CN112920199B - Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition - Google Patents

Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition Download PDF

Info

Publication number
CN112920199B
CN112920199B CN202010492337.4A CN202010492337A CN112920199B CN 112920199 B CN112920199 B CN 112920199B CN 202010492337 A CN202010492337 A CN 202010492337A CN 112920199 B CN112920199 B CN 112920199B
Authority
CN
China
Prior art keywords
alkyl
methyl
mmol
cycloalkyl
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202010492337.4A
Other languages
Chinese (zh)
Other versions
CN112920199A (en
Inventor
王宁宇
余洛汀
左玮琼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan University
Original Assignee
Sichuan 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
Application filed by Sichuan University filed Critical Sichuan University
Priority to CN202010492337.4A priority Critical patent/CN112920199B/en
Priority to PCT/CN2021/095581 priority patent/WO2021244346A1/en
Publication of CN112920199A publication Critical patent/CN112920199A/en
Application granted granted Critical
Publication of CN112920199B publication Critical patent/CN112920199B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General 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)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A piperazinone substituent or a derivative thereof, a preparation method and application thereof, and a pharmaceutical composition belong to the technical field of antitumor drugs. The application designs a piperazinone substituted heterocyclic small molecule compound, which has good affinity to PI3K delta, can down-regulate the activity of a PI3K pathway in a tumor cell, has good inhibitory activity to PI3K delta-dependent tumors, and is expected to be used as one of components of a pharmaceutical preparation for treating different tumors. The preparation method of the piperazinone substituent is simple and convenient, and the yield is high.

Description

Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition
Technical Field
The application relates to the technical field of antitumor drugs, in particular to a piperazinone substituent or a derivative thereof, a preparation method and application thereof, and a pharmaceutical composition.
Background
Phosphatidylinositol 3-kinases (PI 3 Ks) are lipid kinases, major downstream effector proteins of cell surface Receptor Tyrosine Kinases (RTKs) and G-protein coupled receptors (GPCRs), which upon receiving upstream signals, catalyze phosphorylation of phosphatidylinositol 4, 5-bisphosphates (PI (4, 5) P2) to phosphatidylinositol 3,4, 5-triphosphates (phosphoinositides 3,4, 5-triphosphates, PI (3, 4, 5) P3), which serves as a second messenger by linking to downstream effector proteins including various serine/threonine and tyrosine kinases, adaptor proteins (GTPase-activator proteins), and GTPase-activator proteins (GAPs), etc., to induce the downstream effector proteins to activate the functions of cell proliferation, and regulate cell metabolism. Since PI3Ks play a very critical role in multiple links of cell life activities, mutation, abnormal expression or activation of the protein are often related to the occurrence and development of different diseases, and small molecule inhibitors developed aiming at the pathway are promising drugs for a plurality of cancers and inflammatory diseases.
PI3ks can be generally classified into 3 major groups according to differences in structure and substrate specificity: (1) the first class of PI3K family comprises PI3K alpha, PI3K beta, PI3K gamma and PI3K delta, and the four proteins are heterodimers consisting of a regulatory subunit and a catalytic subunit; (2) the second class of PI3K family mainly comprises PI3K-C2 alpha, PI3K-C2 beta and PI3K-C2 gamma, the class of proteins only have catalytic subunits and do not have regulatory subunits, and the combination of the proteins and the lipids is independent of calcium ions; (3) the third class of PI3K family, structurally similar to the first class of PI3Ks, is also a heterodimer composed of a catalytic subunit and a regulatory subunit, but the protein only catalyzes the conversion of PI to PI (3) P, and its primary function is related to the transport of intracellular proteins and vesicles.
PI3K δ is an important member of the first PI3K family, consisting of the catalytic subunit p110 δ and the regulatory subunit p85, which are distributed mainly in the blood system. Different from other three subtypes of the first PI3K family, the upstream protein of the protein is mainly proteins related to immune cell development and functions, such as BCR/TCR, CD28, CD19, TLRs, BCAP and the like. Therefore, the protein is mainly involved in the development and functions of B, T and NK cells. Protein loss does not cause embryonic lethality, but impairs immune system function, and abnormal activation or amplification of proteins is associated with the development of hematologic malignancies, allergies, chronic inflammation and autoimmune diseases. The inhibitor aiming at the protein can be used for treating the tumor of the blood system and the autoimmune disease.
Disclosure of Invention
The application provides a piperazinone substituent or a derivative thereof, a preparation method and application thereof, and a pharmaceutical composition, wherein the piperazinone substituent or the derivative thereof can reduce the activity of a PI3K pathway in tumor cells, so that the piperazinone substituent or the derivative thereof can show good inhibitory activity on PI3K delta-dependent tumors.
The embodiment of the application is realized as follows:
in a first aspect, the present application illustratively provides a piperazinone substituent or derivative thereof, the piperazinone substituent having the formula:
Figure BDA0002520468220000021
wherein X comprises N or S;
y comprises NR 3 Or CR 4
R 1 Included
Figure BDA0002520468220000031
R 2 Including heteroaryl, wherein heteroaryl is substituted with any one or more of the following substituents: halogen, cyano, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 7 R 8
R 3 Comprises H and C 1-6 Alkyl or C 3-6 A cycloalkyl group;
R 4 including H, halo, cyano, methyl, monofluoromethyl, difluoromethyl, trifluoromethyl, ethyl, n-propyl, isopropyl, or cyclopropyl;
R 5 comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, saturated ring containing 1 heteroatom, spirocycloalkyl containing 0 to 3 heteroatoms, cyclic ketone, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -C (O) aryl, -C (O) heteroaryl, -SO 2 C 1-6 Alkyl, -SO 2 C 3-6 Cycloalkyl, -SO 2 Aryl or-SO 2 Heteroaryl of which C 1-6 Alkyl is substituted with any one or more of the following substituents: hydrogen, halogen, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 9 R 10 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 11 R 12 ,C 3-6 The cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, oxygen, halogen and-NR 13 R 14 and the heteroatoms of the saturated ring containing 1 heteroatom are substituted by any one or more of the following substituents: hydrogen, methyl and oxygen;
R 6 comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, spirocycloalkyl containing 0 to 3 heteroatoms, C 4-6 Cyclic ketones, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl or heteroaryl, wherein C 1-6 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 15 R 16 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 17 R 18
R 7 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 8 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 7 、R 8 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 9 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 10 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 11 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 12 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 11 、R 12 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 13 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 14 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 13 、R 14 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 15 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 16 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 15 、R 16 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 17 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 18 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 17 、R 18 And N atom to form a 4-to 7-membered saturated ring containing 1 or 2 heteroatoms
Derivatives include solvates of the piperazinone substituent, pharmaceutically acceptable salts of the piperazinone substituent, or solvates of the pharmaceutically acceptable salts of the piperazinone substituent.
In the technical scheme, the piperazinone substituted heterocyclic small molecule compound is designed, the compound shows good affinity to PI3K delta, and the compound can reduce the activity of a PI3K pathway in tumor cells, so that the compound shows good inhibitory activity to PI3K delta-dependent tumors, and is expected to be used as one of components of a pharmaceutical preparation for treating different tumors.
In a third aspect, the present application illustrates a method for preparing a piperazinone substituent comprising reacting a piperazinone with R 1 Substituted 2-chloro-4-morpholinyl thiophenes [3,2-d]-OH in pyrimidine-6-methanol as R 2 Substituted 2-chloro-4-morpholinyl thiophenes [3,2-d]-Cl in pyrimidine-6-methanol;
wherein R is 1 Included
Figure BDA0002520468220000041
R 2 Including heteroaryl, wherein heteroaryl is substituted with any one or more of the following substituents: halogen, cyano, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 7 R 8
R 5 Comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, saturated ring containing 1 heteroatom, spirocycloalkyl containing 0 to 3 heteroatoms, cyclic ketone, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -C (O) aryl, -C (O) heteroaryl, -SO 2 C 1-6 Alkyl, -SO 2 C 3-6 Cycloalkyl, -SO 2 Aryl or-SO 2 Heteroaryl of which C is 1-6 Alkyl is substituted with any one or more of the following substituents: hydrogen, halogen, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 9 R 10 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 11 R 12 ,C 3-6 The cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, oxygen, halogen and-NR 13 R 14 And the heteroatoms of the saturated ring containing 1 heteroatom are substituted by any one or more of the following substituents: hydrogen, methyl and oxygen;
R 6 comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, spirocycloalkyl containing 0 to 3 heteroatoms, C 4-6 Cyclic ketones, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl or heteroaryl, wherein C 1-6 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 15 R 16 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 17 R 18
R 7 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 8 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 7 、R 8 And N atoms forming a ring containing 1 or 2 hetero atomsA saturated ring of (a);
R 9 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 10 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 11 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 12 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 11 、R 12 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 13 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 14 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 13 、R 14 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 15 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 16 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 15 、R 16 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 17 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 18 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 17 、R 18 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
alternatively, 2-chloro-4-morpholinylthiophene [3,2-d]the-OH group in pyrimidine-6-methanol needs to be substituted first with-Cl to give 4- (2-chloro-6- (chloromethyl) thiophene [3,2-d]Pyrimidine-4-yl) morpholine, with R 1 substituted-Cl;
alternatively, 2-chloro-4-morpholinothieno [3,2-d ] pyrimidine-6-methanol is prepared by the following method:
reacting a mixture containing 2-chloro-4-morpholinyl thiophene [3,2-d ] pyrimidine-6-formaldehyde, methanol and sodium borohydride at-5 ℃ to-15 ℃ for 0.5-3h;
alternatively, 2-chloro-4-morpholinothieno [3,2-d ] pyrimidine-6-carbaldehyde is prepared by the following method:
reacting a mixture comprising 4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine, tetrahydrofuran and n-butyllithium at-78 ℃ for 1h;
alternatively, 4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine is prepared by:
reacting a mixture containing 2, 4-dichlorothieno [3,2-d ] pyrimidine, methanol and morpholine at 20-22 ℃ for 30-40min.
In the technical scheme, the preparation method of the piperazinone substituent is simple and convenient, and the yield is high.
In a third aspect, the present application illustrates a method for preparing a piperazinone substituent comprising reacting a piperazinone with R 1 By substituting-OH in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol with R 2 substituted-Cl in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol;
wherein R is 1 Included
Figure BDA0002520468220000061
R 2 Including heteroaryl, wherein heteroaryl is substituted with any one or more of the following substituents: halogen, cyano, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 7 R 8
R 5 Comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, saturated ring containing 1 heteroatom, spirocycloalkyl containing 0 to 3 heteroatoms, cyclic ketone, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -C (O) aryl, -C (O) heteroaryl, -SO 2 C 1-6 Alkyl, -SO 2 C 3-6 Cycloalkyl, -SO 2 Aryl or-SO 2 Heteroaryl of which C is 1-6 Alkyl is substituted with any one or more of the following substituents: hydrogen, halogen, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 9 R 10 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 11 R 12 ,C 3-6 Cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, oxygen, halogen and-NR 13 R 14 And the heteroatom of the saturated ring containing 1 heteroatom is substituted by any one or more of the following substituents: hydrogen, methyl and oxygen;
R 6 comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, spirocycloalkyl containing 0 to 3 heteroatoms, C 4-6 Cyclic ketones, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl or heteroaryl, wherein C 1-6 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 15 R 16 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 17 R 18
R 7 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 8 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 7 、R 8 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 9 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 10 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 11 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 12 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 11 、R 12 And N atomForming a saturated ring containing 1 or 2 heteroatoms;
R 13 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 14 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 13 、R 14 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 15 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 16 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 15 、R 16 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 17 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 18 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 17 、R 18 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
alternatively, the-OH group of 2-chloro-9-methyl-6-morpholinyl-9H-purin-8-methanol needs to be substituted first with-Cl to give 4- (2-chloro-8- (chloromethyl) -9-methyl-9H-purin-6-yl) morpholine, followed by R 1 substituted-Cl;
alternatively, 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol is prepared by:
reacting a mixture containing 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-formaldehyde, methanol and sodium borohydride at the temperature of between 5 ℃ below zero and 15 ℃ below zero for 0.5 to 3 hours;
alternatively, 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-carbaldehyde was prepared by:
reacting a mixture comprising 4- (2-chloro-9H-purin-6-yl) morpholine, tetrahydrofuran and n-butyllithium at-78 ℃ for 1H;
alternatively, 4- (2-chloro-9H-purin-6-yl) morpholine is prepared by:
reacting a mixture containing 2, 6-dichloropurine, methanol and morpholine at 20-22 ℃ for 30-40min.
In the technical scheme, the preparation method of the piperazinone substituent is simple and convenient, and the yield is high.
In a fourth aspect, the present application provides, as an example, a pharmaceutical composition comprising a piperazinone substituent or derivative thereof as described above.
Alternatively, the pharmaceutical composition comprises a pharmaceutically acceptable carrier for the piperazinone substituent or a pharmaceutically acceptable carrier for the derivative of the piperazinone substituent.
In the technical scheme, the pharmaceutical composition prepared by using the piperazinone substituent or the derivative thereof as a raw material can be used for treating tumors.
In a fifth aspect, the present application provides an application of the piperazinone substituent or the derivative thereof in the preparation of a medicament for treating an autoimmune disease or cancer responsive to inhibition of PI3K δ.
Drawings
In order to more clearly illustrate the technical solutions of the embodiments of the present application, the drawings that are required to be used in the embodiments will be briefly described below, it should be understood that the following drawings only illustrate some embodiments of the present application and therefore should not be considered as limiting the scope, and for those skilled in the art, other related drawings can be obtained from the drawings without inventive effort.
FIG. 1 is a graph showing the inhibition of proliferation of SU-DHL-6 in example 67 of the present application;
FIG. 2 is a graph showing the growth inhibition of SU-DHL-4 in example 67 of the present application;
FIG. 3 is a graph showing the inhibition of proliferation of JEKO-1 in example 67 of the present application;
FIG. 4 is a graph showing the growth inhibition of NAMALVA in example 67 of the present application;
FIG. 5 is a graph of the in vivo anti-tumor profile of example 67 of the present application against a SU-DHL-6 xenograft tumor model;
FIG. 6 is a graph of the in vivo antitumor effect of example 67 on a JEKO-1 xenograft tumor model of the present application;
FIG. 7 is a graph of tumor weight distribution in the SU-DHL-6 xenograft tumor model following treatment in example 67 of the present application;
FIG. 8 is a tumor weight distribution map of a JEKO-1 xenograft tumor model after the action of example 67 of the present application;
FIG. 9 is a graph showing the body weight change of mice in the SU-DHL-6 xenograft tumor model after the treatment in example 67 of the present application;
FIG. 10 is a graph showing the body weight change of mice in the JEKO-1 xenograft tumor model after the action in example 67 of the present application.
Detailed Description
Embodiments of the present application will be described in detail below with reference to examples, but those skilled in the art will appreciate that the following examples are only illustrative of the present application and should not be construed as limiting the scope of the present application. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are conventional products which are not indicated by manufacturers and are commercially available.
PI3K delta is a promising drug target with good druggability, and the problems of safety, selectivity and metabolic stability of the existing drugs are still to be further optimized.
The piperazinone substituent or the derivative thereof, the preparation method, the application and the pharmaceutical composition thereof according to the embodiments of the present application are specifically described as follows:
the application provides a piperazinone substituent or a derivative thereof, wherein the chemical formula of the piperazinone substituent is as follows:
Figure BDA0002520468220000101
wherein X comprises N or S;
y comprises NR 3 Or CR 4
R 1 Included
Figure BDA0002520468220000102
R 2 Including heteroaryl, wherein heteroaryl is substituted with any one or more of the following substituents: halogen, cyano, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 7 R 8
R 3 Comprises H and C 1-6 Alkyl or C 3-6 A cycloalkyl group;
R 4 including H, halo, cyano, methyl, monofluoromethyl, difluoromethyl, trifluoromethyl, ethyl, n-propyl, isopropyl, or cyclopropyl;
R 5 comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, saturated ring containing 1 heteroatom, spirocycloalkyl containing 0 to 3 heteroatoms, cyclic ketone, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -C (O) aryl, -C (O) heteroaryl, -SO 2 C 1-6 Alkyl, -SO 2 C 3-6 Cycloalkyl, -SO 2 Aryl or-SO 2 Heteroaryl of which C is 1-6 Alkyl is substituted with any one or more of the following substituents: hydrogen, halogen, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 9 R 10 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 11 R 12 ,C 3-6 Cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, oxygen, halogen and-NR 13 R 14 And the heteroatom of the saturated ring containing 1 heteroatom is substituted by any one or more of the following substituents: hydrogen, methyl and oxygen;
R 6 comprising hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, spirocycloalkyl containing 0 to 3 heteroatoms, C 4-6 Cyclic ketones, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl or heteroaryl, wherein C 1-6 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, aryl, heteroaryl, -C (O) OC 1-6 Alkyl and-C (O) NR 15 R 16 Aryl and heteroaryl groups are substituted with any one or more of the following substituents: halogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl and-NR 17 R 18
R 7 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 8 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 7 、R 8 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 9 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 10 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 11 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 12 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 11 、R 12 And the N atom forms a saturated ring containing 1 or 2 heteroatoms;
R 13 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 14 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 13 、R 14 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 15 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 16 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 15 、R 16 And the N atom forms a 4-7 membered saturated ring containing 1 or 2 heteroatoms;
R 17 comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 18 Comprises H and C 1-6 Alkyl or C 3-6 Cycloalkyl, or R 17 、R 18 And N atom to form a 4-to 7-membered saturated ring containing 1 or 2 heteroatoms
Derivatives include solvates of the piperazinone substituent, pharmaceutically acceptable salts of the piperazinone substituent, or solvates of the pharmaceutically acceptable salts of the piperazinone substituent.
Alternatively, structural units of piperazinone substituents
Figure BDA0002520468220000121
Included
Figure BDA0002520468220000122
Figure BDA0002520468220000123
Alternatively, R 2 Included
Figure BDA0002520468220000124
Wherein Z comprises N or CH;
R 19 including halogen, cyano, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -OC 1-6 Alkyl or-NR 17 R 18
R 20 Including H, halogen, methyl, fluoromethyl, difluoromethyl, or trifluoromethyl.
Alternatively, R 5 Including hydrogen, methyl, ethyl, allyl, propargyl, C 3-6 Cycloalkyl, saturated ring containing 1 heteroatom, spirocycloalkyl containing 0 to 3 heteroatoms, C 4-6 Cyclic ketones, aryl, heteroaryl, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -C (O) aryl, -C (O) heteroaryl, -SO 2 C 1-3 Alkyl, -SO 2 C 3-6 Cycloalkyl, -SO 2 Aryl or-SO 2 Heteroaryl, wherein methyl and ethyl are substituted with any one or more of the following substituents: hydrogen, halogen, C 3-6 Cycloalkyl, hydroxy, -C (O) OC 1-4 Alkyl and-C (O) NR 9 R 10 ,C 3-6 The cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, halogen and-NR 13 R 14 Containing 1 hetero atomHeteroatoms in a saturated ring of atoms are substituted with any one or more of the following substituents: hydrogen, methyl and oxygen;
R 6 comprising hydrogen, C 1-2 Alkyl radical, C 3-6 Cycloalkyl, spirocycloalkyl containing 0 to 3 heteroatoms, C 4-6 Cyclic ketones, aryl or heteroaryl, of which C 1-2 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl, hydroxy, -C (O) OC 1-4 Alkyl and-C (O) NR 15 R 16
R 9 Comprises H and C 1-2 Alkyl or C 3-6 Cycloalkyl radical, R 10 Comprises H and C 1-2 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And N atom form a 4-to 7-membered saturated ring containing 1 or 2 heteroatoms;
R 13 comprises H and C 1-2 Alkyl or C 3-6 Cycloalkyl radical, R 14 Comprises H and C 1-2 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And N atom form a 4-to 7-membered saturated ring containing 1 or 2 heteroatoms;
R 15 comprises H and C 1-2 Alkyl or C 3-6 Cycloalkyl radical, R 16 Comprises H and C 1-2 Alkyl or C 3-6 Cycloalkyl, or R 9 、R 10 And the N atom forms a 4-to 7-membered saturated ring containing 1 or 2 heteroatoms.
Alternatively, structural units of piperazinone substituents
Figure BDA0002520468220000131
Included
Figure BDA0002520468220000132
Figure BDA0002520468220000133
Alternatively, R 2 Included
Figure BDA0002520468220000134
Figure BDA0002520468220000135
Alternatively, R 5 Including hydrogen, methyl, ethyl, 2-trifluoroethyl, allyl, propargyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2-morpholinyl-2-oxoethyl, 2-amino-2-oxoethyl, 2- (dimethylamino) -2-oxoethyl, 2-methoxy-2-oxoethyl, 2-ethoxy-2-oxoethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 4-hydroxycyclohexyl, trans 4-hydroxycyclohexyl, cis 4-hydroxycyclohexyl, 1, 4-dioxaspiro [4.5]Decan-8-yl, 4-oxocyclohexyl, 4- (dimethylamino) cyclohexyl, 4- (diethylamino) cyclohexyl, 4- (pyrrolidin-1-yl) cyclohexyl, 4-difluorocyclohexyl, 4-dimethylcyclohexyl, 1-methylpiperidin-4-yl, tetrahydro-2H-pyran-4-yl, tetrahydro-2H-thiopyran-4-yl, 1-dioxido-tetrahydro-2H-thiopyran-4-yl, tert-butoxycarbonyl, acetyl, propionyl, butyryl, isobutyryl, tert-butylformyl, 2-hydroxypropionyl, cyclopropaneformyl, cyclobutaneformyl, cyclohexanoyl, acryloyl, methanesulfonyl, ethylsulfonyl, n-propylsulfonyl, isopropylsulfonyl or cyclopropylsulfonyl;
R 6 including hydrogen, methyl, ethyl, 2-trifluoroethyl, allyl, propargyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2-morpholinyl-2-oxoethyl, 2- (dimethylamino) -2-oxoethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 4-hydroxycyclohexyl, trans 4-hydroxycyclohexyl, cis 4-hydroxycyclohexyl, 1, 4-dioxyspiro [4.5] ethyl]Decan-8-yl, 4-oxocyclohexyl, 4- (dimethylamino) cyclohexyl, 4- (diethylamino) cyclohexyl, 4- (pyrrolidin-1-yl) cyclohexyl, 4-difluorocyclohexyl, 4-dimethylcyclohexyl, phenyl, pyridin-2-yl, thiazol-2-yl, 1-methyl-1H-pyrazol-4-yl.
Alternatively, the structural formula of the piperazinone substituent comprises:
Figure BDA0002520468220000141
Figure BDA0002520468220000151
Figure BDA0002520468220000161
Figure BDA0002520468220000171
Figure BDA0002520468220000181
Figure BDA0002520468220000191
Figure BDA0002520468220000201
Figure BDA0002520468220000211
Figure BDA0002520468220000221
Figure BDA0002520468220000231
the application provides a preparation method of piperazinone substituent, which comprises the step of using R 1 Substituted 2-chloro-4-morpholinyl thiophene [3,2-d]OH in pyrimidine-6-methanol with R 2 Substituted 2-chloro-4-morpholinyl thiophene [3,2-d]-Cl in pyrimidine-6-methanol.
Wherein 2-chloro-4Morpholinyl thiophene [3,2-d]the-OH group of pyrimidine-6-methanol is first substituted with-Cl to give 4- (2-chloro-6- (chloromethyl) thiophene [3,2-d ]]Pyrimidine-4-yl) morpholine, or by substitution with other halogen, mesylate, tosylate, etc., to give the corresponding derivative, which is then substituted with R 1 Replacing the leaving group.
2-chloro-4-morpholinothieno [3,2-d ] pyrimidine-6-methanol was prepared by the following method:
reacting a mixture containing 2-chloro-4-morpholinyl thiophene [3,2-d ] pyrimidine-6-formaldehyde, methanol and sodium borohydride at-15 ℃ to-5 ℃ for 0.5-3h.
The 2-chloro-4-morpholinyl thiophene [3,2-d ] pyrimidine-6-formaldehyde is prepared by the following method:
reacting a mixture comprising 4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine, tetrahydrofuran and n-butyllithium at-70 ℃ to-80 ℃ for 0.5 to 3h.
4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine was prepared by the following method:
reacting a mixture containing 2, 4-dichlorothieno [3,2-d ] pyrimidine, methanol and morpholine at 20-22 ℃ for 30-40min.
The following is 1-substituted piperazine-2-ketone or 4-substituted piperazine-2-ketone as R 1 2-substituted benzimidazol-1-yl is R 2 The synthetic route for the example is schematically:
Figure BDA0002520468220000241
the application provides a preparation method of piperazinone substituent, which comprises the step of using R 1 Substituted 2-chloro-4-morpholinyl thiophenes [3,2-d]-OH in pyrimidine-6-methanol as R 2 Substituted 2-chloro-4-morpholinyl thiophene [3,2-d]-Cl in 6-methanol of pyrimidine-co.
Wherein 2-chloro-4-morpholinyl thiophene [3,2-d]the-Cl group in the pyrimidine-6-methanol is required to be first substituted by R 2 Substitution to give 2-substituted-4-morpholinothiophene [3,2-d ]]Pyrimidine-6-methanol, with R 1 substituted-OH.
wherein-OH in 2-substituted-4-morpholinyl thiophene [3,2-d ] pyrimidine-6-methanol needs to be substituted into-Cl to obtain 4- (2-substituted-6- (chloromethyl) thiophene [3,2-d ] pyrimidine-4-yl) morpholine, or is substituted by other leaving groups such as halogen, mesylate, p-toluenesulfonate and the like to obtain corresponding derivatives, and then 1-substituted piperazine-2-ketone or piperazine-2-ketone is used for substituting the leaving groups.
2-chloro-4-morpholinothieno [3,2-d ] pyrimidine-6-methanol was prepared by the following method:
reacting a mixture containing 2-chloro-4-morpholinyl thiophene [3,2-d ] pyrimidine-6-formaldehyde, methanol and sodium borohydride at-15 ℃ to-5 ℃ for 0.5-3h.
2-chloro-4-morpholinyl-thieno [3,2-d ] pyrimidine-6-carbaldehyde was prepared by the following method:
reacting a mixture comprising 4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine, tetrahydrofuran and n-butyllithium at-70 ℃ to-80 ℃ for 0.5 to 3h.
4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine was prepared by the following method:
reacting a mixture containing 2, 4-dichlorothieno [3,2-d ] pyrimidine, methanol and morpholine at 20-22 ℃ for 30-40min.
The following is 1-substituted piperazine-2-ketone or 4-substituted piperazine-2-ketone as R 1 2-substituted benzimidazol-1-yl is R 2 The synthetic route for the example is schematically:
Figure BDA0002520468220000251
the application provides a preparation method of piperazinone substituent, which comprises the step of using R 1 By substituting-OH in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol with R 2 Substituted for-Cl in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol.
wherein-OH in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol needs to be replaced by-Cl to obtain 4- (2-chloro-8- (chloromethyl) -9-methyl-9H-purine-6-yl) morpholine, or replaced by other leaving groups such as halogen, mesylate, p-toluenesulfonate and the like to obtain corresponding derivatives, and then R is used 1 Replacing the leaving group.
2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol is prepared by the following method:
reacting a mixture containing 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-formaldehyde, methanol and sodium borohydride at-5 ℃ to-15 ℃ for 0.5-3H.
2-chloro-9-methyl-6-morpholinyl-9H-purine-8-carbaldehyde was prepared by the following method:
the mixture comprising 4- (2-chloro-9H-purin-6-yl) morpholine, tetrahydrofuran and n-butyllithium is reacted at-70 ℃ to-80 ℃ for 0.5 to 3H.
4- (2-chloro-9H-purin-6-yl) morpholine was prepared by the following method:
reacting a mixture containing 2, 6-dichloropurine, methanol and morpholine at 20-22 ℃ for 30-40min.
The following is 1-substituted piperazine-2-ketone or 4-substituted piperazine-2-ketone as R 1 2-substituted benzimidazol-1-yl is R 2 The synthetic route for the example is schematic:
Figure BDA0002520468220000261
if necessary, a part of the functional groups in the reaction may be protected with a conventional protecting agent and deprotected by an appropriate method.
In addition, the piperazinone substituents of the present application can also be prepared by the following synthetic route:
Figure BDA0002520468220000271
the present application provides a pharmaceutical composition comprising a piperazinone substituent or derivative thereof as described above.
Alternatively, the pharmaceutical composition comprises a pharmaceutically acceptable carrier for the piperazinone substituent or a pharmaceutically acceptable carrier for the derivative of the piperazinone substituent.
The application provides an application of the piperazinone substituent or the derivative thereof in preparing a medicament for treating autoimmune diseases or cancers responding to inhibition of PI3K delta.
Wherein the autoimmune disease comprises rheumatoid arthritis, chronic obstructive pulmonary disease, allergic rhinitis or asthma, and the cancer comprises solid tumor or hematological malignancy.
Alternatively, the cancer comprises an autologous leukemia, lymphoma, or a multiple-onset myeloma.
The piperazinone substituted compound or derivative thereof, the preparation method, the application and the pharmaceutical composition thereof are further described in detail with reference to the following examples.
The following abbreviations are used in the following examples and test examples, as shown in table 1:
table 1 english abbreviation of examples and test examples
Figure BDA0002520468220000281
Example 1
Example 1 of the present application provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 1) and a method for its preparation, compound 1 having the following structural formula:
Figure BDA0002520468220000282
(1) Preparation of 4- (2-chlorothieno [3,2-d ] pyrimidin-4-yl) morpholine (1-1)
Figure BDA0002520468220000283
Placing 2, 4-dichlorothiophene [3,2-d ] pyrimidine (compound 1-0) (20.505g, 100mmol) in a three-necked bottle, adding MeOH (250 mL), cooling the reaction liquid to 0-5 ℃, dropwise adding morpholine (200 mmol), heating to 20-22 ℃ after dropwise adding, reacting for 30-40min (TLC detection reaction is finished, EA: n-hexane = 1.
ESI-MS[M+H] + (m/z):256.0.
(2) Preparation of 2-chloro-4-morpholinothieno [3,2-d ] pyrimidine-6-carbaldehyde (1-2)
Figure BDA0002520468220000291
Placing the compound 1-1 (5.112g, 20mmol) in a three-necked bottle, vacuumizing, introducing nitrogen for 3 times, adding ultra-dry THF (50 mL), vacuumizing, introducing nitrogen for 1 time, cooling to-78 ℃, dropwise adding 2.5M n-BuLi (9.6 mL,24mmol, keeping the temperature of a reaction solution below-75 ℃, vacuumizing, introducing nitrogen for 1 time after dropwise adding, stirring at-78 ℃ for reaction for 1 hour, dropwise adding ultra-dry DMF (40 mmol), stirring at-78 ℃ for reaction for 2 hours (HPLC detection reaction is finished), heating to room temperature, slowly pouring the reaction solution into a cooled saturated ammonium chloride aqueous solution, stirring at room temperature for 30 minutes, performing suction filtration, soaking and washing a filter cake with ice water, and drying to obtain a khaki product 1-2 (5.431 g), wherein the yield is 95.7%.
ESI-MS[M+H] + (m/z):284.0.
(3) Preparation of 2-chloro-4-morpholinothieno [3,2-d ] pyrimidine-6-methanol (1-3)
Figure BDA0002520468220000292
Putting the intermediate 1-2 (5.709g, 20mmol) in a three-necked bottle, adding methanol, cooling to about-10 ℃, adding sodium borohydride (30 mmol) in portions, keeping the temperature at-10 ℃ for reaction for 1h after the addition is finished, dropwise adding 90mL of ice water, stirring for 10min at-10 ℃, performing suction filtration, washing a filter cake with ice water, and drying to obtain alcohol 1-3 (5.537g, 19.428mmol) with the yield of 97%.
ESI-MS[M+Na] + (m/z):308.1.
(4) Preparation of (2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothien [3,2-d ] opyrimidin-6-yl) methanol (1-4)
Figure BDA0002520468220000301
The compound 1-3 (4.275g, 15mmol), 2-ethylbenzimidazole (18 mmol), pd 2 (dba) 3 (1.5mmol),Xphos(3mmol),CsCO 3 (30 mmol) is placed in a three-necked flask, dioxane (100 mL) is added, nitrogen is introduced in the flask for 3 times by vacuum pumping, the temperature is raised to 110 ℃ for reaction for 4-5h, (DCM: EA =1 for basic reaction completion by TLC), the mixture is filtered while hot, the filter cake is washed with DCM, the filtrate is concentrated and stirred with silica gel, and column chromatography gives the target product 1-4 (2.646 g) in 45% yield.
ESI-MS[M+H] + (m/z):396.2.
(5) Preparation of 4- (6- (chloromethyl) -2- (2-ethyl-1H-benzimidazol-1-yl) thiophen [3,2-d ] opyrimidin-4-yl) morpholine (1-5)
Figure BDA0002520468220000302
Placing the compound 1-4 (1.975g, 5 mmol) in a three-necked bottle at room temperature, adding 50mL of dichloromethane, cooling to 0 ℃, slowly adding thionyl chloride (5 mL) in a stirring state, heating to room temperature after the addition to react for 2h (TLC detection reaction is finished, n-hexane: EA = 2.
ESI-MS[M+H] + (m/z):414.3.
(6) Preparation of 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (Compound 1)
Figure BDA0002520468220000311
Putting piperazinone (1.5 mmol) and the intermediate 1-5 (1.5 mmol) into a round-bottomed flask, adding isopropanol and diisopropylethylamine (5 mmol), heating to 85 ℃, reacting and stirring for 24h, detecting complete reaction (EA) by TLC, concentrating the reaction solution under reduced pressure, stirring the sample with silica gel, and performing column chromatography to obtain the target product compound 1 (0.480g, 1mmol) with the yield of 67%.
1 H NMR(400MHz,CDCl 3 )δ7.94–7.88(m,1H),7.72–7.65(m,1H), 7.27(s,1H),7.22–7.17(m,2H),6.36(s,1H),4.03–3.93(m,4H),3.87(s,2H), 3.85–3.79(m,4H),3.35(d,J=4.0Hz,2H),3.32–3.22(m,4H),2.78–2.68 (m,2H),1.36(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ168.74,162.78,158.50,156.93,153.31, 149.71,142.58,134.58,123.44,122.96,122.76,119.09,113.19,112.05,66.73, 56.86,56.40,48.83,46.51,41.33,23.66,12.28.
ESI-MS[M+H] + (m/z):478.2.
Example 2
Example 2 of the present application provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -1-methylpiperazin-2-one (compound 2) and a process for its preparation, compound 2 having the following structural formula:
Figure BDA0002520468220000321
1-methylpiperazin-2-one (0.5 mmol) and the intermediates 1 to 5 (0.5 mmol) of example 1 were placed in a round-bottomed flask, isopropanol (5 mL) and diisopropylethylamine (2.5 mmol) were added, the mixture was heated to 85 ℃ and stirred for 24h, completion of the reaction (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure, and after stirring with silica gel, column chromatography was performed to obtain the objective compound 2 (0.177 g, 0.36mmol) in a yield of 72%.
1 H NMR(400MHz,CDCl 3 )δ8.01–7.96(m,1H),7.78–7.72(m,1H), 7.32(s,1H),7.29–7.24(m,2H),4.09–4.00(m,4H),3.92–3.84(m,6H),3.39 -3.31(m,4H),3.30(s,2H),2.98(s,3H),2.82(t,J=5.4Hz,2H),1.43(t,J= 7.5Hz,3H).
ESI-MS[M+H] + (m/z):492.2.
Example 3
Example 3 of the present application provides 1-ethyl-4- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one and a method of making the same, compound 3 having the following structural formula:
Figure BDA0002520468220000322
1-ethylpiperazin-2-one (0.2 mmol) and intermediates 1 to 5 (0.5 mmol) from example 1 were placed in a round-bottomed flask, isopropanol (5 mL) and diisopropylethylamine (2.5 mmol) were added, the mixture was heated to 85 ℃ and stirred for 24h, reaction completion (EA) was detected by TLC, the reaction mixture was concentrated under reduced pressure, and after sample-mixing with silica gel, column chromatography was performed to obtain the objective compound 3 (0.087g, 0.17mmol) with a yield of 86%.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.95(m,1H),7.79–7.72(m,1H), 7.33(s,1H),7.29–7.22(m,2H),4.08–3.99(m,4H),3.93–3.84(m,6H),3.46 (q,J=7.2Hz,2H),3.40–3.31(m,4H),3.29(s,2H),2.85–2.78(m,2H),1.43 (t,J=7.5Hz,3H),1.17(t,J=7.2Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ165.80,162.74,158.49,156.92,153.27, 149.62,142.48,134.55,123.48,122.95,122.75,119.03,113.23,112.07,66.71, 57.29,56.41,49.55,46.50,45.76,41.19,23.63,12.27,12.12.
ESI-MS[M+H] + (m/z):506.3.
Example 4
Example 4 of the present application provides 1- (cyclopropylmethyl) -4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 4) and a method for its preparation, compound 4 having the following structural formula:
Figure BDA0002520468220000331
placing the compound 1 (0.095g, 0.2mmol) in a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding the intermediate 3-bromopropylene (0.6 mmol) in portions, reacting for 4-5h at 30-32 ℃, detecting by TLC until the reaction is complete (EA), cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in the water layer, washing the organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and performing column chromatography to obtain the target product compound 4 (0.095mmol, 0.049 g) with the yield of 47%.
1 H NMR(400MHz,CDCl 3 )δ8.04–7.93(m,1H),7.79–7.70(m,1H), 7.33(s,1H),7.29–7.23(m,2H),5.78(ddd,J=16.9,5.9,4.4Hz,1H),5.22 (ddd,J=12.6,7.3,1.4Hz,2H),4.11–3.96(m,6H),3.95–3.85(m,6H),3.39 –3.27(m,6H),2.86–2.76(m,2H),1.43(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ166.05,162.75,158.49,156.92,153.27, 149.57,142.49,134.55,132.13,123.48,122.96,122.76,119.04,118.07,113.22, 112.06,66.71,57.22,56.39,49.53,48.64,46.50,45.82,23.63,12.27.
ESI-MS[M+H] + (m/z):518.3.
Example 5
Example 5 of the present application provides 1- (cyclopropylmethyl) -4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 5) and a method for its preparation, compound 5 having the following structural formula:
Figure BDA0002520468220000341
placing the compound 1 (0.095g, 0.2mmol) in example 1 into a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding an intermediate bromomethylcyclopropane (0.6 mmol) in portions, reacting for 4-5h at 30-32 ℃, keeping the reaction temperature at 30-32 ℃, detecting the basic reaction (EA) completely, cooling the reaction liquid, adding water and ethyl acetate, extracting until no product exists in a water layer, washing an organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and performing column chromatography to obtain the target product compound 5 (0.8mmol, 0.06g) with the yield of 64%.
1 H NMR(400MHz,CDCl 3 )δ7.74(dd,J=6.0,3.1Hz,1H),7.49(dd,J= 6.1,2.9Hz,1H),7.08(s,1H),7.05–6.97(m,2H),3.84–3.74(m,4H),3.67–3.58(m,6H),3.22(t,J=5.3Hz,2H),3.13–3.02(m,6H),2.57(t,J=5.4Hz, 2H),1.18(t,J=7.5Hz,3H),0.81–0.67(m,1H),0.32–0.25(m,2H),0.01(d, J=5.4Hz,2H).
13 C NMR(100MHz,CDCl 3 )δ165.98,162.71,158.46,156.90,153.23, 149.61,142.47,134.54,123.48,122.94,122.72,119.00,113.24,112.06,66.69, 57.27,56.41,50.47,49.53,46.48,46.36,23.63,12.26,8.98,3.57.
ESI-MS[M+H] + (m/z):532.3.
Example 6
Example 6 of the present application provides 1-cyclopentyl-4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 6) and a method for its preparation, compound 6 having the following structural formula:
Figure BDA0002520468220000351
1-cyclopentylpiperazine-2-one (0.3 mmol) and the intermediates 1 to 5 (0.2 mmol) of example 1 were placed in a round-bottom flask, isopropanol (5 mL) and diisopropylethylamine (1 mmol) were added, the mixture was heated to 85 ℃ to react and stir for 24h, the reaction (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure, and the target product compound 6 (0.065g, 0.119mmol) was obtained by silica gel-batch column chromatography with a yield of 60%.
1 H NMR(400MHz,CDCl 3 )δ8.03–7.95(m,1H),7.79–7.71(m,1H), 7.32(s,1H),7.32–7.22(m,2H),4.95(p,J=8.4Hz,1H),4.09–3.99(m,4H), 3.88(d,J=5.1Hz,6H),3.39–3.26(m,6H),2.82–2.74(m,2H),1.85(d,J= 5.2Hz,2H),1.73–1.48(m,6H),1.43(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ166.11,162.73,158.46,156.91,153.26, 149.59,142.56,134.57,123.47,122.92,122.71,119.05,113.24,112.03,66.71, 57.53,56.42,53.86,49.70,46.49,40.94,27.87,24.07,23.66,12.27.
ESI-MS[M+H] + (m/z):546.3.
Example 7
Example 7 of the present application provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -1- (trans-4-hydroxycyclohexyl) piperazin-2-one (compound 7) and a process for its preparation, compound 7 having the following structural formula:
Figure BDA0002520468220000361
1- (trans-4-hydroxycyclohexyl) piperazin-2-one (0.3 mmol) and intermediates 1-5 of example 1 (0.2 mmol) were placed in a round bottom flask, isopropanol (5 mL) and diisopropylethylamine (1 mmol) were added, the temperature was raised to 85 ℃ and the reaction stirred for 24h, reaction completion (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure, and then column chromatography was performed after silica gel-mixing to give the desired product compound 7 (0.041 g, 0.71 mmol) with a yield of 36%.
1 H NMR(400MHz,CDCl 3 )δ7.99(d,J=4.7Hz,1H),7.74(d,J=4.5Hz, 1H),7.33(s,1H),7.27(d,J=6.8Hz,2H),4.44(s,1H),4.04(s,4H),3.89(s, 6H),3.55(s,1H),3.38–3.26(m,6H),2.77(s,2H),2.04(d,J=9.1Hz,2H), 1.72(s,2H),1.55–1.40(m,7H).
13 C NMR(100MHz,CDCl 3 )δ165.82,162.72,158.48,156.92,153.27, 149.42,142.52,134.56,123.56,122.96,122.76,119.04,113.21,112.07,69.77, 66.72,57.53,56.40,51.29,49.66,46.50,41.13,34.39,27.10,23.63,12.30.
ESI-MS[M+H] + (m/z):576.3.
Example 8
Example 8 of the present application provides 2- (4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -2-oxopiperazin-1-yl) acetamide (compound 8) and a process for its preparation, compound 8 having the following structural formula:
Figure BDA0002520468220000371
placing the compound 1 (0.095g, 0.2mmol) of example 1 in a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting, stirring for 20min, cooling to 0 ℃, adding intermediate iodoacetamide (0.3 mmol) in portions, keeping 0 ℃ for reaction for 2h, detecting by TLC until the reaction is almost complete (EA), cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in the water layer, washing the organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and performing column chromatography to obtain the target product compound 8 (0.105mmol, 0.056 g), wherein the yield is 52%.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.96(m,1H),7.79–7.73(m,1H), 7.34(s,1H),7.27(d,J=6.4Hz,2H),4.05(s,6H),3.98–3.83(m,6H),3.54(t, J=5.1Hz,2H),3.40–3.30(m,4H),2.88(t,J=5.1Hz,2H),1.43(t,J=7.4Hz, 3H).
13 C NMR(100MHz,CDCl 3 )δ170.28,167.33,162.74,158.51,156.91, 153.35,149.18,142.55,134.56,123.67,122.97,122.78,119.10,113.20,112.07, 66.73,57.06,56.27,50.55,49.35,48.15,46.51,23.66,12.28.
ESI-MS[M+H] + (m/z):535.3.
Example 9
Example 9 of the present application provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) -1- (2-morpholinyl-2-oxoethyl) piperazin-2-one (compound 9) and a method for its preparation, compound 9 having the following structural formula:
Figure BDA0002520468220000381
placing the compound 1 (0.095g, 0.2mmol) of example 1 in a round-bottom flask, adding THF and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding an intermediate chloroacetyl morpholine (0.3 mmol) in portions, keeping 30 ℃ for reaction for 2-3h, detecting by TLC that the reaction is basically complete (EA), cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in an aqueous layer, washing an organic phase by saturated saline, drying the organic phase by anhydrous magnesium sulfate, mixing the organic phase with silica gel, and carrying out column chromatography to obtain the target product compound 9 (0.101mmol, 0.061g) with the yield of 50%.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.96(m,1H),7.78–7.72(m,1H), 7.34(s,1H),7.31–7.23(m,2H),4.21(s,2H),4.10–4.00(m,4H),3.96(s,2H), 3.93–3.83(m,4H),3.70(d,J=3.3Hz,4H),3.63(d,J=4.3Hz,2H),3.55 -3.45(m,4H),3.39(s,2H),3.34(q,J=7.5Hz,2H),2.91(t,J=5.3Hz,2H), 1.43(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ166.94,165.96,162.75,158.50,156.93, 153.28,149.62,142.54,134.57,123.51,122.94,122.74,119.06,113.22,112.09, 66.73,56.96,56.19,49.29,47.65,47.21,46.50,45.40,42.26,23.64,12.27.
ESI-MS[M+H] + (m/z):605.3.
Example 10
Example 10 of the present application provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one (compound 10) and a method for its preparation, compound 10 having the following structural formula:
Figure BDA0002520468220000391
(1) Preparation of 1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one hydrochloride (10-1)
Figure BDA0002520468220000392
Placing 4-Boc-piperazine-2-one (10 mmol), 1-methyl-4-bromo-1H-pyrazole (10 mmol), potassium carbonate (20 mmol) and cuprous iodide (1 mmol) in a round-bottom flask, adding toluene (50 mL), replacing the air in the flask with nitrogen for 3 times at room temperature, injecting N, N-dimethylethylenediamine (2 mmol) into the flask under the protection of nitrogen, heating to 110 ℃, reacting and stirring for 24H, evaporating the solvent under reduced pressure, then adding 100mL of water into the flask, stirring vigorously at room temperature for 1H, filtering, washing with saturated ammonium chloride solution (50 mL) and water (50 mL), and drying to obtain a crude product, namely 1- (1-methyl-1H-pyrazol-4-yl) -4-tert-butoxycarbonyl-piperazine-2-one.
The crude 1- (1-methyl-1H-pyrazol-4-yl) -4-tert-butoxycarbonyl-piperazin-2-one was placed in a round bottom flask, then dioxane hydrochloride solution (30 mL) was added thereto, stirred at room temperature for 2H, filtered rapidly and the product was washed with cold dioxane to give the compound 1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one hydrochloride (10-1) (8.71mmol, 1.887g) in 87% yield after drying.
ESI-MS[M+H] + (m/z):181.1.
(2) Preparation of 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] opyrimidin-6-yl) methyl) -1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one (Compound 10)
Figure BDA0002520468220000401
1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one hydrochloride (10-1) (0.3 mmol) and intermediates 1-5 (0.2 mmol) of example 1 were placed in a round-bottomed flask, isopropanol (5 mL) and diisopropylethylamine (1 mmol) were added, the temperature was raised to 85 ℃ and the reaction was stirred for 12h, completion of reaction (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure, and then subjected to silica gel sampling and column chromatography to obtain the target product compound 10 (0.079g, 0.14mmol) with a yield of 71%.
1 H NMR(400MHz,CDCl 3 )δ8.04(s,1H),8.02–7.97(m,1H),7.78– 7.72(m,1H),7.49(s,1H),7.35(s,1H),7.29–7.24(m,2H),4.08–3.99(m, 4H),3.95(s,2H),3.91–3.85(m,7H),3.75(t,J=5.4Hz,2H),3.46(d,J=6.1 Hz,4H),3.35(q,J=7.5Hz,2H),1.43(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ164.13,162.74,158.49,156.92,153.28, 149.47,142.47,134.53,129.48,124.90,123.50,123.07,122.98,122.78,119.03, 113.24,112.08,66.70,57.70,56.30,49.22,47.49,46.49,39.33,23.64,12.27.
ESI-MS[M+H] + (m/z):558.3.
Example 11
Example 11 of the present application provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -1- (thiazol-2-yl) piperazin-2-one (compound 11) and a method for its preparation, compound 11 having the following structural formula:
Figure BDA0002520468220000411
(1) Preparation of 1- (thiazol-2-yl) piperazin-2-one hydrochloride (11-1)
Figure BDA0002520468220000412
Putting 4-Boc-piperazine-2-one (10 mmol), 2-bromothiazole (10 mmol), potassium carbonate (20 mmol) and cuprous iodide (1 mmol) into a round-bottom flask, adding toluene (30 mL), replacing the air in the flask with nitrogen for 3 times at room temperature, injecting N, N-dimethylethylenediamine (2 mmol) into the flask under the protection of nitrogen, heating to 110 ℃, reacting and stirring for 24h, evaporating the solvent under reduced pressure, then adding 100mL of water into the flask, vigorously stirring for 1h at room temperature, filtering, washing with saturated ammonium chloride solution (50 mL) and water (50 mL) in sequence, and drying to obtain a crude product, namely 1- (thiazole-2-yl) -4-tert-butoxycarbonyl-piperazine-2-one.
Crude 1- (thiazol-2-yl) -4-tert-butoxycarbonyl-piperazin-2-one was placed in a round bottom flask, then dioxane hydrochloride solution (30 mL) was added thereto, stirred at room temperature for 2h, filtered rapidly and the product was washed with cold dioxane to give after drying the compound 1- (thiazol-2-yl) piperazin-2-one hydrochloride (11-1) (6.33mmol, 1.391g) in 63% yield.
ESI-MS[M+H] + (m/z):184.1.
(2) Preparation of 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) -1- (thiazol-2-yl) piperazin-2-one (Compound 11)
Figure BDA0002520468220000421
1- (thiazol-2-yl) piperazin-2-one hydrochloride (11-1) (0.3 mmol) and intermediate 1-5 of example 1 (0.2 mmol) were placed in a round bottom flask, isopropanol (5 mL) and diisopropylethylamine (1 mmol) were added, the reaction was stirred at 85 ℃ for 12h, reaction completion (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure, and the product was subjected to silica gel-loading and column chromatography to give the desired product compound 11 (0.049 g,0.14 mmol) with a yield of 71%.
1 H NMR(400MHz,CDCl 3 )δ7.96–7.88(m,1H),7.72–7.65(m,1H), 7.46(d,J=3.5Hz,1H),7.29(s,1H),7.22–7.16(m,2H),6.99(d,J=3.5Hz, 1H),4.25–4.15(m,2H),4.01–3.91(m,6H),3.87–3.76(m,4H),3.51(s,2H), 3.28(q,J=7.5Hz,2H),2.96(t,J=5.5Hz,2H),1.36(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ165.45,162.79,158.64,158.51,156.92, 153.35,149.27,142.49,137.22,134.55,123.53,122.99,122.80,119.08,115.36, 113.23,112.08,66.72,57.30,56.22,49.30,47.08,46.51,23.66,12.29.
ESI-MS[M+H] + (m/z):561.2.
Example 12
Example 12 of the present application provides 4- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -1- (pyridin-3-yl) piperazin-2-one (compound 12) and a method for its preparation, compound 12 having the following structural formula:
Figure BDA0002520468220000431
(1) Preparation of 1- (pyridin-3-yl) piperazin-2-one dihydrochloride (12-1)
Figure BDA0002520468220000432
Putting 4-Boc-piperazine-2-one (10 mmol), 3-bromopyridine (10 mmol), potassium carbonate (20 mmol) and cuprous iodide (1 mmol) into a round-bottom flask, adding toluene (30 mL), replacing the air in the flask with nitrogen for 3 times at room temperature, injecting N, N-dimethylethylenediamine (2 mmol) into the flask under the protection of nitrogen, heating to 110 ℃, reacting and stirring for 24h, evaporating the solvent under reduced pressure, then adding 100mL of water into the flask, vigorously stirring for 1h at room temperature, filtering, washing with saturated ammonium chloride solution (50 mL) and water (50 mL) in sequence, and drying to obtain a crude product, namely 1- (pyridin-3-yl) -4-tert-butoxycarbonyl-piperazine-2-one.
Crude 1- (pyridin-3-yl) -4-tert-butoxycarbonyl-piperazin-2-one was placed in a round bottom flask, then dioxane hydrochloride solution (30 mL) was added thereto, stirred at room temperature for 2h, filtered rapidly and the product washed with cold dioxane to give after drying the compound 1- (pyridin-3-yl) piperazin-2-one bis-hydrochloride (12-1) (6.91mmol, 1.728g) in 69% yield.
ESI-MS[M+H] + (m/z):178.1.
(2) Preparation of 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophene [3,2-d ] pyrimidin-6-yl) methyl) -1- (pyridin-3-yl) piperazin-2-one (Compound 12)
Figure BDA0002520468220000441
1- (thiazol-2-yl) piperazin-2-one hydrochloride (12-1) (0.3 mmol) and intermediate 1-5 of example 1 (0.2 mmol) were placed in a round bottom flask, isopropanol and diisopropylethylamine (1 mmol) were added, the temperature was raised to 85 ℃ to react and stir for 12h, tlc detected for completion of reaction (EA), the reaction solution was concentrated under reduced pressure, and after sample stirring with silica gel, column chromatography was performed to give the target product compound 12 (0.085g, 0.15 mmol) in 76% yield.
1 H NMR(400MHz,CDCl 3 )δ8.44(dd,J=4.8,1.0Hz,1H),7.99(dd,J= 11.6,4.9Hz,2H),7.78–7.69(m,2H),7.36(s,1H),7.28(d,J=3.9Hz,2H), 7.13(dd,J=6.9,5.3Hz,1H),4.13–4.08(m,2H),4.08–4.03(m,4H),4.00(s, 2H),3.92–3.87(m,4H),3.53(s,2H),3.35(q,J=7.5Hz,2H),3.02–2.95(m, 2H),1.44(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ166.89,162.85,158.53,156.93,153.17, 149.74,147.77,142.67,137.36,134.62,126.58,123.38,122.92,122.72,120.95, 119.93,119.14,113.20,112.06,66.75,58.36,56.36,49.89,46.52,46.41,23.69, 12.28.
ESI-MS[M+H] + (m/z):555.3.
Example 13
Example 13 of the present application provides 1-allyl-4- ((2- (2-methyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 13) and a process for its preparation, compound 13 having the following structural formula:
Figure BDA0002520468220000451
(1) Preparation of (2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinothieno [3,2-d ] pyrimidin-6-yl) methanol (13-1)
Figure BDA0002520468220000452
Intermediates 1 to 3 (4.275g, 15mmol) in example 1, 2-methylbenzimidazole (18 mmol), pd 2 (dba) 3 (1.5mmol),Xphos(3mmol),CsCO 3 (30 mmol) was placed in a three-necked flask, dioxane (100 mL) was added, nitrogen was introduced 3 times under vacuum, the temperature was raised to 110 ℃ for reaction for 4-5h, (DCM: EA =1 for basic completion of the reaction by TLC), the mixture was filtered while hot, the filter cake was washed with DCM, the filtrate was concentrated and stirred with silica gel, and column chromatography gave the desired product 13-1 (3.029 g,7.95mmol, 53% yield).
ESI-MS[M+H] + (m/z):382.2.
(2) Preparation of 4- (6- (chloromethyl) -2- (2-methyl-1H-benzimidazol-1-yl) thiophen [3,2-d ] opyrimidin-4-yl) morpholine (13-2)
Figure BDA0002520468220000453
Placing the compound 13-1 (1.905g, 5 mmol) in a three-necked bottle at room temperature, adding 50mL of dichloromethane, cooling to 0 ℃, slowly adding thionyl chloride (5 mL) into the three-necked bottle under stirring, after the addition, heating to room temperature for reaction for 2h (TLC detection reaction is finished, n-hexane: EA = 2).
ESI-MS[M+H] + (m/z):400.3.
(3) Preparation of 4- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (13-3)
Figure BDA0002520468220000461
Putting piperazinone (1.5 mmol) and intermediate 13-2 (1.5 mmol) into a round bottom flask, adding isopropanol and diisopropylethylamine (5 mmol), heating to 85 ℃, reacting and stirring for 24h, detecting complete reaction (EA) by TLC, concentrating the reaction solution under reduced pressure, stirring with silica gel, and performing column chromatography to obtain intermediate 13-3 (0.574g, 1.24mmol) with yield of 82.7%.
ESI-MS[M+H] + (m/z):464.2.
(4) Preparation of 1-allyl-4- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinylthiophene [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (Compound 13)
Placing a compound 13-3 (0.093g, 0.2mmol) in a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding an intermediate 3-bromopropylene (0.6 mmol) in portions, keeping 30-32 ℃ for reaction for 4-5h, detecting by TLC that the reaction is complete (EA), cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in a water layer, washing an organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, stirring the organic phase with silica gel, and carrying out column chromatography to obtain a target product compound 13 (0.103mmol, 0.052g), wherein the yield is 52%.
1 H NMR(400MHz,CDCl 3 )δ8.12–8.03(m,1H),7.71(dd,J=6.3,2.7 Hz,1H),7.33(s,1H),7.29–7.26(m,2H),5.83–5.71(m,1H),5.27–5.10(m, 2H),4.08–3.95(m,6H),3.95-3.83(m,6H),3.35(d,J=8.4Hz,4H),2.93(s, 3H),2.85–2.76(m,2H).
13 C NMR(100MHz,CDCl 3 )δ166.03,162.71,158.45,153.40,152.30, 149.56,142.46,134.41,132.14,123.47,123.01,122.88,118.87,118.07,113.54, 111.98,66.71,57.24,56.40,49.52,48.64,46.53,45.82,17.75.
ESI-MS[M+H] + (m/z):504.3.
Example 14
Example 14 of the present application provides 1- (cyclopropylmethyl) -4- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 14) and a method for its preparation, compound 14 having the following structural formula:
Figure BDA0002520468220000471
placing the intermediate 13-3 (0.093 g, 0.2mmol) obtained in example 13 into a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding the intermediate bromomethylcyclopropane (0.6 mmol) in portions, keeping 30-32 ℃ for reaction for 4-5h, detecting by TLC that the reaction is basically complete (EA), cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in the water layer, washing the organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and performing column chromatography to obtain the target product compound 14 (0.145mmol, 0.075g) with the yield of 73%.
1 H NMR(400MHz,CDCl 3 )δ8.11–8.03(m,1H),7.74–7.66(m,1H), 7.33(s,1H),7.27(dd,J=6.0,3.2Hz,2H),4.10–3.99(m,4H),3.94–3.84(m, 6H),3.50–3.44(m,2H),3.31(d,J=6.8Hz,4H),2.93(s,3H),2.85–2.79(m, 2H),1.04–0.94(m,1H),0.54(q,J=5.6Hz,2H),0.27(t,J=5.1Hz,2H).
13 C NMR(100MHz,CDCl 3 )δ165.99,162.68,158.43,153.38,152.31, 149.57,142.53,134.42,123.48,122.98,122.83,118.86,113.54,111.97,66.70, 57.30,56.42,53.47,50.49,49.52,46.51,17.78,8.99,3.58.
ESI-MS[M+H] + (m/z):518.3.
Example 15
Example 15 of the present application provides 1-cyclopentyl-4- ((2- (2-methyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 15) and a method for its preparation, compound 15 having the following structural formula:
Figure BDA0002520468220000481
1-cyclopentylpiperazin-2-one (0.3 mmol) and the intermediate 13-3 (0.2 mmol) of example 13 were placed in a round-bottomed flask, isopropanol (5 mL) and diisopropylethylamine (1 mmol) were added, the temperature was raised to 85 ℃ and the reaction was stirred for 24h, the reaction (EA) was detected by TLC and the reaction solution was concentrated under reduced pressure, followed by silica gel stirring and column chromatography to give the desired compound 15 (0.069 g, 0.130mmol) in 65% yield.
1 H NMR(400MHz,CDCl 3 )δ8.08(s,1H),7.71(s,1H),7.33(s,1H),7.28 (s,2H),5.04–4.84(m,1H),4.04(s,4H),3.89(s,6H),3.31(d,J=10.1Hz,4H), 2.93(s,3H),2.79(s,2H),1.94–1.55(m,8H).
13 C NMR(100MHz,CDCl 3 )δ166.11,162.70,158.43,153.40,152.30, 149.57,142.56,134.43,123.47,122.97,122.83,118.89,113.55,111.94,66.71, 57.56,56.43,53.86,49.69,46.52,40.94,27.87,24.08,17.81.
ESI-MS[M+H] + (m/z):532.3.
Example 16
Example 16 of the present application provides 4- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -1- (2-morpholino-2-oxoethyl) piperazin-2-one (compound 16) and a process for its preparation, compound 16 having the following structural formula:
Figure BDA0002520468220000491
placing the compound 13-3 (0.095g, 0.2mmol) of example 13 into a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding an intermediate chloroacetyl morpholine (0.3 mmol) in portions, keeping the temperature at 30 ℃ for reaction for 2-3h, detecting the basic reaction (EA) by TLC, cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in an aqueous layer, washing an organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and performing column chromatography to obtain the target product compound 16 (0.120mmol, 0.071g) with the yield of 60%.
1 H NMR(400MHz,CDCl 3 )δ8.11–8.03(m,1H),7.74–7.67(m,1H), 7.34(s,1H),7.31–7.23(m,2H),4.21(s,2H),4.08–4.00(m,4H),3.95(s,2H), 3.92–3.85(m,4H),3.73–3.66(m,4H),3.62(d,J=4.3Hz,2H),3.54–3.45 (m,4H),3.38(s,2H),2.93(s,3H),2.92–2.88(m,2H).
13 C NMR(100MHz,CDCl 3 )δ166.91,165.95,162.67,158.44,153.36, 152.30,149.67,142.49,134.41,123.44,122.98,122.83,118.84,113.57,112.00, 66.69,56.95,56.16,49.26,47.62,47.20,46.51,45.37,42.23,17.76.
ESI-MS[M+H] + (m/z):591.3.
Example 17
Example 17 of the present application provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4-methylpiperazin-2-one (compound 17) and a method for its preparation, compound 17 having the following structural formula:
Figure BDA0002520468220000501
4-methylpiperazin-2-one (0.5 mmol) was placed in a round bottom flask, THF (5 mL) and potassium tert-butoxide (0.75 mmol) were added at 0 ℃ and stirred for 30min, then the intermediates 1 to 5 (0.5 mmol) of example 1 were slowly added to the reaction solution and warmed to room temperature, the reaction was stirred for 6h, the reaction (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure and extracted with ethyl acetate and water, the organic layer was sample-stirred with silica gel and column-chromatographed to give the desired product compound 17 (0.146g, 0.297 mmol) in 59% yield.
1 H NMR(400MHz,CDCl 3 )δ7.94–7.87(m,1H),7.70–7.62(m,1H), 7.26(s,1H),7.23–7.15(m,2H),4.77(s,2H),3.99–3.89(m,4H),3.83–3.75 (m,4H),3.35(t,J=5.4Hz,2H),3.26(q,J=7.5Hz,2H),3.13(s,2H),2.60(t, J=5.4Hz,2H),2.27(s,3H),1.35(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ167.25,162.34,158.54,156.92,153.32, 147.55,142.52,134.55,124.28,122.96,122.76,119.04,113.24,112.46,66.69, 59.12,51.67,46.53,46.47,45.24,45.07,23.66,12.27.
ESI-MS[M+H] + (m/z):492.2.
Example 18
Example 18 of the present application provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (2, 2-trifluoroethyl) piperazin-2-one (compound 18) and a method for its preparation, compound 18 having the following structural formula:
Figure BDA0002520468220000502
(1) Preparation of tert-butyl 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophene [3,2-d ] pyrimidin-6-yl) methyl) -3-oxopiperazine-1-carboxylate (18-1)
Figure BDA0002520468220000511
Placing N-boc-piperazinone (1.505g, 7.5 mmol) in a three-neck bottle, adding THF (5 mL) and t-BuOK (10 mmol) at 0 ℃, heating to 30-32 ℃, reacting and stirring for 20min, adding the intermediate 1-5 (2.070g, 5 mmol) of example 1 in portions, gradually changing the system color from light yellow to black, reacting for 4-5h at 30-32 ℃, detecting by TLC that the reaction is basically complete (EA), cooling the reaction liquid, adding water and ethyl acetate, extracting until no product exists in a water layer, washing an organic phase with saturated saline water, drying the organic phase with anhydrous magnesium sulfate, and concentrating to dryness to obtain a crude compound 18-1. The crude product is subjected to silica gel sample mixing and column chromatography to obtain a target product 18-1 (2.388g, 4.139mmol), and the yield is 83%.
ESI-MS[M+H] + (m/z):578.3.
(2) Preparation of 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one trifluoroacetate (18-2)
Figure BDA0002520468220000512
Placing the intermediate 18-1 (1.870 g, 4.001mmol) in a three-necked bottle, adding TFA (20 mL) at 0 ℃, slowly raising the temperature to room temperature, reacting and stirring for 2h, distilling the solvent under reduced pressure, adding methanol (20 mL) to the residual oily substance, distilling the solvent under reduced pressure, repeating the steps for three times, and drying the residual oily substance in vacuum to obtain a foamy light yellow solid 18-2 with the yield of 100%.
(3) Preparation of 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophene [3,2-d ] pyrimidin-6-yl) methyl) -4- (2, 2-trifluoroethyl) piperazin-2-one (Compound 18)
Figure BDA0002520468220000521
Putting the intermediate 18-2 (0.2 mmol) into a three-necked bottle, adding potassium carbonate (0.5 mmol), acetonitrile (5 mL) and trifluoroethyl trifluoromethanesulfonate (0.4 mmol), slowly heating to a reflux temperature (85 ℃), stirring for 2h, distilling out the solvent under reduced pressure after TLC detection reaction is complete, adding water and ethyl acetate into the residue, extracting until no product exists in an aqueous layer, washing an organic phase with saturated saline solution, drying the organic phase with anhydrous magnesium sulfate, concentrating, mixing with silica gel, and performing column chromatography to obtain the target product compound 18 (0.053 g,0.095 mmol) with the yield of 47%.
1 H NMR(400MHz,CDCl 3 )δ8.01–7.96(m,1H),7.77–7.72(m,1H), 7.35(s,1H),7.30–7.23(m,2H),4.84(s,2H),4.08–3.98(m,4H),3.92–3.82 (m,4H),3.51(s,2H),3.47–3.40(m,2H),3.34(q,J=7.5Hz,2H),3.13-3.03 (m,2H),2.97(t,J=5.3Hz,2H),1.43(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ162.90,158.50,156.92,153.19,151.50, 142.64,134.63,124.93(q,J=278Hz),124.45,122.94,122.87,122.66,119.08, 113.21,111.97,67.51,66.76,57.59,53.08,52.11,46.51,30.32,23.66,12.27.
19 F NMR(376MHz,CDCl 3 )δ-69.08.
ESI-MS[M+H] + (m/z):560.2.
Example 19
Example 19 of the present application provides 1- (cyclopropylmethyl) -4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 19) and a process for its preparation, compound 19 having the following structural formula:
Figure BDA0002520468220000531
compound 5 (0.15 mmol) from example 5 was dissolved in DMSO (1mL, 0.15mol/L), zinc trifluoromethanesulphinate (TFMS, 0.165g, 0.5mmol) was added thereto at room temperature, after both were completely dissolved, t-butanol peroxide (TBHP, 70%,0.064mL, 0.5 mmol) was slowly added to the reaction solution, the reaction solution was warmed to 50 ℃ and stirred for 6h, TFMS (1.5 eq) and TBHP (1.5 eq) were added again to the reaction solution and the reaction was continued for 12h. After the reaction solution was cooled, extraction was performed with ethyl acetate and water, the ethyl acetate layer was washed with water and saturated brine, and the organic layer was dried over anhydrous magnesium sulfate and then subjected to silica gel column chromatography to obtain compound 19 (0.019g, 0.032mmol) in 21% yield.
ESI-MS[M+H] + (m/z):600.2.
Example 20
Example 20 of the present application provides 4- (cyclopropylmethyl) -1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 20) and a process for its preparation, compound 20 having the formula:
Figure BDA0002520468220000532
cyclopropylcarboxaldehyde (0.4 mmol) and intermediate 18-2 from example 18 (0.2 mmol) were placed in a round bottom flask, dichloromethane was added and cooled to 0 ℃ after which sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added, the reaction stirred at room temperature for 24h, TLC checked for completion, the reaction was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and the product, compound 20 (0.071g, 0.134mmol) was obtained in 67% yield by silica gel stirring and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ7.88–7.80(m,1H),7.61(dd,J=5.0,3.8 Hz,1H),7.20(s,1H),7.12(t,J=3.7Hz,2H),4.70(s,2H),3.92–3.84(m,4H), 3.76–3.69(m,4H),3.30(t,J=5.4Hz,2H),3.19(p,J=7.5Hz,4H),2.64(t,J =5.4Hz,2H),2.18(d,J=6.6Hz,2H),1.28(t,J=7.5Hz,3H),0.75–0.66(m, 1H),0.42(q,J=5.6Hz,2H),-0.01(q,J=4.9Hz,2H).
13 C NMR(100MHz,CDCl 3 )δ167.54,162.37,158.56,156.91,153.35, 147.59,142.57,134.57,124.30,122.95,122.76,119.10,113.21,112.49,66.72, 62.57,57.33,49.77,46.58,46.49,45.28,23.67,12.28,8.13,3.88.
ESI-MS[M+H] + (m/z):532.3.
Example 21
Example 21 provides 4-cyclopentyl-1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 21) and a method for its preparation, compound 21 having the formula:
Figure BDA0002520468220000541
cyclopentanone (0.4 mmol) and intermediate 18-2 (0.2 mmol) from example 18 were placed in a round-bottomed flask, dichloromethane (5 mL) was added and cooled to 0 deg.C, then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, the reaction was completed by TLC, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and the product compound 21 (0.043 g, 0.079mmol) was obtained as a product by stirring with silica gel and column chromatography, with a yield of 39%.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.95(m,1H),7.79–7.72(m,1H), 7.34(s,1H),7.27(dd,J=4.4,2.7Hz,2H),4.83(s,2H),4.08–3.97(m,4H), 3.93–3.81(m,4H),3.42(t,J=5.4Hz,2H),3.39–3.29(m,4H),2.75(t,J= 5.4Hz,2H),2.62-2.52(m,1H),1.93–1.80(m,2H),1.71(dd,J=6.8,5.1Hz, 2H),1.58(dd,J=7.5,4.8Hz,2H),1.49–1.37(m,5H).
13 C NMR(100MHz,CDCl 3 )δ167.56,162.36,158.56,153.34,147.52, 142.53,124.30,122.95,122.75,119.08,113.22,112.51,66.70,66.58,56.71, 48.73,46.62,46.48,45.22,30.35,24.05,23.65,12.26.
ESI-MS[M+H] + (m/z):546.3.
Example 22
Example 22 of the present application provides 4-cyclohexyl-1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 22) and a process for its preparation, compound 22 having the following structural formula:
Figure BDA0002520468220000551
cyclohexanone (0.4 mmol) and intermediate 18-2 from example 18 (0.2 mmol) were placed in a round bottom flask, dichloromethane (5 mL) was added and cooled to 0 deg.C, then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, TLC checked for completion, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and the product, compound 22 (0.049g, 0.088mmol) was obtained by silica gel stirring and column chromatography in 44% yield.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.95(m,1H),7.79–7.71(m,1H), 7.34(s,1H),7.29–7.24(m,2H),4.82(s,2H),4.06–3.97(m,4H),3.90–3.83 (m,4H),3.42–3.29(m,6H),2.78(t,J=5.4Hz,2H),2.35–2.27(m,1H), 1.87-1.76(m,4H),1.42(t,J=7.5Hz,3H),1.29-1.19(m,6H).
13 C NMR(100MHz,CDCl 3 )δ168.12,162.36,158.55,156.91,153.31, 147.69,142.54,134.55,124.23,122.93,122.73,119.05,113.22,112.49,66.69, 62.29,53.66,47.00,46.47,46.01,45.24,28.63,26.06,25.45,23.64,12.27.
ESI-MS[M+H] + (m/z):560.3.
Example 23
Example 23 of the present application provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (1, 4-dioxospiro [4.5] decan-8-yl) piperazin-2-one (compound 23) and a method for its preparation, compound 23 having the following structural formula:
Figure BDA0002520468220000561
1, 4-Dioxospiro [4.5] decan-8-one (0.8 mmol) and intermediate 18-2 of example 18 (0.5 mmol) were placed in a round bottom flask, dichloromethane (5 mL) was added and cooled to 0 ℃ after which sodium acetate (0.5 mmol) and sodium triacetoxyborohydride (1.0 mmol) were added, the reaction stirred at room temperature for 24h, TLC check for completion of the reaction, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and product 23 (0.138g, 0.224mmol) was obtained by stirring with silica gel and column chromatography with a yield of 47%.
1 H NMR(400MHz,CDCl 3 )δ8.06–7.91(m,1H),7.79–7.68(m,1H), 7.34(s,1H),7.27(dd,J=6.5,2.5Hz,2H),4.82(s,2H),4.07–3.97(m,4H), 3.93(s,4H),3.90–3.82(m,4H),3.41–3.29(m,6H),2.78(t,J=5.3Hz,2H), 2.44–2.33(m,1H),1.80(t,J=7.9Hz,4H),1.67–1.49(m,4H),1.42(t,J= 7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ167.89,162.34,158.54,156.90,153.28, 147.61,142.49,134.53,124.25,122.93,122.73,119.01,113.23,112.49,108.17, 66.67,64.34,64.26,60.47,54.05,46.98,46.46,46.00,45.23,33.14,25.36, 23.62,12.26.
ESI-MS[M+H] + (m/z):618.3.
Example 24
Example 24 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (4-oxocyclohexyl) piperazin-2-one (compound 24) and a process for its preparation, compound 24 having the formula:
Figure BDA0002520468220000571
compound 23 (0.080g, 0.13mmol) from example 23 was placed in a round-bottom flask, dichloromethane (5 mL) was added and cooled to 0 ℃, after which THF (10 mL) and concentrated hydrochloric acid (2 mL) were added, stirred at reflux temperature for 4h, the reaction was checked by tlc for completion, concentrated under reduced pressure, and then stirred on silica gel and column chromatographed to give compound 24 (0.061g, 0.106mmol), a yield of 82%.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.94(m,1H),7.76(dd,J=5.9,2.9 Hz,1H),7.36(s,1H),7.30–7.26(m,2H),4.86(s,2H),4.08–3.96(m,4H), 3.92–3.81(m,4H),3.49-3.38(m,4H),3.34(q,J=7.5Hz,2H),2.87–2.80(m, 2H),2.74(s,1H),2.54–2.45(m,2H),2.38–2.28(m,2H),2.05(d,J=11.1Hz, 2H),1.95–1.85(m,2H),1.43(t,J=7.5Hz,3H).
ESI-MS[M+H] + (m/z):574.3.
Example 25
Example 25 of the present application provides 4- (4, 4-difluorocyclohexyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 25) and methods of making the same, compound 25 having the following structural formula:
Figure BDA0002520468220000581
4, 4-Difluorocyclohexanone (0.4 mmol) and intermediate 18-2 (0.5 mmol) from example 18 were placed in a round-bottomed flask, dichloromethane (5 mL) was added and cooled to 0 deg.C, and then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, the reaction was completed by TLC, the reaction solution was extracted with dichloromethane and water, concentrated under reduced pressure by organic phase, and then subjected to silica gel stirring and column chromatography to give product compound 25 (0.064g, 0.108mmol) in 54% yield.
1 H NMR(400MHz,CDCl 3 )δ8.03–7.95(m,1H),7.78–7.72(m,1H), 7.35(s,1H),7.30–7.23(m,2H),4.84(s,2H),4.07–3.98(m,4H),3.92–3.82 (m,4H),3.44–3.38(m,2H),3.34(dd,J=14.0,6.5Hz,4H),2.82–2.74(m, 2H),2.44(t,J=8.7Hz,1H),2.19–2.07(m,2H),1.87–1.63(m,6H),1.43(t,J =7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ167.48,162.37,158.57,156.90,153.38, 147.39,142.57,134.56,124.35,122.96,122.77,119.11,113.20,112.48,66.70, 59.42,54.14,46.86,46.48,46.08,45.24,31.86,24.35,23.67,12.27.
ESI-MS[M+H] + (m/z):596.3.
Example 26
Example 26 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (4-hydroxycyclohexyl) piperazin-2-one (compound 26) and a process for its preparation, compound 26 having the following structural formula:
Figure BDA0002520468220000591
compound 24 (0.058 g, 0.1mmol) from example 24 was placed in a round bottom flask, methanol (3 mL) was added and cooled to 0 ℃, then sodium borohydride (0.2 mmol) was added thereto, slowly warmed to room temperature and stirred for 2h, the reaction was complete by tlc, concentrated under reduced pressure, and stirred over silica gel and column chromatography to give compound 26 (0.039g, 0.068mmol) in 68% yield.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.92(m,1H),7.80–7.69(m,1H), 7.34(s,1H),7.27(dd,J=8.7,4.8Hz,2H),4.82(s,2H),4.12–3.97(m,4H), 3.93–3.82(m,4H),3.56(d,J=4.2Hz,1H),3.43–3.28(m,6H),2.99(q,J= 7.3Hz,1H),2.83–2.71(m,2H),2.37–2.26(m,1H),2.01(s,2H),1.88(s,2H), 1.42(t,J=7.5Hz,3H),1.37–1.21(m,4H).
13 C NMR(100MHz,CDCl 3 )δ167.86,162.33,158.54,156.90,153.28, 147.57,142.48,134.53,124.25,122.96,122.76,119.02,113.20,112.49,69.93, 66.68,61.36,53.86,46.46,46.29,45.23,34.08,31.49,26.26,23.60,12.28.
ESI-MS[M+H] + (m/z):576.3.
Example 27
Example 27 of the present application provides 4- (4- (dimethylamino) cyclohexyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 27) and a process for its preparation, compound 27 having the following structural formula:
Figure BDA0002520468220000601
dimethylamine hydrochloride (02 mmol) and the compound 24 (0058g, 01mmol) of example 24 were placed in a round-bottom flask, dichloromethane (5 mL) was added thereto and cooled to 0 ℃ and then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.3 mmol) were added thereto, the reaction was stirred at room temperature for 24h, TLC was carried out for completion of the reaction, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then, the product 27 (0.021g, 0.035 mmol) was obtained in 35% yield by silica gel stirring and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ8.01–7.93(m,1H),7.77–7.69(m,1H), 7.35(d,J=3.9Hz,1H),7.27(dd,J=9.2,5.4Hz,2H),4.84(d,J=6.7Hz,2H), 4.03(d,J=4.6Hz,4H),3.95–3.81(m,4H),3.46(s,1H),3.45–3.26(m,6H), 3.06(q,J=7.3Hz,1H),2.77(d,J=4.9Hz,2H),2.59(d,J=5.2Hz,6H),1.91 –1.75(m,2H),1.42(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ167.51,162.37,158.54,156.90,153.31, 147.44,142.51,134.53,124.30,122.95,122.75,119.03,113.21,112.43,66.68, 63.90,57.32,54.89,46.47,40.43,26.79,26.02,25.98,23.62,22.17,12.26,8.74.
ESI-MS[M+H] + (m/z):603.4.
Example 28
Example 28 of this application provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (4- (pyrrolidin-1-yl) cyclohexyl) piperazin-2-one (compound 28) and a process for its preparation, compound 28 having the following structural formula:
Figure BDA0002520468220000611
tetrahydropyrrole (0.2 mmol) and the compound 24 from example 24 (0.058g, 0.1mmol) were placed in a round-bottomed flask, dichloromethane (5 mL) was added and cooled to 0 ℃ and then glacial acetic acid (0.2 mmol) and sodium triacetoxyborohydride (0.3 mmol) were added thereto, the reaction was stirred at room temperature for 24h, the reaction was completed by TLC, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then the product compound 28 (0.026g, 0.041mmol) was obtained in 41% yield by silica gel stirring and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ7.99(dd,J=6.3,2.8Hz,1H),7.78–7.68 (m,1H),7.34(s,1H),7.31–7.23(m,2H),4.83(d,J=5.2Hz,2H),4.09–3.98 (m,4H),3.92–3.82(m,4H),3.48–3.40(m,3H),3.36–3.29(m,4H),3.11– 2.92(m,4H),2.84–2.74(m,2H),1.99(s,7H),1.43(t,J=7.5Hz,3H),1.26(t, J=8.8Hz,4H).
13 C NMR(100MHz,CDCl 3 )δ167.69,162.38,158.55,156.92,153.25, 147.66,142.52,134.56,124.22,122.93,122.69,118.99,113.23,112.48,66.69, 62.81,54.99,51.48,51.44,46.90,46.46,45.73,45.14,25.26,23.60,23.44, 20.55,12.26.
ESI-MS[M+H] + (m/z):629.4.
Example 29
Example 29 of the present application provides ethyl 2- (4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) -3-oxopiperazin-1-yl) acetate (compound 29) and a process for its preparation, compound 29 having the following structural formula:
Figure BDA0002520468220000621
intermediate 18-2 (0.2 mmol) obtained in example 18 was placed in a round-bottom flask, potassium carbonate (0.5 mmol), acetonitrile (5 mL) and ethyl bromoacetate (0.4 mmol) were added thereto, the mixture was stirred at reflux temperature (85 ℃) for 2 hours, after completion of the reaction was detected by tlc, the solvent was distilled off under reduced pressure, water and ethyl acetate were added to the residue to extract a water layer free from the product, the organic phase was washed with saturated saline, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography with silica gel to give the objective compound 29 (0.087g, 0.155mmol) in 77% yield.
1 H NMR(400MHz,CDCl 3 )δ7.95–7.88(m,1H),7.71–7.63(m,1H), 7.26(s,1H),7.19(dd,J=8.4,4.3Hz,2H),4.76(s,2H),4.12(q,J=7.1Hz,2H), 4.01–3.90(m,4H),3.82–3.77(m,4H),3.37(d,J=8.4Hz,4H),3.29–3.19 (m,4H),2.84(t,J=5.3Hz,2H),1.35(t,J=7.5Hz,3H),1.19(d,J=7.1Hz, 3H).
ESI-MS[M+H] + (m/z):564.3.
Example 30
Example 30 of the present application provides 2- (4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -3-oxopiperazin-1-yl) acetamide (compound 30) and a method for its preparation, compound 30 having the following structural formula:
Figure BDA0002520468220000631
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottom flask, potassium carbonate (0.5 mmol), acetonitrile (5 mL) and iodoacetamide (0.4 mmol) were added thereto, the mixture was slowly warmed to 80 ℃ and stirred for 2h, after completion of TLC detection reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated saline, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give compound 30 (0.069 g,0.129 mmol) as an aimed product in 65% yield.
1 H NMR(400MHz,MeOD)δ7.94(d,J=7.1Hz,1H),7.61(d,J=7.0Hz, 1H),7.35(s,1H),7.28–7.19(m,2H),4.86(s,4H),3.94(s,4H),3.79(s,4H), 3.50(s,2H),3.36-3.22(m,4H),3.14(s,2H),2.83(s,2H),1.32(t,J=7.4Hz, 3H).
13 C NMR(100MHz,MeOD)δ173.42,167.73,161.89,158.41,156.99, 152.74,148.20,141.41,134.08,123.62,123.05,122.84,117.78,113.40,112.44, 66.22,59.09,56.35,49.39,46.36,46.30,44.75,22.98,11.35.
ESI-MS[M+H] + (m/z):535.
Example 31
Example 31 provides 4-acetyl-1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 31) and a process for its preparation, compound 31 having the formula:
Figure BDA0002520468220000641
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round bottom flask, dichloromethane (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 deg.C, acetic anhydride (0.3 mmol) was slowly added dropwise thereto, followed by warming to room temperature and stirring for 2h, after completion of TLC detection reaction, solvent was distilled off under reduced pressure, water and dichloromethane were added to the residue to extract an aqueous layer free of product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give the objective product compound 31 (0.073g, 0.141mmol) in 70% yield.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.96(m,1H),7.78–7.71(m,1H), 7.36(d,J=7.8Hz,1H),7.29–7.23(m,2H),4.87(s,2H),4.29(d,J=43.2Hz, 2H),4.09–3.97(m,4H),3.93–3.65(m,6H),3.47(s,2H),3.34(q,J=7.5Hz, 2H),2.12(d,J=3.6Hz,3H),1.43(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ168.95,164.86,162.36,158.53,156.90, 153.43,146.46,142.54,134.51,124.75,124.59,122.98,122.80,119.07,113.25, 66.66,49.69,46.46,45.86,45.73,38.55,23.69,21.35,12.26.
ESI-MS[M+H] + (m/z):520.3.
Example 32
Example 32 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (2-hydroxypropionyl) piperazin-2-one (compound 32) and a process for its preparation, compound 32 having the formula:
Figure BDA0002520468220000651
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, L-lactic acid (0.3 mmol) was slowly added dropwise thereto at room temperature, HOBT (0.3 mmol) and EDCI (0.4 mmol) were then added thereto, stirring was carried out at room temperature for 12h, the solvent was distilled off under reduced pressure after completion of the TLC detection reaction, water and methylene chloride were added to the residue to extract a water layer free from the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give the objective product compound 32 (0.059 g, 0.107 mmol) in 54% yield.
1 H NMR(400MHz,CDCl 3 )δ8.01–7.94(m,1H),7.74(dd,J=5.6,3.2 Hz,1H),7.37(s,1H),7.30–7.23(m,2H),4.87(d,J=4.4Hz,2H),4.51-4.47 (m,2H),4.28(d,J=18.1Hz,1H),4.05–3.96(m,4H),3.90–3.83(m,4H), 3.69(s,2H),3.55–3.39(m,2H),3.33(q,J=7.5Hz,2H),1.42(t,J=7.5Hz, 3H),1.35(d,J=6.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ175.18,173.78,162.33,158.53,156.91, 153.45,146.25,142.50,134.50,124.84,123.01,122.84,119.07,113.24,112.37, 66.66,64.59,48.46,46.47,45.76,45.59,39.65,23.67,21.02,12.28.
ESI-MS[M+H] + (m/z):550.3.
Example 33
Example 33 provides 4- (cyclopropylformyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 33) and a process for its preparation, compound 33 having the formula:
Figure BDA0002520468220000661
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and then cyclopropylcarbonyl chloride (0.3 mmol) was added dropwise thereto slowly, followed by warming to room temperature and stirring for 2h, after completion of the TLC detection reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free from the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and then subjected to column chromatography over silica gel to give the desired product, compound 33 (0.078 g, 0.143mmol), in a yield of 72%.
1 H NMR(400MHz,CDCl 3 )δ7.96–7.87(m,1H),7.72–7.63(m,1H), 7.29(s,1H),7.19(dd,J=8.9,5.2Hz,2H),4.81(s,2H),4.38(s,2H),3.95(d,J =4.6Hz,4H),3.81(d,J=4.5Hz,6H),3.40(s,2H),3.26(q,J=7.4Hz,2H), 1.60(td,J=7.8,4.0Hz,1H),1.35(t,J=7.4Hz,3H),1.00–0.88(m,2H),0.77 (s,2H).
13 C NMR(100MHz,CDCl 3 )δ172.51,165.24,162.36,158.55,156.91, 153.46,146.62,142.56,134.53,124.68,122.98,122.80,119.10,113.23,66.68, 50.66,49.06,46.48,45.72,39.12,23.70,12.27,11.15,8.22.
ESI-MS[M+H] + (m/z):546.3.
Example 34
Example 34 of the present application provides 4- (cyclobutylformyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 34) and a method for its preparation, compound 34 having the following structural formula:
Figure BDA0002520468220000671
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ cyclobutylcarbonyl chloride (0.3 mmol) was added dropwise slowly, followed by warming to room temperature and stirring for 2h, TLC detection was performed after completion of the reaction and then the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract to an aqueous layer without the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give the objective product compound 34 (0.072g, 0.129mmol) in 64% yield.
1 H NMR(400MHz,CDCl 3 )δ7.93–7.84(m,1H),7.69–7.62(m,1H), 7.30(s,1H),7.18(dd,J=8.9,5.2Hz,2H),4.82(s,2H),4.39(s,2H),3.97(d,J =4.6Hz,4H),3.82(d,J=4.5Hz,6H),3.39(s,2H),3.25(q,J=7.4Hz,2H), 2.90-2.85(m,1H),,2.35-2.27(m,2H),2.15-1.95(m,4H),1.35(t,J=7.4Hz, 3H).
13 C NMR(100MHz,CDCl 3 )δ165.32,162.16,162.04,158.48,156.92, 153.05,146.76,141.78,134.20,124.55,124.41,123.20,123.03,118.56,113.04, 66.59,46.40,41.92,38.84,37.03,25.19,24.83,23.20,18.23,17.79,12.10.
ESI-MS[M+H] + (m/z):560.3.
Example 35
Example 35 provides 4- (cyclopentacacyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 35) and a method for its preparation, compound 35 having the formula:
Figure BDA0002520468220000681
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and then cyclopentylcarbonyl chloride (0.3 mmol) was slowly added dropwise thereto, followed by warming to room temperature and stirring for 2h, detection by TLC after completion of the reaction, distillation under reduced pressure of the solvent, extraction of the residue to dryness by addition of water and methylene chloride to an aqueous layer, washing of the organic phase with saturated brine, drying of the organic phase over anhydrous magnesium sulfate, concentration and chromatography over silica gel column to give the objective compound 35 (0.065g, 0.113mmol) in a yield of 57%.
1 H NMR(400MHz,CDCl 3 )δ8.02–7.96(m,1H),7.78–7.72(m,1H), 7.36(s,1H),7.27(dt,J=5.6,1.9Hz,2H),4.88(s,2H),4.39–4.27(m,2H), 4.06–3.98(m,4H),3.91–3.80(m,6H),3.45(s,2H),3.34(q,J=7.5Hz,2H), 2.91–2.81(m,1H),1.84–1.72(m,6H),1.60(d,J=6.8Hz,2H),1.43(t,J= 7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ174.78,165.23,158.55,156.90,153.48, 146.65,142.59,134.54,124.69,122.97,122.80,119.12,113.23,66.68,49.23, 46.49,45.94,45.74,41.27,39.03,30.02,26.04,23.70,12.27.
ESI-MS[M+H] + (m/z):574.3.
Example 36
Example 36 of the present application provides 4- (cyclohexanoyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthio [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 36) and a process for its preparation, compound 36 having the formula:
Figure BDA0002520468220000691
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and cyclohexylcarbonyl chloride (0.3 mmol) was added dropwise slowly, then warmed to room temperature and stirred for 2h, TLC detection was performed after completion of the reaction and the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract the aqueous layer without the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give the objective compound 36 (0.061g, 0.104mmol) in 52% yield.
1 H NMR(400MHz,CDCl 3 )δ7.98(dd,J=5.8,3.2Hz,1H),7.77(dd,J=5.6,3.1Hz,1H),7.36(s,1H),7.28(dd,J=9.2,5.3Hz,2H),4.87(s,2H),4.30 (s,2H),4.08–3.96(m,4H),3.93–3.79(m,6H),3.44(s,2H),3.34(q,J=7.5 Hz,2H),2.44(t,J=10.9Hz,1H),1.86–1.45(m,10H),1.42(t,J=7.5Hz, 3H).
13 C NMR(100MHz,CDCl 3 )δ174.70,165.25,162.35,158.53,156.94, 153.38,146.62,142.30,134.43,124.68,123.04,122.88,119.03,113.21,66.67, 49.12,46.47,45.94,45.72,43.04,40.72,38.82,29.04,25.67,25.48,23.57, 12.30.
ESI-MS[M+H] + (m/z):588.3.
Example 37
Example 37 of the present application provides 4-acryloyl-1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 37) and a process for its preparation, compound 37 having the following structural formula:
Figure BDA0002520468220000701
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ acryloyl chloride (0.4 mmol) was slowly added dropwise thereto, followed by warming to room temperature and stirring for 2h, after TLC detection reaction was completed, solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give the desired product compound 37 (0.064g, 0.121mmol) in a yield of 60%.
1 H NMR(400MHz,CDCl 3 )δ8.03–7.95(m,1H),7.78–7.71(m,1H), 7.36(s,1H),7.31–7.24(m,2H),6.51(dd,J=16.7,10.3Hz,1H),6.38(d,J= 15.8Hz,1H),5.80(d,J=8.7Hz,1H),4.87(s,2H),4.35(s,2H),4.12–3.97(m, 4H),3.97–3.76(m,6H),3.48(t,J=5.3Hz,2H),3.34(q,J=7.5Hz,2H),1.43 (t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ164.95,162.34,158.54,156.90,153.46, 146.53,142.59,134.54,129.88,126.27,124.71,122.96,122.78,119.09,113.25, 112.39,77.42,77.10,76.78,66.66,53.46,46.47,45.79,23.71,12.27,8.65.
ESI-MS[M+H] + (m/z):532.3.
Example 38
Example 38 of the present application provides tert-butyl 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -3-oxopiperazine-1-carbonate (compound 38) and a method for its preparation, compound 38 having the following structural formula:
Figure BDA0002520468220000711
placing N-boc-piperazinone (1.505g, 7.5 mmol) in a three-neck bottle, adding THF (5 mL) and t-BuOK (10 mmol) at 0 ℃, heating to 30-32 ℃, reacting and stirring for 20min, adding the intermediate 1-5 (2.070g, 5 mmol) of example 1 in portions, gradually changing the system color from light yellow to black, reacting for 4-5h at 30-32 ℃, detecting by TLC that the reaction is basically complete (EA), cooling the reaction liquid, adding water and ethyl acetate, extracting until no product exists in a water layer, washing an organic phase with saturated saline water, drying the organic phase with anhydrous magnesium sulfate, and concentrating to dryness to obtain a crude compound. The crude product is stirred by silica gel and then subjected to column chromatography to obtain a target product (2.388g, 4.139mmol), and the yield is 83%.
1 H NMR(400MHz,CDCl 3 )δ7.94–7.90(m,1H),7.71–7.66(m,1H), 7.29(s,1H),7.19(dd,J=3.5,2.1Hz,2H),4.81(s,2H),4.12(s,2H),3.99– 3.93(m,4H),3.84–3.78(m,4H),3.61(t,J=5.3Hz,2H),3.38(t,J=5.1Hz, 2H),3.27(q,J=7.5Hz,2H),1.40(s,9H),1.36(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ165.05,161.36,157.54,155.89,152.42, 145.94,141.53,133.51,123.53,121.96,121.78,118.09,112.19,98.97,80.15, 65.67,45.47,45.28,44.70,28.68,27.30,22.66,11.25.
ESI-MS[M+H] + (m/z):578.3.
Example 39
Example 39 of the present application provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (methylsulfonyl) piperazin-2-one (compound 39) and a method of making the same, compound 39 having the following structural formula:
Figure BDA0002520468220000721
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and methanesulfonyl chloride (0.3 mmol) was added dropwise slowly thereto, followed by warming to room temperature and stirring for 2h, TLC detection was performed after completion of the reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract to an aqueous layer without the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography with silica gel to give target product compound 39 (0.085 g, 0.153mmol) in 77% yield.
1 H NMR(400MHz,CDCl 3 )δ7.92(dd,J=5.9,3.2Hz,1H),7.68(dd,J= 5.8,3.1Hz,1H),7.29(s,1H),7.21–7.17(m,2H),4.81(s,2H),3.96(d,J=7.0 Hz,6H),3.85–3.75(m,4H),3.47(s,4H),3.27(q,J=7.5Hz,2H),2.81(s,3H), 1.36(t,J=7.5Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ164.14,162.32,158.56,156.91,153.49, 146.34,142.58,134.53,124.77,122.99,122.82,119.13,113.23,112.44,66.69, 48.69,46.54,46.49,45.70,42.72,36.19,23.71,12.27.
ESI-MS[M+H] + (m/z):556.2.
Example 40
Example 40 of the present application provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (ethylsulfonyl) piperazin-2-one (compound 40) and a method for its preparation, compound 40 having the following structural formula:
Figure BDA0002520468220000731
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round bottom flask, dichloromethane (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ ethylsulfonyl chloride (0.3 mmol) was added dropwise slowly, after warming to room temperature and stirring for 2h, after completion of TLC detection reaction, solvent was distilled off under reduced pressure, water and dichloromethane were added to the residue to extract an aqueous layer free of product, an organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give target product compound 40 (0.072g, 0.126mmol) in 63% yield.
1 H NMR(400MHz,CDCl 3 )δ7.99(d,J=5.9Hz,1H),7.75(d,J=5.7Hz, 1H),7.36(s,1H),7.27(d,J=5.2Hz,2H),4.87(s,2H),4.05(d,J=15.9Hz, 6H),3.88(s,4H),3.65–3.44(m,4H),3.34(q,J=7.2Hz,2H),3.03(q,J=7.2 Hz,2H),1.46–1.34(m,6H).
13 C NMR(100MHz,CDCl 3 )δ164.42,162.32,158.55,156.92,153.46, 146.50,142.58,134.54,124.70,122.98,122.80,119.11,113.24,112.44,66.69, 48.66,46.88,46.49,45.73,45.41,42.85,23.70,12.27,7.91.
ESI-MS[M+H] + (m/z):570.2.
Example 41
Example 41 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (propanesulfonyl) piperazin-2-one (compound 41) and a method for its preparation, compound 41 having the formula:
Figure BDA0002520468220000741
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round bottom flask, dichloromethane (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃, propylsulfonyl chloride (0.3 mmol) was slowly added dropwise thereto, followed by warming to room temperature and stirring for 2h, after completion of the tlc detection reaction, the solvent was distilled off under reduced pressure, water and dichloromethane were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give target product compound 41 (0.075g, 0.129mmol) in 64% yield.
1 H NMR(400MHz,CDCl 3 )δ8.03–7.96(m,1H),7.78–7.72(m,1H), 7.36(s,1H),7.27(dd,J=6.3,2.6Hz,2H),4.86(s,2H),4.03(dd,J=10.3,5.8 Hz,6H),3.94–3.82(m,4H),3.56(d,J=5.1Hz,2H),3.52(d,J=5.1Hz,2H), 3.34(q,J=7.5Hz,2H),3.00–2.90(m,2H),1.84(dd,J=15.4,7.6Hz,2H), 1.43(t,J=7.5Hz,3H),1.06(t,J=7.4Hz,3H).
13 C NMR(100MHz,CDCl 3 )δ164.43,162.30,158.53,156.92,153.41, 146.52,142.49,134.51,124.67,123.00,122.82,119.06,113.26,112.45,66.67, 52.40,48.61,46.83,46.47,45.71,42.74,23.67,16.94,13.01,12.27.
ESI-MS[M+H] + (m/z):584.2.
Example 42
Example 42 of the present application provides 4- (cyclopropylsulfonyl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 42) and a method for its preparation, compound 42 having the following structural formula:
Figure BDA0002520468220000751
intermediate 18-2 (0.2 mmol) from example 18 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ and cyclopropylsulfonyl chloride (0.3 mmol) was slowly added dropwise thereto, followed by warming to room temperature and stirring for 2h, detection by TLC after completion of the reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give compound 42 (0.089g, 0.153mmol), which is the objective compound, in a yield of 77%.
1 H NMR(400MHz,CDCl 3 )δ8.05–7.93(m,1H),7.80–7.70(m,1H), 7.36(s,1H),7.31–7.22(m,2H),4.88(s,2H),4.16–3.95(m,6H),3.89(d,J= 4.3Hz,4H),3.62–3.50(m,4H),3.34(q,J=7.4Hz,2H),2.30(dd,J=8.2,3.7 Hz,1H),1.43(t,J=7.4Hz,3H),1.22(d,J=2.7Hz,2H),1.05(d,J=6.0Hz, 2H).
13 C NMR(100MHz,CDCl 3 )δ164.43,162.32,158.55,156.91,153.47, 146.44,142.58,134.54,124.70,122.99,122.81,119.12,113.23,112.42,66.69, 49.11,46.65,46.49,45.67,43.07,26.40,23.71,12.27,4.78.
ESI-MS[M+H] + (m/z):582.2.
Example 43
Example 43 provides 1- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (2, 2-trifluoroethyl) piperazin-2-one (compound 43) and a process for its preparation, compound 43 having the following structural formula:
Figure BDA0002520468220000761
placing N-boc-piperazinone (1.505g, 7.5 mmol) in a three-necked bottle, adding THF (5 mL) and t-BuOK (10 mmol) at 0 ℃, heating to 30-32 ℃, reacting and stirring for 20min, adding 4- (6- (chloromethyl) -2- (2-methyl-1H-benzimidazole-1-yl) thiophene [3,2-d ] pyrimidine-4-yl) morpholine (13-2) (5 mmol) in portions, gradually changing the system color from light yellow to black, reacting for 4-5H at 30-32 ℃, detecting the basic reaction completion (EA) by TLC, adding water and ethyl acetate into the cooled reaction liquid, extracting until no product exists in an aqueous layer, washing an organic phase by using saturated saline, drying the organic phase by using anhydrous magnesium sulfate, and concentrating to obtain an intermediate, namely, tert-butyl 4- ((2- (2-methyl-1H-benzimidazole-1-yl) -4-morpholinothiophene [3,2-d ] pyrimidine-6-yl) methyl) -3-oxo-piperazine-1-formic ester crude product. The crude product is subjected to silica gel sample mixing and column chromatography to obtain tert-butyl 4- ((2- (2-methyl-1H-benzimidazole-1-yl) -4-morpholinyl thiophene [3,2-d ] pyrimidine-6-yl) methyl) -3-oxo-piperazine-1-formic ester, and the yield is 78%.
Placing an intermediate tert-butyl 4- ((2- (2-methyl-1H-benzimidazole-1-yl) -4-morpholinyl thiophene [3,2-d ] pyrimidine-6-yl) methyl) -3-oxopiperazine-1-formate (4.000 mmol) in a three-neck flask, adding TFA (20 mL) at 0 ℃, slowly heating to room temperature, reacting and stirring for 2H, then distilling off the solvent under reduced pressure, then adding methanol (20 mL) to the residual oily substance, distilling off the solvent under reduced pressure, repeating the steps for three times, and drying the residual oily substance in vacuum to obtain a foamy pale yellow solid 1- ((2- (2-methyl-1H-benzimidazole-1-yl) -4-morpholinyl thiophene [3,2-d ] pyrimidine-6-yl) methyl) piperazine-2-one trifluoroacetate with the yield of 100%.
The intermediate 1- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinothiophene [3,2-d ] pyrimido-6-yl) methyl) piperazin-2-one trifluoroacetate (0.2 mmol) was placed in a three-necked flask, potassium carbonate (0.5 mmol), acetonitrile (5 mL) and trifluoroethyl trifluoromethanesulfonate (0.4 mmol) were added thereto, the mixture was slowly heated to reflux temperature (85 ℃) and stirred for 2H, after completion of the TLC detection reaction, the solvent was distilled off under reduced pressure, water and ethyl acetate were added to the residue to extract a water layer without a product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to obtain the target product 1- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinothiophene [3,2-d ] pyrimido-6-yl) methyl) -4- (2, 2-trifluoroethyl) piperazin-2-one (compound 43) (0.045g, 0.08342% yield).
1 H NMR(400MHz,CDCl 3 )δ8.07(dd,J=6.1,3.1Hz,1H),7.71(dd,J= 6.1,2.9Hz,1H),7.36(s,1H),7.31–7.27(m,2H),4.85(s,2H),4.07–3.99(m, 4H),3.92–3.84(m,4H),3.51(s,2H),3.47–3.41(m,2H),3.08(d,J=9.2Hz, 2H),2.98(t,J=5.3Hz,2H),2.93(s,3H).
13 C NMR(100MHz,CDCl 3 )δ166.32,162.32,158.54,153.53,152.28, 147.13,142.54,134.40,124.46,123.01,122.90,118.93,113.52,112.41,66.69, 57.14,56.89,50.07,46.52,46.41,45.28,17.79.
ESI-MS[M+H] + (m/z):546.2.
Example 44
Example 44 of the present application provides 1- ((2- (2-methyl-1H-benzoimidazol-1-yl) -4-morpholinylthiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (methylsulfonyl) piperazin-2-one (compound 44) and methods for its preparation, compound 44 having the following structural formula:
Figure BDA0002520468220000771
1- ((2- (2-methyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one trifluoroacetate (0.2 mmol) was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ and slow dropwise addition of methanesulfonyl chloride (0.3 mmol) thereto, followed by warming to room temperature and stirring for 2H, detection of reaction by TLC and evaporation of the solvent under reduced pressure, addition of water and methylene chloride to the residue and extraction until no product was present in the aqueous layer, washing of the organic phase with saturated brine, drying of the organic phase over anhydrous magnesium sulfate, concentration and column chromatography gave the target product compound 44 (0.085g, 0.157mmol) in a yield of 79%.
1 H NMR(400MHz,CDCl 3 )δ8.11–8.03(m,1H),7.75–7.68(m,1H), 7.37(s,1H),7.29(t,J=4.8Hz,2H),4.89(s,2H),4.04(d,J=6.2Hz,6H),3.93 –3.83(m,4H),3.59–3.47(m,4H),2.93(s,3H),2.89(s,3H).
13 C NMR(100MHz,CDCl 3 )δ164.13,162.29,158.53,153.62,152.28, 146.33,142.55,134.39,124.77,123.04,122.94,118.96,113.54,112.36,66.68, 48.70,46.53,45.71,42.73,36.24,17.86.
ESI-MS[M+H] + (m/z):542.2.
Example 45
Example 45 of the present application provides 4- (cyclopropylsulfonyl) -1- ((2- (2-methyl-1H-benzimidazol-1-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 45) and a method for its preparation, compound 45 having the following structural formula:
Figure BDA0002520468220000781
1- ((2- (2-methyl-1H-benzoimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one trifluoroacetate (0.2 mmol) was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ and dropwise addition of cyclopropylsulfonyl chloride (0.3 mmol) thereto, followed by warming to room temperature and stirring for 2H, detection of completion of the reaction by TLC and distillation of the solvent under reduced pressure, water and methylene chloride were added to the residue to extract a water layer until no product was present, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography to give the objective product compound 45 (0.081g, 0.143mmol) in 71% yield.
1 H NMR(400MHz,CDCl 3 )δ8.10–8.04(m,1H),7.74–7.68(m,1H), 7.37(s,1H),7.30–7.26(m,2H),4.87(s,2H),4.08(s,2H),4.06–4.00(m,4H), 3.92–3.84(m,4H),3.63–3.48(m,4H),2.93(s,3H),2.35–2.25(m,1H),1.24 –1.19(m,2H),1.08–1.01(m,2H).
13 C NMR(100MHz,CDCl 3 )δ164.43,162.27,158.51,153.56,152.29, 146.46,142.46,134.36,124.68,123.06,122.95,118.90,113.56,112.36,66.67, 49.11,46.64,46.51,45.66,43.07,26.42,17.81,4.78.
ESI-MS[M+H] + (m/z):568.2.
Example 46
Example 46 of the present application provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (1-methylpiperidin-4-yl) piperazin-2-one (compound 46) and a method for its preparation, compound 46 having the following structural formula:
Figure BDA0002520468220000791
n-methylpiperidin-4-one (0.4 mmol) and intermediate 18-2 (0.2 mmol) from example 18 were placed in a round-bottomed flask, dichloromethane (5 mL) was added and cooled to 0 ℃ and then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, the reaction was completed by TLC, the reaction mixture was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then the product compound 46 (0.031g, 0.054mmol) was obtained in 27% yield by stirring with silica gel and column chromatography.
1 H NMR(400MHz,Chloroform-d)δ8.07–7.94(m,1H),7.83–7.70(m, 1H),7.35(s,1H),7.33–7.22(m,2H),4.83(s,2H),4.03(t,J=6.0Hz,4H), 3.87(t,J=5.7Hz,4H),3.46–3.28(m,4H),3.27–3.12(m,2H),2.78(q,J= 5.4,2H),2.53–2.35(m,5H),2.02(s,3H),1.95-1.79(m,4H),1.42(t,J=7.5 Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ167.34,162.33,158.53,156.89,153.30, 147.35,142.46,134.51,124.32,122.97,122.77,119.03,113.20,112.46,66.68, 65.76,58.31,54.23,46.80,46.46,45.52,45.23,44.65,26.43,23.61,12.28.
ESI-MS[M+H] + (m/z):575.3.
Example 47
Example 47 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (tetrahydro-2H-pyran-4-yl) piperazin-2-one (compound 47) and a method for its preparation, compound 47 having the following structural formula:
Figure BDA0002520468220000801
tetrahydro-2H-pyran-4-one (0.4 mmol) and intermediate 18-2 (0.2 mmol) of example 18 were placed in a round bottom flask, dichloromethane (5 mL) was added and cooled to 0 deg.C, after which sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added, the reaction was stirred at room temperature for 24h, the reaction was complete by TLC, the reaction was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then product 47 (0.042g, 0.075 mmol) was obtained by silica gel stirring and column chromatography with a yield of 37%.
1 H NMR(400MHz,Chloroform-d)δ8.04–7.94(m,1H),7.84–7.75(m, 1H),7.35(s,1H),7.34–7.24(m,2H),4.84(s,2H),4.12–3.98(m,6H),3.88(t, J=5.8Hz,4H),3.50–3.29(m,8H),2.80(q,J=6.2Hz,2H),2.51(tt,J=11.3, 3.8Hz,1H),1.82–1.72(m,2H),1.66–1.50(m,2H),1.43(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ167.54,162.30,158.54,156.82,153.16, 147.51,141.71,134.30,124.32,123.19,123.02,118.85,113.25,112.62,67.00, 66.69,65.66,59.71,53.74,46.48,45.26,35.51,29.24,23.46,12.31.
ESI-MS[M+H] + (m/z):562.3.
Example 48
Example 48 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -4-morpholinothiophen [3,2-d ] pyrimidin-6-yl) methyl) -4- (tetrahydro-2H-thiopyran-4-yl) piperazin-2-one (compound 48) and a process for its preparation, compound 48 having the formula:
Figure BDA0002520468220000811
tetrahydro-2H-thiopyran-4-one (0.4 mmol) and intermediate 18-2 (0.2 mmol) of example 18 were placed in a round-bottomed flask, methylene chloride (5 mL) was added and cooled to 0 ℃ to which sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added, the reaction was stirred at room temperature for 24h, TLC check was completed, the reaction solution was extracted with methylene chloride and water, the organic phase was concentrated under reduced pressure, and then, by silica gel stirring and column chromatography, the product compound 48 (0.049g, 0.088mmol) was obtained in 44% yield.
1 H NMR(400MHz,Chloroform-d)δ8.02–7.95(m,1H),7.80–7.72(m, 1H),7.34(s,1H),7.32–7.24(m,2H),4.84(s,2H),4.03(t,J=5.8Hz,4H), 3.87(t,J=5.7Hz,4H),3.43–3.29(m,6H),2.80(t,J=5.4Hz,2H),2.78– 2.63(m,4H),2.41(tt,J=11.3,3.8Hz,1H),2.11(dd,J=13.0,3.4Hz,2H), 1.77–1.65(m,2H),1.43(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ167.77,162.35,158.56,156.89,153.34, 147.49,142.49,134.52,124.31,122.96,122.78,119.09,113.18,112.49,66.71, 61.42,53.21,47.01,46.48,45.60,45.28,29.97,28.36,23.64,12.28.
ESI-MS[M+H] + (m/z):578.2.
Example 49
Example 49 provides 4-cyclopentyl-1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (compound 49) and a method for its preparation, compound 49 having the following structural formula:
Figure BDA0002520468220000821
(1) Preparation of 4- (2-chloro-9-methyl-9H-purin-6-yl) morpholine (49-1)
Figure BDA0002520468220000822
Placing 2, 6-dichloropurine (18.900g, 100mmol) in a three-necked bottle, adding MeOH (500 mL), cooling the reaction solution to 0-5 ℃, dropwise adding morpholine (150 mmol), heating to 20-22 ℃ after dropwise adding, reacting for 2H, performing suction filtration, washing a filter cake with water (50 mL) and methanol (30 mL) in sequence, and performing vacuum drying at 45 ℃ to obtain the product 4- (2-chloro-9H-purin-6-yl) morpholine (21.384 g) with the yield of 90%.
ESI-MS[M+Na] + (m/z):262.2.
Putting 4- (2-chloro-9H-purin-6-yl) morpholine (11.950g, 50mmol) in a three-necked bottle, adding acetonitrile (300 mL) and cesium carbonate (75 mmol), cooling the reaction liquid to 0-5 ℃, slowly dropwise adding methyl iodide (150 mmol), heating to room temperature after dropwise adding, reacting for 4 hours, distilling off the solvent under reduced pressure, adding 500mL of dichloromethane into the residual solid, vigorously stirring for 1 hour, carrying out suction filtration, washing a filter cake with dichloromethane (50 mL), carrying out vacuum concentration on the filtrate, recrystallizing with ethyl acetate, and carrying out vacuum drying at 45 ℃ to obtain a product 49-1 (11.556 g, 45.675mmol) with the yield of 91%.
ESI-MS[M+Na] + (m/z):276.1.
(2) Preparation of 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-carbaldehyde (49-2)
Figure BDA0002520468220000831
Putting 49-1 (10.120g, 40mmol) in a three-neck flask, vacuumizing and introducing nitrogen for 3 times, adding ultra-dry THF, vacuumizing and introducing nitrogen for 1 time, cooling to-78 ℃, dropwise adding 2.5M n-BuLi (19.2mL, 48mmol), keeping the temperature of the reaction liquid below-75 ℃ and stirring at-78 ℃ for reaction for 1h, dropwise adding ultra-dry DMF, keeping the temperature at-78 ℃ and stirring for reaction for 3-4h (HPLC detection reaction is finished), heating to room temperature, slowly pouring the reaction liquid into a cooled saturated ammonium chloride aqueous solution, stirring at room temperature for 30min, extracting the reaction liquid with dichloromethane until no product exists in the aqueous layer, combining organic layers, drying with anhydrous magnesium sulfate, concentrating under reduced pressure to obtain a crude product 49-2 (7.010g, 24.947mmol), obtaining the yield of 62%, and directly using the crude product in the next reaction without purification.
ESI-MS[M+H] + (m/z):282.0.
(3) Preparation of 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol (49-3)
Figure BDA0002520468220000832
Putting the intermediate 49-2 (5.620g, 20mmol) into a three-necked bottle, adding methanol, cooling to about-10 ℃, adding sodium borohydride in portions, keeping the temperature of-10 ℃ after the addition to react for 1h, after the reaction is finished, dropwise adding 90mL of ice water, stirring at-10 ℃ for 10min, performing suction filtration, washing a filter cake with the ice water, and drying to obtain the alcohol 49-3 (5.502g, 19.442mmol) with the yield of 97%.
ESI-MS[M+Na] + (m/z):306.1.
(4) Preparation of 4- (2-chloro-8- (chloromethyl) -9-methyl-9H-purin-6-yl) morpholine (49-4)
Figure BDA0002520468220000841
Placing 49-3 (2.830g and 10mmol) in a three-necked bottle at room temperature, adding 100mL of dichloromethane, cooling to 0 ℃, slowly adding thionyl chloride (10 mL) into the three-necked bottle while stirring, heating to room temperature after the addition for reacting for 2h (TLC detection reaction is completed, n-hexane: EA = 2).
ESI-MS[M+H] + (m/z):302.1.
(5) Preparation of tert-butyl 4- ((2-chloro-9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -3-oxopiperazine-1-carboxylate (49-5)
Figure BDA0002520468220000842
Placing N-boc-piperazinone (1.505g, 7.5 mmol) in a three-neck bottle, adding THF (5 mL) and t-BuOK (10 mmol) at 0 ℃, heating to 30-32 ℃, reacting and stirring for 20min, adding an intermediate 49-4 (2.070g, 5 mmol) in portions, gradually changing the system color from light yellow to black, reacting for 4-5h at 30-32 ℃, detecting by TLC (detection of basic reaction (EA), evaporating the solvent under reduced pressure, extracting the residue with water and dichloromethane until no product exists in a water layer, washing an organic phase with saturated saline solution, drying the organic phase with anhydrous magnesium sulfate, and concentrating to dryness to obtain a crude compound 49-5. The crude product is mixed with silica gel and then subjected to column chromatography to obtain a target product 49-5 (1.909g, 4.097mmol), and the yield is 82%.
ESI-MS[M+H] + (m/z):466.2.
(6) Tert-butyl 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -3-oxopiperazine-1-carboxylate (49-6)
Figure BDA0002520468220000851
Compound 49-5 (1.864g, 4mmol), 2-ethylbenzimidazole (4.8 mmol), pd 2 (dba) 3 (0.4mmol),Xphos(0.8mmol),CsCO 3 (8 mmol) was placed in a three-necked flask, dioxane (50 mL) was added, nitrogen was introduced 3 times under vacuum, the temperature was raised to 110 ℃ for 4-5h, (DCM: EA =1 for basic completion of the reaction by TLC), the mixture was filtered while hot, the filter cake was washed with DCM, the filtrate was concentrated and stirred with silica gel, and column chromatography gave 49-6 (1.223g, 2.127mmol, 53% yield) as the target product.
ESI-MS[M+H] + (m/z):576.3.
(7) 1- ((2- (2-Ethyl-1H-benzimidazol-1-yl) -4-morpholinylthiophene [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one trifluoroacetate (49-7)
Figure BDA0002520468220000852
Putting the intermediate 49-6 (1.150g, 2mmol) in a three-necked flask, adding TFA (15 mL) at 0 ℃, slowly heating to room temperature, reacting and stirring for 2h, then distilling off the solvent under reduced pressure, then adding methanol (20 mL) to the residual oil, distilling off the solvent under reduced pressure, repeating the steps for three times, and drying the residual oil in vacuum to obtain a foamy light yellow solid 49-7 with the yield of 100%.
(8) 4-cyclopentyl-1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (Compound 49)
Figure BDA0002520468220000861
Cyclopentanone (0.4 mmol) and intermediate 49-7 (0.2 mmol) were placed in a round-bottom flask, dichloromethane (5 mL) was added and cooled to 0 deg.C, then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, TLC detected for completion, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then the product compound 49 (0.068g, 0.125mmol) was obtained in 63% yield by silica gel stirring and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ8.07–7.93(m,1H),7.84–7.68(m,1H), 7.32–7.19(m,2H),4.87(s,2H),4.48–4.19(s br,4H),3.92–3.79(m,7H), 3.49–3.24(m,6H),2.75(q,J=5.4,2H),2.57(t,J=7.6Hz,1H),1.98–1.78 (m,2H),1.78–1.65(m,2H),1.63–1.51(m,2H),1.50–1.35(m,5H).
13 C NMR(101MHz,CDCl 3 )δ167.15,156.95,153.67,153.49,150.78, 146.24,142.47,134.55,122.89,122.72,119.08,117.27,113.24,67.02,66.60, 56.77,48.66,46.52,45.82,41.89,30.37,29.33,24.05,23.77,12.39.
ESI-MS[M+H] + (m/z):544.3.
Example 50
Example 50 provides 4-cyclohexyl-1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (compound 50) and a method for its preparation, compound 50 having the following structural formula:
Figure BDA0002520468220000871
cyclohexanone (0.4 mmol) and intermediate 49-7 of example 49 (0.2 mmol) were placed in a round bottom flask, dichloromethane (5 mL) was added and cooled to 0 ℃, after which sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added, the reaction was stirred at room temperature for 24h, tlc checked for completion, the reaction solution was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and the product compound 50 (0.061g, 0.110mmol) was obtained with silica gel stirring and column chromatography in 55% yield.
1 H NMR(400MHz,CDCl 3 )δ8.04–7.96(m,1H),7.80–7.72(m,1H), 7.32–7.22(m,2H),4.87(s,2H),4.30(s br,4H),3.95–3.78(m,7H),3.45– 3.28(m,6H),2.76(q,J=5.4,2H),2.32(dd,J=13.5,6.1Hz,1H),1.95–1.78 (m,4H),1.44(t,J=7.5Hz,3H),1.35–1.05(m,6H).
13 C NMR(101MHz,CDCl 3 )δ167.66,156.93,153.58,153.49,150.74, 146.34,142.39,134.53,122.91,122.74,119.05,117.27,113.25,67.02,62.36, 53.67,46.86,45.90,41.98,29.31,28.60,26.04,25.43,23.75,12.39.
ESI-MS[M+H] + (m/z):558.3.
Example 51
Example 51 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (tetrahydro-2H-pyran-4-yl) piperazin-2-one (compound 51) and a method for its preparation, compound 51 having the following structural formula:
Figure BDA0002520468220000881
tetrahydro-2H-pyran-4-one (0.4 mmol) and intermediate 49-7 (0.2 mmol) of example 49 were placed in a round-bottom flask, dichloromethane (5 mL) was added and cooled to 0 ℃ and then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, the reaction was completed by TLC, the reaction mixture was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then the product compound 51 (0.048g, 0.086 mmol) was obtained in 43% yield by silica gel stirring and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ8.07–7.94(m,1H),7.83–7.70(m,1H), 7.37–7.20(m,2H),4.87(s,2H),4.31(s br,4H),4.03(dd,J=11.5,2.9Hz,2H), 3.95–3.76(m,7H),3.53–3.29(m,8H),2.79(q,J=5.4,2H),2.48(m,1H), 1.91–1.70(m,2H),1.55(m,2H),1.44(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ167.21,156.92,153.66,153.45,150.72, 146.21,142.25,134.47,122.95,122.87,118.98,117.28,113.26,66.99,65.69, 59.68,53.83,46.73,45.89,45.57,41.96,29.30,29.24,23.71,12.39.
ESI-MS[M+H] + (m/z):560.3.
Example 52
Example 52 of the present application provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (tetrahydro-2H-thiopyran-4-yl) piperazin-2-one (compound 52) and a method for its preparation, compound 52 having the following structural formula:
Figure BDA0002520468220000891
tetrahydro-2H-thiopyran-4-one (0.4 mmol) and intermediate 49-7 of example 49 (0.2 mmol) were placed in a round bottom flask, dichloromethane (5 mL) was added and cooled to 0 deg.C, to which was added sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol), the reaction was stirred at room temperature for 24h, the reaction was complete by TLC, the reaction was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then the product compound 52 (0.072g, 0.125mmol) was obtained in 63% yield by silica gel stirring and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ8.00(d,J=6.8Hz,1H),7.76(d,J=6.6Hz, 1H),7.32(d,J=32.2Hz,2H),4.87(s,2H),4.35(s br,4H),3.95–3.75(m,7H), 3.45–3.30(m,6H),2.80–2.57(m,6H),2.47–2.32(m,1H),2.21–2.02(m, 2H),1.72-1.64(m,2H),1.44(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ167.44,156.90,153.66,153.44,150.65, 146.29,142.00,134.40,122.94,122.86,118.92,117.31,113.28,67.00,61.39, 53.23,46.95,45.64,45.43,42.01,29.93,29.29,28.34,23.65,12.42.
ESI-MS[M+H] + (m/z):576.3.
Example 53
Example 53 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (1-methylpiperidin-4-yl) piperazin-2-one (compound 53) and a method for its preparation, compound 53 having the following structural formula:
Figure BDA0002520468220000901
4-methylpiperidin-4-one (0.4 mmol) and intermediate 49-7 (0.2 mmol) from example 49 were placed in a round-bottomed flask, dichloromethane (5 mL) was added and cooled to 0 ℃ and then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, the reaction was completed by TLC, the reaction mixture was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and then the product compound 53 (0.041g, 0.072mmol) was obtained in 36% yield by stirring with silica gel and column chromatography.
1 H NMR(400MHz,CDCl 3 )δ8.01(d,J=4.7Hz,1H),7.74(d,J=4.5Hz, 1H),7.27(dd,J=8.8,5.3Hz,2H),4.86(s,2H),3.95-3.75(m,7H),3.44(d,J= 13.1Hz,4H),3.33(d,J=12.5Hz,4H),3.00(d,J=10.9Hz,2H),2.77(q,J= 5.4,2H),2.35(s,5H),2.12(t,J=10.3Hz,3H),1.84(d,J=9.8Hz,2H),1.68 (dd,J=30.0,19.6Hz,2H),1.41(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ167.27,156.94,153.65,153.43,150.73, 146.20,142.43,134.52,122.89,122.79,119.00,117.26,113.24,66.99,59.41, 54.42,54.15,46.79,45.90,45.53,45.49,41.94,29.27,27.28,23.73,12.38.
ESI-MS[M+H] + (m/z):573.3.
Example 54
Example 54 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (1, 4-dioxaspiro [4.5] decan-8-yl) piperazin-2-one (compound 54), and a method for its preparation, compound 54 having the following structural formula:
Figure BDA0002520468220000911
1, 4-Dioxospiro [4.5] decan-8-one (0.6 mmol) and intermediate 49-7 of example 49 (0.3 mmol) were placed in a round bottom flask, dichloromethane (5 mL) was added and cooled to 0 ℃ after which sodium acetate (0.3 mmol) and sodium triacetoxyborohydride (0.6 mmol) were added, the reaction was stirred at room temperature for 24h, TLC check for completion of the reaction, the reaction was extracted with dichloromethane and water, the organic phase was concentrated under reduced pressure, and the product compound 54 (0.102g, 0.166mmol) was obtained in 55% yield by stirring with silica gel and column chromatography.
1 H NMR(400MHz,Chloroform-d)δ8.00(d,J=5.4Hz,1H),7.82–7.69 (m,1H),7.26(d,J=4.9Hz,2H),4.86(s,2H),4.35(s br,4H),3.93(s br,4H), 3.87(s,4H),3.83(s,3H),3.46–3.24(m,6H),2.77(q,J=5.4,2H),2.45–2.35 (m,1H),1.88–1.49(m,8H),1.44(t,J=7.5,3H).
13 C NMR(101MHz,CDCl 3 )δ167.55,156.92,153.66,153.47,150.74, 146.29,142.45,134.54,122.87,122.70,119.04,117.27,113.24,108.17,67.00, 64.37,64.29,60.51,54.12,46.95,45.91,41.99,33.18,29.28,25.36,23.76, 12.38.
ESI-MS[M+H] + (m/z):616.3.
Example 55
Example 55 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (2, 2-trifluoroethyl) -piperazin-2-one (compound 55) and a method for its preparation, compound 55 having the following structural formula:
Figure BDA0002520468220000921
the intermediate 49-7 (0.2 mmol) obtained in example 49 was placed in a three-necked flask, potassium carbonate (0.5 mmol), acetonitrile (5 mL) and trifluoroethyl trifluoromethanesulfonate (0.4 mmol) were added thereto, the mixture was slowly heated to a reflux temperature (85 ℃ C.) and stirred for 2 hours, after completion of the TLC detection reaction, the solvent was distilled off under reduced pressure, water and ethyl acetate were added to the residue to extract a water layer free from the product, the organic phase was washed with a saturated saline solution, dried over anhydrous magnesium sulfate, concentrated, sample-stirred with silica gel and subjected to column chromatography to obtain the objective compound 55 (0.053 g,0.095 mmol) in a yield of 47%.
1 H NMR(400MHz,Chloroform-d)δ8.07–7.95(m,1H),7.82–7.70(m, 1H),7.33–7.21(m,2H),4.87(s,2H),4.35(s br,4H),3.86(t,J=7.2Hz,4H), 3.84(s,3H),3.50(t,J=5.8Hz,2H),3.48(s,2H),3.35(q,J=7.5Hz,2H),3.08 (q,J=9.2Hz,2H),2.97(t,J=5.4Hz,2H),1.44(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ166.02,156.91,153.68,150.83,145.96, 134.54,122.89,122.72,119.08,117.27,113.23,66.99,57.18,56.96,50.03, 46.48,45.93,42.03,29.25,23.78,12.37.
19 F NMR(376MHz,CDCl 3 )δ-69.17.
ESI-MS[M+H] + (m/z):558.2.
Example 56
Example 56 of the present application provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (4-oxocyclohexyl) -piperazin-2-one (compound 56) and a process for its preparation, compound 56 having the following structural formula:
Figure BDA0002520468220000931
compound 54 (0.080g, 0.13mmol) from example 54 was placed in a round-bottom flask, dichloromethane (5 mL) was added and cooled to 0 ℃, after which THF (10 mL) and concentrated hydrochloric acid (2 mL) were added thereto, stirred at reflux temperature for 4h, the reaction was checked by tlc for completion, concentrated under reduced pressure, and product 56 (0.065g, 0.114mmol) was obtained in 88% yield by stirring over silica gel and column chromatography.
1 H NMR(400MHz,Chloroform-d)δ8.07–7.95(m,1H),7.82–7.69(m, 1H),7.27(q,J=2.2Hz,2H),4.88(s,2H),4.35(s br,4H),3.94–3.78(m,7H), 3.47(t,J=6.3Hz,2H),3.43–3.27(m,4H),2.82(t,J=6.3Hz,2H),2.77– 2.67(m,1H),2.55–2.45(m,2H),2.38–2.27(m,2H),2.14–1.98(m,2H), 1.94–1.82(m,2H),1.44(t,J=7.5Hz,3H).
13 C NMR(101MHz,Chloroform-d)δ209.89,166.97,156.91,153.68, 153.48,150.84,146.10,142.52,134.55,122.87,122.72,119.11,117.27,113.23, 67.00,59.03,54.37,46.76,46.20,45.80,41.95,38.43,29.29,27.74,23.80, 12.38.
ESI-MS[M+H] + (m/z):572.3.
Example 57
Example 57 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (4-hydroxycyclohexyl) -piperazin-2-one (compound 57) and a method for its preparation, compound 57 having the following structural formula:
Figure BDA0002520468220000941
compound 56 (0.058g, 0.1mmol) from example 56 was placed in a round-bottom flask, and after adding methanol (3 mL) and cooling to 0 ℃, sodium borohydride (0.2 mmol) was added thereto, and after slowly warming to room temperature and stirring for 2 hours, the reaction was complete by tlc, and after concentration under reduced pressure, the product compound 57 (0.041g, 0.072mmol) was obtained in a yield of 72% by stirring with silica gel and column chromatography.
1 H NMR(400MHz,Chloroform-d)δ8.09–7.94(m,1H),7.84–7.69(m, 1H),7.41–7.18(m,2H),4.87(d,J=3.3Hz,2H),4.35(s br,4H),3.92–3.73 (m,7H),3.66–3.52(m,1H),3.52–3.23(m,6H),2.77(t,J=5.5Hz,2H),2.42 –2.26(m,1H),2.16–1.96(m,2H),1.96–1.50(m,4H),1.44(t,J=7.4Hz, 3H),1.37–1.21(m,2H).
13 C NMR(101MHz,CDCl 3 )δ167.42,156.92,153.66,153.48,150.79, 146.25,142.52,134.56,122.86,122.70,119.08,117.26,113.22,70.09,67.01, 61.35,53.96,46.87,46.25,41.97,34.08,31.50,29.29,26.25,23.78,12.38.
ESI-MS[M+H] + (m/z):574.3.
Example 58
Example 58 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (4, 4-difluorocyclohexyl) -piperazin-2-one (compound 58) and a method for its preparation, compound 58 having the following structural formula:
Figure BDA0002520468220000951
4, 4-Difluorocyclohexanone (0.4 mmol) and intermediate 49-7 (0.2 mmol) of example 49 were placed in a round bottom flask, and dichloromethane (5 mL) was added thereto and cooled to 0 ℃ after which sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, and the completion of the reaction was checked by TLC, and the reaction solution was extracted with dichloromethane and water, concentrated under reduced pressure by the action of organic phase, and then stirred with silica gel and column chromatography to give product compound 58 (0.058g, 0.098mmol) in 49% yield.
1 H NMR(400MHz,Chloroform-d)δ8.09–7.96(m,1H),7.82–7.70(m, 1H),7.33–7.20(m,2H),4.87(s,2H),4.35(s br,4H),3.99–3.78(m,7H),3.43 (dd,J=6.3,4.4Hz,2H),3.40–3.27(m,4H),2.77(t,J=5.3Hz,2H),2.42(d,J =9.7Hz,1H),2.25–2.04(m,2H),1.91–1.58(m,6H),1.44(t,J=7.4Hz, 3H).
13 C NMR(101MHz,CDCl 3 )δ167.12,156.92,153.67,153.45,150.79, 146.16,142.48,134.54,122.87,122.70,119.05,117.26,113.24,66.99,59.42, 54.17,46.83,45.96,41.97,31.86,29.27,24.43,24.35,23.78,12.38.
ESI-MS[M+H] + (m/z):594.3.
Example 59
Example 59 provides 4- (1, 1-tetrahydro-2H-thiopyran-4-yl) -1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (compound 59) and a method for its preparation, compound 59 having the following structural formula:
Figure BDA0002520468220000961
tetrahydro-4H-thiopyran-4-one-1, 1-dioxide (0.3 mmol) and intermediate 49-7 of example 49 (0.2 mmol) were placed in a round-bottomed flask, methylene chloride (5 mL) was added thereto and cooled to 0 ℃ and then sodium acetate (0.2 mmol) and sodium triacetoxyborohydride (0.5 mmol) were added thereto, the reaction was stirred at room temperature for 24h, TLC showed completion of the reaction, the reaction solution was extracted with methylene chloride and water, the organic phase was concentrated under reduced pressure, and then, stirring and column chromatography were performed with silica gel to obtain 59 (0.032g, 0.053 mmol) as a product in a yield of 26%.
1 H NMR(400MHz,Chloroform-d)δ8.06–7.96(m,1H),7.81–7.70(m, 1H),7.33–7.20(m,2H),4.87(s,2H),4.35(s br,4H),3.99–3.78(m,7H),3.48 (dd,J=6.3,4.4Hz,2H),3.41–3.29(m,4H),3.27–3.12(m,2H),2.99–2.86 (m,2H),2.78(t,J=6.3Hz,2H),2.58(d,J=3.2Hz,1H),2.33–2.12(m,4H), 1.44(t,J=7.4Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ166.30,156.90,153.70,153.44,150.87, 145.89,142.50,134.54,122.89,122.73,119.10,117.30,113.25,66.99,57.10, 54.27,48.64,46.63,45.75,41.92,29.30,25.80,23.81,12.38.
ESI-MS[M+H] + (m/z):608.3.
Example 60
Example 60 provides 1-cyclopentyl-4- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (compound 60) and a method for its preparation, compound 60 having the following structural formula:
Figure BDA0002520468220000971
(1) Preparation of (2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methanol (60-1)
Figure BDA0002520468220000972
The compound 49-3 (1.415g, 5mmol), 2-ethylbenzimidazole (6 mmol), pd 2 (dba) 3 (0.5mmol),Xphos(1.0mmol),CsCO 3 (10 mmol) is put into a three-necked flask, dioxane (60 mL) is added, nitrogen is pumped in for 3 times, the temperature is raised to 110 ℃ for reaction for 4-5h, then the hot reaction product is filtered, a filter cake is washed by DCM, the filtrate is concentrated and concentrated, silica gel is used for sample mixing, and column chromatography is carried out to obtain the target product 60-1 (1.317g, 3.351mmol), wherein the yield is 67%.
ESI-MS[M+H] + (m/z):394.2.
(2) Preparation of 4- (8- (chloromethyl) -2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-9H-purin-6-yl) morpholine (60-2)
Figure BDA0002520468220000973
Placing 60-1 (0.786 g, 2mmol) of the compound in a three-necked bottle at room temperature, adding 30mL of dichloromethane, cooling to 0 ℃, slowly adding thionyl chloride (2 mL) while stirring, heating to room temperature to react for 2h (TLC detection reaction is finished, n-hexane: EA = 2).
ESI-MS[M+H] + (m/z):412.2.
(3) Preparation of 1-cyclopentyl-4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (Compound 60)
Figure BDA0002520468220000981
Placing 4-cyclopentylpiperazine-2-one (0.3 mmol) and intermediate 60-2 (0.2 mmol) in a round-bottomed flask, adding isopropanol and diisopropylethylamine (1 mmol), heating to 85 ℃, reacting and stirring for 24h, detecting complete reaction (EA) by TLC, concentrating the reaction solution under reduced pressure, stirring with silica gel, and performing column chromatography to obtain compound 60 (0.074g, 0.136mmol) with yield of 68%.
1 H NMR(400MHz,CDCl 3 )δ8.05–7.94(m,1H),7.75(dd,J=5.7,3.3 Hz,1H),7.33–7.19(m,2H),5.03–4.83(m,2H),3.89–3.70(m,9H),3.65– 3.56(m,1H),3.50(d,J=5.4Hz,1H),3.42–3.12(m,7H),2.86–2.71(m,2H), 1.92–1.76(m,4H),1.74–1.57(m,4H),1.45(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ166.14,160.30,156.94,153.70,150.73, 146.61,142.60,134.62,122.73,122.65,119.10,116.99,113.23,67.02,57.60, 54.22,53.85,49.66,45.89,40.92,29.20,27.87,24.07,23.77,12.38.
ESI-MS[M+H] + (m/z):544.3.
Example 61
Example 61 provides 1-cyclopropylmethyl-4- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (compound 61) and a process for its preparation, compound 61 having the formula:
(1) Preparation of 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (61-1)
Figure BDA0002520468220000991
Piperazinone (1.2 mmol) and intermediate 60-2 (1.0 mmol) from example 60 were placed in a round bottom flask, isopropanol (5 mL) and diisopropylethylamine (5 mmol) were added, the temperature was raised to 85 ℃ to react and stir 24h, completion of reaction (EA) was detected by tlc, the reaction solution was concentrated under reduced pressure, and after sample mixing with silica gel, column chromatography was performed to give compound 61-1 (0.312g, 0.657mmol) with a yield of 66%.
ESI-MS[M+H] + (m/z):476.2.
(2) 1-Cyclopropylmethyl-4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (Compound 61)
Figure BDA0002520468220000992
Placing a compound 61-1 (0.095g, 0.2mmol) in a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding an intermediate bromomethylcyclopropane (0.6 mmol) in portions, reacting for 4-5h at 30-32 ℃, keeping the reaction for 4-5h, detecting the basic reaction (EA) by TLC, cooling the reaction solution, adding water and ethyl acetate, extracting until no product exists in an aqueous layer, washing an organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and performing column chromatography to obtain a target product compound 61 (0.054g, 0.102mmol), wherein the yield is 51%.
1 H NMR(400MHz,Chloroform-d)δ8.11–7.97(m,1H),7.87–7.72(m, 1H),7.41–7.22(m,2H),4.36(s br,4H),3.89(d,J=6.8Hz,6H),3.84(s,3H), 3.45(t,J=5.6Hz,2H),3.37(q,J=7.5Hz,2H),3.31(d,J=8.9Hz,4H),2.84 (t,J=5.6Hz,2H),1.46(t,J=7.5Hz,3H),0.98(d,J=6.9Hz,1H),0.54(d,J= 7.6Hz,2H),0.26(d,J=5.1Hz,2H).
13 C NMR(101MHz,CDCl 3 )δ166.02,156.93,153.69,150.69,146.62, 142.49,134.58,122.84,122.67,119.04,117.00,113.22,67.01,57.35,54.23, 50.50,49.49,46.40,45.60,29.21,23.74,12.39,8.96,3.55.
ESI-MS[M+H] + (m/z):530.3.
Example 62
Example 62 provides 4- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one (compound 62) and a method for its preparation, compound 62 having the following structural formula:
Figure BDA0002520468220001001
1- (1-methyl-1H-pyrazol-4-yl) piperazin-2-one hydrochloride (10-1) (0.3 mmol) and intermediate 60-2 (0.2 mmol) of example 60 were placed in a round-bottomed flask, isopropanol (5 mL) and diisopropylethylamine (1 mmol) were added, the temperature was raised to 85 ℃ and the reaction was stirred for 12h, completion of reaction (EA) was detected by TLC, the reaction solution was concentrated under reduced pressure, and then subjected to silica gel sample-mixing and column chromatography to give the objective product 62 (0.089 g, 0.160 mmol) in 80% yield.
1 H NMR(400MHz,Chloroform-d)δ8.03(s,1H),8.02–7.97(m,1H), 7.80–7.71(m,1H),7.46(s,1H),7.34–7.22(m,2H),4.36(s,4H),4.01–3.77 (m,12H),3.71(t,J=5.5Hz,2H),3.43(s,2H),3.35(q,J=7.5Hz,2H),2.96(t, J=5.5Hz,2H),1.45(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ164.10,156.93,153.72,150.78,146.31, 142.56,134.59,129.45,124.84,123.08,122.84,122.68,119.08,117.01,113.22, 67.01,57.76,54.11,49.10,47.44,45.89,39.34,29.22,23.78,12.38.
ESI-MS[M+H] + (m/z):556.3.
Example 63
Example 63 provides 2- (4- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -2-oxopiperazin-1-yl) acetamide (compound 63) and a process for its preparation, compound 63 having the following structural formula:
Figure BDA0002520468220001011
placing a compound 61-1 (0.095g, 0.2mmol) in a round-bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, then cooling to 0 ℃, slowly adding an intermediate iodoacetamide (0.3 mmol) in a fractional manner, slowly heating to room temperature, reacting for 4-5h, detecting by TLC (TLC) that the reaction is basically complete (EA), concentrating the reaction solution under reduced pressure, extracting the residue with water and dichloromethane until no product exists in a water layer, washing an organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, mixing the organic phase with silica gel, and carrying out column chromatography to obtain a target product compound 63 (0.048g, 0.090mmol), wherein the yield is 45%.
1 H NMR(400MHz,Chloroform-d)δ8.08–7.96(m,1H),7.82–7.69(m, 1H),7.27(q,J=3.7Hz,2H),6.27(s,1H),5.56(s,1H),4.35(s br,4H),4.03(s, 2H),3.99–3.80(m,9H),3.50(t,J=5.5Hz,2H),3.35(q,J=7.7Hz,2H),3.34 (s,2H),2.88(t,J=5.5Hz,2H),1.45(t,J=7.5,3H).
13 C NMR(101MHz,CDCl 3 )δ170.22,167.32,156.93,153.72,153.67, 150.76,146.28,142.50,134.57,122.86,122.70,119.06,117.00,113.22,67.00, 57.03,54.01,50.39,49.33,48.05,45.89,29.21,23.75,12.38.
ESI-MS[M+H] + (m/z):533.3.
Example 64
Example 64 of the present application provides 4-acetyl-1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) piperazin-2-one (compound 64) and a method for its preparation, compound 64 having the following structural formula:
Figure BDA0002520468220001021
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round bottom flask and dichloromethane (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ acetic anhydride (0.3 mmol) was added dropwise slowly, after warming to room temperature and stirring for 2h, TLC detection completed the reaction was followed by distilling off the solvent under reduced pressure, water and dichloromethane were added to the residue and extracted to an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and chromatographed over silica gel to give the desired product compound 64 (0.082g, 0.158mmol) in 79% yield.
1 H NMR(400MHz,Chloroform-d)δ8.06–7.95(m,1H),7.81–7.70(m, 1H),7.33–7.22(m,2H),4.88(d,J=4.6Hz,2H),4.33(s br,6H),3.92–3.72 (m,6H),3.85(s,3H),3.63–3.53(m,2H),3.34(q,J=7.5Hz,2H),2.13(d,J= 2.5Hz,3H),1.44(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ168.91,165.64,164.68,156.91,153.70, 150.91,145.53,142.52,134.53,122.90,122.74,119.10,117.29,113.22,66.97, 49.68,46.05,43.28,42.46,38.54,29.23,23.80,21.34,12.37.
ESI-MS[M+H] + (m/z):518.3.
Example 65
Example 65 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-isobutyrylpiperazin-2-one (compound 65) and a method for its preparation, compound 65 having the following structural formula:
Figure BDA0002520468220001031
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and isobutyryl chloride (0.3 mmol) was added dropwise slowly, then warmed to room temperature and stirred for 2h, after completion of TLC detection reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give target product 65 (0.066 g, 0.121mmol) in 60% yield.
1 H NMR(400MHz,Chloroform-d)δ8.05–7.95(m,1H),7.82–7.73(m, 1H),7.27(q,J=4.7Hz,2H),4.88(s,2H),4.30(s br,6H),3.92–3.72(m,6H), 3.85(s,3H),3.56(s,2H),3.35(q,J=7.5Hz,2H),2.76(hept,J=6.6Hz,1H), 1.44(t,J=7.5Hz,3H),1.15(d,J=6.8Hz,6H).
13 C NMR(101MHz,CDCl 3 )δ170.08,165.96,156.89,153.68,153.40, 150.82,142.20,134.44,122.98,122.83,119.01,117.31,113.23,66.97,49.11, 46.13,42.39,38.88,33.68,30.55,29.24,23.70,19.17,19.01,12.38.
ESI-MS[M+H] + (m/z):546.3.
Example 66
Example 66 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-cyclopropylformylpiperazin-2-one (compound 66) and its preparation, compound 66 having the following structural formula:
Figure BDA0002520468220001041
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and then cyclopropylcarbonyl chloride (0.3 mmol) was added dropwise slowly, followed by warming to room temperature and stirring for 2h, after completion of the TLC detection reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give the desired product compound 66 (0.075g, 0.138mmol) in 69% yield.
1 H NMR(400MHz,CDCl 3 )δ8.05–7.95(m,1H),7.81–7.69(m,1H), 7.31–7.21(m,2H),4.89(s,2H),4.35(m,4H),3.95–3.82(m,9H),3.57(s, 2H),3.35(q,J=7.5Hz,2H),1.74–1.58(m,1H),1.44(t,J=7.5Hz,3H),1.25 (s,2H),1.04–1.00(m,2H),0.87(dd,J=13.3,6.1Hz,2H).
13 C NMR(101MHz,CDCl 3 )δ172.0,165.1,156.93,153.68,153.41, 150.84,145.15,142.38,134.49,122.94,122.77,119.02,117.30,113.24,66.98, 48.97,46.63,46.02,42.48,39.04,29.27,23.75,12.38,11.12,8.23.
ESI-MS[M+H] + (m/z):544.3.
Example 67
Example 67 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-cyclobutylformylpiperazin-2-one (compound 67) and a method for its preparation, compound 67 having the following structural formula:
Figure BDA0002520468220001051
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and cyclobutylcarbonyl chloride (0.3 mmol) was added dropwise slowly, then warmed to room temperature and stirred for 2h, after completion of the TLC detection reaction the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue and extracted to an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to silica gel column chromatography to give the desired product compound 67 (0.061g, 0.109mmol) in 55% yield.
1 H NMR(400MHz,CDCl 3 )δ8.04–7.96(m,1H),7.80–7.72(m,1H), 7.32–7.22(m,2H),4.87(s,2H),4.35(s,2H),3.95–3.78(m,7H),3.47–3.40 (m,2H),3.39–3.30(m,4H),2.84–2.75(m,2H),2.32(dd,J=13.5,6.1Hz, 1H),2.07(s,1H),1.93–1.71(m,6H),1.64(d,J=12.3Hz,1H),1.44(t,J=7.5 Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ173.26,164.92,156.91,153.68,153.40, 150.83,145.61,142.32,134.47,122.96,122.80,119.02,117.30,113.24,66.97, 48.56,46.28,46.07,42.48,38.80,37.11,29.21,24.89,23.73,17.91,12.38.
ESI-MS[M+H] + (m/z):558.3.
Example 68
Example 68 provides 1- ((2- (2-ethyl-1H-benzimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-cyclopentacarbyl piperazin-2-one (compound 68) and a method for its preparation, compound 68 having the following structural formula:
Figure BDA0002520468220001061
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, cooled to 0 ℃ and cyclopentyl carbonyl chloride (0.3 mmol) was added dropwise slowly, then warmed to room temperature and stirred for 2h, after completion of the TLC detection reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue and extracted to an aqueous layer without the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give the desired product compound 68 (0.067 g, 0.117mmol) in 59% yield.
1 H NMR(400MHz,CDCl 3 )δ7.99(dd,J=5.7,3.2Hz,1H),7.81–7.71 (m,1H),7.35–7.20(m,2H),4.88(s,2H),4.47–4.16(m,6H),3.90–3.83(m, 7H),3.63–3.49(m,2H),3.35(q,J=7.5Hz,2H),2.96–2.70(m,2H),1.89– 1.69(m,7H),1.59(dd,J=6.6,4.2Hz,2H),1.43(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ174.70,165.0,156.96,153.69,153.41, 150.83,145.60,142.29,134.46,122.96,122.80,119.03,117.30,113.22,66.98, 49.13,46.52,46.10,42.48,41.28,38.94,30.00,29.23,26.04,23.69,12.40
ESI-MS[M+H] + (m/z):572.3.
Example 69
Example 69 of the present application provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4- (2-hydroxypropionyl) piperazin-2-one (compound 69), and a method for its preparation, compound 69 having the following structural formula:
Figure BDA0002520468220001071
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, L-lactic acid (0.3 mmol) was slowly added dropwise thereto at room temperature, HOBT (0.3 mmol) and EDCI (0.4 mmol) were then added thereto, stirring was carried out at room temperature for 12h, the solvent was distilled off under reduced pressure after completion of the TLC detection reaction, water and methylene chloride were added to the residue to extract a water layer free from the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give the objective product compound 69 (0.059 g,0.108 mmol) in 54% yield.
1 H NMR(400MHz,Chloroform-d)δ8.06–7.95(m,1H),7.80–7.71(m, 1H),7.27(q,J=3.9Hz,2H),4.87(s,2H),4.51–4.09(m,8H),3.92–3.72(m, 7H),3.79–3.55(m,4H),3.34(q,J=7.5Hz,2H),1.82(s,4H),1.44(t,J=7.4 Hz,3H),1.36(d,J=6.7Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ169.49,165.01,156.91,153.70,153.40, 150.93,145.41,142.47,134.50,122.92,122.77,119.08,117.28,113.22,66.96, 64.51,48.39,46.03,45.84,42.49,39.72,29.26,23.79,21.06,12.37.
ESI-MS[M+H] + (m/z):548.3.
Example 70
Example 70 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-methanesulfonylpiperazin-2-one (compound 70) and a method for its preparation, compound 70 having the following structural formula:
Figure BDA0002520468220001081
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ methanesulfonyl chloride (0.3 mmol) was added dropwise slowly thereto, followed by warming to room temperature and stirring for 2h, detection by TLC after completion of the reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free of the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give the desired product compound 70 (0.080g, 0.144mmol) in a yield of 72%.
1 H NMR(400MHz,Chloroform-d)δ8.08–7.95(m,1H),7.84–7.69(m, 1H),7.28(q,J=5.8Hz,2H),4.88(s,2H),4.35(s br,4H),4.00(s,2H),3.92– 3.72(m,4H),3.85(s,3H),3.66(d,J=5.4Hz,2H),3.55(t,J=5.4Hz,2H), 3.35(q,J=7.5Hz,2H),2.89(s,3H),1.44(t,J=7.5Hz,3H).
13 C NMR(101MHz,CDCl 3 )δ163.99,156.92,153.71,153.41,150.93, 145.45,142.54,134.55,122.90,122.73,119.10,117.27,113.23,66.98,48.63, 46.78,45.83,42.66,42.40,36.41,29.25,23.81,12.37.
ESI-MS[M+H] + (m/z):554.2.
Example 71
Example 71 provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-cyclopropylsulfonylpiperazin-2-one (compound 71) and its preparation, compound 71 having the following structural formula:
Figure BDA0002520468220001091
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottomed flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, after cooling to 0 ℃ and dropwise addition of cyclopropylsulfonyl chloride (0.3 mmol) thereto, followed by warming to room temperature and stirring for 2h, detection by TLC after completion of the reaction, removal of the solvent under reduced pressure, addition of water and methylene chloride to the residue and extraction to an aqueous layer free of product, washing of the organic phase with saturated brine, drying of the organic phase over anhydrous magnesium sulfate, concentration and column chromatography over silica gel to give the desired product 71 (0.069g, 0.119mmol) in 59% yield.
1 H NMR(400MHz,Chloroform-d)δ8.08–7.96(m,1H),7.81–7.71(m, 1H),7.35–7.22(m,2H),4.88(s,2H),4.35(s,4H),4.06(s,2H),3.94–3.78(m, 7H),3.72–3.55(m,4H),3.35(q,J=7.5Hz,2H),2.37–2.25(m,1H),1.44(t, J=7.5Hz,3H),1.25–1.19(m,2H),1.10–0.99(m,2H).
13 C NMR(101MHz,CDCl 3 )δ164.25,156.91,153.71,153.42,150.91, 145.52,142.46,134.52,122.91,122.75,119.09,117.27,113.24,66.98,49.08, 46.83,45.88,43.02,42.36,29.27,26.58,23.79,12.38,4.79.
ESI-MS[M+H] + (m/z):580.3.
Example 72
Example 72 of the present application provides 1- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -9-methyl-6-morpholinyl-9H-purin-8-yl) methyl) -4-methanesulfonylpiperazin-2-one (compound 72) and its preparation, compound 72 having the following structural formula:
Figure BDA0002520468220001101
intermediate 49-7 (0.2 mmol) from example 49 was placed in a round-bottom flask, methylene chloride (5 mL) and triethylamine (0.5 mmol) were added thereto, and after cooling to 0 ℃ propylsulfonyl chloride (0.3 mmol) was added dropwise slowly, followed by warming to room temperature and stirring for 2h, detection by TLC after completion of the reaction, the solvent was distilled off under reduced pressure, water and methylene chloride were added to the residue to extract an aqueous layer free from the product, the organic phase was washed with saturated brine, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography over silica gel to give compound 72 (0.051g, 0.088mmol), a target product, in 44% yield.
1 H NMR(400MHz,Chloroform-d)δ8.00(dd,J=7.5,2.1Hz,1H),7.82– 7.70(m,1H),7.32–7.21(m,2H),4.88(s,2H),4.34(s br,4H),4.04(s,2H), 3.86(d,J=8.1Hz,7H),3.70–3.52(m,4H),3.35(q,J=7.4Hz,2H),3.04– 2.91(m,2H),1.92–1.76(m,2H),1.44(t,J=7.4Hz,3H),1.07(t,J=7.4Hz, 3H).
13 C NMR(101MHz,CDCl 3 )δ164.26,156.91,153.70,153.43,150.89, 145.50,142.46,134.52,122.91,122.75,119.08,117.28,113.23,66.99,52.68, 48.59,47.09,45.89,42.73,42.47,29.24,23.78,16.98,13.00,12.37.
ESI-MS[M+H] + (m/z):582.3.
Example 73
Example 73 provides 1- (cyclopropylmethyl) -4- ((2- (2-ethyl-1H-benzoimidazol-1-yl) -7-methyl-4-morpholinothieno [3,2-d ] pyrimidin-6-yl) methyl) piperazin-2-one (compound 73) and a method for its preparation, compound 73 having the following structural formula:
Figure BDA0002520468220001111
putting 4- ((2- (2-ethyl-1H-benzimidazole-1-yl) -7-methyl-4-morpholinyl thiophene [3,2-d ] pyrimidine-6-yl) methyl) piperazine-2-ketone (0.2 mmol) into a round bottom flask, adding THF (5 mL) and t-BuOK (0.3 mmol), heating to 30-32 ℃, reacting and stirring for 20min, adding intermediate bromomethyl cyclopropane (0.6 mmol) in portions, reacting for 4-5H at 30-32 ℃, detecting by TLC to detect that the reaction is basically complete (EA), cooling the reaction liquid, adding water and ethyl acetate, extracting until no product exists in a water layer, washing an organic phase with saturated saline, drying the organic phase with anhydrous magnesium sulfate, and purifying by column chromatography to obtain a target product 72 (0.031 g,0.057 mmol) with the yield of 28%.
ESI-MS[M+H] + (m/z):546.3.
Example 74
Example 74 of the present application provides 4- ((2- (1H-indol-4-yl) -4-morpholinothieno [3,2-d ] pyrimidin-6-yl) methyl) -1- (cyclopropylmethyl) piperazin-2-one (compound 74) and a method for its preparation, compound 74 having the following structural formula:
Figure BDA0002520468220001112
(1) Preparation of 2-chloro-6-chloromethyl-4-morpholinylthieno [3,2-d ] pyrimidine (74-1)
Figure BDA0002520468220001121
The intermediate 1-3 (4.275g, 15mmol) of example 1 was placed in a three-necked bottle, 50mL of dichloromethane was added, then the mixture was cooled to 0 ℃, thionyl chloride (5 mL) was slowly added thereto while stirring, after the addition, the temperature was raised to room temperature to react for 2h (TLC detection reaction was completed, n-hexane: EA = 2).
ESI-MS[M+H] + (m/z):304.1.
(2) Preparation of 4- ((2-chloro-4-morpholinothieno [3,2-d ] pyrimidin-6-yl) methyl) -1- (cyclopropylmethyl) piperazin-2-one (74-2)
Figure BDA0002520468220001122
Putting 1- (cyclopropylmethyl) piperazine-2-one (5 mmol) and an intermediate 74-1 (5 mmol) in a round-bottom flask, adding isopropanol (5 mL) and diisopropylethylamine (5 mmol), heating to 85 ℃, reacting and stirring for 24h, detecting complete reaction (EA) by TLC, concentrating the reaction solution under reduced pressure, stirring with silica gel, and performing column chromatography to obtain a compound 74-2 (1.242g, 2.943 mmol) with the yield of 59%.
ESI-MS[M+H] + (m/z):422.1.
(3) Preparation of 4- ((2- (1H-indol-4-yl) -4-morpholinylthieno [3,2-d ] pyrimidin-6-yl) methyl) -1- (cyclopropylmethyl) piperazin-2-one (compound 74)
Figure BDA0002520468220001131
The compound 74-2 (0.5 mmol), 1H-indole-4-boronic acid (1 mmol), pdCl 2 (dppf)DCM (0.03mmol),K 3 PO 4 (1.5 mmol) is placed in a round-bottom flask, n-propanol (10 mL) is added, nitrogen is introduced for 3 times by vacuumizing, the temperature is increased to 85 ℃ for reaction for 3-6h, TLC detection is carried out until the reaction is completed, then filtration is carried out while the solution is hot, and a filter cake is washed by DCMThe filtrate was concentrated and stirred with silica gel, and column chromatography was carried out to give the objective compound 74 (0.087g, 0.173mmol) in 35% yield.
ESI-MS[M+H] + (m/z):503.2.
Example 75
Example 75 of the present application provides 4- ((2- (5-fluoro-1H-indol-4-yl) -4-morpholinothieno [3,2-d ] pyrimidin-6-yl) methyl) -1- (cyclopropylmethyl) piperazin-2-one (compound 75) and a method for its preparation, compound 75 having the following structural formula:
Figure BDA0002520468220001132
compound 74-2 (0.5 mmol), 5-fluoro-1H-indole-4-boronic acid (1 mmol), pdCl 2 (dppf)DCM(0.03mmol),K 3 PO 4 (1.5 mmol) is placed in a round-bottom flask, n-propanol (10 mL) is added, nitrogen is introduced for 3 times by vacuumizing, the temperature is raised to 85 ℃ for reaction for 3-6h, TLC is used for detecting that the reaction is completed, then the reaction is filtered while the solution is hot, a filter cake is washed by DCM, a filtrate is concentrated and mixed with silica gel, and column chromatography is carried out to obtain the target product compound 75 (0.062g, 0.119mmol) with the yield of 24%.
ESI-MS[M+H] + (m/z):521.2.
Example 76
Example 76 provides 4- ((2- (1H-indazol-4-yl) -4-morpholinyl thieno [3,2-d ] pyrimidin-6-yl) methyl) -1- (cyclopropylmethyl) piperazin-2-one (compound 76) and a method for its preparation, compound 76 having the following structural formula:
Figure BDA0002520468220001141
compound 74-2 (0.5 mmol), 1H-indazole-4-boronic acid (1 mmol), pdCl 2 (dppf)DCM (0.03mmol),K 3 PO 4 (1.5 mmol) is placed in a round-bottom flask, n-propanol (10 mL) is added, nitrogen is introduced for 3 times by vacuumizing, the temperature is increased to 85 ℃ for reaction for 3-6h, TLC detection is carried out until the reaction is completed, then the hot reaction is carried out, the filter cake is filtered, DCM is used for washing the filter cake, the filtrate is concentrated and mixed with silica gel, and column chromatography is carried out to obtain the targetProduct compound 76 (0.053g, 0.105mmol), 21% yield.
ESI-MS[M+H] + (m/z):504.2.
Test example 1 kinase inhibitory Activity test
Test compounds (0.001-10. Mu.M) or blank solvents were incubated with different protein kinases and the corresponding polypeptide substrates in reaction buffer (8 mM propanesulfonate (MOPS, pH 7.0), 10mM magnesium acetate, 0.2mM ethylenediaminetetraacetic acid (EDTA) and Km concentration of. Gamma.33P-ATP solution). After incubation at room temperature for 40min, the reaction was stopped by adding 3% phosphate solution to the reaction buffer. Then, 10. Mu.L of the reaction solution was dropped onto a P30 filter paper and washed 3 times with 75mM phosphate solution, followed by washing once with methanol, air-drying the P30 filter paper and counting with scintillation liquid. Half inhibitory concentration IC for inhibitory Activity of Compounds 50 And (4) showing.
TABLE 2 examples protein kinase inhibitory Activity (where "D" denotes IC) 50 >1 mu M; "C" means 1. Mu.M>IC 50 >100nM; "B" means 100nM>IC 50 >10nM; "A" represents IC 50 <10nM)
Figure BDA0002520468220001151
Figure BDA0002520468220001161
Figure BDA0002520468220001171
As can be seen from table 2, the piperazinone substitutes prepared in examples 1 to 76 of the present application have a good inhibitory effect on PI3K δ, and also have a partial inhibitory effect on PI3K α, PI3K β, and PI3K γ.
Test example 2 cell growth inhibition test
Collecting cells in logarithmic phase in a centrifuge tube, centrifuging the centrifuge tube in a centrifuge at 1200r/min3min. Resuspend the cells in fresh medium and count the resuspended cells, dilute to a cell concentration of 8X 10 4 ~1×10 5 The cells are inoculated into a 96-well plate according to 100 mu L of cell suspension per well (namely 8000-10000 cells per well), and the inoculated 96-well plate is put into an incubator for incubation. The drug gradient was diluted to an appropriate concentration (generally three-fold dilution), the inoculated cells were treated at 100. Mu.L per well, 3 duplicate wells per drug concentration were set, and the cells were treated with fresh medium containing 0.1% DMSO as a negative control group. Putting the 96-well plate in an incubator for further culture for required time (24h, 48h,72h,96h and 120h), taking out, adding 20 mu L of MTS solution into each well, incubating for 2-4 hours (most suitable for 2 h) at 37 ℃, detecting the absorbance value at 492nm by using an enzyme-linked immunosorbent assay (Spectra MAX M5) after the solution to be detected and the color developing agent generate a colored solution, wherein the absorbance value (A) is in direct proportion to the number of living cells. The inhibition rate of the drug on the tumor cells is calculated according to the following formula: cell growth inhibition rate = (control group A) 495nm Experimental group A 495nm ) Control group A 495nm X100%. Calculating the relationship between the cell growth inhibition rate and the drug concentration by using Graphpad Prism software, fitting a drug growth inhibition curve and calculating the median Inhibitory Concentration (IC) 50 ) The value is obtained.
TABLE 2 example 67 inhibitory Activity on tumor cell lines (IC) on 5 days 50 :nM)
Figure BDA0002520468220001181
The growth inhibition curves of example 67 at 24h, 72h and 120h are shown in FIGS. 1 to 4.
As can be seen from Table 2 and FIGS. 1-4, the piperazinone substituents of example 67 of this application have a superior inhibitory effect on the growth of a wide variety of cells.
Test example 3 in vivo antitumor Activity test
Collecting tumor cells SU-DHL-6 and JEKO-1 in logarithmic growth phase, washing cells with RPMI-1640 blank medium without serum and antibiotics for 3 times, resuspending cells, counting, and suspending cellsDiluting to 1 × 10 7 Individual cells/mL. 100 mu L of cell suspension is inoculated under the right flank of a female NOD-SCID (6-9 weeks old) mouse to establish a mouse subcutaneous tumor model, and the NOD-SCID (6-9 weeks old) mouse used in the research is purchased from Beijing Huafukang bioscience, inc. (Beijing, china). The vaccinated mice were raised under Specific Pathogen Free (SPF) conditions and all animal experiments were approved by the institutional animal care and treatment committee of university of sichuan china (license No.: 20170135) and were performed according to the approved guidelines. The volume of the tumor to be detected is about 50mm 3 ~100mm 3 Thereafter, the mice were randomly divided into four groups: (i) placebo (solvent, oral gavage, n = 6); (ii) Idealisib group (25 mg/kg, gavage oral, n = 6); (iii) JQ1 (50 mg/kg, gavage orally, n = 6); (ii) Combination (25 mg/kg Idelalisib +50mg/kg JQ1, gavage orally, n = 6). The solvent is 2.5% dimethyl sulfoxide, 2.5% ethanol, 10% castor oil in normal saline, and the medicine is prepared into medicine solution with required concentration. The preparation is administered by intragastric administration once daily for 18 or 21 days. The long diameter and the short diameter of the tumor are measured by an electronic vernier caliper every 3 days during the administration period, and the calculation formula is as follows: tumor volume (mm) 3 ) =0.52 × length × width 2 . After administration, mice were euthanized and tumors were removed, and Tumor Growth Inhibition (TGI) values were calculated as: TGI = [1- (T) n -T 0 )/ (C n -C 0 )]×100%,T 0 Mean tumor volume, T, before treatment n Mean tumor volume n days after treatment. C 0 Mean tumor volume, C, before treatment for vehicle group n Mean tumor volume n days after vehicle group treatment.
Example 67 in vivo anti-tumor effects on lymphoma (SU-DHL-6 and JEKO-1) xenograft tumor models are shown in FIGS. 5-10.
As can be seen from FIGS. 5-10, the piperazinone substituents of the examples of the present application have better antitumor effect.
The foregoing description is given for the purpose of illustrating particular embodiments of the present application and is not in any way intended to limit the invention, as numerous modifications and variations will be apparent to those skilled in the art. Any modification, equivalent replacement, improvement and the like made within the spirit and principle of the present application shall be included in the protection scope of the present application.

Claims (14)

1. A piperazinone substituent or derivative thereof, wherein the piperazinone substituent has the formula:
Figure FDA0003888899930000011
wherein X is selected from N;
y is selected from NR 3
R 1 Is selected from
Figure FDA0003888899930000012
R 2 Is selected from
Figure FDA0003888899930000013
Wherein Z is selected from N or CH;
R 3 selected from H, C 1-6 Alkyl or C 3-6 A cycloalkyl group;
R 5 selected from hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, tetrahydro-2H-pyran-4-yl, tetrahydro-2H-thiopyran-4-yl, 1-methylpiperidin-4-yl, 1-tetrahydro-2H-thiopyran-4-yl dioxide, 1, 4-dioxaspiro [4.5] o]Decyl-8-yl, C 4~6 Cyclic ketones, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -SO 2 C 1-6 Alkyl or-SO 2 C 3-6 Cycloalkyl radicals, in which C 1-6 Alkyl is substituted with any one or more of the following substituents: hydrogen, halogen, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) OC 1-6 Alkyl and-C (O) NR 9 R 10 ,C 3-6 Cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, oxygen, halogen and-NR 13 R 14
R 6 Selected from hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 4-6 Cyclic ketones, C 2-6 Alkenyl radical, C 2-6 Alkynyl, wherein, C 1-6 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) OC 1-6 Alkyl and-C (O) NR 15 R 16
R 9 Selected from H, C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 10 Selected from H, C 1-6 Alkyl or C 3-6 A cycloalkyl group;
R 13 selected from H, C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 14 Selected from H, C 1-6 Alkyl or C 3-6 A cycloalkyl group;
R 15 selected from H, C 1-6 Alkyl or C 3-6 Cycloalkyl radical, R 16 Selected from H, C 1-6 Alkyl or C 3-6 A cycloalkyl group;
R 19 selected from halogen, cyano, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl or-OC 1-6 An alkyl group;
R 20 selected from H, halogen, methyl, fluoromethyl, difluoromethyl or trifluoromethyl;
the derivative is selected from pharmaceutically acceptable salts of the piperazinone substituent.
2. The piperazinone substituent or derivative thereof as claimed in claim 1, wherein R is 5 Selected from hydrogen, methyl, ethyl, allyl, propargyl, C 3-6 Cycloalkyl, tetrahydro-2H-pyran-4-yl, tetrahydro-2H-thiopyran-4-yl, 1-methylpiperidin-4-yl, 1-tetrahydro-2H-thiopyran-4-yl dioxide, 1, 4-dioxaspiro [4.5] o]Decyl-8-yl, C 4~6 Cyclic ketones, -C (O) C 1-6 Alkyl, -C (O) C 3-6 Cycloalkyl, -C (O) C 2-6 Alkenyl, -SO 2 C 1-3 Alkyl or-SO 2 C 3-6 Cycloalkyl in which methyl and ethyl are substituted by any one or more ofAnd (3) substituent: hydrogen, halogen, C 3-6 Cycloalkyl, -C (O) OC 1-4 Alkyl and-C (O) NR 9 R 10 ,C 3-6 Cycloalkyl is substituted with any one or more of the following substituents: hydrogen, methyl, hydroxy, halogen and-NR 13 R 14
The R is 6 Selected from hydrogen, C 1-2 Alkyl radical, C 3-6 Cycloalkyl or C 4-6 Cyclic ketones of which C 1-2 Alkyl is substituted with any one or more of the following substituents: halogen, C 3-6 Cycloalkyl, -C (O) OC 1-4 Alkyl and-C (O) NR 15 R 16
Said R is 9 Selected from H, C 1-2 Alkyl or C 3-6 Cycloalkyl radical, R 10 Selected from H, C 1-2 Alkyl or C 3-6 A cycloalkyl group;
the R is 13 Selected from H, C 1-2 Alkyl or C 3-6 Cycloalkyl radical, R 14 Selected from H, C 1-2 Alkyl or C 3-6 A cycloalkyl group;
said R is 15 Selected from H, C 1-2 Alkyl or C 3-6 Cycloalkyl radical, R 16 Selected from H, C 1-2 Alkyl or C 3-6 A cycloalkyl group.
3. A piperazinone substituent or derivative thereof as claimed in claim 2 wherein the structural unit of the piperazinone substituent
Figure FDA0003888899930000031
Is selected from
Figure FDA0003888899930000032
4. The piperazinone substituent or derivative thereof as claimed in claim 3, wherein R is 2 Is selected from
Figure FDA0003888899930000033
Figure FDA0003888899930000034
5. The piperazinone substituent or derivative thereof as claimed in claim 4, wherein R is 5 Selected from the group consisting of hydrogen, methyl, ethyl, 2-trifluoroethyl, allyl, propargyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2-amino-2-oxoethyl, 2- (dimethylamino) -2-oxoethyl, 2-methoxy-2-oxoethyl, 2-ethoxy-2-oxoethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 4-hydroxycyclohexyl, 1, 4-dioxaspiro [ 4.5.5 ] ethyl]Decan-8-yl, 4- (dimethylamino) cyclohexyl, 4- (diethylamino) cyclohexyl, 4-difluorocyclohexyl, 4-dimethylcyclohexyl, 1-methylpiperidin-4-yl, tetrahydro-2H-pyran-4-yl, tetrahydro-2H-thiopyran-4-yl, 1-tetrahydro-2H-thiopyran-4-yl dioxide, acetyl, propionyl, butyryl, isobutyryl, tert-butylformyl, cyclopropylformyl, cyclobutylformyl, cyclopentylcarbonyl, cyclohexanoyl, acryloyl, methanesulfonyl, ethylsulfonyl, n-propylsulfonyl, isopropylsulfonyl or cyclopropylsulfonyl;
said R is 6 Selected from the group consisting of hydrogen, methyl, ethyl, 2-trifluoroethyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2- (dimethylamino) -2-oxoethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
6. A piperazinone substituent or derivative thereof, wherein the piperazinone substituent has a structural formula selected from the group consisting of:
Figure FDA0003888899930000041
Figure FDA0003888899930000051
Figure FDA0003888899930000061
Figure FDA0003888899930000071
the derivative is selected from pharmaceutically acceptable salts of the piperazinone substituent.
7. A process for the preparation of a piperazinone substituent as claimed in any one of claims 1 to 6, wherein the piperazinone substituent is prepared by using R 1 By substituting OH in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol with R 2 substituted-Cl in 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol;
wherein R is 1 、R 2 Is as defined in any one of claims 1 to 6.
8. The method of claim 7 wherein the-OH group of 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol is first replaced by-Cl to provide 4- (2-chloro-8- (chloromethyl) -9-methyl-9H-purin-6-yl) morpholine and R is used to prepare the piperazinone substituent 1 A substitution of-Cl.
9. The process for preparing a piperazinone substituent as claimed in claim 7, wherein 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-methanol is prepared by:
reacting a mixture containing 2-chloro-9-methyl-6-morpholinyl-9H-purine-8-carbaldehyde, methanol and sodium borohydride at-5 ℃ to-15 ℃ for 0.5 to 3 hours.
10. A pharmaceutical composition comprising a piperazinone substituent or derivative thereof as claimed in any one of claims 1 to 6.
11. The pharmaceutical composition according to claim 10 wherein said pharmaceutical composition comprises a pharmaceutically acceptable carrier for said piperazinone substituent or a derivative of said piperazinone substituent.
12. Use of a piperazinone substituent or derivative thereof as claimed in any one of claims 1 to 6 in the manufacture of a medicament for the treatment of an autoimmune disease or cancer responsive to inhibition of PI3K δ.
13. Use of a piperazinone substituent or derivative thereof according to claim 12 in the manufacture of a medicament for the treatment of an autoimmune disease or cancer responsive to inhibition of PI3K δ, wherein the autoimmune disease is selected from rheumatoid arthritis, chronic obstructive pulmonary disease, allergic rhinitis or asthma, and the cancer comprises a solid tumor or a hematologic malignancy.
14. Use of a piperazinone substituent or derivative thereof according to claim 12 in the manufacture of a medicament for the treatment of an autoimmune disease or cancer responsive to inhibition of PI3K δ, wherein the cancer is selected from leukemia, lymphoma or a multiple disease myeloma.
CN202010492337.4A 2020-06-02 2020-06-02 Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition Active CN112920199B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202010492337.4A CN112920199B (en) 2020-06-02 2020-06-02 Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition
PCT/CN2021/095581 WO2021244346A1 (en) 2020-06-02 2021-05-24 Piperazinone substitute or derivative thereof, preparation method therefor and application thereof, and pharmaceutical composition

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010492337.4A CN112920199B (en) 2020-06-02 2020-06-02 Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition

Publications (2)

Publication Number Publication Date
CN112920199A CN112920199A (en) 2021-06-08
CN112920199B true CN112920199B (en) 2023-02-03

Family

ID=76163344

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010492337.4A Active CN112920199B (en) 2020-06-02 2020-06-02 Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition

Country Status (2)

Country Link
CN (1) CN112920199B (en)
WO (1) WO2021244346A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023020604A1 (en) * 2021-08-20 2023-02-23 南京大美生物制药有限公司 Five-membered nitrogen-containing heterocyclic and heteroaryl derivative and use thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0423653D0 (en) * 2004-10-25 2004-11-24 Piramed Ltd Pharmaceutical compounds
GB0608264D0 (en) * 2006-04-26 2006-06-07 Piramed Ltd Pharmaceutical compounds
TWI498332B (en) * 2006-04-26 2015-09-01 Hoffmann La Roche Pyrimidine derivatives for use as inhibitors of pi3k and relevant preparation process, pharmaceutical composition, use, kit,and product
GB0721095D0 (en) * 2007-10-26 2007-12-05 Piramed Ltd Pharmaceutical compounds
WO2009053716A1 (en) * 2007-10-26 2009-04-30 F.Hoffmann-La Roche Ag Purine derivatives useful as pi3 kinase inhibitors
EP2435438A1 (en) * 2009-05-27 2012-04-04 Genentech, Inc. Bicyclic pyrimidine pi3k inhibitor compounds selective for p110 delta, and methods of use

Also Published As

Publication number Publication date
CN112920199A (en) 2021-06-08
WO2021244346A1 (en) 2021-12-09

Similar Documents

Publication Publication Date Title
AU2020201114B2 (en) Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
JP6892444B2 (en) Isoindoline, azaisoindoline, dihydroindenone and dihydroazaindenone inhibitors of MNK1 and MNK2
TWI744225B (en) Tyk2 inhibitors and uses thereof
CN112424185B (en) Benzene ring-containing compound, preparation method and application thereof
JP6888006B2 (en) Pyrazolo, pyrazolo, imidazole-pyrimidine and pyridine compounds that inhibit MNK1 and MNK2
EP3453707A1 (en) Benzazepine derivative, preparation method, pharmaceutical composition and use thereof
CN106928219B (en) Nitrogen-containing fused heterocyclic compound, preparation method, intermediate, composition and application
WO2015017610A1 (en) Syk inhibitors
KR20200098598A (en) Substituted bicyclic heterocyclic compounds as PRMT5 inhibitors
AU2014241152A1 (en) Benzimidazolone derivatives as bromodomain inhibitors
JP2022510980A (en) Heteroaromatic derivative regulator, its production method and use
CA2681326A1 (en) Imidazolopyrimidine analogs and their use as pi3 kinase and mtor inhibitors
TW201127385A (en) N-containing heteroaryl derivatives as JAK3 kinase inhibitors
CN116648452A (en) Preparation of heterocyclic compounds as KRAS inhibitors and methods of use thereof
CA2775942A1 (en) Pi3k (delta) selective inhibitors
WO2020155842A1 (en) Polycyclic compound inhibiting mnk1 and mnk2
JP2022523477A (en) A pharmaceutical composition for preventing or treating a pyrrolopyrimidine derivative and a protein kinase-related disease containing the derivative as an active ingredient.
CN112920199B (en) Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition
TW202115080A (en) Azaaryl compounds and application thereof
CN116801883A (en) Heteroaromatic ring compounds, preparation method and application thereof
CN116670144A (en) Tricyclic compounds and uses thereof
AU2021373162A1 (en) Pyrazolopyridazinone compound, and pharmaceutical composition and use thereof
CN111747927B (en) Compounds as immunomodulators and uses thereof
CN116670136A (en) Tetracyclic compound and medical application thereof
CN114380845A (en) Heterocyclic-fused compound, pharmaceutical composition containing same, preparation method and application thereof

Legal Events

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