CN115925694A - PAK4 kinase inhibitor and preparation method and application thereof - Google Patents

PAK4 kinase inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN115925694A
CN115925694A CN202211282529.8A CN202211282529A CN115925694A CN 115925694 A CN115925694 A CN 115925694A CN 202211282529 A CN202211282529 A CN 202211282529A CN 115925694 A CN115925694 A CN 115925694A
Authority
CN
China
Prior art keywords
substituted
cancer
unsubstituted
compound
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202211282529.8A
Other languages
Chinese (zh)
Inventor
刘冠锋
杨茂
李红波
刘龙飞
彭旭
李英富
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.)
Shenzhen Haibowei Pharmaceutical Co ltd
Chengdu Haibowei Pharmaceutical Co ltd
Original Assignee
Shenzhen Haibowei Pharmaceutical Co ltd
Chengdu Haibowei Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Haibowei Pharmaceutical Co ltd, Chengdu Haibowei Pharmaceutical Co ltd filed Critical Shenzhen Haibowei Pharmaceutical Co ltd
Priority to CN202211282529.8A priority Critical patent/CN115925694A/en
Publication of CN115925694A publication Critical patent/CN115925694A/en
Priority to PCT/CN2023/118200 priority patent/WO2024082872A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a compound as shown in a general formula I, or pharmaceutically acceptable salt, stereoisomer, ester, prodrug and solvate thereof. Test results show that the compound provided by the invention has higher inhibitory activity and PAK I/II selectivity on PAK4 kinase, better liver microsome stability and rat PK pharmacokinetic properties, and especially hERG cardiotoxicityThe risk is low and is a significant improvement over the compounds disclosed in the prior art.

Description

PAK4 kinase inhibitor and preparation method and application thereof
Technical Field
The invention belongs to the technical field of drug synthesis, and particularly relates to a PAK4 kinase inhibitor, and a preparation method and application thereof.
Background
P21-activated protein kinases (PAKs), a class of conserved serine/threonine protein kinases, are effector proteins of the small gtpases CDC42 and Rac1 in the Rho family that mediate transduction of their downstream signaling pathways. According to the differences of sequence homology and activation modes, the method can be divided into two main categories: class I PAKs (PAK 1,2, 3) and class II PAKs (PAK 4,5, 6). As important downstream corresponding molecules of Pho family GTPases Rac and Cdc42, PAKs play an important role in the processes of cell proliferation, cytoskeletal recombination and cell movement. Research shows that each member of PAKs, especially the representative members PAK1 and PAK4, has the phenomena of gene amplification, gene mutation, expression level and activity up-regulation in various tumor cells and tissues, and is closely related to the generation and development of tumors. By inhibiting the abnormal activity of PAKs in the tumor cells, the tumor cells are expected to be inhibited from hyperproliferation, invasion and metastasis and angiogenesis, and the apoptosis of the tumor cells is promoted.
In view of this, the study of inhibitors of PAKs has received extensive attention from medicinal chemists in the last decade. Wang C and other researches show that the expression content of PAK4 in lung cancer, colon cancer, prostatic cancer, pancreatic cancer and breast cancer cells is far higher than that of normal cells, and the PAK4 has important influence on the occurrence, development, invasion and migration of tumors. Therefore, the development of PAK4 inhibitors is one of the effective strategies for the treatment of various tumors.
Recent studies have found that inhibition of class I PAKs has a potential correlation with safety risks such as cardiac acute toxicity, hERG side effects, etc., suggesting that development of inhibitors of PAKs should avoid inhibitory effects on class I PAKs, particularly PAK 1. Therefore, the discovery of highly selective class II inhibitors of PAKs will become the mainstay of future research.
PAK4 is a potential drug development target, and the development of inhibitors thereof provides a new idea for treating related cancers. To date, inhibitors of PAK4 have been in small quantities and the activity of most inhibitors has been unsatisfactory. In addition, the pharmacokinetic property is poor, and the problem of druggability is a common problem of reported molecules. The small molecule inhibitors reported to enter clinical stage are ATG-019 (KPT-9274) developed by De Qi pharmaceutical company and Karyopharm Therapeutics company and PF-3758309 developed by Peprore. Among them, PF-3758309 is a class I PAKs inhibitor with pyrrolopyrazole structure reported in 2009 by feverre corporation, and is the PAKs inhibitor that first entered clinical studies, and its PAK4 IC50 is 19nm, but this compound has stronger inhibitory ability to PAK1, reaching 14nm, and has serious safety risk. In addition, phase I clinical studies were forced to terminate due to poor oral bioavailability, only about 1%, and severe gastrointestinal adverse effects. ATG-019 is a globally initiated p21-activated kinase 4 (PAK 4) and nicotine phosphoribosyl transferase (NAMPT) oral dual-target inhibitor with unknown action mechanism. Several phase I clinical studies are currently being conducted in fields including non-hodgkin's lymphoma, colorectal cancer, lung cancer, melanoma, and the like.
Disclosure of Invention
The compounds developed earlier by the company and capable of serving as PAK4 kinase inhibitors are disclosed in patent WO2022033420A1, and all the compounds have better PAK4 activity and PAK I/II selectivity, and are greatly improved in terms of pharmacokinetic properties compared with the reported PAK4 kinase inhibitors. However, in later continued studies, the inventors have found that these compounds have a strong hERG cardiac inhibitory activity, resulting in an increased risk of cardiotoxic side effects. In view of the above, the invention optimizes the structure of the compound on the basis of the previous research, and screens a series of compounds with higher inhibitory activity and PAK I/II selectivity on PAK4 kinase, wherein the compounds not only have excellent liver microsome stability and rat PK pharmacokinetic properties, but also greatly reduce the hERG heart inhibitory activity and solve the problem of cardiotoxicity risk.
The invention aims to provide a compound with a general formula I and a preparation method thereof, wherein the compound is a PAK4 kinase inhibitor; it is another object of the present invention to provide a use of the compound.
The technical scheme of the invention comprises the following contents:
in a first aspect, the present invention provides a compound of formula i, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure SMS_1
wherein A is 1 、A 2 、A 3 、A 4 Independently selected from C or N, when A 1 、A 2 、A 3 、A 4 Is N, R connected to said N 2 、R 3 、R 4 、R 5 Is absent;
in a preferred embodiment of the present invention, A is 1 、A 2 、A 3 、A 4 Is selected from C.
R 2 、R 3 、R 4 、R 5 Independently selected from-H, halogen, -OH, -CN, -NH 2 、-NO 2 SH, C1-10 straight chain/branched chain alkyl, C3-10 naphthenic base, heterocyclic alkyl, alkynyl, alkenyl, aromatic base, heterocyclic aromatic base, amido, ester group, sulfonyl and phosphoryl, wherein H in the groups can be substituted by the following groups: halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, substituted or unsubstituted aryl or heteroaryl.
The substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyl amino, alkyl, cycloalkyl, heterocyclic alkyl, aryl, heterocyclic aryl, ester group, acyl, carbonyl, amide, sulfonyl, phosphoryl.
The cycloalkyl or heterocycloalkyl described in the present invention includes monocyclic, bridged, spiro or fused cycloalkyl or heterocycloalkyl, and further, the cycloalkyl or heterocycloalkyl includes both saturated and unsaturated forms.
B 1 、B 2 Independently selected from C or N, in a preferred embodiment of the invention, said B 1 、B 2 Is N.
L is selected from-O-, -S-, -NH-or alkylene; preferably, L is-NH-.
R 1 Selected from substituted or unsubstituted five-membered or six-membered aromatic or heterocyclic aromatic groups, substituted or unsubstituted heterocycloalkyl groups containing at least one N and/or O atom.
Preferably, said R 1 Selected from the group consisting of substituted or unsubstituted phenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, furyl, thienyl, pyrrolyl, five or six membered heterocycloalkyl containing at least one N or O atom.
In a particular embodiment of the invention, R 1 Has any one of the following structures:
Figure SMS_2
h on any one or more C atoms in the above structure may be substituted with: -F, -Cl, -Br-OH, -CN, -NH 2 Substituted or unsubstituted amido, substituted or unsubstituted C1-3 alkyl or alkoxy, C3-6 cycloalkyl.
Preferably, the substituent groups are independently selected from-F, -Cl, -Br, -OH, -CN, -NH 2 Methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl, hydroxyethyl.
R 6 Selected from-H, -NH 2 OH, -halogen, amide, sulfonyl, sulfonic group, C1-6 alkyl or alkoxy, C3-6 cycloalkyl, C3-6 heterocycloalkyl, amino, C6-12 aryl, C5-12 heterocycleAn aromatic group; the above alkyl or alkoxy group of C1-6, cycloalkyl group of C3-6, heterocycloalkyl group of C3-6, amino group, aryl group of C6-12, and H on the heteroaryl group of C5-12 may be optionally substituted by one or more halogens, -OH, -CN, -NH 2 、-NO 2 SH and sulfonic acid group.
The ring Ar is fused with the ring Rg, and the fused bond is any bond on the ring Ar;
ring Ar is selected from aromatic five-membered heterocyclic group, aromatic six-membered heterocyclic group or phenyl, the aromatic five-membered heterocyclic group is selected from: imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, furanyl or thienyl; the aromatic six-membered heterocyclic group is selected from: pyridyl, pyridazinyl, pyrimidinyl or pyrazinyl; h on the aromatic five-membered heterocyclic group, the aromatic six-membered heterocyclic group or the phenyl group which is optional may be substituted by: halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, substituted or unsubstituted aryl or heteroaryl. The substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyl amino, alkyl, cycloalkyl, heterocyclic alkyl, aryl, heterocyclic aryl, ester group, acyl, carbonyl, amide, sulfonyl, phosphoryl.
Ring Rg is selected from C3-8 saturated/unsaturated cycloalkyl or C3-8 saturated/unsaturated heterocycloalkyl containing at least one O, N, S, and H on said C3-8 saturated/unsaturated cycloalkyl or C3-8 saturated/unsaturated heterocycloalkyl containing at least one O, N, S may be substituted with: halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, substituted or unsubstituted aryl or heteroaryl. The substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyl amino, alkyl, cycloalkyl, heterocyclic alkyl, aryl, heterocyclic aryl, ester group, acyl, carbonyl, amide, sulfonyl, phosphoryl.
Further, having at least one R on the ring Rg 7 Said R is 7 Independently selected from-H, deuterium, tritium, halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyamino, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, aryl, heteroaryl, ester, acyl, carbonyl, amide, sulfonyl, phosphoryl.
Preferably, the compound has a structure shown in formula II, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure SMS_3
x is selected from substituted or unsubstituted C1-6 straight chain/branched alkyl, substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, substituted or unsubstituted C5-6 aryl or heteroaryl, alkynyl, alkenyl, amido and ester. The substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyl amino, alkyl, alkoxy, hydroxyl alkyl, cycloalkyl, heterocyclic alkyl, aromatic base, heterocyclic aromatic base, ester base, acyl, carbonyl, amide, sulfonyl, phosphoryl.
The cycloalkyl or heterocycloalkyl described in the present invention includes monocyclic, bridged, spiro or fused cycloalkyl or heterocycloalkyl, and further, the cycloalkyl or heterocycloalkyl includes both saturated and unsaturated forms.
Having at least one R on the X group 8 Said R is 8 Independently selected from-H, halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, said substituent being selected from the group consisting of halogen, -OH, -CN, -NH 2 、-NO 2 、-SH、-CF 3 、-CHF 2 、-CH 2 F. Carboxyl, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, phenoxy, pyridyl, amido, sulfonyl, phosphoryl.
Further preferred isWherein the substituted groups are independently selected from-F, -Cl, -Br, -OH, -CN, -NH 2 、-CF 3 Hydroxymethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
R 9 Selected from substituted or unsubstituted phenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, the substituents are independently selected from-F, -Cl, -Br, -OH, -CN, -NH 2 Methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl.
R 10 Is selected from-NH 2 OH, -OH, halogen, C1-6 alkyl or alkoxy, C3-6 cycloalkyl, C3-6 heterocycloalkyl; h on the above C1-6 alkyl or alkoxy, C3-6 cycloalkyl, C3-6 heterocycloalkyl may optionally be substituted with one or more halogens, -OH, -CN, -NH 2 、-NO 2 -SH substitution.
The ring Rg is selected from C3-8 saturated/unsaturated heterocycloalkyl containing at least one of O, N, S, and having at least one R 11 ,R 11 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, -OH, -CN, -NH 2 、-NO 2 SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, phenoxy, sulfonyl, phosphoryl.
In a preferred embodiment of the invention, the compound has the structure shown in formula iii, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure SMS_4
R 12 selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, and,Cyclopentyl, cyclohexyl, hydroxymethyl.
R 13 、R 14 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
R 15 Selected from-H, -F, -Br, -OH, -CN, -NH 2 Substituted or unsubstituted C1-10 straight/branched alkyl, substituted or unsubstituted C1-10 straight/branched alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, wherein H in the above groups may be substituted with: -F, -Cl, -Br-OH, -CN, -NH 2 、-CF 3 Hydroxymethyl, methyl, ethyl, propyl, isopropyl, phenyl, methoxy, ethoxy, propoxy, isopropoxy, phenoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
The cycloalkyl or heterocycloalkyl group includes monocyclic, bridged, spiro or fused cycloalkyl or heterocycloalkyl groups, and further, the cycloalkyl or heterocycloalkyl group includes both saturated and unsaturated forms.
In particular embodiments of the present invention, the substituted or unsubstituted cycloalkyl or heterocycloalkyl includes, but is not limited to
Figure SMS_5
Figure SMS_6
Denotes a position which may be linked to an alkynyl group.
In a preferred embodiment of the invention, the compound has the structure shown in formula iv, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure SMS_7
R 16 、R 17 independently selected from-H, -F, -Cl, -Br, -OH, -CN, -NH 2 -SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxyAlkyl, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and phenyl.
R 18 Selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl.
R 19 、R 20 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
In a preferred embodiment of the invention, the compound has the structure of formula v, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure SMS_8
R 21 selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl.
R 22 、R 23 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
Y is selected from substituted or unsubstituted C1-4 straight chain/branched alkyl, substituted or unsubstituted C3-8 cycloalkyl or heterocycloalkyl, substituted or unsubstituted C5-6 aryl or heteroaryl, amide and ester. The substituent group is selected from-OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyamino, alkyl, alkoxy, hydroxyalkyl, substituted or unsubstituted C3-8 cycloalkyl or heterocycloalkyl, C5-6 aryl or heteroaryl, ester, acyl, carbonyl, amide, sulfonyl, or phosphoryl.
The R is 24 Selected from-H, deuterium, tritium, -F, -Cl, -Br, -OH, -CN, -NH 2 、-NO 2 SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, phenyl, phenoxy, substituted or unsubstituted C3-8 saturated or unsaturated cycloalkyl or heterocycloalkyl.
The substituted or unsubstituted C3-8 cycloalkyl or heterocycloalkyl includes, but is not limited to
Figure SMS_9
Figure SMS_10
Figure SMS_11
Indicating a position to which Y can be attached. H in the above structure may be substituted with: -OH, -CN, -NH 2 、-NO 2 SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, phenyl, phenoxy, sulfonyl, phosphoryl.
In a most preferred embodiment of the invention, the compound has the following structure:
Figure SMS_12
/>
Figure SMS_13
in a second aspect, the present invention provides a pharmaceutical composition comprising a compound of any one of formulas i-v, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof.
Preferably, the pharmaceutical composition further comprises pharmaceutically acceptable excipients, which include but are not limited to: carrier, diluent, adhesive, lubricant and wetting agent.
Preferably, the pharmaceutical composition comprises a therapeutically effective amount of a compound according to any of formulae i-v. In certain embodiments, the pharmaceutical compositions may be used alone or in combination with other agents.
The pharmaceutical compositions are suitable for gastrointestinal or parenteral administration, such as by intravenous, intramuscular, intradermal and subcutaneous routes. Preferably, the pharmaceutical composition further comprises antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the subject, as well as aqueous and non-aqueous sterile suspending agents that may include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
The compounds of any of the general formulae I-V provided herein may be formulated into pharmaceutical preparations in the form of: injections, syrups, elixirs, suspensions, powders, granules, tablets, capsules, lozenges, creams, ointments, lotions, gels, emulsions and the like.
In a third aspect, the present invention provides a use of a compound of any one of formulas i-v, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof, or a pharmaceutical composition comprising a compound of any one of formulas i-v, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof, for the manufacture of a medicament for the treatment of a disease associated with the expression or activity of PAK4 kinase.
The diseases related to the expression or activity of PAK4 kinase comprise cancer, neurodegenerative diseases or immune system diseases.
The cancer includes breast cancer, mantle cell lymphoma, ovarian cancer, esophageal cancer, laryngeal cancer, glioblastoma, neuroblastoma, gastric cancer, hepatocellular cancer, gastric cancer, glioma, endometrial cancer, melanoma, renal cancer, bladder cancer, melanoma, bladder cancer, biliary tract cancer, renal cancer, pancreatic cancer, lymphoma, hairy cell cancer, nasopharyngeal cancer, pharyngeal cancer, large bowel cancer, rectal cancer, brain and central nervous system cancer, cervical cancer, prostate cancer, testicular cancer, genitourinary tract cancer, lung cancer, non-small cell lung cancer, small cell cancer, lung adenocarcinoma, bone cancer, colon cancer, adenoma, pancreatic cancer, adenocarcinoma, thyroid cancer, follicular cancer, hodgkin's leukemia, bronchial cancer, thyroid cancer, uterine body cancer, cervical cancer, multiple myeloma, acute myelogenous leukemia, chronic myelogenous leukemia, lymphocytic leukemia, chronic lymphoid leukemia, myeloid leukemia, myelogenous leukemia, non-hodgkin's lymphoma, primary macroglobulinemia, rhabdomyosarcoma.
Preferably, the compounds of any of said formulae I-V or pharmaceutically acceptable salts, stereoisomers, esters, prodrugs, solvates thereof are used alone or in combination with other types of pharmaceutical agents and/or methods of treatment.
Such other classes of pharmaceutical formulations and/or methods of treatment include, but are not limited to: immunosuppressants, targeted antineoplastic drugs, glucocorticoids, non-steroidal anti-inflammatory drugs, anti-tumor vaccines, adoptive cellular immunotherapy, chemotherapy or radiotherapy.
In a fourth aspect, the invention provides a compound of formula VI,
Figure SMS_14
wherein R is 25 、R 26 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
In a fifth aspect, the present invention provides a compound of formula VII,
Figure SMS_15
wherein R is 27 Selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl.
R 28 、R 29 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
In a sixth aspect, the present invention provides the use of a compound of formula vi and/or formula vii in the preparation of any one of the following compounds, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
(a) A compound of formula I;
(b) A compound described by formula II;
(c) A compound of formula III;
(d) A compound of formula IV;
(e) A compound of formula V.
The technical scheme provided by the invention has the following technical advantages: (1) Compared with the prior art, the compound shown in the general formula I-V or pharmaceutically acceptable salt, stereoisomer, ester, prodrug and solvate thereof provided by the invention have higher inhibitory activity and PAK I/II selectivity as a PAK4 inhibitor, and meanwhile, the stability of liver microsome and rat PK are also improved; (2) Furthermore, the compound of the general formula I or pharmaceutically acceptable salts, stereoisomers, esters, prodrugs and solvates thereof provided by the invention are used as PAK4 inhibitor hERG heart inhibitory activity, so that the risk problem of drug cardiotoxicity is solved.
Detailed Description
The technical solutions in the embodiments of the present invention will be clearly and completely described below, and it is obvious that the described embodiments are only some embodiments of the present invention, and not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
The abbreviations appearing in the following examples have the meanings indicated below:
Et 3 n: triethylamine;
EA: ethyl acetate;
THF: tetrahydrofuran;
MeOH: methanol;
DIEA: n, N-diisopropylethylamine;
n-BuLi: n-butyl lithium;
m: molar concentration units mol/L, e.g., 1M means 1mol/L;
n: normality, for example 1N HCl means hydrochloric acid at a concentration of 1mol/L;
HATU: o- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate;
HBTU: benzotriazole-N, N, N ', N' -tetramethyluronium hexafluorophosphate;
m-CPBA: m-chloroperoxybenzoic acid;
NMO: n-methylmorpholine oxide;
DPPA: diphenyl phosphorazidate;
DMF: n, N-dimethylformamide;
TBDPSCl: tert-butyldiphenylchlorosilane;
DIBAL-H: diisobutylaluminum hydride;
TLC: thin-layer chromatography;
PE: petroleum ether (boiling point 60-90 ℃);
DCM: dichloromethane;
H2O: distilled water;
DMSO, DMSO: dimethyl sulfoxide;
Pd(PPh 3 ) 2 Cl 2 : bis (triphenylphosphine) palladium dichloride;
Pd(dppf)Cl 2 DCM: [1,1' -bis (diphenylphosphino) ferrocene]A palladium dichloride dichloromethane complex;
HCl/1,4-dioxane: hydrochloric acid/1, 4-dioxane solution;
TBAF.3H 2 o: tetrabutylammonium fluoride trihydrate;
1,4-dioxane:1, 4-dioxane.
Preparation of intermediates
The preparation of intermediate 1 was carried out using the following synthetic route:
Figure SMS_16
the preparation of the compound 4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-4-amine dihydrochloride is described in detail as follows:
Figure SMS_17
step 1: preparation of 4- (benzo [ d ] [1,3] dioxo-5-yl) -4-cyanopiperidine-1-carboxylic acid tert-butyl ester
Weighing 2- (benzo [ d ]][1,3]Dioxol-5-yl) acetonitrile (10.00g, 62.03mmol) and tert-butyl bis (2-chloroethyl) carbamate (16.52g, 68.23mmol) were placed in a reaction flask, DMF (120 mL) was added, replaced with nitrogen twice, and cooled to about 0 ℃. Add NaH (9.93g, 248.12mmol,60% in oil) in portions, keeping the system temperature between 0-10 ℃. After the addition, the temperature of the system is raised to 60 ℃ for reaction for 2 hours. The heating was turned off, and the temperature was naturally decreased to room temperature with stirring, and the reaction was carried out for 14 hours. Sampling TLC plate detection, after the raw material reaction is finished, slowly adding the reaction liquid into saturated NH 4 After quenching in aqueous Cl (600 mL), EA (200ml × 2) was added, extracted 2 times, the organic phases were combined and washed 3 times with saturated aqueous NaCl (100ml × 3), dried over anhydrous sodium sulfate and concentrated to give a brown solid (crude) which was used directly in the next reaction.
EM (calculated): 330.2; MS (ESI) M/z (M + H) + :331.3
Step 2: preparation of 4- (benzo [ d ] [1,3] dioxo-5-yl) -4-carbamoylpiperidine-1-carboxylic acid tert-butyl ester
Reacting compound 4- (benzo [ d ]][1,3]Dioxo-5-yl) -4-cyanopiperidine-1-carboxylic acid tert-butyl ester (crude) and NaOH (24.81g, 620.30mmol) are put into a reaction bottle, DMSO (140 mL) is added, after the temperature is raised to 60 ℃, H is slowly dropped 2 O 2 (61 mL). After the addition, a sample was taken to monitor the reaction. After the reaction is finished, cooling to room temperature. The reaction was poured slowly into water (1120 mL) to yield a large amount of a pale yellow solid. After stirring for 20 minutes, filtration was carried out and the filter cake was rinsed 3 times with water. And drying the filter cake to obtain a light yellow solid (crude product) which is directly used for the next reaction.
EM (calculated): 348.2; MS (ESI) M/z (M + H) + :349.3
And 3, step 3: preparation of tert-butyl 4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidine-1-carboxylate
Tert-butyl 4- (benzo [ d ] [1,3] dioxo-5-yl) -4-carbamoylpiperidine-1-carboxylate (crude) was weighed into a reaction flask and acetonitrile (250 mL) was added. An aqueous solution of KOH (13.89g, 248.12mmol) was added (KOH was previously added to 125mL of water and dispersed) with stirring, and the reaction was carried out in an open reaction flask. Dibromo hydantoin (10.64g, 37.22mmol) was slowly added at room temperature, and after the addition, the system was gradually dissolved and clear, the color was light, and the reaction was carried out at room temperature for 2 hours. After the reaction was monitored by TLC, stirring was stopped, liquid was separated, and the organic phase was collected and concentrated to remove most of the solvent. The aqueous phase was extracted 2 times with DCM/MeOH =10/1 (100ml × 2). The concentrated residue was combined with the extract, washed 1 time with saturated aqueous NaCl solution, dried with anhydrous sodium sulfate, and concentrated to dryness to give a black oil (crude) which was used directly in the next reaction.
EM (calculated): 320.2; MS (ESI) M/z (M + H) + :321.3
And 4, step 4: preparation of 4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-4-amine dihydrochloride
Tert-butyl 4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidine-1-carboxylate (crude) was weighed into a reaction flask and HCl/1,4-dioxane (127mL, 4M) was added. Stirring at room temperature for about 10 min to precipitate solid, and stirring for 1 hr to complete the reaction. The reaction solution was directly filtered, and the filter cake was rinsed with a small amount of 1,4-dioxane to give a grayish solid compound (15.70 g, 86.4% in the four-step reaction).
EM (calculated): 220.1 of the total weight of the mixture; MS (ESI) M/z (M + H) + :221.2
Intermediate 2
Preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine
Figure SMS_18
Step 1: preparation of 2-chloro-N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine
The compound 2, 4-dichloro-6-iodoquinazoline (5.00g, 15.39mmol) was dissolved in DMF (30 mL), to which was added 5-cyclopropyl-4-fluoro-1H-pyrazol-3-amine (2.20g, 15.44mmol) and DIEA (7.90g, 61.56mmol) in that order at room temperature. The temperature was raised to 65 ℃ and stirred for 2 hours. After the reaction of the raw materials is finished, cooling the reaction liquid to room temperature. The reaction mixture was slowly poured into water (300 mL) and extracted 2 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution and dried over anhydrous sodium sulfate. Concentrate to dryness, and purify the residue by column chromatography (DCM/MeOH = 50/1) to obtain the target compound (6.02 g, 91.2% yield) as a yellow solid.
EM (calculated): 429.0; MS (ESI) M/z (M + H) + :430.1
Step 2: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine
The compound 2-chloro-N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (6.02g, 14.01mmol) was dissolved in DMF (30 mL), and 4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-4-amine dihydrochloride (4.. 111g, 14.01mmol), KI (4.65g, 28.02mmol) and DIEA (7.23g, 56.04mmol) were added thereto in this order at room temperature. The temperature was raised to 120 ℃ and stirred for 2 hours. After the reaction of the raw materials is finished, cooling the reaction liquid to room temperature. The reaction was poured slowly into water (300 mL) and extracted 2 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl and dried over anhydrous sodium sulfate. Concentrate to dryness, and purify the residue by column chromatography (DCM/MeOH = 50/1) to give the title compound (5.42 g, 63.1% yield) as a yellow solid.
EM (calculated): 613.1; MS (ESI) M/z (M + H) + :614.1
Preparation of the Compounds of examples
Example 1
Preparation of 4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) tetrahydro-2H-pyran-4-ol
Figure SMS_19
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (50mg, 0.08mmol) and 4-ethyltetrahydro-2H-pyran-4-ol (30mg, 0.24mmol) were added to THF (2 mL), to which CuI (4mg, 0.02mmol), et 3 N (24mg, 0.24mmol) and Pd (PPh) 3 ) 2 Cl 2 (15mg, 0.02mmol), and stirred at 50 ℃ for 4 hours under a nitrogen blanket. After completion of the reaction, water (20 mL) was added to the reaction mixture, and extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 25/1) to obtain the target compound (13 mg, yield 26.6%) as a white solid.
EM (calculated): 611.3; MS (ESI) M/z (M + H) + :612.4
1 H NMR(400MHz,DMSO-d 6 )δ0.75-0.79(2H,m),0.91-0.98(2H,m),1.53-1.56(2H,m),1.69-1.81(4H,m),1.83-1.90(3H,m),1.92-2.11(2H,brs),3.42-3.45(2H,m),3.59-3.63(2H,m),3.75-3.82(2H,m),4.24-4.26(2H,m),5.75(1H,s),5.95(2H,s),6.80(1H,d,J=8.0Hz),6.93(1H,d,J=8.0Hz),7.12(1H,s),7.27(1H,d,J=8.0Hz),7.52(1H,d,J=12.0Hz),8.39(1H,s),9.85(1H,s),12.39(1H,s).
The example compounds shown in table 1 below were synthesized according to the method described in example 1:
TABLE 1
Figure SMS_20
/>
Figure SMS_21
/>
Figure SMS_22
/>
Figure SMS_23
/>
Figure SMS_24
Example 10
Preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-ethynyl quinazolin-4-amine
Figure SMS_25
Step 1: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6- ((trimethylsilyl) ethynyl) quinazolin-4-amine
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (90mg, 0.15mmol) and trimethylsilylacetylene (60mg, 0.60mmol) were added to DMF (2 mL), to which CuI (4mg, 0.02mmol), et 3 N (45mg, 0.45mmol) and Pd (PPh) 3 ) 2 Cl 2 (22mg, 0.03mmol) was stirred at 45 ℃ for 4 hours under nitrogen. After completion of the reaction, water (20 mL) was added to the reaction mixture, and extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 40/1) to obtain the target compound (65 mg, yield 74.3%) as a yellow solid.
EM (calculated): 583.3; MS (ESI) M/z (M + H) + :584.4
Step 2: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-ethynyl quinazolin-4-amine
The compound 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6- ((trimethylsilyl) ethynyl) quinazolin-4-amine (65mg, 0.11mmol) was dissolved in methanol (5 mL), to which potassium carbonate (30mg, 0.22mmol) was added, and stirred at room temperature for 4 hours. After completion of the reaction, the reaction solution was filtered, the filtrate was concentrated to dryness, and the obtained crude product was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (15 mg, yield 26.7%) as a white solid.
EM (calculated): 511.2; MS (ESI) M/z (M + H) + :512.3
1 H NMR(400MHz,DMSO-d 6 )δ0.76-0.79(2H,m),0.93-0.95(2H,m),1.52-1.55(2H,m),1.74-1.77(2H,m),1.84-1.86(1H,m),1.87-1.90(2H,brs),3.39-3.42(2H,m),4.13(1H,s),4.25-4.35(2H,m),5.95(2H,s),6.79(1H,d,J=7.6Hz),6.91(1H,d,J=8.4Hz),7.12(1H,s),7.26(1H,d,J=8.4Hz),7.55(1H,dd,J=1.6Hz,8.4Hz),8.41(1H,s),9.83(1H,s),12.31(1H,s).
Example 11
Preparation of 4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) -1, 1-trifluorobutyl-3-yn-2-ol
Figure SMS_26
Step 1: preparation of 1, 1-trifluorobutyl-3-yn-2-ol
The compound trimethylsilylacetylene (5.19g, 52.79mmol) was added to anhydrous THF (30 mL), cooled to-60 deg.C, and n-BuLi (31.67mL, 63.35mmol,2M in n-hexane) was added slowly thereto. After stirring for 1 hour, ethyl trifluoroacetate (5.00g, 35.19mmol) was slowly added dropwise thereto, and stirring was continued for 5 hours while maintaining the temperature. After the reaction is finished, adding saturated NH into the reaction liquid 4 The Cl aqueous solution was quenched and stirred by adding water (50 mL) and EA (50 mL). Separating, extracting the aqueous phase with EA for 1 time, combining the organic phases, washing with saturated NaCl aqueous solution for 1 time, drying with anhydrous sodium sulfate, and concentrating to dryness. The residue was dissolved in methanol (30 mL), cooled to 0 ℃ using an ice-water bath, and then sodium borohydride (4.00g, 105.57mmol) was added in portions and stirred at room temperature overnight. After completion of the reaction, water (50 mL) was slowly added to the reaction solution, followed by extraction with ethyl acetate 2 times. The organic phases were combined, washed 1 time with saturated aqueous NaCl solution, dried over anhydrous sodium sulfate, and concentrated to dryness to give the objective compound (3.90 g, yield 89.4%) as a colorless oil.
EM (calculated): 124.0; MS (ESI) M/z (M + H) + :125.0
Step 2: preparation of 4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) -1, 1-trifluorobutyl-3-yn-2-ol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (100mg, 0.16mmol) and 1, 1-trifluorobutyl-3-yn-2-ol (40mg, 0.32mmol) were added to DMF (5 mL) to which CuI (4mg, 0.02mmol), et 3 N (48mg, 0.48mmol) and Pd (PPh) 3 ) 2 Cl 2 (22mg, 0.03mmol) was stirred at 45 ℃ for 4 hours under nitrogen. After completion of the reaction, water (50 mL) was added to the reaction mixture, and extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, it is concentrated to dryness and the crude product obtained is purified by column chromatography (DCM/MeOH/NH) 3 .H 2 O = 20/1/0.05) to give the target compound (46 mg, yield 47.2%) as a white solid.
EM (calculated): 609.2; MS (ESI) M/z (M + H) + :610.3
1 H NMR(400MHz,DMSO-d 6 )δ0.76-0.78(2H,m),0.93-0.95(2H,m),1.57-1.63(2H,m),1.84-1.87(3H,m),3.45-3.48(2H,m),4.20-4.29(2H,m),5.23-5.28(1H,m),5.96(2H,s),6.82(1H,d,J=8.0Hz),6.93(1H,d,J=2.0Hz),7.13-7.15(2H,m),7.29(1H,d,J=8.8Hz),7.56(1H,d,J=8.8Hz),8.47(1H,s),9.92(1H,s),12.32(1H,s).
Example 12
Preparation of 4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) but-3-yne-1, 2-diol
Figure SMS_27
Step 1: preparation of 1- ((tert-butyldimethylsilyl) oxy) but-3-yn-2-ol
The compound 2- ((tert-butyldimethylsilyl) oxy) acetaldehyde (200mg, 1.15mmol) was added to anhydrous THF (10 mL), cooled to about 0 ℃ with an ice-water bath, to which was slowly added ethynylmagnesium bromide (2.76mL, 1.38mmol,0.5M in THF), allowed to return to room temperature and stirred for 2 hours. After completion of the reaction, water (50 mL) was slowly added to the reaction mixture, followed by extraction with ethyl acetate 2 times. The organic phases were combined, washed 1 time with a saturated aqueous NaCl solution, dried over anhydrous sodium sulfate, and concentrated to dryness to obtain the objective compound (189 mg, yield 82.2%) as a yellow oil.
EM (calculated): 200.1 of the total weight of the mixture; MS (ESI) M/z (M + H) + :201.1
Step 2: preparation of but-3-yne-1, 2-diol
The compound 1- ((tert-butyldimethylsilyl) oxy) but-3-yn-2-ol (189mg, 0.94mmol) was added to 2M HCl/MeOH (5 mL), warmed to 45 ℃ and stirred for 5 h. After the completion of the reaction, the reaction mixture was concentrated to dryness to obtain the objective compound (156 mg, crude product) as a black oil.
And 3, step 3: preparation of 4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) but-3-yne-1, 2-diol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (70mg, 0.11mmol) and but-3-yne-1, 2-diol (156 mg, crude) were added to DMF (5 mL), to which was added CuI (4 mg, 0.02mmol), et 3 N (33mg, 0.33mmol) and Pd (PPh) 3 ) 2 Cl 2 (14mg, 0.02mmol), and stirred at 45 ℃ for 3 hours under nitrogen. After completion of the reaction, water (50 mL) was added to the reaction mixture, and extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (13 mg, yield 20.7%) as a pale yellow solid.
EM (calculated): 571.2, respectively; MS (ESI) M/z (M + H) + :572.3
1 H NMR(400MHz,DMSO-d 6 )δ0.78-0.82(2H,m),0.97-1.01(2H,m),1.56-1.59(2H,m),1.76-1.78(2H,m),1.85-1.90(1H,m),3.41-3.45(2H,m),3.62-3.68(2H,m),4.36-4.40(2H,m),4.49-4.55(1H,m),4.71-4.75(1H,m),5.12(1H,d,J=6.4Hz),5.95(2H,s),6.77(1H,d,J=7.6Hz),6.87(1H,d,J=8.0Hz),7.12(1H,s),7.24(1H,d,J=6.4Hz),7.45(1H,d,J=8.0Hz),8.31(1H,s),9.75(1H,s),12.33(1H,s).
Example 13
Preparation of 4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) cyclohexan-1-ol
Figure SMS_28
Step 1: preparation of methyl 4- (tert-butyldiphenylsilyl) oxy) cyclohexane-1-carboxylate the compound methyl 4-hydroxycyclohexanecarboxylate (2.10 g, 13.28mmol) was added to DCM (50 mL), to which imidazole (0.90g, 13.28mmol), TBDPSCl (3.65g, 13.28mmol) were successively added, and stirred at room temperature for 1 hour. After completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 100/1) to obtain the objective compound (4.80 g, yield 91.3%) as a colorless oil.
EM (calculated): 396.2; MS (ESI) M/z (M + H) + :397.2
Step 2: preparation of 4- ((tert-butyldiphenylsilyl) oxy) cyclohexane-1-carbaldehyde
The compound methyl 4- (tert-butyldiphenylsilyl) oxy) cyclohexane-1-carboxylate (1.00g, 2.52mmol) was added to anhydrous DCM (15 mL), to which DIBAL-H (3.0 mL,3.03mmol,1.0M in n-hexane) was slowly added dropwise, and stirred at room temperature for 1 hour. After completion of the reaction, water was added to the reaction solution and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, the reaction solution was concentrated to dryness to obtain the objective compound (911 mg, yield 98.7%) as a colorless oil.
EM (calculated): 366.2; MS (ESI) M/z (M + H) + :367.2
And step 3: preparation of tert-butyl ((4-ethynylcyclohexyl) oxy) diphenylsilane
Compound 4- ((tert-butyldiphenylsilyl) oxy) cyclohexane-1-carbaldehyde (800mg, 2.18mmol) was added to MeOH (15 mL), to which were added potassium carbonate (752mg, 5.45mmol) and dimethyl (1-diazo-2-oxopropyl) phosphonate (837mg, 4.36mmol) in this order, and the mixture was stirred at room temperature overnight. After completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 80/1) to obtain the objective compound (431 mg, yield 53.9%) as a colorless oil.
EM (calculated): 362.2; MS (ESI) M/z (M + H) + :363.2
And 4, step 4: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -6- (4- ((tert-butyldiphenylsilyl) oxy) cyclohexyl) ethynyl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) quinazolin-4-amine
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (100mg, 0.16mmol) and tert-butyl ((4-ethynylcyclohexyl) oxy) diphenylsilane (116mg, 0.32mmol) were added to DMF (5 mL), to which CuI (4mg, 0.02mmol), et 3 N (48mg, 0.48mmol) and Pd (PPh) 3 ) 2 Cl 2 (22mg, 0.03mmol) was stirred at 45 ℃ for 4 hours under nitrogen. After completion of the reaction, water (50 mL) was added to the reaction mixture, and the mixture was extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 30/1) to obtain the target compound (67 mg, yield 49.4%) as a yellow oil.
EM (calculated): 847.4; MS (ESI) M/z (M + H) + :848.5
And 5: preparation of 4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) cyclohexan-1-ol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -6- (4- ((tert-butyldiphenylsilyl) oxy) cyclohexyl) ethynyl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) quinazolin-4-amine (67mg, 0.08mmol) was dissolved in THF (5 mL), to which TBAF.3H was added 2 O (76mg, 0.24mmol), and stirred at room temperature overnight. After completion of the reaction, water (20 mL) was added to the reaction mixture, and the mixture was extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (13 mg, yield 26.7%) as a pale yellow solid.
EM (calculated): 609.3; MS (ESI) M/z (M + H) + :610.4
1 H NMR(400MHz,DMSO-d 6 )δ0.76-0.80(2H,m),0.93-0.97(2H,m),1.39-1.47(2H,m),1.52-1.58(2H,m),1.59-1.65(1H,m),1.74-1.81(1H,m),1.83-1.87(4H,m),1.94-2.01(4H,m),3.40-3.47(4H,m),4.28-4.39(1H,m),4.57(1H,d,J=4.0Hz),5.96(2H,s),6.881(1H,d,J=8.0Hz),6.94(1H,d,J=8.0Hz),7.13(1H,s),7.24(1H,d,J=8.0Hz),7.46(1H,d,J=8.0Hz),8.32(1H,s),9.76(1H,s),12.28(1H,s).
Example 14
Preparation of (4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) alkynyl) -2-oxacyclo [2.2.2] octan-1-methyl) methanol
Figure SMS_29
/>
Step 1: preparation of 1- ((tert-butyldiphenylsilyl) oxy) methyl) -N-methoxy-N-methyl-2-oxacyclo [2.2.2] octane-4-carboxamide
The compound 1- ((tert-butyldiphenylsilyl) oxy) methyl) -2-oxobicyclo [2.2.2] octane-4-carboxylic acid (1.00g, 2.36mmol) was added to DCM (30 mL), and dimethylhydroxylamine hydrochloride (276 mg, 2.83mmol), HBTU (1.34g, 3.54mmol), and DIEA (760mg, 5.90mmol) were sequentially added thereto and stirred at room temperature overnight. After completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 50/1) to obtain the objective compound (708 mg, yield 64.2%) as a colorless oil.
EM (calculated): 467.2; MS (ESI) M/z (M + H) + :468.2
Step 2: preparation of 1- ((tert-butyldiphenylsilyl) oxy) methyl) -2-oxacyclo [2.2.2] octane-4-carbaldehyde
Compound 1- ((tert-butyldiphenylsilyl) oxy) methyl) -N-methoxy-N-methyl-2-oxacyclo [2.2.2] octane-4-carboxamide (700mg, 1.50mmol) was added to anhydrous THF (10 mL), and DIBAL-H (2.3mL, 2.3mmol,1.0M in N-hexane) was slowly added dropwise thereto after cooling in an ice water bath, followed by stirring at room temperature for 3 hours. After the reaction was completed, water was added to the reaction solution and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, the reaction solution was concentrated to dryness to obtain the objective compound (435 mg, yield 71.0%) as a colorless oil.
EM (calculated): 408.2; MS (ESI) M/z (M + H) + :409.2
And step 3: preparation of tert-butyl ((4-ethynyl-2-oxacyclo [2.2.2] oct-1-yl) methoxy) diphenylsilane
The compound 1- ((tert-butyldiphenylsilyl) oxy) methyl) -2-oxacyclo [2.2.2] octane-4-carbaldehyde (435mg, 1.07mmol) was added to MeOH (10 mL), to which potassium carbonate (443mg, 3.21mmol) and dimethyl (1-diazo-2-oxopropyl) phosphonate (411mg, 2.14mmol) were added in this order, and stirred at room temperature overnight. After the completion of the reaction, the reaction liquid was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 50/1) to obtain the objective compound (386 mg, yield 89.4%) as a colorless oil.
EM (calculated): 404.2; MS (ESI) M/z (M + H) + :405.2
And 4, step 4: preparation of (4-ethynyl-2-oxacyclo [2.2.2] octan-1-yl) methanol
The compound tert-butyl ((4-ethynyl-2-oxocyclo [ 2.2.2)]Octane-1-yl) methoxy) diphenylsilane (386mg, 0.95mmol) was dissolved in THF (10 mL), to which TBAF.3H was added 2 O (899mg, 2.85mmol), and stirred at room temperature overnight. After completion of the reaction, water (30 mL) was added to the reaction mixture, and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Is free ofAfter drying over sodium sulfate hydrate, concentration to dryness afforded the title compound (359 mg, crude) as a colorless oil.
EM (calculated): 166.1; MS (ESI) M/z (M + H) + :167.1;
And 5: preparation of (4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) alkynyl) -2-oxacyclo [2.2.2] octane-1-methyl) methanol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (100mg, 0.16mmol) and (4-ethynyl-2-oxa [ 2.2.2)]Octane-1-yl) methanol (359 mg, crude) was added to DMF (10 mL), to which CuI (4 mg, 0.02mmol), et was added 3 N (48mg, 0.48mmol) and Pd (PPh) 3 ) 2 Cl 2 (22mg, 0.03mmol) was stirred at 45 ℃ for 3 hours under nitrogen. After completion of the reaction, water (100 mL) was added to the reaction mixture, and the mixture was extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (62 mg, yield 59.6%) as a pale yellow solid.
EM (calculated): 651.3 of the container; MS (ESI) M/z (M + H) + :652.4;
1 H NMR(400MHz,DMSO-d 6 )δ0.76-0.80(2H,m),0.93-0.97(2H,m),1.39-1.47(2H,m),1.52-1.58(2H,m),1.59-1.65(1H,m),1.74-1.81(1H,m),1.83-1.87(4H,m),1.94-2.01(4H,m),3.40-3.47(4H,m),4.28-4.39(1H,m),4.57(1H,d,J=4.0Hz),5.96(2H,s),6.881(1H,d,J=8.0Hz),6.94(1H,d,J=8.0Hz),7.13(1H,s),7.24(1H,d,J=8.0Hz),7.46(1H,d,J=8.0Hz),8.32(1H,s),9.76(1H,s),12.28(1H,s).
Example 15
Preparation of 4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) tetrahydro-2H-pyran-3-ol
Figure SMS_30
Step 1: preparation of 3, 7-Dioxolane [4.1.0] Heptane
Compound 3,6 dihydro-2H-pyran (1.00g, 11.89mmol) was added to DCM (20 mL), cooled to 0 ℃ with an ice-water bath, to which m-CPBA (4.10g, 23.78mmol) was added in portions, and stirred at room temperature overnight. After the completion of the reaction, the reaction solution was washed with water, the organic phase was collected and washed with a saturated aqueous NaCl solution 1 time, dried with anhydrous sodium sulfate and concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 20/1) to obtain the objective compound (600 mg, yield 50.4%) as a colorless oil.
EM (calculated): 100.1; MS (ESI) M/z (M + H) + :101.1
Step 2: preparation of 4- ((trimethylsilyl) ethynyl) tetrahydro-2H-pyran-3-ol
The compound trimethylsilylacetylene (1.18g, 11.98mmol) was added to anhydrous THF (10 mL), cooled to-78 deg.C, and n-BuLi (7.20mL, 14.38mmol,2M in n-hexane) was slowly added thereto. After stirring for 1 hour, 3, 7-dioxan [4.1.0] was slowly added dropwise thereto]Heptane (600mg, 5.99mmol) was maintained and stirring continued for 0.5 h at this temperature. Boron trifluoride ether solution was added thereto, and stirring was continued for 0.5 hour. After the reaction is finished, slowly adding saturated NH into the reaction liquid 4 The Cl aqueous solution was quenched and stirred by adding water (20 mL) and EA (20 mL). The aqueous phase was extracted 1 time with EA, the organic phases were combined and washed 1 time with saturated aqueous NaCl solution, dried over anhydrous sodium sulfate, and concentrated to dryness to give the target compound (465 mg, yield 39.2%) as a yellow oil.
EM (calculated): 198.1; MS (ESI) M/z (M + H) + :199.1
And step 3: preparation of 4-ethyltetrahydro-2H-pyran-3-ol
The compound 4- ((trimethylsilyl) ethynyl) tetrahydro-2H-pyran-3-ol (465mg, 2.35mmol) was added to MeOH (15 mL), to which was added potassium carbonate (649mg, 4.70mmol), and stirred at room temperature for 0.5H. After completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 15/1) to obtain the objective compound (236 mg, yield 79.7%) as a yellow oil.
EM (calculated): 126.1; MS (ESI) M/z (M + H) + :127.2
And 4, step 4: preparation of 4- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) tetrahydro-2H-pyran-3-ol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (100mg, 0.16mmol) and 4-ethyltetrahydro-2H-pyran-3-ol (40mg, 0.32mmol) were added to DMF (3 mL), to which was added CuI (4mg, 0.02mmol), et 3 N (48mg, 0.48mmol) and Pd (PPh) 3 ) 2 Cl 2 (22mg, 0.03mmol) was stirred at 45 ℃ for 4 hours under nitrogen. After completion of the reaction, water (100 mL) was added to the reaction mixture, and the mixture was extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 30/1) to obtain the target compound (16 mg, yield 16.4%) as a yellow solid.
EM (calculated): 611.3; MS (ESI) M/z (M + H) + :612.4
1 H NMR(400MHz,DMSO-d 6 )δ0.77-0.79(2H,m),0.95-1.00(2H,m),1.22-1.25(2H,m),1.60-1.63(2H,m),1.86-1.89(2H,m),2.01-2.04(1H,m),2.65-2.70(1H,m),3.07-3.12(1H,m),3.49-3.55(4H,m),3.77-3.84(2H,m),4.18-4.25(2H,m),5.26(1H,d,J=8.0Hz),5.98(2H,s),6.84(1H,d,J=8.0Hz),6.97(1H,d,J=8.0Hz),7.15(1H,s),7.27(1H,d,J=8.0Hz),7.53(1H,d,J=12.0Hz),8.37(1H,s),9.82(1H,s),12.30(1H,s).
Example 16
Preparation of (5- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) tetrahydro-2H-pyran-2-yl) methanol
Figure SMS_31
Step 1: preparation of 6- (((tert-butyldiphenylsilyl) oxy) methyl) -N-methoxy-N-methyltetrahydro-2H-pyran-3-carboxamide
The compound 6- ((tert-butyldiphenylsilyl) oxy) methyl) tetrahydro-2H-pyran-3-carboxylic acid (556 mg, 1.40mmol) was added to DCM (20 mL), and dimethylhydroxylamine hydrochloride (205mg, 2.10mmol), HBTU (637mg, 1.68mmol) and DIEA (452mg, 3.50mmol) were successively added thereto, and stirred at room temperature for 5 hours. After completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 10/1) to obtain the objective compound (524 mg, yield 84.8%) as a colorless oil.
EM (calculated): 441.2; MS (ESI) M/z (M + H) + :442.2
And 2, step: preparation of 6- (((tert-butyldiphenylsilyl) oxy) methyl) tetrahydro-2H-pyran-3-carbaldehyde
The compound 6- (((tert-butyldiphenylsilyl) oxy) methyl) -N-methoxy-N-methyltetrahydro-2H-pyran-3-carboxamide (524mg, 1.19mmol) was added to anhydrous THF (10 mL), and after cooling in an ice-water bath, DIBAL-H (2.4mL, 2.38mmol,1.0M in N-hexane) was slowly added dropwise thereto, and stirring was continued for 0.5 hour while maintaining the temperature. After the reaction was completed, water was added to the reaction solution and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, the reaction solution was concentrated to dryness to give the objective compound (1.20 g, crude product) as a colorless oil.
EM (calculated): 382.2; MS (ESI) M/z (M + H) + :383.3
And step 3: preparation of tert-butyl ((5-ethyltetrahydro-2H-pyran-2-ynyl) methoxy) diphenylsilane
Compound 6- (((tert-butyldiphenylsilyl) oxy) methyl) tetrahydro-2H-pyran-3-carbaldehyde (1.20 g, crude) was added to MeOH (10 mL), to which were added potassium carbonate (493mg, 3.57mmol) and dimethyl (1-diazo-2-oxopropyl) phosphonate (419mg, 2.38mmol) in this order, and stirred at room temperature overnight. After the completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 20/1) to obtain the objective compound (285 mg, total yield of two steps: 63.3%) as a colorless oil.
EM (calculated): 378.2; MS (ESI) M/z (M + H) + :379.3
And 4, step 4: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -6- (6- ((tert-butyldiphenylsilyl) oxy) methyl) tetrahydro-2H-pyran-3-yl) ethynyl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-methyl) quinazolin-4-amine
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (227mg, 0.37mmol) and tert-butyl ((5-ethyltetrahydro-2H-pyran-2-ynyl) methoxy) diphenylsilane (285mg, 0.75mmol) were added to DMF (10 mL), to which CuI (8mg, 0.04mmol), et 3 N (112mg, 1.11mmol) and Pd (PPh) 3 ) 2 Cl 2 (28mg, 0.04mmol), and stirred at 45 ℃ for 4 hours under nitrogen. After completion of the reaction, water (100 mL) was added to the reaction mixture, and extracted 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 20/1) to obtain the target compound (200 mg, yield 62.7%) as a brown-yellow oil.
EM (calculated): 863.4; MS (ESI) M/z (M + H) + :864.5
And 5: preparation of (5- ((2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethynyl) tetrahydro-2H-pyran-2-yl) methanol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxol-5-yl) piperidin-1-yl) -6- (6- ((tert-butyldiphenylsilyl) oxy) methyl) tetrahydro-2H-pyran-3-yl) ethynyl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-methyl) quinazolin-4-amine (200mg, 0.23mmol) was dissolved in THF (10 mL) to which TBAF.3H was added 2 O (218mg, 0.69mmol), was stirred at 40 ℃ for 2 hours. After completion of the reaction, water (30 mL) was added to the reaction mixture, and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained is purified by column chromatography (DCM/MeOH = 20/1),the target compound (97 mg, yield 67.5%) was obtained as a dark yellow solid.
EM (calculated): 625.3 of the total weight of the mixture; MS (ESI) M/z (M + H) + :626.4
1 H NMR(400MHz,DMSO-d 6 )δ0.77-0.80(2H,m),0.94-0.96(2H,m),1.28-1.34(1H,m),1.57-1.67(4H,m),1.84-1.89(3H,m),2.11-2.20(1H,m),3.48-3.63(6H,m),4.02-4.07(1H,m),4.13-4.37(2H,m),4.67(1H,t,J=4.0Hz),5.97(2H,s),6.82(1H,d,J=8.0Hz),6.95(1H,d,J=12.0Hz),7.14(1H,s),7.25(1H,d,J=8.0Hz),7.48(1H,d,J=8.0Hz),8.34(1H,s),9.80(1H,s),12.30(1H,s).
Example 17
Preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6- (1H-pyrazol-4-yl) quinazolin-4-amine
Figure SMS_32
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (30mg, 0.05mmol) and 4-pyrazoleboronic acid pinacol ester (19mg, 0.10mmol) were added to 1,4-dioxane/H 2 O (2mL, 5/1), to which were added sodium carbonate (21mg, 0.20mmol) and Pd (dppf) Cl 2 DCM (7mg, 0.01mmol) was stirred under nitrogen at 80 ℃ for 4 h. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is purified by column chromatography (DCM/MeOH/NH) 3 .H 2 O = 20/1/0.05) to obtain the target compound (5 mg, yield 18.1%) as a white solid.
EM (calculated): 553.2; MS (ESI) M/z (M + H) + :554.3
1 H NMR(400MHz,DMSO-d 6 )δ0.77-0.80(2H,m),0.95-0.96(2H,m),1.54-1.57(2H,m),1.82-1.91(4H,m),1.96-2.01(1H,m),3.41-3.50(2H,m),4.30-4.34(2H,m),5.96(2H,s),6.80(1H,d,J=7.6Hz),6.94(1H,d,J=9.6Hz),7.13(1H,s),7.32(1H,d,J=8.4Hz),7.85(1H,d,J=8.8Hz),8.01-8.15(2H,m),8.48(1H,s),9.72(1H,s),12.32(1H,s),12.94(1H,brs).
Example 18
Preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6- (1-methyl-1H-pyrazol-4-yl) quinazolin-4-amine
Figure SMS_33
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (30mg, 0.05mmol) and 1-methyl-4-pyrazoleboronic acid pinacol ester (21mg, 0.10mmol) were added to 1,4-dioxane/H 2 To O (2mL, 5/1), sodium carbonate (21mg, 0.20mmol) and Pd (dppf) Cl were added 2 DCM (7mg, 0.01mmol) was stirred at 80 ℃ for 4h under nitrogen. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is purified by column chromatography (DCM/MeOH/NH) 3 .H 2 O = 20/1/0.05) to obtain the target compound (4 mg, yield 14.1%) as a white solid.
EM (calculated): 567.3; MS (ESI) M/z (M + H) + :568.3
1 H NMR(400MHz,DMSO-d 6 )δ0.74-0.77(2H,m),0.93-0.95(2H,m),1.44(3H,s),1.50-1.55(2H,m),1.78-1.86(2H,m),1.87-1.89(2H,brs),1.91-1.96(1H,m),3.45-3.52(2H,m),4.33-4.37(2H,m),5.95(2H,s),6.77(1H,d,J=6.4Hz),6.91(1H,d,J=8.0Hz),7.12(1H,s),7.30(1H,d,J=7.2Hz),7.80(1H,d,J=8.0Hz),8.00-8.11(2H,m),8.44(1H,s),9.72(1H,s),12.32(1H,s).
Example 19
Preparation of 3- (4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) phenoxy) propane-1, 2-diol
Figure SMS_34
Step 1: preparation of 2- (4- ((2, 2-dimethyl-1, 3-dioxolan-4-yl) methoxy) phenylboronic acid pinacol ester
The compound, 4-hydroxyphenylboronic acid pinacol ester (2.20g, 10.00mmol) was added to DMSO (40 mL), and potassium carbonate (2.07g, 15.00mmol), 4- (chloromethyl) -2, 2-dimethyl-1, 3-dioxolane (1.80g, 12.00mmol) were sequentially added thereto, and the mixture was stirred at 120 ℃ for 10 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, water (400 mL) was added thereto, and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution, dried over anhydrous sodium sulfate, concentrated to dryness, and the residue was purified by column chromatography (PE/EA = 10/1) to obtain the objective compound (1.7 g, yield 50.9%) as a brown oil.
EM (calculated): 334.2; MS (ESI) M/z (M + H) + :335.2
Step 2: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6- (4- ((2, 2-dimethyl-1, 3-dioxido-4-yl) methoxy) phenyl) quinazolin-4-amine
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (100mg, 0.116mmol) and 2- (4- ((2, 2-dimethyl-1, 3-dioxolan-4-yl) methoxy) phenylboronic acid pinacol ester (107mg, 0.32mmol) were added to 1,4-dioxane/H 2 O (10mL, 5/1), to which were added sodium carbonate (34mg, 0.32mmol) and Pd (dppf) Cl 2 DCM (14mg, 0.02mmol) was stirred at 80 ℃ for 5 h under nitrogen. After completion of the reaction, the reaction solution was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (50 mg, yield 45.1%) as a brown solid.
EM (calculated): 693.3; MS (ESI) M/z (M + H) + :694.4
And step 3: preparation of 3- (4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) phenoxy) propane-1, 2-diol
The compound 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6- (4- ((2, 2-dimethyl-1, 3-dioxan-4-yl) methoxy) phenyl) quinazolin-4-amine (50mg, 0.07mmol) was added to THF (5 mL), concentrated hydrochloric acid (0.5 mL) was added thereto, and stirred at room temperature for 1 hour. After completion of the reaction, the reaction solution was concentrated to dryness, and the obtained crude product was purified by column chromatography (DCM/MeOH = 15/1) to obtain the objective compound (20 mg, yield 42.5%) as a pale yellow solid.
EM (calculated): 653.3; MS (ESI) M/z (M + H) + :654.4
1 H NMR(400MHz,DMSO-d 6 )δ0.76-0.79(2H,m),0.83-0.85(2H,m),1.53-1.56(2H,m),1.79-1.84(2H,m),1.87-1.89(1H,m),1.92-2.01(2H,brs),3.45-3.48(2H,m),3.81-3.82(1H,m),3.89-3.93(1H,m),4.03-4.07(1H,m),4.31-4.39(2H,m),4.70-4.73(1H,m),4.99-5.00(1H,m),5.95(2H,s),6.80(1H,d,J=8.0Hz),6.92(1H,d,J=7.6Hz),7.04(2H,d,J=8.8Hz),7.13(1H,s),7.37(1H,d,J=8.8Hz),7.75(2H,d,J=8.8Hz),7.90(1H,d,J=8.8Hz),8.54(1H,s),9.91(1H,s),12.30(1H,s).
Example 20
Preparation of 1- (4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) phenyl) ethane-1, 2-diol
Figure SMS_35
Step 1: preparation of 1- (4-bromophenyl) ethane-1, 2-diol
The compound 1-bromo-4-vinylbenzene (200mg, 1.10mmol) was added to DCM/H 2 O (10mL, 4/1), NMO (258mg, 2.20mmol) and KOsO were added thereto 4 .2H 2 O (810 mg, 2.20mmol), and the mixture was stirred at room temperature for 3 hours. After the completion of the reaction, the reaction solution was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA = 5/1) to obtain the objective compound (120 mg, yield 50.5%) as a white solid.
EM (calculated): 216.0; MS (ESI) M/z (M + H) + :217.0
Step 2: preparation of 1- (4- (2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) phenyl) ethane-1, 2-diol
The compound 1- (4-bromophenyl) ethane-1, 2-diol (100mg, 0.46mmol) was dissolved in 1,4-dioxane (5 mL), to which was added potassium acetate (90mg, 0.92mmol) and Pd (dppf) Cl in that order 2 DCM (28mg, 0.04mmol) was stirred at 90 ℃ for 5 h under nitrogen. After the reaction was completed, the reaction solution was cooled to room temperature, and 2- (4-amino-4- (benzo [ d ] s) was sequentially added thereto][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (140mg, 0.23mmol), sodium carbonate (49mg, 0.46mmol) and Pd (dppf) Cl 2 DCM (14mg, 0.02mmol) was stirred under nitrogen at 90 ℃ for 6 h. After the reaction was completed, the reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated to dryness, and the obtained crude product was purified by column chromatography and preparative HPLC in this order to obtain the target compound (5 mg, yield 3.5%) as a yellow solid.
EM (calculated): 623.3; MS (ESI) M/z (M + H) + :624.4
1 H NMR(400MHz,DMSO-d 6 )δ0.78-0.81(2H,m),0.93-0.98(2H,m),1.51-1.54(2H,m),1.76-1.80(2H,m),1.81-1.84(1H,m),1.89-1.95(2H,brs),3.41-3.47(2H,m),4.13-4.18(2H,m),4.25-4.32(2H,m),5.00-5.06(1H,m),5.18-5.27(1H,m),5.55-5.60(1H,m),5.96(2H,s),6.81-6.86(2H,m),6.92-6.98(2H,m),7.13(1H,s),7.33(2H,d,J=6.4Hz),7.52-7.58(2H,m),8.41(1H,s),9.87(1H,s),12.33(1H,s).
Example 21
Preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-vinylquinazolin-4-amine
Figure SMS_36
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (60mg, 0.10 mmol) and potassium ethylene trifluoroborate (27mg, 0.20 mmol) were added to 1,4-dioxane/H 2 O (10 mL, 5/1), et was added thereto 3 N (30mg, 0.30mmol) and Pd (dppf) Cl 2 .DCM (14mg, 0.02mmol), and stirred at 80 ℃ for 4 hours under nitrogen. After completion of the reaction, the reaction solution was concentrated to dryness, and the obtained crude product was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (46 mg, yield 89.6%) as a yellow solid.
EM (calculated): 513.2; MS (ESI) M/z (M + H) + :514.3
1 H NMR(400MHz,DMSO-d 6 )δ0.78-0.81(2H,m),0.94-0.97(2H,m),1.51-1.54(2H,m),1.72-1.77(2H,m),1.85-1.88(1H,m),1.90-1.95(2H,brs),3.37-3.43(2H,m),4.25-4.35(2H,m),5.25-5.351(2H,m),5.95(2H,s),6.02-6.05(1H,m),6.83(1H,d,J=6.4Hz),6.90(1H,d,J=8.0Hz),7.13(1H,s),7.29(1H,d,J=8.0Hz),7.51(1H,d,J=8.4Hz),8.37(1H,s),9.81(1H,s),12.30(1H,s).
Example 22
Preparation of 1- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) ethane-1, 2-diol
Figure SMS_37
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-vinylquinazolin-4-amine (40mg, 0.08mmol) was added to DCM/THF/H 2 To O (10mL, 4/1/1), NMO (19mg, 0.16mmol) and KOsO were added 4 .2H 2 O (60mg, 0.16mmol), and stirred at room temperature for 2 hours. After completion of the reaction, the reaction solution was concentrated to dryness, and the obtained crude product was purified by preparative HPLC to give the target compound (10 mg, yield 22.9%) as a yellow solid.
EM (calculated): 547.2; MS (ESI) M/z (M + H) + :548.3
1 H NMR(400MHz,DMSO-d 6 )δ0.75-0.79(2H,m),0.91-0.94(2H,m),1.49-1.52(2H,m),1.75-1.79(2H,m),1.89-1.96(3H,m),3.39-3.44(2H,m),4.11-4.17(2H,m),4.27-4.34(2H,m),5.02-5.09(1H,m),5.20-5.29(1H,m),5.59-5.63(1H,m),5.97(2H,s),6.87(1H,d,J=5.2Hz),6.94(1H,d,J=7.6Hz),7.12(1H,s),7.33(1H,d,J=7.6Hz),7.55(1H,d,J=8.0Hz),8.39(1H,s),9.84(1H,s),12.31(1H,s).
Example 23
Preparation of (5- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) -3, 4-dihydro-2H-pyran-2-yl) methanol
Figure SMS_38
Step 1: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -6- (2- ((tert-butyldiphenylsilyl) oxy) methyl) -3, 4-dihydro-2H-pyran-5-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) quinazolin-4-amine
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (120mg, 0.20 mmol) and tert-butyldiphenyl ((5- (4, 5-tetramethyl-1, 3, 2-dioxobenzofuran-2-yl) -3, 4-dihydro-2H-pyran-2-yl) methoxy) silane (120mg, 0.25mmol) were added to 1,4-dioxane/H 2 To O (10mL, 5/1), sodium carbonate (42mg, 0.40mmol) and Pd (dppf) Cl were added 2 DCM (21mg, 0.03mmol) was stirred overnight at 80 ℃ under nitrogen. After completion of the reaction, the reaction solution was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (80 mg, yield 47.8%) as a yellow solid.
EM (calculated): 837.4; MS (ESI) M/z (M + H) + :838.5
Step 2: preparation of (5- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) -3, 4-dihydro-2H-pyran-2-yl) methanol
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxol-5-yl) piperidin-1-yl) -6- (2- ((tert-butyldiphenylsilyl) oxy) methyl) -3, 4-dihydro-2H-pyran-5-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) quinazolin-4-amine (80mg, 0.10mmol) was dissolved in THF (10 mL), to which TBAF.3H was added 2 O(95mg,0.30 mmol), stirring at 40 ℃ for 3 hours. After completion of the reaction, water (30 mL) was added to the reaction mixture, and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 20/1) to obtain the target compound (50 mg, yield 83.3%) as a white solid.
EM (calculated): 599.3; MS (ESI) M/z (M + H) + :600.4
1 H NMR(400MHz,DMSO-d 6 )δ0.81-0.85(2H,m),0.96-1.00(2H,m),1.43-1.47(2H,m),1.74-1.78(2H,m),1.88-1.92(1H,m),1.99-2.04(2H,m),2.13-2.17(2H,m),3.45-3.52(2H,m),3.79-3.86(2H,m),4.46-4.50(1H,m),4.68-4.77(2H,m),5.33(1H,t,J=4.0Hz),6.02(2H,s),6.38(1H,s),6.92(1H,d,J=8.0Hz),7.04(1H,d,J=8.0Hz),7.21(1H,s),7.32(1H,d,J=8.0Hz),7.72(1H,d,J=12.0Hz),8.12(1H,s),9.87(1H,s),12.34(1H,s).
Example 24
Preparation of (5- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) tetrahydro-2H-pyran-2-yl) methanol
Figure SMS_39
Compound (5- (2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazolin-6-yl) -3, 4-dihydro-2H-pyran-2-yl) methanol (40mg, 0.07mmol) was dissolved in methanol (10 mL), pd/C (10mg, 10% content) was added thereto, and after hydrogen substitution, stirred at room temperature overnight. After completion of the reaction, the reaction solution was filtered, the filtrate was concentrated to dryness, and the obtained crude product was purified by column chromatography (DCM/MeOH = 20/1) to obtain the objective compound (20 mg, yield 47.6%) as a white solid.
EM (calculated): 601.3; MS (ESI) M/z (M + H) + :602.4
1 H NMR(400MHz,DMSO-d 6 )δ0.79-0.82(2H,m),0.93-0.95(2H,m),1.37-1.42(2H,m),1.70-1.784(2H,m),1.85-1.90(1H,m),1.79-2.03(2H,m),2.11-2.16(2H,m),2.35-2.39(1H,m),3.47-3.53(2H,m),3.81-3.87(2H,m),4.49-4.53(1H,m),4.56-4.60(2H,m),4.69-4.76(2H,m),5.34(1H,t,J=4.0Hz),5.96(2H,s),6.88(1H,d,J=7.6Hz),7.05(1H,d,J=8.4Hz),7.15(1H,s),7.28(1H,d,J=6.4Hz),7.68(1H,d,J=8.0Hz),8.21(1H,s),9.69(1H,s),12.28(1H,s).
Example 25
Preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) -N- (1- (hydroxymethyl) -2-oxacyclo [2.2.2] octan-4-yl) quinazoline-6-carboxamide
Figure SMS_40
Step 1: preparation of 1- (((tert-butyldiphenylsilyl) oxy) methyl) -2-oxacyclo [2.2.2] octan-4-amine
The compound 1- ((tert-butyldiphenylsilyl) oxy) methyl) -2-oxabicyclo [2.2.2]Octane-4-carboxylic acid (1.00g, 2.36mmol) was added to toluene (15 mL), to which Et was added 3 N (715mg, 7.08mmol), molecular sieves and DPPA (1.30g, 4.72mmol), was stirred at 100 ℃ for 4 hours. After completion of the reaction, the reaction solution was concentrated to dryness, the residue was dissolved in THF (20 mL), and potassium tert-butoxide (530mg, 4.72mmol) was added thereto and stirred at room temperature overnight. After completion of the reaction, the reaction solution was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA = 5/1) to obtain the objective compound (437 mg, yield 46.9%) as a yellow oil.
EM (calculated): 395.2; MS (ESI) M/z (M + H) + :396.2
Step 2: preparation of methyl 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazoline-6-carboxylate
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -N- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) -6-iodoquinazolin-4-amine (75mg, 0.12mmol) was added to methanol (10 mL), to which Et was added 3 N (36mg, 0.36mmol) and Pd (dppf) Cl 2 DCM (14mg, 0.02mmol) was stirred overnight at 70 ℃ under a carbon monoxide atmosphere. After completion of the reaction, the reaction solution was concentrated to dryness, and the obtained crude product was purified by column chromatography (DCM/MeOH = 30/1) to obtain the objective compound (63 mg, yield 96.3%) as a yellow solid.
EM (calculated): 545.2; MS (ESI) M/z (M + H) + :546.3
And step 3: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazoline-6-carboxylic acid
The compound methyl 2- (4-amino-4- (benzo [ d ]][1,3]Dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazoline-6-carboxylate (63mg, 0.12mmol) was added to methanol/H 2 O (10 mL, 4/1), to which LiOH. H was added 2 O (15mg, 0.36mmol), and the mixture was stirred at 50 ℃ for 2 hours. After the reaction, the reaction solution was concentrated to dryness to obtain the target compound (crude product) as a yellow solid.
EM (calculated): 531.2; MS (ESI) M/z (M-H) - :530.2
And 4, step 4: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -N- (1- ((tert-butyldiphenylsilyl) oxy) methyl) -2-oxacyclo [2.2.2] octan-4-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazoline-6-carboxamide
The compound 2- (4-amino-4- (benzo [ d ] [1,3] dioxo-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazoline-6-carboxylic acid (crude product) was added to THF (10 mL), to which 1- (((tert-butyldiphenylsilyl) oxy) methyl) -2-oxacyclo [2.2.2] octan-4-amine (71mg, 0.18mmol), HATU (68mg, 0.18mmol) and DIEA (31mg, 0.24mmol) were successively added, and stirred at room temperature for 4 hours. After completion of the reaction, the reaction solution was concentrated to dryness, and the residue was purified by column chromatography (DCM/MeOH = 20/1) to obtain the objective compound (61 mg, total yield of two steps 56.0%) as a yellow solid.
EM (calculated): 908.4; MS (ESI) M/z (M + H) + :909.5
And 5: preparation of 2- (4-amino-4- (benzo [ d ] [1,3] dioxol-5-yl) piperidin-1-yl) -4- (5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) -N- (1- (hydroxymethyl) -2-oxacyclo [2.2.2] octan-4-yl) quinazoline-6-carboxamide
Reacting the compound 2- (4-amino-4- (benzo [ d ]][1,3]Dioxol-5-yl) piperidin-1-yl) -N- (1- ((tert-butyldiphenylsilyl) oxy) methyl) -2-oxacyclo [2.2.2]Octane-4-yl) -4- ((5-cyclopropyl-4-fluoro-1H-pyrazol-3-yl) amino) quinazoline-6-carboxamide (61mg, 0.07mmol) was dissolved in THF (10 mL), to which TBAF.3H was added 2 O (66mg, 0.21mmol), and stirred at 50 ℃ for 2 hours. After completion of the reaction, water (30 mL) was added to the reaction mixture, and extracted with EA 2 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. After drying over anhydrous sodium sulfate, concentration to dryness, the crude product obtained was purified by column chromatography (DCM/MeOH = 20/1) to obtain the target compound (15 mg, yield 34.1%) as a yellow solid.
EM (calculated): 670.3; MS (ESI) M/z (M + H) + :671.4
1 H NMR(400MHz,DMSO-d 6 )δ0.79-0.81(2H,m),0.95-0.97(2H,m),1.62-1.83(6H,m),1.87-1.91(1H,m),1.94-2.04(4H,m),2.12-2.31(2H,m),3.18(2H,d,J=4.0Hz),3.44-3.48(4H,m),3.59-3.63(2H,m),4.01(2H,s),4.57(1H,t,J=4.0Hz),6.00(2H,s),6.88(1H,d,J=8.0Hz),7.01(1H,d,J=8.0Hz),7.17(1H,s),7.31(1H,d,J=8.0Hz),7.53-7.57(1H,m),7.92(1H,d,J=12.0Hz),8.65(1H,s),9.84(1H,s),12.33(1H,s).
Test example 1 inhibitory Effect of Compounds on kinase Activity
1: test materials
PAK4(Carna,No.07-126),PAK2(Carna,No.07-124),Kinase substrate31(Cisbio,No.61ST2BLE),DMSO(Sigma,No.D0632),384-well plate(Greiner,No.784075),PF-3758309(selleckchem,No.S709403)
2: experimental method
2.1 preparation of Compounds
Compounds were received by the administrator and the powders were dissolved in 100% DMSO and formulated into 10mM stock stored in nitrogen cabinet protected from light.
2.2 kinase reaction Processes
(1) A1 XKinase buffer was prepared.
(2) Preparation of compound concentration gradient: test compounds were tested at 1000nM, diluted to 100-fold final concentration in 100% DMSO solutions in 384source plates, and compounds were diluted 3-fold with Precision, 10 concentrations. 250nL of 100-fold final concentration of compound was transferred to the 384-well plates of interest using a knockout Echo 550.
(3) A2.5 fold final concentration of Kinase solution was prepared using a1 XKinase buffer.
(4) Add 10. Mu.L of 2.5 fold final concentration kinase solution to the compound well and positive control well, respectively; mu.L of 1 XKinase buffer was added to the negative control wells.
(5) Centrifuge at 1000rpm for 30 seconds, shake the plate and incubate at room temperature for 10 minutes.
(6) A5/3 fold final ATP and Kinase substrate22 mixture was prepared using a1 XKinase buffer.
(7) The reaction was initiated by adding 15. Mu.L of a 5/3 fold final ATP and substrate mixture.
(8) The 384 well plate is centrifuged at 1000rpm for 30 seconds, shaken and mixed, and incubated at room temperature for 60min.
(9) Add 30. Mu.L of termination detection solution to stop the kinase reaction, centrifuge at 1000rpm for 30 seconds, shake and mix.
(10) The conversion was read using a Caliper EZ Reader.
2.3 data analysis
Figure SMS_41
Wherein: conversion% _ sample is the Conversion reading for the sample; conversion% _ min: negative control well mean, representing conversion readings without enzyme live wells; conversion% _ max: positive control wells mean, conversion readings for wells without compound inhibition.
Fitting a dose-response curve:
the log value of the concentration is taken as an X axis, the percent inhibition rate is taken as a Y axis, and the log (inhibitor) v.response-Variable slope of GraphPad Prism 5 of analysis software is adopted to fit a dose-effect curve, so that the IC of each compound to the enzyme activity is obtained 50 The value is obtained. The calculation formula is:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50 -X)*HillSlope))
the test results are shown in table 2:
TABLE 2 inhibitory Activity of the Compounds on PAK4 and PAK2 kinases (IC) 50 )
Figure SMS_42
Test example 2 detection of Compound cardiotoxicity hERG test
1. Principle of experiment
1.1hERG ion channel stably expressed in HEK293 cells. After the hERG current is stable, the influence of the compound on the hERG ion channel can be obtained by comparing the sizes of the hERG current before and after the application of different compound concentrations.
1.2 regulations to follow
This experiment was not performed in compliance with GLP regulations but was performed under standard operating regulations (SOP) of orlistat and related literature published on SCI.
2. Test animals (cell/reagent)
HEK293 cells with steady expression of hERG ion channels.
3. Testing instrument
Patch clamp instrument: PC-505B
A micro-manipulation instrument: MP-225
Drawing an electrode instrument: PC-10 (Narishige, japan)
4. Pharmaceutical formulation
The test compounds were stored at a concentration of 3mM in dimethyl sulfoxide (DMSO). The test day is dissolved in extracellular fluid to prepare the required concentration.
Compounds were purchased from Sigma (st. Louis, MO) company, except for NaOH and KOH for acid-base titration. The final concentrations of test compounds were all prepared the same day and re-dissolved in extracellular fluid. The extracellular fluid (mM) is NaCl,137; KCl,4; caCl2,1.8; mgCl2,1; HEPES,10; glucose 10; pH7.4 (NaOH titration).
All test and control compound solutions contained 0.3% DMSO. Intracellular fluid (mM) was: k asparate, 130; mgCl2,5; EGTA 5; HEPES,10; tris-ATP 4; pH 7.2 (KOH titration).
5. Test method
Cell: all experiments were performed at room temperature. Each cell served as its own control.
Testing of the compounds: the compounds are perfused by adopting a perfusion system utilizing the self gravity. At least 1 cell was tested per concentration. After the current stabilized (or 5 minutes), the blocking effect of the compound was calculated by comparing the change in current level before and after the compound was used.
Positive control: is free of
Electrophysiological properties: the cells were transferred to a perfusion chamber and perfused with extracellular fluid. Intracellular fluid (mM) was: KAspartate,130; mgCl2,5; EGTA 5; HEPES,10; tris-ATP 4; pH 7.2 (KOH titration). The intracellular fluid was stored in small portions in a-80 ℃ freezer and thawed the day of the experiment. The electrode was drawn with PC-10 (Narishige, japan). Whole cell patch clamp recordings were made and noise was filtered at one fifth of the sampling frequency.
Test procedure and results: cells were clamped at-80 mV, then depolarized to 40mV with a 4 second duration square wave, and hyperpolarized to-40 mV with a 2 second duration square wave to give the hERG tail current. This procedure was repeated every 20 seconds. The hERG tail current is pure hERG current. The maximum current induced by the second square wave is detected, after it has stabilized, the test compound is perfused, and when the reaction has stabilized, the intensity of the block is calculated.
The test results are shown in table 3:
TABLE 3 inhibition of hERG channel Current (%) (at 1um concentration)
Figure SMS_43
Figure SMS_44
Using the same experimental conditions and methods, some of the preferred compounds of WO2022033420A1 were tested for hERG inhibition and the results are shown in Table 4 below:
TABLE 4 inhibition of partial compound hERG channel current (%) in WO2022033420A1 patent (at 1um concentration)
Example numbering Inhibition rate Example numbering Inhibition rate
71 85.70% 96 77.12%
79 75.32% 97 84.17%
Higher inhibition rates indicate a greater risk of drug hERG cardiotoxicity. As can be seen from Table 4, the preferred compounds in WO2022033420A1 all have higher hERG inhibition rates than most of the compounds prepared in examples 1-25 provided by the present invention, with greater risk of cardiotoxicity. Through comparison, the structural optimization shows that most of the compounds prepared by the invention have low hERG inhibition rate and smaller cardiotoxicity risk.
Test example 3 detection of Compound liver microsome stability test
1: materials and methods
Buffer solution:
(1) 100mM potassium phosphate buffer, pH 7.4; (2) 10mM MgCl 2
Preparation of compound solution:
(1) Preparation of 100. Mu.M working solution: mu.L of the stock solution (10 mM) of the test or control group was diluted with 495. Mu.L of methanol to give a compound concentration of 100. Mu.M (99% MeOH).
(2) Preparation of 10 μ M working solution: mu.L of 100. Mu.M working solution was diluted with 450. Mu.L of 100mM potassium phosphate buffer to give a compound concentration of 10. Mu.M (9.9% MeOH).
Composition of NADPH (prototype coenzyme II) regeneration System (final concentration of isocitrate dehydrogenase in culture broth 1.0 unit/mL):
β -nicotinamide adenine dinucleotide phosphate, supplier: chem-impex international shipment number: n00616
Preparation of liver microsome solutions (final concentration of 0.5mg protein/mL), liver microsome species are shown in Table 5:
TABLE 5
Figure SMS_45
Stopping liquid:
acetonitrile ice-cold solution containing 100ng/mL tolbutamide and 100ng/mL labetalol as internal standards.
The method comprises the following operation steps:
(1) In each well (T0, T5, T10, T20, T30, T60, and NCF 60) except the blank substrate site well, 10 μ L of working solution of test or control drug was added.
(2) An 80 μ L/well microsome solution was dispensed onto each plate using Apricot and the mixture of microsome solution and compound was incubated at 37 ℃ for about 10 minutes.
(3) To NCF60 was added 10. Mu.L of 100mM potassium phosphate buffer/well, incubated at 37 ℃ and timer 1 was started for the time shown in Table 6.
TABLE 6
Figure SMS_46
(4) After preheating, 10. Mu.L/well of NADPH regeneration system was dispensed to each plate with Apricot to start the reaction.
TABLE 7 Final concentrations of each component in the incubation Medium
Figure SMS_47
(5) Incubate at 37 ℃ and start timer 2, see table 8 for data.
TABLE 8
Figure SMS_48
(6) The reaction was stopped by adding pre-cooled stop solutions (containing 100ng/mL tolbutamide and 100ng/mL albuterol) at 4 ℃ to each well.
(7) The sample plate is then shaken on a shaking trigger for about 10 minutes.
(8) The samples were centrifuged at 4000rpm for 20min at 4 ℃.
(9) And adding 300 mu L of HPLC-grade water into each hole of another 96-hole plate, adding 100 mu L of supernate obtained by centrifugation into corresponding hole positions, and uniformly mixing the two for LC/MS/MS detection.
And (3) data analysis:
calculating t from first order elimination kinetics 1/2 And CL int(mic) Value of
The first order elimination kinetic equation is:
Figure SMS_49
Figure SMS_50
Figure SMS_51
Figure SMS_52
Figure SMS_53
the results of the partial compound liver microsome stability test are shown in table 9:
table 9 partial compound liver microsome stability data
Figure SMS_54
Test example 4 test compound rat PK Properties assay
SD rats, male (purchased from mondsucho laboratory animals ltd). The test compounds were each administered orally (50 mg/kg, 3 per group) to SD rats in a single dose for pharmacokinetic studies. Test compounds were formulated on the day of administration, dissolved using 5% DMSO +10% solutol +85% saline, and formulated into dosing solutions after vortexing for 2min, sonicating for 5 min. Animals were fasted for 10-14 hours prior to oral administration and returned to food 4 hours after administration. After the SD rat is orally taken through the gavage and intravenously administered, pharmacokinetic samples are collected through jugular veins, and the collection time points are as follows: before administration, 5min, 15min, 30min, 1h, 2h, 4h, 6h, 8h and 24h after administration, 3 whole blood samples were collected at each time point, in an amount of about 0.2mL, and anticoagulated with heparin sodium. Immediately after the blood sample was collected, the blood was placed on ice and the plasma was centrifuged within 1 hour (centrifugation conditions: 6800 rpm, 6 minutes, 2-8 ℃). The collected plasma was stored in a-80 ℃ freezer before analysis.
The results of the pharmacokinetic testing of some of the compounds of the invention are shown in table 10 below:
TABLE 10 pharmacokinetic test results for some of the compounds of the invention
Figure SMS_55
Figure SMS_56
Finally, it should be noted that: the above embodiments are only used to illustrate the technical solution of the present invention, and not to limit the same; while the invention has been described in detail and with reference to the foregoing embodiments, it will be understood by those skilled in the art that: the technical solutions described in the foregoing embodiments may still be modified, or some or all of the technical features may be equivalently replaced; and the modifications or the substitutions do not make the essence of the corresponding technical solutions depart from the scope of the technical solutions of the embodiments of the present invention.

Claims (18)

1. A compound of formula I, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure FDA0003898749600000011
wherein A is 1 、A 2 、A 3 、A 4 Independently selected from C or N, when A 1 、A 2 、A 3 、A 4 Any one of which is N, R connected with the N 2 、R 3 、R 4 、R 5 Is absent;
R 2 、R 3 、R 4 、R 5 independently selected from-H, halogen, -OH, -CN, -NH 2 、-NO 2 SH, C1-10 straight chain/branched chain alkyl, C3-10 naphthenic base, heterocyclic alkyl, alkynyl, alkenyl, aromatic group, heterocyclic aromatic group, amido, ester group, sulfonyl and phosphoryl, wherein H in the groups can be substituted by the following groups: halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, substituted or unsubstituted aryl or heteroaryl;
the substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyl amino, alkyl, cycloalkyl, heterocyclic alkyl, aryl, heterocyclic aryl, ester group, acyl, carbonyl, amido, sulfonyl, phosphoryl;
B 1 、B 2 independently selected from C or N; l is selected from-O-, -S-, -NH-or alkylene;
R 1 selected from substituted or unsubstituted five-membered or six-membered aromatic or heterocyclic aromatic groups, substituted or unsubstituted heterocycloalkyl groups containing at least one N and/or O atom;
R 6 selected from-H, -NH 2 OH, halogen, amido, sulfonyl, sulfonic group, C1-6 alkyl or alkoxy, C3-6 cycloalkyl, C3-6 heterocycloalkyl, amino, C6-12 aryl, C5-12 heterocyclic aryl; the above alkyl or alkoxy group of C1-6, cycloalkyl group of C3-6, heterocycloalkyl group of C3-6, amino group, aryl group of C6-12, and H on the heteroaryl group of C5-12 may be optionally substituted by one or more halogens, -OH, -CN, -NH 2 、-NO 2 -SH, sulfonic acid substitution;
the ring Ar is fused with the ring Rg, and the fused bond is any bond on the ring Ar;
ring Ar is selected from aromatic five-membered heterocyclic group, aromatic six-membered heterocyclic group or phenyl, the aromatic five-membered heterocyclic group is selected from: imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, furanyl or thienyl; the aromatic six-membered heterocyclic group is selected from: pyridyl, pyridazinyl, pyrimidinyl or pyrazinyl; h on the aromatic five-membered heterocyclic group, the aromatic six-membered heterocyclic group or the phenyl group which is optional may be substituted by: halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, substituted or unsubstituted aryl or heteroaryl; the substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyamino, alkyl, cycloalkyl, heterocycloalkyl, aryl, heterocycloaryl, ester, acyl, carbonyl, amide, sulfonyl, phosphoryl;
ring Rg is selected from C3-8 saturated/unsaturated cycloalkyl or C3-8 saturated/unsaturated heterocycloalkyl containing at least one O, N, S, and H on said C3-8 saturated/unsaturated cycloalkyl or C3-8 saturated/unsaturated heterocycloalkyl containing at least one O, N, S may be substituted with: halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, substituted or unsubstituted aryl or heteroaryl; the substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyamino, alkyl, cycloalkyl, heterocycloalkyl, aryl, heterocycloaryl, ester, acyl, carbonyl, amide, sulfonyl, phosphoryl;
having at least one R on the ring Rg 7 Said R is 7 Independently selected from-H, deuterium, tritium, halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyamino, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, aryl, heteroaryl, ester, acyl, carbonyl, amide, sulfonyl, phosphoryl.
2. A compound of claim 1, wherein R is 1 Selected from the group consisting of substituted or unsubstituted phenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, furanyl, thienyl, pyrrolyl, five or six membered heterocycloalkyl containing at least one N or O atom; the substituent groups are independently selected from-F, -Cl, -Br, -OH, -CN and-NH 2 Methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl, hydroxyethyl.
3. A compound of claim 2, wherein R is 1 Has any one of the following structures:
Figure FDA0003898749600000031
h on any one or more C atoms in the above structure may be substituted with: -F, -Cl, -Br, -OH, -CN, -NH 2 Substituted or unsubstituted amido, substituted or unsubstituted C1-3 alkyl or alkoxy, C3-6 cycloalkyl; the substituent groups are independently selected from-F, -Cl, -Br, -OH, -CN and-NH 2 Methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl, hydroxyethyl.
4. The compound of claim 1, wherein said compound has the structure of formula II, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure FDA0003898749600000041
/>
x is selected from substituted or unsubstituted C1-6 straight chain/branched alkyl, substituted or unsubstituted C3-10 cycloalkyl or heterocycloalkyl, substituted or unsubstituted C5-6 aryl or heteroaryl, alkynyl, alkenyl, amido and ester groups; the substituent group is selected from halogen, -OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyl amino, alkyl, alkoxy, hydroxyl alkyl, cycloalkyl, heterocyclic alkyl, aromatic group, heterocyclic aromatic group, ester group, acyl, carbonyl, amide, sulfonyl, phosphoryl;
having at least one R on the X group 8 ,R 8 Independently selected from-H, halogen, -OH, -CN, -NH 2 Substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, said substituent being selected from the group consisting of halogen, -OH, -CN, -NH 2 、-NO 2 、-SH、-CF 3 、-CHF 2 、-CH 2 F. Carboxyl, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxyA group, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, phenoxy, pyridyl, amido, sulfonyl, phosphoryl;
R 9 is selected from substituted or unsubstituted phenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, the substituted groups are independently selected from-F, -Cl, -Br, -OH, -CN, -NH 2 Methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl;
R 10 is selected from-NH 2 OH, -OH, halogen, C1-6 alkyl or alkoxy, C3-6 cycloalkyl, C3-6 heterocycloalkyl; h on the above C1-6 alkyl or alkoxy, C3-6 cycloalkyl, C3-6 heterocycloalkyl may optionally be substituted with one or more halogens, -OH, -CN, -NH 2 、-NO 2 -SH substitution;
the ring Rg is selected from C3-8 saturated/unsaturated heterocycloalkyl containing at least one of O, N and S, and at least one R is present on the ring Rg 11 ,R 11 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, -OH, -CN, -NH 2 、-NO 2 SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, phenoxy, sulfonyl, phosphoryl.
5. The compound of claim 4, wherein the compound has a structure according to formula III, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure FDA0003898749600000051
R 12 selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, and cycloHexyl, hydroxymethyl;
R 13 、R 14 independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl;
R 15 selected from-H, -F, -Br, -OH, -CN, -NH 2 Substituted or unsubstituted C1-10 straight/branched alkyl, substituted or unsubstituted C1-10 straight/branched alkoxy, substituted or unsubstituted cycloalkyl or heterocycloalkyl, wherein H in the above groups may be substituted with: -F, -Cl, -Br-OH, -CN, -NH 2 、-CF 3 Hydroxymethyl, methyl, ethyl, propyl, isopropyl, phenyl, methoxy, ethoxy, propoxy, isopropoxy, phenoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
6. The compound of claim 5, wherein said substituted or unsubstituted cycloalkyl or heterocycloalkyl group comprises a monocyclic, bridged, spiro or fused cycloalkyl or heterocycloalkyl group, and includes both saturated and unsaturated forms.
7. The compound of claim 6, wherein said substituted or unsubstituted cycloalkyl or heterocycloalkyl comprises
Figure FDA0003898749600000061
Figure FDA0003898749600000062
Indicates a position to which an alkynyl group may be bonded.
8. The compound of claim 4, wherein said compound has a structure according to formula iv, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure FDA0003898749600000063
R 16 、R 17 independently selected from-H, -F, -Cl, -Br, -OH, -CN and-NH 2 -SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl;
R 18 selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl;
R 19 、R 20 independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
9. The compound of claim 4, wherein the compound has a structure according to formula v, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof:
Figure FDA0003898749600000071
R 21 selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl;
R 22 、R 23 independently selected from H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl;
y is selected from substituted or unsubstituted C1-4 straight chain/branched alkyl, substituted or unsubstituted C3-8 cycloalkyl or heterocycloalkyl, substituted or unsubstituted C5-6 aryl or heteroaryl, amido and ester groups; said substituent groupThe group is selected from-OH, -CN, -NH 2 、-NO 2 SH, carboxyl, hydroxyamino, alkyl, alkoxy, hydroxyalkyl, substituted or unsubstituted C3-8 cycloalkyl or heterocycloalkyl, C5-6 aryl or heteroaryl, ester, acyl, carbonyl, amide, sulfonyl, phosphoryl;
the R is 24 Selected from-H, deuterium, tritium, -F, -Cl, -Br, -OH, -CN, -NH 2 、-NO 2 SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, phenyl, phenoxy, substituted or unsubstituted C3-8 saturated or unsaturated cycloalkyl or heterocycloalkyl.
10. The compound of claim 9, wherein said substituted or unsubstituted C3 _ 8 cycloalkyl or heterocycloalkyl comprises
Figure FDA0003898749600000081
Figure FDA0003898749600000082
Represents a position connectable to Y;
h in the above structure may be substituted with: -OH, -CN, -NH 2 、-NO 2 SH, hydroxylamino, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, phenyl, phenoxy, sulfonyl, phosphoryl.
11. The compound of claims 1-10, wherein the compound has the structure:
Figure FDA0003898749600000083
Figure FDA0003898749600000091
12. a pharmaceutical composition comprising a compound of any one of claims 1-11, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof.
13. Use of a compound of any one of claims 1-11, or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof, or a pharmaceutical composition of claim 12 in the manufacture of a medicament for the treatment of a disease associated with expression or activity of PAK4 kinase.
14. The use according to claim 13, wherein the disease associated with expression or activity of PAK4 kinase comprises cancer, neurodegenerative disease or immune system disease.
15. The use of claim 14, wherein the cancer comprises breast cancer, mantle cell lymphoma, ovarian cancer, esophageal cancer, laryngeal cancer, glioblastoma, neuroblastoma, gastric cancer, hepatocellular cancer, gastric cancer, glioma, endometrial cancer, melanoma, renal cancer, bladder cancer, melanoma, bladder cancer, biliary tract cancer, renal cancer, pancreatic cancer, lymphoma, hairy cell cancer, nasopharyngeal cancer, pharyngeal cancer, large bowel cancer, rectal cancer, brain and central nervous system cancer, cervical cancer, prostate cancer, testicular cancer, genitourinary tract cancer, lung cancer, non-small cell lung cancer, small cell cancer, lung adenocarcinoma, bone cancer, colon cancer, adenoma, pancreatic cancer, adenocarcinoma, thyroid cancer, follicular cancer, hodgkin leukemia, bronchial cancer, thyroid cancer, uterine corpus cancer, cervical cancer, multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia, lymphocytic leukemia, chronic lymphoid leukemia, myeloid leukemia, non-hodgkin lymphoma, primary macroglobulinemia, rhabdomyosarcoma.
16. A compound of the general formula VI,
Figure FDA0003898749600000101
wherein R is 25 、R 26 Independently selected from-H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
17. A compound of the general formula VII,
Figure FDA0003898749600000102
wherein R is 27 Selected from the group consisting of-H, -F, -Cl, -Br, -OH, -CN, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, hydroxymethyl;
R 28 、R 29 independently selected from H, deuterium, tritium, -F, -Cl, -Br, hydroxymethyl, hydroxyethyl, methyl, ethyl, propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl.
18. Use of a compound of formula vi and/or formula vii for the preparation of a compound according to any one of claims 1 to 11 or a pharmaceutically acceptable salt, stereoisomer, ester, prodrug, solvate thereof.
CN202211282529.8A 2022-10-19 2022-10-19 PAK4 kinase inhibitor and preparation method and application thereof Pending CN115925694A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202211282529.8A CN115925694A (en) 2022-10-19 2022-10-19 PAK4 kinase inhibitor and preparation method and application thereof
PCT/CN2023/118200 WO2024082872A1 (en) 2022-10-19 2023-09-12 Pak4 kinase inhibitor, and preparation method therefor and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211282529.8A CN115925694A (en) 2022-10-19 2022-10-19 PAK4 kinase inhibitor and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN115925694A true CN115925694A (en) 2023-04-07

Family

ID=86699711

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211282529.8A Pending CN115925694A (en) 2022-10-19 2022-10-19 PAK4 kinase inhibitor and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN115925694A (en)
WO (1) WO2024082872A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024082872A1 (en) * 2022-10-19 2024-04-25 成都海博为药业有限公司 Pak4 kinase inhibitor, and preparation method therefor and use thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4017211A1 (en) * 1990-05-29 1991-12-05 Merck Patent Gmbh OXAZOLIDINONE
DE60320933D1 (en) * 2002-01-10 2008-06-26 Bayer Healthcare Ag RHO-KINASE INHIBITORS
CN114075175A (en) * 2020-08-14 2022-02-22 成都海博为药业有限公司 Compound serving as PAK4 kinase inhibitor and preparation method and application thereof
CN115925694A (en) * 2022-10-19 2023-04-07 成都海博为药业有限公司 PAK4 kinase inhibitor and preparation method and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024082872A1 (en) * 2022-10-19 2024-04-25 成都海博为药业有限公司 Pak4 kinase inhibitor, and preparation method therefor and use thereof

Also Published As

Publication number Publication date
WO2024082872A1 (en) 2024-04-25

Similar Documents

Publication Publication Date Title
AU2012269648B2 (en) Therapeutically active compositions and their methods of use
EP2990405B1 (en) Deuterated diaminopyrimidine compounds and pharmaceutical compositions comprising such compounds
JP6139782B2 (en) Substituted pyrazolopyrimidine compounds, and pharmaceutically acceptable salts thereof, and solvates, stereoisomers, and tautomers thereof, and pharmaceutical compositions containing them
WO2020094104A1 (en) Nitrogen-containing fused heterocyclic shp2 inhibitor compound, preparation method, and use
EP4056563A1 (en) Compound as shp2 inhibitor and use thereof
EP3904351A1 (en) Fak inhibitor and drug combination thereof
CN109867676B (en) Pyrrolopyrimidine derivative compound, pharmaceutical composition and application thereof
EP3679029B1 (en) Imidazolidine compounds
JP2024505732A (en) Pyridopyrimidinone derivatives and their production methods and uses
EP3929185A1 (en) Nitrogen-containing fused cyclic compound, preparation method therefor and use thereof
EP3939979A1 (en) Jak kinase inhibitor, preparation method for same, and applications thereof in field of medicine
WO2024082872A1 (en) Pak4 kinase inhibitor, and preparation method therefor and use thereof
EP4339193A1 (en) Compound having anti-tumor activity and use thereof
CN114075175A (en) Compound serving as PAK4 kinase inhibitor and preparation method and application thereof
US11891405B2 (en) Furo[3,4-b]pyrrole-containing BTK inhibitor
CN110066272B (en) Substituted benzo [ d ] imidazoles and pharmaceutical compositions thereof
CN112125914B (en) 5-substituted berbamine derivatives, preparation method and application thereof
EP3992193A1 (en) Pyrazolopyrimidine compound, preparation method for same, and applications thereof
JPWO2019235553A1 (en) Azetidine derivatives and their prodrugs
US11236086B2 (en) Substituted pyrrolopyridines as inhibitors of activin receptor-like kinase
TW202417436A (en) Pak4 kinase inhibitor and preparation method and use thereof
CN113993871B (en) BTK inhibitors containing 5-azacycloheptane
CN112209933B (en) BTK inhibitors containing 4-azacycloheptane
TW202233608A (en) A compound as pak4 kinase inhibitor and preparation method and use thereof
EP4082611A1 (en) Azetidine sulfonamide compound

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