WO2024055879A1 - 一类双并环化合物、其制备方法及用途 - Google Patents

一类双并环化合物、其制备方法及用途 Download PDF

Info

Publication number
WO2024055879A1
WO2024055879A1 PCT/CN2023/117167 CN2023117167W WO2024055879A1 WO 2024055879 A1 WO2024055879 A1 WO 2024055879A1 CN 2023117167 W CN2023117167 W CN 2023117167W WO 2024055879 A1 WO2024055879 A1 WO 2024055879A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
hydrogen
compound
amino
cycloalkyl
Prior art date
Application number
PCT/CN2023/117167
Other languages
English (en)
French (fr)
Inventor
刘金明
任云
何婷
李晓勇
何诰
刘叶
田强
宋宏梅
葛均友
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
Publication of WO2024055879A1 publication Critical patent/WO2024055879A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • 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
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • This application is based on the application with CN application number 202211126580.
  • This application belongs to the field of medicinal chemistry, and specifically relates to a class of dicyclic compounds, their preparation methods and uses.
  • TLRs Toll-like receptors
  • PRR Pattern recognition receptor
  • PAMPs pathogen-associated molecular patterns
  • TLR1/2/4/5/6/10 are expressed on the cell surface and can quickly recognize the products of bacterial metabolism.
  • TLR3/7/8/9 is expressed in cellular endosomes and mainly recognizes and monitors exogenous nucleic acid substances from pathogenic cells.
  • TLR7 has a limited expression distribution in humans and is mainly expressed by B cells and plasmacytoid dendritic cells (pDC) in response to viral infection, inducing high levels of interferon- ⁇ and enhancing pDC costimulation and Antigen presentation ability promotes the proliferation of CD4+ T cells and further activates CD8+ T cells to generate adaptive T cell responses.
  • TLR8 expression is relatively more widespread and is mainly expressed by monocytes, NK cells and myeloid dendritic cells (mDC) in humans. Stimulating TLR8 can induce the release of various pro-inflammatory cytokines, such as tumor necrosis factor- ⁇ (TNF- ⁇ ), IL-6, IL-12, and interferon- ⁇ .
  • TNF- ⁇ tumor necrosis factor- ⁇
  • IL-6 interferon- ⁇
  • interferon- ⁇ interferon- ⁇
  • TLR7 and TLR8 play an important role in both the body's innate immunity and acquired immunity, and are popular targets for anti-cancer and anti-viral immunotherapy.
  • TLR7 and TLR8 agonists There have been reports from different companies involving multiple TLR7 and/or TLR8 agonists, but there is still a great demand for TLR7 and/or TLR8 agonists with better activity, safety and physical and chemical properties.
  • Immunostimulatory antibody conjugates combine the precise tumor localization of antibodies with the long-lasting anti-tumor activity and immune memory effects of immune modulators such as TLR7 and/or TLR8 agonists, enabling systemic administration while safely inducing anti-tumor immune responses.
  • the present application provides a class of dicyclic compounds with TLR7 and/or TLR8 agonistic activity.
  • This type of compound has It has better activity, safety and physical and chemical properties, and this type of compound can be further used in immunostimulatory antibody conjugates, which is of great significance for the development of tumor and virus drugs.
  • the present application provides a class of bicyclic compounds, which have a strong agonistic effect on TLR7 and/or TLR8, and therefore have better prevention and/or treatment of related diseases mediated by TLR7 and/or TLR8. use.
  • the compounds were also found to have good physicochemical properties (eg solubility, physical and/or chemical stability) and good safety.
  • the compound is a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, or isotopically labeled compound thereof , metabolites or prodrugs:
  • L 1 is selected from O, S and NR 5 ;
  • L 2 is a covalent bond, or selected from C 1-6 alkylene and -C 1-6 alkylene-O-;
  • R 1 is C 1-6 alkyl, which is optionally substituted by one or more elements selected from hydrogen, halogen, hydroxyl, C 1-6 alkyl, C 1-6 alkoxy, 5-10 membered hetero Aryl and R 6 -C(O)-NH- groups substituted;
  • R 2 is selected from hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl Alkyl, heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1 -6 alkoxy, R 7 and R 7 -L 3 - groups substituted;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 3 is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 alkoxy;
  • R 4 is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 alkoxy;
  • R 5 is selected from hydrogen and C 1-6 alkyl
  • R 6 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl , heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 Substituted with alkoxy groups;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • the application provides a drug-linker described in the following formula, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxidation thereof substances, isotopically labeled compounds, metabolites or prodrugs: D-L'
  • D is a fragment of the compound mentioned above;
  • L’ is the linker fragment.
  • This application also provides immunostimulatory antibody conjugates represented by formula (ISAC-I) or pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates thereof, N-oxides, isotopically labeled compounds, metabolites or prodrugs:
  • Ab is an antibody that targets the target antigen
  • L is the fragment obtained after connecting L' in the drug-linker mentioned above to Ab;
  • z is selected from 1-10.
  • the present application provides a composition
  • a composition comprising the immunostimulatory antibody conjugate or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvent thereof compounds, N-oxides, isotopically labeled compounds, metabolites or prodrugs.
  • the present application provides the use of the compound represented by formula (I) in the preparation of immunostimulatory antibody conjugates.
  • the present application provides a pharmaceutical composition, which contains a preventive and/or therapeutically effective amount of the compound described in any one of the first aspects or its pharmaceutically acceptable salts, esters, stereoisomers, Tautomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs, immunostimulatory antibody conjugates represented by formula (ISAC-I), or the composition and one or more pharmaceutically acceptable carriers.
  • a pharmaceutical composition which contains a preventive and/or therapeutically effective amount of the compound described in any one of the first aspects or its pharmaceutically acceptable salts, esters, stereoisomers, Tautomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs, immunostimulatory antibody conjugates represented by formula (ISAC-I), or the composition and one or more pharmaceutically acceptable carriers.
  • the present application provides a pharmaceutical kit, which contains the compound described in any one of the first aspects or its pharmaceutically acceptable salts, esters, stereoisomers, tautomers, and polymorphs.
  • the present application provides the compound described in any one of the first aspects or its pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, N - Oxides, isotope-labeled compounds, metabolites or prodrugs, immunostimulatory antibody conjugates represented by formula (ISAC-I), the compositions or pharmaceutical compositions are prepared for the prevention and/or treatment of TLR7 and/or Or use in drugs for TLR8-mediated related diseases.
  • the present application provides the compound described in any one of the first aspects or its pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, N - Oxide, isotope-labeled compound, metabolite or prodrug, immunostimulatory antibody conjugate represented by formula (ISAC-I), the composition or pharmaceutical composition, which is used for preventing and/or treating TLR7 and/or or TLR8-mediated related diseases.
  • the present application provides methods for preparing compounds represented by formula (I).
  • alkyl is defined as a straight or branched chain saturated aliphatic hydrocarbon radical.
  • C 1-6 alkyl refers to a straight or branched chain group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl), which is optionally substituted by one or more (such as 1 to 3) suitable substituents such as halogen.
  • C 1-3 alkyl refers to a straight or branched chain group having 1 to 3 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl) optionally replaced by one or Multiple, such as 1 to 3, suitable substituents such as halogen substitution.
  • alkoxy refers to "alkyl-O-" as defined above.
  • C 1-6 alkoxy refers to "C 1-6 alkyl-O-” as defined above.
  • C 1-3 alkoxy group described herein refers to "C 1-3 alkyl-O-", and the "C 1-3 alkyl group” is as defined above.
  • Exemplary C 1-6 alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, n- Pentyloxy or n-hexyloxy.
  • C 1-6 alkylene refers to a divalent group formed by removing two hydrogens from a linear or branched chain alkane containing 1 to 6 carbon atoms, including “C 1- 5 alkylene”, “C 1-4 alkylene”, “C 1-3 alkylene", “C 1-2 alkylene”, specific examples include but are not limited to: -CH 2 -, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 - Wait.
  • cycloalkyl refers to a saturated or partially unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., a monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl , cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, or bicyclo, including spiro, fused or bridged systems, such as bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, bicyclo [3.2.1]octyl or bicyclo[5.2.0]nonyl, decalinyl, etc.), which is optionally substituted by one or more (such as 1 to 3) suitable substituents.
  • a monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl , cyclohexyl, cycloh
  • C 3-6 cycloalkyl refers to a saturated or partially unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring having 3 to 6 ring-forming carbon atoms (e.g., cyclopropyl, cyclopropyl butyl, cyclopentyl or cyclohexyl), optionally substituted by one or more (such as 1 to 3) suitable substituents, for example methyl-substituted cyclopropyl.
  • halogen group is defined to include fluorine, chlorine, bromine or iodine.
  • halogenated means substituted by one or more (such as 1 to 3) the same or different halogen atoms.
  • haloalkyl refers to an alkyl group substituted by one or more (such as 1 to 3) the same or different halogen atoms.
  • C 1-6 haloalkyl refers to a haloalkyl group having 1 to 6 carbon atoms, such as -CF 3 , -C 2 F 5 , -CHF 2 , -CH 2 F, -CH 2 CF 3 , - CH 2 Cl or -CH 2 CH 2 CF 3 , etc.
  • heterocycle or “heterocyclyl” refers to a saturated or partially unsaturated non-aromatic monocyclic or polycyclic group, for example, which has 2, 3, 4, 5, 6 in the ring , 7, 8 or 9 carbon atoms and one or more (e.g.
  • N, O or S(O) t independently selected from N, O or S(O) t (where t is 0, 1 or 2 ) heteroatoms, such as 3-12-membered heterocyclyl, 3-7-membered heterocyclyl, 3-6-membered heterocyclyl, 5-6-membered heterocyclyl, etc., such as 5-10-membered nitrogen-containing heterocyclyl, 6-10-membered oxygen-containing heterocyclic group, 6-8-membered sulfur-containing heterocyclic group, 5-8-membered oxygen-containing heterocyclic group, etc.
  • t is 0, 1 or 2
  • heteroatoms such as 3-12-membered heterocyclyl, 3-7-membered heterocyclyl, 3-6-membered heterocyclyl, 5-6-membered heterocyclyl, etc., such as 5-10-membered nitrogen-containing heterocyclyl, 6-10-membered oxygen-containing heterocyclic group, 6-8-membered sulfur-containing heterocyclic group,
  • heterocyclyl include, but are not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, tetrahydropyrrolidinyl, hexahydropyrrolidyl, Hydrogen-1H-pyrroline, pyrrolidonyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, tetrahydropyridyl, piperidyl, morpholinyl, dithianyl, thiomorpholinyl , piperazinyl, etc.
  • aryl refers to an all-carbon monocyclic or fused-ring polycyclic aromatic group having a conjugated pi electron system.
  • C 6-10 aryl or “C 6-10 aromatic ring” refers to an aromatic group containing 6 to 10 carbon atoms, such as phenyl (ring) or naphthyl (ring).
  • the aryl group or aromatic ring is optionally substituted with 1 or more (such as 1 to 3) suitable substituents (eg, halogen, -OH, -CN, -NO2 , C1-6 alkyl, etc.).
  • heteroaryl refers to an aromatic cyclic group in which at least one ring atom is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom.
  • ring atoms eg carbon atoms, nitrogen atoms or sulfur atoms
  • cyclic structure can be oxo-substituted.
  • the hydrogens in the groups referred to herein may be replaced by deuterium.
  • substituted means that one or more (e.g., 1, 2, 3, or 4) hydrogens on the designated atom are replaced by a selection from the indicated group, provided that no more than the number of atoms designated on the designated atom is substituted. Normal valence in the present case and the substitution forms a stable compound. Combinations of substituents and/or variables are permissible only if such combinations form stable compounds.
  • substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as optionally substituted with one or more of the substituent lists, then one or more hydrogens on the carbon (to the extent of any hydrogen present) may be independently and/or together Selected substituents are substituted or unsubstituted. If you take The nitrogen of a substituent is described as being optionally substituted with one or more of the substituents listed, then one or more hydrogens on the nitrogen (to the extent of any hydrogen present) may each be replaced by an independently selected substituent. or not substituted.
  • each substituent is selected independently of the other.
  • each substituent may be the same as or different from another (other) substituent.
  • one or more means 1 or more than 1 under reasonable conditions, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the point of attachment of a substituent may be from any suitable position on the substituent.
  • the present application also provides all pharmaceutically acceptable isotopically labeled compounds that are identical to the compounds described except that one or more atoms are substituted with the same atomic number but an atomic mass or mass number that is different from the atomic mass predominant in nature or Atomic substitution of mass number.
  • suitable isotopes include, but are not limited to, isotopes of hydrogen (e.g. 2 H, 3 H, deuterium D, tritium T); isotopes of carbon (e.g. 11 C, 13 C and 14 C); isotopes of chlorine (e.g.
  • Isotopes of fluorine such as 18 F
  • Isotopes of iodine such as 123 I and 125 I
  • Isotopes of nitrogen such as 13 N and 15 N
  • Isotopes of oxygen such as 15 O, 17 O and 18 O
  • Phosphorus Isotopes of sulfur e.g. 32 P
  • sulfur isotopes e.g. 35 S
  • Certain isotopically labeled compounds eg, those incorporating radioactive isotopes
  • the radioactive isotopes tritium (i.e. 3 H) and carbon-14 (i.e.
  • positron emitting isotopes eg 11 C, 18 F, 15 O and 13 N
  • PTT positron emission tomography
  • Isotopically labeled compounds may be prepared by methods similar to those described in the accompanying Schemes and/or Examples and Preparations by using appropriate isotopically labeled reagents in place of the previously employed non-labeled reagents.
  • Pharmaceutically acceptable solvates as described herein include those in which the crystallization solvent may be isotopically substituted, for example, D2O , acetone- d6 , or DMSO- d6 .
  • stereoisomer means an isomer formed due to at least one asymmetric center. In compounds with one or more (eg 1, 2, 3 or 4) asymmetric centers, it can give rise to racemic mixtures, single enantiomers, diastereomeric mixtures and Individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds may exist as mixtures of two or more structurally distinct forms in rapid equilibrium (often referred to as tautomers). Representative examples of tautomers include keto-enol tautomers, phenol-ketone tautomers, nitroso-oxime tautomers, and imine-enamine tautomers. wait.
  • This application also provides all possible crystalline forms or polymorphs of the compounds, which may be single polymorphs or mixtures of more than one polymorph in any proportion.
  • compositions herein may exist in free form for therapeutic use, or, where appropriate, as pharmaceutically acceptable derivatives thereof.
  • pharmaceutically acceptable derivatives include, but are not limited to: pharmaceutically acceptable salts, solvates, metabolites or prodrugs that, upon administration to a patient in need thereof, provide, directly or indirectly, The compound or its metabolites or residues. Therefore, when reference is made herein to "the compound", it is also intended to encompass the various derivative forms of the compound described above.
  • Pharmaceutically acceptable salts of the compounds include acid addition salts and base addition salts thereof. Suitable acid addition salts are formed from acids that form pharmaceutically acceptable salts. Suitable base addition salts are formed from bases that form pharmaceutically acceptable salts.
  • suitable salts see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: Properties, Selection, and Use” (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds are known to those skilled in the art.
  • the compound may exist in the form of a solvate, preferably a hydrate, wherein the compound contains a polar solvent as a structural element of the crystal lattice of the compound.
  • the amount of polar solvent, especially water, may be present in stoichiometric or non-stoichiometric ratios.
  • nitrogen-containing heterocycles are capable of forming N-oxides; those skilled in the art will recognize that nitrogen-containing heterocycles are capable of forming N-oxides. Nitrogen-containing heterocycle. Those skilled in the art will also recognize that tertiary amines are capable of forming N-oxides.
  • N-oxides of heterocyclic and tertiary amines are well known to those skilled in the art and include the use of peroxyacids such as peracetic acid and m-chloroperoxybenzoic acid (MCPBA), hydrogen peroxide, alkyl Hydroperoxides such as tert-butyl hydroperoxide, sodium perborate and dioxirane such as dimethyldioxirane are used to oxidize heterocyclic and tertiary amines.
  • MCPBA m-chloroperoxybenzoic acid
  • alkyl Hydroperoxides such as tert-butyl hydroperoxide
  • sodium perborate and dioxirane such as dimethyldioxirane
  • the application also provides metabolites of the compounds, ie, substances formed in the body upon administration of the compounds. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present application also relates to metabolites of said compounds, including by reacting said compounds with mammals A compound produced by exposure to it for a time sufficient to produce its metabolites.
  • the present application further provides prodrugs of said compounds, which are certain derivatives of said compounds which may themselves have little or no pharmacological activity and which when administered into or onto the body may, for example, be hydrolyzed Cleavage converts the compound with the desired activity.
  • prodrugs will be functional group derivatives of the compound that are readily converted in vivo to the desired therapeutically active compound. Additional information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems,” Volume 14, ACS Symposium Series (T. Higuchi and V. Stella) and "Bioreversible Carriers in Drug Design," Pergamon Press, 1987 ( Edited by E.B. Roche, American Pharmaceutical Association).
  • the prodrug may be replaced, for example, by some moiety known to the person skilled in the art as a "pro-moiety” (e.g. described in "Design of Prodrugs", H. Bundgaard (Elsevier, 1985)) Prepared by the presence of appropriate functional groups in the compounds.
  • a pro-moiety e.g. described in "Design of Prodrugs", H. Bundgaard (Elsevier, 1985)
  • the present application also provides said compounds containing protecting groups.
  • protecting groups In any process for preparing the compounds, it may be necessary and/or desirable to protect sensitive or reactive groups on any relevant molecules, thereby forming chemically protected forms of the compounds. This can be achieved by conventional protecting groups, for example, those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 Protecting Groups, these references are incorporated herein by reference.
  • the protecting groups can be removed at an appropriate subsequent stage using methods known in the art.
  • conjugate refers to a substance in which a small molecule drug is linked to a targeting moiety.
  • the small molecule drug is connected to the targeting moiety through a linker.
  • the linker can be broken in a specific environment (such as an intracellular low pH environment, an acidic tumor microenvironment) or a specific action (such as the action of lysosomal proteases), thereby separating the small molecule drug from the targeting moiety .
  • linker refers to the fragment that connects a small molecule drug to a targeting moiety.
  • targeting moiety refers to the portion of the conjugate that is capable of specifically binding to a target (or portion of a target) on the cell surface. Through the interaction of the targeting moiety with the target, the conjugate can be delivered to a specific cell population.
  • the application provides a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N- oxides, isotopically labeled compounds, metabolites, or Prodrugs:
  • L 1 is selected from O, S and NR 5 ;
  • L 2 is a covalent bond, or is selected from C 1-6 alkylene and -C 1-6 alkylene-O-;
  • R 1 is C 1-6 alkyl, which is optionally substituted by one or more elements selected from hydrogen, halogen, hydroxyl, C 1-6 alkyl, C 1-6 alkoxy, 5-10 membered hetero Aryl and R 6 -C(O)-NH- groups substituted;
  • R 2 is selected from hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl Alkyl, heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1 -6 alkoxy, R 7 and R 7 -L 3 - groups substituted;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 3 is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 alkoxy;
  • R 4 is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 alkoxy;
  • R 5 is selected from hydrogen and C 1-6 alkyl
  • R 6 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl , heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 Substituted with alkoxy groups;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • R 1 is C 1-6 alkyl, optionally substituted by one or more hydrogen, halogen, hydroxyl, C 1-3 alkyl, C 1-3 alkoxy , 5-10 membered heteroaryl and R 6 -C(O)-NH- group substituted;
  • R 6 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl , heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 substituted by alkoxy groups.
  • R 1 is C 1-6 alkyl, optionally substituted by one or more hydrogen, halogen, hydroxyl, C 1-3 alkyl, C 1-3 alkoxy , 5-10 membered heteroaryl and R 6 -C(O)-NH- group substituted;
  • R 6 is C 1-6 alkyl.
  • R is C 1-6 alkyl, optionally substituted by one or more members selected from hydrogen, hydroxy, methyl, methoxy, and C 1-3 alkyl-C ( O)-NH- substituted by the group;
  • R 1 is C 1-6 alkyl, which is unsubstituted or substituted with methyl, hydroxy, methoxy, or C 1-3 alkyl-C(O)-NH-.
  • R 1 is C 1-6 alkyl.
  • R1 is n-butyl
  • R is selected from hydrogen, C 1-3 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl
  • the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups are optionally substituted by one or more selected from the group consisting of hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1- Substituted with 6 haloalkyl, C 1-6 alkoxy, R 7 and R 7 -L 3 - groups;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • R is selected from hydrogen, C 1-3 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl
  • the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups are optionally substituted by one or more selected from the group consisting of hydrogen, halogen, amino, hydroxyl, cyano, C 1-3 alkyl, C 1- 3 haloalkyl, C 1-3 alkoxy, R 7 and R 7 -L 3 - groups replace;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • R is selected from hydrogen, C 3-6 cycloalkyl, 3-6 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, the cycloalkyl, heterocyclyl Cyclic groups, aryl groups and heteroaryl groups are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy substituted by R 7 and R 7 -L 3 - groups;
  • R 7 is selected from C 1-3 alkyl, C 3-6 cycloalkyl and 3-6 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy and carboxyl groups;
  • L 3 is selected from C 1-3 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 8 is selected from hydrogen and C 1-3 alkyl.
  • R is selected from hydrogen, C 3-6 cycloalkyl, 3-6 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, the cycloalkyl, heterocyclyl Cyclic, aryl and heteroaryl groups are optionally substituted with one or more groups selected from hydrogen, C 1-3 alkyl, C 1-3 alkoxy, R 7 and R 7 -L 3 - ;
  • R 7 is selected from C 1-3 alkyl and 3-6 membered heterocyclyl, which alkyl and heterocyclyl are optionally substituted by one or more groups selected from hydrogen and C 1-3 alkyl ;
  • L 3 is selected from C 1-3 alkylene and -C 1-3 alkylene -NR 8 -;
  • R 8 is C 1-3 alkyl.
  • R is selected from hydrogen, C 3-6 cycloalkyl, 3-6 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, the cycloalkyl, heterocyclyl Cyclic, aryl and heteroaryl groups are optionally substituted by one or more hydrogen, C 1-3 alkyl, C 1-3 alkoxy, -(CH 2 )N(CH 3 ) 2 , -( CH 2 CH 2 )N(CH 3 ) 2 , 6-membered nitrogen-containing heterocyclyl and 6-membered nitrogen-containing heterocyclyl-methylene-substituted groups, the 6-membered nitrogen-containing heterocyclyl optionally Replaced by one or more hydrogen and methyl groups.
  • L1 is selected from O, S, and NH.
  • L 1 is NH
  • L2 is a covalent bond, or is selected from C 1-3 alkylene and -C 1-3 alkylene-O-.
  • L2 is methylene
  • R3 is selected from hydrogen, halogen, C 1-3 alkyl, and C 1-3 alkoxy.
  • R3 is hydrogen
  • R 4 is selected from hydrogen, halogen, C 1-3 alkyl, and C 1-3 alkoxy.
  • R4 is hydrogen
  • the present invention encompasses compounds of formula (I) obtained by any combination of the above preferred groups.
  • the compound has the structure of Formula (II-A):
  • R 1 is C 1-6 alkyl, optionally substituted by one or more hydrogen, halogen, hydroxyl, C 1-6 alkyl, C 1-6 alkoxy , 5-10 membered heteroaryl and R 6 -C(O)-NH- group substituted;
  • R 2 is selected from hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl Alkyl, heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1 -6 alkoxy, R 7 and R 7 -L 3 - groups substituted;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 6 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl , heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 Substituted with alkoxy groups;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • R 1 is C 1-6 alkyl, optionally substituted by one or more hydrogen, halogen, hydroxyl, C 1-3 alkyl, C 1-3 alkoxy , 5-10 membered heteroaryl and R 6 -C(O)-NH- group substituted;
  • R 6 is selected from C 1-6 alkyl, C 3-10 cycloalkyl, 3-12 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl , heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 Substituted with alkoxy groups;
  • R 2 is selected from hydrogen, C 1-3 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl Alkyl, heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1 -6 alkoxy, R 7 and R 7 -L 3 - groups substituted;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • R 1 is C 1-6 alkyl, optionally substituted by one or more hydrogen, halogen, hydroxyl, C 1-3 alkyl, C 1-3 alkoxy , 5-10 membered heteroaryl and R 6 -C(O)-NH- group substituted;
  • R 6 is C 1-6 alkyl
  • R 2 is selected from hydrogen, C 1-3 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the alkyl, cycloalkyl Alkyl, heterocyclyl, aryl and heteroaryl are optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-3 alkyl, C 1-3 haloalkyl, C 1 -3 alkoxy, R 7 and R 7 -L 3 - groups substituted;
  • R 7 is selected from C 1-6 alkyl, C 3-10 cycloalkyl and 3-12 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and carboxyl groups;
  • L 3 is selected from C 1-6 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 8 is selected from hydrogen and C 1-6 alkyl.
  • R is C 1-6 alkyl, optionally substituted by one or more members selected from hydrogen, hydroxy, methyl, methoxy, and C 1-3 alkyl-C ( O)-NH- substituted by the group;
  • R 2 is selected from hydrogen, C 3-6 cycloalkyl, 3-6 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, the cycloalkyl, heterocyclyl, aryl and Heteroaryl is optionally substituted by one or more hydrogen, halogen, amino, hydroxyl, cyano, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, R 7 and R 7 -L 3 - substituted by the group;
  • R 7 is selected from C 1-3 alkyl, C 3-6 cycloalkyl and 3-6 membered heterocyclyl, the alkyl, cycloalkyl and heterocyclyl are optionally replaced by one or more hydrogen , substituted by halogen, amino, hydroxyl, cyano, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy and carboxyl groups;
  • L 3 is selected from C 1-3 alkylene, O, S, NR 8 , -C 1-6 alkylene-NR 8 -, -C(O)-, -C(O)-NR 8 - and - NR 8 -C(O)-;
  • R 8 is selected from hydrogen and C 1-3 alkyl.
  • R is C 1-6 alkyl, optionally substituted by one or more members selected from hydrogen, hydroxy, methyl, methoxy, and C 1-3 alkyl-C ( O)-NH- substituted by the group;
  • R 2 is selected from hydrogen, C 3-6 cycloalkyl, 3-6 membered heterocyclyl, C 6-10 aryl and 5-6 membered heteroaryl, the cycloalkyl, heterocyclyl, aryl and The heteroaryl group is optionally substituted by one or more hydrogen, methyl, methoxy, -(CH 2 )N(CH 3 ) 2 , -(CH 2 CH 2 )N(CH 3 ) 2 , 6-membered Substituted with nitrogen-containing heterocyclyl and 6-membered nitrogen-containing heterocyclyl-methylene-groups, the 6-membered nitrogen-containing heterocyclyl optionally substituted with one or more hydrogen and methyl.
  • R is selected from n-butyl
  • R 2 is selected from hydrogen
  • R is selected from n-butyl
  • R 2 is selected from hydrogen
  • R is selected from n-butyl
  • R 2 is selected from hydrogen
  • the compound is selected from:
  • the application provides a drug-linker described in the following formula, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxidation thereof substances, isotopically labeled compounds, metabolites or prodrugs: D-L'
  • D is a fragment of any of the compounds described above;
  • L’ is the linker fragment.
  • L' is selected from -L 4 -L 5 -L 6 , wherein,
  • L 5 is a polyethylene glycol fragment, for example where a is selected from an integer from 1 to 10;
  • Z 2 is selected from chemical bonds and C 1-20 alkylene
  • Z 3 is selected from a chemical bond, a C 2-6 alkenylene group and a C 2-6 alkynylene group;
  • A is selected from
  • LG is the leaving group of nucleophilic substitution reaction, selected from halogen, nitro, benzenesulfonate, p-toluenesulfonate, trifluoromethanesulfonate, -S(O) 2 -R 9 and -S( O)-R 9 ;
  • R 9 is selected from the group consisting of C 1-6 alkyl and C 1-6 haloalkyl.
  • L6 is in
  • Z 2 is selected from chemical bonds and C 1-10 alkylene
  • Z 3 is selected from chemical bond, C 2-4 alkenylene and C 2-4 alkynylene;
  • A is selected from
  • LG is selected from halogen, nitro, benzenesulfonate, p-toluenesulfonate, trifluoromethanesulfonate, -S(O) 2 -R 9 and -S(O)-R 9 ;
  • R 9 is selected from the group consisting of C 1-6 alkyl and C 1-6 haloalkyl.
  • L6 is in
  • Z 2 is C 1-6 alkylene
  • Z 3 is a chemical bond
  • A is selected from
  • LG is methanesulfonyl group.
  • L6 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • L' is selected from:
  • the drug-linker is selected from the following structures:
  • the present application also provides an immunostimulatory antibody conjugate of formula (ISAC-I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, or polymorph thereof , solvate, N-oxide, isotopically labeled compound, metabolite or prodrug:
  • Ab is an antibody that can target a target antigen; for example, an antibody that targets tumor cells, such as Trastuzumab monoclonal antibody;
  • L is the fragment obtained by connecting L' as described in any of the preceding paragraphs to Ab;
  • z is selected from 1-10, such as 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • z is selected from 1-8, such as 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4- 8, 5-6, 5-7, 5-8, 6-7, 6-8 or 7-8.
  • L is selected from the following structures:
  • the immunostimulatory antibody conjugate is selected from:
  • z is independently selected from 1-8, such as 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5- 6, 5-7, 5-8, 6-7, 6-8 or 7-8.
  • the present application provides a composition
  • a composition comprising the immunostimulatory antibody conjugate described in any one of the preceding paragraphs or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer thereof, Polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs.
  • the composition may comprise a plurality of immunostimulatory antibody conjugates described herein, wherein each immunostimulatory antibody Conjugates comprise a DL described herein, wherein z is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • each antibody molecule in the composition can be coupled to 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 DLs.
  • the composition may be characterized by a "drug-to-antibody” ratio (DAR) ranging from about 1 to about 10.
  • DAR drug-to-antibody ratio
  • the composition DAR value is an integer or decimal selected from 1-8, such as 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4- 5, 4-6, 4-7, 4-8, 5-6, 5-7, 5-8, 6-7, 6-8 or 7-8.
  • the pharmaceutical composition DAR value is 7.0-8.0, such as 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9 or 8.0.
  • the application provides methods for preparing the compounds of any one of the first aspects.
  • the present application provides a method for preparing the compound of formula (II-A), which includes the following steps:
  • R 1 and R 2 are as defined above.
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from N,N-dimethylformamide, N,N-dimethylacetamide, N-methylpyrrolidone, 1,4-dioxane, tetrahydrofuran and any combination thereof, preferably tetrahydrofuran .
  • the reaction is preferably carried out in the presence of a suitable organic base, which can be selected from N,N-diisopropylethylamine, triethylamine, 4-dimethylaminopyridine and any combination thereof, preferably N,N -Diisopropylethylamine.
  • the reaction is preferably carried out at a suitable temperature, preferably -10-80°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 24 hours.
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from N,N-dimethylformamide, N,N-dimethylacetamide, N-methylpyrrolidone and any combination thereof, preferably N,N-dimethylformamide.
  • the reaction is preferably carried out at a suitable temperature, preferably 50-180°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 12 hours.
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from methanol, ethanol, isopropyl alcohol, n-butanol, tetrahydrofuran and any combination thereof, with ethanol being preferred.
  • the reaction is preferably carried out at a suitable temperature, preferably 20-100°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 24 hours.
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from dichloromethane, chloroform, tetrahydrofuran, ethyl acetate and any combination thereof, with dichloromethane being preferred.
  • the reaction is preferably carried out in the presence of a suitable oxidizing agent, which may be selected from the group consisting of m-chloroperoxybenzoic acid, hydrogen peroxide, tert-butyl hydroperoxide and any combination thereof, with m-chloroperoxybenzoic acid being preferred.
  • the reaction is preferably carried out at a suitable temperature, preferably -10-80°C.
  • the reaction is preferably carried out for a suitable time, for example 1 to 12 hours.
  • the reaction is preferably carried out in a suitable solvent.
  • the solvent may be selected from tetrahydrofuran, 1,4-dioxane, water and any combination thereof, with 1,4-dioxane being preferred.
  • the reaction is preferably carried out at a suitable temperature, preferably 50-150°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 24 hours.
  • this application also provides another method for preparing the compound of formula (II-A), which includes the following steps:
  • R 1 and R 2 are as defined above;
  • LG represents a leaving group, and the leaving group includes but is not limited to a halogen atom, a methanesulfonyloxy group, a p-toluenesulfonyloxy group, and the like.
  • Compound II-A-4 is subjected to a ring-closing reaction with an ammonia solution to obtain compound II-A-8;
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from methanol, ethanol, isopropyl alcohol, n-butanol, tetrahydrofuran and any combination thereof, with methanol being preferred.
  • the reaction is preferably carried out at a suitable temperature, preferably 20-100°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 24 hours.
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from dichloromethane, chloroform, tetrahydrofuran, ethyl acetate and any combination thereof, with dichloromethane being preferred.
  • the reaction is preferably carried out in the presence of a suitable oxidizing agent, which may be selected from the group consisting of m-chloroperoxybenzoic acid, hydrogen peroxide, tert-butyl hydroperoxide and any combination thereof, with m-chloroperoxybenzoic acid being preferred.
  • the reaction is preferably carried out at a suitable temperature, preferably -10-80°C.
  • the reaction is preferably carried out for a suitable time, such as 1 to 12 hours.
  • the reaction is preferably carried out in a suitable solvent.
  • the solvent may be selected from tetrahydrofuran, 1,4-dioxane, water and any combination thereof, with 1,4-dioxane being preferred.
  • the reaction is preferably carried out at a suitable temperature, preferably 50-150°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 24 hours.
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent can be selected from N,N-dimethylformamide, N,N-dimethylacetamide, N-methylpyrrolidone, acetone and any combination thereof, preferably N,N-dimethylformamide.
  • the reaction is preferably carried out in the presence of a suitable organic base or inorganic base, which can be selected from the group consisting of N,N-diisopropylethylamine, triethylamine, 4-dimethylaminopyridine, and sodium carbonate. , potassium carbonate, cesium carbonate, potassium tert-butoxide and any combination thereof, preferably cesium carbonate.
  • the reaction is preferably carried out at a suitable temperature, preferably 20-100°C.
  • the reaction is preferably carried out for a suitable time, such as 2 to 12 hours.
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of the compound described in any one of the preceding paragraphs or a pharmaceutically acceptable salt, ester, stereoisomer, or tautomer thereof.
  • the present application provides a kit comprising a compound as described in any one of the preceding paragraphs or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph thereof, Solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs, immunostimulatory antibody conjugates as described in any of the foregoing, compositions as described in any of the foregoing, or as described in any of the foregoing. the pharmaceutical composition described above.
  • the kit also includes instructions for use.
  • the term "pharmaceutically acceptable carrier” refers to a diluent, adjuvant, excipient, or vehicle with which a therapeutic agent is administered and which is, within the scope of sound medical judgment, suitable Exposure to human and/or other animal tissue without undue toxicity, irritation, allergic reactions, or other problems or complications commensurate with a reasonable benefit/risk ratio.
  • compositions include, but are not limited to, sterile liquids.
  • the pharmaceutical composition may be, for example, in the form of a solid preparation, a semi-solid preparation, a liquid preparation, a gaseous preparation, or the like.
  • compositions may act systemically and/or locally.
  • they may be administered by a suitable route, for example by injection or transdermally; or orally or by inhalation.
  • the content or amount of the compound in the pharmaceutical composition may be from about 0.001 mg to about 1000 mg, suitably from 0.01 to 800 mg, preferably from 0.05 to 500 mg.
  • the application provides methods of preparing the pharmaceutical compositions, the methods comprising combining the compounds or pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs thereof Form, solvate, N- The oxide, isotopically labeled compound, metabolite or prodrug is combined with one or more pharmaceutically acceptable carriers.
  • the immunostimulatory antibody conjugate comprises a small molecule ligand capable of interacting with TLR7 and/or TLR8.
  • Another object of the present application is to provide the compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled Use of the compound, metabolite or prodrug, the immunostimulatory antibody conjugate, the composition, or the pharmaceutical composition in the preparation of a medicament for the prevention and/or treatment of TLR7 and/or TLR8 mediated related diseases.
  • Another object of the present application is to provide the compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled Compounds, metabolites or prodrugs, said immunostimulatory antibody conjugates, said compositions, or said pharmaceutical compositions for preventing and/or treating TLR7 and/or TLR8 mediated related diseases.
  • Another object of the present application is to provide a method for preventing and/or treating TLR7 and/or TLR8-mediated related diseases, which includes administering a preventive or therapeutically effective amount of the compound or a pharmaceutically acceptable amount thereof to an individual in need thereof.
  • a preventive or therapeutically effective amount of the compound or a pharmaceutically acceptable amount thereof to an individual in need thereof.
  • the TLR7 and/or TLR8-mediated disease is preferably a tumor.
  • an effective amount refers to an amount sufficient to achieve the desired preventive or therapeutic effect, for example, an amount that achieves alleviation of one or more symptoms associated with the disease to be treated.
  • Dosage regimens can be adjusted to provide the best desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dosage may be proportionally reduced or increased as the exigencies of the therapeutic situation indicate. It is noted that dosage values may vary depending on the type and severity of the condition to be alleviated, and may include single or multiple doses. It is further understood that, for any particular individual, specific dosage regimens should be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compound.
  • the amount of the compound administered will depend on the individual being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound, and the judgment of the prescribing physician. In some cases, dosage levels no higher than the lower end of the foregoing ranges may be sufficient, while in other cases, larger dosages may still be employed without causing any deleterious side effects, provided that the larger dosage is first The dose is divided into several smaller doses to be administered throughout the day.
  • treating means reversing, alleviating, ameliorating the disorder or condition to which such term is applied, or the progression of one or more symptoms of such disorder or condition.
  • prevention refers to inhibiting and delaying the onset of a disease, including not only prevention before the disease develops, but also prevention of recurrence after treatment.
  • “Individual” as used herein includes humans or non-human animals.
  • Exemplary human subjects include human subjects (referred to as patients) suffering from a disease, such as those described herein, or normal subjects.
  • “Non-human animals” as used herein includes all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals, and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
  • the structure of the compound is determined by nuclear magnetic resonance ( 1 H NMR) or mass spectrometry (MS).
  • the measurement instrument of 1 H NMR is JEOL Eclipse 400 nuclear magnetic instrument.
  • the measurement solvent is deuterated methanol (CD 3 OD), deuterated chloroform (CDCl 3 ) or hexadeuterated dimethyl sulfoxide (DMSO-d 6 ), and the internal standard is For tetramethylsilane (TMS), chemical shifts ( ⁇ ) are given in parts per million (ppm).
  • the measuring instrument for MS is Agilent (ESI) mass spectrometer, manufacturer: Agilent, model: Agilent 6120B.
  • Instrument model Agilent 1260, chromatographic column: Waters SunFire Prep C18 OBD (19mm ⁇ 150mm ⁇ 5.0 ⁇ m); chromatographic column temperature: 25°C; flow rate: 20.0mL/min; detection wavelength: 214nm; elution gradient: (0min: 10% A, 90% B; 16.0min: 90% A, 10% B); mobile phase A: acetonitrile; mobile phase B: 0.05% formic acid aqueous solution.
  • the thin layer chromatography silica gel plate used an aluminum plate (20 ⁇ 20 cm) produced by Merck, and the specification used for thin layer chromatography separation and purification was GF 254 (1 mm) produced in Yantai.
  • the reaction is monitored by thin layer chromatography (TLC) or LC-MS; the developing solvent systems used include: methylene chloride and methanol system, n-hexane and ethyl acetate system, and petroleum ether and ethyl acetate system.
  • TLC thin layer chromatography
  • LC-MS LC-MS
  • the volume of solvent The ratio can be adjusted according to the polarity of the compound or by adding triethylamine, etc.
  • Microwave reaction uses Biotage Initiator+ (400W, RT ⁇ 300°C) microwave reactor.
  • Column chromatography generally uses 200 to 300 mesh silica gel as the carrier.
  • the eluent system includes: methylene chloride and methanol system, as well as petroleum ether and ethyl acetate system.
  • the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of triethylamine can also be added for adjustment.
  • Instrument model Biotage rapid medium-pressure preparative chromatography, chromatographic column: Agela C18 reversed-phase column (Spherical; 20-35 ⁇ m; 100A); chromatographic column temperature: 25°C; flow rate: 28.0mL/min; detection wavelength: 220nm; mobile phase A : acetonitrile; mobile phase B: water.
  • the reaction temperature is room temperature (20°C to 35°C);
  • the reagents used in this article were purchased from Acros Organics, Aldrich Chemical Company, Tebot Chemical and other companies.
  • Step 1 Preparation of 4-(butylamino)-6-methyl-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester
  • 6-Chloro-4-methyl-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester (12g, 48.15mmol), N,N-diisopropylethylamine (12.6g, 96.31mmol) and n-butyl Amine (5.34g, 72.23mmol) was dissolved in tetrahydrofuran (200mL) and stirred at room temperature for 12 hours.
  • Step 2 Preparation of ethyl 4-(butylamino)-6-(2-(dimethylamino)vinyl)-2-(methylthio)pyrimidine-5-carboxylate
  • Step 5 Preparation of 4-(butylamino)-2-((2,4-dimethoxybenzyl)amino)pyrido[4,3-d]pyrimidin-5(6H)-one
  • Step 1 Preparation of 4-(butylamino)-6-methyl-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester
  • 6-Chloro-4-methyl-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester (1.0g, 4.05mmol), N,N-diisopropylethylamine (0.78g, 6.02mmol) and n- Butylamine (0.44g, 6.02mmol) was dissolved in tetrahydrofuran (20mL) and stirred at room temperature for 12 hours.
  • Step 2 Preparation of ethyl 4-(butylamino)-6-(2-(dimethylamino)vinyl)-2-(methylthio)pyrimidine-5-carboxylate
  • Step 4 Preparation of 4-(butylamino)-6-methyl-2-(methylsulfinyl)-5,6-dihydropyridin[4,3-d]pyrimidin-5-one
  • Step 1 Preparation of ethyl 4-((1,3-dimethoxypropan-2-yl)amino)-6-methyl-2-(methylthio)pyrimidine-5-carboxylate
  • Example 2 The synthetic route of Example 1 was adopted, and the first reaction raw material n-butylamine was replaced with 2-amino-1,3-dimethoxypropane to obtain the title compound (1.4 g, yield: 89.3%).
  • Step 2 4-((1,3-dimethoxypropan-2-yl)amino)-6-(2-(dimethylamino)vinyl)-2-(methylthio)pyrimidine-5 -Preparation of ethyl formate
  • Step 3 4-((1,3-dimethoxypropan-2-yl)amino)-6-methyl-2-(methylthio)-5,6-dihydropyridine[4,3- d] Preparation of pyrimidin-5-one
  • Step 4 4-((1,3-dimethoxypropan-2-yl)amino)-6-methyl-2-(methylsulfinyl)-5,6-dihydropyridine[4, Preparation of 3-d]pyrimidin-5-one
  • Step 1 Preparation of ethyl 4-((1,3-dihydroxypropan-2-yl)amino)-6-methyl-2-(methylthio)pyrimidine-5-carboxylate
  • Example 2 The synthetic route of Example 1 was adopted, and n-butylamine, the raw material in the first step, was replaced with 2-amino-1,3-propanediol to obtain the title compound (0.54g, yield: 83.6%).
  • Step 2 4-((1,3-dihydroxypropan-2-yl)amino)-6-(2-(dimethylamino)vinyl)-2-(methylthio)pyrimidine-5-carboxylic acid Preparation of ethyl ester
  • Step 3 4-((1,3-dihydroxypropan-2-yl)amino)-6-methyl-2-(methylthio)-5,6-dihydropyridine[4,3-d] Preparation of pyrimidin-5-one
  • Step 4 4-((1,3-dihydroxypropan-2-yl)amino)-6-methyl-2-(methylsulfinyl)-5,6-dihydropyridine[4,3- d] Preparation of pyrimidin-5-one
  • Step 1 4-((2-Amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)-3-methoxy
  • Step 1 4-((2-Amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)-3-methoxy
  • Step 3 4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)-3-methoxy
  • Step 1 Preparation of tert-butyl 4-(4-(chloromethyl)benzyl)piperazine-1-carboxylate
  • Step 1 Preparation of 2-amino-6-(4-bromobenzyl)-4-(butylamino)pyrido[4,3-d]pyrimidin-5(6H)-one
  • Adopt the synthesis method in the first step of Example 5 replace the reaction raw material 4-(bromomethyl)-3-methoxybenzoic acid methyl ester with 1-bromo-4-(bromomethyl)benzene to obtain the title compound ( 250mg, yield: 58.2%).
  • Step 1 4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)piperidine-1- Preparation of tert-butyl formate
  • Step 4 Preparation of ethyl 4-((1-hydroxy-2-methylhexan-2-yl)amino)-6-methyl-2-(methylthio)pyrimidine-5-carboxylate
  • Step 5 4-((1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)amino)-6-methyl-2-(methylthio) ) Preparation of ethyl pyrimidine-5-carboxylate
  • Step 6 4-((1-((tert-butyldimethylsilyl)oxy)-2-methylhexan-2-yl)amino)-6-(2-(dimethylamino) Preparation of ethyl vinyl)-2-(methylthio)pyrimidine-5-carboxylate
  • Step 7 4-((1-((tert-butyldimethylsilyl)oxy)-2-methylhexan-2-yl)amino)-6-methyl-2-(methylthio) ) Preparation of pyrido[4,3-d]pyrimidin-5(6H)-one
  • Step 8 4-((1-((tert-butyldimethylsilyl)oxy)-2-methylhexan-2-yl)amino)-6-methyl-2-(methylylidene Preparation of sulfonyl)pyrido[4,3-d]pyrimidin-5(6H)-one
  • Step 9 4-((1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)amino)-2-((2,4-dimethoxy Preparation of benzyl)amino)-6-methylpyrido[4,3-d]pyrimidin-5(6H)-one
  • step 6 of Intermediate Preparation Example 1 The synthesis method of step 6 of Intermediate Preparation Example 1 is adopted, and the reaction raw material 4-(butylamino)-2-((2,4-dimethoxybenzyl)amino)pyrido[4,3-d] Pyrimidine-5(6H)-one replaced with 4-((1-((tert-butyldimethylsilyl)oxy)-2-methylhexan-2-yl)amino)-2-((2 ,4-dimethoxybenzyl)amino)-6-methylpyrido[4,3-d]pyrimidin-5(6H)-one to obtain the title compound (25 mg, yield: 39.3%).
  • Step 1 2-((2-Amino-6-methyl-5-oxo-5,6-dihydropyrido[4,3-d]pyrimidin-4-yl)amino)-2-methyl Preparation of hexanal
  • Step 2 2-amino-4-((1-((2,4-dimethoxybenzyl)amino)-2-methylhexan-2-yl)amino)-6-methylpyrido[ Preparation of 4,3-d]pyrimidin-5(6H)-one
  • step 6 of Intermediate Preparation Example 1 The synthesis method of step 6 of Intermediate Preparation Example 1 is adopted, and the reaction raw material 4-(butylamino)-2-((2,4-dimethoxybenzyl)amino)pyrido[4,3-d] Pyrimidine-5(6H)-one replaced with 2-amino-4-((1-((2,4-dimethoxybenzyl)amino)-2-methylhexan-2-yl)amino)-6 -methylpyrido[4,3-d]pyrimidin-5(6H)-one to obtain the title compound (0.23 g, yield: 52.3%).
  • Step 4 N-(2-((2-amino-6-methyl-5-oxo-5,6-dihydropyrido[4,3-d]pyrimidin-4-yl)amino)-2 - Preparation of methylhexyl)acetamide (compound 18)
  • the first step Preparation of (26-oxo-3,6,9,12,15,18,21,24-octaoxahexadecyl)carbamic acid tert-butyl ester
  • Step 2 (26-(4-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl Preparation of tert-butyl)benzyl)piperazin-1-yl)-3,6,9,12,15,18,21,24-octaoxahexadecyl)carbamate
  • Step 3 2-amino-6-(4-((4-(26-amino-3,6,9,12,15,18,21,24-octaoxahexadecyl)piperazine- Preparation of 1-yl)methyl)benzyl)-4-(butylamino)pyrido[4,3-d]pyrimidin-5(6H)-one
  • Step 4 N-(26-(4-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl )Methyl)benzyl)piperazin-1-yl)-3,6,9,12,15,18,21,24-octaoxahexadecyl)-6-(2-(methylsulfonate Preparation of acyl)pyrimidin-5-yl)hex-5-ynamide
  • Step 1 (24-(4-(4-((2-Amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl Preparation of tert-butyl)benzyl)piperazin-1-yl)-24-oxo-3,6,9,12,15,18,21-heptaoxatetradecyl)carbamate
  • Step 2 2-amino-6-(4-((4-(1-amino-3,6,9,12,15,18,21-heptaoxa-24-yl)piperazine) Preparation of -1-yl)methyl)benzyl)-4-(butylamino)pyrido[4,3-d]pyrimidin-5(6H)-one
  • Step 3 N-(24-(4-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl )Methyl)benzyl)piperazin-1-yl)-24-oxo-3,6,9,12,15,18,21-heptaoxatetradecyl)-6-(2-( Preparation of methylsulfonyl)pyrimidin-5-yl)hex-5-ynamide (compound 21)
  • Step 1 (26-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)piper Preparation of tert-butyl (din-1-yl)-3,6,9,12,15,18,21,24-octaoxahexadecyl)carbamate
  • Step 2 2-amino-6-((1-(26-amino-3,6,9,12,15,18,21,24-octaoxahexadecyl)piperidin-4-yl Preparation of )methyl)-4-(butylamino)pyrido[4,3-d]pyrimidin-5(6H)-one
  • Step 3 N-(26-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl) )piperidin-1-yl)-3,6,9,12,15,18,21,24-octaoxahexadecyl)-6-(2-(methylsulfonyl)pyrimidine-5- Preparation of hex-5-ynamide (compound 22)
  • Example 20 1-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)benzyl )-N-(33-(2-(methylsulfonyl)pyrimidin-5-yl)-28-oxo-3,6,9,12,15,18,21,24-octaxa-27- Preparation of azatritri-32-yn-1-yl)piperidine-4-carboxamide (compound 23)
  • Step 1 Preparation of 1-(4-(chloromethyl)benzyl)piperidine-4-carboxylic acid methyl ester
  • Adopt the synthesis method of the first step of Example 6 replace (6-(chloromethyl)pyridin-3-yl)methanol with 1-(4-(chloromethyl)benzyl)piperidine-4-carboxylic acid methyl ester to obtain the title compound (172 mg, yield: 56.5%).
  • Step 4 1-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)benzyl
  • Step 5 1-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)benzyl
  • Step 6 1-(4-((2-amino-4-(butylamino)-5-oxopyrido[4,3-d]pyrimidin-6(5H)-yl)methyl)benzyl )-N-(33-(2-(methylsulfonyl)pyrimidin-5-yl)-28-oxo-3,6,9,12,15,18,21,24-octaxa-27- Preparation of azatritri-32-yn-1-yl)piperidine-4-carboxamide (compound 23)
  • LC-MS was used to determine the ISAC molecular weight and calculate the drug/antibody ratio DAR value.
  • Liquid chromatography column Thermo MAbPac RP 3.0*100mm;
  • Mobile phase A 0.1% FA/H 2 O
  • mobile phase B 0.1% FA/ACN
  • Mass spectrometer model AB Sciex Triple TOF 5600+;
  • CE-SDS calculated the DAR value of immunostimulatory antibody conjugate ISAC-1: 8.0; the DAR value of ISAC-2: 8.0; the DAR value of ISAC-3: 7.99; the DAR value of ISAC-4: 8.0.
  • HEK-Blue hTLR7 cells (Invivogen) were cultured in DMEM medium (Hyclone) containing 10% FBS heat-inactivated fetal bovine serum (Corning). On the day of the test, observe the cell status under a microscope, collect the cells and resuspend them. After counting, adjust the cell concentration to 50 ⁇ L per well, with a total cell volume of 2 ⁇ 10 4 , and seed the cells in a 96-well plate.
  • Emax is the OD 620 at which the activation effect of the test compound reaches the maximum.
  • the activating activity of the compound on HEK-Blue hTLR7 cells was determined according to the above method.
  • the activity of the compound was expressed as EC 50 and Emax.
  • the results are shown in Table 1:
  • HEK-Blue hTLR8 cells (Invivogen) were cultured in DMEM medium (Hyclone) containing 10% FBS heat-inactivated fetal bovine serum (Corning). On the day of the test, observe the cell status under a microscope, collect the cells and resuspend them. After counting, adjust the cell concentration to 50 ⁇ L per well, with a total cell volume of 2 ⁇ 10 4 , and seed the cells in a 96-well plate.
  • Emax is the OD 620 at which the activation effect of the test compound reaches the maximum.
  • the activating activity of the compound on HEK-Blue hTLR8 cells was determined according to the above method.
  • the activity of the compound was expressed as EC 50 and Emax.
  • the results are shown in Table 2:
  • HCC1954 tumor cells (Connuotech) in the logarithmic growth phase, wash them twice with PBS, and resuspend the cells in RPMI1640 + 10% heat-inactivated FBS complete medium to adjust the cell density to 2 ⁇ 10 5 cells /mL, HCC1954 tumor cells were seeded in a 96-well plate (Corning), with 1 ⁇ 10 4 cells per well.
  • PBMC Resuscitate PBMC (Sail Bio), resuspend the cells in RPMI1640+10% heat-inactivated FBS complete medium, adjust the density to 6 ⁇ 10 5 cells/mL, and add it to a 96-well plate inoculated with tumor cells. 3 ⁇ 10 4 PBMC cells.
  • Each SCID-beige mouse (Vital Lever) was subcutaneously inoculated with 5 ⁇ 10 6 HCC1954 cells (mixed with Matrigel in an equal volume of 1:1 and suspended in 0.1 mL of PBS) in the right armpit.
  • mice with irregular, small or large tumors will be eliminated.
  • the remaining mice will be randomly divided into groups according to tumor volume and animal weight, and will be given a single intraperitoneal injection (ip).
  • Physiological saline (vehicle control), Trastuzumab monoclonal antibody, and immunostimulatory antibody conjugate ISAC-1 were used to observe the efficacy of the test compound on the tumor-bearing mouse model and the animal's tolerance to the test compound.
  • TGI (%) (tumor volume) [1-(T Vt -T V0 )/(C Vt -C V0 )] ⁇ 100%
  • T V0 is the average tumor volume of the test compound group when administered in groups
  • T Vt is the average tumor volume of the test compound group on days t after administration
  • C V0 is the average tumor volume of the vehicle group when administered in groups
  • C Vt is The average tumor volume of the vehicle group on day t after administration.
  • TGI (%) (tumor volume) 100% - (T Vt - T V0 )/T V0 ⁇ 100%. If the tumor shrinks from the initial volume, that is, when V t ⁇ V 0 , it is defined as partial tumor regression (PR); if the tumor disappears completely, it is defined as complete tumor regression (CR).
  • PR partial tumor regression
  • CR complete tumor regression

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

提供一类双并环化合物、其制备方法及用途,特别地,提供式(I)所示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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,以及药物组合物、制备方法和用途。

Description

一类双并环化合物、其制备方法及用途
本申请是以CN申请号为202211126580.X,申请日为2022年9月16日的申请为基础,并主张其优先权,该CN申请的公开内容在此作为整体引入本申请中。
技术领域
本申请属于药物化学领域,具体涉及一类双并环化合物、其制备方法及用途。
背景技术
Toll样受体(Toll-like receptors,TLRs)属于模式识别受体(Pattern recognition receptor,PRR)家族,能特异识别病原相关分子模式(PAMP),是参与非特异性免疫的一类重要蛋白质,也是连接非特异性免疫和特异性免疫的桥梁。迄今为止,哺乳动物中共发现13种Toll样受体,在人中发现10种(TLR1-10),其中TLR1/2/4/5/6/10表达于细胞表面,能快速识别细菌代谢的产物,TLR3/7/8/9表达于细胞内体中,主要识别和监测来自病原体细胞内的外源性核酸类物质。
TLR7在人身上具有有限的表达分布,主要通过B细胞和浆细胞样树突状细胞(pDC)表达而对病毒性感染产生应答,诱导高水平的干扰素-α,并增强pDC的共刺激和抗原递呈能力,促进CD4+T细胞的增殖,进一步激活CD8+T细胞从而产生适应性T细胞应答。TLR8表达相对更广泛,在人身上主要通过单核细胞,NK细胞和髓样树突状细胞(mDC)表达。刺激TLR8可以诱导各种不同的促炎细胞因子释放,如肿瘤坏死因子-α(TNF-α)、IL-6、IL-12和干扰素-γ。同时,TLR8信号通路还被证明是逆转Tregs细胞抑制功能所必要且充分的条件。因此TLR7和TLR8在机体的固有免疫和获得性免疫中都起到重要作用,是抗癌和抗病毒免疫疗法的热门靶点。目前已有不同公司涉及多个TLR7和/或TLR8激动剂的报道,但具备更好活性、安全性和理化性质的TLR7和/或TLR8激动剂仍然具有很大需求。
目前一种新的免疫刺激抗体偶联物(Immune-stimulating antibody conjugates,ISAC)正在兴起,其通过可切割或不可切割的连接体将免疫调节剂如TLR7和/或TLR8激动剂与靶向肿瘤的抗体偶联形成。免疫刺激抗体偶联物结合了抗体的精确肿瘤定位与免疫调节剂如TLR7和/或TLR8激动剂的持久抗肿瘤活性和免疫记忆功效,在实现系统给药的同时安全引发抗肿瘤免疫反应。
本申请提供了一类具有TLR7和/或TLR8激动活性的双并环化合物。该类化合物具 有更好的活性,安全性及理化性质,并且该类化合物可进一步用于免疫刺激抗体偶联物,对于肿瘤及病毒药物的开发具有重要意义。
发明内容
一方面,本申请提供一类双并环化合物,这类化合物对TLR7和/或TLR8具有很强的激动作用,因此具有更好的预防和/或治疗TLR7和/或TLR8介导的相关疾病的用途。所述化合物还被发现具有良好的物理化学性质(例如溶解度、物理和/或化学稳定性)和良好的安全性。
所述化合物为式(I)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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
其中,
L1选自O、S和NR5
L2为共价键,或选自C1-6亚烷基和-C1-6亚烷基-O-;
R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-6烷基、C1-6烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R2选自氢、C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R3选自氢、卤素、C1-6烷基和C1-6烷氧基;
R4选自氢、卤素、C1-6烷基和C1-6烷氧基;
R5选自氢和C1-6烷基;
R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基的基团所取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
R8选自氢和C1-6烷基。
在另一个方面,本申请提供下式所述药物-连接体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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
D-L’
其中,D为前文所述化合物的片段;
L’为连接体片段。
本申请还提供式(ISAC-I)所示的免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
其中,
D为前文所述化合物的片段;
Ab是可靶向目标抗原的抗体;
L为前文所述药物-连接体中的L’与Ab连接后得到的片段;
z选自1-10。
另一方面,本申请提供一种组合物,其包含所述免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药。
另一方面,本申请提供了式(I)所示化合物在制备免疫刺激抗体偶联物中的用途。
另一方面,本申请提供了一种药物组合物,其包含预防和/或治疗有效量的第一方面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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、式(ISAC-I)所示的免疫刺激抗体偶联物、或者所述组合物以及一种或多种药学上可接受的载体。
另一方面,本申请提供了一种药盒,其包含第一方面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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、式(ISAC-I)所示的免疫刺激抗体偶联物、所述组合物或者所述药物组合物。
另一方面,本申请提供了第一方面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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、式(ISAC-I)所示免疫刺激抗体偶联物、所述组合物或者药物组合物在制备用于预防和/或治疗TLR7和/或TLR8介导的相关疾病的药物中的用途。
另一方面,本申请提供了第一方面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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、式(ISAC-I)所示免疫刺激抗体偶联物、所述组合物或者药物组合物,其用于预防和/或治疗TLR7和/或TLR8介导的相关疾病。
另一方面,本申请提供了制备式(I)所示化合物的方法。
定义
除非在下文中另有定义,本文中所用的所有技术术语和科学术语的含义意图与本领域技术人员通常所理解的相同。提及本文中使用的技术意图指在本领域中通常所理解的技术,包括那些对本领域技术人员显而易见的技术的变化或等效技术的替换。虽然相信以下术语对于本领域技术人员很好理解,但仍然阐述以下定义以更好地解释本发明。
如本文中所使用,术语“包括”、“包含”、“具有”、“含有”或“涉及”及其在本文中的其它变体形式为包含性的(inclusive)或开放式的,且不排除其它未列举的元素或方法步骤。
如本文中所使用,术语“烷基”定义为直链或支链的饱和脂肪族烃基。例如,如本文中所使用,术语“C1-6烷基”指具有1至6个碳原子的直链或支链的基团(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基),其任选地被一个或多个(诸如1至3个)适合的取代基如卤素取代。术语“C1-3烷基”指具有1至3个碳原子的直链或支链的基团(例如甲基、乙基、正丙基、异丙基),其任选地被一个或多个(诸如1至3个)适合的取代基如卤素取代。
如本文中所使用,术语“烷氧基”是指“烷基-O-”,所述的“烷基”如前文所定义。例如,术语“C1-6烷氧基”是指“C1-6烷基-O-”,所述的“C1-6烷基”如前文所定义。本文所述的“C1-3烷氧基”是指“C1-3烷基-O-”,所述的“C1-3烷基”如前文所定义。示例性的C1-6烷氧基包括甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、仲丁氧基、叔丁氧基、正戊氧基或正己氧基。
如本文中所使用,术语“C1-6亚烷基”是指直链或支链的含1-6个碳原子的烷烃去除两个氢所形成的二价基团,包括“C1-5亚烷基”、“C1-4亚烷基”、“C1-3亚烷基”、“C1-2亚烷基”,具体实例包括但不限于:-CH2-、-CH2CH2-、-CH2CH2CH2-、-CH2CH2CH2CH2-、-CH2CH2CH2CH2CH2-、-CH2CH2CH2CH2CH2CH2-等。
如本文中所使用,术语“环烷基”指饱和或部分不饱和的非芳族单环或多环(诸如双环)烃环(例如单环,诸如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基,或双环,包括螺环、稠合或桥连系统,诸如双环[1.1.1]戊基、双环[2.2.1]庚基、双环[3.2.1]辛基或双环[5.2.0]壬基、十氢化萘基等),其任选地被一个或多个(诸如1至3个)适合的取代基取代。例如,术语“C3-6环烷基”指具有3至6个成环碳原子的饱和或部分不饱和的非芳族单环或多环(诸如双环)烃环(例如环丙基、环丁基、环戊基或环己基),其任选地被一个或多个(诸如1至3个)适合的取代基取代,例如甲基取代的环丙基。
如本文中所使用,术语“卤素”基团定义为包括氟、氯、溴或碘。
如本文中所使用,术语“卤代”是指被一个或多个(诸如1至3个)相同或不同的卤素原子取代。
如本文中所使用,术语“卤代烷基”是指被一个或多个(诸如1至3个)相同或不同的卤素原子取代的烷基。例如,术语“C1-6卤代烷基”指具有1至6个碳原子的卤代烷基,例如-CF3、-C2F5、-CHF2、-CH2F、-CH2CF3、-CH2Cl或-CH2CH2CF3等。
如本文中所使用,术语“杂环”或“杂环基”指饱和或部分不饱和的非芳族单环或多环基团,例如其在环中具有2、3、4、5、6、7、8或9个碳原子和一个或多个(例如1个、2个、3个或4个)独立地选自N、O或S(O)t(其中t是0、1或2)的杂原子,例如3-12元杂环基、3-7元杂环基、3-6元杂环基、5-6元杂环基等,例如5-10元含氮杂环基、6-10元含氧杂环基、6-8元含硫杂环基、5-8元含氧杂环基等。杂环基的代表性实例包括但不限于环氧乙烷基、氮丙啶基、氮杂环丁基(azetidinyl)、氧杂环丁基(oxetanyl)、四氢呋喃基、四氢吡咯烷基、六氢-1H-吡咯啉、吡咯烷酮基、咪唑烷基、吡唑烷基、四氢吡喃基、四氢吡啶基、哌啶基、吗啉基、二噻烷基(dithianyl)、硫吗啉基、哌嗪基等。
如本文中所使用,术语“芳基”或“芳环”指具有共轭π电子系统的全碳单环或稠合环多环芳族基团。例如,术语“C6-10芳基”或“C6-10芳环”指含有6至10个碳原子的芳族基团,诸如苯基(环)或萘基(环)。芳基或芳环任选地被1或多个(诸如1至3个)适合的取代基(例如卤素、-OH、-CN、-NO2、C1-6烷基等)取代。
在本文中,术语“杂芳基”是指具有芳香性的环状基团,其中至少一个环原子为杂原子,例如氮原子、氧原子或硫原子。任选地,环状结构中的环原子(例如碳原子、氮原子或硫原子)可以被氧代。具体实例包括但不限于5-10元杂芳基、5-10元含氮杂芳基、6-10元含氧杂芳基、6-8元含氮杂芳基、5-8元含氧杂芳基等,例如呋喃基、噻吩基、吡咯基、噻唑基、异噻唑基、噻二唑基、噁唑基、异噁唑基、噁二唑基、咪唑基、吡唑基、1,2,3-三唑基、1,2,4-三唑基、1,2,3-噁二唑基、1,2,4-噁二唑基、1,2,5-噁二唑基、1,3,4-噁二唑基、吡啶基、2-吡啶酮基、4-吡啶酮基、嘧啶基、1,4-二氧杂环己二烯基、2H-1,2-噁嗪基、4H-1,2-噁嗪基、6H-1,2-噁嗪基、4H-1,3-噁嗪基、6H-1,3-噁嗪基、4H-1,4-噁嗪基、哒嗪基、吡嗪基、1,2,3-三嗪基、1,3,5-三嗪基、1,2,4,5-四嗪基、氮杂环庚三烯基、1,3-二氮杂环庚三烯基、氮杂环辛四烯基等。
任选地,本文中涉及到的基团中的氢可以被氘取代。
术语“取代”指所指定的原子上的一个或多个(例如1个、2个、3个或4个)氢被从所指出的基团的选择代替,条件是未超过所指定的原子在当前情况下的正常原子价并且所述取代形成稳定的化合物。取代基和/或变量的组合仅仅当这种组合形成稳定的化合物时才是允许的。
如果取代基被描述为“任选地被….取代”,则取代基可(1)未被取代或(2)被取代。如果取代基的碳被描述为任选地被取代基列表中的一个或多个取代,则碳上的一个或多个氢(至存在的任何氢的程度)可单独和/或一起被独立地选择的取代基替代或未替代。如果取 代基的氮被描述为任选地被取代基列表中的一个或多个取代,则氮上的一个或多个氢(至存在的任何氢的程度)可各自被独立地选择的取代基替代或未替代。
如果取代基被描述为“独立地选自”一组基团,则各取代基独立于另一者被选择。因此,各取代基可与另一(其他)取代基相同或不同。
如本文中所使用,术语“一个或多个”意指在合理条件下的1个或超过1个,例如2个、3个、4个、5个、6个、7个、8个、9个或10个。
除非指明,否则如本文中所使用,取代基的连接点可来自取代基的任意适宜位置。
当取代基的键显示为穿过环中连接两个原子的键时,则这样的取代基可键连至该可取代的环中的任一成环原子。
本申请还提供所有药学上可接受的同位素标记的化合物,其与所述化合物相同,除了一个或多个原子被具有相同原子序数但原子质量或质量数不同于在自然界中占优势的原子质量或质量数的原子替代。适合的同位素的实例包括但不限于氢的同位素(例如2H、3H、氘D、氚T);碳的同位素(例如11C、13C及14C);氯的同位素(例如37Cl);氟的同位素(例如18F);碘的同位素(例如123I及125I);氮的同位素(例如13N及15N);氧的同位素(例如15O、17O及18O);磷的同位素(例如32P);及硫的同位素(例如35S)。某些同位素标记的化合物(例如掺入放射性同位素的那些)可用于药物和/或底物组织分布研究(例如分析)中。放射性同位素氚(即3H)及碳-14(即14C)因易于掺入且容易检测而特别可用于该目的。用正电子发射同位素(例如11C、18F、15O及13N)进行取代可在正电子发射断层显像术(PET)研究中用于检验底物受体占据情况。被同位素标记的化合物可通过与描述于随附路线和/或实施例及制备中的那些类似的方法通过使用适当的被同位素标记的试剂代替之前采用的非标记的试剂来制备。本文中所述药学上可接受的溶剂合物包括其中结晶溶剂可被同位素取代的那些,例如,D2O、丙酮-d6或DMSO-d6
术语“立体异构体”表示由于至少一个不对称中心形成的异构体。在具有一个或多个(例如1个、2个、3个或4个)不对称中心的化合物中,其可产生外消旋混合物、单一对映异构体、非对映异构体混合物和单独的非对映异构体。特定个别分子也可以几何异构体(顺式/反式)存在。类似地,所述化合物可以两种或更多种处于快速平衡的结构不同的形式的混合物(通常称作互变异构体)存在。互变异构体的代表性实例包括酮-烯醇互变异构体、苯酚-酮互变异构体、亚硝基-肟互变异构体、亚胺-烯胺互变异构体等。要理解,本申请的范围涵盖所有这样的以任意比例(例如60%、65%、70%、75%、80%、85%、 90%、95%、96%、97%、98%、99%)的异构体或其混合物。
本申请还提供所述化合物的所有可能的结晶形式或多晶型物,其可为单一多晶型物或多于一种多晶型物的任意比例的混合物。
还应当理解,本文中某些化合物可以游离形式存在用于治疗,或适当时,以其药学上可接受的衍生物形式存在。在本文中,药学上可接受的衍生物包括但不限于:药学上可接受的盐、溶剂合物、代谢物或前药,在将它们向需要其的患者给药后,能够直接或间接提供所述化合物或其代谢物或残余物。因此,当在本文中提及“所述化合物”时,也意在涵盖化合物的上述各种衍生物形式。
所述化合物的药学上可接受的盐包括其酸加成盐及碱加成盐。适合的酸加成盐由形成药学上可接受盐的酸来形成。适合的碱加成盐由形成药学上可接受盐的碱来形成。适合的盐的综述参见Stahl及Wermuth的“Handbook of Pharmaceutical Salts:Properties,Selection,and Use”(Wiley-VCH,2002)。用于制备所述化合物的药学上可接受的盐的方法为本领域技术人员已知的。
所述化合物可以溶剂合物(优选水合物)的形式存在,其中所述化合物包含作为所述化合物晶格的结构要素的极性溶剂。极性溶剂特别是水的量可以化学计量比或非化学计量比存在。
本领域技术人员会理解,由于氮需要可用的孤对电子来氧化成氧化物,因此并非所有的含氮杂环都能够形成N-氧化物;本领域技术人员会识别能够形成N-氧化物的含氮杂环。本领域技术人员还会认识到叔胺能够形成N-氧化物。用于制备杂环和叔胺的N-氧化物的合成方法是本领域技术人员熟知的,包括用过氧酸如过氧乙酸和间氯过氧苯甲酸(MCPBA)、过氧化氢、烷基过氧化氢如叔丁基过氧化氢、过硼酸钠和双环氧乙烷(dioxirane)如二甲基双环氧乙烷来氧化杂环和叔胺。这些用于制备N-氧化物的方法已在文献中得到广泛描述和综述,参见例如:T.L.Gilchrist,Comprehensive Organic Synthesis,vol.7,pp 748-750;A.R.Katritzky和A.J.Boulton,Eds.,Academic Press;以及G.W.H.Cheeseman和E.S.G.Werstiuk,Advances in Heterocyclic Chemistry,vol.22,pp 390-392,A.R.Katritzky和A.J.Boulton,Eds.,Academic Press。
本申请还提供所述化合物的代谢物,即在给药所述化合物时体内形成的物质。这样的产物可由例如被给药的化合物的氧化、还原、水解、酰胺化、脱酰胺化、酯化、酶解等产生。因此,本申请还涉及所述化合物的代谢物,包括通过使所述化合物与哺乳动物 接触足以产生其代谢产物的时间的方法制得的化合物。
本申请进一步提供所述化合物的前药,其为自身可具有较小药理学活性或无药理学活性的所述化合物的某些衍生物,当被给药至身体中或其上时可通过例如水解裂解转化成具有期望活性的所述化合物。通常这样的前药会是所述化合物的官能团衍生物,其易于在体内转化成期望的治疗活性化合物。关于前药的使用的其他信息可参见“Pro-drugs as Novel Delivery Systems”,第14卷,ACS Symposium Series(T.Higuchi及V.Stella)及“Bioreversible Carriers in Drug Design,”Pergamon Press,1987(E.B.Roche编辑,American Pharmaceutical Association)。所述前药可例如通过用本领域技术人员已知作为“前-部分(pro-moiety)(例如“Design of Prodrugs”,H.Bundgaard(Elsevier,1985)中所述)”的某些部分替代所述化合物中存在的适当官能团来制备。
本申请还提供含有保护基的所述化合物。在制备所述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,由此形成所述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Protective Groups in Organic Chemistry,ed.J.F.W.McOmie,Plenum Press,1973;和T.W.Greene&P.G.M.Wuts,Protective Groups in Organic Synthesis,John Wiley&Sons,1991中所述的那些保护基,这些参考文献通过援引加入本文。使用本领域已知的方法,可以在适当的后续阶段移除保护基。
术语“约”是指在所述数值的±10%范围内,优选±5%范围内,更优选±2%范围内。
术语“偶联物”是指小分子药物与靶向部分连接得到的物质。在本申请的部分实施方案中,小分子药物与靶向部分通过连接体相连。所述连接体在特定环境(例如胞内低pH值环境、偏酸性的肿瘤微环境)中或特定作用(例如溶酶体蛋白酶的作用)下能够断裂,从而使小分子药物与靶向部分分离。
术语“连接体”是指将小分子药物与靶向部分连接起来的片段。
术语“靶向部分”指偶联物中能够与细胞表面的靶标(或靶标的部分)特异性结合的部分。通过靶向部分与靶标的相互作用,偶联物可以被递送至特定的细胞群。
化合物
在第一个方面,本申请提供一种式(I)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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或 前药:
其中,
L1选自O、S和NR5
L2为共价键,或选自C1-6亚烷基和-C1-6亚烷基-O-;
R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-6烷基、C1-6烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R2选自氢、C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R3选自氢、卤素、C1-6烷基和C1-6烷氧基;
R4选自氢、卤素、C1-6烷基和C1-6烷氧基;
R5选自氢和C1-6烷基;
R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基的基团所取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
R8选自氢和C1-6烷基。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-3烷基、C1-3烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基的基团所取代。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-3烷基、C1-3烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R6为C1-6烷基。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、羟基、甲基、甲氧基和C1-3烷基-C(O)-NH-的基团所取代;
在一些实施方案中,R1为C1-6烷基,所述烷基未被取代或者被甲基、羟基、甲氧基或C1-3烷基-C(O)-NH-取代。
在一些实施方案中,R1为C1-6烷基。
在一些实施方案中,R1为正丁基。
在一些实施方案中,R2选自氢、C1-3烷基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R8选自氢和C1-6烷基。
在一些实施方案中,R2选自氢、C1-3烷基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基、R7和R7-L3-的基团所 取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R8选自氢和C1-6烷基。
在一些实施方案中,R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
R7选自C1-3烷基、C3-6环烷基和3-6元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基和羧基的基团所取代;
L3选自C1-3亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R8选自氢和C1-3烷基。
在一些实施方案中,R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、C1-3烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
R7选自C1-3烷基和3-6元杂环基,所述烷基和杂环基任选地被一个或多个选自氢和C1-3烷基的基团所取代;
L3选自C1-3亚烷基和-C1-3亚烷基-NR8-;
R8为C1-3烷基。
在一些实施方案中,R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、C1-3烷基、C1-3烷氧基、-(CH2)N(CH3)2、-(CH2CH2)N(CH3)2、6元含氮杂环基和6元含氮杂环基-亚甲基-的基团所取代,所述6元含氮杂环基任选地被一个或多个氢和甲基所取代。
在一些实施方案中,L1选自O、S和NH。
在一些实施方案中,L1为NH。
在一些实施方案中,L2为共价键,或选自C1-3亚烷基和-C1-3亚烷基-O-。
在一些实施方案中,L2为亚甲基。
在一些实施方案中,R3选自氢、卤素、C1-3烷基和C1-3烷氧基。
在一些实施方案中,R3为氢。
在一些实施方案中,R4选自氢、卤素、C1-3烷基和C1-3烷氧基。
在一些实施方案中,R4为氢。
本发明涵盖对上述优选基团进行任意组合所得的式(I)的化合物。
在一些实施方案中,所述化合物具有式(II-A)的结构:
其中,各基团R1和R2如上文所定义。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-6烷基、C1-6烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R2选自氢、C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基的基团所取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
R8选自氢和C1-6烷基。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-3烷基、C1-3烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基和C1-6烷氧基的基团所取代;
R2选自氢、C1-3烷基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R8选自氢和C1-6烷基。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-3烷基、C1-3烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
R6为C1-6烷基;
R2选自氢、C1-3烷基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R8选自氢和C1-6烷基。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、羟基、甲基、甲氧基和C1-3烷基-C(O)-NH-的基团所取代;
R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
R7选自C1-3烷基、C3-6环烷基和3-6元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基和羧基的基团所取代;
L3选自C1-3亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
R8选自氢和C1-3烷基。
在一些实施方案中,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、羟基、甲基、甲氧基和C1-3烷基-C(O)-NH-的基团所取代;
R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-6元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、甲基、甲氧基、-(CH2)N(CH3)2、-(CH2CH2)N(CH3)2、6元含氮杂环基和6元含氮杂环基-亚甲基-的基团所取代,所述6元含氮杂环基任选地被一个或多个氢和甲基所取代。
在一些实施方案中,R1选自正丁基、 R2选自氢、
在一些实施方案中,R1选自正丁基、 R2选自氢、
在一些实施方案中,R1选自正丁基、R2选自氢、
在一些实施方案中,所述化合物选自:

在另一个方面,本申请提供下式所述药物-连接体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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
D-L’
其中,D为前文任一项所述的化合物的片段;
L’为连接体片段。
在一些实施方案中,L’选自-L4-L5-L6,其中,
L4选自化学键、-C(=O)-和-NH-;
L5为聚乙二醇片段,例如其中a选自1-10的整数;
L6其中,
Z1选自化学键和-C(=O)-;
Z2选自化学键和C1-20亚烷基;
Z3选自化学键、C2-6亚烯基和C2-6亚炔基;
A选自
LG为亲核取代反应的离去基团,选自卤素、硝基、苯磺酸酯、对甲苯磺酸酯、三氟甲烷磺酸酯、-S(O)2-R9和-S(O)-R9
R9选自选自C1-6烷基和C1-6卤代烷基。
在一些实施方案中,L6其中,
Z1选自化学键和-C(=O)-;
Z2选自化学键和C1-10亚烷基;
Z3选自化学键、C2-4亚烯基和C2-4亚炔基;
A选自
LG选自卤素、硝基、苯磺酸酯、对甲苯磺酸酯、三氟甲烷磺酸酯、-S(O)2-R9和-S(O)-R9
R9选自选自C1-6烷基和C1-6卤代烷基。
在一些实施方案中,L6其中,
Z1为-C(=O)-;
Z2为C1-6亚烷基;
Z3为化学键或
A选自
LG为甲磺酰基。
在一些实施方案中,L6
在一些实施方案中,L’选自:
在一些实施方案中,所述药物-连接体选自以下结构:

前文所述的D-L’通过连接体L’与靶向肿瘤的抗体连接得到本申请所述免疫刺激抗体偶联物。因此,在另一个方面,本申请还提供式(ISAC-I)的免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
其中,
D为前文任一项所述的化合物的片段;
Ab是可靶向目标抗原的抗体;例如靶向肿瘤细胞的抗体,例如Trastuzumab单抗;
L为前文任一项所述的L’与Ab连接后得到的片段;
z选自1-10,例如1、2、3、4、5、6、7、8、9或10。
在一些实施方案中,z选自1-8,例如1-2、1-3、1-4、1-5、1-6、1-7、1-8、2-3、2-4、2-5、2-6、2-7、2-8、3-4、3-5、3-6、3-7、3-8、4-5、4-6、4-7、4-8、5-6、5-7、5-8、6-7、6-8或7-8。
在一些实施方案中,L选自以下结构:

在一些实施方案中,所述免疫刺激抗体偶联物选自:
z独立地选自1-8,例如1-2、1-3、1-4、1-5、1-6、1-7、1-8、2-3、2-4、2-5、2-6、2-7、2-8、3-4、3-5、3-6、3-7、3-8、4-5、4-6、4-7、4-8、5-6、5-7、5-8、6-7、6-8或7-8。
在另一个方面,本申请提供一种组合物,其包含前文任一项所述的免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药。
所述组合物可包含多个本文所述的免疫刺激抗体偶联物,其中每个免疫刺激抗体 偶联物包含本文所述的D-L,其中z独立地为1、2、3、4、5、6、7、8、9或10。换言之,所述组合物中的每个抗体分子可以偶联到1、2、3、4、5、6、7、8、9或10个D-L上。因此,所述组合物的特征可以是范围为约1至约10的“药物抗体”比(DAR)。确定DAR的方法为本领域技术人员所熟知,包括使用反相色谱或HPLC-MS的方法。例如,在一些实施方案中,所述组合物DAR值为选自1-8的整数或小数,例如1-2、1-3、1-4、1-5、1-6、1-7、1-8、2-3、2-4、2-5、2-6、2-7、2-8、3-4、3-5、3-6、3-7、3-8、4-5、4-6、4-7、4-8、5-6、5-7、5-8、6-7、6-8或7-8。又例如,在一些实施方案中,所述药物组合物DAR值为7.0-8.0,例如7.1、7.2、7.3、7.4、7.5、7.6、7.7、7.8、7.9或8.0。
制备方法
在另一个方面,本申请提供制备第一方面任一项所述化合物的方法。特别地,本申请提供了制备式(II-A)的化合物的方法,其包括以下步骤:
其中,R1和R2如上文所定义。
(1)使化合物II-A-1与化合物II-A-2反应以得到化合物II-A-3;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、N-甲基吡咯烷酮、1,4-二氧六环、四氢呋喃及其任意组合,优选四氢呋喃。所述反应优选在适合的有机碱存在下进行,所述有机碱可选自N,N-二异丙基乙胺、三乙胺、4-二甲氨基吡啶及其任意组合,优选N,N-二异丙基乙胺。所述反应优选在适合的温度下进行,所述温度优选为-10-80℃。所述反应优选进行合适的时间,例如2-24小时。
(2)使化合物II-A-3与N,N-二甲基甲酰胺二甲基缩醛进行反应以得到化合物II-A-4;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、N-甲基吡咯烷酮及其任意组合,优选N,N-二甲基甲酰胺。所述反应优选在适合的温度下进行,所述温度优选为50-180℃。所述反应优选进行合适的时间,例如2-12小时。
(3)使化合物II-A-4与化合物II-A-5进行关环反应以得到化合物II-A-6;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自甲醇、乙醇、异丙醇、正丁醇、四氢呋喃及其任意组合,优选乙醇。所述反应优选在适合的温度下进行,所述温度优选为20-100℃。所述反应优选进行合适的时间,例如2-24小时。
(4)使化合物II-A-6进行氧化反应以得到化合物II-A-7;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自二氯甲烷、氯仿、四氢呋喃、乙酸乙酯及其任意组合,优选二氯甲烷。所述反应优选在适合的氧化剂存在下进行,所述氧化剂可选自间氯过氧苯甲酸、双氧水、叔丁基过氧化氢及其任意组合,优选间氯过氧苯甲酸。所述反应优选在适合的温度下进行,所述温度优选为-10-80℃。所述反应优选进行合适的时间,例如1-12小时。
(5)使化合物II-A-7与氨溶液进行反应以得到化合物II-A;
所述反应优选在适合的溶剂中进行。所述溶剂可选自四氢呋喃、1,4-二氧六环、水及其任意组合,优选1,4-二氧六环。所述反应优选在适合的温度下进行,所述温度优选为50-150℃。所述反应优选进行合适的时间,例如2-24小时。
特别地,本申请还提供了另一种制备式(II-A)的化合物的方法,其包括以下步骤:
其中,R1和R2如上文所定义;
LG表示离去基团,所述离去基团包括但不限于卤素原子、甲磺酰基氧基、对甲基苯磺酰基氧基等。
(1)使化合物II-A-4与氨溶液进行关环反应以得到化合物II-A-8;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自甲醇、乙醇、异丙醇、正丁醇、四氢呋喃及其任意组合,优选甲醇。所述反应优选在适合的温度下进行,所述温度优选为20-100℃。所述反应优选进行合适的时间,例如2-24小时。
(2)使化合物II-A-8进行氧化反应以得到化合物II-A-9;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自二氯甲烷、氯仿、四氢呋喃、乙酸乙酯及其任意组合,优选二氯甲烷。所述反应优选在适合的氧化剂存在下进行,所述氧化剂可选自间氯过氧苯甲酸、双氧水、叔丁基过氧化氢及其任意组合,优选间氯过氧苯甲酸。所述反应优选在适合的温度下进行,所述温度优选为-10-80℃。所述反应优选进行合适的时间,例如1-12小时。
(3)使化合物II-A-9与氨溶液进行反应以得到化合物II-A-10;
所述反应优选在适合的溶剂中进行。所述溶剂可选自四氢呋喃、1,4-二氧六环、水及其任意组合,优选1,4-二氧六环。所述反应优选在适合的温度下进行,所述温度优选为50-150℃。所述反应优选进行合适的时间,例如2-24小时。
(4)使化合物II-A-10与化合物II-A-11进行反应以得到化合物II-A;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自N,N-二甲基甲酰胺、 N,N-二甲基乙酰胺、N-甲基吡咯烷酮、丙酮及其任意组合,优选N,N-二甲基甲酰胺。所述反应优选在适合的有机碱或无机碱存在下进行,所述有机碱或无机碱可选自N,N-二异丙基乙胺、三乙胺、4-二甲氨基吡啶、碳酸钠,碳酸钾、碳酸铯、叔丁醇钾及其任意组合,优选碳酸铯。所述反应优选在适合的温度下进行,所述温度优选为20-100℃。所述反应优选进行合适的时间,例如2-12小时。
上述各反应步骤的具体条件为本领域公知,对此本文不作具体限定。根据本申请的教导结合本领域公知常识,本领域技术人员可以对通式中的各取代基进行选择替换以制备得到不同的化合物,这些选择和替换均在本发明的保护范围之内。
药物组合物和药盒
在另一个方面,本申请提供一种药物组合物,其包含预防或治疗有效量的前文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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、前文任一项所述的免疫刺激抗体偶联物、或者前文任一项所述的组合物,以及一种或多种药学上可接受的载体。
在另一个方面,本申请提供一种药盒,其包含前文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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、前文任一项所述的免疫刺激抗体偶联物、前文任一项所述的组合物、或者前文任一项所述的药物组合物。任选地,所述药盒还包括使用说明。
如本文中所使用的,术语“药学上可接受的载体”是指与治疗剂一同给药的稀释剂、辅剂、赋形剂或媒介物,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、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。
所述药物组合物中可使用的药学上可接受的载体包括但不限于无菌液体。所述药物组合物可以例如是固体制剂、半固体制剂、液体制剂或气态制剂等的形式。
所述药物组合物可以系统地作用和/或局部地作用。为此目的,它们可以适合的途径给药,例如通过注射或经皮给药;或者通过口服或通过吸入给药。
所述化合物在药物组合物中的含量或用量可以是约0.001mg至约1000mg,适合地是0.01-800mg,优选0.05-500mg。
在一些实施方案中,本申请提供制备所述药物组合物的方法,所述方法包括将所述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、溶剂合物、N- 氧化物、同位素标记的化合物、代谢物或前药与一种或多种药学上可接受的载体组合。
治疗方法和用途
本文所述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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、或者所述药物组合物,其对靶点TLR7和/或TLR8具有很强的激动作用。
因此,本申请的另一目的在于提供本文所述化合物在制备免疫刺激抗体偶联物中的用途。在一些实施方案中,所述免疫刺激抗体偶联物包含能够与TLR7和/或TLR8作用的小分子配体。
本申请的另一目的在于提供所述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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、所述免疫刺激抗体偶联物、所述组合物、或所述药物组合物在制备药物中的用途,所述药物用于预防和/或治疗TLR7和/或TLR8介导的相关疾病。
本申请的另一目的在于提供所述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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、所述免疫刺激抗体偶联物、所述组合物、或所述药物组合物,其用于预防和/或治疗TLR7和/或TLR8介导的相关疾病。
本申请的另一目的在于提供预防和/或治疗TLR7和/或TLR8介导的相关疾病的方法,其包括向有此需要的个体给予预防或治疗有效量的所述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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、所述免疫刺激抗体偶联物、所述组合物或药物组合物。
在一些实施方案中,所述TLR7和/或TLR8介导相关疾病优选地为肿瘤。
如本文中所使用的术语“有效量”是指足以实现所需预防或治疗效果的量,例如,实现减轻与待治疗疾病相关的一或多种症状的量。
可调整给药方案以提供最佳所需响应。例如,可给药单次推注,可随时间给药数个分剂量,或可如治疗情况的急需所表明而按比例减少或增加剂量。要注意,剂量值可随要减轻的病况的类型及严重性而变化,且可包括单次或多次剂量。要进一步理解,对于任何特定个体,具体的给药方案应根据个体需要及给药所述化合物或监督所述化合物的给药的人员的专业判断来随时间调整。
所给药的所述化合物的量会取决于所治疗的个体、病症或病况的严重性、给药的速率、化合物的处置及处方医师的判断。在一些情况下,不高于前述范围的下限的剂量水平可以是足够的,而在其它情况下,仍可在不引起任何有害副作用的情况下采用较大剂量,条件是首先将所述较大剂量分成数个较小剂量以在一整天中给药。
除非另外说明,否则如本文中所使用,术语“治疗”意指逆转、减轻、改善这样的术语所应用的病症或病况或者这样的病症或病况的一或多种症状的进展。
术语“预防”指抑制和延迟疾病的发作,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。
如本文所使用的“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本文中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)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)。
本文中所使用的当其连接在双键任意一端时,表示该双键构型为顺式或反式构型。当其以的形式出现时,表示位置的键为连接键。
具体实施方式
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。所用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。
化合物的结构是通过核磁共振(1H NMR)或质谱(MS)来确定的。1H NMR的测定仪器为JEOL Eclipse 400核磁仪,测定溶剂为氘代甲醇(CD3OD)、氘代氯仿(CDCl3)或六氘代二甲基亚砜(DMSO-d6),内标为四甲基硅烷(TMS),化学位移(δ)以百万分之一(ppm)的单位给出。
MS的测定仪器为Agilent(ESI)质谱仪,生产商:Agilent,型号:Agilent 6120B。
制备高效液相色谱仪的制备方法:
仪器型号:Agilent 1260,色谱柱:Waters SunFire Prep C18 OBD(19mm×150mm×5.0μm);色谱柱温:25℃;流速:20.0mL/min;检测波长:214nm;洗脱梯度: (0min:10%A,90%B;16.0min:90%A,10%B);流动相A:乙腈;流动相B:0.05%甲酸水溶液。
薄层色谱硅胶板(TLC)使用Merck产的铝板(20×20cm),薄层层析分离纯化采用的规格是烟台产GF 254(1mm)。
反应的监测采用薄层色谱法(TLC)或LC-MS;使用的展开剂体系包括:二氯甲烷和甲醇体系,正己烷和乙酸乙酯体系,以及石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节或者加入三乙胺等进行调节。
微波反应使用Biotage Initiator+(400W,RT~300℃)微波反应器。
柱层析一般使用200~300目硅胶为载体。洗脱剂体系包括:二氯甲烷和甲醇体系,以及石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺进行调节。
反相柱层析的制备方法:
仪器型号:Biotage快速中压制备色谱,色谱柱:Agela C18反相柱(Spherical;20-35μm;100A);色谱柱温:25℃;流速:28.0mL/min;检测波长:220nm;流动相A:乙腈;流动相B:水。
实施例中无特殊说明,反应的温度为室温(20℃~35℃);
本文所使用的试剂购自Acros Organics、Aldrich Chemical Company、特伯化学等公司。
在常规的合成法以及实施例、和中间体合成例中,各缩写的意思如以下所示。

一、化合物实施例
中间体制备例1:2-氨基-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
第一步:4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
将6-氯-4-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯(12g,48.15mmol)、N,N-二异丙基乙胺(12.6g,96.31mmol)和正丁胺(5.34g,72.23mmol)溶于四氢呋喃(200mL)中,室温搅拌12小时。向反应体系中加入水(1L),乙酸乙酯(200mL)萃取三次,合并有机相,依次用稀盐酸水溶液(1N,100mL)和饱和食盐水洗涤有机相,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(13.3g,收率:97.4%)。
MS m/z(ESI):284.2[M+H]+
第二步:4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
将4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯(11g,38.8mmol)和N,N-二甲基甲酰胺二甲基缩醛(23g,194mmol)溶于N,N-二甲基甲酰胺(100mL)中,升温至150℃搅拌4小时。将反应体系冷却至室温后,向反应体系中加入水(500mL),二氯甲烷(200mL)萃取三次,合并有机相,饱和食盐水洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(11g,收率:83.8%)。
MS m/z(ESI):339.2[M+H]+
第三步:4-(丁基氨基)-2-(甲硫基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯(11g,32.5mmol)、氨甲醇溶液(21mL,7M)和乙醇(100mL)加入高压釜中,升温至80℃搅拌12小时。将反应体系浓缩干,所得剩余物经甲基叔丁基醚打浆纯化后,得到标题化合物(7.8g,收率:90.5%)。
MS m/z(ESI):265.1[M+H]+
第四步:4-(丁基氨基)-2-(甲基亚磺酰基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将4-(丁基氨基)-2-(甲硫基)吡啶并[4,3-d]嘧啶-5(6H)-酮(7.8g,29.3mmol)溶于二氯甲烷(80mL)中,冰水浴降温,0℃下向反应液中缓慢分批加入间氯过氧苯甲酸(8.95g,44.2mmol),缓慢升温至室温搅拌4小时。将反应液倒入水(200mL)中,并用二氯甲烷(100mL)萃取三次,合并有机相,饱和碳酸氢钠水溶液(100mL)洗涤两次,饱和亚硫酸钠水溶液(100mL)洗涤两次,饱和食盐水(50mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(8.3g,收率:100%)。
MS m/z(ESI):281.0[M+H]+
第五步:4-(丁基氨基)-2-((2,4-二甲氧基苄基)氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将4-(丁基氨基)-2-(甲基亚磺酰基)吡啶并[4,3-d]嘧啶-5(6H)-酮(8.3g,29.3mmol)和2,4-二甲氧基苄胺(5.9g,35.2mmol)溶于N,N-二甲基甲酰胺(100mL)中,升温至80℃搅拌12小时。将反应体系冷却至室温后,向反应体系中加入水(500mL),二氯甲烷(100mL) 萃取三次,合并有机相,饱和食盐水洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物粗品(12g,收率:100%)。
MS m/z(ESI):384.2[M+H]+
第六步:2-氨基-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将4-(丁基氨基)-2-((2,4-二甲氧基苄基)氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮(12g,29.3mmol)溶于三氟乙酸(60mL)中,室温搅拌12小时。将反应体系浓缩干后,向反应体系中缓慢加入饱和碳酸氢钠水溶液(100mL),大量固体析出,过滤并用水洗涤滤饼。滤饼干燥后经甲基叔丁基醚打浆纯化,得到标题化合物(5.6g,收率:81.7%)。
MS m/z(ESI):234.2[M+H]+
中间体制备例2:(6-(氯甲基)吡啶-3-基)甲醇的制备
将6-(氯甲基)烟酸甲酯(1g,5.39mmol)溶于干燥四氢呋喃(3mL)中,降温至0℃,滴加氢化铝锂(408.93mg,10.78mmol)的四氢呋喃(3mL)溶液,0℃搅拌0.5小时。向反应体系中加冰水(2mL)和氢氧化钠水溶液(15%,0.5mL),二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(30mL)洗涤2次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(200mg,收率:23.6%)。
MS m/z(ESI):158.0[M+H]+
实施例1:2-氨基-4-(丁基氨基)-6-甲基-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(化合物1)的制备
第一步:4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
将6-氯-4-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯(1.0g,4.05mmol)、N,N-二异丙基乙胺(0.78g,6.02mmol)和正丁胺(0.44g,6.02mmol)溶于四氢呋喃(20mL)中,室温搅拌12小时。向反应体系中加入水(150mL),乙酸乙酯(60mL)萃取三次,合并有机相,依次用稀盐酸水溶液(1N,15mL)和饱和食盐水(15mL)洗涤有机相,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(1.1g,收率:95.8%)。
MS m/z(ESI):284.2[M+H]+
第二步:4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
将4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯(1.1g,3.88mmol)和N,N-二甲基甲酰胺二甲基缩醛(4.62g,38.8mmol)溶于N,N-二甲基甲酰胺(10mL)中,升温至150℃搅拌4小时。将反应体系冷却至室温后,向反应体系中加入水(100mL),二氯甲烷(60mL)萃取三次,合并有机相,饱和食盐水(45mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(1.1g,收率:83.8%)。
MS m/z(ESI):339.2[M+H]+
第三步:4-(丁基氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮的制备
将4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯(0.5g,1.48mmol)和30%甲胺乙醇溶液(4mL)溶于乙醇(10mL)中,升温至80℃搅拌12小时。将反应体系冷却至室温后,向反应体系中加入水(100mL),二氯甲烷(60mL)萃取三次,合并有机相,饱和食盐水(45mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经甲基叔丁基醚(5mL)打浆纯化后,得到标题化合物(0.25g,收率:60.7%)。
MS m/z(ESI):279.1[M+H]+
第四步:4-(丁基氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮的制备
将4-(丁基氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(0.1g,0.36mmol)溶于二氯甲烷(6mL)中,冰水浴降温,0℃下向反应液中缓慢分批加入间氯过氧苯甲酸(93.0mg,0.54mmol),缓慢升温至室温搅拌4小时。将反应液倒入水(50mL)中,并用二氯甲烷(30mL)萃取三次,合并有机相,饱和碳酸氢钠水溶液(20mL)洗涤两次,饱和亚硫酸钠水溶液(20mL)洗涤两次,饱和食盐水(30mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物(0.1g,收率:94.4%)。
MS m/z(ESI):295.1[M+H]+
第五步:2-氨基-4-(丁基氨基)-6-甲基-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(化合物1)的制备
将4-(丁基氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(0.1g,0.34mmol)溶于氨的1,4-二氧六环溶液(0.5N,5mL)中,升温至100℃搅拌12小时。将反应体系冷却至室温后,向反应体系中加入水(60mL),二氯甲烷(40mL)萃取三次,合并有机相,饱和食盐水(30mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(20mg,收率:23.8%)。
MS m/z(ESI):248.1[M+H]+
1H NMR(400MHz,CD3OD):δ7.49(d,J=7.6Hz,1H),6.15(d,J=7.6Hz,1H),3.53-3.47(m,2H),3.46(s,3H),1.65-1.61(m,2H),1.48-1.42(m,2H),0.98(t,J=7.6Hz,3H)。
实施例2:2-氨基-4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(化合物2)的制备
第一步:4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
采用实施例1的合成路线,将第一步反应原料正丁胺替换为2-氨基-1,3-二甲氧基丙烷,得到标题化合物(1.4g,收率:89.3%)。
MS m/z(ESI):330.1[M+H]+
第二步:4-((1,3-二甲氧基丙烷-2-基)氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
采用实施例1的合成路线,将第二步反应原料4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯替换为4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯,得到标题化合物(1.2g,收率:80.9%)。
MS m/z(ESI):385.1[M+H]+
第三步:4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮的制备
采用实施例1的合成路线,将第三步反应原料4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯替换为4-((1,3-二甲氧基丙烷-2-基)氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯,得到标题化合物(0.7g,收率:65.4%)。
MS m/z(ESI):325.1[M+H]+
第四步:4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮的制备
采用实施例1的合成路线,将第四步反应原料4-(丁基氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮替换为4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮,得到标题化合物(45mg,收率:47.8%)。
MS m/z(ESI):341.1[M+H]+
第五步:2-氨基-4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(化合物2)的制备
采用实施例1的合成路线,将第五步反应原料4-(丁基氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮替换为4-((1,3-二甲氧基丙烷-2-基)氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮,得到标题化合物(6mg,收率:20.1%)。
MS m/z(ESI):294.2[M+H]+
1H NMR(400MHz,CD3OD):δ7.54(d,J=7.6Hz,1H),6.18(d,J=7.6Hz,1H),3.63-3.50(m,5H),3.46(s,3H),3.39(s,6H)。
实施例3:2-氨基-4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(化合物3)的制备
第一步:4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
采用实施例1的合成路线,将第一步反应原料正丁胺替换为2-氨基-1,3-丙二醇,得到标题化合物(0.54g,收率:83.6%)。
MS m/z(ESI):302.0[M+H]+
第二步:4-((1,3-二羟基丙烷-2-基)氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯的制备
采用实施例1的合成路线,将第二步反应原料4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯替换为4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯,得到标题化合物(0.75g,收率:82.3%)。
MS m/z(ESI):357.1[M+H]+
第三步:4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮的制备
采用实施例1的合成路线,将第三步反应原料4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯替换为4-((1,3-二羟基丙烷-2-基)氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯,得到标题化合物(0.14g,收率:58.8%)。
MS m/z(ESI):297.0[M+H]+
第四步:4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮的制备
采用实施例1的合成路线,将第四步反应原料4-(丁基氨基)-6-甲基-2-(甲硫基)-5,6- 二氢吡啶[4,3-d]嘧啶-5-酮替换为4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮,得到标题化合物(30mg,收率:44.5%)。
MS m/z(ESI):313.0[M+H]+
第五步:2-氨基-4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-5,6-二氢吡啶[4,3-d]嘧啶-5-酮(化合物3)的制备
采用实施例1的合成路线,将第五步反应原料4-(丁基氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮替换为4-((1,3-二羟基丙烷-2-基)氨基)-6-甲基-2-(甲基亚磺酰基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮,得到标题化合物(5mg,收率:26.6%)。
MS m/z(ESI):266.1[M+H]+
1H NMR(400MHz,DMSO-d6):δ9.61-9.59(m,1H),7.57(d,J=7.6Hz,1H),6.53(s,2H),6.01(d,J=7.6Hz,1H),4.79(s,2H),4.18-4.16(m,1H),3.59-3.47(m,4H),3.41(s,3H)。
实施例4:2-氨基-4-丁基氨基)-6-(4-((二甲基氨基)甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物4)的制备
将2-氨基-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮(18.4mg,0.079mmol)、碳酸铯(39.1mg,0.12mmol)和1-(4-(氯甲基)苯基)-N,N-二甲基甲胺(16.5mg,0.09mmol)加入到N,N-二甲基甲酰胺(2mL)中,升温至50℃搅拌8小时。将反应体系冷却至室温后,向反应体系中加入水(60mL),乙酸乙酯(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(12.2mg,收率:40.6%)。
MS m/z(ESI):381.1[M+H]+
1H NMR(400MHz,CD3OD):δ7.57(d,J=7.6Hz,1H),7.33-7.28(m,4H),6.18(d,J=7.6Hz,1H),5.10(s,2H),3.52-3.49(m,4H),2.27(s,6H),1.68-1.59(m,2H),1.47-1.42(m, 2H),0.98(t,J=7.6Hz,3H)。
实施例5:2-氨基-4-(丁基氨基)-6-(4-((二甲基氨基)甲基)-2-甲氧基苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物5)的制备
第一步:4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)-3-甲氧基苯甲酸甲酯的制备
将2-氨基-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮(200mg,0.85mmol)、碳酸钾(240mg,1.7mmol)、碘化钾(13mg,0.08mmol)和4-(溴甲基)-3-甲氧基苯甲酸甲酯(266mg,1.02mmol)加入到N,N-二甲基甲酰胺(6mL)中,升温至60℃搅拌8小时。将反应体系冷却至室温后,向反应体系中加入水(50mL),二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=10/1,体积比),得到标题化合物(150mg,收率:42.5%)。
MS m/z(ESI):412.2[M+H]+
第二步:2-氨基-4-(丁基氨基)-6-(4-(羟甲基)-2-甲氧基苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)-3-甲氧基苯甲酸甲酯(115mg,0.28mmol)加入四氢呋喃(3mL)和无水乙醇(3mL)中,再加入硼氢化钠(32mg,0.85mmol)和无水氯化钙(31mg,0.28mmol),升温至60℃搅拌6小时。将反应体系冷却至室温后,向反应体系中加入饱和氯化铵水溶液(50mL),二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=10/1,体积比),得到标题化合物(60mg,收率:55.7%)。
MS m/z(ESI):384.2[M+H]+
第三步:4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)-3-甲氧基苄基甲磺酸酯的制备
将2-氨基-4-(丁基氨基)-6-(4-(羟甲基)-2-甲氧基苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(60mg,0.16mmol)和三乙胺(32mg,0.32mmol)依次加入到二氯甲烷(3mL)中,冰水浴冷却下滴加甲烷磺酰氯(24mg,0.21mmol),反应液于室温搅拌12小时。向反应体系中加入饱和氯化铵水溶液(50mL),二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物粗品(70mg,收率:94.5%)。
MS m/z(ESI):462.2[M+H]+
第四步:2-氨基-4-(丁基氨基)-6-(4-((二甲基氨基)甲基)-2-甲氧基苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物5)的制备
将4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)-3-甲氧基苄基甲磺酸酯(70mg,0.15mmol)、碳酸铯(100mg,0.30mmol)和二甲胺盐酸盐(18mg,0.23mmol)加入到N,N-二甲基甲酰胺(2mL)中,升温至50℃搅拌8小时。将反应体系冷却至室温后,向反应体系中加入水(60mL),乙酸乙酯(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(10mg,收率:16.1%)。
MS m/z(ESI):411.2[M+H]+
1H NMR(400MHz,CD3OD)δ7.67(d,J=8.0Hz,1H),7.26(d,J=8.0Hz,1H),7.12(s,1H),7.00(d,J=8.0Hz,1H),6.21(d,J=8.0Hz,1H),5.08(s,2H),4.12(s,2H),3.92(s,3H),3.56-3.47(m,2H),2.71(s,6H),1.66-1.59(m,2H),1.46-1.38(m,2H),0.96(t,J=8.0Hz,3H)。
实施例6:2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物6)的制备
第一步:4-(4-(氯甲基)苄基)哌嗪-1-羧酸叔丁酯的制备
将1,4-双(氯甲基)苯(1g,5.75mmol)、N,N-二异丙基乙胺(1.1g,8.62mmol)和哌嗪-1-羧酸叔丁酯(1.07g,5.75mmol)溶于N,N-二甲基甲酰胺(10mL)中,缓慢升温至60℃搅拌6小时。将反应液倒入水(100mL)中,并用二氯甲烷(50mL)萃取三次,合并有机相,饱和食盐水洗涤三次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=1/1,体积比),得到标题化合物(1g,收率:53.8%)。
MS m/z(ESI):325.2[M+H]+
第二步:4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-羧酸叔丁酯的制备
采用实施例5第一步的合成方法,将反应原料4-(溴甲基)-3-甲氧基苯甲酸甲酯替换为4-(4-(氯甲基)苄基)哌嗪-1-羧酸叔丁酯,得到标题化合物(0.3g,收率:58.6%)。
MS m/z(ESI):522.3[M+H]+
第三步:2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物6)的制备
将4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-羧酸叔丁酯(300mg,0.57mmol)溶于甲醇(4mL)和氯化氢的1,4-二氧六环溶液(4N,2mL)的混合溶剂中,室温搅拌12小时。反应完全后,反应液浓缩,得到的剩余物经反相柱层析纯化(洗脱梯度:A%=5%-15%),得到标题化合物(0.21g,收率:87.5%)。
MS m/z(ESI):422.3[M+H]+
1H NMR(400MHz,CD3OD):δ7.82-7.79(m,1H),7.35-7.31(m,4H),6.30-6.27(m,1H),5.14(s,2H),3.59-3.53(m,4H),3.19-3.17(m,4H),2.66-2.65(m,4H),1.66-1.63(m,2H),1.46-1.43(m,2H),0.96(t,J=8.0Hz,3H)。
实施例7:2-氨基-4-(丁基氨基)-6-(4-((4-甲基哌嗪-1-基)甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物7)的制备
将2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(50mg,0.12mmol)和甲醛(30%水溶液,15mg,0.15mmol)依次加入甲醇(4mL)中,然后加入氰基硼氢化钠(22mg,0.36mmol)。反应体系在室温下搅拌2小时。将反应液倒入水(50mL)中,并用二氯甲烷(20mL)萃取三次,合并有机相,饱和食盐水(15mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(10mg,收率:19.4%)。
MS m/z(ESI):436.3[M+H]+
1H NMR(400MHz,CD3OD):δ7.63(d,J=8.0Hz,1H),7.34-7.29(m,4H),6.20(d,J=8.0Hz,1H),5.11(s,2H),3.60(s,2H),3.53-3.49(m,2H),3.12-2.64(m,4H),2.60-2.48(m,7H),1.66-1.63(m,2H),1.47-1.43(m,2H),0.96(t,J=8.0Hz,3H)。
实施例8:2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物8)的制备
第一步:2-氨基-6-(4-溴苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用实施例5第一步的合成方法,将反应原料4-(溴甲基)-3-甲氧基苯甲酸甲酯替换为1-溴-4-(溴甲基)苯,得到标题化合物(250mg,收率:58.2%)。
MS m/z(ESI):402.1[M+H]+
第二步:4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苯基)哌嗪-1-羧酸叔丁酯的制备
将2-氨基-6-(4-溴苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮(50mg,0.12mmol)、哌嗪-1-羧酸叔丁酯(46mg,0.24mmol)、2-二环己基磷-2',6'-二异丙氧基-1,1'-联苯(11mg,0.02mmol)和叔丁醇钠(24mg,0.24mmol)加入到N,N-二甲基甲酰胺(2mL)和甲苯(4mL)的混合溶剂中,氮气置换后加入三(二亚苄基丙酮)二钯(22mg,0.02mmol),升温至110℃搅拌12小时。将反应体系冷却至室温后,向反应体系中加入水(50mL),二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(25mg,收率:39.7%)。
MS m/z(ESI):508.3[M+H]+
第三步:2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物8)的制备
采用实施例6第三步的合成方法,将反应原料4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-羧酸叔丁酯替换为4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苯基)哌嗪-1-羧酸叔丁酯,得到标题化合物(16mg,收率:80%)。
MS m/z(ESI):408.2[M+H]+
1H NMR(400MHz,CD3OD)δ8.02(d,J=8.0Hz,1H),7.33(d,J=8.0Hz,2H),7.01(d,J=8.0Hz,2H),6.39(d,J=8.0Hz,1H),5.13(s,2H),3.65-3.61(m,2H),3.42-3.34(m,8H),1.71-1.63(m,2H),1.47-1.42(m,2H),0.99(t,J=8.0Hz,3H)。
实施例9:2-氨基-4-(丁基氨基)-6-(4-(4-甲基哌嗪-1-基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物9)的制备
采用实施例8第二步的合成方法,将反应原料哌嗪-1-羧酸叔丁酯替换为N-甲基哌嗪,得到标题化合物(6mg,收率:10.8%)。
MS m/z(ESI):422.3[M+H]+
1H NMR(400MHz,CD3OD):δ7.51(d,J=8.0Hz,1H),7.20(d,J=8.0Hz,1H),6.92(d,J=8.0Hz,1H),6.14(d,J=8.0Hz,1H),4.98(s,2H),3.49-3.46(m,2H),3.29-3.16(m,4H),2.67-2.59(m,4H),2.36-2.32(m,5H),1.65-1.61(m,2H),1.50-1.39(m,2H),0.96(d,J=8.0Hz,3H)。
实施例10:2-氨基-4-(丁基氨基)-6-((5-((二甲基氨基)甲基)吡啶-2-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物11)的制备
第一步:2-氨基-4-(丁基氨基)-6-((5-(羟甲基)吡啶-2-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将2-氨基-4-(丁基氨基)-吡啶并[4,3-d]嘧啶-5(6H)-酮(200mg,0.86mmol)、碳酸铯(560.73mg,1.7mmol)和(6-(氯甲基)吡啶-3-基)甲醇(148.63mg,0.63mmol)加入到N,N-二甲基甲酰胺(6mL)中,升温至80℃搅拌16小时。将反应体系冷却至室温后,向反应体系中加入水(60mL),乙酸乙酯(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(160mg,收率:53.2%)。
MS m/z(ESI):355.2[M+H]+
第二步:2-氨基-4-(丁基氨基)-6-((5-(氯甲基)吡啶-2-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将2-氨基-4-(丁基氨基)-6-((5-(羟甲基)吡啶-2-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(160.00mg,0.45mmol)溶于二氯甲烷(4mL)中,滴加氯化亚砜(537.10mg,4.51mmol),在室温下搅拌4小时。向反应体系中加入水(60mL),乙酸乙酯(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=10/1,体积比),得到标题化合物(12mg,收率:7.1%)。
MS m/z(ESI):373.1[M+H]+
第三步:2-氨基-4-(丁基氨基)-6-((5-((二甲基氨基)甲基)吡啶-2-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物11)的制备
将2-氨基-4-(丁基氨基)-6-((5-(氯甲基)吡啶-2-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(12mg,32.18μmol)、N,N-二甲基甲胺(3.94mg,48.28μmol)、碘化钠(0.49mg,3.22μmol)和碳酸铯(21.26mg,64.37μmol)加入乙腈(2mL)中,室温反应16小时。浓缩反应液所得剩余物经制备高效液相色谱纯化,得到标题化合物(5mg,收率:40.7%)。
MS m/z(ESI):382.2[M+H]+
1H NMR(400MHz,CD3OD)δ9.49(t,J=6.0Hz,1H),8.44(d,J=2.0Hz,1H),7.76(dd,J=8.0,2.0Hz,1H),7.63(d,J=8.0Hz,1H),7.29(d,J=8.0Hz,1H),6.21(d,J=8.0Hz,1H),5.19(s,2H),3.51-3.45(m,4H),2.24(s,6H),1.66-1.27(m,5H),0.96(t,J=8.0Hz,3H)。
实施例11:2-氨基-4-(丁基氨基)-6-((4-((二甲基氨基)甲基)环己基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物12)的制备
第一步:4-(羟甲基)-N,N-二甲基环己烷-1-甲酰胺的制备
将4-(羟甲基)环己烷-1-甲酸(0.5g,3.13mmol)、N,N,N′,N′-四甲基-O-(7-氮杂苯并三唑-1-基)六氟磷酸脲(1.56g,4.07mmol)、N,N-二异丙基乙胺(1.21g,9.4mmol)和二甲胺盐酸盐(0.38g,4.69mmol)依次加入到N,N-二甲基甲酰胺(8mL)中,反应液于室温搅拌12小时。向反应体系中加入乙酸乙酯(60mL)稀释反应液,依次用1N氢氧化钠溶液和1N盐酸溶液洗涤,再用饱和食盐水洗涤两次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物粗品(0.52g,收率:89.6%)。
MS m/z(ESI):186.1[M+H]+
第二步:(4-((二甲基氨基)甲基)环己基)甲醇的制备
将4-(羟甲基)-N,N-二甲基环己烷-1-甲酰胺(250mg,1.34mmol)溶入四氢呋喃(10mL)中,冰水浴冷却下缓慢加入四氢铝锂(256mg,6.68mmol),反应液升温至50℃搅拌12小时。向反应体系中加入十水硫酸钠固体(0.2g)搅拌20分钟,反应液垫硅藻土过滤并用乙酸乙酯洗涤滤饼,合并有机相,浓缩,得到标题化合物粗品(230mg,收率:100%)。
MS m/z(ESI):172.2[M+H]+
第三步:1-(4-(氯甲基)环己基)-N,N-二甲基甲胺的制备
将(4-((二甲基氨基)甲基)环己基)甲醇(230mg,1.34mmol)溶入二氯甲烷(10mL)中,加入氯化亚砜(0.8g,6.68mmol),反应液升温至45℃并搅拌12小时。反应液直接浓缩,得到标题化合物粗品(200mg,收率:78.7%)。
MS m/z(ESI):190.1[M+H]+
第四步:2-氨基-4-(丁基氨基)-6-((4-((二甲基氨基)甲基)环己基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物12)的制备
采用实施例5第一步的合成方法,将反应原料4-(溴甲基)-3-甲氧基苯甲酸甲酯替换为1-(4-(氯甲基)环己基)-N,N-二甲基甲胺,得到标题化合物(12mg,收率:9.6%)。
MS m/z(ESI):387.3[M+H]+
1H NMR(400MHz,CD3OD):δ7.46(d,J=8.0Hz,1H),6.15(d,J=8.0Hz,1H),3.86(d,J=8.0Hz,2H),3.52-3.47(m,2H),2.47(d,J=8.0Hz,2H),2.40-2.25(m,6H),1.70-1.31(m,14H),0.98(t,J=8.0Hz,3H)。
实施例12:2-氨基-4-(丁基氨基)-6-(哌啶-4-基甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物13)的制备
第一步:4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)哌啶-1-甲酸叔丁酯的制备
采用实施例10第一步的合成方法,将(6-(氯甲基)吡啶-3-基)甲醇替换为4-(氯甲基)哌啶-1-甲酸叔丁酯(551.10mg,2.33mmol),得到标题化合物(833mg,收率:91.2%)。
MS m/z(ESI):431.3[M+H]+
第二步:2-氨基-4-(丁基氨基)-6-(哌啶-4-基甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物13)的制备
将4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)哌啶-1-甲酸叔丁 酯(703mg,1.47mmol)溶于甲醇(10mL)中,加入氯化氢的1,4-二氧六环溶液(4M,11.02mL),在室温下搅拌2小时。浓缩反应液所得剩余物经制备高效液相色谱纯化,得到标题化合物(400mg,收率:74.2%)。
实施例13:2-氨基-4-(丁基氨基)-6-((1-甲基哌啶-4-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物14)的制备
采用实施例10第一步的合成方法,将(6-(氯甲基)吡啶-3-基)甲醇替换为4-(氯甲基)-1-甲基哌啶(69.62mg,0.47mmol),得到标题化合物(3.5mg,收率:2.1%)。
MS m/z(ESI):345.2[M+H]+
1H NMR(400MHz,CD3OD)δ9.73(s,1H),8.54(s,1H),7.62-7.56(m,1H),6.24-6.22(m,1H),4.05-3.85(m,2H),3.58-3.32(m,5H),3.08-2.97(m,1H),2.95-2.72(m,4H),2.49-2.06(m,2H),1.90-1.75(m,3H),1.67-1.60(m 2H),1.49-1.40(m,2H),0.98(t,J=8.0Hz,3H)。
实施例14:2-氨基-4-(丁基氨基)-6-((1-(2-(二甲基氨基)乙基)哌啶-4-基)甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(化合物15)的制备
将2-氨基-4-(丁基氨基)-6-(哌啶-4-基甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮(190mg,466.08μmol)、2-氯-N,N-二甲基乙醇胺(55.16mg,512.69μmol)、碘化钠(6.99mg,46.61μmol)和碳酸铯(303.72mg,932.17μmol)加入N,N-二甲基甲酰胺(4mL)中,升温至80℃搅拌16小时。向反应体系中加入水(30mL),乙酸乙酯(15mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(5mg,收率:2.7%)。
MS m/z(ESI):402.3[M+H]+
1H NMR(400MHz,CD3OD)δ9.61(t,J=6.0Hz,1H),7.48(d,J=8.0Hz,1H),6.15(d,J=8.0Hz,1H),3.79(d,J=8.0Hz,2H),3.53-3.46(m,2H),2.96(d,J=12.0Hz,2H),2.55-2.46(m,4H),2.27(s,6H),2.04-1.99(m,2H),1.89-1.83(m,1H),1.68-1.60(m,4H),1.50-1.28(m,5H),0.98(t,J=6.0Hz,3H)。
实施例15:2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)-6-甲基吡啶酮[4,3-d]嘧啶-5(6H)-酮(化合物16)的制备
第一步:2-氨基-2-甲基己腈的制备
将2-己酮(10g,0.1mol)和氯化铵(15.9g,0.3mol)加入到氨甲醇溶液(100mL,7N)中,缓慢向反应中滴加三甲基氰硅烷(20g,0.2mol),反应体系于室温搅拌72小时。反应液浓缩,再向体系中加入乙酸乙酯(100mL),垫硅藻土过滤,滤液浓缩得到标题化合物粗品(12g,收率:86.6%)。
MS m/z(ESI):127.1[M+H]+
第二步:2-氨基-2-甲基己酸的制备
将2-氨基-2-甲基己腈(5g,39.7mmol)加入到浓盐酸(30mL)中,反应体系升温至80℃搅拌12小时,反应液浓缩得到标题化合物盐酸盐粗品(6.5g,收率:91.7%)。
MS m/z(ESI):146.1[M+H]+
第三步:2-氨基-2-甲基己-1-醇的制备
将2-氨基-2-甲基己酸(2g,10.9mmol)加入到四氢呋喃(20mL)中,冰水浴冷却下,缓慢加入四氢铝锂(2.1g,54.5mmol),反应体系升温至60℃搅拌12小时。向反应体系中加入十水硫酸钠粉末(5g)搅拌20分钟,反应液垫硅藻土过滤并用乙酸乙酯洗涤滤饼,合并有机相,浓缩,得到标题化合物粗品(1.1g,收率:76.9%)。
MS m/z(ESI):132.1[M+H]+
第四步:4-((1-羟基-2-甲基己-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-羧酸乙酯的制备
采用实施例1第一步的合成方法,将反应原料正丁胺替换为2-氨基-2-甲基己-1-醇,得到标题化合物(0.8g,收率:45.6%)。
MS m/z(ESI):342.2[M+H]+
第五步:4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-羧酸乙酯的制备
将4-((1-羟基-2-甲基己-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-羧酸乙酯(0.65g,1.71mmol)、咪唑(0.24g,3.43mmol)和叔丁基二甲基氯硅烷(0.31g,2.06mmol)加入到N,N-二甲基甲酰胺(6mL)中,反应在室温搅拌12小时。向反应体系中加入水(50mL),二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=10/1,体积比),得到标题化合物(0.8g,收率:100%)。
MS m/z(ESI):456.3[M+H]+
第六步:4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-羧酸乙酯的制备
采用实施例1第二步的合成方法,将反应原料4-(丁基氨基)-6-甲基-2-(甲硫基)嘧啶-5-甲酸乙酯替换为4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-甲基-2-(甲硫基)嘧啶-5-羧酸乙酯,得到标题化合物(0.8g,收率:89.8%)。
MS m/z(ESI):511.3[M+H]+
第七步:4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-甲基-2-(甲硫基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用实施例1第三步的合成方法,将反应原料4-(丁基氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-甲酸乙酯替换为4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-(2-(二甲基氨基)乙烯基)-2-(甲硫基)嘧啶-5-羧酸乙酯,得到标题化合物(0.65g,收率:92.8%)。
MS m/z(ESI):451.2[M+H]+
第八步:4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-甲基-2-(甲基亚磺酰基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用实施例1第四步的合成方法,将反应原料4-(丁基氨基)-6-甲基-2-(甲硫基)-5,6-二氢吡啶[4,3-d]嘧啶-5-酮替换为4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-甲基-2-(甲硫基)吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(0.6g,收率:100%)。
MS m/z(ESI):467.2[M+H]+
第九步:4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-2-((2,4-二甲氧基苄基)氨基)-6-甲基吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用中间体制备例1第五步的合成方法,将反应原料4-(丁基氨基)-2-(甲基亚磺酰基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-6-甲基-2-(甲基亚磺酰基)吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(0.5g,收率:78.3%)。
MS m/z(ESI):570.3[M+H]+
第十步:2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)-6-甲基吡啶酮[4,3-d]嘧啶-5-(6H)-酮(化合物16)的制备
采用中间体制备例1第六步的合成方法,将反应原料4-(丁基氨基)-2-((2,4-二甲氧基苄基)氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为4-((1-((叔丁基二甲基甲硅烷基)氧基)-2-甲基己-2-基)氨基)-2-((2,4-二甲氧基苄基)氨基)-6-甲基吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(25mg,收率:39.3%)。
MS m/z(ESI):306.2[M+H]+
1H NMR(400MHz,DMSO-d6):δ9.82(s,1H),7.68(d,J=8.0Hz,1H),6.91(s,2H),6.09(br,1H),5.03(s,1H),3.67(d,J=10.0Hz,1H),3.55(d,J=12.0Hz,1H),3.39(s,3H), 1.92-1.89(m,1H),1.76-1.64(m,1H),1.34(s,3H),1.25-1.18(m,4H),0.86(t,J=6.0Hz,3H)。
实施例16:N-(2-((2-氨基-6-甲基-5-氧代-5,6-二氢吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己基)乙酰胺(化合物18)的制备
第一步:2-((2-氨基-6-甲基-5-氧代-5,6-二氢吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己醛的制备
将2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)-6-甲基吡啶酮[4,3-d]嘧啶-5(6H)-酮(0.5g,1.64mmol)和戴斯-马丁氧化剂(1.04g,2.46mmol)依次加入无水二氯甲烷(10mL)中,反应体系在室温下搅拌12小时。将反应液缓慢加入饱和硫代硫酸钠溶液和饱和碳酸氢钠溶液配置的混合溶液中(20mL,体积比1:1),混合体系室温搅拌0.5小时,并用二氯甲烷(15mL)萃取三次,合并有机相,饱和食盐水(15mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物粗品(0.5g,收率:100%)。
MS m/z(ESI):304.2[M+H]+
第二步:2-氨基-4-((1-((2,4-二甲氧基苄基)氨基)-2-甲基己-2-基)氨基)-6-甲基吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
将2-((2-氨基-6-甲基-5-氧代-5,6-二氢吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己醛(0.45g,1.48mmol)、2,4-二甲氧基苄胺(0.37g,2.23mmol)和钛酸异丙酯(0.64g,2.23mmol)依次加入无水甲醇(10mL)中,室温搅拌2小时,加入氰基硼氢化钠(0.38g,5.92mmol),反应体系在室温下搅拌2小时。将反应液倒入水(50mL)中,并用二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(15mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,得到标题化合物粗品(0.7g,收率:100%)。
MS m/z(ESI):455.3[M+H]+
第三步:2-氨基-4-((1-氨基-2-甲基己-2-基)氨基)-6-甲基吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用中间体制备例1第六步的合成方法,将反应原料4-(丁基氨基)-2-((2,4-二甲氧基苄基)氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为2-氨基-4-((1-((2,4-二甲氧基苄基)氨基)-2-甲基己-2-基)氨基)-6-甲基吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(0.23g,收率:52.3%)。
MS m/z(ESI):305.2[M+H]+
第四步:N-(2-((2-氨基-6-甲基-5-氧代-5,6-二氢吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己基)乙酰胺(化合物18)的制备
将2-氨基-4-((1-氨基-2-甲基己-2-基)氨基)-6-甲基吡啶并[4,3-d]嘧啶-5(6H)-酮(40mg,0.13mmol)和三乙胺(66mg,0.65mmol)加入无水二氯甲烷(4mL)中,冰水浴冷却下加入醋酸酐(26.5mmol,0.26mmol),反应体系在室温下搅拌4小时。将反应液倒入水(50mL)中,并用二氯甲烷(30mL)萃取三次,合并有机相,饱和食盐水(15mL)洗涤三次,有机相经无水硫酸钠干燥、浓缩,所得剩余物经制备高效液相色谱纯化,得到标题化合物(7mg,收率:15.4%)。
MS m/z(ESI):347.2[M+H]+
1H NMR(400MHz,CD3OD):δ7.61(d,J=8.0Hz,1H),6.20(d,J=8.0Hz,1H),3.88(d,J=12.0Hz,1H),3.73(d,J=12.0Hz,1H),3.49(s,3H),2.23-2.10(m,1H),1.94(s,3H),1.61-1.51(m,1H),1.44-1.26(m,7H),0.91(t,J=6.0Hz,3H)。
实施例17:N-(26-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)-6-(2-(甲基磺酰基)嘧啶-5-基)己-5-炔酰胺(化合物20)的制备
第一步:(26-氧代-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯的制备
向(26-羟基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯(1g,19.5mmol)的二氯甲烷(20mL)溶液中加入戴斯-马丁氧化剂(1.23g,29.3mmol),室温搅拌过夜。反应完全后,用二氯甲烷稀释反应。再依次加入饱和硫代硫酸钠溶液(10mL)和碳酸氢钠溶液(10mL)并搅拌20分钟,用二氯甲烷萃取两次,合并有机相,用盐水洗涤,有机相浓缩得到的剩余物用硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=12/1,体积比)得到标题化合物(0.9g,收率:90.0%)。
MS m/z(ESI):512.3[M+H]+
第二步:(26-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯的制备
向2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(90mg,0.21mmol)的二氯乙烷(4mL)溶液中加入(26-氧代-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯(163mg,0.32mmol)、冰醋酸(20mg,0.21mmol)和氰基硼氢化钠(69mg,1.06mmol),反应混合物于室温反应12小时。反应完毕后,用水稀释,二氯甲烷萃取,合并有机相后用盐水洗涤,有机相浓缩得到的剩余物用制备高效液相色谱纯化,得到标题化合物(30mg,收率:18.6%)。
MS m/z(ESI):917.6[M+H]+
第三步:2-氨基-6-(4-((4-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用实施例6第三步的合成方法,将反应原料4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-羧酸叔丁酯替换为(26-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯,得到标题化合物(28mg,收率:100%)。
MS m/z(ESI):817.5[M+H]+
第四步:N-(26-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)-6-(2-(甲基磺酰基)嘧啶-5-基)己-5-炔酰胺的制备
将2-氨基-6-(4-((4-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮(28mg,34.3μmol)、N,N-二异丙基乙胺(14mg,108.3μmol)和2,5-二氧代吡咯烷-1-基-6-(2-(甲基磺酰基)嘧啶-5-基)己基-5-炔酸酯(15mg,41.1μmol)依次加入N,N-二甲基甲酰胺(2mL)中,室温搅拌1小时。反应液经制备高效液相色谱纯化,得到标题化合物(17mg,收率:46.4%)。
MS m/z(ESI):1067.6[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.93(s,1H),10.08(s,1H),9.46(s,1H),9.12(s,2H),8.14(s,1H),7.95(t,J=5.2Hz,1H),7.30(s,4H),6.41(d,J=8.0Hz,1H),5.16(s,2H),3.72(s,2H),3.60-3.39(m,40H),3.20(q,J=6.0Hz,3H),2.87(s,3H),2.67(s,2H),2.35(s,2H),2.26(t,J=8.0Hz,3H),1.82-1.75(m,2H),1.63-1.51(m,2H),1.45-1.30(m,2H),0.91(t,J=6.0Hz,3H)。
实施例18:N-(24-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-24-氧代-3,6,9,12,15,18,21-七氧杂二十四烷基)-6-(2-(甲基磺酰基)嘧啶-5-基)己-5-炔酰胺(化合物21)的制备
第一步:(24-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-24-氧代-3,6,9,12,15,18,21-七氧杂二十四烷基)氨基甲酸叔丁酯的制备
向2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮(60mg,0.14mmol)的N,N-二甲基甲酰胺(4mL)溶液中加入2,2-二甲基-4-氧代-3,8,11,14,17,20,23,26-八氧杂-5-氮杂二十九烷-29-酸(76mg,0.15mmol)、三乙胺(70mg,0.7mmol)和N,N,N′,N′-四甲基-O-(7-氮杂苯并三唑-1-基)六氟磷酸脲(80mg,0.21mmol),反应混合物于室温反应12小时。反应完毕后,用水稀释,二氯甲烷萃取,合并有机相后用盐水洗涤,有机相浓缩得到的剩余物通过制备高效液相色谱纯化,得到标题化合物(80mg,收率:63.4%)。
MS m/z(ESI):901.5[M+H]+
第二步:2-氨基-6-(4-((4-(1-氨基-3,6,9,12,15,18,21-七氧杂二十四烷-24-酰基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用实施例6第三步的合成方法,将反应原料4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-羧酸叔丁酯替换为(24-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-24-氧代-3,6,9,12,15,18,21-七氧杂二十四烷基)氨基甲酸叔丁酯,得到标题化合物(70mg,收率:85.6%)。
MS m/z(ESI):801.5[M+H]+
第三步:N-(24-(4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-基)-24-氧代-3,6,9,12,15,18,21-七氧杂二十四烷基)-6-(2-(甲基磺酰基)嘧啶-5-基)己-5-炔酰胺(化合物21)的制备
采用实施例17第四步的合成方法,将反应原料2-氨基-6-(4-((4-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为2-氨基-6-(4-((4-(1-氨基-3,6,9,12,15,18,21-七氧杂二十四烷-24-酰基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(11mg,收率:15.9%)。
MS m/z(ESI):1051.5[M+H]+
1H NMR(400MHz,CD3OD)δ8.97(s,2H),7.66(d,J=8.0Hz,1H),7.31(q,J=8.0Hz,4H),6.21(d,J=8.0Hz,1H),5.11(s,2H),3.87-3.44(m,39H),3.36(s,6H),2.65-2.55(m,4H),2.50-2.33(m,6H),2.00-1.89(m,2H),1.68-1.60(m,2H),1.49-1.38(m,2H),0.98(t,J=8.0Hz,3H)。
实施例19:N-(26-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)哌啶-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)-6-(2-(甲基磺酰基)嘧啶-5-基)己-5-炔酰胺(化合物22)的制备
第一步:(26-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)哌啶-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯的制备
采用实施例17第二步的合成方法,将反应原料2-氨基-4-(丁基氨基)-6-(4-(哌嗪-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为2-氨基-4-(丁基氨基)-6-(哌啶-4-基甲基)吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(40mg,收率:19.5%)。
MS m/z(ESI):826.5[M+H]+
第二步:2-氨基-6-((1-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌啶-4-基)甲基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮的制备
采用实施例6第三步的合成方法,将反应原料4-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌嗪-1-羧酸叔丁酯替换为(26-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)哌啶-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)氨基甲酸叔丁酯,得到标题化合物(30mg,收率:81.3%)。
MS m/z(ESI):726.5[M+H]+
第三步:N-(26-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)哌啶-1-基)-3,6,9,12,15,18,21,24-八氧杂二十六烷基)-6-(2-(甲基磺酰基)嘧啶-5-基)己-5-炔酰胺(化合物22)的制备
采用实施例17第四步的合成方法,将反应原料2-氨基-6-(4-((4-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为2-氨基-6-((1-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌啶-4-基)甲基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮,得到标题化合物(20mg,收率:37.3%)。
MS m/z(ESI):976.5[M+H]+
1H NMR(400MHz,CD3OD)δ9.72(t,J=4.0Hz,1H),8.98(s,2H),8.50(s,1H),7.58(d,J=8.0Hz,1H),6.21(d,J=8.0Hz,1H),3.90(d,J=8.0Hz,2H),3.83-3.79(m,2H),3.65-3.50(m,36H),3.38-3.34(m,5H),3.29-3.26(m,2H),2.99-2.89(t,J=12.0Hz,2H),2.59(t,J=8.0Hz,2H),2.40(t,J=8.0Hz,2H),2.16(s,1H),1.98-1.86(m,4H),1.67-1.56(m,4H),1.49-1.40(m,2H),0.98(t,J=8.0Hz,3H)。
实施例20:1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)-N-(33-(2-(甲基磺酰基)嘧啶-5-基)-28-氧代-3,6,9,12,15,18,21,24-八氧杂-27-氮杂三十三-32-炔-1-基)哌啶-4-甲酰胺(化合物23)的制备
第一步:1-(4-(氯甲基)苄基)哌啶-4-羧酸甲酯的制备
将1,4-双(氯甲基)苯(500mg,2.86mmol)、哌啶-4-羧酸甲酯(449.86mg,3.14mmol)和碳酸钾(1.18g,8.57mmol)加入乙腈(4mL)中,反应升温至75℃搅拌5小时。向反应液中加入水(30mL),乙酸乙酯(15mL)萃取三次,合并有机相,饱和食盐水(20mL)洗涤两次,有机相经无水硫酸钠干燥、浓缩所得剩余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=10/1,体积比),得到标题化合物(205mg,收率:25.5%)。
MS m/z(ESI):282.1[M+H]+
第二步:1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌啶-4-羧酸甲酯的制备
采用实施例6第一步的合成方法,将(6-(氯甲基)吡啶-3-基)甲醇替换为1-(4-(氯甲基)苄基)哌啶-4-羧酸甲酯,得到标题化合物(172mg,收率:56.5%)。
MS m/z(ESI):479.3[M+H]+
第三步:1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌啶-4-羧酸的制备
将1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌啶-4-羧酸甲酯(100mg,188.05μmol)溶于甲醇(5mL)和水(1mL)中,加入氢氧化钠(112.83mg,2.82mmol),室温搅拌4小时。浓缩反应液,所得剩余物经制备高效液相色谱纯化,得到标题化合物(21mg,收率:24.0%)。
MS m/z(ESI):465.3[M+H]+
第四步:1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)-N-(26-叠氮基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌啶-4-甲酰胺的制备
将1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)哌啶-4-羧酸(21mg,45.20μmol)、26-叠氮基-3,6,9,12,15,18,21,24-八氧杂二十六烷-1-胺(25.77mg,58.77μmol)、N,N-二异丙基乙胺(17.53mg,135.61μmol)和2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(25.78mg,67.81μmol)加入N,N-二甲基甲酰胺(2mL)中,室温搅拌2小时。浓缩反应液,所得剩余物经制备高效液相色谱纯化,得到标题化合物(20mg,收率:50.0%)。
MS m/z(ESI):885.5[M+H]+
第五步:1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)-N-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌啶-4-甲酰胺的制备
将1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)-N-(26-叠氮基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌啶-4-甲酰胺(20mg,22.60μmol)溶于氨水(0.5mL)和四氢呋喃(3mL)中,加入三苯基膦(7.70mg,29.38μmol),反应体系升温至50℃搅拌3小时。浓缩反应液,所得剩余物经制备高效液相色谱纯化,得到标题化合物(10mg,收率:51.5%)。
MS m/z(ESI):859.5[M+H]+
第六步:1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)-N-(33-(2-(甲基磺酰基)嘧啶-5-基)-28-氧代-3,6,9,12,15,18,21,24-八氧杂-27-氮杂三十三-32-炔-1-基)哌啶-4-甲酰胺(化合物23)的制备
采用实施例17第四步的合成方法,将反应原料2-氨基-6-(4-((4-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌嗪-1-基)甲基)苄基)-4-(丁基氨基)吡啶并[4,3-d]嘧啶-5(6H)-酮替换为1-(4-((2-氨基-4-(丁基氨基)-5-氧代吡啶并[4,3-d]嘧啶-6(5H)-基)甲基)苄基)-N-(26-氨基-3,6,9,12,15,18,21,24-八氧杂二十六烷基)哌啶-4-甲酰胺,得到标题化合物(4.1mg,收率:30.5%)。
MS m/z(ESI):1109.6[M+H]+
1H NMR(400MHz,CD3OD)δ9.62(s,1H),8.98(s,2H),8.50(s,1H),7.63(d,J=8.0Hz,1H),7.37(q,J=8.0Hz,4H),6.20(d,J=8.0Hz,1H),5.12(s,2H),3.85(s,2H),3.61-3.59(m,26H),3.54-3.49(m,6H),3.38-3.33(m,7H),3.20-3.11(m,2H),2.61-2.28(m,9H),1.98-1.76(m,7H),1.67-1.59(m,2H),1.49-1.39(m,2H),0.97(t,J=6.0Hz,3H)。
二、免疫刺激抗体偶联物(ISAC)的制备
实施例2-1 ISAC-1
用1M磷酸氢二钠溶液调节Trastuzumab单抗储备液使其pH=6.51,再用pH=6.5-6.6磷酸缓冲盐溶液(PBS)定容至10mg/mL,得Trastuzumab单抗缓冲溶液。向所得Trastuzumab单抗缓冲溶液加入0.1M EDTA,同时加入TCEP(6eq,10mM),摇匀,25℃反应2小时后,向其中加入Trastuzumab单抗12倍摩尔当量的化合物20的DMSO溶液(10mM),摇匀,室温反应2小时,25℃反应2小时,用NAP-10凝胶柱纯化,并用pH=6.0-6.1盐酸组氨酸缓冲溶液洗涤,收集滤液,得到ISAC-1的盐酸组氨酸缓冲液(1.9mg/mL,1.5mL),于4℃储存。
实施例2-2 ISAC-2
用1M磷酸氢二钠溶液调节Trastuzumab单抗储备液使其pH=6.51,再用pH=6.5-6.6磷酸缓冲盐溶液(PBS)定容至10mg/mL,得Trastuzumab单抗缓冲溶液。向所得Trastuzumab单抗缓冲溶液加入0.1M EDTA,同时加入TCEP(6eq,10mM),摇匀,25℃反应2小时后,向其中加入Trastuzumab单抗12倍摩尔当量的化合物21的DMSO溶液(10mM),摇匀,室温反应2小时,25℃反应2小时,用NAP-10凝胶柱纯化,并用pH=6.0-6.1盐酸组氨酸缓冲溶液洗涤,收集滤液,得到ISAC-2的盐酸组氨酸缓冲液(3.0mg/mL,1.5mL),于4℃储存。
实施例2-3 ISAC-3
用1M磷酸氢二钠溶液调节Trastuzumab单抗储备液使其pH=6.51,再用pH=6.5-6.6磷酸缓冲盐溶液(PBS)定容至10mg/mL,得Trastuzumab单抗缓冲溶液。向所得Trastuzumab单抗缓冲溶液加入0.1M EDTA,同时加入TCEP(6eq,10mM),摇匀,25℃反应2小时后,向其中加入Trastuzumab单抗11倍摩尔当量的化合物22的DMSO溶液(10mM),摇匀,室温反应2小时,25℃反应2小时,用NAP-10凝胶柱纯化,并用pH=6.0-6.1盐酸组氨酸缓冲溶液洗涤,收集滤液,得到ISAC-3的盐酸组氨酸缓冲液(3.2mg/mL,1.5mL),于4℃储存。
实施例2-4 ISAC-4
用1M磷酸氢二钠溶液调节Trastuzumab单抗储备液使其pH=6.51,再用pH=6.5-6.6磷酸缓冲盐溶液(PBS)定容至10mg/mL,得Trastuzumab单抗缓冲溶液。向所得Trastuzumab单抗缓冲溶液加入0.1M EDTA,同时加入TCEP(6eq,10mM),摇匀,25℃反应2小时后,向其中加入Trastuzumab单抗10倍摩尔当量的化合物23的DMSO溶液(10mM),摇匀,室温反应2小时,25℃反应2小时,用NAP-10凝胶柱纯化,并用pH=6.0-6.1盐酸组氨酸缓冲溶液洗涤,收集滤液,得到ISAC-4的盐酸组氨酸缓冲液(3.2 mg/mL,1.5mL),于4℃储存。
三、免疫刺激抗体偶联物(ISAC)的药物/抗体比值:DAR值的测定
LC-MS测定ISAC分子量,计算药物/抗体比DAR值。
色谱测定条件:
液相色谱柱:Thermo MAbPac RP 3.0*100mm;
流动相A:0.1%FA/H2O;流动相B:0.1%FA/ACN;
流速:0.25ml/min;样品室温度:8℃;柱温:60℃;进样量:1μl;
质谱测定条件:
质谱型号:AB Sciex Triple TOF 5600+;
GS1 35;GS2 35;CUR 30;TEM 350;ISVF 5500;DP 250;CE 10;Accumulation time 0.5s;
m/z 600-4000;Time bins to sum 40。
CE-SDS计算免疫刺激抗体偶联物ISAC-1的DAR值:8.0;ISAC-2的DAR值:8.0;ISAC-3的DAR值:7.99;ISAC-4的DAR值:8.0。
二、生物学测试
实验例1:化合物对HEK-Blue hTLR7细胞激动活性的测定
实验步骤:
1.HEK-Blue hTLR7细胞(Invivogen)培养于含有10%FBS热灭活胎牛血清(Corning)的DMEM培养基(Hyclone)中。检测当天,显微镜下观察细胞状态,将细胞收集重悬,计数后,调整细胞浓度,每孔50μL,细胞总量为2×104,接种细胞于96孔板。
2.细胞贴壁过夜后,将50μL不同浓度待测化合物加入孔板中,使其终浓度分别为100μM,33.3μM,11.11μM,3.70μM,1.23μM,0.41μM,0.14μM,0.05μM,0μM,DMSO终浓度为1%。化合物与细胞在37℃、5%CO2培养箱中孵育20h。
3.化合物孵育结束后,在显微镜下观察最大信号孔或化合物最高和最低浓度的细胞状态以及高浓度是否会有化合物结晶析出。取10μL细胞培养上清液转移到新的96孔透明板中,每孔加入90μL QUANTI-Blue(Invivogen)检测试剂,37℃孵育3h,用多功能酶标仪(BMG LABTECH)读取OD620
4.EC50通过Graphpad Prism软件log(agonist)vs.response--Variable slope拟合计算,Emax为本次试验化合物激活作用达到最大的OD620
实验结果:
按照上述方法测定化合物对HEK-Blue hTLR7细胞的激活活性,化合物的活性以EC50和Emax表示,结果如表1所示:
表1.化合物对HEK-Blue hTLR7细胞的激活作用实验结果
结果表明化合物1、4、5、6、7、8、9、11、14和16对人源TLR7具有较强的激活作用。
实验例2:化合物对HEK-Blue hTLR8细胞激动活性的测定
实验步骤:
1.HEK-Blue hTLR8细胞(Invivogen)培养于含有10%FBS热灭活胎牛血清(Corning)的DMEM培养基(Hyclone)中。检测当天,显微镜下观察细胞状态,将细胞收集重悬,计数后,调整细胞浓度,每孔50μL,细胞总量为2×104,接种细胞于96孔板。
2.细胞贴壁过夜后,将50μL不同浓度待测化合物加入孔板中,使其终浓度分别为100μM,33.3μM,11.11μM,3.70μM,1.23μM,0.41μM,0.14μM,0.05μM,0μM,DMSO终浓度为1%。化合物与细胞在37℃、5%CO2培养箱中孵育20h。
3.化合物孵育结束后,在显微镜下观察最大信号孔或化合物最高和最低浓度的细胞状态以及高浓度是否会有化合物结晶析出。取10μL细胞培养上清液转移到新的96孔透明板中,每孔加入90μL QUANTI-Blue(Invivogen-)检测试剂,37℃孵育3h,用多功能酶标仪(BMG LABTECH)读取OD620
4.EC50通过Graphpad Prism软件log(agonist)vs.response--Variable slope拟合计算,Emax为本次试验化合物激活作用达到最大的OD620
实验结果:
按照上述方法测定化合物对HEK-Blue hTLR8细胞的激活活性,化合物的活性以EC50和Emax表示,结果如表2所示:
表2.化合物对HEK-Blue hTLR8细胞的激活作用实验结果

结果表明化合物1、4、5、6、7、8、9、11、12、13、14、15、16和18对人源TLR8具有较强的激活作用。
实验例3:免疫刺激抗体偶联物(ISAC)在肿瘤细胞存在的情况下刺激外周血单个核细胞(PBMC)分泌TNF-α能力的测定
实验步骤:
1.收集表达HER2的对数生长期的HCC1954肿瘤细胞(康诺泰),用PBS清洗两遍,并用RPMI1640+10%热灭活FBS完全培养基重悬细胞调整细胞密度为2×105个/mL,将HCC1954肿瘤细胞接种于96孔板(康宁)中,每孔1×104个细胞。
2.复苏PBMC(赛笠生物),用RPMI1640+10%热灭活FBS完全培养基重悬细胞,调整密度为6×105个/mL,加入接种了肿瘤细胞的96孔板中,每孔3×104个PBMC细胞。
3.将100μL不同浓度待测物Trastuzumab单抗和免疫刺激抗体偶联物ISAC-1加入孔板中,使其终浓度分别为500nM,166.7nM,55.6nM,18.52nM,6.17nM,2.06nM,0.69nM,空白孔加入100μL RPMI1640+10%热灭活FBS完全培养基。
4.放在5%CO2,37℃培养箱中培养,培养24小时后,各孔取50μL上清检测,根据human TNF-αELISA检测试剂盒(Invitrogen)步骤孵育一抗抗体及HRP标记的二抗,显色,并读取450nm波长下的吸光度。使用Graphpad Prism软件根据标准曲线,计算出TNF-α的浓度。
表3.体外HCC1954肿瘤细胞和PBMC共孵育体系中ISAC刺激TNF-α的分泌结果
结果表明各ISAC在体外HCC1954肿瘤细胞和PBMC共孵育体系下均能有效刺激 PBMC分泌TNF-α。
实验例4.ISAC在SCID-beige小鼠异源移植瘤模型中药效评价
实验步骤:
1.每只SCID-beige小鼠(维通利华)右侧腋下处皮下接种5×106个HCC1954细胞(与Matrigel基质胶1:1等体积混合,悬浮于0.1mL PBS)。
2.待平均肿瘤体积生长至约150mm3时,剔除瘤体积不规则及过小或过大的小鼠,剩余小鼠根据肿瘤体积和动物体重进行随机分组,分别单次腹腔注射(i.p.)给予生理盐水(溶媒对照)、Trastuzumab单抗、免疫刺激抗体偶联物ISAC-1,观察受试化合物对该荷瘤鼠模型的药效及动物对受试化合物的耐受性。
3.试验中每周2次称量小鼠体重并测量瘤体积,记录数据。
4.数据统计,肿瘤体积(V)计算公式:V=1/2×a×b2,其中a和b分别表示长和宽。抗肿瘤药效用肿瘤生长抑制率TGI(%)评价,计算公式:TGI(%)(瘤体积)=[1-(TVt-TV0)/(CVt-CV0)]×100%,TV0为分组给药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;CV0为分组给药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-TV0)/TV0×100%。如果肿瘤比起始体积缩小,即Vt<V0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)。
上述实施例不以任何方式限定本发明的方案。除本文中描述的那些外,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。本申请的保护范围由所附权利要求及其任何等同物给出。

Claims (21)

  1. 式(I)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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
    其中,
    L1选自O、S和NR5
    L2为共价键,或选自C1-6亚烷基和-C1-6亚烷基-O-;
    R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-6烷基、C1-6烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
    R2选自氢、C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
    L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
    R3选自氢、卤素、C1-6烷基和C1-6烷氧基;
    R4选自氢、卤素、C1-6烷基和C1-6烷氧基;
    R5选自氢和C1-6烷基;
    R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基的基团所取代;
    R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选 地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
    R8选自氢和C1-6烷基。
  2. 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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中:
    R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-3烷基、C1-3烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
    R6选自C1-6烷基、C3-10环烷基、3-12元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基的基团所取代;
    优选地,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、卤素、羟基、C1-3烷基、C1-3烷氧基、5-10元杂芳基和R6-C(O)-NH-的基团所取代;
    R6为C1-6烷基;
    优选地,R1为C1-6烷基,所述烷基任选地被一个或多个选自氢、羟基、甲基、甲氧基和C1-3烷基-C(O)-NH-的基团所取代;
    优选地,R1为C1-6烷基,所述烷基未被取代或者被甲基、羟基、甲氧基或C1-3烷基-C(O)-NH-取代。
  3. 权利要求1-2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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中:
    R2选自氢、C1-3烷基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、R7和R7-L3-的基团所取代;
    R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
    L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
    R8选自氢和C1-6烷基;
    优选地,R2选自氢、C1-3烷基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,所述烷基、环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
    R7选自C1-6烷基、C3-10环烷基和3-12元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-6烷基、C1-6卤代烷基、C1-6烷氧基和羧基的基团所取代;
    L3选自C1-6亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
    R8选自氢和C1-6烷基;
    优选地,R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
    R7选自C1-3烷基、C3-6环烷基和3-6元杂环基,所述烷基、环烷基和杂环基任选地被一个或多个选自氢、卤素、氨基、羟基、氰基、C1-3烷基、C1-3卤代烷基、C1-3烷氧基和羧基的基团所取代;
    L3选自C1-3亚烷基、O、S、NR8、-C1-6亚烷基-NR8-、-C(O)-、-C(O)-NR8-和-NR8-C(O)-;
    R8选自氢和C1-3烷基;
    优选地,R2选自氢、C3-6环烷基、3-6元杂环基、C6-10芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基任选地被一个或多个选自氢、C1-3烷基、C1-3烷氧基、R7和R7-L3-的基团所取代;
    R7选自C1-3烷基和3-6元杂环基,所述烷基和杂环基任选地被一个或多个选自氢和C1-3烷基的基团所取代;
    L3选自C1-3亚烷基和-C1-3亚烷基-NR8-;
    R8为C1-3烷基。
  4. 权利要求1-3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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中:
    L1选自O、S和NH。
  5. 权利要求1-4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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中:
    L2为共价键,或选自C1-3亚烷基和-C1-3亚烷基-O-。
  6. 权利要求1-5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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中:
    R3选自氢、卤素、C1-3烷基和C1-3烷氧基。
  7. 权利要求1-6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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中:
    R4选自氢、卤素、C1-3烷基和C1-3烷氧基。
  8. 权利要求1-7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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有式(II-A)的结构:
    其中,R1和R2如权利要求1-7任一项所定义。
  9. 权利要求1-8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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,所述化合物具有如下结构:

  10. 下式所述药物-连接体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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
    D-L’
    其中,D为权利要求1-9中任一项所述的化合物的片段;
    L’为连接体片段;
    优选地,L’选自-L4-L5-L6,其中,
    L4选自共价键、-C(=O)-和-NH-;
    L5为聚乙二醇片段,例如其中a选自1-10的整数;
    L6其中,
    Z1选自化学键和-C(=O)-;
    Z2选自化学键和C1-20亚烷基;
    Z3选自化学键、C2-6亚烯基和C2-6亚炔基;
    A选自
    LG为亲核取代反应的离去基团,选自卤素、硝基、苯磺酸酯、对甲苯磺酸酯、三氟甲烷磺酸酯、-S(O)2-R9和-S(O)-R9
    R9选自选自C1-6烷基和C1-6卤代烷基;
    优选地,L’选自:
  11. 权利要求10所述的药物-连接体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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,所述药物-连接体选自以下结构:

  12. 式(ISAC-I)所示的免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药:
    其中,
    D为权利要求1至9中任一项所述的化合物的片段;
    Ab是可靶向目标抗原的抗体;
    L为权利要求10或11中的L’与Ab连接后得到的片段;
    z选自1-10,例如1-8。
  13. 权利要求12所述的免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述免疫刺激抗体偶联物选自:

    z独立地选自1-8,例如1-2、1-3、1-4、1-5、1-6、1-7、1-8、2-3、2-4、2-5、2-6、2-7、2-8、3-4、3-5、3-6、3-7、3-8、4-5、4-6、4-7、4-8、5-6、5-7、5-8、6-7、6-8或7-8。
  14. 化合物在制备免疫刺激抗体偶联物中的用途,所述化合物选自权利要求1-9中任一项所述的化合物。
  15. 组合物,其包含权利要求12或13所述的免疫刺激抗体偶联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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药。
  16. 权利要求15所述的组合物,其DAR值为选自1-8的整数或小数,例如1-2、1-3、1-4、1-5、1-6、1-7、1-8、2-3、2-4、2-5、2-6、2-7、2-8、3-4、3-5、3-6、3-7、3-8、4-5、4-6、4-7、4-8、5-6、5-7、5-8、6-7、6-8或7-8;
    优选地,其DAR值为7.0-8.0,例如7.1、7.2、7.3、7.4、7.5、7.6、7.7、7.8、7.9或8.0。
  17. 药物组合物,其包含预防或治疗有效量的权利要求1-9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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,或者权利要求12或13所述的免疫刺激抗体偶联物,或者权利要求15或16所述的组合物,以及一种或多种药学上可接受的载体。
  18. 权利要求1-9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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、 权利要求12或13所述的免疫刺激抗体偶联物、或者权利要求15或16所述的组合物、或者权利要求17所述的药物组合物在制备用于预防和/或治疗TLR7和/或TLR8介导的诸如癌症和病毒感染等相关疾病的药物中的用途。
  19. 一种药盒,其包含权利要求1-9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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,或者权利要求12或13所述的免疫刺激抗体偶联物,或者权利要求15或16所述的组合物,或者权利要求17所述的药物组合物。
  20. 权利要求1-9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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、权利要求12或13所述的免疫刺激抗体偶联物、或者权利要求15或16所述的组合物、或者权利要求17所述的药物组合物,其用于预防和/或治疗TLR7和/或TLR8介导的诸如癌症和病毒感染等相关疾病。
  21. 一种预防和/或治疗TLR7和/或TLR8介导的诸如癌症和病毒感染等相关疾病的方法,包括向有此需要的个体施用预防或治疗有效量的权利要求1-9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、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药、权利要求12或13所述的免疫刺激抗体偶联物、或者权利要求15或16所述的组合物、或者权利要求17所述的药物组合物。
PCT/CN2023/117167 2022-09-16 2023-09-06 一类双并环化合物、其制备方法及用途 WO2024055879A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211126580.X 2022-09-16
CN202211126580 2022-09-16

Publications (1)

Publication Number Publication Date
WO2024055879A1 true WO2024055879A1 (zh) 2024-03-21

Family

ID=90274228

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/117167 WO2024055879A1 (zh) 2022-09-16 2023-09-06 一类双并环化合物、其制备方法及用途

Country Status (1)

Country Link
WO (1) WO2024055879A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010019637A1 (en) * 2008-08-12 2010-02-18 Smithkline Beecham Corporation Chemical compounds
CN102695416A (zh) * 2009-10-29 2012-09-26 金纳斯克公司 激酶抑制剂
WO2016119707A1 (en) * 2015-01-29 2016-08-04 Hutchison Medipharma Limited Novel heteroaryl and heterocycle compounds, compositions and methods
WO2018095426A1 (zh) * 2016-11-28 2018-05-31 江苏恒瑞医药股份有限公司 吡唑并杂芳基类衍生物、其制备方法及其在医药上的应用
WO2020007275A1 (zh) * 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2023104165A1 (zh) * 2021-12-08 2023-06-15 上海维申医药有限公司 作为TLR7/8激动剂的吡啶[4,3-d]嘧啶类化合物

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010019637A1 (en) * 2008-08-12 2010-02-18 Smithkline Beecham Corporation Chemical compounds
CN102695416A (zh) * 2009-10-29 2012-09-26 金纳斯克公司 激酶抑制剂
WO2016119707A1 (en) * 2015-01-29 2016-08-04 Hutchison Medipharma Limited Novel heteroaryl and heterocycle compounds, compositions and methods
WO2018095426A1 (zh) * 2016-11-28 2018-05-31 江苏恒瑞医药股份有限公司 吡唑并杂芳基类衍生物、其制备方法及其在医药上的应用
WO2020007275A1 (zh) * 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2023104165A1 (zh) * 2021-12-08 2023-06-15 上海维申医药有限公司 作为TLR7/8激动剂的吡啶[4,3-d]嘧啶类化合物

Similar Documents

Publication Publication Date Title
US10323037B2 (en) Aminopyridazinone compounds as protein kinase inhibitors
JP6523251B2 (ja) C−結合ヘテロシクロアルキル置換ピリミジン類及びそれらの用途
WO2015039612A1 (zh) 抑制btk和/或jak3激酶活性的化合物
KR20180026537A (ko) Irak-4 저해제로서 치환된 아자 화합물
TWI823959B (zh) 吡啶并嘧啶類衍生物、其製備方法及其在醫藥上的應用
JP2021152056A (ja) 疾患を治療するためのmct4阻害剤
AU2011228703A1 (en) Spirocyclic compounds and their use as therapeutic agents and diagnostic probes
WO2017088746A1 (zh) 新的表皮生长因子受体抑制剂及其应用
WO2019158070A1 (zh) A2a和/或a2b受体拮抗剂
WO2020182159A1 (zh) Jak激酶抑制剂及其制备方法和在医药领域的应用
WO2019076336A1 (zh) 含吡唑基的三并环类衍生物、其制备方法和应用
WO2022242767A1 (zh) 螺环类化合物及其用途
EP4194457A1 (en) Compound for targeting and degrading protein, and preparation method therefor and use thereof
WO2021202652A1 (en) Tyrosine kinase 2 inhibitors, preparation methods and medicinal uses thereof
WO2022213980A1 (zh) Tyk2抑制剂及其用途
CN109071550B (zh) 具有ido/tdo抑制活性的稠合咪唑衍生物及其制备方法和应用
US20220402867A1 (en) Sulfo-substituted biaryl compound or salt thereof, preparation method therefor, and use thereof
WO2021083383A1 (zh) 一类含氮稠环类sting调节剂类化合物、制备方法和用途
WO2023109751A1 (zh) 嘧啶或吡啶类衍生物及其医药用途
CN113698408A (zh) Jnk抑制剂、其药物组合物和用途
WO2024055879A1 (zh) 一类双并环化合物、其制备方法及用途
WO2022111636A1 (zh) 稠合三环化合物、其制备方法及其在医药上的应用
WO2023030335A1 (zh) 作为tyk2/jak1假激酶结构域抑制剂的化合物及合成和使用方法
WO2023125121A1 (zh) 一类三并环化合物、其制备方法及用途
CN114805341B (zh) 一类芳杂环类化合物及其制备方法和用途

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: 23864611

Country of ref document: EP

Kind code of ref document: A1