CN113061142A - Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof - Google Patents

Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof Download PDF

Info

Publication number
CN113061142A
CN113061142A CN202010002951.8A CN202010002951A CN113061142A CN 113061142 A CN113061142 A CN 113061142A CN 202010002951 A CN202010002951 A CN 202010002951A CN 113061142 A CN113061142 A CN 113061142A
Authority
CN
China
Prior art keywords
alkyl
cycloalkyl
substituted
added
halogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010002951.8A
Other languages
Chinese (zh)
Inventor
张小虎
何苏丹
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.)
Aikono biomedical (Hong Kong) Co.,Ltd.
Original Assignee
Accro Bioscience Inc
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 Accro Bioscience Inc filed Critical Accro Bioscience Inc
Priority to CN202010002951.8A priority Critical patent/CN113061142A/en
Priority to EP21736180.7A priority patent/EP4085059B1/en
Priority to PCT/US2021/012124 priority patent/WO2021138694A1/en
Priority to US17/790,728 priority patent/US20230121233A1/en
Publication of CN113061142A publication Critical patent/CN113061142A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D495/20Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D497/00Heterocyclic compounds containing in the condensed system at least one hetero ring having oxygen and sulfur atoms as the only ring hetero atoms
    • C07D497/02Heterocyclic compounds containing in the condensed system at least one hetero ring having oxygen and sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D497/10Spiro-condensed systems

Abstract

The invention provides a compound of formula (I) or pharmaceutically acceptable salts, esters, solvates, prodrugs, isotopic labels, isomers thereof, wherein the compound of formula (I) has the structure:
Figure DDA0002354170480000011
the invention also provides application of the heterocyclic compound with the programmed cell necrosis pathway inhibitory activity. The heterocyclic compound having a programmed cell necrosis pathway inhibitory activity of the present invention, as a potent inhibitor of programmed cell necrosis pathway, can be used for the treatment or prevention of a disorder responsive to the RIP3 receptor.

Description

Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof
Technical Field
The present invention relates to a heterocyclic compound having an inhibitory activity on the programmed cell necrosis pathway, a composition comprising said compound, a process for the preparation of said compound and the use of said compound in medicine, in particular for the treatment of a variety of diseases responsive to inhibition of the RIP3 receptor, such as tumors, autoimmune diseases, neurodegenerative diseases, metabolic diseases and aging. The invention belongs to the technical field of medicines.
Background
Receptor-interacting protein 3(RIP3) is a member of the receptor-interacting protein family, and its gene is located on chromosome 11 of human (FEBS Lett.2000; 473:285-291) and is a polypeptide consisting of 518 amino acid residues. RIP3 is a serine/threonine protein kinase whose substrates include the homologous family kinase RIP1 and metabolic enzymes (Nat Rev Mol Cell biol. 2010; 11: 700-714). RIP3 has profound effects on physiological and pathological response processes such as cell survival, ontogeny, immunity, etc. (Nature.2011; 471: 368-. In particular, RIP3 is found to play a key role in exogenous apoptosis and programmed necrosis in recent years (science.2009; 325: 332-336; cell.2009; 137: 1100-1111; Nature.2011; 471: 363-367; cell.2009; 137:1112-1123), and the research on RIP3 is greatly promoted. The research shows that: RIP1 and RIP3 can trigger apoptosis by acting through their homotypic interaction Regions (RHIM) and composing complex II with aspartate proteolytic enzyme 8(caspase8) that recruits FADD and cysteine. When Caspase8 activity is inhibited or deleted, complex II will be converted into necrosis inducing signal complex with RIP1 and RIP3 as main components, i.e. necrosis complex. RIP3 is autophosphorylated in a complex state, and in addition to autophosphorylation, phosphorylation of RIP3 also modulates the kinase activity of RIP 1. The main phosphorylation sites of RIP3 are serine and threonine, wherein Ser227 is the most critical, MLKL of RIP3 substrate can be recruited and activated through phosphorylation to generate phosphorylation oligomers, and the movement of the MLKL oligomers from Cell sap to Cell membrane promotes the generation of programmed necrosis (Nature.2011; 471: 363-367; Cell,2012,150: 339-350; Nat Immu.2018; 19; 912-922).
Studies have shown that dysregulation of RIP3 is associated with a variety of pathological states and diseases. RIP3 signaling is involved in the control of a variety of viral infections, including Influenza A Virus (IAV) (FEBS J.2016; 283; 2616-2625), vaccinia virus (cell.2014; 137; 1112-1123), herpes simplex virus-1 (HSV-1) (Cell Host Microbe.2015; 17; 229-242), Murine Cytomegalovirus (MCMV) (Cell Host Microbe.2010; 7; 302-313) and West Nile Virus (WNV) (cell.2017; 169; 1-13). RIP3 mediated necrotic liver injury caused by excessive paracetamol (hepatology.2013; 58; 2099-2108) and alcohol-induced liver injury (hepatology.2013; 57; 1773-1783); thus, elimination of RIP3 may reduce or prevent such liver damage. TNF acts as a mediator that triggers inflammation in septic shock, suggesting that RIP3 is also involved in the development of bacterially induced sepsis. RIP 3-/-mice were found not to develop TNF-induced systemic inflammation in experimental models directed to clinical sepsis (Immunity.2011; 35: 908-918; Mol Med.2012; 18: 577-586). RIP3 also mediates epithelial cell necrosis and chronic (enteritis) enteritis (Nature.2011; 477: 330-) (334). Knock-out of RIP3 can reduce acute necrotizing pancreatitis induced by ranulin peptide (science.2009; 325: 332-. In an animal model of arterial thickening, the knock-out of RIP3 reduced macrophage necrosis in the foci (Cell Rep.2013; 3: 200-210). Deletion of RIP3 inhibited photoreceptor cell death and cone cell death in a model of retinal detachment (Proc Natl Acad Sci.2010; 107: 21695-21700; Proc Natl Acad Sci.2012; 109: 14598-14603). A slowing of reproductive senescence was observed in a male mouse model deficient in RIP3 or MLKL (elife.2017; 6: 6-12). RIP3 plays an important role in the apoptotic pathway, and downregulation of RIP3 leads to a blockage of the apoptotic pathway, as can be observed in acute myeloid leukemia, breast and colorectal cancers (Cell Res.2015; 25: 707-725; Cell Death.2017; 8: e 3084; Cell Death.2014.5: e 1384; Neoplasta.2015; 62: 592-601). Deficiency in RIP3 may also contribute to TAK1 deletion-induced hepatogenesis (Cell Rep.2014; 4: 776-790).
RIP3 is associated with a variety of diseases including tumors, autoimmune diseases, neurodegenerative diseases, metabolic diseases, and aging, and RIP3 is a potential therapeutic target for many diseases. Therefore, the development of the small-molecule inhibitor for inhibiting the activity of the RIP3 kinase can block programmed necrosis dependent on RIP3, slow down diseases or pathological states caused by programmed necrosis, generate a prevention or treatment effect, and have wide clinical application prospects.
Disclosure of Invention
Problems to be solved by the invention
In view of the above-mentioned drawbacks of the prior art, the present invention aims to provide a heterocyclic compound having RIP3 inhibitory activity, which is effective in inhibiting the cellular necrosis pathway and can be used for treating or preventing a disorder responsive to the RIP3 receptor, and use thereof.
Means for solving the problems
The purpose of the invention is realized by the following technical scheme:
a compound of formula (I) or a pharmaceutically acceptable salt, ester, solvate, prodrug, isotopic label, isomer thereof, wherein the compound of formula (I) has the structure:
Figure BDA0002354170460000031
wherein the content of the first and second substances,
n is 1 and 2;
x is selected from N or CR6
Y is selected from O, NR7、CR7R8
A is unsubstituted or substituted by 1 to 4R9Substituted C6-10Aryl or 5-10 membered heteroaryl, said heteroaryl containing 1-4 heteroatoms independently selected from O, N, S;
R1selected from hydrogen atom, deuterium atom, C1-6Alkyl radical, C3-6Cycloalkyl, said alkyl, cycloalkyl being unsubstituted or substituted by 1 to 3 halogen or deuterium atoms, preferably, R1Selected from hydrogen atom, deuterium atom, C1-6An alkyl group;
R2、R3,R7selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted with hydroxy, 1-3 halo or cyclopropyl; or R2And R3May be linked to form a ring B, preferably, R2、R3,R7Selected from hydrogen atom, deuterium atom, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted with hydroxy, 1-3 halo or cyclopropyl; or R2And R3May be linked to form ring B;
R4,R5,R6independently hydrogen atom, deuterium atom, cyano group, halogen, hydroxyl group, amino group, C1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C3-6Cycloalkyl, OC1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by 1 to 3 halogen, hydroxyOr, preferably, R4,R5,R6Independently hydrogen atom, deuterium atom, cyano, halogen, hydroxyl, amino;
R8selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, sulfone group, sulfoxide group, C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by hydroxy, OC1-3Alkyl, cyclopropyl, 1-3 halogen or deuterium atom substitutions;
or R8Selected from unsubstituted or substituted by 1-3R10Substituted C6-10Aryl or 5-10 membered heteroaryl, said heteroaryl containing 1-3 heteroatoms independently selected from O, N, S;
or R7And R8May be composed of ═ O and ═ CH2Or R7And R8May be linked to form ring C;
the ring B and the ring C are respectively and independently selected from 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 4-8 membered cycloalkenyl, 4-8 membered heterocycloalkenyl, the 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 4-8 membered heterocycloalkenyl are substituted by 1-3R11Substitution; the 3-8 membered heterocycloalkyl, 4-8 membered heterocycloalkenyl containing 1-3 heteroatoms independently selected from O, N, S or selected from-C (═ O) N (R)11)-、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)-、-S(=O)2and-NHC (═ O) NH-;
R9,R10each independently selected from hydrogen atom, deuterium atom, cyano, halogen, hydroxyl, amino, C1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C3-6Cycloalkyl, OC1-8Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, COOC1-6Alkyl, said amino, alkyl, cycloalkyl, alkenyl and alkynyl groups being unsubstituted or substituted with 1-3 halogen, hydroxy, amino, acetyl or deuterium atoms;
R11selected from hydrogen atomsDeuterium atom, C1-3Alkyl radical, C3-6Cycloalkyl, said alkyl, cycloalkyl being unsubstituted or substituted by 1 to 3 halogen or deuterium atoms.
In the present invention, preferably, X is selected from N atoms.
In the present invention, preferably, R is7And R8Is composed of ═ O and ═ CH2Or R7And R8When joined to form a ring C, R2、R3Selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by hydroxy, 1-3 halogen or cyclopropyl, or R2And R3May be linked to form ring B;
or R2And R3When joined to form a ring B, R8Selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by hydroxy, OC1-3Alkyl, cyclopropyl, 1-3 halogen or deuterium atoms substituted, or R8Selected from unsubstituted or substituted by 1-3R10A substituted 5-10 membered aromatic or heteroaromatic ring containing 1-3 heteroatoms independently selected from O, N, S;
in the present invention, preferably, A 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 BDA0002354170460000051
in the present invention, preferably, the heterocyclic compound includes:
Figure BDA0002354170460000061
the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of one or more of the compounds of formula (I) as described above, or a pharmaceutically acceptable salt, ester, solvate, prodrug, isotopic label, isomer thereof, and further comprising at least one pharmaceutically acceptable carrier.
The invention also provides a composition, which comprises the compound shown in the formula (I) or pharmaceutically acceptable salt, ester, solvate, prodrug, isotope label, isomer or the pharmaceutical composition and one or more of antitumor drugs, autoimmune disease resistant drugs, neurodegenerative disease resistant drugs, metabolic disease resistant drugs and anti-aging drugs.
In order to more clearly describe the context of the present invention, the terms referred to will now be defined as follows:
the term "halogen" denotes fluorine, chlorine, bromine or iodine, especially fluorine, chlorine or bromine, alone or in combination.
The term "C1-6Alkyl "represents, alone or in combination, a saturated, linear or branched alkyl group containing from 1 to 6, in particular from 1 to 3, carbon atoms, including methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, n-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-pentyl, 2, 3-dimethyl-2-butyl, 3, -dimethyl-2-butyl, and the like. Preferably, "C1-C10The alkyl group "is any of methyl, ethyl, n-propyl, isopropyl, and tert-butyl. Similarly, the term "C1-3Alkyl "alone or in combination means a saturated straight or branched chain alkyl group containing 1 to 3 carbon atoms, including methyl, ethyl, propyl, isopropyl, and the like.
The term "OC1-6Alkyl "alone or in combination denotes the radical C1-6alkyl-O-in which "C1-6Alkyl represents as defined above, including but not limited to methoxy (-OCH)3) Ethoxy (-OCH)2CH3) N-propoxy group (-OCH)2CH2CH3) I-propoxy (-OCH (CH)3)2) N-butoxy (-OCH)2CH2CH2CH3) Sec-butoxy (-OCH (CH)3)CH2CH3) Isobutoxy (-OCH)2CH(CH3)2) T-butoxy (-OC (CH))3)3) N-pentyloxy (-OCH)2CH2CH2CH2CH3) Neopentyloxy (-OCH)2C(CH3)3) And the like.
The term "3-8 membered cycloalkyl" refers to saturated or partially unsaturated monocyclic or polycyclic cycloalkyl groups having 3 to 8, especially 3-6, carbon atoms, either alone or in combination, including cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. Similarly, the term "C3-6Cycloalkyl "means, alone or in combination, a saturated or partially unsaturated monocyclic or polycyclic cycloalkyl group having 3 to 6 carbon atoms.
The term "3-8 membered heterocyclyl" means a saturated or partially unsaturated monocyclic or polycyclic heterocyclyl group comprising 3-8, particularly 3-6, more particularly 5-6 carbon atoms and a heteroatom or heteroatom group selected from N, NH, O, C (O), S (O)m(wherein m is 0, 1 or 2); the 3-to 8-membered heterocyclic group includes aziridinyl, azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuryl, tetrahydrothienyl, piperidinyl, morpholinyl, piperazinyl, thiomorpholinyl, tetrahydropyranyl, 1-dioxothiomorpholinyl, butyrolactam, valerolactam, caprolactam, butyrolactone, valerolactam or caprolactone, and the like. Similarly, the term "C3-6Heterocyclyl "means a saturated or partially unsaturated monocyclic or polycyclic heterocyclyl group comprising 3 to 6, more particularly 5 to 6, carbon atoms and a heteroatom or heteroatom groupThe heteroatom or heteroatom group is selected from N, NH, O, C (O), S (O)m(wherein m is 0, 1 or 2).
The term "aryl" denotes any stable 6-10 membered monocyclic or bicyclic aromatic group including phenyl, naphthyl, tetrahydronaphthyl, 2, 3-indanyl or biphenyl and the like. The hydrogen atoms on the "aryl" are independently optionally substituted with one or more substituents described herein.
The term "heteroaryl" denotes an aromatic cyclic group formed by replacement of a carbon atom on the ring by at least one heteroatom selected from sulfur, oxygen or nitrogen, which aromatic cyclic group may be a 5-7 membered monocyclic or 7-12 bicyclic group. In the present invention, the number of hetero atoms in the heteroaryl group is preferably 1,2,3 or 4, and examples thereof include thienyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyridin-2 (1H) -onyl, pyridin-4 (1H) -onyl, pyrrolyl, pyrazolyl, thiazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, imidazolyl, tetrazolyl, isothiazolyl, oxazolyl, isoxazolyl, thiadiazolyl, oxadiazolyl, naphthyl, benzothienyl, indolyl, benzimidazolyl, benzothiazolyl, benzofuranyl, quinolyl, isoquinolyl, quinazolinyl and the like. The hydrogen atoms on the "heteroaryl" groups are independently optionally substituted with one or more substituents as described herein.
The term "C6-10Aryl "denotes an aryl group having 6 to 10 carbon atoms, wherein aryl denotes as defined above.
The term "5-10 membered heteroaryl" denotes a heteroaromatic ring having 5-10 carbon atoms and heteroatoms, wherein heteroaromatic ring denotes as defined above.
The term "amino" denotes, alone or in combination, a primary amino group (-NH)2) Secondary amino (-NH-) or tertiary amino
Figure BDA0002354170460000081
The term "NHC1-6Alkyl and the term "N (C)1-6Alkyl radical)2"alone or in combination denotes an amino group as defined above, wherein the hydrogen atoms of the amino group are substituted by one and two C, respectively1-6Alkyl is substituted in which "C1-6Alkyl "denotes as defined above.
The term "isomer" encompasses all isomeric forms including enantiomers, diastereomers, tautomers and geometric isomers (including cis-trans isomers). Thus, individual stereochemical isomers of the contemplated compounds of the present invention or mixtures of enantiomers, diastereomers, tautomers or geometric isomers (or cis-trans isomers) thereof are intended to be within the scope of the present invention.
The term "pharmaceutically acceptable salts" means that the compounds of the present invention exist in the form of their pharmaceutically acceptable salts, including acid addition salts and base addition salts. Pharmaceutically acceptable salts are described in pharmaceutical salts described in J.pharmaceutical Sciences (Vol.66: pp.1-19, 1977) by S.M.Berge. In the present invention, pharmaceutically acceptable non-toxic acid addition salts mean salts of the compounds of the present invention with organic or inorganic acids including, but not limited to, hydrochloric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, phosphoric acid, nitric acid, perchloric acid, acetic acid, oxalic acid, maleic acid, fumaric acid, tartaric acid, benzenesulfonic acid, methanesulfonic acid, salicylic acid, succinic acid, citric acid, lactic acid, propionic acid, benzoic acid, p-toluenesulfonic acid, malic acid and the like. Pharmaceutically acceptable non-toxic base addition salts mean salts of the compounds of the invention with organic or inorganic bases, including but not limited to alkali metal salts, such as lithium, sodium or potassium salts; alkaline earth metal salts, such as calcium or magnesium salts; salts of organic bases, e.g. ammonium salts formed by reaction with organic bases containing N groups or N+(C1-6Alkyl radical)4The salt is preferably lithium hydroxide, sodium hydroxide, potassium hydroxide, sodium carbonate, sodium hydrogencarbonate, potassium carbonate, potassium hydrogencarbonate, magnesium carbonate, calcium carbonate, ammonia water, triethylamine, tetrabutylammonium hydroxide, or the like. The "pharmaceutically acceptable salt" can be synthesized by a general chemical method.
The term "solvate" refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, methanol, ethanol, isopropanol, ethyl acetate, tetrahydrofuran, N-dimethylformamide, dimethylsulfoxide, and the like.
The term "ester" is used to denote organic esters, including monoesters, diesters, triesters, and more generally polyesters.
The term "prodrug" means a chemical derivative which is a compound of the present invention and is converted into a compound represented by the general formula I in vivo by a chemical reaction.
The term "isotopic label" means an isotope including, but not limited to2H,3H,11C,13C,14C,15N,17O,18O,18F,32P,35S,36Cl, and the like.
Effects of the invention
The heterocyclic compound with the RIP3 inhibitory activity, as a potent inhibitor of RIP3, can be used for treating or preventing diseases responding to RIP3 receptor.
Drawings
FIG. 1 is a graph showing the results of example 16 on heterocyclic compound B4;
FIG. 2 is a graph showing the results of example 17 on heterocyclic compound B4;
FIG. 3 is a graph showing the results of example 18 on heterocyclic compound B1.
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 used in Qingdao oceanic chemical plant and tobacco station chemical industryA factory product; 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); mobile phase 0.1% NH4HCO3Aqueous solution: acetonitrile (from 5: 95 to 95: 5 in 5 minutes); the flow rate was 1.8 mL/min.
Example 1
A heterocyclic compound B1 synthesized by the method comprising:
Figure BDA0002354170460000101
1) synthesis of intermediate B1-1
2-chloro-5-nitrobenzaldehyde (20.0g,108mmol) was dissolved in N, N-dimethylformamide (200mL), potassium carbonate (29.8g,216mmol) was added, and methyl thioglycolate (12.6g,119mmol) was added dropwise to the reaction system at 0 ℃ via a constant pressure dropping funnel. After the addition was complete, the mixture was stirred at room temperature overnight. After completion of the reaction, the reaction mixture was poured into water (1000mL) as it was to precipitate a solid, filtered, and the filter cake was washed with water (200mL × 2) and dried to obtain yellow solid B1-1(22g, 86%).1H NMR(400MHz,DMSO-d6)δ8.99(s,1H),8.43(s,1H),8.39-8.28(m,2H),3.92(s,3H).
2) Synthesis of intermediate B1-2
B1-1(22.0g,93mmol) was dissolved in a mixed solvent of methanol (200mL) and water (200mL), and sodium hydroxide (14.8g,371mmol) was added to the reaction system to react at 70 ℃ for 3 hours. After the reaction was completed, the pH was slowly adjusted to about 2 with concentrated hydrochloric acid (100mL) under ice-bath conditions, a solid precipitated, the filtrate was filtered, the filter cake was washed with water (100mL), and the filter cake was dried to give B1-2(19.0g, 92%) as a white solid.1H NMR(400MHz,DMSO-d6)δ8.97(s,1H),8.37-8.25(m,3H).
3) Synthesis of intermediate B1-3
B1-2(19.0g,85mmol) was dissolved in quinoline (100mL), copper powder (5.5g,85mmol) was added, and the mixture was reacted at 170 ℃ for 3 hours under a nitrogen atmosphere. After the reaction was completed, ethyl acetate (400mL) was added to the reaction mixture to return the reaction mixture to room temperature, 6N hydrochloric acid (300mL) was added to the filtrate to adjust the pH to acidic, and the organic phase was separated. The organic phase was washed successively with 2N hydrochloric acid (200mL) and saturated sodium bicarbonate solution (200mL), dried and the organic phase was concentrated. The residue was slurried with ethyl acetate (100mL) for 2 hours, filtered, and the filter cake was slurried with water (200mL) for 2 hours, filtered, and dried to give solid B1-3(13.1g, 86%).1H NMR(400MHz,DMSO-d6)δ8.84(s,1H),8.29(d,J=9.2Hz,1H),8.17(d,J=8.8Hz,1H),8.05(d,J=5.2Hz,1H),7.72(d,J=5.6Hz,1H).
4) Synthesis of intermediate B1-4
Crude product B1-3(1.4g, 7.8mmol) was dissolved in dichloromethane (25mL), m-chloroperoxybenzoic acid (4.0g, 19.5mmol) was added slowly under ice bath, allowed to return to room temperature after 10 min, and stirred overnight. The reaction was quenched with saturated aqueous sodium sulfite, and the organic phases were extracted with dichloromethane (40mL x 3), combined and dried over anhydrous sodium sulfate to give crude product B1-4(1.2g, 73%). LC-MS (M/z) 212.9[ M + H]+.
5) Synthesis of intermediate B1-5
B1-4(1.2g, 5.7mmol) was dissolved in ethanol (20mL), iron powder (1.3g, 22.8mmol) was added, the temperature was gradually raised to 85 ℃ and ammonium chloride (1.2g, 22.8mmol) was dissolved in water (10mL) and added to the reaction mixture, and the mixture was stirred at 85 ℃ for 2 hours. The reaction mixture was diluted with dichloromethane (50mL), the iron powder was removed by celite filtration, the filter cake was washed with dichloromethane, water (30mL) was added to the liquid phase, the phases were separated, the organic phases were further extracted with dichloromethane (200mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give a brown solid. Ethyl acetate (10mL) was added for slurrying and filtered to give intermediate B1-5(0.8g, 77%) as a grey solid.1H NMR(400MHz,CDCl3)δ8.42(d,J=8.4Hz,1H),8.22(s,1H),7.90(d,J=8.0Hz,1H),7.33(d,J=6.8Hz,1H),6.92(d,J=6.8Hz,1H).
6) Synthesis of intermediate B1-6
B1-5(800mg, 4.4mmol) was dissolved in ethanol (10mL), trimethyl orthoformate (560mg, 5.3mmol) and 2, 2-dimethyl-1, 3-dioxane-4, 6-dione (760mg, 5.3mmol) were added, the mixture was stirred at 85 ℃ for 2 hours, cooled to room temperature, a solid precipitated, filtered, the filter cake was washed with ethanol (5mL) and dried to give intermediate B1-6(1.4g, 90%) as a pale yellow solid. LC-MS (M/z) 333.7[ M-H]-.
7) Synthesis of intermediate B1-7
Diphenyl ether (10mL) was added to a 25mL round bottom flask, heated to 220 ℃ and stirred for 10 minutes while maintaining the temperature to remove the solvent water, B1-6(837mg, 2.5mmol) was added to the reaction in portions, and the mixture was stirred for 30 minutes at 220 ℃. Cooling to room temperature precipitated a solid, which was filtered and the filter cake was washed with ether to give intermediate B1-7(450mg, 77%) as a crude grey product. LC-MS (M/z):233.8[ M + H]+.
8) Synthesis of intermediate B1-8
Crude product B1-7(450mg, 1.93mmol) was dissolved in phosphorus oxychloride (6mL), the mixture was stirred at 110 ℃ for 2 hours, cooled to room temperature, concentrated to remove phosphorus oxychloride, the residue was dissolved in ethyl acetate (20mL) to form a suspension, the remaining phosphorus oxychloride and hydrochloric acid were neutralized with saturated aqueous sodium bicarbonate, the organic phases were extracted with ethyl acetate (10mL x 3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated, and purified on a silica gel column to give intermediate B1-8 as a gray solid (152mg, 32%).1H NMR(400MHz,DMSO-d6)δ8.99(d,J=4.8Hz,1H),8.62(s,1H),8.26(s,1H),7.93(d,J=4.8Hz,1H),7.89(d,J=6.8Hz,1H),7.67(d,J=6.8Hz,1H).
9) Synthesis of intermediate B1-9
B1-8(50mg, 0.2mmol) and benzothiazole (33mg, 0.22mmol) were dissolved in ethanol (4mL), the mixture was stirred in a microwave reactor at 130 ℃ for 30 minutes, a solid precipitated, filtered, the filter cake was washed with a small amount of ethanol and dried to give the yellow end product B1-9(55mg, 68%).1H NMR(400MHz,DMSO-d6)δ14.79(br s,1H),11.15(s,1H),9.54(s,1H),9.31(s,1H),8.56(d,J=7.2Hz,1H),8.39(d,J=8.4Hz,1H),8.22(s,1H),8.16(s,1H),8.00(d,J=7.6Hz,1H),7.81(d,J=6.8Hz,1H),7.61(d,J=8.4Hz,1H),7.00(d,J=6.8Hz,1H).LC-MS(m/z):365.7[M+H]+.
10) Synthesis of end product B1
B1-9(50mg, 0.14mmol) was dissolved in methanol (30mL), 5% palladium on carbon (5mg, 0.05mmol) was added, and the mixture was stirred under hydrogen at room temperature for 24 hours. Filtration, concentration of the filtrate under reduced pressure and purification of the residue through a silica gel column gave final product B1(24mg, 47%) as a pale yellow solid.
Example 2
A heterocyclic compound B2 synthesized by the method comprising:
Figure BDA0002354170460000131
1) synthesis of intermediate B2-1
B1-3(12.0g,67.0mmol) was dissolved in N, N-dimethylformamide (150mL) and N-bromosuccinimide (13.1g,74.0mmol) was added to the solution, and the mixture was reacted at 60 ℃ for 3 hours under a nitrogen atmosphere. After completion of the reaction, N-dimethylformamide was spin-dried using an oil pump, and the residue was slurried with ethyl acetate (150mL) overnight, filtered, and the filter cake was slurried with water (150mL) for 1 hour, filtered, and dried to give solid B2-1(13g, 75%).1H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.40(d,J=8.8Hz,1H),8.32-8.25(m,2H).
2) Synthesis of intermediate B2-2
B2-1(21.0g,81.4mmol) was dissolved in 300mL of dichloromethane, stirred under ice-bath, added with m-chloroperoxybenzoic acid (42.0g,203.5mmol), allowed to warm to room temperature and stirred overnight. After completion of the reaction, filtration was carried out, the filtrate was spin-dried, a saturated aqueous sodium bicarbonate solution (200mL) was added thereto, the pH was adjusted to 8, the aqueous phase was extracted with methylene chloride (200mL), the organic phase was separated, dried, spin-dried, and slurried with ethyl acetate (150mL) to give B2-2(1.3g, 96%) as a white solid.1H NMR(400MHz,DMSO-d6)δ8.56(dd,J=8.4Hz,J=1.2Hz,1H),8.34(s,1H),8.30(d,J=8.4Hz,1H),8.20(d,J=1.6Hz,1H).
3) Synthesis of intermediate B2-3
B2-2(20.8g, 71.7mmol) was dissolved in ethanol/water (300mL:100mL),ammonium chloride solid (15.3g,287mmol) was added at 85 deg.C and iron powder (16.1g,287mmol) was added slowly. After the reaction was complete, it was cooled to room temperature, filtered, the filtrate was spin-dried, slurried with water (500mL), filtered, and the filter cake was dried to give B2-3(15g, 81%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ7.84(s,1H),7.47(s,1H),6.89-6.59(m,2H),6.44(s,2H).
4) Synthesis of intermediate B2-4
Intermediate B2-3(13.5g, 51.9mmol) was dissolved in ethanol (120mL), and 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (14.3g,77.9mmol) was slowly added thereto at room temperature, followed by stirring at room temperature. After the reaction was complete, it was directly filtered and the filter cake was dried to give B2-4(18g, 86%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ11.42(d,J=12.8Hz,1H),8.68(d,J=13.6Hz,1H),8.13(s,1H),7.99(s,1H),7.90(s,2H),1.69(s,6H).
5) Synthesis of intermediate B2-5
Diphenyl ether (720mL) was heated to 220 deg.C and intermediate B2-4(18.0g,43.6mmol) was added portionwise. The reaction mixture was cooled to room temperature, petroleum ether (500mL) was added thereto, and after stirring for 0.5 hour, the mixture was filtered, and the filter cake was slurried with ethyl acetate (100 mL). The filter cake was dried to give intermediate B2-5(5.5g, 41%).1H NMR(400MHz,DMSO-d6)δ12.31(s,1H),8.35(s,1H),8.25(s,1H),8.08(d,J=7.2Hz,1H),7.77(s,1H),6.23(d,J=6.8Hz,1H).
6) Synthesis of intermediate B2-6
B2-5(5.5g,17.7mmol) was dissolved in phosphorus oxychloride (50mL) and stirred at 110 ℃ overnight. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, a dichloromethane (50mL) solution was added at 0 ℃ and a saturated aqueous sodium bicarbonate solution (50mL) was added to adjust the pH to 8, the organic phase was extracted and separated, and the organic phase was spin-dried to obtain a yellow solid product B2-6(5g, 86%).1H NMR(400MHz,DMSO-d6)δ9.06(d,J=4.0Hz,1H),8.76(s,1H),8.36(s,1H),8.16(s,1H),8.01(d,J=4.0Hz,1H).
7) Synthesis of intermediate B2-7
B2-6(5.5g,17.7mmol) was dissolved in phosphorus oxychloride (50mL) and stirred at 110 ℃ overnight. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in isopropanol (20mL),5-aminobenzothiazole (3.2g,21.3mmol) was added and the reaction stirred at 95 ℃ for 4 hours. After cooling to room temperature, filtration was carried out and the filter cake was washed with isopropanol (50 mL). The filter cake was dried to give product B2-7(6g, 77%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ11.37(s,1H),9.55(s,1H),9.43(s,1H),8.63(d,J=6.8Hz,1H),8.50(s,1H),8.40(d,J=8.8Hz,1H),8.27-8.21(m,2H),7.62(d,J=8.8Hz,1H),7.04(d,J=7.2Hz,1H).
8) Synthesis of end product B2
Intermediate B2-7(450mg,0.9mmol) was dissolved in ethylene glycol (10mL), cesium carbonate (826mg,2.5mmol) was added, and the mixture was stirred at 60 ℃ for 5 hours. The reaction solution was spin dried, and the residue was purified by column chromatography (dichloromethane: methanol ═ 100:1) to give a crude product, which was then dissolved in acetone (50mL), 2N ethyl acetate hydrochloride (2mL) was added, and a solid precipitated, which was stirred for 10 minutes, filtered, and the filter cake was dried to give the final product B2 hydrochloride (55mg, 13%).
According to this synthesis method, a heterocyclic compound B29 was synthesized using different substrates.
Example 3
A heterocyclic compound B3 synthesized by the method comprising:
Figure BDA0002354170460000151
1) synthesis of intermediate B3-1
Intermediate B2-7(300mg,0.63mmol) was dissolved in methanol (300mL), cesium carbonate (462mg,1.42mmol) was added, and the mixture was stirred at room temperature for 20 hours. The reaction solution was spin dried, and the residue was purified by column chromatography (dichloromethane: methanol ═ 100:1) to give a crude product, which was dissolved in acetone (50mL), 2N ethyl acetate hydrochloride (2mL) was added, and a solid was precipitated, which was stirred for 10 minutes, filtered, and the cake was dried to give intermediate B3-1 hydrochloride (105mg, 39%).1H NMR(400MHz,DMSO-d6)δ14.88(s,1H),11.25(s,1H),9.54(s,1H),9.34(s,1H),8.60(d,J=6.8Hz,1H),8.40(d,J=8.4Hz,1H),8.23(s,1H),8.17(s,1H),7.62(d,J=8.4Hz,1H),7.16(s,1H),7.01(d,J=7.2Hz,1H),4.07(s,3H).LCMS(ESI/APCI)m/z:395.7[M+H]+.
2) Synthesis of end product B3
Intermediate B3-1 hydrochloride (15mg, 0.04mmol) was dissolved in acetonitrile (2mL), concentrated hydrochloric acid (0.5mL) was added, the mixture was stirred at 80 ℃ overnight, TLC monitored for reaction completion, water (20mL) was added to the reaction solution, a solid precipitated, the solid was filtered and washed with isopropanol (5mL), dried to give the final product B3(12mg, 83%) as a yellow solid.
Example 4
A heterocyclic compound B4 synthesized by the method comprising:
Figure BDA0002354170460000161
1) synthesis of intermediate B4-1
Methyl 2-fluoro-5-nitrobenzoate (6.0g,30mmol) was dissolved in dimethyl sulfoxide (50mL), sodium methylsulfite (3.06g,30mmol) was added, and the mixture was stirred at room temperature for 4 hours. 60% sodium hydride (1.2g,30mmol) was added and stirred at room temperature overnight. The pH was adjusted to 2 by adding 1N aqueous hydrochloric acid solution, and water (200mL) was added and stirred for 2 hours. Filtration and washing of the filter cake with water (50mL x 3). The filter cake was dried to give intermediate B4-1 as a yellow solid (crude 6.96 g).1H NMR(400MHz,CDCl3)δ8.82(s,1H),8.78(d,J=8.4Hz,1H),8.21(d,J=8.4Hz,1H),4.25(s,2H).
2) Synthesis of intermediate B4-2
Intermediate B4-1(2.27g,10m mol) was dissolved in N, N-dimethylformamide (15mL), methyl iodide (5.68g,40mmol) and DBU (6.09g,40mmol) or potassium carbonate (5.52g,40mmol) were added, the mixture was stirred at room temperature for 3 hours, and the mixture was heated to 40 ℃ and stirred overnight. After the reaction solution cooled to room temperature, it was quenched by addition of water (20mL) and extracted by addition of ethyl acetate (30mL × 2). The organic phases were combined and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 10:1) to give intermediate B4-2(2.0g, 78%) as a yellow solid.1H NMR(400MHz,CDCl3)δ8.81(s,1H),8.77(d,J=8.4Hz,1H),8.23(d,J=8.4Hz,1H),1.66(s,6H).LC-MS(m/z):277.7[M+Na]+.
3) Synthesis of intermediate B4-3
Intermediate B4-2(200mg,0.78 m)mol) was dissolved in isopropanol (20mL), and wet palladium on carbon (200mg) with a 10% content and concentrated hydrochloric acid (1mL) were added, and the atmosphere of hydrogen was replaced and stirred overnight. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was extracted with saturated sodium carbonate solution (3mL) and ethyl acetate (20 mL). The organic phase was concentrated under reduced pressure to give intermediate B4-3(180mg, 99%) as a yellow solid.1H NMR(400MHz,CDCl3)δ7.75(d,J=8.4Hz,1H),7.12-7.07(m,1H),7.07-7.05(m,1H),4.77(br s,2H),1.58(s,6H).
4) Synthesis of intermediate B4-4
Intermediate B4-3(1.52g,6.75mmol) was dissolved in a mixed solvent of methanol (30mL) and water (10mL), potassium hydrogen persulfate (4.15g,6.75mmol) was added thereto at room temperature, and a 10mL aqueous solution of sodium bromide (0.69g,6.75mmol) was slowly added dropwise thereto, followed by stirring at room temperature overnight. Saturated sodium sulfite solution was added to adjust the pH to 8, ethyl acetate (100mL) was added for extraction, and the ethyl acetate phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane: methanol ═ 100:1) to give intermediate B4-4(1.53g, 75%) as a yellow solid product.1H NMR(400MHz,CDCl3)δ7.74(d,J=8.4Hz,1H),7.18(d,J=8.0Hz,1H),4.95(br s,2H),1.60(s,6H).
5) Synthesis of intermediate B4-5
Intermediate B4-4(165mg,0.54mmol) was dissolved in methanol (5mL), and 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (151mg,0.81mmol) was added and stirred at room temperature overnight. The reaction solution was filtered, and the filter cake was washed with ethanol (6 mL). The filter cake was dried to give intermediate B4-5(235mg, 94%) as a white solid.1H NMR(400MHz,CDCl3)δ12.04(s,1H),8.77-8.66(m,1H),8.08(d,J=7.6Hz,1H),7.95-7.85(m,1H),1.80(s,6H),1.65(s,6H).
6) Synthesis of intermediate B4-6
Diphenyl ether (5mL) was heated to 220 deg.C and intermediate B4-5(215mg,0.514mmol) was added portionwise and stirred for 1 hour with constant temperature. The reaction mixture was cooled to room temperature, petroleum ether (20mL) was added with stirring, the mixture was filtered, and the filter cake was washed with petroleum ether (30 mL). The filter cake was purified by silica gel column chromatography (dichloromethane: methanol ═ 50:1) to give intermediate B4-6(30mg, 18%) as a yellow solid product.1H NMR(400MHz,DMSO-d6)δ11.78(s,1H),8.60(s,1H),8.09-8.02(m,1H),6.34(d,J=7.2Hz,1H),1.54(s,3H).LC-MS(m/z):355.6[M+H]+
7) Synthesis of intermediate B4-7
B4-6(30mg,0.084mmol) was dissolved in phosphorus oxychloride (6mL) and stirred at 110 ℃ overnight. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in isopropanol (6mL), and 5-aminobenzothiazole (15mg,0.101mmol) was added to stir the reaction mixture at 90 ℃ overnight. After cooling to room temperature, filtration was carried out and the filter cake was washed with isopropanol (6 mL). The filter cake was dried to give intermediate B4-7 (crude 45mg) as a yellow solid.
8) Synthesis of final product B4:
intermediate B4-7 (crude 45mg,0.084mmol) was dissolved in isopropanol (6mL) and water (0.5mL), and 10% content of wet palladium on carbon (100mg) and triethylamine (0.5mL) were added to replace the hydrogen atmosphere, and the reaction was stirred at 90 ℃ overnight. The reaction was cooled to room temperature, filtered, and the filter cake was washed with methanol (30 mL). The filtrate was concentrated under reduced pressure and the residue was purified twice by thin layer preparative plate chromatography (dichloromethane: methanol ═ 25:1) to give final product B4(5mg, 14%) as a yellow solid.
Example 5
A heterocyclic compound B5 synthesized by the method comprising:
Figure BDA0002354170460000181
1) synthesis of intermediate B5-1
Methyl 2-fluoro-5-nitrobenzoate (4.0g,20mmol) was dissolved in 50mL of dimethyl sulfoxide, sodium ethylsulfonate (2.32g,20mmol) was added, and the mixture was stirred at room temperature overnight. Sodium hydride (805mg,20moL) was added portionwise under ice bath conditions and stirring was continued for 3 h. Iodothane (9.36g, 60mmol) was added and stirring continued for 5 h. Ethyl acetate (300mL) was added, the pH was adjusted to 5-6 with 1N hydrochloric acid, the organic phase was separated, concentrated under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate: 10:1-7:1) to give intermediate B5-1(2.4g, 45%) as a white gray solid.1H NMR(400MHz,CDCl3)δ8.80(s,1H),8.75(d,J=8.4Hz,1H),8.22(d,J=8.4Hz,1H),2.26-1.95(m,2H),1.64(s,3H),1.10(t,J=7.2Hz,3H).
2) Synthesis of intermediate B5-2
Intermediate B5-1(1.0g,3.7mmol) was dissolved in 40mL of ethanol, 5mL of water, ammonium chloride (980mg,18.5mmol) and iron powder (829mg,14.8mmol) were added, and the mixture was heated to 70 ℃ for reaction for 4 h. The hot solution was filtered, the filter cake was washed three times with dichloromethane (80mL x 3), saturated aqueous sodium bicarbonate (100mL) was added and the organic phase was concentrated under reduced pressure to give intermediate B5-2(700mg, 60%) as a yellow solid.1H NMR(400MHz,CDCl3)δ7.74(d,J=8.0Hz,1H),7.10-7.00(m,2H),4.38(s,2H),2.20-1.98(m,2H),1.56(s,3H),1.06(t,J=7.2Hz,3H).
3) Synthesis of intermediate B5-3
Intermediate B5-2(567mg,2.4mmol) was dissolved in a mixed solvent of methanol (20mL) and water (10mL), potassium hydrogen persulfate (1.46mg,2.4mmol) was added thereto at room temperature, and sodium bromide (243mg,2.4mmol) was slowly added dropwise to the solution in 2mL of water, followed by stirring at room temperature for 4 hours. The reaction was quenched by addition of saturated sodium sulfite solution (30 mL). Extraction was performed by the addition of dichloromethane (40mL) and the organic phase was concentrated under reduced pressure to give intermediate B5-3(500mg, crop) as a yellow solid. 4) Synthesis of intermediate B5-4 (0085-
Intermediate B5-3(375mg,1.1mmol) was dissolved in 20mL ethanol, 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (255mg,1.4mmol) was added, and the mixture was stirred at room temperature for 20 minutes. The reaction solution was filtered to obtain a white solid. The diphenyl ether was warmed to 240 ℃ and the white solid was added in portions, reacted for 10 minutes and returned to room temperature. Filtration and washing of the filter cake three times with ether (3mL x 3) gave B5-4 as a brown solid (100mg, 19%).1H NMR(400MHz,DMSO-d6)δ11.77(s,1H),8.59(s,1H),8.05(s,1H),6.35(s,1H),2.10-1.98(m,2H),1.53(s,3H),0.96(t,J=7.2Hz,3H).
5) Synthesis of intermediate B5-5
Intermediate B5-4(90mg,0.28mmol) was dissolved in isopropanol (15mL), added to water (1.5mL), and the atmosphere of 10% palladium on carbon (18mg) and triethylamine (1.0mL) was replaced with hydrogen and stirred at room temperature for 1.5 h. Saturated aqueous sodium bicarbonate (20mL) and dichloromethane (40mL) were added, the organic phase was separated and concentrated under reduced pressure to give intermediate B5-5(50mg, 61%) as a gray solid.1H NMR(400MHz,DMSO-d6)δ12.41(br s,1H),8.62(s,1H),8.22-8.10(m,2H),6.27(d,J=7.6Hz,1H),2.10-1.90(m,2H),1.51(s,3H),0.97(t,J=7.6Hz,3H).LCMS(ESI/APCI)m/z:291.7[M+H]+.
6) Synthesis of end product B5
B5-5(35mg,0.12mmol) was dissolved in phosphorus oxychloride (2mL) and stirred at 110 ℃ for 30 min. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in isopropanol (5mL), to which was added 5-aminobenzothiazole (22mg,0.14 mmol). The reaction was stirred at 95 ℃ for 1 hour. After cooling to room temperature, filtration and washing of the filter cake twice with ether (3mL × 2) gave B5(15mg, 30%) as a yellow solid.
Example 6
A heterocyclic compound B6 synthesized by the method comprising:
Figure BDA0002354170460000201
1) synthesis of intermediate B6-1
Methyl 2-fluoro-5-nitrobenzoate (4.0g,20mmol) was dissolved in dimethyl sulfoxide (30mL), sodium ethyl sulfite (2.32g,20mmol) was added, and the mixture was stirred at room temperature for 16 hours. 60% sodium hydride (0.8g,20mmol) was added and stirred at room temperature for 2 hours. Allyl bromide (3.63g,30mmol) was added and stirred at room temperature for 1 hour. The pH was adjusted to 2 by adding 1N aqueous hydrochloric acid solution, and extraction was performed by adding ethyl acetate (200 mL). The organic phase was concentrated under reduced pressure and the residue was purified by column chromatography on silica gel (petroleum ether: ethyl acetate: 20:1) to give intermediate B6-1(3.5g, 62%) as a white solid.1H NMR(400MHz,CDCl3)δ8.87-8.66(m,2H),8.30-8.13(m,1H),5.92-5.67(m,1H),5.32-5.11(m,2H),2.94-2.54(m,2H),1.68-1.58(m,3H).
2) Synthesis of intermediate B6-2
Intermediate B6-1(3.5g,12.4mmol) and ammonium chloride (2.66g,50mmol) were dissolved in ethanol (100mL) and water (10mL), and reduced iron powder (2.8g,50mmol) was added, and the mixture was heated at 80 ℃ and stirred for 1 hour. After the reaction solution was cooled to room temperature, it was filtered and the filter cake was washed with methanol (100 mL). The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol ═ 20:1) to give intermediate B6-2(3.14g, 100%) as a yellow oily product.1H NMR(400MHz,CDCl3)δ7.75(d,J=8.4Hz,1H),7.08(d,J=8.4Hz,1H),7.05(s,1H),5.93-5.80(m,1H),5.24-5.13(m,2H),4.38(s,2H),2.81-2.64(m,2H),1.54(s,3H).
3) Synthesis of intermediate B6-3
Intermediate B6-2(3.12g,12.4mmol) was dissolved in a mixed solvent of methanol (100mL) and water (40mL), oxone (7.65g,12.4mmol) was added thereto at room temperature, and sodium bromide (1.28g,12.4mmol) was slowly added dropwise to a 10mL aqueous solution, followed by stirring at room temperature for 2 hours. Saturated sodium sulfite solution was added to adjust pH to 8, and ethyl acetate (20mL × 3) was added for extraction. The organic phases were combined and the organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane: methanol ═ 100:1) to give intermediate B6-3(2.7g, 66%) as a yellow solid.1H NMR(400MHz,CDCl3)δ7.73(d,J=8.0Hz,1H),7.16(d,J=8.4Hz,1H),5.92-5.79(m,1H),5.24-5.15(m,2H),4.94(s,2H),2.81-2.65(m,2H),1.56(s,3H).
4) Synthesis of intermediate B6-4
Intermediate B6-3(2.7g,8.2mmol) was dissolved in ethanol (20mL), 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (2.28g,12.3mmol) was added, and the mixture was stirred at room temperature for 7 hours. The reaction solution was filtered, and the filter cake was washed with ethanol (6 mL). The filter cake was dried to give intermediate B6-4(3.0g, 76%) as a white solid.1H NMR(400MHz,CDCl3)δ12.03(d,J=13.2Hz,1H),8.71(d,J=13.2Hz,1H),8.06(d,J=8.8Hz,1H),7.89(d,J=8.4Hz,1H),5.86-5.73(m,1H),5.25-5.18(m,2H),2.84-2.68(m,2H),1.78(s,6H),1.60(s,3H).
5) Synthesis of intermediate B6-5
Diphenyl ether (100mL) was heated to 220 deg.C and intermediate B6-4(3.0g,6.2mmol) was added portionwise with stirring for 0.5 h at RT. The reaction mixture was cooled to room temperature, petroleum ether (150mL) was added with stirring, the mixture was filtered, and the filter cake was washed with petroleum ether (100 mL). The filter cake was dried under reduced pressure to give intermediate B6-5(1.9g, 80%) as a brown solid.1H NMR(400MHz,DMSO-d6)δ11.77(s,1H),8.60(s,1H),8.05(d,J=6.8Hz,1H),6.35(d,J=8.0Hz,1H),5.82-5.64(m,1H),5.28-5.09(m,2H),2.81-2.63(m,2H),1.52(s,3H).
6) Synthesis of intermediate B6-6
Intermediate B6-5(500mg,1.31mmol) was dissolved in acetic acid (15mL), zinc powder (855mg,13.1mmol) was added, and the mixture was heated to 110 ℃ and stirred for 1 hour. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol ═ 100:1) to give intermediate B6-6(200mg, 50%) as a yellow solid product.1H NMR(400MHz,DMSO-d6)δ11.60(br s,1H),8.30(s,1H),8.02(d,J=7.6Hz,1H),7.74(s,1H),6.79-6.59(m,1H),6.14(d,J=7.2Hz,1H),5.98-5.79(m,1H),5.30-5.01(m,3H),2.84-2.58(m,2H),1.90(s,3H).
7) Synthesis of intermediate B6-7
Intermediate B6-6(168mg,0.55mmol) was dissolved in N, N-dimethylformamide (1mL) and dichloromethane (20mL), dessimutan's oxidant (260mg,0.61mmol) was added and the mixture was stirred at room temperature for 4 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol: 30:1) to give intermediate B6-7(70mg, 42%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ12.41(s,1H),8.62(s,1H),8.20-8.12(m,2H),6.27(d,J=8.0Hz,1H),5.80-5.71(m,1H),5.29-5.11(m,2H),2.77-2.66(m,2H),1.50(s,3H).
8) Synthesis of end product B6
B6-7(70mg,0.23mmol) was dissolved in phosphorus oxychloride (6mL) and stirred at 110 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in isopropanol (6mL) and 5-aminobenzothiazole (42mg,0.28mmol) was added. The reaction was stirred at 90 ℃ for 4 hours. After cooling to room temperature, filtration was carried out and the filter cake was washed with isopropanol (6 mL). The filter cake was dried to give final product B6(65mg, 65%) as a yellow solid.
According to this synthesis method, a heterocyclic compound B7 was synthesized using different substrates.
Example 7
A heterocyclic compound B8 synthesized by the method comprising:
Figure BDA0002354170460000221
1) synthesis of end product B8
Methyltriphenylphosphonium bromide (260mg,0.73mmol) was dissolved in tetrahydrofuran (5mL), and after stirring at-70 ℃ for 15 minutes under a nitrogen atmosphere, n-butyllithium (0.33mL,0.83mmol) was added dropwise to the reaction system. After the dropwise addition, the reaction solution was transferred to 0 ℃ and stirred for 1.5 hours, and then the solution became clear. After stirring at-70 ℃ for 20 minutes, a suspension of the final product B4(100mg,0.22mmol) in tetrahydrofuran (5mL) was added dropwise to the reaction system. After the addition was completed, the reaction solution was stirred at room temperature overnight, quenched with water (1mL), spun-dried, and the residue was purified by column chromatography (dichloromethane: methanol ═ 20:1) to give a crude product. The crude product was purified by silica gel prep. (dichloromethane: methanol ═ 30:1) to give final product B8(10mg, 11%).
Example 8
A heterocyclic compound B9 synthesized by the method comprising:
Figure BDA0002354170460000222
1) synthesis of end product B9
Final product B4(50mg,0.11mmol) was dissolved in methanol (3mL) and dichloromethane (5mL), sodium borohydride (11mg,0.28mmol) was added, and the mixture was stirred at room temperature for 30 min. The reaction was quenched by the addition of saturated ammonium chloride solution (1mL), spun dry, and the residue was purified by column chromatography (dichloromethane: methanol ═ 20:1) to give final product B9(8mg, 16%).
Example 9
A heterocyclic compound B10 synthesized by the method comprising:
Figure BDA0002354170460000231
1) synthesis of end product B10
Final product B4(60mg,0.13mmol) was dissolved in tetrahydrofuran (5mL), and after stirring at 0 ℃ for 0.5 hour under a nitrogen atmosphere, methylmagnesium bromide (0.4mL,0.40mmol) was added dropwise to the reaction system. After stirring for 1.0 h, the reaction was quenched with water (1mL), spun dry, and the residue was purified by column chromatography (dichloromethane: methanol 50:1) to give final product B10(30mg, 52%).
According to this synthesis method, a heterocyclic compound B11 was synthesized using different substrates.
Example 10
A heterocyclic compound B12 synthesized by the method comprising:
Figure BDA0002354170460000232
1) synthesis of end product B12
B4-7(49mg,0.1mmol), cyclopropylboronic acid (17mg,0.2mmol) and potassium phosphate trihydrate (83mg,0.35mmol) were dissolved in a mixed solution of toluene (3mL) and water (0.2mL), potassium acetate (2mg,0.005mmol) and cyclohexylphosphine (3mg,0.01mmol) were added, and the mixture was heated to 100 ℃ under nitrogen and stirred overnight. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol 100:1) and purified by combi-flash C18 to give intermediate B12(4mg, 9%) as a yellow solid.
Example 11
A heterocyclic compound B15 synthesized by the method comprising:
Figure BDA0002354170460000241
1) synthesis of intermediate B15-1
Intermediate B4-6(6g,16.9mmol) was dissolved in isopropanol (300mL) and water (27mL), triethylamine (27mL) and 10% wet palladium on carbon (1g) were added, the atmosphere of hydrogen was replaced, and the reaction was stirred at room temperature for 3 hours. Celite was filtered and the filter cake was washed once with methanol (50 mL). The filtrate was concentrated under reduced pressure, and the residue was subjected to column chromatography (dichloromethane: methanol ═ 50:1) to give intermediate B15-1(4g, 86%).1H NMR(400MHz,DMSO-d6)δ12.42(s,1H),8.62(s,1H),8.20-8.10(m,2H),6.26(d,J=7.2Hz,1H),1.53(s,6H).
2) Synthesis of intermediate B15-2
Intermediate B15-1(1g,3.6mmol) was dissolved in phosphorus oxychloride (10mL) and stirred at 110 ℃ for 0.5 h. The reaction solution was concentrated under reduced pressure to obtain intermediate B15-2 (crude)Product 1.8g,3.6 mmol).1H NMR(400MHz,DMSO-d6)δ9.16(d,J=4.4Hz,1H),8.97(s,1H),8.72(s,1H),8.15(d,J=4.0Hz,1H),1.57(s,6H).
3) Synthesis of end product B15
Intermediate B15-2 (crude 90mg,0.18mmol) was dissolved in isopropanol (2mL), 2, 4-difluoro-5-trifluoromethoxyaniline (31mg,0.22mmol) was added and the reaction was allowed to proceed at 95 ℃ for 1 hour. After cooling to room temperature, a solid precipitated, filtered, and the filter cake was dissolved with methanol (3mL) and dichloromethane (3 mL). The solution was adjusted to PH 8 with saturated sodium bicarbonate solution (2mL), spun dry and the residue was purified by column chromatography (dichloromethane: methanol 20:1) to give final product B15(10mg, 14%).
According to the synthesis method, heterocyclic compounds B13, B14, B16, B17, B18 and B19 are synthesized by using different substrates.
Example 12
A heterocyclic compound B21 synthesized by the method comprising:
Figure BDA0002354170460000251
1) synthesis of intermediate B21-1
Intermediate B4-1(3.565g,15.7mmol) was dissolved in N, N-dimethylformamide (35mL), allyl bromide (7.59g,62.7mmol) and potassium carbonate (8.66g,62.7mmol) were added, and the mixture was stirred at room temperature overnight. Quench with 3N hydrochloric acid (20mL) and extract with ethyl acetate (50 mL. times.2). The organic phases were combined and washed with saturated sodium chloride solution (20mL) and the organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 10:1) to give intermediate B21-1(2.09g, 43%) as a yellow solid.1H NMR(400MHz,CDCl3)δ8.79-8.69(m,2H),8.20(d,J=8.4Hz,1H),5.83-5.71(m,2H),5.27-5.18(m,4H),2.91-2.78(m,4H).
2) Synthesis of intermediate B21-2
Grubbs-1st catalyst (212mg,0.258mmol) was dissolved in dichloromethane (200mL), intermediate B21-1(2.09g,6.8mmol) was slowly added dropwise in dichloromethane (50mL) and stirred at room temperature overnight. Concentrating the reaction solution under reduced pressure, and passing the residue through silica gelColumn chromatography (petroleum ether: ethyl acetate: 10:1) gave intermediate B21-2(1.88g, 99%) as a yellow solid.1H NMR(400MHz,CDCl3)δ8.83(s,1H),8.76(d,J=8.8Hz,1H),8.24(d,J=8.4Hz,1H),5.77(s,2H),3.46(d,J=17.2Hz,2H),3.01(d,J=17.2Hz,2H).
3) Synthesis of intermediate B21-3
Intermediate B21-2(1.88g,6.73mmol) was dissolved in isopropanol (20mL), 10% content wet palladium on charcoal (500mg) and concentrated hydrochloric acid (2mL) were added, and the atmosphere of hydrogen was replaced and stirred overnight. The reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was extracted with saturated sodium carbonate solution (10mL) and ethyl acetate (30mL × 6). The organic phases were combined and concentrated under reduced pressure to give intermediate B21-3 (crude 1.66g, 99%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ7.78(d,J=6.4Hz,1H),7.13(d,J=9.2Hz,1H),6.97(s,1H),6.56(s,2H),5.73(s,2H),3.18(d,J=17.6Hz,2H),2.82(d,J=17.6Hz,2H).
4) Synthesis of intermediate B21-4
Intermediate B21-3(660mg,3.07mmol) was dissolved in a mixed solvent of methanol (15mL), tetrahydrofuran (15mL) and water (6mL), oxone (1.89g,3.07mmol) was added thereto at room temperature, and sodium bromide (316mg,3.07mmol) was slowly added dropwise to 3mL of an aqueous solution thereof, followed by stirring at room temperature for 2 hours. Saturated sodium carbonate solution was added to adjust pH to 8 and extracted by adding dichloromethane (50mL x 3). The organic phases were combined and the organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane: methanol ═ 200:1) to give intermediate B21-4(450mg, 45%) as a yellow solid product.1H NMR(400MHz,CDCl3)δ7.75(d,J=8.4Hz,1H),7.19-7.13(m,1H),5.72(s,2H),4.95(s,2H),3.39(d,J=18.0Hz,2H),2.95(d,J=17.6Hz,2H).LCMS(ESI/APCI)m/z:344.6[M+NH4]+.
5) Synthesis of intermediate B21-5
Intermediate B21-4(450mg,1.37mmol) was dissolved in ethanol (6mL), 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (357mg,1.92mmol) was added, and the mixture was stirred at room temperature for 7 hours. The reaction solution was filtered, and the filter cake was washed with ethanol (6 mL). The filter cake was dried to give intermediate B21-5(573mg, 87%) as a white solid.1H NMR(400MHz,CDCl3)δ12.04(d,J=13.2Hz,1H),8.70(d,J=13.6Hz,1H),8.13-8.05(m,1H),7.92-7.86(m,1H),5.75(s,2H),3.42(d,J=17.6Hz,2H),3.00(d,J=17.2Hz,2H),1.57(s,6H).
6) Synthesis of intermediate B21-6
Diphenyl ether (10mL) was heated to 220 deg.C and a solution of intermediate B21-5(425mg,0.88mmol) in 3mL of methylene chloride was added dropwise, stirring for 0.5 h with incubation. The reaction mixture was cooled to room temperature, petroleum ether (10mL) was added with stirring, the mixture was filtered, and the filter cake was washed with petroleum ether (30 mL). The filter cake was dried under reduced pressure to give intermediate RR-164-6 (crude 330mg) as a gray solid product.1H NMR(400MHz,DMSO-d6)δ11.77(s,1H),8.62(s,1H),8.14-7.99(m,1H),6.44-6.25(m,1H),5.77(s,2H),3.24(d,J=17.6Hz,2H),2.94(d,J=17.6Hz,2H).
7) Synthesis of end product B21
B21-6(330mg,0.87mmol) was dissolved in phosphorus oxychloride (6mL) and stirred at 110 ℃ overnight. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in isopropanol (15mL) and 5-aminobenzothiazole (201mg,1.34mmol) was added. The reaction was stirred at 90 ℃ overnight. After cooling to room temperature, filtration was carried out and the filter cake was washed with isopropanol (6 mL). The filter cake was dried to give final product B21(260mg, 64%) as a yellow solid.
Example 13
A heterocyclic compound B22 synthesized by the method comprising:
Figure BDA0002354170460000271
1) synthesis of end product B22
Intermediate B21(75mg,0.16mmol) was dissolved in isopropanol (15mL) and water (1mL), 10% content of wet palladium on carbon (300mg) and triethylamine (0.5mL) were added, the atmosphere of hydrogen was replaced, and the reaction was stirred at 90 ℃ overnight. The reaction was cooled to room temperature, filtered, and the filter cake was washed with methanol (30 mL). The filtrate was concentrated under reduced pressure and the residue was purified by thin layer preparative plate chromatography (dichloromethane: methanol 20:1) and the resulting product slurried with methanol (2mL) for 6 h, filtered, the filter cake washed with ether (15mL) and dried under reduced pressure to give the final product B22 as a yellow solid (12mg, 17%).
Example 14
A heterocyclic compound B23 synthesized by the method comprising:
Figure BDA0002354170460000272
1) synthesis of intermediate B23-1
B2-7(33mg,0.0743mmol), phenylboronic acid (14mg,0.111mmol) and potassium carbonate (31mg,0.223mmol) were dissolved in a mixed solution of dioxane (6mL) and water (0.5mL), and Pd (dppf) Cl was added2(6mg,0.00743mmol), and stirred at 110 ℃ for 6 hours under nitrogen. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol 100:1) to give intermediate B23-1(15mg, 46%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ9.61(s,1H),9.46(s,1H),9.12(s,1H),8.57(d,J=5.2Hz,1H),8.24(d,J=8.8Hz,1H),8.08(s,1H),7.90(s,1H),7.81-7.76(m,2H),7.74(s,1H),7.69-7.62(m,3H),7.57(d,J=8.8Hz,1H),7.11(d,J=5.2Hz,1H).LCMS(ESI/APCI)m/z:441.6[M+H]+.
2) Synthesis of final product B23:
intermediate B23-1(60mg,0.14mmol) was dissolved in methanol (10mL), 10% Pd/C (100mg) was added, and the reaction was carried out overnight at room temperature under a hydrogen atmosphere. Filtering, and spin-drying the filtrate. The residue was purified on preparative silica gel plates (dichloromethane: methanol ═ 100:3) to give the final product B23(17mg, 27%) as a yellow solid.
According to this synthesis method, a heterocyclic compound B24 was synthesized using different substrates.
Example 15
A heterocyclic compound B25 synthesized by the method comprising:
Figure BDA0002354170460000281
1) synthesis of intermediate B25-1
3, 4-difluoronitrobenzene (3.18g,20mmol) was dissolved in N, N-dimethylformamide (30mL) and placed under an ice-water bath. Reacting mercaptoethaneMethyl ester (2.33g,22mmol) was added dropwise to the reaction and stirring was continued overnight. 200mL of water was added and a white solid precipitated, which was filtered and washed twice with water (20 mL. times.2) to give intermediate B25-1(3.5g, 71%) as a white particulate solid.1H NMR(400MHz,CDCl3)δ8.02(d,J=8.4Hz,1H),7.93(d,J=9.6Hz,1H),7.50(t,J=8.0Hz,1H),3.79(s,2H),3.76(s,3H).
2) Synthesis of intermediate B25-2
Intermediate B25-1(3.5mg,14mmol) was dissolved in 50mL of ethanol, and iron powder (3.2mg,57mmol), 10mL of water, and ammonium chloride (3.8g,71mmol) were added, and the mixture was heated to 75 ℃ for reaction for 3 hours. The mixture was filtered while hot, washed three times with dichloromethane (100mL x 3), the filtrate was washed once with saturated aqueous sodium chloride (200mL), the organic phase was concentrated under reduced pressure, and the ether was slurried to give intermediate B25-2(2.4g, 78%) as a white solid.1H NMR(400MHz,CDCl3)δ7.31-7.27(m,1H),6.48-6.30(m,2H),3.89(s,2H),3.67(s,3H),3.44(s,2H).
3) Synthesis of intermediate B25-3
B25-2(2.0g,11.1mmol) was dissolved in 14mL of acetic acid, placed under an ice-water bath, acetic anhydride (1.4g,13mmol) was added dropwise to the reaction system, and stirring was continued for 4 hours. 150mL of water was added to precipitate a white solid, which was filtered to obtain intermediate B25-3(2.0g, loud).
4) Synthesis of intermediate B25-4
B25-3(1.8g,7.0mmol) was dissolved in 130mL of dichloromethane, placed under an ice-water bath, and m-chloroperoxybenzoic acid (85%) (3.8g,19mmol) was added portionwise to the reaction and stirring was continued overnight. The reaction was quenched with saturated aqueous sodium sulfite, washed once with saturated aqueous sodium bicarbonate (60mL), concentrated under reduced pressure, slurried with ethyl acetate (15mL), and filtered to give intermediate B25-4 as a white solid (1.2g, 60%).1H NMR(400MHz,CDCl3)δ7.90-7.77(m,2H),7.65(s,1H),7.19(d,J=8.4Hz,1H),4.28(s,2H),3.70(s,3H),2.23(s,3H).
5) Synthesis of intermediate B25-5
Intermediate B25-4(500mg,1.7mmol) was dissolved in 8mL of N, N-dimethylformamide, and 1, 2-dibromoethane (632mg,6.8mmol) and potassium carbonate (580mg,4.2mmol) were added thereto, followed by reaction at 60 ℃ for 4 hours. Cooling to room temperature, adding BEthyl acetate (100mL) was diluted, washed three times with saturated aqueous sodium chloride (30mL × 3), the organic phase was concentrated under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate 1:1) to give B25-5(880mg, 71%) as a yellow oil.1H NMR(400MHz,CDCl3)δ8.75(s,1H),7.80-7.65(m,2H),7.34(d,J=9.2Hz,1H),3.71(s,3H),2.19(s,3H),1.64-1.62(m,4H).
6) Synthesis of intermediate B25-6
Intermediate B25-5(365mg,1.2mmol) was dissolved in 40mL of methanol, and sodium borohydride (287mg,7.6mmol) was added in portions in an ice-water bath and stirred at room temperature overnight. Saturated aqueous sodium bicarbonate (30mL) was added, extracted twice with dichloromethane (40mL x 2), the organic phase was separated, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give B25-6(200mg, 73%) as a yellow oily liquid.1H NMR(400MHz,CDCl3)δ7.90-7.75(m,2H),7.60(s,1H),7.20(d,J=8.4Hz,1H),3.67(d,J=6.0Hz,2H),2.60(t,J=6.4Hz,2H),2.23(s,3H),1.78-1.67(m,2H),1.15-1.05(m,2H).
7) Synthesis of intermediate B25-7
Intermediate B25-6(200mg,0.70mmol) was dissolved in 4mL of dimethyl sulfoxide, potassium tert-butoxide (94mg,0.84mmol) was added, and the reaction was allowed to warm to 60 ℃ for 3 hours. Cooled to room temperature, diluted with ethyl acetate (50mL), washed twice with saturated aqueous sodium chloride (30mL × 2), the organic phase was separated, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a yellow oil. The oil was dissolved in 6mL of absolute ethanol, 3mL of concentrated HCl was added, and the reaction was allowed to warm to 80 ℃ overnight. The solvent was dried, diluted with ethyl acetate (100mL), adjusted to pH 7-8 with saturated aqueous sodium bicarbonate, the organic phase was separated, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate 1:1) to give B25-7(130mg, 86%) as a white solid.1H NMR(400MHz,CDCl3)δ7.52(d,J=8.4Hz,1H),6.36(d,J=8.4Hz,1H),6.15(s,1H),4,56(s,2H),4.05(s,2H),1.65(s,2H),1.14(s,2H).
8) Synthesis of intermediate B25-8
Intermediate B25-7(130mg,0.60mmol) was dissolved in 10mL of ethanol, and 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (134mg,0.72mmol) was added thereto, followed by stirring at room temperature for 12 hours. The reaction solution is passed throughFiltering to obtain white solid. The diphenyl ether was warmed to 240 ℃ and the white solid was added portionwise. The reaction was carried out for 10 minutes. The temperature was naturally returned to room temperature, filtered, and the filter cake was purified by column chromatography (dichloromethane: acetone ═ 3:1) to give B25-8(20mg, 8.6%) as a white solid.1H NMR(400MHz,DMSO-d6)δ11.82(s,1H),8.37(s,1H),8.00-7.85(m,1H),7.10(s,1H),6.01(d,J=7.6Hz,1H),4.79(s,2H),1.53-1.35(m,4H).
9) Synthesis of end product B25
B25-8(20mg,0.07mmol) was dissolved in phosphorus oxychloride (5mL) and stirred at 100 ℃ for 30 min. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in isopropanol (6mL), to which was added 5-aminobenzothiazole (13mg,0.08 mmol). The reaction was stirred at 90 ℃ for 2 hours. After cooling to room temperature, filtration was carried out, 40mL of dichloromethane was added, the pH was adjusted to 7-8 with a saturated aqueous solution of sodium hydrogencarbonate, the organic phase was dried over anhydrous sodium sulfate, filtration was carried out, the solvent was dried by spinning, and slurried with ethyl acetate (4mL) to give B25(20mg, 70%) as a yellow solid.
According to this synthesis method, a heterocyclic compound B26 was synthesized using different substrates.
Example 16
A heterocyclic compound B27 synthesized by the method comprising:
Figure BDA0002354170460000301
1) synthesis of intermediate B27-1
Intermediate B2-1(5.0g,19.4mmol) was dissolved in N, N-dimethylformamide (50mL), sodium bicarbonate (3.3g, 39mmol) was added followed by 2-dicyclohexylphosphonium-2 ',4',6' -triisopropylbiphenyl (954mg, 2.0mmol), palladium acetate (220mg, 1.0mmol), stirred at 120 ℃ for 4 hours under nitrogen, the reaction was concentrated, and the residue was purified by silica gel column (pure dichloromethane) to give intermediate B27-1 as a pale yellow solid (3.6g, 71%).1H NMR(400MHz,CDCl3)δ8.89(s,1H),8.28(d,J=8.0Hz,1H),8.07–7.87(m,3H),6.58(d,J=16.0Hz,1H),3.87(s,3H).
2) Synthesis of intermediate B27-2
Intermediate B27-1(3.6g, 13.7mmol) in tetrahydrofuran (100mL), slowly adding sodium borohydride (4.2g, 109.6mmol) under ice-bath, returning to room temperature after the addition is complete, stirring overnight, subsequently adding aluminum trichloride (3.6g, 27.4mmol) under ice-bath, refluxing and continuing to stir for 2 days; the reaction was placed in an ice bath and saturated ammonium chloride was slowly added to quench the reaction, the solid was filtered, the filter cake was washed with ethyl acetate (50mL), saturated aqueous sodium chloride solution was added to the filtrate, the organic phases were extracted with ethyl acetate (50mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated and purified on a silica gel column (dichloromethane: methanol 50:1) to give intermediate B27-2(1.8g, 64%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ8.07(s,1H),8.02(d,J=8.4Hz,1H),7.47(d,J=8.4Hz,1H),5.10(s,1H),3.73–3.53(m,4H),3.44–3.37(m,2H).
3) Synthesis of intermediate B27-3
Intermediate B27-2(1.8g, 8.7mmol) was dissolved in dichloromethane (50mL), 85% m-chloroperoxybenzoic acid (4.4g, 21.8mmol) was added, the mixture was stirred at room temperature overnight, a white solid precipitated, the solid was filtered, the filtrate was quenched with saturated sodium sulfite, saturated sodium bicarbonate was then added to adjust the pH to 7, the organic phases were extracted with dichloromethane (40mL x 3), the organic phases were combined, concentrated, and purified on silica gel column (dichloromethane: methanol ═ 50:1) to give intermediate B27-3(800mg, 34%) as a pale yellow solid.1H NMR(400MHz,CDCl3)δ8.43(d,J=7.6Hz,1H),8.31(s,1H),7.90(d,J=8.0Hz,1H),6.66(s,1H),3.90–3.76(m,2H),2.84(t,J=6.8Hz,2H),2.04–1.91(m,2H).
4) Synthesis of intermediate B27-4
Intermediate B27-3(800mg, 3.0mmol) was dissolved in methanol (40mL), cesium carbonate (1.47g, 4.5mmol) was added, the solution quickly turned dark yellow, stirring was continued at room temperature for 2 hours, the reaction was concentrated, and purification was performed on silica gel column (petroleum ether: ethyl acetate: 5:1) to give intermediate B27-4(200mg, 25%) as a pale yellow solid.1H NMR(400MHz,CDCl3)δ8.45–8.32(m,2H),7.90(d,J=8.4Hz,1H),4.30–4.12(m,2H),3.70-3.52(m,2H),2.67–2.50(m,1H),2.42–2.27(m,1H),2.25–2.07(m,2H).
5) Synthesis of intermediate B27-5
B27-4(200mg, 0.74mmol) was dissolved in ethanol (10mL), iron powder (166mg, 2.96mmol) was added, the temperature was gradually raised to 80 ℃, ammonium chloride (157mg, 2.96mmol) was dissolved in water (5mL) and added to the reaction mixture, and the mixture was stirred at 80 ℃ for 1 hour. After cooling to room temperature, the reaction mixture was diluted with dichloromethane (20mL), the iron powder was removed by filtration with celite, the filter cake was washed with dichloromethane, water (20mL) was added to the liquid phase, the liquid phase was separated, the organic phase was further extracted with dichloromethane (20mL × 2), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column (petroleum ether: ethyl acetate ═ 1:1) to obtain intermediate B27-5(120mg, 68%) as a yellow solid.1H NMR(400MHz,CDCl3)δ7.47(d,J=8.0Hz,1H),6.71(d,J=8.0Hz,1H),6.66(s,1H),4.18(s,2H),4.15–4.04(m,2H),3.57-3.34(m,2H),2.56-2.43(m,1H),2.31–2.14(m,1H),2.16–2.01(m,2H).
6) Synthesis of intermediate B27-6
Dissolving B27-5(120mg, 0.50mmol) in ethanol (5mL), slowly adding 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (140mg,0.75mmol), stirring at room temperature for 20 min, precipitating a large amount of solid, filtering, washing the filter cake with a small amount of ethanol, and drying to obtain a light yellow solid intermediate B27-6(140mg, 72%)1H NMR(400MHz,CDCl3)δ11.38(d,J=13.6Hz,1H),8.70(d,J=13.6Hz,1H),7.78(d,J=8.0Hz,1H),7.40(s,1H),7.37(d,J=8.8Hz,1H),4.30–4.08(m,2H),3.60-3.45(m,2H),2.63–2.52(m,1H),2.37–2.26(m,1H),2.24–2.02(m,2H),1.77(s,6H).
7) Synthesis of intermediate B27-7
Diphenyl ether (10mL) was added to the round bottom flask, heated to 240 ℃ and stirred for 10 minutes while maintaining the temperature to remove the solvent water, B27-6(140mg,0.36mmol) was added to the reaction in portions, and the mixture was stirred for 5 minutes at 240 ℃. Cooling to room temperature precipitated a solid which was filtered and the filter cake washed with ether and dried to give intermediate B27-7 as a pale yellow solid (52mg, 50%).1H NMR(400MHz,DMSO-d6)δ12.08(s,1H),8.29(s,1H),8.12-7.96(m,1H),7.73(s,1H),6.14(d,J=7.2Hz,1H),4.13–3.93(m,3H),3.97(d,J=13.6Hz,1H),3.61(d,J=13.2Hz,1H),2.46–2.37(m,1H),2.23–2.11(m,3H).
8) Synthesis of end product B27
B27-7(20mg, 0.07mmol) was dissolved in phosphorus oxychloride (2mL) and stirred at 110 ℃ for 2 h. Concentration to remove phosphorus oxychloride, dissolving the solid residue in ethanol (2mL), adding 5-aminobenzothiazole (13mg,0.08mmol) and one drop of concentrated hydrochloric acid, stirring the mixture at 80 ℃ for 2 hours, cooling to room temperature, precipitating a solid, filtering, pulping the filter cake with ether, and drying to obtain a yellow solid end product B27(15mg, 51%).
Example 17
A heterocyclic compound B28 synthesized by the method comprising:
Figure BDA0002354170460000331
1) synthesis of intermediate B28-1
Methyl o-fluorophenylacetate (10.3g, 61.3mmol) was dissolved in anhydrous tetrahydrofuran (200mL), dimethyl carbonate (16.6g, 184mmol) was added, 60% sodium hydrogen (9.8g, 245mmol) was slowly added under ice bath conditions, followed by stirring overnight at 70 ℃ under nitrogen atmosphere, TLC showed reaction completion, saturated ammonium chloride was quenched under ice bath, the organic phases were extracted with dichloromethane (100mL x 3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated, and purified on silica gel column (petroleum ether: ethyl acetate ═ 20:1) to give pale yellow liquid intermediate B28-1(11.2g, 81%).1H NMR(400MHz,CDCl3)δ7.50–7.39(m,1H),7.37–7.27(m,1H),7.20–7.12(m,1H),7.12–7.01(m,1H),5.01(s,1H),3.77(s,6H).
2) Synthesis of intermediate B28-2
Intermediate B28-1(11.2g, 49.6mmol) was dissolved in N, N-dimethylformamide (130mL), cesium carbonate (32.3g, 99.2mmol) was added followed by the slow dropwise addition of chloromethyl benzyl ether (11.7g, 74.4mmol), the mixture was stirred at room temperature for 1 hour, the solvent was concentrated, water (400mL) was added, the organic phase was extracted with ethyl acetate (200mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified on silica gel column (petroleum ether: ethyl acetate ═ 20:1) to give intermediate B28-2(12.6g, 73%) as a colorless transparent liquid.1H NMR(400MHz,CDCl3)δ7.51–7.41(m,1H),7.35–7.27(m,3H),7.25–7.18(m,3H),7.16–7.09(m,1H),7.07–6.98(m,1H),4.58(s,2H),4.19(s,2H),3.78(s,6H).
3) Synthesis of intermediate B28-3
Intermediate B28-2(8.6g, 24.9mmol) was dissolved in tetrahydrofuran (100mL), lithium aluminum hydride (3.8g, 99.6mmol) was added slowly under ice bath, then gradually returned to room temperature and stirring was continued for 2 hours; water (3.8mL), a 15% aqueous solution of sodium hydroxide (3.8mL), and water (11.4mL) were sequentially added under ice-cooling to precipitate a granular solid, the solid was filtered, the cake was washed with a large amount of methanol, the filtrate was concentrated, and the product was purified with a silica gel column (petroleum ether: ethyl acetate ═ 2:1) to obtain intermediate B28-3(1.8g, 25%) as a colorless transparent liquid.1H NMR(400MHz,CDCl3)δ7.47–7.38(m,1H),7.38–7.29(m,3H),7.29–7.20(m,3H),7.17–7.08(m,1H),7.07–6.96(m,1H),4.53(s,2H),4.16(d,J=11.2Hz,2H),4.07(d,J=10.4Hz,2H),3.94(s,2H),2.67(s,2H).
4) Synthesis of intermediate B28-4
Dissolving intermediate B28-3(1.8g, 6.2mmol) in tetrahydrofuran (20mL), adding 2.5mmol/mL n-butyllithium (2.5mL, 6.2mmol) dropwise under ice bath condition under nitrogen protection, after 10 min, slowly adding p-toluenesulfonyl chloride (1.2g, 6.2mmol) in tetrahydrofuran (5mL), stirring for ten min, adding 2.5mmol/mL n-butyllithium (2.5mL, 6.2mmol) again, heating to 55 deg.C, stirring overnight, cooling to room temperature, quenching reaction with saturated ammonium chloride, extracting organic phase with ethyl acetate (30mL x 3), combining organic phases, drying with anhydrous sodium sulfate, concentrating, purifying with silica gel column (petroleum ether: ethyl acetate: 20:1) to obtain colorless transparent liquid B28-4(1.1g, 61%).1H NMR(400MHz,CDCl3)δ7.32–7.21(m,4H),7.21–7.15(m,2H),7.15–7.08(m,1H),7.08–6.96(m,2H),5.00(d,J=5.6Hz,2H),4.79(d,J=5.2Hz,2H),4.52(s,2H),3.88(s,2H).
5) Synthesis of intermediate B28-5
Dissolving intermediate B28-4(1.1g, 4.0mmol) in methanol (20mL), adding 10% palladium on carbon (21mg, 0.2mmol), stirring the mixture under hydrogen atmosphere for 12 hr, filtering to remove palladium on carbon, washing the solid with dichloromethane, and concentrating the filtrate to obtain colorless transparent liquid B28-5(650mg, 89%) and was directly administered to the next step.1H NMR(400MHz,CDCl3)δ7.32–7.27(m,1H),7.20–7.12(m,1H),7.10–6.96(m,2H),5.01(d,J=6.0Hz,2H),4.76(d,J=6.0Hz,2H),4.10(s,2H).
6) Synthesis of intermediate B28-6
Intermediate B28-5(420mg, 2.3mmol), triphenylphosphine (1.2g, 4.6g) were dissolved in carbon tetrachloride (20mL) and the mixture was stirred at 90 ℃ for 24 h; the solvent was concentrated and purified by silica gel column (petroleum ether: ethyl acetate: 40:1) to obtain intermediate B28-6(440mg, 96%) as a colorless transparent liquid.1H NMR(400MHz,CDCl3)δ7.34–7.27(m,1H),7.23-7.10(m,1H),7.10–6.99(m,2H),5.02(d,J=6.4Hz,2H),4.74(d,J=6.4Hz,2H),4.11(s,2H).
7) Synthesis of intermediate B28-7
Dissolving intermediate B28-6(440mg, 2.2mmol) in concentrated sulfuric acid (5mL) under ice bath condition, dropwise adding concentrated nitric acid (1mL), returning to room temperature and continuing stirring for 5 minutes; the reaction was poured into cold water, the organic phases were extracted with ethyl acetate (10mL x 3), the organic phases were combined, dried over anhydrous sodium sulfate and concentrated to give crude pale yellow product B28-7 (loud 520mg) which was directly put to the next step.1H NMR(400MHz,CDCl3)δ8.30–8.19(m,1H),8.03–7.95(m,1H),7.24–7.18(m,1H),5.03(d,J=5.2Hz,2H),4.75(d,J=5.6Hz,2H),4.16(s,2H).
7) Synthesis of intermediate B28-8
The crude product B28-7 (2.1 mmol) was dissolved in dimethylsulfoxide (30mL), sodium sulfide nonahydrate (768mg, 3.2mmol) was added, the mixture was stirred at room temperature for 3 hours, water (80mL) was added to the reaction solution, ethyl acetate (20mL × 2) was added to extract the organic phase, the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column (petroleum ether: ethyl acetate ═ 10:1) to obtain intermediate B28-8(240mg, 51%) as a yellow solid.1H NMR(400MHz,CDCl3)δ8.40(s,1H),8.07(d,J=8.8Hz,1H),7.28(d,J=8.8Hz,1H),4.85(d,J=6.4Hz,2H),4.82(d,J=6.4Hz,2H),3.78(s,2H).
8) Synthesis of intermediate B28-9
Intermediate B28-8(240mg, 1.08mmol) was dissolved in dichloromethane (15mL) and 85% m-chloroperoxy was addedBenzoic acid (543g, 2.69mmol), the mixture was stirred at room temperature overnight, a white solid precipitated, the solid was filtered, the filtrate was quenched with saturated sodium sulfite, followed by addition of saturated sodium bicarbonate to adjust the pH to 7, dichloromethane (20mL × 3) was extracted from the organic phases, the organic phases were combined, concentrated, and purified on silica gel column (dichloromethane: methanol ═ 50:1) to give intermediate B28-9(220mg, 80%) as a white solid.1H NMR(400MHz,CDCl3)δ8.85(s,1H),8.44(d,J=7.6Hz,1H),7.92(d,J=8.4Hz,1H),5.04(d,J=6.8Hz,2H),4.95(d,J=6.8Hz,2H),3.91(s,2H).
9) Synthesis of intermediate B28-10
B28-9(220mg, 0.86mmol) was dissolved in ethanol (8mL), iron powder (193mg, 3.44mmol) was added, the temperature was gradually raised to 80 ℃ and ammonium chloride (182mg, 3.44mmol) was dissolved in water (3mL) and added to the reaction mixture, and the mixture was stirred at 80 ℃ for 2 hours. Cooling to room temperature, adding dichloromethane (20mL) to dilute the reaction solution, filtering with diatomite to remove iron powder, washing the filter cake with dichloromethane, adding water (20mL) to the solution phase, separating, extracting the organic phase with dichloromethane (20mL × 2), combining the organic phases, drying with anhydrous sodium sulfate, concentrating to obtain crude product B28-10(200mg), and directly feeding into the next step.1H NMR(400MHz,CDCl3)δ7.47(d,J=8.4Hz,1H),7.10(s,1H),6.74(d,J=8.0Hz,1H),4.95(d,J=6.4Hz,2H),4.87(d,J=6.0Hz,2H),4.30(s,2H),3.75(s,2H).
10) Synthesis of intermediate B28-11
B28-10 (crede 200mg, 0.8mmol) was dissolved in ethanol (4mL), followed by slow addition of 5- (methoxymethylene) -2, 2-dimethyl-1, 3-dioxahexane-4, 6-dione (223mg,1.2mmol), stirring at room temperature for 20 minutes, precipitation of a large amount of solid, filtration, washing of the filter cake with a small amount of ethanol, and drying to give intermediate B28-11(240mg, 79%) as a pale yellow solid.1H NMR(400MHz,CDCl3)δ11.47(d,J=12.8Hz,1H),8.76(d,J=13.6Hz,1H),7.87–7.74(m,2H),7.43(d,J=6.8Hz,1H),5.03(d,J=6.0Hz,2H),4.89(d,J=6.0Hz,2H),3.84(s,2H),1.79(s,6H).
11) Synthesis of intermediate B28-12
Diphenyl ether (6mL) was added to a round bottom flask, heated to 240 ℃ and stirred for 10 minutes while maintaining temperature to remove solvent waterSeparately, B28-11(240mg,0.63mmol) was added portionwise to the reaction solution, and the mixture was stirred at 240 ℃ for 5 minutes. Cooling to room temperature precipitated a solid which was filtered and the filter cake washed with ether and dried to give intermediate B28-12 as a pale yellow solid (110mg, 63%).1H NMR(400MHz,DMSO-d6)δ12.34(s,1H),8.27(s,1H),8.17(s,1H),8.05(d,J=7.6Hz,1H),6.15(d,J=7.2Hz,1H),4.96(d,J=6.0Hz,2H),4.77(d,J=6.4Hz,2H),4.13(s,2H).
12) Synthesis of intermediate B28-13
Intermediate B28-12(30mg, 0.11mmol) was dissolved in anhydrous dichloromethane (4mL), pyridine (0.1mL) was added followed by dropwise addition of triflic anhydride (30 drops), the reaction was complete by TLC, and the solvent was concentrated to give crude product B28-13 which was directly fed to the next step. LC-MS (M/z) 409.6[ M + H]+.
13) Synthesis of end product B28
The crude product B28-13(0.11mmol), Pd2(dba)3(18mg, 0.02), xanthphos (12mg, 0.02mmol), cesium carbonate (359mg, 1.1mmol) were dissolved in 1, 4-dioxane (4mL) and stirred at 100 ℃ under nitrogen. The solvent was removed by concentration and purified on a silica gel column (dichloromethane: methanol ═ 20:1) to give the crude product as a pale yellow solid, which was slurried with dichloromethane (2mL), the solid was filtered and washed with ether and dried to give the final product B28 as a pale yellow solid (15mg, 33%).
TABLE 1 analytical Structure and spectral data of heterocyclic Compounds B1-B26
Figure BDA0002354170460000371
Figure BDA0002354170460000381
Figure BDA0002354170460000391
Figure BDA0002354170460000401
Figure BDA0002354170460000411
Example 16
This example was conducted to examine the effect of heterocyclic compounds B1-B26 prepared in examples 1-15 on the inhibition of TNF α -induced HT29 cell necrosis.
HT29 cells were loaded into 96-well plates, then pre-treated with different concentrations (0.3, 1.0, 3.0, 10.0, 20.0. mu.M) of test compound for two hours, and then the viability was quantified after treating the cells with a combination of TNF-. alpha. (40ng/mL), Smac mimetic (100nM) and z-VAD (20. mu.M) (caspase inhibitor, calbeiochem) for 48 hours. DMSO pre-treatment group was used as a negative control, and half inhibitory concentrations (50% Inhibitory Concentration (IC) as exemplified by compound B4 (FIG. 1) were calculated from survival rate curves measured at different concentrations50Value) was defined as the concentration of test compound required to inhibit TNF α -induced HT-29 cell necrosis by 50% relative to untreated control wells. The results are shown in Table 2.
TABLE 2 results of experiments for the inhibition of TNF α -induced necrosis of HT-29 cells by heterocyclic compounds B1-B26
Figure BDA0002354170460000412
Figure BDA0002354170460000421
Example 17
This example was conducted to measure the inhibitory effect of the heterocyclic compounds B2, B4, and B8 prepared in examples 2,4, and 7 on TNF α -induced MEF cell necrosis.
MEF cells were added to 96-well plates and then pre-treated with different concentrations (0.3, 1.0, 3.0, 10.0, 20.0 μ M) of test compound for two hours, and the viability was quantitated after 12 hours of combined treatment of the cells with TNF- α (40ng/mL), Smac mimetic (100nM) and z-VAD (20 μ M) (caspase inhibitor, calbeiochem). The DMSO pretreatment group was used as a negative control, and half inhibitory concentrations were calculated from a working curve of survival rates measured at different concentrations, as exemplified by compound B4 (fig. 2). The results are shown in table 3 (experiment results of inhibition of TSZ-induced MEF apoptosis by heterocyclic compounds B2, B4, and B8).
TABLE 3 inhibition experiment results of TNF α -induced MEF apoptosis by heterocyclic compounds B2, B4 and B8
Numbering EC50(μM) Numbering EC50(μM) Numbering EC50(μM)
B2 0.435 B4 9.33 B8 1.77
Example 18
This example was conducted to measure the inhibitory effects of the heterocyclic compounds B1 and B2 prepared in examples 1 and 2 on TNF α -induced L929 cell necrosis.
L929 cells were added to 96-well plates and then pre-treated with different concentrations (0.3, 1.0, 3.0, 10.0, 20.0. mu.M) of test compound for two hours, followed by treatment of the cells with a combination of TNF-. alpha. (40ng/mL), Smac mimetic (100nM) and z-VAD (20. mu.M) (caspase inhibitor, calbeiochem) for 16 hours, and then assayed for viability quantitatively. The DMSO pretreatment group was used as a negative control, and half inhibitory concentrations were calculated from a working curve of survival rates measured at different concentrations, as exemplified by compound B1 (fig. 3). The results are shown in Table 4.
TABLE 4 inhibition of TNF α -induced L929 cell necrosis by heterocyclic compounds B1, B2
Figure BDA0002354170460000422
Figure BDA0002354170460000431
As can be seen from tables 2,3 and 4, the heterocyclic compounds of the present invention have a strong inhibitory effect on TNF α -induced necrosis of HT29 cells, MEF cells and L929 cells, are potent inhibitors of RIPK3 kinase, and can be used for the treatment or prevention of disorders responsive to necrosis.

Claims (8)

1. A heterocyclic compound with programmed cell necrosis pathway inhibition activity, and pharmaceutically acceptable salts, esters, solvates, prodrugs, isotopic labels, isomers thereof, wherein the compound of formula (I) has the structure:
Figure FDA0002354170450000011
wherein the content of the first and second substances,
n is 1 and 2;
x is selected from N or CR6
Y is selected from O, NR7、CR7R8
A is unsubstitutedOr by 1-4R respectively9Substituted C6-10Aryl or 5-10 membered heteroaryl, said heteroaryl containing 1-4 heteroatoms independently selected from O, N, S;
R1selected from hydrogen atom, deuterium atom, C1-6Alkyl radical, C3-6Cycloalkyl, said alkyl, cycloalkyl being unsubstituted or substituted by 1 to 3 halogen or deuterium atoms;
R2、R3,R7selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted with hydroxy, 1-3 halo or cyclopropyl; or R2And R3May be linked to form ring B;
R4,R5,R6independently hydrogen atom, deuterium atom, cyano group, halogen, hydroxyl group, amino group, C1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C3-6Cycloalkyl, OC1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted with 1-3 halogen, hydroxy;
R8selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, sulfone group, sulfoxide group, C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by hydroxy, OC1-3Alkyl, cyclopropyl, 1-3 halogen or deuterium atom substitutions;
or R8Selected from unsubstituted or substituted by 1-3R10Substituted C6-10Aryl or 5-10 membered heteroaryl, said heteroaryl containing 1-3 heteroatoms independently selected from O, N, S;
or R7And R8May be composed of ═ O and ═ CH2Or R7And R8May be linked to form ring C;
the ring B and the ring C are respectively and independently selected from 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 4-8 membered cycloalkenyl, 4-8 membered heterocycloalkenyl, the 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 4-8 membered heterocycloalkenyl are substituted by 1-3R11Substitution; the 3-8 membered heterocycloalkyl, 4-8 membered heterocycloalkenyl containing 1-3 heteroatoms independently selected from O, N, S or selected from-C (═ O) N (R)11)-、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)-、-S(=O)2and-NHC (═ O) NH-;
R9,R10each independently selected from hydrogen atom, deuterium atom, cyano, halogen, hydroxyl, amino, C1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C3-6Cycloalkyl, OC1-8Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, COOC1-6Alkyl, said amino, alkyl, cycloalkyl, alkenyl and alkynyl groups being unsubstituted or substituted with 1-3 halogen, hydroxy, amino, acetyl or deuterium atoms;
R11selected from hydrogen atom, deuterium atom, C1-3Alkyl radical, C3-6Cycloalkyl, said alkyl, cycloalkyl being unsubstituted or substituted by 1 to 3 halogen or deuterium atoms.
2. The heterocyclic compound having an inhibitory activity on a programmed cell necrosis pathway according to claim 1, and pharmaceutically acceptable salts, esters, solvates, prodrugs, isotopic labels, isomers thereof, wherein:
x is selected from N atoms.
3. The heterocyclic compound having an inhibitory activity on a programmed cell necrosis pathway according to claims 1 and 2, and pharmaceutically acceptable salts, esters, solvates, prodrugs, isotopic labels, isomers thereof, wherein:
R7and R8Is composed of ═ O and ═ CH2Or R7And R8When combined C, R2、R3Selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by hydroxy, 1-3 halogen or cyclopropyl, or R2And R3May be linked to form ring B;
or R2And R3When joined to form a ring B, R8Selected from hydrogen atom, deuterium atom, halogen, amino group, hydroxyl group, carboxyl group, cyano group, C1-6Alkyl radical, C3-6Cycloalkyl, OC1-6Alkyl, NHC1-6Alkyl, N (C)1-6Alkyl radical)2、C2-6Alkenyl radical, C2-6Alkynyl, said alkyl, cycloalkyl, alkenyl and alkynyl being unsubstituted or substituted by hydroxy, OC1-3Alkyl, cyclopropyl, 1-3 halogen or deuterium atoms substituted, or R8Selected from unsubstituted or substituted by 1-3R10A substituted 5-10 membered aromatic or heteroaromatic ring containing 1-3 heteroatoms independently selected from O, N, S.
4. The heterocyclic compound having an inhibitory activity on programmed cell necrosis pathway according to claims 1 and 2, wherein a is unsubstituted or 1 to 3 independently selected from deuterium atom, halogen, hydroxyl group, C, and pharmaceutically acceptable salts, esters, solvates, prodrugs, isotopic labels, isomers thereof1-3Alkyl radical, C1-3Substituent of alkoxy group any of the following groups:
Figure FDA0002354170450000031
5. the heterocyclic compound having an inhibitory activity on a programmed cell necrosis pathway according to any one of claims 1 to 4, and pharmaceutically acceptable salts, esters, solvates, prodrugs, isotopic labels, isomers thereof, characterized in that the heterocyclic compound comprises:
Figure FDA0002354170450000041
6. a pharmaceutical composition comprising a therapeutically effective amount of one or more compounds of formula (I) according to any one of claims 1 to 5 or a pharmaceutically acceptable salt, ester, solvate, prodrug, isotopic label, isomer thereof, and further comprising at least one pharmaceutically acceptable carrier.
7. A composition comprising a compound of formula (I) according to any one of claims 1 to 5 or a pharmaceutically acceptable salt, ester, solvate, prodrug, isotopic label, isomer or a pharmaceutical composition according to claim 6 and one or more of an anti-neoplastic agent, an anti-autoimmune disorder agent, an anti-neurodegenerative disorder agent, an anti-metabolic disorder agent and an anti-aging agent.
8. Use of a compound of formula (I), or a pharmaceutically acceptable salt, ester, solvate, prodrug, isotopic label, isomer thereof, as claimed in any one of claims 1 to 5, or a composition as claimed in claim 6 or 7, for the manufacture of a medicament for the treatment of a condition by antagonising the programmed cell necrosis pathway, said condition comprising: uveitis, dermatitis, acute lung injury, type II diabetes, arthritis, ulcerative colitis, Crohn's disease, early-onset inflammatory bowel disease, extraintestinal inflammatory bowel disease, prevention of ischemia reperfusion injury in solid organ transplantation, non-alcoholic fatty liver disease, autoimmune hepatitis, asthma, systemic lupus erythematosus, sarcoidosis, Wegener's granulomatosis and interstitial lung disease, pulmonary fibrosis, renal fibrosis, hepatic fibrosis, myocardial infarction, asthma, hypersensitivity pneumonitis, interstitial lung disease, ankylosing spondylitis, multiple sclerosis, systemic sclerosis, polymyositis, rheumatoid arthritis, myasthenia gravis, juvenile onset diabetes, glomerulonephritis, autoimmune thyroiditis, graft rejection, Crohn's disease, scleroderma, psoriasis, dermatitis, retinitis pigmentosa, proliferative vitreoretinopathy, proliferative disorders, Bests ' vitelliform macular degeneration, eczema, urticaria, vasculitis, eosinophilic fasciitis, wet and dry age-related macular degeneration (ARMD), diabetic retinopathy, retinopathy of prematurity (ROP), diabetic macular redness, uveitis, retinal vein occlusion, cystoid macular edema, glaucoma, parkinson, alzheimer's disease, huntington's disease, breast cancer, lung cancer, bladder cancer, pancreatic cancer, liver cancer, head and neck squamous carcinoma, thyroid cancer, sarcoma, osteosarcoma, desmoid tumor, melanoma, prostate cancer, colorectal cancer, ovarian cancer, cervical cancer, esophageal cancer, gastric cancer, myeloma, lymphoma, mantle cell lymphoma, cutaneous T-cell lymphoma, chronic and non-progressive anemia, idiopathic or essential thrombocythemia, leukemia, acute leukemia, chronic leukemia, lymphatic leukemia, multiple sclerosis, myeloid leukemia, myelodysplastic syndrome, myeloproliferative disorders, brain tumor, astrocytoma, medulloblastoma, Schwannoma, primary neuroectoblastoma or pituitary tumor.
CN202010002951.8A 2020-01-02 2020-01-02 Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof Pending CN113061142A (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN202010002951.8A CN113061142A (en) 2020-01-02 2020-01-02 Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof
EP21736180.7A EP4085059B1 (en) 2020-01-02 2021-01-04 Heteroaryl compounds as inhibitors of programmed necrosis pathway, composition and method using the same
PCT/US2021/012124 WO2021138694A1 (en) 2020-01-02 2021-01-04 Heteroaryl compounds as inhibitors of programmed necrosis pathway, composition and method using the same
US17/790,728 US20230121233A1 (en) 2020-01-02 2021-01-04 Heteroaryl compounds as inhibitors of programmed necrosis pathway, composition and method using the same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010002951.8A CN113061142A (en) 2020-01-02 2020-01-02 Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof

Publications (1)

Publication Number Publication Date
CN113061142A true CN113061142A (en) 2021-07-02

Family

ID=76558222

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010002951.8A Pending CN113061142A (en) 2020-01-02 2020-01-02 Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof

Country Status (1)

Country Link
CN (1) CN113061142A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060167035A1 (en) * 2004-12-22 2006-07-27 Wolfgang Schwede Quinoline derivative, its use, production and pharmaceutical agents containing the latter
CN101087788A (en) * 2004-12-22 2007-12-12 拜耳先灵医药股份有限公司 Quinoline derivative, its use, production and pharmaceutical agents containing the latter
WO2007147578A1 (en) * 2006-06-21 2007-12-27 Bayer Schering Pharma Aktiengesellschaft Quinoline derivatives, their preparation, their use, and medicaments comprising them
CN103857288A (en) * 2011-03-04 2014-06-11 葛兰素史密斯克莱知识产权(第2号)有限公司 Amino-quinolines as kinase inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060167035A1 (en) * 2004-12-22 2006-07-27 Wolfgang Schwede Quinoline derivative, its use, production and pharmaceutical agents containing the latter
CN101087788A (en) * 2004-12-22 2007-12-12 拜耳先灵医药股份有限公司 Quinoline derivative, its use, production and pharmaceutical agents containing the latter
WO2007147578A1 (en) * 2006-06-21 2007-12-27 Bayer Schering Pharma Aktiengesellschaft Quinoline derivatives, their preparation, their use, and medicaments comprising them
CN103857288A (en) * 2011-03-04 2014-06-11 葛兰素史密斯克莱知识产权(第2号)有限公司 Amino-quinolines as kinase inhibitors

Similar Documents

Publication Publication Date Title
CN106661000B (en) EGFR inhibitor and its preparation and application
CN107793413B (en) Pyrimidine heterocyclic compound and preparation method and application thereof
CN107840846B (en) Pyrimidine ring-containing compound, EGFR inhibitor and application thereof
KR102520543B1 (en) pyridine compounds
CN111377918B (en) KRAS inhibitor compound
JP7337951B2 (en) Nitrogen-containing aromatic heterocyclic amide derivatives for treating cancer
CN109310671B (en) Bruton's tyrosine kinase inhibitors
CN106749233B (en) Sulfonamide derivatives and application thereof
KR102386428B1 (en) Heterocyclic compounds used as FGFR inhibitors
CN107674013A (en) Polycyclic compound, its preparation method, pharmaceutical composition and application
CN114728926A (en) ROCK inhibitor and preparation method and application thereof
CN102134234B (en) Indazolyl urea compounds and preparation method and medicinal use thereof
WO2021197250A1 (en) Novel compound as rearranged during transfection kinase inhibitor
CN112479956A (en) Method for preparing adenosine receptor inhibitor intermediate
CN109384767B (en) Preparation method and intermediate of pyridopyrimidine derivative
CN114605391B (en) Quinoxaline derivative, preparation method and application thereof
CN113061142A (en) Heterocyclic compounds with programmed cell necrosis pathway inhibition activity and application thereof
CN113173924B (en) Pyridine acetamide derivative as CDK inhibitor, and preparation method and application thereof
CN115466289A (en) Compound with TYK2 inhibitory activity, pharmaceutical composition containing same, and application thereof
KR20230133396A (en) Heteroaryl compounds as RIP2 kinase inhibitors, compositions and uses thereof
CN111032659B (en) Imidazo [1, 2-a ] imidazol-2-one derivatives for the treatment of diseases such as parkinson's disease
KR20210024551A (en) Determination of benzoxazole derivatives
CN111377873A (en) Aminopyrimidine compounds, their preparation and use
CN110684048A (en) Novel phosphoinositide 3-kinase inhibitor and preparation method and application thereof
KR102342803B1 (en) Novel pyrimidine derivatives and composition for preventing or treating neurodegenerative disease and cancer comprising thereof

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: 20211123

Address after: Room 2808, 28 / F, Wu Chung House, 213 Queen's Road East, Wanchai, Hong Kong, China

Applicant after: Aikono biomedical (Hong Kong) Co.,Ltd.

Address before: Mailbox 2547, Caster Chamber of Commerce 5-204,23, Governor's Square, Lemon Tree Bay Street, Grand Cayman, Cayman Islands

Applicant before: ACCRO BIOSCIENCE Inc.

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40052898

Country of ref document: HK

SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination