CN109553604B - 4-aminopyrimidine derivatives as CXCR4 inhibitors and uses thereof - Google Patents

4-aminopyrimidine derivatives as CXCR4 inhibitors and uses thereof Download PDF

Info

Publication number
CN109553604B
CN109553604B CN201811034891.7A CN201811034891A CN109553604B CN 109553604 B CN109553604 B CN 109553604B CN 201811034891 A CN201811034891 A CN 201811034891A CN 109553604 B CN109553604 B CN 109553604B
Authority
CN
China
Prior art keywords
dichloromethane
added
synthesis
dried
cxcr4
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201811034891.7A
Other languages
Chinese (zh)
Other versions
CN109553604A (en
Inventor
张小虎
郑计岳
马海阔
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shengshi Taike Biopharmaceutical Technology (Suzhou) Co.,Ltd.
Original Assignee
Cgenetech Suzhou China Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cgenetech Suzhou China Co Ltd filed Critical Cgenetech Suzhou China Co Ltd
Priority to US16/649,983 priority Critical patent/US11396501B2/en
Priority to EP18859565.6A priority patent/EP3687540A4/en
Priority to JP2020538760A priority patent/JP7282786B2/en
Priority to PCT/US2018/052503 priority patent/WO2019060860A1/en
Priority to KR1020207010206A priority patent/KR20200058443A/en
Publication of CN109553604A publication Critical patent/CN109553604A/en
Application granted granted Critical
Publication of CN109553604B publication Critical patent/CN109553604B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered

Abstract

The invention provides a heterocyclic compound with CXCR4 signal channel inhibition activity and pharmaceutically acceptable salts, isotopes, isomers and crystal structures thereof, wherein the compound has a structure shown in a general formula I:
Figure DDA0001789890170000011
the invention also provides the application of the heterocyclic compound with CXCR4 signal channel inhibition activity. The heterocyclic compounds of the present invention having CXCR4 signaling pathway inhibitory activity, as potent antagonists of the CXCR4 signaling pathway, can be used for the treatment or prevention of conditions responsive to CXCR4 receptor antagonism.

Description

4-aminopyrimidine derivatives as CXCR4 inhibitors and uses thereof
Technical Field
The present invention relates to heterocyclic compounds having CXCR4 inhibitory activity, compositions comprising the compounds, processes for preparing the compounds, and uses of the compounds in medicine, particularly for the treatment of conditions responsive to CXCR4 receptor antagonism such as cancer, cancer metastasis, HIV-related diseases, encephalitis, diabetic retinopathy, age-related macular degeneration and retinal cardiovascular disease; the invention also relates to the use of said compounds in stem cell harvesting methods, including, for example, facilitating the release and mobilization of stem cells, including hematopoietic stem cells, non-hematopoietic stem cells, and progenitor stem cells, prior to harvesting. The invention belongs to the technical field of medicines.
Background
CXCR4 belongs to a 7-transmembrane G-protein coupled receptor (GPCR), and its endogenous ligand is the chemokine SDF-1 (stromal cell derived factor-1), also known as CXCL 12. CXCR4 was first discovered to be expressed on T cells as a co-receptor for the CD4 antigen, which binds to the glycoprotein gp120 of Human Immunodeficiency Virus (HIV), mediating the process of viral entry into host T cells. Subsequent studies have shown that inhibition of CXCR4 helps to promote stem cell release and mobilization, and that in combination with granulocyte colony-stimulating factor (G-CSF), stem cell mobilization results for hematopoietic stem cells and endothelial progenitor cells can be improved. The CXCR4/SDF-1 interaction is also a major regulator of cancer stem cell trafficking in humans, playing an important role in the progression and metastasis of many types of cancer cells. Because of these important biological functions mediated by CXCR4, CXCR4 inhibitors are promising for stem cell transplantation and for the treatment of diseases (cancer, HIV, diabetic retinopathy, inflammation, etc.).
Hematopoietic stem cell mobilization
Hematopoietic Stem Cells (HSCs) are widely used in the treatment of diseases of the hematopoietic system, such as multiple myeloma and non-hodgkin's lymphoma. The cytotoxic drug can kill tumor cells and hematopoietic system cells at the same time. Mobilization and harvesting of hematopoietic stem cells allows for reinfusion of the stem cells back into the body after drug treatment for reintroduction into the hematopoietic system.
In general, stem and progenitor cells highly express CXCR4, while bone marrow has a high concentration of SDF-1. Thus, stem and progenitor cells will be attracted to and retained in the bone marrow. Inhibition of CXCR4 interaction with SDF-1 allows stem or progenitor cells to leave the bone marrow and enter the peripheral blood, so that stem cells for transplantation can be mobilized from donors (allografts) or patients (autografts) (Prog Mol Biol trans sci.2012; 111, 243-. CXCR4 inhibitors can also be combined with another mobilizing agent, G-CSF, to improve its mobilization efficiency and success.
HIV and HIV-related pain
CXCR4 and CCR5 are co-receptors for HIV entry into host cells (cell. 1996; 87, 745-56.). Inhibition of CXCR4 may reduce infectivity of the virus X4 strain. Thus CXCR4 inhibitors may be effective in the treatment of HIV infection, particularly in combination with CCR5 inhibitors. The X4 tropic HIV virus is most pathogenic and tends to predominate in the late stages of infection, where neuropathic pain becomes an increasingly serious problem for patients. CXCR4 inhibitors have antiviral and neuropathic pain relieving properties and can be used in combination with other HIV therapeutic agents and analgesic agents to treat HIV and HIV-associated pain.
Pain and inflammation
There is CXCR4 expression on primary sensory neurons, so CXCR4 inhibitors can act as analgesics to control pain (J neurosci.2001, 21, 5027-35). Inhibition of CXCR4 reduced eosinophil infiltration and airway responses in allergic airway diseases (j. immunological.2000, 165, 499-508).
The SDF-1/CXCR4 pathway mediates inflammatory responses by activating integrin-induced cell rolling and close adhesion to activated endothelial cells, promoting inflammatory cells to reach inflammatory sites through vascular endothelium through high chemotaxis of inflammatory cells such as neutrophils, lymphocytes and monocytes (J.Immunol.2000, 164, 5035-5040.). The synovium of rheumatoid arthritis expresses SDF-1, attracts memory T cells expressing CXCR4 to be gathered in a large amount in the synovium, and inhibits the apoptosis of the T cells. In addition, SDF-1 in the blood of the systemic lupus erythematosus patient is obviously increased, peripheral blood mononuclear cells of the patient with the spondyloarthropathy, the rheumatoid arthritis and the psoriatic arthritis express abnormally increased CXCR4, and the patient with the polymyositis and the dermatomyositis also detects the abnormality of SDF-1/CXCR4, which indicates that the SDF-1/CXCR4 plays an important role in the immune and inflammatory processes. Inhibition of CXCR4 interaction with SDF-1 will help to alleviate the inflammatory response.
Retinal neovascularization
Retinal neovascularization is a major cause of blindness in diabetic patients and age-related macular degeneration patients. The SDF-1/CXCR4 bio-axis is significantly involved in ocular neovascularization and is a target for treatment of related retinopathies. Blockade of CXCR4 receptors can prevent recruitment of endothelial progenitor cells, thereby preventing the formation of new microvessels (FASEB j.2007, 21, 3219-30.). Also, CXCR4 inhibitors and VEGFR antibodies have potential additive effects in inhibiting angiogenesis.
Cancer and cancer metastasis
SDF-1/CXCR4 plays an important role in the development, metastasis and recurrence of tumors. SDF-1 and CXCR4 are highly expressed in tumor tissue. High concentrations of SDF-1 in the tumor microenvironment attract CXCR4 positive immunosuppressive cells Treg and MDSC to migrate, accumulate within the tumor, suppress effector T cells and M1-type macrophages in the tumor microenvironment from attacking tumor cells, and thus achieve immune escape (Cancer Immunology Research 2014, 2, 187-193.). In addition, in FAP protein positive tumors such as pancreatic cancer, high concentrations of SDF-1 will reject effector T cells into the tumor microenvironment, leaving the tumor tissue free from T cell infiltration, thus enabling immune escape (Proc Natl Acad Sci usa.2013, 110, 20212-. The inhibition of CXCR4 can prevent the accumulation of immune suppressor cells Treg and MDSC in tumor and make effector T cell enter tumor tissue, thus enhancing the killing ability of immune system to tumor. The CXCR4 inhibitor has addition effect with other tumor immunotherapy such as PD-1 antibody or PD-L1 antibody, CTLA-4 antibody, etc., and can expand the applicable population of the tumor immunotherapy and improve response rate.
SDF-1 is combined with CXCR4 on tumor cells to activate a plurality of signal paths such as downstream EGFR, PI3K, MAPK, Wnt and the like to promote cell proliferation, and simultaneously activates an NF-KB signal path and up-regulates the expression of BCL-2 gene to inhibit apoptosis, thereby directly promoting the growth of tumors. Meanwhile, SDF-1/CXCR4 can promote tumor angiogenesis and indirectly promote tumor growth by up-regulating VEGF expression, recruiting endothelial progenitor cells and other modes. SDF-1 activates CXCR4 downstream pathway, promotes epithelial intercellular transformation, and enhances tumor cell invasion ability. Meanwhile, SDF-1 is continuously and highly expressed in normal tissues such as bone marrow, lymph nodes, liver, lung and brain endothelium, and attracts and recruits CXCR4 positive tumor cells, particularly tumor stem cells, to migrate to the normal tissues. The clinical findings show that the most common tissues and organs of tumor metastasis are bone, liver, brain, lung and adrenal gland, which are tissues with high SDF-1 expression, and the proportion of CXCR4 positive tumor stem cells in a metastasis range is obviously higher than that of a primary range, which indicates that SDF-1/CXCR4 has an important role in tumor metastasis. Blockade of CXCR4 helps to inhibit tumor proliferation and metastasis (Oncogene2015, 1-11.).
CXCR4 and SDF-1 are also involved in maintenance of tumor stem cells and in tumor recurrence and drug resistance after radiotherapy/chemotherapy. The use of CXCR4 inhibitors at the same time or after the end of radiation or chemotherapy helps to increase the sensitivity of tumors to radiation and chemotherapy and reduce the recurrence rate (Clinical Cancer Research 2011, 17, 2074-.
There is AMD3100 as a marketed CXCR4 inhibitor, but AMD3100 will form 4 salts at physiological pH and have poor bio-permeability, resulting in no oral bioavailability. AMD3100 structure optimization, get a series of four hydrogen isoquinoline compounds with long-chain amino, wherein the representative compound is AMD070(J.Med.chem.2010, 53, 3376-3388, WO 2004106493). AMD070 has a certain oral bioavailability, but has strong inhibitory activity on liver enzymes due to a strong basic center (amino side chain) in the structure (the inhibition rate on CYP2D6 is 100% at 1. mu.M, Bioorg Med Chem Lett.2015, 25, 4950-4955.). AMD070, although it entered the second phase of the clinic, eventually terminated the clinical trial because of hepatotoxicity observed in preclinical trials.
Figure BDA0001789890150000031
Disclosure of Invention
In view of the above-mentioned drawbacks of the prior art, it is an object of the present invention to propose a heterocyclic compound having CXCR4 inhibitory activity, which is able to effectively inhibit the CXCR4 pathway, and which can be used for the treatment or prevention of conditions responsive to CXCR4 receptor antagonism, and uses thereof.
The purpose of the invention is realized by the following technical scheme:
a heterocyclic compound having CXCR4 inhibitory activity, and pharmaceutically acceptable salts, isotopes, isomers and crystal forms thereof, having the structure shown in formula I:
Figure BDA0001789890150000041
wherein, A is1,A2,A3Each independently selected from N or CR10And A is1,A2,A3At least one is N;
w is
Figure BDA0001789890150000042
U is
Figure BDA0001789890150000043
Q is a bond or CR23R24
R1,R2,R3,R4Are respectively and independently selected from hydrogen atom, deuterium atom, cyano, halogen, hydroxyl, amino and NHC1-6Alkyl, N (C)1-6Alkyl radical)2、NHCOC1-6Alkyl, NHCOOC1-6Alkyl, NHSO2C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-8Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, said alkyl and alkoxy being unsubstituted or substituted with 1 to 3 halogen or deuterium atoms; r1,R2,R3,R4Preferably independently selected from hydrogen atom, deuterium atom, cyano, halogen, hydroxy, C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-8Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, said alkyl and alkoxy being unsubstituted or substituted with 1 to 3 halogen or deuterium atoms;
R5,R6each independently selected from hydrogen atom, deuterium atom, C1-6Alkyl, or C3-6Cycloalkyl, said alkyl being unsubstituted or substituted by 1 to 3 substituents selected from the group consisting of halogen, deuterium atom, hydroxy, amino, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、NHCOC1-6Alkyl, NHCOOC1-6Alkyl radical, C1-3Substituent substitution of alkoxy; or R4And R5And the atoms to which they are attached may be linked to each other to form a ring;
R7selected from hydrogen atoms, C1-6Alkyl radical, C3-6Cycloalkyl, or C3-6Heterocycloalkyl, said alkyl being unsubstituted or substituted by 1 to 3 atoms selected from halogen, deuterium, C3-6Cycloalkyl, or C3-6A substituent of a heterocycloalkyl group, said heterocycloalkyl group containing 1O atom;
R8,R9each independently selected from hydrogen atom, deuterium atom, cyano group、C1-6Alkyl, or C3-6Cycloalkyl, said alkyl being unsubstituted or substituted by 1 to 3 substituents selected from halogen, deuterium atom, hydroxy, C1-3Substituent substitution of alkoxy; or R8,R9And the atoms to which they are attached may be linked to each other to form a ring;
R10selected from hydrogen atom, deuterium atom, halogen, cyano, hydroxy, amino, C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-8Alkoxy, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C(O)NHC1-6Alkyl, C (O) N (C)1-6Alkyl radical)2、SC1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, 4-7 membered heterocyclyl, 6 membered aryl, 5-6 membered heteroaryl, said alkyl, alkoxy, heterocyclyl, aryl, heteroaryl being unsubstituted or substituted by 1-3 substituents selected from halogen, deuterium atom, hydroxy, C1-3Substituent substitution of alkoxy;
R11selected from hydrogen atoms, C1-6Alkyl radical, C3-6Cycloalkyl, said alkyl being unsubstituted or substituted by 1 to 3 substituents selected from the group consisting of halogen, deuterium atom, hydroxy, amino, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C1-3Substituent substitution of alkoxy;
R12,R13,R14,R15,R18,R19,R20,R21,R23,R24each independently selected from hydrogen atom, deuterium atom, cyano group, hydroxyl group, C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-3Alkoxy, said alkyl and alkoxy being unsubstituted or substituted by 1 to 3 substituents selected from the group consisting of halogen, deuterium atom, hydroxy, amino, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、NHCOC1-6Alkyl, NHCOOC1-6Alkyl radical, C1-3Substituent substitution of alkoxy; or R11And R14And the atoms to which they are attached may be linked to each other to form a ring; or R19And R12Or R15And the atoms to which they are attached may be linked to each other to form a ring;
R16,R17are respectively independentIs selected from hydrogen atom and C1-6Alkyl radical, C3-6Cycloalkyl radical, C3-6Heterocycloalkyl, COC1-6Alkyl, COOC1-6Alkyl, CONHC1-6Alkyl which is unsubstituted or substituted by 1 to 3 substituents selected from halogen, deuterium atom, hydroxy, cyano, C3-6Cycloalkyl radical, C1-3Alkoxy radical, C3-6(ii) a substituent of a heterocycloalkyl group containing 1 heteroatom selected from O or N; or R16And R17And the atoms to which they are attached may be joined to each other to form a 5-7 membered heterocyclic ring; or R16And R15And the atoms to which they are attached may be joined to each other to form a 5-7 membered heterocyclic ring;
R22selected from hydrogen atoms, C1-6Alkyl radical, C3-6Cycloalkyl radical, C3-8Heterocycloalkyl, said alkyl, cycloalkyl or heterocycloalkyl being unsubstituted or substituted by 1 to 3 atoms chosen from halogen, deuterium, hydroxy, cyano, C3-6Cycloalkyl radical, C1-3Alkoxy radical, C1-6Sulfone group, C1-6Acyl radical, C3-6(ii) a substituent of a heterocycloalkyl group containing 1 heteroatom selected from O or N; or R22And R15And the atoms to which they are attached may be linked to each other to form a ring.
In the above heterocyclic compounds having CXCR4 inhibitory activity, preferably, W thereof is selected from
Figure BDA0001789890150000061
Figure BDA0001789890150000062
Wherein X, Y and Z are respectively and independently selected from CR25R26Or O; r25,R26Each independently selected from hydrogen atom, deuterium atom, halogen, C1-3An alkyl group; or R25And R26And the carbon atoms to which they are attached may be linked to each other to form a ring.
In the above heterocyclic compounds having CXCR4 inhibitory activity, preferably, X, Y, Z are each independently selected from CH2,O,
Figure BDA0001789890150000063
And at least one of X, Y and Z is
Figure BDA0001789890150000064
Among the above heterocyclic compounds having CXCR4 signaling pathway inhibitory activity, A is preferred3Is independently selected from CR10,A1,A2Each independently selected from N or CR10And A is1,A2At least one is N;
w is
Figure BDA0001789890150000065
In the above heterocyclic compounds having CXCR4 inhibitory activity, W is preferably selected from unsubstituted or substituted by 1 to 3 groups selected from deuterium atom, cyano group, halogen, C1-3Alkyl radical, C1-3Substituents of alkoxy groups the following groups are substituted:
Figure BDA0001789890150000071
in the above heterocyclic compounds having CXCR4 inhibitory activity, preferably, U is unsubstituted or 1 to 3 independently selected from deuterium atom, halogen, hydroxy, C1-3Alkyl radical, C1-3Substituent of alkoxy group any of the following groups:
Figure BDA0001789890150000081
among the above heterocyclic compounds having CXCR4 inhibitory activity, preferred are heterocyclic compounds including:
Figure BDA0001789890150000082
Figure BDA0001789890150000091
Figure BDA0001789890150000101
Figure BDA0001789890150000111
Figure BDA0001789890150000121
Figure BDA0001789890150000131
Figure BDA0001789890150000141
the present invention also provides a pharmaceutical composition comprising a therapeutically effective amount of one or more heterocyclic compounds having CXCR4 pathway inhibitory activity of any of the above and pharmaceutically acceptable salts, isotopes, isomers, crystalline forms thereof, and further comprising at least one pharmaceutically acceptable carrier.
The invention also provides a combined application composition, which comprises a composition obtained by combining the heterocyclic compound with CXCR4 channel inhibitory activity and pharmaceutically acceptable salts, isotopes, isomers or crystal forms thereof with one or more of antitumor drugs, antibacterial drugs, antiviral drugs, central nervous system drugs and diabetes drugs.
The invention also provides the heterocyclic compound with CXCR4 inhibitory activity and the application of the pharmaceutically acceptable salt, isotope, isomer or crystal form thereof in the preparation of CXCR4 antagonistic drugs;
according to a specific embodiment, preferred isotopes of the invention include, but are not limited to2H,3H,11C,13C,14C,15N,17O,18O,18F,32P,35S,36Cl, and the like. Various isomers, including but not limited to stereoisomers, cis-trans isomers, tautomers, and the like.
The invention has the outstanding effects that:
the heterocyclic compounds having CXCR4 inhibitory activity of the present invention are the preferred results obtained after a large number of compound screens, in which the position of the U group attachment is important when the U group is replaced with A3When linked (comparative compound B4), the inhibitory activity of CXCR4 was lost; furthermore, the two N's on the U group are also important, and the removal of one N (comparative compound B3) or the absence of N (comparative compound B2) both lose the inhibitory activity of CXCR 4; at the same time, the position of the N on the pyrimidine ring is critical for CXCR4 activity, and substitution of the pyrimidine ring for a pyridine ring (comparative compounds B5 and B6) or for other linked pyrimidine rings (comparative compounds B7 and B8) all lost the inhibitory activity of CXCR 4. The heterocyclic compounds of the present invention, as potent antagonists of CXCR4, can be used to treat or prevent conditions responsive to CXCR4 receptor antagonism; the heterocyclic compound with CXCR4 inhibitory activity has weak inhibition capability on CYP liver enzyme, and has better drug interaction safety compared with a clinical compound AMD 070.
Drawings
FIG. 1 is a graph showing the results of the test for heterocyclic compound A42 in example 62;
FIG. 2 is a graph showing the results of the test for heterocyclic compound A43 in example 62;
FIG. 3 is a graph showing the results of the test for heterocyclic compound A78 in example 63;
FIG. 4 is a graph showing the results of the test for heterocyclic compound A83 in example 63.
Detailed Description
The technical solutions of the present invention will be described in detail below in order to clearly understand the technical features, objects, and advantages of the present invention, but the present invention is not limited to the practical scope of the present invention. The experimental methods described in the following examples are all conventional methods unless otherwise specified; the reagents and materials are commercially available, unless otherwise specified.
In the following examples, the solvents and drugs used are either analytically or chemically pure; the solvent is redistilled before use; the anhydrous solvent is treated according to standard or literature methods. Column chromatography silica gel (100-200 mesh) and thin layer chromatography silica gel (GF254) are products of Qingdao oceanic plant and tobacco station chemical plant; petroleum ether (60-90 ℃)/ethyl acetate (v/v) were used as eluents unless otherwise specified; the color developing agent is an ethanol solution of iodine or phosphomolybdic acid; all extraction solvents are, unless otherwise stated, anhydrous Na2SO4And (5) drying.1HNMR were recorded using a varian-400 NMR spectrometer with TMS as an internal standard. LC-MS was recorded using an Agilent model 1100 high performance liquid chromatography-ion trap Mass spectrometer (LC-MSDTrap), Diode Array Detector (DAD), detection wavelengths 214nm and 254nm, ion trap Mass Spectrometry (ESI source). HPLC column is AgelaDurashellC18 (4.6X 50mm, 3.5 μm); the mobile phase was 0.1% aqueous NH4HCO3 solution acetonitrile (5: 95 to 95: 5 in 5 minutes); the flow rate was 1.8 mL/min.
Example 1
A heterocyclic compound a1 synthesized by the method comprising:
Figure BDA0001789890150000161
1) synthesis of intermediate A1-2:
intermediate A1-1(280mg, 1.9mmol, see WO2016128529) was dissolved in dichloromethane (10mL), thionyl chloride (280mg, 2.3mmol) was added dropwise over ice, stirring was continued overnight, sodium bicarbonate solution was added, dichloromethane (50mL) was extracted three times, the dichloromethane phase was dried and spun dry to give a pale yellow oil (180mg, 58%).
2) Synthesis of intermediate A1-4:
intermediate a1-3(160mg, 1mmol, see synthetic literature WO2006026703), intermediate a1-2(170mg, 1.05mmol), potassium iodide (16mg, 0.1mmol) and N, N-diisopropylethylamine (320mg, 2.5mmol) were dissolved in 10mL acetonitrile, stirred at room temperature overnight, extracted with 100mL dichloromethane 50mL aqueous sodium bicarbonate solution, the dichloromethane phase was dried and spun dry, and the residue was purified by column chromatography (dichloromethane: methanol 100: 1 to 50: 1) to give a brown oily liquid (200mg, 69%).
3) Synthesis of product a 1:
intermediate a1-4(100mg, 0.35mmol) was dissolved in 4mL ethanol, triethylamine (350mg, 3.5mmol) and N-methylpiperazine (40mg, 0.38mmol) were added, the mixture was stirred at 80 ℃ overnight, 100mL dichloromethane 50mL aqueous sodium bicarbonate solution was added for extraction, the dichloromethane phase was dried and spun dry, and the residue was purified by column chromatography (dichloromethane: methanol 50: 1) to give a colorless oily liquid (98mg, 80%).
Example 2
Heterocyclic compounds a2, a5, a6, a22-a26, a39 and a60 were synthesized according to the synthesis method of example 1, substituting the corresponding amines for N-methylpiperazine in the last step.
Example 3
A heterocyclic compound a3 synthesized by the method comprising:
Figure BDA0001789890150000171
1) synthesis of intermediate A3-2:
ethyl 4-chloroacetoacetate (13g, 79mmol), S-methylisothiouronium sulfate (20g, 72mmol), dissolved in 200mL of water, sodium carbonate (11.5g, 108mmol) was added, stirred overnight at room temperature, pH adjusted to acidic with 6N HCl, the solid precipitated, filtered, and dried to give the product (11.1g, 73%) as a white solid.
2) Synthesis of intermediate A3-3:
intermediate A3-2(2.8g, 14.7mmol), dissolved in 10mL phosphorus oxychloride, stirred at 100 ℃ for 1h, dried, extracted with 100mL ethyl acetate 100mL aqueous sodium bicarbonate solution, and the ethyl acetate phase dried and dried. The residue was purified by column chromatography (ethyl acetate: petroleum ether: 100: 3) to give the product (2.4g, 80%) as a yellow oily liquid.
3) Synthesis of intermediate A3-4:
intermediate A1-3(400mg, 2.5mmol), A3-3(540mg, 2.6mmol), potassium iodide (41mg, 0.25mmol) and N, N-diisopropylethylamine (800mg, 6.2mmol) were dissolved in 20mL acetonitrile, stirred overnight at room temperature, extracted with 100mL dichloromethane and 100mL aqueous sodium bicarbonate, and the dichloromethane phase was dried and dried. The residue was purified by column chromatography (dichloromethane: methanol 200: 1) to give the product (450mg, 54%) as a brown liquid.
4) Synthesis of product a 3:
intermediate a3-4(450mg, 1.3mmol) was dissolved in 10mL ethanol, triethylamine (1.3g, 13mmol) and N-methylpiperazine (150mg, 1.4mmol) were added, stirred overnight at 80 ℃, 100mL dichloromethane and 10mL aqueous sodium bicarbonate solution were added for extraction, the dichloromethane phase was dried and spun dry to give a liquid which was purified by column chromatography (dichloromethane: methanol 50: 1) to give the product (500mg, 96%) as a brown liquid.
Example 4
A heterocyclic compound a7 synthesized by the method comprising:
Figure BDA0001789890150000181
1) synthesis of intermediate A7-1:
intermediate A1-4(200mg, 0.7mmol) was dissolved in 10mL ethanol, triethylamine (700mg, 7mmol) and N-Boc piperazine (140mg, 0.77mmol) were added, and the mixture was stirred at 80 ℃ overnight. 50mL of dichloromethane and 50mL of aqueous sodium bicarbonate solution were added and the dichloromethane phase was dried and spun dry. The residue was purified by column chromatography (dichloromethane: methanol 50: 1 to 25: 1) to give the product (290mg, 94%) as a colorless oil.
2) Synthesis of product a 7:
intermediate A7-1(270mg, 0.6mmol), dissolved in 5mL of dichloromethane and hydrogen chloride in ethyl acetate (5mL), was stirred overnight at room temperature, extracted with 50mL of dichloromethane and 50mL of aqueous sodium bicarbonate, and the dichloromethane phase was dried and spun dry. The residue was purified by column chromatography (dichloromethane: methanol 50: 1 to 25: 1) to give a colorless oily liquid (180mg, 90%).
Example 5
A heterocyclic compound A8 synthesized by the method comprising:
Figure BDA0001789890150000182
a7(50mg, 0.13mmol) was dissolved in 2mL of methanol, triethylamine (15mg, 0.13mmol) and acrylonitrile (20mg, 0.26mmol) were added, and the mixture was stirred at room temperature overnight. 50mL of dichloromethane and 50mL of aqueous sodium hydrogencarbonate are added and the dichloromethane phase is dried and dried. The residue was purified by column chromatography (dichloromethane: methanol 50: 1 to 25: 1) to give a colorless oily liquid (40mg, 78%).
Example 6
A heterocyclic compound a9 synthesized by the method comprising:
Figure BDA0001789890150000191
1) synthesis of intermediate A9-2:
a9-1(300mg, 1.7mmol) and acetamidine hydrochloride (320mg, 3.4mmol) were dissolved in 10mL of water, potassium carbonate (940mg, 6.8mmol) was added, and the mixture was stirred at room temperature overnight. Acetic acid was added to adjust the pH to acidity, dichloromethane (100mL) was extracted three times, and the organic phase was dried by spin drying. The residue was purified by column chromatography (dichloromethane: methanol 50: 1) to give a white solid (110mg, 35%).
2) Synthesis of intermediate A9-3:
a9-2(110mg, 0.6mmol), triethylamine (600mg, 6mmol), N-methylpiperazine (90mg, 0.9mmol) were dissolved in 10mL acetonitrile, PyBOP (340mg, 0.7mmol) was added, stirring was performed at reflux overnight, 100mL dichloromethane and 100mL aqueous sodium bicarbonate were added for extraction, and the dichloromethane phase was dried and dried. The residue was purified by column chromatography (dichloromethane: methanol 100: 3) to give a yellow oily liquid (140mg, 88%).
3) Synthesis of intermediate A9-4:
a9-3(140mg, 0.5mmol) was dissolved in 5mL of 20% aqueous sulfuric acid, stirred at reflux overnight, and then an aqueous solution of sodium bicarbonate was added to adjust the pH to basic, followed by extraction with dichloromethane, and the dichloromethane phase was dried and spun dry to give a yellow oily liquid (110mg, 97%).
4) Synthesis of product a 9:
a1-3(37mg, 0.23mmol), A9-4(46mg, 0.21mmol) and acetic acid (13mg, 0.21mmol) were dissolved in dichloroethane (5mL), and after stirring at room temperature for 10min, sodium borohydride acetate (66mg, 0.3mmol) was added, and stirring was continued overnight. 100mL of dichloromethane and 100mL of aqueous sodium bicarbonate solution are added and the dichloromethane phase is dried and dried. The residue was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 50: 1) to give a colorless oily liquid (30mg, 39%).
Example 7
The heterocyclic compounds A10, A11, A28 and A29 were synthesized according to the synthesis method of example 6, substituting the corresponding substrate for acetamidine hydrochloride in step 1.
Example 8
According to the synthesis method of example 6, using the corresponding substrate in place of A9-1 and S-methylisothiourea sulfate in place of acetochlor hydrochloride in step 1, a heterocyclic compound A27 was synthesized.
Example 9
According to the synthesis method of example 6, a heterocyclic compound A48 was synthesized by substituting trifluoroacetamide for acetamide hydrochloride in step 1 and A42-1 for A1-3 in step 4.
Example 10
The heterocyclic compounds A57 and A59 were synthesized according to the synthesis method of example 6, substituting the corresponding substrate for A1-3 in step 4.
Example 11
A heterocyclic compound a12 synthesized by the method comprising:
Figure BDA0001789890150000201
1) synthesis of intermediate A12-1:
a9-1(3g, 17mmol), S-methylisothiouronium sulfate (9.5g, 34mmol), dissolved in 100mL of water, potassium carbonate (17.6g, 76.5mmol) added, stirred overnight at room temperature, adjusted to acidic pH by the addition of acetic acid, extracted three times with dichloromethane (100 mL). The organic phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a white solid (3.4g, 92%).
2) Synthesis of intermediate A12-2:
a12-1(3.4g, 16mmol), triethylamine (16g, 160mmol), N-methylpiperazine (2.4g, 40mmol) were dissolved in 100mL acetonitrile, PyBOP (9g, 17.6mmol) was added, stirring was performed at reflux overnight, extraction was performed with 200mL dichloromethane and 200mL aqueous sodium bicarbonate, and the dichloromethane phase was dried and dried. The residue was purified by column chromatography (dichloromethane: methanol 100: 3) to give a yellow oily liquid (2.8g, 59%).
3) Synthesis of intermediate A12-3:
a12-2(2.8g, 9.4mmol) was dissolved in 90mL tetrahydrofuran and 4.5mL water, and Oxone (7g, 11mmol) was added under stirring at room temperature, and stirred at room temperature for 4h, followed by extraction with 200mL ethyl acetate and 200mL aqueous sodium bicarbonate. The ethyl acetate phase was dried and spun dry to give a yellow oily liquid (2.4g, 78%).
4) Synthesis of intermediate A12-4:
a12-3(340mg, 1mmol), tetrahydrofuran solution in dimethylamine (5mL), was stirred under reflux overnight in a sealed tube and extracted by adding 100mL of dichloromethane and 100mL of aqueous sodium bicarbonate. The organic phase was dried and the residue purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 100: 3: 1) to give a yellow oily liquid (260mg, 88%).
5) Synthesis of intermediate A12-5:
a12-4(260mg, 1.3mmol) was dissolved in 5mL of 20% aqueous sulfuric acid, stirred at reflux overnight, an aqueous solution of sodium bicarbonate was added to adjust the pH to alkaline, and dichloromethane was added for extraction. The dichloromethane phase was dried and spun dry to give a yellow solid (210mg, 65%).
6) Synthesis of product a 12:
a1-3(150mg, 0.9mmol), A12-5(210mg, 0.84mmol) and acetic acid (50mg, 0.84mmol) were dissolved in dichloroethane (5mL), and after stirring at room temperature for 10min, sodium borohydride acetate (270mg, 1.3mmol) was added, stirring was continued overnight, and 100mL of dichloromethane and 100mL of aqueous sodium bicarbonate were added for extraction. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 50: 1) to give a colourless oily liquid (60mg, 18%).
Example 12
The heterocyclic compounds a13-a16 and a18-a20 were synthesized according to the synthesis of example 11, substituting the corresponding amine for dimethylamine in step 4.
Example 13
A heterocyclic compound a17 synthesized by the method comprising:
Figure BDA0001789890150000211
1) synthesis of intermediate A17-1:
in a 25mL two-necked flask, A12-2(298mg, 1.00mmol), phenylboronic acid (244mg, 2.00mmol), cuprous thiophene-2-carboxylate (497mg, 2.60mmol), palladium tetrakistriphenylphosphine (115mg, 0.10mmol) and tetrahydrofuran (20mL) were added in that order. After nitrogen substitution was carried out three times, the reaction was carried out at 80 ℃ for 12 hours. The solvent was dried and the solute was purified by column chromatography (mobile phase dichloromethane: methanol 100: 1.5) to give a colorless oil (240mg, 73%).1H NMR(400MHz,CDCl3)δ8.41(d,J=3.6Hz,2H),7.44-7.43(m,3H),6.70(s,1H),5.24(s,1H),3.82-3.80(m,4H),3.47(s,6H),2.53-2.52(m,4H),2.37(s,3H).
2) Synthesis of intermediate A17-2:
a17-1(240mg, 0.73mmol) was dissolved in 20% sulfuric acid solution (10 mL). After reaction at 80 ℃ for 12h, it was cooled to room temperature, the aqueous phase was washed with diethyl ether (10mL × 1), the aqueous phase was adjusted to pH 10 with 6M sodium hydroxide solution and extracted with dichloromethane (30mL × 3). The organic phases were combined, dried over anhydrous sodium sulfate and spin-dried. The solute was purified by column chromatography (mobile phase dichloromethane: methanol 100: 2) to give a colorless oil (170mg, 82%).
2) Synthesis of product a 17:
a17-2(170mg, 0.605mmol), A1-3(98mg, 0.605mmol) and acetic acid (36mg, 0.60mmol) were dissolved in 1mL 1, 2-dichloroethane. After stirring at room temperature for 30min, sodium triacetoxyborohydride (321mg, 1.51mmol) was added. Stirring at normal temperature for 12h, and spin-drying the solvent. The solute was purified by column chromatography (mobile phase dichloromethane: methanol 100: 1) to give a colorless oil (52mg, 20%).
Example 14
According to the synthesis method of example 13, a heterocyclic compound a21 was synthesized by substituting 1-methyl-1H-pyrazole-4 boronic acid for phenylboronic acid in step 1.
Example 15
A heterocyclic compound a30 synthesized by the method comprising:
Figure BDA0001789890150000221
1) synthesis of intermediate A30-1:
a12-3(200mg, 0.61mmol) was dissolved in ethanol (5mL), sodium ethoxide (206mg, 3mmol) was added, and stirring was performed at reflux overnight. 100mL of methylene chloride and 100mL of aqueous sodium bicarbonate solution were added and extracted, and the organic phase was dried by spin-drying. The residue was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 100: 3: 1) to give a yellow oily liquid (160mg, 89%).
2) Synthesis of intermediate A30-2:
a30-1(160mg, 0.5mmol) was dissolved in 5mL of 20% aqueous sulfuric acid and stirred at reflux overnight. Adding sodium bicarbonate aqueous solution to adjust the pH to be alkaline, and extracting by dichloromethane. The dichloromethane phase was dried and spun dry to give a yellow solid (100mg, 80%).
3) Synthesis of product a 30:
a1-3(36mg, 0.22mmol), A30-2(50mg, 0.2mmol) and acetic acid (12mg, 0.2mmol) were dissolved in dichloroethane (5mL), and after stirring at room temperature for 10min, sodium borohydride acetate (64mg, 0.3mmol) was added, and stirring was continued overnight. 100mL of methylene chloride and 100mL of aqueous sodium bicarbonate solution were added and extracted. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 50: 1) to give a colourless oily liquid (40mg, 51%).
Example 16
The synthesis according to example 15, substituting isopropanol for ethanol in step 1, gave heterocyclic compound a 31.
Example 17
Heterocyclic compounds a32 and a33, which were synthesized by the following method:
Figure BDA0001789890150000231
1) synthesis of intermediate A32-2:
a32-1(5g, 26mmol), 2, 2-dimethyl-1, 3-dioxane-4, 6-dione (4.2g, 29mmol) and DMAP (5.0g, 40mmol) were dissolved in 100mL of dichloromethane, and DCC (6g, 29mmol) was added with stirring in an ice bath and stirred at room temperature for 48 h. Filtration was carried out, and the filtrate was extracted with 200mL of methylene chloride and 100mL of 1N hydrochloric acid solution. The dichloromethane phase was dried and spun to give a yellow oily liquid (6g, crude) which was used directly in the next step.
2) Synthesis of intermediate A32-3:
a32-2(6g, crude) was dissolved in 100mL absolute ethanol and stirred under reflux for 2 h. Concentrated under reduced pressure, and the residue was purified by column chromatography (petroleum ether: ethyl acetate 4: 1) to give a yellow oily liquid (640mg, 10% yield in two steps).
3) Synthesis of intermediate A32-4:
a32-3(640mg, 2.5mmol) and S-methylisothiouronium sulfate (696mg, 5mmol) were dissolved in 40mL of water, potassium carbonate (1.3g, 10mmol) was added, and the mixture was stirred at room temperature overnight. Acetic acid was added to adjust the pH to acidity, and dichloromethane (100mL) was extracted three times. The organic phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 20: 1) to give a colourless oily liquid (560mg, 74%).
4) Synthesis of intermediate A32-5:
a32-4(556mg, 1.9mmol), triethylamine (1.9g, 19mmol), N-methylpiperazine (280mg, 2.8mmol) were dissolved in 5mL acetonitrile, PyBOP (1g, 2mmol) was added and the mixture was stirred at reflux overnight. 50mL of methylene chloride and 50mL of aqueous sodium bicarbonate solution were added and extracted. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a yellow oily liquid (362mg, 52%).
5) Synthesis of intermediate A32-6:
a32-5(362mg, 1mmol) was dissolved in 5mL of ethyl acetate, and a solution of ethyl acetate hydrogen chloride (5mL) was added and stirred at room temperature overnight. Concentrated under reduced pressure and extracted by adding 100mL of methylene chloride and 50mL of aqueous sodium bicarbonate solution. The organic phase was dried and concentrated under reduced pressure to give a colorless oily liquid (170mg, 64%).1H NMR(400MHz,CDCl3)δ6.20(s,1H),3.85-3.83(m,1H),3.67(s,4H),2.51(s,3H),2.46(s,4H),2.34(s,3H),1.36(d,J=6.4Hz,3H).
6) Synthesis of intermediate A32-8:
a32-6(60mg, 0.24mmol), A32-7(47mg, 0.32mmol) and catalytic amount of acetic acid were dissolved in 1, 2-dichloroethane and the reaction was stirred for half an hour. Sodium triacetoxyborohydride (136mg, 0.64mmol) was added and the reaction was allowed to proceed overnight at room temperature. Adding water for quenching. Filtration and three extractions of the filtrate with dichloromethane (10mL x 3). The combined organic phases were dried, filtered and concentrated under reduced pressure to give a yellow oily liquid (90mg, crude).
7) Synthesis of product a 32:
a32-8(80mg, crude), 37% formaldehyde (33mg, 0.4mmol) and acetic acid (18mg, 0.3mmol) were dissolved in dichloroethane (4 mL). After stirring for half an hour, sodium triacetoxyborohydride (94mg, 0.3mmol) was added and stirred at room temperature overnight. Add water to quench and filter. The filtrate was extracted three times with dichloromethane (10mL x 3). The combined organic phases were dried, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (dichloromethane: methanol: aqueous ammonia 250: 1) to give product a32(14mg, 16%) and product a33(20mg, 23%) as colorless oily liquids.
Example 18
Heterocyclic compounds a34 and a35, which were synthesized by the following method:
Figure BDA0001789890150000241
1) synthesis of intermediate A34-2:
A34-1(500mg, 2.6mmol) was dissolved in tetrahydrofuran (30mL), sodium hydride (60%, 624mg, 15.6mmol) was added, the mixture was stirred for 10 minutes, methyl iodide (3.7g, 26mmol) was slowly added, and the reaction was carried out at room temperature under nitrogen atmosphere overnight. The reaction was quenched by the addition of water (3mL) and ethyl acetate (2 mL). Concentrated under reduced pressure and extracted with ethyl acetate (2 x10 mL). The aqueous phase was adjusted to PH 3.5 with 2N HCl, extracted with ethyl acetate (3 x10 mL), and the organic phases were combined, dried, filtered, and concentrated under reduced pressure to give a yellow solid (640mg, crude).1H NMR(400MHz,CDCl3)δ4.78-4.47(m,1H),2.84(s,3H),1.46-1.42(m,12H).
2) Synthesis of intermediate A34-3:
a34-2(640mg, 3.1mmol), 2, 2-dimethyl-1, 3-dioxane-4, 6-dione (490mg, 3.4mmol) and DMAP (562mg, 4.6mmol) were dissolved in 5mL of dichloromethane, and DCC (701mg, 3.4mmol) was added with stirring in an ice bath and stirred at room temperature for 48 h. After filtration, 20mL of methylene chloride and 10mL of 1N hydrochloric acid solution were added to the filtrate to separate layers, followed by extraction. The dichloromethane phase was dried and spun dry and the residue was dissolved in 10mL of absolute ethanol and stirred under reflux for 2 h. The ethanol was concentrated under reduced pressure, and the residue was purified by column chromatography (petroleum ether: ethyl acetate 4: 1) to give a yellow oily liquid (590mg, 70%).
3) Synthesis of intermediate A34-4:
a34-3(570mg, 2.1mmol) and S-methylisothiouronium sulfate (584mg, 4.2mmol) were dissolved in 6mL of water, potassium carbonate (1.2g, 8.4mmol) was added, and the mixture was stirred at room temperature overnight. Acetic acid was added to adjust the pH to acidity, and dichloromethane (20mL) was extracted three times. The organic phases were combined, dried and spin dried. The residue was purified by column chromatography (dichloromethane: methanol 20: 1) to give a white solid (380mg, 60%).
4) Synthesis of intermediate A34-5:
a34-4(380mg, 1.2mmol), triethylamine (1.2g, 12mmol), N-methylpiperazine (190mg, 1.9mmol) were dissolved in 5mL acetonitrile, PyBOP (728mg, 1.4mmol) was added and the mixture was stirred at reflux overnight. 50mL of methylene chloride and 50mL of aqueous sodium bicarbonate solution were added and extracted. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a yellow oily liquid (340mg, 74%).1H NMR(400MHz,CDCl3)δ6.05(s,1H),5.2-5.0(m,1H),3.63(s,4H),3.16(s,1H),2.78(s,3H),2.49(s,3H),2.46-2.05(m,4H),2.33(s,3H),1.44(s,12H).
5) Synthesis of intermediate A34-6:
a34-5(340mg, 0.89mmol) was dissolved in 5mL of ethyl acetate, and an ethyl acetate solution (5mL) of hydrogen chloride was added thereto and the mixture was stirred at room temperature overnight. The ethyl acetate was dried by spinning, and extracted with 100mL of dichloromethane and 50mL of aqueous sodium bicarbonate. The organic phase was dried and spun to give a yellow oily liquid (170mg, 68%).1H NMR(400MHz,CDCl3)δ6.20(s,1H),3.68(s,4H),2.50(s,3H),2.47-2.45(m,4H),2.34(s,3H),2.32(s,3H),1.32(d,J=6.4Hz,3H).
6) Synthesis of products a34 and a 35:
a34-6(100mg, 0.35mmol), A34-7(117mg, 0.7mmol), potassium iodide (20mg, 0.12mmol), N, N-diisopropylethylamine (1.4g, 3.5mmol) was dissolved in N-methylpyrrolidone (4mL) and reacted at 120 ℃ overnight. Cooled to room temperature and extracted with ethyl acetate (5 × 10 mL). The combined organic phases were washed 5 times with saturated brine, dried, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (dichloromethane: methanol: ammonia 250: 1) to give a34(10mg, 7%) and a35(20mg, 14%) as colorless oily liquids.
Example 19
A heterocyclic compound a37 synthesized by the method comprising:
Figure BDA0001789890150000261
1) synthesis of intermediate A37-1:
ethyl 4-chloroacetoacetate (16.5g, 100mmol) and acetamidine hydrochloride (10g, 106mmol) were dissolved in ethanol (100mL) successively. DBU (30.4g, 200mmol) was added slowly in an ice-water bath and stirred overnight at room temperature. The solvent was drained, diluted with dichloromethane (120mL), washed with saturated brine (30mL × 3), the organic phase was dried and the solvent was drained. The residue was dissolved in POCl3(20mL) was stirred at 110 ℃ under reflux for 30 min. After cooling to room temperature, most of the POCl was removed3Saturated carbon for residueThe sodium hydrogen carbonate was adjusted to neutral, extracted with ethyl acetate (30mL x 3), the organic phases were combined and dried. After the solvent was dried by evaporation, the residue was purified by column chromatography on silica gel (PE: EA: 5: 1) to give a pale yellow solid (4.0g, 23%).1H NMR(400MHz,CDCl3)δ7.41(s,1H),4.55(s,2H),2.71(s,3H).
2) Synthesis of intermediate A37-3:
2-bromopyridine (1.5g, 9.7mmol) and TMEDA (2.1g, 17.4mmol) were dissolved in 30mL of anhydrous tetrahydrofuran, and after displacing nitrogen, the mixture was stirred at-78 ℃ for 10 minutes. 2.5M n-butyllithium (3.2mL, 8.1mmol) was added thereto, and after stirring for 2 hours, 1-Boc-2-pyrrolidone (1g, 5.4mmol) was added thereto, and the reaction was continued for 2 hours, followed by quenching with water (10mL), returning to room temperature, and then extraction with ethyl acetate. And drying the ethyl acetate phase and spin-drying. The residue was purified by column chromatography (dichloromethane: methanol 250: 1) to give a white solid (900mg, 63%).
2) Synthesis of intermediate A37-4:
a37-3(500mg, 1.9mmol) was dissolved in methylamine in tetrahydrofuran (7.6mL), tetraethyl titanate (1.1g, 3.8mmol) was added, and after stirring at room temperature for 30 minutes, sodium borohydride (290mg, 7.6mmol) was added. Stirring was continued for 2 hours, quenched by the addition of aqueous sodium bicarbonate (100mL), and extracted three times with dichloromethane (100 mL). The organic phase was dried and dried, and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a colorless oily liquid (270mg, 51%).
3) Synthesis of intermediate A37-5:
a37-4(100mg, 0.36mmol), A37-1(70mg, 0.39mmol), potassium iodide (6mg, 0.04mmol) and N, N-diisopropylethylamine (120mg, 0.9mmol) were dissolved in 5mL of acetonitrile and stirred at room temperature overnight. 50mL of methylene chloride and 50mL of aqueous sodium bicarbonate solution were added and extracted. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a colorless oily liquid (110mg, 63%).
4) Synthesis of product a 37:
a37-5(110mg, 0.2mmol), triethylamine (230mg, 2mmol), N-methylpiperazine (115mg, 1mmol) were dissolved in 3mL ethanol and stirred at reflux overnight. 100mL of methylene chloride and 100mL of aqueous sodium bicarbonate solution were added and extracted. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1) to give a colourless oily liquid (90mg, 93%).
Example 20
The synthesis according to example 19, substituting 1-Boc-2-piperidone for 1-Boc-2-pyrrolidone in step 2, gave heterocyclic compound A36.
Example 21
A heterocyclic compound a38 synthesized by the method comprising:
Figure BDA0001789890150000271
a37(60mg, 0.12mmol) was dissolved in 3mL of ethyl acetate, and a solution of ethyl acetate hydrogen chloride (4mL) was added thereto and the mixture was stirred at room temperature overnight. Filtration and drying gave A38 as a white solid as the hydrochloride salt (90 mg).
Example 22
A heterocyclic compound a41 synthesized by the method comprising:
Figure BDA0001789890150000281
1) synthesis of intermediate A41-1:
to a50 mL two-necked flask was added sodium hydride solid (60%, 420mg, 10.5mmol) and the solvent DMF (10 mL). Nitrogen blanket and replace air. A32-7(441mg, 3.0mmol) and 1, 2-dibromoethane (1.95g, 10.5mmol) were dissolved in DMF under ice-water bath and then added to the reaction system in this order. After stirring for 1 hour, the mixture was diluted with dichloromethane (40mL) and washed with saturated brine (40 mL). The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE: EA: 10: 3) to give a white solid (110mg, 21%).1H NMR(400MHz,CDCl3)δ8.68(s,1H),7.64(s,1H),7.36(s,1H),3.02(d,J=4.4Hz,2H),2.01(d,J=4.8Hz,2H),1.50(s,2H),0.87(s,2H).
2) Synthesis of intermediate A41-2:
a41-1(110mg, 0.64mmol), a tetrahydrofuran solution (3mL) in methylamine, tetraethyl titanate (1.14g, 5mmol) added and stirred at 70 ℃ overnight, sodium cyanoborohydride (126mg, 1.3mmol) added and stirred at ambient temperature for 8 hours. The reaction was quenched by addition of water and filtered. The filtrate was diluted with dichloromethane (10mL), washed with water (10mL), the organic phase dried and dried, and purified by silica gel column chromatography (DCM: MeOH ═ 40: 1) to give a colorless oily liquid (40mg, 30%).
3) Synthesis of intermediate A41-3:
a41-2(30mg, 0.16mmol) and intermediate A37-1(31mg, 0.18mmol) were dissolved in acetonitrile (5mL), and potassium iodide (3mg, 0.016mmol) and DIPEA (52mg, 0.4mmol) were added in this order, followed by stirring at room temperature overnight. The solvent was dried by evaporation and the residue was purified by silica gel column chromatography (DCM: MeOH: 200: 3) to give a yellow oily liquid (30mg, 58%).
4) Synthesis of product a 41:
a41-3(35mg, 0.11mmol) and N-methylpiperazine (110mg, 1.1mmol) were dissolved in ethanol (5ml), triethylamine (101mg, 1.1mmol) was added, and the reaction was refluxed at 85 ℃ for 3 hours. After cooling to room temperature, methylene chloride (15ml) was added thereto for dilution, followed by washing with water (20 ml). The organic phase is dried, spun dry and chromatographed over silica gel (DCM: MeOH: NH)4OH 100: 3: 1) to give a colorless oily liquid (25mg, 64%).
Example 23
The heterocyclic compound a44 was synthesized according to the synthesis method of example 22, substituting ethylamine for methylamine in step 2.
Example 24
The synthesis of example 22 was followed, substituting N-ethylpiperazine for N-methylpiperazine in step 4, to afford heterocyclic compound a 45.
Example 25
A heterocyclic compound a40 synthesized by the method comprising:
Figure BDA0001789890150000291
1) synthesis of intermediate A40-1:
a12-2(100mg, 0.33mmol) was dissolved in 20% sulfuric acid solution (10mL) and reacted at 80 ℃ for 12 h. After cooling to room temperature, the aqueous phase was washed with ether (10mL), adjusted to pH 10 with 6M sodium hydroxide solution and extracted with dichloromethane (30mL × 3). The dichloromethane phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a colorless oil as a liquid (59mg, 71%).
2) Synthesis of product a 40:
a41-2(40mg, 0.21mmol) and A40-1(59mg, 0.23mmol) were dissolved in 1, 2-dichloroethane (3mL), and a catalytic amount of glacial acetic acid (0.2mL) was added thereto and the mixture was stirred at room temperature for 5 hours. Sodium triacetoxyborohydride (90mg, 0.42mmol) was then added thereto, and the mixture was stirred overnight. Dichloromethane (10mL) was added to dilute and the organic phase was washed with saturated aqueous sodium bicarbonate (20mL) and dried. The residue is chromatographed on silica gel (DCM: MeOH: NH)3H2O100: 1) to give a colorless oily liquid (49mg, 54%).
Example 26
A heterocyclic compound a42 synthesized by the method comprising:
Figure BDA0001789890150000292
1) synthesis of intermediate A42-2:
a42-1(90mg, 0.56mmol), A37-1(108mg, 0.61mmol), potassium iodide (10mg, 0.06mmol) and N, N-diisopropylethylamine (180mg, 1.4mmol) were dissolved in 10mL of acetonitrile, stirred overnight at room temperature, extracted with 100mL of dichloromethane and 50mL of aqueous sodium bicarbonate. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a pale yellow oily liquid (110mg, 65%).
2) Synthesis of product a 42:
a42-2(30mg, 0.1mmol) was dissolved in 2mL ethanol, triethylamine (100mg, 1mmol) and N-methylpiperazine (50mg, 0.5mmol) were added, the mixture was reacted overnight at 80 ℃, and extracted with 50mL dichloromethane and 50mL aqueous sodium bicarbonate. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a yellow oily liquid (36mg, 98%).
Example 27
The heterocyclic compounds a43, a47, a49 and a63-a74 were synthesized according to the synthesis of example 26 substituting the corresponding amine for N-methylpiperazine in step 2.
Example 28
According to the synthesis method of example 26, substituting the corresponding substrate for A42-1 in step 1, a heterocyclic compound A58 was synthesized.
Example 29
According to the synthesis method of example 26, substituting the corresponding substrate for A37-1 in step 1, a heterocyclic compound A61 was synthesized.
Example 30
A heterocyclic compound a46 synthesized by the method comprising:
Figure BDA0001789890150000301
1) synthesis of intermediate A46-2:
a46-1(667mg, 6mmol) was dissolved in ethanol (25mL) and propargylamine (1.3g, 24mmol) and sodium chloroaurate dihydrate (60mg, 0.15mmol) were added in that order. Stirring was carried out at 85 ℃ for 24 hours under nitrogen. After cooling to room temperature, the solvent was evaporated, and the mixture was dissolved in ethyl acetate (30mL), and washed once with a saturated sodium bicarbonate solution (15mL) and once with a saturated saline solution (15 mL). The organic phase was dried, the solvent was dried by evaporation, and the residue was purified by column chromatography on silica gel (petroleum ether: ethyl acetate 12: 1) to give a colorless oily liquid (118mg, 12%).1H NMR(400MHz,CDCl3)δ8.37(s,1H),7.30(d,J=7.2Hz,1H),7.03(t,J=4.0Hz,1H),3.00(t,J=6.4Hz,2H),2.64(s,2H),1.69(t,J=6.0Hz,2H),0.43(d,J=10.0Hz,4H).
2) Synthesis of intermediate A46-3:
a46-2(100mg, 0.63mmol) was dissolved in 1, 2-dichloromethane and trichloroisocyanuric acid (219mg, 0.95mmol) was added slowly. Stirring at 30 deg.C for five hours, cooling to room temperature, filtering, spin-drying the filtrate, and purifying by silica gel column chromatography (petroleum)Ether: ethyl acetate 10: 1) gave a colorless liquid (79mg, 65%).1H NMR(400MHz,CDCl3)δ8.54(s,1H),7.38(d,J=8.0Hz,1H),7.16(t,J=4.8Hz,1H),2.80(d,J=16Hz,1H),2.64(d,J=16Hz,1H),2.44-2.39(m,1H),2.09-2.04(m,1H),0.94-0.85(m,1H),0.65-0.56(m,1H),0.48-0.40(m,2H).
3) Synthesis of intermediate A46-4:
a46-3(30mg, 0.16mmol) was dissolved in an alcoholic methylamine solution (5 mL). The mixture was stirred at room temperature overnight, and then warmed to 60 ℃ and stirred for 5 hours. After cooling to room temperature, the solvent was spin-dried to give a colorless oily liquid (30mg, 99%).1H NMR(400MHz,CDCl3)δ8.45(s,1H),7.43(d,J=6.8Hz,1H),7.20-7.18(m,1H),4.34-4.24(m,1H),3.27(d,J=11.2Hz,1H),2.85(s,1H),2.71(s,3H),2.44(m,J=12.0Hz,1H),1.78-1.71(m,1H),0.67-0.43(m,4H).
4) Synthesis of intermediate A46-5:
a46-4(30mg, 0.17mmol) and A37-1(33mg, 0.19mmol) were dissolved in acetonitrile (2 mL). Potassium iodide (3mg, 0.017mmol) and DIPEA (55mg, 0.43mmol) were added in this order, followed by stirring at room temperature overnight. After drying the solvent, a liquid was obtained which was purified by column chromatography (DCM: MeOH: 100: 1) to give a pale yellow oily liquid (50mg, 89%).
6) Synthesis of product a 46:
a46-5(50mg, 0.15mmol) and N-methylpiperazine (76mg, 0.76mmol) were dissolved in ethanol (4 ml). Triethylamine (151mg, 1.5mmol) was added and the reaction was refluxed at 85 ℃ for 3 hours. After cooling to room temperature, methylene chloride (15ml) was added thereto for dilution, followed by washing with water (20 ml). The organic phase was dried and spun to give a liquid which was purified by column chromatography (DCM: MeOH: 100: 1 to 50: 1) to give a colorless oily liquid (25mg, 40%).
Example 31
A heterocyclic compound a50 synthesized by the method comprising:
Figure BDA0001789890150000321
1) synthesis of intermediate A50-2:
2-methyl-4-chloropyrimidine (370mg, 2.9mmol), NBS (566mg, 3.2mmol) and AIBN (50mg, 0.3mmol) were dissolved in carbon tetrachloride (10mL), stirred under reflux overnight, extracted with 50mL of methylene chloride and 50mL of aqueous sodium bicarbonate solution. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (50: 1 petroleum ether: ethyl acetate) to give a white solid (130mg, 22%).
2) Synthesis of intermediate A50-3:
a50-2(94mg, 0.58mmol), A1-3(130mg, 0.6mmol), potassium iodide (10mg, 0.06mmol) and N, N-diisopropylethylamine (740mg, 5.8mmol) were dissolved in 10mL acetonitrile, stirred overnight at 50 deg.C, extracted with 100mL dichloromethane and 50mL aqueous sodium bicarbonate. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a brown oily liquid (140mg, 84%).
3) Synthesis of product a 50:
a50-3(50mg, 0.17mmol) dissolved in 2mL NMP was added N, N-diisopropylethylamine (244mg, 1.7mmol), N-methylpiperazine (87mg, 0.85mmol), stirred at 80 ℃ overnight, extracted with 50mL dichloromethane and 50mL aqueous sodium bicarbonate. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1 to 100: 3) to give a pale yellow oily liquid (34mg, 57%).
Example 32
A heterocyclic compound a51 synthesized by the method comprising:
Figure BDA0001789890150000322
1) synthesis of intermediate A51-2:
NaH (60%, 936mg, 23.4mmol) was added to 50mL of diethyl ether, ethyl fluoroacetate (5g, 47.2mmol) was added dropwise at ordinary temperature, and then reacted at 40 ℃ for 4 hours. The reaction was poured into 2M H2SO4(15mL) at 0 ℃, followed by extraction with ether (50mL × 3), combined organic phases, dried over anhydrous sodium sulfate, and column chromatographed (petroleum ether: ethyl acetate 10: 1 to 2: 1) to give a yellow oil (2g, 25%).
2) Synthesis of intermediate A51-3:
a51-2(1.9g, 11.4mmol), acetamidine hydrochloride (2.2g, 22.8mmol) and sodium ethoxide (2.3g, 34.2mmol) were added to ethanol (40mL) and reacted at 80 ℃ overnight. Cooled to room temperature, 6N HCl (2mL) was added, concentrated under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate 3: 1 to 1: 1) to give a yellow solid (800mg, 43%).1H NMR(400MHz,CDCl3)δ13.07(br s,1H),5.35(d,J=46.8Hz,2H),2.53(s,3H).
3) Synthesis of intermediate A51-4:
a51-3(800mg, 5mmol), N-methylpiperazine (750mg, 7.5mmol), PyBOP (2.9g, 5.5mmol) and triethylamine (1.5g, 15mmol) were added to 40mL of acetonitrile, reacted overnight at 80 ℃, concentrated under reduced pressure, 100mL of dichloromethane was added, followed by washing with saturated brine (50 mL. multidot.3), and after drying over anhydrous sodium sulfate, a liquid was obtained which was purified by column chromatography (ethyl acetate) to obtain a yellow oil (1g, 90%).1H NMR(400MHz,CDCl3)δ5.36(d,J=47.2Hz,2H),3.87-3.73(m,4H),2.57-2.50(m,4H),2.49(s,3H),2.33(s,3H).
4) Synthesis of intermediate A51-5:
a51-4(1g, 4.1mmol), 2M methylamine in methanol (6mL, 12mmol), water (15mL) and isopropanol (15mL) were added to the lock tube. The reaction was allowed to proceed overnight at 80 ℃ and concentrated under reduced pressure to remove most of the alcohol, extracted with dichloromethane (30mL _ 3), the organic phases combined, dried over anhydrous sodium sulfate and spun to give a liquid which was purified by column chromatography (dichloromethane: methanol: ammonia: 400: 10: 1 to 200: 10: 1) to give a yellow oil (800mg, 76%).1H NMR(400MHz,CDCl3)δ3.80-3.74(m,4H),3.73(s,2H),2.48-2.46(m,4H),2.44(s,6H),2.31(s,3H).
5) Synthesis of product a 51:
a51-5(100mg, 0.4mmol), A34-7(202mg, 1.2mmol) and DIPEA (155mg, 1.2mmol) were added to 5mL of isopropanol and reacted at 90 ℃ for 3 days. Concentrated under reduced pressure, diluted with dichloromethane (100mL), washed with saturated brine (50mL), and the organic phase was dried over anhydrous sodium sulfate and then spin-dried to give a liquid which was purified by column chromatography (dichloromethane: methanol: ammonia: 200: 10: 1) to give a yellow oil (73mg, 48%).
Example 33
A heterocyclic compound a52 synthesized by the method comprising:
Figure BDA0001789890150000341
1) synthesis of intermediate A52-2:
a52-1(5g, 41mmol), methylamine in tetrahydrofuran (80mL), acetic acid (2.5g, 41mmol) in 100mL dichloromethane was stirred at ambient temperature for 1h, sodium borohydride acetate (13g, 62mmol) was added and stirring was continued overnight. Aqueous sodium bicarbonate solution was added and the layers were separated. The aqueous phase was extracted 3 times with dichloromethane and the organic phases were combined, dried and spun dry. The residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a yellow oily liquid (2.7g, 48%).
2) Synthesis of intermediate A52-3:
a52-2(100mg, 0.73mmol), A37-1(143mg, 0.8mmol), potassium iodide (12mg, 0.07mmol) and N, N-diisopropylethylamine (950mg, 7.3mmol) were dissolved in 10mL of acetonitrile, stirred at room temperature overnight, extracted with 100mL of dichloromethane and 50mL of aqueous sodium bicarbonate solution, and the dichloromethane phase was dried and dried. The residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a pale yellow oily liquid (140mg, 69%).
3) Synthesis of product a 52:
a52-3(50mg, 0.18mmol) was dissolved in 2mL ethanol, triethylamine (182mg, 1.8mm0l) and N-methylpiperazine (91mg, 0.9mmol) were added, the reaction was allowed to proceed overnight at 80 ℃, and 50mL dichloromethane and 50mL aqueous sodium bicarbonate were added for extraction. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a yellow oily liquid (63mg, 99%).
Example 34
The heterocyclic compound A53-A56 was synthesized according to the synthesis method of example 33, substituting the corresponding substrate for A52-1 in step 1.
Example 35
According to the synthetic method of example 33, substituting the corresponding substrate for A37-1 in step 2, a heterocyclic compound A4 was synthesized.
Example 36
A heterocyclic compound a62 synthesized by the method comprising:
Figure BDA0001789890150000351
1) synthesis of intermediate A62-2:
intermediate a62-1(500mg, 4.8mmol) was dissolved in 10mL toluene, and methyl magnesium chloride (4.8mL, 14.4mmol) was added with stirring at 0 ℃, the reaction solution was heated under reflux for 18 hours, cooled to room temperature, quenched with ammonia, filtered, the filtrate dried, spun-dried, and the residue was purified by column chromatography (dichloromethane: methanol: ammonia 100: 2: 1) to give a yellow oily liquid (390mg, 60%).1H NMR(400MHz,CDCl3)δ8.56(d,J=4.4Hz,1H),7.64(t,J=7.8Hz,1H),7.45(d,J=8.0Hz,1H),7.13(t,J=6.4Hz,1H),1.51(s,6H).
2) Synthesis of intermediate A62-3:
intermediate a62-2(350mg, 2.6mmol) was dissolved in 10mL of toluene, formic acid (237mg, 5.2mmol) was added and stirred under reflux for 6h, aqueous sodium bicarbonate (50mL) was added, dichloromethane (50mL) was extracted, the dichloromethane phase was dried and spun dry to give a liquid which was purified by column chromatography (dichloromethane: methanol 100: 1 to 100: 3) to give a yellow oily liquid (300mg, 70%).
3) Synthesis of intermediate A62-4:
intermediate a62-3(300mg, 1.8mmol) was dissolved in anhydrous tetrahydrofuran, sodium hydride (222mg, 2.7mmol) was added in portions, after stirring at room temperature for 15 minutes, iodomethane (390mg, 5.4mmol) was added, the reaction was continued, 50mL of water was added for quenching, dichloromethane (50mL) was added for extraction three times, the organic phase was dried and spin-dried to obtain a liquid, which was purified by column chromatography (dichloromethane: methanol ═ 100: 1) to obtain a yellow oily liquid (280mg, 87%).1H NMR(400MHz,CDCl3)δ8.59(d,J=4.4Hz,1H),8.55(s,1H),7.69(t,J=7.4Hz,1H),7.26(d,J=7.7Hz,1H),7.21-7.18(m,1H),2.72(s,3H),1.75(s,6H).
4) Synthesis of intermediate A62-5:
intermediate a62-4(280mg, 1.6mmol) was dissolved in 5mL methanol 1mL water, sodium hydroxide (192mg, 4.8mmol) was added, the mixture was stirred under reflux overnight, 50mL dichloromethane 50mL aqueous sodium bicarbonate was added for extraction, the dichloromethane phase was dried and spun dry to give a liquid which was purified by column chromatography (dichloromethane: methanol 100: 1) to give a yellow oily liquid (216mg, 90%).1H NMR(400MHz,CDCl3)δ8.59(d,J=4.4Hz,1H),7.64(t,J=7.4Hz,1H),7.39(d,J=8.0Hz,1H),7.13(dd,J=7.2,4.8Hz,1H),2.13(s,3H),1.47(s,6H).
5) Synthesis of intermediate A62-6:
intermediate A62-5(50mg, 0.33mmol), intermediate A37-1(65mg, 0.36mmol), potassium iodide (6mg, 0.03mmol), N, N-diisopropylethylamine (430mg, 3mmol) in acetonitrile (1.5mL) was reacted at room temperature overnight. The reaction solution was spin dried, 20mL of aqueous sodium bicarbonate solution was added, extracted with dichloromethane (3 × 10mL), and the organic phases were combined, dried, filtered, and spin dried. The resulting liquid was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1) to give a colorless oily liquid (56mg, 59%).
6) Synthesis of product a 62:
intermediate a62-6(56mg, 0.19mmol), triethylamine (195mg, 1.9mmol), N-methylpiperazine (97mg, 0.95mmol) were dissolved in 1mL ethanol, stirred at 80 ℃ overnight, extracted with 20mL dichloromethane 20mL aqueous sodium bicarbonate solution, the dichloromethane phase was dried and spun to dryness to give a liquid, which was purified by column chromatography (dichloromethane: methanol: ammonia 100: 1) to give a pale yellow oily liquid (60mg, 89%).
Example 37
A heterocyclic compound a75 synthesized by the method comprising:
Figure BDA0001789890150000361
1) synthesis of intermediates A75-1a and A75-1 b:
raw material A52-1(605mg, 5mmol) was dissolved in dichloromethane (30mL), and sodium borohydride acetate (2.12g, 10mmol) and (S) -1- (4-methoxyphenyl) ethylamine (755mg, 5mmol) were added in this order at 0 ℃ and stirred at room temperature overnight. Saturated aqueous sodium bicarbonate (20mL) was added to quench the reaction, the organic phase was separated, dried, and purified by silica gel column chromatography (petroleum ether: acetone: aqueous ammonia 200: 5: 2) to give A75-1a (1.0g, 78%) and A75-1b (70mg, 5.5%) as yellow oily liquids.
A75-1a:1H NMR(400MHz,CDCl3)δ8.60(d,J=3.2Hz,1H),7.61(t,J=7.6Hz,1H),7.20-7.12(m,3H),7.06(d,J=7.6Hz,1H),6.86(d,J=8.0Hz,2H),3.81(s,3H),3.61-3.55(m,1H),3.46-3.35(m,1H),1.31-1.25(m,6H).
A75-1b:1H NMR(400MHz,CDCl3)68.54(s,1H),7.61-7.50(m,1H),7.26-7.11(m,4H),6.81(d,J=7.2Hz,2H),3.90-3.80(m,1H),3.80-3.70(m,4H),1.42-1.30(m,6H).
2) Synthesis of intermediate A75-2:
a75-1a (500mg, 2mmol), 37% aqueous formaldehyde (150mg, 5mmol), sodium borohydride acetate (636mg, 3mmol) dissolved in 30mL of dichloromethane was stirred overnight at room temperature, and 100mL of dichloromethane and 50mL of aqueous sodium bicarbonate were added and extracted. The dichloromethane phase was dried and spun dry and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a colorless oily liquid (400mg, 70%).1H NMR(400MHz,CDCl3)δ8.56(d,J=3.6Hz,1H),7.66(t,J=7.2Hz,1H),7.49(d,J=7.6Hz,1H),7.33-7.27(m,2H),7.15(t,J=6.0Hz,1H),6.88(d,J=8.0Hz,2H),3.99-3.93(m,1H),3.87-3.72(m,4H),2.02(s,3H),1.39-1.25(m,6H).
3) Synthesis of intermediate A75-3:
a75-2(400mg, 1.48mmol) was dissolved in dichloromethane (10mL) and trifluoroacetic acid (5mL) was slowly added dropwise thereto. Stirring at room temperature overnight. Concentrated under reduced pressure, and the residue was washed three times with 1N HCl (15mL) and ethyl acetate (10mL × 3). Adjusting the pH of the aqueous phase to 1N NaOH solution9, dichloromethane extraction (10mL 4). The combined organic phases were dried and concentrated under reduced pressure to give a colorless oily liquid (90mg, 45%).1H NMR(400MHz,CDCl3)δ8.56(d,J=4.0Hz,1H),7.65(t,J=7.2Hz,1H),7.28(d,J=8.0Hz,1H),7.16(t,J=6.0Hz,1H),3.81-3.70(m,1H),2.31(s,3H),1.38(d,J=6.4Hz,3H).
4) Synthesis of intermediate A75-4:
a75-3(50mg, 0.37mmol) and A37-1(71mg, 0.40mmol) were dissolved in acetonitrile (5mL), and potassium iodide (6mg, 0.037mmol) and DIPEA (119mg, 0.93mmol) were added in this order, followed by stirring at room temperature overnight. Concentrated under reduced pressure and purified by silica gel column chromatography (dichloromethane: methanol 150: 1) to give a pale yellow oily liquid (90mg, 88%).
5) Synthesis of intermediate A75-5:
a75-4(90mg, 0.33mmol) and N-methylpiperazine (163mg, 1.6mmol) were dissolved in ethanol (10mL), triethylamine (333mg, 3.3mmol) was added, and the reaction was refluxed at 85 ℃ for 3 h. Cooled to room temperature, concentrated under reduced pressure, and purified by silica gel column chromatography (dichloromethane: methanol: ammonia 100: 2: 1) to give a colorless oily liquid (70mg, 63%).
Example 38
According to the synthetic method of example 37, a heterocyclic compound a76 was synthesized by substituting a75-1b for a75-1a in step 2.
Example 39
A heterocyclic compound a77 synthesized by the method comprising:
Figure BDA0001789890150000381
1) synthesis of intermediate A77-2:
a77-1(5.0g, 27mmol), acetamidine hydrochloride (3.9g, 41mmol) and potassium carbonate (11.3g, 82mmol) were added to ethanol (80mL) and stirred at room temperature overnight. Ethanol was removed by concentration under reduced pressure, 3N HCl was added to adjust PH to 5, and extraction was performed with N-butanol (50mL × 6) to give a pale yellow solid (3.5g, 87%).
2) Synthesis of intermediate A77-3:
a77-2(2.5g, 16.8mmol), N-Boc piperazine (4.7g, 25.2mmol), PyBOP (9.6g, 18.5mmol) and triethylamine (5.1g, 50.4mmol) were added to 60mL of acetonitrile and reacted at 80 ℃ overnight. Concentrated under reduced pressure and the residue purified by column chromatography (5: 1-3: 1 petroleum ether: ethyl acetate) to give a yellow solid (4.3g, 81%).
3) Synthesis of intermediate A77-4:
a77-3(4.0g, 12.6mmol) was dissolved in CH2Cl2(100mL), trichloroisocyanuric acid (2.9g, 12.6mmol) is added in portions in an ice-water bath, the mixture is stirred and reacted for 1 hour, then the temperature is raised to the normal temperature, and the reaction is carried out for 6 hours. The reaction was quenched by addition of saturated aqueous sodium thiosulfate and the solid was removed by filtration. The filtrates were separated and the aqueous phase was extracted with dichloromethane (50mL x 3). The organic phases are combined, washed with saturated salt water and anhydrous Na2SO4Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (petroleum ether: ethyl acetate: 10: 1-5: 1) to give a pale yellow solid (2.4g, 54%).
4) Synthesis of intermediate A77-5:
intermediate A1-3(421mg, 2.6mmol), intermediate A77-4(1.0g, 2.8mmol), sodium iodide (45mg, 0.3mmol) and N, N-diisopropylethylamine (671mg, 5.2mmol) were dissolved in 10mL of acetonitrile and stirred at room temperature overnight. Concentrated under reduced pressure and the residue purified by column chromatography (petroleum ether: ethyl acetate 1: 1-0: 1) to give a pale yellow liquid (800mg, 64%).
5) Synthesis of intermediate A77-6:
intermediate A77-4(600mg, 1.3mmol) was dissolved in dichloromethane (2mL) and trifluoroacetic acid (1mL) was added and reacted at room temperature for 2 h. Concentration under reduced pressure gave a reddish brown oil (700mg, crude) which was used directly in the next reaction.
6) Synthesis of product a 77:
intermediate A77-6(700mg, crude) was dissolved in methanol (1mL) and 37% aqueous formaldehyde (1mL) and sodium cyanoborohydride (125mg, 2.0mmol) were added in that order. Reacting for 2h at normal temperature, and adding saturated NaHCO3Aqueous (1mL) and extracted with dichloromethane (10mL x 3). The organic phases are combined, washed with saturated salt water and anhydrous Na2SO4Drying, and concentrating under reduced pressure. Purifying the residue by column chromatography (basic alumina)Conversion (5: 1-1: 0) of the residue to give a pale yellow solid (200mg, 39%).
Example 40
Heterocyclic compounds a78, a79 were synthesized according to the synthesis of example 37, substituting the corresponding amine for N-methylpiperazine in the last step.
EXAMPLE 41
According to the synthesis method of example 37, acetaldehyde was used instead of formaldehyde in step 2 to synthesize a heterocyclic compound a 80.
Example 42
A heterocyclic compound a81 synthesized by the method comprising:
Figure BDA0001789890150000391
1) synthesis of intermediate A81-1:
a9-1(531mg, 3mmol), 2-hydroxyacetamidine hydrochloride (400mg, 3.6mmol) and potassium carbonate (1.2g, 9mmol) were added to methanol (20mL) and stirred at 70 ℃ for 16 h. The pH was adjusted to about 5-6 with 36% acetic acid, then adjusted to 8 with saturated aqueous sodium bicarbonate solution, concentrated under reduced pressure, and purified by column chromatography (dichloromethane: methanol 50: 1 to 15: 1) to give yellow solid A81-1(268mg, 31%).1H NMR(400MHz,CDCl3)δ6.56(s,1H),5.09(s,1H),4.70(s,2H),3.38(s,6H).
2) Synthesis of intermediate A81-2:
intermediate a81-1(268mg, 1.34mmol), N-methylpiperazine (268mg, 2.68mmol), triethylamine (541mg, 5.36mmol) and benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (766mg, 1.47mmol) were added to acetonitrile (20mL) and stirred at 80 ℃ overnight. Cooled to room temperature, concentrated under reduced pressure, and purified by column chromatography (dichloromethane: methanol 50: 1 to 20: 1) to give intermediate a81-2(355mg, 93%) as a yellow oil. LC-MS (m/z): 282.9[ M + H]+.
3) Synthesis of intermediate A81-3:
intermediate A81-2(355mg, 1.26mmol) was added to 20% sulfuric acid (10mL) and stirred at 80 ℃ overnight. After cooling to room temperature, saturated aqueous sodium bicarbonate was added to adjust pH 8, and the mixture was extracted with dichloromethane (40mL × 5), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give intermediate a81-3(200mg, 67%) as a yellow oil. LC-MS (m/z): 258.9[ M + Na ]]+.
4) Synthesis of product a 81:
intermediate A81-3(140mg, 0.6mmol), intermediate A75-3(106mg, 0.78mmol) and acetic acid (36mg, 0.6mmol) were added to dichloromethane (15mL) and stirred at room temperature for 1 hour. Sodium triacetoxyborohydride (382mg, 1.8mmol) was added under ice-water bath, and reacted at room temperature overnight. Saturated aqueous sodium bicarbonate (5mL) was added, concentrated under reduced pressure, and purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 50: 1: 0.5) to give product a81(78mg, 36%) as a yellow oil.
Example 43
According to the synthesis method of example 42, (R) -octahydropyrrolo [1, 2-a ] pyrazine was used instead of N-methylpiperazine in step 2 to synthesize a heterocyclic compound a 82.
Example 44
According to the synthesis method of example 11, a heterocyclic compound a83 was synthesized by substituting ammonia water for dimethylamine in step 4 and a75-3 for a1-3 in the last step.
Example 45
A heterocyclic compound a84 synthesized by the method comprising:
Figure BDA0001789890150000401
1) synthesis of intermediate A84-2:
intermediate a75-4(260mg, 0.95mmol) was dissolved in absolute ethanol, followed by addition of a84-1(616mg, 2.85mmol) and triethylamine (288mg, 2.85mol) in that order, stirring overnight at 80 ℃, concentration under reduced pressure, and purification by silica gel column chromatography (dichloromethane: methanol: aqueous ammonia 100: 1) gave a yellow oil a84-2(380mg, 88%).1H NMR(400MHz,CDCl3)δ8.58(s,1H),7.70(s,1H),7.43(d,J=7.2Hz,1H),7.26-6.68(m,2H),4.52(s,1H),4.20-3.50(m,8H),3.40-3.00(m,4H),2.80-2.40(m,6H),1.60-1.43(m,12H).
2) Synthesis of intermediate A84-3:
a84-2(200mg, 0.44mmol) was dissolved in dichloromethane, 4mL trifluoroacetic acid was added dropwise at room temperature, and the mixture was stirred overnight, followed by spin-drying of the solvent to obtain a yellow gelatinous solid intermediate A84-3 (crude 500 mg).
3) Synthesis of product a 84:
intermediate A84-3 (crude 500mg) was dissolved in methanol, 37% aqueous formaldehyde (356mg, 4.4mmol) was added, stirring was carried out at room temperature for 20min, sodium cyanoborohydride (82mg, 1.32mmol) was added, and stirring was carried out at room temperature overnight. Saturated sodium bicarbonate was added to adjust the pH to about 7 to 8, the organic phases were extracted three times with dichloromethane (20mL x 3), combined, concentrated under reduced pressure and purified by silica gel column chromatography (dichloromethane: methanol: aqueous ammonia 50: 1: 0.5) to give product a84(80mg, 49%) as a yellow oil.
Example 46
A heterocyclic compound a85 synthesized by the method comprising:
Figure BDA0001789890150000411
1) synthesis of intermediate A85-1:
n, N-dimethylformamide (9.8g, 134.8mmol) was added dropwise to POCl at 0 deg.C3(60.6g, 396.5mmol) and reacted at this temperature for 1 h. 2-methyl-3, 6-dihydroxypyrimidine (10g, 79.3mmol) was added to the reaction mixture in portions, and the reaction was continued at room temperature for 1 hour and then warmed to 105 ℃ for overnight reaction. The reaction mixture was spin-dried, 100mL of ethyl glacial acetate was added, and the mixture was added dropwise to ice water. The suspension was filtered and the filtrate extracted with ethyl acetate (200 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was rotary dried to give yellow intermediate A85-1 (crude 10 g).
2) Synthesis of intermediate A85-2:
intermediate A85-1(10g, 52.3mmol) was dissolved in THF/H2O (50mL/10mL) in a mixed solution, and NaBH is added under ice bath4(4g, 104.7mmol) were added portionwise at this temperatureThe reaction was continued for 30 minutes. Quench with water (50mL), extract with ethyl acetate (50mL), dry the organic phase over anhydrous sodium sulfate, filter, and spin dry the filtrate. Purification by column chromatography (petroleum ether: ethyl acetate: 10: 1) gave intermediate a85-2(2g, 20%) as a yellow solid.1H NMR(400MHz,CDCl3)δ4.91(s,2H),2.69(s,3H).
3) Synthesis of intermediate A85-3:
intermediate A85-2(2g, 10.3mmol) and imidazole (770mg, 11.3mmol) were dissolved in dichloromethane (20mL) and TBSCl (1.7g, 11.3mmol) was added in portions and reacted at room temperature overnight. Water (20mL) and methylene chloride (20mL) were added and the mixture was extracted. The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. Purification by column chromatography (100: 1) gave intermediate a85-3(2.6g, 84%) as a colourless oil.1H NMR(400MHz,CDCl3)δ4.85(s,2H),2.68(s,3H),0.91(s,9H),0.14(s,6H).
4) Synthesis of intermediate A85-4:
intermediate A85-3(1.5g, 4.8mmol), vinylboron trifluoride potassium salt (655mg, 4.88mmol), cesium carbonate (2.4g, 7.32mmol) and tetratriphenylphosphine palladium (566mg, 0.49mmol) were added to 1, 4-dioxane/water (20mL/4mL) and reacted overnight at 100 ℃ under nitrogen. The reaction mixture was spin-dried and purified by column chromatography (100: 1 petroleum ether: ethyl acetate) to give a crude product (900 mg). The crude intermediate was dissolved in a mixed solution of methanol/dichloromethane (30mL/7mL) and ozonized at-65 ℃ for 4 h. After monitoring the reaction, dimethyl sulfide was quenched. The reaction mixture was spin-dried, and extracted with water (20mL) and methylene chloride (50 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was rotary dried to give intermediate A85-4 as a white solid (crude 610 mg).
5) Synthesis of intermediate A85-5:
intermediate A85-4(610mg, 2.0mmol), A75-3(544mg, 4.0mmol) and glacial acetic acid (360mg, 6.0mmol) were added to dichloromethane (10mL) and, after stirring at ambient temperature for 1h, NaBH (OAc)3(1.3g, 6.0mmol) was added in portions and reacted at room temperature overnight. Water was added for quenching (10mL) and dichloromethane (10mL) was extracted. The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. By column chromatography (dichloromethane: A)Alcohol 100: 1/100: 2) to yield intermediate a85-5(480mg, 80%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.66-8.55(m,1H),7.82-7.65(m,1H),7.38-7.30(m,1H),7.25-7.18(m,1H),4.77(s,2H),4.15-3.85(m,3H),2.66(s,3H),2.19(s,3H),1.56(d,J=7.2Hz,3H).
6) Synthesis of product a 85:
intermediate A85-5(50mg, 0.16mmol), N-methylpiperazine (48mg, 0.48mmol) and triethylamine (80mg, 0.8mmol) were dissolved in ethanol (5mL) and reacted overnight at 80 ℃. The reaction mixture was spin-dried and purified by column chromatography (dichloromethane: methanol: ammonia 100: 1 to 100: 2: 1) to give the final product A85(26mg, 80%) as a colorless oil.
Example 47
The heterocyclic compounds a86 and a87 were synthesized according to the synthesis method of example 46, substituting the corresponding amines (R) -octahydropyrrolo [1, 2-a ] pyrazine and (S) -octahydropyrrolo [1, 2-a ] pyrazine for N-methylpiperazine in the last step.
Example 48
A heterocyclic compound a88 synthesized by the method comprising:
Figure BDA0001789890150000431
1) synthesis of intermediate A88-2:
a88-1(500mg, 2.5mmol) and 37% aqueous formaldehyde (1.0g, 12.5mmol) were added to dichloromethane (10mL) and reacted for 1h at room temperature. Then NaBH (OAc)3(1g, 5.0mmol) was added in portions and stirred at room temperature overnight. Quenched by addition of saturated sodium chloride solution (50mL) and extracted with dichloromethane (50 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. The residue was dissolved in dichloromethane (5mL), and trifluoroacetic acid (2mL) was added dropwise at room temperature and stirred at room temperature overnight. And (3) spin-drying the reaction solution, adding a saturated sodium bicarbonate solution to adjust the pH to 8, and spin-drying. Column chromatography (dichloromethane: methanol: ammonia 10: 1) gave intermediate a88-2 as a yellow oil (crude 120 mg).1HNMR(400MHz,CDCl3)δ4.07(s,1H),2.93-2.89(m,1H),2.88-2.82(m,1H),2.78-2.72(m,1H),2.52-2.42(m,1H),2.27(s,3H),2.22-2.10(m,1H),2.05-1.92(m,1H),1.03(d,J=6.0Hz,3H).
2) Synthesis of product a 88:
intermediate A85-5(40mg, 0.13mmol), A88-2(44mg, 0.39mmol) and triethylamine (66mg, 0.65mmol) were dissolved in ethanol (5mL) and allowed to warm to 80 ℃ for reaction overnight. The reaction mixture was spin-dried and purified by column chromatography (dichloromethane: methanol: ammonia 100: 1) to give the final product A88(40mg, 80%) as a colorless oil.
Example 49
According to the synthesis method of example 48, the heterocyclic compound a89 was synthesized by substituting the enantiomer of a88-1 for a88-1 in the first step.
Example 50
A heterocyclic compound a90 synthesized by the method comprising:
Figure BDA0001789890150000441
1) synthesis of intermediate A90-2:
a90-1(1.0g, 5.75mmol), benzyl bromide (980mg, 5.75mmol), and potassium carbonate (1.1g, 7.80mmol) were dissolved in N-N dimethylformamide (50mL) and stirred at 110 ℃ for 0.5 h. After completion of the reaction, the residue was filtered off, the filtrate was dried by rotary evaporation, 2mL of aqueous sodium hydroxide solution was added, extraction was performed with 50mL of ethyl acetate, the organic phase was dried over anhydrous sodium sulfate, and the solvent was dried by rotary evaporation to obtain intermediate A90-2(1.4g, 93%) as a yellow oily substance.1H NMR(400MHz,CDCl3)δ8.09-7.88(m,1H),7.47-7.45(m,2H),7.42-7.39(m,2H),7.36-7.34(m,1H),7.21-7.12(s,2H),5.19(s,2H).
2) Synthesis of intermediate A90-3:
cuprous iodide (101mg, 0.53mmol), dichlorobistriphenylphosphine palladium (300mg, 0.43mmol) was placed in a two-necked flask. Nitrogen was replaced, and A90-2(1.4g, 5.34mmol), propargyl alcohol (890mg, 16mmol) and triethylamine (2.2g, 21mmol) in acetonitrile were injected into a two-necked flask with a syringe and stirred at 50 ℃ until the mixture was saturatedAnd (4) at night. After completion of the reaction, the solvent was dried by evaporation, water (200mL) was added, extraction was performed with ethyl acetate (100mL), the organic phases were combined, the organic phase was dried over anhydrous sodium sulfate, filtration was performed, the filtrate was dried by evaporation, and the residue was purified by column chromatography (pure ethyl acetate) to obtain intermediate a90-3(1.1g, 85%) as a yellow oily substance.1H NMR(400MHz,CDCl3)δ8.17(s,1H),7.44-7.42(m,2H),7.40-7.36(m,2H),7.34-7.30(m,1H),7.22-7.13(m,2H),5.18(s,2H),4.56(d,J=2.8Hz,2H),3.01(s,1H).
3) Synthesis of intermediate A90-4:
a90-3(800mg, 3.35mmol) was dissolved in 10mL of ethanol, and palladium on carbon (0.2g) was added to replace it with hydrogen. After completion of the reaction, the filtrate was filtered, the residue was purified by column chromatography (pure ethyl acetate) to give intermediate A90-4(150mg, 29%) as a yellow oil.1H NMR(400MHz,DMSO-d6)δ9.68(s,1H),7.91(d,J=4.4Hz,1H),7.09(d,J=7.6Hz,1H),7.05-6.97(m,1H),4.47(s,1H),3.43(t,J=6.4Hz,2H),2.70(t,J=7.6Hz,2H),1.81-1.72(m,2H).
4) Synthesis of intermediate A90-5:
tributylphosphine (315mg, 1.56mmol) was dissolved in tetrahydrofuran (10mL), stirred at 0 ℃ under nitrogen, DIAD (315mg, 1.56mmol) was added dropwise, and stirred for 5 min. A90-4(200mg, 1.30mmol) in tetrahydrofuran was slowly added dropwise and reacted for 48 hours. The solvent was dried off and the residue was subjected to column chromatography (pure ethyl acetate) to give intermediate A90-5(48mg, 27%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.10(s,1H),7.11-7.03(m,2H),4.26-4.07(m,2H),3.02-2.85(m,2H),2.17-2.00(m,2H).
5) Synthesis of intermediate A90-6:
a90-5(180mg, 1.33mmol) was dissolved in dichloromethane (20mL) and stirred at room temperature, trichloroisocyanuric acid (460mg, 1.99mmol) was added and stirred at room temperature overnight. After the reaction was completed, the residue was filtered off, 20mL of a saturated aqueous solution of sodium bicarbonate was added, extraction was performed with 30mL of a dichloromethane solution, the organic phase was dried over anhydrous sodium sulfate, and the solution was spin-dried. The residue is subjected to column chromatography (petroleum ether: ethyl acetate: 4: 1)Intermediate A90-6(40mg, 18%) was obtained as a yellow oil.1H NMR(400MHz,CDCl3)δ8.23(s,1H),7.17(s,2H),5.25(m,1H),4.55-4.43(m,1H),4.41-4.36(m,1H),2.63-2.54(m,1H),2.46-2.37(m,1H).
6) Synthesis of intermediate A90-7
Ethylchloroacetoacetate (16.5g, 100mmol) and acetamidine hydrochloride (10g, 106mmol) were dissolved in ethanol (150mL) sequentially. 1, 8-diazabicycloundecen-7-ene (30.4g, 200mmol) was added slowly in an ice-water bath and stirred overnight at ambient temperature. The solvent was removed by concentration under reduced pressure, diluted with dichloromethane (120mL), washed with saturated brine (30mL × 3), the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to remove the solvent to give a residue, intermediate a90-7(7.93g, 50%).1H NMR(400MHz,CDCl3) Delta 13.01(s, 1H), 6.53(s, 1H), 4.37(s, 2H), 2.50(s, 3H).7) Synthesis of intermediate A90-8
Intermediate A90-7(1.7g, 11mmol), methylamine alcohol solution (687mg, 22mmol), diisopropylethylamine (7.1g, 55mmol), potassium iodide (183mg, 0.1mmol) were dissolved in 50mL acetonitrile and heated in a sealed tube at 60 deg.C and stirred overnight. The acetonitrile was spin dried and the residue was purified by column chromatography (dichloromethane: methane: 10: 1) to give intermediate a90-8(600mg, 35%) as a yellow solid.1H NMR(400MHz,DMSO-d6)66.36(s,1H),3.80(s,2H),2.66(s,1H),2.55(s,3H),2.43(s,2H).
8) Synthesis of intermediate A90-9
Intermediate A90-8(300mg, 1.96mmol), triethylamine (1.9g, 19.6mmol), N-methylpiperazine (980mg, 9.8mmol) were dissolved in 20mL acetonitrile, benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (1.1g, 2.15mmol) was added, and the mixture was stirred under reflux overnight. Acetonitrile was spin-dried to give an oily liquid and the residue was purified by column chromatography (dichloromethane: methanol 20: 1) to give intermediate a90-9(300mg, 65%) as a yellow oil.1H NMR(400MHz,CDCl3)66.38(s,1H),3.67-3.63(m,6H),2.47-2.45(m,10H),2.32(s,3H).
9) Synthesis of product a 90:
a90-6(40mg, 0.24mmol), A90-9(83mg, 0.36mmol), and potassium carbonate (79mg, 0.47mmol) were dissolved in acetonitrile (10mL) and reacted at 85 ℃ for 22 h. The solvent was directly spun dry and the residue was purified by column chromatography (dichloromethanol: methanol 40: 1) to give product a90 as a yellow oil (50mg, 57%).
Example 51
A heterocyclic compound a91 synthesized by the method comprising:
Figure BDA0001789890150000461
1) synthesis of intermediate A91-2:
a91-1(5g, 29mmol), 3-buten-1-ol (2.1g, 29mmol) and triphenylphosphine (9.1g, 35mmol) were dissolved in tetrahydrofuran (50mL) under nitrogen exchange and stirred at 0 ℃. DIAD (6.5g, 32mmol) was injected via syringe and stirred at 60 ℃ overnight. After completion of the reaction, the reaction mixture was cooled to room temperature, tetrahydrofuran was dried, 50mL of a saturated aqueous sodium chloride solution was added, and extraction was performed with 50mL of ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate, ethyl acetate was dried, and the residue was purified by column chromatography (petroleum ether: ethyl acetate: 10: 1) to give intermediate a91-2(4.8g, 69%) as a crude yellow oil.
2) Synthesis of intermediate A91-3:
a91-2(4.8g, 22.64mmol), triphenylphosphine (1.8g, 6.79mmol), palladium acetate (760mg, 2.26mmol), triethylamine (4.6g, 45mmol), potassium acetate (17.8g, 181mmol) were dissolved in DMF (50mL) and stirred at 110 ℃ overnight. After completion of the reaction, 50mL of a saturated aqueous solution of sodium chloride was added, extraction was performed with 50mL of ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, ethyl acetate was dried by spinning, and the residue was purified by column chromatography (petroleum ether: ethyl acetate: 10: 1) to obtain intermediate a91-3(1.5g, 48%) as a yellow oily substance.1H NMR(400MHz,CDCl3)δ8.20(s,1H),7.16-7.11(m,2H),5.07(s,1H),4.84(s,1H),4.25(t,J=5.2Hz,2H),2.82(t,J=6.0Hz,2H).
3) Synthesis of intermediate A91-4:
a91-3(1.5g, 10.2mmol) was dissolved in a mixed solution of methanol and chloroform (methanol)Chloroform to 5: 1), adding 10mg sodium bicarbonate solid, stirring at-78 deg.C, and introducing ozone with ozone generator. After completion of the reaction, 20mL of a saturated aqueous solution of sodium chloride was added, extraction was performed with 20mL of dichloromethane, and the organic phases were combined, dried over anhydrous sodium sulfate, and dichloromethane was spin-dried. The residue was purified by column chromatography (pure ethyl acetate) to give intermediate A91-4(1g, 67%) as a yellow solid.1H NMR(400MHz,CDCl3)δ8.46(s,1H),7.40(s,2H),4.62(t,J=6.4Hz,2H),2.98(t,J=6.4Hz,2H).
4) Synthesis of intermediate A91-5:
a91-4(1g, 6.71mmol) was dissolved in dichloromethane (50mL), sodium borohydride acetate (2.62g, 12.40mmol) was added, stirring was carried out at 0 ℃ for 10min, and (S) - (-) -1- (4-methoxyphenylamine) ethylamine (950mg, 6.17mmol) in dichloromethane was added, and stirring was carried out at room temperature overnight. After completion of the reaction, 40mL of a saturated aqueous solution of sodium hydrogencarbonate was added, extraction was performed with 50mL of dichloromethane, the organic phase was dried over anhydrous sodium sulfate, the organic phase was dried, and column chromatography was performed on the residue (DCM: MeOH: 100: 1) to obtain intermediate A91-5(800mg, 45%) as a colorless oil.1H NMR(400MHz,CDCl3)δ8.16(s,1H),7.40(d,J=7.6Hz,2H),7.09(s,2H),6.87(d,J=7.6Hz,2H),4.32-4.11(m,2H),4.07-3.96(m,1H),3.93(t,J=5.2Hz,1H),3.81(s,3H),1.82-1.74(m,2H),1.49(d,J=5.6Hz,3H).
5) Synthesis of intermediate A91-6:
a91-5(400mg, 1.41mmol) was dissolved in 50mL of dichloromethane, stirred at 0 deg.C, aqueous formaldehyde (285mg, 3.50mmol) was added, and after stirring for 10min, sodium borohydride acetate (746mg, 3.50mmol) was added. After completion of the reaction, 30mL of saturated aqueous sodium bicarbonate was added, extraction was performed with 50mL of dichloromethane, and the organic phase was dried over anhydrous sodium sulfate and dichloromethane was spun dry to give intermediate A91-6(330mg, 79%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.22(s,1H),7.40(d,J=8.4Hz,2H),7.06(s,2H),6.86(d,J=8.4Hz,2H),4.41-4.33(m,2H),4.13-4.05(m,2H),3.80(s,3H),2.31-2.22(m,1H),2.04(s,3H),1.93-1.86(m,1H),1.42(d,J=6.4Hz,3H).
6) Synthesis of intermediate A91-7:
a91-6(330mg, 1.11mmol) was dissolved in dichloromethane (10mL), stirred at room temperature, trifluoroacetic acid (6mL) was added slowly and the reaction was allowed to proceed overnight. After completion of the reaction, dichloromethane and trifluoroacetic acid were dried by spinning, hydrochloric acid (0.5mol/mL, 5mL) was added, extraction was performed with 10mL of dichloromethane, a saturated aqueous solution of sodium bicarbonate was added to the aqueous phase, the pH was adjusted to 8, extraction was performed with 20mL of dichloromethane, drying was performed with anhydrous sodium sulfate, and dichloromethane was spun dry to give intermediate A91-7(120mg, 66%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.15(s,1H),7.11(s,2H),4.50-4.14(m,2H),3.76(s,1H),2.56(s,3H),2.24-2.20(m,2H).
7) Synthesis of intermediate A91-8:
a91-7(120mg, 0.73mmol), A37-1(145mg, 0.80mmol), diisopropylethylamine (188mg, 1.46mmol), and potassium iodide (13mg, 0.07mmol) were dissolved in acetonitrile (10mL) and stirred at room temperature. After completion of the reaction, the acetonitrile was dried by rotary evaporation and the residue was purified by column chromatography (dichloromethanol: methanol 70: 1) to give intermediate a91-8(120mg, 54%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.21(s,1H),7.53(s,1H),7.09(s,2H),4.51-4.34(m,1H),4.28-4.13(m,1H),4.10-3.97(m,1H),3.81(s,2H),2.65(s,3H),2.42(s,3H),2.32-2.21(m,1H),2.21-2.12(m,1H).
8) Synthesis of product a 91:
a91-8(120mg, 0.39mmol), N-methylpiperazine (197mg, 1.97mmol) and triethylamine (394mg, 3.94mmol) were dissolved in absolute ethanol (20mL) and the reaction was stirred at 80 ℃ overnight. After completion of the reaction, the ethanol was spin-dried and the residue was purified by column chromatography (dichloromethanol: methanol 60: 1) to give product a91 as a yellow oil (130mg, 94%).
Example 52
A heterocyclic compound a92 synthesized by the method comprising:
Figure BDA0001789890150000481
1) synthesis of intermediate A92-2:
a92-1(5g, 41mmol) was dissolved in anhydrous methanol (50mL), and sodium methoxide (4.4g, 82mmol) was added, and the reaction was stirred at 0 ℃ overnight. After completion of the reaction, the methanol was dried by evaporation, the residue was extracted with 50mL of ethyl acetate, and the residue was purified by column chromatography (petroleum ether: ethyl acetate: 10: 1) to give intermediate a92-2(4.6g, 78%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.27(d,J=2.8Hz,1H),7.53-7.45(m,1H),7.39-7.31(m,1H),3.97(s,3H).
2) Synthesis of intermediate A92-3:
a92-2(500mg, 3.73mmol) was dissolved in 50mL tetrahydrofuran, stirred at 0 ℃ under nitrogen, and 3N methylmagnesium chloride in diethyl ether (3.73mL, 11.2mmol) was injected via syringe. After completion of the reaction, 0.5N diluted hydrochloric acid (5mL) was added, and the mixture was extracted with 50mL of ethyl acetate. Ethyl acetate was rotary dried to give intermediate A92-3(400mg, 63%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.25(s,1H),7.61-7.30(m,2H),3.91(s,3H),2.66(s,3H).
3) Synthesis of intermediate A92-4:
a92-3(450mg, 2.98mmol) was dissolved in 50mL of dichloromethane, and sodium borohydride acetate (1.26g, 5.96mmol) was added thereto, followed by stirring at 0 ℃ for 10min, followed by addition of (S) - (-) -1- (4-methoxyphenylamine) (400mg, 2.65mmol) dissolved in dichloromethane and stirring at room temperature overnight. After completion of the reaction, 50mL of aqueous sodium bicarbonate solution was added, extraction was performed with 50mL of dichloromethane, dichloromethane was dried by spinning, and the residue was purified by column chromatography (dichloromethanol: methanol 50: 1) to obtain intermediate a92(300mg, 35%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.27-8.16(m,1H),7.45-7.32(m,2H),7.31-7.27(m,1H),7.21-7.17(m,1H),6.88(d,J=8.4Hz,2H),4.27(s,1H),3.81(s,3H),3.79-3.70(m,4H),1.60-1.38(m,6H).
4) Synthesis of intermediate A92-5:
a92-4(130mg, 0.46mmol) was dissolved in 10mL of dichloromethane, an aqueous formaldehyde solution (92mg, 1.14mmol) was added thereto, and the mixture was stirred at 0 ℃ and then added with sodium borohydride acetate (241mg, 1.14mmol), followed by stirring at room temperature overnight. After completion of the reaction, 50mL of saturated aqueous sodium bicarbonate was added, extracted with 40mL of dichloromethane, and the dichloromethane was spun dry to give intermediate A92-5(90mg, 66%) as a crude yellow oil.
5) Synthesis of intermediate A92-6:
a92-5(90mg, 0.30mmol) was dissolved in 20mL of dichloromethane, 2mL of trifluoroacetic acid was added, the mixture was stirred at room temperature overnight, after completion of the reaction, dichloromethane and trifluoroacetic acid were dried by spinning, 0.5N hydrochloric acid (0.5mol/mL, 5mL) was added, the aqueous phase was washed with 10mL of dichloromethane, a saturated aqueous solution of sodium bicarbonate was added to the aqueous phase to adjust the pH to 10, extraction was performed with 20mL of dichloromethane, drying was performed with anhydrous sodium sulfate, filtration was performed, and the filtrate was dried by spinning to obtain intermediate A92-6(25mg, 50%) as a yellow oily substance.1H NMR(400MHz,CDCl3)δ8.17(s,1H),7.15(s,2H),4.23(s,1H),3.84(s,3H),2.58(s,3H),2.30(s,3H).
6) Synthesis of intermediate A92-7:
a92-6(40mg, 0.24mmol), A37-1(50mg, 0.26mmol), DIPEA (63mg, 0.48mmol) and KI (4mg, 0.24mmol) were dissolved in 10mL of acetonitrile and stirred at room temperature overnight. After completion of the reaction, the acetonitrile was dried by rotary evaporation and the residue was purified by column chromatography (dichloromethanol: methanol 100: 1) to give intermediate a92-7(35mg, 47.9%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.18(s,1H),7.45(s,1H),7.18-7.11(m,2H),4.53-4.44(m,1H),3.84(s,3H),3.76(d,J=16.4Hz,1H),3.68(d,J=16.4Hz,1H),2.66(s,3H),2.29(s,3H),1.46(d,J=6.8Hz,3H).
7) Synthesis of product a 92:
a92-7(30mg, 0.10mmol), N-methylpiperazine (49mg, 0.49mmol) and triethylamine (100mg, 0.98mmol) were dissolved in ethanol (8mL) and the reaction was stirred at 80 ℃ overnight. After completion of the reaction, the ethanol was spin-dried and the residue was purified by column chromatography (dichloromethanol: methanol 60: 1) to give product a92(30mg, 71.4%) as a yellow oil.
Example 53
A heterocyclic compound a93 synthesized by the method comprising:
Figure BDA0001789890150000501
1) synthesis of intermediate A93-1:
a92-1(10g, 82mmol), 4-methoxybenzylamine (16.8g, 123.0mmol) and cesium carbonate (40g, 123.0mmol) were added to DMF (50mL) and the mixture was stirred at 70 ℃ overnight. Ethyl acetate (50mL) was added and the saturated sodium bicarbonate solution (20mL x 3) was washed three times with water. The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. Purification by column chromatography (20: 1 petroleum ether: ethyl acetate) gave intermediate a93-1 as a yellow solid (crude 11 g).1H NMR(400MHz,CDCl3)δ7.98(s,1H),7.28-7.18(m,3H),7.00(d,J=8.8Hz,1H),6.90(d,J=8.0Hz,2H),5.05(s,1H),4.36(d,J=5.6Hz,2H),3.81(s,3H).
2) Synthesis of intermediate A93-2:
a93-1(11g, 46.0mmol) was dissolved in anhydrous tetrahydrofuran (200mL) and a 3M solution of methyl magnesium chloride in diethyl ether (77mL, 230.1mmol) was added dropwise at 0 deg.C and stirred at this temperature for 30 min. Quenched by addition of saturated ammonium chloride solution (50mL) and extracted with ethyl acetate (100 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. Purification by column chromatography (5: 1 petroleum ether: ethyl acetate) gave intermediate a93-2(4g, 34%) as a yellow solid.1H NMR(400MHz,CDCl3)δ9.02(s,1H),8.03-7.90(m,1H),7.28-7.15(m,2H),7.08-6.98(m,1H),6.93-6.83(m,2H),4.50-4.30(m,2H),3.79(s,3H),2.72(s,3H).
3) Synthesis of intermediate A93-3:
intermediate A93-2(1.5g, 5.8mmol) was dissolved in trifluoroacetic acid (5mL) and reacted overnight at room temperature. The reaction was spin dried, adjusted to pH 8 with saturated sodium bicarbonate solution and extracted with dichloromethane (20 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. The residue was again dissolved in glacial acetic acid (10mL), acetic anhydride (20mL) was added and stirred at room temperature overnight. The reaction was spin dried, adjusted to pH 8 with saturated sodium bicarbonate solution and extracted with dichloromethane (20 mL). The organic phase was dried over anhydrous sodium sulfate,filtration and spin-drying of the filtrate gave intermediate A93-3(1g, 97%) as a white solid.1H NMR(400MHz,CDCl3)δ11.54(s,1H),9.10(d,J=8.8Hz,1H),8.37(d,J=4.4Hz,1H),7.48(dd,J=8.8Hz,4.4Hz,1H),2.80(s,3H),2.26(s,3H).
4) Synthesis of intermediate A93-4:
intermediate A93-3(1g, 5.6mmol), (S) - (-) -1- (4-methoxyphenyl) ethylamine (1.3g, 8.4mmol) and tetraethyltitanate (3.8g, 16.8mmol) were added to 1, 4-dioxane (30mL) and reacted under nitrogen at 100 ℃ overnight. After cooling to room temperature, methanol (30mL) was added and sodium borohydride (638mg, 16.8mmol) was added in portions at 0 ℃ and reacted at room temperature for 2 h. Filtering with diatomite, and spin-drying the filtrate. Saturated sodium chloride solution (10mL) was added and extracted with dichloromethane (10mL x 3). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was spin-dried. Purification by column chromatography (1: 1 petroleum ether: ethyl acetate) gave intermediate a93-4(380mg, 22%) as a white solid.1H NMR(400MHz,CDCl3)δ11.78(s,1H),8.62(s,1H),8.21(s,1H),7.23-7.16(m,1H),7.15-7.04(m,2H),6.87(d,J=8.0Hz,2H),4.10(s,1H),3.79(s,3H),3.61(s,1H),2.20(s,3H),1.50-1.33(m,6H).
5) Synthesis of intermediate A93-5:
intermediate A93-4(352mg, 1.1mmol) was dissolved in 20mL of methanol, and 37% aqueous formaldehyde (0.9g, 11mmol) and acetic acid (66mg, 1.1mmol) were added, followed by stirring at room temperature for 1 hour, followed by addition of sodium cyanoborohydride (0.69g, 11mmol), and reaction at room temperature overnight. The reaction was concentrated to dryness under reduced pressure, DCM (50mL) was added to dissolve it, saturated sodium bicarbonate was added to adjust the pH to 8, the organic phase was concentrated to dryness under reduced pressure, and the residue was purified by column chromatography (dichloromethane: methanol 50: 1) to give intermediate a93-5(332mg, 92%) as a yellow solid.1H NMR(400MHz,CDCl3)δ11.40(s,1H),8.63(s,1H),8.26(s,1H),7.22-7.17(m,2H),7.01-6.85(m,3H),4.17(s,1H),3.94(s,1H),3.81(s,3H),2.21(s,3H),2.00(s,3H),1.50-1.40(m,6H).
6) Synthesis of intermediate A93-6:
intermediate A93-5(332g, 1mmol) was dissolved in 5mL DCM, and 5mL trifluoroacetic acid was added thereto and stirred at room temperature overnight. The reaction system was concentrated to dryness under reduced pressure to give yellow oil A93-6 (crude 600 mg). LC-MS (m/z): 194.0[ M + H ]]+.
7) Synthesis of intermediate A93-7:
intermediate A93-6 (crude 0.6g, 1mmol), intermediate A37-1(212mg, 1.2mmol) and potassium iodide (17mg, 0.1mmol) were dissolved in 10mL acetonitrile, diisopropylethylamine (0.645g, 5mmol) was added and the reaction was allowed to proceed at room temperature overnight. The reaction was concentrated to dryness under reduced pressure, saturated sodium bicarbonate was added to adjust the pH to 8, and dichloromethane was extracted (20 mL). The organic phase was concentrated under reduced pressure and the residue was purified by column chromatography (dichloromethane: methanol: aqueous ammonia 100: 1: 0.1) to give intermediate a93-7 as a yellow solid (crude 190 mg).
8) Synthesis of product a 93:
intermediate A93-7 (crude 170mg, 0.51mmol), N-methylpiperazine (204mg, 2.04mmol) and diisopropylethylamine (657mg, 5.1mmol) were dissolved in 10mL ethanol and stirred at 80 ℃ for 4 h. The reaction was concentrated to dryness under reduced pressure, and the residue was purified by column chromatography (dichloromethane: methanol: ammonia 20: 1: 0..05) to give a colorless oil a93(85mg, 5% yield in three steps).
Example 54
According to the synthesis method of example 53, a heterocyclic compound a94 was synthesized by using methanesulfonyl triethylamine condition instead of acetic anhydride acetic acid condition in step 3.
Example 55
Comparative compound B1, which was synthesized by the following method:
Figure BDA0001789890150000521
1) synthesis of intermediate B1-2:
b1-1(200mg, 1.2mmol), 4-dimethylaminopyridine (14mg, 0.1mmol) and triethylamine (370mg, 3.6mmol) were dissolved in dichloromethane (15mL), methanesulfonyl chloride (210mg, 1.8mmol) was dissolved in dichloromethane at 0 ℃ and added slowly, stirred at room temperature for 2h, extracted with saturated brine and dichloromethane, separated, the dichloromethane phase dried and dried. The residue was purified by column chromatography (petroleum ether: ethyl acetate 1: 1) to give a colorless oily liquid (220mg, 76%).
2) Synthesis of intermediate B1-3:
b1-2(200mg, 0.83mmol), tetrahydrofuran solution in 5mL of methylamine, N-diisopropylethylamine (540mg, 4.2mmol), stirred under reflux overnight, extracted with 100mL of dichloromethane, 100mL of aqueous sodium bicarbonate, the dichloromethane phase dried and dried, and the residue purified by column chromatography (dichloromethane: methanol 100: 1 to 25: 1) to give a brown oily liquid (100mg, 68%).
3) Synthesis of intermediate B1-4:
b1-3(90mg, 0.5mmol) was dissolved in 3mL ethanol, triethylamine (75mg, 0.8mmol) and 4, 6-dichloropyrimidine (110mg, 0.8mmol) were added, the mixture was stirred at room temperature overnight, 100mL dichloromethane 100mL aqueous sodium bicarbonate solution was added and extracted, the dichloromethane phase was dried and spun dry, and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a brown oily liquid (80mg, 55%).
4) Synthesis of product B1:
b1-4(80mg, 0.28mmol) was dissolved in 3mL ethanol, triethylamine (280mg, 28mmol) and N-methylpiperazine (30mg, 0.31mmol) were added, the mixture was stirred overnight at 80 ℃, 50mL dichloromethane and 50mL aqueous sodium bicarbonate solution were added and extracted, the dichloromethane phase was dried and spun dry, and the residue was purified by column chromatography (dichloromethane: methanol 100: 1 to 50: 1) to give a colorless oily liquid (20mg, 20%).
Example 56
Comparative compound B2, which was synthesized by the following method:
Figure BDA0001789890150000531
a1-4(70mg, 0.24mmol) was dissolved in 5mL methanol, 5% wet palladium on carbon (15mg) was added, hydrogen was replaced, the mixture was stirred overnight at 50 ℃, the filtrate was filtered off, the filtrate was spin-dried, and the residue was purified by column chromatography (dichloromethane: methanol 100: 1) to give a colorless oily liquid (50mg, 82%).
Example 57
Comparative compound B3 was synthesized according to the synthesis procedure of example 1, substituting dimethylamine for N-methylpiperazine in the last step.
Example 58
Comparative compound B4 was synthesized according to the synthesis of example 31 substituting 2-chloro-4-methylpyrimidine for 2-methyl-4-chloropyrimidine in step 1.
Example 59
Comparative compound B6, which was synthesized by the following method:
Figure BDA0001789890150000532
1) synthesis of intermediate B6-1:
a1-3(100mg, 0.62mmol), 6-bromopyridine-2-carbaldehyde (162mg, 0.68mmol), acetic acid (48mg, 0.62mmol) were dissolved in 10mL of dichloroethane, and after stirring at room temperature for 10 minutes, sodium borohydride acetate (252mg, 0.93mmol) was added, stirring was continued overnight, 100mL of dichloromethane and 50mL of an aqueous sodium bicarbonate solution were added for extraction, the dichloromethane phase was dried and dried, and the residue was purified by column chromatography (dichloromethane: methanol: 100: 1) to give a pale yellow oily liquid (180mg, 87%).
2) Synthesis of product B6:
b6-1(50mg, 0.15mmol), dissolved in 2mL NMP, added DIPEA (193mg, 1.5mmol), N-methylpiperazine (73mg, 0.75mmol), stirred at 120 ℃ overnight, extracted with 50mL dichloromethane and 50mL aqueous sodium bicarbonate, the dichloromethane phase dried and spun dry, and the residue purified by column chromatography (dichloromethane: methanol 100: 1) to give a colorless oily liquid (12mg, 23%).
Example 60
Comparative compound B5 was synthesized according to the synthesis of example 59, substituting 2-chloropyridine-4-carbaldehyde for 6-bromopyridine-2-carbaldehyde in step 1.
Example 61
Comparative compounds B7 and B8, synthesized according to the procedure of patent WO 2009121063.
TABLE 1 analytical Structure and spectral data for heterocyclic Compounds A1-A94 and comparative Compounds B1-B8
Figure BDA0001789890150000541
Figure BDA0001789890150000551
Figure BDA0001789890150000561
Figure BDA0001789890150000571
Figure BDA0001789890150000581
Figure BDA0001789890150000591
Figure BDA0001789890150000601
Figure BDA0001789890150000611
Figure BDA0001789890150000621
Figure BDA0001789890150000631
Figure BDA0001789890150000641
Figure BDA0001789890150000651
Figure BDA0001789890150000661
Figure BDA0001789890150000671
Figure BDA0001789890150000681
Example 62
This example demonstrates the binding of heterocyclic compounds a1-a77 and B1-B8 prepared in examples 1-61 to CXCR 4.
HPB-ALL cells were cultured in RPMI-1640 medium containing 10% heat-inactivated fetal bovine serum (Hyclone) +1X penicillin/streptomycin +1X non-essential amino acids +1X sodium pyruvate + 50. mu.M β -mercaptoethanol. HPB-ALL cells were washed once with ice-cold assay buffer (duchenne phosphate buffer + 10% heat-inactivated fetal bovine serum) and then resuspended in assay buffer to a final concentration of 1 × 106And/ml. 95. mu.l/well of the above cell suspension was added to a 96-well plate, and then human CXCR4 monoclonal antibody 12G5(SungeneH31841-11H) conjugated with APC fluorescent label was added to wells 2-12, and a corresponding amount of murine IgG antibody conjugated with APC fluorescent label was added to well 1 as a control. The test compound was diluted stepwise with DMSO and then diluted to the desired concentration with assay buffer. Mu.l of each test compound solution was added to wells 2-11, and the corresponding amount of assay buffer was added to well 1 and well 12, followed by incubation at 4 ℃ for 3h in the absence of light. Mu.l of 4% paraformaldehyde solution was added to each well, and incubated in ice for 10 minutes in the dark. The cells were washed with test buffer and thenResuspended in test buffer, fluorescence signal detected by flow cytometry (Guavasoft6/8HT, Millipore), and IC of compounds calculated from the inhibition of luminescence signal by compounds at different concentrations50The value is obtained. Taking the figure 1 as an example, when the concentration is more than 100nM, the inhibition rate of the heterocyclic compound A42 on the fluorescence signal of 12G5 is nearly 100%, which shows that the compound A42 has nearly completely replaced 12G5 from CXCR4 protein; when the concentration is 0.3nM, the inhibition rate of the heterocyclic compound A42 on the fluorescence signal of 12G5 is nearly 0%, indicating that compound A42 can not nearly replace 12G5 from CXCR4 protein; it can be read from the curve that when the concentration is 9.8nM, the inhibition ratio of the heterocyclic compound A42 to the fluorescence signal of 12G5 is 50%, which indicates that when the concentration is that the heterocyclic compound A42 just replaces half of 12G5 from CXCR4 protein, the concentration is the half Inhibition Concentration (IC) of the heterocyclic compound A4250)。IC50The lower the value, the higher the ability of the heterocyclic compound to bind CXCR4, the better the activity. If IC50> 10000nM, indicating that the compound is unable to displace 12G5 from CXCR4 protein at 10000nM, and the compound can be considered to have no binding ability to CXCR4, i.e. no activity.
The results are shown in table 2 (results of the binding capacity assay for heterocyclic compounds a1-a77 to CXCR 4) and table 3 (results of the binding capacity assay for comparative compounds B1-B8 to CXCR 4).
TABLE 2 results of the binding capacity assay experiment of heterocyclic compounds A1-A77 to CXCR4
Figure BDA0001789890150000691
Figure BDA0001789890150000701
As can be seen from table 2, the heterocyclic compounds of the present invention, which have a good ability to bind to CXCR4, are potent inhibitors of CXCR4 and can be used for the treatment or prevention of conditions responsive to CXCR4 receptor inhibition.
TABLE 3 results of the binding capacity assay experiment of comparative compounds B1-B8 to CXCR4
Numbering IC50(nM) Numbering IC50(nM) Numbering IC50(nM)
B1 >10000 B2 >10000 B3 >10000
B4 >10000 B5 >10000 B6 >10000
B7 >10000 B8 >10000
As can be seen from Table 3, in the heterocyclic compounds of the present invention, the position of the connection of the U group is important when the U group is replaced with A3When linked (comparative compound B4), the inhibitory activity of CXCR4 was lost; furthermore, the two N's on the U group are also important, and the removal of one N (comparative compound B3) or the absence of N (comparative compound B2) both lose the inhibitory activity of CXCR 4; at the same time, the position of the N on the pyrimidine ring is critical for CXCR4 activity, and substitution of the pyrimidine ring for a pyridine ring (comparative compounds B5 and B6) or for other linked pyrimidine rings (comparative compounds B7 and B8) all lost the inhibitory activity of CXCR 4.
Example 63
This example measured the ability of heterocyclic compounds A1, A22, A42, A61, A75 and A78-A94 prepared in examples 1 to 61 to inhibit the induction of calcium ion flux in T cells by SDF-1 α.
Human CD4+ T cells were isolated from human whole blood and subsequently activated and amplified for use using the CD3/CD28 amplification kit (Life Technologies). The pre-cultured cells were suspended in a balanced salt solution containing 20mM HEPES, HEPES/0.005% to a cell concentration of 5X106cells/mL. In 384 well plates, 20. mu.M cells (5X 10) were added per well6cell/mL), after allowing the cells to equilibrate at room temperature for 10min, 20. mu.M Fluo-4 fluorescent calcium indicator dye was added to each well, which was allowed to equilibrate at room temperature for 10min at 37 ℃ with 5% O2/CO2Culturing for 30 min. Calcium efflux was stimulated by adding 25. mu.L (40nM) SDF-1. alpha. per well, followed by addition of test compound (test concentration range 10. mu.M-0.035 nM) to the wells of the test plate using FLIPR Tetra. The test plate is transferred to a fluorescent integrated phase plate reader (FLIPR) and the change in calcium ion by the test compound is detected to determine the ability of the compound to antagonize CXCR 4. Test values fluorescence test values were normalized to untreated control wells, using compounds a78 and a83 as examples (fig. 3, fig. 4), at 50% Inhibitory Concentrations (IC)50Value) is givenDefined as the concentration of test compound required to inhibit SDF-1-induced calcium ion by 50% relative to untreated control wells. The results are shown in Table 4 (results of experiments on inhibition of calcium ion flux of SDF-1. alpha. induced T cells by heterocyclic compound A78-A94).
TABLE 4 results of experiments on inhibition of calcium flux of SDF-1 alpha-induced T cells by heterocyclic compounds A1, A22, A42, A61, A75 and A78-A94
Numbering IC50(nM) Numbering IC50(nM) Numbering IC50(nM)
A1 4.5 A22 3.4 A42 0.021
A61 0.024 A75 0.090 A78 0.062
A79 0.22 A80 0.18 A81 4.5
A82 2.5 A83 0.93 A84 3.0
A85 0.99 A86 0.13 A87 0.18
A88 0.30 A89 0.13 A90 0.046
A91 0.032 A92 118 A93 20000
A94 20000
Example 64
This example measured the inhibitory activity of the heterocyclic compounds A1-A94 prepared in examples 1-61 on CYP liver enzymes.
The two major CYP isozymes and their respective probe substrates are: CYP3a4 (midazolam, 1 μ M) and CYP2D6 (dextromethorphan, 5 μ M). All probes were used at concentrations near or below their KMS. The mixture (200. mu.L) was incubated in a thermostatted water bath at 37 ℃. The mixture contained HLM (0.2mg/mL), phosphate buffer (100mM, pH 7.4), NADPH (1. mu.M), test compound (10. mu.M) and the respective CYP probe substrates. The mixture was preincubated for 10min before the reaction with NADPH started, and inhibitor-enzyme interaction was performed. After 10 minutes, the reaction was quenched by addition to 100 μ L containing the appropriate amount of cold acetonitrile. Then centrifuging and injecting the sample to be tested into LC-MS/MS to quantitatively analyze the concentration of the specific metabolites formed by the substrate and the CYP enzyme. At least three tests are performed independently for each test compound. The results are shown in table 5 (the results of the test for measuring the inhibition ability of some representative compounds on CYP liver enzymes), and the lower the inhibition rate, the weaker the inhibition ability of the compounds on CYP liver enzymes is, and the better the safety of drug interaction is.
TABLE 5
Compound numbering Inhibition rate of CYP3A4 Inhibition rate of CYP2D6
A9 52% 53%
A10 45% 39%
A41 58% 28%
A43 48% 56%
As shown in Table 5, the heterocyclic compound of the present invention has an inhibition rate of less than 60% on CYP liver enzyme at 10 μ M, which indicates that the compound of the present invention has better safety of drug interaction and is greatly superior to clinical compound AMD070 (the inhibition rate of 100% on CYP2D6 at 1 μ M).

Claims (10)

1. A heterocyclic compound with CXCR4 signal channel inhibition activity and pharmaceutically acceptable salts and isomers thereof have a structure shown in a general formula I:
Figure FDA0003136300010000011
wherein A is3Selected from the group consisting of CR10;A1,A2Each independently selected from N or CR10And A is1,A2One is N;
w is
Figure FDA0003136300010000012
U is
Figure FDA0003136300010000013
Q is a bond or CR23R24
R1,R2,R3,R4Each independently selected from hydrogen atom, deuterium atom, C1-6Alkyl or C1-8An alkoxy group;
R5,R6each independently selected from a hydrogen atom, a deuterium atom or C1-6Alkyl which is unsubstituted or substituted by 1 to 3 groups selected from amino or NHCOOC1-6Alkyl substituent substitution; or R4And R5And the atoms to which they are attached are linked to each other to form a ring;
R7is selected from C1-6Alkyl which is unsubstituted or substituted by 1 to 3 substituents selected from C3-6Cycloalkyl substituents;
R8,R9each independently selected from a hydrogen atom, a deuterium atom or C1-6An alkyl group;
R10selected from hydrogen atom, deuterium atom, halogen, cyano, amino, C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-8Alkoxy, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、SC1-6An alkyl, 4-7 membered heterocyclyl, 6 membered aryl or 5-6 membered heteroaryl, said alkyl, alkoxy, heterocyclyl, aryl or heteroaryl being unsubstituted or substituted with 1-3 substituents selected from halogen or hydroxy;
R11selected from hydrogen atoms or C1-6An alkyl group;
R12,R13,R14,R15,R18,R19,R20,R21,R23,R24each independently selected from a hydrogen atom, a deuterium atom or C1-6An alkyl group which is unsubstituted or substituted with 1 to 3 substituents selected from the group consisting of hydroxy; or R11And R14And the atoms to which they are attached are linked to each other to form a ring;
R16,R17are each independently selected from C1-6An alkyl group; or R16And R17And the atoms to which they are attached are joined to form a 5-7 membered heterocyclic ring;
R22selected from hydrogen atoms, C1-6Alkyl radical, C3-6Cycloalkyl or C3-8Heterocycloalkyl, said alkyl, cycloalkyl or heterocycloalkyl being unsubstituted or substituted by 1 to 3 substituents selected from hydroxy, cyano, C3-6Cycloalkyl or C1-3Alkoxy, said heterocycloalkyl containing 1 heteroatom selected from O or N; or R22And R15And the atoms to which they are attached are linked to each other to form a ring.
2. The heterocyclic compounds having CXCR4 signaling pathway inhibitory activity according to claim 1 and the pharmaceutically acceptable salts and isomers thereof, wherein W is selected from the group consisting of
Figure FDA0003136300010000021
Wherein X, Y and Z are respectively and independently selected from CR25R26Or O;
R25,R26each independently selected from a hydrogen atom; or R25And R26And the carbon atoms to which they are attached are linked to each other to form a ring.
3. The heterocyclic compounds having CXCR4 signaling pathway inhibitory activity according to claim 2 and the pharmaceutically acceptable salts and isomers thereof, wherein X, Y, Z are each independently selected from CH2O or
Figure FDA0003136300010000022
And at least one of X, Y and Z is
Figure FDA0003136300010000023
4. Heterocyclic compounds having CXCR4 signaling pathway inhibitory activity according to claim 1 and the pharmaceutically acceptable salts and isomers thereof, characterized in that W is selected from any of the following groups:
Figure FDA0003136300010000024
5. heterocyclic compounds with CXCR4 signaling pathway inhibitory activity according to any of the claims from 1 to 4, as well as the pharmaceutically acceptable salts and isomers thereof, characterized in that U is selected from any of the following groups:
Figure FDA0003136300010000031
6. the following heterocyclic compounds and pharmaceutically acceptable salts and isomers thereof:
Figure FDA0003136300010000032
Figure FDA0003136300010000041
Figure FDA0003136300010000051
Figure FDA0003136300010000061
Figure FDA0003136300010000071
Figure FDA0003136300010000081
Figure FDA0003136300010000091
7. a pharmaceutical composition comprising a therapeutically effective amount of one or more heterocyclic compounds having CXCR4 signaling pathway inhibitory activity according to any one of claims 1-6 and pharmaceutically acceptable salts or isomers thereof, and further comprising at least one pharmaceutically acceptable carrier.
8. A combination composition comprising a combination of a heterocyclic compound having CXCR4 signaling pathway inhibitory activity according to any one of claims 1 to 6 and a pharmaceutically acceptable salt or isomer thereof with one or more of an anti-tumor drug, an antibacterial drug, an antiviral drug, a central nervous system drug, and a diabetes drug.
9. Use of a heterocyclic compound according to any one of claims 1-6 and pharmaceutically acceptable salts or isomers thereof, or of a composition according to claims 7 or 8 for the preparation of a medicament for the treatment of disorders by antagonizing the CXCR4 pathway, stem cell mobilization, wound healing and burn treatment, wherein said disorders are selected from the group consisting of: HIV infection, myocardial infarction, diseases associated with hematopoiesis, inflammation, allergic diseases, interstitial lung diseases, lupus erythematosus, multiple sclerosis, systemic sclerosis, myasthenia gravis, juvenile onset diabetes, transplant rejection, retinitis pigmentosa, proliferative vitreoretinopathy, Berster's vitelliform macular degeneration, wet and dry age-related macular degeneration, diabetic retinopathy, retinopathy of prematurity, diabetic macular erythroma, retinal vein occlusion, cystoid macular edema, glaucoma, vein branch occlusion, breast cancer, lung cancer, bladder cancer, pancreatic cancer, liver cancer, head and neck squamous cell carcinoma, thyroid cancer, sarcoma, osteosarcoma, sclerofibroma, melanoma, prostate cancer, colorectal cancer, ovarian cancer, cervical cancer, esophageal cancer, gastric cancer, myeloma, lymphoma, cervical cancer, esophageal cancer, myeloma, and other cancers, Chronic and non-progressive anemia, idiopathic or primary thrombocythemia, idiopathic myelofibrosis, pulmonary fibrosis, kidney fibrosis, liver fibrosis, cirrhosis, macroglobulinemia, leukemia, myelodysplastic syndrome, myeloproliferative disorders, brain tumors, astrocytomas, medulloblastomas, schwannoma, primary neuroectoblastoma, or pituitary tumors.
10. The use according to claim 9, wherein the condition is selected from: asthma, allergic pneumonia, polymyositis, ankylosing spondylitis, rheumatoid arthritis, glomerulonephritis, autoimmune thyroiditis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, scleroderma, psoriasis, dermatitis, eczema, urticaria, vasculitis, eosinophilic fasciitis or uveitis, mantle cell lymphoma, cutaneous T-cell lymphoma, acute leukemia, chronic leukemia, lymphatic leukemia or myeloid leukemia.
CN201811034891.7A 2017-09-25 2018-09-05 4-aminopyrimidine derivatives as CXCR4 inhibitors and uses thereof Active CN109553604B (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US16/649,983 US11396501B2 (en) 2017-09-25 2018-09-24 Heteroaryl compounds as CXCR4 inhibitors, composition and method using the same
EP18859565.6A EP3687540A4 (en) 2017-09-25 2018-09-24 Heteroaryl compounds as cxcr4 inhibitors, composition and method using the same
JP2020538760A JP7282786B2 (en) 2017-09-25 2018-09-24 Heteroaryl compounds, compositions and methods using same as CXCR4 inhibitors
PCT/US2018/052503 WO2019060860A1 (en) 2017-09-25 2018-09-24 Heteroaryl compounds as cxcr4 inhibitors, composition and method using the same
KR1020207010206A KR20200058443A (en) 2017-09-25 2018-09-24 Heteroaryl compounds as CXCR4 inhibitors, compositions and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710875041 2017-09-25
CN2017108750419 2017-09-25

Publications (2)

Publication Number Publication Date
CN109553604A CN109553604A (en) 2019-04-02
CN109553604B true CN109553604B (en) 2021-08-27

Family

ID=65864655

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811034891.7A Active CN109553604B (en) 2017-09-25 2018-09-05 4-aminopyrimidine derivatives as CXCR4 inhibitors and uses thereof

Country Status (1)

Country Link
CN (1) CN109553604B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115403510B (en) * 2022-08-11 2023-04-25 南方医科大学 PD-L1/CXCL12 double-target inhibitor, preparation method and application

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101052635A (en) * 2004-09-02 2007-10-10 史密丝克莱恩比彻姆公司 Chemical compounds
WO2008008854A2 (en) * 2006-07-11 2008-01-17 Emory University Cxcr4 antagonists including diazine and triazine structures for the treatment of medical disorders
CN101242848A (en) * 2005-06-14 2008-08-13 太景生物科技股份有限公司 Pyrimidine compounds
CN101565404A (en) * 2008-04-21 2009-10-28 太景生物科技股份有限公司 Heterocyclic compounds
CN102015717A (en) * 2008-03-28 2011-04-13 奥蒂瑞斯治疗公司 Chemokine receptor modulators
CN103282360A (en) * 2010-10-14 2013-09-04 普罗克斯马根有限公司 Cxcr4 receptor antagonists
WO2017106291A1 (en) * 2015-12-15 2017-06-22 Bristol-Myers Squibb Company Cxcr4 receptor antagonists

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101052635A (en) * 2004-09-02 2007-10-10 史密丝克莱恩比彻姆公司 Chemical compounds
CN101242848A (en) * 2005-06-14 2008-08-13 太景生物科技股份有限公司 Pyrimidine compounds
WO2008008854A2 (en) * 2006-07-11 2008-01-17 Emory University Cxcr4 antagonists including diazine and triazine structures for the treatment of medical disorders
CN102015717A (en) * 2008-03-28 2011-04-13 奥蒂瑞斯治疗公司 Chemokine receptor modulators
CN101565404A (en) * 2008-04-21 2009-10-28 太景生物科技股份有限公司 Heterocyclic compounds
CN103282360A (en) * 2010-10-14 2013-09-04 普罗克斯马根有限公司 Cxcr4 receptor antagonists
WO2017106291A1 (en) * 2015-12-15 2017-06-22 Bristol-Myers Squibb Company Cxcr4 receptor antagonists

Also Published As

Publication number Publication date
CN109553604A (en) 2019-04-02

Similar Documents

Publication Publication Date Title
DE60028354T2 (en) CHEMOKINE RECEPTOR BINDING HETEROCYCLIC COMPOUNDS
US7872020B2 (en) TGF-β inhibitors
JP6522646B2 (en) Substituted 4,5,6,7-tetrahydro-pyrazolo [1,5-α] pyrazine derivatives as ROS1 inhibitors and 5,6,7,8-tetrahydro-4H-pyrazolo [1,5-α] [1,1 4] Diazepine derivatives
KR20150133172A (en) Mk2 inhibitors and uses thereof
WO2019060860A1 (en) Heteroaryl compounds as cxcr4 inhibitors, composition and method using the same
JP2017508779A5 (en)
US20040132730A1 (en) Inhibitors of TGFbeta
CN110156656B (en) Five-membered heteroaromatic ring derivative, preparation method thereof, pharmaceutical composition and application
EP1628957B1 (en) Sulfopyrroles
JP2023504866A (en) Fluorine-containing heterocyclic derivative having macrocyclic structure and use thereof
CN113773335A (en) Compound as protein kinase inhibitor and preparation method and application thereof
JP2022517723A (en) Macrocycle compound as a CDK inhibitor, its production method and its application in pharmaceutical products
JP2022515309A (en) Substituted aryl compounds, their production methods and uses
CN109553604B (en) 4-aminopyrimidine derivatives as CXCR4 inhibitors and uses thereof
US9078899B2 (en) Pyrazolyl-based carboxamides II
KR20210108555A (en) 1,3,4-Oxadiazol Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
CN110669036B (en) Heterocyclic compounds with CXCR4 signal channel inhibition activity and application thereof
CN111875583B (en) Triazole derivative and preparation method and application thereof
CN109081818B (en) Novel indoleamine 2, 3-bis-oxidase inhibitors
CN110317191B (en) Pyridine heterocyclic compound and application thereof as CXCR4 inhibitor
AU2022288945A1 (en) Heteroaryl compounds as inhibitors of tyk2, composition and application thereof
AU2018337138B2 (en) 2-substituted pyrazole amino-4-substituted amino-5-pyrimidine formamide compound, composition, and application thereof
CN107522634B (en) Biaryl urea carboxylic acid derivative or salt thereof, and preparation method and application thereof
CN107522641B (en) Biaryl urea derivative or salt thereof, and preparation method and application thereof
US11396501B2 (en) Heteroaryl compounds as CXCR4 inhibitors, composition and method using the same

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20200619

Address after: Unit 101 and 102, C11 building, phase I project of biomedical industrial park, 218 Xinghu street, Suzhou Industrial Park, Suzhou area, Suzhou pilot Free Trade Zone, Jiangsu Province

Applicant after: CGENE TECH (SUZHOU, CHINA) Co.,Ltd.

Address before: 215123 Xinghu Street 218 Bio-nano Park, Suzhou Industrial Park, Jiangsu Province, B1 Floor 708

Applicant before: SUZHOU YUNXUAN PHARMACEUTICAL Co.,Ltd.

SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address
CP03 Change of name, title or address

Address after: 215123 units 101 and 102, C11 building, phase I project of biomedical industrial park, No. 218 Xinghu street, Suzhou Industrial Park, China (Jiangsu) pilot Free Trade Zone, Suzhou, Jiangsu Province

Patentee after: Shengshi Taike Biopharmaceutical Technology (Suzhou) Co.,Ltd.

Address before: 215000 units 101 and 102, C11 building, phase I project of biomedical industrial park, 218 Xinghu street, Suzhou Industrial Park, Suzhou pilot Free Trade Zone, Suzhou, Jiangsu Province

Patentee before: CGENE TECH (SUZHOU, CHINA) Co.,Ltd.