WO2021259077A1 - 取代吡嗪类化合物,包含其的药物组合物及其用途 - Google Patents

取代吡嗪类化合物,包含其的药物组合物及其用途 Download PDF

Info

Publication number
WO2021259077A1
WO2021259077A1 PCT/CN2021/099561 CN2021099561W WO2021259077A1 WO 2021259077 A1 WO2021259077 A1 WO 2021259077A1 CN 2021099561 W CN2021099561 W CN 2021099561W WO 2021259077 A1 WO2021259077 A1 WO 2021259077A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
amino
membered
compound
cycloalkyl
Prior art date
Application number
PCT/CN2021/099561
Other languages
English (en)
French (fr)
Inventor
易磊
田强
陈寿军
宋立强
王太津
刘谦
葛勇
杨禹
陈慧萍
宋宏梅
王晶翼
Original Assignee
四川科伦博泰生物医药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 四川科伦博泰生物医药股份有限公司 filed Critical 四川科伦博泰生物医药股份有限公司
Priority to JP2022571308A priority Critical patent/JP2023530838A/ja
Priority to EP21829137.5A priority patent/EP4169913A1/en
Priority to US17/926,763 priority patent/US20230212180A1/en
Priority to CN202180036771.4A priority patent/CN115667239A/zh
Publication of WO2021259077A1 publication Critical patent/WO2021259077A1/zh

Links

Images

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/20Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/10Spiro-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present invention requires an invention patent application filed in China on June 22, 2020, entitled “Substituted Pyrazine Compounds, Pharmaceutical Compositions Containing It and Their Uses", and the application number is 202010576572.X, and November 27, 2020
  • the present invention belongs to the field of medicinal chemistry, and relates to a substituted pyrazine compound used as an inhibitor of SHP2 (s rc h omology 2 domain-containing phosphotyrosine p hosphatase 2 ), a pharmaceutical composition containing it, and its use for prevention and/ Or the medical use of treating SHP2 related diseases.
  • SHP2 genes of a PTPN11 (p rotein t yrosine p hosphatase n onreceptor 11) encoded a protein tyrosine phosphatases (PTPs), intracellular PTPs non-receptor family members, their catalytic tyrosine proteins The acid undergoes a dephosphorylation reaction.
  • SHP2 has two SH2 ( s rc h omology 2 ) domains (N-SH2 at the N-terminus and C-SH2 at the C-terminus), a catalytic domain (PTP) and a proline-rich group and tyrosine phosphorylation The C-terminal tail of the site.
  • SHP2 In the inactivated state, SHP2 is in a self-inhibiting state, and N-SH2 and PTP bind to each other, thereby inhibiting phosphatase activity.
  • growth factors such as PDGF (p latelet- d erived g rowth f actor) and FGF (f ibroblast g rowth f actor ) such stimulation, phosphorylated tyrosine residues Tyr542 (Y542 ) And Tyr580 (Y580), and combined with N-SH2 to expose the catalytically active site of the PTP domain, thereby releasing the self-inhibition state, activating the PTP activity of SHP2, and triggering the signal initiated by tyrosine phosphorylation Conduction cascade reaction.
  • PDGF p latelet- d erived g rowth f actor
  • FGF f ibroblast g rowth f actor
  • SHP2 is widely expressed in the human body and participates in multiple signal pathways such as Ras-Erk, PI3K-Akt, Jak-Stat, Met, FGFR, EGFR and NF-kB, and regulates cell proliferation, differentiation, migration, apoptosis and other physiological functions.
  • SHP2 activating mutants are related to the occurrence of many diseases, such as Noonan syndrome, breast cancer, melanoma, etc. Overexpression of SHP2 will increase chronic myeloid leukemia, mastocytosis, and malignant glioma
  • SHP2 inhibitors to prevent and/or treat cancer and other related diseases.
  • the present invention has surprisingly discovered a series of substituted pyrazine compounds through a large amount of research, which have high SHP2 inhibitory activity and can be used as SHP2 inhibitors to prevent and/or treat SHP2 related diseases, especially tumor diseases. Shows good application prospects.
  • the present invention provides a compound having the structure of Formula I or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, isotope label, Metabolites or prodrugs, of which
  • X is selected from chemical bond, S, O, NH and CH 2 ;
  • R 1 is selected from hydrogen, hydroxy, halogen, amino, C 1-6 alkyl, 3-6 membered heterocycloalkyl and C 3-6 cycloalkyl, wherein the alkyl, heterocycloalkyl and cycloalkyl Each is optionally substituted with one or more substituents selected from halogen, hydroxyl and amino;
  • R 2 is selected from hydrogen, hydroxy, amino, cyano, halogen, C 1-6 alkyl, 3-6 membered heterocycloalkyl and C 3-6 cycloalkyl, wherein the alkyl, heterocycloalkyl and Each cycloalkyl is optionally substituted with one or more substituents selected from halogen, oxo, hydroxy, and amino;
  • A is selected from C 6-12 arylene, 5-12 membered heteroarylene, 3-12 membered heterocycloalkylene and C 3-8 cycloalkylene, wherein the arylene, heteroarylene , alkylene cycloalkyl alkylene and heterocycloalkyl are each optionally substituted with one or more substituents R a;
  • X is NH or CH 2 when any one of R a and X, and atoms attached thereto together form a 5-10 membered cycloaliphatic, 5-10 membered heteroalicyclic, 5-6 membered heteroaryl ring or a benzene ring, wherein said alicyclic ring, heteroalicyclic ring, heteroaromatic ring and benzene ring are each optionally one or more selected from amino, -NH (C 1-6 alkyl), -N (C 1-6 alkyl) 2. Substituent substitution of halogen, cyano, oxo, hydroxy, -O-(C 1-6 alkyl), C 1-6 haloalkyl and C 1-6 alkyl;
  • R b is selected from hydrogen, halogen, amino, cyano, C 1-6 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, 3-10 membered heterocycloalkenyl and 3-10 membered hetero Cycloalkyl, wherein the alkyl, cycloalkyl, cycloalkenyl, heterocycloalkenyl, and heterocycloalkyl are each optionally selected from hydrogen, halogen, oxo, hydroxy, and cyano. And amino substituents;
  • C is selected from C 6-12 arylene, 5-12 membered heteroarylene, 3-12 membered heterocycloalkylene, C 3-12 cycloalkenylene, 3-12 membered heterocycloalkenylene and C 3-8 cycloalkylene, wherein the arylene, heteroarylene, heterocycloalkylene, cycloalkenylene, heterocyclylene alkenylene and cycloalkyl are each optionally substituted with one or more R c replaces;
  • Each of the groups is optionally substituted with one or more substituents selected from halogen, oxo, hydroxy,
  • R 3 and R 4 and the atoms to which they are connected together form a 4-8 membered alicyclic ring or a 4-8 membered heteroalicyclic ring, wherein the alicyclic ring and the heteroalicyclic ring are each optionally substituted by one or more R 4a , Or the alicyclic and heteroalicyclic rings are each optionally fused with one or more C 6-10 aromatic rings or 5-12 membered heteroaromatic rings, each of the aromatic rings and heteroaromatic rings is optionally fused with one Or multiple R 4a substitutions;
  • g 0, 1 or 2;
  • n 0, 1, 2, 3, 4, or 5.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least one of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, formula IVd, Va, Vb Or a compound of Vc structure or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, isotope label, metabolite or prodrug, and one or more A pharmaceutically acceptable carrier and/or one or more additional pharmaceutical active ingredients.
  • the present invention provides a medicine, which comprises:
  • the present invention provides a compound having the structure of Formula I, Formula IIa, Formula IIb, Formula IIIa, Formula IIIb, Formula IVa, Formula IVb, Formula IVc, Formula IVd, Va, Vb, or Vc, or a pharmaceutically acceptable compound thereof Salts, esters, stereoisomers, tautomers, polymorphs, solvates, isotope markers, metabolites or prodrugs of, or the pharmaceutical composition, or the drug, which is used as SHP2 Inhibitors are used to prevent and/or treat diseases or disorders (especially cancer) mediated at least in part by SHP2.
  • the present invention provides a compound having the structure of Formula I, Formula IIa, Formula IIb, Formula IIIa, Formula IIIb, Formula IVa, Formula IVb, Formula IVc, Formula IVd, Va, Vb, or Vc, or a pharmaceutically acceptable compound thereof Salts, esters, stereoisomers, tautomers, polymorphs, solvates, isotope markers, metabolites or prodrugs, or the pharmaceutical composition, or the drug, used as SHP2 inhibitor The purpose of the agent.
  • the present invention provides a compound having the structure of Formula I, Formula IIa, Formula IIb, Formula IIIa, Formula IIIb, Formula IVa, Formula IVb, Formula IVc, Formula IVd, Va, Vb, or Vc, or a pharmaceutically acceptable compound thereof Salts, esters, stereoisomers, tautomers, polymorphs, solvates, isotopic markers, metabolites or prodrugs of, or the pharmaceutical composition, or the drug, in the preparation for Use in medicine for preventing and/or treating diseases or disorders (especially cancer) mediated at least in part by SHP2.
  • the present invention provides a method for preventing and/or treating a disease or condition (especially cancer) mediated at least in part by SHP2, which comprises the following steps: a preventive and/or therapeutically effective amount of Compounds of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, formula IVd, Va, Vb or Vc structure or pharmaceutically acceptable salts, esters, stereoisomers thereof , Tautomers, polymorphs, solvates, isotope markers, metabolites or prodrugs, or the pharmaceutical composition, or the drug, is administered to individuals in need thereof.
  • the present invention provides a combined method for preventing and/or treating diseases or conditions (especially cancer) mediated at least in part by SHP2, which includes:
  • the present invention provides a substituted pyrazine compound with a novel structure, which can be used as a highly active SHP2 inhibitor and can achieve at least one of the following technical effects:
  • Figure 1 shows the comparison of the in vivo efficacy of the positive compound and the compound of Example 12 in the KYSE-520 cell transplantation tumor model.
  • Figure 2 shows the comparison of the in vivo efficacy of the positive compound and the compound of Example 12 in the NCI-H358 cell transplantation tumor model.
  • any numerical value or any subrange falling within the range means that it is specifically disclosed.
  • each numerical range of the parameter disclosed herein should be understood to encompass Each of these values and sub-ranges.
  • “C 1-6” should be understood as covering any sub-range and each point value, such as C 2-5 , C 3-4 , C 1-2 , C 1-3 , C 1-4 , C 1-5, etc., and C 1 , C 2 , C 3 , C 4 , C 5 , C 6, etc.
  • 3-10 yuan should be understood as covering any sub-range and each point value, such as 3-4 yuan, 3-5 yuan, 3-6 yuan, 3-7 yuan, 3-8 yuan, 3-9 yuan, 4-5 yuan, 4-6 yuan, 4-7 yuan, 4-8 yuan, 5-7 yuan, 5-8 yuan, 6-7 yuan, etc., and 3, 4, 5, 6, 7, 8, 9, 10 yuan, etc.
  • pharmaceutically acceptable salt refers to a salt of the compound of the present invention that is substantially non-toxic to living organisms.
  • Pharmaceutically acceptable salts generally include (but are not limited to) the salts formed by the reaction of the compounds of the present invention with pharmaceutically acceptable inorganic acids/organic acids/acidic amino acids or inorganic bases/organic bases/basic amino acids, such salts Also known as acid addition salt or base addition salt.
  • suitable acid addition salts include, but are not limited to, acetates.
  • esters refers to an ester that is substantially non-toxic to the organism and is hydrolyzed to form the compound of the invention or its salt in the organism.
  • Pharmaceutically acceptable esters generally include, but are not limited to, esters formed by the compounds of the present invention and pharmaceutically acceptable carboxylic acids or sulfonic acids, and such esters are also called carboxylic acid esters or sulfonic acid esters.
  • the compound of the present invention may itself be an ester.
  • isomers refers to compounds that have the same molecular weight because of the same number of atoms and atomic types, but differ in the arrangement or configuration of the atoms in space.
  • stereoisomer refers to having at least one chiral factor (including chiral center, chiral axis, chiral plane, etc.) resulting in a vertical asymmetric plane, So that it can rotate the stable isomer of plane polarized light. Since the compounds of the present invention have asymmetric centers and other chemical structures that may lead to stereoisomerism, the present invention also includes these stereoisomers and mixtures thereof. Since the compounds of the present invention (or pharmaceutically acceptable salts thereof) include asymmetric carbon atoms, they can be in the form of single stereoisomers, racemates, enantiomers, and mixtures of diastereomers Form exists. Generally, these compounds can be prepared as racemates.
  • such compounds can be prepared or separated to obtain pure stereoisomers, that is, single enantiomers or diastereomers, or enrichment of single stereoisomers (purity ⁇ 99%, ⁇ 98%, ⁇ 97%, ⁇ 96%, ⁇ 95%, ⁇ 90%, ⁇ 85%, ⁇ 80%, ⁇ 75%, ⁇ 70%, ⁇ 65% or ⁇ 60%).
  • a single stereoisomer of a compound is synthetically prepared from an optically active starting material containing the desired chiral center, or is prepared by preparing a mixture of enantiomeric products and then separating or resolving.
  • the obtained for example, is converted into a mixture of diastereomers and then subjected to separation or recrystallization, chromatographic treatment, chiral resolution reagents, or direct separation of the enantiomers on a chiral chromatography column.
  • Starting compounds with specific stereochemistry are either commercially available or can be prepared according to the methods described below and then resolved by methods well known in the art.
  • enantiomers refers to a pair of stereoisomers that have non-superimposable mirror images of each other.
  • diastereomer or “diastereomer” refers to optical isomers that do not constitute mirror images of each other.
  • racemic mixture or “racemate” refers to a mixture containing equal parts of a single enantiomer (ie, a mixture of two R and S enantiomers in equimolar amounts).
  • non-racemic mixture refers to a mixture containing unequal parts of single enantiomers. Unless otherwise indicated, all stereoisomeric forms of the compounds of the present invention are within the scope of the present invention.
  • Solid lines used in this article Solid wedge Virtual wedge To describe the covalent chemical bonds of the compounds of the present invention.
  • a solid line is used to depict a bond to a chiral atom, it means that all possible stereoisomers at the chiral atom (e.g., specific enantiomers, racemic mixtures, etc.) are included.
  • real or imaginary wedges are used to depict bonds to chiral atoms, it indicates the presence of the indicated stereoisomers.
  • the stereoisomers of the compounds of the present invention may encompass specific enantiomers, diastereomers, racemates, or mixtures thereof in any ratio.
  • tautomers refers to structural isomers with different energies that can be converted into each other through a low energy barrier. If tautomerism is possible (as in solution), the chemical equilibrium of tautomers can be reached.
  • proton tautomers include (but are not limited to) interconversion through proton migration, such as keto-enol isomerization, imine-enamine isomerization Isomerization, amide-imino alcohol isomerization, nitroso-oxime isomerization, etc. Unless otherwise indicated, all tautomeric forms of the compounds of the present invention are within the scope of the present invention.
  • polymorph refers to a solid crystal form of a compound or complex.
  • Those skilled in the art can obtain molecular polymorphs by many known methods. These methods include, but are not limited to, melt recrystallization, melt cooling, solvent recrystallization, desolvation, rapid evaporation, rapid cooling, slow cooling, vapor phase diffusion, and sublimation.
  • well-known techniques can be used to detect, classify and identify polymorphs.
  • DSC Differential Scanning Calorimetry
  • TGA Thermogravimetric Analysis
  • XRPD X-ray Powder Diffraction
  • SCXRD Single crystal X-ray diffraction
  • NMR solid-state nuclear magnetic resonance
  • IR infrared spectroscopy
  • SEM scanning electron microscopy
  • solvate refers to a substance formed by the combination of a compound of the present invention (or a pharmaceutically acceptable salt thereof) and at least one solvent molecule through non-covalent intermolecular force.
  • the compound of the present invention may exist in the form of a solvate, which contains a polar solvent as a structural element of the crystal lattice.
  • the amount of polar solvent can be in the form of a stoichiometric ratio or a non-stoichiometric ratio.
  • isotopic label refers to a derivative compound formed by replacing a specific atom in the compound of the present invention with its isotope atom.
  • the compounds of the present invention include various isotopes of H, C, N, O, F, P, S, Cl, such as 2 H (D), 3 H (T), 13 C, 14 C, 13 N , 15 N, 17 O, 18 O, 18 F, 31 P, 32 P, 34 S, 35 S, 36 S, 37 Cl and 125 I.
  • 12 C can be replaced by 13 C or 14 C; 1 H can be replaced by 2 H (D, deuterium) or 3 H (T, tritium); 16 O can be replaced by 18 O, etc.
  • the term "metabolite” refers to a derivative compound formed after the compound of the present invention is metabolized, such as oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis and other reactions.
  • metabolite refers to a derivative compound formed after the compound of the present invention is metabolized, such as oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis and other reactions.
  • the present invention covers all possible metabolite forms of the compounds of the present invention, that is, substances formed in the body of the individual to whom the compounds of the present invention are administered.
  • the metabolites of compounds can be identified by well-known techniques in the art, and their activity can be characterized by experiments.
  • prodrug refers to a derivative compound capable of directly or indirectly providing the compound of the present invention after administration to an individual.
  • Particularly preferred derivative compounds or prodrugs are compounds that can increase the bioavailability of the compounds of the present invention when administered to an individual (e.g., more easily absorbed into the blood), or promote delivery of the parent compound to the site of action (e.g., lymphatic system) compound of.
  • all prodrug forms of the compounds of the present invention are within the scope of the present invention, and various prodrug forms are known in the art, for example, see T. Higuchi, V. Stella, Pro-drugs as Novel Drug Delivery Systems[J], American Chemical Society, Vol. 14, 1975.
  • the present invention also covers the compounds of the present invention containing a protecting group.
  • a protecting group In any process of preparing the compounds of the present invention, protection of sensitive groups or reactive groups on any relevant molecule may be necessary and/or desirable, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting groups, such as those described in T.W.Greene, P.G.M.Wuts, Protective Groups in Organic Synthesis[M], John Wiley & Sons, 2006. Using methods known in the art, these protecting groups can be removed at appropriate subsequent stages.
  • chemical bond refers to the strong force between two or more atoms (or ions) adjacent to each other in the molecule or crystal of the pure substance, which mainly includes covalent bonds, ionic bonds, metal bonds, and coordination. Key etc. Unless otherwise specified, the chemical bonds in the compounds of the present invention in free form are mostly covalent bonds.
  • alkyl refers to a linear or branched saturated aliphatic hydrocarbon group.
  • C 1-6 alkyl refers to an alkyl group (such as methyl, Ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, etc.), which are optionally substituted by one or more (e.g., 1-3) described herein as substituents (e.g., halo substituted, the group is "C 1- 6 haloalkyl", such as -CF 3, -C 2 F 5, - CHF 2 , -CH 2 F, -CH 2 CF 3 , -CH 2 Cl, -CH 2 CH 2 CF 3, etc.; when substituted by
  • alkylene refers to a linear or branched divalent saturated aliphatic hydrocarbon group, and the two groups (or fragments) to which it is connected can either be connected to the same A carbon atom can be connected to different carbon atoms.
  • C 1-6 alkylene group refers to an alkylene group having 1-6 carbon atoms (such as methylene, 1,1-ethylene, 1,2-ethylene Group, 1,2-propylene, 1,3-butylene, etc.), which are optionally substituted with one or more (such as 1-3) substituents described herein (such as when substituted by halogen,
  • the group is "C 1-6 haloalkylene", such as -CF 2 -, -C 2 F 4 -, -CHF-, etc.).
  • cycloalkyl refers to a monocyclic or polycyclic (e.g., bicyclic) alkyl group (e.g., monocyclic cycloalkyl, such as cyclopropyl, cyclobutyl, Cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, etc.; or bicyclic cycloalkyl, including fused ring, bridged ring or spiro ring, such as decahydronaphthyl, bicyclo[2.2.1]heptyl , Spiro[4.5]decyl, etc.).
  • monocyclic cycloalkyl such as cyclopropyl, cyclobutyl, Cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, etc.
  • bicyclic cycloalkyl
  • C 3- 6 cycloalkyl means a cycloalkyl group having 3 to 6 ring carbon atoms, which is optionally substituted by one or more (e.g., 1-3) described herein Substituents described in (such as substituted by halogen, the group is "C 1-6 halocycloalkyl", such as 2-fluorocyclopropyl, 3-chlorocyclobutyl, 4-bromocyclohexyl Etc.; when substituted by hydroxy, the group is "C 1-6 hydroxycycloalkyl", such as 2-hydroxycyclopropyl, 3-hydroxycyclobutyl, 4-hydroxycyclohexyl, etc.).
  • cycloalkylene refers to a monocyclic or polycyclic (such as bicyclic) divalent alkyl group, and the two groups (or fragments) to which it is connected are both The same ring-forming carbon atom can be connected, and different ring-forming carbon atoms can be connected.
  • C 3-6 cycloalkylene refers to a cycloalkylene having 3-6 ring-forming carbon atoms, which is optionally substituted by one or more (such as 1-3 ) Substituent substitutions described herein (e.g. ).
  • heterocycloalkyl refers to a saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) group with one or Multiple carbon atoms (such as 1, 2, 3, 4, 5, 6, 7, 8 or 9) and one or more (such as 1, 2, 3 or 4) are each independently selected from N, O,
  • the heteroatoms of P and S for example, have 3-15 (preferably 3-8, more preferably 3-6) ring atoms in total.
  • the ring system in the heterocycloalkyl group can be a fused ring, a bridged ring or a spiro ring system. If the valence bond requirements are met, the heterocycloalkyl group can be connected to other groups (or fragments) through any carbon atom or heteroatom in the ring.
  • 3-6 membered heterocycloalkyl refers to a heterocycloalkyl group having 3-6 ring atoms (including one or more heteroatoms), which is optionally substituted by one or Multiple (e.g., 1 to 3) substituents as described herein (e.g., oxirane, sulfiethane, azaethyl, azetidinyl, oxetanyl , Thietanyl, Tetrahydrofuranyl, Dioxolanyl, Tetrahydrothienyl, Pyrrolidinyl, Pyrrolidone, Imidazolidinyl, Pyrazolidinyl, Tetrahydropyranyl, Tetrahydrothiopyran Alkyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,4-thiooxanyl, 1,4-dioxanyl, 1,4-dithianyl, piperazinyl
  • heterocycloalkylene refers to a monocyclic or polycyclic (such as bicyclic) divalent group with one or more carbon atoms (such as 1, 2, 3, 4, 5, 6, 7, 8, or 9) and one or more (such as 2, 3, or 4) heteroatoms each independently selected from N, O, P and S, which
  • the two connected groups (or fragments) can be connected to the same ring-forming carbon atom, or can be connected to the ring-forming carbon atom and the ring-forming heteroatom (such as N atom or P atom) respectively.
  • 3-10 membered heterocycloalkylene refers to a heterocycloalkylene group having 3-10 ring atoms (including one or more heteroatoms), which is optionally One or more (e.g. 1-3) substituents as described herein are substituted (e.g. ).
  • heterocycloalkenyl refers to a monocyclic or polycyclic (such as bicyclic) group with one or more carbon-carbon double bonds, which has One or more carbon atoms (such as 1, 2, 3, 4, 5, 6, 7, 8 or 9) and one or more (such as 1, 2, 3 or 4) are each independently selected from N,
  • the heteroatoms of O, P and S for example, have 3-15 (preferably 3-8, more preferably 3-6) ring atoms in total.
  • the ring system in the heterocycloalkenyl group can be a fused ring, a bridged ring or a spiro ring system. If the valence bond requirements are met, the heterocycloalkenyl group can be connected to other groups (or fragments) through any carbon atom or heteroatom in the ring.
  • 3-6 membered heterocycloalkenyl refers to a heterocycloalkyl group having 3-6 ring atoms (including one or more heteroatoms), which is optionally substituted by one or Multiple (e.g., 1 to 3) substituents described herein are substituted.
  • heterocyclenylene refers to a monocyclic or polycyclic (such as bicyclic) divalent group with one or more carbon-carbon double bonds, which One or more carbon atoms (such as 1, 2, 3, 4, 5, 6, 7, 8, or 9) and one or more (such as 1, 2, 3, or 4) in the ring are independently selected
  • the heteroatoms from N, O, P and S for example, have 3-15 (preferably 3-8, more preferably 3-6) ring atoms in total.
  • aryl refers to a monocyclic or condensed ring aromatic hydrocarbon group having a conjugated ⁇ -electron system.
  • C 6-12 aryl refers to an aryl group having 6-12 carbon atoms (such as phenyl, naphthyl, etc.), which is optionally substituted by one or more of the Substituents described are substituted (such as tolyl substituted by C 1-6 alkyl, chlorophenyl substituted by halogen, etc.).
  • arylene refers to a monocyclic or condensed ring divalent aromatic hydrocarbon group having a conjugated ⁇ -electron system.
  • C 6-10 arylene refers to an arylene group having 6-10 carbon atoms, which is optionally substituted with one or more substituents described herein (such as C 1-6 alkyl substituted tolylene group, halogen substituted chlorophenyl group, etc.).
  • heteroaryl refers to a monocyclic or condensed ring aromatic group with a conjugated ⁇ -electron system, with one or more carbon atoms in the ring (such as 1, 2, 3, 4, 5, 6, 9 or 10 carbon atoms) and one or more (e.g. 1, 2, 3, or 4) each independently selected from N, O, P, and S hetero Atoms, for example, have 5-12 (preferably 5-10, more preferably 5, 6, 9 or 10) ring atoms in total. If the valence bond requirements are met, the heterocycloalkyl group can be connected to the parent molecular moiety through any ring atom.
  • the heteroaryl group can be connected to other groups (or fragments) through any carbon atom or heteroatom (such as a N atom) in the ring. And, the heteroaryl group may optionally be further condensed with benzene.
  • the term "5-12 membered heteroaryl” as used herein refers to a heteroaryl group having 5-12 ring atoms (such as furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl , Isoxazolyl, isothiazolyl, pyrazolyl, oxadiazolyl, thiadiazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, quinolinyl, Isoquinolinyl, benzofuranyl, isobenzofuranyl, benzothienyl, isobenzothienyl, indolyl, isoindolyl or benzo derivatives thereof, etc.), which are optionally substituted by one Or substituted with multiple substituents described herein (such as methylpyridyl substituted by C 1-6 al
  • heteroarylene refers to a monocyclic or condensed divalent aromatic group with a conjugated ⁇ -electron system, with one or more Carbon atoms (e.g., 1, 2, 3, 4, 5, 6, 9, or 10 carbon atoms) and one or more (e.g., 1, 2, 3, or 4) are each independently selected from N, O, P, and
  • the heteroatom of S has, for example, 5-12 (preferably 5-10, more preferably 5, 6, 9 or 10) ring atoms in total.
  • 5-12 membered heteroarylene refers to a heteroarylene group having 5-12 ring atoms, which is optionally substituted by one or more substituents described herein (As substituted by C 1-6 alkyl Substituted by halogen Wait).
  • alkenyl refers to a linear or branched aliphatic hydrocarbon group having one or more carbon-carbon double bonds.
  • C 2-6 alkenyl refers to an alkenyl group having 2-6 carbon atoms and one, two or three (preferably one) carbon-carbon double bonds (such as vinyl, 1-propenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexene Group, 4-hexenyl, 5-hexenyl, 2-methyl-2-propenyl, 4-methyl-3-pentenyl, etc.), which are optionally combined with one or more (such as 1- 3) Substituents as described herein are substituted.
  • alkenylene refers to a straight or branched divalent aliphatic hydrocarbon group with one or more carbon-carbon double bonds, two of which are connected The groups (or fragments) can be connected to the same carbon atom or different carbon atoms.
  • C 2-6 alkenylene refers to an alkenylene group having 2-6 carbon atoms (such as Etc.), which are optionally substituted with one or more (such as 1-3) substituents described herein.
  • cycloalkenyl refers to a monocyclic or polycyclic (e.g., bicyclic) alkenyl group (e.g., monocyclic cycloalkenyl) having one or more carbon-carbon double bonds. Groups, such as cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, etc.).
  • C 3-6 cycloalkenyl refers to a cycloalkenyl group having 3-6 ring-forming carbon atoms, which is optionally substituted by one or more (such as 1-3) herein Substituent substitution described in (e.g. ).
  • cycloalkenylene refers to a monocyclic or polycyclic (such as bicyclic) divalent alkenyl group with one or more carbon-carbon double bonds, which is The two groups (or fragments) that are connected can be connected to the same ring-forming carbon atom or different ring-forming carbon atoms.
  • C 3-6 cycloalkenylene refers to a cycloalkenylene having 3-6 ring-forming carbon atoms, which is optionally substituted by one or more (such as 1-3 ) Substituent substitution as described in this article (e.g. ).
  • alkynyl refers to a linear or branched aliphatic hydrocarbon group having one or more carbon-carbon triple bonds.
  • C 2-6 alkynyl refers to an alkynyl group having 2-6 carbon atoms and one, two, or three (preferably one) carbon-carbon triple bonds (such as ethynyl , 1-propynyl, 2-propynyl, 2-butynyl, 3-butynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 2-hexynyl, 3 -Hexynyl, 4-hexynyl, 5-hexynyl, etc.), which are optionally substituted by one or more (such as 1-3) substituents described herein.
  • alkynylene refers to a straight or branched divalent aliphatic hydrocarbon group with one or more carbon-carbon triple bonds, two of which are connected The groups (or fragments) are respectively connected to different carbon atoms.
  • C 2-6 alkynylene refers to an alkynylene group having 2-6 carbon atoms (such as Etc.), which are optionally substituted with one or more (such as 1-3) substituents described herein.
  • alicyclic or "aliphatic ring” refers to monocyclic or polycyclic (including fused, bridged, and spirocyclic, such as bicyclic) aliphatic hydrocarbons
  • the ring system includes cycloalkane ring, cycloalkene ring and cycloalkyne ring.
  • heteroalicyclic ring or “heteroaliphatic hydrocarbon ring” refers to monocyclic or polycyclic (including fused ring, bridged ring and spiro ring, such as bicyclic) aliphatic Hydrocarbon ring system with one or more carbon atoms (such as 1, 2, 3, 4, 5, 6, 7, 8 or 9) and one or more (such as 1, 2, 3 or 4) in the ring A) heteroatoms each independently selected from N, O, P, and S, including heterocycloalkane ring, heterocycloalkene ring, heterocycloalkyne ring and the like.
  • aromatic ring or “aromatic hydrocarbon ring” refers to a monocyclic or polycyclic (such as bicyclic) aromatic hydrocarbon ring system.
  • heteromatic ring or “heteroaromatic ring” refers to a monocyclic or polycyclic (such as bicyclic) aromatic hydrocarbon ring system, which has one or Multiple carbon atoms (such as 1, 2, 3, 4, 5, 6, 7, 8 or 9) and one or more (such as 1, 2, 3 or 4) are each independently selected from N, O, Heteroatoms of P and S.
  • halogen refers to fluorine (F), chlorine (Cl), bromine (Br), or iodine (I).
  • hydroxyl refers to -OH.
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • amino refers to -NH 2 .
  • the substituent X and the substituent Y are each independently hydrogen, halogen, hydroxy, cyano, alkyl, or aryl.
  • the substituent Y can be either hydrogen or halogen. Hydroxy, cyano, alkyl or aryl; in the same way, when the substituent Y is hydrogen, the substituent X can be either hydrogen or halogen, hydroxy, cyano, alkyl or aryl.
  • substitution and its other variants in this context means that one or more (such as 1, 2, 3, or 4) atoms or groups of atoms (such as hydrogen atoms) on the designated atoms are replaced by other equivalents , The condition is that the normal valence of the specified atom or group of atoms in the current situation is not exceeded, and a stable compound can be formed. If an atom or group of atoms is described as "optionally substituted by”, it can be either substituted or unsubstituted. Unless otherwise specified, the attachment point of the substituent herein can be from any suitable position of the substituent. When the connecting bond in a substituent is shown as a chemical bond passing through the two atoms connected to each other in the ring system, it means that the substituent can connect to any ring atom in the ring system.
  • the present invention provides a compound of formula I or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, isotope label, metabolite or prodrug thereof, wherein
  • X is selected from chemical bond, S, O, NH and CH 2 ;
  • R 1 is selected from hydrogen, hydroxy, halogen, amino, C 1-6 alkyl, 3-6 membered heterocycloalkyl and C 3-6 cycloalkyl, wherein the alkyl, heterocycloalkyl and cycloalkyl Each is optionally substituted with one or more substituents selected from halogen, hydroxyl and amino;
  • R 2 is selected from hydrogen, hydroxy, amino, cyano, halogen, C 1-6 alkyl, 3-6 membered heterocycloalkyl and C 3-6 cycloalkyl, wherein the alkyl, heterocycloalkyl and Each cycloalkyl group is optionally substituted with one or more substituents selected from halogen, oxo, hydroxy and amino;
  • A is selected from C 6-12 arylene, 5-12 membered heteroarylene, 3-12 membered heterocycloalkylene and C 3-8 cycloalkylene, wherein the arylene, heteroarylene , alkylene cycloalkyl alkylene and heterocycloalkyl are each optionally substituted with one or more substituents R a;
  • X is NH or CH 2 when any one of R a and X, and atoms attached thereto together form a 5-10 membered cycloaliphatic, 5-10 membered heteroalicyclic, 5-6 membered heteroaryl ring or a benzene ring, wherein said alicyclic ring, heteroalicyclic ring, heteroaromatic ring and benzene ring are each optionally one or more selected from amino, -NH (C 1-6 alkyl), -N (C 1-6 alkyl) 2. Substituent substitution of halogen, cyano, oxo, hydroxy, -O-(C 1-6 alkyl), C 1-6 haloalkyl and C 1-6 alkyl;
  • R b is selected from hydrogen, halogen, amino, cyano, C 1-6 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkenyl, 3-10 membered heterocycloalkenyl and 3-10 membered hetero Cycloalkyl, wherein the alkyl, cycloalkyl, cycloalkenyl, heterocycloalkenyl, and heterocycloalkyl are each optionally selected from hydrogen, halogen, oxo, hydroxy, and cyano. And amino substituents;
  • C is selected from C 6-12 arylene, 5-12 membered heteroarylene, 3-12 membered heterocycloalkylene, C 3-12 cycloalkenylene, 3-12 membered heterocycloalkenylene and C 3-8 cycloalkylene, wherein the arylene, heteroarylene, heterocycloalkylene, cycloalkenylene, heterocyclylene alkenylene and cycloalkyl are each optionally substituted with one or more R c replaces;
  • Each of the groups is optionally substituted with one or more substituents selected from halogen, oxo, hydroxy,
  • R 3 and R 4 and the atoms to which they are connected together form a 4-8 membered alicyclic ring or a 4-8 membered heteroalicyclic ring, wherein the alicyclic ring and the heteroalicyclic ring are each optionally substituted by one or more R 4a , Or the alicyclic and heteroalicyclic rings are each optionally fused with one or more C 6-10 aromatic rings or 5-12 membered heteroaromatic rings, each of the aromatic rings and heteroaromatic rings is optionally fused with one Or multiple R 4a substitutions;
  • g 0, 1 or 2;
  • n 0, 1, 2, 3, 4, or 5.
  • X in the compound of formula I above is selected from a chemical bond and S; preferably, X is S.
  • R 1 in the compound of formula I above is selected from hydrogen, hydroxyl, halogen, amino, C 1-6 alkyl and C 3-6 cycloalkyl; preferably, R 1 is selected from hydrogen , Halogen, amino and C 1-6 alkyl (such as hydrogen, halogen and C 1-6 alkyl); more preferably, R 1 is selected from hydrogen, fluorine, amino and methyl; further preferably, R 1 is selected from Hydrogen, fluorine and methyl.
  • R 2 in the compound of formula I above is selected from hydrogen, hydroxy, halogen, C 1-6 alkyl and C 3-6 cycloalkyl, wherein the alkyl and cycloalkyl are each Optionally substituted by one or more substituents selected from halogen and hydroxy; preferably, R 2 is selected from hydrogen, hydroxy, fluorine, hydroxymethyl (-CH 2 OH) and fluoromethyl (-CH 2 F) ; More preferably, R 2 is selected from hydrogen, hydroxymethyl and fluoromethyl; further preferably, R 2 is hydroxymethyl.
  • a in the compound of formula I above is selected from C 6-12 arylene and 5-12 membered heteroarylene, wherein the arylene and heteroarylene are each optionally is substituted with one or more R a; preferably, a is selected from phenylene, alkylene pyridyl and pyrimidyl, wherein the phenylene, pyridyl, and pyrimidinyl are each alkylene optionally substituted with one or more R a substituents; more preferably, a is selected from phenylene and pyridinyl, wherein the phenylene and pyridinyl each optionally substituted with one or more substituents R a.
  • B in the compound of formula I above includes G, and G is selected from C 1-6 alkylene, C 3-6 cycloalkylene and 3-6 membered heterocycloalkylene, so
  • the alkylene, cycloalkylene and heterocycloalkylene groups are each optionally selected by one or more of hydrogen, fluorine, oxo, hydroxy, cyano, -OR b , C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 hydroxyalkyl substituents are substituted, preferably each is optionally substituted by one or more substituents selected from hydrogen and fluorine; preferably, G is C 1-3 alkylene
  • the alkylene group is optionally substituted by one or more groups selected from hydrogen, fluorine, oxo, hydroxy, cyano, -OR b , C 1-6 alkyl, C 1-6 haloalkyl and C 1
  • the -6 hydroxyalkyl substituent is substituted, preferably optionally substituted,
  • B in the compound of formula I above comprises R b , and R b is selected from hydrogen, fluorine, chlorine, amino, C 1-3 alkyl, cyclopropyl and 3-6 membered heterocycloalkane Group, wherein the alkyl group and heterocycloalkyl group are each optionally substituted with one or more substituents selected from hydrogen, fluorine, oxo, hydroxy, and cyano.
  • B in the compound of formula I above comprises C, and C is selected from 3-6 membered heterocycloalkylene and C 3-6 cycloalkylene, wherein said heterocycloalkylene and Each cycloalkylene group is optionally substituted with one or more R c ; preferably, C is selected from the group consisting of pyrrolidinylene, azetidinylene, piperidinylene and piperazinylene, wherein the sub Pyrrolidinyl, azetidinyl, piperidinylene and piperazinylene are each optionally substituted with one or more R c .
  • each R c in B in the above formula I compound is independently selected from hydrogen, fluorine, cyano, methyl, ethyl, fluoromethyl, Hydroxymethyl and hydroxyl; preferably, each R c is independently selected from hydrogen, fluorine and hydroxyl.
  • R 3 and R 4 in the compound of formula I are not connected to each other, and R 3 is selected from fluorine, chlorine, bromine, -OR z , hydroxyl, cyano, C 1-6 alkyl, -(C 1-4 alkylene)-R z , -(C 1-4 alkylene)-OR z , C 3-6 cycloalkyl, 3-8 membered heterocycloalkyl (such as 3-6 member Heterocycloalkyl), C 2-4 alkenyl, C 2-4 alkynyl, phenyl and 5-6 membered heteroaryl, wherein the alkyl, alkylene, cycloalkyl, heterocycloalkyl, Alkenyl, alkynyl, phenyl and heteroaryl are each optionally selected by one or more selected from fluorine, chlorine, cyano, oxo, -OR z , hydroxyl, -N(R z ) 2
  • -CH CH 2 , -CH 2 C ⁇ CH, -C ⁇ CH, pyridyl, pyrimidinyl, oxazolyl, thiazolyl, pyridazinyl, pyrazolyl and thienyl, wherein the pyridyl, Pyrimidine, oxazolyl, thiazolyl, pyridazinyl, pyrazolyl and thienyl are each optionally selected from fluorine, chlorine, cyano, -OR z , hydroxyl, -N (R z ). 2.
  • R 3 is selected from fluorine, chlorine, hydroxyl, cyano, methyl, ethyl, cyclopropyl, oxetanyl, Pyridyl, pyrimidinyl, oxazolyl, thiazolyl, pyridazinyl, pyrazolyl and thienyl; more preferably, R 3 is selected from fluorine, chlorine, methyl, ethyl, cyclopropyl and oxetanyl Alkyl; further preferably, R 3 is selected from fluorine and methyl.
  • R 3 in the compound of formula I above includes a plurality of R z , and each R z is independently selected from cyano, C 1-4 alkyl, C 3-6 cycloalkyl , 3-6 membered heterocycloalkyl, phenyl, benzyl and 5-6 membered heteroaryl, wherein the alkyl, cycloalkyl, heterocycloalkyl, phenyl, benzyl and heteroaryl are each any Optionally by one or more selected from amino, fluorine, chlorine, cyano, oxo, hydroxy, -O-(C 1-3 alkyl), methyl, ethyl, difluoromethyl, trifluoromethyl Substituents of phenyl group, C 3-6 cycloalkyl and phenyl; preferably, each R z is independently selected from the group consisting of cyano, methyl, ethyl, isopropyl, cycloprop
  • R 3 and R 4 in the compound of formula I described above are connected to each other, and R 3 and R 4 and the atoms connected thereto form a ring, choose from any of the following:
  • n 0, 1 or 2;
  • R 3 and R 4 and the atoms connected to them form a ring choose from any of the following:
  • R 3 and R 4 and the atoms to which they are connected together form a ring system simultaneously containing a plurality of R 4a
  • the compound of Formula I contains a plurality of R 5, not connected to any two of R 5 and the atom the same atom attached thereto form a C 4-8 alicyclic ring or a 4-8 membered heteroaryl with Alicyclic ring, preferably C 4-6 alicyclic ring or 4-6 membered nitrogen-containing heteroalicyclic ring, or any two R 5 connected to the same atom and the atoms connected with it together form a C 3-6 alicyclic ring or 3-6 membered
  • the heteroalicyclic ring is preferably a cyclopropane ring.
  • g is 0 or 2.
  • n is 0, 1, 2, or 3; preferably, n is 0, 1, or 2.
  • the above-mentioned compound of formula I has a structure as shown in formula IIa or formula IIb, wherein
  • A, B, R 3 and R 4 are as defined above.
  • the above-mentioned compound of formula I has a structure as shown in formula IIIa or formula IIIb, wherein
  • A, B, R 3 and R 4 are as defined above.
  • the above-mentioned compound of formula I has a structure as shown in formula IVa, formula IVb, formula IVc or formula IVd, wherein
  • A, B, R 3 and R 4 are as defined above.
  • the above-mentioned compound of formula I has a structure as shown in formula Va, formula Vb or formula Vc, wherein
  • A, B, R 3 and R 4 are as defined above.
  • the above-mentioned compound of formula I has a structure as shown in any one of formula IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from C 6-12 arylene group, and a 5-12 membered heteroaryl group, wherein the arylene and heteroarylene group each optionally substituted with one or more substituents R a;
  • G is selected from C 1-6 alkylene, C 3-6 cycloalkylene and 3-6 membered heterocycloalkylene, each of which is optionally Substituted by one or more substituents selected from hydrogen, fluorine, oxo, hydroxy, cyano, -OR b , C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 hydroxyalkyl, Preferably each is optionally substituted by one or more substituents selected from hydrogen and fluorine;
  • R b is selected from hydrogen, fluorine, chlorine, amino, C 1-3 alkyl, cyclopropyl and 3-6 membered heterocycloalkyl, wherein each of said alkyl and heterocycloalkyl is optionally substituted by one or more One substituent selected from hydrogen, fluorine, oxo, hydroxyl and cyano;
  • C is selected from 3-6 membered heterocycloalkylene and C 3-6 cycloalkylene, wherein each of said heterocycloalkylene and cycloalkylene is optionally substituted with one or more R c ;
  • each R c is independently selected from hydrogen, fluorine, cyano, methyl, ethyl, fluoromethyl, hydroxymethyl and hydroxyl;
  • R 3 is selected from fluorine, chlorine, bromine, -OR z , hydroxyl, cyano, C 1-6 alkyl, -(C 1-4 alkylene) -R z , -(C 1-4 alkylene) -OR z , C 3-6 cycloalkyl, 3-8 membered heterocycloalkyl (such as 3-6 membered heterocycloalkyl), C 2-4 alkenyl, C 2-4 alkynyl, phenyl and 5 -6 membered heteroaryl, wherein the alkyl, alkylene, cycloalkyl, heterocycloalkyl, alkenyl, alkynyl, phenyl and heteroaryl groups are each optionally selected by one or more fluorine , Chloro, cyano, oxo, -OR z , hydroxyl, -N(R z ) 2 , -NHR z , amino, C 1-4 alkyl, C 3-6
  • each R z is independently selected from cyano, C 1-4 alkyl, C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, phenyl, benzyl and 5-6 membered hetero Aryl, wherein the alkyl, cycloalkyl, heterocycloalkyl, phenyl, benzyl and heteroaryl groups are each optionally selected by one or more groups selected from amino, fluorine, chlorine, cyano, oxo , Hydroxy, -O-(C 1-3 alkyl), methyl, ethyl, difluoromethyl, trifluoromethyl, C 3-6 cycloalkyl and phenyl substituents;
  • n 0, 1 or 2;
  • the above-mentioned compound of formula I has a structure as shown in any one of formula IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from phenylene, alkylene pyridyl and pyrimidyl, wherein the phenylene, pyridyl, and pyrimidinyl are each alkylene optionally substituted with one or more substituents R a;
  • each R a is independently selected from hydrogen, fluoro, chloro, bromo, hydroxy, methoxy, cyclopropyloxy, cyano, methyl, ethyl, n-propyl, isopropyl, difluoromethyl Methoxy, trifluoromethoxy, trifluoromethyl, difluoromethyl, cyclopropyl, pyrrolidinyl, morpholinyl, amino, methylamino, dimethylamino, methylthio, methylsulfonyl and ammonia Sulfonyl
  • G is a C 1-3 alkylene group, and the alkylene group is optionally substituted by one or more selected from hydrogen, fluorine, oxo, hydroxy, cyano, -OR b , C 1-6 alkyl, C Substituent substitution of 1-6 haloalkyl and C 1-6 hydroxyalkyl;
  • R b is selected from hydrogen, fluorine, chlorine, amino, C 1-3 alkyl, cyclopropyl and 3-6 membered heterocycloalkyl, wherein each of said alkyl and heterocycloalkyl is optionally substituted by one or more One substituent selected from hydrogen, fluorine, oxo, hydroxyl and cyano;
  • C is selected from pyrrolidinylene, azetidinyl, piperidinylene and piperazinylene, wherein the pyrrolidinylene, azetidinyl, piperidinylene and piperazine Each of the groups is optionally substituted with one or more R c ;
  • each R c is independently selected from hydrogen, fluorine and hydroxyl
  • each R z is independently selected from cyano, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, morpholinyl, piperazinyl, oxa Cyclobutanyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, azetidinyl, phenyl, benzyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, oxazolyl , Thiazolyl, triazolyl, pyrazinyl, pyrimidinyl and pyridyl, wherein the piperazinyl, azetidinyl, phenyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazole Group, oxazolyl, thiazolyl,
  • R 4 is selected from fluorine, chlorine, methyl, ethyl, isopropyl, cyclopropyl, methoxymethyl, hydroxymethyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, cyanogen Methyl, hydroxymethyl, 2-methylfuryl, thiazolyl, pyridyl and pyrimidinyl;
  • n 0, 1 or 2;
  • each R 4a is independently selected from hydrogen, fluorine, chlorine, oxo, methyl, acetyl and cyclopropyl, wherein each of the methyl and cyclopropyl is optionally substituted by one or more Substituents selected from halogen, cyano, hydroxyl, -O-(C 1-6 alkyl) and C 1-6 alkyl are substituted.
  • the above-mentioned compound of formula I has a structure as shown in any one of formula IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from phenylene, alkylene pyridyl and pyrimidyl, wherein the phenylene, pyridyl, and pyrimidinyl are each alkylene optionally substituted with one or more substituents R a;
  • each R a is independently selected from hydrogen, fluoro, chloro, bromo, hydroxy, cyano, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, pyrrolidinyl, morpholinyl , Amino, methylamino and dimethylamino;
  • R 3 is selected from fluorine, chlorine, hydroxyl, cyano, methyl, ethyl, cyclopropyl, oxetanyl, pyridyl, pyrimidinyl, oxazolyl, thiazolyl, pyridazinyl, pyrazolyl And thienyl, preferably fluorine, chlorine, methyl, ethyl, cyclopropyl and oxetanyl;
  • R 4 is selected from methyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, methoxymethyl, fluoroethyl, cyanomethyl and hydroxymethyl;
  • the above-mentioned compound of formula I has a structure as shown in any one of formulas IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from phenylene, alkylene pyridyl and pyrimidyl, wherein the phenylene, pyridyl, and pyrimidinyl are each alkylene optionally substituted with one or more substituents R a;
  • each R a is independently selected from fluoro, chloro, bromo, cyano, methyl and amino;
  • R 3 is selected from fluorine and methyl
  • R 4 is selected from methyl, fluoromethyl, fluoroethyl, cyanomethyl, hydroxymethyl and methoxymethyl;
  • the above-mentioned compound of formula I has a structure as shown in any one of formula IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from 5-12 membered heteroarylene groups, wherein the heteroarylene groups are optionally substituted with one or more halogens;
  • C is selected from 3-6 membered heterocycloalkylene, which is optionally substituted by one or more fluorine, cyano, methyl or fluoromethyl;
  • R 3 is selected from C 1-6 alkyl
  • R 4 is selected from C 1-6 alkyl
  • the alkyl is optionally substituted by -O-(C 1-3 alkyl )replace
  • n 0, 1 or 2;
  • each R 4a is independently selected from hydrogen and fluorine.
  • the above-mentioned compound of formula I has a structure as shown in any one of formula IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from pyridylene, which is optionally substituted with one or more chlorines;
  • C is selected from the group consisting of azetidinyl and pyrrolidinylene, each of which is optionally substituted by one or more fluorine, cyano, methyl or fluoromethyl replace;
  • R 3 is selected from methyl
  • R 4 is selected from methyl
  • said methyl is optionally substituted by methoxy
  • the above-mentioned compound of formula I has a structure as shown in any one of formula IIa-IIb, IIIa-IIIb, IVa-IVd, and Va-Vc, wherein
  • A is selected from pyridylene, which is optionally substituted with one or more chlorines;
  • R 3 is selected from methyl
  • R 4 is selected from methyl
  • said methyl is optionally substituted by methoxy
  • the above-mentioned compound of formula I has a structure as shown in formula IIb, wherein
  • A is selected from 5-12 membered heteroarylene groups, wherein the heteroarylene group is optionally substituted by one or more halogens; preferably, A is selected from pyridylene groups, wherein the pyridylene group is optionally substituted by One or more chlorine substitutions;
  • B is selected from Wherein C is selected from 3-6 membered heterocycloalkylene, Y is selected from C 1-3 alkylene, R d is selected from hydroxyl; preferably, B is selected from
  • R 3 and R 4 and the atoms to which they are connected form a ring, and Selected from
  • the present invention provides the following compounds covered by the compounds of Formula I, Formula IIa, Formula IIb, Formula IIIa, Formula IIIb, Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula Va, Formula Vb, and Formula Vc:
  • the present invention provides two preparation methods of the compound of formula I;
  • Method 1 includes the following steps:
  • a compound of formula S-1 is reacted with a compound of formula S-2 to produce a compound of formula M-1;
  • Method 2 includes the following steps:
  • LG 1 and LG 2 are each independently a halogen leaving group or a C 1-6 alkylsulfonate leaving group optionally substituted by halogen (such as a triflate leaving group); In addition, LG 2 can also be a hydroxyl group;
  • R x is hydrogen or a leaving group
  • R f is hydrogen, hydroxymethyl, fluoromethyl, or a group converted into hydroxymethyl or fluoromethyl through one or more reactions;
  • PG 1 is a protective group for hydrogen or amino (such as methyl, tert-butoxycarbonyl, tert-butyldimethylsilyl, triisopropylsilyl, benzyl, methoxymethyl, etc.);
  • LG 1 is a halogen leaving group (such as iodine or bromine).
  • LG 2 is a halogen leaving group (such as bromine or chlorine) or a hydroxyl group.
  • R x is hydrogen, halogen, boronic acid group, boronic acid ester group, substituted silicon group, substituted metal group, or C 1-6 alkyl sulfonate optionally substituted by halogen
  • the group is preferably a boric acid group or a borate ester group.
  • the reaction in the above step (1) is carried out in the presence of a base.
  • the base is an inorganic base, such as potassium phosphate.
  • the reaction in the above step (1) is carried out in the presence of a condensation reagent and a base.
  • the base is an organic base, such as DIPEA.
  • the condensation reagent is BOP, HATU or PyBOP, preferably BOP.
  • the reaction in the above step (2) is carried out in the presence of a metal catalyst.
  • the metal catalyst is a palladium catalyst or a copper catalyst, such as tetrakis(triphenylphosphine)palladium, palladium acetate, tris(dibenzylideneacetone)dipalladium, [1,1'- Bis(diphenylphosphino)ferrocene]palladium dichloride, 1,2-bis(diphenylphosphino)ethane palladium dichloride and bis(triphenylphosphine)palladium dichloride, iodide Cuprous and so on.
  • the functional group conversion in the above step (3) includes (but is not limited to) the following reactions: 1) reduction reaction (reagents used such as LiBH 4 , DIBAL-H); 2) metal Catalyzed coupling reaction; 3) Hydrolysis reaction.
  • the deprotection in the above step (3) is carried out under acid hydrolysis or catalytic hydrogenolysis in the presence of a metal catalyst.
  • the acid used in the acid hydrolysis is an organic acid, preferably trifluoroformic acid.
  • the metal catalyst used in the catalytic hydrogenation is palladium/carbon or palladium hydroxide/carbon.
  • one or more steps in the above preparation method can be omitted, and the order of the reaction steps can also be appropriately adjusted as needed, and the protection/deprotection reaction steps can be added or omitted.
  • composition refers to a composition that can be used as a medicine, which comprises a pharmaceutical active ingredient (API) (or therapeutic agent) and optionally one or more pharmaceutically acceptable carriers, with the purpose of promoting The administration of is conducive to the absorption of active ingredients, and then exert biological activity.
  • pharmaceutically acceptable carrier refers to an excipient that is administered with the therapeutic agent, and is suitable for contact with human and/or other animal tissues within the scope of reasonable medical judgment without excessive toxicity, irritation, Allergic reactions or other problems or complications associated with a reasonable benefit/risk ratio.
  • the pharmaceutically acceptable carriers that can be used in the present invention include (but are not limited to) a) diluent; b) lubricant; c) binder; d) disintegrant; e) absorbent, coloring agent, and flavoring And sweeteners; f) emulsifiers or dispersants; and/or g) agents that enhance the absorption of the compound.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least one of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, formula IVd, formula Va, and formula Vb of the present invention
  • a compound of formula Vc or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (such as hydrate), isotope label, metabolite or prodrug thereof Or a compound of formula Vc or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (such as hydrate), isotope label, metabolite or prodrug thereof.
  • the aforementioned pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  • the above-mentioned pharmaceutical composition further comprises one or more additional pharmaceutically active ingredients (such as pharmaceutically active ingredients for preventing and/or treating SHP2-related diseases).
  • the above-mentioned pharmaceutical composition also includes one or more pharmaceutically acceptable carriers and one or more additional pharmaceutically active ingredients (such as for the prevention and/or treatment of SHP2). Active ingredients of drugs for related diseases).
  • the above-mentioned pharmaceutical composition may act systemically and/or locally.
  • they can be administered by suitable routes, such as parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular , Inhalation route or all methods well known to those skilled in the medical field.
  • suitable routes such as parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular , Inhalation route or all methods well known to those skilled in the medical field.
  • the above-mentioned pharmaceutical composition can be administered in combination with at least one other therapeutic agent having a therapeutic effect on a disease or condition.
  • the above-mentioned administration route can be achieved by a suitable dosage form.
  • the above-mentioned pharmaceutical composition may contain 0.01 mg to 1000 mg of at least one of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc,
  • the present invention also provides a preparation method of the above-mentioned pharmaceutical composition or its corresponding preparation form, which comprises combining at least one of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, and formula of the present invention.
  • drug refers to a combination product comprising a therapeutic agent, optionally other therapeutic agents, and optionally packaging and/or instructions.
  • pharmaceutical active ingredient refers to a chemical entity that can effectively prevent and/or treat the target disease or one or more symptoms thereof.
  • the present invention provides a medicine, which comprises:
  • the instructions list the relevant information of the pharmaceutical active ingredients or pharmaceutical compositions in the above-mentioned drugs, and preferably specifically list the indications for which the pharmaceutical active ingredients or pharmaceutical compositions are allowed to be used.
  • the instructions are printable materials (such as paper, plastic, metal foil, adhesive paper, etc.) on which the required information can be formed (such as printing or application).
  • the above-mentioned medicine may contain 0.01 mg to 1000 mg of at least one formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, and formula IVd of the present invention.
  • the present invention also provides a method for preparing the above-mentioned medicine, which comprises combining at least one of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, formula IVd, formula Va,
  • the compound of the present invention can show a strong inhibitory effect on SHP2, and can be used as an SHP2 inhibitor. Therefore, the present invention provides a compound of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, formula IVd, formula Va, formula Vb or formula Vc of the present invention, or a pharmaceutically acceptable compound thereof Accepted salts, esters, stereoisomers, tautomers, polymorphs, solvates (such as hydrates), isotope markers, metabolites or prodrugs, or pharmaceutical compositions, or drugs, used as Use of SHP2 inhibitors.
  • the present invention also provides the compound of formula I, formula IIa, formula IIb, formula IIIa, formula IIIb, formula IVa, formula IVb, formula IVc, formula IVd, formula Va, formula Vb or formula Vc of the present invention or its pharmaceutically Acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates (such as hydrates), isotope markers, metabolites or prodrugs, or pharmaceutical compositions, or drugs, in Use in preparing medicines for preventing and/or treating diseases or disorders mediated at least in part by SHP2 (or SHP2 related diseases, SHP2 enzyme related diseases or SHP2 phosphatase related diseases, especially cancer).
  • SHP2 or SHP2 related diseases, SHP2 enzyme related diseases or SHP2 phosphatase related diseases, especially cancer.
  • disease or disorder mediated at least in part by SHP2 refers to a disease that includes at least a part of SHP2-related factors in its pathogenesis, especially diseases that are sensitive or responsive to SHP2 phosphatase inhibition.
  • Diseases include (but are not limited to) tumor diseases.
  • the above-mentioned tumor-like disorders include (but are not limited to) breast cancer, colorectal cancer, colon cancer, lung cancer (including small cell lung cancer, non-small cell lung cancer and bronchioloalveolar cancer), prostate cancer, Cholangiocarcinoma, bone cancer, bladder cancer, head and neck cancer, kidney cancer, liver cancer, gastrointestinal tissue cancer, esophageal cancer, ovarian cancer, pancreatic cancer, skin cancer, testicular cancer, thyroid cancer, uterine cancer, cervical cancer, vulvar cancer, multiple Myeloma, lymphoma and leukemia, such as chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL) and chronic myelogenous leukemia (CML).
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • the present invention provides a method for preventing and/or treating diseases (especially cancer) mediated at least in part by SHP2, which comprises the following steps: a preventive and/or therapeutically effective amount of at least one formula of the present invention I.
  • Formula IIa, Formula IIb, Formula IIIa, Formula IIIb, Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula Va, Formula Vb, or Formula Vc Compounds or their pharmaceutically acceptable salts, esters, stereoisomers , Tautomers, polymorphs, solvates (such as hydrates), isotope markers, metabolites or prodrugs, or pharmaceutical compositions, or drugs, administered to individuals in need thereof.
  • the term "effective amount" refers to a dose that can induce a biological or medical response in cells, tissues, organs, or organisms (for example, an individual), and is sufficient to achieve the desired preventive and/or therapeutic effects.
  • the dosage regimen can be adjusted to provide the best response. For example, it can be administered in a single dose, can be administered in divided doses over time, or can be administered after a proportional reduction or increase in the dose according to the actual situation. It is understandable that, for any particular individual, the specific dosing regimen should be adjusted according to needs and the professional judgment of the administering personnel. In addition, it is necessary to distinguish between preventive applications and therapeutic applications. In prophylactic applications, relatively low doses are usually given over a long period of time at relatively long intervals. In therapeutic applications, relatively high doses are usually given at relatively short intervals until the progression of the disease is delayed or stopped, preferably until the individual shows partial or complete amelioration of symptoms.
  • the effective amount of the compound sufficient to achieve the preventive and/or therapeutic effect is from about 0.001 mg/kg body weight/day to about 10,000 mg/kg body weight/day.
  • prevention includes inhibiting and delaying the onset of the disease, and includes not only the prevention before the development of the disease, but also the prevention of the recurrence of the disease after the treatment.
  • treatment refers to reversing, alleviating or eliminating the targeted disease or condition. If the subject receives a therapeutic amount of the compound of the present invention or its pharmaceutically acceptable form or the pharmaceutical composition of the present invention, at least one of the indicators and symptoms of the subject exhibits observable and/or possible Remission and/or improvement detected indicate that the subject has been successfully "treated". It is understandable that treatment not only includes complete treatment, but also includes not achieving complete treatment, but achieving some biological or medical-related results.
  • treatment means that the compound of the present invention or its pharmaceutically acceptable form or the pharmaceutical composition of the present invention can achieve at least one of the following effects: (1) When experiencing or showing disease pathology or symptoms Inhibition of disease (ie, prevent the further development of pathology and/or symptomatology) in animals with scientific studies; (2) Improve disease (ie, reverse pathology and/or symptomatology ).
  • administration refers to the application of pharmaceutical active ingredients (such as the compound of the present invention) or a pharmaceutical composition containing the pharmaceutical active ingredient (such as the pharmaceutical composition of the present invention) to an individual or its cells, tissues, organs, biological fluids, etc. , In order to make the active ingredient of the medicine or the medicine composition come into contact with the individual or its cells, tissues, organs, biological fluids and other parts.
  • pharmaceutical active ingredients such as the compound of the present invention
  • a pharmaceutical composition containing the pharmaceutical active ingredient such as the pharmaceutical composition of the present invention
  • the term "has a need for it" refers to the doctor or other nursing staff's judgment on the individual's needs or the benefit from the prevention and/or treatment process. This judgment is based on the doctor's or other nursing staff's respective expertise Kinds of factors.
  • the term "individual" refers to a human or non-human animal.
  • the individuals of the present invention include individuals (patients) suffering from diseases and/or disorders and normal individuals.
  • the non-human animals of the present invention include all vertebrates, such as non-mammals, such as birds, amphibians, reptiles, etc., and mammals, such as non-human primates, domestic animals, and/or domesticated animals (such as sheep, dogs, Cats, cows, pigs, etc.).
  • the present invention also provides a combined method for preventing and/or treating diseases or conditions (especially cancer) mediated at least in part by SHP2, which includes: a) the present invention for preventing and/or treating at least partly Methods for diseases or conditions mediated by SHP2 (especially cancer); and b) additional therapies.
  • the additional therapy in the aforementioned combination method includes (but is not limited to) radiotherapy, chemotherapy, immunotherapy, or any combination thereof.
  • the compound, pharmaceutical composition or medicine of the present invention can be administered before, during or after the additional therapy.
  • the implementation of the additional therapy and the administration of the compound, pharmaceutical composition or medicine of the present invention can be carried out at the same time, or can be carried out in close connection before and after, or can be carried out at intervals of time.
  • the method and order of administration can be selected according to the specific treatment situation. And adjustment.
  • the reagents or instruments used in the examples where the manufacturer is not indicated are all conventional products that can be purchased commercially. If no specific conditions are specified, the normal conditions or the conditions recommended by the manufacturer shall be used.
  • the term "room temperature” as used herein refers to 20°C ⁇ 5°C.
  • the term “about” used herein refers to the numerical value or numerical range and the error range acceptable to those skilled in the art of the numerical value or numerical range, for example, the error range It is ⁇ 10%, ⁇ 5%, ⁇ 4%, ⁇ 3%, ⁇ 2%, ⁇ 1%, ⁇ 0.5%, etc. Unless otherwise stated, concentrations are by weight and ratios (including percentages) are by moles.
  • the measuring instrument of nuclear magnetic resonance uses Bruker 400MHz nuclear magnetic resonance instrument.
  • the measuring solvent is deuterated methanol (CD 3 OD), deuterated chloroform (CDCl 3 ), hexadeuterated dimethyl sulfoxide (DMSO-d) 6 ) or deuterated water (D 2 O), the internal standard substance is tetramethylsilane (TMS).
  • the reaction is monitored by thin-layer chromatography or liquid chromatography-mass spectrometry.
  • the mobile phase system includes (but is not limited to) dichloromethane-methanol system, n-hexane-ethyl acetate system and petroleum ether-ethyl acetate system.
  • the volume ratio can be adjusted according to the polarity of the compound, or adjusted by adding a small amount of triethylamine.
  • Thin-layer chromatography uses GF 254 silica gel plate (0.4 ⁇ 0.5nm, Yantai Jiangyou Silica Gel Development Co., Ltd.).
  • the measuring instrument of preparative high performance liquid chromatography uses Shimadzu LC-8A preparative liquid chromatograph (YMC, ODS, 250 ⁇ 20mm ⁇ 5 ⁇ m C18 chromatographic column).
  • the mass spectrometer used Agilent 6120B mass spectrometer, and the ion source was an electrospray ion source (ESI).
  • ESI electrospray ion source
  • the mobile phase system includes (but is not limited to) the dichloromethane-methanol system and the n-hexane-ethyl acetate system.
  • the volume ratio of the solvent can be based on the compound The polarity is different to adjust, or add a small amount of triethylamine, etc. to adjust.
  • reaction temperature in the following examples is room temperature (20-30°C).
  • reagents in the following examples were purchased from Acros Organics, Aldrich Chemical, Nanjing Yaoshi Technology, Shanghai Shuya Pharmaceutical Technology, etc.
  • Fragment preparation example 1 Preparation of 2-fluoro-1-(4-methylpiperidin-4-yl)ethylamine (Compound A-4).
  • compound B1-1 (5.00 g, 20.72 mmol) was added to THF (50 mL), the temperature was lowered to -78 °C, LDA (2M tetrahydrofuran solution, 34.19 mL) was added dropwise, and the temperature was maintained at this temperature and stirred for 10 minutes after dropping. minute. Then TMSCl (7.43g, 68.37mmol) was dropped into the system, the temperature was controlled at -78°C, and after the dropping, stirred for 1 hour.
  • the reaction solution was poured into saturated sodium bicarbonate aqueous solution, extracted with methyl tert-butyl ether twice, the organic phases were combined, washed with saturated brine, dried with anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness to obtain an oily substance.
  • the oil was dissolved in THF (25 mL), sodium carbonate (4.39 g, 41.44 mmol) was added, the temperature was lowered to 0° C., NBS (4.06 g, 22.79 mmol) was added in batches, and then the temperature was raised to 25° C., and the reaction was carried out for 4 hours.
  • Fragment preparation example 2 Preparation of 2-methoxy-1-(4-methylpiperidin-4-yl)ethylamine (Compound A-5).
  • Step 2 Preparation of tert-butyl 4-(1-hydroxy-2-methoxyethyl)-4-methylpiperidine-1-carboxylate (Compound B2-3):
  • Step 5 Preparation of tert-butyl 4-(1-amino-2-methoxyethyl)-4-methylpiperidine-1-carboxylate (Compound B2-6):
  • Step 6 Preparation of 2-methoxy-1-(4-methylpiperidin-4-yl)ethylamine (Compound A-5):
  • Example 1 (3-((3S,4S)-4-amino-3-methyl-2-oxo-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(2-hydroxymethyl)pyrrolidin-1-yl)pyridin-4-ylsulfanyl)-5-methylpyrazin-2-yl)methanol (compound 1).
  • the sixth step (3-((3S,4S)-4-amino-3-methyl-2-oxo-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro -2-(2-(Hydroxymethyl)pyrrolidin-1-yl)pyridin-4-ylsulfanyl)-5-methylpyrazin-2-yl)methanol (compound 1) of trifluoroacetate preparation:
  • Example 2 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(2-hydroxymethyl)pyrrolidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 2).
  • Step 2 to Step 4 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6- Preparation of (3-chloro-2-(2-hydroxymethyl)pyrrolidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (compound 2):
  • the fourth step (1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)- Preparation of 6-(fluoromethyl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)pyrrolidin-2-yl)methanol (compound 22):
  • Example 5 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl ) Preparation of-6-(hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)acetonitrile (compound 11).
  • Example 6 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(hydroxymethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 12).
  • Example 7 (3-((S)-4-amino-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro-2-(2-( Preparation of hydroxymethyl)pyrrolidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 21).
  • the crude compound 21 was synthesized by a method similar to that described in the first to fourth steps in Example 2
  • the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetic acid salt of compound 21.
  • Example 8 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(hydroxymethyl)pyrrolidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 37).
  • Example 9 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-((S)-2-(hydroxymethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 33).
  • Example 10 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl ) Preparation of-6-(hydroxymethyl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)pyrrolidin-3-yl)acetonitrile (compound 14).
  • Example 11 2-(1-(5-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl )-6-(Hydroxymethyl)pyrazin-2-ylthio)-6-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (Compound 130).
  • the crude product of compound 130 was synthesized by a method similar to the second to sixth steps in Example 6.
  • the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain compound 130 The trifluoroacetate.
  • Example 12 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl )-6-(Hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (Compound 67).
  • Dissolve compound 12-4 (5.15g, 7.77mmol) in ethanol (80mL), add calcium chloride (3.45g, 31.10mmol), add sodium borohydride (1.18g, 31.10mmol) in batches, and slowly heat to 35 React at °C for 3 hours.
  • the sixth step 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl )-6-(Hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (Compound 67):
  • Example 13 (S)-2-(1-(4-(5-(4-Amino-oxa-8-azaspiro[4.5]decane-8-yl)-6-(hydroxymethyl) Preparation of pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (Compound 68).
  • Example 14 (S)-2-(1-(4-(5-(4-(1-amino-2-methoxyethyl)-4-methylpiperidin-1-yl)-6- Preparation of (hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 69).
  • the crude product of compound 69 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 69.
  • Example 15 1-((4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)- Preparation of 6-(hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-ylamino)methyl)cyclopropan-1-ol (compound 58).
  • the crude product of compound 58 was synthesized by a method similar to Example 6, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 58.
  • Example 16 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(fluoromethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 125).
  • the crude product of compound 125 was synthesized using a method similar to Example 6, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 125.
  • Example 17 1-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl ) Preparation of-6-(hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)ethanol (compound 70).
  • the crude product of compound 70 was synthesized using a method similar to Example 6, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 70.
  • Example 18 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl ) Preparation of-6-(hydroxymethyl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)pyrrolidin-2-yl)acetonitrile (compound 5).
  • the crude product of compound 5 was synthesized using a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetic acid salt of compound 5.
  • Example 19 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl ) Preparation of 6-(hydroxymethyl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)pyrrolidin-2-yl)acetamide (compound 4).
  • Example 20 (R)-2-(1-(4-(5-(3-amino-5-fluoro-3H-spiro[benzofuran-2,4'-piperidin]-1'-yl) Preparation of 6-(hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 97).
  • the crude product of compound 97 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 97.
  • Example 21 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(methoxymethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 71).
  • the crude product of compound 71 was synthesized using a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 71.
  • Example 22 (R)-2-(1-(4-(5-(3-amino-3H-spiro[benzofuran-2,4'-piperidine]-1'-yl)-6-( Preparation of hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 72).
  • the crude product of compound 72 was synthesized by a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 72.
  • Example 23 (S)-2-(1-(4-(5-(1-amino-1,3-dihydrospiro[indene-2,4'-piperidine]-1'-yl)-6 -(Hydroxymethyl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 73) preparation.
  • the crude product of compound 73 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 73.
  • Example 24 2-(1-(4-(5-(4-(1-aminoethyl)-4-methylpiperidin-1-yl)-6-(hydroxymethyl)pyrazine-2- Preparation of thiol)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 74).
  • the crude product of compound 74 was synthesized by a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 74.
  • Example 25 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro -2-(3-(Methylsulfonylmethyl)azetidin-1-yl)pyridin-4-ylthio)-5-methylpyrazin-2-yl)methanol (Compound 75) preparation.
  • the crude product of compound 75 was synthesized using a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 75.
  • Example 26 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(methylsulfonylmethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 76).
  • the crude product of compound 76 was synthesized by a method similar to the fourth and fifth steps of Example 5 and purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroethane of compound 76 Acid salt.
  • Example 27 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(hydroxymethyl)-3-methylazetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 77).
  • the crude product of compound 77 was synthesized by a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetic acid salt of compound 77.
  • Example 28 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl ) Preparation of pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 78).
  • the crude product of compound 78 was synthesized by a method similar to the fourth step and the fifth step of Example 5.
  • the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 78.
  • Example 29 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro -2-(3-Methyl-3-(methylsulfonylmethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 79) preparation.
  • Step 4 Preparation of 3-methyl-3-(methylsulfonylmethyl)azetidine (Compound 29-5):
  • Step 5 to Step 9 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6- (3-Chloro-2-(3-methyl-3-(methylsulfonylmethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol ( Compound 79) Preparation.
  • the crude product of compound 79 was synthesized by a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 79.
  • Example 30 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl )-6-(Hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)-2-methylpropionitrile (Compound 80) preparation.
  • the crude product of compound 80 was synthesized using a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 80.
  • Example 31 3-((4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)- Preparation of 6-(hydroxymethyl)-3-methylpyrazin-2-ylthio)-3-chloropyridin-2-ylamino)methyl)oxetan-3-ol (compound 81) .
  • the crude product of compound 81 was synthesized by a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 81.
  • Example 32 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl )-6-(hydroxymethyl)-3-methylpyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 82) Preparation.
  • the crude product of compound 82 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain compound 82 trifluoroacetic acid salt.
  • Example 33 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-(methoxymethyl)-3-methylazetidine-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 83) .
  • the crude product of compound 83 was synthesized by a method similar to the second to sixth steps of Example 6, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 83.
  • Example 34 2-(1-(4-(5-(4-(1-amino-2-fluoroethyl)-4-methylpiperidin-1-yl)-6-(hydroxymethyl)pyridine Preparation of azin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (Compound 84).
  • the crude product of compound 84 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 84.
  • Example 35 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(oxetan-3-ylmethoxy)pyridin-4-ylthio)pyrazin-2-yl)methanol (compound 59).
  • the crude product of compound 59 was synthesized using a method similar to Example 5, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 59.
  • Example 36 2-(1-(4-(3-Amino-5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane Preparation of -8-yl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 85).
  • Example 37 (1-(4-(3-Amino-5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8 Preparation of -yl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)pyrrolidin-2-yl)methanol (compound 86).
  • the crude product of compound 86 was synthesized according to a similar method in Example 36, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 86.
  • Example 38 (3S,4S)-8-(6-amino-5-(3-chloro-2-(3-(methylsulfonylmethyl)azetidin-1-yl)pyridine-4 Preparation of -ylthio)pyrazin-2-yl)-3-methyl-2-oxa-8-azaspiro[4.5]decane-4-amine (compound 87).
  • the crude product of compound 87 was synthesized according to a similar method in Example 36, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 87.
  • Example 39 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro Preparation of -2-(3-fluoro-3-(hydroxymethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 88).
  • the crude product of compound 88 was synthesized by a method similar to Example 6, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 88.
  • Example 40 2-(1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl )-6-(Hydroxymethyl)pyrazin-2-ylthio)-3-fluoropyridin-2-yl)azetidin-3-yl)propan-2-ol (Compound 90).
  • the crude product of compound 90 was synthesized by a method similar to Example 6, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% formic acid aqueous solution) to obtain compound 90.
  • the crude product of compound 107 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 107.
  • Example 42 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6-(3-chloro -2-(3-(fluoromethyl)-3-(hydroxymethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol (Compound 124) preparation.
  • Step 4 to Step 9 (3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6- (3-Chloro-2-(3-(fluoromethyl)-3-(hydroxymethyl)azetidin-1-yl)pyridin-4-ylthio)pyrazin-2-yl)methanol( Preparation of compound 124):
  • the crude product of compound 124 was synthesized using the similar methods described in the first to sixth steps of Example 12.
  • the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain compound 124 The trifluoroacetate.
  • Example 43 (S)-2-(1-(4-(5-(5-amino-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine] -1'-yl)-6-(hydroxymethyl)pyrazin-2-ylsulfanyl)-3-chloropyridin-2-yl)azetidine-3-yl)propan-2-ol (compound 89) Preparation.
  • the crude product of compound 89 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 89.
  • Example 44 (R)-2-(1-(4-(5-(3-amino-3H-spiro[furo[2,3-b]pyridine-2,4'-piperidine]-1' -Yl)-6-(hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)propan-2-ol (compound 101) preparation.
  • the crude product of compound 101 was synthesized using a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 101.
  • the crude product of compound 103 was synthesized by a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 103.
  • Example 46 (S)-2-(1-(4-(3-amino-5-(5-amino-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4' -Piperidine)-1'-yl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidine-3-yl)propan-2-ol (Compound 115) .
  • Example 47 1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)-6 -(Hydroxymethyl)pyrazin-2-ylthio)-3-chloropyridin-2-yl)-3-(hydroxymethyl)azetidine-3-carbonitrile (Compound 126).
  • Example 48 (1-(4-(5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decane-8-yl)pyridine Preparation of azin-2-ylthio)-3-chloropyridin-2-yl)-3-fluoroazetidin-3-yl)methanol (compound 138).
  • the crude product of compound 138 was synthesized by a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 138.
  • the crude product of compound 139 was synthesized by a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 139.
  • Example 50 (S)-2-(1-(4-(5-(5-amino-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine] -1'-yl)-6-(hydroxymethyl)-3-methylpyrazin-2-ylthio)-3-chloropyridin-2-yl)azetidin-3-yl)prop- Preparation of 2-alcohol (Compound 140).
  • the crude product of compound 140 was synthesized by a method similar to Example 13, and the crude product was purified by HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% trifluoroacetic acid aqueous solution) to obtain the trifluoroacetate salt of compound 140.
  • the synthesis of comparative compounds 1, 2 and TNO-155 The comparative compounds 1, 2 and TNO-155 were synthesized with reference to the methods described in WO2019075265, WO2018013597, and WO2015107495, respectively.
  • Test Example 1 SHP2 (protein phosphatase) in vitro enzymatic activity inhibition test.
  • Phosphatase human PTPN11 full-length sequence recombinant protein (SHP2), active (SignalChem);
  • Substrate 6,8-difluoro-4-methylumbelliferyl phosphate (DiFMUP) (Invitrogen);
  • IRS1_pY1172(dPEG8)pY1222(IRS1) BPS Bioscience
  • Termination reagent bpv(phen)(bpv)(Abcam).
  • IRS-1 concentration 0.5 ⁇ M
  • Buffer system 60mM Hepes pH7.2; 75mM NaCl; 75mM KCl; 0.05%surfactant P20; 1mM EDTA; 5mM DTT;
  • Reaction time between enzyme and substrate 30 minutes
  • Microplate reader parameters BMG PHERAstar fluorescence microplate reader, excitation wavelength 340nm, emission wavelength 450nm.
  • Test group Incubate the mixture of test compound and phosphatase SHP2 and the activation peptide IRS-1 in a buffer system at room temperature for 60 minutes, add the substrate DiFMUP to start the reaction, incubate at room temperature for 30 minutes, and then add bpv to stop the reaction.
  • the reaction plate is put into the microplate reader, and the fluorescence value of each well in the plate is read by the endpoint method.
  • Negative group Use 0.05% DMSO aqueous solution to replace the test compound, and the experimental method is the same as that of the test group.
  • Blank group Use 0.05% DMSO aqueous solution to replace the test compound without adding SHP2.
  • the experimental method is the same as that of the test group.
  • the inhibition rate (%) 100%-(fluorescence value of the test group-fluorescence value of the blank group)/(fluorescence value of the negative group-fluorescence value of the blank group) ⁇ 100%.
  • the curve was fitted according to the four-parameter model, and the half inhibitory concentration (IC 50 ) of the compound was calculated.
  • Example 1 Group IC 50 (nM) Group IC 50 (nM) Example 1 6.03 Example 25 7.00 Example 2 1.82 Example 26 0.70 Example 3 3.48 Example 27 4.10 Example 4 4.10 Example 28 2.45 Example 5 1.22 Example 29 1.68 Example 6 1.76 Example 30 4.00 Example 7 4.60 Example 32 3.70 Example 8 0.88 Example 33 4.10 Example 9 1.30 Example 37 2.40 Example 10 1.22 Example 38 5.30 Example 11 9.30 Example 39 1.40 Example 12 2.50 Example 40 4.13 Example 13 4.90 Example 41 8.40 Example 15 2.40 Example 42 10.19 Example 16 1.00 Example 43 2.60 Example 17 2.50 Example 44 2.49 Example 18 1.90 Example 45 4.10 Example 19 1.50 Example 46 2.60 Example 20 6.90 Example 47 3.20 Example 21 1.15 Example 48 2.20 Example 22 4.30 Example 49 2.30 Example 23 1.90 Example 50 4.84
  • the compound of the present invention showed strong inhibitory activity.
  • Test Example 2 KYSE-520 cell (human esophageal squamous cell carcinoma) proliferation activity inhibition test.
  • Reagent test kit Luminescence cell viability analysis kit (Promega).
  • Plating medium KYSE-520:1640+10% FBS;
  • Detection temperature room temperature
  • BMG PHERAstar FS detects chemiluminescence.
  • the cells were cultured in a medium containing 10% fetal bovine serum and placed at 37°C and 5% CO 2 for culture. Pour an appropriate amount of cells into a 96-well plate and culture overnight in an incubator to allow the cells to adhere to the wall. The next day, the medium was removed, complete medium containing the pre-diluted compound was added, and incubated at 37°C for 5 days. On the fifth day, add detection reagents to each well Chemiluminescence detects the relative luminescence unit (RLU) of each well.
  • RLU relative luminescence unit
  • the curve was fitted according to the four-parameter model, and the half inhibitory concentration (IC 50 ) of the compound was calculated.
  • the inhibitory activity of the compound of the present invention on the proliferation of KYSE-520 cells was determined according to the above method, and the results are shown in Table 2.
  • the compound of the present invention has strong cell proliferation inhibitory activity on KYSE-520 cells.
  • Test Example 3 Biochemical hERG inhibition test.
  • Kit Predictor TM hERG fluorescence polarization analysis kit (ThermoFisher), which contains:
  • Microplate reader parameters BMG PHERAstar FS fluorescence microplate reader.
  • Test group add different concentrations of test compounds to the microtiter plate containing hERG cell membrane, add a tracer with high hERG affinity to each well, incubate the microtiter plate at 25°C for 2 hours, use more
  • the functional microplate reader detects the change of fluorescence polarization (excitation wavelength: 540; emission wavelength: 590nm) value (mP).
  • Positive control group 30 ⁇ M compound E4031 was used instead of the test compound, and the experimental method was the same as that of the test group.
  • Blank control group Replace the test compound with hERG buffer and no hERG cell membrane.
  • the experimental method is the same as that of the test group.
  • Percent inhibition rate (%) (1-(mP of the test compound-mP of the positive control group)/(mP of the blank control group-mP of the positive control group)) ⁇ 100%.
  • test results show that the compound of the present invention has a low affinity with hERG, and the IC 50 that competes with the affinity tracer is greater than 10 ⁇ M.
  • Test Example 4 NCI-H358 cell (human non-small cell lung cancer cell) proliferation activity inhibition test.
  • Reagent test kit Luminescence cell viability analysis kit (Promega).
  • Plating medium NCI-H358:1640+10% FBS;
  • Detection temperature room temperature
  • BMG PHERAstar FS detects chemiluminescence.
  • the cells were cultured in a medium containing 10% fetal bovine serum and placed at 37°C and 5% CO 2 for culture. Pour an appropriate amount of cells into a 96-well plate and culture overnight in an incubator to allow the cells to adhere to the wall. The next day, the medium was removed, complete medium containing the pre-diluted compound was added, and incubated at 37°C for 5 days. On the fifth day, add detection reagents to each well Chemiluminescence detects the relative luminescence unit (RLU) of each well.
  • RLU relative luminescence unit
  • the curve was fitted according to the four-parameter model, and the half inhibitory concentration (IC 50 ) of the compound was calculated.
  • the inhibitory activity of the compound of the present invention on the proliferation of NCI-H358 cells was determined according to the above method, and the results are shown in Table 4.
  • Example number NCI-H358, IC 50 ( ⁇ M) Example number NCI-H358, IC 50 ( ⁇ M)
  • Example 2 0.040
  • Example 25 0.150
  • Example 5 0.034
  • Example 26 0.032
  • Example 6 0.074
  • Example 27 0.030
  • Example 12 0.018
  • Example 28 0.130
  • Example 13 0.260
  • Example 29 0.040
  • Example 15 0.040
  • Example 40 0.030
  • Example 43 0.008 Example 21 0.030
  • Example 44 0.050 Example 22 0.031
  • Example 46 0.028
  • Example 23 0.005
  • Example 47 0.023
  • test results show that the compound of the present invention has strong cell proliferation inhibitory activity on NCI-H358 cells.
  • Test Example 5 Biochemical CYP enzyme (cytochrome P450) inhibition test.
  • Test group add different concentrations of test compounds to the microtiter plate, add Luciferin-ME (100 ⁇ M), K 3 PO 4 (100mM) and CYP1A2 (0.01pmol/ ⁇ L) to each well, and pre-incubate at room temperature 10min, then add the NADPH regeneration system, react at room temperature for 30min, finally add an equal volume of detection buffer, incubate at room temperature for 20min, and then perform chemiluminescence detection.
  • Luciferin-ME 100 ⁇ M
  • K 3 PO 4 100mM
  • CYP1A2 CYP1A2 (0.01pmol/ ⁇ L
  • Negative control group The experimental method is the same as that of the test group, except that the test compound is not added.
  • Blank control group The experimental method is the same as that of the test group, except that the test compound is not added, and CYP1A2 Membrance (0.01pmol/ ⁇ L) is used instead of CYP1A2.
  • Test group add different concentrations of test compounds to the microtiter plate, add Luciferin-ME EGE (3 ⁇ M), K 3 PO 4 (100mM) and CYP2D6 (5nM) to each well, pre-incubate at room temperature for 10 min, Then add the NADPH regeneration system, react at 37°C for 30 minutes, and finally add an equal volume of detection buffer, incubate at room temperature for 20 minutes, and then perform chemiluminescence detection.
  • Luciferin-ME EGE 3 ⁇ M
  • K 3 PO 4 100mM
  • CYP2D6 5nM
  • Negative control group The experimental method is the same as that of the test group, except that the test compound is not added.
  • Blank control group The experimental method is the same as that of the test group, except that no test compound is added, and CYP2D6 Membrance (5nM) is used instead of CYP2D6.
  • Test group add different concentrations of test compounds to the microtiter plate, add Luciferin-IPA (3 ⁇ M), K 3 PO 4 (100mM) and CYP3A4 (2nM) to each well, pre-incubate at room temperature for 10 minutes, and then Add NADPH regeneration system, react at room temperature for 30 minutes, finally add an equal volume of detection buffer, incubate at room temperature for 20 minutes, and then perform chemiluminescence detection.
  • Luciferin-IPA 3 ⁇ M
  • K 3 PO 4 100mM
  • CYP3A4 2nM
  • Negative control group The experimental method is the same as that of the test group, except that the test compound is not added.
  • Blank control group The experimental method is the same as the test group, except that no test compound is added, and CYP3A4 Membrance (2nM) is used instead of CYP3A4.
  • Percent inhibition rate (1-(chemiluminescence signal value of the test compound concentration group-chemiluminescence signal value of the blank control group)/(chemiluminescence signal value of the negative control group-chemiluminescence signal value of the blank control group)) ⁇ 100%.
  • Test Example 6 SD rat pharmacokinetics (PK) study.
  • the compound of Example 12 and the comparative compound 2 were administered intravenously (IV) and intragastrically (PO) to male SD rats to investigate the pharmacokinetic properties.
  • the doses of IV and PO are 1mg/kg and 5mg/kg, respectively.
  • the solvent of IV is 5% DMSO + 5% Solutol (15-hydroxystearate polyethylene glycol ester) + 90% Saline (normal saline) , PO solvent is 0.5% MC (sodium methyl cellulose).
  • Blood was collected at different time points after IV and PO administration. The blood was anticoagulated with EDTA.K2, and a plasma sample was obtained after centrifugation and stored at -80°C.
  • Plasma samples were processed with precipitated protein and then analyzed by LC-MS/MS. Using WinNonlin 6.3 software, the non-compartmental model was used to calculate the pharmacokinetic parameters. The results are shown in Table 6 below.
  • Example 12 of the present invention achieves a higher exposure in rat plasma under the conditions of an injection dose of 1 mg/kg and an oral dose of 5 mg/kg, which is much higher than that of the comparative compound 2, and the clearance rate It is much lower than the comparative compound 2, showing excellent pharmacokinetic properties.
  • Test Example 7 Balb/c mouse pharmacokinetics (PK) study.
  • Example 12 The compound of Example 12, Comparative Compound 1, Comparative Compound 2, and TNO-155 were administered to female Balb/c mice by intravenous (IV) and intragastric (PO), respectively, to investigate the pharmacokinetic properties.
  • IV and PO are 1 mg/kg and 10 mg/kg, respectively, the solvent of IV is 5% DMSO+5% Solutol+90% Saline, and the solvent of PO is 0.5% MC (sodium methylcellulose). Blood was collected at different time points after IV and PO administration.
  • IV intravenous
  • PO intragastric
  • the blood of the comparative compound 1 and comparative compound 2 test groups were anticoagulated with EDTA.K2 and stored at -80°C; the blood of the TNO-155 test group was anticoagulated with EDTA.K2, and a plasma sample was obtained after centrifugation , Store at -80°C.
  • the blood or plasma samples are processed by the precipitated protein and then analyzed by LC-MS/MS.
  • WinNonlin 6.3 software the non-compartmental model was used to calculate the pharmacokinetic parameters. The results are shown in Table 7 below.
  • Example 12 of the present invention has a higher exposure in the mouse systemic circulation, a lower clearance rate and a longer half-life at a dosage of 1 mg/kg administered by injection and a dosage of 10 mg/Kg administered by oral administration. Significantly better than the comparative compounds 1, 2 and TNO-155, showing excellent pharmacokinetic properties.
  • Test Example 8 Beagle dog pharmacokinetics (PK) study.
  • Example 12 The compound of Example 12, comparative compound 1, comparative compound 2, and TNO-155 were administered to male beagle dogs via intravenous (IV) and intragastric administration (PO), respectively, to investigate the pharmacokinetic properties.
  • IV and PO are 0.5 mg/kg and 2.5 mg/kg, respectively, the solvent of IV is 5% DMSO+5% Solutol+90% Saline, and the solvent of PO is 0.5% MC (sodium methylcellulose).
  • Blood was collected at different time points after IV and PO administration. The blood was anticoagulated with EDTA.K2, and a plasma sample was obtained after centrifugation and stored at -80°C.
  • Plasma samples were processed with precipitated protein and then analyzed by LC-MS/MS. Using WinNonlin 6.3 software, the non-compartmental model was used to calculate the pharmacokinetic parameters. The results are shown in Table 8 below.
  • Example 12 of the present invention has a higher exposure in beagle plasma at a dosage of 0.5 mg/kg for injection and 2.5 mg/Kg for oral administration, and has a longer half-life and a lower half-life.
  • the clearance rate and comprehensive properties are better than comparative compound 1, comparative compound 2 and TNO-155, showing better pharmacokinetic properties.
  • Test Example 9 Tumor inhibition test on KYSE-520 transplanted tumor model.
  • a CDX xenograft animal model was constructed by subcutaneously inoculating KYSE-520 cells in nu-nu mice, and the tumor was administered orally once a day to evaluate the efficacy of different test compounds in vivo.
  • KYSE-520 cells were cultured in a monolayer in vitro, and the culture conditions were RPMI 1640 medium with 10% fetal calf serum and cultured in an incubator at 37°C and 5% CO 2 air. Use trypsin for digestion and passage twice a week. When the cells are in the exponential growth phase, the cells are collected and counted.
  • the KYSE-520 cell line cultured to the logarithmic growth phase was prepared into a single cell suspension (cells were resuspended in PBS, mixed with phenol red-free matrigel 1:1, and the final cell density was 5 ⁇ 10 7 /ml).
  • Nude mice were inoculated with 0.1ml and subcutaneously inoculated into the right axillary of nude mice to construct a nude mouse xenograft model of esophageal cancer. When the tumor grew to 130mm 3 , the nude mice were randomly divided into 5 groups for administration. The dosing schedule is shown in the table As shown in 9 (orally administered once a day for 21 days), the vehicle is 5% DMSO + 5% Solutol + 90% H 2 O.

Abstract

本发明属于药物化学领域,涉及取代吡嗪类化合物,包含其的药物组合物及其用途。具体地,本发明涉及一种具有式(I)结构的化合物,其表现出较好的SHP2抑制活性,可以作为高效的SHP2抑制剂,用于预防和/或治疗SHP2相关疾病。

Description

取代吡嗪类化合物,包含其的药物组合物及其用途
相关申请的引用
本发明要求2020年6月22日在中国提交的、名称为“取代吡嗪类化合物,包含其的药物组合物及其用途”、申请号为202010576572.X的发明专利申请以及2020年11月27日在中国提交的、名称为“取代吡嗪类化合物,包含其的药物组合物及其用途”、申请号为202011359022.9的发明专利申请的优先权,通过引用的方式将该专利申请的全部内容并入本文。
技术领域
本发明属于药物化学领域,涉及一种用作SHP2( src  homology 2 domain-containing phosphotyrosine  phosphatase  2)抑制剂的取代吡嗪类化合物,包含其的药物组合物,及其用于预防和/或治疗SHP2相关疾病的医药用途。
背景技术
SHP2是一种由基因PTPN11( protein  tyrosine  phosphatase  nonreceptor  11)编码的蛋白质酪氨酸磷酸酶(PTPs),是PTPs家族中的胞内非受体型成员,其催化蛋白质中的酪氨酸发生去磷酸化反应。SHP2具有两个SH2( src  homology  2)结构域(N端的N-SH2和C端的C-SH2)、一个催化结构域(PTP)和一个富含脯氨酸基团和酪氨酸磷酸化位点的C末端尾巴。这两个SH2结构域控制SHP2的亚细胞定位和功能调节。在未激活状态下,SHP2处于自抑制状态,N-SH2与PTP相互结合,从而抑制磷酸酶活性。然而,在生长因子、细胞因子或炎症因子,如PDGF( platelet- derived  growth  factor)和FGF( fibroblast  growth  factor)等的刺激下,磷酸化酪氨酸残基Tyr542(Y542)和Tyr580(Y580),并与N-SH2结合,使得PTP结构域的催化活性位点暴露,进而解除自抑制状态,激活了SHP2的PTP活性,从而引发由酪氨酸磷酸化所启动的信号传导级联反应。
SHP2在人体中广泛表达,参与Ras-Erk、PI3K-Akt、Jak-Stat、Met、FGFR、EGFR和NF-kB等多条信号通路,进而调节细胞增殖、分化、迁移、凋亡等生理学功能。SHP2的激活突变体与多种疾病的发生相关,如努南综合征(Noonan syndrome)、乳腺癌、黑色素瘤等,SHP2的过表达会增加慢性粒细胞白血病、肥大细胞增多症、恶性胶质瘤、肺癌和乳腺癌等癌症的风险,提示SHP2在不同类型癌症及癌症的不同发展阶段中均有广泛的作用。因此,有必要使用SHP2抑制剂来预防和/或治疗癌症和其他相关疾病。
目前,已发现嘧啶酮类、吡嗪类、羧酸类、醌类、喹啉类和吲哚类等化合物具有抑制SHP2活性的功能(例如,见WO2018013597A1),但本领域仍然亟需新的SHP2抑制剂,特别是具有高活性以及其他优异性质的SHP2抑制剂。
发明内容
发明要解决的问题
本发明通过大量的研究,惊喜地发现一系列取代吡嗪类化合物,该类化合物具有较高的SHP2抑制活性,其作为SHP2抑制剂可用于预防和/或治疗SHP2相关疾病,尤其是肿瘤疾病,展示出良好的应用前景。
用于解决问题的方案
第一方面,本发明提供了一种具有式I结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
Figure PCTCN2021099561-appb-000001
X选自化学键、S、O、NH和CH 2
R 1选自氢、羟基、卤素、氨基、C 1-6烷基、3-6元杂环烷基和C 3-6环烷基,其中所述烷基、杂环烷基和环烷基各自任选地被一个或多个选自卤素、羟基和氨基的取代基取代;
R 2选自氢、羟基、氨基、氰基、卤素、C 1-6烷基、3-6元杂环烷基和C 3-6环烷基,其中所述烷基、杂环烷基和环烷基各自任选地被一个或多个选自卤素、氧代基、羟基、氨基的取代基取代;
A选自C 6-12亚芳基、5-12元亚杂芳基、3-12元亚杂环烷基和C 3-8亚环烷基,其中所述亚芳基、亚杂芳基、亚杂环烷基和亚环烷基各自任选地被一个或多个R a取代;
若存在,每一个R a各自独立地选自氢、卤素、羟基、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、氰基、C 1-6烷基、C 3-6环烷基、C 2-6烯基、-S(=O) g-(C 1-6烷基)、-S(=O) gNH 2、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、3-12元杂环烷基、C 6-10芳基和5-12元杂芳基,其中所述烷基、环烷基、烯基、杂环烷基、芳基和杂芳基各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、-S(=O) g-(C 1-6烷基)和C 1-6烷基的取代基取代;
或者,当X为NH或CH 2时,任意一个R a和X以及与其连接的原子一起形成5-10元脂环、5-10元杂脂环、5-6元杂芳环或苯环,其中所述脂环、杂脂环、杂芳环和苯环各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代;
B选自-N(R b)-C(=O)-G-R d、-N(R b)-S(=O) g-G-R d、-N(R b)-C(=O)-N(R b)-G-R d、-N(R b)-S(=O) g-N(R b)-G-R d、-C(=O)-N(R b)-G-R d、-S(=O) g-N(R b)-G-R d、-S(=O) g-G-R d、-O-G-(3-10元杂环烷基)、-O-G-(C 3-6环烷基)、-O-(C 3-6亚环烷基)-G-H、-O-(3-10元亚杂环烷基)-G-H、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b、-G-N(R b)-C(=O)-R b、-G-N(R b)-C(=O)-OR b
Figure PCTCN2021099561-appb-000002
其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);
G选自C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、C 3-6亚环烷基、C 3-6亚环烯基、3-10元亚杂环烯基和3-10元亚杂环烷基,其中所述亚烷基、亚环烷基、亚环烯基、亚杂环烯基和亚杂环烷基各自任选地被一个或多个选自氢、卤素、氧代基、羟基、氰基、氨基、-OR b、-NHR b、-N(R b) 2、-N(R b)-C(=O)-R b、-C(=O)-NH 2、-C(=O)-N(R b) 2、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 3-6环烷基、C 3-6卤代环烷基、C 3-6羟环烷基、3-10元卤代杂环烷基、3-10元羟杂环烷基和3-10元杂环烷基的取代基取代;
R b选自氢、卤素、氨基、氰基、C 1-6烷基、C 3-6环烷基、C 3-6环烯基、3-10元杂环烯基和3-10元杂环烷基,其中所述烷基、环烷基、环烯基、杂环烯基和杂环烷基各自任选地被一个或多个选自氢、卤素、氧代基、羟基、氰基和氨基的取代基取代;
C选自C 6-12亚芳基、5-12元亚杂芳基、3-12元亚杂环烷基、C 3-12亚环烯基、3-12元亚杂环烯基和C 3- 8亚环烷基,其中所述亚芳基、亚杂芳基、亚杂环烷基、亚环烯基、亚杂环烯基和亚环烷基各自任选地被一个或多个R c取代;
若存在,每一个R c各自独立地选自氢、卤素、羟基、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、氰基、C 1-6烷基、C 2-6烯基、-S(=O) g-(C 1-6烷基)、-S(=O) gNH 2、氨基、-NH(C 1-6烷基)和-N(C 1-6烷基) 2,其中所述烷基和烯基各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、-S(=O) g-(C 1-6烷基)和C 1-6烷基的取代基取代;
Y选自C 1-6亚烷基、C 3-6亚环烷基、3-6元亚杂环烷基、C 3-6亚环烯基、3-6元亚杂环烯基、C 6-10亚芳 基、5-12元亚杂芳基、C 2-6亚烯基、-S(=O) g-(C 1-6亚烷基)-、-S(=O) g-N(R b)-、-C(=O)-(C 1-6亚烷基)-、-C(=O)-(C 3-6亚环烷基)-、-C(=O)-(3-6元亚杂环烷基)-和-C(=O)-,其中所述亚烷基、亚杂环烷基、亚环烯基、亚杂环烯基和亚环烷基各自任选地被一个或多个选自卤素、氧代基、羟基和氨基的取代基取代;
R d选自卤素、氨基、羟基、氰基、-S(=O) g-(C 1-6烷基)、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、-NH-(C 1-6烷基)、-NH(C 3-6环烷基)、-N(C 1-6烷基) 2、-NH-C(=O)-O-(C 1-6烷基)、-N(C 1-6烷基)-C(=O)-O-(C 1-6烷基)、-O-C(=O)-NH 2、-O-C(=O)-NH(C 1-6烷基)和-N(C 1-6烷基)-C(=O)-(C 1-6烷基),其中所述烷基和环烷基各自任选地被一个或多个选自卤素、氧代基、羟基、氨基和-O-(C 1-6烷基)的取代基取代;
R 3选自卤素、-OR z、羟基、氰基、-C(=O)-OR z、-C(=O)-N(R z) 2、C 1-6烷基、-(C 1-6亚烷基)-R z、-(C 1- 6亚烷基)-OR z、-(C 1-6亚烷基)-OH、-(C 1-6亚烷基)-N(R z) 2、C 3-6环烷基、3-12元杂环烷基、C 3-6环烯基、3-12元杂环烯基、C 2-6烯基、C 2-6炔基、C 6-12芳基、5-12元杂芳基、-S(=O) g-(C 1-6烷基)、氨基和-N(R z) 2,其中所述烷基、亚烷基、环烷基、杂环烷基、环烯基、杂环烯基、烯基、炔基、杂芳基和芳基各自任选地被一个或多个选自卤素、氰基、氧代基、-OR z、羟基、-N(R z) 2、-NHR z、氨基、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基、5-10元杂芳基、-C(=O)-OR z、-C(=O)-N(R z) 2、-C(=O)-NH 2和硝基的取代基取代;
若存在,每一个R z各自独立地选自氢、氰基、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、卤素、氰基、氧代基、硝基、羟基、-O-(C 1-6烷基)、C 1-6烷基、C 1-6卤代烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
R 4选自卤素、C 1-6烷基、C 3-6环烷基、-C(=O)-R z、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-10芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、卤素、氰基、羟基、氧代基、-O-(C 1-6烷基)、C 1-6烷基、C 1-6卤代烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
或者,R 3和R 4以及与其连接的原子一起形成4-8元脂环或4-8元杂脂环,其中所述脂环和杂脂环各自任选地被一个或多个R 4a取代,或者所述脂环和杂脂环各自任选地与一个或多个C 6-10芳环或5-12元杂芳环稠合,所述芳环和杂芳环各自任选地被一个或多个R 4a取代;
若存在,每一个R 4a各自独立地选自氢、卤素、氰基、-C(=O)H、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、-NH-C(=O)-(C 1-6烷基)、-NH-C(=O)-(C 3-8环烷基)、-NH-C(=O)-(3-8元杂环烷基)、氧代基、C 1-6烷基、C 3-6环烷基、C 3-6环烯基、3-12元杂环烯基、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、环烯基、杂环烯基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
若存在,每一个R 5各自独立地选自氢、卤素、氧代基、氰基、羟基、羧基、-C(=O)-NH 2、-C(=O)-O-(C 1-6烷基)、C 1-6烷基、C 3-6环烷基、C 2-6烯基、C 2-6炔基、-S(=O) g-(C 1-6烷基)、3-6元杂环烷基、C 6-10芳基和5-10元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、羟基、氧代基、卤素、氰基、-O-(C 1-6烷基)、-NH(C 1-6烷基)和-N(C 1-6烷基) 2的取代基取代;或者任意两个R 5以及与其连接的原子一起形成C 3-10脂环或4-12元杂脂环;
g为0、1或2;
n为0、1、2、3、4或5。
第二方面,本发明提供了具有式I结构的化合物的具体实例,其包括:
(1)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-基)甲醇;
(2)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(3)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(氟甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(4)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)乙酰胺;
(5)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)乙腈;
(6)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(7)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(甲氧基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(8)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-2-基)乙腈;
(9)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(10)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(甲基磺酰基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(11)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)乙腈;
(12)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(13)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(2-(3-(氨基甲基)氮杂环丁烷-1-基)-3-氯吡啶-4-基硫基)吡嗪-2-基)甲醇;
(14)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-3-基)乙腈;
(15)N-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基)-2-氰基乙酰胺;
(16)(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)哌啶-4-基)甲醇;
(17)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(18)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(2-(氟甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(19)2-(1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)乙腈;
(20)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(2-氯-6-(2-(羟基甲基)氮杂环丁烷-1-基)嘧啶-3-基硫基)吡嗪-2-基)甲醇;
(21)(3-((S)-4-氨基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(22)(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(氟甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇;
(23)(1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-3-氟-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇;
(24)(4S)-4-氨基-8-(5-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-3-(羟基甲基)吡嗪-2-基)-2-环丙基-2,8-二氮杂螺[4.5]癸烷-3-酮;
(25)1-((4S)-4-氨基-8-(5-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-3-(羟基甲基)吡嗪-2-基)-2,8-二氮杂螺[4.5]癸烷-2-基)乙酮;
(26)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)嘧啶-4-基)-5-甲基吡嗪-2-基)甲醇;
(27)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(2,3-二氯-4-(3-(羟基甲基)氮杂环丁烷-1-基)苯基)-5-甲基吡嗪-2-基)甲醇;
(28)(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇;
(29)2-(1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)丙-2-醇;
(30)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(31)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(2-(3-(氨基甲基)氮杂环丁烷-1-基)-3-氯吡啶-4-基硫基)吡嗪-2-基)甲醇;
(32)1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-甲酰胺;
(33)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-((S)-2-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(34)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(2-(2-(氨基甲基)吡咯烷-1-基)-3-氯吡啶-4-基硫基)吡嗪-2-基)甲醇;
(35)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(3-(羟基甲基)吡咯烷-1-基)嘧啶-4-基)吡嗪-2-基)甲醇;
(36)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(3-(羟基甲基)吡咯烷-1-基)嘧啶-4-基)-5-甲基吡嗪-2-基)甲醇;
(37)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(38)(1-(4-(5-(4-(1-氨基-2-氟乙基)-4-甲基哌啶-1-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇;
(39)(1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-3-氟-6-(羟基甲基)吡嗪-2-基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇;
(40)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(4-(羟基甲基)哌啶-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(41)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(4-(羟基甲基)哌啶-1-基)嘧啶-4-基)-5-甲基吡嗪-2-基)甲醇;
(42)N-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)-2-羟基-N,2-二甲基丙酰胺;
(43)(3-(4-(1-氨基-2-甲氧基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(2-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(44)(3-(4-(1-氨基-2-甲氧基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(45)(3-(4-(1-氨基-2-甲氧基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(46)(3-(4-(1-氨基乙基)-4-氟哌啶-1-基)-6-(3-氯-2-(3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(47)1-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-2-基)环丙腈;
(48)3-(4-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)哌嗪-1-基)-3-氧代丙腈;
(49)1-((4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基氧基)甲基)环丙醇;
(50)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3,6-二氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(51)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(2,3-二氯-6-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(52)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(5-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(53)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(2,3-二氯-6-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(54)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(5-氯-2-(2-(氟甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(55)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)吡啶-2-基)吡咯烷-2-基)乙腈;
(56)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(2-(3-(氟甲基)氮杂环丁烷-1-基)嘧啶-3-基硫基)吡嗪-2-基)甲醇;
(57)(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(2-(3-(氟甲基)氮杂环丁烷-1-基)嘧啶-3-基硫基)吡嗪-2-基)甲醇;
(58)1-((4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基氨基)甲基)环丙醇;
(59)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(氧杂环丁烷-3-基甲氧基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(60)(3-((S)-5-氨基-5,7-二氢螺[环戊烷并[b]吡啶-6,4'-哌啶]-1'-基)-6-(2-(2-(氟甲基)氮杂环丁烷-1-基)吡啶-3-基硫基)吡嗪-2-基)甲醇;
(61)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氟-2-(2-(氟甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(62)(1-(3-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)吡啶-2-基)吡咯烷-2-基)甲醇;
(63)2-(1-(3-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)吡啶-2-基)吡咯烷-3-基)乙腈;
(64)(5-氨基-3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(2-(2-(氟甲基)吡咯烷-1-基)吡啶-3-基硫基)吡嗪-2-基)甲醇;
(65)(1-(3-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)吡啶-2-基)吡咯烷-3-基)甲醇;
(66)1-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)环丙醇;
(67)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(68)(S)-2-(1-(4-(5-(4-氨基-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(69)(S)-2-(1-(4-(5-(4-(1-氨基-2-甲氧基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(70)1-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)乙醇;
(71)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲氧基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(72)(R)-2-(1-(4-(5-(3-氨基-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(73)(S)-2-(1-(4-(5-(1-氨基-1,3-二氢螺[茚-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(74)2-(1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(75)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-基)甲醇;
(76)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(77)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(羟基甲基)-3-甲基氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(78)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(79)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-甲基-3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(80)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)-2-甲基丙腈;
(81)3-((4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基氨基)甲基)氧杂环丁烷-3-醇;
(82)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(83)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲氧基甲基)-3-甲基氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(84)2-(1-(4-(5-(4-(1-氨基-2-氟乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(85)2-(1-(4-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(86)(1-(4-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇;
(87)(3S,4S)-8-(6-氨基-5-(3-氯-2-(3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-胺;
(88)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-氟-3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(89)(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(90)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氟吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(91)(S)-2-(1-(4-(5-(7-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(92)(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[c]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(93)(S)-2-(1-(4-(5-(4-氨基-2-氯-4,6-二氢螺[环戊二烯并[d]噻唑-5,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(94)(S)-2-(1-(4-(5-(4-氨基-4,6-二氢螺[环戊二烯并[d]噻唑-5,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(95)(S)-2-(1-(4-(5-(4-氨基-4,6-二氢螺[环戊二烯并[d]噁唑-5,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(96)(S)-2-(1-(4-(5-(6-氨基-4,6-二氢螺[环戊二烯并[d]噁唑-5,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(97)(R)-2-(1-(4-(5-(3-氨基-5-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(98)(R)-2-(1-(4-(5-(3-氨基-5-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氟吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(99)(R)-2-(1-(4-(5-(3-氨基-6-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(100)(R)-2-(1-(4-(5-(3-氨基-6-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氟吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(101)(R)-2-(1-(4-(5-(3-氨基-3H-螺[呋喃并[2,3-b]吡啶-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(102)(S)-2-(1-(4-(5-(7-氨基-5,7-二氢螺[环戊二烯并[c]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(103)(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(104)(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[c]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(105)(S)-2-(1-(4-(5-(7-氨基-5,7-二氢螺[环戊二烯[b]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(106)(R)-2-(1-(4-(5-(3-氨基-3H-螺[呋喃并[2,3-b]吡啶-2,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(107)(R)-2-(1-(4-(5-(3-氨基-5-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(108)(R)-2-(1-(4-(5-(3-氨基-6-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(109)(S)-2-(1-(4-(5-(4-氨基-2-氯-4,6-二氢螺[环戊二烯并[d]噻唑-5,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(110)(S)-2-(1-(4-(5-(4-氨基-4,6-二氢螺[环戊二烯并[d]噻唑-5,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(111)(S)-2-(1-(4-(5-(4-氨基-4,6-二氢螺[环戊二烯并[d]噁唑-5,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(112)(S)-2-(1-(4-(5-(6-氨基-4,6-二氢螺[环戊二烯并[d]噁唑-5,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(113)(S)-2-(1-(4-(3-氨基-5-(7-氨基-5,7-二氢螺[环戊二烯并[c]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(114)(S)-2-(1-(4-(3-氨基-5-(7-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(115)(S)-2-(1-(4-(3-氨基-5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(116)(S)-2-(1-(4-(3-氨基-5-(5-氨基-5,7-二氢螺[环戊二烯并[c]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(117)(S)-2-(1-(4-(3-氨基-5-(4-氨基-2-氯-4,6-二氢螺[环戊二烯并[d]噻唑-5,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(118)(S)-2-(1-(4-(3-氨基-5-(4-氨基-4,6-二氢螺[环戊二烯[d]噻唑-5,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(119)(R)-2-(1-(4-(3-氨基-5-(3-氨基-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(120)(R)-2-(1-(4-(3-氨基-5-(3-氨基-5-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(121)(R)-2-(1-(4-(5-(4-氨基-8-氮杂二螺[2.1.5.2]十二烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(122)(S)-2-(1-(4-(5-(1-氨基-2,2-二氟-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(123)(S)-2-(1-(4-(5-(4-氨基-8-氮杂-12-氧杂二螺[2.1.5.2]十二烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(124)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(氟甲基)-3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(125)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(氟甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(126)1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)-3-(羟基甲基)氮杂环丁烷-3-甲腈;
(127)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-乙基-3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇;
(128)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(氟甲基)吡嗪-2-基硫基)-3-氟吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(129)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-5-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(130)2-(1-(5-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-6-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(131)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氟吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(132)(R)-2-(1-(4-(3-氨基-5-(4-氨基-8-氮杂二螺[2.1.5.2]十二烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(133)(S)-2-(1-(4-(3-氨基-5-(1-氨基-2,2-二氟-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(134)(S)-2-(1-(4-(3-氨基-5-(4-氨基-8-氮杂-12-氧杂二螺[2.1.5.2]十二烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(135)(1-(4-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)-3-(氟甲基)氮杂环丁烷-3-基)甲醇;
(136)(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(氟甲基)-3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-基)甲醇;
(137)2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)嘧啶-2-基)氮杂环丁烷-3-基)丙-2-醇;
(138)(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)-3-氟氮杂环丁烷-3-基)甲醇;
(139)2-(1-(4-(5-(4-氨基-8-氮杂二螺[2.1.5.2]十二烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇;和
(140)(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇。
第三方面,本发明提供了一种药物组合物,其包含至少一种具有式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、Va、Vb或Vc结构的化合物或其药学上可接受的盐、酯、立体 异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,以及一种或多种药学上可接受的载体和/或一种或多种额外的药物活性成分。
第四方面,本发明提供了一种药品,其包含:
a)容器;
b)位于所述容器中的至少一种具有式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、Va、Vb或Vc结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药或者药物组合物;和
c)任选存在的包装和/或说明书。
第五方面,本发明提供了具有式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、Va、Vb或Vc结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,或者所述药物组合物,或者所述药品,其用作SHP2抑制剂,用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)。
第六方面,本发明提供了具有式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、Va、Vb或Vc结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,或者所述药物组合物,或者所述药品,用作SHP2抑制剂的用途。
第七方面,本发明提供了具有式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、Va、Vb或Vc结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,或者所述药物组合物,或者所述药品,在制备用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)的药物中的用途。
第八方面,本发明提供了一种用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)的方法,其包括下列步骤:将预防和/或治疗有效量的具有式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、Va、Vb或Vc结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,或者所述药物组合物,或者所述药品,施用于对其有需求的个体。
第九方面,本发明提供了一种用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)的组合方法,其包括:
a)所述用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)的方法;和
b)额外的方法。
发明的效果
本发明提供了一种结构新颖的取代吡嗪类化合物,其可以作为高活性的SHP2抑制剂,并且能够实现下述技术效果中的至少一种:
(1)针对SHP2的高抑制活性;
(2)优异的药物代谢动力学性质(如良好的生物利用度、合适的半衰期和作用持续时间);
(3)优异的安全性(较低的毒副作用、较宽的治疗窗)。
附图说明
图1示出了阳性化合物及实施例12的化合物在KYSE-520细胞移植瘤模型中体内药效比较。
图2示出了阳性化合物及实施例12的化合物在NCI-H358细胞移植瘤模型中体内药效比较。
具体实施方式
一般术语和定义
除非另有定义,本文中所使用的术语的含义与本领域技术人员通常所理解的相同。本文中所使用的技术意图是指本领域中通常所理解的技术,包括对于本领域技术人员显而易见的技术的变化或等效替换。虽然下列术语对于本领域技术人员容易理解,但仍然阐述如下,以便更好地解释本发明。
术语“包括”、“包含”、“具有”或“涉及”及其在本文中的其它变体形式是指包含性的或开放式的集合概念,且不排除其它未列举的元素或方法步骤。本领域技术人员应当理解,上述术语如“包括”涵盖 “由…组成”的含义。
术语“一个(种)或多个(种)”或者类似的表述“至少一个(种)”是指例如1、2、3、4、5、6、7、8、9、10个(种)或更多个(种)。
当数值范围的下限和上限被公开时,落入该范围中的任何数值或任何亚范围都表示被具体公开。特别地,本文中所公开的参数的每一个数值范围(例如,以“约a至b”,或同等的“大约a至b”,或同等的“约a-b”的形式)均应理解为涵盖其中的每一个数值和亚范围。例如,“C 1-6”应理解为涵盖其中的任意亚范围以及每一个点值,如C 2-5、C 3-4、C 1-2、C 1-3、C 1-4、C 1-5等,以及C 1、C 2、C 3、C 4、C 5、C 6等。又例如,“3-10元”应理解为涵盖其中的任意亚范围以及每一个点值,例如3-4元、3-5元、3-6元、3-7元、3-8元、3-9元、4-5元、4-6元、4-7元、4-8元、5-7元、5-8元、6-7元等,以及3、4、5、6、7、8、9、10元等。
术语“药学上可接受的盐”是指对生物体基本上无毒性的,本发明的化合物的盐。药学上可接受的盐通常包括(但不限于)本发明的化合物与药学上可接受的无机酸/有机酸/酸性氨基酸或无机碱/有机碱/碱性氨基酸反应而形成的盐,此类盐又被称为酸加成盐或碱加成盐。适合的酸加成盐实例包括(但不限于)乙酸盐。适合的盐的综述参见,例如,Jusiak,Soczewinski,et al.,Remington’s Pharmaceutical Sciences[M],Mack Publishing Company,2005和Stahl,Wermuth,Handbook of Pharmaceutical Salts:Properties,Selection,and Use[M],Wiley-VCH,2002。用于制备本发明的化合物的药学上可接受的盐的方法是本领域技术人员已知的。
术语“药学上可接受的酯”是指对生物体基本上无毒性的,在生物体体内水解成本发明的化合物或其盐的酯。药学上可接受的酯通常包括(但不限于)本发明的化合物与药学上可接受的羧酸或磺酸形成的酯,此类酯又被称为羧酸酯或磺酸酯。另外,本发明的化合物本身也可以是酯。
术语“异构体”是指因具有相同的原子数和原子类型而具有相同的分子量,但原子的空间排列或构型不同的化合物。
术语“立体异构体”(或称“旋光异构体”)是指由于具有至少一个手性因素(包括手性中心、手性轴、手性面等)而导致具有垂直的不对称平面,从而能够使平面偏振光旋转的稳定异构体。由于本发明的化合物存在可能导致立体异构的不对称中心以及其他化学结构,因此本发明也包括这些立体异构体及其混合物。由于本发明的化合物(或其药学上可接受的盐)包括不对称碳原子,因而能够以单一立体异构体形式、外消旋物、对映异构体和非对映异构体的混合物形式存在。通常,这些化合物能够以外消旋物的形式制备。然而,如果需要的话,可以将这类化合物制备或分离后得到纯的立体异构体,即单一对映异构体或非对映异构体,或者单一立体异构体富集化(纯度≥99%、≥98%、≥97%、≥96%、≥95%、≥90%、≥85%、≥80%、≥75%、≥70%、≥65%或≥60%)的混合物。如下文中所述,化合物的单一立体异构体是由含有所需手性中心的旋光起始原料合成制备得到的,或者是通过制备得到对映异构体产物的混合物之后再分离或拆分制备得到的,例如转化为非对映异构体的混合物之后再进行分离或重结晶、色谱处理、使用手性拆分试剂,或者在手性色谱柱上将对映异构体进行直接分离。具有特定立体化学的起始化合物既可以商购得到,也可以按照下文中描述的方法制备再通过本领域熟知的方法拆分得到。术语“对映异构体”是指彼此具有不能重叠的镜像的一对立体异构体。术语“非对映异构体”或“非对映体”是指彼此不构成镜像的旋光异构体。术语“外消旋混合物”或“外消旋物”是指含有等份的单一对映异构体的混合物(即两种R和S对映体的等摩尔量混合物)。术语“非外消旋混合物”是指含有不等份的单一对映异构体的混合物。除非另外指出,本发明的化合物的所有立体异构体形式都在本发明的范围之内。
本文中使用实线
Figure PCTCN2021099561-appb-000003
实楔形
Figure PCTCN2021099561-appb-000004
或虚楔形
Figure PCTCN2021099561-appb-000005
以描绘本发明的化合物的共价化学键。当使用实线以描绘键连至手性原子的键时,表示包括该手性原子处的所有可能的立体异构体(例如,特定的对映异构体、外消旋混合物等)。当使用实或虚楔形以描绘键连至手性原子的键时,表示存在所示的立体异构体。除非另外指明,本发明的化合物的立体异构体可以涵盖特定的对映异构体、非对映异构体、外消旋体或其任意比例的混合物。
术语“互变异构体”(或称“互变异构形式”)是指具有不同能量的,可通过低能垒互相转化的结构异构体。若互变异构是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异 构体(或称质子转移互变异构体)包括(但不限于)通过质子迁移来进行的互相转化,如酮-烯醇异构化、亚胺-烯胺异构化、酰胺-亚胺醇异构化、亚硝基-肟异构化等。除非另外指出,本发明的化合物的所有互变异构体形式都在本发明的范围之内。
术语“多晶型物”(或称“多晶型形式”)是指化合物或复合物的固体晶体形式。本领域技术人员可通过许多已知的方法获得分子的多晶型物。这些方法包括(但不限于)熔融重结晶、熔融冷却、溶剂重结晶、去溶剂化、快速蒸发、快速冷却、慢速冷却、汽相扩散和升华。另外,可用熟知的技术检测、分类和鉴定多晶型物,这些技术包括(但不限于)差示扫描量热法(DSC)、热重分析法(TGA)、X射线粉末衍射法(XRPD)、单晶X射线衍射法(SCXRD)、固态核磁共振(NMR)、红外光谱法(IR)、拉曼光谱法和扫描电镜术(SEM)等。本发明涵盖本发明的化合物的所有可能的结晶形式或多晶型物,其可以为单一的多晶型物或多种多晶型物以任意比例混合而得的混合物。
术语“溶剂化物”是指由本发明的化合物(或其药学上可接受的盐)与至少一种溶剂分子通过非共价分子间作用力结合而形成的物质。本发明的化合物可以溶剂化物的形式存在,其中包含作为晶格结构要素的极性溶剂。极性溶剂的量可以化学计量比或非化学计量比的形式存在。
术语“同位素标记物”是指将本发明的化合物中的特定原子替换为其同位素原子而形成的衍生化合物。除非另外指出,本发明的化合物包括H、C、N、O、F、P、S、Cl的各种同位素,如 2H(D)、 3H(T)、 13C、 14C、 13N、 15N、 17O、 18O、 18F、 31P、 32P、 34S、 35S、 36S、 37Cl和 125I。例如, 12C可被 13C或 14C替代; 1H可被 2H(D,氘)或 3H(T,氚)替代; 16O可被 18O替代等。
术语“代谢物”是指本发明的化合物经代谢后形成的衍生化合物,例如经过氧化、还原、水解、酰胺化、脱酰胺化、酯化、酶解等反应而产生。关于代谢的进一步信息参见Goodman and Gilman's:The Pharmacological Basis of Therapeutics[M],McGraw-Hill International Editions,1996。本发明涵盖本发明的化合物的所有可能的代谢物形式,即在施用本发明的化合物的个体体内形成的物质。化合物的代谢物可以通过所属领域的公知技术来鉴定,其活性可以通过试验来表征。
术语“前药”是指在施用于个体后能够直接或间接地提供本发明的化合物的衍生化合物。特别优选的衍生化合物或前药是在施用于个体时可以提高本发明的化合物的生物利用度的化合物(例如,更易吸收入血),或者促进母体化合物向作用位点(例如,淋巴***)递送的化合物。除非另外指出,本发明的化合物的所有前药形式都在本发明的范围之内,且各种前药形式是本领域已知的,例如参见T.Higuchi,V.Stella,Pro-drugs as Novel Drug Delivery Systems[J],American Chemical Society,Vol.14,1975。此外,本发明还涵盖含有保护基的本发明的化合物。在制备本发明的化合物的任何过程中,保护在任何有关分子上的敏感基团或反应基团可能是必需的和/或期望的,由此形成本发明的化合物的化学保护的形式。这可以通过常规的保护基实现,例如在T.W.Greene,P.G.M.Wuts,Protective Groups in Organic Synthesis[M],John Wiley & Sons,2006中描述的保护基。使用本领域已知的方法,在适当的后续阶段可以移除这些保护基。
术语“化学键”是指纯净物分子内或晶体内相邻两个或多个原子(或离子)间使其相互结合的强烈的作用力,主要包括共价键、离子键、金属键、配位键等。除非另有说明,以游离形式存在的本发明的化合物中的化学键大多为共价键。
在本文中单独或与其他基团组合使用时,术语“烷基”是指直链或支链的饱和脂肪族烃基。例如,本文中所使用的术语“C 1-6烷基”是指具有1-6个碳原子(如1、2、3、4、5或6个碳原子)的烷基(如甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基、正戊基、异戊基、新戊基、正己基等),其任选地被一个或多个(如1-3个)本文中所描述的取代基取代(如被卤素取代时,该基团为“C 1- 6卤代烷基”,如-CF 3、-C 2F 5、-CHF 2、-CH 2F、-CH 2CF 3、-CH 2Cl、-CH 2CH 2CF 3等;如被羟基取代时,该基团为“C 1-6羟烷基”,如-CH 2OH、-CH 2CH 2OH、-CH(OH)CH 3等)。
在本文中单独或与其他基团组合使用时,术语“亚烷基”是指直链或支链的二价饱和脂肪族烃基,其所连接的两个基团(或片段)既可以连接同一个碳原子,又可以连接不同的碳原子。例如,本文中所使用的术语“C 1-6亚烷基”是指具有1-6个碳原子的亚烷基(如亚甲基、1,1-亚乙基、1,2-亚乙基、1,2-亚丙基、1,3-亚丁基等),其任选地被一个或多个(如1-3个)本文中所描述的取代基取代(如被卤素取代时,该基团为“C 1-6卤代亚烷基”,如-CF 2-、-C 2F 4-、-CHF-等)。
在本文中单独或与其他基团组合使用时,术语“环烷基”是指单环或多环(如双环)的烷基(例如单环环烷基,如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基等;或双环环烷基,包括稠环、桥环或螺环,如十氢萘基、双环[2.2.1]庚基、螺[4.5]癸基等)。例如,本文中所使用的术语“C 3- 6环烷基”是指具有3-6个成环碳原子的环烷基,其任选地被一个或多个(如1-3个)本文中所描述的取代基取代(如被卤素取代时,该基团为“C 1-6卤代环烷基”,如2-氟环丙基、3-氯环丁基、4-溴环己基等;如被羟基取代时,该基团为“C 1-6羟环烷基”,如2-羟基环丙基、3-羟基环丁基、4-羟基环己基等)。
在本文中单独或与其他基团组合使用时,术语“亚环烷基”是指单环或多环(如双环)的二价烷基,其所连接的两个基团(或片段)既可以连接同一个成环碳原子,又可以连接不同的成环碳原子。例如,本文中所使用的术语“C 3-6亚环烷基”是指具有3-6个成环碳原子的亚环烷基,其任选地被一个或多个(如1-3个)本文中所描述的取代基取代(例如
Figure PCTCN2021099561-appb-000006
)。
在本文中单独或与其他基团组合使用时,术语“杂环烷基”是指饱和或非饱和的非芳香性的单环或多环(如双环)的基团,其环中具有一个或多个碳原子(如1、2、3、4、5、6、7、8或9个)以及一个或多个(如1、2、3或4个)各自独立地选自N、O、P和S的杂原子,例如一共具有3-15个(优选3-8个,更优选3-6个)环原子。该术语还涵盖下述情况,其中环中的C原子、N原子和/或P原子可以被氧代基(=O)取代和/或环中的S原子可以被1个或2个氧代基(=O)取代。杂环烷基中的环系可以是稠环、桥环或螺环体系。如果满足价键要求,杂环烷基可以通过环中的任意一个碳原子或杂原子连接至其他基团(或片段)。例如,本文中所使用的术语“3-6元杂环烷基”是指具有3-6个环原子(其中包含一个或多个杂原子)的杂环烷基,其任选地被一或多个(例如1至3个)本文中所描述的取代基取代(如环氧乙烷基、环硫乙烷基、环氮乙烷基、氮杂环丁烷基、氧杂环丁烷基、硫杂环丁烷基、四氢呋喃基、二氧杂环戊烷基、四氢噻吩基、吡咯烷基、吡咯烷酮基、咪唑烷基、吡唑烷基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫吗啉基、1,4-噻噁烷基、1,4-二氧六环基、1,4-二噻烷基、哌嗪基、三噁烷基、三噻烷基、噻嗪烷基等;如被卤素取代时,该基团为“3-6元卤代杂环烷基”,如3-氟吡咯烷-1-基等;如被羟基取代时,该基团为“3-6元羟杂环烷基”,如4-羟基四氢呋喃-2-基等)。
在本文中单独或与其他基团组合使用时,术语“亚杂环烷基”是指单环或多环(如双环)的二价基团,其环中具有一个或多个碳原子(如1、2、3、4、5、6、7、8或9个)以及一个或多个(如2、3或4个)各自独立地选自N、O、P和S的杂原子,其所连接的两个基团(或片段)既可以连接同一个成环碳原子,又可以分别连接成环碳原子和成环杂原子(如N原子或P原子)。例如,本文中所使用的术语“3-10元亚杂环烷基”是指具有3-10个环原子(其中包含一个或多个杂原子)的亚杂环烷基,其任选地被一或多个(如1-3个)本文中所描述的取代基取代(例如
Figure PCTCN2021099561-appb-000007
)。
在本文中单独或与其他基团组合使用时,术语“杂环烯基”是指单环或多环(如双环)的具有一个或多个碳-碳双键的基团,其环中具有一个或多个碳原子(如1、2、3、4、5、6、7、8或9个)以及一个或多个(如1、2、3或4个)各自独立地选自N、O、P和S的杂原子,例如一共具有3-15个(优选3-8个,更优选3-6个)环原子。该术语还涵盖下述情况,其中环中的C原子、N原子和/或P原子可以被氧代基(=O)取代和/或环中的S原子可以被1个或2个氧代基(=O)取代。杂环烯基中的环系可以是稠环、桥环或螺环体系。如果满足价键要求,杂环烯基可以通过环中的任意一个碳原子或杂原子连接至其他基团(或片段)。例如,本文中所使用的术语“3-6元杂环烯基”是指具有3-6个环原子(其中包含一个或多个杂原子)的杂环烷基,其任选地被一或多个(例如1至3个)本文中所描述的取代基取代。
在本文中单独或与其他基团组合使用时,术语“亚杂环烯基”是指单环或多环(如双环)的具有一个或多个碳-碳双键的二价基团,其环中具有一个或多个碳原子(如1、2、3、4、5、6、7、8或9个)以及一个或多个(如1、2、3或4个)各自独立地选自N、O、P和S的杂原子,例如一共具有3-15个(优选3-8个,更优选3-6个)环原子。该术语还涵盖下述情况,其中环中的C原子、N原子和/或P原子可以被氧代基(=O)取代和/或环中的S原子可以被1个或2个氧代基(=O)取代。
在本文中单独或与其他基团组合使用时,术语“芳基”是指具有共轭π电子***的单环或稠环的芳香族烃基。例如,本文中所使用的术语“C 6-12芳基”是指具有6-12个碳原子的芳基(如苯基、萘基等),其任选地被一个或多个本文中所描述的取代基取代(如被C 1-6烷基取代的甲苯基、被卤素取代的氯苯基等)。
在本文中单独或与其他基团组合使用时,术语“亚芳基”是指具有共轭π电子***的单环或稠环的二价芳香族烃基。例如,本文中所使用的术语“C 6-10亚芳基”是指具有6-10个碳原子的亚芳基,其任选地被一个或多个本文中所描述的取代基取代(如被C 1-6烷基取代的亚甲苯基、被卤素取代的亚氯苯基等)。
在本文中单独或与其他基团组合使用时,术语“杂芳基”是指具有共轭π电子***的单环或稠环的芳香族基团,其环中具有一个或多个碳原子(如1、2、3、4、5、6、9或10个碳原子)以及一个或多个(例如1、2、3或4个)各自独立地选自N、O、P和S的杂原子,例如一共具有5-12个(优选5-10个,更优选5、6、9或10个)环原子。如果满足价键要求,杂环烷基可以通过任意一个环原子连接至母体分子部分。如果满足价键要求,杂芳基可以通过环中的任意一个碳原子或杂原子(如N原子)连接至其他基团(或片段)。并且,杂芳基可以任选地进一步与苯稠合。例如,本文中所使用的术语“5-12元杂芳基”是指具有5-12个环原子的杂芳基(如呋喃基、噻吩基、吡咯基、噁唑基、噻唑基、咪唑基、异噁唑基、异噻唑基、吡唑基、噁二唑基、噻二唑基、***基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、喹啉基、异喹啉基、苯并呋喃基、异苯并呋喃基、苯并噻吩基、异苯并噻吩基、吲哚基、异吲哚基或其苯并衍生物等),其任选地被一个或多个本文中所描述的取代基取代(如被C 1-6烷基取代的甲基吡啶基、被卤素取代的氯吡啶基等)。
在本文中单独或与其他基团组合使用时,术语“亚杂芳基”是指具有共轭π电子***的单环或稠环的二价芳香族基团,其环中具有一个或多个碳原子(如1、2、3、4、5、6、9或10个碳原子)以及一个或多个(例如1、2、3或4个)各自独立地选自N、O、P和S的杂原子,例如一共具有5-12个(优选5-10个,更优选5、6、9或10个)环原子。例如,本文中所使用的术语“5-12元亚杂芳基”是指具有5-12个环原子的亚杂芳基,其任选地被一个或多个本文中所描述的取代基取代(如被C 1-6烷基取代的
Figure PCTCN2021099561-appb-000008
被卤素取代的
Figure PCTCN2021099561-appb-000009
等)。
如本文中单独或与其他基团组合使用时,术语“烯基”是指具有一个或多个碳-碳双键的直链或支链的脂肪族烃基。例如,本文中所使用的术语“C 2-6烯基”是指具有2-6个碳原子以及一个、两个或三个(优选一个)碳-碳双键的烯基(如乙烯基、1-丙烯基、2-丙烯基、2-丁烯基、3-丁烯基、2-戊烯基、3-戊烯基、4-戊烯基、2-己烯基、3-己烯基、4-己烯基、5-己烯基、2-甲基-2-丙烯基、4-甲基-3-戊烯基等),其任选地被一个或多个(如1-3个)本文中所描述的取代基取代。
在本文中单独或与其他基团组合使用时,术语“亚烯基”是指具有一个或多个碳-碳双键的直链或支链的二价脂肪族烃基,其所连接的两个基团(或片段)既可以连接同一个碳原子,又可以连接不同的碳原子。例如,本文中所使用的术语“C 2-6亚烯基”是指具有2-6个碳原子的亚烯基(如
Figure PCTCN2021099561-appb-000010
Figure PCTCN2021099561-appb-000011
等),其任选地被一个或多个(如1-3个)本文中所描述的取代基取代。
在本文中单独或与其他基团组合使用时,术语“环烯基”是指具有一个或多个碳-碳双键的单环或多环(如双环)的烯基(例如单环环烯基,如环丁烯基、环戊烯基、环己烯基、环庚烯基等)。例如, 本文中所使用的术语“C 3-6环烯基”是指具有3-6个成环碳原子的环烯基,其任选地被一个或多个(如1-3个)本文中所描述的取代基取代(如
Figure PCTCN2021099561-appb-000012
)。
在本文中单独或与其他基团组合使用时,术语“亚环烯基”是指具有一个或多个碳-碳双键的单环或多环(如双环)的二价烯基,其所连接的两个基团(或片段)既可以连接同一个成环碳原子,又可以连接不同的成环碳原子。例如,本文中所使用的术语“C 3-6亚环烯基”是指具有3-6个成环碳原子的亚环烯基,其任选地被一个或多个(如1-3个)本文中所描述的取代基取代(如
Figure PCTCN2021099561-appb-000013
)。
如本文中单独或与其他基团组合使用时,术语“炔基”是指具有一个或多个碳-碳三键的直链或支链的脂肪族烃基。例如,如本文中所使用的术语“C 2-6炔基”是指具有2-6个碳原子以及一个、两个或三个(优选一个)碳-碳三键的炔基(如乙炔基、1-丙炔基、2-丙炔基、2-丁炔基、3-丁炔基、2-戊炔基、3-戊炔基、4-戊炔基、2-己炔基、3-己炔基、4-己炔基、5-己炔基等),其任选地被一个或多个(如1-3个)本文中所描述的取代基取代。
在本文中单独或与其他基团组合使用时,术语“亚炔基”是指具有一个或多个碳-碳三键的直链或支链的二价脂肪族烃基,其所连接的两个基团(或片段)分别连接不同的碳原子。例如,本文中所使用的术语“C 2-6亚炔基”是指具有2-6个碳原子的亚炔基(如
Figure PCTCN2021099561-appb-000014
等),其任选地被一个或多个(如1-3个)本文中所描述的取代基取代。
在本文中单独或与其他基团组合使用时,术语“脂环”或“脂烃环”是指单环或多环(包括稠环、桥环和螺环,如双环)的脂肪族烃类环系,包括环烷环、环烯环和环炔环等。
在本文中单独或与其他基团组合使用时,术语“杂脂环”或“杂脂烃环”是指单环或多环(包括稠环、桥环和螺环,如双环)的脂肪族烃类环系,其环中具有一个或多个碳原子(如1、2、3、4、5、6、7、8或9个)以及一个或多个(如1、2、3或4个)各自独立地选自N、O、P和S的杂原子,包括杂环烷环、杂环烯环和杂环炔环等。
在本文中单独或与其他基团组合使用时,术语“芳环”或“芳烃环”是指单环或多环(如双环)的芳香族烃类环系。
在本文中单独或与其他基团组合使用时,术语“杂芳环”或“杂芳烃环”是指单环或多环(如双环)的芳香族烃类环系,其环中具有一个或多个碳原子(如1、2、3、4、5、6、7、8或9个)以及一个或多个(如1、2、3或4个)各自独立地选自N、O、P和S的杂原子。
在本文中单独或与其他基团组合使用时,术语“卤素”是指氟(F)、氯(Cl)、溴(Br)或碘(I)。
在本文中单独或与其他基团组合使用时,术语“羟基”是指-OH。
在本文中单独或与其他基团组合使用时,术语“氰基”是指-CN。
在本文中单独或与其他基团组合使用时,术语“硝基”是指-NO 2
在本文中单独或与其他基团组合使用时,术语“氨基”是指-NH 2
在本文中单独或与其他基团组合使用时,术语“氧代基”是指=O。
本文中所使用的术语“各自独立地”是指结构中存在的取值范围相同或相近的至少两个基团(或片段)可以在特定情形下具有相同或不同的含义。例如,取代基X和取代基Y各自独立地为氢、卤素、羟基、氰基、烷基或芳基,则当取代基X为氢时,取代基Y既可以为氢,也可以为卤素、羟基、氰基、烷基或芳基;同理,当取代基Y为氢时,取代基X既可以为氢,也可以为卤素、羟基、氰基、烷基或芳基。
术语“取代”及其在本文中的其它变体形式是指所指定的原子上的一个或多个(如1、2、3或4个)原子或原子团(如氢原子)被其他等同物代替,条件是未超过所指定的原子或原子团在当前情况下的正常化合价,并且能够形成稳定的化合物。如果某一原子或原子团被描述为“任选地被……取代”,则其既可以被取代,又可以未被取代。除非另有说明,本文中取代基的连接位点可以来自取代基的任意适宜位置。当取代基中的连接键显示为穿过环系中相互连接的两个原子之间的化学键时,则表示该取 代基可以连接该环系中的任意一个成环原子。
通式化合物
本发明提供了一种式I化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
Figure PCTCN2021099561-appb-000015
X选自化学键、S、O、NH和CH 2
R 1选自氢、羟基、卤素、氨基、C 1-6烷基、3-6元杂环烷基和C 3-6环烷基,其中所述烷基、杂环烷基和环烷基各自任选地被一个或多个选自卤素、羟基和氨基的取代基取代;
R 2选自氢、羟基、氨基、氰基、卤素、C 1-6烷基、3-6元杂环烷基和C 3-6环烷基,其中所述烷基、杂环烷基和环烷基各自任选地被一个或多个选自卤素、氧代基、羟基和氨基的取代基取代;
A选自C 6-12亚芳基、5-12元亚杂芳基、3-12元亚杂环烷基和C 3-8亚环烷基,其中所述亚芳基、亚杂芳基、亚杂环烷基和亚环烷基各自任选地被一个或多个R a取代;
若存在,每一个R a各自独立地选自氢、卤素、羟基、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、氰基、C 1-6烷基、C 3-6环烷基、C 2-6烯基、-S(=O) g-(C 1-6烷基)、-S(=O) gNH 2、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、3-12元杂环烷基、C 6-10芳基和5-12元杂芳基,其中所述烷基、环烷基、烯基、杂环烷基、芳基和杂芳基各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、-S(=O) g-(C 1-6烷基)和C 1-6烷基的取代基取代;
或者,当X为NH或CH 2时,任意一个R a和X以及与其连接的原子一起形成5-10元脂环、5-10元杂脂环、5-6元杂芳环或苯环,其中所述脂环、杂脂环、杂芳环和苯环各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代;
B选自-N(R b)-C(=O)-G-R d、-N(R b)-S(=O) g-G-R d、-N(R b)-C(=O)-N(R b)-G-R d、-N(R b)-S(=O) g-N(R b)-G-R d、-C(=O)-N(R b)-G-R d、-S(=O) g-N(R b)-G-R d、-S(=O) g-G-R d、-O-G-(3-10元杂环烷基)、-O-G-(C 3-6环烷基)、-O-(C 3-6亚环烷基)-G-H、-O-(3-10元亚杂环烷基)-G-H、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b、-G-N(R b)-C(=O)-R b、-G-N(R b)-C(=O)-OR b
Figure PCTCN2021099561-appb-000016
其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);
G选自C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、C 3-6亚环烷基、C 3-6亚环烯基、3-10元亚杂环烯基和3-10元亚杂环烷基,其中所述亚烷基、亚环烷基、亚环烯基、亚杂环烯基和亚杂环烷基各自任选地被一个或多个选自氢、卤素、氧代基、羟基、氰基、氨基、-OR b、-NHR b、-N(R b) 2、-N(R b)-C(=O)-R b、-C(=O)-NH 2、-C(=O)-N(R b) 2、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 3-6环烷基、C 3-6卤代环烷基、C 3-6羟环烷基、3-10元卤代杂环烷基、3-10元羟杂环烷基和3-10元杂环烷基的取代基取代;
R b选自氢、卤素、氨基、氰基、C 1-6烷基、C 3-6环烷基、C 3-6环烯基、3-10元杂环烯基和3-10元杂环烷基,其中所述烷基、环烷基、环烯基、杂环烯基和杂环烷基各自任选地被一个或多个选自氢、卤素、氧代基、羟基、氰基和氨基的取代基取代;
C选自C 6-12亚芳基、5-12元亚杂芳基、3-12元亚杂环烷基、C 3-12亚环烯基、3-12元亚杂环烯基和C 3- 8亚环烷基,其中所述亚芳基、亚杂芳基、亚杂环烷基、亚环烯基、亚杂环烯基和亚环烷基各自任选地被一个或多个R c取代;
若存在,每一个R c各自独立地选自氢、卤素、羟基、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、氰基、C 1-6 烷基、C 2-6烯基、-S(=O) g-(C 1-6烷基)、-S(=O) gNH 2、氨基、-NH(C 1-6烷基)和-N(C 1-6烷基) 2,其中所述烷基和烯基各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、-S(=O) g-(C 1-6烷基)和C 1-6烷基的取代基取代;
Y选自C 1-6亚烷基、C 3-6亚环烷基、3-6元亚杂环烷基、C 3-6亚环烯基、3-6元亚杂环烯基、C 6-10亚芳基、5-12元亚杂芳基、C 2-6亚烯基、-S(=O) g-(C 1-6亚烷基)-、-S(=O) g-N(R b)-、-C(=O)-(C 1-6亚烷基)-、-C(=O)-(C 3-6亚环烷基)-、-C(=O)-(3-6元亚杂环烷基)-和-C(=O)-,其中所述亚烷基、亚杂环烷基、亚环烯基、亚杂环烯基和亚环烷基各自任选地被一个或多个选自卤素、氧代基、羟基和氨基的取代基取代;
R d选自卤素、氨基、羟基、氰基、-S(=O) g-(C 1-6烷基)、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、-NH-(C 1-6烷基)、-NH(C 3-6环烷基)、-N(C 1-6烷基) 2、-NH-C(=O)-O-(C 1-6烷基)、-N(C 1-6烷基)-C(=O)-O-(C 1-6烷基)、-O-C(=O)-NH 2、-O-C(=O)-NH(C 1-6烷基)和-N(C 1-6烷基)-C(=O)-(C 1-6烷基),其中所述烷基和环烷基各自任选地被一个或多个选自卤素、氧代基、羟基、氨基和-O-(C 1-6烷基)的取代基取代;
R 3选自卤素、-OR z、羟基、氰基、-C(=O)-OR z、-C(=O)-N(R z) 2、C 1-6烷基、-(C 1-6亚烷基)-R z、-(C 1- 6亚烷基)-OR z、-(C 1-6亚烷基)-OH、-(C 1-6亚烷基)-N(R z) 2、C 3-6环烷基、3-12元杂环烷基、C 3-6环烯基、3-12元杂环烯基、C 2-6烯基、C 2-6炔基、C 6-12芳基、5-12元杂芳基、-S(=O) g-(C 1-6烷基)、氨基和-N(R z) 2,其中所述烷基、亚烷基、环烷基、杂环烷基、环烯基、杂环烯基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自卤素、氰基、氧代基、-OR z、羟基、-N(R z) 2、-NHR z、氨基、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基、5-10元杂芳基、-C(=O)-OR z、-C(=O)-N(R z) 2、-C(=O)-NH 2和硝基的取代基取代;
若存在,每一个R z各自独立地选自氢、氰基、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、卤素、氰基、氧代基、硝基、羟基、-O-(C 1-6烷基)、C 1-6烷基、C 1-6卤代烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
R 4选自卤素、C 1-6烷基、C 3-6环烷基、-C(=O)-R z、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-10芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、卤素、氰基、羟基、氧代基、-O-(C 1-6烷基)、C 1-6烷基、C 1-6卤代烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
或者,R 3和R 4以及与其连接的原子一起形成4-8元脂环或4-8元杂脂环,其中所述脂环和杂脂环各自任选地被一个或多个R 4a取代,或者所述脂环和杂脂环各自任选地与一个或多个C 6-10芳环或5-12元杂芳环稠合,所述芳环和杂芳环各自任选地被一个或多个R 4a取代;
若存在,每一个R 4a各自独立地选自氢、卤素、氰基、-C(=O)H、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、-NH-C(=O)-(C 1-6烷基)、-NH-C(=O)-(C 3-8环烷基)、-NH-C(=O)-(3-8元杂环烷基)、氧代基、C 1-6烷基、C 3-6环烷基、C 3-6环烯基、3-12元杂环烷基、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、环烯基、杂环烯基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
若存在,每一个R 5各自独立地选自氢、卤素、氧代基、氰基、羟基、羧基、-C(=O)-NH 2、-C(=O)-O-(C 1-6烷基)、C 1-6烷基、C 3-6环烷基、C 2-6烯基、C 2-6炔基、-S(=O) g-(C 1-6烷基)、3-6元杂环烷基、C 6-10芳基和5-10元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、羟基、氧代基、卤素、氰基、-O-(C 1-6烷基)、-NH(C 1-6烷基)和-N(C 1-6烷基) 2的取代基取代;或者任意两个R 5以及与其连接的原子一起形成C 3-10脂环或4-12元杂脂环;
g为0、1或2;
n为0、1、2、3、4或5。
在本发明的一些实施方案中,上述式I化合物中的X选自化学键和S;优选地,X为S。
在本发明的一些实施方案中,上述式I化合物中的R 1选自氢、羟基、卤素、氨基、C 1-6烷基和C 3-6环烷基;优选地,R 1选自氢、卤素、氨基和C 1-6烷基(例如氢、卤素和C 1-6烷基);更优选地,R 1选自氢、 氟、氨基和甲基;进一步优选地,R 1选自氢、氟和甲基。
在本发明的一些实施方案中,上述式I化合物中的R 2选自氢、羟基、卤素、C 1-6烷基和C 3-6环烷基,其中所述烷基和环烷基各自任选地被一个或多个选自卤素和羟基的取代基取代;优选地,R 2选自氢、羟基、氟、羟基甲基(-CH 2OH)和氟甲基(-CH 2F);更优选地,R 2选自氢、羟基甲基和氟甲基;进一步优选地,R 2为羟基甲基。
在本发明的一些实施方案中,上述式I化合物中的A选自C 6-12亚芳基和5-12元亚杂芳基,其中所述亚芳基和亚杂芳基各自任选地被一个或多个R a取代;优选地,A选自亚苯基、亚吡啶基和亚嘧啶基,其中所述亚苯基、亚吡啶基和亚嘧啶基各自任选地被一个或多个R a取代;更优选地,A选自亚苯基和亚吡啶基,其中所述亚苯基和亚吡啶基各自任选地被一个或多个R a取代。
在本发明的一些实施方案中,式I中同时存在多个R a,每一个R a各自独立地选自氢、卤素、羟基、-O-(C 1-3烷基)、-O-(C 3-6环烷基)、氰基、C 1-3烷基、C 3-6环烷基、3-6元杂环烷基、氨基、-NH(C 1-3烷基)、-N(C 1-3烷基) 2、-S(=O) g-(C 1-3烷基)和-S(=O) gNH 2;优选地,每一个R a各自独立地选自氢、氟、氯、溴、羟基、甲氧基、环丙氧基、氰基、甲基、乙基、正丙基、异丙基、二氟甲氧基、三氟甲氧基、三氟甲基、二氟甲基、环丙基、吡咯烷基、吗啉基、氨基、甲氨基、二甲氨基、甲硫基、甲磺酰基和氨磺酰基;更优选地,每一个R a各自独立地选自氢、氟、氯、溴、羟基、氰基、甲基、乙基、正丙基、异丙基、环丙基、吡咯烷基、吗啉基、氨基、甲氨基和二甲氨基;进一步优选地,每一个R a各自独立地选自氟、氯、溴、氰基、甲基和氨基。
在本发明的一些实施方案中,式I中的X为NH或CH 2且同时存在多个R a,任意一个R a和X以及与其连接的原子一起形成5-7元脂环或5-7元杂脂环,优选5-6元脂环或5-6元杂脂环,其中所述脂环和杂脂环各自任选地被一个或多个选自氨基、氟、氯、氰基、氧代基、羟基、-O-(C 1-3烷基)、C 1-3卤代烷基和C 1- 3烷基的取代基取代,优选各自任选地被一个或多个选自氨基、氟、氯、氧代基、羟基、甲氧基和甲基的取代基取代。
在本发明的一些实施方案中,式I中的X为NH且同时存在多个R a,任意一个R a和X以及与其连接的原子一起形成吗啉环。
在本发明的一些实施方案中,上述式I化合物中的B选自-N(R b)-C(=O)-G-R d、-N(R b)-S(=O) 2-G-R d、-O-G-(3-10元杂环烷基)、-O-G-(C 3-6环烷基)、-O-(C 3-6亚环烷基)-G-H、-O-(3-10元亚杂环烷基)-G-H、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b、-G-N(R b)-C(=O)-R b、-G-N(R b)-C(=O)-OR b
Figure PCTCN2021099561-appb-000017
其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);优选地,B选自-N(R b)-C(=O)-G-R d、-O-G-(3-10元杂环烷基)、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b
Figure PCTCN2021099561-appb-000018
其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1- 6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基)。
在本发明的一些实施方案中,上述式I化合物中的B包含G,G选自C 1-6亚烷基、C 3-6亚环烷基和3-6元亚杂环烷基,所述亚烷基、亚环烷基和亚杂环烷基各自任选地被一个或多个选自氢、氟、氧代基、羟基、氰基、-OR b、C 1-6烷基、C 1-6卤代烷基和C 1-6羟烷基的取代基取代,优选各自任选地被一个或多个选自氢和氟的取代基取代;优选地,G为C 1-3亚烷基,所述亚烷基任选地被一个或多个选自氢、氟、氧代基、羟基、氰基、-OR b、C 1-6烷基、C 1-6卤代烷基和C 1-6羟烷基的取代基取代,优选任选地被一个或多个选自氢和氟的取代基取代。
在本发明的一些实施方案中,上述式I化合物中的B包含R b,R b选自氢、氟、氯、氨基、C 1-3烷基、环丙基和3-6元杂环烷基,其中所述烷基和杂环烷基各自任选地被一个或多个选自氢、氟、氧代基、羟基和氰基的取代基取代。
在本发明的一些实施方案中,上述式I化合物中的B包含C,C选自3-6元亚杂环烷基和C 3-6亚环烷基,其中所述亚杂环烷基和亚环烷基各自任选地被一个或多个R c取代;优选地,C选自亚吡咯烷基、亚氮杂环丁烷基、亚哌啶基和亚哌嗪基,其中所述亚吡咯烷基、亚氮杂环丁烷基、亚哌啶基和亚哌嗪基各自任选地被一个或多个R c取代。
在本发明的一些实施方案中,上述式I化合物中的B中同时存在多个R c,每一个R c各自独立地选自氢、氟、氰基、甲基、乙基、氟甲基、羟基甲基和羟基;优选地,每一个R c各自独立地选自氢、氟和羟基。
在本发明的一些实施方案中,上述式I化合物中的B包含Y,Y选自C 1-3亚烷基、C 3-6亚环烷基、3-6元亚杂环烷基、C 2-6亚烯基、-C(=O)-(C 1-3亚烷基)-、-C(=O)-(C 3-6亚环烷基)-、-C(=O)-(3-6元亚杂环烷基)-和-C(=O)-,其中所述亚烷基、亚杂环烷基和亚环烷基各自任选地被一个或多个选自氟、羟基和氨基的取代基取代;优选地,Y选自-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、亚环丙基和-C(=O)-CH 2-,其中所述-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、亚环丙基和-C(=O)-CH 2-各自任选地被一个或多个选自氟、羟基和氨基的取代基取代。
在本发明的一些实施方案中,上述式I化合物中的B包含R d,R d选自氟、氨基、羟基、氰基、-S(=O) 2-(C 1-6烷基)、-O-(C 1-6烷基)、-NH-C(=O)-O-(C 1-6烷基)、-O-C(=O)-NH 2、-O-C(=O)-NH(C 1-6烷基)和-N(C 1- 6烷基)-C(=O)-(C 1-6烷基),其中所述烷基任选地被一个或多个选自氟、氯、氧代基、羟基和-O-(C 1-6烷基)的取代基取代;优选地,R d选自氟、氨基、羟基、氰基、-S(=O) 2-CH 3、-O-CH 3、-NH-C(=O)-O-CH 3和-O-C(=O)-NH 2,其中所述-S(=O) 2-CH 3、-O-CH 3和-NH-C(=O)-O-CH 3各自任选地被一个或多个选自氟、氯、氧代基、羟基和-O-(C 1-6烷基)的取代基取代。
在本发明的一些实施方案中,上述式I化合物中的B选自
Figure PCTCN2021099561-appb-000019
Figure PCTCN2021099561-appb-000020
Figure PCTCN2021099561-appb-000021
-NH-C(=O)-CH 2-CN和-N(CH 3)-C(=O)-C(CH 3) 2-OH;优选地,B选自
Figure PCTCN2021099561-appb-000022
Figure PCTCN2021099561-appb-000023
Figure PCTCN2021099561-appb-000024
-NH-C(=O)-CH 2-CN和-N(CH 3)-C(=O)-C(CH 3) 2-OH。
在本发明的一些实施方案中,上述式I化合物中的R 3和R 4不相互连接,R 3选自氟、氯、溴、-OR z、羟基、氰基、C 1-6烷基、-(C 1-4亚烷基)-R z、-(C 1-4亚烷基)-OR z、C 3-6环烷基、3-8元杂环烷基(如3-6元杂环烷基)、C 2-4烯基、C 2-4炔基、苯基和5-6元杂芳基,其中所述烷基、亚烷基、环烷基、杂环烷基、烯基、炔基、苯基和杂芳基各自任选地被一个或多个选自氟、氯、氰基、氧代基、-OR z、羟基、-N(R z) 2、-NHR z、氨基、C 1-4烷基、C 3-6环烷基、3-6元杂环烷基、苯基、5-6元杂芳基的取代基取代;优选地,R 3选自氟、氯、-OR z、羟基、氰基、甲基、乙基、-CH 2-R z、-CH 2-OR z、环丙基、氧杂环丁烷基、-CH 2CH=CH 2、-CH=CH 2、-CH 2C≡CH、-C≡CH、吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基,其中所述吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基各自任选地被一个或两个选自氟、氯、氰基、-OR z、羟基、-N(R z) 2、-NHR z、氨基和吡唑基的取代基取代;更优选地,R 3选自氟、氯、羟基、氰基、甲基、乙基、环丙基、氧杂环丁烷基、吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基;更优选地,R 3选自氟、氯、甲基、乙基、环丙基和氧杂环丁烷基;进一步优选地,R 3选自氟和甲基。
在本发明的一些的实施方案中,上述式I化合物中的R 3包含多个R z,每一个R z各自独立地选自氰基、C 1-4烷基、C 3-6环烷基、3-6元杂环烷基、苯基、苄基和5-6元杂芳基,其中所述烷基、环烷基、杂环烷基、苯基、苄基和杂芳基各自任选地被一个或多个选自氨基、氟、氯、氰基、氧代基、羟基、-O-(C 1-3烷基)、甲基、乙基、二氟甲基、三氟甲基、C 3-6环烷基和苯基的取代基取代;优选地,每一个R z各自独立地选自氰基、甲基、乙基、异丙基、环丙基、环丁基、环戊基、环己基、吗啉基、哌嗪基、氧杂环丁烷基、四氢呋喃基、四氢吡喃基、吡咯烷基、氮杂环丁烷基、苯基、苄基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、噻唑基、三氮唑基、吡嗪基、嘧啶基和吡啶基,其中所述哌嗪基、氮杂环丁烷基、苯基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、噻唑基、三氮唑基、吡嗪基、嘧啶基和吡啶基各自任选地被一个或两个选自氟、氯、氰基、羟基、甲氧基、甲基和乙基的取代基取代。
在本发明的一些实施方案中,上述式I化合物中的R 3和R 4不相互连接,R 4选自卤素、C 1-6烷基、C 3- 6环烷基、-C(=O)-R z、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、芳基和杂芳基各自任选地被一个或多个选自氨基、氟、氯、氰基、羟基、氧代基、-O-(C 1-3烷基)、甲基、乙基、二氟甲基、三氟甲基和C 3-6环烷基的取代基取代;优选地,R 4选自氟、氯、甲基、乙基、异丙基、环丙基、环丁基、环氧乙烷基、氧杂环丁烷基、苯基、吡咯基、呋喃基、噻吩 基、咪唑基、噁唑基、噻唑基、吡唑基、吡啶基、哒嗪基和嘧啶基,其中所述苯基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、噻唑基、吡唑基、吡啶基、哒嗪基和嘧啶基各自任选地被一个或多个选自氨基、氟、氯、氰基、羟基、甲氧基、甲基、乙基、二氟甲基和三氟甲基的取代基取代;更优选地,R 4选自氟、氯、甲基、乙基、异丙基、环丙基、甲氧甲基、羟基甲基、氟甲基、二氟甲基、三氟甲基、氟乙基、氰甲基、羟基甲基、2-甲基呋喃基、噻唑基、吡啶基和嘧啶基;更优选地,R 4选自甲基、氟甲基、二氟甲基、三氟甲基、羟基甲基、甲氧甲基、氟乙基、氰甲基和羟基甲基;进一步优选地,R 4选自甲基、氟甲基、氟乙基、甲氧甲基、氰甲基、和羟基甲基。
在本发明的一些实施方案中,上述式I化合物中的R 3与R 4相互连接,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000025
选自以下任意一种:
Figure PCTCN2021099561-appb-000026
Figure PCTCN2021099561-appb-000027
优选以下任意一种:
Figure PCTCN2021099561-appb-000028
Figure PCTCN2021099561-appb-000029
Figure PCTCN2021099561-appb-000030
其中m为0、1或2;优选地,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000031
选自以下任意一种:
Figure PCTCN2021099561-appb-000032
Figure PCTCN2021099561-appb-000033
优选以下任意一种:
Figure PCTCN2021099561-appb-000034
Figure PCTCN2021099561-appb-000035
其中m为0、1或2;
更优选地,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000036
选自以下任意一种:
Figure PCTCN2021099561-appb-000037
Figure PCTCN2021099561-appb-000038
优选以下任意一种:
Figure PCTCN2021099561-appb-000039
在本发明的一些实施方案中,R 3和R 4以及与其连接的原子一起形成同时包含多个R 4a的环系,每一个R 4a各自独立地选自氢、氟、氯、氰基、-C(=O)H、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、氧代基、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 2-6烯基和C 2-6炔基,其中所述烷基、环烷基、杂环烷基、烯基和炔基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)和C 1-6烷基的取代基取代;优选地,每一个R 4a各自独立地选自氢、氟、氯、氧代基、甲基、乙酰基和环丙基,其中所述甲基和环丙基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)和C 1- 6烷基的取代基取代。
在本发明的一些实施方案中,上述式I化合物同时包含多个R 5,并且任意两个R 5不互相连接,每一个R 5各自独立地选自氢、氟、氯、氧代基、氰基、-C(=O)-O-(C 1-4烷基)和C 1-3烷基,其中所述烷基各自任选地被一个或多个选自氨基、羟基、氧代基和卤素的取代基取代;优选地,每一个R 5各自独立地选自氢、氟、氯、氰基、甲基和-C(=O)-O-CH 3;更优选地,每一个R 5各自独立地选自氢、氟和甲基;进一步优选地,每一个R 5均为氢。
在本发明的一些实施方案中,上述式I化合物同时包含多个R 5,任意两个不连接同一个原子的R 5以及与其连接的原子一起形成C 4-8脂环或4-8元杂脂环,优选C 4-6脂环或4-6元含氮杂脂环,或者任意两个连接同一个原子的R 5以及与其连接的原子一起形成C 3-6脂环或3-6元杂脂环,优选环丙烷环。
在本发明的一些实施方案中,g为0或2。
在本发明的一些实施方案中,n为0、1、2或3;优选地,n为0、1或2。
在本发明的一些实施方案中,上述式I化合物具有如式IIa或式IIb所示的结构,其中
Figure PCTCN2021099561-appb-000040
A、B、R 3和R 4如上文所定义。
在本发明的一些实施方案中,上述式I化合物具有如式IIIa或式IIIb所示的结构,其中
Figure PCTCN2021099561-appb-000041
A、B、R 3和R 4如上文所定义。
在本发明的一些实施方案中,上述式I化合物具有如式IVa、式IVb、式IVc或式IVd所示的结构,其中
Figure PCTCN2021099561-appb-000042
A、B、R 3和R 4如上文所定义。
在本发明的一些实施方案中,上述式I化合物具有如式Va、式Vb或式Vc所示的结构,其中
Figure PCTCN2021099561-appb-000043
A、B、R 3和R 4如上文所定义。
在本发明的一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自C 6-12亚芳基和5-12元亚杂芳基,其中所述亚芳基和亚杂芳基各自任选地被一个或多个R a取代;
若存在,每一个R a各自独立地选自氢、卤素、羟基、-O-(C 1-3烷基)、-O-(C 3-6环烷基)、氰基、C 1-3烷基、C 3-6环烷基、3-6元杂环烷基、氨基、-NH(C 1-3烷基)、-N(C 1-3烷基) 2、-S(=O) g-(C 1-3烷基)和-S(=O) gNH 2
B选自-N(R b)-C(=O)-G-R d、-N(R b)-S(=O) 2-G-R d、-O-G-(3-10元杂环烷基)、-O-G-(C 3-6环烷基)、-O-(C 3-6亚环烷基)-G-H、-O-(3-10元亚杂环烷基)-G-H、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b、-G-N(R b)-C(=O)-R b、-G-N(R b)-C(=O)-OR b
Figure PCTCN2021099561-appb-000044
其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);
G选自C 1-6亚烷基、C 3-6亚环烷基和3-6元亚杂环烷基,所述亚烷基、亚环烷基和亚杂环烷基各自任选地被一个或多个选自氢、氟、氧代基、羟基、氰基、-OR b、C 1-6烷基、C 1-6卤代烷基和C 1-6羟烷基的取代基取代,优选各自任选地被一个或多个选自氢和氟的取代基取代;
R b选自氢、氟、氯、氨基、C 1-3烷基、环丙基和3-6元杂环烷基,其中所述烷基和杂环烷基各自任选地被一个或多个选自氢、氟、氧代基、羟基和氰基的取代基取代;
C选自3-6元亚杂环烷基和C 3-6亚环烷基,其中所述亚杂环烷基和亚环烷基各自任选地被一个或多个R c取代;
若存在,每一个R c各自独立地选自氢、氟、氰基、甲基、乙基、氟甲基、羟基甲基和羟基;
Y选自C 1-3亚烷基、C 3-6亚环烷基、3-6元亚杂环烷基、C 2-6亚烯基、-C(=O)-(C 1-3亚烷基)-、-C(=O)-(C 3-6亚环烷基)-、-C(=O)-(3-6元亚杂环烷基)-和-C(=O)-,其中所述亚烷基、亚杂环烷基和亚环烷基各自任选地被一个或多个选自氟、羟基和氨基的取代基取代;
R d选自氟、氨基、羟基、氰基、-S(=O) 2-(C 1-6烷基)、-O-(C 1-6烷基)、-NH-C(=O)-O-(C 1-6烷基)、-O- C(=O)-NH 2、-O-C(=O)-NH(C 1-6烷基)和-N(C 1-6烷基)-C(=O)-(C 1-6烷基),其中所述烷基任选地被一个或多个选自氟、氯、氧代基、羟基和-O-(C 1-6烷基)的取代基取代;
R 3和R 4不相互连接时,
R 3选自氟、氯、溴、-OR z、羟基、氰基、C 1-6烷基、-(C 1-4亚烷基)-R z、-(C 1-4亚烷基)-OR z、C 3-6环烷基、3-8元杂环烷基(如3-6元杂环烷基)、C 2-4烯基、C 2-4炔基、苯基和5-6元杂芳基,其中所述烷基、亚烷基、环烷基、杂环烷基、烯基、炔基、苯基和杂芳基各自任选地被一个或多个选自氟、氯、氰基、氧代基、-OR z、羟基、-N(R z) 2、-NHR z、氨基、C 1-4烷基、C 3-6环烷基、3-6元杂环烷基、苯基、5-6元杂芳基的取代基取代;
若存在,每一个R z各自独立地选自氰基、C 1-4烷基、C 3-6环烷基、3-6元杂环烷基、苯基、苄基和5-6元杂芳基,其中所述烷基、环烷基、杂环烷基、苯基、苄基和杂芳基各自任选地被一个或多个选自氨基、氟、氯、氰基、氧代基、羟基、-O-(C 1-3烷基)、甲基、乙基、二氟甲基、三氟甲基、C 3-6环烷基和苯基的取代基取代;
R 4选自卤素、C 1-6烷基、C 3-6环烷基、-C(=O)-R z、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、芳基和杂芳基各自任选地被一个或多个选自氨基、氟、氯、氰基、羟基、氧代基、-O-(C 1-3烷基)、甲基、乙基、二氟甲基、三氟甲基和C 3-6环烷基的取代基取代;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000045
选自以下任意一种:
Figure PCTCN2021099561-appb-000046
优选以下任意一种:
Figure PCTCN2021099561-appb-000047
m为0、1或2;
若存在,每一个R 4a各自独立地选自氢、氟、氯、氰基、-C(=O)H、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3- 8环烷基)、-C(=O)-(3-8元杂环烷基)、氧代基、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 2-6烯基和C 2-6炔基,其中所述烷基、环烷基、杂环烷基、烯基和炔基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)和C 1-6烷基的取代基取代。
在本发明的一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自亚苯基、亚吡啶基和亚嘧啶基,其中所述亚苯基、亚吡啶基和亚嘧啶基各自任选地被一个或多个R a取代;
若存在,每一个R a各自独立地选自氢、氟、氯、溴、羟基、甲氧基、环丙氧基、氰基、甲基、乙基、正丙基、异丙基、二氟甲氧基、三氟甲氧基、三氟甲基、二氟甲基、环丙基、吡咯烷基、吗啉基、氨基、甲氨基、二甲氨基、甲硫基、甲磺酰基和氨磺酰基;
B选自-N(R b)-C(=O)-G-R d、-O-G-(3-10元杂环烷基)、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b
Figure PCTCN2021099561-appb-000048
其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);
G为C 1-3亚烷基,所述亚烷基任选地被一个或多个选自氢、氟、氧代基、羟基、氰基、-OR b、C 1-6烷基、C 1-6卤代烷基和C 1-6羟烷基的取代基取代;
R b选自氢、氟、氯、氨基、C 1-3烷基、环丙基和3-6元杂环烷基,其中所述烷基和杂环烷基各自任选地被一个或多个选自氢、氟、氧代基、羟基和氰基的取代基取代;
C选自亚吡咯烷基、亚氮杂环丁烷基、亚哌啶基和亚哌嗪基,其中所述亚吡咯烷基、亚氮杂环丁烷基、亚哌啶基和亚哌嗪基各自任选地被一个或多个R c取代;
若存在,每一个R c各自独立地选自氢、氟和羟基;
Y选自-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、亚环丙基和-C(=O)-CH 2-,其中所述-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、亚环丙基和-C(=O)-CH 2-各自任选地被一个或多个选自氟、羟基和氨基的取代基取代;
R d选自氟、氨基、羟基、氰基、-S(=O) 2-CH 3、-O-CH 3、-NH-C(=O)-O-CH 3和-O-C(=O)-NH 2,其中所述-S(=O) 2-CH 3、-O-CH 3和-NH-C(=O)-O-CH 3各自任选地被一个或多个选自氟、氯、氧代基、羟基和-O-(C 1-6烷基)的取代基取代;
R 3和R 4不相互连接时,
R 3选自氟、氯、-OR z、羟基、氰基、甲基、乙基、-CH 2-R z、-CH 2-OR z、环丙基、氧杂环丁烷基、-CH 2CH=CH 2、-CH=CH 2、-CH 2C≡CH、-C≡CH、吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基,其中所述吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基各自任选地被一个或两个选自氟、氯、氰基、-OR z、羟基、-N(R z) 2、-NHR z、氨基和吡唑基的取代基取代;
若存在,每一个R z各自独立地选自氰基、甲基、乙基、异丙基、环丙基、环丁基、环戊基、环己基、吗啉基、哌嗪基、氧杂环丁烷基、四氢呋喃基、四氢吡喃基、吡咯烷基、氮杂环丁烷基、苯基、苄基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、噻唑基、三氮唑基、吡嗪基、嘧啶基和吡啶基,其中所述哌嗪基、氮杂环丁烷基、苯基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、噻唑基、三氮唑基、吡嗪基、嘧啶基和吡啶基各自任选地被一个或两个选自氟、氯、氰基、羟基、甲氧基、甲基和乙基的取代基取代;
R 4选自氟、氯、甲基、乙基、异丙基、环丙基、甲氧甲基、羟基甲基、氟甲基、二氟甲基、三氟甲基、氟乙基、氰甲基、羟基甲基、2-甲基呋喃基、噻唑基、吡啶基和嘧啶基;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000049
选自以下任意一种:
Figure PCTCN2021099561-appb-000050
优选以下任意一种:
Figure PCTCN2021099561-appb-000051
Figure PCTCN2021099561-appb-000052
m为0、1或2;
若存在,每一个R 4a各自独立地选自氢、氟、氯、氧代基、甲基、乙酰基和环丙基,其中所述甲基和环丙基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)和C 1-6烷基的取代基取代。
在本发明的一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自亚苯基、亚吡啶基和亚嘧啶基,其中所述亚苯基、亚吡啶基和亚嘧啶基各自任选地被一个或多个R a取代;
若存在,每一个R a各自独立地选自氢、氟、氯、溴、羟基、氰基、甲基、乙基、正丙基、异丙基、环丙基、吡咯烷基、吗啉基、氨基、甲氨基和二甲氨基;
B选自
Figure PCTCN2021099561-appb-000053
Figure PCTCN2021099561-appb-000054
Figure PCTCN2021099561-appb-000055
-NH-C(=O)-CH 2-CN和-N(CH 3)-C(=O)-C(CH 3) 2-OH;
R 3和R 4不相互连接时,
R 3选自氟、氯、羟基、氰基、甲基、乙基、环丙基、氧杂环丁烷基、吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基,优选氟、氯、甲基、乙基、环丙基和氧杂环丁烷基;
R 4选自甲基、氟甲基、二氟甲基、三氟甲基、羟基甲基、甲氧甲基、氟乙基、氰甲基和羟基甲基;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000056
选自以下任意一种:
Figure PCTCN2021099561-appb-000057
以下任意一种:
Figure PCTCN2021099561-appb-000058
在本发明的另一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自亚苯基、亚吡啶基和亚嘧啶基,其中所述亚苯基、亚吡啶基和亚嘧啶基各自任选地被一个或多个R a取代;
若存在,每一个R a各自独立地选自氟、氯、溴、氰基、甲基和氨基;
B选自
Figure PCTCN2021099561-appb-000059
Figure PCTCN2021099561-appb-000060
Figure PCTCN2021099561-appb-000061
Figure PCTCN2021099561-appb-000062
-NH-C(=O)-CH 2-CN和-N(CH 3)-C(=O)-C(CH 3) 2-OH;
优选以下任意一种:
Figure PCTCN2021099561-appb-000063
Figure PCTCN2021099561-appb-000064
Figure PCTCN2021099561-appb-000065
-NH-C(=O)-CH 2-CN和-N(CH 3)-C(=O)-C(CH 3) 2-OH;
R 3和R 4不相互连接时,
R 3选自氟和甲基;
R 4选自甲基、氟甲基、氟乙基、氰甲基、羟基甲基和甲氧甲基;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000066
选自以下任意一种:
Figure PCTCN2021099561-appb-000067
优选以下任意一种:
Figure PCTCN2021099561-appb-000068
在本发明的一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自5-12元亚杂芳基,其中所述亚杂芳基任选地被一个或多个卤素取代;
B选自
Figure PCTCN2021099561-appb-000069
C选自3-6元亚杂环烷基,所述亚杂环烷基任选地被一个或多个氟、氰基、甲基或氟甲基取代;
Y选自C 1-3亚烷基和-C(=O)-(C 1-3亚烷基)-;
R d选自氟、氨基、羟基、氰基、-S(=O) 2-(C 1-6烷基)和-O-(C 1-6烷基);
R 3和R 4不相互连接时,R 3选自C 1-6烷基,R 4选自C 1-6烷基,所述烷基任选地被-O-(C 1-3烷基)取代;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000070
选自
Figure PCTCN2021099561-appb-000071
Figure PCTCN2021099561-appb-000072
m为0、1或2;
若存在,每一个R 4a各自独立地选自氢和氟。
在本发明的一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自亚吡啶基,所述亚吡啶基任选地被一个或多个氯取代;
B选自
Figure PCTCN2021099561-appb-000073
C选自亚氮杂环丁烷基和亚吡咯烷基,所述亚氮杂环丁烷基和亚吡咯烷基各自任选地被一个或多个氟、氰基、甲基或氟甲基取代;
Y选自-CH 2-、-CH(CH 3)-、-C(CH 3) 2-和-C(=O)-CH 2-;
R d选自氟、氨基、羟基、氰基、S(=O) 2-CH 3和-O-CH 3
R 3和R 4不相互连接时,R 3选自甲基,R 4选自甲基,所述甲基任选地被甲氧基取代;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000074
选自
Figure PCTCN2021099561-appb-000075
Figure PCTCN2021099561-appb-000076
在本发明的一些实施方案中,上述式I化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,其中
A选自亚吡啶基,所述亚吡啶基任选地被一个或多个氯取代;
B选自
Figure PCTCN2021099561-appb-000077
Figure PCTCN2021099561-appb-000078
R 3和R 4不相互连接时,R 3选自甲基,R 4选自甲基,所述甲基任选地被甲氧基取代;
或者,R 3和R 4以及与其连接的原子成环时,
Figure PCTCN2021099561-appb-000079
选自
Figure PCTCN2021099561-appb-000080
Figure PCTCN2021099561-appb-000081
在本发明的一些实施方案中,上述式I化合物具有如式IIb所示的结构,其中
A选自5-12元亚杂芳基,其中所述亚杂芳基任选地被一个或多个卤素取代;优选地,A选自亚吡啶基,其中所述亚吡啶基任选地被一个或多个氯取代;
B选自
Figure PCTCN2021099561-appb-000082
其中C选自3-6元亚杂环烷基,Y选自C 1-3亚烷基,R d选自羟基;优选地,B选自
Figure PCTCN2021099561-appb-000083
R 3和R 4以及与其连接的原子成环,且
Figure PCTCN2021099561-appb-000084
选自
Figure PCTCN2021099561-appb-000085
本领域技术人员应当理解,本发明涵盖针对各个实施方案进行任意组合所得的化合物。由一个实施方案中的技术特征或优选技术特征与另外的实施方案中的技术特征或优选技术特征组合得到的实 施方案也包括在本发明的范围内。
本发明提供了式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb和式Vc化合物所涵盖的下列化合物:
Figure PCTCN2021099561-appb-000086
Figure PCTCN2021099561-appb-000087
Figure PCTCN2021099561-appb-000088
Figure PCTCN2021099561-appb-000089
Figure PCTCN2021099561-appb-000090
Figure PCTCN2021099561-appb-000091
Figure PCTCN2021099561-appb-000092
Figure PCTCN2021099561-appb-000093
Figure PCTCN2021099561-appb-000094
化合物的制备方法
本发明提供了式I化合物的两种制备方法;
方法1包括下列步骤:
(1)式S-1化合物与式S-2化合物反应,以生成式M-1化合物;
Figure PCTCN2021099561-appb-000095
(2)式M-1化合物与式S-3化合物反应,以生成式M-2化合物;
Figure PCTCN2021099561-appb-000096
(3)式M-2化合物进行脱保护和功能基团转化,以生成式I化合物;
Figure PCTCN2021099561-appb-000097
方法2包括下列步骤:
(1)式S-1化合物与式S-3化合物反应,以生成式M-3化合物;
Figure PCTCN2021099561-appb-000098
(2)式M-3化合物与式S-2化合物反应,再经可选的脱保护和功能基团转化,以生成式I化合物;
Figure PCTCN2021099561-appb-000099
其中
LG 1和LG 2各自独立地为卤素离去基团或任选被卤素取代的C 1-6烷基磺酸酯基离去基团(如三氟甲磺酸酯基离去基团);另外,LG 2也可以为羟基;
R x为氢或离去基团;
R f为氢、羟基甲基、氟甲基或通过一步或多步反应转化为羟基甲基或氟甲基的基团;
PG 1为氢或氨基的保护基团(如甲基、叔丁氧羰基、叔丁基二甲基硅基、三异丙基硅基、苄基和甲氧甲基等);
其余基团如上文所定义。
在本发明的一些实施方案中,LG 1为卤素离去基团(如碘或溴)。
在本发明的一些实施方案中,LG 2为卤素离去基团(如溴或氯)或羟基。
在本发明的一些实施方案中,R x为氢、卤素、硼酸基、硼酸酯基、取代的硅基、取代的金属基团或任选被卤素取代的C 1-6烷基磺酸酯基,优选硼酸基或硼酸酯基。
在本发明的一些实施方案中,R f为氢、卤素(如氟、氯、溴或碘)、氟甲基或酯基(如-C(=O)-OC 2H 5)。
在本发明的一些实施方案中,上述步骤(1)中的反应在碱的存在下进行。在本发明的一些优选的实施方案中,碱为无机碱类,如磷酸钾。
在本发明的另一些实施方案中,上述步骤(1)中的反应在缩合试剂和碱存在下进行。在本发明的一些优选的实施方案中,碱为有机碱类,如DIPEA。在本发明的另一些优选的实施方案中,缩合试剂为BOP、HATU或PyBOP,优选BOP。
在本发明的一些实施方案中,上述步骤(2)中的反应在金属催化剂的存在下进行。在本发明的 一些优选的实施方案中,金属催化剂为钯催化剂或铜催化剂,如四(三苯基膦)钯、醋酸钯、三(二亚苄基丙酮)二钯、[1,1'-双(二苯基膦基)二茂铁]二氯化钯、1,2-双(二苯基膦基)乙烷二氯化钯和双(三苯基膦)二氯化钯、碘化亚铜等。
在本发明的一些实施方案中,上述步骤(3)中的功能基团转化包括(但不限于)下列反应:1)还原反应(所使用的试剂如LiBH 4、DIBAL-H);2)金属催化的偶联反应;3)水解反应。
在本发明的一些实施方案中,上述步骤(3)中的脱保护在酸水解或金属催化剂存在下的催化氢解的条件下进行。在本发明的一些优选的实施方案中,酸水解所使用的酸为有机酸,优选三氟甲酸。在本发明的另一些优选的实施方案中,催化氢化所使用的金属催化剂为钯/碳或氢氧化钯/碳。
本领域技术人员应当理解,根据期望获得的产物结构,可以省略上述制备方法中的一个或多个步骤,也可根据需要适当地调整反应步骤的顺序以及增加或省略保护/脱保护反应步骤。
本发明的其他通式化合物均可通过类似的方法合成。
药物组合物
术语“药物组合物”是指可以用作药物的组合物,其包含药物活性成分(API)(或治疗剂)以及可选的一种或多种药学上可接受载体,目的是促进对生物体的给药,利于活性成分的吸收,进而发挥生物活性。术语“药学上可接受的载体”是指与治疗剂一同给药的辅料,并且其在合理的医学判断的范围内适于接触人类和/或其它动物的组织而没有过度的毒性、刺激性、过敏反应或与合理的益处/风险比相应的其它问题或并发症。在本发明中可使用的药学上可接受的载体包括(但不限于)a)稀释剂;b)润滑剂;c)粘合剂;d)崩解剂;e)吸收剂、着色剂、调味剂和甜味剂;f)乳化剂或分散剂;和/或g)增强化合物的吸收的试剂。
本发明提供了一种药物组合物,其包含至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药。
在本发明的一些实施方案中,上述药物组合物还包含一种或多种药学上可接受的载体。
在本发明的另一些实施方案中,上述药物组合物还包含一种或多种额外的药物活性成分(如用于预防和/或治疗SHP2相关疾病的药物活性成分)。
在本发明的一些优选的实施方案中,上述药物组合物还同时包含一种或多种药学上可接受的载体以及一种或多种额外的药物活性成分(如用于预防和/或治疗SHP2相关疾病的药物活性成分)。
在本发明的一些实施方案中,上述药物组合物可以***地作用和/或局部地作用。为此目的,它们可以通过适合的途径给药,例如通过胃肠外、局部、静脉内、口服、皮下、动脉内、真皮内、经皮、直肠、颅内、腹膜内、鼻内、肌内、吸入途径或者医药领域技术人员熟知的所有方式施用。上述药物组合物可以与至少一种具有疾病或病症治疗效果的其他治疗剂联合给药。
在本发明的一些实施方案中,上述给药途径可以通过适合的剂型来实现。
在本发明的一些实施方案中,上述药物组合物可以包含0.01mg至1000mg的至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药。
本发明还提供了上述药物组合物或其相应的制剂形式的制备方法,其包括将至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药与一种或多种药学上可接受的载体和/或一种或多种额外的药物活性成分(如用于预防和/或治疗SHP2相关疾病的药物活性成分)组合。
药品
术语“药品”是指包含治疗剂、任选存在的其他治疗剂和任选存在的包装和/或说明书的组合产品。
术语“药物活性成分”、“药物活性物质”、“药物活性剂”或“治疗剂”是指一种化学实体,其可以有效地预防和/或治疗目标疾病或其一种或多种症状。
本发明提供了一种药品,其包含:
a)容器;
b)位于容器中的至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药或者药物组合物;和
c)任选存在的包装和/或说明书。
说明书列出了上述药品中药物活性成分或药物组合物的相关信息,优选具体列出了药物活性成分或药物组合物获准使用的适应症。说明书为可印刷材料(如纸、塑料、金属箔、胶粘纸等),其上可形成(如印刷或施涂)所需的信息。
在本发明的一些实施方案中,上述药品可以包含0.01mg至1000mg的至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药。
本发明还提供了上述药品的制备方法,其包括将至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药,或者药物组合物,与容器以及任选存在的包装和/或说明书组合。
医药用途
本发明的化合物能够对SHP2表现出较强的抑制作用,可以用作SHP2抑制剂。因此,本发明提供了本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药,或者药物组合物,或者药品,用作SHP2抑制剂的用途。
另外,本发明还提供了本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药,或者药物组合物,或者药品,在制备用于预防和/或治疗至少部分由SHP2介导的疾病或病症(或称SHP2相关疾病、SHP2酶相关疾病或SHP2磷酸酶相关疾病,尤其是癌症)的药物中的用途。
术语“至少部分由SHP2介导的疾病或病症(或SHP2相关疾病)”是指发病机理中至少包含一部分与SHP2有关的因素的疾病,特别是对SHP2磷酸酶抑制敏感或有响应的疾病,这些疾病包括(但不限于)肿瘤类疾病。
在本发明的一些实施方案中,上述肿瘤类病症包括(但不限于)乳腺癌、结肠直肠癌、结肠癌、肺癌(包括小细胞肺癌、非小细胞肺癌和细支气管肺泡癌)、***癌、胆管癌、骨癌、膀胱癌、头颈癌、肾癌、肝癌、胃肠组织癌、食道癌、卵巢癌、胰腺癌、皮肤癌、睾丸癌、甲状腺癌、子宫癌、***、外阴癌、多发性骨髓瘤、淋巴瘤和白血病,如慢性淋巴细胞性白血病(CLL)、急性淋巴细胞性白血病(ALL)和慢性骨髓性白血病(CML)。
治疗方法
本发明提供了一种用于预防和/或治疗至少部分由SHP2介导的疾病(尤其是癌症)的方法,其包括下列步骤:将预防和/或治疗有效量的至少一种本发明的式I、式IIa、式IIb、式IIIa、式IIIb、式IVa、式IVb、式IVc、式IVd、式Va、式Vb或式Vc化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物(如水合物)、同位素标记物、代谢物或前药,或者药物组合物,或者药品,施用于对其有需求的个体。
术语“有效量”是指能够诱发细胞、组织、器官或生物体(例如个体)产生生物或医学反应,并且足以实现所需预防和/或治疗效果的剂量。可调整给药方案以提供最佳响应。例如,可单次给药,可随时间分剂量给药,或可根据实际情况按比例减少或增加剂量后给药。可以理解的是,对于任何特定个体,具体的给药方案应根据需要以及给药人员的专业判断而调整。另外,也要区分是预防性应用,还是治疗性应用。在预防性应用中,通常以相对长的间隔长期给予相对低的剂量。在治疗性应用中,则通常以相对短的间隔给予相对高的剂量,直至疾病的进展被延缓或停止,优选直至个体表现出症状的 部分或完全改善。
在本发明中,进行合适的体外或体内测定来确定化合物、药物组合物和/或药品的效果以及给药是否适用于治疗个体所患疾病或病症。这些测定的实例将在下文非限制性实施例中进行描述。通常,足以实现预防和/或治疗效果的化合物的有效量为约0.001mg/千克体重/天至约10,000mg/千克体重/天。
术语“预防”包括抑制和延迟疾病的发作,并且不仅包括在发展疾病之前的预防,还包括在治疗后预防疾病的复发。
术语“治疗”是指逆转、减轻或消除所针对的疾病或病症。如果受试者接受了治疗量的本发明的化合物或其药学上可接受的形式或者本发明的药物组合物,该受试者的至少一种指标和症状表现出可观察到的和/或可检测出的缓解和/或改善,则表明该受试者已被成功地“治疗”。可以理解的是,治疗不仅包括完全地治疗,还包括未达到完全地治疗,但实现了一些生物学或医学相关的结果。具体而言,“治疗”表示本发明的化合物或其药学上可接受的形式或者本发明的药物组合物可以实现下列效果中的至少一种:(1)在正在经历或显示疾病病理学或症状学的动物中抑制疾病(即阻止病理学和/或症状学的进一步发展);(2)在正在经历或显示疾病病理学或症状学的动物中改善疾病(即逆转病理学和/或症状学)。
术语“施用”是指将药物活性成分(比如本发明的化合物)或包含药物活性成分的药物组合物(例如本发明的药物组合物)应用于个体或其细胞、组织、器官、生物流体等部位,以便使药物活性成分或药物组合物与个体或其细胞、组织、器官、生物流体等部位接触的过程。
术语“对其有需求”是指医生或其他护理人员对个体需要或者将要从预防和/或治疗过程中获益的判断,该判断的得出基于医生或其他护理人员在其专长领域中的各种因素。
术语“个体”(或称受试者)是指人类或非人动物。本发明的个体包括患有疾病和/或病症的个体(患者)和正常的个体。本发明的非人动物包括所有脊椎动物,例如非哺乳动物,例如鸟类、两栖类、爬行类等,和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
本发明还提供了一种用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)的组合方法,其包括:a)本发明的用于预防和/或治疗至少部分由SHP2介导的疾病或病症(尤其是癌症)的方法;和b)额外的疗法。
在本发明的一些实施方案中,上述组合方法中的额外的疗法包括(但不限于)放射疗法、化疗疗法、免疫疗法或其任意组合。可以在额外的疗法实施前、实施中或实施后施用本发明的化合物、药物组合物或药品。额外的疗法的实施与本发明的化合物、药物组合物或药品的施用可以同时进行,也可以前后紧密连接地进行,还可以间隔一段时间进行,其施用的方式与顺序可以根据具体治疗情况进行选择和调整。
为了使本发明的目的和技术方案更加清楚,以下结合实施例对本发明的实施方案进行详细描述。但是本领域技术人员将会理解,下列实施例仅用于说明本发明,而不应视为限定本发明的范围。
实施例中所使用的试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。未注明具体条件者,均按照常规条件或制造商建议的条件进行。本文中所使用的术语“室温”是指20℃±5℃。在用于修饰某一数值或数值范围时,本文中所使用的术语“约”是指包括该数值或数值范围以及该数值或数值范围的本领域技术人员可接受的误差范围,例如该误差范围为±10%、±5%、±4%、±3%、±2%、±1%、±0.5%等。除非另有声明,浓度以重量计,比例(包括百分比)以摩尔量计。
在常规的合成法以及实施例和中间体合成例中,各缩写的含义如下表所示。
Figure PCTCN2021099561-appb-000100
以下实施例中记载的化合物的结构通过核磁共振( 1H-NMR)和/或质谱(MS)来确定。
核磁共振( 1H-NMR)的测定仪器使用Bruker 400MHz核磁共振仪,测定溶剂为氘代甲醇(CD 3OD)、氘代氯仿(CDCl 3)、六氘代二甲基亚砜(DMSO-d 6)或氘代水(D 2O),内标物质为四甲基硅烷(TMS)。
以下实施例中的核磁共振(NMR)数据中的缩写代表的含义如下:
s:单峰(singlet)、d:二重峰(doublet)、t:三重峰(triplet)、q:四重峰(quartet)、dd:双二重峰(double doublet)、qd:四二重峰(quartet doublet)、ddd:双双二重峰(double double doublet)、ddt:双双三重峰(double double triplet)、dddd:双双双二重峰(double double double doublet)、m:多重峰(multiplet)、br:宽峰(broad)、J:偶合常数、Hz:赫兹、δ:化学位移。
全部化学位移(δ)值以百万分之一(ppm)的单位给出。
反应的监测采用薄层色谱或液相色谱-质谱联用,流动相体系包括(但不限于)二氯甲烷-甲醇体系、正己烷-乙酸乙酯体系和石油醚-乙酸乙酯体系,溶剂的体积比可以根据化合物的极性不同而进行调节,或者加入少量的三乙胺等进行调节。
薄层色谱采用GF 254硅胶板(0.4~0.5nm,烟台江友硅胶开发有限公司)。
制备型高效液相色谱的测定仪器使用岛津LC-8A制备型液相色谱仪(YMC,ODS,250×20mm×5μm C18色谱柱)。
质谱的测定仪器使用Agilent 6120B质谱仪,离子源为电喷雾离子源(ESI)。
柱色谱采用200~300目硅胶(青岛海洋化工有限公司)为固定相,流动相体系包括(但不限于)二氯甲烷-甲醇体系和正己烷-乙酸乙酯体系,溶剂的体积比可以根据化合物的极性不同而进行调节,或者加入少量的三乙胺等进行调节。
除非另有说明,以下实施例中的反应温度为室温(20~30℃)。
除非另有说明,以下实施例中的试剂购自Acros Organics、Aldrich Chemical、南京药石科技、上海书亚医药科技等。
中间体制备例1:6-溴-3-((3S,4S)-4-(叔丁氧羰基氨基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-5-甲基吡嗪-2-羧酸乙酯(化合物IM-1)和(3S,4S)-8-(5-溴-3-(羟基甲基)-6-甲基吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物IM-7)的制备。
Figure PCTCN2021099561-appb-000101
第一步:6-溴-3-((3S,4S)-4-(叔丁氧羰基氨基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-5-甲基吡嗪-2-羧酸乙酯(化合物IM-1)的制备:
将化合物A-1(200mg,766.07μmol)、DMF(4mL)、化合物A-2(195.60mg,804.37μmol)、BOP(312.74mg,1.53mmol)和DIPEA(594.04mg,4.60mmol)混合后,升温至40℃,反应2小时。反应结束后,加入二碳酸二叔丁酯(250.79mg,1.15mmol),继续反应2小时。加入水和乙酸乙酯进行萃取,有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,得到粗品。粗品经薄层色谱纯化,得到化合物IM-1(290mg,收率73%)。
第二步:(3S,4S)-8-(5-溴-3-(羟基甲基)-6-甲基吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物IM-7)的制备:
将化合物IM-1(5.7g,11.74mmol)溶解在CH 2Cl 2(100mL)中,降温至-78℃。缓慢加入DIBAL-H(35.23mL,35.23mmol),温度缓慢升至0℃,反应10分钟。反应完毕后,降温至-78℃,加入饱和酒石酸钾钠水溶液(200mL),待反应液升至室温搅拌2小时后分液,CH 2Cl 2萃取,有机相合并、干燥、浓缩,得到粗品。粗品经硅胶柱色谱纯化,得到化合物IM-7(2.16g,收率39%)。
中间体制备例2:(S)-6-溴-3-(1-(叔丁氧羰基氨基)-1,3-二氢螺[茚-2,4'-哌啶]-1'-基)吡嗪-2-羧酸甲酯(化合物IM-2)的制备。
Figure PCTCN2021099561-appb-000102
将化合物2-1(300mg,1.01mmol)加入到NMP(5mL)中,再加入DIPEA(786.14mg,6.08mmol),再分批加入化合物A-3(215.34mg,1.06mmol),于25℃反应6小时。然后加入二碳酸二叔丁酯(442.52mg,2.03mmol),于40℃反应2小时。反应完毕后,加入水(15mL),再用乙酸乙酯萃取,有机相浓缩,得到粗品,经柱层析纯化,得到化合物IM-2(310mg,收率56%)。
片段制备例1:2-氟-1-(4-甲基哌啶-4-基)乙胺(化合物A-4)的制备。
Figure PCTCN2021099561-appb-000103
第一步:4-(2-溴乙酰基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B1-2)的制备:
氮气保护下,将化合物B1-1(5.00g,20.72mmol)加入到THF(50mL)中,降温至-78℃,滴入LDA(2M的四氢呋喃溶液,34.19mL),滴完维持该温度搅拌10分钟。然后向体系中滴入TMSCl(7.43g,68.37mmol),控温-78℃,滴完后,搅拌1小时。将反应液倒入饱和碳酸氢钠水溶液中,甲基叔丁基醚萃取2次,合并有机相,饱和盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩干得到油状物。将该油状物溶于THF(25mL)中,加入碳酸钠(4.39g,41.44mmol),降温至0℃,分批加入NBS(4.06g,22.79mmol),随后升温至25℃,反应4小时。反应完成后,向反应液中加入甲基叔丁基醚及10%碳酸钠水溶液,搅拌后分液,有机相用食盐水洗涤1次,无水硫酸钠干燥,过滤,滤液浓缩后,经柱层析纯化,得到化合物B1-2(6.60g,收率99%)。
第二步:4-(2-氟乙酰基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B1-3)的制备:
将化合物B1-2(6.60g,20.61mmol)、18-冠-6(6.86g,25.97mmol)、无水氟化钾(3.23g,55.65mmol)加入甲苯(100mL)中,升温至85℃,反应6小时。反应完成后,冷至室温,向体系中加入水,分出有机相,食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩干,得粗品化合物B1-3(4.68g),未经纯化直接用于下一步反应。
第三步:4-(1-(叔丁基亚磺酰氨基)-2-氟乙基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B1-4)的制备:
将化合物B1-3(4.68g,16.24mmol)溶于钛酸四异丙酯(50mL),加入叔丁基亚磺酰胺(2.95g,24.36mmol),氮气置换后,升温至90℃,反应5小时,冷却至室温后,加入饱和氯化钠溶液,乙酸乙酯萃取,合并有机相,干燥后减压浓缩。残留物溶于甲醇(50mL),冰浴下缓慢加入硼氢化钠(1.69g,44.67mmol),升温至25℃,反应2小时。反应完成后,降温至0℃,加入饱和氯化铵淬灭反应,乙酸乙酯萃取,合并有机相,干燥,减压浓缩后经柱层析纯化,得化合物B1-4(4.85g,收率80%)。
第四步:2-氟-1-(4-甲基哌啶-4-基)乙胺(化合物A-4)的制备:
将化合物B1-4(0.30g,0.82mmol)溶于盐酸二氧六环溶液(4M,3mL),25℃反应2小时。浓缩反应液,得粗品化合物A-4的盐酸盐(0.19g),未经纯化直接用于下一步反应。
片段制备例2:2-甲氧基-1-(4-甲基哌啶-4-基)乙胺(化合物A-5)的制备。
Figure PCTCN2021099561-appb-000104
第一步:4-甲基-4-(环氧乙烷-2-基)哌啶-1-羧酸叔丁酯(化合物B2-2)的制备:
氮气保护下,将三甲基碘化亚砜(12.20g,55.4mmol)加入到DMSO(50mL)中,冰水降温至0℃;分批加入NaH(1.27g,55.4mmol),加完保温10~20℃,反应0.5小时;一次性加入4-甲酰基-4-甲基哌啶-1-羧酸叔丁酯(7.00g,30.8mmol),保温10~20℃,反应4小时。冰水降温至0℃,依次加入水(150ml)、乙酸乙酯(200ml),搅拌10分钟,分出有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩至干,得到化合物B2-2粗品(5.37g,粗品收率72%),未进一步纯化,直接用作下一步。
第二步:4-(1-羟基-2-甲氧基乙基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B2-3)的制备:
将化合物B2-2(1.40g,5.80mmol)溶于MeOH(60mL),加入NaOMe(10.44g,58.0mmol,30%的MeOH溶液),然后40℃搅拌5h。LCMS显示原料基本消失,冷至室温,加1N HCl淬灭反应,乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,抽滤,浓缩,得化合物B2-3粗品(1.4g,粗品收率88%),未进一步纯化,直接用作下一步。
第三步:4-(2-甲氧基乙酰基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B2-4)的制备:
将化合物B2-3(300mg,1.10mmol)溶于DCM(20mL),加入碳酸氢钠(277mg,3.30mmol)和Dess-Martin试剂(710mg,1.65mmol),然后室温搅拌5h。LCMS显示原料基本消失,冷至室温,加饱和亚硫酸钠溶液淬灭反应,二氯甲烷萃取,合并有机相,无水硫酸钠干燥,抽滤,浓缩,得化合物B2-4粗品(250mg,粗品收率84%),未进一步纯化,直接用作下一步。
第四步:(Z)-4-(1-(肟基)-2-甲氧基乙基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B2-5)的制备:
将化合物B2-4(230mg,0.85mmol)溶于EtOH(10mL),加入羟胺盐酸盐(118mg,1.70mmol)和三乙胺(257mg,2.54mmol),然后60℃搅拌2h。LCMS显示原料基本消失,冷至室温,加乙酸乙酯稀释,用1N盐酸溶液洗涤,合并有机相,无水硫酸钠干燥,抽滤,浓缩,得化合物B2-5粗品(230mg,粗品收率95%),未进一步纯化,直接用作下一步。
第五步:4-(1-氨基-2-甲氧基乙基)-4-甲基哌啶-1-羧酸叔丁酯(化合物B2-6)的制备:
将化合物B2-5(230mg,0.85mmol)溶于MeOH(10mL),加入兰尼镍(94mg,1.61mmol),氢气球置换三次后,室温搅拌10小时。LCMS显示原料基本消失,垫硅藻土抽滤,浓缩,得化合物B2-6粗品(200mg,粗品收率91%),未进一步纯化,直接用作下一步。
第六步:2-甲氧基-1-(4-甲基哌啶-4-基)乙胺(化合物A-5)的制备:
将化合物B2-6(200mg,0.73mmol)溶于DCM(10mL),加入三氟乙酸(3mL),室温搅拌1小时。LCMS显示原料基本消失,浓缩,得化合物A-5粗品的三氟乙酸盐(200mg,粗品收率95%),未进一步纯化,直接用作下一步。
参照中间体制备例2的方法,采用相应的起始原料合成中间体化合物IM-3、IM-4、IM-5、IM-6、IM-8、IM-9、IM-10、IM-11、IM-12、IM-13和IM-14;参照中间体制备例1的方法,采用相应的起始原料合成中间体化合物IM-15。
Figure PCTCN2021099561-appb-000105
实施例1:(3-((3S,4S)-4-氨基-3-甲基-2-氧代-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-基)甲醇(化合物1)的制备。
Figure PCTCN2021099561-appb-000106
第一步:(1-(3-氯-4-碘吡啶-2-基)吡咯烷-2-基)甲醇(化合物1-2)的制备:
将化合物1-1(1.00g,3.88mmol)、DMSO(12mL)、吡啶烷-2-基甲醇(432.20mg,4.28mmol)及K 2CO 3(1.61g,11.65mmol)混合后,升温至80℃,反应4小时。冷至室温,加入水和乙酸乙酯,萃取产品,有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,得到化合物1-2粗品(1.29g,收率98%),未经纯化直接用于下一步反应。
第二步:3-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)丙酸2-乙基己酯(化合物1-3)的制备:
将化合物1-2(0.50g,1.48mmol)、3-巯基丙酸2-乙基己酯(370.83mg,1.70mmol)、DIPEA(381.72mg,2.95mmol)、Xantphos(85.45mg,147.68μmol)及Pd 2(dba) 3(67.62mg,73.84μmol)加入到二氧六环(5mL)中,抽真空,氮气置换三次,随后升温至100℃,反应4小时。冷至室温,滤除固体,浓缩滤液,加入水和乙酸乙酯,萃取产品,有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,得到化合物1-3粗品(632.00mg,收率99%),未经纯化直接用于下一步反应。
第三步:3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-硫酚钠(化合物1-4)的制备:
将乙醇钠(20%的乙醇溶液,0.35mL)加入到化合物1-3(300mg,559.42μmol)的THF)(5mL)溶液中,于25℃反应1小时。浓缩除去反应液中的绝大部分溶剂,加入二氯甲烷(5mL),搅拌10分钟,过滤,干燥滤饼,得到化合物1-4粗品(149.77mg,收率100%),未经纯化直接用于下一步反应。
第四步:3-((3S,4S)-4-(叔丁氧羰基氨基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-羧酸乙酯(化合物1-5)的制备:
将化合物IM-1(100.0mg,194.77μmol)、化合物1-4(143.40mg,428.50μmol)、CuI(37.09mg,194.77μmol)、1,10-菲罗啉(35.10mg,194.77μmol)及K 3PO 4(124.03mg,584.31μmol)加入到二氧六环(5mL)中,于微波反应器中升温至100℃,反应1.5小时。冷至室温,滤除不溶物,浓缩滤液,得到粗品,粗品经薄层色谱法纯化,得到化合物1-5(65.00mg,收率49.28%)。
第五步:(3S,4S)-8-(5-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-3-(羟基甲基)-6-甲基吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物1-6)的制备:
将化合物1-5(65mg,95.98μmol)溶于二氯甲烷(2mL)中,于-25℃下缓慢加入DIBAL-H(1M的己烷溶液,0.67mL),保温反应1小时。反应完成后,向反应液中加入二氯甲烷(10mL)和饱和酒石酸钾钠水溶液(5mL),搅拌2小时,萃取产品,有机相用无水硫酸钠干燥,过滤,浓缩滤液,得到粗品,粗品经薄层色谱法纯化,得到化合物1-6(42mg,收率68.89%)。
第六步:(3-((3S,4S)-4-氨基-3-甲基-2-氧代-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-基)甲醇(化合物1)的三氟乙酸盐的制备:
将化合物1-6(42mg,66.12μmol)溶于二氯甲烷(1mL)和TFA(150.78mg,1.32mmol)的混合物中,于25℃反应1小时。浓缩除去反应液中的溶剂,得到粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物1的三氟乙酸盐(27.10mg,收率61%)。
MS(ESI):m/z 535.0[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ7.92(s,3H),7.80(d,J=5.2Hz,1H),5.90(d,J=5.2Hz,1H),4.48 (s,2H),4.43-4.40(m,1H),4.23-4.19(m,1H),3.92(m,1H),3.82(m,2H),3.70(d,J=8.8Hz,1H),3.52(dd,J=10.4,3.6Hz,1H),3.46-3.36(m,2H),3.23(m,1H),3.08(m,2H),2.42(s,3H),2.07-1.98(m,1H),1.96-1.67(m,6H),1.61(m,1H),1.22(d,J=6.4Hz,3H)。
实施例2:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物2)的制备。
Figure PCTCN2021099561-appb-000107
第一步:6-溴-3-((3S,4S)-4-(叔丁氧羰基氨基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-羧酸甲酯(化合物2-2)的制备:
将化合物2-1(120.0mg,0.41mmol)、化合物A-2(103.54mg,0.43mmol)、DIPEA(314.46mg,2.43mmol)加入到NMP(3mL)中,于25℃反应12小时。然后向反应液中加入二碳酸二叔丁酯(177.01mg,811.04μmol),于25℃继续反应2小时。加入水和乙酸乙酯,萃取产品,有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,得到粗品,粗品经薄层色谱法纯化,得到化合物2-2(162mg,收率82%)。
第二步至第四步:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物2)的制备:
除在第二步中使用化合物2-2代替实施例1的第四步中的化合物IM-1外,采用与实施例1中第四步至第六步所描述的类似方法合成化合物2的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物2的三氟乙酸盐。
MS(ESI):m/z 521.3[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.36(s,1H),7.94(s,3H),7.82(d,J=5.2Hz,1H),6.07(d,J=5.2Hz,1H),4.52(s,2H),4.44-4.39(m,1H),4.23-4.20(m,1H),3.89-3.81(m,3H),3.70(d,J=8.8Hz,1H),3.51(dd,J=10.4,3.6Hz,1H),3.46-3.34(m,2H),3.22(m,1H),3.09(m,2H),2.07-1.99(m,1H),1.96-1.79(m,4H),1.78-1.67(m,2H),1.60(m,1H),1.22(d,J=6.4Hz,3H)。
实施例3:(3-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物17)的制备。
Figure PCTCN2021099561-appb-000108
除在第一步中使用1-(4-甲基哌啶-4-基)乙胺二盐酸盐(化合物3-0)代替实施例2的第一步中的化合物A-2外,采用与实施例2中第一步至第四步所描述的类似方法合成化合物17的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物17的三氟乙酸盐。
MS(ESI):m/z 493.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.35(s,1H),7.83(d,J=5.2Hz,1H),7.71(s,3H),6.08(d,J=5.2Hz,1H),4.53(s,2H),4.46-4.40(m,2H),3.84(m,3H),3.53(dd,J=10.4,3.6Hz,1H),3.41-3.35(m,1H),3.25-3.19(m,2H),3.18-3.13(m,1H),2.07–2.00(m,1H),1.97-1.88(m,1H),1.86-1.78(m,1H),1.76-1.68(m,1H),1.65–1.53(m,3H),1.44(m,1H),1.16(d,J=6.4Hz,3H),1.03(s,3H)。
实施例4:(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(氟甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇(化合物22)的制备。
Figure PCTCN2021099561-appb-000109
第一步:(3S,4S)-8-(5-溴-3-(羟基甲基)吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物4-1)的制备:
将化合物2-2(60mg,0.12mmol)溶于二氯甲烷(2mL)中,于-25℃缓慢加入DIBAL-H(1M的己烷溶液,0.84mL),保温反应1小时。反应完成后,向反应液中加入二氯甲烷(10mL)和饱和酒石酸钾钠水溶液(5mL),搅拌2小时,萃取产品,有机相用无水硫酸钠干燥,过滤,浓缩滤液,得到粗品,粗品经薄层色谱法纯化,得到化合物4-1(40mg,收率71%)。
第二步:(3S,4S)-8-(5-溴-3-(氟甲基)吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物4-2)的制备:
将化合物4-1(40mg,87.46μmol)加入二氯甲烷(4mL)中,缓慢加入DAST(28.19mg,174.92μmol),于室温反应2小时。将反应液缓慢倒入饱和碳酸氢钠水溶液(5ml)和二氯甲烷(5ml)的混 合液中,萃取产品,有机相用饱和食盐水(2ml)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,得到粗品,粗品经薄层色谱法纯化,得到化合物4-2(32mg,收率80%)。
第三步:(3S,4S)-8-(5-(3-氯-2-(2-羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)-3-(氟甲基)吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物4-3)的制备:
除在本步骤中使用4-2代替实施例1中第四步的IM-1外,采用实施例1中第四步所描述的类似方法合成化合物4-3。
第四步:(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(氟甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇(化合物22)的制备:
将化合物4-3(20mg,0.03mmol)溶于二氯甲烷(1mL)和TFA(1mL)的混合物中,于25℃反应1小时。浓缩除去反应液中的溶剂,得到粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物22的三氟乙酸盐(5mg,收率24%)。
MS(ESI):m/z 523.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.41(d,J=1.6Hz,1H),7.74(d,J=6.0Hz,1H),6.28(dd,J=6.0,1.6Hz,1H),5.47(d,J=47.6Hz,2H),4.68-4.53(m,1H),4.33-4.27(m,1H),3.99-3.86(m,4H),3.67-3.61(m,2H),3.56(dd,J=11.2,5.2Hz,1H),3.46(d,J=4.0Hz,1H),3.27-3.14(m,3H),2.22-2.16(m,1H),2.08-2.02(m,1H),1.98-1.84(m,5H),1.76-1.72(m,1H),1.32(d,J=6.4Hz,3H)。
实施例5:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)乙腈(化合物11)的制备。
Figure PCTCN2021099561-appb-000110
除在第一步中使用3-氰甲基氮杂环丁烷代替实施例1的第一步中的2-羟基甲基吡咯烷,第四步中使用化合物4-1代替实施例1的第四步中的化合物IM-1外,采用与实施例1中第一步至第四步以及第六步所描述的类似方法合成化合物11的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物11的甲酸盐。
MS(ESI):m/z 516.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.53(brs,1H),8.30(s,1H),7.73(d,J=5.6Hz,1H),6.14(d,J=5.6Hz,1H),4.67(s,2H),4.41(t,J=8.8Hz,2H),4.32-4.26(m,1H),4.03-4.00(m,2H),3.96-3.94(m,1H),3.89-3.80(m,3H),3.39(d,J=4.0Hz,1H),3.20-3.08(m,2H),3.02-2.94(m,1H),2.83(d,J=6.8Hz,2H),1.98-1.86(m,3H),1.74-1.71(m,1H),1.30(d,J=6.4Hz,3H)。
实施例6:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物12)的制备。
Figure PCTCN2021099561-appb-000111
除在第一步中使用3-羟基甲基氮杂环丁烷盐酸盐代替实施例1的第一步中的2-羟基甲基吡咯烷,第四步中使用化合物2-2代替实施例1的第四步中的化合物IM-1外,采用与实施例1中第一步至第六步所描述的类似方法合成化合物12的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物12的三氟乙酸盐。
MS(ESI):m/z 507.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.35(s,1H),7.97(s,3H),7.80(d,J=5.2Hz,1H),6.04(d,J=5.2Hz,1H),4.52(s,2H),4.25-4.12(m,3H),3.98-3.65(m,8H),3.56(d,J=6.0Hz,1H),3.47-3.40(m,1H),3.20-3.01(m,2H),2.76-2.62(m,1H),1.90–1.50(m,4H),1.17(d,J=6.4Hz,3H)。
实施例7:(3-((S)-4-氨基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(2-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物21)的制备。
Figure PCTCN2021099561-appb-000112
除在第一步中使用化合物7-1代替实施例2的第一步中的化合物A-2外,采用与实施例2中第一步至第四步所描述的类似方法合成化合物21的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物21的三氟乙酸盐。
MS(ESI):m/z 507.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.33(s,1H),7.72(d,J=6.0Hz,1H),6.28(d,J=6.0Hz,1H),4.68(s,2H),4.63-4.53(m,1H),4.17(dd,J=10.8,5.6Hz,1H),4.02-3.81(m,6H),3.71-3.50(m,4H),3.27-3.19(m,1H),3.18-3.10(m,1H),2.22-2.15(m,1H),2.07-1.99(m,1H),1.99-1.73(m,6H)。
实施例8:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(羟基甲基)吡咯烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物37)的制备。
Figure PCTCN2021099561-appb-000113
除在第一步中使用3-羟基甲基吡咯烷代替实施例1的第一步中的2-羟基甲基吡咯烷,第四步中使用化合物2-2代替实施例1的第四步中的化合物IM-1外,采用与实施例1中第一步至第六步所描述的类似方法合成化合物37的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物37的甲酸盐。
MS(ESI):m/z 521.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.48(brs,1H),8.30(s,1H),7.72(d,J=4.0Hz,1H),6.10(d,J=4Hz,1H),4.67(s,2H),4.32-4.21(m,1H),3.97-3.94(m,1H),3.92-3.78(m,3H),3.74-3.65(m,3H),3.62-3.50(m,3H),3.42(d,J=4Hz,1H),3.20-3.06(m,2H),2.48-2.37(m,1H),2.09-2.01(m,1H),1.98-1.81(m,3H),1.77-1.68(m,2H),1.31(d,J=8Hz,3H)。
实施例9:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-((S)-2-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物33)的制备。
Figure PCTCN2021099561-appb-000114
除在第一步中使用(S)-2-羟基甲基氮杂环丁烷代替实施例1的第一步中的2-羟基甲基吡咯烷,第四步中使用化合物2-2代替实施例1的第四步中的化合物IM-1外,采用与实施例1中第一步至第六步所描述的类似方法合成化合物33的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物33的三氟乙酸盐。
MS(ESI):m/z 507.1[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.35(s,1H),7.67(d,J=6.4Hz,1H),6.31(d,J=6.4Hz,1H),4.89-4.82(m,1H),4.68(s,2H),4.67-4.52m,2H),4.36-4.25(m,1H),4.02-3.79(m,6H),3.46(d,J=4.0Hz,1H),3.25-3.07(m,2H),2.54-2.43(m,1H),2.33-2.20(m,1H),2.04-1.67(m,4H),1.32(d,J=6.4Hz,3H)。
实施例10:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-3-基)乙腈(化合物14)的制备。
Figure PCTCN2021099561-appb-000115
除在第一步中使用2-(吡咯烷-3-基)乙腈代替实施例1的第一步中的2-羟基甲基吡咯烷,第四步中使用化合物4-1代替实施例1中的化合物IM-1外,采用与实施例1中第一步至第四步以及第六步所描述的类似方法合成化合物14的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物14的三氟乙酸盐。
MS(ESI):m/z 530.2[M+H] +
1H-NMR(400MHz,DMSO-d6):δ8.35(s,1H),7.95(s,3H),7.82(d,J=5.2Hz,1H),6.07(d,J=5.2Hz,1H),4.52(s,2H),4.23-4.18(m,1H),3.91-3.82(m,3H),3.72-3.64(m,4H),3.44-3.39(m,2H),3.14-3.04(m,2H),2.78-2.69(m,2H),2.14-2.06(m,1H),1.85-1.59(m,6H),1.22(d,J=6.4Hz,3H)。
实施例11:2-(1-(5-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-6-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物130)的制备。
Figure PCTCN2021099561-appb-000116
第一步:2-(1-(6-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物11-2)的制备:
将2,6-二氯吡啶(800mg,5.41mmol)溶于DMSO(10mL),加入化合物12-0(983.64mg,6.49mmol)、碳酸钾(2.24g,16.22mmol),加热至80℃,反应4小时,LCMS检测反应完成后,向反应液加入水,乙酸乙酯萃取,合并有机相并干燥、浓缩,得粗品化合物11-2(1.06g),未经纯化直接用于下一步反应。
第二步:2-(1-(6-氯-5-碘吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物11-3)的制备:
将化合物11-2(1.06g,4.68mmol)溶于乙腈(20mL),加入NIS(1.16g,5.14mmol),25℃反应16小时,LCMS检测反应完成后,将反应液浓缩,粗品经薄层制备法纯化,得化合物11-3(700mg,收 率42%)。
第三步至第七步:2-(1-(5-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-6-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物130)的制备:
采用与实施例6中的第二步至第六步类似的方法合成化合物130的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物130的三氟乙酸盐。
MS(ESI):m/z 535.0[M+H] +
1H-NMR(400MHz,DMSO-d6):δ7.94(s,3H),7.72(s,1H),7.71(d,J=8.4Hz,1H),6.38(d,J=8.4Hz,1H),4.46(s,2H),4.29–4.11(m,1H),3.97–3.86(m,4H),3.82(d,J=8.8Hz,1H),3.65(d,J=8.8Hz,1H),3.63–3.56(m,3H),2.97–2.81(m,2H),2.72(dt,J=14.8,7.6Hz,1H),1.91–1.65(m,3H),1.62–1.51(m,1H),1.20(d,J=7.2Hz,3H),1.07(s,6H)。
实施例12:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物67)的制备。
Figure PCTCN2021099561-appb-000117
第一步:2-(1-(3-氯-4-碘吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物12-1)的制备:
将化合物1-1(25.0g,97.1mmol)、DMSO(200mL)、化合物12-0(18.8g,111.7mmol)及K 2CO 3(40.3g,291.3mmol)混合后,升温至80℃,反应4小时。冷至室温,加入水和乙酸乙酯,萃取产品,有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩,得到化合物12-1粗品(36.7g),未经纯化直接用于下一步反应。
第二步:3-(3-氯-2-(3-(2-羟基丙-2-基)氮杂环丁烷-1-基)吡啶-4-基硫基)丙酸2-乙基己酯(化合物12-2)的制备:
将化合物12-1(36.7g,93.68mmol)、3-巯基丙酸2-乙基己酯(24.6g,112.40mmol)、DIPEA(24.2g,187.4mmol)、Xantphos(542.0mg,0.94mmol)及Pd 2(dba) 3(428.9mg,0.47mmol)加入到二氧六环(300mL)中,抽真空,氮气置换三次,随后升温至100℃反应4小时。冷至室温,滤除固体,滤液浓缩得到粗品,经柱层析纯化,得到化合物12-2(37.8g,收率86%)。
第三步:3-氯-2-(3-(2-羟基丙-2-基)氮杂环丁烷-1-基)吡啶-4-硫酚钠(化合物12-3)的制备:
将甲醇钠(5.85g,30%的甲醇溶液)加入到化合物12-2(12.00g,27.1mmol)的THF(120mL)溶液中,于25℃反应1小时。浓缩除去反应液中的绝大部分溶剂,加入二氯甲烷(100mL)、石油醚(50mL),搅拌20分钟,过滤,真空干燥滤饼,得到化合物12-3粗品(7.85g),未经纯化直接用于下一步反应。
第四步:3-((3S,4S)-4-(叔丁氧羰基氨基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(2-羟基丙-2-基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-羧酸甲酯(化合物12-4)的制备:
将化合物2-2(5.00g,10.30mmol)、化合物12-3(5.80g,20.60mmol)、CuI(0.98g,5.15mmol)、1,10-菲罗啉(1.02g,5.15mmol)及K 3PO 4(4.37g,20.6mmol)加入到二氧六环(100mL)中,升温 至100℃反应3小时。冷至室温,滤除不溶物,浓缩滤液得到粗品,粗品经柱层析纯化,得到化合物12-4(5.15g,收率71%)。
第五步:(3S,4S)-8-(5-(3-(2-羟基丙-2-基)氮杂环丁烷-1-基)吡啶-4-基硫基)-3-(羟基甲基)吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-基氨基甲酸叔丁酯(化合物12-5)的制备:
将化合物12-4(5.15g,7.77mmol)溶于乙醇(80mL)中,加入氯化钙(3.45g,31.10mmol),分批加入硼氢化钠(1.18g,31.10mmol),缓慢加热至35℃反应3小时。缓慢加入水(50mL),柠檬酸水调PH=7~8,浓缩掉大部分乙醇,用乙酸乙酯(150ml)萃取产品,有机相浓缩干得到粗品,经柱层析纯化,得到化合物12-5(2.67g,收率51%)。
第六步:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物67)的制备:
将化合物12-5(40mg,0.06mmol)溶于二氯甲烷(2mL)中,缓慢加入TFA(4mL),升温至30℃反应1小时。浓缩除去反应液中的溶剂得到粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物67。
MS(ESI):m/z 535.2[M+H] +
1H-NMR(400MHz,DMSO-d 6)δ8.31(s,1H),7.79(d,J=5.6Hz,1H),6.01(d,J=5.2Hz,1H),4.51(s,2H),4.14–4.02(m,5H),3.75–3.62(m,3H),3.53(d,J=8.4Hz,1H),3.30–3.15(m,2H),3.00(d,J=5.2Hz,1H),2.62(p,J=7.6Hz,1H),1.90–1.79(m,1H),1.79–1.68(m,1H),1.67–1.50(m,2H),1.10(d,J=6.4Hz,3H),1.05(s,6H)。
实施例13:(S)-2-(1-(4-(5-(4-氨基-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物68)的制备。
Figure PCTCN2021099561-appb-000118
除在第一步中使用化合物12-3代替实施例7的第二步中的化合物1-4外,采用与实施例7中第二步至第四步所描述的类似方法合成化合物68的粗品,粗品经Pre-TLC(MeOH/DCM=1:5,V/V)纯化,得到化合物68。
MS(ESI):m/z 521.2[M+H] +
1H-NMR(400MHz,DMSO)δ8.33(s,1H),7.79(d,J=5.2Hz,1H),6.01(d,J=5.2Hz,1H),5.51(t,J=6.0Hz,1H),4.51(d,J=5.2Hz,2H),4.45(s,1H),4.12–4.02(m,4H),3.95(dd,J=8.4,6.4Hz,1H),3.83–3.66(m,3H),3.59(d,J=8.4Hz,1H),3.30–3.28(m,1H),3.21–3.10(m,2H),3.06(t,J=6.0Hz,1H),2.66–2.56(m,1H),1.81–1.74(m,1H),1.73–1.63(m,1H),1.52–1.42(m,2H),1.05(s,6H)。
实施例14:(S)-2-(1-(4-(5-(4-(1-氨基-2-甲氧基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物69)的制备。
Figure PCTCN2021099561-appb-000119
采用实施例13类似的方法合成化合物69的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物69的三氟乙酸盐。
MS(ESI):m/z 537.2[M+H] +
1H-NMR(400MHz,DMSO)δ8.33(s,1H),7.85(br,3H),7.79(d,J=5.6Hz,1H),6.01(d,J=5.2Hz,1H),4.51(s,2H),4.12–4.05(m,4H),3.82–3.77(m,2H),3.63(dd,J=10.8,3.2Hz,1H),3.54–3.43(m,1H),3.33(s,3H),3.26–3.13(m,3H),2.62(p,J=7.6Hz,1H),1.76–1.62(m,2H),1.62–1.53(m,1H),1.53–1.41(m,1H),1.13–0.99(m,9H)。
实施例15:1-((4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基氨基)甲基)环丙烷-1-醇(化合物58)的制备。
Figure PCTCN2021099561-appb-000120
采用实施例6类似的方法合成化合物58的粗品,粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物58的三氟乙酸盐。
MS(ESI):m/z 507.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.35(s,1H),7.63(d,J=6.4Hz,1H),6.24(d,J=6.4Hz,1H),4.68(s,2H),4.33-4.26(m,1H),4.00-3.82(m,4H),3.60(s,2H),3.46(d,J=4.4Hz,1H),3.27-3.14(m,2H),2.01-1.67(m,4H),1.32(d,J=6.4Hz,3H),0.81–0.75(m,2H),0.74–0.67(m,2H)。
实施例16:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(氟甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物125)的制备。
Figure PCTCN2021099561-appb-000121
采用实施例6类似的方法合成化合物125的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物125的三氟乙酸盐。
MS(ESI):m/z 509.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.35(s,1H),8.02(s,3H),7.82(d,J=5.2Hz,1H),6.08(d,J=5.2Hz,1H),4.68(d,J=5.6Hz,1H),4.56(d,J=5.6Hz,1H),4.52(s,2H),4.30-4.17(m,4H),4.00-3.78(m,4H),3.70(d,J=9.2Hz,1H),3.48-3.40(m,1H),3.15-2.90(m,3H),1.93-1.55(m,4H),1.22(d,J=6.4Hz,3H)。
实施例17:1-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)乙醇(化合物70)的制备。
Figure PCTCN2021099561-appb-000122
采用实施例6类似的方法合成化合物70的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物70的三氟乙酸盐。
MS(ESI):m/z 521.3[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.34(s,1H),7.95(s,3H),7.80(d,J=5.6Hz,1H),6.03(d,J=5.6Hz,1H),4.52(s,2H),4.25-4.18(m,1H),4.17-4.10(m,2H),4.08-4.01(m,1H),3.90-3.45(m,7H),3.15-3.01(m,2H),2.57-2.52(m,1H),1.92–1.55(m,4H),1.22(d,J=6.4Hz,3H),1.02(d,J=6.4Hz,3H)。
实施例18:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)乙腈(化合物5)的制备。
Figure PCTCN2021099561-appb-000123
采用实施例5类似的方法合成化合物5的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物5的的三氟乙酸盐。
MS(ESI):m/z 530.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.37(s,1H),7.98(s,3H),7.88(d,J=5.2Hz,1H),6.18(d,J=5.2Hz,1H),4.52(s,2H),4.45–4.44(m,1H),4.24–4.18(m,1H),3.97–3.81(m,4H),3.70(d,J=9.2Hz,1H),3.43–3.37(m,2H),3.14–3.04(m,2H),2.89–2.78(m,2H),2.22–2.16(m,1H),2.06–1.95(m,1H),1.89–1.74(m,5H),1.63–1.59(m,1H),1.20(t,J=6.4Hz,3H)。
实施例19:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)乙酰胺(化合物4)的制备。
Figure PCTCN2021099561-appb-000124
将化合物5(20mg,37.73μmol)溶于二氯甲烷(1mL)、水(0.5mL)和TFA(80mg,0.69mmol)的混合物中,于35℃反应2小时。浓缩除去反应液中的溶剂,得到粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物4的三氟乙酸盐(7.20mg,收率30%)。
MS(ESI):m/z 548.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.36(s,1H),7.94(s,3H),7.86(d,J=5.2Hz,1H),7.23(s,1H),6.79(s,1H),6.10(d,J=5.2Hz,1H),4.59–4.49(m,3H),4.25–4.18(m,1H),3.90–3.79(m,3H),3.76–3.69(m,3H),3.46–3.44(m,1H),3.36–3.32(m,1H),3.14–3.04(m,2H),2.55(dd,J=14.0,3.6Hz,1H),2.13–2.09(m,1H),1.95–1.81(m,3H),1.67–1.59(m,4H),1.22(d,J=6.4Hz,3H)。
实施例20:(R)-2-(1-(4-(5-(3-氨基-5-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物97)的制备。
Figure PCTCN2021099561-appb-000125
采用实施例13类似的方法合成化合物97的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物97的三氟乙酸盐。
MS(ESI):m/z 586.9[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.54(s,3H),8.38(s,1H),7.80(d,J=5.6Hz,1H),7.38(dd,J=8.0,2.8Hz,1H),7.23(td,J=8.8,4.4Hz,1H),7.01(dd,J=8.8,4.4Hz,1H),6.05(d,J=5.6Hz,1H),4.77–4.69(m,1H),4.56(s,2H),4.20–4.02(m,5H),3.98–3.85(m,2H),3.41–3.27(m,2H),2.70–2.57(m,1H),2.16(t,J=13.2,4.4Hz,1H),2.07–1.76(m,3H),1.05(s,6H)。
实施例21:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲氧基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物71)的制备。
Figure PCTCN2021099561-appb-000126
采用实施例5类似的方法合成化合物71的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物71的三氟乙酸盐。
MS(ESI):m/z 521.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.33(s,1H),7.99(s,3H),7.80(d,J=5.6Hz,1H),6.06(d,J=5.2Hz,1H),4.52(s,2H),4.24–4.20(m,3H),3.92–3.85(m,4H),3.71–3.69(s,2H),3.51(d,J=6.8Hz,2H),3.45–3.41(m,1H),3.28(s,3H),3.14–3.03(m,2H),2.89–2.81(m,1H),1.88–1.83(m,2H),1.76–1.73(m,1H),1.63–1.60(m,1H),1.22(d,J=6.8Hz,3H)。
实施例22:(R)-2-(1-(4-(5-(3-氨基-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物72)的制备。
Figure PCTCN2021099561-appb-000127
采用实施例13类似的方法合成化合物72的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物72的三氟乙酸盐。
MS(ESI):m/z 569.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.44(s,3H),8.38(s,1H),7.80(d,J=5.4Hz,1H),7.57(d,J=7.4Hz,1H),7.38(t,J=7.4Hz,1H),7.02(dd,J=16.4,8.0Hz,2H),6.05(d,J=5.4Hz,1H),4.73–4.68(m,1H),4.56(s,2H),4.10(d,J=7.6Hz,4H),3.97–3.92(m,1H),3.45–3.43(m,2H),3.33–3.30(m,1H),2.66–2.60(m,1H),2.21–2.12(m,1H),2.05–1.89(m,2H),1.82–1.77(m,1H),1.05(s,6H)。
实施例23:(S)-2-(1-(4-(5-(1-氨基-1,3-二氢螺[茚-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物73)的制备。
Figure PCTCN2021099561-appb-000128
采用实施例13类似的方法合成化合物73的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物73的三氟乙酸盐。
MS(ESI):m/z 567.3[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.35(s,1H),8.23(s,3H),7.80(d,J=5.4Hz,1H),7.51(d,J=7.2Hz,1H),7.40–7.29(m,3H),6.03(d,J=5.6Hz,1H),4.54(s,2H),4.44–4.40(m,1H),4.08(d,J=7.6Hz,4H),4.01–3.95(m,1H),3.87–3.84(s,1H),3.29–3.19(m,2H),3.17–3.13(m,1H),3.04–3.00(m,1H),2.65–2.58(m,1H),1.89–1.80(m,2H),1.61–1.51(m,2H),1.05(s,6H)。
实施例24:2-(1-(4-(5-(4-(1-氨基乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物74)的制备。
Figure PCTCN2021099561-appb-000129
采用实施例13类似的方法合成化合物74的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物74的三氟乙酸盐。
MS(ESI):m/z 507.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.33(s,1H),7.79(d,J=5.2Hz,1H),7.69(s,3H),6.01(d,J=5.2Hz,1H),4.51(s,2H),4.08(d,J=7.6Hz,4H),3.85–3.78(m,2H),3.25–3.10(m,3H),2.64–2.58(m,1H),1.69–1.53(m,3H),1.44–1.41(m,1H),1.14(d,J=6.8Hz,3H),1.05(s,6H),1.01(s,3H)。
实施例25:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)-5-甲基吡嗪-2-基)甲醇(化合物75)的制备。
Figure PCTCN2021099561-appb-000130
采用实施例5类似的方法合成化合物75的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物75的三氟乙酸盐。
MS(ESI):m/z 583.1[M+H] +
1H-NMR(400MHz,CD3OD):δ7.64–7.58(m,1H),6.11–6.08(m,1H),4.72–4.60(m,4H),4.41–4.27(m,3H),4.01–3.83(m,4H),3.58(d,J=7.6Hz,2H),3.45(d,J=4.0Hz,1H),3.41–3.33(m,1H),3.21–3.06(m,2H),3.01(s,3H),2.48(s,3H),2.03–1.87(m,3H),1.74–1.71(m,1H),1.32(d,J=6.4Hz,3H)。
实施例26:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物76)的制备。
Figure PCTCN2021099561-appb-000131
采用实施例5第四步和第五步类似的方法合成化合物76的粗品粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物76的三氟乙酸盐。
MS(ESI):m/z 569.0[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.32(s,1H),7.66(d,J=6.0Hz,1H),6.22(d,J=6.0Hz,1H),4.66(s,2H),4.62-4.58(m,2H),4.32-4.25(m,3H),3.97-3.84(m,4H),3.56(d,J=7.6Hz,2H),3.45(d,J=4.0Hz,1H),3.38-3.32(m,1H),3.20-3.07(m,2H),3.00(s,3H),1.99-1.88(m,3H),1.77-1.68(m,1H),1.31(d,J=6.5Hz,3H)。
实施例27:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(羟基甲基)-3-甲基氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物77)的制备。
Figure PCTCN2021099561-appb-000132
采用实施例5类似的方法合成化合物77的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物77的三氟乙酸盐。
MS(ESI):m/z 521.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.34(s,1H),7.95(s,3H),7.79(d,J=5.6Hz,1H),6.03(d,J=5.2Hz,1H),4.52(s,2H),4.25-4.16(m,1H),4.03-4.00(m,2H),3.93-3.81(m,3H),3.76-3.73(m,2H),3.71-3.68(m,1H),3.48-3.40(m,3H),3.13-3.03(m,2H),1.88-1.76(m,2H),1.76-1.71(m,1H),1.65-1.55(m,1H),1.22-1.21(m,6H)。
实施例28:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物78)的制备。
Figure PCTCN2021099561-appb-000133
采用实施例5第四步和第五步类似的方法合成化合物78的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物78。
MS(ESI):m/z 505.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.48(s,1H),8.29(s,1H),8.24(s,1H),7.78(d,J=5.2Hz,1H),5.92(d,J=5.2Hz,1H),4.12-4.03(m,5H),4.00-3.95(m,2H),3.75-3.70(m,1H),3.55-3.53(m,1H),3.37-3.34(m,2H),3.05-3.00(m,1H),2.65-2.58(m,1H),1.79-1.50(m,4H),1.12(d,J=6.4Hz,3H),1.05(s,6H)。
实施例29:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-甲基-3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物79)的制备。
Figure PCTCN2021099561-appb-000134
第一步:3-甲基-3-(对甲苯磺酰氧甲基)-氮杂环丁烷-1-羧酸叔丁酯(化合物29-2)的制备:
将化合物29-1(1g,4.97mmol)溶解在吡啶(10mL)中,降温至0℃,加入TsCl(516mg,5.96mmol)。加完后,升至25℃反应3小时。反应完毕后,将反应液加到水中淬灭,二氯甲烷萃取。合并有机相并干燥、浓缩,得到粗品。粗品经柱层析纯化,得到化合物29-2(900mg,收率48%)。
第二步:3-甲基-3-(甲硫基甲基)-氮杂环丁烷-1-羧酸叔丁酯(化合物29-3)的制备:
将化合物29-2(900mg,2.53mmol)溶解在NMP(10mL)中,20℃加入甲硫醇钠(355mg,5.06mmol)。加完后,加热到70℃反应4小时。反应完毕后,冷却至室温,加入水稀释,乙酸乙酯萃取。合并有机相并干燥、浓缩,得到粗品化合物29-3(600mg),未经纯化直接用于下一步反应。
第三步:3-甲基-3-(甲基磺酰基甲基)氮杂环丁烷-1-羧酸叔丁酯(化合物29-4)的制备:
将化合物29-3(600mg,2.07mmol)溶解在DCM(20mL)中,降温至5℃。分批次加入m-CPBA(787mg,4.56mmol)。加完后,自然升至室温反应4小时。反应完毕后,加入15%碳酸钾水溶液(10mL),搅拌5分钟。分液后,有机相用饱和食盐水洗涤,干燥、浓缩,得到粗品。粗品经柱层析纯化,得到化合物29-4(300mg,收率52%)。
第四步:3-甲基-3-(甲基磺酰基甲基)氮杂环丁烷(化合物29-5)的制备:
将化合物29-4(300mg,1.14mmol)溶解在DCM(2mL)中,加入TFA(1mL),20℃反应1小时。反应完毕后,浓缩除去溶剂,得到化合物29-5(300mg)。
第五步至第九步:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-甲基-3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物79)的制备。
第五步至第九步采用实施例5类似的方法合成化合物79的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物79。
MS(ESI):m/z 583.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.32(s,1H),7.81(d,J=5.2Hz,1H),6.08(d,J=5.2Hz,1H),4.50(s,2H),4.20(d,J=8.4Hz,2H),4.13-4.05(m,1H),3.90(d,J=8.4Hz,2H),3.72-3.66(m,3H),3.60(s,2H),3.54(d,J=8.8Hz,1H),3.27-3.15(m,2H),3.04(d,J=4.8Hz,1H),3.01(m,3H),1.90-1.70(m,2H),1.64-1.60(m,1H),1.56-1.50(m,4H),1.12(d,J=6.4Hz,3H)。
实施例30:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)-2-甲基丙腈(化合物80)的制备。
Figure PCTCN2021099561-appb-000135
采用实施例5类似的方法合成化合物80的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物80。
MS(ESI):m/z 544.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.33(s,1H),8.26(s,1H),7.83(d,J=5.2Hz,1H),6.09(d,J=5.2Hz,1H),5.56-5.49(m,1H),4.51(s,2H),4.29(t,J=8.8Hz,2H),4.11-4.06(m,1H),4.03-3.97(m,2H),3.72-3.63(m,3H),3.52-3.50(m,1H),3.23-3.17(m,2H),2.96(d,J=5.2Hz,1H),2.82-2.75(m,2H),1.87-1.79(m,1H),1.76-1.68(m,1H),1.62-1.49(m,2H),1.29(s,6H),1.09(d,J=6.4Hz,3H)。
实施例31:3-((4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基氨基)甲基)氧杂环丁基-3-醇(化合物81)的制备。
Figure PCTCN2021099561-appb-000136
采用实施例5类似的方法合成化合物81的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物81的三氟乙酸盐。
MS(ESI):m/z 537.1[M+H] +
1H-NMR(400MHz,CD 3OD):δ7.61(d,J=7.2Hz,1H),6.23(d,J=7.2Hz,1H),4.65(s,2H),4.63-4.28(m,2H),4.18-4.10(m,1H),4.05-3.90(m,3H),3.89-3.84(m,1H),3.67-3.60(m,3H),3.50-3.43(m,2H),3.24-3.09(m,2H),2.52(s,3H),2.03-1.80(m,3H),1.76-1.66(m,1H),1.33(d,J=6.8Hz,3H)。
实施例32:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物82)的制备。
Figure PCTCN2021099561-appb-000137
采用实施例13类似的方法合成化合物82的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物82三氟乙酸盐。
MS(ESI):m/z 549.3[M+H] +
1H-NMR(400MHz,DMSO-d 6)δ7.94(s,3H),7.77(d,J=5.4Hz,1H),5.85(d,J=5.4Hz,1H),4.48(s,2H),4.24–4.18(m,1H),4.09–4.07(m,4H),3.96–3.81(m,3H),3.70–3.68(m,1H),3.45–3.43(m,1H),3.12–3.02(m,2H),2.66–2.59(m,1H),2.41(s,3H),1.89–1.71(m,3H),1.62–1.59(m,1H),1.22(d,J=6.4Hz,3H),1.05(s,6H)。
实施例33:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲氧基甲基)-3-甲基氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物83)的制备。
Figure PCTCN2021099561-appb-000138
第一步:3-氯-4-碘-2-(3-(甲氧基甲基)-3-甲基氮杂环丁烷-1-基)吡啶(化合物33-1)的制备:
将化合物27-1(0.30g,0.88mmol)溶于THF(3mL),加入NaH(70.89mg,1.77mmol),25℃反应20分钟后,加入碘甲烷(377.31mg,2.66mmol),25℃反应小时;LCMS检测反应完成后,向反应液中加入水,乙酸乙酯萃取,合并有机相并干燥、浓缩,经薄层色谱法纯化,得化合物33-1(100mg,收率32%)。
第二步至第六步:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(甲氧基甲基)-3-甲基氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物83)的制备:
采用与实施例6第二步至第六步类似的方法合成化合物83的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物83。
MS(ESI):m/z 535.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.27(s,1H),7.68(d,J=5.2Hz,1H),6.07(d,J=5.2Hz,1H),4.66(s,2H),4.28–4.16(m,1H),4.09(d,J=8.4Hz,2H),3.85(dd,J=8.4,4.0Hz,3H),3.77–3.63(m,3H),3.41(s,2H),3.39(s,3H),3.29–3.12(m,2H),3.03(d,J=5.2Hz,1H),1.99–1.81(m,2H),1.77–1.65(m,2H),1.31(s,3H),1.22(d,J=6.4Hz,3H)。
实施例34:2-(1-(4-(5-(4-(1-氨基-2-氟乙基)-4-甲基哌啶-1-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物84)的制备。
Figure PCTCN2021099561-appb-000139
采用实施例13类似的方法合成化合物84的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物84的三氟乙酸盐。
MS(ESI):m/z 525.3[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.33(s,1H),7.60(d,J=5.6Hz,1H),6.22(d,J=5.2Hz,1H),4.81–4.70(m,2H),4.67(s,2H),4.53–4.33(m,4H),3.93–3.77(m,2H),3.51–3.37(m,2H),3.25–3.16(m,1H),2.88–2.73(m,1H),1.91–1.56(m,4H),1.19(m,9H)。
实施例35:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(氧杂环丁烷-3-基甲氧基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物59)的制备。
Figure PCTCN2021099561-appb-000140
采用实施例5类似的方法合成化合物59的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物59的三氟乙酸盐。
MS(ESI):m/z 508.1[M+H] +
1H-NMR(400MHz,CD 3OD-d 4):δ8.39(s,1H),7.94(d,J=7.2Hz,1H),6.90(d,J=6.8Hz,1H),4.94–4.93(m,1H),4.71–4.65(m,3H),4.59(dd,J=13.2,4.2Hz,1H),4.42(dd,J=13.6,8.4Hz,1H),4.34–4.27(m,1H),4.01–3.86(m,4H),3.77–3.71(m,2H),3.46(d,J=4.0Hz,1H),3.24–3.10(m,2H),2.72–2.66(m,1H),2.02–1.87(m,3H),1.76–1.73(m,1H),1.32(d,J=6.8Hz,3H)。
实施例36:2-(1-(4-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物85)的制备。
Figure PCTCN2021099561-appb-000141
第一步:2-(1-(4-(3-氨基-5-氯吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物36-2)的制备:
将化合物36-1(135mg,647.66μmol)、化合物12-3(272.74mg,917.49μmol)、CuI(123.35mg,647.66μmol)、1,10-菲罗啉(116.71mg,647.66μmol)及K 3PO 4(412.43mg,1.94mmol)加入到二氧六环(8mL)中,于微波反应器中升温至105℃,反应2小时。冷至室温,滤除不溶物,浓缩滤液,得到粗品,粗品经薄层色谱法纯化,得到化合物36-2(130mg,收率52%)。
第二步:2-(1-(4-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物85)的制备:
将化合物36-2(50mg,129.43μmol)、化合物A-2(63mg,258.87μmol)、DIPEA(134mg,1.04mmol)加入DMF(3mL)中,体系升温至100℃反应16小时。反应完成后,浓缩反应液得到粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物85(8.10mg,收率11%)。
MS(ESI):m/z 520.2[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ7.76(d,J=5.6Hz,1H),7.63(s,1H),6.14(s,2H),5.84(d,J=5.6Hz,1H),4.10-4.00(m,6H),3.90-3.78(m,3H),3.67(d,J=8.4Hz,1H),3.49(d,J=8.4Hz,1H),2.92(d,J=5.2Hz,1H),2.66–2.56(m,1H),1.76–1.42(m,4H),1.08(d,J=6.8Hz,3H),1.05(s,6H)。
实施例37:(1-(4-(3-氨基-5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)吡咯烷-2-基)甲醇(化合物86)的制备。
Figure PCTCN2021099561-appb-000142
参照实施例36类似的方法合成化合物86的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物86的三氟乙酸盐。
MS(ESI):m/z 506.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ7.98(s,3H),7.80(d,J=5.2Hz,1H),7.69(s,1H),6.23(s,2H),5.92(d,J=5.2Hz,1H),4.46–4.39(m,1H),4.25–4.18(m,2H),4.16–4.12(m,1H),3.88–3.80(m,2H),3.69(d,J=9.2Hz,1H),3.52(dd,J=10.4,3.6Hz,1H),3.43–3.34(m,2H),3.22(dd,J=10.4,7.2Hz,1H),3.13–3.00(m,2H),2.08–1.99(m,1H),1.97–1.89(m,1H),1.86–1.78(m,1H),1.70–1.67(m,4H),1.57–1.54(m,1H),1.22(d,J=6.4Hz,3H)。
实施例38:(3S,4S)-8-(6-氨基-5-(3-氯-2-(3-(甲基磺酰基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-4-胺(化合物87)的制备。
Figure PCTCN2021099561-appb-000143
参照实施例36类似的方法合成化合物87的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物87。
MS(ESI):m/z 554.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ7.79(d,J=5.2Hz,1H),7.63(s,1H),6.14(s,2H),5.91(d,J=5.2Hz,1H),4.31(t,J=8.4Hz,2H),4.10–4.03(m,1H),4.03–3.96(m,2H),3.87–3.79(s,2H),3.66(d,J=8.4Hz,1H),3.53–3.47(m,3H),3.43–3.38(m,1H),3.20–3.07(m,2H),2.98(s,3H),2.89(d,J=5.2Hz,1H),1.76–1.69(m,1H),1.64–1.58(m,1H),1.53–1.42(m,2H),1.08(d,J=6.4Hz,3H)。
实施例39:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-氟-3-(羟基甲基) 氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物88)的制备。
Figure PCTCN2021099561-appb-000144
采用实施例6类似的方法合成化合物88的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物88。
MS(ESI):m/z 525.2[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.35(s,1H),7.95(s,3H),7.86(d,J=5.6Hz,1H),6.15(d,J=5.6Hz,1H),4.52(s,2H),4.33(d,J=10.4Hz,1H),4.28(d,J=10.4Hz,1H),4.24-4.10(m,4H),3.9-3.60(m,6H),3.48-3.38(m,1H),3.17-3.30(m,2H),1.92-1.55(m,4H),1.22(d,J=6.4Hz,3H)。
实施例40:2-(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氟吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物90)的制备。
Figure PCTCN2021099561-appb-000145
采用实施例6类似的方法合成化合物90的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的甲酸水溶液)纯化,得到化合物90。
MS(ESI):m/z 519.1[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.23(s,1H),7.61(d,J=5.6Hz,1H),6.40–6.05(m,1H),4.64(s,2H),4.28–4.17(m,1H),4.17–4.03(m,4H),3.85(d,J=8.8Hz,1H),3.71(d,J=8.8Hz,1H),3.69–3.59(m,2H),3.25–3.07(m,2H),3.03(d,J=5.2Hz,1H),2.86–2.76(m,1H),1.98–1.81(m,2H),1.78–1.64(m,2H),1.22(d,J=6.8Hz,3H),1.18(s,6H)。
实施例41:(R)-2-(1-(4-(5-(3-氨基-5-氟-3H-螺[苯并呋喃-2,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物107)的制备。
Figure PCTCN2021099561-appb-000146
采用实施例13类似的方法合成化合物107的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物107的三氟乙酸盐。
MS(ESI):m/z 557.0[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.59(d,J=0.8Hz,1H),8.55(s,3H),8.36(d,J=0.8Hz,1H),7.80(d,J=5.2Hz,1H),7.38(dd,J=8.0,2.8Hz,1H),7.23(td,J=8.8,2.8Hz,1H),7.01(d,J=9.2,4.4Hz,1H),5.95(d,J=5.2Hz,1H),4.77–4.65(m,1H),4.60–4.51(m,1H),4.46–4.36(m,1H),4.08(d,J=7.6Hz,4H),3.42–3.25(m,2H),2.70–2.55(m,1H),2.10–1.94(m,2H),1.90–1.76(m,2H),1.05(s,6H)。
实施例42:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(氟甲基)-3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物124)的制备。
Figure PCTCN2021099561-appb-000147
第一步:1-叔丁基3-甲基3-(氟甲基)氮杂环丁烷-1,3-二羧酸酯(化合物42-2)的制备:
将42-1(450mg,1.93mmol)用DMF(5mL)溶解,加入碳酸钾(533mg,3.86mmol)和碘甲烷(411mg,2.89mmol),25℃反应2小时。加入饱和氯化铵淬灭反应,乙酸乙酯萃取,合并有机相,干燥,减压浓缩后经硅胶柱层析纯化,得化合物42-2(345mg,收率72%)。
第二步:3-(氟甲基)-3-(羟基甲基)氮杂环丁烷-1-羧酸叔丁酯(化合物42-3)的制备:
将化合物42-2(320mg,1.29mmol)加入甲醇(10mL)中,加入硼氢化钠(143mg,3.88mmol),25℃反应2小时。反应完成后,冷至室温,向体系中加入水,分出有机相,食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩干,得粗品化合物42-3(280mg),未经纯化直接用于下一步反应。
第三步:(3-(氟甲基)氮杂环丁烷-3-基)甲醇盐酸盐(化合物42-4)的制备:
将化合物42-3(280mg,1.27mmol)用4M HCl/EA溶液(5mL)溶解,25℃反应2小时。减压抽干溶剂,得化合物42-4粗品(210mg),未经纯化直接用于下一步反应。
第四步至第九步:(3-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(3-氯-2-(3-(氟甲基)-3-(羟基甲基)氮杂环丁烷-1-基)吡啶-4-基硫基)吡嗪-2-基)甲醇(化合物124)的制备:
采用实施例12第一步至第六步所描述的类似的方法合成化合物124的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物124的三氟乙酸盐。
MS(ESI):m/z 538.9[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.35(s,1H),7.97(brs,3H),7.81(d,J=5.6Hz,1H),6.06(d,J=5.6Hz,1H),4.65(s,1H),4.53(s,1H),4.52(s,2H),4.25–4.17(m,1H),4.05–3.78(m,7H),3.69(d,J=8.8Hz,1H),3.60(s,2H),3.38–3.34(m,1H),3.17–3.02(m,2H),1.92–1.79(m,2H),1.79–1.70(m,1H),1.65–1.54(m,1H),1.21(d,J=6.8Hz,3H)。
实施例43:(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物89)的制备。
Figure PCTCN2021099561-appb-000148
采用实施例13类似的方法合成化合物89的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物89的三氟乙酸盐。
MS(ESI):m/z 568.0[M+H] +
1H NMR(400MHz,DMSO)δ8.56(dd,J=5.2,1.6Hz,1H),8.44(br,3H),8.36(s,1H),7.96(d,J=7.6Hz,1H),7.80(d,J=5.6Hz,1H),7.39(dd,J=8.0,5.2Hz,1H),6.05(d,J=5.2Hz,1H),4.51(s,2H),4.59–4.45(m,1H),4.17–4.06(m,4H),4.05–3.84(m,2H),3.36–3.17(m,3H),3.12(d,J=17.2Hz,1H),2.68–2.57(m,1H),1.97–1.82(m,2H),1.67–1.50(m,2H),1.05(s,6H)。
实施例44:(R)-2-(1-(4-(5-(3-氨基-3H-螺[呋喃并[2,3-b]吡啶-2,4'-哌啶]-1'-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物101)的制备。
Figure PCTCN2021099561-appb-000149
采用实施例13类似的方法合成化合物101的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物101的三氟乙酸盐。
MS(ESI):m/z 570.0[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.60(brs,3H),8.39(s,1H),8.22(dd,J=5.2,1.6Hz,1H),7.97(dd,J=7.2,1.2Hz,1H),7.80(d,J=5.2Hz,1H),7.09(dd,J=7.6,5.2Hz,1H),6.06(d,J=5.2Hz,1H),4.84–4.73(m,1H),4.57(s,2H),4.20–4.10(m,1H),4.08(d,J=7.6Hz,4H),4.02–3.94(m,1H),3.44–3.28(m,2H),2.70–2.57(m,1H),2.25–2.12(m,1H),2.08–1.90(m,2H),1.90–1.77(m,1H),1.05(s,6H)。
实施例45:(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物103)的制备。
Figure PCTCN2021099561-appb-000150
采用实施例13类似的方法合成化合物103的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物103的三氟乙酸盐。
MS(ESI):m/z 538.0[M+H] +
1H-NMR(400MHz,CD 3OD):δ8.64-8.54(m,1H),8.41(d,J=1.2Hz,1H),8.33(d,J=1.2Hz,1H),7.98(d,J=7.6Hz,1H),7.59(d,J=6.4Hz,1H),7.42(dd,J=7.6,5.2Hz,1H),6.14(d,J=6.4Hz,1H),4.64–4.32(m,7H),3.52–3.30(m,4H),2.88–2.78(m,1H),1.95–1.75(m,3H),1.74–1.65(m,1H),1.20(s,6H)。
实施例46:(S)-2-(1-(4-(3-氨基-5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物115)的制备。
Figure PCTCN2021099561-appb-000151
参照实施例36类似的方法合成化合物115的粗品,粗品经Pre-TLC(MeOH/DCM=1:5,V/V)纯化,得到化合物115。
MS(ESI):m/z 553.0[M+H] +
1H NMR(400MHz,DMSO)δ8.31(d,J=4.0Hz,1H),7.77(d,J=5.2Hz,1H),7.71–7.61(m,2H),7.21–7.13(m,1H),6.15(s,2H),5.86(d,J=5.6Hz,1H),4.44(s,1H),4.30–4.17(m,2H),4.12–3.99(m,4H),3.89(s,1H),3.22–3.06(m,3H),2.75(d,J=16.4Hz,1H),2.67–2.56(m,1H),1.82–1.62(m,2H),1.57–1.48(m,1H),1.18–1.07(m,1H),1.05(s,6H)。
实施例47:1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)-3-(羟基甲基)氮杂环丁烷-3-甲腈(化合物126)的制备。
Figure PCTCN2021099561-appb-000152
除在第一步中使用3-(羟基甲基)氮杂环丁烷-3-碳腈代替实施例1的第一步中的2-羟基甲基吡咯烷,第四步中使用化合物4-1代替实施例1的第四步中的化合物IM-1外,采用与实施例1中第一步至第四步以及第六步所描述的类似方法合成化合物126的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化得到化合物126的三氟乙酸盐。
MS(ESI):m/z 532.0[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.35(s,1H),7.96(s,3H),7.87(d,J=5.6Hz,1H),6.18(d,J=5.6Hz,1H),4.52(s,2H),4.35(d,J=8.4Hz,2H),4.22(d,J=8.8Hz,2H),4.21–4.16(m,1H),3.96–3.83(m,1H),3.87(d,J=8.8Hz,1H),3.81(s,2H),3.70(d,J=8.8Hz,1H),3.50–3.40(m,1H),3.16–3.01(m,2H),1.92–1.55(m,4H),1.22(d,J=6.4Hz,3H)。
实施例48:(1-(4-(5-((3S,4S)-4-氨基-3-甲基-2-氧杂-8-氮杂螺[4.5]癸烷-8-基)吡嗪-2-基硫基)-3-氯吡啶-2-基)-3-氟氮杂环丁烷-3-基)甲醇(化合物138)的制备。
Figure PCTCN2021099561-appb-000153
采用实施例13类似的方法合成化合物138的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物138的三氟乙酸盐。
MS(ESI):m/z 495.0[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.53(s,1H),8.33(s,1H),7.95(s,3H),7.84(d,J=5.6Hz,1H)6.04(d,J=5.2Hz,1H),4.33-4.10(m,7H),3.91-3.88(m,1H),3.73-3.68(m,3H),3.23-3.14(m,3H),1.79-1.69(m,3H),1.60-1.57(m,1H),1.21(d,J=6.8Hz,3H)。
实施例49:2-(1-(4-(5-(4-氨基-8-氮杂二螺[2.1.5.2]十二烷-8-基)-6-(羟基甲基)吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物139)的制备
Figure PCTCN2021099561-appb-000154
采用实施例13类似的方法合成化合物139的粗品,粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物139的三氟乙酸盐。
MS(ESI):m/z 545.0[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.34(s,1H),7.78(d,J=5.2Hz,1H),7.68(brs,3H),6.01(d,J=5.2Hz,1H),4.52(s,2H),4.07(d,J=7.6Hz,4H),3.90–3.70(m,2H),3.35–3.24(m,1H),3.22–3.12(m,1H),2.94–2.82(m,1H),2.66–2.58(m,1H),2.05–1.70(m,6H),1.66–1.50(m,2H),1.05(s,6H),1.00–0.90(m,1H),0.84–0.73(m,1H),0.64–0.53(m,1H),0.53–0.45(m,1H)。
实施例50:(S)-2-(1-(4-(5-(5-氨基-5,7-二氢螺[环戊二烯并[b]吡啶-6,4'-哌啶]-1'-基)-6-(羟基甲基)-3-甲基吡嗪-2-基硫基)-3-氯吡啶-2-基)氮杂环丁烷-3-基)丙-2-醇(化合物140)的制备。
Figure PCTCN2021099561-appb-000155
采用实施例13类似的方法合成化合物140的粗品,粗品经HPLC(流动相A:乙腈,流动相B:0.05%的三氟乙酸水溶液)纯化,得到化合物140的三氟乙酸盐。
MS(ESI):m/z 582.3[M+H] +
1H-NMR(400MHz,DMSO-d 6):δ8.55(d,J=4.8Hz,1H),8.38(s,3H),7.93(d,J=7.6Hz,1H),7.77(d,J=5.2Hz,1H),7.37(dd,J=4.8Hz,7.6Hz,1H),5.87(d,J=5.2Hz,1H),4.50(s,3H),4.10(d,J=7.6Hz,2H),4.06-3.92(m,2H),3.28-3.20(m,3H),3.12-3.08(m,1H),2.66-2.59(m,1H),252-2.50(m,2H),2.42(s,3H),1.92-1.83(m,2H),1.64-1.55(m,2H),1.05(s,6H)。
对比化合物1、2和TNO-155的合成:分别参照WO2019075265、WO2018013597、WO2015107495中所描述的方法合成对比化合物1、2和TNO-155。
Figure PCTCN2021099561-appb-000156
试验例1:SHP2(蛋白磷酸酶)体外酶学活性抑制试验。
1、试验***:
磷酸酶:人源PTPN11全长序列重组蛋白(SHP2),有活性的(SignalChem);
底物:6,8-二氟-4-甲基伞形酮基磷酸酯(DiFMUP)(Invitrogen);
激活肽:IRS1_pY1172(dPEG8)pY1222(IRS1)(BPS Bioscience);
终止试剂:bpv(phen)(bpv)(Abcam)。
2、试验参数:
SHP2浓度:0.5nM;
DiFMUP浓度:200μM;
IRS-1浓度:0.5μM;
bpv浓度:160μM;
缓冲液体系:60mM Hepes pH7.2;75mM NaCl;75mM KCl;0.05%surfactant P20;1mM EDTA;5mM DTT;
化合物和酶孵育时间:60分钟;
酶与底物反应时间:30分钟;
酶标仪参数:BMG PHERAstar荧光酶标仪,激发波长340nm,发射波长450nm。
3、试验步骤:
测试组:将待测化合物和磷酸酶SHP2的混合物与激活肽IRS-1在缓冲液体系下于室温孵育60分钟,加入底物DiFMUP启动反应,于室温孵育30分钟,然后加入bpv终止反应,将反应板放入酶标仪中,采用终点法读取板中各孔的荧光值。
阴性组:用0.05%DMSO水溶液代替待测化合物,实验方法与测试组相同。
空白组:用0.05%DMSO水溶液代替待测化合物,并且不加SHP2,实验方法与测试组相同。
4、数据处理:
计算各浓度组的相对抑制活性,抑制率(%)=100%-(测试组的荧光值-空白组的荧光值)/(阴性组的荧光值-空白组的荧光值)×100%。按照四参数模型拟合曲线,计算化合物的半数抑制浓度(IC 50)。
5、试验结果:
按照上述方法测定化合物对SHP2活性的抑制作用,结果如表1中所示。
表1.SHP2酶活抑制试验结果
组别 IC 50(nM) 组别 IC 50(nM)
实施例1 6.03 实施例25 7.00
实施例2 1.82 实施例26 0.70
实施例3 3.48 实施例27 4.10
实施例4 4.10 实施例28 2.45
实施例5 1.22 实施例29 1.68
实施例6 1.76 实施例30 4.00
实施例7 4.60 实施例32 3.70
实施例8 0.88 实施例33 4.10
实施例9 1.30 实施例37 2.40
实施例10 1.22 实施例38 5.30
实施例11 9.30 实施例39 1.40
实施例12 2.50 实施例40 4.13
实施例13 4.90 实施例41 8.40
实施例15 2.40 实施例42 10.19
实施例16 1.00 实施例43 2.60
实施例17 2.50 实施例44 2.49
实施例18 1.90 实施例45 4.10
实施例19 1.50 实施例46 2.60
实施例20 6.90 实施例47 3.20
实施例21 1.15 实施例48 2.20
实施例22 4.30 实施例49 2.30
实施例23 1.90 实施例50 4.84
6、结论:
在SHP2酶活抑制试验中,本发明的化合物表现出较强的抑制活性。
试验例2:KYSE-520细胞(人食管鳞癌细胞)增殖活性抑制试验。
1、试验***:
细胞:KYSE-520(JCRB Cell Bank);
试剂盒:
Figure PCTCN2021099561-appb-000157
发光法细胞活力分析试剂盒(Promega)。
2、试验参数:
细胞数量:1500细胞/孔;
铺板培养基:KYSE-520:1640+10%FBS;
加药培养基:KYSE-520:1640+10%FBS;
化合物孵育条件:37℃,5%CO 2
孵育时间:5天;
检测温度:室温;
BMG PHERAstar FS检测化学发光。
3、试验步骤:
将细胞培养在含有10%胎牛血清的培养基中,放置在37℃,5%CO 2培养条件下进行培养。向96孔板中铺入适量细胞,培养箱中过夜培养,使细胞附着贴壁。次日,移除培养基,加入含有预先稀释化合物的完全培养基,于37℃孵育5天。第五天,向每孔中加入检测试剂
Figure PCTCN2021099561-appb-000158
化学发光检测各孔的相对发光单位(RLU)。
4、数据处理:
利用不含细胞的培养基的
Figure PCTCN2021099561-appb-000159
获得背景值。细胞活率(%)=(样品RLU-背景RLU)/(溶媒RLU-背景RLU)×100%,最大抑制率(%)=100%-细胞活率 最大浓度(%)。按照四参数模型拟合曲线,计算化合物的半数抑制浓度(IC 50)。
5、试验结果:
按照上述方法测定本发明的化合物对KYSE-520细胞增殖的抑制活性,结果如表2中所示。
表2.化合物对KYSE-520细胞增殖活性抑制结果
实施例编号 KYSE-520,IC 50(μM) 实施例编号 KYSE-520,IC 50(μM)
实施例1 0.36 实施例21 0.11
实施例2 0.10 实施例22 0.05
实施例3 1.14 实施例23 0.14
实施例4 0.17 实施例26 0.03
实施例5 0.06 实施例27 0.06
实施例6 0.04 实施例29 0.04
实施例7 0.77 实施例33 0.09
实施例8 0.03 实施例39 0.02
实施例9 0.22 实施例42 0.10
实施例10 0.04 实施例43 0.04
实施例12 0.04 实施例44 0.05
实施例15 0.04 实施例45 0.21
实施例16 0.04 实施例46 0.07
实施例17 0.03 实施例47 0.04
实施例18 0.24 实施例48 0.64
实施例19 0.19 实施例49 0.27
6、结论:
本发明的化合物对KYSE-520细胞具有较强的细胞增殖抑制活性。
试验例3:生化hERG抑制试验。
1、试验***:
试剂盒:Predictor TM hERG荧光极化分析试剂盒(ThermoFisher),其含有:
作为阳性对照的化合物E4031;
hERG细胞膜;
亲和性示踪剂;和
hERG缓冲液。
2、试验参数:
hERG浓度:1×;
示踪剂浓度:1nM;
孵育时间:2小时;
酶标仪参数:BMG PHERAstar FS荧光酶标仪。
3、试验步骤:
测试组:将不同浓度的待测化合物加入到含有hERG细胞膜的微孔板中,每孔中再加入具有高hERG亲和性示踪剂,将微孔板在25℃孵育2小时后,使用多功能酶标仪检测荧光偏振(激发波长:540;发射波长:590nm)值(mP)的变化。
阳性对照组:用30μM的化合物E4031代替待测化合物,实验方法与测试组相同。
空白对照组:用hERG缓冲液代替待测化合物,并且不加hERG细胞膜,实验方法与测试组相同。
4、数据处理:
根据数据比值,计算本发明的化合物在不同浓度下对hERG百分比抑制率(%),判断化合物的半数抑制浓度(IC 50)的范围。百分比抑制率(%)=(1-(待测化合物的mP-阳性对照组的mP)/(空白对照组的mP-阳性对照组的mP))×100%。
5、试验结果:
采用上述方法测定本发明的化合物对hERG的抑制,结果如表3中所示。
表3.hERG抑制试验结果
实施例编号 IC 50(μM) 实施例编号 IC 50(μM)
实施例2 >10 实施例27 >10
实施例4 >10 实施例28 >10
实例例7 >10 实施例29 >10
实施例9 >10 实施例30 >10
实施例12 >10 实施例39 >10
实施例13 >10 实施例40 >10
实施例15 >10 实施例42 >10
实施例18 >10 实施例46 >10
实施例19 >10 实施例47 >10
实施例25 >10 实施例48 >10
实施例26 >10    
6、结论:
测试结果表明,本发明的化合物与hERG亲和性低,与亲和性示踪剂竞争的IC 50大于10μM。
试验例4:NCI-H358细胞(人非小细胞肺癌细胞)增殖活性抑制试验。
1、试验***:
细胞:NCI-H358(南京科佰);
试剂盒:
Figure PCTCN2021099561-appb-000160
发光法细胞活力分析试剂盒(Promega)。
2、试验参数:
细胞数量:1500细胞/孔;
铺板培养基:NCI-H358:1640+10%FBS;
加药培养基:NCI-H358:1640+10%FBS;
化合物孵育条件:37℃,5%CO 2
孵育时间:5天;
检测温度:室温;
BMG PHERAstar FS检测化学发光。
3、试验步骤:
将细胞培养在含有10%胎牛血清的培养基中,放置在37℃,5%CO 2培养条件下进行培养。向96孔板中铺入适量细胞,培养箱中过夜培养,使细胞附着贴壁。次日,移除培养基,加入含有预先稀释化合物的完全培养基,于37℃孵育5天。第五天,向每孔中加入检测试剂
Figure PCTCN2021099561-appb-000161
化学发光检测各孔的相对发光单位(RLU)。
4、数据处理:
利用不含细胞的培养基的
Figure PCTCN2021099561-appb-000162
获得背景值。细胞活率(%)=(样品RLU-背景RLU)/(溶媒RLU-背景RLU)×100%,最大抑制率(%)=100%-细胞活率 最大浓度(%)。按照四参数模型拟合曲线,计算化合物的半数抑制浓度(IC 50)。
5、试验结果:
按照上述方法测定本发明的化合物对NCI-H358细胞增殖的抑制活性,结果如表4中所示。
表4.化合物对NCI-H358细胞增殖活性抑制结果
实施例编号 NCI-H358,IC 50(μM) 实施例编号 NCI-H358,IC 50(μM)
实施例2 0.040 实施例25 0.150
实施例5 0.034 实施例26 0.032
实施例6 0.074 实施例27 0.030
实施例12 0.018 实施例28 0.130
实施例13 0.260 实施例29 0.040
实施例15 0.040 实施例40 0.030
实施例17 0.039 实施例42 0.044
实施例19 0.220 实施例43 0.008
实施例21 0.030 实施例44 0.050
实施例22 0.031 实施例46 0.028
实施例23 0.005 实施例47 0.023
6、结论:
测试结果表明,本发明的化合物对NCI-H358细胞具有较强的细胞增殖抑制活性。
试验例5:生化CYP酶(细胞色素P450)抑制试验。
1、试验***:
P450-Glo TMCYP1A2 Screening System,(Promega);
P450-Glo TMCYP2D6 Screening System,(Promega);
P450-Glo TMCYP3A4 Screening System,(Promega)。
2、测试仪器:
BMG PHERAstar FS Luminescent。
3、试验方法:
分别按照试剂盒说明书进行试验,步骤如下:
3.1.对CYP1A2的抑制:
测试组:将不同浓度的待测化合物加入到微孔板中,向每孔中加入Luciferin-ME(100μM)、K 3PO 4(100mM)和CYP1A2(0.01pmol/μL),在室温下预孵育10min,随后加入NADPH再生***,在室温下反应30min,最后加入等体积的检测缓冲液,在室温下孵育20min,然后进行化学发光检测。
阴性对照组:实验方法同测试组,只是不加待测化合物。
空白对照组:实验方法同测试组,只是不加待测化合物,并且用CYP1A2 Membrance(0.01pmol/μL)代替CYP1A2。
3.2.对CYP2D6的抑制:
测试组:将不同浓度的待测化合物加入到微孔板中,向每孔中加入Luciferin-ME EGE(3μM)、K 3PO 4(100mM)和CYP2D6(5nM),在室温下预孵育10min,随后加入NADPH再生***,在37℃下反应30min,最后加入等体积的检测缓冲液,在室温下孵育20min,然后进行化学发光检测。
阴性对照组:实验方法同测试组,只是不加待测化合物。
空白对照组:实验方法同测试组,只是不加待测化合物,并且用CYP2D6 Membrance(5nM)代替CYP2D6。
3.3.对CYP3A4的抑制:
测试组:将不同浓度的待测化合物加入到微孔板中,向每孔中加入Luciferin-IPA(3μM)、K 3PO 4(100mM)和CYP3A4(2nM),在室温下预孵育10min,随后加入NADPH再生***,在室温下反应30min,最后加入等体积的检测缓冲液,在室温下孵育20min,然后进行化学发光检测。
阴性对照组:实验方法同测试组,只是不加待测化合物。
空白对照组:实验方法同测试组,只是不加待测化合物,并且用CYP3A4 Membrance(2nM)代替CYP3A4。
4、数据处理:
百分比抑制率=(1-(待测化合物浓度组的化学发光信号值-空白对照组的化学发光信号值)/(阴性 对照组的化学发光信号值-空白对照组的化学发光信号值))×100%。
当百分比抑制率介于30-80%之间时,根据下述公式,估算化合物对CYP酶的半数抑制浓度(IC 50)或范围:IC 50=X×(1-百分比抑制率)/百分比抑制率,其中X为化合物的测试浓度。
5、试验结果:
按照上述方法测定本发明化合物对三种CYPs的抑制,结果如下表5中所示。
表5.CYPs抑制试验结果
Figure PCTCN2021099561-appb-000163
6、结论:
上述结果表明,本发明化合物对3种主要CYP亚型均无明显抑制作用,表明其潜在的药物相互作用可能性相对较低,优于对比化合物1。
试验例6:SD大鼠药代动力学(PK)研究。
分别通过静脉(IV)和灌胃(PO)向雄性SD大鼠给药实施例12化合物及对比化合物2,考察药代动力学特性。IV和PO的给药剂量分别是1mg/kg和5mg/kg,IV的溶剂为5%DMSO+5%Solutol(15-羟基硬脂酸酯聚乙二醇酯)+90%Saline(生理盐水),PO的溶剂为0.5%MC(甲基纤维素钠)。IV和PO给药后在不同时间点收集血液。血液采用EDTA.K2抗凝,离心后得到血浆样品,于-80℃条件下保存。
血浆样品经沉淀蛋白处理后进行LC-MS/MS分析。应用WinNonlin 6.3软件,采用非房室模型计算药代动力学参数,结果见下表6。
表6.化合物在大鼠体内血液中的药代动力学参数
Figure PCTCN2021099561-appb-000164
注:“/”表示不存在
结论:本发明实施例12的化合物在注射给药剂量1mg/kg和口服给药剂量5mg/kg的条件下,化合物在大鼠血浆中达到较高暴露,远高于对比化合物2,且清除率远低于对比化合物2,表现出优良的药代动力学性质。
试验例7:Balb/c小鼠药代动力学(PK)研究。
分别通过静脉(IV)和灌胃(PO)向雌性Balb/c小鼠给药实施例12化合物、对比化合物1、对比化合物2及TNO-155,考察药代动力学特性。IV和PO的给药剂量分别是1mg/kg和10mg/kg,IV的溶剂为5%DMSO+5%Solutol+90%Saline,PO的溶剂为0.5%MC(甲基纤维素钠)。IV和PO给药后在不同时间点收集血液。实施例12、对比化合物1和对比化合物2试验组的血液采用EDTA.K2抗凝,于-80℃条件下保存;TNO-155试验组的血液采用EDTA.K2抗凝,经离心后得到血浆样品,于-80℃条件下保存。
血液或血浆样品经沉淀蛋白处理后进行LC-MS/MS分析。应用WinNonlin 6.3软件,采用非房室模型计算药代动力学参数,结果见下表7。
表7.化合物在Balb/c小鼠体内血液中的药代动力学参数
Figure PCTCN2021099561-appb-000165
注:“/”表示不存在
结论:本发明实施例12的化合物在注射给药剂量1mg/kg和口服给药剂量10mg/Kg,化合物在小鼠体循环中达到较高暴露,较低的清除率及较长的半衰期,综合性质显著优于对比化合物1、2以及TNO-155,表现出优良的药代动力学性质。
试验例8:比格犬药代动力学(PK)研究。
分别通过静脉(IV)和灌胃(PO)向雄性比格犬给药实施例12化合物、对比化合物1、对比化合物2及TNO-155,考察药代动力学特性。IV和PO的给药剂量分别是0.5mg/kg和2.5mg/kg,IV的溶剂为5%DMSO+5%Solutol+90%Saline,PO的溶剂为0.5%MC(甲基纤维素钠)。IV和PO给药后在不同时间点收集血液。血液采用EDTA.K2抗凝,离心后得到血浆样品,于-80℃条件下保存。
血浆样品经沉淀蛋白处理后进行LC-MS/MS分析。应用WinNonlin 6.3软件,采用非房室模型计算药代动力学参数,结果见下表8。
表8.化合物在比格犬体内血液中的药代动力学参数
Figure PCTCN2021099561-appb-000166
注:“/”表示不存在
结论:本发明实施例12的化合物在注射给药剂量0.5mg/kg和口服给药剂量2.5mg/Kg,化合物在比格犬血浆中达到较高暴露,且具有较长的半衰期及较低的清除率,综合性质优于对比化合物1、对比化合物2及TNO-155,表现出较好的药代动力学性质。
试验例9:KYSE-520移植瘤模型上对肿瘤的抑制试验。
1、试验目的:
通过nu-nu小鼠皮下接种KYSE-520细胞构建CDX移植瘤动物模型,成瘤后每天一次进行口服给药,评价不同受试化合物体内药效。
2、试验试剂:
Figure PCTCN2021099561-appb-000167
3、试验方法:
将KYSE-520细胞体外单层培养,培养条件为RPMI 1640培养基中加10%胎牛血清,于37℃、含5%CO 2空气的培养箱中培养。2次/周用胰蛋白酶进行消化处理传代。当细胞呈指数生长期时,收集细胞并计数。将培养至对数生长期的KYSE-520细胞株制备成单细胞悬液(细胞重悬于PBS中,与无酚红基质胶1:1混合,细胞终密度为5×10 7/ml)每只裸鼠接种0.1ml,接种至裸鼠右侧腋窝皮下后构建食管癌裸鼠移植瘤模型,待肿瘤生长至130mm 3时,将裸鼠进行随机分成5组进行给药,给药方案如表9所示(每天口服给药1次,持续21天),溶媒为5%DMSO+5%Solutol+90%H 2O。
每天监测动物的健康状况及死亡情况,例行检查包括观察药物治疗对动物日常行为表现的影响,如行为活动、摄食(水)量、外观体征或其它异常情况,并做好相应记录。
每周2次称量小鼠体重并测量瘤体积,记录数据。肿瘤体积(V)计算公式:V=1/2×a×b 2,其中a和b分别表示长和宽。小鼠移植瘤体积如图1所示。
抗肿瘤药效用肿瘤生长抑制率TGI(%)评价,其中当肿瘤未出现消退时,TGI(%)(瘤体积)=[1-(TVt-TV0)/(CVt-CV 0)]×100%,TV0为分组时受试化合物组的平均瘤体积,TVt为给药后t天受试化合物组的平均瘤体积;CV 0为分组时溶媒组的平均瘤体积;CVt为给药后t天溶媒组的平均瘤体积;当肿瘤出现消退时,TGI(%)(瘤体积)=100%-(TVt-TV 0)/TV 0×100%。
4、试验结果:
表9.受试化合物对KYSE-520细胞移植瘤模型的疗效(以瘤体积计算)
Figure PCTCN2021099561-appb-000168
注:“-”表示不存在,P<0.05表示与溶媒组相比有显著差异。
5、试验结论:
以上数据显示,口服连续给药21天后,本发明的实施例12化合物抑瘤效果显著,药效优于以相同剂量给药的阳性化合物TNO-155的效果。
试验例10:NCI-H358移植瘤模型上对肿瘤的抑制试验。
1、试验目的:
通过将人非小细胞肺癌细胞NCI-H358皮下接种于Balb/c-nu小鼠右侧肩胛处,构建皮下移植瘤小鼠模型,成瘤后进行口服给药,评价不同受试化合物体内药效。
2、试验试剂:
Figure PCTCN2021099561-appb-000169
3、试验方法:
将NCI-H358细胞体外单层培养,培养条件为RPMI1640培养基中加10%胎牛血清,于37℃、5%CO 2的培养箱中培养。一周2-3次用胰酶-EDTA进行消化处理传代。当细胞呈指数生长期时,收集细胞,计数,接种。
在肿瘤细胞接种前,试验小鼠用小鼠专用编号耳钉依次进行动物标识。
每只小鼠右侧肩胛皮下接种1×10 6个NCI-H358细胞(悬浮于0.1ml PBS+基质胶)。预计第七天肿瘤体积均值为~200mm 3,根据第7天的测量结果,筛选出30只肿瘤形状规则、体积均一的小鼠,并将其进行随机分为5组,并按试验方案给药(给药方案如表10所示(持续21天)),溶媒为5%DMSO+5%Solutol+90%H 2O。
每周3次称量小鼠体重并测量瘤体积,记录数据。肿瘤体积(V)计算公式:V=1/2×a×b 2,其中a和b分别表示长和宽。小鼠移植瘤体积如图2所示。
抗肿瘤药效用肿瘤生长抑制率TGI(%)评价,其中当肿瘤未出现消退时,TGI(%)(瘤体积)=[1-(TV t-TV 0)/(CV t-CV 0)]×100%,TV 0为分组时受试化合物组的平均瘤体积,TV t为给药后t天受试化合物组的平均瘤体积;CV 0为分组时溶媒组的平均瘤体积;CV t为给药后t天溶媒组的平均瘤体积;当肿瘤出现消退时,TGI(%)(瘤体积)=100%-(TV t-TV 0)/TV 0×100%。
如果肿瘤比起始体积缩小,即V t<V 0时,即定义为肿瘤部分消退(PR);如果肿瘤完全消失,即定义为肿瘤完全消退(CR)。
4、试验结果:
表10.受试化合物对NCI-H358细胞移植瘤模型的疗效(以瘤体积计算)
Figure PCTCN2021099561-appb-000170
注:“-”表示不存在,P<0.05表示与溶媒组相比有显著差异;BID表示为每天两次;QD表示为每天一次;mpk表示为mg/kg。
5、试验结论:
以上数据显示,口服连续给药21天后,本发明的实施例12化合物抑瘤效果显著,药效优于以相同剂量或更高剂量给药的阳性化合物TNO155的效果。
试验例11:激酶选择性测试
1、试验目的:通过对本发明化合物进行多种激酶的选择性测试,用于评估本发明化合物的安全性风险。
2、测试单位:ThermoFisher USA
3、测试方法:LanthaScreen binding、Z′-LYTE、Adapta
4、测试靶点:共计100个靶点
Figure PCTCN2021099561-appb-000171
5、试验结论:
Figure PCTCN2021099561-appb-000172
结果显示:TNO-155对以上14个激酶靶点有较强的抑制作用,对剩余86个靶点的抑制作用较弱;实施例12则对100个激酶靶点的抑制作用均较弱,表明实施例12具有较好的激酶选择性,安全性风险低。
总之,本发明提供了一系列具有新颖结构的高活性的SHP2磷酸酶抑制剂,在小鼠KYSE-520 CDX模型及NCI-H358 CDX模型上体现出了良好的药效,有很大的潜力开发成针对肿瘤类疾病的药物。
除本文中描述的那些实施方案外,根据前述描述,本发明的多种修改对本领域技术人员而言会是显而易见的。这样的修改也意图落入所附权利要求书的范围内。本申请中所引用的各参考文献(包括所有专利、专利申请、期刊文章、书籍及任何其它公开)均以其整体援引加入本文。

Claims (22)

  1. 一种具有式I结构的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    Figure PCTCN2021099561-appb-100001
    X选自化学键、S、O、NH和CH 2
    R 1选自氢、羟基、卤素、氨基、C 1-6烷基、3-6元杂环烷基和C 3-6环烷基,其中所述烷基、杂环烷基和环烷基各自任选地被一个或多个选自卤素、羟基和氨基的取代基取代;
    R 2选自氢、羟基、氨基、氰基、卤素、C 1-6烷基、3-6元杂环烷基和C 3-6环烷基,其中所述烷基、杂环烷基和环烷基各自任选地被一个或多个选自卤素、氧代基、羟基、氨基的取代基取代;
    A选自C 6-12亚芳基、5-12元亚杂芳基、3-12元亚杂环烷基和C 3-8亚环烷基,其中所述亚芳基、亚杂芳基、亚杂环烷基和亚环烷基各自任选地被一个或多个R a取代;
    若存在,每一个R a各自独立地选自氢、卤素、羟基、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、氰基、C 1-6烷基、C 3-6环烷基、C 2-6烯基、-S(=O) g-(C 1-6烷基)、-S(=O) gNH 2、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、3-12元杂环烷基、C 6-10芳基和5-12元杂芳基,其中所述烷基、环烷基、烯基、杂环烷基、芳基和杂芳基各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、-S(=O) g-(C 1-6烷基)和C 1-6烷基的取代基取代;
    或者,当X为NH或CH 2时,任意一个R a和X以及与其连接的原子一起形成5-10元脂环、5-10元杂脂环、5-6元杂芳环或苯环,其中所述脂环、杂脂环、杂芳环和苯环各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、羟基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代;
    B选自-N(R b)-C(=O)-G-R d、-N(R b)-S(=O) g-G-R d、-N(R b)-C(=O)-N(R b)-G-R d、-N(R b)-S(=O) g-N(R b)-G-R d、-C(=O)-N(R b)-G-R d、-S(=O) g-N(R b)-G-R d、-S(=O) g-G-R d、-O-G-(3-10元杂环烷基)、-O-G-(C 3-6环烷基)、-O-(C 3-6亚环烷基)-G-H、-O-(3-10元亚杂环烷基)-G-H、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H、-G-O-C(=O)-R b、-G-N(R b)-C(=O)-R b、-G-N(R b)-C(=O)-OR b
    Figure PCTCN2021099561-appb-100002
    其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1-6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);
    G选自C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、C 3-6亚环烷基、C 3-6亚环烯基、3-10元亚杂环烯基和3-10元亚杂环烷基,其中所述亚烷基、亚环烷基、亚环烯基、亚杂环烯基和亚杂环烷基各自任选地被一个或多个选自氢、卤素、氧代基、羟基、氰基、氨基、-OR b、-NHR b、-N(R b) 2、-N(R b)-C(=O)-R b、-C(=O)-NH 2、-C(=O)-N(R b) 2、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 3-6环烷基、C 3-6卤代环烷基、C 3-6羟环烷基、3-10元卤代杂环烷基、3-10元羟杂环烷基和3-10元杂环烷基的取代基取代;
    R b选自氢、卤素、氨基、氰基、C 1-6烷基、C 3-6环烷基、C 3-6环烯基、3-10元杂环烯基和3-10元杂环烷基,其中所述烷基、环烷基、环烯基、杂环烯基和杂环烷基各自任选地被一个或多个选自氢、卤素、氧代基、羟基、氰基和氨基的取代基取代;
    C选自C 6-12亚芳基、5-12元亚杂芳基、3-12元亚杂环烷基、C 3-12亚环烯基、3-12元亚杂环烯基和C 3- 8亚环烷基,其中所述亚芳基、亚杂芳基、亚杂环烷基、亚环烯基、亚杂环烯基和亚环烷基各自任选地被一个或多个R c取代;
    若存在,每一个R c各自独立地选自氢、卤素、羟基、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、氰基、C 1-6烷基、C 2-6烯基、-S(=O) g-(C 1-6烷基)、-S(=O) gNH 2、氨基、-NH(C 1-6烷基)和-N(C 1-6烷基) 2,其中所述烷基和烯基各自任选地被一个或多个选自氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、卤素、氰基、氧代基、 羟基、-O-(C 1-6烷基)、-S(=O) g-(C 1-6烷基)和C 1-6烷基的取代基取代;
    Y选自C 1-6亚烷基、C 3-6亚环烷基、3-6元亚杂环烷基、C 3-6亚环烯基、3-6元亚杂环烯基、C 6-10亚芳基、5-12元亚杂芳基、C 2-6亚烯基、-S(=O) g-(C 1-6亚烷基)-、-S(=O) g-N(R b)-、-C(=O)-(C 1-6亚烷基)-、-C(=O)-(C 3-6亚环烷基)-、-C(=O)-(3-6元亚杂环烷基)-和-C(=O)-,其中所述亚烷基、亚杂环烷基、亚环烯基、亚杂环烯基和亚环烷基各自任选地被一个或多个选自卤素、氧代基、羟基和氨基的取代基取代;
    R d选自卤素、氨基、羟基、氰基、-S(=O) g-(C 1-6烷基)、-O-(C 1-6烷基)、-O-(C 3-6环烷基)、-NH-(C 1-6烷基)、-NH(C 3-6环烷基)、-N(C 1-6烷基) 2、-NH-C(=O)-O-(C 1-6烷基)、-N(C 1-6烷基)-C(=O)-O-(C 1-6烷基)、-O-C(=O)-NH 2、-O-C(=O)-NH(C 1-6烷基)和-N(C 1-6烷基)-C(=O)-(C 1-6烷基),其中所述烷基和环烷基各自任选地被一个或多个选自卤素、氧代基、羟基、氨基和-O-(C 1-6烷基)的取代基取代;
    R 3选自卤素、-OR z、羟基、氰基、-C(=O)-OR z、-C(=O)-N(R z) 2、C 1-6烷基、-(C 1-6亚烷基)-R z、-(C 1- 6亚烷基)-OR z、-(C 1-6亚烷基)-OH、-(C 1-6亚烷基)-N(R z) 2、C 3-6环烷基、3-12元杂环烷基、C 3-6环烯基、3-12元杂环烯基、C 2-6烯基、C 2-6炔基、C 6-12芳基、5-12元杂芳基、-S(=O) g-(C 1-6烷基)、氨基和-N(R z) 2,其中所述烷基、亚烷基、环烷基、杂环烷基、环烯基、杂环烯基、烯基、炔基、杂芳基和芳基各自任选地被一个或多个选自卤素、氰基、氧代基、-OR z、羟基、-N(R z) 2、-NHR z、氨基、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基、5-10元杂芳基、-C(=O)-OR z、-C(=O)-N(R z) 2、-C(=O)-NH 2和硝基的取代基取代;
    若存在,每一个R z各自独立地选自氢、氰基、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、卤素、氰基、氧代基、硝基、羟基、-O-(C 1-6烷基)、C 1-6烷基、C 1-6卤代烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
    R 4选自卤素、C 1-6烷基、C 3-6环烷基、-C(=O)-R z、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-10芳基和5-12元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、卤素、氰基、羟基、氧代基、-O-(C 1-6烷基)、C 1-6烷基、C 1-6卤代烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
    或者,R 3和R 4以及与其连接的原子一起形成4-8元脂环或4-8元杂脂环,其中所述脂环和杂脂环各自任选地被一个或多个R 4a取代,或者所述脂环和杂脂环各自任选地与一个或多个C 6-10芳环或5-12元杂芳环稠合,所述芳环和杂芳环各自任选地被一个或多个R 4a取代;
    若存在,每一个R 4a各自独立地选自氢、卤素、氰基、-C(=O)H、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3-8环烷基)、-C(=O)-(3-8元杂环烷基)、-NH-C(=O)-(C 1-6烷基)、-NH-C(=O)-(C 3-8环烷基)、-NH-C(=O)-(3-8元杂环烷基)、氧代基、C 1-6烷基、C 3-6环烷基、C 3-6环烯基、3-12元杂环烯基、3-12元杂环烷基、C 2-6烯基、C 2-6炔基、C 6-12芳基和5-12元杂芳基,其中所述烷基、环烷基、环烯基、杂环烯基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 6-10芳基和5-10元杂芳基的取代基取代;
    若存在,每一个R 5各自独立地选自氢、卤素、氧代基、氰基、羟基、羧基、-C(=O)-NH 2、-C(=O)-O-(C 1-6烷基)、C 1-6烷基、C 3-6环烷基、C 2-6烯基、C 2-6炔基、-S(=O) g-(C 1-6烷基)、3-6元杂环烷基、C 6-10芳基和5-10元杂芳基,其中所述烷基、环烷基、杂环烷基、烯基、炔基、芳基和杂芳基各自任选地被一个或多个选自氨基、羟基、氧代基、卤素、氰基、-O-(C 1-6烷基)、-NH(C 1-6烷基)和-N(C 1-6烷基) 2的取代基取代;或者任意两个R 5以及与其连接的原子一起形成C 3-10脂环或4-12元杂脂环;
    g为0、1或2;
    n为0、1、2、3、4或5。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    X选自化学键和S,优选S。
  3. 根据权利要求1或2所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    R 1选自氢、羟基、卤素、氨基、C 1-6烷基和C 3-6环烷基,优选氢、卤素、氨基和C 1-6烷基,更优选氢、氟、氨基和甲基。
  4. 根据权利要求1至3中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    R 2选自氢、羟基、卤素、C 1-6烷基和C 3-6环烷基,其中所述烷基和环烷基各自任选地被一个或多个选自卤素和羟基的取代基取代,优选氢、羟基、氟、羟基甲基和氟甲基,更优选羟基甲基。
  5. 根据权利要求1至4中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    A选自C 6-12亚芳基和5-12元亚杂芳基,其中所述亚芳基和亚杂芳基各自任选地被一个或多个R a取代;优选地,A选自亚苯基、亚吡啶基和亚嘧啶基,其中所述亚苯基、亚吡啶基和亚嘧啶基各自任选地被一个或多个R a取代;
    若存在,每一个R a各自独立地选自氢、氟、氯、溴、羟基、甲氧基、环丙氧基、氰基、甲基、乙基、正丙基、异丙基、二氟甲氧基、三氟甲氧基、三氟甲基、二氟甲基、环丙基、吡咯烷基、吗啉基、氨基、甲氨基、二甲氨基、甲硫基、甲磺酰基和氨磺酰基,优选氢、氟、氯、溴、羟基、氰基、甲基、乙基、正丙基、异丙基、环丙基、吡咯烷基、吗啉基、氨基、甲氨基和二甲氨基,更优选氟、氯、溴、氰基、甲基和氨基。
  6. 根据权利要求1至5中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    B选自
    Figure PCTCN2021099561-appb-100003
    -N(R b)-C(=O)-G-R d、-O-G-(3-10元杂环烷基)、-O-G-R d、-N(R b)-G-(3-10元杂环烷基)、-N(R b)-G-(C 3-6环烷基)、-N(R b)-(C 3-6亚环烷基)-G-H、-N(R b)-(3-10元亚杂环烷基)-G-H和-G-O-C(=O)-R b,其中所述杂环烷基、亚杂环烷基、环烷基和亚环烷基各自任选地被一个或多个选自氢、卤素、羟基、氨基、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、氧代基、-O-(C 1-6烷基)、C 1-6卤代烷基和C 1- 6烷基的取代基取代,且B不为-O-(C 1-2卤代烷基);
    G选自C 1-6亚烷基、C 3-6亚环烷基和3-6元亚杂环烷基,所述亚烷基、亚环烷基和亚杂环烷基各自任选地被一个或多个选自氢和氟的取代基取代;优选地,G为C 1-3亚烷基,所述亚烷基任选地被一个或多个选自氢和氟的取代基取代;
    R b选自氢、氟、氯、氨基、C 1-3烷基、环丙基和3-6元杂环烷基,其中所述烷基和杂环烷基各自任选地被一个或多个选自氢、氟、氧代基、羟基和氰基的取代基取代;
    C选自3-6元亚杂环烷基和C 3-6亚环烷基,其中所述亚杂环烷基和亚环烷基各自任选地被一个或多个R c取代;优选地,C选自亚吡咯烷基、亚氮杂环丁烷基、亚哌啶基和亚哌嗪基,其中所述亚吡咯烷基、亚氮杂环丁烷基、亚哌啶基和亚哌嗪基各自任选地被一个或多个R c取代;
    若存在,每一个R c各自独立地选自氢、氟、氰基、甲基、乙基、氟甲基、羟基甲基和羟基;
    Y选自C 1-3亚烷基、C 3-6亚环烷基、3-6元亚杂环烷基、C 2-6亚烯基、-C(=O)-(C 1-3亚烷基)-、-C(=O)-(C 3-6亚环烷基)-、-C(=O)-(3-6元亚杂环烷基)-和-C(=O)-,其中所述亚烷基、亚杂环烷基和亚环烷基各自任选地被一个或多个选自氟、羟基和氨基的取代基取代;优选地,Y选自-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、亚环丙基和-C(=O)-CH 2-,其中所述-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、亚环丙基和-C(=O)-CH 2-各自任选地被一个或多个选自氟、羟基和氨基的取代基取代;
    R d选自氟、氨基、羟基、氰基、-S(=O) 2-(C 1-6烷基)、-O-(C 1-6烷基)、-NH-C(=O)-O-(C 1-6烷基)、-O-C(=O)-NH 2、-O-C(=O)-NH(C 1-6烷基)和-N(C 1-6烷基)-C(=O)-(C 1-6烷基),其中所述烷基任选地被一个或多个选自氟、氯、氧代基、羟基和-O-(C 1-6烷基)的取代基取代;优选地,R d选自氟、氨基、羟基、氰基、-S(=O) 2-CH 3、-O-CH 3、-NH-C(=O)-O-CH 3和-O-C(=O)-NH 2,其中所述-S(=O) 2-CH 3、-O-CH 3和 -NH-C(=O)-O-CH 3各自任选地被一个或多个选自氟、氯、氧代基、羟基和-O-(C 1-6烷基)的取代基取代。
  7. 根据权利要求1至6中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    B选自
    Figure PCTCN2021099561-appb-100004
    Figure PCTCN2021099561-appb-100005
    Figure PCTCN2021099561-appb-100006
    -NH-C(=O)-CH 2-CN、-N(CH 3)-C(=O)-C(CH 3) 2-OH、
    Figure PCTCN2021099561-appb-100007
  8. 根据权利要求1至7任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    R 3和R 4不相互连接时,
    R 3选自氟、氯、羟基、氰基、甲基、乙基、环丙基、氧杂环丁烷基、吡啶基、嘧啶基、噁唑基、噻唑基、哒嗪基、吡唑基和噻吩基,优选氟、氯、甲基、乙基、环丙基和氧杂环丁烷基,更优选氟和甲基;
    R 4选自氟、氯、甲基、乙基、异丙基、环丙基、甲氧甲基、羟基甲基、氟甲基、二氟甲基、三氟甲基、氟乙基、氰甲基、羟基甲基、2-甲基呋喃基、噻唑基、吡啶基和嘧啶基,优选甲基、氟甲基、二氟甲基、三氟甲基、羟基甲基、甲氧甲基、氟乙基、氰甲基和羟基甲基,更优选甲基、氟甲基、氟乙基、甲氧甲基、氰甲基和羟基甲基。
  9. 根据权利要求1至7任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    R 3和R 4以及与其连接的原子成环时,
    Figure PCTCN2021099561-appb-100008
    选自以下任意一种:
    Figure PCTCN2021099561-appb-100009
    Figure PCTCN2021099561-appb-100010
    其中m为0、1或2;
    优选地,
    Figure PCTCN2021099561-appb-100011
    选自以下任意一种:
    Figure PCTCN2021099561-appb-100012
    Figure PCTCN2021099561-appb-100013
    其中m为0、1或2;
    更优选地,
    Figure PCTCN2021099561-appb-100014
    选自以下任意一种:
    Figure PCTCN2021099561-appb-100015
    若存在,每一个R 4a各自独立地选自氢、氟、氯、氰基、-C(=O)H、-C(=O)-(C 1-6烷基)、-C(=O)-(C 3- 8环烷基)、-C(=O)-(3-8元杂环烷基)、氧代基、C 1-6烷基、C 3-6环烷基、3-12元杂环烷基、C 2-6烯基和C 2-6炔基,其中所述烷基、环烷基、杂环烷基、烯基和炔基各自任选地被一个或多个选自卤素、氰基、羟基、-O-(C 1-6烷基)和C 1-6烷基的取代基取代;优选地,每一个R 4a各自独立地选自氢、氟、氯、氧代基、甲基、乙酰基和环丙基。
  10. 根据权利要求1至9任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    同时包含多个R 5,并且任意两个R 5不互相连接,每一个R 5各自独立地选自氢、氟、氯、氧代基、氰基、-C(=O)-O-(C 1-4烷基)和C 1-3烷基,其中所述烷基各自任选地被一个或多个选自氨基、羟基、氧代基和卤素的取代基取代;优选地,每一个R 5各自独立地选自氢、氟、氯、氰基、甲基和-C(=O)-O-CH 3,优选氢、氟和甲基,更优选氢。
  11. 根据权利要求1至9任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    同时包含多个R 5,任意两个不连接同一个原子的R 5以及与其连接的原子一起形成C 4-8脂环或4-8元杂脂环,优选C 4-6脂环或4-6元含氮杂脂环,或者任意两个连接同一个原子的R 5以及与其连接的原子一起形成C 3-6脂环或3-6元杂脂环,优选环丙烷环。
  12. 根据权利要求1至11任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,所述化合物具有如式IIa-IIb、IIIa-IIIb、IVa-IVd、Va-Vc中任一种所示的结构,
    Figure PCTCN2021099561-appb-100016
    A、B、R 3和R 4如权利要求1至11任一项所定义。
  13. 根据权利要求1至12任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    A选自5-12元亚杂芳基,其中所述亚杂芳基任选地被一个或多个卤素取代;
    B选自
    Figure PCTCN2021099561-appb-100017
    C选自3-6元亚杂环烷基,所述亚杂环烷基任选地被一个或多个氟、氰基、甲基或氟甲基取代;
    Y选自C 1-3亚烷基和-C(=O)-(C 1-3亚烷基)-;
    R d选自氟、氨基、羟基、氰基、-S(=O) 2-(C 1-6烷基)和-O-(C 1-6烷基);
    R 3和R 4不相互连接时,R 3选自C 1-6烷基,R 4选自C 1-6烷基,所述烷基任选地被-O-(C 1-3烷基)取代;
    或者,R 3和R 4以及与其连接的原子成环时,
    Figure PCTCN2021099561-appb-100018
    选自
    Figure PCTCN2021099561-appb-100019
    Figure PCTCN2021099561-appb-100020
    m为0、1或2;
    若存在,每一个R 4a各自独立地选自氢和氟。
  14. 根据权利要求1至13任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    A选自亚吡啶基,所述亚吡啶基任选地被一个或多个氯取代;
    B选自
    Figure PCTCN2021099561-appb-100021
    C选自亚氮杂环丁烷基和亚吡咯烷基,所述亚氮杂环丁烷基和亚吡咯烷基各自任选地被一个或多个氟、氰基、甲基或氟甲基取代;
    Y选自-CH 2-、-CH(CH 3)-、-C(CH 3) 2-和-C(=O)-CH 2-;
    R d选自氟、氨基、羟基、氰基、S(=O) 2-CH 3和-O-CH 3
    R 3和R 4不相互连接时,R 3选自甲基,R 4选自甲基,所述甲基任选地被甲氧基取代;
    或者,R 3和R 4以及与其连接的原子成环时,
    Figure PCTCN2021099561-appb-100022
    选自
    Figure PCTCN2021099561-appb-100023
    Figure PCTCN2021099561-appb-100024
  15. 根据权利要求1至14任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,其中
    A选自亚吡啶基,所述亚吡啶基任选地被一个或多个氯取代;
    B选自
    Figure PCTCN2021099561-appb-100025
    Figure PCTCN2021099561-appb-100026
    Figure PCTCN2021099561-appb-100027
    R 3和R 4不相互连接时,R 3选自甲基,R 4选自甲基,所述甲基任选地被甲氧基取代;
    或者,R 3和R 4以及与其连接的原子成环时,
    Figure PCTCN2021099561-appb-100028
    选自
    Figure PCTCN2021099561-appb-100029
    Figure PCTCN2021099561-appb-100030
  16. 下列化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药:
    Figure PCTCN2021099561-appb-100031
    Figure PCTCN2021099561-appb-100032
    Figure PCTCN2021099561-appb-100033
    Figure PCTCN2021099561-appb-100034
    Figure PCTCN2021099561-appb-100035
    Figure PCTCN2021099561-appb-100036
    Figure PCTCN2021099561-appb-100037
    Figure PCTCN2021099561-appb-100038
    Figure PCTCN2021099561-appb-100039
  17. 一种药物组合物,其包含权利要求1至16中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药,以及一种或 多种药学上可接受的载体。
  18. 一种药品,其包含:
    a)容器;
    b)位于所述容器中的至少一种根据权利要求1至16中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药或者根据权利要求17所述的药物组合物;和
    c)任选存在的包装和/或说明书。
  19. 根据权利要求1至16中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药或者根据权利要求17所述的药物组合物或者根据权利要求18所述的药品在制备用于预防和/或治疗至少部分由SHP2介导的疾病或病症优选癌症的药物中的用途。
  20. 根据权利要求1至16中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药或者根据权利要求17所述的药物组合物或者根据权利要求18所述的药品,其用作SHP2抑制剂或者用于预防和/或治疗至少部分由SHP2介导的疾病或病症优选癌症。
  21. 一种用于预防和/或治疗至少部分由SHP2介导的疾病或病症优选癌症的方法,其包括下列步骤:将预防和/或治疗有效量的根据权利要求1至16中任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂化物、同位素标记物、代谢物或前药或者根据权利要求17所述的药物组合物或者根据权利要求18所述的药品施用于对其有需求的个体。
  22. 一种用于预防和/或治疗至少部分由SHP2介导的疾病或病症优选癌症的组合方法,其包括:
    a)根据权利要求21所述的方法;和
    b)额外的方法。
PCT/CN2021/099561 2020-06-22 2021-06-11 取代吡嗪类化合物,包含其的药物组合物及其用途 WO2021259077A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2022571308A JP2023530838A (ja) 2020-06-22 2021-06-11 置換されたピラジン化合物、当該化合物を含む医薬組成物、及びその使用
EP21829137.5A EP4169913A1 (en) 2020-06-22 2021-06-11 Substituted pyrazine compound, pharmaceutical composition comprising same, and use thereof
US17/926,763 US20230212180A1 (en) 2020-06-22 2021-06-11 Substituted pyrazine compound, pharmaceutical composition comprising same, and use thereof
CN202180036771.4A CN115667239A (zh) 2020-06-22 2021-06-11 取代吡嗪类化合物,包含其的药物组合物及其用途

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202010576572.X 2020-06-22
CN202010576572 2020-06-22
CN202011359022 2020-11-27
CN202011359022.9 2020-11-27

Publications (1)

Publication Number Publication Date
WO2021259077A1 true WO2021259077A1 (zh) 2021-12-30

Family

ID=79281930

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/099561 WO2021259077A1 (zh) 2020-06-22 2021-06-11 取代吡嗪类化合物,包含其的药物组合物及其用途

Country Status (5)

Country Link
US (1) US20230212180A1 (zh)
EP (1) EP4169913A1 (zh)
JP (1) JP2023530838A (zh)
CN (1) CN115667239A (zh)
WO (1) WO2021259077A1 (zh)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022237367A1 (zh) * 2021-05-13 2022-11-17 中国科学院上海药物研究所 抑制shp2活性的杂环化合物、其制备方法及用途
CN115521305A (zh) * 2022-09-20 2022-12-27 中国药科大学 Shp2&nampt双靶向化合物及其药物组合物和用途
WO2023071314A1 (zh) * 2021-10-29 2023-05-04 中国药科大学 Shp2与cdk4/6双靶点抑制化合物合成及其制备方法与应用
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015107495A1 (en) 2014-01-17 2015-07-23 Novartis Ag N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
CN105899493A (zh) * 2014-01-17 2016-08-24 诺华股份有限公司 用于抑制shp2活性的1-(三嗪-3-基/哒嗪-3-基)-哌(-嗪)啶衍生物及其组合物
WO2018013597A1 (en) 2016-07-12 2018-01-18 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric shp2 inhibitors
CN107922388A (zh) * 2015-06-19 2018-04-17 诺华股份有限公司 用于抑制shp2活性的化合物和组合物
CN109415360A (zh) * 2016-06-14 2019-03-01 诺华股份有限公司 用于抑制shp2活性的化合物和组合物
WO2019075265A1 (en) 2017-10-12 2019-04-18 Revolution Medicines, Inc. PYRIDINE, PYRAZINE AND TRIAZINE COMPOUNDS AS ALLOSTERIC INHIBITORS OF SHP2
WO2019182960A1 (en) * 2018-03-21 2019-09-26 Synblia Therapeutics, Inc. Shp2 inhibitors and uses thereof
WO2020076723A1 (en) * 2018-10-08 2020-04-16 Revolution Medicines, Inc. Shp2 inhibitor compositions for use in treating cancer
WO2020073949A1 (zh) * 2018-10-10 2020-04-16 江苏豪森药业集团有限公司 含氮杂芳类衍生物调节剂、其制备方法和应用

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015107495A1 (en) 2014-01-17 2015-07-23 Novartis Ag N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
CN105899493A (zh) * 2014-01-17 2016-08-24 诺华股份有限公司 用于抑制shp2活性的1-(三嗪-3-基/哒嗪-3-基)-哌(-嗪)啶衍生物及其组合物
CN107922388A (zh) * 2015-06-19 2018-04-17 诺华股份有限公司 用于抑制shp2活性的化合物和组合物
CN109415360A (zh) * 2016-06-14 2019-03-01 诺华股份有限公司 用于抑制shp2活性的化合物和组合物
WO2018013597A1 (en) 2016-07-12 2018-01-18 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric shp2 inhibitors
WO2019075265A1 (en) 2017-10-12 2019-04-18 Revolution Medicines, Inc. PYRIDINE, PYRAZINE AND TRIAZINE COMPOUNDS AS ALLOSTERIC INHIBITORS OF SHP2
CN111212834A (zh) * 2017-10-12 2020-05-29 锐新医药公司 作为变构shp2抑制剂的吡啶、吡嗪和三嗪化合物
WO2019182960A1 (en) * 2018-03-21 2019-09-26 Synblia Therapeutics, Inc. Shp2 inhibitors and uses thereof
WO2020076723A1 (en) * 2018-10-08 2020-04-16 Revolution Medicines, Inc. Shp2 inhibitor compositions for use in treating cancer
WO2020073949A1 (zh) * 2018-10-10 2020-04-16 江苏豪森药业集团有限公司 含氮杂芳类衍生物调节剂、其制备方法和应用

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
GOODMANGILMAN'S: "The Pharmacological Basis of Therapeutics [M", 1996, MCGRAW-HILL
JUSIAK, SOCZEWINSKI ET AL.: "Remington's Pharmaceutical Sciences [M", 2005, MACK PUBLISHING COMPANY
STAHL, WERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use [M", 2002, WILEY-VCH
T. HIGUCHIV. STELLA: "Prodrugs as Novel Drug Delivery Systems [J", vol. 14, 1975, AMERICAN CHEMICAL SOCIETY
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis [M", 2006, JOHN WILEY & SONS

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022237367A1 (zh) * 2021-05-13 2022-11-17 中国科学院上海药物研究所 抑制shp2活性的杂环化合物、其制备方法及用途
WO2023071314A1 (zh) * 2021-10-29 2023-05-04 中国药科大学 Shp2与cdk4/6双靶点抑制化合物合成及其制备方法与应用
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
CN115521305A (zh) * 2022-09-20 2022-12-27 中国药科大学 Shp2&nampt双靶向化合物及其药物组合物和用途

Also Published As

Publication number Publication date
CN115667239A (zh) 2023-01-31
JP2023530838A (ja) 2023-07-20
US20230212180A1 (en) 2023-07-06
EP4169913A1 (en) 2023-04-26

Similar Documents

Publication Publication Date Title
WO2021259077A1 (zh) 取代吡嗪类化合物,包含其的药物组合物及其用途
WO2021043077A1 (zh) 一种取代吡嗪化合物、其制备方法和用途
CN115448923B (zh) 嘧啶并环化合物及其制备方法和应用
CA2787360C (en) Pyrrol0[3,2-c]pyridinyl-4-benzamide compounds and their use as apoptosis signal-regulating kinase 1 inhibitors
WO2019158019A1 (zh) 嘧啶并环化合物及其制备方法和应用
CA3054455C (en) Fgfr inhibitor and application thereof
TW201716415A (zh) 作為蛋白質激酶之調節劑的掌性二芳基巨環
WO2019037640A1 (en) HETEROCYCLIC COMPOUNDS AS KINASE INHIBITORS, COMPOSITIONS COMPRISING THE HETEROCYCLIC COMPOUND, AND METHODS OF USING THE SAME
TW201406761A (zh) 做爲jak抑制劑之哌啶基環丁基取代之吡咯并吡啶及吡咯并嘧啶衍生物
EP3035800A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
KR20220130168A (ko) 피리미딘-4(3h)-케톤 헤테로시클릭 화합물, 그의 제조 방법, 및 의약 및 약리학에서의 그의 용도
JP2013500972A (ja) Sykキナーゼ阻害剤としての2,7−ナフチリジン−1−オン誘導体
US11161854B2 (en) Indazolyl-spiro[2.2]pentane-carbonitrile derivatives as LRRK2 inhibitors, pharmaceutical compositions, and uses thereof
WO2021139775A1 (zh) 吡啶酮化合物及应用
CN113330009B (zh) 氮杂环化合物、其制备方法及用途
AU2013209586A1 (en) Substituted pyrimidine compounds and their use as SYK inhibitors
WO2021132422A1 (ja) ピリダジニルチアアゾールカルボキシアミド化合物
CN113677680A (zh) Egfr抑制剂及其组合物和应用
WO2023088408A1 (zh) 选择性parp1抑制剂及其应用
CN115515949A (zh) 新型氨基嘧啶类egfr抑制剂
CN113939294A (zh) 化合物、组合物和方法
CN113968856B (zh) 一类具有激酶抑制活性的化合物
WO2021197467A1 (zh) 多靶点的抗肿瘤化合物及其制备方法和应用
WO2020207419A1 (zh) 哌嗪酰胺衍生物,其制备方法及其在医药上的用途
JP7010443B2 (ja) インドリジン系化合物、その製造方法及び用途

Legal Events

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

Ref document number: 21829137

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022571308

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021829137

Country of ref document: EP

Effective date: 20230123