WO2022194096A1 - 雌激素受体调节剂 - Google Patents

雌激素受体调节剂 Download PDF

Info

Publication number
WO2022194096A1
WO2022194096A1 PCT/CN2022/080680 CN2022080680W WO2022194096A1 WO 2022194096 A1 WO2022194096 A1 WO 2022194096A1 CN 2022080680 W CN2022080680 W CN 2022080680W WO 2022194096 A1 WO2022194096 A1 WO 2022194096A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
alkyl
methyl
group
alkylamino
Prior art date
Application number
PCT/CN2022/080680
Other languages
English (en)
French (fr)
Inventor
杨铉
朱程刚
张朝春
泰利约翰•杰弗瑞
陈超乐
包丽茗
闵祥燕
徐良亮
Original Assignee
深圳福沃药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳福沃药业有限公司 filed Critical 深圳福沃药业有限公司
Priority to JP2023557230A priority Critical patent/JP2024511044A/ja
Priority to EP22770451.7A priority patent/EP4310079A1/en
Priority to KR1020237035079A priority patent/KR20230157439A/ko
Priority to AU2022240650A priority patent/AU2022240650A1/en
Publication of WO2022194096A1 publication Critical patent/WO2022194096A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the present invention relates to certain novel compounds, or pharmaceutically acceptable salts thereof, which possess anticancer activity and are therefore potentially useful in methods of treating the human or animal body.
  • the present invention also relates to methods for the manufacture of the compounds, pharmaceutical compositions containing them and their use in methods of treatment, such as for the manufacture of medicaments for the prevention or treatment of cancer in warm-blooded animals such as humans, including For the prevention or treatment of estrogen receptor-dependent or estrogen receptor-mediated diseases.
  • the present invention also relates to compounds that are selective down-modulators of the estrogen receptor.
  • Estrogen receptor belongs to the steroid hormone receptor, including two subtypes, ER ⁇ and ER ⁇ . ER is involved in the regulation and development of the female reproductive tract; in cancers such as breast cancer, tumor growth is related to the action of estrogen and ER receptors, for example, the expression of ER is elevated in most breast cancer patients.
  • SERMs selective estrogen receptor modulators
  • WO2017/182493A1 also discloses a SERM compound.
  • the inventors of the present invention found that the compound represented by the formula (I) has the basic structure of tetrahydrooxazoloisoquinolin-one, which has significant ER degradation activity, and therefore unexpectedly has significantly better properties than the prior art Compounds are known for their ER inhibitory activity; such compounds also have excellent metabolic stability. Therefore, the compounds of the present invention can be used for the treatment of estrogen receptor-dependent or estrogen receptor-mediated diseases, thereby completing the present invention.
  • the present invention relates to the following.
  • One aspect of the present invention provides a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof:
  • Z 1 is selected from CR a R b , C(O) and a bond
  • Z 2 is selected from O, S, NR c , C(O) and bonds, or C 1 -C 6 alkylene, O-(C 1 -C optionally substituted with one or more of the same or different R d 6 alkylene) or NH-(C 1 -C 6 alkylene);
  • Cy 1 is selected from C 6-14 arylene, C 3-8 cycloalkylene, C 5-14 heteroarylene, C 3-14 heterocycloalkylene, C 3-14 heterocycloalkenylene, each of which is optionally substituted with a group selected from the group consisting of halogen atoms, hydroxy, amino, cyano and C 1-6 alkyl or C 1-6 alkoxy optionally substituted with halogen atoms;
  • Cy 2 is selected from bond, C 3-10 cycloalkylene, C 3-14 heterocycloalkylene, each of which is optionally substituted with a group selected from the group consisting of halogen atoms (including F, Cl, Br or I atoms) ), hydroxyl, amino, cyano, and C 1-6 alkyl or C 1-6 alkoxy optionally substituted by halogen atoms;
  • halogen atoms including F, Cl, Br or I atoms
  • R 1 and R 2 are each independently H, halogen atom, hydroxyl, amino, cyano, C 1-6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino , C 3 -C 8 cycloalkyl, said C 1-6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino or C 3 -C 8 cycloalkyl optionally substituted by one or more groups selected from halogen atoms, hydroxy, amino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, cyano or oxo;
  • R 4 is H, halogen atom, hydroxyl, amino, cyano, C 1-6 alkyl, C 1-6 alkylamino, amino C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkene amino, C 3 -C 8 cycloalkyl, the C 1-6 alkyl, C 1-6 alkylamino, amino C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkenyl Amino or C 3 -C 8 cycloalkyl is optionally replaced by one or more selected from halogen atoms, hydroxyl, amino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, cyano, oxo group, C 6-14 aryl, C 5-14 heteroaryl, C 3-14 heterocycloalkylene or
  • R 4 is H, halogen atom, hydroxyl, amino, cyano, C 1-6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino, C 3 -C 8 cycloalkyl, said C 1-6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino or C 3 -C 8 cycloalkyl are optionally Substituted with one or more groups selected from halogen atom, hydroxyl, amino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, cyano or oxo;
  • R 3 is -(CR e R f ) m -CR 31 R 32 R 33 , wherein m is 1, 2 or 3;
  • R 31 , R 32 and R 33 are independently selected from: H, a C 1-6 alkyl group, a halogen atom, a cyano group, and R 31 and R 32 may together form a C 3-8 cycloalkylene group, the C 1-8 6 alkyl and C 3-8 cycloalkylene are optionally substituted by hydroxyl, cyano, amino or halogen atoms;
  • R a , R b , R c , R d , Re and R f are each independently H, a halogen atom, a hydroxyl group, an amino group, a cyano group, a C 1-6 alkyl group optionally substituted with a halogen atom.
  • heteroaryl/heteroarylene, heterocycloalkyl/heterocycloalkylene, heterocycloalkenyl/heterocycloalkenyl contain 1-4 selected from N, O, S
  • the heteroatoms are used as ring constituent atoms.
  • the cycloalkyl/cycloalkylene, heterocycloalkyl/heterocycloalkylene, heterocycloalkenyl/heterocycloalkenylene may be monocyclic, bicyclic or tricyclic (preferably monocyclic or bicyclic) ring systems, wherein bicyclic or tricyclic ring systems include spiro, bridged or fused rings.
  • the aryl/arylene and heteroaryl/heteroarylene may be monocyclic, fused bicyclic or fused tricyclic (preferably monocyclic or fused bicyclic) ring systems, and wherein the fused bicyclic or The fused tricyclic ring system may contain non-aromatic ring structures.
  • the halogen atoms are selected from F, Cl, Br or I atoms.
  • Z 1 is selected from CR a R b , C(O) and a bond
  • Z 2 is selected from O, S, NR c , C(O) and bonds, or C 1 -C 6 alkylene, O-(C 1 -C optionally substituted with one or more of the same or different R d 6 alkylene) or NH-(C 1 -C 6 alkylene);
  • Cy 1 is selected from C 6-14 arylene (preferably C 6-10 arylene) and C 5-14 heteroarylene (preferably C 5-10 heteroarylene), each of which is optionally selected from the following The group substitution of: halogen atom, and C 1-6 alkyl or C 1-6 alkoxy optionally substituted by halogen atom;
  • Cy 2 is selected from bonds, C 3-10 cycloalkylene (preferably C 3-8 cycloalkylene), C 3-14 heterocycloalkylene (preferably C 3-10 heterocycloalkylene);
  • R 1 and R 2 are each independently H or a C 1-6 alkyl group, and the C 1-6 alkyl group is optionally substituted by one or more groups selected from halogen atoms, hydroxyl groups or cyano groups;
  • R 3 is -(CR e R f ) m -CR 31 R 32 R 33 , wherein m is 1, 2 or 3;
  • R 31 , R 32 and R 33 are independently selected from: H, a C 1-6 alkyl group, a halogen atom, a cyano group, and R 31 and R 32 may together form a C 3-8 cycloalkylene group, the C 1-8 6 alkyl and C 3-8 cycloalkylene are optionally substituted by hydroxyl, cyano, amino or halogen atoms;
  • R a , R b , R c , R d , Re , R f are each independently H or a halogen atom
  • R 4 is C 1-6 alkyl, C 1-6 alkylamino, amino C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkenylamino, C 3 -C 8 cycloalkyl,
  • the C 1-6 alkyl, C 1-6 alkylamino, amino C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkenylamino or C 3 -C 8 cycloalkyl are optional by one or more selected from halogen atom, hydroxyl, amino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, cyano, oxo and
  • R 4 is C 1-6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino, C 3 -C 8 cycloalkyl, the C 1- 6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino, or C 3 -C 8 cycloalkyl optionally by one or more selected from halogen atoms, hydroxyl, amino , C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, cyano or oxo group substitution.
  • Z 1 is the key
  • Z 2 is selected from -O-CH 2 -CH 2 -, -O-CH 2 -, -NH-CH 2 -CH 2 -, -NH-CH 2 -, -NH- or -O-;
  • Cy 1 is a C 6-10 arylene group (preferably phenyl) and a C 5-10 heteroarylene group (preferably pyridyl), which are optionally replaced by a halogen atom (preferably F) or a C 1-6 alkoxy group (preferably methoxy) substituted;
  • Cy 2 is selected from bond, azetidine, pyrrolidylene, piperazinylene,
  • R 1 is C 1-6 alkyl (preferably methyl);
  • R 2 is H
  • R 3 is -CH 2 -CR 31 R 32 R 33 ;
  • R 31 , R 32 and R 33 are independently selected from: H, C 1-6 alkyl (preferably methyl) optionally substituted with hydroxy or halogen atom (preferably F), halogen atom (preferably F), cyano, and R 31 and R 32 may together form a C 3-8 cycloalkylene group (preferably cyclopropylidene) optionally substituted by a hydroxyl group or a halogen atom (preferably F);
  • R 4 is optionally selected from halogen atom (preferably F), oxo group, C 1-6 alkylamino (preferably methylamino), (C 1-6 alkyl) 2 amino (preferably dimethylamino) ,and
  • Cy 2 is selected from bond, azetidine, pyrrolidylene, and R 4 is optionally selected from halogen atom (preferably F), oxo, C 1-6 alkylamino (preferably methylamino) and (C 1-6 alkyl) 2 amino (preferably dimethylamino) groups substituted C 1-6 alkyl, C 1-6 alkylamino, C 2-6 alkenyl, C 2-6 alkenylamino.
  • halogen atom preferably F
  • oxo oxo
  • C 1-6 alkylamino preferably methylamino
  • C 1-6 alkyl 2 amino preferably dimethylamino
  • Z 1 is the key
  • Z 2 is selected from -NH- or -O-;
  • Cy 1 is phenyl or pyridyl (preferably phenyl) optionally substituted with F or methoxy;
  • Cy 2 is selected from azetidine, pyrrolidylene, piperazinylene;
  • R 1 is methyl
  • R 2 is H
  • R 3 is -CH 2 -CR 31 R 32 R 33 ;
  • R 31 and R 32 are each F, or R 31 and R 32 together form a cyclopropylene
  • R 33 is selected from: H, hydroxymethyl, fluoromethyl, preferably selected from F, hydroxymethyl, fluoromethyl;
  • R 4 is F-(CH 2 ) 3 -or
  • Cy 2 is selected from azetidine and pyrrolidylene, and R 4 is F-(CH 2 ) 3 -.
  • the compounds of formula (I) of the present invention are in the form of stereoisomers, eg, the carbon at position 1 of the tetrahydroisoquinoline ring (ie, the carbon to which Z is attached) may be in the S configuration , and/or the carbon at position 3 (ie, the carbon to which R 1 is attached) may be in the R configuration.
  • the present invention relates to the following compounds or stereoisomers or pharmaceutically acceptable salts thereof:
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising any one or more of the foregoing compounds of the present invention, or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention relates to the use of a compound of the present invention in the manufacture of a medicament for the treatment of an estrogen receptor-dependent or estrogen receptor-mediated disease in a mammal.
  • Step 3 Synthesis of (R)-7-2-aminopropyl)(2,2,2-trifluoroethyl-3-tritylbenzo[d]oxazol-2(3H)-one
  • Step 6 3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-3-trityl-2, 3,6,7,8,9-Six Synthesis of Hydroxazolo[5,4-f]isoquinolin-6yl)pyridin-3-yl)amino)azetidine-1-carboxylic acid tert-butyl ester
  • Step 7 (6S,8R)-6-(5-(azetidine-3-amino)pyridin-2-yl)-8-methyl-7-(2,2,2-trifluoroethyl yl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Trifluoroacetic acid (9.00 mL) and water (1.00 mL) were added to 3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2) at 0°C -Trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6yl)pyridin-3-yl ) amino) azetidine-1-carboxylic acid tert-butyl ester (120 mg, 154 ⁇ mol, 1 eq), the mixture was reacted at 20° C. for 2 hours, and the completion of the reaction was monitored by TLC.
  • Step 8 (6S,8R)-6-(5-((1-(3-fluoropropane)azetidin-3-yl)amino)pyridin-2-yl)-8-methyl -Synthesis of 7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one (compound 5)
  • Step 1 (S)-3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-3-trityl Base-2,3,6,7,8,9- Synthesis of tert-butyl hexahydrooxazolo[5,4-f]isoquinolin-6-yl)pyridin-3-yl)oxy)pyrrolidine-1-carboxylate
  • Trifluoroacetic acid (1.54 g, 13.4 mmol, 997 uL, 87.3 eq) was added to (S)-3-((6-((6S,8R)-8-methane dissolved in dichloromethane (2 mL) at 0 °C yl-2-oxo-7-(2,2,2-trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4 -f] isoquinolin-6-yl)pyridin-3-yl)oxy)pyrrolidine-1-carboxylic acid tert-butyl ester (120 mg, 154 ⁇ mol, 1 eq) at 20°C for 2 hours, TLC and LCMS monitoring the reaction Finish.
  • Step 1 (6S,8R)-6-(3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl) )-3-trityl -2,3,6,7,8,9-Hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenyl)-2,6-diazaspiro[3.3]heptane- Synthesis of tert-butyl 2-carboxylate to make
  • Step 3 (6S,8R)-6-(2,6-difluoro-4-(6-(2-fluoroethyl)-2,6-diazaspiro[3.3]heptane-2-yl)benzene base)-8-methyl Synthesis of -7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 4 (6S,8R)-6-(2,6-difluoro-4-(6-(2-fluoroethyl)-2,6-diazaspiro[3.3]heptane-2-yl)benzene base)-8-methyl Resolution of -7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one ( Compound 8)
  • Example 13-15 were synthesized; the compound in Example 5, step 3, namely “(R) -7-2-Aminopropyl)(2,2,2-trifluoroethyl-3-tritylbenzo[d]oxazol-2(3H)-one ⁇ as starting material, the following Example 10 was synthesized -12, 16, 18, 25; the compound of step 4 in Example 1 is -(R)-7-(2-aminopropyl)-3-tritylbenzo[d]o
  • Step 3 (6S,8R)-6-(4-((1-(3-Fluoropropyl)azetidin-3-yl)amino)-2-methoxyphenyl)-8-methyl yl-7-(2,2,2-trifluoroethyl)-3-trityl-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinoline-2( Synthesis of 3H)-ketone
  • Trifluoroacetic acid (15.4 g, 135 mmol) and (6S,8R)-6-(4-((1-(3-fluoropropyl)azetidin-3-yl)amino)-2-methoxy phenyl)-8-methyl-7-(2,2,2-trifluoroethyl)-3-trityl-6,7,8,9-tetrahydrooxazolo[5,4- f]
  • Isoquinolin-2(3H)-one (1 g, 1.31 mmol) was dissolved in water (1 mL), and the reaction solution was reacted at 20° C. under nitrogen protection for 1 hour. The end of the reaction was detected by LCMS.
  • Step 6 3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-3-trityl-2, 3,6,7,8,9-Hexahydrooxazolo[5,4-f]isoquinolin-6yl)pyridin-3-yl)amino)azetidine-1-carboxylate tert-butyl ester synthesis
  • Step 7 (6S,8R)-6-(5-(azetidin-3-ylamino)pyridin-2-yl)-8-methyl-7-(2,2,2-trifluoroethyl yl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Trifluoroacetic acid (9.00 mL) and water (1.00 mL) were added to 3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2) at 0°C -Trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6yl)pyridin-3-yl ) amino) azetidine-1-carboxylic acid tert-butyl ester (120 mg, 154 umol, 1 eq), the mixture was reacted at 20° C. for 2 hours, and the completion of the reaction was monitored by TLC.
  • Step 8 (6S,8R)-6-(5-((1-(3-fluoropropane)azetidin-3-yl)amino)pyridin-2-yl)-8-methyl-7- Synthesis of (2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 4 (6S,8R)-6-(3-Fluoro-5-((1-(3-Fluoropropyl)azetidin-3-yl)amino)pyridin-2-yl)-8- Synthesis of methyl-7-(2,2,2-trifluoromethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Trifluoroacetic acid (2.02 g, 17.6 mmol) was dissolved in water (0.05 mL) and (6S,8R)-6-(3-fluoro-5-((1-(3-fluoropropyl)azetidine) was added Alkyl-3-yl)amino)pyridin-2-yl)-8-methyl-7-(2,2,2-trifluoroethyl)-3-trityl-6,7,8,9- In tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one (120 mg, 138 umol), the reaction was carried out at 20°C for 5 hours under nitrogen protection. LCMS detected the end of the reaction.
  • Step 5 (6S,8R)-6-(3-Fluoro-5-((1-(3-Fluoropropyl)azetidin-3-yl)amino)pyridin-2-yl)-8- Synthesis of methyl-7-(2,2,2-trifluoromethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 1 (6S,8R)-6-(5-bromo-3-methoxypyridin-2-yl)-8-methyl-7-(2,2,2-trifluoroethyl)-6, Synthesis of 7,8,9-Tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 3 (6S,8R)-6-(5-((1-(3-Fluoropropyl)azetidin-3-yl)amino)-3-methoxypyridin-2-yl) -8-Methyl-7-(2,2,2-trifluoroethyl)-3-trityl-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinoline Synthesis of Lin-2(3H)-ones
  • Step 1 (S)-3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-3-trityl tert-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)pyridin-3-yl)oxy)pyrrolidine-1-carboxylic acid Synthesis of Butyl Ester
  • Trifluoroacetic acid (1.54 g, 13.4 mmol, 997 uL, 87.3 eq) was added to (S)-3-((6-((6S,8R)-8-methane dissolved in dichloromethane (2 mL) at 0 °C yl-2-oxo-7-(2,2,2-trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4 -f] isoquinolin-6-yl)pyridin-3-yl)oxy)pyrrolidine-1-carboxylate tert-butyl ester (120mg, 154umol, 1eq) at 20°C for 2 hours, monitored by TLC and LCMS The reaction is complete.
  • Step 1 (S)-3-(3-methoxy-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)- 3-Trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenoxy)pyrrolidine-1-carboxylic acid tert.
  • Step 3 (6S,8R)-6-(4-(((S)-1-(3-fluoropropyl)pyrrolidin-3-yl)oxy)-2-methoxyphenyl)-8 -Methyl-7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one synthesis
  • Step 2 (6S,8R)-7-((1-Fluorocyclopropyl)methyl)-6-(4-((1-(3-Fluoropropyl)azetidin-3-yl) ) Amino)-2-methoxyphenyl)-8-methyl-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 1 3-((3,5-Difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-3- Trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenyl)amino)azetidine-1- Synthesis of tert-butyl formate
  • Step 2 (6S,8R)-6-(4-(azetidin-3-ylamino)-2,6-difluorophenyl)-8-methyl-7-(2,2,2 Synthesis of -trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Trifluoroacetic acid (15.5 g, 135 mmol) was dissolved in water (1 mL) and 3-((3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7- (2,2,2-Trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinoline-6- (400 mg, 493 umol) in tert-butyl) phenyl) amino) azetidine-1-carboxylate (400 mg, 493 umol), and reacted under nitrogen protection at 20°C for 2 hours.
  • LCMS detected the end of the reaction.
  • Step 3 (E)-4-(3-((3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-tri Fluoroethyl)-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenyl)amino)azetidine-1 -base)-N,N-dimethylbut-2-enamide synthesis
  • Example 36 (6S,8R)-6-(4-((1-Acryloylazetidin-3-yl)amino)-2,6-difluorophenyl)-8-methyl-7 -(2,2,2-Trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one (Compound 36)
  • Step 1 (6S,8R)-6-(4-((1-acryloylazetidin-3-yl)amino)-2,6-difluorophenyl)-8-methyl-7- Synthesis of (2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 1 (R)-3-(3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl) -3-Trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenoxy)pyrrolidine-1-carboxylic acid Synthesis of tert-butyl ester
  • Trifluoroacetic acid (6.16 g, 54.0 mmol) was dissolved in water (0.4 mL) and (R)-3-(3,5-difluoro-4-((6S,8R)-8-methyl-2- Oxo-7-(2,2,2-trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]iso quinolin-6-yl)phenoxy)pyrrolidine-1-carboxylate tert-butyl ester (503mg, 609umol), react under nitrogen protection at 20°C for 2 hours. LCMS detected the end of the reaction.
  • Step 3 (E)-4-((R)-3-((3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2, 2-Trifluoroethyl)-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenoxy)pyrrolidin-1-yl Synthesis of )-N,N-dimethylbut-2-enamide
  • Step 1 (6S,8R)-6-(4-(((R)-1-acryloylpyrrolidin-3-yl)oxy)-2,6-difluorophenyl)-8-methyl- Synthesis of 7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydrooxazolo[5,4-f]isoquinolin-2(3H)-one
  • Step 1 4-(3,5-Difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-3-tri Synthesis of Benzyl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenyl)piperazine-1-carboxylic acid tert-butyl ester
  • Trifluoroacetic acid (12.3 g, 107 mmol) was dissolved in water (0.1 mL) and 4-(3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7- (2,2,2-Trifluoroethyl)-3-trityl-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinoline-6- (413 mg, 500 umol) in tert-butyl) phenyl) piperazine-1-carboxylate (413 mg, 500 umol), react under nitrogen protection at 20°C for 2 hours. LCMS detected the end of the reaction.
  • Step 3 (E)-4-(4-(3,5-difluoro-4-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoro) ethyl)-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)phenyl)piperazin-1-yl)-N,N - Synthesis of dimethylbut-2-enamide
  • Example 42 3-(4-((2-(4-(3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-tri Fluoroethyl)-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)pyridin-3-yl)oxy)ethyl)piperidine Azin-1-yl)ethyl)amino)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (Compound 42)
  • Step 1 4-(2-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoroethyl)-2,3,6, Synthesis of 7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)pyridin-3-yl)oxy)ethyl)piperazine-1-carboxylic acid tert-butyl ester
  • Step 3 Synthesis of 3-(4-((2-bromoethyl)amino)-1-oxoisoindolin-2-yl)piperidine-2,6-dione
  • Step 4 3-(4-((2-(4-(3-((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-trifluoro) ethyl)-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)pyridin-3-yl)oxy)ethyl)piperazine Synthesis of -1-yl)ethyl)amino)-1-oxoisoindolin-2-yl)piperidine-2,6-dione
  • Example 43 Similar to the synthetic route of the previous Example 42, the following Example 43 was synthesized by selecting the corresponding reactants and the synthetic methods known to those skilled in the art.
  • Example 43 3-(4-((2-(4-(3-(((6-((6S,8R)-8-methyl-2-oxo-7-(2,2,2-tris) Fluoroethyl)-2,3,6,7,8,9-hexahydrooxazolo[5,4-f]isoquinolin-6-yl)pyridin-3-yl)oxy)propyl)piperidine Azin-1-yl)ethyl)amino)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (Compound 43)
  • AZD9496 catalog number: HY-12870
  • AZD9833 catalog number: HY-136255
  • tamoxifen catalog number: HY-13757A
  • Comparative Compound 1 chemical structure: ( 6S,8R)-6-(2,6-Difluoro-4-(2-(3-(fluoromethyl)azetidin-1-yl)ethoxy)phenyl)-7-(2 -Fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline) using the example in WO2017/182493A1 1 prepared by the method.
  • X is the "665 value to 615 value” for each concentration.
  • Min is the mean value of "665 to 615" with the addition of 0.2 mM positive control compound and 5 nl of 3 nM (1.5 nM) estradiol.
  • Max is the "665 value to 615 value” average of DMSO and 5nl 3nM (1.5nM) estradiol. Import the data into Graphpad Prism and use Log(agonist) vs.response--variable slope for curve fitting.
  • Example 1 9.00 100.2
  • Example 9 21.6 100.1
  • Example 12 15.6 100.3
  • Example 14 40.3 100.2
  • Example 25 11.4 100.8
  • Example 30 8.85 99.8
  • Example 31 45.6 100.9
  • Example 32 14.7 100.3
  • Example 33 11.8 99.9
  • Example 34 19.4 100.2 AZD9833 62.0 99.8 Tamoxifen 578.1 100.4
  • Human breast cancer cells MCF-7 (HTB-22, purchased from ATCC) were seeded in 96-well plates in medium containing 10% FBS (Gibco, 10099-141), 1% pyruvate (Sigma, S8636), 1% Non-essential amino acids (Sigma, M7145) in MEM (Hyclone, SH30024.01B) medium, seeded at a density of 4,000 cells/well, and cells were cultured at 37°C, 5% CO 2 .
  • Compounds were prepared as 20 mM stock solutions, serially diluted in 100% DMSO (Sigma, D2650) to a final concentration of 1000, and then diluted 20-fold in medium containing 2% FBS.
  • Example 1 0.39
  • Example 9 0.19
  • Example 10 0.20
  • Example 11 0.38
  • Example 12 0.49
  • Example 13 0.30
  • Example 14 0.44
  • Example 16 0.80
  • Example 25 0.24
  • Example 26 0.55
  • Example 27 1.63
  • Example 28 0.40
  • Example 29 0.71
  • Example 30 0.21
  • Example 31 0.85
  • Example 32 0.39
  • Example 33 0.39
  • Example 34 0.19
  • Example 35 0.82
  • Example 37 0.61
  • Example 39 0.49
  • Example 43 0.69 AZD9496* 3.84 AZD9833* 2.45 Tamoxifen* 274.50 Comparative Compound 1* 2.68
  • This test example uses ER ⁇ DuoSet IC ELISA Kit (R&D, DYC5715) to detect the degradation of ER ⁇ by compounds.
  • human breast cancer cells MCF-7 (source as above) were inoculated in 24-well plates; the medium was 10% activated carbon-treated FBS (Tocyto, UT61204), 1% pyruvate, and 1% non-essential amino acids.
  • FBS Tocyto, UT61204
  • 1% pyruvate 1% non-essential amino acids.
  • MEM medium seeding density was 150,000 cells/well; cells were cultured at 37°C, 5% CO2 for 24 hours.
  • the compounds to be tested were serially diluted and added to 24-well plates containing cells. Cells were incubated for 24 hours at 37°C, 5% CO2. ERa capture antibody was diluted to 1 ⁇ g/ml with PBS (Corning, 21-040-CVR), added at 100 ⁇ L/well to a 96-well plate, sealed and coated overnight at room temperature.
  • the coated 96-well plate was washed three times with PBS, 300 ⁇ L of blocking solution (R&D, DY995) was added to each well, and the plate was blocked at room temperature for 1 hour.
  • the 24-well plate was washed once with PBS, the residual liquid was removed, and 60 ⁇ L of lysate (6M urea (Sigma, U5378), 1 mM EDTA (Sigma, E9884), 0.5% TritonX-100 (Sigma, T8787), 1 mM urea (Sigma, T8787), 1 mM lysis buffer) was added to each well.
  • the T0 plate sample was first added with 600 ⁇ L of stop solution (acetonitrile: methanol (95:5, v/v) solution containing 100 ng/mL tolbutamide (Sigma, T0891)), and then NADPH regeneration system working solution was added.
  • stop solution acetonitrile: methanol (95:5, v/v) solution containing 100 ng/mL tolbutamide (Sigma, T0891)
  • test product supernatant was diluted into 200 ⁇ L of aqueous solution containing 0.3% formic acid for LC-MS/MS analysis, and 100 ⁇ L of the reference product supernatant was diluted to 300 ⁇ L of pure water was used for LC-MS/MS analysis and calculated according to the following formula:

Landscapes

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

Abstract

一种式(I)的化合物或其立体异构体、互变异构体或药用盐,及其在制备预防和/或治疗雌激素受体(ER)相关疾病或病症的药物中的用途。

Description

雌激素受体调节剂 技术领域
本发明涉及某些新颖化合物或其药用盐,它们具有抗癌活性并且因此潜在有用于治疗人体或动物体的方法。本发明还涉及用于制造所述化合物的方法、含有它们的药物组合物以及它们在治疗方法中的用途,例如用于制造供预防或治疗温血动物(如人)的癌症用的药物,包括用于预防或治疗雌激素受体依赖性或由雌激素受体介导的疾病。
本发明还涉及为雌激素受体的选择性下调调节剂的化合物。
背景技术
雌激素受体(ER)属于类固醇激素受体,包括ERα和ERβ两种亚型。ER参与女性生殖道的调节和发育;在乳腺癌等癌症中,肿瘤的生长与雌激素和ER受体的作用有关,例如多数乳腺癌患者的ER表达都有所升高。
已知一些化合物能够结合至ER,从而竞争性抑制ER与内源性雌激素配体(例如雌性性别类固醇激素17b雌二醇(E2))的结合,从而被用作ER抑制剂。例如,选择性雌激素受体调节剂(SERM)如他莫昔芬(Tamoxifen)、AZD9496、AZD9833等就被用作ER抑制剂,以治疗乳腺癌等。WO2017/182493A1也公开了一种SERM化合物。
尽管现有SERM疗法对乳腺癌治疗具有一定作用,但仍需要抑制活性更强并且代谢稳定性提高的ER抑制剂。
发明内容
本发明的发明人发现,如式(I)所示的化合物具有四氢噁唑并异喹啉-酮的基本结构,该结构具有显著的ER降解活性,因此出人意料地具有显著优于现有技术已知化合物的ER抑制活性;同时此类化合物还具有优异的代谢稳定性。因此,本发明的化合物能够用于治疗雌激素受体依赖性或由雌激素受体介导的疾病,由此完成了本发明。
具体地,本发明涉及以下内容。
本发明的一个方面提供式(I)的化合物,或其立体异构体或药学上可接受的盐:
Figure PCTCN2022080680-appb-000001
其中:
Z 1选自CR aR b、C(O)和键;
Z 2选自O、S、NR c、C(O)和键,或任选被一个或多个相同或不同的R d取代的C 1-C 6亚烷基、O-(C 1-C 6亚烷基)或NH-(C 1-C 6亚烷基);
Cy 1选自C 6-14亚芳基、C 3-8亚环烷基、C 5-14亚杂芳基、C 3-14亚杂环烷基、C 3-14亚杂环烯基,其各自任选被选自以下的基团取代:卤素原子、羟基、氨基、氰基以及任选被卤素原子取代的C 1-6烷基或C 1-6烷氧基;
Cy 2选自键、C 3-10亚环烷基、C 3-14亚杂环烷基,其各自任选被选自以下的基团取代:卤素原子(包括F、Cl、Br或I原子)、羟基、氨基、氰基以及任选被卤素原子取代的C 1-6 烷基或C 1-6烷氧基;
R 1、R 2各自独立地为H、卤素原子、羟基、氨基、氰基、C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基或氧代基的基团取代;
R 4为H、卤素原子、羟基、氨基、氰基、C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基、氧代基、C 6-14芳基、C 5-14杂芳基、C 3-14亚杂环烷基或
Figure PCTCN2022080680-appb-000002
(例如
Figure PCTCN2022080680-appb-000003
)
的基团取代;
优选地,R 4为H、卤素原子、羟基、氨基、氰基、C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基或氧代基的基团取代;
R 3为-(CR eR f) m-CR 31R 32R 33,其中m为1、2或3;
R 31、R 32和R 33独立选自:H、C 1-6烷基、卤素原子、氰基,且R 31和R 32可以共同形成C 3-8亚环烷基,所述C 1-6烷基和C 3-8亚环烷基任选被羟基、氰基、氨基或卤素原子取代;
R a、R b、R c、R d、R e、R f各自独立地为H、卤素原子、羟基、氨基、氰基、任选被卤素原子取代的C 1-6烷基。
在本说明书上下文中,所述杂芳基/亚杂芳基、杂环烷基/亚杂环烷基、杂环烯基/亚杂环烯基含有1-4个选自N、O、S的杂原子作为环构成原子。所述环烷基/亚环烷基、杂环烷基/亚杂环烷基、杂环烯基/亚杂环烯基可为不含有芳香性环结构的单环、双环或三环(优选单环或双环)环系统,其中双环或三环环系统包括螺环、桥接环或稠合环。所述芳基/亚芳基和杂芳基/亚杂芳基可为单环、稠合双环或稠合三环(优选单环或稠合双环)环系统,且其中所述稠合双环或稠合三环环系统中可含有非芳香性环结构。所述卤素原子选自F、Cl、Br或I原子。所述氧代基表示―=O‖基团。
在式(I)化合物的优选的实施方案中:
Z 1选自CR aR b、C(O)和键;
Z 2选自O、S、NR c、C(O)和键,或任选被一个或多个相同或不同的R d取代的C 1-C 6亚烷基、O-(C 1-C 6亚烷基)或NH-(C 1-C 6亚烷基);
Cy 1选自C 6-14亚芳基(优选C 6-10亚芳基)和C 5-14亚杂芳基(优选C 5-10亚杂芳基),其各自任选被选自以下的基团取代:卤素原子、以及任选被卤素原子取代的C 1-6烷基或C 1-6烷氧基;
Cy 2选自键、C 3-10亚环烷基(优选C 3-8亚环烷基)、C 3-14亚杂环烷基(优选C 3-10亚杂环烷基);
R 1、R 2各自独立地为H或C 1-6烷基,所述C 1-6烷基任选被一个或多个选自卤素原子、羟基或氰基的基团取代;
R 3为-(CR eR f) m-CR 31R 32R 33,其中m为1、2或3;
R 31、R 32和R 33独立选自:H、C 1-6烷基、卤素原子、氰基,且R 31和R 32可以共同形 成C 3-8亚环烷基,所述C 1-6烷基和C 3-8亚环烷基任选被羟基、氰基、氨基或卤素原子取代;
R a、R b、R c、R d、R e、R f各自独立地为H或卤素原子;
R 4为C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基、氧代基和
Figure PCTCN2022080680-appb-000004
的基团取代;
优选地,R 4为C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基或氧代基的基团取代。
在式(I)化合物的更优选的实施方案中:
Z 1为键;
Z 2选自-O-CH 2-CH 2-、-O-CH 2-、-NH-CH 2-CH 2-、-NH-CH 2-、-NH-或-O-;
Cy 1为C 6-10亚芳基(优选苯基)和C 5-10亚杂芳基(优选吡啶基),其任选被卤素原子(优选F)或C 1-6烷氧基(优选甲氧基)取代;
Cy 2选自键、亚氮杂环丁基、亚吡咯烷基、亚哌嗪基、
Figure PCTCN2022080680-appb-000005
R 1为C 1-6烷基(优选甲基);
R 2为H;
R 3为-CH 2-CR 31R 32R 33
R 31、R 32和R 33独立选自:H、任选被羟基或卤素原子(优选F)取代的C 1-6烷基(优选甲基)、卤素原子(优选F)、氰基,且R 31和R 32可以共同形成任选被羟基或卤素原子(优选F)取代的C 3-8亚环烷基(优选亚环丙基);
R 4为任选被选自卤素原子(优选F)、氧代基、C 1-6烷基氨基(优选甲基氨基)、(C 1-6烷基) 2氨基(优选二甲基氨基)、和
Figure PCTCN2022080680-appb-000006
的基团取代的C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基。
优选地,Cy 2选自键、亚氮杂环丁基、亚吡咯烷基,且R 4为任选被选自卤素原子(优选F)、氧代基、C 1-6烷基氨基(优选甲基氨基)和(C 1-6烷基) 2氨基(优选二甲基氨基)的基团取代的C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基。
在式(I)化合物的特别优选的实施方案中:
Z 1为键;
Z 2选自-NH-或-O-;
Cy 1为苯基或吡啶基(优选苯基),其任选被F或甲氧基取代;
Cy 2选自亚氮杂环丁基、亚吡咯烷基、亚哌嗪基;
R 1为甲基;
R 2为H;
R 3为-CH 2-CR 31R 32R 33
R 31、R 32各为F,或R 31和R 32共同形成亚环丙基;
R 33选自:H、羟甲基、氟甲基,优选选自F、羟甲基、氟甲基;
R 4为F-(CH 2) 3-或
Figure PCTCN2022080680-appb-000007
进一步优选地,Cy 2选自亚氮杂环丁基和亚吡咯烷基,且R 4为F-(CH 2) 3-。
在一些实施方案中,本发明的式(I)的化合物呈立体异构体的形式,例如在四氢异喹啉环的1位的碳(即Z 1所连接的碳)可为S构型,和/或3位的碳(即R 1所连接的碳)可为R构型。
在一些实施方案中,本发明涉及以下化合物或其立体异构体或药学上可接受的盐:
Figure PCTCN2022080680-appb-000008
Figure PCTCN2022080680-appb-000009
Figure PCTCN2022080680-appb-000010
在另一方面中,本发明涉及一种药物组合物,其包含本发明的前述任一种或多种化合物或其立体异构体或药学上可接受的盐,以及药学上可接受的载体。
在另一方面中,本发明涉及本发明的化合物在制备药物中的用途,所述药物用于治疗哺乳动物的雌激素受体依赖性或由雌激素受体介导的疾病。
具体实施方案
下面通过具体的制备实施例和测试例进一步说明本发明。应认识到所述实施例和测试例并非意在限制本发明的范围。实施例中所用的原料、试剂等,均为本领域技术人员所公知并且可通过市售或文献方法获得的物质;所用的试验或表征方法也是本领域技术人员公知的方法。
实施例1:(6S,8R)-6-(2,6-二氟苯基-4-(2-(3-(氟甲基)-氮杂环丁烷-1-基-乙氧基-苯基)-7-异丁基-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物1)
Figure PCTCN2022080680-appb-000011
步骤一:7-溴苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000012
将2-氨基-6-溴苯酚(5g,26.6mmol,1eq)溶于120ml四氢呋喃,在20℃下加入二羰基咪唑(5.17g,31.9mmol,1.2eq),反应液在80℃条件下反应2小时。TLC检测反应结束,将反应液倒入冰水中用盐酸调节pH=2,乙酸乙酯萃取干燥浓缩,粗品柱层析分离得到红色固体7-溴苯并[d]噁唑-2(3H)-酮(5.1g,23.8mmol,89.6%收率)。
1H NMR(400MHz,DMSO-d6):δ=12.0(s,1H),7.30–7.27(m,1H),7.11–7.09(m,2H)ppm。
步骤二:7-溴-3-三苯甲基苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000013
将7-溴苯并[d]噁唑-2(3H)-酮(5.1g,23.8mmol,1eq)溶于50mL N,N-二甲基甲酰胺,在0℃下加入氢化钠(1.43g,35.7mmol,60%纯度,1.5eq),反应液在0℃搅拌30分钟后加入三苯甲基氯甲烷(6.64g,23.8mmol,1eq),反应液在20℃下反应2小时。TLC检测反应结束,将反应液倒入冰水中用乙酸乙酯萃取干燥浓缩,粗品柱层析分离得到白色固体7-溴-3-三苯甲基苯并[d]噁唑-2(3H)-酮(9.5g,20.8mmol,87.4%收率)。
1H NMR(400MHz,CDCl 3):δ=7.40-7.37(m,6H),7.25-7.20(m,9H),7.12(d,J=8.0Hz,1H),6.62(t,J=8.4Hz,1H),5.93(d,J=8.0Hz,1H)ppm。
步骤三:(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]噁唑-7-基)丙烷-2-基)氨基甲酸叔 丁酯的合成
Figure PCTCN2022080680-appb-000014
将7-溴-3-三苯甲基苯并[d]噁唑-2(3H)-酮(4.0g,8.77mmol,1eq)溶于四氢呋喃(20mL)在-78℃氮气保护下加入正丁基锂(2.5M,4.03mL,1.15eq)搅拌30分钟。将(R)-叔丁基4-甲基-1,2,3-噁唑烷-3-羧酸2,2-二氧化物(2.50g,10.5mmol,1.2eq)的四氢呋喃(10mL)溶液在-78℃加入体系,然后自然恢复到0℃反应2个小时。TLC检测反应完成,将反应液倒入冰水中淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到白色固体(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]噁唑-7-基)丙烷-2-基)氨基甲酸叔丁酯(4.5g,8.42mmol,76.8%收率)。
1H NMR(400MHz,CDCl 3):δ=7.50-7.48(m,6H),7.33-7.28(m,9H),6.88-6.78 (m,1H),6.76–6.74(m,1H),5.92(br d,J=8.0Hz,1H),4.22-4.11(m,1H),2.84(br d,J=6.4Hz,2H),1.58(s,9H),1.16(d,J=6.4Hz,3H)ppm。
步骤四:(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000015
在20℃下将(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]噁唑-7-基)丙烷-2-基)氨基甲酸叔丁酯(4.3g,8.04mmol,1eq)加入到盐酸甲醇溶液(4M,30mL,14.9eq).反应2个小时,TLC和LCMS检测反应完成,反应液浓缩,加入饱和碳酸氢钠中和,乙酸乙酯萃取浓缩得到粗品,用30ml石油醚:乙酸乙酯=3:1的混合溶剂打浆过滤得到白色固体(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮(1.9g,4.37mmol,54.4%收率)。
1H NMR(400MHz,DMSO-d6):δ=7.50-7.48(m,6H),7.35-7.25(m,9H),6.92-6.87(m,1H),6.86-6.78(m,1H),5.92(d,J=7.2Hz,1H),3.11-3.06(m,1H),2.60-2.57(m,2H),0.97(d,J=6.4Hz,3H)ppm。
LCMS:m/z(M+H) +=435.1。
步骤五:(R)-7-(2-(异丁胺)丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000016
将(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮(1.65g,3.38mmol,1eq)和2-甲基丙醛(292mg,4.06mmol,370uL,1.2eq)溶于30ml甲醇中,在20℃下向反应液中加入乙酸(406mg,6.76mmol,387uL,2eq)和氰基硼氢化钠(425mg,6.76mmol,2eq)。反应两个小时后,TLC和LCMS监测反应完成,反应液浓缩后加入饱和碳酸氢钠中和,乙酸乙酯萃取干燥浓缩,柱层析分离得到黄色固体(R)-7-(2-(异丁胺)丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮(1.70g,100%收率)。
1H NMR(400MHz,CDCl 3):δ=7.46-7.48(m,6H),7.30-7.28(m,9H),6.93(d,J=7.2Hz,1H),6.82(t,J=8.0Hz,1H),5.99(d,J=8.0Hz,1H),4.13(q,J=7.2Hz,1H),3.57-3.53(m,1H),3.26-3.21(m,1H),2.99–2.94(m,1H),2.79-2.73(m,2H),1.31-1.29(m,3H),0.97-0.94(m,6H)ppm。
LCMS:m/z(M+H) +=491.4。
步骤六:(R)-7-(2-(异丁胺)丙基)苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000017
将(R)-7-(2-(异丁胺)丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮(1.7g,3.46mmol,1eq)溶于20ml三氟乙酸和2ml水的混合溶剂中,在20℃下反应3个小时。TLC和LCMS监测反应完全,反应液在50℃下浓缩后加入饱和碳酸氢钠调节pH成中性,乙酸乙酯萃取,干燥浓缩有机相,粗品经柱层析分离得到黄色固体(R)-7-(2-(异丁胺)丙基)苯并[d]噁唑-2(3H)-酮(650mg,2.62mmol,75.5%收率)。
1H NMR(400MHz,CDCl 3):δ=7.08-7.06(m,1H),6.96–6.93(m,2H),3.25(br d,J=5.6Hz,1H),3.04–2.99(m,1H),2.83-2.80(m,1H),2.61-2.58(m,2H),1.84–1.77 (m,1H),1.18(d,J=6.4Hz,3H),0.90(t,J=6.4Hz,6H)ppm。
LCMS:m/z(M+H) +=249.2。
步骤七:(6S,8R)-6-(4-溴-2,6-二氟苯基)-7-异丁基-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异 喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000018
将(R)-7-(2-(异丁胺)丙基)苯并[d]噁唑-2(3H)-酮(600mg,1eq)和4-溴-2,6-二氟苯甲醛(800mg,1.5eq)溶于30ml甲苯并加入醋酸(450mg,3eq)。反应夜在115℃条件下反应24个小时。TLC和LCMS检测到25%原料剩余和50%的产物生成,反应液在60℃下浓缩,粗品柱层析分离得到黄色固体(6S,8R)-6-(4-溴-2,6-二氟苯基)-7-异丁基-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(460mg,42.2%收率)。
1H NMR(400MHz,CDCl 3):δ=7.59(br s,1H),6.94(br d,J=8.4Hz,2H),6.67(d,J=8.4Hz,1H),6.49(d,J=8.4Hz,1H),5.04(s,1H),3.45-3.39(m,1H),3.09-3.04(m,1H),2.77–2.72(m,1H),2.38–2.34(m,1H),1.89-1.84(m,1H),1.60-1.58(m,1H),0.91(br d,J=6.4Hz,3H),0.74(br d,J=6.4Hz,3H),0.60(br d,J=6.4Hz,3H)ppm。
LCMS:m/z(M+H) +=450.9
步骤八:(6S,8R)-6-(4-溴-2,6-二氟苯基)-7-异丁基-8-甲基-3-三苯甲基-6,7,8,9-四氢噁 唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000019
将(6S,8R)-6-(4-溴-2,6-二氟苯基)-7-异丁基-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(240mg,532μmol,1eq)溶于10mlN,N-二甲基甲酰胺,在0℃下加入氢化钠(42.5mg,1.06mmol,60%纯度,2eq)。然后0℃向反应液中加入三苯甲基氯甲烷(163mg,585μmol,1.1eq),反应缓慢升至20℃反应2个小时。TLC和LCMS检测反应完成,将反应液倒入水中淬灭,乙酸乙酯萃取,有机相干燥浓缩,粗品柱层析分离得到黄色固体(6S,8R)-6-(4-溴-2,6-二氟苯基)-7-异丁基-8-甲基-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(420mg,460μmol,86.5%收率)。
1H NMR(400MHz,CDCl 3):δ=7.48-7.46(m,6H),7.31-7.29(m,9H),6.99(d,J=8.4Hz,2H),6.24-6.16(m,1H),5.66-5.62(m,1H),5.00(s,1H),3.48-3.41(m,1H),3.09-3.04(m,1H),2.79–2.75(m,1H),2.42–2.37(m,1H),1.90–1.85(m,1H),1.64-1.60(m,1H),0.95-0.92(m,3H),0.78(d,J=6.4Hz,3H),0.66(d,J=6.4Hz,3H)ppm。
LCMS:m/z(M+H) +=695.0。
步骤九:2-(3-(氟甲基)氮杂环丁烷-1-基)乙醇的合成
Figure PCTCN2022080680-appb-000020
将3-(氟甲基)氮杂环丁烷盐酸盐(3g,23.9mmol,1eq)溶于四氢呋喃(50mL)在20℃下加入1,8-二氮杂环十一烯(10.9g,71.7mmol,10.8mL,3eq)。在搅拌15分钟以后加入2-溴乙醇(5.97g,47.8mmol,2eq)反应12个小时。TLC检测反应完成,将反应液调节pH大于9,乙酸乙酯萃取,有机相干燥浓缩,粗品柱层析分离得到黄色油状产物2-(3-(氟甲基)氮杂环丁烷-1-基)乙醇(950mg,7.13mmol,29.8%收率)。
1H NMR(400MHz,CDCl 3):δ=4.57-4.42(m,1H),4.41–4.38(m,1H),3.53–3.50(m,2H),3.43–3.40(m,2H),3.10–3.08(m,2H)2.85–2.80(m,1H),2.61–2.59(m,2H)。
步骤十:(6S,8R)-6-(2,6-二氟苯基-4-(2-(3-(氟甲基)-氮杂环丁烷-1-基-乙氧基-苯基)-7- 异丁基-8-甲基-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000021
将(6S,8R)-6-(4-溴-2,6-二氟苯基)-7-异丁基-8-甲基-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(300mg,329μmol,1eq)和2-(3-(氟甲基)氮杂环丁烷-1-基)乙醇(109mg,822μmol,2.5eq)溶于甲苯(9ml)加入[(2-2-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯)]钯(II)甲磺酸盐(27.6mg,32.8μmol,0.1eq)和碳酸铯(321mg,986μmol,3eq),反应液在110℃反应24小时。TLC和LCMS监测反应完成,反应液浓缩后柱层析分离得到黄色固体(6S,8R)-6-(2,6-二氟苯基-4-(2-(3-(氟甲基)-氮杂环丁烷-1-基-乙氧基-苯基)-7-异丁基-8-甲基-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(90mg,121μmol,36.7%收率)。
1H NMR(400MHz,CDCl 3):δ=7.47-7.46(m,6H),7.26-7.23(m,9H),6.34(d,J=10.4Hz,2H),6.23(d,J=8.4Hz,1H),5.61(d,J=8.4Hz,1H),4.95-4.93(m,1H),4.57(d,J=5.6Hz,1H),4.45(d,J=5.6Hz,1H),3.91(q,J=5.6Hz,2H),3.50-3.42(m,5H),3.17-3.16(m,2H),3.06-3.04(m,1H),2.84–2.77(m,2H),2.76–2.72(m,1H),2.37-2.32(m,1H),1.92(m,1H),0.95-0.92(m,3H),0.78(d,J=6.4Hz,3H),0.69-0.65(m,3H)ppm。
LCMS:m/z(M+H) +=742.2。
步骤十一:(6S,8R)-6-(2,6-二氟苯基-4-(2-(3-(氟甲基)-氮杂环丁烷-1-基-乙氧基-苯 基)-7-异丁基-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成(化合物1)
Figure PCTCN2022080680-appb-000022
将(6S,8R)-6-(2,6-二氟苯基-4-(2-(3-(氟甲基)-氮杂环丁烷-1-基-乙氧基-苯基)-7-异丁 基-8-甲基-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(90mg,120.66μmol,1eq)溶于三氟乙酸(9mL)和水(1mL)的混合溶液,在20℃反应2个小时。LCMS和TLC监测反应完成,反应液在40℃下浓缩,浓缩后加饱和碳酸氢钠中和,乙酸乙酯萃取,干燥浓缩有机相,粗品通过薄层层析硅胶板分离得到黄色固体(6S,8R)-6-(2,6-二氟苯基-4-(2-(3-(氟甲基)-氮杂环丁烷-1-基-乙氧基-苯基)-7-异丁基-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(35mg,67.4μmol,55.8%收率,97%纯度,89%ee)。
1H NMR(400MHz,DMSO-d6):δ=6.70(d,J=8.0Hz,1H),6.57(d,J=8.0Hz,1H),6.31(br d,J=10.4Hz,2H),5.06(s,1H),4.56(d,J=5.4Hz,1H),4.44(d,J=5.4Hz,1H),3.93-3.92(m,2H),3.56–3.54(m,2H),3.45–3.42(m,1H),3.20-3.10(m,3H),2.89–2.86(m,2H),2.82–2.77(m,1H),2.40-2.32(m,1H),2.01–1.96(m,1H),1.26(s,2H),0.99(d,J=6.4Hz,3H),0.81(d,J=6.4Hz,3H),0.68(d,J=6.4Hz,3H)ppm。
LCMS:m/z(M+H) +=504.4。
实施例5:(6S,8R)-6-(5-((1-(3-氟丙烷)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物5)
Figure PCTCN2022080680-appb-000023
步骤一:(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]噁唑-7-基)丙烷-2-基)氨基甲酸叔 丁酯的合成
Figure PCTCN2022080680-appb-000024
将7-溴-3-三苯甲基-苯并噁唑-2酮(100g,219mmol,1eq)溶于四氢呋喃(1000mL)在-78℃氮气保护下加入正丁基锂(2.5M,87.6mL,1eq)搅拌30分钟。将(R)-叔丁基4-甲基-1,2,3-噁唑烷-3-羧酸2,2-二氧化物(52.0g,219mmol,1eq)的四氢呋喃(100mL)溶液在-78℃加入体系,然后自然恢复到0℃反应2个小时。TLC检测反应完成,将反应液倒入饱和氯化铵水溶液中淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到黄色固体(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]噁唑-7-基)丙烷-2-基)氨基甲酸叔丁酯(360g,673mmol,74.9%收率)。
1H NMR(400MHz,CDCl 3):δ=7.52-7.47(m,6H),7.35-7.27(m,9H),6.92-6.85(m,1H),6.76(t,J=8.0Hz,1H),5.93(br d,J=8.0Hz,1H),4.00(s,1H),3.81-3.72(m,1H),2.84(br d,J=6.0Hz,2H),1.40(s,9H),1.16(d,J=6.4Hz,3H)ppm。
步骤二:(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000025
在0℃氮气保护下将(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]噁唑-7-基)丙烷-2-基) 氨基甲酸叔丁酯(240g,448mmol,1eq)溶于甲醇(400ml),然后加入到盐酸甲醇溶液(6M,800mL,10.6eq).在0℃反应2个小时,TLC检测反应完成。反应液浓缩,加入饱和碳酸氢钠中和,乙酸乙酯萃取浓缩得到粗品,用石油醚:乙酸乙酯=1:1打浆过滤得到黄色固体(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮(198g,455mmol,84.6%收率)。
1H NMR(400MHz,CDCl 3):δ=7.48-7.34(m,6H),7.23-7.14(m,9H),6.81-6.73(m,1H),6.66(t,J=8.0Hz,1H),5.88(m,1H),3.33(s,J=6.4Hz,1H),2.82-2.65(m,2H),1.12(d,J=6.4Hz,3H)ppm。
步骤三:(R)-7-2-氨基丙基)(2,2,2-三氟乙基-3-三苯甲基苯并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000026
在20℃氮气保护下将(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮(10.0g,23.0mmol,1eq),2,2,2-三氟乙基三氟甲磺酸酯(6.14g,26.5mmol,1.15eq),二异丙基乙二胺(8.92g,69.0mmol,12.0mL,3eq)溶于二噁烷(100mL)里,然后在80℃氮气保护下反应10小时。TLC检测反应完成,反应液浓缩,粗产品柱层析分离得到白色固体(R)-7-2-氨基丙基)(2,2,2-三氟乙基-3-三苯甲基苯并[d]噁唑-2(3H)-酮(9.00g,17.4mmol,75.7%收率)。
1H NMR(400MHz,CDCl 3):δ=7.48(br d,J=7.2Hz,6H),7.33-7.27(m,9H),6.86(br d,J=7.6Hz,1H),6.79-6.73(m,1H),5.95(br d,J=8.0Hz,1H),3.50(s,1H),3.26(br d,J=9.3Hz,3H),2.98-2.85(m,1H),2.82-2.71(m,1H),1.15(br d,J=5.6Hz,3H)ppm。
步骤四:(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢异喹啉并 [5,4-f]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000027
将(R)-7-2-氨基丙基)(2,2,2-三氟乙基-3-三苯甲基苯并[d]噁唑-2(3H)-酮(1.00g,1.94mmol,1eq)和5-溴-2-吡啶甲醛(540mg,2.90mmol,1.5eq)溶于甲苯(50mL)中加入三氟乙酸(662mg,5.81mmol,430uL,3eq),混合物在90℃反应20小时。TLC检测反应完成,反应液浓缩,粗产品by-TLC(石油醚:乙酸乙酯=10:1)纯化得黄色固体(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢异喹啉并[5,4-f]噁唑-2(3H)-酮(780mg,1.76mmol,91.1%收率)。
1H NMR(400MHz,CDCl 3):δ=8.89(s,1H),8.54(d,J=2.4Hz,1H),7.84-7.80(m,1H),7.48(d,J=8.4Hz,1H),6.79-6.74(m,2H),5.09(s,1H),4.13(q,J=7.2Hz,1H),3.50-3.39(m,1H),3.35-3.22(m,1H),3.09-3.03(m,1H),3.00-2.89(m,1H),2.70-2.64(m,1H),1.13(d,J=6.4Hz,3H)ppm。
步骤五:(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四 氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000028
将(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢异喹啉[5,4-f]噁唑-2(3H)-酮(1.00g,2.26mmol,1eq)溶于N,N-二甲基甲酰胺(20mL)中,在0℃加入氢化钠(181mg,4.52mmol,60%纯度,2eq),然后在20℃加入三苯甲基氯甲烷(693mg,2.49mmol,1.1eq)并反应2小时。TLC检测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩,粗品柱层析分离得到白色固体(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(1.20g,1.56mmol,69.0%收率)。
1H NMR(400MHz,CDCl 3):δ=8.52(s,1H),7.75(dd,J 1=2.4Hz,J 2=8.4Hz,1H),7.49-7.45(m,6H),7.36(d,J=8.4Hz,1H),7.32-7.27(m,9H),6.39(d,J=8.4Hz,1H),5.75(d,J=8.4Hz,1H),4.94(s,1H),3.53-3.42(m,1H),3.29-3.18(m,1H),3.01(dd,J 1=4.8Hz,J 2=16.8Hz,1H),2.90(br dd,J 1=9.2Hz,J 2=15.6Hz,1H),2.68(dd,J 1=6.8Hz,J 2=16.8Hz,1H),1.10(d,J=6.4Hz,3H)ppm。
步骤六:3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六 氢噁唑并[5,4-f]异喹啉-6基)吡啶-3-基)氨基)氮杂环丁烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000029
(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢异喹啉并[5,4-f]噁唑-2(3H)-酮(200mg,292μmol,1eq)和3-氨基氮杂环丁烷-1-甲酸叔丁酯(126mg,730μmol,2.5eq)溶于8ml甲苯中,加入[(2-2-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯)]钯(II)甲磺酸盐(24.5mg,29.2μmol,0.1eq)和碳酸铯(286mg,876μmol,3eq),反应液在110℃反应15小时。TLC检测反应完成,反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到黄色固体3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6基)吡啶-3-基)氨基)氮杂环丁烷-1-甲酸叔丁酯(120mg,137μmol,47.2%收率)。
1H NMR(400MHz,CDCl 3):δ=7.77(d,J=2.4Hz,1H),7.52-7.42(m,7H),7.32-7.28(m,4H),7.26-7.17(m,9H),6.89-6.69(m,1H),6.37(d,J=8.4Hz,1H),5.80-5.64(m,1H),5.39-5.20(m,1H),4.98-4.77(m,1H),4.33-4.27(m,2H),4.23-4.11(m,1H),4.08-3.98(m,1H),3.74(m,2H),3.53-3.43(m,2H),3.26-3.11(m,1H),3.04-2.85(m,2H),2.72-2.59(m,1H),1.45(s,9H),1.09(d,J=6.4Hz,3H)ppm。
步骤七:(6S,8R)-6-(5-(氮杂环丁烷-3-氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙 基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000030
在0℃下将三氟乙酸(9.00mL)和水(1.00mL)加入3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6基)吡啶-3-基)氨基)氮杂环丁烷-1-甲酸叔丁酯(120mg,154μmol,1eq)中,混合物在20℃反应2小时,TLC监测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩,得到黄色固体(6S,8R)-6-(5-(氮杂环丁烷-3-氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(120mg,粗产品)。
1H NMR(400MHz,CDCl 3):δ=8.21-7.93(m,2H),7.60-7.35(m,2H),6.95-6.62(m,1H),5.55-5.17(m,1H),4.39-4.13(m,1H),4.06-3.90(m,1H),3.63-3.42(m,1H),2.33-2.13(m,2H),2.07-1.96(m,2H),1.14-1.08(m,2H),0.88(br d,J=4.8Hz,3H)ppm。
步骤八:(6S,8R)-6-(5-((1-(3-氟丙烷)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基 -7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成(化合物5)
Figure PCTCN2022080680-appb-000031
在20℃氮气保护下将(6S,8R)-6-(5-(氮杂环丁烷-3-氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(66.0mg,152μmol,1eq)和3-氟-1-碘丙烷(28.6mg,152μmol,1eq)溶于N,N-二甲基甲酰胺(1mL)中加入二异丙基乙二胺(98.4mg,761μmol,132uL,5eq),混合物在20℃反应4小时。LCMS监测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗产品通过反相柱分离,然后再经过手型柱分离得到黄色固体(6S,8R)-6-(5-((1-(3-氟丙烷)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(27.0mg,54.2μmol,35.6%收率,97.7%纯度,100%ee)。
1H NMR(400MHz,MeOH-d 4):δ=7.76-7.71(m,1H),7.13(d,J=8.4Hz,1H),6.93(m,1H),6.80(d,J=8.0Hz,1H),6.56(d,J=8.0Hz,1H),4.52(t,J=6.0Hz,1H),4.40(t,J=6.0Hz,1H),4.13-4.06(m,1H),3.82-3.76(m,2H),3.54-3.47(m,1H),3.15-3.08(m,1H),3.00-2.91(m,3H),2.76(m,1H),2.64(t,J=7.6Hz,2H),1.83-1.69(m,2H),1.06(d,J=6.6Hz,3H)ppm。
LCMS:m/z(M+H) +=494.3。
实施例7:(6S,8R)-6-(5-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物7)
Figure PCTCN2022080680-appb-000032
步骤一:(S)-3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9- 六氢噁唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000033
(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(150mg,219μmol,1eq)和(S)-3-羟基吡咯烷-1-甲酸叔丁酯(102mg,547μmol,2.5eq)溶于甲苯(20mL)后加入(18.3mg,21.9μmol,0.1eq)和碳酸铯(214mg,657μmol,3eq),反应液在115℃反应15小时。TLC监测反应完成,反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到黄色固体(S)-3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧)吡咯烷-1-甲酸叔丁酯(120mg,138μmol,31.5%收率)。
1H NMR(400MHz,CDCl 3):δ=8.02(d,J=2.8Hz,1H),7.39(d,J=7.2Hz,6H),7.24-7.20(m,5H),7.18-7.14(m,4H),7.08-7.04(m,1H),6.37-6.25(m,1H),5.66(d,J=8.4Hz,1H),4.86-4.80(m,2H),3.55-3.34(m,2H),3.34-3.23(m,1H),3.16-3.04(m,1H),3.01-2.89(m,1H),2.83(br dd,J 1=8.8Hz,J 2=15.6Hz,1H),2.59(dd,J 1=6.4Hz,J 2=16.8Hz,1H),2.15-2.01(m,2H),1.95-1.81(m,2H),1.40(s,9H),1.02(d,J=6.4Hz,3H)ppm。
步骤二:(6S,8R)-8-甲基-6-(5-((S)-吡咯烷-3-氧基)吡啶-2-基)-7-(2,2,2-三氟乙 基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000034
在0℃下将三氟乙酸(1.54g,13.4mmol,997uL,87.3eq)加入溶于二氯甲烷(2mL)的(S)-3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧)吡咯烷-1-甲酸叔丁酯(120mg,154μmol,1eq)中,在20℃下反应2小时,TLC和LCMS监测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗产品薄层层析硅胶板分离得到黄色固体(6S,8R)-8-甲基-6-(5-((S)-吡咯烷-3-氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(105mg,粗产品)。
1H NMR(400MHz,DMSO-d6):δ=8.13(d,J=2.8Hz,1H),7.45-7.34(m,1H),7.32-7.26(m,1H),6.87-6.82(m,1H),6.74-6.67(m,1H),5.14-5.02(m,2H),3.00-2.92(m,2H),2.90-2.78(m,2H),2.72-2.71(m,1H),2.74-2.58(m,1H),2.21-2.07(m,2H),2.06-1.93(m,1H),1.23(s,1H),1.06(d,J=6.4Hz,3H)ppm。
LCMS:m/z(M+H) +=449.2。
步骤三:(6S,8R)-6-(5-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)吡啶-2-基)-8-甲基-7-(2,2,2- 三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成(化合物7)
Figure PCTCN2022080680-appb-000035
在20℃氮气保护下将(6S,8R)-8-甲基-6-(5-((S)-吡咯烷-3-氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(105mg,187μmol,1eq)和3-氟-1-碘丙烷(35.2mg,187μmol,1eq)溶于N,N-二甲基甲酰胺(2mL)中加入二异丙基乙二胺(121mg,936μmol,163uL,5eq),混合物在20℃反应4小时。LCMS检测反应完成,反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗产品通过反相柱分离得到白色固体(6S,8R)-6-(5-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(10.5mg,19.8μmol,10.6%收率,96%纯度,100%ee)。
1H NMR(400MHz,MeOH-d 4):δ=8.05(d,J=2.8Hz,1H),7.40-7.30(m,2H),6.83(d,J=8.0Hz,1H),6.63(d,J=8.0Hz,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.53-3.48(m,1H),3.40(br d,J=6.0Hz,1H),3.10(dd,J 1=4.8Hz,J 2=16.8Hz,1H),3.02-2.87(m,4H),2.78(dd,J 1=6.2Hz,J 2=16.8Hz,1H),2.68-2.58(m,2H),2.56-2.49(m,1H),2.38(dt,J 1=7.6Hz,J 2=13.6Hz,1H),1.99-1.84(m,3H),1.29(br s,1H),1.12(d,J=6.4Hz,3H)ppm。
LCMS:m/z(M+H) +=509.4。
实施例8:(6S,8R)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物8)
Figure PCTCN2022080680-appb-000036
步骤一:(6S,8R)-6-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧-7-(2,2,2-三氟乙基)-3-三苯甲基 -2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6-基)苯基)-2,6-二氮螺[3.3]庚烷-2-甲酸叔丁酯的合
Figure PCTCN2022080680-appb-000037
将(6S,8R)-6-(4-溴-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(300mg,417μmol),2,6-二氮螺[3.3]庚烷-2-甲酸叔丁酯(165.33mg,833.88μmol)溶于甲苯(9mL),加入碳酸铯(272mg,834μmol)和[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯基)]合钯(II)甲磺酸盐(34.96mg,41.69μmol),氮气保护,在110℃反应24小时,LCMS检测反应结束,反应液浓缩后用硅胶柱纯化得到(6S,8R)-6-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢异喹啉并[5,4-f]噁唑-6-基)苯基)-2,6-二氮螺[3.3]庚烷-2-甲酸叔丁酯(120mg,143μmol,收率34.4%)。
1H NMR(400MHz,CDCl 3):δ=7.41-7.37(m,6H),7.24-7.19(m,9H),6.17(d,J=8.5Hz,1H),5.77(d,J=10.5Hz,2H),5.56(d,J=8.5Hz,1H),5.03(s,1H),4.02(s,4H),3.88(s,4H),3.50-3.44(m,1H),3.11-2.99(m,2H),2.85-2.75(m,1H),2.71-2.62(m,1H),1.37(s,9H),0.96(d,J=6.4Hz,3H)ppm。
LCMS:m/z 837.3[M+H] +
步骤二:(6S,8R)-6-(2,6-二氟-4-(2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟 乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉2(3H)-酮的合成
Figure PCTCN2022080680-appb-000038
在0℃将6-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6-基)苯基)-2,6-二氮螺[3.3]庚烷-2-甲酸叔丁酯(300mg,358.47μmol)溶于三氟乙酸(4.5mL)和水(0.5mL)中,升温至20℃搅拌2小时,LCMS检测反应结束,浓缩后调pH=7-8,用水和乙酸乙酯萃取,有机相浓缩得到(6S,8R)-6-(2,6--二氟-4-(2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉2(3H)-酮(350mg,粗品)。
LCMS:m/z 495.0[M+H] +
步骤三:(6S,8R)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基 -7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000039
将(6S,8R)-6-(2,6--二氟-4-(2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉2(3H)-酮(300mg,606.73μmol),1-碘-3-氟乙烷(211.09mg,1.21mmol)溶于N,N-二甲基甲酰胺(15mL),然后加入N,N-二异丙基乙胺(313.66mg,2.43mmo)20℃反应2小时,LCMS检测反应结束,用水和乙酸乙酯萃取,有机相用盐水洗三次,浓缩后用硅胶柱纯化得到(6S,8R)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(95mg,175.76μmol,收率28.9%)。
LCMS:m/z 541.3[M+H] +
步骤四:(6S,8R)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基 -7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的拆分(化合物8)
Figure PCTCN2022080680-appb-000040
将(6S,8R)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮手性拆分得到(6S,8R)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(40mg,74.01μmol,收率44.4%)
1H NMR(400MHz,MeCN-d 3)δ=6.87-6.72(m,1H),6.64-6.53(m,1H),6.03-5.86(m,2H),5.21(s,1H),4.46-4.41(m,1H),4.34-4.30(m,1H),3.89(s,4H),3.56-3.46(m,1H),3.35(s,4H),3.33-3.25(m,1H),3.05(dd,J=4.9,16.4Hz,1H),2.93(qd,J=9.8,15.9Hz,1H),2.74(dd,J=5.1,16.5Hz,1H),2.70-2.66(m,1H),2.63-2.58(m,1H),1.06(d,J=6.5Hz,3H)ppm。
实施例9:(6S,8R)-7-(2,2-二氟-3-羟基丙基)-6-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)苯基)-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物9)
Figure PCTCN2022080680-appb-000041
步骤一:(R)-7-(2-((3-((叔丁基苯基硅基)氧基)-2,2-二氟丙基)胺基)丙基)-3-三苯基苯 并[d]噁唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000042
后续步骤:按照与实施例1-8类似的步骤和/或本领域常规的反应步骤,从 (R)-7-(2-((3-((叔丁基苯基硅基)氧基)-2,2-二氟丙基)胺基)丙基)-3-三苯基苯并[d]噁唑 -2(3H)-酮起始合成化合物9。
1H NMR:(400MHz,ACETONITRILE-d3)δ=11.6(s,1H),9.16(s,1H),6.81(d,J=8.0Hz,1H),6.60(br d,J=8.0Hz,1H),6.13-6.10(m,2H),5.18(s,1H),4.61-4.52(m,2H),4.44-4.33(m,2H),4.22-4.12(m,2H),3.81-3.46(m,4H),3.36-3.24(m,2H),3.18-2.98(m,2H),2.80-2.70(m,2H),2.02-1.95(m,2H),1.05(d,J=6.4Hz,3H)ppm。
类似于前述实施例1-9的合成路线,选择相应的反应物和本领域技术人员已知的合成方式,合成以下实施例化合物。具体地,以实施例9步骤一的化合物即―(R)-7-(2-((3-((叔丁基苯基硅基)氧基)-2,2-二氟丙基)胺基)丙基)-3-三苯基苯并[d]噁唑-2(3H)-酮‖作为原料,合成以下实施例13-15;以实施例5步骤三的化合物即―(R)-7-2-氨基丙基)(2,2,2-三氟乙基-3-三苯甲基苯并[d]噁唑-2(3H)-酮‖作为原料,合成以下实施例10-12、16、18、25;以实施例1步骤四的化合物即―(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]噁唑-2(3H)-酮‖作为原料,合成下列实施例20。
实施例10:(6S,8R)-6-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物10)
Figure PCTCN2022080680-appb-000043
1H NMR:(400MHz,CDCl3)δ=8.03-7.74(m,1H),6.79(s,1H),6.67-6.60(m,1H),5.99(d,J=11.0Hz,2H),5.21(s,1H),4.59-4.51(m,1H),4.48-4.40(m,1H),4.38-4.25(m,1H),4.12-3.95(m,1H),3.78-3.67(m,2H),3.63-3.51(m,1H),3.26-3.09(m,2H),3.00-2.86(m,3H),2.78(dd,J=4.4,16.4Hz,1H),2.67-2.56(m,2H),1.83-1.71(m,2H),1.09(d,J=6.6Hz,3H)ppm。
LCMS:m/z(M+H)+=529.4
实施例11:(6S,8R)-6-(2,6-二氟-4-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物11)
Figure PCTCN2022080680-appb-000044
1H NMR:(400MHz,ACETONITRILE-d3)δ=6.81(d,J=8.0Hz,1H),6.62(d,J=8.0Hz,1H),6.44(br d,J=11.2Hz,2H),5.28(s,1H),4.79(br s,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.56-3.46(m,1H),3.42-3.25(m,1H),3.06(br dd,J=4.8,16.6Hz,1H),3.00-2.86(m,1H),2.83-2.69(m,5H),2.53-2.45(m,4H),1.91-1.85(m,1H),1.85-1.76(m,2H),1.07(d,J=6.4Hz,3H)ppm。
实施例12:(6S,8R)-6-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物12)
Figure PCTCN2022080680-appb-000045
1H NMR:(400MHz,CD3Cl)δ=8.02-7.65(m,1H),6.79-6.71(m,1H),6.64-6.54(m,1H),6.27(d,J=10.4Hz,2H),5.29-5.22(m,1H),4.72(m,J=6.0Hz,1H),4.56(t,J=6.0Hz,1H),4.44(t,J=6.0Hz,1H),3.85-3.72(m,2H),3.64-3.50(m,1H),3.26-3.08(m,4H),2.97-2.86(m,1H),2.85-2.76(m,1H),2.65(t,J=7.2Hz,2H),1.83-1.70(m,2H),1.10(d,J=6.4Hz,3H)。
LCMS:m/z(M+H)+=530.2
实施例13:(6S,8R)-7-(2,2-二氟-3-羟基丙基)-6-(2,6-二氟-4-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)苯基)-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物13)
Figure PCTCN2022080680-appb-000046
1H NMR:(400MHz,CHLOROFORM-d)δ=8.36(br d,J=1.1Hz,1H),6.79(d,J=8.1Hz,1H),6.58(d,J=8.1Hz,1H),6.40(d,J=10.4Hz,2H),5.18(s,1H),4.89(br s,1H),4.61(t,J=5.6Hz,1H),4.50(t,J=5.6Hz,1H),3.77-3.61(m,3H),3.29-2.92(m,7H),2.91-2.75(m,3H),2.54-2.38(m,1H),2.24-2.01(m,3H),1.10(d,J=6.6Hz,3H)ppm。
实施例14:(6S,8R)-7-(2,2-二氟-3-羟基丙基)-6-(2,6-二氟-4-(6-(2-氟乙基)-2,6-二氮螺[3.3]庚烷-2-基)苯基)-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物14)
Figure PCTCN2022080680-appb-000047
1H NMR:(400MHz,ACETONITRILE-d3)δ=6.78(br d,J=6.6Hz,1H),6.67-6.44(m,1H),6.07-5.84(m,2H),5.94(br d,J=11.4Hz,1H),5.24-5.04(m,1H),5.13(br s,1H),4.50-4.26(m,2H),3.89(br s,4H),3.73-3.61(m,1H),3.58-3.45(m,2H),3.36(br s,4H),3.13-3.00(m,2H),3.18-2.97(m,1H),2.77-2.59(m,5H),2.80-2.57(m,1H),1.07-0.97(m,3H),1.03(br d,J=3.8Hz,1H)ppm。
实施例15:(6S,8R)-7-(2,2-二氟-3-羟基丙基)-6-(2,6-二氟-4-(3-((3-氟丙基)胺基)氮杂环丁烷-1-基)苯基)-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物15)
Figure PCTCN2022080680-appb-000048
1H NMR:(400MHz,ACETONITRILE-d3)δ=6.85(d,J=8.1Hz,1H),6.65(d,J=8.1Hz,1H),5.95(d,J=11.5Hz,2H),5.15(s,1H),4.56(t,J=5.6Hz,1H),4.44(t,J=5.6Hz,1H),4.05(t,J=7.5Hz,2H),3.91-3.81(m,3H),3.72-3.48(m,3H),3.43-3.38(m,2H),3.18-3.02(m,2H),2.82-2.64(m,2H),2.13-2.01(m,4H),1.03(d,J=6.5Hz,3H)。
实施例16:(E)-4-((2-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧基-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢噁唑并[5,4-f]异喹啉-6-基)苯氧基)乙基)胺基)-N,N-二甲基丁-2-烯酰胺(化合物16)
Figure PCTCN2022080680-appb-000049
1H NMR:(400MHz,ACETONITRILE-d3)δ=6.81(d,J=8.1Hz,1H),6.75-6.65(m,1H),6.61(d,J=8.1Hz,1H),6.56-6.46(m,3H),5.29(s,1H),4.02(t,J=5.3Hz,2H),3.57-3.50(m,2H),3.41-3.38(m,2H),3.37-3.30(m,1H),3.11-3.04(m,1H),3.02(s,3H),2.94-2.91(m,2H),2.90-2.88(m,3H),2.79-2.72(m,1H),1.11(t,J=7.0Hz,2H),1.07(d,J=6.6Hz,3H)ppm。
LCMS:m/z 569.4[M+H] +
实施例18:(6S,8R)-6-(2,6-二氟-4-(3-((3-氟丙基)胺基)氮杂环丁烷-1-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物18)
Figure PCTCN2022080680-appb-000050
1H NMR:(400MHz,ACETONITRILE-d3)δ=6.81(br d,J=8.0Hz,1H),6.61(br d,J=7.9Hz,1H),5.95(d,J=11.6Hz,2H),5.22(s,1H),4.56(t,J=5.9Hz,1H),4.44(br t,J=5.7Hz,1H),4.03(br t,J=7.3Hz,2H),3.75-3.63(m,1H),3.58-3.44(m,3H),3.36-3.24(m,1H),3.10-2.86(m,2H),2.80-2.71(m,1H),2.64(br t,J=6.8Hz,2H),1.86-1.71(m,3H),1.06(d,J=6.6Hz,3H)。
实施例20:(6S,8R)-6-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)苯基)-7-((1-氟环丙基)甲基)-8-甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物20)
Figure PCTCN2022080680-appb-000051
1H NMR:(400MHz,ACETONITRILE-d3)δ=8.12(s,1H),6.82(d,J=8.3Hz,1H),6.61(d,J=8.0Hz,1H),6.10(d,J=11.6Hz,2H),5.38(br d,J=7.3Hz,1H),5.12(s,1H),4.55(t,J=6.1Hz,1H),4.43(t,J=6.0Hz,1H),4.08-3.97(m,1H),3.75(br d,J=6.3Hz,3H),3.12-2.99(m,2H),2.97-2.90(m,2H),2.83-2.70(m,2H),2.62(t,J=7.1Hz,2H),1.78-1.66(m,2H),1.02(d,J=6.6Hz,3H),0.97-0.86(m,2H),0.62-0.40(m,2H)ppm。
实施例25:(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物25)
Figure PCTCN2022080680-appb-000052
步骤一:(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑 并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000053
将化合物(R)-7-(2-((2,2,2-三氟乙基)氨基)丙基)苯并[d]噁唑-2(3H)-酮(3.00g,10.9mmol),4-溴-2-甲氧基苯甲醛(3.29g,5.32mmol),三氟乙酸(6.24g,54.7mmol),溶于甲苯(15mL)中并在110℃氮气保护下反应48小时。LCMS检测反应结束。反应液浓缩后柱层析纯化得到白色固体(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(4.35g,9.23mmol,收率84.4%)。
LCMS:m/z 470.0[M+H] +
1H NMR(400MHz,DMSO-d6)δ=11.53(s,1H),7.23(d,J=1.75Hz,1H),6.94-7.11(m,1H),6.82(d,J=8.00Hz,1H),6.68(d,J=8.13Hz,1H),6.54(d,J=8.13Hz,1H),5.28-5.52(m,1H),3.82-3.91(m,3H),3.27-3.41(m,2H),2.77-3.02(m,2H),2.68(m,J=16.70,7.30Hz,1H),1.02(d,J=6.50Hz,3H)ppm。
步骤二:(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基 -6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000054
将(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(2.77g,5.88mmol)溶于N,N-二甲基甲酰胺(25mL)中,在0℃氮气保护下加入钠氢(353mg,8.82mmol),然后加入三苯基氯甲烷(1.64g,5.88mmol),反应液在25℃反应2小时。TLC检测反应结束。反应液加饱和氯化铵水溶液淬灭,乙酸乙酯萃取,有机相饱和食盐水洗,硫酸钠干燥浓缩。粗品柱层析纯化得到黄色固体(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(3.75g,5.26mmol,收率89.4%)。
LCMS:m/z 712.2[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=7.18-7.42(m,15H),6.92-6.96(m,1H),6.88(dd,J=8.13,1.75Hz,1H),6.70(d,J=8.25Hz,1H),6.14(d,J=8.63Hz,1H),5.55-5.66(m,1H),5.23(s,1H),3.76(s,3H),3.35-3.48(m,1H),2.89-3.04(m,2H),2.64-2.79(m,1H),2.61(br d,J=6.25Hz,1H),0.97(d,J=6.63Hz,3H)ppm。
步骤三:(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000055
将(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(2g,2.80mmol),1-(3-氟丙基)-3-氨基氮杂环丁烷(1.41g,3.92mmol),碳酸铯(2.74g,8.41mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯基)]合钯(II)甲磺酸盐(118mg,140umol)溶于甲苯(20mL),110℃氮气保护下反应12小时。LCMS监测反应结束。浓缩柱层析纯化得黄色固体(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(1g,1.31mmol,收率46.7%)。
LCMS:m/z 764.3[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=7.18(br d,J=2.38Hz,15H),6.58(d,J=8.25Hz,1H),6.13-6.25(m,1H),5.97-6.04(m,1H),5.92(dd,J=8.25,2.13Hz,1H),5.50-5.65(m,1H),5.14-5.18(m,1H),4.48(t,J=5.94Hz,1H),4.36(t,J=5.94Hz,1H),3.90(br d,J=6.75Hz,1H),3.61-3.79(m,6H),3.44(br d,J=5.50Hz,1H),3.00-3.29(m,1H),2.87-3.00(m,2H),2.79(br d,J=6.13Hz,2H),2.49-2.59(m,3H),1.64-1.73(m,2H),0.97(d,J=6.50Hz,3H)ppm。
步骤四:(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑[5,4-f]异喹啉-2(3H)-酮的合成(化合物25)
Figure PCTCN2022080680-appb-000056
将三氟乙酸(15.4g,135mmol)和(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(1g,1.31mmol)溶于水(1mL)中,反应液在20℃氮气保护下反应1小时。LCMS检测反应结束。混合物浓缩后用碳酸氢钠调pH=7,用水和乙酸乙酯萃取,有机相浓缩。粗产物柱层析纯化后送SFC手性拆分得黄色固体(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢噁唑[5,4-f]异喹啉-2(3H)-酮(300mg,571umol,收率43.7%)。
LCMS:m/z 522.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d3)δ=6.79(d,J=8.13Hz,1H),6.44-6.59(m,2H),6.19(d,J=2.00Hz,1H),5.97(dd,J=8.32,2.06Hz,1H),5.31(s,1H),4.75(br d,J=7.00Hz,1H),4.52(t,J=6.07Hz,1H),4.40(t,J=6.07Hz,1H),3.92-4.09(m,1H),3.79(s,3H),3.68(m,J=3.25,2.13Hz,2H),3.43-3.54(m,1H),3.17-3.30(m,1H),2.87-3.01(m,2H),2.76,(m,J=6.38Hz,2H),2.61-2.67(m,1H),2.51(t,J=7.00Hz,2H),1.63-1.76(m,2H),1.04(d,J=6.50Hz,3H)ppm。
实施例26:(6S,8R)-6-(5-((1-(3-氟丙烷)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物26)
Figure PCTCN2022080680-appb-000057
步骤一:(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]恶唑-7-基)丙烷-2-基)氨基甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000058
将7-溴-3-三苯甲基-苯并恶唑-2酮(100g,219mmol,1eq)溶于四氢呋喃(1000mL)在-78℃氮气保护下加入正丁基锂(2.5M,87.6mL,1eq)搅拌30分钟。将(R)-4-甲基-1,2,3-氧杂噻唑烷-3-甲酸叔丁酯-2,2-二氧化物(52.0g,219mmol,1eq)的四氢呋喃(100mL)溶液在-78℃加入体系,然后自然恢复到0℃反应2个小时。TLC检测反应完成,将反应液倒入饱和氯化铵水溶液中淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到黄色固体(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]恶唑-7-基)丙烷-2-基)氨基甲酸叔丁酯(360g,673mmol,收率74.9%)。
1H NMR(400MHz,CHLOROFORM-d)δ=7.52-7.47(m,6H),7.35-7.27(m,9H),6.92-6.85(m,1H),6.76(t,J=8.0Hz,1H),5.93(br d,J=8.0Hz,1H),4.00(s,1H),3.81- 3.72(m,1H),2.84(br d,J=6.0Hz,2H),1.40(s,9H),1.16(d,J=6.4Hz,3H)ppm。
步骤二:(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]恶唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000059
在0℃氮气保护下将(R)-(1-(2-氧代-3-三苯甲基-2,3-二氢苯并[d]恶唑-7-基)丙烷-2-基)氨基甲酸叔丁酯(240g,448mmol,1eq)溶于甲醇(400ml),然后加入到盐酸甲醇溶液(6M,800mL,10.6eq)。在0℃反应2个小时,TLC检测反应完成。反应液浓缩,加入饱和碳酸氢钠中和,乙酸乙酯萃取浓缩得到粗品,用石油醚:乙酸乙酯=1:1打浆过滤得到黄色固体(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]恶唑-2(3H)-酮(198g,455mmol,收率84.6%)。
1H NMR(400MHz,CHLOROFORM-d)δ=7.48-7.34(m,6H),7.23-7.14(m,9H),6.81-6.73(m,1H),6.66(t,J=8.0Hz,1H),5.88(m,1H),3.33(s,J=6.4Hz,1H),2.82-2.65(m,2H),1.12(d,J=6.4Hz,3H)ppm。
步骤三:(R)-7-(2-((2,2,2-三氟乙基)胺基)丙基)-3-三苯甲基苯并[d]恶唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000060
在20℃氮气保护下将(R)-7-(2-氨基丙基)-3-三苯甲基苯并[d]恶唑-2(3H)-酮(10.0g,23.0mmol,1eq),三氟甲磺酸(2,2,2-三氟乙基)酯(6.14g,26.5mmol,1.15eq),二异丙基乙二胺(8.92g,69.0mmol,12.0mL,3eq)溶于二氧六环(100mL)里,然后在80℃氮气保护下反应10小时。TLC检测反应完成,反应液浓缩,粗产品柱层析分离得到白色固体(R)-7-(2-((2,2,2-三氟乙基)胺基)丙基)-3-三苯甲基苯并[d]恶唑-2(3H)-酮(9.00g,17.4mmol,收率75.7%)。
1H NMR(400MHz,CHLOROFORM-d)δ=7.48(br d,J=7.2Hz,6H),7.33-7.27(m,9H),6.86(br d,J=7.6Hz,1H),6.79-6.73(m,1H),5.95(br d,J=8.0Hz,1H),3.50(s,1H),3.26(br d,J=9.3Hz,3H),2.98-2.85(m,1H),2.82-2.71(m,1H),1.15(br d,J=5.6Hz,3H)ppm。
步骤四:(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢异喹啉并[5,4-f]恶唑-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000061
将(R)-7-(2-((2,2,2-三氟乙基)胺基)丙基)-3-三苯甲基苯并[d]恶唑-2(3H)-酮(1.00g,1.94mmol,1eq)和5-溴-2-吡啶甲醛(540mg,2.90mmol,1.5eq)溶于甲苯(50mL)中加入三氟乙酸(662mg,5.81mmol,430uL,3eq),混合物在90℃反应20小时。TLC检测反应完成,反应液浓缩,粗产品by-TLC(石油醚:乙酸乙酯=10:1)纯化得黄色固体(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢异喹啉并[5,4-f]恶唑-2(3H)-酮(780mg, 1.76mmol,收率91.1%)。
1H NMR(400MHz,CHLOROFORM-d)δ=8.89(s,1H),8.54(d,J=2.4Hz,1H),7.84-7.80(m,1H),7.48(d,J=8.4Hz,1H),6.79-6.74(m,2H),5.09(s,1H),4.13(q,J=7.2Hz,1H),3.50-3.39(m,1H),3.35-3.22(m,1H),3.09-3.03(m,1H),3.00-2.89(m,1H),2.70-2.64(m,1H),1.13(d,J=6.4Hz,3H)ppm。
步骤五:(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000062
将(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢异喹啉并[5,4-f]恶唑-2(3H)-酮(1.00g,2.26mmol,1eq)溶于N,N-二甲基甲酰胺(20mL)中,在0℃加入氢化钠(181mg,4.52mmol,60%purity,2eq),然后在20℃加入三苯甲基氯甲烷(693mg,2.49mmol,1.1eq)并反应2小时。TLC检测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩,粗品柱层析分离得到白色固体(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(1.20g,1.56mmol,收率69.0%)。
1H NMR(400MHz,CHLOROFORM-d)δ=8.52(s,1H),7.75(dd,J 1=2.4Hz,J 2=8.4Hz,1H),7.49-7.45(m,6H),7.36(d,J=8.4Hz,1H),7.32-7.27(m,9H),6.39(d,J=8.4Hz,1H),5.75(d,J=8.4Hz,1H),4.94(s,1H),3.53-3.42(m,1H),3.29-3.18(m,1H),3.01(dd,J 1=4.8Hz,J 2=16.8Hz,1H),2.90(br dd,J 1=9.2Hz,J 2=15.6Hz,1H),2.68(dd,J 1=6.8Hz,J 2=16.8Hz,1H),1.10(d,J=6.4Hz,3H)ppm。
步骤六:3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6基)吡啶-3-基)氨基)氮杂环丁烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000063
(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(200mg,292umol,1eq)和3-氨基氮杂环丁烷-1-甲酸叔丁酯(126mg,730umol,2.5eq)溶于8ml甲苯中,加入[(2-2-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯)]钯(II)甲烷磺酸盐(24.5mg,29.2umol,0.1eq)和碳酸铯(286mg,876umol,3eq),反应液在110℃反应15小时。TLC检测反应完成,反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到黄色固体3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6基)吡啶-3-基)氨基)氮杂环丁烷-1-甲酸叔丁酯(120mg,137umol,收率47.2%)。
1H NMR(400MHz,CHLOROFORM-d)δ=7.77(d,J=2.4Hz,1H),7.52–7.42(m,7H),7.32–7.28(m,4H),7.26–7.17(m,9H),6.89–6.69(m,1H),6.37(d,J=8.4Hz,1H),5.80–5.64(m,1H),5.39–5.20(m,1H),4.98–4.77(m,1H),4.33–4.27(m,2H),4.23–4.11(m,1H),4.08–3.98(m,1H),3.74(m,2H),3.53–3.43(m,2H),3.26–3.11(m,1H),3.04– 2.85(m,2H),2.72–2.59(m,1H),1.45(s,9H),1.09(d,J=6.4Hz,3H)ppm。
步骤七:(6S,8R)-6-(5-(氮杂环丁烷-3-基氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000064
在0℃下将三氟乙酸(9.00mL)和水(1.00mL)加入3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6基)吡啶-3-基)氨基)氮杂环丁烷-1-甲酸叔丁酯(120mg,154umol,1eq)中,混合物在20℃反应2小时,TLC监测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩,得到黄色固体(6S,8R)-6-(5-(氮杂环丁烷-3-基氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(120mg,粗产品)。
1H NMR(400MHz,CHLOROFORM-d)δ=8.21-7.93(m,2H),7.60-7.35(m,2H),6.95-6.62(m,1H),5.55-5.17(m,1H),4.39-4.13(m,1H),4.06-3.90(m,1H),3.63-3.42(m,1H),2.33-2.13(m,2H),2.07-1.96(m,2H),1.14-1.08(m,2H),0.88(br d,J=4.8Hz,3H)ppm。
步骤八:(6S,8R)-6-(5-((1-(3-氟丙烷)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000065
在20℃氮气保护下将(6S,8R)-6-(5-(氮杂环丁烷-3-基氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(66.0mg,152umol,1eq)和3-氟-1-碘丙烷(28.6mg,152umol,1eq)溶于N,N-二甲基甲酰胺(1mL)中加入二异丙基乙二胺(98.4mg,761umol,132uL,5eq),混合物在20℃反应4小时。LCMS监测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗产品通过反相柱分离(column:3_Phenomenex Luna C18 75*30mm*3um;mobilephase:[water(0.05%NH 3H 2O+10mM NH 4HCO 3)-ACN];B%:45%-75%,30min),然后再经过手型柱分离(column:REGIS(R,R)WHELK-O1(250mm*25mm,10um);mobile phase:0.1%NH 3H 2O ETOH;B%:30%-30%,0min)得到黄色固体(6S,8R)-6-(5-((1-(3-氟丙烷)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(27.0mg,54.2umol,收率35.6%,纯度97.7%)。
LCMS:m/z 494.3[M+H] +
1H NMR(400MHz,CD 3OD)δ=7.76-7.71(m,1H),7.13(d,J=8.4Hz,1H),6.93(m,1H),6.80(d,J=8.0Hz,1H),6.56(d,J=8.0Hz,1H),4.52(t,J=6.0Hz,1H),4.40(t,J=6.0Hz,1H),4.13-4.06(m,1H),3.82-3.76(m,2H),3.54-3.47(m,1H),3.15-3.08(m,1H),3.00-2.91(m,3H),2.76(m,1H),2.64(t,J=7.6Hz,2H),1.83-1.69(m,2H),1.06(d,J=6.6Hz,3H)ppm。
实施例27:(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟甲基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物27)
Figure PCTCN2022080680-appb-000066
步骤一:(6S,8R)-6-(5-溴-3-氟吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000067
将化合物(R)-7-(2-(2,2,2-三氟乙基)氨基)丙基)苯并[d]恶唑-2(3H)-酮(500mg,1.82mmol),5-溴-3-氟-吡啶-2-甲醛(390mg,1.91mmol),三氟乙酸(1.04g,9.12mmol)溶于甲苯(10mL)中并在90℃氮气保护下反应12小时。LCMS检测反应结束。反应液浓缩后调pH=7-8,用水和乙酸乙酯萃取,有机相用饱和食盐水洗,干燥浓缩后柱层析纯化得到黄色固体(6S,8R)-6-(5-溴-3-氟吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(605mg,1.22mmol,收率66.8%)。
LCMS:m/z 461.8[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=8.20-8.50(m,2H)7.61(dd,J=9.01,1.88Hz,1H)6.80(d,J=8.00Hz,1H)6.65(d,J=8.00Hz,1H)5.37(s,1H)3.56-3.68(m,1H)3.23-3.31(m,1H)2.88-3.13(m,2H)2.67(dd,J=17.13,8.25Hz,1H)1.07-1.18(m,3H)ppm。
步骤二:(6S,8R)-6-(5-溴-3-氟吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000068
将(6S,8R)-6-(5-溴-3-氟吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(600mg,1.21mmol)溶于N,N-二甲基甲酰胺(12mL)中,在0℃氮气保护下加入钠氢(58mg,1.45mmol)并搅拌30分钟,然后加入三苯基氯甲烷(336mg,1.21mmol),反应液在20℃反应30分钟。LCMS检测反应结束。加饱和氯化铵水溶液淬灭,乙酸乙酯萃取,有机相饱和食盐水洗,干燥浓缩。粗品柱层析纯化得到白色固体(6S,8R)-6-(5-溴-3-氟吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(754mg,991umol,收率82.1%)。
LCMS:m/z 704[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=8.27(d,J=1.63Hz,1H),7.49(dd,J=9.01,1.88Hz,1H),7.37-7.43(m,6H),7.18-7.22(m,9H),6.21(d,J=8.50Hz,1H),5.67(d,J=8.38Hz,1H),5.19(s,1H),3.48-3.69(m,1H),3.06-3.19(m,1H),2.80-2.97(m,2H), 2.54(dd,J=17.13,8.25Hz,1H),1.02(d,J=6.75Hz,3H)ppm。
步骤三:(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000069
将(6S,8R)-6-(5-溴-3-氟吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(400mg,526umol),1-(3-氟丙基)氨基氮杂环丁烷(379mg,1.05mmol),碳酸铯(685mg,2.10mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2—氨基联苯基)]合钯(II)甲磺酸盐(44.1mg,52.6umol)溶于甲苯(10mL),110℃氮气保护下反应12小时。LCMS监测反应结束。浓缩柱层析纯化得淡黄色固体(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(163mg,188umol,收率35.8%)。
LCMS:m/z 754.1[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=7.57(d,J=1.88Hz,1H),7.36-7.41(m,6H),7.21(s,9H),7.16(s,1H),6.41(dd,J=11.57,2.31Hz,1H),6.23(d,J=8.50Hz,1H),5.64(d,J=8.50Hz,1H),5.12(s,1H),4.31-4.53(m,2H),4.09-4.19(m,1H),3.93-4.00(m,1H),3.62-3.68(m,2H),3.00-3.14(m,1H),2.78-2.98(m,4H),2.46-2.60(m,3H),1.65(s,2H),1.02(d,J=6.63Hz,3H)ppm。
步骤四:(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟甲基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000070
把三氟乙酸(2.02g,17.6mmol)溶于水中(0.05mL)后加入(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(120mg,138umol)里,20℃氮气保护下反应5小时。LCMS检测反应结束。混合物浓缩后调pH=7-8,用水和乙酸乙酯萃取,有机相饱和食盐水洗,硫酸钠干燥,浓缩。粗产物prep-HPLC谱得黄色固体(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟甲基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(70mg,124.27umol)。
LCMS:m/z 511.2[M+H] +
步骤五:(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟甲基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000071
将(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟甲基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮手性拆分得(6S,8R)-6-(3-氟-5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟甲基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(42.7mg,83.5umol,收率61.0%)。
1H NMR(400MHz,ACETONITRILE-d3)δ=7.56-7.59(m,1H),6.82(d,J=8.00Hz,1H),6.61-6.67(m,2H),5.28(s,1H),5.14-5.19(m,1H),4.36-4.54(m,2H),3.93-4.02(m,1H),3.57-3.71(m,3H),3.27-3.40(m,1H),2.83-3.01(m,2H),2.76-2.82(m,2H),2.63(d,J=9.01Hz,1H),2.50(t,J=7.00Hz,2H),1.61-1.76(m,2H),1.06(d,J=6.75Hz,3H)ppm。
实施例28:(6S,8R)-6-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物28)
Figure PCTCN2022080680-appb-000072
步骤一:(6S,8R)-6-(5-溴-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000073
将(R)-7-(2-((2,2,2-三氟乙基)胺基)丙基)苯并[d]恶唑-2(3H)-酮(500mg,1.82mmol)和5-溴-3-甲氧基吡啶甲醛(400mg,1.85mmol)溶于甲苯(10mL)中,然后加入三氟乙酸(1.04g,9.12mmol)。在90℃反应12小时。LCMS检测反应结束,反应液浓缩后硅胶柱纯化得到(6S,8R)-6-(5-溴-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(750mg,1.59mmol,收率87.1%)。
步骤二:(6S,8R)-6-(5-溴-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000074
将(6S,8R)-6-(5-溴-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(700mg,1.48mmol)溶于N,N-二甲基甲酰胺(14mL)。在0℃加入钠氢(71.1mg,1.78mmol,60%纯度)。搅拌30分钟后加入三苯甲基氯甲烷(371mg,1.33mmol)并升温至20℃反应2小时。LCMS检测反应结束。反应液用冰水萃灭并用乙酸乙酯萃取。有机相用饱和食盐水洗三次,浓缩后用硅胶柱纯化得到(6S,8R)-6-(5-溴-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(750mg,1.05mmol,收率70.8%)。
1H NMR(400MHz,CHLOROFORM-d)δ=8.12(d,J=1.9Hz,1H),7.54-7.40(m,6H),7.32-7.26(m,9H),6.28(d,J=8.5Hz,1H),5.73(d,J=8.4Hz,1H),5.53-5.31(m,1H),3.82(s,3H),3.77-3.69(m,1H),3.25-3.09(m,1H),3.02-2.83(m,2H),2.64-2.50(m,1H),1.10(d,J=6.6Hz,3H)ppm。
步骤三:(6S,8R)-6-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000075
将(6S,8R)-6-(5-溴-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(400mg,560umol),1-(3-氟丙基)氮杂环丁烷-3-胺(403mg,1.12mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2—氨基联苯基)]合钯(II)甲磺酸盐(46.9mg,56.0umol)和碳酸铯(730mg,2.24mmol,)溶于甲苯(2mL)中。氮气保护,110℃反应12小时。LCMS检测反应结束。反应液浓缩后用硅胶柱纯化得到(6S,8R)-6-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(160mg,209umol,收率33.2%)。
步骤四:(6S,8R)-6-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000076
将(6S,8R)-6-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(160mg,208umol)溶于水(0.5mL)和三氟乙酸(4.5mL)中。20℃反应2小时,LCMS检测反应结束。用碳酸氢钠调pH=7-8,水和乙酸乙酯萃取,有机相用盐水洗三次,浓缩后用硅胶柱纯化得到(6S,8R)-6-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-3-甲氧基吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(70mg,134umol,收率64.0%)。
1H NMR(400MHz,ACETONITRILE-d 3)δ=7.32(d,J=2.3Hz,1H),6.80(d,J=8.0Hz,1H),6.63-6.51(m,2H),5.46(s,1H),4.89(d,J=7.0Hz,1H),4.54(t,J=6.1Hz,1H),4.42(t,J=6.1Hz,1H),4.20-3.96(m,1H),3.82(s,4H),3.74-3.65(m,2H),3.40-3.26(m,1H),3.02-2.84(m,2H),2.82-2.74(m,2H),2.64-2.56(m,1H),2.52(t,J=6.9Hz,2H), 1.77-1.65(m,2H),1.09(d,J=6.8Hz,3H)ppm。
实施例29:(6S,8R)-6-(5-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物29)
Figure PCTCN2022080680-appb-000077
步骤一:(S)-3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000078
(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(150mg,219umol,1eq)和(S)-3-羟基吡咯烷-1-甲酸叔丁酯(102mg,547umol,2.5eq)溶于甲苯(20mL)后加入(18.3mg,21.9umol,0.1eq)和碳酸铯(214mg,657umol,3eq),反应液在115℃反应15小时。TLC监测反应完成,反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗品柱层析分离得到黄色固体(S)-3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)吡咯烷-1-甲酸叔丁酯(120mg,138umol,31.5%收率)。
1H NMR(400MHz,CHLOROFORM-d)δ=8.02(d,J=2.8Hz,1H),7.39(d,J=7.2Hz,6H),7.24-7.20(m,5H),7.18-7.14(m,4H),7.08-7.04(m,1H),6.37-6.25(m,1H),5.66(d,J=8.4Hz,1H),4.86-4.80(m,2H),4.39(br s,1H),3.55-3.34(m,2H),3.34-3.23(m,1H),3.16-3.04(m,1H),3.01-2.89(m,1H),2.83(br dd,J 1=8.8Hz,J 2=15.6Hz,1H),2.59(dd,J 1=6.4Hz,J 2=16.8Hz,1H),2.15-2.01(m,2H),1.95-1.81(m,2H),1.40(s,9H),1.02(d,J=6.4Hz,3H)ppm。
步骤二:(6S,8R)-8-甲基-6-(5-((S)-吡咯烷-3-基)氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000079
在0℃下将三氟乙酸(1.54g,13.4mmol,997uL,87.3eq)加入溶于二氯甲烷(2mL)的(S)-3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)吡咯烷-1-甲酸叔丁酯(120mg,154umol,1eq)中,在20℃下反应2小时,TLC和LCMS监测反应完成。反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗产品薄层层析硅胶板分离得到黄色固体(6S,8R)-8-甲基-6-(5-((S)-吡咯烷-3-基)氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(105mg,粗产品)。
LCMS:m/z(M+H) +=449.2
1H NMR(400MHz,DMSO-d 6)δ=8.13(d,J=2.8Hz,1H),7.45-7.34(m,1H),7.32-7.26(m,1H),6.87-6.82(m,1H),6.74-6.67(m,1H),5.14-5.02(m,2H),3.00-2.92(m,2H),2.90-2.78(m,2H),2.72-2.71(m,1H),2.74-2.58(m,1H),2.21-2.07(m,2H),2.06-1.93(m,1H),1.23(s,1H),1.06(d,J=6.4Hz,3H)ppm。
步骤三:(6S,8R)-6-(5-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000080
在20℃氮气保护下将(6S,8R)-8-甲基-6-(5-((S)-吡咯烷-3-基)氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(105mg,187umol,1eq)和3-氟-1-碘丙烷(35.2mg,187umol,1eq)溶于N,N-二甲基甲酰胺(2mL)中加入二异丙基乙二胺(121mg,936umol,163uL,5eq),混合物在20℃反应4小时。LCMS检测反应完成,反应液加水淬灭,乙酸乙酯萃取,干燥浓缩。粗产品通过反相柱分离(column:Phenomenex Gemini-NX C18 75*30mm*3um;mobile phase:[water(0.04%NH 3H 2O+10mM NH 4HCO 3)-ACN];B%:18%-58%,14min)and SFC(column:REGIS(R,R)WHELK-O1(250mm*25mm,10um);mobile phase:0.1%NH 3H 2O ETOH;B%:30%-30%,10min)得到白色固体(6S,8R)-6-(5-(((S)-1-(3-氟丙烷)吡咯烷-3-基)氧基)吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(10.5mg,19.8umol,收率10.6%,纯度96%)。
LCMS:m/z(M+H) +=509.4
1H NMR(400MHz,CD 3OD)δ=8.05(d,J=2.8Hz,1H),7.40-7.30(m,2H),6.83(d,J=8.0Hz,1H),6.63(d,J=8.0Hz,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.53-3.48(m,1H),3.40(br d,J=6.0Hz,1H),3.10(dd,J 1=4.8Hz,J 2=16.8Hz,1H),3.02-2.87(m,4H),2.78(dd,J 1=6.2Hz,J 2=16.8Hz,1H),2.68-2.58(m,2H),2.56-2.49(m,1H),2.38(dt,J 1=7.6Hz,J 2=13.6Hz,1H),1.99-1.84(m,3H),1.29(br s,1H),1.12(d,J=6.4Hz,3H)ppm。
实施例30:(6S,8R)-6-(4-(((S)-1-(3-氟丙基)吡咯烷-3-基)氧基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物30)
Figure PCTCN2022080680-appb-000081
步骤一:(S)-3-(3-甲氧基-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000082
将(6S,8R)-6-(4-溴-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(500mg,700umol),(S)-3-羟基吡咯烷-1-甲酸叔丁酯(236mg,1.26mmol),碳酸铯(685mg,2.10mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯基)]合钯(II)甲磺酸盐(23.5mg,28.0umol)溶于甲苯(8mL),110℃氮气保护下反应16小时。平行投两个反应,LCMS监测反应结束。合并处理浓缩柱层析纯化得黄色固体(S)-3-(3-甲氧基-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-甲酸叔丁酯(180mg,收率15.7%)。
1H NMR(400MHz,CHLOROFORM-d)δ=7.51-7.43(m,6H),7.33-7.28(m,4H),7.27-7.19(m,5H),6.90-6.73(m,1H),6.43(br s,1H),6.36-6.19(m,2H),5.68(br d,J=8.5Hz,1H),5.29(s,1H),4.85(br s,1H),3.85-3.75(m,3H),3.71-3.43(m,5H),3.14-2.96(m,2H),2.85(qd,J=9.5,15.4Hz,1H),2.66(br dd,J=6.1,16.9Hz,1H),2.16(br s,2H),1.48(s,9H),1.07(br d,J=6.5Hz,3H)ppm。
步骤二:(6S,8R)-6-(2-甲氧基-4-((S)-吡咯烷-3-基氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000083
把(S)-3-(3-甲氧基-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-甲酸叔丁酯(180mg,220umol)溶于三氟乙酸(2ml)和水(0.2mL)中,20℃下反应2小时。LCMS检测反应结束。混合物浓缩后调pH=7-8,用水和乙酸乙酯萃取,有机相饱和食盐水洗,硫酸钠干燥,浓缩。柱层析分离得到黄色固体(6S,8R)-6-(2-甲氧基-4-((S)-吡咯烷-3-基氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(110mg,收率77.7%)。
1H NMR(400MHz,CHLOROFORM-d)δ=6.73(dd,J=8.3,12.6Hz,2H),6.54(d,J=8.1Hz,1H),6.45(d,J=2.4Hz,1H),6.24(dd,J=2.3,8.6Hz,1H),5.35(s,1H),3.84(s,3H),3.50-3.35(m,5H),3.16-3.01(m,3H),2.90-2.78(m,1H),2.66(br dd,J=6.5,16.9Hz,1H),2.37-2.27(m,1H),2.26-2.15(m,1H),1.06(d,J=6.6Hz,3H)ppm。
步骤三:(6S,8R)-6-(4-(((S)-1-(3-氟丙基)吡咯烷-3-基)氧基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000084
将(6S,8R)-6-(2-甲氧基-4-((S)-吡咯烷-3-基氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(110mg,170umol)和二异丙基乙胺(66.1mg,511umol)溶于N,N-二甲基甲酰胺(3mL),在20℃下加入3-氟-1-碘丙烷(25.6mg,136umol)反应3小时,TLC监测反应完成,通过HPLC分离得到60mg粗产物,然后经过SFC手性分离得到黄色固体(6S,8R)-6-(4-(((S)-1-(3-氟丙基)吡咯烷-3-基)氧基)-2-甲氧基苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(47.3mg,收率51.4%)。
1H NMR(400MHz,CHLOROFORM-d)δ=8.62-8.02(m,1H),6.81-6.68(m,2H),6.59(d,J=8.1Hz,1H),6.45(d,J=2.4Hz,1H),6.25(dd,J=2.4,8.5Hz,1H),5.38(s,1H),4.81(br s,1H),4.61-4.43(m,2H),3.84(s,3H),3.61-3.51(m,1H),3.16-3.06(m,2H),2.97-2.79(m,4H),2.76-2.62(m,3H),2.31(br dd,J=6.8,13.8Hz,1H),2.05-1.88(m,3H),1.30-1.23(m,1H),1.10(d,J=6.6Hz,3H)ppm。
实施例31:(6S,8R)-7-((1-氟环丙基)甲基)-6-(4-((1-(3-氟丙基)氮杂环丁环-3-基))氨基)-2-甲氧基苯基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮
Figure PCTCN2022080680-appb-000085
步骤一:(6S,8R)-6-(4-溴-2-甲氧基苯基)-7-((1-氟环丙基)甲基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000086
把(R)-7-(2-(((1-氟环丙基)甲基)氨基)丙基)苯并[d]恶唑-2(3H)-酮(1.05g,3.97mmol),4-溴-2-甲氧基苯甲醛(1.54g,7.15mmol)和三氟乙酸(2.26g,19.8mmol)溶于甲苯(8mL)后,在110℃反应120小时。LCMS监测反应结束。反应液浓缩柱层析纯化得黄色固体(6S,8R)-6-(4-溴-2-甲氧基苯基)-7-((1-氟环丙基)甲基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(249mg,539umol,收率13.5%)。
LCMS:m/z 462.8[M+H] +
步骤二:(6S,8R)-7-((1-氟环丙基)甲基)-6-(4-((1-(3-氟丙基)氮杂环丁环-3-基))氨基)-2-甲氧基苯基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000087
把((6S,8R)-6-(4-溴-2-甲氧基苯基)-7-((1-氟环丙基)甲基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(140mg,303umol),1-(3-氟丙基)氨基氮杂环丁烷(112mg,849umol),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(27.5mg,30.3umol)和叔丁醇钠(87.4mg,910umol)溶于二氧六环(1mL)后并在60℃反应2小时。LCMS监测反应结束。反应液过滤后通过prep-HPLC纯化,然后产品用手性SFC纯化,得到黄色固体(6S,8R)-7-((1-氟环丙基)甲基)-6-(4-((1-(3-氟丙基)氮杂环丁环-3-基))氨基)-2-甲氧基苯基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(7.2mg,收率4.49%)。
LCMS:m/z 513.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=6.75(d,J=8.13Hz,1H),6.67(d,J=8.38Hz,1H),6.54(d,J=8.13Hz,1H),6.18(d,J=2.00Hz,1H),6.00(dd,J=8.38,2.00Hz,1H),5.20(s,1H),4.69(m,J=6.90Hz,1H),4.52(t,J=6.07Hz,1H),4.40(t,J=6.07Hz,1H),3.95-4.04(m,1H),3.80(s,3H),3.64-3.72(m,3H),3.03(dd,J=16.26,4.88Hz,1H),2.86-2.97(m,1H),2.68-2.78(m,3H),2.56-2.65(m,1H),2.50(t,J=7.00Hz,2H),1.64-1.74(m,2H),0.99(d,J=6.63Hz,3H),0.86-0.94(m,2H),0.46-0.58(m,2H)ppm。
类似于前述实施例25的合成路线,选择相应的反应物和本领域技术人员已知的合成方式,合成以下实施例32,33及34。
实施例32:(6S,8R)-7-(2,2-二氟丙基)-6-(4-((1-(3-氟丙基)氮杂环丁环-3-基)-2-甲氧基苯基)-8-甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮
Figure PCTCN2022080680-appb-000088
LCMS:m/z 519.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=6.75(d,J=8.1Hz,1H),6.48(dd,J=8.2,19.9Hz,2H),6.18(d,J=2.1Hz,1H),5.97(dd,J=2.1,8.4Hz,1H),5.22(s,1H),4.73(br s,1H),4.55-4.37(m,2H),4.02-3.95(m,1H),3.79(s,3H),3.69-3.64(m,2H),3.54-3.38(m,2H),2.96-2.85(m,2H),2.74(q,J=6.4Hz,2H),2.65-2.55(m,2H),2.49(t,J=7.0Hz,2H),2.21-2.11(m,1H),1.75-1.62(m,2H),1.54(t,J=19.2Hz,3H),1.00(d,J=6.6Hz,3H)ppm。
实施例33:(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-2-甲氧基-6-氯-苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物33)
Figure PCTCN2022080680-appb-000089
LCMS:m/z 557.4[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=6.73(br d,J=8.1Hz,1H),6.55-6.30(m,1H),6.25-5.95(m,2H),5.60-5.33(m,1H),5.04(br d,J=6.1Hz,1H),4.59-4.34(m,2H),4.11-3.97(m,1H),3.92-3.63(m,4H),3.59(br s,1H),3.36-3.12(m,3H),2.95-2.72(m,4H),2.58(t,J=7.1Hz,2H),1.78-1.67(m,2H),1.01(d,J=6.5Hz,3H)ppm。
实施例34:(6S,8R)-6-(4-((1-(3-氟丙基)氮杂环丁烷-3-基)胺基)-2-甲氧基-6-氟-苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-6,7,8,9-四氢噁唑并[5,4-f]异喹啉-2(3H)-酮(化合物34)
Figure PCTCN2022080680-appb-000090
LCMS:m/z 541.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ=11.46(br s,1H),6.75(d,J=8.0Hz,1H),6.51-6.36(m,2H),5.99(br s,1H),5.75(br d,J=12.8Hz,1H),5.22(br s,1H),4.51(t,J=5.9Hz,1H),4.40(t,J=6.0Hz,1H),4.03-3.89(m,1H),3.70(br s,5H),3.50-3.39(m,1H),3.39-3.33(m,1H),3.31-3.25(m,1H),3.08-2.62(m,6H),1.74-1.59(m,2H),1.01(br d,J=6.5Hz,3H)ppm。
实施例35:(E)-4-(3-((3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)胺基)氮杂环丁烷-1-基)-N,N-二甲基丁-2-烯酰胺(化合物35)
Figure PCTCN2022080680-appb-000091
步骤一:3-((3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)氨基)氮杂环丁烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000092
将(6S,8R)-6-(4-溴-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(1.0g,1.39mmol),3-氨基氮杂环丁烷-1-甲酸叔丁酯(359mg,2.08mmol),碳酸铯(996mg,3.06mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2-氨基联苯基)]合钯(II)甲磺酸盐(58.3mg,69.5umol)溶于甲苯(20mL),110℃氮气保护下反应12小时。LCMS监测反应结束。混合物浓缩后用水和乙酸乙酯萃取,有机相饱和食盐水洗,硫酸钠干燥,浓缩。粗产物浓缩柱层析纯化得黄色固体3-((3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)氨基)氮杂环丁烷-1-甲酸叔丁酯(880mg,1.09mmol,收率78.1%)。
LCMS:m/z 810.3[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=7.36-7.42(m,7H),7.17-7.22(m,10H),6.15-6.24(m,1H),5.87(d,J=10.76Hz,2H),5.58(d,J=8.63Hz,1H),5.05(s,1H),4.21(t,J=8.00Hz,2H),3.65(ddd,J=8.88,4.63,2.13Hz,2H),3.43-3.52(m,1H),3.06(m,J=16.70,5.70Hz,2H),2.62-2.85(m,2H),1.37(s,9H),0.98(d,J=6.50Hz,3H)ppm。
步骤二:(6S,8R)-6-(4-(氮杂环丁烷-3-基氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000093
把三氟乙酸(15.5g,135mmol)溶于水中(1mL)后加入3-((3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)氨基)氮杂环丁烷-1-甲酸叔丁酯(400mg,493umol)里,20℃氮气保护下反应2小时。LCMS检测反应结束。混合物浓缩后得粗产物黄色固体(6S,8R)-6-(4-(氮杂环丁烷-3-基氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(400mg,粗品)。
LCMS:m/z 468.2[M+H] +
步骤三:(E)-4-(3-((3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)胺基)氮杂环丁烷-1-基)-N,N-二甲基丁-2-烯酰胺合成
Figure PCTCN2022080680-appb-000094
将(6S,8R)-6-(4-(氮杂环丁烷-3-基氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(100mg,213umol),(E)-4-溴-N,N-二甲基丁 -2-烯酰胺(41mg,213umol),二异丙基乙胺(60.7mg,469umol)溶于N,N-二甲基甲酰胺(3mL),20℃氮气保护下反应2小时。LCMS检测反应结束,反应液过滤送prep-HPLC纯化得到的产物送SFC分离纯化得黄色固体(E)-4-(3-((3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)胺基)氮杂环丁烷-1-基)-N,N-二甲基丁-2-烯酰胺(38.8mg,65.3umol,收率30.6%)。
LCMS:m/z 579.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=6.81(d,J=8.00Hz,1H),6.50-6.66(m,2H),6.40-6.48(m,1H),6.08(d,J=11.88Hz,2H),5.27(m,J=7.00Hz,1H),5.20(s,1H),3.92-4.04(m,1H),3.69(t,J=6.82Hz,2H),3.43-3.58(m,1H),3.22-3.38(m,1H),3.19(dd,J=5.19,1.31Hz,2H),3.00-3.09(m,4H),2.91-2.99(m,1H),2.88-2.91(m,3H),2.80-2.87(m,2H),2.73(dd,J=16.45,4.94Hz,1H),1.06(d,J=6.50Hz,3H)ppm。
实施例36:(6S,8R)-6-(4-((1-丙烯酰氮杂环丁烷-3-基)氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物36)
Figure PCTCN2022080680-appb-000095
步骤一:(6S,8R)-6-(4-((1-丙烯酰氮杂环丁烷-3-基)氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000096
将(6S,8R)-6-(4-(氮杂环丁烷-3-基氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(250mg,533umol)和二异丙基乙胺(827mg,6.40mmol)溶于N,N-二甲基甲酰胺(2mL),然后在0℃下加丙烯酰氯(9.66mg,106umol)后,在0℃反应2小时。LCMS检测反应结束。反应液过滤送prep-HPLC纯化得到的产物送SFC分离纯化得黄色固体(6S,8R)-6-(4-((1-丙烯酰氮杂环丁烷-3-基)氨基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(29.4mg,56.1umol,收率10.5%)。
LCMS:m/z 522.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=8.96(s,1H),6.82(d,J=8.13Hz,1H),
6.63(d,J=8.13Hz,1H),6.21-6.36(m,1H),6.12-6.19(m,1H),6.09(d,J=11.63Hz,2H),5.63(dd,J=10.19,1.94Hz,1H),5.44(m,J=6.38Hz,1H),5.22(s,1H),4.49-4.59(m,1H),4.15-4.34(m,2H),3.94(dt,J=9.10,4.64Hz,1H),3.68-3.78(m,1H),3.45-3.57(m,1H),3.23-3.38(m,1H),2.87-3.09,(m,2H),2.74(dd,J=16.32,5.19Hz,1H),1.06(d,J=6.63Hz,3H)ppm。
实施例37:(E)-4-((R)-3-((3,5-二氟4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-基)-N,N-二甲基丁-2-烯酰 胺(化合物37)
Figure PCTCN2022080680-appb-000097
步骤一:(R)-3-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000098
将(6S,8R)-6-(4-溴-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(1.7g,2.36mmol),R-3-羟基-1-Boc-吡咯烷(663mg,3.54mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2—氨基联苯基)]合钯(II)甲磺酸盐(99.0mg,118umol),碳酸铯(1.69g,5.20mmol)溶于甲苯(20mL),110℃氮气保护下反应12小时。LCMS监测反应结束。混合物浓缩后柱层析纯化得淡黄色固体(R)-3-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-甲酸叔丁酯(503mg,609umol,收率25.7%)。
LCMS:m/z 825.3[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=7.19-7.42(m,15H),6.26(d,J=10.38Hz,2H),6.20(d,J=8.63Hz,1H),5.60(d,J=8.50Hz,1H),5.09(s,1H),3.46-3.59(m,4H),3.20-3.44(m,2H),2.86-3.17(m,3H),2.62-2.80(m,2H),2.08(m,J=6.80Hz,1H),1.38-1.41(m,9H),0.98(d,J=6.50Hz,3H)ppm。
步骤二:(6S,8R)-6-[2,6-二氟-4-(((R)-吡咯烷-3-基)氧基)苯基]-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000099
把三氟乙酸(6.16g,54.0mmol)溶于水中(0.4mL)后加入(R)-3-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-甲酸叔丁酯(503mg,609umol)里,20℃氮气保护下反应2小时。LCMS检测反应结束。混合物浓缩后用饱和碳酸氢钠调pH=7,乙酸乙酯萃取,有机相过滤浓缩,柱层析得黄色固体(6S,8R)-6-[2,6-二氟-4-(((R)-吡咯烷-3-基)氧基)苯基]-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(183mg,378umol,收率62.2%)。
LCMS:m/z 483.2[M+H] +
步骤三:(E)-4-((R)-3-((3,5-二氟4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-基)-N,N-二甲基丁-2-烯酰胺的合成
Figure PCTCN2022080680-appb-000100
将(6S,8R)-6-[2,6-二氟-4-(((R)-吡咯烷-3-基)氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(90mg,186umol),(E)-4-溴-N,N-二甲基丁-2-烯酰胺(35.7mg,186umol),N,N-二异丙基乙胺(52.9mg,409umol)溶于N,N-二甲基甲酰胺(1mL),0℃氮气保护下反应1小时。LCMS检测反应结束,反应液过滤送prep-HPLC分离得到的产物送SFC纯化得黄色固体(E)-4-((R)-3-((3,5-二氟4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-基)-N,N-二甲基丁-2-烯酰胺(18.5mg,30.7umol,收率16.5%)。
LCMS:m/z 579.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=6.82(d,J=8.25Hz,1H),6.60-6.71(m,2H),6.45-6.54(m,2H),6.44(s,1H),5.29(s,1H),4.81(m,J=7.30,4.60Hz,1H),3.52(m,J=6.00Hz,1H),3.31(s,1H),3.22(m,J=6.00Hz,2H),3.04-3.09(m,1H),3.01(s,3H),2.93-2.98(m,1H),2.89(s,3H),2.66-2.87(m,5H),2.42(m,J=7.50Hz,1H),2.30(m,J=7.60Hz,1H),1.07(d,J=6.50Hz,3H)ppm。
实施例38:(6S,8R)-6-(4-(((R)-1-丙烯酰吡咯烷-3-基)氧基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物38)
Figure PCTCN2022080680-appb-000101
步骤一:(6S,8R)-6-(4-(((R)-1-丙烯酰吡咯烷-3-基)氧基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000102
将(6S,8R)-6-[2,6-二氟-4-(((R)-吡咯烷-3-基)氧基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(90mg,186umol),二异丙基乙胺(240mg,1.86mmol)溶于N,N-二甲基甲酰胺(1mL),然后在0℃下加丙烯酰氯(16.8mg,186umol)后,在0℃反应1小时。LCMS检测反应结束,反应液过滤送prep-HPLC分离得到的产物送SFC纯化得白色固体(6S,8R)-6-(4-(((R)-1-丙烯酰吡咯烷-3-基)氧基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(24.8mg,45.9umol,收率 24.6%)。
LCMS:m/z 537.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=8.70-9.00(m,1H),6.82(d,J=8.13Hz,1H),6.63(d,J=8.00Hz,1H),6.44-6.60(m,3H),6.15-6.25(m,1H),5.64(ddd,J=10.35,8.10,2.31Hz,1H),5.30(s,1H),4.92-5.06(m,1H),3.54-3.88(m,4H),3.31-3.52(m,2H),3.07(dd,J=16.45,4.94Hz,1H),2.94(dq,J=15.90,9.63Hz,1H),2.76(dd,J=16.51,5.13Hz,1H),2.16-2.25(m,2H),1.07(d,J=6.63Hz,3H)ppm。
类似于前述实施例37及38的合成路线,选择相应的反应物和本领域技术人员已知的合成方式,合成以下实施例39及40。
实施例39:(E)-4-((S)-3-((3,5-二氟4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯氧基)吡咯烷-1-基)-N,N-二甲基丁-2-烯酰胺(化合物39)
Figure PCTCN2022080680-appb-000103
LCMS:m/z 579.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d3)δ=6.82(d,J=8.13Hz,1H),6.60-6.71(m,2H),6.48-6.53(m,1H),6.45-6.48(m,1H),6.43-6.45(m,1H),5.29(s,1H),4.77-4.84(m,1H),3.47-3.57(m,1H),3.32(m,J=9.60Hz,1H),3.22(dd,J=6.07,1.44Hz,2H),3.07(dd,J=16.76,4.75Hz,1H),3.00-3.04(m,3H),2.93-2.99(m,1H),2.89(s,3H),2.83(s,5H),2.37-2.49(m,1H),2.26-2.36(m,1H),1.07(d,J=6.50Hz,3H)ppm。
实施例40:(6S,8R)-6-(4-(((S)-1-丙烯酰吡咯烷-3-基)氧基)-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(化合物40)
Figure PCTCN2022080680-appb-000104
LCMS:m/z 537.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=6.82(d,J=8.00Hz,1H),6.62(d,J=8.00Hz,1H),6.44-6.59(m,3H),6.16-6.26(m,1H),5.64(m,J=10.40,10.40,2.30Hz,1H),5.30(s,1H),4.92-5.06(m,1H),3.55-3.88(m,4H),3.45-3.53(m,1H),3.34(m,J=16.10,9.60Hz,1H),3.03-3.12(m,1H),2.89-3.00(m,1H),2.76(dd,J=16.70,5.32Hz,1H),1.77(m,J=4.90,2.40Hz,2H),1.07(d,J=6.63Hz,3H)ppm。
实施例41:(E)-4-(4-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)哌嗪-1-基)-N,N-二甲基丁-2-烯酰胺(化合物41)
Figure PCTCN2022080680-appb-000105
步骤一:4-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)哌嗪-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000106
将(6S,8R)-6-(4-溴-2,6-二氟苯基)-8-甲基-7-(2,2,2-三氟乙基)-3-三苯甲基-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(700mg,972umo),N-Boc哌嗪(272mg,1.46mmol),(2-二环己基膦基-2’,4’,6’-三-异丙基-1,1’-联苯基)(2’-氨基-1,1’-联苯-2-基)钯(II)甲烷磺酸盐((41.2mg,48.6umol),碳酸铯(697mg,2.14mmol)溶于甲苯(15mL),110℃氮气保护下反应12小时。LCMS监测反应结束。混合物浓缩后用水和乙酸乙酯萃取,有机相饱和食盐水洗,硫酸钠干燥,浓缩。粗产物柱层析纯化得白色固体4-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)哌嗪-1-甲酸叔丁酯(413mg,501umol,收率51.5%)。
LCMS:m/z 824.3[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ=7.27-7.53(m,15H),6.24-6.42(m,3H),5.63-5.74(m,1H),5.17(s,1H),3.52-3.68(m,5H),3.10-3.39(m,6H),2.77(m,J=16.40,4.30Hz,2H),1.52(s,9H),0.89-1.11(m,3H)ppm。
步骤二:(6S,8R)-6-(2,6-二氟-4-(哌嗪-1-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000107
把三氟乙酸(12.3g,107mmol)溶于水中(0.1mL)后加入4-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-3-三苯甲基-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)哌嗪-1-甲酸叔丁酯(413mg,500umol)里,20℃氮气保护下反应2小时。LCMS检测反应结束。粗产物柱层析得黄色固体(6S,8R)-6-(2,6-二氟-4-(哌嗪-1-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑[5,4-f]异喹啉-2(3H)-酮(110mg,228umol,收率45.5%)。
LCMS:m/z 482.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ=6.81(d,J=8.00Hz,1H),6.48-6.60(m,3H),5.16(s,1H),3.06-3.10(m,4H),2.86-2.99(m,3H),2.67-2.80(m,6H),1.05(d,J=6.50Hz,3H.)ppm。
步骤三:(E)-4-(4-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)哌嗪-1-基)-N,N-二甲基丁-2-烯酰胺的合成
Figure PCTCN2022080680-appb-000108
将6S,8R)-6-(2,6-二氟-4-(哌嗪-1-基)苯基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑[5,4-f]异喹啉-2(3H)-酮(100mg,207umol),(E)-4-溴-N,N-二甲基丁-2-烯酰胺(39.8mg,207.3umol),二异丙基乙胺(58.9m,456umol)溶于N,N-二甲基甲酰胺(2mL),20℃氮气保护下反应1小时。LCMS检测反应结束,反应液过滤送prep-HPLC分离得到的产物送SFC纯化得黄色固体(E)-4-(4-(3,5-二氟-4-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)苯基)哌嗪-1-基)-N,N-二甲基丁-2-烯酰胺(37mg,62.3umol,收率30.1%)。
LCMS:m/z 593.2[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=8.78-8.97(m,1H),6.81(d,J=8.13Hz,1H),6.50-6.69(m,3H),6.38-6.47(m,2H),5.25(s,1H),3.45-3.59(m,1H),3.24-3.40(m,1H),3.12-3.22(m,6H),3.04-3.10(m,1H),3.03(s,3H),2.92-3.00(m,1H),2.90(s,3H),2.70-2.79(m,1H),2.46-2.60(m,4H),1.07(d,J=6.63Hz,3H)ppm。
实施例42:3-(4-((2-(4-(3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)乙基)哌嗪-1-基)乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(化合物42)
Figure PCTCN2022080680-appb-000109
步骤一:4-(2-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)乙基)哌嗪-1-甲酸叔丁酯的合成
Figure PCTCN2022080680-appb-000110
把(6S,8R)-6-(5-溴吡啶-2-基)-8-甲基-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑[5,4-f]异喹啉-2(3H)-酮(700mg,1.58mmol),4-(2-羟乙基)哌嗪-1-甲酸叔丁酯(546mg,2.37mmol),[(2-双-叔丁基膦-3-甲氧基-6-甲基-2,4,6-三异丙基-1,1-联苯)-2-(2—氨基联苯基)]合钯(II)甲磺酸盐(132mg,158umol)和叔丁醇钠(456mg,4.75mmol)溶于甲苯(10mL)后,氮气保护下在60℃反应12小时。TLC监测反应结束。反应液浓缩。粗产物柱层析纯化得黄色固体4-(2-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)乙基)哌嗪-1-甲酸叔丁酯(400mg,676umol,收率42.7%)。
LCMS:m/z 591.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ=11.36-11.63(m,1H),7.93-8.18(m,1H),7.37(dd,J=8.69,2.94Hz,1H),7.11-7.27(m,1H),6.84(d,J=8.13Hz,1H),6.68(d,J=8.13Hz,1H),5.02(s,1H),4.01-4.19(m,2H),3.37-3.64(m,3H),3.17(d,J=5.13Hz,1H),2.89-3.03(m,1H),2.80-2.89(m,1H),2.55-2.76(m,3H),2.24-2.45(m,6H),1.38(s,9H),1.06(m,J=6.50Hz,3H)ppm。
步骤二:(6S,8R)-8-甲基-6-(5-(2-(哌嗪-1-基)乙氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮的合成
Figure PCTCN2022080680-appb-000111
把4-(2-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)乙基)哌嗪-1-甲酸叔丁酯(392mg,662.5umol)和三氟乙酸(6.16g,54.0mmol)溶于二氯甲烷(10mL)中,在20℃反应1小时。LCMS监测反应结束。反应液浓缩后用饱和的碳酸氢钠溶液调pH=7-8,用水和乙酸乙酯萃取,有机相饱和食盐水洗,硫酸钠干燥,浓缩。粗产品柱层析纯化得黄色固体(6S,8R)-8-甲基-6-(5-(2-(哌嗪-1-基)乙氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(156mg,317umol,收率47.9%)。
LCMS:m/z 491.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ=8.08-8.20(m,1H),7.36(dd,J=8.63,2.88Hz,1H),7.25(d,J=8.63Hz,1H),6.84(d,J=8.13Hz,1H),6.68(d,J=8.13Hz,1H),5.03(s,1H),4.11(br t,J=5.44Hz,2H),3.52-3.59(m,1H),3.34-3.42(m,1H),2.80-2.99(m,6H),2.63-2.76(m,3H),2.53-2.63(m,4H),1.06(d,J=6.63Hz,3H)ppm。
步骤三:3-(4-((2-溴乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮的合成
Figure PCTCN2022080680-appb-000112
把3-(4-氨基-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(2.00g,7.71mmol),1,2-二溴乙烷(17.3g,92.5mmol)和N,N-二甲基异丙胺(2.99g,23.1mmol)溶于N-甲基吡咯烷酮(20mL)后,置换氮气,然后在100℃反应4小时。LCMS监测反应结束。反应液水洗,乙酸乙酯萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,浓缩。粗产品柱层析纯化得黄色固体3-(4-((2-溴乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(403mg,1.10mmol,收率14.27%)。
LCMS:m/z 365.0[M+H] +
1H NMR(400MHz,DMSO-d 6)δ=11.00(s,1H),7.31(t,J=7.75Hz,1H),6.98(d,J=7.25Hz,1H),6.84(d,J=8.00Hz,1H),5.93(br s,1H),5.11(dd,J=13.26,5.00Hz,1H),4.18-4.28(m,1H),4.09-4.17(m,1H),3.53-3.64(m,4H),2.86-2.99(m,1H),2.64(br s,1H),2.23-2.37(m,1H),1.98-2.09(m,1H)ppm。
步骤四:3-(4-((2-(4-(3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)乙基)哌嗪-1-基)乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮的合成
Figure PCTCN2022080680-appb-000113
把(6S,8R)-8-甲基-6-(5-(2-(哌嗪-1-基)乙氧基)吡啶-2-基)-7-(2,2,2-三氟乙基)-6,7,8,9-四氢恶唑并[5,4-f]异喹啉-2(3H)-酮(70mg,142umol),3-(4-((2-溴乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(57.3mg,156umol),N,N-二异丙基乙胺(55.2mg,427umol)溶于二氧六环(2mL)里,然后在氮气保护下80℃反应12小时。LCMS监测反应结束。反应液旋干后送prep-HPLC纯化,得到黄色固体3-(4-((2-(4-(3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)乙基)哌嗪-1-基)乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(20mg,25.8umol,收率18.1%)。
LCMS:m/z 776.3[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=8.91(br s,1H),8.10(d,J=2.50Hz,1H),7.31-7.37(m,1H),7.28(s,1H),7.26(br d,J=2.75Hz,1H),7.05(d,J=7.38Hz,1H),6.83(t,J=7.69Hz,2H),6.70(d,J=8.00Hz,1H),4.98-5.18(m,2H),4.54-4.91(m,1H),4.22-4.31(m,1H),4.19(s,1H),4.13(t,J=5.57Hz,2H),3.36-3.49(m,2H),3.28-3.35(m,2H),2.84-3.07(m,3H),2.69-2.83(m,7H),2.68(br s,8H),2.39-2.43(m,1H),1.28(br d,J=6.25Hz,1H),1.08(d,J=6.75Hz,3H)ppm。
类似于前述实施例42的合成路线,选择相应的反应物和本领域技术人员已知的合成方式,合成以下实施例43。
实施例43:3-(4-((2-(4-(3-((6-((6S,8R)-8-甲基-2-氧代-7-(2,2,2-三氟乙基)-2,3,6,7,8,9-六氢恶唑并[5,4-f]异喹啉-6-基)吡啶-3-基)氧基)丙基)哌嗪-1-基)乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(化合物43)
Figure PCTCN2022080680-appb-000114
LCMS:m/z 790.3[M+H] +
1H NMR(400MHz,ACETONITRILE-d 3)δ=8.71-9.01(m,1H),8.09(d,J=2.50Hz,1H),7.40(s,1H),7.26-7.30(m,1H),7.20-7.26(m,1H),7.04(d,J=7.38Hz,1H),6.83(t,J=7.69Hz,2H),6.70(d,J=8.13Hz,1H),5.02-5.17(m,2H),4.50(br s,1H),4.18(q,J=16.38Hz,2H),4.06(t,J=6.32Hz,2H),3.43-3.53(m,1H),3.33-3.43(m,1H),3.26(br d,J=5.25Hz,2H),2.90-3.06(m,2H),2.73-2.87(m,2H),2.70(br dd,J=4.57,2.69Hz,1H),2.59(br t,J=6.19Hz,3H),2.29-2.53(m,12H),1.28(br d,J=6.13Hz,1H),1.08(d,J=6.63Hz,3H)ppm。
测试例
以下各测试例中,AZD9496(目录号:HY-12870),AZD9833(目录号:HY-136255)和他莫昔芬(目录号:HY-13757A)购自MedChemExpress;对比化合物1(化学结构:(6S,8R)-6-(2,6-二氟-4-(2-(3-(氟甲基)氮杂环丁烷-1-基)乙氧基)苯基)-7-(2-氟-2-甲基丙基)-8-甲基-6,7,8,9-四氢-3H-吡唑并[4,3-f]异喹啉)采用WO2017/182493A1中实施例1的方法制备。
测试例1:ERαTR-FRET测试
准备1X Tris-HCl(Sigma,PHG0002)蛋白缓冲液,混合后待用。将待检测化合物配置成浓度为2mM的储存液,再依次3倍梯度稀释,共稀释10个浓度。用Echo 550分别往反应板加入稀释好的化合物,每孔100nL,同时每孔加入5nL雌二醇(Sigma,491187;终浓度为1.5nM)。
准备1x蛋白混合液:先按照下表准备2x GST-ERα-LBD(Invitrogen,A15677)/MAb抗GST-Eu(Cisbio,61GSTKLA)混合溶液。
物质 最终浓度(nM) 工作浓度(nM) 原液浓度(nM)
GST-ERα-LBD 2 4 20100
MAb抗GST-Eu 2.5ng/孔 50nl/孔 50μg/ml
配置2x生物素-SRC2/抗生蛋白链菌素-XL665(Cisbio,610SAXLA)混合溶液。
物质 最终浓度(nM) 工作浓度(nM) 原液浓度(nM)
生物素-SRC2 75 150 1000000
抗生蛋白链菌素-XL665 50ng/孔 50nl/孔 1mg/ml
把以上两种2x GST-NR-LBD/MAb抗GST-Eu溶液和2x生物素-SRC2/抗生蛋白链菌素-XL665溶液按体积1:1混合均匀;将1x蛋白混合溶液加入384孔板每个孔,每孔加20μL,并将384孔板放入离心机室温1000转离心10秒,取出后在室温放置3小时后使用EnVision多功能酶标仪读数。
读数665和615(纳米)值,并以615值做校正值,最终数值表示为665值/615值;抑制率按照以下公式计算:
Figure PCTCN2022080680-appb-000115
X是每个浓度的―665值比615值‖。Min是加了0.2mM阳性对照化合物和5nl 3nM (1.5nM)雌二醇的―665值比615值‖平均值。Max是DMSO和5nl 3nM(1.5nM)雌二醇的―665值比615值‖平均值。将数据导入Graphpad Prism并使用Log(agonist)vs.response--variable slope进行曲线拟合。
拟合公式:Y=Bottom+(Top-Bottom)/(1+10^((LogEC 50-X)*HillSlope))
ERαTR-FRET实验数据
化合物 IC 50(nM) 最大抑制率
实施例1 9.00 100.2
实施例9 21.6 100.1
实施例10 7.80 100.2
实施例12 15.6 100.3
实施例14 40.3 100.2
实施例25 11.4 100.8
实施例26 58.4 100.4
实施例30 8.85 99.8
实施例31 45.6 100.9
实施例32 14.7 100.3
实施例33 11.8 99.9
实施例34 19.4 100.2
AZD9833 62.0 99.8
他莫昔芬 578.1 100.4
测试例2:MCF-7细胞增殖试验
在96孔板中接种人乳腺癌细胞MCF-7(HTB-22,购自ATCC),培养基为含10%FBS(Gibco,10099-141),1%丙酮酸(Sigma,S8636),1%非必需氨基酸(Sigma,M7145)的MEM(Hyclone,SH30024.01B)培养基,接种密度为4,000个细胞/孔,细胞在37℃、5%CO 2条件下培养。将化合物配置成20mM的储存液,用100%DMSO(Sigma,D2650)梯度稀释成1000终浓度,再用含2%FBS的培养基稀释20倍。培养24小时后去掉培养基,每孔加入90μL含2%FBS的培养基和10μL药物,对照组加入10μL DMSO,轻轻振荡混匀,空白组只含100μL 2%FBS培养基,放置37℃、5%CO 2培养箱中培养,72小时后加入50μL混合后的Cell Titer-Glo(Promega,G7571),振荡混匀,室温放置10min,测定化学发光信号值,阴性对照为0.5%DMSO孔。
MCF-7细胞增殖试验实验数据
化合物 IC 50(nM)
实施例1 0.39
实施例9 0.19
实施例10 0.20
实施例11 0.38
实施例12 0.49
实施例13 0.30
实施例14 0.44
实施例16 0.80
实施例25 0.24
实施例26 0.55
实施例27 1.63
实施例28 0.40
实施例29 0.71
实施例30 0.21
实施例31 0.85
实施例32 0.39
实施例33 0.39
实施例34 0.19
实施例35 0.82
实施例37 0.61
实施例39 0.49
实施例43 0.69
AZD9496* 3.84
AZD9833* 2.45
他莫昔芬* 274.50
对比化合物1* 2.68
测试例3:化合物对ERα的降解作用
本测试例使用ERαDuoSet IC ELISA Kit(R&D,DYC5715)检测化合物对ERα的降解作用。试验第一天,在24孔板中接种人乳腺癌细胞MCF-7(来源同上);培养基为含10%的活性炭处理FBS(Tocyto,UT61204),1%丙酮酸,1%非必需氨基酸的MEM培养基;接种密度为150,000个细胞/孔;细胞在37℃、5%CO2条件下培养24小时。
试验第二天,将待测化合物梯度稀释,加入到含有细胞的24孔板中。细胞在37℃、5%CO2条件下继续培养24小时。ERα捕获抗体用PBS(Corning,21-040-CVR)稀释至1μg/ml,以100μL/孔加入96孔板中,封板并在室温条件下包被过夜。
试验第三天,将包被后的96孔板用PBS洗3次,每孔加入300μL封闭液(R&D,DY995),室温封闭1小时。24孔板用PBS洗1次,吸掉残余液体,向每孔加入60μL裂解液(6M尿素(Sigma,U5378)、1mM EDTA(Sigma,E9884)、0.5%TritonX-100(Sigma,T8787)、1mM PMSF(Sigma,93482)、蛋白酶抑制剂混合物(Sigma,05892970001))于冰上裂解15分钟后,加入稀释液(1mM EDTA,0.5%TritonX-100溶于PBS);取稀释的细胞裂解液加入封闭后的96孔板中,每孔100μL,室温孵育2小时;用洗液(R&D,WA126)洗板3次后加入一抗,孵育1小时后,将96孔板洗4次,加入二抗,室温避光孵育30分钟;用洗液洗板4次,加入TMB(R&D,DY999)显色15分钟后用终止液(R&D,DY994)终止反应,读取450nm光吸收。
化合物对ERα降解作用的IC 50
Figure PCTCN2022080680-appb-000116
Figure PCTCN2022080680-appb-000117
测试例4:体外人体肝微粒体稳定性试验
准备8块96孔孵育板,分别命名为T0、T5、T15、T30、T45、T60、Blank60和NCF60。前6块孵育板对应的反应时间点分别为0、5、15、30、45和60分钟。Blank60板中不加入待测化合物或对照化合物并在孵育60分钟后取样。NCF60板中用磷酸钾盐缓冲液代替NADPH再生体系溶液(Sigma,N0505)进行孵育60分钟。
分别在T0、T5、T15、T30、T45、T60和NCF60板上添加2μL待测化合物或对照品工作液和100μL人体肝脏微粒体工作液(Corning,452117),微粒体蛋白浓度为1mg/mL)。在Blank60板中只添加微粒体工作液,然后将除T0和NCF60外的孵育板Blank60、T5、T15、T30、T45和T60放置于37℃水浴锅中预孵育大约10分钟。
T0板样品先加入600μL的终止液(含100ng/mL甲苯磺丁脲(Sigma,T0891)的乙腈:甲醇(95:5,v/v)溶液)后,再添加NADPH再生体系工作液。
在NCF60板上每孔添加98μL磷酸钾盐缓冲液,孵育60分钟。
孵育板Blank60、T5、T15、T30、T45和T60预孵育结束后,每个样品孔内添加98μL NADPH再生体系工作液以启动反应。
孵育适当时间(如5、15、30、45和60分钟)后,分别在Blank60、T5、T15、T30、T45、T60和NCF60板的每个样品孔和对照品样品孔中加入600μL的终止液以终止反应。
所有样品板摇匀并在3220×g离心20分钟,分别取200μL试验品上清液稀释到200μL含0.3%甲酸的水溶液中用于LC-MS/MS分析,取100μL对照品上清液稀释到300μL纯水中用于LC-MS/MS分析,并按照如下公式计算:
Figure PCTCN2022080680-appb-000118
Figure PCTCN2022080680-appb-000119
Figure PCTCN2022080680-appb-000120
Figure PCTCN2022080680-appb-000121
Figure PCTCN2022080680-appb-000122
体外微粒体稳定性实验数据
Figure PCTCN2022080680-appb-000123
Figure PCTCN2022080680-appb-000124

Claims (10)

  1. 式(I)的化合物,或其立体异构体或药学上可接受的盐:
    Figure PCTCN2022080680-appb-100001
    其中:
    Z 1选自CR aR b、C(O)和键;
    Z 2选自O、S、NR c、C(O)和键,或任选被一个或多个相同或不同的R d取代的C 1-C 6亚烷基、O-(C 1-C 6亚烷基)或NH-(C 1-C 6亚烷基);
    Cy 1选自C 6-14亚芳基、C 3-8亚环烷基、C 5-14亚杂芳基、C 3-14亚杂环烷基、C 3-14亚杂环烯基,其各自任选被选自以下的基团取代:卤素原子、羟基、氨基、氰基以及任选被卤素原子取代的C 1-6烷基或C 1-6烷氧基;
    Cy 2选自键、C 3-10亚环烷基、C 3-14亚杂环烷基,其各自任选被选自以下的基团取代:卤素原子(包括F、Cl、Br或I原子)、羟基、氨基、氰基以及任选被卤素原子取代的C 1-6烷基或C 1-6烷氧基;
    R 1、R 2各自独立地为H、卤素原子、羟基、氨基、氰基、C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基或氧代基的基团取代;
    R 4为H、卤素原子、羟基、氨基、氰基、C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基、氧代基、C 6-14芳基、C 5-14杂芳基、C 3-14亚杂环烷基或
    Figure PCTCN2022080680-appb-100002
    (例如
    Figure PCTCN2022080680-appb-100003
    )
    的基团取代;
    优选地,R 4为H、卤素原子、羟基、氨基、氰基、C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基或氧代基的基团取代;
    R 3为-(CR eR f) m-CR 31R 32R 33,其中m为1、2或3;
    R 31、R 32和R 33独立选自:H、C 1-6烷基、卤素原子、氰基,且R 31和R 32可以共同形成C 3-8亚环烷基,所述C 1-6烷基和C 3-8亚环烷基任选被羟基、氰基、氨基或卤素原子取代;
    R a、R b、R c、R d、R e、R f各自独立地为H、卤素原子、羟基、氨基、氰基、任选被卤素原子取代的C 1-6烷基。
  2. 根据权利要求1所述的化合物,或其立体异构体或药学上可接受的盐,其中
    Z 1选自CR aR b、C(O)和键;
    Z 2选自O、S、NR c、C(O)和键,或任选被一个或多个相同或不同的R d取代的C 1-C 6亚烷基、O-(C 1-C 6亚烷基)或NH-(C 1-C 6亚烷基);
    Cy 1选自C 6-14亚芳基(优选C 6-10亚芳基)和C 5-14亚杂芳基(优选C 5-10亚杂芳基),其各自任选被选自以下的基团取代:卤素原子、以及任选被卤素原子取代的C 1-6烷基或C 1-6烷氧基;
    Cy 2选自键、C 3-10亚环烷基(优选C 3-8亚环烷基)、C 3-14亚杂环烷基(优选C 3-10亚杂环烷基);
    R 1、R 2各自独立地为H或C 1-6烷基,所述C 1-6烷基任选被一个或多个选自卤素原子、羟基或氰基的基团取代;
    R 3为-(CR eR f) m-CR 31R 32R 33,其中m为1、2或3;
    R 31、R 32和R 33独立选自:H、C 1-6烷基、卤素原子、氰基,且R 31和R 32可以共同形成C 3-8亚环烷基,所述C 1-6烷基和C 3-8亚环烷基任选被羟基、氰基、氨基或卤素原子取代;
    R a、R b、R c、R d、R e、R f各自独立地为H或卤素原子;
    R 4为C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基、氧代基和
    Figure PCTCN2022080680-appb-100004
    的基团取代。
  3. 根据权利要求2所述的化合物,或其立体异构体或药学上可接受的盐,其中,R 4为C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基、C 3-C 8环烷基,所述C 1-6烷基、
    C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基或C 3-C 8环烷基任选被一个或多个选自卤素原子、羟基、氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、氰基或氧代基的基团取代。
  4. 根据权利要求1-3中任一项所述的化合物,或其立体异构体或药学上可接受的盐,其中
    Z 1为键;
    Z 2选自-O-CH 2-CH 2-、-O-CH 2-、-NH-CH 2-CH 2-、-NH-CH 2-、-NH-或-O-;
    Cy 1为C 6-10亚芳基(优选苯基)和C 5-10亚杂芳基(优选吡啶基),其任选被卤素原子(优选F)或C 1-6烷氧基(优选甲氧基)取代;
    Cy 2选自键、亚氮杂环丁基、亚吡咯烷基、亚哌嗪基、
    Figure PCTCN2022080680-appb-100005
    R 1为C 1-6烷基(优选甲基);
    R 2为H;
    R 3为-CH 2-CR 31R 32R 33
    R 31、R 32和R 33独立选自:H、任选被羟基或卤素原子(优选F)取代的C 1-6烷基(优选甲基)、卤素原子(优选F)、氰基,且R 31和R 32可以共同形成任选被羟基或卤素原子(优选F)取代的C 3-8亚环烷基(优选亚环丙基);
    R 4为任选被选自卤素原子(优选F)、氧代基、C 1-6烷基氨基(优选甲基氨基)、(C 1-6烷基) 2氨基(优选二甲基氨基)、和
    Figure PCTCN2022080680-appb-100006
    的基团取代的C 1-6烷基、C 1-6烷基氨基、氨基C 1-6烷基、C 2-6烯基、C 2-6烯基氨基。
  5. 根据权利要求4所述的化合物,或其立体异构体或药学上可接受的盐,其中
    Cy 2选自键、亚氮杂环丁基、亚吡咯烷基,且
    R 4为任选被选自卤素原子(优选F)、氧代基、C 1-6烷基氨基(优选甲基氨基)和(C 1-6烷基) 2氨基(优选二甲基氨基)的基团取代的C 1-6烷基、C 1-6烷基氨基、C 2-6烯基、C 2-6烯基氨基。
  6. 根据权利要求1-5中任一项所述的化合物,或其立体异构体或药学上可接受的盐,其中
    Z 1为键;
    Z 2选自-NH-或-O-;
    Cy 1为苯基或吡啶基(优选苯基),其任选被F或甲氧基取代;
    Cy 2选自亚氮杂环丁基、亚吡咯烷基、亚哌嗪基;
    R 1为甲基;
    R 2为H;
    R 3为-CH 2-CR 31R 32R 33
    R 31、R 32各为F,或R 31和R 32共同形成亚环丙基;
    R 33选自:H、羟甲基、氟甲基,优选选自F、羟甲基、氟甲基;
    R 4为F-(CH 2) 3-或
    Figure PCTCN2022080680-appb-100007
  7. 根据权利要求6所述的化合物,或其立体异构体或药学上可接受的盐,其中Cy 2选自亚氮杂环丁基和亚吡咯烷基,且R 4为F-(CH 2) 3-。
  8. 根据权利要求1所述的化合物,或其立体异构体或药学上可接受的盐,其中所述化合物选自以下化合物:
    Figure PCTCN2022080680-appb-100008
    Figure PCTCN2022080680-appb-100009
    Figure PCTCN2022080680-appb-100010
  9. 一种药物组合物,其包含根据权利要求1-8中任一项所述的化合物或其立体异构体或药学上可接受的盐,以及药学上可接受的载体。
  10. 根据权利要求1-8中任一项所述的化合物或其立体异构体或药学上可接受的盐在制备用于治疗哺乳动物的雌激素受体依赖性或由雌激素受体介导的疾病的药物中的用途。
PCT/CN2022/080680 2021-03-15 2022-03-14 雌激素受体调节剂 WO2022194096A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2023557230A JP2024511044A (ja) 2021-03-15 2022-03-14 エストロゲン受容体モジュレーター
EP22770451.7A EP4310079A1 (en) 2021-03-15 2022-03-14 Estrogen receptor antagonist
KR1020237035079A KR20230157439A (ko) 2021-03-15 2022-03-14 에스트로겐 수용체 조절제
AU2022240650A AU2022240650A1 (en) 2021-03-15 2022-03-14 Estrogen receptor antagonist

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110277849 2021-03-15
CN202110277849.3 2021-03-15

Publications (1)

Publication Number Publication Date
WO2022194096A1 true WO2022194096A1 (zh) 2022-09-22

Family

ID=83246765

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/080680 WO2022194096A1 (zh) 2021-03-15 2022-03-14 雌激素受体调节剂

Country Status (6)

Country Link
EP (1) EP4310079A1 (zh)
JP (1) JP2024511044A (zh)
KR (1) KR20230157439A (zh)
CN (1) CN115073493A (zh)
AU (1) AU2022240650A1 (zh)
WO (1) WO2022194096A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016202161A1 (zh) * 2015-06-16 2016-12-22 江苏恒瑞医药股份有限公司 哌啶类衍生物、其制备方法及其在医药上的应用
US20170305909A1 (en) * 2016-04-20 2017-10-26 Astrazeneca Ab Chemical compounds
CN109219604A (zh) * 2016-04-08 2019-01-15 豪夫迈·罗氏有限公司 四氢异喹啉雌激素受体调节剂及其用途
WO2019192533A1 (zh) * 2018-04-04 2019-10-10 深圳福沃药业有限公司 用于治疗乳腺癌的雌激素受体降解剂
WO2019223715A1 (zh) * 2018-05-23 2019-11-28 江苏恒瑞医药股份有限公司 苯并哌啶或杂芳基并哌啶类衍生物、其制备方法及其在医药上的应用

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016202161A1 (zh) * 2015-06-16 2016-12-22 江苏恒瑞医药股份有限公司 哌啶类衍生物、其制备方法及其在医药上的应用
CN109219604A (zh) * 2016-04-08 2019-01-15 豪夫迈·罗氏有限公司 四氢异喹啉雌激素受体调节剂及其用途
US20170305909A1 (en) * 2016-04-20 2017-10-26 Astrazeneca Ab Chemical compounds
WO2017182493A1 (en) 2016-04-20 2017-10-26 Astrazeneca Ab Indazole derivatives for use in down-regulation of the estrogen receptor for the treatment of cancer
WO2019192533A1 (zh) * 2018-04-04 2019-10-10 深圳福沃药业有限公司 用于治疗乳腺癌的雌激素受体降解剂
WO2019223715A1 (zh) * 2018-05-23 2019-11-28 江苏恒瑞医药股份有限公司 苯并哌啶或杂芳基并哌啶类衍生物、其制备方法及其在医药上的应用

Also Published As

Publication number Publication date
KR20230157439A (ko) 2023-11-16
CN115073493A (zh) 2022-09-20
EP4310079A1 (en) 2024-01-24
JP2024511044A (ja) 2024-03-12
AU2022240650A1 (en) 2023-10-19

Similar Documents

Publication Publication Date Title
KR102614939B1 (ko) 피리미딘-융합고리 화합물 및 그의 제조방법과 용도
CN110156786B (zh) 嘧啶并环化合物及其制备方法和应用
CN114085212B (zh) 异吲哚啉类化合物、其制备方法、药物组合物及用途
CN113454083A (zh) Kras突变蛋白抑制剂
ES2902050T3 (es) Derivado 1,2-dihidro-3H-pirazolo[3,4-D]pirimidin-3-ona como inhibidor de Wee1
JP2022533398A (ja) Kras g12cインヒビター及びその使用
CN113286794A (zh) Kras突变蛋白抑制剂
CN110078741B (zh) 稠环嘧啶氨基衍生物﹑其制备方法、中间体、药物组合物及应用
US20230212170A1 (en) Inhibitors of kras g12c protein and uses thereof
CN106749233B (zh) 一类磺酰胺衍生物及其应用
CN112778276A (zh) 作为shp2抑制剂的化合物及其应用
CA2993096A1 (en) Fused ring pyrimidine compound, intermediate, and preparation method, composition and use thereof
CN111065639A (zh) 一类细胞周期依赖性激酶的降解剂、其制备方法、药物组合物及其用途
JP2020519564A (ja) アミノピリミジン縮合5員複素環化合物、その中間体、製造方法、薬物組成物および使用
TW202237597A (zh) 新型egfr降解劑
CN110655520A (zh) 嘧啶并环化合物及其制备方法和应用
CN114805311A (zh) 螺环茚
CN114805361B (zh) 一类氨基取代的芳香杂环并吡唑类化合物、制备方法和用途
WO2022127827A1 (zh) Kras g12c蛋白突变抑制剂、其制备方法、药物组合物及其应用
CA3234693A1 (en) Novel modulators of ehmt1 and ehmt2 and therapeutic use thereof
WO2022194096A1 (zh) 雌激素受体调节剂
EP4206197A1 (en) Preparation method for novel rho-related protein kinase inhibitor and intermediate in preparation method
CN102190669A (zh) 二氢喋啶酮类衍生物、其制备方法及其在医药上的应用
WO2024006776A1 (en) Estrogen receptor alpha degraders and medical use thereof
CN116600808A (zh) 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用

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: 22770451

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 18282147

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2023557230

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: AU2022240650

Country of ref document: AU

Ref document number: 2022240650

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 20237035079

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020237035079

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2022770451

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022240650

Country of ref document: AU

Date of ref document: 20220314

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2022770451

Country of ref document: EP

Effective date: 20231016