WO2022206730A1 - 嘧啶并吡嗪酮化合物及其用途 - Google Patents

嘧啶并吡嗪酮化合物及其用途 Download PDF

Info

Publication number
WO2022206730A1
WO2022206730A1 PCT/CN2022/083568 CN2022083568W WO2022206730A1 WO 2022206730 A1 WO2022206730 A1 WO 2022206730A1 CN 2022083568 W CN2022083568 W CN 2022083568W WO 2022206730 A1 WO2022206730 A1 WO 2022206730A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cycloalkyl
alkylene
substituted
independently selected
Prior art date
Application number
PCT/CN2022/083568
Other languages
English (en)
French (fr)
Other versions
WO2022206730A8 (zh
Inventor
张学军
臧杨
李群
陈浩民
陈登辉
刘礼飞
安丹
刘哲
李莉娥
杨俊�
Original Assignee
武汉人福创新药物研发中心有限公司
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 武汉人福创新药物研发中心有限公司 filed Critical 武汉人福创新药物研发中心有限公司
Publication of WO2022206730A1 publication Critical patent/WO2022206730A1/zh
Publication of WO2022206730A8 publication Critical patent/WO2022206730A8/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • 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 the fields of chemistry and medicine, in particular, the present invention relates to pyrimidopyrazinone compounds and uses thereof.
  • Methionine adenosyltransferase 2A (MAT2A), a member of the MAT family of proteins, is widely distributed in the body and is expressed in non-parenchymal cells of the liver and all extrahepatic tissues (Murray B, et al. World J Gastroenterol 2019;25(31):4300-4319.), the MAT family also has two members, MAT1A and MAT2B.
  • MAT2A inhibitors can reduce S-adenosylmethionine (SAM) levels, so MAT2A inhibitors are also called SAM inhibitors.
  • SAM is the main methyl donor in cells.
  • Protein arginine methyltransferase 5 (PRMT5) is a methylase that utilizes the methyl donor of SAM. SAM plays an important role in the PRMT5 pathway and can affect the activity of PRMT5. Studies have shown that MAT2A is a "synthetic lethal" target in MTAP-deficient tumors.
  • MTA a substrate of the MTAP enzymatic reaction, accumulates abundantly in MTAP-deficient cancers
  • MTA Is a potent and selective inhibitor of PRMT5 resulting in reduced PRMT5 methylation activity in MTAP-deficient cells
  • MAT2A produces PRMT5 substrate SAM
  • MAT2A deletion selectively reduces the growth and PRMT5 methylation activity of MTAP-deficient cells
  • MAT2A is selectively required in MTAP-deficient cancers (Marjon K, et al. Cell Rep. 2016;15(3):574-587).
  • MAT2A inhibitors offer a new treatment option for patients with MTAP-deficient tumors.
  • MAT2A is involved in the metabolism of cancer stem cells, and cancer stem cells require a large amount of methionine to maintain their own histone methylation, which is crucial for the growth and tumorigenicity of cancer stem cells. Inhibition of MAT2A, a key enzyme in the methionine cycle, can significantly inhibit the growth of cancer stem cells and tumor formation. And in human non-small cell lung cancer tissues, MAT2A protein is abnormally high expressed. When tumor stem cells from non-small cell lung cancer were transplanted into mice, MAT2A inhibitor almost completely suppressed tumor growth, while the chemotherapeutic drug cisplatin had little effect (Wang Z, et al. Nature medicine, 2019, 25(5): 1-13.).
  • MAT1A Conversion of MAT1A and MAT2A in liver cancer. Many studies have shown that MAT plays an important role in the occurrence of chronic liver disease and liver cancer. Under normal circumstances, MAT2A is mainly expressed in the fetal liver, and is gradually replaced by MAT1A with growth and development after birth. MAT1A maintains the differentiation state of hepatocytes. There is a dynamic balance between MAT1A and MAT2A in normal hepatocytes, which together maintain the homeostasis of intracellular SAM.
  • MAT1A:MAT2A transition In hepatocellular carcinoma, down-regulation of MAT1A expression levels and up-regulation of MAT2A occur, termed MAT1A:MAT2A transition, which dedifferentiates the liver, reduces SAM biosynthesis, and enhances proliferative signaling in the liver.
  • MAT1A:MAT2A transition In human hepatocellular carcinoma, the expression ratio of MAT1A:MAT2A is negatively correlated with cell growth and genomic instability, and directly correlated with hepatoma cell apoptosis and overall DNA methylation; a reduced ratio is associated with a higher degree of malignancy and a lower survival rate in HCC. prognostic markers.
  • chr9p21 chromosome 9p21
  • the chromosomal chr9p21 locus includes the CDKN2A gene, which encodes the key tumor suppressors p19-ARF and p16-INK4a, and Chr9p21 deletion frequently involves co-deletion of genes proximal to CDKN2A, the most important of which is methylthioadenosine phosphorylase (MTAP) Gene.
  • MTAP methylthioadenosine phosphorylase
  • MTAP deficiency is not only present in tissue culture cells, but also in primary leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer (NSLC), bladder cancer, astrocytoma, osteosarcoma, In head and neck cancer, mucinous chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-Hodgkin lymphoma, and mesothelioma.
  • NSC non-small cell lung cancer
  • the present invention aims to provide a compound that can effectively inhibit MAT2A, which can be used as an improvement or replacement of current drugs or MAT2A inhibitors.
  • the present invention proposes a compound represented by formula III, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug :
  • L 1 is selected from -O-, -S-, -N(R L1 )- and single bond;
  • R L1 is selected from hydrogen and C 1 -C 6 alkyl;
  • R 1 is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl ) and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkenyl), the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkane radicals, -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkenyl) are optionally One or more R 1a is substituted, and said R 1a is selected from halogen; when R 1a is more than one, said R 1a is the same or different;
  • R L1 and R 1 can be combined with L to form a 3- to 6-membered heterocycloalkyl optionally substituted with one or more R L1 ; when R L1 When there are more than one, the R L1 are the same or different; the 3- to 6-membered heterocycloalkyl, wherein 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S and N, and the rest are carbon atom;
  • R 2 is selected from unsubstituted or R 2a substituted C 6 -C 10 -aryl, unsubstituted or R 2a substituted 5- to 10-membered heteroaryl; when R 2a is multiple, the R 2a are the same or different; the 5- to 10-membered heteroaryl group, wherein 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S and N, and the rest are carbon atoms;
  • R 3 is selected from unsubstituted or R 3a -substituted C 6 -C 10 -aryl, unsubstituted or R 3a -substituted 5- to 10-membered heteroaryl, unsubstituted or R 3a -substituted ring A; when R When there are multiple 3a , the R 3a are the same or different; the ring A is wherein Ring Cx and Ring Cy are each independently selected from C5 - C6 -aryl, 5- to 6-membered heteroaryl, 5- to 6-membered cycloalkyl, and 5- to 6-membered heterocycloalkyl, and D and E are each independently is selected from C, CH, N, G and J are each independently selected from C, CH, CH2 , N, NH, O, S;
  • R A and R B are each independently selected from H, -CN, -OH, oxo, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 6 alkoxy, C 2 - C 6 alkenyl, C 2 -C 6 alkynyl, -NH 2 , -S(O) 0-2 -(C 1 -C 6 alkyl), -S(O) 0-2 -(C 6 -C 10 aryl), -CO-(C 1 -C 6 alkyl), -CO-(C 3 -C 14 cycloalkyl), -C 3 -C 14 cycloalkyl, -(C 1 -C 6 alkylene) alkyl)-(C 3 -C 14 cycloalkyl), C 6 -C 10 aryl, 3- to 14-membered heterocycloalkyl, 5- to 10-membered heteroaryl; the 3- to 14-membered hetero
  • R A and R B C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 Alkynyl, -NH 2 , -S(O) 0-2 -(C 1 -C 6 alkyl), -S(O) 0-2 -(C 6 -C 10 aryl), -CO-(C 1 -C 6 alkyl), -CO-(C 3 -C 14 cycloalkyl), -C 3 -C 14 cycloalkyl, -(C 1 -C 6 alkylene)-(C 3 -C 14 cycloalkyl), C 6 -C 10 aryl, 3- to 14-membered heterocycloalkyl, and 5- to 10-membered heteroaryl groups are each independently and optionally substituted with one or more R Aa ; when R Aa is multiple , the R Aa are
  • the R Aa are each independently selected from -OH, deuterium, halogen, C 1 -C 6 alkyl, -N(R An ) 2 , wherein each R An is independently selected from H, C 1 -C 6 alkane base, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 6 -C 10 aryl, 3 to 14 membered heterocycloalkyl, -(C 1 -C 6 alkylene)-(3 to 14-membered heterocycloalkyl), and 5- to 10-membered heteroaryl, -NHC(O)-(OC 1 -C 6 alkyl), -NO 2 , -CN, oxo, -C(O)OH, -C(O)-O-(C 1 -C 6 alkyl), -(C 1 -C 6 alkylene)-(C 1 -C 6 -alkoxy), -C(O)NH 2 , -C(O
  • Each alkyl, alkenyl, aryl, and heterocycloalkyl in said R Aa is each independently and optionally substituted with one or more R Abs each independently selected from -OH, -O -(C 1 -C 6 alkyl), halogen, -NH 2 , -(C 1 -C 6 alkylene)-NH 2 , -COOH, -CN and oxo;
  • R 4 is selected from hydrogen, halogen, -OH, -CN, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) are optionally
  • One or more R 4a is substituted, and said R 4a is selected from halogen; when R 4a is more than one, said R 4a is the same or different.
  • the alkyl group includes straight chain alkyl group and branched chain alkyl group.
  • L 1 is selected from -O-, -S-, -N(R L1 )- or a single bond;
  • R L1 is selected from hydrogen, C 1 -C 3 Alkyl; according to a preferred embodiment of the present invention, R L1 is selected from hydrogen, methyl, and ethyl.
  • L 1 is selected from -O-, or -NH-.
  • R 1 is selected from C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)-( C 3 -C 6 cycloalkyl); the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) ) is optionally substituted by one or more R 1a selected from halogen ; according to certain embodiments of the present invention, R 1 is selected from C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 3 alkylene)-(C 3 -C 6 cycloalkyl), said C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 3 cycloalkyl) Alky
  • -L 1 -R 1 is selected from: -NH-cyclopropyl, -O-cyclopropyl, -NH-CH 2 CF 3 , -O-CH 2 CF 3 , - NH- CH2 -cyclopropyl, -O- CH2 -cyclopropyl.
  • R 2 is selected from unsubstituted or substituted phenyl group by R 2a and unsubstituted or substituted by R 2a 5- to 10-membered heteroaryl group, the 5 To 10-membered heteroaryl, one of the ring atoms is N; according to a preferred embodiment of the present invention, R 2 is selected from unsubstituted or substituted phenyl by R 2a , unsubstituted or substituted by R 2a pyridyl, unsubstituted or pyrimidinyl substituted by R 2a .
  • R 3 is a ring A substituted by R 3a ;
  • the R 3a is selected from -C 1 -C 6 alkyl, -(C 1 -C 6 alkylene Alkyl)-5-6 membered heterocycloalkyl, -(C 1 -C 6 alkylene)-S(O) 2 -CH 3 , -(C 1 -C 6 alkylene)-C(O) -N(CH 3 ) 2 ;
  • the ring A is Wherein, the ring Cx is a 5-membered heteroaromatic ring or a 5-membered heterocycloalkyl; the ring Cy is a benzene ring, a 5-6-membered heteroaromatic ring or a 5-6-membered heterocycloalkyl; D and E are each independently selected from C , CH, N; preferably, the ring Cx is a 5-membered heteroaromatic ring
  • R 3 is selected from unsubstituted or substituted phenyl by R 3a , unsubstituted or substituted by R 3a pyridyl, unsubstituted or substituted by R 3a Pyridonyl, unsubstituted or substituted by R 3a pyridazinyl, unsubstituted or substituted by R 3a isoxazolyl, unsubstituted or substituted by R 3a pyrazolyl, and unsubstituted or substituted by R 3a Ring A selected from benzothiazolyl, benzisothiazolyl, benzoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, quinoxalinyl, quinolinyl, quinazolinyl, imidazopyridyl , pyrazolopyridyl, triazolopyridyl, benzopyr
  • R 2a and R 3a are each independently selected from -RA , -OR A , halogen and -CN.
  • R A is independently selected from H, -OH, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -CO-(C 1 -C 6 alkyl), -(C 1 -C 6 alkylene)-(C 3 -C 14 cycloalkyl), the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, - CO-(C 1 -C 6 alkyl), -(C 1 -C 6 alkylene)-(C 3 -C 14 cycloalkyl) are each independently and optionally substituted with one or more R Aa .
  • R Aa is independently selected from -OH, halogen, -N(R An ) 2 , wherein each R An is independently selected from H, methyl , ethyl, isopropyl, n-propyl.
  • R 4 is independently selected from hydrogen, halogen, -OH, -CN, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) is optionally substituted with one or more R 4a selected from fluorine and chlorine; when R 4a is more than one, the R 4a are the same or different.
  • R 4 is selected from hydrogen, halogen, -OH, -CN, methyl, ethyl, n-propyl, propyl, fluoromethyl, fluoro Ethyl, fluoropropyl, cyclopropyl, fluorocyclopropyl, Chloromethyl, Chloroethyl, Chloropropyl, Chlorocyclopropyl, Preferably, R 4 is selected from hydrogen, halogen, methyl, ethyl, -CH 2 -CF 3 , cyclopropyl, R 4 is preferably hydrogen.
  • R 2 is selected from phenyl substituted by R 2a and pyridyl substituted by R 2a , and R 2a is independently selected from fluorine, chlorine, bromine, any -RA substituted with one or more R Aa, -OR A optionally substituted with one or more R Aa independently selected from C 1 -C substituted with one or more R Aa 6 alkyl, C 3 -C 6 cycloalkyl substituted by one or more R Aa , each R Aa is independently selected from -OH, fluorine, chlorine, bromine; according to a preferred embodiment of the present invention, R 2a independently selected from -fluoro, chloro, bromo, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, methoxy, ethoxy, n-propoxy, isopropoxy, Fluoromethoxy; according to certain embodiments of the present invention,
  • R 3 is selected from ring A substituted by R 3a , and said ring A is selected from and R 3a is independently selected from -RA optionally substituted with one or more R Aa independently selected from C 1 -C 6 alkyl substituted with one or more R Aa , each R Aa is independently selected from -OH, fluorine, chlorine, bromine; according to a preferred embodiment of the present invention, R 3a is independently selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl group, tert-butyl, tert-butyl, -butyl, -CH 2 -OH, -(CH 2 ) 2 -OH, -(CH 2 ) 3 -OH, -(CH 2 ) 4 -OH,
  • R 3 is selected from: R 3a is selected from: -C 1 -C 6 alkyl, -C 1 -C 6 alkyl-5-6 membered heterocycloalkyl, -C 1 -C 6 alkyl-S(O) 2 -CH 3 , -C 1 -C 6 alkyl-C(O)-N(CH 3 ) 2 ; preferably, the C 1 -C 6 alkyl group is a straight-chain or branched alkyl group; the C 1 -C 6 alkyl group is a straight-chain or branched-chain alkyl group; C6 alkyl is optionally substituted by -OH; preferably, the 5-6 membered heterocycloalkyl is Preferably, R 3a is selected from: -CH 3 , -
  • R 3 is selected from
  • the compound shown in formula III has the structure shown in formula II:
  • R 1 , R 2a , R 3a , R 4 , and L 1 have the above-mentioned definitions, and Y 1 , Y 2 , and Y 3 are each independently selected from N, S, C, and CH.
  • L 1 is selected from -O-, -S-, -N(R L1 )- and a single bond;
  • R L1 is selected from hydrogen and C 1 -C 6 alkyl;
  • R 1 is selected from C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), said C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) are optionally substituted by one or more R 1a ; when R 1a is When more than one, the R 1a is the same or different; the R 1a is selected from halogen;
  • X 1 is selected from C(R 2a ), N;
  • Y 1 , Y 2 , Y 3 are each independently selected from N, S, C, CH;
  • R 2a is independently selected from -RA , -OR A and halogen
  • R 3a is independently selected from -RA
  • R A is independently selected from H, -CN, -OH, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl substituted with one or more R Aa , C 1 -C 6 alkyl substituted with one or more R Aa or multiple R Aa substituted C 3 -C 6 cycloalkyl; when there are multiple R Aa , the R Aa are the same or different;
  • the R Aa are each independently selected from -OH, deuterium, halogen;
  • R 4 is selected from hydrogen, halogen, -OH, -CN, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) are optionally
  • One or more R 4a is substituted, and said R 4a is selected from halogen; when R 4a is more than one, said R 4a is the same or different.
  • L 1 is selected from -O-, -S-, -N(R L1 )- or a single bond;
  • R L1 is selected from hydrogen, C 1 -C 3 alkyl; preferably, R L1 is selected from hydrogen, methyl, and ethyl.
  • R 1 is selected from methyl, ethyl, propyl, fluoromethyl, fluoroethyl, fluoropropyl, cyclopropyl, According to a preferred embodiment of the present invention, R 1 is selected from methyl, ethyl, -CH 2 -CF 3 , cyclopropyl,
  • X 1 is selected from CH and N.
  • R 2a is independently selected from -fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, methoxy , ethoxy, n-propoxy, isopropoxy, Fluoromethoxy.
  • R 3a is independently selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, tert-butyl group, -CH 2 -OH, -(CH 2 ) 2 -OH, -(CH 2 ) 3 -OH, -(CH 2 ) 4 -OH,
  • R 4 is selected from hydrogen, halogen, -OH, -CN, methyl, ethyl, n-propyl, propyl, fluoromethyl, fluoro Ethyl, fluoropropyl, cyclopropyl, fluorocyclopropyl, Chloromethyl, Chloroethyl, Chloropropyl, Chlorocyclopropyl, Preferably, R 4 is selected from hydrogen, halogen, methyl, ethyl, -CH 2 -CF 3 , cyclopropyl,
  • the compound shown in formula II has the structure shown in formula II-a or II-b:
  • R 1 , L 1 , R 2a , and R 3a have the above-mentioned definitions.
  • the compound shown in formula III can also have the structure shown in formula I:
  • R 1 , L 1 , R 2a , R 3a , R 4 , and n A have the above-mentioned definitions.
  • L 1 is selected from -O-, -S-, -N(R L1 )- and a single bond;
  • R L1 is selected from hydrogen and C 1 -C 6 alkyl;
  • R 1 is selected from C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), said C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) are optionally substituted by one or more R 1a ; when R 1a is When more than one, the R 1a is the same or different; the R 1a is selected from halogen;
  • X 1 is selected from C(R 2a ), N;
  • R 2a is independently selected from -RA , -OR A and halogen
  • R 3a is independently selected from -RA
  • R A is independently selected from H, -CN, -OH, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 6 alkyl substituted with one or more R Aa , C 1 -C 6 alkyl substituted with one or more R Aa or multiple R Aa substituted C 3 -C 6 cycloalkyl; when there are multiple R Aa , the R Aa are the same or different;
  • the R Aa are each independently selected from -OH, deuterium, halogen;
  • R 4 is selected from hydrogen, halogen, -OH, -CN, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl), the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) are optionally
  • One or more R 4a is substituted, and said R 4a is selected from halogen; when R 4a is more than one, said R 4a is the same or different;
  • n A is selected from the integers 1, 2 or 3.
  • L 1 is selected from -O-, -S-, -N(R L1 )- or single bond;
  • R L1 is selected from hydrogen, C 1 -C 3 alkyl; preferably, R L1 is selected from hydrogen, methyl, and ethyl.
  • R 1 is selected from methyl, ethyl, propyl, fluoromethyl, fluoroethyl, fluoropropyl, cyclopropyl, Preferably, R 1 is selected from methyl, ethyl, -CH 2 -CF 3 , cyclopropyl,
  • X 1 is selected from CH and N.
  • R 2a is independently selected from -fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, methoxy , ethoxy, n-propoxy, isopropoxy, Fluoromethoxy.
  • R 3a is independently selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, tert-butyl group, -CH 2 -OH, -(CH 2 ) 2 -OH, -(CH 2 ) 3 -OH, -(CH 2 ) 4 -OH,
  • R 4 is selected from hydrogen, halogen, -OH, -CN, methyl, ethyl, n-propyl, propyl, fluoromethyl, fluoro Ethyl, fluoropropyl, cyclopropyl, fluorocyclopropyl, Chloromethyl, Chloroethyl, Chloropropyl, Chlorocyclopropyl, Preferably, R 4 is selected from hydrogen, halogen, methyl, ethyl, -CH 2 -CF 3 , cyclopropyl,
  • the compound of formula III is selected from any of the following compounds:
  • the present invention provides intermediates M-1 and M-2:
  • R M is selected from halogen
  • L 1 , R 1 , R 2 , R 4 have the above-mentioned definitions.
  • L 1 is selected from -O-, -S-, -N(R L1 )- and a single bond;
  • R L1 is selected from hydrogen and C 1 -C 6 alkyl;
  • R 1 is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)- (C 3 -C 6 cycloalkyl) and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkenyl);
  • the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkyl) and -(C 1 -C 6 alkylene)-(C 3 -C 6 cycloalkenyl) is optionally substituted with one or more R 1a selected from halogen; when R 1a is more than one, the said R 1a is the same or different;
  • R L1 and R 1 can be combined with L to form a 3- to 6-membered heterocycloalkyl optionally substituted with one or more R L1 ; when R L1 When there are more than one, the R L1 are the same or different; the 3- to 6-membered heterocycloalkyl, wherein 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S and N, and the rest are carbon atom;
  • R 2 is selected from unsubstituted or R 2a -substituted C 6 -C 10 aryl, unsubstituted or R 2a substituted 5- to 10-membered heteroaryl; when R 2a is poly When one, the R 2a are the same or different; the 5- to 10-membered heteroaryl group, wherein 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S and N, and the rest are carbon atoms;
  • R A and R B are each independently selected from H, -CN, -OH, oxo, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 6 alkoxy, C 2 - C 6 alkenyl, C 2 -C 6 alkynyl, -NH 2 , -S(O) 0-2 -(C 1 -C 6 alkyl), -S(O) 0-2 -(C 6 -C 10 aryl), -CO-(C 1 -C 6 alkyl), -CO-(C 3 -C 14 cycloalkyl), -C 3 -C 14 cycloalkyl, -(C 1 -C 6 alkylene) alkyl)-(C 3 -C 14 cycloalkyl), C 6 -C 10 aryl, 3- to 14-membered heterocycloalkyl, 5- to 10-membered heteroaryl; the 3- to 14-membered hetero
  • R A and R B C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 Alkynyl, -NH 2 , -S(O) 0-2 -(C 1 -C 6 alkyl), -S(O) 0-2 -(C 6 -C 10 aryl), -CO-(C 1 -C 6 alkyl), -CO-(C 3 -C 14 cycloalkyl), -C 3 -C 14 cycloalkyl, -(C 1 -C 6 alkylene)-(C 3 -C 14 cycloalkyl), C 6 -C 10 aryl, 3- to 14-membered heterocycloalkyl, and 5- to 10-membered heteroaryl groups are each independently and optionally substituted with one or more R Aa ; when R Aa is multiple , the R Aa are
  • the R Aa are each independently selected from -OH, deuterium, halogen, -N(R An ) 2 , wherein each R An is independently selected from H, C 1 -C 6 alkyl, C 2 -C 6 alkene base, C 2 -C 6 alkynyl, C 6 -C 10 aryl, 3- to 14-membered heterocycloalkyl, -(C 1 -C 6 alkylene)-(3- to 14-membered heterocycloalkyl), and 5- to 10-membered heteroaryl, -NHC(O)-(OC 1 -C 6 alkyl), -NO 2 , -CN, oxo, -C(O)OH, -C(O)-O- (C 1 -C 6 alkyl), -(C 1 -C 6 alkylene)-(C 1 -C 6 -alkoxy), -C(O)NH 2 , -C(O)-(C 1 -C 6
  • Each alkyl, alkenyl, aryl, and heterocycloalkyl in said R Aa is each independently and optionally substituted with one or more R Abs each independently selected from -OH, -O -(C 1 -C 6 alkyl), halogen, -NH 2 , -(C 1 -C 6 alkylene)-NH 2 , -COOH, -CN and oxo;
  • RM is selected from fluorine, chlorine, bromine, and iodine; preferably, RM is selected from fluorine, chlorine, and bromine.
  • L 1 is selected from -O-, -S-, -N(R L1 )- or single bond;
  • R L1 is selected from hydrogen, C 1 - C3 alkyl.
  • R L1 is selected from hydrogen, methyl, ethyl.
  • R 1 is selected from C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene base)-(C 3 -C 6 cycloalkyl); the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl and -(C 1 -C 6 alkylene)-(C 3 -C 6cycloalkyl ) is optionally substituted with one or more R 1a selected from halogen.
  • R 1 is selected from methyl, ethyl, propyl, fluoromethyl, fluoroethyl, fluoropropyl, cyclopropyl, Preferably, R 1 is selected from methyl, ethyl, -CH 2 -CF 3 , cyclopropyl,
  • R 2 is selected from unsubstituted or substituted phenyl with R 2a and unsubstituted or substituted by R 2a 5- to 10-membered heteroaryl , one of the ring atoms of the 5- to 10-membered heteroaryl group is N; preferably, R 2 is selected from unsubstituted or substituted phenyl by R 2a , unsubstituted or substituted by R 2a pyridyl, unsubstituted or Pyrimidyl substituted with R 2a .
  • R 2a is independently selected from -RA , -OR A , halogen and -CN.
  • R A is independently selected from H, -OH, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -CO -(C 1 -C 6 alkyl), -(C 1 -C 6 alkylene)-(C 3 -C 14 cycloalkyl), said C 1 -C 6 alkyl, C 3 -C 6 ring Alkyl, -CO-(C 1 -C 6 alkyl), -(C 1 -C 6 alkylene)-(C 3 -C 14 cycloalkyl) each independently and optionally by one or more R Aa substituted.
  • R Aa is independently selected from -OH, halogen, -N(R An ) 2 , wherein each R An is independently selected from H, methyl, ethyl, isopropyl, n-propyl.
  • R 2 is selected from phenyl substituted by R 2a and pyridyl substituted by R 2a
  • R 2a is independently selected from fluorine, chlorine , bromine, -RA optionally substituted with one or more R Aa
  • -OR A optionally substituted with one or more R Aa independently selected from substituted with one or more R Aa C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl substituted with one or more R Aa , each R Aa independently selected from -OH, fluoro, chloro, bromo; according to certain preferences of the present invention
  • R 2a is independently selected from - fluoro, chloro, bromo, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, methoxy, ethoxy, n-propoxy, isopropoxy base, Fluoromethoxy; more preferably,
  • the intermediate M-1 is selected from any of the following compounds:
  • the intermediate M-2 is selected from any of the following compounds:
  • the present invention provides a method for preparing the compound of formula III, comprising:
  • R 1 , R 2 , R 3 , R 4 , and L 1 have the above-mentioned definitions, and RM is selected from fluorine, chlorine, bromine, and iodine; preferably, RM is selected from fluorine, chlorine, and bromine.
  • the halogenation reagent is selected from N-chlorosuccinimide and N-bromosuccinimide.
  • step (2) under the protection of inert gas, the intermediate M-2 is mixed with The palladium catalyst and the base are contacted to obtain the compound represented by formula III.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the compound represented by formula III, its tautomer, stereoisomer, hydrate, solvate, pharmacy at least one of the above acceptable salts or prodrugs.
  • the pharmaceutical composition further includes one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition may further contain one or more additional therapeutic agents.
  • the present invention provides the compound represented by formula III, its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug or said Use of the pharmaceutical composition in the preparation of a medicament for treating or preventing MAT2A-related diseases, the present invention also provides any compound described in the first aspect of the present invention or the pharmaceutical composition described in the fourth aspect of the present invention. Or medicinal use to prevent MAT2A-related diseases.
  • the medicament is for treating or preventing cancer.
  • the cancer is an MTAP-deficient cancer.
  • the cancer is selected from the group consisting of mesothelioma, neuroblastoma, rectal cancer, colon cancer, familiar adenomatous polyposis and hereditary nonpolyposis colorectal cancer, esophageal cancer, lip Cancer, laryngeal cancer, hypopharyngeal cancer, tongue cancer, salivary gland cancer, gastric cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, kidney cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, endometrial cancer, intrauterine cancer Membranous cancer, choriocarcinoma, pancreatic cancer, prostate cancer, bladder cancer, testicular cancer, breast cancer, urinary cancer, melanoma, brain tumor, lymphoma, head and neck cancer, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL) ), acute myeloid leukemia (AML), chronic myeloid leukemia (C ALL), chronic lymphoblastic le
  • the present invention also provides a method for MAT2A-related diseases, comprising administering to a patient a preventive or therapeutically effective amount of the compound represented by formula (I), its tautomer, stereoisomer, hydrate, solvate, pharmaceutically At least one of an acceptable salt or prodrug, or a pharmaceutical composition of the above.
  • the patient is a mammal, preferably a human.
  • the present invention also provides at least one of the compound represented by formula (I), its tautomer, stereoisomer, hydrate, solvate, pharmaceutically acceptable salt or prodrug, or a pharmaceutical combination thereof
  • the application of the drug in the treatment or prevention of MAT2A-related diseases is not limited to, butyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N-phenyl-N
  • the compounds and/or compositions thereof of the present invention can effectively inhibit the activity of MAT2A enzyme, have a good inhibitory effect on the proliferation of HCT116 MTAP-/- cells and the level of SAM in cells, and have better pharmacological effects.
  • Kinetic properties It has broad application prospects in preparing medicines for treating MAT2A-related diseases.
  • groups and their substituents can be selected by those skilled in the art to provide stable moieties and compounds.
  • substituents When substituents are described by conventional chemical formulae written from left to right, the substituents also include the chemically equivalent substituents obtained when the structural formula is written from right to left. For example, CH2O is equivalent to OCH2 .
  • a number from 1 to 10 should be understood as not only reciting each integer of 1, 2, 3, 4, 5, 6, 7, 8, 9 and 10, but also reciting at least that each integer is respectively associated with Sum of 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms that, within the scope of sound medical judgment, are suitable for use in contact with human and animal tissue without more toxicity, irritation, allergic reactions or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts or “pharmaceutically acceptable salts thereof” refers to pharmaceutically acceptable salts of non-toxic acids or bases, including salts of inorganic acids and bases, organic acids and bases.
  • salts are also contemplated by the present invention. They may serve as intermediates in the purification of compounds or in the preparation of other pharmaceutically acceptable salts or may be used in the identification, characterization or purification of the compounds of the present invention.
  • stereoisomer refers to isomers that result from different arrangements of atoms in a molecule in space.
  • Stereochemical definitions and conventions used herein are generally in accordance with S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry of Organic Compounds”, defined by John Wiley & Sons, Inc., New York, 1994.
  • the compounds of the present invention may contain asymmetric centers or chiral centers and therefore exist in different stereoisomeric forms.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often referred to as a mixture of enantiomers.
  • a 50:50 mixture of enantiomers is called a racemic mixture or racemate, which can occur when there is no stereoselectivity or stereospecificity in a chemical reaction or method Spin body.
  • the compounds of the present invention may exist as one of the possible isomers or as a mixture thereof, for example, as pure optical isomers, or as mixtures of isomers, such as racemic and non-isomeric isomers.
  • Optically active (R)- or (S)-isomers can be prepared using chiral synthons or chiral preparations, or resolved using conventional techniques.
  • the substituent may be of E or Z configuration; if the compound contains a disubstituted cycloalkyl, the substituent of the cycloalkyl may be cis or trans (cis- or trans-) structure.
  • Compounds of the present invention containing asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Resolution of racemic mixtures of compounds can be carried out by any of a number of methods known in the art. Exemplary methods include fractional recrystallization using chiral resolving acids, which are optically active salt-forming organic acids. Suitable resolving agents for the fractional recrystallization process are, for example, optically active acids such as tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or various optically active camphorsulfonic acids such as ⁇ - D and L forms of camphorsulfonic acid.
  • optically active acids such as tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or various optically active camphorsulfonic acids such as ⁇ - D and L forms of camphorsulfonic
  • resolving agents suitable for fractional crystallization methods include ⁇ -methyl-benzylamine in stereoisomerically pure form (eg, S and R forms or diastereomerically pure form), 2-phenylglycinol, Norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, etc.
  • Resolution of the racemic mixture can also be performed by elution on a column packed with an optically active resolving agent (eg, dinitrobenzoylphenylglycine). It can be carried out by high performance liquid chromatography (HPLC) or supercritical fluid chromatography (SFC).
  • tautomer refers to an isomer of a functional group resulting from the rapid movement of an atom in two positions in a molecule.
  • the compounds of the present invention may exhibit tautomerism.
  • Tautomeric compounds can exist as two or more interconvertible species.
  • Proton tautomers arise from the migration of covalently bonded hydrogen atoms between two atoms.
  • Tautomers generally exist in equilibrium, and attempts to separate individual tautomers usually result in a mixture whose physicochemical properties are consistent with a mixture of compounds. The position of equilibrium depends on the chemical properties within the molecule.
  • the ketone form predominates; in phenols, the enol form predominates.
  • the present invention encompasses all tautomeric forms of the compounds.
  • composition means a mixture of one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, such as a physiologically/pharmaceutically acceptable carrier and excipients.
  • the purpose of a pharmaceutical composition is to facilitate the administration of a compound to an organism.
  • solvate means that a compound of the present invention or a salt thereof includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces, and when the solvent is water, it is a hydrate.
  • prodrug refers to a compound of the invention that can be converted under physiological conditions or by solvolysis to a biologically active compound.
  • the prodrugs of the present invention are prepared by modifying functional groups in the compounds, which modifications can be removed by conventional procedures or in vivo to yield the parent compounds.
  • Prodrugs include compounds formed by connecting a hydroxyl or amino group in the compounds of the present invention to any group. When the prodrugs of the compounds of the present invention are administered to mammalian individuals, the prodrugs are cleaved to form free hydroxyl, free the amino group.
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute the compound.
  • compounds can be labeled with radioisotopes, such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C). All transformations of the isotopic composition of the compounds of the present invention, whether radioactive or not, are included within the scope of the present invention.
  • excipient refers to a pharmaceutically acceptable inert ingredient.
  • classes of the term “excipient” include, without limitation, binders, disintegrants, lubricants, glidants, stabilizers, fillers, diluents, and the like.
  • C 1 -C 6 alkyl is understood to mean a straight-chain or branched saturated monovalent hydrocarbon radical having 1, 2, 3, 4, 5 or 6 carbon atoms and a branched chain having 3 to 6 carbon atoms. Saturated monovalent hydrocarbon group.
  • the alkyl group is, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl , 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-dimethylbutyl or 1,2-dimethylbutyl, etc.
  • the groups have 1, 2 or 3 carbon atoms (" C1 -C3 alkyl”), eg methyl, ethyl, n-propyl or isopropyl.
  • C1 -C3 alkyl 1, 2 or 3 carbon atoms
  • alkyl can include “alkylene” groups.
  • alkylene should be understood to mean a straight-chain saturated divalent hydrocarbon radical having 1 to 6 carbon atoms or a branched saturated divalent hydrocarbon radical having 3 to 6 carbon atoms, unless otherwise specified, such as methylene, Ethylene, propylene, 1-methylpropylene, butylene, etc.
  • C 2 -C 6 alkenyl is understood to mean a straight-chain monovalent hydrocarbon group containing one or more carbon-carbon double bonds, having 2, 3, 4, 5 or 6 carbon atoms or a A branched monovalent hydrocarbon group having 3 to 6 carbon atoms of a double bond such as an alkenyl group.
  • C 2 -C 6 alkynyl is to be understood as meaning a straight-chain, branched or cyclic hydrocarbon group containing 2 to 6 carbon atoms and at least one carbon-carbon triple bond, such as ethynyl, propynyl, butynyl and 3-methylbutynyl, etc.
  • C3 - C6cycloalkyl is understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring having 3 to 6 carbon atoms, including fused or bridged polycyclic ring systems. Such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
  • C 1 -C 6 alkoxy is to be understood as -O-(C 1 -6 alkyl), wherein “C 1 -6 alkyl” has the above definition.
  • halo or halogen refers to fluorine, chlorine, bromine or iodine, preferably fluorine or chlorine.
  • haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoropropyl, and heptachloropropyl.
  • C6 - C10 -aryl refers to a monovalent monocyclic or bicyclic aromatic hydrocarbon group of 6 to 10 ring atoms, such as phenyl or naphthyl.
  • 5- to 10-membered heteroaryl refers to a monovalent monocyclic or bicyclic aromatic group having 5 to 10 ring atoms comprising one or two aromatic rings, one or more of which (in certain implementations) In the example, 1, 2, 3 or 4) ring atoms are heteroatoms independently selected from O, S and N, and the remainder are carbon atoms.
  • bicyclic or “fused” means that two rings are joined together by a bond between two atoms (eg, naphthalene), and are joined together by a series of atoms to form a bridge (eg, quinuclidine) ) or the individual atoms together form a spiro compound (eg, 1,4-dioxa-8-aza-spiro[4.5]decane and N,3,3-dimethyl-1,5-dioxa spiro[5.5]undecan-9-yl).
  • a spiro compound eg, 1,4-dioxa-8-aza-spiro[4.5]decane and N,3,3-dimethyl-1,5-dioxa spiro[5.5]undecan-9-yl.
  • 3 to 6 membered heterocycloalkyl refers to a saturated monocyclic group of 3 to 6 ring atoms, wherein 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S and N, The remainder are carbon atoms; in addition, one or two ring carbon atoms in the heterocyclyl ring may be optionally substituted with a -CO- group.
  • patient refers to any animal including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses or primates, most preferably humans.
  • terapéuticaally effective amount refers to the amount of active compound or drug that a researcher, veterinarian, physician or other clinician is seeking to elicit a biological or medical response in a tissue, system, animal, individual or human, and includes one of the following or more of: (1) Prevention of disease: eg, prevention of disease, disorder or condition in individuals susceptible to a disease, disorder or condition but not yet experiencing or developing disease pathology or symptoms. (2) Inhibiting a disease: eg, inhibiting a disease, disorder or condition (ie preventing further progression of the pathology and/or condition) in an individual who is experiencing or developing the pathology or symptom of the disease, disorder or condition. (3) Alleviating disease: eg, alleviating a disease, disorder or condition (ie, reversing the pathology and/or symptoms) in an individual who is experiencing or experiencing the pathology or symptoms of the disease, disorder or condition.
  • the compounds of the present invention are identified by nuclear magnetic resonance (NMR) and/or mass spectrometry (MS).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the units of NMR shifts are 10-6 (ppm).
  • the solvents for NMR measurement are deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and the internal standard is tetramethylsilane (TMS).
  • DIPEA can also be written as DIEA, diisopropylethylamine, that is, N,N-diisopropylethylamine
  • NCS N-Chlorosuccinimide, also known as chlorosuccinimide
  • NBS N-bromosuccinimide
  • EC 80 concentration for 80% of maximal effect, the concentration that can cause 80% of the maximal effect
  • IC 50 half inhibitory concentration, refers to the concentration at which half of the maximum inhibitory effect is achieved
  • N equivalent concentration, for example, 2N hydrochloric acid means 2mol/L hydrochloric acid solution
  • the first step Synthesis of 2-chloro-N-(4-(difluoromethoxy)phenyl)-5-nitropyrimidin-4-amine (I-1B)
  • the second step synthesis of N 2 -cyclopropyl-N 4 -(4-(difluoromethoxy)phenyl)-5-nitropyrimidine-2,4-diamine (I-1C)
  • the third step synthesis of N 2 -cyclopropyl-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-2,4,5-triamine (I-1D)
  • the fourth step the synthesis of 2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one (I-1E)
  • N 2 -cyclopropyl-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-2,4,5-triamine (1.00 g, 3.25 mmol), ethyl glyoxylate (431.8 mg) , 4.23 mmol) and acetic acid (195.4 mg, 3.25 mmol) were added to ethanol (20 mL), the nitrogen was replaced, and the reaction was heated to 80° C. for 3 hours.
  • the reaction solution was cooled to room temperature, water (30 mL) was added, extracted with ethyl acetate (90 mL ⁇ 3), the organic phases were combined, washed with saturated brine (90 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated.
  • the fifth step the synthesis of 6-chloro-2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one (I-1F)
  • Step 6 2-(Cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(2-methyl-2H-indazol-5-yl)pteridine-7 (8H)-ketone (target compound I-1)
  • 6-Chloro-2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one (380 mg, 1.00 mmol), 2 -methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)-2H-indazole (387.4 mg, 1.50 mmol), 1, 1'-Bis(diphenylphosphonium)ferrocene palladium chloride (73.2 mg, 100.0 ⁇ mol) and potassium carbonate (414.8 mg, 3.00 mmol) were dissolved in 1,4-dioxane (10 mL) and water (1 mL) ), nitrogen was replaced, and the temperature was raised to 90° C.
  • the synthetic route is as follows:
  • the first step Synthesis of 2-cyclopropoxy-N-(4-(difluoromethoxy)phenyl)-5-nitropyrimidin-4-amine
  • the second step Synthesis of 2-cyclopropoxy-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-4,5-diamine
  • reaction solution was filtered, washed with methanol (30.0 mL*2), and the reaction solution was concentrated to obtain 2-cyclopropoxy-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-4 , 5-diamine (520 mg yellow solid). used directly in the next step.
  • the third step Synthesis of 2-cyclopropoxy-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one
  • reaction mixture was diluted with water (20.0 mL), then extracted with ethyl acetate (20.0 mL*3), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain compound 2-cyclopropoxy-8 -(4-(Difluoromethoxy)phenyl)pteridine-7(8H)-one (500 mg, 1.44 mmol, yellow solid, 89.0% yield).
  • the fourth step the synthesis of 6-chloro-2-cyclopropoxy-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one
  • the synthetic route is as follows:
  • the first step synthesis of N-(4-(difluoromethoxy)phenyl)-5-nitro-2-(2,2,2-trifluoroethoxy)pyrimidin-4-amine
  • the second step synthesis of N 4 -(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pyrimidine-4,5-diamine
  • N-(4-(difluoromethoxy)phenyl)-5-nitro-2-(2,2,2-trifluoroethoxy)pyrimidin-4-amine (900 mg, 2.37 mmol) in methanol (20.0 mL) solution was added platinum vanadium carbon (501 mg, 192 ⁇ mol, content of 10%), then the reaction system was replaced with hydrogen 3 times, and the reaction was stirred at 15° C. for 5 hours under a hydrogen pressure of 15 psi.
  • the third step Synthesis of 8-(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pteridine-7(8H)-one
  • N 4 -(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pyrimidine-4,5-diamine 500 mg, 1.43 mmol
  • ethanol 5.00 mL
  • ethyl glyoxylate 379 mg, 1.86 mmol, 50.0% concentration
  • glacial acetic acid 85.7 mg, 1.43 mmol
  • reaction mixture was diluted with water (30.0 mL), then extracted with ethyl acetate (30.0 mL*3), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain compound 8-(4-(difluoro) Methoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pteridine-7(8H)-one (550 mg yellow solid). used directly in the next step.
  • the fourth step Synthesis of 6-chloro-8-(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pteridine-7(8H)-one
  • N,N to 8-(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pteridine-7(8H)-one 250 mg, 644 ⁇ mol
  • N-chlorosuccinimide 112 mg, 837 ⁇ mol
  • 6-Chloro-8-(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pteridine-7(8H)-one (260mg yellow oily compound) . used directly in the next step.
  • the fifth step 8-(4-(difluoromethoxy)phenyl)-6-(2-methyl-2H-indazol-5-yl)-2-(2,2,2-trifluoroethyl Synthesis of Oxy)pteridine-7(8H)-one (I-3)
  • 6-chloro-8-(4-(difluoromethoxy)phenyl)-2-(2,2,2-trifluoroethoxy)pteridine-7(8H)-one (220 mg, 520 ⁇ mol) in 1,4-dioxane (3.00 mL) and water (500 ⁇ L) was added 2-methyl-5-(4,4,5,5-tetramethyl-1,3) , 2-dioxaborol-2-yl)-2H-indazole (202 mg, 781 ⁇ mol), potassium carbonate (216 mg, 1.56 mmol) and 1,1-bis(diphenylphosphonium)ferrocene Palladium chloride (19.0 mg, 26.0 ⁇ mol), the reaction was stirred at 70° C.
  • the synthetic route is as follows:
  • the first step Synthesis of 2-chloro-N-(4-(difluoromethoxy)phenyl)-5-nitropyrimidin-4-amine
  • the second step N 4 -(4-(difluoromethoxy)phenyl)-5-nitro-N 2 -(2,2,2-trifluoroethyl)pyrimidine-2,4-diamine synthesis
  • reaction solution was cooled to room temperature, poured into water (50 mL), filtered, and the solid was collected and dried to obtain N 4 -(4-(difluoromethoxy)phenyl)-5-nitro-N 2 -(2, 2,2-Trifluoroethyl)pyrimidine-2,4-diamine (1.00 g, yellow solid, 83.5% yield).
  • the third step synthesis of N 4 -(4-(difluoromethoxy)phenyl)-N 2 -(2,2,2-trifluoroethyl)pyrimidine-2,4,5-triamine
  • N 4 -(4-(difluoromethoxy)phenyl)-5-nitro-N 2 -(2,2,2-trifluoroethyl)pyrimidine-2,4-diamine (1.00 g, 2.64 mmol) and palladium on carbon (0.10 g, content of 10%) were added to methanol (10 mL), replaced with hydrogen, heated to 30° C., and the reaction was stirred at 50 psi for 3 hours.
  • the fourth step synthesis of 8-(4-(difluoromethoxy)phenyl)-2-((2,2,2-trifluoroethyl)amino)pteridine-7(8H)-one
  • N 4 -(4-(difluoromethoxy)phenyl)-N 2 -(2,2,2-trifluoroethyl)pyrimidine-2,4,5-triamine 700 mg, 2.00 mmol
  • Ethyl glyoxylate 613.8 mg, 3.01 mmol
  • acetic acid 120.4 mg, 2.00 mmol
  • Step 6 8-(4-(Difluoromethoxy)phenyl)-6-(2-methyl-2H-indazol-5-yl)-2-((2,2,2-trifluoro Ethyl)amino)pteridine-7(8H)-one (Compound 1-4)
  • 6-Chloro-8-(4-(difluoromethoxy)phenyl)-2-((2,2,2-trifluoroethyl)amino)pteridine-7(8H)- Ketone 100 mg, 237.1 ⁇ mol
  • 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborol-2-yl)-2H-indone azole 91.8 mg, 355.7 ⁇ mol
  • 1,1-bis(diphenylphosphonium)ferrocene palladium dichloride 17.4 mg, 23.7 ⁇ mol
  • potassium carbonate 98.3 mg, 711.4 ⁇ mol
  • the synthetic route is as follows:
  • the first step Synthesis of 2-chloro-N-(4-(difluoromethoxy)phenyl)-5-nitropyrimidin-4-amine
  • the second step synthesis of N 2 -(cyclopropylmethyl)-N 4 -(4-(difluoromethoxy) phenyl)-5-nitropyrimidine-2,4-diamine
  • reaction solution was cooled to room temperature, poured into water (50 mL), filtered, and the solid was collected and dried to obtain N 2 -(cyclopropylmethyl)-N 4 -(4-(difluoromethoxy)phenyl)- 5-Nitropyrimidine-2,4-diamine (1.00 g, yellow solid, 90.1% yield).
  • the third step synthesis of N 2 -(cyclopropylmethyl)-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-2,4,5-triamine
  • the fourth step the synthesis of 2-((cyclopropylmethyl)amino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one
  • N 2 -(cyclopropylmethyl)-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-2,4,5-triamine 600 mg, 1.87 mmol
  • ethyl glyoxylate 571.9 mg, 2.80 mmol
  • acetic acid 112.1 mg, 1.87 mmol
  • the reaction solution was cooled to room temperature, water (20 mL) was added, extracted with ethyl acetate (20 mL*3), the organic phases were combined, washed with saturated brine (30 mL*2), dried over anhydrous sodium sulfate, filtered and concentrated.
  • the fifth step the synthesis of 6-chloro-2-((cyclopropylmethyl)amino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one
  • Step 6 2-((Cyclopropylmethyl)amino)-8-(4-(difluoromethoxy)phenyl)-6-(2-methyl-2H-indazol-5-yl) Pteridin-7(8H)-one (Compound I-5)
  • 6-Chloro-2-((cyclopropylmethyl)amino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one (100 mg, 253.9 ⁇ mol), 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborol-2-yl)-2H-indazole (98.3 mg, 380.9 ⁇ mol), 1,1-bis(diphenylphosphonium)ferrocene palladium dichloride (18.6 mg, 25.4 ⁇ mol) and potassium carbonate (105.3 mg, 761.9 ⁇ mol) were dissolved in dioxane (1 mL) and water (0.2 mL), nitrogen was replaced, and the temperature was raised to 60° C.
  • the synthetic route is as follows:
  • the first step the synthesis of 2-chloro-N-(4-chlorophenyl)-5-nitropyrimidin-4-amine
  • reaction mixture was diluted with water (50.0 mL), then extracted with ethyl acetate (50.0 mL*3), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain compound 2-chloro-N-(4 -Chlorophenyl)-5-nitropyrimidin-4-amine (6.50 g, 22.8 mmol, yellow solid, 88.5% yield).
  • the second step the synthesis of N 4 -(4-chlorophenyl)-N 2 -cyclopropyl-5-nitropyrimidine-2,4-diamine
  • the third step the synthesis of N 4 -(4-chlorophenyl)-N 2 -cyclopropylpyrimidine-2,4,5-triamine
  • the fourth step the synthesis of 8-(4-chlorophenyl)-2-(cyclopropylamino)pteridine-7(8H)-one
  • the fifth step the synthesis of 6-chloro-8-(4-chlorophenyl)-2-(cyclopropylamino)pteridine-7(8H)-one
  • reaction mixture was diluted with water (20.0 mL), a solid was precipitated, the reaction solution was filtered, the solid was dried, the residue was slurried with methanol (10.0 mL), and then separated and purified by high performance liquid chromatography.
  • the separation method was (column).
  • the synthetic route is as follows:
  • the third step Synthesis of 2-chloro-N-(6-(difluoromethoxy)pyridin-3-yl)-5-nitropyrimidin-4-amine
  • reaction mixture was diluted with water (50.0 mL), then extracted with ethyl acetate (50.0 mL*3), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain compound 2-chloro-N-(6 -(difluoromethoxy)pyridin-3-yl)-5-nitropyrimidin-4-amine (4.80 g, crude brown solid). used directly in the next step.
  • the fourth step synthesis of N 2 -cyclopropyl-N 4 -(6-(difluoromethoxy)pyridin-3-yl)-5-nitropyrimidine-2,4-diamine
  • reaction mixture was diluted with water (150 mL), a solid was precipitated, the reaction solution was filtered, the solid was dried, and then slurried with methyl tert-butyl ether (30.0 mL) to obtain N 2 -cyclopropyl-N 4 -( 6-(Difluoromethoxy)pyridin-3-yl)-5-nitropyrimidine-2,4-diamine (2.30 g, 6.80 mmol, yellow solid, 86.4% yield).
  • the fifth step synthesis of N 2 -cyclopropyl-N 4 -(6-(difluoromethoxy)pyridin-3-yl)pyrimidine-2,4,5-triamine
  • the sixth step the synthesis of 2-(cyclopropylamino)-8-(6-(difluoromethoxy)pyridin-3-yl)pteridine-7(8H)-one
  • the seventh step the synthesis of 6-chloro-2-(cyclopropylamino)-8-(6-(difluoromethoxy)pyridin-3-yl)pteridine-7(8H)-one
  • N-chlorosuccinimide 174 mg, 1.30 mmol was added to the N-dimethylformamide (2.00 mL) solution, followed by stirring at 60°C for 4 hours.
  • reaction mixture was diluted with water (20.0 mL), then extracted with ethyl acetate (20.0 mL*3), the organic layers were combined, washed with water (20.0 mL*3), dried over anhydrous sodium sulfate, filtered, and concentrated to give compound 6- Chloro-2-(cyclopropylamino)-8-(6-(difluoromethoxy)pyridin-3-yl)pteridine-7(8H)-one (330 mg yellow solid). used directly in the next step.
  • the eighth step 2-(cyclopropylamino)-8-(6-(difluoromethoxy)pyridin-3-yl)-6-(2-methyl-2H-indazol-5-yl)pteroid Synthesis of Pyridin-7(8H)-one (I-7)
  • 6-chloro-2-(cyclopropylamino)-8-(6-(difluoromethoxy)pyridin-3-yl)pteridine-7(8H)-one 330 mg, 867 ⁇ mol
  • dioxane 5.00 mL
  • water 1.00 mL
  • Pentan-2-yl)-2H-indazole 336 mg, 1.30 mmol
  • 1,1-bis(diphenylphosphonium)ferrocene palladium dichloride (63.4 mg, 86.7 ⁇ mol) and potassium carbonate (359 mg, 2.60 mmol) and the reaction was stirred at 70°C for 2 hours.
  • reaction mixture was diluted with water (30.0 mL), a solid was precipitated, the reaction solution was filtered, the solid was dried, slurried with ethyl acetate (10.0 mL), and then separated and purified by high performance liquid chromatography.
  • the separation method was (column).
  • the synthetic route is as follows:
  • the first step the synthesis of 2-chloro-N-(4-cyclopropylphenyl)-5-nitropyrimidin-4-amine
  • the second step the synthesis of N 2 -cyclopropyl-N 4 -(4-cyclopropylphenyl)-5-nitropyrimidine-2,4-diamine
  • the third step synthesis of N 2 -cyclopropyl-N 4 -(4-cyclopropylphenyl)pyrimidine-2,4,5-triamine
  • N 2 -cyclopropyl-N 4 -(4-cyclopropylphenyl)-5-nitropyrimidine-2,4-diamine (1.60 g, 5.14 mmol) and platinum vanadium carbon (160 mg, 10.0% content ) was added to methanol (16.0 mL), replaced with hydrogen, heated to 30°C, and stirred at 30°C, 50 psi for 3 hours.
  • the reaction solution was cooled to room temperature and filtered, and the filtrate was spin-dried. Obtained N2 -cyclopropyl-N4-( 4 -cyclopropylphenyl)pyrimidine-2,4,5-triamine (1.35 g, tan solid, 93.3% yield).
  • the fourth step the synthesis of 2-(cyclopropylamino)-8-(4-cyclopropylphenyl)pteridine-7(8H)-one
  • N 2 -cyclopropyl-N 4 -(4-cyclopropylphenyl)pyrimidine-2,4,5-triamine (1.20g, 4.27mmol), ethyl glyoxylate (1.13g, 5.54mmol) and acetic acid (256 mg, 4.27 mmol) were added to ethanol (12.0 mL), nitrogen was replaced, the mixture was heated to 80°C, and stirred at 80°C for 3 hours.
  • the reaction solution was cooled to room temperature, water (20.0 mL) was added, extracted with ethyl acetate (45.0 mL*3), the organic phases were combined, washed with saturated brine (30.0 mL*2), dried over anhydrous sodium sulfate, filtered, concentrate.
  • the fifth step the synthesis of 6-chloro-2-(cyclopropylamino)-8-(4-cyclopropylphenyl)pteridine-7(8H)-one
  • 6-Chloro-2-(cyclopropylamino)-8-(4-cyclopropylphenyl)pteridine-7(8H)-one 160 mg, 452 ⁇ mol
  • 2-methyl-5-(4, 4,5,5-Tetramethyl-1,3,2-dioxaborolane-2-yl)indazole 140 mg, 542 ⁇ mol
  • 1,1-bis(diphenylphosphorus)ferrocene Palladium dichloride (18.4 mg, 22.6 ⁇ mol) and potassium carbonate (187 mg, 1.36 mmol) were dissolved in dioxane (2.00 mL) and water (0.20 mL). under stirring for 2 hours.
  • the synthetic route is as follows:
  • the synthetic route of compound I-10 refers to the synthetic method of I-1 to obtain 2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(2-methylbenzoyl) [d]thiazol-6-yl)pteridine-7(8H)-one (I-10)
  • the synthetic route is as follows:
  • the first step Synthesis of 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-2H-indazole
  • the synthetic route is as follows:
  • the first step Synthesis of 2-(cyclopropylmethoxy)-N-(4-(difluoromethoxy)phenyl)-5-nitropyrimidin-4-amine
  • the second step Synthesis of 2-(cyclopropylmethoxy)-N 4 -(4-(difluoromethoxy)phenyl)pyrimidine-4,5-diamine
  • the third step Synthesis of 2-(cyclopropylmethoxy)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one
  • reaction mixture was diluted with water (20.0 mL), then extracted with ethyl acetate (20.0 mL*3), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain compound 2-(cyclopropylmethoxy) yl)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one (450 mg, 1.25 mmol, yellow solid, 89.5% yield).
  • the fourth step the synthesis of 6-chloro-2-(cyclopropylmethoxy)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one
  • N,N-Dimethylmethane to 2-(cyclopropylmethoxy)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one 350 mg, 971 ⁇ mol
  • N-chlorosuccinimide 195 mg, 1.46 mmol
  • reaction mixture was diluted with water (10.0 mL), then extracted with ethyl acetate (10.0 mL*3), the organic layers were combined, washed with water (10.0 mL*3), dried over anhydrous sodium sulfate, filtered, concentrated, 6-Chloro-2-(cyclopropylmethoxy)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one (300 mg yellow oily compound) was obtained. used directly in the next step.
  • the synthetic route is as follows:
  • the synthetic route is as follows:
  • the first step Synthesis of 3-amino-2-methyl-2,6-dihydropyrrolo[3,4-c]pyrazole-5(4H)-carboxylic acid tert-butyl ester
  • tert-butyl 3-cyano-4-oxopyrrolidine-1-carboxylate (3.00 g, 14.2 mmol) was dissolved in ethanol (30.0 mL), and methylhydrazine (788 mg, 17.1 mmol) was added and stirred well , and reacted at 85°C for 17 hours. After the reaction, it was directly filtered and concentrated, and the residue was separated and purified by high performance liquid chromatography.
  • the third step Synthesis of 2-methyl-2,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole
  • the synthetic route is as follows:
  • the first step 2-methyl-1-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-1H-indazole Synthesis of -2-yl)propan-2-ol
  • Step 2 2-(Cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(1-(2-hydroxy-2-methylpropyl)-1H-indium Synthesis of oxazol-5-yl)pteridine-7(8H)-one (I-15)
  • the synthetic route is as follows:
  • the first step synthesis of (Z)-3-((dimethylamino)methylene)-4-oxopyrrolidine-1-carboxylic acid tert-butyl ester
  • tert-butyl 3-oxopyrrolidine-1-carboxylate (4.00 g, 21.6 mmol) was dissolved in tetrahydrofuran (10.0 mL), and N,N-dimethylformamide dimethylacetal (7.72 mmol) was added. g, 64.9 mmol), replaced with nitrogen, stirred well, and reacted at 70° C. for 16 hours. After the reaction, it was directly concentrated, and the residue was separated and purified by high performance liquid chromatography.
  • the third step synthesis of 1-methyl-1,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole
  • the synthetic route is as follows:
  • the first step (2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-2,3-dihydro Synthesis of Benzofuran-2-yl)methanol
  • Step 2 2-(Cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(2-(hydroxymethyl)-2-methyl-2,3-di Hydrobenzofuran-5-yl)pteridine-7(8H)-one (I-17)
  • the synthetic route is as follows:
  • the first step the synthesis of 1-(4-bromo-1H-pyrazol-1-yl)-2-methylpropan-2-ol
  • the third step 2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(1-(2-hydroxy-2-methylpropyl)-1H-pyridine Azol-4-yl)pteridine-7(8H)-one (I-18)
  • the synthetic route is as follows:
  • the first step the synthesis of 4-(4-bromo-1H-pyrazol-1-yl)-2-methylbutan-2-ol
  • the third step 2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(1-(3-hydroxy-3-methylbutyl)-1H-pyridine Azol-4-yl)pteridine-7(8H)-one (target compound I-19)
  • 6-chloro-2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)pteridine-7(8H)-one 500 mg, 1.32 mmol
  • 2-Methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborol-2-yl)-1H-pyrazol-1-yl ) butan-2-ol 553 mg, 1.97 mmol
  • 1,1-bis(diphenylphosphonium)ferrocene palladium dichloride 107 mg, 132 ⁇ mol
  • potassium carbonate 364 mg, 2.63 mmol
  • the synthetic route is as follows:
  • the first step synthesis of 1-(5-bromo-2H-indazol-2-yl)propan-2-ol
  • the synthetic route is as follows:
  • the first step the synthesis of methyl 2-(5-bromo-2H-indazol-2-yl) propionate
  • the third step 2-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-2H-indazol-2-yl) Synthesis of Propane-1-ol
  • the synthetic route is as follows:
  • the first step the synthesis of 1-(5-bromo-2H-indazol-2-yl)-2-methylpropan-2-ol
  • the third step 8-(4-(difluoromethoxy)phenyl)-6-(2-(2-hydroxy-2-methylpropyl)-2H-indazol-5-yl)-2- ((2,2,2-Trifluoroethyl)amino)pteridine-7(8H)-one (I-22)
  • the synthetic route is as follows:
  • the first step Synthesis of 5-bromo-2-(2-(methylsulfonyl)ethyl)-2H-indazole
  • the third step 2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(2-(2-(methylsulfonyl)ethyl)-2H-indium Synthesis of oxazol-5-yl)pteridine-7(8H)-one (I-23)
  • the synthetic route is as follows:
  • the first step the synthesis of 2-(5-bromo-2H-indazol-2-yl)-N,N-dimethylacetamide
  • the third step 2-(5-(2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-7-oxo-7,8-dihydropteridine-6 -yl)-2H-indazol-2-yl)-N,N-dimethylacetamide (I-24)
  • N,N-dimethyl-2-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl) -2H-Indazol-2-yl)acetamide 300 mg, 911 ⁇ mol
  • potassium carbonate 504 mg, 3.65 mmol
  • 6-chloro-2-(cyclopropylamino)-8-(4-(difluoromethoxy) yl)phenyl)pteridine-7(8H)-one (242 mg, 638 ⁇ mol)
  • 1,1-bis(diphenylphosphonium)ferrocene palladium dichloride (66.7 mg, 91.1 ⁇ mol) was added to tetrahydrofuran (6.00 mL) and water (1.00 mL), reacted at 25°C for 8 hours under nitrogen protection, the reaction solution was filtered and concentrated to obtain the crude product.
  • the synthetic route is as follows:
  • the first step Synthesis of 5-bromo-1-(3-methylbut-2-en-1-yl)pyridin-2(1H)-one
  • reaction mixture was diluted with water (20.0 mL), then extracted with ethyl acetate (20.0 mL*3), the organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain 1-(3-hydroxy-3- Methylbutyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)pyridin-2(1H)-one (350mg brown oil). used directly in the next step.
  • the fourth step 2-(cyclopropylamino)-8-(4-(difluoromethoxy)phenyl)-6-(1-(3-hydroxy-3-methylbutyl)-6-oxygen Synthesis of Substituted-1,6-dihydropyridin-3-yl)pteridine-7(8H)-one (I-25)
  • the synthetic route is as follows:
  • the first step Synthesis of 4-(2-(5-bromo-2H-indazol-2-yl)ethyl)morpholine
  • BPS Bioscience MAT2A Inhibitor Screening Kit was used to detect the IC 50 of compounds inhibiting MAT2A enzymatic activity.
  • the compounds to be tested were dissolved in DMSO, and all compounds were diluted in DMSO to an initial concentration of 1 mM, 3-fold dilution, and 10 concentration gradients. Transfer 200 nL of the diluted compound to each well of the reaction plate (784075, Greiner) with an Echo 550, seal the plate with a sealing film, centrifuge at 1000 g for 1 minute, and the final concentration of DMSO is 1%.
  • %inhibition 100-(Signal cmpd-Signal Ave_PC )/(Signal Ave_VC -Signal Ave_PC ) ⁇ 100.
  • Signal Ave_PC the fluorescent signal of the positive control well in the reaction plate
  • Signal Ave_VC the fluorescent signal of the negative control well in the reaction plate.
  • the IC50 50% inhibitory concentration of the compound was obtained using the following nonlinear fitting formula:
  • the experimental results show that the compound of the present invention has a good inhibitory activity on MAT2A enzyme, and is better than that of the control compound.
  • Test Example 2 Effects of Compounds on the Proliferation of HCT116 MTAP-/- Cells
  • control compounds and compounds of the present invention were determined according to the following experimental methods.
  • HCT116 MTAP-/- cells were cultured in MCCOYS 5A medium, supplemented with 10% FBS and 1% Penicillin-Streptomycin, and cultured at 37°C and 5% CO 2 . The cells were routinely cultured until the cell saturation was 80%-90%, and the cells were harvested. Resuspend in the corresponding medium to prepare a cell suspension of appropriate density. Transfer 150nL of diluted compounds to 384 cell culture plate with Echo 550; seed the cells into 384 cell culture plate, 400/well, 30uL.
  • the initial concentration of the upper limit of the final concentration of the compound was 20 ⁇ M, which was diluted according to 4-fold gradient, with a total of 10 concentrations, and the starting concentration of the control compound AGI-24512 was 30 ⁇ M, which was diluted according to 3-fold gradient, with a total of 10 concentrations.
  • the cell plate to be tested was placed at room temperature for 30 minutes to equilibrate, 30 ⁇ L of CTG reagent (CelltiterGlo kit) was added to each well, and the chemiluminescence signal value was read with an Envision instrument after being placed in the dark at room temperature for 30 minutes.
  • the inhibitory activity IC 50 of the compound on HCT116 MTAP-/- cell proliferation was calculated by detecting the luminescence value.
  • the experimental results show that the compound of the present invention has a good inhibitory effect on the proliferation of HCT116 MTAP-/- cells, and its inhibitory activity is better than that of the control compound.
  • Test Example 3 Effects of Compounds on SAM Levels in HCT116 MTAP-/- Cells
  • control compounds and compounds of the present invention were determined according to the following experimental methods.
  • HCT116 MTAP-/- cells were cultured in MCCOYS 5A medium, 10% FBS and 1% Penicillin-Streptomycin were added, and cultured at 37°C and 5% CO 2 .
  • the cells were routinely cultured until the cell saturation was 80%-90%, and the cells were harvested.
  • the cells were resuspended in the corresponding medium to prepare a cell suspension of appropriate density, inoculated in a 96-well culture dish for 24 hours, and incubated with the test compound and cells at 37°C and 5% CO 2 for 4 hours.
  • Detection of SAM levels in cells after compound treatment cells were gently washed once in ammonium carbonate buffer (75 mM, pH 7.4), placed on dry ice, and washed with metabolite extraction buffer (containing 50 ng/ml deuterated d3SAM). 80% cold methanol and 20% acetic acid) cleavage. After centrifugation at 3200 rpm for 30 min at 4°C, the supernatant was collected and stored at -80°C until the level of SAM was analyzed by LC/MS.
  • ammonium carbonate buffer 75 mM, pH 7.4
  • metabolite extraction buffer containing 50 ng/ml deuterated d3SAM
  • 80% cold methanol and 20% acetic acid cleavage. After centrifugation at 3200 rpm for 30 min at 4°C, the supernatant was collected and stored at -80°C until the level of SAM was analyzed by LC/MS.
  • the human liver microsomal stability of the control compounds and the compounds of the present invention was determined according to the following experimental method.
  • the liver microsome stability assay of the compounds was performed by in vitro co-incubation of the compounds with human liver microsomes.
  • the compounds to be tested were first formulated as 10 mM stock solutions in DMSO solvent, and then the compounds were diluted to 0.5 mM using acetonitrile.
  • Liver microsomes (Corning) were diluted with PBS to a microsome/buffer solution, and this solution was used to dilute 0.5 mM of the compound to make a working solution with a compound concentration of 1.5 ⁇ M and a liver microsome concentration of 0.75 mg/ml.
  • Test Example 5 Evaluation of Compound Toxicity to Human Normal Hepatocytes
  • control compounds and compounds of the present invention were determined according to the following experimental methods.
  • Human normal hepatocytes LO2 were cultured in DMEM medium containing 10% FBS. When the cells were in good growth state, they were seeded in a 384-well plate at a density of 3000/well and 30 ⁇ L/well. Place in a 37 °C, 5% CO 2 incubator overnight.
  • IC50 values were calculated using Graphpad 5 software.
  • the experimental results show that the compound of the present invention has no inhibitory activity on human normal hepatocytes LO2, indicating that there is no risk of toxicity to human normal hepatocytes.
  • mice pharmacokinetic properties of the control compounds and the compounds of the present invention were determined according to the following experimental methods.
  • mice show that the compounds of the present invention exhibit excellent pharmacokinetic properties in mice.

Landscapes

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

Abstract

一种有效抑制MAT2A的新化合物,其为式III所示化合物,或者式III所示化合物的互变异构体、立体异构体、水合物、溶剂化物、药学可接受的盐或前药:其中,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;R 1、R 2、R 3、R 4的定义如说明书中所述;所述化合物能够有效抑制MAT2A酶活性,具有广阔的应用前景。

Description

嘧啶并吡嗪酮化合物及其用途
本发明要求享有于2021年3月29日向中国国家知识产权局提交的,专利申请号为202110335375.3,名称为“嘧啶并哒嗪酮化合物及其用途”的在先申请的优先权。该在先申请的全文通过引用的方式结合于本发明中。
技术领域
本发明涉及化学及医药领域,具体地,本发明涉及嘧啶并吡嗪酮化合物及其用途。
背景技术
甲硫氨酸腺苷转移酶2A(methionine adenosyltransferase 2A,MAT2A)是MAT家族蛋白成员之一,体内分布广泛,在肝脏的非实质细胞和所有肝外组织中均有表达(Murray B,et al.World J Gastroenterol 2019;25(31):4300-4319.),MAT家族还有MAT1A和MAT2B两个成员。MAT2A抑制剂可降低S-腺苷甲硫氨酸(SAM)水平,故MAT2A抑制剂也称为SAM抑制剂。SAM是细胞中主要的甲基供体,蛋白精氨酸甲基转移酶5(PRMT5)是利用SAM的甲基供体的甲基化酶,SAM在PRMT5通路有重要作用,可影响PRMT5活性,研究表明MAT2A是MTAP缺失型肿瘤的“合成致死”靶点。使用shRNA筛选确定MAT2A和PRMT5是MTAP缺失细胞中的易感基因,代谢学和生化研究揭示了这种合成致死性的机制基础:MTAP酶反应的底物MTA在MTAP缺失的癌症中大量积聚,MTA是PRMT5的一种有效的选择性抑制剂,导致MTAP缺失细胞的PRMT5甲基化活性降低,MAT2A产生PRMT5的底物SAM,MAT2A缺失可选择性地降低MTAP缺失细胞的生长和PRMT5甲基化活性,MAT2A在MTAP缺失的癌症中是选择性必需的(Marjon K,et al.Cell Rep.2016;15(3):574-587)。MAT2A抑制剂为MTAP缺失的肿瘤患者提供了一种新的治疗方法。
MAT2A参与肿瘤干细胞的代谢,肿瘤干细胞需要大量的甲硫氨酸以维持自身组蛋白的甲基化,这对于肿瘤干细胞的生长和致瘤作用至关重要。抑制甲硫氨酸循环上的关键酶MAT2A,可以大幅抑制肿瘤干细胞的生长和肿瘤的形成。并且在人类非小细胞肺癌组织中,MAT2A蛋白有异常高表达。将非小细胞肺癌的肿瘤干细胞移植到小鼠身上,MAT2A抑制剂几乎能完全抑制肿瘤的生长,而化疗药物顺铂几乎没什么效果(Wang Z,et al.Nature medicine,2019,25(5):1- 13.)。
肝癌中MAT1A与MAT2A进行转换。许多研究表明MAT在慢性肝病和肝癌的发生中起着重要作用,正常情况下,胎儿肝脏中主要表达MAT2A,出生后随着生长发育逐渐被MAT1A替代,MAT1A维持肝细胞的分化状态。在正常肝细胞中MAT1A与MAT2A存在动态平衡,共同维持细胞内SAM稳态。在肝细胞癌中,发生MAT1A的表达水平下调和MAT2A的上调,被称为MAT1A:MAT2A转换,肝脏去分化,使SAM生物合成减少,增强肝脏的增殖信号。在人类肝癌中,MAT1A:MAT2A的表达比率与细胞生长和基因组不稳定呈负相关,与肝癌细胞凋亡和整体DNA甲基化直接相关;比率降低是肝癌恶性程度更高、生存率更低的预后标志。有研究表明,使用小干扰RNA沉默MAT2A基因能抑制肝癌细胞的生长并诱导细胞凋亡(Liu Q,et al.Hepatol Res.2007;37(5):376-388.)。
2018年全球新增1810万例癌症病例,死亡人数达960万。大约15%的人类癌症染色体9p21(chr9p21)基因组纯合缺失,在多形性胶质母细胞瘤中的缺失频率高达>50%。染色体chr9p21位点包括CDKN2A基因,它编码关键的肿瘤抑制因子p19-ARF和p16-INK4a,Chr9p21缺失经常涉及CDKN2A近端基因的共缺失,其中最重要的是甲硫腺苷磷酸化酶(MTAP)基因。研究发现许多人恶性细胞缺乏MTAP活性。MTAP缺陷不仅存在于组织培养细胞中,而且该缺陷也存在于原发性白血病、胶质瘤、黑色素瘤、胰腺癌、非小细胞肺癌(NSLC)、膀胱癌、星形细胞瘤、骨肉瘤、头颈癌、粘液性软骨肉瘤、卵巢癌、子宫内膜癌、乳腺癌、软组织肉瘤、非霍奇金淋巴瘤和间皮瘤中。
目前MAT2A抑制剂在癌症等领域的研究取得了一定的进展,但仍需进一步开发新型的药物,优化目前在研MAT2A药物,为癌症治疗领域未被满足的临床需求提供新的治疗选择。
发明内容
本发明旨在提出一种能够有效抑制MAT2A的化合物,其能够作为目前药物或者MAT2A抑制剂的改进或者替换。
为此,在本发明的第一方面,本发明提出了一种式III所示化合物,其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药:
Figure PCTCN2022083568-appb-000001
其中,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
R 1选自C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基),所述C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基)任选被一个或多个R 1a取代,所述R 1a选自卤素;当R 1a为多个时,所述R 1a相同或不同;
或当L 1为-N(R L1)-时,R L1和R 1可以与L组合在一起形成任选地被一个或多个R L1取代的3至6元杂环烷基;当R L1为多个时,所述R L1相同或不同;所述3至6元杂环烷基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
R 2选自无取代或被R 2a取代的C 6-C 10-芳基、无取代或被R 2a取代的5至10元杂芳基;当R 2a为多个时,所述R 2a相同或不同;所述5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
R 3选自无取代或被R 3a取代的C 6-C 10-芳基、无取代或被R 3a取代的5至10元杂芳基、无取代或被R 3a取代的环A;当R 3a为多个时,所述R 3a相同或不同;所述环A为
Figure PCTCN2022083568-appb-000002
其中环Cx和环Cy各自独立地选自C 5-C 6-芳基、5至6元杂芳基、5至6元环烷基和5至6元杂环烷基,D和E各自独立地选自C、CH、N,G和J各自独立地选自C、CH、CH 2、N、NH、O、S;
所述R 2a选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A和-CN;
所述R 3a选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A、-CN、-(C 1-C 6亚烷基)-5-6元杂环烷基、-(C 1-C 6亚烷基)-S(O) 2-(C 1-C 3烷基)、-(C 1-C 6亚烷基)-C(O)-NR AR B
R A和R B各自独立地选自H、-CN、-OH、氧代、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基;所述3至14元杂环烷基和5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
所述R A和R B中的每个C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、 -C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基各自独立且任选地被一个或多个R Aa取代;当R Aa为多个时,所述R Aa相同或不同;
所述R Aa各自独立地选自-OH、氘、卤素、C 1-C 6烷基、-N(R An) 2,其中每个R An各自独立地选自H、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 6-C 10芳基、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和5至10元杂芳基、-NHC(O)-(OC 1-C 6烷基)、-NO 2、-CN、氧代、-C(O)OH、-C(O)-O-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 1-C 6-烷氧基)、-C(O)NH 2、-C(O)-(C 1-C 6烷基)、-O-(C 1-C 6烷基)、-Si(C 1-C 6烷基) 3、-S(O) 0-2-(C 1-C 6烷基)、C 6-C 10芳基、-(C 1-C 6亚烷基)-(C 6-C 10芳基)、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和-O(C 6-C 14芳基);
所述R Aa中的每个烷基、烯基、芳基和杂环烷基各自独立且任选地被一个或多个R Ab取代,所述R Ab各自独立地选自-OH、-O-(C 1-C 6烷基)、卤素、-NH 2、-(C 1-C 6亚烷基)-NH 2、-COOH、-CN和氧代;
R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同。
所述烷基包括直链烷基、支链烷基。
本领域技术人员可以理解,根据本领域中使用的惯例,在本申请的结构式中,
Figure PCTCN2022083568-appb-000003
用于描绘化学键,所述化学键为部分或取代基与核心结构或骨架结构相连的点。
根据本发明的某些实施例,式III所示化合物中,L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;根据本发明的一优选实施例,R L1选自氢、甲基、乙基。较佳地,L 1选自-O-、或-NH-。
根据本发明的某些实施例,式III所示化合物中,R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基);所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代,所述R 1a选自卤素;根据本发明的某些实施例,R 1选自C 1-C 3烷基、C 3-C 6环烷基、-(C 1-C 3亚烷基)-(C 3-C 6环烷基),所述C 1-C 3烷基、C 3-C 6环烷基、-(C 1-C 3亚烷基)-(C 3-C 6环烷基)任选地被一个或多个R 1a取代;当R 1a为多个时,所述R 1a相同或不同;所述R 1a选自卤素;较佳地,所述卤素为F;根据本发明的一优选实施例,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
Figure PCTCN2022083568-appb-000004
根据本发明的又一优选实施例,R 1选自 甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000005
根据本发明的又一优选实施例,-L 1-R 1选自:-NH-环丙基、-O-环丙基、-NH-CH 2CF 3、-O-CH 2CF 3、-NH-CH 2-环丙基、-O-CH 2-环丙基。
根据本发明的某些实施例,式III所示化合物中,R 2选自无取代或被R 2a取代的苯基和无取代或被R 2a取代的5至10元杂芳基,所述5至10元杂芳基其中一个环原子为N;根据本发明的一优选实施例,R 2选自无取代或被R 2a取代的苯基、无取代或被R 2a取代的吡啶基、无取代或被R 2a取代的嘧啶基。
根据本发明的某些实施例,式III所示化合物中,R 3为被R 3a取代的环A;所述R 3a选自-C 1-C 6烷基、-(C 1-C 6亚烷基)-5-6元杂环烷基、-(C 1-C 6亚烷基)-S(O) 2-CH 3、-(C 1-C 6亚烷基)-C(O)-N(CH 3) 2;所述环A为
Figure PCTCN2022083568-appb-000006
其中,环Cx为5元杂芳环或5元杂环烷基;环Cy为苯环、5-6元杂芳环或5-6元杂环烷基;D和E各自独立地选自C、CH、N;较佳地,所述环Cx为5元杂芳环,所述杂芳环含有2个N或所述杂芳环含有1个N和1个S;较佳地,所述环Cx为5元杂环烷基,所述杂环烷基含有1个O;较佳地,所述环Cy含有1或2个N;
根据本发明的某些实施例,式III所示化合物中,R 3选自无取代或被R 3a取代的苯基、无取代或被R 3a取代的吡啶基、无取代或被R 3a取代的吡啶酮基、无取代或被R 3a取代的哒嗪基、无取代或被R 3a取代的异噁唑基、无取代或被R 3a取代的吡唑基、和无取代或被R 3a取代的环A,所述环A选自
Figure PCTCN2022083568-appb-000007
苯并噻唑基、苯并异噻唑基、苯并噁唑基、苯并咪唑基、苯并三唑基、吲唑基、喹喔啉基、喹啉基、喹唑啉基、咪唑并吡啶基、吡唑并吡啶基、三唑并吡啶基、苯并哒嗪基、苯并呋喃基、二氢苯并呋喃基、二氢苯并二氧乙烯基和四氢苯并二氧乙烯基,其中n A选自整数1、2或3;根据本发明的一优选实施例,所述环A选自苯并噻唑基、苯并咪唑基、吲唑基、苯并哌啶基、苯并哌嗪基、苯并吗啉基、二氢茚、苯并二氢呋喃。
根据本发明的某些实施例,式III所示化合物中,R 2a和R 3a各自独立地选自-R A、-OR A、卤素和-CN。
根据本发明的某些实施例,式III所示化合物中,R A独立地选自H、-OH、C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、 -CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)各自独立且任选地被一个或多个R Aa取代。
根据本发明的某些实施例,式III所示化合物中,R Aa各自独立地选自-OH、卤素、-N(R An) 2,其中每个R An各自独立地选自H、甲基、乙基、异丙基、正丙基。
根据本发明的某些实施例,式III所示化合物中,R 4独立地选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自氟和氯;当R 4a为多个时,所述R 4a相同或不同。
根据本发明的一优选实施例,式III所示化合物中,R 4选自氢、卤素、-OH、-CN、甲基、乙基、正丙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、氟代环丙基、
Figure PCTCN2022083568-appb-000008
Figure PCTCN2022083568-appb-000009
氯代甲基、氯代乙基、氯代丙基、氯代环丙基、
Figure PCTCN2022083568-appb-000010
较佳地,R 4选自氢、卤素、甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000011
R 4优选为氢。
根据本发明的某些实施例,式III所示化合物中,R 2选自被R 2a取代的苯基和被R 2a取代的吡啶基,并且R 2a独立地选自氟、氯、溴、任选被一个或多个R Aa取代的-R A、任选被一个或多个R Aa取代的-OR A,所述R A独立地选自被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基,每个R Aa独立地选自-OH、氟、氯、溴;根据本发明的一优选实施例,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
Figure PCTCN2022083568-appb-000012
氟代甲氧基;根据本发明的某些实施例,R 2
Figure PCTCN2022083568-appb-000013
R 2a选自:氟、氯、溴、-O-C 1-C 6烷基或C 3-C 6环烷基;所述C 1-C 6烷基任选地被一个或多个卤素取代;较佳地,所述卤素为F;较佳地,R 2a选自:氟、氯、-O-CF 2H或环丙基;根据本发明的又一优选实施例,R 2选自
Figure PCTCN2022083568-appb-000014
Figure PCTCN2022083568-appb-000015
根据本发明的某些实施例,式III所示化合物中,R 3选自被R 3a取代的环A,所述环A 选自
Figure PCTCN2022083568-appb-000016
Figure PCTCN2022083568-appb-000017
并且R 3a独立地选自任选被一个或多个R Aa取代的-R A,所述R A独立地选自被一个或多个R Aa取代的C 1-C 6烷基,每个R Aa各自独立地选自-OH、氟、氯、溴;根据本发明的一优选实施例,R 3a独立地选自甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、叔丁基、-CH 2-OH、-(CH 2) 2-OH、-(CH 2) 3-OH、-(CH 2) 4-OH、
Figure PCTCN2022083568-appb-000018
根据本发明的某些实施例,式III所示化合物中,R 3选自:
Figure PCTCN2022083568-appb-000019
Figure PCTCN2022083568-appb-000020
Figure PCTCN2022083568-appb-000021
R 3a选自:-C 1-C 6烷基、-C 1-C 6烷基-5-6元杂环烷基、-C 1-C 6烷基-S(O) 2-CH 3、-C 1-C 6烷基-C(O)-N(CH 3) 2;较佳地,所述C 1-C 6烷基为直链或含支链的烷基;所述C 1-C 6烷基任选地被-OH取代;较佳地,所述5-6元杂环烷基为
Figure PCTCN2022083568-appb-000022
较佳地,R 3a选自:-CH 3、-
Figure PCTCN2022083568-appb-000023
Figure PCTCN2022083568-appb-000024
根据本发明的一优选实施例,式III所示化合物中,R 3选自
Figure PCTCN2022083568-appb-000025
Figure PCTCN2022083568-appb-000026
根据本发明的某些实施例,式III所示化合物具有式II所示结构:
Figure PCTCN2022083568-appb-000027
其中,R 1、R 2a、R 3a、R 4、L 1具有上文所述的定义,Y 1、Y 2、Y 3各自独立地选自N、S、C、CH。
根据本发明的某些实施例,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3- C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代;当R 1a为多个时,所述R 1a相同或不同;所述R 1a选自卤素;
X 1选自C(R 2a)、N;
Y 1、Y 2、Y 3各自独立地选自N、S、C、CH;
R 2a独立地选自-R A、-OR A和卤素;
R 3a独立地选自-R A
R A独立地选自H、-CN、-OH、C 1-C 6烷基、C 3-C 6环烷基、被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基;当R Aa为多个时,所述R Aa相同或不同;
所述R Aa各自独立地选自-OH、氘、卤素;
R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同。
根据本发明的某些实施例,式II所示化合物中,L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;较佳地,R L1选自氢、甲基、乙基。
根据本发明的某些实施例,式II所示化合物中,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
Figure PCTCN2022083568-appb-000028
根据本发明的优选实施例,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000029
根据本发明的某些实施例,式II所示化合物中,X 1选自CH、N。
根据本发明的某些实施例,式II所示化合物中,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
Figure PCTCN2022083568-appb-000030
氟代甲氧基。
根据本发明的某些实施例,式II所示化合物中,R 3a独立地选自甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、叔丁基、-CH 2-OH、-(CH 2) 2-OH、-(CH 2) 3-OH、-(CH 2) 4-OH、
Figure PCTCN2022083568-appb-000031
根据本发明的某些实施例,式II所示化合物中,R 4选自氢、卤素、-OH、-CN、甲基、乙 基、正丙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、氟代环丙基、
Figure PCTCN2022083568-appb-000032
氯代甲基、氯代乙基、氯代丙基、氯代环丙基、
Figure PCTCN2022083568-appb-000033
较佳地,R 4选自氢、卤素、甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000034
根据本发明的某些实施例,式II所示化合物具有式II-a或II-b所示结构:
Figure PCTCN2022083568-appb-000035
其中,R 1、L 1、R 2a、R 3a具有上文所述的定义。
根据本发明的某些实施例,式III所示化合物还可以具有式I所示结构:
Figure PCTCN2022083568-appb-000036
其中,R 1、L 1、R 2a、R 3a、R 4、n A具有上文所述的定义。
根据本发明的某些实施例,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代;当R 1a为多个时,所述 R 1a相同或不同;所述R 1a选自卤素;
X 1选自C(R 2a)、N;
R 2a独立地选自-R A、-OR A和卤素;
R 3a独立地选自-R A
R A独立地选自H、-CN、-OH、C 1-C 6烷基、C 3-C 6环烷基、被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基;当R Aa为多个时,所述R Aa相同或不同;
所述R Aa各自独立地选自-OH、氘、卤素;
R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同;
n A选自整数1、2或3。
根据本发明的某些实施例,式I所示化合物中,L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;较佳地,R L1选自氢、甲基、乙基。
根据本发明的某些实施例,式I所示化合物中,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
Figure PCTCN2022083568-appb-000037
较佳地,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000038
根据本发明的某些实施例,式I所示化合物中,X 1选自CH、N。
根据本发明的某些实施例,式I所示化合物中,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
Figure PCTCN2022083568-appb-000039
氟代甲氧基。
根据本发明的某些实施例,式I所示化合物中,R 3a独立地选自甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、叔丁基、-CH 2-OH、-(CH 2) 2-OH、-(CH 2) 3-OH、-(CH 2) 4-OH、
Figure PCTCN2022083568-appb-000040
根据本发明的某些实施例,式I所示化合物中,R 4选自氢、卤素、-OH、-CN、甲基、乙基、正丙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、氟代环丙基、
Figure PCTCN2022083568-appb-000041
氯代甲基、氯代乙基、氯代丙基、氯代环丙基、
Figure PCTCN2022083568-appb-000042
较佳地,R 4选自氢、卤素、甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000043
根据本发明的某些实施例,所述式III化合物选自下列任一化合物:
Figure PCTCN2022083568-appb-000044
Figure PCTCN2022083568-appb-000045
在本发明的第二方面,本发明提供了中间体M-1和M-2:
Figure PCTCN2022083568-appb-000046
其中,R M选自卤素,L 1、R 1、R 2、R 4具有上文所述的定义。
根据本发明的某些实施例,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
根据本发明的某些实施例,R 1选自C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基);
所述C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基)任选被一个或多个R 1a取代,所述R 1a选自卤素;当R 1a为多个时,所述R 1a相同或不同;
或当L 1为-N(R L1)-时,R L1和R 1可以与L组合在一起形成任选地被一个或多个R L1取代的3至6元杂环烷基;当R L1为多个时,所述R L1相同或不同;所述3至6元杂环烷基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
根据本发明的某些实施例,R 2选自无取代或被R 2a取代的C 6-C 10芳基、无取代或被R 2a取代的5至10元杂芳基;当R 2a为多个时,所述R 2a相同或不同;所述5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
所述R 2a独立地选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A和-CN;
R A和R B各自独立地选自H、-CN、-OH、氧代、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基;所述3至14元杂环烷基和5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
所述R A和R B中的每个C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基各自独立且任选地被一个或多个R Aa取代;当R Aa为多个时,所述R Aa相同或不同;
所述R Aa各自独立地选自-OH、氘、卤素、-N(R An) 2,其中每个R An各自独立地选自H、C 1- C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 6-C 10芳基、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和5至10元杂芳基、-NHC(O)-(OC 1-C 6烷基)、-NO 2、-CN、氧代、-C(O)OH、-C(O)-O-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 1-C 6-烷氧基)、-C(O)NH 2、-C(O)-(C 1-C 6烷基)、-O-(C 1-C 6烷基)、-Si(C 1-C 6烷基) 3、-S(O) 0-2-(C 1-C 6烷基)、C 6-C 10芳基、-(C 1-C 6亚烷基)-(C 6-C 10芳基)、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和-O(C 6-C 14芳基);
所述R Aa中的每个烷基、烯基、芳基和杂环烷基各自独立且任选地被一个或多个R Ab取代,所述R Ab各自独立地选自-OH、-O-(C 1-C 6烷基)、卤素、-NH 2、-(C 1-C 6亚烷基)-NH 2、-COOH、-CN和氧代;
且所述中间体M-1不包括下列化合物:
Figure PCTCN2022083568-appb-000047
根据本发明的某些实施例,中间体M-1和M-2中,R M选自氟、氯、溴、碘;较佳地,R M选自氟、氯、溴。
根据本发明的某些实施例,中间体M-1和M-2中,L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基。根据本发明的某些优选实施例,R L1选自氢、甲基、乙基。
根据本发明的某些实施例,中间体M-1和M-2中,R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基);所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代,所述R 1a选自卤素。根据本发明的某些优选实施例,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
Figure PCTCN2022083568-appb-000048
较佳地,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
Figure PCTCN2022083568-appb-000049
根据本发明的某些实施例,中间体M-1和M-2中,R 2选自无取代或被R 2a取代的苯基和无取代或被R 2a取代的5至10元杂芳基,所述5至10元杂芳基其中一个环原子为N;较佳 地,R 2选自无取代或被R 2a取代的苯基、无取代或被R 2a取代的吡啶基、无取代或被R 2a取代的嘧啶基。
根据本发明的某些实施例,中间体M-1和M-2中,R 2a独立地选自-R A、-OR A、卤素和-CN。
根据本发明的某些实施例,中间体M-1和M-2中,R A独立地选自H、-OH、C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)各自独立且任选地被一个或多个R Aa取代。
根据本发明的某些实施例,中间体M-1和M-2中,R Aa各自独立地选自-OH、卤素、-N(R An) 2,其中每个R An各自独立地选自H、甲基、乙基、异丙基、正丙基。
根据本发明的某些实施例,中间体M-1和M-2中,R 2选自被R 2a取代的苯基和被R 2a取代的吡啶基,并且R 2a独立地选自氟、氯、溴、任选被一个或多个R Aa取代的-R A、任选被一个或多个R Aa取代的-OR A,所述R A独立地选自被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基,每个R Aa独立地选自-OH、氟、氯、溴;根据本发明的某些优选实施例,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
Figure PCTCN2022083568-appb-000050
氟代甲氧基;更佳地,R 2选自
Figure PCTCN2022083568-appb-000051
Figure PCTCN2022083568-appb-000052
根据本发明的某些实施例,所述中间体M-1选自下列任一化合物:
Figure PCTCN2022083568-appb-000053
Figure PCTCN2022083568-appb-000054
根据本发明的某些实施例,所述中间体M-2选自下列任一化合物:
Figure PCTCN2022083568-appb-000055
在本发明的第三方面,本发明提供了一种制备式III所述化合物的方法,其包括:
(1)使所述中间体M-1与卤化试剂接触,得到所述中间体M-2;和/或
Figure PCTCN2022083568-appb-000056
(2)使所述中间体M-2与
Figure PCTCN2022083568-appb-000057
接触,得到式III所示化合物,
Figure PCTCN2022083568-appb-000058
其中,R 1、R 2、R 3、R 4、L 1具有上文所述的定义,R M选自氟、氯、溴、碘;较佳地,R M选自氟、氯、溴。
根据本发明的某些实施例,步骤(1)中,所述卤化试剂选自N-氯代丁二酰亚胺、N-溴代琥珀酰亚胺。
根据本发明的某些实施例,步骤(2)中,在惰性气体保护下将所述中间体M-2与
Figure PCTCN2022083568-appb-000059
钯催化剂和碱接触,得到式III所示化合物。
在本发明的第四方面,本发明提供了一种药物组合物,其包含治疗有效量的式III所示的化合物、其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药中的至少一种。
根据本发明的某些实施例,所述药物组合物还包括一种或多种药学上可接受的辅料。
根据本发明的某些实施例,所述药物组合物还可以进一步含有一种或多种额外的治疗剂。
在本发明的第五方面,本发明提供了式III所示的化合物、其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药或所述的药物组合物在制备药物中的用途,所述药物用于治疗或者预防MAT2A相关疾病,本发明还提供了本发明第一方面所述的任意化合物或本发明第四方面所述的药物组合物治疗或者预防MAT2A相关疾病的医药用途。
根据本发明的某些实施例,所述药物用于治疗或者预防癌症。
根据本发明的某些实施例,所述癌症是MTAP缺失的癌症。
根据本发明的某些实施例,所述癌症选自间皮瘤、神经母细胞瘤、直肠癌、结肠癌、熟悉的腺瘤性息肉病和遗传性非息肉性结直肠癌、食道癌、唇癌、喉癌、下咽癌、舌癌、唾液腺癌、胃癌、腺癌、甲状腺髓样癌、甲状腺乳头状癌、肾癌、肾实质癌、卵巢癌、子宫颈癌、子宫体癌、子宫内膜癌、绒毛膜癌、胰腺癌、前列腺癌、膀胱癌、睾丸癌、乳腺癌、泌尿癌、黑素瘤、脑瘤、淋巴瘤、头颈癌、急性淋巴白血病(ALL)、慢性淋巴白血病(CLL)、急性髓样白血病(AML)、慢性粒细胞白血病(CML)、肝细胞癌、胆囊癌、支气管癌、小细胞肺癌、非小细胞肺癌、多发性骨髓瘤、基底肉瘤、畸胎瘤、视网膜母细胞瘤、脉络膜黑色素瘤、精原细胞瘤、横纹肌肉瘤、骨肉瘤、软骨肉瘤、肌瘤、脂肪肉瘤、纤维肉瘤、尤因肉瘤和浆细胞瘤。
本发明还提供一种MAT2A相关疾病的方法,包括给予患者预防或治疗有效量的式(I)所示的化合物、其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药中的至少一种,或上述药物组合物。
根据本发明的某些实施例,所述患者是哺乳动物,优选是人。
本发明还提供式(I)所示的化合物、其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药中的至少一种,或其药物组合物在治疗或预防MAT2A相关疾病中的应用。
有益效果
根据本发明的实施例,本发明所述化合物和/或其组合物能够有效抑制MAT2A酶活性,对HCT116 MTAP-/-细胞增殖及细胞中SAM水平有很好的抑制作用,具有更好的药代动力学性质。在制备治疗与MAT2A相关疾病的药物方面具有广阔的应用前景。
本发明的附加方面和优点将在下面的描述中部分给出,部分将从下面的描述中变得明显,或通过本发明的实践了解到。
术语定义与说明
除非另有说明,本申请说明书和权利要求书中记载的基团和术语定义,包括其作为实例的定义、示例性的定义、优选的定义、表格中记载的定义、实施例中具体化合物的定义等,可以彼此之间任意组合和结合。这样的组合和结合后的基团定义及化合物结构,应当属于本申请说明书记载的范围内。
除非另有定义,否则本文所有科技术语具有的涵义与权利要求主题所属领域技术人员通常理解的涵义相同。除非另有说明,本文全文引用的所有专利、专利申请、公开材料通过引用方 式整体并入本文。如果本文对术语有多个定义,以本章的定义为准。
除非另有说明,否则采用本领域技术范围内的常规方法,如质谱、NMR、IR和UV/Vis光谱法和药理学方法。除非提出具体定义,否则本文在分析化学、有机合成化学以及药物和药物化学的有关描述中采用的术语是本领域已知的。可在化学合成、化学分析、药物制备、制剂和递送,以及对患者的治疗中使用标准技术。例如,可利用厂商对试剂盒的使用说明,或者按照本领域公知的方式或本申请的说明来实施反应和进行纯化。通常可根据本说明书中引用和讨论的多个概要性和较具体的文献中的描述,按照本领域熟知的常规方法实施上述技术和方法。在本说明书中,可由本领域技术人员选择基团及其取代基以提供稳定的结构部分和化合物。当通过从左向右书写的常规化学式描述取代基时,该取代基也同样包括从右向左书写结构式时所得到的在化学上等同的取代基。举例而言,CH 2O等同于OCH 2
本申请说明书和权利要求书记载的数值范围,当该数值范围被理解为“整数”时,应当理解为记载了该范围的两个端点以及该范围内的每一个整数。例如,“1~6的整数”应当理解为记载了0、1、2、3、4、5和6的每一个整数。当该数值范围被理解为“数”时,应当理解为记载了该范围的两个端点以及该范围内的每一个整数以及该范围内的每一个小数。例如,“1~10的数”应当被理解为不仅记载了1、2、3、4、5、6、7、8、9和10的每一个整数,还至少记载了其中每一个整数分别与0.1、0.2、0.3、0.4、0.5、0.6、0.7、0.8、0.9的和。
术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”或者“其药学上可接受的盐”是指药学上可接受的无毒酸或碱的盐,包括无机酸和碱、有机酸和碱的盐。
除了药学可接受的盐外,本发明还考虑其他盐。它们可以在化合物纯化中或在制备其它药学上课接受的盐中充当中间体或可用于本发明化合物的鉴别、表征或纯化。
术语“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体。本发明使用的立体化学定义和惯例大体上按照S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-Hill Book Company,New York;and Eliel,E.and Wilen,S.,“Stereochemistry of Organic Compounds”,John Wiley&Sons,Inc.,New York,1994来定义。本发明化合物可含有不对称中心或手性中心,因此以不同的立体异构形式存在。所预期的是,本发明化合物的所有立体异构体形式,包括但不限于非对映异构体、对映异构体和阻转异构体(atropisomer)和几何(或构象)异构体及它们的混合物,如外消旋混合物,均在本发明的范围之内。
许多有机化合物以光学活性形式存在,即它们具有使平面偏振光的平面发生旋转的能力。当描述具有光学活性的化合物时,使用前缀D和L或R和S来表示就分子中的手性中心(或多个手性中心)而言分子的绝对构型。前缀D和L或(+)和(–)是用于指定化合物所致平面偏振光旋转的符号,其中(–)或L表示化合物是左旋的。前缀为(+)或D的化合物是右旋的。就给定的化学结构而言,除了这些立体异构体互为镜像外,这些立体异构体是相同的。具体的立体异构体也可称为对映异构体,并且所述异构体的混合物通常称作对映异构体的混合物。对映异构体的50:50混合物称为外消旋混合物或外消旋体,当在化学反应或方法中没有立体选择性或立体特异性时,可出现所述外消旋混合物或外消旋体。
依据原料和方法的选择,本发明化合物可以以可能的异构体中的一个或它们的混合物的形式存在,例如作为纯旋光异构体,或作为异构体混合物,如作为外消旋和非对映异构体混合物,这取决于不对称碳原子的数量。旋光性的(R)-或(S)-异构体可使用手性合成子或手性制剂制备,或使用常规技术拆分。如果此化合物含有一个双键,取代基可能为E或Z构型;如果此化合物中含有二取代的环烷基,环烷基的取代基可能为顺式或反式(cis-或trans-)构型。
当将本发明通式中与手性碳的键描写成直线时,应当理解为,手性碳的(R)和(S)两种构型和由此产生的其对映体纯的化合物和混合物两者包括在该通式范围内。本文中消旋体或者对映体纯的化合物的图示法来自Maehr,J.Chem.Ed.1985,62:114-120。除非另有说明,用楔形键和虚线键表示一个立体中心的绝对构型。
含有不对称取代的碳原子的本发明化合物能够以旋光活性形式或外消旋形式分离。化合物的外消旋混合物的拆分可以通过本领域已知的许多方法中的任一种来进行。示例性方法包括使用手性拆分酸的分级重结晶,该手性拆分酸是旋光活性的成盐有机酸。用于分级重结晶方法的适合的拆分剂例如是旋光活性酸,例如酒石酸、二乙酰基酒石酸、二苯甲酰基酒石酸、扁桃酸、苹果酸、乳酸或各种旋光活性樟脑磺酸如β-樟脑磺酸的D和L形式。适合于分级结晶方法的其它的拆分剂包括立体异构纯形式的α-甲基-苄胺(例如,S和R形式或者非对映异构纯形式)、2-苯基甘氨醇、降麻黄碱、麻黄碱、N-甲基麻黄碱、环己基乙胺、1,2-二氨基环己烷等。外消旋混合物的拆分还可以通过在填充有旋光活性拆分剂(例如,二硝基苯甲酰基苯基甘氨酸)的柱子上洗脱来进行。可以采用高效液相色谱(HPLC)法也可以采用超临界流体色谱法(SFC)进行。具体方法的选择以及洗脱条件、色谱柱的选择可以由本领域技术人员根据化合物的结构以及试验结果选择。进一步的,还可以使用已知构型的光学纯的起始原料或试剂,通过立体有机合成,获得本发明所描述化合物的任何对映体或非对映体。
烯烃、C=N双键等的许多几何异构体也可以存在于本文所述的化合物中,且所有这种稳定的异构体在本发明中均被考虑。当本文所描述化合物含有烯双键时,除非另外说明,否则,这种双键包括E和Z几何异构体。
术语“互变异构体”是指因分子中某一原子在两个位置迅速移动而产生的官能团异构体。本发明化合物可表现出互变异构现象。互变异构的化合物可以存在两种或多种可相互转化的种类。质子移变互变异构体来自两个原子之间共价键合的氢原子的迁移。互变异构体一般以平衡形式存在,尝试分离单一互变异构体时通常产生一种混合物,其理化性质与化合物的混合物是一致的。平衡的位置取决于分子内的化学特性。例如,在很多脂族醛和酮如乙醛中,酮型占优势;而在酚中,烯醇型占优势。本发明包含化合物的所有互变异构形式。
术语“药物组合物”表示一种或多种文本所述化合物或其生理学/药学上可接受的盐或前体药物与其它化学组分的混合物,其它组分例如生理学/药学上可接受的载体和赋形剂。药物组合物的目的是促进化合物对生物体的给药。
术语“溶剂化物”指本发明化合物或其盐包括以分子间非共价力结合的化学计量或非化学计量的溶剂,当溶剂为水时,则为水合物。
术语“前药”是指可以在生理条件下或者通过溶剂解转化为具有生物活性的本发明化合物。本发明的前药通过修饰在该化合物中的功能基团来制备,该修饰可以按常规的操作或者在体内被除去,而得到母体化合物。前药包括本发明化合物中的一个羟基或者氨基连接到任何基团上所形成的化合物,当本发明化合物的前药被施予哺乳动物个体时,前药被割裂而分别形成游离的羟基、游离的氨基。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“辅料”是指可药用惰性成分。术语“赋形剂”的种类实例非限制性地包括粘合剂、崩解剂、润滑剂、助流剂、稳定剂、填充剂和稀释剂等。
术语“C 1-C 6烷基”应理解为表示具有1、2、3、4、5或6个碳原子的直链或支链饱和一价烃基具有3至6个碳原子的支链的饱和单价烃基。所述烷基是例如甲基、乙基、丙基、丁基、戊基、己基、异丙基、异丁基、仲丁基、叔丁基、异戊基、2-甲基丁基、1-甲基丁基、1-乙基丙基、1,2-二甲基丙基、新戊基、1,1-二甲基丙基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、2-乙基丁基、1-乙基丁基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、2,3-二甲基丁基、1,3-二甲基丁基或1,2-二甲基丁基等或它们的异构体。特别地,所述基团具有1、 2或3个碳原子(“C 1-C 3烷基”),例如甲基、乙基、正丙基或异丙基。本领域技术人员将认识到,术语“烷基”可包括“亚烷基”基团。
术语“亚烷基”应理解为表示具有1-6个碳原子的直链饱和二价烃基或具有3-6个碳原子的支链饱和二价烃基,除非另有说明,例如亚甲基、亚乙基、亚丙基、1-甲基亚丙基、亚丁基等。
术语“C 2-C 6烯基”应理解为表示含有一个或多个碳-碳双键、具有2、3、4、5或6个碳原子的直链一价烃基或具有丙烯基、丁烯基等的双键的具有3至6个碳原子的支链一价烃基。
术语“C 2-C 6炔基”应理解为表示含有2-6个碳原子和至少一个碳碳三键的直链、支链或者环状烃基,例如乙炔基、丙炔基、丁炔基和3-甲基丁炔基等。
术语“C 3-C 6环烷基”应理解为表示饱和的一价单环或双环烃环,其具有3~6个碳原子,包括稠合或桥接的多环系统。如环丙基、环丁基、环戊基、环己基。
术语“C 1-C 6烷氧基”应理解为-O-(C 1- 6烷基),其中“C 1- 6烷基”具有上述定义。
术语“卤代基”或“卤素”是指氟、氯、溴或碘,优选氟或氯。
术语“卤代烷基”指包括具有特定数目的碳原子、被一或多个卤素取代的支链和直链的饱和脂族烃基(如-CvFw,其中v=1至3,w=1至(2v+1))。卤代烷基的实例包括,但不限于三氟甲基、三氯甲基、五氟乙基、五氯乙基、2,2,2-三氟乙基、七氟丙基和七氯丙基。
术语“C 6-C 10-芳基”是指6至10个环原子的单价单环或双环芳族烃基,例如苯基或萘基。
术语“5至10元杂芳基”是指具有5至10个环原子的单价单环或双环芳族基团,其包含一个或两个芳族环,其中一个或多个(在某些实施例中为1、2、3或4个)环原子为独立选自O、S和N的杂原子,其余为碳原子。
双环情况下,术语“双环”或“稠合”是指两个环经两个原子间的键连接在一起(例如,萘),经一系列原子结合在一起形成一桥(例如,奎宁环)或单个原子在一起形成螺环化合物(例如,1,4-二氧杂-8-氮杂-螺[4.5]癸烷及N,3,3-二甲基-1,5-二氧杂螺[5.5]十一烷-9-基)。
术语“3至6元杂环烷基”是指3至6个环原子的饱和单环基团,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;另外,杂环基环中的一个或两个环碳原子可以任选地被-CO-基团取代。
本文单独或组合使用的“氧代”是指=O。
Figure PCTCN2022083568-appb-000060
表示含2个N原子的五元杂芳环,其中
Figure PCTCN2022083568-appb-000061
表示离域大π键。
术语“患者”是指包括哺乳动物在内的任何动物,优选小鼠、大鼠、其它啮齿类动物、兔、狗、猫、猪、牛、羊、马或灵长类动物,最优选人。
术语“治疗有效量”是指研究人员、兽医、医师或其它临床医师正在组织、系统、动物、个体或人中寻找的引起生物学或医学反应的活性化合物或药物的量,它包括以下一项或多项:(1)预防疾病:例如在易感染疾病、紊乱或病症但尚未经历或出现疾病病理或症状的个体中预防疾病、紊乱或病症。(2)抑制疾病:例如在正经历或出现疾病、紊乱或病症的病理或症状的个体中抑制疾病、紊乱或病症(即阻止病理和/或症状的进一步发展)。(3)缓解疾病:例如在正经历或出现疾病、紊乱或病症的病理或症状的个体中缓解疾病、紊乱或病症(即逆转病理和/或症状)。
具体实施方式
下文将结合具体实施例对本发明的技术方案做更进一步的详细说明。应当理解,下列实施例仅为示例性地说明和解释本发明,而不应被解释为对本发明保护范围的限制。凡基于本发明上述内容所实现的技术均涵盖在本发明旨在保护的范围内。
除非另有说明,以下实施例中使用的原料和试剂均为市售商品,或者可以通过已知方法制备。
如无特别说明,本发明的化合物均是通过核磁共振(NMR)和/或质谱(MS)来确定。NMR位移的单位为10 -6(ppm)。NMR测定的溶剂为氘代二甲基亚砜、氘代氯仿、氘代甲醇等,内标为四甲基硅烷(TMS)。
本发明的缩写定义如下:
试剂:
DIPEA:也可写为DIEA,二异丙基乙胺,亦即N,N-二异丙基乙胺
THF:四氢呋喃
DMSO:二甲基亚砜
NCS:N-氯代丁二酰亚胺,亦即氯代丁二酰亚胺
NBS:N-溴代琥珀酰亚胺
Pd(dppf)Cl 2:[1,1'-双(二苯基膦基)二茂铁]二氯化钯
EA:乙酸乙酯
EtOH:乙醇
AcOH:乙酸
MeOH:甲醇
符号或单位:
EC 80:concentration for 80%of maximal effect,能引起80%最大效应的浓度
IC 50:半数抑制浓度,指达到最大抑制效果一半时的浓度
M:mol/L,摩尔浓度,如1M盐酸表示1mol/L盐酸溶液
N:当量浓度,例如2N盐酸表示2mol/L盐酸溶液
RT:保留时间
试验方法:
LC-MS:液质联用色谱
SFC:超临界流体色谱
TLC:薄层色谱
除非作出相反的指示,本文例举的化合物使用ChemBioDraw Ultra 13.0命名和编号。
实施例1:化合物I-1的制备
2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(化合物I-1)
Figure PCTCN2022083568-appb-000062
化合物I-1的合成路线如下所示:
Figure PCTCN2022083568-appb-000063
第一步:2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(I-1B)的合成
Figure PCTCN2022083568-appb-000064
把2,4-二氯-5-硝基嘧啶(5.00g,25.8mmol)和4-(二氟甲氧基)苯胺(4.51g,28.3mmol)溶解在四氢呋喃(40mL)中,降温到-40℃,缓慢加入N,N-二异丙基乙胺(6.66g,51.5mmol),-40℃反应2小时。反应结束后加水(80mL)淬灭,升至室温,用乙酸乙酯(160mL)萃取,然后用饱和食盐水(160mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩得到黄色固体2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(I-1B)(7.8g,产率88.4%)。
LC-MS,M/Z(ESI):317.0(M+H) +
第二步:N 2-环丙基-N 4-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-2,4-二胺(I-1C)的合成
Figure PCTCN2022083568-appb-000065
把2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(4.00g,12.6mmol),环丙胺(865.4mg,15.2mmol),N,N-二异丙基乙胺(4.08g,31.6mmol)和氟化铯(1.92g,12.63mmol)加入二甲亚砜(15mL)中,置换氮气,加热到80℃,搅拌反应3小时。将反应液降至室温,倒入水(50mL)中,过滤,收集固体,干燥得到黄色固体N 2-环丙基-N 4-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-2,4-二胺(I-1C)(4.00g,产率65.7%)。
LC-MS,M/Z(ESI):338.0(M+H) +
第三步:N 2-环丙基-N 4-(4-(二氟甲氧基)苯基)嘧啶-2,4,5-三胺(I-1D)的合成
Figure PCTCN2022083568-appb-000066
把N 2-环丙基-N 4-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-2,4-二胺(3.50g,7.26mmol)和钯碳(0.35g,含量为10%)加入甲醇(30mL)中,置换氢气,加热到30℃,50psi压力下搅拌反应3小时。将反应液降至室温过滤,将滤液浓缩得到红色固体N 2-环丙基-N 4-(4-(二氟甲氧基)苯基)嘧啶-2,4,5-三胺(I-1D)(2.1g,产率94.0%)。
LC-MS,M/Z(ESI):308.1(M+H) +
第四步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(I-1E)的合成
Figure PCTCN2022083568-appb-000067
把N 2-环丙基-N 4-(4-(二氟甲氧基)苯基)嘧啶-2,4,5-三胺(1.00g,3.25mmol),乙醛酸乙酯 (431.8mg,4.23mmol)和醋酸(195.4mg,3.25mmol)加入乙醇(20mL)中,置换氮气,加热到80℃反应3小时。将反应液降至室温,加入水(30mL),用乙酸乙酯(90mL×3)萃取,合并有机相,用饱和食盐水(90mL×2)洗涤,无水硫酸钠干燥,过滤,浓缩。残留物用硅胶柱纯化(石油醚:乙酸乙酯(V/V)=100:0-1:1),得到红色固体2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(I-1E)(520mg,产率46.2%)。
LC-MS,M/Z(ESI):346.1(M+H) +
第五步:6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(I-1F)的合成
Figure PCTCN2022083568-appb-000068
把2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(400mg,1.16mmol)和氯代丁二酰亚胺(232.0mg,1.74mmol)加入N,N-二甲基甲酰胺(10mL)中,置换氮气,在室温下搅拌3小时。将反应液加入水(30mL),乙酸乙酯(90mL×3)萃取,合并有机相,用饱和食盐水(90mL×2)洗涤。残留物用硅胶柱纯化(石油醚:乙酸乙酯(V/V):100:0-3:1)得到黄色固体6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(I-1F)(400mg,产率90.9%)。
LC-MS,M/Z(ESI):380.1(M+H) +
第六步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(目标化合物I-1)
Figure PCTCN2022083568-appb-000069
在氮气保护下将6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(380mg,1.00mmol),2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-2H-吲唑(387.4mg,1.50mmol),1,1’-双(二苯基磷)二茂铁氯化钯(73.2mg,100.0μmol)和碳酸钾(414.8mg,3.00mmol)溶解在1,4-二氧六环(10mL)和水(1mL)中,置换氮气,升温至90℃反应2小时。降至室温,将 反应液滴加入水(30mL)中,用乙酸乙酯(90mL)萃取。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩。然后将固体用甲醇(20mL)在60℃下打浆,热过滤得到黄色固体化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(176mg,产率36.2%)。
LC-MS,M/Z(ESI):476.2[M+H] +
1H NMR(400MHz,DMSO-d 6):δ8.80-8.82(m,2H),8.49(s,1H),8.09(d,1H),7.65(d,1H),7.17-7.57(m,6H),4.19(s,3H),2.45-2.47(m,1H),0.51-0.65(m,1H),0.42-0.49(m,3H).
实施例2:目标化合物I-2的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000070
第一步:2-环丙氧基-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺的合成
在氮气保护下,向2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(1.00g,3.16mmol)的二甲基亚砜(10.0mL)溶液中加入环丙醇(500mg,8.61mmol)和N,N-二异丙基乙胺(1.04g,8.04mmol)和氟化铯(480mg,3.16mmol),反应在50℃搅拌10小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(30.0mL*3)萃取,合并有机层,用水(30.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=30:1至10:1)得到化合物2-环丙氧基-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(550mg,1.63mmol,黄色固体,收率51.5%)。
LC-MS,M/Z(ESI):339.0(M+H) +
1H NMR(400MHz,CDCl 3)δ10.31(s,1H),9.25(s,1H),7.69-7.76(m,2H),7.15-7.22(m,2H),6.29-6.78(m,1H),4.29-4.34(m,1H),0.88-0.96(m,2H),0.81-0.88(m,2H)。
第二步:2-环丙氧基-N 4-(4-(二氟甲氧基)苯基)嘧啶-4,5-二胺的合成
在氮气保护下,向2-环丙氧基-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(550mg,1.63mmol)的甲醇(10.0mL)溶液中加入铂钒碳(500mg,192μmol,含量为10%),然后用氢气将反应体系置换3次,反应在15psi的氢气压力下,15℃搅拌5小时。反应完成后,将反应液过滤,用甲醇(30.0mL*2)洗涤,反应液浓缩,得到2-环丙氧基-N 4-(4-(二氟甲氧基)苯基)嘧啶-4,5-二胺(520mg黄色固体)。直接用于下一步。
第三步:2-环丙氧基-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮的合成
氮气保护下,向2-环丙氧基-N 4-(4-(二氟甲氧基)苯基)嘧啶-4,5-二胺(500mg,1.62mmol)的乙醇(5.00mL)溶液中加入乙醛酸乙酯的甲苯溶液(431mg,2.11mmol,浓度50.0%)和冰醋酸(97.4mg,1.62mmol),反应在80℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩得到化合物2-环丙氧基-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(500mg,1.44mmol,黄色固体,收率89.0%)。
LC-MS,M/Z(ESI):347.1(M+H) +
第四步:6-氯-2-环丙氧基-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮的合成
向2-环丙氧基-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(300mg,866μmol)的N,N-二甲基甲酰胺(1.00mL)溶液中加入N-氯代丁二酰亚胺(174mg,1.30mmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(10.0mL)稀释,然后用乙酸乙酯(10.0mL*3)萃取,合并有机层,用水(10.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩得到化合物6-氯-2-环丙氧基-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(280mg黄色固体)。直接用于下一步。
LC-MS,M/Z(ESI):381.2(M+H) +
第五步:2-环丙氧基-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-2)的合成
Figure PCTCN2022083568-appb-000071
在氮气保护下,向6-氯-2-环丙氧基-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(180mg,473μmol)的1,4-二氧六环(1.00mL)和水(200μL)的溶液中加入2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(183mg,709μmol),碳酸钾(196mg,1.42mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(34.6mg,47.3μmol),反应在75℃搅拌3小时。反应完成后,将反应混合物用水(10.0mL)稀释,然后用乙酸乙酯(10.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=5:1至1:3),然后用甲醇(5.00mL)打浆,得到化合物2-环丙氧基-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-2)(147mg,296μmol,黄色固体,收率62.6%)。
LC-MS,M/Z(ESI):477.0(M+H) +
1H NMR(400MHz,DMSO_d 6)δ9.11(s,1H),8.89(s,1H),8.53(s,1H),8.11(dd,1H),7.68(d,1H),7.19-7.56(m,5H),4.15-4.25(m,4H),0.69(d,4H).
实施例3:化合物I-3的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000072
第一步:N-(4-(二氟甲氧基)苯基)-5-硝基-2-(2,2,2-三氟乙氧基)嘧啶-4-胺的合成
在氮气保护下,向2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(1.20g,3.79mmol)的二甲基亚砜(10.0mL)溶液中加入2,2,2-三氟乙醇(1.14g,11.4mmol)和N,N-二异丙基乙胺(980mg,7.58mmol)和氟化铯(576mg,3.79mmol),反应在20℃搅拌5小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(30.0mL*3)萃取,合并有机层,用水(30.0mL*3)洗涤,无水硫酸钠干燥,过滤,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=30:1至10:1)得到化合物N-(4-(二氟甲氧基)苯基)-5-硝基-2-(2,2,2-三氟乙氧基)嘧啶-4-胺(900mg,2.37mmol,黄色固体,收率62.5%)。
LC-MS,M/Z(ESI):381.1(M+H) +
1H NMR(400MHz,CDCl 3)δ10.24(s,1H),9.26(s,1H),7.53-7.61(m,2H),7.19-7.25(m,2H),6.35-6.77(m,1H),4.77(q,2H).
第二步:N 4-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)嘧啶-4,5-二胺的合成
在氮气保护下,向N-(4-(二氟甲氧基)苯基)-5-硝基-2-(2,2,2-三氟乙氧基)嘧啶-4-胺(900mg,2.37mmol)的甲醇(20.0mL)溶液中加入铂钒碳(501mg,192μmol,含量为10%),然后用氢气将反应体系置换3次,反应在15psi的氢气压力下,15℃搅拌5小时。反应完成后,将反应液过滤,用甲醇(30.0mL*2)洗涤,反应液浓缩,得到N 4-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)嘧啶-4,5-二胺(920mg黄色固体)。直接用于下一步。
第三步:8-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮的合成
氮气保护下,向N 4-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)嘧啶-4,5-二胺(500mg,1.43mmol)的乙醇(5.00mL)溶液中加入乙醛酸乙酯的甲苯溶液(379mg,1.86mmol,50.0%浓度)和冰醋酸(85.7mg,1.43mmol),反应在90℃搅拌10小时。反应完成后,将反应混合物用水(30.0mL)稀释,然后用乙酸乙酯(30.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩得到化合物8-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮(550mg黄色固体)。直接用于下一步。
LC-MS,M/Z(ESI):389.1(M+H) +
第四步:6-氯-8-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮的合成
向8-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮(250mg,644μmol)的N,N-二甲基甲酰胺(2.00mL)溶液中加入N-氯代丁二酰亚胺(112mg,837μmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL *3)萃取,合并有机层,用水(20.0mL*3)洗涤,无水硫酸钠干燥,过滤,残留物用硅胶板分离纯化(石油醚:乙酸乙酯(V/V)=1:3),得到化合物
6-氯-8-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮(260mg黄色油状化合物)。直接用于下一步。
LC-MS,M/Z(ESI):422.9(M+H) +
第五步:8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮(I-3)的合成
Figure PCTCN2022083568-appb-000073
在氮气保护下,向6-氯-8-(4-(二氟甲氧基)苯基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮(220mg,520μmol)的1,4-二氧六环(3.00mL)和水(500μL)的溶液中加入2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(202mg,781μmol),碳酸钾(216mg,1.56mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(19.0mg,26.0μmol),反应在70℃搅拌3小时。反应完成后,将反应混合物用水(10.0mL)稀释,然后用乙酸乙酯(15.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=5:1至1:3),然后用甲醇(5.00mL)打浆,得到化合物8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)-2-(2,2,2-三氟乙氧基)蝶啶-7(8H)-酮(I-3)(10.9mg,20.2μmol,黄色固体,收率3.89%)。
LC-MS,M/Z(ESI):519.1(M+H) +
1H NMR(400MHz,DMSO_d 6)δ9.15(s,1H),8.91(s,1H),8.55(s,1H),8.11(dd,1H),7.69(d,1H),7.53(d,2H),7.20-7.44(m,3H),4.97(q,2H)4.19(s,3H).
实施例4:化合物I-4的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000074
第一步:2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺的合成
把2,4-二氯-5-硝基嘧啶(5.00g,25.8mmol)和4-(二氟甲氧基)苯胺(4.51g,28.3mmol)溶解在四氢呋喃(30mL)中,降温到-40℃,缓慢加入N,N-二异丙基乙胺(6.66g,51.5mmol),-40℃反应2小时。反应结束后加水(80mL)淬灭,升至室温,用乙酸乙酯(160mL)萃取,然后用饱和食盐水(160mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩得到2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(8.00g,黄色固体,产率98.0%)。
LC-MS,M/Z(ESI):317.2(M+H) +
第二步:N 4-(4-(二氟甲氧基)苯基)-5-硝基-N 2-(2,2,2-三氟乙基)嘧啶-2,4-二胺的合成
把2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(1.00g,3.16mmol),2,2,2-三氟乙胺(375.4mg,3.79mmol),N,N-二异丙基乙胺(1.02g,7.90mmol)和氟化铯(479.7mg,3.16mmol)加入二甲亚砜(5mL)中,置换氮气,加热到80℃,搅拌反应3小时。将反应液降至室温,倒入水(50mL)中,过滤,收集固体,干燥得到N 4-(4-(二氟甲氧基)苯基)-5-硝基-N 2-(2,2,2-三氟乙基)嘧啶-2,4-二胺(1.00g,黄色固体,产率83.5%)。
LC-MS,M/Z(ESI):380.1(M+H) +
第三步:N 4-(4-(二氟甲氧基)苯基)-N 2-(2,2,2-三氟乙基)嘧啶-2,4,5-三胺的合成
把N 4-(4-(二氟甲氧基)苯基)-5-硝基-N 2-(2,2,2-三氟乙基)嘧啶-2,4-二胺(1.00g,2.64mmol)和钯碳(0.10g,含量为10%)加入甲醇(10mL)中,置换氢气,加热到30℃,50psi压力下搅拌反应3小时。将反应液降至室温过滤,将滤液浓缩得到N 4-(4-(二氟甲氧基)苯基)-N 2-(2,2,2-三氟乙基)嘧啶-2,4,5-三胺(800mg,黄色固体,产率86.9%)。
LC-MS,M/Z(ESI):350.1(M+H) +
第四步:8-(4-(二氟甲氧基)苯基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮的合成
把N 4-(4-(二氟甲氧基)苯基)-N 2-(2,2,2-三氟乙基)嘧啶-2,4,5-三胺(700mg,2.00mmol),乙醛酸乙酯(613.8mg,3.01mmol)和醋酸(120.4mg,2.00mmol)加入乙醇(5mL)中,置换氮气,加热到80℃反应3小时。将反应液降至室温,加入水(20mL),用乙酸乙酯(20mL*3)萃取,合并有机相,用饱和食盐水(30mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩。残留物用硅胶柱纯化(石油醚:乙酸乙酯(V/V)=100:0-1:1),得到8-(4-(二氟甲氧基)苯基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(650mg,红色固体,产率83.7%)。
LC-MS,M/Z(ESI):388.1(M+H) +
第五步:6-氯-8-(4-(二氟甲氧基)苯基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮的合成
把8-(4-(二氟甲氧基)苯基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(600mg,1.55mmol)和氯代丁二酰亚胺(310.3mg,2.32mmol)加入N,N-二甲基甲酰胺(5mL)中,置换氮气,在室温下搅拌3小时。将反应液加入水(20mL),乙酸乙酯(20mL*3)萃取,合并有机相,用饱和食盐水(30mL*2)洗涤。用无水硫酸钠干燥,过滤,浓缩后得到6-氯-8-(4-(二氟甲氧基)苯基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(480mg,黄色固体,产率73.5%)。
LC-MS,M/Z(ESI):422.0(M+H) +
第六步:8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(化合物I-4)
Figure PCTCN2022083568-appb-000075
在氮气保护下将6-氯-8-(4-(二氟甲氧基)苯基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(100mg,237.1μmol),2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(91.8mg,355.7μmol),1,1-双(二苯基磷)二茂铁二氯化钯(17.4mg,23.7μmol)和碳酸钾(98.3mg,711.4μmol)溶解在二氧六环(1mL)和水(0.2mL)中,置换氮气,升温至60℃反应2小时。降至室温,将反应液滴加入水(30mL)中,用乙酸乙酯(90mL)萃取。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩。然后将固体用乙酸乙酯(20mL)在65℃下打浆,冷却过滤得到化合物8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(化合物I-4)(36.4mg,黄色固体,产率28.5%)。
LC-MS,M/Z(ESI):518.1(M+H) +
1H NMR(400MHz,DMSO-d 6):δ8.81-8.95(m,2H),8.51(s,1H),8.14-8.47(m,1H),8.09(dd,1H),7.66(d,1H),7.16-7.59(m,5H),4.19(s,4H),3.71-3.87(m,1H).
实施例5:化合物I-5的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000076
第一步:2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺的合成
把2,4-二氯-5-硝基嘧啶(5.00g,25.8mmol)和4-(二氟甲氧基)苯胺(4.51g,28.3mmol)溶解在四氢呋喃(40mL)中,降温到-40℃,缓慢加入N,N-二异丙基乙胺(6.66g,51.5mmol), -40℃反应2小时。反应结束后加水(80mL)淬灭,升至室温,用乙酸乙酯(160mL)萃取,然后用饱和食盐水(160mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩得到2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(7.8g,黄色固体,产率88.4%)。
LC-MS,M/Z(ESI):317.1(M+H) +
第二步:N 2-(环丙基甲基)-N 4-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-2,4-二胺的合成
把2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(1.00g,3.16mmol),环丙基甲胺(269.5mg,297.9μmol),N,N-二异丙基乙胺(1.02g,7.90mmol)和氟化铯(497.7mg,3.16mmol)加入二甲亚砜(5mL)中,置换氮气,加热到80℃,搅拌反应3小时。将反应液降至室温,倒入水(50mL)中,过滤,收集固体,干燥得到N 2-(环丙基甲基)-N 4-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-2,4-二胺(1.00g,黄色固体,产率90.1%)。
LC-MS,M/Z(ESI):352.1(M+H) +
第三步:N 2-(环丙基甲基)-N 4-(4-(二氟甲氧基)苯基)嘧啶-2,4,5-三胺的合成
把N 2-(环丙基甲基)-N 4-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-2,4-二胺(1.00g,2.85mmol)和钯碳(0.10g,含量为10%)加入甲醇(10mL)中,置换氢气,加热到30℃,50psi压力下搅拌反应3小时。将反应液降至室温过滤,将滤液浓缩得到N 2-(环丙基甲基)-N 4-(4-(二氟甲氧基)苯基)嘧啶-2,4,5-三胺(700mg,红色固体,产率76.5%)。
LC-MS,M/Z(ESI):322.1(M+H) +
第四步:2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮的合成
把N 2-(环丙基甲基)-N 4-(4-(二氟甲氧基)苯基)嘧啶-2,4,5-三胺(600mg,1.87mmol),乙醛酸乙酯(571.9mg,2.80mmol)和醋酸(112.1mg,1.87mmol)加入乙醇(5mL)中,置换氮气,加热到80℃反应3小时。将反应液降至室温,加入水(20mL),用乙酸乙酯(20mL*3)萃取,合并有机相,用饱和食盐水(30mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩。残留物用硅胶柱纯化(石油醚:乙酸乙酯(V/V)=100:0-1:1),得到2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(600mg,红色固体,产率89.4%)。
LC-MS,M/Z(ESI):360.1(M+H) +
第五步:6-氯-2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮的合成
把2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(550.0mg,1.53mmol)和 氯代丁二酰亚胺(306.6mg,2.30mmol)加入N,N-二甲基甲酰胺(5mL)中,置换氮气,在室温下搅拌3小时。将反应液加入水(20mL),乙酸乙酯(20mL*3)萃取,合并有机相,用饱和食盐水(30mL*2)洗涤。用无水硫酸钠干燥,过滤,浓缩得到6-氯-2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(460mg,黄色固体,产率76.3%)。
LC-MS,M/Z(ESI):394.0(M+H) +
第六步:2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(化合物I-5)
Figure PCTCN2022083568-appb-000077
在氮气保护下将6-氯-2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(100mg,253.9μmol),2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(98.3mg,380.9μmol),1,1-双(二苯基磷)二茂铁二氯化钯(18.6mg,25.4μmol)和碳酸钾(105.3mg,761.9μmol)溶解在二氧六环(1mL)和水(0.2mL)中,置换氮气,升温至60℃反应2小时。降至室温,将反应液滴加入水(30mL)中,用乙酸乙酯(90mL)萃取。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,浓缩。然后将固体用甲醇(20mL)在60℃下打浆,冷却过滤得到化合物2-((环丙基甲基)氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(74.6mg,黄色固体,产率94.3%)。
LC-MS,M/Z(ESI):490.2(M+H) +
1H NMR(400MHz,DMSO-d 6):δ8.81-8.90(m,2H),8.54(s,1H),8.14(d,1H),7.80-8.10(m,1H),7.70(d,1H),7.22-7.60(m,5H),4.24(s,3H),2.89(t,2H),0.82-1.19(m,1H),0.23-0.47(m,3H),0.00(s,1H).
实施例6:目标化合物I-6的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000078
第一步:2-氯-N-(4-氯苯基)-5-硝基嘧啶-4-胺的合成
在-40℃下,向2,4-二氯-5-硝基嘧啶(2.50g,12.9mmol)的四氢呋喃(50.0mL)溶液中加入4-氯苯胺(1.69g,13.3mmol)和N,N-二异丙基乙胺(3.33g,25.8mmol),反应在-40℃搅拌2小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(50.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩得到化合物2-氯-N-(4-氯苯基)-5-硝基嘧啶-4-胺(6.50g,22.8mmol,黄色固体,产率88.5%)。
LC-MS,M/Z(ESI):285.2(M+H) +
1H NMR(400MHz,DMSO_d 6)δ10.48(s,1H),9.14-9.19(m,1H),7.54-7.60(m,2H),7.48-7.53(m,2H).
第二步:N 4-(4-氯苯基)-N 2-环丙基-5-硝基嘧啶-2,4-二胺的合成
室温下,向2-氯-N-(4-氯苯基)-5-硝基嘧啶-4-胺(2.50g,8.77mmol)的二甲基亚砜(30.0mL)溶液中加入环丙胺(601mg,10.5mmol),N,N-二异丙基乙胺(2.83g,21.9mmol)和氟化铯(1.33g,8.77mmol),然后在80℃搅拌3小时。反应完成后,将反应混合物用水(150mL)稀释,有固体析出,反应液过滤,固体干燥,得到N 4-(4-氯苯基)-N 2-环丙基-5-硝基嘧啶-2,4-二胺(2.80g黄色固体)。直接用于下一步。
LC-MS,M/Z(ESI):306.1(M+H) +
第三步:N 4-(4-氯苯基)-N 2-环丙基嘧啶-2,4,5-三胺的合成
在氮气保护下,向N 4-(4-氯苯基)-N 2-环丙基-5-硝基嘧啶-2,4-二胺(1.50g,4.91mmol)的甲醇(20.0mL)溶液中加入铂钒碳(500mg,4.91mmol,10.0%纯度),反应于15psi的氢气压力下,在20℃搅拌10小时。反应完成后,将反应混合物过滤,滤饼用甲醇(30.0mL*2)洗涤,滤液减压浓缩,得到N 4-(4-氯苯基)-N 2-环丙基嘧啶-2,4,5-三胺(1.30g,4.71mmol,黄色固体,产率96.1%)。直接用于下一步。
LC-MS,M/Z(ESI):276.0(M+H) +
第四步:8-(4-氯苯基)-2-(环丙基氨基)蝶啶-7(8H)-酮的合成
向N 4-(4-氯苯基)-N 2-环丙基嘧啶-2,4,5-三胺(1.10g,3.99mmol)的乙醇(5.00mL)溶液中加入乙醛酸乙酯(1.06g,5.19mmol)和冰醋酸(240mg,3.99mmol),反应在氮气保护下80℃搅拌5小时。反应完成后,将反应混合物用水(30.0mL)稀释,然后用乙酸乙酯/四氢呋喃(1/1,30.0mL*4)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用甲基叔丁基醚(15.0mL)打浆,得到化合物8-(4-氯苯基)-2-(环丙基氨基)蝶啶-7(8H)-酮(1.20g黄色固体)。直接用于下一步。
LC-MS,M/Z(ESI):314.3(M+H) +
第五步:6-氯-8-(4-氯苯基)-2-(环丙基氨基)蝶啶-7(8H)-酮的合成
在氮气保护下,向8-(4-氯苯基)-2-(环丙基氨基)蝶啶-7(8H)-酮(600mg,1.91mmol)的N,N-二甲基甲酰胺(5.00mL)溶液中加入N-氯代丁二酰亚胺(380mg,2.85mmol),然后在25℃搅拌2小时。将反应混合物用水(30.0mL)稀释,有固体析出,反应液过滤,固体干燥,得到黄色固体化合物6-氯-8-(4-氯苯基)-2-(环丙基氨基)蝶啶-7(8H)-酮(450mg,1.29mmol,产率67.6%)。
LC-MS,M/Z(ESI):348.0(M+H) +
第六步:8-(4-氯苯基)-2-(环丙基氨基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-6)的合成
Figure PCTCN2022083568-appb-000079
在氮气保护下,向6-氯-8-(4-氯苯基)-2-(环丙基氨基)蝶啶-7(8H)-酮(200mg,574μmol)的1,4-二氧六环(5.00mL)和水(1.00mL)溶液中加入2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(222mg,862μmol),1,1-双(二苯基磷)二茂铁二氯化钯(21.0mg,28.7μmol)和碳酸钾(238mg,1.72mmol),反应在80℃下搅拌2小时。反应完成后,将反应混合物用水(20.0mL)稀释,有固体析出,反应液过滤,固体干燥,残留物用甲醇(10.0mL)打浆,然后用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex luna C18 250*50mm*10um;溶剂:A=水+0.225体积%甲酸(99.0%),B=乙腈;梯度:38%-68%,19分钟),得到8-(4-氯苯基)-2-(环丙基氨基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-6)(36.8mg,81.9μmol,黄色固体,收率14.3%)。
LC-MS,M/Z(ESI):444.0(M+H) +
1H NMR(400MHz,DMSO_d6)δ8.70-8.95(m,2H),8.49(s,1H),8.05-8.11(m,1H),7.57-7.86(m,4H),7.43-7.55(m,2H),4.18(s,3H),2.73-2.96(m,1H),0.38-0.68(m,4H).
实施例7:化合物I-7的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000080
Figure PCTCN2022083568-appb-000081
第一步:2-(二氟甲氧基)-5-硝基吡啶(1a-2)的合成
Figure PCTCN2022083568-appb-000082
在0℃下,向5-硝基吡啶-2-酚(1a-1)(3.50g,25.0mmol)的乙腈(30.0mL)溶液中加入碳酸钠(5.30g,50.0mmol)和2,2-二氟-2-(氟磺酰)乙酸(6.67g,37.5mmol),反应在30℃下搅拌10小时。反应完成后,将反应混合物用饱和碳酸氢钠水溶液(150mL)稀释,然后用乙酸乙酯(100mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=1:0至50:1)得到2-(二氟甲氧基)-5-硝基吡啶(1a-2)(3.10g,16.3mmol,黄色油状物,收率32.6%)。
1H NMR(400MHz,DMSO_d 6)δ9.15(d,1H),8.69(dd,1H),7.61-8.03(m,1H),7.35(d,1H).
第二步:6-(二氟甲氧基)吡啶-3-胺(1a)的合成
Figure PCTCN2022083568-appb-000083
在氮气保护下,向2-(二氟甲氧基)-5-硝基吡啶(1a-2)(2.00g,10.5mmol)的甲醇(20.0mL)溶液中加入铂钒碳(500mg,10%纯度),反应于15psi的氢气压力下,在30℃搅拌10小时。反应完成后,将反应混合物过滤,滤饼用甲醇(50.0mL*2)洗涤,滤液减压浓缩,得到6-(二氟甲氧基)吡啶-3-胺(1a)(1.65g,10.3mmol,棕色油状物,产率97.9%)。直接用于下一步。
LC-MS,M/Z(ESI):161.0(M+H) +
Figure PCTCN2022083568-appb-000084
第三步:2-氯-N-(6-(二氟甲氧基)吡啶-3-基)-5-硝基嘧啶-4-胺的合成
Figure PCTCN2022083568-appb-000085
在-40℃下,向2,4-二氯-5-硝基嘧啶(2.70g,13.9mmol)的四氢呋喃(30.0mL)溶液中加入6-(二氟甲氧基)吡啶-3-胺(2.23g,13.9mmol)和N,N-二异丙基乙胺(3.60g,27.8mmol),反应在-40℃搅拌2小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(50.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩得到化合物2-氯-N-(6-(二氟甲氧基)吡啶-3-基)-5-硝基嘧啶-4-胺(4.80g,棕色固体粗品)。直接用于下一步。
LC-MS,M/Z(ESI):318.0(M+H) +
1H NMR(400MHz,DMSO_d 6)δ10.58(s,1H),9.15-9.23(m,1H),8.38(d,1H),8.04(dd,1H),7.49-7.95(m,1H),7.20(d,1H).
第四步:N 2-环丙基-N 4-(6-(二氟甲氧基)吡啶-3-基)-5-硝基嘧啶-2,4-二胺的合成
氮气保护下,向2-氯-N-(6-(二氟甲氧基)吡啶-3-基)-5-硝基嘧啶-4-胺(2.50g,7.87mmol)的二甲基亚砜(20.0mL)溶液中加入环丙胺(539mg,9.44mmol),N,N-二异丙基乙胺(2.54g,19.7mmol)和氟化铯(1.20g,7.87mmol),然后在80℃搅拌5小时。反应完成后,将反应混合物用水(150mL)稀释,有固体析出,反应液过滤,固体干燥,然后用甲基叔丁基醚(30.0mL)打浆,得到N 2-环丙基-N 4-(6-(二氟甲氧基)吡啶-3-基)-5-硝基嘧啶-2,4-二胺(2.30g,6.80mmol,黄色固体,产率86.4%)。
LC-MS,M/Z(ESI):339.3(M+H) +
1H NMR(400MHz,DMSO_d 6)δ10.37(s,1H),8.96(s,1H),8.70(d,1H),8.64(d,1H),8.42(dd,1H),7.47-7.91(m,1H),7.08-7.17(m,1H),2.66(td,1H),0.67-0.76(m,2H),0.48-0.61(m,2H).
第五步:N 2-环丙基-N 4-(6-(二氟甲氧基)吡啶-3-基)嘧啶-2,4,5-三胺的合成
在氮气保护下,向N 2-环丙基-N 4-(6-(二氟甲氧基)吡啶-3-基)-5-硝基嘧啶-2,4-二胺(2.30g,6.80mmol)的甲醇(20.0mL)溶液中加入铂钒碳(500mg,10%纯度),反应于15psi的氢气压力下,在20℃搅拌10小时。反应完成后,将反应混合物过滤,滤饼用甲醇(30.0mL*2)洗涤,滤液减压浓缩,得到N 2-环丙基-N 4-(6-(二氟甲氧基)吡啶-3-基)嘧啶-2,4,5-三胺(1.90g,6.16mmol,黄色固体,产率90.6%)。直接用于下一步。
LC-MS,M/Z(ESI):309.1(M+H) +
第六步:2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)蝶啶-7(8H)-酮的合成
向N 2-环丙基-N 4-(6-(二氟甲氧基)吡啶-3-基)嘧啶-2,4,5-三胺(900mg,2.92mmol)的乙醇(10.0mL)溶液中加入乙醛酸乙酯(775mg,3.80mmol)和冰醋酸(175mg,2.92mmol),反 应在氮气保护下70℃搅拌2小时。反应完成后,将反应混合物用水(60.0mL)稀释,然后用乙酸乙酯/四氢呋喃(1/1,50.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用甲基叔丁基醚(20.0mL)打浆,得到化合物2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)蝶啶-7(8H)-酮(900mg,2.60mmol,黄色固体,产率89.0%)。直接用于下一步。
LC-MS,M/Z(ESI):347.1(M+H) +
第七步:6-氯-2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)蝶啶-7(8H)-酮的合成
在氮气保护下,向2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)蝶啶-7(8H)-酮(300mg,866μmol)的N,N-二甲基甲酰胺(2.00mL)溶液中加入N-氯代丁二酰亚胺(174mg,1.30mmol),然后在60℃搅拌4小时。将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,用水(20.0mL*3)洗,无水硫酸钠干燥,过滤,浓缩,得到化合物6-氯-2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)蝶啶-7(8H)-酮(330mg黄色固体)。直接用于下一步。
LC-MS,M/Z(ESI):381.0(M+H) +
第八步:2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-7)的合成
Figure PCTCN2022083568-appb-000086
在氮气保护下,向6-氯-2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)蝶啶-7(8H)-酮(330mg,867μmol)的二氧六环(5.00mL)和水(1.00mL)溶液中加入2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(336mg,1.30mmol),1,1-双(二苯基磷)二茂铁二氯化钯(63.4mg,86.7μmol)和碳酸钾(359mg,2.60mmol),反应在70℃下搅拌2小时。反应完成后,将反应混合物用水(30.0mL)稀释,有固体析出,反应液过滤,固体干燥,用乙酸乙酯(10.0mL)打浆,然后用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex Luna C18 150*25mm*10um;溶剂:A=水+0.225体积%甲酸(99.0%),B=乙腈;梯度:58%-88%,10分钟),得到2-(环丙基氨基)-8-(6-(二氟甲氧基)吡啶-3-基)-6-(2-甲基-2H-吲唑-5-基)蝶啶- 7(8H)-酮(I-7)(125mg,262μmol,黄色固体,收率30.2%)。
LC-MS,M/Z(ESI):477.1(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.82(br s,2H),8.49(s,1H),8.32-8.43(m,1H),7.96-8.23(m,3H),7.76-7.90(m,1H),7.65(d,1H),7.32(d,1H),4.18(s,3H),2.69-3.13(m,1H),0.40-0.69(m,4H).
实施例8:化合物I-8的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000087
第一步:2-氯-N-(4-环丙基苯基)-5-硝基嘧啶-4-胺的合成
把2,4-二氯-5-硝基嘧啶(1.19g,6.14mmol)和4-环丙基苯胺(900mg,6.76mmol)溶解在四氢呋喃(20.0mL)中,降温到-40℃,缓慢加入N,N-二异丙基乙胺(1.59g,12.2mmol),-40℃反应2小时。反应结束后加水(10.0mL)淬灭,升至室温,用乙酸乙酯(45.0mL)萃取,然后用饱和食盐水(20.0mL)洗涤,再用无水硫酸钠干燥,过滤,浓缩,得到粗产物2-氯-N-(4-环丙基苯基)-5-硝基嘧啶-4-胺(1.60g,黄色固体粗品)。
1H NMR(400MHz,CDCl 3)δ10.17(br s,1H),9.17(s,1H),7.48-7.51(m,2H),7.13-7.16(m,2H),1.91-1.98(m,1H),1.00–1.11(m,2H),0.72-0.75(m,2H).
第二步:N 2-环丙基-N 4-(4-环丙基苯基)-5-硝基嘧啶-2,4-二胺的合成
把2-氯-N-(4-环丙基苯基)-5-硝基嘧啶-4-胺(1.60g,粗品),环丙胺(377mg,6.60mmol),N,N-二异丙基乙胺(1.78g,13.7mmol)和氟化铯(836g,5.50mmol)溶解于二甲亚砜(10.0mL)中,置换氮气,加热到80℃,搅拌3小时。将反应液降温至室温,倒入水(30.0mL)中,过滤,收集固体。得到N 2-环丙基-N 4-(4-环丙基苯基)-5-硝基嘧啶-2,4-二胺(1.60g,黄色固体,产率93.3%)。
LC-MS,M/Z(ESI):312.2(M+H) +
第三步:N 2-环丙基-N 4-(4-环丙基苯基)嘧啶-2,4,5-三胺的合成
把N 2-环丙基-N 4-(4-环丙基苯基)-5-硝基嘧啶-2,4-二胺(1.60g,5.14mmol)和铂钒碳(160mg,10.0%含量)加入甲醇(16.0mL)中,置换氢气,加热到30℃,在30℃,50psi搅拌3小时。将反应液降温至室温过滤,将滤液旋干。得到N 2-环丙基-N 4-(4-环丙基苯基)嘧啶-2,4,5-三胺(1.35g,棕褐色固体,产率93.3%)。
LC-MS,M/Z(ESI):282.1(M+H) +
第四步:2-(环丙基氨基)-8-(4-环丙基苯基)蝶啶-7(8H)-酮的合成
把N 2-环丙基-N 4-(4-环丙基苯基)嘧啶-2,4,5-三胺(1.20g,4.27mmol),乙醛酸乙酯(1.13g,5.54mmol)和醋酸(256mg,4.27mmol)加入乙醇(12.0mL)中,置换氮气,加热到80℃,在80℃搅拌3小时。将反应液降至室温,加入水(20.0mL),用乙酸乙酯(45.0mL*3)萃取,合并有机相,用饱和食盐水(30.0mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩。残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=100:1至1:1),浓缩馏分,得到2-(环丙基氨基)-8-(4-环丙基苯基)蝶啶-7(8H)-酮(360mg,黄色固体,产率26.4%)。
LC-MS,M/Z(ESI):320.1(M+H) +
1H NMR(400MHz,DMSO-d6)δ8.67-8.86(m,1H),7.93(s,1H),7.34-7.12(m,4H),2.53(d,1H),1.81-2.09(m,1H),0.97-1.13(m,2H),0.69-0.85(m,2H),0.63(br s,1H),0.32-0.58(m,3H).
第五步:6-氯-2-(环丙基氨基)-8-(4-环丙基苯基)蝶啶-7(8H)-酮的合成
把2-(环丙基氨基)-8-(4-环丙基苯基)蝶啶-7(8H)-酮(400mg,1.16mmol)和氯代丁二酰亚胺(232mg,1.74mmol)加入N,N-二甲基甲酰胺(10.0mL)中,置换氮气,在室温下搅拌3小时。将反应液加入水(30.0mL),用乙酸乙酯(90.0mL*3)萃取,合并有机相,用饱和食盐水(30.0mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩。残留物用硅胶柱分离纯化(石油醚: 乙酸乙酯(V/V)=100:1至3:1),浓缩馏分,得到6-氯-2-(环丙基氨基)-8-(4-环丙基苯基)蝶啶-7(8H)-酮(400mg,黄色固体,产率90.9%)。
LC-MS,M/Z(ESI):354.0(M+H) +
第六步:2-(环丙基氨基)-8-(4-环丙基苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(化合物I-8)
Figure PCTCN2022083568-appb-000088
将6-氯-2-(环丙基氨基)-8-(4-环丙基苯基)蝶啶-7(8H)-酮(160mg,452μmol),2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)吲唑(140mg,542μmol),1,1-双(二苯基磷)二茂铁二氯化钯(18.4mg,22.6μmol)和碳酸钾(187mg,1.36mmol)溶解在二氧六环(2.00mL)和水(0.20mL)中从,置换氮气,升温至60℃,在氮气保护下搅拌2小时。降至室温,将反应液滴加入水(10.0mL)中,用乙酸乙酯(20.0mL)萃取,有机相用饱和食盐水(15.0mL)洗涤,无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex Luna C18 150*25mm*10um;溶剂:A=水+0.225体积%甲酸(99%),B=乙腈;梯度:44%-74%,10分钟),得到化合物2-(环丙基氨基)-8-(4-环丙基苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-8)(18.4mg,黄色固体,产率8.54%)。
LC-MS,M/Z(ESI):450.2(M+H) +
1H NMR(400MHz,DMSO-d6)δ8.83-8.87(m,1H),8.80(s,1H),8.48(s,1H),8.07(dd,1H),7.69-7.78(m,1H),7.64(d,1H),7.17-7.31(m,4H),4.18(s,3H),1.95-2.08(m,1H),1.23(s,1H),0.98-1.07(m,2H),0.70-0.78(m,2H),0.37-0.67(m,4H).
实施例9:化合物I-9的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000089
第一步:(2-甲基咪唑并[1,2-a]吡啶-6-基)硼酸(6a)的合成
Figure PCTCN2022083568-appb-000090
将6-溴-2-甲基咪唑并[1,2-a]吡啶(1.00g,4.74mmol),双联频哪醇硼酸酯(1.80g,7.11mmol),1,1-双(二苯基磷)二茂铁二氯化钯(173mg,236.9μmol)和醋酸钾(1.40g,14.2mmol)溶解在二氧六环(20mL)中,置换氮气,在90℃下搅拌1小时。降至室温,将反应液过滤,母液旋干,得到粗品。将得到的粗品用石油醚:乙酸乙酯(10:1,20mL)打浆,过滤后得到(2-甲基咪唑并[1,2-a]吡啶-6-基)硼酸(6a)(1.10g,产率89.9%)。
LC-MS,M/Z(ESI):177.0(M+H) +
第二步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基咪唑并[1,2-a]吡啶-6-基)蝶啶-7(8H)-酮的合成(I-9)
Figure PCTCN2022083568-appb-000091
将6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(100mg,263.3μmol),(2-甲基咪唑并[1,2-a]吡啶-6-基)硼酸(101.9mg,395.0μmol),1,1-双(二苯基磷)二茂铁二氯化钯(10.8mg,13.2μmol)和碳酸钾(109.2mg,790.0μmol)溶解在二氧六环(2mL)和水(0.4mL)中,置换氮气,升温至90℃,在氮气保护下搅拌2小时。降至室温,将反应液滴加入水(10 mL)中,用乙酸乙酯(30mL)萃取。有机相用饱和食盐水(30mL)洗两次,无水硫酸钠干燥,浓缩,残留物通过反相高效液相色谱法进行分离,分离方法为(柱子:3_Phenomenex Luna C18 75*30mm*3μm;溶剂:A=水+0.225%体积甲酸(99%),B=乙腈;梯度:15%-45%,10分钟),冻干后得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基咪唑并[1,2-a]吡啶-6-基)蝶啶-7(8H)-酮(45mg,黄色固体,产率35.8%)。
LC-MS,M/Z(ESI):476.2(M+H) +
1H NMR(400MHz,DMSO-d 6)δ9.64(s,1H),8.91(d,1H),8.03(d,1H),7.17-7.89(m,8H),3.28-3.32(m,1H),2.33(s,3H),0.39-0.65(m,4H).
实施例10:化合物I-10的制备
2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基苯并[d]噻唑-6-基)蝶啶-7(8H)-酮(I-10)
Figure PCTCN2022083568-appb-000092
化合物I-10的合成路线参考I-1的合成方法,得到2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基苯并[d]噻唑-6-基)蝶啶-7(8H)-酮(I-10)
LC-MS,M/Z(ESI):493.1[M+H] +
实施例11:化合物I-11的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000093
第一步:5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑的合成
在室温下,将5-溴-2H-吲唑(2.00g,10.1mmol)溶解在N,N-二甲基甲酰胺(20.0mL),加入双联频哪醇硼酸酯(3.87g,15.2mmol),醋酸钾(2.99g,30.4mmol)和1,1-双(二苯基磷)二茂铁二氯化钯二氯甲烷混合物(828mg,1.02mmol),充分搅拌,在100℃反应12小时。反应结束后加水(30.0mL)淬灭,用乙酸乙酯(90.0mL)萃取,然后用饱和食盐水(90.0mL)洗涤,有机相用无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=5:1)得到5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(1.64g,棕褐色油状物,产率66.1%)。
LC-MS,M/Z(ESI):244.9(M+H) +
第二步:2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇的合成
Figure PCTCN2022083568-appb-000094
把5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(1.64g,6.72mmol)溶解于N,N-二甲基甲酰胺(16.0mL)中,加入2,2-二甲基环氧乙烷(1.21g,16.8mmol),碳酸钾(1.39g,10.0mmol),置换氮气,在100℃搅拌2小时。反应结束后加水(10.0mL)淬灭,用乙酸乙酯(30.0mL)萃取,然后用饱和食盐水(30.0mL)洗涤,有机相用无水硫酸钠干燥,过滤, 浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Welch Ultimate XB-SiOH 250*50*10μm;溶剂:A=环己烷,B=乙醇;梯度:1%-20%,15分钟),得到物2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇(186mg,无色透明油状,产率2.12%)。
LC-MS,M/Z(ESI):317.1(M+H) +
第三步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-羟基-2-甲基丙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-11)的合成
Figure PCTCN2022083568-appb-000095
2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇(135mg,426μmol)溶解于1,4-二氧六环(4.00mL)中,将6-氯-2-(环丙基氨基)-8-[4-(二氟甲氧基)苯基]蝶啶-7-酮(147mg,388μmol),碳酸钾(160mg,1.16mmol)的水溶液(1.00mL),1,1-双(二苯基磷)二茂铁二氯化钯二氯甲烷混合物(15.8mg,19.4μmol),充分搅拌,在25℃反应12小时。反应结束后加水(15.0mL)淬灭,用乙酸乙酯(30.0mL)萃取,然后用饱和食盐水(30.0mL)洗涤,有机相用无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Waters Xbridge 150*25mm*5μm;溶剂:A=水+0.225体积%甲酸(99%),B=乙腈;梯度:38%-68%,7分钟),得到2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-羟基-2-甲基丙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-11)(75.0mg,黄色固体,产率34.8%)。
LC-MS,M/Z(ESI):534.2(M+H) +
1H NMR(400MHz,CDCl 3)δ8.98(s,1H),8.83-8.95(m,1H),8.26(d,1H),8.02(s,1H),7.78(d,1H),7.30-7.38(m,4H),6.40-6.80(m,1H),5.34-5.63(m,1H),4.37(s,2H),4.25-4.35(m,1H),1.21(s,7H),0.41-0.60(m,4H)
.
实施例12:化合物I-12的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000096
第一步:2-(环丙基甲氧基)-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺的合成
在氮气保护下,向2-氯-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(1.00g,3.16mmol)的二甲基亚砜(10.0mL)溶液中加入环丙基甲醇(600mg,8.32mmol)和N,N-二异丙基乙胺(1.04g,8.04mmol)和氟化铯(480mg,3.16mmol,),反应在30℃搅拌10小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(30.0mL*3)萃取,合并有机层,用水(30.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=30:1至15:1)得到化合物2-(环丙基甲氧基)-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(500mg,1.42mmol,黄色固体,收率44.9%)。
LC-MS,M/Z(ESI):353.0(M+H) +
1H NMR(400MHz,CDCl 3)δ10.27(s,1H),9.24(s,1H),7.58-7.66(m,2H),7.17-7.24(m,2H),6.33-6.75(m,1H),4.21(d,2H),1.22-1.39(m,1H),0.60-0.68(m,2H),0.30-0.42(m,2H).
第二步:2-(环丙基甲氧基)-N 4-(4-(二氟甲氧基)苯基)嘧啶-4,5-二胺的合成
在氮气保护下,向2-(环丙基甲氧基)-N-(4-(二氟甲氧基)苯基)-5-硝基嘧啶-4-胺(500mg,1.42mmol)的甲醇(10.0mL)溶液中加入铂钒碳(500mg,192μmol,含量为10%),然后用氢气将反应体系置换3次,反应在15psi的氢气压力下,15℃搅拌10小时。反应完成后,将 反应液过滤,用甲醇(15.0mL*2)洗涤,反应液浓缩,得到2-(环丙基甲氧基)-N 4-(4-(二氟甲氧基)苯基)嘧啶-4,5-二胺(470mg黄色固体)。直接用于下一步。
LC-MS,M/Z(ESI):323.1(M+H) +
第三步:2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮的合成
氮气保护下,向2-(环丙基甲氧基)-N 4-(4-(二氟甲氧基)苯基)嘧啶-4,5-二胺(450mg,1.40mmol)的乙醇(5.00mL)溶液中加入乙醛酸乙酯的甲苯溶液(371mg,1.82mmol,50.0%浓度)和冰醋酸(83.8mg,1.40mmol),反应在90℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩得到化合物2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(450mg,1.25mmol,黄色固体,收率89.5%)。
LC-MS,M/Z(ESI):361.1(M+H) +
第四步:6-氯-2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮的合成
向2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(350mg,971μmol)的N,N-二甲基甲酰胺(1.00mL)溶液中加入N-氯代丁二酰亚胺(195mg,1.46mmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(10.0mL)稀释,然后用乙酸乙酯(10.0mL*3)萃取,合并有机层,用水(10.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩,得到6-氯-2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(300mg黄色油状化合物)。直接用于下一步。
LC-MS,M/Z(ESI):394.8(M+H) +
第五步:2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-12)的合成
Figure PCTCN2022083568-appb-000097
在氮气保护下,向6-氯-2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(200mg, 507μmol)的1,4-二氧六环(1.00mL)和水(200μL)的溶液中加入2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(196mg,760μmol),碳酸钾(210mg,1.52mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(37.1mg,50.7μmol),反应在75℃搅拌3小时。反应完成后,将反应混合物减压浓缩,残留物用硅胶板分离纯化(石油醚:乙酸乙酯(V/V)=1:2),然后用甲醇(3.00mL)打浆,得到化合物2-(环丙基甲氧基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-12)(3.67mg,7.04μmol,黄色固体,收率1.39%)。
LC-MS,M/Z(ESI):491.1(M+H) +
1H NMR(400MHz,CDCl 3)δ8.98-9.13(m,2H),8.28(br d,1H),8.01(s,1H),7.78(d,1H),7.33-7.43(m,4H),6.38-6.87(m,1H),4.26(s,3H),4.08(d,2H),1.18-1.29(m,1H),0.48-0.65(m,2H),0.18-0.37(m,2H).
实施例13:化合物I-13的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000098
在氮气保护下,向6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(200mg,527μmol)的N,N-二甲基甲酰胺(2.00mL)溶液中加入2-甲基-4,5,6,7-四氢-2H-吡唑并[4,3-c]吡啶盐酸盐(133mg,632μmol)和碳酸铯(515mg,1.58mmol),反应在50℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,用水(20.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex Gemini-NX C18 75*30mm*3μm;溶剂:A=水+0.225体积%甲酸(99.0%),B=乙腈;梯度:35%-65%,7分钟),然后再用甲醇(5.00mL)打浆,得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-6,7-二氢-2H-吡唑并[4,3-c]吡啶-5(4H)-基)蝶啶-7(8H)-酮(I-13)(43.1mg,85.3μmol,黄色固体,收率16.2%)。
LC-MS,M/Z(ESI):481.1(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.45(s,1H),7.47-7.57(m,1H),7.38-7.46(m,2H),7.05-7.38(m,4H),4.63(s,2H),3.93(t,2H),3.75(s,3H),3.41-3.52(m,1H),2.69-2.82(m,2H),0.22-0.66(m,4H).
实施例14:化合物I-14的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000099
第一步:3-氨基-2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-甲酸叔丁酯的合成
在室温下,将3-氰基-4-氧代吡咯烷-1-甲酸叔丁酯(3.00g,14.2mmol)溶解在乙醇(30.0mL),加入甲基肼(788mg,17.1mmol)充分搅拌,在85℃反应17小时。反应结束后直接过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex luna C18150*40mm*15μm;溶剂:A=水+0.225体积%氨水(30%),B=乙腈;梯度:25%-55%,10分钟),得到黄色固体3-氨基-2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-甲酸叔丁酯(1.90g,产率55.8%)。
LC-MS,M/Z(ESI):239.2(M+H) +
第二步:2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-甲酸叔丁酯的合成
把3-氨基-2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-甲酸叔丁酯(1.90g,7.97mmol)溶解于1,4-二氧六环(20.0mL)中,加入亚硝酸异戊酯(0.87g,15.9mmol),置换氮气,在100℃搅拌2小时。反应结束后加水(10.0mL)淬灭,用乙酸乙酯(60.0mL)萃取,然后用饱和食盐水(60.0mL)洗涤,有机相用无水硫酸钠干燥,过滤,浓缩得到黄色油状物2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-甲酸叔丁酯(1.20g,产率67.4%)。
LC-MS,M/Z(ESI):224.5(M+H) +
第三步:2-甲基-2,4,5,6-四氢吡咯并[3,4-c]吡唑的合成
2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-甲酸叔丁酯(1.20g,5.37mmol)溶解于二氯甲烷(12.0mL)中,加入三氟乙酸(30.6g,268mmol),充分搅拌,在25℃反应12小时。反应结束后,浓缩,得到粗产物黄色固体2-甲基-2,4,5,6-四氢吡咯并[3,4-c]吡唑(435mg,产率65.7%)。
LC-MS,M/Z(ESI):124.1(M+H) +
1H NMR(400MHz,DMSO-d6)δ7.58(s,1H),4.27(t,4H),3.84(s,3H).
第四步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-基)蝶啶-7(8H)-酮(I-14)的合成
Figure PCTCN2022083568-appb-000100
2-甲基-2,4,5,6-四氢吡咯并[3,4-c]吡唑(50.0mg,405μmol)溶解于二甲基亚砜(1.00mL)中,加入二异丙基乙胺((262mg,2.03mmol),6-氯-2-(环丙基氨基)-8-[4-(二氟甲氧基)苯基]蝶啶-7(8H)-酮(154mg,405μmol),置换氮气,充分搅拌,在95℃反应2小时。反应结束后加水(15.0mL)淬灭,过滤,收集固体,然后把固体在60℃甲醇溶液中重结晶,得到2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-甲基-2,6-二氢吡咯并[3,4-c]吡唑-5(4H)-基)蝶啶-7(8H)-酮(I-14)(40.0mg,黄色固体,产率20.0%)。
LC-MS,M/Z(ESI):467.1(M+H) +
1H NMR(400MHz,DMSO-d6)δ8.41(s,1H),7.52-7.56(m,1H),7.43(d,2H),7.30-7.35(m,3H),7.11-7.17(m,1H),4.86(s,4H),3.84(s,3H),3.30(s,1H),0.48(d,2H),0.36(s,2H).
实施例15:化合物I-15的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000101
第一步:2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲唑-2-基)丙烷-2-醇的合成
把5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(1.64g,6.72mmol)溶解于N,N-二甲基甲酰胺(16.0mL)中,加入2,2-二甲基环氧乙烷(1.21g,16.8mmol),碳酸钾(1.39g,10.0mmol),置换氮气,在100℃搅拌2小时。反应结束后加水(10.0mL)淬灭,用乙酸乙酯(30.0mL)萃取,然后用饱和食盐水(30.0mL)洗涤,有机相用无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Welch Ultimate XB-SiOH 250*50*10um;溶剂:A=环己烷,B=乙醇;梯度:1%-20%,15分钟),纯化得到2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲唑-2-基)丙烷-2-醇(450mg,无色透明油状物,产率14.3%)。
LC-MS,M/Z(ESI):317.1(M+H) +
第二步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(2-羟基-2-甲基丙基)-1H-吲唑-5-基)蝶啶-7(8H)-酮(I-15)的合成
Figure PCTCN2022083568-appb-000102
2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲唑-2-基)丙烷-2-醇(223mg,706μmol)溶解于1,4二氧六环(4.00mL)中,将6-氯-2-(环丙基氨基)-8-[4-(二氟甲氧基)苯基]蝶啶-7(8H)-酮(244mg,642μmol),碳酸钾(266mg,1.93mmol)的水溶液(1.00mL),1,1-双(二苯基磷)二茂铁二氯化钯二氯甲烷混合物(26.2mg,32.1μmol),充分搅拌,在25℃反应 12小时。反应结束后加水(15.0mL)淬灭,用乙酸乙酯(30.0mL)萃取,然后用饱和食盐水(30.0mL)洗涤,有机相用无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex Gemini-NX C18 75*30mm*3um;溶剂:A=水+0.225体积%甲酸(99%),B=乙腈;梯度:40%-70%,7分钟),得到2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(2-羟基-2-甲基丙基)-1H-吲唑-5-基)蝶啶-7(8H)-酮(I-15)(88.0mg,黄色固体,产率24.6%)。
LC-MS,M/Z(ESI):534.1(M+H) +
1H NMR(400MHz,CDCl 3)δ8.85-8.94(m,2H),8.36(dd,1H),8.12(s,1H),7.51(dd,1H),7.34(s,4H),6.42-6.80(m,1H),5.43-5.75(m,1H),4.36(s,2H),3.57-3.64(m,1H),1.23(s,6H),0.45-0.85(m,4H).
实施例16:化合物I-16的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000103
第一步:(Z)-3-((二甲氨基)亚甲基)-4-氧代吡咯烷-1-甲酸叔丁酯的合成
在室温下,将3-氧代吡咯烷-1-甲酸叔丁酯(4.00g,21.6mmol)溶解在四氢呋喃(10.0mL),加入N,N-二甲基甲酰胺二甲基缩醛(7.72g,64.9mmol),置换氮气,充分搅拌,在70℃反应16小时。反应结束后直接浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Welch Ultimate XB-CN 250*70*10um;溶剂:A=环己烷,B=乙醇;梯度:1%-40%,15分钟),得到(Z)-3-((二甲氨基)亚甲基)-4-氧代吡咯烷-1-甲酸叔丁酯(1.40g,黄色油状物,产率26.9%)。
LC-MS,M/Z(ESI):241.2(M+H) +
1H NMR(400MHz,CDCl 3)δ7.37(s,1H),4.58(s,2H),3.83(s,2H),3.11(s,6H),1.48(s,9H).
第二步:1-甲基-4,6-二氢吡咯并[3,4-c]吡唑-5(1H)-甲酸叔丁酯的合成
(Z)-3-((二甲氨基)亚甲基)-4-氧代吡咯烷-1-甲酸叔丁酯(1.00g,4.16mmol)溶解于乙醇(10.0mL)中,加入甲基肼(230mg,4.99mmol),置换氮气,在85℃搅拌17小时。反应结束后过滤,溶解于石油醚(20.0mL)中,然后先用饱和氯化铵溶液(25.0mL)洗涤,再用饱和碳酸氢钠溶液(25.0mL)洗涤,有机相用无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex luna C18 150*40mm*15um;溶剂:A=水+0.225体积%甲酸(99%),B=乙腈;梯度:25%-55%,10分钟),得到棕1-甲基-4,6-二氢吡咯并[3,4-c]吡唑-5(1H)-甲酸叔丁酯(260mg,褐色油状物,产率27.4%)。
LC-MS,M/Z(ESI):224.1(M+H) +
1H NMR(400MHz,CDCl 3)δ7.31(d,1H),4.40-4.53(m,4H),3.88(s,3H),1.51(s,9H).
第三步:1-甲基-1,4,5,6-四氢吡咯并[3,4-c]吡唑的合成
1-甲基-4,6-二氢吡咯并[3,4-c]吡唑-5(1H)-甲酸叔丁酯(135mg,426μmol)加入三氟乙酸(2.55g,22.3mmol),充分搅拌,在25℃反应12小时。反应结束后直接浓缩,得到1-甲基-1,4,5,6-四氢吡咯并[3,4-c]吡唑(80.0mg,棕褐色油状物,粗品)。
LC-MS,M/Z(ESI):124.1(M+H) +
第四步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-甲基-4,6-二氢吡咯并[3,4-c]吡唑-5(1H)-基)蝶啶-7(8H)-酮(I-16)的合成
Figure PCTCN2022083568-appb-000104
1-甲基-1,4,5,6-四氢吡咯并[3,4-c]吡唑(80.0mg,649μmol)溶解于二甲基亚砜(1.00mL)中,加入二异丙基乙胺(419mg,3.25mmol),6-氯-2-(环丙基氨基)-8-[4-(二氟甲氧基)苯基]蝶啶-7(8H)-酮(246mg,649μmol),置换氮气,充分搅拌,在95℃反应2小时。反应结束后加水(15.0mL)淬灭,过滤,收集固体,然后把固体在60℃甲醇溶液中重结晶,得到2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-甲基-4,6-二氢吡咯并[3,4-c]吡唑-5(1H)-基)蝶啶-7(8H)-酮(I- 16)(4.08mg,黄色固体,产率1.35%)。
LC-MS,M/Z(ESI):467.1(M+H) +
1H NMR(400MHz,DMSO-d 6)δ8.41(s,1H),7.43(d,2H),7.31-7.37(m,3H),7.25(s,1H),7.16(s,1H),4.92-5.10(m,2H),4.78-4.92(m,2H),3.80(s,3H),3.30(s,1H),0.48(dd,2H),0.36(s,2H).
实施例17:目标化合物I-17的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000105
第一步:(2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2,3-二氢苯并呋喃-2-基)甲醇的合成
在室温下,将(5-溴-2-甲基-2,3-二氢苯并呋喃-2-基)甲醇(100mg,411μmol),醋酸钾(121mg,1.23mmol),双联频哪醇硼酸酯(157mg,617μmol)和1,1-双(二苯基磷)二茂铁二氯化钯(30.1mg,41.1μmol)加入到1,4-二氧六环(2.00mL)中,氮气保护下在90℃反应2小时,反应液过滤浓缩,得到化合物(2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2,3-二氢苯并呋喃-2-基)甲醇(115mg,黄色固体粗品)。
LC-MS,M/Z(ESI):291.1(M+H) +
第二步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(羟甲基)-2-甲基-2,3-二氢苯并呋喃-5-基)蝶啶-7(8H)-酮(I-17)
Figure PCTCN2022083568-appb-000106
在氮气保护下,将(2-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2,3-二氢苯并呋喃-2-基)甲醇(115mg,396μmol),碳酸钾(164mg,1.91mmol),6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(151mg,3.96mmol)1,1-双(二苯基磷)二茂铁二氯化钯(29.0mg,39.6μmol)加入到1,4-二氧六环(1.00mL)和水(0.30mL)中,氮气保护下35℃下反应12小 时,反应液过滤,浓缩得到粗品。然后粗品经高效液相色谱纯化,分离方法为(柱子:Phenomenex Synergi C18 75*30mm*4um;溶剂:A=水+0.1体积%三氟甲酸(99%),B=乙腈;梯度:46%-66%B,6分钟),得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(羟甲基)-2-甲基-2,3-二氢苯并呋喃-5-基)蝶啶-7(8H)-酮(I-17)(58.5mg,22.7%产率)。
LC-MS,M/Z(ESI):508.3(M+H) +
1H NMR(400MHz,CDCl 3)δ9.45(br,1H),8.51(s,1H),8.17-8.19(m,2H),7.30-7.34(m,4H),6.42-6.83(m,2H),3.65-3.77(m,2H),3.34-3.38(d,1H),2.96-3.00(d,1H),2.48-2.51(br,1H),1.48(s,3H),0.55-0.64(m,4H).
实施例18:化合物I-18的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000107
第一步:1-(4-溴-1H-吡唑-1-基)-2-甲基丙烷-2-醇的合成
在室温下,将4-溴吡唑(1.00g,6.80mmol),2,2-二甲基环氧乙烷(981mg,13.6mmol)和碳酸铯(2.22g,6.80mmol)加入到二氧六环(20.0mL)中,氮气保护下在80℃反应2小时,反应液过滤,浓缩得到化合物1-(4-溴-1H-吡唑-1-基)-2-甲基丙烷-2-醇(1.20g,白色固体粗品)。
LC-MS,M/Z(ESI):219.0(M+H) +
第二步:2-甲基-1-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丙烷-2-醇的合成
在室温下,将1-(4-溴-1H-吡唑-1-基)-2-甲基丙烷-2-醇(500mg,2.28mmol),醋酸钾(672mg,6.85mmol),双联频哪醇硼酸酯(580mg,2.28mmol)和1,1-双(二苯基磷)二茂铁二氯化钯 (167mg,228μmol)加入到1,4-二氧六环(10.0mL)中,氮气保护下在80℃反应2小时,反应液过滤,浓缩得到2-甲基-1-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丙烷-2-醇(500mg,黄色油状物粗品)。
LC-MS,M/Z(ESI):267.2(M+H) +
第三步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(2-羟基-2-甲基丙基)-1H-吡唑-4-基)蝶啶-7(8H)-酮(I-18)
Figure PCTCN2022083568-appb-000108
在氮气保护下,将2-甲基-1-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丙烷-2-醇(350mg,1.32mmol),碳酸钾(545mg,3.95mmol),6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(250mg,658μmol),1,1-双(二苯基磷)二茂铁二氯化钯(48.1mg,65.8μmol)加入到1,4-二氧六环(8.00mL)和水(1.00mL)中,氮气保护下25℃下反应6小时,反应液过滤浓缩得到粗品。粗品用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex Synergi C18 150*50mm*3um;溶剂:A=水+0.1体积%甲酸(99%),B=乙腈;梯度:30%-60%,15分钟),得到2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(2-羟基-2-甲基丙基)-1H-吡唑-4-基)蝶啶-7(8H)-酮(I-18)(26.1mg,黄色化合物,产率4.08%)。
LC-MS,M/Z(ESI):484.2(M+H) +
1H NMR(400MHz,CDCl 3)δ8.79(br,1H),8.48(s,1H),8.41(s,1H),7.29-7.36(m,4H),6.42-6.78(t,1H),4.11(s,2H),1.60-1.67(m,1H),1.20(s,6H),0.43-0.57(m,4H).
实施例19:目标化合物I-19的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000109
第一步:4-(4-溴-1H-吡唑-1-基)-2-甲基丁烷-2-醇的合成
把4-溴吡唑(2.00g,13.6mmol)和4-溴-2-甲基丁烷-2-醇(3.41g,20.4mmol)加入N,N-二甲基甲酰胺(10mL)中,加入碳酸钾(3.76g,27.2mmol),置换氮气,在25℃下搅拌12小时。将反应液加入水(30mL),乙酸乙酯(30mL*3)萃取,合并有机相,用饱和食盐水(45mL*2)洗涤,无水硫酸钠干燥,过滤,浓缩得到4-(4-溴-1H-吡唑-1-基)-2-甲基丁烷-2-醇(3.00g,黄色油状物,产率94.6%)。
LC-MS,M/Z(ESI):233.1(M+H) +
第二步:2-甲基-4-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丁烷-2-醇的合成
把4-(4-溴-1H-吡唑-1-基)-2-甲基丁烷-2-醇(2.80g,12.0mmol),双联频哪醇硼酸酯(4.58g,18.0mmol),1,1-双(二苯基磷)二茂铁二氯化钯(879mg,1.20μmol)和乙酸钾(2.36g,24.0mmol)加入1,4-二氧六环(20mL)中,置换氮气,在80℃下搅拌1小时。将反应液浓缩,得到2-甲基-4-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丁烷-2-醇(2.80g,黄色油状物,产率83.2%)。直接用于下一步。
LC-MS,M/Z(ESI):281.1(M+H) +
第三步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(3-羟基-3-甲基丁基)-1H-吡唑-4-基)蝶啶-7(8H)-酮(目标化合物I-19)
Figure PCTCN2022083568-appb-000110
在氮气保护下,将6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(500mg,1.32mmol),2-甲基-4-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吡唑-1-基)丁烷-2-醇(553mg,1.97mmol),1,1-双(二苯基磷)二茂铁二氯化钯(107mg,132μmol)和碳酸钾(364mg,2.63mmol)溶解在1,4-二氧六环(4mL)和水(1mL)中,置换氮气,在25℃反应2小时。将反应液滴加入水(20mL)中,用乙酸乙酯(60mL)萃取。有机相用饱和食盐水(60mL)洗涤,无水硫酸钠干燥,过滤,浓缩。残留物通过反相高效液相色谱法进行分离,分离方法为(柱子:Waters Xbridge 150*25mm*5um;溶剂:A=水+0.225体积甲酸(99%),B=乙腈;梯度:34%-64%,10分钟),冻干得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(3-羟基-3-甲基丁基)-1H-吡唑-4-基)蝶啶-7(8H)-酮(I-19)(18.0mg,黄色固体,产率2.71%)。
LC-MS,M/Z(ESI):498.2(M+H) +
1H NMR(400MHz,DMSO-d 6)δ8.62-8.91(m,1H),8.52(s,1H),8.16(s,1H),7.15-7.55(m,5H),4.47(s,1H),4.21-4.30(m,2H),2.70-2.88(m,1H),1.89-1.98(m,2H),1.12(s,6H),0.31-0.71(m,4H).
实施例20:化合物I-20的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000111
第一步:1-(5-溴-2H-吲唑-2-基)丙烷-2-醇的合成
向5-溴-2H-吲唑(1.00g,5.08mmol)的N,N-二甲基甲酰胺(10.0mL)溶液中加入1-溴丙烷-2-醇(1.31g,6.60mmol,70.0%纯度)和碳酸铯(3.31g,10.2mmol)和碘化钠(76.1mg,508μmol),反应在80℃搅拌10小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(30.0mL*3)萃取,合并有机层,用水(30.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化两次,分离方法为(柱子:Welch Ultimate XB-CN 250*70*10um;溶剂:A=正己烷,B=乙醇+0.1体积%氨水(30.0%);梯度:1%-35%,15分 钟)和(柱子:Welch Ultimate XB-SiOH 250*70*10um;溶剂:A=正己烷,B=乙醇+0.1体积%氨水(30.0%);梯度:1%-40%,20分钟),得到化合物1-(5-溴-2H-吲唑-2-基)丙烷-2-醇(500mg,1.96mmol,白色固体,收率38.6%)。
LC-MS,M/Z(ESI):255.1(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.32(d,1H),7.97(dd,1H),7.58(d,1H),7.30(dd,1H),5.00(d,1H),4.24-4.39(m,2H),4.07-4.19(m,1H),1.08(d,3H).
第二步:1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇的合成
在氮气保护下,向1-(5-溴-2H-吲唑-2-基)丙烷-2-醇(450mg,1.76mmol)的1,4-二氧六环(1.00mL)溶液中加入双联频哪醇硼酸酯(538mg,2.12mmol),醋酸钾(519mg,5.29mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(64.5mg,88.2μmol),反应在85℃搅拌10小时。反应完成后,将反应液减压浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=50:1至10:1),得到1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇(450mg,1.49mmol,灰白色固体,收率84.4%)。
LC-MS,M/Z(ESI):303.1(M+H) +
第三步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-羟基丙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-20)的合成
Figure PCTCN2022083568-appb-000112
在氮气保护下,向1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇(200mg,662μmol)的1,4-二氧六环(1.00mL)和水(200uL)的溶液中加入6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(350mg,922μmol),碳酸钾(274mg,1.99mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(24.2mg,33.1μmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(10.0mL)稀释,然后用乙酸乙酯(10.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶板分离纯化(石油醚:乙酸乙酯(V/V)=0:1),得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-羟基丙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-20)(100mg,183μmol,黄色固体,27.6%收率)。
LC-MS,M/Z(ESI):520.1(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.65-9.02(m,2H),8.46(s,1H),8.08(dd,1H),7.42-7.84(m,4H),7.12-7.40(m,3H),5.03(d,1H),4.24-4.40(m,2H),4.09-4.21(m,1H),2.52-3.00(m,1H),1.10(d,3H),0.34-0.71(m,4H).
实施例21:目标化合物I-21的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000113
第一步:2-(5-溴-2H-吲唑-2-基)丙酸甲酯的合成
向5-溴-2H-吲唑(5.00g,25.4mmol)的乙腈(50.0mL)溶液中加入2-溴丙酸甲酯(5.00g,30.0mmol)和碳酸钾(7.01g,50.8mmol),反应在25℃搅拌5小时,然后在80℃搅拌10小时。反应完成后,将反应混合物用水(100mL)稀释,然后用乙酸乙酯(100mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=30:1至10:1),得到2-(5-溴-2H-吲唑-2-基)丙酸甲酯(5.80g,20.5mmol,淡黄色油状物,收率40.4%)。
LC-MS,M/Z(ESI):282.9(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.49(s,1H),7.94-8.07(m,1H),7.60(d,1H),7.33(d,1H),5.67(q,1H),3.65(s,3H),1.79(d,3H).
第二步:2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙酸甲酯的合成
在氮气保护下,向2-(5-溴-2H-吲唑-2-基)丙酸甲酯(2.00g,7.06mmol)的1,4-二氧六环(10.0mL)溶液中加入双联频哪醇硼酸酯(2.15g,8.48mmol)、醋酸钾(2.08g,21.2mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(258mg,353μmol),反应在85℃搅拌10小时。反应完成后,将反应液减压浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=30:1至5:1),得到2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙酸甲酯(2.30g,6.97mmol,黄色油状物,收率98.6%)。
LC-MS,M/Z(ESI):331.0(M+H) +
第三步:2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-1-醇的合成
在氮气保护下,0℃下,向2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙酸甲酯(600mg,1.82mmol)的四氢呋喃(10.0mL)溶液中加入四氢铝锂的四氢呋喃溶液(2.50M,1.82mL),反应在25℃搅拌2小时。反应完成后,在0℃下,将反应液用十水硫酸钠(3.00g)淬灭,过滤,用乙酸乙酯(20.0mL)洗涤,滤液浓缩,得到2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-1-醇(320mg,1.06mmol,黄色油状物,收率58.3%)。
第四步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(1-羟基丙烷-2-基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-21)的合成
Figure PCTCN2022083568-appb-000114
在氮气保护下,向6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(120mg,316μmol)的1,4-二氧六环(5.00mL)和水(1.00mL)的溶液中加入2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-1-醇(153mg,506μmol),碳酸钾(131mg,948μmol)和1,1-双(二苯基磷)二茂铁二氯化钯(23.1mg,31.6μmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层, 无水硫酸钠干燥,过滤,浓缩,残留物用乙腈(20.0mL)和二氯甲烷(20.0mL)打浆,得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(1-羟基丙烷-2-基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-21)(45.8mg,88.1μmol,黄色固体,收率27.9%)。
LC-MS,M/Z(ESI):520.3(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.83(s,2H),8.51(s,1H),8.08(dd,1H),7.44-7.90(m,4H),7.14-7.42(m,3H),5.01(t,1H),4.61-4.75(m,1H),3.70-3.88(m,2H),2.62-2.97(m,1H),1.52(d,3H),0.28-0.76(m,4H).
实施例22:目标化合物I-22的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000115
第一步:1-(5-溴-2H-吲唑-2-基)-2-甲基丙烷-2-醇的合成
在室温下,将5-溴-2H吲唑(1.00g,5.08mmol),2,2-二甲基环氧乙烷(732mg,10.2mmol)和碳酸铯(1.65g,5.08mmol)加入到1,4-二氧六环(20.0mL)中,氮气保护下在80℃反应2小时,反应液过滤,浓缩得到粗品。然后粗品经正相制备,分离方法为(柱子:Phenomenex Synergi XB-SiOH 250*70mm*10um;溶剂:A=正己烷,B=乙醇;梯度:1%-25%,20分钟),得到化合物1-(5-溴-2H-吲唑-2-基)-2-甲基丙烷-2-醇(400mg,白色固体,收率29.3%)。
LC-MS,M/Z(ESI):269.1(M+H) +
第二步:2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)丙烷-2-醇的合成
在室温下,将1-(5-溴-2H-吲唑-2-基)-2-甲基丙烷-2-醇(350mg,1.30mmol),醋酸钾(383mg,3.90mmol),双联频哪醇硼酸酯(396mg,1.56mmol)和1,1-双(二苯基磷)二茂铁二氯化钯 (95.2mg,130μmol)加入到1,4-二氧六环(15.0mL)中,氮气保护下在80℃反应4小时,反应液过滤,浓缩得到2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吡唑-1-基)丙烷-2-醇(400mg,黄色油状化合物粗品)。
LC-MS,M/Z(ESI):317.0(M+H) +
第三步:8-(4-(二氟甲氧基)苯基)-6-(2-(2-羟基-2-甲基丙基)-2H-吲唑-5-基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(I-22)
Figure PCTCN2022083568-appb-000116
在氮气保护下,将2-甲基-1-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吡唑-1-基)丙烷-2-醇(400mg,1.27mmol),碳酸钾(525mg,3.80mmol),6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(533mg,1.27mmol)1,1-双(二苯基磷)二茂铁二氯化钯(92.6mg,127μmol)加入到1,4-二氧六环(8.00mL)和水(1.00mL)中,氮气保护下25℃下反应8小时,反应液过滤,浓缩得到粗品。然后粗品用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex Synergi C18 150*25mm*10um;溶剂:A=水+0.1体积%甲酸(99%),B=乙腈;梯度:38%-68%,10分钟),得到化合物8-(4-(二氟甲氧基)苯基)-6-(2-(2-羟基-2-甲基丙基)-2H-吲唑-5-基)-2-((2,2,2-三氟乙基)氨基)蝶啶-7(8H)-酮(I-22)(45.4mg,黄色固体,产率6.24%)。
LC-MS,M/Z(ESI):576.3(M+H) +
1H NMR(400MHz,CDCl 3)δ8.99(s,1H),8.92(s,1H),8.25-8.27(d,1H),8.03(s,1H),7.77-7.79(m,1H),7.34(s,4H),6.44-6.80(t,1H),5.47-5.86(m,1H),4.22(s,2H),4.20(s,1H),3.83(s,1H),1.21(s,6H).
实施例23:目标化合物I-23的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000117
第一步:5-溴-2-(2-(甲磺酰)乙基)-2H-吲唑的合成
向5-溴-2H-吲唑(2.00g,10.2mmol)的乙腈(20.0mL)溶液中加入1-溴-2-(甲基磺酰基)乙烷(1.90g,10.2mmol)和碳酸钾(2.81g,20.3mmol),反应在80℃搅拌10小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(50.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=15:1至5:1),得到化合物5-溴-2-(2-(甲基磺酰基)乙基)-2H-吲唑(600mg,1.98mmol,白色固体,收率19.5%)。
1H NMR(400MHz,DMSO_d 6)δ8.47(s,1H),8.00(d,1H),7.60(d,1H),7.33(dd,1H),4.88(t,2H),3.86(t,2H),2.93(s,3H).
第二步:2-(2-(甲基磺酰基)乙基)-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑的合成
在氮气保护下,向5-溴-2-(2-(甲基磺酰基)乙基)-2H-吲唑(300mg,990μmol)的1,4-二氧六环(10.0mL)溶液中加入双联频哪醇硼酸酯(302mg,1.19mmol)、醋酸钾(291mg,2.97mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(36.2mg,49.5μmol),反应在85℃搅拌10小时。反应完成后,将反应液减压浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=5:1至二氯甲烷:甲醇(V/V)=10:1),得到2-(2-(甲基磺酰基)乙基)-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(320mg,914μmol,灰白色固体,收率92.3%)。
LC-MS,M/Z(ESI):351.2(M+H) +
第三步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-(甲基磺酰基)乙基)-2H-吲唑-5-基) 蝶啶-7(8H)-酮(I-23)的合成
Figure PCTCN2022083568-appb-000118
在氮气保护下,向6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(300mg,790μmol)的1,4-二氧六环(5.00mL)和水(1.00mL)的溶液中加入2-(2-(甲基磺酰基)乙基)-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑(300mg,857μmol)(3),碳酸钾(328mg,2.37mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(57.8mg,79.0μmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(10.0mL)稀释,然后用二氯甲烷/甲醇(10/1,10.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物依次用甲醇(20.0mL)、乙腈(20.0mL)和二氯甲烷(20.0mL)打浆,得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-(甲基磺酰基)乙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-23)(94.5mg,153μmol,黄色固体,收率19.4%)。
LC-MS,M/Z(ESI):568.3(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.69-9.02(m,2H),8.62(s,1H),8.10(dd,1H),7.58-7.94(m,2H),7.13-7.57(m,5H)4.89(t,2H),3.89(t,2H),2.69-3.04(m,4H),0.27-0.87(m,4H).
实施例24:目标化合物I-24的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000119
第一步:2-(5-溴-2H-吲唑-2-基)-N,N-二甲基乙酰胺的合成
在室温下,将5-溴-2H-吲唑(1.00g,5.08mmol),N,N-二甲基-2-氯乙酰胺(925mg,7.61mmol)和碳酸铯(3.31g,10.2mmol)加入到1,4-二氧六环(15.0mL)中,氮气保护下在70℃反应8小时,反应液过滤浓缩得到粗品。然后粗品经正相制备纯化,纯化方法为(柱子:Phenomenex Synergi XB-SiOH 250*70mm*10um;溶剂:A=正己烷,B=99.9%乙醇+0.1%氨水;梯度:1%-40%,20分钟),得到化合物2-(5-溴-2H-吲唑-2-基)-N,N-二甲基乙酰胺(350mg,白色固体,收率24.4%)。
LC-MS,M/Z(ESI):282.1(M+H) +
第二步:N,N-二甲基-2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)乙酰胺的合成
在室温下,将2-(5-溴-2H-吲唑-2-基)-N,N-二甲基乙酰胺(300mg,1.06mmol),醋酸钾(313mg,3.19mmol),双联频哪醇硼酸酯(405mg,1.59mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(77.8mg,106μmol)加入到1,4-二氧六环(3.00mL)中,氮气保护下在85℃反应7小时,反应液过滤,浓缩得到化合物N,N-二甲基-2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)乙酰胺(300mg,黄色油状粗品)。
LC-MS,M/Z(ESI):330.2(M+H) +
第三步:2-(5-(2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-7-氧代-7,8-二氢蝶啶-6-基)-2H-吲唑-2-基)-N,N-二甲基乙酰胺(I-24)
Figure PCTCN2022083568-appb-000120
在氮气保护下,将N,N-二甲基-2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)乙酰胺(300mg,911μmol),碳酸钾(504mg,3.65mmol),6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(242mg,638μmol),1,1-双(二苯基磷)二茂铁二氯化钯(66.7mg,91.1μmol)加入到四氢呋喃(6.00mL)和水(1.00mL)中,氮气保护下25℃下反应8小时,反应液过滤浓缩得到粗品。然后粗品用高效液相色谱仪分离纯化,分离方法为(柱子: Phenomenex Synergi C18 150*30mm*7um;溶剂:A=水+0.1体积%盐酸(99%),B=乙腈;梯度:40%-60%,7分钟),得到化合物2-(5-(2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-7-氧代-7,8-二氢蝶啶-6-基)-2H-吲唑-2-基)-N,N-二甲基乙酰胺(I-24)(71.2mg,黄色固体,产率14.2%)。
LC-MS,M/Z(ESI):547.4(M+H) +
1H NMR(400MHz,CDCl 3)δ8.78-8.90(m,2H),8.44(s,1H),7.93-8.10(m,2H),7.63-7.65(d,1H),7.18-7.54(m,5H),5.46(s,2H),3.09(s,3H),2.87(s,3H),2.81-2.82(s,1H),0.40-0.66(m,4H).
实施例25:化合物I-25的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000121
第一步:5-溴-1-(3-甲基丁-2-烯-1-基)吡啶-2(1H)-酮的合成
在氮气保护下,0℃下,向5-溴吡啶-2(1H)-酮(2.00g,11.5mmol)的四氢呋喃(20.0mL)溶液中加入钠氢(690mg,17.3mmol,含量60.0%),反应在0℃搅拌1小时,然后加入1-溴-3-甲基丁-2-烯(2.23g,14.9mmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(50.0mL*3)萃取,合并有机层,用水(50.0mL*3)洗涤,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=10:1至3:1),得到5-溴-1-(3-甲基丁-2-烯-1-基)吡啶-2(1H)-酮(2.20g,9.09mmol,黄色油状物,收率79.1%)。
LC-MS,M/Z(ESI):242.1(M+H) +
1H NMR(400MHz,CDCl 3)δ6.99-7.89(m,2H),6.48(d,1H),5.29(br s,1H),4.51(d,2H),1.55-2.00(m,6H).
第二步:5-溴-1-(3-羟基-3-甲基丁基)吡啶-2(1H)-酮的合成
向5-溴-1-(3-甲基丁-2-烯-1-基)吡啶-2(1H)-酮(1.00g,4.13mmol)的1,4-二氧六环(20.0mL)溶液中加入硫酸溶液(7.36g,37.5mmol,50.0%浓度),反应在80℃搅拌10小时。反应完成后,0℃下,将反应混合物用氢氧化钠水溶液(4.00M,30.0mL)淬灭,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,得到5-溴-1-(3-羟基-3-甲基丁基)吡啶-2(1H)-酮(800mg,3.08mmol,灰色油状物,收率74.5%)。
LC-MS,M/Z(ESI):244.1(M-18+H) +
1H NMR(400MHz,CDCl 3)δ7.48(d,1H),7.35(dd,1H),6.49(d,1H),4.03-4.11(m,2H),1.87-1.95(m,2H),1.28(s,6H).
第三步:1-(3-羟基-3-甲基丁基)-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)吡啶-2(1H)-酮的合成
在氮气保护下,向5-溴-1-(3-羟基-3-甲基丁基)吡啶-2(1H)-酮(300mg,1.15mmol)的1,4-二氧六环(2.00mL)溶液中加入双联频哪醇硼酸酯(360mg,1.42mmol)、醋酸钾(340mg,3.46mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(42.2mg,57.7μmol),反应在85℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩得到1-(3-羟基-3-甲基丁基)-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)吡啶-2(1H)-酮(350mg棕色油状物)。直接用于下一步。
LC-MS,M/Z(ESI):308.2(M+H) +
第四步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(3-羟基-3-甲基丁基)-6-氧代-1,6-二氢吡啶-3-基)蝶啶-7(8H)-酮(I-25)的合成
Figure PCTCN2022083568-appb-000122
在氮气保护下,向6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(340mg,895 μmol)的1,4-二氧六环(4.00mL)和水(1.00mL)的溶液中加入1-(3-羟基-3-甲基丁基)-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)吡啶-2(1H)-酮(4)(340mg,1.11mmol),碳酸钾(371mg,2.68mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(65.5mg,89.5μmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(20.0mL)稀释,然后用乙酸乙酯(20.0mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用高效液相色谱仪分离纯化,分离方法为(柱子:Phenomenex luna C18 150*40mm*15um;溶剂:A=水+0.225体积%甲酸(99.0%),B=乙腈;梯度:35%-65%,10分钟),得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(1-(3-羟基-3-甲基丁基)-6-氧代-1,6-二氢吡啶-3-基)蝶啶-7(8H)-酮(I-25)(104mg,183μmol,黄色固体,收率20.4%)。
LC-MS,M/Z(ESI):525.3(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.99(br s,1H),8.68-8.89(m,1H),8.28(dd,1H),7.15-7.58(m,6H),6.53(d,1H),4.47(s,1H),3.92-4.13(m,2H),2.67-2.98(m,1H),1.61-1.91(m,2H),1.13(s,6H),0.35-0.71(m,4H).
实施例26:化合物I-26的制备
合成路线如下所示:
Figure PCTCN2022083568-appb-000123
第一步:4-(2-(5-溴-2H-吲唑-2-基)乙基)吗啉的合成
向5-溴-2H-吲唑(16.0g,81.2mmol)的乙腈(400mL)溶液中加入4-(2-氯乙基)吗啉(14.6g,97.4mmol)和碳酸钾(22.4g,162mmol),反应在80℃搅拌10小时,然后在95℃搅拌10小时。反应完成后,将反应混合物浓缩,残留物用水(300mL)稀释,然后用乙酸乙酯(300mL*3)萃取,合并有机层,无水硫酸钠干燥,过滤,浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=10:1至3:1),得到化合物4-(2-(5-溴-2H-吲唑-2-基)乙基)吗啉(7.00g,22.6 mmol,灰白色固体,收率27.8%)。
LC-MS,M/Z(ESI):310.1(M+H) +
1H NMR(400MHz,CDCl 3)δ7.97(s,1H),7.82(d,1H),7.58(d,1H),7.33(dd,1H),4.51(t,2H),3.62-3.75(m,4H),2.94(t,2H),2.42-2.57(m,4H).
第二步:4-(2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)乙基)吗啉的合成
在氮气保护下,向4-(2-(5-溴-2H-吲唑-2-基)乙基)吗啉(6.00g,19.3mmol)的1,4-二氧六环(70.0mL)溶液中加入双联频哪醇硼酸酯(5.89g,23.2mmol)、醋酸钾(5.70g,58.0mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(708mg,967μmol),反应在85℃搅拌10小时。反应完成后,将反应液减压浓缩,残留物用硅胶柱分离纯化(石油醚:乙酸乙酯(V/V)=20:1至5:1),得到4-(2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)乙基)吗啉(6.50g,18.2mmol,黄色油状物,收率94.1%)。
LC-MS,M/Z(ESI):358.2(M+H) +
第三步:2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-吗啉代乙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-26)的合成
Figure PCTCN2022083568-appb-000124
在氮气保护下,向4-(2-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2H-吲唑-2-基)乙基)吗啉(2.50g,7.00mmol)的1,4-二氧六环(30.0mL)和水(5.00mL)的溶液中加入6-氯-2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)蝶啶-7(8H)-酮(2.66g,7.00mmol),碳酸钾(2.90g,21.0mmol)和1,1-双(二苯基磷)二茂铁二氯化钯(256mg,350μmol),反应在25℃搅拌10小时。反应完成后,将反应混合物用水(50.0mL)稀释,然后用乙酸乙酯(50.0mL*3)萃取,合并有机层,用盐水(50.0mL*2)洗涤,干燥,残留物用乙酸乙酯/甲醇/二氯甲烷(5/1/1,70.0mL)打浆,得到化合物2-(环丙基氨基)-8-(4-(二氟甲氧基)苯基)-6-(2-(2-吗啉代乙基)-2H-吲唑-5-基)蝶啶-7(8H)-酮(I-26)(1.97g,3.29mmol,黄色固体,收率47.0%)。
LC-MS,M/Z(ESI):575.4(M+H) +
1H NMR(400MHz,DMSO_d 6)δ8.60-9.07(m,2H),8.54(s,1H),8.08(dd,1H),7.15-7.89(m,7H),4.55(t,2H),3.52-3.54(m,4H),2.65-3.01(m,3H),2.44(br s,4H),0.35-0.73(m,4H).
本发明测试例中的对照化合物的制备参考专利WO2019/191470A1得到,结构如下所示:
Figure PCTCN2022083568-appb-000125
对照化合物
测试例1:MAT2A酶活性抑制试验
采用BPS Bioscience MAT2A抑制剂筛选试剂盒,检测化合物对MAT2A酶活性抑制的IC 50,首先用DMSO溶解待测化合物,所有化合物在DMSO稀释至起始浓度1mM,3倍稀释,10个浓度梯度。用Echo 550向反应板(784075,Greiner)每孔转移200nL稀释好的化合物,用封板膜封板,1000g离心1分钟,DMSO终浓度为1%。用1X的酶反应缓冲液配制准备2X MAT2A酶液,向384-反应板(Corning3702)中每孔加入10μL 2X MAT2A酶液,用封板膜封板,1000g离心60秒,室温孵育10分钟。用1X MAT2A激酶反应缓冲液配制2X L-Methionine和ATP混合液,向384-反应板中每孔加入10μL 2X L-Methionine和ATP混合液,用封板膜封住板,总反应体系为20μL。1000g离心60秒,室温孵育60分钟。每孔加入20μL检测缓冲液Colorimetric Detection Reagent,1000g离心30秒,室温反应15分钟。用Envision 2104读630nm的荧光信号,通过如下公式计算抑制率:
%inhibition=100-(Signal cmpd-Signal Ave_PC)/(Signal Ave_VC-Signal Ave_PC)×100.
注:Signal Ave_PC:反应板中的阳性对照孔的荧光信号;Signal Ave_VC:反应板中的阴性对照孔的荧光信号。
利用以下非线性拟合公式来得到化合物的IC 50(半数抑制浓度):
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*HillSlope))
注:X:化合物浓度log值;Y:Inhibition%
按照上述实验方法测定对照化合物和本发明化合物对MAT2A酶活性抑制效果,结果如下表1所示:
表1测试化合物对MAT2A酶活性抑制结果
测试化合物 IC 50(nM)
对照化合物 89.04
I-1 14.9
I-2 33.49
I-3 32.93
I-4 57.18
I-5 52.13
I-6 38.33
I-7 137.3
I-8 41.34
I-11 20.50
I-12 74.61
I-17 21.1
I-20 23.23
I-21 25.26
I-22 32.22
I-23 22.53
I-24 23.78
I-25 128.4
I-26 39.98
实验结果表明,本发明化合物对MAT2A酶具有很好的抑制活性,并且优于对照化合物。
测试例2:化合物对HCT116 MTAP-/-细胞增殖的影响
按照下述实验方法测定对照化合物和本发明化合物对HCT116 MTAP-/-细胞增殖的影响。
HCT116 MTAP-/-细胞培养于MCCOYS 5A培养基中,加10%FBS和1%Penicillin-Streptomycin,置于37℃、5%CO 2条件下培养。细胞常规培养至细胞饱和度为80%-90%,收取细胞。用相应的培养基重悬,配制成合适密度的细胞悬液。将稀释好的化合物用Echo 550转移150nL至384细胞培养板;将细胞种到384细胞培养板中,400/孔,30uL。化合物的终浓度上限起始浓度为20μM,按照4倍的梯度稀释,共10个浓度,对照化合物AGI-24512起始浓度为30μM,按照3倍的梯度稀释,共10个浓度。将培养板放置细胞培养箱,37℃,5%CO 2环境中培养5天。将细胞待测板放置室温平衡30分钟,每孔加30μL CTG试剂(CelltiterGlo 试剂盒),室温避光放置30分钟后用Envision仪器读取化学发光信号值。通过检测发光值计算化合物对HCT116 MTAP-/-细胞增殖的抑制活性IC 50
表2测试化合物对HCT116 MTAP-/-细胞增殖的抑制结果
测试化合物 IC 50(nM)
对照化合物 266
I-1 27
I-11 87
I-20 115
I-22 100
实验结果显示,本发明化合物对HCT116 MTAP-/-细胞增殖有很好的抑制作用,其抑制活性优于对照化合物。
测试例3:化合物对HCT116 MTAP-/-细胞中SAM水平作用
按照下述实验方法测定对照化合物和本发明化合物对HCT116 MTAP-/-细胞中SAM水平作用。
HCT116 MTAP-/-细胞培养于MCCOYS 5A培养基中,加10%FBS和1%Penicillin-Streptomycin,置于37℃、5%CO 2条件下培养。细胞常规培养至细胞饱和度为80%-90%,收取细胞。用相应的培养基重悬,配制成合适密度的细胞悬液,接种在96孔培养皿中24小时后,用待测化合物与细胞在37℃、5%CO 2条件下共同孵育4小时。对化合物处理后细胞中SAM水平进行检测:将细胞在碳酸铵缓冲液(75mM,pH7.4)中轻轻洗涤一次,置于干冰上,并用代谢物提取缓冲液(含有50ng/ml氘化d3SAM的80%冷甲醇和20%乙酸)裂解。在4℃、3200rpm离心30分钟后,收集上清液并储存在-80℃直至通过LC/MS分析SAM的水平。
表3 80nM化合物对HCT116MTAP-/-细胞中SAM水平抑制
测试化合物 IC 50(nM)
I-11 31.35
I-26 32.96
实验结果显示,化合物对HCT116 MTAP-/-细胞中SAM水平有明显的抑制作用,并且优于对照化合物。
测试例4:人肝微粒体稳定性
按照下述实验方法测定对照化合物和本发明化合物的人肝微粒体稳定性。化合物的肝微粒体稳定性试验采用化合物与人肝微粒体体外共孵育进行检测。首先将待测化合物在DMSO溶 剂中配制成10mM的储备液,随后使用乙腈将化合物稀释至0.5mM。使用PBS稀释肝微粒体(Corning)成微粒体/缓冲液溶液,并使用该溶液稀释0.5mM的化合物成为工作溶液,工作溶液中化合物浓度为1.5μM,肝微粒体浓度为0.75mg/ml。取深孔板,每孔加入30μL工作溶液,然后加入15μL预热好的6mM NADPH溶液启动反应,37℃孵育。在孵育的0、5、15、30、45分钟时,加入135μL乙腈至相应的孔中终止反应。在最后45分钟时间点用乙腈终止反应后,深孔板涡旋振动10分钟(600rpm/min),然后离心15分钟。离心后取上清,1:1加入纯化水后进行LC-MS/MS检测,获得每个时间点化合物峰面积与内标峰面积比值,将5、15、30、45分钟时化合物的峰面积比值与0分钟时的峰面积比值进行比较,计算每个时间点化合物的剩余百分比,使用Graphpad 5软件计算T 1/2
表4化合物的肝微粒体稳定性结果
化合物 种属 T 1/2(minute) Cl int(mL/min/kg)
I-1 >1200 ≤0.01
实验结果显示,化合物I在人肝微粒体中表现出较好的稳定性,代谢慢,成药性好。
测试例5:化合物对人正常肝细胞毒性评价
按照下述实验方法测定对照化合物和本发明化合物对人正常肝细胞的毒性。
人正常肝细胞LO2在含10%FBS的DMEM培养基中培养,在细胞生长状态良好时,按照3000/孔,30μL/孔密度接种于384孔板。放入37℃,5%CO 2培养箱过夜。
每孔加入梯度稀释的化合物或DMSO,另设置不接种细胞加入培养基的孔作为空白对照。放入37℃,5%CO 2培养箱培养20小时后,根据CytoTox-Glo TM Cytotoxicity Assay试剂盒(Promega,Cat.No.G9290)说明书准备CytoTox-Glo TM Cytotoxicity检测试剂和裂解试剂。按10μL/孔加入CytoTox-Glo TM Cytotoxicity检测试剂,室温放置15分钟,在多功能酶标仪上读取死细胞luminescence信号。按10μL/孔加入裂解试剂,室温放置15分钟后在多功能酶标仪上读取总的luminescence信号。活细胞信号=总信号-死细胞信号。计算细胞活力抑制率:
细胞活力抑制率=(DMSO组-测试化合物)/(DMSO组-空白对照组)×100%
采用Graphpad 5软件计算IC 50值。
实验结果显示,本发明化合物对人正常肝细胞LO2未见抑制活性,表明对人正常肝细胞无毒性风险。
测试例6:小鼠药代动力学
按照下述实验方法测定对照化合物和本发明化合物的小鼠药代动力学性质。
采用雄性CD-1小鼠3只,剂量为10mg/kg,给药途径为灌胃,溶媒为5%DMSO+5%NMP+10%Solutol+80%(40%HB-CD),禁食过夜,采血时间点为给药前和在给药后15、30分钟以及1、2、4、6、8、24小时。血液样品6800g 2-8℃离心6分钟,收集血浆,于-80℃保存。取各时间点血浆30μL加入300μL含100ng/mL内标的甲醇,涡旋混匀后2-8℃18000g离心7分钟。取200μL转移至96孔进样板中,进行LC-MS/MS定量分析。主要药代动力学参数用WinNonlin 7.0软件非房室模型分析。
表5小鼠灌胃给药的主要药代动力学参数(均值)
化合物 T 1/2(h) T max(h) C max(ng/mL) AUC (0-t)(h·ng/mL)
I-1 2.37 0.5 11631 51974
实验结果表明,本发明化合物在小鼠体内表现出优良的药代动力学性质。
以上对本发明技术方案的实施方式进行了示例性的说明。应当理解,本发明的保护范围不拘囿于上述实施方式。凡在本发明的精神和原则之内,本领域技术人员所做的任何修改、等同替换、改进等,均应包含在本申请权利要求书的保护范围之内。

Claims (28)

  1. 一种式III所示的化合物,其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药:
    Figure PCTCN2022083568-appb-100001
    其中,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
    R 1选自C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基),所述C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基)任选被一个或多个R 1a取代,所述R 1a选自卤素;当R 1a为多个时,所述R 1a相同或不同;
    或当L 1为-N(R L1)-时,R L1和R 1可以与L组合在一起形成任选地被一个或多个R L1取代的3至6元杂环烷基;当R L1为多个时,所述R L1相同或不同;所述3至6元杂环烷基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    R 2选自无取代或被R 2a取代的C 6-C 10-芳基、无取代或被R 2a取代的5至10元杂芳基;当R 2a为多个时,所述R 2a相同或不同;所述5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    R 3选自无取代或被R 3a取代的C 6-C 10-芳基、无取代或被R 3a取代的5至10元杂芳基、无取代或被R 3a取代的环A;当R 3a为多个时,所述R 3a相同或不同;所述环A为
    Figure PCTCN2022083568-appb-100002
    其中环Cx和环Cy各自独立地选自C 5-C 6-芳基、5至6元杂芳基、5至6元环烷基和5至6元杂环烷基,D和E各自独立地选自C、CH、N,G和J各自独立地选自C、CH、CH 2、N、NH、O、S;
    所述R 2a选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A和-CN;
    所述R 3a选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A、-CN、-(C 1-C 6亚烷基)-5-6元杂环烷基、-(C 1-C 6亚烷基)-S(O) 2-(C 1-C 3 烷基)、-(C 1-C 6亚烷基)-C(O)-NR AR B
    R A和R B各自独立地选自H、-CN、-OH、氧代、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基;所述3至14元杂环烷基和5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    所述R A和R B中的每个C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基各自独立且任选地被一个或多个R Aa取代;当R Aa为多个时,所述R Aa相同或不同;
    所述R Aa各自独立地选自-OH、氘、卤素、C 1-C 6烷基、-N(R An) 2,其中每个R An各自独立地选自H、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 6-C 10芳基、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和5至10元杂芳基、-NHC(O)-(OC 1-C 6烷基)、-NO 2、-CN、氧代、-C(O)OH、-C(O)-O-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 1-C 6-烷氧基)、-C(O)NH 2、-C(O)-(C 1-C 6烷基)、-O-(C 1-C 6烷基)、-Si(C 1-C 6烷基) 3、-S(O) 0-2-(C 1-C 6烷基)、C 6-C 10芳基、-(C 1-C 6亚烷基)-(C 6-C 10芳基)、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和-O(C 6-C 14芳基);
    所述R Aa中的每个烷基、烯基、芳基和杂环烷基各自独立且任选地被一个或多个R Ab取代,所述R Ab各自独立地选自-OH、-O-(C 1-C 6烷基)、卤素、-NH 2、-(C 1-C 6亚烷基)-NH 2、-COOH、-CN和氧代;
    R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同。
  2. 如权利要求1所述的化合物,其特征在于,
    L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
    R 1选自C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基),所述C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基)任选被一个或多个R 1a取代,所述R 1a选自卤素;当R 1a为多个时,所述R 1a相同或不同;
    或当L 1为-N(R L1)-时,R L1和R 1可以与L组合在一起形成任选地被一个或多个R L1取代的3至6元杂环烷基;当R L1为多个时,所述R L1相同或不同;所述3至6元杂环烷基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    R 2选自无取代或被R 2a取代的C 6-C 10-芳基、无取代或被R 2a取代的5至10元杂芳基;当R 2a为多个时,所述R 2a相同或不同;所述5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    R 3选自无取代或被R 3a取代的C 6-C 10-芳基、无取代或被R 3a取代的5至10元杂芳基、无取代或被R 3a取代的环A;当R 3a为多个时,所述R 3a相同或不同;所述环A为
    Figure PCTCN2022083568-appb-100003
    其中环Cx和环Cy各自独立地选自C 5-C 6-芳基、5至6元杂芳基、5至6元环烷基和5至6元杂环烷基,D和E各自独立地选自C、CH、N,G和J各自独立地选自C、CH、CH 2、N、NH、O、S;
    所述R 2a和R 3a各自独立地选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A和-CN;
    R A和R B各自独立地选自H、-CN、-OH、氧代、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基;所述3至14元杂环烷基和5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    所述R A和R B中的每个C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基各自独立且任选地被一个或多个R Aa取代;当R Aa为多个时,所述R Aa相同或不同;
    所述R Aa各自独立地选自-OH、氘、卤素、-N(R An) 2,其中每个R An各自独立地选自H、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 6-C 10芳基、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和5至10元杂芳基、-NHC(O)-(OC 1-C 6烷基)、-NO 2、-CN、氧代、-C(O)OH、-C(O)-O-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 1-C 6-烷氧基)、-C(O)NH 2、-C(O)-(C 1-C 6烷基)、-O-(C 1-C 6烷基)、-Si(C 1-C 6烷基) 3、-S(O) 0-2-(C 1-C 6烷基)、C 6-C 10芳基、-(C 1-C 6亚烷基)-(C 6-C 10芳基)、 3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和-O(C 6-C 14芳基);
    所述R Aa中的每个烷基、烯基、芳基和杂环烷基各自独立且任选地被一个或多个R Ab取代,所述R Ab各自独立地选自-OH、-O-(C 1-C 6烷基)、卤素、-NH 2、-(C 1-C 6亚烷基)-NH 2、-COOH、-CN和氧代;
    R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同。
  3. 如权利要求1或2所述的化合物,其特征在于:
    L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;较佳地,R L1选自氢、甲基、乙基;
    和/或,R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基);所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代,所述R 1a选自卤素;
    和/或,R 2选自无取代或被R 2a取代的苯基和无取代或被R 2a取代的5至10元杂芳基,所述5至10元杂芳基其中一个环原子为N;较佳地,R 2选自无取代或被R 2a取代的苯基、无取代或被R 2a取代的吡啶基、无取代或被R 2a取代的嘧啶基;
    和/或,R 3选自无取代或被R 3a取代的苯基、无取代或被R 3a取代的吡啶基、无取代或被R 3a取代的吡啶酮基、无取代或被R 3a取代的哒嗪基、无取代或被R 3a取代的异噁唑基、无取代或被R 3a取代的吡唑基、和无取代或被R 3a取代的环A,所述环A选自
    Figure PCTCN2022083568-appb-100004
    苯并噻唑基、苯并异噻唑基、苯并噁唑基、苯并咪唑基、苯并三唑基、吲唑基、喹喔啉基、喹啉基、喹唑啉基、咪唑并吡啶基、吡唑并吡啶基、三唑并吡啶基、苯并哒嗪基、苯并呋喃基、二氢苯并呋喃基、二氢苯并二氧乙烯基和四氢苯并二氧乙烯基,其中n A选自整数1、2或3;较佳地,所述环A选自苯并噻唑基、苯并咪唑基、吲唑基、苯并哌啶基、苯并哌嗪基、苯并吗啉基、二氢茚、苯并二氢呋喃;
    和/或,R 2a和R 3a各自独立地选自-R A、-OR A、卤素和-CN;
    和/或,R A独立地选自H、-OH、C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)各自独立且任选地被一个或多个R Aa取代;
    和/或,R Aa各自独立地选自-OH、卤素、-N(R An) 2,其中每个R An各自独立地选自H、甲 基、乙基、异丙基、正丙基;
    和/或,R 4独立地选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自氟和氯;当R 4a为多个时,所述R 4a相同或不同。
  4. 如权利要求1-3任一项所述的化合物,其特征在于:
    L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、甲基、乙基;
    和/或,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
    Figure PCTCN2022083568-appb-100005
    较佳地,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100006
    和/或,R 2选自被R 2a取代的苯基和被R 2a取代的吡啶基,并且R 2a独立地选自氟、氯、溴、任选被一个或多个R Aa取代的-R A、任选被一个或多个R Aa取代的-OR A,所述R A独立地选自被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基,每个R Aa独立地选自-OH、氟、氯、溴;较佳地,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
    Figure PCTCN2022083568-appb-100007
    氟代甲氧基;更佳地,R 2选自
    Figure PCTCN2022083568-appb-100008
    Figure PCTCN2022083568-appb-100009
    和/或,R 3选自被R 3a取代的环A,所述环A选自
    Figure PCTCN2022083568-appb-100010
    Figure PCTCN2022083568-appb-100011
    Figure PCTCN2022083568-appb-100012
    并且R 3a独立地选自任选被一个或多个R Aa取代的-R A,所述R A独立地选自被一个或多个R Aa取代的C 1-C 6烷基,每个R Aa各自独立地选自-OH、氟、氯、溴;较佳地,R 3a独立地选自甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、叔丁基、-CH 2- OH、-(CH 2) 2-OH、-(CH 2) 3-OH、-(CH 2) 4-OH、
    Figure PCTCN2022083568-appb-100013
    和/或,R 4选自氢、卤素、-OH、-CN、甲基、乙基、正丙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、氟代环丙基、
    Figure PCTCN2022083568-appb-100014
    氯代甲基、氯代乙基、氯代丙基、氯代环丙基、
    Figure PCTCN2022083568-appb-100015
    较佳地,R 4选自氢、卤素、甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100016
  5. 如权利要求1-4任一项所述的化合物,其特征在于,R 4为氢;
    R 2
    Figure PCTCN2022083568-appb-100017
    R 2a选自:氟、氯、溴、-O-C 1-C 6烷基或C 3-C 6环烷基;所述C 1-C 6烷基任选地被一个或多个卤素取代;
    R 3为被R 3a取代的环A;
    所述R 3a选自-C 1-C 6烷基、-(C 1-C 6亚烷基)-5-6元杂环烷基、-(C 1-C 6亚烷基)-S(O) 2-CH 3、-(C 1-C 6亚烷基)-C(O)-N(CH 3) 2
    所述环A为
    Figure PCTCN2022083568-appb-100018
    其中,环Cx为5元杂芳环或5元杂环烷基;
    环Cy为苯环、5-6元杂芳环或5-6元杂环烷基;
    D和E各自独立地选自C、N;
    较佳地,所述环Cx为5元杂芳环,所述杂芳环含有2个N或所述杂芳环含有1个N和1个S;
    较佳地,所述环Cx为5元杂环烷基,所述杂环烷基含有1个O;
    较佳地,所述环Cy含有1或2个N;
    较佳地,所述卤素为F。
  6. 如权利要求1-5任一项所述的化合物,其特征在于,R 2
    Figure PCTCN2022083568-appb-100019
    Figure PCTCN2022083568-appb-100020
    R 2a选自:氟、氯、-O-CF 2H或环丙基;
    较佳地,R 2选自
    Figure PCTCN2022083568-appb-100021
    Figure PCTCN2022083568-appb-100022
  7. 如权利要求1-6任一项所述的化合物,其特征在于,R 3选自:
    Figure PCTCN2022083568-appb-100023
    Figure PCTCN2022083568-appb-100024
    R 3a选自:-C 1-C 6烷基、-C 1-C 6烷基-5-6元杂环烷基、-C 1-C 6烷基-S(O) 2-CH 3、-C 1-C 6烷基-C(O)-N(CH 3) 2
    较佳地,所述C 1-C 6烷基为直链或含支链的烷基;所述C 1-C 6烷基任选地被-OH取代。
  8. 如权利要求1-7任一项所述的化合物,其特征在于:R 3选自
    Figure PCTCN2022083568-appb-100025
    Figure PCTCN2022083568-appb-100026
    Figure PCTCN2022083568-appb-100027
  9. 如权利要求1-8任一项所述化合物,其特征在于,所述化合物为式II所示化合物或式II所示化合物的互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药:
    Figure PCTCN2022083568-appb-100028
    其中,R 1、R 2a、R 3a、R 4、L 1具有权利要求1-8任一项所述的定义;
    较佳地,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
    R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代;当R 1a为多个时,所述R 1a相同或不同;所述R 1a选自卤素;
    X 1选自C(R 2a)、N;
    Y 1、Y 2、Y 3各自独立地选自N、S、C;
    R 2a独立地选自-R A、-OR A和卤素;
    R 3a独立地选自-R A
    R A独立地选自H、-CN、-OH、C 1-C 6烷基、C 3-C 6环烷基、被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基;当R Aa为多个时,所述R Aa相同或不同;
    所述R Aa各自独立地选自-OH、氘、卤素;
    R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a 取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同。
  10. 根据权利要求1-9任一项所述的化合物,其特征在于,
    L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;较佳地,R L1选自氢、甲基、乙基;
    和/或,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
    Figure PCTCN2022083568-appb-100029
    较佳地,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100030
    和/或,X 1选自CH、N;
    和/或,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
    Figure PCTCN2022083568-appb-100031
    氟代甲氧基;
    和/或,R 3a独立地选自甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、叔丁基、-CH 2-OH、-(CH 2) 2-OH、-(CH 2) 3-OH、-(CH 2) 4-OH、
    Figure PCTCN2022083568-appb-100032
    和/或,R 4选自氢、卤素、-OH、-CN、甲基、乙基、正丙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、氟代环丙基、
    Figure PCTCN2022083568-appb-100033
    氯代甲基、氯代乙基、氯代丙基、氯代环丙基、
    Figure PCTCN2022083568-appb-100034
    较佳地,R 4选自氢、卤素、甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100035
  11. 如权利要求1-10任一项所述的化合物,其特征在于,所述化合物具有结构II-a或II-b所示的结构:
    Figure PCTCN2022083568-appb-100036
    其中,R 1、L 1、R 2a、R 3a具有权利要求1-10所述的定义。
  12. 如权利要求1-11任一项所述的化合物,其特征在于,L 1选自-O-、或-NH-;
    R 1选自C1-C3烷基、C3-C6环烷基、-(C1-C3亚烷基)-(C3-C6环烷基),所述C1-C3烷基、C3-C6环烷基、-(C1-C3亚烷基)-(C3-C6环烷基)任选地被一个或多个R 1a取代;当R 1a为多个时,所述R 1a相同或不同;所述R 1a选自卤素;
    较佳地,所述卤素为F;
    较佳地,所述R 1选自:甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
    Figure PCTCN2022083568-appb-100037
    更佳地,R 1选自:甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100038
    较佳地,-L 1-R 1选自:-NH-环丙基、-O-环丙基、-NH-CH 2CF 3、-O-CH 2CF 3、-NH-CH 2-环丙基、-O-CH 2-环丙基。
  13. 如权利要求1-12任一项所述的化合物,其特征在于,R 3a选自:-C 1-C 6烷基、-C 1-C 6烷基-5-6元杂环烷基、-C 1-C 6烷基-S(O) 2-CH 3、-C 1-C 6烷基-C(O)-N(CH 3) 2
    所述C 1-C 6烷基为直链或含支链的烷基;
    所述C 1-C 6烷基任选地被-OH取代;
    较佳地,所述5-6元杂环烷基为
    Figure PCTCN2022083568-appb-100039
    较佳地,R 3a选自:-CH 3
    Figure PCTCN2022083568-appb-100040
    Figure PCTCN2022083568-appb-100041
  14. 如权利要求1-13任一项所述的化合物,其特征在于,所述化合物为式I所示化合物或式I所示化合物的互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药:
    Figure PCTCN2022083568-appb-100042
    其中,R 1、L 1、R 2a、R 3a、R 4、n A具有权利要求1-13所述的定义;
    较佳地,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
    R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代;当R 1a为多个时,所述R 1a相同或不同;所述R 1a选自卤素;
    X 1选自C(R 2a)、N;
    R 2a独立地选自-R A、-OR A和卤素;
    R 3a独立地选自-R A
    R A独立地选自H、-CN、-OH、C 1-C 6烷基、C 3-C 6环烷基、被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基;当R Aa为多个时,所述R Aa相同或不同;
    所述R Aa各自独立地选自-OH、氘、卤素;
    R 4选自氢、卤素、-OH、-CN、C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 4a取代,所述R 4a选自卤素;当R 4a为多个时,所述R 4a相同或不同;
    n A选自整数1、2或3。
  15. 根据权利要求1-14任一项所述的化合物,其特征在于,
    L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;较佳地,R L1选自氢、甲基、乙基;
    和/或,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
    Figure PCTCN2022083568-appb-100043
    较佳地,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100044
    和/或,X 1选自CH、N;
    和/或,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、 乙氧基、正丙氧基、异丙氧基、
    Figure PCTCN2022083568-appb-100045
    氟代甲氧基;
    和/或,R 3a独立地选自甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、叔丁基、-CH 2-OH、-(CH 2) 2-OH、-(CH 2) 3-OH、-(CH 2) 4-OH、
    Figure PCTCN2022083568-appb-100046
    和/或,R 4选自氢、卤素、-OH、-CN、甲基、乙基、正丙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、氟代环丙基、
    Figure PCTCN2022083568-appb-100047
    氯代甲基、氯代乙基、氯代丙基、氯代环丙基、
    Figure PCTCN2022083568-appb-100048
    较佳地,R 4选自氢、卤素、甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100049
  16. 如权利要求1-15任一项所述的化合物,其特征在于,所述化合物选自为下列所示化合物或下列所示化合物的互变异构体、立体异构体、水合物、溶剂化物、药学可接受的盐或前药:
    Figure PCTCN2022083568-appb-100050
    Figure PCTCN2022083568-appb-100051
    Figure PCTCN2022083568-appb-100052
  17. 如下所示的中间体M-1和M-2:
    Figure PCTCN2022083568-appb-100053
    其中,
    R M选自卤素;L 1、R 1、R 2、R 4具有权利要求1-16任一项所述的定义;
    较佳地,L 1、R 1、R 2的定义如权利要求1中所述;
    较佳地,L 1选自-O-、-S-、-N(R L1)-和单键;R L1选自氢和C 1-C 6烷基;
    较佳地,R 1选自C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基);
    所述C 1-C 6烷基、C 2-C 6烯基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基)和-(C 1-C 6亚烷基)-(C 3-C 6环烯基)任选被一个或多个R 1a取代,所述R 1a选自卤素;当R 1a为多个时,所述R 1a相同或不同;
    或当L 1为-N(R L1)-时,R L1和R 1可以与L组合在一起形成任选地被一个或多个R L1取代的3至6元杂环烷基;当R L1为多个时,所述R L1相同或不同;所述3至6元杂环烷基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    较佳地,R 2选自无取代或被R 2a取代的C 6-C 10芳基、无取代或被R 2a取代的5至10元杂芳基;当R 2a为多个时,所述R 2a相同或不同;所述5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    所述R 2a独立地选自-R A、-OR A、卤素、-N=N-R A、NR AR B、-(C 1-C 6亚烷基)-NR AR B、-C(O)OR A、-C(O)NR AR B、-OC(O)R A和-CN;
    R A和R B各自独立地选自H、-CN、-OH、氧代、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、 3至14元杂环烷基、5至10元杂芳基;所述3至14元杂环烷基和5至10元杂芳基,其中1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子;
    所述R A和R B中的每个C 1-C 6烷基、C 3-C 6环烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、-NH 2、-S(O) 0-2-(C 1-C 6烷基)、-S(O) 0-2-(C 6-C 10芳基)、-CO-(C 1-C 6烷基)、-CO-(C 3-C 14环烷基)、-C 3-C 14环烷基、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)、C 6-C 10芳基、3至14元杂环烷基、5至10元杂芳基各自独立且任选地被一个或多个R Aa取代;当R Aa为多个时,所述R Aa相同或不同;
    所述R Aa各自独立地选自-OH、氘、卤素、-N(R An) 2,其中每个R An各自独立地选自H、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 6-C 10芳基、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和5至10元杂芳基、-NHC(O)-(OC 1-C 6烷基)、-NO 2、-CN、氧代、-C(O)OH、-C(O)-O-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 1-C 6-烷氧基)、-C(O)NH 2、-C(O)-(C 1-C 6烷基)、-O-(C 1-C 6烷基)、-Si(C 1-C 6烷基) 3、-S(O) 0-2-(C 1-C 6烷基)、C 6-C 10芳基、-(C 1-C 6亚烷基)-(C 6-C 10芳基)、3至14元杂环烷基、-(C 1-C 6亚烷基)-(3至14元杂环烷基)、和-O(C 6-C 14芳基);
    所述R Aa中的每个烷基、烯基、芳基和杂环烷基各自独立且任选地被一个或多个R Ab取代,所述R Ab各自独立地选自-OH、-O-(C 1-C 6烷基)、卤素、-NH 2、-(C 1-C 6亚烷基)-NH 2、-COOH、-CN和氧代;
    且所述中间体M-1不包括下列化合物:
    Figure PCTCN2022083568-appb-100054
  18. 如权利要求17所述的中间体M-1和M-2,其特征在于:
    R M选自氟、氯、溴、碘;较佳地,R M选自氟、氯、溴;
    L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、C 1-C 3烷基;较佳地,R L1选自氢、甲基、乙基;
    和/或,R 1选自C 1-C 6烷基、C 3-C 6环烷基、-(C 1-C 6亚烷基)-(C 3-C 6环烷基);所述C 1-C 6烷基、C 3-C 6环烷基和-(C 1-C 6亚烷基)-(C 3-C 6环烷基)任选被一个或多个R 1a取代,所述R 1a选自卤素;
    和/或,R 2选自无取代或被R 2a取代的苯基和无取代或被R 2a取代的5至10元杂芳基,所述5至10元杂芳基其中一个环原子为N;较佳地,R 2选自无取代或被R 2a取代的苯基、无取代或被R 2a取代的吡啶基、无取代或被R 2a取代的嘧啶基;
    和/或,R 2a独立地选自-R A、-OR A、卤素和-CN;
    和/或,R A独立地选自H、-OH、C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基),所述C 1-C 6烷基、C 3-C 6环烷基、-CO-(C 1-C 6烷基)、-(C 1-C 6亚烷基)-(C 3-C 14环烷基)各自独立且任选地被一个或多个R Aa取代;
    和/或,R Aa各自独立地选自-OH、卤素、-N(R An) 2,其中每个R An各自独立地选自H、甲基、乙基、异丙基、正丙基。
  19. 如权利要求17或18所述的中间体M-1和M-2,其特征在于:
    L 1选自-O-、-S-、-N(R L1)-或单键;R L1选自氢、甲基、乙基;
    和/或,R 1选自甲基、乙基、丙基、氟代甲基、氟代乙基、氟代丙基、环丙基、
    Figure PCTCN2022083568-appb-100055
    较佳地,R 1选自甲基、乙基、-CH 2-CF 3、环丙基、
    Figure PCTCN2022083568-appb-100056
    和/或,R 2选自被R 2a取代的苯基和被R 2a取代的吡啶基,并且R 2a独立地选自氟、氯、溴、任选被一个或多个R Aa取代的-R A、任选被一个或多个R Aa取代的-OR A,所述R A独立地选自被一个或多个R Aa取代的C 1-C 6烷基、被一个或多个R Aa取代的C 3-C 6环烷基,每个R Aa独立地选自-OH、氟、氯、溴;较佳地,R 2a独立地选自-氟、氯、溴、甲基、乙基、正丙基、异丙基、环丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、
    Figure PCTCN2022083568-appb-100057
    氟代甲氧基;更佳地,R 2选自
    Figure PCTCN2022083568-appb-100058
    Figure PCTCN2022083568-appb-100059
  20. 如权利要求17-19任一项所述的中间体M-1,其特征在于,其选自下列任一化合物:
    Figure PCTCN2022083568-appb-100060
  21. 如权利要求17-19任一项所述的中间体M-2,其特征在于,其选自下列任一化合物:
    Figure PCTCN2022083568-appb-100061
    Figure PCTCN2022083568-appb-100062
  22. 权利要求1-16任一项所述化合物的制备方法,其包括:
    (1)使中间体M-1与卤化试剂接触,得到所述中间体M-2;和/或
    Figure PCTCN2022083568-appb-100063
    (2)使所述中间体M-2与
    Figure PCTCN2022083568-appb-100064
    接触,得到式III所示化合物,
    Figure PCTCN2022083568-appb-100065
    其中,R 1、R 2、R 3、R 4、L 1具有权利要求1-16任一项所述的定义,R M选自氟、氯、溴、碘;较佳地,R M选自氟、氯、溴。
  23. 如权利要求22所述的方法,其特征在于:
    步骤(1)中,所述卤化试剂选自N-氯代丁二酰亚胺、N-溴代琥珀酰亚胺;和/或
    步骤(2)中,在惰性气体保护下将中间体M-2与
    Figure PCTCN2022083568-appb-100066
    钯催化剂和碱接触,得到式III所示化合物。
  24. 一种药物组合物,其包含治疗有效量的权利要求1-16任一项所示的化合物、其互变异构体、立体异构体、水合物、溶剂化物、药学上可接受的盐或前药中的至少一种。
  25. 权利要求1-16任一项所述的化合物或者权利要求24的药物组合物在制备药物中的用途,所述药物用于治疗或者预防MAT2A相关疾病。
  26. 权利要求1-16任一项所述的化合物或者权利要求24的药物组合物在制备药物中的 用途,所述药物用于治疗或者预防癌症。
  27. 权利要求26所述的用途,其中所述癌症是MTAP缺失的癌症。
  28. 权利要求26所述的用途,其中所述癌症选自间皮瘤、神经母细胞瘤、直肠癌、结肠癌、熟悉的腺瘤性息肉病和遗传性非息肉性结直肠癌、食道癌、唇癌、喉癌、下咽癌、舌癌、唾液腺癌、胃癌、腺癌、甲状腺髓样癌、甲状腺乳头状癌、肾癌、肾实质癌、卵巢癌、子宫颈癌、子宫体癌、子宫内膜癌、绒毛膜癌、胰腺癌、前列腺癌、膀胱癌、睾丸癌、乳腺癌、泌尿癌、黑素瘤、脑瘤、淋巴瘤、头颈癌、急性淋巴白血病(ALL)、慢性淋巴白血病(CLL)、急性髓样白血病(AML)、慢性粒细胞白血病(CML)、肝细胞癌、胆囊癌、支气管癌、小细胞肺癌、非小细胞肺癌、多发性骨髓瘤、基底肉瘤、畸胎瘤、视网膜母细胞瘤、脉络膜黑色素瘤、精原细胞瘤、横纹肌肉瘤、骨肉瘤、软骨肉瘤、肌瘤、脂肪肉瘤、纤维肉瘤、尤因肉瘤和浆细胞瘤。
PCT/CN2022/083568 2021-03-29 2022-03-29 嘧啶并吡嗪酮化合物及其用途 WO2022206730A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110335375.3 2021-03-29
CN202110335375 2021-03-29

Publications (2)

Publication Number Publication Date
WO2022206730A1 true WO2022206730A1 (zh) 2022-10-06
WO2022206730A8 WO2022206730A8 (zh) 2024-02-22

Family

ID=83406799

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/083568 WO2022206730A1 (zh) 2021-03-29 2022-03-29 嘧啶并吡嗪酮化合物及其用途

Country Status (2)

Country Link
CN (1) CN115141202A (zh)
WO (1) WO2022206730A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11999713B2 (en) 2021-10-20 2024-06-04 Insilico Medicine Ip Limited Methionine adenosyltransferase 2a (MAT2A) inhibitors and uses thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020123395A1 (en) * 2018-12-10 2020-06-18 Ideaya Biosciences, Inc. 2-oxoquinazoline derivatives as methionine adenosyltransferase 2a inhibitors
WO2020139991A1 (en) * 2018-12-27 2020-07-02 Agios Pharmaceuticals, Inc. Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
WO2020139992A1 (en) * 2018-12-27 2020-07-02 Agios Pharmaceuticals, Inc. Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
CN111936499A (zh) * 2018-03-30 2020-11-13 安吉奥斯医药品有限公司 Mat2a的杂二环抑制剂和用于治疗癌症的方法
WO2020243376A1 (en) * 2019-05-31 2020-12-03 Agios Pharmaceuticals, Inc. Heterobicyclic inhibitors of mat2a and methods of use for treating cancer
WO2021139775A1 (zh) * 2020-01-10 2021-07-15 江苏先声药业有限公司 吡啶酮化合物及应用

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111936499A (zh) * 2018-03-30 2020-11-13 安吉奥斯医药品有限公司 Mat2a的杂二环抑制剂和用于治疗癌症的方法
WO2020123395A1 (en) * 2018-12-10 2020-06-18 Ideaya Biosciences, Inc. 2-oxoquinazoline derivatives as methionine adenosyltransferase 2a inhibitors
WO2020139991A1 (en) * 2018-12-27 2020-07-02 Agios Pharmaceuticals, Inc. Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
WO2020139992A1 (en) * 2018-12-27 2020-07-02 Agios Pharmaceuticals, Inc. Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
WO2020243376A1 (en) * 2019-05-31 2020-12-03 Agios Pharmaceuticals, Inc. Heterobicyclic inhibitors of mat2a and methods of use for treating cancer
WO2021139775A1 (zh) * 2020-01-10 2021-07-15 江苏先声药业有限公司 吡啶酮化合物及应用

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11999713B2 (en) 2021-10-20 2024-06-04 Insilico Medicine Ip Limited Methionine adenosyltransferase 2a (MAT2A) inhibitors and uses thereof

Also Published As

Publication number Publication date
WO2022206730A8 (zh) 2024-02-22
CN115141202A (zh) 2022-10-04

Similar Documents

Publication Publication Date Title
WO2021073439A1 (zh) 用于抑制shp2活性的吡嗪衍生物
CN113454085B (zh) Mat2a的aza杂双环抑制剂和用于治疗癌症的方法
TW202110841A (zh) Mat2a之雜雙環抑制劑及用於治療癌症之使用方法
WO2018113584A1 (zh) Fgfr4抑制剂、其制备方法与药学上的应用
TW202033526A (zh) 用作酪胺酸激酶抑制劑的化合物、包含其的藥物組合物及其用途
WO2021244609A1 (zh) 具有大环结构的化合物及其用途
CN115260187A (zh) 吡啶酮化合物及其用途
WO2022095904A1 (zh) 吡唑并哒嗪酮化合物、其药物组合物及其用途
WO2022206730A1 (zh) 嘧啶并吡嗪酮化合物及其用途
WO2019233457A1 (zh) Erk抑制剂及其应用
TW202017928A (zh) 新穎三環化合物
TWI691500B (zh) 作為tyro3、axl和mertk(tam)家族受體酪胺酸激酶抑制劑之雜環化合物
WO2020156319A1 (zh) N-甲酰胺衍生物、其制备方法及其在医药上的用途
WO2023036252A1 (zh) 吡咯并嘧啶类或吡咯并吡啶类衍生物及其医药用途
WO2023280254A1 (zh) 一种tead抑制剂
TW202028195A (zh) 作為TGF-βR1抑制劑的化合物及其應用
WO2023165581A1 (zh) 一种吡啶类衍生物及其用途
WO2022206939A1 (zh) 作为fgfr抑制剂的杂环化合物及其应用
WO2022222911A1 (zh) 嘧啶酮化合物及其用途
WO2022148196A1 (zh) 多激酶抑制剂及其用途
WO2022089219A1 (zh) 芳基酰胺化合物、包含其的药物组合物及其制备方法和用途
TW202112783A (zh) 三環類化合物及其用途
WO2020233645A1 (zh) 大环类衍生物、及其制备方法和用途
WO2020156162A1 (zh) Mek抑制剂及其在医药上的应用
WO2023078267A1 (zh) 作为蛋白激酶调节剂的含氨基大环化合物

Legal Events

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

Ref document number: 22778912

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22778912

Country of ref document: EP

Kind code of ref document: A1