WO2020244613A1 - 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物 - Google Patents

作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物 Download PDF

Info

Publication number
WO2020244613A1
WO2020244613A1 PCT/CN2020/094532 CN2020094532W WO2020244613A1 WO 2020244613 A1 WO2020244613 A1 WO 2020244613A1 CN 2020094532 W CN2020094532 W CN 2020094532W WO 2020244613 A1 WO2020244613 A1 WO 2020244613A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
haloalkyl
nrr
compound
heterocyclic group
Prior art date
Application number
PCT/CN2020/094532
Other languages
English (en)
French (fr)
Inventor
赵焰平
王红军
刘彬
姜媛媛
仲伟婷
徐慧芬
张宏雷
黄闯闯
田娜娜
赵静
李晶
刘伟娜
周丽莹
刘亚男
Original Assignee
北京泰德制药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 北京泰德制药股份有限公司 filed Critical 北京泰德制药股份有限公司
Priority to KR1020217043346A priority Critical patent/KR20220016225A/ko
Priority to JP2021572544A priority patent/JP7485701B2/ja
Priority to CN202080040989.2A priority patent/CN113906026A/zh
Priority to US17/616,571 priority patent/US20220378799A1/en
Priority to EP20818265.9A priority patent/EP3967694A4/en
Publication of WO2020244613A1 publication Critical patent/WO2020244613A1/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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to 2,4,6-trisubstituted pyrimidine compounds as inhibitors of ATR kinase. More specifically, the compounds of the present invention can be effectively used in the treatment of ATR kinase-mediated diseases, such as proliferative diseases, such as cancer.
  • the present invention also provides a pharmaceutical composition of the compound, its use for treating ATR kinase-mediated diseases, and preparation thereof.
  • ATR Alzheimer's disease telangiectasia and Rad3-related protein
  • ATR is a type of protein kinase involved in genome stability and DNA damage repair, and belongs to the PIKK family.
  • the activation of ATR can be activated by stalled replication forks or single-stranded DNA damage (SSB).
  • the activated ATR will recruit repair proteins or repair factors to repair the damaged parts, delay the mitotic process (especially in the G2/M phase of mitosis), which not only stabilizes the replication fork, but also ensures the stability of the genome.
  • the inhibition of ATR can be combined with radiotherapy or chemotherapy drugs to synergistically enhance the effect.
  • Widely used chemotherapy drugs include antimetabolites (such as gemcitabine), DNA crosslinkers (such as cisplatin, carboplatin), alkylating agents (such as temozolomide), topoisomerase inhibitors (such as topotecan, irinotecan) )Wait.
  • antimetabolites such as gemcitabine
  • DNA crosslinkers such as cisplatin, carboplatin
  • alkylating agents such as temozolomide
  • topoisomerase inhibitors such as topotecan, irinotecan
  • the present invention provides compounds of general formula (I), which can be used to treat ATR kinase-mediated diseases, such as proliferative diseases, such as cancer.
  • the present invention provides a compound of general formula (A), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CR 5 or N; Y is CR 6 or N; and wherein at least one of X and Y is N;
  • Z is CR # or N
  • R 1 , R 2 , R 3 , R 4 and R # are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; or R 1 and R 2 , R 3 and R 4 are connected to form a bond, C 1-6 alkylene, C 2-6 alkenylene or C 2-6 alkynylene;
  • R 5 and R 6 are independently selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl;
  • R # and R 1 may be connected to form a C 3-5 membered cycloalkyl group or a 3-5 membered heterocyclic group;
  • Ring A is selected from C 6-10 aryl and 5-10 membered heteroaryl
  • Ring B is selected from 5-10 membered heteroaryl groups
  • R* is selected from C 1-6 alkyl, C 1-6 haloalkyl, -L'-NRR', -L'-OR, -L'-C 3-7 cycloalkyl and -L'-4-8 Membered heterocyclic group; wherein L'is independently selected from bond, C 1-6 alkylene, C 2-6 alkenylene, C 2-6 alkynylene, -OC 1-6 alkylene and -NH- C 1-6 alkylene;
  • R b is independently selected from H, halogen, -CN, -L-OR, -LC 1-6 alkyl, -LC 1-6 alkoxy, -LC 3-7 cycloalkane optionally substituted by R** Group, -L-3-8 membered heterocyclic group, -NRR', -C(O)R, -C(O)OR, -C(O)NRR', -OC(O)R', -NRC( O)R', -OC(O)NRR' and -NRC(O)NRR';
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, or R, R'and the nitrogen atom to which they are attached form a 4-8 membered heterocyclic group;
  • n 0, 1, 2, 3, 4 or 5;
  • n 0, 1, 2, 3, 4, or 5.
  • the present invention provides a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , Polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N;
  • Y is CH or N; and wherein at least one of X and Y is N;
  • R 1 , R 2 , R 3 and R 4 are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; or R 1 and R 2 , R 3 and R 4 are connected to form a bond, C 1- 6 alkylene, C 2-6 alkenylene or C 2-6 alkynylene;
  • Ring A is selected from C 6-10 aryl and 5-10 membered heteroaryl
  • Ring B is selected from 5-10 membered heteroaryl groups
  • R b is independently selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group , -NRR', -C(O)R, -C(O)OR, -C(O)NRR', -OC(O)R', -NRC(O)R', -OC(O)NRR' And -NRC(O)NRR';
  • n 0, 1, 2, 3, 4 or 5;
  • n 0, 1, 2, 3, 4 or 5;
  • L is independently selected from bond, -O-, -S-, -NR-, -C(O)-, C 1-6 alkylene, C 2-6 alkenylene and C 2-6 alkynylene ;
  • R* is selected from C 1-6 alkyl, C 1-6 haloalkyl, -L'-NRR', -L'-OR, -L'-C 3-7 cycloalkyl and -L'-4-8 Membered heterocyclic group;
  • L' is independently selected from bond, C 1-6 alkylene, C 2-6 alkenylene, C 2-6 alkynylene, -OC 1-6 alkylene and -NH-C 1-6 alkylene base;
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, or R, R'and the nitrogen atom to which they are attached form a 4-8 membered heterocyclic group;
  • the present invention provides a pharmaceutical composition containing a compound of the present invention, and optionally a pharmaceutically acceptable excipient.
  • the present invention provides a pharmaceutical composition containing a compound of the present invention and a pharmaceutically acceptable excipient, which also contains other therapeutic agents.
  • the invention provides a kit comprising a compound of the invention, and other therapeutic agents, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the present invention provides the use of the compound of the present invention in the preparation of a medicament for the treatment and/or prevention of ATR kinase-mediated diseases.
  • the present invention provides a method of treating and/or preventing ATR kinase-mediated diseases in a subject, comprising administering a compound of the present invention or a composition of the present invention to the subject.
  • the present invention provides a compound of the invention or a composition of the invention for use in the treatment and/or prevention of ATR kinase-mediated diseases.
  • the diseases include proliferative diseases (such as cancer), especially solid tumors (such as cancer and sarcoma) and leukemia and lymphoma, especially for breast cancer, colorectal cancer, lung cancer (including small cell lung cancer, Non-small cell lung cancer and bronchioloalveolar cancer) and prostate cancer and bile duct cancer, bone cancer, bladder cancer, head and neck cancer, kidney cancer, liver cancer, gastrointestinal cancer, esophageal cancer, ovarian cancer, pancreatic cancer, skin cancer, Testicular cancer, thyroid cancer, uterine cancer, cervical cancer and vulvar cancer, as well as leukemia [including acute lymphoblastic leukemia (ALL) and chronic myeloid leukemia (CML)], multiple myeloma and lymphoma.
  • proliferative diseases such as cancer
  • solid tumors such as cancer and sarcoma
  • leukemia and lymphoma especially for breast cancer, colorectal cancer
  • lung cancer including small cell lung cancer, Non-small cell lung cancer and bro
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1 -2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 and C 5 -6 alkyl.
  • C 1-6 alkyl refers to a straight or branched chain saturated hydrocarbon group having 1 to 6 carbon atoms. In some embodiments, C 1-4 alkyl is preferred. Examples of C 1-6 alkyl groups include: methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), tert-butyl Base (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl ( C 5 ), 3-methyl-2-butyl (C 5 ), tert-amyl (C 5 ) and n-hexyl (C 6 ).
  • C 1-6 alkyl also includes heteroalkyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • the alkyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • alkyl abbreviations include: Me(-CH 3 ), Et(-CH 2 CH 3 ), iPr(-CH(CH 3 ) 2 ), nPr(-CH 2 CH 2 CH 3 ), n-Bu(-CH 2 CH 2 CH 2 CH 3 ) or i-Bu(-CH 2 CH(CH 3 ) 2 ).
  • C 2-6 alkenyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C 2-4 alkenyl is preferred. Examples of C 2-6 alkenyl groups include: vinyl (C 2 ), 1-propenyl (C 3 ), 2-propenyl (C 3 ), 1-butenyl (C 4 ), 2-butenyl (C 4 ), butadienyl (C 4 ), pentenyl (C 5 ), pentadienyl (C 5 ), hexenyl (C 6 ), etc.
  • C 2-6 alkenyl also includes heteroalkenyl, in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • Alkenyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 2-6 alkynyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C 2-4 alkynyl is preferred. Examples of C 2-6 alkynyl groups include but are not limited to: ethynyl (C 2 ), 1-propynyl (C 3 ), 2-propynyl (C 3 ), 1-butynyl (C 4 ), 2-butynyl (C 4 ), pentynyl (C 5 ), hexynyl (C 6 ), etc.
  • C 2-6 alkynyl also includes heteroalkynyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • An alkynyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 1-6 alkylene, C 2-6 alkenylene or C 2-6 alkynylene refers to the above-defined "C 1-6 alkyl, C 2-6 alkenyl or C 2-6 The divalent group of "alkynyl”.
  • C 1-6 alkylene group refers to a divalent group formed by removing another hydrogen of the C 1-6 alkyl group, and may be a substituted or unsubstituted alkylene group. In some embodiments, C 1-4 alkylene is particularly preferred.
  • the unsubstituted alkylene group includes but is not limited to: methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), butylene Group (-CH 2 CH 2 CH 2 CH 2 -), pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) ,and many more.
  • alkylene groups substituted by one or more alkyl groups (methyl) include, but are not limited to: substituted methylene (-CH(CH 3 )- , -C(CH 3 ) 2 -), substituted ethylene (-CH(CH 3 )CH 2 -, -CH 2 CH(CH 3 )-, -C(CH 3 ) 2 CH 2 -, -CH 2 C(CH 3 ) 2- ), substituted propylene (-CH(CH 3 )CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, -CH 2 CH 2 CH(CH 3 ) -, -C(CH 3 ) 2 CH 2 CH 2 -, -CH 2 C(CH 3 ) 2 CH 2 -, -CH 2 CH 2 C(CH 3 ) 2 -), etc.
  • C 2-6 alkynylene group refers to a divalent group formed by removing another hydrogen of the C 2-6 alkynyl group, and may be a substituted or unsubstituted alkynylene group. In some embodiments, C 2-4 alkynylene groups are particularly preferred. Exemplary alkynylene groups include, but are not limited to, ethynylene (-C ⁇ C-), substituted or unsubstituted propynylene (-C ⁇ CCH 2 -), and the like.
  • C 1-6 alkoxy refers to the group -OR, where R is a substituted or unsubstituted C 1-6 alkyl group. In some embodiments, C 1-4 alkoxy is particularly preferred. Specific alkoxy groups include but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, N-hexyloxy and 1,2-dimethylbutoxy. The alkoxy group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • Halo or "halogen” refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
  • C 1-6 haloalkyl and “C 1-6 haloalkoxy” refer to the above-mentioned “C 1-6 alkyl” and “C 1-6 alkoxy", which are substituted by one or more halogen groups. ⁇ Replacement.
  • C 1-4 haloalkyl is particularly preferred, and C 1-2 haloalkyl is more preferred.
  • C 1-4 haloalkoxy is particularly preferred, and C 1-2 haloalkoxy is more preferred.
  • haloalkyl groups include, but the are not limited to: -CF 3, -CH 2 F, -CHF 2, -CHFCH 2 F, -CH 2 CHF 2, -CF 2 CF 3, -CCl 3, -CH 2 Cl , -CHCl 2 , 2,2,2-trifluoro-1,1-dimethyl-ethyl, etc.
  • exemplary halogenated alkoxy groups include, but are not limited to: -OCH 2 F, -OCHF 2 , -OCF 3 , and the like.
  • Haloalkyl and haloalkoxy groups can be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 3-7 cycloalkyl refers to a non-aromatic cyclic hydrocarbon group having 3 to 7 ring carbon atoms and zero heteroatoms. In some embodiments, C 3-5 cycloalkyl is preferred. In other embodiments, C 3-6 cycloalkyl is preferred. In other embodiments, C 5-6 cycloalkyl is preferred. Cycloalkyl also includes ring systems in which the above-mentioned cycloalkyl ring is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring, and in this case, the number of carbons continues to indicate The number of carbons in the cycloalkyl system.
  • Exemplary cycloalkyl groups include, but are not limited to: cyclopropyl (C 3 ), cyclopropenyl (C 3 ), cyclobutyl (C 4 ), cyclobutenyl (C 4 ), cyclopentyl ( C 5 ), cyclopentenyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ), cycloheptyl (C 7 ), cycloheptene Group (C 7 ), cycloheptadienyl (C 7 ), cycloheptatrienyl (C 7 ), etc.
  • the cycloalkyl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • 3-11 membered heterocyclic group refers to a group of 3 to 11 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, Sulfur, boron, phosphorus and silicon.
  • the point of attachment may be a carbon or nitrogen atom.
  • a 3-9 membered heterocyclic group is preferred, which is a 3 to 9 membered non-aromatic ring system with ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, 3-8 membered Heterocyclic group, which is a 3 to 8-membered non-aromatic ring system with ring carbon atoms and 1 to 4 ring heteroatoms; preferably 3-6 membered heterocyclic group, which is a ring system with ring carbon atoms and 1 to 3 ring heteroatoms A 3 to 6 membered non-aromatic ring system of atoms; preferably a 3-5 membered heterocyclic group, which is a 3 to 5 membered non-aromatic ring system with ring carbon atoms and 1 to 2 ring heteroatoms; preferably 4-8 membered hetero Cyclic group, which is a 4 to 8-membered non-aromatic ring system with ring carbon atoms and 1 to 3
  • Heterocyclyl also includes a ring system in which the above heterocyclyl ring is fused with one or more cycloalkyl groups, wherein the point of attachment is on the cycloalkyl ring, or wherein the above heterocyclyl ring is connected to one or more aryl groups or Heteroaryl fused ring systems in which the point of attachment is on the heterocyclyl ring; and in this case, the number of ring members continues to indicate the number of ring members in the heterocyclyl ring system.
  • Exemplary 3-membered heterocyclic groups containing one heteroatom include, but are not limited to: aziridinyl, oxiranyl, and thiorenyl.
  • Exemplary 4-membered heterocyclic groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl, and thietane.
  • Exemplary 5-membered heterocyclic groups containing one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl and pyrrolyl-2, 5-dione.
  • Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to: dioxolane, oxasulfuranyl, disulfuranyl, and oxasulfuranyl.
  • Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl, and thiadiazolinyl.
  • Exemplary 6-membered heterocyclic groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridyl, and thianyl.
  • Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithiacyclohexyl, dioxanyl.
  • Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to: hexahydrotriazinyl (triazinanyl).
  • Exemplary 7-membered heterocyclic groups containing one heteroatom include, but are not limited to: azepanyl, oxepanyl, and thieppanyl.
  • Exemplary 5-membered heterocyclic groups fused to a C 6 aryl ring include, but are not limited to: indoline, isoindoline , Dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinone, etc.
  • Exemplary 6-membered heterocyclic groups fused with C 6 aryl rings include, but are not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, and many more.
  • the heterocyclyl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 6-10 aryl refers to a monocyclic or polycyclic (e.g., bicyclic) 4n+2 aromatic ring system (e.g., having 6-10 ring carbon atoms and zero heteroatoms) (e.g., having a ring arrangement Shared 6 or 10 ⁇ electrons) groups.
  • an aryl group having six ring carbon atoms ( “C 6 aryl”; e.g., phenyl).
  • the aryl group has ten ring carbon atoms ("C 10 aryl”; for example, naphthyl, for example, 1-naphthyl and 2-naphthyl).
  • the aryl group also includes a ring system in which the above-mentioned aryl ring is fused with one or more cycloalkyl or heterocyclic groups, and the point of attachment is on the aryl ring.
  • the number of carbon atoms continues to indicate The number of carbon atoms in the aryl ring system.
  • the aryl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • 5-10 membered heteroaryl group refers to a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms (for example, having a shared ring arrangement 6 or 10 ⁇ electrons), where each heteroatom is independently selected from nitrogen, oxygen and sulfur.
  • heteroaryl groups containing one or more nitrogen atoms as long as the valence allows, the point of attachment may be a carbon or nitrogen atom.
  • Heteroaryl bicyclic ring systems can include one or more heteroatoms in one or both rings.
  • Heteroaryl groups also include ring systems in which the above-mentioned heteroaryl ring is fused with one or more cycloalkyl or heterocyclic groups, and the point of attachment is on the heteroaryl ring, in this case, the carbon atom The number continues to indicate the number of carbon atoms in the heteroaryl ring system.
  • a 5-6 membered heteroaryl group is particularly preferred, which is a 5-6 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms.
  • Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furyl, and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl (for example, 1,2,4-oxadiazolyl), and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyridyl.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively.
  • Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepinyl, oxepinyl, and thiepinyl.
  • Exemplary 5,6-bicyclic heteroaryl groups include but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl , Benzisofuryl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazole, Indenazinyl and purinyl.
  • Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to: naphthyridinyl, pterridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl .
  • Heteroaryl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • heteroaryl groups include: pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl ( 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-l,2,3-triazolyl, pyranyl, 2-furanyl, 3-furan, etc., 2-Thienyl, 3-thienyl, oxazolyl, isoxazolyl, oxazolyl (1,2,4-oxazolyl, 1,3,4-oxazolyl, 1,2,5-oxazolyl Azolyl, thiazolyl, thiadiazolyl (1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl).
  • Carbonyl whether used alone or in combination with other terms (such as aminocarbonyl), is represented as -C(O)-.
  • alkyl group, alkenyl group, alkynyl group, carbocyclic group, heterocyclic group, aryl group, heteroaryl group, etc. defined herein are optionally substituted groups.
  • Each of Raa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclic, heterocyclyl, aryl and heteroaryl, or two Raa groups are combined to form a heterocyclic group or Heteroaryl ring, where each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl group is independently covered by 0, 1, 2, 3, 4 or 5 R dd groups Group replacement
  • Each of R cc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl, or two R cc groups are combined to form a heterocyclic ring Group or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl group is independently covered by 0, 1, 2, 3, 4 or 5 R dd group substitution;
  • R ee is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclic, aryl, heterocyclyl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbon Cyclic, heterocyclic, aryl and heteroaryl groups are independently substituted with 0, 1, 2, 3, 4 or 5 R gg groups;
  • Each of R ff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclic, heterocyclic, aryl, and heteroaryl, or two R ff groups are combined to form a heterocyclic group Or heteroaryl ring, where each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl group is independently covered by 0, 1, 2, 3, 4 or 5 R gg Group substitution
  • cancer includes but is not limited to the following cancers: breast, ovary, cervix, prostate, testis, esophagus, stomach, skin, lung, bone, colon, pancreas, thyroid, bile duct, buccal cavity and pharynx (mouth), lips, Tongue, oral cavity, pharynx, small intestine, colorectal, large intestine, rectum, brain and central nervous system cancer, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, adenocarcinoma, Adenoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder cancer, liver cancer, kidney cancer, bone marrow disorder, lymphatic disorder, Hodgkin's disease, hair cell carcinoma and leukemia.
  • treatment relates to reversing, reducing, inhibiting the progression of or preventing the disorder or condition to which the term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of verb therapy, the latter being as just defined.
  • pharmaceutically acceptable salt refers to those carboxylates and amino acid addition salts of the compounds of the present invention, which are suitable for contact with patient tissues within the scope of reliable medical judgment, and will not cause inappropriate toxicity, The irritation, allergies, etc., commensurate with a reasonable benefit/risk ratio, are effective for their intended application, including (where possible) the zwitterionic form of the compounds of the invention.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali metal and alkaline earth metal hydroxides or organic amines.
  • metals used as cations are sodium, potassium, magnesium, calcium and the like.
  • suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine and procaine.
  • Base addition salts of acidic compounds can be prepared by contacting the free acid form with a sufficient amount of the required base in a conventional manner to form the salt.
  • the free acid can be regenerated by contacting the salt form with the acid in a conventional manner, and then separating the free acid.
  • the free acid forms are somewhat different from their respective salt forms in certain physical properties, such as solubility in polar solvents, but for the purposes of the present invention, the salts are still equivalent to their respective free acids.
  • the salt can be sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate prepared from inorganic acid Salt, chloride, bromide, iodide, acid such as hydrochloric acid, nitric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, phosphoric acid, etc.
  • Representative salts include: hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurel Acid salt, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthoate, Methanesulfonate, glucoheptonate, lactobionate, laurylsulfonate and isethionate, etc.
  • Salts can also be prepared from organic acids, such as aliphatic mono- and di-carboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like.
  • organic acids such as aliphatic mono- and di-carboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like.
  • Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, horse Lysoate, mandelate, benzoate, chlorobenzoic acid basin, methylbenzoate, dinitrobenzoate, naphthoate, benzenesulfonate, tosylate, phenylethyl Acid salt, citrate, lactate, maleate, tartrate, methanesulfonate, etc.
  • Pharmaceutically acceptable salts may include cations based on alkali metals and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, etc., and non-toxic ammonium, quaternary ammonium and amine cations, including but not limited to ammonium, tetramethyl Ammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, etc. Also encompasses salts of amino acids, such as arginine, gluconate, galacturonate, etc. (see, for example, Berge SMet al., "Pharmaceutical Salts," J. Pharm. Sci., 1977; 66: 1- 19. This is incorporated as a reference).
  • Examples of pharmaceutically acceptable non-toxic amides of the compounds of the present invention include C 1 -C 6 alkyl esters, where the alkyl group is linear or branched. Acceptable esters also include C 5 -C 7 cycloalkyl esters and aryl alkyl esters, such as but not limited to benzyl esters. C 1 -C 4 alkyl esters are preferred.
  • the esters of the compounds of the present invention can be prepared according to conventional methods, for example: March's Advanced Organic Chemistry, 5 Edition" MB Smith & J. March, John Wiley & Sons, 2001.
  • Examples of pharmaceutically acceptable non-toxic amides of the compounds of the present invention include amides derived from ammonia, primary C 1 -C 6 alkyl amines, and secondary C 1 -C 6 dialkyl amines, where the alkyl group is linear or branched. Chained.
  • the amine may also be in the form of a 5- or 6-membered heterocyclic ring containing one nitrogen atom. Amides derived from ammonia, C 1 -C 3 alkyl primary amines and C 1 -C 2 dialkyl secondary amines are preferred.
  • the amides of the compounds of the present invention can be prepared according to conventional methods, for example: March's Advanced Organic Chemistry, 5 Edition", MBSmith & J. March, John Wiley & Sons, 2001.
  • Subjects to be administered include, but are not limited to: humans (ie, men or women of any age group, for example, pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young Adults, middle-aged adults or older adults)) and/or non-human animals, for example, mammals, for example, primates (for example, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep , Goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "human", “patient” and “subject” are used interchangeably herein.
  • treatment includes the effect that occurs when a subject suffers from a specific disease, disorder, or condition, which reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder Or the development of a condition ("therapeutic treatment"), and also includes effects that occur before the subject begins to suffer from a specific disease, disorder, or condition ("prophylactic treatment").
  • the "effective amount" of a compound refers to an amount sufficient to cause a target biological response.
  • the effective amount of the compound of the present invention may vary according to the following factors: for example, the biological target, the pharmacokinetics of the compound, the disease to be treated, the mode of administration, and the subject’s Age health and symptoms.
  • the effective amount includes a therapeutically effective amount and a preventive effective amount.
  • the "therapeutically effective amount” of the compound used herein is an amount sufficient to provide a therapeutic benefit during the treatment of a disease, disorder, or condition, or to cause one or more symptoms associated with the disease, disorder, or condition The amount of delay or minimization.
  • the therapeutically effective amount of a compound refers to the amount of the therapeutic agent when used alone or in combination with other therapies, which provides therapeutic benefits in the process of treating diseases, disorders, or conditions.
  • the term “therapeutically effective amount” may include an amount that improves overall treatment, reduces or avoids the symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
  • the “prophylactically effective amount” of the compound used herein is an amount sufficient to prevent a disease, disorder, or condition, or an amount sufficient to prevent one or more symptoms related to the disease, disorder, or condition, or prevent a disease The amount of recurrence of the disorder or condition.
  • the prophylactically effective amount of a compound refers to the amount of the therapeutic agent when used alone or in combination with other agents, which provides preventive benefits in the process of preventing diseases, disorders or conditions.
  • the term “prophylactically effective amount” may include an amount that improves overall prevention, or an amount that enhances the preventive effect of other preventive agents.
  • Combination and related terms refer to the simultaneous or sequential administration of the compound of the present invention and other therapeutic agents.
  • the compound of the present invention can be administered simultaneously or sequentially in separate unit dosage forms with other therapeutic agents, or simultaneously administered with other therapeutic agents in a single unit dosage form.
  • compounds of the present invention refers to the following compounds of formula (I) to formula (VI), pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, Solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof.
  • the present invention relates to a compound of general formula (A), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , Polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CR 5 or N; Y is CR 6 or N; and wherein at least one of X and Y is N;
  • Z is CR # or N
  • R 1 , R 2 , R 3 , R 4 and R # are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; or R 1 and R 2 , R 3 and R 4 are connected to form a bond, C 1-6 alkylene, C 2-6 alkenylene or C 2-6 alkynylene;
  • R 5 and R 6 are independently selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl;
  • R # and R 1 may be connected to form a C 3-5 membered cycloalkyl group or a 3-5 membered heterocyclic group;
  • Ring A is selected from C 6-10 aryl and 5-10 membered heteroaryl
  • Ring B is selected from 5-10 membered heteroaryl groups
  • R* is selected from C 1-6 alkyl, C 1-6 haloalkyl, -L'-NRR', -L'-OR, -L'-C 3-7 cycloalkyl and -L'-4-8 Membered heterocyclic group; wherein L'is independently selected from bond, C 1-6 alkylene, C 2-6 alkenylene, C 2-6 alkynylene, -OC 1-6 alkylene and -NH- C 1-6 alkylene;
  • R b is independently selected from H, halogen, -CN, -L-OR, -LC 1-6 alkyl, -LC 1-6 alkoxy, -LC 3-7 cycloalkane optionally substituted by R** Group, -L-3-8 membered heterocyclic group, -NRR', -C(O)R, -C(O)OR, -C(O)NRR', -OC(O)R', -NRC( O)R', -OC(O)NRR' and -NRC(O)NRR';
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, or R, R'and the nitrogen atom to which they are attached form a 4-8 membered heterocyclic group;
  • n 0, 1, 2, 3, 4 or 5;
  • n 0, 1, 2, 3, 4, or 5.
  • the present invention relates to a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate Substances, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N;
  • Y is CH or N; and wherein at least one of X and Y is N;
  • R 1 , R 2 , R 3 and R 4 are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; or R 1 and R 2 , R 3 and R 4 are connected to form a bond, C 1- 6 alkylene, C 2-6 alkenylene or C 2-6 alkynylene;
  • Ring A is selected from C 6-10 aryl and 5-10 membered heteroaryl
  • Ring B is selected from 5-10 membered heteroaryl groups
  • R b is independently selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group , -NRR', -C(O)R, -C(O)OR, -C(O)NRR', -OC(O)R', -NRC(O)R', -OC(O)NRR' And -NRC(O)NRR';
  • n 0, 1, 2, 3, 4 or 5;
  • n 0, 1, 2, 3, 4 or 5;
  • L is independently selected from bond, -O-, -S-, -NR-, -C(O)-, C 1-6 alkylene, C 2-6 alkenylene and C 2-6 alkynylene ;
  • R* is selected from C 1-6 alkyl, C 1-6 haloalkyl, -L'-NRR', -L'-OR, -L'-C 3-7 cycloalkyl and -L'-4-8 Membered heterocyclic group;
  • L' is independently selected from bond, C 1-6 alkylene, C 2-6 alkenylene, C 2-6 alkynylene, -OC 1-6 alkylene and -NH-C 1-6 alkylene base;
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl, or R, R'and the nitrogen atom to which they are attached form a 4-8 membered heterocyclic group;
  • X is CR 5 and Y is N; in another specific embodiment, X is CH and Y is N; in another specific embodiment, X is N, and Y is CR 6 ; In another specific embodiment, X is N, and Y is CH; in another specific embodiment, X is N, and Y is N.
  • Z is CR # ; in another specific embodiment, Z is N.
  • R 1 is H; in another specific embodiment, R 1 is C 1-6 alkyl, such as (R)-C 1-6 alkyl, such as (R)-methyl; In another specific embodiment, R 1 is C 1-6 haloalkyl, such as (R)-C 1-6 haloalkyl.
  • R 2 is H; in another specific embodiment, R 2 is C 1-6 alkyl, such as (R)-C 1-6 alkyl, such as (R)-methyl; In another specific embodiment, R 2 is C 1-6 haloalkyl, such as (R)-C 1-6 haloalkyl.
  • R 3 is H; in another specific embodiment, R 3 is C 1-6 alkyl, such as (R)-C 1-6 alkyl, such as (R)-methyl; In another specific embodiment, R 3 is C 1-6 haloalkyl, such as (R)-C 1-6 haloalkyl. In a specific embodiment, R 4 is H; in another specific embodiment, R 4 is C 1-6 alkyl, such as (R)-C 1-6 alkyl, such as (R)-methyl; In another specific embodiment, R 4 is C 1-6 haloalkyl, such as (R)-C 1-6 haloalkyl.
  • R # is H; in another specific embodiment, R # is C 1-6 alkyl, such as (R)-C 1-6 alkyl, such as (R)-methyl; In another specific embodiment, R # is C 1-6 haloalkyl, such as (R)-C 1-6 haloalkyl.
  • At least one of R 1 and R 2 is C 1-6 alkyl, such as (R)-C 1-6 alkyl, such as (R)-methyl.
  • R 1 and R 2 , or R 3 and R 4 are connected to form a bond; in another specific embodiment, R 1 and R 2 , or R 3 and R 4 are connected to form C 1-6 Alkylene, such as methylene, ethylene or propylene; in another embodiment, R 1 and R 2 , or R 3 and R 4 are connected to form a C 2-6 alkenylene group; in another In a specific embodiment, R 1 and R 2 , or R 3 and R 4 are connected to form a C 2-6 alkynylene group.
  • ring A is a C 6-10 aryl group, such as phenyl; in another specific embodiment, ring A is a 5-10 membered heteroaryl group, such as a 5-6 membered heteroaryl group.
  • ring A is selected from In another specific embodiment, ring A is selected from In another specific embodiment, ring A is selected from In another specific embodiment, ring A is selected from Where R a1 -R a11 are as defined in the context.
  • ring B is a 5-10 membered heteroaryl group, such as a bicyclic 9-10 membered heteroaryl group; in another specific embodiment, ring B is Indicates the position of attachment to the rest of the molecule.
  • ring B is selected from In another specific embodiment, ring B is selected from In another specific embodiment, ring B is selected from In another specific embodiment, ring B is ring B is
  • Z 1 to Z 12 are N atoms; preferably, at least one of Z 1 to Z 12 is a N atom; in another above specific embodiment, Z 1 -Z 3 contains at most one N atom; in another above-mentioned specific embodiment, Z 4 -Z 6 contains at most two N atoms; in another above-mentioned specific embodiment, Z 4 is an N atom.
  • R b is independently selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -NRR' and -NRC(O)R'; In another specific embodiment, one of R b is H; in another specific embodiment, one of R b is halogen; in another specific embodiment, one of R b is -CN; in another specific embodiment In an embodiment, one of R b is -L-OR; in another specific embodiment, one of R b is -OH; in another specific embodiment, one of R b is -LC 1-6 alkyl ; In another specific embodiment, one of R b is C 1-6 alkyl; in another specific embodiment, one of R b is -LC 1-6 alkoxy; in another specific embodiment wherein R b is a C 1-6 alkoxy group; in another particular embodiment, wherein R b is a -LC 3-7 cycloalkyl; in another particular embodiment, wherein R b is a - L
  • L is independently selected from bond, -NR-, -C(O)- and C 1-6 alkylene; in another specific embodiment, L is a bond; in another specific embodiment In the scheme, L is the bond -O-; in another specific embodiment, L is the bond -S-; in another specific embodiment, L is the bond -NR-; in another specific embodiment, L is Bond -C(O)-; in another specific embodiment, L is a bond C 1-6 alkylene; in another specific embodiment, L is a bond C 2-6 alkenylene; in another specific embodiment In an embodiment, L is a bond C 2-6 alkynylene.
  • R* is C 1-6 alkyl; in another specific embodiment, R* is C 1-6 haloalkyl; in another specific embodiment, R* is -L'- NRR'; in another specific embodiment, R* is -L'-OR; in another specific embodiment, R* is -L'-C 3-7 cycloalkyl; in another specific embodiment , R* is -L'-4-8 membered heterocyclic group.
  • any technical solution or any combination of any of the above specific embodiments can be combined with any technical solution or any combination of other specific embodiments.
  • any one aspect or any combination of X may be Y, R 1 -R 4, Ring A, Ring B, R a, R b, R *, R **, L, L ', m, n , P, R and R', etc., or any combination thereof.
  • the present invention is intended to include all the combinations of these technical solutions, limited to space, and will not be listed one by one.
  • the present invention provides a compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate thereof , Hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Y, ring A, ring B, R b and n are as defined above.
  • the present invention provides a compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Y is CH or N
  • Ring A is selected from:
  • R a1 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a2 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a3 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a9 is selected from H and -NRC(O)NRR', wherein R and R'are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl;
  • R a10 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R a11 is selected from H and -NRR', wherein R and R'are independently selected from H, C 1-6 alkyl, and C 1-6 haloalkyl;
  • R b is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 haloalkyl
  • n 0, 1, 2, 3, or 4.
  • the present invention provides a compound of general formula (II), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is selected from:
  • R a1 is H or methyl
  • R a2 is methyl or isopropyl
  • R a3 is H or methyl
  • R a4 is H or -SO 2 Me
  • R a9 is H or -NH 2 C(O)NH 2 Et;
  • R a10 is CF 2 H or C 1-6 alkoxy
  • R a11 is H
  • R b is H or methyl
  • n 0, 1, or 2.
  • the present invention provides a compound of general formula (III), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Y, ring A, R a and m are as defined above.
  • the present invention provides a compound of general formula (III), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is selected from:
  • R a1 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a2 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a3 is selected from C 1-6 alkyl and C 1-6 haloalkyl.
  • the present invention provides a compound of general formula (III), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is selected from:
  • R a1 is methyl
  • R a2 is methyl or isopropyl
  • R a3 is methyl
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • Ring A is selected from:
  • R a1 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a3 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a5 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R a6 is H
  • R a7 is H
  • R a8 is selected from C 1-6 alkyl, C 1-6 haloalkyl and C 3-7 cycloalkyl;
  • R a10 is selected from C 1-6 alkyl and C 1-6 haloalkyl optionally substituted by -CN;
  • R a11 is H
  • R b is selected from hydrogen, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR', -C(O)R;
  • n 0, 1, 2, or 3.
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • Ring A is selected from:
  • R a1 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a2 is selected from C 1-6 alkyl, C 1-6 haloalkyl, -L-NRR', -L-OR and -L-4-8 membered heterocyclyl, where L is a bond or C 1-6 Alkyl, R and R'are independently H, C 1-6 alkyl or C 1-6 haloalkyl;
  • R a3 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a5 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R a6 is H
  • R a7 is H
  • R a8 is selected from C 1-6 alkyl, C 1-6 haloalkyl and C 3-7 cycloalkyl;
  • R a10 is selected from C 1-6 alkyl and C 1-6 haloalkyl optionally substituted by -CN;
  • R a11 is H
  • R b is selected from hydrogen, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR', -C(O)R;
  • n 0, 1, 2, or 3.
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • Ring A is selected from:
  • R a1 is H or methyl
  • R a2 is methyl, isopropyl, -CH 2 CH 2 CH 2 OH, -CH 2 CH 2 OMe, -CH 2 CH 2 NMe 2 , -CH 2 CH 2 CH 2 NMe 2 , azetidine- 3-ylmethyl, oxetan-3-ylmethyl, pyran-4-yl, tetrahydropyrrolyl or piperidinyl;
  • R a3 is H, methyl or CF 3 ;
  • R a4 is -SO 2 N(Me) 2 , -SO 2 Me, -SO 2 iPr or -SO 2 -piperidin-4-yl;
  • R a5 is methyl, methoxy, isopropyl or trifluoromethyl
  • R a6 is H
  • R a7 is H
  • R a8 is Me
  • R a10 is selected from isopropyl optionally substituted by -CN;
  • R a11 is H
  • R b is hydrogen, halogen, methoxy or -CH 2 -azetidin-3-yl
  • n 0, 1, 2, or 3.
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • Ring A is selected from:
  • R a1 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a3 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a5 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R a10 is selected from C 1-6 alkyl and C 1-6 haloalkyl optionally substituted by -CN;
  • R a11 is H
  • R b is selected from hydrogen, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR', -C(O)R;
  • n 0, 1, 2, or 3.
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a1 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a2 is selected from C 1-6 alkyl, C 1-6 haloalkyl and -L-4-8 membered heterocyclic group;
  • R a3 is selected from H, C 1-6 alkyl and C 1-6 haloalkyl
  • R a5 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R a10 is selected from C 1-6 alkyl and C 1-6 haloalkyl optionally substituted by -CN;
  • R a11 is H
  • R b is selected from hydrogen, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR', -C(O)R;
  • L is selected from bond and C 1-6 alkylene
  • n 0, 1, 2, or 3.
  • the present invention provides a compound of general formula (IV), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a1 is methyl
  • R a2 is -CH 2 CH 2 CH 2 OH, -CH 2 CH 2 NMe 2 , -CH 2 CH 2 CH 2 NMe 2 , -CH 2 CH 2 OMe, azetidine-3-ylmethyl, oxygen Etidine-3-ylmethyl, pyran-4-yl, tetrahydropyrrolyl or piperidinyl;
  • R a3 is H, methyl, CF 3 ;
  • R a4 is -SO 2 N(Me) 2 , -SO 2 Me, -SO 2 iPr or -SO 2 -piperidin-4-yl;
  • R a5 is methyl, isopropyl, methoxy or trifluoromethyl
  • R b is hydrogen, halogen, methoxy or -CH 2 -azetidin-3-yl
  • n 0, 1, 2, or 3.
  • the present invention provides a compound of general formula (V), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a1 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a3 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R b is selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR' and -NRC(O)R';
  • n 0, 1, 2, 3, 4 or 5;
  • R* is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • L is selected from bond, -O-, -S-, -NR-, -C(O)- or C 1-6 alkylene;
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl.
  • the present invention provides a compound of general formula (V), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a1 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • R a2 is selected from C 1-6 alkyl, C 1-6 haloalkyl and -L-4-8 membered heterocyclic group;
  • R a3 is selected from C 1-6 alkyl and C 1-6 haloalkyl
  • Ring B is selected from:
  • R b is selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR' and -NRC(O)R', wherein R and R'are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl;
  • L is selected from bond and C 1-6 alkylene
  • n 0, 1, 2, 3, 4, or 5.
  • the present invention provides a compound of general formula (V), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a1 is methyl
  • R a2 is methyl, isopropyl or piperidinyl
  • R a3 is methyl or trifluoromethyl
  • Ring B is selected from:
  • R b is selected from H, halogen, methyl, methoxy, -CH 2 -azetidin-3-yl, -NHMe and -NHC(O)Me;
  • n 0, 1, or 2.
  • the present invention provides a compound of general formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a5 is selected from methyl, methoxy, isopropyl or trifluoromethyl
  • Ring B is selected from:
  • R b is selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR' and -NRC(O)R', wherein R and R'are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl;
  • n 0, 1, 2, 3, 4, or 5.
  • the present invention provides a compound of general formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a4 is selected from -S(O) 2 R*, wherein R* is selected from C 1-6 alkyl, -L'-NRR' and 4-8 membered heterocyclyl, and wherein R and R'are independently selected
  • a 4-8 membered heterocyclic group is formed from H, C 1-6 alkyl and C 1-6 haloalkyl or R, R'and the nitrogen atom to which they are connected;
  • L' is selected from bond and C 1-6 alkylene;
  • R a5 is selected from methyl, methoxy, isopropyl or trifluoromethyl
  • Ring B is selected from:
  • R b is selected from H, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group,- NRR' and -NRC(O)R', wherein R and R'are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl;
  • L is selected from bond and C 1-6 alkylene
  • n 0, 1, or 2.
  • the present invention provides a compound of general formula (VI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X is CH or N
  • R a4 is -SO 2 N(Me) 2 , -SO 2 Me, -SO 2 iPr or -SO 2 -piperidin-4-yl;
  • R a5 is selected from methyl, methoxy, isopropyl or trifluoromethyl
  • Ring B is selected from:
  • R b is selected from H, halogen, methyl, methoxy, -CH 2 -azetidin-3-yl, -NHMe and -NHC(O)Me;
  • n 0, 1, or 2.
  • the present invention provides a compound of general formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is substituted by R a12 is a C 3-7 cycloalkyl group or a 4-8 membered heterocyclic group (preferably R a12 substituted 5-6 membered heterocyclyl, preferably substituted with R a12 N atom-containing 5- 6-membered heterocyclic group);
  • R a1 is C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy or C 1-6 haloalkoxy (preferably methyl, trifluoromethyl or methoxy);
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a12 is H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 5 is H, halogen, C 1-6 alkyl or C 1-6 haloalkyl
  • Z 1 is N or CR b1 ;
  • Z 2 is N or CR b2 ;
  • Z 3 is N or CR b3 ;
  • Z 4 is NR b4 or C(R b4 ) 2 ;
  • Z 5 is N or CR b5 ;
  • Z 6 is N or CR b6 ;
  • R b1 , R b2 , R b3 , R b4 , R b5 and R b6 are independently selected from H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and C 1- 6 Haloalkoxy (preferably selected from H, halogen and methoxy);
  • At least one of R a1 and R a3 is a C 1-6 haloalkyl group.
  • the present invention provides a compound of general formula (VII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is substituted by R a12 is a C 3-7 cycloalkyl group or a 4-8 membered heterocyclic group (preferably R a12 substituted 5-6 membered heterocyclyl, preferably substituted with R a12 N atom-containing 5- 6-membered heterocyclic group);
  • R a1 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a12 is H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 5 is H, halogen, C 1-6 alkyl or C 1-6 haloalkyl
  • Z 1 is N or CR b1 ;
  • Z 2 is N or CR b2 ;
  • Z 3 is N or CR b3 ;
  • Z 4 is NR b4 or C(R b4 ) 2 ;
  • Z 5 is N or CR b5 ;
  • Z 6 is N or CR b6 ;
  • R b2 is H or halogen (preferably H or F);
  • R b3 is H
  • R b1 , R b4 , R b5 and R b6 are independently selected from H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy (preferably Ground is selected from H, halogen and methoxy);
  • At least one of R a1 and R a3 is a C 1-6 haloalkyl group.
  • the present invention provides a compound of general formula (VIII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is The connection point with the rest of the molecule is at one of the positions shown by *;
  • R a1 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a12 is H or C 1-6 alkyl (preferably H or methyl);
  • R 5 is H or halogen (preferably H or F);
  • Ring B is selected from
  • R b is H, methoxy or halogen (preferably H or halogen);
  • n 0, 1, 2 or 3;
  • At least one of R a1 and R a3 is a C 1-6 haloalkyl group.
  • the present invention provides a compound of general formula (VIII), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is The connection point with the rest of the molecule is at one of the positions shown by *;
  • R a1 is methyl or trifluoromethyl
  • R a3 is methyl or trifluoromethyl
  • R a12 is H or methyl
  • R 5 is H or F
  • Ring B is selected from
  • R b is H
  • n 0, 1, 2 or 3;
  • At least one of R a1 and R a3 is a C 1-6 haloalkyl group.
  • the present invention provides a compound of general formula (IX), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is substituted by R a12 is a C 3-7 cycloalkyl group or a 4-8 membered heterocyclic group (preferably R a12 substituted 5-6 membered heterocyclyl, preferably substituted with R a12 N atom-containing 5- 6-membered heterocyclic group);
  • R a1 is C 1-6 alkyl or C 1-6 haloalkyl
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl
  • R a12 is H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 5 is H, halogen, C 1-6 alkyl or C 1-6 haloalkyl
  • Z 7 is N or CR b7 ;
  • Z 8 is N or CR b8 ;
  • Z 9 is N or CR b9 ;
  • Z 10 is N or CR b10 ;
  • Z 11 is N or CR b11 ;
  • Z 12 is N or CR b12 ;
  • R b7 is H, -L-OR, -LC 1-6 alkyl, -LC 3-7 cycloalkyl, -NRR', -NRC(O)R' or -NRC(O)NRR';
  • R b8 , R b9 , R b10 , R b11 and R b12 are independently selected from H, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and C 1- 6 Haloalkoxy (preferably selected from H, halogen, -CN and methoxy);
  • L is independently selected from bond, -O-, -S-, -NR-, -C(O)-, C 1-6 alkylene, C 2-6 alkenylene and C 2-6 alkynylene;
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl.
  • the present invention provides a compound of general formula (IX), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is substituted by R a12 is a C 3-7 cycloalkyl group or a 4-8 membered heterocyclic group (preferably R a12 substituted 5-6 membered heterocyclyl, preferably substituted with R a12 N atom-containing 5- 6-membered heterocyclic group);
  • R a1 is C 1-6 alkyl or C 1-6 haloalkyl
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl
  • R a12 is H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 5 is H, halogen, C 1-6 alkyl or C 1-6 haloalkyl
  • Z 7 is N or CR b7 ;
  • Z 8 is N or CR b8 ;
  • Z 9 is N or CR b9 ;
  • Z 10 is N or CR b10 ;
  • Z 11 is N or CR b11 ;
  • Z 12 is N or CR b12 ;
  • R b7 is selected from -NRR', -NRC(O)R' or -NRC(O)NRR';
  • R b8 , R b9 , R b10 , R b11 and R b12 are independently selected from H, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy and C 1- 6 Haloalkoxy (preferably selected from H, halogen, -CN and methoxy);
  • L is independently selected from bond, -O-, -S-, -NR-, -C(O)-, C 1-6 alkylene, C 2-6 alkenylene and C 2-6 alkynylene;
  • R and R' are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl.
  • the present invention provides a compound of general formula (X), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is The connection point with the rest of the molecule is at one of the positions shown by *;
  • R a1 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a12 is H or C 1-6 alkyl (preferably H or methyl);
  • R 5 is H
  • R b7 is selected from -NRR', -NRC(O)R' or -NRC(O)NRR'; wherein R and R'are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl ( Preferably, R b7 is selected from NH 2 , NHMe, NHEt, NHC(O)Me or NHC(O)Et);
  • R b is H or halogen (preferably H or F).
  • the present invention provides a compound of general formula (X), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is The connection point with the rest of the molecule is at one of the positions shown by *;
  • R a1 is methyl or trifluoromethyl
  • R a3 is methyl or trifluoromethyl
  • R a12 is H or methyl
  • R 5 is H
  • R b7 is selected from NH 2 , NHMe or NHC(O)Me;
  • R b is H or F.
  • the present invention provides a compound of general formula (XI), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvent Complexes, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring C is substituted by R a12 is a C 3-7 cycloalkyl group or a 4-8 membered heterocyclic group (preferably R a12 substituted 5-6 membered heterocyclyl, preferably substituted with R a12 N atom-containing 5- 6-membered heterocyclic group);
  • R a1 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a3 is C 1-6 alkyl or C 1-6 haloalkyl (preferably methyl or trifluoromethyl);
  • R a12 is H, C 1-6 alkyl or C 1-6 haloalkyl (preferably H or methyl);
  • R 5 is H, halogen, C 1-6 alkyl or C 1-6 haloalkyl (preferably H or F);
  • Ring B is selected from:
  • R b is selected from H, halogen, -CN, -LC 1-6 alkyl, C 1-6 alkoxy, -LC 3-7 cycloalkyl, -L-3-8 membered heterocyclic group, -NRR' , -NRC(O)R' and -NRC(O)NRR', wherein R and R'are independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl.
  • Preferred compounds of the present invention include but are not limited to the compounds listed below, or their pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs Types, prodrugs or isotopic variants, and their mixtures:
  • the compounds of the present invention may include one or more asymmetric centers, and therefore may exist in various stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the present invention may be individual enantiomers, diastereomers, or geometric isomers (such as cis and trans isomers), or may be in the form of a mixture of stereoisomers, Including racemate mixtures and mixtures rich in one or more stereoisomers.
  • Isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and formation and crystallization of chiral salts; or preferred isomers can be Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • an organic compound can form a complex with a solvent, which reacts in the solvent or precipitates or crystallizes out of the solvent. These complexes are called “solvates”. When the solvent is water, the complex is called “hydrate”. The present invention covers all solvates of the compounds of the present invention.
  • solvate refers to a compound or a salt form thereof combined with a solvent, which is usually formed by a solvolysis reaction. This physical association may include hydrogen bonding.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether and the like.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be capable of separation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid.
  • “Solvate” includes solvates in solution and isolatable solvates. Representative solvates include hydrates, ethanolates and methanolates.
  • hydrate refers to a compound that binds to water. Generally, the ratio of the number of water molecules contained in the hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Therefore, a hydrate of a compound can be represented by, for example, the general formula R ⁇ x H 2 O, where R is the compound, and x is a number greater than zero.
  • a given compound can form more than one type of hydrate, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5 H 2 O)) and polyhydrates (x is a number greater than 1, for example, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • monohydrate x is 1
  • lower hydrate x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5 H 2 O)
  • polyhydrates x is a number greater than 1, for example, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • the compounds of the invention may be in amorphous or crystalline form (polymorphs).
  • the compounds of the present invention may exist in one or more crystalline forms. Therefore, the present invention includes all amorphous or crystalline forms of the compound of the present invention within its scope.
  • polymorph refers to a crystalline form (or a salt, hydrate or solvate thereof) of a compound in a specific crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, photoelectric properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature and other factors can cause one crystalline form to dominate.
  • Various polymorphs of the compound can be prepared by crystallization under different conditions.
  • the present invention also includes isotopically-labeled compounds, which are equivalent to those described in formula (I), but one or more atoms are replaced by atoms having atomic mass or mass number different from those commonly found in nature.
  • isotopes that can be introduced into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • Isotopically-labeled compounds of formula (I) of the present invention and their prodrugs can generally be prepared in this way.
  • easily available isotope-labeled reagents are used instead of non-isotopes. Labeled reagents.
  • prodrugs are also included in the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted into its active form with medical effects by, for example, hydrolysis in the blood in the body.
  • the pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, ASSymposium Series Vol. 14, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: solubility limits overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each introduced This article serves as a reference.
  • a prodrug is any covalently bonded compound of the invention, and when such a prodrug is administered to a patient, it releases the parent compound in the body.
  • Prodrugs are usually prepared by modifying functional groups, and the modification is performed in such a way that the modification can be performed by conventional operations or cleavage in vivo to produce the parent compound.
  • Prodrugs include, for example, the compounds of the present invention in which a hydroxyl, amino, or sulfhydryl group is bonded to any group, which can be cleaved to form a hydroxyl, amino, or sulfhydryl group when administered to a patient.
  • prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of the hydroxyl, sulfhydryl, and amino functional groups of the compound of formula (I).
  • esters such as methyl and ethyl esters can be used.
  • the ester itself can be active and/or can be hydrolyzed under human body conditions.
  • Suitable pharmaceutically acceptable in vivo hydrolyzable ester groups include those groups that are easily decomposed in the human body to release the parent acid or salt thereof.
  • the present invention also provides a pharmaceutical preparation comprising a therapeutically effective amount of a compound of formula (I) or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent or excipient. All these forms belong to the present invention.
  • compositions preparations and kits
  • the invention provides a pharmaceutical composition comprising a compound of the invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of a compound of the invention.
  • the pharmaceutical composition comprises a therapeutically effective amount of a compound of the invention.
  • the pharmaceutical composition comprises a prophylactically effective amount of a compound of the invention.
  • the pharmaceutically acceptable excipient used in the present invention refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that can be used in the composition of the present invention include (but are not limited to) ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum white Protein), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, partial glyceride mixture of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate , Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene- Block polymers, polyethylene glycol and
  • kits e.g., pharmaceutical packaging.
  • the kit provided may include the compound of the present invention, other therapeutic agents, and the first and second containers (for example, vials, ampoules, bottles, syringes, and/or dispersible packages or other Suitable container).
  • the provided kits may also optionally include a third container, which contains pharmaceutical excipients for diluting or suspending the compound of the present invention and/or other therapeutic agents.
  • the compound of the invention and the other therapeutic agent provided in the first container and the second container are combined to form a unit dosage form.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration , Intracerebrospinal drug delivery, intralesional drug delivery, and intracranial injection or infusion technology.
  • an effective amount of a compound provided herein is administered.
  • the doctor can determine the amount of the compound actually administered .
  • the compounds provided herein are administered to subjects at risk of developing the conditions, typically based on the doctor's advice and under the supervision of the doctor, and the dosage levels are as described above.
  • Subjects at risk of developing a particular disorder generally include subjects with a family history of the disorder, or those subjects who are particularly sensitive to the development of the disorder as determined by genetic testing or screening.
  • the pharmaceutical compositions provided herein can also be administered chronically ("long-term administration").
  • Long-term administration refers to the administration of the compound or its pharmaceutical composition over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or the administration can be continued indefinitely, For example, the rest of the subject's life.
  • chronic administration is intended to provide a constant level of the compound in the blood over a long period of time, for example, within a therapeutic window.
  • the pharmaceutical composition may be administered as a bolus, for example, to increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic level of the active ingredient through the body. For example, an intramuscular or subcutaneous bolus dose allows the active ingredient to be released slowly, while a bolus injection delivered directly to a vein (for example, by IV intravenous infusion) ) Can be delivered more quickly, so that the concentration of the active component in the blood quickly rises to an effective level.
  • the pharmaceutical composition may be administered as a continuous infusion, for example, by IV infusion, to provide a steady-state concentration of the active ingredient in the subject's body.
  • a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
  • Oral compositions can take the form of bulk liquid solutions or suspensions or bulk powders. However, more generally, in order to facilitate precise dosing, the composition is provided in unit dosage form.
  • unit dosage form refers to a physically discrete unit suitable as a unit dosage for human patients and other mammals, each unit containing a predetermined number of active substances suitable for producing the desired therapeutic effect and suitable pharmaceutical excipients.
  • Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or pills, tablets, capsules, etc. in the case of solid compositions.
  • the compound is usually a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), and the remaining part is useful for forming the desired administration form.
  • Kinds of carriers or excipients and processing aids are used for forming the desired administration form.
  • the representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides about 0.01 to about 20 mg/kg of the compound of the present invention, with preferred doses each providing about 0.1 to about 10 mg/kg, especially about 1 to about 5 mg/kg.
  • the transdermal dose is usually selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, preferably about 0.1 To about 10% by weight, and more preferably about 0.5 to about 15% by weight.
  • the injection dose level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more can also be given.
  • the maximum total dose cannot exceed approximately 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous carriers as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • the solid form may include, for example, any of the following components, or compounds with similar properties: binders, for example, microcrystalline cellulose, tragacanth, or gelatin; excipients, for example, starch or lactose, disintegrants, For example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, for example, colloidal silicon dioxide; sweeteners, for example, sucrose or saccharin; or flavoring agents, for example, mint, water Methyl salicylate or orange flavoring agent.
  • binders for example, microcrystalline cellulose, tragacanth, or gelatin
  • excipients for example, starch or lactose, disintegrants, For example, alginic acid, Primogel or corn starch
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • the transdermal composition is typically formulated as a topical ointment or cream containing the active ingredient.
  • the active ingredient When formulated as an ointment, the active ingredient is typically combined with paraffin or a water-miscible ointment base.
  • the active ingredient can be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art, and usually include other components for enhancing the active ingredient or stable skin penetration of the formulation. All such known transdermal preparations and components are included within the scope provided by the present invention.
  • transdermal administration can be achieved using a reservoir or porous membrane type, or a variety of solid matrix patches.
  • compositions for oral administration, injection or topical administration are only representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, Part 8, which is incorporated herein by reference.
  • the compounds of the invention can also be administered in a sustained release form or from a sustained release drug delivery system.
  • Descriptions of representative sustained-release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention also relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation contains water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are ⁇ -, ⁇ - and ⁇ -cyclodextrins composed of 6, 7 and 8 ⁇ -1,4-linked glucose units, respectively, which optionally include one on the linked sugar moiety Or multiple substituents, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substituted.
  • the cyclodextrin is a sulfoalkyl ether ⁇ -cyclodextrin, for example, sulfobutyl ether ⁇ -cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation includes hexapropyl- ⁇ -cyclodextrin (e.g., 10-50% in water).
  • ATR kinase As described herein, ATR kinase is known to play a role in tumorigenesis and many other diseases. We have found that the compound of formula (I) has potent anti-tumor activity, which is believed to be obtained by inhibiting ATR kinase.
  • the compound of the present invention has value as an antitumor agent.
  • the compounds of the present invention have the value as anti-proliferation, apoptosis and/or anti-invasive agents in the suppression and/or treatment of solid and/or liquid tumor diseases.
  • the compounds of the present invention are expected to be useful in the prevention or treatment of those tumors that are sensitive to inhibition of ATR.
  • the compounds of the present invention are expected to be useful in the prevention or treatment of those tumors mediated solely or in part by ATR. Therefore, the compounds can be used to produce ATR enzyme inhibition in warm-blooded animals in need of such treatment.
  • inhibitors of ATR kinase should be used for the treatment of proliferative diseases (such as cancer), especially solid tumors (such as cancer and sarcoma) and leukemia and lymphoma, especially for breast cancer, colorectal cancer, lung cancer (including Small cell lung cancer, non-small cell lung cancer and bronchioloalveolar cancer) and prostate cancer and cholangiocarcinoma, bone cancer, bladder cancer, head and neck cancer, kidney cancer, liver cancer, gastrointestinal tissue cancer, esophagus cancer, ovarian cancer, pancreatic cancer , Skin cancer, testicular cancer, thyroid cancer, uterine cancer, cervical cancer and vulvar cancer, as well as leukemia [including acute lymphocytic leukemia (ALL) and chronic myeloid leukemia (CML)], multiple myeloma and lymphoma
  • proliferative diseases such as cancer
  • solid tumors such as cancer and sarcoma
  • leukemia and lymphoma especially for breast cancer, colorectal cancer
  • Anticancer effects useful for treating cancer in patients include, but are not limited to, antitumor effects, response rate, time to disease progression, and survival rate.
  • the anti-tumor effect of the treatment method of the present invention includes, but is not limited to, inhibition of tumor growth, delay of tumor growth, tumor degeneration, tumor shrinkage, prolonged tumor regeneration after treatment is stopped, and slowing of disease progression.
  • Anti-cancer effects include preventive treatment and treatment of existing diseases.
  • ATR kinase inhibitors or pharmaceutically acceptable salts thereof are also useful in the treatment of cancer patients, including but not limited to blood cancers, such as leukemia and multiple myeloma; lymphomas, such as Hodgkin’s disease and non-Hodgkin’s Lymphoma (including mantle cell lymphoma) and myelodysplastic syndromes, as well as solid tumors and their metastases (metastases), such as breast cancer, lung cancer (non-small cell lung cancer (NSCL), small cell lung cancer (SCLC), Squamous cell carcinoma), endometrial cancer, central nervous system tumors (such as glioma, embryonal dysplastic neuroepithelial tumor, glioblastoma multiforme, mixed glioma, medulloblastoma, Retinoblastoma, neuroblastoma, germ cell tumor and teratoma, gastrointestinal cancer (such as stomach cancer), esophageal cancer, hepatocellular (liver
  • the compound of the present invention and the treatment method comprising the administration or use of an ATR kinase inhibitor or a pharmaceutically acceptable salt thereof are particularly useful for treating patients with lung cancer, prostate cancer, melanoma, ovarian cancer, breast cancer, endometrial cancer, Patients with renal cancer, gastric cancer, sarcoma, head and neck cancer, central nervous system tumors and their metastases, as well as patients with acute myeloid leukemia.
  • the effective amount of the compound of the present invention is usually 0.01 mg to 50 mg of compound per kilogram of patient body weight at an average daily dose, preferably 0.1 mg to 25 mg of compound per kilogram of patient weight, in single or multiple administration.
  • the compound of the present invention can be administered to the patient in need of such treatment at a daily dose ranging from about 1 mg to about 3500 mg per patient, preferably 10 mg to 1000 mg.
  • the daily dose per patient can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000mg. It can be administered once or multiple times daily, weekly (or at intervals of several days), or on an intermittent schedule.
  • the compound can be administered on a weekly basis (e.g., every Monday), one or more times per day, variably or for several weeks, for example 4-10 weeks.
  • it may be administered daily for several days (for example, 2-10 days), and then for several days (for example, 1-30 days) without administering the compound, the cycle may be repeated indefinitely or a given number of times, such as 4-10 Cycles.
  • the compound of the present invention can be administered daily for 5 days, then 9 days intermittently, and then administered daily for 5 days, then 9 days intermittently, and so on, repeating the cycle indefinitely or repeating 4-10 times in total.
  • the treatment defined herein may be applied as a stand-alone treatment, or may include conventional surgery or radiotherapy or chemotherapy in addition to the compounds of the invention. Therefore, the compounds of the present invention can also be used in combination with existing therapeutic agents for the treatment of cancer.
  • Suitable agents for combined use include:
  • Anti-proliferative/anti-tumor drugs and their combinations used in medical oncology such as alkylating agents (such as cisplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, Busulfan (Busulphan and nitrosoureas); antimetabolites (e.g.
  • antifolates such as fluorouracils such as 5-fluorouracil and tegafur, raltitrexed, methamphetamine Chlorin, cytarabine, hydroxyurea and gemcitabine
  • anti-tumor antibiotics e.g., anthracyclines, such as adriamycin, bleomycin, doxorubicin, doxorubicin) Normycin (daunomycin), epirubicin (epirubicin), idarubicin (idarubicin), mitomycin C (mitomycin-C), actinomycin D (dactinomycin) and mithramycin (mithramycin)
  • Anti-mitotic agents for example, vinca alkaloids, such as vincristine, vinblastine, vindesine, and vinorelbine; and taxanes, such as paclitaxel and docetaxel (taxotere)
  • topoisomerase Inhibitors e.g
  • Cell growth inhibitors such as anti-estrogens (such as tamoxifen, toremifene, raloxifene, droloxifene, and idoxifene) (iodoxyfene)), estrogen receptor downregulators (e.g. fulvestrant (fulvestrant)), anti-androgens (e.g. bicalutamide, flutamide, nilutamide, and Cyproterone acetate), LHRH antagonists or LHRH agonists (e.g. goserelin, leuprorelin, and buserelin), progestins (e.g.
  • anti-estrogens such as tamoxifen, toremifene, raloxifene, droloxifene, and idoxifene
  • estrogen receptor downregulators e.g. fulvestrant (fulvestrant)
  • anti-androgens e.g. bicalutamide,
  • Aromatase inhibitors such as Anastrozole, letrozole, vorazole and exemestane
  • 5 ⁇ -reductase inhibitors such as finastere Amine (finasteride);
  • Anti-invasive agents such as c-Src kinase family inhibitors, such as AZD0530 and dasatinib) and metalloproteinase inhibitors, such as marimastat; and urokinase-type plasminogen activator Inhibitors of body function;
  • Growth factor function inhibitors such as growth factor antibodies and growth factor receptor antibodies (eg anti-erbB2 antibody trastuzumab [Herceptin TM ] and anti-erbBl antibody cetuximab [C225]); such inhibition Agents also include, for example, tyrosine kinase inhibitors, such as inhibitors of the epidermal growth factor family (e.g., EGFR family tyrosine kinase inhibitors, such as gefitinib, erlotinib and CI 1033; and erbB2 tyrosine kinase Inhibitors, such as lapatinib; inhibitors of the hepatocyte growth factor family; inhibitors of the platelet-derived growth factor family, such as imatinib; inhibitors of serine/threonine kinases (such as Ras/Raf signaling Inhibitors, such as farnesyltransferase inhibitors, such as sorafenib (BAY 43
  • Anti-angiogenic agents such as those that inhibit the effect of vascular endothelial growth factor [for example, the anti-vascular endothelial growth factor antibody bevacizumab (Avastin TM ); and VEGF receptor tyrosine kinase inhibitors, such as ZD6474, AZD2171, vatalanib and sunitinib, and compounds that act by other mechanisms (such as linomide, integrin ⁇ v ⁇ 3 function inhibitors and angiostatin) ];
  • vascular endothelial growth factor for example, the anti-vascular endothelial growth factor antibody bevacizumab (Avastin TM ); and VEGF receptor tyrosine kinase inhibitors, such as ZD6474, AZD2171, vatalanib and sunitinib, and compounds that act by other mechanisms (such as linomide, integrin ⁇ v ⁇ 3 function inhibitors and angiostatin)
  • Vascular disruptors such as combretastatin (combretastatin);
  • Antisense therapy such as those targeting the targets listed above, such as ISIS 2503;
  • Gene therapy methods including methods to replace abnormal genes (such as abnormal p53 or abnormal BRCA1 or BRCA2); GDEPT (gene-directed enzyme prodrug therapy) methods, such as the use of cytosine deaminase, breast cancer Those methods of glycoside kinase or bacterial nitroreductase; and methods to improve the tolerance of patients to chemotherapy or radiotherapy, such as multidrug resistance gene therapy; and
  • Immunotherapy methods including in vitro and in vivo methods to improve the immunogenicity of patients’ tumor cells, such as transfection with cytokines (such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor); reducing T -Anergy method; a method using transfected immune cells (such as cytokine-transfected dendritic cells); a method using cytokine-transfected tumor cell lines and a method using anti-idiotypic antibodies.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • T -Anergy method a method using transfected immune cells (such as cytokine-transfected dendritic cells)
  • cytokine-transfected tumor cell lines such as anti-idiotypic antibodies.
  • the intermediate b8-2 (8.24mmol, 1.5g) was dissolved in 30mL of dichloromethane, m-CPBA (20mmol, 3.4g) was slowly added, and the reaction was carried out at room temperature overnight. Add 50 mL of water and extract with dichloromethane to obtain intermediate b8-1 with a quantitative yield.
  • intermediate b8-1 was obtained with a yield of 42%.
  • the intermediate b10-3 (0.91mmol, 300mg) and diboronic acid pinacol ester (1.09mmol, 276mg) were dissolved in 12mL 1,4-dioxane, and potassium acetate (2.7mmol, 266mg) and Pd(dppf)Cl 2 (0.18mmol, 132mg), react at 100°C for 2 hours. The reaction was stopped, filtered, the organic solvent was evaporated under reduced pressure, and the column chromatography was separated to obtain intermediate b10 (240 mg, yield: 70%).
  • the raw material b13-1 (23.5 mmol, 2.4 g) was dissolved in 15 mL of THF, the raw material p-toluenesulfonyl chloride (7.85 mmol, 1.5 g) was slowly added dropwise, and the reaction was stopped after 2 hours at room temperature. Add 40 mL of water, extract with ethyl acetate, and separate by flash column chromatography to obtain intermediate b13-2 (2.0 g, yield: 99%).
  • the intermediate b16-1 (1.81 mmol, 300 mg) was dissolved in a mixed solution of 10 mL methanol and 2 mL water, potassium monopersulfate Oxone (4.5 mmol, 278 mg) was slowly added, and the reaction was carried out at room temperature for 4 hours. 30 mL of water was added to the reaction solution, extracted with ethyl acetate, and separated by flash column chromatography to obtain intermediate b16-2 (350 mg, yield: 99%).
  • intermediate b16-2 (1.76 mmol, 350 mg) in 3 mL of concentrated sulfuric acid, add N-bromosuccinimide (1.33 mmol, 235 mg) in batches, and continue the reaction at room temperature for 3 hours.
  • the reaction solution was slowly poured into a 30mL ice-water mixture, and the pH was adjusted to about 8 with 1N NaOH solution, extracted with dichloromethane, dried with anhydrous sodium sulfate, and separated by flash column chromatography to obtain intermediate b16-3 (350mg, yield : 95%).
  • the intermediate b16-3 (0.9mmol, 250mg) and diboronic acid pinacol ester (1.08mmol, 276mg) were dissolved in 8mL 1,4-dioxane, and potassium acetate (1.8mmol, 176mg) and Pd(dppf)Cl 2 (0.09mmol, 66mg), react at 100°C for 2 hours. The reaction was stopped, filtered, and the solvent was evaporated under reduced pressure to obtain intermediate b16, which was directly used in the next reaction.
  • the intermediate c8-6 (1.46mmol, 500mg) was dissolved in 15mL of anhydrous THF, and butyllithium (0.7mL, 2.5M) was added dropwise. After 30 minutes, isopropoxyboronic acid was added. Naol ester c3-4 (1.75mmol, 326mg), reacted at -78°C for 3 hours, 50mL saturated aqueous ammonium chloride solution was added to quench the reaction, and extracted with dichloromethane. Dry with anhydrous sodium sulfate to obtain the crude intermediate c8 (500 mg, yield: 87%). LC-MS: [M+H] + : 390.
  • the intermediate c11-2 (0.4mmol, 170mg) was dissolved in 10mL of anhydrous THF, and butyllithium (0.8mL, 2.5M) was added dropwise. After 30 minutes, isopropoxyboronic acid was added. Naol ester c3-4 (1mmol, 200 ⁇ L), reacted at -78°C for 3 hours, 50mL saturated aqueous ammonium chloride was added to quench the reaction, and extracted with dichloromethane. Drying with anhydrous sodium sulfate gave the crude intermediate c11 (100mg, yield: 53%). Used directly in the next reaction. LC-MS: [M+H] + : 474.
  • the intermediate c14-4 (1.3mmol, 500mg), pinacol diborate (1.52mmol, 300mg), DIEA (3.8mmol, 0.66mL) and Pd(Amphos)Cl were added to the microwave reaction flask 2 (0.13 mmol, 90 mg), 20 mL of a mixed solution of 2-Me-THF and MeOH was dissolved (v/v, 1/1). The reaction was stopped after heating to 100°C by microwave for 1 hour. Filter, add 30 mL of water, and extract with ethyl acetate to obtain the crude intermediate c14. Used directly in the next reaction.
  • the intermediate a1 (0.8mmol, 200mg) and the raw material 4-indoleboronic acid pinacol ester d4-1 (0.88mmol, 214mg) were dissolved in a 10mL mixture of 1,4-dioxane and water Medium (v/v: 9/1), potassium carbonate (1.6 mmol, 221 mg) and Pd(dppf)Cl 2 (0.08 mmol, 59 mg) were added, and the mixture was heated to 110° C. to react for 2 hours. Stop the reaction, filter, add 30 mL of water, extract with ethyl acetate, and separate by column chromatography to obtain intermediate d4 (50 mg). Yield: 19%, LC-MS: [M+H] + : 329.
  • the intermediate e1-2 (3.23 mmol, 730 mg) was dissolved in 16 mL of a mixture of methanol/acetonitrile (1/1, v/v), and an aqueous KF solution (749 mg/3 mL) was slowly added. After the system was stirred for 10 minutes, L-tartaric acid (6.46 mmol, 968 mg) and tetrahydrofuran (350 ⁇ L) were added, and the reaction was continued at room temperature for 1.5 hours. The reaction was stopped, filtered, and the filtrate was concentrated to obtain intermediate e1-3 (600 mg, crude product).
  • compound D8-1 (0.16mmol, 70mg) and intermediate c3 (0.32mmol, 150mg) were dissolved in a 5mL mixture of 1,4-dioxane and water (v/v: 9/1 ), potassium carbonate (0.32mmol, 44mg), Pd(dppf)Cl 2 (0.016mmol, 12mg) were added, and the reaction was heated in a microwave at 100°C for 1 hour.
  • compound D23-1 (0.24mmol, 95mg) and intermediate c3 (0.48mmol, 223mg) were dissolved in a 5mL mixture of 1,4-dioxane and water (v/v: 9/1 ), potassium carbonate (0.48mmol, 66mg), Pd(dppf)Cl 2 (0.024mmol, 18mg) were added, and the reaction was heated in a microwave at 100°C for 1.5 hours.
  • compound D27-4 (0.38mmol, 150mg) and intermediate c3 (0.76mmol, 348mg) were dissolved in a 5mL mixture of 1,4-dioxane and water (v/v: 9/1 ), potassium carbonate (1.14mmol, 157mg), Pd(dppf)Cl 2 (0.04mmol, 28mg) were added, and the reaction was heated in a microwave at 100°C for 1 hour.
  • the intermediate D30a-1 was replaced with D30b-1 in the second step to synthesize the target compound D30b.
  • the intermediate D10-1 (0.53mmol, 200mg) and the intermediate c9a/c9b mixture (1.06mmol, 432mg) were dissolved in 10mL of a mixture of 1,4-dioxane and water (v/v : 9/1), potassium carbonate (1.59 mmol, 219 mg) and Pd(dppf)Cl 2 (0.05 mmol, 39 mg) were added, and the temperature was raised to 105° C. for microwave reaction for 1 hour.
  • the intermediate d7 (0.28mmol, 100mg) and the intermediate c9a/c9b mixture (0.56mmol, 227mg) were dissolved in 10mL of 1,4-dioxane and water mixture (v/v, 9 /1), potassium carbonate (0.84mmol, 116mg) and Pd(dppf)Cl 2 (0.03mmol, 20mg) were added, and the temperature was raised to 105°C in microwave for reaction for 1 hour.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

通式(A)化合物,它们可用于治疗ATR激酶介导的疾病,例如增生性疾病,例如癌症。还提供通式(A)化合物的药物组合物、其用于治疗ATR激酶介导的疾病的用途、及其制备。

Description

作为ATR激酶抑制剂的2,4,6-三取代的嘧啶化合物 发明领域
本发明涉及作为ATR激酶的抑制剂的2,4,6-三取代的嘧啶化合物。更具体地,本发明的化合物可有效用于ATR激酶介导的疾病,例如增生性疾病,例如癌症的治疗。本发明还提供所述化合物的药物组合物、其用于治疗ATR激酶介导的疾病的用途、及其制备。
背景技术
ATR(Ataxia telangiectasia and Rad3-related protein)是一类参与基因组稳定性及DNA损伤修复的蛋白激酶,属于PIKK家族成员。ATR的激活可被停滞的复制叉或者DNA单链损伤(SSB)所激活。激活的ATR将招募修复蛋白或修复因子对受损部位进行修复,延缓有丝分裂过程(特别在有丝分裂的G2/M期),既稳定了复制叉,又保障了基因组的稳定性。
此外,大部分肿瘤细胞中的DNA损伤修复系统异常,通常缺失一定的修复通路(如p53或ATM的突变),使得其更加依赖ATR来存活。而在正常细胞中,由于其健全完整的修复通路,单独抑制ATR激酶不会产生较大影响。因此,抑制ATR可能对癌症的治疗具有更显著的效果,而对正常细胞则不会产生较大毒副作用。
而且,ATR的抑制可同时与放疗或化疗药物联用来协同增强效果。广泛应用的化疗药物包括抗代谢药(如吉西他滨),DNA交联剂(如顺铂、卡铂)、烷化剂(如替莫唑胺),拓扑异构酶抑制剂(如拓扑替康、伊立替康)等。肿瘤细胞在受到化疗或放疗的影响时,会较大程度上激活ATR信号通路来修复受损的DNA。因此,利用放疗或化疗药治疗癌症时,同时对ATR进行抑制,可使得癌症的治疗效果大大增强。
到目前为止,仍无ATR抑制剂上市,因此发现更加有效且安全的ATR抑制剂仍十分必要。
发明内容
本发明提供通式(I)化合物,它们可用于治疗ATR激酶介导的疾病,例如增生性疾病,例如癌症。
在一个方面,本发明提供了通式(A)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000001
其中,
X为CR 5或N;Y为CR 6或N;并且其中X和Y中至少有一个为N;
Z为CR #或N;
R 1、R 2、R 3、R 4和R #独立地选自H、C 1-6烷基和C 1-6卤代烷基;或者R 1与R 2,R 3与R 4连接形 成键、C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;
R 5和R 6独立地选自H、卤素、C 1-6烷基或C 1-6卤代烷基;
当Z为CR #时,其中R #与R 1可以连接形成C 3-5元环烷基或3-5元杂环基;
环A选自C 6-10芳基和5-10元杂芳基;
环B选自5-10元杂芳基;
R a独立地选自任选被R**取代的H、卤素、-CN、-L-NRR’、-L-OR、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(=NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基;
其中L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;p=1或2;
R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR、-L’-C 3-7环烷基和-L’-4-8元杂环基;其中L’独立地选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、-O-C 1-6亚烷基和-NH-C 1-6亚烷基;
R b独立地选自任选被R**取代的H、卤素、-CN、-L-OR、-L-C 1-6烷基、-L-C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’和-NRC(O)NRR’;
R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基,或者R、R’与它们连接的氮原子形成4-8元杂环基;
m=0、1、2、3、4或5;
n=0、1、2、3、4或5。
在另一个方面,本发明提供了通式(I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000002
其中,
X为CH或N;Y为CH或N;并且其中X和Y中至少有一个为N;
R 1、R 2、R 3和R 4独立地选自H、C 1-6烷基和C 1-6卤代烷基;或者R 1与R 2,R 3与R 4连接形成键、C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;
环A选自C 6-10芳基和5-10元杂芳基;
环B选自5-10元杂芳基;
R a独立地选自任选被R**取代的H、卤素、-CN、-L-NRR’、-L-OR、-C(O)R、-C(O)OR、-C(O)NRR’、 -OC(O)R’、-NRC(O)R’、-OC(O)NRR’、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(=NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基;
R b独立地选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’和-NRC(O)NRR’;
m=0、1、2、3、4或5;
n=0、1、2、3、4或5;
其中L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;
R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR、-L’-C 3-7环烷基和-L’-4-8元杂环基;
L’独立地选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、-O-C 1-6亚烷基和-NH-C 1-6亚烷基;
R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基,或者R、R’与它们连接的氮原子形成4-8元杂环基;
p=1或2。
在另一个方面,本发明提供了一种药物组合物,所述药物组合物含有本发明化合物,和任选地药学上可接受的赋形剂。
在另一个方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物,其还含有其它治疗剂。
在另一个方面,本发明提供了包含本发明化合物,和其它治疗剂以及药学上可接受的载剂、佐剂或媒剂的试剂盒。
在另一个方面,本发明提供了本发明化合物在制备用于治疗和/或预防ATR激酶介导的疾病的药物中的用途。
在另一个方面,本发明提供了在受试者中治疗和/或预防ATR激酶介导的疾病的方法,包括向所述受试者给药本发明化合物或本发明组合物。
在另一个方面,本发明提供了本发明化合物或本发明组合物,其用于治疗和/或预防ATR激酶介导的疾病。
在具体实施方案中,所述疾病包括增生性疾病(如癌症),尤其实体瘤(如癌和肉瘤)及白血病和淋巴癌,尤其对于例如乳腺癌、结肠直肠癌、肺癌(包括小细胞肺癌、非小细胞肺癌和细支气管肺泡癌)和前列腺癌及胆管癌、骨癌、膀胱癌、头及颈癌、肾癌、肝癌、胃肠组织癌、食道癌、卵巢癌、胰腺癌、皮肤癌、睾丸癌、甲状腺癌、子宫癌、子宫颈癌和外阴癌、以及白血病[包括急性淋巴细胞白血病(ALL)和慢性髓细胞源性白血病(CML)]、多发性骨髓瘤和淋巴癌。
由随后的具体实施方案、实施例和权利要求,本发明的其它目的和优点将对于本领域技术人员显而易见。
定义
化学定义
下面更详细地描述具体官能团和化学术语的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C 1-6烷基”包括C 1、C 2、C 3、C 4、C 5、C 6、C 1-6、C 1-5、C 1-4、C 1-3、C 1-2、C 2-6、C 2-5、C 2-4、C 2-3、C 3-6、C 3-5、C 3-4、C 4-6、C 4-5和C 5-6烷基。
“C 1-6烷基”是指具有1至6个碳原子的直链或支链饱和烃基团。在一些实施方案中,C 1-4烷基是优选的。C 1-6烷基的例子包括:甲基(C 1)、乙基(C 2)、正丙基(C 3)、异丙基(C 3)、正丁基(C 4)、叔丁基(C 4)、仲丁基(C 4)、异丁基(C 4)、正戊基(C 5)、3-戊基(C 5)、戊基(C 5)、新戊基(C 5)、3-甲基-2-丁基(C 5)、叔戊基(C 5)和正己基(C 6)。术语“C 1-6烷基”还包括杂烷基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。常规烷基缩写包括:Me(-CH 3)、Et(-CH 2CH 3)、iPr(-CH(CH 3) 2)、nPr(-CH 2CH 2CH 3)、n-Bu(-CH 2CH 2CH 2CH 3)或i-Bu(-CH 2CH(CH 3) 2)。
“C 2-6烯基”是指具有2至6个碳原子和至少一个碳碳双键的直链或支链烃基团。在一些实施方案中,C 2-4烯基是优选的。C 2-6烯基的例子包括:乙烯基(C 2)、1-丙烯基(C 3)、2-丙烯基(C 3)、1-丁烯基(C 4)、2-丁烯基(C 4)、丁二烯基(C 4)、戊烯基(C 5)、戊二烯基(C 5)、己烯基(C 6),等等。术语“C 2-6烯基”还包括杂烯基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烯基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 2-6炔基”是指具有2至6个碳原子、至少一个碳-碳叁键以及任选地一个或多个碳-碳双键的直链或支链烃基团。在一些实施方案中,C 2-4炔基是优选的。C 2-6炔基的例子包括但不限于:乙炔基(C 2)、1-丙炔基(C 3)、2-丙炔基(C 3)、1-丁炔基(C 4)、2-丁炔基(C 4),戊炔基(C 5)、己炔基(C 6),等等。术语“C 2-6炔基”还包括杂炔基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。炔基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基”指的是上述定义的“C 1-6烷基、C 2-6烯基或C 2-6炔基”的二价基团。
“C 1-6亚烷基”是指除去C 1-6烷基的另一个氢而形成的二价基团,并且可以是取代或未取代的亚烷基。在一些实施方案中,C 1-4亚烷基是特别优选的。未取代的所述亚烷基包括但不限于:亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-)、亚丙基(-CH 2CH 2CH 2-)、亚丁基(-CH 2CH 2CH 2CH 2-)、亚戊基(-CH 2CH 2CH 2CH 2CH 2-)、亚己基(-CH 2CH 2CH 2CH 2CH 2CH 2-),等等。示例性的取代的所述亚烷基,例如,被一个或多个烷基(甲基)取代的所述亚烷基,包括但不限于:取代的亚甲基(-CH(CH 3)-、-C(CH 3) 2-)、取代的亚乙基(-CH(CH 3)CH 2-、-CH 2CH(CH 3)-、-C(CH 3) 2CH 2-、-CH 2C(CH 3) 2-)、取代的亚丙基(-CH(CH 3)CH 2CH 2-、-CH 2CH(CH 3)CH 2-、-CH 2CH 2CH(CH 3)-、-C(CH 3) 2CH 2CH 2-、-CH 2C(CH 3) 2CH 2-、-CH 2CH 2C(CH 3) 2-),等等。
“C 2-6亚烯基”是指除去C 2-6烯基的另一个氢而形成的二价基团,并且可以是取代或未取代的亚烯基。在一些实施方案中,C 2-4亚烯基是特别优选的。示例性的未取代的所述亚烯基包括但不限于:亚乙烯基(-CH=CH-)和亚丙烯基(例如,-CH=CHCH 2-、-CH 2-CH=CH-)。示例性的取代的所述亚烯基,例如,被一个或多个烷基(甲基)取代的亚烯基,包括但不限于:取代的亚乙基(-C(CH 3)=CH-、-CH=C(CH 3)-)、取代的亚丙烯基(-C(CH 3)=CHCH 2-、-CH=C(CH 3)CH 2-、-CH=CHCH(CH 3)-、-CH=CHC(CH 3) 2-、-CH(CH 3)-CH=CH-、-C(CH 3) 2-CH=CH-、-CH 2-C(CH 3)=CH-、-CH 2-CH=C(CH 3)-),等等。
“C 2-6亚炔基”是指除去C 2-6炔基的另一个氢而形成的二价基团,并且可以是取代或未取代的亚炔基。在一些实施方案中,C 2-4亚炔基是特别优选的。示例性的所述亚炔基包括但不限于:亚乙炔基(-C≡C-)、取代或未取代的亚丙炔基(-C≡CCH 2-),等等。
“C 1-6烷氧基”是指基团-OR,其中,R为取代或未取代的C 1-6烷基。在一些实施方案中,C 1-4烷氧基是特别优选的。具体的所述烷氧基包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、叔丁氧基、仲丁氧基、正戊氧基、正己氧基和1,2-二甲基丁氧基。烷氧基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“卤代”或“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。
因此,“C 1-6卤代烷基”和“C 1-6卤代烷氧基”是指上述“C 1-6烷基”和“C 1-6烷氧基”,其被一个或多个卤素基团取代。在一些实施方案中,C 1-4卤代烷基是特别优选的,更优选C 1-2卤代烷基。在一些实施方案中,C 1-4卤代烷氧基是特别优选的,更优选C 1-2卤代烷氧基。示例性的所述卤代烷基包括但不限于:-CF 3、-CH 2F、-CHF 2、-CHFCH 2F、-CH 2CHF 2、-CF 2CF 3、-CCl 3、-CH 2Cl、-CHCl 2、2,2,2-三氟-1,1-二甲基-乙基,等等。示例性的所述卤代烷氧基包括但不限于:-OCH 2F、-OCHF 2、-OCF 3,等等。卤代烷基和卤代烷氧基基团可以在任何可用的连接点上被取代,例如,1至5个取代基、1至3个取代基或1个取代基。
“C 3-7环烷基”是指具有3至7个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C 3-5环烷基是优选的。在另一些实施方案中,C 3-6环烷基是优选的。在另一些实施方案中,C 5-6环烷基是优选的。环烷基还包括其中上述环烷基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基(C 3)、环丙烯基(C 3)、环丁基(C 4)、环丁烯基(C 4)、环戊基(C 5)、环戊烯基(C 5)、环己基(C 6)、环己烯基(C 6)、环已二烯基(C 6)、环庚基(C 7)、环庚烯基(C 7)、环庚二烯基(C 7)、环庚三烯基(C 7),等等。环烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“3-11元杂环基”是指具有环碳原子和1至5个环杂原子的3至11元非芳香环系的基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,优选3-9元杂环基,其为具有环碳原子和1至5个环杂原子的3至9元非芳香环系;在一些实施方案中,优选3-8元杂环基,其为具有环碳原子和1至4个环杂原子的3至8元非芳香环系;优选3-6元杂环基,其为具有环碳原子和1至3个环杂原子 的3至6元非芳香环系;优选3-5元杂环基,其为具有环碳原子和1至2个环杂原子的3至5元非芳香环系;优选4-8元杂环基,其为具有环碳原子和1至3个环杂原子的4至8元非芳香环系;更优选5-6元杂环基,其为具有环碳原子和1至3个环杂原子的5至6元非芳香环系。杂环基还包括其中上述杂环基环与一个或多个环烷基稠合的环体系,其中连接点在环烷基环上,或其中上述杂环基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在杂环基环上;且在这样的情况下,环成员的数目继续表示在杂环基环体系中环成员的数目。示例性的包含一个杂原子的3元杂环基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基(thiorenyl)。示例性的含有一个杂原子的4元杂环基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环基包括但不限于:二氧杂环戊烷基、氧硫杂环戊烷基(oxasulfuranyl)、二硫杂环戊烷基(disulfuranyl)和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环基包括但不限于:三唑啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环基包括但不限于:哌啶基、四氢吡喃基、二氢吡啶基和硫杂环己烷基(thianyl)。示例性的包含两个杂原子的6元杂环基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性的包含三个杂原子的6元杂环基包括但不限于:六氢三嗪基(triazinanyl)。示例性的含有一个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的与C 6芳基环稠合的5元杂环基(在本文中也称作5,6-双环杂环基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与C 6芳基环稠合的6元杂环基(本文还指的是6,6-双环杂环基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。杂环基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 6-10芳基”是指具有6-10个环碳原子和零个杂原子的单环或多环的(例如,双环)4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“C 6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C 10芳基”;例如,萘基,例如,1-萘基和2-萘基)。芳基还包括其中上述芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述芳基环上,在这种情况下,碳原子的数目继续表示所述芳基环系统中的碳原子数目。芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“5-10元杂芳基”是指具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。杂芳基双环系统在一个或两个环中可以包括一个或多个杂原子。杂芳基还包括其中上述杂芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述杂芳基环上,在这种情况下,碳原子的数目继续表示所述杂芳基环系统中的碳原子数目。在一些实施方案中,5-6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环或双环的4n+2芳族环体系。示例性的含有一个杂原子的5元杂芳基包括但 不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:三唑基、噁二唑基(例如,1,2,4-噁二唑基)和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括但不限于:吡啶基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并三唑基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。杂芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
优选的杂芳基的具体例子包括:吡咯基、咪唑基、吡唑基、2-吡啶基、3-吡啶基、4-吡啶基、嘧啶基、吡嗪基、哒嗪基、三唑基(4H-l,2,4-三唑基、1H-1,2,3-三唑基、2H-l,2,3-三唑基、吡喃基、2-呋喃基、3-呋喃等、2-噻吩基、3-噻吩基、噁唑基、异噁唑基、噁唑基(1,2,4-噁唑基、1,3,4-噁唑基、1,2,5-噁唑基、噻唑基、噻二唑基(1,2,4-噻二唑基、1,3,4-噻二唑基、1,2,5-噻二唑基)。
“羰基”无论单独使用或与其他术语连用(如氨基羰基),表示为-C(O)-。
“氧代”表示=O。
“硫代”表示=S。
本文定义的烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基等为任选取代的基团。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR aa、-ON(R bb) 2、-N(R bb) 2、-N(R bb) 3 +X -、-N(OR cc)R bb、-SH、-SR aa、-SSR cc、-C(=O)R aa、-CO 2H、-CHO、-C(OR cc) 2、-CO 2R aa、-OC(=O)R aa、-OCO 2R aa、-C(=O)N(R bb) 2、-OC(=O)N(R bb) 2、-NR bbC(=O)R aa、-NR bbCO 2R aa、-NR bbC(=O)N(R bb) 2、-C(=NR bb)R aa、-C(=NR bb)OR aa、-OC(=NR bb)R aa、-OC(=NR bb)OR aa、-C(=NR bb)N(R bb) 2、-OC(=NR bb)N(R bb) 2、-NR bbC(=NR bb)N(R bb) 2、-C(=O)NR bbSO 2R aa、-NR bbSO 2R aa、-SO 2N(R bb) 2、-SO 2R aa、-SO 2OR aa、-OSO 2R aa、-S(=O)R aa、-OS(=O)R aa、-Si(R aa) 3、-OSi(R aa) 3、-C(=S)N(R bb) 2、-C(=O)SR aa、-C(=S)SR aa、-SC(=S)SR aa、-SC(=O)SR aa、-OC(=O)SR aa、-SC(=O)OR aa、-SC(=O)R aa、-P(=O) 2R aa、-OP(=O) 2R aa、-P(=O)(R aa) 2、-OP(=O)(R aa) 2、-OP(=O)(OR cc) 2、-P(=O) 2N(R bb) 2、-OP(=O) 2N(R bb) 2、-P(=O)(NR bb) 2、-OP(=O)(NR bb) 2、-NR bbP(=O)(OR cc) 2、-NR bbP(=O)(NR bb) 2、-P(R cc) 2、-P(R cc) 3、-OP(R cc) 2、-OP(R cc) 3、-B(R aa) 2、-B(OR cc) 2、-BR aa(OR cc)、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(R bb) 2、=NNR bbC(=O)R aa、=NNR bbC(=O)OR aa、 =NNR bbS(=O) 2R aa、=NR bb或=NOR cc取代;
R aa的每个独立地选自烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R aa基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R bb的每个独立地选自:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R bb基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R cc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R cc基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R dd的每个独立地选自:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR ee、-ON(R ff) 2、-N(R ff) 2,、-N(R ff) 3 +X -、-N(OR ee)R ff、-SH、-SR ee、-SSR ee、-C(=O)R ee、-CO 2H、-CO 2R ee、-OC(=O)R ee、-OCO 2R ee、-C(=O)N(R ff) 2、-OC(=O)N(R ff) 2、-NR ffC(=O)R ee、-NR ffCO 2R ee、-NR ffC(=O)N(R ff) 2、-C(=NR ff)OR ee、-OC(=NR ff)R ee、-OC(=NR ff)OR ee、-C(=NR ff)N(R ff) 2、-OC(=NR ff)N(R ff) 2、-NR ffC(=NR ff)N(R ff) 2、-NR ffSO 2R ee、-SO 2N(R ff) 2、-SO 2R ee、-SO 2OR ee、-OSO 2R ee、-S(=O)R ee、-Si(R ee) 3、-OSi(R ee) 3、-C(=S)N(R ff) 2、-C(=O)SR ee、-C(=S)SR ee、-SC(=S)SR ee、-P(=O) 2R ee、-P(=O)(R ee) 2、-OP(=O)(R ee) 2、-OP(=O)(OR ee) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基、杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代,或者两个偕R dd取代基可结合以形成=O或=S;
R ee的每个独立地选自烷基、卤代烷基、烯基、炔基、碳环基、芳基、杂环基和杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R ff的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R ff基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R gg的每个独立地是:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OC 1-6烷基、-ON(C 1-6烷基) 2、-N(C 1-6烷基) 2、-N(C 1-6烷基) 3 +X -、-NH(C 1-6烷基) 2 +X -、-NH 2(C 1-6烷基) +X -、-NH 3 +X -、-N(OC 1-6烷基)(C 1-6烷基)、-N(OH)(C 1-6烷基)、-NH(OH)、-SH、-SC 1-6烷基、-SS(C 1-6烷基)、-C(=O)(C 1-6烷基)、-CO 2H、-CO 2(C 1-6烷基)、-OC(=O)(C 1-6烷基)、-OCO 2(C 1-6烷基)、-C(=O)NH 2、-C(=O)N(C 1-6烷基) 2、-OC(=O)NH(C 1-6烷基)、-NHC(=O)(C 1-6烷基)、-N(C 1-6烷基)C(=O)(C 1-6烷基)、-NHCO 2(C 1-6烷基)、-NHC(=O)N(C 1-6烷基) 2、-NHC(=O)NH(C 1-6烷基)、-NHC(=O)NH 2、-C(=NH)O(C 1-6烷基)、-OC(=NH)(C 1-6烷基)、-OC(=NH)OC 1-6烷基、-C(=NH)N(C 1-6烷基) 2、-C(=NH)NH(C 1-6烷基)、-C(=NH)NH 2、 -OC(=NH)N(C 1-6烷基) 2、-OC(NH)NH(C 1-6烷基)、-OC(NH)NH 2、-NHC(NH)N(C 1-6烷基) 2、-NHC(=NH)NH 2、-NHSO 2(C 1-6烷基)、-SO 2N(C 1-6烷基) 2、-SO 2NH(C 1-6烷基)、-SO 2NH 2、-SO 2C 1-6烷基、-SO 2OC 1-6烷基、-OSO 2C 1-6烷基、-SOC 1-6烷基、-Si(C 1-6烷基) 3、-OSi(C 1-6烷基) 3、-C(=S)N(C 1-6烷基) 2、C(=S)NH(C 1-6烷基)、C(=S)NH 2、-C(=O)S(C 1-6烷基)、-C(=S)SC 1-6烷基、-SC(=S)SC 1-6烷基、-P(=O) 2(C 1-6烷基)、-P(=O)(C 1-6烷基) 2、-OP(=O)(C 1-6烷基) 2、-OP(=O)(OC 1-6烷基) 2、C 1-6烷基、C 1-6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 7碳环基、C 6-C 10芳基、C 3-C 7杂环基、C 5-C 10杂芳基;或者两个偕R gg取代基可结合形成=O或=S;其中,X -为反离子。
示例性的氮原子上取代基包括但不局限于:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR bb)R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者连接至氮原子的两个R cc基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代,且其中R aa、R bb、R cc和R dd如上所述。
其它定义
术语“癌症”包括但不限于下列癌症:乳腺、卵巢、子宫颈、前列腺、睾丸、食道、胃、皮肤、肺、骨、结肠、胰腺、甲状腺、胆道、颊腔与咽(口)、唇、舌、口腔、咽、小肠、结肠直肠、大肠、直肠、脑与中枢神经系统的癌症、成胶质细胞瘤、神经母细胞瘤、角化棘皮瘤、表皮样癌、大细胞癌、腺癌、腺瘤、滤泡癌、未分化的癌、乳头状癌、精原细胞瘤、黑色素瘤、肉瘤、膀胱癌、肝癌、肾癌、骨髓障碍、淋巴障碍、霍奇金氏病、毛细胞癌和白血病。
本文所用的术语“治疗”涉及逆转、减轻、抑制该术语适用的障碍或病症的进展或者预防之,或者这类障碍或病症的一种或多种症状。本文所用的名词“治疗”涉及动词治疗的动作,后者是如刚才所定义的。
本文所用的术语“药学上可接受的盐”表示本发明化合物的那些羧酸盐、氨基酸加成盐,它们在可靠的医学判断范围内适用于与患者组织接触,不会产生不恰当的毒性、刺激作用、变态反应等,与合理的益处/风险比相称,就它们的预期应用而言是有效的,包括(可能的话)本发明化合物的两性离子形式。
药学上可接受的碱加成盐是与金属或胺生成的,例如碱金属与碱土金属氢氧化物或有机胺。用作阳离子的金属的实例有钠、钾、镁、钙等。适合的胺的实例有N,N′-二苄基乙二胺、氯普鲁卡因、胆碱、二乙醇胺、乙二胺、N-甲基葡糖胺和普鲁卡因。
酸性化合物的碱加成盐可以这样制备,按照常规方式使游离酸形式与足量所需的碱接触,生成盐。按照常规方式使盐形式与酸接触,再分离游离酸,可以使游离酸再生。游离酸形式在某些物理性质上多少不同于它们各自的盐形式,例如在极性溶剂中的溶解度,但是出于本发明的目的,盐还是等价于它们各自的游离酸。
盐可以是从无机酸制备的硫酸盐、焦硫酸盐、硫酸氢盐、亚硫酸盐、亚硫酸氢盐、硝酸盐、磷酸 盐、磷酸一氢盐、磷酸二氢盐、偏磷酸盐、焦磷酸盐、氯化物、溴化物、碘化物,酸例如盐酸、硝酸、硫酸、氢溴酸、氢碘酸、磷酸等。代表性盐包括:氢溴酸盐、盐酸盐、硫酸盐、硫酸氢盐、硝酸盐、乙酸盐、草酸盐、戊酸盐、油酸盐、棕榈酸盐、硬脂酸盐、月桂酸盐、硼酸盐、苯甲酸盐、乳酸盐、磷酸盐、甲苯磺酸盐、柠檬酸盐、马来酸盐、富马酸盐、琥珀酸盐、酒石酸盐、萘甲酸盐、甲磺酸盐、葡庚糖酸盐、乳糖酸盐、月桂基磺酸盐和羟乙磺酸盐等。盐也可以是从有机酸制备的,例如脂肪族一元与二元羧酸、苯基取代的烷酸、羟基烷酸、烷二酸、芳香族酸、脂肪族与芳香族磺酸等。代表性盐包括乙酸盐、丙酸盐、辛酸盐、异丁酸盐、草酸盐、丙二酸盐、琥珀酸盐、辛二酸盐、癸二酸盐、富马酸盐、马来酸盐、扁桃酸盐、苯甲酸盐、氯苯甲酸盆、甲基苯甲酸盐、二硝基苯甲酸盐、萘甲酸盐、苯磺酸盐、甲苯磺酸盐、苯乙酸盐、柠檬酸盐、乳酸盐、马来酸盐、酒石酸盐、甲磺酸盐等。药学上可接受的盐可以包括基于碱金属与碱土金属的阳离子,例如钠、锂、钾、钙、镁等,以及无毒的铵、季铵和胺阳离子,包括但不限于铵、四甲基铵、四乙基铵、甲胺、二甲胺、三甲胺、三乙胺、乙胺等。还涵盖氨基酸的盐,例如精氨酸盐、葡糖酸盐、半乳糖醛酸盐等(例如参见Berge S.M.et al.,″Pharmaceutical Salts,”J.Pharm.Sci.,1977;66:1-19,引入此作为参考)。
本发明化合物的药学上可接受的无毒酰胺的实例包括从C 1-C 6烷基酯,其中烷基是直链或支链的。可接受的酯还包括C 5-C 7环烷基酯以及芳基烷基酯,例如但不限于苄基酯。C 1-C 4烷基酯是优选的。本发明化合物的酯可以按照常规方法制备,例如:March′s Advanced Organic Chemistry,5 Edition”M.B.Smith&J.March,John Wiley&Sons,2001。
本发明化合物的药学上可接受的无毒性酰胺的实例包括从氨、伯C 1-C 6烷基胺和仲C 1-C 6二烷基胺衍生的酰胺,其中烷基是直链或支链的。在仲胺的情况下,胺也可以是含有一个氮原子的5或6元杂环的形式。从氨、C 1-C 3烷基伯胺和C 1-C 2二烷基仲胺衍生的酰胺是优选的。本发明化合物的酰胺可以按照常规方法制备,例如:March′s Advanced Organic Chemistry,5 Edition”,M.B.Smith&J.March,John Wiley&Sons,2001。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。本文可互换使用术语“人”、“患者”和“受试者”。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括在受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药代动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗有效量和预防有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足 以提供治疗益处的量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化的量。化合物的治疗有效量是指单独使用或与其它疗法联用时,治疗剂的量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其它治疗剂的治疗效果的量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的量,或足以预防与疾病、障碍或病症有关的一或多种症状的量,或防止疾病、障碍或病症复发的量。化合物的预防有效量是指单独使用或与其它药剂联用时,治疗剂的量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的量,或增强其它预防药剂的预防效果的量。
“组合”以及相关术语是指同时或依次给药本发明化合物和其它治疗剂。例如,本发明化合物可以与其它治疗剂以分开的单位剂型同时或依次给药,或与其它治疗剂一起在单一单位剂型中同时给药。
具体实施方案
本文中,“本发明化合物”指的是以下的式(I)至式(VI)化合物、其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物。
本文中,化合物使用标准的命名法命名。具有非对称中心的化合物,应该明白(除非另有说明)所有的光学异构体及其混合物均包含在内。此外,除非另有规定,本发明所包括的所有异构体化合物与碳碳双键可能以Z和E的形式出现。在不同的互变异构形式存在的化合物,一个所述化合物并不局限于任何特定的互变异构体,而是旨在涵盖所有的互变异构形式。某些化合物所使用的一般公式,包括描述、变量。除非另有规定,在这样的公式中的每个变量被定义独立于任何其他变量和在每个发生时独立地定义了一个公式中的任何一个变量的多个变量。
在一个实施方案中,本发明涉及通式(A)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000003
其中,
X为CR 5或N;Y为CR 6或N;并且其中X和Y中至少有一个为N;
Z为CR #或N;
R 1、R 2、R 3、R 4和R #独立地选自H、C 1-6烷基和C 1-6卤代烷基;或者R 1与R 2,R 3与R 4连接形成键、C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;
R 5和R 6独立地选自H、卤素、C 1-6烷基或C 1-6卤代烷基;
当Z为CR #时,其中R #与R 1可以连接形成C 3-5元环烷基或3-5元杂环基;
环A选自C 6-10芳基和5-10元杂芳基;
环B选自5-10元杂芳基;
R a独立地选自任选被R**取代的H、卤素、-CN、-L-NRR’、-L-OR、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(=NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基;
其中L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;p=1或2;
R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR、-L’-C 3-7环烷基和-L’-4-8元杂环基;其中L’独立地选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、-O-C 1-6亚烷基和-NH-C 1-6亚烷基;
R b独立地选自任选被R**取代的H、卤素、-CN、-L-OR、-L-C 1-6烷基、-L-C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’和-NRC(O)NRR’;
R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基,或者R、R’与它们连接的氮原子形成4-8元杂环基;
m=0、1、2、3、4或5;
n=0、1、2、3、4或5。
在另一个实施方案中,本发明涉及通式(I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000004
其中,
X为CH或N;Y为CH或N;并且其中X和Y中至少有一个为N;
R 1、R 2、R 3和R 4独立地选自H、C 1-6烷基和C 1-6卤代烷基;或者R 1与R 2,R 3与R 4连接形成键、C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;
环A选自C 6-10芳基和5-10元杂芳基;
环B选自5-10元杂芳基;
R a独立地选自任选被R**取代的H、卤素、-CN、-L-NRR’、-L-OR、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(=NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基;
R b独立地选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’和-NRC(O)NRR’;
m=0、1、2、3、4或5;
n=0、1、2、3、4或5;
其中L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;
R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR、-L’-C 3-7环烷基和-L’-4-8元杂环基;
L’独立地选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、-O-C 1-6亚烷基和-NH-C 1-6亚烷基;
R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基,或者R、R’与它们连接的氮原子形成4-8元杂环基;
p=1或2。
X、Y和Z
在一个具体实施方案中,X为CR 5,且Y为N;在另一个具体实施方案中,X为CH,且Y为N;在另一个具体实施方案中,X为N,且Y为CR 6;在另一个具体实施方案中,X为N,且Y为CH;在另一个具体实施方案中,X为N,且Y为N。
在一个具体实施方案中,Z为CR #;在另一个具体实施方案中,Z为N。
R 1、R 2、R 3、R 4和R #
在一个具体实施方案中,R 1为H;在另一个具体实施方案中,R 1为C 1-6烷基,例如(R)-C 1-6烷基,例如(R)-甲基;在另一个具体实施方案中,R 1为C 1-6卤代烷基,例如(R)-C 1-6卤代烷基。在一个具体实施方案中,R 2为H;在另一个具体实施方案中,R 2为C 1-6烷基,例如(R)-C 1-6烷基,例如(R)-甲基;在另一个具体实施方案中,R 2为C 1-6卤代烷基,例如(R)-C 1-6卤代烷基。在一个具体实施方案中,R 3为H;在另一个具体实施方案中,R 3为C 1-6烷基,例如(R)-C 1-6烷基,例如(R)-甲基;在另一个具体实施方案中,R 3为C 1-6卤代烷基,例如(R)-C 1-6卤代烷基。在一个具体实施方案中,R 4为H;在另一个具体实施方案中,R 4为C 1-6烷基,例如(R)-C 1-6烷基,例如(R)-甲基;在另一个具体实施方案中,R 4为C 1-6卤代烷基,例如(R)-C 1-6卤代烷基。在一个具体实施方案中,R #为H;在另一个具体实施方案中,R #为C 1-6烷基,例如(R)-C 1-6烷基,例如(R)-甲基;在另一个具体实施方案中,R #为C 1-6卤代烷基,例如(R)-C 1-6卤代烷基。
在另一个具体实施方案中,R 1和R 2中至少一个为C 1-6烷基,例如(R)-C 1-6烷基,例如(R)-甲基。
在另一个具体实施方案中,R 1与R 2,或R 3与R 4连接形成键;在另一个具体实施方案中,R 1与R 2,或R 3与R 4连接形成C 1-6亚烷基,例如亚甲基、亚乙基或亚丙基;在另一个具体实施方案中,R 1与R 2,或R 3与R 4连接形成C 2-6亚烯基;在另一个具体实施方案中,R 1与R 2,或R 3与R 4连接形成C 2-6亚炔基。
在另一个具体实施方案中,当Z为CR #时,其中R #、R 1与它们连接的原子一起形成C 3-5元环烷基;在另一个具体实施方案中,所述C 3-5元环烷基为环丙基;在另一个具体实施方案中,所述C 3-5元 环烷基为环丁基;在另一个具体实施方案中,所述C 3-5元环烷基为环戊基;在另一个具体实施方案中,当Z为CR #时,其中R #、R 1与它们连接的原子一起形成3-5元杂环基;在另一个具体实施方案中,所述3-5元杂环基为氧杂环丙基、氮杂环丙基或硫杂环丙基;在另一个具体实施方案中,所述3-5元杂环基为氧杂环丁基、氮杂环丁基或硫杂环丁基;在另一个具体实施方案中,所述3-5元杂环基为四氢呋喃基、吡咯烷基或硫杂环戊基。
环A
在一个具体实施方案中,环A为C 6-10芳基,例如苯基;在另一个具体实施方案中,环A为5-10元杂芳基,例如5-6元杂芳基。
在另一个具体实施方案中,环A选自
Figure PCTCN2020094532-appb-000005
Figure PCTCN2020094532-appb-000006
在另一个具体实施方案中,环A选自
Figure PCTCN2020094532-appb-000007
在另一个具体实施方案中,环A选自
Figure PCTCN2020094532-appb-000008
其中R a1-R a11如上下文所定义。
环B
在一个具体实施方案中,环B为5-10元杂芳基,例如双环的9-10元杂芳基;在另一个具体实施方案中,环B为
Figure PCTCN2020094532-appb-000009
表示与分子其余部分的连接位置,Z 1-Z 12中有0、1、2或3个为选自O、S和N的杂原子,只要化学上允许;并且Z 1-Z 12任选被氧化形成=O基团;优选地,其中Z 1-Z 12中至少一个为选自O、S和N的杂原子,只要化学上允许;并且Z 1-Z 12任选被氧化形成=O基团;在另一个具体实施方案中,环B选自
Figure PCTCN2020094532-appb-000010
Figure PCTCN2020094532-appb-000011
在另一个具体实施方案中,环B选自
Figure PCTCN2020094532-appb-000012
在另一个具体实施方案中,环 B为
Figure PCTCN2020094532-appb-000013
在上述具体实施方案中,Z 1-Z 12中有0、1、2或3个为N原子;优选地,Z 1-Z 12中至少一个为N原子;在另一上述具体实施方案中,Z 1-Z 3中最多含有一个N原子;在另一上述具体实施方案中,Z 4-Z 6中最多含有两个N原子;在另一上述具体实施方案中,Z 4为N原子。
R a
在一个具体实施方案中,R a独立地选自任选被R**取代的H、-L-NRR’、-L-OR、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(=O)(=NR)R*、-L-S(O) pR*、-L-C 3-7环烷基和-L-4-8元杂环基;在另一个具体实施方案中,其中一个R a为H;在另一个具体实施方案中,其中一个R a为卤素;在另一个具体实施方案中,其中一个R a为-CN;在另一个具体实施方案中,其中一个R a为-L-NRR’;在另一个具体实施方案中,其中一个R a为-L-OR;在另一个具体实施方案中,其中一个R a为-C(O)R;在另一个具体实施方案中,其中一个R a为-C(O)OR;在另一个具体实施方案中,其中一个R a为-C(O)NRR’;在另一个具体实施方案中,其中一个R a为-OC(O)R’;在另一个具体实施方案中,其中一个R a为-NRC(O)R’;在另一个具体实施方案中,其中一个R a为-OC(O)NRR’;在另一个具体实施方案中,其中一个R a为-NRC(O)NRR’;在另一个具体实施方案中,其中一个R a为C 1-6烷基;在另一个具体实施方案中,其中一个R a为C 1-6卤代烷基;在另一个具体实施方案中,其中一个R a为C 1-6烷氧基;在另一个具体实施方案中,其中一个R a为C 1-6卤代烷氧基;在另一个具体实施方案中,其中一个R a为-L-S(O) pR*;在另一个具体实施方案中,其中一个R a为-L-S(=O)(=NR)R*;在另一个具体实施方案中,其中一个R a为-L-C 3-7环烷基;在另一个具体实施方案中,其中一个R a为-L-4-8元杂环基;在上述具体实施方案中,其中所述R a被一个、两个或三个R**取代。
R b
在一个具体实施方案中,R b独立地选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-NRR’和-NRC(O)R’;在另一个具体实施方案中,其中一个R b为H;在另一个具体实施方案中,其中一个R b为卤素;在另一个具体实施方案中,其中一个R b为-CN;在另一个具体实施方案中,其中一个R b为-L-OR;在另一个具体实施方案中,其中一个R b为-OH;在另一个具体实施方案中,其中一个R b为-L-C 1-6烷基;在另一个具体实施方案中,其中一个R b为C 1-6烷基;在另一个具体实施方案中,其中一个R b为-L-C 1-6烷氧基;在另一个具体实施方案中,其中一个R b为C 1-6烷氧基;在另一个具体实施方案中,其中一个R b为-L-C 3-7环烷基;在另一个具体实施方案中,其中一个R b为-L-3-8元杂环基;在另一个具体实施方案中,其中一个R b为-NRR’;在另一个具体实施方案中,其中一个R b为-C(O)R;在另一个具体实施方案中,其中一个R b为-C(O)OR;在另一个具体实施方案中,其中一个R b为-C(O)NRR’;在另一个具体实施方案中,其中一个R b为-OC(O)R’;在另一个具体实施方案中,其中一个R b为-NRC(O)R’;在另一个具体实施方案中,其中一个R b为-OC(O)NRR’;在另一个具体实施方案中,其中一个R b为-NRC(O)NRR’。
m
在一个具体实施方案中,m=0;在另一个具体实施方案中,m=1;在另一个具体实施方案中,m=2;在另一个具体实施方案中,m=3;在另一个具体实施方案中,m=4;在另一个具体实施方案中,m=5。
n
在一个具体实施方案中,n=0;在另一个具体实施方案中,n=1;在另一个具体实施方案中,n=2;在另一个具体实施方案中,n=3;在另一个具体实施方案中,n=4;在另一个具体实施方案中,n=5。
L
在一个具体实施方案中,L独立地选自键、-NR-、-C(O)-和C 1-6亚烷基;在另一个具体实施方案中,L为键;在另一个具体实施方案中,L为键-O-;在另一个具体实施方案中,L为键-S-;在另一个具体实施方案中,L为键-NR-;在另一个具体实施方案中,L为键-C(O)-;在另一个具体实施方案中,L为键C 1-6亚烷基;在另一个具体实施方案中,L为键C 2-6亚烯基;在另一个具体实施方案中,L为键C 2-6亚炔基。
R*
在一个具体实施方案中,R*为C 1-6烷基;在另一个具体实施方案中,R*为C 1-6卤代烷基;在另一个具体实施方案中,R*为-L’-NRR’;在另一个具体实施方案中,R*为-L’-OR;在另一个具体实施方案中,R*为-L’-C 3-7环烷基;在另一个具体实施方案中,R*为-L’-4-8元杂环基。
L’
在一个具体实施方案中,L’为键;在另一个具体实施方案中,L’为C 1-6亚烷基;在另一个具体实施方案中,L’为C 2-6亚烯基;在另一个具体实施方案中,L’为C 2-6亚炔基;在另一个具体实施方案中,L’为-O-C 1-6亚烷基;在另一个具体实施方案中,L’为-NH-C 1-6亚烷基。
R**
在一个具体实施方案中,R**为H;在另一个具体实施方案中,R**为卤素;在另一个具体实施方案中,R**为-CN;;在另一个具体实施方案中,R**为-C(=O)R;在另一个具体实施方案中,R**为C 1-6烷基;在另一个具体实施方案中,R**为C 1-6卤代烷基;在另一个具体实施方案中,R**为C 1-6烷氧基;在另一个具体实施方案中,R**为C 1-6卤代烷氧基;在另一个具体实施方案中,R**为4-8元杂环基;在另一个具体实施方案中,R**为-S(O) p-C 1-6烷基;在另一个具体实施方案中,R**为-S(O) p-C 1-6卤代烷基;在另一个具体实施方案中,R**为-NRR’。
R和R’
在一个具体实施方案中,R和R’独立地为H;在另一个具体实施方案中,R和R’独立地为C 1-6烷基;在另一个具体实施方案中,R和R’独立地为C 1-6卤代烷基;在另一个具体实施方案中,R、R’ 与它们连接的氮原子形成4-8元杂环基。
p
在一个具体实施方案中,p=1;在另一个具体实施方案中,p=2。
以上任一具体实施方案中的任一技术方案或其任意组合,可以与其它具体实施方案中的任一技术方案或其任意组合进行组合。例如,X的任一技术方案或其任意组合,可以与Y、R 1-R 4、环A、环B、R a、R b、R*、R**、L、L’、m、n、p、R和R’等的任一技术方案或其任意组合进行组合。本发明旨在包括所有这些技术方案的组合,限于篇幅,不再一一列出。
在更具体的实施方案中,本发明提供了通式(II)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000014
其中,Y、环A、环B、R b和n如上文中所定义。
在另一更具体的实施方案中,本发明提供了通式(II)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
Y为CH或N;
环A选自:
Figure PCTCN2020094532-appb-000015
R a1选自H、C 1-6烷基和C 1-6卤代烷基;
R a2选自C 1-6烷基和C 1-6卤代烷基;
R a3选自H、C 1-6烷基和C 1-6卤代烷基;
R a4选自H和S(O) pR*,其中p=1或2,R*选自C 1-6烷基和C 1-6卤代烷基;
R a9选自H和-NRC(O)NRR’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
R a10选自C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R a11选自H和-NRR’,其中R和R’独立地选自H、C 1-6烷基,C 1-6卤代烷基;
B选自
Figure PCTCN2020094532-appb-000016
R b选自氢、卤素、C 1-6烷基和C 1-6卤代烷基;
n=0、1、2、3或4。
在另一更具体的实施方案中,本发明提供了通式(II)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
环A选自:
Figure PCTCN2020094532-appb-000017
R a1为H或甲基;
R a2为甲基或异丙基;
R a3为H或甲基;
R a4为H或-SO 2Me;
R a9为H或-NH 2C(O)NH 2Et;
R a10为CF 2H或C 1-6烷氧基;
R a11为H;
B选自
Figure PCTCN2020094532-appb-000018
R b为H或甲基;
n=0、1或2。
在另一更具体的实施方案中,本发明提供了通式(III)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000019
其中,Y、环A、R a和m如上文所定义。
在另一更具体的实施方案中,本发明提供了通式(III)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
环A选自:
Figure PCTCN2020094532-appb-000020
R a1选自C 1-6烷基和C 1-6卤代烷基;
R a2选自C 1-6烷基和C 1-6卤代烷基;
R a3选自C 1-6烷基和C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(III)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
环A选自:
Figure PCTCN2020094532-appb-000021
R a1为甲基;
R a2为甲基或异丙基;
R a3为甲基。
在另一更具体的实施方案中,本发明提供了通式(IV)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000022
其中,
X为CH或N;
环A选自:
Figure PCTCN2020094532-appb-000023
R a1选自H、C 1-6烷基和C 1-6卤代烷基;
R a2选自任选被R**取代的C 1-6烷基、C 1-6卤代烷基、-L-NRR’、-L-OR、-L-4-8元杂环基和-L-S(O) pR*;其中L选自键、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基,p=1或2,R*选自C 1-6烷基、C 3-7环烷基、4-8元杂环基、C 1-6卤代烷基和-NRR’;R**选自H、卤素、-OR、-C(O)R、C 1-6烷基、C 1-6卤代烷基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
R a3选自H、C 1-6烷基和C 1-6卤代烷基;
R a4选自任选被R**取代的C 1-6烷基、-L-S(=O)(=NR)R*、-L-S(O) pR*和-L-4-8元杂环基;其中,p=1或2,L选自键、-NR-、-C(O)-和C 1-6亚烷基,R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR和-L’-4-8元杂环基;并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自H、-CN、-NRR’、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基和-S(O) p-C 1-6卤代烷基,其中p=1或2;
R a5选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R a6为H;
R a7为H;
R a8选自C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基;
R a10选自任选被-CN取代的C 1-6烷基和C 1-6卤代烷基;
R a11为H;
R b选自氢、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R;
n=0、1、2或3。
在另一更具体的实施方案中,本发明提供了通式(IV)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
环A选自:
Figure PCTCN2020094532-appb-000024
R a1选自H、C 1-6烷基和C 1-6卤代烷基;
R a2选自C 1-6烷基、C 1-6卤代烷基、-L-NRR’、-L-OR和-L-4-8元杂环基,其中L为键或C 1-6亚烷基,R和R’独立地为H、C 1-6烷基或C 1-6卤代烷基;
R a3选自H、C 1-6烷基和C 1-6卤代烷基;
R a4选自-S(O) pR*,其中p=1或2,R*选自C 1-6烷基、4-8元杂环基、C 1-6卤代烷基和-L’-NRR’,并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;
R a5选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R a6为H;
R a7为H;
R a8选自C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基;
R a10选自任选被-CN取代的C 1-6烷基和C 1-6卤代烷基;
R a11为H;
R b选自氢、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R;
n=0、1、2或3。
在另一更具体的实施方案中,本发明提供了通式(IV)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
环A选自:
Figure PCTCN2020094532-appb-000025
R a1为H或甲基;
R a2为甲基、异丙基、-CH 2CH 2CH 2OH、-CH 2CH 2OMe、-CH 2CH 2NMe 2、-CH 2CH 2CH 2NMe 2、氮杂环丁烷-3-基甲基、氧杂环丁烷-3-基甲基、吡喃-4-基、四氢吡咯基或哌啶基;
R a3为H、甲基或CF 3
R a4为-SO 2N(Me) 2、-SO 2Me、-SO 2iPr或-SO 2-哌啶-4-基;
R a5为甲基、甲氧基、异丙基或三氟甲基;
R a6为H;
R a7为H;
R a8为Me;
R a10选自任选被-CN取代的异丙基;
R a11为H;
R b为氢、卤素、甲氧基或-CH 2-氮杂环丁烷-3-基;
n=0、1、2或3。
在另一更具体的实施方案中,本发明提供了通式(IV)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000026
其中,
X为CH或N;
环A选自:
Figure PCTCN2020094532-appb-000027
R a1选自C 1-6烷基和C 1-6卤代烷基;
R a2选自任选被R**取代的C 1-6烷基、C 1-6卤代烷基、-L-NRR’、-L-4-8元杂环基、-L-S(O) pR*,其中L选自键、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基,p=1或2,R*选自C 1-6烷基、C 1-6卤代烷基和-NRR’,并且其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自卤素、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基和-S(O) p-C 1-6卤代烷基,其中p=1或2;
R a3选自H、C 1-6烷基和C 1-6卤代烷基;
R a4选自任选被R**取代的C 1-6烷基、-L-S(=O)(=NR)R*、-L-S(O) pR*和-L-4-8元杂环基;其中,p=1或2,L选自键、-NR-、-C(O)-和C 1-6亚烷基,R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR和-L’-4-8元杂环基;并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自-CN、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基和-S(O) p-C 1-6卤代烷基,其中p=1或2;
R a5选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R a10选自任选被-CN取代的C 1-6烷基和C 1-6卤代烷基;
R a11为H;
R b选自氢、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R;
n=0、1、2或3。
在另一更具体的实施方案中,本发明提供了通式(IV)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
环A为
Figure PCTCN2020094532-appb-000028
R a1选自C 1-6烷基和C 1-6卤代烷基;
R a2选自C 1-6烷基、C 1-6卤代烷基和-L-4-8元杂环基;
R a3选自H、C 1-6烷基和C 1-6卤代烷基;
R a4选自-S(O) pR*,其中p=1或2,R*选自C 1-6烷基、C 1-6卤代烷基、-4-8元杂环基和-L’-NRR’,并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、 C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;
R a5选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R a10选自任选被-CN取代的C 1-6烷基和C 1-6卤代烷基;
R a11为H;
R b选自氢、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R;
L选自键和C 1-6亚烷基;
n=0、1、2或3。
在另一更具体的实施方案中,本发明提供了通式(IV)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
环A为
Figure PCTCN2020094532-appb-000029
R a1为甲基;
R a2为-CH 2CH 2CH 2OH、-CH 2CH 2NMe 2、-CH 2CH 2CH 2NMe 2、-CH 2CH 2OMe、氮杂环丁烷-3-基甲基、氧杂环丁烷-3-基甲基、吡喃-4-基、四氢吡咯基或哌啶基;
R a3为H、甲基、CF 3
R a4为-SO 2N(Me) 2、-SO 2Me、-SO 2iPr或-SO 2-哌啶-4-基;
R a5为甲基、异丙基、甲氧基或三氟甲基;
R b为氢、卤素、甲氧基或-CH 2-氮杂环丁烷-3-基;
n=0、1、2或3。
在另一更具体的实施方案中,本发明提供了通式(V)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000030
其中,
X为CH或N;
R a1选自C 1-6烷基和C 1-6卤代烷基;
R a2选自任选被R**取代的-L-S(O) pR*、-L-S(=O)(=NR)R*、C 1-6烷基和-L-4-8元杂环基;
R a3选自C 1-6烷基和C 1-6卤代烷基;
环B为
Figure PCTCN2020094532-appb-000031
Figure PCTCN2020094532-appb-000032
R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’;
n=0、1、2、3、4或5;
p=1或2;
R*选自C 1-6烷基或C 1-6卤代烷基;
L选自键、-O-、-S-、-NR-、-C(O)-或C 1-6亚烷基;
R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(V)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
R a1选自C 1-6烷基和C 1-6卤代烷基;
R a2选自C 1-6烷基、C 1-6卤代烷基和-L-4-8元杂环基;
R a3选自C 1-6烷基和C 1-6卤代烷基;
环B选自:
Figure PCTCN2020094532-appb-000033
Figure PCTCN2020094532-appb-000034
R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
L选自键和C 1-6亚烷基;
n=0、1、2、3、4或5。
在另一更具体的实施方案中,本发明提供了通式(V)的化合物,或其药学上可接受的盐、对映异 构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
R a1为甲基;
R a2为甲基、异丙基或哌啶基;
R a3为甲基或三氟甲基;
环B选自:
Figure PCTCN2020094532-appb-000035
Figure PCTCN2020094532-appb-000036
R b选自H、卤素、甲基、甲氧基、-CH 2-氮杂环丁烷-3-基、-NHMe和-NHC(O)Me;
n=0、1或2。
在另一更具体的实施方案中,本发明提供了通式(VI)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000037
其中,
X为CH或N;
R a4选自任选被R**取代的-L-S(O) 2R*基团;其中,L选自键和C 1-6亚烷基,R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’和-L’-4-8元杂环基;并且其中L’选自键和C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自氢、-CN、C(=O)R、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’,其中p=1或2;
R a5选自甲基、甲氧基、异丙基或三氟甲基;
环B选自:
Figure PCTCN2020094532-appb-000038
Figure PCTCN2020094532-appb-000039
R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
n=0、1、2、3、4或5。
在另一更具体的实施方案中,本发明提供了通式(VI)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
R a4选自-S(O) 2R*,其中,R*选自C 1-6烷基、-L’-NRR’和4-8元杂环基,并且其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;L’选自键和C 1-6亚烷基;
R a5选自甲基、甲氧基、异丙基或三氟甲基;
环B选自:
Figure PCTCN2020094532-appb-000040
Figure PCTCN2020094532-appb-000041
R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
L选自键和C 1-6亚烷基;
优选地,n=0、1或2。
在另一更具体的实施方案中,本发明提供了通式(VI)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
X为CH或N;
R a4为-SO 2N(Me) 2、-SO 2Me、-SO 2iPr或-SO 2-哌啶-4-基;
R a5选自甲基、甲氧基、异丙基或三氟甲基;
环B选自:
Figure PCTCN2020094532-appb-000042
Figure PCTCN2020094532-appb-000043
R b选自H、卤素、甲基、甲氧基、-CH 2-氮杂环丁烷-3-基、-NHMe和-NHC(O)Me;
n=0、1或2。
在另一更具体的实施方案中,本发明提供了通式(VII)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000044
其中,
环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
R a1为C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基或C 1-6卤代烷氧基(优选甲基、三氟甲基或甲氧基);
R a3为C 1-6烷基或C 1-6卤代烷基(优选甲基或三氟甲基);
R a12为H、C 1-6烷基或C 1-6卤代烷基;
R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基;
Z 1为N或CR b1
Z 2为N或CR b2
Z 3为N或CR b3
Z 4为NR b4或C(R b4) 2
Z 5为N或CR b5
Z 6为N或CR b6
R b1、R b2、R b3、R b4、R b5和R b6独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基(优选地选自H、卤素和甲氧基);
优选地,
R a1和R a3中至少一个为C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(VII)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12 取代的含N原子的5-6元杂环基);
R a1为C 1-6烷基或C 1-6卤代烷基(优选甲基或三氟甲基);
R a3为C 1-6烷基或C 1-6卤代烷基(优选甲基或三氟甲基);
R a12为H、C 1-6烷基或C 1-6卤代烷基;
R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基;
Z 1为N或CR b1
Z 2为N或CR b2
Z 3为N或CR b3
Z 4为NR b4或C(R b4) 2
Z 5为N或CR b5
Z 6为N或CR b6
R b2为H或卤素(优选H或F);
R b3为H;
R b1、R b4、R b5和R b6独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基(优选地选自H、卤素和甲氧基);
优选地,
R a1和R a3中至少一个为C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(VIII)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000045
其中,
环C为
Figure PCTCN2020094532-appb-000046
其中与分子其余部分的连接点在其中一个*所示的位置;
R a1为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
R a3为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
R a12为H或C 1-6烷基(优选为H或甲基);
R 5为H或卤素(优选为H或F);
环B选自
Figure PCTCN2020094532-appb-000047
R b为H、甲氧基或卤素(优选为H或卤素);
n为0、1、2或3;
优选地,
R a1和R a3中至少一个为C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(VIII)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
环C为
Figure PCTCN2020094532-appb-000048
其中与分子其余部分的连接点在其中一个*所示的位置;
R a1为甲基或三氟甲基;
R a3为甲基或三氟甲基;
R a12为H或甲基;
R 5为H或F;
环B选自
Figure PCTCN2020094532-appb-000049
R b为H;
n为0、1、2或3;
优选地,
R a1和R a3中至少一个为C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(IX)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000050
其中,
环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
R a1为C 1-6烷基或C 1-6卤代烷基;
R a3为C 1-6烷基或C 1-6卤代烷基;
R a12为H、C 1-6烷基或C 1-6卤代烷基;
R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基;
Z 7为N或CR b7
Z 8为N或CR b8
Z 9为N或CR b9
Z 10为N或CR b10
Z 11为N或CR b11
Z 12为N或CR b12
其中,
R b7为H、-L-OR、-L-C 1-6烷基、-L-C 3-7环烷基、-NRR’、-NRC(O)R’或-NRC(O)NRR’;
R b8、R b9、R b10、R b11和R b12独立地选自H、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基(优选地选自H、卤素、-CN和甲氧基);
L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(IX)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
R a1为C 1-6烷基或C 1-6卤代烷基;
R a3为C 1-6烷基或C 1-6卤代烷基;
R a12为H、C 1-6烷基或C 1-6卤代烷基;
R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基;
Z 7为N或CR b7
Z 8为N或CR b8
Z 9为N或CR b9
Z 10为N或CR b10
Z 11为N或CR b11
Z 12为N或CR b12
其中,
R b7选自-NRR’、-NRC(O)R’或-NRC(O)NRR’;
R b8、R b9、R b10、R b11和R b12独立地选自H、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和 C 1-6卤代烷氧基(优选地选自H、卤素、-CN和甲氧基);
L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;
R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
在另一更具体的实施方案中,本发明提供了通式(X)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000051
其中,
环C为
Figure PCTCN2020094532-appb-000052
其中与分子其余部分的连接点在其中一个*所示的位置;
R a1为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
R a3为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
R a12为H或C 1-6烷基(优选为H或甲基);
R 5为H;
R b7选自-NRR’、-NRC(O)R’或-NRC(O)NRR’;其中,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基(优选地,R b7选自NH 2、NHMe、NHEt、NHC(O)Me或NHC(O)Et);
R b为H或卤素(优选为H或F)。
在另一更具体的实施方案中,本发明提供了通式(X)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
环C为
Figure PCTCN2020094532-appb-000053
其中与分子其余部分的连接点在其中一个*所示的位置;
R a1为甲基或三氟甲基;
R a3为甲基或三氟甲基;
R a12为H或甲基;
R 5为H;
R b7选自NH 2、NHMe或NHC(O)Me;
R b为H或F。
在另一更具体的实施方案中,本发明提供了通式(XI)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000054
其中,
环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
R a1为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
R a3为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
R a12为H、C 1-6烷基或C 1-6卤代烷基(优选为H或甲基);
R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基(优选为H或F);
环B选自:
Figure PCTCN2020094532-appb-000055
R b选自H、卤素、-CN、-L-C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-NRC(O)R’和-NRC(O)NRR’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
优选的本发明化合物包括但不限于下面列举的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020094532-appb-000056
Figure PCTCN2020094532-appb-000057
Figure PCTCN2020094532-appb-000058
Figure PCTCN2020094532-appb-000059
Figure PCTCN2020094532-appb-000060
Figure PCTCN2020094532-appb-000061
Figure PCTCN2020094532-appb-000062
Figure PCTCN2020094532-appb-000063
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理缔合可包括氢键键合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、乙醚等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂合物包括药学上可接受的溶剂合物且进一步包括化学计量的溶剂合物和非化学计量的溶剂合物。在一些情况下,所述溶剂合物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂合物”包括溶液状态的溶剂合物和可分离的溶剂合物。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R·x H 2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R·0.5 H 2O))和多水合物(x为大于1的数,例如,二水合物(R·2 H 2O)和六水合物(R·6 H 2O))。
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元 素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。
本发明还包括同位素标记的化合物,它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如 3H和 14C)的那些可用于药物和/或底物组织分布测定。氚、即 3H和碳-14、即 14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即 2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(I)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(I)化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
本发明还提供药物制剂,包含治疗有效量的式(I)化合物或其治疗学上可接受的盐和其药学上可接受的载体、稀释剂或赋形剂。所有这些形式都属于本发明。
药物组合物、制剂和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的本发明化合物。在一些实施方案中,所述药物组合物包含治疗有效量的本发明化合物。在一些实施方案中,所述药物组合物包含预防有效 量的本发明化合物。
用于本发明的药学上可接受的赋形剂是指不会破坏一起调配的化合物的药理学活性的无毒载剂、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括(但不限于)离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
给药
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度提高至有效水平。推注剂量取决于通过身体的活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注(例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药物组 合物,而后持续输液。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington′s Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材 料的描述可在Remington′s Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。
治疗
如本文中所述,已知ATR激酶在肿瘤发生及许多其它疾病中起作用。我们已经发现式(I)化合物具有强有力的抗肿瘤活性,所述抗肿瘤活性被认为通过抑制ATR激酶获得。
从而,本发明化合物具有作为抗肿瘤药剂的价值。尤其,本发明化合物具有在实体和/或液体肿瘤疾病的遏制和/或治疗中作为抗增殖、凋亡和/或抗侵袭药剂的价值。尤其,预期本发明化合物有用于预防或治疗对抑制ATR敏感的那些肿瘤。此外,预期本发明化合物有用于预防或治疗单独或部分由ATR介导的那些肿瘤。因此,所述化合物可用于在需要此类治疗温血动物中产生ATR酶抑制作用。
如本文中所述,ATR激酶的抑制剂应对增生性疾病(如癌症),尤其实体肿瘤(如癌和肉瘤)及白血病和淋巴癌的治疗,尤其对于例如乳腺癌、结肠直肠癌、肺癌(包括小细胞肺癌、非小细胞肺癌和细支气管肺泡癌)和前列腺癌及胆管癌、骨癌、膀胱癌、头及颈癌、肾癌、肝癌、胃肠组织癌、食道癌、卵巢癌、胰腺癌、皮肤癌、睾丸癌、甲状腺癌、子宫癌、子宫颈癌和外阴癌、以及白血病[包括急性淋巴细胞白血病(ALL)和慢性髓细胞源性白血病(CML)]、多发性骨髓瘤和淋巴癌的治疗具有治疗价值。
有用于治疗患者中的癌症的抗癌作用包括但不局限于抗肿瘤作用、响应率、疾病进展的时间及存活率。本发明治疗方法的抗肿瘤作用包括但不局限于肿瘤生长的抑制、肿瘤生长的延迟、肿瘤的退化、肿瘤的收缩、治疗停止后肿瘤再生长时间的延长及疾病进展的减慢。抗癌作用包括预防性治疗以及现存在疾病的治疗。
ATR激酶抑制剂或其药学上可接受的盐还有用于治疗癌症患者,所述癌症包括但不局限于血癌,如白血病、多发性骨髓瘤;淋巴瘤,如霍奇金病、非霍奇金淋巴瘤(包括套细胞淋巴瘤)和骨髓增生异常综合征,以及还有实体肿瘤及其转移灶(metastases),如乳腺癌、肺癌(非小细胞肺癌(NSCL)、小细胞肺癌(SCLC)、鳞状细胞癌)、子宫内膜癌、中枢神经系统肿瘤(如胶质瘤、胚胎期发育不良性神经上皮肿瘤、多形性成胶质细胞瘤、混合型胶质瘤、髓母细胞瘤、成视网膜细胞瘤、成神经细胞瘤、生殖细胞瘤和畸胎瘤、胃肠道癌(如胃癌)、食道癌、肝细胞(肝)癌、胆管癌、结肠和直肠癌、小肠癌、胰腺癌、皮肤癌如黑素瘤(尤其转移性黑素瘤)、甲状腺癌、头及颈癌和唾液腺癌、前列腺癌、睾丸癌、卵巢癌、子宫颈癌、子宫癌、外阴癌、膀胱癌、肾癌(包括肾细胞癌、明细胞和肾嗜酸细胞瘤)、鳞状细胞癌、肉瘤如骨肉瘤、软骨肉瘤、平滑肌肉瘤、软组织肉瘤、尤因肉瘤、胃肠道间质瘤(GIST)、卡波西肉瘤和儿科癌如横纹肌肉瘤和成神经细胞瘤。
预期本发明化合物以及包括给予或使用ATR激酶抑制剂或其药学上可接受的盐的治疗方法特别有用于治疗患有肺癌、前列腺癌、黑素瘤、卵巢癌、乳腺癌、子宫内膜癌、肾癌、胃癌、肉瘤、头及颈癌、中枢神经系统的肿瘤及其转移灶的患者,还有用于治疗患有急性髓细胞白血病的患者。
本发明化合物的有效量通常在平均日剂量为0.01mg至50mg化合物/千克患者体重,优选0.1mg至25mg化合物/千克患者体重,以单次或多次给药。通常,本发明化合物可向该有此治疗需要的患者以每位患者约1mg至约3500mg的日剂量范围给药,优选10mg至1000mg。例如,每位患者的日剂量可为10、20、30、40、50、60、70、80、90、100、150、200、250、300、350、400、500、600、700、800、900或1000mg。可每天、每周(或间隔数天)或以间歇时间表,给药一次或多次。例如,可在每周的基础上(例如每周一),每天给予所述化合物一次或多次,不定地或持续几周,例如4-10周。或者,可每天给药持续几天(例如2-10天),然后几天(例如1-30天)不给药所述化合物,不定地重复该循环或重复给定的次数,例如4-10个循环。例如,本发明化合物可每天给药持续5天,然后间断9天,然后再每天给药持续5天,然后间断9天,以此类推,不定地重复该循环或共重复4-10次。
组合治疗
本文中定义的治疗可作为单独治疗应用,或除本发明化合物之外,可包括常规外科手术或放疗或化疗。因此,本发明化合物还可与用于治疗癌症的现有治疗药剂联合使用。
联合使用的合适的药剂包括:
(1)内科肿瘤学所用的抗增殖/抗肿瘤药物及其组合,如烷化剂(例如顺铂、卡铂、环磷酰胺、氮芥、美法仑(melphalan)、苯丁酸氮芥、白消安(Busulphan)和亚硝基脲);抗代谢药(例如抗叶酸剂,如诸如5-氟尿嘧啶和替加氟(tegafur)之类的氟尿嘧啶、雷替曲塞(raltitrexed)、甲氨碟呤、阿糖胞苷、羟基脲和吉西他滨(gemcitabine));抗肿瘤抗生素(例如蒽环类,如阿霉素(adriamycin)、博莱霉素(bleomycin)、多柔比星(doxorubicin)、道诺霉素(daunomycin)、表柔比星(epirubicin)、伊达比星(idarubicin)、丝裂霉素C(mitomycin-C)、放线菌素D(dactinomycin)和光神霉素(mithramycin));抗有丝分裂药剂(例如长春花生物碱,如长春新碱、长春碱、长春地辛和长春瑞滨;和紫杉烷类,如紫杉醇和多西他赛(taxotere));和拓扑异构酶抑制剂(例如表鬼臼毒素,如依托泊苷(etoposide)和替尼泊苷(teniposide)、安吖啶(amsacrine)、拓扑替康(topotecan)和喜树碱(camptothecin));
(ii)细胞生长抑制剂,如抗雌激素(例如他莫昔芬(tamoxifen)、托瑞米芬(toremifene)、雷洛昔芬(raloxifene)、屈洛昔芬(droloxifene)和艾多昔芬(iodoxyfene))、雌激素受体下调剂(例如氟维司群(fulvestrant))、抗雄激素(例如比卡鲁胺(bicalutamide)、氟他胺(flutamide)、尼鲁米特(nilutamide)和乙酸环丙孕酮)、LHRH拮抗剂或LHRH激动剂(例如戈舍瑞林(goserelin)、亮丙瑞林(leuprorelin)和布舍瑞林(buserelin))、孕激素(例如乙酸甲地孕酮)、芳化酶抑制剂(例如阿那曲唑(Anastrozole)、来曲唑(letrozole)、伏氯唑(vorazole)和依西美坦(exemestane))和5α-还原酶的抑制剂,如非那雄胺(finasteride);
(iii)抗侵袭药剂(例如c-Src激酶家族抑制剂,如AZD0530和达沙替尼)和金属蛋白酶抑制剂,如马立马司他(marimastat);以及尿激酶型纤溶酶原激活物受体功能的抑制剂);
(iv)生长因子功能抑制剂:例如生长因子抗体和生长因子受体抗体(例如抗erbB2抗体曲妥珠单抗 [Herceptin TM]和抗erbBl抗体西妥昔单抗[C225]);此类抑制剂还包括例如酪氨酸激酶抑制剂,例如表皮生长因子家族的抑制剂(例如EGFR家族酪氨酸激酶抑制剂,如吉非替尼、厄洛替尼和CI 1033;和erbB2酪氨酸激酶抑制剂,如拉帕替尼);肝细胞生长因子家族的抑制剂;血小板衍生生长因子家族的抑制剂,如伊马替尼;丝氨酸/苏氨酸激酶的抑制剂(例如Ras/Raf信号传导抑制剂,如法尼基转移酶抑制剂,例如索拉非尼(BAY 43-9006))和通过MEK和/或Akt激酶的细胞信号传导的抑制剂;
(v)抗血管生成药剂,如抑制血管内皮生长因子的作用的那些药剂[例如抗血管内皮细胞生长因子抗体贝伐珠单抗(Avastin TM);和VEGF受体酪氨酸激酶抑制剂,如ZD6474、AZD2171、伐他拉尼(vatalanib)和舒尼替尼(sunitinib),和通过其它机理起作用的化合物(例如利诺胺(linomide)、整联蛋白αvβ3功能抑制剂和血管生成抑制素)];
(vi)血管破坏剂,如考布他汀(combretastatin);
(vii)反义疗法,例如针对以上列出的靶标的那些,如ISIS 2503;
(viii)基因治疗方法,包括置换异常基因(如异常ρ53或异常BRCA1或BRCA2)的方法;GDEPT(基因导向的(gene-directed)酶前药治疗)方法,如使用胞嘧啶脱氨酶、胸苷激酶或细菌硝基还原酶的那些方法;和提高患者对化疗或放疗的耐受性的方法,如多药耐药性基因疗法;和
(ix)免疫治疗方法,包括提高患者的肿瘤细胞的免疫原性的体外和体内方法,如用细胞因子(如白介素2、白介素4或粒细胞-巨噬细胞集落刺激因子)转染;降低T-细胞麻痹(anergy)的方法;使用转染的免疫细胞(如细胞因子转染的树突细胞)的方法;使用细胞因子转染的肿瘤细胞系的方法和使用抗独特型抗体的方法。
实施例
本文所用的材料或试剂为可购买到的或由本领域通常已知的合成方法制备。以下反应路线例示了本发明化合物的具体合成方法。
关键中间体a1-a13的制备:
Figure PCTCN2020094532-appb-000064
在反应瓶中加入2,4,6-三氯嘧啶(13.74mmol,2.5g),(R)-2-甲基吗啉(13.8mmol,1.4g)和DIPEA(30mmol,3.87g),50mL乙腈溶解后,90℃反应2小时。停止反应,旋干溶剂,加水50mL,乙酸乙酯萃取,无水Na 2SO 4干燥,柱层析分离,得到中间体a1(2.3g,收率:68%),LC-MS:[M+H] +:248。
Figure PCTCN2020094532-appb-000065
冰浴条件下,在反应瓶中加入4-溴-7-氮杂吲哚(25.5mmol,5g)和对甲苯磺酰氯(38.25mmol,7.27g),100mL无水四氢呋喃溶解,缓慢加入60%NaH(30mmol,1.2g),室温反应6小时。停止反应,缓慢加水50mL,旋干有机溶剂,二氯甲烷萃取,无水Na 2SO 4干燥,柱层析分离,得到中间体a2(9.0g,收率定量),LC-MS:[M+H] +:350。
氮气保护下,将中间体a2(25.5mmol,9.0g)和双联硼酸频哪醇酯(30.6mmol,7.77g)溶于1,4-二氧六环中,加入醋酸钾(51mmol,5g)和Pd(dppf)Cl 2(2.5mmol,1.83g),90℃反应12小时。停止反应,过滤,旋干有机溶剂,柱层析分离,得到中间体a3(10.0g,收率:98%),LC-MS:[M+H] +:399。
参照中间体a3合成如下中间体:
Figure PCTCN2020094532-appb-000066
Figure PCTCN2020094532-appb-000067
氮气保护下,将中间体a1(4mmol,1.0g)和中间体a3(6mmol,2.4g)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钠(8mmol,848mg)和Pd(dppf)Cl 2(0.4mmol,293mg),加热回流反应12小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到中间体a4(700mg)和中间体a5(400mg),总收率:57%,LC-MS:[M+H] +:484。
Figure PCTCN2020094532-appb-000068
氮气保护下,将中间体a1(2.0mmol,500mg)和3,5-二甲基-1H-吡唑-4-硼酸频哪醇酯a6(2.0mmol, 440mg)溶于15mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(6.1mmol,830mg)和Pd(dppf)Cl 2(0.4mmol,293mg),加热回流反应12小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到中间体a7(200mg,收率:33%)和中间体a8(60mg,收率:10%),LC-MS:[M+H] +:308。
Figure PCTCN2020094532-appb-000069
氮气保护下,将中间体a4(1.04mmol,500mg)和中间体a6(1.1mmol,253mg)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钾(3.2mmol,430mg)和Pd(dppf)Cl 2(0.2mmol,151mg),加热回流反应12小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到中间体a9(320mg,收率:57%),LC-MS:[M+H] +:544。
Figure PCTCN2020094532-appb-000070
氮气保护下,将中间体a1(2.0mmol,500mg)和1,3,5-二甲基-1H-吡唑-4-硼酸频哪醇酯a10(2.0mmol,472mg)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钠(6.0mmol,636mg)和Pd(dppf)Cl 2(0.2mmol,151mg),加热回流反应16小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到中间体a11(280mg,收率:43%)和中间体a12(110mg,收率:17%)LC-MS:[M+H] +:322。
关键中间体b1-b18的制备:
Figure PCTCN2020094532-appb-000071
冰浴条件下,在反应瓶中加入4-溴-7-氮杂吲哚(10.2mmol,2g),50mL DMF溶解,缓慢加入60%NaH(15.2mmol,610mg)。半小时后,加入SEMCl(15.2mmol,2.5g),室温下继续反应1小时。停止反应,缓慢加水100mL,乙酸乙酯萃取,无水Na 2SO 4干燥,得到中间体b1(收率定量),LC-MS:[M+H] +:327。
氮气保护下,将中间体b1(10.2mmol,3.3g)和双联硼酸频哪醇酯(20.3mmol,5.2g)溶于1,4- 二氧六环中,加入醋酸钾(20.3mmol,2.0g)和Pd(dppf)Cl 2(1.1mmol,742mg),90℃反应3小时。停止反应,过滤,旋干有机溶剂,柱层析分离,得到中间体b2(5.8g,收率:70%),LC-MS:[M+H] +:375。
Figure PCTCN2020094532-appb-000072
在-78℃,将1,5-二甲基-4-溴-3-三氟甲基-1-H-吡唑(0.41mmol,100mg)溶于10mL无水THF中,逐滴加入丁基锂(0.18mL,2.5M),30分钟后,加入异丙氧基硼酸频哪醇酯(0.49mmol,91mg),-78℃继续反应3小时后,加入20mL饱和氯化铵水溶液淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,直接用于下一步反应。LC-MS:[M+H] +:291。
Figure PCTCN2020094532-appb-000073
氮气保护下,将化合物3-溴-4-甲基苯基甲基砜(2.0mmol,500mg)和双联硼酸频哪醇酯(4.0mmol,1.0g)溶于10mL 1,4-二氧六环中,加入醋酸钾(6.0mmol,590mg)和Pd(dppf)Cl 2(0.2mmol,150mg),90℃反应2小时。停止反应,过滤,减压蒸除有机溶剂,得到中间体b4(收率定量),LC-MS:[M+H] +:297。
Figure PCTCN2020094532-appb-000074
在冰浴条件下,将化合物N,N-二甲基对甲苯磺酰胺b5-2(5mmol,1g)溶于12mL浓硫酸中,加入NBS(5.5mmol,983mg),缓慢升温至室温,并搅拌3小时。将该混合液缓慢倒入冰水中淬灭反应,抽滤,干燥,得到化合物b5-1(1.3g,收率:93%),LC-MS:[M+H] +:278。
氮气保护下,将化合物b5-1(2.2mmol,600mg)和双联硼酸频哪醇酯(4.3mmol,1.1g)溶于15mL 1,4-二氧六环中,加入醋酸钾(6.5mmol,635mg)和Pd(dppf)Cl 2(0.22mmol,158mg),100℃反应2小时。停止反应,过滤,减压蒸除有机溶剂,柱层析分离,得到中间体b5(收率定量),LC-MS:[M+H] +:326。
Figure PCTCN2020094532-appb-000075
将中间体a6(1.0mmol,222mg)溶于10mL DMF中,加入碳酸铯(1.0mmol,326mg)和3-溴 丙基苄醚(1.2mmol,275mg),70℃反应12小时。停止反应,加水,乙酸乙酯萃取,饱和食盐水洗涤,无水硫酸钠干燥,过滤,柱层析分离,得到油状中间体b6-1(260mg,收率:70%),LC-MS:[M+H] +:371。
将中间体b6-1(0.27mmol,100mg)溶于5mL的乙醇溶液中,加入20mg钯碳,催化加氢下(4atm)反应过夜。抽滤,得到白色固体b6(88mg,收率定量),LC-MS:[M+H] +:281。
Figure PCTCN2020094532-appb-000076
氮气保护下,将化合物4-溴-2-二氟甲基吡啶(1.7mmol,350mg)和四羟基二硼酸(3.2mmol,288mg)溶于10mL乙醇中,加入醋酸钾(6.4mmol,630mg),X-Phos(0.21mmol,102mg)和X-Phos-Pd-G3(0.11mmol,90mg),80℃反应3小时。停止反应,过滤,减压蒸除有机溶剂,得到中间体b7,LC-MS:[M+H] +:174。
Figure PCTCN2020094532-appb-000077
在反应瓶中加入对甲氧基苯硫醇(7.2mmol,1.0g),氢氧化钾(10.8mmol,604mg),20mL乙醇溶解,逐滴加入溴代异丙烷(7.2mmol,878mg),50℃反应。TLC监测反应完全,过滤,减压蒸除溶剂,柱层析分离,得到中间体b8-2,收率定量。
冰浴下,将中间体b8-2(8.24mmol,1.5g)溶于30mL二氯甲烷中,缓慢加入m-CPBA(20mmol,3.4g),室温反应过夜。加水50mL,二氯甲烷萃取,得到中间体b8-1,收率定量。
以中间体b8-1为原料,参照中间体b5的合成,得到中间体b8,收率:42%。
Figure PCTCN2020094532-appb-000078
在反应瓶中加入中间体a6(0.23mmol,50mg),NaH(0.25mmol,9mg),8mL DMF溶解,逐滴加入1,2-环氧基-2-甲基丙烷(0.25mmol,18mg),80℃反应过夜,加水15mL,10mL乙酸乙酯萃取,减压蒸除溶剂,柱层析分离,得到中间体b9,收率:44%,LC-MS:[M+H] +:295。
Figure PCTCN2020094532-appb-000079
冰浴下,将中间体b10-1(2.06mmol,500mg)溶于10mL DMSO中,缓慢加入异丙基硫醇(2.47mmol,188mg),室温反应2小时。加水50mL,乙酸乙酯萃取,得到粗品中间体b10-2,直接进行下一步反应。
冰浴下,将上步粗品b10-2溶于30mL二氯甲烷中,缓慢加入m-CPBA(12.5mmol,2.1g),室温反应10小时。停止反应,加水50mL,二氯甲烷萃取,flash柱层析分离,得到690mg中间体b10-3,两步总收率:99%。
氮气保护下,将中间体b10-3(0.91mmol,300mg)和双联硼酸频哪醇酯(1.09mmol,276mg)溶于12mL 1,4-二氧六环中,加入醋酸钾(2.7mmol,266mg)和Pd(dppf)Cl 2(0.18mmol,132mg),100℃下反应2小时。停止反应,过滤,减压蒸除有机溶剂,柱层析分离,得到中间体b10(240mg,收率:70%)。
Figure PCTCN2020094532-appb-000080
冰浴下,将原料甲基乙基胺(3.57mmol,211mg)溶于8mL THF中,缓慢滴加原料b11-1(2.38mmol,680mg),室温反应2小时后停止反应。加水30mL,乙酸乙酯萃取,flash柱层析分离,得到中间体b11-2(450mg,收率:62%)。LC-MS:[M+H] +:309。
氮气保护下,将中间体b11-2(1.46mmol,450mg)和双联硼酸频哪醇酯(1.75mmol,445mg)溶于12mL 1,4-二氧六环中,加入醋酸钾(2.92mmol,291mg)和Pd(dppf)Cl 2(0.10mmol,107mg),100℃下反应2小时。停止反应,过滤,减压蒸除溶剂,flash柱层析分离,得到中间体b11(480mg,收率:93%)。LC-MS:[M+H] +:356。
Figure PCTCN2020094532-appb-000081
冰浴下,将原料甲基乙基胺(20.3mmol,1.2g)溶于15mL THF中,缓慢滴加原料b12-1(6.8mmol,1.5g),室温反应2小时后停止反应。加水40mL,乙酸乙酯萃取,flash柱层析分离,得到中间体 b12-2(1.6g,收率:97%)。
冰浴下,缓慢将中间体b12-2(2.07mmol,500mg)溶于3mL浓硫酸中,分批加入N-溴代琥珀酰亚胺(2.18mmol,388mg),室温反应2小时后停止反应。将反应液缓慢倒入30mL冰水混合物中,加入饱和碳酸氢钠水溶液调节pH~10左右,乙酸乙酯萃取,得到粗品中间体b12-3。
氮气保护下,将粗品b12-3(550mg)和双联硼酸频哪醇酯(2.6mmol,655mg)溶于10mL 1,4-二氧六环中,加入醋酸钾(5.1mmol,505mg)和Pd(dppf)Cl 2(0.17mmol,126mg),100℃下反应2小时。停止反应,过滤,减压蒸除溶剂,得到中间体b12,直接用于下一步反应。LC-MS:[M+H] +:368。
Figure PCTCN2020094532-appb-000082
冰浴下,将原料b13-1(23.5mmol,2.4g)溶于15mL THF中,缓慢滴加原料对甲苯磺酰氯(7.85mmol,1.5g),室温反应2小时后停止反应。加水40mL,乙酸乙酯萃取,flash柱层析分离,得到中间体b13-2(2.0g,收率:99%)。
冰浴下,缓慢将中间体b13-2(1.95mmol,500mg)溶于3mL浓硫酸中,分批加入N-溴代琥珀酰亚胺(2.05mmol,365mg)后,室温反应2小时。将反应液缓慢倒入30mL冰水混合物中,加入饱和碳酸氢钠水溶液调节pH~10左右,乙酸乙酯萃取,得到900mg油状粗品中间体b13-3。
氮气保护下,将中间体b13-3(550mg,粗品)和双联硼酸频哪醇酯(2.5mmol,626mg)溶于10mL 1,4-二氧六环中,加入醋酸钾(4.9mmol,483mg)和Pd(dppf)Cl 2(0.16mmol,120mg),100℃下反应2小时。停止反应,过滤,减压蒸除溶剂,得到中间体b13,直接用于下一步反应。LC-MS:[M+H] +:383。
参照化合物b13合成如下中间体:
Figure PCTCN2020094532-appb-000083
Figure PCTCN2020094532-appb-000084
将原料对甲苯硫酚(0.81mmol,100mg),碳酸钾(2.41mmol,333mg)和4-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.97mmol,270mg)溶于8mL DMF中,升温至70℃反应2小时。停止反应,加水30mL,乙酸乙酯萃取,浓缩,得到230mg粗品中间体b15-1。
冰浴下,将中间体b15-1(3.09mmol,1.0g)溶于50mL二氯甲烷中,缓慢加入间氯过氧苯甲酸(15.4mmol,2.66g),室温下反应4小时。向反应液中加入50mL冰水,乙酸乙酯萃取,flash柱层析分离,得到中间体b15-2(600mg,收率:58%)。LC-MS:[M+H] +:340。
将中间体b15-2(1.77mmol,600mg)溶于6mL二氯甲烷中,缓慢加入2mL三氟乙酸,室温下反应1小时。停止反应,减压蒸除溶剂。向反应液中加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,得到中间体b15-3。LC-MS:[M+H] +:240。
将中间体b15-3(1.77mmol,423mg)溶于5mL甲醇中,加入2mL 40%甲醛水溶液和4滴醋酸。搅拌10分钟后加入NaCNBH 3(8.8mmol,555mg),室温下反应3小时。停止反应,减压蒸除溶剂。向反应液中加入30mL饱和碳酸氢钠水溶液,二氯甲烷萃取,flash柱层析分离,得到中间体b15-4(收率:89%)。LC-MS:[M+H] +:254。
冰浴下,缓慢将中间体b15-4(1.58mmol,400mg)溶于3mL浓硫酸中,分批加入N-溴代琥珀酰亚胺(1.74mmol,307mg),室温继续反应3小时。将反应液缓慢倒入30mL冰水混合物中,并用1N NaOH溶液调节体系pH~8左右,二氯甲烷萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体b15-5(300mg,收率:58%)。LC-MS:[M+H] +:333。
氮气保护下,将中间体b15-5(0.57mmol,190mg)和双联硼酸频哪醇酯(0.68mmol,174mg)溶于5mL 1,4-二氧六环中,加入醋酸钾(1.72mmol,168mg)和Pd(dppf)Cl 2(0.11mmol,84mg),100℃下反应2小时。停止反应,过滤,减压蒸除溶剂,得到中间体b15,直接用于下一步反应。LC-MS:[M+H] +:380。
Figure PCTCN2020094532-appb-000085
将原料对甲苯硫酚(4.01mmol,500mg),氢氧化钾(10.05mmol,553mg)和2-溴丙烷(6.04mmol, 737mg)溶于12mL乙醇中,升温至50℃反应8小时。停止反应,加水30mL,二氯甲烷萃取,得到粗品中间体b16-1。
冰浴下,将中间体b16-1(1.81mmol,300mg)溶于10mL甲醇和2mL水的混合溶液中,缓慢加入单过硫酸氢钾Oxone(4.5mmol,278mg),室温下反应4小时。向反应液中加入30mL水,乙酸乙酯萃取,flash柱层析分离,得到中间体b16-2(350mg,收率:99%)。
冰浴下,缓慢将中间体b16-2(1.76mmol,350mg)溶于3mL浓硫酸中,分批加入N-溴代琥珀酰亚胺(1.33mmol,235mg),室温下继续反应3小时。将反应液缓慢倒入30mL冰水混合物中,并用1N NaOH溶液调节pH~8左右,二氯甲烷萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体b16-3(350mg,收率:95%)。
氮气保护下,将中间体b16-3(0.9mmol,250mg)和双联硼酸频哪醇酯(1.08mmol,276mg)溶于8mL 1,4-二氧六环中,加入醋酸钾(1.8mmol,176mg)和Pd(dppf)Cl 2(0.09mmol,66mg),100℃下反应2小时。停止反应,过滤,减压蒸除溶剂,得到中间体b16,直接用于下一步反应。
Figure PCTCN2020094532-appb-000086
-10℃下,将原料3-溴-4-甲基-苯胺(5.37mmol,1.0g)和CuCl 2(0.54mmol,60mg)溶于20mL浓盐酸中,缓慢加入2mL的亚硝酸钠(5.9mmol,410mg)水溶液并搅拌30分钟。将二氯亚砜(24mmol,2.89g)缓慢溶于10mL水中并将该溶液缓慢滴加至反应体系中,继续在-10℃下搅拌1小时。升温至室温,反应2小时后停止反应。向反应液中加入40mL冰水,二氯甲烷萃取,无水硫酸钠干燥,得到中间体b17-1(250mg,收率:17%)。
冰浴下,将原料4-羟基丁胺(1.13mmol,84mg)和三乙胺(1.85mmol,188mg)溶于10mL二氯甲烷中,缓慢加入中间体b17-1(0.93mmol,250mg),室温反应6小时后停止反应。加入30mL水,二氯甲烷萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体b17-2(150mg,收率:53%)。LC-MS:[M+H] +:309。
氮气保护下,将中间体b17-2(0.49mmol,150mg)和双联硼酸频哪醇酯(0.58mmol,150mg)溶于8mL 1,4-二氧六环中,加入醋酸钾(1.46mmol,160mg)和Pd(dppf)Cl 2(0.097mmol,70mg),100℃下反应2小时。停止反应,过滤,减压蒸除溶剂,得到中间体b17,直接用于下一步反应。LC-MS:[M+H] +:356。
Figure PCTCN2020094532-appb-000087
冰浴下,将原料a6(0.45mmol,100mg)溶于8mL DMF中,缓慢加入NaH(0.54mmol,22mg)。搅拌30分钟后,逐滴加入氯甲基硫醚(0.54mmol,52mg),升温至60℃反应10小时后停止反应。缓慢向体系加入30mL水,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体b18(60mg,收率:47%)。LC-MS:[M+H] +:283。
关键中间体c1-c15的制备:
Figure PCTCN2020094532-appb-000088
操作步骤同上一致。
参照中间体a3合成的化合物如下:
Figure PCTCN2020094532-appb-000089
Figure PCTCN2020094532-appb-000090
在反应瓶中加入原料c3-1(36.7mmol,7g),碳酸铯(92.1mmol,30.03g),100mL DMF溶解,缓慢加入原料c3-2(36.8mmol,8.43g)。升温至80℃反应4小时后停止反应,缓慢加水300mL,乙酸乙酯萃取,无水Na 2SO 4干燥,flash柱层析分离,得到中间体c3-3(3.4g,收率:23%)和中间体c4-1(7.0g,收率:47%),LC-MS:[M+H] +:412。
Figure PCTCN2020094532-appb-000091
在-78℃,将中间体c3-3(6.3mmol,2.6g)溶于60mL无水THF中,逐滴加入丁基锂(3.0mL,2.5M),30分钟后,加入异丙氧基硼酸频哪醇酯c3-4(9.5mmol,1.88mL),-78℃继续反应3小时后,加入150mL饱和氯化铵水溶液淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,得到粗品中间体c3。LC-MS:[M+H] +:460。
参照中间体c3的合成路线,以c4-1为原料,得到粗品中间体c4,LC-MS:[M+H] +:460。
参照中间体c3合成如下化合物:
Figure PCTCN2020094532-appb-000092
Figure PCTCN2020094532-appb-000093
在反应瓶中将原料4-溴-7-氮杂吲哚氮氧化物c5-1(4.7mmol,1g)溶于15mL乙酸酐中,135℃下反应8小时。冷却至室温后,加入50mL水,二氯甲烷萃取,无水硫酸钠干燥,柱层析分离,得到中间体c5-2(1.3g,收率:93%)。LC-MS:[M+H] +:297。
将中间体c5-2(4.36mmol,1.3g),碳酸钾(17.5mmol,2.4g)溶于20mL甲醇中,加入5mL水,室温反应12小时后过滤,减压蒸除溶剂,柱层析分离,得到中间体c5-3(235mg,收率:25%)。 LC-MS:[M+H] +:213。
在反应瓶中加入中间体c5-3(0.236mmol,50mg),碳酸钾(1.18mmol,162mg),5mL丙酮溶解。缓慢滴加碘甲烷(0.236mmol,14μL),50℃下反应过夜。停止反应,过滤,减压蒸除溶剂,柱层析分离,得到中间体c5-4(16mg,收率:30%)。LC-MS:[M+H] +:227。
氮气保护下,将中间体c5-4(0.11mmol,25mg)和双联硼酸频哪醇酯(0.13mmol,33mg)溶于4mL 1,4-二氧六环中,加入醋酸钾(0.33mmol,32mg)和Pd(dppf)Cl 2(0.011mmol,9mg),加热回流反应3小时。停止反应,过滤,减压蒸除溶剂后,得到中间体粗品c5。LC-MS:[M+H] +:276。
Figure PCTCN2020094532-appb-000094
-20℃下,在反应瓶中加入原料c6-1(1.0mmol,200mg),12mL乙醇溶解。逐滴加入(R)-2-甲基吗啉(1.1mmol,111mg)的2mL乙醇溶液,室温反应12小时。停止反应,减压蒸出溶剂,柱层析分离,得到中间体c6(127mg,收率:48%),LC-MS:[M+H] +:266。
Figure PCTCN2020094532-appb-000095
在反应瓶中加入原料c3-1(2.19mmol,500mg)和原料c7-1(2.63mmol,731mg),10mL DMF溶解。加入碳酸铯(6.57mmol,2.14g)后80℃下反应5小时。停止反应,过滤,减压蒸出溶剂,flash柱层析分离,得到中间体c7-2(240mg,收率:27%),LC-MS:[M+H-tBu] +:356。
在-78℃,将中间体c7-2(0.6mmol,247mg)溶于6mL无水THF中,逐滴加入丁基锂(0.29mL,2.5M),30分钟后,加入异丙氧基硼酸频哪醇酯c3-4(0.78mmol,155μL),-78℃继续反应3小时后,加入100mL饱和氯化铵水溶液淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,得到粗品中间体c7(240mg,收率:88%)。LC-MS:[M+H] +:460。
Figure PCTCN2020094532-appb-000096
将原料N-甲基-4-哌啶酮c8-1(17.6mmol,2g)和Boc-肼(17.6mmol,2.3g)溶于60mL乙醇中,室温下反应2天。减压蒸除溶剂后,将此混合物置于冰浴下并用20mL醋酸和5mL水溶解,缓慢加入NaBH 3CN(26.5mmol,1.67g),继续反应3小时。停止反应,减压蒸除溶剂,用饱和NaHCO 3水溶液调pH~9左右,二氯甲烷萃取。无水硫酸钠干燥,得到中间体c8-2(3.1g,收率:77%)。LC-MS:[M+H] +:230。
将中间体c8-2(13.5mmol,3.1g)溶于30mL二氯甲烷中,逐滴加入6mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,得到中间体c8-3(收率定量)。
在反应瓶中加入中间体c8-3(6.76mmol,878mg)和三氟乙酰乙酸甲酯(7.44mmol,1.26g),12mL甲醇溶解后升温至回流反应1.5小时。向该混合物中加入1mL浓盐酸后,继续反应12小时。停止反应,减压蒸除溶剂,flash柱层析分离得到中间体c8-4(1.68g,收率定量)。LC-MS:[M+H] +:250。
冰浴下,将中间体c8-4(6.7mmol,1.68g)和碳酸钾(20.2mmol,4.82g)溶于20mL DMF中,缓慢加入碳酸二甲酯(13.4mmol,1.2g),室温反应12小时后,停止反应,加水50mL,乙酸乙酯萃取,减压蒸除溶剂,flash柱层析分离,得到中间体c8-5(400mg,收率:23%)。LC-MS:[M+H] +:264。
冰浴下,将中间体c8-5(1.14mmol,300mg)溶于6mL浓硫酸中,缓慢分批加入NBS(1.14mmol,200mg),室温反应8小时后,将该混合物倾倒至50mL冰水中,用饱和NaHCO 3水溶液调pH至9左右,二氯甲烷萃取,减压蒸除溶剂,无水硫酸钠干燥,得到中间体c8-6(400mg,收率定量)。LC-MS:[M+H] +:342。
在-78℃,将中间体c8-6(1.46mmol,500mg)溶于15mL无水THF中,逐滴加入丁基锂(0.7mL,2.5M),30分钟后,加入异丙氧基硼酸频哪醇酯c3-4(1.75mmol,326mg),-78℃继续反应3小时后,加入50mL饱和氯化铵水溶液淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,得到粗品中间体c8(500mg,收率:87%)。LC-MS:[M+H] +:390。
Figure PCTCN2020094532-appb-000097
冰浴,氮气保护下,在反应瓶中加入原料c3-1(5mmol,1.15g)和2-(三甲基硅烷基)乙氧甲基氯SEMCl(5mmol,0.9mL),25mL THF溶解。分批加入NaH(5mmol,200mg),搅拌1小时后停止反应。缓慢向体系加入50mL冰水淬灭反应,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到混合中间体c9a-1和c9b-1(1.3g,收率:73%),LC-MS:[M+H] +:359。
在-78℃,将混合中间体c9a-1和c9b-1(3.63mmol,1.3g)溶于35mL无水THF中,逐滴加入丁基锂(1.74mL,2.5M),30分钟后,加入异丙氧基硼酸频哪醇酯c3-4(5.45mmol,1.1mL)。-78℃下继续反应3小时,加入100mL饱和氯化铵水溶液淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,得 到粗品中间体c9a和c9b(1.8g)。直接用于下一步反应。LC-MS:[M+H] +:407。
Figure PCTCN2020094532-appb-000098
冰浴,氮气保护下,将原料a6(2.25mmol,500mg)溶于15mL DMF中,分批加入NaH(4.5mmol,180mg),搅拌30分钟后,缓慢加入4-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(3.37mmol,943mg),升温至90℃继续反应2小时。停止反应,缓慢向体系加入50mL冰水淬灭反应,乙酸乙酯萃取,无水硫酸钠干燥,得到粗品中间体c10(2.0g),直接用于下一步反应。LC-MS:[M+H] +:406。
Figure PCTCN2020094532-appb-000099
在反应瓶中加入原料c3-1(2.19mmol,500mg)和原料c11-1(2.63mmol,769mg),10mL DMF溶解。加入碳酸铯(6.57mmol,2.14g)后,80℃下加热反应10小时。停止反应,过滤,减压蒸除溶剂,flash柱层析分离,得到中间体c11-2(170mg,收率:19%),LC-MS:[M+H-tBu] +:331。
在-78℃,将中间体c11-2(0.4mmol,170mg)溶于10mL无水THF中,逐滴加入丁基锂(0.8mL,2.5M),30分钟后,加入异丙氧基硼酸频哪醇酯c3-4(1mmol,200μL),-78℃继续反应3小时后,加入50mL饱和氯化铵水溶液淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,得到粗品中间体c11(100mg,收率:53%)。直接用于下一步反应。LC-MS:[M+H] +:474。
Figure PCTCN2020094532-appb-000100
将原料c14-1(14.3mmol,3.3g)和N-Boc-肼(15.7mmol,2.1g)溶于30mL醋酸中。搅拌5分钟后,缓慢加入NaBH 3CN(43mmol,2.7g),室温下反应10小时。停止反应,向体系中加入100mL冰水淬灭反应,减压蒸除溶剂。用饱和NaHCO 3水溶液调体系pH~9左右,二氯甲烷萃取。无水硫酸钠干燥,flash柱层析分离,得到中间体c14-2(3.2g,收率:65%)。
在反应瓶中加入中间体c14-2(8.83mmol,3.1g)和乙酰丙酮(10.6mmol,1.1mL),30mL醋酸溶解。向该混合物中缓慢加入3mL氢溴酸(45%),室温反应2小时。停止反应,减压蒸除溶剂,得到粗品中间体c14-3(3g,粗品)。LC-MS:[M+H] +:216。
将粗品中间体c14-3(7.8mmol,1.68g)、Boc酸酐(11.7mmol,2.5g)、三乙胺(23.4mmol,3.4mL)和DMAP(0.78mmol,95mg)溶于20mL四氢呋喃中,室温反应12小时。停止反应,加水100 mL,乙酸乙酯萃取,饱和氯化钠水溶液洗涤,无水硫酸钠干燥,得到黄色油状产品(1.37g)。将该黄色油状物溶于10mL DMF中并置于冰浴下,缓慢分批加入NBS(5mmol,882mg),室温反应8小时后,加入50mL冰水,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体c14-4(1.1g,三步总收率:32%)。LC-MS:[M+H] +:395。
氮气保护下,在微波反应瓶中加入中间体c14-4(1.3mmol,500mg),双联硼酸频那醇酯(1.52mmol,300mg),DIEA(3.8mmol,0.66mL)和Pd(Amphos)Cl 2(0.13mmol,90mg),20mL 2-Me-THF和MeOH的混合溶液溶解(v/v,1/1)。微波升温至100℃反应1小时后停止反应。过滤,加水30mL,乙酸乙酯萃取,得到粗品中间体c14。直接用于下一步反应。LC-MS:[M+H] +:442。
Figure PCTCN2020094532-appb-000101
冰浴下,将原料a6(2.0mmol,444mg),N-Boc-哌嗪(3.0mmol,559mg),DIEA(5.0mmol,645mg)溶于15mL四氢呋喃中。搅拌5分钟后,缓慢向体系加入三光气(4.0mmol,1.08g),冰浴下搅拌2小时后停止反应。向该反应液加水40mL,二氯甲烷萃取,flash柱层析分离,得到中间体c15(180mg,收率:21%)。LC-MS:[M+H] +:435。
关键中间体d1-d9的制备:
Figure PCTCN2020094532-appb-000102
操作步骤同上一致。
Figure PCTCN2020094532-appb-000103
在微波反应瓶中加入中间体a1(2.02mmol,500mg)和原料2-氨基苯并咪唑d1-1(2.11mmol,282mg),10mL DMF溶解。加入碳酸铯(4.03mmol,1.3g)后微波100℃下反应1.5小时。停止反应,加水40mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体d1(600mg,收率:86%),LC-MS:[M+H] +:345。
Figure PCTCN2020094532-appb-000104
在微波反应瓶中加入中间体a1(2.02mmol,500mg)和原料2-硝基-4-氟-苯胺d2-1(2.4mmol,375mg),8mL DMF溶解。加入叔丁醇钠(4mmol,384mg)后微波100℃下反应1小时。停止反应,加水30mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体d2-2(700mg,收率:95%),LC-MS:[M+H] +:368。
在反应瓶中加入中间体d2-2(2.72mmol,1g)、还原铁粉(9.5mmol,532mg)和氯化铵(9.5mmol,508mg),加入20mL乙醇和4mL水溶解。100℃下反应1.5小时。停止反应,抽滤,加水60mL,乙酸乙酯萃取,无水硫酸钠干燥,得到中间体d2-3(900mg,收率:98%),LC-MS:[M+H] +:338。
在反应瓶中加入中间体d2-3(1.78mmol,600mg)、溴腈(5.3mmol,565mg),10mL甲醇和4mL水溶解。微波60℃下反应1.5小时。停止反应,加水40mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体d2(400mg,收率:62%),LC-MS:[M+H] +:363。
参照中间体d2合成的化合物如下:
Figure PCTCN2020094532-appb-000105
Figure PCTCN2020094532-appb-000106
氮气保护下,将中间体c6(1mmol,265mg)和中间体a3(1mmol,398mg)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钾(3mmol,414mg)和Pd(dppf)Cl 2(0.1mmol,73mg),微波加热110℃反应30分钟。停止反应,过滤,加水10mL,乙酸乙酯萃取,柱层析分离,得到中间 体d3(100mg)收率:20%,LC-MS:[M+H] +:502。
Figure PCTCN2020094532-appb-000107
氮气保护下,将中间体a1(0.8mmol,200mg)和原料4-吲哚硼酸频哪醇酯d4-1(0.88mmol,214mg)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钾(1.6mmol,221mg)和Pd(dppf)Cl 2(0.08mmol,59mg),加热至110℃反应2小时。停止反应,过滤,加水30mL,乙酸乙酯萃取,柱层析分离,得到中间体d4(50mg)收率:19%,LC-MS:[M+H] +:329。
Figure PCTCN2020094532-appb-000108
氮气保护下,将中间体a1(1.28mmol,317mg)和中间体c1(2.57mmol,1.02g)溶于1,4-二氧六环和水的15mL混合液中(v/v:9/1),加入碳酸钠(5.12mmol,543mg)和Pd(dppf)Cl 2(0.13mmol,95mg),加热回流反应12小时。停止反应,过滤,旋干有机溶剂,加水30mL,乙酸乙酯萃取,柱层析分离,得到中间体d5(300mg),收率:49%,LC-MS:[M+H] +:484。
Figure PCTCN2020094532-appb-000109
在反应瓶中加入中间体d2-3(6.25mmol,2.0g),20mL吡啶溶解。缓慢加入甲基异硫氰酸酯(7.5mmol,548mg),90℃下反应0.5小时。降温至室温,加入EDCI(9.38mmol,1.8g),继续升温至90℃并反应10小时,停止反应。向该反应液加入100mL水,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体d7(1.2g,收率:54%),LC-MS:[M+H] +:359。
Figure PCTCN2020094532-appb-000110
氮气保护下,将中间体a1(14.0mmol,3.47g)和原料6-氟-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-吲哚d9-1(21.0mmol,5.48g)溶于1,4-二氧六环和水的80mL混合液中(v/v:9/1),加入碳酸钠(42mmol,4.4g)和Pd(PPh 3)Cl 2(1.4mmol,984mg),加热至90℃并连续搅拌6小时。停止反应,过滤,加水200mL,乙酸乙酯萃取,柱层析分离,得到中间体d9(1.75g,收率:37%), LC-MS:[M+H] +:347。
关键中间体e1的制备:
Figure PCTCN2020094532-appb-000111
氮气保护,-5℃下,将原料e1-1(5mmol,1.05g)溶于15mL氟代苯中,缓慢加入二乙基锌(12.5mL,25mmol,2N的甲苯溶液)和氯碘甲烷的氟苯溶液(4.4g/1.8mL,25mmol,分三次加入),室温反应12小时。将体系置于冰浴下,加饱和氯化铵水溶液40mL淬灭反应,乙酸乙酯萃取,无水硫酸钠干燥,柱层析分离,得到中间体e1-2(730mg,收率:65%)。
氮气保护下,将中间体e1-2(3.23mmol,730mg)溶于16mL甲醇/乙腈(1/1,v/v)的混合液中,缓慢加入KF的水溶液(749mg/3mL)。体系搅拌10分钟后,加入L-酒石酸(6.46mmol,968mg)和四氢呋喃(350μL),室温继续反应1.5h。停止反应,过滤,滤液浓缩,得到中间体e1-3(600mg,粗品)。
氮气保护下,将中间体e1-3(7.4mmol,1.5g),碳酸铯(22.2mmol,7.2g)和2,4,6-三氯嘧啶(7.4mmol,1.35g)溶于36mL甲苯/水(5/1,v/v)的混合液中,加入Pd(dppf)Cl 2(0.74mmol,540mg)。体系加热至110℃反应过夜,停止反应,过滤,浓缩,flash柱层析分离,得到黄色油状中间体e1(500mg,收率:27%),LC-MS:[M+H] +:246。
实施例1:
Figure PCTCN2020094532-appb-000112
氮气保护下,将中间体a4(0.21mmol,100mg)和1-甲基-1H-吡唑-5-硼酸频哪醇酯(0.4mmol,84mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.5mmol,848mg)和Pd(dppf)Cl 2(0.02mmol,15mg),微波下100℃加热反应1小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物A1-1。将化合物A1-1溶于四氢呋喃和甲醇的混合溶液中,加入碳酸铯(0.4mmol,131mg),回流反应过夜至反应完全。过滤,旋干溶剂,制备色谱分离,得到目标化合物A1(收率:41%),LC-MS:[M+H] +:376。
以实施例1路线合成的化合物如下:
Figure PCTCN2020094532-appb-000113
Figure PCTCN2020094532-appb-000114
Figure PCTCN2020094532-appb-000115
实施例2:
Figure PCTCN2020094532-appb-000116
在反应瓶中加入中间体a7(0.33mmol,100mg),碳酸铯(0.66mmol,215mg)和2-溴乙基甲基醚(0.39mmol,59mg),乙腈溶解后,升温回流反应2小时。停止反应,抽滤,柱层析分离,得到化合物A14-2(100mg,收率:83%),LC-MS:[M+H] +:366。
氮气保护下,将中间体A14-2(0.27mmol,100mg)和中间体b2(0.33mmol,123mg)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钾(0.54mmol,75mg),Pd(dppf)Cl 2(0.02mmol,15mg),90℃加热反应12小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物A14-1(120mg,收率:77%),LC-MS:[M+H] +:578。
将化合物A14-1(120mg,0.17mmol)溶于5mL二氯甲烷中,滴加2mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。再次将混合物溶于5mL甲醇中,加入氨水3mL,室温反应过夜。减压蒸除溶剂,制备色谱分离,得到目标化合物A14(50mg,收率:66%),LC-MS:[M+H] +:448。
以实施例2路线合成的化合物如下:
Figure PCTCN2020094532-appb-000117
实施例3:
Figure PCTCN2020094532-appb-000118
在反应瓶中加入中间体a9(0.16mmol,85mg),碳酸铯(0.47mmol,153mg)和N-Boc-4-羟基哌啶甲磺酸酯(0.19mmol,52mg),DMF溶解后,升温至80℃持续反应12小时。停止反应,过滤,加水,乙酸乙酯萃取,柱层析分离,得到化合物A16-1(70mg,收率:61%),LC-MS:[M+H] +:727。
将中间体A16-1(0.1mmol,70mg)溶于6mL二氯甲烷中,滴加2mL三氟乙酸,室温反应2小时。停止反应,减压蒸除有机溶剂。将该混合物再次溶解于10mL甲醇中,加入4N NaOH水溶液2mL,50℃反应2小时。过滤,乙酸乙酯萃取,制备色谱分离,得到目标化合物A16(15mg,收率: 30%),LC-MS:[M+H] +:473。
以实施例3路线合成的化合物如下:
Figure PCTCN2020094532-appb-000119
Figure PCTCN2020094532-appb-000120
Figure PCTCN2020094532-appb-000121
实施例4:
Figure PCTCN2020094532-appb-000122
氮气保护下,将中间体a5(0.21mmol,100mg)和中间体a10(0.4mmol,94mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.5mmol,848mg)和Pd(dppf)Cl 2(0.02mmol,15mg),100℃加热反应12小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物B1-1(80mg,收率:68%),LC-MS:[M+H] +:558。
将化合物B1-1(80mg)溶于6mL乙醇中,加入4N NaOH水溶液3mL,回流反应至反应完全。过滤,加入乙酸乙酯萃取,减压蒸除溶剂,制备色谱分离,得到目标化合物B1(收率定量),LC-MS:[M+H] +:404。
以实施例4路线合成的化合物如下:
Figure PCTCN2020094532-appb-000123
Figure PCTCN2020094532-appb-000124
实施例5:
Figure PCTCN2020094532-appb-000125
-50℃下,将三聚氯氰(110mmol,1.97g)和DIPEA(210mmol,2.76g)溶于70mL二氯甲烷中,逐滴缓慢加入(R)-2-甲基吗啉(110mmol,1.08g)。-50℃搅拌1小时后,加入1N HCl水溶液50mL淬灭反应,二氯甲烷萃取。无水硫酸钠干燥,过滤,旋干溶剂,得到C1-3(2.5g,收率:92%)。LC-MS:[M+H] +:249。
在氮气保护下,将C1-3(0.4mmol,100mg),中间体a10(0.4mmol,95mg),碳酸钾(1.2mmol,166mg)和Pd(PPh 3) 4(0.04mmol,46mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),80℃加热反应12小时。停止反应,过滤,减压蒸除有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物C1-2(80mg,收率:41%),LC-MS:[M+H] +:323。
在氮气保护下,将C1-2(0.25mmol,80mg),中间体b2(0.3mmol,112mg),碳酸钾(0.75mmol,103mg)和Pd(dppf)Cl 2(0.025mmol,18mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),100℃反应3小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物C1-1(90mg,收率:68%),LC-MS:[M+H] +:535。
将C1-1(0.17mmol,90mg)溶于5mL二氯甲烷中,滴加2mL三氟乙酸,室温下反应2小时。停止反应,减压蒸除有机溶剂。再次将该混合物溶于5mL甲醇中,滴加氨水2mL,室温反应12小时。减压蒸除溶剂,制备色谱分离,得到化合物C1(37mg,收率:53%),LC-MS:[M+H] +:405。
1H NMR(400MHz,DMSO-d 6)δ11.91(s,1H),8.40(d,J=5.0Hz,1H),8.05(d,J=4.9Hz,1H),7.65(t,J=2.9Hz,1H),7.21(d,J=2.6Hz,1H),4.03(d,J=11.0Hz,1H),3.83(d,J=12.7Hz,1H),3.75(s,3H),3.71-3.66(m,1H),3.52(d,J=11.4Hz,1H),3.30(d,J=6.7Hz,2H),2.73(s,3H),2.54(s,4H),1.34(s,3H).
实施例6:
Figure PCTCN2020094532-appb-000126
在氮气保护下,将中间体a12(0.19mmol,60mg),中间体b7(0.37mmol,64mg),碳酸钾(0.57mmol,78mg)和Pd(dppf)Cl 2(0.02mmol,14mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),100℃反应2小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,制备色谱分离,得到化合物B8(24mg,收率:31%),LC-MS:[M+H] +:415。
1H NMR(400MHz,DMSO-d 6)δ8.85(d,J=5.2Hz,1H),8.47(s,1H),8.35(d,J=5.2Hz,1H),7.33(s,1H),7.05(t,J=55.0Hz,1H),4.66(s,1H),4.24(d,J=13.4Hz,1H),4.00(d,J=11.7Hz,1H),3.79(d,J=11.4Hz,1H),3.73(s,3H),3.69-3.63(m,1H),3.50(d,J=10.0Hz,1H),3.30-3.24(m,1H),2.66(s,3H),2.49(d,J=1.6Hz,3H),1.25(d,J=6.8Hz,3H).
实施例7:
Figure PCTCN2020094532-appb-000127
将实施例6中的中间体a12替换为中间体a11,参照其路线得到目标化合物A25(17mg,收率:20%),LC-MS:[M+H] +:415。
1H NMR(400MHz,DMSO-d 6)δ8.84(d,J=5.1Hz,1H),8.49(s,1H),8.43-8.38(m,1H),7.08(t,J=54.9Hz,1H),6.69(s,1H),4.56(s,1H),4.26(d,J=13.8Hz,1H),4.00(d,J=10.1Hz,1H),3.79(d,J=11.5Hz,1H),3.74(s,3H),3.70-3.64(m,1H),3.52(t,J=11.3Hz,1H),3.29-3.20(m,1H),2.52(s,3H),2.38(s,3H),1.26(d,J=6.7Hz,3H).
实施例8:
Figure PCTCN2020094532-appb-000128
在氮气保护下,将中间体a11(0.31mmol,100mg),4-Boc-氨基-苯硼酸(0.34mmol,81mg),碳酸钾(0.93mmol,129mg)和Pd(dppf)Cl 2(0.06mmol,45mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),90℃反应12小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层 析分离,得到化合物A26-2(124mg,收率:84%),LC-MS:[M+H] +:479。
将中间体A26-2(0.25mmol,124mg)溶于5mL二氯甲烷中,滴加三氟乙酸2mL,室温反应2小时。减压蒸除溶剂,得到A26-1,LC-MS:[M+H] +:379。
冰浴条件下,将中间体A26-1(0.25mmol)和三乙胺(1.4mmol,135mg)溶于10mL无水四氢呋喃中,滴加乙基异氰酸酯(0.29mmol,20mg),连续搅拌反应1小时后用饱和氯化铵水溶液20mL淬灭反应,减压蒸除溶剂,制备色谱分离,得到化合物A26(40mg,收率:36%),LC-MS:[M+H] +:450。
实施例9:
Figure PCTCN2020094532-appb-000129
参照化合物A18的合成,同时得到化合物A27,LC-MS:[M+H]+:458。
1H NMR(400MHz,DMSO-d 6)δ11.92(s,1H),8.35(d,J=5.0Hz,1H),7.99(d,J=5.1Hz,1H),7.59(d,J=3.4Hz,1H),7.20(s,1H),6.83(s,1H),4.69-4.47(m,5H),4.28(s,1H),4.03(d,J=11.2Hz,1H),3.82(d,J=12.0Hz,1H),3.70(d,J=11.1Hz,1H),3.55(t,J=12.4Hz,1H),3.35(s,1H),3.12(s,2H),2.64(s,6H),2.00(d,J=8.3Hz,1H),1.31(d,J=6.7Hz,3H).
实施例10:
Figure PCTCN2020094532-appb-000130
氮气保护下,将中间体a4(0.1mmol,48mg)和中间体c3(0.15mmol,69mg)溶于3mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.3mmol,41mg)和Pd(dppf)Cl 2(0.01mmol,8mg),微波下100℃加热反应1小时。停止反应,过滤,减压蒸出有机溶剂,加水10mL,乙酸乙酯萃取,柱层析分离,得到化合物D1-1(77mg,收率:98%)。LC-MS:[M+H] +:781。
将化合物D1-1(35mg)溶于2mL二氯甲烷中,加入0.5mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。再将此混合物溶于水(0.5mL)和甲醇(3mL)的混合溶液中,加入氢氧化钠(0.45mmol,18mg),回流反应至反应完全。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物D1(6mg,两步 收率:26%),LC-MS:[M+H] +:527。
参照实施例10路线合成的化合物如下:
Figure PCTCN2020094532-appb-000131
Figure PCTCN2020094532-appb-000132
实施例11:D4
Figure PCTCN2020094532-appb-000133
将化合物D1-1(0.098mmol,77mg)溶于3mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,得化合物D4-1(66mg,收率定量)。LC-MS:[M+H] +:682。
将上步所得化合物D4-1溶于5mL甲醇中,加入30%的甲醛水溶液(0.2mmol,20μL)和NaBH 3CN(0.3mmol,20mg),滴加1滴醋酸后,室温反应3小时。减压蒸除溶剂,TLC色谱分离,得到化合物D4-2(40mg,收率:97%)。LC-MS:[M+H] +:695。
将化合物D4-2(40mg)溶于溶于四氢呋喃(1.5mL)和甲醇(2mL)的混合溶液中,加入碳酸铯(0.1mmol,33mg),回流反应至反应完全。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物D4(16mg,收率:93%),LC-MS:[M+H] +:541。
1H NMR(400MHz,DMSO-d 6)δ11.76(s,1H),8.32(d,J=5.0Hz,1H),7.97(d,J=5.0Hz,1H),7.56(t,J=2.9Hz,1H),7.20(dd,J=3.4,2.0Hz,1H),6.88(s,1H),4.55(s,1H),4.31-4.18(m,2H),4.01(d,J=10.2Hz,1H),3.79(d,J=11.4Hz,1H),3.68(d,J=10.9Hz,1H),3.54(t,J=12.0Hz,1H),2.92(d,J= 11.3Hz,2H),2.27(s,3H),2.21(d,J=15.9Hz,5H)[Me,CH 2],2.08(d,J=11.9Hz,2H),2.04-1.97(m,1H),1.90(d,J=11.8Hz,2H),1.28(d,J=6.7Hz,3H).
实施例12:D3
Figure PCTCN2020094532-appb-000134
氮气保护下,将中间体a1(1mmol,247mg)和中间体c3(1.7mmol,780mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(3mmol,414mg)和Pd(PPh 3) 4(0.1mmol,115mg),微波下100℃加热反应1小时。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物D3-1。LC-MS:[M+H] +:545。
氮气保护下,将化合物D3-1(0.11mmol,100mg)和中间体c5(0.22mmol,60mg)溶于3mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.33mmol,45mg)和Pd(dppf)Cl 2(0.011mmol,8mg),微波100℃下加热反应30分钟。停止反应,过滤,旋干有机溶剂,加水,乙酸乙酯萃取,柱层析分离,得到化合物D3-2(20mg,收率:28%)。LC-MS:[M+H] +:657。
将化合物D3-2(0.036mmol,24mg)溶于2mL二氯甲烷中,加入0.5mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D3(8mg,收率:40%),LC-MS:[M+H] +:557。
1H NMR(400MHz,DMSO-d 6)δ11.58(s,1H),7.43(s,1H),7.27(t,J=2.9Hz,1H),7.09-7.06(m,1H),6.89(s,1H),4.54(s,1H),4.35-4.21(m,2H),4.00(d,J=9.4Hz,1H),3.91(s,3H),3.79(d,J=11.6Hz,1H),3.68(d,J=10.4Hz,1H),3.58-3.50(m,1H),3.29(s,1H),3.08(d,J=12.6Hz,2H),2.61(t,J=12.4Hz,2H),2.27(s,3H),2.05-1.98(m,2H),1.86(d,J=12.0Hz,2H),1.27(d,J=6.7Hz,3H).
实施例13:D5
Figure PCTCN2020094532-appb-000135
氮气保护下,将中间体a1(1mmol,247mg)和中间体c4(1.6mmol,734mg)溶于1,4-二氧六环和水的10mL混合液中(v/v:9/1),加入碳酸钾(3mmol,414mg),Pd(PPh 3) 4(0.1mmol,115mg),微波100℃加热反应30分钟。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,柱层析分离,得到化合物D5-1(150mg,收率:28%),LC-MS:[M+H] +:545。
氮气保护下,将化合物D5-1(0.18mmol,100mg)和中间体c2(0.26mmol,109mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.54mmol,74mg),Pd(dppf)Cl 2(0.018mmol,13mg),微波100℃加热反应30分钟。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D5-2(110mg,收率:76%),LC-MS:[M+H] +:805。
将化合物D5-2(0.137mmol,110mg)溶于4mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。再次将混合物溶于5mL甲醇和5mL的四氢呋喃的混合溶液中,加入碳酸铯(1.37mmol,446mg),70℃反应1小时。减压蒸除溶剂,制备色谱分离,得到目标化合物D5(24mg,收率:32%),LC-MS:[M+H] +:551。
1H NMR(400MHz,DMSO-d 6)δ12.11(s,1H),8.21(d,J=8.0Hz,1H),7.70(d,J=8.0Hz,1H),7.58(d,J=2.8Hz,1H),7.47(d,J=3.0Hz,1H),6.75(s,1H),4.56-4.38(m,2H),4.22(d,J=13.3Hz,1H),4.06-3.98(m,1H),3.80(d,J=11.5Hz,1H),3.72-3.65(m,1H),3.58-3.50(m,1H),3.29(s,1H),3.06(d,J=12.4Hz,2H),2.68-2.60(m,2H),1.87(s,4H),1.29(d,J=6.7Hz,3H).
实施例14:D6
Figure PCTCN2020094532-appb-000136
氮气保护下,将中间体d1(0.15mmol,50mg)和中间体c3(0.17mmol,63mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.29mmol,41mg),Pd(dppf)Cl 2(0.014mmol,11mg),微波100℃加热反应30分钟。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D6-1(50mg,收率:52%),LC-MS:[M+H] +:642。
将化合物D6-1(0.078mmol,50mg)溶于4mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D6(14mg,收率:34%),LC-MS:[M+H] +:542。
参照实施例14路线合成的化合物如下:
Figure PCTCN2020094532-appb-000137
实施例15:D8
Figure PCTCN2020094532-appb-000138
将中间体d1(0.58mmol,200mg)和4-二甲氨基吡啶DMAP(0.029mmol,3.5mg)溶于10mL二氯甲烷中,加入三乙胺(2.9mmol,294mg),醋酸酐(261mg,4.4mL)后,室温反应12小时。停止反应,加水30mL,二氯甲烷萃取,无水硫酸钠干燥,得到化合物D8-1(200mg,收率:81%),LC-MS:[M+H] +:429。
氮气保护下,将化合物D8-1(0.16mmol,70mg)和中间体c3(0.32mmol,150mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.32mmol,44mg),Pd(dppf)Cl 2(0.016mmol,12mg),微波100℃加热反应1小时。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D8-2(40mg,收率:37%),LC-MS:[M+H] +:684。
将化合物D8-2(0.058mmol,40mg)溶于4mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D8(11mg,收率:33%),LC-MS:[M+H] +:584。
参照实施例15路线合成的化合物如下:
Figure PCTCN2020094532-appb-000139
Figure PCTCN2020094532-appb-000140
实施例16:D10
Figure PCTCN2020094532-appb-000141
将中间体d2(0.27mmol,100mg)和碳酸铯(0.69mmol,224mg)溶于10mL DMF中,逐滴加入碘甲烷(0.33mmol,47mg),55℃下反应2小时。停止反应,加水30mL,乙酸乙酯萃取,无水硫酸钠干燥,TLC色谱分离,得到化合物D10-1(80mg,收率:82%),LC-MS:[M+H] +:363。
氮气保护下,将化合物D10-1(0.21mmol,80mg)和中间体c3(0.42mmol,195mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.42mmol,59mg),Pd(dppf)Cl 2(0.021mmol,16mg),微波100℃加热反应1小时。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D10-2(40mg,收率:30%),LC-MS:[M+H] +:674。
将化合物D10-2(0.06mmol,40mg)溶于4mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D10(10mg,收率:29%),LC-MS:[M+H] +:574。
参照实施例16路线合成的化合物如下:
Figure PCTCN2020094532-appb-000142
Figure PCTCN2020094532-appb-000143
实施例17:D11
Figure PCTCN2020094532-appb-000144
氮气保护下,将中间体c6(1mmol,265mg)和中间体a3(1mmol,398mg)溶于1,4-二氧六环和水的12mL混合液中(v/v:9/1),加入碳酸钾(3mmol,414mg),Pd(dppf)Cl 2(0.1mmol,73mg),微波100℃加热反应30分钟。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,柱 层析分离,得到化合物D11-1(100mg,收率:20%),LC-MS:[M+H] +:502。
氮气保护下,将化合物D11-1(0.2mmol,100mg)和中间体c3(0.24mmol,110mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.6mmol,83mg),Pd(dppf)Cl 2(0.02mmol,16mg),微波100℃加热反应30分钟。停止反应,过滤,减压蒸除有机溶剂,加水10mL,乙酸乙酯萃取,flash柱层析分离,得到化合物D11-2(30mg,收率:20%),LC-MS:[M+H] +:799。
将化合物D11-2(0.05mmol,40mg)溶于4mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。再次将混合物溶于3mL甲醇和3mL的四氢呋喃的混合溶液中,加入碳酸铯(0.3mmol,98mg),70℃反应2小时。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物D11(16mg,两步收率:59%),LC-MS:[M+H] +:545。
1H NMR(400MHz,DMSO-d 6)δ11.80(s,1H),8.32(d,J=5.1Hz,1H),7.90(s,1H),7.58(t,J=3.0Hz,1H),7.16-7.11(m,1H),4.59(d,J=7.7Hz,1H),4.25(d,J=13.4Hz,2H),4.03-3.96(m,1H),3.76(d,J=2.2Hz,2H),3.67-3.46(m,3H),3.08(d,J=12.5Hz,2H),2.61(t,J=12.2Hz,2H),2.26(s,3H),2.02(t,J=13.0Hz,2H),1.87(d,J=11.6Hz,2H),1.38(d,J=6.8Hz,3H).
实施例18:D15
Figure PCTCN2020094532-appb-000145
将化合物D10(0.66mmol,380mg)溶于10mL甲醇中,加入30%的甲醛水溶液(1.3mmol,140μL)和NaBH 3CN(1.95mmol,122mg),滴加5滴醋酸后,室温反应3小时。减压蒸除溶剂,TLC色谱分离,得到化合物D15(105mg,收率:28%)。LC-MS:[M+H] +:588。
1H NMR(400MHz,DMSO-d 6)δ8.58(d,J=5.1Hz,1H),8.14(dd,J=8.8,5.2Hz,1H),7.06(dd,J=9.7,2.7Hz,1H),6.86(s,1H),6.79(td,J=9.4,2.6Hz,1H),4.45(s,1H),4.23(s,1H),4.10(d,J=13.1Hz,1H),4.01(d,J=10.9Hz,1H),3.79(d,J=11.7Hz,1H),3.69(d,J=11.0Hz,1H),3.55(t,J=11.4Hz,1H),3.37(d,J=11.4Hz,1H),3.03(d,J=4.8Hz,3H),2.92(d,J=11.2Hz,2H),2.28(s,3H),2.22(s,3H),2.20(s,1H),2.16(d,J=11.6Hz,1H),2.11-2.03(m,2H),1.90(d,J=11.7Hz,2H),1.30(d,J=6.7Hz,3H).
实施例19:D16
Figure PCTCN2020094532-appb-000146
将中间体d2(0.28mmol,100mg)和碳酸铯(0.83mmol,270mg)溶于12mL DMF中,逐滴加入碘甲烷(0.28mmol,40mg),60℃下反应2小时。停止反应,加水30mL,二氯甲烷萃取,无水硫酸钠干燥,TLC色谱分离,得到化合物D16-1(50mg,收率:48%),LC-MS:[M+H] +:377。
氮气保护下,将化合物D16-1(0.14mmol,50mg)和中间体c4(0.22mmol,102mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.22mmol,32mg),Pd(dppf)Cl 2(0.011mmol,8mg),微波100℃加热反应1小时。停止反应,过滤,减压蒸除有机溶剂,加水15mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D16-2(80mg,收率:85%),LC-MS:[M+H] +:674。
将化合物D16-2(80mg)溶于14mL二氯甲烷中,滴加2mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D16(20mg,收率:30%),LC-MS:[M+H] +:574。
1H NMR(400MHz,DMSO-d 6)δ8.65(d,J=5.1Hz,1H),8.16(dd,J=8.8,5.3Hz,1H),7.06(dd,J=9.6,2.7Hz,1H),6.79(td,J=9.3,2.7Hz,1H),6.73(s,1H),4.53-4.40(m,2H),4.06(dd,J=25.9,11.4Hz,2H),3.80(d,J=11.6Hz,1H),3.73-3.67(m,1H),3.58-3.52(m,1H),3.36(s,1H),3.10(d,J=12.5Hz,2H),3.03(d,J=4.8Hz,3H),2.69(t,J=10.6Hz,2H),2.47(s,3H),2.00(q,J=6.4,5.2Hz,1H),1.93-1.88(m,3H),1.31(d,J=6.7Hz,3H).
实施例20:D17
Figure PCTCN2020094532-appb-000147
氮气保护下,将中间体d4(0.14mmol,45mg)和中间体c3(0.25mmol,115mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.3mmol,41mg)和Pd(dppf)Cl 2(0.014mmol,11mg),微波下100℃加热反应1小时。停止反应,过滤,减压蒸出有机溶剂,加水20mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D17-1(50mg,收率:58%)。LC-MS:[M+H] +:626。
将化合物D17-1(50mg)溶于2mL二氯甲烷中,加入0.5mL三氟乙酸,室温反应2小时后, 减压蒸除溶剂。制备色谱分离,得到目标化合物D17(31mg,收率:74%),LC-MS:[M+H] +:526。
参照实施例20路线合成的化合物如下:
Figure PCTCN2020094532-appb-000148
实施例21:D19
Figure PCTCN2020094532-appb-000149
氮气保护下,将中间体d5(0.31mmol,150mg)和中间体c3(0.60mmol,275mg)溶于15mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.9mmol,124mg)和Pd(dppf)Cl 2(0.03mmol,25mg),微波下100℃加热反应1小时。停止反应,过滤,减压蒸出有机溶剂,加水10mL,乙酸乙酯萃取,柱层析分离,得到化合物D19-1(66mg,收率:28%)。LC-MS:[M+H] +:781。
将化合物D19-1(66mg)溶于5mL二氯甲烷中,加入1mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。再将此混合物溶于四氢呋喃(3mL)和甲醇(3mL)的混合溶液中,加入碳酸铯(0.2mmol,65mg),回流反应至反应完全。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物D19(20mg,两 步收率:46%),LC-MS:[M+H] +:527。
参照实施例21路线合成的化合物如下:
Figure PCTCN2020094532-appb-000150
实施例22:D23
Figure PCTCN2020094532-appb-000151
将中间体d2(0.28mmol,100mg)和碳酸铯(0.69mmol,225mg)溶于10mL DMF中,逐滴加入碘乙烷(0.33mmol,51mg),60℃下反应1.5小时。停止反应,加水30mL,二氯甲烷萃取,无水硫酸钠干燥,TLC色谱分离,得到化合物D23-1(95mg,收率:87%),LC-MS:[M+H] +:391。
氮气保护下,将化合物D23-1(0.24mmol,95mg)和中间体c3(0.48mmol,223mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(0.48mmol,66mg),Pd(dppf)Cl 2(0.024mmol,18mg),微波100℃加热反应1.5小时。停止反应,过滤,减压蒸除有机溶剂,加水15mL,乙酸乙 酯萃取,TLC色谱分离,得到化合物D23-2(100mg,收率:61%),LC-MS:[M+H] +:688。
将化合物D23-2(100mg)溶于15mL二氯甲烷中,滴加3mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D23(36mg,收率:44%),LC-MS:[M+H] +:588。
1H NMR(400MHz,DMSO-d 6)δ8.65(t,J=5.6Hz,1H),8.15(dd,J=8.9,5.3Hz,1H),7.05(dd,J=9.7,2.7Hz,1H),6.87(s,1H),6.78(td,J=9.4,2.7Hz,1H),4.44(s,1H),4.32(s,1H),4.09(d,J=12.9Hz,1H),4.05-3.98(m,1H),3.79(d,J=11.7Hz,1H),3.69(d,J=11.4Hz,1H),3.56(d,J=12.2Hz,1H),3.50(dd,J=13.3,6.7Hz,2H),3.40-3.35(m,1H),3.07(d,J=12.7Hz,2H),2.60(t,J=12.3Hz,2H),2.27(s,3H),2.01(t,J=9.0Hz,2H),1.85(d,J=11.6Hz,2H),1.30(d,J=6.7Hz,3H),1.19(d,J=7.1Hz,3H).
实施例23:A29
Figure PCTCN2020094532-appb-000152
氮气保护下,将中间体a4(0.41mmol,200mg)和中间体b10(0.49mmol,190mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钾(1.24mmol,170mg)和Pd(dppf)Cl 2(0.08mmol,60mg),升温至90℃搅拌3小时。停止反应,过滤,加水30mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到化合物A29-1,(240mg,收率:69%),LC-MS:[M+H] +:700。
将化合物A29-1(0.286mmol,200mg)溶于8mL四氢呋喃和甲醇的混合溶液中(v/v,1/1),加入碳酸铯(0.86mmol,280mg),回流反应至反应完全。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物A29(收率:42%),LC-MS:[M+H] +:546。
1H NMR(400MHz,DMSO-d 6)δ11.77(s,1H),8.32(d,J=5.0Hz,1H),8.22(q,J=8.4Hz,2H),8.12(s,1H),7.98(d,J=5.0Hz,1H),7.56(t,J=3.0Hz,1H),7.18(dd,J=3.4,1.9Hz,1H),7.03(s,1H),4.63(s,1H),4.26(d,J=13.1Hz,1H),4.08-3.97(m,1H),3.81(d,J=11.6Hz,1H),3.74-3.62(m,2H),3.61-3.48(m,1H),3.35(dd,J=12.9,3.8Hz,1H),1.31(d,J=6.7Hz,3H),1.24(d,J=2.1Hz,6H).
参照实施例23路线合成如下化合物:
Figure PCTCN2020094532-appb-000153
Figure PCTCN2020094532-appb-000154
Figure PCTCN2020094532-appb-000155
实施例24:A31
Figure PCTCN2020094532-appb-000156
在反应瓶中加入中间体a9(0.78mmol,425mg),碳酸铯(2.35mmol,765mg)和3-((甲基磺酰基)氧基)吡咯烷-1-甲酸叔丁酯(0.94mmol,249mg),10mL DMF溶解。升温至80℃反应2小时。停止反应,向体系加水40mL,乙酸乙酯萃取,flash柱层析分离,得到化合物A31-1(380mg,收率:69%),LC-MS:[M+H] +:713。
将中间体A31-1(380mg)溶于6mL二氯甲烷中,滴加2mL三氟乙酸,室温反应2小时。停止反应,减压蒸除溶剂。向体系加入饱和碳酸氢钠水溶液20mL,二氯甲烷萃取,得到粗品中间体A31-2。直接进行下一步反应。LC-MS:[M+H] +:613。
冰浴下,将粗品中间体A31-2和10滴40%乙醛水溶液溶于5mL甲醇中,滴加4滴冰醋酸。搅拌5分钟后,缓慢加入NaCNBH 3(0.82mmol,51mg),继续搅拌2小时后停止反应。向反应液加入4M氯化铵水溶液40mL,乙酸乙酯萃取,flash柱层析分离,得到中间体A31-3,两步总收率:73%。LC-MS:[M+H] +:641。
将中间体A31-3(0.125mmol,80mg)溶解于8mL甲醇和THF的混合溶液中(v/v,1/1),加入碳酸铯(0.375mmol,122mg),60℃下反应2小时。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物A31(45mg,收率:74%),LC-MS:[M+H] +:487。
1H NMR(400MHz,DMSO-d 6)δ11.75(s,1H),8.33(d,J=5.0Hz,1H),8.01(d,J=5.0Hz,1H),7.57(t,J=3.0Hz,1H),7.24(dd,J=3.4,1.9Hz,1H),6.65(s,1H),4.92(p,J=7.2Hz,1H),4.57(s,1H),4.22(d,J=13.2Hz,1H),4.02(d,J=9.1Hz,1H),3.81(d,J=11.5Hz,1H),3.69(d,J=10.7Hz,1H),3.59-3.50(m,1H),3.29(s,1H),3.09(t,J=8.4Hz,1H),2.78(q,J=7.0Hz,1H),2.65-2.58(m,2H),2.54(s,3H),2.46(dd,J=7.1,3.1Hz,2H),2.39(s,3H),2.23(s,2H),1.29(d,J=6.7Hz,3H),1.05(t,J=7.2Hz,3H).
实施例25:A32
Figure PCTCN2020094532-appb-000157
在反应瓶中加入中间体a9(0.27mmol,140mg),碳酸铯(0.81mmol,263mg)和3-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.32mmol,90mg),8mL DMF溶解。升温至100℃反应4小时。停止反应,向体系加水40mL,乙酸乙酯萃取,flash柱层析分离,得到化合物A32-1(收率:55%),LC-MS:[M+H] +:727。
将中间体A32-1(75mg)溶于4mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时。停止 反应,减压蒸除溶剂。向体系加入饱和碳酸氢钠水溶液20mL,二氯甲烷萃取,得到粗品中间体A32-2。直接进行下一步反应。LC-MS:[M+H] +:627。
冰浴下,将粗品中间体A32-2和1mL 40%甲醛水溶液溶于3mL甲醇中,滴加2滴冰醋酸。搅拌5分钟后,缓慢加入NaCNBH 3(0.5mmol,33mg),继续搅拌2小时后停止反应。向体系加入4M氯化铵水溶液20mL,乙酸乙酯萃取,TLC色谱分离,得到中间体A32-3,两步总收率:54%。LC-MS:[M+H] +:641。
将中间体A32-3(0.05mmol,33mg)溶解于4mL甲醇和THF的混合溶液中(v/v,1/1),加入碳酸铯(0.2mmol,65mg),60℃下反应2小时。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物A32(22mg,收率:91%),LC-MS:[M+H] +:487。
1H NMR(400MHz,DMSO-d 6)δ11.75(s,1H),8.33(d,J=5.1Hz,1H),8.01(d,J=5.1Hz,1H),7.56(t,J=3.0Hz,1H),7.24(t,J=2.7Hz,1H),6.66(s,1H),4.56(s,1H),4.22(d,J=14.7Hz,2H),4.02(d,J=10.8Hz,1H),3.81(d,J=11.4Hz,1H),3.69(d,J=9.7Hz,1H),3.53(d,J=14.3Hz,1H),2.89(s,1H),2.81(s,2H),2.54(s,3H),2.37(s,3H),2.23(s,3H),1.99(s,1H),1.85(d,J=30.5Hz,3H),1.77(d,J=12.5Hz,1H),1.67(s,1H),1.30(s,3H).
实施例26:A37
Figure PCTCN2020094532-appb-000158
在反应瓶中加入中间体a9(0.38mmol,210mg),碳酸铯(1.16mmol,378mg)和4-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.46mmol,129mg),8mL DMF溶解后,升温至90℃反应5小时。停止反应,向体系加水30mL,乙酸乙酯萃取,flash柱层析分离,得到中间体A37-1(收率:41%),LC-MS:[M+H] +:727。
将中间体A37-1(115mg)溶于3mL二氯甲烷中,滴加1mL三氟乙酸,室温反应2小时。停止反应,减压蒸除有机溶剂。向体系加入饱和碳酸氢钠水溶液20mL,二氯甲烷萃取,得到粗品中间体A37-2。直接进行下一步反应。LC-MS:[M+H] +:627。
冰浴下,将粗品中间体A37-2和1mL 40%甲醛水溶液溶于2mL甲醇中,滴加2滴冰醋酸。搅拌5分钟后,缓慢加入NaCNBH 3(072mmol,45mg),继续搅拌2小时后停止反应。向体系加入4M氯化铵水溶液20mL,乙酸乙酯萃取,TLC色谱分离,得到中间体A37-3,两步总收率:51%。LC-MS:[M+H] +:641。
将中间体A37-3(0.08mmol,50mg)溶解于4mL甲醇和THF的混合溶液中(v/v,1/1),加入碳酸铯(0.23mmol,76mg),60℃下反应2小时。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物A37(9mg,收率:22%),LC-MS:[M+H] +:487。
实施例27:A38
Figure PCTCN2020094532-appb-000159
在反应瓶中加入中间体a9(0.12mmol,66mg),碳酸铯(0.24mmol,79mg)和原料A38-1(0.24mmol,50mg),5mL乙腈溶解后,升温至80℃反应2小时。停止反应,向体系加水40mL,乙酸乙酯萃取,flash柱层析分离,得到中间体A38-2(80mg,收率:99%),LC-MS:[M+H] +:655。
将中间体A38-2(0.12mmol,80mg)溶于4mL甲醇和THF的混合溶液中(v/v,1/1),加入碳酸铯(0.24mmol,79mg),80℃下反应2小时。过滤,减压蒸除溶剂,制备色谱分离,得到目标化合物A38(15mg,收率:25%),LC-MS:[M+H] +:501。
1H NMR(400MHz,DMSO-d 6)δ11.75(s,1H),8.33(d,J=5.0Hz,1H),8.01(d,J=4.9Hz,1H),7.56(t,J=2.9Hz,1H),7.23(dd,J=3.5,1.9Hz,1H),6.67(s,1H),4.59(s,1H),4.22(d,J=12.8Hz,1H),4.06-3.92(m,3H),3.81(d,J=11.5Hz,1H),3.69(d,J=11.3Hz,1H),3.55(t,J=11.4Hz,1H),3.28(s,1H),2.96(dt,J=50.5,23.5Hz,4H),2.39(s,3H),2.16(s,1H),2.13(s,3H),2.08(d,J=11.3Hz,4H),1.46(s,2H),1.29(d,J=6.7Hz,3H).
实施例28:A41
Figure PCTCN2020094532-appb-000160
氮气保护下,将中间体a4(0.18mmol,85mg)和中间体b18(0.18mmol,50mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.53mmol,73mg)和Pd(dpPf)Cl 2(0.02mmol,15mg),100℃微波加热反应30分钟。停止反应,过滤,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到化合物A41-1(77mg,收率:73%),LC-MS:[M+H] +:604。
在反应瓶中将化合物A41-1(0.127mmol,77mg)溶于6mL甲醇和水的混合液中(v/v,5/1),缓慢加入单过硫酸氢钾Oxone(0.38mmol,233mg),室温反应3小时后停止反应。加水30mL,二氯甲烷萃取,得到粗品化合物A41-2(70mg),LC-MS:[M+H] +:636。
将粗品化合物A41-2(0.03mmol,15mg)溶于3mL甲醇和四氢呋喃的混合液中(v/v,1/1),加入碳酸铯(0.07mmol,23mg),升温至60℃反应3小时,停止反应。加水20mL,二氯甲烷萃取,制备色谱分离,得到目标化合物A41(3mg,收率:27%),LC-MS:[M+H] +:482。
1H NMR(400MHz,DMSO-d 6)δ11.76(s,1H),8.34(d,J=5.0Hz,1H),8.01(d,J=5.0Hz,1H),7.58(t,J=3.0Hz,1H),7.23(dd,J=3.4,1.9Hz,1H),6.72(s,1H),5.73(s,2H),4.61(s,1H),4.25(d,J=13.3Hz,1H),4.06-3.98(m,1H),3.81(d,J=11.4Hz,1H),3.70(d,J=9.9Hz,1H),3.58-3.50(m,1H),3.28(d,J=9.7Hz,1H),3.09(s,3H),2.59(s,3H),2.43(s,3H),1.30(d,J=6.6Hz,3H).
实施例29:D24
Figure PCTCN2020094532-appb-000161
氮气保护下,将中间体d7(0.92mmol,330mg)和中间体c3(1.8mmol,845mg)溶于15mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(1.83mmol,254mg)和Pd(dppf)Cl 2(0.09mmol,67mg),100℃下加热反应2小时。停止反应,过滤,加水40mL,乙酸乙酯萃取,flash柱层析分离, 得到化合物D24-1(400mg,收率:67%),LC-MS:[M+H] +:656。
将化合物D24-1(0.6mmol,400mg)溶于6mL二氯甲烷中,加入2mL三氟乙酸,室温反应3小时,停止反应。缓慢加入饱和碳酸氢钠水溶液30mL,二氯甲烷萃取,制备色谱分离,得到目标化合物D24(收率:55%),LC-MS:[M+H] +:556。
参照实施例29路线合成如下化合物:
Figure PCTCN2020094532-appb-000162
实施例30:D26
Figure PCTCN2020094532-appb-000163
在微波反应瓶中加入中间体a1(2.02mmol,500mg)和原料2-硝基-3-氨基-吡啶(2.4mmol,337mg),8mL DMA溶解。加入叔丁醇钠(4mmol,384mg)后微波100℃下反应1.5小时。停止反应,加水30mL,乙酸乙酯萃取,无水硫酸钠干燥,得到粗品中间体D26-1,LC-MS:[M+H] +:351。
在反应瓶中加入上步中间体D26-1(粗品)、还原铁粉(12mmol,672mg)和氯化铵(12mmol,642mg),加入22mL乙醇和4mL水溶解。80℃下反应2小时。停止反应,抽滤,加水60mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D26-2(两步收率:91%),LC-MS:[M+H] +:321。
在反应瓶中加入中间体D26-2(0.65mmol,210mg)、三乙胺(3.3mmol,331mg)和羰基二咪唑(3.3mmol,530mg),10mL四氢呋喃溶解。升温至50℃下反应10小时。停止反应,加水40mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D26-3(200mg,收率:89%),LC-MS:[M+H] +:347。
中间体D26-3(0.35mmol,120mg)溶于5mL三氯氧磷中,升温至100℃反应10小时。停止反应,将反应液置于冰浴下,缓慢加入冰水20mL,并用饱和碳酸钠水溶液调节pH~10左右。乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D26-4(70mg,收率:55%),LC-MS:[M+H] +:366。
将中间体D26-4(0.16mmol,60mg)溶于10mL四氢呋喃中,缓慢加入甲基胺溶液(1mL,2.5M),升温至70℃反应4小时。停止反应,flash柱层析分离,得到中间体D26-5(30mg,收率:53%),LC-MS:[M+H] +:360。
氮气保护下,将中间体D26-5(0.083mmol,30mg)和中间体c3(0.15mmol,69mg)溶于3mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.17mmol,23mg)和Pd(dppf)Cl 2(0.008mmol,7mg),100℃加热反应2小时。停止反应,过滤,加水10mL,乙酸乙酯萃取,TLC色谱分离,得到 化合物D26-6(50mg,收率:67%),LC-MS:[M+H] +:657。
将化合物D26-6(0.076mmol,50mg)溶于3mL二氯甲烷中,加入1.0mL三氟乙酸,室温反应3小时,停止反应。缓慢加入饱和碳酸氢钠水溶液20mL,二氯甲烷萃取,制备色谱分离,得到目标化合物D26(14mg,收率:34%),LC-MS:[M+H] +:557。
1H NMR(400MHz,DMSO-d 6)δ8.88(d,J=26.7Hz,1H),8.40(d,J=7.9Hz,1H),8.13(dd,J=5.3,1.6Hz,1H),7.03(dd,J=7.9,5.1Hz,1H),6.89(s,1H),4.74(s,1H),4.45(s,1H),4.07(dd,J=33.0,11.6Hz,2H),3.80(d,J=11.6Hz,1H),3.72-3.67(m,1H),3.59-3.53(m,1H),3.51(s,1H),3.21(d,J=9.7Hz,2H),3.10(d,J=4.8Hz,3H),2.36(s,2H),2.30(s,3H),2.17-1.98(m,4H),1.31(d,J=6.8Hz,3H).
实施例31:D27
Figure PCTCN2020094532-appb-000164
在微波反应瓶中加入中间体c6(1mmol,265mg)和原料2-硝基-4-氟-苯胺d2-1(1mmol,171mg),8mL DMA溶解。加入叔丁醇钠(2mmol,192mg)后微波100℃下反应1小时。停止反应,加水30mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D27-1(150mg,收率:39%),LC-MS:[M+H] +:386。
在反应瓶中加入中间体D27-1(0.39mmol,150mg)、还原铁粉(1.95mmol,109mg)和氯化铵(3.9mmol,206mg),加入10mL乙醇和2mL水溶解。100℃下反应2小时。停止反应,抽滤,加水30mL,乙酸乙酯萃取,无水硫酸钠干燥,得到粗品中间体D27-2(70mg),LC-MS:[M+H] +:356。
在反应瓶中加入中间体D27-2(0.2mmol,70mg)、溴腈(0.4mmol,42mg),4mL甲醇和1mL水溶解。升温至80℃下反应2小时。停止反应,加水40mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D27-3(60mg,收率:79%),LC-MS:[M+H] +:381。
将中间体D27-3(0.47mmol,180mg)和碳酸铯(1.42mmol,462mg)溶于6mL DMF中,逐滴加入碘甲烷(0.47mmol,0.029mL),50℃下反应3小时。停止反应,加水20mL,二氯甲烷萃取, 无水硫酸钠干燥,TLC色谱分离,得到化合物D27-4(150mg,收率:81%),LC-MS:[M+H] +:395。
氮气保护下,将化合物D27-4(0.38mmol,150mg)和中间体c3(0.76mmol,348mg)溶于1,4-二氧六环和水的5mL混合液中(v/v:9/1),加入碳酸钾(1.14mmol,157mg),Pd(dppf)Cl 2(0.04mmol,28mg),微波100℃加热反应1小时。停止反应,过滤,减压蒸除溶剂,加水15mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D27-5(112mg,收率:43%),LC-MS:[M+H] +:692。
将化合物D27-5(112mg)溶于5mL二氯甲烷中,滴加2mL三氟乙酸,室温反应2小时后,减压蒸除溶剂,制备色谱分离,得到目标化合物D27(30mg,收率:32%),LC-MS:[M+H] +:592。
1H NMR(400MHz,DMSO-d 6)δ8.19(d,J=5.2Hz,1H),8.05(dd,J=8.8,5.2Hz,1H),7.07(dd,J=9.6,2.7Hz,1H),6.80(td,J=9.3,2.7Hz,1H),4.49(d,J=7.3Hz,1H),4.30(d,J=11.5Hz,1H),4.10(d,J=11.9Hz,1H),4.03-3.97(m,1H),3.76(d,J=2.2Hz,2H),3.67-3.55(m,2H),3.09(s,1H),3.06(s,1H),3.04(d,J=5.0Hz,3H),2.61(t,J=12.1Hz,2H),2.25(s,3H),2.04(dd,J=13.0,8.9Hz,2H),1.87(d,J=11.8Hz,2H),1.40(d,J=6.8Hz,3H).
实施例32:D28
Figure PCTCN2020094532-appb-000165
氮气保护下,将中间体d9(4.0mmol,1.4g)和中间体c3(4.0mmol,3.7g)溶于50mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(12mmol,1.67g)和Pd(dppf)Cl 2(0.4mmol,296mg),升温至100℃加热反应2小时。停止反应,过滤,减压蒸除溶剂,加水80mL,乙酸乙酯萃取,柱层析分离,得到化合物D28-1(2.4g,收率:93%)。LC-MS:[M+H] +:644。
将化合物D28-1(2.4g)溶于30mL二氯甲烷中,加入10mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入80mL饱和碳酸钠水溶液,二氯甲烷萃取,柱层析分离。得到目标化合物D28(收率:90%),LC-MS:[M+H] +:544。
参照实施例42路线合成如下化合物:
Figure PCTCN2020094532-appb-000166
Figure PCTCN2020094532-appb-000167
Figure PCTCN2020094532-appb-000168
实施例33:D29
Figure PCTCN2020094532-appb-000169
冰浴下,将中间体d2(0.55mmol,200mg)和溴化铜(0.82mmol,184mg)溶于10mL乙腈中。缓慢加入亚硝酸叔丁酯(0.83mmol,85mg),冰浴下连续搅拌5小时后停止反应。过滤,flash柱层析分离,得到中间体D29-1(30mg,收率:13%),LC-MS:[M+H] +:427。
氮气保护下,将中间体D29-1(0.13mmol,56mg)和环丙基氨(1.3mmol,74mg)溶于5mL乙醇中,升温至80℃反应10小时。停止反应,flash柱层析分离,得到中间体D29-2(35mg,收率:67%)。LC-MS:[M+H] +:403。
氮气保护下,将中间体D29-2(0.087mmol,35mg)和中间体c3(0.10mmol,48mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.26mmol,36mg)和Pd(dppf)Cl 2(0.01mmol,13mg),升温至100℃反应2小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,柱层析分离,得到化合物D29-3(40mg,收率:66%)。LC-MS:[M+H] +:682。
将化合物D29-3(40mg)溶于6mL二氯甲烷中,加入1.5mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D29(23mg,收率:68%),LC-MS:[M+H] +:582。
1H NMR(400MHz,DMSO-d 6)δ8.77(d,J=2.8Hz,1H),8.15(dd,J=8.8,5.3Hz,1H),7.12(dd,J= 9.7,2.6Hz,1H),6.86(s,1H),6.84-6.76(m,1H),4.43(s,1H),4.34(d,J=12.2Hz,1H),4.04(dd,J=24.4,11.8Hz,2H),3.79(d,J=11.6Hz,1H),3.69(d,J=11.9Hz,1H),3.55(t,J=11.8Hz,1H),3.37(d,J=11.8Hz,1H),3.08(d,J=12.5Hz,2H),2.86(s,1H),2.62(t,J=12.4Hz,2H),2.26(s,3H),2.06-1.97(m,2H),1.85(d,J=12.2Hz,2H),1.30(d,J=6.6Hz,3H),0.79(d,J=5.7Hz,2H),0.54-0.46(m,2H).
实施例34:D30a+D30b
Figure PCTCN2020094532-appb-000170
将中间体a1(0.81mmol,200mg)和原料5-氟苯并咪唑(0.88mmol,121mg)溶于6mL 1,4-二氧六环中,加入碳酸铯(1.61mmol,525mg)。升温至100℃反应4小时。停止反应,减压蒸除溶剂,flash柱层析分离。得到中间体D30a-1(70mg,收率:25%)和D30b-1(100mg,收率:36%)。LC-MS:[M+H] +:348。
氮气保护下,将中间体D30a-1(0.2mmol,70mg)和中间体c3(0.24mmol,111mg)溶于5mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.6mmol,84mg)和Pd(dppf)Cl 2(0.04mmol,29mg),升温至100℃加热反应2小时。停止反应,过滤,加水30mL,乙酸乙酯萃取,flash柱层析分离,得到化合物D30a-2(30mg,收率:24%)。LC-MS:[M+H] +:645。
将化合物D30a-2(30mg)溶于6mL二氯甲烷中,加入1.5mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D30a(20mg,收率:79%),LC-MS:[M+H] +:545。
1H NMR(400MHz,DMSO-d 6)δ9.15(s,1H),8.41(dd,J=8.9,5.1Hz,1H),7.59(dd,J=9.2,2.5Hz,1H),7.25(td,J=9.4,2.5Hz,1H),6.87(s,1H),4.54(s,1H),4.38-4.24(m,2H),4.03-3.95(m,1H),3.78(d,J=11.6Hz,1H),3.68(dd,J=11.7,3.1Hz,1H),3.53(dd,J=12.7,9.7Hz,1H),3.27(d,J=5.0Hz,1H),3.08(d,J=12.7Hz,2H),2.62(t,J=12.2Hz,2H),2.29(s,3H),2.02(dd,J=14.0,9.0Hz,3H),1.86(d,J=11.3Hz,2H),1.29(d,J=6.8Hz,3H).
参照化合物D30a的合成路线,在第二步将中间体D30a-1替换为D30b-1,合成目标化合物D30b。
1H NMR(400MHz,DMSO-d 6)δ9.11(s,1H),8.16(dd,J=9.9,2.7Hz,1H),7.78(dd,J=8.8,5.0Hz,1H),7.20(td,J=9.1,2.6Hz,1H),6.89(s,1H),4.54(s,1H),4.32(d,J=13.0Hz,2H),4.04-3.96(m,1H),3.78(d,J=11.6Hz,1H),3.71-3.64(m,1H),3.58-3.49(m,1H),3.33-3.35(m,1H),3.08(d,J=12.7Hz,2H),2.62(t,J=12.4Hz,2H),2.29(s,3H),2.07-1.97(m,2H),1.87(d,J=12.1Hz,2H),1.29(d,J=6.7Hz,3H).
参照实施例34路线合成如下化合物:
Figure PCTCN2020094532-appb-000171
实施例35:D31
Figure PCTCN2020094532-appb-000172
氮气保护下,将中间体d2(0.55mmol,200mg)和二甲基硫迷(0.72mmol,68mg)溶于8mL二氯乙烷中。升温至60℃后,将亚硝酸叔丁酯(0.72mmol,74mg)溶于1mL二氯乙烷中,缓慢加入体系并连续搅拌5小时。停止反应,flash柱层析分离,得到中间体D31-1(210mg,收率:78%),LC-MS:[M+H] +:394。
氮气保护下,将中间体D31-1(0.25mmol,100mg)和中间体c3(0.31mmol,140mg)溶于8mL1,4-二氧六环和水的混合液中(v/v:7/1),加入碳酸钾(0.76mmol,105mg)和Pd(dppf)Cl 2(0.05mmol,37mg),升温至100℃反应2小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到化合物D31-2(30mg,收率:18%)。LC-MS:[M+H] +:691。
将化合物D31-2(30mg)溶于3mL二氯甲烷中,加入1.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合 物D31(11mg,收率:47%),LC-MS:[M+H] +:591。HPLC纯度:94.73%。
实施例36:D32
Figure PCTCN2020094532-appb-000173
将中间体d2-3(2.96mmol,1.0g)溶于5g乙醇酸中。升温至140℃并搅拌4小时。停止反应,向体系缓慢加入30mL饱和碳酸钠水溶液,二氯甲烷萃取,flash柱层析分离,得到中间体D32-1(80mg,收率:7%),LC-MS:[M+H] +:378。
氮气保护下,将中间体D32-1(0.21mmol,80mg)和中间体c3(0.25mmol,232mg)溶于8mL 1,4-二氧六环和水的混合液中(v/v:7/1),加入碳酸钾(0.63mmol,87mg)和Pd(dppf)Cl 2(0.04mmol,31mg),升温至90℃反应3小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到化合物D32-2(35mg,收率:25%)。LC-MS:[M+H] +:675。
将化合物D32-2(25mg)溶于3mL二氯甲烷中,加入1.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D32(17mg,收率:57%),LC-MS:[M+H] +:575。HPLC纯度:99.71%。
实施例37:D33+D34/
Figure PCTCN2020094532-appb-000174
氮气保护下,将中间体D10-1(0.53mmol,200mg)和中间体c9a/c9b混合物(1.06mmol,432mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(1.59mmol,219mg)和Pd(dppf)Cl 2(0.05mmol,39mg),微波升温至105℃反应1小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到混合物D33a-1和D33b-1(200mg,收率:61%)。LC-MS:[M+H] +:621。
将混合物D33a-1和D33b-1(0.32mmol,200mg)溶于5mL二氯甲烷中,加入2.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。将该混合物溶于3mL甲醇中,缓慢加入3mL的7M氨水溶液,室温搅拌2小时,停止反应。二氯甲烷萃取,TLC色谱分离。得到中间体D33-2(130mg,收率:83%),LC-MS:[M+H] +:491。
将中间体D33-2(0.26mmol,130mg)、3-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.29mmol,81mg)和碳酸铯(0.80mmol,259mg)溶于5mL DMF中,升温至80℃反应1小时后,再次添加3-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.29mmol,81mg)至反应体系并继续搅拌5小时。停止反应,加入40mL水,二氯甲烷萃取,TLC色谱分离。得到混合物D33-3和D34-1(150mg,收率:86%),LC-MS:[M+H] +:674。
将混和物D33-3和D34-1(0.148mmol,100mg)溶于3mL二氯甲烷中,加入1.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D33(15mg,收率:18%),和目标化合物D34(60mg,收率:71%),LC-MS:[M+H] +:574。
D33: 1H NMR(400MHz,DMSO-d 6)δ9.24(s,1H),9.12(s,1H),8.84(s,1H),8.26(dd,J=8.9,5.1Hz, 1H),7.20(dd,J=9.1,2.6Hz,1H),6.98(dt,J=10.4,5.3Hz,1H),6.78(s,1H),4.76(d,J=10.1Hz,1H),4.42(s,1H),4.14-3.99(m,2H),3.82(d,J=11.6Hz,1H),3.70(dd,J=11.7,3.1Hz,1H),3.60-3.52(m,2H),3.43-3.24(m,3H),3.10(d,J=4.8Hz,3H),2.48(s,3H),2.14(s,2H),2.05-1.93(m,2H),1.85(t,J=13.0Hz,1H),1.32(d,J=6.7Hz,3H).
D34: 1H NMR(400MHz,DMSO-d 6)δ9.02(d,J=15.1Hz,2H),8.83(s,1H),8.22(dd,J=8.9,5.0Hz,1H),7.17(dd,J=9.1,2.6Hz,1H),6.92(d,J=10.8Hz,2H),4.75(s,1H),4.45(s,1H),4.11(d,J=13.0Hz,1H),4.05-4.00(m,1H),3.81(d,J=11.6Hz,1H),3.72-3.67(m,1H),3.55(m,2H),3.45(dd,J=21.6,10.7Hz,2H),3.38-3.29(m,2H),3.09(d,J=4.9Hz,3H),2.31(s,3H),2.13(s,2H),2.00(dd,J=15.0,7.5Hz,2H),1.82(s,1H),1.31(d,J=6.7Hz,3H).
实施例38:D35
Figure PCTCN2020094532-appb-000175
氮气保护下,将中间体D10-1(0.39mmol,150mg)和中间体a6(0.48mmol,106mg)溶于10mL1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.80mmol,110mg)和Pd(dppf)Cl 2(0.04mmol,30mg),微波升温至105℃反应1小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到化合物D35-1(150mg,收率:86%)。LC-MS:[M+H] +:437。
将中间体D35-1(0.34mmol,150mg)、3-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.52mmol,145mg)和碳酸铯(0.68mmol,221mg)溶于10mL乙腈中,升温至80℃反应1小时后,再次添加3-((甲基磺酰基)氧基)哌啶-1-甲酸叔丁酯(0.52mmol,145mg)至反应体系并继续搅拌5小时。停止反应,加入40mL水,二氯甲烷萃取,TLC色谱分离。得到化合物D35-2(30mg,收率:15%),LC-MS:[M+H] +:619。
将化合物物D35-2(0.048mmol,30mg)溶于2mL二氯甲烷中,加入0.5mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入10mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D35(7mg,收率:28%),LC-MS:[M+H] +:519。
1H NMR(400MHz,DMSO-d 6)δ8.84-8.75(m,1H),8.17(dd,J=8.9,5.3Hz,1H),7.06(dd,J=9.7,2.6Hz,1H),6.80(td,J=9.5,2.6Hz,1H),6.59(s,1H),4.45(s,1H),4.17-3.98(m,3H),3.80(d,J=11.6Hz,1H),3.69(d,J=10.9Hz,1H),3.56(d,J=11.2Hz,1H),3.08(d,J=4.8Hz,3H),3.02(d,J=11.8Hz,1H),2.90(d,J=12.5Hz,1H),2.81(t,J=11.2Hz,1H),2.47(s,3H),2.33(s,3H),2.00(dd,J=14.7,7.0 Hz,3H),1.73(d,J=13.3Hz,1H),1.50(d,J=38.4Hz,2H),1.30(d,J=6.6Hz,3H).
实施例39:D36
Figure PCTCN2020094532-appb-000176
氮气保护下,将中间体D10-1(0.27mmol,100mg)和中间体c10(0.35mmol,140mg)溶于10mL1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钾(0.53mmol,73mg)和Pd(dppf)Cl 2(0.03mmol,20mg),微波升温至110℃反应1小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D36-1(120mg,收率:72%)。LC-MS:[M+H] +:619。
将化合物D36-1(0.19mmol,120mg)溶于5mL二氯甲烷中,加入2mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D36(74mg,收率:76%),LC-MS:[M+H] +:519。
1H NMR(400MHz,DMSO-d 6)δ8.79(d,J=5.1Hz,1H),8.17(dd,J=8.8,5.3Hz,1H),7.06(dd,J=9.7,2.6Hz,1H),6.79(td,J=9.4,2.7Hz,1H),6.59(s,1H),4.45(s,1H),4.27(d,J=11.6Hz,1H),4.10-3.99(m,2H),3.80(d,J=11.5Hz,1H),3.69(dd,J=11.8,3.1Hz,1H),3.59-3.50(m,1H),3.36(d,J=4.0Hz,1H),3.13(d,J=12.4Hz,2H),3.08(d,J=4.8Hz,3H),2.72(t,J=12.3Hz,2H),2.48(s,3H),2.33(s,3H),1.97(td,J=13.3,10.5,6.8Hz,3H),1.82(d,J=10.7Hz,2H),1.30(d,J=6.7Hz,3H).
实施例40:D37
Figure PCTCN2020094532-appb-000177
将中间体d1(0.35mmol,120mg)和三乙胺(1.75mmol,177mg)溶于8mL二氯甲烷中,缓慢加入乙基异氰酸酯(1.75mmol,124mg),室温反应10小时,停止反应。减压蒸除溶剂,缓慢加入20mL水,二氯甲烷萃取,TLC色谱分离。得到中间体D37-1(120mg,收率:83%),LC-MS:[M+H] +:416。
氮气保护下,将中间体D37-1(0.22mmol,90mg)和中间体c3(0.66mmol,298mg)溶于10mL1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钾(0.44mmol,61mg)和Pd(dppf)Cl 2(0.02mmol,16mg),微波升温至120℃反应1小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D37-2(100mg,收率:64%)。LC-MS:[M+H] +:713。
将化合物物D37-2(0.14mmol,100mg)溶于5mL二氯甲烷中,加入2mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D37(48mg,收率:56%),LC-MS:[M+H] +:613。
实施例41:D39
Figure PCTCN2020094532-appb-000178
氮气保护下,将中间体D10-1(0.26mmol,100mg)和中间体c11(0.48mmol,226mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钾(0.53mmol,73mg)和Pd(dppf)Cl 2(0.03mmol,19mg),微波升温至110℃反应1小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,TLC色谱分离,得到化合物D39-1(100mg,收率:56%)。LC-MS:[M+H] +:688。
将化合物D39-1(0.14mmol,100mg)溶于5mL二氯甲烷中,加入2mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。缓慢加入20mL饱和碳酸氢钠水溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D39(17mg,收率:21%),LC-MS:[M+H] +:588。
参照实施例41路线合成如下化合物:
Figure PCTCN2020094532-appb-000179
Figure PCTCN2020094532-appb-000180
实施例42:D41+D42
Figure PCTCN2020094532-appb-000181
将中间体D33-2(0.61mmol,300mg)、氯甲基硫醚(1.83mmol,176mg)和碳酸铯(1.83mmol,596mg)溶于10mL DMF中,升温至80℃反应2小时。停止反应,过滤,加入40mL水,二氯甲烷萃取,flash柱层析分离。得到混合物D41-1和D42-1(170mg,总收率:51%),LC-MS:[M+H] +:551。
将混和物D41-1和D42-1(0.148mmol,100mg)溶于12mL甲醇和水的混合溶液中(v/v,5/1),缓慢加入单过硫酸氢钾Oxone(0.93mmol,569mg),室温反应2小时。过滤,加入30mL饱和硫代硫酸钠溶液,二氯甲烷萃取,制备色谱分离。得到目标化合物D41(20mg,收率:23%)和目标化合物D42(40mg,收率:47%),LC-MS:[M+H] +:583。
参照实施例42路线合成如下化合物:
Figure PCTCN2020094532-appb-000182
其中:D43-2的合成如下所示:
Figure PCTCN2020094532-appb-000183
氮气保护下,将中间体d7(0.28mmol,100mg)和中间体c9a/c9b混合物(0.56mmol,227mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钾(0.84mmol,116mg)和Pd(dppf)Cl 2(0.03mmol,20mg),微波升温至105℃反应1小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到混合物D43a-1和D43b-1(300mg,收率:89%)。LC-MS:[M+H] +:603。
将混合物D43a-1和D43b-1(0.5mmol,300mg)溶于8mL二氯甲烷中,加入2.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。将该混合物再次溶于4mL甲醇中,缓慢加入3mL的7M氨水溶液,室温搅拌2小时,停止反应。二氯甲烷萃取,flash柱层析分离。得到化合物D43-2(220mg,收率:93%),LC-MS:[M+H] +:473。
实施例43:D48
Figure PCTCN2020094532-appb-000184
氮气保护下,将中间体e1(1.39mmol,340mg)和中间体c3(1.53mmol,703mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v:9/1),加入碳酸钠(4.17mmol,442mg)和Pd(dppf)Cl 2(0.14mmol,103mg),升温至110℃反应2小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到中间体D48-1(450mg,收率:60%)。LC-MS:[M+H] +:543。
在反应瓶中加入中间体D48-1(0.83mmol,450mg)和原料2-硝基-苯胺(0.99mmol,138mg),8mL DMA溶解。加入叔丁醇钠(1.66mmol,159mg)后,升温至100℃反应2小时。停止反应,加水30mL,乙酸乙酯萃取,饱和氯化钠水溶液洗涤,得到粗品中间体D48-2,LC-MS:[M+H] +:644。
在反应瓶中加入中间体D48-2(500mg,粗品)、还原铁粉(3.89mmol,218mg)和氯化铵(7.8mmol,413mg),30mL乙醇和5mL水溶解,80℃下反应2小时。停止反应,抽滤,加水100mL,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D48-3(260mg,两步收率:52%),LC-MS:[M+H] +:614。
将中间体D48-3(0.42mmol,260mg)溶于8mL吡啶中,缓慢加入甲基异硫氰酸酯(0.5mmol,37mg),90℃下反应0.5小时。降温至室温,加入EDCI(0.63mmol,120mg),继续升温至90℃并连续搅拌4小时,停止反应。向体系内加入50mL水,乙酸乙酯萃取,无水硫酸钠干燥,flash柱层析分离,得到中间体D48-4(130mg,收率:48%),LC-MS:[M+H] +:653。
将中间体D48-4(0.38mmol,250mg)溶于8mL二氯甲烷中,加入2.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。制备色谱分离,得到化合物D48-5(130mg,收率:62%),LC-MS:[M+H] +:553。
化合物D48-5经手性色谱分离,得到目标化合物D48。
1H NMR(400MHz,DMSO-d 6)δ8.34(q,J=5.7,5.1Hz,1H),8.22(d,J=8.4Hz,1H),7.43(s,1H),7.29(dd,J=7.7,0.9Hz,1H),7.13(td,J=7.8,1.3Hz,1H),7.03-6.98(m,1H),4.40-4.32(m,1H),3.99(dd,J=11.6,4.5Hz,1H),3.90(d,J=12.5Hz,1H),3.62(dt,J=11.2,5.6Hz,1H),3.48-3.41(m,1H),3.09(s,1H),3.06(d,J=4.8Hz,3H),2.71-2.66(m,1H),2.63-2.57(m,2H),2.04-1.96(m,2H),1.88(d,J=13.9Hz,2H),1.60(dd,J=9.2,3.9Hz,1H),1.35(dd,J=5.9,4.8Hz,1H).
实施例44:D49
Figure PCTCN2020094532-appb-000185
氮气保护下,将中间体D48-1(0.35mmol,190mg)和中间体d9-1(0.52mmol,137mg)溶于5mL  1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钾(1.05mmol,145mg)和Pd(dppf)Cl 2(0.04mmol,23mg),升温至110℃反应2小时。停止反应,过滤,减压蒸除溶剂,加水20mL,乙酸乙酯萃取,flash柱层析分离,得到中间体D49-1(200mg,收率:89%)。LC-MS:[M+H] +:641。
将中间体D49-1(0.31mmol,200mg)溶于6mL二氯甲烷中,加入2.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。制备色谱分离,得到化合物D49-2(100mg,收率:60%),LC-MS:[M+H] +:541。
化合物D49-2经手性色谱分离,得到目标化合物D49。
1H NMR(400MHz,DMSO-d 6)δ11.40(s,1H),7.94(dd,J=11.6,2.6Hz,1H),7.50(t,J=2.2Hz,1H),7.42(s,1H),7.37(dd,J=9.2,2.3Hz,1H),7.35-7.29(m,1H),4.54-4.43(m,1H),3.99(dd,J=11.4,4.6Hz,1H),3.90(d,J=11.4Hz,1H),3.68-3.57(m,1H),3.49-3.39(m,1H),3.34(s,2H),3.20(d,J=12.6Hz,2H),2.86-2.73(m,2H),2.30(s,3H),2.18-1.93(m,5H),1.60(dd,J=9.8,2.9Hz,1H),1.31-1.24(m,1H).
实施例45:D50
Figure PCTCN2020094532-appb-000186
氮气保护下,将中间体a1(1.0mmol,247mg)和中间体a13(1.0mmol,415mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钠(3mmol,318mg)和Pd(dppf)Cl 2(0.1mmol,73mg),升温至100℃反应10小时。停止反应,过滤,减压蒸除溶剂,加水30mL,乙酸乙酯萃取,flash柱层析分离,得到中间体D50-1(124mg,收率:25%)。LC-MS:[M+H] +:501。
氮气保护下,将中间体D50-1(0.64mmol,320mg)和中间体c9a和c9b的混合物(1.28mmol,520mg)溶于10mL 1,4-二氧六环和水的混合液中(v/v,9/1),加入碳酸钾(1.9mmol,265mg)和Pd(dppf)Cl 2(0.06mmol,47mg),升温至110℃反应5小时。停止反应,过滤,减压蒸除溶剂,加水30mL,乙酸乙酯萃取,flash柱层析分离,得到混合物D50a-2和D50b-2(收率:99%)。LC-MS:[M+H] +:745。
将混合物D50a-2和D50b-2(0.71mmol,530mg)溶于12mL二氯甲烷中,加入4.0mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。将该混合物再次溶于10mL甲醇中,缓慢加入5mL的7M氨水溶液,室温搅拌2小时,停止反应。二氯甲烷萃取,flash柱层析分离。得到中间体D50-3(400mg,收率:91%),LC-MS:[M+H] +:615。
在反应瓶中加入中间体D50-3(0.16mmol,100mg)和原料D50-4(0.33mmol,71mg),5mL DMF溶解。加入碳酸铯(0.49mmol,159mg)后,80℃下加热反应10小时。停止反应,过滤,减压蒸除溶剂,flash柱层析分离,得到中间体D50-5(60mg,收率:47%),LC-MS:[M+H-tBu] +:810。
将中间体D50-5(55mg)溶于3mL二氯甲烷中,加入0.5mL三氟乙酸,室温反应2小时后,减压蒸除溶剂。将该混合物再次溶于3mL甲醇和四氢呋喃的混合溶液中(v/v,1/1),加入碳酸铯(0.3mmol,93mg),升温至80℃搅拌2小时,停止反应。二氯甲烷萃取,制备色谱分离。得到目标化合物D50(11mg,收率:30%;微量异构体),LC-MS:[M+H] +:556。HPLC纯度:93.2%。
参照实施例45路线合成如下化合物:
Figure PCTCN2020094532-appb-000187
实施例46:ATR激酶活性实验:
用ATR激酶磷酸化衍生自p53生物素化的蛋白(Eurofins,货号:14-952)。该实验利用时间分辨荧光测定磷酸化蛋白的量。利用抗-p53-phospho-(丝氨酸15)-K-特异性抗体(Cisbio,货号:61GSTDLA)和d2-标记的抗-GST抗体(Cisbio,货号:61P08KAE)来检测磷酸化蛋白的量。实验开始前,根据需要配制如下工作液:1×反应缓冲液(20mM HEPES PH8.0、1%甘油、0.01%Brij-35),稀释缓冲液(20mM HEPES PH8.0、1%甘油、0.01%Brij-35、5mM DTT和1%BSA),终止液(20mM HEPES PH8.0、1%甘油、0.01%Brij-35、250mM EDTA),检测缓冲液(50mM HEPES PH7.0、150mM NaCl、267mM KF、0.1%胆酸钠、0.01%Tween-20、0.0125%叠氮化钠)。上述所用试剂除特别提到生产商,其它试剂均购于Sigma或英潍捷基。
进行照如下操作:
用1×反应缓冲液制备4×梯度稀释化合物溶液,得到9个不同浓度的化合物,加2.5μL 4×梯度稀释化合物溶液到384孔分析板(784075,Greiner)。用1×反应缓冲液制备4×p53底物工作液(40nM),加2.5μL 4×p53底物工作液到384孔分析板中。用稀释缓冲液制备4×ATR/ATRIP工作液(12.8ng/μL),加2.5μL 4×ATR/ATRIP工作液至384孔分析板。用去离子水制备4×ATP工作液(2mM),加2.5μL 4×ATP工作液到384孔分析板,在避光条件下室温培育30分钟。加5μL终止液到384分析板。最后加5μL检测混合物(0.084ng/μL的Anti-phospho-p53(ser15)-K和5ng/μL的Anti-GST-d2)到384孔分析板。室温孵育过夜,用ENVISION(Perkinelmer)仪器检测荧光信号(激发波长为320nm,发射波长为665nm和615nm)。通过各孔荧光强度值计算出每个孔中的抑制率:ER(Emission Ratio,发射光比率)=(665nm处荧光强度/615nm处荧光强度);抑制率=(ER阳性-ER待测化合物)/(ER阳性-ER阴性)×100%,利用参数拟合标准软件(GraphPad Prism 6.0)计算化合物的IC 50数值。
实施例的激酶数据结果:
Figure PCTCN2020094532-appb-000188
Figure PCTCN2020094532-appb-000189
实施例47:体外细胞增殖抑制实验:
本实验通过检测化合物在肿瘤细胞系TOV21G(卵巢癌)和MV4-11(白血病)中对体外细胞活性的影响,研究化合物抑制细胞增殖的作用。
TOV21G细胞与MV4-11细胞均购于美国典型培养物保藏中心(American Type Culture Collection,ATCC)。
TOV21G细胞培养于MCDB105/M199培养基(含15%FBS)中,细胞融合度达到85%以上时,用于试验。96-孔培养板每孔接种约1000个细胞,培养24小时,加入不同浓度待测化合物(0-10μM)处理细胞,每组设置3个平行复孔。设置空白孔(只含有培养基)和对照孔(接种细胞,不用药物处理)。培养120小时,每孔加入40μL Cell Titer-Glo溶液(Promega,#G7573),避光震荡孵育20min,每孔取100μL转移至96-孔白板中(Corning,#3917),用Biotek Synergy H1多功能酶标仪读取发光值。
MV-4-11细胞培养于IMDM培养基(含有20%FBS)中,96-孔培养板每孔接种约10000个细胞,加入不同浓度待测化合物(0-10μM)处理细胞,每组设置3个平行复孔。设置空白孔(只含有培养基)和对照孔(接种细胞,不用药物处理)。培养120小时,每孔加入40μL Cell Titer-Glo溶液,避光震荡孵育20min,每孔取100μL转移至96-孔白板中,用Biotek Synergy H1多功能酶标仪读取发光值。
抑制率(%)=100%×(对照孔-测试孔)/(对照孔-空白孔)
增殖试验结果证明测试化合物在研究的人肿瘤细胞中具有效力,以IC 50值反映(IC 50为在50%最大效力下的抑制浓度)。
CellTiter-Glo发光法细胞活性检测结果
编号 TOV21G/IC 50/μM MV4-11/IC 50/μM
D1 0.26 0.12
D2 0.92 0.78
D3 0.28 0.29
D4 0.33 0.19
D6 0.24 0.17
D7 0.46 0.31
D8 0.43 0.22
D10 0.38 0.33
D11 0.64 0.37
D12 0.28 0.29
D13 0.80 0.25
D14 1.20 0.81
D15 0.77 0.25
D16 1.05 1.36
D17 0.23 0.14
D18 0.51 0.28
D19 0.11 0.08
D20 0.16 0.07
D21 0.14 0.19
D22 0.85 0.92
D23 0.58 0.69
D24 0.12 0.08
D25 1.03 0.99
D26 3.58 3.09
D27 0.72 0.97
D28 0.28 0.23
D29 0.71 0.50
D30a 2.09 0.41
D30b 2.22 0.33
D31 NT NT
D32 NT NT
D33 1.45 1.31
D34 NT NT
D35 0.74 0.84
D36 0.66 0.64
D37 NT NT
D38 0.14 0.23
D39 0.86 1.23
D40 0.72 0.58
D41 0.94 0.57
D42 2.01 1.73
D43 0.21 0.54
D44 0.79 0.68
D45 0.92 0.64
D46 1.33 0.53
D47 0.60 0.55
D48 0.55 0.20
D49 0.50 0.55
D50 NT 0.81
D51 0.68 0.66
D52 0.37 0.59
D53 0.71 0.62
D54a 0.18 0.09
D54b 0.44 0.26
D55 0.19 0.22
D56 0.44 0.32
D57 0.41 0.22
实施例48:化合物体外肝微粒体稳定性实验:
对本发明化合物进行肝微粒体稳定性实验研究。将待测化合物(终浓度2.0nM)在加入或不加入NADPH情况下与人/小鼠的肝微粒体进行共孵育,检测60分钟内孵育上清中的化合物浓度。代表性化合物的结果如下:
Figure PCTCN2020094532-appb-000190
实施例49:化合物体内药代动力学实验:
对本发明化合物进行体内药代动力学研究。
实验方法:
雄性ICR小鼠(3只/组)口服灌胃给药,给药剂量为10mg/kg。收集给药前(0小时)和给药后(0.25,0.5,1,2,4,6,8,24小时)的血浆样品,对收集的样品进行LC/MS/分析并采集数据,采集的分析数据用Analyst v1.6.2,(AB Applied Biosystems Company,USA)软件计算相关药代动力学参数。
代表性化合物的结果如下:
Figure PCTCN2020094532-appb-000191
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。

Claims (26)

  1. 通式(A)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2020094532-appb-100001
    其中,
    X为CR 5或N;Y为CR 6或N;并且其中X和Y中至少有一个为N;
    Z为CR #或N;
    R 1、R 2、R 3、R 4和R #独立地选自H、C 1-6烷基和C 1-6卤代烷基;或者R 1与R 2,R 3与R 4连接形成键、C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;
    R 5和R 6独立地选自H、卤素、C 1-6烷基或C 1-6卤代烷基;
    当Z为CR #时,其中R #、R 1与它们连接的原子一起形成C 3-5元环烷基或3-5元杂环基;
    环A选自C 6-10芳基和5-10元杂芳基;
    环B选自5-10元杂芳基;
    R a独立地选自任选被R**取代的H、卤素、-CN、-L-NRR’、-L-OR、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基;
    其中L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;p=1或2;
    R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR、-L’-C 3-7环烷基和-L’-4-8元杂环基;其中L’独立地选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、-O-C 1-6亚烷基和-NH-C 1-6亚烷基;
    R b独立地选自任选被R**取代的H、卤素、-CN、-L-OR、-L-C 1-6烷基、-L-C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’和-NRC(O)NRR’;
    R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
    R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基,或者R、R’与它们连接的氮原子形成4-8元杂环基;
    m=0、1、2、3、4或5;
    n=0、1、2、3、4或5。
  2. 通式(I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2020094532-appb-100002
    其中,
    X为CH或N;Y为CH或N;并且其中X和Y中至少有一个为N;
    R 1、R 2、R 3和R 4独立地选自H、C 1-6烷基和C 1-6卤代烷基;或者R 1与R 2,R 3与R 4连接形成键、C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;
    环A选自C 6-10芳基和5-10元杂芳基;
    环B选自5-10元杂芳基;
    R a独立地选自任选被R**取代的H、卤素、-CN、-L-NRR’、-L-OR、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基;
    其中L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;p=1或2;
    R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR、-L’-C 3-7环烷基和-L’-4-8元杂环基;其中L’独立地选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基、-O-C 1-6亚烷基和-NH-C 1-6亚烷基;
    R b独立地选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R、-C(O)OR、-C(O)NRR’、-OC(O)R’、-NRC(O)R’、-OC(O)NRR’和-NRC(O)NRR’;
    R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
    R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基,或者R、R’与它们连接的氮原子形成4-8元杂环基;
    m=0、1、2、3、4或5;
    n=0、1、2、3、4或5。
  3. 权利要求1或2的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R 1和R 2中至少一个为C 1-6烷基,优选地为(R)-C 1-6烷基,更优选为(R)-甲基。
  4. 权利要求1-3中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,环A选自以下基团:
    Figure PCTCN2020094532-appb-100003
    更优选地,环A选自以下基团:
    Figure PCTCN2020094532-appb-100004
    更优选地,环A选自:
    Figure PCTCN2020094532-appb-100005
    其中
    Figure PCTCN2020094532-appb-100006
    表示与分子其余部分的连接位置,R a1-R a11如权利要求1中R a所定义。
  5. 权利要求1-4中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,环B选自双环的9-10元杂芳基;优选地,环B选自以下基团:
    Figure PCTCN2020094532-appb-100007
    其中
    Figure PCTCN2020094532-appb-100008
    表示与分子其余部分的连接位置,Z 1-Z 12中有0、1、2或3个为选自O、S和N的杂原子,只要化学上允许;并且Z 1-Z 12任选被氧化形成=O基团;优选地,Z 1-Z 12中至少一个为选自O、S和N的 杂原子,只要化学上允许;并且Z 1-Z 12任选被氧化形成=O基团;
    优选地,Z 1-Z 12中有0、1、2或3个为N原子;优选地,Z 1-Z 12中至少一个为N原子;优选地,Z 1-Z 3中最多含有一个N原子;优选地,Z 4-Z 6中最多含有两个N原子;优选地,Z 4为N原子;
    更优选地,环B选自以下基团:
    Figure PCTCN2020094532-appb-100009
    更优选地,环B选自
    Figure PCTCN2020094532-appb-100010
    更优选地,环B选自
    Figure PCTCN2020094532-appb-100011
  6. 权利要求1-5中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R a独立地选自任选被R**取代的H、卤素、-L-NRR’、-L-OR、-NRC(O)NRR’、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-L-S(O) pR*、-L-S(=O)(NR)R*、-L-C 3-7环烷基和-L-4-8元杂环基。
  7. 权利要求1-6中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R b独立地选自任选被R**取代的H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’,优选地,R b独立地选自任选被R**取代的H、卤素、-CN、-L-OR、-L-C 1-6烷基、-L-C 1-6烷氧基、-L-C 3-7环烷基、-NRR’和-NRC(O)R’。
  8. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(II)的结构:
    Figure PCTCN2020094532-appb-100012
    其中,Y、环A、环B、R b和n如权利要求1-7中所定义;
    优选地,
    环A选自:
    Figure PCTCN2020094532-appb-100013
    R a1选自H、C 1-6烷基和C 1-6卤代烷基;优选地为H或甲基;
    R a2选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基或异丙基;
    R a3选自H、C 1-6烷基和C 1-6卤代烷基;优选地为H或甲基;
    R a4选自H和S(O) pR*,其中p=1或2,R*选自C 1-6烷基和C 1-6卤代烷基;优选地为H或-SO 2Me;
    R a9选自H和-NRC(O)NRR’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;优选地为H或-NH 2C(O)NH 2Et;
    R a10选自C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基;优选地为CF 2H或C 1-6烷氧基;
    R a11选自H和-NRR’,其中R和R’独立地选自H、C 1-6烷基,C 1-6卤代烷基;优选地为H;
    B选自
    Figure PCTCN2020094532-appb-100014
    R b选自氢、卤素、C 1-6烷基和C 1-6卤代烷基;优选地为甲基、卤素或甲氨基;
    n=0、1、2、3或4;优选地,n=0、1或2。
  9. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(III)的结构:
    Figure PCTCN2020094532-appb-100015
    其中,Y、环A、R a和m如权利要求1-7中所定义;
    优选地,
    环A选自:
    Figure PCTCN2020094532-appb-100016
    R a1选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基;
    R a2选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基或异丙基;
    R a3选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基。
  10. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(IV)的结构:
    Figure PCTCN2020094532-appb-100017
    其中,
    X为CH或N;
    环A选自:
    Figure PCTCN2020094532-appb-100018
    R a1选自H、C 1-6烷基和C 1-6卤代烷基;优选地为H或甲基;
    R a2选自任选被R**取代的C 1-6烷基、C 1-6卤代烷基、-L-NRR’、-L-OR、-L-4-8元杂环基和-L-S(O) pR*;其中L选自键、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基,p=1或2,R*选自C 1-6烷基、C 3-7环烷基、4-8元杂环基、C 1-6卤代烷基和-NRR’;R**选自H、卤素、-OR、-C(O)R、C 1-6烷基、C 1-6卤代烷基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
    优选地,R a2选自C 1-6烷基、C 1-6卤代烷基、-L-NRR’、-L-OR和-L-4-8元杂环基,其中L为键或C 1-6亚烷基,R或R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;优选地为甲基、异丙基、-CH 2CH 2CH 2OH、-CH 2CH 2OMe、-CH 2CH 2NMe 2、-CH 2CH 2CH 2NMe 2、氮杂环丁烷-3-基甲基、氧杂环丁烷-3-基甲基、吡喃-4-基、四氢吡咯基或哌啶基;
    R a3选自H、C 1-6烷基和C 1-6卤代烷基;优选地为H、甲基、CF 3
    R a4选自任选被R**取代的C 1-6烷基、-L-S(=O)(NR)R*、-L-S(O) pR*和-L-4-8元杂环基;其中,p=1或2,L选自键、-NR-、-C(O)-和C 1-6亚烷基,R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR和-L’-4-8元杂环基;并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自H、-CN、-NRR’、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基和-S(O) p-C 1-6卤代烷基,其中p=1或2;
    优选地,R a4选自-S(O) pR*,其中p=1或2,R*选自C 1-6烷基、4-8元杂环基、C 1-6卤代烷基和-L’-NRR’,并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;优选地为-SO 2N(Me) 2、-SO 2Me、-SO 2iPr或-SO 2-哌啶-4-基;
    R a5选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基和C 1-6卤代烷氧基;优选地选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基、甲氧基、异丙基或三氟甲基;
    R a6为H;
    R a7为H;
    R a8选自C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基;优选地为C 1-6烷基和C 1-6卤代烷基;优选地为Me;
    R a10选自任选被-CN取代的C 1-6烷基和C 1-6卤代烷基;
    R a11为H;
    R b选自氢、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R;优选地为氢、卤素、甲氧基或-CH 2-氮杂环丁烷-3-基;
    n=0、1、2或3。
  11. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(IV)的结构:
    Figure PCTCN2020094532-appb-100019
    其中,
    X为CH或N;
    环A选自:
    Figure PCTCN2020094532-appb-100020
    优选地为
    Figure PCTCN2020094532-appb-100021
    R a1选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基;
    R a2选自任选被R**取代的C 1-6烷基、C 1-6卤代烷基、-L-NRR’、-L-4-8元杂环基、-L-S(O) pR*,其中L选自键、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基,p=1或2,R*选自C 1-6烷基、C 1-6卤代烷基和-NRR’,并且其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自卤素、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基和-S(O) p-C 1-6卤代烷基,其中p=1或2;
    优选地,R a2选自C 1-6烷基、C 1-6卤代烷基和-L-4-8元杂环基;优选地为甲基、异丙基、-CH 2CH 2CH 2NMe 2、吡喃-4-基、四氢吡咯基或哌啶基;
    R a3选自H、C 1-6烷基和C 1-6卤代烷基;优选地为甲基、CF 3
    R a4选自任选被R**取代的C 1-6烷基、-L-S(=O)(NR)R*、-L-S(O) pR*和-L-4-8元杂环基;其中,p=1或2,L选自键、-NR-、-C(O)-和C 1-6亚烷基,R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’、-L’-OR和-L’-4-8元杂环基;并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自-CN、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基和-S(O) p-C 1-6卤代烷基,其中p=1或2;
    优选地,R a4选自-S(O) pR*,其中p=1或2,R*选自C 1-6烷基、C 1-6卤代烷基、-4-8元杂环基和-L’-NRR’,并且其中L’选自键、C 1-6亚烷基、C 2-6亚烯基、C 2-6亚炔基和-NH-C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;优选地为-SO 2N(Me) 2、-SO 2Me、-SO 2iPr或-SO 2-哌啶-4-基;
    R a5选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基和C 1-6卤代烷氧基;优选地选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基、异丙基、甲氧基或三氟甲基;
    R a10选自任选被-CN取代的C 1-6烷基和C 1-6卤代烷基;
    R a11为H;
    R b选自氢、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-C(O)R;优选地为氢、卤素、甲氧基或-CH 2-氮杂环丁烷-3-基;
    n=0、1、2或3。
  12. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(V)的结构:
    Figure PCTCN2020094532-appb-100022
    其中,
    X为CH或N;
    R a1选自C 1-6烷基和C 1-6卤代烷基;
    R a2选自任选被R**取代的-L-S(O) pR*、-L-S(=O)(NR)R*、C 1-6烷基和-L-4-8元杂环基;
    R a3选自C 1-6烷基和C 1-6卤代烷基;
    环B为
    Figure PCTCN2020094532-appb-100023
    Figure PCTCN2020094532-appb-100024
    R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’;
    n=0、1、2、3、4或5;
    p=1或2;
    R*选自C 1-6烷基或C 1-6卤代烷基;
    L选自键、-O-、-S-、-NR-、-C(O)-或C 1-6亚烷基;
    R**独立地选自H、卤素、-CN、-C(=O)R、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、4-8元杂环基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’;
    R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
  13. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(V)的结构:
    Figure PCTCN2020094532-appb-100025
    其中,
    X为CH或N;
    R a1选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基;
    R a2选自C 1-6烷基和-L-4-8元杂环基;优选地为甲基或哌啶基;
    R a3选自C 1-6烷基和C 1-6卤代烷基;优选地为甲基或三氟甲基;
    环B选自:
    Figure PCTCN2020094532-appb-100026
    Figure PCTCN2020094532-appb-100027
    R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
    L选自键和C 1-6亚烷基;
    n=0、1、2、3、4或5;优选地,n=0、1或2。
  14. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(VI)的结构:
    Figure PCTCN2020094532-appb-100028
    其中,
    X为CH或N;
    R a4选自任选被R**取代的-L-S(O) 2R*基团;其中,L选自键和C 1-6亚烷基,R*选自C 1-6烷基、C 1-6卤代烷基、-L’-NRR’和-L’-4-8元杂环基;并且其中L’选自键和C 1-6亚烷基,R和R’独立地选自H、C 1-6烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;R**选自氢、-CN、C(=O)R、C 1-6烷基、C 1-6卤代烷基、-S(O) p-C 1-6烷基、-S(O) p-C 1-6卤代烷基和-NRR’,其中p=1或2;
    优选地,R a4选自-S(O) 2R*,其中,R*选自C 1-6烷基、-L’-NRR’和4-8元杂环基,并且其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基或者R、R’与它们连接的氮原子形成4-8元杂环基;L’选自键和C 1-6亚烷基;优选地为-SO 2N(Me) 2、-SO 2Me、-SO 2iPr或-SO 2-哌啶-4-基;
    R a5选自甲基、甲氧基、异丙基或三氟甲基;
    环B选自:
    Figure PCTCN2020094532-appb-100029
    Figure PCTCN2020094532-appb-100030
    R b选自H、卤素、-CN、-OH、C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’和-NRC(O)R’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基;
    n=0、1、2、3、4或5;优选地,n=0、1或2。
  15. 权利要求1的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(VII)的结构:
    Figure PCTCN2020094532-appb-100031
    其中,
    环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
    R a1为C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基或C 1-6卤代烷氧基(优选甲基、三氟甲基或甲氧基);
    R a3为C 1-6烷基或C 1-6卤代烷基(优选甲基或三氟甲基);
    R a12为H、C 1-6烷基或C 1-6卤代烷基;
    R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基;
    Z 1为N或CR b1
    Z 2为N或CR b2
    Z 3为N或CR b3
    Z 4为NR b4或C(R b4) 2
    Z 5为N或CR b5
    Z 6为N或CR b6
    R b1、R b2、R b3、R b4、R b5和R b6独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基(优选地选自H、卤素和甲氧基);
    优选地,
    R a1为C 1-6烷基或C 1-6卤代烷基(优选甲基或三氟甲基);
    R b2为H或卤素(优选H或F);
    R b3为优选H;
    优选地,
    R a1和R a3中至少一个为C 1-6卤代烷基。
  16. 权利要求1的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(VIII)的结构:
    Figure PCTCN2020094532-appb-100032
    其中,
    环C为
    Figure PCTCN2020094532-appb-100033
    其中与分子其余部分的连接点在其中一个*所示的位置;
    R a1为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
    R a3为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
    R a12为H或C 1-6烷基(优选为H或甲基);
    R 5为H或卤素(优选为H或F);
    环B选自
    Figure PCTCN2020094532-appb-100034
    R b为H、甲氧基或卤素(优选为H或卤素);
    n为0、1、2或3;
    优选地,
    R a1和R a3中至少一个为C 1-6卤代烷基。
  17. 权利要求1的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(IX)的结构:
    Figure PCTCN2020094532-appb-100035
    其中,
    环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
    R a1为C 1-6烷基或C 1-6卤代烷基;
    R a3为C 1-6烷基或C 1-6卤代烷基;
    R a12为H、C 1-6烷基或C 1-6卤代烷基;
    R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基;
    Z 7为N或CR b7
    Z 8为N或CR b8
    Z 9为N或CR b9
    Z 10为N或CR b10
    Z 11为N或CR b11
    Z 12为N或CR b12
    其中,
    R b7为H、-L-OR、-L-C 1-6烷基、-L-C 3-7环烷基、-NRR’、-NRC(O)R’或-NRC(O)NRR’;
    R b8、R b9、R b10、R b11和R b12独立地选自H、卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基和C 1-6卤代烷氧基(优选地选自H、卤素、-CN和甲氧基);
    L独立地选自键、-O-、-S-、-NR-、-C(O)-、C 1-6亚烷基、C 2-6亚烯基和C 2-6亚炔基;
    R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
  18. 权利要求17的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中Z 7为N或CR b7
    其中,
    R b7选自-NRR’、-NRC(O)R’或-NRC(O)NRR’;
    其中,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
  19. 权利要求1的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(X)的结构:
    Figure PCTCN2020094532-appb-100036
    其中,
    环C为
    Figure PCTCN2020094532-appb-100037
    其中与分子其余部分的连接点在其中一个*所示的位置;
    R a1为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
    R a3为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
    R a12为H或C 1-6烷基(优选为H或甲基);
    R 5为H;
    R b7选自-NRR’、-SR、-NRC(O)R’或-NRC(O)NRR’;其中,R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基(优选地,R b7选自NH 2、NHMe、NHEt、NHC(O)Me或NHC(O)Et);
    R b为H或卤素(优选为H或F)。
  20. 权利要求1的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其具有通式(XI)的结构:
    Figure PCTCN2020094532-appb-100038
    其中,
    环C为被R a12取代的C 3-7环烷基或4-8元杂环基(优选被R a12取代的5-6元杂环基,优选被R a12取代的含N原子的5-6元杂环基);
    R a1为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
    R a3为C 1-6烷基或C 1-6卤代烷基(优选为甲基或三氟甲基);
    R a12为H、C 1-6烷基或C 1-6卤代烷基(优选为H或甲基);
    R 5为H、卤素、C 1-6烷基或C 1-6卤代烷基(优选为H或F);
    环B选自:
    Figure PCTCN2020094532-appb-100039
    R b选自H、卤素、-CN、-L-C 1-6烷基、C 1-6烷氧基、-L-C 3-7环烷基、-L-3-8元杂环基、-NRR’、-NRC(O)R’和-NRC(O)NRR’,其中R和R’独立地选自H、C 1-6烷基和C 1-6卤代烷基。
  21. 权利要求1的化合物,其选自:
    Figure PCTCN2020094532-appb-100040
    Figure PCTCN2020094532-appb-100041
    Figure PCTCN2020094532-appb-100042
    Figure PCTCN2020094532-appb-100043
    Figure PCTCN2020094532-appb-100044
    Figure PCTCN2020094532-appb-100045
    Figure PCTCN2020094532-appb-100046
  22. 药物组合物,其含有权利要求1-21中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
  23. 权利要求1-21中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体在制备用于治疗和/或预防ATR激酶介导的疾病的药物中的用途。
  24. 一种在受试者中治疗和/或预防ATR激酶介导的疾病的方法,所述方法包括向所述受试者给药权利要求1-21中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求22的药物组合物。
  25. 权利要求1-21中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求22的药物组合物,其用于治疗和/或预防ATR激酶介导的疾病。
  26. 权利要求23的用途或权利要求24的方法或权利要求25的化合物或组合物的用途,其中所述ATR激酶介导的疾病包括增生性疾病(如癌症),尤其实体瘤(如癌和肉瘤)及白血病和淋巴癌,尤其对于例如乳腺癌、结肠直肠癌、肺癌(包括小细胞肺癌、非小细胞肺癌和细支气管肺泡癌)和前列腺癌及胆管癌、骨癌、膀胱癌、头及颈癌、肾癌、肝癌、胃肠组织癌、食道癌、卵巢癌、胰腺癌、皮肤癌、睾丸癌、甲状腺癌、子宫癌、子宫颈癌和外阴癌、以及白血病[包括急性淋巴细胞白血病(ALL)和慢性髓细胞源性白血病(CML)]、多发性骨髓瘤和淋巴癌。
PCT/CN2020/094532 2019-06-06 2020-06-05 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物 WO2020244613A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
KR1020217043346A KR20220016225A (ko) 2019-06-06 2020-06-05 Atr 키나제 억제제로서의 2,4,6-삼치환된 피리미딘 화합물
JP2021572544A JP7485701B2 (ja) 2019-06-06 2020-06-05 Atrキナーゼ阻害剤としての2,4,6-三置換ピリミジン化合物
CN202080040989.2A CN113906026A (zh) 2019-06-06 2020-06-05 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
US17/616,571 US20220378799A1 (en) 2019-06-06 2020-06-05 2, 4, 6-tri-substituted pyrimidine compound as atr kinase inhibitor
EP20818265.9A EP3967694A4 (en) 2019-06-06 2020-06-05 TRI-SUBSTITUTED PYRIMIDINE COMPOUND IN POSITIONS 2, 4, 6 USED AS ATR KINASE INHIBITOR

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201910490080.6 2019-06-06
CN201910490080 2019-06-06
CN201910876919.X 2019-09-17
CN201910876919 2019-09-17
CN202010441539 2020-05-22
CN202010441539.6 2020-05-22

Publications (1)

Publication Number Publication Date
WO2020244613A1 true WO2020244613A1 (zh) 2020-12-10

Family

ID=73608860

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/094532 WO2020244613A1 (zh) 2019-06-06 2020-06-05 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物

Country Status (7)

Country Link
US (1) US20220378799A1 (zh)
EP (1) EP3967694A4 (zh)
JP (1) JP7485701B2 (zh)
KR (1) KR20220016225A (zh)
CN (2) CN112047938B (zh)
TW (1) TWI778366B (zh)
WO (1) WO2020244613A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4155307A4 (en) * 2020-05-20 2023-09-13 Beijing Tide Pharmaceutical Co., Ltd. 2,4,6-TRI-SUBSTITUTED PYRIMIDINE COMPOUNDS AS ATR KINASE INHIBITORS

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114213424B (zh) * 2021-12-30 2023-05-26 杭州高光制药有限公司 一种呋喃[3,2-b]并吡啶衍生物的合成方法
CN115745995A (zh) * 2022-01-10 2023-03-07 苏州浦合医药科技有限公司 Atr抑制剂及其用途
WO2023215133A1 (en) * 2022-05-02 2023-11-09 AcuraStem Incorporated Pikfyve kinase inhibitors
CN115043770B (zh) * 2022-07-21 2023-09-08 南京大学 一种吲哚/氮杂吲哚类化合物的光诱导合成方法

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CN101010318A (zh) * 2004-07-09 2007-08-01 阿斯利康(瑞典)有限公司 作为磷脂酰肌醇(pi)3-激酶抑制剂的2,4,6-三取代的嘧啶及其在癌症治疗中的用途
WO2008032077A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab Pyrimidine derivatives
WO2009093981A1 (en) * 2008-01-23 2009-07-30 S Bio Pte Ltd Triazine compounds as kinase inhibitors
CN101535296A (zh) * 2006-09-14 2009-09-16 阿斯利康(瑞典)有限公司 作为p13k和mtor抑制剂用于治疗增殖性疾病的2-苯并咪唑基-6-吗啉代-4-苯基嘧啶衍生物
CN101765597A (zh) * 2007-04-12 2010-06-30 霍夫曼-拉罗奇有限公司 作为磷脂酰肌醇-3-激酶抑制剂的嘧啶衍生物
CN101790525A (zh) * 2007-04-12 2010-07-28 霍夫曼-拉罗奇有限公司 药物化合物
WO2011005119A1 (en) * 2009-07-07 2011-01-13 Pathway Therapeutics Limited Pyrimidinyl and 1,3,5-triazinyl benzimidazoles and their use in cancer therapy
WO2012044641A1 (en) * 2010-09-29 2012-04-05 Pathway Therapeutics Inc. 1,3,5-triazinyl benzimidazole sulfonamides and their use in cancer therapy
CN102448958A (zh) * 2009-03-27 2012-05-09 帕斯维医疗公司 嘧啶基和1,3,5-三嗪基苯并咪唑磺酰胺和其在癌症治疗中的用途
WO2019050889A1 (en) * 2017-09-08 2019-03-14 Bluevalley Pharmaceutical Llc SUBSTITUTED PYRROLOPYRIDINES AS ATR INHIBITORS
WO2019154365A1 (zh) * 2018-02-07 2019-08-15 南京明德新药研发有限公司 一种atr抑制剂及其应用
CN110343095A (zh) * 2018-04-08 2019-10-18 中国科学院上海药物研究所 一类精氨酸甲基转移酶抑制剂及其药物组合物和用途
CN110467610A (zh) * 2018-05-10 2019-11-19 四川科伦博泰生物医药股份有限公司 一种取代嘧啶化合物、其制备方法和用途

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK1020462T3 (da) * 1997-07-24 2004-04-26 Zenyaku Kogyo Kk Heterocykliske forbindelser og antitumormiddel indeholdende disse som aktiv ingrediens
MA40933A (fr) * 2014-11-11 2017-09-19 Piqur Therapeutics Ag Difluorométhyl-aminopyridines et difluorométhyl-aminopyrimidines
CN108602802B (zh) * 2016-07-26 2020-01-21 深圳市塔吉瑞生物医药有限公司 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
CN111867590B (zh) * 2017-07-13 2023-11-17 德州大学系统董事会 Atr激酶的杂环抑制剂
US10800774B2 (en) * 2017-08-17 2020-10-13 Board Of Regents, The University Of Texas System Heterocyclic inhibitors of ATR kinase
CN109627263B (zh) * 2017-12-21 2022-05-20 深圳市塔吉瑞生物医药有限公司 用于抑制激酶活性的二苯氨基嘧啶类化合物
BR112022001393A2 (pt) * 2019-08-06 2022-03-22 Wuxi Biocity Biopharmaceutics Co Ltd Forma cristalina do inibidor de atr e seu uso
WO2021233376A1 (zh) * 2020-05-20 2021-11-25 北京泰德制药股份有限公司 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
AU2021282122B2 (en) * 2020-05-29 2023-09-21 Wuxi Life Fountain Biotech Co., Ltd Fluoropyrrolopyridine compound and application thereof

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CN101010318A (zh) * 2004-07-09 2007-08-01 阿斯利康(瑞典)有限公司 作为磷脂酰肌醇(pi)3-激酶抑制剂的2,4,6-三取代的嘧啶及其在癌症治疗中的用途
WO2008032077A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab Pyrimidine derivatives
CN101535296A (zh) * 2006-09-14 2009-09-16 阿斯利康(瑞典)有限公司 作为p13k和mtor抑制剂用于治疗增殖性疾病的2-苯并咪唑基-6-吗啉代-4-苯基嘧啶衍生物
CN101790525A (zh) * 2007-04-12 2010-07-28 霍夫曼-拉罗奇有限公司 药物化合物
CN101765597A (zh) * 2007-04-12 2010-06-30 霍夫曼-拉罗奇有限公司 作为磷脂酰肌醇-3-激酶抑制剂的嘧啶衍生物
WO2009093981A1 (en) * 2008-01-23 2009-07-30 S Bio Pte Ltd Triazine compounds as kinase inhibitors
CN102448958A (zh) * 2009-03-27 2012-05-09 帕斯维医疗公司 嘧啶基和1,3,5-三嗪基苯并咪唑磺酰胺和其在癌症治疗中的用途
WO2011005119A1 (en) * 2009-07-07 2011-01-13 Pathway Therapeutics Limited Pyrimidinyl and 1,3,5-triazinyl benzimidazoles and their use in cancer therapy
WO2012044641A1 (en) * 2010-09-29 2012-04-05 Pathway Therapeutics Inc. 1,3,5-triazinyl benzimidazole sulfonamides and their use in cancer therapy
WO2019050889A1 (en) * 2017-09-08 2019-03-14 Bluevalley Pharmaceutical Llc SUBSTITUTED PYRROLOPYRIDINES AS ATR INHIBITORS
WO2019154365A1 (zh) * 2018-02-07 2019-08-15 南京明德新药研发有限公司 一种atr抑制剂及其应用
CN110343095A (zh) * 2018-04-08 2019-10-18 中国科学院上海药物研究所 一类精氨酸甲基转移酶抑制剂及其药物组合物和用途
CN110467610A (zh) * 2018-05-10 2019-11-19 四川科伦博泰生物医药股份有限公司 一种取代嘧啶化合物、其制备方法和用途

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences,", MACK PUBLISHING COMPANY
BERGE S. M. ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
D. FLEISHERS. RAMONH. BARBRA: "Advanced Drug Delivery Reviews", vol. 19, 1996, article "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", pages: 115 - 130
M. B. SMITHJ. MARCH: "March' s Advanced Organic Chemistry", 2001, JOHN WILEY & SONS
See also references of EP3967694A4
T. HIGUCHIV. STELLA: "Prodrugs as Novel Delivery Systems, A.C.S. Symposium Series", 1987, PERGAMON PRESS

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4155307A4 (en) * 2020-05-20 2023-09-13 Beijing Tide Pharmaceutical Co., Ltd. 2,4,6-TRI-SUBSTITUTED PYRIMIDINE COMPOUNDS AS ATR KINASE INHIBITORS

Also Published As

Publication number Publication date
CN112047938B (zh) 2022-11-22
TW202112764A (zh) 2021-04-01
KR20220016225A (ko) 2022-02-08
JP7485701B2 (ja) 2024-05-16
EP3967694A1 (en) 2022-03-16
EP3967694A4 (en) 2022-12-07
US20220378799A1 (en) 2022-12-01
TWI778366B (zh) 2022-09-21
JP2022536313A (ja) 2022-08-15
CN113906026A (zh) 2022-01-07
CN112047938A (zh) 2020-12-08

Similar Documents

Publication Publication Date Title
WO2021219091A2 (zh) 喹喔啉酮衍生物作为kras g12c突变蛋白的不可逆抑制剂
WO2020244613A1 (zh) 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
TWI758832B (zh) 抑制並誘導降解egfr和alk的化合物
BR112019012239A2 (pt) compostos de aminotiazol como inibidores de c-kit
CN108699039B (zh) 用于抑制蛋白激酶活性的(杂)芳基酰胺类化合物
WO2019201131A1 (zh) 用于抑制蛋白激酶活性的二(杂)芳基大环化合物
WO2023134266A1 (zh) 2-哌啶基或2-吡唑基取代的嘧啶化合物作为egfr抑制剂
TW202102511A (zh) 噻吩并雜環類衍生物、其製備方法及其在醫藥上的應用
CN113348170A (zh) 联苯类衍生物抑制剂、其制备方法和应用
WO2021233376A1 (zh) 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
WO2022012484A1 (zh) 作为atr激酶抑制剂的吡唑并嘧啶化合物
WO2021057796A1 (zh) 取代的稠合三环衍生物及其组合物及用途
WO2021023104A1 (zh) 1h-[1,2,3]三唑并[4,5-h]喹唑啉类化合物作为蛋白激酶抑制剂
WO2023198180A1 (zh) Egfr降解剂
CN111349084B (zh) 用于抑制蛋白激酶活性的氨基嘧啶类化合物
WO2023207556A1 (zh) Prmt5-mta抑制剂
TW202334095A (zh) Sos1抑制劑

Legal Events

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

Ref document number: 20818265

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021572544

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020818265

Country of ref document: EP

Effective date: 20211206

ENP Entry into the national phase

Ref document number: 20217043346

Country of ref document: KR

Kind code of ref document: A