CN110372693B - Compound for targeted degradation of BET protein and application thereof - Google Patents

Compound for targeted degradation of BET protein and application thereof Download PDF

Info

Publication number
CN110372693B
CN110372693B CN201910594817.9A CN201910594817A CN110372693B CN 110372693 B CN110372693 B CN 110372693B CN 201910594817 A CN201910594817 A CN 201910594817A CN 110372693 B CN110372693 B CN 110372693B
Authority
CN
China
Prior art keywords
formula
compound
pharmaceutical composition
treating
independently
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
CN201910594817.9A
Other languages
Chinese (zh)
Other versions
CN110372693A (en
Inventor
饶燏
李孟鸿
兰天龙
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.)
Tsinghua University
Original Assignee
Tsinghua University
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
Priority claimed from CN201810726513.9A external-priority patent/CN108690020A/en
Application filed by Tsinghua University filed Critical Tsinghua University
Publication of CN110372693A publication Critical patent/CN110372693A/en
Application granted granted Critical
Publication of CN110372693B publication Critical patent/CN110372693B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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

Abstract

The invention discloses a compound for targeted degradation of BET protein and application thereof, and provides a compound which is a compound shown in a formula I or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof: X-Y-Z formula I wherein X represents a ligand for BET protein, Z represents a ligand for E3 ligase, and Y represents a chain linking X and Z.

Description

Compound for targeted degradation of BET protein and application thereof
Technical Field
The invention relates to the field of biomedicine, in particular to a compound for targeted degradation of BET protein and application thereof.
Background
Different types of cells that possess common gene sequences within an individual perform different biological functions because there are open and closed states of transcription of different genes between different types of cells. The epigenetic regulation of expression of this gene is regulated, in part, by acetylation of specific lysine residues on histones and other proteins. Bromodomain (BRD) is a transcription coactivator containing a conserved protein domain capable of specifically recognizing acetylated lysine (KAc) in histone, can recognize acetylated lysine and recruit transcription factors and other related proteins to specific gene transcription sites in chromatin, changes the activity of RNA polymerase II, and promotes gene transcription and downstream signal transmission. In some tumor and inflammatory states, epigenetic regulation of gene transcription is aberrant, resulting in aberrant expression of growth-promoting genes and proinflammatory factor genes. BET (Bromodomain and extra-terminal) proteins represent an entire family of proteins, the members of which include BRD2, BRD3, BRD4 and BRDT, each of which contains two Bromodomain domains BD I and BD II at the N-terminus and an extra-terminal domain (extra-terminal domain) at the C-terminus.
The most studied of the BET protein family members is the BRD4 protein at present. BRD4 is often located in a super enhancer region upstream of several important protooncogenes, such as c-MYC and bcl-xL, and plays an important role in the regulation of expression of these genes. Based on the important role played by BRD4 in the regulation and control of important protooncogene expression, BRD4 is expected to become an important target for the treatment of various tumors, such as acute myelocytic leukemia, multiple myeloma, Burkit's lymphoma and prostate cancer. Inhibitors of BRD4, such as (+) -JQ-1, i-BET and OTX-015, have been developed to show great therapeutic potential in preclinical animal models of a variety of tumors, including Burkit's lymphoma.
However, new drugs that are effective in inhibiting or degrading BET proteins are yet to be developed.
Disclosure of Invention
The present invention aims to solve at least to some extent one of the technical problems of the prior art:
the inventors have found that (+) -JQ-1 and OTX-015 lead to a rapid several-fold increase in BRD4 protein levels in a concentration-dependent manner, making inhibition of BRD4 more difficult. Even above IC50At ten-fold concentrations, (+) -JQ-1 and OTX-015 still do not sufficiently inhibit c-MYC. In addition, (+) -JQ-1 caused a decrease in c-MYC in AML cells, but the level of c-MYC recovered rapidly after (+) -JQ-1 withdrawal and was higher than before the dosing treatment. Although a number of BET small molecule inhibitors including OTX-015 and I-BET726 have been introduced into clinical trials. However, the effectiveness of these drugs remains to be tested due to insufficient inhibition of c-MYC and the ability of feedback mechanisms to upregulate BRD4 gene expression. Furthermore, in some The phenomenon of resistance of tumors to BET inhibitors was observed in preclinical studies of BET inhibitors.
Based on the discovery of the above problems, the inventors propose a novel compound, which adopts a double-target molecular structure, the structure of which is shown in fig. 1, the structure of one end of the molecule is targeted and combined with E3 ligase, the structure of the other end is targeted and combined with BET protein, the structures of the two ends are connected through a chain (linker) to form a complete compound molecule, the compound ubiquitinates a target protein through E3 and guides the target protein to enter a degradation pathway, and the specific degradation effect on the target protein is strong. The compound can efficiently degrade BET protein in various cell lines at a lower concentration, and can more effectively block the expression of protooncogene c-MYC; BET protein inhibitors do not affect BET protein abundance, BET proteins may rapidly regain activity upon withdrawal, whereas BET proteins require a certain amount of time to restore intracellular protein abundance even after withdrawal of the PROTAC molecule. In addition, the PROTAC molecule designed aiming at the BET protein can induce the artificial ubiquitination degradation of the BET protein when the natural degradation pathway of the BET protein fails, so that the drug resistance of tumor cells to a BET protein inhibitor is overcome.
To this end, in a first aspect of the present invention, the present invention provides a compound which is a compound represented by formula I or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof:
X-Y-Z
formula I
Wherein X represents a ligand of BET protein, Z represents a ligand of E3 ligase, and Y represents a chain connecting X and Z.
According to an embodiment of the present invention, the above compound may further comprise at least one of the following additional technical features:
according to an embodiment of the invention, X is a compound of formula II-1 or II-2,
Figure BDA0002117302440000021
Cy1or Cy2Each independently is a benzene ring, C6-12Aryl, heteroaryl of 5 to 12 ring atoms, C3-12Cycloalkyl or heterocyclyl consisting of 3 to 12 ring atoms;
L1is- (CR)mRw)g-O-(CRmRw)g-,-(CRmRw)g-S-(CRmRw)g-,-(CRmRw)g-N(R1a)-(CRmRw)g-,-(CRmRw)n-,-(CRmRw)g-(CR1a=CR1a)n-(CRmRw)g-,-(CRmRw)g-(C≡C)n-(CRmRw)g-,-(CRmRw)g-S(=O)p-(CRmRw)g-,-(CRmRw)g-C(=O)-(CRmRw)g-,-(CRmRw)g-C(=O)-O-(CRmRw)g-,-(CRmRw)g-S(=O)p-N(R1a)-(CRmRw)g-,-(CRmRw)g-C(=O)-N(R1a)-(CRmRw)g-;
Each Cy1Or Cy2Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh1Substituted;
each L1Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh2Substituted;
each Rh1Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, heterocyclyl consisting of 3 to 12 ring atoms, heteroaryl consisting of 5 to 12 ring atoms, Rg-(CRmRw)g-O-(CRmRw)g-,Rg-(CRmRw)g-S-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-,Rg-(CRmRw)g-(CR1a=CR1a)n-(CRmRw)g-,Rg-(CRmRw)g-(C≡C)n-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-O-(CRmRw)g-,Rg-(CRmRw)g-O-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-N(R1a)-(CRmRw)g-or Rg-(CRmRw)g-N(R1a)-C(=O)-(CRmRw)g-;
Each Rh2Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, heterocyclyl consisting of 3 to 12 ring atoms, heteroaryl consisting of 5 to 12 ring atoms, Rg-(CRmRw)g-O-(CRmRw)g-,Rg-(CRmRw)g-S-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-,Rg-(CRmRw)g-(CR1a=CR1a)n-(CRmRw)g-,Rg-(CRmRw)g-(C≡C)n-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-O-(CRmRw)g-,Rg-(CRmRw)g-O-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-N(R1a)-(CRmRw)g-or Rg-(CRmRw)g-N(R1a)-C(=O)-(CRmRw)g-;
Each Rh1Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh3Substituted;
each Rh2Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh4Substituted;
each Rh3Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl or C3-6A cycloalkyl group;
each Rh4Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl or C3-6A cycloalkyl group;
each R1aEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms;
each Rm、RwOr RgEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C 2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms;
each n is independently 1, 2, 3 or 4;
each g is independently 0, 1, 2, 3 or 4;
each p is independently 1 or 2.
According to an embodiment of the invention, Cy1Or Cy2Each independently is a benzene ring, C6-10Aryl, heteroaryl of 5 to 10 ring atoms, C3-6Cycloalkyl or heterocyclyl consisting of 3 to 12 ring atoms.
According to an embodiment of the invention, each Rh1Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms, heteroaryl consisting of 5 to 10 ring atoms, Rg-(CRmRw)g-O-(CRmRw)g-,Rg-(CRmRw)g-S-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-,Rg-(CRmRw)g-(CR1a=CR1a)n-(CRmRw)g-,Rg-(CRmRw)g-(C≡C)n-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-O-(CRmRw)g-,Rg-(CRmRw)g-O-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-N(R1a)-(CRmRw)g-or Rg-(CRmRw)g-N(R1a)-C(=O)-(CRmRw)g-;
Each Rh2Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms, heteroaryl consisting of 5 to 10 ring atoms, Rg-(CRmRw)g-O-(CRmRw)g-,Rg-(CRmRw)g-S-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-,Rg-(CRmRw)g-(CR1a=CR1a)n-(CRmRw)g-,Rg-(CRmRw)g-(C≡C)n-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-O-(CRmRw)g-,Rg-(CRmRw)g-O-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-N(R1a)-(CRmRw)g-or Rg-(CRmRw)g-N(R1a)-C(=O)-(CRmRw)g-;
Each Rh1Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh3Substituted;
each Rh2Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh4Substituted;
each R h3Each independently hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl or C3-6A cycloalkyl group;
each Rh4Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl or C3-6A cycloalkyl group.
According to an embodiment of the invention, each R1aEach independently is H, deuterium, F, Cl, Br, I, CN, NO2OH, amino, carboxyl, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms;
each Rm、RwOr RgEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms.
According to an embodiment of the invention, Cy1Or Cy2Are each independently
Figure BDA0002117302440000051
Figure BDA0002117302440000052
According to an embodiment of the invention, each Rh1Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2COOH, methyl, ethyl, n-propyl, isopropyl, n-propyl The group consisting of butyl, isobutyl, tert-butyl,
Figure BDA0002117302440000053
Figure BDA0002117302440000054
Rg-(CRmRw)g-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-O-(CRmRw)g-,Rg-(CRmRw)g-O-C(=O)-(CRmRw)g-,Rg-(CRmRw)g-S(=O)p-N(R1a)-(CRmRw)g-,Rg-(CRmRw)g-N(R1a)-S(=O)p-(CRmRw)g-,Rg-(CRmRw)g-C(=O)-N(R1a)-(CRmRw)g-or Rg-(CRmRw)g-N(R1a)-C(=O)-(CRmRw)g-;
Each Rh2Each independently hydrogen, deuterium, F, Cl, Br, I, oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
each Rh1Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh3Substituted;
each Rh2Are each independently substituted by 1, 2, 3, 4, 5 or 6Rh4Substituted;
each Rh3Each independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2COOH, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
each Rh4Independently represent hydrogen, deuterium, F, Cl, Br, I, CN,OH,NO2,NH2COOH, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
According to an embodiment of the invention, each R1aEach independently is H, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
each Rm、RwOr RgEach independently is H, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
According to an embodiment of the invention, X is a compound of formula III-1, III-2, III-3, III-4, III-5 or III-6,
Figure BDA0002117302440000061
according to an embodiment of the invention, Z is a compound of formula IV,
Figure BDA0002117302440000071
Wherein Q is N or CR2
M is C (R)eRf),N(R1b) O or S;
w and K are each independently C (R)eRf) NH, O or S;
R2hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl or C2-6An alkynyl group;
each R2aOr R2bEach independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2COOH, oxo, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, hetero having 3 to 12 ring atomsCyclyl or heteroaryl of 5 to 12 ring atoms;
each R2cEach independently is hydrogen, deuterium, F, Cl, Br, I, CN, OH, NO2,NH2,COOH,C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 12 ring atoms;
each Re、RfEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms;
each R1bEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-6Alkyl radical, C 1-6Haloalkyl, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 12 ring atoms;
when W or K is NH, the R2a(ii) is not substituted at the position of said W or K;
n1、n2each independently is 0, 1, 2 or 3;
n3is 0, 1, 2, 3, 4 or 5.
According to an embodiment of the invention, each Re、RfEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms;
each R1bEach independently is H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-6Cycloalkyl radical, C6-10Aryl, heterocyclyl consisting of 3 to 12 ring atoms or heteroaryl consisting of 5 to 10 ring atoms.
According to an embodiment of the invention, R2Hydrogen, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
According to an embodiment of the invention, each R2a、R2bEach independently is hydrogen, deuterium, oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
Each R2cEach independently hydrogen, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
According to an embodiment of the present invention, each Re、RfEach independently hydrogen, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl;
each R1bEach independently is H, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
According to an embodiment of the invention, Z is a compound of formula V-1, V-2, V-3 or V-4,
Figure BDA0002117302440000081
according to an embodiment of the present invention, Y is a group containing 1 to 30 atoms including at least one selected from a carbon atom, a sulfur atom, an oxygen atom, a nitrogen atom, and a selenium atom.
According to an embodiment of the invention, said Y is optionally substituted C1-20Alkyl radical, C1-20Haloalkyl, C1-20Alkoxy radical, C2-20Alkenyl radical, C2-20Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, heterocyclic group composed of 3-12 ring atoms or heteroaryl composed of 5-12 ring atoms. In some embodiments, each C1-20Alkyl radical, C1-20Haloalkyl, C1-20Alkoxy radical, C2-20Alkenyl radical, C2-20Alkynyl, C3-12Cycloalkyl radical, C6-12Aryl, heterocyclyl of 3 to 12 ring atoms or heteroaryl of 5 to 12 ring atoms independently by C 1-8alkyl-C (═ O) -NH-.
According to an embodiment of the invention, said Y is optionally substituted
Figure BDA0002117302440000082
Wherein x is1-x23Each of which is independently a key of the electronic device,
Figure BDA0002117302440000083
Figure BDA0002117302440000091
R1dis H, deuterium, F, Cl, Br, I, CN, -NO2OH, amino, carboxyl or C1-4An alkyl group.
In some embodiments, each x1-x23Independently by C1-6alkyl-C (═ O) -NH-.
According to an embodiment of the invention, Y is a compound of formula VI-1 or VI-2,
Figure BDA0002117302440000092
each r is an integer of 0-12 independently;
each k is an integer of 0-12 independently;
each j is an integer of 0-12 independently;
each t1Or t3Each of which is independently a key, and each of which is independently a bond,
Figure BDA0002117302440000093
optionally substituted
Figure BDA0002117302440000094
Each t2Or t4Each of which is independently a bond, and each of which is independently a bond,
Figure BDA0002117302440000095
optionally substituted
Figure BDA0002117302440000096
Figure BDA0002117302440000097
t5Is a bond or optionally substituted
Figure BDA0002117302440000098
R1dIs H, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
In some embodiments, each t1、t2、t3、t4Or t5Independently by C1-4alkyl-C (═ O) -NH-.
According to an embodiment of the present invention, when Y is a compound represented by formula VI-1,
Figure BDA0002117302440000099
wherein:
Figure BDA0002117302440000101
is composed of
Figure BDA0002117302440000102
Figure BDA0002117302440000103
k=k1+2 or
k=k2+1 or
k=k3+1 or
k=k4+2。
According to an embodiment of the invention, said Y is a compound represented by at least one of the following,
Figure BDA0002117302440000104
Figure BDA0002117302440000111
each t6、t7、t8、t9Or t10Each independently is an integer of 0 to 11.
In a second aspect of the present invention, the present invention provides a compound represented by any one of formulas 1 to 50, or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug thereof,
Figure BDA0002117302440000112
Figure BDA0002117302440000121
Figure BDA0002117302440000131
Figure BDA0002117302440000141
Figure BDA0002117302440000151
Figure BDA0002117302440000161
Figure BDA0002117302440000171
Figure BDA0002117302440000181
Figure BDA0002117302440000191
Figure BDA0002117302440000201
Figure BDA0002117302440000211
In a third aspect of the invention, a pharmaceutical composition is provided. According to an embodiment of the invention, the pharmaceutical composition comprises a compound according to any of the above.
According to an embodiment of the present invention, the above pharmaceutical composition may further comprise at least one of the following additional technical features:
according to an embodiment of the invention, the pharmaceutical composition further comprises an adjuvant.
According to an embodiment of the present invention, the pharmaceutical composition further comprises other drugs for treating or preventing non-hodgkin's lymphoma, Burkitt's lymphoma, acute myelogenous leukemia, multiple myeloma, lung cancer, prostate cancer, and NUT midline cancer.
According to an embodiment of the invention, the other drug for treating or preventing non-hodgkin's lymphoma comprises ibrutinib.
According to an embodiment of the invention, the other medicament for treating or preventing Burkitt's lymphoma comprises at least one selected from cyclophosphamide, doxofigucine.
According to an embodiment of the present invention, the other agent for treating or preventing acute myelogenous leukemia comprises at least one selected from cytarabine, Azacitidine, Decitabine.
According to an embodiment of the present invention, the other medicament for treating or preventing multiple myeloma comprises at least one selected from carfilzomib, thalidomide, lenalidomide, pomalidomide.
According to an embodiment of the present invention, the other drug for treating or preventing lung cancer comprises at least one selected from gefitinib, erlotinib, osthole, afatinib.
According to an embodiment of the present invention, the other medicament for treating or preventing prostate cancer comprises at least one selected from flutamide and nilutamide.
In a fourth aspect of the invention, the invention proposes the use of a compound according to any one of the above or a pharmaceutical composition according to any one of the above for the manufacture of a medicament for degrading BET proteins.
In a fifth aspect, the invention provides the use of a compound as described in any one of the preceding claims or a pharmaceutical composition as described in any one of the preceding claims in the manufacture of a medicament for the treatment or prophylaxis of non-hodgkin's lymphoma, Burkitt's lymphoma, acute myeloid leukaemia, multiple myeloma, lung cancer, prostate cancer and NUT midline cancer.
In a sixth aspect of the invention, a method of degrading a BET protein is provided. According to an embodiment of the invention, the method comprises: contacting a BET protein with a compound described above or a pharmaceutical composition described in any of the above.
In a seventh aspect of the invention, the invention features a method of treating or preventing non-hodgkin's lymphoma, Burkitt's lymphoma, acute myelogenous leukemia, multiple myeloma, lung cancer, prostate cancer, and NUT midline cancer. According to an implementation of the invention, the method comprises: administering to the patient a compound according to any one of the above or a pharmaceutical composition according to any one of the above.
The HBL-1 cell line is a non-Hodgkin lymphoma cell, ibrutinib is the most commonly used drug for clinically treating non-Hodgkin lymphoma and has an excellent treatment effect, the target point of ibrutinib is BTK protein, and the ibrutinib occupies an ATP binding pocket of the BTK protein and inhibits the activity of BTK by covalent crosslinking of an acrylamide functional group at the tail end of a molecule and a cysteine residue at the 481-position of the BTK protein. The C481S mutation in the BTK protein caused the cell to acquire resistance to ibrutinib. In one aspect, compounds according to embodiments of the invention exhibit strong degradation of BET proteins (DCs) from a variety of cell lines50<100nM) and can significantly degrade the expression product c-MYC protein of the protooncogene c-MYC downstream of BRD 4. On the other hand, the compounds according to the examples of the present invention showed a very strong Degradation (DC) of the BET protein of HBL-1(BTK-C481S) cell line50<1nM) and shows a very strong killing effect (IC) on HBL-1(BTK-C481S) cell line in MTT experiment50As low as 0.66 nM). Thus, the compounds of the present embodiments have the potential to treat ibrutinib-resistant non-hodgkin's lymphoma.
Drawings
FIG. 1 is a schematic diagram of the basic technical route for PROTACs (proteolytic targeting chimeras) according to an embodiment of the present invention;
FIG. 2 is a schematic diagram of the preparation of compounds of the present invention by a click reaction (click reaction) or an amide condensation reaction according to an embodiment of the present invention;
FIG. 3 is a schematic representation of the degradation of Jurkat cell lines BRD4 and BRD2 by compounds of formula 1, formula 9 and formula 13 and a literature report compound ARV-825, according to an example of the present invention;
FIG. 4-1 is a graphical representation of the results of the BET protein degradation of the compound of formula 2 as affected by other compounds, such as carfilzomib (Cfz), in accordance with an embodiment of the present invention;
FIG. 4-2 is a graphical representation of the results of the BET protein degradation of the compound of formula 2 as affected by other compounds such as (+) -JQ-1, ABBV-075 and pomalidomide (Poma) in accordance with an embodiment of the present invention;
FIG. 5-1 is a schematic diagram of the degradation of BET protein by a compound of formula 2, formula 5, formula 10, formula 13, or formula 15 in cell line HBL-1 according to an embodiment of the present invention;
FIG. 5-2 is a schematic diagram of the degradation of BET protein by Ramos in a cell line of a compound of formula 2, formula 5, formula 10, formula 13, or formula 15 according to an embodiment of the present invention;
FIGS. 5-3 are schematic illustrations of the degradation of BET protein by compounds of formula 2, formula 5, formula 10, formula 13, or formula 15 in cell line IgE MM, according to embodiments of the present invention;
FIGS. 5-4 are schematic illustrations of the degradation of BET protein by compounds of formula 2, formula 5, formula 10, formula 13, or formula 15 in cell line RPMI, in accordance with embodiments of the present invention;
FIG. 6-1 is a graph showing the results of the effect of the compound of formula 2 and (+) -JQ-1, ARV-825, ABBV-075, etc. on the protooncoprotein c-myc in Ramos cell lines treated for 2 hours according to the example of the present invention;
FIG. 6-2 is a graph showing the results of the effect of the compound of formula 2 and (+) -JQ-1, ARV-825, ABBV-075, etc. on the protooncoprotein c-myc in a Ramos cell line treated for 6 hours according to the example of the present invention;
FIG. 7 is a graph showing the BET protein reduction effect of the compound represented by formula 10 and ARV-825 (Amersham pharmacia Biotech) on HBL-1(BTK-C481S) cell line (incubated for 2 hours) at low concentration according to the example of the present invention;
FIG. 8-1 is a graph showing the killing effect of the negative control of formula 2, the compounds of formula 2 and 10, (+) -JQ-1, ARV-825 and ABBV-075 etc. on cells in HBL-1(BTK-C481S) cell line according to the example of the present invention; and
FIG. 8-2 is a graph showing the results of cell killing by the negative control of formula 2, the compounds of formula 2 and 10, (+) -JQ-1, ARV-825 and ABBV-075 etc. in HBL-1(BTK-C481S) cell line according to the example of the present invention;
FIG. 9-1 is a schematic diagram showing the degradation of BET protein by the compounds of formula 16, formula 18, formula 19, formula 20, formula 22, and formula 23 in the cell line Jurkat according to the example of the present invention;
FIG. 9-2 is a graph showing the degradation of BET protein by the compounds of formula 24, formula 25, formula 26, formula 27, formula 28, formula 29, and formula 30 in the cell line Jurkat according to the example of the present invention;
FIG. 9-3 is a graph showing the degradation of BET protein by the compounds of formula 16, formula 18, formula 19, formula 20, formula 21, formula 22, and formula 23 in the cell line Jurkat according to the example of the present invention;
FIG. 10-1 is a graph showing the comparison of the activities of the compounds represented by formula 2, formula 10, formula 21 and formula 24 on the lung cancer K562 cell line according to the example of the present invention;
fig. 10-2 is a graphical representation comparing the activity of compounds of formula 2, formula 10, formula 21, and formula 24 on prostate cancer LNcap cell lines, according to embodiments of the present invention.
Detailed Description
The following detailed description of embodiments of the invention is intended to be illustrative, and is not to be construed as limiting the invention.
As used herein, the term "administering to a patient a previously described compound, or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof, or a previously described pharmaceutical composition" refers to introducing a predetermined amount of a substance into the patient by some suitable means. The compound of formula I, formula II, formula III, formula IV, formula V or formula VI of the present invention or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof, or pharmaceutical composition thereof may be administered by any common route as long as it can reach the intended tissue. Various modes of administration are contemplated, including peritoneal, intravenous, intramuscular, subcutaneous, cortical, oral, topical, nasal, pulmonary and rectal, but the invention is not limited to these exemplified modes of administration.
The administration frequency and dose of the pharmaceutical composition of the present invention can be determined by a number of relevant factors, including the type of disease to be treated, the administration route, the age, sex, body weight and severity of the disease of the patient and the type of drug as an active ingredient.
The term "treatment" is used to refer to obtaining a desired pharmacological and/or physiological effect. The effect may be prophylactic in terms of complete or partial prevention of the disease or symptoms thereof, and/or may be therapeutic in terms of a partial or complete cure for the disease and/or adverse effects caused by the disease. As used herein, "treatment" encompasses the treatment of diseases in mammals, particularly humans, including: (a) preventing the occurrence of a disease or disorder in an individual who is susceptible to the disease but has not yet been diagnosed with the disease; (b) inhibiting the disease; or (c) alleviating the disease, e.g., alleviating symptoms associated with the disease. As used herein, "treatment" encompasses any administration of a drug or compound to a subject to treat, cure, alleviate, ameliorate, reduce or inhibit a disease in the subject, including, but not limited to, administration of a compound or pharmaceutical composition comprising formula I-VI or formulae 1-18 as described herein to a subject in need thereof.
According to the embodiment of the invention, the auxiliary materials comprise medicinal excipients, lubricants, fillers, diluents, disintegrating agents, stabilizers, preservatives, emulsifiers, cosolvents, colorants and sweeteners which are well known in the field of preparation, and are prepared into different dosage forms such as tablets, pills, capsules, injections and the like.
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied, unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be referred to as described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and Jerry March, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"chiral" is a molecule having the property of not overlapping its mirror image; and "achiral" refers to a molecule that can overlap with its mirror image.
"enantiomer" refers to two isomers of a compound that are not overlapping but are in mirror image relationship to each other.
"diastereomer" refers to a stereoisomer having two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow the general definitions of S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E.and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
Many organic compounds exist in an optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of a molecule with respect to one or more of its chiral centers. The prefixes d and l or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or l indicates that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. A particular stereoisomer is an enantiomer and a mixture of such isomers is referred to as an enantiomeric mixture. A50: 50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process.
Any asymmetric atom (e.g., carbon, etc.) of a compound disclosed herein can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Depending on the choice of starting materials and methods, the compounds of the invention may exist as one of the possible isomers or as mixtures thereof, for example as racemates and diastereomeric mixtures (depending on the number of asymmetric carbon atoms). The optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituents may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have cis or trans configuration.
Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
The racemates of any of the resulting end products or intermediates can be resolved into the optical enantiomers by known methods using methods familiar to those skilled in the art, e.g., by separation of the diastereomeric salts obtained. The racemic product can also be separated by chiral chromatography, e.g., High Performance Liquid Chromatography (HPLC) using a chiral adsorbent. In particular, Enantiomers can be prepared by asymmetric synthesis, for example, see Jacques, et al, Enantiomers, racemes and solutions (Wiley Interscience, New York, 1981); principles of Asymmetric Synthesis (2) nd Ed.Robert E.Gawley,Jeffrey Aubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);Wilen,S.H.Tables of Resolving Agents and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of Notre Dame Press,Notre Dame,IN 1972);Chiral Separation Techniques:A Practical Approach(Subramanian,G.Ed.,Wiley-VCH Verlag GmbH&Co.KGaA,Weinheim,Germany,2007)。
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (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 (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers (valenctautomers) include interconversion by recombination of some of the bonding electrons. A specific example of keto-enol tautomerism is the tautomerism of the pentan-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the tautomerism of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention. It is understood that the term "optionally substituted" may be used interchangeably with the term "substituted or unsubstituted". In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a particular substituent. Unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently.
As described herein, the ring system formed by the ring with the substituent R 'bonded to the center represents that the substituent R' may be substituted at any substitutable position on the ring. For example, formula a represents that any possible substituted position on the ring of B 'may be substituted by R', as shown in formula B, formula c and formula d.
Figure BDA0002117302440000271
Attachment points on the ring as described in the present invention
Figure BDA0002117302440000272
The attachment to the rest of the molecule can be at any point on the ring that is attachable. For example, formula i represents any possible attachment position on the B' ring as indicated by formulas d, e and f.
Figure BDA0002117302440000273
Two attachment points on the same ring as described in the present invention
Figure BDA0002117302440000274
The two other parts of the molecule can be attached separately at any two different locations on the ring that can be attached, and the attachment at the two ends can be interchanged. For example, formula j represents that any of the two different positions on the D ring that may be linked can be used as linkers to link the remaining two moieties of the molecule, as shown in formulae g, h and k.
Figure BDA0002117302440000275
As described herein, in some embodiments, when Y is a compound of formula VI-1,
Figure BDA0002117302440000276
Figure BDA0002117302440000277
is composed of
Figure BDA0002117302440000278
Figure BDA0002117302440000279
And k is k1+2, or k ═ k2+1, or k ═ k3+1, or k ═ k4+2。
In addition, k is defined as k 1+2”、“k=k2+1”、“k=k3+1 "or" k ═ k4+2 "is for k1、k2、k3Or k4The value range of (2) is limited. As described above, formula
Figure BDA00021173024400002710
Wherein k is an integer of 0 to 12, and represents t3Is k, and each t3The structural formulas of (A) and (B) may be the same or different. Thus, formula
Figure BDA00021173024400002711
When t denotes3Is composed of
Figure BDA00021173024400002712
When the temperature of the water is higher than the set temperature,
Figure BDA0002117302440000281
the number of the repetitions of (2) is k; formula (II)
Figure BDA0002117302440000282
Where k is k1+2, means when t is3Is composed of
Figure BDA0002117302440000283
Figure BDA0002117302440000284
In the case of the combination of (a) and (b),
Figure BDA0002117302440000285
is k1The number of the main components is one,
Figure BDA0002117302440000286
the number of the repeated units is 2, the total number of the repeated units still keeps k, and the two repeated units have the connection relationship shown in the formula; formula (II)
Figure BDA0002117302440000287
Where k is k2+1, means when t is3Is composed of
Figure BDA0002117302440000288
In the case of the combination of (a) and (b),
Figure BDA0002117302440000289
is k2The number of the main components is one,
Figure BDA00021173024400002810
the number of the repeated units is 1, the total number of the repeated units still keeps k, and the two repeated units have the connection relationship shown in the formula; formula (II)
Figure BDA00021173024400002811
Where k is k3+1, means when t is3Is composed of
Figure BDA00021173024400002812
In the case of the combination of (a) and (b),
Figure BDA00021173024400002813
the number of the repetitions of (a) is 1,
Figure BDA00021173024400002814
is k3The total number of the repeating units still keeps k, and the two repeating units have the connection relation shown in the formula; formula (II)
Figure BDA00021173024400002815
Where k is k4+2, means when t is3Is composed of
Figure BDA00021173024400002816
In the combination of (1), the connection relationship of the formula is that the repeated number is k4Of
Figure BDA00021173024400002817
With 1 connection repetition
Figure BDA00021173024400002818
Reconnection repetition number of 1
Figure BDA00021173024400002819
And the total number of repeating units remains k.
If two points of attachment are available on the chain structure for attachment to the rest of the molecule, the attachment at both ends may be interchanged, as described herein. As described herein, for example, "- (CR)mRw)g-O-C(=O)-(CRmRw)g- (CR)mRw)g-S(=O)p-N(R1a)-(CRmRw)g- "the connection of the two ends may be interchanged.
In addition, unless otherwise explicitly indicated, the descriptions of the terms "… independently" and "… independently" and "… independently" used in the present invention are interchangeable, and should be understood in a broad sense, which means that the specific items expressed between the same symbols do not affect each other in different groups, or that the specific items expressed between the same symbols in the same groups do not affect each other. For example, the formula "- (CR)mRw)g-O-C(=O)-(CRmRw)g- "and the formula" - (CR)mRw)g-S(=O)p-N(R1a)-(CRmRw)g- "R between the twomAnd RwAre not affected by each other even if they are of the same structural formula as "- (CR)mRw)g-O-C(=O)-(CRmRw)g- ", each RmAre also unaffected by each other.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example The term "C1-6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group. As another example, integers between 0 and 12 include any of the endpoints 0 and 12, and integers between 0 and 12, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
Unless otherwise indicated, the term "alkyl" denotes a saturated straight or branched chain monovalent hydrocarbon group of 1 to 20 carbon atoms, or 1 to 10 carbon atoms, or 1 to 8 carbon atoms, or 1 to 6 carbon atoms, or 1 to 4 carbon atoms, or 1 to 3 carbon atoms, wherein the alkyl group may be independently and optionally substituted with one or more substituents described herein, including, but not limited to, deuterium, amino, hydroxyl, cyano, F, Cl, Br, I, mercapto, nitro, oxo (═ O), and the like. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH) 3) Ethyl (Et, -CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl group (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) T-butyl (t-Bu, -C (CH)3)3) N-pentyl (-CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH(CH3)2) 3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) N-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2) 2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2) 3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)3) N-heptyl, n-octyl, and the like. The term "alkyl" and its prefix "alkane" as used herein, both include straight and branched saturated carbon chains. The term "alkylene" is used herein to denote a saturated divalent hydrocarbon radical resulting from the elimination of two hydrogen atoms from a straight or branched chain saturated hydrocarbon, examples of which include, but are not limited to, methylene, ethylidene, and the like.
The term "C1-6The "alkyl" in alkyl-C (═ O) -NH- "is as defined herein, and such examples include, but are not limited to, CH 3-C(=O)-NH-、CH3CH2-C(=O)-NH-、CH3CH2CH2-C(=O)-NH-、(CH3)3C-C (═ O) -NH-, and the like.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 15 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described hereinIt includes the positioning of "cis" and "tans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 8 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 15 carbon atoms, wherein there is at least one site of unsaturation, i.e. a carbon-carbon sp triple bond, wherein said alkynyl radical may optionally be substituted with one or more substituents as described herein. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH) 2C.ident.CH), 1-propynyl (-C.ident.C-CH)3) And so on.
The terms "cycloalkyl", "cycloaliphatic", "carbocyclic", or "carbocyclyl" refer to a mono-or polyvalent, non-aromatic, saturated or partially unsaturated ring, and contain no heteroatoms, including monocyclic rings of 3 to 12 carbon atoms or bicyclic rings of 7 to 12 carbon atoms. The bicyclic carbocyclic ring having 7 to 12 atoms may be bicyclo [4,5 ]]、[5,5]、[5,6]Or [6,6 ]]The bicyclic carbocyclic ring having 9 or 10 atoms may be bicyclo [5,6 ]]Or [6,6 ]]And (4) preparing the system. Suitable cyclic aliphatic groups include, but are not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl. Examples of cycloaliphatic radicals include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. And said "cycloaliphatic" or "carbocycle", "carbocyclyl", "cycloalkyl" may be substituted or unsubstituted, wherein The substituent may be, but is not limited to, deuterium, hydroxyl, amino, halogen, cyano, aryl, heteroaryl, alkoxy, alkylamino, alkyl, alkenyl, alkynyl, heterocyclic, mercapto, nitro, aryloxy, hydroxyl-substituted alkoxy, hydroxyl-substituted alkyl-C (═ O) -, alkyl-S (═ O) —, alkyl-S (═ O)2-, hydroxy-substituted alkyl-S (═ O)2-, carboxy-substituted alkoxy, and the like.
The terms "heterocycle", "heterocyclyl", "heteroalicyclic" or "heterocyclic" are used interchangeably herein and all refer to a monocyclic, bicyclic or tricyclic ring system in which one or more carbon atoms in the ring are independently and optionally substituted with a heteroatom having the meaning as set forth herein, the ring may be fully saturated or contain one or more degrees of unsaturation, but is by no means aromatic, and has one or more points of attachment to the rest of the molecule. One or more of the ring hydrogen atoms are independently and optionally substituted with one or more substituents as described herein. Some of these embodiments are "heterocycle", "heterocyclyl", "heteroalicyclic" or "heterocyclic" groups are monocyclic (1-6 carbon atoms and 1-3 heteroatoms selected from N, O, P, S) having 3-7 members rings, where S or P is optionally substituted with one or more oxygen atoms to yield, for example, SO 2、PO、PO2When the ring is a three-membered ring, in which there is only one heteroatom), or a 7-to 10-membered bicyclic ring (4-9 carbon atoms and 1-3 heteroatoms selected from N, O, P, S, where S or P is optionally substituted with one or more oxygen atoms to give, for example, SO2、PO、PO2The group of (1).
The heterocyclic group may be a carbon-based or heteroatom group. "Heterocyclyl" also includes heterocyclic groups fused to saturated or partially unsaturated rings or heterocycles. Examples of heterocycles include, but are not limited to, pyrrolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, thiaxalkyl, thiazolidinyl, oxazolidinyl, piperazinyl, homopiperazinyl, azapineCyclobutyl, oxetanyl, thietanyl, homopiperidinyl, epoxypropyl, azepinyl, oxepanyl, thiepinyl, 4-methoxy-piperidin-1-yl, 1,2,3, 6-tetrahydropyridin-1-yl, oxazepinyl
Figure BDA0002117302440000301
Radical diaza
Figure BDA0002117302440000302
Radical, sulfur nitrogen hetero
Figure BDA0002117302440000303
A group, pyrrolin-1-yl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxacyclohexyl, 1, 3-dioxolanyl, pyrazolinyl, dithianyl, dithienoalkyl, dihydrothienyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1,2, 6-thiadiazinane 1, 1-dioxo-2-yl, 4-hydroxy-1, 4-azaphosphane 4-oxide-1-yl, 2-hydroxy-1- (piperazin-1-yl) ethanone-4-yl, 2-hydroxy-1- (5, 6-dihydro-1, 2, 4-triazin-1 (4H) -yl) ethanon-4-yl, 5, 6-dihydro-4H-1, 2, 4-oxadiazin-4-yl, 2-hydroxy-1- (5, 6-dihydropyridin-1 (2H) -yl) ethanon-4-yl, 3-azabicyclo [3.1.0 ]Hexyl, 3-azabicyclo [4.1.0]Heptyl, azabicyclo [2.2.2 ] s]Hexyl, 2-methyl-5, 6,7, 8-tetrahydro- [1.2.4]Triazole [1,5-c ]]Pyrimidin-6-yl, 4,5,6, 7-tetrahydroisoxazole [4,3-c ] as intermediates]Pyridin-5-yl, 3H-indolyl 2-oxo-5-azabicyclo [2.2.1]Heptane-5-yl, 2-oxo-5-azabicyclo [2.2.2 ]]Octane-5-yl, quinolizinyl and N-pyridyl urea. Examples of heterocyclic groups also include 1, 1-dioxothiomorpholinyl and wherein two carbon atoms of the ring are replaced by oxygen atoms such as pyrimidinedione. And the heterocyclic group may be substituted or unsubstituted, wherein the substituent may be, but is not limited to, deuterium, oxo (═ O), hydroxyl, amino, halogen, cyano, heteroaryl, alkoxy, alkylamino, alkyl, alkenyl, alkynyl, heterocyclic group, mercapto, nitro, aryloxy, hydroxyl-substituted alkoxy, hydroxyl-substituted alkyl-C (═ O) -, alkyl-S (═ O)2-, hydroxyl-substituted alkyl-S (═ O) -, hydroxyl-substituted alkyl-C (═ O) -, hydroxyl-substituted alkyl-S (═ O) -, hydroxyl-substituted alkyl-ralkyl-S (═ O)2-, carboxy-substituted alkoxy, and the like.
The term "heteroatom" means one or more of O, S, N, P and Si atoms, including N, S and any oxidation state form of P; primary, secondary, tertiary amines and quaternary ammonium salt forms; or a form in which a hydrogen on a nitrogen atom in the heterocycle is substituted, e.g., N (e.g., N in 3, 4-dihydro-2H-pyrrolyl), NH (e.g., NH in pyrrolidinyl), or NR (e.g., NR in N-substituted pyrrolidinyl).
The term "aryl" may be used alone or as a majority of an "aralkyl", "aralkoxy", or "aryloxyalkyl" group, and refers to monocyclic, bicyclic, and tricyclic carbon ring systems containing a total of 6 to 14 ring members, wherein at least one of the ring systems is aromatic, wherein each ring system contains 3 to 7 ring members with one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring", e.g., aromatic rings may include phenyl, naphthyl and anthracenyl. And the aryl group may be substituted or unsubstituted, wherein the substituent may be, but is not limited to, deuterium, hydroxyl, amino, halogen, cyano, aryl, heteroaryl, alkoxy, alkylamino, alkyl, alkenyl, alkynyl, heterocyclic, mercapto, nitro, aryloxy, hydroxyl-substituted alkoxy, hydroxyl-substituted alkyl-C (═ O) -, alkyl-S (═ O) —, alkyl-S (═ O)2-, hydroxy-substituted alkyl-S (═ O)2-, carboxy-substituted alkoxy, and the like.
The term "heteroaryl" may be used alone or as a majority of "heteroarylalkyl" or "heteroarylalkoxy" and denotes monocyclic, bicyclic and tricyclic ring systems containing a total of 5-14 membered rings, wherein at least one ring system is aromatic and at least one ring system contains one or more heteroatoms, wherein the heteroatoms have the meaning described herein, and wherein each ring system contains 3-7 membered rings and one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic" or "heteroaromatic". And the heteroaryl group may be substituted or unsubstituted, wherein a substituent may And are, but are not limited to, deuterium, hydroxy, amino, halogen, cyano, aryl, heteroaryl, alkoxy, alkylamino, alkyl, alkenyl, alkynyl, heterocyclyl, mercapto, nitro, aryloxy, hydroxy-substituted alkoxy, hydroxy-substituted alkyl-C (═ O) -, alkyl-S (═ O)2-, hydroxy-substituted alkyl-S (═ O)2-, carboxy-substituted alkoxy, and the like.
In other embodiments, the aromatic heterocycle includes, but is not limited to, the following monocyclic rings: 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 4-methylisoxazol-5-yl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, pyrimidin-5-yl, pyridazinyl (e.g. 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g. 5-tetrazolyl), triazolyl (e.g. 2-triazolyl and 5-triazolyl), and the like, 2-thienyl, 3-thienyl, pyrazolyl (e.g. 2-pyrazolyl), isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, 1,3, 4-thiadiazol-2-yl, pyrazinyl, pyrazin-2-yl, 1,3, 5-triazinyl, benzo [ d ] thiazol-2-yl, imidazo [1,5-a ] pyridin-6-yl; the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), and isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl).
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula I, formula II, formula III, formula IV, formula V, or formula VI. Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, in the prior invention, the ester can be used as the phenyl ester of the prodrug,aliphatic radical (C)1-24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel Delivery Systems, Vol.14of the A.C.S.Symphosis Series, Edward B.Roche, ed., Bioreversible Carriers in Drug designs, American Pharmaceutical Association and Pergamon Press,1987, J.Rautio et al, Prodrugs in Design and Clinical Applications, Nature Review Delivery, 2008,7,255 and 270, S.J.Herer et al, Prodrugs of pharmaceuticals and pharmaceuticals, Journal of chemical Chemistry,2008,51,2328 and 5.
"metabolite" refers to the product of a particular compound or salt thereof that is metabolized in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assays as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting the compounds of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, description of the scientific acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, inorganic acid salts formed by reaction with amino groups such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, and organic acid salts such as acetate, oxalate, maleate, tartrate Citrate, succinate, malonate, or by other methods described in the literature such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical) 4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
The term "treating" or "treatment" as used herein refers, in some embodiments, to ameliorating a disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
Pharmaceutically acceptable acid addition salts may be formed with inorganic and organic acids, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheophylline, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, and the like, Polysilonolactates, propionates, stearates, succinates, sulfosalicylates, tartrates, tosylates and trifluoroacetates.
Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, sulfosalicylic acid, and the like.
Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of groups I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (cholinate), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of Pharmaceutical Salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable Salts.
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms.
Any formulae given herein are also intended to represent the non-isotopically enriched forms of these compounds in order toAnd isotopically enriched forms. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H,3H,11C,13C,14C,15N,17O,18O,18F,31P,32P,35S,36Cl and125I。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present3H,14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13C. the isotopically enriched compounds can be used for metabolic studies (use) 14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula I, formula II, formula III, formula IV, formula V or formula VI can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stability. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. If a substituent of a compound of the invention is designated as deuterium, the compound has at least 3500 for each designated deuterium atom (at each designated deuterium atom) Deuterium incorporation of 52.5%), at least 4000 (60%), at least 4500 (67.5%), at least 5000 (75%), at least 5500 (82.5%), at least 6000 (90%), at least 6333.3 (95%), at least 6466.7 (97%), at least 6600 (99%) or at least 6633.3 (99.5%) deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D2O, acetone-d6、DMSO-d6Those solvates of (a).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention, a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle, or combination thereof. In some embodiments, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
The following examples of the present invention are described in further detail, and are intended to be illustrative, but not limiting, of the present invention.
The experimental procedures used in the following examples are all conventional procedures unless otherwise specified.
Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
The compounds of the present invention are prepared by a click reaction or an amide condensation reaction, as shown in figure 2.
Preparation of intermediate
Figure BDA0002117302440000351
To a 10mL round-bottomed flask, 120mg of starting material 1 was added and 3mL of N, N-dimethylformamide was added to dissolve the starting material, and then 70mg of 2, 4-difluorophenol and 375mg of K were added in this order2CO3. The reaction was kept stirring at 110 degrees celsius for about two hours until TLC monitored the disappearance of starting material 1. The reaction solution was returned to room temperature. The reaction solution was added to a large amount of water and extracted with ethyl acetate several timesThe organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was spin-dried. To the residue was added 10mL methanol/water-3: 1 and diluted hydrochloric acid activated and dried iron powder 100mg and ammonium chloride solid 50mg, and the mixture was stirred at 80 ℃ under reflux for 1.5 hours. Cooling to room temperature and filtering with diatomaceous earth to remove the remaining iron powder, adding appropriate amount of water to the filtrate and extracting with ethyl acetate for 3 times, 20mL each time, combining the organic phases and drying with anhydrous sodium sulfate, removing the solvent by rotary evaporation, and separating and purifying the residue with a 200-mesh and 300-mesh silica gel chromatographic column, wherein the mobile phase is dichloromethane: methanol 20: 1, 90mg of intermediate 2 are obtained, with a yield of 90%.
To a 10mL round bottom flask were added 33mg of 5-hexynoic acid, 120mg of 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethyluronium Hexafluorophosphate (HATU), 2mL of dichloromethane, 2mL of N, N-dimethylformamide, and 100. mu.L of triethylamine in this order, and the mixture was stirred at room temperature for 5 minutes, 90mg of intermediate 2 was added to the reaction solution, and reacted at room temperature until intermediate 2 was substantially disappeared. The reaction was added to 200mL of water and extracted with ethyl acetate (3 times 20mL each), the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was isolated and purified by 200-300 mesh silica gel chromatography with the mobile phase dichloromethane: methanol 30: 1, 95mg of intermediate 3 are obtained, with a yield of 84%.
Figure BDA0002117302440000361
To a 10mL round bottom flask was added 88mg of starting material 1 and 2mL of N, N-dimethylformamide to dissolve, followed by 88mg of 3- (propane-2-yn-1-oxy) phenol (3- (prop-2-yn-1-yloxy) phenol) and 275mg of potassium carbonate. The reaction was kept stirring at 110 ℃ for about two hours until disappearance of starting material 1 was monitored by TLC. The reaction solution was returned to room temperature. The reaction solution was added to a large amount of water and extracted several times with ethyl acetate, and the organic phases were combined and dried using anhydrous sodium sulfate, and the solvent was spin-dried. To the residue was added 10mL methanol/water-3: 1 and diluted hydrochloric acid, 100mg of iron powder and 50mg of ammonium chloride solid which are activated and dried, and the reaction solution is refluxed and stirred for 1.5 hours at the temperature of 80 ℃. Cooling to room temperature and filtering with diatomaceous earth to remove the remaining iron powder, adding appropriate amount of water to the filtrate and extracting with ethyl acetate 3 times, 20mL each time, combining the organic phases and drying with anhydrous sodium sulfate, rotary evaporating to remove the solvent, separating and purifying the residue with a 200-mesh and 300-mesh silica gel chromatographic column, the mobile phase is dichloromethane: methanol 20: 1, 56mg of intermediate 4 are obtained, with a yield of 73%.
50mg of intermediate 4 was placed in a 10mL round-bottom flask, 10mL of methylene chloride was added to dissolve it, the reaction flask was placed in an ice-water bath, and 330mg of ethylsulfonyl chloride and 360. mu.L of triethylamine were sequentially added to the reaction solution and the reaction was stirred at this temperature until the starting material substantially disappeared. The reaction was added to 150mL of water and extracted with dichloromethane (3 times 20mL each), the organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. The residue was transferred to a 10mL round-bottomed flask, and then 22mg of potassium fluoride, 90mg of potassium carbonate and 43mg of 1, 3-diol were added to the reaction solution in this order, and the reaction solution was heated and stirred at 110 ℃ for 2 hours. Cooled to room temperature, the reaction mixture was added to 150mL of water and extracted with ethyl acetate (3 times 20mL), the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was isolated and purified by 200-mesh 300-mesh silica gel chromatography using dichloromethane as the mobile phase: methanol 30: 1, 45mg of intermediate 5 are obtained, with a yield of 73%.
1H-NMR(400MHz,CDCl3)δ(ppm)10.66(s,1H),7.50(s,1H),7.29(m,1H),7.15-7.05(m,3H),6.60(d,J=8.60Hz,1H),6.48-6.41(m,3H),4.57(s,2H),3.56(s,3H),3.19(q,J=6.96Hz,2H),1.42(t,J=7.20Hz,3H);
Figure BDA0002117302440000371
A10 mL round bottom flask was charged with 66mg of starting material 1 and 2mL of N, N-dimethylformamide to dissolve, followed by the sequential addition of 90mg of N- (3-hydroxyphenyl) -5-yne-hexanamide and 206mg of potassium carbonate. The reaction was kept stirring at 110 ℃ for about two hours until disappearance of starting material 1 was monitored by TLC. The reaction solution was returned to room temperature. The reaction solution was added to a large amount of water and extracted several times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. To the residue was added 10mL methanol/water-3: 1 and diluted hydrochloric acid, 100mg of iron powder and 50mg of ammonium chloride solid which are activated and dried, and the reaction solution is refluxed and stirred for 1.5 hours at the temperature of 80 ℃. Cooling to room temperature and filtering with diatomaceous earth to remove the remaining iron powder, adding appropriate amount of water to the filtrate and extracting with ethyl acetate 3 times, 20mL each time, combining the organic phases and drying with anhydrous sodium sulfate, rotary evaporating to remove the solvent, separating and purifying the residue with a 200-mesh and 300-mesh silica gel chromatographic column, the mobile phase is dichloromethane: methanol 20: 1 to yield 52mg of intermediate 6 in 79% yield.
50mg of intermediate 6 was placed in a 10mL round-bottom flask, 10mL of methylene chloride was added to dissolve it, the reaction flask was placed in an ice-water bath, and 290mg of ethylsulfonyl chloride and 320. mu.L of triethylamine were sequentially added to the reaction solution and the reaction was stirred at this temperature until the starting material substantially disappeared. The reaction was added to 150mL of water and extracted with dichloromethane (3 times 20mL each), the organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. The residue was transferred to a 10mL round-bottomed flask, and then 20mg of potassium fluoride, 78mg of potassium carbonate and 38mg of 1, 3-diol were added to the reaction solution in this order, and the reaction solution was heated and stirred at 110 ℃ for 2 hours. Cooled to room temperature, the reaction mixture was added to 100mL of water and extracted with ethyl acetate (3 times 20mL), the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was isolated and purified by 200-mesh 300-mesh silica gel chromatography using dichloromethane as the mobile phase: methanol 30: 1, 40mg of intermediate 7 are obtained, with a yield of 66%.
Figure BDA0002117302440000381
To a 50mL round-bottomed flask was added 300mg of starting material 1 and 10mL of N, N-dimethylformamide was added to dissolve it, and then 300mg of 4-aminophenol and 280mg of K were added in this order2CO3. The reaction was stirred at 110 ℃ for two hours until disappearance of starting material 1 was monitored by TLC. The reaction solution was returned to room temperature. The reaction solution was added to a large amount of water and extracted with ethyl acetateThe organic phases were combined several times and dried over anhydrous sodium sulfate, the solvent was spin-dried. The residue was separated and purified by 200-mesh 300-mesh silica gel chromatography column, and the mobile phase was dichloromethane: methanol 20: 1, 170mg of intermediate 8 are obtained, with a yield of 67%.
To a 50mL round bottom flask was added 300mg of starting material 1 and 10mL of N, N-dimethylformamide to dissolve it, then 300mg of 4-aminophenol and 280mg of K were added in that order2CO3. The reaction was stirred at 110 ℃ for two hours until TLC monitored the disappearance of starting material 1. The reaction solution was returned to room temperature. The reaction solution was added to a large amount of water and extracted several times with ethyl acetate, the organic phases were combined and dried using anhydrous sodium sulfate, and the solvent was spin-dried. The residue was separated and purified by 200-mesh 300-mesh silica gel chromatography column, and the mobile phase was dichloromethane: methanol 20: 1 to yield 190mg of intermediate 9 in 74% yield.
Preparation of bis, CRBN terminal ligand
Figure BDA0002117302440000382
To a round bottom flask was added 2- (2, 6-diketopiperidin-3-yl) -4-fluorobenzoxazolinyl-1, 3-dione, 2.0 equivalents of DIEA, an appropriate amount of N, N-dimethylformamide as solvent and 2.0 equivalents of an amine starting material terminated with an azide functional group or an amine starting material terminated with a carboxylic acid. The reaction solution was stirred at 80 ℃ for 3 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was separated and purified by 200-mesh 300-mesh silica gel chromatography with ethyl acetate as mobile phase: petroleum ether is 1: 1 to 3: 1, the yield is 20-40%.
Figure BDA0002117302440000391
CRBN ligand used for negative control of formula 2 preparation 2- (2, 6-diketopiperidin-3-yl) -4-fluorobenzoxazolin-1, 3-dione, 2.0 equivalents of potassium carbonate, appropriate amount of N, N-dimethylformamide as solvent and 2.0 equivalents of ethyl iodide were added to a round bottom flask and the reaction was stirred at 80 ℃ for 2 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was separated and purified by 200-mesh 300-mesh silica gel chromatography with ethyl acetate as mobile phase: petroleum ether is 3: 1, yield 90%.
To a round bottom flask was added 2- (1-ethyl-2, 6-diketopiperidin-3-yl) -4-fluorobenzoxazolin-1, 3-dione, 2.0 equivalents of DIEA, the appropriate amount of N, N-dimethylformamide as solvent and 2.0 equivalents of the azido-terminated amine starting material. The reaction solution was stirred at 80 ℃ for 3 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was separated and purified by 200-mesh 300-mesh silica gel chromatography with ethyl acetate as mobile phase: petroleum ether is 1: 1 to 3: 1, the yield is 20-40%.
Figure BDA0002117302440000392
A round-bottom flask was charged with raw material A, 5.0 equivalents of terminal alkyne raw material, 10% equivalents of 1,1' -bisdiphenylphosphinoferrocene palladium dichloride, 20% equivalents of cuprous iodide and a suitable volume of N, N-dimethylformamide as solvent, the flask was sealed with a rubber stopper and the air in the flask was replaced with argon, and triethylamine in a volume of 1/3N, N-dimethylformamide was added to the reaction flask with a syringe. The reaction solution was stirred at 70 ℃ for 6 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was isolated and purified by 200-mesh 300-mesh silica gel chromatography with a mobile phase of dichloromethane: methanol 30: 1 to 10: 1, yield about 50%.
And adding the intermediate in the previous step, 3-5 equivalents of sodium azide and a proper amount of N, N-dimethylformamide into a round-bottom flask, and stirring the reaction solution at 80 ℃ for 6 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was isolated and purified by 200-mesh 300-mesh silica gel chromatography with a mobile phase of dichloromethane: methanol 30: 1 to 10: 1, yield 100%.
Figure BDA0002117302440000401
To a round bottom flask was added 300mg of 2- (2, 6-diketopiperidin-3-yl) -4-fluorobenzoxazolin-1, 3-dione, 2.0 equivalents of DIEA, the appropriate amount of dimethyl sulfoxide as solvent and 2.0 equivalents of Boc-L-lysine. The reaction solution was stirred at 80 ℃ for 3 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was separated and purified by 200-mesh 300-mesh silica gel chromatography with ethyl acetate as mobile phase: petroleum ether is 1: 1 to 3: 1, 244mg of intermediate was obtained with a yield of 44%. 35mg of this intermediate was taken in a 10ml round bottom flask, 5ml of dichloromethane and 200. mu.l of trifluoroacetic acid were added, stirring was carried out at room temperature for 2 hours, the solvent was dried by spinning, 3ml of N, N-dimethylformamide was added to the residue to dissolve it, and 100. mu.l of acetic anhydride was added. Stirred at room temperature for 2 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was isolated and purified by 200-mesh 300-mesh silica gel chromatography with a mobile phase of dichloromethane: methanol 20: 1, 24mg of product is obtained, with a yield of 77%.
Figure BDA0002117302440000402
To a round bottom flask was added 2- (2, 6-diketopiperidin-3-yl) -3-fluorobenzoxazolinyl-1, 3-dione, 2.0 equivalents of DIEA, an appropriate amount of N, N-dimethylformamide as solvent and 2.0 equivalents of an amine starting material terminated with an azide functional group or an amine starting material terminated with a carboxylic acid. The reaction solution was stirred at 80 ℃ for 3 hours. The reaction was quenched by addition of saturated brine, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was separated and purified by 200-mesh 300-mesh silica gel chromatography with ethyl acetate as mobile phase: petroleum ether is 1: 1 to 3: 1, the yield is 20-40%.
Preparation of the Compound of example 1
Preparation of compounds represented by formulas 1 to 17
Synthesis of a compound of formula 1:
Figure BDA0002117302440000411
taking 15mg of intermediate 3 and 14mg of CRBN end ligand, putting the intermediate 3 and the CRBN end ligand into a 5mL round-bottom flask, adding 50mg of sodium ascorbate, and then adding N, N-dimethylformamide/water to obtain a mixture of 5: 1, 2mL of the mixed solvent, and finally 2mg of anhydrous copper sulfate was added to the mixture to carry out a reaction at room temperature for 6 hours. Saturated brine was added to the reaction solution, the mixture was extracted three to five times with ethyl acetate, the organic phases were combined and dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation, and the residue was separated and purified by a 200-mesh 300-mesh silica gel chromatography column, the mobile phase was dichloromethane: methanol 30: 1 to 10: 1, 13mg of the title compound are obtained in a yield of 56%.
1H-NMR(400MHz,CDCl3)δ(ppm):10.15(s,1H),9.47(s,1H),8.94(s,1H),7.64(m,2H),7.47(t,J=7.60Hz,1H),7.34(s,1H),7.21(s,1H),7.12(s,1H),7.07(d,J=7.20Hz,1H),6.87-6.78(m,4H),6.69-6.65(m,1H),6.43(s,1H),6.19(s,1H),4.95-4.91(m,1H),4.27(t,J=7.20Hz,2H),3.63(s,3H),3.26-3.21(m,2H),2.89-2.73(m,5H),2.47-2.44(m,2H),2.13-2.07(m,3H),1.85(m,2H),1.76(m,5H),1.64-1.61(m,2H),1.38-1.25(m,8H);
LC-MS(ESI+):m/z calculated for C47H48F2N9O7(M+H)+:888.36found 888.92.
According to the preparation method, the compounds shown in the formulas 2-15 and 17 can be prepared from the intermediate 3/5/7 and the CRBN terminal ligand.
Figure BDA0002117302440000421
1H-NMR(400 MHz,CDCl3)δ(ppm)10.50(s,1H),9.48(s,1H),8.96(s,1H),7.64(s,1H),7.59(t,J=8.80 Hz,1H),7.54(s,1H),7.43(t,J=8.00 Hz,1H),7.19(s,1H),7.11(s,1H),7.05(d,J=7.20 Hz,1H),6.86-6.77(m,4H),6.66(t,J=8.40 Hz,1H),6.46(t,J=5.20 Hz,1H),6.41(s,1H),4.95-4.91(m,1H),4.43(t,J=4.80 Hz,2H),3.80(t,J=4.80 Hz,2H),3.67(t,J=4.20Hz,2H),3.62-3.55(m,12H),3.43-3.39(m,2H),2.86-2.72(m,5H),2.44-2.41(m,2H),2.09-2.07(m,2H);
LC-MS(ESI+):m/z calculated for C47H48F2N9O10(M+H)+:936.34 found 936.52.
Figure BDA0002117302440000422
1H-NMR(400 MHz,CDCl3)δ(ppm)10.72(s,1H),9.70(s,1H),9.03(s,1H),7.70(d,J=7.24 Hz,1H),7.67(s,1H),7.60(d,J=8.80 Hz,1H),7.52(s,1H),7.41-7.33(m,2H),7.19(s,1H),7.11(s,1H),6.84-6.77(m,3H),6.66(t,J=7.88 Hz,1H),6.40(s,1H),5.24(dd,J=13.08Hz,J=4.92 Hz,1H),4.43(d,J=7.40 Hz,2H),4.42(d,J=13.36 Hz,1H),4.27(d,J=16.12 Hz,1H),3.80(t,J=4.72 Hz,2H),3.64(s,3H),3.56-3.52(m,8H),3.43-3.38(m,2H),2.86-2.78(m,3H),2.67(t,J=7.32 Hz,2H),2.43(s,2H),2.38-2.28(m,1H),2.05-2.00(m,4H),1.91-1.84(m,2H);
LC-MS(ESI+):m/z calculated for C48H51F2N8O9(M+H)+:921.37 found 921.60.
Figure BDA0002117302440000431
1H-NMR(400 MHz,CDCl3)δ(ppm)10.68(s,1H),9.55(s,1H),8.97(s,1H),7.72(d,J=7.08Hz,1H),7.67(s,1H),7.60(d,J=8.88Hz,1H),7.44-7.37(m,2H),7.33(s,1H),7.23(s,1H),7.14(s,1H),6.85-6.78(m,3H),6.68(t,J=7.72Hz,1H),6.44(s,1H),5.30-5.25(m,1H),4.46(d,J=16.04Hz,1H),4.31-4.23(m,3H),3.64(s,3H),3.39-3.34(m,6H),2.87-2.80(m,3H),2.71(t,J=7.40Hz,2H),2.44(t,J=6.72Hz,2H),2.39-2.32(m,4H),2.08-1.98(m,7H),1.93-1.86(m,2H),1.65-1.59(m,2H),1.53-1.50(m,2H);
LC-MS(ESI+):m/z calculated for C50H55F2N8O7(M+H)+:917.41 found 917.99.
Figure BDA0002117302440000432
1H-NMR(400 MHz,CDCl3)δ(ppm)11.09(s,1H),8.87(s,1H),7.70(s,1H),7.46-7.42(m,2H),7.35(m,1H),7.20(dd,J=8.72 Hz,J=2.68Hz,1H),7.13-7.02(m,3H),6.85(d,J=8.52Hz,1H),6.61(dd,J=8.28 Hz,J=1.88 Hz,1H),6.44(m,2H),6.39(s,1H),5.06(s,2H),4.91(dd,J=11.72 Hz,J=5.20 Hz,1H),4.52(t,J=4.84 Hz,2H),3.85(t,J=4.84 Hz,2H),3.66(m,5H),3.61(s,4H),3.58(s,4H),3.40(t,J=5.00 Hz,2H),3.18(q,J=7.40 Hz,2H),2.88-2.70(m,3H),2.13-2.09(m,1H),1.41(t,J=7.32 Hz,3H);
LC-MS(ESI+):m/z calculated for C46H50N9O12S(M+H)+:952.32 found 952.67.
Figure BDA0002117302440000433
1H-NMR(400 MHz,CDCl3)δ(ppm)11.43(s,1H),8.54(s,1H),7.73(s,1H),7.45-7.41(m,3H),7.20-7.17(m,2H),7.12(t,J=8.24 Hz,1H),7.06(d,J=7.04 Hz,1H),7.01(d,J=8.72 Hz,1H),6.87(s,1H),6.83(d,J=8.52 Hz,1H),6.60(d,J=8.08 Hz,1H),6.48-6.45(m,2H),6.37(s,1H),5.06(s,2H),4.89(dd,J=11.36 Hz,J=5.00 Hz,1H),4.55(t,J=4.84 Hz,2H),3.89(t,J=4.92 Hz,2H),3.70(s,3H),3.67(t,J=5.00 Hz,2H),3.62(s,4H),3.41(t,J=5.08 Hz,2H),3.18(q,J=7.32 Hz,2H),2.90-2.70(m,3H),2.05-2.00(m,1H),1.42(t,J=7.36 Hz,3H);
LC-MS(ESI+):m/z calculated for C44H46N9O11S(M+H)+:908.30 found 908.59.
Figure BDA0002117302440000441
1H-NMR(400 MHz,CDCl3)δ(ppm)10.28(s,1H),9.74(s,1H),8.77(s,1H),.55-7.40(m,3H),7.24-7.21(m,1H),7.15-7.02(m,6H),6.86(s,1H),6.60(t,J=6.68 Hz,2H),6.34(s,1H),4.93-4.88(m,1H),4.39(t,J=5.12 Hz,2H),3.83-3.81(m,2H),3.70-3.67(m,2H),3.61-3.36(m,6H),3.49(s,3H),3.43-3.40(m,2H),3.25(q,J=7.20 Hz,2H),2.71-2.66(m,2H),2.35-2.28(m,2H),1.99-1.94(m,2H),1.44(t,J=7.36 Hz,3H);
LC-MS(ESI+):m/z calculated for C47H52N20O11S(M+H)+:963.34 found 963.78.
Figure BDA0002117302440000442
1H-NMR(400 MHz,CDCl3)δ(ppm)10.45(s,1H),9.50(s,1H),7.65(s,1H),7.54(s,1H),7.49-7.45(m,2H),7.27-7.24(m,1H),7.21(s,1H),7.11-7.05(m,3H),7.03(m,1H),6.85(d,J=8.60 Hz,1H),6.61-6.59(m,1H),6.45-6.42(m,2H),6.38(s,1H),6.20(t,J=5.52 Hz,1H),5.06(s,2H),4.95-4.91(m,1H),4.32(t,J=7.08 Hz,2H),3.56(s,3H),3.25-3.09(m,5H),2.14-2.11(m,1H),1.89-1.85(m,2H),1.65-1.59(m,2H),1.42-1.26(m,10H);
LC-MS(ESI+):m/z calculated for C46H50N9O9S(M+H)+:904.34 found 904.88.
Figure BDA0002117302440000451
1H-NMR(400 MHz,CDCl3)δ(ppm)10.28(s,1H),9.25(s,1H),7.54(s,1H),7.49-7.45(m,2H),7.27(s,1H),7.25-7.22(m,2H),7.13-7.04(m,4H),6.87(d,J=8.52 Hz,1H),6.63-6.60(m,1H),6.46-6.44(m,2H),6.40-6.39(m,1H),6.21(m,1H),5.06(s,2H),4.93-4.90(m,1H),4.33(t,J=7.20 Hz,1H),3.58(s,3H),3.27-3.16(m,4H),2.86-2.76(m,3H),2.12-2.11(m,1H),1.89-1.86(m,2H),1.66-1.63(m,2H),1.42(t,J=7.36 Hz,3H),1.32-1.25(m,14H);
LC-MS(ESI+):m/z calculated for C49H56N9O9S(M+H)+:946.38 found 947.35.
Figure BDA0002117302440000452
1H-NMR(400 MHz,CDCl3)δ(ppm)10.54(s,1H),9.58(s,1H),7.77(d,J=6.84 Hz,1H),7.67-7.63(m,3H),7.55(s,1H),7.43(d,J=2.52 Hz,1H),7.24(d,J=2.76 Hz,1H),7.21(s,1H),7.07-7.04(m,2H),6.98(s,1H),6.56(dd,J=8.0 Hz,J=1.80 Hz,1H),6.39(dd,J=8.24 Hz,J=1.92Hz,1H),6.33(m,2H),5.02(s,2 H),4.99(m,1H),4.37(t,J=7.24 Hz,2 H),3.54(s,3H),3.17(dd,J=14.76 Hz,J=7.36 Hz,2H),2.90-2.74(m,3H),2.50(t,J=6.56 Hz,2H),2.14-2.11(m,1H),1.99-1.92(m,2H),1.69-1.58(m,4H),(t,J=7.32 Hz,3H);
LC-MS(ESI+):m/z calculated for C45H43N8O9S(M+H)+:871.28 found 871.63.
Figure BDA0002117302440000453
Figure BDA0002117302440000461
1H-NMR(400 MHz,CDCl3)δ(ppm)10.33(s,1H),9.35(s,1H),8.98(s,1H),7.66-7.61(m,2H),7.50-7.46(m,1H),7.33(s,1H),7.21(t,J=2.68 Hz,1H),7.12(s,1H),7.08(d,J=7.04 Hz,1H),6.87(s,1H),6.85(s,1H),6.84-6.78(m,2H),6.70-6.66(m,1H),6.44(t,J=2.36Hz,1H),6.21(t,J=5.40Hz,1H),4.95-4.90(m,1H),4.28(t,J=7.20Hz,2H),3.64(s,3H),3.24(dd,J=12.52 Hz,J=6.60 Hz,2H),2.90-2.70(m,5H),2.45(t,J=6.76Hz,2H),2.15-2.05(m,3H),1.85(m,2H),1.63(m,2H),1.38-1.25(m,14H);
LC-MS(ESI+):m/z calculated for C50H54F2N9O7(M+H)+:930.40 found 931.96.
Figure BDA0002117302440000462
1H-NMR(400 MHz,CDCl3)δ(ppm)10.25(s,1H),9.66(s,1H),9.04(s,1H),7.79(dd,J=6.32 Hz,J=2.04 Hz,1H),7.71-7.66(m,3H),7.60(s,1H),7.39(s,1H),7.23(s,1H),7.15(s,1H),6.90-6.81(m,3H),6.70(m,1H),6.44(s,1H),5.03(dd,J=12.04 Hz,J=5.36 Hz,1H),4.36-4.31(m,2H),3.65(s,3H),2.92-2.76(m,5H),2.53(t,J=6.64 Hz,2H),2.47(t,J=5.96 Hz,2H),2.17-2.07(m,3H),1.96(m,2H),1.69(m,2H),1.60(m,2H);
LC-MS(ESI+):m/z calculated for C46H41F2N8O7(M+H)+:855.30 found 855.86.
Figure BDA0002117302440000463
1H-NMR(400 MHz,CDCl3)δ(ppm)10.45(s,1H),9.85(s,1H),9.04(s,1H),7.78(d,J=7.36 Hz,1H),7.69(dd,J=8.76 Hz,J=2.36 Hz,1H),7.58(d,J=2.32 Hz,1H),7.53(d,J=7.48 Hz,1H),7.41(t,J=7.64 Hz,1H),7.32(s,1H),7.20(t,J=2.56 Hz,3H),7.12(s,1H),6.87-6.79(m,1H),6.68(t,J=8.80 Hz,1H),6.41(s,1H),5.29(dd,J=13.32 Hz,J=5.12 Hz,1H),4.51(d,J=12.84 Hz,1H),4.32(d,J=17.00 Hz,1H),4.29(m,2H),3.63(s,3H),2.92-2.81(m,2H),2.77(t,J=6.36 Hz,2H),2.51-2.41(m,5H),2.22-2.18(m,2H),2.09-2.01(m,2H),1.94-1.87(m,2H),1.62(m,2H),1.46(m,2H);
LC-MS(ESI+):m/z calculated for C46H43F2N8O6(M+H)+:841.32found 841.89.
Figure BDA0002117302440000471
1H-NMR(400MHz,CDCl3)δ(ppm)10.33(s,1H),9.97(s,1H),8.95(s,1H),7.83(d,J=7.52Hz,1H),7.66-7.59(m,3H),7.45(t,J=8.08Hz,1H),7.32(s,1H),7.21(s,1H),7.13(s,1H),6.84(m,3H),6.68(s,1H),6.43(s,1H),5.29(d,J=10.44Hz,1H),4.53(d,J=17.04Hz,1H),4.36-4.27(m,4H),3.65(s,3H),3.52(t,J=5.59Hz,2H),2.88-2.81(m,4H),2.44(m,2H),2.22(m,1H),2.06(m,2H),1.88(m,2H),1.68-1.63(m,4H),1.39(m,2H);
LC-MS(ESI+):m/z calculated for C47H45F2N8O7(M+H)+:871.33found 871.91.
Figure BDA0002117302440000472
Dissolving a carboxylic acid end raw material and 1.2 equivalents of HATU (condensing agent) in a DCM/DMF mixed solvent, adding 2.0 equivalents of triethylamine, stirring for 5-10 minutes at room temperature, dissolving 1.0 equivalent of the intermediate 2 in a proper amount of DCM/DMF mixed solvent, quickly dripping the intermediate into the reaction solution, and stirring overnight at room temperature. The reaction solution was added to 50mL of water and extracted with ethyl acetate (3X 10mL), the organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. The residue was separated and purified by 200-mesh 300-mesh silica gel chromatography column, and the mobile phase was dichloromethane: methanol 30: 1, obtaining the formula 16.
1H-NMR(400MHz,CDCl3)δ(ppm)10.02(s,1H),8.60(s,1H),7.63(d,J=2.32Hz,1H),7.57(s,1H),7.48-7.44(m,2H),7.18(t,J=2.40Hz,1H),7.14(s,1H),7.07(d,J=7.04Hz,1H),6.86-6.80(m,4H),6.70(t,J=7.72Hz,1H),6.42(s,1H),6.22(t,J=5.24Hz,1H),4.90(dd,J=12.28Hz,J=5.52Hz,1H),3.26(dd,J=12.60Hz,J=5.28Hz,1H),2.88-2.66(m,3H),2.38(t,J=7.32Hz,2H),2.12-2.09(m,1H),1.78(m,2H),1.67(m,2H),1.49(m,2H);
LC-MS(ESI+):m/z calculated for C39H35F2N6O7(M+H)+:736.25found 737.96.
Figure BDA0002117302440000481
1H-NMR(400MHz,DMSO-d6)δ(ppm)12.01(s,1H),11.11(s,1H),9.84(s,1H),8.20(s,1H),7.48(t,J=7.64Hz,1H),7.39(d,J=2.64Hz,1H),7.29(t,J=2.68Hz,1H),7.27(s,1H),7.21(dd,J=8.76Hz,J=2.60Hz,1H),7.14(t,J=8.20Hz,1H),7.05-6.98(m,3H),6.76(t,J=6.12Hz,1H),6.63(dd,J=8.16Hz,J=1.84Hz,1H),6.48(t,J=2.16Hz,1H),6.40(dd,J=8.20Hz,J=1.80Hz,1H),6.26(t,J=2.16Hz,1H),5.06-5.01(m,1H),4.99(s,2H),4.59(t,J=5.68Hz,2H),3.80(q,J=6.00Hz,2H),3.11(t,J=7.28Hz,2H),2.90-2.81(m,1H),2.59-2.45(m,2H),2.02-2.00(m,1H),1.22(t,J=7.24Hz,3H);
LC-MS(ESI+):m/z calculated for C40H38N9O9S(M+H)+:820.24found 820.90.
Preparation of compound represented by formula 18-50
Synthesis of a compound represented by formula 18:
Figure BDA0002117302440000482
dissolving a carboxylic acid end raw material and 1.2 equivalents of HATU (condensing agent) in a DCM/DMF mixed solvent, adding 2.0 equivalents of triethylamine, stirring at room temperature for 5-10 minutes, dissolving 1.0 equivalent of intermediate 2 in a proper amount of DCM/DMF mixed solvent, quickly dropping the intermediate into the reaction solution, and stirring at room temperature overnight. The reaction was added to 50mL of water and extracted with ethyl acetate (3 times 10mL each), the organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. The residue was separated and purified by 200-mesh 300-mesh silica gel chromatography column, and the mobile phase was dichloromethane: methanol 30: 1, obtaining the formula 18.
1H-NMR(400MHz,CDCl3)δ(ppm)9.86(s,1H),7.76(s,1H),7.56(d,J=2.48Hz,1H),7.50-7.45(m,2H),7.18(t,J=2.52Hz,1H),7.14-7.11(m,2H),7.09(d,J=8.36Hz,1H),6.99(d,J=8.36Hz,1H),6.87-6.81(m,3H),6.71(t,J=7.68Hz,1H),6.50(d,J=6.32Hz,1H),6.40(s,1H),4.89-4.85(m,1H),3.76-3.73(m,2H),3.63(s,3H),2.87-2.70(m,3H),2.08-2.05(m,1H),1.58-1.53(m,2H);
LC-MS(ESI+):m/z calculated for C36H29F2N6O7(M+H)+:695.20,found 695.96.
Compounds of formula 19 to 23 can be synthesized from intermediate 2 according to the above-described method
Figure BDA0002117302440000491
1H-NMR(400MHz,CDCl3)δ(ppm)7.66(s,1H),7.47-7.42(m,2H),7.21(d,J=1.80Hz,1H),7.14(s,1H),7.05(d,J=5.04Hz,1H),6.92(d,J=8.52Hz,1H),6.85-6.80(m,3H),6.68(t,J=7.48Hz,1H),6.40(s,1H),4.88-4.84(m,1H),3.62(s,3H),3.37(t,J=5.88Hz,2H),2.45(t,J=7.12Hz,2H),2.10-2.03(m,3H);
LC-MS(ESI+):m/z calculated for C37H31F2N6O7(M+H)+:709.21,found 709.84.
Figure BDA0002117302440000492
1H-NMR(400MHz,CDCl3)δ(ppm)9.82(s,1H),8.29(s,1H),7.66(d,J=2.32Hz,1H),7.52-7.43(m,3H),7.21(t,J=2.76Hz,1H),7.15(s,1H),7.07(d,J=7.12Hz,1H),6.89-6.81(m,4H),6.71(t,J=7.80Hz,1H),6.44(s,1H),6.25(s,1H),4.91-4.86(m,1H),3.64(s,3H),3.32(m,2H),2.89-2.69(m,3H),2.44(t,J=6.92Hz,2H),2.13-2.05(m,1H),1.90-1.84(m,2H),1.79-1.73(m,2H);
LC-MS(ESI+):m/z calculated for C38H33F2N6O7(M+H)+:723.23,found 723.42.
Figure BDA0002117302440000493
Figure BDA0002117302440000501
1H-NMR(400 MHz,CDCl3)δ(ppm)9.68(d,J=4.96Hz,1H),7.73(s,1H),7.44-7.38(m,2H),7.26-7.20(m,1H),7.18(s,1H),7.12(s,1H),7.01(d,J=7Hz,1H),6.84-6.78(m,4H),6.68-6.67(m,1H),6.38(d,J=2.04Hz,1H),4.87-4.84(m,1H),4.58(s,1H),3.58(s,3H),3.20(t,J=6.4Hz,2H),2.83-2.68(m,3H),1.99(s,3H),1.87(s,1H),1.65(m,2H),1.46(m,2H),1.23(m,2H);
LC-MS(ESI+):m/z calculated for C41H38F2N7O8(M+H)+:794.27,found 794.77.
Figure BDA0002117302440000502
1H-NMR(400 MHz,CDCl3)δ(ppm)9.92(s,1H),8.48(s,1H),7.66(d,J=2.40Hz,1H),7.49-7.42(m,3H),7.20(t,J=2.64Hz,1H),7.15(s,1H),7.07(d,J=7.08Hz,1H),6.88-6.80(m,4H),6.70(d,J=7.24Hz,1H),6.44(s,1H),6.21(t,J=5.20Hz,1H),4.92-4.87(m,1H),3.64(s,3H),3.27(q,J=6.28Hz,2H),2.89-2.66(m,3H),2.35(t,J=7.44Hz,2H),2.14-2.10(m,1H),1.79-1.71(m,2H),1.46-1.42(m,2H),0.89-0.83(m,4H);
LC-MS(ESI+):m/z calculated for C40H37F2N6O7(M+H)+:751.26,found 751.92.
Figure BDA0002117302440000503
1H-NMR(400 MHz,CDCl3)δ(ppm)9.98(s,1H),8.62(s,1H),7.69(d,J=2.48Hz,1H),7.48(d,J=7.56Hz,1H),7.42-7.38(m,2H),7.21(t,J=2.64Hz,1H),7.16(s,1H),7.07(d,J=7.08Hz,1H),6.87-6.81(m,4H),6.70(d,J=8.68Hz,1H),6.45(s,1H),6.21(t,J=5.40Hz,1H),4.92-4.88(m,1H),3.65(s,3H),3.25(q,J=6.72Hz,2H),2.89-2.68(m,3H),2.36(t,J=7.36Hz,2H),2.14-2.10(m,1H),1.75-1.71(m,2H),1.45-1.40(m,4H),0.89-0.83(m,4H);
LC-MS(ESI+):m/z calculated for C41H39F2N6O7(M+H)+:765.28,found 765.79.
Figure BDA0002117302440000511
The carboxylic acid end material was dissolved with 1.0 equivalent of intermediate 8, 1.2 equivalents of EDCI (condensing agent), 2.0 equivalents of HOBt, and 0.1 equivalent in a DCM/DMF mixed solvent, 2.0 equivalents of triethylamine was added, and stirred at room temperature overnight. The reaction was added to 50mL of water and extracted with ethyl acetate (3 times 10mL each), the organic phases were combined and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. The residue was separated and purified by 200-mesh 300-mesh silica gel chromatography column, and the mobile phase was dichloromethane: methanol 30: 1, obtaining the formula 24.
1H-NMR(400MHz,CDCl3)δ(ppm),8.36(d,J=2.76Hz,1H),8.12(dd,J=2.8Hz,J=9.08Hz,1H),7.52-7.45(m,3H),7.12(s,1H),7.07(d,J=7.08Hz,1H),6.97(d,J=8.56Hz,1H),6.91(t,J=8.12Hz,3H),6.31(d,J=2.84Hz,1H),4.89-4.85(m,1H),3.68(t,J=6.4Hz,2H),3.65(s,3H),2.85-2.68(m,3H),2.65(t,J=6Hz,2H),2.11-2.06(m,1H);
LC-MS(ESI+):m/z calculated for C36H30N7O9(M+H)+:704.20,found 704.86.
Compounds of formula 25 to 27 can be synthesized from intermediate 8 according to the above-described method
Figure BDA0002117302440000512
1H-NMR(400MHz,CDCl3:CD3OD=10:1)δ(ppm)8.35(d,J=2.68Hz,1H),8.11(dd,J=9.12Hz,J=2.76Hz,1H),7.51(d,J=8.84Hz,2H),7.46(t,J=7.28Hz,1H),7.13(s,1H),7.06(d,J=7.08Hz,1H),6.93-6.89(m,4H),6.31(d,J=2.80Hz,1H),4.89-4.85(m,1H),3.66(s,3H),3.36(m,2H),2.84-2.66(m,3H),2.44(t,J=7.08Hz,2H),2.07-2.03(m,3H);
LC-MS(ESI+):m/z calculated for C37H32N7O9(M+H)+:718.22,found 718.86.
Figure BDA0002117302440000521
1H-NMR(400MHz,CDCl3:CD3OD=10:1)δ(ppm),8.36(d,J=2.62Hz,1H),8.12(dd,d,J=11.20Hz,d,J=2.56Hz,1H),7.51-7.44(m,3H),7.26(s,1H),7.12(s,1H),7.05(d,J=7.08Hz,1H),6.94-6.88(m,4H),6.32(d,J=2.76Hz,1H),4.89-4.85(m,1H),3.66(s,3H),3.29(t,J=6.84Hz,2H),2.86-2.73(m,5H),2.12-2.07(m,1H),1.84-1.78(m,2H),1.75-1.70(m,2H);
LC-MS(ESI+):m/z calculated for C38H34N7O9(M+H)+:732.23,found 732.81.
Figure BDA0002117302440000522
1H-NMR(400MHz,CDCl3:CD3OD=10:1)δ(ppm)8.36(d,J=2.92Hz,1H),8.11(dd,J=9.04Hz,J=2.76Hz,1H),7.51(d,J=8.88Hz,2H),7.46(t,J=7.56Hz,1H),7.13(s,1H),7.05(d,J=7.12Hz,1H),6.92(d,J=8.80Hz,2H),6.86(d,J=8.52Hz,1H),6.32(d,J=2.76Hz,1H),4.90-4.85(m,1H),3.66(s,3H),3.26(t,J=6.84Hz,2H),2.86-2.68(m,3H),2.35(t,J=7.44Hz,2H),2.09-2.07(m,1H),1.77-1.65(m,4H),1.51-1.43(m,2H);
LC-MS(ESI+):m/z calculated for C39H36N7O9(M+H)+:746.25,found 746.79.
Compounds of formula 28 to formula 31 can be synthesized from intermediate 9 according to the above-described procedure
Figure BDA0002117302440000523
1H-NMR(400MHz,CDCl3:CD3OD=10:1)δ(ppm)8.38(d,J=2.64Hz,1H),8.16(dd,J=2.6Hz,J=9.08,1H),7.51(s,1H),7.46(t,J=7.64,1H),7.24(d,J=2Hz,1H),7.21(t,J=0.84Hz,1H),7.13(s,1H),7.05(q,J=7.16Hz,3H),6.94(d,J=8.8Hz,1H),6.67(d,J=7.6Hz,1H),6.33(d,J=2.48Hz,1H),4.90-4.85(m,1H),3.67(t,J=6.08Hz,2H),3.64(s,3H),2.85-2.73(m,3H),2.65(t,J=5.84,2H),2.10-2.02(m,1H);
LC-MS(ESI+):m/z calculated for C36H30N7O9(M+H)+:703.20,found 703.79.
Figure BDA0002117302440000531
1H-NMR(400 MHz,CDCl3:CD3OD=10:1)δ(ppm)8.35(d,J=2.76Hz,1H),8.10(dd,J=9.08Hz,J=2.80Hz,1H),7.46(t,J=1.96Hz,1H),7.41(dd,J=8.44Hz,J=7.20Hz,1H),7.24(d,J=2.84Hz,1H),7.21(d,J=8.04Hz,1H),7.13-7.11(m,2H),7.02(d,J=7.08Hz,1H),6.96(d,J=9.08Hz,1H),6.86(d,J=8.56Hz,1H),6.65(dd,J=7.92Hz,J=1.52Hz,1H),6.31(d,J=2.84Hz,1H),4.88-4.83(m,1H),3.64(s,3H),3.32(t,J=6.48Hz,2H),2.83-2.69(m,3H),2.41(t,J=7.16Hz,2H),2.08-1.97(m,3H);
LC-MS(ESI+):m/z calculated for C37H32N7O9(M+H)+:718.22,found 718.77.
Figure BDA0002117302440000532
1H-NMR(400 MHz,CDCl3)δ(ppm),8.38(s,1H),8.17-8.14(m,1H),7.48-7.44(m,2H),7.25-7.20(m,3H),7.13(s,1H),7.10-7.02(m,3H),6.86(d,J=8.6Hz,1H),6.67(d,J=7.24Hz,1H),6.33(s,1H),4.90-4.86(m,1H),3.66(s,3H),3.29(t,J=6.44,2H),2.87-2.70(m,3H),2.37(t,J=7.28Hz,2H),2.13-2.07(m,1H),1.82-1.78(m,2H),1.75-1.69(m,2H);
LC-MS(ESI+):m/z calculated for C38H34N7O9(M+H)+:731.23,found 731.86.
Figure BDA0002117302440000541
1H-NMR(400MHz,CDCl3:CD3OD=10:1)δ(ppm)8.37(d,J=2.76Hz,1H),8.13(dd,J=9.08Hz,J=2.80Hz,1H),7.50(s,1H),7.45(dd,J=8.44Hz,J=7.24Hz,1H),7.25(d,J=2.80Hz,1H),7.22(d,J=8.12Hz,1H),7.14(s,1H),7.11(d,J=7.96Hz,1H),7.05(d,J=7.08Hz,1H),6.99(d,J=9.08Hz,1H),6.84(d,J=8.52Hz,1H),6.67(dd,J=7.96Hz,J=1.64Hz,1H),6.32(d,J=2.84Hz,1H),4.88-4.84(m,1H),3.65(s,3H),3.24(t,J=6.80Hz,2H),2.85-2.68(m,3H),2.32(t,J=7.36Hz,2H),2.10-2.05(m,1H),1.75-1.64(m,4H),1.48-1.41(m,2H);
LC-MS(ESI+):m/z calculated for C39H36N7O9(M+H)+:746.25,found 746.79.
Figure BDA0002117302440000542
Formula 32 can be prepared by the process shown in formula 1. LC-MS (ESI)+):m/z calculated for C47H48F2N9O10(M+H)+:936.34,found 936.57.
Figure BDA0002117302440000543
Formula 33 can be prepared by the process shown in formula 1. LC-MS (ESI)+):m/z calculated for C43H32F2N9O8(M+H)+:848.29,found 848.95.
Figure BDA0002117302440000551
Formula 34 can be prepared by the process shown in formula 1. LC-MS (ESI)+):m/z calculated for C48H48N9O11S(M+H)+:958.31,found 958.43.
Figure BDA0002117302440000552
Formula 35 can be prepared by the process shown in formula 1. LC-MS (ESI)+):m/z calculated for C46H44N9O10S(M+H)+:914.29,found 914.73.
Figure BDA0002117302440000553
Formula 36 can be prepared by the process shown in formula 1. LC-MS (ESI)+):m/z calculated for C44H41N8O10S(M+H)+:873.26,found 873.79.
Figure BDA0002117302440000554
Formula 37 can be prepared by the process shown in formula 1. LC-MS (ESI)+):m/z calculated for C44H46N9O11S(M+H)+:908.30,found 908.49.
Figure BDA0002117302440000561
Formula 38 can be prepared by the method shown in formula 1. LC-MS (ESI)+):m/z calculated for C46H50N9O12S(M+H)+:952.32,found 952.93.
Figure BDA0002117302440000562
Formula 39 can be prepared by the method shown in formula 1. LC-MS (ESI) +):m/z calculated for C43H39N8O10S(M+H)+:859.24,found 859.71.
Figure BDA0002117302440000563
Formula 40 can be prepared by the method shown in formula 1. LC-MS (ESI)+):m/z calculated for C46H43F2N8O8(M+H)+:873.31,found 873.95.
Figure BDA0002117302440000564
Formula 41 can be prepared by the method shown in formula 1. LC-MS (ESI)+):m/z calculated for C45H47F2N8O7(M+H)+:843.30,found 843.89.
Figure BDA0002117302440000571
Formula 42 can be prepared by the process shown in formula 28. LC-MS (ESI)+):m/z calculated for C39H36N7O9(M+H)+:746.25,found 746.79.
Figure BDA0002117302440000572
Formula 43 can be prepared by the process shown in formula 28. LC-MS (ESI)+):m/z calculated for C39H36N7O9(M+H)+:746.25,found 746.92.
Figure BDA0002117302440000573
Formula 44 can be prepared by the process shown in formula 28. LC-MS (ESI)+):m/z calculated for C40H38N7O9(M+H)+:760.27,found 760.73.
Figure BDA0002117302440000574
Formula 45 can be prepared by the process shown in formula 28. LC-MS (ESI)+):m/z calculated for C40H38N7O9(M+H)+:760.27,found 760.57.
Figure BDA0002117302440000581
Formula 46 can be prepared by the process shown in formula 28. LC-MS (ESI)+):m/z calculated for C41H40N7O9(M+H)+:774.28,found 774.90.
Figure BDA0002117302440000582
Formula 47 can be prepared by the process shown in formula 28. LC-MS (ESI)+):m/z calculated for C41H40N7O9(M+H)+:774.28,found 774.82.
Figure BDA0002117302440000583
Formula 48 can be prepared by the process shown in formula 19. LC-MS (ESI)+):m/z calculated for C38H37F2N6O7(M+H)+:723.23,found 723.78.
Figure BDA0002117302440000584
Formula 49 can be prepared by the process shown in formula 19. LC-MS (ESI)+):m/z calculated for C39H37F2N6O7(M+H)+:737.25,found 737.48.
Figure BDA0002117302440000591
Formula 50 can be prepared by the method shown in formula 19. LC-MS (ESI)+):m/z calculated for C40H37F2N6O7(M+H)+:750.26,found 750.99.
Example 2 biological Activity test of Compounds represented by formulas 1 to 30 at the level of Western blotting experiment
Cell treatment:
taking Ramos or HBL-1 or IgE MM or Jurkat cells in logarithmic growth phase in a 6-well plate, and adding a compound for treatment; after incubation for the corresponding time, the cells were harvested.
Extracting cell whole protein:
collecting cells: the treated cells were scraped off in a culture medium, suspended thoroughly, centrifuged at 300g for 5 minutes and collected, washed once with PBS, and the PBS was discarded.
Cell lysis: adding 100 mu L of 2 XLoading Buffer into each sample, fully shaking and uniformly mixing, denaturing at 100 ℃ for 15 minutes, and storing at-20 ℃ after uniformly mixing or directly using for Western Blot detection.
The formula of 5 × Loading Buffer is as follows: 250mM Tris-HCl (pH6.8), 10% (W/V) SDS, 0.5% (W/V) bromophenol blue, 50% (V/V) glycerol, 5% (W/V) beta-mercaptoethanol (2-ME). The 2 XLoading Buffer is prepared by adding 1.5 times volume of dd water into 5 Xloading Buffer.
The specific steps of detection in Western blotting experiments (Western Blot, WB) are as follows:
1) SDS-PAGE gels were prepared at appropriate concentrations. The concentration of the separation gel is 8 percent, and the concentration of the concentrated gel is 5 percent.
2) Samples were prepared. Protein samples were prepared according to experimental requirements, denatured at 95 ℃ for 15 minutes, centrifuged, mixed and loaded onto SDS-PAGE gel wells. The loading volume is adjusted appropriately according to the protein quantification results, typically 8 μ L per well.
3) And (4) electrophoresis. And (3) switching on the power supply, wherein the voltage of the protein sample in the concentrated gel is 80 volts, and when the protein sample enters the separation gel, the voltage is adjusted to 120 volts to continue electrophoresis. The electrophoresis was stopped when bromophenol blue almost completely ran off the PAGE gel.
4) And (5) transferring the film. And (3) taking down the gel after electrophoresis is finished, and installing a film transfer device according to the following sequence: (negative electrode), filter paper, gel, activated PVDF membrane, filter paper, (positive electrode). Then the clamping and transferring device is placed in a film transferring buffer solution, and finally the ice box is placed in a refrigerator with the temperature of 4 ℃ and the constant voltage power-on is carried out for 1.5 hours at 100V.
5) And (5) sealing. After the completion of the membrane transfer, the PVDF membrane was removed, immersed in TBST buffer containing 5% skimmed milk powder, and shaken on a shaker at room temperature for 1 hour.
6) Primary antibody incubation. After blocking, the cells were washed 3 times with TBST buffer, and then primary antibody was added at a moderate dilution rate overnight at 4 ℃. The primary antibody was recovered and the PVDF membrane was washed 3 times with TBST buffer with 10 minutes shaking.
7) And (5) incubating a secondary antibody. The TBST buffer is discarded, secondary antibodies (mouse or rabbit antibodies, determined as primary antibodies) are added at a dilution ratio (usually 1: 3000-1: 5000), and the mixture is shaken in a shaker at room temperature for 1 hour. The secondary antibody was discarded and the PVDF membrane was washed 3 times with TBST buffer with shaking for 10 minutes. Finally, the mixture is washed with TBST buffer for 10 minutes.
8) Color development and tabletting. And (3) uniformly covering the ECL chromogenic substrate on the PVDF membrane, and developing for 0.5-15 minutes at room temperature.
The compounds of the examples of the invention have the following BET-degrading activity:
in the Jurkat cell line, the degradation of BET protein by the compounds represented by formulas 1, 9 and 13 was clearly observed as a result of Western Blotting (WB) as shown in FIG. 3 (Jurkat cell line: 2X 10)6Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; final DMSO concentration is one thousandth).
In the Rescue experiment, the BET protein degradation effect of the compound of formula 2 was clearly observed in the results of the Western blotting experiment (WB) in Ramos cell lines, as shown in FIGS. 4-1 and 4-2 (the concentration of formula 2 was 100 nM; the concentration of Cfz was 0.4. mu.M.; (+) -JQ-1, ABBV-075 and the concentration of Poma was 10. mu.M; the results of the protein degradation effect of the compound of formula 2 were 2X 10M in Ramos cell lines, as shown in Cfz, (+) -JQ-1, ABBV-075 and Poma in Western blotting experiment (WB)6Cells per well (6 well plate), 5% CO at 37 ℃2Adding Cfz, (+) -JQ-1, ABBV-075 and Poma, incubating for 2 hr, adding the compound of formula 2, 5% CO at 37 deg.C2Incubating for 2 hours; DMSO final concentration of 1%); no degradation of BET protein by Cfz, (+) -JQ-1, ABBV-075 and Poma was observed in the results of Western Blotting (WB) experiments.
In the HBL-1, Ramos, IgEMM and RPMI cell lines, the degradation of BET protein by the compounds of formulae 2, 5, 10, 13 and 15 (each compound concentration was 100nM) was clearly observed in the results of Western Blotting (WB) experiments, as shown in FIGS. 5-1, 5-2, 5-3 and 5-4 (HBL-1 cell line: 2X 10 cell line)6Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; the final concentration of DMSO is one thousandth; ramos cell line: 2X 10 6Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; the final concentration of DMSO is one thousandth; IgEMM cell lines: 2X 106Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; the final concentration of DMSO is one thousandth; RPMI cell lines: 2X 106Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; final DMSO concentration in thousandths).
In the Ramos cell line, it was clearly observed in the results of the western blotting experiment (WB) that the compound represented by formula 2 (at a concentration of 100nM) exhibited a decrease in the abundance of the downstream protooncogene c-MYC protein due to the degradation of the BET protein, as shown in FIG. 6-1 or FIG. 6-2. Wherein FIG. 6-1 shows that incubation of formula 2 for 2 hours causes down-regulation of c-MYC expression, and FIG. 6-2 shows that incubation of formula 2 for 6 hours causes more significant down-regulation of c-MYC expression (Ramos cell line: 2X 10)6Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; final DMSO concentration in thousandths).
In the HBL-1(BTK-C481S) cell line, it was clearly observed in the results of Western blotting experiment (WB) that the BET proteolysis effect of the compound of formula 2 incubated at low concentration (0.1nM, 0.5nM, 1nM, 2nM) for 2 hours was compared with the BET proteolysis effect of cells treated at the same time with the same concentration of ARV-825 as shown in FIG. 7 (HBL-1(BTK-C481S cell line: 2X 10) 6Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 2 hours: final DMSO concentration in thousandths).
Degradation of BET protein by the compounds represented by formula 16, formula 18, formula 19, formula 20, formula 21, formula 22, formula 23, formula 24, formula 25, formula 26, formula 27, formula 28, formula 29, and formula 30 was clearly observed in the results of Western blotting experiments (WB) in the Jurkat cell line, wherein formula 16, formula 19, and formula 21 exhibited a certain selectivity for BRD4 degradation, while formula 24 exhibited no selectivity for BRD2, and the results were shown in FIGS. 9-1, 9-2, and 9-3 (Jurkat cell line: 2X 10)6Cells per well (6 well plate), 5% CO at 37 ℃2Culturing for 24 hours; final DMSO concentration in thousandths).
Example 3 biological Activity test at cellular level of Compounds represented by formulas 1 to 50
MTT test reagent:
reagent: RPIM 1640 medium; DMEM medium; 100 × non-essential amino acids (NEAA); 100 times streptomycin mixed liquor; 50mM beta mercaptoethanol; fetal bovine serum (FBS, previously inactivated).
Medium a (500 mL): RPIM 1640 medium (450mL) +100 XNEAA (5mL) +100 Xpenicillin mixed solution (5mL) + fetal calf serum (50mL) +50mM beta-mercaptoethanol (0.5 mL).
B Medium (500 mL): DMEM medium (450mL) +100 XNEAA (5mL) +100 Xstreptomycin mixture (5mL) + fetal bovine serum (50mL) +50mM beta mercaptoethanol (0.5 mL). .
CCK-8 Kit (cell counting Kit-8)
MTT Experimental protocol (HBL-1 (BTK-C481S)):
1) cells were collected in log phase and cell suspension concentration was adjusted to 6.6X 10 using A medium4/mL。
2) The small molecule solution is prepared by diluting the small molecule with a 2-fold gradient of the A culture medium to 50nM to 0.15 nM.
3) 45 μ L of cell suspension was added to a 96-well plate (marginal wells filled with sterile PBS, 3000 cells/well). Negative controls (45. mu.L of cell suspension and 45. mu.L of A medium) were set for each plate, and 3 wells were set for each group.
4) Standing at 37 deg.C for 5% CO2After 1 hour of incubation, 45 μ L of the corresponding small molecule solution was added to each well of the 96-well plate. Then at 37 ℃ with 5% CO2Incubate for 72-96 hours. Add 10. mu.L cck-8 solution per well and continue incubation for 4 h. The absorbance of each well was measured by direct enzyme linked immunosorbent assay (OD 490 nm).
The BET protein degradation and cell proliferation inhibition results of the compounds of the present invention measured by the above-described method are shown in table 1, fig. 8-1, or fig. 8-2 below.
In addition, FIGS. 10-1 and 10-2 show the activity of compounds of formulae 2, 10, 21 and 24 compared to the activity of the lung cancer K562 cell line and the prostate cancer LNcap cell line. It can be seen that formula 21, which selectively degrades BRD4, and formula 24, which does not degrade BRD2, are less cytotoxic than non-selective formulas 2 and 10, and thus formula 21 and formula 24 may have greater safety and therapeutic prospects.
Table 1: the half Degradation Concentration (DC) of compounds shown in formula 1-formula 50 and compounds such as ABBV-075, (+) -JQ-1 and ARV-825 reported in the literature on BET protein of HBL-1(BTK-C481S) cell line50) And MTT assay for inhibitory potency (IC)50Value):
Figure BDA0002117302440000621
Figure BDA0002117302440000631
Figure BDA0002117302440000641
Figure BDA0002117302440000651
Figure BDA0002117302440000661
Figure BDA0002117302440000671
Figure BDA0002117302440000681
remarking: n.d indicates no detectable inhibitory activity.
As can be seen from the analysis in table 1 above, the compounds of the present invention have significant inhibitory effects on BET protein degradation and cell proliferation.
In the description of the specification, reference to the description of "one embodiment," "some embodiments," "an example," "a specific example," or "some examples" or the like means that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above are not necessarily intended to refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, various embodiments or examples and features of different embodiments or examples described in this specification can be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it will be understood that the above embodiments are exemplary and not to be construed as limiting the present invention, and that changes, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (9)

1. A compound that is a compound of formula I or a stereoisomer, geometric isomer, tautomer, or pharmaceutically acceptable salt thereof:
X-Y-Z
formula I
Wherein X represents a ligand for BET protein, Z represents a ligand for E3 ligase, and Y represents a chain linking X and Z;
wherein X is a compound shown as a formula III-2, III-4 or III-6,
Figure DEST_PATH_IMAGE002
z is a compound shown as a formula V-3 or V-4,
Figure FDA0003540419590000012
Figure DEST_PATH_IMAGE004
the Y is a compound represented by at least one of the following compounds,
Figure FDA0003540419590000022
each t6、t7、t8、t9Or t10Each independently is an integer of 0 to 11.
2. A compound represented by any one of formulas 1 to 50, or a stereoisomer, a geometric isomer, a tautomer, or a pharmaceutically acceptable salt thereof,
Figure FDA0003540419590000031
Figure FDA0003540419590000041
Figure FDA0003540419590000051
Figure FDA0003540419590000061
Figure FDA0003540419590000071
Figure FDA0003540419590000081
Figure FDA0003540419590000091
Figure FDA0003540419590000101
Figure FDA0003540419590000111
Figure FDA0003540419590000121
3. a pharmaceutical composition comprising a compound of claim 1 or 2.
4. The pharmaceutical composition of claim 3, further comprising an adjuvant.
5. The pharmaceutical composition of claim 3, further comprising other agents for treating or preventing non-Hodgkin's lymphoma, Burkitt's lymphoma, acute myelogenous leukemia, multiple myeloma, lung cancer, prostate cancer, and NUT midline cancer.
6. The pharmaceutical composition of claim 5, wherein the other drugs for treating or preventing non-Hodgkin's lymphoma comprise ibrutinib;
optionally, the other agent for treating or preventing Burkitt's lymphoma comprises at least one selected from cyclophosphamide, doxorubicine;
optionally, the other agent for treating or preventing acute myelogenous leukemia comprises at least one selected from cytarabine, Azacitidine, Decitabine;
optionally, the other agent for treating or preventing multiple myeloma comprises at least one selected from carfilzomib, thalidomide, lenalidomide, pomalidomide;
optionally, the other medicament for treating or preventing lung cancer comprises at least one selected from gefitinib, erlotinib, osthole, afatinib;
optionally, the other agent for treating or preventing prostate cancer comprises at least one selected from flutamide and nilutamide.
7. Use of a compound as claimed in claim 1 or 2 or a pharmaceutical composition as claimed in any one of claims 3 to 6 in the manufacture of a medicament for degrading BET proteins.
8. Use of a compound as claimed in claim 1 or claim 2 or a pharmaceutical composition as claimed in any one of claims 3 to 6 in the manufacture of a medicament for the treatment or prophylaxis of non-hodgkin's lymphoma, Burkitt's lymphoma, acute myelogenous leukemia, multiple myeloma, lung cancer, prostate cancer and NUT midline cancer.
9. A non-diagnostic therapeutic method for degrading BET protein, comprising: contacting a BET protein with a compound according to claim 1 or 2 or a pharmaceutical composition according to any one of claims 3 to 6.
CN201910594817.9A 2018-07-04 2019-07-03 Compound for targeted degradation of BET protein and application thereof Active CN110372693B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2018107265139 2018-07-04
CN201810726513.9A CN108690020A (en) 2018-07-04 2018-07-04 It is a kind of targeting degradation BET albumen compound and its application
CN2018094550 2018-07-04
CNPCT/CN2018/094550 2018-07-04

Publications (2)

Publication Number Publication Date
CN110372693A CN110372693A (en) 2019-10-25
CN110372693B true CN110372693B (en) 2022-05-03

Family

ID=68251800

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910594817.9A Active CN110372693B (en) 2018-07-04 2019-07-03 Compound for targeted degradation of BET protein and application thereof

Country Status (2)

Country Link
CN (1) CN110372693B (en)
WO (1) WO2020007322A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4083037A4 (en) * 2019-12-26 2024-01-24 Yonsei Univ Univ Industry Foundation Uif Pyrrolidine derivative, and pharmaceutical composition for preventing or treating beta-amyloid or tau protein-associated diseases containing same
CN113402520A (en) * 2020-03-16 2021-09-17 中国科学院上海药物研究所 WEE1 protein degradation agent
JP2023539463A (en) 2020-08-17 2023-09-14 アリゴス セラピューティクス インコーポレイテッド Methods and compositions for targeting PD-L1
CN113143930B (en) * 2021-04-08 2023-05-30 深圳湾实验室 Use of compound in preparing SARS-Cov-2E protein inhibitor
CN113603676B (en) * 2021-04-28 2022-05-24 浙江工业大学 Erlotinib-based EGFR protein targeted degradation small molecule compound and preparation method and application thereof
CN115551861A (en) * 2021-04-30 2022-12-30 成都苑东生物制药股份有限公司 Novel BRD4 bromodomain PROTAC protein degradation agent, preparation method and medical application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104136435A (en) * 2011-12-30 2014-11-05 艾伯维公司 Bromodomain inhibitors
WO2017223452A1 (en) * 2016-06-23 2017-12-28 Dana-Farber Cancer Institute, Inc. Degradation of bromodomain-containing protein 9 (brd9) by conjugation of brd9 inhibitors with e3 ligase ligand and methods of use
CN108690020A (en) * 2018-07-04 2018-10-23 清华大学 It is a kind of targeting degradation BET albumen compound and its application

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014125408A2 (en) * 2013-02-12 2014-08-21 Aurigene Discovery Technologies Limited Substituted 1h-pyrrolopyridinone derivatives as kinase inhibitors
JP6778114B2 (en) * 2014-04-14 2020-10-28 アルビナス・オペレーションズ・インコーポレイテッドArvinas Operations, Inc. Imid-based proteolysis modulators and related uses
MA40943A (en) * 2014-11-10 2017-09-19 Constellation Pharmaceuticals Inc SUBSTITUTED PYRROLOPYRIDINES USED AS BROMODOMA INHIBITORS
MA40940A (en) * 2014-11-10 2017-09-19 Constellation Pharmaceuticals Inc SUBSTITUTED PYRROLOPYRIDINES USED AS BROMODOMA INHIBITORS

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104136435A (en) * 2011-12-30 2014-11-05 艾伯维公司 Bromodomain inhibitors
WO2017223452A1 (en) * 2016-06-23 2017-12-28 Dana-Farber Cancer Institute, Inc. Degradation of bromodomain-containing protein 9 (brd9) by conjugation of brd9 inhibitors with e3 ligase ligand and methods of use
CN108690020A (en) * 2018-07-04 2018-10-23 清华大学 It is a kind of targeting degradation BET albumen compound and its application

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Discovery ofN-(4-(2,4-Difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)ethanesulfonamide (ABBV-075/Mivebresib), a Potent and Orally Available Bromodomain and Extraterminal Domain (BET) Family Bromodomain Inhibitor;Keith F. McDaniel等;《J. Med. Chem.》;20170926;第60卷;第8369页摘要 *

Also Published As

Publication number Publication date
CN110372693A (en) 2019-10-25
WO2020007322A1 (en) 2020-01-09

Similar Documents

Publication Publication Date Title
CN110372693B (en) Compound for targeted degradation of BET protein and application thereof
CN110724143B (en) Preparation of target BTK protein degradation compound and application of target BTK protein degradation compound in treatment of autoimmune system diseases and tumors
CN108690020A (en) It is a kind of targeting degradation BET albumen compound and its application
EP3083589B1 (en) Substituted piperazine compounds and methods of use thereof
WO2021093758A1 (en) Pyrimido derivative and application thereof in medicine
CN110845500B (en) Application of target BTK degradation compound in treatment of autoimmune system diseases
TWI828712B (en) Heterocyclic compounds as TRK inhibitors
CN106243088B (en) Substituted diethylenediamine compound and its application method and purposes
WO2019109415A1 (en) Compound for targeted degradation of hmgcr and application thereof
WO2022100624A1 (en) Oxy-substituted aminocarbonate thiophene-based compound and use thereof
CN114846005A (en) SHP2 inhibitor and application thereof
CN111303133A (en) Small molecule compound for degrading EZH2 protein
JPWO2003070730A1 (en) Pyrrolopyrimidine derivatives
AU2020401560A1 (en) Antagonists of the muscarinic acetylcholine receptor M4
WO2020182018A1 (en) Nitrogen heterocyclic compound, preparation method therefor and use thereof
JP4958379B2 (en) 1- [alkyl], 1-[(heteroaryl) alkyl] and 1-[(aryl) alkyl] -7-pyridinyl-imidazo [1,2-a] pyrimidin-5 (1H) -one derivatives
WO2021249463A1 (en) Bicyclic compound and application thereof
TW201629025A (en) Compounds and their use as BACE inhibitors
TW202128649A (en) 2-azaspiro[3.4]octane derivatives as m4 agonists
CN107759620B (en) Octahydropyrrolo [3,4-c ] pyrrole derivatives, methods of use, and uses thereof
WO2022174765A1 (en) Fused ring compound as wee-1 inhibitor
JP2023524863A (en) tricyclic heterocycle
WO2020006724A1 (en) Compound for targeted degradation of fak protein and use thereof
CN112851622A (en) Macrolide brefeldin A ester derivative and application thereof
CA3128419A1 (en) Dezocine derivative and medical 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
GR01 Patent grant
GR01 Patent grant