WO2024061118A1 - 大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用 - Google Patents

大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用 Download PDF

Info

Publication number
WO2024061118A1
WO2024061118A1 PCT/CN2023/119030 CN2023119030W WO2024061118A1 WO 2024061118 A1 WO2024061118 A1 WO 2024061118A1 CN 2023119030 W CN2023119030 W CN 2023119030W WO 2024061118 A1 WO2024061118 A1 WO 2024061118A1
Authority
WO
WIPO (PCT)
Prior art keywords
fragment
methyl
amino
benzo
amine
Prior art date
Application number
PCT/CN2023/119030
Other languages
English (en)
French (fr)
Inventor
朱振东
牛德强
凡明
李旭珂
Original Assignee
科辉智药生物科技(无锡)有限公司
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 科辉智药生物科技(无锡)有限公司 filed Critical 科辉智药生物科技(无锡)有限公司
Publication of WO2024061118A1 publication Critical patent/WO2024061118A1/zh

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/16Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • the present invention relates to the field of medicinal chemistry, specifically to a class of macrocyclic nitrogen-containing crown ether compounds and their stereoisomers, tautomers or pharmaceutically acceptable salts, their preparation methods and medicines containing the compounds. combination.
  • the invention also relates to the use of such compounds as protein kinase inhibitors.
  • Protein kinase is an enzyme protein widely present in cells and on the cell surface. So far, more than 500 protein kinases have been discovered and identified. They belong to a family of structurally related proteins whose members are known to be involved in nearly all cellular signaling activities.
  • the catalytic function of protein kinase is to transfer the ⁇ -phosphate group in the ATP (or GTP) molecule to a specific threonine, serine or tyrosine group on the target protein, causing the target protein conformation to change, leading to the target protein function Transition from resting state to active state.
  • protein kinases are divided into two major categories: serine/threonine protein kinases and tyrosine protein kinases.
  • protein kinases play an extremely important role in the normal functions of cells and organs, including cell growth, differentiation, proliferation, angiogenesis, apoptosis, cytoskeletal arrangement, regulation of metabolic reactions, membrane transport and Cell movement, etc.
  • the non-catalytic functions of protein kinases also play an indispensable role, including allosteric effects, subcellular targeting, protein complex scaffolding, protein competitive interactions, and DNA binding.
  • protein kinases undergo genetic mutations or protein kinases are overexpressed, dysregulated protein kinases can lead to a variety of pathological changes, including cancer, inflammation, autoimmune diseases, cardiovascular system and neurological diseases. Therefore, protein kinases have become one of the most important targets in today's drug development. In recent years, protein kinase inhibitors have continued to achieve success in clinical treatment and have been approved by drug regulatory organizations around the world, bringing major breakthroughs in clinical disease treatment.
  • the present invention specifically relates to a class of macrocyclic nitrogen-containing crown ether compounds and their application as inhibitors of protein kinases, including but not limited to protein kinases such as LRRK2, JAK1, JAK2, JAK3, EGFR and CDK9.
  • LRRK2 Leucine-rich repeat kinase 2
  • PD Parkinson's disease
  • LRRK2 gene mutations are commonly found in G2019S and I2020T in the kinase active region and R1441G/C/H and Y1699C in the GTPase region Mutations, these mutations can lead to increased LRRK2 protein kinase activity, leading to a variety of pathological changes, including increased -synuclein and TAU protein phosphorylation, nervous system inflammation, and neuronal mitochondrial dysfunction.
  • PD is the second most common neurodegenerative disease after Alzheimer’s Disease, with more than 10 million patients worldwide, but no effective treatment has been found yet. 10% of PD patients have a significant family history, with LRRK2 mutations being the most common cause. This mutation occurs in approximately 5% of familial PD and 1% of non-familial PD, and the clinical manifestations caused by these mutations are indistinguishable from sporadic PD. In addition, LRRK2 mutations can also lead to pathological changes in TAU protein, and pathological changes in TAU are a major feature of Alzheimer's disease. Therefore, LRRK2 may be located in the upstream stage of the pathogenesis of neurodegenerative diseases and also plays an important role in other neurodegenerative diseases. Therefore, the development of LRRK2-specific inhibitors has become one of the effective ways to treat PD and other neurodegenerative diseases.
  • Janus kinase is a non-receptor tyrosine protein kinase. Its family contains four members: JAK1, JAK2, JAK3 and TYK2. It is a cytoplasmic tyrosine kinase that transduces cytokine signals from membrane receptors to STAT transcription factors. Acid kinase. JAK-STAT related diseases include rheumatoid arthritis (RA), asthma, ankylosing spondylitis, lupus erythematosus, psoriasis, vitiligo and other autoimmune diseases.
  • RA rheumatoid arthritis
  • asthma asthma
  • ankylosing spondylitis lupus erythematosus
  • psoriasis vitiligo and other autoimmune diseases.
  • pan-JAK inhibitors show good efficacy in the treatment of inflammatory and tumor diseases, they can produce off-target effects at higher doses, causing some serious side effects and causing adverse reactions. Therefore, there is still a need to develop new inhibitors targeting different JAK subtypes for the treatment of autoimmune diseases.
  • highly selective JAK1 inhibitors are being studied clinically for the treatment of rheumatoid arthritis (RA), asthma, and dermatitis, and may be safer.
  • Cyclin-dependent kinase belongs to the serine/threonine kinase family and is a family of protein kinases that regulate the cell cycle and gene transcription. During different cell cycle stages, CDKs phosphorylate specific cell cycle proteins to regulate cellular activities.
  • CDK9 is one of the most important CDKs that cooperates with four cyclins, including cyclin T1, cyclin K, cyclin T2a and T2b. CDK9 binds to cell cyclins to form a heterodimer of positive transcription elongation factor b (P-TEFb).
  • P-TEFb phosphorylates the C-terminal domain (CTD) of RNA polymerase II, allowing transcription to extend from the start site, and is the core molecule for transcription elongation.
  • CTD C-terminal domain
  • Dysregulation of CDK9 kinase activity can lead to a variety of diseases, including highly proliferative diseases (such as cancer), viral infections, or cardiovascular diseases.
  • CDK9 Given the critical role of CDK9 in disease, downregulation of CDK9 provides an excellent opportunity to develop targeted therapies for cancer and other diseases. Although many small molecule CDK inhibitors have been reported, most of them lack selectivity for specific CDKs and thus exhibit low efficacy and high adverse event rates in clinical practice. Therefore, the development of CDK9-specific targeting inhibitors is crucial for the treatment of human diseases.
  • heterocyclic compounds containing macrocyclic nitrogen crown ethers of the present invention exhibit good inhibitory activity against protein kinases; moreover, such compounds also exhibit good water solubility.
  • the present invention provides a compound having the structure of formula (I) or its stereoisomer, tautomer or pharmaceutically acceptable salt:
  • Z means absence (covalent bond) or -OCH 2 CH 2 -;
  • L represents the absence of (covalent bond), -O-, -NH- or -N(C 1 ⁇ 4 alkyl)-;
  • A is an aryl group optionally substituted by 1 or 2 substituents selected from the following, 5-12 membered heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocycle group or 5-18 membered bridged ring group, wherein the substituent is selected from: deuterium, C 1-4 alkyl group, C 1-10 alkoxy group, C 3-6 cycloalkyl group, 3-8 membered heterocyclyl group , C 1 ⁇ 4 alkylphosphoryl, C 1 ⁇ 4 alkylsulfonyl, aminosulfonyl, C 1 ⁇ 4 alkylsulfamoyl, cyano C 3 ⁇ 6 cycloalkyl, C 2 ⁇ 4 alkenylmethyl Acyl group, C 2 to 4 alkenylformamido, cyano C 1 to 4 alkylcarbamoyl group, cyano C 1 to 4 alkyl group, C 1 to 4 alkylformyl-3 to 8-member
  • R 1 is hydrogen, or optionally substituted C 1 to 4 alkyl, C 3 to 6 cycloalkyl, C 1 to 4 alkyl-CO-, -CO or C 1 to 4 alkylsulfonyl, wherein The substituent is selected from: deuterium, C 1 to 4 alkyl group, C 1 to 10 alkoxy group, C 3 to 6 cycloalkyl group, oxo, cyano group, hydroxyl group, amino group, dimethylamino group, and hydroxylamine group;
  • R 2 is independently selected from hydrogen, C 1 to 3 alkyl, trifluoromethyl, C 1 to 3 alkoxy, cyano and halogen;
  • n 1 or 2;
  • Ra is selected from hydrogen, amino, trifluoromethyl, halogen, cyano, C 1 ⁇ 3 alkyl, acetyl, C 1 ⁇ 3 alkylphosphoryl and C 1 ⁇ 3 alkylsulfonyl;
  • Rb is selected from hydrogen, amino and C 1-3 alkylamino
  • Ra, Rb and the C atoms to which they are connected can together form a 5-6 membered aromatic ring, a 5-12 membered heteroaromatic ring or a 5-8 membered heterocyclic ring;
  • the compound of Formula (I) has the following structure of Formula (II) or Formula (III):
  • L, A, R 1 , R 2 , Ra and Rb are as defined above.
  • the compound of Formula I has the following structure of Formula (IV):
  • X and Y are each independently selected from C and N;
  • Z means absence (covalent bond) or -OCH 2 CH 2 ;
  • L, A, R 1 , R 2 and n are as defined above.
  • A may preferably have the following structure:
  • a pharmaceutical composition comprising the compound of formula (I) according to the present invention or its stereoisomer, tautomer or pharmaceutically acceptable salt, and any Select a pharmaceutically acceptable carrier.
  • the compound of Formula (I) has structures (II)-(IV) described above.
  • the compound of formula (I) is the specific compound described above.
  • the above compounds of the present invention or their stereoisomers, tautomers or pharmaceutically acceptable salts or pharmaceutical compositions thereof are provided for use in the treatment or prevention of protein kinase-related diseases. application in medicines.
  • the protein kinase-related diseases are diseases in which protein kinases are involved in signaling, mediating, modulating or modulating disease progression or symptoms.
  • the protein kinase is selected from LRRK2, JAK1, JAK2, JAK3, EGFR and CDK9.
  • protein kinase-related diseases include the following disease types: cancer, autoimmune diseases, metabolic diseases, inflammation, infections (bacteria, viruses, yeast, Fungi, etc.), central nervous system diseases, degenerative neurological diseases, allergies/asthma, skin diseases, vasculogenesis, neovascularization, angiogenesis, cardiovascular diseases, etc.
  • the protein kinase-related diseases of the present invention are selected from neurodegenerative diseases, autoimmune diseases and tumors. In other preferred embodiments, the protein kinase-related diseases of the present invention are selected from Parkinson's disease, asthma, dermatitis, non-small cell lung cancer, acute myeloid leukemia (AML) and liver cancer.
  • transplant rejection e.g., kidney, liver, heart, lung, pancreas (islet cells), bone marrow, cornea, small intestine, skin allograft transplant or xenograft
  • graft-versus-host disease osteoarthritis, rheumatoid arthritis, multiple sclerosis, diabetes, diabetic retinopathy, asthma, inflammatory bowel disease (Crohn's disease, ulcers colitis), kidney disease, cachexia, septic shock, lupus, diabetes, myasthenia gravis, psoriasis, dermatitis, eczema, seborrhea, Alzheimer's disease, Parkinson's disease, stem cell protection in chemotherapy, autologous Or in vitro selection or purging during allogeneic bone marrow transplantation, leukemia (acute myeloid leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia, and the like.
  • leukemia acute myeloid leukemia, chronic myelogen
  • compositions and methods described herein are used to treat or prevent cancer, eye disease, or retinopathy.
  • compositions and methods described herein are used to treat or prevent rheumatoid arthritis, transplant rejection, asthma or allergy, or symptoms thereof.
  • compositions and methods described herein are used to treat or prevent diseases or disease symptoms associated with hyperproliferative diseases, or, alternatively, with vasculogenesis.
  • Optionally substituted in this article means that the groups described in A, R 1 , etc. may or may not be substituted by substituents. That is, it is not limited to the case of being substituted by the listed substituents, but also includes not being substituted by substituents. Substitution of the listed substituents.
  • R is substituted or unsubstituted C 1 to 10 alkyl, C 3 to 6 cycloalkyl or heterocycloalkyl, phenyl, naphthyl or indolyl, where the substituent is C 1 to "4 alkyl, C 1 to 4 alkoxy, cyano group, hydroxyl, mercapto, amino or halogen" are expressed in the same way, but the limitation of substituted or unsubstituted does not only refer to C 1 to 10 alkyl in a narrow sense, but Applies to all groups described.
  • the substituent may be deuterium, C 1 to 4 alkyl, C 1 to 10 alkoxy, C 3 to 6 cycloalkyl, 3 to 8 membered heterocyclyl, C 1 to 4 alkylphosphoryl, C 1 to 4 alkylsulfonyl, aminosulfonyl, C 1 to 4 alkylsulfamoyl, cyano, C 3 to 6 cycloalkyl, C 2 to 4 alkenylformyl , C 2 ⁇ 4 alkenylcarbamoyl group, cyano C 1 ⁇ 4 alkylcarbamoyl group, Cyano C 1 to 4 alkyl, C 1 to 4 alkylformyl-3 to 8-membered heterocyclyl, C 3 to 6 cycloalkylformyl, 3 to 8-membered heterocyclyl-sulfonyl, C 1 to 4 Alkylsulfonyl-3 ⁇ 8-membere
  • alkyl is used to represent a straight-chain or branched saturated hydrocarbon group.
  • C 1 to 3 alkyl refers to a saturated hydrocarbon group containing 1 to 3 carbon atoms
  • C 1 to 4 alkyl refers to a saturated hydrocarbon group containing 1 to 4 carbon atoms.
  • An alkyl group with 1 to 10 carbon atoms refers to a saturated hydrocarbon group with 1 to 10 carbon atoms, including straight, branched or cyclic alkyl groups.
  • alkoxy refers to alkyl-O-.
  • C 1-6 alkoxy is intended to include C 1 , C 2 , C 3 , C 4 , C 5 , C 6 alkoxy. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (eg, n-propoxy and isopropoxy), and tert-butoxy.
  • C 1-10 alkoxy refers to an -O-alkyl group, which includes straight-chain, branched-chain and cyclic alkyl groups having 1 to 10 carbon atoms.
  • alkoxy groups include methoxy, ethoxy, propoxy (eg, n-propoxy, isopropoxy and cyclopropoxy), tert-butoxy and the like.
  • Preferred alkoxy groups here are C 1 to 6 alkoxy groups and C 1 to 4 alkoxy groups.
  • cycloalkyl refers to a non-aromatic carbocyclic group, including cyclized alkyl groups.
  • the cycloalkyl group may include a monocyclic, bicyclic or polycyclic system. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, etc.
  • C3-8 cycloalkyl groups refer to cycloalkyl groups containing 3 to 8 carbon atoms. Preferred cycloalkyl groups herein are C3-8 cycloalkyl groups and C3-6 cycloalkyl groups.
  • heterocycloalkyl or “heterocyclyl” refers to a non-aromatic heterocycloalkyl group in which 1 or 2 or 3 ring-forming carbon atoms are replaced by heteroatoms such as O, N or S atoms. Heterocycloalkyl groups preferably have 3, 4, 5, 6 or 7 ring atoms. The preferred heterocyclyl groups herein are C 3-8 heterocyclyl groups.
  • Aryl refers to an aromatic carbocyclic group, including monocyclic, bicyclic, tricyclic or polycyclic aromatic hydrocarbons, such as phenyl, naphthyl, anthracenyl, phenanthrenyl, etc.
  • Aryl groups are preferably monocyclic, bicyclic or tricyclic ring systems having 6 to 14 or 6 to 12 ring members, wherein at least one ring in said system is aromatic and wherein each ring in said system Rings contain 3 to 7 ring members.
  • Substituted aryl means that at least one hydrogen atom on the benzene ring of the aryl is substituted by a non-hydrogen moiety.
  • the substituent of the aryl can be halogen, C 1 to 10 alkoxy, -CN, -OH, -SH , -NH 2 , C 1 ⁇ 6 alkyl.
  • Preferred aryl groups include phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl and tetrahydronaphthyl.
  • aralkyl or "arylalkyl” refers to an alkyl residue attached to an aryl ring. Non-limiting examples include benzyl, phenethyl, and the like.
  • the fused aryl group can be attached to another group at a suitable position on the cycloalkyl ring or aromatic ring.
  • heteroaryl means a stable 5-12 membered aromatic monocyclic or aromatic bicyclic or aromatic polycyclic heterocyclic ring, which is completely unsaturated or partially unsaturated, and which contains carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from N, O and S. It includes 5-membered, 6-membered or 7-membered aromatic monocyclic rings or 8-membered, 9-membered, 10-membered, 11-membered or 12-membered aromatic bicyclic or aromatic polycyclic heterocyclic rings; preferably any heterocyclic ring defined above and Fused benzene rings. Nitrogen and sulfur heteroatoms may optionally be oxidized.
  • Nitrogen atoms are substituted or unsubstituted (ie, N or NR, where R is H or another substituent, if defined).
  • Heterocycles can be attached to their pendant groups at any heteroatom or carbon atom that results in a stable structure. If the resulting compound is stable, the heterocyclyl groups described herein may be substituted on the carbon or nitrogen atom.
  • the nitrogen in the heterocycle may optionally be quaternized. Preferably, when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to each other.
  • aryl hetero groups include, but are not limited to, acridinyl, azetidinyl, azecinyl, benzimidazolyl, benzofuryl, benzothiofuranyl, benzothienyl, benzox Azolyl, benzoxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, quinolyl, decahydroquinolyl, dihydrofura[2,3-b]tetrahydrofuranyl, furanyl, furanyl, imidazole Alkyl, imidazolinyl, imidazolyl, 1H-indazolyl, imidazopyridyl, indolyl, indolin
  • heteroaryl may also include biaryl groups formed from an “aryl” as defined above and a monocyclic “heteroaryl” Structures, such as but not limited to "-phenylbipyridyl-", “-phenylbipyrimidinyl”, “-pyridylbiphenyl”, “-pyridylbipyrimidinyl-", “-pyrimidinylbiphenyl”base-”.
  • C 1-4 alkylphosphoryl refers to a phosphoryl group substituted by C 1-4 alkyl groups.
  • the two alkyl groups on the phosphoryl group may be the same or different.
  • C 1-4 alkylsulfonyl refers to a C 1-4 alkyl substituted sulfonyl group.
  • halogen includes fluorine, chlorine, bromine and iodine.
  • Bridged cyclic group refers to a polycyclic group with 5 to 18 members, preferably 5 to 14 members, containing two or more cyclic structures and sharing two carbon atoms that are not directly connected to each other. More preferably, it is a 6- to 14-membered bridged ring group, and still more preferably, a 7- to 10-membered bridged ring group. According to the number of constituent rings, it can be divided into bicyclic, tricyclic, pyridone or polycyclic bridged heterocyclyl groups, preferably bicyclic, tricyclic or pyridone, more preferably bicyclic or tricyclic. Bridged cyclic groups herein allow for 1, 2 or 3 ring atoms to be heteroatoms selected from nitrogen, oxygen or S(O) n (where n is selected from 0, 1 and 2).
  • the groups or substituents are connected to the parent nucleus at their rightmost end in the customary order from left to right.
  • the position where it is connected to the parent nucleus of the compound as a substituent is the rightmost "formyl".
  • pharmaceutically acceptable salt or “pharmaceutically acceptable salt” used herein refers to certain salts that enable the compounds of the present invention to maintain their original biological activity and are suitable for pharmaceutical use.
  • Pharmaceutically acceptable salts of the compounds represented by formula (I) can be salts formed by carboxyl or amine groups (mainly amine groups) and suitable bases or acids, such as salts formed with suitable bases (including metal salts) , ammonium salt), or a salt with a suitable acid.
  • the compounds of the invention preferably form salts with suitable acids which may be selected from the group consisting of: hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid, nitric acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid , fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid, trifluoroacetic acid and aspartic acid , preferably methanesulfonic acid or hydrochloric acid.
  • suitable acids may be selected from the group consisting of: hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid, nitric acid, formic acid, acetic acid
  • treatment includes any effect resulting in amelioration of a condition, disease, disorder, etc., such as alleviation, reduction, regulation, amelioration or elimination, or amelioration of symptoms thereof.
  • pharmaceutically acceptable carrier means a pharmaceutical substance, composition or vehicle such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc, magnesium stearate , calcium stearate or zinc stearate or stearic acid) or solvent encapsulated substances which involve carrying or transporting the subject compound from one organ or part of the body to another.
  • manufacturing aid e.g., lubricant, talc, magnesium stearate , calcium stearate or zinc stearate or stearic acid
  • solvent encapsulated substances which involve carrying or transporting the subject compound from one organ or part of the body to another.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the patient.
  • composition means a composition comprising a compound of the invention and at least one other pharmaceutically acceptable carrier.
  • “Pharmaceutically acceptable carrier” refers to a medium generally accepted in the art for the delivery of biologically active agents to animals, particularly mammals, including (i.e.) adjuvants, excipients or vehicles such as diluents, preservatives , fillers, flow regulators, disintegrants, wetting agents, emulsifiers, suspending agents, sweeteners, flavoring agents, aromatics, antibacterial agents, antifungal agents, lubricants and dispersants, depending on Mode of administration and nature of dosage form.
  • Brettphos Pd G3 is methane sulfonate (2-dicyclohexylphosphine)-3,6-dimethoxy-2',4',6'-triisopropyl-1,1'- Benzene)(2'-amino-1,1'-biphenyl-2-yl)palladium(II);
  • TLC refers to thin layer chromatography;
  • the chemical shift value ( ⁇ ) is in ppm, using tetramethylsilane (TMS) or residual solvent peak as the internal standard, the coupling constant (J) is in Hertz (Hz), and the multiplex of the peak shape in the 1 H NMR spectrum
  • TMS tetramethylsilane
  • J Hertz
  • the instrument used for liquid chromatography-mass spectrometry is Shimadzu LCMS-2020, and the instrument used for preparative high-performance liquid chromatography (Prep-HPLC) is Bonna-Agela FLEXA FL-H100G.
  • the thin layer chromatography silica gel plate model used in thin layer chromatography is Yantai Huanghai HSGF254 thin layer chromatography silica gel plate.
  • the specification used for reaction monitoring is 2.5 ⁇ 8cm, and the coating thickness is 0.2 ⁇ 0.03mm. It is used for separation and purification.
  • the specification is 20 ⁇ 20cm, and its coating thickness is 0.4–0.5mm.
  • Silica gel column chromatography uses Qingdao Marine Silica Gel 100-200 mesh or 200-300 mesh silica gel as the carrier.
  • reaction solution was concentrated and the residue was purified by recrystallization in methanol (30ml) to obtain a white solid (((4-nitrophenyl)sulfonyl)aminodiyl)bis(ethane-2,1-diol)bis(4-nitrobenzenesulfonate) (5.2g, 83%).
  • Step 4 Intermediate Amine-1-e 4-methyl-9-nitro-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline synthesis
  • Step 5 Synthesis of intermediate Amine-1 4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline-9-amine
  • Step 3 Intermediate Amine-2-d 4-(4-nitro-2-(2-(2-(toluenesulfonyloxy)ethoxy)ethoxy)phenoxy)4-methylbenzene Synthesis of ethyl sulfonate
  • Step 4 Synthesis of the intermediate Amine-2-e 7-(4-methoxybenzyl)-13-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10]azacyclododecane
  • Step 5 Synthesis of intermediate Amine-2-f 13-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10]azacyclododecane
  • the reaction mixture was partitioned between water (20ml) and dichloromethane (30ml). The organic layer was collected, and the aqueous layer was extracted with dichloromethane (10ml x 5). The organic layers were combined, washed with saturated sodium chloride solution (30 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • the crude product was purified by silica gel column chromatography (eluted with 5%-10% methanol in dichloromethane solution). Yellow solid 13-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10]azadodecyl (620mg ,56%).
  • Step 6 Intermediate Amine-2-g 7-methyl-13-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]tri Synthesis of oxa[10]azacyclododecane
  • Step 7 Synthesis of Amine-2 7-methyl-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10]azacyclododecane-13-amine
  • Step 3 Synthesis of intermediate Amine-3-d ethyl 2-(2-(2-hydroxy-4-nitrophenoxy)ethoxy)acetate
  • the crude product obtained was separated and purified by silica gel column chromatography (eluted with 2-5% methanol in dichloromethane solution) to obtain a light 2-(2-(2-(2-hydroxyethoxy)-4-nitrophenoxy)ethoxy)ethanol (500 mg, 57%) as a yellow oil.
  • Step 5 Synthesis of intermediate Amine-3-f 2-(5-nitro-2-(2-(2-(tosyloxy)ethoxy)ethoxy)phenoxy)ethyl-4-toluenesulfonate
  • the reaction mixture was partitioned between water (20 ml) and dichloromethane (30 ml). The organic layer was collected, and the aqueous phase was extracted with dichloromethane (10mlx2). The organic phase layers were combined, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • the crude product was separated and purified by silica gel column chromatography (eluted with 2%-5% methanol in dichloromethane solution). Obtained yellow solid 12-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10]azacyclododecane ( 479 mg, 64%).
  • Step 8 Intermediate Amine-3-i 7-methyl-12-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]tri Synthesis of oxa[10]azacyclododecane
  • Step 9 Amine-3 7-methyl-12-nitro-2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10 ]Synthesis of azacyclododecane-12-amine
  • Step 1 Synthesis of fragment 1-c N-(tert-butyl)-3-((2-chloro-5-fluoropyrimidin-4-yl)amino)benzenesulfonamide
  • reaction solution was concentrated under reduced pressure, and the crude product obtained was separated and purified by silica gel column chromatography (eluting with 15%-25% ethyl acetate in n-hexane solution) to obtain a white solid N-(tert-butyl)-3-((2-chloro -5-Fluoropyrimidin-4-yl)amino)benzenesulfonamide (362 mg, 33%).
  • Step 2 Compound 1N-(tert-butyl)-3-((5-fluoro-2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1, Synthesis of 4,7]dioxazolin-9-yl)amino)pyrimidin-4-yl)amino)benzenesulfonamide
  • Step 1 Synthesis of fragment 2-b(4-((cyanomethyl)carbamoyl)phenyl)boronic acid
  • Step 2 Synthesis of fragment 2-d 2,4-dichloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine
  • Step 3 Fragment 2-e 4-(2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine-4- Synthesis of -N-(cyanomethyl)benzamide
  • the reaction mixture was partitioned between ethyl acetate (10ml) and water (10ml).
  • the organic phase was collected and saturated with chlorination Wash with sodium solution (10 ml), dry over anhydrous sodium sulfate, and concentrate under reduced pressure.
  • the crude product obtained is separated and purified by silica gel column chromatography (eluting with 0.5-2% methanol in dichloromethane solution) to obtain a white solid 4-(2-chloro -7-((2-(Trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-N-(cyanomethyl)benzyl Amide (300 mg, 34%).
  • Step 4 Fragment 2-f N-(cyanomethyl)-4-(2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4, 7]dioxazolin-9-yl)amino)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine-4- Synthesis of benzamide
  • TLC monitored the reaction to be complete.
  • the reaction mixture was cooled to room temperature, partitioned into water (20 ml), and extracted with ethyl acetate (10 ml x 2). Combine the organic phases, wash with saturated sodium chloride solution (20 ml), dry over anhydrous sodium sulfate, and concentrate under reduced pressure.
  • Step 5 N-(cyanomethyl)-4-(2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxan Synthesis of oxazolin-9-yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)benzamide
  • Step 1 Fragment 3-b 3-((2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine- Synthesis of tert-butyl 4-yl)amino)piperidine-1-carboxylate
  • reaction mixture was concentrated under reduced pressure, and the crude product was separated and purified by silica gel column chromatography (eluted with petroleum ether containing 25% ethyl acetate) to give tert-butyl 3-(((2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)piperidine-1-carboxylate (130 mg, 85%) as a white solid.
  • Step 2 Fragment 3-c 3-((2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline- 9-yl)amino)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)piperidine -Synthesis of 1-tert-butyl formate
  • Step 3 Fragment 3-d N 2 -(4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazolin-9-yl) Synthesis of -N 4 -(piperidin-3-yl)-7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine
  • reaction mixture was concentrated and the residue was added to aqueous ammonia (5 ml) and ethyl acetate (5 ml). The reaction mixture was stirred at 40°C for 16 hours. TLC monitored the reaction to be complete. The organic layer was collected, washed with saturated sodium chloride solution (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 4 1-(3-((2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline-9 Synthesis of -yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)piperidin-1-yl)prop-2-en-1-one
  • the reaction mixture was partitioned between water (5 ml) and 10% methanol in dichloromethane (10 ml). The organic layer was collected and the aqueous layer was extracted with 10% methanol in dichloromethane (10ml x 3). The organic layers were combined, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 1 Fragment 4-b N-(tert-butyl)-3-((6-chloro-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4-d Synthesis of ]pyrimidin-4-yl)amino)benzenesulfonamide
  • Fragment 1-b containing 4,6-dichloro-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4-d]pyrimidine (196 mg, 0.83 mmol): A solution of 3-amino-N-(tert-butyl)benzenesulfonamide (189mg, 0.83mmol) and N,N-diisopropylethylamine (128mg, 0.99mmol) in isopropyl alcohol (10ml) was heated to reflux and stirred for reaction 6 Hour. TLC monitored the reaction to be complete. The reaction solution was concentrated, and the residue was partitioned between ethyl acetate (15 ml) and water (20 ml).
  • Step 2 Fragment 4-c N-(tert-butyl)-3-((6-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4 ,7]dioxazolin-9-yl)amino)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino )Synthesis of benzene sulfonamide
  • the resulting mixture was replaced with nitrogen three times and heated to 110°C with stirring for 16 hours.
  • the reaction was monitored by TLC to be complete.
  • the reaction solution was cooled to room temperature and partitioned between water (10 ml) and ethyl acetate (10 ml).
  • the organic layer was collected and the aqueous layer was extracted with ethyl acetate (10 ml x 2).
  • the organic layers were combined, washed with saturated sodium chloride solution (20 ml), and dried over anhydrous sodium sulfate.
  • Step 3 N-(tert-butyl)-3-((6-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]di Synthesis of oxazolin-9-yl)amino)-1H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)benzenesulfonamide
  • Step 1 Synthesis of fragment 5-c 6-chloro-4-(1-methyl-1H-indol-3-yl)-1H-pyrazolo[3,4-d]pyrimidine
  • 1,2-dichloro-pyrazolo[3,4-d]pyrimidine 500 mg, 2.64 mmol
  • N-methyl indole 347 mg, 2.64 mmol
  • aluminum trichloride 500 mg, 2.64 mmol
  • the ethyl chloride (10 ml) solution was heated to reflux for 12 hours. TLC monitored the reaction to be complete.
  • the reaction mixture was partitioned between dichloromethane (10 ml) and water (20 ml).
  • Step 2 Fragment 5-d 6-chloro-4-(1-methyl-1H-indol-3-yl)-1-(2-(trimethylsilyl)ethoxy)methyl)-1H -Synthesis of pyrazolo[3,4-d]pyrimidines
  • reaction mixture was partitioned between ethyl acetate (100 ml) and water (100 ml). Collect the organic layer, wash with saturated sodium chloride solution (100ml x 2), dry with anhydrous sodium sulfate, and concentrate under reduced pressure.
  • the crude product obtained is separated and purified by silica gel column chromatography (eluting with 0.5-1% methanol in dichloromethane solution). Obtained as a white solid 6-chloro-4-(1-methyl-1H-indol-3-yl)-1-(2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolo [3,4-d]pyrimidine (245 mg, 22% in two steps).
  • Step 3 Fragment 5-e 4-methyl-N-(4-(1-methyl-1H-indol-3-yl)-1-((2-(trimethylsilyl)ethoxy) )methyl)-1H-pyrazolo[3,4-d]pyrimidin-6-yl)-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]di Synthesis of oxazoline-9-amine
  • the resulting mixture was replaced with nitrogen three times, heated to 110°C and stirred for 12 hours.
  • the reaction was complete when monitored by TLC.
  • the reaction mixture was cooled to room temperature and partitioned between water (20 ml) and ethyl acetate (10 ml).
  • the organic layer was collected and the aqueous phase was extracted with ethyl acetate (10 ml x 2).
  • the organic layers were combined, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 4 4-Methyl-N-(4-(1-methyl-1H-indol-3-yl)-1H-pyrazolo[3,4-d]pyrimidin-6-yl)-3, Synthesis of 4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline-9-amine
  • reaction solution was concentrated, and the residue was added to a mixed solution containing ammonia water (2 ml) and ethyl acetate (4 ml). The resulting mixture was stirred and reacted at 30°C for 16 hours. TLC monitored the reaction to be complete. The organic layer was collected, washed with saturated sodium chloride solution (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude product was separated and purified by preparative TLC (eluted with 10% methanol in dichloromethane solution) to obtain 4-methane as a yellow solid.
  • Step 3 Fragments 6-d(1r,4r)-1-methylcyclohexane-1,4-diol and 6-e(1s,4s)-1-methylcyclohexane-1,4-diol Synthesis
  • Step 4 Fragment 6-f(1s,4s)-4-((2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2, Synthesis of 3-d]pyrimidin-4-yl)oxy)-1-methylcyclohexan-1-ol
  • the aqueous phase was extracted with ethyl acetate (10ml x 2) . Combine the organic layers, wash with saturated sodium chloride solution (30 ml), dry over anhydrous sodium sulfate, and concentrate under reduced pressure.
  • the crude product obtained is separated and purified by silica gel column chromatography (eluted with n-hexane containing 25% ethyl acetate) to obtain Colorful oil (1s,4s)-4-((2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d] Pyrimidin-4-yl)oxy)-1-methylcyclohexan-1-ol (100 mg, 79%).
  • Step 5 Fragment 6-g(1s,4s)-1-methyl-4-((2-((7-methyll-2,3,6,7,8,9-hexahydro-5H-benzene And[b][1,4,7]trioxa[10]azacyclododecyl-12-yl)amino)-7-((2-(trimethylsilyl)ethoxy) Synthesis of methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)cyclohexan-1-ol
  • Reaction time is 14 hours. TLC monitored the reaction to be complete.
  • the reaction mixture was cooled to room temperature and partitioned between water (20 ml) and ethyl acetate (10 ml). The organic layer was collected and the aqueous layer was extracted with ethyl acetate (10ml x 2). The organic layers were combined, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 6 (1s,4s)-1-methyl-4-((2-((7-methyl-2,3,6,7,8,9-hexahydro-5H-benzo[b][ 1,4,7]trioxa[10]azacyclododecyl-12-yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)cyclohexan- Synthesis of 1-alcohol
  • Step 1 Fragment 7-b(1s,4s)-4-((2-chloro-5-(trifluoromethyl)pyrimidin-4-yl)oxy)-1-methylcyclohexan-1-ol and fragments Synthesis of 7-c(1s,4s)-4-((4-chloro-5-(trifluoromethyl)pyrimidin-2-yl)oxy)-1-methylcyclohexan-1-ol
  • the reaction mixture was quenched with saturated ammonium chloride solution (10ml) and extracted with ethyl acetate (10ml x 2).
  • the organic layer was collected, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • the crude product was separated and purified by silica gel column chromatography (eluted with n-hexane containing 6%-12.5% ethyl acetate).
  • Step 2 (1s,4s)-1-methyl-4-((2-((7-methyl-2,3,6,7,8,9-hexahydro-5H-benzo[b][ 1,4,7]trioxa[10]azacyclododecyl-12-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)oxy)cyclohexan-1-ol Synthesis
  • TLC monitored the reaction to be complete.
  • the reaction mixture was cooled to room temperature, Partition between water (10 ml) and ethyl acetate (10 ml).
  • the organic layer was collected and the aqueous layer was extracted with ethyl acetate (10ml x 2).
  • the organic layers were combined, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 1 Fragment 8-a N-(4-chloro-5-(trifluoromethyl)pyrimidin-2-yl)-4-methyl-3,4,5,6-tetrahydro-2H-benzo[ Synthesis of b][1,4,7]dioxazoline-9-amine
  • the second eluting fraction was yellow solid N-(4-chloro-5-(trifluoromethyl)pyrimidin-2-yl)-4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazolin-9-amine (406 mg, 28%);
  • Step 2 2-Methyl-1-((2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline Synthesis of -9-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)propan-2-ol
  • the implementation method is the same as method 1, except that fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 2-d is used to replace fragment 4-a, fragment 10-a: 2-(isopropylsulfonyl)aniline is used to replace fragment 1-b, and fragment Amine-2 is used to replace fragment Amine-1.
  • fragment 2-d is used to replace fragment 4-a
  • fragment 10-a 2-(isopropylsulfonyl)aniline is used to replace fragment 1-b
  • fragment Amine-2 is used to replace fragment Amine-1.
  • the title compound was obtained.
  • the implementation method is the same as method 4, except that fragment 4-a is replaced with fragment 2-d, fragment 1-b is replaced with fragment 2-(isopropylsulfonyl)aniline, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 4-a is replaced with fragment 2-d, fragment 1-b is replaced with fragment 2-(isopropylsulfonyl)aniline, and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment 5-a is replaced with fragment 1-a to obtain the title compound.
  • the implementation method is the same as method 2, replacing fragment 2-d with fragment 1-a, replacing fragment 2-b with fragment 14-e, and replacing fragment Amine-1 with fragment Amine-2 to obtain the title compound.
  • Step 1 Synthesis of Fragment 14-a 2-(azetidine-3-alkylene)acetonitrile trifluoroacetate
  • Step 2 Synthesis of fragment 14-c 2-(1-(ethylsulfonyl)-3-azetidinylidene)acetonitrile
  • Step 3 Fragment 14-e 2-(1-(ethylsulfonyl)-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborane-2) -Synthesis of -1H-pyrazol-1-yl)azetidin-3-yl)acetonitrile
  • the implementation method is the same as method 2, except that fragment 2-d is replaced with fragment 1-a to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 1-a is used to replace fragment 2-d, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 4-a is used to replace fragment 2-d, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 4-a is replaced with fragment 2-d and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 4-a is replaced with fragment 2-d and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 4, replacing fragment 4-a with fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 4, replacing fragment Amine-1 with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment 5-a is replaced with fragment 2-c and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 25-b is used to replace fragment 2-b and fragment 1-a is used to replace fragment 2-d to obtain the title compound.
  • Step 1 Fragment 25-b 3-cyclopentyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)-1H- Synthesis of pyrazol-1-yl)propionitrile
  • the implementation method is the same as method 2, replacing fragment 2-b with fragment 25-b, and replacing fragment Amine-1 with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 2-b is replaced with fragment 25-b to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 2-b is replaced with fragment 14-a to obtain the title compound.
  • the implementation method is the same as method 2, replacing fragment 2-b with fragment 14-a and replacing fragment 2-d with fragment 4-a to obtain the title compound.
  • the implementation method is the same as method 1, except that fragment 1-b is replaced with fragment 1H-indole-5-amine, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 31-b is used to replace fragment 2-b, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • Step 1 Fragment 31-b 4-fluoro-1-isopropyl-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborane-2 Synthesis of -yl)-1H-benzo[d]imidazole
  • the implementation method is the same as method 4, except that fragment 4-a is replaced with fragment 2-d, fragment 1-b is replaced with fragment (2-aminophenyl)dimethylphosphine oxide, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment (2-aminophenyl)dimethylphosphine oxide, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 4-bromo-2-fluoroaniline is used to replace fragment 1-b, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 4-a is replaced with fragment 2-d and fragment 1-b is replaced with fragment 2-(isopropylsulfonyl)aniline to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment 2-(isopropylsulfonyl)aniline to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 14-a is used to replace fragment 2-b and fragment 1-a is used to replace fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 2-d is replaced with fragment 4-a to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment 2-c is used to replace fragment 5-a to obtain the title compound.
  • the implementation method is the same as method 1, except that fragment Amine-3 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment 5-a is replaced with fragment 1-a and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 25-b is used to replace fragment 2-b, fragment 1-a is used to replace fragment 2-d, and fragment Amine-3 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, replacing fragment 2-b with fragment 14-a, replacing fragment 2-d with fragment 1-a, and replacing fragment Amine-1 with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 1-a replaces fragment 2-d, and fragment Amine-3 replaces fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment 5-a is replaced with fragment 2-c and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 5, except that fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 2, replacing fragment 2-b with fragment 25-b, and replacing fragment Amine-1 with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 14-a is used to replace fragment 2-b, and fragment Amine-3 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, replacing fragment 2-b with fragment 14-a, replacing fragment 2-d with fragment 4-a, and replacing fragment Amine-1 with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 2-d is replaced with fragment 4-a and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment 2-(isopropylsulfonyl)aniline, fragment 4-a is replaced with fragment 2-d, and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 3. Replace fragment 3-a with fragment (3-aminophenyl)carbamate tert-butyl ester, replace fragment 2-d with fragment 1-a, replace fragment Amine-1 with fragment Amine-2, and obtain the title compound.
  • the implementation method is the same as method 3, except that fragment 3-a is replaced with fragment (3-aminophenyl)carbamate tert-butyl ester, and fragment 2-d is replaced with fragment 1-a to obtain the title compound.
  • the implementation method is the same as method 3. Replace fragment 3-a with fragment (3-aminophenyl)carbamate tert-butyl ester, replace fragment 2-d with fragment 1-a, and replace fragment Amine-1 with fragment Amine-3 to obtain the title. compound.
  • the implementation method is the same as method 3.
  • the implementation method is the same as method 3, replacing fragment 3-a with fragment (3-aminophenyl)carbamate tert-butyl ester, replacing fragment 2-d with fragment 1-a, and replacing fragment 3-e with fragment propionyl chloride to obtain the title compound .
  • the implementation method is the same as method three, replacing fragment 3-a with fragment (3-aminophenyl)carbamic acid tert-butyl ester, replacing fragment 2-d with fragment 1-a, replacing fragment 3-e with fragment propionyl chloride, and replacing fragment Amine-1 with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment 2-(isopropylsulfonyl)aniline, fragment 4-a is replaced with fragment 60-b, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • Step 1 Synthesis of fragment 60-b 2,6-dichloro-9-(tetrahydro-2H-pyran-2-yl)-9H-purine
  • the implementation method is the same as method 4, replacing fragment 1-b with fragment N-(2-aminophenyl)methanesulfonamide, replacing fragment 4-a with fragment 2-d, and replacing fragment Amine-1 with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 14-e is used to replace fragment 2-b, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 63-a is used to replace fragment 2-b, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • Step 1 Fragment 63-a 1-(methylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborocyclan-2-yl)-1H -Synthesis of pyrazole
  • the implementation method is the same as method 4, replacing fragment 1-b with fragment methylamine hydrochloride and replacing fragment 4-a with fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment 3-(isopropylsulfonyl)aniline, fragment 4-a is replaced with fragment 2-d, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment (3-aminophenyl)dimethylphosphine oxide, fragment 4-a is replaced with fragment 2-d, and fragment Amine-1 is replaced with fragment Amine-2 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 3-aminobenzenesulfonamide is used to replace fragment 1-b, fragment 2-d is used to replace fragment 4-a, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 68-h is used to replace fragment 1-b, and fragment 2-d is used to replace fragment 4-a to obtain the title compound.
  • Step 1 Synthesis of fragment 68-b(1r,3r)-3-(methylamino)cyclobutan-1-ol
  • Lithium aluminum hydride (812 mg, 21.36 mmol) was added dropwise to a solution of tert-butyl ((1r, 3r)-3-hydroxycyclobutyl)carbamate (1 g, 5.34 mmol) in anhydrous tetrahydrofuran (15 ml) at 0°C. The resulting mixture was stirred at 65°C for 18 hours. TLC monitored the reaction to be complete. The reaction mixture was quenched by adding water (0.8 ml), sodium hydroxide (0.8 ml, 15% aqueous solution) and water (2.4 ml) at 0°C, and the reaction was continued to stir at room temperature for 10 minutes.
  • Step 2 Synthesis of fragment 68-c benzyl ((1r,3r)-3-hydroxycyclobutyl)(methyl)carbamate
  • Step 3 Synthesis of fragment 68-d(1r,3r)-3-(((benzyloxy)carbonyl)(methyl)amino)cyclobutyl 4-methylbenzenesulfonic acid
  • reaction mixture was diluted with dichloromethane (10ml), washed with water (20ml %-15% ethyl acetate (n-hexane elution) was separated and purified to obtain a colorless oily substance (1r, 3r)-3-(((benzyloxy)carbonyl)(methyl)amino)cyclobutyl 4-methyl Benzenesulfonic acid (1.63g, 89%).
  • Step 4 Synthesis of fragment 68-e S-((1s,3s)-3-(((benzyloxy)carbonyl)(methyl)amino)cyclobutyl)ethanethiolate
  • Step 5 Synthesis of fragment 68-f benzyl ((1s,3s)-3-(chlorosulfonyl)cyclobutyl)(methyl)carbamate
  • Acetonitrile (5 ml) containing S-((1s,3s)-3-(((benzyloxy)carbonyl)(methyl)amino)cyclobutyl)ethanethiolate (1g, 3.4mmol) was added to the solution at room temperature.
  • the solution was slowly added dropwise to the stirred N-chlorosuccinimide (983mg, 7.15mmol), acetic acid (0.12ml), water (0.12ml) solution in acetonitrile (5ml), maintaining a stable and slow dropping rate.
  • the temperature of the reaction solution should not exceed 30°C.
  • the resulting mixture was stirred at room temperature for 15 minutes. TLC monitored the reaction to be complete.
  • the reaction mixture was diluted with ethyl acetate (10 ml), washed with saturated sodium bicarbonate solution (20 ml), washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain benzyl ((1s, 3s)-3-(chlorosulfonyl)cyclobutyl)(methyl)carbamate (622 mg, crude product), the crude product was used directly in the next reaction without purification.
  • Step 6 Synthesis of fragment 68-g benzyl ((1s,3s)-3-(azetidin-1-ylsulfonyl)cyclobutyl)(methyl)carbamate
  • reaction mixture was diluted with dichloromethane (15ml), washed with water (10ml %-1.5% methanol (dichloromethane elution) was separated and purified to obtain a colorless oily substance benzyl ((1s,3s)-3-(azetidin-1-ylsulfonyl)cyclobutyl)(methyl) ) carbamate (120 mg, 10% two steps).
  • Step 7 Synthesis of fragment 68-h(1s,3s)-3-(azetidin-1-ylsulfonyl)-N-methylcyclobutan-1-amine
  • Step 1 Synthesis of Fragment 69-b tert-butyl 3-(tosyloxy)azetidine-1-carboxylate
  • Step 2 Fragment 69-c Synthesis of 2-chloro-4-(1H-pyrazol-4-yl)-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine
  • Step 3 Fragment 69-d tert-butyl 3-(4-(2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3 Synthesis of -d]pyrimidin-4-yl)-1H-pyrazol-1-yl)azetidine-1-carboxylic acid
  • the reaction mixture was cooled to Room temperature, partitioned into water (20ml), extracted with ethyl acetate (20ml x 2). The organic layer was collected, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude product was separated and purified by silica gel column chromatography (eluted with dichloromethane containing 2% methanol) to obtain a white solid tert.
  • Step 4 Fragment 69-e tert-butyl 3-(4-(2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7] Dioxazolin-9-yl)amino)-7-(((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine-4- Synthesis of 1H-pyrazol-1-yl)azetidine-1-carboxylic acid
  • TLC monitored the reaction to be complete.
  • the reaction mixture was cooled to room temperature, partitioned into water (20ml), and extracted with ethyl acetate (10ml x 2).
  • the organic layer was collected, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • the crude product was separated and purified by preparative TLC (eluting with n-hexane containing 4% methanol and 38% ethyl acetate).
  • Step 5 Fragment 69-f N-(4-(1-(azetidin-3-yl)-1H-pyrazol-4-yl)-7-((2-(trimethylsilyl) )ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl)-4-methyl-3,4,5,6-tetrahydro-2H-benzo[b] Synthesis of [1,4,7]dioxazoline-9-amine
  • Step 6 Fragment 69-g 4-methyl-N-(4-(1-(1-(1-(methylsulfonyl)azetidin-3-yl)-1H-pyrazole-4- base)-7-(((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl)-3,4,5, Synthesis of 6-tetrahydro-2H-benzo[b][1,4,7]dioxazolin-9-amine
  • Step 7 4-Methyl-N-(4-(1-(1-(methylsulfonyl)azetidin-3-yl)-1H-pyrazol-4-yl)-7H-pyrrolo Synthesis of [2,3-d]pyrimidin-2-yl)-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazoline-9-amine
  • the reaction mixture was cooled to room temperature and partitioned between water (10 ml) and ethyl acetate (10 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (10ml x 2). The organic layers were combined, washed with saturated sodium chloride solution (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 1 Synthesis of 1-(3-(4-(2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1,4,7]dioxazolin-9-yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)azetidin-1-yl)ethanone
  • reaction mixture was concentrated under reduced pressure, and the residue was added to a solution of ammonia water (3ml) and ethyl acetate (2ml). The resulting mixture was stirred and reacted at 30°C for 16 hours. TLC monitored the reaction to be complete. The organic layer was collected and saturated sodium chloride solution (10ml ), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 1 Fragment 71-c tert-butyl ((1s,3s)-3-((3-((5-chloro-2-((4-methyl-3,4,5,6-tetrahydro-2H) -Synthesis of benzo[b][1,4,7]dioxazolin-9-yl)amino)pyrimidin-4-yl)amino)phenyl)carbamoyl)cyclobutyl)carbamate
  • N 4 -(3-aminophenyl)-5-chloro-N 2 -(4-methyl-3,4,5,6-tetrahydro-2H-benzo[b][1 ,4,7]dioxazolin-9-yl)pyrimidine-2,4-diamine (30mg, 0.07mmol), (1s,3s)-3-((tert-butoxycarbonyl)amino)cyclobutanecarboxylic Acid (15 mg, 0.07 mmol), 2-(7-azobenzotris) was added to a solution of N,N-diisopropylethylamine (36 mg, 0.28 mmol) in N,N-dimethylformamide (1 ml).
  • Step 2 (1s,3s)-3-amino-N-(3-((5-chloro-2-((4-methyl-3,4,5,6-tetrahydro-2H-benzo[b Synthesis of ][1,4,7]dioxazolin-9-yl)amino)pyrimidin-4-yl)amino)phenyl)cyclobutanecarboxamide
  • Example 71 The implementation was the same as Example 71, except that fragment 71-b was replaced with (1r,3r)-3-((tert-butoxycarbonyl)amino)cyclobutanecarboxylic acid to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment (2-aminophenyl)dimethylphosphine oxide, and fragment 4-a is replaced with fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 2, using fragment 74-a(R)-3-cyclopentyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaboroheterocycle) Pentan-2-yl)-1H-pyrazol-1-yl)propionitrile (Shanghai Bid Pharmaceutical Technology Co., Ltd.) replaced fragment 2-b, and fragment Amine-2 replaced fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, using fragment 75-a(S)-3-cyclopentyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaboroheterocycle) Pentan-2-yl)-1H-pyrazol-1-yl)propionitrile (Shanghai Bid Pharmaceutical Technology Co., Ltd.) was used to replace fragment 2-b, and fragment 1-a was used to replace fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 74-a is used to replace fragment 2-b to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 2-b is replaced with fragment 75-a to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 6-d is replaced with fragment (2-aminophenyl)dimethylphosphine oxide to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 79-d is used to replace fragment 1-b and fragment 2-d is used to replace fragment 4-a to obtain the title compound.
  • Fragment 79-d is implemented as follows:
  • Step 1 Synthesis of Fragment 79-c 8-(cyclopropanecarbonyl)-3,8-diazabicyclo[3.2.1]octane-3-carboxylic acid tert-butyl ester
  • reaction mixture was partitioned between water (20ml) and ethyl acetate (20ml). Collect the organic layer, wash with saturated sodium chloride solution (10ml Octane-3-carboxylic acid tert-butyl ester (600 mg, crude product), the crude product was directly used in the next reaction without purification.
  • Step 2 Synthesis of fragment 79-d 3,8-diazabicyclo[3.2.1]oct-8-yl(cyclopropyl)methanone
  • the implementation method is the same as method 4.
  • Use fragment 80-f 2-methoxy-3-(1-methyl-1H-1,2,4-triazol-3-yl)aniline to replace fragment 1-b.
  • 2-d was substituted for fragment 4-a to obtain the title compound.
  • Fragment 80-f is implemented as follows:
  • Step 3 Fragment 80-d 3-(2-methoxy-3-nitrophenyl)-1H-1,2,4-triazole
  • N,N-dimethylformamide dimethyl acetal (11.9g, 100mmol) containing 2-methoxy-3-nitrobenzamide (2g, 10mmol) was stirred and reacted at 90°C for 30 minutes. . TLC monitored the reaction to be complete. Concentrate under reduced pressure to remove a large amount of N,N-dimethylformamide dimethyl acetal, dissolve the residue in ethanol (10 ml), and then add dropwise ethanol (6.25 g, 100 mmol) containing hydrazine hydrate (6.25 g, 100 mmol) at 0°C. 50ml) and acetic acid (12ml) mixed solution. The resulting mixture was warmed to room temperature and stirred for 4 hours.
  • Step 4 Fragment 80-e 3-(2-methoxy-3-nitrophenyl)-1-methyl-1H-1,2,4-triazole
  • Step 5 Fragment 80-f 2-methoxy-3-(1-methyl-1H-1,2,4-triazol-3-yl)aniline
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment ethylamine hydrochloride to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment 2-(isopropylsulfonyl)aniline to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment (2-aminophenyl)dimethylphosphine oxide to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment 2-(methylsulfonyl)ethane-1-amine, and fragment 4-a is replaced with fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 4, except that fragment 1-b is replaced with fragment (2-aminoethyl)dimethylphosphine oxide, and fragment 4-a is replaced with fragment 2-d to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment (2-aminoethyl)dimethylphosphine oxide to obtain the title compound.
  • the implementation method is the same as method 6, except that fragment 6-d is used to replace fragment 6-e to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment (S)-oxetan-2-ylmethylamine, and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment 2-(methylsulfonyl)ethane-1-amine, and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 3, replacing fragment 3-a with fragment 1-amino-2-methylpropan-2-ol, and replacing fragment Amine-1 with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 7, replacing fragment 6-d with fragment aniline, and replacing fragment Amine-3 with fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment (S)-oxetan-2-ylmethylamine to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment Amine-1 is used to replace fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 6-d is replaced with fragment 2-(methoxymethyl)aniline, and fragment Amine-3 is replaced with fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 2-methoxyaniline is used to replace fragment 6-d, and fragment Amine-1 is used to replace fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 8, except that fragment 8-b is replaced with fragment 1-(aminomethyl)cyclopropane-1-carbonitrile to obtain the title compound.
  • Step 1 Intermediate 97-a 13-nitro-2,3,5,6,8,9-hexahydro-7H-benzo[b][1,4,7]trioxa[10]azepine Synthesis of tert-butyl cyclododecane-7-carboxylate
  • Step 2 Intermediate 97-b 13-amino-2,3,5,6,8,9-hexahydro-7H-benzo[b][1,4,7]trioxa[10]azeterocycle Synthesis of tert-butyl dodecane-7-carboxylate
  • Step 3 Intermediate 97-c 13-((4-((2-(isopropylsulfonyl)phenyl)amino)-7-((2-(trimethylsilyl)ethoxy)methyl yl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl)amino)-2,3,5,6,8,9-hexahydro-7H-benzo[b][1,4, Synthesis of tert-butyl 7]trioxa[10]azacyclododecane-7-carboxylate
  • TLC monitored the reaction to be complete.
  • the reaction mixture was cooled to room temperature and partitioned between water (20 ml) and ethyl acetate (10 ml).
  • the organic layer was collected and the aqueous phase was extracted with ethyl acetate (10ml x 2).
  • the organic layer was collected, washed with saturated sodium chloride solution (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 4 N 2 -(2,3,6,7,8,9-hexahydro-5H-benzo[b][1,4,7]trioxa[10]azacyclododecane-13 Synthesis of -base-N 4 -(2-(isopropylsulfonyl)phenyl)-7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine
  • Example 97 The implementation is the same as Example 97, except that di-tert-butyl dicarbonate is replaced with methylsulfonyl chloride to obtain the title compound LC_MS: (ES + ): m/z 632.0 [M+H] + .
  • the implementation method is the same as method 1, except that fragment 1-b is replaced with fragment 6-e to obtain the title compound.
  • the implementation method is the same as method 1, replacing fragment 1-a with fragment 2,4-dichloro-5-methylpyrimidine, and replacing fragment 1-b with fragment 6-e to obtain the title compound.
  • the implementation method is the same as method 1, except that fragment 1-a is replaced with fragment 2,4,5-trichloropyrimidine, and fragment 1-b is replaced with fragment 6-e to obtain the title compound.
  • the implementation method is the same as method 1, except that fragment 1-b is replaced with fragment 6-e and fragment Amine-1 is replaced with fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 1, except that fragment 2,4,5-trichloropyrimidine is used to replace fragment 1-a, fragment 6-e is used to replace fragment 1-b, and fragment Amine-3 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 1, replace fragment 1-a with fragment 2,4-dichloro-5-methylpyrimidine, replace fragment 1-b with fragment 6-e, replace fragment Amine-1 with fragment Amine-3, and obtain the title compound.
  • the implementation method is the same as method 1, except that fragment 79-d is used to replace fragment 1-b to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 7-a is replaced with fragment 2,6-dichloro-N-methylpyrimidin-4-amine to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 7-a is replaced with fragment 2,6-dichloro-N-methylpyrimidin-4-amine, and fragment Amine-3 is replaced with fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 7-a is replaced with fragment 2,4-dichloroquinazoline to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 7-a is replaced with fragment 2,4-dichloroquinoline to obtain the title compound.
  • the implementation method is the same as method 3, except that the fragment 3-a is replaced with the fragment (2-amino-5-methylphenyl)carbamate tert-butyl ester, and the fragment Amine-1 is replaced with the fragment Amine-3 to obtain the title compound.
  • the implementation method is the same as method 3, except that fragment 3-a is replaced with fragment (2-amino-5-methylphenyl)carbamate tert-butyl ester, and fragment 2-d is replaced with fragment 7-a to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 6-d is replaced with fragment N-(2-aminophenyl)methanesulfonamide, and fragment Amine-3 is replaced with fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 5-fluoro-2-methoxyphenylboronic acid is used to replace fragment 2-b, fragment 1-a is used to replace fragment 2-d, and fragment Amine-3 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 2, except that fragment 5-fluoro-2-methoxyphenylboronic acid is used to replace fragment 2-b, fragment 1-a is used to replace fragment 2-d, and fragment Amine-2 is used to replace fragment Amine-1 to obtain the title compound.
  • the implementation method is the same as method 7, except that fragment 6-d is replaced with fragment 2-(methoxymethyl)aniline to obtain the title compound.

Landscapes

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

Abstract

本发明提供了式(I)所示的一类大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用。

Description

大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用
本申请要求于2022年9月21日提交中国专利局、申请号为202211155875.X、发明名称为“大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。
技术领域
本发明涉及药物化学领域,具体涉及一类含有大环含氮冠醚化合物及其立体异构体、互变异构体或药学上可接受的盐,它们的制备方法以及含有所述化合物的药物组合物。本发明还涉及该类化合物用作为蛋白激酶抑制剂的用途。
背景技术
蛋白激酶(Protein kinase)是广泛存在于细胞内和细胞表面的酶蛋白,至目前为止被发现和鉴定的蛋白激酶超过500种。它们属于一个结构相关的蛋白家族,其已知成员与几乎所有细胞信号转导活动均有关。蛋白激酶的催化功能是将ATP(或GTP)分子中的γ-磷酸基团转移到靶蛋白上特定的苏氨酸、丝氨酸或酪氨酸基团,使靶蛋白构象发生变化,导致靶蛋白功能由静止转变到活化状态。按照蛋白激酶的靶点氨基酸的特异性,蛋白激酶被分为两大类:丝氨酸/苏氨酸蛋白激酶和酪氨酸蛋白激酶。
蛋白激酶参与的信号传导和调节在细胞和器官的正常功能中起着极为重要的作用,包括细胞生长、分化、增殖、血管生成、细胞凋亡、细胞骨架排列、代谢反应的调节、膜转运和细胞运动等。除此之外,蛋白激酶的非催化功能也起着不可缺少的作用,包括别构效应,亚细胞靶向,蛋白质复合物支架,蛋白质竞争相互作用和DNA结合。但在另一方面,当蛋白激酶发生基因突变或蛋白激酶过度表达时,失调的蛋白激酶会导致多种病理改变,包括癌症,炎症,自身免疫性病,心血管系统和神经系统疾病。因此,蛋白激酶已经成为当今药物开发中最重要靶点之一,近年来蛋白激酶抑制剂在临床治疗上不断取得成功并获得世界各国药监组织的批准,给临床疾病治疗带来重大突破。
本发明具体涉及一类大环含氮冠醚化合物及其作为蛋白激酶,包括但不限于LRRK2、JAK1、JAK2、JAK3、EGFR和CDK9等蛋白激酶的抑制剂的应用。
富亮氨酸重复激酶2(LRRK2)是LRRK2基因的蛋白产物,属于丝氨酸/苏氨酸蛋白激酶。虽然LRRK2蛋白的功能在目前尚不完全清楚,但早在2004年LRRK2基因突变就被发现与帕金森氏病(PD)有关。LRRK2基因突变常见于激酶活性区域的G2019S和I2020T以及GTP酶区域的R1441G/C/H和Y1699C 突变,这些突变均可导致LRRK2蛋白激酶活性升高,引发多种病理改变,包括增加-synuclein和TAU蛋白磷酸化、神经系统炎症以及神经元线粒体功能失调。
PD是仅次于老年痴呆症(Alzheimer’s Disease)之后第二大神经系统退行性疾病,在全球有1000多万患者,但至今尚未发现有效治疗。10%的PD患者有明显家族史,其中LRRK2突变是最常见的原因。该突变发生在约5%的家族性PD以及1%的非家族性PD,这些突变引起的临床表现与散发型PD没有区别。此外LRRK2突变还可以导致TAU蛋白的病理改变,而TAU的病理改变是老年痴呆症的一大特征。因此LRRK2可能位于神经退行性疾病发病机理的上游阶段,对其他神经退行性疾病也起着重要作用。因此开发LRRK2特异性抑制剂成为治疗PD以及其它神经退行性疾病的有效途径之一。
Janus激酶(JAK)是一种非受体型酪氨酸蛋白激酶,其家族含有JAK1、JAK2、JAK3和TYK2四个成员,是转导细胞因子信号从膜受体到STAT转录因子的细胞质酪氨酸激酶。JAK-STAT相关疾病有风湿性关节炎(RA)、哮喘、强直脊柱炎、红斑狼疮、牛皮癣、白癜风等自身免疫疾病。虽然泛JAK抑制剂在治疗炎症和肿瘤性疾病显示较好疗效,但在较高剂量下会产生脱靶效应,从而带来一些较严重副作用,引起不良反应发生。因此仍需要研发新型具有靶向不同JAK亚型的抑制剂,用于治疗自身免疫疾病。比如高选择性JAK1抑制剂正在临床中开展治疗风湿性关节炎(RA)、哮喘、皮炎的研究,可能具有更高安全性。
细胞周期蛋白依赖性激酶(CDK)归属于丝氨酸/苏氨酸激酶,是一族调节细胞周期和基因转录的蛋白激酶。在不同的细胞周期阶段,CDK磷酸化特定的细胞周期蛋白以调控细胞活动。CDK9是最重要的CDK之一,它可与四种细胞周期蛋白合作,包括细胞周期蛋白T1、细胞周期蛋白K、细胞周期蛋白T2a和T2b。CDK9与细胞周期蛋白结合,形成正转录延伸因子b(P-TEFb)的异源二聚体。P-TEFb磷酸化RNA聚合酶II的C端结构域(CTD),使转录从起始位点延伸,是转录延伸的核心分子。CDK9激酶活性失调会导致多种疾病发生,包括高度增殖性疾病(例如癌症)、病毒性传染病或心血管疾病。
鉴于CDK9在疾病中的关键作用,下调CDK9为开发针对癌症和其他疾病的靶向治疗提供了极好的机会。虽然目前已经报道了许多小分子CDK抑制剂,但它们中的大多数缺乏对特定CDK的选择性,因而在临床上表现出低疗效和高不良事件率。因此,开发具有CDK9特异性靶向抑制剂,对于人类疾病治疗至关重要。
现有的蛋白激酶化合物在酶抑制活性上还不能让人满意,或者水溶性不够好,不能够满足药物开发的要求。因此,现有技术中仍迫切需要具有良好的蛋白激酶抑制活性和/或良好的水溶性的化合物。
发明内容
本发明人经研究发现,本发明的含有大环氮冠醚的杂环化合物针对蛋白激酶展现了很好的抑制活性;而且,该类化合物也展示了良好的水溶性。
基于上述研究发现,在第一方面,本发明提供了一种具有式(I)结构的化合物或其立体异构体、互变异构体或药学上可接受的盐:
其中,
Z表示不存在(共价键)或者-OCH2CH2-;
L表示不存在(共价键)、-O-、-NH-或者-N(C1~4烷基)-;
A是任选地被1或2个选自以下的取代基取代的芳基、5~12元杂芳基、C1~10烷基、C3~8环烷基、3~8元杂环基或5~18元桥环基,其中所述取代基选自:氘、C1~4烷基、C1~10烷氧基、C3~6环烷基、3~8元杂环基、C1~4烷基磷酰基、C1~4烷基磺酰基、氨基磺酰基、C1~4烷基氨基磺酰基、氰基C3~6环烷基、C2~4烯基甲酰基、C2~4烯基甲酰氨基、氰基C1~4烷基氨基甲酰基、氰基C1~4烷基、C1~4烷基甲酰基-3~8元杂环基、C3~6环烷基甲酰基、3~8元杂环基-磺酰基、C1~4烷基磺酰基-3~8元杂环基、氨基C3~6环烷基甲酰氨基、C1~4烷基磷酰氨基、C1~4烷基磺酰氨基、氰基、羟基、氧代、巯基、氨基、C2~4烯基和卤素;
R1是氢,或者任选取代的C1~4烷基、C3~6环烷基、C1~4烷基-CO-、-CO或C1~4烷基磺酰基,其中所述取代基选自:氘、C1~4烷基、C1~10烷氧基、C3~6环烷基、氧代、氰基、羟基、氨基、二甲氨基、羟胺基;
R2独立地选自氢、C1~3烷基、三氟甲基、C1~3烷氧基、氰基和卤素;
n是1或2;
Ra选自氢、氨基、三氟甲基、卤素、氰基、C1~3烷基、乙酰基、C1~3烷基磷酰基和C1~3烷基磺酰基;
Rb选自氢、氨基和C1~3烷基氨基;
Ra、Rb和它们所连接的C原子可以一起形成5-6元芳环、5~12元杂芳环或5~8元杂环;
更具体地,在一些实施方案中,所述式(I)化合物具有以下式(II)或式(III)结构:
其中,L、A、R1、R2、Ra、Rb如上述定义。
在另一些实施方案中,所述式I化合物具有以下式(IV)结构:
其中,X、Y各自独立地选自C和N;
Z表示不存在(共价键)或者-OCH2CH2;-
L、A、R1、R2和n如上述定义。
在本发明中,对于上述式(I)化合物或者式(II)-(IV)化合物而言,
A可优选为以下的结构:
本发明中优选以下化合物或其可药用盐:













在本发明的另一个方面,提供了一种药物组合物,其包含本发明所述的式(I)化合物或其立体异构体、互变异构体或药学上可接受的盐,以及任选的可药用载体。在一些实施方案中,所述式(I)化合物具有上述(II)-(IV)结构。在另一些实施方案中,所述式(I)化合物为上述具体的化合物。
在本发明的又一个方面,提供了本发明的上述化合物或其立体异构体、互变异构体或药学上可接受的盐或其药物组合物在制备用作蛋白激酶抑制剂的药物中的应用。
在本发明的又一个方面,提供了本发明的上述化合物或其立体异构体、互变异构体或药学上可接受的盐或其药物组合物在制备用于治疗或预防蛋白激酶相关疾病的药物中的应用。所述蛋白激酶相关疾病是其中的蛋白激酶与疾病发展或症状的信号传递、介导、调节或调整有关的疾病。所述蛋白激酶选自LRRK2、JAK1、JAK2、JAK3、EGFR和CDK9。蛋白激酶相关疾病的实例有下列疾病类型:癌症、自身免疫性疾病、代谢性疾病、炎症、感染(细菌、病毒、酵母菌、 真菌等)、中枢神经系统疾病、退行性神经系统疾病、过敏/哮喘、皮肤病、脉管发生、新血管化、血管生成、心血管病等。
在一些实施方案中,本发明所述的蛋白激酶相关疾病选自神经退行性疾病、自身免疫疾病和肿瘤。在另一些优选实施方案中,本发明所述的蛋白激酶相关疾病选自帕金森病、哮喘、皮炎、非小细胞肺癌、急性髓系白血病(AML)和肝癌。
本文所述化合物、组合物和方法用于治疗或预防疾病或其症状,包括,移植排斥反应(例如,肾、肝、心脏、肺、胰(胰岛细胞)、骨髓、角膜、小肠、皮肤同种移植或异种皮移植),移植体抗宿主性疾病,骨关节炎,类风湿性关节炎,多发性硬化,糖尿病、糖尿病性视网膜病,哮喘,炎性肠道疾病(克罗恩氏病、溃疡性结肠炎),肾脏疾病,恶病质,脓毒性休克,狼疮,糖尿病,重症肌无力,牛皮癣,皮炎,湿疹,皮脂溢,阿尔茨海默氏病,帕金森氏病,化疗中的干细胞保护,自身或异体骨髓移植时的体外选择或体外清洗(purging),白血病(急性骨髓性白血病、慢性骨髓性白血病、急性成淋巴细胞性白血病等),癌(乳癌,肺癌,结肠直肠癌,卵巢癌,前列腺癌,肾癌,鳞状上皮细胞癌,前列腺癌,成胶质细胞瘤,黑素瘤,胰癌,卡波济氏肉瘤等),眼病,视网膜病(例如,黄斑变性、糖尿病性视网膜病),角膜病,青光眼,细菌性感染,病毒性感染,真菌性感染和心脏病,包括但不限于再狭窄。在一个实施方案中,本文所述的组合物和方法用于治疗或预防癌症、眼病,或视网膜病。在另一个实施方案中,本文所述组合物和方法用于治疗或预防类风湿性关节炎、移植排斥反应、哮喘或过敏,或其症状。在其它实施方案中,本文所述组合物和方法用于治疗或预防与高增殖性疾病相关的,或者,与脉管生成相关的疾病或疾病症状。
本文中“任选取代”是指A、R1等所述基团可以被取代基取代,也可以不被取代基取代,即并不仅限于被所列举的取代基取代的情况,也包括不被所列举的取代基取代的情况。这种表达方式与“R为取代或未取代的C1~10烷基、C3~6环烷基或杂环烷基、苯基、萘基或吲哚基,其中取代基为C1~4烷基、C1~4烷氧基、氰基、羟基、疏基、氨基或卤素”的表达方式相同,但是取代或未取代的限定并非仅狭义地指C1~10烷基,而是适用于所有所述的基团。在本文中,提及“任选取代”时,取代基可以是氘、C1~4烷基、C1~10烷氧基、C3~6环烷基、3~8元杂环基、C1~4烷基磷酰基、C1~4烷基磺酰基、氨基磺酰基、C1~4烷基氨基磺酰基、氰基C3~6环烷基、C2~4烯基甲酰基、C2~4烯基甲酰氨基、氰基C1~4烷基氨基甲酰基、 氰基C1~4烷基、C1~4烷基甲酰基-3~8元杂环基、C3~6环烷基甲酰基、3~8元杂环基-磺酰基、C1~4烷基磺酰基-3~8元杂环基、氨基C3~6环烷基甲酰氨基、C1~4烷基磷酰氨基、C1~4烷基磺酰氨基、氰基、羟基、氧代(=O)、巯基、氨基、二甲氨基、羟胺基、C2~4烯基和卤素;其中所述作为取代基的芳基可以被氘、C1~4烷基、C1~10烷氧基、C3~6环烷基、氰基、羟基、巯基、氨基和卤素等取代基进一步取代。
术语“烷基”用于表示直链或支链的饱和烃基,例如,C1~3烷基是指含有1~3个碳原子的饱和烃基,C1~4烷基是指含有1~4个碳原子的饱和烃基,C1~10的烷基是指含有1~10个碳原子的饱和烃基,包括直连、支链或者环状的烷基。
术语“烷氧基”是指烷基-O-。“C1~6烷氧基”意欲包括C1、C2、C3、C4、C5、C6烷氧基。烷氧基的实例包括但不限于甲氧基、乙氧基、丙氧基(例如正丙氧基和异丙氧基)和叔丁氧基。类似地,“C1~10烷氧基”是指-O-烷基基团,烷基包括直链、支链和环状烷基,其碳原子数为1~10个。烷氧基的例子包括甲氧基,乙氧基,丙氧基(如,正丙氧基、异丙氧基和环丙氧基),叔丁氧基等。本文中优选的烷氧基是C1~6烷氧基、C1~4烷氧基。
术语“环烷基”指非芳族碳环基,包括环化的烷基。环烷基可以包括单环、二环或多环系统。环烷基的例子包括环丙基、环丁基、环戊基、环己基、环庚基等,C3~8环烷基是指含有3~8个碳原子的环烷基。本文中优选的环烷基是C3~8环烷基、C3~6环烷基。
术语“杂环烷基”或“杂环基”是指非芳族杂环烷基,其中1个或2个或3个成环碳原子被杂原子例如O、N或S原子取代。杂环烷基优选具有3,4,5,6或7个成环原子。本文中优选的杂环基是C3-8杂环基。
“芳基”是指芳族碳环基,包括单环、二环、三环或多环芳烃,例如苯基,萘基,蒽基,菲基等。芳基优选具有6至14个或6至12个环成员的单环、二环或三环的环系统,其中所述系统中的至少一个环为芳族的且其中所述系统中的每个环含有3至7个环成员。“取代的芳基”是指在芳基的苯环上至少一个氢原子被非氢部分取代,芳基的取代基可以是卤素、C1~10烷氧基、-CN、-OH、-SH、-NH2、C1~6烷基。优选的芳基包括苯基、联苯基、茚满基、1-萘基、2-萘基和四氢萘基。术语“芳烷基”或“芳基烷基”是指连接至芳基环的烷基残基。非限制性实例包括苄基、苯乙基等。稠合的芳基可在环烷基环或芳族环的合适位置上连接至另一基团。
术语“杂芳基”意指稳定的5-12元的芳香单环或芳香二环或芳香多环杂环,其为完全不饱和的、部分不饱和的,且其含有碳原子和1个、2个、3个或4个独立地选自N、O和S的杂原子。其包括5元、6元或7元的芳香单环或8元、9元、10元、11元、12元的芳香二环或芳香多环杂环;优选上文所定义的任意杂环与苯环稠合。氮和硫杂原子可任选地被氧化。氮原子为取代的或未取代的(即N或NR,其中R为H或如果被定义,则为另一取代基)。杂环可在得到稳定结构的任何杂原子或碳原子处连接至其侧基。如果所得化合物是稳定的,则本文所述的杂环基可在碳或氮原子上被取代。杂环中的氮可任选地被季铵化。优选地,当杂环中S和O原子的总数超过1时,则这些杂原子彼此不相邻。芳杂基的实施例包括但不限于吖啶基、氮杂环丁基、吖辛因基、苯并咪唑基、苯并呋喃基、苯并硫代呋喃基、苯并噻吩基、苯并噁唑基、苯并噁唑啉基、苯并噻唑基、苯并三唑基、苯并四唑基、苯并异噁唑基、苯并异噻唑基、苯并咪唑啉基、咔唑基、4aH-咔唑基、咔啉基、色满基、色烯基、喹啉基、十氢喹啉基、二氢呋喃并[2,3-b]四氢呋喃基、呋喃基、呋咱基、咪唑烷基、咪唑啉基、咪唑基、1H-吲唑基、咪唑并吡啶基、二氢吲哚基、吲嗪基、吲哚基、3H-吲哚基、异苯并呋喃基、异色满基、异吲唑基、异二氢吲哚基、异吲哚基、异喹啉基、异噻唑基、异噻唑并吡啶基、异噁唑基、异噁唑并吡啶基、亚甲基二氧基苯基、吗啉基、二氮杂萘基、八氢异喹啉基、噁二唑基、1,2,3-噁二唑基、1,2,4-噁二唑基、1,2,5-噁二唑基、1,3,4-噁二唑基、噁唑烷基、噁唑基、噁唑并吡啶基、噁唑烷基、萘嵌间二氮杂苯基、羟吲哚基、嘧啶基、菲啶基、菲咯啉基、吩嗪基、吩噻嗪基、吩噁噻基、吩噁嗪基、酞嗪基、哌嗪基、哌啶基、哌啶酮基、4-哌啶酮基、胡椒基、喋啶基、嘌呤基、吡喃基、吡嗪基、吡唑烷基、吡唑啉基、吡唑并吡啶基、吡唑基、哒嗪基、吡啶并噁唑基、吡啶并咪唑基、吡啶并噻唑基、吡啶基、嘧啶基、吡咯烷基、吡咯啉基、2-吡咯烷酮基、2H-吡咯基、吡咯基、喹唑啉基、喹啉基、4H-喹嗪基、喹喔啉基、奎宁环基、四唑基、四氢呋喃基、四氢异喹啉基、四氢喹啉基、6H-1,2,5-噻二嗪基、1,2,3-噻二唑基、1,2,4-噻二唑基、1,2,5-噻二唑基、1,3,4-噻二唑基、噻蒽基、噻唑基、噻吩基、噻唑并吡啶基、噻吩并噻唑基、噻吩并噁唑基、噻吩并咪唑基、噻吩基、三嗪基、1,2,3-三唑基、1,2,4-三唑基、1,2,5-三唑基、1,3,4-三唑基和呫吨基、喹啉基、异喹啉基、酞嗪基、喹唑啉基、吲哚基、1H-吲唑基、苯并咪唑基、1,2,3,4-四氢喹啉基、1,2,3,4-四氢异喹啉基、5,6,7,8-四氢-喹啉基、2,3-二氢-苯并呋喃基、1,2,3,4-四氢-喹喔啉基和1,2,3,4-四氢-喹唑啉基。术语“杂芳基”还可以包括由上述所定义的“芳基”与单环“杂芳基”所形成的联芳基 结构,例如但不限于“-苯基联吡啶基-”、“-苯基联嘧啶基”、“-吡啶基联苯基”、“-吡啶基联嘧啶基-”、“-嘧啶基联苯基-”。
术语“C1~4烷基磷酰基”是指C1~4烷基取代的磷酰基,磷酰基上的两个烷基可以相同也可以不同。
术语“C1~4烷基磺酰基”是指C1~4烷基取代的磺酰基。
术语“卤素”包括氟、氯、溴和碘。
“桥环基”指5至18元、优选5至14元,含有两个或两个以上环状结构,彼此共用两个不直接相连接的碳原子的多环基团。更优选为6至14元,还更优选为7至10元桥环基。根据组成环的数目可以分为双环、三环、吡啶酮或多环桥杂环基,优选为双环、三环或吡啶酮,更优选为双环或三环。本文中的桥环基允许其中1个、2个或3个环原子为选自氮、氧或S(O)n(其中n选自0、1和2)的杂原子。
在本文中,如未特别指明,所述基团或取代基均按习惯从左至右的顺序,在其最右端与母核相连接。例如,对于氰基C1~4烷基氨基甲酰基而言,其作为取代基与化合物母核相连的位置是最右侧的“甲酰基”。
本文中所用术语“药学上可接受的盐”或“可药用盐”是指使本发明化合物能保持原有生物活性并且适合于医药用途的某些盐类。式(I)所表示的化合物的可药用的盐可以是通过羧基或胺基(主要是胺基)与合适的碱或酸所形成的盐,例如与合适的碱形成的盐(包括金属盐、铵盐)、或者与合适的酸形成的盐。本发明的化合物优选与合适的酸形成盐,所述酸可选自:盐酸、氢溴酸、氢氟酸、硫酸、磷酸、硝酸、甲酸、乙酸、丙酸、草酸、丙二酸、琥珀酸、富马酸、马来酸、乳酸、苹果酸、酒石酸、柠檬酸、苦味酸、甲磺酸、乙磺酸、甲苯磺酸、苯磺酸、萘磺酸、三氟乙酸和天冬氨酸,优选为甲磺酸或盐酸。
本文使用的术语“治疗”包括导致改善病症、疾病、障碍等的任何效果,例如减轻、减少、调节、改善或消除,或改善其症状。
本文使用的短语“可药用载体”意指药用物质、组合物或媒介物,诸如液体或固体填充剂、稀释剂、赋形剂、制造助剂(例如润滑剂、滑石、硬脂酸镁、硬脂酸钙或硬脂酸锌或硬脂酸)或溶剂包囊物质,其涉及将主题化合物从一个器官或身体的部分携带或运送至另一个器官或身体的部分。每种载体在与制剂的其它成分相容和对患者无害的意义上必须是“可接受的”。
术语“药物组合物”意指包含本发明化合物与至少一种其它药用载体的组合物。“药用载体”是指本领域中通常接受用于将生物活性剂递送至动物(具体为哺乳动物)的介质,包括(即)佐剂、赋形剂或媒介物,诸如稀释剂、防腐剂、填充剂、流动调控剂、崩解剂、润湿剂、乳化剂、悬浮剂、增甜剂、矫味剂、芳香剂、抗细菌剂、抗真菌剂、润滑剂和分散剂,这取决于给药模式和剂型的性质。
术语:“Brettphos Pd G3”是指甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II);“TLC”是指薄层色谱;
为更好地阐述本发明所采取的技术手段及其效果,以下结合非限制性实施例来进一步说明本发明。本发明的实施例,包括实施例中提供的描述,旨在说明本发明的实施方式,其并非旨在限制任何权利要求的范围。根据本发明,本领域技术人员将理解,在不脱离本发明的精神和范围的情况下,可以对所公开的具体实施方案进行许多改变并仍然能获得相同或相似的结果。
除非另有说明,所有材料/试剂均从商业供应商处获得,无需进一步纯化即可使用。下述实施例的化合物结构是通过核磁共振(NMR)和/或液相色谱质谱联用(LC_MS)来表征和确定的。
1H NMR波谱通过Bruker Avance 400MHz波谱仪上在室温下记录,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD)或氘水(D2O)。化学位移值(δ)以ppm为单位,用四甲基硅烷(TMS)或残留溶剂峰作为内标,耦合常数(J)以赫兹(Hz)为单位,1H NMR谱图中峰型的多重性缩写如下:s(单峰)、d(双重峰)、t(三重峰)、q(四重峰)、qn(五重峰)、m(多重峰)、br(宽峰)。
液相色谱质谱联用(LC_MS)使用的仪器是Shimadzu LCMS-2020,制备型高效液相色谱(Prep-HPLC)使用的仪器是Bonna-Agela FLEXA FL-H100G。薄层色谱(TLC)所使用的薄层层析硅胶板型号是烟台黄海HSGF254薄层层析硅胶板,反应监测所用的规格为2.5×8cm,其涂层厚度为0.2±0.03mm,分离纯化所用的规格为20×20cm,其涂层厚度为0.4–0.5mm。硅胶柱层析色谱法使用的是青岛海洋硅胶100–200目或200–300目硅胶为载体。
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。
合成实施例:
片段Amine-1
实施方式:
步骤1:中间体Amine-1-b(((4-硝基苯基)磺酰基)氮杂二基)双(乙烷-2,1-二基)双(4-硝基苯磺酸酯)的合成
0℃下向含有二乙醇胺(1.0g,9.51mmol)和三乙胺(3.4g,33.28mmol)的四氢呋喃(5ml)溶液中,缓慢滴加含有4-硝基苯磺酰氯(7.0g,31.38mmol)的四氢呋喃(10ml)溶液。所得混合物升至室温搅拌16小时。TLC监测反应完全。浓缩反应液,剩余物在甲醇(30ml)中重结晶纯化得到白色固体(((4-硝基苯基)磺酰基)氨二基)双(乙-2,1-二醇)双(4-硝基苯磺酸酯)(5.2g,83%)。
1H NMR(400MHz,DMSO-d6):δ8.46(d,J=8.8Hz,4H),8.34(d,J=8.8Hz,2H),8.13(d,J=8.8Hz,4H),8.04(d,J=8.8Hz,2H),4.19(t,J=5.2Hz,4H),3.50(t,J=4.8Hz,4H).
步骤2:中间体Amine-1-c 9-硝基-4-((4-硝基苯基)磺酰基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉的合成
将含有(((4-硝基苯基)磺酰基)氮杂二基)双(乙烷-2,1-二基)双(4-硝基苯磺酸酯)(5.2g,7.9mmol),4-硝基儿茶酚(1.1g,7.2mmol),碳酸钾(2.1g,15.1mmol)和碘化钾(112mg,0.7mmol)的N,N-二甲基甲酰胺(20ml)溶液在70℃反应12小时。TLC监测反应完全。混合物分配于乙酸乙酯(150ml)和水(200ml)中。收集有机相,饱和氯化钠溶液(100ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(33%二氯甲烷的正己烷洗脱溶液)纯化得到白色固体9-硝基-4-((4-硝基苯基)磺酰基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉(556mg,15%)。
1H NMR(400MHz,DMSO-d6):δ8.35(d,J=8.8Hz,2H),8.01-8.04(m,2H),7.91(d,J=2.8Hz,1H),7.84-7.87(m,1H),7.14(d,J=9.2Hz,1H),4.67(t,J=4.6Hz,2H),4.41(t,J=4.6Hz,2H),3.52-3.56(m,4H).
步骤3:中间体Amine-1-d 9-硝基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉合成
向含有9-硝基-4-((4-硝基苯基)磺酰基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉(556.0mg,1.36mmol)的N,N-二甲基甲酰胺(10ml)溶液中,加入4-甲基硫酚(203.0mg,1.63mmol)和碳酸钾(564.0mg,4.08mmol)。所得混合物在室温下搅拌14小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(100ml)和水(100ml)中。收集有机相,饱和氯化钠溶液(100ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用20%-33%乙酸乙酯的正己烷溶液洗脱)纯化得到黄色固体9-硝基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉(267mg,87%)。
LC_MS:(ES+):m/z 225.15[M+H]+.
1H NMR(400MHz,CDCl3):δ7.74-7.77(m,1H),7.63(d,J=2.8Hz,1H),6.62(d,J=8.8Hz,1H),4.24(t,J=4.4Hz,2H),3.90-3.94(m,2H),3.57-3.62(m,4H),3.57-3.62(m,4H).
步骤4:中间体Amine-1-e 4-甲基-9-硝基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉的合成
0℃下向含有9-硝基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉(600mg,2.68mmol),N,N-二甲基甲酰胺(5ml)溶液中加入氢化钠(60%,118mg,2.95mmol)和碘甲烷(457mg,3.22mmol)。所得混合物在0℃下反应30分钟,然后升至室温反应3小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(50ml)和水(50ml)中。收集有机层,饱和氯化钠溶液(30ml x 2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用15%-20%乙酸乙酯的正己烷溶液洗脱)纯化得到黄色固体4-甲基-9-硝基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉(583mg,91%)。
LC_MS:(ES+):m/z 239.30[M+H]+.
1H NMR(400MHz,CDCl3):δ7.78-7.81(m,1H),7.67(d,J=2.4Hz,1H),6.59(d,J=8.8Hz,1H),4.21(t,J=4.4Hz,2H),3.56-3.63(m,6H),3.36(s,3H).
步骤5:中间体Amine-1 4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
将含有4-甲基-9-硝基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉(583mg,2.45mmol)和Pd/C(10%,60mg)的甲醇(10ml)和乙酸乙酯(10ml)溶液在室温氢气氛围(氢气球)下反应2小时。TLC监测反应完全。Pd/C通过过滤移除,滤渣用甲醇(10ml x 2)洗,合并的滤液减压浓缩,得到紫色油状物粗品4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(497mg,97%),无需进一步纯化即可用于下一步。
LC_MS:(ES+):m/z 208.85[M+H]+.
1H NMR(400MHz,CDCl3):δ6.53-6.56(m,1H),6.23-6.25(m,2H),4.19(t,J=4.4Hz,2H),3.57(t,J=5.8Hz,2H),3.30-3.36(m,9H).
片段Amine-2
实施方式:
实施方式:
步骤1:中间体Amine-2-b 2-(2-羟基乙氧基)-5-硝基苯酚的合成
0℃下向含有金属钠(551mg,24mmol)的乙二醇(10ml)溶液中加入5-硝基苯并[d][1,3]二恶唑(2g,12mmol)。所得混合物在120℃下搅拌3小时。TLC监测反应完全。反应混合物冷至室温,用2N盐酸酸化至pH=3~4,过滤悬浮物,收集固体,水洗后减压干燥得到棕色固体2-(2-羟基乙氧基)-5-硝基苯酚(2.1g,88%)。
LC_MS:(ES+):m/z 199.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.86(s,1H),7.75-7.72(m,1H),7.62-7.61(m,1H),7.14-7.12(m,1H),4.90(s,1H),4.13-4.11(m,2H),3.78-3.76(m,2H).
步骤2:中间体Amine-2-c 2-(2-(2-(2-羟基乙氧基)-5-硝基苯氧基)乙氧基)乙醇的合成
将含有2-(2-羟基乙氧基)-5-硝基苯酚(2.15g,10.75mmol),2-(2-氯乙氧基)乙烷-1-醇(2g,16.13mmol),碘化钾(1.8g,10.75mmol),碳酸钾(4.45g,32.25mmol)的N,N-二甲基甲酰胺(20ml)溶液在70℃下搅拌反应2天。TLC监测反应完全。反应混合物冷至室温,分配于水(200ml)和乙酸乙酯(100ml)中。收集有机层,水相用乙酸乙酯(50ml)萃取。合并有机层,饱和氯化钠溶液(100ml)洗涤,无水硫酸钠干燥,减压浓缩得到橘色油状物2-(2-(2-(2-羟基乙氧基)-5-硝基苯氧基)乙氧基)乙醇(2.1g,67%),无需进一步纯化即可用于下一步。
LC_MS:(ES+):m/z 288.2[M+H]+.
1HNMR(400MHz,CDCl3):δ7.95-7.92(m,1H),7.77-7.76(m,1H),6.94-6.92(m,1H),4.24-4.22(m,2H),4.16-4.14(m,2H),4.04-4.01(m,2H),3.96-4.00(m,2H),3.84-3.76(m,4H).
步骤3:中间体Amine-2-d 4-(4-硝基-2-(2-(2-(甲苯磺酰氧基)乙氧基)乙氧基)苯氧基)4-甲基苯磺酸乙酯的合成
0℃下向含有2-(2-(2-(2-羟基乙氧基)-5-硝基苯氧基)乙氧基)乙醇(2.1g,7.3mmol)和三乙胺(3.3g,21.9mmol)的二氯甲烷溶液中加入4-甲苯磺酰氯(3.5g,18.25mmol)。所得混合物在室温下搅拌反应14小时。TLC监测反应完全。反应 混合物分配于二氯甲烷(10ml)和水(20ml)中。收集有机层,饱和氯化钠溶液(20ml x 2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析分离纯化(洗脱液:50%乙酸乙酯的石油醚溶液至1.6%乙酸乙酯的二氯甲烷溶液)得到棕色油状物4-(4-硝基-2-(2-(2-(甲苯磺酰氧基)乙氧基)乙氧基)苯氧基)4-甲基苯磺酸乙酯(3.5g,80%)。
LC_MS:(ES+):m/z 596.1[M+H]+.
1H NMR(400MHz,CDCl3):δ7.86-7.72(m,6H),7.34-7.29(m,4H),6.86-6.84(m,1H),4.42-4.39(m,2H),4.31-4.29(m,2H),4.20-4.18(m,2H),4.15-4.13(m,2H),3.85-3.79(m,4H),2.44-2.42(d,J=8.4Hz,6H).
步骤4:中间体Amine-2-e 7-(4-甲氧基苄基)-13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷的合成
将含有4-甲基苯磺酸2-(4-硝基-2-(2-(2-(甲苯磺酰基氧基乙氧基)乙氧基)苯氧基)乙基(3.5g,5.9mmol),(4-甲氧基苯基)甲胺(806mg,5.9mmol),碘化钾(488mg,2.94mmol)和碳酸钾(4.05g,29.38mmol)的乙腈(100ml)溶液回流反应16小时。TLC监测反应完全。反应混合物冷至室温,分配于水(100ml)和乙酸乙酯(50ml)中。收集有机层,水层用乙酸乙酯(50ml x 2)萃取。合并有机层,饱和氯化钠溶液(80ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析分离纯化(洗脱液:33%乙酸乙酯的石油醚溶液)得到黄色固体7-(4-甲氧基苄基)-13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(1.6g,70%)。
LC_MS:(ES+):m/z 389.5[M+H]+.
步骤5:中间体Amine-2-f 13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷的合成
将含有7-(4-甲氧基苄基)-13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(1.6g,4.12mmol)和1-氯乙基氯甲酸酯(883mg,6.18mmol)的1,2-二氯乙烷(30ml)溶液在室温下搅拌反应13小时。TLC监测反应完全。反应混合物浓缩,剩余物溶于甲醇(8ml)。所得混合物回流反应1小时。TLC监测反应完全。反应混合物分配于水(20ml)和二氯甲烷(30ml)。收集有机层,水层用二氯甲烷(10ml x 5)萃取。合并有机层,饱和氯化钠溶液(30ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用5%-10%的甲醇的二氯甲烷溶液洗脱)纯化得到黄色固体13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂十二烷基(620mg,56%)。
LC_MS:(ES+):m/z 269.2[M+H]+.
1H NMR(400MHz,CDCl3):δ7.93-7.91(m,1H),7.86-7.85(m,1H),7.12-7.10(d,J=8.8Hz,1H),4.31-4.28(m,4H),3.81-3.79(m,2H),3.72-3.69(m,2H),3.00-2.98(m,2H),2.82-2.79(m,2H).
步骤6:中间体Amine-2-g 7-甲基-13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷的合成
将含有13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂十二烷基(515mg,1.9mmol),三乙胺(567mg,3.8mmol)和碘甲烷(409mg,2.88mmol)的N,N-二甲基甲酰胺溶液70℃下在密封管中反应三小时。TLC监测反应完全。反应混合物冷至室温,分配于水(50ml)和乙酸乙酯(30ml)中。收集有机层,水层用乙酸乙酯(20ml x 2)萃取。合并有机层,饱和氯化钠溶液(30ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用5%-10%的甲醇的二氯甲烷溶液洗脱)纯化得到黄色固体7-甲基-13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(267mg,49%)。
LC_MS:(ES+):m/z 283.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ7.96-7.92(m,2H),7.28-7.26(d,J=8.8Hz,1H),4.34-4.25(m,4H),3.71-3.70(m,4H),3.05-2.84(m,4H),2.48-2.40(m,3H).
步骤7:Amine-2 7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺的合成
室温下向含有7-甲基-13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(267mg,0.95mmol)的二氯甲烷(3ml)溶液中加入锌粉(870mg,13.3mmol)和醋酸(1.6g,26.6mmol)。反应混合物在室温氮气氛围下搅拌10分钟。TLC监测反应完全。反应混合物用饱和碳酸氢钠溶液碱化至pH=7-8,过滤。滤液分配于10%甲醇的二氯甲烷溶液(20ml)和水(10ml)中。收集有机层,水层用10%甲醇的二氯甲烷(20ml x 3)溶液萃取。合并有机层,饱和氯化钠溶液(30ml)洗涤,无水硫酸钠干燥,减压浓缩得到黄色油状粗品7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺(230mg,96%)。
LC_MS:(ES+):m/z 252.8[M+H]+.
片段Amine-3
实施方式:
步骤1:中间体Amine-3-b 2-(烯丙氧基)-1-氯-4-硝基苯的合成
将2-氯-5-硝基苯酚(5.0g,28.8mmol),3-溴丙烯(3.5g,28.8mmol)和碳酸钾(6g,43.2mmol)加入到反应瓶中,加入20ml N,N-二甲基甲酰胺,升温至70℃反应12小时。LTC监测反应完全,反应液冷却至室温,加水(200ml)和乙酸乙酯(100ml)萃取,水相再加乙酸乙酯(50ml)萃取,合并的有机相用饱和氯化钠溶液(100ml)洗涤,无水硫酸钠干燥,浓缩。粗品经重结晶得到褐色固体2-(烯丙氧基)-1-氯-4-硝基苯(5.5g,90%)。
1H NMR(400MHz,CDCl3):δ7.78-7.82(m,2H),7.53(d,J=8.4Hz,1H),6.03-6.12(m,1H),5.49-5.54(m,1H),5.37-5.40(m,1H),4.71-4.73(m,2H).
步骤2:中间体Amine-3-c 2-(2-(2-(烯丙氧基)-4-硝基苯氧基)乙氧基)乙-1-醇的合成
0℃下将二乙二醇(10g,93.6mmol)加入含有10ml N,N-二甲基甲酰胺反应瓶中,加入氢化钠(60%,562mg,14mmol)。反应液升至室温反应1小时,然后冷却至0℃,滴加2-(烯丙氧基)-1-氯-4-硝基苯(2g,9.36mmol),反应液在氮气保护50℃下搅拌反应2天。TLC监测反应完全,反应液加乙酸乙酯(100ml)和水(100ml)萃取,有机相分别用的饱和氯化钠溶液(100ml)洗涤2次,无水硫酸钠干燥,浓缩,粗品经柱层析分离纯化(洗脱液:20%-50%乙酸乙酯的正己烷溶液)得到褐色油状物2-(2-(2-(烯丙氧基)-4-硝基苯氧基)乙氧基)乙-1-醇(2g,75%)。
1H NMR(400MHz,CDCl3):δ7.88-7.91(m,1H),7.76(d,J=2.8Hz,1H),6.94(d,J=8.8Hz,1H),6.04-6.13(m,1H),5.44-5.49(m,1H),5.33-5.36(m,1H),4.66-4.68(m,2H),4.27(t,J=4.6Hz,2H),3.96(t,J=4.8Hz,2H),3.69-3.78(m,4H),2.28(s,1H).
步骤3:中间体Amine-3-d 2-(2-(2-羟基-4-硝基苯氧基)乙氧基)乙酸乙酯的合成
在室温氮气保护下,向含有2-(2-(2-(烯丙氧基)-4-硝基苯氧基)乙氧基)乙-1-醇(1g,3.53mmol)的醋酸(5ml)悬浮液中加入四(三苯基膦)钯(408mg,0.35mmol),所得混合物氮气置换3次,升温至120℃搅拌反应2小时。TLC监测反应完全。反应混合物减压浓缩,分配于水(20ml)和乙酸乙酯(10ml)中。收集有机层,水相用乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,得到棕色油状粗品2-(2-(2-羟基-4-硝基苯氧基)乙氧基)乙酸乙酯(860mg,粗品),无需进一步纯化即可用于下一步。
LC_MS:(ES+):m/z 286.10[M+H]+.
1H NMR(400MHz,CDCl3):δ7.77-7.80(m,2H),6.92(d,J=9.6Hz,1H),6.86(s,1H),4.26-4.33(m,4H),3.88-3.90(m,2H),3.76-3.78(m,2H),2.11(s,3H).
步骤4:中间体Amine-3-e 2-(2-(2-(2-羟基乙氧基)-4-硝基苯氧基)乙氧基)乙-1-醇的合成
将含有2-(2-(2-羟基-4-硝基苯氧基)乙氧基)乙酸乙酯(860mg,3mmol),2-溴乙醇(1.5g,12mmol)和碳酸钾(3.7g,27.1mmol)的N,N-二甲基甲酰胺(10ml)溶液在70℃下搅拌反应14小时。TLC监测反应完全。反应混合物冷却至室温,分配于水(100ml)和乙酸乙酯(50ml)中。收集有机层,水相用乙酸乙酯(50mlx2)萃取。合并有机层,饱和氯化钠溶液(100ml)洗涤,无水硫酸钠干燥,减压浓缩;剩余物溶于甲醇(5ml)中,室温下加入碳酸钾(416mg,3mmol),所得混合物室温搅拌反应30分钟。TLC监测反应完全。反应混合物分配于二氯甲烷(30ml)和水(20ml)中。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用2-5%甲醇的二氯甲烷溶液洗脱)分离纯化得到淡黄色油状物2-(2-(2-(2-羟乙氧基)-4-硝基苯氧基)乙氧基)乙醇(500mg,57%)。
1H NMR(400MHz,CDCl3):δ7.91-7.94(m,1H),7.78(d,J=2.4Hz,1H),6.92(d,J=8.8Hz,1H),4.75(s,1H),4.24(t,J=4.2Hz,2H),4.15(t,J=4.2Hz,2H),4.02(t,J=4.2Hz,2H),3.96(s,2H),3.75-3.82(m,4H),1.74(s,1H).
步骤5:中间体Amine-3-f 2-(5-硝基-2-(2-(2-(甲苯磺酰氧基)乙氧基)乙氧基)苯氧基)乙基-4-甲苯磺酸酯的合成
0℃下向含有2-(2-(2-(2-羟乙氧基)-4-硝基苯氧基)乙氧基)乙-1-醇(500mg,1.74mmol),三乙胺(528mg,5.22mmol)和4-二甲氨基吡啶(21mg,0.17mmol)的二氯甲烷(5ml)溶液中加入对甲苯磺酰氯(730mg,3.83mmol)。所得混合物升至室温搅拌反应2小时,TLC监测反应完全。反应混合物分配于二氯甲烷(20ml)和水(20ml)中。收集有机相,饱和氯化钠溶液(10ml x 2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用50-100%乙酸乙酯的己烷溶液洗脱)分离纯化 得到褐色油状物2-(5-硝基-2-(2-(2-(甲苯磺酰氧基)乙氧基)乙氧基)苯氧基)乙基-4-甲苯磺酸酯(830mg,80%)。
1H NMR(400MHz,CDCl3):δ7.88-7.91(m,1H),7.76-7.81(m,4H),7.64(d,J=2.8Hz,1H),7.30-7.35(m,4H),6.91(d,J=9.2Hz,1H),4.19(t,J=4.6Hz,4H),4.38-4.40(m,2H),4.23-4.26(m,2H),3.86(t,J=4.6Hz,2H),3.81(t,J=4.6Hz,2H),2.43(d,J=8.0Hz,6H).
步骤6:中间体Amine-3-g 7-(4-甲氧基苄基)-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷的合成
将含有2-(5-硝基-2-(2-(2-(甲苯磺酰氧基)乙氧基)乙氧基)苯氧基)乙基-4-甲苯磺酸酯(630mg,1.06mmol),4-甲氧基苄胺(145mg,1.06mmol),碳酸钾(731mg,5.3mmol)和碘化钾(88mg,0.53mmol)的乙腈(30ml)溶液回流反应14小时。TLC监测反应完全。反应混合物冷却至室温,分配于水(30mL)和乙酸乙酯(10ml)中。收集有机层,水相用乙酸乙酯(10ml x 2)萃取。合并有机相,饱和氯化钠溶液(30ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用20%-50%乙酸乙酯的己烷溶液洗脱)分离纯化得到黄色油状物7-(4-甲氧苄基)-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(210mg,51%)。
1H NMR(400MHz,CDCl3):δ7.90-7.93(m,1H),7.75(d,J=2.0Hz,1H),7.13(s,2H),7.00(d,J=8.8Hz,1H),6.78(d,J=7.2Hz,2H),4.31(t,J=4.0Hz,2H),4.17(s,2H),3.87(t,J=3.8Hz,2H),3.78(s,5H),3.64(s,2H),2.87(d,J=6.8Hz,4H).
步骤7:中间体Amine-3-h 12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂十二烷基的合成
将含有7-(4-甲氧苄基)-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(1.08g,2.78mmol)和1-氯乙基氯甲酸酯(596mg,4.17mmol)的1,2-二氯乙烷(15ml)溶液室温下搅拌反应3小时。TLC监测反应完全。浓缩反应混合物,剩余物中加入甲醇(3ml),所得混合物加热回流1小时。TLC监测完全反应完。反应混合物分配于水(20ml)和二氯甲烷(30ml)中。收集有机层,水相用二氯甲烷(10mlx2)萃取。合并有机相层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用2%-5%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(479mg,64%)。
1H NMR(400MHz,DMSO-d6):δ8.03(t,J=5.4Hz,2H),7.44(s,1H),7.35(d,J=8.8Hz,1H),4.30-4.35(m,4H),3.81(t,J=4.0Hz,2H),3.70(t,J=4.6Hz,2H),3.05-3.12(m.4H).
步骤8:中间体Amine-3-i 7-甲基-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷的合成
0℃下向含有12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(265mg,0.98mmol)和碘甲烷(167mg,1.18mmol)的N,N-二甲基甲酰胺(5ml)中,加入氢化钠(60%,43mg,1.08mmol)。反应混合物在0℃下搅拌反应30分钟,然后升至室温继续搅拌反应3小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(30ml)和水(50ml)中。收集有机层,水相用乙酸乙酯(20ml x 4)萃取。合并有机层,饱和氯化钠溶液(50ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用2%-10%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色油状物7-甲基-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(153mg,56%)。
LC_MS:(ES+):m/z 283.20[M+H]+.
1H NMR(400MHz,CDCl3):δ7.89-7.91(m,1H),7.85(d,J=2.4Hz,1H),6.99(d,J=8.8Hz,1H),4.28(t,J=4.2Hz,2H),4.21(t,J=4.8Hz,2H),3.88(t,J=4.2Hz,2H),3.74(t,J=5Hz,2H),2.82(t,J=4.6Hz,2H),2.68(t,J=4.8Hz,2H),2.37(s,3H).
步骤9:Amine-3 7-甲基-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺的合成
室温下向含有7-甲基-12-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂十二烷(153mg,0.63mmol)的二氯甲烷(7ml)溶液加入锌粉(574mg,8.78mmol)和醋酸(1.05g,17.55mmol)。所得混合物在室温氮气保护下搅拌反应30分钟。TLC监测反应完全。反应混合物过滤,滤液分配于二氯甲烷(20ml)和水(10ml)中。收集有机层,水相用10%甲醇的二氯甲烷溶液(10ml x 3)萃取。合并有机相,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩得到粗品浅棕色油状物7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺(123mg,90%),无需进一步纯化即可用于下一步。
LC_MS:(ES+):m/z 253.20[M+H]+.
实施例1
N-(叔丁基)-3-((5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯磺酰胺
实施步骤(实施方法一):
步骤1:片段1-c N-(叔丁基)-3-((2-氯-5-氟嘧啶-4-基)氨基)苯磺酰胺的合成
将含有2,4-二氯-5-氟嘧啶(500mg,3mmol)和3-氨基-N-(叔丁基)苯磺酰胺(685mg,3mmol)的甲醇(10ml)溶液加热至50℃搅拌反应3小时。TLC监测反应完全。反应液减压浓缩,所得粗品经硅胶柱层析(用15%-25%乙酸乙酯的正己烷溶液洗脱)分离纯化得白色固体N-(叔丁基)-3-((2-氯-5-氟嘧啶-4-基)氨基)苯磺酰胺(362mg,33%)。
LC_MS:(ES+):m/z 360.0[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.27(s,1H),8.38(d,J=3.6Hz,1H),8.21(t,J=2.4Hz,1H),7.88-7.91(m,1H),7.55-7.59(m,3H),1.14(s,9H),
步骤2:化合物1N-(叔丁基)-3-((5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯磺酰胺的合成
在室温氮气氛围下向含有N-(叔丁基)-3-((2-氯-5-氟嘧啶-4-基)氨基)苯磺酰胺(40mg,0.11mmol),4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(23mg,0.11mmol)和磷酸钾(70mg,0.33mmol)的1,4-二氧六环(1ml)悬浮液中加入BrettPhos Pd G3(10mg,0.011mmol),所得混合物氮气置换3次,加热至110℃搅拌反应14小时。TLC监测反应完全。反应液冷却至室温,分配于水(10ml)和乙酸乙酯(10ml)中。收集有机层,水层用乙酸乙酯(10ml)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用5%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体目标化合物(21.1mg,35%)。
LC_MS:(ES+):m/z 531.5[M+H]+.
1H NMR(400MHz,CDCl3):δ8.27(s,1H),7.95(d,J=2.8Hz,1H),7.76(d,J=8.0Hz,1H),7.59(d,J=7.6Hz,1H),7.41(t,J=8.0Hz,1H),6.94-6.99(m,2H),6.86-6.90(m,1H),6.69(d,J=8.0Hz,1H),4.65(brs,1H),4.24(t,J=4.4Hz,2H),3.62(t,J=5.6Hz,2H),3.42-3.48(m,4H),3.37(s,3H),1.20(s,9H).
实施例2
N-(氰甲基)-4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺
实施步骤(实施方法二):
步骤1:片段2-b(4-((氰甲基)氨基甲酰基)苯基)硼酸的合成
在0℃下向含有4-羧基苯硼酸(2.0g,12.0mmol)和N,N-二甲基甲酰胺(2ml)的二氯甲烷(80ml)溶液中加入草酰氯(2.4ml,26.5mmol)。所得混合物加热回流3小时。TLC监测反应完全。反应液浓缩,剩余物溶于N,N-二甲基甲酰胺(5ml)中,在0℃氮气保护下,滴加到含有氨基乙腈盐酸盐(1.23g,13.2mmol)和N,N-二异 丙基乙胺(3.9g,30.1mmol)的N,N-二甲基甲酰胺(10ml)溶液中。所得混合物在室温下搅拌反应12小时。TLC监测反应完全。反应混合物中加入水(150ml),乙酸乙酯萃取(50ml x 5)。合并有机层,无水硫酸钠干燥,过滤,减压浓缩,所得粗品在乙酸乙酯中重结晶得到淡黄色固体(4-((氰甲基)氨基甲酰基)苯基)硼酸(950mg,38%)
1H NMR(400MHz,DMSO-d6):δ9.18(t,J=5.4Hz,1H),8.23(s,2H),7.81-7.90(m,4H),4.31(d,J=5.6Hz,2H).
步骤2:片段2-d 2,4-二氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶的合成
在0℃下向含有2,4-二氯-7H-吡咯并[2,3-d]嘧啶(5g,26.6mmol)的N,N-二甲基甲酰胺(50ml)溶液中加入氢化钠(60%,1.3g,31.9mmol)。所得混合物升至室温搅拌反应1小时后,冷却至0℃,然后逐滴加入加2-(三甲基硅烷基)乙氧甲基氯(4.9g,29.3mmol)。升至室温,氮气保护下搅拌反应14小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(150ml)和水(300ml)中。收集有机层,饱和氯化钠溶液(100ml x 2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用1.5%乙酸乙酯的正己烷溶液洗脱)分离纯化得到无色油状物2,4-二氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(2.3g,27%)。
1H NMR(400MHz,CDCl3):δ7.37(d,J=3.6Hz,1H),6.66(d,J=3.6Hz,1H),5.60(s,2H),3.53(t,J=8.2Hz,2H),0.92(t,J=8.4Hz,2H),-0.04(s,9H).
步骤3:片段2-e 4-(2-氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-N-(氰基甲基)苯甲酰胺的合成
在室温氮气氛围下向含有(4-((氰甲基)氨基甲酰基苯硼酸(200mg,0.98mmol),2,4-二氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(624mg,1.96mmol)和碳酸钠水溶液(1ml,2N)的乙腈(2ml)中加入四(三苯基膦)钯(113mg,0.1mmol),所得反应混合物氮气置换3次,升温至90℃搅拌反应12小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(10ml)和水(10ml)。收集有机相,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用0.5-2%甲醇的二氯甲烷溶液洗脱)分离纯化得到白色固体4-(2-氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-N-(氰基甲基)苯甲酰胺(300mg,34%)。
LC_MS:(ES+):m/z 442.1[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.41(t,J=5.4Hz,1H),8.28(d,J=8.4Hz,2H),8.08(d,J=8.4Hz,2H),7.92(d,J=3.6Hz,1H),7.10(d,J=3.6Hz,1H),5.64(s,2H),4.37(d,J=5.6Hz,2H),3.56(d,J=8.0Hz,2H),0.86(t,J=8.0Hz,2H),-0.08(s,9H).
步骤4:片段2-f N-(氰甲基)-4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺的合成
在室温氮气氛围下向含有4-(2-氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-N-(氰基甲基)苯甲酰胺(60.0mg,0.1mmol),4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(35.4mg,0.2mmol)和磷酸钾(89.0mg,0.4mmol)的1,4-二氧六环(1ml)溶液中,加入BrettPhos Pd G3(9.1mg,0.01mmol),所得混合物氮气置换3次,升温至110℃搅拌反应13小时。TLC监测反应完全。反应混合物冷却至室温,分配于水(20ml)中,乙酸乙酯萃取(10ml x2)。合并有机相,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用5%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体N-(氰甲基)-4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺(62mg,72%)。
LC_MS:(ES+):m/z 614.4[M+H]+.
步骤5:N-(氰甲基)-4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺的合成
将含有N-(氰甲基)-4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺(60mg,0.1mmol)和四丁基氟化铵(0.5ml,1M四氢呋喃溶液)的四氢呋喃(0.5ml)溶液升温至70℃搅拌反应5小时。TLC监测反应完全。反应混合物冷却至室 温,分配于水(10ml)中。收集有机相,水相用乙酸乙酯(10ml x2)萃取。合并有机相,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用10%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体目标化合物(16.5mg,34%)。
LC_MS:(ES+):m/z 484.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.63(s,1H),9.35(t,J=5.6Hz,1H),8.83-9.23(m,1H),8.24(d,J=7.6Hz,2H),8.05(d,J=8.4Hz,2H),7.07-7.68(m,3H),6.68(s,2H),4.36(d,J=5.2Hz,2H),3.92-4.28(m,2H),3.49-3.59(m,2H),3.32-3.41(m,2H),3.29-3.31(m,2H),3.28(s,3H).
实施例3
1-(3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-基)丙-2-烯-1-酮
实施步骤(实施方法三):
步骤1:片段3-b 3-((2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-甲酸叔丁酯的合成
向含有片段2-d:2,4-二氯-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(100mg,0.315mmol),3-氨基哌啶-1-甲酸叔丁酯(94.6mg,0.473mmol)和N-乙基-N-异丙基丙-2-胺(61mg,0.473mmol)的乙醇(2ml)溶液在90℃下搅拌反应18小时。TLC监测反应完全。反应混合物减压浓缩,所得粗品经硅胶柱层析(用含25%乙酸乙酯的石油醚洗脱)分离纯化得到白色固体3-(((2-氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-甲酸叔丁酯(130mg,85%)。
LC_MS:(ES+):m/z 482.2[M+H]+.
步骤2:片段3-c 3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-甲酸叔丁酯的合成
室温氮气氛围下向含有3-(((2-氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-甲酸叔丁酯(84mg,0.17mmol),4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(36mg,0.17mmol)和磷酸钾(111mg,0.53mmol)的1,4-二氧六环(2ml)悬浮液中加入BrettPhos Pd G3(16mg,0.017mmol),反应混合物氮气置换三次,110℃下搅拌反应20小时。TLC监测反应完全。反 应混合物冷至室温,分配于水(10ml)和乙酸乙酯(10ml)中。收集有机层,水层用乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含6%甲醇和50%乙酸乙酯的石油醚洗脱)分离纯化得到棕色固体叔丁基3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-甲酸(60mg,47%)。
LC_MS:(ES+):m/z 654.9[M+H]+.
步骤3:片段3-d N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-N4-(哌啶-3-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺的合成
将含有3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-甲酸叔丁酯(75mg,0.11mmol)和三氟乙酸(1ml)的二氯甲烷(2ml)溶液在室温下搅拌反应5小时。TLC监测反应完全。反应混合物浓缩,剩余物加入到氨水(5ml)和乙酸乙酯(5ml)中。反应混合物在40℃下搅拌反应16小时。TLC监测反应完全。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇的二氯甲烷洗脱)分离纯化得到黄色固体N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-N4-(哌啶-3-基)-7H-吡咯[2,3-d]嘧啶-2,4-二胺(40mg,82%)。
LC_MS:(ES+):m/z 424.0[M+H]+.
步骤4:1-(3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-基)丙-2-烯-1-酮的合成
0℃下向含有N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-N4-(哌啶-3-基)-7H-吡咯[2,3-d]嘧啶-2,4-二胺(40mg,0.09mmol)和三乙胺(28mg,0.28mmol)的二氯甲烷(2ml)溶液中加入丙烯酰氯(8.5mg,0.09mmol)。反应混合物在室温下搅拌反应4小时。TLC监测反应完全。反应混合物分配于水(5ml)和10%甲醇的二氯甲烷(10ml)中。收集有机层,水层要用含10%甲醇的二氯甲烷(10ml x 3)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇的二氯甲烷洗脱)分离纯化得到棕色固体1-(3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)哌啶-1-基)丙-2-烯-1-酮(12.1mg,26%)。
LC_MS:(ES+):m/z 478.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.06(s,1H),8.49(brs,1H),7.23-7.35(m,2H),7.10(brs,1H),6.83-7.01(m,2H),6.65-6.74(m,1H),6.52(s,1H),6.06-6.20(m,1H),5.57-5.77(m,1H),4.54(d,J=10.8Hz,1H),4.04-4.17(m,5H),3.57(t,J=5.6Hz,2H),3.41(brs,2H),3.32(s,3H),3.20-3.26(m,1H),3.08-3.16(m,1H),2.73-2.98(m,1H),2.05-2.11(m,1H),1.88-1.90(m,1H),1.52-1.70(m,2H).
实施例4
N-(叔丁基)-3-((6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺
实施步骤(实施方法四):
步骤1:片段4-b N-(叔丁基)-3-((6-氯-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺的合成
将含有4,6-二氯-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶(196mg,0.83mmol),片段1-b:3-氨基-N-(叔丁基)苯磺酰胺(189mg,0.83mmol)和N,N-二异丙基乙胺(128mg,0.99mmol)的异丙醇(10ml)溶液加热回流搅拌反应6小时。TLC监测反应完全。反应液浓缩,剩余物分配于乙酸乙酯(15ml)和水(20ml)中。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用20%-33%乙酸乙酯的正己烷溶液洗脱)分离纯化得到白色固体N-(叔丁基)-3-((6-氯-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺(150mg,45%)。
LC_MS:(ES+):m/z 465.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.77(s,1H),8.34(s,1H),8.19(s,1H),8.02-8.11(m,1H),7.61-7.66(m,3H),5.81-5.84(m,1H),3.94,3.97(two singles,1H),3.67-3.73(m,1H),2.33-2.44(m,1H),1.74-2.03(m,3H),1.50-1.62(m,2H),1.15(s,9H).
步骤2:片段4-c N-(叔丁基)-3-((6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺的合成
在室温氮气氛围下向含有N-(叔丁基)-3-((6-氯-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺(44.6mg,0.096mmol),4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(20mg,0.096mmol)和磷酸钾(61mg,0.29mmol)的1,4-二氧六环(1ml)悬浮液中加入BrettPhos Pd G3(8.7mg,0.009mmol),所得混合物氮气置换3次,加热至110℃搅拌反应16小时。TLC监测反应完全。反应液冷却至室温,分配于水(10ml)和乙酸乙酯(10ml)中。收集有机层,水层用乙酸乙酯(10mlx2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干 燥,减压浓缩,所得粗品经制备TLC(用5%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体N-(叔丁基)-3-((6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺(52mg,80%)。
LC_MS:(ES+):m/z 637.2[M+H]+.
步骤3:N-(叔丁基)-3-((6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺的合成
将含有N-(叔丁基)-3-((6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺(52mg,0.082mmol)和三氟乙酸(0.5ml)的二氯甲烷(1.5ml)溶液在室温下搅拌反应2小时。TLC监测反应完全。减压浓缩有机溶剂,剩余物加入碳酸钠溶液调节pH至7-8,然后加入二氯甲烷(10ml×2)萃取。合并有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用7.5%甲醇的二氯甲烷溶液洗脱)分离纯化得黄色固体N-(叔丁基)-3-((6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺(20mg,46%)。
LC_MS:(ES+):m/z 553.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ12.96(s,1H),9.90(s,1H),8.81(s,1H),8.46(d,J=7.6Hz,1H),8.28(brs,1H),8.11(s,1H),7.68(s,1H),7.47-7.53(m,2H),7.31(brs,1H),7.06-7.09(m,1H),6.66(d,J=8.8Hz,1H),4.15(t,J=4.2Hz,2H),3.53(t,J=5.6Hz,2H),3.40(t,J=5.4Hz,2H),3.33-3.35(m,1H),3.29-3.30(m,1H),3.27(s,3H),1.14(s,9H).
实施例5
4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶-6-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺
实施步骤(实施方法五):
步骤1:片段5-c 6-氯-4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶的合成
将含有4,6-二氯-吡唑并[3,4-d]嘧啶(500mg,2.64mmol),N-甲基吲哚(347mg,2.64mmol)和三氯化铝的1,2-二氯乙烷(10ml)溶液加热回流反应12小时。TLC监测反应完全。反应混合物分配于二氯甲烷(10ml)和水(20ml)中。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩得到淡黄色粗品6-氯-4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶(1.4g),无需进一步纯化即可用于下一步。
步骤2:片段5-d 6-氯-4-(1-甲基-1H-吲哚-3-基)-1-(2-(三甲基硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-d]嘧啶的合成
在0℃下向含有6-氯-4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶(1.4g,4.9mmol)的N,N-二甲基甲酰胺(10ml)溶液中,加入氢化钠(60%,294mg,7.3mmol)。混合物升至室温搅拌反应1小时,然后冷至0℃,将2-(三甲基硅烷基)乙氧甲基氯(980g,5.8mmol)滴加到反应液中。所得混合物在室温氮气氛围下搅拌下反应14小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(100ml)和水(100ml)中。收集有机层,饱和氯化钠溶液(100ml x 2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用0.5-1%甲醇的二氯甲烷溶液洗脱)分离纯化得到白色固体6-氯-4-(1-甲基-1H-吲哚-3-基)-1-(2-(三甲基硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-d]嘧啶(245mg,两步22%).
LC_MS:(ES+):m/z 414.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.90(d,J=2.8Hz,2H),8.62-8.64(m,1H),7.61-7.63(m,1H),7.31-7.38(m,2H),5.70(s,2H),3.98(s,3H),3.64(t,J=8.0Hz,2H),0.86(t,J=8.0Hz,2H),-0.08(s,9H).
步骤3:片段5-e 4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-d]嘧啶-6-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
在室温氮气保护下向含有6-氯-4-(1-甲基-1H-吲哚-3-基)-1-(2-(三甲基硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-d]嘧啶(100mg,0.24mmol),4-甲基-3,4,5,6-四氢-2H- 苯并[b][1,4,7]二恶唑啉-9-胺(55mg,0.24mmol)和磷酸钾(154mg,0.72mmol)的1,4-二氧六环(2ml)溶液中,加入BrettPhos Pd G3(22mg,0.024mmol),所得混合物氮气置换3次,升温至110℃搅拌反应12小时。TLC监测反应完全。反应混合物冷却至室温,分配于水(20ml)和乙酸乙酯(10ml)中。收集有机层,水相用乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩得,所得粗品经制备TLC(用5%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1-(2-(三甲基硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-d]嘧啶-6-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(38mg,27%)
LC_MS:(ES+):m/z 586.5[M+H]+.
步骤4:4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶-6-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
在室温下将含有4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1-(2-(三甲基硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-d]嘧啶-6-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(38mg,0.065mmol)和三氟乙酸(0.5ml)的二氯甲烷(0.5ml)溶液搅拌反应3小时。TLC监测反应完全。反应液浓缩,剩余物加入到含有氨水(2ml)和乙酸乙酯(4ml)的混合溶液中。所得混合物在30℃下搅拌反应16小时。TLC监测反应完全。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用10%甲醇的二氯甲烷溶液洗脱)分离纯化得到黄色固体4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶-6-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(18.5mg,62%)。
LC_MS:(ES+):m/z 456.2[M+H]+.
1H NMR(400MHz,CDCl3):δ8.62(d,J=7.6Hz,1H),8.14(s,1H),7.91(s,1H),7.28-7.41(m,6H),7.06-7.08(m,1H),6.68(d,J=8.4Hz,1H),4.26(t,J=4.2Hz,2H),3.93(s,3H),3.61(t,J=5.6Hz,2H),3.42-3.55(m,4H),3.38(s,3H).
实施例6
1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)环己醇
实施步骤(实施方法六):
步骤1:片段6-b 8-甲基-1,4-二氧杂螺[4.5]癸-8-醇的合成
-60℃下向含有1,4-环己二酮单乙二醇缩酮(2g,12.8mmol)的无水四氢呋喃(20ml)的悬浮液中加入甲基溴化镁(3M溶在四氢呋喃中,7.7ml,23mmol),反应 混合物在-60℃下搅拌反应20分钟,然后升至-30℃搅拌30分钟,再在0℃搅拌反应30分钟。TLC监测反应完全。反应混合物用饱和氯化铵溶液(10ml)淬灭,乙酸乙酯(20ml)萃取。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩得到无色油状物8-甲基-1,4-二氧杂螺[4.5]癸-8-醇(1.4g,63%),未经进一步纯化直接用于下一步。
步骤2:片段6-c 4-羟基-4-甲基环己酮的合成
将含有8-甲基-1,4-二氧杂螺[4.5]癸-8-醇(1.4g,8.1mmol)和对甲苯磺酸吡啶鎓(408.6mg,1.63mmol)的丙酮(16ml)和水(8ml)的悬浮物在60℃氮气保护下搅拌反应13小时。TLC监测反应完全。反应混合物冷至室温,加入水(50ml)淬灭,乙酸乙酯(30ml x 2)萃取。合并有机层,保饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含15%乙酸乙酯的正己烷洗脱)分离纯化得到黄色油状物4-羟基-4-甲基环己酮(630mg,57%)。
步骤3:片段6-d(1r,4r)-1-甲基环己烷-1,4-二醇和6-e(1s,4s)-1-甲基环己烷-1,4-二醇的合成
0℃下向含有4-羟基-4-甲基环己酮(630mg,4.9mmol)的甲醇(6ml)悬浮液中加入硼氢化钠(372.1mg,9.5mmol),反应混合物在室温下搅拌反应12小时。TLC监测反应完全。反应混合物用水(0.5ml)淬灭,减压浓缩,所得粗品经硅胶柱层析(用含4%甲醇的二氯甲烷洗脱)分离纯化得到白色固体6-d(1r,4r)-1-甲基环己烷-1,4-二醇(80mg,12%),
1H NMR(400MHz,CDCl3):δ4.04–3.71(m,1H),1.89(dq,J=16.0,9.6,6.7Hz,2H),1.89(dq,J=16.0,9.6,6.7Hz,2H),1.59–1.36(m,6H),1.26(s,3H).
以及白色固体6-e(1s,4s)-1-甲基环己烷-1,4-二醇(400mg,60%);
1H NMR(400MHz,CDCl3):δ3.60(tt,J=9.7,4.1Hz,1H),1.81–1.72(m,2H),1.72–1.58(m,4H),1.55–1.37(m,4H),1.22(s,3H).
步骤4:片段6-f(1s,4s)-4-((2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-1-甲基环己烷-1-醇的合成
0℃氮气保护下向含有(1s,4s)-1-甲基环己烷-1,4-二醇(40.4mg,0.3mmol)和2,4-二氯-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(100mg,0.3mmol)的无水四氢呋喃(5ml)溶液中加入叔丁醇钾(70.7mg,0.6mmol),反应混合物在室温下搅拌反应13小时。TLC监测反应完全。反应混合物分配于水(20ml)和乙酸乙酯中(20ml)中。水相用乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(30ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含25%乙酸乙酯的正己烷洗脱)分离纯化得到无色油状物(1s,4s)-4-((2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-1-甲基环己烷-1-醇(100mg,79%)。
LC_MS:(ES+):m/z 412.2[M+H]+.
步骤5:片段6-g(1s,4s)-1-甲基-4-((2-((7-甲基l-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)环己-1-醇的合成
室温氮气氛围下向含有(1s,4s)-4-((2-氯-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-1-甲基环己醇(100mg,0.24mmol),Amine-3:6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺(61.2mg, 0.24mmol),磷酸钾(154.8mg,0.73mmol)的1,4-二氧六环(2ml)悬浮液中加入BrettPhos Pd G3(21.8mg,0.02mmol),反应混合物氮气置换三次,110℃下搅拌反应14小时。TLC监测反应完全。反应混合物冷至室温,分配于水(20ml)和乙酸乙酯(10ml)中。收集有机层,水层用乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇和1%氨水的二氯甲烷洗脱)得到黄色固体(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)己醇(70mg,46%)。
LC_MS:(ES+):m/z 628.3[M+H]+.
步骤6:(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)环己-1-醇的合成
将含有(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)己醇(70mg,0.11mmol)和三氟乙酸(1ml)的二氯甲烷溶液(1ml)在0℃下搅拌反应3小时。TLC监测反应完全。减压移除挥发物,剩余物用饱和碳酸钠溶液调节pH=7~8,二氯甲烷(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,浓缩。剩余物加入到含有氨水(2ml)和乙酸乙酯(4ml)的混合溶剂中。反应混合物在30℃下搅拌反应12小时。TLC监测反应完全。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇的二氯甲烷洗脱)分离纯化得到橘色固体1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)环己醇(42.6mg,76%)。
LC_MS:(ES+):m/z 498.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.33(s,1H),8.90(s,1H),7.66(s,1H),7.35(brs,1H),6.95-6.98(m,2H),6.25-6.27(m,1H),5.16-5.22(m,1H),4.08-4.22(m,5H),3.72(s,4H),3.29(s,3H),2.82-3.05(m,4H),1.86-1.87(m,4H),1.66-1.69(m,2H),1.41-1.48(m,2H),1.17(s,3H).
实施例7
(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)-5-(三氟甲基)嘧啶-4-基)氧基)环己-1-醇
实施步骤(实施方法七):
步骤1:片段7-b(1s,4s)-4-((2-氯-5-(三氟甲基)嘧啶-4-基)氧基)-1-甲基环己-1-醇和片段7-c(1s,4s)-4-((4-氯-5-(三氟甲基)嘧啶-2-基)氧基)-1-甲基环己-1-醇的合成
0℃氮气保护下向含有(1s,4s)-1-甲基环己烷-1,4-二醇(100mg,0.77mmol)的四氢呋喃(3ml)溶液加入氢化钠(60%,68mg,1.69mmol),混合物缓慢升至室温后搅拌反应30分钟,然后将反应液滴加到2,4-二氯-5-(三氟甲基)嘧啶(334mg,1.54mmol)的四氢呋喃(2ml)溶液中,反应液在室温下搅拌反应16小时。TLC监测反应完全。反应混合物用饱和氯化铵溶液(10ml)淬灭,乙酸乙酯(10ml x 2)萃取。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含6%-12.5%乙酸乙酯的正己烷洗脱)分离纯化得到两组化合物:无色油状物7-b(第一流出组分)(1s,4s)-4-((2-氯-5-(三氟甲基)嘧啶-4-基)氧基)-1-甲基环己-1-醇(28mg,12%);
LC_MS:(ES+):m/z 311.0[M+H]+.
和无色油状物7-c(第二流出组分)(1s,4s)-4-((4-氯-5-(三氟甲基)嘧啶-2-基)氧基)-1-甲基环己-1-醇(52mg,21%)。
LC_MS:(ES+):m/z 311.0[M+H]+.
步骤2:(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)-5-(三氟甲基)嘧啶-4-基)氧基)环己-1-醇的合成
室温氮气氛围下向含有(1s,4s)-4-((2-氯-5-(三氟甲基)嘧啶-4-基)氧基)-1-甲基环己-1-醇(20.0mg,0.064mmol),6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺(19.4mg,0.077mmol),磷酸钾(154.8mg,0.73mmol)的1,4-二氧六环(2ml)悬浮液中加入BrettPhos Pd G3(21.8mg,0.02mmol),反应混合物氮气置换三次,110℃下搅拌反应12小时。TLC监测反应完全。反应混合物冷至室温, 分配于水(10ml)和乙酸乙酯(10ml)中。收集有机层,水层用乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇和1%氨水的二氯甲烷洗脱)得到灰白色固体(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)-5-(三氟甲基)嘧啶-4-基)氧基)环己-1-醇(10mg,30%)。
LC_MS:(ES+):m/z 527.5[M+H]+.
1H NMR(400MHz,CD3OD):δ8.34(s,1H),7.54(brs,1H),7.28-7.30(m,1H),7.09(d,J=8.8Hz,1H),5.18-5.22(m,1H),4.35(t,J=4.8Hz,2H),4.19(t,J=4Hz,2H),3.82-3.97(m,4H),3.43-3.53(m,2H),2.91(s,3H),1.90-1.96(m,4H),1.77-1.81(m,2H),1.50-1.57(m,2H),1.25(s,3H).
实施例8
2-甲基-1-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)丙-2-醇
实施步骤(实施方法八):
步骤1:片段8-a N-(4-氯-5-(三氟甲基)嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
将含有2,4-二氯-5-(三氟甲基)嘧啶(800mg,3.69mmol),4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(768mg,3.69mmol)和三乙胺(750mg,7.40mmol)的四氢呋喃(30ml)溶液室温下搅拌反应16小时。TLC监测反应完全。反应液浓缩,剩余物分配于乙酸乙酯(15ml)和水(20ml)中。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用20-33%乙酸乙酯的正己烷溶液洗脱)分离纯化得到黄色固体,然后经制备HPLC分离(流动相:30%~90%乙腈的水溶液梯度洗脱25分钟),得到第一个流出组分为黄色固体N-(2-氯-5-(三氟甲基)嘧啶-4-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(430mg,30%);
LC_MS:(ES+):m/z 389.0[M+H]+.
1H NMR(400MHz,CDCl3):δ8.34(d,J=1.2Hz,1H),6.97(d,J=2.5Hz,1H),6.93(dd,J=8.7,2.6Hz,1H),6.87(s,1H),6.65(d,J=8.6Hz,1H),4.27–4.18(m,2H),3.60(t,J=5.6Hz,2H),3.46(dt,J=8.9,5.0Hz,4H),3.37(s,3H).
第二个流出组分为黄色固体N-(4-氯-5-(三氟甲基)嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(406mg,28%);
LC_MS:(ES+):m/z 389.0[M+H]+.
1H NMR(400MHz,CDCl3):δ8.48(s,1H),7.35(s,1H),7.03(d,J=2.5Hz,1H),6.93(d,J=7.8Hz,1H),6.64(d,J=8.7Hz,1H),4.30–4.17(m,2H),3.59(t,J=5.6Hz,2H),3.45(dq,J=11.9,6.1,4.5Hz,4H),3.36(s,3H).
步骤2:2-甲基-1-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)丙-2-醇的合成
将含有N-(4-氯-5-(三氟甲基)嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(30mg,77.2μmol),1-氨基-2-甲基丙-2-醇(14mg,155μmol)和三乙胺(16mg,155μmol)的1,4-二氧六环(1ml)溶液80℃下搅拌反16小时。TLC监测反应完全。反应液浓缩,剩余物分配于乙酸乙酯(15ml)和水(20ml)中。TLC监测反应完全。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含50%乙酸乙酯的石油醚洗脱)分离纯化得到橘色固体2-甲基-1-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)丙-2-醇(18mg,52%)。
LC_MS:(ES+):m/z 442.1[M+H]+.
实施例9
N-(叔丁基)-3-((5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-13-基)氨基)嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法一,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 575.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.67(s,1H),9.10(s,1H),8.19-8.14(m,3H),7.60(s,1H),7.52-7.49(m,2H),7.43(s,1H),7.22-7.19(m,1H),6.96-6.94(d,J=8.4Hz,1H),4.13-4.09(m,2H),3.96(s,2H),3.73(s,4H),3.30(s,3H),3.18-2.85(m,4H),1.12(s,9H).
实施例10
N4-(2-(异丙基磺酰基)苯基)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段2-d替换片段4-a,用片段10-a:2-(异丙基磺酰)苯胺替换片段1-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 567.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.43(s,1H),9.48(s,1H),8.88-8.93(m,2H),7.82(d,J=8.0Hz,1H),7.73(t,J=7.8Hz,1H),7.58(s,1H),7.27-7.31(m,2H),7.04(d,J=3.2Hz,1H),6.91(d,J=8.4Hz,1H),6.25(d,J=2.8Hz,1H),4.01-4.12(m,4H),3.61-3.77(m,4H),3.42-3.51(m,1H),3.29(s,3H),2.72-2.87(m,2H),2.26-2.38(m,2H),1.18(d,J=6.8Hz,6H).
实施例11
N4-(2-(异丙基磺酰基)苯基)-N6-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-1H-吡唑并[3,4-d]嘧啶-4,6-二胺
实施方法同方法四,用片段2-d替换片段4-a,用片段2-(异丙基磺酰)苯胺替换片段1-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 568.6[M+H]+.
1H NMR(400MHz,CDCl3):δ9.86(s,1H),8.75-8.77(m,1H),7.89-7.92(m,2H),7.66-7.70(m,1H),7.56(s,1H),7.25-7.29(m,1H),7.08-7.11(m,1H),6.94-6.96(m, 1H),4.41(s,2H),4.14-4.16(m,2H),4.07(s,2H),3.90(s,2H),3.62(s,4H),3.22-3.29(m,1H),2.96(s,3H),1.30-1.32(d,J=6.8Hz,6H).
实施例12
N4-(2-(异丙基磺酰基)苯基)-N6-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)-1H-吡唑并[3,4-d]嘧啶-4,6-二胺
实施方法同方法四,用片段2-d替换片段4-a,用片段2-(异丙基磺酰)苯胺替换片段1-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 568.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.11(s,1H),9.65(s,1H),9.15(s,1H),8.34-8.50(m,1H),7.90-7.92(m,1H),7.78-7.82(m,2H),7.44-7.48(m,2H),7.25-7.28(m,1H),6.90(d,J=8.4Hz,1H),3.98-4.10(m,4H),3.62-3.74(m,4H),3.41-3.46(m,1H),3.30(s,3H),2.73-2.92(m,2H),2.31-2.46(m,2H),1.14(d,J=6.4Hz,6H).
实施例13
N-(5-氟-4-(1-甲基-1H-吲哚-3-基)嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺
实施方法同方法五,用片段1-a替换片段5-a,获得标题化合物。
LC_MS:(ES+):m/z 434.3[M+H]+.
1HNMR(400MHz,CDCl3):δ8.69(d,J=8.0Hz,1H),8.15(d,J=4.0Hz,1H),7.93(d,J=2.4Hz,1H),7.30-7.38(m,2H),7.23-7.26(m,1H),7.13(d,J=2.4Hz,1H),7.02-7.04(m,1H),6.83(s,1H),6.68(d,J=8.4Hz,1H),4.26(t,J=4.4Hz,2H),3.88(s,3H),3.62(t,J=5.6Hz,2H),3.42-3.48(m,4H),3.38(s,3H).
实施例14
2-(1-(乙基磺酰基)-3-(4-(5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段1-a替换片段2-d,用片段14-e替换片段2-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 601.6[M+H]+.
1HNMR(400MHz,DMSO-d6):δ9.57(s,1H),8.72(s,1H),8.56(d,J=2.0Hz,1H),8.25(s,1H),7.59(s,1H),7.33(d,J=8.8Hz,1H),7.03(d,J=8.4Hz,1H),4.53(d,J=9.2Hz,2H),4.24(d,J=8.8Hz,2H),4.21-4.10(m,4H),3.89-3.74(m,4H),3.69(s,2H),3.30(s,3H),3.26-3.21(m,2H),3.14-2.91(m,2H),2.67-2.56(m,2H),1.24(t,J=7.2Hz,3H).
片段14-e的合成方法如下:
步骤1:片段14-a 2-(氮杂环丁烷-3-亚烷基)乙腈三氟乙酸盐的合成
将含有3-(氰基亚甲基)-氮杂环丁烷-1-甲酸叔丁酯(5g,25.7mmol)和三氟乙酸(20ml)的二氯甲烷(20ml)溶液,室温下搅拌反应2.5小时。TLC监测反应完全。反应液减压浓缩得到褐色油状物2-(3-氮杂环丁基亚基)乙腈三氟醋酸盐(5g,粗品),无需进一步纯化即可用于下一步。
步骤2:片段14-c 2-(1-(乙基磺酰基)-3-氮杂环丁亚基)乙腈的合成
0℃下向含有2-(3-氮杂环丁基亚基)乙腈三氟醋酸盐(5g,26.2mmol)和N,N-二异丙基二胺(11.8g,91.6mmol)的二氯甲烷(50ml)溶液中加入乙基磺酰氯(4g,31.4mmol)。所得混合物升至室温反应14小时。TLC监测反应完全。反应混合物分配于二氯甲烷(50ml)和水(100ml)中。收集有机层,饱和氯化钠溶液(100mlx2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用20%-33%乙酸乙酯的己烷溶液洗脱)分离纯化得到黄色固体2-(1-(乙基磺酰基)-3-氮杂环丁亚基)乙腈(3g,两步合并收率62%)
1H NMR(400MHz,DMSO-d6):δ5.89-5.92(m,1H),4.77-4.79(m,2H),4.68-4.70(m,2H),3.20-3.25(q,2H),1.23(t,J=7.4Hz,3H).
步骤3:片段14-e 2-(1-(乙基磺酰基)-3-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈的合成
将含有2-(1-(乙基磺酰基)-3-氮杂环丁亚基)乙腈(2.9g,15.6mmol),4-吡唑硼酸频哪醇酯(3.02g,15.6mmol)和1,8-二氮杂二环十一碳-7-烯(2.37g,15.6mmol)的乙腈(40ml)溶液加热回流反应2小时。TLC监测反应完全。反应混合物减压浓缩,所得粗品经硅胶柱层析(用30%-50%乙酸乙酯的己烷溶液洗脱)分离纯化得到白色固体2-(1-(乙基磺酰基)-3-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈(3g,51%)
1H NMR(400MHz,DMSO-d6):δ8.36(s,1H),7.78(s,1H),4.45(d,J=8.8Hz,2H),4.15(d,J=9.2Hz,2H),3.60(s,2H),3.17-3.23(q,2H),1.28(s,12H),1.23(t,J=7.4Hz,3H).
实施例15
N-(氰基甲基)-4-(5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段1-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 463.3[M+H]+.
1H NMR(400MHz,CDCl3):δ8.31(d,J=3.2Hz,1H),8.18(d,J=8.4Hz,2H),7.89(d,J=8.4Hz,2H),7.14(d,J=2.4Hz,1H),6.94-7.05(m,2H),6.65-6.74(m,2H),4.41(d,J=5.6Hz,2H),4.24(t,J=4.4Hz,2H),3.60(t,J=5.6Hz,2H),3.41-3.48(m,4H),3.37(s,3H).
实施例16
N-(氰基甲基)-4-(5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段1-a替换片段2-d,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 507.8[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.68(s,1H),8.40-8.37(m,1H),8.66-8.65(m,1H),8.16-8.14(m,2H),8.06-8.04(m,2H),7.60(m,1H),7.24-7.22(m,1H),6.97-6.94(m,1H),4.37-4.36(m,2H),4.14-4.12(m,2H),4.07-4.04(m,2H),3.74(m,2H),3.64-3.63(m,2H),2.73-2.62(m,4H),2.28(s,3H).
实施例17
N-(氰基甲基)-4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 528.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.71(s,1H),9.41-9.38(m,1H),9.27(s,1H),8.28-8.26,(m,2H),8.08-8.06(m,2H),7.81(s,1H),7.33-7.32(m,2H),6.96-6.94(m,1H),6.73(m,1H),4.38-4.36(m,2H),4.17-4.08(m,4H),3.78-3.68(m,4H),3.31(s,3H),2.71-2.62(m,2H),2.37-2.30(m,2H).
实施例18
N-(氰基甲基)-4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段4-a替换片段2-d,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 529.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.74(s,1H),9.44(t,J=1.6Hz,1H),8.43-8.36(m,3H),8.10(d,J=8.4Hz,2H),7.73(s,1H),7.39(d,J=8.8Hz,1H),7.00(d,J=8.8 Hz,1H),4.37(d,J=4.8Hz,2H),4.17-4.10(m,4H),3.73(d,J=32.8Hz,4H),2.82-2.68(m,4H),2.37-2.33(m,3H).
实施例19
N-(叔丁基)-3-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法四,用片段2-d替换片段4-a,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 596.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.27(s,1H),9.42(s,1H),8.71(s,1H),8.44(d,J=7.2Hz,1H),8.28(s,1H),7.65(s,1H),7.61(brs,1H),7.51-7.42(m,2H),7.35(d,J=8.0Hz,1H),7.00-6.90(m,2H),6.70(s,1H),4.16-3.99(m,4H),3.77-3.61(m,4H),3.29(s,3H),2.88-2.74(m,2H),2.38-2.23(m,2H),1.15(s,9H).
实施例20
N-(叔丁基)-3-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法四,用片段2-d替换片段4-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 596.7[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.31(s,1H),9.48(s,1H),8.82(s,1H),8.44(d,J=8.0Hz,1H),8.28(s,1H),7.77(s,1H),7.66(s,1H),7.51-7.43(m,2H),7.36-7.33(m,1H),7.02-6.96(m,2H),6.72-6.71(m,1H),4.23(s,2H),4.07(s,2H),3.82(brs,4H),3.32(s,3H),2.80(brs,4H),1.14(s,9H).
实施例21
N-(叔丁基)-3-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法四,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 552.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.24(s,1H),9.42(brs,1H),8.41-8.62(m,2H),8.34(s,1H),7.66(s,1H),7.41-7.49(m,2H),7.27-7.35(m,1H),7.01-7.14(m,1H), 6.82-6.99(m,1H),6.54-6.78(m,2H),4.02-4.28(m,2H),3.52-3.58(m,2H),3.34-3.48(m,4H),3.27(s,3H),1.15(s,9H).
实施例22
N-(叔丁基)-3-((6-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法四,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 597.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.04(s,1H),10.00(s,1H),9.08(s,1H),8.50-8.48(m,1H),8.22(brs,1H),8.13(s,1H),7.70(s,1H),7.56-7.49(m,3H),7.35-7.32(m,1H),6.95(d,J=8.8Hz,1H),4.09-4.05(m,4H),3.72-3.65(m,4H),3.32(m,3H),2.77-2.74(m,1H),2.64-2.60(m,1H),2.33-2.29(m,2H),1.14(s,9H).
实施例23
7-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺
实施方法同方法五,用片段2-c替换片段5-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 499.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.44(s,1H),9.03(brs,1H),8.87(d,J=8.0Hz,1H),8.41(s,1H),7.83(brs,1H),7.53(d,J=8.0Hz,1H),7.46-7.43(m,1H),7.30-7.26(m,1H),7.22-7.18(m,2H),7.00(d,J=8.0Hz,1H),6.87-6.86(m,1H),4.22-4.10(m,4H),3.95(s,3H),3.85-3.70(m,4H),3.31(s,3H),3.13-2.93(m,2H),2.76-2.54(m,2H).
实施例24
7-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶-6-基)-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺
实施方法同方法五,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 500.8[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.17(s,1H),9.38(s,1H),8.90(d,J=8.0Hz,1H),8.66(s,1H),8.50(s,1H),7.69(brs,1H),7.56(d,J=8.0Hz,1H),7.46(d,J=8.4Hz,1H),7.33-7.23(m,2H),6.99(d,J=8.4Hz,1H),4.21-4.06(m,4H),3.97(s,3H),3.81-3.63(m,4H),3.29(s,3H),2.85-2.71(m,2H),2.39-2.30(m,2H).
实施例25
3-环戊基-3-(4-(5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基))氨基)嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段25-b替换片段2-b,片段1-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 492.3[M+H]+.
1H NMR(400MHz,CDCl3):δ8.26(s,1H),8.21(brs,1H),8.17(s,1H),7.14(brs,1H),6.84-7.02(m,2H),6.58-6.73(m,1H),4.19-4.25(m,3H),3.60(t,J=5.6Hz,2H),3.38-3.45(m,2H),3.37(s,3H),3.09-3.15(m,1H),2.91-2.96(m,1H),2.51-2.62(m,1H),1.92-2.00(m,1H),1.48-1.75(m,6H),1.19-1.31(m,3H).
片段25-b的合成方法如下:
步骤1:片段25-b 3-环戊基-3-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑-1-基)丙腈的合成
将含有3-环戊基丙烯腈(100mg,0.8mmol)和4-吡唑硼酸頻哪醇酯(161mg,0.8mmol)的乙腈(1ml)溶液在0℃下搅拌反应10分钟后,然后在0℃下加入1,8-二氮杂二环十一碳-7-烯(126mg,0.8mmol)。反应体系升温至100℃搅拌反应12小时。TLC监测反应完全。反应混合物冷却至室温,减压浓缩,所得粗品经硅胶 柱层析(用30%乙酸乙酯的石油醚溶液洗脱)分离纯化得到无色油状物3-环戊基-3-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑-1-基)丙腈(85mg,32%)。
1H NMR(400MHz,CDCl3):δ7.83(s,1H),7.78(s,1H),4.11-4.18(m,1H),3.03-.309(m,1H),2.85-2.91(m,1H),2.45-2.56(m,1H),1.88-1.96(m,1H),1.61-1.72(m,2H),1.56-1.59(m,1H),1.43-1.54(m,2H),1.32(s,12H),1.25-1.29(m,1H),1.08-1.16(m,1H).
实施例26
3-环戊基-3-(4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段25-b替换片段2-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 557.6[M+H]+.
1HNMR(400MHz,DMSO-d6):δ11.50(s,1H),9.02(s,1H),8.68(s,1H),8.29(s,1H),7.77(s,1H),7.38-7.36(m,1H),7.23-7.22(m,1H),6.98-6.96(m,1H),6.79-6.78(m,1H),4.58-4.52(m,1H),4.16-4.12(m,4H),3.79-3.72(m,4H),3.29(s,3H),3.25-3.16(m,2H),2.97-2.81(m,2H),2.43-2.42(m,2H),1.85-1.82(m,1H),1.25-1.23(m,8H).
实施例27
3-环戊基-3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段25-b替换片段2-b,获得标题化合物。
LC_MS:(ES+):m/z 513.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.45(s,1H),8.72(s,1H),8.66(s,1H),8.26(s,1H),7.40(d,J=2.4Hz,1H),7.14-7.20(m,2H),6.74(d,J=2.8Hz,1H),6.65(d,J=8.8Hz,1H),4.50-4.57(m,1H),4.15(t,J=3.4Hz,2H),3.53(t,J=5.6Hz,2H),3.38(t,J=5.4Hz,2H),3.28-3.30(m,1H),3.27(s,3H),3.24(d,J=9.2Hz,1H),3.17-3.22(m,1H),2.38-2.46(m,1H),1.78-1.86(m,1H),1.51-1.65(m,3H),1.39-1.46(m,1H),1.29-1.38(m,2H),1.14-1.23(m,2H),
实施例28
2-(1-(乙基磺酰基)-3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段14-a替换片段2-b,获得标题化合物。
LC_MS:(ES+):m/z 578.6[M+H]+.
1H NMR(400MHz,CDCl3):δ9.36(s,1H),8.37(s,1H),8.27(s,1H),7.26-7.28(m,2H),6.93-7.14(m,2H),6.77-6.88(m,1H),6.66(d,J=8.4Hz,1H),6.47-6.59(m, 1H),4.62(d,J=9.2Hz,2H),4.24(d,J=8.8Hz,4H),3.60(t,J=5.4Hz,2H),3.37-3.51(m,8H),3.06-3.11(q,2H),1.41(t,J=7.4Hz,3H).
实施例29
2-(1-(乙基磺酰基)-3-(4-(6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段14-a替换片段2-b,用片段4-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 579.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.26(s,1H),9.31(s,1H),8.94(s,1H),8.44,8.51(two singles,2H),7.39(brs,1H),7.19(d,J=7.2Hz,1H),6.66-6.69(m,1H),4.59(d,J=9.2Hz,2H),4.26(d,J=8.8Hz,2H),4.11-4.20(m,2H),3.71(brs,2H),3.49-3.57(m,2H),3.37-3.45(m,2H),3.23-3.32(m,7H),1.25(t,J=7.0Hz,3H).
实施例30
5-氟-N4-(1H-吲哚-5-基)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)嘧啶-2,4-二胺
实施方法同方法一,用片段1H-吲哚-5-胺替换片段1-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 479.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.07(s,1H),9.14(s,1H),8.96(s,1H),8.00(d,J=4.0Hz,1H),7.82(s,1H),7.43(s,1H),7.34-7.36(m,2H),7.27-7.30(m,1H),7.11-7.13(m,1H),6.79(d,J=8.0Hz,1H),6.39(s,1H),4.01(s,2H),3.62(s,4H),3.47(s,2H),2.67-2.95(m,4H),2.33(s,3H).
实施例31
N-(4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺
实施方法同方法二,用片段31-b替换片段2-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 560.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.69(s,1H),9.23(s,1H),8.16(s,1H),7.81(s,1H),7.74(d,J=12.0Hz,1H),7.38-7.41(m,1H),7.32-7.33(m,1H),6.97(d,J=8.4Hz,1H),6.67-6.68(m,1H),4.82-4.93(m,1H),4.13-4.17(m,4H),3.74-3.80(m,4H),3.32(s,3H),2.94(brs,4H),2.65(s,3H),1.64(d,J=7.2Hz,6H).
31-b实施方式如下:
步骤1:片段31-b 4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-苯并[d]咪唑的合成
室温氮气氛围下向含有6-溴-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(300mg,1.11mmol),4,4,4',4',5,5,5',5'-八甲基-2,2'-双(1,3,2-二氧杂硼烷)(338mg,1.33mmol)和乙酸钾的二氧六环(5ml)悬浮液中加入1,1'-双(二苯基膦基)二茂铁二氯化钯(II)(80mg,0.11mmol),反应混合物氮气置换三次,100℃下搅拌反应14小时。TLC监测反应完全。反应混合物浓缩,所得粗品经硅胶柱层析(用含50%乙酸乙酯的正己烷洗脱)分离纯化得到白色固体4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-苯并[d]咪唑(300mg,85%)。
LC_MS:(ES+):m/z 319.1[M+H]+.
实施例32
二甲基(2-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯基)氧化膦
实施方法同方法四,用片段2-d替换片段4-a,用片段(2-氨基苯基)二甲基氧化膦替换片段1-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 537.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.62(s,1H),11.27(s,1H),9.10-9.13(m,1H),8.81(s,1H),7.48-7.59(m,3H),7.32-7.35(m,1H),7.06(t,J=7.2Hz,1H),6.90-6.94(m,2H),6.35-6.37(m,1H),4.11(t,J=3.8Hz,2H),4.04(t,J=5.0Hz,2H),3.72(t,J=3.8Hz,2H),3.64(t,J=4.8Hz,2H),2.26(s,3H),2.68-2.76(m,2H),2.58-2.65(m,2H),1.83(d,J=13.6Hz,6H).
实施例33
二甲基(2-((6-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯基)氧化膦
实施方法同方法四,用片段(2-氨基苯基)二甲基氧化膦替换片段1-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 538.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.05(s,1H),12.06(s,1H),9.19(s,1H),9.02(s,1H),7.75(s,1H),7.52-7.66(m,3H),7.35(d,J=8Hz,1H),7.15(t,J=7.2Hz,1H),6.96(d,J=8.8Hz,1H),4.09-4.11(m,4H),3.67-3.74(m,4H),3.29(s,3H),2.67-2.82(m,2H),2.32-2.33(m,2H),1.85(d,J=13.6Hz,6H).
实施例34
N4-(4-溴-2-氟苯基)-N6-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-1H-吡唑并[3,4-d]嘧啶-4,6-二胺
实施方法同方法四,用片段4-溴-2-氟苯胺替换片段1-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 558.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ12.99(s,1H),9.76(s,1H),9.05(s,1H),8.07(s,1H),7.80(t,J=8.2Hz,1H),7.64-7.69(m,2H),7.44(d,J=8.8Hz,1H),7.27-7.29(m,1H),6.97(d,J=8.8Hz,1H),4.17-4.33(m,2H),3.75-4.01(m,6H),3.31(s,3H),2.66-3.05(m,4H).
实施例35
N4-(2-(异丙基磺酰基)苯基)-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段2-d替换片段4-a,用片段2-(异丙基磺酰基)苯胺替换片段1-b,获得标题化合物。
LC_MS:(ES+):m/z 523.6[M+H]+.
1H NMR(400MHz,CDCl3):δ9.58(brs,1H),9.16(brs,1H),8.87(d,J=8.4Hz,1H),7.84-7.86(m,1H),7.59-7.64(m,1H),7.13-7.19(m,2H),6.89-7.00(m,1H),6.77-6.86(m,1H),6.64-6.72(m,2H),6.41(d,J=3.6Hz,1H),4.24(t,J=4.2Hz,2H),3.60(t,J=5.8Hz,2H),3.41-3.52(m,4H),3.37(s,3H),3.22-3.29(m,1H),1.30(d,J=7.2Hz,6H).
实施例36
N4-(2-(异丙基磺酰基)苯基)-N6-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-1H-吡唑并[3,4-d]嘧啶-4,6-二胺
实施方法同方法四,用片段2-(异丙基磺酰基)苯胺替换片段1-b,获得标题化合物。
LC_MS:(ES+):m/z 524.6[M+H]+.
1H NMR(400MHz,CDCl3):δ9.83(brs,1H),8.78(d,J=8.4Hz,1H),7.87-7.91(m,2H),7.64(t,J=7.8Hz,1H),7.21-7.26(m,1H),7.17(brs,1H),6.85-7.05(m,1H),6.52-6.78(m,1H),4.25(brs,2H),3.61(t,J=5.4Hz,2H),3.41-3.56(m,4H),3.38(s,3H),3.20-3.27(m,1H),1.31(d,J=6.8Hz,6H),
实施例37
2-(1-(乙基磺酰基)-3-(4-(5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段14-a替换片段2-b,用片段1-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 557.1[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.22-8.30(m,3H),7.17(s,1H),4.59(d,J=9.2Hz,2H),4.23(d,J=9.6Hz,4H),3.60(t,J=5.2Hz,1H),3.37,3.38(two singles,5H),3.05-3.11(q,2H),1.53-1.70(m,2H),1.41(t,J=7.4Hz,3H),1.19-1.34(m,2H).
实施例38
N-(氰基甲基)-4-(6-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段4-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 484.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.39(s,1H),9.51(s,1H),9.43(t,J=5.6Hz,1H),8.37(s,1H),8.34(d,J=8.4Hz,2H),8.10(d,J=8.4Hz,2H),7.39(d,J=2.0Hz,1H),7.18-7.20(m,1H),6.69(d,J=8.8Hz,1H),4.37(t,J=2.8Hz,2H),4.16(t,J=4.2Hz,2H),3.53(t,J=5.6Hz,2H),3.41(t,J=5.6Hz,2H),3.32-3.35(m,2H),3.28(s,3H).
实施例39
4-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺
实施方法同方法五,用片段2-c替换片段5-a,获得标题化合物。
LC_MS:(ES+):m/z 455.4[M+H]+.
1H NMR(400MHz,CDCl3):δ9.21(s,1H),8.51(d,J=4.8Hz,1H),7.91(s,1H),7.38(d,J=8.0Hz,1H),7.30-7.34(m,1H),7.27-7.29(m,2H),7.07(d,J=6.8Hz,1H),6.77(brs,1H),6.66(d,J=8.4Hz,1H),6.60(d,J=2.4Hz,1H),4.24(t,J=4.4Hz,2H),3.91(s,3H),3.60(t,J=5.6Hz,2H),3.38-3.51(m,4H),3.36(s,3H).
实施例40
N-(叔丁基)-3-((5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法一,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 575.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.68(s,1H),9.11(s,1H),8.20-8.16(m,3H),7.63(s,1H),7.52-7.41(m,3H),7.23(d,J=8.8Hz,1H),6.96(d,J=8.4Hz,1H),4.07-4.03(m,4H),3.70(s,4H),3.32(s,3H),3.17-2.67(m,4H),1.12(s,9H).
实施例41
N-(5-氟-4-(1-甲基-1H-吲哚-3-基)嘧啶-2-基)-7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺
实施方法同方法五,用片段1-a替换片段5-a,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 478.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.38(s,1H),9.74(d,J=8.0Hz,1H),8.40(d,J=3.6Hz,1H),8.23(d,J=2.8Hz,1H),7.58-7.54(m,2H),7.32(t,J=7.2Hz,2H),7.22(t,J=7.6Hz,1H),7.03(d,J=8.8Hz,1H),4.23-4.04(m,4H),3.93(s,3H),3.83-3.64(m,4H),3.31(s,3H),3.18-2.70(m,4H).
实施例42
3-环戊基-3-(4-(5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段25-b替换片段2-b,片段1-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 536.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.57(s,1H),8.57(d,J=1.6Hz,1H),8.52(d,J=2.8Hz,1H),8.18(s,1H),7.59(d,J=1.6Hz,1H),7.40(d,J=8.8Hz,1H),7.08(d,J=8.8Hz,1H),4.66-4.60(m,1H),4.29(s,2H),4.10(brs,2H),3.82-3.72(m,4H), 3.32(s,3H),3.23-3.20(m,2H),2.79(brs,2H),2.43-2.36(m,2H),1.85-1.79(m,1H),1.65-1.52(m,4H),1.33-1.25(m,2H),1.20-1.11(m,2H).
实施例43
2-(1-(乙基磺酰基)-3-(4-(5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段14-a替换片段2-b,片段1-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 601.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.61(s,1H),8.74(s,1H),8.57(d,J=2.4Hz,1H),8.26(s,1H),7.59(s,1H),7.36(d,J=7.6Hz,1H),7.07(d,J=7.6Hz,1H),4.54(d,J=9.2Hz,2H),4.25(d,J=9.2Hz,4H),4.10-4.09(m,2H),3.74-3.69(m,6H),3.31(s,3H),3.26-3.21(m,2H),2.82(brs,4H),1.24(t,J=7.2Hz,3H).
实施例44
N-(氰基甲基)-4-(5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)苯甲酰胺
实施方法同方法二,片段1-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 507.8[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.79(s,1H),9.52-9.50(m,1H),8.68(d,J=3.2Hz,1H),8.17-8.15(m,2H),8.10-8.07(m,2H),7.65(s,1H),7.33-7.31(m,1H),7.08-7.06(m,1H),4.37-4.35(m,2H),4.28-4.22(m,2H),4.17-4.09(m,2H),3.83-3.70(m,4H),3.32(s,3H),2.92-2.66(m,4H).
实施例45
N-(叔丁基)-3-((6-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法四,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 597.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.08-13.06(m,1H),10.04(s,1H),9.13(s,1H),8.50-8.48(m,1H),8.22-8.16(m,2H),7.70(s,1H),7.61(s,1H),7.56-7.50(m,2H),7.40-7.39(m,1H),7.05-7.03(m,1H),4.17-4.11(m,4H),3.73(s,4H),3.32(s,3H),2.94-2.62(m,4H),1.14(s,9H).
实施例46
7-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺
实施方法同方法五,用片段2-c替换片段5-a,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 499.8[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.43(s,1H),9.00(s,1H),8.87(d,J=7.6Hz,1H),8.41(s,1H),7.76(s,1H),7.30-7.18(m,3H),7.00(d,J=8.8Hz,2H),6.87(s,1H),4.17-4.11(m,4H),3.95(s,3H),3.76-3.71(m,4H),3.12-2.87(m,4H),2.67(s,3H).
实施例47
7-甲基-N-(4-(1-甲基-1H-吲哚-3-基)-1H-吡唑并[3,4-d]嘧啶-6-基)-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺
实施方法同方法五,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 500.7[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.18(s,1H),9.37(s,1H),8.91(d,J=7.6Hz,1H),8.66(s,1H),8.50(s,1H),7.65-7.64(m,1H),7.56(d,J=8.0Hz,1H),7.47-7.45(m,1H),7.34-7.23(m,2H),7.00(d,J=8.8Hz,1H),4.12-4.09(m,4H),3.96(s,3H),3.71-3.64(m,4H),3.30(s,3H),2.78(brs,2H),2.63(brs,2H).
实施例48
3-环戊基-3-(4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段25-b替换片段2-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 557.8[M+H]+.
1HNMR(400MHz,DMSO-d6):δ11.51(s,1H),9.04(s,1H),8.69(s,1H),8.30(s,1H),7.74(s,1H),7.73-7.40(m,1H),7.24-7.22(m,1H),7.00-6.98(m,1H),6.80-6.79(m,1H),4.59-4.53(m,1H),4.26-4.09(m,4H),3.71(s,4H),3.31(s,3H),3.26-3.21(m,2H),2.95-2.75(m,2H),2.46-2.40(m,2H),1.85-1.81(m,1H),1.63-1.51(m,3H),1.47-1.43(m,1H),1.35-1.17(m,4H).
实施例49
2-(1-(乙基磺酰基)-3-(4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段14-a替换片段2-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 622.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.55(s,1H),9.08(s,1H),8.82(s,1H),8.39(s,1H),7.72(s,1H),7.40(d,J=8.0Hz,1H),7.26-7.25(m,1H),6.98(d,J=8.4Hz,1H),6.89(d,J=3.2Hz,1H),4.59(d,J=9.2Hz,2H),4.25(d,J=8.8Hz,2H),4.16-4.08(m,4H),3.70(s,6H),3.31(s,3H),3.28-3.22(m,2H),2.82(brs,2H),2.39(brs,2H),1.25(t,J=7.2Hz,3H).
实施例50
2-(1-(乙基磺酰基)-3-(4-(6-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-3-基)乙腈
实施方法同方法二,用片段14-a替换片段2-b,用片段4-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 623.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.37(s,1H),9.62(s,1H),8.98(s,1H),8.58(s,1H),8.47(s,1H),7.74(s,1H),7.50(d,J=8.0Hz,1H),7.09(d,J=8.0Hz,1H),4.60(d,J=9.2Hz,2H),4.44-4.26(m,4H),4.12(brs,2H),3.92-3.72(m,6H),3.32(s,3H),3.28-3.23(m,2H),2.85(brs,4H),1.26(t,J=7.6Hz,3H).
实施例51
N-(氰基甲基)-4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 528.8[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.74(s,1H),9.45(t,J=5.2Hz,1H),9.34(s,1H),8.27(d,J=8.4Hz,2H),8.09(d,J=8.4Hz,2H),7.84(s,1H),7.43(d,J=8.0Hz,1H),7.34(d,J=3.2Hz,1H),7.04(d,J=8.4Hz,1H),6.74(d,J=2.8Hz,1H),4.37-4.29(m,4H),4.13-4.10(m,2H),3.81-3.74(m,4H),3.31(s,3H),3.02-2.62(m,4H).
实施例52
N-(氰基甲基)-4-(6-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)苯甲酰胺
实施方法同方法二,用片段4-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 529.7[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.48(s,1H),9.75(s,1H),9.44(t,J=5.6Hz,1H),8.43-8.36(m,3H),8.11(d,J=8.4Hz,2H),7.71(d,J=1.6Hz,1H),7.42(d,J=8.4Hz,1H),7.03(d,J=8.4Hz,1H),4.38-4.37(m,2H),4.17-4.09(m,4H),3.71(s,4H),3.30(s,3H),2.97-2.79(m,2H),2.45-2.33(m,2H).
实施例53
N4-(2-(异丙基磺酰基)苯基)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段2-(异丙基磺酰基)苯胺替换片段1-b,用片段2-d替换片段4-a,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 567.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.45(s,1H),9.48(s,1H),8.89-8.93(m,2H),7.81-7.84(m,1H),7.72-7.76(m,1H),7.56(s,1H),7.28-7.31(m,2H),7.04-7.05(m,1H),6.92-6.94(m,1H),6.25-6.26(m,1H),4.03-4.08(m,4H),3.67-3.69(m,4H),3.44-3.48(m,1H),3.31(s,3H),2.75-2.90(m,2H),2.33-2.36(m,2H),1.17-1.19(d,J=6.8Hz,6H).
实施例54
N-(3-((5-氯-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)嘧啶-4-基)氨基)苯基)丙烯酰胺
实施方法同方法三,用片段(3-氨基苯基)氨基甲酸叔丁酯替换片段3-a,用片段1-a替换片段2-d,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 525.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.35(s,1H),9.33(s,1H),8.89(s,1H),8.13(s,1H),7.96(s,1H),7.56-7.57(m,1H),7.42(s,1H),7.20-7.29(m,3H),6.81-6.83(m,1H),6.48-6.55(m,1H),6.22-6.27(m,1H),5.73-5.76(m,1H),4.06(s,2H),3.68-3.81(m,6H),3.32(s,3H),2.91(brs,4H).
实施例55
N-(3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)丙烯酰胺
实施方法同方法三,用片段(3-氨基苯基)氨基甲酸叔丁酯替换片段3-a,用片段1-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 481.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.12(s,1H),8.91(s,1H),8.80(s,1H),8.06(s,1H),7.84(s,1H),7.49(d,J=8.0Hz,1H),7.36(d,J=7.6Hz,1H),7.29-7.25(m,1H),7.00-6.97(m,2H),6.49-6.42(m,2H),6.28-6.23(m,1H),5.76-5.74(m,1H),4.07(s,2H),3.51-3.46(m,2H),3.25(brs,7H).
实施例56
N-(3-((5-氯-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)氨基)苯基)丙烯酰胺
实施方法同方法三,用片段(3-氨基苯基)氨基甲酸叔丁酯替换片段3-a,用片段1-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 526.0[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.37(s,1H),9.34(s,1H),8.89(s,1H),8.77(s,1H),8.13(s,1H),7.97(s,1H),7.59(s,1H),7.43(s,1H),7.29(s,2H),6.56-6.48(m,1H),6.27-6.22(m,1H),5.76-5.73(m,1H),4.14-3.95(m,4H),3.77-3.69(m,4H),3.31(s,3H),2.77(m,4H).
实施例57
N-(3-((5-氯-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)嘧啶-4-基)氨基)苯基)丙酰胺
实施方法同方法三,用片段(3-氨基苯基)氨基甲酸叔丁酯替换片段3-a,用片段1-a替换片段2-d,用片段丙酰氯替换片段3-e,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 527.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.01(s,1H),9.34(s,1H),8.87(s,1H),8.13(s,1H),7.87(s,1H),7.46(s,2H),7.19-7.25(m,3H),6.86-6.88(m,1H),4.14(s,2H),3.70-3.79(m,6H),3.32(s,3H),2.50-2.79(m,4H),2.29-2.35(m,2H),1.07(t,J=7.6Hz,3H).
实施例58
N-(3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)丙酰胺
实施方法同方法三,用片段(3-氨基苯基)氨基甲酸叔丁酯替换片段3-a,用片段1-a替换片段2-d,用片段丙酰氯替换片段3-e,获得标题化合物。
LC_MS:(ES+):m/z 483.8[M+H]+.
1HNMR(400MHz,DMSO-d6):δ9.83(s,1H),8.89(s,1H),8.75(s,1H),8.06(s,1H),7.78(s,1H),7.38-7.34(m,2H),7.25-7.21(m,1H),7.02-6.97(m,2H),6.49(d,J=8.4Hz,1H),4.10-4.08(m,2H),3.48(t,J=5.4Hz,2H),3.41-3.34(m,2H),3.29-3.28(m,2H),3.25(s,3H),2.34-2.29(m,2H),1.07(t,J=7.4Hz,3H).
实施例59
N-(3-((5-氯-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)氨基)苯基)丙酰胺
实施方法同方法三,用片段(3-氨基苯基)氨基甲酸叔丁酯替换片段3-a,用片段1-a替换片段2-d,用片段丙酰氯替换片段3-e,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 527.2[M+H]+.
1HNMR(400MHz,DMSO-d6):δ9.87(s,1H),9.17(s,1H),8.86(s,1H),8.11(s,1H),7.77(s,1H),7.42-7.40(m,1H),7.32-7.25(m,3H),7.15(d,J=8.8Hz,1H),6.78(d,J=8.4Hz,1H),4.05-3.98(m,2H),3.83-3.73(m,2H),3.69-3.56(m,4H),3.33(s,3H),2.67-2.59(m,2H),2.33-2.28(m,4H),1.07(t,J=7.6Hz,3H).
实施例60
N6-(2-(异丙基磺酰基)苯基)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-9H-嘌呤-2,6-二胺
实施方法同方法四,用片段2-(异丙基磺酰基)苯胺替换片段1-b,用片段60-b替换片段4-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 568.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ12.80(s,1H),9.84(s,1H),9.14(s,1H),9.02(d,J=8.4Hz,1H),8.00(s,1H),7.82-7.84(m,1H),7.74(t,J=7.2Hz,1H),7.56(s,1H),7.28-7.32(m,2H),6.96(d,J=8.8Hz,1H),4.05-4.10(m,4H),3.70-3.73(m,4H),3.41-3.48(m,1H),3.31(s,3H),2.84(s,2H),2.33-2.41(m,2H),1.19(d,J=6.8Hz,6H).
片段60-b的合成方法如下:
步骤1:片段60-b 2,6-二氯-9-(四氢-2H-吡喃-2-基)-9H-嘌呤的合成
将含有2,6-二氯-9H-嘌呤(1g,5.32mmol),3,4-二氢-2H-吡喃(1.2g,14.36mmol),对甲苯磺酸(27mg,0.16mmol)的乙酸乙酯溶液在50℃下搅拌反应2小时。TLC监测反应完全。反应混合物冷至室温,减压浓缩,所得粗品经硅胶柱层析(用含2%乙酸乙酯的石油醚洗脱)分离纯化得到白色固体2,6-二氯-9-(四氢-2H-吡喃-2-基)-9H-嘌呤(1g,68%)。
LC_MS:(ES+):m/z 272.9[M+H]+.
1H NMR(400MHz,CDCl3):δ8.33(s,1H),5.75-5.78(m,1H),4.17-4.21(m,1H),3.75-3.82(m,1H),2.15-2.19(m,1H),2.08-2.11(m,1H),1.92-2.02(m,1H),1.66-1.85(m,3H).
实施例61
N-(2-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯基)甲磺酰胺
实施方法同方法四,用片段N-(2-氨基苯基)甲磺酰胺替换片段1-b,用片段2-d替换片段4-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 554.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.26(s,1H),9.24(s,1H),8.84(s,1H),8.73(s,1H),7.85-7.87(d,J=8Hz,1H),7.61(s,1H),7.39-7.41(d,J=8Hz,1H),7.28-7.32(t,J=8.0Hz,1H),7.18-7.23(m,2H),6.95(s,1H),6.85-6.87(d,J=8.0Hz,1H),6.43(s,1H),4.07(s,2H),3.90(s,2H),3.70(s,4H),3.28(s,3H),2.97(s,1H),2.86(s,3H),2.75(s,1H),2.42(s,2H).
实施例62
N-(4-(1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺
实施方法同方法二,用片段14-e替换片段2-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 436.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ13.30(s,1H),11.45(s,1H),8.96(s,1H),8.26-8.53(m,2H),7.72(s,1H),7.36-7.38(m,1H),7.17-7.19(m,1H),6.93(d,J=8.8Hz,1H),6.79(d,J=3.2Hz,1H),4.06-4.16(m,4H),3.66-3.76(m,4H),3.30(s,3H),2.67-2.77(m,2H),2.31-2.33(m,2H).
实施例63
7-甲基-N-(4-(1-(甲基磺酰基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺
实施方法同方法二,用片段63-a替换片段2-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 514.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.68(s,1H),9.28(s,1H),8.87(s,1H),8.70(s,1H),7.96-7.97(m,1H),7.20-7.32(m,2H),7.06-7.08(m,1H),6.90-6.91(m,1H),4.26-4.30(m,3H),4.11-4.15(m,3H),3.98-4.07(m,2H),3.79-3.83(m,2H),3.70(s,3H),3.55-3.61(m,1H),3.43-3.48(m,2H),3.19-3.23(m,1H),2.91-2.92(m,3H).
片段63-a的合成方法如下:
步骤1:片段63-a 1-(甲基磺酰基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环烷-2-基)-1H-吡唑的合成
0℃下向含有4-(4,4,5,5-四甲基-1,3,2-二氧杂硼硼烷-2-基)-1H-吡唑(300mg,1.55mmol)和三乙胺(470mg,4.65mmol)的二氯甲烷(5ml)中加入甲磺酰氯(212mg,1.86mmol)。反应混合物升至室温,搅拌反应3小时。TLC监测反应完全。反应混合物分配于水(20ml)中,二氯甲烷(10ml x 3)萃取。合并有机层,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含30%乙酸乙酯的石油醚)分离纯化得到无色油状物1-(甲基磺酰基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吡唑(200mg,47%)。
1H NMR(400MHz,DMSO-d6):δ8.35(s,1H),8.04(s,1H),3.57(s,3H),1.29(s,12H).
实施例64
N4-甲基-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段甲胺盐酸盐替换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 355.1[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.96(s,1H),8.36(s,1H),7.38(s,1H),7.22-7.26(m,1H),7.10(d,J=8.0Hz,1H),6.73(s,1H),6.60(d,J=8.8Hz,1H),6.36(s,1H),4.11-4.13(m,2H),3.51(t,J=5.6Hz,2H),3.34-3.37(m,2H),3.29(s,2H),3.26(s,3H),2.96(d,J=4.4Hz,3H).
实施例65
N4-(3-(异丙基磺酰基)苯基)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段3-(异丙基磺酰基)苯胺替换片段1-b,用片段2-d替换片段4-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 567.6[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.28(s,1H),9.53(s,1H),8.90-8.92(m,1H),8.76(s,1H),7.94-7.95(m,1H),7.57-7.61(m,2H),7.43(d,J=8.0Hz,1H),7.29-7.32(m,1H),6.92-6.96(m,2H),6.67-6.68(m,1H),4.05-4.10(m,4H),3.70-3.74(m,4H),3.36-3.43(m,1H),3.30(s,3H),2.88(brs,2H),2.39-2.46(m,2H),1.19(d,J=6.8Hz,6H).
实施例66
二甲基(3-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯基)氧化膦
实施方法同方法四,用片段(3-氨基苯基)二甲基氧化膦替换片段1-b,用片段2-d替换片段4-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 537.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.22(s,1H),9.37(s,1H),8.74-8.80(m,1H),8.46-8.52(m,1H),7.90(d,J=12.4Hz,1H),7.65-7.73(m,1H),7.44-7.48(m,1H),7.29-7.38(m,2H),6.93-6.96(m,2H),6.68(s,1H),4.14-4.18(m,2H),3.94-4.03(m,2H),3.65-3.80(m,4H),3.31(s,3H),2.74-3.08(m,4H),1.64(d,J=13.2Hz,6H).
实施例67
3-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯磺酰胺
实施方法同方法四,用片段3-氨基苯磺酰胺替换片段1-b,用片段2-d替换片段4-a,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 540.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.31(s,1H),9.52(s,1H),8.93-8.94(m,1H),8.48(s,1H),8.47(d,J=8.0Hz,1H),8.17(s,1H),7.78-7.79(m,1H),7.35-7.52(m,4H),7.03(d,J=8.8Hz,1H),6.96-6.97(m,1H),6.71-6.72(m,1H),4.25-4.30(m,2H),3.92-4.11(m,4H),3.77-3.80(m,2H),3.56(brs,1H),3.43-3.46(m,1H),3.16-3.23(m,2H),2.91(s,3H).
实施例68
N4-((1s,3s)-3-(氮杂环丁烷-1-基磺酰基)环丁基)-N4-甲基-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段68-h换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 528.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.19(s,1H),8.43(brs,1H),7.27(brs,1H),6.99-7.14(m,1H),6.81-6.92(m,1H),6.47-6.73(m,2H),5.14-5.23(m,1H),4.09-4.17(m,1H),3.87(t,J=7.6Hz,4H),3.46-3.55(m,2H),3.29-3.44(m,6H),3.17-3.25(m,6H),2.55-2.69(m,4H),2.17-2.25(m,2H).
片段68-h的合成方法如下:
步骤1:片段68-b(1r,3r)-3-(甲基氨基)环丁-1-醇的合成
在0℃向含有((1r,3r)-3-羟基环丁基)氨基甲酸叔丁酯(1g,5.34mmol)的无水四氢呋喃(15ml)溶液中滴加氢化铝锂(812mg,21.36mmol)。所得混合液在65℃下搅拌反应18小时。TLC监测反应完全。反应混合液在0℃加水(0.8ml),氢氧化钠(0.8ml,15%水溶液)和水(2.4ml)淬灭,在室温下继续搅拌反应10分钟。过滤除去固体,滤饼用含10%甲醇的二氯甲烷(20ml×3)洗涤。合并滤液,减压浓缩得到无色油状物(1r,3r)-3-(甲基氨基)环丁-1-醇(540mg,粗品),粗品直接用于下一步反应。
步骤2:片段68-c苄基((1r,3r)-3-羟基环丁基)(甲基)氨基甲酸酯的合成
在0℃下向含有粗品(1r,3r)-3-(甲基氨基)环丁-1-醇(540mg,5.34mmol)和碳酸钠(1.1g,10.7mmol)的四氢呋喃(10ml)和水(2.5ml)溶液中加入氯甲酸苄酯。所得混合物升至室温搅拌反应15小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(10ml)和水(15ml)。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,粗品经硅胶柱层析(用含20%-100%乙酸乙酯的正己烷洗脱)分离纯化得到无色油状物苄基((1r,3r)-3-羟基环丁基)(甲基)氨基甲酸酯(1.1g,87%两步)。
LC_MS:(ES+):m/z 236.1[M+H]+.
1H NMR(400MHz,DMSO-d6):δ7.31-7.39(m,5H),5.05(s,2H),4.97(d,J=4.4Hz,1H),4.71-4.80(m,1H),4.15-4.17(m,1H),2.82(s,3H),2.30-2.37(m,2H),1.95-2.01(m,2H).
步骤3:片段68-d(1r,3r)-3-(((苄氧基)羰基)(甲基)氨基)环丁基4-甲基苯磺酸的合成
在0℃下向含有苄基((1r,3r)-3-羟基环丁基)(甲基)氨基甲酸酯(1.1g,4.66mmol),三乙胺(944mg,9.32mmol),4-二甲氨基吡啶(57mg,0.47mmol)的二氯甲烷(15ml)溶液中加入4-甲苯磺酰氯(977mg,5.13mmol)。所得混合物升温至室温搅拌反应16小时。TLC监测反应完全。反应混合物加二氯甲烷(10ml)稀释,水(20ml x 2)洗涤,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含10%-15%乙酸乙酯的正己烷洗脱)分离纯化得到无色油状物(1r,3r)-3-(((苄氧基)羰基)(甲基)氨基)环丁基4-甲基苯磺酸(1.63g,89%)。
1H NMR(400MHz,CDCl3):δ7.76(d,J=8.4Hz,2H),7.31-7.36(m,7H),5.09(s,2H),4.87-4.97(m,1H),4.71-4.79(m,1H),2.84(s,3H),2.48-2.58(m,2H),2.44(s,3H),2.39-2.45(m,2H).
步骤4:片段68-e S-((1s,3s)-3-(((苄氧基)羰基)(甲基)氨基)环丁基)乙硫醇酸酯的合成
将含有(1r,3r)-3-(((苄氧基)羰基)(甲基)氨基)环丁基4-甲基苯磺酸(1.63g,4.58mmol)和硫代乙酸钾(2.62g,22.93mmol)的二甲基亚砜(16ml)溶液在60℃下搅拌反应17小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(10ml)和水(15ml)。水层乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml x 3)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含10-20%乙酸乙酯的正己烷洗脱)分离纯化得到无色油状物S-((1s,3s)-3-(((苄氧基)羰基)(甲基)氨基)环丁基)乙硫醇酸酯(1g,81%)。
LC_MS:(ES+):m/z 294.1[M+H]+.
步骤5:片段68-f苄基((1s,3s)-3-(氯磺酰基)环丁基)(甲基)氨基甲酸酯的合成
室温下将含有S-((1s,3s)-3-(((苄氧基)羰基)(甲基)氨基)环丁基)乙硫醇酸酯(1g,3.4mmol)的乙腈(5ml)溶液缓慢滴加到搅拌反应的N-氯代丁二酰亚胺(983mg,7.15mmol),醋酸(0.12ml),水(0.12ml)的乙腈(5ml)溶液中,保持稳定缓慢滴加的速度使反应液的温度不超过30℃。所得的混合物在室温下搅拌反应15分钟。TLC监测反应完全。反应混合物加乙酸乙酯(10ml)稀释,饱和碳酸氢钠溶液(20ml)洗涤,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩得到黄色油状物苄基((1s,3s)-3-(氯磺酰基)环丁基)(甲基)氨基甲酸酯(622mg,粗品),粗品不经纯化直接用于下一步反应。
步骤6:片段68-g苄基((1s,3s)-3-(氮杂环丁烷-1-基磺酰基)环丁基)(甲基)氨基甲酸酯的合成
将含有粗品苄基((1s,3s)-3-(氯磺酰基)环丁基)(甲基)氨基甲酸酯(622mg,1.96mmol),杂氮环丁烷(112mg,1.96mmol),N,N-二异丙基乙胺(505mg,3.91mmol)的二氯甲烷(3ml)溶液室温下搅拌反应16小时。TLC监测反应完全。反应混合物加二氯甲烷(15ml)稀释,水(10ml x 2)洗,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含1%-1.5%甲醇的二氯甲烷洗脱)分离纯化得到无色油状物苄基((1s,3s)-3-(氮杂环丁烷-1-基磺酰基)环丁基)(甲基)氨基甲酸酯(120mg,10%两步)。
1H NMR(400MHz,CDCl3):δ7.32-7.37(m,5H),5.12(s,2H),4.69-4.81(m,1H),3.96(t,J=7.6Hz,4H),3.33-3.39(m,1H),2.94(s,1H),2.57-2.65(m,2H),2.42-2.53(m,2H),2.22-2.30(m,2H).
步骤7:片段68-h(1s,3s)-3-(氮杂环丁烷-1-基磺酰基)-N-甲基环丁-1-胺的合成
室温氢气(氢气球)氛围下将含有苄基((1s,3s)-3-(氮杂环丁烷-1-基磺酰基)环丁基)(甲基)氨基甲酸酯(90mg,0.26mmol)和钯/碳(100%,90mg)的甲醇(4ml)溶液搅拌反应24小时。TLC监测反应完全。过滤除去钯/碳,甲醇(5ml x 2)洗涤。合并滤液,减压浓缩得到褐色油状物(1s,3s)-3-(氮杂环丁烷-1-基磺酰基)-N-甲基环丁-1-胺(55mg,粗品),粗品不经纯化直接用于下一步反应。
实施例69
4-甲基-N-(4-(1-(1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺
实施方式:
步骤1:片段69-b 3-(甲苯磺酰氧基)氮杂环丁烷-1-羧酸叔丁酯的合成
0℃下向含有3-羟基氮杂环丁烷-1-羧酸叔丁酯(500mg,2.9mmol),三乙胺(862.4mg,5.78mmol),4-二甲氨基吡啶(36.7mg,0.3mmol)的二氯甲烷(5ml)溶液中加入4-甲苯磺酰氯(606.4mg,3.2mmol),反应混合物在室温下搅拌反应12小时。TLC监测反应完全。反应混合物分配于二氯甲烷(20ml)和水(20ml)。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含30%乙酸乙酯的正己烷洗脱)分离纯化得到无色油状物3-(甲苯磺酰氧基)氮杂环丁烷-1-羧酸叔丁酯(700mg,74%)。
1H NMR(400MHz,CDCl3):δ7.78-7.80(m,2H),7.37(d,J=8.0Hz,2H),4.98-5.03(m,1H),4.08-4.11(m,2H),3.87-3.97(m,2H),2.47(s,3H),1.41(s,9H).
步骤2:片段69-c 2-氯-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶的合成
室温氮气氛围下向含有2,4-二氯-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(500mg,1.57mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼硼烷-2-基)-1H-吡唑(304.6mg,1.57mmol),碳酸钠(2N,1ml)溶液和乙腈(5ml)中加入四(三苯基膦)钯(184.8mg,0.16mmol),反应混合物氮气置换三次,55℃下搅拌反应12小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(20ml)和水(20ml)中。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含50%乙酸乙酯的正己烷洗脱)分离纯化得到灰白色固体2-氯-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(200mg,36%)。
LC_MS:(ES+):m/z 350.4[M+H]+.
步骤3:片段69-d叔丁基3-(4-(2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-羧酸的合成
氮气氛围下将含有2-氯-4-(1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(200mg,0.57mmol),3-(甲苯磺酰氧基)氮杂环丁烷-1-羧酸叔丁酯(282mg,0.86mmol),碳酸铯(371mg,1.14mmol)的N,N-二甲基甲酰胺(2ml)溶液60℃下搅拌反应13小时。TLC监测反应完全。反应混合物冷却至 室温,分配于水(20ml),乙酸乙酯(20ml x 2)萃取。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含2%甲醇的二氯甲烷洗脱)分离纯化得到白色固体叔丁基3-(4-(2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-羧酸(200mg,65%)。
步骤4:片段69-e叔丁基3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-甲酸的合成
室温氮气氛围下向含有3-(4-(2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-羧酸(100mg,0.2mmol),4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(42mg,0.2mmol),磷酸钾(127mg,0.6mmol)的1,4-二氧六环(1ml)溶液中加入BrettPhos Pd G3(18mg,0.02mmol),反应混合物氮气置换三次,110℃下搅拌反应13小时。TLC监测反应完全。反应混合物冷至室温,分配于水(20ml),乙酸乙酯(10ml x 2)萃取。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含4%甲醇和38%乙酸乙酯的正己烷洗脱)分离纯化得到黄色固体叔丁基3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-甲酸(100mg,74%)。
LC_MS:(ES+):m/z 677.7[M+H]+.
步骤5:片段69-f N-(4-(1-(氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
室温下向含有叔丁基3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-甲酸(100mg,0.15mmol)的二氯甲烷(1ml)溶液中加入盐酸/二氧六环(4M,1ml)溶液,反应液在室温下搅拌反应1小时。TLC监测反应完全。减压移除挥发物,剩余物用碳酸钠溶液碱化至pH=7-8,二氯甲烷(10ml x 2)萃取。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇的二氯甲烷洗脱)分离纯化得到黄色固体N-(4-(1-(氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(62mg,70%)。
LC_MS:(ES+):m/z 577.6[M+H]+.
步骤6:片段69-g 4-甲基-N-(4-(1-(1-(1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
0℃下向含有N-(4-(1-(氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(62mg,0.11mmol)和三乙胺(16.2mg,0.16mmol)的二氯 甲烷(2ml)溶液中加入甲基磺酰氯(12.6mg,0.11mmol),所得混合物升温至室温搅拌反应3小时。TLC监测反应完全。反应混合物分配于二氯甲烷(10ml)和水(10ml)。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含5%甲醇的二氯甲烷洗脱)分离纯化得到黄色固体4-甲基-N-(4-(1-(1-(1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(60mg,80%)。
LC_MS:(ES+):m/z 655.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.96(s,1H),8.69(s,1H),8.35(s,1H),7.41(d,J=2.0Hz,1H),7.33-7.34(m,1H),7.28-7.30(m,1H),6.87(d,J=3.6Hz,1H),6.65(d,J=8.8Hz,1H),5.50(s,2H),5.39-5.46(m,1H),4.32-4.39(m,4H),4.13-4.14(m,2H),3.51-3.57(m,4H),3.27-3.38(m,4H),3.27(s,3H),3.16(s,3H),0.84-0.89(m,2H),-0.11(s,9H).
步骤7:4-甲基-N-(4-(1-(1-(甲基磺酰基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺的合成
将含有N-(4-(1-(氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(60mg,0.13mmol)和四丁基氟化铵(0.5ml,1M四氢呋喃溶液)的四氢呋喃(0.5ml)溶液50℃下搅拌反应48小时。TLC监测反应完全。反应混合液冷却至室温,分配于水(10ml)和乙酸乙酯(10ml)。收集有机层,水层乙酸乙酯(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇和1%氨水的二氯甲烷洗脱)分离纯化得到4-甲基-N-(4-(1-(1-(甲基磺酰 基)氮杂环丁烷-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-基)-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-胺(10mg,14%)。
LC_MS:(ES+):m/z 525.2[M+H]+.
1H NMR(400MHz,CDCl3):δ8.83(s,1H),8.28-7.32(m,2H),6.92-7.02(m,2H),6.59-6.66(m,2H),5.19-5.22(m,1H),4.43-4.51(m,4H),4.24(s,2H),3.60(t,J=5.6Hz,2H),3.41-3.53(m,4H),3.37(s,3H),3.07(s,3H).
实施例70
1-(3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H)-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-基)乙烷-1-酮
实施方式:
步骤1:1-(3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁-1-基)乙酮的合成
将含有叔丁基3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁烷-1-甲酸(56mg,0.08mmol)和三氟乙酸(1ml)的二氯甲烷(2ml)溶液在室温下搅拌反应4小时。TLC监测反应完全。反应混合物减压浓缩,剩余物加到氨水(3ml)和乙酸乙酯(2ml)溶液,所得混合物在30℃下搅拌反应16小时。TLC监测反应完全。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇的二氯甲烷洗脱)分离纯化得到黄色固体1-(3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)氮杂环丁-1-基)乙酮(11mg,27%)。
LC_MS:(ES+):m/z 489.3[M+H]+.
1H NMR(400MHz,CDCl3):δ8.81(brs,1H),8.28(s,2H),7.28(s,1H),6.93-7.02(m,2H),6.57-6.67(m,2H),5.17-5.23(m,1H),4.59-4.67(m,2H),4.45-4.56(m,2H),4.24(s,2H),3.58-3.61(m,2H),3.37-3.44(m,6H),1.97(s,3H).
实施例71
(1s,3s)-3-氨基-N-(3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)环丁烷-1-甲酰胺
实施方式:
步骤1:片段71-c叔丁基((1s,3s)-3-((3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)氨基甲酰基)环丁基)氨基甲酸酯的合成
在0℃下向含有N4-(3-氨基苯基)-5-氯-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)嘧啶-2,4-二胺(30mg,0.07mmol),(1s,3s)-3-((叔丁氧羰基)氨基)环丁烷羧酸(15mg,0.07mmol),N,N-二异丙基乙胺(36mg,0.28mmol)的N,N-二甲基甲酰胺(1ml)溶液中加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(53mg,0.14mmol);所得混合物室温下搅拌反应30分钟。TLC监测反应完全。反应混合物分配于水(10ml)和乙酸乙酯(10ml)。收集有机层,饱和氯化钠溶液(20ml x 2)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含5%甲醇的二氯甲烷洗脱)分离纯化得到白色固体叔丁基((1s,3s)-3-((3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑酮-9-基)氨基)嘧啶-4-基)氨基)苯基)氨基甲酰基)环丁基)氨基甲酸酯(35mg,79%)。
LC_MS:(ES+):m/z 624.4[M+H]+.
步骤2:(1s,3s)-3-氨基-N-(3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)环丁烷甲酰胺的合成
将含有叔丁基((1s,3s)-3-((3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)氨基甲酰基)环丁基)氨基甲酸酯(35mg,0.056mmol)的4M盐酸/二氧六环(1ml)和二氯甲烷(1ml)室温下搅拌反应3小时。TLC监测反应完全。减压浓缩,剩余物用碳酸钠溶液调节pH=8~9,然后用二氯甲烷(10ml×2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇和1%氨水的二氯甲烷洗脱)分离纯化得到灰色固体(1s,3s)-3-氨基-N-(3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)环丁烷甲酰胺(20.4mg,69%)。
LC_MS:(ES+):m/z 524.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.90(s,1H),8.94(s,1H),8.71(s,1H),8.06(s,1H),7.86(s,1H),7.34(d,J=8.0Hz,1H),7.34(d,J=7.2Hz,1H),7.21-7.25(m,1H),7.00-7.03(m,2H),6.49(d,J=8.4Hz,1H),5.10(s,2H),4.08(t,J=4.0Hz,2H),3.49(t,J=5.6Hz,2H),3.34-3.41(m,4H),3.27-3.29(m,1H),3.26(s,3H),2.82-2.86(m,1H),2.32-2.39(m,2H),1.98-2.06(m,2H).
实施例72
(1r,3r)-3-氨基-N-(3-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氨基)苯基)环丁烷-1-甲酰胺
实施方式同实施例71,用(1r,3r)-3-((叔丁氧羰基)氨基)环丁烷羧酸替换片段71-b获得标题化合物。
LC_MS:(ES+):m/z 524.7[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.90(s,1H),8.94(s,1H),8.71(s,1H),8.06(s,1H),7.86(s,1H),7.34(d,J=8.0Hz,1H),7.34(d,J=7.2Hz,1H),7.21-7.25(m,1H),7.00-7.03(m,2H),6.49(d,J=8.4Hz,1H),5.10(s,2H),4.08(t,J=4.0Hz,2H),3.49(t,J=5.6Hz,2H),3.34-3.41(m,4H),3.27-3.29(m,1H),3.26(s,3H),2.82-2.86(m,1H),2.32-2.39(m,2H),1.98-2.06(m,2H).
实施例73
二甲基(2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯基)氧化膦
实施方法同方法四,用片段(2-氨基苯基)二甲基氧化膦替换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 493.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.59(s,1H),11.20(s,1H),9.14-9.11(m,1H),8.57(s,1H),7.57-7.46(m,1H),7.32(s,1H),7.07-7.05(m,2H),6.88(s,1H),6.65-6.64(m,1H),6.34(s,1H),4.15(s,2H),3.55-3.53(m,2H),3.38-3.32(m,4H),3.28(s,3H),1.83(d,J=13.6Hz,6H).
实施例74
(R)-3-环戊基-3-(4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段74-a(R)-3-环戊基-3-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丙腈(上海毕得医药科技股份有限公司)替换片段2-b,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 557.7[M+H]+.
1H NMR(400MHz,CDCl3):δ9.31(s,1H),8.27(s,2H),7.49(s,1H),7.11-7.07(m,2H),6.95-6.92(m,2H),4.27-4.25(m,1H),4.19-4.14(m,4H),3.80-3.75(m,4H),3.16-3.10(m,1H),2.97-2.92(m,1H),2.82(d,J=8.0Hz,4H),2.58(m,1H),2.42(s,3H),1.97(m,2H),1.72(m,2H),1.56(m,2H),1.26(m,2H).
实施例75
(S)-3-环戊基-3-(4-(2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-13-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段75-a(S)-3-环戊基-3-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丙腈(上海毕得医药科技股份有限公司)替换片段2-b,片段1-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 557.7[M+H]+.
1H NMR(400MHz,CDCl3):δ9.06(s,1H),8.27(d,J=1.8Hz,2H),7.49(s,1H),7.12-7.09(m,1H),7.02(s,1H),6.96-6.94(m,2H),6.61(m,1H),4.27-4.25(m,1H),4.20-4.15(m,4H),3.82-3.77(m,4H),3.16-3.10(m,1H),2.97-2.92(m,1H),2.85(d,J=16.2Hz,4H),2.60-2.58(m,1H),2.44(s,3H),1.98-1.96(m,1H),1.73-1.68(m,2H),1.58-1.56(m,2H),1.33-1.26(m,2H).
实施例76
(R)-3-环戊基-3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段74-a替换片段2-b,获得标题化合物。
LC_MS:(ES+):m/z 513.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.42(s,1H),8.72(s,1H),8.66(s,1H),8.26(s,1H),7.40(d,J=2.8Hz,1H),7.20-7.15(m,2H),6.75-6.74(m,1H),6.65(d,J=8.8Hz,1H),4.58-4.52(m,1H),4.15(d,J=4.4Hz,2H),3.53(t,J=5.6Hz,2H),3.38(t,J=5.6Hz,2H),3.29(s,2H),3.27(s,3H),3.22-3.20(m,1H),2.46-2.39(m,1H),1.83-1.80(m,1H),1.63-1.43(m,4H),1.36-1.19(m,4H).
实施例77
(S)-3-环戊基-3-(4-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡唑-1-基)丙腈
实施方法同方法二,用片段75-a替换片段2-b,获得标题化合物。
LC_MS:(ES+):m/z 513.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.42(s,1H),8.72(s,1H),8.66(s,1H),8.26(s,1H),7.40-7.39(m,1H),7.20-7.15(m,2H),6.75-6.74(m,1H),6.65(d,J=8.8Hz,1H),4.57-4.52(m,1H),4.15(d,J=4.0Hz,2H),3.53(t,J=5.6Hz,2H),3.38(t,J=5.6Hz,2H),3.29(s,2H),3.27(s,3H),3.25-3.20(m,1H),2.46-2.39(m,1H),1.86-1.79(m,1H),1.63-1.51(m,3H),1.46-1.43(m,1H),1.36-1.28(m,4H).
实施例78
二甲基(2-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)苯基)氧化膦
实施方法同方法七,用片段(2-氨基苯基)二甲基氧化膦替换片段6-d,获得标题化合物。
LC_MS:(ES+):m/z 566.20[M+H]+.
1H NMR(400MHz,CD3OD):δ8.34(s,1H),8.06-7.92(m,1H),7.76-7.71(m,1H),7.62(t,J=7.8Hz,1H),7.42-7.31(m,2H),7.12-7.09(m,1H),6.94(d,J=8.4Hz, 1H),4.19-4.08(m,4H),3.96-3.79(m,4H),3.52-3.34(m,4H),2.92(s,3H),1.79(d,J=13.6Hz,6H).
实施例79
环丙基((1R,5S)-3-(2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3,8-二氮杂双环[3.2.1]辛烷-8-基)甲酮
实施方法同方法四,用片段79-d替换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 504.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.31(s,1H),8.32(brs,1H),7.27(s,1H),7.11-7.06(m,1H),6.84(s,1H),6.61(brs,1H),6.44-6.43(m,1H),4.76-4.63(m,2H),4.54-4.41(m,2H),4.13(s,2H),3.52(t,J=5.2Hz,2H),3.32(brs,5H),3.26(s,3H),3.21-3.16(m,1H),2.04-1.96(m,2H),1.80-1.77(m,2H),1.71-1.69(m,1H),0.79-0.72(m,4H).
片段79-d实施方式如下:
步骤1:片段79-c 8-(环丙烷羰基)-3,8-二氮杂双环[3.2.1]辛烷-3-羧酸叔丁酯的合成
在0℃下向含有3,8-二氮杂双环[3.2.1]辛烷-3-羧酸叔丁酯(500mg,2.35mmol),环丙烷羧酸(203mg,2.35mmol),N,N-二异丙基乙胺(1.22g,9.44mmol)的N,N-二甲基甲酰胺的搅拌反应溶液中加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.79g,4.72mmol);所得混合物升温至室温搅拌反应30分钟。TLC监测反应完全。反应混合物分配于水(20ml)和乙酸乙酯(20ml)。收集有机层,饱和氯化钠溶液(10ml x 3)洗涤,无水硫酸钠干燥,减压浓缩得到黄色油状物8-(环丙烷羰基)-3,8-二氮杂双环[3.2.1]辛烷-3-羧酸叔丁酯(600mg,粗品),粗品不经纯化直接用于下一步反应。
步骤2:片段79-d 3,8-二氮杂双环[3.2.1]辛-8-基(环丙基)甲酮的合成
将含有8-(环丙烷羰基)-3,8-二氮杂双环[3.2.1]辛烷-3-羧酸叔丁酯(600mg,2.14mmol)和三氟乙酸(1.5ml)的二氯甲烷(3ml)溶液在室温下搅拌反应5小时。TLC监测反应完全。加压移除挥发物,剩余物用碳酸钠溶液调节pH值至7-8,然后用二氯甲烷(10ml×3)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,得到黄色油状物3,8-二氮杂双环[3.2.1]辛-8-基(环丙基)甲酮(350mg,粗品)。
实施例80
N4-(2-甲氧基-3-(1-甲基-1H-1,2,4-三唑-3-基)苯基)-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段80-f:2-甲氧基-3-(1-甲基-1H-1,2,4-三唑-3-基)苯胺替换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 528.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.14(s,1H),8.54(s,1H),8.34-8.38(m,1H),8.26(d,J=4.0Hz,1H),7.55(d,J=3.8Hz,1H),7.27(s,1H),7.19(t,J=8.0Hz,1H),7.09(s,1H),6.86(s,1H),6.56-6.58(m,1H),6.51(s,1H),4.13(s,2H),3.95(s,3H),3.72(s,3H),3.50-3.53(m,2H),3.36(s,2H),3.30(s,2H),3.27(s,3H).
片段80-f实施方式如下:
步骤1:片段80-b 2-甲氧基-3-硝基苯甲酸甲酯
将含有3-硝基水杨酸甲酯(2g,10.15mmol),碘甲烷(2.9g,20.29mmol),碳酸钾(2.8g,20.29mmol)的N,N-二甲基甲酰胺(20ml)溶液在60℃下搅拌反应1小时。TLC监测反应完全。反应混合物加冰水(20ml)淬灭,然后乙酸乙酯(10ml x 2)萃取。有机层饱和氯化钠溶液(30ml)洗涤,无水硫酸钠干燥,减压浓缩,得到淡黄色固体2-甲氧基-3-硝基苯甲酸甲酯(2.09g,粗品)。
LC_MS:(ES+):m/z 212.0[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.11-8.14(m,1H),8.03-8.05(m,1H),7.44(t,J=4.0Hz,1H),3.89(d,J=7.2Hz,6H).
步骤2:片段80-c 2-甲氧基-3-硝基苯甲酰胺
在0℃下向含有2-甲氧基-3-硝基苯甲酸甲酯(2.09g,9.90mmol)的氢氧化铵(20ml)溶液中加入甲胺醇溶液(35ml);反应混合物室温下搅拌反应15小时。TLC监测反应完全。反应混合物减压浓缩得到黄色固体2-甲氧基-3-硝基苯甲酰胺(2g,粗品)。
LC_MS:(ES+):m/z 196.9[M+H]+.
1H NMR(400MHz,DMSO-d6):δ7.95-7.98(m,2H),7.76-7.78(m,2H),7.37(t,J=4.0Hz,1H),3.88(s,3H).
步骤3:片段80-d 3-(2-甲氧基-3-硝基苯基)-1H-1,2,4-三唑
将含有2-甲氧基-3-硝基苯甲酰胺(2g,10mmol)的N,N-二甲基甲酰胺二甲基缩醛(11.9g,100mmol)溶液在90℃下搅拌反应30分钟。TLC监测反应完全。减压浓缩移除大量的N,N-二甲基甲酰胺二甲基缩醛,剩余物溶于乙醇(10ml),然后在0℃下滴加含有水合肼(6.25g,100mmol)的乙醇(50ml)和乙酸(12ml)混合溶液。所得混合物升温至室温搅拌反应4小时。TLC监测反应完全。反应混合物浓缩,剩余物分配于水(50ml)和乙酸乙酯(20ml)。水层乙酸乙酯(10ml x 3)洗涤。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品在室温下加到30ml 2%乙酸乙酯的石油醚溶液中搅拌反应3小时,过滤,收集固体,真空干燥得到黄色固体3-(2-甲氧基-3-硝基苯基)-1H-1,2,4-三唑(1.56g,71%)。
LC_MS:(ES+):m/z 221.1[M+H]+.
步骤4:片段80-e 3-(2-甲氧基-3-硝基苯基)-1-甲基-1H-1,2,4-三唑
将含有3-(2-甲氧基-3-硝基苯基)-1H-1,2,4-三氮唑(1.56g,7.085mmol),碳酸钾(1.31g,9.49mmol),碘甲烷(1.31g,9.21mmol)的N,N-二甲基甲酰胺(10ml)溶液在室温下搅拌反应3小时。TLC监测反应完全。反应混合物分配于乙酸乙酯(30ml)和水(30ml)。收集有机层,无水硫酸钠干燥,减压浓缩,所得粗品加到乙醇(3ml)中并在45℃下搅拌反应直至固体消失。混合物冷却至室温;过滤收集固体,滤饼用-30℃乙醇(2ml)洗涤,真空干燥,得到黄色固体3-(2-甲氧基-3-硝基苯基)-1-甲基-1H-1,2,4-三唑(255mg,15%)。
LC_MS:(ES+):m/z 235.1[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.53(s,1H),8.12-8.14(m,1H),7.86-7.88(m,1H),7.40(t,J=4.0Hz,1H),4.05(s,3H),3.85(s,3H).
步骤5:片段80-f 2-甲氧基-3-(1-甲基-1H-1,2,4-三唑-3-基)苯胺
室温氢气(氢气球)氛围下将含有3-(2-甲氧基-3-硝基苯基)-1-甲基-1H-1,2,4-三氮唑(255mg,1.08mmol)和钯/碳(10%,100mg)的乙醇(5ml)在室温下搅拌反应1小时。TLC监测反应完全。过滤除去钯/碳,乙醇(5ml x 2)洗涤。合并滤液,减压浓缩,所得粗品经硅胶柱层析(用含3%甲醇的二氯甲烷洗脱)分离纯化得到灰白色固体2-甲氧基-3-(1-甲基-1H-1,2,4-三唑-3-基)苯胺(140mg,63%)。
LC_MS:(ES+):m/z 205.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.08(s,1H),7.33-7.35(m,1H),6.99(t,J=3.8Hz,1H),6.80-6.82(m,1H),3.99(s,3H),3.77(s,3H).
实施例81
N4-乙基-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法八,用片段乙胺盐酸盐替换片段8-b,获得标题化合物。
LC_MS:(ES+):m/z 398.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.10-8.17(m,1H),7.18-7.23(m,1H),6.96-7.10(m,2H),6.84-6.86(m,1H),6.63-6.66(m,1H),4.13(t,J=4.4Hz,2H),3.52(t,J= 5.6Hz,2H),3.41(t,J=5.4Hz,2H),3.34-3.36(m,2H),3.26(s,3H),3.10-3.26(m,2H),1.05(t,J=7.0Hz,3H).
实施例82
N4-(2-(异丙基磺酰基)苯基)-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法八,用片段2-(异丙基磺酰基)苯胺替换片段8-b,获得标题化合物。
LC_MS:(ES+):m/z 552.1[M+H]+.
1H NMR(400MHz,CDCl3):δ9.34(s,1H).8.42-8.44(d,J=4.2Hz,1H),8.32(s,1H),7.89-7.91(d,J=4.0Hz,1H),7.58(t,J=3.8Hz,1H),7.00(s,1H),6.83-6.86(d,J=4.4Hz,1H),6.59-6.61(d,J=4.2Hz,1H),4.23(t,J=2.2Hz,2H),3.60(t,J=3.0Hz,2H),3.42-3.46(m,4H),3.37(s,1H),3.16-3.23(m,1H),1.28-1.30(d,J=3.4Hz,6H).
实施例83
二甲基(2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)苯基)氧化膦
实施方法同方法八,用片段(2-氨基苯基)二甲基氧化膦替换片段8-b,获得标题化合物。
LC_MS:(ES+):m/z 522.30[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.62(s,1H),9.43(s,1H),8.34(s,1H),7.90-7.92(m,1H),7.55-7.57(m,1H),7.49(s,1H),7.19(t,J=8.0Hz,1H),6.93(s,2H),6.59(d,J=8.8Hz,1H),4.13(s,2H),3.51(t,J=5.6Hz,2H),3.39(t,J=5.6Hz,2H),3.30(s,2H),3.26(s,3H),1.74(d,J=13.2Hz,6H),
实施例84
N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-N4-(2-(甲基磺酰基)乙基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方法同方法四,用片段2-(甲基磺酰基)乙烷-1-胺替换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 447.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.96(s,1H),8.25(s,1H),7.41(s,1H),7.30(s,1H),7.11(s,1H),6.74(s,1H),6.61(s,1H),6.34(s,1H),4.12(s,2H),3.81(d,J=5.2Hz,2H),3.49(d,J=6.1Hz,5H),3.26(s,6H),3.02(s,3H).
实施例85
二甲基(2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)乙基)氧化膦
实施方法同方法四,用片段(2-氨基乙基)二甲基氧化膦替换片段1-b,用片段2-d替换片段4-a,获得标题化合物。
LC_MS:(ES+):m/z 445.3[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.92(s,1H),8.23(s,1H),7.39-7.27(m,2H),7.10-7.08(m,1H),6.76-6.69(m,1H),6.61-6.58(m,1H),6.35(brs,1H),4.18-4.06(m,2H),3.75-3.63(m,2H),3.55-3.48(m,2H),3.29-3.23(m,4H),2.12-2.06(m,2H),1.76(s,3H),1.43(d,J=12.8Hz,6H).
实施例86
二甲基(2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)乙基)氧化膦
实施方法同方法八,用片段(2-氨基乙基)二甲基氧化膦替换片段8-b,获得标题化合物。
LC_MS:(ES+):m/z 474.2[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.29(s,1H),8.12(s,1H),7.21(s,1H),7.11-7.07(m,2H),6.64(d,J=8.8Hz,1H),4.12(t,J=4.4Hz,2H),3.73-3.65(m,2H),3.50(t,J=5.2Hz,2H),3.37(t,J=5.6Hz,2H),3.32-3.30(m,2H),3.26(s,3H),2.07-2.00(m,2H),1.40(d,J=12.8Hz,6H).
实施例87
(1r,4r)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)环己-1-醇
实施方法同方法六,用片段6-d替换片段6-e,获得标题化合物。
LC_MS:(ES+):m/z 498.20[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.31(s,1H),8.87(s,1H),7.60(m,1H),7.32-7.30(m,1H),6.95-6.91(m,1H),6.27-6.26(m,1H),5.37(m,1H),4.25(s,2H),4.10-4.06(m,4H),3.68(m,4H),3.00-2.64(m,4H),2.35-2.33(m,3H),2.03-1.96(m,2H),1.76-1.63(m,4H),1.51-1.45(m,2H),1.19(s,3H).
实施例88
(S)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)-N4-(氧杂环丁烷-2-基甲基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法八,用片段(S)-氧杂环丁烷-2-基甲胺替换片段8-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 484.30[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.60(s,1H),8.95(s,1H),8.19(s,1H),7.62(s,1H),7.37-7.27(m,2H),7.16-7.05(m,2H),4.95-4.92(m,1H),4.52-4.41(m,2H),4.29-4.02(m,4H),3.85-3.62(m,6H),3.51-3.44(m,1H),3.33-3.18(m,1H),2.88(s,3H),2.63-2.57(m,1H),2.41(brs,1H).
实施例89
N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷基-12-基)-N4-(2-(甲基磺酰基)乙基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法八,用片段2-(甲基磺酰基)乙烷-1-胺替换片段8-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 520.20[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.64(s,1H),8.99(s,1H),8.22(s,1H),7.52(s,1H),7.38(brs,1H),7.25-7.08(m,1H),4.31(s,2H),4.16-3.83(m,6H),3.70-3.47(m,6H),3.18(brs,2H),3.02(s,3H),2.90(s,3H).
实施例90
2-甲基-1-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)丙-2-醇
实施方法同方法三,用片段1-氨基-2-甲基丙-2-醇替换片段3-a,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 457.25[M+H]+.
1H NMR(400MHz,DMSO-d6):δ10.93(s,1H),8.43(s,1H),7.70(d,J=2.4Hz,1H),7.25-7.23(m,1H),6.95(brs,1H),6.86(d,J=8.8Hz,1H),6.76(t,J=2.0Hz,1H),6.50-6.49(m,1H),4.69(s,1H),4.08-4.03(m,4H),3.67-3.62(m,4H),3.50(d,J =5.6Hz,2H),2.76(t,J=4.4Hz,2H),2.61(t,J=4.4Hz,2H),2.28(s,3H),1.16(s,6H).
实施例91
N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-N4-苯基-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法七,用片段苯胺替换片段6-d,用片段Amine-1替换片段Amine-3,获得标题化合物。
LC_MS:(ES+):m/z 446.1[M+H]+.
实施例92
(S)-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-N4-(氧杂环丁烷-2-基甲基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法八,用片段(S)-氧杂环丁烷-2-基甲胺替换片段8-b,获得标题化合物。
LC_MS:(ES+):m/z 440.0[M+H]+.
实施例93
(1s,4s)-1-甲基-4-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氧基)环己-1-醇
实施方法同方法七,用片段Amine-1替换片段Amine-3,获得标题化合物。
LC_MS:(ES+):m/z 483.20[M+H]+.
1H NMR(400MHz,CDCl3):δ8.29(s,1H),7.16(s,1H),6.85(d,J=8.4Hz,1H),6.62(d,J=8.8Hz,1H),5.15-5.12(m,1H),4.22(t,J=4.4Hz,2H),3.59(t,J=5.6Hz,2H),3.46-3.40(m,4H),3.37(s,3H)1.97-1.92(m,4H),1.79-1.76(m,3H),1.63-1.56(m,2H),1.28(s,3H).
实施例94
N4-(2-(甲氧基甲基)苯基)-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法七,用片段2-(甲氧基甲基)苯胺替换片段6-d,用片段Amine-1替换片段Amine-3,获得标题化合物。
LC_MS:(ES+):m/z 490.1[M+H]+.
1H NMR(400MHz,CDCl3):δ8.50(s,1H),8.27(s,1H),8.19(d,J=8.2Hz,1H),7.33(t,J=7.8Hz,1H),7.22(dd,J=7.6,1.7Hz,1H),7.08(td,J=7.5,1.2Hz,1H),7.00(d,J=2.5Hz,1H),6.91(dd,J=8.6,2.5Hz,2H),6.58(d,J=8.7Hz,1H),4.49(s,2H),4.28–4.16(m,2H),3.59(t,J=5.7Hz,2H),3.47–3.39(m,7H),3.36(s,3H).
实施例95
N4-(2-甲氧基苯基)-N2-(4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法七,用片段2-甲氧基苯胺替换片段6-d,用片段Amine-1替换片段Amine-3,获得标题化合物。
LC_MS:(ES+):m/z 476.1[M+H]+.
1H NMR(400MHz,CDCl3):δ8.31(d,J=8.0Hz,1H),8.16(s,1H),7.14–7.05(m,1H),7.01(d,J=2.5Hz,1H),6.95–6.88(m,3H),6.62(d,J=8.7Hz,1H),4.27–4.21(m,2H),3.92(s,3H),3.61(t,J=5.6Hz,2H),3.46(dt,J=8.8,4.9Hz,4H),3.38(s,3H).
实施例96
1-(((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)甲基)环丙烷-1-甲腈
实施方法同方法八,用片段1-(氨基甲基)环丙烷-1-甲腈替换片段8-b,获得标题化合物。
LC_MS:(ES+):m/z 449.1[M+H]+.
1H NMR(400MHz,CDCl3):δ8.11(s,1H),7.00(d,J=2.5Hz,1H),6.82(d,J=8.6Hz,1H),6.61(d,J=8.7Hz,1H),4.27–4.17(m,2H),3.72(d,J=6.0Hz,2H),3.58 (t,J=5.6Hz,2H),3.47–3.39(m,4H),3.36(s,3H),1.25(d,J=2.0Hz,2H),1.05(q,J=5.1Hz,2H).
实施例97
N2-(2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-N4-(2-(异丙基磺酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方式:
步骤1:中间体97-a 13-硝基-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯的合成
将含有13-硝基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷(140mg,0.52mmol),二碳酸二叔丁酯(170mg,0.78mmol),N,N-二甲基吡啶-4- 胺(6mg,0.05mmol)和三乙胺(105mg,1.0mmol)的二氯甲烷(2ml)溶液在室温下搅拌反应12小时。TLC监测反应完全。反应混合物分配于水(20ml)和二氯甲烷(10ml)中。收集有机层,水相用二氯甲烷(10ml x 2)萃取。合并有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经硅胶柱层析(用含1%甲醇的二氯甲烷洗脱)分离纯化得到白色固体13-硝基-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯(146mg,76%)。
步骤2:中间体97-b 13-氨基-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯的合成
将含有13-硝基-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯(146mg,0.4mmol),铁粉(224mg,4.0mmol)和氯化铵(107mg,2.0mmol)和水(0.2ml)的乙醇(2ml)溶液回流反应12小时。TLC监测反应完全。反应混合物过滤,减压浓缩滤液得到黑色油状物13-氨基-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯(100mg,73%),未经进一步纯化,直接用于下一步。
步骤3:中间体97-c 13-((4-((2-(异丙基磺酰基)苯基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)氨基)-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯的合成
室温氮气氛围下向含有2-氯-N-(2-(异丙基磺酰基)苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-胺(71.1mg,0.15mmol),13-氨基 -2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯(50mg,0.15mmol),磷酸钾(95.4mg,0.45mmol)的1,4-二氧六环(1ml)中加入BrettPhos Pd G3(13.6mg,0.015mmol),反应混合物氮气置换三次,110℃下搅拌反应12小时。TLC监测反应完全。反应混合物冷至室温,分配于水(20ml)和乙酸乙酯(10ml)中。收集有机层,水相用乙酸乙酯(10ml x 2)萃取。收集有机层,饱和氯化钠溶液(20ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含3%甲醇的二氯甲烷洗脱)分离纯化得到黄色固体13-((4-((2-(异丙基磺酰基)苯基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)氨基)-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯(70mg,59%)。
LC_MS:(ES+):m/z 783.7[M+H]+.
步骤4:N2-(2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基-N4-(2-(异丙基磺酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺的合成
将含有13-((4-((2-(异丙基磺酰基)苯基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-基)氨基)-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-羧酸叔丁酯(70mg,0.09mmol)和三氟乙酸(2ml)的二氯甲烷(2ml)溶液在室温下搅拌反应3小时。TLC监测反应完全。浓缩反应液,剩余物加入到氨水(2ml)和乙酸乙酯(4ml)的混合溶液中。反应混合物在30℃下搅拌反应16小时。TLC监测反应完全。收集有机层,饱和氯化钠溶液(10ml)洗涤,无水硫酸钠干燥,减压浓缩,所得粗品经制备TLC(用含10%甲醇的二氯甲烷洗脱)分离纯化得到黄色固体N2-(2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基-N4-(2-(异丙基磺酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺(18.7mg,37%)。
LC_MS:(ES+):m/z 553.9[M+H]+.
1H NMR(400MHz,CDCl3):δ9.58(s,1H),9.10(s,1H),8.79-8.81(m,1H),7.86-7.88(m,1H),7.60-7.64(m,1H),7.48-7.49(m,1H),7.16-7.20(m,1H),7.00-7.05(m,2H),6.92(s,1H),6.84-6.85(d,J=3.6Hz,1H),6.44-6.45(d,J=3.6Hz,1H),4.18-4.21(m,2H),4.09-4.11(m,2H),3.77-3.81(m,4H),3.24-3.30(m,1H),3.00-3.02(m,2H),2.92-2.94(m,2H),1.30-1.32(d,J=7.2Hz,6H).
实施例98
1-(13-((4-((2-(异丙基磺酰基)苯基)氨基)-7H-吡咯并[2,3-d]嘧啶-2-基)氨基)-2,3,5,6,8,9-六氢-7H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-7-基)乙-1-酮
实施方式同实施例97,用乙酰氯替换二碳酸二叔丁酯,获得标题化合物
LC_MS:(ES+):m/z 595.6[M+H]+.
1H NMR(400MHz,CDCl3):δ9.60(s,1H),8.74-8.80(m,2H),7.85-7.88(m,1H),7.60-7.65(m,1H),7.41-7.47(m,1H),7.09-7.20(m,2H),6.84-6.97(m,3H),6.45-6.46(m,1H),4.24-4.33(m,2H),4.12-4.15(m,1H),4.06-4.10(m,2H),3.80-3.82(m,1H),3.72-3.76(m,4H),3.55-3.63(m,2H),3.22-3.29(m,1H),2.17-2.23(m,3H),1.29-1.31(d,J=6.8Hz,6H).
实施例99
N4-(2-(异丙基磺酰基)苯基)-N2-(7-(甲基磺酰基)-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺
实施方式同实施例97,用甲基磺酰氯替换二碳酸二叔丁酯,获得标题化合物LC_MS:(ES+):m/z 632.0[M+H]+.
1H NMR(400MHz,DMSO-d6):δ11.44(s,1H),9.48(s,1H),8.88-8.93(m,2H),7.81-7.84(m,1H),7.72-7.76(m,1H),7.62-7.63(d,J=2.4Hz,1H),7.28-7.34(m,2H),7.03-7.04(m,1H),6.92-6.94(d,J=8.4Hz,1H),6.25-6.26(m,1H),4.07-4.13(m,4H),3.72-3.76(m,4H),3.51-3.54(t,J=2.6Hz,2H),3.44-3.49(m,1H),3.38-3.41(t,J=2.4Hz,2H),2.95(s,3H),1.17-1.19(d,J=6.8Hz,6H).
实施例100
(1s,4s)-4-((5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑唑啉-9-基)氨基)嘧啶-4-基)氧基)-1-甲基环己-1-醇
实施方法同方法一,用片段6-e替换片段1-b,获得标题化合物。
LC_MS:(ES+):m/z 433.2[M+H]+.
1H NMR(400MHz,CDCl3):δ8.07–7.88(m,1H),7.23–7.09(m,1H),7.23–7.09(m,1H),7.09–6.33(m,3H),5.07(tt,J=9.3,5.2Hz,1H),4.21(t,J=4.4Hz,2H),3.59(t,J=5.5Hz,2H),3.49–3.27(m,4H),3.36(s,3H),2.05–1.82(m,4H),1.82–1.71(m,2H),1.60(ddd,J=13.9,10.7,5.7Hz,2H),1.28(s,3H).
实施例101
(1s,4s)-1-甲基-4-((5-甲基-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氧基)环己-1-醇
实施方法同方法一,用片段2,4-二氯-5-甲基嘧啶替换片段1-a,用片段6-e替换片段1-b,获得标题化合物。
LC_MS:(ES+):m/z 429.2[M+H]+.
1H NMR(400MHz,CDCl3):δ7.90(s,1H),7.22(d,J=2.4Hz,1H),7.01–6.74(m,2H),6.60(d,J=8.6Hz,1H),5.06(tt,J=9.2,4.3Hz,1H),4.21(t,J=4.4Hz,2H),3.59(t,J=5.7Hz,2H),3.50–3.37(m,4H),3.36(s,3H),1.96–1.82(m,4H),1.77(dd,J=12.8,3.9Hz,2H),1.61(ddd,J=14.3,11.5,4.5Hz,2H),1.28(s,3H).
实施例102
(1s,4s)-4-((5-氯-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)氧基)-1-甲基环己-1-醇
实施方法同方法一,用片段2,4,5-三氯嘧啶替换片段1-a,用片段6-e替换片段1-b,获得标题化合物。
LC_MS:(ES+):m/z 449.1[M+H]+.
1H NMR(400MHz,CDCl3):δ8.06(s,1H),7.16(d,J=2.5Hz,1H),6.82(dd,J=8.6,2.5Hz,2H),6.61(d,J=8.7Hz,1H),5.15–4.99(m,1H),4.22(dd,J=5.2,3.6Hz,2H),3.59(t,J=5.7Hz,2H),3.43(t,J=5.7Hz,2H),3.41–3.37(m,2H),3.36(s,3H),1.95(td,J=10.2,8.7,4.4Hz,4H),1.78(dh,J=9.6,2.5Hz,2H),1.62–1.55(m,2H),1.28(s,3H).
实施例103
(1s,4s)-4-((5-氟-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)氧基)-1-甲基环己-1-醇
实施方法同方法一,用片段6-e替换片段1-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 477.55[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.37(s,1H),8.24(d,J=2.8Hz,1H),7.48(d,J=2.4Hz,1H),7.21-7.19(m,1H),6.97(d,J=8.8Hz,1H),5.08-5.01(m,1H),4.25(s,1H),4.12-4.06(m,4H),3.70(brs,4H),2.99-2.85(m,4H),2.46(s,3H),1.88-1.84(m,4H),1.67-1.64(m,2H),1.46-1.41(m,2H),1.15(s,3H).
实施例104
(1s,4s)-4-((5-氯-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)氧基)-1-甲基环己-1-醇
实施方法同方法一,用片段2,4,5-三氯嘧啶替换片段1-a,用片段6-e替换片段1-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 493.40[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.54(s,1H),8.26(s,1H),7.48(d,J=1.2Hz,1H),7.25-7.23(m,1H),7.01(d,J=8.8Hz,1H),5.09-5.02(m,1H),4.26(s,1H),4.17-4.08(m,4H),3.72(brs,4H),3.07-2.87(m,4H),2.57(s,3H),1.85-1.84(m,4H),1.68-1.64(m,2H),1.46-1.40(m,2H),1.16(s,3H).
实施例105
(1s,4s)-1-甲基-4-((5-甲基-2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)嘧啶-4-基)氧基)环己-1-醇
实施方法同方法一,用片段2,4-二氯-5-甲基嘧啶替换片段1-a,用片段6-e替换片段1-b,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 473.3[M+H]+.
1HNMR(400MHz,DMSO-d6):δ9.15(s,1H),8.00(s,1H),7.55(d,J=2.0Hz,1H),7.29-7.27(m,1H),6.99(d,J=8.8Hz,1H),5.06-4.99(m,1H),4.22-4.08(m,5H),3.78-3.67(m,4H),3.10-2.58(m,4H),2.50(s,3H),1.96(s,3H),1.83-1.80(m,4H),1.70-1.62(m,2H),1.46-1.39(m,2H),1.16(s,3H).
实施例106
环丙基((1R,5S)-3-(5-氟-2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)嘧啶-4-基)-3,8-二氮杂双环[3.2.1]辛-8-基)甲酮
实施方法同方法一,用片段79-d替换片段1-b,获得标题化合物。
LC_MS:(ES+):m/z 483.4[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.77(s,1H),7.94(d,J=6.8Hz,1H),7.01-7.06(m,2H),6.61(d,J=8.8Hz,1H),4.73-4.74(m,1H),4.57-4.58(m,1H),4.10-4.22(m,4H),3.56(t,J=5.6Hz,2H),3.35(t,J=5.6Hz,2H),3.29(s,2H),3.26(s,3H),3.21(d,J=12.4Hz,1H),3.09(d,J=12.4Hz,1H),1.96-2.01(m,2H),1.70-1.82(m,2H),1.26-1.34(m,1H),0.70-0.79(m,4H).
实施例107
(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)-6-(甲基氨基)嘧啶-4-基)氧基)环己-1-醇
实施方法同方法七,用片段2,6-二氯-N-甲基嘧啶-4-胺替换片段7-a,获得标题化合物。
LC_MS:(ES+):m/z 488.35[M+H]+.
1H NMR(400MHz,DMSO-d6):δ8.71(s,1H),7.66(s,1H),7.17(d,J=8.4Hz,1H),6.89(d,J=8.8Hz,1H),6.71(d,J=8.4Hz,1H),6.24(d,J=2.0Hz,1H),5.16(s,1H),4.89-4.85(m,1H),4.06-4.05(m,2H),3.98-3.94(m,2H),3.66-3.62(m,4H),2.84-2.67(m,7H),2.37-2.33(m,3H),1.75-1.60(m,6H),1.42-1.36(m,2H),1.13(s,3H).
实施例108
(1s,4s)-1-甲基-4-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-6-(甲基氨基)嘧啶-4-基)氧基)环己-1-醇
实施方法同方法七,用片段2,6-二氯-N-甲基嘧啶-4-胺替换片段7-a,用片段Amine-1替换片段Amine-3,获得标题化合物。
LC_MS:(ES+):m/z 444.55[M+H]+.
1H NMR(400MHz,CDCl3):δ7.24(d,J=2.4Hz,1H),6.86-6.84(m,1H),6.75(brs,1H),6.59(d,J=8.8Hz,1H),5.18(s,1H),4.99-4.94(m,1H),4.84(brs,1H),4.22-4.20(m,2H),3.59(t,J=5.6Hz,2H),3.43-3.37(m,4H),3.36(s,3H),2.85(d,J=5.2Hz,3H),1.95-1.91(m,2H),1.88-1.81(m,2H),1.78-1.74(m,2H),1.63-1.55(m,2H),1.27(s,3H).
实施例109
(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)喹唑啉-4-基)氧基)环己-1-醇
实施方法同方法七,用片段2,4-二氯喹唑啉替换片段7-a,获得标题化合物。
LC_MS:(ES+):m/z 509.40[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.35(s,1H),7.93(d,J=8.0Hz,1H),7.77-7.69(m,2H),7.52(d,J=8.4Hz,1H),7.37(d,J=8.4Hz,1H),7.26(t,J=7.6Hz,1H),6.98(t,J=8.8Hz,1H),5.29-5.27(m,1H),4.30(s,1H),4.13-4.08(m,4H),3.68-3.66(m,4H),2.86-2.67(m,4H),2.35(s,3H),1.93-1.91(m,4H),1.73-1.70(m,2H),1.50-1.43(m,2H),1.17(s,3H).
实施例110
(1s,4s)-1-甲基-4-((2-((7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)氨基)喹啉-4-基)氧基)环己-1-醇
实施方法同方法七,用片段2,4-二氯喹啉替换片段7-a,获得标题化合物。
LC_MS:(ES+):m/z 508.35[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.16(s,1H),7.96(s,1H),7.90(d,J=8.4Hz,1H),7.60-7.52(m,2H),7.30-7.21(m,2H),7.00(d,J=8.8Hz,1H),6.50(s,1H),4.48-4.42(m,1H),4.31(s,1H),4.20(brs,2H),4.12-4.09(m,2H),3.70(brs,4H),3.03-2.77(m,4H),2.49(s,3H),1.93-1.89(m,4H),1.73-1.69(m,2H),1.51-1.44(m,2H),1.18(s,3H).
实施例111
N-(5-甲基-2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)苯基)丙烯酰胺
实施方法同方法三,用片段(2-氨基-5-甲基苯基)氨基甲酸叔丁酯替换片段3-a,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 500.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.95(s,1H),8.57(s,1H),7.74(s,1H),6.95(dd,J=48.3,41.0Hz,2H),6.80(d,J=26.0Hz,1H),6.60(d,J=33.7Hz,2H),6.30(d,J=16.8Hz,1H),6.11(dd,J=29.9,19.3Hz,1H),5.94(s,1H),5.64(d,J=9.9Hz,1H),5.34(s,1H),4.12(dd,J=21.4,14.4Hz,2H),3.59(d,J=30.4Hz,2H),3.35(s,5H),2.33(d,J=17.2Hz,2H),1.75(s,3H).
实施例112
N-(5-甲基-2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)苯基)丙烯酰胺
实施方法同方法三,用片段(2-氨基-5-甲基苯基)氨基甲酸叔丁酯替换片段3-a,用片段7-a替换片段2-d,获得标题化合物。
LC_MS:(ES+):m/z 529.0[M+H]+.
1H NMR(400MHz,MeOD)δ8.13(s,1H),7.53(d,J=8.0Hz,1H),7.15(d,J=8.5Hz,2H),6.77(ddd,J=18.1,15.6,11.2Hz,3H),6.37(dd,J=5.9,3.9Hz,2H),5.77(dd,J=8.4,3.5Hz,1H),4.17–4.12(m,2H),3.58(t,J=5.6Hz,2H),3.49–3.36(m,4H),3.35(s,3H),2.39(s,3H).
实施例113
N-(2-((2-((4-甲基-3,4,5,6-四氢-2H-苯并[b][1,4,7]二恶唑啉-9-基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)苯基)甲磺酰胺
实施方法同方法七,用片段N-(2-氨基苯基)甲磺酰胺替换片段6-d,用片段Amine-1替换片段Amine-3,获得标题化合物。
LC_MS:(ES+):m/z 539.20[M+H]+.
1H NMR(500MHz,DMSO-d6):δ9.14(s,1H),8.60(s,1H),8.51(s,1H),8.28(s,1H),7.82(d,J=7.0Hz,1H),7.28(dd,J=7.6,1.6Hz,1H),7.11–7.03(m,2H),6.82–6.74(m,2H),6.67(d,J=8.7Hz,1H),4.19–4.13(m,2H),3.54(t,J=5.5Hz,2H),3.45(t,J=5.4Hz,2H),3.41–3.37(m,2H),3.28(s,3H),2.89(s,3H).
实施例114
N-(5-氟-4-(5-氟-2-甲氧基苯基)嘧啶-2-基)-7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-胺
实施方法同方法二,用片段5-氟-2-甲氧基苯硼酸替换片段2-b,片段1-a替换片段2-d,用片段Amine-3替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 473.5[M+H]+.
1H NMR(400MHz,DMSO-d6):δ9.66(s,1H),8.54(d,J=2.0Hz,1H),7.58(brs,1H),7.39-7.36(m,2H),7.23-7.19(m,2H),6.97-6.95(m,1H),4.08(brs,4H),3.80(s,3H),3.73-3.69(m,4H),3.29(s,3H),2.89-2.52(m,4H).
实施例115
N-(5-氟-4-(5-氟-2-甲氧基苯基)嘧啶-2-基)-7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-13-胺
实施方法同方法二,用片段5-氟-2-甲氧基苯硼酸替换片段2-b,片段1-a替换片段2-d,用片段Amine-2替换片段Amine-1,获得标题化合物。
LC_MS:(ES+):m/z 473.6[M+H]+.
1H NMR(400MHz,CDCl3):δ9.67(s,1H),8.55(d,J=2.0Hz,1H),7.57(d,J=2.4Hz,1H),7.41-7.36(m,2H),7.23-7.20(m,2H),6.99(d,J=8.8Hz,1H),4.13-4.07(m,4H),3.80(s,3H),3.70-3.62(m,4H),3.30(s,3H),3.08-2.75(m,4H).
实施例116
N4-(2-(甲氧基甲基)苯基)-N2-(7-甲基-2,3,6,7,8,9-六氢-5H-苯并[b][1,4,7]三氧杂[10]氮杂环十二烷-12-基)-5-(三氟甲基)嘧啶-2,4-二胺
实施方法同方法七,用片段2-(甲氧基甲基)苯胺替换片段6-d,获得标题化合物。LC_MS:(ES+):m/z 534.75[M+H]+.
1H NMR(400MHz,CD3OD):δ8.28(s,1H),7.94-7.91(m,1H),7.85-7.79(m,1H),7.47-7.33(m,4H),7.21-7.19(m,1H),7.08-7.06(m,1H),6.92-6.90(m,1H),4.50(s,2H),4.15-4.13(m,2H),3.98-3.95(m,2H),3.81-3.78(m,4H),3.39(s,3H),3.09-2.92(m,4H),2.58(s,3H).
实施例117:化合物在水和稀盐酸中溶解度的测定
标准样品的配制:
将化合物溶解于乙腈(LCMS纯)中,配成如下浓度梯度标准样品:1000μg/mL,500μg/mL,250μg/mL,125μg/mL,62.5μg/mL,31.25μg/mL。
水中待测样品制备:
称取待测物粉末1.0mg左右,加入0.3mL超纯水(pH~6)中,室温充分震荡过夜,离心,取上清,用乙腈稀释10倍。混合均匀后进样分析。
稀盐酸中待测样品制备:
吸取153.8μL 6M HCl,加入20mL水,混匀制成稀盐酸溶液A,(pH~1.5)。称取待测物粉末1.0mg左右,加入0.3mL稀盐酸溶液A,室温充分震荡1h,离心,取上清,用乙腈稀释10倍。混合均匀后进样分析。
分析方法:
将标准样品和待测样品用岛津LCMS-2020分析,色谱柱:Skim-psck GIST 5μm C18 2.1*50nm;流动相:A:水,B:甲醇;流速:0.3mL/min。B的梯度:0min:10%;0-1.0min:10%;1.0-2.5min:95%;3.0-4.0min:95%;4.0-4.1min:10%;4.1-6.0min:10%。
数据处理:
将标准样品的浓度/峰面积的关系制成标准曲线,用此标准曲线定标待测样品的浓度。
化合物溶解度=待测样品的浓度×10
化合物的溶解度如下表:
由上述结果可知,本发明的化合物在水中具备良好的溶解度,在模拟胃酸的稀盐酸中的溶解度更好。
生物活性测试实施例:
生物测试实施例-1:激酶活性抑制实验:
检测受试化合物在JAK1,JAK2,JAK3,TYK2,CDK7,CDK9,EGFR(T790M)和LRRK2激酶上的抑制率。化合物测试浓度为10nM和100nM,复孔检测,或10μM起,3倍稀释,10个浓度,复孔检测。
采用Caliper迁移率变动检测技术(Caliper mobility shift assay)的方法检测化合物对JAK1,JAK2,JAK3,CDK7和CDK9激酶的活性抑制作用。使用分液器Echo 550向目的板384-well-plate转移250nL 100倍终浓度的化合物。用1×激酶缓冲液配制2.5倍终浓度的激酶溶液。在化合物孔和阳性对照孔中分别加10μL的2.5倍终浓度的激酶溶液;在阴性对照孔中加入10μL的1×激酶缓冲液。1000rpm离心30秒后在室温下孵育10分钟。用1×激酶缓冲液配制5/3倍终 浓度的ATP和激酶底物的混合溶液,加入15μL的5/3倍终浓度的ATP和底物的混合溶液,起始反应。将384孔板1000rpm离心30秒,振荡混匀后室温孵育相应的时间,加入30μL终止检测液终止激酶反应,1000rpm离心30秒,振荡混匀。用Caliper EZ Reader读取转化率。换算抑制率%=(阳性对照转化率均值%-样品转化率%/(阳性对照转化率均值%-阴性对照转化率均值%)。其中:阴性对照孔,代表激酶缓冲液背景孔的转化率读数;阳性对照孔,代表没有化合物抑制孔的转化率读数。以浓度的log值作为x轴,百分比抑制率为y轴,采用分析软件GraphPad Prism 5的log(inhibitor)vs.response-Variable slope拟合量效曲线,从而得出各个化合物对酶活性的IC50值。计算公式:y=Bottom+(Top-Bottom)/(1+10^((LogIC50-x)ⅹhillslope)),x:抑制剂浓度的对数;y:%抑制率。
下表1中提供了一些化合物在100nM对一些激酶活性的抑制率:
(区间:A>80%;B:40-79%;C:25-39%)


下表2中提供了一些化合物在10nM对一些激酶活性的抑制率:
(区间:A>80%;B:40-79%;C:25-39%)


在下表3中提供了一些化合物在一些激酶活性测定中的IC50区间:
(IC50区间:A<0.05μM;B:0.05-0.2μM;C:0.2-1μM):


生物测试实施例-2:LRRK2激酶活性抑制实验:
采用ADP-Glo方法检测化合物对LRRK2激酶的活性抑制作用。将化合物稀释液转移到384孔板(784075,Greiner)中,密封孔板,以1000g的转速离心1分钟。用1×激酶缓冲液配制2倍终浓度的激酶溶液,加入2.5μL 2倍终浓度的激酶溶液,以1000g的转速离心孔板30秒,室温放置10分钟。在1×激酶缓冲液中配制2倍终浓度的ATP和激酶底物的混合溶液,加入2.5μL 2倍终浓度的ATP和激酶底物的混合溶液,开始反应。以1000g转速离心孔板30秒,密封孔板,室温放置2小时。加入4μL ADP-Glo试剂,室温下孵育40分钟;再加入8μL激酶检测试剂,室温下孵育40分钟。在Envision 2104微孔板检测仪上检测发光信号值。换算抑制率%=100-(化合物信号值-阳性对照信号值)/(空白对照信号值-阳性对照信号值)×100
用GraphPad 6.0计算IC50并绘制化合物的量效曲线:y=Bottom+(Top-Bottom)/(1+10^((logic50-x)ⅹhillslope)),x:抑制剂浓度的对数;y:%抑制率。
在下表4中提供了一些化合物在LRRK2激酶活性测定中的IC50区间:
(IC50区间:A<0.1μM;B:0.1-0.5μM;C:0.5-1μM):

生物测试实施例-3:EGFR激酶活性抑制实验:
采用TR-FRET方法检测化合物对EGFR激酶的活性抑制作用。将化合物稀释液转移到384孔板(784075,Greiner)中,密封孔板,以1000g的转速离心1分钟。在1×激酶缓冲液中配制2×EGFR wt,向384孔板中加入5μL 2×EGFR wt,以1000g的转速离心孔板30秒,室温放置10分钟。在1×激酶缓冲液中配制2×TK-substrate-biotin(2μM)和ATP混合物。加入5μL 2×TK-substrate-biotin和ATP混合物开始反应。以1000g转速离心孔板30秒,密封孔板,室温放置40分钟。在HTRF检测缓冲液中配制4×Sa-XL 665,向孔板中加入5μL Sa-XL 665和5μL TK-antibody-Cryptate,以1000g的转速离心孔板30秒,室温放置1小时。用Envision 2104微孔板检测仪检测615nm(Cryptate)和665nm(XL665)下的荧光信号值。换算抑制率%=100-(化合物信号值-阳性对照信号值)/(空白对照信号值-阳性对照信号值)×100,用GraphPad 8.0计算IC50并绘制化合物的量效曲线:y=Bottom+(Top-Bottom)/(1+10^((logic50-x)ⅹhillslope)),x:抑制剂浓度的对数;y:%抑制率。
在下表5中提供了一些化合物在EGFR激酶活性测定中的IC50区间:
IC50区间:A<0.05μM;B:0.05-1μM;C:1-5μM):
生物测试实施例-4:
CDK9细胞活性测试
实验所用细胞系MV-4-11,用含10%胎牛血清的IMDM培养液(含100U/mL青霉素及0.1g/L链霉素)培养,细胞系HCC1187,用含10%胎牛血清的RPMI-1640培养液(含100U/mL青霉素及0.1g/L链霉素)培养,细胞系Mia Paca-2,用含10%胎牛血清和2.5%马血清的DMEM(含100U/mL青霉素及0.1g/L链霉素)培养,细胞系A375,用含10%胎牛血清的RPMI-1640培养液(含100U/mL青霉素及0.1g/L链霉素)培养,所有细胞均于37℃、5%CO2的饱和湿度培养箱中培养。
MV-4-11细胞、HCC1187细胞、Mia Paca-2细胞、A375细胞生长至80%-90%融合度时用胰酶消化、重悬、计数并用完全培养基稀释至一定的细胞密度,每孔100μL接种于96孔板中,以完全培养基作为背景对照,于37℃、5%CO2的饱和湿度培养箱中培养过夜。用DMSO稀释化合物为终浓度的200倍,取3μL上述化合物的DMSO储存液加入197μL完全培养基,每孔加入50μL培养基稀释的化合物于细胞中,于37℃、5%CO2的饱和湿度培养箱中培养72小时。72小时后将培养板放置于室温平衡,每孔加入40μL试剂,振荡器混匀2分钟以裂解细胞,室温孵育60分钟以稳定化学发光信号,用PE EnVision酶标仪读取化学发光。计算细胞生长抑制率Inh%=(DMSO孔-化合物处理孔)/(DMSO孔-背景孔)ⅹ100,用GraphPad Prism 5拟合得到细胞生长IC50值。
在下表6中提供了一些化合物在细胞活性测定中的IC50区间:
(IC50区间:A<0.1μM;B:0.1-1μM;C:1-5μM),NT未测:

生物测试实施例-5:
JAKs细胞活性测试
JAK1_IL-4诱导的THP1细胞中pSTAT6的检测
离心收集THP1细胞,用1ⅹHBSS重悬,计数并稀释至一定的细胞密度。接种于384孔板中,于37℃、5%CO2的饱和湿度培养箱中培养。将化合物(DMSO终体积为0.3%)加入到THP1细胞中(10个剂量,两个复孔),于37℃、5%CO2的饱和湿度培养箱中培养。加入hIL-4刺激细胞,于37℃、5%CO2的饱和湿度培养箱中培养。加入裂解液,室温孵育。根据AlphaLISA SureFire Ultra p-STAT6检测试剂盒说明,将裂解液转移至pSTAT6的检测体系,PE Envision酶标仪读取ALPHA信号,计算抑制率Inh%=((DMSO-样本)/(DMSO-背景))×100,IC50通过Graphpad Prism non-linear regression equation拟合得到,y=Bottom+(Top-Bottom)/(1+10^((LogIC50-x)ⅹhillslope)),x为化合物浓度,y为抑制率。
JAK2_pSTAT5按上述步骤在IL-3诱导的BAF3细胞中进行检测,JAK3_pSTAT5按上述步骤在IL-2诱导的CTLL-2细胞(无血清刺激)中进行检测。
在下表7中提供了一些化合物在细胞活性测定中的IC50区间:
(IC50区间:A<0.05μM;B:0.05-0.5μM;C:0.5-5μM),NT未测:

生物测试实施例-6:
HEK293T过表达全长LRRK2(G2019S)或野生型LRRK2磷酸化S935的检测
HEK293T接种于6孔板中,过夜培养。次日用Superfect转染试剂瞬时转入pcDNA5-FRT/TO-FLAG LRRK2(G2019S,全长)或pCMV-flag-WT LRRK2(全长)质粒,37度,5%二氧化碳条件下培养20-24小时。次日,收集细胞,以0.48×10^6细胞/毫升的密度重悬,50微升每孔接种于384孔板中。使用Tecan加入化合物或DMSO(体积比为0.5%)。37度,5%二氧化碳条件下培养1.5小时。1.5小时后弃上清,50微升每孔8%PFA固定细胞,室温孵育1小时。用EL406洗板机清洗三次,每孔115微升PBST,拍干培养板。用50微升每孔的Licor Odyssey封闭液(0.1%Tween 20)封闭细胞,室温1.5小时。弃封闭液拍干培养板。每孔加入20微升一抗(1:1000(pS935)一抗,用Licor Odyssey缓冲液(0.1%Tween20)稀释),4度过夜孵育。次日,弃一抗,用PBST洗五遍。每孔加入20微升二抗(用Licor Odyssey缓冲液(0.1%Tween20)按照1:500稀释,DNA染色1:2000),室温孵育1小时。弃二抗,PBST洗三遍。LICOR Odyssey imager Scanner测累积强度,数据分析。
在下表8中提供了一些化合物在LRRK2(G2019S)或野生型LRRK2细胞活性测定中的IC50区间:
(IC50区间:A<0.5μM;B:0.5-1μM;C:1-5μM):

生物测试实施例-7:
A549细胞中内源性磷酸化LRRK2(S935)的检测
A549细胞按1×10^6细胞/毫升的密度重悬于无血清的1640培养基中,每孔1毫升接种于6孔板中,37度,5%二氧化碳条件下培养6-8小时。加入1微升化合物的DMSO储存液或DMSO(体积比为0.1%),混匀,37度,5%二氧化碳条件下培养15小时。收集细胞,每个样品加入50微升RIPA III裂解液冰上裂解细胞15分钟,4度13000rpm离心30分钟,取上清,BCA测细胞裂解液浓度,所有样品浓度归一化。加入5×上样缓冲液,95度10分钟。WB检测内源性LRRK2磷酸化S935的含量,磷酸化LRRK2(S935)抗体为Abcam#133450,。Image J统计磷酸化LRRK2(S935)信号强度,用beta actin来归一样品中磷酸化LRRK2(S935)信号,数据分析。
下表9中提供了一些化合物在2uM浓度下对A549细胞中LRRK2(S935)活性的抑制率:
(区间:A>80%;B:40-79%;C:25-39%;)
下表10中提供了一些化合物在1uM浓度下对A549细胞中LRRK2(S935)活性的抑制率:
(区间:A>80%;B:40-79%;C:25-39%;)
下表11中提供了一些化合物在A549细胞中LRRK2(S935)活性的抑制活性IC50区间:
(IC50区间:A<0.1μM;B:0.1-0.5μM;C:0.5-5μM)

Claims (13)

  1. 一种式(I)化合物,或其立体异构体、互变异构体或药学上可接受的盐:
    其中,
    Z表示不存在(共价键)或者-OCH2CH2-;
    L表示不存在(共价键)、-O-、-NH-或者-N(C1~4烷基)-;
    A是任选地被1或2个选自以下的取代基取代的C6~14芳基、5~12元杂芳基、C1~10烷基、C3~8环烷基、3~8元杂环基或5~18元桥环基,其中所述取代基选自:氘、C1~4烷基、C1~10烷氧基、C3~6环烷基、3~8元杂环基、C1~4烷基磷酰基、C1~4烷基磺酰基、氨基磺酰基、C1~4烷基氨基磺酰基、氰基C3~6环烷基、C2~4烯基甲酰基、C2~4烯基甲酰氨基、氰基C1~4烷基氨基甲酰基、氰基C1~4烷基、C1~4烷基甲酰基-3~8元杂环基、C3~6环烷基甲酰基、3~8元杂环基-磺酰基、C1~4烷基磺酰基-3~8元杂环基、氨基C3~6环烷基甲酰氨基、C1~4烷基磷酰氨基、C1~4烷基磺酰氨基、氰基、羟基、氧代、巯基、氨基、C2~4烯基和卤素;
    R1是氢,或者任选取代的C1~4烷基、C3~6环烷基、C1~4烷基-CO-、-CHO或C1-4烷基磺酰基,其中所述取代基选自:氘、C1~4烷基、C1~10烷氧基、C3~6环烷基、氧代、氰基、羟基、氨基、二甲氨基、羟胺基;
    R2独立地选自氢、C1~3烷基、三氟甲基、C1~3烷氧基、氰基和卤素;
    n是1或2;
    Ra选自氢、氨基、三氟甲基、卤素、氰基、C1~3烷基、乙酰基、C1~3烷基磷酰基和C1~3烷基磺酰基;
    Rb选自氢、氨基和C1~3烷基氨基;
    Ra、Rb和它们所连接的C原子可以一起形成5-6元芳环、5~12元杂芳环或5~8元杂环。
  2. 根据权利要求1所述的式(I)化合物或其立体异构体、互变异构体或药学上可接受的盐,其中所述式(I)化合物具有式(II)所示的结构:
    其中,L、A、R1、R2、Ra、Rb和n具有如权利要求1中所述的定义。
  3. 根据权利要求1所述的式(I)化合物或其立体异构体、互变异构体或药学上可接受的盐,其中所述式(I)化合物具有式(III)所示的结构:
    其中,L、A、R1、R2、Ra、Rb和n具有如权利要求1中所述的定义。
  4. 根据权利要求1所述的式(I)化合物或其立体异构体、互变异构体或药学上可接受的盐,其中所述式(I)化合物具有式(IV)所示的结构:
    其中,L、A、R1、R2和n具有如权利要求1中所述的定义;
    X、Y各自独立地选自C和N;
    Z表示不存在(共价键)或者-OCH2CH2-。
  5. 根据权利要求1-4中任一项所述的化合物或其立体异构体、互变异构体或药学上可接受的盐,其中A选自以下结构:
  6. 根据权利要求1-5中任一项所述的化合物或其立体异构体、互变异构体或药学上可接受的盐,其中所述化合物选自:













  7. 一种药物组合物,其包含权利要求1至6中任一项所述的化合物或其立体异构体、互变异构体或药学上可接受的盐,以及任选的可药用载体。
  8. 根据权利要求1至7中任一项所述的化合物或其立体异构体、互变异构体或药学上可接受的盐在制备用作蛋白激酶抑制剂的药物中的应用。
  9. 根据权利要求1至7中任一项所述的化合物或其立体异构体、互变异构体或药学上可接受的盐在制备用于治疗或预防蛋白激酶相关疾病的药物中的应用。
  10. 根据权利要求9的应用,其中所述蛋白激酶相关疾病是其中的疾病发展或症状与蛋白激酶的信号传递、介导、调节或调整有关的疾病。
  11. 根据权利要求8-10中任一项所述的应用,其中所述蛋白激酶选自LRRK2、JAK1、JAK2、JAK3、EGFR和CDK9。
  12. 根据权利要求9-11中任一项所述的应用,其中所述疾病选自神经退行性疾病、自身免疫疾病和肿瘤。
  13. 根据权利要求8-11中任一项所述的应用,其中所述疾病选自帕金森病、哮喘、皮炎、非小细胞肺癌、急性髓系白血病(AML)和肝癌。
PCT/CN2023/119030 2022-09-21 2023-09-15 大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用 WO2024061118A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211155875.X 2022-09-21
CN202211155875.XA CN117736198A (zh) 2022-09-21 2022-09-21 大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用

Publications (1)

Publication Number Publication Date
WO2024061118A1 true WO2024061118A1 (zh) 2024-03-28

Family

ID=90255050

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/119030 WO2024061118A1 (zh) 2022-09-21 2023-09-15 大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用

Country Status (2)

Country Link
CN (1) CN117736198A (zh)
WO (1) WO2024061118A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1486312A (zh) * 2000-12-21 2004-03-31 ��̩��˹ҩ��ɷ����޹�˾ 可用作蛋白激酶抑制剂的吡唑化合物
CN102260248A (zh) * 2003-09-16 2011-11-30 诺瓦提斯公司 作为zap-70和/或syk抑制剂的2,4-二(杂)芳基氨基嘧啶衍生物
WO2016197987A1 (zh) * 2015-06-12 2016-12-15 杭州英创医药科技有限公司 作为Syk抑制剂和/或Syk-HDAC双重抑制剂的杂环化合物
WO2017118438A1 (zh) * 2016-01-08 2017-07-13 杭州英创医药科技有限公司 作为fgfr抑制剂的杂环化合物
US20170342088A1 (en) * 2014-08-25 2017-11-30 Salk Institute For Biological Studies Novel ulk1 inhibitors and methods using same
CN111683662A (zh) * 2017-08-28 2020-09-18 陈志宏 取代嘧啶类化合物及其药物组合物和治疗方法

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1486312A (zh) * 2000-12-21 2004-03-31 ��̩��˹ҩ��ɷ����޹�˾ 可用作蛋白激酶抑制剂的吡唑化合物
CN102260248A (zh) * 2003-09-16 2011-11-30 诺瓦提斯公司 作为zap-70和/或syk抑制剂的2,4-二(杂)芳基氨基嘧啶衍生物
US20170342088A1 (en) * 2014-08-25 2017-11-30 Salk Institute For Biological Studies Novel ulk1 inhibitors and methods using same
WO2016197987A1 (zh) * 2015-06-12 2016-12-15 杭州英创医药科技有限公司 作为Syk抑制剂和/或Syk-HDAC双重抑制剂的杂环化合物
WO2017118438A1 (zh) * 2016-01-08 2017-07-13 杭州英创医药科技有限公司 作为fgfr抑制剂的杂环化合物
CN111683662A (zh) * 2017-08-28 2020-09-18 陈志宏 取代嘧啶类化合物及其药物组合物和治疗方法

Also Published As

Publication number Publication date
CN117736198A (zh) 2024-03-22

Similar Documents

Publication Publication Date Title
JP7005582B2 (ja) ブルトン型チロシンキナーゼ(btk)インヒビターとしての多フルオロ置換化合物
US20220127281A1 (en) Heteroaryl compounds that inhibit g12c mutant ras proteins
CN105732636B (zh) 杂芳化合物及其在药物中的应用
KR102131612B1 (ko) Pi3k 억제제로서 헤테로시클릴아민
KR101792837B1 (ko) 키나아제 억제제로서 사용을 위한 이미다조피라진
JP2022523981A (ja) 抗癌剤として有用な縮合三環式化合物
ES2945212T3 (es) Utilización de derivados de pirazolopirimidinas para el tratamiento de trastornos relacionados con PI3K
CN107108644B (zh) 二氟甲基-氨基吡啶和二氟甲基-氨基嘧啶
CN114450284B (zh) 作为bcl-xl蛋白抑制剂的6,7-二氢-5h-吡啶并[2,3-c]哒嗪衍生物
ES2654288T3 (es) Moduladores de P2X7
US10513523B2 (en) Imidazopyrazines and pyrazolopyrimidines and their use as AMPA receptor modulators
WO2011028685A1 (en) Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
AU2014249192A1 (en) BET bromodomain inhibitors and therapeutic methods using the same
KR101947289B1 (ko) 신규 피롤로피리미딘 화합물 또는 그의 염, 및 이것을 함유하는 의약 조성물, 특히 nae 저해 작용에 기초하는 종양 등의 예방제 및/또는 치료제
CN115991679A (zh) 作为a2a抑制剂的2-氧代噻唑衍生物和用于治疗癌症的化合物
KR20130029756A (ko) N-7 치환된 퓨린 및 피라졸로피리미딘 화합물, 조성물 및 사용 방법
JP2021518379A (ja) Jak阻害剤
WO2020207476A1 (zh) 一种吡唑并吡嗪衍生的化合物、药物组合物以及其用途
CN111655690B (zh) 吡唑并吡啶酮化合物
JP2023517680A (ja) Kcc2調節剤としての縮合ピリミジン化合物
WO2016001077A1 (en) Compounds inhibiting the enzyme monopolar spindle 1 kinase,pharmaceutical compositions and uses thereof
CN117897384A (zh) Cdk2抑制剂
US20240101535A1 (en) Dihydroisoquinolinone derivative and application thereof
WO2024061118A1 (zh) 大环含氮冠醚化合物及其作为蛋白激酶抑制剂的应用
AU2018337138B2 (en) 2-substituted pyrazole amino-4-substituted amino-5-pyrimidine formamide compound, composition, and application thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23867407

Country of ref document: EP

Kind code of ref document: A1