CN116444523A - Compounds as SMARCA2 inhibitors and uses thereof - Google Patents

Compounds as SMARCA2 inhibitors and uses thereof Download PDF

Info

Publication number
CN116444523A
CN116444523A CN202210005363.9A CN202210005363A CN116444523A CN 116444523 A CN116444523 A CN 116444523A CN 202210005363 A CN202210005363 A CN 202210005363A CN 116444523 A CN116444523 A CN 116444523A
Authority
CN
China
Prior art keywords
alkyl
alkoxy
hydroxy
amino
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210005363.9A
Other languages
Chinese (zh)
Inventor
陈耀忠
全旭
刘琪
张小猛
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.)
Nanjing Sanhome Pharmaceutical Co Ltd
Original Assignee
Nanjing Sanhome Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Sanhome Pharmaceutical Co Ltd filed Critical Nanjing Sanhome Pharmaceutical Co Ltd
Priority to CN202210005363.9A priority Critical patent/CN116444523A/en
Publication of CN116444523A publication Critical patent/CN116444523A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • 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

Landscapes

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

Abstract

The invention belongs to the field of pharmaceutical chemistryThe invention provides a compound shown in a formula (I) or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, a preparation method of the compound, a pharmaceutical composition containing the compound and application of the compound or the composition in treating SMARCA2 and/or SMARCA4 mediated diseases such as cancers.

Description

Compounds as SMARCA2 inhibitors and uses thereof
Technical Field
The present invention is in the field of medicinal chemistry, and in particular relates to compounds that are SMARCA2 inhibitors or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, methods for their preparation, pharmaceutical compositions containing these compounds and the use of these compounds or compositions for the treatment of SMARCA2 and/or SMARCA4 mediated diseases such as cancer.
Background
Genomic DNA in eukaryotic nuclei is entangled with histone octamers to form nucleosomes and further concentrated to form chromosomes, enabling the storage of large amounts of genetic information in a very compact form. However, the structure of chromatin limits the binding of DNA to various transcription regulators, so chromatin needs to be accessible to enhance chromosomal DNA by a remodeling (chromatin remodeling) process. A variety of protein Chromatin-Remodeling Complex (CRC) remodelling complexes can use the energy generated by ATP hydrolysis to slide or pop up nucleosomes, exposing DNA, allowing transcription factors and transcriptional regulatory proteins to bind, initiating gene transcription. Of these CRC, the SWI/SNF complex is one of the most studied complexes at present.
The SWI/SNF complex consists of core, catalytic and regulatory subunits, and contains three subtypes in total, namely classical BRG1/BRM associated factor (cBAF), polybroma-associated BAF (PBAF) and non-classical BAF (ncBAF). The core complex of SWI/SNF CRC for all subtypes consists of four core subunits BAF155 (encoded by SMARCC1 gene), BAF170 (encoded by SMARCC2 gene), INI1 (SNF 5 or BAF47, encoded by SMARCB1 gene), and ATPase subunit BRM (encoded by SMARCA2 gene)/BRG 1 (BRM/SWI 2-associated gene 1, encoded by SMARCA4 gene). BRM is a paralog to BRG1 and both proteins act as mutually exclusive ATP-dependent subunits in mammalian SWI/SNF chromatin remodeling complexes. The cells require BRM or BRG1 to assemble the catalytically active SWI/SNF complex. Multiple variants of SWI/SNF complexes have been characterized as having different subunit compositions, but only one catalytic subunit (BRM or BRG 1) is present in each complex. SWI/SNF gene mutations were found in approximately 20% of cancers, and sequencing data from 669 patients containing 24 tumors showed that SWI/SNF averaged mutations were next to TP53 (26%) and were located second in all genes. SWI/SNF gene mutations occur mainly in catalytic and regulatory subunits, such as SMARCA4, SMARCA2, ARID1A, ARID1B, and the like. Wherein SMARCA4 has function deletion mutation in small cell ovarian cancer, urothelial cancer, non-small cell lung cancer, gastric cancer, sarcoma, melanoma, head and neck cancer, liver cancer, cervical cancer, bile duct cancer, etc.; and 10-25% of mutations in non-small cell lung cancer cells, including 70-90% homozygous deletion mutations, are higher than in other types of tumor cells.
The deletion mutation of SMARCA4 has high correlation with the hypodifferentiation of non-small cell lung cancer tumor and is mutually exclusive with the mutation of EGFR, KRAS, ALK, RET and ROS1 genes. Thus, SMARCA4 deficient patients are likely not to benefit from inhibitors against the mutations described above. The total survival of SMARCA4 homozygous deletion mutant NSCLC patients was far worse than SMARCA4 heterozygous mutant and wild type patients under existing targeted molecular therapy and tumor immunotherapy treatment.
When SMARCA4 is inactivated, compensatory upregulation of SMARCA2 expression levels occurs. Proliferation of SMARCA4 homozygous deletion mutant tumor cells is highly sensitive to SMARCA2shRNA, such as NCI-H8383, NCI-H1299, and a549. However, SMARCA2shRNA had less effect on SMARCA4 wild type or cells with SMARCA4 heterozygous mutations and did not affect expression of other core SWI/SNF modules. In vivo experiments, the effect of SMARCA2 deletion on tumor growth was also significantly different for SMARCA4 mutation (NCI-H1299) and SMARCA4WT (NCI-H460). SMARCA2 knockout resulted in a significant decrease in Ki67 (proliferation marker) in SMARCA4 deleted NCI-H1299 tumors, but no significant change in SMARCA2 WT tumors.
In view of the above, SMARCA2 selective inhibitors have clear mechanism in treating SMARCA4 mutant non-small cell lung cancer and other tumor related diseases, and have great potential to be a new therapeutic means in the tumor therapeutic field, so safer and more effective SMARCA2 inhibitors need to be developed to meet clinical demands.
Disclosure of Invention
It is an object of the present invention to provide a class of compounds having SMARCA2 inhibitory activity represented by the general formula (I) or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
it is a further object of the present invention to provide a process for the preparation of the compounds of formula (I) of the present invention or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof.
It is a further object of the present invention to provide compositions comprising a compound of formula (I) of the present invention or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof and a pharmaceutically acceptable carrier, as well as compositions comprising a compound of formula (I) of the present invention or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof and another drug or drugs.
It is a further object of the present invention to provide a method of treating SMARCA2 and/or SMARCA4 mediated diseases such as cancer by the compound of the general formula (I) or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, and the use of the compound of the general formula (I) or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof for the preparation of a medicament for treating SMARCA2 and/or SMARCA4 mediated diseases such as cancer.
Aiming at the purpose of the invention, the invention provides the following technical scheme:
in a first aspect, the present invention provides a compound of formula (I) or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
wherein,,
R 1 selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylAmino, alkanoylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkylsulfonyl, aminosulfonyl and alkylaminosulfonyl;
R 2 selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, alkoxyalkyl;
a is selected from the group consisting of six-membered and five-membered heteroaromatic rings, six-membered and six-membered heteroaromatic rings, and five-membered and six-membered heteroaromatic rings, optionally substituted with one or more groups selected from halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylamido, alkanoyl, alkylsulfonyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, haloalkylacyl, hydroxyalkylacyl, cycloalkylacyl, heterocyclylacyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo groups;
L is a bond, -S-, -O-, -N-, alkylene, -C (O) -, -S (O) -or-S (O) 2 -; and
b is selected from the group consisting of alkyl, aryl, heteroaryl, cycloalkyl and heterocyclyl, optionally substituted with one or more groups selected from the group consisting of halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylamido, alkanoyl, alkylsulfonyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, haloalkylacyl, hydroxyalkylacyl, cycloalkylacyl, heterocyclylacyl, cycloalkyl, heterocyclyl, aryl, heteroaryl and oxo groups.
In some preferred embodiments, the compounds of the invention are compounds of formula (I) or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein:
R 1 selected from hydrogen, halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkanoyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, C 1-6 Alkylsulfonyl, aminosulfonyl and C 1-6 An alkylamino sulfonyl group;
further preferably, R 1 Selected from hydrogen, halogen, hydroxy, C 1-3 Alkyl, halogenated C 1-3 Alkyl, hydroxy C 1-3 Alkyl, C 1-3 Alkoxy, halo C 1-3 Alkoxy, hydroxy C 1-3 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-3 Alkylamino, C 1-3 Alkylacylamino, C 1-3 Alkanoyl, aminoacyl, C 1-3 Alkylaminoacyl, bis C 1-3 Alkylamino, C 1-3 Alkylsulfonyl, aminosulfonyl and C 1-3 An alkylamino sulfonyl group;
still more preferably, R 1 Selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, t-butyl, halogenated C 1-3 Alkyl, hydroxy C 1-3 Alkyl, C 1-3 Alkoxy, halo C 1-3 Alkoxy, hydroxy C 1-3 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-3 Alkylamino, C 1-3 Alkylacylamino, C 1-3 Alkanoyl, aminoacyl, C 1-3 Alkylaminoacyl, bis C 1-3 Alkylamino, methylsulfonyl, ethylsulfonyl, methylaminosulfonyl, ethylaminosulfonyl.
In some preferred embodiments, the compounds of the invention are compounds of formula (I) or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein:
R 2 Selected from hydrogen, halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro,carboxyl, cyano, amino, C 1-6 Alkoxy C 1-6 An alkyl group;
further preferably, R 2 Selected from hydrogen, halogen, hydroxy, C 1-3 Alkyl, halogenated C 1-3 Alkyl, hydroxy C 1-3 Alkyl, C 1-3 Alkoxy, halo C 1-3 Alkoxy, hydroxy C 1-3 Alkoxy, nitro, carboxyl, cyano, amino, C 1-3 Alkoxy C 1-3 An alkyl group;
still more preferably, R 2 Selected from hydrogen, hydroxy, methyl, ethyl, propyl, methoxy, ethoxy, methoxymethyl, ethoxymethyl.
In some preferred embodiments, the compounds of the invention are compounds of formula (I) or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein:
a is selected from Optionally substituted with one or more groups selected from halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkyl acyl, C 1-6 Alkylsulfonyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, alkenyl, alkynyl, halo C 1-6 Alkyl acyl, hydroxy C 1-6 Group substitution of alkyl acyl and oxo groups;
further preferably, A is selected from
In some preferred embodiments, the compounds of the invention are compounds of formula (I) or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein:
l is a bond, -S-, -O-, -N-, or sub-C 1-6 Alkyl, -C (O) -, -S (O) -or-S (O) 2 -;
Further preferably, L is a bond, methylene, ethylene, -C (O) -, -S (O) -.
In some preferred embodiments, the compounds of the invention are compounds of formula (I) or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein:
b is selected from C 1-6 Alkyl, aryl, heteroaryl, C 3-8 Cycloalkyl and C 3-8 Heterocyclyl, optionally substituted with one or more substituents selected from halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkyl acyl, C 1-6 Alkylsulfonyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, halo C 1-6 Alkyl acyl, hydroxy C 1-6 Group substitution of alkyl acyl and oxo groups;
Further preferably, B is selected fromAnd trifluoromethyl, optionally substituted with one or more groups selected from halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkyl acyl, C 1-6 Alkylsulfonyl, aminoacyl, C 1-6 Alkylamino groupAcyl, bis C 1-6 Alkylamino, halo C 1-6 Alkyl acyl, hydroxy C 1-6 Group substitution of alkyl acyl and oxo groups;
still more preferably, B is selected from trifluoromethyl,
In some preferred embodiments, the present invention provides compounds of formula (I) or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein formula (I) has the structure of formula (II),
wherein R is 1 、R 2 L and B have the definitions given for the general formula (I) above.
In some preferred embodiments, the present invention provides compounds of formula (I) or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein formula (I) has the structure of formula (IIa),
wherein,,
R 3 selected from hydrogen, halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkanoyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, C 1-6 Alkylsulfonyl, aminosulfonyl and C 1-6 An alkylamino sulfonyl group; preferably, R 3 Selected from hydrogen, halogen, hydroxy, C 1-3 Alkyl, halogenated C 1-3 Alkyl, hydroxy C 1-3 Alkyl, C 1-3 Alkoxy, halo C 1-3 Alkoxy, hydroxy C 1-3 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-3 Alkylamino, C 1-3 Alkylacylamino, C 1-3 Alkanoyl, aminoacyl, C 1-3 Alkylaminoacyl, bis C 1-3 Alkylamino, C 1-3 Alkylsulfonyl, aminosulfonyl and C 1 -3-alkylaminosulfonyl; still more preferably, R 1 Selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, t-butyl, halogenated C 1-3 Alkyl, hydroxy C 1-3 Alkyl, C 1-3 Alkoxy, halo C 1-3 Alkoxy, hydroxy C 1-3 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-3 Alkylamino, C 1-3 Alkylacylamino, C 1-3 Alkanoyl, aminoacyl, C 1-3 Alkylaminoacyl groups.
n is selected from 1, 2, 3 or 4;
R 1 and R is 2 Has the definition of the general formula (I).
In some specific embodiments, the compounds of formula (IIa) according to the invention or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof have the following IIaa structure:
wherein R is 1 、R 2 、R 3 And n has the definition given for the general formula (IIa) above.
The present invention provides the following specific compounds or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof:
in a second aspect, the present invention provides a pharmaceutical composition comprising a compound of the invention or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof.
In some embodiments, the invention provides a compound of the invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutical composition comprising a compound of the invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, for use in the treatment of SMARCA2 and/or SMARCA4 mediated diseases, such as cancer.
In some embodiments, the present invention provides pharmaceutical compositions comprising a compound of the present invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
The compounds of the present invention, or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, may be admixed with a pharmaceutically acceptable carrier, diluent or excipient to prepare a pharmaceutical formulation suitable for oral or parenteral administration. Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, and oral routes. The formulation may be administered by any route, for example by infusion or bolus injection, by absorption through the epithelial or skin mucosa (e.g. oral mucosa or rectum, etc.). Administration may be systemic or local. Examples of formulations for oral administration include solid or liquid dosage forms, specifically including tablets, pills, granules, powders, capsules, syrups, emulsions, suspensions and the like. The formulations may be prepared by methods known in the art and comprise carriers, diluents or excipients conventionally used in the art of pharmaceutical formulations.
In a third aspect, the present invention provides a method for treating SMARCA2 and/or SMARCA4 mediated diseases and the use of a compound of formulae (I), (II), (IIa) and (IIaa) of the present invention or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for treating SMARCA2 and/or SMARCA4 mediated diseases. In some specific embodiments, the disease is a cancer characterized by SMARCA2 and/or SMARCA4 defects or mutations. In some specific embodiments, the level of SMARCA4 activity or function in the cancer cells is reduced.
In some preferred embodiments, the present invention provides methods for treating SMARCA2 and/or SMARCA4 mediated diseases such as cancer and the use of the compounds of formula (I), (II), (IIa) or (IIaa) of the present invention or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, or pharmaceutical compositions comprising the same, for the manufacture of a medicament for treating SMARCA2 and/or SMARCA4 mediated diseases such as cancer. In some specific embodiments, the cancer is a cancer in which SMARCA2 and/or SMARCA4 defects or mutations are present. In other specific embodiments, the level of SMARCA4 activity or function in the cancer cell is reduced.
In some preferred embodiments, the present invention provides methods for treating cancer, including but not limited to: non-small cell lung cancer, lung adenocarcinoma, lung cancer, large cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, small cell lung cancer, melanoma, fibroproliferative melanoma, uveal melanoma, ovarian small cell carcinoma, rhabdoid tumor of ovary, cutaneous squamous cell carcinoma, glioma, uterine cancer, endometrial cancer, ovarian serous cyst adenocarcinoma, bladder urothelial carcinoma, primary central nervous system lymphoma, esophageal cancer, bladder cancer, gastric adenocarcinoma, adenoid cyst carcinoma, diffuse large B-cell lymphoma of lymphoid tumor, pancreatic cancer, colorectal adenocarcinoma, cholangiocarcinoma, sarcoma, head and neck cancer, cervical and cervical canal carcinoma, medulloblastoma, cutaneous T-cell lymphoma, hepatocellular carcinoma, renal papillary cell carcinoma, breast cancer, mantle cell lymphoma gall bladder cancer, testicular germ cell cancer, renal cell clear cell carcinoma, prostate cancer, pediatric Ewing's sarcoma, thymoma, renal chromocytoma, renal non-clear cell carcinoma, pheochromocytoma and paraganglioma, thyroid cancer, malignant peripheral nerve sheath tumor, neuroendocrine prostate cancer, head and neck squamous cell carcinoma, adrenocortical carcinoma, cervical cancer and cervical canal cancer, cutaneous squamous cell carcinoma, testicular germ cell carcinoma, glioblastoma multiforme, ewing's sarcoma, clear cell renal cell carcinoma, neuroblastoma, diffuse large B cell lymphoma, acute myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, malignant rhabdoid tumor, epithelioid sarcoma, renal medullary carcinoma, synovial sarcoma, meningioma and soft tissue sarcoma.
Definition of terms
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
The terms "hydrogen", "carbon", "oxygen" in the compounds of the present invention include all isotopes thereof. Isotopes are understood to include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include protium, tritium, and deuterium, isotopes of carbon include 12 C、 13 C and C 14 Isotopes of C, oxygen include 16 O and 18 o, etc.
"isomer" in the present invention refers to molecules of the same atomic composition and manner of attachment, but differing in their three-dimensional spatial arrangement, including, but not limited to, diastereomers, enantiomers, cis-trans isomers, and mixtures thereof, such as racemic mixtures. Many organic compounds exist in optically active form, i.e. they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefix D, L or R, S is used to denote the absolute configuration of the chiral center of the molecule. The prefix D, L or (+), (-) is used to name the sign of the compound plane polarized light rotation, (-) or L means that the compound is left-handed and the prefix (+) or D means that the compound is right-handed. The chemical structures of these stereoisomers are identical, but the stereoisomers are not identical. The particular stereoisomer may be an enantiomer, and the mixture of isomers is commonly referred to as an enantiomeric mixture. The 50:50 enantiomeric mixture is known as a racemic mixture or racemate, which may result in the absence of stereoselectivity or stereospecificity during chemical reactions. The terms "racemic mixture" and "racemate" refer to a mixture of two enantiomers in equimolar amounts, lacking optical activity.
Depending on the choice of starting materials and methods, the compounds of the invention may be present in the form of one of the possible isomers or mixtures thereof, for example racemates and non-corresponding isomer mixtures, depending on the number of asymmetric carbon atoms. Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
The resulting mixture of any stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, e.g., by chromatography and/or fractional crystallization, depending on the differences in the physicochemical properties of the components.
The "halogen" in the present invention means fluorine, chlorine, bromine, iodine. "halo" in the present invention means substituted with fluorine, chlorine, bromine or iodine.
"alkyl" in the present invention refers to a straight or branched saturated aliphatic hydrocarbon group, preferably a straight or branched group having 1 to 6 carbon atoms, further preferably a straight or branched group having 1 to 3 carbon atoms, non-limiting examples of which include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl and the like. Alkyl groups may be substituted or unsubstituted, and when substituted, the substituents may be at any useful point of attachment.
The term-C (O) -as used herein means "carbonyl".
The invention is-S (O) 2 -means "sulfonyl".
"haloalkyl" in the present invention refers to an alkyl group substituted with at least one halogen.
"hydroxyalkyl" in the present invention refers to an alkyl group substituted with at least one hydroxy group.
"alkoxy" in the present invention refers to an-O-alkyl group. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, n-propoxy, isopropoxy, isobutoxy, sec-butoxy, and the like. Alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents may be at any useful point of attachment.
"cycloalkyl" in the present invention refers to a cyclic saturated hydrocarbon group. Suitable cycloalkyl groups may be substituted or unsubstituted, monocyclic, bicyclic or tricyclic saturated hydrocarbon groups having 3 to 12 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
"heterocyclyl" of the present invention refers to groups of 3-to 12-membered non-aromatic ring systems ("3-12 membered heterocyclyl") having 1 to 4 ring heteroatoms, each of which is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus and silicon. In heterocyclyl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as long as the valence permits. The heterocyclyl groups may either be monocyclic ("monocyclic heterocyclyl") or fused, bridged or spiro ring systems (e.g., bicyclic systems (also known as "bicyclic heterocyclyl")) and may be saturated or may be partially unsaturated. Suitable heterocyclyl groups include, but are not limited to, piperidinyl, azetidinyl, aziridinyl, tetrahydropyrrolyl, piperazinyl, dihydroquinazolinyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, Etc. Each instance of a heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent may be at any useful point of attachment.
"aryl" in the present invention refers to aromatic systems which may comprise a single ring or a fused multiple ring, preferably a single ring or a fused double ring, containing from 6 to 12 carbon atoms, preferably from about 6 to about 10 carbon atoms. Suitable aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, fluorenyl, indanyl. Aryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents may be at any useful point of attachment.
"heteroaryl" or "heteroaryl ring" in the present invention refers to an aryl group having at least one carbon atom replaced with a heteroatom, preferably consisting of 5 to 12 atoms (5-12 membered heteroaryl), more preferably 5 to 10 atoms (5-10 membered heteroaryl), said heteroatom being O, S, N. The heteroaryl group includes, but is not limited to, imidazolyl, pyrrolyl, furanyl, thienyl, pyrazolyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, indolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, isoindolyl, benzopyrazolyl, benzimidazolyl, benzofuranyl, benzopyranyl, benzothienyl, benzoxazolyl, benzothiazolyl, benzisoxazolyl, benzisothiazolyl, quinolinyl, isoquinolinyl, quinazolinyl, cinnolinyl, quinoxalinyl, benzoxazinyl, benzothiazinyl, imidazopyridinyl, pyrimidopyrazyl, pyrimidoimidazolyl, Etc. Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents may be at any useful point of attachment.
The term "pharmaceutically acceptable salts" as used herein refers to salts of the compounds of the present invention which are safe and effective when used in a mammal, and which possess the desired biological activity.
"solvate" according to the present invention is intended in the conventional sense to mean a complex formed by the combination of a solute (e.g. active compound, salt of active compound) and a solvent (e.g. water). The solvent refers to a solvent known to or easily determined by those skilled in the art. In the case of water, the solvate is generally referred to as a hydrate, such as a hemihydrate, a monohydrate, a dihydrate, a trihydrate, or an alternative amount thereof, and the like.
The in vivo effect of the compound of formula (I) may be exerted in part by one or more metabolites formed in the human or animal body following administration of the compound of formula (I). As mentioned above, the in vivo effects of the compounds of formula (I) may also be exerted via metabolism of the precursor compounds ("prodrugs"). The "prodrug" of the present invention means a compound which is converted into a compound of the present invention by reaction with an enzyme, gastric acid or the like under physiological conditions in an organism, that is, a compound which is converted into a compound of the present invention by oxidation, reduction, hydrolysis or the like of an enzyme and/or a compound which is converted into a compound of the present invention by hydrolysis reaction of gastric acid or the like, or the like.
The "crystal" of the present invention means a solid whose internal structure is formed by repeating constituent atoms (or groups thereof) regularly in three dimensions, unlike an amorphous solid which does not have such a regular internal structure.
The term "pharmaceutical composition" according to the instant invention shall mean a mixture comprising any one of the compounds of the instant invention, including the corresponding isomer, prodrug, solvate, pharmaceutically acceptable salt or chemically protected form thereof, and one or more pharmaceutically acceptable carriers and/or another drug or drugs. The purpose of the pharmaceutical composition is to facilitate the administration of the compound to the organism. The compositions are generally useful in the manufacture of medicaments for the treatment and/or prophylaxis of diseases mediated by one or more kinases.
By "pharmaceutically acceptable carrier" is meant a carrier that does not cause significant irritation to the organism and does not interfere with the biological activity and properties of the compound being administered, and that comprises all solvents, diluents or other excipients, dispersants, surfactant isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like. Unless any conventional carrier medium is incompatible with the compounds of the present invention. Some examples of pharmaceutically acceptable carriers include, but are not limited to, sugars such as lactose, glucose, and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, cellulose and cellulose acetate; malt, gelatin, and the like.
The "excipient" of the present invention refers to an inert substance added to a pharmaceutical composition to further facilitate administration of the compound. Excipients may include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols.
Detailed Description
The present invention will be described in further detail with reference to examples, but the present invention is not limited to these examples. The materials used in the examples below are commercially available unless otherwise specified.
EXAMPLE 1- (tert-butyl) -N- (2- ((4- (3- ((2S, 6R) -2, 6-dimethylmorpholinyl) imidazo [1,2-b ] pyridazin-6-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
Step 1 preparation of (2S, 6R) -4- (6-chloroimidazo [1,2-b ] pyridazin-3-yl) -2, 6-dimethylmorpholine
3-bromo-6-chloroimidazo [1,2-b]Pyridazine (1G, 4.3 mmol), (2R, 6S) -2, 6-dimethylmorpholine (0.75G, 6.5 mmol), methanesulfonic acid (2-di-tert-butylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (tBuXphos Pd G3,0.34G,0.43 mmol), sodium t-butoxide (t-Buona, 1.24G,12.9 mmol) were dissolved in anhydrous 1, 4-dioxane (10 mL) and then reacted at 60℃for 3 hours under nitrogen protection. After the reaction is finished, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, purifying by column chromatography to obtain the title compound, LC-MS M/z [ M+1 ] ] + :267.1。
Step 2: preparation of tert-butyl (2- ((4-bromothiazol-2-yl) amino) -2-oxoethyl) carbamate
4-bromothiazol-2-amine (0.68 g,3.8 mmol), N-t-butoxycarbonylamino (1 g,5.7 mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 2.9g,7.6 mmol), N, N-diisopropylethylamine (1.47 g,11.4 mmol) were dissolved in anhydrous dichloromethane (6.5 mL) and stirred overnight at room temperature. After the reaction is finished, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, purifying by column chromatography to obtain the title compound, LC-MS M/z [ M+1 ]] + :336.0。
Step 3: preparation of (2- (2- ((tert-butoxycarbonyl) amino) acetamido) thiazol-4-yl) boronic acid
Tert-butyl (2- ((4-bromothiazol-2-yl) amino) -2-oxoethyl) carbamate (0.6 g,1.8 mmol), pinacol biborate (2.28 g,9.0 mmol), tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 0.17g,0.18 mmol), 2-dicyclohexylphosphine-2 ',4',6' -triisopropylbiphenyl (X-Phos, 0.17g,0.36 mmol), potassium acetate (0.53 mg,5.4 mmol) were dissolved in anhydrous 1, 4-dioxane (5 mL) and reacted at 101℃for 4 hours under nitrogen protection. After the reaction is finished, concentrating the reaction solution, dissolving ethyl acetate, filtering by a Buchner funnel, filtering the filtrate by an organic filter membrane, concentrating the obtained filtrate, preparing and separating the title compound by prep-HPLC (acidity) (preparation liquid phase), and LC-MS M/z [ M+1 ] ] + :302.1。
Step 4: preparation of tert-butyl (2- ((4- (3- ((2 s,6 r) -2, 6-dimethylmorpholinyl) imidazo [1,2-b ] pyridazin-6-yl) thiazol-2-yl) amino) -2-oxoethyl) carbamate
(2- (2- ((tert-Butoxycarbonyl) amino) acetamido) thiazol-4-yl) boronic acid (451.6 mg,1.5 mmol), (2S, 6R) -4- (6-chloroimidazo [1, 2-b)]Pyridazin-3-yl) -2, 6-dimethylmorpholine (200.0mg,0.75 mmol), potassium phosphate (477.6 mg,2.25 mmol), 1-bis (diphenylphosphine) dicyclopentadienyl iron palladium dichloride (54.9 mg,0.075 mmol) was dissolved in anhydrous 1, 4-dioxane (4 mL) and water (0.5 mL) and reacted at 90℃for 2 hours under nitrogen. After the reaction, the reaction solution is concentrated, dissolved in ethyl acetate, filtered by a Buchner funnel, and the concentrated reaction solution is purified by column chromatography to obtain the title compound, LC-MS M/z [ M+1 ]] + :488.2。
Step 5: preparation of 2-amino-N- (4- (3- ((2S, 6R) -2, 6-dimethylmorpholinyl) imidazo [1,2-b ] pyridazin-6-yl) thiazol-2-yl) acetamide
Tert-butyl (2- ((4- (3- ((2S, 6R) -2, 6-dimethylmorpholinyl) imidazo [1, 2-b)]Pyridazin-6-yl) thiazol-2-yl) amino) -2-oxoethyl carbamate (110.0 mg,0.23 mmol) was dissolved in anhydrous dichloromethane (1 mL), cooled to 0℃and hydrochloric acid/dioxane (4M, 0.56mL,2.3 mmol) was slowly added and stirred at room temperature for 1 hour. After the reaction, directly concentrating the reaction solution to obtain the title compound, LC-MS M/z [ M+1 ] ] + :388.2。
Step 6: preparation of 1- (tert-butyl) -N- (2- ((4- (3- ((2 s,6 r) -2, 6-dimethylmorpholinyl) imidazo [1,2-b ] pyridazin-6-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
2-amino-N- (4- (3- ((2S, 6R) -2, 6-dimethylmorpholinyl) imidazo [1, 2-b)]Pyridazin-6-yl) thiazol-2-yl) acetamide (90.0 mg,0.23 mmol), 1-tert-butyl-1H-pyrrole-3-carboxylic acid (50.2 mg,0.3 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 174.9mg,0.46 mmol) and N, N-diisopropylethylamine (118.9 mg,0.92 mmol) were dissolved in anhydrous dichloromethane (1.5 mL) and stirred at room temperature for 2 hours. After the reaction is finished, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtainThe title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.45(s,1H),8.21-8.18(m,1H),8.08(d,J=9.5Hz,1H),7.92(s,1H),7.64(d,J=9.5Hz,1H),7.54-7.50(m,1H),7.35(s,1H),7.00-6.95(m,1H),6.50-6.46(m,1H),4.12(d,J=5.5Hz,2H),3.92-3.88(m,3H),3.68-3.66(m,3H),1.50(s,9H),1.17(d,J=6.1Hz,6H).LC-MS m/z[M+1] + :537.1。
Example 2:1- (tert-butyl) -N- (2- (4- (1-methyl-2-oxo-3- ((tetrahydro-2H-pyran-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of 5-bromo-1-methyl-3- ((tetrahydro-2H-pyran-4-yl) methyl) -1, 3-dihydro-2H-benzo [ d ] imidazol-2-one
5-bromo-1-methyl-1, 3-dihydro-2H-benzo [ d ]]Imidazol-2-one (1 g,4.42 mmol), 4- (bromomethyl) tetrahydro-2H-pyran (3.15 g,17.68 mmol) were dissolved in anhydrous dichloromethane (10 mL), cooled to 0deg.C, cesium carbonate (6.48 g,19.89 mmol) was slowly added, stirred for 2 min, brought to 70deg.C and reacted overnight at reflux. After the reaction is finished, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, purifying by column chromatography to obtain the title compound, LC-MS M/z [ M+1 ] ] + :325.1。
Step 2: preparation of 1-methyl-3- ((tetrahydro-2H-pyran-4-yl) methyl) -5- (4, 5-tetramethyl-1, 3, 2-dioxabenzaldehyde-2-yl) -1, 3-dihydro-2H-benzo [ d ] imidazol-2-one
5-bromo-1-methyl-3- ((tetrahydro-2H-pyran-4-yl) methyl) -1, 3-dihydro-2H-benzo [ d ]]Imidazol-2-one (1.23 g,3.79 mmol), unionPinacol borate (5.79 g,22.8 mmol), catalyst 1, 1-bis (diphenylphosphine) dicyclopentadienyl iron palladium dichloride (0.28 g,0.379 mmol) and potassium acetate (0.74 g,7.59 mmol) were dissolved in anhydrous 1, 4-dioxane (13 mL) and reacted under reflux under nitrogen for 6 hours. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ]] + :373.2。
Step 3: preparation of tert-butyl (2- ((4-bromothiazol-2-yl) amino) -2-oxoethyl) carbamate
/>
4-bromothiazol-2-amine (0.68 g,3.8 mmol), N-t-butoxycarbonylamino (1 g,5.7 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 2.9g,7.6 mmol), N, N-diisopropylethylamine (1.47 g,11.4mm mol) were dissolved in anhydrous dichloromethane (6.5 mL) and stirred overnight at room temperature. After the reaction is finished, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, purifying by column chromatography to obtain the title compound, LC-MS M/z [ M+1 ] ] + :336.0。
Step 4: preparation of tert-butyl (2- ((4- (1-methyl-2-oxo-3- ((tetrahydro-2H-pyran-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) thiazol-2-yl) amino) -2-oxoethyl) carbamate
Tert-butyl (2- ((4-bromothiazol-2-yl) amino) -2-oxoethyl) carbamate (225 mg,0.67 mmol), 1-methyl-3- ((tetrahydro-2H-pyran-4-yl) methyl) -5- (4, 5-tetramethyl-1, 3, 2-dioxan-2-yl) -1, 3-dihydro-2H-benzo [ d ]]Imidazole-2-one (500 mg,1.34 mmol), catalyst 1, 1-bis (diphenylphosphine) dipyridyl iron palladium dichloride (49 mg,0.067 mmol) and potassium phosphate (447 mg,2.01 mmol) were dissolved in anhydrous 1, 4-dioxane (5 mL) and reacted overnight under reflux under nitrogen. Reaction completionThen, the mixture was washed with saturated sodium chloride, extracted with ethyl acetate, dried over anhydrous sodium sulfate, and purified by column chromatography to give the title compound. LC-MS M/z [ M+1 ]] + :502.3。
Step 5: preparation of 2-amino-N- (4- (1-methyl-2-oxo-3- ((tetrahydro-2H-pyran-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) thiazol-2-yl) acetamide
Tert-butyl (2- ((4- (1-methyl-2-oxo-3- ((tetrahydro-2H-pyran-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d)]Imidazol-5-yl) thiazol-2-yl) amino) -2-oxoethyl carbamate (226 mg,0.45 mmol) was dissolved in anhydrous dichloromethane (2 mL), cooled to 0 ℃, then hydrochloric acid/dioxane (4 m,1.1mL,4.5 mmol) was slowly added dropwise, and stirred at room temperature overnight. After the reaction, directly concentrating the reaction solution to obtain the title compound, LC-MS M/z [ M+1 ] ] + :402.2。
Step 6: preparation of 1- (tert-butyl) -N- (2- (4- (1-methyl-2-oxo-3- ((tetrahydro-2H-pyran-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
2-amino-N- (4- (1-methyl-2-oxo-3- ((tetrahydro-2H-pyran-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ]]Imidazol-5-yl) thiazol-2-yl) acetamide (57 mg,0.13 mmol) was dissolved in anhydrous dichloromethane (1 mL), cooled to 0deg.C, N, N-diisopropylethylamine (50 mg,0.39 mmol) was added dropwise followed by 1-tert-butyl-1H-pyrrole-3-carboxylic acid (32.7 mg,0.20 mmol) and 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 99.2mg,0.26 mmol) and stirred at room temperature for 3 hours. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.26(s,1H),8.18-8.15(m,1H),7.69(s,1H),7.66(d,J=8.2Hz,1H),7.56(s,1H),7.52(s,1H),7.19(d,J=8.1Hz,1H),7.00-6.95(m,1H),6.51-6.45(m,1H),4.10(d,J=5.6Hz,2H),3.84-3.82(m,2H),3.76(d,J=7.0Hz,2H),3.35(s,3H),3.24(t,J=11.2Hz,2H),2.12-2.00(m,1H),1.49(s,9H),1.38-1.26(m,4H).LC-MS m/z[M+1] + :551.3。
EXAMPLE 3 1- (tert-butyl) -N- (2-oxo-2- ((4- (1- ((tetrahydro-2H-pyran-4-yl) methyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) thiazol-2-yl) amino) ethyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of 6-bromo-1- ((tetrahydro-2H-pyran-4-yl) methyl) -1H-pyrrolo [2,3-b ] pyridine
6-bromo-1H-pyrrole [2,3-b ]]Pyridine (500 mg,2.55 mmol), 4- (bromomethyl) tetrahydro-2H-pyran (545 mg,3.06 mmol) was dissolved in anhydrous tetrahydrofuran (5 mL), cesium carbonate (325.8 mg,6.38 mmol) was added under ice-bath, and then transferred to heat reflux and stirred overnight. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ] ] + :295.1。
Step 2-step 6: the synthesis was identical to steps 2 to 6 of example 1, thereby obtaining the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.33(s,1H),8.19(m,1H),8.01(d,J=8.1Hz,1H),7.80(s,1H),7.75(d,J=8.1Hz,1H),7.57(d,J=3.2Hz,1H),7.52(s,1H),7.02-6.93(m,1H),6.54-6.42(m,2H),4.21(d,J=7.0Hz,2H),4.11(d,J=5.6Hz,2H),3.86-3.78(m,2H),3.28-3.19(m,2H),1.50(s,9H),1.45-1.37(m,3H),1.33-1.27(m,2H).LC-MS m/z[M+1] + :521.2。
EXAMPLE 4 1- (tert-butyl) -N- (2-oxo-2- ((4- (1- (2, 2-trifluoroethyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) thiazol-2-yl) amino) ethyl) -1H-pyrrole-3-carboxamide
/>
Step 1: preparation of 6-bromo-1- (2, 2-trifluoroethyl) -1H-pyrrolo [2,3-b ] pyridine
6-bromo-1H-pyrrole [2,3-b ]]Pyridine (0.5 g,2.55 mmol) was dissolved in anhydrous N, N-dimethylformamide (5 mL), cooled to 0deg.C, 1-trifluoro-2-iodoethane (1.07 g,5.1 mmol) was added, after stirring for ten minutes sodium hydrogen (0.1 g,2.55 mmol) was slowly added, followed by transfer to room temperature and stirring for 3 hours, and then transfer to 40℃for oil bath reaction overnight. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ]] + :279.0。
Step 2-step 6: the synthesis was identical to steps 2 to 6 of example 1, thereby obtaining the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.34(s,1H),8.20-8.16(t,J=8Hz,1H),8.09(d,J=8Hz,1H),7.86(s,1H),7.83(d,J=8Hz,1H),7.62(d,J=4Hz,1H),7.52(s,1H),6.97(s,1H),6.62(d,J=4Hz,1H),6.50-6.46(m,1H),5.26-5.20(m,2H),4.11(d,J=5.7Hz,2H),1.50(s,9H).LC-MS m/z[M+1] + :505.1。
EXAMPLE 5 1- (tert-butyl) -N- (2- ((4- (1- (oxetan-3-ylmethyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
The procedure was followed in accordance with example 4 except that 2-iodo-1, 1-trifluoroethane was changed to 3- (bromomethyl) oxetane, thereby obtaining the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.33(s,1H),8.20-8.17(m,1H),8.03(d,J=8.1Hz,1H),7.80(s,1H),7.77(d,J=8Hz,1H),7.61(d,J=3.0Hz,1H),7.55-7.51(m,1H),7.00-6.96(m,1H),6.51-6.46(m,2H),4.67-4.64(m,2H),4.61(d,J=7.1Hz,2H),4.54-4.51(m,2H),4.11(d,J=5.5Hz,2H),3.55-3.49(m,1H),1.50(s,9H).LC-MS m/z[M+1] + :493.2。
EXAMPLE 6 1- (tert-butyl) -N- (2-oxo-2- ((4- (1- ((tetrahydro-2H-pyran-4-yl) methyl) -1H-indol-6-yl) thiazol-2-yl) amino) ethyl) -1H-pyrrole-3-carboxamide
The synthesis was identical to that of example 2, except that 5-bromo-1-methyl-1, 3-dihydro-2H-benzo [ d ]]The title compound was prepared by substituting imidazol-2-one with 6-bromoindole. 1 H NMR(400MHz,DMSO-d 6 )δ12.27(s,1H),8.17(t,J=5.2Hz,1H),7.99(s,1H),7.65-7.47(m,4H),7.40(d,J=2.2Hz,1H),7.04-6.92(m,1H),6.53-6.45(m,1H),6.43(d,J=1.7Hz,1H),4.18-3.98(m,4H),3.88-3.78(m,2H),3.27-3.17(m,2H),2.16-2.02(m,1H),1.50(s,9H),1.43-1.26(m,4H).LC-MS m/z[M+1] + :520.2。
EXAMPLE 7 1- (tert-butyl) -N- (2- ((4- (1-methyl-3- (morpholinomethyl) -1H-indol-5-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of 4- ((5-bromo-1-methyl-1H-indol-3-yl) methyl) morpholine
5-bromo-1-methylindole-3-carbaldehyde (0.4 g,1.69 mmol) and morpholine (0.19 g,2.19 mmol) were dissolved in anhydrous dichloromethane (4 mL), cooled to 0deg.C, slowly added sodium triacetoxyborohydride (1 g,5.07 mmol) and stirred overnight at room temperature. After the reaction was completed, the mixture was cooled on iceWater quenching is added under the bath, dichloromethane extraction, saturated sodium chloride washing, anhydrous sodium sulfate drying and direct concentration of the organic phase to obtain the title compound. LC-MS M/z [ M+1 ]] + :309.1。
Step 2-step 6: the synthesis was identical to steps 2 to 6 of example 2, to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.27(s,1H),8.20-8.13(m,2H),7.71(d,J=8.3Hz,1H),7.52(s,1H),7.44-7.42(m,2H),7.26(s,1H),6.97(s,1H),6.48(s,1H),4.10(d,J=5.5Hz,2H),3.77(s,3H),3.63-3.65(m,6H),2.45-2.34(m,4H),1.50(s,9H).LC-MS m/z[M+1] + :535.3。
EXAMPLE 8 1- (tert-butyl) -N- (2- ((4- (1-methyl-3- (morpholine-4-carbonyl) -1H-indol-5-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of (5-bromo-1-methyl-1H-indol-3-yl) (morpholinyl) methanone
5-bromo-1-methyl-1H-indole-3-carboxylic acid (0.5 g,1.97 mmol) was dissolved in anhydrous dichloromethane (5 mL), cooled to 0deg.C, N-diisopropylethylamine (1.02 g,7.9 mmol) was slowly added dropwise, stirred for 3 min and then transferred to room temperature. Morpholine (0.22 g,2.56 mmol) and 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (HATU, 1.5g,3.94 mmol) were added and reacted for 4 hours. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ]] + :323.0。
Step 2-step 6: the synthesis was identical to steps 2 to 6 of example 2, to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.35(s,1H),8.36(s,1H),8.17(t,J=5.5Hz,1H),7.84-7.73(m,2H),7.60-7.38(m,3H),6.97(s,1H),6.48(s,1H),4.09(d,J=5.6Hz,2H),3.84(s,3H),3.72-3.57(m,8H),1.50(s,9H).LC-MS m/z[M+1] + :549.2。
EXAMPLE 9 1- (tert-butyl) -N- (2- ((4- (1-methyl-3- (morpholinomethyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of 4- ((5-bromo-1H-pyrrolo [2,3-b ] pyridin-3-yl) methyl) morpholine
5-bromo-1H-pyrrole [2,3-b ]]Pyridine-3-carbaldehyde (0.5 g,2.23 mmol) was dissolved in anhydrous dichloromethane (5.0 mL), morpholine (0.25 g,2.9 mmol) was added, cooled to 0deg.C, sodium triacetoxyborohydride (1.42 g,6.69 mmol) was added, and then transferred to room temperature and the reaction stirred for 3.5 hours. After the reaction was completed, water quenching was added under ice bath, extraction was performed with methylene chloride, washing was performed with saturated sodium chloride, drying was performed with anhydrous sodium sulfate, and the organic phase was directly concentrated to obtain the title compound. LC-MS M/z [ M+1 ] ] + :296.0。
Step 2: preparation of 4- ((5-bromo-1-methyl-1H-pyrrolo [2,3-b ] pyridin-3-yl) methyl) morpholine
4- ((5-bromo-1H-pyrrole [2, 3-b)]Pyridin-3-yl) methyl morpholine (200 mg,0.68 mmol) was dissolved in anhydrous N, N-dimethylformamide (2.0 mL), cooled to 0deg.C, sodium hydrogen (60%) (37.94 mg,0.95 mmol) was slowly added and the reaction stirred for 1 hour. Methyl iodide (96.23 mg,0.68 mmol) was then added, followed by transfer to room temperature and stirring for 4 hours. After the reaction was completed, water quenching was added under ice bath, extraction was performed with methylene chloride, washing was performed with saturated sodium chloride, drying was performed with anhydrous sodium sulfate, and the organic phase was directly concentrated to obtain the title compound. LC-MS m-z[M+1] + :310.1。
Step 3-step 7: the synthesis was identical to steps 2 to 6 of example 2, to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.35(s,1H),8.83(s,1H),8.48(d,J=0.5Hz,1H),8.23-8.11(m,1H),7.62(s,1H),7.55-7.49(m,1H),7.46(s,1H),6.97(s,1H),6.48(s,1H),4.10(d,J=5.8Hz,2H),3.81(s,3H),3.68-3.59(m,2H),3.60-3.53(m,4H),2.42-2.36(m,4H),1.50(s,9H).LC-MS m/z[M+1] + :536.2。
EXAMPLE 10N- (2- ((4- (6-benzoyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-3-yl) thiazol-2-yl) amino) -2-oxoethyl) -1- (tert-butyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of (3-bromo-7, 8-dihydro-1, 6-naphthyridin-6 (5H) -yl) (phenyl) methanone
3-bromo-5, 6,7, 8-tetrahydro-1, 6-naphthyridine (0.3 g,1.42 mmol) was dissolved in anhydrous dichloromethane (3 mL), cooled to 0deg.C, benzoic acid (0.26 g,2.12 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 1.1g,2.83 mmol), N, N-diisopropylethylamine (0.55 g,4.25 mmol) was added, followed by transfer to room temperature and stirring reaction continued overnight. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ] ] + :317.0。
Step 2-step 6: the synthesis was identical to steps 2 to 6 of example 2, to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.37(s,1H),8.94(s,1H),8.18(s,1H),7.80-7.63(m,2H),7.58-7.39(m,6H),6.97(s,1H),6.47(s,1H),4.91-4.84(m,2H),4.17-4.02(m,2H),3.71-3.65(m,1H),3.63-3.57(m,1H),3.02-2.93(m,2H),1.49(s,9H).LC-MS m/z[M+1] + :543.2。
EXAMPLE 11 1- (tert-butyl) -N- (2-oxo-2- ((4- (7-oxo-8- ((tetrahydro-2H-pyran-4-yl) methyl) -5,6,7, 8-tetrahydro-1, 8-naphthyridin-3-yl) thiazol-2-yl) amino) ethyl) -1H-pyrrole-3-carboxamide
Step 1: preparation of 6-bromo-1- ((tetrahydro-2H-pyran-4-yl) methyl) -3, 4-dihydro-1, 8-naphthyridin-2 (1H) -one
6-bromo-3, 4-dihydro-1, 8-naphthyridin-2 (1H) -one (260.0 mg,0.12 mmol) was dissolved in anhydrous N, N-dimethylformamide (3 mL), cooled to 0deg.C, 4- (bromomethyl) tetrahydro-2H-pyran (53.4 mg,0.3 mmol) was added, sodium hydrogen (60%) (7.2 mg,0.18 mmol) was slowly added, and the mixture was transferred to room temperature and stirred for 3 hours. After the reaction, water quenching is carried out under ice bath, extraction is carried out by ethyl acetate, saturated sodium chloride is used for washing, anhydrous sodium sulfate is used for drying, and the title compound is obtained through column chromatography. LC-MS M/z [ M+1 ]] + :325.0。
Step 2-step 6: the synthesis was identical to steps 2 to 6 of example 2, to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.34(s,1H),8.75(s,1H),8.20-8.17(m,1H),8.08(s,1H),7.66(s,1H),7.52(s,1H),7.00-6.94(m,1H),6.48(s,1H),4.10(d,J=5.6Hz,2H),4.02(d,J=7.0Hz,2H),3.82-3.79(m,2H),3.22-3.16(m,4H),2.96-2.93(m,2H),2.68-2.65(m,2H),2.05-1.92(m,1H),1.49(s,9H),1.46-1.42(m,2H).LC-MS m/z[M+1] + :551.2。
EXAMPLE 12 1- (tert-butyl) -N- ((S) -1- ((4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrole [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) -1H-pyrrole-3-carboxamide
Step 1: preparation of (2S, 6R) -4- (6-bromopyrrole [2,1-f ] [1,2,4] triazin-4-yl) -2, 6-dimethylmorpholine
6-bromo-4-chloropyrrole [2,1-f][1,2,4]Triazine (1 g,4.3 mmol), (2R, 6S) -2, 6-dimethylmorpholine (0.74 g,6.45 mmol), N-diisopropylethylamine (2.2 g,17.2 mmol) were dissolved in dimethyl sulfoxide (10 mL) and then reacted for 3 hours at 120℃with sealing. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ]] + :311.0。
Step 2: preparation of (2S, 6R) -2, 6-dimethyl-4- (6- (4, 5-tetramethyl-1, 3, 2-dioxabenzaldehyde-2-yl) pyrrole [2,1-f ] [1,2,4] triazin-4-yl) morpholine
(2S, 6R) -4- (6-bromopyrrole [2, 1-f)][1,2,4]Triazin-4-yl) -2, 6-dimethylmorpholine (0.5 g,1.6 mmol), pinacol biborate (2.85 g,8 mmol), 1-bis (diphenylphosphine) dicyclopentadienyl iron palladium dichloride (0.12 g,0.16 mmol), 2-dicyclohexylphosphine-2 ',4',6' -triisopropylbiphenyl (Xphos, 0.15g,0.32 mmol), potassium acetate (0.47 g,4.8 mmol) were dissolved in anhydrous 1, 4-dioxane (5 mL) and reacted at 101 ℃ for 4 hours under nitrogen protection. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ] ] + :359.2。
Step 3: preparation of tert-butyl- ((S) -1- ((4-bromothiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) carbamate
/>
2-amino-4-bromothiazole (1 g,5.62 mmol) was dissolved in anhydrous dichloromethane (10 mL), cooled to 0℃and N, N-diisopropylethylamine (2.9 g,22.48 mmol) was added dropwise, followed by addition of 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 4.3g,11.24 mmol) and N- (tert-butoxycarbonyl) -O-methyl-L-serine (1.48 g,6.74 mmol) followed by stirring overnight at room temperature. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ]] + :380.0。
Step 4: preparation of tert-butyl ((S) -1- ((4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrole [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) carbamate
(2S, 6R) -2, 6-dimethyl-4- (6- (4, 5-tetramethyl-1, 3, 2-dioxabenzaldehyde-2-yl) pyrrole [2, 1-f)][1,2,4]Triazin-4-yl) morpholine (0.45 g,1.7 mmol), tert-butyl- ((S) -1- ((4-bromothiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) carbamate (0.4 g,1.05 mmol), 1-bis (diphenylphosphine) bis-iron palladium dichloride (0.07 g,0.1 mmol) and cesium carbonate (1.02 g,3.15 mmol) were dissolved in a 1, 4-dioxane/water (4 mL/0.8 mL) mixed solvent and reacted at 90℃for 4 hours under nitrogen protection. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. LC-MS M/z [ M+1 ] ] + :532.2。
Step 5: preparation of (S) -2-amino-N- (4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrolo [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) -3-methoxypropionamide
Tert-butyl ((S) -1- ((4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrole [2, 1-f)][1,2,4]Triazin-6-yl) thiazol-2-yl) amino)3-methoxy-1-oxopropan-2-yl) carbamate (280 mg,0.53 mmol) was dissolved in anhydrous dichloromethane (2 mL), cooled to 0deg.C, hydrochloric acid/1, 4-dioxane (4M, 1.5mL,6 mmol) was slowly added dropwise, followed by transfer to room temperature for 2 hours. After the reaction, the supernatant was separated, the solid was retained, and the solid was directly concentrated to give the title compound. LC-MS M/z [ M+1 ]] + :432.1。
Step 6: preparation of 1- (tert-butyl) -N- ((S (-1- ((4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrolo [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) -1H-pyrrole-3-carboxamide
(S) -2-amino-N- (4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrole [2, 1-f)][1,2,4]Triazin-6-yl) thiazol-2-yl) -3-methoxypropionamide (224 mg,0.52 mmol) was dissolved in anhydrous dichloromethane (3 mL), cooled to 0℃and N, N-diisopropylethylamine (218 mg,2.08 mmol) was slowly added dropwise, stirred at this temperature for 10 minutes, followed by the addition of 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 399mg, 1.04 mmol) and 1-tert-butyl-1H-pyrrole-3-carboxylic acid (104 mg,0.63 mmol) for 3 hours. After the reaction, washing with saturated sodium chloride, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and purifying by column chromatography to obtain the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.40(s,1H),8.05(s,1H),7.95-7.92(m,1H),7.91(s,1H),7.64-7.60(m,1H),7.54(s,1H),7.32(s,1H),6.96(s,1H),6.50(s,1H),4.91(dd,J=13.0,6.2Hz,1H),4.66(d,J=12.4Hz,2H),3.73-3.64(m,4H),3.31(s,3H),2.92-2.87(m,2H),1.49(s,9H),1.21(d,J=6.0Hz,6H).LC-MS m/z[M+1] + :581.2。
EXAMPLE 13 1- (tert-butyl) -N- (2- ((4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrolo [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxamide
The procedure of example 12 was followed except that N- (t-butoxycarbonyl) -O-methyl-L-serine was changed to N-t-butoxycarbonyl glycine to obtain the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.30(s,1H),8.16(t,J=5.4Hz,1H),8.03(s,1H),7.91(s,1H),7.58-7.47(m,2H),7.31(s,1H),6.97(s,1H),6.47(s,1H),4.65(d,J=12.5Hz,2H),4.08(d,J=5.6Hz,2H),3.81-3.54(m,2H),3.00-2.74(m,2H),1.49(s,9H),1.21(d,J=6.1Hz,6H).LC-MS m/z[M+1] + :537.2。
EXAMPLE 14N- ((S) -1- ((4- (4- ((2S, 6R) -2, 6-dimethylmorpholinyl) pyrrolo [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) -1- (methylsulfonyl) -1H-pyrrole-3-carboxamide
The procedure was followed in the same manner as in example 12 except for replacing 1-tert-butyl-1H-pyrrole-3-carboxylic acid with 1- (methylsulfonyl) -1H-pyrrole-3-carboxylic acid to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.50(s,1H),8.49(d,J=7.1Hz,1H),8.05(s,1H),7.99(s,1H),7.91(s,1H),7.55(s,1H),7.32(s,1H),7.31-7.28(m,1H),6.79(s,1H),4.97-4.88(m,1H),4.66(d,J=12.8Hz,2H),3.76-3.64(m,4H),3.57(s,3H),2.92-2.87(m,2H),2.46(s,3H),1.21(d,J=6.0Hz,6H).LC-MS m/z[M+1] + :603.1。
Example 15 (S) -1- (tert-butyl) -N- (3-methoxy-1- ((4- (4-morpholinopyrrol [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -1-oxopropan-2-yl) -1H-pyrrole-3-carboxamide
The procedure was followed in accordance with example 12 except that (2R, 6S) -2, 6-dimethylmorpholine was replaced with morpholine to prepare the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.38(s,1H),8.05(s,1H),7.93-7.88(m,2H),7.62(s,1H),7.50(s,1H),7.33(s,1H),7.00-6.93(m,1H),6.50(s,1H),4.96-4.86(m,1H),4.05-3.93(m,3H),3.84-3.76(m,3H),3.72-3.69(m,2H),3.40-3.33(m,5H),1.49(s,9H).LC-MS m/z[M+1] + :553.2。
EXAMPLE 16 (S) -N- (3-methoxy-1-oxo-1- ((4- (4- (pyrrolidin-1-yl) pyrrole [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) propan-2-yl) -1- (methylsulfonyl) -1H-pyrrole-3-carboxamide
The procedure was followed except for replacing 1-tert-butyl-1H-pyrrole-3-carboxylic acid with 1- (methylsulfonyl) -1H-pyrrole-3-carboxylic acid and replacing (2R, 6S) -2, 6-dimethylmorpholine with tetrahydropyrrole in example 12 to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.49(s,1H),8.49(d,J=7.1Hz,1H),8.01-7.97(m,2H),7.81(s,1H),7.48(s,1H),7.32-7.28(m,1H),7.25(s,1H),6.80(s,1H),4.96-4.87(m,1H),4.03-3.96(m,2H),3.77-3.62(m,4H),3.57(s,3H),3.31(s,3H),2.15-1.85(m,4H).LC-MS m/z[M+1] + :559.1。
EXAMPLE 17 (S) -1- (tert-butyl) -N- (3-methoxy-1- ((4- (4- (3-methoxyazetidin-1-yl) pyrrole [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -1-oxopropan-2-yl) -1H-pyrrole-3-carboxamide
The procedure was followed in accordance with example 12, except that (2R, 6S) -2, 6-dimethylmorpholine was replaced with 3-methoxy-azetidine, thereby obtaining the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.39(s,1H),8.00(s,1H),7.91(d,J=7.4Hz,1H),7.84(s,1H),7.64-7.59(m,1H),7.46(s,1H),7.04(s,1H),6.99-6.94(m,1H),6.50(s,1H),4.94-4.89(m,1H),4.47-4.38(m,3H),4.10-4.03(m,3H),3.72-3.68(m,2H),3.62-3.58(m,2H),3.31(s,3H),1.49(s,9H).LC-MS m/z[M+1] + :553.2。
EXAMPLE 18 (S) -N- (1- ((4- (4- (3, 3-difluoroazetidin-1-yl) pyrrole [2,1-f ] [1,2,4] triazin-6-yl) thiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) -1- (methylsulfonyl) -1H-pyrrole-3-carboxamide
The procedure was followed in accordance with example 12 except that (2R, 6S) -2, 6-dimethylmorpholine was replaced with 3, 3-difluoroazetidine and 1-tert-butyl-1H-pyrrole-3-carboxylic acid was replaced with 1- (methylsulfonyl) -1H-pyrrole-3-carboxylic acid, to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.53(s,1H),8.49(d,J=7.1Hz,1H),8.10(s,1H),8.01-7.97(m,1H),7.96(s,1H),7.47(s,1H),7.32-7.26(m,1H),7.07(s,1H),6.80(s,1H),5.00-4.80(m,4H),3.76-3.68(m,2H),3.63-3.59(m,1H),3.57(s,3H),3.28(s,3H).LC-MS m/z[M+1] + :581.1。
EXAMPLE 19N- ((S) -1- ((4- (1- ((2S, 6R) -2, 6-dimethylmorpholino) pyrrolo [1,2-a ] pyrazin-7-yl) thiazol-2-yl) amino) -3-methoxy-1-oxopropan-2-yl) -1- (methylsulfonyl) -1H-pyrrole-3-carboxamide
The synthesis was identical to that of example 12, except that 6-bromo-4-chloropyrrole [2,1-f][1,2,4]Triazine replacement with 7-bromo-1-chloropyrrolo [1,2-A]Pyrazine, 1-tert-butyl-1H-pyrrole-3-carboxylic acid was replaced with 1- (methylsulfonyl) -1H-pyrrole-3-carboxylic acid to give the title compound. 1 H NMR(400MHz,DMSO-d 6 )δ12.45(s,1H),8.48(d,J=7.2Hz,1H),7.99(s,1H),7.93(s,1H),7.83(d,J=4.2Hz,1H),7.51(s,1H),7.30(d,J=2.2Hz,1H),7.21-7.14(m,1H),7.12(d,J=4.7Hz,1H),6.79(s,1H),4.93(m,1H),4.19(d,J=12.7Hz,2H),3.81-3.65(m,4H),3.56(s,3H),3.32(s,3H),2.77-2.61(m,2H),1.17(d,J=6.1Hz,6H).LC-MS m/z[M+1] + :602.2。
Experimental example 1: evaluation of SMARCA2/SMARCA4 enzyme Activity
1. Experimental materials
1.1 reagent: SMARCA2 Chromatin Remodeling Enzyme (Human BRM) (EpiCypher, 15-1015); SMARCA4Chromatin Remodeling Enzyme (Human BRG 1) (EpiCypher, 15-1014); heLa Mononucleosomes, purified (EpiCypher, 16-0002); ADP-Glo reagent (Promega, V9102).
1.2 preparation of solutions of Compounds
All compounds were formulated in DMSO as 10mM stock solutions. Subsequently, 10 concentration gradients were diluted 4-fold with DMSO from the highest initial concentration of 10. Mu.M and centrifuged at 1000rpm for 1min for subsequent experiments.
2. Experimental method
200nL of DMSO or test compound solution was added to 384 well assay plates by Echo followed by 10. Mu.L of 2 XSMARCA 2 or SMARCA4 enzyme solution to the wells and incubated for 30min at room temperature. After the incubation was completed, 10 μl of 2 x substrate mixture was added to the wells to initiate the reaction, and the solution was concentrated at the bottom of the wells by centrifugation. The cover was incubated at room temperature for 60min.
During the reaction, 5. Mu.L of the reaction solution was transferred to another 384-well plate. At 60min incubation, 5. Mu.L of ADP-Glo reagent was added to the experimental mixture, rapidly centrifuged at room temperature and incubated for 150min. After the incubation was completed, 10 μl of kinase assay reagent was added, centrifuged rapidly and incubated for a further 60min at room temperature. Fluorescence values were read by EnVision plate reader.
Calculation of IC using GraphPad Prism software 50 And dose response curves for the compounds were plotted. The inhibition rate calculation formula is as follows:
Inhibition(%)=(1-(Cmpd-DMSO no enzyme )/(DMSO enzyme -DMSO no enzyme ) X 100). IC of partial Compound 50 The results are shown in Table 1. The compounds of the invention exhibit significant selective inhibition activity against SMARCA2 enzymes.
List one
"-" indicates no measurement.
From the above experimental results, it can be seen that the compounds of the present invention have a good inhibitory effect on the atpase activity of SMARCA 2.
Experimental example 2 evaluation of in vitro cell Activity of Compounds
1. Experimental materials
Experimental cells: human non-small cell lung cancer cell NCI-H1944 was purchased from Nanjac, bai Biotech Co.
Test compounds: the compounds of the invention prepared in the above examples were each prepared in 20mM stock solution in DMSO and finally diluted to 9 concentrations for detection, and the final concentrations of the compounds for NCI-H1944 cell experiments were 20000.00nM, 5000.00nM, 1250.00nM, 312.50nM, 78.13nM, 19.53nM, 4.88nM, 1.22nM and 0.31nM.
2. Experimental method
2.1 cell resuscitation:
NCI-H1944 cell resuscitation: the NCI-H1944 cell cryopreservation tube was removed from the liquid nitrogen tank and placed in a 37℃water bath with gentle shaking to thaw it as soon as possible. Thawing, taking out the frozen storage tube, sterilizing with alcohol cotton ball, unscrewing the cover, sucking out the cell sap, injecting into the centrifuge tube, adding 1mL of RPMI-1640 culture medium containing 10% FBS, mixing, placing into a centrifuge, centrifuging at 1000rpm for 5min. And then the supernatant is discarded, and the complete culture medium is added for repeated blowing until the cells are completely blown away and resuspended. The culture dish was inoculated at a suitable concentration. Placing at 37deg.C, 5% CO 2 Is cultured in an incubator of (a).
2.2 cell culture and passaging:
NCI-H1944 cell culture and passaging: cells were grown to about 80-90%, and medium (RPMI-1640 Medium+10% FBS+1% Streptomyces lividans) was transferred to a 15mL centrifuge tube, 1000rpm, and centrifuged for 5min. Removing supernatant, re-suspending cells with complete medium, inoculating into culture dish at desired density, placing at 37deg.C, 5% CO 2 Is replaced or passaged every 2-3 days depending on the cell growth.
2.3 experimental procedure:
day 1 of the experiment:
NCI-H1944 cells were resuspended in complete medium at corresponding densities after passaging, 1X10 3 Number/well, inoculated into 96-well culture plates: the wells were filled with 200. Mu.L PBS to prevent faster evaporation of the edge mediumResulting in excessive differences in culture conditions in the interior plate wells; the leftmost column of the 60 holes in the inner part is blank hole, no cells are added, and the blank hole is filled with PBS with equal volume; the remaining 54 wells were cell plated with a row gun, 100. Mu.L of each well, and 5% CO was added 2 Is cultured in an incubator at 37 ℃ for 24 hours.
Day 2 of the experiment:
NCI-H1944 cells were added with 100. Mu.L of (2X) drug based on the original medium (100. Mu.L), two wells were set for each concentration group, and 5% CO was further added 2 The incubator was cultured for 7 days. The compound solutions were formulated as follows: 1-2mg of the compound was weighed in advance and prepared into a 20mM mother liquor using DMSO. The drug was diluted with complete medium, the final drug concentration was up to 40000nM starting maximum concentration, diluted sequentially to 9 concentration gradients in a 1:3 gradient: 10000.00nM, 2500.00nM, 625.00nM, 156.25nM, 39.06nM, 9.77nM, 2.44nM, 0.61nM and 0.15nM.
Day 8 of the experiment
100 mu L of culture medium is sucked and removed from each well of a 96-well plate, then 100 mu L (1X) of medicine is added, sufficient nutrition of cells is ensured, and 5% CO is continuously added 2 Culturing in an incubator for 7 days.
Day 15 of the experiment:
after 14 days of NCI-H1944 cell treatment, cellTiter-Glo Luminescent Cell Viabillity Assay was removed 30min earlier and equilibrated to room temperature. The medium in the wells was aspirated, and Celltiter-Glo reagent was diluted 1:1 (equal volume) with complete medium, and 150. Mu.L of diluted Celltiter-Glo reagent was added to each well and shaken at room temperature for 2min. After incubation for 10min at room temperature, 100. Mu.L of detection chemiluminescent signal is taken, and the detection condition of Read sample injection is 500ms. The inhibition ratio of each well relative to the solvent control well was calculated from the a.u. values derived by the microplate reader: inhibition (%) = 100- (a.u. experimental well-a.u. blank well)/(a.u. solvent control well-a.u. blank well) ×100. According to different drug concentrations and the corresponding inhibition ratios, graghPad 5.0 software is used for fitting a four-parameter logic S-shaped curve, and data are analyzed to obtain the IC 50 Values, experimental results are shown in table 2.
TABLE 2
"NA means inactive"
From the above experimental results, it can be seen that the compounds of the present invention exhibit good inhibitory activity against proliferation of NCI-H1944 non-small cell lung cancer cells (SMARCA 4 deletion mutation), and are highly promising as therapeutic agents for SMARCA4 mutated non-small cell lung cancer.
Experimental example 3 evaluation of mouse PK
1. Experimental animal
SPF-class male BALB/c mice were kept in the experimental environment for 2-3 days after the animals reached.
2. Experimental method
2.1 animal preparation
The experimental animals were randomly divided into a single oral administration (PO) group and a single intravenous administration (IV) group (n=3) 1 day before the start of the experiment, and the experimental animals were orally administered, fasted for 24 hours.
2.2 pharmaceutical formulation
The PO group compound solvent is 10% Solutol+5% DMSO+85% physiological saline, and the drug solution is prepared according to the administration dosage of 10mg/kg and the administration volume of 0.2mL/10g (mouse body weight); group IV compounds were formulated into drug solutions at a dose of 2% Solutol+1% DMSO+97% physiological saline, 0.1mL/10g (mouse body weight). The composition of the transparent solution was formulated in accordance with a drug administration volume of 1mg/kg,0.1mL/10 g. When in preparation, the organic solvents are added one by one, and the mixture is uniformly mixed or sonicated until the compound is dissolved, and the corresponding physiological saline is replenished after the compound is dissolved.
2.3 administration and blood sampling
The PO group is administrated by the method of gastric administration, and the IV group is administrated by the method of tail vein injection. Orbital blood collection was performed with capillaries at 0.083h, 0.25h, 0.5h, 1h, 2h, 6h, 10h, and 24h sampling points after dosing. The amount of blood collected from each mouse was about 40 μl per time point, and then the capillary was transferred into heparin sodium anticoagulation tube and placed on ice. After the experiment was completed, the animals were sacrificed by cervical spine removal and cadavers were frozen in a-20 ℃ refrigerator. The collected blood sample was centrifuged at 8000rpm for 5min at 4℃and plasma was separated after centrifugation. Transfer 15 μl of plasma to shallow 96 well plates and freeze store in-80 ℃ refrigerator.
2.4 biological sample analysis
2.4.1 development of methods for analysis of Compounds
The development of methods for compound analysis should be performed the day before or the day of initiation of administration. Standard compounds were first dissolved in DMSO to 5mg/mL and then gradually diluted with methanol to 100ng/mL for development of LC-MS/MS assay methods. After the determination, plasma test samples with the concentrations of 10, 20 and 50ng/mL are respectively prepared for sample injection detection, whether the concentration of the compound and the peak area are in direct proportion is determined, in addition, the dilution of the sample is adjusted according to the response of the test sample, and the peak area of the lower limit of quantification is preferably 500-1500 and cannot be higher than 3000 during formal sample injection.
2.4.2 analysis of samples
mu.L of the test solution was taken and gradually diluted to 10ng/mL with methanol. LC-MS/MS analysis was performed on 10ng/mL of standard solution and test sample. Test article accuracy (%) =test article peak area/working fluid peak area X100.
2.4.3 biological sample treatment
Thawing 15 μl of plasma before sampling, adding 150 μl of precipitant, shaking by vortex, centrifuging at 4500rpm for 5min, diluting the supernatant (dilution multiple is the same as that of blank matrix and standard yeast) according to method development result, shaking by vortex after dilution, centrifuging at 4500rpm for 5min, and sampling 200 μl of supernatant.
2.4.4 standard Curve preparation
Blank matrix preparation: blank plasma 50. Mu.L, adding 500. Mu.L of precipitant, vortex shaking, centrifuging at 15000rpm for 5min, and collecting supernatant for dilution with dilution factor identical to that of standard curve and sample.
Preparing standard yeast: blank plasma 45. Mu.L, 5. Mu.L standard solution (50. Mu.g/mL) and 500. Mu.L precipitant were added, vortexed and centrifuged at 15000rpm for 5min, and the supernatant was diluted (dilution factor was the same as that of blank matrix, sample) to give the maximum concentration of standard curve. The standard curve is diluted into standard curve by using blank matrix according to the proportion of 5000, 2000, 1000, 500, 200, 100, 50, 20, 10 and 5ng/mL, 200 mu L of supernatant is sampled.
2.4.5 data analysis
The pharmacokinetic parameters were calculated by applying Phoenix WinNonlin non-compartmental model to the blood concentration data at different time points, providing the appropriate parameters. The experimental results are shown in Table 3.
TABLE 3 Table 3
From the above results, it can be seen that the compound of example 12 of the present invention has higher Cmax and exposure to T in the plasma after oral administration 1/2 And Tmax are good, exhibiting good pharmacokinetic properties. At the same time, example 13 also shows acceptable pharmacokinetic properties, both compounds having good pharmaceutical potential.
Although the invention has been described in detail hereinabove, those skilled in the art will appreciate that various modifications and changes can be made thereto without departing from the spirit and scope of the invention. The scope of the invention is not limited by the detailed description set forth above, but rather is to be attributed to the claims.

Claims (10)

1. A compound of formula (I) or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
wherein,,
R 1 selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkyl Acylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkylsulfonyl, aminosulfonyl and alkylaminosulfonyl;
R 2 selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, alkoxyalkyl;
a is selected from the group consisting of six-membered and five-membered heteroaromatic rings, six-membered and six-membered heteroaromatic rings, and five-membered and six-membered heteroaromatic rings, optionally substituted with one or more groups selected from halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylamido, alkanoyl, alkylsulfonyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, haloalkylacyl, hydroxyalkylacyl, cycloalkylacyl, heterocyclylacyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo groups;
l is a bond, -S-, -O-, -N-, alkylene, -C (O) -, -S (O) -or-S (O) 2 -; and
b is selected from the group consisting of alkyl, aryl, heteroaryl, cycloalkyl and heterocyclyl, optionally substituted with one or more groups selected from the group consisting of halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylamido, alkanoyl, alkylsulfonyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, haloalkylacyl, hydroxyalkylacyl, cycloalkylacyl, heterocyclylacyl, cycloalkyl, heterocyclyl, aryl, heteroaryl and oxo groups.
2. The compound according to claim 1, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 1 Selected from hydrogen, halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxyRadical C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkanoyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, C 1-6 Alkylsulfonyl, aminosulfonyl and C 1-6 Alkyl sulfamoyl.
3. The compound according to claim 1 or 2, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 2 Selected from hydrogen, halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, C 1-6 Alkoxy C 1-6 An alkyl group.
4. A compound according to any one of claims 1-3, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein a is selected from Optionally substituted with one or more groups selected from halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkyl acyl, C 1-6 Alkylsulfonyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, alkenyl, alkynyl, halo C 1-6 Alkyl acyl, hydroxy C 1-6 Group substitution of alkyl acyl and oxo groups.
5. The compound according to any one of claims 1-4, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein L is a bond, -S-, -O-, -N-, sub-C 1-6 Alkyl, -C (O) -, -S (O) -or-S (O) 2 -。
6. The compound according to any one of claims 1-5, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein B is selected from C 1-6 Alkyl, aryl, heteroaryl, C 3-8 Cycloalkyl and C 3-8 Heterocyclyl, optionally substituted with one or more substituents selected from halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkyl acyl, C 1-6 Alkylsulfonyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, halo C 1-6 Alkyl acyl, hydroxy C 1-6 Group substitution of alkyl acyl and oxo groups.
7. The compound according to claim 1, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein the general formula (I) has the structure of the following general formula (IIa),
wherein,,
R 3 selected from hydrogen, halogen, hydroxy, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, nitro, carboxyl, cyano, amino, mono C 1-6 Alkylamino, C 1-6 Alkylacylamino, C 1-6 Alkyl groupAcyl, aminoacyl, C 1-6 Alkylaminoacyl, bis C 1-6 Alkylamino, C 1-6 Alkylsulfonyl, aminosulfonyl and C 1-6 An alkylamino sulfonyl group;
n is selected from 1, 2, 3 or 4; and
R 1 and R is 2 Having the definition in the general formula (I) as claimed in claims 1 to 3.
8. The compound of claim 1, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein the compound is a compound selected from the group consisting of:
9. a pharmaceutical composition comprising a compound of any one of claims 1-8, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
10. Use of a compound according to any one of claims 1 to 8, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, or a pharmaceutical composition according to claim 9 in the manufacture of a medicament for the treatment of cancer.
CN202210005363.9A 2022-01-05 2022-01-05 Compounds as SMARCA2 inhibitors and uses thereof Pending CN116444523A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210005363.9A CN116444523A (en) 2022-01-05 2022-01-05 Compounds as SMARCA2 inhibitors and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210005363.9A CN116444523A (en) 2022-01-05 2022-01-05 Compounds as SMARCA2 inhibitors and uses thereof

Publications (1)

Publication Number Publication Date
CN116444523A true CN116444523A (en) 2023-07-18

Family

ID=87120666

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210005363.9A Pending CN116444523A (en) 2022-01-05 2022-01-05 Compounds as SMARCA2 inhibitors and uses thereof

Country Status (1)

Country Link
CN (1) CN116444523A (en)

Similar Documents

Publication Publication Date Title
EP4328225A1 (en) Heterocyclic derivative inhibitor and preparation method therefor and application thereof
EP3841102B1 (en) Tetrahydropyridopyrimidine derivatives as ahr modulators
US20230065740A1 (en) Cyclin-dependent kinase inhibitors
CN113527335A (en) Macrocyclic compound as EGFR inhibitor and application thereof
JP2019520410A (en) Compound as isocitrate dehydrogenase inhibitor and application thereof
EP3878852A1 (en) Substituted pyrazolo[1,5-a]pyridine compound, composition containing the same and use thereof
CN114656482A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
JP5820080B2 (en) Tricyclic PI3K and / or mTOR inhibitors
CN116535401A (en) Novel PARP1 inhibitors and uses thereof
BR112021002979A2 (en) keap1-nrf2 protein-protein interaction inhibitors
US11897871B1 (en) Methods for treating cancer
US20230339902A1 (en) Tricyclic ligands for degradation of ikzf2 or ikzf4
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
JP2024508868A (en) Terpyridine diketone compounds, or salts thereof, and methods of preparation and use thereof
CN111592541B (en) Macrocyclic kinase inhibitors and uses thereof
KR20220085735A (en) Isoxazolidine derivative compounds, and uses thereof
CN114644627A (en) AhR inhibitor and application thereof
CN115768769A (en) Tetrahydroisoquinoline compound and application thereof
CN114369097B (en) Heteroaromatic AhR inhibitors
CN116444523A (en) Compounds as SMARCA2 inhibitors and uses thereof
KR20230104782A (en) 7-Azaindole compounds for inhibition of BCR-ABL tyrosine kinase
CN115433207A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
KR20200078510A (en) Compounds having ERK kinase inhibitory activity and uses thereof
CN103596953A (en) Pyridonaphthyridine PI3K/mTOR dual inhibitors and preparation and use thereof
CN104230960B (en) Four-ring anaplastic lymphoma kinase inhibitor

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication