WO2021104441A1 - 作为egfr激酶抑制剂的多芳基化合物 - Google Patents

作为egfr激酶抑制剂的多芳基化合物 Download PDF

Info

Publication number
WO2021104441A1
WO2021104441A1 PCT/CN2020/132188 CN2020132188W WO2021104441A1 WO 2021104441 A1 WO2021104441 A1 WO 2021104441A1 CN 2020132188 W CN2020132188 W CN 2020132188W WO 2021104441 A1 WO2021104441 A1 WO 2021104441A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
amino
formula
methyl
methoxy
Prior art date
Application number
PCT/CN2020/132188
Other languages
English (en)
French (fr)
Inventor
刘磊
刘扬
周峰
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 CN202080081283.0A priority Critical patent/CN114728932A/zh
Publication of WO2021104441A1 publication Critical patent/WO2021104441A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This application relates to polyaryl compounds as EGFR kinase inhibitors, their preparation methods, pharmaceutical compositions containing the compounds, and their use in the treatment of EFGR kinase-related diseases.
  • EGFR Epithelial growth factor Receptor
  • EGF epithelial growth factor
  • EGFR belongs to the ErbB receptor family, which includes EGFR (ErbB-1), HER2/c-neu (ErbB-2), HER3 (ErbB-3) and HER4 (ErbB-4).
  • EGFR is also called HER1, ErbB1.
  • EGFR is widely distributed on the surface of mammalian epithelial cells, fibroblasts, glial cells, keratinocytes and other cells. The EGFR signaling pathway plays an important role in the physiological processes of cell growth, proliferation and differentiation.
  • EGFR is divided into three regions: extracellular ligand binding region, transmembrane region and intracellular kinase region.
  • extracellular ligand binding region When EGFR is bound by the corresponding ligands, it will induce the formation of homotypic or heterodimers, thereby activating the intracellular tyrosine kinase pathway and making it autophosphorylate, thereby guiding downstream phosphorylation, including MAPK, Akt and JNK Pathway to induce cell proliferation.
  • EGFR tyrosine kinase inhibitors (Tyrosine Kinase Inhibitor, TKI) block tumors by blocking the binding of endogenous ATP to the intracellular kinase region, thereby inhibiting receptor phosphorylation and the activation of downstream signal transduction molecules Proliferation of cells.
  • TKI Tyrosine Kinase Inhibitor
  • This application relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof,
  • R 1 is selected from a phenyl group and a 5-6 membered heteroaryl group, the phenyl group and a 5-6 membered heteroaryl group are optionally substituted by R 7;
  • R 1 is wherein R 11 and R 12 are independently selected from C 1-4 alkyl, and the C 1-4 alkyl is optionally substituted by hydroxy or C 1-4 alkoxy;
  • R 2 is a 4-14 membered heterocycloalkyl group, and the 4-14 membered heterocycloalkyl group is optionally substituted by R 8;
  • U, V, and W are independently selected from CR 6 and N;
  • Ring A is selected from phenyl and 5-6 membered heteroaryl groups
  • R 3 is selected from H, halogen, cyano, C 1-4 alkyl and C 1-4 alkoxy;
  • R 4 and R 5 are independently selected from C 1-4 alkyl and C 1-4 alkoxy;
  • R 6 is independently selected from H, C 1-4 alkyl, C 1-4 alkoxy and halogen
  • R 7 is independently selected from halogen, hydroxy, amino, C 1-4 alkyl and C 1-4 alkoxy, the C 1-4 alkyl and C 1-4 alkoxy are optionally substituted by hydroxy;
  • R 8 is independently selected from halogen, OH, C 1-6 alkyl, C 1-6 alkoxy, amino, C 3-8 cycloalkyl and 3-8 membered heterocycloalkyl, the amino, C 3 -8 cycloalkyl and 3-8 membered heterocycloalkyl are optionally substituted by R 9;
  • R 9 is independently selected from halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 acyl and oxo.
  • R 1 is selected from 5-6 membered heteroaryl, which is optionally substituted by R 7.
  • R 1 is selected from 5-membered heteroaryl, which is optionally substituted by R 7.
  • R 1 is selected from phenyl, pyridyl, pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, tetrazolyl and triazolyl, the phenyl, pyridine , pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, tetrazolyl and triazolyl optionally substituted with R 7.
  • R 1 is selected from pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, tetrazolyl, and triazolyl, the pyrrolyl, furyl, thienyl, imidazole , oxazolyl, pyrazolyl, tetrazolyl and triazolyl optionally substituted with R 7.
  • R 1 is selected from phenyl, pyridyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl and triazolyl, the phenyl, pyridyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl and triazolyl optionally substituted with R 7.
  • R 1 is selected from phenyl, pyridyl, pyrazolyl, and imidazolyl, and the phenyl, pyridyl, pyrazolyl, and imidazolyl are optionally substituted with R 7 .
  • R 1 is selected from the group consisting of pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, and triazolyl, the pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl and triazole The group is optionally substituted by R 7.
  • R 7 is independently selected from halogen, hydroxy, and C 1-4 alkyl, which C 1-4 alkyl is optionally substituted with hydroxy.
  • R 7 is independently selected from C 1-4 alkyl, said C 1-4 alkyl is optionally substituted with hydroxy.
  • R 7 is independently selected from methyl and hydroxyethyl.
  • R 7 is independently selected from methyl.
  • R 1 is selected from phenyl
  • R 1 is selected from phenyl
  • R 1 is selected from
  • R 1 is Wherein R 11 is selected from C 1-4 alkyl, R 12 is selected from optionally hydroxy substituted C 1-4 alkyl.
  • R 1 is Wherein R 11 is methyl and R 12 is methyl or hydroxyethyl.
  • R 2 is a 5-12 membered heterocycloalkyl group, which is optionally substituted with R 8 .
  • R 2 is a 6-11 membered heterocycloalkyl group, which is optionally substituted with R 8 .
  • R 2 is selected from piperidinyl, tetrahydropyranyl, morpholinyl, piperazinyl, 1,4-thiaoxanyl, 1,4-dioxanyl, thiomethanyl Linyl, 1,3-dithianyl, 1,4-dithianyl, azepanyl, oxepanyl, thieppanyl, 3,9-diazaspiro [5.5] Undecyl, 2,9-diazaspiro[5.5] Undecyl, The piperidinyl, tetrahydropyranyl, morpholinyl, piperazinyl, 1,4-thiooxanyl, 1,4-dioxanyl, thiomorpholinyl, 1,3-bis Thianyl, 1,4-dithianyl, azepanyl, oxepanyl, thiepanyl, 3,9-diazaspiro[5.5]undec
  • R 2 is selected from piperidinyl, tetrahydropyranyl, morpholinyl, piperazinyl, 1,4-thiaoxanyl, 1,4-dioxanyl, thiomethanyl Linyl, 1,3-dithianyl, 1,4-dithianyl, azepanyl, oxepanyl, thieppanyl, 3,9-diazaspiro [5.5] Undecyl, 2,9-diazaspiro[5.5] Undecyl, The piperidinyl, tetrahydropyranyl, morpholinyl, piperazinyl, 1,4-thiooxanyl, 1,4-dioxanyl, thiomorpholinyl, 1,3-bis Thianyl, 1,4-dithianyl, azepanyl, oxepanyl, thiepanyl, 3,9-diazaspiro[5.5]undec
  • R 2 is selected from piperidinyl, piperazinyl, morpholinyl, 3,9-diazaspiro[5.5]undecyl, The piperidinyl, piperazinyl, morpholinyl, 3,9-diazaspiro[5.5]undecyl, Optionally substituted by R 8.
  • R 2 is selected from piperidinyl, piperazinyl, morpholinyl, 3,9-diazaspiro[5.5]undecyl, The piperidinyl, piperazinyl, morpholinyl, 3,9-diazaspiro[5.5]undecyl, Optionally substituted by R 8.
  • R 2 is selected from piperidinyl, piperazinyl, morpholinyl, The piperidinyl, piperazinyl, morpholinyl, Optionally substituted by R 8.
  • R 2 is selected from piperidinyl, morpholinyl and 3,9-diazaspiro[5.5]undecyl, the piperidinyl, morpholinyl and 3,9-diaza hetero-spiro [5.5] undec-alkyl is optionally substituted with R 8.
  • R 8 is independently selected from halogen, C 1-6 alkyl, C 1-6 alkoxy, amino, C 3-8 cycloalkyl, and 3-8 membered heterocycloalkyl, the Amino, C 3-8 cycloalkyl, and 3-8 membered heterocycloalkyl are optionally substituted with R 9.
  • R 8 is independently selected from halogen, OH, C 1-4 alkyl, amino, C 3-7 cycloalkyl, and 3-7 membered heterocycloalkyl, the amino, C 3-7 cycloalkyl, and 3-7 membered heterocycloalkyl is optionally substituted with R 9.
  • R 8 is independently selected from halogen, OH, amino, and 3-7 membered heterocycloalkyl, which amino and 3-7 membered heterocycloalkyl are optionally substituted with R 9.
  • R 8 is independently selected from C 1-4 alkyl, amino, C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl, the amino, C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl is optionally substituted with R 9.
  • R 8 is independently selected from F, OH, methyl, amino, piperazinyl, 1,3-oxazinyl, 1,4-diazepanyl, 1,3- Oxazepinyl, 1,4-oxazepinyl, oxetanyl, morpholinyl, pyrrolidinyl, tetrahydropyranyl, azetidinyl, The amino group, piperazinyl group, 1,3-oxazinyl group, 1,4-diazepanyl group, 1,3-oxazepinyl group, 1,4-oxazepine group Cycloheptanyl, oxetanyl, morpholinyl, pyrrolidinyl, tetrahydropyranyl, azetidinyl are optionally substituted with R 9.
  • R 8 is independently selected from F, OH, methyl, amino, piperazinyl, 1,3-oxazinyl, 1,4-diazepanyl, 1,3- Oxazepanyl, 1,4-oxazepanyl, oxetanyl, morpholinyl, pyrrolidinyl, tetrahydropyranyl, the amino group, piperazinyl , 1,3-oxazinyl, 1,4-diazepanyl, 1,3-oxazepinyl, 1,4-oxazepinyl, oxazepin Cyclobutanyl, morpholinyl, pyrrolidinyl, and tetrahydropyranyl are optionally substituted with R 9.
  • R 8 is independently selected from F, OH, amino, piperazinyl, 1,4-oxazepanyl, oxetanyl, morpholinyl, pyrrolidinyl, Tetrahydropyranyl, the amino group, piperazinyl group, 1,4-oxazepinyl group, oxetanyl group, morpholinyl group, pyrrolidinyl group, tetrahydropyranyl group optionally Replaced by R 9.
  • R 8 is independently selected from methyl, amino, piperazinyl, 1,3-oxazinyl, 1,4-diazepanyl, 1,3-oxazepine Cycloheptanyl and 1,4-oxazepanyl, the amino group, piperazinyl, 1,3-oxazinyl, 1,4-diazepanyl, 1,3 - oxa azepanyl and 1,4-oxa-azepanyl R 9 is optionally substituted.
  • R 9 is independently selected from halogen, C 1-4 alkyl, C 1-4 acyl, C 1-4 alkoxy, and oxo.
  • R 9 is independently selected from halogen, C 1-4 alkyl, and C 1-4 acyl.
  • R 9 is independently selected from halogen, C 1-4 alkyl, C 1-4 alkoxy, and oxo.
  • R 9 is independently selected from F, methyl, acetyl, and oxo.
  • R 9 is independently selected from F, methyl, and acetyl.
  • R 9 is independently selected from methyl and oxo.
  • R 2 is selected from
  • R 2 is selected from
  • R 2 is selected from
  • R 2 is selected from
  • U is CR 6 .
  • R 6 is independently selected from C 1-4 alkoxy.
  • R 6 is methoxy
  • V is selected from CH and N.
  • W is selected from CH and N.
  • ring A is selected from phenyl, pyrazinyl, and pyridyl.
  • ring A is selected from phenyl and pyrazinyl.
  • R 3 is selected from halogen and C 1-4 alkyl.
  • R 3 is selected from Cl and Br.
  • R 4 and R 5 are independently selected from C 1-3 alkyl.
  • R 4 and R 5 are methyl groups.
  • the compound of formula (I) of the present application is selected from the compound of formula (II),
  • R 1 , R 2 , R 3 , R 6 and V are as defined above.
  • the compound of formula (I) of the present application is selected from the compound of formula (III),
  • R 1 , R 2 , R 3 , R 6 and V are as defined above.
  • the compound of formula (I) of the present application is selected from the following compounds:
  • the application relates to a pharmaceutical composition, which comprises the compound of formula (I) of the application or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable excipients.
  • this application relates to a method for treating a disease mediated by EGFR in a mammal, including administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof to a mammal in need of such treatment, preferably a human, Or its pharmaceutical composition.
  • this application relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof in the preparation of a medicine for preventing or treating EGFR-mediated diseases.
  • this application relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt or a pharmaceutical composition thereof in the prevention or treatment of EGFR-mediated diseases.
  • this application relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for preventing or treating EGFR-mediated diseases.
  • 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
  • C mn in this document means that the part has an integer number of carbon atoms in a given range.
  • C 1-6 means that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms.
  • any variable such as R
  • its definition in each case is independent. For example, if a group is replaced by 2 Rs, then each R has independent options.
  • halo or halogen refers to fluorine, chlorine, bromine and iodine.
  • hydroxy refers to the -OH group.
  • cyano refers to the -CN group.
  • amino refers to the -NH 2 group.
  • alkyl refers to a hydrocarbon group of the general formula C n H 2n+1.
  • the alkyl group may be linear or branched.
  • C 1 - 6 alkyl refers to (e.g., methyl, ethyl, n-propyl, isopropyl, alkyl containing 1 to 6 carbon atoms, n-butyl, isobutyl, sec-butyl, Tert-butyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, neopentyl, hexyl, 2-methylpentyl, etc.).
  • the alkyl moiety (ie, alkyl) of alkoxy, alkylamino, dialkylamino, alkylsulfonyl, and alkylthio have the same definition as described above.
  • alkoxy refers to -O-alkyl.
  • C 1-4 alkoxy can be methoxy, ethoxy, n-propyloxy, isopropyloxy, n-butyloxy, isobutyloxy, sec-butyloxy or Tert-butyloxy and so on.
  • cycloalkyl refers to a carbocyclic ring that is fully saturated and may exist as a monocyclic, bridged, or spiro ring. Unless otherwise indicated, the carbocyclic ring is usually a 3 to 10 membered ring.
  • Non-limiting examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl (bicyclo[2.2.1]heptyl), bicyclo[2.2.2]octyl, adamantane Alkyl and so on.
  • "cycloalkyl” includes "C 3-8 cycloalkyl” and "C 3-7 cycloalkyl”.
  • C 3-8 cycloalkyl should be understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring, which consists of 3 to 8 carbon atoms forming a 3 to 8 membered carbocyclic ring, such as cyclopropyl, cyclobutyl, Cyclopentyl, cyclohexyl, etc.
  • C 3-8 cycloalkyl preferably includes “C 3-7 cycloalkyl”.
  • C 3-7 cycloalkyl should be understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring, which consists of 3 to 7 carbon atoms forming a 3 to 7 membered carbocyclic ring.
  • heterocycloalkyl refers to a cyclic group that is fully saturated and may exist as a monocyclic ring, a fused ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the heterocyclic ring usually contains 1 to 5 heteroatoms (preferably 1 or 2 or 3 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen as ring atoms, and the total number of ring atoms is usually 3. , 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14.
  • heterocycloalkyl in the present application includes but is not limited to 3-membered heterocycloalkanes, such as oxiranyl, sulfiethane, cycloazaethyl, and 4-membered heterocycloalkyls, such as azetidinyl, oxa Butyl, thiabutanyl, 5-membered heterocycloalkyl such as tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidine Group, tetrahydropyrazolyl, 6-membered heterocycloalkyl such as piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1,4-thiooxanyl, 1, 4-d
  • 4-14 membered heterocycloalkyl refers to fully saturated, containing 1-5, preferably containing 1-3 heteroatoms independently selected from N, O and/or S as ring atoms, and the number of ring atoms Respectively 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 single ring, parallel ring, bridge ring or spiro ring.
  • 5-12 membered heterocycloalkyl refers to fully saturated, containing 1-5, preferably containing 1-3 heteroatoms independently selected from N, O and/or S as ring atoms, and the number of ring atoms Respectively 5, 6, 7, 8, 9, 10, 11 or 12 monocyclic rings, parallel rings, bridged rings or spiro rings.
  • the "4-14 membered heterocycloalkyl group” and "5-12 membered heterocycloalkyl group” of the present application preferably include "6-11 membered heterocycloalkyl group", "3-8 membered heterocycloalkyl group” or " 3-7 membered heterocycloalkyl” and so on.
  • heteroaryl refers to a monocyclic or condensed polycyclic ring system, which contains at least one ring atom selected from N, O, and S, the remaining ring atoms are C, and have at least one aromatic ring.
  • Preferred heteroaryl groups have a single 4 to 8 membered ring, especially a single 5 to 8 membered ring, or multiple fused rings containing 6 to 14, especially 6 to 10 ring atoms.
  • heteroaryl groups include, but are not limited to, pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolinyl , Tetrazolyl, triazolyl, triazinyl, benzofuranyl, benzothienyl, indolyl, isoindolyl, etc.
  • heteroaryl includes "5-6 membered heteroaryl”.
  • 5-6 membered heteroaryl refers to an aromatic ring system having 5 or 6 ring atoms, and it contains 1-3, preferably 1-2 heteroatoms independently selected from N, O and S.
  • “5-6 membered heteroaryl” includes, but is not limited to, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxazolyl, Azolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, 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 specific disease, condition or disorder, (ii) reduction, amelioration or elimination of one or more symptoms of a specific disease, condition or disorder, or (iii) prevention or delay
  • the amount of the compound of the present application 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.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms that are within the scope of reliable medical judgment and are suitable for use in contact with human and animal tissues, but not Many toxicity, irritation, allergic reactions or other problems or complications are commensurate with a reasonable benefit/risk ratio.
  • salts for example, metal salts, ammonium salts, salts with organic bases, salts with inorganic acids, salts with organic acids, salts with basic or acidic amino acids, etc. can be mentioned. .
  • pharmaceutical composition refers to a mixture of one or more of the compounds of the application or their salts and pharmaceutically acceptable excipients.
  • the purpose of the pharmaceutical composition is to facilitate the administration of the compound of the present application to the organism.
  • 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 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.
  • 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 may be partial or complete, and partial deuterium substitution refers to the substitution of at least one hydrogen by deuterium.
  • the compound of the present application 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, and geometric isomers are all included in the scope of the present invention.
  • the schematic diagram of the racemate or enantiomerically pure compound herein is from Maehr, J. Chem. Ed. 1985, 62: 114-120. Unless otherwise specified, use wedge keys and virtual wedge keys Represents the absolute configuration of a three-dimensional center. Additional asymmetric carbon atoms, asymmetric sulfur atoms, asymmetric nitrogen atoms, or asymmetric phosphorus atoms may be present in substituents such as alkyl groups. All these isomers and their mixtures are included in the scope of the present invention.
  • the compounds of the present application containing asymmetric atoms or asymmetric double bonds can be isolated in an optically pure form or in 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.
  • tautomers refers to structural isomers of different energies that can interconvert via a low energy barrier.
  • proton tautomers also called proton transfer tautomers
  • proton migration such as keto-enol and imine-enamine isomerization.
  • a specific example of a proton tautomer is the imidazole moiety, in which protons can migrate between two ring nitrogens.
  • Valence tautomers include interconversion through the recombination of some bonding electrons.
  • 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 compounds of the application or their pharmaceutically acceptable salts or pharmaceutical compositions 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 methods, dissolution methods, granulation methods, emulsification methods, freeze-drying methods, and the like.
  • 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 200 mg/kg body weight, preferably 0.05 to 40 mg/kg body weight, more preferably 0.1 to 20 mg/kg body weight, in single or divided doses form.
  • the compound of the present application 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 replacement manners, preferred implementation manners include but are not limited to the embodiments of the present application.
  • the compound of general formula (I) of the present application can be prepared by those skilled in the art of organic synthesis through Route 1:
  • the compound of formula (a) and the compound of formula (b) react in the presence of sodium carbonate and [1,1'-bis(diphenylphosphine)ferrocene]dichloropalladium dichloromethane complex to produce formula (c)
  • the compound, the compound of formula (c) is catalytically hydrogenated to produce the compound of formula (d), and the compound of formula (d) reacts with the compound of formula (e) to produce the compound of formula (I).
  • 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.
  • N,N-dimethylpiperidin-4-amine (1-5) 500mg, 2.71mmol
  • 1-bromo-2-fluoro-4-methoxy-5-nitrobenzene 604mg, 2.44mmol
  • acetonitrile 15mL
  • potassium carbonate 5.3g, 8.13mmol
  • the target compound (3-((5-bromo-2-((4-(4-(dimethylamino)piperidin-1-yl)-2-methoxy-5-(1-methyl-1H -Pyrazol-4-yl)phenyl)amino)pyrimidin-4-yl)amino)quinolin-4-yl)dimethylphosphine oxide (compound 1) trifluoroacetate (29.8mg, yield: 14.1 %).
  • the residue was purified by high performance liquid chromatography (eluent gradient: the same as in Example 1).
  • the target compound (6-((5-bromo-2-((2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)-6-morpholinylpyridin-3-yl) )Amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 4) trifluoroacetate (25 mg, yield: 9.6%).
  • reaction system was replaced with argon three times and then reacted at 90°C for 5 hours. After the reaction was completed, the reaction solution was cooled to room temperature and filtered, the filtrate was extracted with ethyl acetate (80 mL ⁇ 3), the combined organic phase was washed with saturated brine (60 mL), and then dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure.
  • the system was replaced with argon three times and reacted at 105°C for 5 hours until the reaction was complete.
  • the reaction solution was cooled to room temperature and filtered, the filtrate was extracted with ethyl acetate (30 mL ⁇ 3), and the combined organic phase was washed with saturated brine (20 mL), then dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure.
  • Example 7 (3-((5-Bromo-2-((5-(1-(2-hydroxyethyl)-1H-pyrazol-4-yl)-2-methoxy-4-morpholine (Phenyl)amino)pyrimidin-4-yl)amino)quinolin-4-yl)dimethylphosphine oxide (compound 7) preparation of trifluoroacetate
  • the reaction system was replaced with argon three times and then reacted at 100°C for 16 hours. After the reaction was completed, the reaction solution was cooled to room temperature, the compound was filtered, the filtrate was extracted with ethyl acetate (30mL X 3), the combined organic phases were washed with saturated brine (20mL), then dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure .
  • Example 8 (6-((5-Bromo-2-((6-(4-(dimethylamino)piperidin-1-yl)-2-methoxy-5-(1-methyl-1H -Pyrazol-4-yl)pyridin-3-yl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 8) preparation of trifluoroacetate
  • reaction solution was heated to 120°C and stirred for 16 hours. After the reaction was completed, the reaction solution was cooled to room temperature and diluted with water (80 mL), and the mixture was extracted with ethyl acetate (100 mL x 3). The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure.
  • the residue was purified by high performance liquid chromatography (eluent gradient: the same as in Example 1) to obtain the target product 1-(6-methoxy-3-(1-methyl-1H-pyrazol-4-yl) )-5-nitropyridin-2-yl)-N,N-dimethylpiperidin-4-amine (8-3) (90mg, yield: 20%) and 1-(6-methoxy- 5-(1-methyl-1H-pyrazol-4-yl)-3-nitropyridin-2-yl)-N,N-dimethylpiperidin-4-amine (8-3A) (25mg, Yield: 20%).
  • the reaction solution was cooled to room temperature, the mixture was filtered, the filtrate was extracted with ethyl acetate (30mL X 3), the organic phases were combined and washed with saturated brine (20mL), then dried over anhydrous sodium sulfate and filtered, and the filtrate was decompressed concentrate.
  • the reaction solution was heated to 105°C and stirred for 16 hours. After the completion of the reaction was detected by LCMS, the reaction solution was cooled to room temperature. The reaction solution was added to water (60mL), the mixture was diluted with ethyl acetate (70mL), filtered through a pad of celite, the filtrate was extracted with ethyl acetate (70mL X 3), the organic phase was washed with saturated brine and then washed with water. The aqueous sodium sulfate was dried and filtered, and the filtrate was concentrated under reduced pressure.
  • Example 13 (6-((5-Bromo-2-((2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)-6-(4-(4-methyl Piperazine-1-yl)piperidin-1-yl)pyridin-3-yl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 13) trifluoro Preparation of acetate
  • reaction solution was cooled to room temperature, filtered, and the mother liquor was concentrated to obtain the target product 2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)-6-(4-(4-methylpiperazine) -1-yl)piperidin-1-yl)pyridin-3-amine (13-4) (80 mg, yield: 98.9%).
  • the crude product was directly used in the next reaction.
  • Example 14 (6-((5-Bromo-2-((2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)-4-(4-(4-methyl Preparation of phenylpiperazin-1-yl)piperidin-1-yl)phenyl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 14)
  • Example 15 ((6-((5-Bromo-2-((4-(3-fluoro-4-morpholinopiperidin-1-yl)-2-methoxy-5-(1-methyl Preparation of phenyl-1H-pyrazol-4-yl)phenyl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 83) trifluoroacetate
  • reaction solution was filtered, concentrated, and separated by column chromatography to obtain the target product 4-(1-(2-bromo-5-methoxy-4-nitrophenyl)-3-fluoropiperidin-4-yl) Morinoline (83-4) (200.00 mg, yield: 33.8%).
  • reaction solution was filtered through Celite to remove palladium-carbon, and concentrated to obtain the target product 4-(3-fluoro-4-morpholinepiperidin-1-yl)-2-methoxy-5-(1-methyl- 1H-pyrazol-4-yl)aniline (83-6) (150.00 mg, yield: 89.8%).
  • the reaction solution was separated and purified by high performance liquid chromatography (eluent gradient reference example 1) to obtain the target product ((6-((5-bromo-2-((4-(3-fluoro-4-morpholine) Piperidin-1-yl)-2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)phenyl)amino)pyrimidin-4-yl)amino)quinoxaline-5 -Yl)dimethylphosphine oxide (Compound 83) trifluoroacetate (60.00 mg, yield: 20.3%).
  • the morpholine was replaced with cis 2,6-dimethylmorpholine, and (3-((2,5-dichloropyrimidin-4-yl)amino)quinoline-4- Yl) dimethyl phosphine oxide (2-1) is replaced with (6-((5-bromo-2-chloropyrimidin-4-yl) amino) quinoxalin-5-yl) dimethyl phosphine oxide (4- 9), the target product (6-((5-bromo-2-((4-((2S,6R)-2,6-dimethylmorpholine)-2-methoxy-5-(1- Methyl-1H-pyrazol-4-yl)phenyl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 85).
  • Example 2 replace (3-((2,5-dichloropyrimidin-4-yl)amino)quinolin-4-yl)dimethylphosphine oxide (2-1) with (6-( (5-Bromo-2-chloropyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (4-9), to obtain the target product 1-(4-(1-(4-( (5-Bromo-4-((5-(dimethylphosphoryl)quinoxalin-6-yl)amino)pyrimidin-2-yl)amino)-5-methoxy-2-(1-methyl -1H-pyrazol-4-yl)phenyl)piperidin-4-yl)piperazin-1-yl)ethane-1-one (Compound 90).
  • the 1-methyl-4-(piperidin-4-yl)piperazine was replaced with cis 2,6-dimethylmorpholine to obtain the target compound (6-((5- Bromo-2-((6-((2S,6R)-2,6-dimethylmorpholine)-2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)pyridine -3-yl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 92).
  • Example 13 replace 1-methyl-4-(piperidin-4-yl)piperazine with morpholine, and replace 1-methyl-4-pyrazole boronic acid pinacol ester with 4- Pyrazolboronic acid pinacol ester to obtain the target compound (6-((5-bromo-2-((2-methoxy-6-morpholine-5-(1H-pyrazol-4-yl)pyridine-3 -Yl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 94) trifluoroacetate.
  • the 1-methyl-4-(piperidin-4-yl)piperazine was replaced with 4-(4-piperidinyl)morpholine to obtain the target compound (6-((5- Bromo-2-((2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)-6-(4-morpholinopiperidin-1-yl)pyridin-3-yl )Amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (compound 95).
  • the compound (6-aminoquinolin-5-yl) dimethyl phosphine oxide (100-3) (1.0 g, yellow solid) was obtained by elution.
  • reaction mixture was concentrated under reduced pressure, the residue was added to N,N-dimethylformamide (5 mL), filtered, and purified by preparative high performance liquid chromatography (eluent gradient reference example 1) to obtain compound (6 -((5-Bromo-2-((2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)-4-(4-morpholinopiperidin-1-yl) Phenyl) amino) pyrimidin-4-yl) amino) quinoxalin-5-yl) dimethyl phosphine oxide trifluoroacetate.
  • Example 15 replace tert-butyl 3-fluoro-4-oxopiperidine-1-carboxylate with N-tert-butoxycarbonyl-4-piperidone, and replace morpholine with homomorpholine .
  • the target compound (6-((2-((4-(4-(1,4-oxazepan-4-yl)piperidin-1-yl)-2-methoxy-5-(1 -Methyl-1H-pyrazol-4-yl)phenyl)amino)-5-bromopyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide (Compound 24).
  • the tert-butyl 3-fluoro-4-oxopiperidine-1-carboxylate was replaced by N-tert-butoxycarbonyl-4-piperidone, and the morpholine was replaced by 3,3 -Difluoropyrrolidine to obtain the target compound (6-((5-Bromo-2-((4-(4-(3,3-difluoropyrrolidin-1-yl)piperidin-1-yl)))- 2-Methoxy-5-(1-methyl-1H-pyrazol-4-yl)phenyl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethylphosphine oxide ( Compound 102).
  • reaction solution was spin-dried directly, and the intermediate (6-((5-chloro-2-((2-methoxy-5-(1- Methyl-1H-pyrazol-4-yl)-6-(piperazin-1-yl)pyridin-3-yl)amino)pyrimidin-4-yl)amino)quinoxalin-5-yl)dimethyl Phosphine oxide (109-3) (150 mg, yield: 50%).
  • reaction solution was spin-dried under reduced pressure, DCM (10 mL) was added and spin-dried again under reduced pressure to obtain the intermediate (6-((2-((6-(4-(azetidin-3-yl)piperazine-1- Yl)-2-methoxy-5-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)amino)-5-bromopyrimidin-4-yl)amino)quinoxaline-
  • reaction was stirred at room temperature for 5 hours, then sodium triacetoxyborohydride (118 mg, 0.555 mmol) was added, and the reaction was carried out at room temperature for 11 hours.
  • the reaction mixture was adjusted to pH ⁇ 7 with aqueous sodium hydroxide solution, extracted with dichloromethane (20mL X 3), the organic phase was spin-dried, and the residue was added to DMF (5mL).
  • the present invention also synthesized the following compounds, the characterization data of which is:
  • the compounds in the following test examples are all prepared according to the method of the above-mentioned examples of the present application.
  • the compounds of the test examples can be in the form of free base or its trifluoroacetate salt, and are determined according to the specific forms of the products prepared in the above examples.
  • Test Example 1 Enzymatic EGFR inhibition experiment
  • HTRF KinEASE-TK kit was purchased from CisBio (France).
  • the 384-well assay plate was purchased from Greiner bio-one (Germany).
  • the 384-well source plate was purchased from LABCYTE (USA).
  • MgCl 2 , MnCl 2 , DTT, Triton X-100, HEPES, and BSA were purchased from Sigma (USA).
  • EGFR WT-WT EGFR WT-del19/T790M/C797S
  • EGFR WT-L858R/T790M/C797S EGFR WT-del19/T790M
  • EGFR WT-L858R/T790M refer to Nature.2016; 534(7605):129-132 Methods of expression and purification.
  • Multilabel detection analyzer Envision Multilabel Reader (PerkinElmer, USA)
  • kinase buffer solution (1x Enzyme buffer, 1mM MnCl2, 1mM MgCl2, 1mM DTT, 0.01% BSA).
  • the final detection concentration of the compound is 10 ⁇ M, and it is formulated to a 100-fold concentration using DMSO, that is, 1 mM.
  • DMSO dimethyl methoxysulfoxide
  • Precision PRC384U automatic micropipette
  • kinase reaction Use kinase buffer to prepare 2X enzyme reaction solution (WT-WT: 3nM, WT-d19/TM/CS: 1nM, WT-LR/TM/CS: 0.1nM, WT-LR/TM: 0.5nM, WT-d19/ TM: 1nM) Transfer 5 ⁇ l of enzyme reaction solution to the reaction wells of a 384-well plate, and add 5 ⁇ l of kinase buffer to the negative control wells. Shake and mix at 450 rpm and incubate at room temperature for 10 minutes.
  • reaction stop solution (Streptavidin-XL665, TK Antibody-Cryptate) to the 384-well reaction plate, centrifuge at 1000 rpm for 1 minute, leave it at room temperature for 60 minutes, and read the plate with Envision.
  • Enzymatic inhibition activity of the compounds of the present invention 50 is shown below IC (del19 abbreviated as d19, T790M abbreviated as TM, L858R abbreviated to LR, C797S abbreviated as CS).
  • RPMI1640 was purchased from Gibco (USA).
  • FBS was purchased from Excell (China).
  • IL-3 Recombinant Mouse Protein was purchased from Gibco (USA).
  • BaF3 cells were purchased from Riken (Japan).
  • BaF3/EGFR-WT, BaF3/EGFR-Del19/T790M/C797S cells were constructed by Sino-American Crown Biotechnology (Taicang) Co., Ltd.
  • BaF3 medium RPMI1640+10%FBS+8ng/ml IL-3 Recombinant Mouse Protein.
  • BaF3/EGFR-WT, BaF3/EGFR-Del19/T790M/C797S medium RPMI1640+10% FBS.
  • Test Example 3 PC9 EGFR-Del19/T790M/C797S and HEK293T cell proliferation experiment
  • RPMI1640 was purchased from Gibco (USA).
  • DMEM was purchased from Gibco (USA).
  • FBS was purchased from Gibco (USA).
  • Puromycin was purchased from Invitrogen (USA).
  • Pancreatin was purchased from Invitrogen (USA).
  • DMSO was purchased from Sigma (USA).
  • Cell Counting Kit-8 (CCK-8) cell proliferation toxicity detection kit was purchased from Dongren Chemical Technology (Shanghai) Co., Ltd.
  • PC9 EGFR-Del19/T790M/C797S cells were constructed by Sino-American Crown Biotechnology (Taicang) Co., Ltd.
  • HEK293T cells were purchased from ATCC (USA).
  • PC9 EGFR-Del19/T790M/C797S medium RPMI1640+10% FBS+0.5ug/mL puromycin.
  • HEK293T medium DMEM+10% FBS.
  • the cells are cultured for 2-3 days to reach 80-90% confluence. Digest the cells with trypsin, resuspend by centrifugation and count, adjust the cell concentration to 1000 cells per well (HEK293T) and 3000 cells per well (PC9EGFR-Del19/T790M/C797S), 90 ⁇ l volume, 5% CO 2 , and incubate overnight at 37°C .
  • Inhibition% (OD cell control –OD sample )/(OD cell control –OD media control )*100
  • CD-1 mice were purchased from Sibefu (Beijing) Biotechnology Co., Ltd.
  • DMSO, MC methyl cellulose
  • Tween80 purchased from Sigma (USA).
  • Acetonitrile was purchased from Merck (USA).
  • HP- ⁇ -CD was purchased from Japan Food and Chemical Co., Ltd.
  • mice Six female CD-1 mice (20-30g, 4-6 weeks, three in each group) were randomly divided into two groups, with three in each group. The first group was given compound 82 by tail vein injection at a dose of 1 mg/kg, and the solvent was 5% DMSO in 10% HP- ⁇ -CD in water, and the second group was given orally with compound 82 at a dose of 5 mg/kg and the vehicle was 0.5% MC/0.25 %Tween80in water. Feed and water normally before the animal experiment. Mice in each group were subjected to intravenous blood sampling at 0.083 (intravenous injection group only), 0.25, 0.5, 1, 2, 4, 6, 8 and 24 h before and after administration. The collected whole blood samples were placed in a K2EDTA anticoagulation tube, and after centrifugation for 5 minutes (4000 rpm, 4°C), plasma was taken for testing.
  • intravenous blood sampling at 0.083 (intravenous injection group only), 0.25, 0.5, 1, 2, 4, 6, 8 and 24 h before and after
  • the PK test results are as follows:

Abstract

公开了第四代(T790M/C797S突变)EGFR激酶抑制剂及其医药用途,具体公开了式(I)所示化合物或其药学上可接受的盐,所述化合物在EGFR Del19/T790M/C797S和L858R/T790M/C797S异常突变引起的疾病上有着较好的疗效。

Description

作为EGFR激酶抑制剂的多芳基化合物 技术领域
本申请涉及作为EGFR激酶抑制剂的多芳基化合物、其制备方法、含有该化合物的药物组合物、以及其在治疗EFGR激酶相关疾病中的用途。
背景技术
EGFR(Epidermal Growth Factor Receptor)是上皮生长因子(EGF)细胞增殖和信号传导的受体。EGFR属于ErbB受体家族的一种,该家族包括EGFR(ErbB-1)、HER2/c-neu(ErbB-2)、HER3(ErbB-3)和HER4(ErbB-4)。EGFR也被称作HER1、ErbB1。EGFR广泛分布于哺乳动物上皮细胞、成纤维细胞、胶质细胞、角质细胞等细胞表面,EGFR信号通路对细胞的生长、增殖和分化等生理过程发挥重要的作用。EGFR分为三区:胞外配体结合区,跨膜区和胞内激酶区。当EGFR受到相应配体结合后,会诱导其形成同型或者异型二聚体,从而激活胞内的酪氨酸激酶通路,使其自身磷酸化,从而引导下游的磷酸化,包括MAPK、Akt和JNK通路,诱导细胞增殖。
EGFR酪氨酸激酶抑制剂(Tyrosine Kinase Inhibitor,TKI)是通过阻断内源性的ATP结合到胞内的激酶区域,从而抑制受体磷酸化及其下游信号转导分子的活化,阻断肿瘤细胞的增殖。尽管EGFR靶向治疗已经成功进入临床阶段,并已有药物上市,但是EGFR的基因突变导致药物出现耐药。其突变主要发生在18-21号外显子,其中19号外显子缺失和21号外显子的L858R的点突变是最常见的突变亚型,占所有突变类型的90%。随着药物的开发使用,大部分耐药出现在激酶的gatekeeper区域T790M的二次突变。最近几年开发的三代不可逆抑制剂对T790M突变的抑制活性很好,但是也不可避免地出现了C797S突变,而且是明星药物AZD9291的主要耐药机制(40%左右)。2018年AZD9291已经进入一线治疗,针对C797S突变,人们急需开发新型的、更加安全有效的EGFR TKI。
发明内容
本申请涉及式(I)化合物或其药学上可接受的盐,
Figure PCTCN2020132188-appb-000001
其中,
R 1选自苯基和5-6元杂芳基,所述苯基和5-6元杂芳基任选被R 7取代;
或R 1
Figure PCTCN2020132188-appb-000002
其中R 11、R 12独立地选自C 1-4烷基,所述C 1-4烷基任选地被羟基或C 1-4烷氧基取代;
R 2为4-14元杂环烷基,所述4-14元杂环烷基任选被R 8取代;
U、V、W独立地选自CR 6和N;
环A选自苯基和5-6元杂芳基;
R 3选自H、卤素、氰基、C 1-4烷基和C 1-4烷氧基;
R 4、R 5独立地选自C 1-4烷基和C 1-4烷氧基;
R 6独立地选自H、C 1-4烷基、C 1-4烷氧基和卤素;
R 7独立地选自卤素、羟基、氨基、C 1-4烷基和C 1-4烷氧基,所述C 1-4烷基和C 1-4烷氧基任选地被羟基取代;
R 8独立地选自卤素、OH、C 1-6烷基、C 1-6烷氧基、氨基、C 3-8环烷基和3-8元杂环烷基,所述氨基、C 3-8环烷基和3-8元杂环烷基任选地被R 9取代;
R 9独立地选自卤素、C 1-4烷基、C 1-4烷氧基、C 1-4酰基和氧代。
在一些实施方案中,R 1选自5-6元杂芳基,所述5-6元杂芳基任选被R 7取代。
在一些实施方案中,R 1选自5元杂芳基,所述5元杂芳基任选被R 7取代。
在一些实施方案中,R 1选自苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、四唑基和三唑基,所述苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、四唑基和三唑基任选被R 7取代。
在一些实施方案中,R 1选自吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、四唑基和三唑基,所述吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、四唑基和三唑基任选被R 7取代。
在一些实施方案中,R 1选自苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基和三唑基,所述苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基和三唑基任选被R 7取代。
在一些实施方案中,R 1选自苯基、吡啶基、吡唑基和咪唑基,所述苯基、吡啶基、吡唑基和咪唑基任选被R 7取代。
在一些实施方案中,R 1选自吡咯基、呋喃基、噻吩基、咪唑基、吡唑基和三唑基,所述吡咯基、呋喃基、噻吩基、咪唑基、吡唑基和三唑基任选被R 7取代。
在一些实施方案中,R 7独立地选自卤素、羟基和C 1-4烷基,所述C 1-4烷基任选地被羟基取代。
在一些实施方案中,R 7独立地选自C 1-4烷基,所述C 1-4烷基任选地被羟基取代。
在一些实施方案中,R 7独立地选自甲基和羟乙基。
在一些实施方案中,R 7独立地选自甲基。
在一些实施方案中,R 1选自苯基、
Figure PCTCN2020132188-appb-000003
Figure PCTCN2020132188-appb-000004
在一些实施方案中,R 1选自苯基、
Figure PCTCN2020132188-appb-000005
在一些实施方案中,R 1选自
Figure PCTCN2020132188-appb-000006
Figure PCTCN2020132188-appb-000007
在一些实施方案中,R 1
Figure PCTCN2020132188-appb-000008
其中R 11选自C 1-4烷基、R 12选自任选被羟基取代的C 1-4烷基。
在一些实施方案中,R 1
Figure PCTCN2020132188-appb-000009
其中R 11为甲基、R 12为甲基或羟乙基。
在一些实施方案中,R 2为5-12元杂环烷基,所述5-12元杂环烷基任选被R 8取代。
在一些实施方案中,R 2为6-11元杂环烷基,所述6-11元杂环烷基任选被R 8取代。
在一些实施方案中,R 2选自哌啶基、四氢吡喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基、氮杂环庚烷基、氧杂环庚烷基、硫杂环庚烷基、3,9-二氮杂螺[5.5]十一烷基、2,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000010
Figure PCTCN2020132188-appb-000011
所述哌啶基、四氢吡喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基、氮杂环庚烷基、氧杂环庚烷基、硫杂环庚烷基、3,9-二氮杂螺[5.5]十一烷基、2,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000012
任选被R 8取代。
在一些实施方案中,R 2选自哌啶基、四氢吡喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基、氮杂环庚烷基、氧杂环庚烷基、硫杂环庚烷基、3,9-二氮杂螺[5.5]十一烷基、2,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000013
所述哌啶基、四氢吡喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基、氮杂环庚烷基、氧杂环庚烷基、硫杂环庚烷基、3,9-二氮杂螺[5.5]十一烷基、2,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000014
任选被R 8取代。
在一些实施方案中,R 2选自哌啶基、哌嗪基、吗啉基、3,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000015
Figure PCTCN2020132188-appb-000016
所述哌啶基、哌嗪基、吗啉基、3,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000017
Figure PCTCN2020132188-appb-000018
任选被R 8取代。
在一些实施方案中,R 2选自哌啶基、哌嗪基、吗啉基、3,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000019
Figure PCTCN2020132188-appb-000020
所述哌啶基、哌嗪基、吗啉基、3,9-二氮杂螺[5.5]十一烷基、
Figure PCTCN2020132188-appb-000021
Figure PCTCN2020132188-appb-000022
任选被R 8取代。
在一些实施方案中,R 2选自哌啶基、哌嗪基、吗啉基、
Figure PCTCN2020132188-appb-000023
所述哌啶基、哌嗪基、吗啉基、
Figure PCTCN2020132188-appb-000024
任选被R 8取代。
在一些实施方案中,R 2选自哌啶基、吗啉基和3,9-二氮杂螺[5.5]十一烷基,所述哌啶基、吗啉基和3,9-二氮杂螺[5.5]十一烷基任选被R 8取代。
在一些实施方案中,R 8独立地选自卤素、C 1-6烷基、C 1-6烷氧基、氨基、C 3-8环烷基和3-8元杂环烷基,所述氨基、C 3-8环烷基和3-8元杂环烷基任选地被R 9取代。
在一些实施方案中,R 8独立地选自卤素、OH、C 1-4烷基、氨基、C 3-7环烷基和3-7元杂环烷基,所述氨基、C 3-7环烷基和3-7元杂环烷基任选地被R 9取代。
在一些实施方案中,R 8独立地选自卤素、OH、氨基和3-7元杂环烷基,所述氨基和3-7元杂环烷基任选地被R 9取代。
在一些实施方案中,R 8独立地选自C 1-4烷基、氨基、C 3-7环烷基和3-7元杂环烷基,所述氨基、C 3-7环烷基和3-7元杂环烷基任选地被R 9取代。
在一些实施方案中,R 8独立地选自F、OH、甲基、氨基、哌嗪基、1,3-噁嗪烷基、1,4-二 氮杂环庚烷基、1,3-氧杂氮杂环庚烷基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基、氮杂环丁烷基,所述氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基、氮杂环丁烷基任选地被R 9取代。
在一些实施方案中,R 8独立地选自F、OH、甲基、氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基,所述氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基任选地被R 9取代。
在一些实施方案中,R 8独立地选自F、OH、氨基、哌嗪基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基,所述氨基、哌嗪基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基任选地被R 9取代。
在一些实施方案中,R 8独立地选自甲基、氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基和1,4-氧杂氮杂环庚烷基,所述氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基和1,4-氧杂氮杂环庚烷基任选地被R 9取代。
在一些实施方案中,R 9独立地选自卤素、C 1-4烷基、C 1-4酰基、C 1-4烷氧基和氧代。
在一些实施方案中,R 9独立地选自卤素、C 1-4烷基和C 1-4酰基。
在一些实施方案中,R 9独立地选自卤素、C 1-4烷基、C 1-4烷氧基和氧代。
在一些实施方案中,R 9独立地选自F、甲基、乙酰基和氧代。
在一些实施方案中,R 9独立地选自F、甲基和乙酰基。
在一些实施方案中,R 9独立地选自甲基和氧代。
在一些实施方案中,R 2选自
Figure PCTCN2020132188-appb-000025
Figure PCTCN2020132188-appb-000026
在一些实施方案中,R 2选自
Figure PCTCN2020132188-appb-000027
Figure PCTCN2020132188-appb-000028
在一些实施方案中,R 2选自
Figure PCTCN2020132188-appb-000029
Figure PCTCN2020132188-appb-000030
在一些实施方案中,R 2选自
Figure PCTCN2020132188-appb-000031
Figure PCTCN2020132188-appb-000032
在一些实施方案中,U为CR 6
在一些实施方案中,R 6独立地选自C 1-4烷氧基。
在一些实施方案中,R 6为甲氧基。
在一些实施方案中,V选自CH和N。
在一些实施方案中,W选自CH和N。
在一些实施方案中,环A选自苯基、吡嗪基和吡啶基。
在一些实施方案中,环A选自苯基和吡嗪基。
在一些实施方案中,
Figure PCTCN2020132188-appb-000033
选自
Figure PCTCN2020132188-appb-000034
在一些实施方案中,
Figure PCTCN2020132188-appb-000035
选自
Figure PCTCN2020132188-appb-000036
在一些实施方案中,
Figure PCTCN2020132188-appb-000037
选自
Figure PCTCN2020132188-appb-000038
在一些实施方案中,R 3选自卤素和C 1-4烷基。
在一些实施方案中,R 3选自Cl和Br。
在一些实施方案中,R 4、R 5独立地选自C 1-3烷基。
在一些实施方案中,R 4、R 5为甲基。
在一些实施方案中,本申请的式(I)化合物选自式(II)化合物,
Figure PCTCN2020132188-appb-000039
其中R 1、R 2、R 3、R 6和V如上定义。
在一些实施方案中,本申请的式(I)化合物选自式(III)化合物,
Figure PCTCN2020132188-appb-000040
其中R 1、R 2、R 3、R 6和V如上定义。
在一些实施方案中,本申请的式(I)化合物选自以下化合物:
Figure PCTCN2020132188-appb-000041
Figure PCTCN2020132188-appb-000042
Figure PCTCN2020132188-appb-000043
Figure PCTCN2020132188-appb-000044
Figure PCTCN2020132188-appb-000045
Figure PCTCN2020132188-appb-000046
另一方面,本申请涉及药物组合物,其包含本申请的式(I)化合物或其药学上可接受的盐和药学上可接受的辅料。
另一方面,本申请涉及治疗哺乳动物由EGFR介导的疾病的方法,包括对需要该治疗的哺乳动物,优选人类,给予治疗有效量的式(I)化合物或其药学上可接受的盐、或其药物组合物。
另一方面,本申请涉及式(Ⅰ)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EGFR介导的疾病的药物中的用途。
另一方面,本申请涉及式(Ⅰ)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EGFR介导的疾病中的用途。
另一方面,本申请涉及预防或者治疗EGFR介导的疾病的式(Ⅰ)化合物或其药学上可接受的盐、或其药物组合物。
定义
除非另有说明,本申请中所用的下列术语具有下列含义。一个特定的术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照本领域普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
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。
本文中的C m-n,是指该部分具有给定范围中的整数个碳原子。例如“C 1-6”是指该基团可具有1个碳原子、2个碳原子、3个碳原子、4个碳原子、5个碳原子或6个碳原子。
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。例如,如果一个基团被2个R所取代,则每个R都有独立的选项。
术语“卤”或“卤素”是指氟、氯、溴和碘。
术语“羟基”指-OH基团。
术语“氰基”指-CN基团。
术语“氨基”指-NH 2基团。
术语“烷基”是指通式为C nH 2n+1的烃基。该烷基可以是直链或支链的。例如,术语“C 1- 6烷基”指含有1至6个碳原子的烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、1-甲基丁基、2-甲基丁基、3-甲基丁基、新戊基、己基、2-甲基戊基等)。类似地,烷氧基、烷基氨基、二烷基氨基、烷基磺酰基和烷硫基的烷基部分(即烷基)具有上述相同定义。
术语“烷氧基”指-O-烷基。术语“C 1-4烷氧基”可以是甲氧基、乙氧基、正丙基氧基、异丙基氧基、正丁基氧基、异丁基氧基、仲丁基氧基或叔丁基氧基等。
术语“酰基”表示式-C(=O)R的基团,其中R为氢或如本文所定义的烷基;术语“C 1-4酰基”是指包含1-4个碳原子的-C(=O)R,如甲酰基、乙酰基、丙酰基、丁酰基、异丁酰基。
术语“环烷基”指完全饱和的并且可以以呈单环、桥环或螺环存在的碳环。除非另有指示,该碳环通常为3至10元环。环烷基非限制性实例包括但不限于环丙基、环丁基、环戊基、环己基、降冰片基(双环[2.2.1]庚基)、双环[2.2.2]辛基、金刚烷基等。优选地,“环烷基”包含了“C 3-8环烷基”和“C 3-7环烷基”。
术语“C 3-8环烷基”应理解为表示饱和的一价单环或双环烃环,其由3~8个碳原子形成3~8元碳环,如环丙基、环丁基、环戊基、环己基等。“C 3-8环烷基”优选地包含了“C 3-7环烷基”。“C 3-7环烷基”应理解为表示饱和的一价单环或双环烃环,其由3~7个碳原子形成3~7元碳环。
术语“杂环烷基”是指完全饱和的并且可以以单环、并环、桥环或螺环存在的环状基团。除非另有指示,该杂环通常含有1至5个独立地选自硫、氧和/或氮的杂原子(优选1或2或3个杂原子)作为环原子,且环原子总数通常为3、4、5、6、7、8、9、10、11、12、13或14。
本申请的“杂环烷基”包括但不限于3元杂环烷,如环氧乙烷基、环硫乙烷基、环氮乙烷基,4元杂环烷基,如吖丁啶基、噁丁环基、噻丁环基,5元杂环烷基如四氢呋喃基、四氢噻吩基、吡咯烷基、异噁唑烷基、噁唑烷基、异噻唑烷基、噻唑烷基、咪唑烷基、四氢吡唑基,6元杂环烷基如哌啶基、四氢吡喃基、四氢噻喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基,7元杂环烷基如氮杂环庚烷基、氧杂环庚烷基、硫杂环庚烷基。
术语“4-14元杂环烷基”是指完全饱和的,含有1-5个,优选含有1-3个独立选自N、O和/或S的杂原子作为环原子,且环原子数分别为4、5、6、7、8、9、10、11、12、13或14的单环、并环、桥环或螺环。
术语“5-12元杂环烷基”是指完全饱和的,含有1-5个,优选含有1-3个独立选自N、O和/或S的杂原子作为环原子,且环原子数分别为5、6、7、8、9、10、11或12单环、并环、桥环或螺环。
本申请“4-14元杂环烷基”和“5-12元杂环烷基”优选地包含了“6-11元杂环烷基”、“3-8元杂环烷基”或“3-7元杂环烷基”等。
术语“杂芳基”是指单环或稠合多环体系,其中含有至少一个选自N、O、S的环原子,其余环原子为C,并且具有至少一个芳香环。优选的杂芳基具有单个4至8元环,尤其是单个5至8元环,或包含6至14个,尤其是6至10个环原子的多个稠合环。杂芳基的非限制性实例包括但不限于吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、吡啶基、嘧啶基、吡嗪基、喹啉基、异喹啉基、四唑基、三唑基、三嗪基、苯并呋喃基、苯并噻吩基、吲哚基、异吲哚基等。优选地,“杂芳基”包含了“5-6元杂芳基”。
术语“5-6元杂芳基”指具有5或6个环原子的芳族环系,且其包含1-3个,优选1-2个独立选自N、O和S的杂原子。特别地,“5-6元杂芳基”包括但不限于噻吩基、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、吡啶基、哒嗪基、嘧啶基、吡嗪基或三嗪基等。
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申请化合物的用量。构成“治疗有效量”的本申请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“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
作为药学上可接受的盐,例如,可以提及金属盐、铵盐、与有机碱形成的盐、与无机酸形成的盐、与有机酸形成的盐、与碱性或者酸性氨基酸形成的盐等。
术语“药物组合物”是指一种或多种本申请的化合物或其盐与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本申请的化合物。
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)”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氘取代。
本申请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 PCTCN2020132188-appb-000047
表示一个立体中心的绝对构型。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或不对称双键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。
本申请的化合物和中间体还可以以不同的互变异构体形式存在,并且所有这样的形式包含于本申请的范围内。术语“互变异构体”是指可经由低能垒互变的不同能量的结构异构体。例如,质子互变异构体(也称为质子转移互变异构体)包括经由质子迁移的互变,如酮-烯醇及亚胺-烯胺异构化。质子互变异构体的具体实例是咪唑部分,其中质子可在两个环氮间迁移。价互变异构体包括通过一些成键电子的重组的互变。
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、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200mg/kg体重,优选为0.05到40mg/kg体重,更优选0.1到20mg/kg体重,以单独或分开剂量的形式。
本申请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申请的实施例。
本申请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申请的化学变化及其所需的试剂和物料。为了获得本申请的化合物,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。
在一些实施方案中,本申请通式(I)的化合物可以由有机合成领域技术人员通过路线1来制备:
Figure PCTCN2020132188-appb-000048
式(a)化合物与式(b)化合物在碳酸钠和[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物存在下反应生成式(c)化合物,式(c)化合物催化氢化生成式(d)化合物,式(d)化合物与式(e)化合物反应生成式(I)化合物。
为清楚起见,进一步用实施例来阐述本申请,但是实施例并非限制本申请的范围。本申请所使用的所有试剂是市售的,无需进一步纯化即可使用。
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。
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。
实施例1、(3-((5-溴-2-((4-(4-(二甲基氨基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物1)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000049
化合物1-2:
Figure PCTCN2020132188-appb-000050
室温下将喹啉-3-胺(1-1)(1g,6.9mmol)溶于N,N-二甲基甲酰胺(10mL)中,再在0℃下滴加碘代丁二酰亚胺(1.7g,7.6mmol)的N,N-二甲基甲酰胺(10mL)溶液,反应在室温下搅拌1小时。反应完全后将反应混合物倒入水(100mL)中,混合物用乙酸乙酯(50mL X3)萃取,有机相用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:石油醚:乙酸乙酯=3:1,Rf=0.5)纯化,得到目标产品4-碘喹啉-3-胺(1-2)(670mg,收率:36%)。
LCMS:Rt:1.505min;MS m/z(ESI):271.0[M+H] +
化合物1-3:
Figure PCTCN2020132188-appb-000051
室温下将4-碘喹啉-3-胺(1-2)(370mg,1.37mmol),二甲基氧化膦(214mg,2.74mmol),碳酸铯(1.34g,4.11mmol),三(二亚苄基丙酮)二钯(247mg,0.27mmol)和4,5-双二苯基膦-9,9-二甲基氧杂蒽(258mg,0.54mmol)加到N,N-二甲基甲酰胺(6mL)中,体系在氮气保护下加热至100℃下搅拌反应12小时。反应完全后将反应混合物过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=20:1,Rf=0.5)纯化,得到目标产品(3-氨基喹啉-4-基)二甲基氧化膦(1-3)(180mg,收率:60%)。
LCMS:Rt:1.235min;MS m/z(ESI):221.3[M+H] +
化合物1-4:
Figure PCTCN2020132188-appb-000052
室温下将(3-氨基喹啉-4-基)二甲基氧化膦(1-3)(180mg,0.82mmol),5-溴-2,4-二氯嘧啶(374mg,1.64mmol)和二异丙基乙基胺(1.06g,8.2mmol)溶于乙醇(5mL)中,体系在氮气保护下加热至100℃搅拌反应12小时。反应完全后将反应混合物减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=20:1,Rf=0.5)纯化,得到目标产品(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)(120mg,收率:36%)。
LCMS:Rt:1.620min;MS m/z(ESI):413.2[M+H] +
化合物1-6:
Figure PCTCN2020132188-appb-000053
室温下将N,N-二甲基哌啶-4-胺(1-5)(500mg,2.71mmol)和1-溴-2-氟-4-甲氧基-5-硝基苯(604mg,2.44mmol)溶于乙腈(15mL)中,然后加入碳酸钾(5.3g,8.13mmol)。反应液在氩气保护下加热至80℃搅拌反应4小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:甲醇:二氯甲烷=1:30~1:10)纯化,得到目标产物1-(2-溴-5-甲氧基-4-硝基苯基)-N,N-二甲基哌啶-4-胺(1-6)(560mg,收率:57.9%)。
LCMS:Rt:0.952min;MS m/z(ESI):358.1[M+H] +
化合物1-7:
Figure PCTCN2020132188-appb-000054
室温下将1-(2-溴-5-甲氧基-4-硝基苯基)-N,N-二甲基哌啶-4-胺(1-6)(300mg,0.84mmol),1-甲基-4-1H-吡唑-硼酸频那醇酯(524mg,2.52mmol),碳酸钠(268mg,2.52mmol)和[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(55.2mg,0.16mmol)溶于二氧六环(10mL)和水(1mL)的混合液中,氮气保护下将反应液升温至100℃搅拌反应12小时。反应完全后将反应混合物过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=20:1,Rf=0.5)纯化,得到目标产品1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-N,N-二甲基哌啶-4-胺(1-7)(250mg,收率:82.7%)。
LCMS:Rt:0.980min;MS m/z(ESI):360.5[M+H] +
化合物1-8:
Figure PCTCN2020132188-appb-000055
室温下将1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-N,N-二甲基哌啶-4-胺(1-7)(250mg,0.69mmol)溶于乙醇(15mL)中并加入钯碳催化剂(25mg),反应液在氢气氛下升温至50℃搅拌反应3h至反应完全。将反应液过滤,滤液减压浓缩,得到粗品目标产物1-(4-氨基-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)-N,N-二甲基哌啶-4-胺(1-8)(190mg,收率:83.7%)。
LCMS:Rt:0.382min;MS m/z(ESI):330.5[M+H] +
化合物1的三氟乙酸盐:
Figure PCTCN2020132188-appb-000056
室温下将1-(4-氨基-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)-N,N-二甲基哌啶-4-胺(1-8)(100mg,0.30mmol)和(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)(80mg,0.20mmol)溶于异丙醇(3mL)中,再加入三氟乙酸(228mg,2.0mmol);将反应液加热至100℃搅拌反应24小时。反应完全后将反应液冷却至室温,然后减压浓缩,残余物用高效液相制备色谱法纯化,洗脱剂梯度如下表所示:
Figure PCTCN2020132188-appb-000057
得到目标化合物(3-((5-溴-2-((4-(4-(二甲基氨基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物1)三氟乙酸盐(29.8mg,收率:14.1%)。
1H NMR(400MHz,CD 3OD)δ:9.46(d,J=4.8Hz,1H),8.23(s,1H),8.15-8.13(m,1H),7.82(brs,2H),7.68(d,J=6.0Hz,3.2Hz,2H),7.56(s,1H),7.38(brs,1H),6.76(s,1H),3.88(s,3H),3.51(s,3H),3.20-3.14(m,3H),2.87(s,6H)2.70-2.63(m,2H),2.19(s,3H),2.16(s,3H),2.11-2.07(m,2H),1.81-1.75(m,2H).
19F NMR(376.5MHz,CD 3OD):δ-76.89.
31P NMR(162.0MHz,CD 3OD):δ47.59.
LCMS:Rt:1.021min;MS m/z(ESI):704.1,706.1[M+H] +
实施例2、(3-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物2)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000058
化合物2-1:
Figure PCTCN2020132188-appb-000059
室温下将(3-氨基喹啉-4-基)二甲基氧化膦(1-3)(1.0g,4.52mmol)溶于乙醇(10mL)中,加入2,4,5-三氯嘧啶(1.65g,9.04mmol)和N,N-二异丙基乙胺(3.5g,27.1mmol),反应液在氩气保护下加热至120℃搅拌反应72小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:10~1:1)纯化,得到目标产物(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)(0.56g,收率:30%)。
LCMS:Rt:1.517min;MS m/z(ESI):367.2[M+H] +
化合物2-3:
Figure PCTCN2020132188-appb-000060
室温下将吗啉(2-2)(2.0g,22.98mmol)和1-溴-2-氟-4-甲氧基-5-硝基苯(5.17g,20.68mmol)溶于乙腈(25mL)中,然后加入碳酸钾(9.5g,68.94mmol)。反应液在氩气保护下加热至80℃搅拌反应4小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:甲醇:二氯甲烷=1:30~1:10)纯化,得到目标产物4-(2-溴-5-甲氧基-4-硝基苯基)吗啉(2-3)(3.4g,收率:79.2%)。
LCMS:Rt:1.715min;MS m/z(ESI):319.0[M+H] +
化合物2-4:
Figure PCTCN2020132188-appb-000061
室温下将4-(2-溴-5-甲氧基-4-硝基苯基)吗啉(2-3)(2.0g,6.33mmol),1-甲基-4-1H-吡唑-硼酸频那醇酯(3.94mg,18.9mmol),碳酸钠(2.0g,18.9mmol)和[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(926mg,1.26mmol)溶于二氧六环(10mL)和水(1mL)的混合液中,反应体系在氮气保护下升温至100℃下搅拌12小时。反应完全后将反应混合物过滤,滤液减压浓缩,残余物通过硅胶柱色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=20:1,Rf=0.5)纯化,得到目标产品4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)吗啉(2-4)(800mg,收率:40%)。
LCMS:Rt:1.410min;MS m/z(ESI):319.1[M+H] +
化合物2-5:
Figure PCTCN2020132188-appb-000062
室温下将4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)吗啉(2-4)(500mg,1.57mmol)溶于乙醇(15mL)并加入钯碳催化剂(25mg),反应液在氢气氛下升温至50℃搅拌反应3h至反应完全。将反应液过滤,滤液减压浓缩,得到粗品目标产物2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯胺(2-5)(450mg,收率:99%)。
LCMS:Rt:0.901min;MS m/z(ESI):289.4[M+H] +
化合物2的三氟乙酸盐:
Figure PCTCN2020132188-appb-000063
室温下将2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯胺(2-5)(112mg,0.309mmol)和(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)(150mg,0.309mmol)溶于异丙醇(8mL)中,再加入三氟乙酸(352.26mg,3.09mmol);将反应液加热至100℃搅拌反应24小时。反应完全后将反应液冷却至室温,然后减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标化合物(3-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物2)三氟乙酸盐(29.8mg,收率:15.7%)。
1H NMR(400MHz,CD 3OD)δ:9.46(brs,1H),8.21-8.17(m,2H),7.98-7.94(m,1H),7.82-7.65(m,4H),7.41(s,1H),6.81(s,1H),3.90(s,3H),3.83-3.60(m,7H),2.85(brs,4H),2.18(s,3H),2.14(s,3H).
19F NMR(376.5MHz,CD 3OD):δ-77.37.
31P NMR(162.0MHz,CD 3OD):δ47.45.
LCMS:Rt:1.320min;MS m/z(ESI):619.1[M+H] +
实施例3、(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物3)的三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000064
化合物3的三氟乙酸盐:
Figure PCTCN2020132188-appb-000065
室温下将(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)(150mg,0.363mmol),2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯胺(2-5)(104.6mg,0.363mmol)和三氟乙酸(413.82mg,3.63mmol)溶于异丙醇(5mL)中,反应在100℃下搅24小时。反应完全后将反应液冷却并减压浓缩,残余物通过高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得纯品目标产物(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物3)三氟乙酸盐(30.1mg,收率:7.1%)。
1H NMR(400MHz,CD 3OD)δ:9.35(brs,1H),8.22-8.20(m,2H),7.96(s,1H),7.83-7.54(m,4H),7.42(s,1H),6.79(s,1H),3.90(brs,3H),3.76-3.73(m,7H),2.84(brs,4H),2.17(s,3H),2.14(s,3H).
19F NMR(376.5MHz,CD 3OD):δ-77.32.
31P NMR(162.0MHz,CD 3OD):δ46.95.
LCMS:Rt:1.388min;MS m/z(ESI):663.3,665.3[M+H] +
实施例4、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-吗啉基吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物4)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000066
化合物4-2:
Figure PCTCN2020132188-appb-000067
0℃下将2,3,6-三氯-5-硝基吡啶(4-1)(625mg,2.75mmol)和甲醇(89mg,2.75mmol)溶于四氢呋喃(20mL)中,再加入钠氢(98.8mg,4.13mmol);反应在0℃下继续搅拌反应0.5小时后,反应用水(50mL)淬灭,混合物用乙酸乙酯(30mL X 3)萃取,有机相用无水硫酸钠干燥过滤,滤液减压浓缩,得到粗品目标产品2,3-二氯-6-甲氧基-5-硝基吡啶(4-2)和2,5-二氯-6-甲氧基-3-硝基吡啶(4-2A)的混合物(500mg,收率:82%)。
化合物4-3:
Figure PCTCN2020132188-appb-000068
室温下将2,3-二氯-6-甲氧基-5-硝基吡啶(4-2)和2,5-二氯-6-甲氧基-3-硝基吡啶混合物(4-2A)(500mg,2.24mmol),吗啉(391mg,4.48mmol)加入到N,N-二甲基甲酰胺(5mL)中。反应液在室温下搅拌反应5小时。反应完全后将反应液倒入水(10mL)中,混合物用乙酸乙酯(30mL X 3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱机梯度:二氯甲烷:甲醇=10:1)纯化,得到目标产物4-(3-氯-6-甲氧基-5-硝基吡啶-2-基)吗啉(4-3)(80mg,收率:45.8%)。
LCMS:Rt:1.683min;MS m/z(ESI):274.2[M+H] +
化合物4-4:
Figure PCTCN2020132188-appb-000069
室温下将4-(3-氯-6-甲氧基-5-硝基吡啶-2-基)吗啉(4-3)(170mg,0.62mmol),1-甲基-4-1H-吡唑-硼酸频那醇酯(259mg,1.24mmol),碳酸钠(606mg)加入到二氧六环(3mL)和水(1mL)的混合溶剂中,体系抽真空充氩气三次。然后加入二氯双[二叔丁基-(4-二甲基氨基苯基)膦]钯(88mg,0.124mmol),再抽真空充氩气三次。将反应液加热至105℃搅拌反应16小时。反应完全后,将反应液冷却至室温并减压浓缩,残余物通过制备薄层色谱层析法(洗脱机梯度:二氯甲烷:甲醇=10:1)纯化,得到目标产物4-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)吗啉(4-4)(170.26mg,收率:86%)。
LCMS:Rt:1.457min;MS m/z(ESI):320.1[M+H] +
化合物4-5:
Figure PCTCN2020132188-appb-000070
室温下将4-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)吗啉(4-4)(170.26mg,0.53mmol)加入到乙醇(5mL)中,加入钯碳催化剂(45mg),反应体系抽真空充氢气三次,然后将反应液加热至50℃反应3小时。反应完全后将反应液过滤,滤液减压浓缩,得到目标产物2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-吗啉基吡啶-3-胺(4-5)(140mg,收率:90.7%)。
LCMS:Rt:1.058min;MS m/z(ESI):290.1[M+H] +
化合物4-7:
Figure PCTCN2020132188-appb-000071
室温下将喹喔啉-6-胺(4-6)(5.0g,34.48mmol)溶于乙酸(150mL)中,将氯化碘(6.1g,37.58mmol)的乙酸(55mL)溶液缓慢滴加到反应液中,反应液在氩气氛下在20℃搅拌反应2小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:20~1:3)纯化,得到目标产物5-碘喹喔啉-6-胺(4-7)(6.0g,收率:67%)。
LCMS:Rt:1.325min;MS m/z(ESI):272.1[M+H] +
化合物4-8:
Figure PCTCN2020132188-appb-000072
室温下将5-碘喹喔啉-6-胺(4-7)(6.0g,22.1mmol),二甲基膦氧(2.6g,33.2mmol)和磷酸钾(7.0g,33.2mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(1.2g,2.2mmol),醋酸钯(494mg,2.2mmol)溶于N,N-二甲基甲酰胺(100mL)和水(20mL)中。反应液在氩气氛下加热至120℃搅拌反应24小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:10~1:1)纯化,得到目标产物(6-氨基喹喔啉-5-基)二甲基氧化膦(4-8)(4.0g,收率:67%)。
LCMS:Rt:0.902min;MS m/z(ESI):221.9[M+H] +
化合物4-9:
Figure PCTCN2020132188-appb-000073
室温下将(6-氨基喹喔啉-5-基)二甲基氧化膦(4-8)(1.0g,4.52mmol)溶于乙醇(20mL)中,再加入5-溴-2,4-二氯嘧啶(2.0g,9.04mmol)和N,N-二异丙基乙胺(3.5g,27.1mmol)。反应液在氩气保护下加热至90℃搅拌反应72小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:10~1:1)纯化,得到目标产物(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(0.5g,收率:27%)。
LCMS:Rt:1.453min;MS m/z(ESI):411.9[M+H] +
化合物4的三氟乙酸盐:
Figure PCTCN2020132188-appb-000074
室温下将2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-吗啉基吡啶-3-胺(4-5)(140mg,0.48mmol),(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(160mg,0.39mmol),加入到异丙醇(3mL)中,再加入三氟乙酸(547mg,4.8mmol),将反应液加热至100℃搅拌反应16小时。反应完全后,将反应液冷却至室温,然后减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化。得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-吗啉基吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物4)的三氟乙酸盐(25mg,收率:9.6%)。
1H NMR(400MHz,CD 3OD):δ8.87(d,J=1.6Hz,1H),8.84(d,J=1.6Hz,1H),8.70(brs,1H),8.29(s,1H),7.91(s,1H),7.84(s,1H),7.71(brs,1H),7.52(brs,1H),3.99(s,3H),3.80-3.78(m,7H),3.18-3.12(m,4H),2.17(s,3H),2.13(s,3H).
31P NMR(162.0MHz,CD 3OD):δ53.12.
19F NMR(376.5MHz,CD 3OD):δ-77.40.
LCMS:Rt:1.572min;MS m/z(ESI):665.1,667.1[M+H] +
实施例5、(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(哌啶-4-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物5)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000075
化合物5-2:
Figure PCTCN2020132188-appb-000076
室温下将1-氯-2-碘-5-甲氧基-4-硝基苯(5-1)(2.0g,6.38mmol),1-甲基-4-1H-吡唑-硼酸频那醇酯(1.32g,6.38mmol)和碳酸钠(2.0g,19.14mmol)加入到二氧六环(20mL)和水(6mL)中,氩气置换三次后,加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(930mg,1.27mmol),氩气再置换三次后将反应液升温到90℃搅拌反应4小时。反应完全后将反应液直接减压浓缩,残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯/石油醚=1/4)纯化,得目标化合物4-(2-氯-4-甲氧基-5-硝基苯基)-1-甲基-1H-吡唑(5-2)(1.5g,收率:88%)。
LCMS:Rt:0.76min;MS m/z(ESI):268.1[M+H] +
化合物5-3:
Figure PCTCN2020132188-appb-000077
室温下将4-(2-氯-4-甲氧基-5-硝基苯基)-1-甲基-1H-吡唑(5-2)(1.3g,4.86mmol),4-(4,4,5,5-四甲基-1,3,2-二氧硼-2-基)-3,6-二氢吡啶-1(2H)-甲酸叔丁酯(1.5g,4.86mmol)和碳酸钠(1.55g,14.58mmol)加入到二氧六环(20mL)和水(6mL)中,氩气置换三次,再加入二氯二叔丁基-(4-二甲基氨基苯基)磷钯(II)(344mg,0.486mmol),氩气再置换三次后将反应液升温到105℃搅拌反应3小时。反应完全后将反应液减压浓缩,残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯/石油醚=1/4)纯化,得到目标中间体4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-3,6-二氢吡啶-1(2H)-甲酸叔丁酯(5-3)(1.0g,收率:50%)。
LCMS:Rt:1.815min;MS m/z(ESI):415.1[M+H] +
化合物5-4:
Figure PCTCN2020132188-appb-000078
室温下将4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-3,6-二氢吡啶-1(2H)-甲酸叔丁酯(5-3)(900mg,2.17mmol)加入到甲醇(2mL)溶解,再加盐酸甲醇溶液(2N,11mL),反应在室温下搅拌反应2h。反应完全后将反应液直接减压浓缩,得到粗品目标产物4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-1,2,3,6-四氢吡啶(5-4)(700mg,收率:100%)。
LCMS:Rt:0.876min;MS m/z(ESI):315.3[M+H] +
化合物5-5:
Figure PCTCN2020132188-appb-000079
室温下将4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-1,2,3,6-四氢吡啶(5-4)(700mg,2.23mmol)和4-二甲氨基吡啶(272mg,2.23mmol)溶于二氯甲烷(20mL)中,再加入三氟乙酸酐(560mg,2.67mmol),反应在室温下搅拌反应6小时。反应完全后将反应液减压浓缩,残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯/石油醚=1/3)纯化,得到目标中间体2,2,2-三氟乙酰-1-(4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-3,6-二氢吡啶(5-5)(600mg,收率:66%)。
LCMS:Rt:1.561min;MS m/z(ESI):411.0[M+H] +
化合物5-6:
Figure PCTCN2020132188-appb-000080
室温下将2,2,2-三氟乙酰-1-(4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)-3,6-二氢吡啶(5-5)(600mg,1.46mmol),钯碳催化剂(150mg)加入到甲醇(30mL)中,反应体系用 氢气置换四次后升温到70℃搅拌反应28小时。反应液过滤,滤液减压浓缩,得到粗品目标中间体2,2,2-三氟乙酰-1-4-(4-氨基-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)哌啶(5-6)(450mg,收率:80%)。
LCMS:Rt:1.275min;MS m/z(ESI):383.1[M+H] +
化合物5-7:
Figure PCTCN2020132188-appb-000081
室温下将1-(2,2,2-三氟乙酰)-4-(4-氨基-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)哌啶(5-6)(100mg,0.26mmol),(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)(86mg,0.21mmol)和三氟乙酸(300mg,2.6mmol)溶于异丙醇(20mL)中,反应液在氩气氛下加热至105℃搅拌反应16小时。将反应混合物减压浓缩,残余物加入乙酸乙酯(10mL)打浆,悬浊液过滤,固体干燥得到中间体1-三氟乙酰-4-(4-((5-溴-4-((4-(二甲基磷酰基)喹啉-3-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)哌啶(5-7)(80g,收率:40%)。
LCMS:Rt:1.33min;MS m/z(ESI):759.3[M+H] +
化合物5的三氟乙酸盐:
Figure PCTCN2020132188-appb-000082
室温下将1-三氟乙酰-4-(4-((5-溴-4-((4-(二甲基磷酰基)喹啉-3-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)哌啶(5-7)(60mg,0.079mmol),氢氧化钾(44mg,0.79mmol)加入到甲醇(5mL)和水(2mL)中。反应液在氩气氛下加热至60℃搅拌反应16小时。将反应混合物减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标化合物(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(哌啶-4-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物5)三氟乙酸盐(46mg,收率:88%)。
1H NMR(400MHz,CD 3OD):δ9.44(d,J=4.0Hz,1H),8.28(s,1H),8.20(d,J=8.4Hz,1H),7.99(d,J=8.0Hz,1H),7.81-7.71(m,2H),7.59(d,J=4.0Hz,1H),7.33(s,1H),7.23(s,1H),6.93(s,1H),3.94(s,3H),3.67(s,3H),3.46(d,J=12.8Hz,2H),3.13-3.10(m,1H),3.06-2.98(m,2H),2.18(s,3H),2.14(s,3H),1.93-1.90(m,4H).
31P NMR(162.0MHz,CD 3OD):δ47.33.
19F NMR(376.5MHz,CD 3OD):δ-77.24.
LCMS:Rt:0.946min;MS m/z(ESI):661.1,663.1[M+H] +
实施例6、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌啶-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物6)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000083
化合物6-2:
Figure PCTCN2020132188-appb-000084
室温下将2,3-二氯-6-甲氧基-5-硝基吡啶(4-2)(5.0g,22.42mmol),4-(4,4,5,5-四甲基-1,3,2-二氧硼-2-基)-3,6-二氢吡啶-1(2H)-甲酸叔丁酯(6.93g,22.42mmol),碳酸钠(5.94g,56.05mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(1.6g,2.242mmol)加入到二氧六环(50mL)和水(12mL)中。反应体系用氩气置换三次后在90℃下反应5小时。反应完全后将反应液冷却至室温并过滤,滤液用乙酸乙酯(80mL X 3)萃取,合并有机相用饱和食盐水(60mL)洗涤,然后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:50)纯化,得到目标产物3-氯-6-甲氧基-5-硝基-3',6'-二氢-[2,4'-联吡啶]-1'(2'H)-甲酸叔丁酯(6-2)(450mg,收率:5.4%)。
1H NMR(400MHz,CDCl 3):δ8.35(s,1H),6.55(brs,1H),4.16(br,1H),4.10(s,3H),3.66-3.64(m,2H),2.63(br,2H),1.50(s,9H).
化合物6-3:
Figure PCTCN2020132188-appb-000085
室温下将3-氯-6-甲氧基-5-硝基-3',6'-二氢-[2,4'-联吡啶]-1'(2'H)-甲酸叔丁酯(6-2)(450mg,0.95mmol),1-甲基-4-1H-吡唑-硼酸频那醇酯(197mg,0.95mmol),碳酸铯(770mg,2.37mmol)和二氯二叔丁基-(4-二甲基氨基苯基)磷钯(II)(67mg,0.09mmol)加入到二氧六环(5mL)和水(2mL)中。体系用氩气置换三次后在105℃下反应5小时至反应完全。将反应液冷却至室温并过滤,滤液用乙酸乙酯(30mL X 3)萃取,合并有机相用饱和食盐水(20mL)洗涤,然后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:20)纯化,得到目标产物6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基-3',6'-二氢-[2,4'-二氢]-1'(2'H)-甲酸叔丁酯(6-3)(210mg,收率:53.41%)。
化合物6-4:
Figure PCTCN2020132188-appb-000086
室温下将6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基-3',6'-二氢-[2,4'-二氢]-1'(2'H)-甲酸叔丁酯(6-3)(210mg,0.51mmol)溶于二氯甲烷(6mL)中,然后加入三氟乙酸(3mL)。反应体系在室温下反应3小时至反应完全。将反应液减压浓缩,得到目标产物6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基-1',2',3',6'-四氢-2,4'-联吡啶(6-4)(160mg,粗产品)。直接用于下一步反应。
LCMS:Rt:0.907min;MS m/z(ESI):315.9[M+H] +
化合物6-5:
Figure PCTCN2020132188-appb-000087
室温下将6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基-1',2',3',6'-四氢-2,4'-联吡啶(6-4)(160mg,0.507mmol),三乙胺(256mg,2.53mmol)和N,N-二甲基-4-氨基吡啶(6mg)溶于二氯甲烷(8mL)中,再加入三氟乙酸酐(160mg,0.761mmol)。反应在室温下反应12小时至反应完全。将反应液减压浓缩,残余物通过制备薄层色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:1)纯化,得到目标产物6-5(80mg,收率:38.5%)。
LCMS:Rt:1.797min;MS m/z(ESI):412.0[M+H] +
化合物6-6:
Figure PCTCN2020132188-appb-000088
室温下将中间体化合物6-5(40mmol,0.097mmol))溶于乙醇(5mL)中,再加入钯碳催化剂(5mg,10%wt)。反应体系用氢气置换3次后加热到50℃反应5小时至反应完全。将反应液过滤,滤液减压浓缩,得到目标产物三氟乙酰-1-(4-(5-氨基-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌啶(6-6)(30mg,粗产品)。直接用于下一步反应。
LCMS:Rt:1.297min;MS m/z(ESI):384.0[M+H] +
化合物6-7:
Figure PCTCN2020132188-appb-000089
室温下将1-三氟乙酰-4-(5-氨基-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌啶(6-6)(30mg,0.078mmol),(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(32mg,0.078mmol)溶解于异丙醇(5mL)中,再加入三氟乙酸(89mg,0.78mmol),将反应加热至100℃反应16小时至反应完全。将反应液冷却并减压浓缩,残余物通过制备薄层色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=10:1)纯化,得到目标产物1-三氟乙酰-(4-(5-((5-溴-4-((5-(甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌啶(6-7)(27mg,45.4%)。
LCMS:Rt:1.567min;MS m/z(ESI):761.4[M+H] +
化合物6的三氟乙酸盐:
Figure PCTCN2020132188-appb-000090
室温下将1-三氟乙酰-(4-(5-((5-溴-4-((5-(甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌啶(6-7)(27mg,0.035mmol)溶解于甲醇(3mL)和水(0.5mL)中,再加入碳酸钾(48mg,0.35mmol),将反应加热至70℃反应16小时至反应完全。将反应液冷却并减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标产物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌啶-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物6)三氟乙酸盐(17mg,收率:72.07%)。
1H NMR(400MHz,CD 3OD):δ8.85-8.83(m,2H),8.74(d,J=4.0Hz,1H),8.33(s,1H),8.28(s,1H),7.60(d,J=12.0Hz,1H),7.59(s,1H),7.23(s,1H),4.07(s,3H),3.81(s,3H),3.49-3.46(m,2H),3.25-3.22(m,1H),3.11-3.05(m,2H),2.29-2.19(m,2H),2.16(s,3H),2.12(s,3H),1.95-1.92(m,2H).
31P NMR(162.0MHz,CD 3OD):δ52.86.
19F NMR(376.5MHz,CD 3OD):δ-77.12.
LCMS:Rt:1.000min;MS m/z(ESI):663.1,665.1[M+H] +
实施例7、(3-((5-溴-2-((5-(1-(2-羟基乙基)-1H-吡唑-4-基)-2-甲氧基-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物7)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000091
化合物7-2:
Figure PCTCN2020132188-appb-000092
室温下将4-吡唑硼酸频哪醇酯(7-1)(1.00g,5.154mmol),(2-溴乙氧基)-叔丁基二甲基硅烷(1.85g,7.730mmol)和碳酸钾(1.42g,10.308mmol)加入到乙腈(10mL)中。反应液在90℃下反应16小时。反应完全后将反应液冷却至室温并过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯:石油醚=1:10)纯化,得到目标产物1-(2-((叔丁基二甲基硅基)氧基)乙基)-1H-吡唑-硼酸频那醇酯(7-2)(570mg,收率:31.4%)。
LCMS:Rt:2.115min;MS m/z(ESI):353.5[M+H] +
化合物7-3:
Figure PCTCN2020132188-appb-000093
室温下将1-(2-((叔丁基二甲基硅基)氧基)乙基)-1H-吡唑-硼酸频那醇酯(7-2)(250.00mg,0.708mmol),4-(2-溴-5-甲氧基-4-硝基苯基)吗啉(224.60mg,0.708mmol),碳酸铯(461.35mg,1.416mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(51.81mg,0.071mmol)加入到二氧六环(7.5mL)和水(1.5mL)中。反应体系用氩气置换三次后在100℃下反应16小时。反应完全后将反应液冷却至室温,化合物过滤,滤液用乙酸乙酯(30mL X 3)萃取,合并有机相用饱和食盐水(20mL)洗涤,然后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:乙酸乙酯)纯化,得到目标产物4-(2-(1-(2-((叔丁基二甲基硅基)氧基)乙基)-1H-吡唑-4-基)-5-甲氧基-4-硝基苯基)吗啉(7-3)(175mg,收率:53.5%)。
LCMS:Rt:2.046min;MS m/z(ESI):463.0[M+H] +
化合物7-4:
Figure PCTCN2020132188-appb-000094
室温下将4-(2-(1-(2-((叔丁基二甲基硅基)氧基)乙基)-1H-吡唑-4-基)-5-甲氧基-4-硝基苯基)吗啉(7-3)(175mg,0.379mmol),钯碳催化剂(40mg)加入到乙醇(4mL)中。反应在氢气氛下加热至50℃反应6小时。反应完全后将反应液过滤,滤液减压浓缩,得到目标产物5-(1-(2-((叔丁基二甲基硅基)氧基)乙基)-1H-吡唑-4-基)-2-甲氧基-4-吗啉苯胺(7-4)(100mg,收率:61%)。
LCMS:Rt:1.562min;MS m/z(ESI):433.2[M+H] +
化合物7的三氟乙酸盐:
Figure PCTCN2020132188-appb-000095
室温下将5-(1-(2-((叔丁基二甲基硅基)氧基)乙基)-1H-吡唑-4-基)-2-甲氧基-4-吗啉苯胺(7-4)(100.00mg,0.231mmol),(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)(76.06mg,0.185mmol)和三氟乙酸(263.53mg,2.311mmol)溶于异丙醇(6mL)中。反应在100℃下 反应16小时至反应完全。将反应液冷却后减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标化合物(3-((5-溴-2-((5-(1-(2-羟基乙基)-1H-吡唑-4-基)-2-甲氧基-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物7)三氟乙酸盐(35mg,收率:21.8%)。
1H NMR(400MHz,CD 3OD):δ9.36(s,1H),8.22(s,1H),8.21(d,J=8.0Hz,1H),7.98(d,J=8.0Hz,1H),7.88(brs,1H),7.80-7.72(m,2H),7.61(brs,1H),7.45(s,1H),6.79(s,1H),4.07(brs,2H),3.89(s,3H),3.81-3.76(m,6H),2.84(brs,4H),2.17(s,3H),2.13(s,3H).
31P NMR(162.0MHz,CD 3OD):δ46.97.
19F NMR(376.5MHz,CD 3OD):δ-77.29.
LCMS:Rt:1.258min;MS m/z(ESI):693.4,695.4[M+H] +
实施例8、(6-((5-溴-2-((6-(4-(二甲氨基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物8)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000096
化合物8-2:
Figure PCTCN2020132188-appb-000097
室温下将2,3-二氯-6-甲氧基-5-硝基吡啶(4-2)和2,5-二氯-6-甲氧基-3-硝基吡啶(4-2A)的混合物(1.0g,4.48mmol),N,N-二甲基哌啶-4-胺(1.14g,8.96mmol)加入到N,N-二甲基甲酰胺(10mL)中。反应液在室温下搅拌5小时。反应完全后,向反应液中加入水(80mL),混合物用乙酸乙酯(90mL X 3)萃取。有机相用饱和食盐水洗涤,用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=10:1)纯化,得到目标产物1-(3-氯-6-甲氧基-5-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-2)和1-(5-氯-6-甲氧基-3-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-2)的混合物(590mg,收率:41.84%)。
LCMS:Rt:0.943min;MS m/z(ESI):315.2[M+H] +
化合物8-3:
Figure PCTCN2020132188-appb-000098
室温下将1-(3-氯-6-甲氧基-5-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-2)和1-(5-氯-6-甲氧基-3-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-2A)的混合物(490mg,1.56mmol),1-甲基吡唑-4-硼酸频哪醇酯(649mg,3.12mmol)和碳酸铯(1.53g,4.68mmol)加入到N,N-二甲基甲酰胺:水(3:1,8mL)中,反应体系抽真空充氩气三次,再加入二氯双[二叔丁基-(4-二甲基氨基苯基)膦]钯(220mg,0.312mmol),反应体系再抽真空充氩气三次。将反应液加热至120℃搅拌反应16小时。反应完全后将反应液冷却至室温并用水(80mL)稀释,混合物用乙酸乙酯(100mL X3)萃取。有机相用饱和食盐水洗涤后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标产物1-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-3)(90mg,收率:20%)和1-(6-甲氧基-5-(1-甲基-1H-吡唑-4-基)-3-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-3A)(25mg,收率:20%)。
LCMS:Rt:0.920min;MS m/z(ESI):361.1[M+H]。
化合物8-4:
Figure PCTCN2020132188-appb-000099
室温下将1-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)-N,N-二甲基哌啶-4-胺(8-3)(90mg,0.25mmol)和钯碳催化剂(13.5mg)加入到乙醇(5mL)中,体系抽真空充氢气三次。将反应液加热至50℃搅拌反应5小时。反应完全后,将反应液过滤,滤液减压浓缩。得到粗品目标产物6-(4-(二甲氨基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-胺(8-4)(28mg,收率:33.89%)。
LCMS:Rt:0.560min;MS m/z(ESI):331.1[M+H]。
化合物8的三氟乙酸盐:
Figure PCTCN2020132188-appb-000100
室温下将6-(4-(二甲氨基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-胺(8-4)(28mg,0.085mmol)和(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(34.96mg,0.085mmol)溶于异丙醇(2mL)中,再加入三氟乙酸(96.92mg,0.85mmol)。将反应液加热至100℃反应16小时。反应完全后,将反应液冷却至室温,然后减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标化合物(6-((5-溴-2-((6-(4-(二甲基氨基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物8)三氟乙酸盐(12mg,收率:20%)。
1H NMR(400MHz,CD 3OD)δ:8.85(s,1H)),8.85-8.79(m,2H),8.28(s,1H),8.09(s,1H),7.72(s,1H),7.61(br,1H),7.52(s,1H),4.00(s,3H),3.73(s,3H),3.67-3.64(m,2H),3.35-3.31(m,1H),2.93(s,6H),2.93-2.84(m,2H),2.17(s,3H),2.11(s,3H),2.17-2.09(m,2H),1.88-1.84(m,8.2Hz,2H).
19F NMR(376.5MHz,CD 3OD):δ-77.22.
31P NMR(162.0MHz,CD 3OD):δ52.93.
LCMS:Rt:1.107min;MS m/z(ESI):706.4,708.4[M+H] +
实施例9、(E)-(3-((5-溴-2-((4-(4-(二甲氨基)哌啶-1-基)-2-甲氧基-5-(1-(甲氧基亚氨基)乙基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物9)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000101
化合物9-2:
Figure PCTCN2020132188-appb-000102
室温下将1-(2-溴-5-甲氧基-4-硝基苯基)-N,N-二甲基哌啶-4-胺(1-6)(600.00mg,1.675mmol),三丁基(1-乙氧基乙烯基)锡(907.34mg,2.519mmol)和四(三苯基膦)钯(193.55mg,0.168mmol)加入到1,4-二氧六环(12mL)中。体系用氩气置换三次后加热至110℃反应16小时。LCMS显示反应完全,将反应液冷却至室温过滤。向滤液中加入稀盐酸并在常温下搅拌30分钟至反应完全,再向滤液中加入氟化钾水溶液并继续搅拌30分钟。将反应液过滤,滤液用乙酸乙酯(30mL X 3)萃取,有机相干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:甲醇:二氯甲烷=1:20)纯化,得到目标产物1-(2-(4-(二甲氨基)哌啶-1-基)-4-甲氧基-5-硝基苯基)乙基-1-酮(9-2)(520mg,收率:96.6%)。
LCMS:Rt:1.069min;MS m/z(ESI):350.4[M+H] +
化合物9-3:
Figure PCTCN2020132188-appb-000103
室温下将1-(2-(4-(二甲氨基)哌啶-1-基)-4-甲氧基-5-硝基苯基)乙基-1-酮(9-2)(520.00mg,1.618mmol)溶入乙醇(8mL)和水(8mL)的混合液中,加入氯化铵(259.64mg,4.854mmol)和铁粉(271.10mg,4.854mmol)。反应液在80℃下反应16小时。反应完全后将反应液冷却至室温,混合物过滤,滤液用乙酸乙酯(30mL X 3)萃取,有机相合并后用饱和食盐水(20mL)洗涤,然后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:甲醇:二氯甲烷=1:20)纯化,得到目标产物1-(5-氨基-2-(4-(二甲氨基)哌啶-1-基)-4-甲氧基苯基)乙基-1-酮(9-3)(300mg,收率:63.5%)。
LCMS:Rt:0.355min;MS m/z(ESI):292.5[M+H] +
化合物9-4:
Figure PCTCN2020132188-appb-000104
室温下将1-(5-氨基-2-(4-(二甲氨基)哌啶-1-基)-4-甲氧基苯基)乙基-1-酮(9-3)(300.00mg,1.030mmol)和(6-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-5-基)二甲基氧化膦(1-4)(300.00mg,0.729mmol)溶于异丙醇中,再加入三氟乙酸(1000.00mg,8.770mmol)中。反应液在100℃下反应24小时至反应完全。将反应液过滤,滤液减压浓缩,残余物通过硅胶柱色谱层析法(洗脱剂梯度:甲醇:二氯甲烷=1:10)纯化,得到目标产物1-(5-((5-溴-4-((4-(二甲基磷酰基)喹啉-3-基)氨基)嘧啶-2-基)氨基)-2-(4-(二甲基氨基)哌啶-1-基)-4-甲氧基苯基)乙基-1-酮(9-4)(300mg)。
LCMS:Rt:0.893min;MS m/z(ESI):666.1[M+H] +
化合物9的三氟乙酸盐:
Figure PCTCN2020132188-appb-000105
室温下将1-(5-((5-溴-4-((4-(二甲基磷酰基)喹啉-3-基)氨基)嘧啶-2-基)氨基)-2-(4-(二甲基氨基)哌啶-1-基)-4-甲氧基苯基)乙基-1-酮(9-4)(125.00mg,0.188mmol),甲氧基胺盐酸盐(46.99mg,0.563mmol)和冰醋酸(112.894mg,1.880mmol)溶于1,2-二氯乙烷(2mL)中。反应在50℃下反应16小时至反应完全。先将反应液冷却后减压浓缩,残余物通过高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得纯品目标产物(E)-(3-((5-溴-2-((4-(4-(二甲氨基)哌啶-1-基)-2-甲氧基-5-(1-(甲氧基亚氨基)乙基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物9)三氟乙酸盐(65mg,收率:49.7%)。
1H NMR(400MHz,CD 3OD)δ:9.41(d,J=4.4Hz,1H),8.26(d,J=6.0Hz,1H),8.26(s,1H),8.09(d,J=8.4Hz,1H),7.82-7.71(m,2H),7.53(s,1H),6.82(s,1H),3.92(s,3H),3.61(s,3H),3.38-3.26(m,4H),2.92(s,6H),2.86-2.79(m,1H),2.21-2.12(m,2H),2.18(s,3H),2.12(s,3H),2.02(s,3H),1.86-1.76(m,2H).
19F NMR(376.5MHz,CD 3OD):δ-77.18.
31P NMR(162.0MHz,CD 3OD):δ47.08.
LCMS:Rt:1.058min;MS m/z(ESI):695.5,697.5[M+H] +
实施例10、(E)-(3-((5-溴-2-((4-(4-(二甲氨基)哌啶-1-基)-5-(1-((2-羟基乙氧基)亚氨基)乙基)-2-甲氧苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物10)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000106
化合物10的三氟乙酸盐:
Figure PCTCN2020132188-appb-000107
室温下将1-(5-((5-溴-4-((4-(二甲基磷酰基)喹啉-3-基)氨基)嘧啶-2-基)氨基)-2-(4-(二甲基氨基)哌啶-1-基)-4-甲氧基苯基)乙基-1-酮(9-4)(125.00mg,0.188mmol),2-氨氧基乙醇(43.40mg,0.563mmol)和冰醋酸(112.894mg,1.880mmol)溶于1,2-二氯乙烷(2mL)中。反应在50℃下反应16小时至反应完全。先将反应液冷却后减压浓缩,残余物通过高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得目标产物(E)-(3-((5-溴-2-((4-(4-(二甲氨基)哌啶-1-基)-5-(1-((2-羟基乙氧基)亚氨基)乙基)-2-甲氧基苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物10)三氟乙酸盐(35mg,收率:25.7%)。
1H NMR(400MHz,CD3OD)δ:9.42-9.40(m,1H),8.27(d,J=8.4Hz,1H),8.25(s,1H),8.10(d,J=8.3Hz,1H),7.80-7.72(m,2H),7.53(s,1H),6.81(d,J=6.0Hz,1H),3.96-3.87(m,2H),3.88(s,3H),3.66-3.64(m,2H),3.37-3.34(m,3H),2.94(s,6H),2.90-2.75(m,2H),2.17-2.13(m,2H),2.17(s,3H),2.13(s,3H),2.03(s,3H),1.85(brs,2H).
19F NMR(376.5MHz,CD3OD):δ-77.22.
31P NMR(162.0MHz,CD3OD):δ47.08.
LCMS:Rt:1.016min;MS m/z(ESI):725.4,727.4[M+H] +
实施例11、(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物82)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000108
化合物82-2:
Figure PCTCN2020132188-appb-000109
室温下将1-溴-2-氟-4-甲氧基-5-硝基苯(4.2g,16.80mmol)和1-甲基-4-(哌啶-4-基)哌嗪(82-1)(3.08g,16.80mmol)溶于N,N-二甲基甲酰胺(20mL)中,加入碳酸钾(6.97g,50.4mmol),反应液在氩气保护下加热至60℃搅拌反应4小时至反应完全。将反应液直接减压浓缩,残余物用硅胶柱色谱层析法(洗脱剂梯度:甲醇:二氯甲烷=1:30~1:10)纯化,得到目标产物1-(1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)-4-甲基哌嗪(82-2)(1.5g,收率:22%)。
化合物82-3:
Figure PCTCN2020132188-appb-000110
室温下将1-(1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)-4-甲基哌嗪(82-2)(500mg,1.21mmol),1-甲基吡唑-4-硼酸频哪醇酯(504mg,2.42mmol),碳酸钠(385mg,3.63mmol)加入到1,4-二氧六环和水的混合溶剂(1,4-二氧六环:水=3:1.8mL)中,体系抽真空充氩气三次,加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(89mg,0.121mmol),再抽真空充氩气三次。将反应液加热至105℃搅拌反应16小时。用LCMS检测反应完全后,将反应液冷却至室温。将反应液加入到水(60mL)中,混合物用乙酸乙酯(70mL)稀释后,垫硅藻土过滤,滤液用乙酸乙酯(70mL X 3)萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥过滤,滤液减压浓缩。残余物通过硅胶柱色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=15:1)纯化,得到目标产物1-(1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)哌啶-4-基)-4-甲基哌嗪(82-3)(164mg,收率:33.0%)。
LCMS:Rt:0.880min;MS m/z(ESI):415.1[M+H] +
化合物82-4:
Figure PCTCN2020132188-appb-000111
室温下将1-(1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)哌啶-4-基)-4-甲基哌嗪(82-3)(164mg,0.4mmol)和钯碳催化剂(30mg)加入到乙醇(3mL)中,体系抽真空充氢气置换三次。将反应液加热至50℃反应6小时。用LCMS检测反应,待反应完全后,将反应液冷却至室温并过滤,滤液减压浓缩,得到粗品目标产物2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯胺(82-4)(118mg,收率:76.7%)。
LCMS:Rt:0.300min;MS m/z(ESI):385.2[M+H] +
化合物82的三氟乙酸盐:
Figure PCTCN2020132188-appb-000112
室温下将2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯胺(82-4)(118mg,0.31mmol),(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)(115mg,0.28mmol)溶于异丙醇(3mL)中,加入三氟乙酸(353mg,3.1mmol),将反应液加热至100℃搅拌反应13小时至LCMS检测反应完全。将反应液冷却至室温并减压浓缩,残余物通过高效液相制备色谱法(洗脱剂梯度:同实施例1)纯化,得到目标化合物(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物82)三氟乙酸盐(26mg,收率:11%)。
1H NMR(400MHz,CD 3OD):δ9.43(d,J=4.4Hz,1H),8.22(s,1H),8.14-8.10(m,1H),7.82-7.78(m,2H),7.70-7.66(m,2H),7.48(d,J=6.4Hz,2H),6.75(s,1H),3.87(s,3H),3.53(brs,3H),3.08(d,J=11.6Hz,2H),2.80-2.40(m,9H),2.32-2.24(m,2H),2.31(s,3H),2.18(s,3H),2.15(s,3H),1.96-1.92(m,2H),1.60-1.55(m,2H).
19F NMR(377MHz,CD 3OD):δ-76.94.
31P NMR(162MHz,CD 3OD):δ47.58.
LCMS:Rt:0.788min;MS m/z(ESI):759.2,761.2[M+H] +
实施例12、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉代苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物12)的制备
Figure PCTCN2020132188-appb-000113
参考实施例2的合成方法,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉代苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物12)。
1H NMR(400MHz,CD 3OD)δ:8.81(s,2H),8.76(s,1H),8.24(s,1H),7.93(s,1H),7.86(s,1H),7.52-7.43(m,2H),6.82(s,1H),3.91(s,3H),3.80(br,4H),3.68(s,3H),2.88(br,4H),2.16(s,3H),2.12(s,3H).
31P NMR(162.0MHz,CD 3OD)δ:52.84.
LCMS:Rt:6.757min;MS m/z(ESI):664.1,666.1[M+H] +
实施例13、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(4-甲基哌嗪-1-基)哌啶-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物13)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000114
化合物13-2:
Figure PCTCN2020132188-appb-000115
室温下将2,3-二氯-6-甲氧基-5-硝基吡啶(4-2)(4.0g,17.9mmol)和1-甲基-4-(哌啶-4-基)哌嗪(3.62g,19.7mmol)加入到N,N-二甲基甲酰胺(30ml)中,室温下反应12小时。TLC显示反应完全,向反应液中加入水100mL,用乙酸乙酯(40mL X 4)萃取,合并有机相,用饱和食盐 水(50mL)洗涤,无水硫酸钠干燥,减压浓缩。柱层析色谱法(洗脱剂:二氯甲烷:甲醇50:1)纯化,得到目标产物1-(1-(3-氯-6-甲氧基-5-硝基吡啶-2-基)哌啶-4-基)-4-甲基哌嗪(13-2)(3.7g,收率:55.77%)。
LCMS:Rt:0.940min;MS m/z(ESI):370.1[M+H] +
化合物13-3:
Figure PCTCN2020132188-appb-000116
室温下将1-(1-(3-氯-6-甲氧基-5-硝基吡啶-2-基)哌啶-4-基)-4-甲基哌嗪(13-2)(1.0g,2.71mmol)加入到N,N-二甲基甲酰胺(10mL)和水(2.5mL)中,加入1-甲基-4-吡唑硼酸频哪醇酯(733mg,3.52mmol),碳酸铯(1.8g,5.42mmol)和二氯二叔丁基-(4-二甲基氨基苯基)磷钯(II)(191mg,0.27mmol)。用氮气置换三次,在120℃下反应12小时。TLC显示反应完全,将反应液冷却至室温,过滤,乙酸乙酯(30mL X 3)萃取,合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,减压浓缩。柱层析色谱法(洗脱剂:二氯甲烷:甲醇50:1)纯化,得到目标产物1-(1-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌啶-4-基)-4-甲基哌嗪(13-3)(90mg,收率:8%)。
LCMS:Rt:0.843min;MS m/z(ESI):416.2[M+H] +
化合物13-4:
Figure PCTCN2020132188-appb-000117
室温下将1-(1-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌啶-4-基)-4-甲基哌嗪(13-3)溶于甲醇/四氢呋喃(2mL/2mL)中,加入钯/碳(10mg,10%wt)。用氢气置换3次,室温反应12小时,LCMS检测反应完全。将反应液降至室温,过滤,母液浓缩得到目标产物2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(4-甲基哌嗪-1-基)哌啶-1-基)吡啶-3-胺(13-4)(80mg,收率:98.9%)。粗品直接用于下一步反应。
LCMS:Rt=0.443min;MS m/z(ESI):386.2[M+H] +
化合物13的三氟乙酸盐:
Figure PCTCN2020132188-appb-000118
室温下将2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(4-甲基哌嗪-1-基)哌啶-1-基)吡啶-3-胺(13-4)(80mg,0.207mmol)和(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(85mg,0.207mmol)溶解于3mL异丙醇中,加入三氟乙酸(0.5mL),反应加热至105℃,反应12小时。先将反应液冷却然后浓缩,残余物通过prep-HPLC(洗脱剂梯度参考实施例1)纯化得到目标产物化合物13的三氟乙酸盐(30mg,19.1%)。
1H NMR(400MHz,CD 3OD)δ8.87-8.71(m,3H),8.30(s,1H),7.95(s,1H),7.80(s,1H),7.68(s,1H),7.48(s,1H),3.99(s,3H),3.79(s,3H),3.67(d,J=12.1Hz,2H),3.52(br,8H),3.24(br,1H),2.97–2.82(m,5H),2.17-2.14(m,8H),1.88-1.86(m,2H).
19F NMR(162MHz,CD 3OD)δ-77.26.
31P NMR(162MHz,CD 3OD)δ53.07.
LCMS:Rt:1.020min;MS m/z(ESI):761.2,763.2[M+H] +
实施例14、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物14)的制备
Figure PCTCN2020132188-appb-000119
化合物14:
Figure PCTCN2020132188-appb-000120
室温下将2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯胺(82-4)(156mg,0.41mmol)、(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(140mg,0.34mmol)溶于异丙醇(3mL)中,然后再加入三氟乙酸(389mg,3.41mmol)。将反应液加热至100℃反应16小时。用LCMS检测反应,待反应完全后,将反应液冷却至室温。然后减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度参考实施例1)纯化,得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦三氟乙酸盐(50mg),产物溶解在5mL的二氯甲烷中,用饱和碳酸钠水溶液洗涤(5mL X 3),再用10mL水洗涤,干燥,旋干得目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物14)(25mg,收率:9.68%)。
1H NMR(400MHz,MeOD):δ8.81-8.77(m,3H),8.24(s,1H),7.91(s,1H),7.84(s,1H),7.45-7.36(m,2H),6.82(s,1H),3.90(s,3H),3.69(s,3H),3.15-3.12(m,2H),2.63–2.58(m,10H),2.31(br,4H),2.16(s,3H),2.11(s,3H),1.98-1.95(m,2H),1.64-1.59(m,2H).
31P NMR(162MHz,MeOD):δ52.85.
LCMS:Rt:1.014min;MS m/z(ESI):760.1,762.1[M+H] +
实施例15、((6-((5-溴-2-((4-(3-氟-4-吗啉代哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物83)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000121
化合物83-2:
Figure PCTCN2020132188-appb-000122
室温下将3-氟-4-氧代哌啶-1-羧酸叔丁酯(500.00mg,2.302mmol),吗啉(240.66mg,2.762mmol)溶于1,2-二氯乙烷(10mL)中,加入冰醋酸(207.18mg,3.453mmol),50℃下反应5小时,之后降至室温,缓慢加入三乙酰氧基硼氢化钠(976.05mg,4.604mmol),加入完毕后将反应在室温下搅拌1小时,TLC显示反应完全,加入饱和碳酸钠水溶液调节PH至9~10,用二氯甲烷(30mL X 3)萃取,饱和氯化钠水溶液(30mL)洗涤,无水硫酸钠干燥,浓缩,得到目标产物3-氟-4-吗啉哌啶-1-羧酸叔丁酯(83-2)(650.00mg,收率:97.9%)。
LCMS:Rt=0.820min;MS m/z(ESI):289.1[M+H] +
化合物83-3:
Figure PCTCN2020132188-appb-000123
室温下将3-氟-4-吗啉哌啶-1-羧酸叔丁酯(83-2)(650mg,2.254mmol)加入到盐酸二氧六环溶液(4M,15mL)中,在室温下反应2小时。LCMS检测反应完全,将反应液浓缩,得到目标产物4-(3-氟哌啶-4-基)吗啉盐酸盐(83-3)(420.00mg,收率:62.7%)。
LCMS:Rt=0.445min;MS m/z(ESI):189.5[M+H] +
化合物83-4:
Figure PCTCN2020132188-appb-000124
室温下将4-(3-氟哌啶-4-基)吗啉盐酸盐(83-3)(420.00mg,1.414mmol),1-溴-2-氟-4-甲氧基-5-硝基苯(353.50mg,1.414mmol)溶于N,N-二甲基甲酰胺(8mL)中,加入碳酸钾(585.40mg,4.242mmol),在50℃下搅拌反应16小时,LCMS检测反应完全。将反应液过滤,浓缩,柱层析分离,得到目标产物4-(1-(2-溴-5-甲氧基-4-硝基苯基)-3-氟哌啶-4-基)吗啉(83-4)(200.00mg,收率:33.8%)。
LCMS:Rt=1.010min;MS m/z(ESI):418.0[M+H] +
化合物83-5:
Figure PCTCN2020132188-appb-000125
室温下将4-(1-(2-溴-5-甲氧基-4-硝基苯基)-3-氟哌啶-4-基)吗啉(83-4)(200.00mg,0.478mmol),1-甲基-4-吡唑硼酸频哪醇酯(99.42mg,0.478mmol和碳酸钠(55.09mg,0.956mmol)溶于1,4-二氧六环(5mL)和水(1mL)的混合溶液中,再加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(35.09mg,0.048mmol),置换氮气三次,在100℃下搅拌16小时,LCMS检测反应完全。将反应液过滤,浓缩,得到目标产物4-(3-氟-1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)哌啶-4-基)吗啉(83-5)(180.00mg,收率:89.8%),直接用于下一步。
LCMS:Rt=1.030min;MS m/z(ESI):420.5[M+H] +
化合物83-6:
Figure PCTCN2020132188-appb-000126
室温下将4-(3-氟-1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)哌啶-4-基)吗啉(83-5)(180.00mg,0.429mmol)溶于无水乙醇(30mL)中,加入湿钯/碳(100mg(Pd>=10%,H 2Owt%=50%)),置换氢气三次,在60℃下反应4小时。将反应液通过硅藻土过滤除去钯碳,浓缩,得到目标产物4-(3-氟-4-吗啉哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯胺(83-6)(150.00mg,收率:89.8%)。
LCMS:Rt=0.430min;MS m/z(ESI):390.5[M+H] +
化合物83的三氟乙酸盐:
Figure PCTCN2020132188-appb-000127
室温下将4-(3-氟-4-吗啉哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯胺(83-6)(150.00mg,0.385mmol)和(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(158.91mg,0.385mmol)溶于异丙醇(5mL)中,加入三氟乙酸(0.8mL),在105℃下反应16小时。将反应液通过高效液相制备色谱法(洗脱剂梯度参考实施例1)分离纯化得到目标产物((6-((5-溴-2-((4-(3-氟-4-吗啉代哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物83)三氟乙酸盐(60.00mg,收率:20.3%)。
1H NMR(400MHz,CDCl 3)δ12.59(s,1H),8.96(d,J=9.5,4.2Hz,1H),8.71(d,J=10.4,1.8Hz,2H),8.29(d,J=3.9Hz,2H),7.98(br,1H),7.55(br,1H),7.40-7.36(m,2H),6.67(s,1H),5.26(br,1H),3.95-3.90(m,7H),3.79(s,3H),3.54-3.49(m,1H),3.26-3.25(m,1H),2.97–2.57(m,7H),2.14–2.10(m,7H),1.91(br,1H).
19F NMR(377MHz,CDCl 3)δ-198.78.
31P NMR(162MHz,CDCl 3)δ49.33.
LCMS:Rt:1.010min;MS m/z(ESI):765.2,767.2[M+H] +
实施例16、(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)-二甲基氧化膦(化合物84)的制备
Figure PCTCN2020132188-appb-000128
化合物84-2:
Figure PCTCN2020132188-appb-000129
室温下将(6-胺基喹喔啉-5-基)二甲氧化膦(4-8)(1.0g,4.52mmol)和2,4,5-三氯嘧啶(2.81g,15.32mmol)溶于乙醇(30mL)并在室温下搅拌,再加入二异丙基乙胺(4.0mL,22.87 mmol)。待加料完毕后,室温下搅拌10分钟,再将反应置于100℃油浴中,加热回流反应22小时。停止反应并冷却至室温,旋干。将所得粗品经硅胶柱层析(二氯甲烷:甲醇=30:1)分离纯化,得到(6-((2,5-二氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲氧化膦(84-2)(150mg,收率:49.8%)。
LCMS:Rt:1.500min;MS m/z(ESI):368.0[M+H] +
化合物84:
Figure PCTCN2020132188-appb-000130
室温下将(6-((2,5-二氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲氧化膦(84-2)(179mg,0.485mmol)和4-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)吗啉(2-4)(140mg,0.485mmol)溶于异丙醇(6mL)并于室温下搅拌,再加入三氟乙酸(0.4mL,4.85mmol)。待加料完毕后,将反应置于100℃油浴中,加热回流反应14小时。将反应液冷却至室温并减压浓缩,所得粗品通过高效液相制备色谱法纯化,得到目标产物(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-吗啉苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)-二甲基氧化膦(化合物84)(110mg,收率:36.6%)。
1H NMR(400MHz,CDCl 3)δ:12.78(s,1H),9.16-9.13(m,1H),8.73(d,J=1.9Hz,1H),8.70(d,J=1.9Hz,1H),8.23(s,1H),8.18(s,1H),7.77(s,1H),7.64(s,1H),7.60-7.58(m,1H),7.34(s,1H),6.71(s,1H),3.93(s,3H),3.81-3.78(m,4H),3.78(s,3H),2.95–2.86(m,4H),2.14(s,3H),2.10(s,3H).
31P NMR(162.0MHz,CDCl 3)δ:49.739.
LCMS:Rt:6.667min;MS m/z(ESI):620.1[M+H] +
实施例17、(6-((5-溴-2-((4-((2S,6R)-2,6-二甲基吗啉)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物85)的制备
Figure PCTCN2020132188-appb-000131
参考实施例2的合成方法,将吗啉替换为顺式2,6-二甲基吗啉,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((5-溴-2-((4-((2S,6R)-2,6-二甲基吗啉)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物85)。
1H NMR(400MHz,MeOD):δ8.89(s,3H),8.31(s,1H),8.02(s,1H),7.63–7.31(m,3H),6.88(s,1H),3.94–3.81(m,8H),3.05-3.03(m,2H),2.46-2.43(m,2H),2.18(s,3H),2.14(s,3H) 1.28-1.16(m,6H).
31P NMR(162.0MHz,MeOD)δ:53.32.
HPLC:96.01%@214nm,99.80%@254nm
LCMS:Rt:1.640min;MS m/z(ESI):692.1,694.1[M+H] +
实施例18、(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物86)的制备
Figure PCTCN2020132188-appb-000132
参考实施例2的合成方法,将吗啉替换为1-甲基-4-(哌啶-4-基)哌嗪,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((2,5-二氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲氧化膦(84-2),得到目标产物(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物86)。
1H NMR(400MHz,MeOD):δ9.00–8.91(m,1H),8.80(d,J=1.8Hz,1H),8.76(d,J=1.8Hz,1H),8.12(s,1H),7.92(s,1H),7.87(s,1H),7.48–7.44(m,2H),6.79(s,1H),3.89(s,3H),3.71(s,3H),3.15(d,J=11.5Hz,2H),2.94–2.38(m,10H),2.36–2.25(m,4H),2.14(s,3H),2.11(s,3H),1.96(d,J=11.5Hz,2H),1.85-1.57(m,2H).
31PNMR(162MHz,MeOD):δ53.27
LCMS:Rt:0.960min;MS m/z(ESI):716.3[M+H] +
实施例19、(6-((5-溴-2-((4-((1R,5S)-3-羟基-8-氮杂双环[3.2.1]辛烷-8-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物87)的制备
Figure PCTCN2020132188-appb-000133
参考实施例2的合成方法,将吗啉替换为去甲托品醇,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((5-溴-2-((4-((1R,5S)-3-羟基-8-氮杂双环[3.2.1]辛烷-8-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物87)。
1H NMR(400MHz,CDCl 3)δ12.72(s,1H),8.98-8.94(m,1H),8.75-8.69(m,2H),8.26-7.41(m, 6H),6.56(br,1H),4.18-3.62(m,7H),2.30-2.15(m,2H),2.13(s,3H),2.09(s,3H),2.00(s,3H),1.96-1.64(m,6H).
31P NMR(162MHz,CDCl 3)δ49.42(s).
HPLC:99.36%@214nm,99.54%@254nm
LCMS:Rt:1.593min;MS m/z(ESI):704.8,706.8[M+H] +
实施例20、(6-((2-((4-((1R,5S)-3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物88)的制备
Figure PCTCN2020132188-appb-000134
参考实施例2的合成方法,将吗啉替换为3-氧杂-8-氮杂-二环[3.2.1]辛烷,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((2-((4-((1R,5S)-3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物88)。
1H NMR(400MHz,MeOD):δ8.90(br,1H),8.82(d,J=2Hz,1H),8.78(d,J=1.6Hz,1H),8.23(s,1H),7.76-7.73(m,2H),7.57-7.48(m,2H),6.64(s,1H),3.89(s,3H),3.86(d,J=10.8Hz,2H),3.72(s,3H),3.62(s,2H),3.60-3.53(m,2H),2.16(s,3H),2.12(s,3H),1.93(br,4H)
31P NMR(162MHz,MeOD):δ52.86
LCMS:Rt:6.187min;MS m/z(ESI):690.1,692.1[M+H] +
实施例21、(6-((2-((4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物89)的制备
Figure PCTCN2020132188-appb-000135
参考实施例2的合成方法,将吗啉替换为8-氧杂-3-氮杂双环[3.2.1]辛烷,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((2-((4-(8-氧杂-3-氮杂双环[3.2.1]辛烷-3-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物89)。
1H NMR(400MHz,MeOD):δ8.85–8.80(m,3H),8.24(s,1H),7.83(s,1H),7.62(s,1H),7.56(br,1H),7.39(s,1H),6.88(s,1H),4.28(br,2H),3.91(s,3H),3.74(s,3H),2.95-2.92(m,2H),2.75-2.72(m,2H),2.15(s,3H),2.11(s,3H),1.97-1.84(m,4H)
31PNMR(162MHz,MeOD):δ52.85
LCMS:Rt:6.357min;MS m/z(ESI):690.1,692.1[M+H] +
实施例22、1-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)哌啶-4-基)哌嗪-1-基)乙烷-1-酮(化合物90)的制备
Figure PCTCN2020132188-appb-000136
化合物90-2:
Figure PCTCN2020132188-appb-000137
室温下将哌嗪-1-羧酸叔丁酯(8.0g,42.88mmol),4-氧代哌啶-1-羧酸苄酯(5.0g,21.44mmol),醋酸(1.9g,32.16mmol)分别加入到二氯乙烷(100mL)中,升温到50度搅拌反应5h。把反应体系冷却到室温,加入醋酸硼氢化钠(9.0g,42.88mmol),室温反应1.5h。加入氢氧化钠水溶液调剂pH至11,用二氯甲烷(100mL X 3)萃取,饱和食盐水洗涤,无水硫酸钠干燥,过滤,旋干,得粗品4-(1-((苄氧基)羰基)哌啶-4-基)哌嗪-1-羧酸叔丁酯(90-2)(7.0g,收率:81%),直接用于下一步。
LCMS:Rt:0.470min;MS m/z(ESI):404.2[M+H] +
化合物90-3:
Figure PCTCN2020132188-appb-000138
室温下将4-(1-((苄氧基)羰基)哌啶-4-基)哌嗪-1-羧酸叔丁酯(90-2)(7.0g,17.35mmol)溶解到二氧六环(10mL)中,再加入盐酸二氧六环(4M,50mL)室温搅拌反应16小时。旋干,得中间体4-(哌嗪-1-基)哌啶-1-羧酸苄酯(90-3)(10.0g),粗品直接用于下一步。
LCMS:Rt:0.450min;MS m/z(ESI):304.2[M+H] +
化合物90-4:
Figure PCTCN2020132188-appb-000139
室温下将4-(哌嗪-1-基)哌啶-1-羧酸苄酯(90-3)(6.0g,19.77mmol),三乙胺(6.0g,59.33mmol)加入到二氯甲烷(120mL)中搅拌5分钟。加入乙酸酐(4.0g,39.54mmol)室温反应2h。加入100ml水,用二氯甲烷(100mL X 2)萃取,饱和食盐水洗涤,无水硫酸钠干燥,过滤,旋干,得中间体4-(4-乙酰哌嗪-1-基)哌啶-1-羧酸苄酯(90-4)(7.0g,收率:100%),直接用于下一步。
LCMS:Rt:0.440min;MS m/z(ESI):346.2[M+H]。
化合物90-5:
Figure PCTCN2020132188-appb-000140
室温下将4-(4-乙酰哌嗪-1-基)哌啶-1-羧酸苄酯(90-4)(7.0g,20.26mmol),湿钯/碳(1.5g(Pd>=10%,H 2O wt%=50%))。加入到甲醇(100mL)中,加料完毕后,将反应置于氢气(~15psi)氛下,升温到60℃搅拌反应4小时。过滤,旋干,得中间体1-(4-(哌啶-4-基)哌嗪-1-基)乙酮(90-5)(4.0g,收率:93%)。
LCMS:Rt:0.350min;MS m/z(ESI):212.2[M+H] +
化合物90-6:
Figure PCTCN2020132188-appb-000141
室温下将1-(4-(哌啶-4-基)哌嗪-1-基)乙酮(90-5)(2.0g,9.46mmol),1-溴-2-氟-4-甲氧基-5-硝基苯(2.36g,9.46mmol)分别加入到乙腈(40mL)中,升温到50℃搅拌反应16小时。LCMS检测反应完全,旋干,硅胶柱(洗脱剂梯度:二氯甲烷/甲醇=20/1)纯化得中间体1-(4- (1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)哌嗪-1-基)乙酮(6)(2.0g,收率:48%)。
LCMS:Rt:0.833min;MS m/z(ESI):441.1[M+H] +
其余步骤参考实施例2,将(3-((2,5-二氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(2-1)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物1-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1-甲基-1H-吡唑-4-基)苯基)哌啶-4-基)哌嗪-1-基)乙烷-1-酮(化合物90)。
1H NMR(400MHz,CDCl 3):δ12.58(s,1H),9.01-8.97(m,1H),8.73(d,J=2.0Hz,1H),8.69(d,J=2.0Hz,1H),8.29(s,1H),8.21(s,1H),7.69(s,1H),7.61-7.56(m,2H),7.31(s,1H),6.70(s,1H),3.90(s,3H),3.75(s,3H),3.67-3.64(m,2H),3.51-3.48(m,2H),3.21(d,J=11.6Hz,2H),2.61-2.56(m,6H),2.34-2.27(m,1H),2.14(s,3H),2.11(s,6H),1.89(d,J=11.2Hz,2H),1.65-1.56(m,2H).
31P NMR(162.0MHz,CDCl 3):δ49.26.
LCMS:Rt:1.040min;MS m/z(ESI):788.1,790.1[M+H] +
实施例23、(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物91)的制备
Figure PCTCN2020132188-appb-000142
参考实施例15的合成方法,将3-氟-4-氧代哌啶-1-羧酸叔丁酯替换为N-叔丁氧羰基-4-哌啶酮,将(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)替换为(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4),得到目标化合物(3-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(化合物91)。
1H NMR(400MHz,CDCl 3)δ11.99(s,1H),9.91(d,J=4.8Hz,1H),8.27-8.25(m,2H),8.03(d,J=8.0Hz,1H),7.77(s,1H),7.67-7.56(m,4H),7.25(s,1H),6.66(s,1H),3.88(s,3H),3.76-3.72(m,7H),3.21-3.18(m,2H),2.58-2.53(m,5H),2.18(s,3H),2.15(s,3H),1.91(d,J=11.6Hz,2H),1.67-1.47(m,4H).
31P NMR(162MHz,CDCl 3)δ43.52.
LCMS:Rt:1.615min;MS m/z(ESI):746.2,748.2[M+H] +
实施例24、(6-((5-溴-2-((6-((2S,6R)-2,6-二甲基吗啉)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物92)的合成
Figure PCTCN2020132188-appb-000143
参考实施例13的合成方法,将1-甲基-4-(哌啶-4-基)哌嗪替换为顺式2,6-二甲基吗啉,得到目标化合物(6-((5-溴-2-((6-((2S,6R)-2,6-二甲基吗啉)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物92)。
1H NMR(400MHz,MeOD)δ8.81-8.77(m,3H),8.26(s,1H),8.10(s,1H),7.74(s,1H),7.59(s,1H),7.44(s,1H),3.99(s,3H),3.84(br,2H),3.71(s,3H),3.24(br,2H),2.51(t,J=11.2Hz,2H),2.16(s,3H),2.16(s,3H),1.15(s,3H),1.13(s,3H).
31P NMR(162MHz,MeOD)δ52.84.
LCMS:Rt:1.760min;MS m/z(ESI):693.1,695.1[M+H] +
实施例25、(6-((2-((6-((1R,5S)-3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物93)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000144
参考实施例13的合成方法,将1-甲基-4-(哌啶-4-基)哌嗪替换为3-氧杂-8-氮杂-二环[3.2.1]辛烷,得到目标化合物(6-((2-((6-((1R,5S)-3-氧杂-8-氮杂双环[3.2.1]辛烷-8-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物93)三氟乙酸盐。
1H NMR(400MHz,MeOD)δ8.87–8.71(m,3H),8.27(s,1H),7.78(br,3H),7.52(s,1H),4.03(s,2H),3.95(s,3H),3.88(d,J=10.5Hz,2H),3.82(s,3H),3.54(d,J=10.0Hz,2H),2.17(s,3H),2.13(s,3H),1.91(s,4H).
19F NMR(162MHz,MeOD)δ-77.42.
31P NMR(162MHz,MeOD)δ53.18.
LCMS:Rt:1.577min;MS m/z(ESI):691.1,693.1[M+H] +
实施例26、(6-((5-溴-2-((2-甲氧基-6-吗啉-5-(1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物94)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000145
参考实施例13的合成方法,将1-甲基-4-(哌啶-4-基)哌嗪替换为吗啉,将1-甲基-4-吡唑硼酸频哪醇酯替换为4-吡唑硼酸频哪醇酯,得到目标化合物(6-((5-溴-2-((2-甲氧基-6-吗啉-5-(1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物94)三氟乙酸盐。
1H NMR(400MHz,DMSO):δ12.82(s,1H),8.87-8.70(m,4H),8.30(s,1H),8.02-8.00(m,2H),7.86(s,1H),7.57(s,1H),3.88(s,3H),3.74-3.72(m,4H),3.09-3.07(m,4H),2.03(s,3H),2.00(s,3H).
19F NMR(376.5MHz,DMSO):δ-74.893
31P NMR(162.0MHz,DMSO):δ48.569
LCMS:Rt:1.491min;MS m/z(ESI):651.4,653.4[M+H] +
实施例27、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-吗啉代哌啶-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物95)的制备
Figure PCTCN2020132188-appb-000146
参考实施例13的合成方法,将1-甲基-4-(哌啶-4-基)哌嗪替换为4-(4-哌啶基)吗啉,得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-吗啉代哌啶-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物95)。
1H NMR(400MHz,MeOD):δ8.82-8.75(m,3H),8.25(s,1H),8.08(s,1H),7.77(s,1H),7.59(br,1H),7.43(s,1H),3.98(s,3H),3.74-3.72(m,7H),3.52-3.49(m,2H),2.79-2.73(m,2H),2.62(br,4H),2.32-2.26(m,1H),2.16(s,3H),2.12(s,3H),1.96(d,J=11.3Hz,2H),1.63-1.55(m,2H).
31P NMR(162MHz,MeOD):δ52.85(s).
LCMS:Rt:4.117min;MS m/z(ESI):748.1,750.1[M+H] +
实施例28、(6-((5-溴-2-((6-(3-氟-4-吗啉代哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物96)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000147
参考实施例13的合成方法,将1-甲基-4-(哌啶-4-基)哌嗪替换为4-(3-氟哌啶-4-基)吗啉盐酸盐(83-3),得到目标化合物(6-((5-溴-2-((6-(3-氟-4-吗啉代哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物96)三氟乙酸盐。
1H NMR(400MHz,CDCl 3)δ13.50(s,1H),8.79(s,1H),8.75(s,1H),8.64(d,J=5.4Hz,1H),8.05(s,1H),7.87-7.85(m,2H),7.58(d,J=9.4Hz,1H),7.52(s,1H),5.31–5.19(m,1H),4.06(s,3H),4.01–3.96(m,5H),3.93-3.85(m,5H),3.59–3.55(m,2H),3.50-3.47(m,2H),3.35–2.96(m,2H),2.35-2.33(m,1H),2.14(s,3H),2.11(s,3H),1.99-1.96(m,1H).
19F NMR(377MHz,CDCl 3)δ-75.83(s).-196.61(s),
31P NMR(162MHz,CDCl 3)δ51.16(s).
LCMS:Rt:1.003min;MS m/z(ESI):766.1,768.1[M+H] +
实施例29、(6-((5-溴-2-((6-(3-氟-4-(1,4-噁吖庚环-4-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物97)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000148
参考实施例15的合成方法,将吗啉替换为高吗啉,将1-溴-2-氟-4-甲氧基-5-硝基苯替换为2,3-二氯-6-甲氧基-5-硝基吡啶(4-2),得到目标化合物(6-((5-溴-2-((6-(3-氟-4-(1,4-噁吖庚环-4-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物97)三氟乙酸盐。
1H NMR(400MHz,MeOD)δ8.85–8.70(m,3H),8.26(d,J=7.4Hz,1H),8.17(s,1H),7.74(s,1H),7.57(s,1H),7.43(s,1H),5.30(d,J=50.5Hz,1H),4.00(s,3H),3.93(d,J=3.7Hz,2H),3.85(dd,J=13.1,7.3Hz,2H),3.78(s,1H),3.69(s,3H),3.59-3.51(s,6H),3.11(dd,J=40.1,14.2Hz,1H),3.10-2.89(m,1H),2.29-2.26(m,1H),2.17-2.09(m,9H).
19F NMR(162MHz,MeOD)δ-76.92,-201.27.
31P NMR(162MHz,MeOD)δ52.85.
LCMS:Rt:1.027min;MS m/z(ESI):780.1,782.1[M+H] +
实施例30、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(氧杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物98)的制备
Figure PCTCN2020132188-appb-000149
化合物98-2:
Figure PCTCN2020132188-appb-000150
室温下将哌嗪-1-羧酸叔丁酯(2.0g,10.73mmol),2,3-二氯-6-甲氧基-5-硝基吡啶(4-2)(2.39g,10.73mmol)分别加入到四氢呋喃(50mL)中,室温搅拌反应4h。反应液减压浓缩,硅胶柱纯化(石油醚/乙酸乙酯=5/1)得4-(3-氯-6-甲氧基-5-硝基吡啶-2-基)哌嗪-1-羧酸叔丁酯(98-2)(2.0g,收率:50%)。
LCMS:Rt:1.550min;MS m/z(ESI):373.0[M+H] +
化合物98-3:
Figure PCTCN2020132188-appb-000151
室温下将4-(3-氯-6-甲氧基-5-硝基吡啶-2-基)哌嗪-1-羧酸叔丁酯(98-2)(2.0g,5.36mmol),1-甲基-4-吡唑硼酸频哪醇酯(1.67g,8.04mmol),磷酸钾(3.41g,16.08mmol)分别加入到二氧六环(25mL)和水(2.5mL)中,氩气置换四次,加入双(二-叔丁基(4-二甲氨基苯基)膦)二氯化钯(II)(380mg,0.536mmol),氩气置换四次,升温到105℃搅拌反应4小时。反应 液减压浓缩,硅胶柱纯化(洗脱剂梯度:石油醚/乙酸乙酯=20/1)得中间体4-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌嗪-1-羧酸叔丁酯(98-3)(2.0g,收率:89%)。
LCMS:Rt:0.963min;MS m/z(ESI):419.1[M+H] +
化合物98-4:
Figure PCTCN2020132188-appb-000152
室温下将4-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌嗪-1-羧酸叔丁酯(98-3)(2.0g,4.78mmol)溶解到二氧六环(10mL)中,加入盐酸二氧六环溶液(4M,20mL),室温搅拌反应4小时。反应液减压浓缩至干,得中间体1-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌嗪(98-4)(2.0g)。
LCMS:Rt:0.880min;MS m/z(ESI):319.1[M+H] +
化合物98-5:
Figure PCTCN2020132188-appb-000153
室温下将1-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌嗪(98-4)(2.0g,6.28mmol),4-二甲氨基吡啶(766mg,6.28mmol)加入到二氯甲烷(20mL)中搅拌5分钟,再加入三氟乙酸酐(2.64g,12.56mmol)室温搅拌反应2小时,反应液直接减压浓缩至干,硅胶柱纯化,得中间体2,2,2-三氟-1-(4-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌嗪-1-基)乙酮(98-5)(2.0g,收率:77%)。
LCMS:Rt:1.253min;MS m/z(ESI):415.5[M+H] +
化合物98-6:
Figure PCTCN2020132188-appb-000154
室温下将2,2,2-三氟-1-(4-(6-甲氧基-3-(1-甲基-1H-吡唑-4-基)-5-硝基吡啶-2-基)哌嗪-1-基)乙酮(98-5)(2.0g,4.83mmol),湿钯/碳(500mg(Pd>=10%,H 2O wt%=50%)),分别加入到乙醇(30mL)中,加料完毕后,将反应置于氢气(~15psi)氛下,升温到55℃搅拌反应2小时。过滤除去催化剂,滤液减压浓缩至干,得中间体1-(4-(5-氨基-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(98-6)(1.7g,收率:92%)。
LCMS:Rt:1.560min;MS m/z(ESI):385.1[M+H] +
化合物98-7:
Figure PCTCN2020132188-appb-000155
室温下将1-(4-(5-氨基-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(98-6)(1.0g,2.6mmol),(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(859mg,2.08mmol),三氟乙酸(2.96g,26mmol)分别加入到异丙醇(30mL)中,氩气保护升温到95℃搅拌反应16小时,反应液浓缩,硅胶柱(洗脱剂梯度:二氯甲烷/甲醇=20/1)纯化得中间体得中间体1-(4-(5-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(98-7)(1.5g,收率:76%)。
LCMS:Rt:1.500min;MS m/z(ESI):762.1[M+H] +
化合物98-8:
Figure PCTCN2020132188-appb-000156
室温下将1-(4-(5-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(98-7)(500mg,0.657mmol),氢氧化钾(370mg,6.57mmol)分别加入到甲醇(20mL)和水(2mL)中,升温到60度搅拌反应5h。加入50mL二氯甲烷和30mL水稀释,用二氯甲烷(50mL X 3)萃取,有机相用无水硫酸钠干燥后浓缩至干,得中间体(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(98-8)(300mg,收率:69%)。
LCMS:Rt:0.843min;MS m/z(ESI):666.1[M+H] +
化合物98:
Figure PCTCN2020132188-appb-000157
室温下将(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(98-8)(300mg,0.451mmol),3-氧杂环丁酮(33mg,0.451mmol),醋酸(41mg,0.677mmol)分别加入到1,2-二氯乙烷(10mL)中。反应液加热至50℃搅拌反应5小时。加入三乙酰氧基硼氢化钠(191mg,0.902mmol),室温下反应1小时。将反应混合物用氢氧化钠水溶液调节pH至11,用二氯甲烷萃取,浓缩至干,残余物加入到N,N-二甲基甲酰胺(5mL)中,过滤后用高效液相制备色谱法(洗脱剂梯度参考实施例1)纯化,得到(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(氧杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦三氟乙酸盐。产物冻干后溶解在5mL的二氯甲烷中,用饱和碳酸钠(10mL X 2)洗涤,用水(10mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩至干,得到目标产物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(氧杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物98)(15mg,收率:4.64%)。
1H NMR(400MHz,MeOD):δ8.82(d,J=2.0Hz,1H),8.78-8.75(m,2H),8.25(s,1H),8.12(s,1H),7.73(s,1H),7.56(br,1H),7.43(s,1H),4.74-4.70(m,2H),4.66-4.62(m,2H),3.99(s,3H),3.70(s,3H),3.64-3.58(m,1H),3.17(br,4H),2.49(br,4H),2.16(s,3H),2.12(s,3H).
31P NMR(162.0MHz,MeOD):δ52.83.
LCMS:Rt:1.008min;MS m/z(ESI):720.1,722.1[M+H] +
实施例31、(6-((5-溴-2-((6-(3-氟-4-(4-甲基哌嗪-1-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物99)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000158
参考实施例15的合成方法,将吗啉替换为N-甲基哌嗪,将1-溴-2-氟-4-甲氧基-5-硝基苯替换为2,3-二氯-6-甲氧基-5-硝基吡啶(4-2),得到目标化合物(6-((5-溴-2-((6-(3-氟-4-(4-甲基哌嗪-1-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物99)三氟乙酸盐。
1H NMR(400MHz,CDCl 3)δ13.26(s,1H),8.80(d,J=1.8Hz,1H),8.76–8.65(m,2H),8.07(s,1H),7.81(d,J=13.8Hz,2H),7.65(d,J=8.9Hz,1H),7.47(s,1H),5.12-5.01(m,1H),3.97(s,3H),3.87(s,3H),3.69-3.65(m,1H),3.56(s,1H),3.34-3.22(m,6H),3.04-3.02(m,1H),2.92(d,J=14.2Hz,1H),2.88–2.80(m,6H),2.79-2.74(m,1H),2.20-2.18(m,1H),2.14(s,3H),2.11(s,3H),1.87-1.85(m,1H).
19F NMR(377MHz,CDCl 3)δ-75.81,-198.61,
31P NMR(162MHz,CDCl 3)δ50.78.
LCMS:Rt:0.946min;MS m/z(ESI):779.2,781.2[M+H] +
实施例32、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-吗啉代哌啶-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹啉-5-基)二甲基氧化膦(化合物100)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000159
化合物100-2:
Figure PCTCN2020132188-appb-000160
室温下将喹啉-6-胺(4.0g,27.78mmol)和四丁基溴化铵(13.39g,27.78mmol)加入二氯甲烷/甲醇(75ml/25ml)中。反应在室温下反应2小时。反应完全后将反应液用饱和硫代硫酸钠溶液调节pH至8左右,再用乙酸乙酯(80mL X 4)萃取。合并有机相,用饱和食盐水洗涤后加入无水硫酸钠干燥,减压浓缩后通过硅胶柱(石油醚:乙酸乙酯=8:1)洗脱,得到5-溴喹啉-6-胺(100-2)(4.0g,黄色固体)。
LCMS:Rt:0.860min;MS m/z(ESI):223.0[M+H] +
化合物100-3:
Figure PCTCN2020132188-appb-000161
室温下将化合物5-溴喹啉-6-胺(100-2)(1.0g,4.48mmol),二甲基氧磷(480mg,6.73mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(260mg,0.45mmol),醋酸钯(100mg,0.45mmol)和磷酸钾(1.43g,6.73mmol)溶于N,N-二甲基甲酰胺和水(40mL/8mL)中,将反应液加热至120℃搅拌反应16小时。将反应液冷却至室温,然后加入150mL水,用乙酸乙酯(50mL X 3)萃取,合并有机相,饱和食盐水洗涤并用无水硫酸钠干燥,通过硅胶柱(石油醚:乙酸乙酯=1:1)洗 脱得到化合物(6-氨基喹啉-5-基)二甲基氧化膦(100-3)(1.0g,黄色固体)。
化合物100-4:
Figure PCTCN2020132188-appb-000162
室温下将(6-氨基喹喔啉-5-基)二甲基氧化膦(100-3)(40mg,0.18mmol)溶于乙醇(5mL)中,再加入5-溴-2,4-二氯嘧啶(82mg,0.36mmol)和N,N-二异丙基乙胺(3.5g,27.1mmol)。反应液在氩气保护下加热至90℃搅拌反应16小时至反应完全。将反应液直接减压浓缩,残余物用硅胶板层析法(乙酸乙酯:石油醚=1:1)纯化,得到目标产物(6-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-5-基)二甲基氧化膦(100-4)(40mg,收率:54%)。
LCMS:Rt:1.245min;MS m/z(ESI):413.0[M+H] +
其余步骤参考实施例13的合成方法,将1-甲基-4-(哌啶-4-基)哌嗪替换为4-(4-哌啶基)吗啉,将(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-5-基)二甲基氧化膦(100-4),得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-吗啉代哌啶-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹啉-5-基)二甲基氧化膦(化合物100)三氟乙酸盐。
1H NMR(400MHz,MeOD):δ8.84(d,J=4.3Hz,1H),8.74(d,J=8.8Hz,1H),8.48–8.32(m,1H),8.26(s,1H),7.95(s,1H),7.75(s,1H),7.65(dd,J=8.8,4.3Hz,1H),7.56(s,1H),7.48(s,1H),4.14–4.11(m,2H),3.97(s,3H),3.80-3.74(m,2H),3.69(s,3H),3.66–3.49(m,4H),3.32(br,1H),3.13-3.09(m,2H),2.82(t,J=12.0Hz,2H),2.19-2.13(m,8H),1.86-1.78(m,2H).
19F NMR(377MHz,MeOD):δ-77.11.
31P NMR(162MHz,MeOD):δ47.37.
LCMS:Rt:3.797min;MS m/z(ESI):747.1,749.1[M+H] +
实施例33、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉代哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物101)的制备
Figure PCTCN2020132188-appb-000163
化合物101-2:
Figure PCTCN2020132188-appb-000164
室温下将4-(哌啶-4-基)吗啉(540mg,3.17mmol),1-溴-2-氟-4-甲氧基-5-硝基苯(793mg,3.17mmol)分别加入到乙腈(20mL)中,升温到50度搅拌反应16小时。将反应液减压浓缩至干,硅胶柱(洗脱剂梯度:二氯甲烷/甲醇=20/1)纯化得4-(1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)吗啉(101-2)(900mg,收率:71%)。
LCMS:Rt:0.843min;MS m/z(ESI):400.4[M+H] +
化合物101-3:
Figure PCTCN2020132188-appb-000165
室温下将4-(1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)吗啉(101-2)(800mg,1.99mmol),1-甲基吡唑-4-硼酸频哪醇酯(499mg,2.39mmol),碳酸铯(1.96g,5.99mmol)分别加入到二氧六环(20mL)和水(5mL)中,氩气置换四次,加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(280mg,0.39mmol),氩气置换四次,升温到105℃搅拌反应16小时。通过LCMS检测反应完全。反应液减压浓缩,硅胶柱(洗脱剂梯度:二氯甲烷/甲醇=20/1)纯化得中间体4-(1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)哌啶-4-基)吗啉(101-3)(450mg,收率:56%)。
LCMS:Rt:1.020min;MS m/z(ESI):402.1[M+H] +
化合物101-4:
Figure PCTCN2020132188-appb-000166
室温下将4-(1-(5-甲氧基-2-(1-甲基-1H-吡唑-4-基)-4-硝基苯基)哌啶-4-基)吗啉(101-3)(450mg,1.12mmol),湿钯/碳(200mg(Pd>=10%,H 2O wt%=50%)),分别加入到乙醇(20mL)中,加料完毕后,将反应置于氢气(~15psi)氛下,升温到50度搅拌反应2小时。用硅藻土过滤除去催化剂,滤液减压浓缩至干,得中间体2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉哌啶-1-基)苯胺(101-4)(200mg,收率:48%)。
LCMS:Rt:0.445min;MS m/z(ESI):372.1[M+H] +
化合物101:
Figure PCTCN2020132188-appb-000167
室温下将2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉哌啶-1-基)苯胺(101-4)(200mg,0.54mmol),(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9)(185mg,0.45mmol),三氟乙酸(614mg,5.38mmol)分别加入到异丙醇(20mL)中。反应液加热至95℃搅拌反应16小时。将反应混合物减压浓缩,残余物加入到N,N-二甲基甲酰胺(5mL)中,过滤后用高效液相制备色谱法(洗脱剂梯度参考实施例1)纯化,得到化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉代哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦三氟乙酸盐,产物冻干后溶解在5mL的二氯甲烷中,用饱和碳酸钠(10mL X 2)洗涤两次,后用水(10mL)洗涤,无水硫酸钠干燥,减压浓缩至干,得到目标产物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-4-(4-吗啉代哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物101)(141mg,收率:19%)。
1H NMR(400MHz,CDCl 3):δ12.58(s,1H),8.99(dd,J=4.4,9.6Hz,1H),8.84-8.70(m,2H),8.29(s,1H),8.22(s,1H),7.76(s,1H),7.58(br,1H),7.50(br,1H),7.32(s,1H),6.71(s,1H),3.90(s,3H),3.77(br,7H),3.21(d,J=11.2Hz,2H),2.59(s,6H),2.37-2.18(m,1H),2.14(s,3H),2.11(s,3H),1.95(d,J=10.8Hz,2H),1.63-1.57(m,2H).
31P NMR(162.0MHz,CDCl 3):δ49.29.
LCMS:Rt:1.019min;MS m/z(ESI):747.1,749.1[M+H] +
实施例34、(6-((2-((4-(4-(1,4-噁吖庚环-4-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物24)的制备
Figure PCTCN2020132188-appb-000168
参考实施例15的合成方法,将3-氟-4-氧代哌啶-1-羧酸叔丁酯替换为N-叔丁氧羰基-4-哌啶酮,将吗啉替换为高吗啉,得到目标化合物(6-((2-((4-(4-(1,4-噁吖庚环-4-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物24)。
1H NMR(400MHz,CDCl 3):δ12.59(s,1H),8.98-8.95(m,1H),8.74-8.71(m,2H),8.29(s,1H),8.21(s,1H),7.62-7.52(m,3H),7.33(s,1H),6.65(s,1H),3.95-3.80(m,7H),3.75(s,3H),3.27-2.62(m,10H),2.30-2.27(m,2H),2.15(s,3H),2.13(s,3H),2.06-1.91(m,3H)
31P NMR(162.0MHz,CDCl 3)δ:49.38.
LCMS:Rt:0.920min;MS m/z(ESI):761.1,763.1[M+H] +
实施例35、(6-((5-溴-2-((4-(4-(3,3-二氟吡咯烷-1-基)哌啶-1-基))-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物102)的制备
Figure PCTCN2020132188-appb-000169
参考实施例15的合成方法,将3-氟-4-氧代哌啶-1-羧酸叔丁酯替换为N-叔丁氧羰基-4-哌啶酮,将吗啉替换为3,3-二氟吡咯烷,得到目标化合物(6-((5-溴-2-((4-(4-(3,3-二氟吡咯烷-1-基)哌啶-1-基))-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物102)。
1H NMR(400MHz,MeOD):δ8.83(s,1H),8.77(d,J=1.9Hz,1H),8.26(s,1H),7.93(s,1H),7.80(s,1H),7.49-7.46(m,2H),7.36-7.34(m,1H),6.83(s,1H),3.91(s,3H),3.69(s,3H),3.24-3.21(m,3H),2.71-2.70(m,2H),2.55-2.53(m,4H),2.17-2.13(m,10H),1.86(s,2H).
31P NMR(162.0MHz,MeOD)δ:52.89.
LCMS:Rt:1.120min;MS m/z(ESI):767.1,769.1[M+H] +.
实施例36、(6-((5-溴-2-((4-(3-氟-4-(4-甲基哌嗪-1-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物103)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000170
参考实施例15的合成方法,将吗啉替换为N-甲基哌嗪,得到目标化合物(6-((5-溴-2-((4-(3-氟-4-(4-甲基哌嗪-1-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物103)三氟乙酸盐。
1H NMR(400MHz,CDCl 3)δ13.34(s,1H),10.66(s,1H),8.83(s,1H),8.76(d,J=1.6Hz,1H),8.73-8.68(m,1H),8.06(s,1H),7.93(s,1H),7.63(s,1H),7.52-7.47(m,2H),6.69(s,1H),5.40-5.22(m,1H),3.90(s,3H),3.89(s,3H),3.63-3.32(m,10H),3.24-3.15(m,1H),2.95-2.77(m,5H),2.31-2.23(m,1H),2.17(s,3H),2.13(s,3H),2.05–2.04(m,1H).
19F NMR(377MHz,CDCl 3)δ-75.88,-197.22.
31P NMR(162MHz,CDCl3)δ52.33.
LCMS:Rt:1.100min;MS m/z(ESI):778.1,780.1[M+H] +
实施例37、(6-((5-溴-2-((4-(3,3-二氟-4-(吡咯烷-1-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑- 4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物104)的制备
Figure PCTCN2020132188-appb-000171
参考实施例15的合成方法,将3-氟-4-氧代哌啶-1-羧酸叔丁酯替换为3,3-二氟-4-氧代哌啶-1-羧酸叔丁酯,将吗啉替换为吡咯烷,得到目标化合物(6-((5-溴-2-((4-(3,3-二氟-4-(吡咯烷-1-基)哌啶-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物104)。
1H NMR(400MHz,MeOD)δ:8.83-8.77(m,3H),8.27(s,1H),8.02(s,1H),7.90(s,1H),7.52-7.48(m,1H),7.30(s,1H),6.85(s,1H),3.93(s,3H),3.70(s,3H),3.13-2.97(m,2H),2.85-2.69(m,7H),2.17(s,3H),2.13(s,3H),2.10-1.93(m,2H),1.84-1.76(m,4H).
19F NMR(376.5MHz,MeOD)δ:-101.05,-101.69.
31P NMR(162.0MHz,MeOD)δ:52.89.
LCMS:Rt:4.102min;MS m/z(ESI):767.2,769.2[M+H] +.
实施例38、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡咯-3-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物36)的制备
Figure PCTCN2020132188-appb-000172
参考实施例11的合成方法,将1-甲基吡唑-4-硼酸频哪醇酯替换为1-甲基吡咯-3-硼酸频哪醇酯,将(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡咯-3-基)-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物36)。
1H NMR(400MHz,CDCl 3):δ12.53(s,1H),9.00(dd,J=9.5,4.2Hz,1H),8.71(d,J=1.8Hz,1H),8.67(d,J=1.8Hz,1H),8.27(s,1H),8.14(s,1H),7.54(br,1H),7.23(s,1H),7.01(s,1H),6.66(s,1H),6.31(s,1H),6.24(s,1H),3.87(s,3H),3.48(s,3H),3.30(d,J=11.0Hz,2H),2.66-2.52(m,10H),2.32-225(m,4H),2.13(s,3H),2.09(s,3H),1.95-1.92(m,2H),1.65-1.62(m,2H).
31PNMR(162MHz,CDCl 3):δ49.08
LCMS:Rt:0.960min;MS m/z(ESI):759.5,761.5[M+H] +
实施例39、6-((5-溴-2-((2-甲氧基-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)-5-(噻吩-3-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物45)的制备
Figure PCTCN2020132188-appb-000173
参考实施例11的合成方法,将1-甲基吡唑-4-硼酸频哪醇酯替换为3-噻吩硼酸,将(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物6-((5-溴-2-((2-甲氧基-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)-5-(噻吩-3-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物45)。
1H NMR(400MHz,CDCl 3)δ12.48(s,1H),8.88-8.85(m,1H),8.72-8.68(m,2H),8.27(s,1H),8.20(s,1H),7.60-7.58(m,1H),7.39(d,J=4.8Hz,1H),7.30(s,1H),7.26(m,1H),7.03(s,1H),6.64(s,1H),3.90(s,3H),3.16-3.13(m,2H),2.71-2.51(m,10H),2.34(s,3H),2.22(s,1H),2.13(s,3H),2.09(s,3H),1.88-1.85(m,2H),1.58-1.50(m,2H).
31P NMR(162MHz,CDCl 3)δ59.38.
LCMS:Rt:2.036min;MS m/z(ESI):762.2,764.2[M+H] +
实施例40、(6-((5-溴-2-((5-(呋喃-3-基)-2-甲氧基-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物54)的制备
Figure PCTCN2020132188-appb-000174
参考实施例11的合成方法,将1-甲基吡唑-4-硼酸频哪醇酯替换为3-呋喃硼酸,将(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((5-溴-2-((5-(呋喃-3-基)-2-甲氧基-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物54)。
1H NMR(400MHz,CDCl 3):δ12.50(s,1H),8.90-887(m,1H),8.74(d,J=1.6Hz,1H),8.70(d,J=2.0Hz,1H),8.28(s,1H),8.19(s,1H),7.69(s,2H),7.33(s,1H),7.03(s,1H),6.69(s,1H),6.55(s,1H),3.90(s,3H),3.20(d,J=12Hz,2H),2.71-2.56(m,10H),2.33(s,3H),2.32-2.45(m,1H),2.14(s,3H),2.11(s,3H),1.93(d,J=11.2Hz,2H),1.67-1.59(m,2H).
31P NMR(162.0MHz,CDCl 3):δ49.15.
LCMS:Rt:1.090min;MS m/z(ESI):746.1,748.1[M+H] +
实施例41、(6-((5-溴-2-((4-甲氧基-6-(4-(4-甲基哌嗪-1-基)哌啶-1-基)-[1,1’-联苯基]-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)-二甲基氧化膦(化合物105)的制备
Figure PCTCN2020132188-appb-000175
参考实施例11的合成方法,将1-甲基吡唑-4-硼酸频哪醇酯替换为苯硼酸,将(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((5-溴-2-((4-甲氧基-6-(4-(4-甲基哌嗪-1-基)哌啶-1-基)-[1,1’-联苯基]-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)-二甲基氧化膦(化合物105)。
1H NMR(400MHz,CDCl 3)δ:12.45(s,1H),8.83(d,J=5.5Hz,1H),8.70(d,J=16.8Hz,2H),8.26(s,1H),8.11(s,1H),7.56(s,1H),7.46(d,J=7.2Hz,2H),7.32(s,1H),7.07–7.00(m,3H),6.64(s,1H),3.92(s,3H),3.13-3.10(m,2H),2.76–2.37(m,10H),2.30(s,3H),2.21-2.14(m,1H),2.12(s,3H),2.08(s,3H),1.77-1.74(m,2H),1.48–1.34(m,2H).
31P NMR(162.0MHz,CDCl 3)δ:49.12.
LCMS:Rt:4.157min;MS m/z(ESI):756.2,758.2[M+H] +
实施例42、(6-((5-溴-2-((2-甲氧基-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)-5-(吡啶-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物106)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000176
参考实施例11的合成方法,将1-甲基吡唑-4-硼酸频哪醇酯替换为4-吡啶硼酸,将(3-((5-溴-2-氯嘧啶-4-基)氨基)喹啉-4-基)二甲基氧化膦(1-4)替换为(6-((5-溴-2-氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(4-9),得到目标产物(6-((5-溴-2-((2-甲氧基-4-(4-(4-甲基哌嗪-1-基)哌啶-1-基)-5-(吡啶-4-基)苯基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物106)三氟乙酸盐。
1H NMR(400MHz,MeOD)δ8.82(d,J=2.0Hz,1H),8.76(d,J=2.0Hz,1H),8.68(br,1H),8.25(s,1H),8.20(d,J=4.4Hz,2H),8.06(s,1H),7.70(d,J=5.2Hz,2H),7.53(br,1H),6.85(s,1H),3.96(s,3H),3.15-2.98(m,9H),2.76(s,3H),2.72-2.66(m,4H),2.15(s,3H),2.11(s,3H),1.89(d,J=10.8Hz,2H),1.55-1.52(m,2H).
19F NMR(376.5MHz,MeOD):δ-76.88
31P NMR(162MHz,MeOD):δ52.74
LCMS:Rt:3.347min;MS m/z(ESI):757.1,759.1[M+H] +
实施例43、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(四氢-2H-吡喃-4-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物108)的制备
Figure PCTCN2020132188-appb-000177
化合物108:
Figure PCTCN2020132188-appb-000178
室温下将(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(98-8)(100mg,0.15mmol),四氢吡喃酮(30mg,0.30mmol),冰醋酸(14mg,0.225mmol)分别加入到1,2-二氯乙烷(8mL)中。反应液在氩气氛下加热至50℃搅拌反应3小时,降温至室温,再加入三乙酰氧基硼氢化钠(64mg,0.30mmol),室温下反应1h。将反应混合物用氢氧化钠水溶液调节pH≈7,用二氯甲烷(5ml X 3)萃取。有机相减压浓缩至干,残余物加入到DMF(5mL)中,过滤后用高效液相制备色谱法(洗脱剂梯度:
Figure PCTCN2020132188-appb-000179
)纯化,得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(四氢-2H-吡喃- 4-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物108)(15mg,收率:14%)。
1H NMR(400MHz,MeOD):δ8.82(d,J=2.0Hz,1H),8.77(d,J=2.0Hz,2H),8.25(s,1H),8.12(s,1H),7.73(s,1H),7.56(br,1H),7.46(s,1H),4.03-4.02(m,2H),3.99(s,3H),3.70(s,3H),3.48-3.40(m,2H),3.17-3.15(m,4H),2.73(br,4H),2.53-2.47(m,1H),2.16(s,3H),2.13(s,3H),1.91(d,J=10.8Hz,2H),1.61-1.50(m,2H).
31P NMR(162.0MHz,MeOD):δ52.88.
LCMS:Rt:1.076min;MS m/z(ESI):748.0,750.0[M+H] +
实施例44、(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(氧杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物109)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000180
化合物109-2:
Figure PCTCN2020132188-appb-000181
室温下将1-(4-(5-氨基-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(98-6)(200mg,0.52mmol),(6-((2,5-二氯嘧啶-4-基)氨基)喹喔啉-5-基)二甲氧化膦(84-2)(155mg,0.42mmol)和三氟乙酸(593mg,5.2mmol)分别加入到异丙醇(10mL)中,氩气保护升温到95℃搅拌反应16小时。反应液直接旋干,用薄层制备色谱(二氯甲烷:甲醇=20:1)制备得中间体1-(4-(5-((5-氯-4-((5-(二甲基磷基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(109-2)(350mg,收率:94%)。
LCMS:Rt:1.388min;MS m/z(ESI):716.5[M+H] +
化合物109-3:
Figure PCTCN2020132188-appb-000182
室温下将1-(4-(5-((5-氯-4-((5-(二甲基磷基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)-2,2,2-三氟乙烷-1-酮(109-2)(350mg,0.488mmol),氢氧化钾(273mg,4.88mmol)分别加入到甲醇(25mL)和水(2mL)中,升温到60度搅拌反应5h。反应液直接旋干,用薄层制备色谱(二氯甲烷:甲醇=20:1)制备得中间体(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(109-3)(150mg,收率:50%)。
LCMS:Rt:0.903min;MS m/z(ESI):620.6[M+H] +
化合物109的三氟乙酸盐:
Figure PCTCN2020132188-appb-000183
室温下将(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(109-3)(150mg,0.242mmol),3-氧杂环丁酮(35mg,0.484mmol),醋酸(30mg,0.484mmol)分别加入到1,2-二氯甲烷(15mL)中。反应液在氩气氛下加热至50℃搅拌反应5小时。降温到室温,再加入三乙酰氧基硼氢化钠(103mg,0.484mmol),室温下反应1h。将反应混合物用氢氧化钠水溶液调节pH≈7,用二氯甲烷萃取,有机相旋干,残余物加入到DMF(5mL)中,过滤后用高效液相制备色谱法(洗脱剂梯度:参考实施例1)纯化,得到目标标题化合物(6-((5-氯-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(氧杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物109)三氟乙酸盐(10mg,收率:6.1%)。
1H NMR(400MHz,MeOD):δ8.89-8.88(m,1H),8.86(d,J=2.0Hz,1H),8.81(d,J=2.0Hz,1H),8.21(s,1H),8.18(s,1H),7.84(s,1H),7.62-7.60(m,1H),7.54(s,1H),4.95-4.82(m,4H),4.56-4.49(m,1H),4.02(s,3H),3.77(s,3H),3.48-3.13(m,8H),2.17(s,3H),2.13(s,3H).
19F NMR(376.5MHz,CD 3OD):δ-77.23.
31P NMR(162.0MHz,MeOD):δ53.45.
LCMS:Rt:1.007min;MS m/z(ESI):676.0[M+H] +
实施例45、(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(1-甲基氮杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物110)的制备
Figure PCTCN2020132188-appb-000184
化合物110-2:
Figure PCTCN2020132188-appb-000185
室温下将(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(98-8)(100mg,0.15mmol),3-氧代氮杂环丁-1-羧酸叔丁酯(51mg,0.30mmol),冰醋酸(14mg,0.225mmol)分别加入到1,2-二氯乙烷(10mL)中,升温到50℃搅拌反应2小时。降温到室温,加入三乙酰氧基硼氢化钠(64mg,0.30mmol),室温下搅拌反应14h。饱和碳酸钠溶液调节pH≈7,用二氯甲烷萃取(20mL X 3),有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后滤液减压浓缩,用薄层制备色谱(二氯甲烷:甲醇=20:1)制备得中间体叔丁基3-(4-(5-((5-溴-4-((5-(二甲基磷基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)氮杂环丁烷-1-羧酸酯(110-2)(100mg,收率:81%)。
LCMS:Rt:1.222min;MS m/z(ESI):819.7,821.7[M+H] +
化合物110-3:
Figure PCTCN2020132188-appb-000186
室温下将叔丁基3-(4-(5-((5-溴-4-((5-(二甲基磷基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-6-甲氧基-3-(1-甲基-1H-吡唑-4-基)吡啶-2-基)哌嗪-1-基)氮杂环丁烷-1-羧酸酯(110-2)(100mg,0.121mmol)加入到盐酸二氧六环(1M,1.2mL,1.22mmol)溶液中,室温搅拌反应1h。反应液减压旋干,加入DCM(10mL)再次减压旋干,得到中间体(6-((2-((6-(4-(氮杂环丁-3-基)哌嗪-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(110-3)粗品,(100mg,收率:100%),直接用于下一步反应。
LCMS:Rt:1.050min;MS m/z(ESI):719.1,721.1[M+H] +
化合物110:
Figure PCTCN2020132188-appb-000187
室温下将(6-((2-((6-(4-(氮杂环丁-3-基)哌嗪-1-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)-5-溴嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(110-3)(80mg,0.111mmol),甲醛水溶液(1M,1.2mL,1.11mmol),三乙胺(111mg,0.111mmol)分别加入到甲醇(3mL)中。室温下搅拌反应5小时,再加入三乙酰氧基硼氢化钠(118mg,0.555mmol),室温下反应11h。将反应混合物用氢氧化钠水溶液调节pH≈7,用二氯甲烷萃取(20mL X 3),有机相旋干,残余物加入到DMF(5mL)中,过滤后滤液用高效液相制备色谱法(洗脱剂梯度:参考实施例43)纯化,得到目标化合物(6-((5-溴-2-((2-甲氧基-5-(1-甲基-1H-吡唑-4-基)-6-(4-(1-甲基氮杂环丁-3-基)哌嗪-1-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物110)(4mg,收率:4.9%)。
1H NMR(400MHz,MeOD):δ8.82(d,J=1.6Hz,1H),8.77(d,J=2.0Hz,2H),8.26(s,1H),8.12(s,1H),7.75(s,1H),7.56(br,1H),7.42(s,1H),3.99(s,3H),3.71(s,3H),3.60(t,J=5.4Hz,2H),3.14(br,4H),3.11-3.01(m,3H),3.46(br,4H),2.39(s,3H),2.16(s,3H),2.13(s,3H).
31P NMR(162.0MHz,MeOD):δ52.880.
LCMS:Rt:1.115min;MS m/z(ESI):733.4,735.4[M+H] +
实施例46、(6-((5-溴-2-((6-(2-(二甲氨基)-7-氮杂螺[3.5]壬烷-7-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物111)三氟乙酸盐的制备
Figure PCTCN2020132188-appb-000188
化合物111-2:
Figure PCTCN2020132188-appb-000189
室温下将2-氧代-7-氮杂螺[3.5]壬烷-7-羧酸叔丁酯(200mg,0.837mmol),二甲胺(0.5mL,1.0mmol,2M in THF)溶于甲醇(5mL)中,加入冰醋酸(75mg,1.255mmol),室温条件下反应2小时,缓慢加入三乙酰氧基硼氢化钠(354mg,1.67mmol,2.0eq),加入完毕后将反应在室温下搅拌16小时。TLC(乙酸乙酯/石油醚=1/2)显示反应完全,将反应液倒入饱和碳酸氢钠水溶液(50mL)中,用二氯甲烷(100mL X 3)萃取,饱和氯化钠水溶液(50mL)洗涤,无水硫酸钠干燥,过滤后滤液浓缩,得到目标产物2-(二甲氨基)-7-氮杂螺[3.5]壬烷-7-羧酸叔丁酯(111-2)(200mg,粗品),直接用于下一步。
LCMS:Rt=0.920min;MS m/z(ESI):269.2[M+H] +
化合物111-3:
Figure PCTCN2020132188-appb-000190
室温下将2-(二甲氨基)-7-氮杂螺[3.5]壬烷-7-羧酸叔丁酯(111-2)(200mg,0.746mmol)溶入无水二氯甲烷(4mL)中,加入三氟乙酸(0.5mL,4.386mmol),在室温条件下反应2小时。LCMS检测反应完全,将反应液浓缩,得到目标产物N,N-二甲基-7-氮杂螺[3.5]壬烷-2-胺(111-3)(0.60g粗品),直接用于下一步。
LCMS:Rt=0.310min;MS m/z(ESI):169.2[M+H] +
化合物111-4:
Figure PCTCN2020132188-appb-000191
室温下将粗品N,N-二甲基-7-氮杂螺[3.5]壬烷-2-胺(111-3)(0.60g,3.55mmol),2,3-二氯-6-甲氧基-5-硝基吡啶(165mg,0.744mmol)溶于N,N二甲基甲酰胺(6mL)中,加入碳酸钾(308mg,2.232mmol),二异丙基乙胺(192mg,1.488mmol)。在50℃下搅拌反应16小时,LCMS检测反应完全。将反应液过滤浓缩,所得残余物通过硅胶板色谱层析法(洗脱剂梯度:二氯甲烷:甲醇=10:1)纯化,得到目标产品7-(3-氯-6-甲氧基-5-硝基吡啶-2-基)-N,N-二甲基-7-氮杂螺[3.5]壬烷-2-胺(111-4)(220mg,收率:83.7%)。
LCMS:Rt=1.107min;MS m/z(ESI):355.1[M+H] +
其余步骤参考实施例13的合成方法,将1-(1-(3-氯-6-甲氧基-5-硝基吡啶-2-基)哌啶-4-基)-4-甲基哌嗪(13-2)替换为7-(3-氯-6-甲氧基-5-硝基吡啶-2-基)-N,N-二甲基-7-氮杂螺[3.5]壬烷-2-胺(111-4),得到目标产物(6-((5-溴-2-((6-(2-(二甲氨基)-7-氮杂螺[3.5]壬烷-7-基)-2-甲氧基-5-(1-甲基-1H-吡唑-4-基)吡啶-3-基)氨基)嘧啶-4-基)氨基)喹喔啉-5-基)二甲基氧化膦(化合物111)三氟乙酸盐。
1H NMR(400MHz,MeOD)δ8.84(d,J=1.9Hz,1H),8.80(d,J=1.9Hz,1H),8.74(brs,1H),8.27(s,1H),7.99(s,1H),7.76(s,1H),7.63(brs,1H),7.50(s,1H),3.98(s,3H),3.74-3.67(m,4H),3.13-3.08(m,2H),3.07-3.01(m,2H),2.81(s,6H),2.44-2.38(m,2H),2.16(s,3H),2.13(s,3H),1.98-1.96(m,2H),1.80-1.67(m,4H).
31P NMR(376.5MHz,MeOD)δ53.02.
19F NMR(162MHz,MeOD)δ-77.10.
LCMS:Rt:4.377min;MS m/z(ESI):746.1,748.1[M+H] +
参考上述实施例的合成方法,本发明还合成了以下化合物,其表征数据为:
Figure PCTCN2020132188-appb-000192
Figure PCTCN2020132188-appb-000193
Figure PCTCN2020132188-appb-000194
Figure PCTCN2020132188-appb-000195
Figure PCTCN2020132188-appb-000196
Figure PCTCN2020132188-appb-000197
Figure PCTCN2020132188-appb-000198
Figure PCTCN2020132188-appb-000199
生物学活性及相关性质测试例
以下测试例中的化合物均根据本申请上述实施例的方法制备获得,测试例化合物可以是游离碱或其三氟乙酸盐形式,根据上文实施例制得的产物具体形式确定。
以下测试例中“NA”表示未测试。
测试例1:酶学EGFR抑制实验
实验材料:
HTRF KinEASE-TK kit购自于CisBio(France)。
384-well assay plate板购自于greiner bio-one(Germany)。
384-well source plate板购自于LABCYTE(USA)。
MgCl 2,MnCl 2,DTT,TritonX-100,HEPES,BSA购自于Sigma(USA)。
EGFR WT-WT,EGFR WT-del19/T790M/C797S,EGFR WT-L858R/T790M/C797S,EGFR WT-del19/T790M,EGFR WT-L858R/T790M参考Nature.2016;534(7605):129–132的方法表达和纯化。
实验仪器:
自动微孔移液器:Precision PRC384U(BioTek,USA)
纳升级声波移系统:
Figure PCTCN2020132188-appb-000200
HANDLE RS(LABCYTE,USA)
多标记检测分析仪:Envision Multilabel Reader(PerkinElmer,USA)
实验方法:
1.配制激酶缓冲溶液(1x Enzyme buffer,1mM MnCl2,1mM MgCl2,1mM DTT,0.01%BSA)。
2.化合物的准备。化合物的检测终浓度为10μM,使用DMSO配制成100倍浓度,即1mM。使用自动微孔移液器(Precision PRC384U)将待测化合物做4倍梯度稀释,获得浓度从1mM到3.8nM的10个剂量浓度,将待测化合物转移到384孔板中,每孔100nL化合物。
3.激酶反应。使用激酶缓冲液配制2X酶反应溶液(WT-WT:3nM,WT-d19/TM/CS:1nM,WT-LR/TM/CS:0.1nM,WT-LR/TM:0.5nM,WT-d19/TM:1nM)转移5μl酶反应溶液到384孔板反应孔中,阴性对照孔加入5μl激酶缓冲液。450rpm振荡混匀,室温下静置孵育10分钟。使用激酶缓冲液配制2X底物/ATP溶液(ATP浓度:WT-WT:4.8μM,WT-d19/TM/CS:4μM,WT-LR/TM/CS:0.24μM,WT-LR/TM:0.86μM,WT-d19/TM:3.8μM),转移5μl底物/ATP溶液到384孔板反应孔中起始反应,450rpm振荡混匀,室温下孵育60分钟。
4.终止反应。向384孔反应板中加入10μl反应终止液(Streptavidin-XL665,TK Antibody-Cryptate),1000rpm离心1分钟,室温下放置60分钟,Envision读板。
5.曲线拟合。从Envision程序上复制发光读数的数据,将发光读数665nM/615nM的比值通过公式转换为抑制百分率:Percent inhibition=(max-signal ratio)/(max-min)*100。“min”为不加酶进行反应的对照样荧光读数比值;“max”为加入DMSO作为对照的样品荧光读数比值。将数据导入MS Excel并使用XLFit excel add-in version 5.4.0.8进行曲线拟合:Y=Bottom+(Top-Bottom)/(1+(IC50/X)^HillSlope)。
本发明化合物的酶学活性抑制IC 50如下所示(del19简写为d19,T790M简写为TM,L858R简写为LR,C797S简写为CS)。
Figure PCTCN2020132188-appb-000201
测试例2:BaF3细胞抗增殖实验
实验材料:
RPMI1640购自Gibco(USA)。
FBS购自Excell(China)。
IL-3 Recombinant Mouse Protein购自Gibco(USA)。
Cell
Figure PCTCN2020132188-appb-000202
Luminescent Cell Viability kit购自Promega(USA)。
BaF3细胞购自Riken(Japan)。
BaF3/EGFR-WT,BaF3/EGFR-Del19/T790M/C797S细胞由中美冠科生物技术(太仓)有限公司构建。
BaF3培养基:RPMI1640+10%FBS+8ng/ml IL-3Recombinant Mouse Protein。
BaF3/EGFR-WT,BaF3/EGFR-Del19/T790M/C797S培养基:RPMI1640+10%FBS。
读板仪器:EnVision(PerkinElmer,USA)。
实验方法:
调整细胞浓度,96孔板每孔种2000个BaF3,BaF3/EGFR-WT或者BaF3/EGFR-Del19/T790M/C797S细胞,90μl体积,用DMSO配制1000X待测化合物母液,并用DMSO将待测化合物做3倍梯度稀释(共9个浓度),再用相应细胞培养基做100倍稀释后,每孔加 入10μl(10X浓度)的待测化合物(每个化合物每个浓度三个复孔),5%CO 2,37℃培养72小时。72小时后,每孔加入50μl Cell Titer-Glo,在摇板仪上2min混匀,孵育10min后,用EnVision读数。
数据分析:
细胞活性用GraphPad Prism version 5作图,曲线用nonlinear regression model with a sigmoidal dose response拟合,由此计算IC50数据。测试结果见下表。
Figure PCTCN2020132188-appb-000203
Figure PCTCN2020132188-appb-000204
测试例3:PC9 EGFR-Del19/T790M/C797S及HEK293T细胞增殖实验
实验材料:
RPMI1640购自Gibco(USA)。
DMEM购自Gibco(USA)。
FBS购自Gibco(USA)。
Puromycin购自Invitrogen(USA)。
胰酶购自Invitrogen(USA)。
DMSO购自Sigma(USA)。
Cell Counting Kit-8(CCK-8)细胞增殖毒性检测试剂盒购自东仁化学科技(上海)有限公司。
PC9 EGFR-Del19/T790M/C797S细胞由中美冠科生物技术(太仓)有限公司构建。
HEK293T细胞购自ATCC(USA)。
PC9 EGFR-Del19/T790M/C797S培养基:RPMI1640+10%FBS+0.5ug/mL puromycin。
HEK293T培养基:DMEM+10%FBS。
读板仪器:EnVision(PerkinElmer,USA)。
实验方法:
细胞培养2-3天至融合度达到80-90%。使用胰酶消化细胞,离心重悬后计数,调整细胞浓度至1000个每孔(HEK293T)及3000个每孔(PC9EGFR-Del19/T790M/C797S),90μl体积,5%CO 2,37℃培养过夜。用DMSO配制200X待测化合物母液,并用DMSO将待测化合物做3倍梯度稀释(共8个浓度),再用无血清培养基做20倍稀释后,每孔加入10μl(10X浓度)的待测化合物(每个化合物每个浓度两个复孔),5%CO 2,37℃培养72小时。72小时后,每孔加入10μl CCK-8,离心混匀后,37℃培养1-4小时,在摇板仪上2min混匀,孵育10min后,用EnVision读取OD450。
数据分析:
根据以下公式计算抑制率:
Inhibition%=(OD cell control–OD sample)/(OD cell control–OD media control)*100
使用IDBS Xlfit Model 205进行4参数拟合计算IC50数据
测试结果如下表所示:
Figure PCTCN2020132188-appb-000205
Figure PCTCN2020132188-appb-000206
Figure PCTCN2020132188-appb-000207
测试例4:小鼠药代动力学实验
实验材料:
CD-1小鼠购自斯贝福(北京)生物技术有限公司。
DMSO、MC(甲基纤维素)、Tween80购自Sigma(USA)。
乙腈购自Merck(USA)。
HP-β-CD购自日本食品化工株氏会社。
实验方法:
雌性CD-1小鼠6只(20-30g,4-6周,每组三只),随机分成2组,每组3只。第1组尾静脉注射给予化合物82,剂量为1mg/kg,溶媒5%DMSO in 10%HP-β-CD in water,第2组口服给予化合物82,剂量5mg/kg,溶媒0.5%MC/0.25%Tween80in water。动物实验前正常喂食喂水。每组小鼠于给药前及给药后0.083(仅静脉注射组)、0.25、0.5、1、2、4、6、8和24h进行静脉采血。收集的全血样品置于K2EDTA抗凝管中,离心5min后(4000rpm,4℃)取血浆待测。
取小鼠血浆样品30μL,补3μL空白溶剂,加入200μL乙腈溶剂(其中含内标化合物)沉淀蛋白,涡旋0.5min后,离心(4700rpm,4℃)15min,上清液用水稀释3倍,进样10μL于LC-MS/MS系统(AB Sciex Triple Quad 5500)进行定量检测。在测定样品浓度时随行CD-1小鼠血浆标准曲线(线性范围:0.5-1000ng/mL)和质控样品。对10x稀释样品,取3μL样品加入27μL的空白血浆;3x稀释样品,取10μL样品加入20μL空白血浆,其余处理步骤同不稀释样品。
PK测试结果如下所示:
Figure PCTCN2020132188-appb-000208
Figure PCTCN2020132188-appb-000209

Claims (36)

  1. 式(I)化合物或其药学上可接受的盐,
    Figure PCTCN2020132188-appb-100001
    其中,
    R 1选自苯基和5-6元杂芳基,所述苯基和5-6元杂芳基任选被R 7取代;
    或R 1
    Figure PCTCN2020132188-appb-100002
    其中R 11、R 12独立地选自C 1-4烷基,所述C 1-4烷基任选地被羟基或C 1-4烷氧基取代;
    R 2选自C 1-6烷基、C 4-14环烷基、4-14元杂环烷基、苯基和5-6元杂芳基,所述C 1-6烷基、C 4-14环烷基、4-14元杂环烷基、苯基和5-6元杂芳基任选被R 8取代;
    U、V、W独立地选自CR 6和N;
    环A选自苯基和5-6元杂芳基;
    R 3选自H、卤素、氰基、C 1-4烷基和C 1-4烷氧基;
    R 4、R 5独立地选自C 1-4烷基和C 1-4烷氧基;
    R 6独立地选自H、C 1-4烷基、C 1-4烷氧基和卤素;
    R 7独立地选自卤素、羟基、氨基、C 1-4烷基和C 1-4烷氧基,所述C 1-4烷基和C 1-4烷氧基任选地被羟基取代;
    R 8独立地选自卤素、OH、C 1-6烷基、C 1-6烷氧基、氧代、氨基、C 3-8环烷基和3-8元杂环烷基,所述氨基、C 3-8环烷基和3-8元杂环烷基任选地被R 9取代;
    R 9独立地选自卤素、C 1-4烷基、C 1-4烷氧基、C 1-4酰基和氧代。
  2. 权利要求1所述的式(I)化合物,其中,R 1选自5-6元杂芳基,所述5-6元杂芳基任选被R 7取代。
  3. 权利要求2所述的式(I)化合物,其中,R 1选自5元杂芳基,所述5元杂芳基任选被R 7取代。
  4. 权利要求3所述的式(I)化合物,其中,R 1选自苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、四唑基和三唑基,所述苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、吡唑基、四唑基和三唑基任选被R 7取代。
  5. 权利要求4所述的式(I)化合物,其中,R 1选自苯基、吡啶基、吡咯基、呋喃基、噻吩 基、咪唑基、吡唑基和三唑基,所述苯基、吡啶基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基和三唑基任选被R 7取代。
  6. 权利要求1-5任一项所述的式(I)化合物,其中,R 7独立地选自卤素、羟基和C 1-4烷基,所述C 1-4烷基任选地被羟基取代。
  7. 权利要求6所述的式(I)化合物,其中,R 7独立地选自C 1-4烷基,所述C 1-4烷基任选地被羟基取代。
  8. 权利要求7所述的式(I)化合物,其中,R 7独立地选自甲基和羟乙基。
  9. 权利要求1所述的式(I)化合物,其中,R 1选自苯基、
    Figure PCTCN2020132188-appb-100003
    Figure PCTCN2020132188-appb-100004
  10. 权利要求1所述的式(I)化合物,其中,R 1
    Figure PCTCN2020132188-appb-100005
    其中R 11选自C 1-4烷基、R 12选自任选被羟基取代的C 1-4烷基。
  11. 权利要求10所述的式(I)化合物,其中,R 11为甲基、R 12为甲基或羟乙基。
  12. 权利要求1-11任一项所述的式(I)化合物,其中,R 2选自C 5-12环烷基和5-12元杂环烷基,所述C 5-12环烷基和5-12元杂环烷基任选被R 8取代。
  13. 权利要求12所述的式(I)化合物,其中,R 2为5-12元杂环烷基,所述5-12元杂环烷基任选被R 8取代。
  14. 权利要求13所述的式(I)化合物,其中,R 2选自哌啶基、四氢吡喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基、氮杂环庚烷基、氧杂环庚烷基、硫杂环庚烷基、3,9-二氮杂螺[5.5]十一烷基、2,9-二氮杂螺[5.5]十一烷基、
    Figure PCTCN2020132188-appb-100006
    所述哌啶基、四氢吡喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基、1,4-二噻烷基、氮杂环庚烷基、氧杂环庚烷基、 硫杂环庚烷基、3,9-二氮杂螺[5.5]十一烷基、2,9-二氮杂螺[5.5]十一烷基、
    Figure PCTCN2020132188-appb-100007
    Figure PCTCN2020132188-appb-100008
    任选被R 8取代。
  15. 权利要求14所述的式(I)化合物,其中,R 2选自哌啶基、哌嗪基、吗啉基、3,9-二氮杂螺[5.5]十一烷基、
    Figure PCTCN2020132188-appb-100009
    所述哌啶基、哌嗪基、吗啉基、3,9-二氮杂螺[5.5]十一烷基、
    Figure PCTCN2020132188-appb-100010
    任选被R 8取代。
  16. 权利要求1-15任一项所述的式(I)化合物,其中,R 8独立地选自卤素、OH、C 1-4烷基、氨基、C 3-7环烷基和3-7元杂环烷基,所述氨基、C 3-7环烷基和3-7元杂环烷基任选地被R 9取代。
  17. 权利要求16所述的式(I)化合物,其中,R 8独立地选自F、OH、甲基、氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基、氮杂环丁烷基,所述氨基、哌嗪基、1,3-噁嗪烷基、1,4-二氮杂环庚烷基、1,3-氧杂氮杂环庚烷基、1,4-氧杂氮杂环庚烷基、氧杂环丁烷基、吗啉基、吡咯烷基、四氢吡喃基、氮杂环丁烷基任选地被R 9取代。
  18. 权利要求1-17任一项所述的式(I)化合物,其中,R 9独立地选自卤素、C 1-4烷基、C 1-4酰基、C 1-4烷氧基和氧代。
  19. 权利要求18所述的式(I)化合物,其中,R 9独立地选自F、甲基、乙酰基和氧代。
  20. 权利要求1-19任一项所述的式(I)化合物,其中,R 2选自
    Figure PCTCN2020132188-appb-100011
    Figure PCTCN2020132188-appb-100012
  21. 权利要求1-20任一项所述的式(I)化合物,其中,U为CR 6
  22. 权利要求21所述的式(I)化合物,其中,R 6独立地选自C 1-4烷氧基。
  23. 权利要求22所述的式(I)化合物,其中,R 6为甲氧基。
  24. 权利要求1-23任一项所述的式(I)化合物,其中,V选自CH和N。
  25. 权利要求1-24任一项所述的式(I)化合物,其中,W选自CH和N。
  26. 权利要求1-25任一项所述的式(I)化合物,其中,环A选自苯基、吡咯基、吡唑基、咪唑基、吡啶基、吡嗪基和哒嗪基。
  27. 权利要求26所述的式(I)化合物,其中,环A选自苯基和吡嗪基。
  28. 权利要求1-27任一项所述的式(I)化合物,其中,
    Figure PCTCN2020132188-appb-100013
    选自
    Figure PCTCN2020132188-appb-100014
  29. 权利要求28所述的式(I)化合物,其中,
    Figure PCTCN2020132188-appb-100015
    选自
    Figure PCTCN2020132188-appb-100016
  30. 权利要求1-29任一项所述的式(I)化合物,其中,R 3选自卤素和C 1-4烷基。
  31. 权利要求30所述的式(I)化合物,其中,R 3选自Cl和Br。
  32. 权利要求1-31任一项所述的式(I)化合物,其中,R 4、R 5独立地选自C 1-3烷基。
  33. 权利要求32所述的式(I)化合物,其中,R 4、R 5为甲基。
  34. 权利要求1所述的式(I)化合物,选自以下化合物:
    Figure PCTCN2020132188-appb-100017
    Figure PCTCN2020132188-appb-100018
    Figure PCTCN2020132188-appb-100019
    Figure PCTCN2020132188-appb-100020
    Figure PCTCN2020132188-appb-100021
    Figure PCTCN2020132188-appb-100022
  35. 药物组合物,其包含权利要求1-34任一项所述的式(I)化合物或其药学上可接受的盐和药学上可接受的辅料。
  36. 权利要求1-34任一项所述的式(I)化合物或其药学上可接受的盐、或权利要求35所述的药物组合物在制备预防或者治疗EGFR介导的疾病的药物中的用途。
PCT/CN2020/132188 2019-11-29 2020-11-27 作为egfr激酶抑制剂的多芳基化合物 WO2021104441A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080081283.0A CN114728932A (zh) 2019-11-29 2020-11-27 作为egfr激酶抑制剂的多芳基化合物

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201911205549 2019-11-29
CN201911205549.3 2019-11-29
CN202010063931.1 2020-01-20
CN202010063931 2020-01-20
CN202010523861 2020-06-10
CN202010523861.3 2020-06-10

Publications (1)

Publication Number Publication Date
WO2021104441A1 true WO2021104441A1 (zh) 2021-06-03

Family

ID=76129129

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/132188 WO2021104441A1 (zh) 2019-11-29 2020-11-27 作为egfr激酶抑制剂的多芳基化合物

Country Status (2)

Country Link
CN (1) CN114728932A (zh)
WO (1) WO2021104441A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022262857A1 (zh) * 2021-06-17 2022-12-22 微境生物医药科技(上海)有限公司 芳基氧膦类化合物
WO2023020600A1 (zh) * 2021-08-19 2023-02-23 贝达药业股份有限公司 Egfr抑制剂的盐、晶型及其组合物和应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009143389A1 (en) * 2008-05-21 2009-11-26 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
WO2012051587A1 (en) * 2010-10-14 2012-04-19 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in egfr-driven cancers
WO2012151561A1 (en) * 2011-05-04 2012-11-08 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in egfr-driven cancers
WO2019015655A1 (zh) * 2017-07-19 2019-01-24 正大天晴药业集团股份有限公司 作为egfr激酶抑制剂的芳基磷氧化合物
WO2020216371A1 (zh) * 2019-04-26 2020-10-29 江苏先声药业有限公司 Egfr抑制剂及其应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009143389A1 (en) * 2008-05-21 2009-11-26 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
WO2012051587A1 (en) * 2010-10-14 2012-04-19 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in egfr-driven cancers
WO2012151561A1 (en) * 2011-05-04 2012-11-08 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in egfr-driven cancers
WO2019015655A1 (zh) * 2017-07-19 2019-01-24 正大天晴药业集团股份有限公司 作为egfr激酶抑制剂的芳基磷氧化合物
WO2020216371A1 (zh) * 2019-04-26 2020-10-29 江苏先声药业有限公司 Egfr抑制剂及其应用

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022262857A1 (zh) * 2021-06-17 2022-12-22 微境生物医药科技(上海)有限公司 芳基氧膦类化合物
WO2023020600A1 (zh) * 2021-08-19 2023-02-23 贝达药业股份有限公司 Egfr抑制剂的盐、晶型及其组合物和应用

Also Published As

Publication number Publication date
CN114728932A (zh) 2022-07-08

Similar Documents

Publication Publication Date Title
JP6496376B2 (ja) 阻害剤化合物
CN113166103B (zh) Egfr抑制剂及其应用
CN112334451A (zh) 作为激酶抑制剂的杂环化合物
US10280170B2 (en) Substituted 4,5,6,7-tetrahydro-pyrazolo[1,5-a]pyrazine derivatives and 5,6,7,8-tetrahydro-4H-pyrazolo[1,5-a][1,4]diazepine derivatives as Ros1 inhibitors
KR20160116033A (ko) 2,4-이치환 페닐-1,5-디아민 유도체, 이의 응용, 및 이로 제조한 약물 조성물
KR20120044966A (ko) 1,2,4-트리아졸로〔1,5a〕 피리딘 유도체의 제조 및 용도
CN114502536A (zh) 作为激酶抑制剂的杂环化合物
EP3640247B1 (en) Syk inhibitor and use method therefor
EP3053923B1 (en) Triazolopyrazine derivatives as tyrosin kinase inhibitors
TWI580679B (zh) 雜芳基並嘧啶類衍生物、其製備方法和用途
JP2019507766A (ja) 線維症の治療のための新規化合物及びその医薬組成物
JP2017508779A5 (zh)
JP2024505732A (ja) ピリドピリミジノン系誘導体及びその製造方法と使用
WO2021139775A1 (zh) 吡啶酮化合物及应用
WO2021104441A1 (zh) 作为egfr激酶抑制剂的多芳基化合物
CN114340634A (zh) 作为激酶抑制剂的杂环化合物
CN116888108B (zh) 新型egfr降解剂
JP2022511236A (ja) 置換キナゾリノン誘導体、及びmGluR4のポジティブアロステリック調節剤としてのその使用
WO2021244502A1 (zh) 多芳基化合物及应用
KR20240019071A (ko) N2-페닐피리미딘-2,4-디아민 화합물, 및 그 제조 방법 및 사용 방법
CN113348170A (zh) 联苯类衍生物抑制剂、其制备方法和应用
JP7440710B1 (ja) G12d変異krasタンパクに作用する複素環化合物
WO2022127807A1 (zh) 一种芳基磷氧化物类衍生物自由碱的晶型及其制备方法和应用
CN116891502A (zh) Egfr降解剂
TW202300152A (zh) Egfr抑制劑

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: 20894216

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: 20894216

Country of ref document: EP

Kind code of ref document: A1