WO2021147953A1 - 嘧啶并五元环类衍生物及其应用 - Google Patents

嘧啶并五元环类衍生物及其应用 Download PDF

Info

Publication number
WO2021147953A1
WO2021147953A1 PCT/CN2021/073098 CN2021073098W WO2021147953A1 WO 2021147953 A1 WO2021147953 A1 WO 2021147953A1 CN 2021073098 W CN2021073098 W CN 2021073098W WO 2021147953 A1 WO2021147953 A1 WO 2021147953A1
Authority
WO
WIPO (PCT)
Prior art keywords
membered
compound
amino
alkyl
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2021/073098
Other languages
English (en)
French (fr)
Inventor
张国宝
陈家隽
周峰
蒋蕾
严玉玺
唐锋
古鹏
唐任宏
任晋生
Original Assignee
江苏先声药业有限公司
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 江苏先声药业有限公司 filed Critical 江苏先声药业有限公司
Priority to CN202180010745.4A priority Critical patent/CN115244055A/zh
Publication of WO2021147953A1 publication Critical patent/WO2021147953A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

  • the present invention relates to a novel pyrimido five-membered ring derivative or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing them, and its use in the prevention or treatment of kinase-related diseases such as Janus kinase (JAK, especially JAK3) and / Or use in Bruton's tyrosine kinase (BTK) related diseases.
  • kinase-related diseases such as Janus kinase (JAK, especially JAK3) and / Or use in Bruton's tyrosine kinase (BTK) related diseases.
  • Autoimmune disease is a type of disease that is caused by abnormal immune function to attack one's own cells or tissues, leading to inflammation and tissue damage, including rheumatoid arthritis (RA), inflammatory bowel disease (IBD) and systemic lupus erythematosus (SLE) Wait.
  • RA rheumatoid arthritis
  • IBD inflammatory bowel disease
  • SLE systemic lupus erythematosus Wait.
  • BTK and JAK3 are two important targets for autoimmune diseases.
  • BTK is a member of the TEC family of non-receptor tyrosine kinases, and its structure includes PH domain, TH domain, SH3 domain, SH2 domain and SH1 domain.
  • BCR B cell antigen receptor
  • BTK has become one of the important targets for the treatment of diseases related to abnormal B cell activation, including autoimmune diseases and B-cell lymphoma.
  • Ibrutinib, Acalabrutinib and Zanubrutinib are three approved BTK inhibitors, which mainly treat B-cell lymphoma. They have obvious effects in some patients. However, serious side effects and drug-resistant mutations have also been observed clinically.
  • ibrutinib was approved by the US FDA for the treatment of graft-versus-host disease (GVHD), while other BTK inhibitors are currently being actively explored clinically to treat autoimmune diseases, including RA, SLE and multiple sclerosis (MS).
  • GVHD graft-versus-host disease
  • MS multiple sclerosis
  • JAK3 is a member of the JAK family of non-receptor tyrosine kinases.
  • the JAK kinase family has 4 members: JAK-1, JAK-2, JAK-3 and TYK-2.
  • Signal transducer and activator of transcription STAT are the downstream substrates of JAK3.
  • JAK3 activates STAT to make it a dimer into the nucleus and regulate the transcription and expression of specific genes.
  • the JAK-STAT signaling pathway plays an important role in the proliferation and differentiation of lymphocytes, as well as the expression of pro-inflammatory cytokines (JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov.
  • JAK3 has become one of the targets of autoimmune diseases and malignant tumors.
  • Tofacitinib is a JAK3 inhibitor approved by the FDA, which has shown good clinical efficacy in RA and IBD.
  • certain adverse reactions including serious infections, liver damage, etc., which are considered to be related to Tofacitinib's insufficient selectivity for JAK1/2 (JAK inhibition as a therapeutic strategy for immune and inflammatory diseases.
  • Nat Rev Drug Discov JAK3 inhibitor approved by the FDA, which has shown good clinical efficacy in RA and IBD.
  • JAK1/2 JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov.
  • the present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof:
  • R 1 is selected from H, Or the following groups optionally substituted by R a1 : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclyl, C 6 -C 10 aryl or 5-10 membered hetero Aryl;
  • R 2 is selected from hydrogen, F, Cl, Br, I, CN, OH, NO 2 , Or the following groups optionally substituted by Ra2 : NH 2 , SH, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclyl, C 1 -C 10 alkoxy , C 3 -C 10 cycloalkyloxy, 3-10 membered heterocyclyloxy, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, C 6 -C 10 aryl, 5-10 membered Heteroaryl, C 6 -C 10 aryloxy or 5-10 membered heteroaryloxy;
  • R 5 , R 6 , R 7 are independently selected from hydrogen, F, Cl, Br, I, CN, or the following groups optionally substituted by Ra3 : C 1 -C 10 alkyl, C 3 -C 10 cycloalkane Group or 3-10 membered heterocyclic group;
  • X 1 , X 2 , X 3 are independently selected from CR 8 or N;
  • R 8 is selected from H, F, Cl, Br, I, CN, OH or the following groups optionally substituted by R a4 : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl or 3-10 member Heterocyclic group;
  • R 9 , R 10 , and R 12 are independently selected from the following groups optionally substituted by Ra5 : OH, NH 2 , C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered hetero Cyclic, C 6 -C 10 aryl or 5-10 membered heteroaryl;
  • R 11 is selected from H or the following groups optionally substituted by R a6 : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclic group, C 6 -C 10 aryl or 5-10 membered heteroaryl;
  • Y and Z are independently selected from NH, O, S,
  • n1, n2, n3, q1, q2 are independently selected from 0, 1 or 2, and p1 is selected from 1, 2, 3 or 4;
  • the condition is that when Y is NH, and when R 3 is H, F, difluoromethyl or trifluoromethyl, R 1 is not methyl; when Y is O, and when R 3 and R 4 are both H When R 1 is not a methyl group; when L is selected from , R 2 is not H, CN,
  • Ra1 is selected from F, Cl, Br, I, or OH.
  • Ra3 is selected from F, Cl, Br, I.
  • R b is selected from the following groups optionally substituted by R c : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclyl, C 6 -C 10 aryl or 5-10 membered heteroaryl.
  • R 1 is selected from H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, or 3-10 membered heterocyclyl, the C 1 -C 10 alkyl, C 3- C 10 cycloalkyl or 3-10 membered heterocyclic group is optionally substituted by R a1.
  • R 1 is selected from C 1 -C 6 alkyl or 5-6 membered heterocycloalkyl, and the C 1 -C 6 alkyl, 5-6 membered heterocycloalkyl is optionally selected by Ra1 replace.
  • R 1 is selected from a C 1 -C 3 alkyl group or a 5-6 membered heterocycloalkyl group, the 5-6 membered heterocycloalkyl group contains 1 to 2 O atoms, and the C 1- C 3 alkyl or 5-6 membered heterocycloalkyl is optionally substituted by Ra1.
  • R 1 is selected from a C 1 -C 3 alkyl group or a 5-6 membered heterocycloalkyl group, and the 5-6 membered heterocycloalkyl group contains 1 to 2 O atoms.
  • R 1 is selected from methyl, ethyl, hydroxyethyl, oxetanyl, or tetrahydrofuranyl.
  • R 1 is selected from methyl, ethyl
  • R 1 is selected from methyl, ethyl or
  • R 1 is selected from ethyl.
  • R 2 is selected from H, F, Cl, Br, I, CN, NH 2 , C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, or 3-10 membered heterocyclyl, The NH 2 , C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl or 3-10 membered heterocyclic group is optionally substituted by Ra2.
  • R 2 is selected from H, F, Cl, Br, I, CN, C 1 -C 6 alkyl, COOH or CONH 2 , the C 1 -C 6 alkyl, COOH or CONH 2 is any Choose to be replaced by R b.
  • R 2 is selected from H, F, Cl, Br, or I.
  • R 2 is selected from H, F, or Cl.
  • R 2 is selected from H or Cl.
  • R 3 and R 4 are independently selected from H, F, Cl, Br, I, OH, CN, or the following groups optionally substituted by R c : NH 2 , C 1 -C 10 alkyl , C 3 -C 10 cycloalkyl, 3-10 membered heterocyclic group.
  • R 3 and R 4 are independently selected from H, F, Cl, Br, I, OH, CN, or C 1 -C 6 alkyl optionally substituted with R c.
  • R 3 and R 4 are independently selected from H, F, Cl, Br, I, OH, or methyl.
  • R 3 and R 4 are independently selected from H, F, Cl, Br, I, or methyl.
  • R 5 , R 6 , R 7 are independently selected from H, F, Cl, Br, I, CN, or C 1 -C 6 alkyl optionally substituted with Ra3.
  • R 5, R 6, R 7 are independently selected from H, F, Cl, Br, I, CN , or optionally substituted R a3 is methyl, ethyl or t-butyl.
  • R 5 , R 6 , R 7 are independently selected from H, F, Cl, Br, I, CN, methyl, or ethyl.
  • R 5 , R 6 , R 7 are independently selected from H, F, Cl, Br, I, CN, CH 2 F, or tert-butyl.
  • R 5 , R 6 , R 7 are independently selected from H, F, or CN.
  • R 5 is selected from H, F, or CN.
  • R 5 is selected from H or F.
  • R 6 and R 7 are selected from H, CH 2 F, or tert-butyl.
  • R 6 and R 7 are selected from H.
  • R 8 is selected from H, F, Cl, Br, I, CN, or C 1 -C 6 alkyl optionally substituted with Ra4.
  • R 8 is selected from H, F, Cl, Br, I, CN, methyl, or ethyl.
  • R 8 is selected from H.
  • X 1 , X 2 , X 3 are independently selected from CH or N.
  • X 2 is selected from N.
  • X 3 is selected from CH.
  • R 9 , R 10 , R 12 are independently selected from the following groups optionally substituted by Ra5 : OH, NH 2 , C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl , Phenyl or 5-6 membered heteroaryl.
  • R 11 is selected from H or the following groups optionally substituted with Ra6 : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, phenyl, or 5-6 membered heteroaryl .
  • m, n1, n2, n3, q2 are independently selected from 0 or 1, and/or p1, q1 are selected from 1 or 2.
  • n1, n2, n3 are independently selected from 0 or 1, and/or q1 is selected from 1 or 2, and/or m, p1 are independently selected from 1, and/or q2 is selected from 0.
  • q1 and p1 are independently selected from 1, and q2 is selected from 0.
  • Y and Z are independently selected from NH or O.
  • L is selected from
  • L is selected from
  • L is selected from And the NH in L is the same as Connected.
  • L is selected from When, the NH in L and Connected.
  • L is selected from
  • L is selected from
  • L is selected from
  • L is selected from
  • L is selected from
  • L is selected from
  • L is selected from
  • L is selected from
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the compound represented by formula (II) or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 5 , X 1 , and L are as defined above.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the compound represented by formula (III) or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , X 1 , and L are as defined above.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or a pharmaceutically acceptable salt thereof:
  • the present invention also provides a pharmaceutical composition, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable auxiliary materials.
  • the present invention also provides a pharmaceutical composition, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier and/or excipient.
  • the present invention relates to a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for preparing prevention or treatment of Janus kinase (JAK, especially JAK3) and/or Bruton's tyrosine
  • JAK3 Janus kinase
  • BTK Bruton's tyrosine
  • the present invention relates to a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof in the prevention or treatment of Janus kinase (JAK, especially JAK3) and/or Bruton's tyrosine kinase (BTK) Use in related diseases.
  • JAK3 Janus kinase
  • BTK Bruton's tyrosine kinase
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof for preventing or treating diseases related to Janus kinase (JAK, especially JAK3) and/or Bruton's tyrosine kinase (BTK), or Pharmaceutical composition.
  • JAK3 Janus kinase
  • BTK Bruton's tyrosine kinase
  • the present invention also relates to a method for treating diseases related to Janus kinase (JAK, particularly JAK3) and/or Bruton's tyrosine kinase (BTK), which method comprises administering to a patient a therapeutically effective dose containing the compound of the present invention A pharmaceutical preparation of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • JAK3 Janus kinase
  • BTK Bruton's tyrosine kinase
  • the diseases related to Janus kinase (JAK) and/or Bruton's tyrosine kinase (BTK) include but are not limited to tumors (such as B-cell lymphoma) and autoimmune diseases (such as rheumatism). Arthritis, inflammatory bowel disease and systemic lupus erythematosus) and so on.
  • the place indicates the connection site.
  • pharmaceutically acceptable salts refers to pharmaceutically acceptable salts of non-toxic acids or bases, including salts of inorganic acids and bases, and organic acids and bases.
  • stereoisomer refers to the isomers produced by the different arrangements of atoms in the molecule in space, including cis and trans isomers, enantiomers, diastereomers and conformational isomers.
  • the compound of the present invention may have an asymmetric atom such as a carbon atom, a sulfur atom, a nitrogen atom, a phosphorus atom (optical center), or an asymmetric double bond. Racemates, enantiomers, diastereomers, geometric isomers and individual isomers are all included in the scope of the present invention.
  • the compounds of the present invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers Conformers, (D)-isomers, (L)-isomers, and their racemic mixtures and other mixtures, such as enantiomers or diastereomer-enriched mixtures, all of these mixtures belong to Within the scope of the present invention.
  • Additional asymmetric carbon atoms, asymmetric sulfur atoms, asymmetric nitrogen atoms, or asymmetric phosphorus atoms may be present in substituents such as alkyl groups.
  • the asymmetric atom-containing compound of the present application can be isolated in an optically pure form or a racemic form.
  • the optically active pure form can be resolved from the racemic mixture or synthesized by using chiral raw materials or chiral reagents.
  • stereoisomers include, but are not limited to:
  • tautomer refers to an isomer of a functional group resulting from the rapid movement of an atom in a molecule at two positions.
  • the compounds of the present invention may exhibit tautomerism.
  • Tautomeric compounds can exist in two or more mutually convertible species.
  • Proton shift tautomers result from the migration of covalently bonded hydrogen atoms between two atoms.
  • Tautomers generally exist in an equilibrium form, and an attempt to separate a single tautomer usually produces a mixture whose physical and chemical properties are consistent with a mixture of compounds. The position of equilibrium depends on the chemical properties of the molecule.
  • the ketone type is dominant; in phenol, the enol type is dominant.
  • the present invention encompasses all tautomeric forms of the compound.
  • pharmaceutical composition means a mixture of one or more of the compounds described in the text or their physiologically/pharmaceutically acceptable salts or prodrugs and other chemical components, such as physiologically/pharmaceutically acceptable carriers And excipients.
  • the purpose of the pharmaceutical composition is to facilitate the administration of the compound to the organism.
  • substituted means that any one or more hydrogen atoms on a specific atom are replaced by a substituent, as long as the valence of the specific atom is normal and the substituted compound is stable.
  • it means that two hydrogen atoms are replaced, and the oxo will not occur on the aromatic group.
  • the term “optional” or “optionally” means that the event or situation described later can occur or not occur, and the description includes occurrence of said event or situation and non-occurrence of said event or situation.
  • the ethyl group is "optionally" substituted by halogen, meaning that the ethyl group can be unsubstituted (CH 2 CH 3 ), monosubstituted (such as CH 2 CH 2 F), or polysubstituted (such as CHFCH 2 F, CH 2 CHF 2 etc.) or completely substituted (CF 2 CF 3 ).
  • CH 2 CH 3 unsubstituted
  • monosubstituted such as CH 2 CH 2 F
  • polysubstituted such as CHFCH 2 F, CH 2 CHF 2 etc.
  • CF 2 CF 3 completely substituted
  • halo or halogen refers to fluorine, chlorine, bromine and iodine.
  • C 1 -C 10 alkyl should be understood to mean a linear or branched saturated monovalent hydrocarbon group having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms.
  • the alkyl group is, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl Group, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-d
  • alkoxy can be understood as “alkyloxy” or “alkyl-O", preferably, "C 1 -C 10 alkoxy” may include “C 1 -C 6 alkoxy” and " C 1 -C 3 alkoxy”.
  • C 2 -C 10 alkenyl should be understood to preferably mean a linear or branched monovalent hydrocarbon group, which contains one or more double bonds and has 2, 3, 4, 5, 6, 7, 8, 9 , 10 carbon atoms, preferably "C 2 -C 6 alkenyl", more preferably “C 2 -C 4 alkenyl", still more preferably C 2 or C 3 alkenyl. It should be understood that where the alkenyl group contains more than one double bond, the double bonds may be separated from each other or conjugated.
  • alkenyl groups such as vinyl, allyl, (E)-2-methylvinyl, (Z)-2-methylvinyl, (E)-but-2-enyl, (Z)-butanyl -2-enyl, (E)-but-1-enyl, (Z)-but-1-enyl, isopropenyl, 2-methylprop-2-enyl, 1-methylprop-2 -Alkenyl, 2-methylprop-1-enyl, (E)-1-methylprop-1-enyl, (Z)-1-methylprop-1-enyl.
  • C 2 -C 10 alkynyl should be understood to mean a linear or branched monovalent hydrocarbon group containing one or more triple bonds and having 2, 3, 4, 5, 6, 7, 8, 9, 10 carbon atoms, "C 2 -C 6 alkynyl” is preferred, “C 2 -C 4 alkynyl” is more preferred, and C 2 or C 3 alkynyl is still more preferred.
  • the alkynyl group is, for example, ethynyl, prop-1-ynyl, prop-2-ynyl, but-1-ynyl, but-2-ynyl, but-3-ynyl, 1-methylprop- 2-alkynyl.
  • C 3 -C 10 cycloalkyl should be understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring, which has 3 to 10 carbon atoms. Such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or a bicyclic hydrocarbon group such as decalin ring.
  • C 3 -C 6 cycloalkyl should be understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring having 3 to 6 carbon atoms.
  • the "C 3 -C 10 cycloalkyl group” may include a "C 3 -C 6 cycloalkyl group”.
  • cycloalkyloxy can be understood as “cycloalkyl-O", preferably, "C 3 -C 10 cycloalkyloxy” may include “C 3 -C 6 cycloalkyloxy”.
  • heterocyclic group refers to a monocyclic heterocyclic group, a fused heterocyclic group system; a fused heterocyclic group includes a heterocyclic group, a spiro heterocyclic group, a bridged heterocyclic group, which may be saturated, partially saturated or non- Saturated, but not aromatic; the pentaheterocyclic group, spiro heterocyclic group, and bridged heterocyclic group include but are not limited to 6-18 membered hexaheterocyclic group, 5-18 membered spiro heterocyclic group or 5-18
  • the membered bridge heterocyclic group, the pentaheterocyclic group, spiro heterocyclic group, and bridge heterocyclic group may contain 1 to 5 heteroatoms, and the heteroatoms are selected from N, O, B and S.
  • 3-10 membered heterocyclic group means a saturated or partially saturated monovalent monocyclic, fused ring, spiro ring or bridged ring, which contains 1-5, preferably 1-3 selected from N, O, Heteroatoms of B and S.
  • the heterocyclic group may include but is not limited to: 4-membered ring, such as azetidinyl, oxetanyl; 5-membered ring, such as tetrahydrofuranyl, dioxolyl, pyrrole Alkyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl; or 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithiaalkyl, thiomorpholinyl, piperazinyl Or trithiaalkyl; or partially saturated 6-membered ring such as tetrahydropyridyl; or 7-membered ring such as diazacycloheptanyl.
  • 4-membered ring such as azetidinyl, oxetanyl
  • 5-membered ring such as tetrahydrofuranyl, dioxolyl, pyrrol
  • the heterocyclic group may be benzo-fused.
  • the heterocyclic group may be bicyclic, such as but not limited to a 5, 5-membered ring, such as hexahydrocyclopenta[c]pyrrole-2(1H)-yl ring, or a 5, 6-membered bicyclic ring, such as hexahydropyrrole And [1,2-a]pyrazine-2(1H)-yl ring.
  • the ring containing the nitrogen atom may be partially unsaturated, that is, it may contain one or more double bonds, such as but not limited to 2,5-dihydro-1H-pyrrolyl, 4H-[1,3,4]thiadi Azinyl, 4,5-dihydrooxazolyl or 4H-[1,4]thiazinyl, or it may be benzo-fused, such as but not limited to dihydroisoquinolinyl.
  • the 3-10 membered heterocyclic group may be a "3-10 membered heterocycloalkyl", which means a saturated monovalent monocyclic, fused ring, spiro ring or bridge containing 1-5 heteroatoms. Ring;
  • "3-10 membered heterocycloalkyl” includes 5-6 membered heterocycloalkyl; according to the present invention, the heterocyclic group is non-aromatic.
  • heterocyclyloxy can be understood as “heterocyclyl-O", preferably, “3-10 membered heterocyclyloxy” may include “3-6 membered heterocyclyloxy”.
  • C 6 -C 10 aryl should be understood to preferably mean a monovalent or partially aromatic monocyclic or bicyclic hydrocarbon ring having 6, 7, 8, 9, 10 carbon atoms.
  • a ring having 6 carbon atoms such as phenyl; or a ring having 9 carbon atoms (“C 9 aryl”), such as indanyl or indenyl, or having 10
  • a ring of three carbon atoms such as tetrahydronaphthyl, dihydronaphthyl, or naphthyl.
  • C 6 -C 10 aryloxy can be understood as “C 6 -C 10 aryl-O”.
  • 5-10 membered heteroaryl should be understood to include monovalent monocyclic, bicyclic or tricyclic aromatic ring systems having 5, 6, 7, 8, 9, 10 ring atoms, especially 5 or 6 or 9 or 10 ring atoms, and it contains 1 to 5, preferably 1 to 3 heteroatoms independently selected from N, O and S and, in addition, may be benzo-fused in each case.
  • the heteroaryl group is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiol Diazolyl, etc.
  • treatment means administering the compound or formulation described in this application to prevent, ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
  • terapéuticaally effective amount means (i) treatment or prevention of a particular disease, condition or disorder, (ii) reduction, amelioration or elimination of one or more symptoms of a particular disease, condition or disorder, or (iii) prevention or delay
  • the amount of the compound of the present invention that constitutes a “therapeutically effective amount” varies depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but it can be routinely determined by those skilled in the art. Determined by its own knowledge and this disclosure.
  • excipients refers to pharmaceutically acceptable inert ingredients.
  • examples of types of the term “excipient” include, without limitation, binders, disintegrants, lubricants, glidants, stabilizers, fillers, diluents, and the like. Excipients can enhance the handling characteristics of the pharmaceutical preparation, that is, make the preparation more suitable for direct compression by increasing fluidity and/or adhesion.
  • examples of typical "pharmaceutically acceptable carriers” suitable for the above formulations are: sugars, starches, cellulose and its derivatives and other auxiliary materials commonly used in pharmaceutical formulations.
  • pharmaceutically acceptable excipients refers to those excipients that have no obvious stimulating effect on the organism and will not damage the biological activity and performance of the active compound.
  • Suitable auxiliary materials are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and those well known to those skilled in the art Equivalent alternatives, preferred implementations include but are not limited to the embodiments of the present invention.
  • the present application also includes compounds of the present application that are the same as those described herein, but have one or more atoms replaced by an isotope-labeled atom having an atomic weight or mass number different from the atomic weight or mass number commonly found in nature.
  • isotopes that can be bound to the compounds of the present application include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl, etc.
  • isotope-labeled compounds of the application can be used in compound and/or substrate tissue distribution analysis. Tritiated (ie 3 H) and carbon-14 (ie 14 C) isotopes are especially preferred due to their ease of preparation and detectability. Positron emission isotopes such as 15 O, 13 N, 11 C, and 18 F can be used in positron emission tomography (PET) studies to determine substrate occupancy.
  • PET positron emission tomography
  • the isotopically-labeled compounds of the present application can be prepared by the following procedures similar to those disclosed in the schemes and/or examples below, by replacing non-isotopically-labeled reagents with isotope-labeled reagents.
  • substitution with heavier isotopes can provide certain therapeutic advantages resulting from higher metabolic stability (for example, increased in vivo half-life or reduced dosage requirements), and therefore in certain situations
  • deuterium substitution can be partial or complete, and partial deuterium substitution refers to the substitution of at least one hydrogen by at least one deuterium.
  • the pharmaceutical composition of the present application can be prepared by combining the compound of the present application with suitable pharmaceutically acceptable excipients, for example, can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, and powders. , Granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols.
  • Typical routes for administering the compound of the present application or a pharmaceutically acceptable salt or pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, Intramuscular, subcutaneous, and intravenous administration.
  • the pharmaceutical composition of the present application can be manufactured by methods well known in the art, such as conventional mixing method, dissolution method, granulation method, sugar-coated pill method, grinding method, emulsification method, freeze-drying method, etc.
  • the pharmaceutical composition is in an oral form.
  • the pharmaceutical composition can be formulated by mixing the active compound with pharmaceutically acceptable excipients well known in the art. These auxiliary materials enable the compound of the present application to be formulated into tablets, pills, lozenges, sugar-coated agents, capsules, liquids, gels, slurries, suspensions, etc., for oral administration to patients.
  • the solid oral composition can be prepared by conventional mixing, filling or tableting methods. For example, it can be obtained by the following method: mixing the active compound with solid excipients, optionally grinding the resulting mixture, adding other suitable excipients if necessary, and then processing the mixture into granules to obtain tablets Or the core of the dragee.
  • suitable excipients include but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
  • the pharmaceutical composition may also be suitable for parenteral administration, such as a sterile solution, suspension or lyophilized product in a suitable unit dosage form.
  • the daily dose is 0.01 to 100 mg/kg body weight, preferably 0.05 to 50 mg/kg body weight, more preferably 0.1 to 30 mg/kg body weight, in single or divided doses form.
  • Figure 1 is the curve of the clinical arthritis score of each group of mice over time.
  • Figure 2 is a histogram of the area under the curve of the clinical arthritis score of each group of mice over time.
  • Figure 3 is a representative section of joint pathology of mice in each group.
  • Figure 4 is a histogram of joint pathology scores of mice in each group.
  • the solvent used in the present invention is commercially available. Commercially available compounds use supplier catalog names.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) and/or mass spectrometry (MS).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the unit of NMR shift is 10 -6 (ppm).
  • the solvents measured by NMR are deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and the internal standard is tetramethylsilane (TMS);
  • TMS tetramethylsilane
  • IC 50 refers to the half inhibitory concentration, which refers to the half of the maximum inhibitory effect concentration.
  • the following eluent can be formed by two or more solvents to form a mixed eluent, and the percentage is the volume ratio of each solvent.
  • the volumetric dosage of ethyl acetate in the stripping agent: petroleum ether is 0:100 ⁇ 10:100.
  • the present invention uses the following acronyms:
  • reaction solution was quenched with saturated ammonium chloride solution (20mL), and then extracted with ethyl acetate (20mL*3).
  • the combined organic phase was dried and filtered with anhydrous sodium sulfate.
  • the filtrate was concentrated under reduced pressure to obtain the crude title product 1f (580mg ), the crude product was directly used in the next reaction.
  • reaction solution was stirred at room temperature for 1 hour until the reaction was complete.
  • the reaction solution was diluted with water (30 mL), extracted with ethyl acetate (30 mL*2), the combined organic phase was dried over anhydrous sodium sulfate, filtered and spin-dried to obtain the crude product 1i (310 mg), which was directly used in the next step.
  • Preparative high performance liquid chromatography Waters 2767 preparative chromatograph, mobile phase A: 0.1% NH 4 HCO 3 aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10 ⁇ m), gradient: 10% ACN Run for 3 minutes, 65%-70% ACN runs to 13 minutes, 95% ACN runs to 16 minutes, and 10% ACN runs to 18 minutes to complete purification and obtain the target compound.
  • the third step 5-chloro-N-(1-ethyl-1H-pyrazol-4-yl)-4-(1-phenylethoxy)-7-(((2-(trimethylform Synthesis of (silyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidin-2-amine 2f
  • Step 5 Synthesis of 4,5-Dichloro-N-(1-ethyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-2-amine (2h)
  • the seventh step 4-(3,8-diazabicyclo[4.2.0]octane-8-yl)-5-chloro-N-(1-ethyl-1H-pyrazol-4-yl)- Synthesis of 7H-pyrrolo[2,3-d]pyrimidin-2-amine 2k
  • Preparative high performance liquid chromatography Waters 2767 preparative chromatograph, mobile phase A: 10mmol% FA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10 ⁇ m), gradient: 10% ACN running for 3 minutes, 30%-35% ACN was run for 12 min, 95% ACN was run for 17 min, and 10% ACN was run for 19 min to complete purification, and the target compound was obtained.
  • Triethylamine (13mg, 0.13mmol) was added to the crude 5-chloro-N-(1-ethyl-1H-pyrazol-4-yl)-4-(1,6-diazaspiro[3.5 ]Non-1-yl)-7H pyrrolo[2,3-d]pyrimidin-2-amine 3f (10 mg) hydrochloride in dichloromethane (3 mL). The resulting solution was stirred at room temperature for 10 minutes. Then the solution was cooled with an ice water bath, and a tetrahydrofuran solution (1 mL) of 3 g (2.4 mg, 0.027 mmol) of acryloyl chloride was slowly added dropwise.
  • reaction solution was raised to room temperature and stirred for 1 hour.
  • the reaction solution was concentrated under reduced pressure, and the crude product obtained was dissolved in dimethylformamide by Waters 2767 preparative chromatograph, mobile phase A: 0.1% FA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN for 3 min, 30%-35% ACN for 10 min, 95% ACN for 12 min, and 10% ACN for 15 min to complete purification to obtain product 003 (2.0 mg).
  • High performance liquid chromatography Waters 2767 preparative chromatograph, mobile phase A: 0.1% FA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10 ⁇ m), gradient: 10% ACN running for 3 minutes, 30%-35% ACN was run for 13 min, 95% ACN was run for 16 min, and 10% ACN was run for 18 min to complete purification, and the target compound 004 was obtained.
  • reaction solution was diluted with water (10 mL), extracted with dichloromethane (20 mL*2), the combined organic phase was washed once with saturated brine, dried over anhydrous sodium sulfate, filtered and spin-dried to obtain the crude product. Purified by HPLC to obtain product 005 (11.54 mg).
  • Preparative high performance liquid chromatography Waters 2767 preparative chromatograph, mobile phase A: 10mmol% NaHCO 3 aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10 ⁇ m), gradient: 10% ACN for 3 min , 30%-40% ACN was run to 16 min, 95% ACN was run to 15 min, and 10% ACN was run to 20 min to complete purification, and compound 005 was obtained.
  • Trifluoroacetic acid (3 mL) was added to a dichloromethane solution (10 mL) of compound tert-butyl 6a (800 mg, 3.55 mmol) at room temperature and stirred for 2 hours. The reaction was found to be complete (iodine smoked) by spotting. This solution was concentrated to dryness under reduced pressure to obtain crude product 6b (450 mg).
  • Step 2 Synthesis of benzyl 4-carbonyl-6-azaspiro[2.5]octane-6-carboxylate 6c
  • the third step the synthesis of benzyl (R,Z)-4-((tert-butylsulfinyl)imino)-6-azaspiro[2.5]octane-6-carboxylate 6e
  • Step 5 Synthesis of 6g benzyl 4-amino-6-azaspiro[2.5]octane-6-carboxylate
  • the seventh step Benzyl 4-((5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-4 -Yl)amino)-6-azaspiro[2.5]octane-6-carboxylate 6k
  • reaction solution was concentrated under reduced pressure and then passed through Waters 2767 preparative chromatograph, mobile phase A: 0.1% FA aqueous solution, mobile phase B: acetonitrile, chromatographic column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN to run for 3 minutes, 30% to 35% ACN to run to 15 minutes, 95% ACN to run to 18 minutes, 10% ACN to run to 20 minutes to complete purification to obtain the title product 6k (170 mg).
  • mobile phase A 0.1% FA aqueous solution
  • mobile phase B acetonitrile
  • chromatographic column Sunfire (Prep C18 OBD 19*250mm 10um)
  • gradient 10% ACN to run for 3 minutes, 30% to 35% ACN to run to 15 minutes, 95% ACN to run to 18 minutes, 10% ACN to run to 20 minutes to complete purification to obtain the title product 6k (170 mg).
  • the ninth step (R)-1-(4-((5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7Hpyrrolo[2,3-d] (Pyrimidine-4-yl)amino)-6-azaspirocyclo[2.5]octane-6-yl)prop-2-en-1-one 006-1 and (S)-1-(4-((5 -Chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7Hpyrrolo[2,3-d]pyrimidin-4-yl)amino)-6-azaspiro[ 2.5] Synthesis of octane-6-yl)prop-2-en-1-one 006-2
  • Triethylamine (0.1mL, 0.6mmol) was added to compound (R)-5-chloro-N2-(1-ethyl-1H-pyrazol-4-yl)-N4-(6-azaspiro) at room temperature
  • Octan-4-yl)-7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine 61 120 mg, trifluoroacetate
  • dichloromethane (10 mL). The resulting solution was stirred at room temperature for 10 minutes.
  • the reaction solution was concentrated under reduced pressure, and the crude product obtained was dissolved in dimethylformamide by Waters 2767 preparative chromatograph, mobile phase A: 0.1% FA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18OBD 19*250mm 10um ), gradient: 10% ACN runs for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 15 minutes, and 10% ACN runs to 20 minutes to complete purification to obtain the target product 006 (40 mg, yield 29.2%), 006 ( 25 mg) was resolved by SFC to obtain the title product 006-1 (8.0 mg, 5.5%), 006-2 (4.0 mg, 2.8%).
  • SFC preparation method instrument: Waters SFC80, preparation column: ChiralPak AD, 250 ⁇ 25mm, 10 ⁇ m, mobile phase: A for CO 2 and B for MeOH (0.1% DEA), gradient: B 0-40%, flow rate: 70g /min, pressure: 100bar, temperature: 35°C, wavelength: 214nm, time: 8min, sample dissolution method: dissolve in 5 ml methanol, injection volume: 3.5 ml/needle.
  • Step 2 Synthesis of benzyl 7-carbonyl-5-azaspiro[2.4]heptane-5-carboxylate 7c
  • the third step Synthesis of benzyl (R,Z)-7-((tert-butylsulfinyl)imino)-5-azaspiro[2.4]heptane-5-carboxylate 7e
  • Step 5 Synthesis of 7g benzyl 7-amino-5-azaspiro[2.4]heptane-5-carboxylate
  • the seventh step Benzyl 7-((5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-4 -Yl)amino)-5-azaspiro[2.4]heptane-5-carboxylate 7k
  • the ninth step 1-(7-(5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7H pyrrolo[2,3-d]pyrimidin-4-yl )Amino)-5-azaspiro[2.4]heptane-5-yl)prop-2-en-1-one 007; (R)-1-(7-(5-chloro-2-((1- Ethyl-1H-pyrazol-4-yl)amino)-7H pyrrolo[2,3-d]pyrimidin-4-yl)amino)-5-azaspiro[2.4]heptan-5-yl)propane -2-en-1-one 007-1; and (S)-1-(7-(5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7H pyrrole Synthesis of 2-[2,3-d]pyrimidin-4-yl)amino
  • triethylamine 51mg, 0.50mmol was added to the crude product compound 5-chloro-N 2 -(1-ethyl-1H-pyrazol-4-yl)-N 4 -(5-aza Spiro[2.4]heptane-7-yl)-7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine 7l (182mg) in dichloromethane (5mL) solution, stir at this temperature 10 minutes. Then, a dichloromethane solution (0.5 mL) containing acryloyl chloride 7m (30.4 mg, 0.33 mmol) was slowly added dropwise. The temperature of the reaction solution was raised to room temperature and stirred for 2 hours.
  • Preparation method instrument: Waters SFC80, preparation column: ChiralPak OD, 250 ⁇ 25mm, 10 ⁇ m, mobile phase: A for CO 2 and B for EtOH (0.1% DEA), gradient: B 0-40%, flow rate: 70g/ min, pressure: 100bar, temperature: 35°C, wavelength: 214nm, time: 13min, sample dissolution method: dissolve in 5ml methanol, injection volume: 3.5ml/needle.
  • Trifluoroacetic acid (4 mL) was added to a dichloromethane (6 mL) solution of (R)-8e (65.3 mg, 0.15 mmol) in a single-necked flask at room temperature, and the mixture was stirred at room temperature for 1 hour. Point the board to monitor the reaction is complete. The solution was concentrated under reduced pressure, dichloromethane (6 mL) was added again, and concentrated again, and the resulting crude product 8f was directly used in the next reaction.
  • Triethylamine (89 mg, 0.88 mmol) was added to a solution of (R)-8f (0.146 mmol, trifluoroacetate) in tetrahydrofuran (8 mL) at room temperature. The resulting solution was stirred at room temperature for 10 minutes. Then the solution was cooled with an ice water bath, and a tetrahydrofuran solution (2.0 mL) containing 8 g (13.2 mg, 0.146 mmol) of acryloyl chloride was slowly added dropwise. The temperature of the reaction solution was raised to room temperature and stirred for 2 hours. The reaction solution was concentrated under reduced pressure, and the resulting crude product was dissolved in dimethylformamide and purified by reverse phase Flash to obtain the title product 008 (21.4 mg).
  • the first step the synthesis of tert-butyl 4-((2,5-dichloro-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)azepane-1-carboxylate 9c
  • Triethylamine (0.15 mL, 2.0 mmol) was added to a solution of 9f (250 mg, trifluoroacetate) in dichloromethane (10 mL) at room temperature. The resulting solution was stirred at room temperature for 10 minutes. Then the solution was cooled with an ice water bath, and a tetrahydrofuran solution (2.0 mL) containing acryloyl chloride for 9 h (90 mg, 1.0 mmol) was slowly added dropwise. The temperature of the reaction solution was raised to room temperature and stirred overnight. The reaction solution was concentrated under reduced pressure to obtain compound 009, which was dissolved in dimethylformamide, purified by reverse phase Flash and resolved by SFC to obtain title products 009-1 (25.3 mg) and 009-2 (39.5 mg).
  • Preparation method instrument: Waters SFC80, preparation column: ChiralPak AY, 250 ⁇ 25mm, 10 ⁇ m, mobile phase: A for CO 2 and B for MeOH (0.1% DEA), gradient: B 0-40%, flow rate: 70g/ min, pressure: 100bar, temperature: 35°C, wavelength: 214nm, time: 13min, sample dissolution method: dissolve in 15 ml methanol, injection volume: 5 ml/needle.
  • the first step (tert-butyl(3-((2,5-dichloro-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenyl)carbamate)010- b synthesis
  • Trifluoroacetic acid (37.0mg, 0.38mmol) was added to isopropanol dissolved with 010-b (150.0mg, 0.38mmol) and 1-ethyl-1H-pyrazole-4-amine (84.0mg, 0.76mmol) (5mL), heat the solution to 110°C in a sealed tube and continue to stir for 10 hours. After the temperature of the reaction system dropped to room temperature and concentrated under reduced pressure, the crude product obtained was purified by reverse phase Flash to obtain 010-c (50 mg, yield: 28%, purity: 90%).
  • the third step (4-(3-aminophenoxy)-5-chloro-N-(1-ethyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine -2-amine) 010-d synthesis
  • the dioxane hydrochloride solution (4.0M, 2mL) was added to a single-necked flask containing 010-c (50mg, 0.11mmol). After stirring for 30 minutes, the reaction solution was concentrated under reduced pressure to obtain the crude product 010-d (50 mg, hydrochloride, purity: 55%), which was used in the next step without further purification.
  • the fourth step (N-(3-((5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7Hpyrrolo[2,3-d]pyrimidine-4 -Oxy) phenyl) acrylamide) 010 synthesis
  • Triethylamine (36 mg, 0.36 mmol) was added to a solution of compound 010-d (50 mg, hydrochloride) in dichloromethane (2 mL) at about 5°C.
  • a dichloromethane solution (1.0 mL) containing acryloyl chloride (12 mg, 0.13 mmol) was slowly added dropwise.
  • the temperature of the reaction solution was raised to room temperature and stirred for 30 minutes.
  • the reaction solution was concentrated under reduced pressure for preparation and purification (conditions: acetonitrile/water (0.04% formic acid)) and purification to obtain the target product 010 (7.02 mg, yield: 13%).
  • Step 1 Synthesis of 2,4,5-trichloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H pyrrolo[2,3-d]pyrimidine 011-1-b
  • the fifth step 1-((3S,4R)-3-((5-chloro-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7Hpyrrolo[2,3- d) Synthesis of pyrimidin-4-yl)oxy)-4-fluoropyrrolidin-1-yl)prop-2-en-1-one 011-1
  • Step 1 Synthesis of 2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H pyrrolo[2,3-d]pyrimidine 012-1-b
  • 012-1-d (150mg, 0.45mmol) was dissolved in a mixture of acetone (4mL) and H 2 O (2mL), potassium carbonate solid (186mg, 1.35mmol) was added and stirred at room temperature, the acryloyl chloride (60mg, 0.67 mmol) was added dropwise, stirred at room temperature for 2 hours, filtered, prepared and purified (conditions: acetonitrile/water (0.04% formic acid)) to obtain 012-1 (42.5 mg, yield 25.3% in three steps).
  • Step 1 Synthesis of 4,6-dichloro-1-tetrahydropyran-7H-pyrazole[2,3-D]pyrimidine 013-1-b
  • the fifth step (1-((3S,4R)-3-((2-((1-ethyl-1H-pyrazol-4-yl)amino)-7Hpyrazolo[2,3-d] Synthesis of (pyrimidin-4-yl)oxy)-4-fluoropyrrolidin-1-yl)prop-2-en-1-one 013-1
  • the crude product 013-1-e was dissolved in acetone and water (2.0 mL). Potassium carbonate (49.7 mg, 0.36 mol) was added and stirred at this temperature for 10 minutes; then, acetone solution (0.5 mL) containing acryloyl chloride (39.1 mg, 0.43 mmol) was slowly added dropwise. The temperature of the reaction solution was raised to room temperature and stirred for 2 hours. The crude product was filtered and purified by reverse phase Flash (conditions: acetonitrile/water (0.04% formic acid)) to obtain 013-1 (29.4 mg, 42.3% yield in two steps).
  • the first step tert-butyl (R)-3-((2,5-dichloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7Hpyrrolo[2,3-d ]Pyrimidine-4-yl)oxy)pyrrolidine-1-carboxylate 015-1-b synthesis
  • 015-1-d (281 mg, 0.74 mmol) was added to a methanol solution (8 mL) of DIPEA (4 mL) at room temperature, and the reaction solution was stirred at 50°C overnight. The reaction solution was concentrated under reduced pressure to obtain 015-1-e (259 mg, yield: >99%). The crude product was used directly in the next step without further purification.
  • the first step tert-butyl (R)-3-((2,5-dichloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7Hpyrrolo[2,3-d ]Pyrimidine-4-yl)oxy)pyrrolidine-1-carboxylate synthesis 016-1-b synthesis
  • reaction solution was concentrated under reduced pressure and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN Run for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 15 minutes, and 10% ACN runs to 20 minutes to complete purification to obtain 016-1-c (289 mg, yield: 69.5%).
  • the third step 5-chloro-4-(((R)-pyrrolidin-3-yl)oxy)-N-(1-((S)-tetrahydrofuran-3-yl)-1H-pyrazole-4 -Yl)-7H-pyrrolo[2,3-d]pyrimidin-2-amine 016-1-d synthesis
  • 016-1-c (289 mg, 0.46 mmol) was added to a dichloromethane solution (4 mL) of trifluoroacetic acid (2 mL) at room temperature, and the reaction solution was stirred at room temperature for 2 hours.
  • the reaction solution was concentrated under reduced pressure, added to a methanol solution (4 mL) of DIPEA (3 mL), and the reaction solution was stirred at 50°C overnight.
  • the fourth step 1-((R)-3-((5-chloro-2-((1-((S)-tetrahydrofuran-3-yl)-1H-pyrazol-4-yl)amino)-7H -Pyrrolo[2,3-d]pyrimidin-4-yl)oxy)pyrrolidin-1-yl)prop-2-en-1-one 016-1
  • 016-1-d (160 mg, 0.41 mmol) was dissolved in a mixture of acetone (4 mL) and H 2 O (2 mL) at room temperature, and potassium carbonate solid (169.7 mg, 1.38 mmol) was added and stirred.
  • Step 1 Synthesis of (S)-3-p-toluenesulfonyltetrahydrofuran 025-1-a
  • the intermediate 025-1-h (420 mg) was dissolved in acetone and water (1:1, 5.0 mL, potassium carbonate (243.0 mg, 1.6 mol) was added, and the mixture was stirred at this temperature for 10 minutes. Then dropped slowly.
  • acetone solution 0.5 mL
  • acryloyl chloride 95.5 mg, 983.6 mmol.
  • the temperature of the reaction solution was raised to room temperature and stirred for 2 hours.
  • the reaction solution was washed with saturated brine (5.0 mL), and the crude product obtained was dissolved in DMF.
  • the crude product 025-2-f was dissolved in acetone and water (1:1, 5.0 mL), potassium carbonate (322.9 mg, 2.4 mol) was added, and the mixture was stirred at this temperature for 10 minutes. Then, an acetone solution (0.5 mL) containing acryloyl chloride (130.3 mg, 1.5 mmol) was slowly added dropwise. The temperature of the reaction solution was raised to room temperature and stirred for 2 hours.
  • reaction solution was washed with saturated brine (10.0 mL), and the crude product obtained was dissolved in DMF and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19 *250mm 10um), gradient: 10% ACN runs for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 16 minutes, and 10% ACN runs to 20 minutes to complete purification to obtain the product 025-2 (154.9 mg, yield: 41.8%).
  • Trifluoroacetic acid (2 mL) was added to the dichloromethane solution (5.0 mL) of Intermediate 026-c (256 mg, 0.539 mmol) at 0°C, and the mixture was stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to obtain intermediate 026-d (202 mg, yield: 99%), which was directly used in the next reaction without purification.
  • reaction solution was filtered and purified (Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18OBD 19*250mm 10um), gradient: 10% ACN for 3min, 30 %-35% ACN runs to 13 min, 95% ACN runs to 15 min, and 10% ACN runs to 20 min to complete purification) to obtain product 026 (7.78 mg, yield: 12%).
  • 011-1-b (351 mg, 1.0 mmol), 028-b (200 mg, 1.0 mmol) and DIPEA (0.3 mL, 2.0 mmol) were added to the isopropanol (10.0 mL) solution at room temperature.
  • the reaction system was sealed and heated to 100°C, and stirred at this temperature for 16 hours.
  • the reaction solution was concentrated under reduced pressure to obtain a crude product, which was purified to obtain Intermediate 028-c (440 mg, yield: 85.4%).
  • reaction solution was filtered, preparative and purified (Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN for 3 min, Run 30%-35% ACN for 12 minutes, 95% ACN for 16 minutes, and 10% ACN for 20 minutes to complete purification) to obtain 028 (28 mg, purity: 94%).
  • the first step 7-((2,5-Dichloro-7-((2-(trimethylsilyl)ethoxy)7H-pyrrolo[2,3-d]pyrimidin-4-yl) Synthesis of amino)-5-azaspirocyclo[2.4]heptane-5-carboxylic acid benzyl ester 029-c
  • the second step 7-((5-chloro-2-((1-methyl-1H-pyrazol-4-yl)amino)-7-((2-(trimethylsilyl)ethoxy )-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)-5-azaspirocyclo[2.4]heptane-5-carboxylic acid benzyl ester 029-e
  • the crude intermediate 030-e (0.519mmol) was dissolved in a mixed solvent of acetone (3.0ml) and water (3.0mL), the pH value was adjusted to 10 with potassium carbonate, the reaction solution was cooled to 0°C, and then the acryloyl chloride (54 mg, 0.6 mmol) was dissolved in acetone (1.0 mL) and added dropwise to the reaction solution. After the addition was completed, the reaction solution was stirred at 0°C for 1 h.
  • reaction solution was filtered and sent directly to the Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10 %ACN was run for 3 minutes, 30%-35% ACN was run for 13 minutes, 95% ACN was run for 15 minutes, and 10% ACN was run for 20 minutes to complete purification, and product 030 (7.04 mg) was obtained.
  • mobile phase A 1 ⁇ formic acid aqueous solution
  • mobile phase B acetonitrile
  • column Sunfire (Prep C18 OBD 19*250mm 10um)
  • gradient 10 %ACN was run for 3 minutes, 30%-35% ACN was run for 13 minutes, 95% ACN was run for 15 minutes, and 10% ACN was run for 20 minutes to complete purification, and product 030 (7.04 mg) was obtained.
  • 031-e 500 mg, 0.84 mmol was added to trifluoroacetic acid (3.0 mL) at 25°C, and the reaction solution was stirred at 25°C for 30 minutes.
  • the reaction solution was concentrated under reduced pressure, and lithium hydroxide monohydrate (157 mg, 3.7 mmol of tetrahydrofuran (3.0 mL)/water (1.0 mL) solution was added, and the reaction solution was stirred at 25° C. for 1 hour.
  • the reaction solution was concentrated under reduced pressure , Purified by Prep-TLC (Flash) to obtain 031-f (90 mg, yield: 82%).
  • reaction solution was concentrated under reduced pressure and purified by preparation (Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN Run for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 15 minutes, and 10% ACN runs to 20 minutes to complete purification) to obtain 031 (85.0 mg, yield 33%).
  • preparation Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN Run for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 15 minutes, and 10% ACN runs to 20 minutes to complete purification
  • reaction solution was poured into ice In water (5.0mL), the mixture was concentrated under reduced pressure and then preparatively purified (Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um) , Gradient: 10% ACN runs for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 15 minutes, and 10% ACN runs to 20 minutes to complete purification) to obtain 032 (6.28 mg, yield: 6.3%).
  • Step 2 (R)-3-((2-((1-ethyl-1H-pyrazol-4-yl)amino)-7-((2-(trimethylsilyl)ethoxy )Methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)pyrrolidine-1-carboxylate 034-d synthesis
  • Step 2 Benzyl 7-((2-chloro-7-((2-(trimethylsilyl)ethoxy)methyl-pyrrolo[2,3-d]pyrimidin-4-yl) (Oxy)-5-azaspiro[2.4]heptane-5-carboxylate 035-f
  • the third step benzyl 7-((2-((1-ethyl-1H-pyrazol-4-yl)amino-7-((2-(trimethylsilyl)ethoxy)methyl -Pyrrolo[2,3-d]pyrimidin-4-yl)oxy)-5-azaspiro[2.4]heptane-5-carboxylate 035-h
  • the fourth step 4-((5-azaspiro[2.4]heptane-7-yl)amino)-N-((1-ethyl-1H-pyrazol-4-yl)amino)-7(( Synthesis of 2-(trimethylsilyl)ethoxy)methyl-7H pyrrolo[2,3-d]pyrimidine-2-amino 035-i
  • the fifth step 4-((5-azaspiro[2.4]heptane-7-yl)oxy)-N-((1-ethyl-1H-pyrazol-4-yl)amino)-7Hpyrrole Synthesis of and [2,3-d]pyrimidin-2-amine 035-j
  • the sixth step 1-(7-((-2-((1-ethyl-1H-pyrazol-4-yl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl) (Oxy)-5-azaspiro[2.4]heptane-5-yl)prop-2-en-1-one 035
  • the filtrate is prepared by Waters 2767 chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, Column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN for 3 min, 30%-35% ACN for 13 min, 95% ACN for 15 min, 10% ACN for 20 min to complete the purification, and the compound was obtained 035, compound 035 was resolved by SFC [column: DAICEL CHIRALCEL OD (250mm*30mm, 10um); mobile phase: A: CO 2 ; B: MeOH (0.1% DEA), 30%-30%, 20min] to obtain 035- 1 (23.55 mg, yield: 22%) and 035-2 (21.68 mg, yield: 20%).
  • the third step 5-chloro-N-(1-methyl-1H-pyrazol-4-yl)-4-((((3R,5S)-5-methylpyrrolidin-3-yl)oxy )-7H-pyrrolo[2,3-d]pyrimidin-2-amine 036-f
  • Dissolve 036-f 150 mg, 0.43 mmol
  • a mixture of tetrahydrofuran 2.5 mL
  • water 0.5 mL
  • potassium phosphate 182 mg, 0.86 mmol
  • a solution of acryloyl chloride 39mg, 0.43mmol
  • tetrahydrofuran 0.5mL was added dropwise and stirred at 25°C for 1 hour.
  • reaction solution was concentrated under reduced pressure and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN running for 4 minutes , 30%-35% ACN run to 13min, 95% ACN run to 16min, 10% ACN run to 19min to complete purification to obtain 036 (22.3mg, yield: 9%).
  • the fifth step 5-chloro-N-(1-(difluoromethyl)-1H-pyrazol-4-yl)-4-(((((3R,5S)-5-methylpyrrolidine-3 -Yl)oxy)-7H-pyrrolo[2,3-d]pyrimidin-2-amine 037-f
  • reaction solution was prepared by Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, chromatographic column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN for 3 min, 30%- 35% ACN was run for 11 minutes, 95% ACN was run for 14 minutes, and 10% ACN was run for 19 minutes to complete purification to obtain 037 (38.63 mg, yield 38%).
  • reaction solution was poured into ice water (10.0mL), the reaction solution was concentrated under reduced pressure, and the residue was subjected to Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18) OBD 19*250mm 10um), gradient: 10% ACN runs for 3 minutes, 30%-35% ACN runs to 13 minutes, 95% ACN runs to 15 minutes, and 10% ACN runs to 19 minutes to complete purification.
  • Intermediate 038-b (125mg, Yield: 41.2%).
  • Step 2 (2S,4R)-4-((2-((1-ethyl-1H-pyrazol-4-yl)amino)-7-((2-(trimethylsilyl)ethoxy (Yl)methyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)-2-methyltetrahydropyrrole-1-carboxylic acid tert-butyl ester 038-d
  • reaction solution was cooled to room temperature, concentrated under reduced pressure, and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18OBD 19*250mm 10um), gradient: 10% ACN Run for 3 minutes, 30%-35% ACN runs to 12 minutes, 95% ACN runs to 16 minutes, and 10% ACN runs to 20 minutes to complete the purification of intermediate 038-d (115 mg, yield: 99.5%).
  • reaction solution was concentrated under reduced pressure, and the residue was subjected to Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ TFA aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN operation For 3 minutes, 30%-35% ACN was run to 12 minutes, 95% ACN was run to 14 minutes, and 10% ACN was run to 20 minutes to complete purification to obtain intermediate 038-e (65 mg, yield: 96.5%).
  • reaction solution was concentrated under reduced pressure and passed on a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, chromatographic column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN running for 3 minutes, 30%-35% ACN was run for 12 minutes, 95% ACN was run for 16 minutes, and 10% ACN was run for 19 minutes to complete purification to obtain 038 (28.1 mg, yield: 40.2%).
  • reaction solution is cooled and concentrated, and the residue is prepared and purified (Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN was run for 3 min, 30%-35% ACN was run to 13 min, 95% ACN was run to 15 min, and 10% ACN was run to 20 min to complete purification) to obtain intermediate 039-e (300 mg, 82.2%).
  • the first step 5-bromo-2,4-dichloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine 040 -b synthesis
  • 040-b 150mg, 0.38mmol
  • 040-c 105mg, 0.42mmol
  • potassium tert-butoxide 64mg, 0.57mmol
  • the reaction solution was stirred at 25°C for 30 minutes .
  • the fifth step 2-((1-methyl-1H-pyrazol-4-yl)amino)-4-((((3R,5S)-5-methylpyrrolidin-3-yl)oxy) Synthesis of -7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile 040-h
  • the sixth step 4-(((3R,5S)-1-acryloyl-5-methylpyrrolidin-3-yl)oxy)-2-((1-methyl-1H-pyrazole-4- (Yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile 040 synthesis
  • reaction solution was concentrated under reduced pressure and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ ammonia solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN for 3min , 30%-35% ACN run to 13min, 95% ACN run to 15min, 10% ACN run to 20min to complete purification to obtain 040 (2.01mg, yield 3%).
  • Dissolve 041-f (340mg, 1.0mmol) in a mixture of tetrahydrofuran (10.0mL) and water (2.0mL), add potassium phosphate (424mg, 2.0mmol) and stir, add acryloyl chloride (110mg, 1.2mmol) in tetrahydrofuran (1.0mL) The solution was added dropwise and stirred at 0°C for 1 hour.
  • reaction solution was concentrated under reduced pressure and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN for 3min , 30%-35% ACN run to 13min, 95% ACN run to 15min, 10% ACN run to 20min to complete purification to obtain 041 (55.8mg, yield: 14.1%).
  • reaction solution was concentrated under reduced pressure and passed through a Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, chromatographic column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN running for 3 minutes, 30%-35% ACN was run for 12 minutes, 95% ACN was run for 15 minutes, and 10% ACN was run for 18 minutes to complete purification to obtain intermediate 042-f (55 mg, yield: 27%).
  • mobile phase A 1 ⁇ formic acid aqueous solution
  • mobile phase B acetonitrile
  • chromatographic column Sunfire (Prep C18 OBD 19*250mm 10um)
  • gradient 10% ACN running for 3 minutes, 30%-35% ACN was run for 12 minutes, 95% ACN was run for 15 minutes, and 10% ACN was run for 18 minutes to complete purification to obtain intermediate 042-f (55 mg, yield: 27%).
  • reaction solution was concentrated under reduced pressure and purified by preparation (Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN Run for 3 minutes, 30%-35% ACN runs to 10 minutes, 95% ACN runs to 13 minutes, and 10% ACN runs to 20 minutes to complete purification) to obtain product 042 (4.1 mg, yield: 6.8%).
  • preparation Waters 2767 preparative chromatograph, mobile phase A: 1 ⁇ formic acid aqueous solution, mobile phase B: acetonitrile, column: Sunfire (Prep C18 OBD 19*250mm 10um), gradient: 10% ACN Run for 3 minutes, 30%-35% ACN runs to 10 minutes, 95% ACN runs to 13 minutes, and 10% ACN runs to 20 minutes to complete purification
  • potassium tert-butoxide 143.1mg, 1.275mmol was added to tert-butyl(2S,4R)-4-hydroxy-2-methyltetrahydropyrrole-1-carboxylate 017-a(171.2 mg, 0.850 mmol) in a tetrahydrofuran solution (10 mL) and stirred at room temperature for 30 minutes. Then 2,4,5-trichloro-7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-d]pyrimidine 011-1-b( 300 mg, 0.850 mmol) was added to the reaction solution, and the reaction was stirred at 25°C for 1 hour.
  • reaction solution was concentrated to dryness under reduced pressure, methanol (5 mL) was added to the residue to dissolve, cooled to 0°C, DIEA (1 mL) was added, and the reaction was stirred at 30°C for 2 hours. Ice water (1 mL) was added to the reaction solution to quench the reaction, the reaction solution was concentrated to dryness under reduced pressure, and the residue was purified by reverse-phase Flash (acetonitrile/0.1% TFA) to obtain 5-chloro-N-(1-ethyl) -1H-pyrazol-4-yl)-4-(((3R,5S)-5-methyltetrahydropyrrol-3-yl)oxy)-7H-pyrrolo[2,3-d]pyrimidine- 2-amine 017-e (125 mg).

Abstract

本申请提供了式(I)所示的嘧啶并五元环类化合物或其药学可接受的盐、药物组合物及其制备方法,以及作为JAK3和/或BTK抑制剂的用途。

Description

嘧啶并五元环类衍生物及其应用
本申请要求2020年1月21日向中国国家知识产权局提交的,专利申请号为202010072567.5,发明名称为“嘧啶并五元环类衍生物及其应用”以及2020年4月7日向中国国家知识产权局提交的,专利申请号为202010263801.2,发明名称为“嘧啶并五元环类衍生物及其应用”的两件在先申请的优先权。所述两件申请的全文通过引用的方式结合于本申请中。
技术领域
本发明涉及一种新型的嘧啶并五元环类衍生物或其药学可接受的盐,含有它们的药物组合物以及其在预防或者治疗激酶相关性疾病如Janus激酶(JAK,特别是JAK3)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病中的用途。
背景技术
自身免疫病是由免疫功能异常引起对自身细胞或组织的攻击,导致炎症和组织损伤的一类疾病,包括风湿性关节炎(RA)、炎症性肠炎(IBD)和系统性红斑狼疮(SLE)等。BTK和JAK3是针对自身免疫病的两个重要靶点。
BTK是非受体型酪氨酸激酶TEC家族的一员,其结构上包括了PH结构域、TH结构域、SH3结构域、SH2结构域和SH1结构域。BTK在B细胞抗原受体(BCR)信号通路的活化过程中起着关键的作用,调控着B细胞的发育和活化,对B细胞的增殖、促炎细胞因子的表达和抗体的分泌发挥着重要的作用(Targeting Bruton's tyrosine kinase in B cell malignancies.Nat Rev Cancer.2014Apr;14(4):219-32),因此BTK成为治疗B细胞异常活化相关疾病的重要靶点之一,包括自身免疫病和B细胞淋巴瘤。Ibrutinib、Acalabrutinib和Zanubrutinib是已经获批的三个BTK抑制剂,主要治疗B细胞淋巴瘤,在部分病人中有明显疗效,但是临床上也观察到存在严重的副作用和耐药突变。2017年ibrutinib被美国FDA批准用于治疗移植物抗宿主病(GVHD),而其它的BTK抑制剂目前正在临床上积极探索治疗自身免疫疾病,包括RA、SLE和多发性硬化症(MS)。
JAK3是非受体酪氨酸激酶JAK家族的一员。JAK激酶家族有4个成员:JAK-1、JAK-2、JAK-3和TYK-2。信号转导子和转录激活子(STAT)是JAK3的下游底物,JAK3活化STAT使其成为二聚体进入细胞核内,对特定基因的转录表达进行调节。JAK-STAT信号通路对淋巴细胞增殖、分化以及促炎细胞因子的表达具有重要作用(JAK inhibition as a therapeutic strategy for immune and inflammatory diseases.Nat Rev Drug Discov.2017 December 28;17(1):78;The JAK-STAT Pathway:Impact on Human Disease and Therapeutic Intervention.Annual Review of Medicine.Vol.66:311-328),因此JAK3成为自身免疫病及恶性肿瘤的靶点之一。Tofacitinib是FDA批准的JAK3抑制剂,其在RA和IBD上展现出良好的临床疗效。但也有一定的不良反应,包括严重的感染、肝损伤等,这被认为与Tofacitinib对JAK1/2选择性不足有关(JAK inhibition as a therapeutic strategy for immune and inflammatory diseases.Nat Rev Drug Discov.2017 December 28;17(1):78;JAK-inhibitors.New players in the field of immune-mediated diseases,beyond rheumatoid arthritis.Rheumatology(Oxford).2019 Feb 1;58(Suppl 1):i43-i54)。
除了BTK和JAK3抑制剂各自单独的临床作用,同时抑制BTK/JAK3信号通路则会表现出协同的疗效。数个研究表明,在胶原蛋白诱导的大鼠关节炎模型(CIA)中同时抑制BTK和JAK,观察到关节肿胀有明显缓解,破骨细胞数量减少,病理评分也显著改善,疗效优于单药作用(2016 ACR/ARHP Annual Meeting.Abstract 484;2013 ACR/ARHP Annual Meeting.Abstract 2353)。Abbvie公司于2018年9月和2019年6月分别启动了ABBV599(BTK抑制剂和JAK抑制剂联用)针对RA和SLE的临床二期实验。另一针对BTK/JAK3双靶点抑制剂DWP212525也在小鼠CIA模型中展现出了对疾病的缓解和对关节的保护作用(2019 ACR/ARHP Annual Meeting.Abstract 965)。
鉴于庞大的自身免疫病市场以及未被满足的市场需求,基于BTK和JAK3在自身免疫病上的功能以及已有的临床效果,有必要开发针对BTK和JAK3具有良好活性,且选择性好、毒副作用低的双靶点小分子抑制剂。
发明内容
本发明提供一种式(I)所示化合物或其药学上可接受的盐:
Figure PCTCN2021073098-appb-000001
其中,
R 1选自H、
Figure PCTCN2021073098-appb-000002
或任选被R a1取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
R 2选自氢、F、Cl、Br、I、CN、OH、NO 2
Figure PCTCN2021073098-appb-000003
或任选被R a2取代的以下基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
R 5、R 6、R 7独立地选自氢、F、Cl、Br、I、CN或任选被R a3取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
X 1、X 2、X 3独立地选自CR 8或N;
R 8选自H、F、Cl、Br、I、CN、OH或任选被R a4取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
R 9、R 10、R 12独立地选自任选被R a5取代的以下基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
R 11选自H或任选被R a6取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
每一个R a1、R a2、R a3、R a4独立选自F、Cl、Br、I、OH、CN、=O、NO 2
Figure PCTCN2021073098-appb-000004
Figure PCTCN2021073098-appb-000005
硼酸基、硼酸酯基或任选被R b取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
每一个R a5、R a6独立选自F、Cl、Br、I、OH、CN、=O、NO 2或任选被R b取代的下列基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
每一个R b独立选自F、Cl、Br、I、OH、CN、=O、NO 2或任选被R c取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
每一个R c独立选自F、Cl、Br、I、OH、CN、=O、NO 2、NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
L选自
Figure PCTCN2021073098-appb-000006
R 3、R 4独立地选自H、F、Cl、Br、I、OH、CN、=O、NO 2或任选被R c取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基;
Y、Z独立选自NH、O、S、
Figure PCTCN2021073098-appb-000007
m、n1、n2、n3、q1、q2独立选自0、1或2,p1选自1、2、3或4;
条件是,当Y为NH,且当R 3为H、F、二氟甲基或三氟甲基时,R 1不为甲基;当Y为O,且当R 3、R 4均为H时,R 1不为甲基;当L选自
Figure PCTCN2021073098-appb-000008
时,R 2不为H、CN、
Figure PCTCN2021073098-appb-000009
在一些实施方案中,当L选自
Figure PCTCN2021073098-appb-000010
时,L中的NH与
Figure PCTCN2021073098-appb-000011
相连。
在一些实施方案中,当L选自
Figure PCTCN2021073098-appb-000012
且Y、Z不为NH时,L中的环N原子与
Figure PCTCN2021073098-appb-000013
相连。
在一些实施方案中,R a1选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基。
在一些实施方案中,R a1选自F、Cl、Br、I或OH。
在一些实施方案中,R a2选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基。
在一些实施方案中,R a3选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基。
在一些实施方案中,R a3选自F、Cl、Br、I。
在一些实施方案中,R a4选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基。
在一些实施方案中,R a5选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基。
在一些实施方案中,R a6选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基。
在一些实施方案中,R b选自F、Cl、Br、I、OH、CN、=O或任选被R c取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基。
在一些实施方案中,R b选自任选被R c取代的下列基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基。在一些实施方案中,R c选自F、Cl、Br、I、OH、CN、=O、NH 2或C 1-C 10烷基。
在一些实施方案中,R 1选自H、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基,所述C 1-C 10烷基、C 3-C 10 环烷基或3-10元杂环基任选被R a1取代。
在一些实施方案中,R 1选自C 1-C 6烷基或5-6元杂环烷基,所述C 1-C 6烷基、5-6元杂环烷基任选被R a1取代。
在一些实施方案中,R 1选自C 1-C 3烷基或5-6元杂环烷基,所述5-6元杂环烷基含有1~2个O原子,所述C 1-C 3烷基或5-6元杂环烷基任选被R a1取代。
在一些实施方案中,R 1选自C 1-C 3烷基或5-6元杂环烷基,所述5-6元杂环烷基含有1~2个O原子。
在一些实施方案中,R 1选自甲基、乙基、羟乙基、氧杂环丁烷基或四氢呋喃基。
在一些实施方案中,R 1选自甲基、乙基
Figure PCTCN2021073098-appb-000014
在一些实施方案中,R 1选自甲基、乙基或
Figure PCTCN2021073098-appb-000015
在一些实施方案中,R 1选自乙基。
在一些实施方案中,R 2选自H、F、Cl、Br、I、CN、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基,所述NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基任选被R a2取代。
在一些实施方案中,R 2选自H、F、Cl、Br、I、CN、C 1-C 6烷基、COOH或CONH 2,所述C 1-C 6烷基、COOH或CONH 2任选被R b取代。
在一些实施方案中,R 2选自H、F、Cl、Br或I。
在一些实施方案中,R 2选自H、F或Cl。
在一些实施方案中,R 2选自H或Cl。
在一些实施方案中,R 3、R 4独立地选自H、F、Cl、Br、I、OH、CN或任选被R c取代的下列基团:NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基。
在一些实施方案中,R 3、R 4独立地选自H、F、Cl、Br、I、OH、CN或任选被R c取代的C 1-C 6烷基。
在一些实施方案中,R 3、R 4独立地选自H、F、Cl、Br、I、OH或甲基。
在一些实施方案中,R 3、R 4独立地选自H、F、Cl、Br、I或甲基。
在一些实施方案中,R 5、R 6、R 7独立地选自H、F、Cl、Br、I、CN或任选被R a3取代的C 1-C 6烷基。
在一些实施方案中,R 5、R 6、R 7独立地选自H、F、Cl、Br、I、CN或任选被R a3取代的甲基、乙基或叔丁基。
在一些实施方案中,R 5、R 6、R 7独立地选自H、F、Cl、Br、I、CN、甲基或乙基。
在一些实施方案中,R 5、R 6、R 7独立地选自H、F、Cl、Br、I、CN、CH 2F或叔丁基。
在一些实施方案中,R 5、R 6、R 7独立地选自H、F或CN。
在一些实施方案中,R 5选自H、F或CN。
在一些实施方案中,R 5选自H或F。
在一些实施方案中,R 6、R 7选自H、CH 2F或叔丁基。
在一些实施方案中,R 6、R 7选自H。
在一些实施方案中,R 8选自H、F、Cl、Br、I、CN或任选被R a4取代的C 1-C 6烷基。
在一些实施方案中,R 8选自H、F、Cl、Br、I、CN、甲基或乙基。
在一些实施方案中,R 8选自H。
在一些实施方案中,X 1、X 2、X 3独立地选自CH或N。
在一些实施方案中,X 2选自N。
在一些实施方案中,X 3选自CH。
在一些实施方案中,R 9、R 10、R 12独立地选自任选被R a5取代的以下基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基、苯基或5-6元杂芳基。
在一些实施方案中,R 11选自H或任选被R a6取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基、苯基或5-6元杂芳基。
在一些实施方案中,m、n1、n2、n3、q2独立选自0或1,和/或p1、q1选自1或2。
在一些实施方案中,n1、n2、n3独立选自0或1,和/或q1选自1或2,和/或m、p1独立选自1,和/或q2选自0。
在一些实施方案中,q1、p1独立选自1,q2选自0。
在一些实施方案中,Y、Z独立选自NH或O。
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000016
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000017
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000018
且L中的NH与
Figure PCTCN2021073098-appb-000019
相连。
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000020
时,L中的NH与
Figure PCTCN2021073098-appb-000021
相连。
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000022
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000023
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000024
Figure PCTCN2021073098-appb-000025
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000026
Figure PCTCN2021073098-appb-000027
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000028
Figure PCTCN2021073098-appb-000029
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000030
Figure PCTCN2021073098-appb-000031
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000032
Figure PCTCN2021073098-appb-000033
在一些实施方案中,L选自
Figure PCTCN2021073098-appb-000034
Figure PCTCN2021073098-appb-000035
在一些实施方案中L选自
Figure PCTCN2021073098-appb-000036
Figure PCTCN2021073098-appb-000037
在一些实施方案中L选自
Figure PCTCN2021073098-appb-000038
Figure PCTCN2021073098-appb-000039
在一些实施方案中L选自
Figure PCTCN2021073098-appb-000040
Figure PCTCN2021073098-appb-000041
在一些实施方案中L选自
Figure PCTCN2021073098-appb-000042
Figure PCTCN2021073098-appb-000043
在一些实施方案中L选自
Figure PCTCN2021073098-appb-000044
Figure PCTCN2021073098-appb-000045
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自式(II)所示化合物或其药学可接受的盐:
Figure PCTCN2021073098-appb-000046
其中,R 1、R 2、R 5、X 1、L如上文定义。
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自式(III)所示化合物或其药学可接受的盐:
Figure PCTCN2021073098-appb-000047
其中,R 1、R 2、X 1、L如上文定义。
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自以下化合物或其药学可接受的盐:
Figure PCTCN2021073098-appb-000048
Figure PCTCN2021073098-appb-000049
Figure PCTCN2021073098-appb-000050
Figure PCTCN2021073098-appb-000051
Figure PCTCN2021073098-appb-000052
本发明还提供药物组合物,其包含式(I)所示化合物或其药学可接受的盐和药学上可接受的辅料。
本发明还提供药物组合物,其包含式(I)所示化合物或其药学可接受的盐、药学可接受的载体和/或赋形剂。
进一步,本发明涉及式(I)所示的化合物或其药学上可接受的盐,或其药物组合物在制备预防或者治疗Janus激酶(JAK,特别是JAK3)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病的药物中的用途。
进一步,本发明涉及式(I)所示的化合物或其药学上可接受的盐,或其药物组合物在预防或者治疗Janus激酶(JAK,特别是JAK3)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病中的用途。
进一步,本发明涉及预防或者治疗Janus激酶(JAK,特别是JAK3)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病的式(I)化合物或其药学上可接受的盐,或其药物组合物。
本发明还涉及治疗Janus激酶(JAK,特别是JAK3)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病的方法,该方法包括给以患者治疗上有效剂量的包含本发明所述的式(I)化合物或其药学上可接受的盐的药物制剂。
本发明的优选方案,其中所述的Janus激酶(JAK)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病包括但不限于肿瘤(如B细胞淋巴瘤)和自身免疫病(如风湿性关节炎、炎症性肠炎和系统性红斑狼疮)等。
术语定义和说明
除非另有说明,本发明说明书和权利要求书中记载的基团和术语定义,包括其作为实例的定义、示例性的定义、优选的定义、表格中记载的定义、实施例中具体化合物的定义等,可以彼此之间任意组合和结合。这样的组合和结合后的基团定义及化合物结构,应当属于本发明说明书记载的范围内。
本发明中
Figure PCTCN2021073098-appb-000053
处表示连接位点。
术语“药学上可接受的盐”是指药学上可接受的无毒酸或碱的盐,包括无机酸和碱、有机酸和碱的盐。
术语“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体、非对应异构体和构象异构体。
本发明的化合物可以具有不对称原子如碳原子、硫原子、氮原子、磷原子(光学中心)或不对称双键。外消旋体、对映异构体、非对映异构体、几何异构体和单个的异构体都包括在本发明的范围之内。
本文中消旋体或者对映体纯的化合物的图示法来自Maehr,J.Chem.Ed.1985,62:114-120。除非另有说明,用楔形键和虚楔键
Figure PCTCN2021073098-appb-000054
表示一个立体中心的绝对构型,用黑实键和虚键
Figure PCTCN2021073098-appb-000055
表示脂 环化合物的顺反构型。当本文所述化合物含有烯属双键或其它几何不对称中心,除非另有规定,它们包括E、Z几何异构体。同样地,所有的互变异构形式均包括在本发明的范围之内。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子、不对称硫原子、不对称氮原子或不对称磷原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。本申请的含有不对称原子的化合物可以以光学活性纯的形式或外消旋形式被分离出来。光学活性纯的形式可以从外消旋混合物拆分,或通过使用手性原料或手性试剂合成。立体异构体的非限制性实例包括但不限于:
Figure PCTCN2021073098-appb-000056
术语“互变异构体”是指因分子中某一原子在两个位置迅速移动而产生的官能团异构体。本发明化合物可表现出互变异构现象。互变异构的化合物可以存在两种或多种可相互转化的种类。质子移变互变异构体来自两个原子之间共价键合的氢原子的迁移。互变异构体一般以平衡形式存在,尝试分离单一互变异构体时通常产生一种混合物,其理化性质与化合物的混合物是一致的。平衡的位置取决于分子内的化学特性。例如,在很多脂族醛和酮如乙醛中,酮型占优势;而在酚中,烯醇型占优势。本发明包含化合物的所有互变异构形式。
术语“药物组合物”表示一种或多种文本所述化合物或其生理学/药学上可接受的盐或前体药物与其它化学组分的混合物,其它组分例如生理学/药学上可接受的载体和赋形剂。药物组合物的目的是促进化合物对生物体的给药。
术语“被取代”是指特定原子上的任意一个或多个氢原子被取代基取代,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧代(即=O)时,意味着两个氢原子被取代,氧代不会发生在芳香基上。
术语“任选”或“任选地”是指随后描述的事件或情况可以发生或不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。例如,乙基“任选”被卤素取代,指乙基可以是未被取代的(CH 2CH 3)、单取代的(如CH 2CH 2F)、多取代的(如CHFCH 2F、CH 2CHF 2等)或完全被取代的(CF 2CF 3)。本领域技术人员可理解,对于包含一个或多个取代基的任何基团,不会引入任何在空间上不可能存在和/或不能合成的取代或取代模式。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。
术语“卤”或“卤素”是指氟、氯、溴和碘。
术语“C 1-C 10烷基”应理解为表示具有1、2、3、4、5、6、7、8、9或10个碳原子的直链或支链饱和一价烃基。所述烷基是例如甲基、乙基、丙基、丁基、戊基、己基、异丙基、异丁基、仲丁基、叔丁基、异戊基、2-甲基丁基、1-甲基丁基、1-乙基丙基、1,2-二甲基丙基、新戊基、1,1-二甲基丙基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、2-乙基丁基、1-乙基丁基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、2,3-二甲基丁基、1,3-二甲基丁基或1,2-二甲基丁基等;“C 1-C 6烷基”应理解为表示具有1、2、3、4、5、6个碳原子的直链或支链饱和一价烃基。“C 1-C 3烷基”应理解为表示具有1、2、3个碳原子的直链或支链饱和一价烃基。
术语“烷氧基”可理解为“烷基氧基”或“烷基-O”,优选地,“C 1-C 10烷氧基”可以包含“C 1-C 6烷氧基”和“C 1-C 3烷氧基”。
术语“C 2-C 10烯基”应理解为优选表示直链或支链的一价烃基,其包含一个或多个双键并且具有2、3、4、5、6、7、8、9、10个碳原子,优选“C 2-C 6烯基”,进一步优选“C 2-C 4烯基”,更进一步优选C 2或C 3烯基。应理解,在所述烯基包含多于一个双键的情况下,所述双键可相互分离或共轭。所述烯基例如乙烯基、烯丙基、(E)-2-甲基乙烯基、(Z)-2-甲基乙烯基、(E)-丁-2-烯基、(Z)-丁-2-烯基、(E)-丁-1-烯基、(Z)-丁-1-烯基、异丙烯基、2-甲基丙-2-烯基、1-甲基丙-2-烯基、2-甲基丙-1-烯基、(E)-1-甲基丙-1-烯基、(Z)-1-甲基丙-1-烯基。
术语“C 2-C 10炔基”应理解为表示直链或支链的一价烃基,其包含一个或多个三键并且具有2、3、4、5、6、7、8、9、10个碳原子,优选“C 2-C 6炔基”,进一步优选“C 2-C 4炔基”,更进一步优选C 2或C 3炔基。所述炔基是例如乙炔基、丙-1-炔基、丙-2-炔基、丁-1-炔基、丁-2-炔基、丁-3-炔基、1-甲基丙-2-炔基。
术语“C 3-C 10环烷基”应理解为表示饱和的一价单环或双环烃环,其具有3~10个碳原子。如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基或环癸基,或者是双环烃基如十氢化萘环。术语“C 3-C 6环烷基”应理解为表示饱和的一价单环或双环烃环,其具有3~6个碳原子。优选地,“C 3-C 10环烷基”可以包含“C 3-C 6环烷基”。
术语“环烷基氧基”可理解为“环烷基-O”,优选地,“C 3-C 10环烷基氧基”可以包含“C 3-C 6环烷基氧基”。
术语“杂环基”,是指单环杂环基、稠杂环基系统;稠杂环包括并杂环基、螺杂环基、桥杂环基,可以是饱和的、部分饱和的或不饱和的,但不是芳香性的;所述并杂环基、螺杂环基、桥杂环基包括但不限于6-18元并杂环基、5-18元螺杂环基或5-18元桥杂环基,所述并杂环基、螺杂环基、桥杂环基可含有1~5个杂原子,所述杂原子选自N、O、B和S。
术语“3-10元杂环基”意指饱和的或部分饱和的一价单环、并环、螺环或桥环,其包含1-5个,优选1-3个选自N、O、B和S的杂原子。特别地,所述杂环基可以包括但不限于:4元环,如氮杂环丁烷基、氧杂环丁烷基;5元环,如四氢呋喃基、二氧杂环戊烯基、吡咯烷基、咪唑烷基、吡唑烷基、吡咯啉基;或6元环,如四氢吡喃基、哌啶基、吗啉基、二噻烷基、硫代吗啉基、哌嗪基或三噻烷基;或部分饱和的6元环如四氢吡啶基;或7元环,如二氮杂环庚烷基。任选地,所述杂环基可以是苯并稠合的。所述杂环基可以是双环的,例如但不限于5,5元环,如六氢环戊并[c]吡咯-2(1H)-基环,或者5,6元双环,如六氢吡咯并[1,2-a]吡嗪-2(1H)-基环。含氮原子的环可以是部分不饱和的,即它可以包含一个或多个双键,例如但不限于2,5-二氢-1H-吡咯基、4H-[1,3,4]噻二嗪基、4,5-二氢噁唑基或4H-[1,4]噻嗪基,或者,它可以是苯并稠合的,例如但不限于二氢异喹啉基。任选地,所述3-10元杂环基可以是“3-10元杂环烷基”,意指饱和的含1-5个杂原子的一价单环、并环、螺环或桥环;优选地,“3-10元杂环烷基”包括5-6元杂环烷基;根据本发明,所述杂环基是无芳香性的。
术语“杂环基氧基”可理解为“杂环基-O”,优选地,“3-10元杂环基氧基”可以包含“3-6元杂环基氧基”。
术语“C 6-C 10芳基”应理解为优选表示具有6、7、8、9、10个碳原子的一价芳香性或部分芳香性的单环或双环烃环。特别是具有6个碳原子的环(“C 6芳基”),例如苯基;或者具有9个碳原子的环(“C 9芳基”),例如茚满基或茚基,或者具有10个碳原子的环(“C 10芳基”),例如四氢化萘基、二氢萘基或萘基。
术语“C 6-C 10芳基氧基”可理解为“C 6-C 10芳基-O”。
“5-10元杂芳基”应理解为包括这样的一价单环、双环或三环芳族环系:其具有5、6、7、8、9、10个环原子,特别是5或6或9或10个环原子,且其包含1-5个,优选1-3个独立选自N、O和S的杂原子并且,另外在每一种情况下可为苯并稠和的。特别地,杂芳基选自噻吩基、呋喃基、吡咯基、噁唑基、噻唑基、咪唑基、吡唑基、异噁唑基、异噻唑基、噁二唑基、三唑基、噻二唑基等以及它们的苯并衍生物,例如苯并呋喃基、苯并噻吩基、苯并噻唑基、苯并噁唑基、苯并异噁唑基、苯并咪唑基、苯并三唑基、吲唑基、吲哚基、异吲哚基等;或吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基等,以及它们的苯并衍生物,例如喹啉基、喹唑啉基、异喹啉基等;或吖辛因基、吲嗪基、嘌呤基等以及它们的苯并衍生物;或噌啉基、酞嗪基、喹唑啉基、喹喔啉基、萘啶基、蝶啶基等。
术语“5-10元杂芳基氧基”可理解为“5-10元杂芳基-O”。
术语“治疗”意为将本申请所述化合物或制剂进行给药以预防、改善或消除疾病或与所述疾病相关的一 个或多个症状,且包括:
(i)预防疾病或疾病状态在哺乳动物中出现,特别是当这类哺乳动物易患有该疾病状态,但尚未被诊断为已患有该疾病状态时;
(ii)抑制疾病或疾病状态,即遏制其发展;
(iii)缓解疾病或疾病状态,即使该疾病或疾病状态消退。
术语“治疗有效量”意指(i)治疗或预防特定疾病、病况或障碍,(ii)减轻、改善或消除特定疾病、病况或障碍的一种或多种症状,或(iii)预防或延迟本文中所述的特定疾病、病况或障碍的一种或多种症状发作的本发明化合物的用量。构成“治疗有效量”的本发明化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。
术语“辅料”是指可药用惰性成分。术语“赋形剂”的种类实例非限制性地包括粘合剂、崩解剂、润滑剂、助流剂、稳定剂、填充剂和稀释剂等。赋形剂能增强药物制剂的操作特性,即通过增加流动性和/或粘着性使制剂更适于直接压缩。适用于上述制剂的典型的“药学上可接受的载体”的实例为:糖类,淀粉类,纤维素及其衍生物等在药物制剂中常用到的辅料。
术语“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。合适的辅料是本领域技术人员熟知的,例如碳水化合物、蜡、水溶性和/或水可膨胀的聚合物、亲水性或疏水性材料、明胶、油、溶剂、水等。
词语“包括(comprise)”、“含有(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应理解为开放的、非排他性的意义,即“包括但不限于”。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本申请还包括与本文中记载的那些相同的,但一个或多个原子被原子量或质量数不同于自然中通常发现的原子量或质量数的原子置换的同位素标记的本申请化合物。可结合到本申请化合物的同位素的实例包括氢、碳、氮、氧、磷、硫、氟、碘和氯的同位素,诸如分别为 2H、 3H、 11C、 13C、 14C、 13N、 15N、 15O、 17O、 18O、 31P、 32P、 35S、 18F、 123I、 125I和 36Cl等。
某些同位素标记的本申请化合物(例如用 3H及 14C标记的那些)可用于化合物和/或底物组织分布分析中。氚化(即 3H)和碳-14(即 14C)同位素对于由于它们易于制备和可检测性是尤其优选的。正电子发射同位素,诸如 15O、 13N、 11C和 18F可用于正电子发射断层扫描(PET)研究以测定底物占有率。通常可以通过与公开于下文的方案和/或实施例中的那些类似的下列程序,通过同位素标记试剂取代未经同位素标记的试剂来制备同位素标记的本申请化合物。
此外,用较重同位素(诸如氘(即 2H))取代可以提供某些由更高的代谢稳定性产生的治疗优点(例如增加的体内半衰期或降低的剂量需求),并且因此在某些情形下可能是优选的,其中氘取代可以是部分或完全的,部分氘取代是指至少一个氢被至少一个氘取代。
本发明的化合物的所有的同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
本申请的药物组合物可通过将本申请的化合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本申请化合物或其药学上可接受的盐或其药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。
本申请的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等。
在一些实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本申请的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。
可以通过常规的混合、填充或压片方法来制备固体口服组合物。例如,可通过下述方法获得:将所述 的活性化合物与固体辅料混合,任选地碾磨所得的混合物,如果需要则加入其它合适的辅料,然后将该混合物加工成颗粒,得到了片剂或糖衣剂的核心。适合的辅料包括但不限于:粘合剂、稀释剂、崩解剂、润滑剂、助流剂、甜味剂或矫味剂等。
药物组合物还可适用于肠胃外给药,如合适的单位剂型的无菌溶液剂、混悬剂或冻干产品。
本文所述的通式Ⅰ化合物的所有施用方法中,每天给药的剂量为0.01到100mg/kg体重,优选为0.05到50mg/kg体重,更优选0.1到30mg/kg体重,以单独或分开剂量的形式。
本发明具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本发明的化学变化及其所需的试剂和物料。为了获得本发明的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。
附图说明
图1为各组小鼠的临床关节炎评分随时间变化的曲线。
图2为各组小鼠的临床关节炎评分随时间变化的曲线下面积柱状图。
图3为各组小鼠的关节病理代表性切片图。
图4为各组小鼠的关节病理评分柱状图。
具体实施方式
以下实施例详细说明发明的技术方案,但本发明的保护范围包括但不限于此。
本发明所使用的溶剂可经市售获得。市售化合物采用供应商目录名称。
化合物的结构是通过核磁共振(NMR)和/或质谱(MS)来确定的。NMR位移的单位为10 -6(ppm)。NMR测定的溶剂为氘代二甲基亚砜、氘代氯仿、氘代甲醇等,内标为四甲基硅烷(TMS);“IC 50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。下文的洗脱剂可由两种或多种溶剂形成混合洗脱剂,其百分比为各溶剂的体积比,如“乙酸乙酯/石油醚:0-10%”表示梯度洗脱过程中,混合洗脱剂中的乙酸乙酯:石油醚的体积用量为0:100~10:100。本发明采用以下缩略词:
Figure PCTCN2021073098-appb-000057
Figure PCTCN2021073098-appb-000058
实施例1化合物001的制备
1-丙烯酰基-3-(2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯
Figure PCTCN2021073098-appb-000059
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000060
第一步:2,4-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶(1b)的合成
0℃下将钠氢(510mg,12.76mmol,60%)加入到N,N-二甲基甲酰胺(15mL)中,然后缓慢加入2,4-二氯-7H-吡咯并[2,3-d]嘧啶1a(2g,10.63mmol)的N,N-二甲基甲酰胺(5mL)溶液。待加入完毕后,2-(三甲基硅烷基)乙氧甲基氯(2.45mL,13.83mmol)慢慢加入该反应液中,然后在氮气氛围下0℃搅拌1.5小时至反应完全。混合物倒入水中用乙酸乙酯(20.0mL*3)萃取,有机相合并后用无水硫酸钠干燥过滤,滤液减压浓缩,得到的粗品用硅胶柱纯化(PE/EtOAc=1/0~7/1)纯化得到标题产物1b(1.9g)。
LCMS:Rt:2.378min;MS m/z(ESI):318.0[M+H] +
第二步:3-(2-氯-7-((2-(三甲硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲醛(1d)的 合成
室温下将2,4-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶1b(1.7g,5.363mmol)和3-甲酰基苯硼酸1c(724mg,4.826mmol)溶于1,4-二氧六环(20mL)和水(5mL)中,加入碳酸钠(1.14g,10.726mmol)和四三苯基膦钯(619mg,0.5363mmol),反应液在氮气氛下110℃搅拌1小时至反应完全。混合物倒入水中用乙酸乙酯(20.0mL*3)萃取,有机相合并后用无水硫酸钠干燥过滤,滤液减压浓缩,得到的粗品用硅胶柱纯化(PE/EtOAc=7/1)纯化得到标题产物1d(1.7g)。
LCMS:Rt:2.293min;MS m/z(ESI):388.1[M+H] +
第三步:1-(1-羟基-2-丙烯)-3-(2-氯-7-((2-(三甲硅基)乙氧基)甲基)-7-吡咯并[2,3-d]嘧啶-4-基)苯1f的合成
将3-(2-氯-7-((2-(三甲硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯甲醛1d(500mg,1.295mmol)溶解到四氢呋喃(10mL)中,在0℃冰浴氮气氛下慢慢滴入乙烯基溴化镁溶液1e(1.94mL,1.94mmol,1M溶解在四氢呋喃中)。加完后反应液在0℃下继续反应30分钟至反应完全。TLC板(PE/EtOAc=4/1)监测反应完毕。反应液用饱和氯化铵溶液(20mL)淬灭,然后乙酸乙酯(20mL*3)萃取,合并的有机相用无水硫酸钠干燥过滤,滤液减压浓缩,得到的粗品标题产物1f(580mg),该粗品直接用于下步反应。
第四步:1-(1-羟基-2-丙烯)-3-(2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)苯1h的合成
室温下将1-(1-羟基-2-丙烯)-3-(2-氯-7-((2-(三甲硅基)乙氧基)甲基)-7-吡咯并[2,3-d]嘧啶-4-基)苯1f(580mg,1.397mmol)和1-乙基-1H-吡唑-4-胺1g(264mg,2.38mmol)溶于1,4-二氧六环(10mL)中,加入碳酸铯(1.36g,4.191mmol)、1,1'-联萘-2,2'-双二苯膦(87mg,0.140mmol)和三(二亚苄基丙酮)二钯(128mg,0.1397mmol),反应液在氮气氛下120℃搅拌6小时至反应完全。混合物直接减压浓缩,得到的粗品用硅胶柱纯化(PE/EtOAc=5/1~1/1)纯化得到1h(370mg)。
LCMS:Rt:1.829min;MS m/z(ESI):491.3[M+H] +
第五步:1-(1-羟基-2-丙烯)-3-(2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯(1i)的合成
0℃下将三氟乙酸(2mL)缓慢滴加到1-(1-羟基-2-丙烯)-3-(2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)苯1h(340mg,0.694mmol)的二氯甲烷(6mL)溶液中。反应液在室温搅拌2小时。LCMS监测反应完全。混合物直接减压浓缩,得到中间体。向该中间体中加入四氢呋喃(3mL)和2M氢氧化钠溶液(3mL)。该反应液室温搅拌1小时至反应完全。反应液用水(30mL)稀释,乙酸乙酯(30mL*2)萃取,合并的有机相无水硫酸钠干燥,过滤旋干得到粗产物1i(310mg),该粗品直接用于下一步。
LCMS:Rt:0.949min;MS m/z(ESI):361.2[M+H] +
第六步:1-丙烯酰基-3-(2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯001的合成
室温下将1-(1-羟基-2-丙烯)-3-(2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)苯1i(320mg,0.89mmol)溶于乙酸乙酯(10mL)中,加入2-碘酰基苯甲酸(497mg,1.78mmol),反应液在80℃搅拌3小时至反应完全。混合物过滤,滤饼用乙酸乙酯(20mL)洗涤,合并的滤液直接减压浓缩,得到的粗品高效液相色谱法纯化得到001(49.74mg)。
高效液相制备色谱法:Waters 2767制备色谱仪,流动相A:0.1%NH 4HCO 3水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10μm),梯度:10%ACN运行3min,65%-70%ACN运行至13min,95%ACN运行至16min,10%ACN运行至18min结束纯化,得到目标化合物。
LCMS:Rt:1.090min;MS m/z(ESI):359.0[M+H] +.
1H NMR(400MHz,DMSO)δ11.57(br s,1H),9.75(d,J=7.6Hz,1H),9.16(s,1H),8.41(s,1H),8.22(d,J=7.6Hz,1H),7.96(dd,J=15.2,19.6Hz,3H),7.69(t,J=7.6Hz,1H),7.60(s,1H),7.25(d,J=3.6Hz,1H),6.95(dd,J=7.6,16.0Hz,1H),6.70(d,J=3.6Hz,1H),4.11(q,J=7.2Hz,2H),1.39(t,J=7.2Hz,3H).
实施例2化合物002的制备
1-(8-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3,8-二氮杂双环[4.2.0]辛-3-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000061
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000062
第一步:2,5-二氯-4-(1-苯基乙氧基)-7H-吡咯并[2,3-d]嘧啶2c的合成
室温下将2,4,5-三氯-7H-吡咯并[2,3-d]嘧啶2a(3g,13.49mmol)溶解在1,4-二氧六环(45mL)中,添加2-甲基丁-2-醇钾(53mL,54mmol)和1-苯乙醇2b(1.62mL,13.49mmol)。混合物在室温下搅拌1个小时。随后用水淬灭反应,乙酸乙酯萃取,有机相被收集,干燥,浓缩得到粗产物2,5-二氯-4-(1-苯基乙氧基)-7H-吡咯并[2,3-d]嘧啶2c(4.17g)。
LCMS:Rt:2.105min;MS m/z(ESI):309.2[M+H] +
第二步:2,5-二氯-4-(1-苯基乙氧基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶2d的合成
室温下将氢化钠(1.908g,47.66mmol)加入到冷却到0℃氮气氛围下的无水四氢呋喃(60mL)中,随后添加2,5-二氯-4-(1-苯基乙氧基)-7H-吡咯并[2,3-d]嘧啶2c(3.67g,11.92mmol)。混合物在该体系下搅拌30分钟,然后(2-(氯甲氧基)乙基)三甲基硅烷(4.012g,23.83mmol)被注入。在16个小时后,用饱和的氯化铵溶液淬灭,乙酸乙酯萃取,有机相被收集,干燥,浓缩。剩余物使用薄层柱色谱法(乙酸乙酯/石油醚:0-10%)纯化得到2d(2.44g)。
LCMS:Rt:2.344min;MS m/z(ESI):438.1[M+H] +
第三步:5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(1-苯基乙氧基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-2-胺2f的合成
室温下将2d(2.14g,4.88mmol),1-乙基-1H-吡唑-4-胺2e(860mg,7.32mmol),1,1’-联萘-2,2’-二苯膦(290mg,0.49mmol),碳酸铯(4.85g,14.64mmol),三(二亚苄基丙酮)二钯(430mg,0.49mmol)和1,4-二氧六环(20mL)添加到反应瓶中,混合物在氮气,120℃的条件下反应16h。反应结束后,溶剂被旋干,剩余物利用薄层柱色谱法(乙酸乙酯/石油醚:0-40%)纯化得到2f(1.86g)。
LCMS:Rt:1.843min;MS m/z(ESI):513.2[M+H] +
第四步:(4,5-二氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-7-基)甲醇2g的合成
室温下将2f(1.86g,3.62mmol)加入到三氯氧磷(10mL)中,混合物在70℃下反应40分钟,然后在100℃下反应5小时。随后将混合物到入到温水中淬灭,用乙酸乙酯萃取,有机相被收集,干燥,浓缩。剩余物用制备薄层柱色谱法(石油醚/乙酸乙酯=1:1)得到标题产物2g(330mg)。
LCMS:Rt:1.183min;MS m/z(ESI):327.0[M+H] +
第五步:4,5-二氯-N-(1-乙基-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-胺(2h)的合成
室温下将2g(330mg,1.01mmol)加入到四氢呋喃(4mL)中,然后添加2M氢氧化钠(4mL)。在反应2小时后,加水稀释,乙酸乙酯(15.0mL*3)萃取,有机相被收集,干燥,浓缩得到标题产物2h(300mg)。
LCMS:Rt:1.533min;MS m/z(ESI):297.0[M+H] +
第六步:叔丁基8-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3,8-二氮杂双环[4.2.0]辛烷-3-羧酸酯2j的合成
室温下将叔丁酯2i(110mg,0.51mmol),二异丙基乙胺(70mg,0.51mmol)和DMSO(1mL)加到微波管中,随后在微波120℃条件下反应15分钟。反应结束后,用水稀释,乙酸乙酯萃取,有机相被收集,干燥,浓缩。剩余物用制备薄层色谱法(石油醚/乙酸乙酯=2:1)纯化得到标题产物2j(91mg)。
LCMS:Rt:1.253min;MS m/z(ESI):473.1[M+H] +
第七步:4-(3,8-二氮杂双环[4.2.0]辛烷-8-基)-5-氯-N-(1-乙基-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-胺2k的合成
室温下将叔丁基2j(91mg,0.19mmol)溶解在乙酸乙酯(0.5mL)中,然后添加3M盐酸/乙酸乙酯(1.5mL),混合物在室温条件下反应1个小时。溶剂被旋干得到粗产物2k(72mg)直接用于下一步反应。
LCMS:Rt:0.746min;MS m/z(ESI):373.3[M+H] +
第八步:1-(8-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3,8-二氮杂双环[4.2.0]辛-3-基)丙-2-烯-1-酮002的合成
室温下将2k(72mg,0.19mmol)和二异丙基乙胺(75mg,0.58mmol)溶解在二氯甲烷中(2mL),混合物被冷却到-30℃添加丙烯基酰氯(21mg,0.23mmol)并搅拌1个小时。反应结束后,溶剂被旋干。剩余物溶解在乙腈中,使用制备高效液相色谱法纯化得到产物002(2.30mg)。
高效液相制备色谱法:Waters 2767制备色谱仪,流动相A:10mmol%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10μm),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至17min,10%ACN运行至19min结束纯化,得到目标化合物。
LCMS:Rt:0.933min;MS m/z(ESI):427.1[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.30(s,1H),8.60(d,J=18.4Hz,1H),7.86(d,J=11.6Hz,1H),7.47(d,J=4.4Hz,1H),6.98–6.97(m,1H),6.81-6.74(m,1H),6.08(dd,J=16.8,2.4Hz,1H),5.67-5.41(m,1H),4.87(dd,J=30.2,8.4Hz,1H),4.45-4.22(m,3H),4.07(q,J=7.2Hz,2H),3.93–3.71(m,2H),3.63(dd,J=14.0,2.0Hz,1H),2.94(s,1H),2.11–1.86(m,2H),1.36(t,J=8.0Hz,3H).
实施例3化合物003的制备
1-(1-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1,6-二氮杂螺[3.5]壬-6-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000063
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000064
第一步:1-(2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)-1,6-二氮杂螺[3.5]壬烷-6-羧酸叔丁酯(3c)的合成
室温下将2,4,5-三氯-7H-吡咯并[2,3-d]嘧啶3a(236mg,1.06mmol),1,6-二氮杂螺[3.5]壬烷-6-羧酸叔丁酯3b(120mg,0.53mmol)溶解到异丙醇(4mL)中,随后添加二异丙基乙胺(274mg,2.12mmol),该体系在110℃,反应16小时。反应结束后,旋干溶剂,剩余物溶解在甲醇中,利用高效液相色谱法(乙腈-水:0-80%)纯化得到标题产物3c(110mg)。
LCMS:Rt:1.910min;MS m/z(ESI):412.3[M+H] +
第二步:1-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)-1,6-二氮杂螺[3.5]壬-6-羧酸叔丁酯3e的合成
室温下将3c(40mg,0.097mmol)加入到化合物1-乙基-1H-吡唑-4-胺3d(21mg,0.19mmol)和三氟乙酸(11mg,0.097mmol)的异丙醇(6.0mL)溶液中。将反应体系密封并加热到110℃在此温度下搅拌过夜。待反应体系降至室温后,将反应液在减压下浓缩得到粗产品,后经硅胶板纯化得到中间体3e(13mg)。
LCMS:Rt:1.53min;MS m/z(ESI):487.2[M+H] +
第三步:5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(1,6-二氮杂螺[3.5]壬-1-基)-7H吡咯并[2,3-d]嘧啶-2-胺3f的合成
室温下将中间体1-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)-1,6-二氮杂螺[3.5]壬-6-羧酸叔丁酯3e(13mg,0.027mmol)加入到盐酸的1,4-二氧六环(2mL,3M)溶液中(1.0mL)中。反应液在室温下搅拌直至反应完全。减压下将反应液浓缩,得到标题产物3f的盐酸盐(10mg),未经纯化直接用于下一步。
LCMS:Rt:0.810min;MS m/z(ESI):387.0[M+H] +
第四步:1-(1-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)-1,6-二氮杂螺[3.5]壬-6-基)丙-2-烯-1-酮003的合成
室温下将三乙胺(13mg,0.13mmol)加入到粗品5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(1,6-二氮杂螺[3.5]壬-1-基)-7H吡咯并[2,3-d]嘧啶-2-胺3f(10mg)盐酸盐的二氯甲烷(3mL)溶液中。所得溶液在室温下搅拌10分钟。然后将该溶液用冰水浴冷却,并缓慢滴加丙烯酰氯3g(2.4mg,0.027mmol)的四氢呋喃溶液(1mL)。反应液温度升至室温,并搅拌1小时。减压下将反应液浓缩,所得粗品用二甲基甲酰胺溶解经Waters 2767制备色谱仪,流动相A:0.1%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至10min,95%ACN运行至12min,10%ACN运行至15min结束纯化得到产物003(2.0mg)。
LCMS:Rt:1.164min;MS m/z(ESI):441.0[M+H] +
1H NMR(400MHz,CDCl3)δ7.67-7.63(m,1H),7.48-7.45(m,1H),6.68-6.35(m,1H),6.30-6.10(m,1H),5.60-5.25(m,2H),4.16-4.08(m,1H),3.92-3.84(m,1H),3.71-3.50(m,2H),2.38-2.20(m,2H),2.19-2.06(m,3H),2.05-1.98(m,2H),1.87-1.71(m,2H),1.65-1.50(m,1H),1.47-1.39(m,2H),1.36(t,J=8Hz,3H).
实施例4化合物004的制备
1-(6-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)八氢-1H-吡咯并[2,3-c]吡啶-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000065
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000066
第一步:叔丁基6-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)八氢-1H-吡咯并[2,3-c]吡啶-1-羧酸酯4c的合成
室温下将2h(80mg,0.27mmol),4b(104mg,0.81mmol)和N-乙基-N-异丙基丙-2-胺(104mg,0.81mmol)于二甲亚砜(3mL)的溶液微波加热至120℃搅拌30分钟至反应完全。反应液用20mL乙酸乙酯稀释后,20mL饱和食盐水洗一次,无水硫酸钠干燥后减压浓缩,得到的粗品用硅胶柱色谱(MeOH:DCM=0~10%)纯化得到标题产物4c(80mg)。
LCMS:Rt:1.676min;MS m/z(ESI):487.4[M+H] +
第二步:5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(八氢-6H-吡咯并[2,3-c]吡啶-6-基)-7H-吡咯并[2,3-d]嘧啶-2-胺4d的合成
将叔丁基6-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)八氢-1H-吡咯并[2,3-c]吡啶-1-羧酸酯4c(80mg,0.16mmol)加入到盐酸二氧六环溶液(4M,5mL)中,室温搅拌反应2小时。反应液减压浓缩后直接用于下一步反应(62mg)。
LCMS:Rt:1.009min;MS m/z(ESI):387.4[M+H] +
第三步:1-(6-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)八氢-1H-吡咯并[2,3-c]吡啶-1-基)丙-2-烯-1-酮004的合成
将4d(62mg,0.16mmol)溶解到二氯甲烷(5mL)中,在0℃冰浴下加入N,N-二异丙基乙胺(62mg,0.18mmol)和丙烯酰氯(20mg,0.18mmol)。所得反应液在0℃下继续反应2小时至反应完全。反应液旋干得到的粗品用高效液相制备色谱法纯化得到标题产物004(7mg)。
高效液相制备色谱法:Waters 2767制备色谱仪,流动相A:0.1%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10μm),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至16min,10%ACN运行至18min结束纯化,得到目标化合物004。
LCMS:Rt:1.272min;MS m/z(ESI):441.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.46-11.40(d,1H),8.79(s,1H),7.95-7.86(d,1H),7.48,7.43(s,1H),7.08(dd,J=16.0Hz,1H),6.71,6.58(dd,J=16.0Hz,8Hz,1H),6.22-6.12(m,1H),5.75-5.60(m,1H),4.45-4.18(m,1H),4.09-3.97(m,2H),3.97-3.86(m,1H),3.78-3.72(m,1H),3.60-3.48(m,1H),3.48-3.39(m,1H),3.18-3.00(m,1H),2.82-2.71(m,1H),2.67-2.57(m,1H),2.33-2.10(m,2H),1.92-1.72(m,2H),1.40-1.25(m,3H).
实施例5化合物005的制备
N-(3-(6-((1-乙基-1H-吡唑-4-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)苯基)丙烯酰胺
Figure PCTCN2021073098-appb-000067
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000068
第一步:4,6-二氯-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶5b的制备
室温下将5a(1g,5.3mmol),3,4-二氢-吡喃(2.4mL,5.3mmol)和对甲苯磺酸(65mg,0.53mmol)溶于乙酸乙酯(15mL)中,反应液在氮气氛下50℃搅拌16个小时至反应完全。混合物用饱和碳酸氢钠溶液调碱至pH=10,用乙酸乙酯萃取,有机相合并后用无水硫酸钠干燥过滤,滤液减压浓缩,得到的粗品用硅胶柱(EA:PE=0~30%)纯化得到标题产物5b(1.4g)。
LCMS:Rt:1.854min;MS m/z(ESI):273.0[M+H] +
第二步:6-氯-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶5d的合成
室温下将5b(500mg,1.8mmol)、(3-((叔丁氧基羰基)氨基)苯基)硼酸5c(350mg,1.8mmol)、醋酸钯(10mg,0.07mmol)、三苯基磷(20mg,0.07mmol)和碳酸钠(200mg,1.8mmol,1M)溶于二氧六环(6mL)中,反应液在氮气氛下80℃搅拌16小时至反应完全。过滤,滤液用水和乙酸乙酯(20.0mL*3)萃取,有机相合并后用无水硫酸钠干燥过滤,滤液减压浓缩,得到的粗品用硅胶柱(EA:PE=0~30%)纯化得到产物5d(520mg)。
LCMS:Rt:2.090min;MS m/z(ESI):430.1[M+H] +
第三步:4-(3-氨基苯基)-N-(1-乙基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-6-胺5f的合成
室温下将5d(500mg,1.17mmol),1-乙基-1H-吡唑-4-胺5e(129mg,1.17mmol)和三氟乙酸(133mg,1.17mmol)溶解到异丙醇(5mL)中,反应液在110℃下搅拌16小时至反应完全。反应液用清水(50mL)稀释,加入碳酸钠固体调碱至pH=10,用乙酸乙酯(20.0mL*3)萃取,有机相合并后用无水硫酸钠干燥,过 滤旋干,得到的粗品用硅胶柱(EA:PE=0~60%)纯化得到标题产物5f(270mg)。
LCMS:Rt:1.170min;MS m/z(ESI):321.5[M+H] +
第四步:N-(3-(6-((1-乙基-1H-吡唑-4-基)氨基)-1H-吡唑并[3,4-d]嘧啶-4-基)苯基)丙烯酰胺005的合成
将4-(3-氨基苯基)-N-(1-乙基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-6-胺5f(160mg,0.5mmol)溶解到二氯甲烷(2mL)中,在0℃冰浴下加入三乙胺(152mg,1.5mmol)和丙烯酰氯5g(45mg,0.5mmol)。加完后反应液在0℃下继续反应30分钟至反应完全。反应液用清水(10mL)稀释,二氯甲烷(20mL*2)萃取,合并的有机相用饱和食盐水洗涤一次,无水硫酸钠干燥,过滤旋干得到的粗品。用高效液相制备色谱法纯化得到产物005(11.54mg)。
高效液相制备色谱法:Waters 2767制备色谱仪,流动相A:10mmol%NaHCO 3水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10μm),梯度:10%ACN运行3min,30%-40%ACN运行至16min,95%ACN运行至15min,10%ACN运行至20min结束纯化,得到化合物005。
LCMS:Rt:1.177min;MS m/z(ESI):375.0[M+H] +
1H NMR(400MHz,DMSO)δ13.32(s,1H),10.43(s,1H),9.71(s,1H),8.66(s,1H),8.33(s,1H),8.07(s,1H),7.93(s,1H),7.80(s,1H),7.57(t,J=8.0Hz,2H),6.49(dd,J=17.0,10.0Hz,1H),6.33(d,J=15.2Hz,1H),5.83(d,J=11.7Hz,1H),4.15(d,J=5.9Hz,2H),1.39(t,J=7.3Hz,3H).
实施例6化合物006、化合物006-1和化合物006-2的制备
1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺环[2.5]辛烷-6-基)丙-2-烯-1-酮006
(R)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺环[2.5]辛烷-6-基)丙-2-烯-1-酮006-1
(S)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺环[2.5]辛烷-6-基)丙-2-烯-1-酮006-2
Figure PCTCN2021073098-appb-000069
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000070
第一步:6-氮杂螺[2.5]辛烷-4-酮6b的合成
室温下将三氟乙酸(3mL)加入到化合物叔丁基6a(800mg,3.55mmol)的二氯甲烷溶液(10mL)中搅 拌2小时,点板发现反应完全(碘熏)。将此溶液在减压下浓缩干得到粗品6b(450mg)。
第二步:苯甲基4-羰基-6-氮杂螺[2.5]辛烷-6-羧酸酯6c的合成
室温下将DIEA(2.0mL,10.8mmol)加入到6-氮杂螺[2.5]辛烷-4-酮6b(450mg,3.6mmol)和CbzCl(0.8mL,4.5mmol)的四氢呋喃(15mL)溶液中搅拌2小时。点板监控反应完全。将此溶液在减压下浓缩,所得粗产品用硅胶柱(乙酸乙酯:石油醚=1:10)纯化得到产物6c(890mg)。
LCMS:Rt:1.646min;MS m/z(ESI):260.1[M+H] +
第三步:苯甲基(R,Z)-4-((叔丁基亚磺酰基)亚氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6e的合成
室温下将Ti(OEt) 4(2.32g,10.2mmol)加入到化合物苯甲基4-羰基-6-氮杂螺[2.5]辛烷-6-羧酸酯6c(880mg,3.4mmol)和化合物(R)-2-甲基丙烷-2-亚磺酰胺6d(820mg,6.8mmol)的四氢呋喃(15mL)溶液中搅拌过夜。TLC监控反应完全。加入水(20mL),乙酸乙酯(50mL)过滤,将此溶液在减压下浓缩,所得粗产品用硅胶柱纯化得到标题产物6e(600mg)。
LCMS:Rt:1.831min;MS m/z(ESI):363.1[M+H] +
第四步:苯甲基4-(((R)-叔丁基亚磺酰基)氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6f的合成
-10℃下将NaBH 4(235mg,6.0mmol)加入到化合物苯甲基(R,Z)-4-((叔丁基亚磺酰基)亚氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6e(560mg,1.5mmol)的四氢呋喃(10mL)溶液中,零度搅拌反应2小时。点板监控反应完全。加入水(20mL),乙酸乙酯(50mL),有机层用饱和食盐水洗,无水硫酸钠干燥,将此溶液在减压下浓缩,所得粗产品产物6f(560mg)。
LCMS:Rt:1.658min;MS m/z(ESI):365.1[M+H] +
第五步:苯甲基4-氨基-6-氮杂螺[2.5]辛烷-6-羧酸酯6g的合成
室温下将化合物苯甲基4-(((R)-叔丁基亚磺酰基)氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6f(560mg,1.5mmol)加入到HCl/二氧六环(5mL)的溶液里反应1小时,将反应液浓缩干得到标题产物6g(400mg)。
第六步:苯甲基4-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6i的合成
室温下分别将苯甲基4-氨基-6-氮杂螺[2.5]辛烷-6-羧酸酯6g(400mg,1.5mmol),化合物2,4,5-三氯-7H-吡咯并[2,3-d]嘧啶6h(660mg,3.0mmol)加入到DIEA(0.6mL,4.5mmol)的异丙醇(20.0mL)溶液内。将反应体系密封并加热到90℃在此温度下搅拌过夜。待反应体系降至室温后,将反应液在减压下浓缩得到粗产品,后经Waters 2767制备色谱仪,流动相A:0.1%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化得到标题产物6i(250mg)。
LCMS:Rt:1.887min;MS m/z(ESI):446.0[M+H] +
第七步:苯甲基4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6k的合成
室温下分别将中间体苯甲基4-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺[2.5]辛烷-6-羧酸酯6i(250mg,0.56mmol),化合物1-乙基-1H-吡唑-4-胺6j(123mg,1.12mmol)加入到三氟乙酸(100mg,1.1mmol)的异丙醇(15.0mL)溶液内。将反应体系密封并加热到90℃在此温度下搅拌过夜。待反应体系降至室温后,将反应液在减压下浓缩后经Waters 2767制备色谱仪,流动相A:0.1%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至15min,95%ACN运行至18min,10%ACN运行至20min结束纯化得到标题产物6k(170mg)。
LCMS:Rt:1.565min;MS m/z(ESI):521.1[M+H] +
第八步:5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-(6-氮杂螺[2.5]辛烷-4-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺6l的合成
室温下将中间体6k(60mg,0.3mmol)加入到三氟乙酸溶液(5mL)中,反应液加热到80度搅拌2小时。减压下将反应液浓缩,得到标题产物6l的三氟乙酸盐(120mg)。
LCMS:Rt:0.785min;MS m/z(ESI):387.1[M+H] +
第九步:(R)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺环[2.5]辛烷-6-基)丙-2-烯-1-酮006-1以及(S)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-6-氮杂螺环[2.5]辛烷-6-基)丙-2-烯-1-酮006-2的合成
室温下将三乙胺(0.1mL,0.6mmol)加入到化合物(R)-5-氯-N2-(1-乙基-1H-吡唑-4-基)-N4-(6-氮杂螺[2.5]辛烷-4-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺6l(120mg,三氟乙酸盐)的二氯甲烷(10mL)溶液中。所得溶液在室温下搅拌10分钟。然后将该溶液用冰水浴冷却,并缓慢滴加含丙烯酰氯6m(33mg,0.37mmol)的四氢呋喃溶液(2.0mL)。反应液温度升至室温,并搅拌2小时。减压下将反应液浓缩,所得粗品用二甲基甲酰胺溶解经Waters 2767制备色谱仪,流动相A:0.1%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化得到目标产物006(40mg,收率29.2%),006(25mg)经过SFC拆 分得到标题产物006-1(8.0mg,5.5%),006-2(4.0mg,2.8%)。
LCMS:Rt:1.139min;MS m/z(ESI):441.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.25(s,1H),δ8.67(s,1H),7.84(s,1H),7.46(s,1H),6.91(s,1H),6.55-6.48(m,1H),6.11-5.96(m,1H),5.75-5.70(m,1H),5.44-5.41(m,1H),4.29(d,1H),4.05(dd,J=14.4Hz,J=7.2Hz,2H),3.92-3.89(m,1H),3.62-3.60(m,1H),3.52-3.50(m,1H),2.98(t,J=10.0Hz,1H),1.84-1.80(m,1H),1.35(t,J=7.2Hz,3H),1.25-1.15(m,1H),1.05(d,J=11.6Hz,1H),0.74-0.52(m,2H),0.40(s,1H).
1H NMR(400MHz,DMSO-d 6)δ11.25(s,1H),8.67(s,1H),7.84(s,1H),7.45(s,1H),6.91(s,1H),6.55-6.48(m,1H),6.11-5.96(m,1H),5.89-5.73(m,1H),5.44-5.39(m,1H),4.31(d,1H),4.08(dd,J=14.4Hz,J=7.2Hz,2H),3.92-3.89(m,1H),3.66-3.64(m,1H),3.53-3.49(m,1H),2.97(t,J=10.0Hz,1H),2.02-1.87(m,1H),1.35(t,J=7.2Hz,3H),1.25-1.05(m,2H),0.87-0.62(m,2H),0.40(s,1H).
SFC制备方法:仪器:Waters SFC80,制备柱:ChiralPak AD,250×25mm,10μm,流动相:A for CO 2and B for MeOH(0.1%DEA),梯度:B 0-40%,流动速率:70g/min,压力:100bar,温度:35℃,波长:214nm,时间:8min,样品溶解方法:溶解于5毫升甲醇中,进样量:3.5毫升/针。
实施例7化合物007、化合物007-1和化合物007-2的制备
1-(7-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮007
(R)-1-(7-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮007-1
(S)-1-(7-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮007-2
Figure PCTCN2021073098-appb-000071
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000072
第一步:苯甲基5,8-二氧杂-10-氮杂二螺[2.0.4 4.3 3]十一烷-10-羧酸酯7b的合成
0℃下,将DIPEA(1.7g,13mmol)加入到5,8-二氧杂-10-氮杂二螺[2.0.4.3]十一烷7a(1g,6.5mmol)的四氢呋喃溶液(15mL)中,在此温度下,搅拌10分钟后加入CbzCl,然后室温搅拌2h。反应液减压浓缩,所得粗产品通过柱层色谱纯化,得到标题产物7b(900mg)。
第二步:苯甲基7-羰基-5-氮杂螺[2.4]庚烷-5-羧酸酯7c的合成
室温下,将对甲苯磺酸(2.7g,15.3mmol)加入到中间体7b(900mg,3.1mmol)的丙酮/水(1:1,15mL)溶液的中。反应体系室温搅拌16h后,减压浓缩除去丙酮,然后加入20mL DCM,饱和食盐水洗两遍,浓缩所得粗产品通过柱层色谱纯化得到标题产物7c(720mg,纯度:90%)
第三步:苯甲基(R,Z)-7-((叔丁基亚磺酰基)亚氨基)-5-氮杂螺[2.4]庚烷-5-羧酸酯7e的合成
室温下,将中间体苯甲基7-羰基-5-氮杂螺[2.4]庚烷-5-羧酸酯7c(720mg,2.9mmol)和化合物(R)-2-甲基丙烷-2-亚磺酰胺7d(423mg,3.5mmol)溶入到无水四氢呋喃(20mL)中,加入钛酸四乙酯(2.0g,8.7mmol),在氮气保护下,反应体系在65℃下搅拌16h。待反应体系降至室温后,加入500mg水和20mL EtOAc,过滤浓缩,粗产品通过柱层色谱(乙酸乙酯:石油醚=1:10)纯化,得到标题产物7e(780mg,纯度:90%).
第四步:苯甲基7-(((R)-叔丁基亚磺酰基)氨基)-5-氮杂螺[2.4]庚烷-5-羧酸酯7f的合成
中间体7e(780mg,2.2mmol)溶于无水四氢呋喃中,零度以下,加入NaBH 4(124.8mg,3.3mmol)。氮气保护下,反应体系在-48~0℃搅拌4h后,加入20mL EA,用饱和食盐水洗两遍,浓缩得到标题产物7f粗产品(800mg)。
LCMS:Rt:1.652min;MS m/z(ESI):351.4[M+H] +
第五步:苯甲基7-氨基-5-氮杂螺[2.4]庚烷-5-羧酸酯7g的合成
粗产品中间体苯甲基7-(((R)-叔丁基亚磺酰基)氨基)-5-氮杂螺[2.4]庚烷-5-羧酸酯7f(800mg)溶于HCl/二氧六环(4M,2.0mL)中,室温搅拌2h,减压浓缩,得到标题产物7g(810mg)。
LCMS:Rt:1.310min;MS m/z(ESI):247.2[M+H] +
第六步:苯甲基7-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-羧酸酯7i的合成
在50mL闷罐中,室温下粗产品中间体苯甲基7-氨基-5-氮杂螺[2.4]庚烷-5-羧酸酯7g(810mg)溶于异丙醇(15mL)中,加入2,4,5-三氯-7H-吡咯并[2,3-D]嘧啶(746mg,3.4mmol)和DIEA(582mg,4.5mmol)。反应体系于120℃下搅拌12h。反应液冷却至室温后,浓缩所得粗产品通过柱层色谱(甲醇:二氯甲烷=1:10)纯化,得到标题产物7i(360mg,纯度:95%)。
LCMS:Rt:10.695min;MS m/z(ESI):432.3[M+H] +
第七步:苯甲基7-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-羧酸酯7k的合成
在25mL闷罐中,室温下7i(360mg,0.84mmol)溶于异丙醇(10mL)中,加入4-氨基-1-乙基吡唑(93.2mg,0.84mmol)和TFA(95.8mg,0.84mmol)。反应体系于110℃下搅拌12h。反应液冷却至室温后,浓缩所得粗产品通过柱层色谱纯化,得到标题产物7k(170mg,纯度:95%)。
LCMS:Rt:1.600min;MS m/z(ESI):507.1[M+H] +
第八步:5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-(5-氮杂螺[2.4]庚烷-7-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺7l的合成
室温下中间体苯甲基7-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-羧酸酯7k(170mg,0.34mmol)溶于三氟乙酸(3mL)中,反应体系于80℃下搅拌2h。浓缩得到标题产物7l(182mg)。
LCMS:Rt:1.290min;MS m/z(ESI):373.1[M+H] +
第九步:1-(7-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮007;(R)-1-(7-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮007-1;及(S)-1-(7-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮007-2的合成
0℃下,将三乙胺(51mg,0.50mmol)加入到粗产品化合物5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-(5-氮杂螺[2.4]庚烷-7-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺7l(182mg)的二氯甲烷(5mL)溶液中,在此温度下搅拌10分钟。然后缓慢滴加含丙烯酰氯7m(30.4mg,0.33mmol)的二氯甲烷溶液(0.5mL)。反应液温度 升至室温,并搅拌2小时后。反应液减压浓缩,所得粗品用DMF溶解经反相Flash得到标题化合物007,化合物007经SFC拆分后得到标题产物007-1(15.5mg,收率21%)和007-2(14.2mg,收率21%)。
LCMS:Rt:7.255min;MS m/z(ESI):427.4[M+H] +
LCMS:Rt:7.290min;MS m/z(ESI):427.4[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.22(s,1H),8.66(s,1H),7.83(s,1H),7.45(s,1H),6.91(s,1H),6.59-6.48(m,1H),6.13(d,J=16Hz,1H),5.85(d,J=4Hz,1H),5.68-5.64(m,1H),4.51-4.29(m,1H),4.07-4.01(m,2H),3.87-3.79(m,2H),3.62-3.59(m,1H),3.52-3.37(m,1H),1.35-1.31(t,J=8Hz,3H),1.21(s,1H),0.87-0.82(m,1H),0.76-0.65(m,2H).
1H NMR(400MHz,DMSO-d 6)δ11.24(s,1H),8.68(s,1H),7.82(s,1H),7.45(s,1H),7.16-7.15(m,1H),6.99-6.91(m,1H),6.56-6.47(m,1H),6.14-6.10(m,1H),5.82(d,J=4Hz,1H),5.67-5.63(m,1H),4.51-4.28(m,1H),4.06-4.01(m,2H),3.87-3.79(m,1H),3.62-3.60(m,1H),3.51-3.41(m,1H),1.34-1.31(t,J=8Hz,3H),1.20(s,1H),0.87-0.81(m,1H),0.74-0.65(m,2H).
制备方法:仪器:Waters SFC80,制备柱:ChiralPak OD,250×25mm,10μm,流动相:A for CO 2and B for EtOH(0.1%DEA),梯度:B 0-40%,流动速率:70g/min,压力:100bar,温度:35℃波长:214nm,时间:13min,样品溶解方法:溶解于5毫升甲醇中,进样量:3.5毫升/针。
实施例8化合物008的制备
1-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000073
合成路线及具体合成步骤:
Figure PCTCN2021073098-appb-000074
第一步:叔-丁基(R)-3-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸酯8c的合成
室温下在封管体系中将二异丙基乙胺(516mg,4mmol)加入到化合物2,4,5-三氯-7H-吡咯并[2,3-d]嘧啶8a(442mg,2mmol)和化合物叔-丁基(R)-3-氨基吡咯烷-1-羧酸酯8b(744mg,4mmol)的乙醇溶液(8mL)中,密封,110℃搅拌过夜,TLC点板发现反应完全。冷却,过滤,滤饼用8mL的四氢呋喃洗2次,将此滤液在减压下浓缩干得到粗品,过柱得到标题产物8c(594mg,收率:80%)。
LCMS:Rt:0.924min;MS m/z(ESI):373.2[M+H] +
第二步:叔丁基(R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸酯8e的合成
室温下在单口烧瓶中将三氟乙酸(77mg,0.792mmol)加入到(R)-8c(294mg,0.792mmol)和1-乙基-1H-吡唑-4-胺8d(88mg,0.792mmol)的异丙醇(6mL)溶液中,升温至95度搅拌过夜。点板监控反应完全。冷却至室温,将此溶液在减压下浓缩,所得粗产品用硅胶柱纯化得到标题产物8e(101mg,收率:28.6%)
LCMS:Rt:1.032min;MS m/z(ESI):447.6[M+H] +
第三步:(R)-5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-(吡咯烷-3-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺8f的合成
室温下在单口烧瓶中将三氟乙酸(4mL)加入到(R)-8e(65.3mg,0.15mmol)的二氯甲烷(6mL)溶液 中,室温搅拌1小时。点板监控反应完全。将此溶液在减压下浓缩,再次加入二氯甲烷(6mL),再次浓缩,所得粗产品8f直接用于下一步反应。
第四步:1-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮008的合成
室温下将三乙胺(89mg,0.88mmol)加入到(R)-8f(0.146mmol,三氟乙酸盐)的四氢呋喃(8mL)溶液中。所得溶液在室温下搅拌10分钟。然后将该溶液用冰水浴冷却,并缓慢滴加含丙烯酰氯8g(13.2mg,0.146mmol)的四氢呋喃溶液(2.0mL)。反应液温度升至室温,并搅拌2小时。减压下将反应液浓缩,所得粗品用二甲基甲酰胺溶解经反相Flash纯化得到标题产物008(21.4mg)。
LCMS:Rt:1.12min;MS m/z(ESI):401.4[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.22(s,1H),8.69(s,1H),7.86(s,1H),7.48(s,1H),6.91(s,1H),6.66-6.53(m,1H),6.18-6.04(m,2H),5.70-5.64(m,1H),4.81-4.63(m,1H),4.08-4.02(m,2H),3.88-3.74(m,1H),3.68-3.60(m,1H),3.47-3.38(m,1H),2.43-2.03(m,3H),1.34(t,J=7.2Hz,3H).
实施例9化合物009、化合物009-1及化合物009-2的制备
1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-基)丙-2-烯-1-酮009
(R)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-基)丙-2-烯-1-酮009-1
(S)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-基)丙-2-烯-1-酮009-2
Figure PCTCN2021073098-appb-000075
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000076
第一步:叔-丁基4-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-羧酸酯9c的合成
室温下分别将中间体9a(500mg,2.26mmol),9b(726mg,3.39mmol)加入到DIPEA(830mg,6.78mmol)的异丙醇(20.0mL)溶液内。将反应体系密封并加热到90℃在此温度下搅拌过夜。待反应体系降至室温后,将反应液在减压下浓缩得到粗产品,后经反相FLASH纯化得到标题产物9c 650mg。
LCMS:Rt:1.915min;MS m/z(ESI):400.0[M+H] +
第二步:叔丁基4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-羧酸酯9e的合成
室温下分别将9c(650mg,1.63mmol),化合物1-乙基-1H-吡唑-4-胺9d(363mg,3.26mmol)加入到三 氟乙酸(160mg,1.63mmol)的异丙醇(30.0mL)溶液内。将反应体系密封并加热到90℃在此温度下搅拌过夜。待反应体系降至室温后,将反应液在减压下浓缩得到粗产品,后经反相FLASH纯化得到产物9e(310mg)。
LCMS:Rt:1.520min;MS m/z(ESI):475.1[M+H] +
第三步:N 4-(吖庚环-4-基)-5-氯-N 2-(1-乙基-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺9f的合成
室温下将嘧啶9e(310mg,0.65mmol)加入到三氟乙酸(3mL)的二氯甲烷溶液(10mL)中,反应液室温搅拌过夜。减压下将反应液浓缩,得到产物9f的三氟乙酸盐(250mg)。
LCMS:Rt:0.784min;MS m/z(ESI):375.0[M+H] +
第四步:1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-基)丙-2-烯-1-酮009及光学异构体(R)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-基)丙-2-烯-1-酮009-1和(S)-1-(4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吖庚环-1-基)丙-2-烯-1-酮009-2的合成
室温下将三乙胺(0.15mL,2.0mmol)加入到9f(250mg,三氟乙酸盐)的二氯甲烷(10mL)溶液中。所得溶液在室温下搅拌10分钟。然后将该溶液用冰水浴冷却,并缓慢滴加含丙烯酰氯9h(90mg,1.0mmol)的四氢呋喃溶液(2.0mL)。反应液温度升至室温,并搅拌过夜。减压下将反应液浓缩得到化合物009,化合物009用二甲基甲酰胺溶解经反相Flash纯化和SFC拆分后得到标题产物009-1(25.3mg)和009-2(39.5mg)。
LCMS:Rt:1.095min;MS m/z(ESI):429.0[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.19(s,1H),8.58(s,1H),7.88(s,1H),7.49(s,1H),6.88-6.62(m,2H),6.12-5.87(m,1H),5.68(s,1H),5.41-5.32(m,1H),4.81-4.48(m,1H),4.28-3.95(m,3H),3.19-3.12(m,1H),3.16-2.75(m,2H),2.42-1.97(m,1H),1.67-1.54(m,5H),1.46-1.34(m,3H). 1H NMR(400MHz,DMSO-d 6)δ11.15(s,1H),8.59(s,1H),7.84(s,1H),7.47(s,1H),6.87(s,1H),6.82-6.75(m,1H),6.18-6.12(m,1H),5.87-5.80(m,1H),5.70-5.66(m,1H),4.27(s,1H),4.09-4.03(m,2H),3.79-3.32(m,4H),2.12-1.68(m,6H),1.35(t,J=7.2Hz,3H),.
制备方法:仪器:Waters SFC80,制备柱:ChiralPak AY,250×25mm,10μm,流动相:A for CO 2and B for MeOH(0.1%DEA),梯度:B 0-40%,流动速率:70g/min,压力:100bar,温度:35℃,波长:214nm,时间:13min,样品溶解方法:溶解于15毫升甲醇中,进样量:5毫升/针。
实施例10化合物010的制备
N-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)丙烯酰胺
Figure PCTCN2021073098-appb-000077
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000078
第一步:(叔丁基(3-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯)010-b合成
室温下将010-a(550.0mg,2.5mmol)加入到溶有N-Boc-3-氨基苯酚(784.5mg,3.8mmol)和碳酸钾(1.0g,7.5mmol)的DMF(20mL)溶液中,将此混合物在90℃下搅拌16小时。将此溶液用饱和食盐水(100mL)稀释,然后用乙酸乙酯(30mL*3)萃取。所得有机相在减压下浓缩,所得油状物在经反相Flash纯化得到目标产物010-b(150mg,收率:15.2%)。
LCMS:Rt:1.47min;MS m/z(ESI):339.0,341.0[M+H-(t-Bu)].
1H NMR(400MHz,DMSO-d 6)δ12.78(s,1H),9.64(s,1H),7.77(s,1H),7.50(s,1H),7.38-7.43(m,2H),6.95-6.98(m,1H),1.53(s,9H).
第二步:(叔丁基(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯)010-c的合成
将三氟乙酸(37.0mg,0.38mmol)加入到溶有010-b(150.0mg,0.38mmol)和1-乙基-1H-吡唑-4-胺(84.0mg,0.76mmol)的异丙醇(5mL)中,将此溶液在封管中加热到110℃,并持续搅拌10小时。待反应体系温度降至室温后减压浓缩后,所得粗产品用反相Flash纯化得到010-c(50mg,收率:28%,纯度:90%)。
LCMS:Rt:1.40min;MS m/z(ESI):470.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.62(s,1H),9.55(s,1H),9.10(s,1H),7.34-7.38(m,3H),7.17(s,1H),6.86-6.88(d,J=7.2Hz,1H),3.81-3.82(m,2H),1.44(s,9H),1.56-1.21(m,3H).
第三步:(4-(3-氨基苯氧基)-5-氯-N-(1-乙基-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-胺)010-d的合成
约5℃下,将盐酸二氧六环溶液(4.0M,2mL)加入到含有010-c(50mg,0.11mmol)的单口瓶中。搅拌30分钟后,将反应液在减压下浓缩,得到粗产品010-d(50mg,盐酸盐,纯度:55%),未经进一步纯化直接于下一步使用。
LCMS:Rt:0.94min;MS m/z(ESI):370.2,372.1[M+H].
第四步:(N-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)丙烯酰胺)010的合成
约5℃下将三乙胺(36mg,0.36mmol)加入到化合物010-d(50mg,盐酸盐)的二氯甲烷(2mL)溶液中。缓慢滴加含丙烯酰氯(12mg,0.13mmol)的二氯甲烷溶液(1.0mL)。反应液温度升至室温,并搅拌30分钟。减压下将反应液浓缩,制备纯化(条件:乙腈/水(0.04%甲酸))纯化得到目标产物010(7.02mg,收率:13%)。
LCMS:Rt:3.99min;MS m/z(ESI):424.4,426.4[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.66(s,1H),10.38(s,1H),9.14(s,1H),7.67-7.68(m,2H),7.47-7.48(m,1H),7.21-7.22(m,2H),7.01-7.03(d,J=8.8Hz,2H),6.47(dd,J=10,10.6Hz,1H),6.24(dd,J=1.2,1.6Hz,1H),5.76-5.79(dd,J=2,8Hz,1H),3.77-3.82(m,2H),1.20(t,J=7.2Hz,3H)。
实施例11化合物011-1的制备
1-((3S,4R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000079
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000080
第一步:2,4,5-三氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶011-1-b合成
在0℃条件下,将NaH(100mg,2.49mmol)分多批次加入2,4,5-三氯-7H-吡咯并[2,3-d]嘧啶011-1-a(500mg,2.26mmol)的四氢呋喃(6mL)溶液中,加完搅拌15分钟后,将SEMCl(604mg,3.62mmol)滴加进去,0℃反应1个小时。待LCMS监测反应结束后将15mL饱和NH 4Cl溶液加入反应液,用二氯甲烷萃取三次(15mL*3),有机相合并,用水(5mL)和饱和食盐水(5mL)各洗一次,有机相用无水硫酸钠干燥1个小时,过滤,此滤液在减压下浓缩过柱(乙酸乙酯:石油醚=1:10)得到011-1-b(639mg,收率:80.5%)。
LCMS:Rt:2.01min;MS m/z(ESI):353.9[M+H]。
1H NMR(400MHz,CDCl 3)δ7.37(s,1H),5.60(s,2H),3.56(t,J=8.0Hz,2H),0.96(t,J=8.4Hz,2H),0.02(s,9H).
第二步:(3S,4R)-3-((2,5-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-羧酸叔丁酯011-1-c的合成
室温下,将NaH(100mg,2.49mmol)分多批次加入(3R,4S)-3-氟-4-羟基吡咯烷-1-羧酸叔丁酯(331mg,1.58mmol)的DMSO(10mL)溶液中,加完搅拌15分钟,然后将2,4,5-三氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶011-1-b(555mg,1.58mmol)加进去,反应体系升温到55度,反应一个小时,将50mL饱和NH 4Cl溶液加入反应液,用二氯甲烷萃取三次(50mL*3),有机相合并,用水(50mL)和饱和食盐水(50mL)各洗一次,有机相用无水硫酸钠干燥1个小时,过滤,此滤液在减压下浓缩后过柱纯化(乙酸乙酯:石油醚=1:8)得到纯品011-1-c(644.4mg,收率:78.2%)。
LCMS:Rt:2.25min;MS m/z(ESI):521.5[M+H].
第三步:(3S,4R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-羧酸叔丁酯011-1-d的合成
室温氮气保护下,将Pd 2(dba) 3(115mg,0.124mmol)与BINAP(73mg,0.124mmol)加入到(3S,4R)-3-((2,5-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-羧酸叔丁酯011-1-c(644mg,1.24mmol),1-乙基-1H-吡唑-4-胺(277mg,2.48mmol)和Cs 2CO 3(815mg,2.48mmol)的1,4-二氧六环(15mL)溶液中,反应体系升温至110℃搅拌16小时,冷却至室温。过滤,滤饼用60mL四氢呋喃洗涤,将此溶液在减压下浓缩后所得粗产品用Chemflash纯化(乙酸乙酯:石油醚=1:8)得到纯品011-1-d(309.2mg,收率:42%)。
LCMS:Rt:1.82min;MS m/z(ESI):596.7[M+H].
第四步:5-氯-N-(1-乙基-1H-吡唑-4-基)-4-((3S,4R)-4-氟吡咯烷-3-基)氧基)-7H吡咯并[2,3-d]嘧啶-2-胺011-1-e的合成
室温下,将TFA(2.5mL)加入到011-1-d(309.2mg,0.52mmol)的二氯甲烷(5mL)溶液中,室温搅拌1个小时,点板原料消失,在减压下浓缩后加入5mL二氯甲烷。在减压下浓缩带走多余三氟乙酸。所得粗品加入5mL甲醇,和1mL DIPEA。升温至50℃,搅拌一个小时,在减压下浓缩得到粗品011-1-e,直接用于下一步反应。
LCMS:Rt:0.961min;MS m/z(ESI):366.4[M+H].
第五步:1-((3S,4R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮011-1的合成
将粗品011-1-e溶于5mL丙酮和5mL水的混合溶剂中,用碳酸钾调节pH值到10,反应液冷却至0℃,然后将丙稀酰氯(70.2mg,0.78mmol)溶解在丙酮(1mL)中滴加到反应液中。滴加完毕后,反应液在0℃下搅拌1h,LCMS检测到原料消失。反应结束后,将反应液过滤后制备纯化(条件:乙腈/水(0.04%甲酸)),得到011-1(24.4mg,纯度:99.2%)。
LCMS:Rt:2.84min;MS m/z(ESI):420.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.58(s,1H),9.12(s,1H),7.90(s,1H),7.51(s,1H),7.09(s,1H),6.68-6.59(m,1H),6.18(d,J=16.8Hz,1H),5.76-5.51(m,3H),4.39-4.16(m,1H),4.10-4.05(m,2H),4.04-3.93(m,1H),3.86-3.65(m,2H),1.35(t,J=7.2Hz,3H).
实施例12化合物012-1的制备
1-((3S,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000081
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000082
第一步:2-氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶012-1-b合成
在20℃下搅拌,将叔丁基(3R,4S)-3-氟-4-羟基吡咯烷-1-羧酸盐(350mg,1.71mmol)加入到DMSO(20mL)溶液内,氢化钠(70mg,1.71mmol)分批加入反应液,继续搅拌15分钟,将2,4-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶1b(541mg,1.71mmol)加入反应液中,升温到55℃搅拌1小时,点板监控反应完全,水(50mL)加进去,乙酸乙酯(50mL*2)萃取,合并有机相,用饱和食盐水洗,将有机相在减压下浓缩过柱(乙酸乙酯:石油醚=1:1)得到012-1-b(300mg,收率:36.2%)。
LCMS:Rt:1.89min;MS m/z(ESI):487.2[M+H].
第二步:叔丁基(3S,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-羧酸酯012-1-c的合成
室温在氮气保护下,将Pd 2(dba) 3(55mg,0.06mmol),BINAP(37mg,0.06mmol)加入到012-1-b(300mg,0.62mmol),1-乙基-1H-吡唑-4-胺(137mg,1.24mmol)和碳酸铯(606mg,1.86mmol)的1,4-二氧六环(20mL)溶液中,反应体系升温至120℃搅拌过夜,冷却至室温。将反应液在减压下浓缩后过柱纯化(乙酸乙酯:石油醚=1:1)得到012-1-c(255mg,收率:73.7%)。
LCMS:Rt:1.99min;MS m/z(ESI):562.2[M+H].
1H NMR(400MHz,CDCl 3)δ7.86(s,1H),7.58(s,1H),6.91(s,1H),6.56(s,1H),6.49(s,1H),5.52(s,2H), 5.46-5.32(m,1H),4.15-3.53(m,8H),1.54-1.52(m,3H),1.50(s,9H),0.94-0.86(m,2H),0.02(s,9H).
第三步:N-(1-乙基-1H-吡唑-4-基)-4-((3S,4R)-4-氟吡咯烷-3-基)氧基)-7H吡咯并[2,3-d]嘧啶-2-胺012-1-d的合成
(1)室温下将012-1-c(255mg,0.45mmol)加入到三氟乙酸(2mL)的二氯甲烷溶液(5mL)中,反应液室温搅拌2小时。减压下将反应液浓缩得到粗品(164mg,收率:>99%),未经纯化直接进行下一步
LCMS:Rt:0.76min;MS m/z(ESI):362.0[M+H].
(2)室温下将粗品(164mg,0.45mmol)加入到DIPEA(2mL)的甲醇溶液(4mL)中,反应液50℃搅拌过夜。减压下将反应液浓缩,得到012-1-d(150mg,收率:>99%)。
LCMS:Rt:0.76min;MS m/z(ESI):332.1[M+H].
第四步:1-((3S,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮012-1的合成
将012-1-d(150mg,0.45mmol)溶解在丙酮(4mL)和H 2O(2mL)的混合液中,加入碳酸钾固体(186mg,1.35mmol)室温搅拌,将丙烯酰氯(60mg,0.67mmol)滴加进去,室温搅拌2小时,过滤制备纯化(条件:乙腈/水(0.04%甲酸))得到012-1(42.5mg,三步收率25.3%)。
LCMS:Rt:5.71min;MS m/z(ESI):386.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.38(s,1H),8.98(s,1H),7.92(s,1H),7.50(s,1H),6.97(s,1H),6.68-6.59(m,1H),6.27-6.18(m,2H),5.76-5.47(m,3H),4.40-4.21(m,1H),4.10-3.76(m,4H),3.69-3.64(m,1H),1.35(t,J=7.2Hz,3H).
实施例13化合物013-1的制备
1-((3S,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡唑并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000083
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000084
第一步:4,6-二氯-1-四氢吡喃-7H-吡唑[2,3-D]嘧啶013-1-b合成
室温下,将4,6-二氯-7H-吡唑[2,3-D]嘧啶013-1-a(5.0g,26.6mmol)溶于乙酸乙酯中,加入对甲苯磺酸(4.6g,26.6mmol)和DHP(1.1g,13.3mmol);然后在50℃下搅拌3小时。减压蒸馏,粗产品过柱纯化(乙酸乙酯:石油醚=1:6),得到013-1-b(5.0g,收率:68.8%)。
1H NMR(400MHz,DMSO-d 6)δ8.61(s,1H),5.93-5.96(dd,J=8,10Hz,1H),3.97(d,J=12Hz,1H),3.77-3.71(m,1H),2.44-2.35(m,1H),2.02(d,J=4Hz,1H),1.95(dd,J=4,8Hz,1H),1.81-1.74(m,1H),1.65-1.59(m,2H).
第二步:1-((3S,4R)-3-(6-氯-1-(四氢-2H-吡喃-2-基)-1H吡唑并[2,3-d]嘧啶-4-基)氧基)-4-氟 吡咯烷-1-基)羧酸叔丁酯013-1-c的合成
室温下,将4,6-二氯-1-四氢吡喃-7H-吡唑[2,3-D]嘧啶013-1-b(330mg,1.2mmol)和(3S,4R)-3-羟基-4-氟吡咯烷-1-羧酸叔丁酯(248.7mg,1.2mmol)溶于DMSO中(8.0mL)中,加入NaH(34.6mg,1.4mmol),然后55℃搅拌2小时。反应液减压浓缩,所得粗产品通过柱层色谱纯化(乙酸乙酯:石油醚=1:6),得到013-1-c(180.0mg,收率:34.2%)。
LCMS:Rt:1.1.803min;MS m/z(ESI):442.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ8.40(s,1H),5.89(d,J=6.0Hz,1H),5.76(s,1H),5.56-5.42(m,1H),3.96-3.93(m,2H),3.75-3.55(m,3H),3.41-3.32(m,1H),2.41-2.38(m,1H),2.01-1.89(m,2H),1.77(s,1H),1.57(s,2H),1.43(s,9H).
第三步:(3S,4R)-3-((6-(1-乙基-1H-吡唑-4-基)氨基)-1-(四氢-2H-吡喃-2-基)-1H-吡唑并[3,4-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-羧酸叔丁酯013-1-d的合成
室温下,将中间体013-1-c(180mg,0.41mmol)溶于二氧六环(10.0mL)中,加入4-氨基-1-乙基吡唑(54.6mg,0.49mmol),随后在搅拌下,加入Pd 2(dba) 3(36.6mg,0.04mmol),BINAP(49.8mg,0.08mmol),Cs 2CO 3(260.6mg,0.8mmol)。在氮气保护下,于100℃搅拌12小时。减压蒸馏,粗产品通过柱层色谱纯化(乙酸乙酯:石油醚=1:15),得到013-1-d(95mg,收率:43.9%)。
LCMS:Rt:1.628min;MS m/z(ESI):517.4[M+H].
1H NMR(400MHz,DMSO-d 6)δ9.74(s,1H),8.06-7.97(m,2H),7.62(s,1H),5.80-5.61(m,3H),4.13(s,2H),4.04-4.00(s,2H),3.67-3.61(m,3H),2.02-1.99(m,2H),1.88-1.79(m,2H),1.58(m,2H),1.50-1.43(m,13H).
第四步:1-((3S,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡唑并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷013-1-e的合成
室温下,中间体013-1-d(95mg,0.18mmol)溶于二氯甲烷中(2.0mL),加入HCl/二氧六环(4.0M,1.0mL)中,室温搅拌3小时,减压浓缩,得到粗产品013-1-e(110mg),不经纯化,直接用于下一步。
LCMS:Rt:1.085min;MS m/z(ESI):333.1[M+H].
第五步:(1-((3S,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡唑并[2,3-d]嘧啶-4-基)氧基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮013-1的合成
0℃下,将粗产品013-1-e溶于丙酮和水中(2.0mL)。加入碳酸钾(49.7mg,0.36mol),在此温度下搅拌10分钟;然后缓慢滴加含丙烯酰氯(39.1mg,0.43mmol)的丙酮溶液(0.5mL)。反应液温度升至室温,并搅拌2小时。粗产品过滤,通过经反相Flash纯化(条件:乙腈/水(0.04%甲酸)),得到013-1(29.4mg,两步收率42.3%)。
LCMS:Rt:4.967min;MS m/z(ESI):387.5[M+H].
1H NMR(400MHz,DMSO-d 6)δ13.19(s,1H),9.49(s,1H),7.96-7.91(m,2H),7.57(s,1H),6.66(t,J=6.0,10.0Hz,1H),6.23(d,J=12Hz,1H),5.79-5.76(m,3H),4.35-4.33(m,1H),4.11-3.95(m,2H),3.89-3.64(m,2H),3.45-3.40(m,1H),1.36(s,3H).
实施例14化合物015-1的制备
(R)-1-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000085
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000086
第一步:叔丁基(R)-3-((2,5-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯015-1-b合成
将叔丁基(R)-3-羟基吡咯烷-1-羧酸酯(295mg,1.58mmol)加入到DMSO(20mL)溶液内,在20℃下搅拌,氢化钠(63mg,1.58mmol)分批加入反应液,继续搅拌15分钟后,将2,4,5-三氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶011-1-b(500mg,1.58mmol)加入反应液中,并升温到55℃搅拌2小时。点板监控反应完全后,水(50mL)加进去,乙酸乙酯(50mL*2)萃取,合并有机相,用饱和食盐水洗,将有机相在减压下浓缩,过硅胶柱(乙酸乙酯:石油醚=1:1)得到015-1-b(450mg,收率:56.9%)。
LCMS:Rt:2.40min;MS m/z(ESI):447.1[M+H].
1H NMR(400MHz,CDCl 3)δ7.13(s,1H),5.84(d,J=8.4Hz,1H),5.54(s,1H),3.76-3.53(m,6H),2.28(s,2H),1.45(s,9H),0.97-0.93(m,3H),0.02(s,9H).
第二步:叔丁基(R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯015-1-c的合成
室温在氮气保护下,将Pd 2(dba) 3(82mg,0.09mmol),BINAP(56mg,0.09mmol)加入到015-1-b(450mg,0.9mmol),1-乙基-1H-吡唑-4-胺(150mg,1.35mmol)和碳酸铯(880mg,2.70mmol)的1,4-二氧六环(50mL)溶液中,反应体系升温至120℃搅拌过夜,冷却至室温。将反应液在减压下浓缩后过柱纯化(乙酸乙酯:石油醚=1:1)得到015-1-c(430mg,收率:83.1%)。
LCMS:Rt:2.145min;MS m/z(ESI):578.3[M+H].
1H NMR(400MHz,CDCl 3)δ7.90(s,1H),7.66(s,1H),6.89(s,1H),6.70(d,J=8.0Hz,1H),5.77-5.73(m,1H),5.52(s,2H),4.28-4.18(m,2H),3.81-3.59(m,6H),2.35-2.25(m,2H),1.61-1.55(m,11H),1.34(t,J=6.8Hz,1H),1.00(t,J=8.0Hz,2H),0.02(s,9H).
第三步:(R)-(5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-4-(吡咯烷-3-氧基)-7H吡咯并[2,3-d]嘧啶-7-基)甲醇015-1-d的合成
室温下将叔丁基(R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯015-1-c(430mg,0.74mmol)加入到三氟乙酸(4mL)的二氯甲烷溶液(8mL)中,反应液室温搅拌2小时。减压下将反应液浓缩得到015-1-d(281mg,收率:>99%)。粗品未经纯化直接使用于下一步。
LCMS:Rt:0.782min;MS m/z(ESI):378.1[M+H].
第四步:(R)-5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(吡咯烷-3-氧基)-7H吡咯并[2,3-d]嘧啶-2-胺015-1-e的合成
室温下将015-1-d(281mg,0.74mmol)加入到DIPEA(4mL)的甲醇溶液(8mL)中,反应液50℃搅拌过夜。减压下将反应液浓缩,得到015-1-e(259mg,收率:>99%)。粗品未再经纯化直接使用于下一步。
LCMS:Rt:0.830min;MS m/z(ESI):348.1[M+H]。
第五步:(R)-1-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基) 吡咯烷-1-基)丙-2-烯-1-酮015-1的合成
将粗品015-1-e(258mg,0.74mmol)溶解在丙酮(8mL)和H 2O(4mL)的混合液中,加入碳酸钾固体(306mg,2.22mmol)室温搅拌,将丙烯酰氯(100mg,1.11mmol)滴加进去,室温搅拌2小时,过滤制备纯化(条件:乙腈/水(0.04%甲酸))得到015-1(23.5mg,收率:8.0%)。
LCMS:Rt:6.267min;MS m/z(ESI):402.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.50(s,1H),9.09(s,1H),7.89(s,1H),7.53(s,1H),7.04(s,1H),6.70-6.53(m,1H),6.17-6.12(m,1H),5.78-5.64(m,2H),4.08(dd,J=7.2,14.4Hz,2H),3.85-3.66(m,4H),2.34-2.15(m,2H),1.36(t,J=7.2Hz,3H).
实施例15化合物016-1的制备
Figure PCTCN2021073098-appb-000087
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000088
第一步:叔丁基(R)-3-((2,5-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯的合成016-1-b合成
室温下将叔丁基(R)-3-羟基吡咯烷-1-羧酸酯(295.0mg,1.58mmol)加入到二甲基亚砜(20.0mL)溶液内。氩气保护下在20℃下搅拌10分钟,然后将氢化钠(63.0mg,1.58mmol)加入反应液中搅拌30分钟,把2,4,5-三氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶(500.0mg,1.58mmol)加入反应体系中,将温度升高到55℃反应2小时,加水(30ml)淬灭,用乙酸乙酯(50ml*2)萃取,将有机相浓缩后经过柱(乙酸乙酯:石油醚=1:3)纯化得到016-1-b(680mg,收率:85.7%)。
LCMS:Rt:2.080min;MS m/z(ESI):447.0[M+H].
1H NMR(400MHz,DMSO-d 6)δ6.97(s,1H),5.94(s,1H),5.46(s,2H),4.90-4.81(m,1H),3.82-3.78(m,1H),3.51(t,J=8.0Hz,3H),3.39-3.32(m,1H),2.36-2.28(m,1H),1.47(s,9H),0.93-0.85(m,4H),0.03(s,9H).
第二步:叔丁基(R)-3-((5-氯-2-((1-((S)-四氢呋喃-3-基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯016-1-c的合成
在氮气保护下,于室温将Pd 2(dba) 3(62.0mg,0.067mmol),BINAP(41.6mg,0.067mmol)加入到016-1-b(340mg,0.67mmol),(R)-1-(四氢呋喃-3-基)-1H-吡唑-4-胺(102.5mg,0.67mmol)和碳酸铯(655.2mg,2.01mmol)的1,4-二氧六环(30mL)溶液中,反应体系升温至120℃搅拌16小时,冷却至室温。将反应液在减压下浓缩后经Waters 2767制备色谱仪,流动相A:1‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运 行至15min,10%ACN运行至20min结束纯化得到016-1-c(289mg,收率:69.5%)。
LCMS:Rt:1.670min;MS m/z(ESI):620.0[M+H].
第三步:5-氯-4-(((R)-吡咯烷-3-基)氧基)-N-(1-((S)-四氢呋喃-3-基)-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2-胺016-1-d的合成
室温下将016-1-c(289mg,0.46mmol)加入到三氟乙酸(2mL)的二氯甲烷溶液(4mL)中,反应液室温搅拌2小时。减压下将反应液浓缩,加入DIPEA(3mL)的甲醇溶液(4mL)中,反应液50℃搅拌过夜。减压下将反应液浓缩,通过硅胶柱色谱(乙酸乙酯:石油醚=1:10)纯化得到016-1-d(160mg,收率:89%)。
LCMS:Rt:1.130min;MS m/z(ESI):390.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.61(s,1H),9.43(s,1H),9.16(s,1H),7.94(t,J=1.2Hz,1H),7.58(s,1H),7.10(s,1H),5.72(s,1H),5.00(d,J=1.2Hz,1H),3.43-3.29(m,4H),2.40-2.23(m,6H),1.99(d,J=6.8Hz,2H).
第四步:1-((R)-3-((5-氯-2-((1-((S)-四氢呋喃-3-基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-基)丙-2-烯-1-酮016-1的合成
室温下将016-1-d(160mg,0.41mmol)溶解在丙酮(4mL)和H 2O(2mL)的混合液中,加入碳酸钾固体(169.7mg,1.38mmol)搅拌。将丙烯酰氯(51.8mg,0.57mmol)滴加进去,室温搅拌12小时后,过滤浓缩后制备纯化(Waters 2767制备色谱仪,流动相A:0.04%FA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到016-1(50.1mg,收率:27.5%)。
LCMS:Rt:7.940min;MS m/z(ESI):444.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.52(s,1H),9.13(s,1H),7.94(s,1H),7.55(s,1H),7.05(s,1H),6.66-6.62(m,1H),6.15(dd,J=8.0,16.0Hz,2H),5.71-5.64(d,J=8.0Hz,2H),4.97(s,1H),3.96(t,J=8.0Hz,4H),3.90-3.98(m,4H)2.38-2.21(m,4H).
实施例16化合物025-1的制备
(R)-1-(3-(5-氯-2-(((R)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯-5-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000089
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000090
第一步:(S)-3-对甲苯磺酰基四氢呋喃025-1-a的合成
室温下,将(S)-3-羟基四氢呋喃(1.0g,11.4mmol)加入到无水四氢呋喃(10.0mL)溶液中,零度下,加入氢化钠(0.9g,22.8mmol),在此温度搅拌0.5h后加入对甲苯磺酰氯(2.6g,13.6mmol),室温搅拌过夜。通过饱和食盐水(30.0mL)和乙酸乙酯(30.0mL*2)洗涤,浓缩所得粗产品,纯化(乙酸乙酯:石油醚=1:5)得到中间体025-1-a(2.4g,收率:87.3%)。
LCMS:Rt:1.428min;MS m/z(ESI):243.0[M+H].
1H NMR(400MHz,DMSO-d 6)δ7.82(d,J=8.0Hz,2H),7.50(d,J=8.0Hz,2H),5.14-5.11(m,1H),3.79-3.72(m,1H),3.70-3.64(m,3H),2.43(s,3H),2.13-2.03(m,1H),1.92-1.86(m,1H).
第二步:(R)-3-(4-硝基吡唑)四氢呋喃025-1-b的合成
室温、在氮气保护下,将中间体025-1-a(2.4g,9.6mmol)、4-硝基吡唑(0.9g,8.0mmol)和碳酸铯(5.2g,16.0mmol)依序加入DMF(15.0mL)中,反应升温至100℃下搅拌12h。待反应体系降至室温后,减压浓缩,粗产品纯化(乙酸乙酯:石油醚=1:5),得到中间体025-1-b(1.3g,收率:71.6%)。
LCMS:Rt:1.068min;MS m/z(ESI):184.0[M+H].
1H NMR(400MHz,DMSO-d 6)δ8.92(s,1H),8.30(s,1H),5.13-5.09(m,1H),4.02-3.93(m,3H),3.85-3.79(m,1H),2.44-2.32(m,2H).
第三步:(R)-3-(4-氨基吡唑)四氢呋喃025-1-c的合成
室温下,中间体025-1-b(1.3g,8.5mmol)溶于无水乙醇中,加入Pd/C(130mg),在氢气的氛围中,反应体系于室温搅拌3h,过滤浓缩,得到粗品纯化(乙酸乙酯:石油醚=1:4)得到025-1-c(1.0g,收率:92%)。
LCMS:Rt:0.395min;MS m/z(ESI):154.0[M+H].
1H NMR(400MHz,DMSO-d 6)δ7.05(s,1H),6.92(s,1H),4.82-4.78(m,1H),3.93-3.74(m,6H),2.32-2.23(m,1H),2.18-2.11(m,1H).
第四步:(R)-3-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸叔丁酯025-1-f的合成
室温下,将2,4,5-三氯-7H-吡咯并[2,3-D]嘧啶025-1-d(3.8g,17.2mmol)和(R)-3-氨基吡咯烷-1-甲酸叔丁酯025-1-e(3.8g,20.6mmol)溶于乙醇溶液(50.0mL)中,加入DIPEA(4.4mL,34.4mmol),升温至110℃搅拌12h。降温后反应液减压浓缩,所得粗产品纯化得到中间体025-1-f(5.0g,收率:78.6%),不经纯化直接用于下一步反应。
LCMS:Rt:1.868min;MS m/z(ESI):372[M+H].
1H NMR(400MHz,DMSO-d 6)δ12.16(s,1H),7.36(s,1H),6.67-6.63(m,1H),4.71-4.62(m,1H),3.68-3.63(m,1H),3.45-3.42(m,1H),3.35-3.24(m,2H),2.52-2.51(m,1H),2.22-2.19(m,1H),1.41(s,9H).
第五步:(R)-1-(3-(5-氯-2-(((R)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯)甲酸叔丁酯025-1-g的合成
室温下,中间体(R)-3-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸叔丁酯025-1-f(1.0g,2.7mmol)和中间体(R)-3-(4-氨硝基吡唑)四氢呋喃025-1-c(495mg,3.2mmol)加入异丙醇(15.0mL)的闷罐中,后加入三氟乙酸(307mg,2.7mmol),反应升温110℃下搅拌12h。反应体系降至室温后,减压浓缩纯化后得到中间体025-1-g(400mg,收率:30.5%)。
LCMS:Rt:1.333min;MS m/z(ESI):489.3[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.24(s,1H),8.73(s,1H),7.93(s,1H),7.50(s,1H),6.92(s,1H),6.02(s,1H),5.76(s,1H),4.94(s,1H),4.70-4.64(m,1H),4.04-3.94(m,2H),3.89-3.70(m,3H),3.44(s,1H),3.24-3.20(m,1H),2.41-2.32(m,1H),2.21(s,2H),2.05-1.99(m,1H),1.40(d,J=8Hz,9H).
第六步:(R)-1-(3-(5-氯-2-(((R)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯025-1-h的合成
中间体025-1-g(400mg,819.7mmol)溶于二氯甲烷中(5.0mL),加入HCl/二氧六环(4M,4.0mL)中,室温搅拌2h,减压浓缩,得到中间体025-1-h(420mg),不用纯化,直接用于下一步。
LCMS:Rt:0.347min;MS m/z(ESI):432.3[M+H]。
第七步:(R)-1-(3-(5-氯-2-(((R)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯-5-基)丙-2-烯-1-酮025-1的合成
0℃下,将中间体025-1-h(420mg)溶于丙酮和水中(1:1,5.0mL,加入碳酸钾(243.0mg,1.6mol),在此温度下搅拌10分钟。然后缓慢滴加含丙烯酰氯(95.5mg,983.6mmol)的丙酮溶液(0.5mL)。反应液温度升至室温,并搅拌2小时后。反应液通过饱和食盐水(5.0mL)洗涤,所得粗品用DMF溶解,通过经Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化后得到产物025-1(261.6mg,收率:70.7%)。
LCMS:Rt:4.557min;MS m/z(ESI):443.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.24(s,1H),8.73(s,1H),7.93(s,1H),7.50(s,1H),6.92(s,1H),6.66-6.53(m,1H),6.17-6.06(m,2H),5.70-6.4(m,1H),4.93-4.66(m,2H),4.06-3.74(m,5H),3.69-3.37(m,3H),2.40-2.03(m,4H).
实施例17化合物025-2的制备
(S)-1-(3-(5-氯-2-(((R)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯-5-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000091
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000092
第一步:(R)-3-对甲苯磺酰基四氢呋喃025-2-a的合成
室温下,将(R)-3-羟基四氢呋喃(1.0g,11.4mmol)加入到无水四氢呋喃溶液中,0℃下,加入氢化钠(0.9g,22.8mmol),在此温度搅拌0.5h后加入对甲苯磺酰氯(2.6g,13.6mmol),室温搅拌过夜。通过饱和食盐水和乙酸乙酯洗涤两遍,浓缩所得粗产品纯化(PE:EA=6:1)得到中间体025-2-a(2.5g,收率:90.9%)。
LCMS:Rt:1.448min;MS m/z(ESI):243.0[M+H].
1H NMR(400MHz,DMSO-d 6)δ7.81(d,J=8.0Hz,1H),7.49(d,J=8.0Hz,1H),5.12(s 1H),3.78-3.72(m,1H),3.69-3.64(m,3H),2.43(s,3H),2.10-2.03(m,1H),1.92-1.85(m,1H).
第二步:(S)-3-(4-硝基吡唑1-基)四氢呋喃025-2-b的合成
室温下,将中间体025-2-a(2.5g,10.3mmol)和4-硝基吡唑(0.97g,8.6mmol)溶入到DMF(15mL)中,加入碳酸铯(5.6g,17.2mmol),在氮气保护下,反应体系在100℃下搅拌12h。待反应体系降至室温后,减压浓缩,粗产品纯化(PE:EA=10:1),得到中间体025-2-b(1.5g,收率:82.7%)。
LCMS:Rt:1.143min;MS m/z(ESI):184.0[M+H].
第三步:(S)-3-(4-氨基吡唑1-基)四氢呋喃025-2-c的合成
室温下,中间体025-2-b(1.5g,8.2mmol)溶于无水乙醇(30mL)中,加入湿Pd/C(150mg),在氢气的氛围中反应体系于室温搅拌3h。过滤浓缩,粗产品纯化(DCM:MeOH=10:1),得到中间体025-2-c(1.0g,收率:92%)。
LCMS:Rt:0.440min;MS m/z(ESI):154.0[M+H].
第四步:(R)-1-(3-(5-氯-2-(((S)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯-5-基)甲酸叔丁酯025-2-e的合成
室温下,将中间体025-2-d(1.0g,2.7mmol)和中间体025-2-c(495mg,3.2mmol)溶入到异丙醇(15.0mL)中,加入三氟乙酸(307mg,2.7mmol),在闷罐中,反应体系在110℃下搅拌12h。反应降至室温后,减压浓缩纯化(EA:PE=1:1),得到中间体025-2-e(570mg,收率:43%)。
LCMS:Rt:1.518min;MS m/z(ESI):489.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.35(s,1H),8.91(s,1H),7.94(s,1H),7.51(s,1H),6.95(s,1H),6.17(s,1H),5.76(s,1H),4.95(s,1H),4.71-4.66(m,1H),4.00-3.94(m,2H),3.89-3.77(m,3H),3.44(s,1H),3.26-3.22(m,1H),2.41-2.04(m,4H),1.40(d,J=8Hz,9H).
第五步:(R)-1-(3-(5-氯-2-(((S)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯025-2-f的合成
中间体025-2-e(570mg,819.7mmol)溶于二氯甲烷中(5.0mL),加入HCl/二氧六环(4.0M,4.0mL)中,室温搅拌2h,减压浓缩,得到中间体025-2-f(420mg,收率:92.6%),直接用于下一步。
LCMS:Rt:1.440min;MS m/z(ESI):389.2[M+H].
第六步:(S)-1-(3-(5-氯-2-(((R)-1-四氢呋喃-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-吡咯-5-基)丙-2-烯-1-酮025-2的合成
0℃下,将粗品025-2-f溶于丙酮和水中(1:1,5.0mL),加入碳酸钾(322.9mg,2.4mol),在此温度下搅拌10分钟。然后缓慢滴加含丙烯酰氯(130.3mg,1.5mmol)的丙酮溶液(0.5mL)。反应液温度升至室 温,并搅拌2小时。反应液通过饱和食盐水(10.0mL)洗涤,所得粗品用DMF溶解,通过经Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至16min,10%ACN运行至20min结束纯化得到产物025-2(154.9mg,收率:41.8%)。
LCMS:Rt:4.44min;MS m/z(ESI):443.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.25(s,1H),8.74(s,1H),7.93(s,1H),7.50(s,1H),6.92(s,1H),6.66-6.53(m,1H),6.17-6.06(m,1H),5.70-5.64(m,1H),4.93-4.66(m,2H),4.06-3.78(m,5H),3.67-3.37(m,4H),2.37-2.06(m,4H).
实施例18化合物026的制备
(R)-1-(3-((5-氯-2-((1-(氧杂环丁烷-3-基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)四氢吡咯-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000093
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000094
第一步:4-硝基-1-(环氧丁烷-3-基)-1H-吡唑026-e的合成
将3-溴环氧丁烷(400mg,2.918mmol)缓慢加入到4-硝基-1H-吡唑(330mg,2.918mmol)和碳酸铯(1.14g,3.502mmol)的DMF(5.0mL)溶液中,加热到110℃,在此温度下搅拌过夜。待反应体系降至室温后,加入5.0mL冰水淬灭,将混合液用乙酸乙酯萃取,合并萃取液,用氯化钠溶液洗涤有机相,有机相用无水硫酸钠干燥,过滤,滤液在减压下浓缩得到粗产品,经柱层析纯化(DCM/甲醇=100:1)得到产物026-e(350mg,收率:70.9%)。
1H NMR(400MHz,CDCl 3)δ8.31(s,1H),8.17(s,1H),5.49-5.43(m,1H),5.10-5.03(m,4H).
第二步:1-(环氧丁烷-3-基)-1H-吡唑-4-胺026-b的合成
将湿钯/碳(10mg)加入到4-硝基-1-(环氧丁烷-3-基)-1H-吡唑026-e(350mg,2.071mmol)的乙醇(10.0mL)溶液中,在氢气下室温搅拌过夜。将混合液在减压下过滤,滤液浓缩得到中间体026-b(265mg,收率: 92.0%)。
LCMS:Rt:0.355min;MS m/z(ESI):140.4[M+H].
第三步:叔丁基(R)-3-((5-氯-2-((1-(氧杂环丁烷-3-基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)四氢吡咯-1-羧酸酯026-c的合成
在50mL微波管中,将三氟乙酸(91.9mg,0.806mmol)缓慢加入到叔丁基(R)-3-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)四氢吡咯-1-羧酸酯026-a(300mg,0.806mmol)和1-(氧杂环丁烷-3-基)-1H-吡唑-4-胺026-b(112mg,0.806mmol)的异丙醇(3.0mL)溶液中,在氩气下将反应体系密封,加热到100℃,在此温度下搅拌过夜。待反应体系降至室温后,加入5mL冰水淬灭,将混合液在减压下浓缩得到粗产品,经Waters2767制备色谱仪,流动相A:0.5‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化得到化合物026-c(256mg,收率:66.8%)。
第四步:(R)-5-氯-N 2-(1-(氧杂环丁烷-3-基)-1H-吡唑-4-基)-N 4-(四氢吡咯-3-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺026-d的合成
0℃下将三氟乙酸(2mL)加入到中间体026-c(256mg,0.539mmol)的二氯甲烷溶液(5.0mL)中,室温搅拌2小时。减压下将反应液浓缩得到中间体026-d(202mg,收率:99%),不经纯化直接用于下一步反应。
第五步:(R)-1-(3-((5-氯-2-((1-(氧杂环丁烷-3-基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)四氢吡咯-1-基)丙-2-烯-1-酮026的合成
将中间体026-d(202mg,0.539mmol)溶解在丙酮(5.0mL)和水(2.0mL)的混合液中,加入碳酸钾(186.2mg,1.348mmol)。丙烯酰氯(73mg,0.808mmol)缓慢滴加入后室温搅拌2小时。反应液过滤,制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到产物026(7.78mg,收率:12%)。
LCMS:Rt:4.567min;MS m/z(ESI):429.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.26(brs,1H),8.79(s,1H),8.04(s,1H),7.62(s,1H),6.92(s,1H),6.66-6.53(m,1H),6.16-6.07(m,2H),5.67(t,J=7.0,7.0,1H),5.51-5.44(m,1H),4.92-4.83(m,4H),4.05-3.84(m,1H),3.77-3.62(m,2H),3.57-3.38(m,2H),2.36-2.23(m,1H),2.17-2.00(m,1H).
实施例19化合物027的制备
1-((3S,4R)-3-((2-((1-(2-羟乙基)-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000095
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000096
第一步:2-(4-硝基-1H-吡唑-1-基)乙烷-1-醇027-f的合成
将2-溴代-1-乙醇(1.0g,8.0mmol),4-硝基-1H-吡唑(1.35g,12mmol)和Cs 2CO 3(7.8g,24mmol)加入到DMF(20.0mL)溶液内,在100℃下搅拌12小时。降温加水(50.0mL)终止反应,乙酸乙酯(50.0mL*2)萃取。有机相用饱和食盐水洗,减压浓缩后纯化(PE/EA=3/1)得到中间027-f(810mg,收率:64.5%)。
LCMS:Rt:1.270min;MS m/z(ESI):158.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ8.81(s,1H),8.26(s,1H),4.98(s,1H),4.21(t,2H),3.77(t,2H).
第二步:2-(4-氨基-1H-吡唑-1-基)乙烷-1-醇027-d的合成
室温氢气保护下,将中间体027-f(400mg,2.54mmol),湿Pd/C(40.0mg)加入到乙醇(6.0mL)溶液中。氢化反应搅拌3小时后,反应液过滤,溶液减压浓缩得到中间体027-d(314mg,收率:97.3%)。
LCMS:Rt:0.34min;MS m/z(ESI):128.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ7.01(s,1H),6.88(s,1H),4.79(s,1H),3.93(t,2H),3.76(s,1H),3.63(d,2H).
第三步:叔丁基(R)-3-((2,5-二氯-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸酯027-c的合成
室温下将中间体027-a(4.89g,22.0mmol)、027-b(4.1g,22.0mmol)和DIPEA(7.7g,60.0mmol)加入到异丙醇(20.0mL)中。闷罐加热到110℃,反应16小时回至室温。减压浓缩纯化(PE/EA=5:1),得到中间体027-c(6.5g,收率:87.6%)。
LCMS:Rt:1.545min;MS m/z(ESI):372.1[M+H].
1H NMR(400MHz,CDCl 3)δ11.91(s,1H),7.00(s,1H),6.01(s,1H),4.86(s,1H),3.81(s,1H),3.51(s,3H),2.33(s,1H),1.99(s,1H),1.48(s,9H).
第四步:叔丁基(R)-3-((5-氯-2-((1-(2-羟乙基)-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸酯027-e的合成
室温下将中间体027-c(200mg,0.54mmol),2-(4-氨基-1H-吡唑-1-基)乙烷-1-醇027-d(82mg,0.64mmol)加入到三氟乙酸(92.3mg,0.81mmol)的异丙醇溶液(5.0mL)中,在闷罐反应95℃搅拌18小时后降至室温。减压下将反应液浓缩,纯化得到中间体027-e(150mg,收率:60.0%)。
LCMS:Rt:1.41min;MS m/z(ESI):463.1[M+H].
第五步:(R)-2-(4-((5-氯-4-(吡咯烷-3-基氨基)-7H吡咯并[2,3-d]嘧啶-2-基)氨基)-1H-吡唑-1- 基)乙烷-1-醇027-f的合成
将中间体027-e(150mg,0.32mmol)溶解在三氟乙酸(2.0mL)和DCM(4.0mL)的混合液中,室温搅拌2小时后浓缩得到粗品中间体027-f(149mg),不纯化直接做下一步反应。
LCMS:Rt:1.235min;MS m/z(ESI):363.4[M+H].
第六步:(R)-1-(3-((5-氯-2-((1-(2-羟乙基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮027的合成
将中间体027-f(149mg,0.41mmol)溶解在丙酮(4.0mL)和H 2O(2.0mL)的混合液中,加入碳酸钾固体(113.8mg,0.825mmol)室温搅拌,将丙烯酰氯(27.2mg,0.30mmol)滴加进去,室温搅拌2小时,过滤制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到产物027(7.89mg,收率:5%)。
LCMS:Rt:4.06min;MS m/z(ESI):417.3[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.84(s,1H),9.67(s,1H),7.94(s,1H),7.56(s,1H),7.07(s,1H),6.66-6.52(m,1H),6.15-6.13(d,J=16.4Hz,1H),5.70(t,J=2.0Hz,1H),4.86-4.75(m,2H),4.10(s,7H),3.51-3.49(m,1H),2.38-2.10(m,3H).
实施例20化合物028的制备
1-((2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000097
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000098
第一步:叔丁基(2S,4R)-4-((2,5-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-羧酸酯028-c的合成
室温下分别将011-1-b(351mg,1.0mmol),028-b(200mg,1.0mmol)和DIPEA(0.3mL,2.0mmol)加入到异丙醇(10.0mL)溶液内。将反应体系密封并加热到100℃,在此温度下搅拌16小时。反应液减压浓缩得到粗品,纯化得到中间体028-c(440mg,收率:85.4%)。
LCMS:Rt:2.520min;MS m/z(ESI):516.2[M+H].
1H NMR(400MHz,CDCl 3)δ7.00(s,1H),6.01(d,J=7.2Hz,1H),5.49(s,2H),4.79(dd,J=13.2,6.4Hz,1H),4.00(s,2H),3.55(dd,J=14.0,8.4Hz,2H),3.30(dd,J=11.2,5.6Hz,1H),2.66-2.59(m,1H),2.28(s,2H),1.48(s,9H),0.98-0.86(m,4H),0.02(s,9H).
第二步:叔丁基(2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧 基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-羧酸酯028-e的合成
室温在氮气保护下,将Pd 2(dba) 3(78mg,0.085mmol),BINAP(53mg,0.085mmol)加入到中间体028-c(440mg,0.85mmol),1-乙基-1H-吡唑-4-胺028-d(190mg,1.70mmol)和碳酸铯(830mg,2.55mmol)的1,4-二氧六环(30.0mL)溶液中,反应体系升温至100℃搅拌16小时,冷却至室温。反应液减压浓缩后纯化得到中间体028-e(400mg,收率:87.1%)。
LCMS:Rt:2.325min;MS m/z(ESI):591.3[M+H].
1H NMR(400MHz,CDCl 3)δ7.89(s,1H),7.62(s,1H),6.79(s,1H),6.61(s,1H),5.81(d,J=6.4Hz 1H),5.48(s,2H),4.70(s,2H),4.20(dd,J=11.2,7.2Hz,2H),4.03(s,2H),3.60(t,J=8.4Hz,2H),3.32(dd,J=11.2,5.6Hz,1H),2.67-2.60(m,1H),1.49(s,9H),1.38(s,3H),1.26(t,J=7.2Hz,3H),0.91(t,J=8.0Hz,2H),0.02(s,9H).
第三步:(2-((1-乙基-1H-吡唑-4-基)氨基)-4-((3R,5S)-5-甲基吡咯烷-3-基)氨基)-7H吡咯并[2,3-d]嘧啶-7-基)甲醇028-f的合成
室温下将中间体028-e(400mg,0.68mmol)加入到三氟乙酸(5.0mL)的二氯甲烷溶液(10.0mL)中,反应液室温搅拌2小时。减压下将反应液浓缩得到中间体028-f(264mg,收率:99%)。
LCMS:Rt:0.900min;MS m/z(ESI):391.1[M+H].
第四步:5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-((3R,5S)-5-甲基吡咯烷-3-基)-7H吡咯并[2,3-d]嘧啶-2,4-二胺028-g的合成
室温下将中间体028-f(264mg,0.68mmol)加入到DPIEA(5.0mL)的甲醇溶液(10.0mL)中,反应液50℃搅拌过夜。减压下将反应液浓缩,得到中间体028-g(250mg,收率:99%)。
LCMS:Rt:0.840min;MS m/z(ESI):361.1[M+H].
第五步:1-((2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮028的合成
室温下,中间体028-g(250mg,0.68mmol)溶解在丙酮(10.0mL)和H 2O(5.0mL)的混合液中,加入碳酸钾(272mg,2.04mmol),将丙烯酰氯(90mg,1.0mmol)滴加进去,室温搅拌2小时。反应液过滤,制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至16min,10%ACN运行至20min结束纯化)得到028(28mg,纯度:94%)。
LCMS:Rt:6.892min;MS m/z(ESI):413.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.32(s,1H),8.86(brs,1H),7.86(s,1H),7.48(s,1H),6.95(s,1H),6.68-6.53(m,1H),6.31(brs,1H),6.12(t,J=14.4Hz,1H),5.72-5.64(m,1H),4.69-4.59(m,1H),4.27-4.01(m,4H),2.01-1.96(m,1H),1.88-1.81(m,1H),1.33-1.30(m,7H).
实施例21化合物029、化合物029-1及化合物029-2的制备
1-(7-(5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺环[2.4]庚烷-5-基)丙-2-烯-1-酮029
(R)-1-(7-(5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺环[2.4]庚烷-5-基)丙-2-烯-1-酮029-1
(S)-1-(7-(5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺环[2.4]庚烷-5-基)丙-2-烯-1-酮029-2
Figure PCTCN2021073098-appb-000099
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000100
第一步:7-((2,5-二氯-7-((2-(三甲基甲硅烷基)乙氧基)7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺环[2.4]庚烷-5-羧酸苄酯029-c的合成
室温下,将029-b(650mg)溶于异丙醇(15.0mL)中,然后加入2,4,5-三氯-7-((2-(三甲基甲硅烷基)乙氧基)-7H-吡咯并[2,3-D]嘧啶011-1-b(628.8mg,1.8mmol)和DIPEA(462.9mg,3.6mmol)。反应体加热至120℃。搅拌12h后冷却至室温后,浓缩所得粗品纯化(石油醚:乙酸乙酯=30:1~10:1),得到中间体029-c(634mg,收率:63.4%)。
LCMS:Rt:2.053min;MS m/z(ESI):562.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ7.67(s,1H),7.47-7.38(m,5H),6.57(d,J=6.6Hz,1H),5.49(s,2H),5.15(s,2H),4.55–4.29(m,1H),3.96–3.83(m,1H),3.82–3.68(m,2H),3.61–3.52(m,2H),1.41(dd,J=18.0,13.5Hz,3H),0.92–0.78(m,4H),0.00(s,9H).
第二步:7-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺环[2.4]庚烷-5-羧酸苄酯029-e的合成
室温在氮气保护下,将中间体029-c(634mg,1.1mmol)和1-甲基-1H-吡唑-4-胺029-d(163.0mg,1.7mmol)溶于1,4-二氧六环(20.0mL),然后在搅拌下,加入Pd(dppf)Cl 2(103.5mg,0.1mmol)、BINAP(141.0mg,0.2mmol)和碳酸铯(736.4mg,2.2mmol),反应体系升温至120℃,搅拌12小时后,冷却至室温,过滤,滤饼用乙酸乙酯(60.0mL)洗涤后浓缩,纯化(石油醚:乙酸乙酯=10:1~2:1)得到029-e(520mg,收率:74.1%)。
LCMS:Rt:2.017min;MS m/z(ESI):623.2[M+H].
1H NMR(400M Hz,DMSO-d 6)δ9.05(d,J=6.1Hz,1H),8.01(brs,1H),7.61(d,J=2.4Hz,1H),7.48(t,J=5.7Hz,2H),7.43(t,J=5.4Hz,3H),7.28(s,1H),6.00(s,1H),5.53(s,2H),5.20(s,2H),4.51–4.39(m,1H),3.99–3.82(m,5H),3.82–3.69(m,1H),3.63(t,J=8.1Hz,2H),3.44–3.37(m,1H),1.02–0.88(m,4H),0.88–0.75(m,2H),0.00(d,J=2.4Hz,9H).
第三步:5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-5-氮杂螺环[2.4]庚烷-7H-吡咯并[2,3-d]嘧啶029-f的合成
在0℃下,将三氟乙酸(4.5mg,3.05mmol)加入到029-e(520mg,0.84mmol)的二氯甲烷溶液(5.0mL)中,80℃搅拌72小时。LCMS监测反应结束后,减压下将反应液浓缩,粗产品溶于THF/H 2O(6.0mL,2:1)的混合液中,加入LiOH(175.4mg,4.2mmol)室温搅拌3h,过滤粗品纯化后得中间体029-f(120mg,收率:40%)。
LCMS:Rt:0.693min;MS m/z(ESI):359.5[M+H].
第四步:(R,S)-1-(7-(5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-5-氮杂螺环[2.4]庚烷-5-基)丙-2-烯-1-酮029-1和029-2的合成
在0℃下,将中间体029-f(120mg,0.34mmol)加入到四氢呋喃(4.0mL)和H 2O(2.0mL)的混合液中,再加入磷酸钾(148.6mg,0.68mmol),搅拌10分钟后,缓缓滴加丙烯酰氯的四氢呋喃溶液(46.2mg, 0.51mmol),室温搅拌2小时,过滤,滤液浓缩后制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到化合物029,化合物029经手性拆分[column:DAICEL CHIRALCEL OD(250mm*30mm,10um);mobile phase:A:CO 2;B:MeOH(0.1%DEA),30%-30%,20min]得到产物029-1(18.9mg,收率:23%)和029-2(25.3mg,收率:29%)。
029-1:
LCMS:Rt:5.107min;MS m/z(ESI):413.1[M+H].
1H NMR 400MHz,DMSO-d 6)δ11.30(s,1H),8.78(s,1H),7.82(s,1H),7.48(d,J=3.1Hz,1H),6.96(d,J=2.4Hz,1H),6.56(ddd,J=19.9,16.7,10.3Hz,1H),6.15(dd,J=16.8,2.4Hz,1H),5.91(s,1H),5.68(ddd,J=10.3,4.9,2.3Hz,1H),4.54–4.30(m,1H),3.87(s,1H),3.84–3.82(m,1H),3.79(s,3H),3.67–3.54(m,1H),3.52–3.39(m,1H),0.90–0.59(m,4H).
029-2:
LCMS:Rt:5.064min;MS m/z(ESI):413.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.26(s,1H),8.70(s,1H),7.81(s,1H),7.47(d,J=2.5Hz,1H),6.94(d,J=1.6Hz,1H),6.56(ddd,J=20.1,16.8,10.3Hz,1H),6.15(dd,J=16.8,2.3Hz,1H),5.82(d,J=6.5Hz,1H),5.68(ddd,J=10.4,5.0,2.4Hz,1H),4.58–4.20(m,1H),4.10–3.89(m,1H),3.88–3.82(m,1H),3.78(s,3H),3.59(dd,J=41.4,11.5Hz,1H),3.41(d,J=12.5Hz,1H),0.93–0.63(m,4H).
实施例22化合物030的制备
1-((3S,4S)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000101
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000102
第一步:(3S,4S)-叔丁基3-((2,5-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-4-氟吡咯烷-1-羧酸酯030-b的合成
在微波管中将2,4,5-三氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶011-1-b(305mg,0.87mmol)加至(3S,4S)-叔丁基3-氨基-4-氟吡咯烷-1-羧酸酯030-a(265mg,1.3mmol)和DIPEA(224mg, 1.74mmol)的EtOH溶液(10.0mL)。反应液加热到110℃搅拌6小时,冷却到室温。四氢呋喃(10.0mL)加入并过滤,滤饼用四氢呋喃(10.0mL*2)洗涤,滤液减压浓缩后得到粗品。纯化得到中间体030-b(378.2mg,收率:87.3%)。
LCMS:Rt:1.980min;MS m/z(ESI):518.3[M+H].
第二步:(3S,4S)-叔丁基3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-4-氟吡咯烷-1-羧酸酯030-d的合成
室温下在单口烧瓶中将TFA(71mg,0.729mmol)加入到030-b(378.2mg,0.729mmol)和030-c(162mg,1.458mmol)的异丙醇(10.0mL)溶液中。升温至95℃搅拌过夜后冷却至室温,将此溶液在减压下浓缩,所得粗产品用硅胶柱纯化得到中间体030-d(100mg,收率:23%)。
LCMS:Rt:2.125min;MS m/z(ESI):595.7[M+H].
第三步:5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-((3S,4S)-4-氟吡咯烷-3-基)-7H吡咯并[2,3-d]嘧啶-2,4-二胺030-e的合成
室温下,将TFA(2.5mL)加入到中间体030-d(100mg,0.168mmol)的二氯甲烷(5.0mL)溶液中。搅拌1个小时后,减压浓缩。二氯甲烷加入(5.0mL),在减压下浓缩除去多余TFA。所得粗品加入甲醇(5.0mL)和DIPEA(1.0mL)。反应液升温至50℃,搅拌一个小时后,减压浓缩得到粗品030-e,直接用于下一步反应。
LCMS:Rt:0.42min;MS m/z(ESI):365.0[M+H].
第四步:1-((3S,4S)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氨基)-4-氟吡咯烷-1-基)丙-2-烯-1-酮030的合成
将粗品中间体030-e(0.519mmol)溶于丙酮(3.0ml)和水(3.0mL)的混合溶剂中,用碳酸钾调节pH值到10,反应液冷却至0℃,然后将丙稀酰氯(54mg,0.6mmol)溶解在丙酮(1.0mL)中滴加到反应液中。滴加完毕后,反应液在0℃下搅拌1h。反应结束后,将反应液过滤后直接送制备Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化,得到产物030(7.04mg)。
LCMS:Rt:1.51min;MS m/z(ESI):419.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.28(s,1H),8.75(s,1H),8.44(s,1H),7.90(s,1H),7.47(s,1H),6.94(s,1H),6.66-6.60(m,1H),6.25-6.16(m,2H),5.72(d,J=10.4Hz,1H),5.52-5.33(m,1H),4.94-4.80(m,1H),4.05-4.01(m,2H),3.96-3.78(m,3H),1.35(t,J=7.2Hz,3H).
实施例23化合物031的制备
1-((3S,4R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-4-羟基吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000103
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000104
第一步:(3S,4R)-3-((2,5-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-4-羟基吡咯烷-1-羧酸叔丁酯031-c的合成
将2,4,5-三氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶011-1-b(370mg,1.05mmol)和(3S,4R)-3-氨基-4-羟基吡咯烷-1-羧酸叔丁酯031-b(234mg,1.16mmol)加入到DIPEA(338mg,2.62mmol)的异丙醇(8.0mL)溶液中,反应液升温到110℃下搅拌16小时,将反应液用水(30.0mL)稀释,然后用乙酸乙酯(10.0mL*2)萃取。将有机相减压浓缩后经柱层析(石油醚:乙酸乙酯=1:1)纯化得到中间体031-c(502mg,收率:80%)。
LCMS:Rt:1.843min;MS m/z(ESI):518.1[M+H].
第二步:(3S,4R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-4-羟基吡咯烷-1-羧酸叔丁酯031-e的合成
将中间体031-c(450mg,0.87mmol),1-乙基-1H-吡唑-4-胺031-d(125mg,1.13mmol),Pd(dppf)Cl 2(79mg,0.087mmol),Xantphos(54.1mg,0.087mmol)和碳酸铯(709mg,2.17mmol)加入到1,4-二氧六环(20.0mL)中,反应液在氩气保护下120℃搅拌反应16小时。将反应液用硅藻土过滤,并用乙酸乙酯(20.0mL)洗涤滤饼。滤液减压浓缩后,纯化(石油醚/乙酸乙酯=1:2)得到031-e(500mg,收率:88%)。
LCMS:Rt:1.837min;MS m/z(ESI):593.3[M+H].
第三步:(3R,4S)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-3-醇031-f的合成
25℃下将031-e(500mg,0.84mmol)加入到三氟乙酸(3.0mL)中,反应液25℃下搅拌30分钟。减压下将反应液浓缩,加入一水合氢氧化锂(157mg,3.7mmol的四氢呋喃(3.0mL)/水(1.0mL)溶液中,反应液在25℃搅拌1小时。减压下将反应液浓缩,通过Prep-TLC(Flash)纯化得到031-f(90mg,收率:82%)。
LCMS:Rt:1.05min;MS m/z(ESI):363.2[M+H].
第四步:1-((3S,4R)-3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-4-羟基吡咯烷-1-基)丙-2-烯-1-酮031的合成
将031-f(200mg,0.61mmol)溶解在四氢呋喃(8.0mL)和水(2.0mL)的混合液中,加入磷酸钾(323.5mg,1.52mmol)搅拌。将丙烯酰氯(60mg,0.67mmol)的四氢呋喃(0.5mL)溶液逐滴滴加进去,搅拌1小时。反应液减压浓缩后经制备纯化(Waters 2767制备色谱仪,流动相A:1‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到031(85.0mg,收率33%)。
1H NMR(400MHz,DMSO-d 6)δ11.31(s,1H),8.81(s,1H),7.87(s,1H),7.47(d,J=2.0Hz,1H),6.96(t,J=2.4Hz,1H),6.61-6.55(m,1H),6.41(s,1H),6.18-6.12(m,1H),5.71-5.64(m,1H),4.68-4.55(m,1H),4.42-4.33(m,1H),3.84(dd,J=4.4Hz,J=11.6Hz,1H),3.61-3.51(m,2H),3.40-3.20(m,1H),1.32(t,J=6.4Hz,3H).
实施例24化合物032的制备
(R,E)-1-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)四氢吡咯-1-基)-4-氟-丁-2-烯-1-酮
Figure PCTCN2021073098-appb-000105
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000106
第一步:(R,E)-1-(3-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)四氢吡咯-1-基)-4-氟-丁-2-烯-1-酮032的合成
在0℃下,将(R-5-氯-N 2-(1-乙基-1H-吡唑-4-基)-N 4-(四氢吡咯-3-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺8f(80.0mg,0.231mmol),(E)-4-氟-丁-2-烯酸(032-a,28.8mg,0.276mmol)和DIPEA(74.6mg,0.578mmol)加入到无水DMF(5.0mL)中,然后将HATU(131.7mg,0.346mmol)搅拌下加入到反应液中,室温下搅拌反应16小时。将反应液倒入到冰水(5.0mL)中,混合液减压浓缩后制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到032(6.28mg,收率:6.3%)。
LCMS:Rt:4.888min;MS m/z(ESI):433.1[M+H].
1H NMR(400M Hz,DMSO-d 6)δ11.81(brs,1H),9.62(brs,1H),7.92(s,1H),7.55(s,1H),7.08(s,1H),6.95(brs,1H),6.82-6.72(m,1H),6.48(dd,J=29.2,16.4Hz,1H),5.19(dd,J=10.8,5.6Hz,1H),5.07(dd,J=9.2,3.6Hz,1H),4.86-4.72(m,1H),4.08(q,J=7.2Hz,2H),3.88-3.73(m,1H),3.67-3.62(m,1H),3.50-3.42(m,1H),2.38-2.33(m,1H),2.27-2.21(m,1H),2.14-2.05(m,1H),1.35(t,J=7.2Hz,3H).
实施例25化合物033的制备
(R)-1-(4-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)氮杂环庚烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000107
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000108
第一步:(R)-4-((2,5-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)氮杂环庚烷-1-甲酸叔丁酯033-b的合成
室温下,在闷罐里分别将2,4,5-三氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶011-1-b(500mg,1.42mmol),(R)-4-氨基氮杂环庚烷-1-甲酸叔丁酯033-a(335mg,1.56mmol)和DIPEA(818mg,4.26mmol)加入到异丙醇(10.0mL)溶液内。反应液在110℃下搅拌16小时。降温后,反应液减压浓缩纯化(乙酸乙酯/石油醚=10/1)得到中间体033-b(680mg,收率:90%)。
LCMS:Rt:1.78min;MS m/z(ESI):530.2[M+H].
第二步:(R)-4-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)氮杂环庚烷-1-甲酸叔丁酯033-d的合成
室温在氮气保护下,将Pd 2(dba) 3(113.5mg,0.12mmol),BINAP(77.0mg,0.12mmol)加入到中间体033-b(660mg,1.24mmol),1-乙基-1H-吡唑-4-胺033-c(145mg,1.49mmol)和碳酸铯(1.2g,3.72mmol)的1,4-二氧六环(25.0mL)溶液中。反应体系升温至120℃搅拌16小时,冷却至室温。反应液减压浓缩后纯化(乙酸乙酯/石油醚=1/2)得到中间体033-d(730mg,收率:95%)。
LCMS:Rt:1.925min;MS m/z(ESI):591.8[M+H].
1H NMR(400MHz,DMSO-d 6)δ8.92(s,1H),7.98(brs,1H),7.59(s,1H),7.20(s,1H),6.01(brs,1H),5.48(s,2H),4.34(s,1H),3.90(s,3H),3.61(t,J=7.6Hz,3H),3.48(s,2H),3.32(s,1H),2.19(s,1H),2.04(s,1H),1.90(t,J=5.6Hz,2H),1.79(d,J=8.4Hz,2H),1.51(s,9H),0.94(t,J=8.4Hz,2H),0.00(s,9H).
第三步:(R)-N 4-(氮杂环庚烷-4-基)-5-氯-N 2-(1-甲基-1H-吡唑-4-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺033-e的合成
室温下将中间体033-d(700mg,1.18mmol)加入到三氟乙酸(2.0mL)的二氯甲烷溶液(5.0mL)中。反应液搅拌2小时减压浓缩后,加入LiOH(247mg,5.9mmol)到四氢呋喃/水(4.0/1.0mL)的溶液中,室温反应16小时,得到中间体033-e(350mg,收率:82%),未经纯化直接下一步使用。
LCMS:Rt:1.182min;MS m/z(ESI):361.1[M+H].
第四步:(R)-1-(4-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)氮杂环庚烷-1-基)丙-2-烯-1-酮033的合成
将中间体033-e(350mg,0.97mmol)溶解在四氢呋喃(5.0mL)和水(2.0mL)的混合液中,加入碳酸钾(514mg,2.42mmol)室温搅拌2分钟。丙烯酰氯(122mg,1.36mmol)滴加进去。搅拌2小时后,过滤制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到产物033(16.2mg,收率:4%)。
LCMS:Rt:6.587min;MS m/z(ESI):415.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.20(s,1H),8.66(s,1H),7.81(s,1H),7.45(s,1H),6.88(d,J=2.4Hz,1H),6.82-6.75(m,1H),6.18-6.11(m,1H),5.88(d,J=23.2Hz,1H),5.68(d,J=10.4Hz,1H),4.23(s,1H),3.78(s,4H),3.61(t,J=4.8Hz,2H),2.16-2.11(m,1H),1.95(s,1H),1.84(s,2H),1.81-1.70(m,1H),1.33(d,J=6.4Hz,1H),1.23(s,1H).
实施例26化合物034的制备
(R)-1-(3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000109
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000110
第一步:叔丁基(R)-3-((2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯034-b的合成
室温下将(R)-3-羟基吡咯烷-1-羧酸叔丁酯034-a(324.4mg,1.7mmol)加入到DMSO(20.0mL)溶液内。氩气保护下在20℃下搅拌10分钟,然后将NaH(68.0mg,1.7mmol)加入反应液中搅拌30分钟,把2,4-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶1b(500.0mg,1.57mmol)加入反应体系中,将温度升高到55℃反应2小时,加水(30.0mL)淬灭,用乙酸乙酯(50.0mL*2)萃取,将有机相浓缩后经过柱纯化(乙酸乙酯:石油醚=1:10)得到中间体034-b(309.0mg,收率:42%)。
LCMS:Rt:1.320min;MS m/z(ESI):413.2,415.2[M+H].
1H NMR(400M Hz,DMSO-d 6)δ7.65(s,1H),6.69(s,1H),5.79(s,2H),5.62(s,2H),3.72(s,1H),3.59(d,J=16.0Hz,5H),2.31-2.26(m,3H),1.49(m,9H),0.91(t,J=7.6Hz,2H),0.00(s,9H).
第二步:(R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯034-d的合成
室温在氮气保护下,将Pd 2(dba) 3(60.5mg,0.066mmol),BINAP(41.1mg,0.066mmol)加入到中间体034-b(309.0mg,0.66mmol),1-乙基-1H-吡唑-4-胺034-c(80.6mg,0.72mmol)和碳酸铯(645.0mg,1.98mmol)的1,4-二氧六环(20.0mL)溶液中,反应体系升温至100℃搅拌16小时,冷却至室温。加水(30.0ml)淬灭,用乙酸乙酯(50.0ml*2)萃取,将有机相浓缩后经过柱(乙酸乙酯/石油醚=2/1)纯化得到中间体034-d(296mg,收率:71.1%)。
LCMS:Rt:0.860min;MS m/z(ESI):544.3[M+H].
第三步:(R)-N-(1-乙基-1H-吡唑-4-基)-4-(吡咯烷-3-基-氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺034-e的合成
室温下将中间体034-d(296mg,0.54mmol)加入到三氟乙酸(2.0mL)的二氯甲烷溶液(4mL)中,反应液室温搅拌2小时。减压下将反应液浓缩,加入DIPEA(3.0mL)的甲醇溶液(4.0mL)中,反应液50℃搅拌反应6小时。减压下将反应液浓缩,通过反相制备(乙腈/0.1%TFA)纯化得到中间体034-e(200mg,收率:98%)。
LCMS:Rt:1.060min;MS m/z(ESI):314.2[M+H].
第四步:(R)-1-(3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-基)丙-2-烯-1-酮034的合成
将034-e(80mg,0.25mmol)溶解在丙酮(4.0mL)和H 2O(2.0mL)的混合液中,加入碳酸钾固体(96.0mg,0.62mmol)室温搅拌,将丙烯酰氯(31.0mg,0.35mmol)滴加进去。室温搅拌2小时后过滤,滤液制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至16min,10%ACN运行至19min结束纯化)得到产物034(3.14mg)。
LCMS:Rt:7.337min;MS m/z(ESI):368.1[M+H].
1H NMR(400M Hz,DMSO-d 6)δ11.28(s,1H),8.91(s,1H),7.89(s,1H),7.52(s,1H),6.91(s,1H),6.68-6.53(m,1H),6.23(d,J=1.2Hz,1H),6.20(dd,J=21.2,1.2Hz,1H),5.78-5.64(m,1H),4.09-4.01(q,J=4.8Hz,2H),3.81-3.66(m,3H),2.32-2.14(m,3H),3.36(t,J=7.2Hz,3H).
实施例27化合物035、化合物035-1和化合物035-2的制备
1-(7-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮035
(R)-1-(7-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮035-1
(S)-1-(7-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮035-2
Figure PCTCN2021073098-appb-000111
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000112
第一步:035-d的合成
在0℃氮气保护下,将中间体035-c(500mg,2.0mmol),溶于甲醇15.0mL中,加入硼氢化钠(153.8mg,4.0mmol),然后室温搅拌12h,浓缩,粗产品用乙酸乙酯和饱和食盐水洗涤,浓缩后反相制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到中间体035-d(500mg,收率:99.2%)。
LCMS:Rt:1.263min;MS m/z(ESI):248.1[M+H].
第二步:苄基7-((2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基-吡咯并[2,3-d]嘧啶-4-基)氧基)-5-氮杂 螺[2.4]庚烷-5-羧酸酯035-f的合成
在0℃气保护下,将035-d(500.0mg,2.0mmol)溶于干燥的四氢呋喃(15.0mL)中,加入叔丁醇钾(454.2mg,4.0mmol),0℃下搅拌0.5h后,2,4-二氯-7((2-(三甲基甲硅烷基)乙氧基)甲基)-吡咯并[2,3-d]嘧啶1b(705.9mg,2.00mmol)的四氢呋喃(3.0mL)溶液缓慢滴加,然后室温搅拌2h。加入饱和氯化铵溶液淬灭,60.0mL乙酸乙酯洗涤,滤液在减压下浓缩,所得粗产品用柱层色谱纯化(乙酸乙酯:石油醚=30:1至15:1),得到035-f(860mg,收率:80.4%)。
LCMS:Rt:2.327min;MS m/z(ESI):529.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ7.66(d,J=3.5Hz,1H),7.46(d,J=4.3Hz,2H),7.43-7.37(m,3H),6.71(t,J=3.1Hz,1H),5.62(s,2H),5.32(dd,J=10.2,5.7Hz,1H),5.22-5.10(m,2H),4.01(ddd,J=21.9,12.8,4.2Hz,1H),3.91-3.76(m,2H),3.58(t,J=8.1Hz,2H),3.31(dd,J=15.1,10.5Hz,1H),1.09-0.97(m,2H),0.96-0.78(m,4H),0.00(d,J=4.0Hz,9H).
第三步:苄基7-((2-((1-乙基-1H-吡唑-4-基)氨基-7-((2-(三甲基甲硅烷基)乙氧基)甲基-吡咯并[2,3-d]嘧啶-4-基)氧基)-5-氮杂螺[2.4]庚烷-5-羧酸酯035-h的合成
在室温氮气保护下,将035-f(860mg,1.6mmol)和035-g(269.0mg,2.4mmol)溶于1,4-二氧六环(20.0mL),然后在搅拌下,加入Pd(dppf)Cl 2(149.1mg,0.16mmol),BINAP(203.0mg,0.32mmol)和碳酸铯(1.1g,3.2mmol),反应体系升温至120℃,搅拌12小时后,冷却至室温,过滤,滤饼用乙酸乙酯(60.0mL)洗涤,滤液减压浓缩所得粗品纯化(石油醚:乙酸乙酯=10:1至2:1),得到中间体035-h(700mg,收率:71.3%)。
LCMS:Rt:1.987min;MS m/z(ESI):604.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ9.20(d,J=6.9Hz,1H),8.09(s,1H),7.64(s,1H),7.54-7.40(m,5H),7.19(t,J=17.1Hz,1H),6.48(s,1H),5.62(s,2H),5.34(d,J=3.5Hz,1H),5.28-5.15(m,2H),4.28-4.17(m,2H),4.10-3.97(m,1H),3.91(dt,J=25.6,23.4Hz,3H),3.64(t,J=7.9Hz,2H),3.35(dd,J=14.8,10.6Hz,1H),1.49(dd,J=11.5,7.1Hz,3H),1.11(s,1H),1.03-0.83(m,5H),0.00(d,J=3.8Hz,9H).
第四步:4-((5-氮杂螺[2.4]庚烷-7-基)氨基)-N-((1-乙基-1H-吡唑-4-基)氨基)-7((2-(三甲基甲硅烷基)乙氧基)甲基-7H吡咯并[2,3-d]嘧啶-2-氨035-i的合成
在室温下,将035-h(700mg,1.2mmol)溶于异丙醇(10.0mL)中,加入无水Pd/C(70.0mg),在氢气氛围下(4PSI)室温搅拌48h。过滤浓缩,粗品经反相制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到035-i(400.0mg,收率:73.5%)。
LCMS:Rt:1.247min;MS m/z(ESI):470.2[M+H].
第五步:4-((5-氮杂螺[2.4]庚烷-7-基)氧基)-N-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-2-胺035-j的合成
在0℃下,将三氟乙酸(3.0g,3.05mmol)加入到035-i(400.0mg,0.85mmol)二氯甲烷溶液(10.0mL)中,室温搅拌24小时。LCMS监测反应结束后,减压下将反应液浓缩,粗产品溶于甲醇/N,N-二异丙基乙胺的混合液(10.0mL,1:1)的混合液中,50℃下搅拌16h,得到产品035-j(190.0mg,收率:65.7%)。
LCMS:Rt:0.847min;MS m/z(ESI):340.2[M+H].
第六步:1-(7-((-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-5-氮杂螺[2.4]庚烷-5-基)丙-2-烯-1-酮035的合成
在0℃下,将035-j(190.0mg,0.56mmol)加入到四氢呋喃(4.0mL)和H 2O(2.0mL)的混合液中,再加入磷酸钾(594.9mg,2.8mmol),搅拌10分钟后,缓缓滴加丙烯酰氯的四氢呋喃溶液(76.1mg,0.84mmol),室温搅拌2小时,过滤,滤液用Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化,得到化合物035,化合物035经SFC拆分[column:DAICEL CHIRALCEL OD(250mm*30mm,10um);mobile phase:A:CO 2;B:MeOH(0.1%DEA),30%-30%,20min]得到035-1(23.55mg,收率:22%)和035-2(21.68mg,收率:20%)。
035-1:
LCMS:Rt:5.787min;MS m/z(ESI):394.2[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.27(s,1H),8.84(s,1H),7.87(s,1H),7.50(s,1H),6.92(s,1H),6.57(dd,J=28.5,11.3Hz,1H),6.25(s,1H),6.16(d,J=17.0Hz,1H),5.77-5.58(m,1H),5.28(d,J=45.7Hz,1H),4.20-3.97(m,3H),3.97-3.73(m,2H),3.46(d,J=10.2Hz,1H),1.37(t,J=7.2Hz,3H),1.06-0.83(m,2H),0.75(d,J=14.9Hz,2H).
035-2:
1H NMR(400MHz,DMSO-d 6)δ11.28(s,1H),8.85(s,1H),7.87(s,1H),7.50(s,1H),6.93(dd,J=10.8,7.7Hz,1H),6.57(ddd,J=16.7,12.9,10.2Hz,1H),6.27-6.22(m,1H),6.20-6.10(m,1H),5.73-5.63(m,1H),5.29(dd,J=46.2,4.1Hz,1H),4.19-3.91(m,3H),3.89-3.74(m,2H),3.40(dd,J=48.2,11.1Hz,1H),1.37(t,J=7.3Hz,4H),1.05-0.83(m,2H),0.82-0.68(m,2H).
实施例28化合物036的制备
1-((2S,4R)-4-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000113
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000114
第一步:(2S,4R)-4-((2,5-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-羧酸叔丁酯036-c的合成
将2,4,5-三氯-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶011-1-b(20mg,0.06mmol),化合物036-b(12mg,0.06mmol),叔丁醇钾(10mg,0.09mmol)加入到四氢呋喃(0.55mL)中。反应液在25℃下搅拌30分钟。将反应液减压浓缩后纯化(石油醚:乙酸乙酯=10:1)得到中间体036-c(15mg,收率:48%)。
LCMS:Rt:1.62min;MS m/z(ESI):461.2,462.2[M+H].
1H NMR(400MHz,CDCl 3)δ7.14(s,1H),5.83(brs,1H),5.54(s,2H),4.15-4.09(m,1H),3.86-3.75(m,2H),3.55(t,J=8.0Hz,2H),2.47(brs,1H),2.07-2.05(m,1H),1.51-1.46(m,12H),0.95(t,J=8.4Hz,2H),0.00(s,9H).
第二步:(2S,4R)-4-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧 基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-羧酸叔丁酯036-e的合成
将036-c(220mg,0.43mmol),1-甲基-1H-吡唑-4-胺036-d(84mg,0.86mmol),Pd(dppf)Cl 2(31mg,0.043mmol),Xantphos(25mg,0.043mmol)和碳酸铯(280mg,0.86mmol)加入到1,4-二氧六环(10.0mL)中,反应液在氩气保护下95℃搅拌反应16小时。将反应液用硅藻土过滤,并用乙酸乙酯(15.0mL)洗涤滤饼。将滤液在减压下浓缩,纯化(石油醚/乙酸乙酯=1:1)得到036-e(250mg,收率:99%)。LCMS:Rt:1.26min;MS m/z(ESI):578.3[M+H]. 1H NMR(400MHz,CDCl 3)δ7.83(s,1H),7.61(s,1H),7.03(s,1H),6.87(s,1H),6.69(s,1H),5.70(m,1H),5.50(s,2H),4.17-4.15(m,1H),3.96(s,3H),3.82-3.68(m,2H),3.59(t,J=8.0Hz,1H),2.46(m,1H),2.11(m,1H),2.09(m,1H),1.54(m,11H),0.98(t,J=4.0Hz,2H),0.00(s,9H).
第三步:5-氯-N-(1-甲基-1H-吡唑-4-基)-4-((((3R,5S)-5-甲基吡咯烷-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺036-f的合成
25℃下将036-e(250mg,0.43mmol)加入到三氟乙酸(3.0mL)中,反应液25℃下搅拌30分钟。减压浓缩后,加入氢氧化锂(361mg,8.60mmol)的四氢呋喃(2.0mL)/水(1.0mL)溶液中,反应液在25℃搅拌3小时。减压下将反应液浓缩,纯化(二氯甲烷/甲醇=10:1)得到036-f(200mg,收率:99%)。
LCMS:Rt:3.91min;MS m/z(ESI):348.2[M+H].
第四步:1-((2S,4R)-4-((5-氯-2-((1-甲基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮036的合成
将036-f(150mg,0.43mmol)溶解在四氢呋喃(2.5mL)和水(0.5mL)的混合液中,加入磷酸钾(182mg,0.86mmol)搅拌。将丙烯酰氯(39mg,0.43mmol)的四氢呋喃(0.5mL)溶液逐滴滴加进去,25℃下搅拌1小时。将反应液减压浓缩后经Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行4min,30%-35%ACN运行至13min,95%ACN运行至16min,10%ACN运行至19min结束纯化得到036(22.3mg,收率:9%)。
LCMS:Rt:5.38min;MS m/z(ESI):402.3[M+H].
1H NMR(400M Hz,DMSO-d 6)δ11.53(s,1H),9.10(s,1H),7.85(s,1H),7.51(s,1H),7.06(s,1H),6.70-6.53(m,1H),6.16(t,J=18.8Hz,1H),5.81-5.64(m,2H),4.40-4.27(m,1H),4.06-3.89(m,1H),3.80(s,3H),3.69-3.66(m,1H),2.11-1.94(m,1H),1.43-1.39(m,3H),1.26-1.24(m,1H).
实施例29化合物037的制备
1-((2S,4R)-4-((5-氯-2-((1-(二氟甲基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000115
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000116
第一步:1-(二氟甲基)-4-硝基-1H-吡唑037-i的合成
在0℃下,将溴二氟甲烷037-h(3.93g,30.0mmol)滴入到4-硝基-1H-吡唑037-g(1.13mg,10.0mmol)和Cs 2CO 3(6.52g,20.0mmol)的DMF(10.0mL)溶液中,加完后升温到室温,将20.0mL饱和NH 4Cl溶液加入反应液淬灭,用乙酸乙酯萃取两次(50.0mL*2),有机相合并,用水(20.0mL)和饱和氯化钠溶液(20.0mL)各洗一次,有机相用无水硫酸钠干燥过滤,减压浓缩后纯化(石油醚:乙酸乙酯=5:1)得到中间体037-i(922mg,收率:56.6%)。
1H NMR(400M Hz,CDCl 3)δ8.51(s,1H),8.21(s,1H),7.21(t,J=60.0Hz,1H).
第二步:1-(二氟甲基)-1H-吡唑-4-胺037-d的合成
室温下,将湿Pd/C(10%,46mg)加入037-i(459mg,2.82mmol)的乙醇(10.0mL)溶液中,加完后用水泵抽真空,用氢气填充,重复5次,反应体系搅拌过夜,过滤,滤饼用四氢呋喃(10.0mL*3)洗涤,滤液浓缩得到中间体037-d(309mg,收率:82.3%)。
LCMS:Rt:0.339min;MS m/z(ESI):134.1[M+H].
第三步:(2S,4R)-4-((2,5-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-羧酸叔丁酯037-c的合成
将011-1-b(438mg,1.24mmol),化合物037-b(250mg,1.24mmol),叔丁醇钾(209mg,1.86mmol)加入到四氢呋喃(10.0mL)中,该反应液在25℃下搅拌30分钟。将反应液用水(30.0mL)稀释,然后用乙酸乙酯(10.0mL*2)萃取。将有机相减压浓缩后纯化(石油醚:乙酸乙酯=5:1)得到中间体037-c(530mg,收率:83%)。
LCMS:Rt:1.65min;MS m/z(ESI):461.2[M+H-56].
第四步:(2S,4R)-叔丁基4-((5-氯-2-((1-(二氟甲基)-1H-吡唑-4-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-甲酸酯037-e的合成
将中间体037-c(250mg,0.48mmol),1-(二氟甲基)-1H-吡唑-4-胺037-d(96mg,0.72mmol),Pd(dppf)Cl 2(35mg,0.048mmol),Xantphos(28mg,0.048mmol)和碳酸铯(313mg,0.96mmol)加入到1,4-二氧六环(10.0mL)中,反应液在氩气保护下100℃搅拌反应16小时。将反应液用硅藻土过滤,并用乙酸乙酯(20.0mL)洗涤滤饼。将滤液在减压下浓缩,纯化(石油醚:乙酸乙酯=3:1)得到中间体037-e(230mg,收率:77%)。
LCMS:Rt:1.52min;MS m/z(ESI):614.3[M+H].
1H NMR(400M Hz,CDCl 3)δ8.37(s,1H),7.79(s,1H),6.94(s,1H),6.85(s,1H),5.72(brs,1H),5.53(s,2H),4.17-3.78(m,3H),3.62(t,J=8.0Hz,2H),2.48(m,1H),2.16(m,1H),1.55(m,11H),1.33(t,J=7.6Hz, 3H),0.99(t,J=8.0Hz,2H),0.00(s,9H).
第五步:5-氯-N-(1-(二氟甲基)-1H-吡唑-4-基)-4-(((((3R,5S)-5-甲基吡咯烷-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺037-f的合成
25℃下将中间体037-e(230mg,0.37mmol)加入到三氟乙酸(3.0mL)中,反应液25℃下搅拌30分钟。反应液减压浓缩后,加入氢氧化锂(157mg,3.7mmol的四氢呋喃(3.0mL)/水(1.0mL)溶液中,反应液在25℃搅拌1小时。减压浓缩,纯化(二氯甲烷:甲醇=10:1)得到中间体037-f(90mg,收率:63%)。
LCMS:Rt:4.33min;MS m/z(ESI):384.2,386.2[M+H].
第六步:1-((2S,4R)-4-((5-氯-2-((1-(二氟甲基)-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮037的合成
将中间体037-f(900mg,0.23mmol)溶解在四氢呋喃(2.5mL)和水(0.5mL)的混合液中,加入磷酸钾固体(98mg,0.46mmol)后在25℃下搅拌,将丙烯酰氯(21mg,0.23mmol)的四氢呋喃(0.5mL)溶液逐滴滴加进去,25℃下搅拌1小时。反应液经制备Waters 2767制备色谱仪,流动相A:1‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至11min,95%ACN运行至14min,10%ACN运行至19min结束纯化得到037(38.63mg,收率38%)。
LCMS:Rt:6.25min;MS m/z(ESI):438.2,440.2[M+H].
1H NMR(400M Hz,DMSO-d 6)δ11.69(s,1H),9.46(s,1H),8.36(s,1H),7.93-7.64(m,2H),7.14(s,1H),6.70-6.53(m,1H),6.22-6.13(m,1H),5.82-5.72(m,1H),5.71-5.65(m,1H),4.40-4.30(m,1H),4.06-3.68(m,2H),2.60-2.43(m,1H),2.13-1.96(m,1H),1.42(t,J=6.8Hz,3H).
实施例30化合物038的制备
1-((2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-基)-丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000117
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000118
第一步:(2S,4R)-4-((2-氯-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-甲酸叔丁酯038-b的合成
在0℃下,将NaH(37.7mg,60%,0.942mmol)加入到(2S,4R)-4-羟基-2-甲基四氢吡咯-1-甲酸叔丁酯038-a(126.5mg,0.628mmol)的DMSO溶液(5.0mL)中,室温搅拌30分钟后,将2,4-二氯-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶1b(200mg,0.628mmol)加入到反应液中搅拌反应2小 时。反应液倒入到冰水(10.0mL)中,反应液减压浓缩,残留物过Waters 2767制备色谱仪,流动相A:1‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至19min结束纯化,得到中间体038-b(125mg,收率:41.2%)。
LCMS:Rt:1.520min;MS m/z(ESI):483.2[M-H].
第二步:(2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-甲酸叔丁酯038-d的合成
在氮气保护下,将Pd 2(dba) 3(9.5mg,0.010mmol)和BINAP(6.5mg,0.010mmol)加入到038-b(100mg,0.207mmol),1-乙基4-氨基吡唑038-c(23.0mg,0.207mmol)和碳酸铯(168.6mg,0.518mmol)的1,4-二氧六环(5.0mL)溶液中。反应体系升温至100℃搅拌反应16小时。反应液冷却至室温,减压浓缩,经Waters 2767制备色谱仪,流动相A:1‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至16min,10%ACN运行至20min结束纯化中间体038-d(115mg,收率:99.5%)。
LCMS:Rt:1.895min;MS m/z(ESI):558.7[M+H]
第三步:N-(1-乙基-1H-吡唑-4-基)-4-(((3R,5S)-5-甲基四氢吡咯-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺038-e的合成
在0℃下,将三氟乙酸(1.0mL)加入到中间体038-d(115mg,0.206mmol)的二氯甲烷溶液(5.0mL)中,搅拌16小时。减压浓缩,残留物中加入到四氢呋喃(2.0mL)和水(3.0mL),冷却至0℃,再加入氢氧化锂(24.7mg),在25℃下搅拌反应1小时。反应液减压浓缩,残留物经Waters 2767制备色谱仪,流动相A:1‰TFA水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至14min,10%ACN运行至20min结束纯化得到中间体038-e(65mg,收率:96.5%)。
LCMS:Rt:0.807min;MS m/z(ESI):328.2[M+H].
第四步:1-((2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-基)-丙-2-烯-1-酮038的合成
在0℃下,将中间体038-e(60mg,0.183mmol)溶解在四氢呋喃(5.0mL)和水(5.0mL)中,加入磷酸钾(97.1mg,0.458mmol)搅拌。丙烯酰氯(19.9mg,0.219mmol)缓慢加入,室温搅拌反应2小时。反应液减压浓缩,经Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至16min,10%ACN运行至19min结束纯化得到038(28.1mg,收率:40.2%)。
LCMS:Rt:5.875min;MS m/z(ESI):382.1[M+H].
1H NMR(400M Hz,DMSO-d 6)δ11.29(brs,1H),8.92(s,1H),7.89(s,1H),7.52(s,1H),6.92(s,1H),6.70-6.53(m,1H),6.23-6.22(m,1H),6.16(t,J=14.8Hz,1H),5.78-5.71(m,1H),5.71-5.64(m,1H),4.40-4.22(m,2H),4.08(q,J=7.2Hz,2H),3.95-3.86(m,1H),3.66(d,J=14.8Hz,1H),2.02(dd,J=7.2,14.4Hz,1H),1.38-1.33(m,6H).
实施例31化合物039的制备
(R,E)-2-(3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羰基)-4,4-二甲基戊-2-烯腈
Figure PCTCN2021073098-appb-000119
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000120
第一步:(R)-叔丁基3-((2-氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯039-b的合成
0℃下,将(R)-3-羟基吡咯烷-1-羧酸叔丁酯039-a(971mg,5.20mmol)溶于四氢呋喃(50.0mL),加入氢化钠(245mg,60%w/w)。搅拌30分钟后加入2,4-二氯-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶1b(1.50g,4.7mmol),0℃下搅拌2小时。加入饱和氯化铵水(20.0mL)溶液淬灭反应,使用乙酸乙酯(15.0mL*3)进行萃取,有机相用饱和氯化钠水溶液(20.0mL)洗涤,干燥过滤浓缩,残留物纯化(乙酸乙酯/石油醚=1/4),得到中间体039-b(2.00g,90.5%)。
LCMS:Rt:1.068min;MS m/z(ESI):413.1[M-56+H].
1H NMR(400MHz,CHLOROFORM-d)δ=7.15(d,J=3.5Hz,1H),6.54(d,J=3.8Hz,1H),5.86-5.75(m,1H),5.55(s,2H),3.79-3.44(m,6H),2.27-2.17(m,2H),1.46(br s,9H),0.96-0.83(t,J=8.0Hz,2H),-0.05(s,9H).
第二步:(R)-叔丁基3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羧酸酯039-d的合成
将中间体039-b(1.00g,2.10mmol)溶于1,4-二氧六环(20.0mL)中,加入1-乙基-1H-吡唑-4-胺039-c(261mg,2.40mmol),三二亚苄基丙酮二钯(195mg,213μmol),碳酸铯(1.40g,4.30mmol)和联萘二苯磷(133mg,213μmol)。氮气氛围下100℃反应16小时。降温后加入水(50.0mL)稀释反应,使用乙酸乙酯(25.0mL*3)进行萃取,有机相用饱和氯化钠水溶液(25.0mL)洗涤。有机相溶液过滤浓缩,残留物纯化(乙酸乙酯/石油醚=1/4),得到中间体039-d(600mg,51.8%)。
LCMS:Rt:1.073min;MS m/z(ESI):544.3[M+H].
1H NMR(400MHz,CHLOROFORM-d)δ=7.93-7.80(m,1H),7.59(s,1H),6.88(d,J=3.4Hz,1H),6.62(br d,J=6.6Hz,1H),6.42(d,J=3.5Hz,1H),5.78-5.64(m,1H),5.50(s,2H),4.25-4.12(m,2H),3.81-3.47(m,6H),2.32-2.13(m,2H),1.56-1.43(m,12H),0.97-0.87(t,J=8.0Hz,2H),-0.06(s,9H).
第三步:(R)-N-(1-乙基-1H-吡唑-4-基)-4-(吡咯烷-3-氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺039-e的合成
将中间体039-d(600mg,1.10mmol)溶于二氯甲烷(10.0mL)中,加入三氟乙酸(1.90g,16.2mmol),25℃搅拌反应16小时。反应液浓缩后,溶于乙醇(20.0mL)和水(10.0mL)中,加入氢氧化钠(233mg,5.80mmol),50℃搅拌反应2小时。反应液降温后浓缩,残留物制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化)得到中间体039-e(300mg,82.2%)。
LCMS:Rt:0.667min;MS m/z(ESI):314.1[M+H].
第四步:(R,E)-2-(3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)吡咯烷-1-羰基)-4,4-二甲基戊-2-烯腈039的合成
将中间体039-e(150mg,479μmol)溶于DMF(5.0mL)中,加入三乙胺(145mg,1.40mmol)和2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(182mg,479μmol),50℃搅拌2小时。降温后加入水稀释,使用乙酸乙酯(10.0mL)进行萃取,有机相用饱和氯化钠水溶液(20.0mL)洗涤,干燥过滤浓缩,残留物制备纯化(经Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至14min,95%ACN运行至16min,10%ACN运行至19min结束纯化),得到产物039(15.0mg,7.0%)。
LCMS:Rt:2.720min;MS m/z(ESI):449.3[M+H].
1H NMR(400MHz,DMSO-d6)δ=11.30(s,1H),8.93(s,1H),7.90(d,J=10.3Hz,1H),7.52(d,J=2.6Hz,1H),7.07(s,1H),7.03-6.98(m,1H),7.00(s,1H),7.03-6.98(m,1H),6.94(s,1H),6.27-6.22(m,1H),5.74(s,1H),4.14-4.07(q,J=7.4Hz,2H),3.91(dd,J=5.0,13.8Hz,1H),3.83-3.69(m,2H),3.68-3.52(m,2H),2.37-2.19(m,2H),1.37(t,J=7.2Hz,3H),1.27-1.13(m,9H).
实施例32化合物040的制备
4-(((3R,5S)-1-丙烯酰基-5-甲基吡咯烷-3-基)氧基)-2-((1-甲基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-5-腈
Figure PCTCN2021073098-appb-000121
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000122
第一步:5-溴-2,4-二氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶040-b的合成
将040-a(100mg,0.37mmol),NaH(18mg,0.44mmol)在氩气保护下的无水四氢呋喃(3.0mL)中0℃下搅拌1小时。2-(三甲基硅烷基)乙氧甲基氯(74mg,0.44mmol)随后缓慢加入加入。反应液在25℃下搅拌1小时。将反应液用水(15.0mL)淬灭,然后用乙酸乙酯(5.0mL*3)萃取。有机相减压浓缩后得到中间体040-b(150mg,收率:99%)。
LCMS:Rt:1.15min;MS m/z(ESI):398.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ8.18(s,1H),5.57(s,2H),3.54(t,J=8.0Hz,2H),0.85(t,J=8.0Hz,2H), 0.00(s,9H).
第二步:(2S,4R)-叔丁基4-((5-溴-2-氯-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-羧酸酯040-d的合成
将040-b(150mg,0.38mmol),040-c(105mg,0.42mmol),叔丁醇钾(64mg,0.57mmol),加入到四氢呋喃(3.0mL)中,反应液在25℃搅拌反应30分钟。反应液减压浓缩,纯化(石油醚/乙酸乙酯=10:1)得到中间体040-d(100mg,收率:47%)。
LCMS:Rt:1.72min;MS m/z(ESI):507.1[M+H-56].
1H NMR(400M Hz,DMSO-d 6)δ7.89(s,1H),5.78(s,1H),5.58(s,2H),4.05-3.87(m,2H),3.61-3.58(m,3H),2.03-1.97(m,1H),1.48-1.43(m,12H),1.30(s,1H),0.91(t,J=8.0Hz,2H),0.00(s,9H).
第三步:(2S,4R)-叔丁基4-((5-溴-2-((1-甲基-1H-吡唑-4-基)氨基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-羧酸酯040-f的合成
将040-d(100mg,0.18mmol),Pd(dppf)Cl 2(13mg,0.018mmol),Xantphos(10mg,0.018mmol),碳酸铯(117mg,0.36mmol),040-e(35mg,0.36mmol)加入到1,4-二氧六环(10.0mL)中,反应液在氩气保护下100℃搅拌16小时。反应液通过硅藻土过滤,用乙酸乙酯(15.0mL)洗涤。滤液浓缩纯化(石油醚/乙酸乙酯=3:1)得到中间体040-f(30mg,收率:27%)。
LCMS:Rt:0.96min;MS m/z(ESI):624.3[M+H].
1H NMR(400M Hz,DMSO-d 6)δ9.44(s,1H),8.35(m,1H),7.65(s,1H),7.43(s,1H),5.80-5.79(m,1H),5.61-5.56(m,2H),4.16-4.14(m,2H),4.08-4.05(m,3H),3.67-3.63(m,3H),2.09-2.04(m,1H),1.53-1.42(m,12H),1.32(s,1H),0.91(t,J=8.0Hz,2H),0.00(s,9H).
第四步:(2S,4R)-叔丁基4-((5-氰基-2-((1-甲基-1H-吡唑-4-基)氨基)-7-(((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-羧酸酯040-g的合成
将040-f(150mg,0.24mmol),Pd 2(dba) 3(88mg,0.096mmol),dppf(53mg,0.096mmol),氰化亚铜(215mg,2.4mmol)加入到1,4-二氧六环(10.0mL)中,反应液在氩气保护下100℃搅拌16小时。反应液通过硅藻土过滤,用乙酸乙酯(15.0mL)洗涤。滤液减压浓缩纯化(石油醚/乙酸乙酯=2:1)得到040-g(120mg,收率:88%)。
LCMS:Rt:1.52min;MS m/z(ESI):569.3[M+H].
第五步:2-((1-甲基-1H-吡唑-4-基)氨基)-4-((((3R,5S)-5-甲基吡咯烷-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-5-腈040-h的合成
将040-g(100mg,0.18mmol)加入到三氟乙酸(3.0mL)中,反应液25℃下搅拌30分钟。减压下将反应液浓缩,加入到一水合氢氧化锂(151mg,3.6mmol)的四氢呋喃(5.0mL)/水(1.0mL)溶液中,反应液在25℃搅拌1小时后,减压浓缩,通过Prep-HPLC(1‰TFA)纯化得到040-h(65mg,收率:98%)。
LCMS:Rt:3.68min;MS m/z(ESI):339.3[M+H].
第六步:4-(((3R,5S)-1-丙烯酰基-5-甲基吡咯烷-3-基)氧基)-2-((1-甲基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-5-腈040的合成
将040-h(65mg,0.19mmol)溶解在四氢呋喃(1.5mL)和水(三滴)的混合液中,加入磷酸钾固体(60mg,0.29mmol)后在25℃下搅拌,将丙烯酰氯(17mg,0.19mmol)的四氢呋喃(0.5mL)溶液逐滴滴加进去,25℃下搅拌1小时。将反应液减压浓缩后经Waters 2767制备色谱仪,流动相A:1‰氨水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化得到040(2.01mg,收率3%)。
LCMS:Rt:4.78min;MS m/z(ESI):393.3[M+H].
1H NMR(400M Hz,DMSO-d 6)δ12.33(s,1H),9.29(s,1H),7.93(s,1H),7.86(s,1H),7.53(s,1H),6.71-6.53(m,1H),6.17(t,J=16.0Hz,1H),5.83-5.78(m,1H),5.72-5.65(m,1H),4.41-4.27(m,1H),4.09-3.90(m,2H),3.81(s,3H),3.69-3.66(m,1H),2.11-1.94(m,1H),1.42-1.38(m,3H).
实施例33化合物041的制备
(R)-1-(3-((2-((1-乙基-1H-吡唑-4-基)氨基)-5-氟-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷 -1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000123
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000124
第一步:(R)-3-((2-氯-5-氟-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸叔丁酯041-c的合成
将2,4-二氯-5-氟-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶041-a(500mg,1.50mmol),(R)-3-氨基吡咯烷-1-羧酸叔丁酯041-b(560mg,3.00mmol)和DIPEA(580mg,4.50mmol),加入到异丙醇(20.0mL)溶液内,90℃下搅拌16小时。有机相减压浓缩后纯化(乙酸乙酯:石油醚=5:1)得到中间体041-c(660mg,收率:91%)。
LCMS:Rt:2.235min;MS m/z(ESI):486.2[M+H].
第二步:(R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-5-氟-7-((2-(三甲基硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)吡咯烷-1-羧酸叔丁酯041-e的合成
室温在氮气保护下,将Pd 2(dba) 3(128mg,0.14mmol),BINAP(87mg,0.14mmol)加入到041-c(660mg,1.40mmol),1-乙基-1H-吡唑-4-胺(041-d,311mg,2.80mmol)和碳酸铯(912mg,2.80mmol)的1,4-二氧六环(40.0mL)溶液中,反应体系升温至100℃搅拌12小时。冷却至室温后过滤,反应液减压浓缩纯化(乙酸乙酯:石油醚=2:1)得到中间体041-e(630mg,收率:82.6%)。
LCMS:Rt:2.090min;MS m/z(ESI):561.3[M+H].
第三步:N 2-(1-乙基-1H-吡唑-4-基)-N 4-((3R,5S)-吡咯烷-3-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺041-f的合成
室温下将041-e(580mg,1.0mmol)加入到三氟乙酸(4.0mL)的二氯甲烷溶液(8.0mL)中,反应液在0℃搅拌6小时。反应液用饱和碳酸氢钠(50ml)淬灭,用乙酸乙酯萃取,将有机相在减压下浓缩,粗品未经纯化加入到LiOH(205mg,5.0mmol)的四氢呋喃/水溶液(10.0mL/2.0mL)中,反应液搅拌2小时。反应液减压液浓缩得到中间体041-f(340mg,收率:99%)。
LCMS:Rt:1.03min;MS m/z(ESI):327.3[M+H].
第四步:1-((2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-吡咯烷-1-基)丙-2-烯-1-酮041的合成
将041-f(340mg,1.0mmol)溶解在四氢呋喃(10.0mL)和水(2.0mL)的混合液中,加入磷酸钾(424mg,2.0mmol)搅拌,将丙烯酰氯(110mg,1.2mmol)的四氢呋喃(1.0mL)溶液逐滴滴加进去,0℃下搅拌1小时。将反应液减压浓缩后经Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至13min,95%ACN运行至15min,10%ACN运行至20min结束纯化得到041(55.8mg,收率:14.1%)。
LCMS:Rt:3.567min;MS m/z(ESI):381.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ11.86(s,1H),10.15(s,1H),9.10(s,1H),7.95(s,1H),7.58(s,1H),6.96(s, 1H),6.75(s,1H),6.71-6.54(m,1H),6.22-6.13(m,1H),5.74-5.55(m,2H),4.69-4.61(m,1H),4.13-4.07(m,3H),3.59-3.47(m,1H),2.67-2.57(m,1H),1.98-1.77(m,1H),1.35(t,J=7.2Hz,3H),1.29(d,J=6.4Hz,3H).
实施例34化合物042的制备
1-((2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-5-氟-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000125
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000126
第一步:(2S,4R)-4-((2-氯-5-氟-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-羧酸叔丁酯042-c的合成
将2,4-二氯-5-氟-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶042-a(300mg,0.98mmol),(2S,4R)-4-氨基-2-甲基吡咯烷-1-羧酸叔丁酯042-b(197mg,0.98mmol)加入到DIPEA(287mg,2.25mmol)的异丙醇(10.0mL)溶液内,在110℃下搅拌16小时。加水(100.0mL)进去,用乙酸乙酯(100.0mL*2)萃取,合并有机相,用饱和食盐水洗,将有机相在减压下浓缩,过硅胶柱(乙酸乙酯:石油醚=1:10)得到042-c(380mg,收率:85.0%)。
LCMS:Rt:1.989min;MS m/z(ESI):500.1[M+H].
第二步:(2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-5-氟-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-羧酸叔丁酯042-e的合成
室温在氮气保护下,将Pd 2(dba) 3(64mg,0.07mmol),BINAP(43mg,0.07mmol)加入到042-c(350g,0.7mmol),1-乙基-1H-吡唑-4-胺042-d(109mg,0.9mmol)和碳酸铯(570,mg,1.75mmol)的1,4-二氧六环(20.0mL)溶液中,反应升温至120℃搅拌12小时,。反应冷却至室温,过滤纯化(乙酸乙酯:石油醚=2:1)得到中间体042-e(341mg,收率:84%)。
LCMS:Rt:2.037min;MS m/z(ESI):575.2[M+H].
第三步:N 2-(1-乙基-1H-吡唑-4-基)-5-氟-N 4-((3R,5S)-5-甲基吡咯烷-3-基)-7H-吡咯并[2,3-d]嘧啶-2,4-二胺042-f的合成
室温下将042-e(340mg,0.59mmol)加入到三氟乙酸(3.0mL)的二氯甲烷溶液(6.0mL)中,反应液在0℃搅拌6小时。反应液用饱和碳酸氢钠(50.0ml)淬灭,乙酸乙酯(20.0mL*2),将有机相在减压下浓缩,粗品未经纯化加入到LiOH(242mg,5.9mmol)的四氢呋喃/水溶液(4.0mL/1.5mL)中,反应液25℃下搅拌2小时。反应液减压浓缩,过Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至12min,95%ACN运行至15min,10%ACN运行至18min结束纯化得到中间体042-f(55mg,收率: 27%)。
LCMS:Rt:1.10min;MS m/z(ESI):345.3[M+H].
第四步:1-((2S,4R)-4-((2-((1-乙基-1H-吡唑-4-基)氨基)-5-氟-7H-吡咯并[2,3-d]嘧啶-4-基)氨基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮042的合成
将042-f(50mg,0.15mmol)溶解在四氢呋喃(6.0mL)和水(2.0mL)的混合液中,加入磷酸钾(77mg,0.36mmol)搅拌,将丙烯酰氯(17mg,0.18mmol)的四氢呋喃(0.5mL)溶液逐滴滴加进去,25℃下搅拌1小时。反应液减压浓缩后经制备纯化(Waters 2767制备色谱仪,流动相A:1‰甲酸水溶液,流动相B:乙腈,色谱柱:Sunfire(Prep C18 OBD 19*250mm 10um),梯度:10%ACN运行3min,30%-35%ACN运行至10min,95%ACN运行至13min,10%ACN运行至20min结束纯化)得到产物042(4.1mg,收率:6.8%)。
LCMS:Rt:6.557min;MS m/z(ESI):399.1[M+H].
1H NMR(400MHz,DMSO-d 6)δ10.68(s,1H),8.62(s,1H),7.84(s,1H),7.45(s,1H),6.64-6.53(m,3H),6.15(t,J=14.0Hz,1H),5.69-5.62(m,1H),4.65(d,J=8.8Hz,1H),4.10-4.01(m,4H),3.46(t,J=10.0Hz,1H),2.43(s,1H),1.95-1.83(m,1H),1.34-1.28(m,6H).
实施例35化合物017-1的制备
1-((2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-基)-丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000127
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000128
第一步:叔丁基(2S,4R)-4-((2,5-二氯-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-羧酸酯017-b的合成
在0℃下,将叔丁醇钾(143.1mg,1.275mmol)加入到叔丁基(2S,4R)-4-羟基-2-甲基四氢吡咯-1-羧酸酯017-a(171.2mg,0.850mmol)的四氢呋喃溶液(10mL)中,室温搅拌30分钟。然后将2,4,5-三氯-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶011-1-b(300mg,0.850mmol)加入到反应液中,25℃下搅拌反应1小时。将反应液倒入到饱和氯化铵溶液(10mL)中,用乙酸乙酯萃取(10mL*3),合并萃取液,用无水硫酸钠干燥,过滤,滤液减压蒸干,残留物过硅胶柱(PE/EtOAc=20:1),得到叔丁基(2S,4R)-4-((2,5-二氯-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-羧酸酯017-b(363mg)。
LCMS:Rt:1.692min;MS m/z(ESI):516.2,518.2[M-H].
第二步:叔丁基(2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-羧酸酯017-d的合成
在氮气保护下,将Pd 2(dba) 3(21.6mg,0.024mmol)和BINAP(14.7mg,0.024mmol)加入到叔丁基(2S,4R)-4-((2,5-二氯-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-羧酸酯017-b(245mg,0.473mmol),1-乙基4-氨基吡唑017-c(52.6mg,0.473mmol)和碳酸铯(385.3mg,1.182mmol)的1,4-二氧六环(5mL)溶液中,反应体系升温至100℃搅拌反应16小时。反应液冷却至室温,在减压下过滤,浓缩,残留物过反相Flash(乙腈/0.05%TFA)纯化得到叔丁基(2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-羧酸酯017-d(238mg)。
LCMS:Rt:1.467min;MS m/z(ESI):592.3,594.3[M+H].
第三步:5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(((3R,5S)-5-甲基四氢吡咯-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺017-e的合成
在0℃下,将三氟乙酸(2mL)加入到叔丁基(2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-羧酸酯017-d(238mg,0.402mmol)的二氯甲烷溶液(5mL)中,室温下搅拌反应16小时。减压下将反应液浓缩至干,残留物中加入甲醇(5mL)溶解,冷却至0℃,再加入DIEA(1mL),在30℃下搅拌反应2小时。反应液中加入冰水(1mL)淬灭反应,减压下将反应液浓缩至干,残留物过反相Flash(乙腈/0.1%TFA)纯化,得到5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(((3R,5S)-5-甲基四氢吡咯-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺017-e(125mg)。
LCMS:Rt:0.905min;MS m/z(ESI):362.1,364.1[M+H]。
第四步:1-((2S,4R)-4-((5-氯-2-((1-乙基-1H-吡唑-4-基)氨基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)-2-甲基四氢吡咯-1-基)-丙-2-烯-1-酮017-1的合成
在0℃下,将5-氯-N-(1-乙基-1H-吡唑-4-基)-4-(((3R,5S)-5-甲基四氢吡咯-3-基)氧基)-7H-吡咯并[2,3-d]嘧啶-2-胺017-e(115mg,0.318mmol)溶解在四氢呋喃(5mL)和水(2mL)中,加入磷酸钾(168.8mg,0.795mmol),再加入丙烯酰氯(31.6mg,0.349mmol),室温搅拌反应2小时。反应液减压蒸干,残留物用Prep-HPLC(甲酸体系)制备纯化得到化合物017-1(40.56mg)。
LCMS:Rt:6.819min;MS m/z(ESI):416.1,418.1[M+H].
1H NMR(400M Hz,DMSO-d 6)δ11.51(brs,1H),9.08(d,J=4.8Hz,1H),7.88(brs,1H),7.52(s,1H),7.05(s,1H),6.70-6.52(m,1H),6.16(t,J=16.8Hz,1H),5.82-5.73(m,1H),5.68(q,J=10.0Hz,1H),4.42-4.25(m,1H),4.08(q,J=7.2Hz,2H),4.06-4.02(m,1H),3.95-3.88(m,1H),3.67(d,J=14.4Hz,1H),2.02(dd,J=42.8,14.4Hz,1H),1.41(dd,J=11.2,6.8Hz,3H),1.36(t,J=7.2Hz,3H).
实施例36化合物023-1的制备
1-((3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯-1-基)丙-2-烯-1-酮
Figure PCTCN2021073098-appb-000129
合成路线和具体合成步骤:
Figure PCTCN2021073098-appb-000130
第一步:(3R,4R)-3-((2-氯-7-((2-三甲基硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯-1-甲酸叔丁酯023-b的合成
室温下,将NaH(106mg,2.7mmol)分多批次加入化合物(3R,4R)-4-氟-3-羟基四氢吡咯-1-羧酸叔丁酯023-a(453mg,2.2mmol)的无水DMSO(10.0mL)溶液中,室温下,搅拌15分钟后,将化合物2,4-二氯-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶1b(700mg,2.2mmol)加入,在55℃搅拌1小时后,LCMS监测反应完毕,反应体系中加入乙酸乙酯30mL稀释,用饱和NH 4Cl溶液洗涤三遍(30mL×3),有机相用无水硫酸钠干燥1个小时,过滤,浓缩干得到粗品,过柱(乙酸乙酯:石油醚=30:1至15:1)得到纯品023-b(400mg)。
LCMS:Rt:2.174min;MS m/z(ESI):487.2,489.2[M+H]。
第二步:(3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯-1-羧酸叔丁酯023-d的合成
室温在氮气保护下,将(3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯-1-羧酸叔丁酯023-b(400mg,0.82mmol)和4-氨基-1-乙基吡唑023-c(136.7mg,1.23mmol)溶于1,4-二氧六环(20.0mL),然后在搅拌下,加入Pd 2(dba) 3(75.3mg,0.082mmol),Binap(25.6mg,0.164mmol)和碳酸铯(644mg,1.64mmol),反应体系升温至120℃搅拌12小时,冷却至室温。过滤,滤饼用60mL四氢呋喃洗涤,溶液在减压下浓缩,所得粗产品用柱层色谱纯化(乙酸乙酯:石油醚=15:1至3:1)得到产物023-d(400mg)。
LCMS:Rt:2.017min;MS m/z(ESI):562.3[M+H]。
第三步:(3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯023-e的合成
室温下,将(3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯-1-羧酸叔丁酯023-d(400mg,1.2mmol),在0℃下,加入到三氟乙酸(5mL)的二氯甲烷溶液(5mL)中,室温搅拌2小时。LCMS监测反应结束后,减压下将反应液浓缩,粗产品通过柱层析纯化,得到023-e(200mg)。
LCMS:Rt:1.226min;MS m/z(ESI):332.1[M+H]。
第四步:1-((3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯-1-基)丙-2-烯-1-酮023-1的合成
室温下,将(3R,4R)-3-((2-((1-乙基-1H-吡唑-4-基)氨基)-7H吡咯并[2,3-d]嘧啶-4-基)氧基)-4-氟-四氢吡咯023-e(200mg,0.60mmol)溶解在四氢呋喃(4.0mL)和H 2O(2.0mL)的混合液中。0℃下,先加入磷酸钾固体(318.4mg,1.5mmol),搅拌十分钟后,缓缓滴加丙烯酰氯的四氢呋喃溶液(54.0mg,0.72mmol),室温搅拌2h,过滤,Prep-HPLC(甲酸体系)制备纯化得到化合物023-1(90.0mg)。
LCMS:Rt:7.592min;MS m/z(ESI):386.1[M+H]。
1H NMR(400MHz,DMSO-d 6)δ11.35(s,1H),9.01(s,1H),7.92(s,1H),7.52(s,1H),7.01–6.84(m,1H), 6.64(dd,J=15.7,11.3Hz,1H),6.40–6.09(m,2H),5.92–5.20(m,3H),3.91(m,6H),1.37(t,J=7.2Hz,3H).
生物学活性及相关性质测试例
测试实施例1:BTK激酶活性抑制实验
实验原理:BTK激酶与化合物共同孵育后,在ATP的作用下与底物反应。使用Promega公司的ADP-GLO检测试剂盒对反应产生的ADP进行定量,从而反映对酶活性的抑制作用。
实验仪器:Labcyte公司Echo650移液系统;Perkin Elmer公司Envision酶标仪;Eppendorf公司5810离心机。
实验材料:
试剂 品牌 货号
Tris hydrochloride溶液 Sigma T2663
BRIJ 35 detergent(10%) Merck 203728
MgCl 2溶液 Sigma M1028
ADP-Glo激酶检测试剂盒 Promega V9102
BTK Carna bioscience 08-180
Poly(4:1Glu,Tyr) Sigma P0275
384孔板 Perkin Elmer 6007290
实验方法:用Echo移液系统将待测化合物转移至384孔板中,并且加入2μL/孔的BTK,孵育30分钟。然后加入3μL/孔的底物Poly(4:1Glu,Tyr)和ATP的混合溶液,启动酶反应。化合物终浓度分别从3μM或300nM或100nM起始,3倍稀释。反应中酶的终浓度为1.7ng/孔,ATP终浓度为36μM,底物的终浓度为0.1mg/mL。反应1小时后,加入5μL/孔ADP-GLO试剂,孵育40分钟。然后加入10μL/孔激酶反应检测试剂,孵育30分钟。用Envision酶标仪读取荧光信号,并计算抑制率、半数抑制浓度(IC 50)。
本发明化合物的生物活性通过以上的试验进行测定,测得的IC 50值见下表1。
表1实施例化合物对BTK激酶活性抑制的IC 50
实施例化合物编号 IC 50(nM) 实施例化合物编号 IC 50(nM)
001 47.75 025-2 0.73
002 75.36 026 1.19
003 8.45 027 2.26
004 57.77 028 2.17
005 2.95 029-1 3.02
006-1 12.62 030 1.15
007-1 1.71 031 2.58
008 1.22 032 6.06
009-1 1.07 033 4.04
010 2.59 034 5.97
011-1 0.97 035-1 9.82
012-1 0.98 036 5.69
013-1 0.91 037 11.90
015-1 2.00 038 3.15
016-1 3.94 039 5.78
017-1 2.22 040 1.39
023-1 4.52 041 1.80
025-1 0.98 042 2.79
测试实施例2:JAK3激酶活性抑制实验
实验原理:JAK3激酶与化合物共同孵育后,在ATP的作用下与底物反应。使用Promega公司的ADP-GLO检测试剂盒对反应产生的ADP进行定量,从而反映对酶活性的抑制作用。
实验仪器:
Labcyte公司Echo650移液系统
Perkin Elmer公司Envision酶标仪
Eppendorf公司5810离心机。
实验材料:
试剂 品牌 货号
Tris hydrochloride溶液 Sigma T2663
BRIJ 35 detergent(10%) Merck 203728
MgCl 2溶液 Sigma M1028
ADP-Glo激酶检测试剂盒 Promega V9102
JAK3 Carna bioscience 08-046
Poly(4:1Glu,Tyr) Sigma P0275
384孔板 Perkin Elmer 6007290
试剂 品牌 货号
Tris hydrochloride溶液 Sigma T2663
BRIJ 35detergent(10%) Merck 203728
MgCl 2溶液 Sigma M1028
ADP-Glo激酶检测试剂盒 Promega V9102
JAK3 Carna bioscience 08-046
Poly(4:1Glu,Tyr) Sigma P0275
384孔板 Perkin Elmer 6007290
实验方法:
用Echo移液系统将待测化合物转移至384孔板中,并且加入2μL/孔的JAK3,孵育30分钟。然后加入3μL/孔的底物Poly(4:1Glu,Tyr)和ATP的混合溶液,启动酶反应。化合物终浓度分别从3μM或300nM或100nM起始,3倍稀释。反应中酶的终浓度为1.9ng/孔,ATP终浓度为36μM,底物的终浓度为0.1mg/mL。反应1小时后,加入5μL/孔ADP-GLO试剂,孵育40分钟。然后加入10μL/孔激酶反应检测试剂,孵育30分钟。用Envision酶标仪读取荧光信号,并计算抑制率、半数抑制浓度(IC 50)。
本发明化合物的生物活性通过以上的试验进行测定,测得的IC 50值见下表2。
表2本发明化合物对JAK3激酶活性抑制的IC 50
实施例化合物编号 IC 50(nM) 实施例化合物编号 IC 50(nM)
001 1.92 025-2 0.71
002 1.58 026 0.66
003 4.28 027 1.22
004 2.12 028 1.69
005 1.05 029-1 1.37
006-1 1.37 030 0.71
007-1 0.96 031 0.59
008 0.90 032 2.59
009-1 0.83 033 3.40
010 2.02 034 0.57
011-1 0.99 035-1 3.58
012-1 0.57 036 0.53
013-1 1.14 037 0.43
015-1 0.29 038 0.48
016-1 0.35 039 10.06
017-1 0.61 040 0.41
023-1 1.21 041 1.69
025-1 1.42 042 0.35
测试实施例3:对Ramos细胞中BTK磷酸化的抑制作用
实验原理:将Ramos细胞与化合物和刺激剂孵育后,使用Cisbio公司的BTK磷酸化检测试剂盒,通过均相时间分辨荧光(HTRF)的方法检测荧光能量的转移,从而反映对磷酸化的抑制作用。
实验仪器:
仪器 品牌 型号
生物安全柜 ESCO CLASSⅡBSC
离心机 Eppendorf 5810
CO 2培养箱 ESCO CCL-170B-8
细胞计数仪 CountStar IC1000
Envision Perkin Elmer /
实验材料:
Figure PCTCN2021073098-appb-000131
Figure PCTCN2021073098-appb-000132
实验方法:
用Echo移液系统将待测化合物转移至384孔板中,将Ramos细胞密度调整为1X10 7细胞/mL,加入10μL/孔细胞悬液,在37℃,5%CO 2的培养箱中孵育1小时。然后加入5μL/孔的刺激剂anti-human IgM抗体,刺激剂终浓度为10μg/mL,孵育10分钟。化合物终浓度为1μM起始,4倍稀释。加入5μL/孔的细胞裂解液,室温孵育30分钟。使用Cisbio公司的BTK phospho-Y223试剂盒对BTK的磷酸化程度进行检测,最终在Envision酶标仪上读取发射光665nm和615nm下的荧光信号,计算抑制率和半数抑制浓度(IC 50)。
本发明化合物的生物活性通过以上的试验进行测定,测得的IC 50值见下表3。
表3本发明化合物对Ramos细胞中BTK磷酸化抑制活性
实施例化合物编号 IC 50(nM) 实施例化合物编号 IC 50(nM)
005 44.96 028 21.91
007-1 0.88 029-1 2.68
008 4.83 030 3.76
009-1 8.59 031 21.41
010 9.66 032 29.49
011-1 4.98 033 53.20
012-1 2.36 034 53.36
013-1 18.24 035-1 20.46
015-1 7.68 036 2.84
016-1 35.58 037 4.15
017-1 4.91 038 13.87
023-1 10.69 039 41.73
025-1 8.82 040 11.85
025-2 5.80 041 8.59
026 12.61 042 1.68
027 19.39    
测试实施例4:对CTLL-2细胞中STAT5磷酸化的抑制作用
实验原理:此实验是评价化合物对JAK3下游底物STAT5磷酸化的影响。将CTLL-2细胞与化合物和刺激剂孵育后,使用Perkin Elmer公司的p-STAT5(Tyr694/699)检测试剂盒,通过时间分辨荧光的方法检测供体微珠和受体微珠之间的荧光能量转移,从而反映对磷酸化的抑制作用。
实验仪器:
仪器 品牌 型号
生物安全柜 Thermo Scientific 1300 Series A2
离心机 Eppendorf 5702
CO 2培养箱 Thermo Scientific 371
细胞计数仪 Invitrogen C10281
Envision Perkin Elmer /
Echo Labcyte 655
实验材料:
Figure PCTCN2021073098-appb-000133
实验方法:
将CTLL-2细胞种在384孔板中,1.5X10 4细胞/15μL/孔,用Echo将化合物转移至384孔板中,在37℃,5%CO 2的培养箱中孵育30分钟。然后加入5μL/孔的刺激剂IL-2,终浓度为1ng/mL,孵育30分钟。化合物终浓度从3μM起始,3倍稀释。加入5μL/孔细胞裂解液,室温孵育10分钟。用Perkin Elmer公司的AlphaLISA p-STAT5(Tyr694/699)检测试剂盒对STAT5的磷酸化程度进行检测,最终在Envision酶标仪上读取AlphaLISA信号,计算抑制率和半数抑制浓度(IC 50)。
本发明化合物的生物活性通过以上的试验进行测定,测得的IC 50值见下表4。
表4本发明化合物对CTLL-2细胞中STAT5磷酸化的抑制活性
实施例化合物编号 IC 50(nM) 实施例化合物编号 IC 50(nM)
005 101.12 027 266.74
007-1 15.78 028 28.54
008 21.02 029-1 186.14
009-1 15.98 030 12.26
010 34.75 031 225.68
011-1 17.84 032 265.45
012-1 20.94 033 24.55
015-1 20.72 034 186.84
016-1 97.99 035-1 271.87
017-1 23.02 036 28.70
023-1 30.94 037 33.15
025-1 48.75 038 155.80
025-2 34.65 040 162.37
026 44.89 042 29.53
测试实施例5:对小鼠脾脏中BTK靶点的占据
实验原理:此实验是评价化合物在小鼠脾脏中对BTK靶点的占据。将冷冻的脾脏样品匀浆,然后与带生物素标记的探针化合物孵育,化合物未占据的BTK蛋白与探针结合,化合物已经占据的BTK蛋白则不能与探针结合,通过ELISA方法检测,从而反映化合物对BTK靶点的占据。
实验仪器:
仪器 品牌 型号
离心机 Eppendorf 5810
离心机 Eppendorf 5430R
组织研磨仪 美壁 LD48
Envision Perkin Elmer /
实验材料:
试剂 品牌 货号
Anti-BTK抗体 Cell signal 8547S
链霉亲和素包被板 R&D Systems CP004
CNX-500 MCE HY-100338
ELISA显色液 R&D Systems DY999
ELISA终止液 R&D Systems DY994
RIPA裂解液 Sigma R0278
Anti-rabbit IgG抗体 Cell signal 7074S
BCA Protein Assay试剂盒 Pierce 23225
实验方法:
使用C57BL/6N雌性小鼠,将待测化合物配制在2%Tween80/0.5%methycellulose溶液中灌胃给药,剂量为10mg/kg,给药后0.5h或者24小时后取脾脏保存于干冰中。将冷冻的脾脏样品匀浆并用BCA试剂盒检测蛋白浓度。将蛋白浓度调整一致后的脾脏匀浆液与CNX-500孵育1小时,CNX-500终浓度为1μM。然后转移100μL/孔至链霉亲和素包被板中孵育过夜。弃上清并清洗,加入anti-BTK抗体孵育2小时。弃上清并清洗,加入HRP标记的Anti-rabbit IgG抗体,孵育1小时。弃上清并清洗,用显色液显色10-15分钟,终止反应后,使用Envision在波长450nm下读取吸光度值。计算占据比率,占据比率计算公式为:
Figure PCTCN2021073098-appb-000134
其中:
信号 max表示:对照组样品加入探针化合物后产生的信号;
信号 min表示:对照组样品不加探针化合物后产生的信号;
信号 待测化合物表示:给待测化合物的样品加入探针化合物后产生的信号。
实验结果如表5所示。
表5本发明化合物对小鼠脾脏中BTK靶点的占据
Figure PCTCN2021073098-appb-000135
测试实施例6:对小鼠全血中IL-2诱导的STAT5磷酸化的抑制作用
实验原理:此实验是评价化合物对JAK3下游底物STAT5磷酸化的影响。小鼠口服给药后取全血,加入刺激剂IL-2孵育15分钟,通过流式细胞技术检测淋巴细胞中STAT5磷酸化水平,从而反映化合物对JAK3靶点的抑制作用。
实验仪器:
仪器 品牌 型号
生物安全柜 ESCO CLASSⅡ BSC
离心机 Eppendorf 5810
CO 2培养箱 ESCO CCL-170B-8
Flow cytometer BD Biosciences Canto II
-4℃冷藏箱 海尔 HYC-650
实验材料:
试剂 品牌 货号
AF647 Mouse Anti-pStat5抗体 BD Biosciences 562076
BV 421 anti-mouse CD8抗体 Biolegend 100738
BV421 Anti-Mouse CD3e抗体 BD Biosciences 740014
FITC Anti-Mouse CD4抗体 BD Biosciences 553047
IL-2 R&D 402-ML-020
小鼠Fc封闭抗体 Biolegend 156603
裂解固定液 BD Biosciences 558049
细胞染色液 Biolegend 420201
96孔板 Corning 3799
Perm Buffer III BD Biosciences 558050
96孔深孔板 Axygen P-96-450V-C
实验方法:
使用C57BL/6N雌性小鼠,将待测化合物配制在2%Tween80/0.5%methycellulose溶液中,灌胃给药,剂量为10mg/kg,给药0.5或4小时后取全血置于肝素钠抗凝管中。将全血80μL/孔种于96孔板中,加入5μL/孔小鼠Fc封闭抗体,然后再加入检测抗体5μL/孔,不同批次实验中检测抗体为CD8抗体或者CD3抗体/CD4抗体的混合液。加入10μL/孔刺激剂IL-2,孵育15分钟,刺激剂终浓度为200ng/mL。将全血转移60μL/孔至96孔深孔板中,并加入裂解固定液350μL/孔,孵育10分钟。离心弃上清后,将细胞重悬于 100μL/孔Perm Buffer III,孵育30分钟。离心弃上清,加入pSTAT5抗体50μL/孔,孵育30分钟。离心弃上清后,将细胞重悬于染色液中,用流式细胞仪检测pSTAT5的信号,并计算抑制率。抑制率计算公式为:
Figure PCTCN2021073098-appb-000136
其中:
信号 max表示:对照组样品加入刺激剂IL-2后产生的信号;
信号 min表示:对照组样品不加刺激剂IL-2后产生的信号;
信号 待测化合物表示:给待测化合物的样品加入刺激剂IL-2后产生的信号。
实验结果如表6所示。
表6本发明化合物对小鼠全血中IL-2诱导的STAT5磷酸化的抑制作用
Figure PCTCN2021073098-appb-000137
测试实施例7 CAIA小鼠模型药效
6-8周龄DBA/1雄性小鼠,第0天,II型胶原的5种单克隆抗体(Arthrogen-CIA 5-Clone Cocktail Kit;Chrondex)的混合液皮下注射到DBA/1小鼠中,抗体混合液的浓度为10mg/ml,每只小鼠注射150μl,相当于每只小鼠注射1.5mg;II型胶原抗体诱导后第3天,小鼠腹腔注射脂多糖(LPS)100μl,浓度为0.5mg/ml,相当于每只小鼠50μg。
小鼠在第4天出现病症,在第7-8天疾病达到高峰期。根据动物临床症状进行评分。0:无红斑和红肿;1:近跗骨附近或踝关节或跖骨出现红斑或轻度红肿,1个脚趾红肿;2:踝关节和跖骨轻微红斑和肿胀,或超过两个脚趾红肿;3:踝、腕关节和跖骨中度红斑和肿胀;4:踝、腕关节,跖骨和脚趾全部严重红肿。
CAIA小鼠治疗方案:动物分组分别为阴性对照组、阳性对照组及实验组。其中实验组设置化合物03610mg/kg和3mg/kg。
阳性对照组给予Dexamethasone 1mg/kg,Evobrutinib 3mg/kg,Tofacitinib 30mg/kg。诱导第5天根据小鼠发病临床评分进行分组,确保在给药干预前每组小鼠的发病的严重程度一致,然后灌胃给药(Dexamethasone腹腔注射给药),阴性对照组在相同时间点灌胃等体积溶媒(2%Tween80/0.5%methycellulose溶液)。每天记录小鼠评分及体重。
根据图1和图2所示的实验结果表明:经过化合物036治疗后,CAIA小鼠的临床症状明显改善,临床评分与阴性对照组有统计学差异,化合物036的两个剂量在对该疾病模型表现出一定的量效关系。
组织形态学鉴定:诱导第13天处死小鼠,取小鼠四肢,浸泡脱钙液中,固定后石蜡包埋,以5μm厚度在整个踝关节处切片,脱腊和梯度水合后进行H&E染色。染色后扫片机扫描片子,并由独立第二人盲法病理评分。分别对关节炎症和骨损伤情况进行评价,关节周边组织炎症评分:0:无炎症细胞或仅有极少量的炎症细胞浸润到软骨区域;1:骨关节周围有轻到中度的炎症细胞浸润;2:骨或骨连接处有严重的炎症细胞浸润;关节骨损伤评分:0:软骨及骨关节处无明显损伤;1:骨及外周骨关节有轻度破坏性损伤;2:骨及骨关节处有严重的结构性破坏。分别对双后肢及前肢进行评价,每个后肢及前肢每项最高评分2分,每个前肢或后肢最高评分4分,总计16分。
图3和图4所示的实验结果表明:各治疗组关节病变有明显改善,与阴性对照组相比具有显著性差异。
以上,对本发明的实施方式进行了说明。但是,本发明不限定于上述实施方式。凡在本发明的精神和原则之内,所做的任何修改、等同替换、改进等,均应包含在本发明的保护范围之内。

Claims (18)

  1. 式(I)所示化合物或其药学上可接受的盐:
    Figure PCTCN2021073098-appb-100001
    其中,
    R 1选自H、
    Figure PCTCN2021073098-appb-100002
    或任选被R a1取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
    R 2选自氢、F、Cl、Br、I、CN、OH、NO 2
    Figure PCTCN2021073098-appb-100003
    或任选被R a2取代的以下基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
    R 5、R 6、R 7独立地选自氢、F、Cl、Br、I、CN或任选被R a3取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
    X 1、X 2、X 3独立地选自CR 8或N;
    R 8选自H、F、Cl、Br、I、CN、OH或任选被R a4取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
    R 9、R 10、R 12独立地选自任选被R a5取代的以下基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
    R 11选自H或任选被R a6取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
    每一个R a1、R a2、R a3、R a4独立选自F、Cl、Br、I、OH、CN、=O、NO 2
    Figure PCTCN2021073098-appb-100004
    Figure PCTCN2021073098-appb-100005
    硼酸基、硼酸酯基或任选被R b取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
    每一个R a5、R a6独立选自F、Cl、Br、I、OH、CN、=O、NO 2或任选被R b取代的下列基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
    每一个R b独立选自F、Cl、Br、I、OH、CN、=O、NO 2或任选被R c取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基、C 2-C 10烯基、C 2-C 10炔基、C 6-C 10芳基、5-10元杂芳基、C 6-C 10芳基氧基或5-10元杂芳基氧基;
    每一个R c独立选自F、Cl、Br、I、OH、CN、=O、NO 2、NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基;
    L选自
    Figure PCTCN2021073098-appb-100006
    R 3、R 4独立地选自H、F、Cl、Br、I、OH、CN、=O、NO 2或任选被R c取代的下列基团:NH 2、SH、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基、3-10元杂环基氧基;
    Y、Z独立选自NH、O、S、
    Figure PCTCN2021073098-appb-100007
    m、n1、n2、n3、q1、q2独立选自0、1或2,p1选自1、2、3或4;
    条件是:当Y为NH,且当R 3为H、F、二氟甲基或三氟甲基时,R 1不为甲基;当Y为O,且当R 3、R 4均为H时,R 1不为甲基;当L选自
    Figure PCTCN2021073098-appb-100008
    时,R 2不为H、CN、
    Figure PCTCN2021073098-appb-100009
    Figure PCTCN2021073098-appb-100010
  2. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R a1、R a2独立选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基。
  3. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R a3、R a4、R a5、R a6独立选自F、Cl、Br、I、CN、=O或任选被R b取代的下列基团:OH、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基。
  4. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R b选自任选被R c取代的下列基团:C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 6-C 10芳基或5-10元杂芳基。
  5. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R c选自F、Cl、Br、I、OH、CN、=O、NH 2或C 1-C 10烷基。
  6. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述L选自
    Figure PCTCN2021073098-appb-100011
    Figure PCTCN2021073098-appb-100012
  7. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述L选自
    Figure PCTCN2021073098-appb-100013
  8. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,m、n1、n2、n3、q2独立选自0或1,和/或p1、q1选自1或2。
  9. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 1选自H、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基,所述C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基任选被R a1取代。
  10. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 1选自C 1-C 3烷基或5-6元杂环烷基,所述5-6元杂环烷基含有1~2个O原子,所述C 1-C 3烷基或5-6元杂环烷基任选被R a1取代。
  11. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 2选自H、F、Cl、Br、I、CN、NH 2、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基,所述NH 2、 C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基任选被R a2取代。
  12. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 2选自H、F、Cl、Br、I、CN、C 1-C 6烷基、COOH或CONH 2,所述C 1-C 6烷基、COOH或CONH 2任选被R b取代。
  13. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 3、R 4独立地选自H、F、Cl、Br、I、OH、CN或任选被R c取代的下列基团:NH 2、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基。
  14. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自式(II)所示化合物或其药学可接受的盐:
    Figure PCTCN2021073098-appb-100014
    其中,R 1、R 2、R 5、X 1、L如权利要求1中所定义。
  15. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自式(III)所示化合物或其药学可接受的盐:
    Figure PCTCN2021073098-appb-100015
    其中,R 1、R 2、X 1、L如权利要求1中所定义。
  16. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自以下化合物或其药学可接受的盐:
    Figure PCTCN2021073098-appb-100016
    Figure PCTCN2021073098-appb-100017
    Figure PCTCN2021073098-appb-100018
    Figure PCTCN2021073098-appb-100019
    Figure PCTCN2021073098-appb-100020
  17. 一种药物组合物,所述组合物包含权利要求1至16任一项的化合物或其药学上可接受的盐,以及药学上可接受的辅料。
  18. 权利要求1至16任一项的化合物或其药学上可接受的盐、或权利要求17所述的药物组合物在制备预防或者治疗Janus激酶(JAK)和/或布鲁顿酪氨酸激酶(BTK)相关性疾病的药物中的用途。
PCT/CN2021/073098 2020-01-21 2021-01-21 嘧啶并五元环类衍生物及其应用 WO2021147953A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180010745.4A CN115244055A (zh) 2020-01-21 2021-01-21 嘧啶并五元环类衍生物及其应用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202010072567.5 2020-01-21
CN202010072567 2020-01-21
CN202010263801 2020-04-07
CN202010263801.2 2020-04-07

Publications (1)

Publication Number Publication Date
WO2021147953A1 true WO2021147953A1 (zh) 2021-07-29

Family

ID=76993289

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/073098 WO2021147953A1 (zh) 2020-01-21 2021-01-21 嘧啶并五元环类衍生物及其应用

Country Status (2)

Country Link
CN (1) CN115244055A (zh)
WO (1) WO2021147953A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114315838A (zh) * 2020-09-30 2022-04-12 江苏先声药业有限公司 嘧啶并吡咯类化合物

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103814030A (zh) * 2011-09-22 2014-05-21 辉瑞大药厂 吡咯并嘧啶及嘌呤衍生物
CN103874699A (zh) * 2011-09-20 2014-06-18 赛尔佐姆有限公司 吡唑并[4,3-c]吡啶衍生物作为激酶抑制剂
CN104311573A (zh) * 2013-09-18 2015-01-28 北京韩美药品有限公司 抑制btk和/或jak3激酶活性的化合物
CN107417684A (zh) * 2013-12-05 2017-12-01 辉瑞公司 杂环丙烯酰胺
CN108864057A (zh) * 2017-05-16 2018-11-23 山东大学 含有4-氨基吡唑结构的jak与hdac双靶点抑制剂及其制备方法和应用
CN109311896A (zh) * 2016-06-30 2019-02-05 株式会社大熊制药 吡唑并嘧啶衍生物作为激酶抑制剂
WO2019132562A1 (ko) * 2017-12-28 2019-07-04 주식회사 대웅제약 카이네이즈 저해제로서의 옥시-플루오로피페리딘 유도체
WO2019132560A1 (ko) * 2017-12-28 2019-07-04 주식회사 대웅제약 카이네이즈 저해제로서의 아미노-플루오로피페리딘 유도체
WO2019132561A1 (ko) * 2017-12-28 2019-07-04 주식회사 대웅제약 카이네이즈 저해제로서의 아미노-메틸피페리딘 유도체

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103874699A (zh) * 2011-09-20 2014-06-18 赛尔佐姆有限公司 吡唑并[4,3-c]吡啶衍生物作为激酶抑制剂
CN103814030A (zh) * 2011-09-22 2014-05-21 辉瑞大药厂 吡咯并嘧啶及嘌呤衍生物
CN104311573A (zh) * 2013-09-18 2015-01-28 北京韩美药品有限公司 抑制btk和/或jak3激酶活性的化合物
CN107417684A (zh) * 2013-12-05 2017-12-01 辉瑞公司 杂环丙烯酰胺
CN109311896A (zh) * 2016-06-30 2019-02-05 株式会社大熊制药 吡唑并嘧啶衍生物作为激酶抑制剂
CN108864057A (zh) * 2017-05-16 2018-11-23 山东大学 含有4-氨基吡唑结构的jak与hdac双靶点抑制剂及其制备方法和应用
WO2019132562A1 (ko) * 2017-12-28 2019-07-04 주식회사 대웅제약 카이네이즈 저해제로서의 옥시-플루오로피페리딘 유도체
WO2019132560A1 (ko) * 2017-12-28 2019-07-04 주식회사 대웅제약 카이네이즈 저해제로서의 아미노-플루오로피페리딘 유도체
WO2019132561A1 (ko) * 2017-12-28 2019-07-04 주식회사 대웅제약 카이네이즈 저해제로서의 아미노-메틸피페리딘 유도체

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114315838A (zh) * 2020-09-30 2022-04-12 江苏先声药业有限公司 嘧啶并吡咯类化合物

Also Published As

Publication number Publication date
CN115244055A (zh) 2022-10-25

Similar Documents

Publication Publication Date Title
CN105732636B (zh) 杂芳化合物及其在药物中的应用
TWI623530B (zh) 雙環稠合之雜芳基或芳基化合物
KR101837223B1 (ko) 피리디논/피라지논, 그의 제조 방법 및 사용 방법
CN103038233B (zh) 吡啶酮和氮杂吡啶酮化合物及使用方法
CN104125959B (zh) 作为btk活性的抑制剂的杂芳基吡啶酮和氮杂‑吡啶酮化合物
CN102458581B (zh) 作为JANUS激酶抑制剂的吡唑-4-基-吡咯并[2,3-d]嘧啶和吡咯-3-基-吡咯并[2,3-d]嘧啶的N-(杂)芳基-吡咯烷衍生物
TWI672304B (zh) 作爲酪胺酸激酶抑制劑之經取代的乙炔基雜雙環化合物
KR20220007889A (ko) Fgfr 저해제 및 이의 사용 방법
CN107344940B (zh) Btk抑制剂及其用途
CN103288833A (zh) 作为激酶抑制剂的咪唑并三嗪和咪唑并嘧啶
TW201632523A (zh) 4,6-經取代之吡唑并[1,5-a]吡
JP7370617B2 (ja) アミノノルカンファン誘導体及びその製造方法と使用
CN113321654B (zh) 作为激酶抑制剂的稠合吡啶酮类化合物
WO2018221433A1 (ja) ヘテロアリールアミン誘導体
CN107531679A (zh) 芳香酰胺类衍生物、其制备方法及其在医药上的应用
WO2022068929A1 (zh) 嘧啶二酮类化合物及其用途
TW202110848A (zh) 取代的稠合雙環類衍生物、其製備方法及其在醫藥上的應用
CN115867346A (zh) 激酶抑制剂
WO2021147952A1 (zh) 嘧啶并吡咯类化合物
WO2021147953A1 (zh) 嘧啶并五元环类衍生物及其应用
CN112739420A (zh) 苯氧基-吡啶基-嘧啶化合物及使用方法
CN111315750B (zh) 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物
WO2023093728A1 (zh) Brm选择性降解剂化合物及其应用
WO2023134629A1 (zh) 核转运调节剂及其用途
WO2022062601A1 (zh) 嘧啶并吡咯类化合物

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: 21744025

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21744025

Country of ref document: EP

Kind code of ref document: A1