CN113801135A - Thienopyrazole compounds, pharmaceutical composition containing same and application thereof - Google Patents

Thienopyrazole compounds, pharmaceutical composition containing same and application thereof Download PDF

Info

Publication number
CN113801135A
CN113801135A CN202010542108.9A CN202010542108A CN113801135A CN 113801135 A CN113801135 A CN 113801135A CN 202010542108 A CN202010542108 A CN 202010542108A CN 113801135 A CN113801135 A CN 113801135A
Authority
CN
China
Prior art keywords
group
hydrogen
alkyl
cycloalkyl
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010542108.9A
Other languages
Chinese (zh)
Inventor
陈寿军
宋帅
蒋小玲
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
Priority to CN202010542108.9A priority Critical patent/CN113801135A/en
Publication of CN113801135A publication Critical patent/CN113801135A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-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
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the field of medicinal chemistry, and relates to a thienopyrazole compound, a medicinal composition containing the thienopyrazole compound and application of the thienopyrazole compound. Specifically, the invention provides a compound with a structure shown in a formula I, which shows better antitumor activity.

Description

Thienopyrazole compounds, pharmaceutical composition containing same and application thereof
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and relates to a thienopyrazole compound with UPS7 inhibitory activity, a pharmaceutical composition containing the thienopyrazole compound, and medical application of the thienopyrazole compound.
Background
Ubiquitin-proteasome system (UPS) is a basic physiological regulatory process in cells, and proteins are ubiquitinated and modified by proteases and degraded by proteases through a series of cascade reactions. The abnormality of UPS is closely related to tumor, neurodegenerative disease, virus infection and other diseases. At present, medicines are mainly developed aiming at five types of targets of protease, E1 activating enzyme, E2 binding enzyme, E3 ligase and Deubiquitinases (DUBs) in a UPS system.
The deubiquitinase can specifically cut off isopeptide bonds formed between glycine residues at the carbon terminal of ubiquitin and target proteins, so that ubiquitin is separated from the target proteins, and the target proteins are prevented from being degraded, relocated or activated.
There are currently approximately 100 DUBs in humans, of which ubiquitin-specific proteases (USPs) are the largest family members of DUBs, including about 85 members, an isopeptidase belonging to the cysteine protease family (Wu and Kumar, Journal of Medicinal Chemistry,2018,61: 422-. More than 40 family members of USPs have been found to be associated with tumor development and progression.
USP7 is located in the nucleus, a key deubiquitinase in UPS, and can specifically cut off the isopeptide bond formed between the carbon terminal of ubiquitin and the target protein, so that ubiquitin is separated from the target protein, and the target protein is protected from degradation, relocation or activation (Turnbull and Ioannidis, Nature,2017,550: 481-486).
USP7 has a wide distribution of human tissues and plays a major role in neural development, cell cycle regulation, epigenetic regulation, DNA damage repair, and immune response. Studies have shown that USP7 is overexpressed in cancer cells such as hepatocellular carcinoma, multiple myeloma, colon cancer, lung cancer, prostate cancer, and bladder cancer, and that this overexpression is directly related to tumor invasion and poor prognosis (Pozhidaeva and Bezsonova, DNA Repair,2019,76: 30-39).
USP7 has a wide variety of substrates, and most of them are proteins involved in Cell cycle regulation, immune response, apoptosis, DNA damage repair, etc., such as MDM2, p53, ERCC6, Foxp3, PTEN, FOXO4, etc. (Chauhan and Tian, Cancer Cell,2012,22:345 and 358). In some tumor cells MDM2 is overexpressed, USP7 protects MDM2 from ubiquitination, and MDM2, after binding to p53 protein, promotes ubiquitination and degradation of p53 protein, promoting tumor growth.
USP7 can also exert tumor promotion effects by directly modulating the expression of tumor suppressor proteins (e.g., p53, PTEN, FOXO4, p114ARF, p16INK4) and tumor promoting proteins (e.g., N-MYC, REST), up-regulating the expression of tumor associated factors (e.g., HIF-1), and modulating tumor associated signaling pathways (e.g., SHH signaling pathway, Wnt/β -catenin signaling pathway, androgen receptor signaling pathway, DNA damage repair signaling pathway) (Zhou and Wang, Medicinal Chemistry,2018,14: 3-18).
In addition, USP7 also plays a role in tumor immune surveillance escape by regulating Treg cell upstream signal molecules (such as transcription factor FOXP3 and epigenetic regulatory factor Tip60), up-regulating Treg cell activity (Wang and Wu, PLoS One,2017,12:1-23) and inhibiting Teff cell (CD8+ T cell) activity.
The development of inhibitors of USP7 is one of the hotspots in the field of tumor research. Currently, no drugs are on the market worldwide for the USP7 target, and the compounds under investigation are all in the preclinical stage of study.
Although companies such as hybrids SA, Forma Therapeutics, inc, Les laboratories server, Almac Discovery Limited have corresponding research on USP7 inhibitors and related patent publications, there is still a great need in the art for new USP7 inhibitors, particularly USP7 inhibitors with high activity and other excellent properties.
Disclosure of Invention
Problems to be solved by the invention
Through a large number of researches, the invention develops a thienopyrazole compound and a corresponding preparation method thereof, the thienopyrazole compound can obviously inhibit the activity of UPS7, and the thienopyrazole compound can be used as an UPS7 inhibitor for preventing and/or treating at least partial diseases mediated by UPS7, especially tumor diseases (cancer).
Means for solving the problems
In a first aspect, the present invention provides a compound having the structure of formula I or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof,
Figure BDA0002539269000000021
wherein the content of the first and second substances,
R0selected from hydrogen, halogen, cyano, C1-6Alkyl and C1-6A haloalkyl group;
L1is absent or selected from C1-6Alkylene radical, C1-6Haloalkylene, C3-8Cycloalkylene and 3-8 membered heterocycloalkylene;
R1selected from hydrogen, hydroxy, halogen, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C6-10Aryl, 5-10 membered heteroaryl, -NRaRb
Figure BDA0002539269000000022
Wherein the content of the first and second substances,
Raand RbEach independently selected from hydrogen and C1-6Alkyl radical, C3-8Cycloalkyl, -C (O) -C1-6Alkyl, -C (O) -C3-8Cycloalkyl, -C (O) -C2-6Alkenyl, -S (O)q-C1-6Alkyl, -S (O)q-C3-8Cycloalkyl and-S (O)q-C2-6An alkenyl group; wherein, the C1-6Alkyl, aryl, heteroaryl, and heteroaryl,C3-8Cycloalkyl and C2-6Each alkenyl group is independently optionally substituted with one or more of the following: hydrogen, -C1-6alkylene-C (O) OH and-C (O) O-C1-6An alkyl group; q is selected from 1 and 2;
b is selected from 3-8 membered heterocycloalkyl, 3-8 membered heterocycloalkenyl and 5-10 membered heteroaryl;
c is selected from C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C4-6Cycloalkenyl, 3-8 membered heterocycloalkenyl, C6-10Aryl and 5-10 membered heteroaryl;
d is selected from C3-8Cycloalkyl and 3-8 membered heterocycloalkyl;
if present, each R9And R10Each independently selected from hydrogen, halogen, cyano, oxo, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, carboxy, -C (O) O-C1-6Alkyl and-C1-6alkylene-C (O) OH; wherein, the C1-6Alkyl radical, C3-8Cycloalkyl and 3-8 membered heterocycloalkyl are each independently optionally substituted with one or more of the following: hydrogen, hydroxy and amino;
z3、z4and z5Each independently selected from 0, 1,2, 3, 4, 5 and 6;
R2、R3、R4、R5and R6Each independently selected from hydrogen, hydroxy, halogen, cyano, carboxy, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl radical, C1-6Alkoxy and C1-6A haloalkoxy group;
L2selected from oxygen, sulfur, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-, -S-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl and-C (O) -C1-6An alkyl group;
a is selected from C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl;
if present, each R7And R8Each independently selected from hydrogen, hydroxy, halogen, cyano, -NRcRd、C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, -C1-6alkylene-OH, -C1-6alkylene-O-C1-6Alkyl, -C (O) -N (C)1-6Alkyl radical)2、-C(O)-C1-6Alkyl and-C (O) -C1-6Haloalkyl, or R7And R8And the atoms to which they are attached together form C3-8Cycloalkyl or 3-8 membered heterocycloalkyl; wherein the content of the first and second substances,
Rcand RdEach independently selected from hydrogen and C1-6Alkyl and-C (O) -C1-6An alkyl group;
z1and z2Each independently selected from 0, 1,2, 3, 4, 5, 6, 7, 8, 9 and 10.
In a second aspect, the present invention provides specific compounds having the structure of formula I, comprising:
(1)3- ((3- (5-chloro-3-methyl-2- (piperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(2)3- ((3- (5-chloro-3-methyl-2- (2-methylpiperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(3)3- ((3- (5-chloro-3-methyl-2- (3-methylpiperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(4)3- ((3- (5-chloro-3-methyl-2- (piperidin-4-ylamino) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(5)3- ((3- (5-chloro-3-methyl-2- (piperidine-4-carbonyl) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(6)3- ((3- (5-chloro-3-methyl-2- (piperidin-4-ylmethyl) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(7)3- ((3- (5-chloro-2- (2, 6-dimethylpiperidin-4-yloxy) -3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(8)3- ((3- (5-chloro-3-methyl-2- (1-methylpiperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(9)3- ((3- (5-chloro-3-methyl-2- (6-methylpiperidin-3-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(10)3- ((3- (5-chloro-3-methyl-2- (quinin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(11)3- ((3- (2- (1-azabicyclo [3.2.1] octan-5-yloxy) -5-chloro-3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(12)3- ((3- (2- (3-aminocyclopentyloxy) -5-chloro-3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(13)3- ((3- (5-chloro-2- (2, 2-dimethylpyrrolidin-3-yloxy) -3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(14)3- ((3- (5-chloro-2- (4-methoxypyrrolidin-3-yloxy) -3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(15)3- ((3- (5-chloro-2- (1, 4-dimethylpyrrolidin-3-yloxy) -3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione;
(16)3- ((3- (2- ((3-aminocyclobutyl) methoxy) -5-chloro-3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione; and
(17)3- ((3- (5-chloro-3-methyl-2- ((3-methylazetidin-3-yl) methoxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione.
In a third aspect, the present invention provides a pharmaceutical composition comprising a compound having the structure of formula I, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, as described above, and one or more pharmaceutically acceptable carriers.
In a fourth aspect, the present invention provides a pharmaceutical product comprising:
a) a compound having the structure of formula I or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, or a pharmaceutical composition thereof, as described above;
b) optionally packaging and/or instructions.
In a fifth aspect, the present invention provides a compound having the structure of formula I, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixtures thereof, as described above, or a pharmaceutical composition as described above, or a pharmaceutical product as described above, for use as an inhibitor of UPS7, for the prevention and/or treatment of a disease or condition mediated at least in part by UPS7 (in particular cancer), or for use in inhibiting UPS7 activity in vitro or in inhibiting tumor cell proliferation in vitro.
In a sixth aspect, the present invention provides a compound having the structure of formula I, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixture thereof, or a pharmaceutical composition thereof, or a medicament thereof, for use as an inhibitor of UPS 7.
In a seventh aspect, the present invention provides a use of a compound having the structure of formula I, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixture thereof, or a pharmaceutical composition thereof, or a pharmaceutical product thereof, for the manufacture of a medicament for the prevention and/or treatment of a disease or condition mediated at least in part by UPS7, particularly cancer.
In an eighth aspect, the present invention provides a method for preventing and/or treating a disease or condition mediated at least in part by UPS7 (particularly cancer), comprising the steps of: administering to a subject in need thereof a prophylactically and/or therapeutically effective amount of a compound having the structure of formula I, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, or a pharmaceutical composition thereof, or a pharmaceutical product thereof.
ADVANTAGEOUS EFFECTS OF INVENTION
The invention provides a thienopyrazole compound with a novel structure, which can be used as a high-efficiency UPS7 inhibitor and has antitumor activity. In addition, the synthesis method of the compound is mild, and the operation is simple and easy.
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, unless otherwise defined, all terms used herein have the same meaning or meaning as commonly understood by one of ordinary skill in the art. Although the meaning or intention of the terms used herein will be easily understood by those skilled in the art, the following description is made in order to better explain the technical aspects of the present invention.
[ definition of terms ]
The terms "comprising", "including", "having", "involving" 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 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 present invention that are substantially non-toxic to organisms. Pharmaceutically acceptable salts generally include, but are not limited to, salts formed by reacting a compound of the invention with a pharmaceutically acceptable inorganic/organic acid or inorganic/organic base, 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 and that hydrolyzes in vivo to 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 esters.
The term "isomers" refers to compounds having the same molecular weight, but differing in the spatial arrangement or configuration of the atoms, due to the same number and type of atoms.
The term "stereoisomer" (or "optical isomer") refers to a stable isomer having a perpendicular plane of asymmetry due to having at least one chiral factor (including chiral center, chiral axis, chiral plane, etc.) that enables rotation of plane polarized light. Since the compounds of the present invention have asymmetric centers as well as other chemical structures that may lead to stereoisomers, the present invention also includes such stereoisomers and mixtures thereof. Since the compounds of the present invention (or pharmaceutically acceptable salts thereof) include asymmetric carbon atoms, they can exist in the form of single stereoisomers, racemates, mixtures of enantiomers and diastereomers. Generally, these compounds can be prepared in the form of racemates. However, if desired, such compounds may 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 hereinafter, individual stereoisomers of compounds are prepared synthetically from optically active starting materials containing the desired chiral center, 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 the enantiomers on chiral chromatographic columns. The starting compounds of a particular stereochemistry are either commercially available or may be prepared according to the methods described hereinafter and resolved by methods well known in the art. The term "enantiomer" refers to a pair of stereoisomers that have non-superimposable mirror images of each other. The term "diastereomer" 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 parts of a single enantiomer. Unless otherwise indicated, all stereoisomeric forms of the compounds of the present invention are within the scope of the present invention.
The term "tautomer" (or "tautomeric form") refers to structural isomers having different energies that can interconvert through a low energy barrier. If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (or proton transfer tautomers) include, but are not limited to, interconversions by proton transfer, such as keto-enol isomerization, imine-enamine isomerization, amide-iminoalcohol isomerization, 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. Polymorphs of a molecule can be obtained by a number of known methods by those skilled in the art. These 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, and Scanning Electron Microscopy (SEM), among others.
The term "solvate" refers to a substance formed by the binding of a compound of the present invention (or a pharmaceutically acceptable salt thereof) to at least one solvent molecule by non-covalent intermolecular forces. Common solvates include, but are not limited to, hydrates (including hemihydrate, monohydrate, dihydrate, trihydrate, etc.), ethanolates, acetonates, and the like.
The term "isotopic label" refers to a derivatized compound formed by replacing a particular atom in a compound of the invention with its isotopic atom. Unless otherwise indicated, the compounds of the present invention include various isotopes of H, C, N, O, F, P, S, Cl, such as2H(D)、3H(T)、13C、14C、15N、17O、18O、18F、31P、32P、35S、36S and37and (4) Cl. For example,12c can be covered12C、13C or14C, replacing;1h can be covered2H (D, deuterium) or3H (T, tritium) substitution;16o can be18O substitution, and the like. The invention includes isotopically-labeled compounds obtained by substituting any atom in the structure with its isotope.
The term "metabolite" refers to a derivative compound formed after the compounds of the present invention are metabolized. Further information on metabolism can be found in Goodman and Gilman's: The pharmaceutical Basis of Therapeutics (9)thed.)[M],McGraw-Hill International Editions,1996。
The term "prodrug" refers to a derivative compound that is capable of providing, directly or indirectly, a compound of the invention upon administration to a subject. Particularly preferred derivative compounds or prodrugs are those that increase the bioavailability of the compounds of the invention when administered to a subject (e.g., more readily absorbed into the blood), or facilitate delivery of the parent compound to the site of action (e.g., the lymphatic system). Unless otherwise indicated, all prodrug forms of the compounds of the present invention are within the scope of the present invention, and various prodrug forms are well known in the art.
The term "independently of each other" means that at least two groups (or ring systems) present in the structure in the same or similar range of values may have the same or different meaning in a particular case. For example, substituent X and substituent Y are each independently hydrogen, halogen, hydroxy, cyano, alkyl or aryl, and when substituent X is hydrogen, substituent Y may be either hydrogen, halogen, hydroxy, cyano, alkyl or aryl; similarly, when the substituent Y is hydrogen, the substituent X may be hydrogen, or may be halogen, hydroxy, cyano, alkyl or aryl.
The term "alkyl" refers to a straight or branched chain saturated aliphatic hydrocarbon group. For example, the term "C" as used in the present invention1-6Alkyl "refers to an alkyl group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl, and the like) optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., when substituted with halogen, the group is" C1-6Haloalkyl radicals ", e.g. -CF3、-C2F5、-CHF2、-CH2F、-CH2CF3、-CH2Cl、-CH2CH2CF3Etc.).
The term "alkylene" refers to a divalent straight or branched chain saturated aliphatic hydrocarbon radical, the two radicals or segments to which it is attached being capable of connecting either the same carbon atom or different carbon atoms. For example, the term "C" as used in the present invention1-6Alkylene "refers to an alkylene group having 1 to 6 carbon atoms (e.g., methylene, 1-ethylene, 1, 2-ethylene, and the like) optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., when substituted with a halogen, the group is" C1-6Haloalkylene ", for example-CF2-、-C2F4-, -CHF-, etc.).
The term "cycloalkyl" refers to a monocyclic or polycyclic (such as bicyclic) aliphatic hydrocarbon group (e.g., monocyclic cycloalkyl 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]Pentyl, bicyclo [2.2.1]Heptyl, etc.), optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., oxo). For example, the term "C" as used in the present invention3-8Cycloalkyl "refers to a cycloalkyl group having 3 to 8 ring-forming carbon atoms (e.g., 2-methylcyclopropyl).
The term "cycloalkylene" refers to a divalent monocyclic or polycyclic (such as bicyclic) aliphatic hydrocarbon group, to which two groups or moieties are attached that can join either the same or different ring-forming carbon atoms, optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., oxo). For example, the term "C" as used in the present invention3-8Cycloalkylene "refers to cycloalkylene groups having 3 to 8 ring-forming carbon atoms (e.g.
Figure BDA0002539269000000071
)。
The term "heterocycloalkyl" refers to a monocyclic or polycyclic (such as bicyclic) aliphatic radical having one or more carbon atoms (e.g., 2, 3, 4, 5, 6, 7, 8, or 9) in the ring and one or more (e.g., 2, 3, or 4) each independently selected from O, S, N, NH, S (O), and S (O)2Optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., oxo). The heterocycloalkyl group, if required for a valence, may be attached to the parent molecular moiety through any one of the carbon atoms or heteroatoms in the ring. For example, the term "3-8 membered heterocycloalkyl" as used herein refers to heterocycloalkyl (e.g., oxiranyl, aziridinyl, aziridyl), azetidine) having 3 to 8 ring atoms (containing one or more heteroatoms or groups of heteroatoms) thereinAn azetidinyl group (azetidinyl), an oxetanyl group (oxolanyl), a tetrahydrofuranyl group (tetrahydrofuryl), a pyrrolidinyl group (pyrrolidinyl), a pyrrolidinonyl group (pyrrolidinyl), an imidazolidinyl group (imidazolidinylidinyl), a pyrazolidinyl group (pyrrolidinyl), a pyrrolidinyl group (pyrrolidinyl), a tetrahydropyranyl group (tetrahydropyranyl) and the like.
The term "heterocycloalkylene" refers to a divalent monocyclic or polycyclic (such as bicyclic) aliphatic hydrocarbon group having one or more carbon atoms (e.g., 2, 3, 4, 5, 6, 7, 8, or 9) in the ring and one or more (e.g., 2, 3, or 4) each independently selected from O, S, N, NH, S (O), and S (O)2The two groups or fragments to which they are attached can be either attached to the same ring-forming carbon atom or to a ring-forming carbon atom and a ring-forming heteroatom (e.g., nitrogen atom), respectively, which is optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., oxo). For example, the term "3-8 membered heterocycloalkylene" as used herein refers to a heterocycloalkylene group (e.g., containing one or more heteroatoms or groups of heteroatoms) having 3 to 8 ring atoms
Figure BDA0002539269000000081
)。
The term "alkoxy" means an "alkyl" or "cycloalkyl" (e.g., C) as defined above attached to the parent molecular moiety through an oxygen atom1-6Alkoxy radical, C3-8Cycloalkoxy and the like, including methoxy, ethoxy, n-propoxy, isopropoxy, cyclopropoxy, n-butoxy, isobutoxy, t-butoxy, sec-butoxy, cyclobutoxy, n-pentoxy, isopentoxy, n-hexoxy, or isomers thereof). For example, the term "C" as used in the present invention1-6Alkoxy "refers to an alkoxy group having 1 to 6 carbon atoms (e.g., methoxy, ethoxy, t-butoxy, etc.) optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., when substituted with a halogen, the group is" C1-6Haloalkoxy ", e.g. -OCF3、-OC2F5Etc.).
The term "aryl" refers toMonocyclic or fused polycyclic aromatic hydrocarbon radicals having a conjugated pi-electron system, optionally substituted by one or more substituents described herein (e.g. by oxo, C)1-6Alkyl (methyl, ethyl) or halo (fluoro, chloro, bromo). For example, the term "C" as used in the present invention6-10Aryl "means an aryl group having 6 to 10 carbon atoms (e.g., phenyl, naphthyl, etc.).
The term "heteroaryl" refers to a monocyclic or fused polycyclic aromatic group having a conjugated pi-electron system, having one or more carbon atoms (e.g., 1,2, 3, 4, 5, 6, 9, or 10 carbon atoms) and one or more (e.g., 2, 3, or 4) heteroatoms or groups of heteroatoms, each independently selected from O, S, N and NH, in the ring, optionally substituted with one or more substituents described herein (e.g., by oxo, C1-6Alkyl (methyl, ethyl) or halo (fluoro, chloro, bromo). A heteroaryl group may be attached to the parent molecular moiety through any one of the ring atoms if the valency requirements are met. For example, the term "5-10 membered heteroaryl" used in the present invention means a heteroaryl group having 5 to 10 ring atoms (e.g., thienyl (thienyl), furyl (furyl), pyrrolyl (pyrrolyl), oxazolyl (oxazoyl), thiazolyl (thiazoyl), imidazolyl (imidazoyl), pyrazolyl (pyrazoyl), isoxazolyl (isoxazoyl), isothiazolyl (isothiazolyl), oxadiazolyl (oxazoyl), triazolyl (triazolyl), thiadiazolyl (thiadiazolyl), pyridyl (pyridyl), pyridazinyl (pyridazinyl), pyrimidinyl (pyrimidiniyl), pyrazinyl (pyrazinyl), triazinyl (triazizyl) or a benzo derivative thereof, etc.).
The term "alkenyl" refers to a straight or branched chain aliphatic hydrocarbon group containing one or more carbon-carbon double bonds, typically containing from about 2 to 15 carbon atoms. For example, the term "C" as used in the present invention2-6Alkenyl "means an alkenyl group containing 2 to 6 carbon atoms (e.g., ethenyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, n-octenyl, n-decenyl, and the like), which is optionally substituted with one or more (e.g., 1 to 3) substituents (e.g., halogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cyano, hydroxy, alkoxy, aryloxyAlkoxyalkyl, alkylthio, amino, alkanoyloxy, arylacyloxy, cycloalkylacyloxy, carboxyl, alkoxycarbonyl, etc.).
The term "cycloalkenyl" refers to a monocyclic or polycyclic (such as bicyclic) aliphatic hydrocarbon group (e.g., monocyclic cycloalkenyl such as cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, and the like) containing one or more carbon-carbon double bonds. For example, the term "C" as used in the present invention4-6Cycloalkenyl "refers to cycloalkenyl groups having 4 to 6 ring-forming carbon atoms, optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., oxo).
The term "heterocycloalkenyl" refers to a monocyclic or polycyclic (such as bicyclic) aliphatic group having one or more carbon atoms (e.g., 2, 3, 4, 5, 6, 7, 8, or 9) in the ring and one or more (e.g., 2, 3, or 4) each independently selected from the group consisting of O, S, N, NH, S (O), and S (O)2And contain one or more carbon-carbon double bonds, optionally substituted with one or more (e.g., 1 to 3) substituents described herein (e.g., oxo). The heterocycloalkenyl group may be attached to the parent molecular moiety through any one of the carbon atoms or heteroatoms in the ring, if the valency requirement is met. For example, the term "3-8 membered heterocycloalkenyl" as used herein refers to a heterocycloalkenyl having 3 to 8 ring atoms (which contain one or more heteroatoms or groups of heteroatoms).
The term "amino" refers to "-NH2A "group, which can be unsubstituted or substituted with one or more substituents described herein (e.g., -NH)2、-NHCH3、-N(CH3)2、-NH(C1-3Alkyl), -N (C)1-3Alkyl radical)2Etc.). The term "amino" as used herein may also be protected by an amino protecting group, unless otherwise indicated. Suitable amino protecting groups include acetyl (Ac), tert-butyloxycarbonyl (Boc), benzyloxycarbonyl (Cbz), and the like.
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
The term "cyano" refers to the "-CN" group.
The term "hydroxy" refers to an "-OH" group.
The term "carboxy" refers to the "-C (O) OH" group.
The term "oxo" refers to an "═ O" group. An oxo group typically occurs at a carbon atom that, in addition to it, also connects two other atoms, radicals (groups) or moieties, which together form a carbonyl group.
The term "carbonyl" refers to a "-C (═ O) -" or "-C (O) -" group.
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) in the specified group are replaced with other atoms or groups of atoms, provided that the specified group in the present case satisfies the valence requirements 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", that substituent may be unsubstituted or substituted. If a first substituent is described as being optionally substituted with one or more of the list of second substituents, one or more hydrogen atoms in the first substituent may be replaced, either individually or each independently, with one or more of the list of second substituents, or not.
When a bond of a substituent is shown through a bond connecting two atoms in a ring, then such substituent may be attached to any ring-forming atom in the ring.
The term "one or more" means 1 or more than 1, for example 2, 3, 4, 5, 6, 7, 8, 9 or 10, under reasonable conditions.
[ Compound of the general formula ]
The present invention provides a compound of formula I or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof,
Figure BDA0002539269000000101
wherein the content of the first and second substances,
R0selected from hydrogen, halogen, cyano, C1-6Alkyl and C1-6A haloalkyl group;
L1is absent or selected from C1-6Alkylene radical, C1-6Haloalkylene, C3-8Cycloalkylene and 3-8 membered heterocycloalkylene;
R1selected from hydrogen, hydroxy, halogen, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C6-10Aryl, 5-10 membered heteroaryl, -NRaRb
Figure BDA0002539269000000102
Wherein the content of the first and second substances,
Raand RbEach independently selected from hydrogen and C1-6Alkyl radical, C3-8Cycloalkyl, -C (O) -C1-6Alkyl, -C (O) -C3-8Cycloalkyl, -C (O) -C2-6Alkenyl, -S (O)q-C1-6Alkyl, -S (O)q-C3-8Cycloalkyl and-S (O)q-C2-6An alkenyl group; wherein, the C1-6Alkyl radical, C3-8Cycloalkyl and C2-6Each alkenyl group is independently optionally substituted with one or more of the following: hydrogen, -C1-6alkylene-C (O) OH and-C (O) O-C1-6An alkyl group; q is selected from 1 and 2;
b is selected from 3-8 membered heterocycloalkyl, 3-8 membered heterocycloalkenyl and 5-10 membered heteroaryl;
c is selected from C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C4-6Cycloalkenyl, 3-8 membered heterocycloalkenyl, C6-10Aryl and 5-10 membered heteroaryl;
d is selected from C3-8Cycloalkyl and 3-8 membered heterocycloalkyl;
if present, each R9And R10Each independently selected from hydrogen, halogen, cyano, oxo, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl, 3-to 8-membered heterocycloalkyl, carboxy、-C(O)O-C1-6Alkyl and-C1-6alkylene-C (O) OH; wherein, the C1-6Alkyl radical, C3-8Cycloalkyl and 3-8 membered heterocycloalkyl are each independently optionally substituted with one or more of the following: hydrogen, hydroxy and amino;
z3、z4and z5Each independently selected from 0, 1,2, 3, 4, 5 and 6;
R2、R3、R4、R5and R6Each independently selected from hydrogen, hydroxy, halogen, cyano, carboxy, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl radical, C1-6Alkoxy and C1-6A haloalkoxy group;
L2selected from oxygen, sulfur, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-, -S-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl and-C (O) -C1-6An alkyl group;
a is selected from C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl;
if present, each R7And R8Each independently selected from hydrogen, hydroxy, halogen, cyano, -NRcRd、C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, -C1-6alkylene-OH, -C1-6alkylene-O-C1-6Alkyl, -C (O) -N (C)1-6Alkyl radical)2、-C(O)-C1-6Alkyl and-C (O) -C1-6Haloalkyl, or R7And R8And the atoms to which they are attached together form C3-8Cycloalkyl or 3-8 membered heterocycloalkyl; wherein the content of the first and second substances,
Rcand RdEach independently selected from hydrogen and C1-6Alkyl and-C (O) -C1-6An alkyl group;
z1and z2Each independently selected from0.1, 2, 3, 4, 5, 6, 7, 8, 9 and 10.
In some embodiments of the invention, in the compounds of formula I above, R0Selected from hydrogen, halogen, C1-4Alkyl and C1-4A haloalkyl group.
In some embodiments of the invention, in the above compound of formula I, L1Is absent or selected from C1-6Alkylene and C1-6A haloalkylene group; preferably, L1Is absent or selected from C1-4Alkylene and C1-4A haloalkylene group.
In some embodiments of the invention, in the compounds of formula I above, R1Is selected from
Figure BDA0002539269000000111
Figure BDA0002539269000000112
Wherein the content of the first and second substances,
b is selected from 3-8 membered heterocycloalkyl and 3-8 membered heterocycloalkenyl;
c is selected from C3-8A cycloalkyl group;
d is selected from C3-8A cycloalkyl group;
if present, each R9And R10Each independently selected from hydrogen, halogen, cyano, oxo, C1-6Alkyl radical, C1-6Haloalkyl and carboxyl; wherein, the C1-6Alkyl is optionally substituted with one or more of the following groups: hydrogen, hydroxy and amino;
z3、z4and z5Each independently selected from 0, 1 and 2.
In some embodiments of the invention, in the compounds of formula I above, R2、R3、R4、R5And R6Each independently selected from hydrogen, halogen, cyano, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy and C1-6A haloalkoxy group.
In some embodiments of the invention, in the above compound of formula I, L2Is selected fromOxygen, sulfur, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen, C1-6Alkyl and C1-6A haloalkyl group.
In some embodiments of the invention, in the compounds of formula I above, A is selected from C3-8Cycloalkyl and 3-8 membered heterocycloalkyl.
In some embodiments of the invention, in the compounds of formula I above, each R, if present7And R8Each independently selected from hydrogen, hydroxy, halogen, cyano, -NRcRd、C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C3-8Cycloalkyl and 3-8 membered heterocycloalkyl, or R7And R8And the atoms to which they are attached together form C3-8Cycloalkyl or 3-8 membered heterocycloalkyl; preferably, if present, each R is7And R8Each independently selected from hydrogen, hydroxy, halogen, cyano, -NRcRd、C1-6Alkyl radical, C1-6Alkoxy and C1-6Haloalkyl, or R7And R8And the atoms to which they are attached together form C3-8Cycloalkyl or 3-8 membered heterocycloalkyl; wherein the content of the first and second substances,
Rcand RdEach independently selected from hydrogen and C1-6Alkyl and-C (O) -C1-6An alkyl group;
z1and z2Each independently selected from 0, 1 and 2.
In some embodiments of the present invention, in the compounds of formula I above,
R0is hydrogen;
L1is C1-6An alkylene group;
R1is selected from
Figure BDA0002539269000000121
Wherein the content of the first and second substances,
b is selected from 3-8 membered heterocycloalkyl and 3-8 membered heterocycloalkenyl;
c is C3-6A cycloalkyl group;
if present, each R9Each independently selected from oxo and C1-6A haloalkyl group;
if present, each R10Each independently selected from hydrogen and C1-6An alkyl group;
R2、R3、R4、R5and R6Each independently selected from hydrogen, halogen and C1-6An alkyl group;
L2selected from-O-, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen and C1-6An alkyl group;
a is selected from C3-8Cycloalkyl and 3-8 membered heterocycloalkyl;
if present, each R7And R8Each independently selected from hydrogen, amino, C1-6Alkyl and C1-6Alkoxy, or R7And R8And the atoms to which they are attached together form C3-8A cycloalkyl group;
z1、z2、z3and z4Each independently selected from 0, 1 and 2.
In some preferred embodiments of the present invention, in the compounds of formula I above,
R0is hydrogen;
L1is methylene;
R1is composed of
Figure BDA0002539269000000122
Wherein the content of the first and second substances,
b is 5-membered heterocycloalkyl;
c is cyclopropyl;
R9is an oxo group;
R10is C1-6An alkyl group;
z3and z4Is 2;
R2、R3、R4、R5and R6Each is independentIs selected from hydrogen, halogen and C1-6An alkyl group;
L2selected from-O-, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen and C1-6An alkyl group;
a is selected from C4-5Cycloalkyl and 4-8 membered heterocycloalkyl;
if present, each R7And R8Each independently selected from hydrogen, amino, C1-6Alkyl and C1-6An alkoxy group;
z1and z2Each independently selected from 0 and 1.
In some more preferred embodiments of the present invention, in the compounds of formula I above,
R0is hydrogen;
L1is methylene;
R1is composed of
Figure BDA0002539269000000131
R2And R4Is hydrogen;
R3is chlorine;
R5and R6Is methyl;
L2selected from the group consisting of-O-, -NH-, -C (O) -, -O-CH2-and-CH2-;
A is selected from
Figure BDA0002539269000000132
Figure BDA0002539269000000133
In some preferred embodiments of the present invention, the compound of formula I above has a structure as shown in formula II, wherein
Figure BDA0002539269000000134
R2、R3、R4、R5、R7、R8、L2、A、z1And z2As defined above.
In other preferred embodiments of the present invention, the above-described compound of formula I has the structure shown in formula III, wherein
Figure BDA0002539269000000135
R0、R1、R6、R7、R8、L1、L2、A、z1And z2As defined above.
In other preferred embodiments of the present invention, the above-described compound of formula I has the structure shown in formula IV, wherein
Figure BDA0002539269000000141
R7、R8、L2、A、z1And z2As defined above.
In some more preferred embodiments of the present invention, the above-described compound of formula I has a structure as shown in any one of formulas IV-1 to IV-5, wherein
Figure BDA0002539269000000142
R7、R8、A、z1And z2As defined above.
In addition, the present invention also provides the following compounds or their pharmaceutically acceptable salts, esters, solvates, stereoisomers, tautomers, polymorphs, isotopic labels, metabolites, prodrugs or mixtures thereof, the structures and names of which are shown in the following table:
Figure BDA0002539269000000143
Figure BDA0002539269000000151
Figure BDA0002539269000000161
[ production method ]
The invention provides a preparation method of the compound shown in the formula I.
The preparation method comprises the following steps:
Figure BDA0002539269000000171
the method comprises the following steps: reacting the compound a with the compound b to obtain a compound c;
step two: carrying out substitution reaction on the compound c to obtain a compound d;
step three: reacting the compound e with the compound f to obtain a compound g;
step four: reacting the compound g with a compound d to obtain a compound h;
step five: reacting (e.g., reducing) compound h to obtain compound i;
step six: carrying out substitution reaction on the compound i to obtain a compound j;
step seven: reacting the compound j with a compound k to obtain a compound shown in the formula I;
wherein R is0、R1、R2、R3、R4、R5、R6、R7、R8、z1、z2、L1、L2And ring A is as defined in formula I; x1、X2、X3And X4Each independently selected from halogen and trifluoromethanesulfonyloxy, preferably X1、X2、X3And X4Each independently selected from halogen; m is selected from boric acid group, C1-6Alkyl-substituted borato, five-membered oxaboryl substituted by one or more methyl groups and C1-6Alkyl-substituted tin, preferably M is tri-n-butyltin.
In some embodiments of the invention, the reaction in step one is carried out in an organic solvent selected from THF, DCM and DMF, preferably THF.
In some embodiments of the invention, the reaction in step one is in PPh3In the presence of (a).
In some embodiments of the invention, the reaction in step one is carried out in the presence of an azo compound selected from the group consisting of DIAD, DEAD and ADDP, preferably DIAD.
In some embodiments of the invention, the reaction in step one is carried out at a temperature of from 0 ℃ to 50 ℃, preferably from 0 ℃ to 25 ℃.
In some embodiments of the invention, the substitution reaction in step two is carried out in an organic solvent selected from the group consisting of Tol, THF and Et2O, preferably Tol.
In some embodiments of the invention, the substitution reaction in step two is carried out in the presence of a base selected from the group consisting of n-BuLi, t-BuLi, LiHMDS and NaHMDS, preferably n-BuLi.
In some embodiments of the invention, the substitution reaction in step two is carried out in the presence of a metal reagent selected from the group consisting of tributyltin chloride, hexa-n-butylditin, and tri-n-butylmethoxy tin, preferably tributyltin chloride.
In some embodiments of the invention, the reaction in step two is carried out at a temperature of-78 ℃ to 50 ℃, preferably-40 ℃ to 25 ℃.
In some embodiments of the invention, the reaction described in step three is carried out in an organic solvent selected from the group consisting of alcohols (e.g., methanol, ethanol, isopropanol, etc.), EA, THF, ACN, DMF, DMSO, and Tol, preferably methanol.
In the inventionIn some embodiments, the reaction of step three is carried out in the presence of a base selected from NaH, t-BuOK, K2CO3、Cs2CO3MeONa, n-BuLi, DIPEA and Et3N, preferably MeONa.
In some embodiments of the invention, the reaction in step three is carried out at a temperature of-20 ℃ to 100 ℃, preferably at a temperature of 0 ℃ to 25 ℃.
In some embodiments of the invention, the reaction described in step four is carried out in an organic solvent selected from the group consisting of Diox, Tol, ACN, DMF and THF, preferably Diox.
In some embodiments of the invention, the reaction of step four is carried out under catalysis of a catalyst selected from the group consisting of Pd (PPh)3)4、Pd(OAc)2、Pd2(dba)3、Pd(PPh3)2Cl2、Pd(PPh3)2Cl2·CH2Cl2And Pd (dppf) Cl2Pd (PPh) is preferred3)4
In some embodiments of the invention, the reaction described in step four is carried out in the presence of a copper reagent selected from the group consisting of CuI, CuCl and CuBr, preferably CuI.
In some embodiments of the invention, the reaction in step four is carried out at a temperature of from-20 ℃ to 200 ℃, preferably from 80 ℃ to 150 ℃.
In some embodiments of the invention, the reaction in step five is a reduction reaction.
In some preferred embodiments of the invention, the reduction in step five is carried out in an organic solvent selected from THF, DMF, DCM, Tol, Diox and ACN, preferably THF.
In some preferred embodiments of the invention, the reduction reaction in step five is carried out in the presence of a reducing agent selected from LiAlH4DIBAL and NaBH4Preferably LiAlH4
In some preferred embodiments of the present invention, the reduction reaction in step five is carried out at a temperature of-20 ℃ to 50 ℃, preferably at a temperature of 0 ℃ to 25 ℃.
In some embodiments of the invention, the substitution reaction described in step six is carried out in an organic solvent selected from DCM, DMF, THF, Et2O and Tol, preferably DCM.
In some embodiments of the invention, the substitution reaction in step six is carried out in the presence of a halogenating agent selected from the group consisting of halogenated alkanes, trihalophosphines, trihalophosphine oxides, hydrohalic acids and DCSO, preferably DCSO.
In some embodiments of the invention, the substitution reaction in step six is carried out at a temperature of-20 ℃ to 50 ℃, preferably at a temperature of 0 ℃ to 25 ℃.
In some embodiments of the invention, the reaction in step seven is carried out in an organic solvent selected from DMF, DCM, THF and Tol, preferably DMF.
In some embodiments of the invention, the reaction in step seven is carried out in the presence of a base selected from the group consisting of NaH, t-BuOK, Cs2CO3And K2CO3NaH is preferred.
In some embodiments of the invention, the reaction in step seven is carried out at a temperature of from-20 ℃ to 50 ℃, preferably from 0 ℃ to 25 ℃.
[ 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, with the purpose of facilitating administration to an organism, facilitating absorption of the active ingredient, and thereby exerting biological activity. The term "pharmaceutically acceptable carrier" refers to an excipient that is administered with a therapeutic agent, and which is, within the scope of sound medical judgment, suitable for contact with the tissues of humans 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 may be used in the present invention include (but are not limited to): a) a diluent; b) a lubricant; c) a binder; d) a disintegrant; e) absorbents, coloring, flavoring and sweetening agents; f) an emulsifier or dispersant; and/or g) an agent that enhances absorption of the compound.
The present invention provides a pharmaceutical composition comprising a compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof.
In some embodiments, the above pharmaceutical compositions further comprise one or more pharmaceutically acceptable carriers.
The pharmaceutical compositions described above may act systemically and/or locally. For this purpose, they may be administered by a suitable route, for example by parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular, inhalation routes or by all means well known to those skilled in the art of medicine. The pharmaceutical compositions described above may be administered in combination with at least one other therapeutic agent having a therapeutic effect on a disease or condition.
The above administration route can be achieved by a suitable dosage form. Dosage forms that may be used 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.
The pharmaceutical composition described above may comprise 0.01mg to 1000mg of at least one compound of formula I described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixtures thereof.
The present invention also provides a process for the preparation of the above pharmaceutical compositions or corresponding formulation forms thereof, which comprises combining at least one compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, with one or more pharmaceutically acceptable carriers.
[ drugs ]
The term "pharmaceutical product" refers to a combination product comprising a therapeutic agent, optionally other therapeutic agents, and optionally packaging and/or instructions.
The present invention provides a pharmaceutical product comprising:
a) a compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, or a pharmaceutical composition as described above;
b) optionally packaging and/or instructions.
The aforementioned pharmaceutical product may comprise 0.01mg to 1000mg of at least one compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof.
The present invention also provides a process for the preparation of the above mentioned pharmaceutical products, which comprises combining a compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g. hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixtures thereof, or a pharmaceutical composition as described above, optionally with packaging and/or instructions.
[ medical use ]
The compound of the invention can show strong inhibition effect on UPS7, and can be used as UPS7 inhibitor. Accordingly, the present invention provides the use of a compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixtures thereof, or a pharmaceutical composition as described above, or a pharmaceutical product as described above, as an inhibitor of UPS 7.
In addition, the present invention provides the use of a compound of formula I as described above, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixtures thereof, or a pharmaceutical composition as described above, or a pharmaceutical product 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 UPS7, particularly cancer.
The term "a disease or condition mediated at least in part by UPS 7" refers to a disease that develops mechanisms that include at least a portion of the factors associated with UPS7, including, but not limited to, cancer, neurodegenerative diseases (e.g., alzheimer's disease, parkinson's disease, etc.), diabetes, bone and joint diseases, joint inflammatory conditions, osteoporosis, immunological conditions, cardiovascular disease, ischemic disease, viral infections and diseases, bacterial infections and diseases, and the like.
[ method of treatment ]
The present invention provides a method for the prevention and/or treatment of a disease, in particular cancer, mediated at least in part by UPS7, comprising the steps of: administering to a subject in need thereof a prophylactically and/or therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixtures thereof, or a pharmaceutical composition thereof, or a pharmaceutical product thereof.
The term "effective amount" refers to a dose that is capable of inducing a biological or medical response in a cell, tissue, organ or organism (e.g., an individual) and is sufficient to achieve a desired prophylactic and/or therapeutic effect. The dosing regimen may be adjusted to provide the best response. For example, it may be administered in a single dose, may be administered in divided doses over time, or may be administered after proportionally decreasing or increasing the dose as the case may be. It will be appreciated that for any particular individual, the specific dosage regimen will be adjusted as required and at the discretion of the attendant person. In addition, a distinction is also made between prophylactic and therapeutic applications. In prophylactic applications, relatively low doses are typically administered chronically at relatively long intervals. In therapeutic applications, then, relatively high doses are typically administered at relatively short intervals until the progression of the disease is delayed or halted, preferably until the individual exhibits a partial or complete improvement in symptoms.
In the present invention, suitable in vitro or in vivo assays are performed to determine the efficacy of the compounds, pharmaceutical compositions and/or medicaments and whether administration is suitable for treating a disease or condition in an individual. Examples of such assays are described in the non-limiting examples below. Typically, an effective amount of a compound sufficient to achieve a prophylactic and/or therapeutic effect is from about 0.001 mg/kg body weight/day to about 10,000 mg/kg body weight/day. Where appropriate, an effective amount is from about 0.01 mg/kg body weight/day to about 1,000 mg/kg body weight/day. In more general terms, an effective amount is about 0.01 to 1,000 mg/kg body weight per day, every second day, or every third day, preferably about 0.1 to 500 mg/kg body weight. Exemplary treatment regimens are once every two days or once a week or once a month.
The term "treating" refers to alleviating or eliminating the disease or disorder in question. A subject is successfully "treated" if the subject receives a therapeutic amount of a compound of the invention or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of the invention, and the subject exhibits an observable and/or detectable remission and/or improvement of at least one of the indications and symptoms. It is understood that treatment includes not only complete treatment, but also less than complete treatment, but achieves some biologically or medically relevant result. In particular, "treatment" means that the compound of the invention or a pharmaceutically acceptable form thereof or the pharmaceutical composition of the invention can achieve at least one of the following effects: (1) preventing disease from occurring in an animal that may be predisposed to the disease but has not yet experienced or exhibited disease pathology or symptomology; (2) inhibiting disease in an animal experiencing or exhibiting disease pathology or symptomatology (i.e., arresting further development of pathology and/or symptomatology); (3) ameliorating the disease (i.e., reversing pathology and/or symptomatology) in an animal experiencing or exhibiting disease pathology or symptomatology.
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 site of a cell, tissue, organ, biological fluid, etc. thereof, such that the pharmaceutically active ingredient or pharmaceutical composition contacts the subject or the site of a cell, tissue, organ, biological fluid, etc. thereof. 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 by a physician or other caregiver that an individual needs or will benefit from a prophylactic and/or therapeutic procedure, the judgment being made based on various factors within the physician's or other caregiver's expertise.
The term "individual" (or subject) refers to a human or non-human animal. The subject of the invention includes both subjects (patients) suffering from a disease and/or disorder and normal subjects. Non-human animals of the invention include all vertebrates, e.g., non-mammals, such as birds, amphibians, reptiles, and the like, and mammals, e.g., non-human primates, livestock, and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, and the like).
In order to make the objects and technical solutions of the present invention clearer, embodiments of the present invention will be described in detail below with reference to examples. It will be understood by those skilled in the art that the following examples are illustrative of the present invention only and should not be taken as limiting the scope of the invention.
The reagents and instruments used in the examples are not indicated by the manufacturer, and are all conventional products commercially available. Those who do not have specific conditions noted are conducted under conventional conditions or conditions recommended by the manufacturer. The term "room temperature" used in the present invention means 20 ℃. + -. 5 ℃. As used herein, the term "about" when used in reference to a value or range of values is intended to encompass the value or range of values and tolerances acceptable to those skilled in the art for that value or range of values, e.g., within ± 10%, ± 5%, ± 4%, ± 3%, ± 2%, ± 1%, ± 0.5%, etc.
In the conventional syntheses as well as in the examples and intermediate syntheses, the meanings of the abbreviations are shown in the following table.
Abbreviations Means of Abbreviations Means of
DMF N, N-dimethylformamide Boc Tert-butyloxycarbonyl radical
Tf Trifluoromethanesulfonyl radical THF Tetrahydrofuran (THF)
DIAD Diisopropyl azodicarboxylate DEAD Azodicarboxylic acid diethyl ester
DCM Methylene dichloride DMSO Dimethyl sulfoxide
DIPEA N, N-diisopropylethylamine Pd2(dba)3 Tris (dibenzylideneacetone) dipalladium
Pd(PPh3)2Cl2 Bis (triphenylphosphine) palladium dichloride Pd(dppf)Cl2 [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
h Hour(s) PPh3 Triphenylphosphine
ADDP Azodicarbonyldipiperidine Tol Toluene
Et2O Ether (A) n-BuLi N-butyl lithium
t-BuLi Tert-butyl lithium LiHMDS Lithium bis (trimethylsilyl) amide
NaHMDS Bis (trimethylsilyl) amide sodium salt EA Ethyl acetate
ACN Acetonitrile Et3N Triethylamine
t-BuOK Potassium tert-butoxide MeONa Sodium methoxide
Diox 1, 4-dioxane Pd(PPh3)4 Tetrakis (triphenylphosphine) palladium
Pd(OAc)2 Palladium acetate DIBAL Diisobutylaluminum hydride
DCSO Thionyl chloride
The structures of the compounds described in the following examples were determined by nuclear magnetic resonance (1H-NMR) and/or Mass Spectrometry (MS).
Nuclear magnetic resonance (1H-NMR) was measured using a Bruker 400MHz NMR spectrometer, and deuterated methanol (CD) was used as a measuring solvent3OD), deuterated chloroform (CDCl)3) Hexadeuterio dimethyl sulfoxide (DMSO-d)6) Or deuterated water (D)2O), the internal standard substance is fourMethylsilane (TMS).
Abbreviations in the Nuclear Magnetic Resonance (NMR) data in the following examples represent the following meanings:
s: unimodal (singlet), d: doublet (doublt), t: triplet (triplet), q: quartet (quartz), dd: doublet (doubledoubledoublet), qd: quartet doubtet, ddd: double doublet (double doublet), ddt: double double triplet (double double triple), dddd: double double doublet (double double doublet), m: multiplet (multiplex), br: broad (broad), J: coupling constant, Hz: hertz, δ: chemical shift.
All chemical shift (δ) values are given in parts per million (ppm).
Mass Spectrometry (MS) was performed using an Agilent 6120B mass spectrometer with an electrospray ion source (ESI).
Preparative thin layer chromatography (Prep-TLC) purification was performed in the examples of the invention using the following procedure:
a thin-layer chromatography silica gel plate (the thickness is 1mm, the specification is 200 multiplied by 200mm, the binder is sodium carboxymethyl cellulose) produced by Nicotiana Summinck chemical company is adopted, a detection instrument is a ZF-20D dark box type ultraviolet analyzer, and the detection wavelength is 254nm and/or 365 nm.
The examples of the invention were purified by preparative high performance liquid chromatography (Prep-HPLC) using the following procedure:
the method A comprises the following steps:
a chromatographic column: waters SunAire Prep C18OBD 5 μm 19X 150 mm;
mobile phase A: acetonitrile; mobile phase B: water (containing 0.05 wt% formic acid);
time (min) Mobile phase A (v%) Mobile phase B (v%) Flow rate (mL/min)
0.00 10.0 90.0 28
18.00 90.0 10.0 28
The method B comprises the following steps:
a chromatographic column: waters SunAire Prep C18OBD 5 μm 19X 150 mm;
mobile phase A: acetonitrile; mobile phase B: water (containing 0.05 wt% ammonium bicarbonate);
time (min) Mobile phase A (v%) Mobile phase B (v%) Flow rate (mL/min)
0.00 10.0 90.0 28
18.00 90.0 10.0 28
[ preparation and identification of Compounds ]
The first embodiment is as follows: synthesis of 3- ((3- (5-chloro-3-methyl-2- (piperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (Compound 1).
Figure BDA0002539269000000231
The method comprises the following steps: synthesis of tert-butyl 4- (2-bromo-4-chloro-6-methylphenoxy) piperidine-1-carboxylate (Compound 1-2):
compound 1-1(8.80g, 39.75mmol), 4-hydroxypiperidine-1-carboxylic acid tert-butyl ester (4.00g, 19.87mmol), PPh3(10.43g, 39.75mmol) and dry THF (240mL) were added to the reaction flask in sequence. DIAD (7.8mL, 39.75mmol) was added dropwise at 0 ℃. After dropping, slowly raising the temperature to room temperature for reaction for 16 h. The reaction solution was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 23/2, v/v) to give the title compound 5.45 g.
ESI-MS:m/z 347.8,349.8[M-56+H]+
Step two: synthesis of tert-butyl 4- (4-chloro-2-methyl-6- (tributyltin) phenoxy) piperidine-1-carboxylate (Compound 1-3):
a solution of n-BuLi in n-hexane (2.5mol/L, 7.41mL) was added dropwise to a solution of compound 1-2(5.00g, 12.35mmol) in Tol (50mL) at-40 ℃. After 0.5h of reaction, tributyltin chloride (4.02mL, 14.82mmol) was added dropwise. After dropping, slowly raising the temperature to 0 ℃ for reaction for 3 h. The reaction was quenched with water (20mL), extracted with EA (3 × 25mL), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 20/1, v/v) to give the title compound 6.31 g.
1H-NMR(400MHz,CDCl3):δ7.16-7.04(m,2H),4.11(brs,2H),3.99-3.90(m,1H),2.69(t,J=11.9Hz,2H),2.23(s,3H),1.89(d,J=11.2Hz,2H),1.59-1.48(m,7H),1.46(s,9H),1.45-1.39(m,1H),1.32(h,J=7.3Hz,6H),1.17-0.98(m,6H),0.89(t,J=7.3Hz,9H)。
Step three: synthesis of methyl 3-iodo-2-methyl-2H-thieno [3,2-c ] pyrazole-5-carboxylate (Compounds 1-5):
compounds 1-4(1.00g, 3.25mmol) were added to methanol (30 mL). A solution of MeONa in methanol (1mL, 33 wt%) was added dropwise at room temperature and the reaction was allowed to proceed for 0.5 h. Further, methyl iodide (0.61mL, 9.74mmol) was added and the reaction was continued for 2 hours, followed by addition of methyl iodide (0.61mL, 9.74mmol) and further reaction for 2 hours. The reaction solution was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: dichloromethane ═ 100%, v) to obtain the title compound 484 mg.
1H-NMR(400MHz,CDCl3):δ7.89(s,1H),4.13(s,3H),3.92(s,3H)。
ESI-MS:m/z 322.9[M+H]+
Step four: synthesis of methyl 3- (2- (1- (tert-butoxycarbonyl) piperidin-4-yloxy) -5-chloro-3-methylphenyl) -2-methyl-2H-thieno [3,2-c ] pyrazole-5-carboxylate (Compounds 1-6):
a reaction flask is sequentially added with compound 1-5(350mg, 1.09mmol), compound 1-3(802mg, 1.30mmol), CuI (414mg, 2.17mmol) and Diox (17.5mL), and Pd (PPh) is added under the protection of nitrogen3)4(251mg, 0.22mmol), and the temperature is raised to 110 ℃ for reaction for 8 h. Stopping heating, and cooling to room temperature. Filtration and concentration of the filtrate under reduced pressure followed by purification by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 3/1, v/v) gave 466mg of the title compound.
ESI-MS:m/z 519.8,521.8[M+H]+
Step five: synthesis of tert-butyl 4- (4-chloro-2- (5- (hydroxymethyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-3-yl) -6-methylphenoxy) piperidine-1-carboxylate (Compounds 1-7):
compound 1-6(400mg, 0.77mmol) was dissolved in THF (10mL) and LiAlH was added dropwise at 0 deg.C4Solution of THF (0.6mL, 1.54 mmol). After dropping, continue to reverseThe time is 1.5 h. The reaction was quenched with water (1mL), and the reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 2/1, v/v) to give the title compound 314 mg.
ESI-MS:m/z 491.8,493.8[M+H]+
Step six: synthesis of tert-butyl 4- (4-chloro-2- (5- (chloromethyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-3-yl) -6-methylphenoxy) piperidine-1-carboxylate (Compounds 1-8):
compounds 1-7(150mg, 0.30mmol) were dissolved in DCM (3mL), cooled to 0 deg.C, DCSO (0.04mL, 0.61mmol) was added and stirring continued for 0.5 h. The reaction solution was concentrated under reduced pressure to give the title compound (150 mg).
ESI-MS:m/z 509.6,511.7[M+H]+
Step seven: synthesis of tert-butyl 4- (4-chloro-2- (5- ((6, 6-dimethyl-2, 4-dioxo-3-azabicyclo [3.1.0] hexan-3-yl) methyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-3-yl) -6-methylphenoxy) piperidine-1-carboxylate (Compounds 1-9):
6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (82mg, 0.59mmol) was dissolved in DMF (1.5mL), cooled to 0 deg.C, and NaH (23mg, 0.59mmol, 60%) was added and reacted for 0.5 h. Then, a solution of 1-8(150mg,0.29mmol) in DMF (1.5mL) was added dropwise, and the mixture was warmed to room temperature and reacted for 4.5 h. The reaction was quenched with water, diluted with EA (2mL), washed with water (3X 5mL), the organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure, purified by preparative high performance liquid chromatography (method A), the preparation was concentrated under reduced pressure, and lyophilized to give the title compound 129 mg.
ESI-MS:m/z 612.8,614.8[M+H]+
Step eight: synthesis of 3- ((3- (5-chloro-3-methyl-2- (piperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (compound 1) hydrochloride salt:
to a reaction flask was added compound 1-9(100mg, 0.13mmol), followed by addition of hydrochloric acid/ethyl acetate solution (2mL, 4mol/L) and reaction at room temperature for 1 h. After the reaction solution was concentrated under reduced pressure, it was separated and purified by preparative high performance liquid chromatography (method A), the preparation solution was concentrated under reduced pressure, hydrochloric acid (0.5mL, 1mol/L) was added to the preparation solution, and freeze-dried to give the title compound hydrochloride as 68 mg.
1H-NMR(400MHz,DMSO-d6):δ8.68(brs,1H),8.33(brs,1H),7.52(d,J=2.5Hz,1H),7.41(d,J=2.6Hz,1H),7.10(s,1H),4.65(s,2H),3.90(s,3H),3.79(dt,J=7.7,4.1Hz,1H),2.87-2.60(m,4H),2.57(s,2H),2.34(s,3H),1.73-1.59(m,2H),1.44-1.28(m,2H),1.16(s,3H),0.99(s,3H)。
ESI-MS:m/z 513.0,515.0[M+H]+
Example two: synthesis of 3- ((3- (5-chloro-3-methyl-2- (1-methylpiperidin-4-yloxy) phenyl) -2-methyl-2H-thieno [3,2-c ] pyrazol-5-yl) methyl) -6, 6-dimethyl-3-azabicyclo [3.1.0] hexane-2, 4-dione (compound 8).
Figure BDA0002539269000000251
The hydrochloride salt of compound 1 (26mg, 0.05mmol) was added to the reaction flask and dissolved in acetonitrile (1 mL). Aqueous formaldehyde (0.05mL, 37-40% purity) was added dropwise and reacted at room temperature for 20 minutes, followed by addition of sodium cyanoborohydride (9mg, 0.14mmol) and reaction at room temperature for 3 hours. The reaction was quenched by addition of saturated aqueous sodium bicarbonate, extracted with EA (3 × 2mL), the organic phases combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated, purified by preparative high performance liquid chromatography (method B), and the preparation lyophilized to give the title compound 14 mg.
1H-NMR(400MHz,CD3OD):δ7.38(d,J=2.4Hz,1H),7.22(d,J=2.6Hz,1H),7.03(s,1H),4.69(s,2H),3.88(s,3H),3.54(brs,1H),2.44(s,2H),2.34(s,3H),2.33-2.11(m,2H),2.06(s,3H),1.89(brs,2H),1.51(brs,2H),1.43(brs,2H),1.20(s,3H),1.04(s,3H)。
ESI-MS:m/z 527.2,529.1[M+H]+
[ pharmacological Activity test ]
The first test example: USP7 (protease) in vitro enzymatic activity inhibition assay.
1. The test system comprises:
protease: his6-USP7(Boston biochem);
substrate: ubiquitin-rhodamine 110(boston biochem);
buffer solution: 5 × USP7 assay Buffer (BPS).
2. Test parameters are as follows:
concentration of USP 7: 3 nM;
ubiquitin-rhodamine 110 concentration: 100 nM;
buffer system: 1.25 × USP7 assay buffer; 0.06% BSA; 1mM DTT; ddH2O;
Reaction temperature: 25 ℃;
compound and enzyme incubation time: 20 min;
enzyme kinetic reaction time: 20 min;
parameters of the microplate reader: the BMG PHORAStar FS luciferase label instrument has an excitation wavelength of 485nm and an emission wavelength of 520 nm.
3. The test method comprises the following steps:
the assay was performed according to kit instructions, with the following steps:
test group: and (3) incubating the mixture of the compound to be detected and the protease USP7 for 20min at 25 ℃ in a buffer solution system, adding a substrate ubiquitin-rhodamine 110 to start reaction, and reading the fluorescence value of each hole by adopting an enzyme kinetics method.
Negative group: the test compound was replaced with 0.2% DMSO in water and the experimental procedure was the same as for the test group.
Blank group: test compounds were replaced with 0.2% aqueous DMSO and the test was performed in the same manner as the test group without protease USP 7.
4. Data processing:
the relative inhibitory activity of each concentration group was calculated, and the inhibition ratio (%) — 100% - (fluorescence value of test group-fluorescence value of blank group)/(fluorescence value of negative group-fluorescence value of blank group) × 100%. Half maximal Inhibitory Concentration (IC) of the compound was calculated according to a four parameter model fitting curve50)。
5. And (3) test results:
the inhibition of USP7 activity by the compounds of the present invention was determined according to the above method and the results are shown in table 1.
TABLE 1 results of enzyme activity inhibition test of USP7
Example numbering IC50(nM)
1 13.39±0.04
2 36.26±2.52
In the enzyme activity inhibition test of USP7, the compound of the invention shows extremely strong inhibitory activity.
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, including all patents, patent applications, journal articles, books, and any other publications, cited in this disclosure is hereby incorporated by reference in its entirety.

Claims (8)

1. A compound having the structure of formula I or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof,
Figure FDA0002539268990000011
wherein the content of the first and second substances,
R0selected from hydrogen, halogen, cyano, C1-6Alkyl and C1-6A haloalkyl group;
L1is absent or selected from C1-6Alkylene radical, C1-6Haloalkylene, C3-8Cycloalkylene and 3-8 membered heterocycloalkylene;
R1selected from hydrogen, hydroxy, halogen, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C6-10Aryl, 5-10 membered heteroaryl, -NRaRb
Figure FDA0002539268990000012
Wherein the content of the first and second substances,
Raand RbEach independently selected from hydrogen and C1-6Alkyl radical, C3-8Cycloalkyl, -C (O) -C1-6Alkyl, -C (O) -C3-8Cycloalkyl, -C (O) -C2-6Alkenyl, -S (O)q-C1-6Alkyl, -S (O)q-C3-8Cycloalkyl and-S (O)q-C2-6An alkenyl group; wherein, the C1-6Alkyl radical, C3-8Cycloalkyl and C2-6Each alkenyl group is independently optionally substituted with one or more of the following: hydrogen, -C1-6alkylene-C (O) OH and-C (O) O-C1-6An alkyl group; q is selected from 1 and 2;
b is selected from 3-8 membered heterocycloalkyl, 3-8 membered heterocycloalkenyl and 5-10 membered heteroaryl;
c is selected from C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C4-6Cycloalkenyl, 3-8 membered heterocycloalkenyl, C6-10Aryl and 5-10 membered heteroaryl;
d is selected from C3-8Cycloalkyl and 3-8 membered heterocycloalkyl;
if present, each R9And R10Each independently selected from hydrogen, halogen, cyano, oxo, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, carboxy, -C (O) O-C1-6Alkyl and-C1-6alkylene-C (O) OH; wherein, the C1-6Alkyl radical, C3-8Cycloalkyl and 3-8 membered heterocycloalkyl are each independently optionally substituted with one or more of the following: hydrogen, hydroxy and amino;
z3、z4and z5Each independently selected from 0, 1,2, 3, 4, 5 and 6;
R2、R3、R4、R5and R6Each independently selected from hydrogen, hydroxy, halogen, cyano, carboxy, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl radical, C1-6Alkoxy and C1-6A haloalkoxy group;
L2selected from oxygen, sulfur, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-, -S-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen, C1-6Alkyl radical, C1-6Haloalkyl, C3-8Cycloalkyl and-C (O) -C1-6An alkyl group;
a is selected from C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl;
if present, each R7And R8Each independently selected from hydrogen, hydroxy, halogen, cyano, -NRcRd、C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, -C1-6alkylene-OH, -C1-6alkylene-O-C1-6Alkyl, -C (O) -N (C)1-6Alkyl radical)2、-C(O)-C1-6Alkyl and-C (O) -C1-6Haloalkyl, or R7And R8And the atoms to which they are attached together form C3-8Cycloalkyl or 3-8 membered heterocycloalkyl; wherein the content of the first and second substances,
Rcand RdEach independently selected from hydrogen and C1-6Alkyl and-C (O) -C1-6An alkyl group;
z1and z2Each independently selected from 0, 1,2, 3, 4, 5, 6, 7, 8, 9 and 10.
2. The compound of claim 1, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof,
R0is hydrogen;
L1is C1-6An alkylene group;
R1is selected from
Figure FDA0002539268990000021
Wherein the content of the first and second substances,
b is selected from 3-8 membered heterocycloalkyl and 3-8 membered heterocycloalkenyl;
c is C3-6A cycloalkyl group;
if present, each R9Each independently selected from oxo and C1-6A haloalkyl group;
if present, each R10Each independently selected from hydrogen and C1-6An alkyl group;
R2、R3、R4、R5and R6Each independently selected from hydrogen, halogen and C1-6An alkyl group;
L2selected from-O-, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen and C1-6An alkyl group;
a is selected from C3-8Cycloalkyl and 3-8 membered heterocycloalkyl;
if present, each R7And R8Each independently selected from hydrogen, amino, C1-6Alkyl and C1-6Alkoxy, or R7And R8And the atoms to which they are attached together form C3-8A cycloalkyl group;
z1、z2、z3and z4Each independently selected from 0, 1 and 2.
3. The compound of claim 1, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof,
R0is hydrogen;
L1is methylene;
R1is composed of
Figure FDA0002539268990000022
Wherein the content of the first and second substances,
b is 5-membered heterocycloalkyl;
c is cyclopropyl;
R9is an oxo group;
R10is C1-6An alkyl group;
z3and z4Is 2;
R2、R3、R4、R5and R6Each independently selected from hydrogen, halogen and C1-6An alkyl group;
L2selected from-O-, -N (R)11)-、-C(O)-、-O-C1-6Alkylene-and C1-6An alkylene group; wherein the content of the first and second substances,
R11selected from hydrogen and C1-6An alkyl group;
a is selected from C4-5Cycloalkyl and 4-8 membered heterocycloalkyl ring;
if present, each R7And R8Each independently selected from hydrogen, amino, C1-6Alkyl and C1-6An alkoxy group;
z1and z2Each independently selected from 0 and 1.
4. The compound of claim 1, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof,
R0is hydrogen;
L1is methylene;
R1is composed of
Figure FDA0002539268990000031
R2And R4Is hydrogen;
R3is chlorine;
R5and R6Is methyl;
L2selected from the group consisting of-O-, -NH-, -C (O) -, -O-CH2-and-CH2-;
A is selected from
Figure FDA0002539268990000032
Figure FDA0002539268990000033
5. The following compounds or pharmaceutically acceptable salts, esters, solvates, stereoisomers, tautomers, polymorphs, isotopic labels, metabolites, prodrugs or mixtures thereof:
Figure FDA0002539268990000034
Figure FDA0002539268990000041
Figure FDA0002539268990000051
6. a pharmaceutical composition comprising a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, and one or more pharmaceutically acceptable carriers.
7. A pharmaceutical product, comprising:
a) a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug, or mixture thereof, or a pharmaceutical composition according to claim 6;
b) optionally packaging and/or instructions.
8. Use of a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, polymorph, isotopic label, metabolite, prodrug or mixture thereof, or a pharmaceutical composition according to claim 6, or a pharmaceutical product according to claim 7, in the manufacture of a medicament for the prevention and/or treatment of a disease or condition mediated at least in part by UPS 7.
CN202010542108.9A 2020-06-15 2020-06-15 Thienopyrazole compounds, pharmaceutical composition containing same and application thereof Pending CN113801135A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010542108.9A CN113801135A (en) 2020-06-15 2020-06-15 Thienopyrazole compounds, pharmaceutical composition containing same and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010542108.9A CN113801135A (en) 2020-06-15 2020-06-15 Thienopyrazole compounds, pharmaceutical composition containing same and application thereof

Publications (1)

Publication Number Publication Date
CN113801135A true CN113801135A (en) 2021-12-17

Family

ID=78892435

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010542108.9A Pending CN113801135A (en) 2020-06-15 2020-06-15 Thienopyrazole compounds, pharmaceutical composition containing same and application thereof

Country Status (1)

Country Link
CN (1) CN113801135A (en)

Similar Documents

Publication Publication Date Title
TWI766882B (en) Novel compounds
EP2964223B1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
US20230128824A1 (en) Spiro ring-containing quinazoline compound
US9688654B2 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
US11407740B2 (en) Antibacterial compounds
CN112390797A (en) Novel spirocyclic K-Ras G12C inhibitor
KR20140059164A (en) Tricyclic gyrase inhibitors
EP4071155A1 (en) Thiazololactam compound as erk inhibitor and use thereof
US20190084988A1 (en) Wdr5 inhibitors and modulators
WO2022095909A1 (en) Compound used as ntrk inhibitor and application thereof
US20220356186A1 (en) Perk inhibiting pyrrolopyrimidine compounds
CN113801135A (en) Thienopyrazole compounds, pharmaceutical composition containing same and application thereof
CN115433207A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN111808105B (en) Pyrimidinone pyrazolo compound containing fused ring group, preparation method and application thereof
EP4098647A1 (en) Disubstituted adamantyl derivative or pharmaceutically acceptable salt thereof, and pharmaceutical composition for suppressing cancer growth comprising same as active ingredient
WO2018075871A1 (en) Novel antibiotics and methods of using same
CN116390923A (en) Heterocyclic derivative and preparation method and application thereof
US20220332722A1 (en) Perk inhibiting compounds
CN113087724B (en) Isothiazolopyrimidinone compounds, pharmaceutical compositions containing the same and uses thereof
EP4353724A1 (en) Compound as cdk kinase inhibitor and use thereof
JP7010443B2 (en) Indolizine compounds, their production methods and uses
CN111247119B (en) Amidine and guanidine derivatives, preparation method and application thereof in medicines
EP4219453A1 (en) Pyrazole compound and preparation method therefor and use thereof
CN116102533A (en) Aromatic heterocyclic compound and application thereof
WO2022253316A1 (en) Quinoxaline derivative, preparation therefor and use 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