CN112694477B - Pyrazolo ring compound, pharmaceutical composition containing pyrazolo ring compound, preparation method and application of pyrazolo ring compound - Google Patents

Pyrazolo ring compound, pharmaceutical composition containing pyrazolo ring compound, preparation method and application of pyrazolo ring compound Download PDF

Info

Publication number
CN112694477B
CN112694477B CN202011084004.4A CN202011084004A CN112694477B CN 112694477 B CN112694477 B CN 112694477B CN 202011084004 A CN202011084004 A CN 202011084004A CN 112694477 B CN112694477 B CN 112694477B
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
formula
hydrogen
acceptable form
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
CN202011084004.4A
Other languages
Chinese (zh)
Other versions
CN112694477A (en
Inventor
陈寿军
宋帅
张毅涛
田强
宋宏梅
薛彤彤
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical 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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN112694477A publication Critical patent/CN112694477A/en
Application granted granted Critical
Publication of CN112694477B publication Critical patent/CN112694477B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The invention belongs to the field of pharmaceutical chemistry, and relates to pyrazolo ring compounds, a pharmaceutical composition containing the pyrazolo ring compounds, a preparation method and application thereof. Specifically, the invention provides a compound with a structure shown in a formula I, which has better TGF beta R1 inhibition activity and higher selectivity, can be used as a high-efficiency and high-selectivity TGF beta R1 inhibitor, and has anti-tumor activity.

Description

Pyrazolo ring compound, pharmaceutical composition containing pyrazolo ring compound, preparation method and application of pyrazolo ring compound
Citation of related application
The present invention claims priority to the invention patent application No. 201911008123.9 filed in china on 10/22 of 2019, entitled "pyrazolo ring compound, pharmaceutical compositions comprising the same, methods of preparation thereof, and uses thereof, the entire contents of which are incorporated herein by reference.
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and relates to a pyrazolo ring compound with TGF beta R1 inhibition activity, a preparation method thereof, a pharmaceutical composition containing the pyrazolo ring compound and medical application thereof.
Background
Transforming growth factor-beta (transforming growth factor-beta, TGF-beta) is a multifunctional cytokine that regulates a variety of cellular responses, such as cell proliferation, differentiation, migration, and apoptosis. TGF-beta superfamily includes TGF-beta 1, TGF-beta 2, TGF-beta 3, activin, inhibin, bone morphogenic proteins, and the like. TGF- β is signaled by two highly conserved single transmembrane serine/threonine kinases tgfβr1 and tgfβr2 (ACS med. Chem. Lett.,2018,9,1117).
Smads are important TGF- β signaling and regulatory molecules in cells that can transduce TGF- β signaling directly from the cell membrane into the nucleus, TGF- β/Smads signaling pathways play an important role in tumor development and progression. In TGF-beta/Smads signaling, activated TGF-beta first binds to TGF-beta R2 at the surface of the cell membrane, forms a heterodimeric complex, and is further recognized and bound by TGF-beta R1. Activated tgfβr1 further phosphorylates Smad2/Smad3 proteins, which in turn bind further to Smad4 to form a heterotrimeric complex that enters the nucleus to act synergistically with co-activators/inhibitors to regulate transcription of target genes (Nature, 2003,425,577). Any change in the TGF-beta/Smads signaling pathway results in an abnormality in the signaling pathway (PNAS, 2019,116,9166).
TGF- β signaling pathways are deregulated in many diseases including cancer, gastric, colorectal, prostate, ovarian, pancreatic, liver, lung, cervical and head and neck cancer cell lines and tumor tissues with significantly elevated levels of tgfβr1 protein. Activation of TGF- β signaling pathways causes significant pathological effects in tumor stroma, including immunosuppression, angiogenesis, and connective tissue hyperplasia. Furthermore, TGF- β signaling pathway can enhance the invasiveness of tumor cells, promote epithelial transformation into the interstitium, and increase the tolerance to treatment by tumor epithelial cells (nat. Neurosci.,2014,17,943).
Currently, the development of inhibitors against the key target tgfβr1 in TGF- β signaling pathways has been gaining attention in the pharmaceutical industry, published patent applications including WO 02/094833 A1, WO 2009/150047 A1, WO 2017/035118 A1, WO 2018/019106 A1, and the like. There remains a need in the art for novel tgfβr1 inhibitors, particularly tgfβr1 inhibitors having high activity and selectivity.
Disclosure of Invention
Problems to be solved by the invention
Through extensive research, the invention unexpectedly discovers that pyrazolo ring compounds and corresponding preparation methods thereof can remarkably inhibit the activity of TGF beta R1, have good selectivity between TGF beta R1 and TGF beta R2, and can be used as TGF beta R1 inhibitors for treating proliferative disorders and apoptosis dysregulated disorders at least partially mediated by TGF-beta signaling pathways, particularly diseases at least partially mediated by TGF beta R1, such as cancers, e.g. liver cancers.
Solution for solving the problem
In a first aspect, the present invention provides a compound having the structure of formula I or a pharmaceutically acceptable form thereof,
wherein,
each X is independently selected from-CH=, -CH 2 -, -NH-and-n=, and satisfies valence requirements;
n is 0, 1 or 2;
when n is a number of times 0,is a single bond; when n is 1, ">Is a single bond or a double bond; when n is 2, the bond between two X's andeach independently is a single bond or a double bond, and is not simultaneously a double bond;
R 1 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 4 Substitution;
R 4 each at each occurrence is independently selected from hydrogen, deuterium, cyano, C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl;
R 2 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 5 Substitution;
R 5 each at each occurrence is independently selected from hydrogen, deuterium, cyano, halogen, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, -OR a 、-NR b R c 、-C(=O)R a 、-C(=O)OR a and-C (=O) NR b R c The C is 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl optionally substituted with one or more R 7 Substitution;
R 3 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 6 Substitution;
R 6 each at each occurrence is independently selected from hydrogen, deuterium, cyano, halogen, C 1-6 Alkyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, -OR a 、-NR b R c 、-C(=O)R a 、-C(=O)OR a 、-C(=O)NR b R c 、-S(=O) q R a and-S (=o) q NR b R c Q is 1 or 2, said C 1-6 Alkyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl optionally substituted with one or more R 7 Substitution;
R 7 each at each occurrence is independently selected from hydrogen, deuterium, halogen, -OR a 、-NR b R c 、-C(=O)R a 、-C(=O)OR a and-C (=O) NR b R c
R a Each at each occurrence is independently selected from hydrogen, C 1-6 Alkyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl;
R b and R is c Each at each occurrence is independently selected from hydrogen, C 1-6 Alkyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl, or R b And R is c Together with the atoms to which they are attached, form a 3-7 membered ring;
the pharmaceutically acceptable form is selected from the group consisting of pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, nitrogen oxides, isotopic labels, metabolites and prodrugs.
In a second aspect, the present invention provides a specific compound having the structure of formula I, comprising:
(1) 3- (benzo [ d ] thiazol-6-yl) -5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one;
(2) 3- (benzo [ d ] thiazol-6-yl) -5- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one;
(3) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -5- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one;
(4) 3- (benzo [ d ] thiazol-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(5) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(6) 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(7) 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(8) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(9) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(10) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (3-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(11) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (3-bromophenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(12) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (pyrazin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one;
(13) 4- (3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -7-oxo-2H-pyrazolo [3,4-c ] pyridin-6 (7H) -yl) -N-methylbenzenesulfonamide;
(14) 3- (benzo [ d ] thiazol-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(15) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(16) 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(17) 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(18) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(19) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(20) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (3-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(21) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (3-bromophenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(22) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (pyrazin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one;
(23) 4- (3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -7-oxo-4, 5-dihydro-2H-pyrazolo [3,4-c ] pyridin-6 (7H) -yl) -N-methylbenzenesulfonamide;
(24) 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [4,3-d ] pyrimidin-7 (6H) -one; and
(25) 3- (benzo [ d ] thiazol-6-yl) -6- (1-methyl-1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2, 6-dihydro-7H-pyrazolo [3,4-c ] pyridin-7-one.
In a third aspect, the present invention provides a method for preparing a compound having the structure of formula I above, comprising:
1) A process for the preparation of a compound of formula I-1 comprising:
step 1-1: cyclizing the compound 1a and oxaloacetate diethyl sodium salt to obtain a compound 2a;
step 1-2: the compound 2a is subjected to hydrolysis reaction to obtain a compound 3a;
step 1-3: the compound 4a and 2-bromo-1, 1-diethoxyethane undergo substitution reaction to obtain a compound 5a;
Step 1-4: the compound 5a and the compound 3a are subjected to condensation reaction to obtain a compound 6a;
step 1-5: cyclizing the compound 6a to obtain a compound 7a;
step 1-6: the compound 7a is subjected to sulfonylation or halogenation to obtain a compound 8a;
step 1-7: the compound 8a and the compound 9a are subjected to a coupling reaction to obtain a compound of the formula I-1;
wherein LG is a leaving group selected from trifluoromethanesulfonyl oxy and halogen, preferably chloro or bromo; w is selected from boric acid group, 4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl and tri-n-butylstannyl; r is R 1 、R 2 And R is 3 As defined above;
2) A process for the preparation of a compound of formula I-2 comprising:
step 2-1: the compound 7a is subjected to reduction reaction to obtain a compound 10a;
step 2-2: the compound 10a is subjected to sulfonylation or halogenation to obtain a compound 11a;
step 2-3: the compound 11a and the compound 9a are subjected to a coupling reaction to obtain a compound of the formula I-2;
wherein R is 1 、R 2 、R 3 LG and W are as defined above;
3) A process for the preparation of a compound of formula I-4 comprising:
step 3-1: the compound 2a is subjected to acylation reaction and halogenation reaction to obtain a compound 12a;
step 3-2: the compound 12a and the compound 13a undergo reductive amination reaction to obtain a compound 14a;
step 3-3: the compound 14a is subjected to hydrolysis reaction to obtain a compound 15a;
Step 3-4: the compound 15a is subjected to condensation reaction to obtain a compound 16a;
step 3-5: coupling the compound 16a and the compound 9a to obtain a compound of the formula I-4;
wherein LG is halogen, preferably chloro or bromo; r is R 1 、R 2 、R 3 And W is as defined above.
In a fourth aspect, the present invention provides a pharmaceutical composition comprising at least one compound having the structure of formula I as described above, or a pharmaceutically acceptable form thereof, and one or more pharmaceutically acceptable carriers.
In a fifth aspect, the present invention provides a kit comprising:
a) At least one compound having the structure of formula I as described above or a pharmaceutically acceptable form thereof, as a first therapeutic agent, or as a first pharmaceutical composition as described above;
b) Optionally at least one other therapeutic agent as a second therapeutic agent, or a pharmaceutical composition comprising the other therapeutic agent as a second pharmaceutical composition; and
c) Optionally package and/or instructions.
In a sixth aspect, the present invention provides a compound having the structure of formula I as described above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, for use as a tgfβr1 inhibitor for use in the prevention and/or treatment of a disease or disorder mediated at least in part by tgfβr1 (in particular cancer, such as liver cancer).
In a seventh aspect, the present invention provides the use of a compound having the structure of formula I as described above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, as an inhibitor of tgfβr1.
In an eighth aspect, the present invention provides the use of a compound having the structure of formula I as described above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, in the manufacture of a medicament for the prevention and/or treatment of a disease or condition mediated at least in part by tgfβr1, in particular cancer, such as liver cancer.
In a ninth aspect, the present invention provides a method for the prevention and/or treatment of a disease or disorder mediated at least in part by tgfβr1 (in particular cancer, such as liver cancer), comprising the steps of: a prophylactically and/or therapeutically effective amount of a compound having the structure of formula I as described above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, is administered to a subject in need thereof.
In a tenth aspect, the present invention provides a pharmaceutical combination composition comprising a compound having the structure of formula I as described above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, together with at least one other tgfβr1 inhibitor.
ADVANTAGEOUS EFFECTS OF INVENTION
The pyrazolo ring compound provided by the invention can be used as a high-efficiency and high-selectivity TGF beta R1 inhibitor, has anti-tumor activity, and has fewer toxic and side effects and drug interactions. The synthesis method is mild, the operation is simple and easy, and the method is suitable for industrial mass production.
Detailed Description
Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described herein; it is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
[ definition of terms ]
Unless otherwise indicated, the following terms have the following meanings in the present invention.
The terms "comprising," "including," "having," or "containing," or any other variation thereof, are intended to cover a non-exclusive or open-ended inclusion. For example, a composition, method, or apparatus that comprises a list of elements is not necessarily limited to only those elements explicitly listed, but may also include other elements not explicitly listed or inherent to such composition, method, or apparatus.
The term "pharmaceutically acceptable salt" refers to salts of the compounds of the invention which are substantially non-toxic to organisms. Pharmaceutically acceptable salts generally include, but are not limited to, salts formed from the compounds of the present invention by reaction with pharmaceutically acceptable inorganic/organic acids or inorganic/organic bases, such salts also being referred to as acid addition salts or base addition salts.
The term "pharmaceutically acceptable ester" refers to an ester that is substantially non-toxic to an organism in which it is hydrolyzed to form a compound of the invention or a salt thereof. Pharmaceutically acceptable esters generally include, but are not limited to, esters of the compounds of the present invention with pharmaceutically acceptable carboxylic or sulfonic acids, such esters also being referred to as carboxylic or sulfonic acid esters.
The term "isomer" refers to a compound that has the same molecular weight due to the same number and type of atoms, but differs in the spatial arrangement or configuration of the atoms.
The term "stereoisomer" (or "optical isomer") refers to a stable isomer that has a perpendicular plane of asymmetry due to at least one chiral factor (including chiral center, chiral axis, chiral plane, etc.), thereby enabling rotation of plane polarized light. The present invention also includes stereoisomers and mixtures thereof, due to the presence of asymmetric centers and other chemical structures which may lead to stereoisomers. Since the compounds of the present invention (or pharmaceutically acceptable salts thereof) include asymmetric carbon atoms, they can exist as single stereoisomers, racemates, mixtures of enantiomers and diastereomers. In general, these compounds can be prepared in the form of racemates. However, if desired, such compounds can be prepared or isolated to give pure stereoisomers, i.e., single enantiomers or diastereomers, or mixtures enriched in single stereoisomers (purity. Gtoreq.98%,. Gtoreq.95%,. Gtoreq.93%,. Gtoreq.90%,. Gtoreq.88%,. Gtoreq.85% or. Gtoreq.80%). As described below, individual stereoisomers of the compounds are prepared synthetically from optically active starting materials containing the desired chiral centers or by preparation of mixtures of enantiomeric products followed by separation or resolution, e.g., conversion to mixtures of diastereomers followed by separation or recrystallization, chromatography, use of chiral resolving agents, or direct separation of enantiomers on chiral chromatographic columns. Starting compounds having specific stereochemistry are either commercially available or prepared according to the methods described below and resolved by methods well known in the art. The term "enantiomer" refers to a pair of stereoisomers that have non-overlapping mirror images of each other. The term "diastereoisomer" or "diastereomer" refers to optical isomers that do not form mirror images of each other. The term "racemic mixture" or "racemate" refers to a mixture containing equal parts of a single enantiomer (i.e., an equimolar mixture of the two R and S enantiomers). The term "non-racemic mixture" refers to a mixture containing unequal portions of individual enantiomers. All stereoisomeric forms of the compounds of the invention are within the scope of the invention unless otherwise indicated.
The term "tautomer" (or "tautomeric form") refers to structural isomers having different energies that can be converted to each other by a low energy barrier. If tautomerism is possible (e.g., in solution), chemical equilibrium of the tautomers can be achieved. For example, proton tautomers (or proton transfer tautomers) include, but are not limited to, interconversions by proton transfer, such as keto-enol isomerisation, imine-enamine isomerisation, amide-imine alcohol isomerisation, and the like. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The term "polymorph" (or "polymorphic form") refers to a solid crystalline form of a compound or complex. The polymorphs of a molecule can be obtained by a number of known methods by a person skilled in the art. Such methods include, but are not limited to, melt recrystallization, melt cooling, solvent recrystallization, desolvation, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, and sublimation. In addition, polymorphs can be detected, classified and identified using well known techniques including, but not limited to, differential Scanning Calorimetry (DSC), thermogravimetric analysis (TGA), X-ray powder diffraction (XRPD), single crystal X-ray diffraction (SCXRD), solid state Nuclear Magnetic Resonance (NMR), infrared spectroscopy (IR), raman spectroscopy, scanning Electron Microscopy (SEM), and the like.
The term "solvate" refers to a substance formed by the association of a compound of the invention (or a pharmaceutically acceptable salt thereof) with at least one solvent molecule by non-covalent intermolecular forces. Common solvates include, but are not limited to, hydrates (including hemihydrate, monohydrate, dihydrate, trihydrate, and the like), ethanolates, acetonates, and the like.
The term "nitroxide" refers to compounds formed by oxidation of nitrogen atoms in tertiary amines or nitrogen (aromatic) containing heterocyclic structures. For example, the nitrogen atom in the 1-position of the parent nucleus of the compound of formula I may form the corresponding nitroxide.
The term "isotopic label" refers to a derivative compound from which a specific atom in a compound of the present invention is replaced by its isotopic atom. Unless otherwise indicated, the compounds of the invention include various isotopes of H, C, N, O, F, P, S, cl, e.g 2 H(D)、 3 H(T)、 13 C、 14 C、 15 N、 17 O、 18 O、 18 F、 31 P、 32 P、 35 S、 36 S and 37 Cl。
the term "metabolite" refers to a derivative compound of the present invention which is formed after metabolism. For further information on metabolism see Goodman and Gilman's The Pharmacological Basis of Therapeutics (9 th ed.)[M],McGraw-Hill International Editions,1996。
The term "prodrug" refers to a derivative compound that is capable of providing a compound of the invention directly or indirectly after administration to a subject. Particularly preferred derivative compounds or prodrugs are compounds that, when administered to an individual, may increase the bioavailability of the compounds of the invention (e.g., are more readily absorbed into the blood) or promote delivery of the parent compound to the site of action (e.g., the lymphatic system). All prodrug forms of the compounds of the invention are within the scope of the invention unless otherwise indicated, and the various prodrug forms are well known in the art.
The term "independently" means that at least two groups (or ring systems) present in the structure that are the same or similar in value range may have the same or different meanings in the particular case. For example, substituent X and substituent Y are each independently hydrogen, halogen, hydroxy, cyano, alkyl or aryl, then when substituent X is hydrogen, substituent Y may be either hydrogen or halogen, hydroxy, cyano, alkyl or aryl; similarly, when the substituent Y is hydrogen, the substituent X may be either hydrogen or halogen, hydroxy, cyano, alkyl or aryl.
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
The term "alkyl" refers to a straight or branched aliphatic hydrocarbon group. For example, the term "C" as used in the present invention 1-6 Alkyl "refers to an alkyl group having 1 to 6 carbon atoms. Common alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl, and the like. The alkyl groups of the present invention are optionally substituted with one or more substituents described herein (e.g., halogen).
The term "haloalkyl" refers to an alkyl group substituted with one or more (such as 1 to 3) identical or different halogen atoms. For example, the term "C" as used in the present invention 1-6 Haloalkyl "refers to haloalkyl groups having 1 to 6 carbon atoms. Common haloalkyl groups include, but are not limited to, -CH 2 F、-CHF 2 、-CF 3 、-CH 2 CF 3 、-CF 2 CF 3 、-CH 2 CH 2 CF 3 、-CH 2 Cl, and the like. Haloalkyl groups in the present invention are optionally substituted with one or more substituents described herein.
The term "alkoxy" refers to an alkyl group attached to the remainder of the molecule through an oxygen atom. Common alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentyloxy, hexyloxy, and the like. The alkoxy groups of the present invention are optionally substituted with one or more substituents described herein.
The term "cycloalkyl" refers to a saturated or partially saturated, monocyclic or polycyclic (such as bicyclic) non-aromatic hydrocarbon group. For example, the term "C" as used in the present invention 3-8 Cycloalkyl "refers to cycloalkyl groups having 3 to 8 carbon atoms. Common cycloalkyl groups include, but are not limited to, monocyclic cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, and the like; or bicyclic cycloalkyl, including fused, bridged or spiro rings, such as bicyclo [1.1.1]Amyl, bicyclo [2.2.1]Heptyl, bicyclo [3.2.1]Octyl and bicyclo [5.2.0 ]Nonyl, decalin, and the like. Cycloalkyl groups in the present invention are optionally substituted with one or more substituents described herein (e.g., methyl).
The term "heterocycloalkyl" refers to a saturated or partially saturated, monocyclic or polycyclic (such as bicyclic) non-aromatic group, the ring atoms of which are made up of carbon atoms and at least one heteroatom selected from nitrogen, oxygen and sulfur. If valence requirements are met, the heterocycloalkyl group may be attached to the remainder of the molecule through any one of the ring atoms. For example, the term "3-8 membered heterocycloalkyl" as used in the present invention refers to a heterocycloalkyl group having 3 to 8 ring atoms. Common heterocycloalkyl groups include, but are not limited to, oxiranyl (oxairanyl), aziridinyl (aziridinyl), azetidinyl (azetidinyl), oxetanyl (oxaetanyl), tetrahydrofuranyl (tetrahydrofuranyl), dioxolyl (dioxanyl), pyrrolidinyl (pyrrosinyl), pyrrolidinonyl (pyrrosinyl), imidazolidinyl (imidazolidinyl), pyrazolidinyl (pyrrosidinyl), tetrahydropyranyl (tetrahydrofuranyl), piperidinyl (piperadinyl), piperazinyl (piperazinyl), morpholinyl (morpholinyl), thiomorpholinyl (thiomorpholinyl), dithianyl (dithianyl) or trithianyl (trithionyl). The heterocycloalkyl groups of the present invention are optionally substituted with one or more substituents described herein.
The term "aryl" refers to a monocyclic or fused polycyclic aromatic hydrocarbon group having a conjugated pi-electron system. For example, the term "C" as used in the present invention 6-10 Aryl "refers to aryl groups having 6 to 10 carbon atoms. Common aryl groups include, but are not limited to, phenyl, naphthyl, anthryl, phenanthryl, acenaphthylenyl, azulenyl, fluorenyl, indenyl, pyrenyl, and the like. Aryl groups of the invention are optionally substituted with one or more substituents described herein (e.g., halogen, hydroxy, cyano, nitro, C 1-6 Alkyl, etc.) substitution.
The term "heteroaryl" refers to an aromatic group having a single ring or a fused multiple ring (in particular a benzo-fused multiple ring) of conjugated pi electron systems, the ring atoms of which consist of carbon atoms and at least one heteroatom selected from nitrogen, oxygen and sulfur. If valence requirements are met, the heterocycloalkyl group may be attached to the remainder of the molecule through any one of the ring atoms. For example, the term "5-10 membered heteroaryl" as used in the present invention refers to heteroaryl groups having 5 to 10 ring atoms. Common heteroaryl groups include, but are not limited to, thienyl (thienyl), furyl (furyl), pyrrolyl (pyrroyl), oxazolyl (oxazolyl), thiazolyl (thiazolyl), imidazolyl (iminozolyl), pyrazolyl (pyrazozolyl), isoxazolyl (isoxazolyl), isothiazolyl (isothiazolyl), oxadiazolyl (oxazoyl), triazolyl (triazolyl), thiadiazolyl (thiadiazolyl), pyridyl (pyridinyl), pyridazinyl (pyridazinyl), pyrimidinyl (pyrimidyl), pyrazinyl (pyraziyl), triazinyl (triazinyl), and benzo derivatives thereof, and the like. Heteroaryl groups of the invention are optionally substituted with one or more substituents described herein (e.g., halogen, C 1-6 Alkyl, etc.) substitution.
The term "alkenyl" refers to a straight or branched aliphatic hydrocarbon group having at least one c=c double bond. For example, the term "C" as used in the present invention 2-6 Alkenyl "refers to alkenyl groups having 2 to 6 carbon atoms. Common alkenyl groups include, but are not limited to, ethenyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, n-octenyl, n-decenyl, and the like. Alkenyl groups in the present invention are optionally substituted with one or more substituents described herein.
The term "alkynyl" refers to a straight or branched aliphatic hydrocarbon group having at least one c≡c triple bond. For example, the term "C" as used in the present invention 2-6 Alkynyl "refers to alkynyl groups having 2 to 6 carbon atoms. Common alkynyl groups include, but are not limited to, ethynyl, 2-propynyl, 2-butynyl, 1, 3-butadiynyl, and the like. Alkynyl groups in the present invention are optionally substituted with one or more substituents described herein.
The term "substituted" means that one or more (e.g., 1, 2, 3, or 4) atoms (e.g., hydrogen atoms) or groups of atoms (e.g., triflate groups) on the specified group are replaced with other atoms or groups of atoms, provided that the specified group meets the valence requirements in the current case and forms a stable compound after substitution. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. If a substituent is described as "optionally substituted with …," the substituent may be unsubstituted or substituted. If a first substituent is described as being optionally substituted with one or more of the second list of substituents, one or more hydrogen atoms in the first substituent may be replaced by one or more of the second list of substituents, either alone (or independently of each other) (independitly).
The term "one or more" means 1 or more than 1, e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10, under reasonable conditions.
[ Compounds of the general formula ]
The present invention provides a compound of formula I or a pharmaceutically acceptable form thereof,
wherein,
each X is independently selected from-CH=, -CH 2 -, -NH-and-n=, and satisfies valence requirements;
n is 0, 1 or 2;
when n is a number of times 0,is a single bond; when n is 1, ">Is a single bond or a double bond; when n is 2, the bond between two X's andeach independently is a single bond or a double bond, and is not simultaneously a double bond;
R 1 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 4 Substitution;
R 4 each at each occurrence is independently selected from hydrogen, deuterium, cyano, C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl;
R 2 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 5 Substitution;
R 5 each at each occurrence is independently selected from hydrogen, deuterium, cyano, halogen, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, -OR a 、-NR b R c 、-C(=O)R a 、-C(=O)OR a and-C (=O) NR b R c The C is 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl optionally substituted with one or more R 7 Substitution;
R 3 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 6 Substitution;
R 6 each at each occurrence is independently selected from hydrogen, deuterium, cyano, halogen, C 1-6 Alkyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, -OR a 、-NR b R c 、-C(=O)R a 、-C(=O)OR a 、-C(=O)NR b R c 、-S(=O) q R a and-S (=o) q NR b R c Q is 1 or 2, said C 1-6 Alkyl, C 3-8 Cycloalkyl or 3-8 membered heterocycloalkyl optionally substituted with one or more R 7 Substitution;
R 7 each at each occurrence is independently selected from hydrogen, deuterium, halogen, -OR a 、-NR b R c 、-C(=O)R a 、-C(=O)OR a and-C (=O) NR b R c
R a Each at each occurrence is independently selected from hydrogen, C 1-6 Alkyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl;
R b and R is c Each at each occurrence is independently selected from hydrogen, C 1-6 Alkyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl, or R b And R is c Together with the atoms to which they are attached, form a 3-7 membered ring;
the pharmaceutically acceptable form is selected from the group consisting of pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, nitrogen oxides, isotopic labels, metabolites and prodrugs.
In some embodiments of the invention, R in the above-described compounds of formula I 1 Is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 4 Substituted, X, n, R 2 、R 3 And R is 4 As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 1 Is a 5-6 membered heteroaryl, said 5-6 membered heteroaryl optionally being substituted with one or more R 4 Substituted, X, n, R 2 、R 3 And R is 4 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 1 Is a 6 membered heteroaryl, said 6 membered heteroaryl optionally being substituted with one or more R 4 Substituted, X, n, R 2 、R 3 And R is 4 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 1 Is a pyridinyl group, optionally substituted with one or more C 1-6 Alkyl or C 1-6 Haloalkyl substitution, X, n, R 2 And R is 3 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I 1 Selected from 6-methylpyridin-2-yl, 6-trifluoromethylpyridin-2-yl and 6-difluoromethylpyridin-2-yl, X, n, R 2 And R is 3 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I 2 Is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 5 Substituted, X, n, R 1 、R 3 And R is 5 As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 2 Is a 5-10 membered fused ring heteroaryl, said 5-10 membered fused ring heteroaryl optionally substituted with one or more R 5 Substituted, X, n, R 1 、R 3 And R is 5 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 2 Selected from benzothiazolyl and triazolopyridinyl, optionally substituted with one or more R 5 Substituted, X, n, R 1 、R 3 And R is 5 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 2 Selected from benzo [ d ]]Thiazolyl and [1,2,4 ]]Triazolo [1,5-a ]]Pyridyl, X, n, R 1 And R is 3 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I 2 Selected from benzo [ d ]]Thiazol-6-yl and [1,2,4 ]]Triazolo [1,5-a ]]Pyridin-6-yl, X, n, R 1 And R is 3 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl and 5-6 membered heteroaryl, optionally substituted with one or more R 6 Substituted, X, n, R 1 、R 2 And R is 6 As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, halogen, C 1-6 Alkyl, -OR a 、-S(=O) q R a and-S (=o) q NR b R c Q is 1 or 2, X, n, R 1 、R 2 、R a 、R b And R is c As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, halogen, -OR a 、-S(=O) q R a and-S (=o) q NR b R c Q is 1 or 2, X, n, R 1 、R 2 、R a 、R b And R is c As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, fluorine, bromine, hydroxyl, C 1-6 Alkyl, -O-C 1-6 Alkyl, -S (=o) 2 -C 1-6 Alkyl and-S (=o) 2 -NH-C 1-6 Alkyl, X, n, R 1 And R is 2 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, fluorine, bromine, hydroxy, -O-C 1-6 Alkyl, -S (=o) 2 -C 1-6 Alkyl and-S (=o) 2 -NH-C 1-6 Alkyl, X, n, R 1 And R is 2 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each occurrence is independently selected from hydrogen, bromo, hydroxy, methyl, methoxy, methanesulfonyl, and methanesulfonyl, X, n, R 1 And R is 2 As defined above.
In some more preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each occurrence is independently selected from hydrogen, bromo, hydroxy, methoxy, methanesulfonyl, and methanesulfonyl, X, n, R 1 And R is 2 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from 4-methoxyphenyl, 4-hydroxyphenyl, 1- (methylsulfonyl) -1H-pyrazol-4-yl, 3-hydroxyphenyl, 3-bromophenyl, pyrazin-2-yl, 4- (N-methylsulfamoyl) phenyl and 1- (methyl) -1H-pyrazol-4-yl, X, N, R 1 And R is 2 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I 3 Selected from 4-methoxyphenyl, 4-hydroxyphenyl, 1- (methylsulfonyl) -1H-pyrazole-4-yl, 1H-pyrazol-4-yl, 3-hydroxyphenyl, 3-bromophenyl, pyrazin-2-yl and 4- (N-methylsulfamoyl) phenyl, X, N, R 1 And R is 2 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I 1 Is one or more R 4 Substitution, said R 4 Selected from hydrogen, deuterium, C 1-6 Alkyl, C 1-6 Haloalkyl and C 3-8 Cycloalkyl, X, n, R 1 、R 2 And R is 3 As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 1 Is one or more R 4 Substitution, said R 4 Selected from methyl, difluoromethyl and trifluoromethyl, X, n, R 1 、R 2 And R is 3 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I 2 Is one or more R 5 Substitution, said R 5 Selected from hydrogen and C 1-6 Alkyl, X, n, R 1 、R 2 And R is 3 As defined above.
In some preferred embodiments of the invention, R 5 Is hydrogen.
In some embodiments of the invention, R in the above-described compounds of formula I 3 Is one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, halogen, C 1-6 Alkyl, C 1-6 Alkoxy, methanesulfonyl and methanesulfonyl groups, X, n, R 1 、R 2 And R is 3 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I 3 Is one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, halogen, C 1-6 Alkoxy, methanesulfonyl and methanesulfonyl groups, X, n, R 1 、R 2 And R is 3 As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 3 Is one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, methyl, methoxy, fluoro, bromo, methanesulfonyl and methylaminosulfonyl, X, n, R 1 、R 2 And R is 3 As defined above.
In some preferred embodiments of the invention, R in the above-described compounds of formula I 3 Is one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, methoxy, fluoro, bromo, methanesulfonyl and methylaminosulfonyl, X, n, R 1 、R 2 And R is 3 As defined above.
In some preferred embodiments of the invention, n is 0 or 1.
When n is 1, andin the case of double bonds, the compounds of formula I may be compounds of formula I-1:
wherein R is 1 、R 2 And R is 3 As defined above.
When n is 1, andwhen a single bond is used, the compound of the formula I can be a compound of the formula I-2:
Wherein R is 1 、R 2 And R is 3 As defined above.
When n is 1, andin the case of double bonds, the compounds of formula I described above may be compounds of formula I-3:
wherein R is 1 、R 2 And R is 3 As defined above.
When n is 0, the compound of formula I may be a compound of formula I-4:
wherein R is 1 、R 2 And R is 3 As defined above.
When n is 1, andwhen a single bond is used, the compound of the formula I can be a compound of the formula I-5:
wherein R is 1 、R 2 And R is 3 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 1 Is a 6 membered heteroaryl, said 6 membered heteroaryl optionally being substituted with one or more R 4 Substituted, R 2 、R 3 And R is 4 As defined above.
In some preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 1 Is a pyridinyl group, optionally substituted with one or more C 1-6 Alkyl substitution, R 2 And R is 3 As defined above.
In some more preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 1 Is 6-methylpyridin-2-yl, R 2 And R is 3 As defined above.
In some embodiments of the invention, the above formula I-1,R in the compound of formula I-2, formula I-3, formula I-4 or formula I-5 2 Is a 5-10 membered fused ring heteroaryl, said 5-10 membered fused ring heteroaryl optionally substituted with one or more R 5 Substituted, R 1 、R 3 And R is 5 As defined above.
In some preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 2 Selected from benzothiazolyl and triazolopyridinyl, optionally substituted with one or more R 5 Substituted, R 1 、R 3 And R is 5 As defined above.
In some more preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 2 Selected from benzo [ d ]]Thiazolyl and [1,2,4 ]]Triazolo [1,5-a ]]Pyridyl, R 1 And R is 3 As defined above.
In some most preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 2 Selected from benzo [ d ]]Thiazol-6-yl and [1,2,4 ]]Triazolo [1,5-a ]]Pyridin-6-yl, R 1 And R is 3 As defined above.
In some embodiments of the invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 3 Selected from phenyl and 5-6 membered heteroaryl, optionally substituted with one or more R 6 Substituted, R 1 、R 2 And R is 6 As defined above.
In some preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, halogen, C 1-6 Alkyl, -OR a 、-S(=O) q R a and-S (=o) q NR b R c Preferably hydrogen, halogen, -OR a 、-S(=O) q R a and-S (=o) q NR b R c Q is 1 or 2, R 1 、R 2 、R a 、R b And R is c As defined above.
In some more preferred embodiments of the present invention, R in the above-described compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each occurrence is independently selected from hydrogen, bromo, hydroxy, methyl, methoxy, methanesulfonyl, and methanesulfonyl, preferably hydrogen, bromo, hydroxy, methoxy, methanesulfonyl, and methanesulfonyl, R 1 And R is 2 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I-1 or formula I-2 3 Selected from 4-hydroxyphenyl, 1- (methylsulfonyl) -1H-pyrazol-4-yl, 3-hydroxyphenyl, 3-bromophenyl, pyrazin-2-yl, 4- (N-methylsulfamoyl) phenyl and 1- (methyl) -1H-pyrazol-4-yl, preferably 4-hydroxyphenyl, 1H-pyrazol-4-yl, 1- (methylsulfonyl) -1H-pyrazol-4-yl, 3-hydroxyphenyl, 3-bromophenyl, pyrazin-2-yl and 4- (N-methylsulfamoyl) phenyl, R 1 And R is 2 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I-3 3 Is 4-hydroxyphenyl, R 1 And R is 2 As defined above.
In some most preferred embodiments of the invention, R in the above-described compounds of formula I-4 3 Selected from 4-methoxyphenyl and 4-hydroxyphenyl, R 1 And R is 2 As defined above.
In some embodiments of the invention, in the compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 described above,
R 1 is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 4 Substitution, said R 4 Selected from hydrogen, deuterium, C 1-6 Alkyl, C 1-6 Haloalkyl and C 3-8 Cycloalkyl;
R 2 is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 5 Substitution, said R 5 Selected from hydrogen and C 1-6 An alkyl group;
R 3 selected from phenyl and 5-6 membered heteroaryl, optionally substituted with one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, halogen, C 1-6 Alkyl, C 1-6 Alkoxy, methanesulfonyl and methanesulfonyl.
In some embodiments of the invention, in the compounds of formula I-1, formula I-2, formula I-3, formula I-4 or formula I-5 described above,
R 1 is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 4 Substitution, said R 4 Selected from hydrogen, deuterium, C 1-6 Alkyl, C 1-6 Haloalkyl and C 3-8 Cycloalkyl;
R 2 is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 5 Substitution, said R 5 Selected from hydrogen and C 1-6 An alkyl group;
R 3 selected from phenyl and 5-6 membered heteroaryl, optionally substituted with one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, halogen, C 1-6 Alkoxy, methanesulfonyl and methanesulfonyl.
In addition, the invention also provides the following compounds or pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, nitrogen oxides, isotopic labels, metabolites or prodrugs thereof, the structures and names of which are shown in the following table:
/>
/>
[ preparation method ]
The invention provides a preparation method of the compound of the formula I, which comprises the following steps:
1) A process for the preparation of a compound of formula I-1 comprising:
step 1-1: cyclizing the compound 1a and oxaloacetate diethyl sodium salt to obtain a compound 2a;
step 1-2: the compound 2a is subjected to hydrolysis reaction to obtain a compound 3a;
step 1-3: the compound 4a and 2-bromo-1, 1-diethoxyethane undergo substitution reaction to obtain a compound 5a;
step 1-4: the compound 5a and the compound 3a are subjected to condensation reaction to obtain a compound 6a;
Step 1-5: cyclizing the compound 6a to obtain a compound 7a;
step 1-6: the compound 7a is subjected to sulfonylation or halogenation to obtain a compound 8a;
step 1-7: the compound 8a and the compound 9a are subjected to a coupling reaction to obtain a compound of the formula I-1;
wherein LG is a leaving group selected from trifluoromethanesulfonyl oxy and halogen, preferably chloro or bromo; w is selected from boric acid group, 4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl and tri-n-butylstannyl; r is R 1 、R 2 And R is 3 As defined above;
2) A process for the preparation of a compound of formula I-2 comprising:
step 2-1: the compound 7a is subjected to reduction reaction to obtain a compound 10a;
step 2-2: the compound 10a is subjected to sulfonylation or halogenation to obtain a compound 11a;
step 2-3: the compound 11a and the compound 9a are subjected to a coupling reaction to obtain a compound of the formula I-2;
wherein R is 1 、R 2 、R 3 LG and W are as defined above;
3) A process for the preparation of a compound of formula I-4 comprising:
step 3-1: the compound 2a is subjected to acylation reaction and halogenation reaction to obtain a compound 12a;
step 3-2: the compound 12a and the compound 13a undergo reductive amination reaction to obtain a compound 14a;
step 3-3: the compound 14a is subjected to hydrolysis reaction to obtain a compound 15a;
step 3-4: the compound 15a is subjected to condensation reaction to obtain a compound 16a;
Step 3-5: coupling the compound 16a and the compound 9a to obtain a compound of the formula I-4;
wherein LG is halogen, preferably chloro or bromo; r is R 1 、R 2 、R 3 And W is as defined above.
In some embodiments of the present invention, step 1-1 of the above-described preparation process is performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., dichloromethane (DCM), chloroform (TCM), 1, 2-dichloroethane (1, 2-DCE), etc.), nitriles (e.g., acetonitrile (AN), etc.), N-methylpyrrolidone (NMP), N-Dimethylformamide (DMF), N-Dimethylacetamide (DMA), tetrahydrofuran (THF), 1, 4-dioxane (Diox), dimethylsulfoxide (DMSO), aromatic hydrocarbons (e.g., toluene (TL), xylene (XY), etc.), and any combination thereof, preferably TL.
In some embodiments of the invention, step 1-1 of the above preparation process is carried out in the presence of a suitable acid, which may be selected from p-toluene sulfonic acid (TsOH) and acetic acid (AcOH), preferably AcOH.
In some embodiments of the invention, step 1-1 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 25-160 ℃.
In some embodiments of the present invention, steps 1-2 in the above preparation process are performed in a suitable organic solvent or a mixed solvent of an organic solvent and water, which may be selected from alcohols (e.g., methanol (MeOH), ethanol (EtOH), n-butanol (n-BuOH), etc.), THF, acOH, diox, and any combination thereof, preferably THF, etOH, or a mixed solvent thereof with water.
In some embodiments of the invention, steps 1-2 of the above preparation process are performed in the presence of a suitable base, which may be selected from sodium hydroxide (NaOH), potassium hydroxide (KOH) and lithium hydroxide (LiOH), preferably LiOH.
In some embodiments of the invention, step 1-2 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-100 ℃, preferably 25-80 ℃.
In some embodiments of the present invention, steps 1-3 of the above preparation process are performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, and any combination thereof, preferably NMP, DMSO, or DMF.
In some embodiments of the present invention, steps 1-3 of the above preparation process are performed in the presence of a suitable base, including an organic base selected from the group consisting of N, N-Diisopropylethylamine (DIPEA), triethylamine (TEA), potassium t-butoxide (t-BuOK) and pyridine (Py), or an inorganic base selected from the group consisting of potassium phosphate (K) 3 PO 4 ) Sodium hydride (NaH), potassium carbonate (K) 2 CO 3 ) Sodium carbonate (Na) 2 CO 3 ) Cesium carbonate (Cs) 2 CO 3 ) And NaOH, preferably Na 2 CO 3 Or t-BuOK.
In some embodiments of the invention, steps 1-3 of the above preparation process are carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 0-120 ℃.
In some embodiments of the present invention, steps 1-4 of the above preparation process are performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, and any combination thereof, preferably NMP, DMSO, or DMF.
In some embodiments of the present invention, steps 1-4 of the above preparation process are performed in the presence of a suitable condensing agent, which may be selected from the group consisting of thionyl chloride (SOCl) 2 ) Oxalyl chloride ((COCl) 2 ) Phosphorus oxychloride (POCl) 3 ) Phosphorus trichloride (PCl) 3 ) Phosphorus pentachloride (PCl) 5 ) Ethyl Chloroformate (ECF), isopropyl chloroformate (IPCF), HATU, HBTU, EEDQ, DEPC, DCC, DIC, EDC, BOP, pyAOP and PyBOP, preferably HATU.
In some embodiments of the present invention, steps 1-4 of the above preparation process are performed in the presence of a suitable base, including an organic base selected from DIPEA, TEA, t-BuOK and Py or an inorganic base selected from K 3 PO 4 、NaH、K 2 CO 3 、Na 2 CO 3 、Cs 2 CO 3 And NaOH, preferably K 2 CO 3 Or t-BuOK.
In some embodiments of the invention, steps 1-4 of the above preparation process are carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 25-120 ℃.
In some embodiments of the present invention, steps 1-5 of the above-described preparation process are performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, aromatic hydrocarbons (e.g., TL, XY), trifluoroacetic acid (TFA), and any combination thereof, preferably TL or TFA.
In some embodiments of the invention, steps 1-5 of the above preparation process are performed in the presence of a suitable acid, which may be selected from TsOH, TFA and AcOH, preferably TsOH or TFA.
In some embodiments of the invention, steps 1-5 of the above preparation process are carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 0-120 ℃.
In some embodiments of the present invention, steps 1-6 of the above-described preparation process are performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, aromatic hydrocarbons (e.g., TL, XY), and any combination thereof, preferably THF or DCM.
In some embodiments of the present invention, steps 1-6 of the above preparation process are performed in the presence of a suitable acylating agent, which may be selected from the group consisting of trifluoromethanesulfonic anhydride (Tf 2 O) and N, N-bis (trifluoromethanesulfonyl) aniline, preferably N, N-bis (trifluoromethanesulfonyl) aniline, the halogenating reagent being selected from phosphorus oxybromide (POBr) 3 ) And POCl 3 POCl is preferred 3
In some embodiments of the present invention, steps 1-6 of the above preparation process are performed in the presence of a suitable base, including an organic base selected from DIPEA, TEA, t-BuOK and Py or an inorganic base selected from K 3 PO 4 、NaH、K 2 CO 3 、Na 2 CO 3 、Cs 2 CO 3 And NaOH, preferably DIPEA or TEA.
In some embodiments of the invention, steps 1-7 of the above preparation process are performed in a suitable organic solvent or a mixed solution of AN organic solvent and water, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), meOH, etOH, t-butanol (t-BuOH), DMF, AN, ethers (e.g., ethylene glycol dimethyl ether (DME), THF, diox), aromatic hydrocarbons (e.g., TL, XY), and any combination thereof, preferably TL or Diox.
In some embodiments of the invention, steps 1-7 of the above preparation process are carried out in the presence of a suitable catalyst, preferably a palladium catalyst, which may be selected from tetrakis (triphenylphosphine) palladium (0) (Pd (PPh) 3 ) 4 ) Palladium (II) acetate (Pd (OAc) 2 )、Pd 2 (dba) 3 、Pd(PPh 3 ) 2 Cl 2 、Pd(PPh 3 ) 2 Cl 2 Dichloromethane complex, pdCl 2 (Amphos) 2 And Pd (dppf) Cl 2 Preferably PdCl 2 (Amphos) 2 Or Pd (PPh) 3 ) 4
In some embodiments of the invention, steps 1-7 of the above preparation method are performed in the presence of a suitable ligand, which may be selected from triphenylphosphine (PPh 3 ) BINAP, tris (o-methylphenyl) phosphine (P (o-tol) 3 ) Tricyclohexylphosphine Tetrafluoroborate (TCHP) and X-PHOS, preferably PPh 3 Or X-PHOS.
In some embodiments of the present invention, steps 1-7 of the above preparation process are performed in the presence of a suitable base, including an organic base selected from DIPEA, TEA, t-BuOK and Py or an inorganic base selected from K 3 PO 4 、NaH、K 2 CO 3 、Na 2 CO 3 、Cs 2 CO 3 And NaOH, preferably K 2 CO 3 Or Cs 2 CO 3
In some embodiments of the invention, steps 1-7 of the above preparation process are carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 50-150 ℃.
In some embodiments of the present invention, step 2-1 of the above-described preparation process is performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), alcohols (e.g., meOH, etOH, n-BuOH), ethyl Acetate (EA), NMP, THF, diox, DMSO, and any combination thereof, preferably MeOH, 1,2-DCE, or EA.
In some embodiments of the invention, step 2-1 of the above preparation process is carried out in the presence of a suitable catalyst, which may be selected from palladium on carbon (Pd/C), palladium hydroxide on carbon (Pd (OH) 2 /C) and platinum oxide (PtO) 2 ) Pd/C or Pd (OH) are preferred 2 /C。
In some embodiments of the invention, step 2-1 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 0-100 ℃.
In some embodiments of the present invention, step 2-2 of the above-described preparation process is performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, aromatic hydrocarbons (e.g., TL, XY), and any combination thereof, preferably THF or DCM.
In some embodiments of the invention, step 2-2 of the above preparation method is performed in the presence of a suitable acylating agent or halogenated agent, which may be selected from Tf 2 O and N, N-bis (trifluoromethanesulfonyl) aniline, preferably N, N-bis (trifluoromethanesulfonyl) aniline, the halogenated reagent being selected from POBr 3 And POCl 3 POCl is preferred 3
In some embodiments of the present invention, step 2-2 of the above preparation method is performed in the presence of a suitable base, including an organic base selected from DIPEA, TEA, t-BuOK and Py or an inorganic base selected from K 3 PO 4 、NaH、K 2 CO 3 、Na 2 CO 3 、Cs 2 CO 3 And NaOH, preferably DIPEA or TEA.
In some embodiments of the invention, step 2-2 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 0-120 ℃.
In some embodiments of the invention, steps 2-3 of the above preparation process are performed in a suitable organic solvent or a mixed solution of an organic solvent and water, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), meOH, etOH, t-BuOH, DMF, AN, ethers (e.g., DME, THF, diox), aromatic hydrocarbons (e.g., TL, XY), and any combination thereof, preferably TL or Diox.
In some embodiments of the invention, steps 2-3 of the above preparation process are carried out in the presence of a suitable catalyst, preferably a palladium catalyst, which may be selected from Pd (PPh 3 ) 4 、Pd(OAc) 2 、Pd 2 (dba) 3 、Pd(PPh 3 ) 2 Cl 2 、Pd(PPh 3 ) 2 Cl 2 Dichloromethane complex, pdCl 2 (Amphos) 2 And Pd (dppf) Cl 2 Preferably PdCl 2 (Amphos) 2 Or Pd (PPh) 3 ) 4
In some embodiments of the present invention, steps 2-3 of the above preparation method are performed in the presence of a suitable ligand selected from PPh 3 、BINAP、P(o-tol) 3 TCHP and X-PHOS, preferably PPh 3 Or X-PHOS.
In some embodiments of the present invention, step 2-3 of the above preparation process is performed in the presence of a suitable base, including an organic base selected from DIPEA, TEA, t-BuOK and Py or an inorganic base selected from K 3 PO 4 、NaH、K 2 CO 3 、Na 2 CO 3 、Cs 2 CO 3 And NaOH, preferably K 2 CO 3 Or Cs 2 CO 3
In some embodiments of the invention, steps 2-3 of the above preparation process are carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 50-150 ℃.
In some embodiments of the present invention, step 3-1 of the above-described preparation process is performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, and any combination thereof, preferably 1,2-DCE, DCM, or DMF.
In some embodiments of the present invention, step 3-1 of the above preparation method is performed in the presence of a suitable acylating agent selected from DMF and N, N-Diethylformamide (DEF), preferably DMF, and a halogenated agent selected from POCl 3 And POBr 3 POCl is preferred 3
In some embodiments of the invention, step 3-1 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 0-100 ℃.
In some embodiments of the present invention, step 3-2 of the above preparation process is performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), THF, meOH, and any combination thereof, preferably DCM or MeOH.
In some embodiments of the invention, step 3-2 of the above preparation process is performed in the presence of a suitable acid, which is AcOH.
In some embodiments of the present invention, step 3-2 of the above preparation method is performed in the presence of a suitable reducing agent, which may be selected from sodium borohydride (NaBH 4 ) Sodium cyanoborohydride (NaBH) 3 CN) and sodium triacetoxyborohydride (NaBH (OAc) 3 ) NaBH (OAc) is preferred 3
In some embodiments of the invention, step 3-2 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-100 ℃, preferably 25-70 ℃.
In some embodiments of the present invention, step 3-3 of the above preparation process is performed in a suitable organic solvent or a mixture of organic solvents and water, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), diox, THF, meOH, and any combination thereof, preferably THF or MeOH.
In some embodiments of the invention, step 3-3 of the above preparation process is carried out in the presence of a suitable base, which may be selected from NaOH, KOH and LiOH, preferably LiOH.
In some embodiments of the invention, step 3-3 of the above preparation process is carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 25-100 ℃.
In some embodiments of the present invention, steps 3-4 of the above preparation process are performed in a suitable organic solvent, which may be selected from the group consisting of halogenated hydrocarbons (e.g., DCM, TCM, 1,2-DCE, etc.), nitriles (e.g., AN, etc.), NMP, DMF, DMA, THF, diox, DMSO, and any combination thereof, preferably THF or DMF.
In some embodiments of the present invention, steps 3-4 of the above preparation process are performed in the presence of a suitable condensing agent, which may be selected from SOCl 2 、(COCl) 2 、POCl 3 、PCl 3 、PCl 5 ECF, IPCF, HATU, HBTU, EEDQ, DEPC, DCC, DIC, EDC, BOP, pyAOP and PyBOP, preferably HATU.
In some embodiments of the present invention, steps 3-4 of the above preparation process are performed in the presence of a suitable base, including an organic base selected from DIPEA, TEA, t-BuOK, NMP and Py or an inorganic base selected from K 3 PO 4 、NaH、K 2 CO 3 、Na 2 CO 3 、Cs 2 CO 3 And NaOH, preferably NMP or DIPEA.
In some embodiments of the invention, steps 3-4 of the above preparation process are carried out at a suitable temperature, said temperature being in the range of 0-200 ℃, preferably 25-100 ℃.
In some embodiments of the invention, steps 3-5 of the above preparation process are performed in a suitable organic solvent. The organic solvent may be selected from the group consisting of halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), methanol, t-butanol, ethanol, DMF, acetonitrile, ethers (e.g., ethylene glycol dimethyl ether, tetrahydrofuran, dioxane), aromatic hydrocarbons (e.g., toluene, xylene), and any combination thereof, preferably dioxane or toluene.
In some embodiments of the invention, steps 3-5 of the above preparation process are performed in the presence of a catalyst. The catalyst is preferably a palladium catalyst, e.g. tetrakis (triphenylphosphine) palladium, palladium acetate, pd 2 (dba) 3 、Pd(PPh 3 ) 2 Cl 2 、Pd(PPh 3 ) 2 Cl 2 Dichloromethane complex or Pd (dppf) Cl 2 Tetrakis (triphenylphosphine) palladium or palladium acetate is preferred.
In some embodiments of the present invention, steps 3-5 of the above preparation process are performed in the presence of a ligand selected from BINAP, tris (o-methylphenyl) phosphorus, triphenylphosphine, tricyclohexylphosphine tetrafluoroborate, X-PHOS, preferably X-PHOS or triphenylphosphine.
In some embodiments of the present invention, steps 3-5 in the above preparation process are preferably carried out in the presence of a suitable base comprising an organic base which may be selected from DIPEA, triethylamine, potassium tert-butoxide and pyridine or an inorganic base which may be selected from potassium phosphate, sodium hydride, potassium carbonate, sodium carbonate, cesium carbonate and sodium hydroxide, preferably potassium carbonate or cesium carbonate.
In some embodiments of the invention, steps 3-5 of the above preparation process are carried out at a suitable temperature, preferably 0-200 ℃, more preferably 50-150 ℃.
[ pharmaceutical composition ]
The term "pharmaceutical composition" refers to a composition that can be used as a medicament comprising a pharmaceutically active ingredient (API) (or therapeutic agent) and optionally one or more pharmaceutically acceptable carriers. The term "pharmaceutically acceptable carrier" refers to an adjuvant that is administered with a therapeutic agent and which is, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and/or other animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable carriers that can be used in the present invention include (but are not limited to): a) Diluents, such as fish oil, docosahexaenoic acid or esters thereof, triglycerides, omega-3 fatty acids or derivatives thereof, dextrose, glucose, glycine, or mixtures thereof; b) Lubricants, such as stearic acid, sodium oleate, sodium chloride, polyethylene glycol, or mixtures thereof; c) Binders, for example gelatin, magnesium carbonate, natural and synthetic gums (such as acacia, sodium alginate), polyvinylpyrrolidone, or mixtures thereof; d) Disintegrants, for example agar, bentonite, xanthan gum, alginic acid or its sodium salt, effervescent agents, or mixtures thereof; e) Absorbents, colorants, flavors and/or sweeteners; f) Emulsifying or dispersing agents, such as caprylic/capric polyethylene glycol glycerides, polyethylene glycol glycerides oleate, glycerol oleate, diethylene glycol monoethyl ester, or other acceptable emulsifying agents: and/or g) substances that enhance absorption of the compound, such as polyethylene glycol 200, polyethylene glycol 400, and the like.
The present invention provides a pharmaceutical composition comprising at least one compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 as described above or a pharmaceutically acceptable form thereof.
In some embodiments of the invention, the above pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
The above pharmaceutical composition may act systematically and/or locally. For this purpose, they may be administered by a suitable route, for example by parenteral, topical, intravenous, oral, subcutaneous, intra-arterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular route or as an inhalant.
The above route of administration may be accomplished by suitable dosage forms. Dosage forms useful in the present invention include (but are not limited to): tablets, capsules, troches, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups and the like.
When administered orally, the above pharmaceutical compositions may be formulated into any orally acceptable dosage form, including, but not limited to, tablets, capsules, aqueous solutions, aqueous suspensions, and the like. The carriers used in tablets generally include lactose and corn starch, and optionally lubricants such as magnesium stearate. Carriers used in capsules generally include lactose and dried corn starch. Aqueous suspensions are typically prepared by mixing the API with suitable emulsifying and suspending agents. Optionally, some sweetening agent, flavoring agent, coloring agent, etc. can be added into the above oral preparation.
The above pharmaceutical compositions may also be administered in the form of sterile injectable preparations, including sterile injectable aqueous or oleaginous suspensions, or sterile injectable aqueous or oleaginous solutions. Among the carriers that may be used include (but are not limited to): water, ringer's solution and isotonic sodium chloride solution. In addition, the sterilized fixed oils may also be used as solvents or suspending media, such as mono-or diglycerides.
The above pharmaceutical composition may comprise 0.01mg to 1000mg of at least one compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 as described above or a pharmaceutically acceptable form thereof.
The present invention also provides a process for the preparation of the above pharmaceutical composition or a corresponding formulation thereof, which comprises combining at least one compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 or a pharmaceutically acceptable form thereof as described above with one or more pharmaceutically acceptable carriers.
[ medicine box ]
The term "kit" refers to a container.
The present invention provides a kit comprising:
a) At least one compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 above, or a pharmaceutically acceptable form thereof, as a first therapeutic agent, or a pharmaceutical composition as described above as a first pharmaceutical composition;
b) Optionally at least one other therapeutic agent as a second therapeutic agent, or a pharmaceutical composition comprising the other therapeutic agent as a second pharmaceutical composition; and
c) Optionally package and/or instructions.
The above kit may comprise 0.01mg to 1000mg of at least one compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 as described above or a pharmaceutically acceptable form thereof.
The invention also provides a method of preparing the above kit comprising combining at least one compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, with optionally at least one other therapeutic agent or a pharmaceutical composition comprising other therapeutic agent, packaging and/or instructions.
[ medical use ]
The compound of the invention can show stronger inhibition effect on TGF beta R1 and IC 50 The values can reach 100nM or less, respectively even 10nM or less, and the TGF beta R2 can be inhibited weakly, so that the TGF beta R1 inhibitor (especially the TGF beta R1 selective inhibitor) can be used. Accordingly, the present invention provides the use of a compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 as defined above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as defined above, as a TGF-beta R1 inhibitor (especially TGF-beta R1 selective Inhibitors).
In addition, the application also provides the use of the compounds of the formula I, the formula I-1, the formula I-2, the formula I-3, the formula I-4 and/or the formula I-5 or pharmaceutically acceptable forms thereof or the pharmaceutical composition in the preparation of medicaments for preventing and/or treating diseases or symptoms at least partially mediated by TGF beta R1.
The term "disease or disorder mediated at least in part by tgfβr1" refers to a disease in which pathogenesis includes at least a portion of the factors associated with tgfβr1, including, but not limited to, cancers such as lung cancer, colorectal cancer, multiple Myeloma (MM), acute Myelogenous Leukemia (AML), acute Lymphoblastic Leukemia (ALL), pancreatic cancer, liver cancer, neuroblastoma, breast cancer, ovarian cancer, melanoma, other solid tumors, or other hematological cancers.
[ method of treatment ]
The present invention provides a method for preventing and/or treating a disease mediated at least in part by rorγ, comprising the steps of: a prophylactically and/or therapeutically effective amount of a compound of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 above, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition as described above, is administered to a subject in need thereof.
The term "effective amount" refers to a dose capable of eliciting a biological or medical response from a cell, tissue, organ or organism (e.g., an individual) and sufficient to achieve a desired prophylactic and/or therapeutic effect.
The dosing regimen may be adjusted to provide the best desired response. For example, it may be administered in a single dose, it may be administered in divided doses over time, or it may be administered after a proportional decrease or increase in dose depending on the actual situation. It will be appreciated that the particular dosage regimen for any particular individual will be adjusted according to the needs and the discretion of the attendant administering or supervising the administration of the compositions.
The amount of the compound of the invention administered will depend on the individual condition, the severity of the disease or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. Generally, the effective amount is about 0.001-10000mg/kg body weight of the subject per day. In suitable cases, the effective amount is about 0.01-1000mg/kg body weight of the subject per day. About 0.01-1000mg/kg of subject body weight, typically about 0.1-500mg/kg of subject body weight, may be administered daily, every two days, or every three days. Exemplary dosing regimens are one or more times per day, or one or more times per week, or one or more times per month. The interval between individual doses may typically be daily, weekly, monthly or yearly when administered multiple times. Alternatively, the administration may be in the form of a slow release formulation, in which case a lower frequency of administration is required. The dosage and frequency of administration may vary depending on the half-life of the drug in the subject, and may also vary depending on whether it is for prophylactic or therapeutic use. In prophylactic applications, relatively low doses are administered chronically at relatively low frequency intervals; in therapeutic applications, it is sometimes desirable to administer relatively high doses at short intervals until the progression of the disease is delayed or stopped, preferably until the individual exhibits a partial or complete improvement in the symptoms of the disease, after which prophylactic applications may be employed.
The term "treatment" refers to the alleviation or elimination of a disease or condition for which it is intended. A subject is indicated to have been successfully "treated" if the subject has received a therapeutic amount of a compound of the invention, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of the invention, at least one indicator and symptom of which exhibits observable and/or detectable remission and/or improvement. It is understood that treatment includes not only complete treatment, but also less than complete treatment, but achieves some biologically or medically relevant results. In particular, "treatment" means that a compound of the invention or a pharmaceutically acceptable form thereof or a pharmaceutical composition of the invention may achieve at least one of the following effects, for example: (1) Preventing disease in animals that may be predisposed to the disease but have not undergone or displayed disease pathology or symptomology; (2) Inhibiting the disease (i.e., preventing further development of pathology and/or symptomology) in an animal experiencing or exhibiting disease pathology or symptomology; (3) Disease is ameliorated (i.e., pathology and/or symptomology is reversed) in an animal that is experiencing or exhibiting pathology or symptomology of the disease.
The term "administering" refers to the process of applying a pharmaceutically active ingredient (such as a compound of the present invention) or a pharmaceutical composition comprising a pharmaceutically active ingredient (e.g., a pharmaceutical composition of the present invention) to a subject or a cell, tissue, organ, biological fluid, etc. thereof, such that the pharmaceutically active ingredient or pharmaceutical composition is in contact with the subject or a cell, tissue, organ, biological fluid, etc. Common modes of administration include, but are not limited to, oral administration, subcutaneous administration, intramuscular administration, intraperitoneal administration, ocular administration, nasal administration, sublingual administration, rectal administration, vaginal administration, and the like.
The term "in need thereof" refers to a judgment of a physician or other caregiver as to the need of an individual or as to the impending benefit from the prevention and/or treatment process based on various factors of the physician or other caregiver in their area of expertise.
The term "individual" (or subject) refers to a human or non-human animal. The subject of the present invention includes subjects (patients) suffering from diseases and/or disorders and normal subjects. Non-human animals of the present invention include all vertebrates, such as non-mammals, e.g., birds, amphibians, reptiles, etc., and mammals, e.g., non-human primates, domestic animals, and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
[ Combined drug administration ]
The compounds of the present invention have no significant inhibitory effect on all 3 major CYP subtypes (CYP 1A2, CYP2D6 and CYP3 A4), indicating a relatively low potential for drug interactions, and therefore the above-described compounds of formula I or pharmaceutically acceptable forms thereof or the above-described pharmaceutical compositions may optionally be administered in combination with other therapeutic agents having at least some effect in the treatment of various diseases.
The present invention provides pharmaceutical combination compositions or combined preparations of the above compounds of formula I, formula I-1, formula I-2, formula I-3, formula I-4 and/or formula I-5 or pharmaceutically acceptable forms thereof or of the above pharmaceutical compositions with at least one other therapeutic agent, especially a TGF beta R1 inhibitor, for simultaneous, separate or sequential use in the prevention and/or treatment of a disease or condition.
In order to make the objects and technical solutions of the present invention more apparent, embodiments of the present invention will be described in detail with reference to examples. It will be appreciated by those skilled in the art that the following examples are illustrative of the present invention and should not be construed as limiting the scope of the invention.
The reagents or apparatus used in the examples are all conventional products commercially available. Those not specifying the specific conditions were carried out according to the conventional conditions or the conditions recommended by the manufacturer. The term "room temperature" as used herein refers to 20 ℃ ± 5 ℃. As used herein, the term "about" when used in reference to a particular value or range of values is intended to encompass the value or range of values as well as ranges of errors that are acceptable to those skilled in the art of the value or range of values, such as, for example, ±10%, ±5%, ±4%, ±3%, ±2%, ±1%, ±0.5%, etc.
In the conventional synthesis methods and examples of intermediate synthesis, the meanings of the abbreviations are shown in the following table.
The structures of the compounds described in the examples below were prepared by nuclear magnetic resonance 1 H-NMR) and/or Mass Spectrometry (MS).
Nuclear magnetic resonance 1 H-NMR) using Bruker 400MHz NMR, the solvent was deuterated methanol (CD) 3 OD), deuterated chloroform (CDCl) 3 ) Hexadeuterated dimethyl sulfoxide (DMSO-d) 6 ) The internal standard substance is Tetramethylsilane (TMS).
Abbreviations in Nuclear Magnetic Resonance (NMR) data in the following examples represent the following meanings:
s: single peak (single), d: dual peak (doubelet), t: triplet (triplet), q: quartet (quaternion), dd: double doublet (double), qd: four doublets (quatet doubelet), ddd: double doublet (double double doublet), ddt: double triplet (double double triplet), dddd: double peak (double double double doublet), m: multiple peaks (multiplet), br: broad peak (broad), J: coupling constant, hz: hertz, delta: chemical shift.
All chemical shift (delta) values are given in parts per million (ppm).
The Mass Spectrum (MS) measuring instrument uses an Agilent 6120B mass spectrometer, and the ion source is an electrospray ion source (ESI).
The examples of the present invention were purified by preparative high performance liquid chromatography (Prep-HPLC) using the methods shown below.
Method A:
chromatographic column: daisogel C 18 ODS(8μm*45mm*450mm)
Mobile phase a: acetonitrile; mobile phase B: water (containing 0.05% ammonium bicarbonate, w/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 0.0 100.0 70
50.00 50.0 50.0 70
Method B:
chromatographic column: waters Sunfire Prep C 18 OBD(5μm*19mm*150mm)
Mobile phase a: acetonitrile; mobile phase B: water (containing 0.05% trifluoroacetic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 40.0 60.0 28
16.00 90.0 10.0 28
Method C:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19 mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (containing 0.05% trifluoroacetic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 40.0 60.0 26
16.00 90.0 10.0 26
Method D:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19 mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 45.0 55.0 24
2.00 45.0 55.0 24
16.00 90.0 10.0 24
Method E:
chromatographic column: waters Sunfire Prep C 18 ODS (5 μm. Times.19 mm. Times.150 mm) mobile phase A: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 28
50.00 90.0 10.0 28
Method F:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (containing 0.05% trifluoroacetic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 30
2.00 30.0 70.0 30
16.00 90.0 10.0 30
Method G:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm. Times.19 mm. Times.150 mm) flowPhase A: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 28
4.00 30.0 70.0 28
16.00 90.0 10.0 28
Method H:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (containing 0.05% trifluoroacetic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 28
2.00 30.0 70.0 28
16.00 90.0 10.0 28
Method I:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 26
2.00 30.0 70.0 26
18.00 90.0 10.0 26
Method J:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 28
3.00 30.0 70.0 28
16.00 80.0 20.0 28
Method K:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 40.0 60.0 28
4.00 40.0 60.0 28
16.00 90.0 10.0 28
Method L:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 10.0 90.0 28
16.00 90.0 10.0 28
Method M:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 28
16.00 90.0 10.0 28
Method N:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 250 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 30
3.00 30.0 70.0 30
16.00 70.0 30.0 30
Method O:
chromatographic column: waters SunFire Prep C 18 OBD (5 μm x 19mm x 250 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 30.0 70.0 28
16.00 70.0 30.0 28
Method P:
chromatographic column: c (C) 18 OBD(5μm*19mm*150mm)
Mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 10 90 28
16.00 90.0 10.0 28
Method Q:
chromatographic column: waters XBridge Prep C 18 OBD (5 μm x 19mm x 150 mm) mobile phase a: acetonitrile; mobile phase B: water (0.05% formic acid, v/v)
Time [ min] Mobile phase A [%] Mobile phase B [%] Flow Rate [ mL/min]
0.00 15.0 85.0 28
2.00 15.0 85.0 28
20.00 90.0 10.0 28
Preparation of the Compounds
Embodiment one: synthesis of 3- (benzo [ d ] thiazol-6-yl) -5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (compound 1).
Step one: synthesis of 2-hydrazino-6-methylpyridine (Compound 1-2):
2-fluoro-6-methylpyridine (50 g,449.98 mmol) and hydrazine hydrate (36.60 g,584.97 mmol) were weighed and dissolved in isopropyl alcohol (200 mL), and the reaction system was reacted at 90℃for 168 hours. Diluting with water (2000 mL), filtering, purifying the filtrate by preparative high performance liquid chromatography (method A), and lyophilizing to obtain the title compound (16.6 g, yield 29.1%).
1 H-NMR(400MHz,DMSO-d 6 ):δ7.33(dd,J=8.3,7.2Hz,1H),7.24(s,1H),6.50(d,J=8.3Hz,1H),6.39(d,J=7.2Hz,1H),2.26(s,3H)。MS(ESI):m/z 124.1[M+H] +
Step two: synthesis of 5-hydroxy-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (Compound 1-3):
2-hydrazino-6-methylpyridine (15 g,121.80 mmol) and diethyl oxaloacetate sodium salt (51.19 g,243.59 mmol) were dissolved in toluene (200 mL), followed by addition of acetic acid (208.98 mL,3.65 mol) and the addition was completed, and the temperature was raised to 100℃for 2 hours. After cooling the reaction solution to room temperature, it was slowly poured into water (400 mL), extracted with ethyl acetate (300 ml×3), the organic phases were combined, washed with saturated brine (200 ml×1), dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give a colorless oil. Purification by flash column chromatography on silica gel (petroleum ether: ethyl acetate=1:1) afforded the title compound (28.0 g, 93.0% yield).
MS(ESI):m/z 247.9[M+H] +
Step three: synthesis of 5-chloro-4-formyl-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (Compound 1-4):
phosphorus oxychloride (1.30 g,8.49 mmol) was added to DMF (5 mL), stirred at 0deg.C for 1 hour, then a solution of 5-hydroxy-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (752 mg,2.83 mmol) in DMF (5 mL) was added to the above reaction solution, stirred at 23deg.C for 16 hours, and heated to 80deg.C for 24 hours. The reaction was cooled to room temperature and poured into water (100 mL), extracted with ethyl acetate (30 mL. Times.3), the organic phases were combined and dried and concentrated to give the crude product. Purification by preparative high performance liquid chromatography (method B) and lyophilization gave the title compound (375 mg, 39.2% yield).
MS(ESI):m/z 294.0[M+H] +
Step four: synthesis of 5-chloro-4- ((4-methoxyphenylamino) methyl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (Compound 1-5):
5-chloro-4-formyl-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (380 mg,1.29 mmol) and p-methoxyaniline (159.34 mg,1.29 mmol) were added to dichloromethane (15 mL), glacial acetic acid (0.1 mL) was added dropwise to the reaction system, after stirring at 23℃for 30 minutes, sodium Triacetoxyborohydride (STAB) (274.21 mg,1.29 mmol) was added, and stirring was continued for 16 hours. After the reaction mixture was concentrated, water (30 mL) was added, extraction was performed with ethyl acetate (20 mL. Times.3), and the organic phases were combined, dried and concentrated to give a crude product. Purification by preparative high performance liquid chromatography (method C) and lyophilization gave the title compound (127 mg, 22.3% yield).
MS(ESI):m/z 401.1[M+H] +
Step five: synthesis of 5-chloro-4- ((4-methoxyphenylamino) methyl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid (Compound 1-6):
5-chloro-4- ((4-methoxyphenylamino) methyl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (117 mg, 291.87. Mu. Mol) was added to ethanol (10 mL), tetrahydrofuran (5 mL) and water (0.66 mL), naOH (25.68 mg, 642.12. Mu. Mol) was added, and the mixture was stirred at 23℃for 16 hours. The reaction solution was concentrated, water (25 mL), ethyl acetate extraction (15 mL. Times.3) and the organic phases were combined, and concentrated by drying to give the crude title compound (107 mg, yield 98.3%) which was used in the next step without purification.
MS(ESI):m/z 373.1[M+H] +
Step six: synthesis of 3-chloro-5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (Compounds 1 to 7):
5-chloro-4- ((4-methoxyphenylamino) methyl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid (100 mg, 268.24. Mu. Mol) was added to N, N-dimethylformamide (10 mL), HATU (117.29 mg, 308.47. Mu. Mol) and N-methylmorpholine (67.83 mg, 670.59. Mu. Mol) were added in this order, and the mixture was stirred for 1 hour at 80℃under nitrogen. The reaction solution was cooled to room temperature, poured into water (100 mL), extracted with methylene chloride (30 mL. Times.3), and concentrated to give the crude title compound (15 mg, yield 15.76%).
MS(ESI):m/z 355.1[M+H] +
Step seven: synthesis of 3- (benzo [ d ] thiazol-6-yl) -5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (compound 1):
3-chloro-5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c]Pyrazol-6 (2H) -one (15 mg, 42.28. Mu. Mol), benzo [ d ]]Thiazol-6-ylboronic acid (7.57 mg, 42.28. Mu. Mol), tetrakis (triphenylphosphine) palladium (4.89 mg, 4.23. Mu. Mol) and K 2 CO 3 (5.84 mg, 42.28. Mu. Mol) was added to a single-necked flask, 1, 4-dioxane (5.00 mL) and water (1 mL), N 2 After the displacement, the temperature was raised to 90℃and stirred for 16 hours. The reaction solution was cooled to room temperature, filtered through celite and concentrated directly to give the crude product. Purification by preparative high performance liquid chromatography (method D) gave the title compound (1.65 mg, yield 8.04%) after lyophilization.
1 H-NMR(400MHz,CDCl 3 ):δ9.19(s,1H),8.17(d,J=8.2Hz,1H),8.01(d,J=5.5Hz,1H),7.79(s,1H),7.70-7.65(m,3H),7.44(d,J=8.3Hz,1H),7.16(d,J=6.8Hz,1H),6.96(d,J=8.6Hz,2H),4.87(s,2H),3.83(s,3H),2.27(s,3H)。MS(ESI):m/z 454.1[M+H] +
Embodiment two: synthesis of 3- (benzo [ d ] thiazol-6-yl) -5- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (Compound 2).
3- (benzo [ d ] thiazol-6-yl) -5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (compound 1) (10 mg, 22.05. Mu. Mol) was added to hydrobromic acid (48%, 2 mL), and the mixture was stirred at 100℃for 1 hour. The reaction was poured into water (10 mL), saturated aqueous sodium bicarbonate adjusted ph=5, extracted 3 times with dichloromethane (10 mL), the organic phases combined and dried and concentrated to give the crude product. The crude product was purified by preparative high performance liquid chromatography (method E) and lyophilized to give the formate salt of the title compound (1.66 mg, 14.7% yield).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.49(s,1H),9.46(s,1H),8.31(d,J=1.7Hz,1H),8.05(d,J=8.6Hz,1H),7.96(t,J=7.8Hz,1H),7.66-7.57(m,3H),7.38-7.31(m,2H),6.87-6.80(m,2H),5.08(s,2H),2.22(s,3H)。MS(ESI):m/z 440.1[M+H] +
Embodiment III: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -5- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (Compound 3).
Step one: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (compound 3-1):
3-chloro-5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c]Pyrazol-6 (2H) -one (30 mg, 84.56. Mu. Mol), [1,2,4]Triazolo [1,5-a ]]Pyridine-6-yl boronic acid pinacol ester (31.09 mg, 126.84. Mu. Mol), tetrakis (triphenylphosphine) palladium (9.77 mg, 8.46. Mu. Mol) and K 2 CO 3 (23.37 mg, 169.11. Mu. Mol) was added to a single-necked flask, 1, 4-dioxane (5.00 mL) and water (1 mL) were added, and the mixture was heated to 90℃after nitrogen substitution and stirred for 16 hours. The reaction solution was cooled to room temperature, filtered through celite and concentrated directly to give the crude product. The crude product was purified by preparative high performance liquid chromatography (method F) and lyophilized to give the title compound (12 mg, yield 31.1%).
MS(ESI):m/z 438.2[M+H] +
Step two: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -5- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (compound 3):
3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -5- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -4, 5-dihydropyrrolo [3,4-c ] pyrazol-6 (2H) -one (12 mg, 27.43. Mu. Mol), lithium bromide (2.38 mg, 27.43. Mu. Mol) and p-toluenesulfonic acid (4.72 mg, 27.43. Mu. Mol) were added to a microwave reaction flask, and N-methylpyrrolidone (1 mL) was added and reacted at 150℃for 1 hour. The reaction solution was cooled to room temperature, purified by preparative high performance liquid chromatography (method G), and lyophilized to give p-toluenesulfonate of the title compound (0.62 mg, yield 3.6%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.50(s,1H),9.31(d,J=1.5Hz,1H),8.60(s,1H),7.97(t,J=7.8Hz,1H),7.84(d,J=9.3Hz,1H),7.73(d,J=8.0Hz,1H),7.65-7.58(m,2H),7.51(dd,J=9.3,1.8Hz,1H),7.47(d,J=7.9Hz,2H),7.33(d,J=7.6Hz,1H),7.11(d,J=7.8Hz,2H),6.87-6.80(m,2H),5.09(s,2H),2.29(s,3H),2.17(s,3H)。MS(ESI):m/z 424.1[M+H] +
Embodiment four: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 4).
Step one: synthesis of 5-hydroxy-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid (Compound 4-1):
5-hydroxy-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester (28 g,113.25 mmol) was dissolved in a mixed solvent of tetrahydrofuran (300 mL) and water (100 mL), lithium hydroxide monohydrate (23.78 g,566.23 mmol) was further added, and the reaction was carried out at room temperature for 2 hours after the addition. After the reaction solution was diluted with water (300 mL), the organic phase was discarded, the aqueous phase ph=3 was adjusted with 2N aqueous hydrochloric acid, at this time, a large amount of white solid was precipitated, the white solid was filtered, washed with clear water (200 mL), and finally the solid obtained by filtration was dried overnight in a constant temperature oven at 50 ℃ to give the title compound (16.1 g, yield 64.9%).
MS(ESI):m/z 220.1[M+H] +
Step two: synthesis of N- (2, 2-diethoxyethyl) -4-methoxyaniline (Compound 4-3):
4-Methoxyaniline (5 g,40.6 mmol), 2-bromo-1, 1-diethoxyethane (9.6 g,48.72 mmol) was dissolved in N-methylpyrrolidone (30 mL), and potassium carbonate (7.29 g,52.78 mmol) was added in one portion, and the mixture was stirred at 95℃for 16 hours. The reaction solution was diluted with water (80 mL), extracted with ethyl acetate (50 ml×3), the combined organic phases were washed with water, dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure to give the crude product, which was purified by flash column chromatography on silica gel (petroleum ether: ethyl acetate=3:1) to give the title compound (5 g, yield 51.4%).
MS(ESI):m/z 240.1[M+H] +
Step three: synthesis of N- (2, 2-diethoxyethyl) -5-hydroxy-N- (4-methoxyphenyl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxamide (Compound 4-4):
5-hydroxy-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid (1.3 g,5.93 mmol), N- (2, 2-diethoxyethyl) -4-methoxyaniline (2.13 g,8.90 mmol) were dissolved in N, N-dimethylformamide (30 mL), HATU (3.38 g,8.90 mmol) was added in one portion, and the addition was complete and stirred for 16 hours at 25 ℃. The reaction solution was diluted with water (50 mL), extracted with ethyl acetate (50 ml×3), the combined organic phases were washed with water, dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure to give a crude product, which was purified by flash column chromatography (acetonitrile/water/trifluoroacetic acid=70/30/0.05, v/v/v), and lyophilized to give the title compound (1.5 g, yield 63.2%).
MS(ESI):m/z 441.2[M+H] +
Step four: synthesis of 3-hydroxy-6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 4-5):
n- (2, 2-Diethoxyethyl) -5-hydroxy-N- (4-methoxyphenyl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxamide (1.5 g,3.41 mmol) was dissolved in trifluoroacetic acid (20 mL), and the temperature was raised to 60℃for 4 hours. The reaction solution was evaporated to dryness under reduced pressure, methyl t-butyl ether (100 mL) was added, and the mixture was stirred vigorously for 30 minutes, whereupon a large amount of solid was precipitated, and filtered to give the title compound (1 g, yield 84.0%).
MS(ESI):m/z 349.1[M+H] +
Step five: synthesis of 6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -7-oxo-6, 7-dihydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonate (Compound 4-6):
3-hydroxy-6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (500 mg,1.44 mmol) was dissolved in tetrahydrofuran (8 mL) and triethylamine (217.86 mg,2.15 mmol) and N, N-bis (trifluoromethanesulfonyl) aniline (769 mg,2.15 mmol) were added and reacted at 25℃for 1 hour. The reaction mixture was evaporated in vacuo, methyl t-butyl ether (10 mL) was added, and the mixture was vigorously stirred for 30 minutes, whereupon a large amount of gray solid was precipitated, filtered and dried to give the title compound (500 mg, yield 72.2%).
MS(ESI):m/z 481.0[M+H] +
Step six: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 4-7):
benzo [ d ] thiazol-6-ylboronic acid (28 mg,0.16 mmol), 6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -7-oxo-6, 7-dihydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonic acid ester (50 mg,0.1 mmol) were dissolved in 1, 4-dioxane (3 mL), potassium phosphate (66 mg,0.3 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium (II) dichloride (7.62 mg, 10.41. Mu. Mol) were added in one portion, and the reaction was stirred for 16 hours after heating to 100℃in an oil bath. The reaction solution was cooled to room temperature, diluted with water (50 mL), extracted with ethyl acetate (50 ml×3), the combined organic phases were washed with water, dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure to give the crude product, which was purified by preparative thin layer chromatography (100% ethyl acetate) to give the title compound (30 mg, yield 64.4%).
MS(ESI):m/z 466.1[M+H] +
Step seven: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 4):
3- (benzo [ d ] thiazol-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (10 mg,0.021 mmol) was dissolved in hydrobromic acid (48%, 1 mL) at 25℃and heated to 100℃for 4 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was purified by preparative high performance liquid chromatography (method H) and lyophilized to give the title compound (1.02 mg, yield 10.7%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.76(s,1H),9.48(s,1H),8.32(d,J=1.7Hz,1H),8.09(d,J=8.5Hz,1H),7.95(t,J=7.8Hz,1H),7.63(d,J=8.0Hz,1H),7.46-7.32(m,2H),7.32-7.20(m,2H),7.17(d,J=7.4Hz,1H),6.95-6.80(m,2H),6.54(d,J=7.4Hz,1H),2.20(s,3H)。MS(ESI):m/z 452.1[M+H] +
Fifth embodiment: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 5).
Step one: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 5-1):
[1,2,4] triazolo [1,5-a ] pyridin-6-ylboronic acid (25 mg,0.16 mmol) and 6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -7-oxo-6, 7-dihydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonic acid ester (50 mg,0.1 mmol) were dissolved in 1, 4-dioxane (1 mL) and water (0.25 mL), potassium carbonate (29 mg,0.21 mmol) and tetrakis (triphenylphosphine) palladium (12 mg,0.01 mmol) were added in one portion, and the reaction was stirred for 2 hours after the addition was completed and the oil bath was warmed to 95 ℃. The reaction solution was cooled to room temperature, diluted with water (50 mL), extracted with ethyl acetate (50 ml×3), the combined organic phases were washed with water, dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure to give the crude product, which was purified by preparative thin layer chromatography (100% ethyl acetate) to give the title compound (20 mg, yield 44.5%).
MS(ESI):m/z 449.8[M+H] +
Step two: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (compound 5):
3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (15 mg,0.033 mmol) was dissolved in hydrobromic acid (48%, 2 mL) and reacted at 100℃for 4 hours. The reaction solution was filtered, and the filtrate was purified by preparative high performance liquid chromatography (method I) and lyophilized to give the hydrobromide salt of the title compound (1.7 mg, yield 11.8%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.23(s,1H),8.61(s,1H),7.97(t,J=7.8Hz,1H),7.88(d,J=9.2Hz,1H),7.80(d,J=8.0Hz,1H),7.58(dd,J=9.2,1.7Hz,1H),7.34(d,J=7.6Hz,1H),7.27-7.22(m,2H),7.17(d,J=7.4Hz,1H),6.94-6.84(m,2H),6.55(d,J=7.4Hz,1H),2.15(s,3H)。MS(ESI):m/z 435.8[M+H] +
Example six: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 6).
Step one: synthesis of 1- (4-methoxybenzyl) -4-nitro-1H-pyrazole (exemplary Compound 6-2):
4-nitro-1H-pyrazole (500 mg,4.42 mmol) and potassium carbonate (1.22 g,8.84 mmol) were weighed into DMF (10 mL), and then 4-methoxybenzyl chloride (1599 mg,4.86 mmol) was added dropwise, and the reaction system was reacted at 25℃for 2 hours. The reaction solution was slowly poured into water (80 mL), extracted with ethyl acetate (50 ml×3), the organic phases were combined, washed once with saturated brine (50 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, the solvent was evaporated under reduced pressure to give a crude product as a colorless oil, and the title compound (985 mg, yield 95.4%) was purified by silica gel column chromatography (petroleum ether: ethyl acetate=2:1).
MS(ESI):m/z 234.3[M+H] +
Step two: synthesis of 1- (4-methoxybenzyl) -1H-pyrazol-4-amine (Compound 6-3):
1- (4-methoxybenzyl) -4-nitro-1H-pyrazole (985 mg,4.22 mmol) was dissolved in methanol (20 mL), pd/C (50 mg) was added, and the reaction system was replaced with a hydrogen balloon three times, followed by reaction at 25℃for 15 hours under a hydrogen atmosphere. The reaction solution was filtered, and the cake was rinsed with methanol (20 mL), and then the filtrate was concentrated under reduced pressure to give a crude title compound (805 mg, yield 93.8%) which was used in the next step without purification.
MS(ESI):m/z 204.0[M+H] +
Step three: synthesis of N- (2, 2-diethoxyethyl) -1- (4-methoxybenzyl) -1H-pyrazol-4-amine (Compound 6-4):
1- (4-methoxybenzyl) -1H-pyrazol-4-amine (700 mg,3.44 mmol) and 2-bromo-1, 1-diethoxy-ethane (1.02 g,5.17 mmol) were dissolved in N-methylpyrrolidone (10 mL), followed by addition of potassium carbonate (951.98 mg,6.89 mmol) and potassium iodide (57.17 mg, 344.42. Mu. Mol), and after the addition, the temperature was raised to 80℃for reaction for 15 hours. The reaction solution was cooled to room temperature, then, it was slowly poured into water (50 mL), extracted with ethyl acetate (50 mL. Times.3), the combined organic phases were washed with saturated brine (50 mL), the organic phases were dried over anhydrous sodium sulfate, the drying agent was filtered off, and the solvent was evaporated under reduced pressure to give a crude colorless oily product. The crude product was purified by column chromatography on silica gel (petroleum ether: ethyl acetate=1:4) to give the title compound (425 mg, yield 38.6%).
MS(ESI):m/z 320.2[M+H] +
Step four: synthesis of N- (2, 2-diethoxyethyl) -5-hydroxy-N- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxamide (Compound 6-5):
5-hydroxy-1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxylic acid (80 mg, 364.97. Mu. Mol) and N- (2, 2-diethoxyethyl) -1- (4-methoxybenzyl) -1H-pyrazol-4-amine (140 mg, 437.96. Mu. Mol) were dissolved in N-dimethylformamide (2 mL), HATU (166.43 mg, 437.96. Mu. Mol) was then added, and the reaction system was reacted at 25℃for 15 hours. The reaction solution was slowly poured into water (15 mL), extracted with ethyl acetate (15 mL. Times.3), the organic phases were combined, washed once with saturated brine (20 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, the filtrate was concentrated under reduced pressure to give a crude colorless oil, and the crude oil was purified by C 18 Column chromatography purification (acetonitrile/water/trifluoro)Acetic acid=68%/32%/0.05%) to give the title compound (82 mg, yield 43.3%).
MS(ESI):m/z 521.3[M+H] +
Step five: synthesis of 3-hydroxy-6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 6-6):
n- (2, 2-Diethoxyethyl) -5-hydroxy-N- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -1- (6-methylpyridin-2-yl) -1H-pyrazole-3-carboxamide (80 mg, 153.68. Mu. Mol) was dissolved in toluene (4 mL), followed by addition of p-toluenesulfonic acid (26.46 mg, 153.68. Mu. Mol), and after the addition, the reaction was carried out at 110℃for 4 hours. The reaction solution was cooled to room temperature, evaporated to dryness under reduced pressure to give a crude product, which was purified by preparative high performance liquid chromatography (method J) and lyophilized to give the title compound (15 mg, yield 22.8%).
1 H-NMR(400MHz,DMSO-d 6 ):δ8.41(s,1H),8.12(d,J=8.3Hz,1H),7.89(s,2H),7.36-7.26(m,3H),7.22(d,J=7.5Hz,1H),6.97-6.89(m,2H),6.57(d,J=7.2Hz,1H),5.30(s,2H),3.74(s,3H),2.54(s,3H)。MS(ESI):m/z 429.1[M+H] +
Step six: synthesis of 6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -7-oxo-6, 7-dihydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonate (Compound 6-7):
3-hydroxy-6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (15 mg, 33.26. Mu. Mol) and triethylamine (5.67 mg, 56.02. Mu. Mol) were dissolved in tetrahydrofuran (1 mL), and then N, N-bis (trifluoromethanesulfonyl) aniline (15.01 mg, 42.01. Mu. Mol) was added, and the reaction was carried out at room temperature for 0.5 hours after the addition. The reaction mixture was diluted with dichloromethane (15 mL), washed once with water (5 mL) and saturated brine (5 mL), and the organic phase was dried over anhydrous sodium sulfate, and the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give the title compound (15 mg, yield 95.5%).
MS(ESI):m/z 561.2[M+H] +
Step seven: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 6-8):
6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -7-oxo-6, 7-dihydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonate (15 mg, 26.76. Mu. Mol) and benzo [ d ] thiazol-6-ylboronic acid pinacol ester (10.48 mg, 40.14. Mu. Mol) were dissolved in 1, 4-dioxane (2 mL), and tetrakis (triphenylphosphine) palladium (3.09 mg, 2.68. Mu. Mol) and potassium carbonate (7.40 mg, 53.52. Mu. Mol) were then added, and the reaction system was replaced with nitrogen 3 times and the oil bath was warmed to 95℃under nitrogen atmosphere to react for 4 hours. The reaction solution was cooled to room temperature, slowly poured into water (10 mL), extracted with ethyl acetate (10 ml×3), the organic phases were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered off the drying agent, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by preparative high performance liquid chromatography (method K), and lyophilized to give the title compound (10 mg, yield 68.5%).
MS(ESI):m/z 546.2[M+H] +
Step eight: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 6):
3- (benzo [ d ] thiazol-6-yl) -6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (10 mg, 18.33. Mu. Mol) was dissolved in trifluoroacetic acid (1 mL), and the temperature was raised to 80℃for reaction for 15 hours. The reaction solution was concentrated under reduced pressure to give a crude product, which was purified by preparative high performance liquid chromatography (method L) and lyophilized to give the trifluoroacetate salt of the title compound (2.12 mg, 21.0%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.48(s,1H),8.32(d,J=1.7Hz,1H),8.13-8.05(m,3H),7.95(t,J=7.8Hz,1H),7.63(d,J=8.0Hz,1H),7.39(dd,J=17.2,7.7Hz,3H),6.61(d,J=7.5Hz,1H),2.20(s,3H)。MS(ESI):m/z 426.1[M+H] +
Embodiment seven: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (compound 7).
3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one trifluoroacetate (20 mg, 37.07. Mu. Mol) and triethylamine (11.25 mg, 111.22. Mu. Mol) were dissolved in dichloromethane (2 mL), methanesulfonyl chloride (607.88 mg, 44.49. Mu. Mol) was added, and the reaction was carried out at room temperature for 2 hours. After the reaction solution was diluted with methylene chloride (30 mL), each was washed with water (15 mL) and saturated brine (15 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, the filtrate was concentrated under reduced pressure to obtain a crude product, which was purified by preparative high performance liquid chromatography (method M), and lyophilized to obtain the title compound (3.36 mg, yield 18.0%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.48(s,1H),8.85(s,1H),8.49(s,1H),8.34(d,J=1.7Hz,1H),8.10(d,J=8.5Hz,1H),7.96(t,J=7.8Hz,1H),7.64(d,J=8.0Hz,1H),7.55(d,J=7.5Hz,1H),7.41-7.33(m,2H),6.68(d,J=7.5Hz,1H),3.65(s,3H),2.21(s,3H)。MS(ESI):m/z 503.7[M+H] +
Example seven-a: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (compound 7) hydrochloride.
Concentrating the filtrate from example seven under reduced pressure, using C 18 Reverse phase column purification (acetonitrile/water/hcl=45%/55%/0.05%) to obtain a preparation, freeze drying, and changing the mobile phase system (acetonitrile/water/hcooh=45%/55%/0.05%) to reuse C 18 Purification by reverse phase column gave the title compound as the hydrochloride salt after lyophilization.
1 H-NMR(400MHz,DMSO-d 6 ):δ9.48(s,1H),8.85(d,J=0.7Hz,1H),8.49(d,J=0.7Hz,1H),8.34(d,J=1.8Hz,1H),8.09(d,J=8.5Hz,1H),7.96(t,J=7.8Hz,1H),7.64(d,J=8.0Hz,1H),7.55(d,J=7.5Hz,1H),7.41-7.31(m,2H),6.68(d,J=7.5Hz,1H),3.66(s,3H),2.20(s,3H)。MS(ESI):m/z 503.7[M+H] +
Example eight: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 14).
Step one: synthesis of 3-hydroxy-6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-tetrahydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 14-1):
3-hydroxy-6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-C ] pyridin-7 (6H) -one (180 mg,0.52 mmol) was dissolved in methanol (20 mL), pd/C (30 mg) was added under nitrogen protection, the air was replaced three times with hydrogen, and the reaction was stirred at 25℃under hydrogen for 24 hours. Filtration through celite, concentration of the filtrate under reduced pressure afforded the crude product, purification by preparative high performance liquid chromatography (method N), and lyophilization afforded the title compound (30 mg, 16.4% yield).
MS(ESI):m/z 351.1[M+H] +
Step two: synthesis of 6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -7-oxo-4, 5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonate (Compound 14-2):
3-hydroxy-6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-tetrahydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (30 mg,0.086 mmol) and triethylamine (13 mg,0.13 mmol) were dissolved in tetrahydrofuran (2 mL) and N, N-bis (trifluoromethanesulfonyl) aniline (46 mg,0.13 mmol) was added in one portion and reacted at 25℃for 1 hour. The reaction solution was dried with nitrogen, methyl tert-butyl ether (6 mL) was added, a large amount of white solid was precipitated, stirred at 25℃for 10 minutes, filtered, and the cake was rinsed with methyl tert-butyl ether (5 mL) and dried to give the title compound (20 mg, yield 47.2%).
MS(ESI):m/z 493.0[M+H] +
Step three: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 14-3):
benzo [ d ] thiazol-6-ylboronic acid pinacol ester (16 mg,0.062 mmol) and 6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -7-oxo-4, 5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridin-3-yl triflate (20 mg,0.041 mmol) were dissolved in 1, 4-dioxane (2 mL) and water (0.5 mL), potassium carbonate (11 mg,0.083 mmol) and tetrakis (triphenylphosphine) palladium (5 mg,0.04 mmol) were added in one portion and the mixture was stirred at 95℃for 2 hours. The reaction solution was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (30 ml×3), the combined organic phases were washed with water, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product, which was purified by preparative thin layer chromatography (petroleum ether: ethyl acetate=1:1) to give the title compound (10 mg, yield 34.5%).
MS(ESI):m/z 467.8[M+H] +
Step four: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 14):
3- (benzo [ d ] thiazol-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (20 mg,0.043 mmol) was dissolved in aqueous hydrobromic acid (48%, 1 mL) and reacted at 100℃for 3 hours. The reaction solution was cooled to room temperature, directly filtered, purified by preparative high performance liquid chromatography (method O) and lyophilized to give the hydrobromide salt of the title compound (1.44 mg, yield 7.4%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.46(s,1H),8.23(d,J=1.7Hz,1H),8.06(d,J=8.5Hz,1H),7.89(t,J=7.8Hz,1H),7.52(d,J=7.9Hz,1H),7.32(dd,J=8.5,1.8Hz,1H),7.27(d,J=7.6Hz,1H),7.24-7.14(m,2H),6.87-6.76(m,2H),3.95(t,J=6.4Hz,2H),2.99(t,J=6.5Hz,2H),2.14(s,3H)。MS(ESI):m/z 454.1[M+H] +
Example nine: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 15).
Step one: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 15-1):
6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -7-oxo-4, 5,6, 7-tetrahydro-2H-pyrazolo [3,4-c]Pyridin-3-yl triflate (55 mg, 114.01. Mu. Mol) and [1,2,4]]Triazolo [1,5-a ] ]Pyridine-6-yl-boric acid (27.86 mg, 171.01. Mu. Mol) was dissolved in 1, 4-dioxane (2 mL), and tetrakis (triphenylphosphine) palladium (13.17 mg, 11.40. Mu. Mol) and potassium carbonate (31.51 mg, 228.01. Mu. Mol) were then added, and the reaction system was replaced with nitrogen 3 times after the addition, and reacted at 95℃for 15 hours under a nitrogen atmosphere. The reaction solution was cooled to room temperature, dichloromethane (30 mL) was added, then washed once with clear water (10 mL) and saturated brine (10 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by C 18 Column chromatography purification (acetonitrile/water/trifluoroacetic acid=50/50/0.05, v/v/v), saturated sodium bicarbonate solution ph=8, dichloromethane extraction (20 ml×2), washing the combined organic phases with saturated brine (10 mL), drying over anhydrous sodium sulfate, filtering off the drying agent, and concentrating the filtrate under reduced pressure gave the title compound (31 mg, yield 60.3%).
MS(ESI):m/z 452.2[M+H] +
Step two: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-hydroxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (compound 15):
3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (4-methoxyphenyl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (31 mg, 68.66. Mu. Mol) was dissolved in hydrobromic acid solution (48%, 1 mL) and reacted at 100℃for 3 hours. After the reaction solution was cooled to room temperature, it was then filtered, and the filtrate was purified by preparative high performance liquid chromatography (method P) and lyophilized to give the hydrobromide salt of the title compound (3.77 mg, yield 10.6%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.53(s,1H),9.23(d,J=1.5Hz,1H),8.58(s,1H),7.92(t,J=7.8Hz,1H),7.84(d,J=9.2Hz,1H),7.68(d,J=8.1Hz,1H),7.50(dd,J=9.3,1.7Hz,1H),7.26(d,J=7.6Hz,1H),7.22-7.15(m,2H),6.84-6.77(m,2H),3.96(t,J=6.5Hz,2H),3.00(t,J=6.5Hz,2H),2.11(s,3H)。MS(ESI):m/z 438.2[M+H] +
Example ten: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 8).
Step one: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 8-1):
6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -7-oxo-6, 7-dihydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonate (156 mg, 278.32. Mu. Mol) and [1,2,4] triazolo [1,5-a ] pyridin-6-ylboronic acid (68 mg, 417.48. Mu. Mol) were dissolved in a mixed solvent of 1, 4-dioxane (6 mL) and water (2 mL), and then dichlorodi-tert-butyl- (4-dimethylaminophenyl) palladium (II) (20 mg, 27.83. Mu. Mol) and potassium carbonate (77 mg, 556.64. Mu. Mol) were added, and the reaction system was replaced with nitrogen 3 times, and the oil bath was warmed to 95℃under nitrogen atmosphere and reacted for 1 hour. The reaction solution was cooled to room temperature, diluted with dichloromethane (30 mL), then washed once with water (10 mL) and saturated brine (10 mL) in this order, the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give a crude product. The crude product was purified by silica gel column chromatography (dichloromethane: methanol=20:1) to give the title compound (82 mg, yield 52.8%).
MS(ESI):m/z 530.2[M+H] +
Step two: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (compound 8):
3- ([ 1,2, 4)]Triazolo [1,5-a ]]Pyridin-6-yl) -6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c]Pyridin-7 (6H) -one (82 mg, 147.11. Mu. Mol) was dissolved in trifluoroacetic acid (2 mL) and reacted at 80℃for 15 hours. Will beConcentrating the reaction solution under reduced pressure to obtain a crude product, and passing through C 18 Purification by column chromatography (acetonitrile/water/trifluoroacetic acid=33%/67%/0.05%) afforded the trifluoroacetate salt of the title compound (45 mg, yield 58.5%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.25(t,J=1.2Hz,1H),8.61(s,1H),8.09(s,2H),7.98(t,J=7.8Hz,1H),7.88(d,J=9.3Hz,1H),7.80(d,J=8.1Hz,1H),7.57(dd,J=9.2,1.7Hz,1H),7.43(d,J=7.5Hz,1H),7.35(d,J=7.6Hz,1H),6.63(d,J=7.5Hz,1H),2.15(s,3H)。MS(ESI):m/z 410.1[M+H] +
Example eleven: synthesis of 3- ([ 1,2,4] triazolo [1,5-a ] pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c ] pyridin-7 (6H) -one (Compound 9).
3- ([ 1,2, 4)]Triazolo [1,5-a ]]Pyridin-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2H-pyrazolo [3,4-c]The trifluoroacetate salt of pyridin-7 (6H) -one (42 mg, 80.24. Mu. Mol) and triethylamine (24 mg, 240.72. Mu. Mol) were dissolved in methylene chloride (2 mL), methanesulfonyl chloride (11 mg, 96.29. Mu. Mol) was added, and the reaction was completed at room temperature for 2 hours. After the reaction solution was diluted with dichloromethane (30 mL), it was washed once with water (10 mL) and saturated brine (10 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to obtain a crude product. The crude product is C 18 Column chromatography purification (acetonitrile/water/trifluoroacetic acid=41/59/0.05, v/v/v) afforded the trifluoroacetate salt of the title compound (24 mg, yield 49.7%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.26(t,J=1.3Hz,1H),8.86(s,1H),8.62(s,1H),8.50(s,1H),7.98(t,J=7.8Hz,1H),7.89(dd,J=9.2,0.9Hz,1H),7.81(d,J=8.0Hz,1H),7.61-7.53(m,2H),7.35(d,J=7.6Hz,1H),6.72(d,J=7.5Hz,1H),3.66(s,3H),2.15(s,3H)。MS(ESI):m/z 488.1[M+H] +
Embodiment twelve: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (compound 16).
Step one: synthesis of 3-hydroxy-6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 16-1):
3-hydroxy-6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2H-pyrazolo [3,4-c]Pyridin-7 (6H) -one (80 mg, 186.72. Mu. Mol) was dissolved in a mixed solvent of ethyl acetate (10 mL) and methanol (10 mL), pd/C (10 mg) was added under nitrogen protection, the reaction system was replaced with hydrogen three times, and the reaction was stirred under a hydrogen atmosphere at 25℃for 24 hours. Filtering the reaction solution with diatomite, evaporating the filtrate under reduced pressure to obtain a crude product, and performing C 18 Column chromatography purification (acetonitrile/water/trifluoroacetic acid=55/45/0.05, v/v/v) afforded the trifluoroacetate salt of the title compound (22 mg, yield 38.3%).
MS(ESI):m/z 431.1[M+H] +
Step two: synthesis of 6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -7-oxo-4, 5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridin-3-yl trifluoromethanesulfonate (Compound 16-2):
3-hydroxy-6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (35 mg, 64.28. Mu. Mol) of trifluoroacetate and triethylamine (19 mg, 192.84. Mu. Mol) were dissolved in methylene chloride (2 mL), and then N, N-bis (trifluoromethanesulfonyl) aniline (34 mg, 96.42. Mu. Mol) was added, and the addition was completed, and the reaction was carried out at room temperature for 0.5 hours. After the reaction solution was diluted with methylene chloride (20 mL), each was washed once with water (10 mL) and saturated brine (10 mL), and the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give the title compound (33 mg, yield 91.6%) which was used in the next step without purification.
MS(ESI):m/z 563.3[M+H] +
Step three: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 16-3):
6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -7-oxo-4, 5,6, 7-tetrahydro-2H-pyrazolo [3,4-c]Pyridin-3-yl triflate (33 mg, 58.71. Mu. Mol) and benzo [ d ]]The thiazole-6-yl boronic acid pinacol ester (25 mg, 90.00. Mu. Mol) was dissolved in a mixed solvent of 1, 4-dioxane (2 mL) and water (0.5 mL), followed by addition of di-tert-butyl (4-dimethylaminophenyl) phosphorus palladium (II) dichloride (4 mg, 5.87. Mu. Mol) and potassium carbonate (16 mg, 117.42. Mu. Mol), displacement of the reaction system with nitrogen was completed 3 times, and the temperature of the oil bath was raised to 95℃under nitrogen atmosphere for reaction for 1 hour. The reaction solution was cooled to room temperature, diluted with dichloromethane (30 mL), washed once with water (10 mL) and saturated brine (10 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give a crude product, which was taken up in C 18 Column chromatography purification (acetonitrile/water/ammonium bicarbonate=56/44/0.05, v/v/v) afforded the title compound (30 mg, yield 88.2%).
MS(ESI):m/z 548.2[M+H] +
Step four: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 16):
3- (benzo [ d)]Thiazol-6-yl) -6- (1- (4-methoxybenzyl) -1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c]Pyridin-7 (4H) -one (30 mg, 54.84. Mu. Mol) was dissolved in trifluoroacetic acid (2 mL) and reacted at 80℃for 15 hours. Concentrating the reaction solution under reduced pressure to obtain crude product, and passing through C 18 Column chromatography purification (acetonitrile/water/trifluoroacetic acid=38/62/0.05, v/v/v) afforded the trifluoroacetate salt of the title compound (15 mg, 44.6%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.46(s,1H),8.24(d,J=1.7Hz,1H),8.06(d,J=8.5Hz,1H),7.97(s,2H),7.90(t,J=7.8Hz,1H),7.53(d,J=8.0Hz,1H),7.37-7.24(m,2H),4.09(t,J=6.8Hz,2H),3.02(t,J=6.5Hz,2H),2.14(s,3H)。MS(ESI):m/z 428.1[M+H] +
Embodiment thirteen: synthesis of 3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1- (methylsulfonyl) -1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c ] pyridin-7 (4H) -one (Compound 17).
3- (benzo [ d)]Thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -5, 6-dihydro-2H-pyrazolo [3,4-c]The trifluoroacetate salt of pyridin-7 (4H) -one (12 mg, 28.07. Mu. Mol) and triethylamine (8 mg, 84.21. Mu. Mol) were dissolved in methylene chloride (2 mL), methanesulfonyl chloride (5 mg, 42.11. Mu. Mol) was added thereto, and the reaction was completed at room temperature for 2 hours. The reaction mixture was diluted with dichloromethane (30 mL), washed once with water (15 mL) and saturated brine (15 mL), the organic phase was dried over anhydrous sodium sulfate, the drying agent was filtered off, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by C 18 Column chromatography purification (acetonitrile/water/trifluoroacetic acid=41/59/0.05, v/v/v) afforded the trifluoroacetate salt of the title compound (9.16 mg, yield 52.6%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.46(s,1H),8.61(s,1H),8.41(s,1H),8.26(d,J=1.7Hz,1H),8.06(d,J=8.5Hz,1H),7.90(t,J=7.8Hz,1H),7.53(d,J=8.0Hz,1H),7.37-7.26(m,2H),4.15(d,J=6.5Hz,2H),3.58(s,3H),3.04(t,J=6.5Hz,2H),2.15(s,3H)。MS(ESI):m/z 506.1[M+H] +
Fourteen examples: synthesis of 3- (benzo [ d ] thiazol-6-yl) -6- (1-methyl-1H-pyrazol-4-yl) -2- (6-methylpyridin-2-yl) -2, 6-dihydro-7H-pyrazolo [3,4-c ] pyridin-7-one (compound 25).
3- (benzo [ d ] thiazol-6-yl) -2- (6-methylpyridin-2-yl) -6- (1H-pyrazol-4-yl) -2, 6-dihydro-7H-pyrazolo [3,4-c ] pyridin-7-one (compound 6,15.00mg, 27.80. Mu. Mol), cesium carbonate (27.18 mg, 84.31. Mu. Mol), methyl 4-methylbenzenesulfonate (10.36 mg, 55.61. Mu. Mol) were dissolved in N, N-dimethylformamide (2 mL), and the temperature was raised to 80℃and the reaction was carried out for 0.5H. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated and purified by preparative high performance liquid chromatography (method Q) to give the title compound (8.26 mg, yield 65.81%).
1 H-NMR(400MHz,DMSO-d 6 ):δ9.48(s,1H),8.33(d,J=1.6Hz,1H),8.27(s,1H),8.09(d,J=8.5Hz,1H),7.95(t,J=7.8Hz,1H),7.85-7.81(m,1H),7.63(d,J=8.0Hz,1H),7.40(d,J=7.5Hz,1H),7.39-7.37(m,1H),7.37-7.35(m,1H),6.61(d,J=7.5Hz,1H),3.91(s,3H),2.20(s,3H)。ESI-MS:m/z440.0[M+H] +
Pharmacological Activity test
Test example one: in vitro enzymatic Activity inhibition assay (TGF. Beta.R1).
The experimental method comprises the following steps: according to ADP-Glo TM Description of kinase assay kit (Promega, cat#v9102) the inhibition of tgfβr1 enzyme activity by the compounds of the invention was determined as follows:
after preincubation of TGF-beta R1 enzyme with test compounds (1000 nM, 100nM, 10 nM) at 30℃for 30min, respectively, TGF-beta R1 peptide and Adenosine Triphosphate (ATP) were added to initiate a reaction. After incubation at 30℃for 3h ADP-Glo was added TM The reagent is incubated for 90min at room temperature, and then kinase detection reagent is added. After incubation for 30min at room temperature, chemiluminescent signal values were detected. The percent inhibition of compounds at different concentrations was calculated using the solvent set (DMSO) as a negative control, the buffer set (without tgfβr1enzyme) as a blank according to the following formula:
percentage inhibition = (1- (chemiluminescent signal value of compound at different concentration-chemiluminescent signal value of blank)/(chemiluminescent signal value of negative control-chemiluminescent signal value of blank)) × 100%;
when the percent inhibition was between 30-80%, the half Inhibition Concentration (IC) of the compound was calculated according to the following formula 50 ) Or range of:
IC 50 =x (1-percent inhibition (%))/percent inhibition (%), wherein: x is the test concentration of the compound.
The experimental results are shown in table 1 below.
TABLE 1 inhibitory Activity of the Compounds of the invention against TGF-beta R1
Examples numbering IC for TGF beta R1 50 (nM)
Example 1 106.05±12.93
Example two 35.37±1.65
Example III 96.29±27.99
Example IV 25.14±4.77
Example five 16.87±2.41
Example six 8.43±5.39
Example seven 1.89±0.29
Example eight 1.98±0.56
Example nine 9.39±0.22
Examples ten 16.76±2.57
Example eleven 3.78±0.29
Example twelve 2.95±0.12
Example thirteen 3.38±0.71
Examples fourteen 7.21±2.08
As can be seen from Table 1, the compounds of the present invention have a significant inhibitory effect on TGF-beta R1.
Test example two: in vitro enzymatic Activity inhibition assay (TGF. Beta.R2).
The experimental method comprises the following steps: according to ADP-Glo TM Description of kinase assay kit (Promega, cat#v9102) the inhibition of tgfβr2 enzyme activity by the compounds of the invention was determined as follows:
after preincubation of tgfβr2 enzyme with test compounds (1000 nM, 100nM, 10 nM) at 30 ℃ for 30min, respectively, myelin Basic Protein (MBP) and Adenosine Triphosphate (ATP) were added to initiate a reaction. After incubation at 30℃for 3h ADP-Glo was added TM The reagent is incubated for 90min at room temperature, and then kinase detection reagent is added. After incubation for 30min at room temperature, the chemiluminescent signal value was measured. The percent inhibition of compounds at different concentrations was calculated using the solvent set (DMSO) as a negative control, the buffer set (without tgfβr2enzyme) as a blank according to the following formula:
percentage inhibition = (1- (chemiluminescent signal value of compound at different concentration-chemiluminescent signal value of blank)/(chemiluminescent signal value of negative control-chemiluminescent signal value of blank)) × 100%;
when the percent inhibition was between 30-80%, the half Inhibition Concentration (IC) of the compound was calculated according to the following formula 50 ) Or range of:
IC 50 =x (1-percent inhibition (%))/percent inhibition (%), wherein: x is the test concentration of the compound.
The experimental results are shown in table 2 below.
TABLE 2 inhibition of TGF-beta R2 enzyme Activity by Compounds of the invention
Examples numbering IC for TGF beta R2 50 (nM)
Example two >1000
Example IV >1000
Example five >1000
Example seven 247.79±43.92
Example eight 971.84±140.65
Example nine >1000
Examples ten >1000
Example eleven >1000
Example twelve 289.96±30.34
Example thirteen 336.28±26.92
As can be seen from Table 2, the compounds of the present invention have a weak inhibitory activity against TGF-beta R2.
As can be seen from tables 1 and 2, the compounds of the present invention have selective inhibition of TGF-beta R1.
Test example three: in vitro cell activity inhibition assay.
The experimental method comprises the following steps: according to Bright-Glo TM Description of luciferase assay kit (Promega, cat#E2620) the inhibition of HEK293-SBE cells TGF beta/Smad signaling pathway by the compounds of the invention was determined as follows:
HEK293-SBE cells (Bps bioscience, cat # 60653) were added to 96-well plates (MEM medium with 10% FBS), 3 ten thousand per well, 37℃at 5% CO 2 Culturing overnight. The medium was changed to 0.5% fbs medium and 0.5% fbs medium diluted test compound was added with a final maximum concentration of 10 μm, 4-fold dilution of compound, total 8 concentration gradients. After 4-5 hours of incubation, 10. Mu.l TGF-beta was added. The final concentration of TGF-beta was 0.5ng/ml. Mu.l of medium was added instead of TGF beta as negative control. No test compound was added to the blank and tgfβ was added. Bright Glo reagent was added to each well and chemiluminescent signal values were read on a microplate reader.
The percent inhibition of compounds at different concentrations was calculated according to the following formula:
percentage inhibition = (1- (chemiluminescent signal value of test compound-chemiluminescent signal value of blank)/(chemiluminescent signal value of negative control-chemiluminescent signal value of blank)) ×100%;
the percent inhibition of compounds at different concentrations was plotted against compound concentration, and IC was calculated by fitting a curve according to a four parameter model, by the following formula 50 Value:
y=Min+(Max-Min)/(1+(x/IC 50 ) Taken together, ≡α (-Hillslope)), wherein: y is the percent inhibition; max and Min are the maximum value and the minimum value of the fitting curve respectively; x is the logarithmic concentration of the compound; and Hillslope is the slope of the curve.
The experimental results are shown in table 3 below.
TABLE 3 inhibition of HEK293-SBE cell TGF beta/Smad signaling pathway by Compounds of the invention
Examples numbering IC 50 (nM)
Example two 37.93±4.88
Example IV 114.40±22.05
Example five 81.55±11.64
Example six 25.11±2.34
Example seven 43.27±4.84
Example eight 15.52±1.57
Example nine 112.30±19.45
Examples ten 29.70±3.84
Example eleven 17.37±1.88
Example twelve 4.08±0.36
Example thirteen 8.43±0.48
Examples fourteen 49.58±2.35
As can be seen from Table 3, the compound HEK293-SBE cell TGF beta/Smad signal pathway of the invention has a remarkable inhibition effect.
Test example four: biochemical hERG inhibition assay.
1. Test system:
the kit comprises: preconductor TM hERG fluorescence polarization detection kit (Thermo Fisher, cat#PV5365), the kit contains the following components: positive control compound hERG potassium channel blocker E4031; hERG cell membrane; affinity Tracer tracker; and hERG buffer.
2. Test parameters:
hERG concentration: 1×; tracer concentration: 1nM; incubation time: 2h; BMG PHERAstar FS FP.
3. The test method comprises the following steps:
the test was performed according to the kit instructions, the steps were as follows:
test group: 10. Mu.M and 1. Mu.M of the test compound was added to a microplate containing hERG cell membranes, a Tracer having high hERG affinity was added to each well, and after incubating the microplate at room temperature for 2 hours, the change in fluorescence polarization (excitation wavelength: 540nm; emission wavelength: 590 nm) was detected using a multifunctional microplate reader.
Positive control group: the test compound was replaced with 30. Mu.M positive control compound E4031, and the experimental procedure was the same as that of the test group.
Blank control group: the test compounds were replaced with hERG buffer and hERG cell membranes were not added, and the experimental procedure was the same as for the test group.
4. And (3) data processing:
based on the data ratio, the percent inhibition (%) of the compounds of the invention against hERG at various concentrations was calculated and the half Inhibition Concentration (IC) of the compounds was determined 50 ) Is not limited in terms of the range of (a).
Percentage inhibition (%) = (1- (fluorescence polarization value of test compound-fluorescence polarization value of positive control)/(fluorescence polarization value of blank control-fluorescence polarization value of positive control)) ×100%;
5. experimental results:
inhibition of hERG by compounds was determined using the methods described above and the results are shown in table 4 below.
TABLE 4 hERG inhibition assay results
Examples numbering IC 50 (μM)
Example two >10
Example IV >10
Example five >10
Example six >10
Example seven >10
Example eight >10
Example nine >10
Examples ten >10
Example eleven >10
Example twelve >10
Example thirteen >10
Examples fourteen >10
The test results show that the compound of the invention has low affinity with hERG and has affinity with hERGIC competing with sex Tracer 50 All greater than 10 μm, demonstrating that the compounds of the invention have lower risk of cardiotoxicity associated with hERG ion channels.
Test example five: CYP enzyme (cytochrome P450) inhibition assay.
1. Test system:
P450-Glo TM CYP1A2 screening System (Promega, cat#V9770);
P450-Glo TM CYP2D6 screening System (Promega, cat#V9890);
P450-Glo TM CYP3A4 screening System (Promega, cat#V9920).
2. Test instrument:
BMG PHERAstar FS Luminescent。
3. the test method comprises the following steps:
the test was performed according to the kit instructions, respectively, as follows:
3.1. inhibition of CYP1 A2:
Test group: the test compounds with different concentrations are added into a microplate, and Luciferin-ME (100 mu M) and K are added into each well 3 PO 4 (100 mM) and CYP1A2 (0.01 pmol/. Mu.L) were pre-incubated at room temperature for 10min, followed by addition of NADPH regeneration system, reaction at room temperature for 30min, final addition of an equal volume of detection buffer, incubation at room temperature for 20min, and chemiluminescent detection.
Negative control group: the experimental procedure was the same as for the test group, but without the test compound.
Blank control group: the experimental procedure was the same as for the test group, except that no test compound was added and CYP1A2 was replaced with CYP1A2 membrane (0.01 pmol/. Mu.L).
3.2. Inhibition of CYP2D 6:
test group: adding test compounds with different concentrations into microplates, adding Luciferin-ME EGE (3 μm), K into each well 3 PO 4 (100 mM) and CYP2D6 (5 nM) were pre-incubated at room temperature for 10min, followed by addition of NADPH regeneration system, reaction at 37℃for 30min, and finally addition of an equal volume of detection buffer, incubation at room temperature for 20min, followed by chemiluminescent detection.
Negative control group: the experimental procedure was the same as for the test group, but without the test compound.
Blank control group: the experimental procedure was the same as for the test group, but without the test compound and with CYP2D6 membrane (5 nM) instead of CYP2D6.
3.3. Inhibition of CYP3 A4:
test group: the test compounds with different concentrations are added into a microplate, and Luciferin-IPA (3 mu M) and K are added into each well 3 PO 4 (100 mM) and CYP3A4 (2 nM) were pre-incubated at room temperature for 10min, followed by addition of NADPH regeneration system, reaction at room temperature for 30min, final addition of an equal volume of detection buffer, incubation at room temperature for 20min, and chemiluminescent detection.
Negative control group: the experimental procedure was the same as for the test group, but without the test compound.
Blank control group: the experimental procedure was the same as for the test group, but without the test compound and with CYP3A4 membrane (2 nM) instead of CYP3A4.
4. And (3) data processing:
percentage inhibition (%) = (1- (chemiluminescent signal value of test compound-chemiluminescent signal value of blank)/(chemiluminescent signal value of negative control-chemiluminescent signal value of blank)) ×100%;
based on the inhibition of CYP enzyme by compounds at different concentrations, half inhibition concentration (IC 50 ) Or range of:
IC 50 =x (1-percent inhibition (%))/percent inhibition (%), wherein: x is the test concentration of the compound.
5. Experimental results:
inhibition of three CYPs by the compounds of the present invention was determined as described above and the results are shown in table 5 below.
TABLE 5 CYPs inhibition test results
The above results indicate that the compounds of the present invention have no significant inhibition of all 3 major CYP subtypes, indicating a relatively low potential for drug interactions.
Test example six: CYP induction test.
1. Test system:
the assay uses HepG2 cells transiently transfected with the CYP3A4 luciferase reporter gene to assess the induction of CYP3A4 by the compounds.
2. The test method comprises the following steps:
1) HepG2 cells were cultured in a T25 cell culture flask until the degree of fusion reached about 80%, and plasmid-entrapped liposomes were prepared according to Lipofectamine 3000. Lipofectamine 3000 in the test System: 7.5 μl, P3000: 20. Mu.L of plasmid pGL4.16-CYP3A4-Promoter and pcDNA3.1 (+) -hPSR 5. Mu.g each. The prepared liposome was added dropwise into a T25 flask to transfect HepG2 cells.
2) 24h after transfection, cells were digested and counted. Cells were diluted to 5.56X10 with MEM medium (10% FBS) 5 cells/mL were spread uniformly at 90. Mu.L/well to Assay plate (96 well plate, clear) and each well contained about 5.0X10 cells 4 And each. Positive rifampicin and test compounds were diluted to 10. Mu.M and 1. Mu.M in MEM medium (10% FBS). The negative control was 0.1% dmso.
3) Adding diluted compound into corresponding well at a ratio of 10 μl/well, tapping Assay plate edge for about 2min, mixing thoroughly with culture medium, standing at 37deg.C, 5% CO 2 Incubation in incubator for 24h.
4) Cell plates and Bright-Glo TM The luciferase reagent was removed and equilibrated to room temperature.
5) At 50. Mu.L/well, bright-Glo was added TM Shake at 650rpm for 5min.
6) The mixture in the well plate was transferred rapidly 100. Mu.l to Detection plate (96 well plate, white opaque), and luminescence values were measured on a PHERAstar FS microplate reader.
3. And (3) data processing:
the activation of CYP3A4 by the test compound was calculated according to the following formula:
percent (%) activation of CYP3A4 = (test compound well luminescence value-negative control well mean luminescence value)/(10 μm positive control rifampicin well mean luminescence value-negative control well mean luminescence value) ×100%;
when the activation percentage is more than or equal to 20%, judging that the compound to be tested has CYP3A4 induction potential; when the percent activation is < 20%, it is judged that the compound CYP3A4 has weak or no induction activity.
4. Experimental results:
the results are shown in table 6 below.
TABLE 6 CYP3A4 Induction test results
Examples numbering Percent activation of CYP3A4 CYP3A4 Induction
Example five 11.33±2.26 Negative of
Example six 3.46±1.47 Negative of
Example seven 2.23±2.43 Negative of
Example eight 19.64±5.56 Negative of
The test result shows that the compound has no obvious induction effect on CYP3A4, and the risk of drug interaction generated by the compound is lower.
Test example seven: SD rat Pharmacokinetic (PK) studies.
The pharmacokinetic profile was examined by administering the compound of example seven-a to male SD rats by Intravenous (IV) and intragastric (PO) administration, respectively. The administration doses of IV and PO were 1mg/kg and 5mg/kg, respectively, the solvent of IV was 5% DMSO+5% solutol (15-hydroxystearate polyethylene glycol ester) +90% physiological saline, and the solvent of PO was 0.5% MC (sodium methylcellulose). Blood was collected at different time points after IV and PO dosing. Blood is anticoagulated by EDTA-K2, and is centrifuged to obtain a plasma sample, which is stored at-80 ℃.
Plasma samples were subjected to LC-MS/MS analysis after treatment with precipitated proteins. Pharmacokinetic parameters were calculated using the non-compartmental model using WinNonlin 6.3 software, the results are given in table 7 below.
TABLE 7 pharmacokinetic parameters of Compounds in rat blood
As shown in Table 7, the exposure (AUC) of the compound of example seven-A to rats by IV administration at a dose of 1mg/kg last ) At 328h ng/mL, the corresponding maximum plasma concentration (C max ) At 684ng/mL, clearance (Cl) was 51.1mL/min/kg, indicating that the compounds of the present invention had excellent drug exposure in rats by IV administration.
AUC in rats of the compound of example seven-A administered at a dose of PO of 5mg/kg last 1949h ng/mL, corresponding C max At 1062ng/mL, the compounds of the present invention were shown to have excellent drug exposure in the rat blood system by PO administration.
The half-life of the seven-a compound of the oral example was calculated to be 0.56h and the bioavailability was calculated to be 119% compared to intravenous administration.
Taken together, the compounds of example seven-a demonstrate excellent pharmacokinetic properties in rats.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in this application (including all patents, patent applications, journal articles, books, and any other publications) is incorporated herein by reference in its entirety.

Claims (21)

1. A compound having the structure of formula I-1 or a pharmaceutically acceptable form thereof,
wherein,
R 1 selected from 5-6 membered heteroaryl, said 5-6 membered heteroaryl optionally substituted with one or more R 4 Substitution;
R 4 each at each occurrence is independently selected from hydrogen, deuterium, and C 1-6 An alkyl group;
R 2 selected from 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 5 Substitution;
R 5 each at each occurrence is independently selected from hydrogen, deuterium, halogen and C 1-6 Alkyl, said C 1-6 Alkyl is optionally substituted with one or more R 7 Substitution;
R 3 selected from C 6-10 Aryl and 5-10 membered heteroaryl, said C 6-10 Aryl or 5-10 membered heteroaryl optionally substituted with one or more R 6 Substitution;
R 6 each at each occurrence is independently selected from hydrogen, deuterium, halogen, C 1-6 Alkyl, -OR a and-S (=o) q R a Q is 1 or 2, said C 1-6 Alkyl is optionally substituted with one or more R 7 Substitution;
R 7 at each timeEach occurrence is independently selected from hydrogen, deuterium, and halogen;
R a each at each occurrence is independently selected from hydrogen and C 1-6 An alkyl group;
the pharmaceutically acceptable form is selected from pharmaceutically acceptable salts.
2. The compound or pharmaceutically acceptable form thereof according to claim 1, wherein,
R 1 is a pyridinyl group, optionally substituted with one or more C 1-6 Alkyl substitution.
3. The compound or pharmaceutically acceptable form thereof according to claim 1, wherein,
R 1 selected from 6-methylpyridin-2-yl.
4. A compound or pharmaceutically acceptable form thereof according to any one of claims 1 to 3, wherein,
R 2 selected from benzothiazolyl and triazolopyridinyl, optionally substituted with one or more R 5 And (3) substitution.
5. The compound or pharmaceutically acceptable form thereof according to claim 4, wherein,
R 2 Selected from benzo [ d ]]Thiazolyl and [1,2,4 ]]Triazolo [1,5-a ]]A pyridyl group.
6. The compound or pharmaceutically acceptable form thereof according to claim 5, wherein,
R 2 selected from benzo [ d ]]Thiazol-6-yl and [1,2,4 ]]Triazolo [1,5-a ]]Pyridin-6-yl.
7. The compound or pharmaceutically acceptable form thereof according to any one of claims 1 to 3 or 5 to 6, wherein,
R 3 selected from phenyl and 5-6 membered heteroaryl groups, said phenyl and 5-6 membered heteroaryl groupsThe radicals optionally being substituted by one or more R 6 And (3) substitution.
8. The compound or pharmaceutically acceptable form thereof according to claim 7, wherein,
R 3 selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, halogen, C 1-6 Alkyl, -OR a and-S (=o) q R a Q is 1 or 2.
9. The compound or pharmaceutically acceptable form thereof according to claim 8, wherein,
R 3 selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each at each occurrence is independently selected from hydrogen, fluorine, bromine, hydroxyl, C 1-6 Alkyl and-S (=o) 2 -C 1-6 An alkyl group.
10. The compound or pharmaceutically acceptable form thereof according to claim 9, wherein,
R 3 Selected from phenyl, pyrazolyl and pyrazinyl, optionally substituted with one or more R 6 Substituted, R 6 Each independently at each occurrence is selected from hydrogen, bromo, hydroxy, methyl, and methanesulfonyl.
11. The compound or pharmaceutically acceptable form thereof according to claim 10, wherein,
R 3 selected from the group consisting of 4-hydroxyphenyl, 1- (methylsulfonyl) -1H-pyrazol-4-yl, 3-hydroxyphenyl, 3-bromophenyl, pyrazin-2-yl and 1- (methyl) -1H-pyrazol-4-yl.
12. The compound or pharmaceutically acceptable form thereof according to claim 1, wherein,
R 4 selected from methyl groups.
13. The compound or pharmaceutically acceptable form thereof according to claim 1, wherein,
R 5 selected from hydrogen and C 1-6 An alkyl group.
14. The compound or pharmaceutically acceptable form thereof according to claim 13, wherein,
R 5 is hydrogen.
15. The compound or pharmaceutically acceptable form thereof according to claim 1, wherein,
R 6 selected from hydrogen, hydroxy, halogen, C 1-6 Alkyl and methanesulfonyl.
16. The compound or pharmaceutically acceptable form thereof according to claim 15, wherein,
R 6 selected from the group consisting of hydrogen, hydroxy, methyl, fluoro, bromo and methanesulfonyl.
17. The compound or pharmaceutically acceptable form thereof according to claim 1, wherein,
R 1 is a 5-6 membered heteroaryl, said 5-6 membered heteroaryl optionally being substituted with one or more R 4 Substitution, said R 4 Selected from hydrogen, deuterium and C 1-6 An alkyl group;
R 2 is a 5-10 membered heteroaryl, said 5-10 membered heteroaryl optionally substituted with one or more R 5 Substitution, said R 5 Selected from hydrogen and C 1-6 An alkyl group;
R 3 selected from phenyl and 5-6 membered heteroaryl, optionally substituted with one or more R 6 Substitution, said R 6 Selected from hydrogen, hydroxy, halogen, C 1-6 Alkyl and methanesulfonyl.
18. The following compounds or pharmaceutically acceptable salts thereof:
19. a pharmaceutical composition comprising at least one compound according to any one of claims 1 to 18, or a pharmaceutically acceptable form thereof, and one or more pharmaceutically acceptable carriers.
20. A kit, comprising:
a) At least one compound according to any one of claims 1 to 18, or a pharmaceutically acceptable form thereof, as a first therapeutic agent, or a pharmaceutical composition according to claim 19 as a first pharmaceutical composition;
b) Optionally at least one other therapeutic agent as a second therapeutic agent, or a pharmaceutical composition comprising the other therapeutic agent as a second pharmaceutical composition; and
c) Optionally package and/or instructions.
21. Use of a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition according to claim 19, in the manufacture of a medicament for the prevention and/or treatment of a disease or condition mediated at least in part by tgfβr1, which disease or condition mediated at least in part by tgfβr1 is cancer.
CN202011084004.4A 2019-10-22 2020-10-12 Pyrazolo ring compound, pharmaceutical composition containing pyrazolo ring compound, preparation method and application of pyrazolo ring compound Active CN112694477B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019110081239 2019-10-22
CN201911008123 2019-10-22

Publications (2)

Publication Number Publication Date
CN112694477A CN112694477A (en) 2021-04-23
CN112694477B true CN112694477B (en) 2024-02-06

Family

ID=75505789

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011084004.4A Active CN112694477B (en) 2019-10-22 2020-10-12 Pyrazolo ring compound, pharmaceutical composition containing pyrazolo ring compound, preparation method and application of pyrazolo ring compound

Country Status (1)

Country Link
CN (1) CN112694477B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004026307A1 (en) * 2002-09-18 2004-04-01 Pfizer Products Inc. Triazole derivatives as transforming growth factor (tgf) inhibitors
CN106795139A (en) * 2014-10-07 2017-05-31 伊莱利利公司 Aminopyridine base epoxide pyrazole compound

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004026863A1 (en) * 2002-09-18 2004-04-01 Pfizer Products Inc. Novel oxazole and thiazole compounds as transforming growth factor (tgf) inhibitors
AR103232A1 (en) * 2014-12-22 2017-04-26 Bristol Myers Squibb Co TGFbR ANTAGONISTS
WO2017015425A1 (en) * 2015-07-23 2017-01-26 Bristol-Myers Squibb Company Tgf beta receptor antagonists
ES2787449T3 (en) * 2016-06-13 2020-10-16 Genfleet Therapeutics Shanghai Inc Benzotriazole-derived unsaturated alpha and beta-amide compound used as TGF-betaRI inhibitor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004026307A1 (en) * 2002-09-18 2004-04-01 Pfizer Products Inc. Triazole derivatives as transforming growth factor (tgf) inhibitors
CN106795139A (en) * 2014-10-07 2017-05-31 伊莱利利公司 Aminopyridine base epoxide pyrazole compound

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
转化生长因子-β 抑制剂研究进展;盖小雄 等;《药学学报》;第50卷(第4期);第413-418页 *

Also Published As

Publication number Publication date
CN112694477A (en) 2021-04-23

Similar Documents

Publication Publication Date Title
JP6523356B2 (en) Macrocyclic compounds as TRK kinase inhibitors
KR20220027879A (en) Regulators of nitrogen-containing heterocyclic derivatives, preparation methods and uses thereof
BR112020011746A2 (en) substituted bicyclic heterocyclic compounds acting as prmt5 inhibitors
WO2021143701A1 (en) Pyrimidine-4(3h)-ketone heterocyclic compound, preparation method therefor and use thereof in medicine and pharmacology
JP6888006B2 (en) Pyrazolo, pyrazolo, imidazole-pyrimidine and pyridine compounds that inhibit MNK1 and MNK2
CN109721600B (en) Nitrogen-containing fused ring compounds and preparation method and application thereof
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
JP2023518609A (en) Novel aminopyrimidine-based EGFR inhibitor
CN113968856B (en) Compounds with kinase inhibitory activity
CN112341477B (en) Imidazo pyrimidine compound containing fused ring group, preparation method and application thereof
CN112694477B (en) Pyrazolo ring compound, pharmaceutical composition containing pyrazolo ring compound, preparation method and application of pyrazolo ring compound
WO2023036252A1 (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical use thereof
CN114380818A (en) Pyrazolo pyridine compound, and pharmaceutical composition, preparation method and application thereof
CN113227100B (en) Thienopyridone compounds
JP2023530768A (en) FGFR and its mutation inhibitor, its production method and application
TW202024070A (en) FGFR inhibitor, preparation method therefor and application thereof
CN116249692A (en) Pyrazole compound as well as preparation method and application thereof
CN108727368A (en) Nitrogenous fused heterocyclic compound, preparation method, intermediate, composition and application
WO2024083111A1 (en) Novel heterocyclic compounds
RU2796605C2 (en) 7h-pyrrolo[2,3-d]pyrimidine-4-amine derivative
CN114380845A (en) Heterocyclic-fused compound, pharmaceutical composition containing same, preparation method and application thereof
CN114149410A (en) Pyridine-fused ring compound and preparation method and application thereof
CN116801880A (en) Nitrogen-containing heterocyclic compounds
CN114369083A (en) Heterocyclic compound, pharmaceutical composition containing same, preparation method and application thereof
CN117247387A (en) Aromatic heterocyclic compound and preparation method 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