WO2023169547A1 - DNA聚合酶θ抑制剂及其用途 - Google Patents

DNA聚合酶θ抑制剂及其用途 Download PDF

Info

Publication number
WO2023169547A1
WO2023169547A1 PCT/CN2023/080706 CN2023080706W WO2023169547A1 WO 2023169547 A1 WO2023169547 A1 WO 2023169547A1 CN 2023080706 W CN2023080706 W CN 2023080706W WO 2023169547 A1 WO2023169547 A1 WO 2023169547A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
membered
cycloalkyl
heteroatoms
ring
Prior art date
Application number
PCT/CN2023/080706
Other languages
English (en)
French (fr)
Inventor
李瑶
张国彪
王耀羚
刘漫
郑登宇
王正
王顺
钟启杰
唐平明
余彦
张晨
严庞科
Original Assignee
四川海思科制药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 四川海思科制药有限公司 filed Critical 四川海思科制药有限公司
Publication of WO2023169547A1 publication Critical patent/WO2023169547A1/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to an inhibitor of DNA polymerase ⁇ and its use in preparing and treating tumors.
  • DNA polymerase ⁇ is a unique multifunctional DNA polymerase. Mammalian cells have evolved multiple pathways to repair DNA double-strand breaks (DSBs) to ensure genome stability.
  • DNA polymerase theta is a key component of the alternative end joining (alt-EJ) pathway, also known as the microhomology-mediated end joining (MMEJ) pathway, involved in DNA double-strand break repair. MMEJ is another repair pathway of cells in addition to non-homologous end joining (NHEJ) and homologous recombination (HR).
  • DNA polymerase ⁇ (Pol ⁇ ) is important for repairing DNA breaks, especially in homologous recombination deficient (HRD) cells.
  • HRD including defects in the BRCA1 and BRCA2 genes, is a clinically important feature of many important types of tumors, including breast, ovarian, prostate, and pancreatic cancer.
  • Pol ⁇ is highly expressed and directs DSB repair toward alt-EJ, turning on microhomology. Mediated end joining (MMEJ) DNA repair process.
  • MMEJ Mediated end joining
  • Pol ⁇ leads to cell death through accumulation of toxic RAD51 intermediates and inhibition of the alt-EJ repair pathway.
  • Pol ⁇ also has the function of reverse transcribing RNA and promoting DNA repair using RNA as a template [7].
  • Pol ⁇ is barely expressed in normal tissues, but is highly expressed in various tumor types (such as breast cancer, ovarian cancer, HNSCC, and lung cancer). At the same time, homologous recombination repair deficiency (HRD) is common in these tumor types; therefore, there is a theoretical basis for the application of Pol ⁇ inhibitors in these tumor types.
  • HRD homologous recombination repair deficiency
  • Pol ⁇ inhibitors have the potential to be used as monotherapy and in combination with PARP inhibitors to treat multiple types of tumors, and their unique mechanisms of action may help solve two types of PARP resistance.
  • Other clinical patient groups may also benefit from combination treatment regimens with Pol ⁇ inhibitors, including potentially with chemotherapy, radiation therapy, and immuno-oncology agents.
  • the present invention provides a Pol ⁇ inhibitor with novel structure and good efficacy, which is used to treat tumor-related diseases and has excellent effects of high bioavailability and low toxic and side effects.
  • the present invention relates to a formula (Ic), (Ia), formula (I), formula (Ib), (Id), (Ie), (If), formula (II), formula (II-a), ( II-d), (II-e), formula (Ib-1), (II-a-1), (II-e-1) or (II-b) or (I-1-a) Compounds, stereoisomers, deuterates or pharmaceutically acceptable salts thereof,
  • D ring is selected from
  • X 1 and X 2 are each independently selected from O or S; or X 2 does not exist; in some embodiments, X 1 and X 2 are each independently selected from O; X 3 is selected from O or NR x3 ;
  • X 4 is selected from O or NH or NR x 4 ; in some embodiments, X 4 is selected from O; in some embodiments, X 4 is selected from NH; in some embodiments, X 4 is selected from NR x 4 ;
  • R x3 is independently selected from COR
  • s is selected from 0, 1, 2, 3, 4, or 5; in some embodiments, s is selected from 0, 1, 2, 3, or 4; in some embodiments, s is selected from 0, 1, 2, or 3 ; In some embodiments, s is selected from 0, 1, or 2; In some embodiments, s is selected from 1 or 2; In some embodiments, s is selected from 1; In some embodiments, s is selected from 2 ;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. Ring; in some embodiments, Ring A is an 8-10 membered bicyclic heteroatom containing 1-3 heteroatoms selected from N, S, and O; in some embodiments, Ring A is an 8-10 membered bicyclic heterocyclic ring containing 1, 2, or 3 A 5-membered monocyclic heteroaryl group selected from N, S, and O heteroatoms, containing 1, 2, or 3 6-membered monocyclic heteroaryl groups selected from N, S, and O heteroatoms, containing 1, 2, or 3 An 8-membered bicyclic heteroatom selected from N, S, and O heteroatoms, a 9-membered bicyclic heteroacyclic ring containing 1, 2, or 3 heteroatoms selected from N, S, and O, or a 9-membered bicyclic
  • Ring A 8-membered bicyclic heteroacyclic ring from N, S, O heteroatoms; in some embodiments, Ring A is thiazolyl, pyrazolyl, In some embodiments, Ring A is In some embodiments, Ring A is or In some embodiments, Ring A is or In some embodiments, Ring A is In some embodiments, Ring A is In some embodiments, Ring A is In some embodiments, Ring A is
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O.
  • connection order of ring B is from left to right;
  • Ring B is phenyl; in some embodiments, Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, Pyridyl, pyrazinyl, pyridazinyl, pyrimidinyl, In a In some embodiments, Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl ,
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl,
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O; in certain embodiments, Ring C is a 5-6-membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, 5-membered monocyclic heteroaryl group with S and O heteroatoms;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;In certain embodiments, R 1 and R 2 are independently N 3 , OH or methoxy; In certain embodiments, R 1 and R 2 are independently OH;
  • R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring; in certain embodiments, R 1 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring; in certain embodiments
  • R 1 and the attached carbon atom form a 5- or 6-membered heterocyclic ring; in certain embodiments, R 1 and the attached carbon atom form a piperidine ring; in certain embodiments, R 2 and the attached carbon atom form a piperidine ring; in certain embodiments, R 2 and the attached carbon atom form a piperidine ring.
  • the connected carbon atoms form a 4-6 membered heterocyclic ring; in certain embodiments, R 2 and the connected carbon atoms form a 5- or 6-membered heterocyclic ring;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Alkynyl and C 3-6 cycloalkyl group substitutions; in certain embodiments, R 3 is C 1-4 alkyl or 4-6 containing 1-3 heteroatoms selected from N, S, O One-membered heterocycloalkyl, the alkyl and heterocycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl Substituted with a group; in certain embodiments, R 3 is
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group; in certain embodiments, R 3 and B ring form a piperidine ring or a tetrahydropyrrole ring;
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl; in certain embodiments, R 4 and R 5 are independently H or C 1-4 alkyl; in certain embodiments, R 4 and R 5 are independently H or methyl;
  • n and m are each independently selected from 0, 1, 2 or 3; in certain embodiments, n and m are each independently 0 or 1; in certain embodiments, n is 0 and m is 0 or 1; In certain embodiments, m is 0 and n is 0 or 1; in certain embodiments, n is 0 and m is 0;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, - C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms) base), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -C(O)-C 3-6 cycloalkyl or - (CH 2 ) r -C 3-6 cycloalkyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl and cycloalkyl groups are optionally selected from 1 to 3 Substituted from halogen, D, CN, OH, C 1-4 alkoxy, C
  • each R 6 is independently halogen, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl , C 1-4 alkoxy, C 2-4 alkynyl , morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl , tetrahydrofuryl, tetrahydropyranyl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl or isoxazolyl, the alkyl group , Alkoxy, alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuranyl, tetrahydropyranyl, cyclopropyl, cyclobutyl, pyrazolyl, Furyl, pyrrolyl, thienyl, imid
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R a is independently selected from H, D, C 1-4 alkyl, C 3-6 cycloalkyl;
  • r is selected from 0, 1, 2 or 3;
  • D is selected from D1 or D4, and the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkylphenyl, 5-membered heteroarylcene containing 1-3 heteroatoms selected from N, S, O benzo containing 1-3 heteroatoms selected from N, S, O 5-membered heterocycloalkyl, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 5-6-membered heteroaryl groups selected from N, S, and O heteroatoms , 5-6 membered cycloalkyl and containing 1-3 heteroatoms selected from N, S, O 5-membered heteroaryl, 5-6 membered cycloalky
  • Or ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • D is selected from D2, and the compound meets one of the following conditions:
  • R 6 is not halogen, C 1-4 alkyl, C 2-4 alkenyl, unsubstituted C3-6 cycloalkyl, unsubstituted containing 1-3 4-7 membered heterocycloalkyl groups selected from N, S, O heteroatoms, -O-CH 3 or OH, and ring B is not
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, Contains 1-3 heterogens selected from N, S, and O 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl, containing 1-3 selected from N, S, O heteroatoms, 5-6 membered heterocycloalkylacene containing 1-3 A 5-membered heteroaryl acene group selected from N, S, and O heteroatoms, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 selected from N, A 5-6-membered heteroaryl group and a 5-6-membered cycloalkyl group containing S and O heteroatoms and containing 1-3 5-membered heteroaryl groups and a 5-6-membered cycloalkyl group selected from N, S and O heteroatoms Acene group, a 5-membered heteroaryl 5-6-membered heterocycloalkyl
  • Or ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • R 6 is selected from R
  • R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C3-6 cycloalkyl, when r is selected from 0, the heterocycloalkyl and cycloalkyl are further replaced by 1-3 selected from halogen, D, CN, OH, C1-4 alkoxy, C1 -4 alkyl, C3-6 cycloalkyl and NH 2 group substitutions.
  • the present invention relates to a compound represented by formula (Ic), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • D ring is selected from
  • X 1 and X 2 are each independently selected from O or S, or X2 does not exist;
  • X 3 is selected from O or NR x3 ;
  • X 4 is selected from O or NH or NR x4 ;
  • R x3 is independently selected from COR
  • the alkyl, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are any The selected one is substituted by 1-3 groups selected from halogen, D, OH, CN, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • z is selected from 0 or 1;
  • s is selected from 0, 1, 2, 3, 4 or 5;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O.
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;
  • R 1 or R 2 forms a 4-6 membered heterocyclic ring with the carbon atom to which they are connected;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Substitution of alkynyl and C 3-6 cycloalkyl groups;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n and m are each independently selected from 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, - C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R a is independently selected from H, D, C 1-4 alkyl, C 3-6 cycloalkyl;
  • r is selected from 0, 1, 2 or 3.
  • the present invention relates to a compound represented by formula (Ic), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • D ring is selected from
  • X 1 and X 2 are each independently selected from O or S;
  • X 3 is selected from O or NR x3 ;
  • X 4 is selected from O or NH or NR x4 ;
  • R x3 is independently selected from COR
  • the alkyl, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are any The selected one is substituted by 1-3 groups selected from halogen, D, OH, CN, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • z is selected from 0 or 1;
  • s is selected from 0, 1, 2, 3, 4 or 5;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O.
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;
  • R 1 or R 2 forms a 4-6 membered heterocyclic ring with the carbon atom to which they are connected;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Substitution of alkynyl and C 3-6 cycloalkyl groups;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n and m are each independently selected from 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 selected from N, S, O heteroatoms 4-7 membered heterocycloalkyl), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O), -(CH 2 ) r -C 3-6 cycloalkyl, -C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl are optionally replaced by 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R a is independently selected from H, D, C 1-4 alkyl, C 3-6 cycloalkyl;
  • r is selected from 0, 1, 2 or 3.
  • the present invention relates to a compound represented by formula (Ic), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • D ring is selected from
  • X 1 and X 2 are each independently selected from O or S;
  • X 3 is selected from O or NR x3 ;
  • X 4 is selected from O or NH or NR x4 ;
  • R x3 is independently selected from COR
  • the alkyl, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are any The selected one is substituted by 1-3 groups selected from halogen, D, OH, CN, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • z is selected from 0 or 1;
  • s is selected from 0, 1, 2, 3, 4 or 5;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O.
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;
  • R 1 or R 2 forms a 4-6 membered heterocyclic ring with the carbon atom to which they are connected;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Substitution of alkynyl and C 3-6 cycloalkyl groups;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n and m are each independently selected from 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, - C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R a is independently selected from H, D, C 1-4 alkyl, C 3-6 cycloalkyl;
  • r is selected from 0, 1, 2 or 3;
  • D is selected from D1 or D4, and the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkylphenyl, 5-membered heteroarylcene containing 1-3 heteroatoms selected from N, S, O benzo containing 1-3 heteroatoms selected from N, S, O 5-membered heterocycloalkyl, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 5-6-membered heteroaryl groups selected from N, S, and O heteroatoms , 5-6 membered cycloalkyl and containing 1-3 heteroatoms selected from N, S, O 5-membered heteroaryl, 5-6 membered cycloalky
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • D is selected from D2, and the compound meets one of the following conditions:
  • R 6 is not halogen, C 1-4 alkyl, C 2-4 alkenyl, unsubstituted C 3-6 cycloalkyl, unsubstituted Contains 1-3 4-7-membered heterocycloalkyl groups selected from N, S, O heteroatoms, -O-CH 3 or OH, and ring B is not
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkyl acene group 5-membered heteroaryl acene group containing 1-3 heteroatoms selected from N, S, and O
  • 5-membered heteroaryl acene group containing 1-3 heteroatoms selected from N, S, and O Heteroaryl and contains 1-3 5-6-membered heteroaryl selected from N, S, O heteroatoms, 5-6-membered cycloalkyl and contains 1-3 selected from N, S, O heteroatoms 5-membered heteroaryl, 5-6-membered cycloalkylocene, 5-membered heteroaryla 5-6-member
  • R 6 is selected from R
  • R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, r is selected from 0, the heterocycloalkyl and cycloalkyl are further replaced by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy , C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 group substitutions.
  • the present invention relates to a compound represented by formula (Ic), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • D ring is selected from
  • X 1 and X 2 are each independently selected from O or S;
  • X 3 is selected from O or NR x3 ;
  • X 4 is selected from O or NH or NR x4 ;
  • R x3 is independently selected from COR
  • the alkyl, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are any The selected one is substituted by 1-3 groups selected from halogen, D, OH, CN, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • z is selected from 0 or 1;
  • s is selected from 0, 1, 2, 3, 4 or 5;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O.
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;
  • R 1 or R 2 forms a 4-6 membered heterocyclic ring with the carbon atom to which they are connected;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Substitution of alkynyl and C 3-6 cycloalkyl groups;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n and m are each independently selected from 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, - C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R a is independently selected from H, D, C 1-4 alkyl, C 3-6 cycloalkyl;
  • r is selected from 0, 1, 2 or 3;
  • the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • At least one R6 is not halogen and C1-4 alkyl
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • the present invention relates to a compound represented by formula (Ic), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • D ring is selected from
  • X 1 and X 2 are each independently selected from O or S;
  • X 3 is selected from O or NR x3 ;
  • X 4 is selected from O or NH
  • R x3 is independently selected from COR
  • z is selected from 0 or 1;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O. Containing 1-3 heteroatoms selected from N, S, O 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl, containing 1-3 selected from N, S, O heteroatoms, 5-6 membered heterocycloalkylacene, containing 1-3 selected A 5-membered heteroarylcene acene group containing 1-3 heteroatoms selected from N, S, O, and a 5-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N , 5-membered heteroaryl group of S and O heteroatoms and containing 1-3 selected from N, S, O heteroatoms, 5-6-membered cycloalkyl group and containing 1-3 selected A 5-membered heteroaryl group selected from N, S, and O heteroatoms, benzo containing 1-3 5-membered heteroaryl groups selected from N, S
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;
  • R 1 or R 2 forms a 4-6 membered heterocyclic ring with the carbon atom to which they are connected;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Substitution of alkynyl and C 3-6 cycloalkyl groups;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n and m are each independently selected from 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, - C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R a is independently selected from H, D, C 1-4 alkyl, C 3-6 cycloalkyl;
  • r is selected from 0, 1, 2 or 3;
  • the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkylphenyl, 5-membered heteroarylcene containing 1-3 heteroatoms selected from N, S, O benzo containing 1-3 heteroatoms selected from N, S, O 5-membered heterocycloalkyl, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 5-6-membered heteroaryl groups selected from N, S, and O heteroatoms , 5-6 membered cycloalkyl and containing 1-3 heteroatoms selected from N, S, O 5-membered heteroaryl, 5-6 membered cycloalky
  • At least one R6 is not halogen and C1-4 alkyl
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • the present invention relates to a compound represented by formula (I-1-a), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • X 1 and X 2 are each independently selected from O or S;
  • X 3 is selected from O or NR x3 ;
  • R x3 is independently selected from COR
  • z is selected from 0 or 1;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O. ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O.
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently N 3 , OH or C 1-4 alkoxy, which is optionally substituted by 1 to 3 groups selected from D, halogen, OH, NH 2 and CN ;
  • R 1 or R 2 forms a 4-6 membered heterocyclic ring with the carbon atom to which they are connected;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or C 3 -6 cycloalkyl, the alkyl, alkenyl, alkynyl, heterocycloalkyl and cycloalkyl are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 Substitution of alkynyl and C 3-6 cycloalkyl groups;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 4a , R 5a , R 4 and R 5 are each independently selected from H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n and m are each independently selected from 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-7 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r - (5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, - C(O)-C 3-6 cycloalkyl or phenyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl, cycloalkyl and phenyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 al
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • r is selected from 0, 1, 2 or 3;
  • the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkylphenyl, 5-membered heteroarylcene containing 1-3 heteroatoms selected from N, S, O benzo containing 1-3 heteroatoms selected from N, S, O 5-membered heterocycloalkyl, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 5-6-membered heteroaryl groups selected from N, S, and O heteroatoms , 5-6 membered cycloalkyl and containing 1-3 5-membered heteroaryl selected from N, S, O heteroatoms, 5-6 membered cycloalky
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • (x)z is selected from 1;
  • the present invention is a compound represented by formula (I-c), its stereoisomer, deuterated compound or pharmaceutically acceptable salt, D is selected from D1 or D4, and the compound satisfies one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkylphenyl, 5-membered heteroarylcene containing 1-3 heteroatoms selected from N, S, O benzo containing 1-3 heteroatoms selected from N, S, O 5-membered heterocycloalkyl, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 5-6-membered heteroaryl groups selected from N, S, and O heteroatoms , 5-6 membered cycloalkyl and containing 1-3 heteroatoms selected from N, S, O 5-membered heteroaryl, 5-6 membered cycloalky
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • the present invention is a compound represented by formula (I-c), its stereoisomer, deuterated compound or pharmaceutically acceptable salt,
  • D is selected from D1 or D4, and the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl or alkynyl and heterocycloalkyl optionally substituted with 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl; or R 3 and B ring form 5-6 One-membered heterocycloalkyl;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkylphenyl, 5-membered heteroarylcene containing 1-3 heteroatoms selected from N, S, O benzo containing 1-3 heteroatoms selected from N, S, O 5-membered heterocycloalkyl, containing 1-3 5-membered heteroaryl groups selected from N, S, and O heteroatoms and containing 1-3 5-6-membered heteroaryl groups selected from N, S, and O heteroatoms , 5-6 membered cycloalkyl and containing 1-3 heteroatoms selected from N, S, O 5-membered heteroaryl, 5-6 membered cycloalky
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • the present invention is a compound represented by formula (I-c), its stereoisomer, deuterated compound or pharmaceutically acceptable salt, wherein,
  • D is selected from D2, and the compound meets one of the following conditions:
  • R 6 is not halogen, C 1-4 alkyl, C 2-4 alkenyl, unsubstituted C3-6 cycloalkyl, unsubstituted containing 1-3 4-7 membered heterocycloalkyl groups selected from N, S, O heteroatoms, -O-CH 3 or OH, and ring B is not
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O
  • One-membered heterocycloalkyl acene group 5-membered heteroaryl acene group containing 1-3 heteroatoms selected from N, S, and O
  • 5-membered heteroaryl acene group containing 1-3 heteroatoms selected from N, S, and O Heteroaryl and contains 1-3 5-6-membered heteroaryl selected from N, S, O heteroatoms, 5-6-membered cycloalkyl and contains 1-3 selected from N, S, O heteroatoms 5-membered heteroaryl, 5-6-membered cycloalkylocene, 5-membered heteroaryla 5-6-member
  • R 6 is selected from R
  • R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, r is selected from 0, the heterocycloalkyl and cycloalkyl are further replaced by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy , C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 group substitution;
  • the compound of the present invention has the formula (I), (Ia), (Ib), (Id), (Ie), ( If) structure:
  • the compound of the present invention has the structure of formula (I), (Ib):
  • the compound of the present invention has the formula (II), (II-d), (II-e), (Ib-1) Structure:
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O.
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • R 1 and R 2 are independently OH
  • R 3 is a C 1-4 alkyl group or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O.
  • the alkyl group and heterocycloalkyl group are optionally substituted by 1-3 Substituted with a group selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • R 4 and R 5 are independently H, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1 or 2;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r -(5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -C(O)-C 3-6 cycloalkyl or -( CH 2 ) r -C 3-6 cycloalkyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl and cycloalkyl are optionally selected from 1 to 3 Group substitution of halogen, D, CN, OH, C 1-4 alkoxy, C 1-4 al
  • two R 6 on adjacent ring atoms and the atoms to which they are connected together form a C 5-6 cycloalkyl group or a 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • Alkyl or heteroaryl, the cycloalkyl, heterocycloalkyl or heteroaryl is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH;
  • r is selected from 0, 1, 2;
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is a C 1-4 alkyl group or a 4-6-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, and the alkyl group is further substituted by a C 2-4 alkenyl group. , C 2-4 alkynyl or C 3-6 cycloalkyl substituted, the heterocycloalkyl is optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl Substituted with C 3-6 cycloalkyl groups;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O. Cyclic heteroaryl;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 6 is not halogen, C 1-4 alkyl, C 2-4 alkenyl, unsubstituted C 3-6 cycloalkyl, unsubstituted Contains 1-3 4-7-membered heterocycloalkyl groups selected from N, S, O heteroatoms, -O-CH 3 or OH, and ring B is not
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O. Cyclic heteroaryl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • R 6 is selected from R, R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, r is selected from 0, the heterocycloalkyl and cycloalkyl are further replaced by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy , C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 group substitution;
  • the compound of the present invention has the formula (II), (II-d), (II-e), (Ib-1) Structure:
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O.
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • R 1 and R 2 are independently OH
  • R 3 is a C 1-4 alkyl group or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O.
  • the alkyl group and heterocycloalkyl group are optionally replaced by 1-3 Substituted with a group selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • R 4 and R 5 are independently H, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1 or 2;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r -(5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -C(O)-C 3- 6 cycloalkyl or -( CH 2 ) r -C 3-6 cycloalkyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl and cycloalkyl optionally substituted by 1-3 groups selected from halogen, D, CN, OH, C 1-4 alkoxy, C 1-4 alky
  • two R 6 on adjacent ring atoms and the atoms to which they are connected together form a C 5-6 cycloalkyl group or a 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • Alkyl or heteroaryl, the cycloalkyl, heterocycloalkyl or heteroaryl is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH;
  • r is selected from 0, 1, 2;
  • the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is a C 1-4 alkyl group or a 4-6-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, and the alkyl group is further substituted by a C 2-4 alkenyl group. , C 2-4 alkynyl or C 3-6 cycloalkyl substituted, the heterocycloalkyl is optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl Substituted with C 3-6 cycloalkyl group;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • At least one R 6 is not halogen and C 1-4 alkyl
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O. Cyclic heteroaryl;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • R 6 is not halogen, C 1-4 alkyl, C 2-4 alkenyl, unsubstituted C 3-6 cycloalkyl, unsubstituted Contains 1-3 4-7-membered heterocycloalkyl groups selected from N, S, O heteroatoms, -O-CH 3 or OH, and ring B is not
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O. Cyclic heteroaryl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl,
  • R 6 is selected from R, R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, r is selected from 0, the heterocycloalkyl and cycloalkyl are further replaced by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy , C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 group substitution;
  • the compound of the present invention has the structure of formula II:
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O.
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl,
  • R 1 and R 2 are independently OH
  • R 3 is a C 1-4 alkyl group or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O.
  • the alkyl group and heterocycloalkyl group are optionally substituted by 1-3 Substituted with a group selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • R 4 and R 5 are independently H, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1 or 2;
  • Each R 6 is independently halogen, D, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkenyl , C 2-4 alkynyl, -(CH 2 ) r - (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms), -C(O)- (containing 1-3 4-6 membered heterocycloalkyl selected from N, S, O heteroatoms) ), -(CH 2 ) r -(5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, O), -C(O)-C 3-6 Cycloalkyl or -(CH 2 ) r -C 3-6 cycloalkyl, the alkyl, alkoxy, alkenyl, alkynyl, heterocycloalkyl, heteroaryl and cycloalkyl groups are optionally 1-3 groups selected from halogen, D, CN, OH, C 1-4 alkoxy, C 1-4 alkyl,
  • two R 6 on adjacent ring atoms and the atoms to which they are connected together form a C 5-6 cycloalkyl group or a 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • Alkyl or heteroaryl, the cycloalkyl, heterocycloalkyl or heteroaryl is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH;
  • r is selected from 0, 1, 2;
  • the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is a C 1-4 alkyl group or a 4-6-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, and the alkyl group is further substituted by a C 2-4 alkenyl group. , C 2-4 alkynyl or C 3-6 cycloalkyl substituted, the heterocycloalkyl is optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl Substituted with C 3-6 cycloalkyl groups;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl,
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O. Cyclic heteroaryl;
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • Ring A is thiazolyl, pyrazolyl,
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl,
  • R 3 is a C 1-4 alkyl group or a 4-membered heterocycloalkyl group containing 1 to 3 heteroatoms selected from N, S, and O.
  • the alkyl group and heterocycloalkyl group are optionally selected from 1 to 3 heteroatoms.
  • R 4 and R 5 are independently H or C 1-4 alkyl
  • n 0 or 1
  • p and q are independently 1, 2, 3 or 4;
  • Each R 6 is independently halogen, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl , C 1-4 alkoxy, C 2-4 alkynyl , morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl , tetrahydrofuryl, tetrahydropyranyl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl or isoxazolyl, the alkyl group , Alkoxy, alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuranyl, tetrahydropyranyl, cyclopropyl, cyclobutyl, pyrazolyl, Furyl, pyrrolyl, thienyl, imid
  • D is selected from D1 or D4, and the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is a C 1-4 alkyl group or a 4-6-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, and the alkyl group is further substituted by a C 2-4 alkenyl group. , C 2-4 alkynyl or C 3-6 cycloalkyl substituted, the heterocycloalkyl is optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl Substituted with C 3-6 cycloalkyl groups;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl,
  • Ring A is thiazolyl, pyrazolyl,
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group
  • D is selected from D2, and the compound meets one of the following conditions:
  • R 6 is not halogen, C 1-4 alkyl, C 2-4 alkenyl, unsubstituted C3-6 cycloalkyl, unsubstituted containing 1-3 4-7 membered heterocycloalkyl groups selected from N, S, O heteroatoms, -O-CH 3 or OH, and ring B is not
  • Ring A is thiazolyl, pyrazolyl,
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazinyl, pyridazinyl,
  • R 6 is selected from R, R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, r is selected from 0, the heterocycloalkyl and cycloalkyl are further replaced by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy , C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 group substitution;
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl, pyrimidinyl, pyridazinyl,
  • R 3 is a C 1-4 alkyl group or a 4-membered heterocycloalkyl group containing 1 to 3 heteroatoms selected from N, S, and O.
  • the alkyl group and heterocycloalkyl group are optionally selected from 1 to 3 heteroatoms.
  • R 4 and R 5 are independently H or C 1-4 alkyl
  • n 0 or 1
  • p and q are independently 1, 2, 3 or 4;
  • Each R 6 is independently halogen, N 3 , CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H , C 1-4 alkyl, C 1-4 alkoxy, C 2-4 alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl , tetrahydrofuryl, tetrahydropyranyl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl or isoxazolyl, the alkyl group , alkoxy, alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuranyl, tetrahydropyranyl, cyclopropyl, cyclo Butyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazo
  • the compound meets one of the following conditions:
  • At least one of R 1 or R 2 is N 3 or C 1-4 alkoxy, or R 1 or R 2 and the carbon atom to which they are connected form a 4-6 membered heterocyclic ring;
  • R 3 is a C 1-4 alkyl group or a 4-6-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, and the alkyl group is further substituted by a C 2-4 alkenyl group. , C 2-4 alkynyl or C 3-6 cycloalkyl substituted, the heterocycloalkyl is optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl Substituted with C 3-6 cycloalkyl groups;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridazinyl,
  • R 3 and B ring form a 5-6 membered heterocycloalkyl group.
  • the compound of the present invention contains the structure of formula (II-a) or (II-b):
  • the compound of the present invention has the structure of formula (II-a-1), (II-e-1):
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl, alkyne and heterocycloalkyl are optionally substituted by 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 6 when R 6 is selected from R, R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, when r is selected from 0, the heterocycloalkyl and cycloalkyl are further substituted by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy group, C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 ; the definitions of other groups are consistent with any of the previous technical solutions.
  • the compound of the present invention has the structure of formula (II-a-1), (II-e-1):
  • R 3 is C 2-4 alkenyl, C 2-4 alkynyl, or a 4-6 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, and O, so alkenyl, alkyne and heterocycloalkyl are optionally substituted by 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • Ring A is A 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic heteroacyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • R 6 when R 6 is selected from R, R is selected from -(CH 2 ) r -(4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O), -(CH 2 ) r -C 3-6 cycloalkyl, when r is selected from 0, the heterocycloalkyl and cycloalkyl are further substituted by 1-3 selected from halogen, D, CN, OH, C 1-4 alkoxy Group substitution of C 1-4 alkyl, C 3-6 cycloalkyl and NH 2 ;
  • Ring A is an 8-10-membered bicyclic heteroatom containing 1-3 heteroatoms selected from N, S, and O. and ring;
  • ring A is an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O. Cycloheterocycloalkyl or bicyclic heteroaryl;
  • the present invention relates to a compound represented by formula (I), (Ia), (Ib), (Id), (Ie), (If) or formula (II), its stereoisomer, deuterated compound or pharmaceutical with acceptable salt,
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O. heterocyclic ring;
  • Ring A is a 5-6 membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, O or an 8-membered heteroaryl containing 1-3 heteroatoms selected from N, S, O. -10-membered bicyclic heterocycloalkyl or bicyclic heteroaryl;
  • Ring A is Or an 8-10 membered bicyclic heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • Ring A is Or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O; the bicyclic heterocycloalkyl group includes but is not limited to The bicyclic heteroaryl groups include but are not limited to
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O. 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O One-membered heterocycloalkyl acene group or benzo 5-6 membered cycloalkyl group;
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl,
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl,
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl, pyrimidinyl, In some embodiments, Ring B is a 5-6 membered heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O; in some embodiments, Ring B is a 5-6 membered heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O. 5-membered heteroaryl from N, S, O heteroatoms;
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, or pyrimidinyl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, or isothiazolyl;
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl, or pyrimidinyl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, or thienyl
  • R 1 and R 2 are independently OH or C 1-4 alkoxy, which is optionally substituted by 1-3 groups selected from D, halogen, OH, NH 2 and CN;
  • R 1 and R 2 are independently OH;
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl, and the alkyl, alkenyl, alkynyl and cycloalkyl groups are optionally 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • R 3 is C 2-4 alkenyl or C 2-4 alkynyl, and the alkenyl and alkynyl groups are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl groups are substituted; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and is further substituted by C 2-4 alkenyl, C 2 -4 alkynyl or C 3-6 cycloalkyl substitution;
  • R 3 is C 1-4 alkyl, said alkyl optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl, and C 3 -6 cycloalkyl group substitution;
  • R 3 is C 1-4 alkyl, further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • R 3 is C 1-4 alkyl, said alkyl optionally substituted by 1-3 selected from D, C 2-4 alkenyl, C 2-4 alkynyl, and C 3-6 Substitution of cycloalkyl groups;
  • R 4 and R 5 are independently H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • R 4 and R 5 are independently H, halogen, C 1-4 alkyl, C 2-4 alkenyl, or C 2-4 alkynyl;
  • R 4 and R 5 are independently H or C 1-4 alkyl
  • n 0, 1, 2, or 3; in some embodiments, m is 0, 1, or 2; in some embodiments, m is 1 or 2; in some embodiments, m is 0 or 1; in some In the embodiment, m is 1;
  • p, q are independently 0, 1, 2, 3 or 4; in some embodiments, p, q are independently 1, 2, 3 or 4; in some embodiments, p is 2, 3 or 4, In some embodiments, q is 1, 2, or 3;
  • Each R is independently halo, D, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR ; in some embodiments, each R is independently halo, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR; in some embodiments, at least one R 6 is not H, halogen, and C 1-4 alkyl;
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • two R 6 on adjacent ring atoms and the atoms to which they are connected together form a C 5-6 cycloalkyl group or a C cycloalkyl group containing 1 to 3 heteroatoms selected from N, S, and O.
  • the cycloalkyl, heterocycloalkyl or heteroaryl is optionally surrounded by 1-3 groups selected from halogen, D, CN, OH and NH 2 Substituted;
  • the cycloalkyl group includes but is not limited to cyclopentyl, cyclohexyl, cyclopentenyl, and cyclohexenyl;
  • the heterocycloalkyl group includes but is not limited to tetrahydropyrrolyl, tetrahydrofuranyl, and tetrahydropyranyl base, morpholinyl, piperidinyl, piperazinyl;
  • the heteroaryl group includes but is not limited to pyrazolyl, pyrrolyl, furyl, pyrimidinyl, pyridyl, pyrazinyl, pyridazinyl, thienyl, Thiazolyl, oxazolyl;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-7 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, containing 1-3 heteroatoms selected from N, S, O 4-6 membered heterocycloalkyl, 5-6 membered heteroaryl or C 3-6 cycloalkyl containing 1-3 heteroatoms selected from N, S, O, the alkyl, alkenyl, alkynyl , heterocycloalkyl, heteroaryl and cycloalkyl are optionally substituted by 1-3 groups selected from halogen, D, CN, OH, C 1-4 alkoxy and NH 2 ;
  • each R is independently H, C 1-4 alkyl, C 2-4 alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuran base, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl or isoxazolyl, the alkyl, alkynyl, morpholinyl, Azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuryl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl Or the isoxazolyl group is optionally substituted by 1-3
  • each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, morpholinyl, azetidinyl, oxetanyl , tetrahydropyrrolyl, tetrahydrofuryl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl or The alkyl, alkenyl, alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuryl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl, iso
  • each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, containing 1-3 heteroatoms selected from N, S, O 4-6 membered heterocycloalkyl, 5-6 membered heteroaryl or C 3-6 cycloalkyl containing 1-3 heteroatoms selected from N, S, O, the alkyl, alkenyl, alkynyl , heterocycloalkyl, heteroaryl and cycloalkyl are optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH 2 ;
  • each R is independently H, C 1-4 alkyl, C 2-4 alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuran base, cyclopropyl, cyclobutyl, pyrazolyl, furyl or pyrrolyl, the alkyl, alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuran base, cyclopropyl, cyclobutyl, pyrazolyl, furyl or pyrrolyl optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R 7 is independently halogen, CN, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, -OC 3-6 cycloalkyl, 4- containing 1-3 heteroatoms selected from N, S, O 7-membered heterocycloalkyl or 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O.
  • alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, hetero Cycloalkyl and heteroaryl are optionally substituted by 1-3 groups selected from halogen, C 1-4 alkyl, D, CN, OH and C 3-6 cycloalkyl;
  • each R is independently C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, -OC 3-6 cycloalkyl, containing 1-3 heteroatoms selected from N, S, O 4-7 membered heterocycloalkyl or 5-6 membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl , heterocycloalkyl and heteroaryl are optionally substituted with 1-3 groups selected from halogen, C 1-4 alkyl, D, CN, OH and C 3-6 cycloalkyl; in some embodiments , at least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl base, C 3-6 cycloalkyl group, 4-7 membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O, or 5-membered heterocycloalkyl group containing 1-3 heteroatoms selected from N, S, O -6-membered heteroaryl, the alkynyl, alkoxy, alkyl, cycloalkyl, heterocycloalkyl and heteroaryl groups are optionally substituted by 1-3 selected from halogen, C 1-4 alkyl, D, CN, OH and C 3-6 cycloalkyl group substitution;
  • each R is independently C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, cyclopropyl, cyclobutyl, -O cyclopropyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl , tetrahydropyrrolyl, pyrazolyl, pyrrolyl, pyridyl, pyrimidinyl or pyridazinyl, the alkyl, alkenyl, alkynyl, alkoxy, cyclopropyl, cyclobutyl, azetidine base, oxetanyl, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, pyrazoly
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl base, cyclopropyl, cyclobutyl, -O cyclopropyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, pyrazolyl, pyrrolyl, pyridyl, Pyrimidinyl or pyridazinyl, the alkyl, alkenyl, alkynyl, alkoxy, cyclopropyl, cyclobutyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl, tetrahydrofuranyl, Hydropyrrolyl, pyrazolyl, pyrroly
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O. heterocyclic ring;
  • Ring B is a 5-6 membered heteroaryl group, phenyl group containing 1-3 heteroatoms selected from N, S, and O. 5-6-membered heterocycloalkyl and 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O, 5-6-membered heteroaryl containing 1-3 heteroatoms selected from N, S, and O One-membered heterocycloalkyl acene group or benzo 5-6 membered cycloalkyl group;
  • Ring C is a 5-6 membered monocyclic heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently OH or C 1-4 alkoxy, which is optionally substituted by 1-3 groups selected from D, halogen, OH, NH 2 and CN;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl, and the alkyl, alkenyl, alkynyl and cycloalkyl groups are optionally 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • R 4 and R 5 are independently H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-7 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R 7 is independently C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1- 4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, -OC 3-6 cycloalkyl, 4-7 membered heterocycle containing 1-3 selected from N, S, O heteroatoms Alkyl or 5-6 membered heteroaryl containing 1-3 selected from N, S, O heteroatoms, the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, heterocycloalkyl and The heteroaryl group is optionally substituted by 1-3 groups selected from halogen, C 1-4 alkyl, D, CN, OH and C 3-6 cycloalkyl;
  • the compound meets one of the following conditions:
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 2-4 alkenyl or C 2-4 alkynyl, and the alkenyl and alkynyl groups are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2- 4 alkynyl and C 3-6 cycloalkyl groups substituted; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkyne base or C 3-6 cycloalkyl substitution;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O
  • Ring A is Or an 8-10 membered bicyclic heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, 4-7-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or 5-6-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O Heteroaryl, the alkynyl, alkoxy, alkyl, cycloalkyl, heterocycloalkyl and heteroaryl groups are optionally substituted by 1-3 selected from halogen, C 1-4 alkyl, D, CN , OH and C 3-6 cycloalkyl group substitution.
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O. heterocyclic ring;
  • Ring B is a 5-6-membered heteroaryl or phenyl group containing 1-3 heteroatoms selected from N, S, and O;
  • R 1 and R 2 are independently OH or C 1-4 alkoxy, which is optionally substituted by 1-3 groups selected from D, halogen, OH, NH 2 and CN;
  • R 3 is C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl, and the alkyl, alkenyl, alkynyl and cycloalkyl groups are optionally 1-3 groups selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl;
  • R 4 and R 5 are independently H, D, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1, 2 or 3;
  • p and q are independently 0, 1, 2, 3 or 4;
  • Each R 6 is independently halogen, D, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-7 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • two R 6 on adjacent ring atoms and their connected atoms together form a C 5-6 carbocyclic ring or a 5-6 membered heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O,
  • the carbocyclic or heterocyclic ring is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R 7 is independently C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1- 4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, -OC 3-6 cycloalkyl, 4-7 membered heterocycle containing 1-3 selected from N, S, O heteroatoms Alkyl or a 5-6 membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O, and the alkyl group Base, alkenyl, alkynyl, alkoxy, cycloalkyl, heterocycloalkyl and heteroaryl are optionally substituted by 1-3 selected from halogen, C 1-4 alkyl, D, CN, OH and C 3-6 cycloalkyl group substitution;
  • the compound meets one of the following conditions:
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 2-4 alkenyl or C 2-4 alkynyl, and the alkenyl and alkynyl groups are optionally substituted by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2- 4 alkynyl and C 3-6 cycloalkyl groups substituted; or R 3 is C 1-4 alkyl or C 3-6 cycloalkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkyne base or C 3-6 cycloalkyl substitution;
  • Ring B is a 5-membered heteroaryl group containing 1-3 heteroatoms selected from N, S, and O;
  • At least one R6 is not halogen and C 1-4 alkyl
  • Ring A is Or an 8-10 membered bicyclic heterocyclic ring containing 1-3 heteroatoms selected from N, S, and O;
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, 4-7-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or 5-6-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O Heteroaryl, the alkynyl, alkoxy, alkyl, cycloalkyl, heterocycloalkyl and heteroaryl groups are optionally substituted by 1-3 selected from halogen, C 1-4 alkyl, D, CN , OH and C 3-6 cycloalkyl group substitution.
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O.
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl, pyrimidinyl, or
  • R 1 and R 2 are independently OH
  • R 3 is C 1-4 alkyl, which is optionally replaced by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl. group substitution;
  • R 4 and R 5 are independently H, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1 or 2;
  • Each R 6 is independently halogen, D, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • two R 6 on adjacent ring atoms and the atoms to which they are connected together form a C 5-6 cycloalkyl group or a 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • Alkyl or heteroaryl, the cycloalkyl, heterocycloalkyl or heteroaryl is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH;
  • the compound meets one of the following conditions:
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 1-4 alkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, or
  • Ring A is Or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O;
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, 4-7-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or 5-6-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O Heteroaryl, the alkynyl, alkoxy, alkyl, cycloalkyl, heterocycloalkyl and heteroaryl groups are optionally substituted by 1-3 selected from halogen, C 1-4 alkyl, D, CN , OH and C 3-6 cycloalkyl group substitution;
  • Ring A is a 5-6-membered monocyclic heteroaryl containing 1-3 heteroatoms selected from N, S, and O or an 8-10-membered bicyclic ring containing 1-3 heteroatoms selected from N, S, and O.
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl, pyrazinyl or pyrimidinyl;
  • R 1 and R 2 are independently OH
  • R 3 is C 1-4 alkyl, which is optionally replaced by 1-3 selected from halogen, D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl. group substitution;
  • R 4 and R 5 are independently H, halogen, C 1-4 alkyl, C 2-4 alkenyl or C 2-4 alkynyl;
  • n 0, 1 or 2;
  • Each R 6 is independently halogen, D, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, 4-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • two R 6 on adjacent ring atoms and the atoms to which they are connected together form a C 5-6 cycloalkyl group or a 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, S, and O.
  • Alkyl or heteroaryl, the cycloalkyl, heterocycloalkyl or heteroaryl is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH;
  • the compound meets one of the following conditions:
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 1-4 alkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, oxazolyl, isoxazolyl or isothiazolyl;
  • Ring A is Or an 8-10-membered bicyclic heterocycloalkyl group or a bicyclic heteroaryl group containing 1-3 heteroatoms selected from N, S, and O;
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, C 3-6 cycloalkyl, 4-7-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O, or 5-6-membered heterocycloalkyl containing 1-3 heteroatoms selected from N, S, O Heteroaryl, the alkynyl, alkoxy, alkyl, cycloalkyl, heterocycloalkyl and heteroaryl groups are optionally substituted by 1-3 selected from halogen, C 1-4 alkyl, D, CN , OH and C 3-6 cycloalkyl group substitution;
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl, pyrimidinyl,
  • R 3 is C 1-4 alkyl, which is optionally surrounded by 1-3 groups selected from D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl replace;
  • R 4 and R 5 are independently H or C 1-4 alkyl
  • n 0 or 1
  • p and q are independently 1, 2, 3 or 4;
  • Each R 6 is independently halogen, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuranyl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl or isoxazolyl, the alkyl, alkynyl, morpholinyl, azetidinyl, Oxetanyl, tetrahydropyrrolyl, tetrahydrofuryl, cyclopropyl, cyclobutyl, pyrazolyl, furyl, pyrrolyl, thienyl, imidazolyl, oxazolyl, thiazolyl or isoxazolyl
  • 1-3 groups selected from halogen, D
  • Each R 7 is independently C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1- 4 alkyl, -CONHC 1-4 alkyl, cyclopropyl, cyclobutyl, -O cyclopropyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, Pyrazolyl, pyrrolyl, pyridyl, pyrimidinyl or pyridazinyl, the alkyl, alkenyl, alkynyl, alkoxy, cyclopropyl, cyclobutyl, azetidinyl, oxetanyl base, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, pyrazolyl
  • the compound meets one of the following conditions:
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 1-4 alkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl, thienyl, or
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, cyclo Propyl, cyclobutyl, -O-cyclopropyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuryl, tetrahydropyrrolyl, pyrazolyl, pyrrolyl, pyridyl, pyrimidinyl or clatter Azinyl, the alkyl, alkenyl, alkynyl, alkoxy, cyclopropyl, cyclobutyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, Pyrazolyl, pyrrolyl, pyridyl,
  • Ring B is phenyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl or pyrimidinyl;
  • R 3 is C 1-4 alkyl, which is optionally surrounded by 1-3 groups selected from D, C 2-4 alkenyl, C 2-4 alkynyl and C 3-6 cycloalkyl replace;
  • R 4 and R 5 are independently H or C 1-4 alkyl
  • n 0 or 1
  • p and q are independently 1, 2, 3 or 4;
  • Each R 6 is independently halogen, CN, R, N(R) 2 , COR, CON(R) 2 , OR, or NRCOR;
  • Each R is independently H, C 1-4 alkyl, C 2-4 alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuranyl, cyclopropyl, cyclobutyl, pyrazolyl, furyl or pyrrolyl, the alkyl, alkynyl, morpholinyl, azetidinyl, oxetanyl, tetrahydropyrrolyl, tetrahydrofuranyl, cyclopropyl, Cyclobutyl, pyrazolyl, furyl or pyrrolyl is optionally substituted by 1-3 groups selected from halogen, D, CN, OH and NH ;
  • Each R 7 is independently C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1- 4 alkyl, -CONHC 1-4 alkyl, cyclopropyl, cyclobutyl, -O cyclopropyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, Pyrazolyl, pyrrolyl, pyridyl, pyrimidinyl or pyridazinyl, the alkyl, alkenyl, alkynyl, alkoxy, cyclopropyl, cyclobutyl, azetidinyl, oxetanyl base, morpholinyl, tetrahydrofuranyl, tetrahydropyrrolyl, pyrazolyl
  • the compound meets one of the following conditions:
  • R 1 or R 2 is C 1-4 alkoxy
  • R 3 is C 1-4 alkyl, and further substituted by C 2-4 alkenyl, C 2-4 alkynyl or C 3-6 cycloalkyl;
  • Ring B is pyrazolyl, imidazolyl, thiazolyl or thienyl
  • At least one R 7 is C 2-4 alkynyl, C 1-4 alkoxy, -COC 1-4 alkyl, -SO 2 C 1-4 alkyl, -CONHC 1-4 alkyl, cyclo Propyl, cyclobutyl, -O-cyclopropyl, azetidinyl, oxetanyl, morpholinyl, tetrahydrofuryl, tetrahydropyrrolyl, pyrazolyl, pyrrolyl, pyridyl, pyrimidinyl or Pyridazinyl, the alkyl, alkenyl, alkynyl, alkoxy, cyclopropyl, cyclobutyl, azetidinyl, oxygen Heterocyclylbutyl, morpholinyl, tetrahydrofuryl, tetrahydropyrrolyl, pyrazolyl, pyrrolyl, pyridyl, pyr
  • the C 1-4 alkyl group includes but is not limited to methyl, ethyl, propyl, isopropyl, butyl, isobutyl, n-butyl;
  • the C 2-4 alkenyl group includes but is not limited to vinyl , propenyl, allyl, 1-butenyl, 2-butenyl, 3-butenyl;
  • the C 2-4 alkynyl group includes but is not limited to ethynyl, propynyl, propargyl, 1 -Butynyl, 2-butynyl, 3-butynyl;
  • the C 3-6 cycloalkyl group includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; the heterocycloalkane
  • the base includes but is not limited to azetidinyl, oxetanyl, morpholinyl, tetrahydrofuranyl
  • the present invention provides a compound selected from one of the following structures in Group I, its stereoisomer, deuterated compound or pharmaceutically acceptable salt:
  • the present invention also provides compounds selected from one of the following Group II structures, their stereoisomers, deuterated compounds or pharmaceutically acceptable salts:
  • the present invention also provides a pharmaceutical composition, which contains a therapeutically effective amount of any compound of the present invention, or its stereoisomer, deuterated compound or pharmaceutically acceptable salt, and a pharmaceutically acceptable salt. Carriers and/or excipients.
  • the present invention also relates to a pharmaceutical use, that is, any compound described in the present invention, or its stereoisomer, deuterated compound or pharmaceutically acceptable salt, or a composition containing them is used in the preparation of the treatment of Pol ⁇ -mediated Use in medicines for diseases.
  • the present invention also relates to a therapeutic use, that is, any compound described in the present invention, or its stereoisomer, deuterated compound or pharmaceutically acceptable salt, or a composition containing them is used in the treatment of Pol ⁇ -mediated diseases. uses in .
  • the Pol ⁇ -mediated diseases include, but are not limited to, liver cancer, breast cancer, ovarian cancer, lung cancer, kidney cancer, prostate cancer, skin cancer, bladder cancer, pancreatic cancer, or head and neck cancer.
  • the present invention relates to a pharmaceutical composition or pharmaceutical preparation, which contains a therapeutically effective amount of the compound of the present invention or its stereoisomer, deuterated product, solvate, pharmaceutically acceptable Salt and carrier and/or excipients.
  • the pharmaceutical composition may be in the form of a unit preparation (the amount of the main drug in a unit preparation is also referred to as "preparation specification").
  • the present invention also provides a method for treating diseases in a mammal, which includes administering to the mammal a therapeutically effective amount of the compound of the present invention or its stereoisomer, deuterated product, solvate, pharmaceutically acceptable Acceptable salts or pharmaceutical compositions.
  • mammals of the present invention include humans.
  • an "effective amount” or a “therapeutically effective amount” refers to administration of a sufficient amount of a compound disclosed herein that will alleviate to some extent one or more symptoms of the disease or condition being treated. In some embodiments where the result is a reduction and/or alleviation of the signs, symptoms or causes of disease, or any other desired change in a biological system.
  • an "effective amount” for therapeutic use is the amount of a composition containing a compound disclosed herein that is required to provide a clinically significant reduction in disease symptoms.
  • therapeutically effective amounts include, but are not limited to: 1-1000 mg, 1-900 mg, 1-800 mg, 1-700 mg, 1-600 mg, 1-500 mg, 1-400 mg, 1-300 mg, 1-250 mg, 1-200 mg, 1-150mg, 1-125mg, 1-100mg, 1-80mg, 1-60mg, 1-50mg, 1-40mg, 1-25mg, 1-20mg, 5-1000mg, 5-900mg, 5-800mg, 5- 700mg, 5-600mg, 5-500mg, 5-400mg, 5-300mg, 5-250mg, 5-200mg, 5-150mg, 5-125mg, 5-100mg, 5-90mg, 5-70mg, 5-80mg, 5-60mg, 5-50mg, 5-40mg, 5-30mg, 5-25mg, 5-20mg, 10-1000mg, 10-900mg, 10-800mg, 10-700mg, 10-600mg, 10-500mg, 10- 450mg, 10-400mg, 10-300mg, 10-
  • the pharmaceutical composition includes, but is not limited to: 1-1000 mg, 5-500 mg, 10-250 mg, 50-250 mg, 100-200 mg, 1 mg, 1.25 mg, 2.5 mg, 5 mg, 10 mg, 12.5 mg, 15mg, 20mg, 25mg, 30mg, 35mg, 40mg, 45mg, 50mg, 60mg, 70mg, 80mg, 90mg, 100mg, 120mg, 125mg, 150mg, 200mg, 250mg, 300mg, 400mg, 500mg, 600mg, 700mg, 800mg, 900mg, 1000 mg of the compound of the present invention or its stereoisomer, deuterated product, solvate, or pharmaceutically acceptable salt.
  • a method for treating diseases in mammals comprising administering to a subject a therapeutically effective amount of a compound of the present invention or its stereoisomer, deuterated product, solvate, or pharmaceutically acceptable salt, the therapeutically effective amount Preferred is 1-1000 mg, and the disease is preferably liver cancer, breast cancer, ovarian cancer, lung cancer, kidney cancer, prostate cancer, skin cancer, bladder cancer, pancreatic cancer or head and neck cancer.
  • a method for treating diseases in mammals includes administering a pharmaceutical compound of the present invention or its stereoisomer, deuterated product, solvate, or pharmaceutically acceptable salt at a daily dose of 1-1000 mg/day.
  • the daily dose can be a single dose or divided dose.
  • the daily dose includes but is not limited to: 1-1000 mg/day, 1-300 mg/day, 5-500 mg/day, 10-500 mg /day, 10-400mg/day, 10-300mg/day, 10-100mg/day, 20-400mg/day, 20-200mg/day, 20-100mg/day, 50-500mg/day, 50-250mg/day , 50-200mg/day, 50-150mg/day, 50-100mg/day, 100-500mg/day, 100-300mg/day, 100-200mg/day;
  • daily dosages include, but are not limited to: 1 mg/day, 2.5 mg/day, 5 mg/day, 10 mg/day, 12.5 mg/day, 15 mg/day, 20 mg/day, 25 mg/day, 30 mg/day , 35mg/day, 40mg/day, 45mg/ day, 50mg/day, 60mg/day, 70mg/day, 80mg/day, 90mg/day, 100mg/day, 120mg/day, 150mg/day, 200mg/day, 250mg/day, 300mg/day, 400mg/day, 500mg/day, 1000mg/day.
  • the present invention relates to a kit, which may include a composition in single-dose or multiple-dose form.
  • the kit contains a compound of the invention or a stereoisomer, deuterate, solvate or pharmaceutically acceptable salt thereof,
  • the amount of the compound of the present invention or its stereoisomer, deuterated product, solvate, or pharmaceutically acceptable salt is the same as that in the above-mentioned pharmaceutical composition.
  • the amount of the compound of the present invention or its stereoisomer, deuterated product, solvate, and pharmaceutically acceptable salt is converted in the form of a free base in each case.
  • Preparation specification refers to the weight of the main drug contained in each tube, tablet or other unit preparation.
  • the carbon, hydrogen, oxygen, sulfur, nitrogen or halogen involved in the groups and compounds of the present invention all include their isotopes, and the carbon, hydrogen, oxygen, sulfur, Nitrogen or halogen is optionally further replaced by one or more of their corresponding isotopes, where carbon isotopes include 12 C, 13 C and 14 C, and hydrogen isotopes include protium (H), deuterium (deuterium, also known as heavy hydrogen).
  • tritium T, also known as superheavy hydrogen
  • oxygen isotopes include 16 O, 17 O and 18 O
  • sulfur isotopes include 32 S, 33 S, 34 S and 36 S
  • nitrogen isotopes include 14 N and 15 N
  • the isotope of fluorine is 19 F
  • the isotopes of chlorine include 35 Cl and 37 Cl
  • the isotopes of bromine include 79 Br and 81 Br.
  • Halogen refers to F, Cl, Br, I or their isotopes.
  • Halo or halogen substitution refers to substitution by one or more selected from F, Cl, Br, I or their isotopes.
  • the upper limit of the number of halogen substituents is equal to the sum of the number of hydrogens that can be substituted by the substituted group. Without special limitation, the number of halogen substituents is any integer between 1 and the upper limit. When the number of halogen substituents is greater than 1, the same or different halogens may be used for substitution. It usually includes 1-5 halogen substitutions, 1-3 halogen substitutions, 1-2 halogen substitutions, and 1 halogen substitution.
  • D and “deuterium” refer to deuterium, an isotope of hydrogen (H).
  • Deuterated or “deuterated” means that the hydrogen atom on the alkyl, cycloalkyl, alkylene, aryl, heteroaryl, mercapto, heterocycloalkyl, alkenyl, alkynyl and other groups is replaced by at least In the case of substitution of a deuterium atom, the upper limit of the number of deuterations is equal to the sum of the number of hydrogens that can be substituted by the substituted group.
  • the number of deuterations is any integer between 1 and the upper limit, such as 1- 20 deuterium atoms, 1-10 deuterium atoms, 1-6 deuterium atoms, 1-3 deuterium atoms, 1-2 deuterium atoms or 1 deuterium atom.
  • C xy group refers to a group containing x to y carbon atoms, such as "C 1-6 alkyl” refers to an alkyl group containing 1 to 6 carbon atoms.
  • Alkyl refers to a monovalent linear or branched saturated aliphatic hydrocarbon group. It is usually an alkyl group of 1 to 20 carbon atoms, or an alkyl group of 1 to 8 carbon atoms, or an alkyl group of 1 to 6 carbon atoms, or an alkyl group of 1 to 4 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, neo-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl etc., the alkyl group may be further substituted by a substituent.
  • Alkylene refers to divalent straight and branched chain saturated alkyl groups. Examples of alkylene include, but are not limited to, methylene, ethylene, and the like.
  • Haloalkyl refers to the situation where one or more hydrogens in the alkyl group are replaced by one or more halogen atoms (such as fluorine, chlorine, bromine, iodine or its isotopes).
  • the upper limit of the number of halogen substituents is equal to the number of halogen atoms in the alkyl group.
  • the sum of the number of hydrogens that can be substituted, and the number of halogen substituents is any integer between 1 and the upper limit unless otherwise specified.
  • the alkyl group is substituted by 1-5 halogens or 1-3 halogens or 1-2 halogens or 1 halogen; when the number of halogen substituents is greater than 1, it can be substituted with the same or different halogens; specific examples Including but not limited to -CF 3 , -CH 2 Cl, -CH 2 CF 3 , -CCl 2 , CF 3 , etc.
  • Alkoxy refers to -O-alkyl.
  • -OC 1-8 alkyl For example -OC 1-8 alkyl, -OC 1-6 alkyl, -OC 1-4 alkyl or -OC 1-2 alkyl.
  • Specific non-limiting examples include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy, n-hexyloxy, Cyclopropoxy and cyclobutoxy, etc.; the alkoxy group may be optionally substituted by a substituent.
  • Haloalkoxy refers to -O-haloalkyl.
  • -O-halogenated C 1-8 alkyl For example -O-halogenated C 1-8 alkyl, -O-halogenated C 1-6 alkyl, -O-halogenated C 1-4 alkyl or -O-halogenated C 1-2 alkyl; halogen
  • the upper limit of the number of substituents is equal to the sum of the number of hydrogens that can be substituted by the substituted group. Without special limitation, the number of halogen substituents is any integer between 1 and the upper limit, preferably 1-5 halogen substitutions.
  • halogen substitutions when the number of halogen substituents is greater than 1, they can be substituted with the same or different halogens; non-limiting examples include monofluoromethoxy, difluoromethoxy Fluoromethoxy, trifluoromethoxy, difluoroethyloxy, etc.
  • Alkynyl refers to a linear or branched chain hydrocarbon group containing at least one carbon-carbon triple bond (C ⁇ C), usually containing 2 to 18 carbon atoms, further containing 2 to 8 carbon atoms, further containing 2 to 6 carbon atoms, further including 2 to 4 carbon atoms, examples of which include but are not limited to ethynyl, 1-propynyl, 2-propynyl, butynyl, 2-butynyl, 3-butynyl base, 1-methyl-2-propynyl, 4-pentynyl, 3-pentynyl, 1-methyl-2-butynyl, 2-hexynyl, 3- Hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonenyl and 4-decynyl, etc.; the alkynyl group can optionally Substituted by substituents.
  • Alkynylene refers to a linear or branched divalent unsaturated hydrocarbon group containing a carbon-carbon triple bond (C ⁇ C), usually containing 2 to 18 carbon atoms, and further containing 2 to 8 carbon atoms, Further comprising 2 to 6 carbon atoms, further comprising 2 to 4 carbon atoms, non-limiting examples include ethynylene, propynylene, butynylene, and the alkynylene may be optionally substituted. replace.
  • Cycloalkyl refers to a saturated or partially unsaturated, non-aromatic carbocyclic hydrocarbon group containing no ring heteroatoms.
  • the cycloalkyl group can be a monocyclic ring, a bicyclic ring or a polycyclic ring.
  • the bicyclic ring or the polycyclic ring can be a branched ring, a spiro ring, a bridged ring or a combination thereof.
  • the bicyclic ring or the polycyclic ring can include one or more aromatic rings, but the ring system as a whole Not aromatic, the attachment site can be on an aromatic ring or a non-aromatic ring.
  • the cycloalkyl group contains 3 to 20 carbon atoms, further containing 3 to 8 carbon atoms, and further containing 3 to 6 carbon atoms; when it is a monocyclic cycloalkyl group, it contains 3 to 15 carbon atoms, or 3 -10 carbon atoms, or 3 to 8 carbon atoms, or 3 to 6 carbon atoms; in the case of a bicyclic or polycyclic cycloalkyl group, 5 to 12 carbon atoms, or 5 to 11 carbon atoms, Or contain 6-10 carbon atoms; non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, butenyl, cyclopentenyl, cyclohexenyl, etc., cycloalkyl groups may be optionally substituted with substituents.
  • Cycloalkylene refers to a divalent group of cycloalkyl.
  • Aryl refers to an aromatic carbocyclic ring that does not contain heteroatoms, including single-ring aryl and fused-ring aryl. Typically contains 6 to 14 carbon atoms, and further contains 6 to 10 carbon atoms. Non-limiting examples include phenyl, naphthyl, fenyl, phenanthrenyl, and aryl groups may be optionally substituted with substituents.
  • Carbocycle or “carbocyclyl” refers to a saturated, partially unsaturated or aromatic carbocyclic ring, and its meaning includes aryl and cycloalkyl.
  • the carbocyclic ring can be a single ring, a bicyclic ring or a polycyclic ring, and the bicyclic ring or polycyclic ring includes bridged rings, parallel rings, spiro rings and combinations thereof.
  • Carbocyclic rings usually have 3 to 12 carbon atoms, or 3 to 10 carbon atoms, or 3 to 6 carbon atoms.
  • monocyclic carbocyclic rings include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or phenyl, etc.
  • bicyclic bridged rings include etc.
  • double rings and rings include etc.
  • bicyclic spiro rings include etc.
  • the carbocyclic ring may be optionally substituted with substituents.
  • Heterocycloalkyl refers to a saturated or partially unsaturated non-aromatic carbocyclic ring containing 1, 2, 3 or 4 heteroatoms selected from N, S, and O.
  • the heterocycloalkyl group can be a single ring, a bicyclic ring, or a polycyclic ring.
  • the bicyclic ring or the polycyclic ring can be a bridged ring, a branched ring, a spiro ring, or a combination thereof.
  • the bicyclic ring or the polycyclic ring can include one or more aromatic rings or heteroaromatic rings.
  • the ring system as a whole is not aromatic, and the connection site can be on an aromatic ring or a non-aromatic ring.
  • the heterocycloalkyl group is a 3- to 20-membered ring.
  • it is usually a 3 to 15-membered ring, or a 3-10-membered ring, or a 3-8-membered ring, or a 3-6-membered ring.
  • it is a bicyclic or polycyclic heterocycloalkyl group, it is usually a 5-12-membered ring, or a 5-11-membered ring, or a 6-9-membered ring.
  • heteroatoms N and S include their oxidation states.
  • Non-limiting examples of heterocycloalkyl groups include azetidine base, morpholinyl, piperazinyl, piperidinyl, tetrahydropyranyl, oxetanyl, pyranyl, azepanyl, azepanyl, oxolenyl , oxacyclohexenyl, etc., the heterocycloalkyl group may be optionally substituted by a substituent.
  • Heteroaryl ring or “heteroaryl group”, unless otherwise specified, refers to an aromatic ring containing 1 to 4 heteroatoms selected from N, O or S and their oxidation states, which can be monocyclic or bicyclic. Or polycyclic, the bicyclic or polycyclic ring can be a bridged ring, a branched ring, a spiro ring, or a combination thereof; when it is a bicyclic or polycyclic ring, it can be a fused heteroaryl group and an aryl group, or a heteroaryl group and a fused aryl group. Condensation of heteroaryl groups, in which either the heteroaryl group or the aryl group can be the attachment site.
  • Non-limiting examples include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, indolyl, purinyl, etc.; the heteroaryl group may be optionally substituted by a substituent.
  • Heterocycle or “heterocyclyl” refers to a saturated or unsaturated, aromatic or non-aromatic ring containing 1 to 4 heteroatoms selected from N, O or S and their oxidation states, and its meaning includes hetero Aryl and heterocycloalkyl.
  • Heterocycles include monocyclic heterocycles, bicyclic bridged heterocycles, bicyclic heterocycles and bicyclic spiroheterocycles or combinations thereof. Usually it is a 3- to 12-membered heterocyclic ring or a 5- to 12-membered heterocyclic ring, or a 5- to 7-membered heterocyclic ring.
  • the heterocyclyl group can be attached to a heteroatom or a carbon atom, and non-limiting examples include oxetyl, aziridyl, oxetanyl, azetidinyl, 1,3-dioxolane base, 1,4-dioxanyl, 1,3-dioxanyl, piperazinyl, azepanyl, pyridyl, furyl, thienyl, pyranyl, N-alkylpyrrole base, pyrimidinyl, pyrazinyl, pyrazolyl, pyridazinyl, imidazolyl, piperidinyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,3-dithiyl, dihydrofuranyl , dihydropyranyl, dithiopentanyl, tetrahydrofuryl, tetrahydropyrrolyl, tetrahydroimidazoly
  • Heterocyclylene refers to a substituted or unsubstituted, saturated or unsaturated, aromatic or non-aromatic divalent heterocyclic group. Non-limiting examples include wait.
  • Spiro ring refers to a polycyclic group that shares one carbon atom (called a spiro atom) between rings. It may contain 0 or more double bonds or triple bonds, and may contain 0 to 5 atoms selected from N, O, S, P, Si and heteroatoms in their oxidation states. Usually the spiro ring is a 6 to 14 membered ring, or a 6 to 12 membered ring, or a 6 to 10 membered ring.
  • the spiro rings are three-spiro-three (meaning three-membered ring spiro-three-membered ring), three-spiro-four, three-spiro-five, three-spiro-six, four-spiro-four, four-spiro-five, four-spiro-six, penta-spiro-five or penta-spiro-6.
  • spiro rings include The spiro ring may be optionally substituted by substituents.
  • “Fused ring” or “condensed ring” refers to a polycyclic group in which the ring shares two adjacent ring atoms and one chemical bond. It can contain one or more double bonds or triple bonds, and the ring can contain 0 to 5 optional Heteroatoms from N, S, O, P, Si and their oxidation states.
  • the combined ring is a 5 to 20 membered ring, or a 5 to 14 membered ring, or a 5 to 12 membered ring, or a 5 to 10 membered ring.
  • the ring is a tri-tetracyclic ring (indicating a ring formed by a three-membered ring and a four-membered ring.
  • IUPC naming rules it may be a three-membered ring as the basic ring or a four-membered ring as the basic ring.
  • the same below. principle three and five rings, three and six rings, four and four rings, four and five rings, four and six rings, five and five rings, five and six rings, and six and six rings.
  • Non-limiting examples of cyclones include purine, quinoline, isoquinoline, benzopyran, benzofuran, benzothiophene, ;
  • the ring may be aromatic or non-aromatic, and may be optionally substituted by substituents.
  • Bicyclic heterocyclic ring refers to a bicyclic group in which two rings share two adjacent ring atoms and one chemical bond. It may contain one or more double bonds or triple bonds, and contains 1 to 5 atoms selected from N, S, and O. , P, Si and heteroatoms in their oxidation state can be saturated, partially saturated or aromatic, and their meanings include bicyclic heterocycloalkyl and bicyclic heteroaryl.
  • the combined ring is a 5- to 20-membered ring, or a 5- to 14-membered ring, or a 5- to 12-membered ring, or an 8- to 10-membered ring.
  • the ring is a six-membered pentacyclic ring (indicating a ring formed by a six-membered ring and a five-membered ring. According to the IUPC naming rules, it may be a six-membered ring as the basic ring or a five-membered ring as the basic ring. The same below. theory), six and six rings, five and five rings, four and four rings, four and five rings, four and six rings, five and five rings, five and six rings, six and six rings.
  • the bicyclic heterocyclic ring is optionally substituted by a substituent.
  • Bridged ring means that two rings share two non-adjacent ring atoms and may contain one or more double or triple bonds.
  • the bridged ring may contain 0 to 5 heteroatoms selected from N, S, O, P, Si and their oxidation states. Usually the ring atoms of the bridged ring range from 5 to 20, or 5 to 14, or 5 to 12, or 5 to 10.
  • Non-limiting examples of bridged rings include adamantane,
  • substitution refers to any substitution at a position permitted by chemical theory, and the number of substituents complies with the rules of chemical bonding.
  • substituents include, but are not limited to: C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 heteroalkyl, C 5-12 aryl, 5-12 yuan Heteroaryl, hydroxyl, C 1-6 alkoxy, C 5-12 aryloxy, thiol, C 1-6 alkylthio, cyano, halogen, C 1-6 alkylthiocarbonyl, C 1 -6 -alkylcarbamoyl, N-carbamoyl, nitro, silyl, sulfinyl, sulfonyl, sulfoxide, halogenated C 1-6 alkyl, halogenated C 1-6 alkoxy, amino , phosphonic acid, -CO 2 (
  • Alkyl optionally substituted by F means that the alkyl group can but does not have to be substituted by F, including the case where the alkyl group is substituted by F and the case where the alkyl group is not substituted by F.
  • “Pharmaceutically acceptable salt” means that the compound of the present invention retains the biological effectiveness and properties of the free acid or free base, and the free acid is mixed with a non-toxic inorganic base or organic base, and the free base is mixed with Salts obtained by the reaction of non-toxic inorganic acids or organic acids.
  • “Pharmaceutical composition” means a mixture of one or more compounds described herein, or their stereoisomers, deuterates, solvates, pharmaceutically acceptable salts or co-crystals, with other constituents, wherein the other constituents Contains physiologically/pharmaceutically acceptable carriers and/excipients.
  • Carrier refers to a vehicle that does not cause significant irritation to the organism and does not eliminate the biological activity and properties of the administered compound. It can change the way the drug enters the human body and its distribution in the body, control the release rate of the drug, and transfer the drug to the body.
  • Non-limiting examples of delivery systems to targeted organs include microcapsules and microspheres, nanoparticles, liposomes, etc.
  • Excipient means an excipient that is not itself a therapeutic agent and is used as a diluent, excipient, binder and/or vehicle and is added to a pharmaceutical composition to improve its handling or storage properties or to allow or facilitate The compounds or pharmaceutical compositions are formed into unit dosage forms for administration.
  • pharmaceutical excipients can serve a variety of functions and can Described are wetting agents, buffers, suspending agents, lubricants, emulsifiers, disintegrants, absorbents, preservatives, surfactants, colorants, flavoring agents and sweeteners.
  • Examples of pharmaceutical excipients include, but are not limited to: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose and its derivatives, such as carboxymethyl Sodium cellulose, ethyl cellulose, cellulose acetate, hydroxypropyl methylcellulose, hydroxypropyl cellulose, microcrystalline cellulose and croscarmellose (such as croscarmellose sodium) ; (4) tragacanth powder; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository wax; (9) oils, such as peanut oil, cottonseed oil, red Flower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as oils Ethyl acid este
  • Stepoisomers refer to isomers produced by different spatial arrangements of atoms in a molecule, including cis-trans isomers, enantiomers and conformational isomers.
  • the compounds of the present invention also include their tautomers.
  • the present invention describes the left-hand compound in which the pyrimidine ring is substituted by OH
  • the right-hand tautomer compound is also included.
  • Solvate refers to a substance formed by a compound of the present invention or a salt thereof and a stoichiometric or non-stoichiometric solvent bound by non-covalent intermolecular forces.
  • the solvent is water, it is a hydrate.
  • Co-crystal refers to a crystal formed by combining an active pharmaceutical ingredient (API) and a co-crystal form (CCF) under the action of hydrogen bonds or other non-covalent bonds.
  • API active pharmaceutical ingredient
  • CCF co-crystal form
  • the pure states of API and CCF are both Solids, and there are fixed stoichiometric ratios between the components.
  • a eutectic is a multicomponent crystal that includes both a binary eutectic formed between two neutral solids and a multicomponent eutectic formed between a neutral solid and a salt or solvate.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS). NMR shifts ( ⁇ ) are given in units of 10 -6 (ppm). NMR was measured using (Bruker Avance III 400 and Bruker Avance 300) nuclear magnetic instruments, and the measurement solvents were deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), and deuterated methanol (CD 3 OD ), the internal standard is tetramethylsilane (TMS);
  • HPLC was measured using Agilent 1260DAD high-pressure liquid chromatograph (Zorbax SB-C 18 100 ⁇ 4.6mm, 3.5 ⁇ M);
  • Thin layer chromatography silica gel plates use Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plates.
  • the specifications of silica gel plates used in thin layer chromatography (TLC) are 0.15mm-0.20mm.
  • the specifications used for thin layer chromatography separation and purification products are 0.4mm. -0.5mm;
  • T3P 1-propylphosphonic anhydride
  • Step 1 Dissolve compound 1A (0.2g, 0.55mmol) (synthesized with reference to the method described in patent WO2021028670) in 1,4-dioxane (10mL), and add compound 1B (0.13g, 0.66mmol) to it in sequence ), Pd 2 (dba) 3 (0.056g, 0.06mmol), XantPhos (0.1g, 0.17mmol), potassium carbonate (0.11g, 0.83mmol), stir evenly, and under nitrogen atmosphere, heat to 95°C and react for 18 hours. After the reaction is complete, cool to room temperature, filter the reaction solution through diatomaceous earth, pour the filtrate into water (10 mL), and extract with ethyl acetate (20 mL ⁇ 3).
  • Step 2 Dissolve compound 1C (0.11g, 0.21mmol) in dichloromethane (10mL), cool to -20°C, slowly add boron trichloride (0.075g, 0.63mmol) dropwise, and naturally rise to room temperature and stir for 3 Hour. The reaction was quenched by adding ice-cold saturated aqueous sodium bicarbonate solution (10 mL), and the residue was washed with ethyl acetate (20 mL ⁇ 3). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
  • Step 1 Dissolve compound 2A (0.5g, 2.42mmol) in 1,4-dioxane (8mL), add compound 2B (0.6g, 2.90mmol), PdCl 2 (dppf) (0.18g, 0.24) in sequence mmol), sodium carbonate (0.51g, 4.84mmol), water (2mL), under nitrogen atmosphere, heat to 100°C and stir for 18 hours.
  • Second step Using 2C and 1A as raw materials, refer to the first step of Example 1 to obtain compound 2D.
  • LC-MS(ESI): m/z 535.1[M+H] + .
  • Step 3 Using 2D as raw material, refer to the second step of Example 1 to obtain compound 2.
  • LC-MS(ESI): m/z 495.1[M+H] + .
  • Step 1 Dissolve compound 3A (2.2g, 10.14mmol) in toluene (30mL), and add potassium fluoroborate (1.48g, 12.17mmol) and tetrakis triphenylphosphorus palladium (0.59g, 0.51mmol) to it in sequence. , water (3mL), under nitrogen atmosphere, heat to 90°C and stir for 18 hours. After the reaction is complete, cool to room temperature. The reaction solution is filtered through diatomaceous earth. The obtained filtrate is poured into water (10 mL) and extracted with ethyl acetate (20 mL ⁇ 3). The combined organic phase is washed with saturated brine.
  • target compound 3B 600 mg, yield 30.1%.
  • Second step Using 3B and 1A as raw materials, refer to the first step of Example 1 to obtain compound 3C.
  • LC-MS(ESI): m/z 524.1[M+H] + .
  • Step 3 Using 3C as raw material, refer to the second step of Example 1 to obtain target compound 3.
  • LC-MS(ESI): m/z 495.1[M+H] + .
  • the first step use 4B and 4A (refer to the method described in patent WO2021028643 for synthesis) as raw materials, and refer to the first step of Example 1 to obtain the target compound 4C.
  • LC-MS(ESI): m/z 492.1[M+H] + .
  • Second step Using 4C as raw material, refer to the second step of Example 1 to obtain target compound 4.
  • LC-MS(ESI): m/z 452.1[M+H] + .
  • Step 1 Dissolve 5A (2g, 7.86mmol) and 3,3-dimethylazetidine (0.73g, 7.86mmol) in toluene (50mL), then add palladium acetate (0.18g, 0.79mmol) ), ( ⁇ )-2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (0.98g, 1.57mmol) and cesium carbonate (5.12g, 15.72mmol), under nitrogen ventilation protection, Stir at 60°C for 16h.
  • Step 2 Dissolve 5B (1.82g, 6.83mmol) in a mixed solvent of ethanol (50mL) and water (50mL), add zinc powder (2.23g, 34.15mmol) and ammonium chloride (3.65g, 68.30mmol) , react at 80°C for 1 hour.
  • Step 3 Dissolve 5C (1.5g, 6.34mmol) in toluene (50mL), add sodium hydroxide (2.54g, 63.40mmol), tetrabutylammonium bromide (0.20g, 0.63mmol) and disulfate sulfate in sequence. Methyl ester (0.88g, 6.97mmol), react at room temperature overnight.
  • Step 4 Dissolve 5E (0.4g, 1.99mmol) (refer to the method described in patent WO2021028670 for synthesis) and 5D (0.6g, 2.39mmol) in pyridine (10mL), and slowly add 50% T3P ethyl acetate at room temperature. solution (7.97mmol), react at 50°C overnight.
  • Step 5 Using 5F and 2-chloro-6-methyl-4-(trifluoromethyl)pyridine as raw materials, refer to the first step of Example 1 to obtain the target compound 5G.
  • LC-MS(ESI): m/z 593.1[M+H] + .
  • Step 6 Using 5G as raw material, refer to the second step of Example 1 to obtain the target compound 5.
  • Step 2 Dissolve 6A (2.16g, 7.41mmol) and 2-chloro-6-methyl-4-(trifluoromethyl)pyridine (1.74g, 8.90mmol) in 1,4-dioxane ( 50 mL), add Pd 2 (dba) 3 (679 mg, 0.74 mmol), 4,5-bis(diphenylphosphine)-9,9-dimethylxanthene (858 mg, 1.48 mmol) and potassium carbonate in sequence. (2.05g, 14.83mmol), react under nitrogen atmosphere at 95°C overnight.
  • Compound 7 was obtained by using 6C and imidazole [1,2-A]pyridine-6-amino as raw materials and operating with reference to Example 6 (fourth, five and six steps of reaction).
  • Step 1 Dissolve 6C (600mg, 1.67mmol) and 13A (310mg, 1.67mmol) in pyridine (15mL), slowly add 50% T3P in ethyl acetate solution (6.68mmol) at room temperature, and react at 50°C overnight.
  • Step 2 Dissolve 13B (200mg, 0.38mmol) and morpholine (50mg, 0.57mmol) in 1,4-dioxane (8mL), then add Pd 2 (dba) 3 (35mg, 0.038mmol) , 4,5-bis(diphenylphosphine)-9,9-dimethylxanthene (44 mg, 0.076 mmol) and potassium carbonate (110 mg, 0.76 mmol), react overnight at 90°C in a nitrogen atmosphere.
  • Pd 2 (dba) 3 35mg, 0.038mmol
  • 4,5-bis(diphenylphosphine)-9,9-dimethylxanthene 44 mg, 0.076 mmol
  • potassium carbonate 110 mg, 0.76 mmol
  • Step 3 Dissolve 13C (125 mg, 0.23 mmol) in 4M hydrogen chloride in 1,4-dioxane solution (6 mL), and react at room temperature overnight.
  • Second step Using 6C and 14B as raw materials, refer to the first step of Example 13 to obtain the title compound 14C (204 mg, 13%).
  • LC-MS(ESI): m/z 560.1[M+H] + .
  • Step 3 Using 14C as raw material, refer to the third step of Example 13 to obtain compound 14.
  • Compound 15 was obtained by using 6C (200 mg, 0.56 mmol) and 15A as raw materials and operating with reference to Example 8 (steps 3, 4, and 5).
  • Step 1 Dissolve 16A (synthesized with reference to the method described in patent WO 2020156445) (580g, 2.66mol) in ethanol (3L), slowly add 6M hydrochloric acid solution at room temperature, and raise the temperature to 100°C to react overnight. After the reaction was cooled to room temperature, the solid produced was filtered, the filter cake was washed with water several times, and then the solid was dried to obtain the title compound 16B (440 g, 88%).
  • LC-MS(ESI): m/z 187.1[M-H]-.
  • Step 2 Dissolve 16B (440g, 2.34mol) and benzyltrimethylammonium chloride (868g, 4.68mol) in phosphorus oxychloride (3L), raise the temperature to 105°C and react overnight. Wait for the reaction to cool to room temperature, concentrate under reduced pressure to remove most of the solvent, add 3L of ethyl acetate to the residue, then slowly add 5L of ice water, extract and separate the liquids, extract the aqueous phase with ethyl acetate (1L ⁇ 2), and combine the organic The layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
  • Step 3 Combine 16C (2g, 8.89mmol), methylboronic acid (0.53g, 8.89mmol), dichloro[1,1′-bis(diphenylphosphine)ferrocene]palladium (0.65g, 0.89mmol) ) and potassium carbonate (2.46g, 17.78mmol) were dissolved in 1,4-dioxane (25mL), and the reaction was carried out overnight at 100°C in a nitrogen atmosphere.
  • Step 4 Using 16D and 4A as raw materials, refer to the first step of Example 1 to obtain the title compound 16E (360 mg, 46%).
  • LC-MS(ESI): m/z 529.1[M+H] + .
  • Step 5 Using 16E as raw material, refer to the second step of Example 1 to obtain the title compound 16 (55 mg, 20%).
  • the first step using 6C and 4-bromo-5-chloro-2-fluoroaniline as raw materials, refer to the first step of Example 13 to obtain the title compound 17A.
  • LC-MS(ESI): m/z 566.0[M+H] + .
  • Step 2 Using 17A as raw material, refer to the fourth step of Example 8 to obtain 17B.
  • LC-MS(ESI): m/z 583.1[M+H] + .
  • Step 3 Using 17B as raw material, refer to the second step of Example 13 to obtain the title compound 17C.
  • LC-MS(ESI): m/z 590.2[M+H] + .
  • Step 4 Using 17C as raw material, refer to the third step of Example 13 to obtain compound 17.
  • Compound 19 was obtained by using 17B and 3,3-difluoropyrrolidine as raw materials and operating with reference to Example 17 (third and fourth step reactions).
  • Step 1 In a single-mouth bottle, add cyclopentanone (9.20g, 109.33mmol), compound 21B (20.13g, 109.33mmol), and ammonium acetate (8.43g, 109.33mmol) in sequence, and raise the temperature to 135°C for 18 hours. Cool to room temperature and continue stirring for 5 hours. If solid precipitates, filter, wash the filter cake three times with petroleum ether, dry the filter cake to obtain a crude product, and recrystallize the crude product with ethanol to obtain the target compound 21C (5.7g, yield 25.66%).
  • LC-MS(ESI): m/z 204.1[M+H] + .
  • Step 2 In a 50 mL single-neck bottle, add compound 21C (2.80 g, 13.78 mmol) and phosphorus oxychloride (20 mL) in sequence, and raise the temperature to 100°C to react overnight. Wait for the reaction to cool to room temperature, concentrate under reduced pressure to remove most of the solvent, add the residue dropwise to an ice-cold saturated aqueous sodium bicarbonate solution, extract three times with ethyl acetate, and wash the combined organic phases with saturated brine. Dry with aqueous sodium sulfate and concentrate under reduced pressure to obtain the crude product. The crude product was purified with a medium-pressure preparation instrument Biotage Isolera One (40g silica gel column, eluent: 0-10% EA/PE) to obtain the target compound 21D (1.1g, yield 36.02%) ).
  • Biotage Isolera One 40g silica gel column, eluent: 0-10% EA/PE
  • Step 3 In a single-mouth bottle, add compound 21D (0.50g, 2.26mmol), 6A (0.66g, 2.26mmol), Pd 2 (dba) 3 (0.13g, 0.23mmol), XantPhos (0.39g, 0.68 mmol), potassium carbonate (0.47g, 3.39mmol), dioxane (10mL), nitrogen replacement three times, the temperature was raised to 95°C, and the reaction was stirred for 18 hours. Cool to room temperature, filter through diatomaceous earth, pour the filtrate into water, and extract three times with ethyl acetate. The combined organic phase is washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product.
  • the crude product is prepared under medium pressure.
  • the target compound 21E (0.38g, yield 35.29%) was purified using Biotage Isolera One (12g silica gel column, eluent: 0-25% EA/PE).
  • LC-MS(ESI): m/z 477.1[M+H] + .
  • Step 4 In a single-neck bottle, add compound 21E (0.38g, 0.80mmol), 10% palladium-carbon (0.85g, 0.80mmol), and anhydrous methanol (20mL) in sequence, and react at room temperature for 1 hour in a hydrogen atmosphere. After the reaction was completed, the mixture was filtered through diatomaceous earth, and the filtrate was concentrated under reduced pressure to obtain the target compound 21F (0.22g, 71.18%). It was used directly in the next reaction without purification.
  • LC-MS(ESI): m/z 387.1[M+H] + .
  • Step 5 In a single-mouth bottle, add compound 21F (0.10g, 0.26mmol), 21G (0.026g, 0.26mmol), 1-propylphosphoric anhydride (50% ethyl acetate solution) (1.07g, 1.68mmol) in sequence. ), pyridine (10 mL), raise the temperature to 50°C and stir for 18 hours. Cool to room temperature, pour the reaction solution into water, and extract three times with ethyl acetate. The combined organic phases are washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product.
  • Step 6 In a 50mL single-neck bottle, add compound 21H (0.12g, 0.23mmol), DMF (5mL), add sodium hydride (60%) (0.014g, 0.35mmol) at 0°C, stir and react for 10 minutes, and then Deuterated methyl iodide (0.05g, 0.35mmol) was added dropwise, and the temperature was slowly raised to room temperature and the reaction was stirred for 2 hours. The reaction solution was added dropwise to an ice-cold dilute acid aqueous solution, and extracted three times with ethyl acetate. The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product.
  • the crude product was washed with medium
  • the target compound 21I (0.10g, yield 79.21%) was purified using Biotage Isolera One (12g silica gel column, eluent: 0-30% EA/PE).
  • LC-MS(ESI): m/z 549.2[M+H] + .
  • Step 7 In a 50mL single-neck bottle, add compound 21I (0.10g, 0.18mmol) and dichloromethane (8mL) in sequence, cool to -20°C, and slowly add boron trichloride (1.33g, 11.35mmol) dropwise. The temperature was slowly raised to room temperature and the reaction was stirred for 30 minutes. The reaction solution was added dropwise to an ice-cold saturated aqueous sodium bicarbonate solution, and extracted three times with ethyl acetate. The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product.
  • Step 2 In a 50mL single-neck bottle, add compound 22B (0.45g, 2.24mmol), 5E (0.50g, 3.36mmol), and pyridine (10mL) in sequence, stir at room temperature for 10min, and then slowly add 1-propylphosphoric anhydride dropwise (50% ethyl acetate solution) (4.28g, 6.73mmol), stir at room temperature for 1 hour after the dropwise addition, and then raise the temperature to 50°C for reaction for 48 hours. Cool to room temperature, concentrate under reduced pressure to remove most of the pyridine, pour the residue into water, and extract three times with ethyl acetate.
  • 1-propylphosphoric anhydride dropwise (50% ethyl acetate solution) (4.28g, 6.73mmol
  • the third step using 22C and 2-chloro-6-methyl-4-(trifluoromethyl)pyridine as raw materials, refer to the first step of Example 1 to obtain the target compound 22D (0.26g, yield 65.19%).
  • LC-MS(ESI): m/z 493.1[M+H] + .
  • Step 4 Using 22D raw material, refer to the second step of Example 1 to obtain the target compound 22.
  • Step 1 In a 50mL single-neck bottle, add compound 26A (0.5g, 2.45mmol), 26B (0.32g, 2.45mmol), DIPEA (0.95g, 7.35mmol), and dichloromethane (20mL) in sequence, and stir at room temperature for 30 minutes, then slowly add HATU (1.12g, 2.94mmol) dropwise, and stir at room temperature for 1 hour after the dropwise addition is completed. Concentrate under reduced pressure to remove most of the solvent, pour the residue into water, and extract three times with ethyl acetate. The combined organic phases are washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product.
  • the crude product is pressed with medium
  • the target compound 26C (0.46g, yield 67.50%) was purified using Biotage Isolera One (12g silica gel column, eluent: 0-15% EA/PE).
  • LC-MS(ESI): m/z 279.0[M+H] + .
  • Step 2 In a 50mL single-neck bottle, add compound 26C (0.46g, 1.65mmol), zinc powder (0.54g, 8.25mmol), and anhydrous methanol (20mL) in sequence, stir at room temperature for 5 minutes, and then add ammonium chloride (0.44 g,8.23 mmol) and continue stirring for 10 minutes. Filter through diatomaceous earth, concentrate the filtrate, pour the residue into water, and extract three times with ethyl acetate. The combined organic phases are washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the target compound 26D (0.33 g, yield 80.59%).
  • LC-MS(ESI): m/z 249.1[M+H] + .
  • the third step using 26D and compound 6C as raw materials, refer to the fourth step of Example 6 to obtain the target compound 26E.
  • LC-MS(ESI): m/z 591.2[M+H] + .
  • Step 4 Using 26E as raw material, refer to the fourth step of Example 8 to obtain the target compound 26F.
  • LC-MS(ESI): m/z 608.2[M+H] + .
  • Step 5 Using 26F as raw material, refer to the second step of Example 1 to obtain the target compound 26.
  • Step 2 At room temperature, add lithium aluminum hydride (5.69g, 150mmol) into the round-bottomed flask, add dry tetrahydrofuran (50mL), and stir evenly. Dissolve 28B (6.36g, 30mmol) in dry THF (25mL). Add the THF solution of 28B dropwise to the suspension of lithium aluminum hydride under an ice bath. After the dropwise addition is completed, reflux at 80°C for 16 hours. TLC monitors the disappearance of raw materials and stops the reaction.
  • Step 3 Using 6C and 28C as raw materials, refer to the first step of Example 13 to obtain 28D.
  • LC-MS(ESI): m/z 469.2[M+H] + .
  • Step 4 Using 28D as raw material, refer to the third step of Example 13 to obtain compound 28 (30 mg, 21.89%).
  • Step 1 Dissolve 29A (1.2g, 5.0mmol) in methanol (30mL) at room temperature, add zinc powder (3.27g, 50mmol), stir evenly, and add ammonium chloride (2.67g) to the reaction system in batches , 50 mmol), react at room temperature for 30 minutes, TLC monitors the disappearance of the raw materials, and stops the reaction. After filtration, the filtrate was concentrated, EA (50 mL) was added to dissolve the residue, the organic phase was washed with water (30 mL ⁇ 3), the organic phase was dried, and concentrated to obtain product 29B (1 g, 96.15%).
  • LC-MS(ESI): m/z 207.9[M+H] + .
  • Step 2 Using 6C and 29B as raw materials, refer to the fourth step of Example 6 to obtain 29C.
  • LC-MS(ESI): m/z 550.0[M+H] + .
  • the third step using 29C as raw material, refer to the fifth step of Example 6 to operate the product 29D.
  • LC-MS(ESI): m/z 569.0[M+H] + .
  • Step 4 Using 29D as raw material, refer to the second step of Example 13 to obtain 29E.
  • LC-MS(ESI): m/z 580.2[M+H] + .
  • Step 5 Using 29E as raw material, refer to the second step of Example 1 to obtain compound 29.
  • the first step using 6C and 30A as raw materials, refer to the fourth step of Example 6 to obtain 30B.
  • LC-MS(ESI): m/z 517.1[M+H] + .
  • Step 2 Using 30B as raw material, refer to the fifth step of Example 6 to obtain product 30C.
  • LC-MS(ESI): m/z 534.1[M+H] + .
  • the third step using 30C as raw material, refer to the first step of Example 29 to obtain product 30D.
  • LC-MS(ESI): m/z 504.1[M+H] + .
  • LC-MS(ESI): m/z 562.2[M+H] + .
  • Second step Using 31A as raw material, refer to the second step of Example 1 to obtain compound 31.
  • the first step using 32A as raw material, refer to the first step of Example 26 to obtain product 32B.
  • LC-MS(ESI): m/z 327.9[M+H] + .
  • Step 4 Using 32D as raw material, refer to the third step of Example 26 to obtain 32E.
  • LC-MS(ESI): m/z 607.1[M+H] + .
  • Step 5 Using 32E as raw material, refer to the fourth step of Example 26 to obtain product 32F.
  • LC-MS(ESI): m/z 624.1[M+H] + .
  • Step 6 Using 32F as raw material, refer to the fifth step of Example 26 to obtain compound 32 (50 mg, 53.52%).
  • the first step using 33B and 6C as raw materials, refer to the first step of Example 13 to obtain the target compound 33A.
  • LC-MS(ESI): m/z 480.0[M+H] + .
  • Step 2 Using 33A as raw material, refer to the third step of Example 13 to obtain compound 33.
  • LC-MS(ESI): m/z 440.2[M+H] + .
  • Step 1 Add compound 17B (0.10g, 0.17mmol), Pd(PPh 3 ) 2 Cl 2 (24.0mg, 0.034mmol), CuI (16.0mg, 0.085mmol), and 5mL DMF into the dry reaction bottle in sequence. Replace with nitrogen three times, continue to add triethylamine (0.10g, 1.02mmol), complete the addition of trimethylsilylacetylene (0.10g, 1.02mmol), replace with nitrogen again, and then heat up to 50°C to react overnight.
  • Step 2 Dissolve compound 34A (84.0 mg, 0.14 mmol) in 1 mL of 1,4-dioxane, then add 5 mL of hydrogen chloride/1,4-dioxane solution, stir at room temperature for 40 hours, and mix the reaction solution Add 20 mL of ethyl acetate to dilute, add saturated sodium bicarbonate solution to adjust the pH to 7-8, separate the organic phase, wash the organic phase with 20 mL of saturated brine, dry over anhydrous sodium sulfate, filter, and concentrate to obtain target compound 34B (52.0 mg , yield 66.21%).
  • LC-MS(ESI): m/z 561.2[M+H] + .
  • Step 1 In a 50mL single-neck bottle, add compound 36A (0.43g, 2.08mmol), potassium vinyl trifluoroborate (0.42g, 3.14mmol), [1,1′-bis(diphenylphosphine)bis Ferrocene]palladium dichloride dichloromethane complex (0.047g, 0.081mmol), sodium carbonate (0.17g, 0.21mmol), dioxane (8mL), water (2mL), replace with nitrogen three times, and heat to The reaction was stirred at 60°C for 18 hours.
  • the third step using 36C as raw material, refer to the first step of Example 1 to obtain compound 36D.
  • LC-MS(ESI): m/z 531.2[M+H] + .
  • Step 4 Using 36C as raw material, operate compound 36 with reference to the second step of Example 1.
  • the first step using compound 6C (0.12g, 0.33mmol) and 2,4-difluoro-5-chloroaniline (0.065g, 0.4mmol) as raw materials, refer to the fourth step of Example 6 to obtain compound 37A (160mg, Yield 95.85%).
  • LC-MS(ESI): m/z 506.1[M+H] + .
  • Step 2 In a 25mL single-neck bottle, add compound 37A (0.06g, 0.12mmol), DMF (5mL), add sodium hydride (60%) (0.014g, 0.36mmol) at 0°C, stir and react for 15 minutes, drop Add allyl bromide (0.044g, 0.36mmol), slowly raise the temperature to room temperature and stir for 1 hour.
  • the third step Using compound 37B (0.07g, 0.13mmol) as raw material, the target compound 37 (24mg, yield 36.5%) was obtained by referring to the second step of Example 1.
  • Step 1 In a 50 mL sealed tube, add compound 40A (0.77 g, 4.0 mmol) and ammonia water (20 mL) in sequence, raise the temperature to 120°C and stir for 2 hours. After the reaction was completed, cooled to room temperature, the reaction solution was poured into water, extracted three times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain compound 40B (0.53g, collected The yield was 69.9%) and was directly used in the next reaction without further purification.
  • LC-MS(ESI): m/z 191.0[M+H] + .
  • Second step Using compound 6C (0.18g, 0.50mmol) and 5-chloro-2-fluoro-4-nitroaniline (9B) as raw materials, refer to the fourth step of Example 6 to obtain compound 40C (230mg, yield 86.33%).
  • Step 3 Using compound 40C as raw material, refer to the fifth step of Example 6 to obtain compound 40D (60 mg, yield 39%).
  • LC-MS(ESI): m/z 538.1[M+H] + .
  • Step 4 Using compound 40D (0.06g, 0.11mmol) as raw material, refer to the first step of Example 29 to obtain compound 40E (60 mg, yield 99.99%), which was directly used in the next reaction without further purification.
  • LC-MS(ESI): m/z 520.1[M+H] + .
  • Step 5 Using compound 40E (0.06g, 0.11mmol) as raw material, refer to the fourth step of Example 30 to obtain compound 40F (70 mg, yield 99.99%), which was directly used in the next reaction without further purification.
  • LC-MS(ESI): m/z 282.3[M/2+H] + .
  • Step 6 Using compound 40F (0.067g, 0.12mmol) as raw material, refer to the second step of Example 1 to obtain compound 40 (20 mg, yield 32%).
  • LC-MS(ESI): m/z 522.1[M+H] + .
  • Step 1 In a 50mL single-neck bottle, add compound 41A (0.6g, 3.67mmol), morpholine (0.96g, 11.02mmol), potassium carbonate (2.53g, 18.3mmol), and acetonitrile (10mL) in sequence, and heat to 80 The reaction was stirred for 48 hours.
  • Second step Using compound 4A (0.1g, 0.27mmol) and compound 41B (0.057g, 0.27mmol) as raw materials, refer to the first step of Example 1 to obtain compound 41C (90 mg, yield 62.08%).
  • LC-MS(ESI): m/z 537.2[M+H] + .
  • the third step Using compound 41C (0.09g, 0.17mmol) as raw material, refer to the second step of Example 1 to obtain compound 41 (15 mg, yield 17.76%).
  • Step 1 In a 50mL single-neck bottle, add compound 44A (0.5g, 2.06mmol), potassium vinyl trifluoroborate (0.41g, 3.09mmol), [1,1′-bis(diphenylphosphine)bis Ferrocene]palladium dichloride dichloromethane complex (0.23g, 0.31mmol), potassium carbonate (0.41g, 3.09mmol), dioxane (10mL), water (1mL), replace with nitrogen three times, and heat to The reaction was stirred at 100°C for 18 hours.
  • Step 2 In a 50mL sealed tube, add compound 44B (0.23g, 1.24mmol), THF (15mL), (trifluoromethyl)trimethylsilane (1.76g, 12.4mmol), under nitrogen protection, and heat to 80 The reaction was stirred for 18 hours. After the reaction was cooled to room temperature, the reaction solution was concentrated under reduced pressure to obtain a crude product. The crude product was purified using a medium-pressure preparation instrument Biotage Isolera One (12g silica gel column, eluent: 0-10% EA/PE) to obtain compound 44C (150 mg, yield 51.44%).
  • Biotage Isolera One (12g silica gel column, eluent: 0-10% EA/PE
  • Step 3 Using compound 44C (0.15g, 0.64mmol) as raw material, refer to the first step of Example 29 to obtain compound 44D (130 mg, yield 99%), which was directly used in the next reaction without further purification.
  • LC-MS(ESI): m/z 206.1[M+H] + .
  • Step 4 Using 6C (0.08g, 0.22mmol) and compound 44D (0.10g, 0.48mmol) as raw materials, refer to the fourth step of Example 6 to obtain compound 44E (70 mg, yield 58.12%).
  • Step 5 Using compound 44E (0.07g, 0.13mmol) as raw material, refer to the fifth step of Example 6 to obtain compound 44F (80 mg, yield 99%), which was directly used in the next reaction without further purification.
  • LC-MS(ESI): m/z 564.1[M+H] + .
  • Step 6 Compound 44F (0.08g, 0.13mmol) was used as raw material and the target compound 44 (22mg, yield 30%) was obtained by referring to the second step of Example 1.
  • Step 1 In a 50mL single-neck bottle, add compound 47A (5.76g, 58.65mmol), ethyl trifluoroacetoacetate (10.8g, 58.65mmol), and ammonium acetate (4.52g, 58.65mmol) in sequence, and raise the temperature to 135°C. The reaction was stirred for 18 hours. Stir slowly and cool to room temperature, precipitate a solid, filter, wash the solid with diethyl ether, and purify the solid by recrystallization from ethanol to obtain compound 47B (5.6 g, yield 43.56%).
  • the third step Using compound 1A (0.12g, 0.33mmol) and compound 47C (0.16g, 0.68mmol) as raw materials, refer to the first step of Example 1 to obtain compound 47D (0.075g, yield 40.37%).
  • LC-MS(ESI): m/z 563.1[M+H] + .
  • Step 4 Using compound 47D (0.075g, 0.13mmol) as raw material, refer to the second step of Example 1 to obtain compound 47 (30 mg, yield 44.14%).
  • Step 1 Dissolve compound 48A (2.5g, 17mmol) in ethanol (15mL) in a 50mL single-neck bottle, add iron nitrate (7.03g, 17mmol), raise the temperature to 50°C and stir for 3 hours. Cool to room temperature, pour the reaction solution into water, and extract with ethyl acetate. The organic phase is washed with water and saturated brine in sequence, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. The crude product is dissolved in toluene (15 mL) and heated to Stir at 50° C.
  • LC-MS(ESI): m/z 242.1[M+H] + .
  • Step 3 Using 48C (160 mg, 0.66 mmol) as raw material, refer to the first step of Example 29 to obtain compound 48D (140 mg, 99%), which can be directly used in the next step of reaction without further purification.
  • LC-MS(ESI): m/z 212.1[M+H] + .
  • Step 4 Using 6C (0.1g, 0.27mmol) and compound 48D (0.11g, 0.52mmol) as raw materials, refer to the fourth step of Example 6 to obtain compound 48E (140 mg, yield 91%).
  • LC-MS(ESI): m/z 554.1[M+H] + .
  • Step 5 Using compound 48E (0.11g, 0.2mmol) as raw material, refer to the fifth step of Example 6 to obtain compound 48F (100 mg, yield 87.6%), which can be directly used in the next reaction without further purification.
  • LC-MS(ESI): m/z 571.1[M+H] + .
  • Step 6 Using compound 48F (0.08g, 0.18mmol) as raw material, refer to the second step of Example 1 to obtain target compound 48 (40 mg, yield 41.86%).
  • Step 1 Dissolve 50A (1g, 4.65mmol) and 3,3-difluoroazetidine (0.65g, 6.98mmol) in N-methylpyrrolidone (20mL), then add potassium carbonate (1.29g , 9.30mmol), reacted at 60°C for 5h.
  • Second step Using 1A (0.25g, 0.61mmol) and 50B (0.17g, 0.61mmol) as raw materials, refer to the first step of Example 1 to obtain the target compound 50C (125mg, 34%).
  • LC-MS(ESI): m/z 600.2[M+H] + .
  • Step 3 Using 50C (125 mg, 0.21 mmol) as raw material, refer to the second step of Example 1 to obtain the title compound 50 (61 mg, 52%).
  • Step 1 Dissolve 51A (8g, 41.48mmol) in DMF (50mL), then add potassium carbonate (17.28g, 125.04mmol), stir at room temperature for 10 minutes, then slowly add methyl iodide (11.83g, 83.36mmol) at room temperature. Reaction was allowed to take place overnight. After 16 h, 100 mL of water was added to the reaction solution, extracted with ethyl acetate (50 mL ⁇ 3), the organic layers were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and concentrated, the residue was subjected to silica gel column chromatography.
  • Step 2 Combine 51B (7.5g, 36.37mmol), methylboronic acid (2.39g, 39.93mmol), dichloro[1,1′-bis(diphenylphosphine)ferrocene]palladium (2.66g, 3.64 mmol) and cesium carbonate (23.70g, 72.74mmol) were dissolved in 1,4-dioxane (200mL), and the reaction was carried out overnight at 100°C under nitrogen atmosphere.
  • Step 3 Dissolve 51C (5.2g, 28.02mmol) and methyl acrylate (24.12g, 280mmol) in DMF (80mL), then add Pd 2 (dba) 3 (2.57g, 2.80mmol), N, N - Dicyclohexylmethylamine (10.95g, 56.04mmol) and tri-tert-butylphosphine (2.27g, 11.22mmol) were reacted overnight at 100°C under nitrogen atmosphere.
  • Step 5 Dissolve 51E (2.66g, 11.21mmol) in a mixed solvent of tetrahydrofuran (50mL) and methanol (2mL), add 60% sodium hydride (1.8g, 45mmol) in batches under a nitrogen atmosphere, and then heat to React at 60°C for 4 hours. Cool to room temperature, concentrate the reaction solution, and obtain the crude residue 51F (3.5 g), which is directly used in the next step of the reaction without further purification.
  • Step 6 Place the crude 51F obtained in the previous step into a round-bottomed flask, slowly add concentrated hydrochloric acid (30 mL) under an ice bath, and then raise the temperature to 115°C for 1 hour. Cool to room temperature, slowly add saturated aqueous sodium bicarbonate solution (200 mL) in an ice bath, extract with ethyl acetate (50 mL ⁇ 3), combine the organic layers, wash with saturated brine (60 mL), dry over anhydrous sodium sulfate, and concentrate.
  • concentrated hydrochloric acid (30 mL) under an ice bath
  • ethyl acetate 50 mL ⁇ 3
  • Step 9 Under nitrogen atmosphere, slowly add phosphorus oxychloride (5 mL) to 51I (0.26 g, 1.41 mmol), and react at 100°C for 3 hours. Cool to room temperature, concentrate to remove part of the solvent, slowly add saturated sodium bicarbonate aqueous solution (15 mL) to the residue, extract with ethyl acetate (10 mL ⁇ 3), combine the organic layers, dry over anhydrous sodium sulfate, and concentrate the residue with silica gel
  • Step 10 Using 1A (0.23g, 0.63mmol) and 51J (128mg, 0.63mmol) as raw materials, refer to the first step of Example 1 to obtain the target compound 51K (115mg, 34%).
  • LC-MS(ESI): m/z 531.1[M+H] + .
  • Step 11 Using 51K (115 mg, 0.22 mmol) as raw material, refer to the first step of Example 1 to obtain the title compound 51 (40 mg, 52%).
  • Step 1 Dissolve 52A (1g, 4.01mmol) in 4M hydrogen chloride in 1,4-dioxane solution (20mL), and react at room temperature overnight. After 16 h, the solid produced was filtered, the filter cake was washed with a small amount of petroleum ether, and the solid was dried to obtain target compound 52B (710 mg, 95%).
  • LC-MS(ESI): m/z 150.1[M+H] + .
  • Step 2 Using 6C (200 mg, 0.56 mmol) and 52B (105 mg, 0.56 mmol) as raw materials, refer to the fourth step of Example 6 to obtain the target compound 52C (170 mg, 62%).
  • LC-MS(ESI): m/z 492.1[M+H] + .
  • Step 3 Using 52C (170 mg, 0.35 mmol) as raw material, refer to the fifth step of Example 6 to obtain the target compound 52D (110 mg, 62%).
  • LC-MS(ESI): m/z 509.1[M+H] + .
  • Step 4 Using 52D (193 mg, 0.37 mmol) as raw material, refer to the sixth step of Example 6 to obtain compound 52 (42 mg, 41%).
  • Step 3 Under nitrogen atmosphere, slowly add phosphorus oxychloride (5 mL) to 53C (246 mg, 1.12 mmol), and react at 100°C for 3 hours. Cool to room temperature, concentrate to remove part of the solvent, slowly add saturated sodium bicarbonate aqueous solution (15 mL) to the residue, extract with ethyl acetate (10 mL ⁇ 3), combine the organic layers, dry over anhydrous sodium sulfate, and concentrate the residue with silica gel
  • Step 4 Dissolve 6A (250mg, 0.85mmol) and 53D (203mg, 0.85mmol) in 1,4-dioxane (15mL), then add Pd 2 (dba) 3 (78mg, 0.085mmol), 4,5-bis(diphenylphosphine)-9,9-dimethylxanthene (104 mg, 0.17 mmol) and potassium carbonate (235 mg, 1.70 mmol) were reacted overnight at 95°C under nitrogen ventilation protection.
  • Pd 2 (dba) 3 78mg, 0.085mmol
  • 4,5-bis(diphenylphosphine)-9,9-dimethylxanthene 104 mg, 0.17 mmol
  • potassium carbonate 235 mg, 1.70 mmol
  • Step 6 Using 53F (132 mg, 0.33 mmol) and 5-chloro-2,4-difluoroaniline (54 mg, 0.33 mmol) as raw materials, refer to the fourth step of Example 6 to obtain the target compound 53G (144 mg, 80% ).
  • LC-MS(ESI): m/z 548.2[M+H] + .
  • Step 7 Use 53G (144 mg, 0.26 mmol) as raw material, and operate the target compound 53H (115 mg, 77%) with reference to the fifth step of Example 6.
  • LC-MS(ESI): m/z 565.2[M+H] + .
  • Step 8 Using 53H (115 mg, 0.20 mmol) as raw material, refer to the sixth step of Example 6 to obtain compound 53 (35 mg, 23%).
  • Example 54 (2S, 3S, 4S)-N-(5-chloro-4-(2,2-difluorocyclopropyl)-2-fluorophenyl)-3,4-dihydroxy-N-( Methyl-d3)-1-(6-methyl-4-(trifluoromethyl)pyridin-2-yl)-5-oxopyrrolidine-2-carboxamide (Compound 54)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

一种式(I-c)所示的含氮杂环化合物,或其立体异构体、氘代化物或者药学上可接受的盐,以及它们的药物组合物,并提供了在制备治疗DNA聚合酶θ介导的疾病的药物中的用途,式(I-c)中各基团如说明书之定义。

Description

DNA聚合酶θ抑制剂及其用途 技术领域
本发明涉及一种DNA聚合酶θ的抑制剂,及其在制备治疗肿瘤中的用途。
背景技术
DNA聚合酶θ(Polθ)是一种独特的多功能DNA聚合酶。哺乳动物细胞已经进化出多种途径来修复DNA双链断裂(DSB)以确保基因组稳定性。DNA聚合酶theta(Polθ)是替代末端连接(alt-EJ)通路的关键组成部分,也称为微同源介导的末端连接(MMEJ)通路,参与DNA双链断裂修复。MMEJ是除了非同源末端连接(NHEJ)和同源重组(HR)之外,细胞的另外一种修复途径。DNA聚合酶θ(Polθ)对修复DNA断裂非常重要,特别是在同源重组缺陷(HRD)细胞中。HRD,包括BRCA1和BRCA2基因的缺陷,是多种重要类型肿瘤的临床重要特征,包括乳腺癌、卵巢癌、前列腺癌和胰腺癌,Polθ高度表达并引导DSB修复朝向alt-EJ,开启微同源介导的末端连接(MMEJ)的DNA修复过程。在HR缺陷的情况下,Polθ的抑制通过毒性RAD51中间体的积累和alt-EJ修复途径的抑制,导致细胞死亡。此外,Polθ还具有逆转录RNA并促进以RNA为模板的DNA修复功能[7]。Polθ在正常组织中的几乎不表达,但在多种肿瘤类型(如乳腺癌、卵巢癌、HNSCC和肺癌)中高表达。同时,在这些瘤种中,又普遍存在同源重组修复缺陷(HRD);因此,Polθ抑制剂在这些瘤种中存在着应用的理论基础。
目前,HRD肿瘤可以用多聚ADP核糖聚合酶(PARP)抑制剂来治疗,这代表了一个快速增长的、数十亿美元的全球市场。但是,有相当一部分患者最初对PARP抑制剂治疗没有反应,绝大多数接受治疗的患者最终出现PARP抑制剂耐药性。Polθ抑制剂同时具有作为单药疗法以及联合PARP抑制剂治疗多种类型肿瘤的潜力,其独特的作用机制可能有助于解决2种类型的PARP耐药。其他临床患者群体也可能有从Polθ抑制剂联合治疗方案中受益,包括可能与化疗、放疗和免疫肿瘤药物的联合治疗方案。
发明内容
本发明提供了一种结构新颖、药效好的Polθ抑制剂,用于治疗与肿瘤相关的疾病,兼具生物利用度高和毒副作用低的优异效果。
本发明涉及一种式式(I-c)、(I-a)、式(I)、式(I-b)、(I-d)、(I-e)、(I-f)、式(II)、式(II-a)、(II-d)、(II-e)、式(I-b-1)、(II-a-1)、(II-e-1)或者(II-b)或者(I-1-a)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,


其中,
D环选自
X1、X2各自独立地选自O或者S;或者X2不存在;在一些实施方案中,X1、X2各自独立地选自O;X3选自O或者NRx3
X4选自O或者NH或者NRx4;在一些实施方案中,X4选自O;在一些实施方案中,X4选自NH;在一些实施方案中,X4选自NRx4
Rx3独立选自COR;
Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的5-6元杂芳基)、-(CH2)s-C3-6环烷基或-(CH2)s-苯基;所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选的被1-3个选自卤素、D、OH、CN、C1-4烷基、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;在一些实施方案中,Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-C3- 6环烷基;所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C1-4烷基的基团取代;在一些实施方案中,Rx4选自C1-4烷基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-C3-6环烷基;所述烷基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C1-2烷基的基团取代;在一些实施方案中,Rx4选自甲基、乙基、丙基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-C3-6环烷基;所述甲基、乙基、丙基、杂环烷基和环烷基任选的被1-3个 选自F、Cl、D(氘)、甲基、乙基的基团取代;在一些实施方案中,Rx4选自-CH3、-CH2CH3、-CH2D、CHD2、-CD3、-CH2-环丙基、-CH2-环丁基、z选自0或者1;在一些实施方案中,z选自0;在一些实施方案中,z选自1;
s选自0、1、2、3、4或者5;在一些实施方案中,s选自0、1、2、3或4;在一些实施方案中,s选自0、1、2或3;在一些实施方案中,s选自0、1或2;在一些实施方案中,s选自1或2;在一些实施方案中,s选自1;在一些实施方案中,s选自2;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;在一些实施方案中,环A为含有1-3个选自N、S、O杂原子的8-10元双环杂并环;在一些实施方案中,环A为含有1、2或3个选自N、S、O杂原子的5元单环杂芳基、含有1、2或3个选自N、S、O杂原子的6元单环杂芳基、含有1、2或3个选自N、S、O杂原子的8元双环杂并环、含有1、2或3个选自N、S、O杂原子的9元双环杂并环或者含有1、2或3个选自N、S、O杂原子的8元双环杂并环;在一些实施方案中,环A为噻唑基、吡唑基、 在一些实施方案中,环A为 在一些实施方案中,环A为或者在一些实施方案中,环A为 或者 在一些实施方案中,环A为在一些实施方案中,环A为在一些实施方案中,环A为
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、含有1-3个选自N、S、O杂原子的6元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
其中,在无特殊说明的情况下,环B的连接顺序为从左到右;
在一些实施方案中,环B为苯基;在一些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基、 在一 些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、
在一些实施方案中,环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、嘧啶基、
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;在某些实施方案中,C环为含有1-3个选自N、S、O杂原子的5元单环杂芳基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;在某些实施方案中,R1和R2独立地为N3、OH或者甲氧基;在某些实施方案中,R1和R2独立地为OH;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;在某些实施方案中,R1与所连接的碳原子形成4-6元杂环;在某些实施方案中,R1与所连接的碳原子形成5或者6元杂环;在某些实施方案中,R1与所连接的碳原子形成哌啶环;在某些实施方案中,R2与所连接的碳原子形成4-6元杂环;在某些实施方案中,R2与所连接的碳原子形成5或者6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;在某些实施方案中,R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;在某些实施方案中,R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;在某些实施方案中,R3为甲基、氘代甲基或者含有1、2或3个选自N、S、O杂原子的4、5或6元杂环烷基;在某些实施方案中,R3为甲基、氘代甲基、-CH2-环丙基、3-丙烯基、3-丙炔基、氧杂环丁基、在某些实施方案中,R3为甲基、氘代甲基;在某些实施方案中,R3为-CH2-环丙基、3-丙烯基、3-丙炔基、氧杂环丁基、
作为选择,R3与B环形成5-6元杂环烷基;在某些实施方案中,R3与B环形成哌啶环或者四氢吡咯环;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;在某些实施方案中,R4和R5独立地为H或者C1-4烷基;在某些实施方案中,R4和R5独立地为H或者甲基;
n、m各自独立地选自0、1、2或3;在某些实施方案中,n、m各自独立为0或1;在某些实施方案中,n为0,m为0或1;在某些实施方案中,m为0,n为0或1;在某些实施方案中,在某些实施方案中,n为0,m为0;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
或者每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-C(O)-C3-6环烷基或者-(CH2)r-C3-6环烷基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;或者每个R独立地为-(CH2)r-(含有1-3个选自N、S、O杂原子的7元杂环烷基),所述杂环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
在某些实施方案中,每个R6独立地为卤素、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基,所述烷基、烷氧基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的基团取代;或者每个R独立地为C2-4烯基、所述烯基、任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
在某些实施方案中,每个R7独立地为=O、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-CH2-环丙基、-O-CH2CH2-环丙基、-O-CH2-环丁基、-O-CH2CH2-环丁基、-O-氧杂环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基、噻吩基、-P(=O)(C1-4烷基)2、-NHS(O)2C1-4烷基、-N(C1-3烷基)S(O)2C1-4烷基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基或噻吩基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基、环丙基和环丁基的基团取代;或者每个R7独立地为卤素;
在某些实施方案中,每个R7独立地为=O、F、Cl、Br、I、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-CH2-环丙基、-O-CH2CH2-环丙基、-O-CH2-环丁基、-O-CH2CH2-环丁基、-O-氧杂环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基、噻吩基、-P(=O)(C1-4烷基)2、-NHS(O)2C1-4烷基、-N(C1-3烷基)S(O)2C1-4烷基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基或噻吩基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基、环丙基和环丁基的基团取代;
每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
r选自0、1、2或3;
D选自D1或D4,所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(c)m或n为1、2或3,或者X2不存在;或者m为1或2;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
或者环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
(i)z选自1;
(j)X1或X2选自S;
D选自D2,所述化合物满足以下条件之一:
(k)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(l)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
(m)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(n)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原 子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
或者环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
(o)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代。
本发明涉及一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,D环选自
X1、X2各自独立地选自O或者S,或者X2不存在;;
X3选自O或者NRx3
X4选自O或者NH或者NRx4
Rx3独立选自COR;
Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的5-6元杂芳基)、-(CH2)s-C3-6环烷基或-(CH2)s-苯基;所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选的被1-3个选自卤素、D、OH、CN、C1-4烷基、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
z选自0或者1;
s选自0、1、2、3、4或者5;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、含有1-3个选自N、S、O杂原子的6元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
作为选择,R3与B环形成5-6元杂环烷基;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
n、m各自独立地选自0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2、-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
r选自0、1、2或3。
本发明涉及一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,D环选自
X1、X2各自独立地选自O或者S;
X3选自O或者NRx3
X4选自O或者NH或者NRx4
Rx3独立选自COR;
Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的5-6元杂芳基)、-(CH2)s-C3-6环烷基或-(CH2)s-苯基;所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选的被1-3个选自卤素、D、OH、CN、C1-4烷基、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
z选自0或者1;
s选自0、1、2、3、4或者5;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、含有1-3个选自N、S、O杂原子的6元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
作为选择,R3与B环形成5-6元杂环烷基;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
n、m各自独立地选自0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的 4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2、-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
r选自0、1、2或3。
本发明涉及一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,D环选自
X1、X2各自独立地选自O或者S;
X3选自O或者NRx3
X4选自O或者NH或者NRx4
Rx3独立选自COR;
Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的5-6元杂芳基)、-(CH2)s-C3-6环烷基或-(CH2)s-苯基;所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选的被1-3个选自卤素、D、OH、CN、C1-4烷基、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
z选自0或者1;
s选自0、1、2、3、4或者5;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、含有1-3个选自N、S、O杂原子的6元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
作为选择,R3与B环形成5-6元杂环烷基;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
n、m各自独立地选自0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2、-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
r选自0、1、2或3;
D选自D1或D4,所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(c)m或n为1、2或3;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷 基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
(i)z选自1;
(j)X1或X2选自S;
D选自D2,所述化合物满足以下条件之一:
(k)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(l)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
(m)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(n)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(o)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代。
本发明涉及一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,D环选自
X1、X2各自独立地选自O或者S;
X3选自O或者NRx3
X4选自O或者NH或者NRx4
Rx3独立选自COR;
Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的5-6元杂芳基)、-(CH2)s-C3-6环烷基或-(CH2)s-苯基;所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选的被1-3个选自卤素、D、OH、CN、C1-4烷基、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
z选自0或者1;
s选自0、1、2、3、4或者5;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
作为选择,R3与B环形成5-6元杂环烷基;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
n、m各自独立地选自0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2、-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
r选自0、1、2或3;
所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(c)m或n为1、2或3;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6 元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
(i)z选自1;
(j)X1或X2选自S。
本发明涉及一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,D环选自
X1、X2各自独立地选自O或者S;
X3选自O或者NRx3
X4选自O或者NH;
Rx3独立选自COR;
z选自0或者1;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的 5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
作为选择,R3与B环形成5-6元杂环烷基;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
n、m各自独立地选自0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2、-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
r选自0、1、2或3;
所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(c)m或n为1、2或3;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
(i)z选自1;
(j)X1或X2选自S。
本发明涉及一种式(I-1-a)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,X1、X2各自独立地选自O或者S;
X3选自O或者NRx3
Rx3独立选自COR;
z选自0或者1;
环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基或者苯并4-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
作为选择,R3与B环形成5-6元杂环烷基;
R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
n、m各自独立地选自0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为=O、卤素、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
r选自0、1、2或3;
所述化合物满足以下条件之一:
(p)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(q)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(r)m或n为1、2或3;
(s)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基或者苯并4-6元环烷基;
(t)至少存在一个R6不为卤素和C1-4烷基;
(u)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(v)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
(w)R3与B环形成5-6元杂环烷基;
(x)z选自1;
(y)X1或X2选自S。
本发明一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,D选自D1或D4,所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(c)m或n为1、2或3;或者X2不存在;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
(i)z选自1;
(j)X1或X2选自S;
基团定义与前文任一技术方案一致。
本发明一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
D选自D1或D4,所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
(c)m或n为1、2或3;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3 个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
(i)z选自1;
(j)X1或X2选自S;
基团定义与前文任一技术方案一致。
本发明一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
D选自D2,所述化合物满足以下条件之一:
(k)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(l)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
(m)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(n)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
(o)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(I)、(I-a)、(I-b)、(I-d)、(I-e)、(I-f)的结构:
基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(I)、(I-b)的结构:
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(II)、(II-d)、(II-e)、(I-b-1)的结构:
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基、
R1和R2独立地为OH;
R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1或2;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-C(O)-C3-6环烷基或者-(CH2)r-C3-6环烷基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;或者每个R独立地为-(CH2)r-(含有1-3个选自N、S、O杂原子的7元杂环烷基),所述杂环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
r选自0、1、2;
在式(II)、(II-d)中,所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
(c)m为1或2;或者X2不存在;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)或者含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
在式(II-e)中,所述化合物满足以下条件之一:
(i)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(j)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
(k)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(l)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
(m)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(II)、(II-d)、(II-e)、(I-b-1)的结构:

其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基、
或环B为
R1和R2独立地为OH;
R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1或2;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-C(O)-C3- 6环烷基或者-(CH2)r-C3-6环烷基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基 和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
r选自0、1、2;
所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
(c)m为1或2;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O- (CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)或者含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基
在式(II-e)中,所述化合物满足以下条件之一
(i)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(j)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
(k)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(l)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
(m)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式II的结构:
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、嘧啶基、
R1和R2独立地为OH;
R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1或2;
每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-C(O)-C3-6 环烷基或者-(CH2)r-C3-6环烷基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
r选自0、1、2;
所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
(c)m为1或2;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
环A为噻唑基、吡唑基、
或环A为
或环A为
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、
或环B为
R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H或者C1-4烷基;
m为0或1;
p、q独立地为1、2、3或4;
每个R6独立地为卤素、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基,所述烷基、烷氧基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的基团取代;或者每个R独立地为C2-4烯基、所述烯基、任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的基团取代;
每个R7独立地为=O、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-氧杂环丁基、-O-CH2-环丙基、-O-CH2CH2-环丙基、-O-CH2-环丁基、-O-CH2CH2-环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基、噻吩基、-P(=O)(C1-4烷基)2、-NHS(O)2C1-4烷基、-N(C1-3烷基)S(O)2C1-4烷基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基或噻吩基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基、环丙基和环丁基的基团取代;或者每个R7独立地为卤素;
D选自D1或D4,所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
(c)m为1;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为噻唑基、吡唑基、
或环A为
或环A为
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-氧杂环丁基、-O-CH2-环丙基、-O-CH2CH2-环丙基、-O-CH2-环丁基、-O-CH2CH2-环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪 基、噻吩基、-P(=O)(C1-4烷基)2、-NHS(O)2C1-4烷基、-N(C1-3烷基)S(O)2C1-4烷基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基或噻吩基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基、环丙基和环丁基的基团取代;
(h)R3与B环形成5-6元杂环烷基;
D选自D2,所述化合物满足以下条件之一:
(i)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(j)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
(k)环A为噻唑基、吡唑基、
或环A为
(l)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、
(m)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
其他基团定义如前文一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
环A为
环A为
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、吡啶基、嘧啶基、哒嗪基、
R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H或者C1-4烷基;
m为0或1;
p、q独立地为1、2、3或4;
每个R6独立地为卤素、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基,所述烷基、烷氧基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环 丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基、任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的基团取代;
每个R7独立地为=O、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-氧杂环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代;
所述化合物满足以下条件之一:
(a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
(b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
(c)m为1;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、哒嗪基、
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为
环A为
(g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-氧杂环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代;
(h)R3与B环形成5-6元杂环烷基。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,包含式(II-a)或(II-b)结构:
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(II-a-1)、(II-e-1)的结构:
在式II-a-1中,所述化合物满足以下条件之一:
(a)、R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(b)、至少存在一个R6不为卤素和C1-4烷基;
(c)、环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(d)、至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
在式II-e-1中,所述化合物满足以下条件之一:
(e)、当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(f)、当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH;
(g)、环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(h)、当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(II-a-1)、(II-e-1)的结构:
在式II-a-1中,所述化合物满足以下条件之一:
(a)、R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(b)、至少存在一个R6不为卤素和C1-4烷基;
(c)、环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(d)、至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O 杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
在式II-e-1中,所述化合物满足以下条件之一:
(e)、当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
(f)、当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH;
(g)、环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(h)、当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中:环A为含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中:环A为含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中:环A为噻唑基、吡唑基、
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中:环A为
其他基团定义与前文任一技术方案一致。
本发明所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中:环A为
其他基团定义与前文任一技术方案一致。
本发明涉及一种式(I)、(I-a)、(I-b)、(I-d)、(I-e)、(I-f)或式(II)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
在一些实施方案中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
在一些实施方案中,环A为或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
在一些实施方案中,环A为或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;所述双环并环杂环烷基包括但不限于所述双环并环杂芳基包括但不限于
在一些实施方案中,环A为
在一些实施方案中,环A为
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基或者苯并5-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
在一些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、嘧啶基、
在一些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、嘧啶基、
在一些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、吡啶基、嘧啶基、在一些实施方案中,环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基或苯基;在一些实施方案中,环B为含有1-3个选自N、S、O杂原子的5元杂芳基;
在一些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基或嘧啶基;
在一些实施方案中,环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基或异噻唑基;
在一些实施方案中,环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、吡啶基或嘧啶基;
在一些实施方案中,环B为吡唑基、咪唑基、噻唑基或噻吩基;
R1和R2独立地为OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
在一些实施方案中,R1和R2独立地为OH;
在一些实施方案中,R1或R2为C1-4烷氧基;
R3为C1-4烷基、C2-4烯基、C2-4炔基或C3-6环烷基,所述烷基、烯基、炔基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
在一些实施方案中,R3为C2-4烯基或C2-4炔基,所烯基和炔基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
在一些实施方案中,R3为C1-4烷基,所述烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
在一些实施方案中,R3为C1-4烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
在一些实施方案中,R3为C1-4烷基,所述烷基任选的被1-3个选自D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
在一些实施方案中,R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
在一些实施方案中,R4和R5独立地为H或者C1-4烷基;
m为0、1、2或3;在一些实施方案中,m为0、1或2;在一些实施方案中,m为1或2;在一些实施方案中,m为0或1;在一些实施方案中,m为1;
p、q独立地为0、1、2、3或4;在一些实施方案中,p、q独立地为1、2、3或4;在一些实施方案中,p为2、3或4,在一些实施方案中,q为1、2或3;
每个R6独立地为卤素、D、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;在一些实施方案中,每个R6独立地为卤素、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;在一些实施方案中,至少存在一个R6不为H、卤素和C1-4烷基;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
在一些实施方案中,可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;所述环烷基包括但不限于环戊基、环己基、环戊烯基、环己烯基,所述杂环烷基包括但不限于四氢吡咯基、四氢呋喃基、四氢吡喃基、吗啉基、哌啶基、哌嗪基;所述杂芳基包括但不限于吡唑基、吡咯基、呋喃基、嘧啶基、吡啶基、吡嗪基、哒嗪基、噻吩基、噻唑基、噁唑基;
每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基、C3-6环烷基或苯基,所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基和NH2的基团取代;
在一些实施方案中,每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基或C3-6环烷基,所述烷基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基和NH2的基团取代;
在一些实施方案中,每个R独立地为H、C1-4烷基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基,所述烷基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基任选地被1-3个选自卤素、D、CN、OH、甲氧基、乙氧基和NH2的基团取代;
在一些实施方案中,每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基、异噁唑基或者所述烷基、烯基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基、异噁唑基或任选地被1-3个选自卤素、D、CN、OH、甲氧基、乙氧基和NH2的基团取代;在一些实施方案中,每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基、C3-6环烷基或苯基,所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
在一些实施方案中,每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基或C3-6环烷基,所述烷基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
在一些实施方案中,每个R独立地为H、C1-4烷基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基或吡咯基,所述烷基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基或吡咯基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、-OC3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
在一些实施方案中,每个R7独立地为C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、-OC3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;在一些实施方案中,至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
在一些实施方案中,至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
在一些实施方案中,每个R7独立地为C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、环丙基、环丁基、-O环丙基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代;
在一些实施方案中,至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、环丙基、环丁基、-O环丙基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代。
作为更具体的第一技术方案,提供了式(I)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基或者苯并5-6元环烷基;
C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
R1和R2独立地为OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
R3为C1-4烷基、C2-4烯基、C2-4炔基或C3-6环烷基,所述烷基、烯基、炔基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基、C3-6环烷基或苯基,所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、-OC3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
所述化合物满足以下条件之一:
(a)R1或R2为C1-4烷氧基;
(b)R3为C2-4烯基或C2-4炔基,所烯基和炔基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(c)m为1、2或3;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环
烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基或者苯并5-6元环烷基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代。
作为更具体的第二技术方案,提供了式(I)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基或苯基;
R1和R2独立地为OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
R3为C1-4烷基、C2-4烯基、C2-4炔基或C3-6环烷基,所述烷基、烯基、炔基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1、2或3;
p、q独立地为0、1、2、3或4;
每个R6独立地为卤素、D、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基、C3-6环烷基或苯基,所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、-OC3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述烷 基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
所述化合物满足以下条件之一:
(a)在一些实施方案中,R1或R2为C1-4烷氧基;
(b)R3为C2-4烯基或C2-4炔基,所烯基和炔基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(c)m为1、2或3;
(d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
(g)至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代。
作为更具体的第三技术方案,式I所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式II的结构:
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、嘧啶基、 或者
R1和R2独立地为OH;
R3为C1-4烷基,所述烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1或2;
每个R6独立地为卤素、D、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基或C3-6环烷基,所述烷基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
所述化合物满足以下条件之一:
(a)R1或R2为C1-4烷氧基;
(b)R3为C1-4烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(c)m为1或2;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、 或者
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(g)至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
其他基团定义如前文任一技术方案。
作为更具体的第四技术方案,式I所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式II的结构:
其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基或嘧啶基;
R1和R2独立地为OH;
R3为C1-4烷基,所述烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
m为0、1或2;
每个R6独立地为卤素、D、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基、含有1-3个选自N、S、O杂原子的5-6元杂芳基或C3-6环烷基,所述烷基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
所述化合物满足以下条件之一:
(a)在一些实施方案中,R1或R2为C1-4烷氧基;
(b)R3为C1-4烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(c)m为1或2;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基或异噻唑基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
(g)至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、C3-6环烷基、含有1-3个选自N、S、O杂原子的4-7元杂环烷基或者含有1-3个选自N、S、O杂原子的5-6元杂芳基,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH和C3-6环烷基的基团取代;
其余基团如第二技术方案所述。
作为更具体的第五技术方案,式I或II的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
环A为
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、吡啶基、嘧啶基、
R3为C1-4烷基,所述烷基任选的被1-3个选自D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H或者C1-4烷基;
m为0或1;
p、q独立地为1、2、3或4;
每个R6独立地为卤素、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基,所述烷基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基任选地被1-3个选自卤素、D、CN、OH、甲氧基、乙氧基和NH2的基团取代;
每个R7独立地为C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、环丙基、环丁基、-O环丙基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代;
所述化合物满足以下条件之一:
(a)R1或R2为C1-4烷氧基;
(b)R3为C1-4烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(c)m为1;
(d)环B为吡唑基、咪唑基、噻唑基、噻吩基、 或者
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为
(g)至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、环丙基、环丁基、-O环丙基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒 嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代。
其他基团定义如前文任意技术方案。
作为更具体的第六技术方案,式I或II的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
环A为
环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、吡啶基或嘧啶基;
R3为C1-4烷基,所述烷基任选的被1-3个选自D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
R4和R5独立地为H或者C1-4烷基;
m为0或1;
p、q独立地为1、2、3或4;
每个R6独立地为卤素、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
每个R独立地为H、C1-4烷基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基或吡咯基,所述烷基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、环丙基、环丁基、吡唑基、呋喃基或吡咯基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
每个R7独立地为C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、环丙基、环丁基、-O环丙基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代;
所述化合物满足以下条件之一:
(a)在一些实施方案中,R1或R2为C1-4烷氧基;
(b)R3为C1-4烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
(c)m为1;
(d)环B为吡唑基、咪唑基、噻唑基或噻吩基;
(e)至少存在一个R6不为卤素和C1-4烷基;
(f)环A为
(g)至少一个R7为C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、环丙基、环丁基、-O环丙基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧 杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基或哒嗪基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、环丙基和环丁基的基团取代;
其余基团如第二或第四技术方案所述。
所述C1-4烷基包括但不限于甲基、乙基、丙基、异丙基、丁基、异丁基、正丁基;所述C2-4烯基包括但不限于乙烯基、丙烯基、烯丙基、1-丁烯基、2-丁烯基、3-丁烯基;所述C2-4炔基包括但不限于乙炔基、丙炔基、炔丙基、1-丁炔基、2-丁炔基、3-丁炔基;所述C3-6环烷基包括但不限于环丙基、环丁基、环戊基、环己基;所述杂环烷基包括但不限于氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、氮杂环己烯基、氧杂环己烯基;所述杂芳基包括但不限于吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基、噻唑基、噻吩基、噁唑基、异噁唑基、噁二唑基、嘧啶并吡唑、吡嗪并吡唑、嘧啶并咪唑、吡嗪并咪唑、吲哚基、嘌呤基等。
本发明提供了选自以下I组中结构之一的化合物,其立体异构体、氘代化物或药学上可接受的盐:
I组:










本发明还提供了选自以下II组结构之一的化合物,其立体异构体、氘代化物或药学上可接受的盐:
II组:








本发明还提供了一种药物组合物,其含有治疗有效量的本发明所述的任一化合物,或其立体异构体、氘代化物或药学上可接受的盐,以及药学上可接受的载体和/或辅料。
本发明还涉及一种制药用途,即本发明所述的任一化合物,或其立体异构体、氘代化物或药学上可接受的盐,或者含有它们的组合物在制备治疗Polθ介导的疾病的药物中的用途。本发明还涉及一种治疗用途,即本发明所述的任一化合物,或其立体异构体、氘代化物或药学上可接受的盐,或者含有它们的组合物在治疗Polθ介导的疾病的中的用途。
所述Polθ介导的疾病包括但不限于肝癌、乳腺癌、卵巢癌、肺癌、肾癌、前列腺癌、皮肤癌、膀胱癌、胰腺癌或头颈癌。
本发明涉及一种药物组合物或药物制剂,所述的药物组合物或药物制剂包含治疗有效量的本发明所述的化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐和载体和/或辅料。该药物组合物可以为单位制剂形式(单位制剂中主药的量也被称为“制剂规格”)。
本发明还提供一种用于治疗哺乳动物的疾病的方法,其包括向所述哺乳动物给予治疗有效量的本发明所述的化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐或药物组合物。一些实施方案中,本发明中所述哺乳动物包括人。
本申请中所述“有效量”或“治疗有效量”是指给予足够量的本申请公开的化合物,其将在某种程度上缓解所治疗的疾病或病症的一种或多种症状。在一些实施方案 中,结果是减少和/或缓和疾病的体征、症状或原因,或生物系统的任何其它希望改变。例如,针对治疗用途的“有效量”是提供临床上显著的疾病症状降低所需的包含本申请公开的化合物的组合物的量。治疗有效量的实例包括但不限于:1-1000mg、1-900mg、1-800mg、1-700mg、1-600mg、1-500mg、1-400mg、1-300mg、1-250mg、1-200mg、1-150mg、1-125mg、1-100mg、1-80mg、1-60mg、1-50mg、1-40mg、1-25mg、1-20mg、5-1000mg、5-900mg、5-800mg、5-700mg、5-600mg、5-500mg、5-400mg、5-300mg、5-250mg、5-200mg、5-150mg、5-125mg、5-100mg、5-90mg、5-70mg、5-80mg、5-60mg、5-50mg、5-40mg、5-30mg、5-25mg、5-20mg、10-1000mg、10-900mg、10-800mg、10-700mg、10-600mg、10-500mg、10-450mg、10-400mg、10-300mg、10-250mg、10-200mg、10-150mg、10-125mg、10-100mg、10-90mg、10-80mg、10-70mg、10-60mg、10-50mg、10-40mg、10-30mg、10-20mg;20-1000mg、20-900mg、20-800mg、20-700mg、20-600mg、20-500mg、20-400mg、20-350mg、20-300mg、20-250mg、20-200mg、20-150mg、20-125mg、20-100mg、20-90mg、20-80mg、20-70mg、20-60mg、20-50mg、20-40mg、20-30mg;50-1000mg、50-900mg、50-800mg、50-700mg、50-600mg、50-500mg、50-400mg、50-300mg、50-250mg、50-200mg、50-150mg、50-125mg、50-100mg;100-1000mg、100-900mg、100-800mg、100-700mg、100-600mg、100-500mg、100-400mg、100-300mg、100-250mg、100-200mg;
在一些实施方案中,该药物组合物包括但不限于:1-1000mg、5-500mg、10-250mg、50-250mg、100-200mg、1mg、1.25mg、2.5mg、5mg、10mg、12.5mg、15mg、20mg、25mg、30mg、35mg、40mg、45mg、50mg、60mg、70mg、80mg、90mg、100mg、120mg、125mg、150mg、200mg、250mg、300mg、400mg、500mg、600mg、700mg、800mg、900mg、1000mg的本发明化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐。
一种用于治疗哺乳动物的疾病的方法,所述方法包括给予受试者治疗有效量的本发明化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐,治疗有效量优选1-1000mg,所述的疾病优选肝癌、乳腺癌、卵巢癌、肺癌、肾癌、前列腺癌、皮肤癌、膀胱癌、胰腺癌或头颈癌。
一种用于治疗哺乳动物的疾病的方法所述方法包括,将药物本发明化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐以1-1000mg/天的日剂量给予受试者,所述日剂量可以为单剂量或分剂量,在一些实施方案中,日剂量包括但不限于:1-1000mg/天、1-300mg/天、5-500mg/天、10-500mg/天、10-400mg/天、10-300mg/天、10-100mg/天、20-400mg/天、20-200mg/天、20-100mg/天、50-500mg/天、50-250mg/天、50-200mg/天、50-150mg/天、50-100mg/天、100-500mg/天、100-300mg/天、100-200mg/天;
在一些实施方案中,日剂量包括但不限于:1mg/天、2.5mg/天、5mg/天、10mg/天、12.5mg/天、15mg/天、20mg/天、25mg/天、30mg/天、35mg/天、40mg/天、45mg/ 天、50mg/天、60mg/天、70mg/天、80mg/天、90mg/天、100mg/天、120mg/天、150mg/天、200mg/天、250mg/天、300mg/天、400mg/天、500mg/天、1000mg/天。
本发明涉及一种试剂盒,该试剂盒可以包括单剂量或多剂量形式的组合物,该试剂盒包含本发明化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐,本发明化合物的或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐量与上述药物组合物中其量相同。
本发明中本发明化合物或者其立体异构体、氘代物、溶剂化物、药学上可接受的盐的量在每种情况下以游离碱的形式换算。
“制剂规格”是指每一支、片或其他每一个单位制剂中含有主药的重量。
合成路线
本领域技术人员可以结合已知的有机合成技术制备本发明的化合物,其起始原料为市售化学品和(或)化学文献中所述的化合物。“市售化学品”是从正规商业来源获得的,供应商包括:泰坦科技、安耐吉化学、上海德默、成都科龙化工、韶远化学科技、南京药石、药明康德和百灵威科技等公司。
通过美国化学会化学文摘社制备的已知化学物质的索引,可以选择性地识别特定和类似的反应物,这些索引可在大多数公共图书馆和大学图书馆以及在线获得。已知但在目录中不可商购的化学品可选地由定制化学合成工厂制备,其中许多标准化学供应工厂提供定制合成服务。
术语
在本发明未特殊说明的情况下,本发明的术语具有以下含义:
本发明所述基团和化合物中所涉及的碳、氢、氧、硫、氮或卤素均包括它们的同位素,及本发明所述基团和化合物中所涉及的碳、氢、氧、硫、氮或卤素任选进一步被一个或多个它们对应的同位素所替代,其中碳的同位素包括12C、13C和14C,氢的同位素包括氕(H)、氘(氘,又称为重氢)、氚(T,又称为超重氢),氧的同位素包括16O、17O和18O,硫的同位素包括32S、33S、34S和36S,氮的同位素包括14N和15N,氟的同位素19F,氯的同位素包括35Cl和37Cl,溴的同位素包括79Br和81Br。
“卤素”在本文中是指F、Cl、Br、I或者它们的同位素。
“卤代”或“卤素取代”是指被一个以上选自F、Cl、Br、I或者它们的同位素取代,卤素取代基数量的上限等于被取代基团可被取代的氢数之和,在未作特殊限定下,卤素取代基数量为1至该上限之间的任意整数,当卤素取代基数量大于1时,可以是相同或不同的卤素进行取代。通常包括1-5个卤素取代、1-3卤素取代、1-2个卤素取代、1个卤素取代的情形。
“D”、“氘”是指氢(H)的同位素氘。
“氘代”或“氘代物”是指烷基、环烷基、亚烷基、芳基、杂芳基、巯基、杂环烷基、烯基、炔基等基团上的氢原子被至少一个氘原子取代的情形,氘代的数量上限等于被取代基团可被取代的氢数之和,在未作特殊限定下,氘代数量为1至该上限之间的任意整数,例如1-20个氘原子取代、1-10个氘原子取代、1-6个氘原子取代、1-3个氘原子取代、1-2个氘原子取代或1个氘原子取代。
“Cx-y”基团是指包含x至y个碳原子的基团,比如“C1-6烷基”指包含1-6个碳原子的烷基。
“烷基”是指一价的直链或支链饱和脂肪族烃基。通常为1至20个碳原子的烷基,或者1至8个碳原子的烷基,或者1至6个碳原子的烷基,或者1至4个碳原子的烷基。非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、仲丁基、新丁基、叔丁基、正戊基、异戊基、新戊基、正己基等,烷基可以进一步被取代基取代。
“亚烷基”是指二价的直链和支链饱和烷基。亚烷基实施例包括但不限于亚甲基、亚乙基等。
“卤代烷基”是指烷基中的一个或多个氢被一个或多个卤素原子(如氟、氯、溴、碘或其同位素)替代的情形,卤素取代基的数量的上限等于烷基中可被取代的氢数之和,在未作特殊限定下,卤素取代基数量为1至该上限之间的任意整数。通常烷基被1-5个卤素取代或者1-3卤素取代或者1-2个卤素取代或1个卤素取代;当卤素取代基数量大于1时,可以是相同或不同的卤素进行取代;具体示例包括但不限于-CF3、-CH2Cl、-CH2CF3、-CCl2、CF3等。
“烷氧基”或“烷基氧基”是指-O-烷基。例如-O-C1-8烷基、-O-C1-6烷基、-O-C1-4烷基或-O-C1-2烷基。具体的非限制性实施例包括甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基、正己氧基、环丙氧基和环丁氧基等;所述的烷氧基可以任选被取代基取代。
“卤代烷氧基”是指-O-卤代烷基。例如-O-卤代C1-8烷基、-O-卤代C1-6烷基、-O-卤代C1-4烷基或-O-卤代C1-2烷基;卤素取代基的数量的上限等于被取代基团可被取代的氢数之和,在未作特殊限定下,卤素取代基数量为1至该上限之间的任意整数,优选1-5个卤素取代、1-3卤素取代、1-2个卤素取代、1个卤素取代;当卤素取代基数量大于1时,可以是相同或不同的卤素进行取代;非限制性实施例包括一氟甲氧基、二氟甲氧基、三氟甲氧基、二氟乙基氧基等。
“烯基”是指包含至少一个碳碳双键(C=C)的直链烃基或支链烃基,通常包含2至18个碳原子,如2至8个碳原子,进一步如2至6个碳原子,再进一步如2至4个碳原子,其示例包括但不限于乙烯基、烯丙基、1-丙烯基、2-丙烯基、1-丁烯基、2-丁烯基、3-丁烯基、1-戊烯基、2-戊烯基、3-戊烯基、4-戊烯基、1-甲基-1-丁烯基、2-甲基-1-丁烯基、2-甲基-3-丁烯基、1-己烯基、2-己烯基、3-己烯基、4-己烯基、5-己烯基、1-甲基-1-戊烯基、2-甲基-1-戊烯基、1-庚烯基、2-庚烯基、3-庚烯基、4-庚烯基、1-辛烯基、3-辛烯基、1-壬烯基、3-壬烯基、1-癸烯基、4-癸烯基、1,3-丁二烯、1,3-戊二烯、1,4-戊二烯和1,4-己二烯等;所述的烯基可以任选进一步被取代基取代。
“亚烯基”是指直链或支链的、含有至少一个碳碳双键(C=C)的二价不饱和烃基,通常包含2至18个碳原子,如2至8个碳原子,进一步如2至6个碳原子,再进一步如2至4个碳原子,非限制性实施例包括亚乙炔基,所述的亚烯基可以任选地被取代基取代。
“炔基”是指含有至少一个碳碳三键(C≡C)的直链烃基或支链烃基,通常包含2至18个碳原子,进一步包含2至8个碳原子,进一步包含2至6个碳原子,再进一步包含2至4个的碳原子,其示例包括但不限于乙炔基、1-丙炔基、2-丙炔基、丁炔基、2-丁炔基、3-丁炔基、1-甲基-2-丙炔基、4-戊炔基、3-戊炔基、1-甲基-2-丁炔基、2-己炔基、3- 己炔基、2-庚炔基、3-庚炔基、4-庚炔基、3-辛炔基、3-壬炔基和4-癸炔基等;所述的炔基可以任选地被取代基取代。
“亚炔基”是指直链或支链的、含有碳碳三键(C≡C)的二价不饱和烃基,,通常包含2至18个碳原子,进一步包含2至8个碳原子,进一步包含2至6个碳原子,进一步包含2-4个碳原子,非限制性实施例包括亚乙炔基、亚丙炔基、亚丁炔基,所述的亚炔基可以任选地被取代基取代。
“环烷基”是指饱和或部分不饱和的、不含环杂原子的、非芳香性的碳环烃基。环烷基可以是单环、双环或多环,双环或多环可以是并环、螺环、桥环或其组合形式,双环或多环中可以包括一个及以上的芳环,但环系统整体不具有芳香性,连接位点可以在芳环上或非芳香环上。通常环烷基含有3至20个碳原子,进一步含有3-8个碳原子,更进一步含有3-6个碳原子;当为单环环烷基时,含有3-15个碳原子,或者3-10个碳原子,或者3-8个碳原子,或者3-6个碳原子;当为双环或多环环烷基时,含有5-12个碳原子,或者含有5-11个碳原子,或者含有6-10个碳原子;非限制性实施例包括环丙基、环丁基、环戊基、环己基、环庚基、丁烯基、环戊烯基、环己烯基、 等,环烷基可以任选地被取代基取代。
“亚环烷基”是指环烷基的二价基团。
“芳基”是指具有芳香性不含有杂原子的碳环,包括单环芳基和稠环芳基。通常含有6至14个碳原子,进一步含有6至10个碳原子。非限制性实施例包含苯基、萘基、葸基、菲基,芳基可以任选地被取代基取代。
“碳环”或“碳环基”是指饱和的、部分不饱和的或者芳香的碳环,其含义包括芳基和环烷基。碳环可以是单环、双环或多环,双环或多环包括桥环、并环和螺环以及它们的组合形式。碳环通常有3至12个碳原子,或者3-10个碳原子,或者3-6个碳原子。非限制性实施例中,单环碳环包括环丙基、环丁基、环戊基、环己基、环庚基或苯基等,双环桥环包括等,双环并环包括 等,双环螺环包括 等,碳环可以任选被取代基所取代。
“杂环烷基”是指包含1、2、3或者4个选自N、S、O杂原子的饱和或部分不饱和的非芳香性碳环。杂环烷基可以是单环、双环或多环,双环或多环可以是桥环、并环、螺环或其组合形式,双环或多环中可以包括一个及以上的芳环或杂芳环,但环系统整体不具有芳香性,连接位点可以在芳香环上或非芳香环上。通常杂环烷基为3至20元环,当为单环杂环烷基时,通常为3至15元环,或者3-10元环,或者3-8元环,或者3-6元环;当为双环或多环环杂环烷基时,通常为5-12元环,或者5-11元环,或者6-9元环。其中的杂原子N、S包括其氧化态。杂环烷基的非限制性实施例包括氮杂环丁 基、吗啉基、哌嗪基、哌啶基、四氢吡喃基、氧杂环丁基、吡喃基、氮杂环戊烯基、氮杂环己烯基、氧杂环戊烯基、氧杂环己烯基等,杂环烷基可以任选地被取代基取代。
“杂芳环”或“杂芳基”未特殊说明时,是指包含1至4个选自N、O或S及其氧化态的杂原子且具有芳香性的环,可以是单环、双环或多环,双环或多环可以是桥环、并环、螺环以及它们的组合形式;当为双环或多环时,可以是杂芳基与芳基稠和,也可以是杂芳基与杂芳基的稠和,其中杂芳基或芳基均可为连接位点。非限制性实施例包括呋喃基、噻吩基、吡咯基、噁唑基、噻唑基、咪唑基、吡唑基、吡啶基、嘧啶基、哒嗪基、吡嗪基、吲哚基、嘌呤基、 等;所述的杂芳基可以任选被取代基所取代。
“杂环”或“杂环基”是指饱和或不饱和的、芳香或者非芳香的、包含1至4个选自N、O或S及其氧化态的杂原子的环,其含义包括杂芳基和杂环烷基。杂环包括单环杂环、双环桥杂环、双环并杂环和双环螺杂环或其组合形式。通常为3至12元杂环或者5至12元杂环,或者5至7元杂环。杂环基可以连接在杂原子或者碳原子上,非限制性实施例包括环氧乙基、氮杂环丙基、氧杂环丁基、氮杂环丁基、1,3-二氧戊环基、1,4-二氧戊环基、1,3-二氧六环基、哌嗪基、氮杂环庚基、吡啶基、呋喃基、噻吩基、吡喃基、N-烷基吡咯基、嘧啶基、吡嗪基、吡唑基、哒嗪基、咪唑基、哌啶基、哌叮基、吗啉基、硫代吗啉基、1,3-二噻基、二氢呋喃基、二氢吡喃基、二噻戊环基、四氢呋喃基、四氢吡咯基、四氢咪唑基、噁唑基、二氢噁唑基、四氢噁唑基、四氢噻唑基、四氢吡喃基、苯并咪唑基、苯并吡啶基、吡咯并吡啶基、苯并二氢呋喃基、氮杂二环[3.2.1]辛烷基、氮杂二环[5.2.0]壬烷基、氧杂三环[5.3.1.1]十二烷基、氮杂金刚烷基和氧杂螺[3.3]庚烷基、 等,杂环可以任选被取代基所取代。
“亚杂环基”是指取代或未取代、饱和或不饱和、芳香或者非芳香的二价杂环基团。非限制性实施例包括等。
“螺环”是指环与环之间共用一个碳原子(称螺原子)的多环基团,其可以包含0或1个以上的双键或三键,可以含有0至5个选自N、O、S、P、Si及其氧化态的杂原子。通常螺环为6至14元环,或者6至12元环,或者6至10元环。通常螺环为三螺三(表示三元环螺三元环)、三螺四、三螺五、三螺六、四螺四、四螺五、四螺六、五螺五或者五螺六。螺环的其非限定性实例包括 所述的螺环可以任选被取代基所取代。
“并环”或“稠环”是指环与环共享毗邻的两个环原子和一个化学键的多环基团,可以含有一个或多个双键或三键,并环可以含0至5个选自N、S、O、P、Si及其氧化态的杂原子。通常并环为5至20元环,或者5至14元环,或者5至12元环,或者5至10元环。通常并环为三并四环(表示三元环与四元环形成的并环,根据IUPC命名规则有可能是三元环作为基本环也可能是四元环作为基本环的并环,以下同理)、三并五环、三并六环,四并四环、四并五环、四并六环、五并五环、五并六环、六并六环。并环的非限定性实例包括嘌呤、喹啉、异喹啉、苯并吡喃、苯并呋喃、苯并噻吩、;所述的并环可以是芳香的或非芳香的,并任选被取代基所取代。
“双环杂并环”是指两个环共享毗邻的两个环原子和一个化学键的双环基团,可以含有一个或多个双键或三键,含1至5个选自N、S、O、P、Si及其氧化态的杂原子,可以是饱和的、部分饱和的或者是芳香的,其含义包括双环并环杂环烷基和双环并环杂芳基。通常并环为5至20元环,或者5至14元环,或者5至12元环,或者8至10元环。通常并环为六并五环(表示六元环与五元环形成的并环,根据IUPC命名规则有可能是六元环作为基本环也可能是五元环作为基本环的并环,以下同理)、六并六环、五并五环、四并四环、四并五环、四并六环、五并五环、五并六环、六并六环。所述的双环杂并环任选被取代基所取代。
“桥环”是指两个环之间共享两个不相邻的环原子,可以含有1个或多个双键或三键。桥环可以含0至5个选自N、S、O、P、Si及其氧化态的杂原子。通常桥环的环原子为5至 20个,或者5至14个,或者5至12个,或者5至10个。桥环的非限定性实例包括金刚烷、
“取代”或“取代基”在未特殊说明时,是指在化学理论允许的位置发生任意取代,取代基个数符合化学键规则。示例性的取代基包括但不限于:C1-6烷基、C2-6烯基、C2-6炔基、C3-8杂烷基、C5-12芳基、5-12元杂芳基、羟基、C1-6烷氧基、C5-12芳氧基、硫醇基、C1-6烷硫基、氰基、卤素、C1-6烷硫代羰基、C1-6烷氨基甲酰基、N-氨基甲酰基、硝基、甲硅烷基、亚磺酰基、磺酰基、亚砜、卤代C1-6烷基、卤代C1-6烷氧基、氨基、膦酸、-CO2(C1-6烷基),-OC(=O)(C1-6烷基),-OCO2(C1-6烷基),-C(=O)NH2,-C(=O)N(C1-6烷基)2,-OC(=O)NH(C1-6烷基),-NHC(=O)(C1-6烷基),-N(C1-6烷基)C(=O)(C1-6烷基),-NHCO2(C1-6烷基),-NHC(=O)N(C1-6烷基)2,-HC(=O)NH(C1-6烷基),-NHC(=O)NH2,-NHSO2(C1-6烷基),-SO2N(C1-6烷基)2,-SO2NH(C1-6烷基),-SO2NH2,-SO2C1-6烷基等。
“任选”或“任选地”是指随后所描述的事件或环境可以但不必须发生,该说明包括该事件或环境发生或不发生的场合。如:“任选被F取代的烷基”指烷基可以但不必须被F取代,说明包括烷基被F取代的情形和烷基不被F取代的情形。
“药学上可接受的盐”是指本发明化合物保持游离酸或者游离碱的生物有效性和特性,且所述的游离酸通过与无毒的无机碱或者有机碱,所述的游离碱通过与无毒的无机酸或者有机酸反应获得的盐。
“药物组合物”表示一种或多种本文所述化合物或其立体异构体、氘代化物、溶剂化物、药学上可接受的盐或共晶,与其他组成成分的混合物,其中其他组分包含生理学/药学上可接受的载体和/赋形剂。
“载体”指的是:不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性,并能改变药物进入人体的方式和在体内的分布、控制药物的释放速度并将药物输送到靶向器官的体系,非限制性的实例包括微囊与微球、纳米粒、脂质体等。
“赋形剂”指的是:其本身并非治疗剂,用作稀释剂、辅料、粘合剂和/或媒介物,用于添加至药物组合物中以改善其处置或储存性质或允许或促进化合物或药物组合物形成用于给药的单位剂型。如本领域技术人员所已知的,药用赋形剂可提供各种功能且可描 述为润湿剂、缓冲剂、助悬剂、润滑剂、乳化剂、崩解剂、吸收剂、防腐剂、表面活性剂、着色剂、矫味剂及甜味剂。药用赋形剂的实例包括但不限于:(1)糖,例如乳糖、葡萄糖及蔗糖;(2)淀粉,例如玉米淀粉及马铃薯淀粉;(3)纤维素及其衍生物,例如羧甲基纤维素钠、乙基纤维素、乙酸纤维素、羟丙基甲基纤维素、羟丙基纤维素、微晶纤维素及交联羧甲基纤维素(例如交联羧甲基纤维素钠);(4)黄蓍胶粉;(5)麦芽;(6)明胶;(7)滑石;(8)武形剂,例如可可脂及栓剂蜡;(9)油,例如花生油、棉籽油、红花油、芝麻油、橄榄油、玉米油及大豆油;(10)二醇,例如丙二醇;(11)多元醇,例如甘油、山梨醇、甘露醇及聚乙二醇;(12)酯,例如油酸乙酯及月桂酸乙酯;(13)琼脂;(14)缓冲剂,例如氢氧化镁及氢氧化铝;(15)海藻酸;(16)无热原水;(17)等渗盐水;(18)林格溶液(Ringer’ssolution);(19)乙醇;(20)pH缓冲溶液;(21)聚酯、聚碳酸酯和/或聚酐;及(22)其他用于药物制剂中的无毒相容物质。
“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体和构象异构体。
本发明的化合物还包括其互变异构体,例如当本发明阐述嘧啶环被OH取代的左侧化合物时,也同时包括右侧的互变异构体化合物。
“溶剂化物”指本发明化合物或其盐与分子间非共价力结合的化学计量或非化学计量的溶剂形成的物质。当溶剂为水时,则为水合物。
“共晶”是指活性药物成分(API)和共晶形成物(CCF)在氢键或其他非共价键的作用下结合而成的晶体,其中API和CCF的纯态在室温下均为固体,并且各组分间存在固定的化学计量比。共晶是一种多组分晶体,既包含两种中性固体之间形成的二元共晶,也包含中性固体与盐或溶剂化物形成的多元共晶。
“X2不存在”表示部分为亚甲基。
具体实施方式
以下将通过实施例对本发明的内容进行详细描述。实施例中未注明具体条件的,按照常规条件的实验方法进行。所举实施例是为了更好地对本发明的内容进行说明,但并不能理解为本发明的内容仅限于所举实例。本领域常规技术人员根据上述发明内容对实施方案进行非本质的改进和调整,仍属于本发明的保护范围。
测试方法
化合物的结构是通过核磁共振(NMR)或(和)质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker Avance III 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS);
MS的测定用(Agilent 6120B(ESI)和Agilent 6120B(APCI));
HPLC的测定使用Agilent 1260DAD高压液相色谱仪(Zorbax SB-C18 100×4.6mm,3.5μM);
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm-0.20mm,薄层层析分离纯化产品采用的规格是0.4mm-0.5mm;
柱层析一股使用烟台黄海硅胶200-300目硅胶为载体。
缩写
XantPhos:4,5-双二苯基膦-9,9-二甲基氧杂葸
T3P:1-丙基磷酸酐
实施例1:
第一步:将化合物1A(0.2g,0.55mmol)(参考专利WO2021028670中描述方法合成)溶于1,4-二氧六环(10mL)中,依次向其中加入化合物1B(0.13g,0.66mmol),Pd2(dba)3(0.056g,0.06mmol),XantPhos(0.1g,0.17mmol),碳酸钾(0.11g,0.83mmol),均匀搅拌,氮气氛围下,升温至95℃反应18小时。待反应完全后,冷却至室温,反应液通过硅藻土过滤,将滤液倒入水(10mL)中,用乙酸乙酯萃取(20mL×3),合并后的有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩后得到粗品,粗品用Biotage Isolera OneTM(12g硅胶柱,洗脱剂:0-27%EA/PE)纯化得到目标化合物1C(110mg,收率38.2%)。LC-MS(ESI):m/z=524.1[M+H]+.
第二步:将化合物1C(0.11g,0.21mmol)溶于二氯甲烷(10mL),冷却至-20℃,缓慢滴加三氯化硼(0.075g,0.63mmol),自然升至室温搅拌3小时。滴加到冰的饱和碳酸氢钠水溶液(10mL)淬灭反应,残余物用乙酸乙酯(20mL×3),合并后的有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用Biotage Isolera OneTM(12g硅胶柱,洗脱剂:0-5%MeOH/DCM)纯化得到目标化合物1(30mg,收率28.2%)。LC-MS(ESI):m/z=484.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.50-8.47(m,1H),7.99-7.83(m,1H),7.50-7.19(m,1H),5.44-4.73(m,1H),4.53-4.20(m,2H),2.71-2.09(m,3H).
实施例2:
第一步:将化合物2A(0.5g,2.42mmol)溶于1,4-二氧六环(8mL),依次加入化合物2B(0.6g,2.90mmol),PdCl2(dppf)(0.18g,0.24mmol),碳酸钠(0.51g,4.84mmol),水(2mL),氮气氛围下,升温至100℃搅拌18小时。待反应完全后,冷却至室温,反应液经硅藻土过滤,得到的滤液倒入水(10mL)中,用乙酸乙酯萃取(20mL×3),合并 后的有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用Biotage Isolera OneTM(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到目标化合物2C(500mg,收率99.5%)。LC-MS(ESI):m/z=208.1[M+H]+.
第二步:以2C和1A为原料,参考实施例1第一步操作得到化合物2D。LC-MS(ESI):m/z=535.1[M+H]+.
第三步:以2D为原料,参考实施例1第二步操作得到化合物2。LC-MS(ESI):m/z=495.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.42-8.36(m,1H),8.16-7.91(m,3H),7.56-7.49(m,1H),7.30-7.17(m,1H),5.82-4.91(m,1H),4.58-4.25(m,2H),3.96(s,3H),2.62-2.43(m,3H).
实施例3:
第一步:将化合物3A(2.2g,10.14mmol)溶于甲苯(30mL)中,依次向其中加入氟硼酸钾(1.48g,12.17mmol),四三苯基磷钯(0.59g,0.51mmol),水(3mL),氮气氛围下,升温至90℃搅拌18小时。待反应完全后,冷却至室温,反应液经硅藻土过滤,得到的滤液倒入水(10mL)中,用乙酸乙酯萃取(20mL×3),合并后的有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用Biotage Isolera OneTM(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到目标化合物3B(600mg,收率30.1%)。
1H NMR(400MHz,CDCl3)δ7.41(s,1H),2.65(m,3H).
第二步:以3B和1A为原料,参考实施例1第一步操作得到化合物3C。LC-MS(ESI):m/z=524.1[M+H]+.
第三步:以3C为原料,参考实施例1第二步操作得到目标化合物3。LC-MS(ESI):m/z=495.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.41-8.39(m,1H),7.80-7.16(m,2H),5.41-4.78(m,1H),4.49-4.15(m,2H),2.52-2.49(m,3H).
实施例4:
第一步:以4B和4A(参考专利WO2021028643中描述方法合成)为原料,参考实施例1第一步操作得到目标化合物4C。LC-MS(ESI):m/z=492.1[M+H]+.
第二步:以4C为原料,参考实施例1第二步操作得到目标化合物4。LC-MS(ESI):m/z=452.1[M+H]+.
1H NMR(400MHz,CD3OD-d4)δ8.14-8.11(m,1H),7.99-7.76(m,2H),7.57-7.24(m,2H),5.25-4.89(m,1H),4.38-4.12(m,2H),3.55-2.15(m,3H),2.37-2.34(m,3H).
实施例5:
第一步:将5A(2g,7.86mmol)、3,3-二甲基氮杂环丁烷(0.73g,7.86mmol)溶于甲苯(50mL)中,随后加入醋酸钯(0.18g,0.79mmol)、(±)-2,2′-双(二苯基膦基)-1,1′-联萘(0.98g,1.57mmol)及碳酸铯(5.12g,15.72mmol),氮气换气保护,于60℃下搅拌16h。待反应冷却至室温,向反应液中加入100mL水,以乙酸乙酯(40mL×3)萃取,合并有机层,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到标题化合物5B(1.82g,87%)。LC-MS(ESI):m/z=267.1[M+H]+.
第二步:将5B(1.82g,6.83mmol)溶于乙醇(50mL)及水(50mL)的混合溶剂中,加入锌粉(2.23g,34.15mmol)及氯化铵(3.65g,68.30mmol),80℃反应1h。冷却至室温,过滤除去不溶物,以乙酸乙酯洗滤饼,减压浓缩除去滤液中大部分有机溶剂,向残余物中加入50mL水,以乙酸乙酯(40mL×3)萃取,合并有机层,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到标题化合物5C(1.5g,93%)。LC-MS(ESI):m/z=237.1[M+H]+.
第三步:将5C(1.5g,6.34mmol)溶于甲苯(50mL)中,依次加入氢氧化钠(2.54g,63.40mmol)、四丁基溴化铵(0.20g,0.63mmol)及硫酸二甲酯(0.88g,6.97mmol),室温反应过夜。向反应液中加入100mL水,以乙酸乙酯(40mL×3)萃取,合并有机层,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到标题化合物5D(0.63g,40%)。LC-MS(ESI):m/z=251.1[M+H]+.
第四步:将5E(0.4g,1.99mmol)(参考专利WO2021028670中描述方法合成)及5D(0.6g,2.39mmol)溶于吡啶(10mL)中,室温缓慢滴加50%T3P的乙酸乙酯溶液(7.97mmol),50℃反应过夜。待反应完全后,向反应液中加入50mL水,以乙酸乙酯(30mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到标题化合物5F(0.35g,40%)。LC-MS(ESI):m/z=434.1[M+H]+.
第五步:以5F和2-氯-6-甲基-4-(三氟甲基)吡啶为原料,参考实施例1第一步操作得到目标化合物5G。LC-MS(ESI):m/z=593.1[M+H]+.
第六步:以5G为原料,参考实施例1第二步操作得到目标化合物5。
1H NMR(400MHz,CDCl3)δ8.40(s,1H),7.69-7.68(m,1H),7.04(s,1H),6.32-6.31(m,1H),5.26-5.25(m,1H),4.42-4.40(m,4H),4.34-4.27(m,1H),4.19-4.18(m,1H),3.20(s,3H),2.39(s,3H).LC-MS(ESI):m/z=553.1[M+H]+.
实施例6:
第一步:将5E(2g,9.94mmol)溶于DMF(25mL)中,加入碳酸钾(2.06g,14.91mmol)后,缓慢滴加苄溴(1.70g,9.94mmol),室温反应过夜。16h后,向反应液中加入50mL水,乙酸乙酯(20mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到标题化合物6A(2.16g,69%)。LC-MS(ESI):m/z=292.1[M+H]+.
第二步:将6A(2.16g,7.41mmol)及2-氯-6-甲基-4-(三氟甲基)吡啶(1.74g,8.90mmol)溶于1,4-二氧六环(50mL)中,依次加入Pd2(dba)3(679mg,0.74mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(858mg,1.48mmol)及碳酸钾(2.05g,14.83mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入100mL水,乙酸乙酯(30mL×3)萃取,合并有机层,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=3∶1)得到标题化合物6B(1.76g,56%)。LC-MS(ESI):m/z=451.1[M+H]+.
第三步:将6B(1.76g,3.91mmol)溶于甲醇(20mL)及四氢呋喃(10mL)的混合溶剂中,加入10%钯碳粉末(200mg),氢气氛围下反应过夜。待反应完全后,过滤,少量甲醇洗滤饼,将滤液减压浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到6C(1.36g,96%)。LC-MS(ESI):m/z=361.0[M+H]+.
第四步:将6C(0.40g,1.11mmol)溶于吡啶(10mL)中,加入2-(7-偶氮苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(0.55g,1.44mmol)和5-氨基-2,4-二氟苄腈(0.26g,1.69mmol),65℃反应1小时。浓缩,残留物用硅胶柱色谱分离提纯(PE∶EA(v/v)=5∶1-2∶1)得到标题化合物6D(0.15g,27.2%)。LC-MS(ESI):m/z=497.0[M+H]+.
第五步:将6D(0.15g,0.3mmol)溶于四氢呋喃(10mL)中,0℃下加入氢化钠(0.014g,0.58mmol),搅拌5分钟后,加入碘甲烷(0.085g,0.60mmol),室温反应1小时。加入水(10mL)淬灭反应,EA(20×3mL)萃取,合并后的有机相用饱和氯化钠 洗(10mL),无水硫酸钠干燥,过滤,浓缩得到标题化合物6E(0.1g,65.3%)。LC-MS(ESI):m/z=511.1[M+H]+.
第六步:将6E(0.10g,0.20mmol)溶于盐酸二氧六环溶液(10mL,4M)中,室温反应16小时。浓缩,残留物用硅胶柱色谱分离提纯(DCM∶MeOH(v/v)=40∶1-10∶1)得到标题化合物6(0.03g,31.9%)。
1H NMR(400MHz,CDCl3)δ8.55(s,1H),8.18-8.15(m,1H),7.29-7.27(m,1H),7.17(s,1H),4.69-4.68(m,1H),4.36-4.34(m,1H),3.73(s,1H),3.29(s,3H),2.66(s,3H).LC-MS(ESI):m/z=471.1[M+H]+.
实施例7:
以6C和咪唑[1,2-A]吡啶-6-氨基为原料,参考实施例6操作(第四,五,六步反应),得到化合物7。
1H NMR(400MHz,CDCl3)δ8.57-8.55(m,1H),8.45(s,1H),7.86(s,1H),7.78(s,1H),7.58(s,1H),7.19(s,1H),7.15-7.13(m,1H),5.01(s,1H),4.46-4.41(m,2H),3.27(s,3H),2.58(s,3H).LC-MS(ESI):m/z=450.2[M+H]+.
实施例8:
第一步:将1-溴-2,4-二氟-5-硝基苯(8A)(1.0g,4.21mmol)溶于二氧六环(15mL)和水(3mL)中,依次加入3-噻吩硼酸(0.81g,6.33mmol),Pd(dppf)Cl2(0.62g,0.84mmol)和碳酸钾(1.16g,8.39mmol),氮气氛围,100℃反应3小时。待反应完全后,冷至室温,过滤,浓缩,残留物用硅胶柱色谱分离提纯(PE∶EA(v/v)=10∶1-5∶1)得到标题化合物8B(0.80g,78.8%)。
第二步:将8B(0.80g,3.32mmol)溶于乙醇(10mL)和水(5mL)中,加入铁粉(0.92g,16.54mmol)和氯化铵(1.78g,33.27mmol),60℃反应1小时。待反应冷至室温,过滤,浓缩,残留物用硅胶柱色谱分离提纯(PE∶EA(v/v)=5∶1-2∶1)得到标题化合物8C(0.51g,72.7%)。LC-MS(ESI):m/z=212.1[M+H]+.
第三步:将8C(0.26g,1.23mmol)溶于吡啶(5mL)中,加入6C(0.30g,0.83mmol)和1-丙基磷酸酐(1.58g,2.49mmol),50℃反应16小时。待反应冷至室温,加入水(50mL),EA(20×3mL)萃取,合并后的有机相用饱和氯化钠洗,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱色谱分离提纯(PE∶EA(v/v)=10∶1-2∶1)得到标题化合物8D(0.25g,54.4%)。LC-MS(ESI):m/z=554.0[M+H]+.
第四步:将8D(0.25g,0.45mmol)溶于N,N-二甲基甲酰胺(10mL)中,0℃下加入氢化钠(0.022g,0.90mmol),搅拌5分钟后,加入氘代碘甲烷(0.078g,0.54mmol),室温反应1小时。加入水(10mL)淬灭反应,EA(20×3mL)萃取,合并后的有机相用饱和氯化钠洗,无水硫酸钠干燥,浓缩得到标题化合物8E(0.15g,58.4%)。LC-MS(ESI):m/z=571.2[M+H]+.
第五步:将8E(0.15g,0.26mmol)溶于盐酸二氧六环溶液(10mL,4M)中,室温反应16小时。浓缩,残留物用硅胶柱色谱分离提纯(DCM∶MeOH(v/v)=40∶1-10∶1)得到标题化合物8(0.02g,14.5%)。
1H NMR(400MHz,CDCl3)δ8.48(s,1H),8.24-8.20(m,1H),7.73-7.72(m,1H),7.50-7.49(m,1H),7.49-7.44(m,1H),7.14-7.09(m,2H),5.43-5.41(m,1H),4.39-4.34(m,1H),4.28-4.24(m,1H),2.48(s,3H).LC-MS(ESI):m/z=531.2[M+H]+.
实施例9:
以6C和9A为原料,参考实施例6操作(第四,五,六反应),得到化合物9。
1H NMR(400MHz,CDCl3)δ8.41(s,1H),7.76(s,1H),7.56-7.55(m,1H),7.41-7.40(m,1H),7.09(s,1H),5.17-5.16(m,1H),4.26-4.25(m,1H),4.21-4.20(m,1H),3.97(s,2H),3.29(s,3H),2.51(s,3H).LC-MS(ESI):m/z=528.1[M+H]+.
实施例10:
以6C和10A为原料,参考实施例6操作(第四,六步反应),得到化合物10。
1H NMR(400MHz,CD3OD)δ8.65-8.64(m,2H),8.44(s,1H),8.01-7.95(m,2H),7.19(s,1H),5.34-5.29(m,1H),4.53-4.49(m,1H),4.49-4.46(m,1H),3.73-3.63(m,3H),2.38(s,3H).LC-MS(ESI):m/z=411.1[M+H]+.
实施例11:
以6C和11A为原料,参考实施例6操作(第四,六步反应),得到化合物11。
1H NMR(400MHz,CDCl3)δ8.42(s,1H),7.78-7.77(m,2H),7.61-7.60(m,2H),7.06(s,1H),4.96(s,1H),4.67(s,1H),4.38(s,1H),3.29(s,3H),2.88(s,2H),2.48(s,3H).LC-MS(ESI):m/z=435.0[M+H]+.
实施例12:
以6C和12A为原料,参考实施例6操作(第四,五,六步反应),得到化合物12。
1H NMR(400MHz,CDCl3)δ8.38(s,1H),7.52(s,1H),7.42(s,1H),7.21(s,1H),5.13-5.07(m,1H),4.83-4.76(m,2H),4.55-4.49(m,2H),4.48-4.42(m,1H),4.16-4.10(m,1H),3.26(s,2H).2.49(s,3H).LC-MS(ESI):m/z=482.1[M+H]+.
实施例13:
第一步:将6C(600mg,1.67mmol)及13A(310mg,1.67mmol)溶于吡啶(15mL)中,室温缓慢滴加50%T3P的乙酸乙酯溶液(6.68mmol),50℃反应过夜。待反应冷至室温,向反应液中加入50mL水,乙酸乙酯(30mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到标题化合物13B(447mg,51%)。LC-MS(ESI):m/z=528.1[M+H]+.
第二步:将13B(200mg,0.38mmol)、吗啉(50mg,0.57mmol)溶于1,4-二氧六环(8mL)中,随后加入Pd2(dba)3(35mg,0.038mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(44mg,0.076mmol)及碳酸钾(110mg,0.76mmol),氮气氛围,90℃下反应过夜。待反应冷至室温,向反应液中加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=2∶1)得到标题化合物13C(125mg,62%)。LC-MS(ESI):m/z=535.2[M+H]+.
第三步:将13C(125mg,0.23mmol)溶于4M氯化氢的1,4-二氧六环溶液(6mL)中,室温反应过夜。减压浓缩除去反应溶剂,向残留物加入15mL水,以乙酸乙酯(10 mL×4)萃取,合并有机层,用无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到化合物13(21mg,18%)。
1H NMR(400MHz,CD3OD)δ8.58(s,1H),7.54-7.53(m,2H),7.31(s,1H),7.18-7.17(m,2H),5.05(s,1H),4.58-4.57(m,1H),4.47-4.45(m,1H),3.92-3.87(m,4H),3.31(s,3H),3.29-3.25(m,4H),2.68(s,3H).LC-MS(ESI):m/z=495.1[M+H]+.
实施例14:
第一步:将14A(1g,6.07mmol)及环丙甲醛(1.06g,15.18mmol)溶于甲醇(20mL)中,缓慢加入乙酸(0.73g,12.14mmol),随后分批加入氰基硼氢化钠(1g,6.07mmol),室温反应过夜。向反应液中加入50mL水,乙酸乙酯(30mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到标题化合物14B(600mg,45%)。LC-MS(ESI):m/z=218.1[M+H]+.
第二步:以6C和14B为原料,参考实施例13第一步操作得到标题化合物14C(204mg,13%)。LC-MS(ESI):m/z=560.1[M+H]+.
第三步:以14C为原料,参考实施例13第三步操作得到化合物14。
1H NMR(400MHz,CD3OD)δ8.38(s,1H),7.48-7.47(m,2H),7.31(s,1H),7.12-7.11(m,2H),5.17(s,1H),4.54-4.43(m,3H),4.14-4.13(m,1H),3.71-3.66(m,1H),3.54-3.50(m,1H),2.61(s,3H),1.39-1.34(m,1H),0.56-0.51(m,4H),.LC-MS(ESI):m/z=520.0[M+H]+.
实施例15:
以6C(200mg,0.56mmol)和15A为原料,参考实施例8操作(第三,四,五步),得到化合物15。
1H NMR(400MHz,CDCl3)δ8.59(s,1H),7.46(s,1H),7.37(s,1H),7.31(s 1H),7.13(s,1H),5.98-5.97(m,1H),5.72-5.71(m,1H),4.51-4.50(m,1H),4.28-4.27(m,1H),2.35(s,3H),1.53-1.51(m,3H).LC-MS(ESI):m/z=509.1[M+H]+.
实施例16:
第一步:将16A(参考专利WO 2020156445中描述方法合成)(580g,2.66mol)溶于乙醇(3L)中,室温缓慢加入6M盐酸溶液,升温至100℃反应过夜。待反应冷至室温,将产生的固体进行过滤,滤饼用水多次洗涤,随后将固体干燥得到标题化合物16B(440g,88%)。LC-MS(ESI):m/z=187.1[M-H]-.
第二步:将16B(440g,2.34mol)及苄基三甲基氯化铵(868g,4.68mol)溶于三氯氧磷(3L)中,升温至105℃反应过夜。待反应冷却至室温,减压浓缩除去大部分溶剂,向残留物中加入3L乙酸乙酯,随后缓慢加入5L冰水,萃取分液,水相用乙酸乙酯萃取(1L×2),合并有机层,无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到标题化合物16C(495g,94%)。LC-MS(ESI):m/z=224.9[M+H]+.
第三步:将16C(2g,8.89mmol)、甲基硼酸(0.53g,8.89mmol)、二氯[1,1′-二(二苯基膦)二茂铁]钯(0.65g,0.89mmol)及碳酸钾(2.46g,17.78mmol)溶于1,4-二氧六环中(25mL)中,氮气氛围,100℃反应过夜。待反应冷至室温,向反应液中加入50mL水,乙酸乙酯(25mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到标题化合物16D(0.49g,27%)。
1H NMR(400MHz,CDCl3)δ2.95-2.87(m,2H),2.66-2.54(m,2H),2.49(s,3H).LC-MS(ESI):m/z=205.1[M+H]+.
第四步:以16D和4A为原料,参考实施例1第一步操作得到标题化合物16E(360mg,46%)。LC-MS(ESI):m/z=529.1[M+H]+.
第五步:以16E为原料,参考实施例1第二步操作得到标题化合物16(55mg,20%)。
1H NMR(400MHz,CDCl3)δ8.66(s,1H),7.86-7.82(m,1H),7.08-7.03(m,1H),4.80-4.75(m,1H),4.70(s,1H),4.39-4.34(m,1H),3.19(s,3H),2.55-2.47(m,2H),2.02(s,3H),1.30-1.23(m,2H).LC-MS(ESI):m/z=489.0[M+H]+.
实施例17:
第一步:以6C和4-溴-5-氯-2-氟苯胺为原料,参考实施例13第一步操作得到标题化合物17A。LC-MS(ESI):m/z=566.0[M+H]+.
第二步:以17A为原料,参考实施例8第四步操作得到17B。LC-MS(ESI):m/z=583.1[M+H]+.
第三步:以17B为原料,参考实施例13第二步操作得到标题化合物17C。LC-MS(ESI):m/z=590.2[M+H]+.
第四步:以17C为原料,参考实施例13第三步操作得到化合物17。
1H NMR(400MHz,CD3OD)δ8.38(s,1H),7.65-7.64(m,1H),7.03(s,1H),6.30-6.29(m,1H),5.27-5.23(m,1H),4.32-4.25(m,1H),4.21-4.17(m,1H),3.83-3.77(m,4H),3.20(s,3H),3.10-3.04(m,4H),2.39(s,3H).LC-MS(ESI):m/z=550.2[M+H]+.
实施例18:
以17B和噻吩-3-硼酸为原料,参考实施例17操作(第三,四步反应),得到化合物18。
1H NMR(400MHz,CD3OD)δ8.48(s,1H),7.96-7.93(m,1H),7.63(s,1H),7.49-7.44(m,2H),7.33-7.29(m,1H),7.21(s,1H),4.96(s,1H),4.45-4.44(m,1H),4.23-4.22(m,1H),2.62(s,3H).LC-MS(ESI):m/z=547.1[M+H]+.
实施例19:
以17B和3,3-二氟吡咯烷为原料,参考实施例17操作(第三,四步反应),得到化合物19。
1H NMR(400MHz,CD3OD)δ8.57(s,1H),7.82-7.81(m,1H),7.30(s,1H),7.01-7.00(m,1H),5.01(s,1H),4.52-4.51(m,1H),4.29-4.28(m,1H),3.93-3.84(m,2H),3.62-3.60(m,2H),2.69(s,3H),2.51-2.42(m,2H).LC-MS(ESI):m/z=570.1[M+H]+.
实施例20:
以17B和氮杂环丁烷为原料,参考实施例17操作(第三,四步反应),得到化合物20。
1H NMR(400MHz,CD3OD)δ8.56(s,1H),7.64-7.63(m,1H),7.29(s,1H),6.51-6.49(m,1H),5.03(s,1H),4.50(s,1H),4.27-4.25(m,1H),2.21-2.11(m,4H),2.68(s,3H),2.37-2.32(m,2H).LC-MS(ESI):m/z=520.2[M+H]+.
实施例21:(2S,3S,4S)-N-(5-氯-2,4-二氟苯基)-3,4-二羟基-N-(甲基-d3)-5-氧代-1-(4-三氟甲基)-6,7-二氢-5H-环戊烷-2-基吡咯烷-2-甲酰胺(化合物21)
(2S,3S,4S)-N-(5-chloro-2,4-difluorophenyl)-3,4-dihydroxy-N-(methyl-d3)-5-oxo-1-(4-(trifluoromethyl)-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)pyrrolidine-2-carboxamide
第一步:在单口瓶中,依次加入环戊酮(9.20g,109.33mmol),化合物21B(20.13g,109.33mmol),乙酸铵(8.43g,109.33mmol),升温至135℃反应18小时。冷却至室温,继续搅拌5小时,有固体析出,过滤,滤饼用石油醚洗涤三次,干燥滤饼得到粗品,粗品用乙醇重结晶得到目标化合物21C(5.7g,收率25.66%)。LC-MS(ESI):m/z=204.1[M+H]+.
第二步:在50mL单口瓶中,依次加入化合物21C(2.80g,13.78mmol),三氯氧磷(20mL),升温至100℃反应过夜。待反应冷却至室温,减压浓缩除去大部分溶剂,将残余物逐滴滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(40g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到目标化合物21D(1.1g,收率36.02%)。
1H NMR(400MHz,CDCl3)δ7.30(s,1H),3.14-3.02(m,4H),2.18-2.16(m,2H).
第三步:在单口瓶中,依次加入化合物21D(0.50g,2.26mmol),6A(0.66g,2.26mmol),Pd2(dba)3(0.13g,0.23mmol),XantPhos(0.39g,0.68mmol),碳酸钾(0.47g,3.39mmol),二氧六环(10mL),氮气置换三次,升温至95℃搅拌反应18小时。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到目标化合物21E(0.38g,收率35.29%)。LC-MS(ESI):m/z=477.1[M+H]+.
第四步:在单口瓶中,依次加入化合物21E(0.38g,0.80mmol),10%钯-碳(0.85g,0.80mmol),无水甲醇(20mL),在氢气氛围中室温反应1小时。反应结束后,垫硅藻土过滤,减压浓缩滤液,得到目标化合物21F(0.22g,71.18%)。无需纯化直接用于下一步反应。LC-MS(ESI):m/z=387.1[M+H]+.
第五步:在单口瓶中,依次加入化合物21F(0.10g,0.26mmol),21G(0.026g,0.26mmol),1-丙基磷酸酐(50%乙酸乙酯溶液)(1.07g,1.68mmol),吡啶(10mL),升温至50℃搅拌反应18小时。冷却至室温,反应液倒入水中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-26%EA/PE)纯化得到目标化合物21H(0.12g,收率86.78%)。LC-MS(ESI):m/z=532.2[M+H]+.
第六步:在50mL单口瓶中,加入化合物21H(0.12g,0.23mmol),DMF(5mL),0℃下加入氢化钠(60%)(0.014g,0.35mmol),搅拌反应10分钟,再滴加氘代碘甲烷(0.05g,0.35mmol),缓慢升温至室温搅拌反应2小时。将反应液逐滴滴加到冰的稀酸水溶液中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到目标化合物21I(0.10g,收率79.21%)。LC-MS(ESI):m/z=549.2[M+H]+.
第七步:在50mL单口瓶中,依次加入化合物21I(0.10g,0.18mmol),二氯甲烷(8mL),冷却至-20℃,缓慢滴加三氯化硼(1.33g,11.35mmol),缓慢升温至室温搅拌反应30分钟。将反应液逐滴滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯萃取三次,合并后的有机相用,饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-7%MeOH/DCM)纯化得到目标化合物21(0.04g,收率43.67%)。LC-MS(ESI):m/z=509.0[M+H]+.
1H NMR(400MHz,Methanol-d4)δ8.53-8.48(m,1H),8.07-7.88(m,7.8Hz,1H),7.55-7.46(m,1H),4.88(s,1H),4.57-4.55(m,1H),4.27-4.28(m,1H),3.27-2.87(m,4H),2.36-2.09(m,2H).
实施例22:
第一步:在单口瓶中,依次加入化合物22A(5g,36.73mmo1),硫酸二甲酯(4.63g,36.73mmol),氢氧化钠(14.69g,367.30mmol),四丁基溴化铵(1.18g,3.67mmol),甲苯(50mL),室温反应过夜。反应完全后,垫硅藻土过滤除去不溶物,减压浓缩滤液得到的粗产物用中压制备仪Biotage Isolera One(40g硅胶柱,洗脱剂:0-12%EA/PE)纯化得到目标化合物22B(2.38g,收率43.15%)。LC-MS(ESI):m/z=151.1[M+H]+.
第二步:在50mL单口瓶中,依次加入化合物22B(0.45g,2.24mmol),5E(0.50g,3.36mmol),吡啶(10mL),室温搅拌10min,然后缓慢滴加1-丙基磷酸酐(50%乙酸乙酯溶液)(4.28g,6.73mmol),滴加完毕后室温搅拌1小时,随后升温至50℃反应48小时。冷却至室温,减压浓缩除去大部分吡啶,将残余物倒入水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%MeOH/DCM)纯化得到目标化合物22C(0.27g,收率36.16%)。LC-MS(ESI):m/z=334.1[M+H]+.
第三步:以22C和2-氯-6-甲基-4-(三氟甲基)吡啶原料,参考实施例1第一步操作得到目标化合物22D(0.26g,收率65.19%)。LC-MS(ESI):m/z=493.1[M+H]+.
第四步:以22D原料,参考实施例1第二步操作得到目标化合物22。
1H NMR(400MHz,Methanol-d4)δ8.43(s,1H),8.09-8.02(m,2H),7.62-7.58(m,1H),7.31(s,1H),5.15-5.12(m,1H),4.26-4.20(m,2H),3.31(s,3H),2.66(s,3H).LC-MS(ESI):m/z=453.1[M+H]+.
实施例23:
以23A和化合物6C为原料,参考实施例8操作(第三,四,五步),得到化合物23。
1H NMR(400MHz,Methanol-d4)δ8.54(s,1H),8.27-7.70(m,2H),7.30(s,1H),4.93(s,1H),4.63-4.46(m,1H),4.27(s,1H),2.68-2.51(m,3H).LC-MS(ESI):m/z=490.1[M+H]+.
实施例24:
以24A和化合物6C为原料,参考实施例8操作(第三,四,五步),得到化合物24。
1H NMR(400MHz,Methanol-d4)δ8.61-8.49(m,1H),8.02-7.94(m,1H),7.66-7.53(m,1H),7.41-7.22(m,1H),4.93-4.86(m,1H),4.61-4.45(m,2H),4.25-4.23(m,1H),2.69-2.45(m,3H).LC-MS(ESI):m/z=533.0[M+H]+.
实施例25:
以化合物17B和化合物25A为原料,参考实施例17操作(第三,四步反应),得到化合物25。
1H NMR(400MHz,Methanol-d4)δ8.47-8.42(m,1H),7.58-7.39(m,1H),7.19-7.06(m,1H),6.48-6.31(m,1H),4.93-4.86(m,1H),4.44-4.40(m,1H),4.32-4.24(m,2H),4.23-4.14(m,2H),3.86-3.72(m,2H),3.24(s,3H),2.58-2.43(m,3H).LC-MS(ESI):m/z=550.2[M+H]+.
实施例26:
第一步:在50mL单口瓶中,依次加入化合物26A(0.5g,2.45mmol),26B(0.32g,2.45mmol),DIPEA(0.95g,7.35mmol),二氯甲烷(20mL),室温搅拌30分钟,然后缓慢滴加HATU(1.12g,2.94mmol),滴加完毕后室温搅拌1小时。减压浓缩除去大部分溶剂,将残余物倒入水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-15%EA/PE)纯化得到目标化合物26C(0.46g,收率67.50%)。LC-MS(ESI):m/z=279.0[M+H]+.
第二步:在50mL单口瓶中,依次加入化合物26C(0.46g,1.65mmol),锌粉(0.54g,8.25mmol),无水甲醇(20mL),室温搅拌5min,然后加入氯化铵(0.44g,8.23 mmol),继续搅拌10分钟。垫硅藻土过滤,将滤液浓缩,残余物倒入水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到目标化合物26D(0.33g,收率80.59%)。LC-MS(ESI):m/z=249.1[M+H]+.
第三步:以26D和化合物6C为原料,参考实施例6第四步操作得到目标化合物26E。LC-MS(ESI):m/z=591.2[M+H]+.
第四步:以26E为原料,参考实施例8第四步操作得到目标化合物26F。LC-MS(ESI):m/z=608.2[M+H]+.
第五步:以26F为原料,参考实施例1第二步操作得到目标化合物26。
1H NMR(400MHz,Methanol-d4)δ8.55-8.52(m,1H),8.25-7.93(m,1H),7.66-7.42(m,1H),7.30-7.22(m,1H),4.95(s,1H),4.68-4.21(m,6H),2.70-2.50(m,3H).LC-MS(ESI):m/z=568.2[M+H]+.
实施例27:
以4A和27A为原料,参考实施例4操作(第一,二步反应),得到化合物27。
1H NMR(400MHz,CD3OD)δ8.12-8.10(m,1H),7.91-7.77(m,1H),7.54-7.42(m,1H),6.57-6.55(m,1H),4.92(s,1H),4.51-4.47(m,1H),4.21-4.20(m,1H),3.90-3.88(m,2H),3.71(s,1H),3.25-3.23(m,3H),2.55-2.30(m,3H),0.65-0.60(m,2H),0.36-0.35(m,2H).LC-MS(ESI):m/z=482.0[M+H]+.
实施例28:
第一步:室温下将28A(11.2g,100mmol)溶解于乙醇(100mL)中,向溶液中加入(Boc)2O(43.6g,200mmol)和三乙胺(20.2g,200mmol),搅拌均匀后,65℃反应16小时,TLC监测原料消失,停止反应。将反应液浓缩,柱层析(PE∶EA=4∶1)得到产物28B(18.5g,87.27%)。LC-MS(ESI):m/z=213.1[M+H]+.
第二步:室温下,将氢化铝锂(5.69g,150mmol)加入圆底烧瓶中,加入干燥的四氢呋喃(50mL),搅拌均匀。将28B(6.36g,30mmol)溶解于干燥的THF(25mL)中,冰浴下,将28B的THF溶液滴加到氢化铝锂的悬浊液中,滴加完毕后,80℃回流16小时。TLC监测原料消失,停止反应。待反应冷至室温,加入15%的氢氧化钠溶液淬灭反应,过滤,滤液浓缩后得到的粗产物用柱层析(PE∶EA=4∶1)分离得到产物28C(2.33g,61.64%)。LC-MS(ESI):m/z=127.1[M+H]+.
第三步:以6C和28C为原料,参考实施例13第一步操作得到28D。LC-MS(ESI):m/z=469.2[M+H]+.
第四步:以28D为原料,参考实施例13第三步操作得到化合物28(30mg,21.89%)。
1H NMR(400MHz,CD3OD)δ8.71-8.30(m,3H),7.58-7.57(m,1H),7.18(s,1H),5.55-4.78(m,1H),4.46-4.45(m,1H),3.95(m,1H),2.49(s,3H),1.19(s,3H).
LC-MS(ESI):m/z=429.1[M+H]+.
实施例29:
第一步:室温下,将29A(1.2g,5.0mmol)溶解于甲醇(30mL)中,加入锌粉(3.27g,50mmol),搅拌均匀,向反应体系中分批加入氯化铵(2.67g,50mmol),室温反应30分钟,TLC监测原料消失,停止反应。过滤,将滤液浓缩后,加入EA(50mL)溶解残余物,水洗有机相(30mL×3),干燥有机相,浓缩得到产物29B(1g,96.15%)。LC-MS(ESI):m/z=207.9[M+H]+.
第二步:以6C和29B为原料,参考实施例6第四步操作得到29C。LC-MS(ESI):m/z=550.0[M+H]+.
第三步:以29C为原料,参考实施例6第五步操作产物29D。LC-MS(ESI):m/z=569.0[M+H]+.
第四步:以29D为原料,参考实施例13第二步操作得到29E。LC-MS(ESI):m/z=580.2[M+H]+.
第五步:以29E为原料,参考实施例1第二步操作得到化合物29。
1H NMR(400MHz,CD3OD)δ8.54(d,1H),7.31-7.22(m,2H),7.01-6.90(m,1H),4.98(d,1H),4.59(m,1H),4.36(m,5H),2.67-2.47(m,3H).LC-MS(ESI):m/z=540.2[M+H]+.
实施例30:
第一步:以6C和30A为原料,参考实施例6第四步操作得到30B。LC-MS(ESI):m/z=517.1[M+H]+.
第二步:以30B为原料,参考实施例6第五步操作得到产物30C。LC-MS(ESI):m/z=534.1[M+H]+.
第三步:以30C为原料,参考实施例29第一步操作得到产物30D。LC-MS(ESI):m/z=504.1[M+H]+.
第四步:室温下,将30D(30mg,0.06mmol)溶解于DCM(2mL)中,冰浴下,向溶液中加入乙酸酐(1mL,10.7mmol),升温至室温,继续反应1小时,TLC监测原料消失,停止反应。将反应液倒入DCM(10mL)中,加入饱和碳酸氢钠溶液洗涤(10mL×3),有机相用饱和食盐水(10mL)洗涤一次,干燥有机相,浓缩,柱层析(PE∶EA=2∶1)分离得到产物30E(31mg,94.72%)。LC-MS(ESI):m/z=546.1[M+H]+.第五步:以30E为原料,参考实施例13第三步操作得到化合物30(8mg,28.78%)。
1H NMR(400MHz,CD3OD)δ8.45-8.36(m,1H),8.19-7.83(m,1H),7.35-7.22(m,1H),7.11(m,1H),4.82(d,1H),4.58-4.42(m,1H),4.40-4.12(m,1H),2.58-2.39(m,3H),2.08(m,3H).LC-MS(ESI):m/z=506.2[M+H]+.
实施例31:
第一步:室温下,将30D(50mg,0.1mmol)溶解于干燥的DCM(10mL)中,向溶液中加入TEA(30mg,0.3mmol),搅拌均匀后,将混合溶液加入到三光气(15mg,0.05mmol)的DCM(5mL)溶液中,继续搅拌10分钟后,向反应体系中加入MeOH(2mL)。室温反应30分钟后,TLC监测原料消失,停止反应。将反应液倒入DCM(10mL)中,用饱和碳酸氢钠溶液(15mL)洗涤有机相两次,再用饱和食盐水(15mL)洗涤有机相一次,干燥有机相,浓缩,柱层析(PE∶EA=2∶1)分离得到产物31A(55mg,97.95%)。LC-MS(ESI):m/z=562.2[M+H]+.
第二步:以31A为原料,参考实施例1第二步操作得到化合物31。
1H NMR(400MHz,CD3OD)δ8.43(d,1H),8.20(m,1H),7.32-6.98(m,2H),4.84(d,1H),4.59-4.42(m,1H),4.39-4.13(m,1H),3.67(d,3H),2.57-2.37(m,3H).LC-MS(ESI):m/z=522.1[M+H]+.
实施例32:
第一步:以32A为原料,参考实施例26第一步操作得到产物32B。LC-MS(ESI):m/z=327.9[M+H]+.
第二步:室温下,将32B(288mg,0.88mmol),溶解于干燥的甲苯(10mL)中,依次加入Boc-NH2(120mg,1.06mmol),Pd2(dba)3(119mg,0.13mmol),BINAP(160mg,0.26mmol),Cs2CO3(860mg,2.64mmol),氮气氛围,100℃下反应16小时,TLC监测原料消失,停止反应。将反应液冷却至室温,倒入EA(15mL)中,用水洗涤有机相(15mL×3),饱和食盐水(15mL)洗涤一次,干燥有机相,浓缩,柱层析(PE∶EA=4∶1)得到产物32C(225mg,70.10%)。LC-MS(ESI):m/z=365.0[M+H]+.
第三步:室温下,将32C(225mg,0.62mmol)溶解于DCM(10mL)中,向溶液中加入三氟乙酸(1mL,13.4mmol),室温反应1h,TLC监测原料消失,停止反应。将反应液倒入DCM(10mL)中,加入饱和碳酸氢钠溶液(10mL),萃取,分液,有机相用饱和食盐水(15mL)洗涤两次,干燥有机相,浓缩,柱层析(PE∶EA=4∶1)分离得到产物32D(160mg,97.52%)。LC-MS(ESI):m/z=265.0[M+H]+.
第四步:以32D为原料,参考实施例26第三步操作得到32E。LC-MS(ESI):m/z=607.1[M+H]+.
第五步:以32E为原料,参考实施例26第四步操作得到产物32F。LC-MS(ESI):m/z=624.1[M+H]+.
第六步:以32F为原料,参考实施例26第五步操作得到化合物32(50mg,53.52%)。
1H NMR(400MHz,CD3OD)δ8.63-8.48(m,1H),8.04(t,1H),7.62(m,1H),7.46-7.19(m,1H),5.55-4.92(m,1H),4.66-4.17(m,6H),2.61(m,3H).LC-MS(ESI):m/z=584.2[M+H]+.
实施例33:
第一步:以33B和6C为原料,参考实施例13第一步操作得到目标化合物33A。LC-MS(ESI):m/z=480.0[M+H]+.
第二步:以33A为原料,参考实施例13第三步操作得到化合物33。LC-MS(ESI):m/z=440.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.46(s,1H),8.06-8.03(m,1H),7.26(s,1H),7.07-7.05(m,1H),6.91-6.86(m,1H),5.57-5.35(m,1H),4.80-4.77(t,1H),4.70-4.63(m,1H),4.51-4.41(m,2H),3.39-3.36(m,2H),2.38(s,3H).
实施例34:
第一步:将化合物17B(0.10g,0.17mmol),Pd(PPh3)2Cl2(24.0mg,0.034mmol),CuI(16.0mg,0.085mmol),5mL DMF依次加入干燥反应瓶。氮气置换三次,继续加入三乙胺(0.10g,1.02mmol),三甲基硅乙炔(0.10g,1.02mmol)加入完毕,再次氮气置换,然后升温至50℃反应过夜。将反应液冷却至室温,然后垫硅藻土过滤,用30mL乙酸乙酯洗涤硅藻土,滤液使用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,然后粗品使用中压制备仪Biotage Isolera One(24g硅胶柱,洗脱剂:0-15%EA/PE)纯化得到目标化合物34A(84.0mg,收率82.20%)。LC-MS(ESI):m/z=601.2[M+H]+.
第二步:将化合物34A(84.0mg,0.14mmol),溶于1mL 1,4-二氧六环,然后加入5mL氯化氢/1,4-二氧六环溶液,室温搅拌40小时,将反应液加入20mL乙酸乙酯稀释,加入饱和碳酸氢钠溶液调节pH至7-8,分出有机相,有机相使用饱和食盐水20mL洗涤,无水硫酸钠干燥,过滤,浓缩得到目标化合物34B(52.0mg,收率66.21%)。LC-MS(ESI):m/z=561.2[M+H]+.
第三步:将化合物34B(56mg,0.10mmol)溶于5mL THF,室温下加入TBAF四氢呋喃溶液(1M,0.2mL,0.20mmol),室温搅拌2h。浓缩除去大部分溶剂,加入20mL乙酸乙酯,饱和食盐水多次洗涤,有机相用无水硫酸钠干燥,过滤,浓缩得到棕色粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%MeOH/DCM)纯化得到化合物34(12.0mg,收率24.55%)。LC-MS(ESI):m/z=489.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.57-8.52(m,1H),8.04-7.41(m,2H),7.31-7.24(m,1H),5.52-4.88(m,1H),4.57-4.26(m,2H),4.14-4.00(m,1H),2.69-2.51(m,3H).
实施例35:
以化合物29D为原料,参考实施例34操作(第一,二,三步),得到化合物35。LC-MS(ESI):m/z=473.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.47-8.42(m,1H),7.99-7.42(m,1H),7.32-7.04(m,2H),5.44-4.77(m,1H),4.48-4.16(m,2H),3.87-3.73(m,1H),2.60-2.42(m,3H).
实施例36:
第一步:在50mL单口瓶中,依次加入化合物36A(0.43g,2.08mmol),乙烯基三氟硼酸钾(0.42g,3.14mmol),[1,1′-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.047g,0.081mmol),碳酸钠(0.17g,0.21mmol),二氧六环(8mL),水(2mL),氮气置换三次,升温至60℃搅拌反应18小时。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-8%EA/PE)纯化得到化合物36B(170mg,收率53.21%)。
1H NMR(400MHz,CDCl3)δ7.12(s,1H),7.04(s,1H),6.62-6.54(m,1H),5.97-5.92(m,1H),5.52-5.49(m,1H),2.52(s,3H).
第二步:在50mL封管中,依次加入化合物36B(0.17g,1.11mmol),碘化钠(58mg,0.22mmol),(三氟甲基)三甲基硅烷(1.58g,11.14mmol),干燥四氢呋喃(13mL),氮气氛围下升温至80℃室温搅拌反应18小时。将反应液冷却至室温,倒入水中,用乙酸乙酯萃取三次,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物36C(130mg,收率57.52%)。LC-MS(ESI):m/z=204.1[M+H]+.
第三步:以36C为原料,参考实施例1第一步操作得到化合物36D。LC-MS(ESI):m/z=531.2[M+H]+.
第四步:以36C为原料,参考实施例1第二步操作化合物36。
1H NMR(400MHz,CD3OD)δ8.04-7.78(m,2H),7.40-7.33(m,1H),6.83-6.76(m,1H),5.71-4.72(m,1H),4.16-4.08(m,2H),2.80-2.74(m,1H),2.47-2.31(m,3H),1.86-1.70(m,2H).LC-MS(ESI):m/z=491.1[M+H]+.
实施例37:
第一步:以化合物6C(0.12g,0.33mmol)和2,4-二氟-5-氯苯胺(0.065g,0.4mmol)为原料,参考实施例6第四步操作得到化合物37A(160mg,收率95.85%)。LC-MS(ESI):m/z=506.1[M+H]+.
第二步:在25mL单口瓶中,加入化合物37A(0.06g,0.12mmol),DMF(5mL),0℃下加入氢化钠(60%)(0.014g,0.36mmol),搅拌反应15分钟,滴加烯丙基溴(0.044g,0.36mmol),缓慢升温至室温搅拌反应1小时。将反应液逐滴滴加到冰的稀盐酸中,用乙酸乙酯萃取三次,合并有机相,依次用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱 剂:0-30%EA/PE)纯化得到化合物37B(70mg,收率99.99%)。LC-MS(ESI):m/z=546.1[M+H]+.
第三步:以化合物37B(0.07g,0.13mmol)为原料,参考实施例1第二步操作得到目标化合物37(24mg,收率36.5%)。
1H NMR(400MHz,CD3OD)δ8.58-8.52(m,1H),7.98-7.87(m,1H),7.52-7.47(m,1H),7.32-7.25(m,1H),5.86-5.81(m,1H),5.51-5.13(m,2H),4.87-4.81(m,1H),4.59-4.31(m,3H),4.29-4.01(m,1H),2.69-2.40(m,3H).LC-MS(ESI):m/z=506.1[M+H]+.
实施例38:
以17B和3-氟氮杂环丁烷盐酸盐为原料,参考实施例17操作(第三,四步反应),得到化合物38。
1H NMR(400MHz,CD3OD)δ8.56-8.08(m,1H),7.71-7.52(m,1H),7.29-7.23(m,1H),6.63-6.59(m,1H),5.51-5.30(m,1H),5.02-4.95(m,1H),4.53-4.44(m,3H),4.27-4.14(m,3H),2.68-2.53(m,3H).LC-MS(ESI):m/z=538.1[M+H]+.
实施例39:
以中间体17B和二甲胺四氢呋喃溶液为起始原料,参考实施例17操作(第三,四步反应),得到化合物39。
1H NMR(400MHz,CD3OD)δ8.57-8.51(m,1H),7.84-7.65(m,1H),7.34-7.23(m,1H),7.14-6.95(m,1H),5.52-4.82(m,1H),4.55-4.28(m,2H),2.88-2.79(m,6H),2.69-2.53(m,3H).LC-MS(ESI):m/z=508.1[M+H]+.
实施例40:
第一步:在50mL封管中,依次加入化合物40A(0.77g,4.0mmol),氨水(20mL),升温至120℃搅拌反应2小时。反应完全后,冷却至室温,反应液倒入水中,用乙酸乙酯萃取三次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物40B(0.53g,收率69.9%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=191.0[M+H]+.
第二步:以化合物6C(0.18g,0.50mmol)和5-氯-2-氟-4-硝基苯胺(9B)为原料,参考实施例6第四步操作得到化合物40C(230mg,收率86.33%)。
第三步:以化合物40C为原料,参考实施例6第五步操作得到化合物40D(60mg,收率39%)。LC-MS(ESI):m/z=538.1[M+H]+.
第四步:以化合物40D(0.06g,0.11mmol)为原料,参考实施例29第一步操作得到化合物40E(60mg,收率99.99%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=520.1[M+H]+.
第五步:以化合物40E(0.06g,0.11mmol)为原料参考实施例30第四步操作得到化合物40F(70mg,收率99.99%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=282.3[M/2+H]+.
第六步:以化合物40F(0.067g,0.12mmol)为原料,参考实施例1第二步操作得到化合物40(20mg,收率32%)。LC-MS(ESI):m/z=522.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.47-8.42(m,1H),8.08-7.99(m,1H),7.88-7.71(m,1H),7.37-6.86(m,2H),5.43-4.83(m,1H),4.47-4.18(m,2H),2.60-2.43(m,3H),2.14-2.10(m,6H).
实施例41:
第一步:在50mL单口瓶中,依次加入化合物41A(0.6g,3.67mmol),吗啉(0.96g,11.02mmol),碳酸钾(2.53g,18.3mmol),乙腈(10mL),升温至80℃搅拌反应48小时。冷却至室温,经硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物41B(160mg,收率20.31%)。
1H NMR(400MHz,CDCl3)δ6.51(d,J=4.0Hz,1H),6.45(d,J=4.0Hz,1H),3.83-3.80(m,4H),3.29-3.27(m,4H).
第二步:以化合物4A(0.1g,0.27mmol)和化合物41B(0.057g,0.27mmol)为原料,参考实施例1第一步操作得到化合物41C(90mg,收率62.08%)。LC-MS(ESI):m/z=537.2[M+H]+.
第三步:以化合物41C(0.09g,0.17mmol)为原料,参考实施例1第二步操作得到化合物41(15mg,收率17.76%)。
1H NMR(400MHz,CD3OD)δ8.17-7.89(m,1H),7.81-7.71(m,1H),7.49-7.27(m,1H),6.57-6.51(m,1H),5.52-4.83(m,1H),4.55-4.20(m,2H),3.83-3.73(m,4H),3.31-3.29(m,4H),2.50-2.32(m,3H).LC-MS(ESI):m/z=497.2[M+H]+.
实施例42:
以17B和3,3-二氟氮杂环丁烷为原料,参考实施例17操作(第三,四步反应),得到化合物42。
1H NMR(400MHz,CDCl3)δ8.53(s,1H),7.64-7.62(m,1H),7.14(s,1H),6.43-6.41(m,1H),5.10(s,1H),4.73-4.71(m,1H),4.52-4.44(m,5H),2.63(s,3H).LC-MS(ESI):m/z=556.1[M+H]+.
实施例43:
以17B和43A为原料,参考实施例17操作(第三,四步反应),得到化合物43。
1H NMR(400MHz,Methanol-d4)δ8.59-8.53(m,1H),7.76-7.58(m,1H),7.31-7.24(m,1H),6.68-6.44(m,1H),5.07-5.00(m,1H),4.57-4.50(m,1H),4.31-4.28(m,1H),3.96-3.91(m,1H),3.14-3.08(m,2H),2.76-2.54(m,5H).LC-MS(ESI):m/z=570.1[M+H]+.
实施例44:
第一步:在50mL单口瓶中,依次加入化合物44A(0.5g,2.06mmol),乙烯基三氟硼酸钾(0.41g,3.09mmol),[1,1′-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.23g,0.31mmol),碳酸钾(0.41g,3.09mmol),二氧六环(10mL),水(1mL),氮气置换三次,升温至100℃搅拌反应18小时。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得到粗 品。粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物44B(230mg,收率60.31%)。
1H NMR(400MHz,CDCl3)δ8.26-8.25(m,1H),7.03-7.02(m,1H),6.82-6.75(m,1H),5.94-5.90(m,1H),5.57-5.54(m,1H).
第二步:在50mL封管中,加入化合物44B(0.23g,1.24mmol),THF(15mL),(三氟甲基)三甲基硅烷(1.76g,12.4mmol),氮气保护,升温至80℃搅拌反应18小时。待反应冷至室温,将反应液减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物44C(150mg,收率51.44%)。
第三步:以化合物44C(0.15g,0.64mmol)为原料,参考实施例29第一步操作得到化合物44D(130mg,收率99%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=206.1[M+H]+.
第四步:以6C(0.08g,0.22mmol)和化合物44D(0.10g,0.48mmol)为原料,参考实施例6第四步操作得到化合物44E(70mg,收率58.12%)。
第五步:以化合物44E(0.07g,0.13mmol)为原料,参考实施例6第五步操作得到化合物44F(80mg,收率99%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=564.1[M+H]+.
第六步:以化合物44F(0.08g,0.13mmol)为原料参考实施例1第二步操作得到目标化合物44(22mg,收率30%)。
1H NMR(400MHz,CD3OD)δ8.55-8.52(m,1H),7.79-7.61(m,1H),7.36-7.09(m,2H),5.54-4.82(m,1H),4.58-4.24(m,2H),2.99-2.89(m,1H),2.68-2.51(m,3H),2.06-1.94(m,1H),1.85-1.79(m,1H).LC-MS(ESI):m/z=525.1[M+H]+.
实施例45:
以17B为原料,参考实施例17操作(第三,四步反应),得到化合物45。
1H NMR(400MHz,CD3OD)δ8.59-8.57(m,1H),8.08-7.90(m,1H),7.76-7.56(m,1H),7.33-7.27(m,1H),5.56-4.82(m,1H),4.61-4.31(m,2H),2.72-2.54(m,6H).LC-MS(ESI):m/z=507.1[M+H]+.
实施例46:
以17B为原料,参考实施例17操作(第三,四步反应),得到化合物46。
1H NMR(400MHz,CD3OD)8.56-8.51(m,1H),7.65-7.46(m,1H),7.29-7.23(m,1H),6.51-6.47(m,1H),5.51-4.96(m,1H),4.51-4.50(m,1H),4.31-4.26(m,3H),3.72-3.68(m,2H),2.79-2.53(m,4H),1.37-1.25(m,4H).LC-MS(ESI):m/z=534.1[M+H]+.
实施例47:
第一步:在50mL单口瓶中,依次加入化合物47A(5.76g,58.65mmol),三氟乙酰乙酸乙酯(10.8g,58.65mmol),乙酸铵(4.52g,58.65mmol),升温至135℃搅拌反应18小时。缓慢搅拌冷却至室温,析出固体,过滤,用乙醚洗涤固体,固体再用乙醇重结晶纯化得到化合物47B(5.6g,收率43.56%)。
1H NMR(400MHz,DMSO-d6)δ11.90(s,1H),6.51(s,1H),3.31-2.45(m,4H),1.73-1.65(m,4H).
第二步:在单口瓶中,依次加入化合物47B(2.5g,11.4mmol),三氯氧磷(10mL),然后升温至100℃搅拌反应18小时。冷却至室温,减压浓缩掉大量反应液,得到粗品倒入水中,用饱和碳酸氢钠调节pH至中性,用乙酸乙酯萃取三次,合并有机相,依次用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用硅胶柱层析分离纯化(洗脱剂:EA/PE=1/10)得到化合物47C(2.7g,收率100%)。LC-MS(ESI):m/z=236.1[M+H]+.
第三步:以化合物1A(0.12g,0.33mmol)和化合物47C(0.16g,0.68mmol)为原料,参考实施例1第一步操作得到化合物47D(0.075g,收率40.37%)。LC-MS(ESI):m/z=563.1[M+H]+.
第四步:以化合物47D(0.075g,0.13mmol)为原料,参考实施例1第二步操作得到化合物47(30mg,收率44.14%)。
1H NMR(400MHz,CD3OD)δ8.42-8.39(m,1H),8.09-7.88(m,1H),7.53-7.46(m,1H),5.19-5.01(m,1H),4.45-4.20(m,1H),3.08-2.89(m,4H),2.01-1.83(m,3H).LC-MS(ESI):m/z=523.20[M+H]+.
实施例48:
第一步:在50mL单口瓶中,将化合物48A(2.5g,17mmol)溶于乙醇(15mL)中,加入硝酸铁(7.03g,17mmol),升温至50℃搅拌反应3小时。冷却至室温,将反应液倒入水中,用乙酸乙酯萃取,有机相依次用水,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品溶于甲苯(15mL)中,加热到50℃搅拌10分钟,缓慢滴加正己烷直至浑浊,然后缓慢冷却至室温搅拌析出大量固体,过滤,减压干燥得到化合物48B(1.6g,收率48.9%)。LC-MS(ESI):m/z=192.1[M+H]+.
第二步:在单口瓶中,依次加入化合物48B(0.3g,1.57mmol),氢氧化钠(63mg,1.57mmol),水(8mL),DMF(12mL),然后升温至80℃,加入二氟氯乙酸钠(0.29g,1.9mmol),搅拌反应5小时。冷却至室温,反应液倒入水中,用乙酸乙酯萃取三次,合并有机相,依次用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用快速柱层析分离纯化(洗脱剂:EA/PE=1/10)得到化合物48C(0.16g,收率42.3%)。LC-MS(ESI):m/z=242.1[M+H]+.
第三步:以48C(160mg,0.66mmol)为原料,参考实施例29第一步操作得到化合物48D(140mg,99%),无需进一步纯化可直接用于下一步反应。LC-MS(ESI):m/z=212.1[M+H]+.
第四步:以6C(0.1g,0.27mmol)和化合物48D(0.11g,0.52mmol)为原料,参考实施例6第四步操作得到化合物48E(140mg,收率91%)。LC-MS(ESI):m/z=554.1[M+H]+.
第五步:以化合物48E(0.11g,0.2mmol)为原料,参考实施例6第五步操作得到化合物48F(100mg,收率87.6%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=571.1[M+H]+.
第六步:以化合物48F(0.08g,0.18mmol)为原料,参考实施例1第二步操作得到目标化合物48(40mg,收率41.86%)。
1H NMR(400MHz,CD3OD)δ8.47-8.42(m,1H),7.99-7.83(m,1H),7.49-7.36(m,1H),7.21-6.75(m,2H),5.44-4.70(m,1H),4.48-4.18(m,2H),2.60-2.43(m,3H).LC-MS(ESI):m/z=531.1[M+H]+.
实施例49:
以17B为原料,参考实施例17操作(第三,四步反应),得到化合物49。
1H NMR(400MHz,CD3OD)δ8.56(s,1H),7.65-7.63(m,1H),7.29(s,1H),6.52-6.50(m,1H),5.03(s,1H),4.50(s,1H),4.29-4.25(m,1H),3.86-3.69(m,2H),3.50-3.42(m,1H),2.68(s,3H),1.92-1.78(m,2H),1.25-1.21(m,3H).LC-MS(ESI):m/z=534.1[M+H]+.
实施例50:
第一步:将50A(1g,4.65mmol)及3,3-二氟氮杂环丁烷(0.65g,6.98mmol)溶于N-甲基吡咯烷酮(20mL)中,随后加入碳酸钾(1.29g,9.30mmol),于60℃反应5h。冷却至室温,向反应液中加入40mL水,以乙酸乙酯(20mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=40∶1)得到目标化合物50B(0.86g,68%)。
1H NMR(400MHz,CDCl3)δ6.83(s,1H),6.33(s,1H),4.36-4.34(m,4H).
第二步:以1A(0.25g,0.61mmol)及50B(0.17g,0.61mmol)为原料,参考实施例1第一步操作得到目标化合物50C(125mg,34%)。LC-MS(ESI):m/z=600.2[M+H]+.
第三步:以50C(125mg,0.21mmol)为原料,参考实施例1第二步操作得到标题化合物50(61mg,52%)。
1H NMR(400MHz,CD3OD)δ7.90-7.84(m,1H),7.78(s,1H),7.68-7.54(m,2H),5.68-5.61(m,1H),4.84-4.75(m,1H),4.51-4.34(m,5H).LC-MS(ESI):m/z=560.1[M+H]+.
实施例51:
第一步:将51A(8g,41.48mmol)溶于DMF(50mL)中,随后加入碳酸钾(17.28g,125.04mmol),室温搅拌10min后,缓慢加入碘甲烷(11.83g,83.36mmol),室温反应过夜。16h后,向反应液中加入100mL水,以乙酸乙酯(50mL×3)萃取,合并有机层,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=20∶1)得到目标化合物51B(7.5g,87%)。LC-MS(ESI):m/z=206.1[M+H]+.
第二步:将51B(7.5g,36.37mmol)、甲基硼酸(2.39g,39.93mmol)、二氯[1,1′-二(二苯基膦)二茂铁]钯(2.66g,3.64mmol)及碳酸铯(23.70g,72.74mmol)溶于1,4-二氧六环中(200mL)中,氮气氛围,于100℃反应过夜。16h后,冷却至室温,向反应液中加入300mL水,以乙酸乙酯(100mL×3)萃取,合并有机层,用饱和食盐水(150 mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到目标化合物51C(5.2g,77%)。LC-MS(ESI):m/z=186.1[M+H]+.
第三步:将51C(5.2g,28.02mmol)及丙烯酸甲酯(24.12g,280mmol)溶于DMF(80mL)中,随后加入Pd2(dba)3(2.57g,2.80mmol)、N,N-二环己基甲胺(10.95g,56.04mmol)及三叔丁基膦(2.27g,11.22mmol),氮气氛围,于100℃反应过夜。16h后冷却至室温,向反应液中加入150mL水,以乙酸乙酯(50mL×3)萃取,合并有机层,用饱和食盐水(60mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=3∶1)得到目标化合物51D(3.1g,47%)。LC-MS(ESI):m/z=236.1[M+H]+.
第四步:将51D(3.1g,13.18mmol)溶于无水乙醇(30mL)中,加入10%钯碳(300mg),氢气氛围下,于60℃反应过夜。16h后冷却至室温,过滤除去不溶物,以乙酸乙酯洗滤饼,合并滤液,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物51E(2.66g,85%)。LC-MS(ESI):m/z=238.1[M+H]+.
第五步:将51E(2.66g,11.21mmol)溶于四氢呋喃(50mL)及甲醇(2mL)的混合溶剂中,氮气氛围下,分批加入60%氢化钠(1.8g,45mmol),随后升温至60℃反应4h。冷却至室温,将反应液浓缩后得到的残留物51F粗品(3.5g),未经进一步纯化,直接用于下一步反应。
第六步:将上一步得到的51F粗品置于圆底烧瓶中,冰浴下缓慢加入浓盐酸(30mL),随后升温至115℃反应1h。冷却至室温,冰浴下缓慢加入饱和碳酸氢钠水溶液(200mL),以乙酸乙酯(50mL×3)萃取,合并有机层,用饱和食盐水(60mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=3∶1)得到目标化合物51G(0.91g,两步收率55%)。LC-MS(ESI):m/z=148.1[M+H]+.
第七步:将51G(0.91g,6.84mmol)溶于二氯甲烷(40mL)中,冰浴下滴加二乙胺基三氟化硫(3.95g,24.50mmol),氮气氛围下,于45℃反应过夜。16h后冷却至室温,缓慢加入饱和碳酸氢钠水溶液(50mL),以二氯甲烷(30mL×3)萃取,合并有机层,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到目标化合物51H(645mg,61%)。LC-MS(ESI):m/z=170.1[M+H]+.
第八步:将51H(0.3g,1.77mmol)溶于二氯甲烷(10mL)中,冰浴下加入间氯过氧苯甲酸(0.61g,3.54mmol),室温反应过夜。16h后,缓慢加入饱和碳酸氢钠水溶液(20mL),以二氯甲烷(15mL×3)萃取,合并有机层,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到目标化合物51I(260mg,79%)。
第九步:氮气氛围下,向51I(0.26g,1.41mmol)缓慢加入三氯氧磷(5mL),于100℃反应3h。冷却至室温,浓缩除去部分溶剂,向残留物缓慢加入饱和碳酸氢钠水溶液(15mL),以乙酸乙酯(10mL×3)萃取,合并有机层,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到目标化合物51J(128mg,45%)。LC-MS(ESI):m/z=204.1[M+H]+.
第十步:以1A(0.23g,0.63mmol)及51J(128mg,0.63mmol)为原料,参考实施例1第一步操作得到目标化合物51K(115mg,34%)。LC-MS(ESI):m/z=531.1[M+H]+.
第十一步:以51K(115mg,0.22mmol)为原料,参考实施例1第一步操作得到标题化合物51(40mg,52%)。
1H NMR(400MHz,CD3OD)δ7.88-7.84(m,1H),7.76(s,1H),7.68-7.62(m,1H),5.69-5.63(m,1H),4.81-4.73(m,1H),4.41-4.27(m,1H),3.03-2.81(m,4H),2.67(s,3H).LC-MS(ESI):m/z=491.1[M+H]+.
实施例52:
第一步:将52A(1g,4.01mmol)溶于4M氯化氢的1,4-二氧六环溶液(20mL)中,室温反应过夜。16h后,将产生的固体过滤,以少量石油醚洗滤饼,将固体干燥得到目标化合物52B(710mg,95%)。LC-MS(ESI):m/z=150.1[M+H]+.
第二步:以6C(200mg,0.56mmol)及52B(105mg,0.56mmol)为原料,参考实施例6第四步操作得到目标化合物52C(170mg,62%)。LC-MS(ESI):m/z=492.1[M+H]+.
第三步:以52C(170mg,0.35mmol)为原料,参考实施例6第五步操作得到目标化合物52D(110mg,62%)。LC-MS(ESI):m/z=509.1[M+H]+.
第四步:以52D(193mg,0.37mmol)为原料,参考实施例6第六步操作得到化合物52(42mg,41%)。
1H NMR(400MHz,CD3OD)δ8.50(s,1H),7.90-7.84(m,1H),7.56(s,1H),7.45-7.39(m,2H),7.28(s,1H),5.25(s,1H),4.56-4.53(m,2H),2.69(s,3H).LC-MS(ESI):m/z=469.1[M+H]+.
实施例53:
第一步:将53A(3g,15.07mmol)溶于4M氯化氢的1,4-二氧六环溶液(25mL)中,室温反应过夜。16h后,将反应液浓缩,残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=50∶1)得到目标化合物53B(1.42g,95%)。
1H NMR(400MHz,CD3Cl)δ10.50(s,2H),7.70-7.65(m,1H),7.59-7.55(m,1H),7.15-7.11(m,1H).
第二步:将53B(200mg,2.02mmol)加入三氟乙酰乙酸乙酯(6mL)中,于130℃反应4h,随后将反应温度调节至100℃反应过夜。16h后冷却至室温,向反应液中加入30mL水,以乙酸乙酯(15mL×3)萃取,合并有机层,无水硫酸钠干燥,浓缩 后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到目标化合物53C(246mg,56%)。LC-MS(ESI):m/z=220.1[M+H]+.
第三步:氮气氛围下,向53C(246mg,1.12mmol)缓慢加入三氯氧磷(5mL),于100℃反应3h。冷却至室温,浓缩除去部分溶剂,向残留物缓慢加入饱和碳酸氢钠水溶液(15mL),以乙酸乙酯(10mL×3)萃取,合并有机层,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=40∶1)得到目标化合物53D(203mg,76%)。LC-MS(ESI):m/z=238.0[M+H]+.
第四步:将6A(250mg,0.85mmol)及53D(203mg,0.85mmol)溶于1,4-二氧六环(15mL)中,随后加入Pd2(dba)3(78mg,0.085mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(104mg,0.17mmol)及碳酸钾(235mg,1.70mmol),氮气换气保护,于95℃反应过夜。16h后冷却至室温,向反应液中加入30mL水,以乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物53E(170mg,40%)。LC-MS(ESI):m/z=493.1[M+H]+.
第五步:将53E(170mg,0.35mmol)溶于四氢呋喃(3mL)及甲醇(3mL)的混合溶剂中,加入10%钯碳(30mg),氢气氛围下,于60℃反应过夜。16h后冷却至室温,过滤除去不溶物,以乙酸乙酯洗滤饼,合并滤液,浓缩后得到目标化合物53F(132mg,95%),未经进一步纯化,直接进行下一步反应。LC-MS(ESI):m/z=403.1[M+H]+.
第六步:以53F(132mg,0.33mmol)和5-氯-2,4-二氟苯胺(54mg,0.33mmol)为原料,参考实施例6第四步操作得到目标化合物53G(144mg,80%)。LC-MS(ESI):m/z=548.2[M+H]+.
第七步:以53G(144mg,0.26mmol)为原料,参考实施例6第五步操作目标化合物53H(115mg,77%)。LC-MS(ESI):m/z=565.2[M+H]+.
第八步:以53H(115mg,0.20mmol)为原料,参考实施例6第六步操作得到化合物53(35mg,23%)。
1H NMR(400MHz,CDCl3)δ8.75(s,1H),7.90-7.84(m,1H),7.88-7.85(m,1H),7.53-7.50(m,1H),7.17-7.12(m,1H),5.08(s,1H),4.73-4.70(m,1H),4.38-4.35(m,1H).LC-MS(ESI):m/z=525.1[M+H]+.
实施例54:(2S,3S,4S)-N-(5-氯-4-(2,2-二氟环丙基)-2-氟苯基)-3,4-二羟基-N-(甲基-d3)-1-(6-甲基-4-(三氟甲基)吡啶-2-基)-5-氧代吡咯烷-2-甲酰胺(化合物54)
(2S,3S,4S)-N-(5-chloro-4-(2,2-difluorocyclopropyl)-2-fluorophenyl)-3,4-dihydroxy-N-(methyl-d3)-1-(6-methyl-4-(trifluoromethyl)pyridin-2-yl)-5-oxopyrrolidine-2-carboxamide
第一步:室温下将54A(1.3g,5.79mmol)溶解于甲苯(20mL)中,向溶液中加入2,5-己二酮(3.31g,28.98mmol),搅拌均匀后加入对甲苯磺酸(0.3g,1.74mmol),100℃反应3小时。TLC监测原料消失,停止反应。冷却至室温,向反应体系加入EA(20mL),用饱和食盐水(15mL)洗涤有机相3次,干燥有机相,过滤,浓缩,残余物经柱层析(PE∶EA=50∶1)分离提纯得到产物54B(420mg,23.97%)。
第二步:室温下将54B(420mg,1.39mmol)溶解于二氧六环(10mL)中,依次加入乙烯基三氟硼酸钾(220mg,1.64mmol),Pd(dppf)Cl2(150mg,0.21mmol),Na2CO3(0.44g,4.15mmol),水(2mL),氮气气氛下,100℃反应16小时,TLC监测原料消失,停止反应。冷却至室温,向反应体系中加入EA(10mL),用水洗涤有机相(10mL×3),再用饱和食盐水(10mL)洗涤有机相一次,干燥有机相,过滤,浓缩,残余物经柱层析(PE∶EA=50∶1)分离提纯得到产物54C(250mg,72.03%)。LC-MS(ESI):m/z=250.1[M+H]+.
第三步:室温下将54C(250mg,1.0mmol)溶解于干燥的THF(20mL)中,向溶液中加入二氟溴甲基三甲基硅烷(2.03g,10.0mmol),搅拌均匀,加入碘化钠(75mg,0.5mmol),65℃反应24h,TLC监测原料消失,停止反应。冷却至室温,向反应体系中加入EA(10mL),有机相用饱和食盐水(10mL)洗涤两次,干燥有机相,过滤,浓缩,残余物经柱层析(PE∶EA=50∶1)分离提纯得到产物54D(220mg,73.40%)。LC-MS(ESI):m/z=300.1[M+H]+.
第四步:室温下,将54D(220mg,0.73mmol)溶解于乙醇当中,向其中加入盐酸羟胺(540mg,7.77mmol),再向其中加入水(2mL),80℃反应20小时,TLC监测原料消失,停止反应。冷却至室温,浓缩反应液,用EA(10mL)萃取反应液三次,有机相用饱和食盐水(20mL)洗涤两次,干燥有机相,过滤,浓缩,残余物经柱层析(PE∶EA=4∶1)分离纯化得到产物54E(80mg,49.18)。LC-MS(ESI):m/z=222.1[M+H]+.
第五步:室温下,将6C(100mg,0.28mmol)溶解于吡啶(5mL)中,加入54E(74mg,0.34mmol),搅拌均匀,向溶液中滴加T3P(1.42g,2.24mmol),50℃反应16小时,TLC监测原料消失,停止反应。向反应液中加入EA(15mL),用饱和食盐水洗涤有机相(10mL×3),干燥有机相,过滤,浓缩,残余物经柱层析(PE∶EA=4∶1)分离纯化 得到54F异构体1(Rf=0.5(PE∶EA=4∶1),35mg,22.36%)和54F异构体2(Rf=0.3(PE∶EA=4∶1),55mg,35.14%)。LC-MS(ESI):m/z=564.1[M+H]+.
第六步:室温下,将54F异构体1(35mg,0.06mmol)溶解于干燥的DMF(5mL)中,冰水浴下,加入NaH(4.7mg,0.2mmol),搅拌10min后,向反应体系中加入CD3I(28mg,0.2mmol),升温至室温,继续反应30min,TLC监测原料消失,停止反应。将反应液滴入1M的盐酸(10mL)淬灭反应,加入EA(15mL)萃取,分液,水相用EA(10mL×2)萃取,合并有机相,有机相用饱和氯化钠溶液(20mL)洗涤两次,干燥有机相,过滤,浓缩,残余物经柱层析(PE∶EA=4∶1)分离纯化得到产物54G异构体1(35mg,97.07%)。LC-MS(ESI):m/z=581.1[M+H]+.
以54F异构体2为原料参考上述合成方法得到54G异构体2。LC-MS(ESI):m/z=581.1[M+H]+.
第七步:室温下将54G异构体1(35mg,0.06mmol)溶解于干燥的DCM(8mL)中,降温至-20℃,向反应体系中滴加BCl3的二氯甲烷溶液(1.5mL,1mol/L),滴加完毕后,升温至室温,继续反应1小时,TLC监测原料消失,停止反应。将反应液滴加入冰的饱和碳酸氢钠溶液(10mL)中,二氯甲烷(15mL)萃取,分液,水相用DCM(10mL×2)萃取,合并后的有机相,用饱和食盐水(20mL)洗涤一次,干燥有机相,过滤,浓缩,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物54,异构体1(25mg,76.72%)。
1H NMR(400MHz,CD3OD)δ8.62-8.46(m,1H),8.09-7.78(m,1H),7.46(m,1H),7.25(m,1H),4.94(d,1H),4.60-4.50(m,1H),4.28(d,1H),3.07(m,1H),2.73-2.45(m,3H),2.09-1.86(m,2H).LC-MS(ESI):m/z=541.2[M+H]+.
以54G异构体2为原料参考上述合成方法得到化合物54,异构体2。
1H NMR(400MHz,CD3OD)δ8.58-8.49(m,1H),8.04-7.78(m,1H),7.42(d,1H),7.24(m,1H),4.93(d,1H),4.60-4.48(m,1H),4.28(d,1H),3.10-2.98(m,1H),2.63(m,3H),2.04
(m,1H),1.97-1.81(m,1H).LC-MS(ESI):m/z=541.2[M+H]+.
实施例55:
以化合物21F为原料,参考实施例17操作(第一到第四步),得到化合物55。
1H NMR(400MHz,Methanol-d4)δ8.59-8.53(m,1H),7.76-7.58(m,1H),7.31-7.24(m,1H),6.68-6.44(m,1H),5.07-5.00(m,1H),4.57-4.50(m,1H),4.31-4.28(m,1H),3.96-3.91(m,1H),3.14-3.08(m,2H),2.76-2.54(m,5H).LC-MS(ESI):m/z=564.1[M+H]+.
实施例56:
以化合物55A为原料,参考实施例34操作(第一,二,三步),得到化合物56。
1H NMR(400MHz,Methanol-d4)δ8.64-8.45(m,1H),8.09-7.77(m,1H),7.68-7.50(m,1H),4.91(s,1H),4.65-4.52(m,1H),4.36-4.22(m,1H),4.18-3.98(m,1H),3.26-2.75(m,4H),2.35-2.11(m,2H).LC-MS(ESI):m/z=515.2[M+H]+.
实施例57:
第一步:以化合物17B(0.15g,0.26mmol)为原料,参考实施例1第二步操作得到目标化合物57A(0.10g,收率56.59%)。LC-MS(ESI):m/z=543.4[M+H]+.
第二步:在单口瓶中,依次加入化合物57A(0.10g,0.18mmol),3-氧杂环丁胺(0.011g,0.15mmol),Pd2(dba)3(0.017g,0.018mmol),BINAP(0.028g,0.045mmol),碳酸铯(0.15g,0.45mmol),甲苯(10mL),氮气置换三次,升温至100℃搅拌反应过夜。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物57(0.05g,收率62.20%)。
1H NMR(400MHz,Methanol-d4)δ8.63-8.48(m,1H),7.79-7.56(m,1H),7.34-7.23(m,1H),6.57-6.25(m,1H),5.10-4.95(m,3H),4.78-4.60(m,3H),4.57-4.25(m,2H),2.74-2.50(m,3H).LC-MS(ESI):m/z=536.3[M+H]+.
实施例58:
以50A和1-甲基-1H-吡唑-4-硼酸为原料,参考实施例50操作(第一、二、三步)得到化合物58。
1H NMR(400MHz,CD3OD-d4)δ9.16(d,1H),9.04-8.70(m,3H),8.55-8.22(m,2H),6.56(d,1H),6.43(d,1H),6.16-5.60(m,1H),5.30-5.08(m,1H),4.74-4.57(m,4H).LC-MS(ESI):m/z=549.0[M+H]+.
实施例59:
以59A为原料,参考实施例53操作(第一步到第八步),得到化合物59。
1H NMR(400MHz,CDCl3)δ8.75(s,1H),7.88-7.82(m,1H),7.78-7.74(m,1H),7.49-7.45(m,1H),7.18-7.14(m,1H),5.10(s,1H),4.74-4.48(m,1H),4.38-4.34(m,1H).LC-MS(ESI):m/z=525.1[M+H]+.
实施例60:
以17B和三丁基丙炔锡烷为原料,参考实施例34实验操作(第一、二步),得到化合物60。
1H NMR(400MHz,CDCl3)δ8.55(s,1H),7.81-7.78(m,1H),7.38-7.35(m,1H),7.14(s,1H),5.05(s,1H),4.70-4.67(m,1H),4.41-4.38(m,1H),2.64(s,3H),2.15(s,3H).LC-MS(ESI):m/z=503.4[M+H]+.
实施例61:
第一步:在50mL单口瓶中,依次加入化合物29B(0.23g,1.11mmol),1,3-噻唑-2-硼酸频哪醇酯(0.28g,1.33mmol),Pd(dppf)Cl2(0.081g,0.11mmol),氟化铯(0.51g,3.36mmol),DMF(10mL),水(1mL),氮气置换三次,升温至100℃搅拌反应18小时。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并有机相, 用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物61A(70mg,收率29.72%)。LC-MS(ESI):m/z=213.1[M+H]+.
第二步:以化合物6C(80mg,0.22mmol)和化合物61A(70mg,0.33mmol)为原料,参考实施例6第四步操作得到化合物61B(80mg,收率65.58%)。LC-MS(ESI):m/z=555.3[M+H]+.
第三步:以化合物61B(83mg,0.15mmol)为原料,参考实施例6第五步操作得到目标化合物61C(80mg,收率93.32%),未经进一步纯化可直接用于下一步反应。LC-MS(ESI):m/z=572.4[M+H]+.
第四步:以化合物61C(80mg,0.14mmol)为原料,参考实施例1第二步操作得到目标化合物61(6mg,收率8.06%)。
LC-MS(ESI):m/z=532.4[M+H]+.
1H NMR(400MHz,CD3OD)δ9.12-9.08(m,1H),8.55-8.53(m,1H),8.38-8.28(m,1H),8.20-8.16(m,1H),7.53-7.48(m,1H),7.30-7.24(m,1H),5.57-4.94(m,1H),4.70-4.29(m,2H),2.62-2.54(m,3H)。
实施例62:
以6C(200mg,0.56mmol)和62A为原料,参考实施例8操作(第三,四,五步),得到化合物62。
1H NMR(400MHz,CD3OD)δ8.47-7.99(m,1H),7.19-7.17(m,1H),6.93-6.84(m,2H),5.54-5.43(m,1H),5.33-5.23(m,1H),4.48-4.26(m,2H),3.26-2.68(m,4H),2.46-2.38(m,3H).LC-MS(ESI):m/z=459.2[M+H]+.
实施例63:
第一步:在50mL封管中,依次加入化合物63A(4.3g,50mmol),三氟乙酰乙酸乙酯(9.21g,50mmol),乙酸铵(3.85g,50mmol),升温至100℃搅拌反应18小时。缓慢搅 拌冷却至室温,析出固体,过滤,用乙醚洗涤固体,固体再用乙醇重结晶纯化得到化合物63B(0.9g,收率8.77%)。
1H NMR(400MHz,DMSO-d6)δ11.88(s,1H),6.70(s,1H),5.00-4.99(m,2H),4.87-4.86(m,2H).LC-MS(ESI):m/z=205.9[M+H]+.
第二步:在单口瓶中,依次加入化合物63B(0.9g,4.39mmol),三氯氧磷(25mL),然后升温至100℃搅拌反应18小时。冷却至室温,减压浓缩掉大量反应液,得到粗品倒入水中,饱和碳酸氢钠水溶液调节pH至中性,乙酸乙酯萃取三次,合并后的有机相,依次用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用柱层析分离纯化(洗脱剂:EA/PE=1/10)得到化合物63C(0.78g,收率78.78%)。
第三步:在单口瓶中,依次加入化合物6A(0.21g,0.72mmol),化合物63C(0.18g,0.81mmol),Pd2(dba)3(0.074g,0.081mmol),XantPhos(0.14g,0.24mmol),碳酸钾(0.17g,1.22mmol),二氧六环(10mL),氮气置换三次,然后升温至95℃搅拌反应18小时。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并有机相,依次用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到化合物63D(0.34g,收率87.74%)。LC-MS(ESI):m/z=479.2[M+H]+.
第四步:在单口瓶中,将63D(0.34g,0.71mmol)溶于甲醇(20mL)中,加入10%钯碳(100mg),氢气氛围,室温下搅拌反应2小时。硅藻土过滤除去不溶物,以乙酸乙酯洗滤饼,合并滤液,浓缩后得到目标化合物63E(0.25g,90.68%),无需进一步纯化直接进行下一步反应。LC-MS(ESI):m/z=389.1[M+H]+.
第五步:以63E(0.25g,0.64mmol)和5-氯-2,4-二氟苯胺(0.16g,0.98mmol)为原料,参考实施例6第四步操作得到化合物63F(0.21g,收率61.47%)。LC-MS(ESI):m/z=534.1[M+H]+.
第六步:以化合物63F(0.07g,0.13mmol)为原料,参考实施例6第五步操作得到目标化合物63G(70mg,收率97.75%),未经进一步纯化可直接用于下一步反应。LC-MS(ESI):m/z=551.1[M+H]+.
第七步:以化合物63G(70mg,0.13mmol)为原料,参考实施例1第二步操作得到目标化合物63(36mg,收率54.21%)。
1H NMR(400MHz,CD3OD)δ8.58-8.53(m,1H),7.83-7.78(m,1H),7.48-7.18(m,1H),5.42-4.88(m,4H),4.75-4.67(m,1H),4.51-4.20(m,2H).LC-MS(ESI):m/z=511.1[M+H]+.实施例64:
第一步:将64A(2g,3.93mmol)溶于DMF(40mL)中,依次加入Pd(dppf)Cl2(0.29g,0.39mmol),dppf(0.33g,0.60mmol),五水合硫代硫酸钠(4.88g,19.66mmol),碳酸铯(3.84g,11.79mmol)。氮气氛围,反应升温至140℃反应6小时,继续在100℃搅拌16小时。待反应冷至室温,加水(100mL),乙酸乙酯(50mL×2)萃取,合并后的有机相用饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(24g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到产物64B(0.49g,收率24.51%)。LC-MS(ESI):m/z=168.1[M+H]+.
第二步:以64B(0.23g,1.38mmol)和6C(0.25g,0.69mmol)为原料,参考实施例6第四步操作得到产物64C(0.18g,收率51.20%)。LC-MS(ESI):m/z=510.1[M+H]+.
第三步:以64C(0.18g,0.35mmol)为原料,参考实施例6第五步操作得到产物64D(0.1g,收率59.69%)。LC-MS(ESI):m/z=527.1[M+H]+.
第四步:以化合物64D(0.11g,0.21mmol)为原料,参考实施例1第二步操作得到目标化合物64(21.0mg,收率20.56%)。
1H NMR(400MHz,CD3OD-d4):δ8.56-8.50(m,1H),7.89-7.85(m,1H),7.73-7.67(m,1H),7.56(s,1H),7.46-7.08(m,2H),5.60-5.22(m,1H),4.59-4.53(m,2H),2.69-2.44(m,3H).LC-MS(ESI):m/z=487.0[M+H]+.
实施例65:
以化合物65A为原料,参考实施例64操作(第一到第四步),得到化合物65。
1H NMR(400MHz,CDCl3)δ8.48(s,1H),7.76-7.75(m,1H),7.49-7.48(m,1H),7.35(s,1H),7.21-7.16(m,1H),7.13(s,1H),5.36(s,1H),4.72-4.71(m,1H),4.56-4.55(m,1H),2.64(s,3H),2.42(s,1H),1.26(s,1H).LC-MS(ESI):m/z=487.1[M+H]+.
实施例66:
以6C和4-溴-3氯-5-氟苯胺为原料,参考实施例17实验操作(第一到第四步),得到化合物66。
1H NMR(400MHz,CD3OD)δ8.58(s,1H),7.50-7.49(d,1H),7.40-7.36(m,1H),7.30(s,1H),5.03(s,1H),4.77(d,1H),4.74-4.73(d,2H),4.71-4.70(d,1H),4.54-4.53(d,1H),4.44-4.43(d,1H),2.66(s,3H).LC-MS(ESI):m/z=556.1[M+H]+.
实施例67:
以66B为原料,参考实施例34实验操作(第一到第三步),得到化合物67。
1H NMR(400MHz,CD3OD)δ8.58(s,1H),7.77(s,1H),7.59-7.56(d,1H),7.31(s,1H),5.04(s,1H),4.56-4.55(d,1H),4.47-4.46(s,1H),2.66(s,3H).LC-MS(ESI):m/z=489.10[M+H]+.
实施例68:
第一步:以化合物55B(0.15g,0.25mmol)为原料,参考实施例13第二步操作得到目标化合物68A(0.08g,收率70.74%)。LC-MS(ESI):m/z=622.2[M+H]+.
第二步:以化合物68A(0.11g,0.18mmol)为原料,参考实施例1第二步操作得到目标化合物68(0.02g,收率19.09%)。
1H NMR(400MHz,Methanol-d4)δ8.62-8.42(m,1H),7.87-7.67(m,1H),6.84-6.64(m,1H),5.50(s,1H),4.98-4.89(m,1H),4.60-4.25(m,5H),3.23-2.90(m,4H),2.33-2.15(m,2H).LC-MS(ESI):m/z=582.2[M+H]+.
实施例69:
以6C和化合物69A为原料,参考实施例17实验操作(第一到第四步),得到化合物69。LC-MS(ESI):m/z=538.6[M+H]+.
1H NMR(400MHz,CD3OD)δ8.55(s,1H),7.65-7.64(m,1H),7.46-7.45(m,1H),7.28(s,1H),6.87-6.84(m,1H),4.98(s,1H),4.53-4.40(m,6H),2.65(s,3H).
实施例70:
第一步:将化合物70A(3g,20.49mmol)溶于乙醇(20mL)中,加入硝酸铁(4.96g,20.49mmol),升温至50℃搅拌反应3小时。冷却至室温,将反应液倒入水中,用乙酸乙酯萃取。有机相依次用水,饱和食盐水洗涤,减压浓缩得到粗品,粗品溶于甲苯(15mL)中,加热到50℃搅拌10分钟,缓慢滴加正己烷直至浑浊,然后缓慢冷却至室温搅拌析出大量固体,过滤,减压干燥得到化合物70B(1.2g,收率30.58%)。
第二步:将化合物70B(0.20g,1.04mmol),3,3-二氟环丁-1-醇(0.11g,1.04mmol),溶于DMF(5mL)中。降温至0℃,加入三苯基膦(0.25g,1.25mmol),搅拌30分钟后,加入DIAD(0.33g,1.25mmol),升至室温反应过夜。将反应液倒入水中,用乙酸乙酯萃取。有机相依次用水,饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到目标化合物70C(0.21g,收率71.70%)。
第三步:将化合物70C(0.21g,0.75mmol),锌粉(0.24g,3.71mmol)溶于甲醇(10mL),随后加入氯化铵(0.20g,3.71mmol),室温搅拌1小时。过滤,将滤液减压浓缩,向残余物中加入乙酸乙酯,有机相依次用水,饱和食盐水洗涤,减压浓缩得到化合物70D(0.14g,收率74.18%)。LC-MS(ESI):m/z=252.1[M+H]+.
第四步:以化合物70D(0.14g,0.56mmol)和6C(0.19g,0.56mmol)为原料,参考实施例6第四步操作得到目标化合物70E(0.14g,收率42.09%)。LC-MS(ESI):m/z=594.5[M+H]+.
第五步:以化合物70E(0.14g,0.24mmol)为原料,参考实施例6第五步操作得到目标化合物70F(0.11g,收率75.02%)。LC-MS(ESI):m/z=611.6[M+H]+.
第六步:以化合物70F(0.11g,0.18mmol)为原料,参考实施例1第二步操作得到目标化合物70(0.04g,收率38.93%)。
1H NMR(400MHz,Methanol-d4)δ8.70-8.54(m,1H),8.06-7.79(m,1H),7.35-7.27(m,1H),7.18-6.93(m,1H),5.06-4.88(m,2H),4.61-4.31(m,2H),3.33-3.22(m,2H),2.93-2.78(m,2H),2.75-2.57(m,3H)LC-MS(ESI):m/z=571.5[M+H]+.
实施例71:
以6C为原料,参考实施例17实验操作(第一到第四步),得到化合物71。
1H NMR(400MHz,Methanol-d4)δ8.55-8.52(m,1H),7.66-7.43(m,1H),7.29-7.24(m,1H),6.72-6.58(m,1H),5.01-4.89(m,1H),4.59-4.20(m,6H),2.69-2.49(d,3H).LC-MS(ESI):m/z=556.5[M+H]+.
实施例72:
以化合物71B为原料,参考实施例34实验操作(第一、二、三步),得到化合物72。
1H NMR(400MHz,Methanol-d4)δ8.57-8.49(m,1H),7.86-7.39(m,2H),7.31-7.23(m,1H),4.99-4.83(M,1H),4.60-4.45(m,2H),4.29-4.00(m,1H),2.69-2.49(m,3H).LC-MS(ESI):m/z=489.1[M+H]+.
实施例73:
第一步:将6A(0.5g,1.72mmol)及73A(0.35g,1.89mmol)溶于1,4-二氧六环(15mL)中,随后加入Pd2(dba)3(162mg,0.17mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(206mg,0.34mmol)及碳酸钾(0.48g,3.44mmol),氮气氛围,于95℃反应过夜。16h后冷却至室温,向反应液中加入30mL水,以乙酸乙酯(15mL×3)萃取,合并后的有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物73B(0.51g,68%)。LC-MS(ESI):m/z=438.1[M+H]+.
第二步:将73B(0.51g,1.17mmol)溶于甲醇(10mL)中,冰浴下加入氢氧化锂(42mg,1.75mmol),室温反应30min。加入20mL水,以乙酸乙酯(10mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到目标化合物73C(102mg,25%)。LC-MS(ESI):m/z=348.1[M+H]+.
第三步:以73C(102mg,0.29mmol)和5-氯-2,4-二氟苯胺(47mg,0.29mmol)为原料,参考实施例6第四步操作得到目标化合物73D(110mg,77%)。LC-MS(ESI):m/z=493.4[M+H]+.
第四步:以73D(110mg,0.22mmol)为原料,参考实施例6第五步操作得到目标化合物73E(94mg,84%)。LC-MS(ESI):m/z=510.0[M+H]+.
第五步:以73E(94mg,0.18mmol)为原料,参考实施例6第五步操作得到化合物73(40mg,47%)。
1H NMR(400MHz,CD3OD)δ8.53(s,1H),8.02-7.97(m,1H),7.30-7.24(m,2H),5.19(s,1H),4.66-4.63(m,1H),4.51-4.88(m,1H).LC-MS(ESI):m/z=470.0[M+H]+.
实施例74:
第一步:将74A(1g,4.79mmol)及硫氰酸钾(1.39g,14.34mmol)溶于DMSO(10mL)中,60℃反应2h。冷却至室温,加入20mL水,以乙酸乙酯(10mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=20∶1)得到目标化合物74B(0.81g,91%)。
1H NMR(400MHz,CD3Cl)δ7.71(d,J=5.6Hz,1H),7.36(d,J=5.6Hz,1H).
第二步:将74B(0.81g,4.35mmol)溶于乙酸(10mL)中,加入铁粉(100mg)后,室温反应过夜。16h后,加入30mL水,以乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物74C(316mg,46%)。LC-MS(ESI):m/z=157.1[M+H]+.
第三步:以6C(450mg,1.25mmol)及74C(200mg,1.25mmol)为原料,参考实施例6第四步操作得到目标化合物74D(280mg,45%)。LC-MS(ESI):m/z=499.0[M+H]+.
第四步:以74D(280mg,0.56mmol)为原料,参考实施例6第五步操作得到目标化合物74E(215mg,74%)。LC-MS(ESI):m/z=516.1[M+H]+.
第五步:以74E(215mg,0.42mmol)为原料,参考实施例6第六步操作得到化合物74(60mg,30%)。
1H NMR(400MHz,CD3OD)δ8.34(s,1H),7.18-7.15(m,1H),7.04(s,1H),7.00-6.97(m,1H),5.39(s,1H),4.41-4.39(m,1H),4.38-4.35(m,1H),2.18(s,3H).LC-MS(ESI):m/z=476.4[M+H]+.
实施例75:
以化合物69C为原料,参考实施例34实验操作(第一、二、三步),得到化合物75。
1H NMR(400MHz,CD3OD)δ8.57(s,1H),7.88(s,1H),7.78-7.76(d,1H),7.59-7.62(d,1H),7.30(s,1H),4.99(s,1H),4.56-4.55(d,1H),4.45(s,1H),4.01(s,1H),2.66(s,3H).LC-MS(ESI):m/z=471.4[M+H]+.
实施例76:
第一步:依次将化合物76A(0.31g,1.22mmol),(1-氟乙烯基)甲基二苯基硅烷(0.24g,1.01mmol),四三苯基磷钯(0.12g,0.1mmol),氟化铯(0.31g,2.01mmol),CuI(0.019g,0.1mmol)加入DMF(15mL)中,氮气氛围,室温下搅拌反应18小时。反应液经硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%EA/PE)纯化得到化合物76B(0.2mg,收率90.2%)。
第二步:室温下,将76B(0.2g,0.91mmol)溶于甲醇(10mL)中,加入锌粉(0.6g,9.1mmol),搅拌均匀,向反应体系中分批加入氯化铵(0.49g,9.1mmol),室温反应2小时。过滤,浓缩滤液,向残余物中加入EA(50mL),有机相用水洗(30mL×3),无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%EA/PE)纯化得到化合物76C(0.16g,收率92.74%)。LC-MS(ESI):
m/z=190.1[M+H]+.
第三步:以化合物6C(0.12g,0.33mmol)和化合物76C(0.063g,0.33mmol)为原料,参考实施例6第四步操作得到化合物76D(0.16g,收率91.16%)。LC-MS(ESI):m/z=532.1[M+H]+.
第四步:以化合物76D(0.16g,0.30mmol)为原料,参考实施例6第五步操作得到化合物76E(60mg,收率36.44%)。LC-MS(ESI):m/z=549.2[M+H]+.
第五步:以化合物76E(0.060g,0.11mmol)为原料,参考实施例1第二步操作得到目标化合物76(36mg,收率64.32%)。
1H NMR(400MHz,CD3OD)δ8.47-8.42(m,1H),8.01-7.76(m,1H),7.58-7.33(m,1H),7.25-7.14(m,1H),5.44-4.77(m,2H),4.49-4.19(m,2H),2.60-2.42(m,3H).LC-MS(ESI):m/z=509.5[M+H]+.
实施例77:
以6C(200mg,0.56mmol)和77A为原料,参考实施例8操作(第三,四,五步),得到化合物77。
1H NMR(400MHz,CD3OD)δ8.48-8.37(m,2H),8.14-7.96(m,1H),7.58-7.33(m,2H),7.16(s,1H),7.12-6.93(m,1H),5.54-5.18(m,1H),4.51-4.44(m,2H),2.52-2.31(m,3H).LC-MS(ESI):m/z=453.1[M+H]+.
实施例78:
第一步:向DMF(30mL)中依次加入氯化锂(0.70g,16.52mmol),氯化亚铜(1.65g,16.67mmol),室温下搅拌1小时,向体系中加入醋酸钾(1.63g,16.64mmol),联硼酸频那醇酯(4.23g,16.64mmol)和78A(1g,15.13mmol),室温搅拌20小时。反应完全后,加入饱和食盐水(200mL)搅拌30分钟,垫硅藻土过滤,滤饼用乙酸乙酯(30ml×3)洗涤,滤液用乙酸乙酯(50mL×3)萃取,合并后的有机相,用饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,过滤,减压浓缩,残留物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到化合物78B(686mg,收率23.36%)。
1H NMR(400MHz,CDCl3)δ5.66-5.51(m,1H),5.50-5.46(m,1H),1.55-1.46(m,1H),1.26(s,12H),0.70-0.64(m,2H),0.60-0.53(m,2H).
第二步:将化合物78B(50mg,0.26mmol)溶于二氧六环(5mL),依次向其中加入化合物17B(182mg,0.31mmol),碳酸钾(90mg,0.65mmol),Pd(dppf)Cl2(9mg,0.013mmol),氮气氛围,升温至85℃搅拌12小时。冷却至室温,加入乙酸乙酯(50mL)稀释,垫硅藻土过滤,乙酸乙酯(10mL×3)洗涤滤饼,合并有机相,减压浓缩得到粗品,粗品用Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到化合物78C(60mg,收率40.42%)。LC-MS(ESI):m/z=571.1[M+H]+.
第三步:以78C(60mg)为原料,参考实施例1第二步操作得到目标化合物78(17mg,收率29.11%)。
1H NMR(400MHz,CD3OD)δ8.50-8.40(m,1H),7.89-7.31(m,1H),7.21(s,1H),7.19-6.94(m,1H),5.45-4.78(m,3H),4.51-4.17(m,2H),2.62-2.40(m,3H),1.65-1.56(m,1H),0.74-0.61(m,2H),0.53-0.35(m,2H).LC-MS(ESI):m/z=531.1[M+H]+.
实施例79:(2S,3S,4S)-N-(5-氯-2,4-二氟苯基)-3,4-二羟基-N-(甲基-d3)-5-氧代-1-(7-(三氟甲基)呋喃并[3,2-b]吡啶-5-基)吡咯烷-2-酰胺(化合物79)
(2S,3S,4S)-N-(5-chloro-2,4-difluorophenyl)-3,4-dihydroxy-N-(methyl-d3)-5-oxo-1-(7-(trifluoromethyl)furo[3,2-b]pyridin-5-yl)pyrrolidine-2-carboxamide
第一步:依次将化合物79A(5.0g,27.29mmol),三氟乙酰乙酸乙酯(12.5g,68.22mmol)溶于冰乙酸(100mL),升温至100℃反应16小时。反应完全后冷至室温,减压浓缩,得到的粗品使用中压制备仪Biotage Isolera One(80g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物79B(4.2g,收率75.77%)。LC-MS(ESI):m/z=204.0[M+H]+.
第二步:将化合物79B(4.2g,20.68mmol)溶于三氯氧磷(25mL),升温至100℃反应8小时。待反应冷至室温,减压浓缩,得到的残余物溶于DCM(50mL),加入碳酸氢钠冰水溶液搅拌10分钟,萃取,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩滤液得到粗品,粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物79C(1.7g,收率37.10%)。LC-MS(ESI):m/z=222.4[M+H]+.
第三步:向干燥的1,4-二氧六环(10mL)中依次加入化合物79C(120.0mg,0.33mmol),化合物1A(110.0mg,0.49mmol),碳酸钾(91.0mg,0.66mmol),Pd2(dba)3(30.0mg,0.033mmol),Xantphos(76.0mg,0.13mmol),氮气氛围,升温至95℃反应过夜。待反应冷至室温,加入乙酸乙酯(30mL)稀释,垫硅藻土过滤,将滤液浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物79D(104.0mg,收率57.42%)。LC-MS(ESI):m/z=549.0[M+H]+.
第四步:将化合物79D(104.0mg,0.19mmol)溶于二氯甲烷(10mL),氮气氛围,降温至-40℃,滴加三氯化硼(1M,二氯甲烷溶液,1.0mL),滴加完毕,反应20分钟,自然升至室温反应3h。将反应液缓慢滴入碳酸氢钠冰水溶液中,搅拌30分钟,分出有机相,水相用二氯甲烷(30mL)萃取,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物79(32.0mg,收率33.10%)。LC-MS(ESI):m/z=509.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.49-8.46(m,1H),8.17-8.11(m,1H),8.09-7.80(m,1H),7.48-7.35(m,1H),7.15-6.92(m,1H),5.80-5.04(m,1H),4.26-4.17(m,2H).
实施例80:
第一步:室温下,将17B(100mg,0.17mmol)溶解于干燥的甲苯(10mL)中,向其中依次加入氨基甲酸叔丁酯(24mg,0.21mmol),Pd2(dba)3(31mg,0.034mmol),BINAP(32mg,0.051mmol),CS2CO3(170mg,0.51mmol),搅拌均匀后,氮气氛围,100℃反应16小时。TLC监测原料消失,停止反应。冷却至室温,向反应液中加入EA(10mL),有机相用水(15mL)洗涤两次,饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩后,残余物经柱层析(PE∶EA=4∶1)分离纯化得到产物80A(95mg,90.13%)。LC-MS(ESI):m/z=620.2[M+H]+.
第二步:室温下,将80A(95mg,0.15mmol)溶解于干燥的二氯甲烷(5mL)中,向溶液中滴加盐酸的甲醇溶液(10mL,4mol/L),滴加完毕后,室温反应4小时。TLC 监测原料消失,停止反应。将反应液浓缩,残余物用二氯甲烷(10mL)溶解,滴加饱和碳酸氢钠水溶液将pH调节为碱性,萃取分液,有机相用饱和食盐水(10mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到产物80B(45mg,62.52%)。LC-MS(ESI):m/z=480.5[M+H]+.
第三步:室温下,将80B(45mg,0.094mmol)溶解于乙腈(5mL)中,冰浴下,滴加亚硝酸叔丁酯(15mg,0.14mmol),搅拌1分钟,滴加叠氮基三甲基硅烷(16mg,0.14mmol),滴加完毕后,室温反应1小时。TLC监测原料消失,停止反应。将反应液浓缩,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物80(15mg,31.55%)。
1H NMR(400MHz,CD3OD)δ8.50-8.38(m,1H),7.87-7.70(m,1H),7.39-7.36(d,1H),7.26-7.14(m,1H),4.88-4.79(d,1H),4.46-4.42(m,1H),4.19-4.18(d,1H),2.60-2.40(m,3H).LC-MS(ESI):m/z=506.1[M+H]+.
实施例81:
第一步:在单口瓶中,依次加入化合物29A(0.3g,1.45mmol),4-异噁唑硼酸频那醇酯(0.43g,2.2mmol),Pd(dppf)Cl2(0.11g,0.14mmol),碳酸钾(0.6g,4.34mmol),二氧六环(10mL),去离子水(2mL),氮气置换三次,升温至85℃搅拌反应过夜。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到目标化合物81A(0.07g,收率24.61%)。LC-MS(ESI):m/z=197.1[M+H]+.
第二步:以化合物81A(0.07g,0.36mmol)和6C(0.15g,0.43mmol)为原料,参考实施例8第三步操作得到目标化合物81B(0.05g,收率25.80%)。LC-MS(ESI):m/z=539.1[M+H]+.
第三步:以化合物81B(0.05g,0.093mmol)为原料,参考实施例8第四步操作得到目标化合物81C(0.045g,收率87.11%)。LC-MS(ESI):m/z=556.6[M+H]+.
第四步:以化合物81C(0.045g,0.08mmol)为原料,参考实施例1第二步操作得到目标化合物81(6mg,收率14.37%)。
1H NMR(400MHz,Methanol-d4)δ9.09-8.95(m,1H),8.88-8.74(m,1H),8.49-8.38(m,1H),8.10-7.93(m,1H),7.45-7.30(m,1H),7.23-7.09(m,1H),4.90-4.80(m,1H),4.62-4.40(m,2H),2.55-2.39(m,3H).LC-MS(ESI):m/z=516.2[M+H]+.
实施例82:
第一步:将82A(1g,4.32mmol)、碳酸氢钾(0.48g,4.75mmol)及碳酸钾(0.66g,4.75mmol)溶于水(10mL)中,冰浴下滴加三光气(0.64g,2.16mmol)的甲苯(10mL)溶液,冰浴下继续反应2h。向反应液中加入20mL水,以乙酸乙酯(15mL×3)萃取,合并后的有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚:乙酸乙酯(v/v)=10∶1)得到82B(0.93g,97%)。LC-MS(ESI):m/z=222.2[M+H]+.
第二步:将82B(0.93g,4.20mmol)及2-氯-6-甲基-4-(三氟甲基)吡啶(0.90g,4.62mmol)溶于1,4-二氧六环(25mL)中,随后加入Pd2(dba)3(380mg,0.42mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂葸(490mg,0.84mmol)及碳酸钾(1.16g,8.40mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入50mL水,乙酸乙酯(25mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到82C(583mg,36%)。LC-MS(ESI):m/z=381.1[M+H]+.
第三步:将82C(583mg,1.53mmol)溶于甲醇(5mL)及四氢呋喃(5mL)的混合溶剂中,加入10%钯碳粉末(40mg),氢气氛围下反应过夜。待反应完全后,过滤,少量甲醇洗滤饼,将滤液减压浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到82D(415mg,93%)。LC-MS(ESI):m/z=291.2[M+H]+.
第四步:以82D(415mg,1.43mmol)及4-溴-5-氯-2-氟苯胺(0.32g,1.43mmol)为原料,参考实施例8第三步操作得到标题化合物82E(502mg,71%)。LC-MS(ESI):m/z=496.0[M+H]+.
第五步:以82E(502mg,1.01mmol)为原料,参考实施例8第四步操作得到82F(318mg,61%)。LC-MS(ESI):m/z=513.0[M+H]+.
第六步:以82F(150mg,0.29mmol)和3,3-二氟环丁胺(32mg,0.35mmol)为原料,参考实施例13第二步操作得到化合物82(77mg,50%)。
1H NMR(400MHz,CDCl3)δ8.34(s,1H),7.56-7.54(m,1H),7.09(s,1H),6.37-6.35(m,1H),5.16-5.12(m,1H),4.52-4.45(m,4H),4.36-4.31(m,2H),2.59(s,3H).LC-MS(ESI):m/z=526.4[M+H]+.
实施例83:
第一步:以82F(130mg,0.25mmol)为原料,参考实施例34第一步操作得到83A(93mg,69%)。LC-MS(ESI):m/z=531.0[M+H]+.
第二步:以83A(93mg,0.17mmol)为原料参考实施例34第三步操作得到化合物83(35mg,44%)。LC-MS(ESI):m/z=459.1[M+H]+.
1H NMR(400MHz,Chloroform-d)δ8.53(s,1H),7.53-7.51(m,1H),7.15(s,1H),7.12-7.10(m,1H),5.66(s,1H),4.84-4.80(m,1H),4.56(s,1H),4.07(s,1H),2.40(s,3H).
实施例84:
以6C(200mg,0.56mmol)和84A为原料,参考实施例17操作(第一到第四步反应),得到化合物84。LC-MS(ESI):m/z=518.6[M+H]+.
1H NMR(400MHz,CD3OD)δ8.40(s,1H),7.39-7.36(m,1H),7.29(s,1H),6.77-6.75(m,1H),5.13-5.12(m,1H),4.36-4.30(m,4H),4.23-4.19(m,2H),2.65(s,3H),2.31(s,3H).
实施例85:
第一步:将6A(600mg,2.06mmol)及85A(667mg,3.09mmol)溶于1,4-二氧六环(50mL)中,随后加入Pd2(dba)3(377mg,0.41mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂葸(477mg,0.82mmol)及碳酸钾(854mg,6.18mmol),氮气氛围,于100℃反应3小时。反应完全后,冷却至室温,减压浓缩除去反应溶剂,残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物85B(738mg,76%)。LC-MS(ESI):m/z=471.1[M+H]+.
第二步:将85B(738mg,1.57mmol)溶于乙酸乙酯(20mL)中,加入氢氧化钯炭(375mg),氢气氛围下反应1小时。过滤,EA(30mL×2)洗涤滤饼,滤液减压浓缩后得到粗 品,粗品经柱层析硅胶分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到85C(463mg,77%)。
1H NMR(400MHz,CD3Cl)δ8.60(s,1H),7.33(s,1H),5.28(d,1H),5.12(t,1H),4.92(d,1H),1.47(s,3H),1.45(s,3H).
第三步:以85C(551mg,1.45mmol)和5-氯-2,4-二氟苯胺(356mg,2.17mmol)为原料,参考实施例6第四步操作得到85D(283mg,37%)。LC-MS(ESI):m/z=523.8[M-H]-.
第四步:以85D(283mg,0.54mmol)及三甲基乙炔基硅(424mg,4.32mmol)为原料,参考实施例34第一步操作得到85E(213mg,67%)。LC-MS(ESI):m/z=588.5[M+H]+.
第五步:以85E(150mg,0.26mmol)为原料,参考实施例8第四步操作得到85F(120mg,87%)。LC-MS(ESI):m/z=533.5[M+H]+.
第六步:以85F(106mg,0.2mmol)为原料,参考实施例1第二步操作得到化合物85(70mg,71%)。LC-MS(ESI):m/z=493.5[M+H]+.
1H NMR(400MHz,CD3OD)δ8.77(s,1H),8.35(m,1H),7.61(s,1H),7.49(m,1H),4.83(s,1H),4.57(d,1H),4.30(d,1H),4.17(s,1H).
实施例86:
第一步:将86A(1g,4.31mmol)溶于吡啶(20mL),降温至0℃,加入HATU(3.28g,8.62mmol),搅拌20min。加入4-溴-5-氯-2-氟苯胺(2.42g,10.77mmol),搅拌5min,随后升温至75℃反应过夜。冷却至室温,减压浓缩除去大部分吡啶,加入乙酸乙酯稀释,有机相用盐酸(0.5M)多次洗涤,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到目标化合物86B(0.74g,收率39.23%)。LC-MS(ESI):m/z=337.0[M-Boc+H]+.
第二步:将86B(0.74g,1.49mmol)溶于二氯甲烷(20mL),降温至0℃,加入咪唑(0.34g,5.04mmol),叔丁基二甲基氯硅烷(1.27g,8.43mmol),自然升至室温反应过夜。减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到目标化合物86C(0.63g,收率67.54%)。LC-MS(ESI):m/z=451.1[M-Boc+H]+.
第三步:以化合物86C(0.63g,1.14mmol)为原料,参考实施例8第四步操作得到目标化合物86D(0.61g,收率94.04%)。LC-MS(ESI):m/z=468.1[M-Boc+H]+.
第四步:以化合物86D(0.2g,0.35mmol)为原料,参考实施例13第二步操作得到目标化合物86E(0.17g,收率83.58%)。LC-MS(ESI):m/z=481.3[M-Boc+H]+.
第五步:将86E(0.17g,0.29mmol)溶于二氯甲烷(10mL),降温至0℃,加入叔丁基二甲硅基三氟甲磺酸酯(0.23g,0.87mmol),搅拌5min,加入卢剔啶(0.09g,0.84mmol),随后升温至室温反应1小时。反应液倒入水中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到目标化合物86F(0.12g,收率86.02%)。LC-MS(ESI):m/z=481.3[M+H]+.
第六步:以86F(0.12g,0.25mmol)和2-氯-6-甲基-4-三氟甲基烟腈(0.08g,0.36mmol)为原料,参考实施例1第一步操作得到目标化合物86G(0.09g,收率54.12%)。LC-MS(ESI):m/z=665.3[M+H]+.
第七步:将化合物86G(0.09g,0.14mmol)溶于THF(50mL),加入TBAF四氢呋喃溶液(1M,0.2mL,0.2mmol),室温搅拌2小时。浓缩,残余物溶于乙酸乙酯(20mL),有机相用饱和食盐水多次洗,无水硫酸钠干燥,过滤,浓缩得到棕色粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物86(0.045g,收率57.76%)。
1H NMR(400MHz,Methanol-d4)δ7.86-7.63(M,1H),7.02-6.89(m,1H),6.74-6.49(m,1H),5.01-4.76(m,1H),4.54-4.39(M,4H),4.30-4.20(m,1H),3.29-3.22(m,2H),2.62-2.42(m,3H),2.13-1.93(m,2H).LC-MS(ESI):m/z=551.6[M+H]+.
实施例87:
第一步:以化合物86D(0.25g,0.44mmol)为原料,参考实施例34第一步操作得到目标化合物87A(0.18g,收率69.78%)。LC-MS(ESI):m/z=414.6[M-Boc-TMS+H]+.
第二步:向化合物87A(0.18g,0.31mmol)中加入4M盐酸-二氧六环(10mL)溶液,室温反应2小时。减压浓缩,得到目标化合物87B的盐酸盐(0.15g)。LC-MS(ESI):m/z=486.3[M+H]+.
第三步:将87B的盐酸盐(0.15g,0.29mmol)溶于N-甲基吡咯烷酮(5mL),依次加入2-氯-6-甲基-4-三氟甲基烟腈(0.1g,0.45mmol),DIPEA(0.08g,0.62mmol),随后升温至65℃反应过夜。冷却至室温,反应液倒入水中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩后得到粗品,粗品用中压制备仪Biotage Isolera One(8g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到目标化合物87C(0.12g,收率61.73%)。LC-MS(ESI):m/z=670.3[M+H]+.
第四步:以化合物87C(0.12g,0.18mmol)为原料,参考实施例34第三步操作得到目标化合物87(0.01g,收率11.48%)。
1H NMR(400MHz,Methanol-d4)δ8.06-7.78(m,1H),7.53-7.23(m,1H),6.96-6.74(m,1H),5.28-5.06(m,1H),4.65-4.51(m,1H),4.27-3.73(m,3H),2.53-2.13(m,3H),2.12-1.87(m,2H).LC-MS(ESI):m/z=484.4[M+H]+.
实施例88:
第一步:室温下,将88A(4.0g,17.93mmol)溶解于干燥的THF(40mL)中,氮气氛围,-78℃下向反应液中滴加LDA(10mL,2mol/L),滴加完毕后,-78℃反应1.5小时。向反应液中滴加1,3,2-二噁唑噻吩-2,2-二氧化物(2.67g,21.52mmol)的THF(5mL)溶液,滴加完毕后,自然升温至室温反应16小时。TLC监测原料消失,冷至0℃,向反应体系中滴加盐酸(8mL,80mmol),滴加完毕后,自然升温至室温反应3小时。将反应液倒入饱和碳酸氢钠溶液中淬灭反应,用EA(40mL×2)萃取水相,合并有机相,有机相用饱和食盐水(40mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化得到化合物88B(3.3g,68.92%)。LC-MS(ESI):m/z=267.9[M+H]+.
第二步:室温下,将88B(3.3g,12.36mmol)溶解于1,4-二氧六环(30mL)中,加入碳酸铯(8.05g,24.73mmol),升温至100℃反应16小时。TLC监测原料消失,停止反应,冷至室温。向反应液中加入EA(30mL),有机相用水(40mL×2)洗涤,饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化得到化合物88C(2.6g,85.15%)。LC-MS(ESI):m/z=248.0[M+H]+.
第三步:室温下,将88C(2.6g,10.53mmol)溶于干燥的DMSO(15mL)中,依次加入(三氟甲基)三甲基硅烷(4.49g,31.59mmol),硼酸三甲酯(3.28g,31.59mmol),碘化亚铜(200mg,1.05mmol),1,10-菲罗啉(190mg,1.05mmol),氟化钾(1.84g,31.59mmol)。氮气氛围下,升温至60℃反应16小时。TLC监测原料消失,停止反应并冷至室温。向反应液中加入EA(20mL),有机相用水(30mL×2)洗涤,饱和食盐水(30mL×2)洗涤,无水硫酸纳干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化得到化合物88D(1.9g,95.40%)。LC-MS(ESI):m/z=190.1[M+H]+.
第四步:室温下,将88D(1.9g,10.05mmol)溶于干燥的DCM(20mL)。冰浴,氮气氛围下,分批加入间氯过氧苯甲酸(2.08g,12.07mmol),加入完毕,自然升至室温反 应1.5小时。TLC监测原料消失,停止反应。向反应液中加入DCM(20mL),有机相依次用饱和碳酸氢钠水溶液(30mL×2)洗涤,饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物88E(1.3g,63.06%)。LC-MS(ESI):m/z=206.1[M+H]+.
第五步:室温下,将88E(1.3g,6.34mmol)溶于三氯氧磷(20mL)中,升温至100℃反应6小时。TLC监测原料消失,停止反应。冷至室温后,浓缩反应液,残余物用EA(20mL)稀释,倒入冰水(20mL)中,滴加饱和碳酸氢钠水溶液调节pH为碱性,萃取,分液,有机相用饱和食盐水(30mL)洗涤,无水硫酸纳干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=50∶1)分离纯化得到化合物88F(150mg,10.85%)。
第六步:室温下,将6A(150mg,0.51mmol)溶于干燥的二氧六环(10mL),依次加入88F(0.14g,0.63mmol),Pd2(dba)3(93mg,0.1mmol),XantPhos(89mg,0.15mmol),碳酸钾(210mg,1.53mmol),氮气氛围,升温至100℃反应16小时。TLC监测原料消失,停止反应。待反应冷至室温,向反应液中加入EA(20mL),有机相用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化得到化合物88G(150mg,61.48%)。LC-MS(ESI):m/z=479.3[M+H]+.
第七步:室温下,将88G(150mg,0.31mmol)溶于甲醇(20mL),向反应液中加入钯碳(66mg,0.062mmol),氢气氛围下,室温反应16小时。TLC监测原料消失,停止反应。将反应液过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物88H(88mg,73.11%)。LC-MS(ESI):m/z=389.3[M+H]+.
第八步:以88H(88mg,0.23mmol)和5-氯-2,4-二氟苯胺(45mg,0.28mmol)为原料,参考实施例8第三步操作得到88I(60mg,48.87%)。LC-MS(ESI):m/z=534.5[M+H]+.
第九步:以88I(60mg,0.11mmol)为原料,参考实施例8第四步操作得到产物88J(55mg,90.76%)。LC-MS(ESI):m/z=551.5[M+H]+.
第十步:以88J(55mg,0.1mmol)为原料,参考实施例1第二步操作得到化合物88(25mg,48.94%)。LC-MS(ESI):m/z=511.4[M+H]+.
1H NMR(400MHz,CD3OD)δ8.11-8.08(m,1H),8.06(s,1H),7.37-7.32(m,1H),4.69-4.66(m,2H),4.56(s,1H),4.49-4.48(d,1H),4.15-4.14(d,1H),3.34-3.30(m,2H).
实施例89:
第一步:以6C(650mg,1.80mmol)和89A(330mg,1.80mmol)为原料,参考实施例8第三步操作得到化合物89B(500mg,收率52.6%)。
1H NMR(400MHz,DMSO-d6)δ10.36(s,1H),8.36(s,1H),7.53-7.51(m,1H),7.49-7.45(m,1H),7.45-7.42(m,1H),7.32-7.29(m,1H),5.31-5.28(m,1H),5.09-5.04(m,1H),5.00-4.97(m,1H),2.38(s,3H),2.29(s,3H),1.34-1.28(m,6H).
第二步:以89B(500mg,0.95mmol)为原料,参考实施例8第四步操作得到目标化合物89C(423mg,收率81.65%)。LC-MS(ESI):m/z=545.6[M+H]+.
第三步:以89C(423mg,0.77mmol)为原料,参考实施例34第一步操作得到目标化合物89D(80mg,收率15.45%)。
第四步:将89D(80mg)溶于干燥的二氯甲烷(2mL),氮气氛围下,冷却至-30℃,滴加BCl3(0.4mL,1M in DCM),滴加完毕后,自然升至室温,反应2小时。反应完毕后,滴加饱和碳酸氢钠水溶液(20mL)淬灭,二氯甲烷(10mL×3)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩得到89E粗品,无需进一步纯化直接进行下一步。
第五步:
将89E粗品溶于干燥的四氢呋喃(2mL),加入TBAF(284mg,1.09mmol),室温搅拌1小时。反应完毕,加入饱和食盐水(10mL),用乙酸乙酯(5mL×3)萃取,合并后的有机相用饱和食盐水洗三次,无水硫酸钠干燥,过滤,浓缩,残余物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%MeOH/DCM)纯化得到目标化合物89(6mg,两步收率9.3%)。LC-MS(ESI):m/z=451.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.41(s,1H),7.65-7.61(m,1H),7.56(s,1H),7.49-7.43(m,1H),7.30(s,1H),5.17-5.12(m,1H),4.27-4.18(m,2H),3.86(s,1H),2.65(s,3H),2.54(s,3H).
实施例90:
第一步:将化合物79C(0.5g,2.26mmol)溶于二氧六环(20mL),依次加入化合物82B(0.55g,2.49mmol),Pd2(dba)3(0.21g,0.23mmol),XantPhos(0.26g,0.45mmol),碳 酸钾(0.62g,4.52mmol),氮气氛围,升温至95℃搅拌反应18小时。冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相,依次用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物90A(0.8g,收率87.12%)。LC-MS(ESI):m/z=407.0[M+H]+.
第二步:将化合物90A(0.22g,0.54mmol)溶于甲醇(10mL),冷却至0℃,滴加6mol/L氢氧化钠水溶液(0.27mL,1.62mmol),升温至室温搅拌反应1小时。冰浴下,向反应液滴加到5%柠檬酸水溶液调节pH至弱酸性。用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物90B(0.16g,收率93.71%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=317.1[M+H]+.
第三步:以90B(0.16g,0.51mmol)和5-氯-2,4-二氟苯胺(0.1g,0.61mmol)为原料,参考实施例8第三步操作得到化合物90C(0.12g,收率50.96%)。LC-MS(ESI):m/z=462.5[M+H]+.
第四步:以化合物90D(0.12g,0.26mmol)为原料,参考实施例8第四步操作得到化合物90(70mg,收率56.23%)。LC-MS(ESI):m/z=479.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.37-8.32(m,1H),8.17-8.10(m,1H),8.07-7.65(m,1H),7.50-7.36(m,1H),7.18-6.92(m,1H),5.15-5.08(m,1H),4.30-4.24(m,2H).
实施例91:
第一步:将化合物91A(3g,11.36mmol)溶于甲醇(14mL),冰浴下滴加二氯亚砜(1mL),滴加完毕升至70℃搅拌3小时。反应完全后,冷至室温,减压浓缩,向残余物中加入二氯甲烷(30mL),水(10mL),萃取,水相用二氯甲烷反萃(10mL×2),合并后的有机相依次用饱和碳酸氢钠水溶液洗,饱和食盐水溶液洗,无水硫酸钠干燥,过滤,浓缩,得到标题化合物91B粗品(2.81g,89%)。LC-MS(ESI):m/z=279.2[M+H]+.
第二步:室温下,将化合物91B(2.81g,10.10mmol)溶于二碳酸二叔丁酯(6mL)中,加入DMAP(123mg,1.01mmol),升温至60℃,反应1小时。浓缩,残留物用硅胶柱色谱分离提纯(石油醚:乙酸乙酯(v/v)=10∶1-5∶1)得到标题化合物91C(3.01g,79%)。LC-MS(ESI):m/z=379.2[M+H]+.
第三步:将化合物91C(3.01g,8.01mmol)溶于四氢呋喃(10mL)及甲醇(10mL)的混合溶剂,加入10%钯碳(300mg),氢气氛围下反应3h。过滤,除去不溶物,乙酸乙酯洗滤饼,合并滤液,浓缩后得到目标化合物91D粗品(1.79g,92%),未经进一步纯化,直接进行下一步反应。LC-MS(ESI):m/z=245.1[M+H]+.
第四步:将化合物91D(1.79g,7.32mmol)及2-溴-6-甲基-4-三氟甲基吡啶(1.75g,7.32mmol)溶于1,4-二氧六环(15mL),依次加入Pd2(dba)3(668mg,0.73mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂葸(828mg,1.46mmol)及碳酸铯(7.14g,21.96mmol),氮气 氛围,于95℃反应4h。冷却至室温,向反应液中加入100mL水,乙酸乙酯(30mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚:乙酸乙酯(v/v)=5∶1)得到目标化合物91E(2.21g,75%)。LC-MS(ESI):m/z=404.0[M+H]+.
第五步:依次将化合物91E(2.21g,5.50mmol)、氢氧化锂(1.16g,27.52mmol)加入四氢呋喃(8mL)和水(8mL)的混合溶剂中,室温下搅拌12h。反应完全后,加入柠檬酸水溶液调pH至4-5,向反应液中加入100mL水,以二氯甲烷(30mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到化合物91F的粗品(1.5g,98%)。不需要进一步纯化直接进行下一步反应。
LCMS m/z=290.2[M+H]+.
第六步:将化合物91F(1.0g,3.34mmol)溶于DMF(25mL),加入碳酸钾(2.06g,14.91mmol),缓慢滴加苄溴(0.64g,3.78mmol),室温反应1.5h。向反应液中加入50mL水,乙酸乙酯(20mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后得到粗品标题化合物91G(1.16g,69%)。LC-MS(ESI):m/z=380.1[M+H]+.
第七步:室温下,将化合物91G(1.16g,3.10mmol)溶于四氢呋喃(5mL),依次加入二碳酸二叔丁酯(14.20g,65.10mmol),DMAP(38mg,0.31mmol),60℃反应1小时。冷至室温后,减压浓缩,残留物用硅胶柱色谱分离提纯(石油醚:乙酸乙酯(v/v)=10∶1-5∶1)得到标题化合物91H(1.30g,87%)。LC-MS(ESI):m/z=480.2[M+H]+.
第八步:将化合物91H(1.30g,2.71mmol)溶于四氢呋喃(10mL)及甲醇(10mL)的混合溶剂,加入10%钯碳(200mg),氢气氛围下反应3h。过滤除去不溶物,以乙酸乙酯洗滤饼,合并滤液,浓缩后得到目标化合物91I(1.0g,94%),未经进一步纯化,直接进行下一步反应。LC-MS(ESI):m/z=390.1[M+H]+.
第九步:以化合物91I(1.0g,2.56mmol)及4-溴-5-氯-2-氟苯胺(630mg,2.81mmol)为原料,参考实施例8第三步操作得到目标化合物91J(700mg,46%)。LC-MS(ESI):m/z=595.1[M+H]+.
第十步:以化合物91J(700mg,1.18mmol)为原料,参考实施例8第四步操作得到目标化合物91K(600mg,83%)。LC-MS(ESI):m/z=612.2[M+H]+.
第十一步:以化合物91K(200mg,0.33mmol)为原料,参考实施例13第二步操作得到标题化合物91L(110mg,54%)。LC-MS(ESI):m/z=625.1[M+H]+.
第十二步:依次将化合物91L(60mg,0.1mmol),2,6二甲基吡啶(23mg,0.21mmol)溶于二氯甲烷(10mL),冰浴下,加入叔丁基二甲硅基三氟甲磺酸酯(40mg,0.15mmol),反应4小时。TLC监测原料消失,加入饱和氯化铵水溶液淬灭。向反应液中加入水(15mL)萃取,有机相用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物91(20mg,收率38%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-55%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,Methanol-d4)δ8.44-8.14(m,1H),7.61(d,1H),6.98(d,1H),6.62-6.52(m,1H),4.96-4.87(m,1H),4.54-4.24(m,4H),3.58-3.32(m,2H),2.45(d,3H).LC-MS(ESI):m/z=525.2[M+H]+.
实施例92:
第一步:以化合物91F(300mg,1.04mmol)及4-溴-5-氯-2-氟苯胺(233mg,1.04mmol)为原料,参考实施例8第三步操作得到目标化合物92A(267mg,52%)。LC-MS(ESI):m/z=495.1[M+H]+.
第二步:以92A(267mg,0.54mmol)为原料,参考实施例8第四步操作得到92B(230mg,81%)。LC-MS(ESI):m/z=529.3[M+H]+.
第三步:以92B(230mg,0.44mmol)及三甲基乙炔基硅(214mg,2.18mmol)为原料,参考实施例34第一步操作得到标题化合物92C(145mg,61%)。LC-MS(ESI):m/z=547.5[M+H]+.
第四步:将化合物92C(145mg,0.27mmol)溶于甲醇(8mL),加入碳酸钾(110mg,0.8mmol),室温搅拌2小时。浓缩除去大部分溶剂,加入20mL乙酸乙酯,有机相用饱和食盐水多次洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到棕色粗品,使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物92(25mg,收率19%)。
1H NMR(400MHz,Methanol-d4)δ8.23(d,1H),7.87(d,1H),7.51(d,1H),6.97(d,1H),5.07-4.77(m,1H),3.63-3.23(m,3H),2.44(d,3H).LC-MS(ESI):m/z=475.4[M+H]+.
实施例93:
以化合物91K为原料,参考实施例34操作(第一,二,三步),得到化合物93。
1H NMR(400MHz,Methanol-d4)δ8.23(d,1H),7.87(d,1H),7.77(d,1H),6.98(d,1H),5.00-4.83(m,1H),4.21-3.37(m,3H),2.45(d,3H).LC-MS(ESI):m/z=458.4[M+H]+.
实施例94:
第一步:将17B(250mg,0.43mmol)、乙烯基三氟硼酸钾(75mg,0.56mmol)溶于1,4-二氧六环(10mL)及水(1mL)的混合溶剂,随后加入二氯[1,1′-二(二苯基膦)二茂铁]钯(31mg,0.043mmol)及碳酸钾(120mg,0.86mmol),氮气氛围,90℃反应过夜。待反应冷至室温,向反应液中加入30mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(25mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=8∶1)得到94A(170mg,74%)。LC-MS(ESI):m/z=531.6[M+H]+.
第二步:将94A(170mg,0.32mmol)溶于四氢呋喃(3mL)及水(3mL)的混合溶剂中,随后加入二水合锇酸钾(12mg,0.032mmol)及高碘酸钠(140mg,0.64mmol),室温反应过夜。待反应完全后,向反应液中加入15mL水,乙酸乙酯(10mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚:乙酸乙酯(v/v)=5∶1)得到94B(95mg,56%)。LC-MS(ESI):m/z=533.5[M+H]+.
第三步:将94B(95mg,0.18mmol)溶于甲醇(5mL)中,加入3,3-二氟氮杂环丁烷(25mg,0.27mmol)、三乙胺(91mg,0.90mmol)及5滴冰醋酸,室温搅拌2h,随后加入氰基硼氢化钠(23mg,0.37mmol),室温反应过夜。待反应完全后,向反应液中加入15mL水,乙酸乙酯(10mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到94C(70mg,64%)。LC-MS(ESI):m/z=610.2[M+H]+.
第四步:以94C(70mg,0.12mmol)为原料,参考实施例1第二步操作得到标题化合物94(32mg,37%)。
1H NMR(400MHz,CDCl3)δ8.48(s,1H),7.74-7.72(m,1H),7.45-7.42(m,1H),7.07(s,1H),4.96(s,1H),4.62-4.60(m,1H),4.35-4.32(m,1H),3.85(s,2H),3.75-3.68(m,4H),2.58(s,3H).LC-MS(ESI):m/z=570.0[M+H]+.
实施例95:
第一步:以55B(270mg,0.44mmol)为原料,参考实施例94第一步操作得到95A(176mg,72%)。LC-MS(ESI):m/z=557.6[M+H]+.
第二步:以95A(176mg,0.32mmol)为原料,参考实施例6第六步操作得到化合物95(68mg,41%)。
1H NMR(400MHz,CDCl3)δ8.49(s,1H),7.80-7.77(m,1H),7.50-7.47(m,1H),7.09-7.01(m,1H),5.85-5.80(m,1H),5.56-5.52(m,1H),5.01(s,1H),4.76-4.74(m,1H),4.44-4.42(m,1H),3.18-3.01(m,4H),2.27-2.18(m,2H).LC-MS(ESI):m/z=517.7[M+H]+.
实施例96:
以化合物55B为原料,参考实施例95操作(第一,二步),得到化合物96。
1H NMR(400MHz,CDCl3)δ8.47(s,1H),7.78-7.76(m,1H),7.19-7.16(m,1H),5.33-5.31(m,1H),5.09(s,1H),5.01(s,1H),4.78-4.76(m,1H),4.45-4.43(m,1H),3.15-3.05(m,4H),2.26-2.20(m,2H),2.13(s,3H).LC-MS(ESI):m/z=531.8[M+H]+.
实施例97:
第一步:将82B(0.6g,2.71mmol)及21D(0.72g,3.25mmol)溶于1,4-二氧六环(15mL),依次加入Pd2(dba)3(250mg,0.27mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(310mg,0.54mmol)及碳酸钾(1.77g,5.42mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入50mL水,乙酸乙酯(25mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到97A(780mg,71%)。LC-MS(ESI):m/z=407.4[M+H]+.
第二步:将97A(780mg,1.92mmol)溶于甲醇(8mL)及四氢呋喃(8mL)的混合溶剂,加入10%钯碳粉末(50mg),氢气氛围下反应过夜。待反应完全后,过滤,少量甲醇洗滤饼,将滤液减压浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到97B(557mg,92%)。LC-MS(ESI):m/z=317.1[M+H]+.
第三步:以97B(557mg,1.76mmol)及4-溴-5-氯-2-氟苯胺(0.40g,1.76mmol)为原料,参考实施例8第三步操作得到标题化合物97C(640mg,70%)。LC-MS(ESI):m/z=522.4[M+H]+.
第四步:以97C(640mg,1.22mmol)为原料,参考实施例8第四步操作得到97D(560mg,85%)。LC-MS(ESI):m/z=539.0[M+H]+.
第五步:以97D(200mg,0.37mmol)和3-氟环丁胺(36mg,0.48mmol)为原料,参考实施例13第二步操作得到化合物97(65mg,33%)。
1H NMR(400MHz,CDCl3)δ8.19(s,1H),7.44-7.41(m,1H),6.26-6.22(m,1H),5.42-5.32(m,1H),5.28-5.22(m,1H),4.45-4.35(m,2H),4.28-4.25(m,2H),4.18-4.08(m,2H),3.01-2.96(m,4H),2.19-2.10(m,2H).LC-MS(ESI):m/z=534.2[M+H]+.
实施例98:
第一步:依次将化合物6A(2.9g,9.96mmol),化合物79C(2.65g,11.95mmol)溶于干燥二氧六环(45mL),向其中依次加入Pd2(dba)3(0.91g,1.0mmol),XantPhos(1.73g,2.99mmol),碳酸钾(2.75g,19.90mmol),氮气氛围下,升温至100℃反应16小时。反应结束后,冷却至室温,加入乙酸乙酯(50mL)稀释,过滤,滤液减压浓缩后得到粗品,粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物98A(2.8g,收率59.01%)。LC-MS(ESI):m/z=477.1[M+H]+.
第二步:依次将醋酸钯(1.40g,6.24mmol),三乙胺(2.29g,22.62mmol)加入二氯甲烷(30mL)中,滴加三乙基硅烷(9.89g,85.05mmol),滴加完毕后室温下搅拌搅拌5分钟。滴加化合物98A(2.7g,5.67mmol)的二氯甲烷(10mL)溶液,滴加完毕,室温搅拌1小时。将反应液过滤,滤液减压浓缩后得到的粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物98B(1.7g,收率77.62%)。LC-MS(ESI):m/z=387.3[M+H]+.
第三步:依次将化合物98B(0.5g,1.29mmol),4-溴-5-氯-2-氟苯胺(0.43g,1.94mmol)溶于吡啶(10mL),滴加T3P(4.12g,50%in EA,6.46mmol),滴加完毕后搅拌30分钟,然后升温至50℃搅拌过夜。反应完毕,减压浓缩除去大部分吡啶,然后加入乙酸乙酯(50mL)稀释,有机相用硫酸氢钾水溶液(1M,30mL)洗涤,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物98C(0.49g,收率64.09%)。LC-MS(ESI):m/z=592.5[M+H]+.
第四步:将化合物98C(0.47g,0.79mmol)溶于DMF(10mL),氮气氛围,0℃下,加入氢化钠(60%)(48.0mg,1.2mmol),搅拌5分钟后,滴加氘代碘甲烷(0.23g,1.59 mmol),滴加完毕后继续反应30分钟。加入冰的稀盐酸(1M,30mL)淬灭,乙酸乙酯(30mL)萃取,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物98D(0.45g,收率93.42%)。LC-MS(ESI):m/z=609.5[M+H]+.
第五步:将化合物98D(100mg,0.16mmol)溶于甲苯(15mL),依次加入3,3-二氟三甲叉亚胺盐酸盐(41.0mg,0.32mmol),Pd2(dba)3(29.0mg,0.032mmol),BINAP(30.0mg,0.048mmol),碳酸铯(0.31g,0.96mmol),氮气氛围,升温至100℃反应16小时。待反应冷至室温,加入乙酸乙酯(20mL)稀释,过滤,滤液减压浓缩后得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物98E(31.0mg,收率31.15%)。LC-MS(ESI):m/z=622.2[M+H]+.
第六步:将化合物98E(31.0mg,0.050mmol)溶于二氯甲烷(5mL),氮气氛围下,降温至-40℃,滴加三氯化硼(1M in DCM,0.10mmol,0.10mL),滴加完毕,继续搅拌30分钟。然后自然升至室温反应2小时。将反应液滴入饱和碳酸氢钠水溶液(20mL)中,搅拌10分钟,分液,水相用二氯甲烷萃取(20mL),合并后的有机相,用无水硫酸钠干燥,过滤,浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物98(5.1mg,收率17.53%)。
1H NMR(400MHz,CD3OD)δ8.80-8.75(m,1H),8.29-8.22(m,1H),7.92-7.61(m,1H),7.28-7.08(m,1H),6.86-6.75(m,1H),5.62-4.97(m,1H),4.64-4.62(m,1H),4.59-4.42(m,4H),4.39-4.32(m,1H).LC-MS(ESI):m/z=582.2[M+H]+.
实施例99:
以化合物98D为原料,参考实施例95操作(第一,二步),得到化合物99。
1H NMR(400MHz,CD3OD)δ8.69-8.64(m,1H),8.18-8.11(m,1H),7.99-7.65(m,1H),7.34-7.23(m,1H),7.19-6.96(m,1H),5.52-4.98(m,1H),4.89-4.71(m,2H),4.55-4.24(m,2H),2.04-2.03(m,3H).LC-MS(ESI):m/z=531.5[M+H]+.
实施例100:
第一步:向化合物100A(3g,32.2mmol)中加入三氟乙酰乙酸乙酯(8.89g,48.32mmol),升温至110℃搅拌反应1小时。冷却至室温,减压浓缩除去未反应的原料,随后加入75%H2SO4溶液,升温至90℃搅拌反应1小时。冷却至室温,将反应液缓慢倒入冰块中,搅拌有白色固体析出,过滤,收集滤饼得到目标化合物100B(3.2g,收率46.62%)。LC-MS(ESI):m/z=214.0[M+H]+.
第二步:向化合物100B(1.8g,8.44mmol)中加入三氯氧磷(10mL),升温至100℃反应过夜。冷却至室温,减压浓缩,将残余物逐滴滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯萃取三次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(40g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到目标化合物100C(1.3g,收率66.51%)。
1H NMR(400MHz,Chloroform-d)δ8.16-8.08(m 2H),7.89-7.80(m,1H),7.75-7.66(m,2H).
第三步:以化合物100C(1g,4.32mmol)为原料,参考实施例98第一步操作得到目标化合物100D(1.2g,收率57.1%)。LC-MS(ESI):m/z=487.4[M+H]+
第四步:以化合物100D(1.2g,2.47mmol)为原料,参考实施例98第二步操作得到目标化合物100E(0.9g,91.94%)。无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=397.1[M+H]+.
第五步:以化合物100E(0.9g,2.27mmol)和5-氯-2,4-二氟苯胺(0.45g,2.72mmol)为原料,参考实施例98第三步操作得到目标化合物100F(0.17g,收率13.82%)。LC-MS(ESI):m/z=542.5[M+H]+.
第六步:以化合物100F(0.12g,0.31mmol)为原料,参考实施例98第四步操作得到目标化合物100G(0.12g,收率69.26%)。LC-MS(ESI):m/z=559.5[M+H]+.
第七步:以化合物100G(0.12g,0.21mmol)为原料,参考实施例13第三步操作得到目标化合物100(7mg,收率6.42%)。
1H NMR(400MHz,Methanol-d4)δ9.04-8.82(m,1H),8.30-7.79(m,4H),7.75-7.47(m,2H),5.58-5.15(m,1H),4.47-4.19(m,2H).LC-MS(ESI):m/z=519.2[M+H]+.
实施例101:
第一步:-78℃,氮气氛围下,向LDA(2M,7.5mL,15mmol)中滴加101A(1.28g,10mmol)的四氢呋喃(25mL)溶液,滴加完毕,继续搅拌反应1小时。滴加3-氧杂环丁酮(1.44g,20mmol)的四氢呋喃(25mL)溶液,滴加完毕,继续搅拌反应1小时。TLC监测原料消失,停止反应。滴加饱和氯化铵水溶液(20mL)淬灭反应,加入EA (50mL)萃取,有机相用饱和食盐水(40mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩得到的粗品经柱层析(PE∶EA=2∶1)分离纯化得到化合物101B(1.25g,62.62%)。LC-MS(ESI):m/z=200.1[M+H]+.
第二步:室温下,将6A(150mg,0.51mmol)溶于干燥的二氧六环(10mL),依次加入101B(0.11g,0.55mmol),Pd2(dba)3(93mg,0.1mmol),Xantphos(93mg,0.15mmol),碳酸钾(210mg,1.53mmol),氮气氛围,100℃反应16小时。TLC监测原料消失,停止反应。待反应冷至室温,加入EA(20mL)稀释,有机相用水(20mL×2)洗,饱和食盐水(20mL×2)洗,无水硫酸钠干燥,过滤,浓缩,残余物经柱层析(PE∶EA=1∶1)分离纯化得到化合物101C(180mg,79.21%)。LC-MS(ESI):m/z=455.3[M+H]+.
第三步:室温下,将101C(180mg,0.4mmol)溶于DCM(10mL),氮气氛围,降温至-78℃,滴加DAST(77mg,0.48mmol),滴加完毕后,-78℃下反应两小时。TLC监测原料消失,停止反应。将反应液倒入饱和碳酸氢钠水溶液中淬灭,加入DCM(30mL),萃取,分液,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物经柱层析(PE∶EA=2∶1)分离纯化得到化合物101D(120mg,65.72%)。LC-MS(ESI):m/z=457.8[M+H]+.
第四步:室温下,将101D(120mg,0.26mmol)溶于甲醇(15mL),加入钯碳(40mg),氢气氛围,室温反应16小时。TLC监测原料消失,停止反应。将反应液过滤,滤液减压浓缩后,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物101E(90mg,94.49%)。LC-MS(ESI):m/z=367.1[M+H]+.
第五步:以101E(90mg,0.25mmol)和5-氯-2,4-二氟苯胺(61mg,0.37mmol)为原料,参考实施例8第三步操作得到101F(80mg,62.51%)。LC-MS(ESI):m/z=512.7[M+H]+.
第六步:以101F(80mg,0.16mmol)为原料,参考实施例8第四步操作得到产物101G(70mg,82.72%)。LC-MS(ESI):m/z=529.1[M+H]+.
第七步:以101G(70mg,0.13mmol)为原料,参考实施例8第五步操作得到化合物101(15mg,23.60%)。LC-MS(ESI):m/z=489.4[M+H]+.
1H NMR(400MHz,CD3OD)δ7.84-7.80(t,1H),7.48-7.46(d,1H),7.41-7.39(d,1H),7.35-7.31(m,1H),4.76-4.75(d,1H),4.73(s,1H),4.65(s,4H),4.56-4.55(d,1H),4.50-4.47(m,3H).
实施例102:
第一步:将102A(5g,24.25mmol)、甲基硼酸(1.59g,26.62mmol)、二氯[1,1′-二(二苯基膦)二茂铁]钯(1.77g,2.43mmol)及碳酸铯(15.80g,48.49mmol)溶于1,4-二氧六环(150mL)中,氮气氛围,于100℃反应过夜。待反应完全后,冷却至室温,向反应液中加入水(200mL),以乙酸乙酯(100mL×3)萃取,合并有机层,用饱和食盐水(150mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到102B(2.66g,59%)。LC-MS(ESI):m/z=186.1[M+H]+.
第二步:将102B(2.66g,14.33mmol)及丙烯酸甲酯(12.34g,143mmol)溶于DMF(50mL)中,随后依次加入Pd2(dba)3(1.31g,1.43mmol)、N,N-二环己基甲胺(5.60g,28.66mmol)及三叔丁基膦(1.16g,5.74mmol),氮气氛围,于100℃反应过夜。待反应完全后,冷却至室温,向反应液中加入水(100mL),以乙酸乙酯(40mL×3)萃取,合并有机层,用饱和食盐水(60mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=3∶1)得到102C(0.86g,25%)。LC-MS(ESI):m/z=236.1[M+H]+.
第三步:将102C(0.86g,3.66mmol)溶于无水乙醇(10mL)中,加入10%钯碳(100mg),氢气氛围下,于60℃反应过夜。待反应完全后,冷却至室温,过滤除去不溶物,以乙酸乙酯洗滤饼,合并滤液,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到102D(0.7g,81%)。LC-MS(ESI):m/z=238.1[M+H]+.
第四步:将102D(0.7g,2.95mmol)溶于四氢呋喃(20mL)及甲醇(1mL)的混合溶剂中,氮气氛围下,分批加入60%氢化钠(474mg,11.84mmol),随后升温至60℃反应4h。冷却至室温,将反应液浓缩后得到的残留物102E粗品(0.97g),未经进一步纯化,直接用于下一步反应。
第五步:将上一步得到的102E粗品置于圆底烧瓶中,冰浴下缓慢加入浓盐酸(10mL),随后升温至115℃反应1h。冷却至室温,冰浴下缓慢加入饱和碳酸氢钠水溶液(150mL),以乙酸乙酯(30mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=3∶1)得到102F(0.22g,两步收率51%)。LC-MS(ESI):m/z=148.1[M+H]+.
第六步:将102F(0.22g,1.65mmol)溶于二氯甲烷(10mL)中,冰浴下滴加二乙胺基三氟化硫(0.96g,5.92mmol),氮气氛围下,于45℃反应过夜。待反应完全后,冷 却至室温,缓慢加入饱和碳酸氢钠水溶液(20mL),以二氯甲烷(15mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到102G(178mg,70%)。LC-MS(ESI):m/z=170.1[M+H]+.
第七步:将102G(178mg,1.05mmol)溶于二氯甲烷(10mL),冰浴下加入间氯过氧苯甲酸(0.36g,2.1mmol),室温反应过夜。待反应完全后,缓慢加入饱和碳酸氢钠水溶液(15mL),以二氯甲烷(10mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到102H(144mg,74%)。
第八步:氮气氛围下,向102H(0.26g,0.63mmol)缓慢加入三氯氧磷(4mL),于100℃反应3h。冷却至室温,减压浓缩,向残留物中缓慢加入饱和碳酸氢钠水溶液(15mL),以乙酸乙酯(10mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到102I(80mg,51%)。LC-MS(ESI):m/z=204.1[M+H]+.
第九步:以1A(0.14g,0.39mmol)及102I(80mg,0.39mmol)为原料,参考实施例1第一步操作得到102J(98mg,46%)。LC-MS(ESI):m/z=531.1[M+H]+.
第十步:以102J(98mg,0.19mmol)为原料,参考实施例13第三步操作得到化合物102(35mg,39%)。
1H NMR(400MHz,DMSO-d6)δ8.45(s,1H),7.98-7.94(m,1H),7.67-7.61(m,1H),5.97-5.94(m,1H),5.70-5.67(m,1H),5.15(s,1H),2.97-2.82(m,2H),2.50(s,3H),2.15-2.07(m,2H).LC-MS(ESI):m/z=491.1[M+H]+.
实施例103:
第一步:将103A(1.8g,11.04mmol)溶于浓硫酸(20mL)中,冰浴下缓慢滴加硝酸(3mL),于室温反应过夜。待反应完全后,将反应液缓慢加入冰水(50mL)中,用饱和碳酸氢钠水溶液调节pH至8左右,以乙酸乙酯(50mL×3)萃取,合并有机层,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到103B(0.87g,38%)。LC-MS(ESI):m/z=209.2[M+H]+.
第二步:将103B(0.87g,4.18mmol)溶于甲醇(20mL)中,加入10%钯碳(200mg),氢气氛围下,于60℃反应过夜。待反应完全后,冷却至室温,过滤除去不溶物,以乙酸乙酯洗滤饼,合并滤液,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到103C(0.61g,82%)。LC-MS(ESI):m/z=179.1[M+H]+.
第三步:将103C(0.61g,3.43mmol)溶于原甲酸三乙酯(15mL),随后加入对甲苯磺酸(60mg,0.35mmol),氮气氛围,150℃反应24h。冷却至室温,向反应液中加入50mL水,乙酸乙酯(25mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到103D(84mg,13%)。LC-MS(ESI):m/z=189.2[M+H]+.
第四步:将103D(84mg,0.45mmol)溶于二氯甲烷(10mL),冰浴下加入间氯过氧苯甲酸(0.12g,0.9mmol),室温反应过夜。待反应完全后,缓慢加入饱和碳酸氢钠水溶液(10mL),以二氯甲烷(10mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到103E(56mg,61%)。
第五步:氮气氛围下,向103E(56mg,0.27mmol)缓慢加入三氯氧磷(3mL),于100℃反应2h。冷却至室温,浓缩除去部分溶剂,向残留物缓慢加入饱和碳酸氢钠水溶液淬灭反应,以乙酸乙酯(10mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到103F(35mg,57%)。LC-MS(ESI):m/z=223.0[M+H]+.
第六步:以1A(57mg,0.16mmol)及103F(35mg,0.16mmol)为原料,参考实施例1第一步操作得到103G(60mg,69%)。LC-MS(ESI):m/z=550.1[M+H]+.
第七步:以103G(60mg,0.11mmol)为原料,参考实施例13第三步操作得到化合物103(24mg,43%)。
1H NMR(400MHz,CDCl3)δ8.82(s,1H),8.01-7.93(m,2H),7.24-7.19(m,1H),5.15(s,1H),4.80-4.77(m,1H),4.46-4.43(m,1H).LC-MS(ESI):m/z=510.1[M+H]+.
实施例104:(2S,3S,4S)-N-(5-氯-4-(2,2-二氟环丙基)-2-氟苯基)-3,4-二羟基-N-(甲基-d3)-5-氧代-1-(7-(三氟甲基)呋喃[3,2-b]吡啶-5-基)吡咯烷-2-甲酰胺(化合物104)(2S,3S,4S)-N-(5-chloro-4-(2,2-difluorocyclopropyl)-2-fluorophenyl)-3,4-dihydroxy-N-(methyl-d3)-5-oxo-1-(7-(trifluoromethyl)furo[3,2-b]pyridin-5-yl)pyrrolidine-2-carboxamide
第一步:将98B(200mg,0.52mmol)及54E(115mg,0.52mmol)溶于吡啶(5mL)中,室温缓慢滴加50%T3P的乙酸乙酯溶液(2.6mmol),然后升温至50℃反应过夜。待反应冷至室温,向反应液中加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和柠檬酸溶液(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶 柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到104A(195mg,64%)。LC-MS(ESI):m/z=590.2[M+H]+.
第二步:将104A(195mg,0.34mmol)溶于四氢呋喃(10mL),冰浴下加入60%氢化钠(20mg,0.50mmol),随后滴加氘代碘甲烷(0.1g,0.68mmol),继续搅拌1h,随后缓慢升温至室温反应1h。加入1M氯化氢溶液淬灭反应,加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到104B(170mg,85%)。LC-MS(ESI):m/z=607.6[M+H]+.
第三步:将104B(170mg,0.28mmol)溶于4M氯化氢的1,4-二氧六环溶液(5mL)中,室温反应过夜。待反应完全后,减压浓缩,向残留物加入15mL水,以乙酸乙酯(10mL×4)萃取,合并有机层,用无水硫酸钠干燥,浓缩后残留物经硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)后,进一步经过高效液相制备进行纯化分离得到化合物104,异构体1(35mg,22%,保留时间:21.420min)和化合物104,异构体2(18mg,11%,保留时间:22.175min)。
高效液相制备分离条件:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含5mmol/L乙酸铵),梯度洗脱,流动相A含量从30%-80%,流量12mL/min。洗脱时间25min。)
化合物104,异构体1:1H NMR(400MHz,CDCl3)δ8.68(s,1H),7.94-7.92(m,1H),7.85-7.82(m,1H),7.21-7.17(m,1H),7.04-7.00(m,1H),5.01(s,1H),4.76-4.73(m,1H),4.41-4.38(m,1H),2.88-2.83(m,1H),2.00-1.89(m,1H),1.64-1.57(m,1H).LC-MS(ESI):m/z=567.6[M+H]+.
化合物104,异构体2:1H NMR(400MHz,CDCl3)δ8.69(s,1H),7.95-7.93(m,1H),7.87-7.84(m,1H),7.14-7.11(m,1H),7.04-7.02(m,1H),5.01(s,1H),4.75-4.73(m,1H),4.41-4.37(m,1H),2.93-2.83(m,1H),1.98-1.89(m,1H),1.68-1.61(m,1H).LC-MS(ESI):m/z=567.6[M+H]+.
实施例105:
第一步:将化合物91K(150mg,0.25mmol)溶于l,4-二氧六环(8mL),滴入盐酸l,4-二氧六环溶液(4M,8mL)。室温搅拌至原料消失,将反应液浓缩得到化合物105A(115mg,92%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=512.1[M+H]+.
第二步:将化合物105A(115mg,0.23mmol)溶于DMF(5mL),冰浴下,加入氢化钠(9mg,0.46mmol),搅拌10分钟,加入溴甲基环丙烷(62mg,0.46mmol),室温反应30分钟。倒入冰水(20mL)淬灭,EA(30mL×2)萃取,合并后的有机相用无水硫酸钠干燥,过滤,浓缩得到的粗品经硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=6∶1)得到标题化合物105B(110mg,85%)。LC-MS(ESI):m/z=566.3[M+H]+.
第三步:以化合物105B(110mg,0.19mmol)为原料,参考实施例34第一步操作得到标题化合物105C(80mg,70%)。LC-MS(ESI):m/z=584.5[M+H]+.
第四步:将化合物105C(80mg,0.14mmol)溶于甲醇(8mL),加入三乙胺(110mg,0.8mmol),室温搅拌过夜。待反应完毕浓缩除去大部分溶剂,加入20mL乙酸乙酯,有机相用饱和食盐水多次洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到棕色粗品,使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物105(35mg,收率50%)。
1H NMR(400MHz,Methanol-d4)δ8.33-8.31(m,1H),8.07-7.34(m,2H),7.07-7.04(m,1H),5.21-4.88(m,1H),4.13-4.11(m,1H),3.87-3.41(m,2H),3.24-2.98(m,2H),2.55-2.53(m,3H),1.16-0.82(m,1H),0.65-0.38(m,2H),0.30-0.20(m,2H).LC-MS(ESI):m/z=512.4[M+H]+.
实施例106:
以98B和106A为原料,参考实施例8实验操作(第三、四、五步),得到化合物106。
1H NMR(400MHz,CD3OD)δ8.62(s,1H),8.29-8.26(m,1H),7.93-7.88(m,1H),7.69(s,1H),7.53-7.47(m,1H),7.26-7.22(m,1H),5.39-5.21(m,1H),4.30-4.25(m,2H).LC-MS(ESI):m/z=491.1[M+H]+.
实施例107:
第一步:将化合物107A(3.00g,16.03mmol)溶于二氯甲烷(60mL),0℃下,加入三乙胺(3.24g,32.06mmol),搅拌10分钟后,滴加甲基磺酰氯(2.75g,24.05mmol),滴加完毕自然至室温反应2小时。反应结束,加水(50mL),搅拌10分钟,萃取分液,水相用乙酸乙酯(50mL×4)萃取,合并后的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩至出现大量固体,向残余物中加入石油醚(10mL),搅拌10分钟,过滤,干燥滤饼后得到化合物107B(2.80g,收率65.84%)。LC-MS(ESI):m/z=266.1[M+H]+.
第二步:将化合物107B(2.8g,10.55mmol)溶于乙腈(40mL),依次加入三甲基氰硅烷(1.26g,12.66mmol),TBAF(3.31g,12.66mmol),滴加完毕,升温至回流反应过夜。反应结束,待反应冷至室温,减压浓缩,得到的粗品使用Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%MeOH/DCM)纯化得到化合物107C(1.40g,收率67.84%)。LC-MS(ESI):m/z=197.1[M+H]+.
第三步:依次将化合物107C(1.4g,7.14mmol),2-氯-6-甲基-4-(三氟甲基)吡啶(2.09g,10.71mmol)溶于干燥二氧六环(20mL),向其中依次加入Pd2(dba)3(0.65g,0.71mmol),XantPhos(1.24g,2.14mmol),碳酸钾(1.97g,14.25mmol),氮气氛围下,升温至100℃反应16小时。反应结束后,冷却至室温,加入乙酸乙酯(50mL)稀释,过滤,滤液减压浓缩后得到粗品,粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物107D(1.39g,收率55.19%)。LC-MS(ESI):m/z=356.30[M+H]+.
第四步:将化合物107D(1.4g,3.94mmol)溶于氯化氢甲醇溶液(4M,30mL),室温搅拌2天。反应结束后,减压浓缩,然后向粗品中加入饱和碳酸氢钠溶液(30mL),乙酸乙酯(30mL×2)萃取,合并后的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到化合物107E粗品(1.5g),粗品直接用于下一步反应。LC-MS(ESI):m/z=349.1[M+H]+.
第五步:将化合物107E(1.5g,4.31mmol)加入甲苯(30mL)中,依次加入2,2-二甲氧基丙烷(4.49g,43.10mmol),对甲苯磺酸吡啶盐(0.65g,2.59mmol),升温至回流反应过夜。待反应冷却至室温,加入乙酸乙酯(30mL)稀释,加入水(30mL)萃取,有 机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩,得到的粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物107F(0.97g,收率57.95%)。LC-MS(ESI):m/z=389.1[M+H]+.
第六步:将化合物107F(0.97g,2.50mmol)溶于甲醇(10mL),依次加入水(5mL),一水合氢氧化锂(0.21g,5mmol),然后搅拌过夜。反应完毕,加入1M稀盐酸调节pH至3-4,乙酸乙酯(20mL×3)萃取,合并后的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩,得到的粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物107G(0.45g,收率48.09%)。LC-MS(ESI):m/z=375.2[M+H]+.
第七步:依次将化合物107G(0.45g,1.20mmol),5-氯-2,4-二氟苯胺(0.29g,1.78mmol)溶于吡啶(10mL),滴加T3P(1.91g,50%in EA,5.99mmol),滴加完毕后搅拌30分钟,然后升温至50℃搅拌过夜。反应完毕,减压浓缩除去大部分吡啶,然后加入乙酸乙酯(30mL)稀释,有机相依次用硫酸氢钾水溶液(1M,20mL)洗涤,饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,浓缩,得到的粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物107H(0.40g,收率64.12%)。LC-MS(ESI):m/z=520.1[M+H]+.
第八步:将化合物107H(0.20g,0.38mmol)溶于DMF(5mL),氮气氛围,0℃下,加入氢化钠(60%)(22.0mg,0.57mmol),搅拌5分钟后,滴加氘代碘甲烷(0.11g,0.76mmol),滴加完毕后继续反应30分钟。加入冰的稀盐酸(1M,15mL)淬灭,乙酸乙酯(20mL)萃取,有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩,得到的粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物107I(0.17g,收率83.33%)。LC-MS(ESI):m/z=537.2[M+H]+.第九步:将化合物107I(170.0mg,0.32mmol)溶于二氯甲烷(10mL),氮气氛围下,降温至-40℃,滴加三氯化硼(1M in DCM,0.19mmol,0.19mL),滴加完毕,继续搅拌30分钟,然后自然升至室温反应2小时。将反应液滴入饱和碳酸氢钠水溶液(20mL)中,搅拌10分钟,分液,水相用二氯甲烷萃取(20mL),合并后的有机相,用无水硫酸钠干燥,过滤,浓缩,得到的粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物107(34.0mg,收率21.39%)。
1H NMR(400MHz,CD3OD)δ8.49-8.09(m,lH),7.67-7.24(m,3H),5.05-4.93(m,lH),4.54-4.40(m,2H),3.03-2.87(m,lH),2.62-2.50(m,4H).LC-MS(ESI):m/z=497.5[M+H]+.
实施例108:
第一步:将化合物108A(10g,39.46mmol)溶于DMF(100mL),加入碳酸钾(10.91g,78.94mmol),0℃下缓慢滴加碘甲烷(10.91g,39.46mmol),滴加完毕,升至室温继续搅拌反应2小时。将反应液倒入水中,用乙酸乙酯(200mL×3)萃取,合并后的有机相,用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用柱层析分离纯化(EA/PE=1/50)得到化合物108B(10.5g,收率99.48%)。
1H NMR(400MHz,CD3Cl)δ7.70-7.68(m,1H),7.64-7.62(m,2H),3.94(s,3H).
第二步:室温下,将108B(7.7g,28.79mmol)溶于干燥的甲苯(100mL),向其中依次加入氨基甲酸叔丁酯(4.05g,34.55mmol),Pd2(dba)3(2.64g,2.88mmol),BINAP(3.59g,5.76mmol),Cs2CO3(18.76g,57.58mmol),搅拌均匀后,氮气氛围,100℃下反应18小时。TLC监测原料消失,停止反应。冷却至室温,垫硅藻土过滤,乙酸乙酯(100mL)洗涤滤饼,滤液用水(200mL×2)洗涤,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,浓缩,得到的粗品用中压制备仪Biotage Isolera One(80g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物108C(8.0g,收率91.5%)。LC-MS(ESI):m/z=304.1[M+H]+.
第三步:将108C(8.0g,1.03mmol)溶于二氯甲烷(20mL),缓慢加入三氟乙酸(10mL),搅拌均匀,室温下搅拌反应2小时。减压浓缩除去大部分溶剂,残余物用饱和碳酸氢钠水溶液调节pH至8-10,二氯甲烷(100mL×3)萃取,合并后的有机相用无水硫酸钠干燥,过滤,浓缩,得到的粗品用乙酸乙酯/石油醚(V1/V2=1/10)重结晶得到108D(4.0g,74.6%)。
1H NMR(400MHz,DMSO-d6)δ7.55-7.52(d,1H),6.83-6.81(d,1H),6.27(s,2H),3.75(s,3H).
第四步:依次将108D(1.27g,6.24mmol),2,5-己二酮(3.56g,31.2mmol),对甲苯磺酸(0.21g,1.25mmol)溶于甲苯(20mL),升温至100℃下搅拌反应3小时。待反应冷却至室温,将反应液倒入水中,用乙酸乙酯(100mL×2)萃取,合并后的有机相用水(100mL×2)洗,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(10g硅胶柱,洗脱剂:0-2%EA/PE)纯化得到化合物108E(1.3g,收率74%)。LC-MS(ESI):m/z=282.3[M+H]+.
第五步:将108E(0.8g,2.84mmol)溶于干燥的四氢呋喃(25mL),加入钛酸四异丙酯(2.42g,8.51mmol),氮气氛围,0℃下滴加乙基溴化镁(2.0M in THF,2.15mL,4.29 mmol),滴加完毕,自然升至室温,搅拌反应24小时。加入饱和氯化铵水溶液淬灭反应,将反应液倒入水中,用乙酸乙酯(50mL×2)萃取,合并后的有机相依次用水(50mL),饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(10g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物108F(0.15g,收率18.88%)。
1H NMR(400MHz,CDCl3)δ7.31-7.25(m,1H),5.92-5.91(m,2H),2.02(s,6H),1.31-1.26(m,2H),1.07-1.04(m,2H).
第六步:依次将108F(0.85g,3.04mmol),三乙胺(0.46g,4.56mmol)溶于二氯甲烷(25mL),0℃下滴加三甲基氯硅烷(0.99g,9.12mmol),滴加完毕,自然升至室温,搅拌反应2小时。将反应液倒入饱和碳酸氢钠水溶液(50mL),乙酸乙酯(50mL×2)萃取,合并后的有机相依次用水(50mL)洗,饱和食盐水(100mL)洗,无水硫酸钠干燥,过滤,减压浓缩得到化合物108G(0.9g,收率84.12%),无需进一步纯化可直接用于下一步反应。
第七步:将108G(0.4g,1.14mmol)溶于二氯甲烷(12mL),0℃下滴加二乙胺基三氟化硫(0.22g,1.37mmol),滴加完毕,搅拌反应30分钟。将反应液倒入饱和碳酸氢钠水溶液(30mL)中,乙酸乙酯(30mL×2)萃取,合并后的有机相用水(30mL),饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩,得到化合物108H(0.36g,收率99.99%),无需进一步纯化可直接用于下一步反应。LC-MS(ESI):m/z=282.2[M+H]+.第八步:依次将108H(0.4g,1.42mmol),盐酸羟胺(1.97g,28.34mmol),三乙胺(0.72g,7.07mmol)溶于乙醇(9mL)和水(3mL)的混合溶剂,升温至100℃搅拌反应18小时。反应完全后,冷至室温,将反应液倒入水(30mL)中,乙酸乙酯(30mL×2)萃取,合并后的有机相用水(30mL)洗,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩得到化合物108I(0.3g,收率99.99%),无需进一步纯化可直接用于下一步反应。
第九步:以化合物6C(0.1g,0.28mmol)和108I(0.1g,0.49mmol)为原料,参考实施例8第三步操作得到化合物108J(70mg,收率45.8%)。LC-MS(ESI):m/z=546.2[M+H]+.
第十步:以化合物108J(70mg,0.13mmol)为原料,参考实施例8第四步操作得到化合物108K(70mg,收率95.6%)。LC-MS(ESI):m/z=563.2[M+H]+.
第十一步:以化合物108K(70mg,0.27mmol)为原料,参考实施例13第三步操作得到目标化合物108(30mg,收率47.8%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-70%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,CD3OD)δ8.57-8.52(m,1H),8.09-7.99(m,1H),7.47-7.22(m,2H),5.74-5.53(m,1H),5.43-5.34(m,1H),5.23-5.17(m,1H),5.11-4.99(m,1H),4.58-4.29(m,3H),2.70-2.25(m,3H).LC-MS(ESI):m/z=523.2[M+H]+.
实施例109:
第一步:室温下,将91A(10.0g,37.85mmol)溶于DMF(50mL),向其中加入碳酸钾(26.16g,189.25mmol),搅拌十分钟后,加入碘甲烷(26.86g,189.25mmol),加入完毕,室温下反应16小时。TLC监测原料消失,停止反应。向反应液中加入EA(100mL),有机相用水(100mL×2)洗涤,饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物经柱层析(PE∶EA=1∶1)分离纯化得到化合物109A(7.0g,63.27%)。LC-MS(ESI):m/z=293.3[M+H]+.
第二步:室温下,将109A(3.0g,10.26mmol)溶于甲醇(20mL),加入钯碳(1.1g,1.03mmol),氢气氛围,室温下反应3小时。TLC监测原料消失,停止反应。过滤,滤液浓缩后得到化合物109B(1.4g,86.27%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=159.2[M+H]+.
第三步:室温下,将109B(700mg,4.43mmol)溶于干燥的二氧六环(30mL),向反应中依次加入79C(1.08g,4.87mmol),Pd2(dba)3(810mg,0.89mmol),Xantphos(770mg,1.33mmol),碳酸钾(1.84g,13.31mmol),氮气氛围下,100℃反应16小时。TLC监测原料消失,停止反应。待反应冷至室温,向反应液中加入EA(50mL),有机相依次用水(50mL×2)洗涤,饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到化合物109C(750mg,49.32%)。LC-MS(ESI):m/z=344.1[M+H]+.
第四步:室温下,将109C(750mg,2.18mmol)溶于甲醇(5mL),依次加入水(5mL),氢氧化锂(100mg,4.36mmol),室温反应20分钟。TLC监测原料消失,停止反应。滴加1M的盐酸调节pH为弱酸性,加入水(20mL),DCM(50mL),萃取分液,有机相用饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物109D(660mg,91.96%)。LC-MS(ESI):m/z=330.1[M+H]+.
第五步:以109D(120mg,0.36mmol)和5-氯-2,4-二氟苯胺(65mg,0.40mmol)为原料,参考实施例8第三步操作得到化合物109E(140mg,81.91%)。LC-MS(ESI):m/z=475.4[M+H]+.
第六步:以109E(140mg,0.29mmol)为原料,参考实施例8第四步操作得到化合物109(70mg,49.98%)。
1H NMR(400MHz,CD3OD)δ8.61-8.55(m,1H),8.21-8.14(m,1H),7.77-7.73(m,1H),7.57-7.47(m,1H),7.20-6.99(m,1H),5.07-4.98(m,1H),3.63-3.61(m,2H),2.92-2.91(m,3H).LC-MS(ESI):m/z=492.1[M+H]+.
实施例110:
第一步:将21D(2g,9.02mmol)溶于二氯甲烷(20mL),降温至0℃,分批加入间氯过氧苯甲酸(5.45g,31.57mmol),然后升至室温反应过夜。将反应液加入饱和碳酸氢钠溶液中,用乙酸乙酯萃取,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到目标化合物110A(2.04g,收率95.18%)。LC-MS(ESI):m/z=238.1[M+H]+.
第二步:将110A(2.04g,8.59mmol)溶于乙酸酐(20mL),升温至100℃反应4小时。冷却至室温,减压浓缩除去大部分溶剂,将残余物滴加到冰的饱和碳酸氢钠水溶液中,用二氯甲烷萃取两次,合并后的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到目标化合物110B(1.6g,收率66.61%)。LC-MS(ESI):m/z=280.1[M+H]+.
第三步:将化合物110B(1.6g,5.72mmol)溶于甲醇(20mL)和水(5mL),加入无水碳酸钾(2.77g,20.04mmol),室温条件下搅拌反应4小时。将反应液倒入水中,用二氯甲烷萃取两次,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩,得到的粗品经中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到目标化物110C(1.2g,收率88.29%)。LC-MS(ESI):m/z=238.1[M+H]+
第四步:将110C(1.2g,5.05mmol)溶于二氯甲烷(20mL),降温至0℃,分批加入戴斯-马丁试剂(3.21g,7.57mmol),然后升至室温反应5小时。将反应液缓慢倒入饱和碳酸氢钠水溶液中,用二氯甲烷萃取两次,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩,得到的粗品经中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到目标化合物110D(0.75g,63.04%)。LC-MS(ESI):m/z=236.1[M+H]+.
第五步:将110D(0.75g,3.18mmol)溶于二氯甲烷(10mL),降温至0℃,加入DAST(2.56g,15.87mmol),然后升至室温反应4小时。将反应液缓慢倒入饱和碳酸氢钠水溶液中,用二氯甲烷萃取两次,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩, 得到的粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到目标化合物110E(0.36g,收率43.95%)。
1H NMR(400MHz,Chloroform-d)δ7.63(s,1H),3.27-3.06(m,2H),2.82-2.54(m,2H).
第六步:依次将化合物110E(0.36g,1.4mmol),6A(0.41g,1.4mmol)溶于二氧六环(10mL),依次加入Pd2(dba)3(0.13g,0.14mmol),XantPhos(0.24g,0.42mmol),碳酸钾(0.25g,4.2mmol),氮气氛围,升温至95℃搅拌反应18小时。待反应完全后,冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品经中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到目标化合物110F(0.32g,收率44.61%)。LC-MS(ESI):m/z=513.4[M+H]+
第七步:将化合物110F(0.32g,0.62mmol)溶于甲醇(10mL),加入10%钯-碳(0.04g),氢气氛围,室温下反应2小时。反应结束后,垫硅藻土过滤,减压浓缩滤液,得到目标化合物110G(0.25g,95.48%)。无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=423.5[M+H]+.
第八步:以化合物110G(0.25g,0.59mmol)和5-氯-2,4-二氟苯胺(0.14g,0.89mmol)为原料,参考实施例8第三步操作得到目标化合物110H(0.21g,收率62.68%)。LC-MS(ESI):m/z=568.5[M+H]+.
第九步:以化合物110H(0.21g,0.37mmol)为原料,参考实施例8第四步操作得到目标化合物110I(0.17g,收率78.55%)。LC-MS(ESI):m/z=585.8[M+H]+.
第十步:以化合物110I(50mg,0.08mmol)为原料,参考实施例6第六步操作得到目标化合物110(20mg,收率43.19%)。
1H NMR(400MHz,Methanol-d4)δ8.87-8.66(m,1H),8.35-7.99(m,1H),7.57-7.38(m,1H),5.21-5.01(m,1H),4.52-4.19(m,2H),3.29-3.12(m,2H),2.87-2.62(m,2H).LC-MS(ESI):m/z=545.0[M+H]+.
实施例111:
第一步:将111A(0.5g,3.70mmol)溶于甲苯(20mL)中,依次加入2,5-己二酮(2.11g,18.5mmol),对甲苯磺酸(0.13g,0.75mmol),升温至100℃下搅拌反应3小时。冷却至室温,将反应液倒入水中,用乙酸乙酯萃取(50mL×2),有机相依次用水(50mL×2),饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(10g硅胶柱,洗脱剂:0-2%EA/PE)纯化得到化合物111B(0.78g,收率99%)。LC-MS(ESI):m/z=212.2[M+H]+.
第二步:将化合物111B(0.4g,1.89mmol)溶于DMF(10mL),冷却至0℃,分批加入氢化钠(60%)(0.054g,2.25mmol),搅拌15分钟,滴加氘代碘甲烷(0.4g,2.82mmol),滴加完毕后,升温至室温搅拌反应1小时。将反应液逐滴滴加到稀盐酸中,用乙酸乙酯萃取(50mL×2),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物111C(0.29g,收率68.11%)。LC-MS(ESI):m/z=226.1[M+H]+.
第三步:将111C(0.29g,1.29mmol)溶于乙醇(9mL)和水(3mL),依次加入盐酸羟胺(1.79g,25.8mmol),三乙胺(0.65g,6.45mmol),升温至100℃搅拌反应18小时。待反应冷至室温,将反应液倒入水中,乙酸乙酯萃取(30mL×2),合并后的有机相依次用水(30mL)洗,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩得到化合物111D(0.2g,收率99.99%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=148.1[M+H]+.
第四步:以化合物98B(0.1g,0.26mmol)和化合物111D(0.093g,0.63mmol)为原料,参考实施例8第三步操作,得到化合物111E(0.07g,收率52.23%)。LC-MS(ESI):m/z=516.2[M+H]+.
第四步:以化合物111E(0.07g,0.14mmol)为原料,参考实施例8第四步操作得到化合物111F(0.07g,收率93.9%)。LC-MS(ESI):m/z=533.6[M+H]+.
第五步:以化合物111F(70mg,0.13mmol)为原料,参考实施例8第五步操作得到目标化合物111(23mg,收率35.93%)。
1H NMR(400MHz,DMSO-d6)δ7.91(s,1H),7.40-7.36(m,2H),6.62-6.61(m,2H),6.29-6.28(m,1H),5.74-5.73(m,1H),4.22(s,1H),3.31(s,1H),3.91-3.90(m,2H),3.13(s,3H).LC-MS(ESI):m/z=493.2[M+H]+.
实施例112:
第一步:将111B(0.3g,1.42mmol)溶于二氯甲烷(20mL),依次加入四丁基溴化铵(0.046g,0.14mmol),对甲苯磺酰氯(4.06g,21.3mmol),室温下反应18小时。将反应液倒入水中,用乙酸乙酯萃取(50mL×2),合并后的有机相依次用水(50mL×2)洗,饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(10g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物112A(0.16g,收率30.8%)。
1H NMR(400MHz,CDCl3)δ8.05-8.03(m,2H),7.93-7.91(m,1H),7.80-7.79(m,1H),7.26-7.24(m,2H),7.08-7.06(m,1H),5.91(s,2H),2.51-2.37(m,3H),2.06-2.01(m,6H).
第二步:将化合物112A(0.16g,0.44mmol)溶于乙醇(9mL)和水(3mL),依次加入盐酸羟胺(0.61g,8.8mmol),三乙胺(0.22g,2.2mmol),升温至100℃反应18小时。待反应冷至室温,将反应液倒入水中,用乙酸乙酯萃取(30mL×2),合并后的有机相依次用水(30mL)洗,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩得到化合物112B(0.12g,收率94.9%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=288.3[M+H]+.
第三步:依次将化合物98B(0.1g,0.25mmol),化合物112B(0.1g,0.49mmol)溶于吡啶(10mL),搅拌15分钟后,将反应液冷却至0℃,滴加1-丙基磷酸酐(50%乙酸乙酯溶液)(1.2g,2.0mmol),搅拌30分钟后,升温至50℃反应18小时。待反应完全后,冷却至室温,反应液倒入冰水中,乙酸乙酯萃取(50mL×2),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到化合物112C(0.13g,收率76.6%)。LC-MS(ESI):m/z=654.1[M-H]-.
第四步:将化合物112C(0.13g,0.2mmol)溶于DMF(10mL),冷却至0℃,加入氢化钠(60%)(0.024g,0.6mmol),搅拌15分钟,滴加氘代碘甲烷(0.087g,0.6mmol),滴加完毕后,升至室温搅拌反应1小时。将反应液逐滴滴加到稀盐酸中,用乙酸乙酯萃取(50mL×3),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物112D(0.14g,收率99%),无需进一步纯化直接用于下一步反应。LC-MS(ESI):m/z=673.2[M+H]+.
第五步:将112D(0.14g,0.21mmol)溶于四氢呋喃(10mL),加入四丁基氟化铵(0.27g,1.05mmol),室温搅拌反应18小时。将反应液倒入水中,用乙酸乙酯萃取(30mL×2),合并后的有机相依次用水(30mL)洗,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到化合物112E(80mg,收率73.5%)。LC-MS(ESI):m/z=519.0[M+H]+.
第六步:将化合物112E(80mg,0.15mmol)溶于二氯甲烷(8mL),冷却至-20℃,滴加1.0M三氯化硼二氯甲烷溶液(1.50mL,1.50mmol),滴加完毕后,自然升至升温搅拌反应2小时。将反应液滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯(30mL×2)萃取,合并后的有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,得到的粗品用高效液相制备纯化得到目标化合物112(12mg,收率16.72%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMSO溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-70%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,CD3OD)δ8.75(s,1H),8.24-8.11(m,2H),7.50-7.49(m,2H),7.18-7.17(m,1H),6.59-6.58(m,1H),5.11(s,1H),4.74-4.69(m,2H).LC-MS(ESI):m/z=479.1[M+H]+.
实施例113:
以98B和113A为原料,参考实施例8实验操作(第三、四、五步),得到化合物113。
LC-MS(ESI):m/z=509.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.60-8.52(m,1H),8.27-8.22(m,1H),7.99-7.69(m,1H),7.40-7.30(m,1H),7.26-7.05(m,1H),5.28-5.11(m,1H),4.38-4.19(m,2H).
实施例114:
以化合物98B和114A为原料,参考实施例8实验操作(第三、四、五步),得到化合物114。LC-MS(ESI):m/z=471.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.57(s,1H),8.25-8.23(m,1H),7.61-7.48(m,2H),7.27-7.22(m,1H),7.21-7.19(m,1H),5.21-5.18(m,1H),4.27-4.19(m,2H),2.39-2.35(m,3H).
实施例115:
以化合物98D为原料,参考实施例98操作(第五,六步),得到化合物115。
1H NMR(400MHz,CD3OD)δ8.81-8.75(m,1H),8.29-8.22(m,1H),7.85-7.54(m,1H),7.28-7.09(m,1H),6.74-6.62(m,1H),5.48-5.32(m,1H),5.05-4.98(m,1H),4.64-4.62(m,1H),4.58-4.44(m,3H),4.33-4.16(m,2H).LC-MS(ESI):m/z=564.1[M+H]+.
实施例116:
以化合物109D和54E为原料,参考实施例8实验操作(第三、四步),得到化合物116。
1H NMR(400MHz,CD3OD)δ8.61-8.54(t,1H),8.21-8.09(m,1H),7.67-7.62(m,1H),7.50-7.39(m,1H),7.20-6.99(m,1H),5.07-4.96(m,1H),3.65-3.63(d,2H),3.12-3.03(m,1H),2.93-2.90(m,3H),2.09-1.89(m,2H).LC-MS(ESI):m/z=550.2[M+H]+.
实施例117:(2S,3S,4S)-N-(3-氯-4-氟-5-(三氟甲基)苯基)-3,4-二羟基-N-(甲基-d3)-5-氧-1-(7-三氟甲基)呋喃并[3,2-b]吡啶-5-基)吡咯烷-2-甲酰胺(化合物117)
(2S,3S,4S)-N-(3-chloro-4-fluoro-5-(trifluoromethyl)phenyl)-3,4-dihydroxy-N-(methyl-d3)-5-oxo-1-(7-(trifluoromethyl)furo[3,2-b]pyridin-5-yl)pyrrolidine-2-carboxamide
第一步:依次将化合物98B(0.12g,0.31mmol),3-氯-4-氟-5-(三氟甲基)苯胺(0.066g,0.31mmol)溶于吡啶(10mL),搅拌15分钟。将反应液冷却至0℃,滴加1-丙基磷酸酐(50%乙酸乙酯溶液)(1.0g,1.57mmol),滴加完毕后,搅拌30分钟,然后升温至50℃搅拌反应18小时。待反应冷却至室温,倒入冰水,乙酸乙酯萃取(50mL×3),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物117A(0.16g,收率88.71%)。LC-MS(ESI):m/z=582.1[M+H]+.
第二步:将化合物117A(80mg,0.14mmol)溶于DMF(10mL)。0℃下,加入氢化钠(60%)(17mg,0.42mmol),搅拌15分钟,滴加氘代碘甲烷(61mg,0.42mmol),滴加完毕后,升温至室温搅拌反应1小时。将反应液滴加到稀盐酸中,用乙酸乙酯萃取(50mL×2),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物117B(80mg,收率95.42%),无需进一步纯化,直接用于下一步反应。LC-MS(ESI):m/z=599.0[M+H]+.
第三步:将117B(80mg,0.13mmol)溶于二氯甲烷(8mL),冷却至-20℃,滴加1.0M三氯化硼二氯甲烷溶液(130mL,1.30mmol),滴加完毕后,缓慢升温至室温搅拌反应2小时。将反应液滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯(30mL×2)萃取,合并后的有机相,依次用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物117(40mg,收率55.06%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-60%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,CD3OD)δ8.52(s,1H),8.17-8.16(m,1H),8.10(s,1H),7.99-7.98(m,1H),7.08(s,1H),5.11(s,1H),4.19-4.17(m,2H).LC-MS(ESI):m/z=559.0[M+H]+.
实施例118:
第一步:将化合物91A(3.0g,11.36mmol)溶于DMF(25mL),加入碳酸钾(3.14g,22.72mmol)后,缓慢滴加苄溴(2.40g,13.63mmo]),室温反应1.5h后,向反应液中加入50mL水,乙酸乙酯(20mL×3)萃取,合并后的有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,浓缩后得到粗品标题化合物118A(3.10g,77%)。LC-MS(ESI):m/z=355.1[M+H]+.
第二步:室温下,将化合物118A(3.10g,8.73mmol)溶于二碳酸二叔丁酯(6mL)中,加入DMAP(106mg,0.87mmol),升温至60℃下反应2小时。反应完全后,冷至室温,浓缩,残留物用硅胶柱色谱分离提纯(石油醚∶乙酸乙酯(v/v)=10∶1-5∶1)得到标题化合物118B(2.58g,65%)。LC-MS(ESI):m/z=455.3[M+H]+.
第三步:将化合物118B(2.58g,5.67mmol)溶于四氢呋喃(10mL)和甲醇(10mL)的混合溶剂,加入10%钯碳(300mg),氢气氛围下反应3h。反应完全后,过滤,乙酸乙酯洗涤滤饼,将滤液减压浓缩后得到目标化合物118C(865mg,66%),未经进一步纯化,直接用于下一步反应。LC-MS(ESI):m/z=231.5[M+H]+.
第四步:将化合物118C(865mg,3.75mmol)溶于DMF(25mL),加入碳酸钾(1.03g,7.49mmol),滴加苄溴(792mg,4.5mmol),室温反应1.5h。向反应液中加入50mL水,乙酸乙酯(20mL×3)萃取,合并后的有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后得到粗品标题化合物118D(250mg,21%)。LC-MS(ESI):m/z=321.4[M+H]+.
第五步:将化合物118D(250mg,0.78mmol)及化合物21D(206mg,0.93mmol)溶于1,4-二氧六环(15mL),随后依次加入Pd2(dba)3(85mg,0.093mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(107mg,0.186mmol)及碳酸铯(760mg,2.34mmol),氮气氛围,于95℃反应4h。待反应冷却至室温,向反应液中加入100mL水,以乙酸乙酯(30mL×3)萃取,合并后的有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物118E(276mg,70%)。LC-MS(ESI):m/z=506.4[M+H]+.
第六步:将化合物118E(276mg,0.55mmol)溶于四氢呋喃(10mL)及甲醇(10mL)的混合溶剂,加入10%钯碳(100mg),氢气氛围下反应3h。反应完毕后,过滤, 乙酸乙酯洗涤滤饼,滤液减压浓缩后得到目标化合物118F(171g,75%),未经进一步纯化,直接进行下一步反应。LC-MS(ESI):m/z=416.5[M+H]+.
第七步:将化合物118F(171mg,0.41mmol)及5-氯-2,4-二氟苯胺(74mg,0.45mmol)溶于吡啶(8mL),室温下滴加T3P(50%in EA,1.64mmol),滴加完毕后,升温至50℃反应过夜。反应完全后,加入50mL水,以乙酸乙酯(20mL×3)萃取,合并后的有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物118G(161mg,70%)。LC-MS(ESI):m/z=561.1[M+H]+.
第八步:将化合物118G(161mg,0.29mmol)溶于四氢呋喃(6mL),氮气氛围,冰浴下,分批加入60%氢化钠(34mg,0.86mmol),随后滴加氘代碘甲烷(126mg,0.86mmol),冰浴下反应1h,随后缓慢升至室温反应1h。将反应液倒入冰水中淬灭,加入15mL水,以乙酸乙酯(10mL×3)萃取,合并后的有机层,用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物118H(129mg,77%)。LC-MS(ESI):m/z=578.2[M+H]+.
第九步:将化合物118H(129mg,0.22mmol)溶于1,4-二氧六环(8mL),滴入盐酸1,4-二氧六环溶液(4M,8mL),室温搅拌至原料消失,浓缩,向残余物中加入20mL乙酸乙酯,有机相用饱和食盐水多次洗涤,无水硫酸钠干燥,过滤,浓缩得到的粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物118(58mg,收率55%)。
1H NMR(400MHz,Methanol-d4)δ8.27-8.21(m,1H),8.09-7.37(m,2H),5.00-4.91(m,1H),3.70-3.34(m,2H),3.25-2.81(m,4H),2.33-2.01(m,2H).LC-MS(ESI):m/z=478.4[M+H]+.
实施例119:
第一步:将化合物91D(500mg,2.05mmol)及化合物79C(498mg,2.25mmol)溶于1,4-二氧六环(15mL),随后加入Pd2(dba)3(210mg,0.23mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(265mg,0.46mmol)及碳酸钾(849mg,6.15mmol),氮气换气保护,于95℃反应4h。待反应冷却至室温,向反应液中加入100mL水,以乙酸乙酯(30mL×3)萃取,合并后的有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物119A(661mg,75%)。LC-MS(ESI):m/z=430.4[M+H]+.
第二步:将化合物119A(661mg,1.54mmol)溶于乙腈(10mL),依次加入溴化锂(1.34g,15.40mmol),三乙胺(777.7mg,7.70mmol)和水(31mg,1.69mmol),室温下反应6h。反应完毕后,加入饱和乙酸铵水溶液(5mL)和水(50mL),以二氯甲烷(20mL×3)萃取,合并后的有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=5∶1)得到目标化合物119B(515mg,80%)。LC-MS(ESI):m/z=416.5[M+H]+.
第三步:将化合物119B(150mg,0.36mmol)及5-氯-2,4-二氟苯胺(88mg,0.40mmol)溶于吡啶(8mL),室温下滴加T3P(50%in EA,3.6mmol),然后升至50℃反应过夜。反应完全后,向反应液中加入50mL水,以乙酸乙酯(20mL×3)萃取,合并后的有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物119C(110mg,55%)。LC-MS(ESI):m/z=561.1[M+H]+.
第四步:将化合物119C(110mg,0.20mmol)溶于N,N-二甲基甲酰胺(6mL)。氮气氛围,冰浴下,分批加入60%氢化钠(24mg,0.59mmol),随后滴加氘代碘甲烷(86mg,0.59mmol),冰浴下反应1h,随后缓慢升至室温反应1h。倒入冰水中淬灭反应,加入15mL水,以乙酸乙酯(10mL×3)萃取,合并后的有机层,用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物119D(82mg,71%)。LC-MS(ESI):m/z=578.2[M+H]+.
第五步:将化合物119D(82mg,0.14mmol)溶于1,4-二氧六环(8mL),滴入盐酸1,4-二氧六环溶液(4M,8mL),待反应完毕后,浓缩,向残余物中加入20mL乙酸乙酯,有机相分别用饱和碳酸氢钠水溶液和饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩后得到的粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物119(25mg,收率38%)。
1H NMR(400MHz,Methanol-d4)δ8.54-8.46(m,1H),8.28-7.65(m,2H),7.58-7.38(m,1H),7.23-6.90(m,1H),5.08-4.98(m,1H),3.75-3.33(m,2H).LC-MS(ESI):m/z=478.4[M+H]+.
实施例120:
第一步:将109B(0.8g,5.06mmol)及2-氯-6-甲基-4-(三氟甲基)吡啶(1.09g,5.57mmol)溶于1,4-二氧六环(30mL),随后加入Pd2(dba)3(466mg,0.50mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(578mg,1.01mmol)及碳酸钾(3.30g,10.12mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入50mL水,乙酸 乙酯(25mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到120A(1.33g,83%)。LC-MS(ESI):m/z=318.33[M+H]+.
第二步:将120A(1.33g,4.19mmol)溶于甲醇(6mL)及四氢呋喃(12mL)的混合溶剂中,加入氢氧化锂(301mg,12.57mmol)的水溶液(10mL),室温反应30min。滴加1M氯化氢水溶液调节pH至6左右,加入40mL水,以乙酸乙酯(25mL×3)萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到120B(1.12g,88%)。LC-MS(ESI):m/z=304.2[M+H]+.
第三步:将120B(200mg,0.66mmol)及54E(146mg,0.66mmol)溶于吡啶(6mL)中,室温缓慢滴加50%T3P的乙酸乙酯溶液(1.32mmol),50℃反应过夜。待反应冷至室温,向反应液中加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和柠檬酸溶液(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到标题化合物120C(260mg,78%)。LC-MS(ESI):m/z=507.4[M+H]+.
第四步:将120C(260mg,0.51mmol)溶于四氢呋喃(6mL)中,冰浴下分批加入60%氢化钠(31mg,0.77mmol),随后滴加氘代碘甲烷(0.15g,1.03mmol),继续搅拌1h,随后缓慢升温至室温反应1h。加入1M氯化氢溶液淬灭反应,加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到化合物120(87mg,32%)。
1H NMR(400MHz,CDCl3)δ8.45(s,1H),7.88-7.84(m,1H),7.16-7.11(m,1H),6.96(s,1H),4.96-4.91(m,1H),3.42-3.34(m,1H),2.95-2.87(m,4H),2.58(s,3H),2.06-1.97(m,1H),1.69-1.63(m,1H),0.89-0.83(m,1H).LC-MS(ESI):m/z=524.1[M+H]+.
实施例121:
第一步:将化合物119B(360mg,0.87mmol)及4-溴-5-氯-2-氟苯胺(253mg,1.13mmol)溶于吡啶(8mL)中,室温缓慢滴加T3P(50%in EA,3.48mmol),50℃反应过夜。反应完全后,向反应液中加入水(50mL),乙酸乙酯(20mL×3),萃取,合并有机层,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离 提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到目标化合物121A(400mg,74%)。LC-MS(ESI):m/z=621.1[M+H]+.
第二步:将121A(400mg,0.65mmol)溶于DMF(8mL)中,冰浴下,加入60%氢化钠(78mg,1.94mmol),搅拌10分钟,加入氘代碘甲烷(281mg,1.94mmol),室温反应30分钟。倒入冰水(20mL)淬灭,用乙酸乙酯(30mL×2)萃取,合并后的有机相用无水硫酸钠干燥,过滤,浓缩得到的粗品经硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=6∶1)得到121B(380mg,92%)。LC-MS(ESI):m/z=638.3[M+H]+.
第三步:室温下,将化合物121B(120mg,0.19mmol)溶于干燥的甲苯(10mL)中,向其中依次加入3,3-二氟三甲叉亚胺盐酸盐(29mg,0.31mmol),Pd2(dba)3(28mg,0.03mmol),BINAP(39mg,0.063mmol),CS2CO3(195mg,0.60mmol),搅拌均匀后,氮气氛围,100℃下反应16小时。TLC监测原料消失,停止反应。冷却至室温,向反应液中加入EA(10mL),有机相用水(15mL×2)洗涤,饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物经柱层析(PE∶EA=4∶1)分离纯化得到标题化合物121C(81mg,66%)。LC-MS(ESI):m/z=651.1[M+H]+.
第四步:将化合物121C(81mg,0.12mmol),2,6二甲基吡啶(26mg,0.24mmol)溶于无水二氯甲烷(10mL)中,冰浴下,加入叔丁基二甲硅基三氟甲磺酸酯(48mg,0.18mmol),反应4小时。TLC监测原料消失,加入饱和氯化铵溶液淬灭。向反应液中加入水(15mL×2)萃取,并用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物121(20mg,收率30%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%乙酸铵),梯度洗脱,流动相A含量从10%-55%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,Chloroform-d)δ8.75-8.55(m,1H),8.20-7.57(m,2H),7.11-6.72(m,1H),6.49-6.34(m,1H),5.08-4.93(m,1H),4.54-4.39(m,4H),4.00-3.61(m,2H).LC-MS(ESI):m/z=551.2[M+H]+.
实施例122:
以化合物121B为原料,参考实施例34操作(第一,二,三步),得到化合物122。
1H NMR(400MHz,Methanol-d4)δ8.45-8.41(m,1H),8.19-7.43(m,3H),7.19-6.80(m,1H),5.13-4.84(m,1H),4.09-3.77(s,1H),3.52-3.43(m,1H),3.41-3.23(m,1H).LC-MS(ESI):m/z=484.3[M+H]+.
实施例123:
第一步:将化合物123A(5g,23.84mmol),锌粉(15.6g,238.4mmol)溶于甲醇(100mL),随后加入氯化铵(12.75g,238.4mmol),室温搅拌1小时。过滤,将滤液减压浓缩,向残余物中加入乙酸乙酯(200mL),有机相依次用水(100mL),饱和食盐水(100mL)洗涤,减压浓缩得到化合物123B(4.29g,收率99.99%),无需进一步纯化,可直接用于下一步反应。LC-MS(ESI):m/z=180.0[M+H]+.
第二步:依次将123B(4.3g,23.92mmol),2,5-己二酮(13.65g,119.6mmol),对甲苯磺酸(0.82g,4.78mmol)溶于甲苯(120mL),升温至100℃下搅拌反应3小时。待反应冷却至室温,将反应液倒入水中,用乙酸乙酯(200mL×2)萃取,合并后的有机相用水(100mL×2)洗,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到的粗品用快速柱层析(PE/EA=50/1)纯化得到化合物123C(4.3g,收率69.6%)。
1H NMR(400MHz,CD3Cl)δ7.21-7.20(m,2H),5.88(s,2H),2.03(s,6H).
第三步:将化合物123C(0.5g,1.94mmol)溶于甲苯(50mL)中,依次向其中加入3,3-二氟吖啶盐酸盐(0.3g,2.31mmol),Pd2(dba)3(0.18g,0.19mmol),BINAP(0.24g,0.39mmol),碳酸铯(1.90g,5.83mmol),氮气氛围,升温至100℃搅拌反应18小时。反应完全后,冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯(100mL×2)萃取,合并后的有机相用饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到目标化合物123D(0.5g,收率80.9%)。
1H NMR(400MHz,CD3Cl)δ6.73-6.71(m,1H),6.23-6.20(m,1H),5.89(s,2H),4.38-4.32(m,1H),2.02(s,6H).
第四步:依次将123D(0.35g,1.11mmol),盐酸羟胺(1.54g,22.2mmol),三乙胺(0.56g,5.55mmol)溶于乙醇(9mL)和水(3mL)的混合溶剂,升温至100℃搅拌反应18小时。反应完全后,冷至室温,将反应液倒入水(30mL)中,乙酸乙酯(30mL×2)萃取,合并后的有机相用水(30mL)洗,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到化合物123E(0.15g,收率57.11%)。LC-MS(ESI):m/z=237.1[M+H]+.
第五步:将化合物98B(0.10g,0.26mmol),化合物123E(0.062g,0.26mmol)溶于吡啶(10mL),搅拌15分钟。将反应液冷却至0℃,逐滴滴入1-丙基磷酸酐(50%乙酸乙酯溶液)(1.0g,1.57mmol),搅拌30分钟后,升温至50℃搅拌反应18小时。冷却至室温,反应液倒入冰水中,用乙酸乙酯萃取(50mL×3),合并后的有机相,用饱和食盐 水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到化合物123F(0.15g,收率95.38%)。LC-MS(ESI):m/z=605.1[M+H]+.
第六步:将化合物123F(120mg,0.20mmol)溶于DMF(10mL),冷却至0℃,缓慢加入氢化钠(60%)(24mg,0.60mmol),搅拌15分钟,缓慢滴加氘代碘甲烷(87mg,0.60mmol),滴加完毕后,升温至室温搅拌反应1小时。将反应液逐滴滴加到稀盐酸中,然后用乙酸乙酯萃取(50mL×2),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物123G(120mg,收率96.47%),无需进一步纯化,直接用于下一步反应。LC-MS(ESI):m/z=622.2[M+H]+.
第七步:将化合物123G(120mg,0.19mmol)溶于二氯甲烷(8mL),冷却至-20℃,缓慢滴加1.0M三氯化硼二氯甲烷溶液(1.20mL,1.20mmol),滴加完毕后,自然升温至室温搅拌反应2小时。将反应液逐滴滴加到冰的饱和碳酸氢钠水溶液中,然后用乙酸乙酯(30mL×2)萃取,合并后的有机相,依次用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物123(12mg,收率10.85%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-60%,流量12mL/min。洗脱时间15min。LC-MS(ESI):m/z=582.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.59(s,1H),8.26-8.25(m,1H),7.24(s,1H),7.19-7.18(m,1H),6.90(s,1H),5.30-5.29(m,1H),4.49-4.38(m,4H),4.32-4.27(m,2H).
实施例124:
第一步:将化合物123C(0.5g,1.94mmol)溶于1,4-二氧六环(18mL),依次向其中加入2-噻吩硼酸频哪醇酯(0.49g,2.33mmol),Pd(dppf)Cl2(0.14g,0.19mmol),碳酸铯(0.76g,2.33mmol),水(2mL),氮气氛围下,升温至100℃搅拌反应18小时。反应完全后,冷却至室温,反应液经硅藻土过滤,滤液倒入水中,并用乙酸乙酯(50mL×2)萃取,合并后的有机相,用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物124A(0.25g,收率42.14%)。
1H NMR(400MHz,CD3Cl)δ7.53-7.52(m,1H),7.44-7.39(m,2H),7.21-7.14(m,2H),5.91(s,2H),2.02(s,6H).
第二步:依次将124A(0.25g,0.82mmol),盐酸羟胺(1.14g,16.4mmol),三乙胺(0.41g,4.1mmol)溶于乙醇(9mL)和水(3mL)的混合溶剂,升温至100℃搅拌反应18小时。反应完全后,冷至室温,将反应液倒入水(30mL)中,乙酸乙酯(30mL×2)萃取,合并后的有机相用水(30mL)洗,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到化合物124B(0.15g,收率80.34%)。LC-MS(ESI):m/z=228.1[M+H]+.
第三步:将化合物98B(0.10g,0.26mmol),化合物124B(0.059g,0.26mmol)溶于吡啶(10mL),搅拌15分钟。将反应液冷却至0℃,逐滴滴入1-丙基磷酸酐(50%乙酸乙酯溶液)(1.0g,1.57mmol),搅拌30分钟,升温至50℃反应18小时。反应完毕后,冷却至室温,反应液倒入冰水中,用乙酸乙酯萃取(50mL×3),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-40%EA/PE)纯化得到化合物124C(0.12g,收率77.45%)。LC-MS(ESI):m/z=438.0[M+H]+.
第四步:将化合物124C(120mg,0.2mmol)溶于DMF(10mL),冷却至0℃,缓慢加入氢化钠(60%)(24mg,0.60mmol),搅拌15分钟,缓慢滴加氘代碘甲烷(87mg,0.60mmol),滴加完毕后,升温至室温搅拌反应1小时。将反应液逐滴滴加到稀盐酸中淬灭,用乙酸乙酯萃取(50mL×2),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物124D(120mg,收率97.88%),无需进一步纯化,直接用于下一步反应。LC-MS(ESI):m/z=613.1[M+H]+.
第五步:将化合物124D(120mg,0.20mmol)溶于二氯甲烷(8mL),冷却至-20℃,缓慢滴加1.0M三氯化硼二氯甲烷溶液(1.20mL,1.20mmol),滴加完毕后,缓慢升温至室温搅拌反应2小时。将反应液逐滴滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯(30mL×2)萃取,合并后的有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物124(35mg,收率30.54%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-55%,流量12mL/min。洗脱时间15min。LC-MS(ESI):m/z=573.1[M+H]+.
1H NMR(400MHz,CD3OD)δ8.61(s,1H),8.25(s,1H),8.03-8.02(m,1H),7.80(s,1H),7.66-7.61(m,1H),7.21-7.18(m,1H),5.31-5.29(m,1H),4.33-4.27(m,2H).
实施例125:
以化合物98B和64B为原料,参考实施例8实验操作(第三、四、五步),得到化合物125。LC-MS(ESI):m/z=513.0[M+H]+.
1H NMR(400MHz,CD3OD)δ8.73(s,1H),8.26-8.25(d,1H),7.90-7.86(m,1H),7.70-7.67(m,1H),7.60(s,1H),7.27(d,1H),7.24-7.19(m,1H),5.29(s,1H),4.61-4.60(d,1H),4.57-4.54(t,1H).
实施例126:
第一步:将79C(0.21g,0.95mmol)及82B(0.23g,1.04mmol)溶于1,4-二氧六环(20mL)中,随后加入Pd2(dba)3(87mg,0.095mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(110mg,0.19mmol)及碳酸钾(0.26g,1.9mmol),氮气氛围下,于95℃反应过夜。反应完毕后,冷却至室温,向反应液中加入水(100mL),以乙酸乙酯(30mL×3)萃取,合并后的有机相,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物126A(0.34g,88.08%)。LC-MS(ESI):m/z=407.1[M+H]+.
第二步:依次将醋酸钯(0.21g,0.93mmol),三乙胺(0.26g,2.53mmol)加入到二氯甲烷(15mL)中,随后滴加三乙基硅烷(1.47g,12.64mmol),搅拌5分钟,然后滴加化合物126A(0.34g,0.84mmol)的二氯甲烷溶液,滴加完毕,室温下搅拌反应1小时。将反应液过滤,滤液浓缩后残留物用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-50%EA/PE)纯化得到化合物126B(0.24g,90.36%)。LC-MS(ESI):m/z=317.0[M+H]+.
第三步:以化合物126B(0.24g,0.76mmol)和4-溴-2-氯-5-氟苯胺(0.20g,0.91mmol)为原料,参考实施例8第三步操作得到化合物126C(0.26g,收率65.5%)。LC-MS(ESI):m/z=521.9[M+H]+.
第四步:以化合物126C(120mg,0.23mmol)为原料,参考实施例8第四步操作得到化合物126D(120mg,收率96.68%)。LC-MS(ESI):m/z=538.9[M+H]+.
第五步:将化合物126D(0.12g,0.22mmol)溶于甲苯(50mL)中,依次向其中加入3,3-二氟吖啶盐酸盐(0.042g,0.33mmol),Pd2(dba)3(0.030g,0.033mmol),BINAP(0.041g,0.066mmol),碳酸铯(0.22g,0.66mmol),氮气氛围,升温至100℃搅拌反应18小时。反应完毕后,冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯(100mL×2)萃取,合并后的有机相用饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物126(30mg,收率24.71%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-65%,流量12mL/min。洗脱时间15min。LC-MS(ESI):m/z=552.6[M+H]+.
1H NMR(400MHz,CD3OD)δ8.36-8.32(m,1H),8.16-8.10(m,1H),7.74-7.32(m,1H),7.11-6.92(m,1H),6.73-6.61(m,1H),5.19-5.12(m,1H),4.48-4.22(m,5H).
实施例127:(2S,3S,4S)-N-(3-氯-4,5-二氟苯基)-3,4-二羟基-N-(甲基-d3)-5-氧代-1-(7-(三氟甲基)呋喃[3,2-b]吡啶-5-基)吡咯烷-2-甲酰胺(化合物127)
(2S,3S,4S)-N-(3-chloro-4,5-difluorophenyl)-3,4-dihydroxy-N-(methyl-d3)-5-oxo-1-(7-(trifluoromethyl)furo[3,2-b]pyridin-5-yl)pyrrolidine-2-carboxamide
第一步:将127A(200mg,1.07mmol)溶于甲醇(10mL),加入氯化铵(572mg,10.70mmol),室温下,分三次加入锌粉(700mg,10.70mmol),每次间隔15分钟,加毕,室温搅拌反应12小时。反应完毕,垫硅藻土过滤,滤饼用乙酸乙酯(10mL)冲洗,收集滤液并减压浓缩,残余物加入乙酸乙酯(10mL),溶解后再次垫硅藻土抽滤,用乙酸乙酯(10mL)冲洗滤饼,收集滤液并减压浓缩,残余物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物127B(124mg,收率70.86%)。
1H NMR(400MHz,DMSO-d6)δ6.51-6.44(m,2H),5.52(s,2H).
第二步:将98B(120mg,0.31mmol)溶于吡啶(5mL),加入127B(61mg,0.37mmol),滴加1-丙基磷酸酐(1mL,50%in EA,1.68mmol),滴加完毕后,升温至50℃反应12小时。反应完毕,减压浓缩,残余物加入水(10mL),乙酸乙酯(10mL),溶解后加稀盐酸(15N)调pH至弱酸性,萃取,分出有机相,水相用乙酸乙酯(10mL×2)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩,残余物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物127C(160mg,收率97.05%)。LC-MS(ESI):m/z=532.0[M+H]+.
第三步:将127C(150mg,0.28mmol)溶于DMF(3mL),0℃下,加入NaH(14mg,0.34mmol,60%Wt),搅拌5分钟,滴加氘代碘甲烷(45mg,0.31mmol),滴加完毕后,升至室温反应0.5小时。反应完毕,将反应液滴入水(20mL)中淬灭,用乙酸乙酯(10 mL×3)萃取,合并后的有机相,用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到127D粗品,无需进一步纯化,直接进行下一步。LC-MS(ESI):m/z=549.0[M+H]+.
第四步:将上一步127D粗品溶于二氯甲烷(5mL),冷却至-30℃,滴加BCl3(1mL,1M in DCM),滴加完毕后,自然升至室温,反应2小时。反应完毕,加入饱和碳酸氢钠水溶液(20mL)淬灭,用二氯甲烷(10mL×3)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,减压浓缩,残余物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%MeOH/DCM)纯化得到化合物127(37mg,两步收率25.97%)。LC-MS(ESI):m/z=509.0[M+H]+.
1H NMR(400MHz,CD3OD)δ8.60(s,1H),8.26-8.24(m,1H),7.79-7.60(m,2H),7.21(s,1H),5.35-5.19(m,1H),4.57-4.23(m,2H).
实施例128:
以化合物120B为原料,参考实施例17实验操作(第一、二、三步),得到化合物128。
1H NMR(400MHz,CD3OD)δ8.25-8.21(m,1H),7.62-7.28(m,1H),7.00-6.95(m,1H),6.66-6.59(m,1H),4.95-4.81(m,1H),4.43-4.36(m,4H),3.49-3.20(m,2H),2.83-2.76(m,3H),2.51-2.38(m,3H).LC-MS(ESI):m/z=539.2[M+H]+.
实施例129:
以化合物128B为原料,参考实施例34实验操作(第一、三步),得到化合物129。
1H NMR(400MHz,CD3OD)δ8.25-8.21(m,1H),7.87-7.30(m,2H),7.00-6.95(m,1H),5.59-4.79(m,1H),4.03-3.83(m,1H),3.50-3.22(m,2H),2.83-2.76(m,3H),2.51-2.38(m,3H).LC-MS(ESI):m/z=472.0[M+H]+.
实施例130:
第一步:室温下,将109D(460mg,1.40mmol)溶于吡啶(10mL),加入5-氯-2,4-二氟苯胺(380mg,1.69mmol),搅拌均匀后,滴加T3P(3.0mL,5.04mmol),滴加完毕后升温至55℃反应16小时。TLC监测原料消失,停止反应。待反应冷至室温,向反应液中加入EA(20mL),有机相用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到130A(480mg,64.01%)。LC-MS(ESI):m/z=535.0[M+H]+.
第二步:室温下,将130A(480mg,0.90mmol)溶于干燥的DMF(10mL),冰水浴下,加入NaH(45mg,1.35mmol),搅拌10min后,加入CD3I(190mg,1.34mmol),随后升至室温,继续反应30min。TLC监测原料消失,停止反应。滴加1M的盐酸(10mL)淬灭反应,EA(20mL)萃取,有机相用饱和氯化钠溶液(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物130B(480mg,96.49%)。LC-MS(ESI):m/z=552.0[M+H]+.
第三步:室温下,将130B(120mg,0.22mmol)溶解于干燥的DMF(5mL),向其中依次加入三甲基硅基乙炔(65mg,0.66mmol),Pd(dppf)Cl2(32mg,0.044mmol),碘化亚铜(13mg,0.066mmol),三乙胺(67mg,0.66mmol),搅拌均匀,氮气氛围下,50℃反应16小时。TLC监测原料消失,停止反应。冷却至室温,向反应液中加入EA(15mL),有机相依次用水(15mL×2)洗涤,饱和食盐水(15mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物130C(108mg,86.12%)。LC-MS(ESI):m/z=570.1[M+H]+.
第四步:室温下,将130C(108mg,0.19mmol)溶于THF(5mL),向其中滴加四丁基氟化铵的四氢呋喃溶液(0.30mL,1mol/L),滴加完毕继续搅拌1小时。TLC监测原料消失,停止反应。向反应液中加入EA(15mL),有机相依次用饱和碳酸氢钠溶液(15mL×3)洗涤,饱和食盐水(15mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到化合物130(60mg,63.43%)。
1H NMR(400MHz,CD3OD)δ8.58-8.53(d,1H),8.20-8.15(m,1H),8.12-8.10(d,1H),7.71-7.61(m,1H),7.17-6.97(m,1H),5.06-4.94(m,1H),4.14(s,1H),3.65-3.61(m,1H),3.41-3.31(m,1H),2.95-2.89(m,3H).LC-MS(ESI):m/z=498.6[M+H]+.
实施例131:
第一步:将79C(0.5g,2.26mmol)溶于四氯化碳(10mL),于冰浴下加入溴素(0.72g,4.52mmol),室温反应过夜。待反应完全后,向反应液中加入30mL水,以乙酸乙酯 (15mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=10∶1)得到目标化合物131A(0.42g,62%)。
1H NMR(400MHz,CDCl3)8.05(s,1H),7.54(s,1H).
第二步:依次将131A(0.36g,1.20mmol)、甲基硼酸(79mg,1.32mmol)、二氯[1,1′-二(二苯基膦)二茂铁]钯(88mg,0.12mmol)及碳酸铯(0.59g,1.80mmol)溶于1,4-二氧六环(10mL)及水(1mL)的混合溶剂中,氮气氛围,100℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入40mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=4∶1)得到131B(164mg,58%)。LC-MS(ESI):m/z=236.1[M+H]+.
第三步:将131B(164mg,0.70mmol)及6A(205mg,0.70mmol)溶于1,4-二氧六环(10mL)中,随后加入Pd2(dba)3(64mg,0.07mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(81mg,0.14mmol)及碳酸钾(193mg,1.4mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入30mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到131C(249mg,72%)。LC-MS(ESI):m/z=491.1[M+H]+.
第四步:将乙酸钯(183mg,0.82mmol)及三乙胺(103mg,1.02mmol)溶于二氯甲烷(10mL)中,冰浴下滴加三乙基硅烷(593mg,5.10mmol),搅拌30min,缓慢滴加131C(249mg,0.51mmol)的二氯甲烷溶液(3mL),滴加完毕后,室温反应1h。过滤除去不溶物,以二氯甲烷(20mL×2)洗滤饼,滤液浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到131D(161mg,79%)。LC-MS(ESI):m/z=401.1[M+H]+.
第五步:将131D(161mg,0.40mmol)及5-氯-2,4-二氟苯胺(72mg,0.44mmol)溶于吡啶(5mL)中,室温下缓慢滴加50%T3P的乙酸乙酯溶液(1.2mmol),滴加完毕后,50℃反应过夜。待反应冷至室温,向反应液中加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和柠檬酸溶液(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到标题化合物131E(171mg,78%)。LC-MS(ESI):m/z=546.1[M+H]+.
第六步:将131E(171mg,0.31mmol)溶于四氢呋喃(6mL)中,冰浴下加入60%氢化钠(19mg,0.47mmol),随后滴加氘代碘甲烷(90mg,0.62mmol),滴加完毕后,继续搅拌1h,随后缓慢升温至室温反应1h。加入1M稀盐酸淬灭反应,加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到131F(124mg,70%)。LC-MS(ESI):m/z=563.1[M+H]+.
第七步:将131F(124mg,0.22mmol)溶于4M氯化氢的1,4-二氧六环溶液(5mL)中,室温反应过夜。待反应完全后,浓缩,向残留物加入15mL水,以乙酸乙酯(10mL×4)萃取,合并有机层,用无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到化合物131(35mg,30%)。
1H NMR(400MHz,CD3Cl)δ8.46(s,1H),8.03-8.98(m,1H),7.18-7.16(m,1H),7.10(s,1H),5.28-5.25(m,1H),4.40-4.36(m,1H),4.30-4.28(m,1H),2.45(s,3H).LC-MS(ESI):m/z=523.1[M+H]+.
实施例132:
以化合物98D为原料,参考实施例98操作(第五,六步),得到化合物132。
1H NMR(400MHz,CDCl3)δ8.75(s,1H),8.00-7.98(m,1H),7.73-7.70(m,1H),7.08-7.06(m,1H),6.43-6.39(m,1H),5.17(s,1H),4.82-4.74(m,2H),4.47-4.45(m,1H),3.88(s,1H),3.20-3.09(m,2H),2.63-2.51(m,2H).LC-MS(ESI):m/z=596.2[M+H]+.
实施例133:
第一步:将化合物121B(1.05g,1.65mmol)溶于DCM(20mL),加入三氟乙酸(5mL),室温下反应2小时。反应完全后,加饱和碳酸氢钠溶液(100mL)调节溶液pH至8,二氯甲烷(100mL×2)萃取,合并后的有机相,用饱和食盐水洗,无水硫酸钠干燥,过滤,滤液浓缩后得到标题化合物133A(0.88g,100%)。LC-MS(ESI):m/z=538.1[M+H]+.
第二步:室温下将化合物133A(0.88g,1.65mmol)溶于DMF(20mL),向其中加入碳酸铯(1.07g,3.3mmol),溴乙酸乙酯(0.41g,2.47mmol),升温至80℃反应2小时。反应完全后冷却至室温,加水(30mL),用乙酸乙酯(50mL×3)萃取,合并有机相,用饱和氯化钠水溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(PE∶EA(v/v)=2∶1)得到目标化合物133B(0.61g,59.3%)。LC-MS(ESI):m/z=624.1[M+H]+.
第三步:将化合物133B(0.55g,0.88mmol)溶于乙醇(20mL),依次加入硼氢化钠(50mg,1.32mmol),氯化锂(56mg,1.32mmol),室温反应2h。反应完全后直接浓缩,残留物经硅胶柱层析分离(PE∶EA(v/v)=1∶1)得到化合物133C(0.33g,64.5%)。LC-MS(ESI):m/z=582.1[M+H]+.
第四步:将133C(0.33g,0.57mmol)溶于DCM(20mL),加入三乙胺(0.17g,1.71mmol),加完后降温至0℃,向其中滴加甲基磺酰氯(0.1g,0.86mmol),滴加 完毕后,升至室温反应2小时。反应完全后,加水(20mL),用DCM(20mL×3)萃取,合并有机相,用饱和氯化钠水溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(PE∶EA(v/v)=2∶1)得到化合物133D(0.33g,收率88%)。LC-MS(ESI):m/z=660.1[M+H]+.
第五步:依次将133D(0.33g,0.5mmol),吗啉(87mg,1.0mmol),DIPEA(0.2g,1.5mmol),溶于乙腈(20mL)中,升温至60℃反应2小时。反应完全后冷却至室温,加水(30mL),用乙酸乙酯(30mL×3)萃取,合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(DCM∶MeOH(v/v)=10∶1)得到目标化合物133E(230mg,收率70%)。LC-MS(ESI):m/z=651.1[M+H]+.
第六步:室温下,将化合物133E(100mg,0.15mmol)溶于干燥的甲苯(10mL),向其中依次加入3,3-二氟三甲叉亚胺盐酸盐(24mg,0.18mmol),Pd2(dba)3(28mg,0.03mmol),BINAP(37mg,0.06mmol),CS2CO3(146mg,0.45mmol),搅拌均匀后,氮气氛围,100℃下反应16小时。TLC监测原料消失,停止反应。冷却至室温,向反应液中加水(30mL),用乙酸乙酯(30mL×3)萃取,合并后的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物经柱层析(DCM∶MeOH(v/v)=10∶1)分离纯化得到标题化合物133(8mg,7.8%)。
1H NMR(400MHz,Chloroform-d)δ8.65-8.57(m,1H),8.02-8.00(m,1H),7.91-7.85(m,1H),6.95-6.87(m,1H),6.47-6.35(m,1H),5.08-5.04(m,1H),4.51-4.45(m,4H),3.72-3.60(m,5H),3.53-3.37(m,3H),2.95(s,2H),2.88(s,2H),2.61-2.52(m,2H).LC-MS(ESI):m/z=664.2[M+H]+.
实施例134:
室温下,将130B(120mg,0.22mmol),溶解于干燥的甲苯(10mL),向其中依次加入3-氟氮杂环丁烷(20mg,0.27mmol),Pd2(dba)3(40mg,0.044mmol),BINAP(41mg,0.066mmol),碳酸铯(280mg,0.87mmol),氮气氛围下,100℃反应16小时。TLC监测原料消失,停止反应。将反应液冷却至室温,向其中加入EA(20mL),有机相用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到化合物134(45mg,37.40%)。
1H NMR(400MHz,CD3OD)δ8.59-8.54(d,1H),8.20-8.13(m,1H),7.80-7.34(m,1H),7.16-6.99(m,1H),6.71-6.59(m,1H),5.50-5.31(m,1H),5.15-5.00(m,1H),4.53-4.43(m,2H),4.24-4.16(m,2H),3.63-3.58(m,1H),2.97-2.90(m,3H).LC-MS(ESI):m/z=547.0[M+H]+.
实施例135:
以化合物98D为原料,参考实施例34操作(第一,二,三步),得到化合物135。
1H NMR(400MHz,Methanol-d4)δ8.69-8.64(m,1H),8.19-7.75(m,2H),7.65-7.18(m,1H),7.03-6.94(m,1H),5.50-4.85(m,1H),4.53-4.22(m,2H),4.05-3.91(m,1H).LC-MS(ESI):m/z=515.1[M+H]+.
实施例136:
以98B(200mg,0.56mmol)和70D为原料,参考实施例8操作(第三,四,五步),得到化合物136。
1H NMR(400MHz,CD3OD)δ8.79-8.74(m,1H),8.29-8.28(m,1H),8.22-8.05(m,1H),7.29-7.19(m,1H),7.11-7.08(m,1H),5.60-4.90(m,1H),4.63-4.58(m,2H),4.33-4.31(m,1H),3.27-3.13(m,2H),2.87-2.79(m,2H).LC-MS(ESI):m/z=597.0[M+H]+.
实施例137:
第一步:室温下,将137A(2.5g,16.40mmol)溶于干燥的DMF(10mL),冰水浴下,分批加入NaH(780mg,19.5mmol),搅拌10min后,滴加碘甲烷(2.56g,18.04mmol),滴加完毕后升至室温,继续反应30min。TLC监测原料消失,停止反应。滴加1M的盐酸(10mL)淬灭反应,EA(20mL×2)萃取,合并后的有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物137B(2.3g,84.17%)。LC-MS(ESI):m/z=167.1[M+H]+.
第二步:室温下,将137B(2.0g,12mmol)溶解于乙腈(12mL),向其中加入乙酰氯(1.39g,18mmol),搅拌均匀,再向其中加入碘化钠(17.99g,120mmol),80℃微波反应3小时。停止反应,向反应液中加入DCM(20mL),有机相依次用饱和碳酸氢钠水溶液(20mL×2)洗涤,饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物137C(2.2g,71.04%)。LC-MS(ESI):m/z=259.0[M+H]+.
第三步:室温下,将137C(2.0g,7.75mmol)溶于DMSO(10mL),依次加入(三氟甲基)三甲基硅烷(3.31g,23.23mmol),硼酸三甲酯(2.42g,23.25mmol),碘化亚铜(150mg,0.77mmol),1,10-菲罗啉(140mg,0.78mmol),氟化钾(1.35g,23.24mmol),氮气氛围,60℃反应16小时。TLC监测原料消失,停止反应。待反应冷至室温,向反应液中加入EA(20mL),有机相用水(30mL×2)洗涤,饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到化合物137D(1.4g,90.25%)。LC-MS(ESI):m/z=201.2[M+H]+.
第四步:室温下,将137D(1.4g,6.99mmol)溶于DCM(20mL),冰浴下,氮气氛围,分批向反应中加入间氯过氧苯甲酸(1.70g,8.39mmol),加入完毕,自然升温至室温反应3小时。TLC监测原料消失,停止反应。向反应液中加入DCM(20mL),有机相依次用饱和碳酸钾溶液(30mL×2)洗涤,饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物137E(1.3g,86.04%)。LC-MS(ESI):m/z=217.2[M+H]+.
第五步:室温下,将137E(1.3g,6.01mmol)溶于三氯氧磷(20mL),100℃反应6小时。TLC监测原料消失,停止反应。待反应冷至室温,浓缩,残余物用DCM(20mL)稀释后倒入冰水(20mL)中,滴加饱和碳酸氢钠水溶液调节pH为碱性,萃取,分液,有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得到化合物137F(980mg,69.50%)。LC-MS(ESI):m/z=235.1[M+H]+.
第六步:室温下,将6A(150mg,0.51mmol)溶解于干燥的二氧六环(10mL),向反应液中依次加入137F(0.14g,0.60mmol),Pd2(dba)3(93mg,0.1mmol),Xantphos(89mg,0.15mmol),碳酸钾(210mg,1.53mmol),氮气氛围,100℃下反应16小时。TLC监测原料消失,停止反应。待反应冷至室温,向反应液中加入EA(20mL),合并后的有机相用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到化合物137G(170mg,68.11%)。LC-MS(ESI):m/z=490.2[M+H]+.
第七步:室温下,将137G(170mg,0.35mmol)溶于甲醇(20mL),向反应液中加入钯碳(37mg,0.035mmol),氢气气氛下,室温反应16小时。TLC监测原料消失,停止反应。将反应液过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物137H(135mg,96.59%)。LC-MS(ESI):m/z=400.1[M+H]+.
第八步:室温下,将137H(135mg,0.34mmol)溶于吡啶(5mL),向反应液中加入5-氯-2,4-二氟苯胺(67mg,0.41mmol),搅拌均匀,向反应液滴加T3P(1.08g,1.70mmol),滴加完毕后,50℃反应16小时。TLC监测原料消失,停止反应。向反应液中加入EA(15mL),有机相用饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩 滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到137I(123mg,66.40%)。LC-MS(ESI):m/z=545.5[M+H]+.
第九步:室温下,将137I(123mg,0.23mmol)溶于干燥的DMF(5mL),冰水浴下,加入NaH(11.0mg,0.28mmol),搅拌10min后,向反应体系中加入CD3I(50mg,0.35mmol),升温至室温,继续反应30min。TLC监测原料消失,停止反应。滴加1M的盐酸(10mL)淬灭反应,EA(20mL×2)萃取,合并后的有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物137J(55mg,90.76%)。LC-MS(ESI):m/z=562.2[M+H]+.
第十步:室温下,将137J(60mg,0.11mmol)溶于DCM(5mL),降温至-20℃,向反应体系中滴加BCl3的二氯甲烷溶液(0.5mL,1mol/L,0.5mmol),滴加完毕后,升温至室温,继续反应1小时。TLC监测原料消失,停止反应。将反应液滴加入冰的饱和碳酸氢钠水溶液(10mL)中淬灭,DCM(10mL×2)萃取,合并后的有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物137(30mg,52.26%)。
1H NMR(400MHz,CD3OD)δ8.71-8.67(d,1H),8.29-7.91(m,1H),7.66-7.48(m,2H),6.87-6.66(m,1H),4.97-4.91(d,1H),4.61-4.57(m,1H),4.30-4.29(d,1H),3.96-3.94(t,3H).LC-MS(ESI):m/z=522.0[M+H]+.
实施例138:
第一步:室温下,将138A(4.3g,30.13mmol)溶于甲醇(40mL),冰浴下,向反应液中加入冰醋酸(3.08g,51.22mmol),搅拌均匀后,向反应液中加入溴素(8.19g,51.22mmol),0℃继续反应30分钟,TLC监测原料消失,停止反应。向反应液中加入饱和硫代硫酸钠溶液淬灭反应,EA(40mL×3)萃取,合并后的有机相用饱和食盐水(60mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化的到产物138B(4.2g,62.94%)。LC-MS(ESI):m/z=221.1[M+H]+.
第二步:室温下,依次将138B(4.2g,18.96mmol),醋酸钾(3.72g,37.91mmol),醋酸(36.43g,606.74mmol)溶于甲苯(50mL),冰浴下,向反应液中滴加亚硝酸异戊 酯(3.33g,28.44mmol),搅拌均匀,氮气氛围,30℃反应4小时,TLC监测原料消失,停止反应。浓缩,残余物用EA(50mL)溶解,用饱和碳酸氢钠溶液将pH调为碱性,有机相用饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化的到产物138C(1.3g,29.49%)。LC-MS(ESI):m/z=231.9[M+H]+.
第三步:室温下,将138C(1.3g,5.59mmol)溶于DMF(10mL),冰水浴下,加入NaH(140mg,6.0mmol),搅拌10min后,加入碘甲烷(950mg,6.69mmol),随后升至室温,继续反应30min。TLC监测原料消失,停止反应。滴加1M的盐酸(10mL)淬灭反应,EA(20mL×2)萃取,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物138D(620mg,45.00%)。LC-MS(ESI):m/z=245.9[M+H]+.
第四步:室温下,将138D(620mg,2.52mmol),三氟甲基(1,10-二氮杂菲)铜(950mg,3.05mmol)溶于DMF(5mL),氮气氛围,100℃反应16小时。TLC监测原料消失,停止反应,待反应冷却至室温,加入EA(20mL),有机相依次用水(30mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物138E(256mg,43.12%)。LC-MS(ESI):m/z=236.1[M+H]+.
第五步:室温下,将6A(180mg,0.62mmol)溶于二氧六环(10mL),向反应液中依次加入138E(0.16g,0.68mmol),Pd2(dba)3(110mg,0.12mmol),Xantphos(110mg,0.19mmol),碳酸钾(260mg,1.86mmol),氮气氛围,100℃反应16小时。TLC监测原料消失,停止反应,待反应冷至室温,向反应液中加入EA(20mL),有机相依次用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物138F(236mg,77.61%)。LC-MS(ESI):m/z=491.40[M+H]+.
第六步:室温下,将138F(236mg,0.48mmol)溶于甲醇(20mL),加入钯碳(51mg,0.048mmol),氢气氛围,室温反应16小时。TLC监测原料消失,停止反应。过滤,滤液浓缩后,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到产物138G(165mg,85.87%)。LC-MS(ESI):m/z=401.1[M+H]+.
第七步:室温下,将138G(165mg,0.41mmol)溶于吡啶(5mL),向反应液中加入5-氯-2,4-二氟苯胺(74mg,0.45mmol),搅拌均匀后,滴加T3P(1.30g,2.05mmol),滴加完毕后,55℃反应16小时。TLC监测原料消失,停止反应。向反应液中加入EA(15mL),有机相用饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,滤液浓缩后,残余物经柱层析(PE∶EA=2∶1)分离纯化得到138H(55mg,24.58%)。LC-MS(ESI):m/z=546.1[M+H]+.
第八步:室温下,将138H(55mg,0.10mmol)溶于DMF(5mL),冰水浴下,加入NaH(5mg,0.11mmol),搅拌10min后,向反应体系中加入CD3I(18mg,0.12mmol),升温至室温,继续反应30min。TLC监测原料消失,停止反应,滴加1M的盐酸(10mL)淬灭反应,EA(20mL×2)萃取,合并后的有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,滤液浓缩后,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物138I(20mg,35.53%)。LC-MS(ESI):m/z=563.1[M+H]+.
第九步:室温下将138I(20mg,0.036mmol)溶于DCM(5mL),降温至-20℃,向反应体系中滴加BCl3的二氯甲烷溶液(0.2mL,1mol/L,0.2mmol),滴加完毕后,升温至室温,继续反应1小时,TLC监测原料消失,停止反应。将反应液滴加到冰的饱和碳酸氢钠水溶液(10mL)中淬灭,DCM(10mL×2)萃取,合并后的有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,滤液浓缩后,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物138(8mg,42.50%)。
1H NMR(400MHz,CD3OD)δ8.80-8.79(d,1H),8.39-8.19(m,1H),8.16-7.90(m,1H),7.61-7.45(m,1H),5.26-5.14(m,1H),4.36-4.28(m,2H),4.23-4.20(m,3H).LC-MS(ESI):m/z=523.5[M+H]+.
实施例139:
以110G(200mg,0.56mmol)和3-氯-4-氟苯胺为原料,参考实施例8操作(第三,四,五步),得到化合物139。LC-MS(ESI):m/z=527.0[M+H]+.
1H NMR(400MHz,CD3OD)δ8.90(s,1H),7.91-7.89(m,1H),7.70-7.68(m,1H),7.42-7.38(t,1H),4.86(s,1H),4.57-4.56(d,1H),4.47-4.46(s,1H),3.20-3.19(t,2H),2.81-2.71(m,2H).
实施例140:
第一步:在100mL封管中,将140A(5g,26.85mmol)溶于DMF(30mL),滴加三乙胺(10.87g,107.42mmol),依次加入CuI(1.02g,5.31mmol),双三苯基磷二氯化钯(1.88g,2.69mmol),三甲基硅基乙炔(7.91g,80.58mmol),氮气氛围,50℃反应72小时。反应完成后,经硅藻土过滤,EA(60mL)冲洗滤饼,向滤液中加入水(60mL),萃取,分液,水相用乙酸乙酯(20mL×2)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩后,粗品用Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到目标化合物140B(250mg,收率4.58%)。
1H NMR(400MHz,DMSO-d6)δ7.05-7.01(m,1H),6.41-6.39(m,1H),6.34-6.30(m,1H),5.42(s,2H),2.20(s,3H),0.19(s,9H).
第二步:将98B(100mg,0.26mmol)溶于吡啶(5mL),加入140B(63mg,0.31mmol),滴加1-丙基磷酸酐(1mL,50%in EA,1.68mmol),滴加完毕后,升温至50℃反应12小时。反应完毕,减压浓缩,向残余物中加入水(10mL),乙酸乙酯(10mL),溶解后加稀盐酸(1.5N)调pH至弱酸性,萃取,分液,水相用乙酸乙酯(10mL×2)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩后,残余物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物140C(122mg,收率82.09%)。
1H NMR(400MHz,DMSO-d6)δ10.43(s,1H),8.52(s,1H),8.50-8.48(m,1H),7.47-7.45(m,1H),7.37-7.29(m,2H),7.20-7.18(m,1H),5.46-5.43(m,1H),5.14-5.10(m,1H),5.04-5.01(m,1H),2.31(s,3H),1.33-1.31(m,6H),0.22(s,9H).
第三步:将140C(122mg,0.21mmol)溶于DMF(5mL),冰水浴下,加入NaH(10mg,0.25mmol,60%Wt),搅拌5分钟厚,滴加CD3I(34mg,0.23mmol),滴加完毕后,升至室温反应0.5小时。反应完毕,将反应液滴加到纯化水(20mL)中淬灭,乙酸乙酯(10mL×3)萃取,合并后的有机相,用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩后,得到残余物140D粗品,无需进一步纯化,直接进行下一步。
第四步:将上一步140D粗品溶于二氯甲烷(5mL),降温至-30℃,滴加BCl3(1mL,1M in DCM),滴加完毕后,自然升至室温,反应2小时。反应完毕,加入饱和碳酸氢钠水溶液(20mL)淬灭,用二氯甲烷(10mL×3)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩后,得到残余物140E粗品,无需进一步纯化,直接进行下一步。
第五步:将上一步140E粗品溶解于干燥四氢呋喃(2mL),滴加TBAF(0.3mL,1M in THF),滴加完毕后,室温搅拌1小时。反应完毕,加入饱和食盐水(10mL),用乙酸乙酯(5mL×3)萃取,合并后的有机相用饱和食盐水洗(5mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩后,得到残余物用Biotage Isolera One(12g硅胶柱,洗脱剂:0-5%MeOH/DCM)纯化得到得到目标化合物140(17mg,三步收率17.00%)。
1H NMR(400MHz,CD3OD)δ8.57(s,1H),8.25-8.23(m,1H),7.65-7.62(m,1H),7.59(s,1H),7.52-7.48(m,1H),7.20-7.18(m,1H),5.24-5.20(m,1H),4.27-4.21(m,2H),3.84(s,1H),2.54(s,3H).LC-MS(ESI):m/z=477.2[M+H]+.
实施例141:
第一步:将141A(参考专利WO2021028670中描述方法合成)(500mg,2.67mmol)及2-氯-6-甲基-4-(三氟甲基)吡啶(520mg,2.67mmol)溶于1,4-二氧六环(10mL)中,随后加入Pd2(dba)3(244mg,0.27mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(310mg, 0.53mmol)及碳酸钾(0.73g,5.29mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入30mL水,以乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到141B(528mg,57%)。LC-MS(ESI):m/z=347.1[M+H]+.
第二步:将141B(528mg,0.82mmol)溶于二氯甲烷(15mL),加入三乙胺(0.31g,3.02mmol),冰浴下滴加对甲苯磺酰氯(0.31g,1.68mmol),然后升至室温反应过夜。待反应完全后,向反应液中加入30mL水,以二氯甲烷(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=10∶1)得到141C(493mg,65%)。LC-MS(ESI):m/z=501.1[M+H]+.
第三步:将141C(493mg,0.98mmol)、5-氯-2,4-二氟苯胺(176mg,1.08mmol)及三乙胺(198mg,1.96mmol)溶于乙腈(20mL)中,升温至80℃反应过夜。待反应完全后,冷至室温,向反应液中加入40mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到141D(287mg,60%)。LC-MS(ESI):m/z=492.1[M+H]+.
第四步:将141D(287mg,0.31mmol)溶于四氢呋喃(6mL),冰浴下加入60%氢化钠(35mg,0.87mmol),随后滴加氘代碘甲烷(130mg,0.87mmol),继续搅拌1h,随后缓慢升温至室温反应1h。加入1M稀盐酸淬灭反应,加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=8∶1)得到141E(133mg,45%)。LC-MS(ESI):m/z=509.1[M+H]+.
第五步:将141E(133mg,0.22mmol)溶于4M氯化氢的1,4-二氧六环溶液(5mL)中,室温反应过夜。待反应完全后,浓缩,向残留物中加入15mL水,以乙酸乙酯(10mL×4)萃取,合并有机层,用无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到化合物141(35mg,29%)。
1H NMR(400MHz,CD3Cl)δ8.54(s,1H),7.65-7.62(m,1H),7.13(s,1H),6.44-6.40(m,1H),5.09(s,1H),4.71-4.69(m,1H),4.52-4.40(m,3H),2.64(s,3H).LC-MS(ESI):m/z=469.0[M+H]+.
实施例142:
第一步:将79A(1.5g,8.19mmol)及4,4-二氟乙酰乙酸乙酯(4.08g,24.57mmol)溶于冰醋酸(10mL),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,浓缩,向残留物中加入30mL饱和碳酸氢钠水溶液,以乙酸乙酯(15mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到目标化合物142A(668mg,42%)。LC-MS(ESI):m/z=186.1[M+H]+.
第二步:将142A(668mg,3.61mmol)溶于三氯氧磷(5mL),氮气氛围,100℃下反应3h。反应完全后冷却至室温,浓缩,向残留物中加入20mL饱和碳酸氢钠水溶液,以乙酸乙酯(10mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=20∶1)得到目标化合物142B(295mg,40%)。LC-MS(ESI):m/z=204.2[M+H]+.
第三步:将142B(295mg,1.45mmol)及6A(422mg,1.45mmol)溶于1,4-二氧六环(10mL)中,随后加入Pd2(dba)3(133mg,0.14mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(168mg,0.29mmol)及碳酸钾(401mg,2.90mmol),氮气氛围,95℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入30mL水,以乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到142C(315mg,47%)。LC-MS(ESI):m/z=459.2[M+H]+.
第四步:将乙酸钯(250mg,1.11mmol)及三乙胺(140mg,1.39mmol)溶于二氯甲烷(6mL),冰浴下滴加三乙基硅烷(802mg,6.88mmol),搅拌30min,缓慢滴加142C(315mg,0.69mmol)的二氯甲烷溶液(2mL),滴加完毕后,室温反应1h。过滤除去不溶物,以二氯甲烷(20mL×2)洗滤饼,合并滤液,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到142D(222mg,87%)。
第五步:将142D(222mg,0.60mmol)及5-氯-2,4-二氟苯胺(110mg,0.66mmol)溶于吡啶(5mL),室温下缓慢滴加50%T3P的乙酸乙酯溶液(3.0mmol),滴加完毕后升温至50℃反应过夜。反应完全后,冷至室温,向反应液中加入30mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和柠檬酸溶液(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到142E(176mg,57%)。LC-MS(ESI):m/z=514.0[M+H]+.
第六步:将142E(176mg,0.34mmol)溶于四氢呋喃(6mL)中,冰浴下加入60%氢化钠(20mg,0.51mmol),随后滴加氘代碘甲烷(74mg,0.51mmol),继续搅拌1h, 随后缓慢升温至室温反应1h。加入1M氯化氢溶液淬灭反应,加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到142F(120mg,66%)。LC-MS(ESI):m/z=531.5[M+H]+.
第七步:将142F(120mg,0.23mmol)溶于4M氯化氢的1,4-二氧六环溶液(5mL)中,室温反应过夜。反应完全后,浓缩,向残留物加入15mL水,以乙酸乙酯(10mL×4)萃取,合并有机层,用无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=20∶1)得到化合物142(32mg,28%)。
1H NMR(400MHz,CD3Cl)δ8.57(s,1H),7.92-7.87(m,2H),7.16-7.10(m,1H),6.98-6.96(m,1H),6.91(s,1H),4.99(s,1H),4.74-4.72(m,1H),4.37-4.35(m,1H).LC-MS(ESI):m/z=491.0[M+H]+.
实施例143:
第一步:将110G(500mg,1.18mmol)及4-溴-5-氯-2-氟苯胺(291mg,1.30mmol)溶于吡啶(6mL),室温下,滴加50%T3P的乙酸乙酯溶液(5.9mmol),50℃反应过夜。反应完全后,冷至室温,向反应液中加入30mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和柠檬酸溶液(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到143A(483mg,65%)。LC-MS(ESI):m/z=628.5[M+H]+.
第二步:将143A(483mg,0.77mmol)溶于四氢呋喃(10mL),冰浴下加入60%氢化钠(46mg,1.16mmol),随后滴加氘代碘甲烷(167mg,1.16mmol),继续搅拌1h,随后缓慢升温至室温反应1h。加入1M氯化氢溶液淬灭反应,加入20mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到143B(244mg,49%)。
1H NMR(400MHz,CD3Cl)δ8.94(s,1H),8.12-8.10(m,1H),7.63-7.60(m,1H),5.00(s,1H),4.96-4.94(m,1H),4.65-4.62(m,1H),3.19-3.12(m,2H),2.80-2.66(m,2H).LC-MS(ESI):m/z=645.0[M+H]+.
第三步:将143B(244mg,0.38mmol)、三甲基硅基乙炔(187mg,1.90mmol)、二三苯基膦二氯化钯(160mg,0.23mmol)及碘化亚铜(72mg,0.38mmol)溶于三乙胺(10mL),氮气氛围,50℃下反应过夜。待反应完全后,冷却至室温,向反应液中加入30 mL水,乙酸乙酯(15mL×3)萃取,合并有机层,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=10∶1)得到143C(225mg,89%)。LC-MS(ESI):m/z=663.6[M+H]+.
第四步:将143C(225mg,0.34mmol)溶于4M氯化氢的1,4-二氧六环溶液(6mL)中,50℃反应过夜。待反应完全后,冷却至室温,浓缩,向残留物加入15mL水,以乙酸乙酯(10mL×4)萃取,合并有机层,用无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到143D(166mg,78%)。LC-MS(ESI):m/z=623.6[M+H]+.
第五步:将143D(166mg,0.27mmol)溶于四氢呋喃(10mL),加入1M四丁基氟化铵的四氢呋喃溶液(0.4mL),室温反应1h。浓缩,向残留物加入20mL水,以乙酸乙酯(10mL×3)萃取,合并有机层,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到化合物143(70mg,47%)。
1H NMR(400MHz,CD3Cl)δ8.86(s,1H),8.04-8.02(m,1H),7.47-7.44(m,1H),4.94(s,1H),4.76-4.74(m,1H),4.48-4.45(m,1H),3.80-3.75(m,1H),3.67-3.62(m,1H),3.50(s,1H),2.80-2.66(m,2H).LC-MS(ESI):m/z=551.1[M+H]+.
实施例144:
以143B和3,3-二氟氮杂环丁烷盐酸盐为原料,参考实施例17操作(第三,四步反应),得到化合物144。
1H NMR(400MHz,Methanol-d4)δ8.92(s,1H),7.93-7.79(m,1H),6.81-6.67(m,1H),4.95(s,1H),4.58-4.52(m,4H),4.51-4.46(m,2H),3.26-3.03(m,2H),2.84-2.61(m,2H).LC-MS(ESI):m/z=618.6[M+H]+.
实施例145:
第一步:室温下,将79C(2.2g,9.93mmol)溶于干燥的THF(20mL),氮气氛围,-78℃向反应液中滴加正丁基锂(8mL,19.86mmol,2.5M),滴加完毕,继续反应1小时,向反应液中滴加三甲基氯硅烷(2.16g,19.83mmol),滴加完毕,继续反应1小时, TLC监测原料消失,停止反应。-78℃下,向反应液中加入饱和氯化铵水溶液淬灭反应,加入EA(20mL),萃取分液,有机相用水(20mL)洗涤,饱和食盐水(30×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得的产物145A(2.0g,68.57%)。LC-MS(ESI):m/z=294.1[M+H]+.
第二步:室温下,将145A(1.0g,3.40mmol)溶于乙腈(10mL),向反应液中依次加入氟化钾(240mg,4.08mmol),N-碘代丁二酰亚胺(7.65g,34mmol),55℃反应3小时。TLC监测原料消失,停止反应。将反应液冷却至室温,用饱和亚硫酸氢钠水溶液淬灭反应,加入EA(20mL),用饱和碳酸氢钠调节pH至碱性,萃取分液,有机相用饱和食盐水(30×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得的产物145B(650mg,55.02%)。LC-MS(ESI):m/z=348.0[M+H]+.
第三步:室温下,将145B(650mg,1.87mmol)溶于干燥的THF(10mL),-78℃下,氮气氛围下,向反应液中滴加正丁基锂(1.13mL,2.81mmol,2.5M),滴加完毕,继续反应1小时。将碘甲烷(660mg,4.66mmol)加入干燥的瓶中,向其中加入无水硫酸镁,搅拌5分钟,过滤,-78℃,将滤液滴加到反应液中,滴加完毕,继续反应3小时。TLC监测原料消失,停止反应。-78℃下,向反应液中加入饱和氯化铵水溶液淬灭反应,加入EA(20mL),萃取分液,有机相用水(20mL)洗涤,饱和食盐水(30×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得的产物145C(110mg,24.97%)。LC-MS(ESI):m/z=236.1[M+H]+.
第四步:室温下,将1A(100mg,0.27mmol)溶于干燥的二氧六环(5mL),向反应液中依次加入145C(70mg,0.30mmol),Pd2(dba)3(49mg,0.054mmol),Xantphos(47mg,0.081mmol),碳酸钾(110mg,0.81mmol),氮气氛围,100℃下反应16小时。TLC监测原料消失,停止反应,冷至室温。向反应液中加入EA(20mL),有机相用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化得到产物145D(110mg,72.38%)。LC-MS(ESI):m/z=563.5[M+H]+.
第五步:室温下将145D(110mg,0.20mmol)溶于DCM(5mL),降温至-20℃,向反应体系中滴加BCl3的二氯甲烷溶液(1.0mL,1mol/L,1.0mmol),滴加完毕后,升温至室温,继续反应1小时。TLC监测原料消失,停止反应。将反应液滴加入冰的饱和碳酸氢钠水溶液(10mL)中淬灭,DCM(10mL×2)萃取,合并后的有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物145(35mg,33.47%)。LC-MS(ESI):m/z=523.5[M+H]+.
1H NMR(400MHz,CD3OD)δ8.46-8.44(d,1H),8.17-7.90(m,1H),7.58-7.44(m,1H),6.88-6.67(d,1H),5.24-5.12(m,1H),4.35-4.26(m,2H),2.61-2.58(d,3H).
实施例146:
第一步:室温下,将146A(300mg,0.85mmol)溶于THF(10mL),向其中加入四丁基氟化铵(240mg,0.94mmol),反应1小时。TLC监测原料消失,停止反应。向反应液中加入EA(20mL),有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得到产物146B(180mg,75.78%)。LC-MS(ESI):m/z=279.9[M+H]+.
第二步:室温下,将146B(180mg,0.64mmol)溶于DMF(10mL),向其中加入氰化亚铜(63mg,0.70mmol),120℃反应16小时。TLC监测原料消失,停止反应。冷却至室温,向反应液中加入EA(20mL),有机相用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得到产物146C(79mg,69.12%)。LC-MS(ESI):m/z=179.1[M+H]+.
第三步:室温下,将1A(100mg,0.27mmol)溶于干燥的二氧六环(10mL),向反应液中依次加入146C(53mg,0.30mmol),Pd2(dba)3(49mg,0.054mmol),Xantphos(47mg,0.081mmol),碳酸钾(110mg,0.81mmol),氮气氛围,100℃下反应16小时。TLC监测原料消失,停止反应。冷却至室温,向反应液中加入EA(20mL),有机相用水(20mL×2)洗涤,饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=4∶1)分离纯化得到产物146D(60mg,43.93%)。LC-MS(ESI):m/z=506.1[M+H]+.
第四步:室温下将146D(60mg,0.12mmol)溶于DCM(5mL),降温至-20℃,滴加BCl3的二氯甲烷溶液(0.5mL,1mol/L,0.5mmol),滴加完毕后,升温至室温,继续反应1小时。TLC监测原料消失,停止反应。将反应液滴加入冰的饱和碳酸氢钠水溶液(10mL)中淬灭,DCM(10mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(DCM∶MeOH=15∶1)分离纯化得到化合物146(25mg,44.72%)。LC-MS(ESI):m/z=466.5[M+H]+.
1H NMR(400MHz,CD3OD)δ8.58-8.57(d,1H),8.30-8.24(m,1H),8.15-7.90(m,1H),7.58-7.45(m,1H),7.27-7.03(m,1H),5.24-5.11(m,1H),4.34-4.24(m,2H).
实施例147:
以化合物110G和114A为原料,参考实施例8实验操作(第三、四、五步),得到化合物147。
1H NMR(400MHz,CD3OD)δ7.40(s,1H),6.12(s,1H),5.98(s,1H),5.69-5.63(m,1H),3.39(s,1H),3.06-3.04(m,1H),2.94-2.92(m,1H),1.71-1.64(m,2H),1.32-1.18(m,2H),0.84-0.79(m,3H).LC-MS(ESI):m/z=507.5[M+H]+.
实施例148:
以化合物110G和140B为原料,参考实施例34操作(第一,二,三步),得到化合物148。
1H NMR(400MHz,CD3OD)δ8.90(s,1H),7.65-7.61(m,1H),7.59-7.53(m,1H),7.48-7.42(m,1H),4.91(s,1H),4.60-4.55(m,1H),4.46-4.43(m,1H),3.81(s,1H),3.22-3.14(m,2H),2.82-2.69(m,2H),2.50(s,3H).LC-MS(ESI):m/z=513.7[M+H]+.
实施例149:
第一步:将70B(0.26g,1.32mmol)溶于DMF(5mL),依次加入碳酸钾(0.36g,2.64mmol),二氟碘甲烷(0.36g,2.03mmol),室温反应过夜。反应结束,加入20mL水,以乙酸乙酯(10mL×3)萃取,合并有机层,用饱和食盐水(10mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物149A(0.31g,94.37%)。LC-MS(ESI):m/z=242.0[M+H]+.
第二步:室温下,将149A(0.31g,1.28mmol)溶于甲醇(10mL),加入锌粉(0.5g,7.64mmol),搅拌均匀,向反应体系中分批加入氯化铵(0.68g,12.71mmol),室温反应2小时。过滤,浓缩滤液,向残余物中加入EA(20mL),有机相用水洗(10mL×2),无水硫酸钠干燥,过滤,浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物149B(0.19g,收率70.16%)。LC-MS(ESI):m/z=212.1[M+H]+.
第三步:依次将化合物110G(0.16g,0.38mmol),化合物149B(0.16g,0.76mmol)溶于吡啶(5mL),搅拌15分钟。将反应液冷却至0℃,滴加1-丙基磷酸酐(50%乙酸乙酯溶液)(0.72g,1.13mmol),搅拌30分钟后,升温至50℃反应18小时。冷却至室温,反应液倒入冰水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物149C(76mg,收率32.05%)。LC-MS(ESI):m/z=616.6[M+H]+.
第四步:将化合物149C(75mg,0.12mmol)溶于DMF(3mL),冷却至0℃,缓慢加入氢化钠(60%)(7.1mg,0.19mmol),搅拌5分钟后,滴加氘代碘甲烷(35mg,0.24mmol),然后升温至室温搅拌反应0.5小时。将反应液滴加到稀盐酸中淬灭,乙酸乙酯(10mL×3)萃取,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-20%EA/PE)纯化得到化合物149D(65mg,收率85.58%)。LC-MS(ESI):m/z=633.70[M+H]+.
第五步:将化合物149D(65mg,0.10mmol)溶于HCl/二氧六环溶液(10mL),室温搅拌30小时。加入饱和碳酸氢钠水溶液调节pH至7-8,加入乙酸乙酯(20mL),萃取分液,有机相用饱和食盐水洗涤(20mL×2),干燥,浓缩得到粗品。粗品用高效液相制备纯化得到目标化合物149(35mg,收率59.04%)。
1H NMR(400MHz,CD3OD)δ8.93-8.92(d,1H),8.16(d,1H),7.51-6.76(m,2H),5.56-4.89(m,1H),4.57(d,1H),4.33(d,1H),3.22-2.73(m,4H).LC-MS(ESI):m/z=593.1[M+H]+.
实施例150:
以化合物98D为原料,参考实施例98操作(第五,六步),得到化合物150。
1H NMR(400MHz,CD3OD)δ8.78-8.73(m,1H),8.27-8.08(m,1H),7.77-7.46(m,1H),7.25-7.05(m,1H),6.60-6.49(m,1H),5.62-4.96(m,1H),4.69-4.29(m,2H),4.11-4.07(m,4H),2.25-2.01(m,4H),1.92-1.84(m,2H).LC-MS(ESI):m/z=586.2[M+H]+.
实施例151:
第一步:依次将4-溴-5-氯-2-氟苯胺(0.22g,0.98mmol),异丙烯基三氟硼酸钾(0.17g,1.15mmol),Pd(dppf)Cl2(0.072g,0.10mmol),碳酸钠(0.16g,1.48mmol),溶于二氧六环(9mL)和水(1mL),氮气氛围下,升温至100℃搅拌反应18小时。待反应完全后,冷却至室温,垫硅藻土过滤,滤液倒入水中,用乙酸乙酯萃取三次,合并后的有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到化合物151B(170mg,收率93.45%)。LC-MS(ESI):m/z=186.1[M+H]+.
第二步:依次将化合物98B(0.15g,0.36mmol),化合物151B(0.13g,0.70mmol)溶于吡啶(10mL),搅拌15分钟。将反应液冷却至0℃,滴加1-丙基磷酸酐(50%乙酸乙酯溶液)(1.38g,2.17mmol),滴加完毕后,搅拌30分钟,然后升温至50℃搅拌反应18小时。反应完全后,冷却至室温,反应液倒入冰水中,用乙酸乙酯萃取(50mL×3), 合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-25%EA/PE)纯化得到化合物151C(180mg,收率84.76%)。LC-MS(ESI):m/z=590.6[M-H]-.
第三步:将化合物151C(150mg,0.25mmol)溶于DMF(10mL),0℃下,缓慢加入氢化钠(60%)(20mg,0.50mmol),搅拌15分钟后,滴加氘代碘甲烷(40mg,0.28mmol),滴加完毕后,升至室温搅拌反应1小时。将反应液滴加到稀盐酸中淬灭,用乙酸乙酯萃取(50mL×2),合并后的有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物151D(60mg,收率39.54%),无需进一步纯化,直接用于下一步反应。LC-MS(ESI):m/z=607.2[M+H]+.
第四步:将化合物151D(60mg,0.1mmol)溶于4.0N盐酸二氧六环溶液(10mL),室温搅拌反应18小时。将反应液滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯(30mL×2)萃取,合并后的有机相,依次用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物151(20mg,收率35.64%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-60%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,CD3OD)δ8.93(s,1H),8.04-8.02(m,1H),7.34-7.31(m,1H),5.36-5.35(m,1H),5.08-4.93(m,1H),4.58-4.34(m,1H),3.77-3.50(m,1H),3.22-3.15(m,2H),2.80-2.75(m,2H),2.15-2.09(m,3H).LC-MS(ESI):m/z=567.1[M+H]+.
实施例152:
第一步:依次将化合物152A(2.0g,18.37mmol),碳酸钠(5.84g,55.11mmol)溶于水(15mL)中,冰浴下加入碘单质(4.66g,18.37mmol),室温下搅拌18h。滴加稀盐酸调节pH至酸性,用乙酸乙酯(30mL×3)萃取,合并后的有机相用饱和硫代硫酸钠水溶液洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到固体152B(2.2g,收率50.96%),无需进一步纯化可直接用于下一步反应。LC-MS(ESI):m/z=236.0[M+H]+.
第二步:将化合物152B(1.5g,6.38mmol)溶于二氧六环(20mL),依次加入Pd(PPh3)2Cl2(0.45g,0.64mmol),CuI(0.24g,1.28mmol),三乙胺(0.97g,9.57mmol),三甲基硅乙炔(0.94g,9.57mmol),氮气氛围,100℃反应过夜。反应完全后,冷却至室温,垫硅藻土过滤,乙酸乙酯(15mL×3)洗涤滤饼,滤液用饱和食盐水洗涤,无水 硫酸钠干燥,过滤,浓缩得到粗品,然后粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到目标化合物152C(0.40g,收率30.50%)。LC-MS(ESI):m/z=206.2[M+H]+.
第三步:室温下,将152C(0.40g,1.95mmol)溶于DCM(20mL)。冰浴,氮气氛围下,分批加入间氯过氧苯甲酸(0.50g,2.92mmol),室温反应18小时。TLC监测原料消失,停止反应。向反应液中加入DCM(20mL),有机相依次用饱和碳酸氢钠水溶液(30mL×2)洗涤,饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩得到粗品,粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%MeOH/DCM)纯化得到目标化合物152D(0.41g,收率95%)。LC-MS(ESI):m/z=222.1[M+H]+.
第四步:氮气氛围下,将152D(0.41g,1.85mmol)溶于三氯氧磷(8mL),然后升温至100℃反应18小时。反应完全后,冷却至室温,浓缩除去溶剂,向残留物中滴加饱和碳酸氢钠水溶液(10mL),滴加完毕后,乙酸乙酯(20mL×3)萃取,合并后的有机相,用无水硫酸钠干燥,过滤,浓缩后得到的粗品使用中压制备仪Biotage Isolera One(12g硅胶柱,洗脱剂:0-10%EA/PE)纯化得到目标化合物152E(0.22g,收率49.6%)。LC-MS(ESI):m/z=240.1[M+H]+.
第五步:依次将化合物152E(80mg,0.33mmol),化合物1A(0.1g,0.27mmol)溶于二氧六环(20mL),向其中依次加入Pd2(dba)3(25mg,0.027mmol),XantPhos(31mg,0.054mmol),碳酸钾(75mg,0.54mmol),氮气氛围下,升温至95℃反应18小时。反应结束后,冷却至室温,加入乙酸乙酯(50mL)稀释,过滤,滤液减压浓缩后得到粗品,粗品使用中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-30%EA/PE)纯化得到化合物152F(100mg,收率65.31%)。LC-MS(ESI):m/z=567.1[M+H]+.
第六步:室温下,将152F(100mg,0.18mmol)溶解于THF(6mL)中,向其中滴加四丁基氟化铵的四氢呋喃溶液(0.27mL,1mol/L),室温下继续搅拌1小时。TLC监测原料消失,停止反应。向反应液中加入乙酸乙酯(15mL),有机相用饱和碳酸氢钠水溶液(15mL×3)洗涤,饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物经中压制备仪Biotage Isolera One(20g硅胶柱,洗脱剂:0-40%EA/PE)分离纯化得到化合物152G(60mg,67.36%)。LC-MS(ESI):m/z=495.1[M+H]+.
第七步:将化合物152G(60mg,0.12mmol)溶于二氯甲烷(8mL),冷却至-20℃,滴加1.0M三氯化硼二氯甲烷溶液(1.20mL,1.20mmol),滴加完毕后,缓慢升温至室温搅拌反应2小时。将反应液滴加到冰的饱和碳酸氢钠水溶液中,用乙酸乙酯(30mL×2)萃取,合并后的有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗品,粗品用高效液相制备纯化得到目标化合物152(22mg,收率40.31%)。
制备方法:仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)。样品用DMF溶解,用0.45μm滤头过滤,制成样品液。制备色谱条件:流动相A,B组成:流动相A:乙腈流动相B:水(含1%TFA),梯度洗脱,流动相A含量从10%-70%,流量12mL/min。洗脱时间15min。
1H NMR(400MHz,CD3OD)δ8.25-8.06(m,1H),7.99-7.91(m,1H),7.56-7.45(m,1H),7.05-6.85(m,1H),5.25-5.14(m,1H),4.54-4.23(m,2H),2.58-2.54(m,3H).LC-MS(ESI):m/z=455.5[M+H]+.
实施例153:
以化合物153A为原料,参考实施例110操作(第五步到第十步),得到化合物153。
1H NMR(400MHz,CD3OD)δ8.43-8.41(m,1H),8.26-8.22(m,1H),7.85-7.83(m,1H),7.47-7.42(m,1H),5.11-5.07(m,1H),4.29-4.27(m,2H),3.05-3.03(m,2H),2.73-2.63(m,2H).LC-MS(ESI):m/z=477.1[M+H]+.
实施例154:(2S,3S,4S)-N-(5-氯-2,4-二氟苯基)-4-羟基-3-甲氧基-N-(甲基-d3)-1-(6-甲基-4-(三氟甲基)吡啶-2-基)-5-氧代吡咯烷-2-甲酰胺(化合物154)
(2S,3S,4S)-N-(5-chloro-2,4-difluorophenyl)-4-hydroxy-3-methoxy-N-(methyl-d3)-1-(6-methyl-4-(trifluoromethyl)pyridin-2-yl)-5-oxopyrrolidine-2-carboxamide
第一步:将6B(1.25g,2.77mmol)溶于4M氯化氢的1,4-二氧六环溶液(25mL),室温反应过夜。待反应完全后,减压浓缩除去溶剂,向残留物加入25mL水,以乙酸乙酯(10mL×4)萃取,合并有机层,用无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到154A(805mg,71%)。LC-MS(ESI):m/z=411.1[M+H]+.
第二步:将154A(805mg,1.95mmol)及三乙胺(592mg,5.85mmol)溶于DMF(15mL),冰浴下缓慢滴加苯甲酰氯(275mg,1.95mmol),滴加完毕后,室温反应过夜。待反应完全后,向反应液中加入40mL水,以乙酸乙酯(25mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=10∶1)得到物154B(300mg,30%)。LC-MS(ESI):m/z=515.2[M+H]+.
第三步:将154B(300mg,0.58mmol)溶于二氯甲烷(14mL),随后加入碘甲烷(330mg,2.33mmol)及氧化银(540mg,2.33mmol),室温反应过夜。待反应完全后,过滤,以乙酸乙酯洗滤饼,滤液浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到154C(255mg,83%)。LC-MS(ESI):m/z=529.2[M+H]+.
第四步:将154C(255mg,0.48mmol)溶于甲醇(8mL),加入10%钯碳粉末(50mg),氢气氛围,60℃反应过夜。反应完全后,冷却至室温,过滤,少量甲醇洗滤饼,将滤液减压浓缩后,残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到154D(203mg,96%)。LC-MS(ESI):m/z=439.5[M+H]+.
第五步:将154D(203mg,0.46mmol)及5-氯-2,4-二氟-N-氘代甲基苯胺(83mg,0.46mmol)溶于吡啶(10mL),室温缓慢滴加50%T3P的乙酸乙酯溶液(2.3mmol),50℃反应过夜。反应完全后,向反应液中加入30mL水,乙酸乙酯(20mL×3)萃取,合并有机层,用饱和柠檬酸溶液(30mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=5∶1)得到154E(104mg,38%)。LC-MS(ESI):m/z=601.2[M+H]+.
第六步:将154E(104mg,0.17mmol)溶于甲醇(6mL),加入碳酸钾(47mg,0.34mmol),室温反应过夜。待反应完全后,向反应液中加入20mL水,以乙酸乙酯(15mL×3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(二氯甲烷∶甲醇(v/v)=40∶1)得到化合物154(35mg,41%)。
1H NMR(400MHz,CD3Cl)δ8.55(s,1H),7.81-7.78(m,1H),7.38-7.35(m,1H),7.14(s,1H),5.05(s,1H),4.70-4.68(m,1H),4.41-4.39(m,1H),2.64(s,3H),2.15(s,3H).LC-MS(ESI):m/z=497.1[M+H]+.
实施例155:
以化合物143B为原料,参考实施例98操作(第五,六步),得到化合物155。
1H NMR(400MHz,CDCl3)δ8.78(s,1H),7.81-7.78(m,1H),6.49-6.45(m,1H),6.09-5.80(m,1H),4.99(s,1H),4.70-4.67(m,1H),4.56-4.46(m,2H),4.41-4.39(m,1H),3.54-3.50(m,1H),3.41-3.35(m,1H),3.08(s,2H),2.72-2.52(m,4H).LC-MS(ESI):m/z=632.2[M+H]+.
实施例156:
以化合物98D为原料,参考实施例98操作(第五,六步),得到化合物156。
1H NMR(400MHz,CDCl3)δ8.65(s,1H),7.92-7.90(m,1H),7.56-7.54(m,1H),7.02-7.01(m,1H),6.33-6.29(m,1H),6.14-5.84(m,1H),5.10-4.99(m,1H),4.78-4.73(m,1H),4.48-4.38(m,1H),4.23-4.17(m,2H),4.12-4.03(m,2H),3.10-2.97(m,1H).LC-MS(ESI):m/z=596.9[M+H]+.
实施例157:
以化合物98B、2,4-二氟-3,5-二氯苯胺为原料,参照实施例117操作(第一到第三步),合成化合物157。
1H NMR(400MHz,CD3OD)δ8.78-8.75(m,1H),8.29-7.61(m,2H),7.35-7.06(m,1H),5.60-4.85(m,1H),4.62-4.55(m,1H),4.31-4.29(m,1H).LC-MS(ESI):m/z=542.9[M+H]+.
实施例158:
以化合物21F、3-氯-4-氟-5-三氟甲基苯胺为原料,参照实施例117操作(第一到第三步),合成化合物158。
1H NMR(400MHz,Methanol-d4)δ8.52(s,1H),8.22-8.19(m,1H),8.02-8.00(m,1H),5.48-4.85(m,1H),4.55-4.53(m,1H),4.46-4.45(m,1H),3.21-3.01(m,4H),2.28-2.20(m,2H).LC-MS(ESI):m/z=559.5[M+H]+.
实施例159:
第一步:室温下,将3-氯-2-氟苯甲酸(5.0g,28.61mmol)溶解于浓硫酸(15mL),冰浴下,向其中滴加硝酸(1.99g,31.55mmol),滴加完毕,室温反应16小时。TLC监测原料消失,停止反应。将反应液倒入冰水中,搅拌,析出固体,过滤,用水洗涤滤饼,滤饼用EA(30mL)溶解,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过 滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物159B(5.5g,87.56%)。
第二步:室温下,将159B(5.5g,25.05mmol)溶解于DMF(20mL),向其中加入碳酸钾(10.38g,75.07mmol),搅拌均匀,再加入碘甲烷(7.11g,50.10mmol),室温反应16小时。TLC监测原料消失,停止反应。向反应液中加入EA(40mL),有机相用水(40mL×2)洗涤,饱和食盐水(40mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物159C(4.7g,80.33%)。
第三步:室温下,将159C(2.30g,9.85mmol)溶解于干燥的DCM(15mL),-78℃,氮气氛围下,向其中缓慢滴加二异丁基氢化铝(1.68g,11.82mmol),滴加完毕,继续反应10min。TLC监测原料消失,用甲醇淬灭反应,然后升温至室温,向反应液中加入饱和酒石酸钾钠溶液(10mL),继续搅拌4小时,加入DCM(25mL),萃取,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物159D(1.7g,84.79%)。
第四步:室温下,将159D(1.7g,8.35mmol)溶解于干燥的DCM(10mL),冰浴下,向反应液中滴加DAST(2.69g,16.73mmol),滴加完毕,室温反应2小时。TLC监测原料消失,停止反应。向反应液中加入DCM(20mL),用饱和碳酸氢钠溶液将pH调为碱性,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物159E(1.7g,90.27%)。
第五步:室温下,将159E(1.7g,7.54mmol)溶于甲醇(20mL),向反应液中加入锌粉(4.93g,75.4mmol),搅拌均匀后,分批加入氯化铵(4.03g,75.32mmol),然后在室温下反应30分钟。TLC监测原料消失,停止反应。将反应液过滤,浓缩滤液,残余物用EA(25mL)溶解,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物159F(470mg,31.87%)。LC-MS(ESI):m/z=196.2[M+H]+.
第六步到第八步:
以98B(150mg,0.39mmol)和159F(110mg,0.56mmol)为原料,参考实施例117的合成路线得到化合物159(20mg,42.02%)。LC-MS(ESI):m/z=541.4[M+H]+.
1H NMR(400MHz,CD3OD)δ8.61(s,1H),8.26(d,1H),8.09-8.06(m,1H),7.97-7.95(m,1H),7.26-6.99(m,2H),5.21-5.20(d,1H),4.28-4.27(d,2H).
实施例160:
第一步:室温下,将160A(1.5g,6.65mmol)溶于DMF(10mL),加入碳酸钾(2.76g,19.87mmol),搅拌均匀,加入碘甲烷(1.89g,13.3mmol),室温反应16小时。TLC监测原料消失,停止反应。向反应液中加入EA(30mL),有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得到产物160B(1.25g,78.49%)。
第二步:以160B(1.25g,5.22mmol)为原料,参考实施例32的第二步的合成方法得到化合物160C(570mg,39.61%)。
第三步:室温下,将160C(0.57g,2.07mmol)溶于干燥的DMF(5mL),冰浴下,加入NaH(99mg,2.48mmol),搅拌10min,加入CD3I(390mg,2.69mmol),继续反应30分钟。TLC监测原料消失,停止反应。加入水淬灭反应,向反应液中加入EA(20mL),萃取,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=10∶1)分离纯化得到产物160D(550mg,90.76%)。
第四步:以160D(550mg,1.88mmol)为原料,参考实施例32的第三步的合成方法得到化合物160E(250mg,69.03%)。LC-MS(ESI):m/z=193.2[M+H]+.
第五步:室温下,将98B(130mg,0.34mmol)溶解于吡啶(5mL)中,加入160E(72mg,0.37mmol),搅拌均匀,向反应液中滴加T3P(1.09g,1.71mmol),50℃反应16小时,TLC监测原料消失,停止反应。向反应液中加入EA(10mL),有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩滤液,残余物经柱层析(PE∶EA=2∶1)分离纯化得到产物160F(70mg,36.71%)。LC-MS(ESI):m/z=561.0[M+H]+.
第六步:以160F(70mg,0.12mmol)为原料,参考实施例8的第五步的合成方法得到化合物160(25mg,40%)。LC-MS(ESI):m/z=521.0[M+H]+.
1H NMR(400MHz,CD3OD)δ8.78(s,1H),8.26(d,1H),7.50-7.48(m,1H),7.46-7.43(m,1H),7.22-7.21(d,1H),5.01(s,1H),4.61-4.59(d,1H),4.52-4.50(d,1H),3.99(s,3H).
实施例161:
以160A(1.0g,4.44mmol)和二氟碘甲烷(0.95g,5.33mmol)为原料,参考实施例160的合成路线合成化合物161(10mg)。LC-MS(ESI):m/z=557.5[M+H]+.
1H NMR(400MHz,CD3OD)δ8.78(s,1H),8.27-8.26(d,1H),7.89-7.87(m,1H),7.74-7.72(d,1H),7.21-7.20(d,1H),7.18-6.82(t,1H),4.97(s,1H),4.60-4.58(d,1H),4.49-4.47(d,1H).
实施例162:
第一步:将162A(2g,14.81mmol)溶于1,2-二氯乙烷(50mL),随后分批加入三溴氧磷(4.25g,14.81mmol),氮气氛围,80℃反应过夜。待反应待反应完全后,冷至室温,向反应液中加入100mL水,乙酸乙酯(40mL×3)萃取,合并有机层,用饱和碳酸氢钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩后残留物用硅胶柱层析分离提纯(石油醚∶乙酸乙酯(v/v)=10∶1)得到162B(1.77g,60%)。LC-MS(ESI):m/z=198.2[M+H]+.
第二步:以162B为原料,参考实施例131第二步的合成步骤,合成得到162C。LC-MS(ESI):m/z=134.1[M+H]+.
第三步~第九步:以162C为原料,参考实施例88第四步~第十步的合成步骤,合成得到化合物162。
1H NMR(400MHz,CD3Cl)δ8.14-8.13(m,1H),7.66-7.64(m,1H),7.18-7.15(m,2H),6.94-6.92(m,1H),5.29-5.26(m,1H),4.41-4.37(m,1H),4.31-4.28(m,1H),2.64(s,3H).LC-MS(ESI):m/z=455.1[M+H]+.
实施例163:
以110G及3-氯-4-氟-5-(三氟甲基)苯胺为原料,参考实施例117的合成步骤,得到化合物163。
1H NMR(400MHz,CD3Cl)δ8.78(s,1H),8.02-7.99(m,1H),7.54-7.51(m,1H),4.87(s,1H),4.70-4.68(m,1H),4.40-4.37(m,1H),3.12-3.00(m,3H),2.72-2.56(m,2H).LC-MS(ESI):m/z=595.4[M+H]+.
生物测试例
1.Polθ酶学活性检测
在DNA模板,引物,dNTP均存在的条件下,Polθ酶可催化合成DNA双链,通过PicoGreen dye对DNA双链进行染色,得到的结果与Polθ的酶活性正相关。试验总体系为10μl,试验缓冲液为25m M Tris-HCl pH 7.5,12.5mM NaCl,0.5mM MgCl2,5% (v/v)glycerol,0.01%v/v Triton x-100,0.01%(w/v)Bovineγ-Globulin,1mM DTT。终止液为25mM Tris-HCl pH 7.5,10mM EDTA。试验开始时,在384孔板(PerkinElmer 6008269)中加入4nM的Polθ蛋白(aa1792~2590),50nM DNA模板(5′-GCT ACA TTG ACA ATG GCA TCA AAT CTC AGA TTG CGT CTT A TG ACA GCC GCG-3′),50nM引物(5′-GCG GCT GTC ATA AG-3′),再加入梯度稀释的受试化合物,室温孵育30min,加入40μM的dNTP(Takara 4030),室温孵育90min,加入4μl PicoGreen dye(lnvitrogen P7581)终止反应(PicoGreen dye:终止液,1∶80),室温避光孵育90min。使用EnspireTM Multilabel Reader酶标仪(PerkinElmer)在激发光485nM/发射光520nM检测。应用GraphPad Prism软件,计算IC50值。
本发明化合物对Polθ具有小于1000nM的IC50值,一些优选化合物的IC50<500nM,一些更优选化合物的IC50<200nM,一些更优选的化合物的IC50<100nM,一些更优选的化合物的IC50<50nM,一些更优选的化合物的IC50<10nM。部分具体化合物的IC50值如表1所示,其中A代表IC50<100nM,B代表100nM≤IC50<500nM,C代表500nM≤IC50
表1化合物对Polθ酶抑制活性

化合物DZ-01按照专利WO2021028670 Example 1中描述方法合成;
结论:本发明的化合物对Polθ酶学活性具有良好的抑制作用;尤其是其中化合物79的IC50<15nM。
2.细胞增殖抑制活性
DLD-1 BRCA2-/-细胞培养基为1640培养基(含10%FBS和1X Penicillin/Streptomycin)。细胞培养于37℃、5%CO2孵箱中。将细胞以500cells/100μL的密度接种到96孔板(Corning,CAT#3603)中,于37℃、5%CO2条件下培养过夜。第二天每孔加入1μL 100x含不同浓度受试化合物的培养基,每个浓度设3个复孔。同时设置DMSO溶媒对照组和阴性对照组,均为3个复孔。在37℃、5%CO2条件下继续培养10天,分别在第4天和第7天更换含化合物的培养基。10天后,每孔加入60μL检测试剂(Cell Viability Assay,Promega,G7573),置于振荡器上避光孵育15分钟,之后于PHERAstar FSX多功能酶标仪(BMG LABTECH)测定荧光信号值。应用GraphPad Prism软件,计算IC50值。
表2 DLD-1 BRCA2-/-细胞增殖抑制活性
化合物DZ-01按照专利WO2021028670 Example 1中描述方法合成;
结论:本发明的化合物对DLD-1 BRCA2-/-细胞增殖具有良好的抑制作用。
3.大鼠药代动力学测试
1.1试验动物:雄性SD大鼠,220g左右,6~8周龄,6只/化合物。购于成都达硕实验动物有限公司。
1.2试验设计:试验当天,24只SD大鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。
于给药前及给药后异氟烷麻醉经眼眶取血0.10mL,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5,15, 30min,1,2,4,6,8,24h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。
结论:本发明化合物,比如实施例化合物对大鼠具有良好的药代动力学性能。
4.小鼠药代动力学测试
1.1试验动物:雄性ICR小鼠,20~25g,18只/化合物。购于成都达硕实验动物有限公司。
1.2试验设计:试验当天,72只ICR小鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。
于给药前及给药后异氟烷麻醉经眼眶取血0.06mL,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5,15,30min,1,2,4,6,8,24h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。
结论:本发明化合物,比如实施例化合物对小鼠具有良好的药代动力学性能。
5.比格犬药代动力学测试
试验动物:雄性比格犬,8~11kg左右,6只/化合物,购于北京玛斯生物技术有限公司。
试验方法:试验当天,12只比格犬按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。于给药前及给药后通过颈静脉或四肢静脉取血1ml,置于EDTAK2离心管中。5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5,15,30min,1,2,4,6,8,10,12,24,48,72h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。
结论:本发明化合物,比如实施例化合物对比格犬具有良好的药代动力学性能。
6.猴药代动力学测试
试验动物:雄性食蟹猴,3~5kg,3~6年龄,6只/化合物。购于苏州西山生物技术有限公司。
试验方法:试验当天,6只猴按体重随机分组。给药前1天禁食不禁水14~18h,给药后4h给食。
于给药前及给药后通过四肢静脉取血1.0mL,置于EDTAK2离心管中。5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5min,15min,30min,1,2,4,6,8,10,12,24h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。
结论:本发明化合物,比如实施例化合物对猴具有良好的药代动力学性能。
7.hERG钾离子通道作用测试
实验平台:电生理手动膜片钳系统
细胞系:稳定表达hERG钾离子通道的中国仓鼠卵巢(CHO)细胞系
实验方法:稳定表达hERG钾通道的CHO(Chinese Hamster Ovary)细胞,在室温下用全细胞膜片钳技术记录hERG钾通道电流。玻璃微电极由玻璃电极毛胚(BF150-86-10,Sutter)经拉制仪拉制而成,灌注电极内液后的尖端电阻为2-5MΩ左右,将玻璃微电极插入放大器探头即可连接至膜片钳放大器。钳制电压和数据记录由pClamp 10软件通过 电脑控制和记录,采样频率为10kHz,滤波频率为2kHz。在得到全细胞记录后,细胞钳制在-80mV,诱发hERG钾电流(IhERG)的步阶电压从-80mV给予一个2s的去极化电压到+20mV,再复极化到-50mV,持续1s后回到-80mV。每10s给予此电压刺激,确定hERG钾电流稳定后(至少1分钟)开始给药过程。化合物每个测试浓度至少给予1分钟,每个浓度至少测试2个细胞(n≥2)。
数据处理:数据分析处理采用pClamp 10,GraphPad Prism 5和Excel软件。不同化合物浓度对hERG钾电流(-50mV时诱发的hERG尾电流峰值)的抑制程度用以下公式计算:
Inhibition%=[1-(I/Io)]×100%
其中,Inhibition%代表化合物对hERG钾电流的抑制百分率,I和Io分别表示在加药后和加药前hERG钾电流的幅度。
化合物IC50使用GraphPad Prism 5软件通过以下方程拟合计算得出:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
其中,X为供试品检测浓度的Log值,Y为对应浓度下抑制百分率,Bottom和Top分别为最小和最大抑制百分率。
结论:本发明化合物,比如实施例化合物对hERG钾通道电流没有明显的抑制作用。
8.肝微粒体稳定性测试
本实验采用人、犬、大鼠和小鼠五种属肝微粒体作为体外模型来评价受试物的代谢稳定性。
在37℃条件下,1μM的受试物与微粒体蛋白、辅酶NADPH共同孵育,反应至一定时间(5,10,20,30,60min)加入冰冷含内标的乙腈终止反应,采用LC-MS/MS方法检测样品中受试物浓度,以孵育体系中药物剩余率的ln值和孵育时间求得T1/2,并进一步计算肝微粒体固有清除率CLint(mic)和肝固有清除率CLint(Liver)
结论:本发明化合物,比如实施例化合物具有良好的肝微粒体稳定性。
9.CYP450酶抑制测试
本项研究的目的是应用体外测试体系评价受试物对人肝微粒体细胞色素P450(CYP)的5种同工酶(CYP1A2、CYP2C9、CYP2C19、CYP2D6和CYP3A4)活性的影响。CYP450同工酶的特异性探针底物分别与人肝微粒体以及不同浓度的受试物共同孵育,加入还原型烟酰胺腺嘌呤二核苷酸磷酸(NADPH)启动反应,在反应结束后,通过处理样品并采用液相色谱-串联质谱联用(LC-MS/MS)法定量检测特异性底物产生的代谢产物,测定CYP酶活性的变化,计算IC50值,评价受试物对各CYP酶亚型的抑制潜能。
表3化合物对于CYP2D6酶的抑制作用参数
化合物DZ-01按照专利WO2021028670 Example 1中描述方法合成。
结论:本发明化合物,比如实施例化合物对人肝微粒体细胞色素P450(CYP)的5种同工酶没有明显的抑制活性,其中化合物127对CYP2D6酶的抑制活性>30μM。
10.Caco2渗透性测试
试验使用单层Caco-2细胞,在96孔Transwell板中采用三平行孵育。将含有本发明化合物(2μM)或对照化合物地高辛(10μM)、纳多洛尔(2μM)和美托洛尔(2μM)的转运缓冲溶液(HBSS,10mM HEPES,pH 7.4±0.05)加入顶端侧或基底侧的给药端孔中。对应接收端孔中加入含DMSO的转运缓冲溶液。在37±1℃条件下孵育2小时后,取出细胞板并从顶端和底端各取出适量样品至新的96孔板中。随后加入含内标的乙腈沉淀蛋白。使用LC MS/MS分析样品并测定本发明化合物和对照化合物的浓度。浓度数据用于计算从单层细胞顶端侧向基底侧、以及基底侧向顶端转运的表观渗透系数,从而计算外排率。用荧光黄的渗漏评价孵育2小时后单层细胞的完整性。
结论:本发明化合物,比如实施例化合物具有良好的Caco2渗透性。

Claims (16)

  1. 一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
    其中,D环选自
    X1、X2各自独立地选自O或者S,或者X2不存在;
    X3选自O或者NRx3
    X4选自O或者NH或者NRx4
    Rx3独立选自COR;
    Rx4选自C1-4烷基、C2-4烯基、C2-4炔基、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的4-6元杂环烷基)、-(CH2)s-(含有1-3个选自N、S、O、Si、P杂原子的5-6元杂芳基)、-(CH2)s-C3-6环烷基或-(CH2)s-苯基;所述烷基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选的被1-3个选自卤素、D、OH、CN、C1-4烷基、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
    z选自0或者1;
    s选自0、1、2、3、4或者5;
    环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
    环B为含有1-3个选自N、S、O杂原子的5-6元杂芳基、苯基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、含有1-3个选自N、S、O杂原子的6元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、苯并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
    C环为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或者苯基;
    R1和R2独立地为N3、OH或C1-4烷氧基,所述烷氧基任选地被1-3个选自D、卤素、OH、NH2和CN的基团取代;
    作为选择,R1或者R2与各自所连接的碳原子形成4-6元杂环;
    R3为C1-4烷基、C2-4烯基、C2-4炔基、含有1-3个选自N、S、O杂原子的4-6元杂环烷基或者C3-6环烷基,所述烷基、烯基、炔基、杂环烷基和环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
    作为选择,R3与B环形成5-6元杂环烷基;
    R4a、R5a、R4和R5各自独立地选自H、D、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
    n、m各自独立地选自0、1、2或3;
    p、q独立地为0、1、2、3或4;
    每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
    每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-(CH2)r-C3-6环烷基、-C(O)-C3-6环烷基或者苯基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基、环烷基和苯基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
    可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6碳环或含有1-3个选自N、S、O杂原子的5-6元杂环,所述碳环或杂环任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
    每个R7独立地为=O、卤素、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2、-NRaS(O)2-Ra,所述烷基、烯基、炔基、烷氧基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
    每个Ra各自独立选自H、D、C1-4烷基、C3-6环烷基;
    r选自0、1、2或3。
  2. 根据权利要求1所述一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,
    D选自D1或D4,所述化合物满足以下条件之一:
    (a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
    (b)R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进 一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;或者R3与B环形成5-6元杂环烷基;
    (c)m或n为1、2或3;或者X2不存在;
    (d)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、苯并含有1-3个选自N、S、O杂原子的5元杂环烷基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
    (e)至少存在一个R6不为卤素和C1-4烷基;
    (f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
    (g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
    (h)R3与B环形成5-6元杂环烷基;
    (i)z选自1;
    (j)X1或X2选自S。
  3. 根据权利要求1所述一种式(I-c)所示的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
    D选自D2,所述化合物满足以下条件之一:
    (k)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
    (l)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
    (m)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
    (n)环B为含有1-3个选自N、S、O杂原子的5元杂芳基、 含有1-3个选自N、S、O杂原子的5-6元杂环烷基并5-6元杂芳基、含有1-3个选自N、S、O杂原子的5-6元杂环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并含有1-3个选自N、S、O杂原子的5-6元杂芳基、5-6元环烷基并含有1-3个选自N、S、O杂原子的5元杂芳基、5-6元环烷基并苯基、含有1-3个选自N、S、O杂原子的5元杂芳基并5-6元杂环烷基或者苯并4-6元环烷基;
    (o)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代。
  4. 根据权利要求1-3任一项所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(I)、(I-a)、(I-b)、(I-d)、(I-e)、(I-f)的结构:
  5. 根据权利要求1-3任一项所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(II)、(II-d)、(II-e)、(I-b-1)的结构:

    其中,环A为含有1-3个选自N、S、O杂原子的5-6元单环杂芳基或含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
    环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基、
    R1和R2独立地为OH;
    R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
    R4和R5独立地为H、卤素、C1-4烷基、C2-4烯基或C2-4炔基;
    m为0、1或2;
    每个R6独立地为卤素、D、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
    每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4烯基、C2-4炔基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-C(O)-(含有1-3个选自N、S、O杂原子的4-6元杂环烷基)、-(CH2)r-(含有1-3个选自N、S、O杂原子的5-6元杂芳基)、-C(O)-C3- 6环烷基或者-(CH2)r-C3-6环烷基,所述烷基、烷氧基、烯基、炔基、杂环烷基、杂芳基和环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;或者每个R独立地为-(CH2)r-(含有1-3个选自N、S、O杂原子的7 元杂环烷基),所述杂环烷基任选地被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代;
    可选地,相邻环原子上的两个R6及其连接的原子一起形成C5-6环烷基或含有1-3个选自N、S、O杂原子的5-6元杂环烷基或杂芳基,所述环烷基、杂环烷基或杂芳基任选地被1-3个选自卤素、D、CN、OH和NH2的基团取代;
    r选自0、1、2;
    在式(II)、(II-d)中,所述化合物满足以下条件之一:
    (a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
    (b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
    (c)m为1或2;或者X2不存在;
    (d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
    (e)至少存在一个R6不为卤素和C1-4烷基;
    (f)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
    (g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O- (CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)或者含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
    (h)R3与B环形成5-6元杂环烷基;
    在式(II-e)中,所述化合物满足以下条件之一:
    (i)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
    (j)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
    (k)环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环并环杂环烷基或双环并环杂芳基;
    (l)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、嘧啶基、
    (m)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代。
  6. 根据权利要求1、2或3所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,
    环A为噻唑基、吡唑基、
    环B为苯基、吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、
    R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4元杂环烷基,所述烷基、杂环烷基任选的被1-3个选自D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
    R4和R5独立地为H或者C1-4烷基;
    m为0或1;
    p、q独立地为1、2、3或4;
    每个R6独立地为卤素、N3、CN、R、N(R)2、COR、CON(R)2、OR或NRCOR;
    每个R独立地为H、C1-4烷基、C1-4烷氧基、C2-4炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基,所述烷基、烷氧基、炔基、吗啉基、氮杂环丁基、氧杂环丁基、四氢吡咯基、四氢呋喃基、四氢吡喃基、环丙基、环丁基、吡唑基、呋喃基、吡咯基、噻吩基、咪唑基、噁唑基、噻唑基或者异噁唑基任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的 基团取代;或者每个R独立地为C2-4烯基、所述烯基、任选地被1-3个选自卤素、D、CN、OH、甲基、甲氧基、乙氧基、环丙基和NH2的基团取代;
    每个R7独立地为=O、CN、C1-4烷基、C2-4烯基、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-CH2-环丙基、-O-CH2CH2-环丙基、-O-CH2-环丁基、-O-CH2CH2-环丁基、-O-氧杂环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基、噻吩基、-P(=O)(C1-4烷基)2、-NHS(O)2C1-4烷基、-N(C1-3烷基)S(O)2C1-4烷基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基或噻吩基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基、环丙基和环丁基的基团取代;或者每个R7独立地为卤素;
    D选自D1或D4,所述化合物满足以下条件之一:
    (a)至少R1或R2其中一个为N3或C1-4烷氧基,或者R1或者R2与各自所连接的碳原子形成4-6元杂环;
    (b)R3为C1-4烷基或含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所述烷基并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代,所述杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;
    (c)m为1;
    (d)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、
    (e)至少存在一个R6不为卤素和C1-4烷基;
    (f)环A为噻唑基、吡唑基、
    (g)至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、环丙基、环丁基、-O-环丙基、-O-环丁基、-O-氧杂环丁基、-O-CH2-环丙基、-O-CH2CH2-环丙基、-O-CH2-环丁基、-O-CH2CH2-环丁基、氮杂环丁基、氧杂环丁基、-CH2-吗啉基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基、噻吩基、-P(=O)(C1-4烷基)2、-NHS(O)2C1-4烷基、-N(C1-3烷基)S(O)2C1-4烷基,所述烷基、烯基、炔基、烷氧基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吗啉基、四氢呋喃基、四氢吡咯基、吡唑基、吡咯基、吡啶基、嘧啶基、哒嗪基或噻吩基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基、环丙基和环丁基的基团取代;
    (h)R3与B环形成5-6元杂环烷基;
    D选自D2,所述化合物满足以下条件之一:
    (i)当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
    (j)当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH,且环B不为
    (k)环A为噻唑基、吡唑基、
    (l)环B为吡唑基、咪唑基、噻唑基、噻吩基、噁唑基、异噁唑基、异噻唑基、吡嗪基、哒嗪基、
    (m)当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代。
  7. 根据权利要求1-3任一项所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,包含式(II-a)或(II-b)结构:
  8. 根据权利要求1-3任一项所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,所述化合物具有式(II-a-1)、(II-e-1)的结构:
    在式II-a-1中,所述化合物满足以下条件之一:
    (a)、R3为C2-4烯基、C2-4炔基或者含有1-3个选自N、S、O杂原子的4-6元杂环烷基,所烯基、炔基和杂环烷基任选地被1-3个选自卤素、D、C2-4烯基、C2-4炔基和C3-6环烷基的基团取代;或者R3为C1-4烷基或C3-6环烷基,并进一步被C2-4烯基、C2-4炔基或C3-6环烷基取代;
    (b)、至少存在一个R6不为卤素和C1-4烷基;
    (c)、环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
    (d)、至少一个R7为=O、C2-4炔基、C1-4烷氧基、-COC1-4烷基、-SO2C1-4烷基、-CONHC1-4烷基、-CON(C1-4烷基)2、-(CH2)r-C3-6环烷基、-OC3-6环烷基、-O-(CH2)r-C3-6环烷基、-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷 基)、-O-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、含有1-3个选自N、S、O杂原子的5-6元杂芳基、-P(=O)Ra 2或者-NRaS(O)2-Ra,所述炔基、烷氧基、烷基、环烷基、杂环烷基和杂芳基任选地被1-3个选自卤素、C1-4烷基、D、CN、OH、C1-4烷氧基和C3-6环烷基的基团取代;
    在式II-e-1中,所述化合物满足以下条件之一:
    (e)、当X4选自NH或者NRx4时,至少存在一个R6不为卤素和C1-4烷基;
    (f)、当X4选自O时,至少存在一个R6不为卤素、C1-4烷基、C2-4烯基、未被取代的C3-6环烷基、未被取代的含有1-3个选自N、S、O杂原子的4-7元杂环烷基、-O-CH3或OH;
    (g)、环A为含有1-3个选自N、S、O杂原子的5元杂芳基或者含有1-3个选自N、S、O杂原子的8-10元双环杂并环;
    (h)、当R6选自R,R选自-(CH2)r-(含有1-3个选自N、S、O杂原子的4-7元杂环烷基)、-(CH2)r-C3-6环烷基,r选自0时,所述杂环烷基、环烷基进一步被1-3个选自卤素、D、CN、OH、C1-4烷氧基、C1-4烷基、C3-6环烷基和NH2的基团取代。
  9. 根据权利要求1-8任一项所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中:环A为含有1-3个选自N、S、O杂原子的8-10元双环杂并环。
  10. 权利要求1所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,所述化合物选自I组中的结构之一。
  11. 根据权利要求1所述的化合物,其立体异构体、氘代化物或药学上可接受的盐,其中,所述化合物选自II组中的结构之一。
  12. 一种药物组合物,其含有权利要求1-11中任意一项所述的化合物,或其立体异构体、氘代化物或药学上可接受的盐,以及药学上可接受的载体和/或辅料。
  13. 权利要求1-11中任意一项所述的化合物,其立体异构体、氘代化物、溶剂化物或者药学上可接受的盐,或者权利要求12所述的组合物在制备治疗Polθ介导的疾病的药物中的用途。
  14. 根据权利要求13所述的用途,其中,所述Polθ介导的疾病为肝癌、乳腺癌、卵巢癌、肺癌、肾癌、前列腺癌、皮肤癌、膀胱癌、胰腺癌或头颈癌。
  15. 一种药物组合物或药物制剂,所述的药物组合物或药物制剂包含选1-1000mg的权利要求1-11任意一项所述的化合物或者其立体异构体、氘代物、溶剂化物或者药学上可接受的盐和载体和/或辅料。
  16. 一种用于治疗哺乳动物的疾病的方法,所述方法包括给予受试者治疗有效量的权利要求1-11任意一项所述的化合物或者其立体异构体、氘代物、溶剂化物或者药学上可接受的盐,治疗有效量优选1-1000mg,所述的疾病优选肝癌、乳腺癌、卵巢癌、肺癌、肾癌、前列腺癌、皮肤癌、膀胱癌、胰腺癌或头颈癌。
PCT/CN2023/080706 2022-03-10 2023-03-10 DNA聚合酶θ抑制剂及其用途 WO2023169547A1 (zh)

Applications Claiming Priority (16)

Application Number Priority Date Filing Date Title
CN202210230303.7 2022-03-10
CN202210230303 2022-03-10
CN202210441233.X 2022-04-25
CN202210441233 2022-04-25
CN202210579356 2022-05-25
CN202210579356.X 2022-05-25
CN202210707891 2022-06-21
CN202210707891.9 2022-06-21
CN202210811425.5 2022-07-11
CN202210811425 2022-07-11
CN202210930668.0 2022-08-02
CN202210930668 2022-08-02
CN202210999788 2022-08-19
CN202210999788.6 2022-08-19
CN202310060809.2 2023-01-16
CN202310060809 2023-01-16

Publications (1)

Publication Number Publication Date
WO2023169547A1 true WO2023169547A1 (zh) 2023-09-14

Family

ID=87936145

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/080706 WO2023169547A1 (zh) 2022-03-10 2023-03-10 DNA聚合酶θ抑制剂及其用途

Country Status (2)

Country Link
TW (1) TW202341998A (zh)
WO (1) WO2023169547A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021028643A1 (en) * 2019-08-09 2021-02-18 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2021028670A1 (en) * 2019-08-09 2021-02-18 Artios Pharma Limited Deuterated compounds for use in the treatment of cancer
CN114127062A (zh) * 2019-05-31 2022-03-01 伊迪亚生物科学有限公司 作为DNA聚合酶Theta抑制剂的噻二唑基衍生物
WO2022167817A1 (en) * 2021-02-07 2022-08-11 Artios Pharma Limited Novel process

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114127062A (zh) * 2019-05-31 2022-03-01 伊迪亚生物科学有限公司 作为DNA聚合酶Theta抑制剂的噻二唑基衍生物
WO2021028643A1 (en) * 2019-08-09 2021-02-18 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2021028670A1 (en) * 2019-08-09 2021-02-18 Artios Pharma Limited Deuterated compounds for use in the treatment of cancer
WO2022167817A1 (en) * 2021-02-07 2022-08-11 Artios Pharma Limited Novel process

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
STOCKLEY MARTIN L., FERDINAND AMANDA, BENEDETTI GIOVANNI, BLENCOWE PETER, BOYD SUSAN M., CALDER MAT, CHARLES MARK D., EDWARDES LUC: "Discovery, Characterization, and Structure-Based Optimization of Small-Molecule In Vitro and In Vivo Probes for Human DNA Polymerase Theta", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 65, no. 20, 27 October 2022 (2022-10-27), US , pages 13879 - 13891, XP093089562, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.2c01142 *

Also Published As

Publication number Publication date
TW202341998A (zh) 2023-11-01

Similar Documents

Publication Publication Date Title
CN106103416B (zh) 作为trpm8拮抗剂的氮杂螺衍生物
CN102036981B (zh) 取代的4-羟基嘧啶-5-甲酰胺
WO2021218110A1 (zh) 一类苯并噻唑基联芳基类化合物、制备方法和用途
WO2020259432A1 (zh) Kras-g12c抑制剂
CN115803030A (zh) 用于kras靶向降解的化合物和方法
CN110753693A (zh) Egfr蛋白水解靶向嵌合分子和相关使用方法
CN104066431B (zh) 吡嗪激酶抑制剂
WO2023051716A1 (zh) 杂芳基衍生物parp抑制剂及其用途
CN107106559A (zh) 自分泌运动因子的取代的螺环抑制剂
CN112552295A (zh) Kras突变蛋白抑制剂
CN110156786A (zh) 嘧啶并环化合物及其制备方法和应用
CN110062758A (zh) 作为rho-激酶抑制剂的二环二氢嘧啶-甲酰胺衍生物
ES2780699T3 (es) Amidas de piperidina fusionadas útiles como moduladores de canales iónicos
WO2019000682A1 (zh) Rho相关蛋白激酶抑制剂、包含其的药物组合物及其制备方法和用途
CN113748114A (zh) 一种喹唑啉化合物及其在医药上的应用
CN104520296B (zh) 作为ttx-s阻断剂的吡咯并吡啶酮衍生物
CN106478497A (zh) 作为ttx‑s 阻滞剂的芳胺衍生物
CN114728962A (zh) 血浆激肽释放酶抑制剂及其用途
CN111434662B (zh) 卤代烯丙基胺类化合物及其应用
CN107207504B (zh) 酞嗪酮衍生物、其制备方法及用途
CN111499634A (zh) 一种喹唑啉化合物及其在医药上的应用
CN110520416B (zh) 多取代吡啶酮类衍生物、其制备方法及其医药用途
WO2021208918A1 (zh) 作为egfr抑制剂的三环化合物
CN106459033B (zh) 作为间变性淋巴瘤酶(alk)抑制剂的吲哚并喹诺酮类化合物
CN113923988A (zh) 取代的芳甲基脲和杂芳甲基脲、其类似物和使用其的方法

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: 23766124

Country of ref document: EP

Kind code of ref document: A1