CN117396485A - PARP7 inhibitors - Google Patents

PARP7 inhibitors Download PDF

Info

Publication number
CN117396485A
CN117396485A CN202280037061.8A CN202280037061A CN117396485A CN 117396485 A CN117396485 A CN 117396485A CN 202280037061 A CN202280037061 A CN 202280037061A CN 117396485 A CN117396485 A CN 117396485A
Authority
CN
China
Prior art keywords
alkyl
alkoxy
amino
hydroxy
cyano
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202280037061.8A
Other languages
Chinese (zh)
Inventor
刘斌
陈博
田金龙
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Xuanzhu Biopharmaceutical Co Ltd
Original Assignee
Xuanzhu Biopharmaceutical Co Ltd
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 Xuanzhu Biopharmaceutical Co Ltd filed Critical Xuanzhu Biopharmaceutical Co Ltd
Publication of CN117396485A publication Critical patent/CN117396485A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic 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 three hetero rings
    • C07D498/14Ortho-condensed systems

Abstract

The invention belongs to the technical field of medicines, and in particular relates to a PARP7 kinase inhibitor compound shown in a general formula (I), a pharmaceutically acceptable salt or an isomer thereof, a pharmaceutical composition containing the compound, the pharmaceutically acceptable salt or the isomer thereof, a method for preparing the compound, the pharmaceutically acceptable salt or the isomer thereof, and application of the compound, the pharmaceutically acceptable salt or the isomer thereof.

Description

PARP7 inhibitors Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a PARP7 inhibitor compound, a pharmaceutically acceptable salt or an isomer thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salt or the isomer thereof, a method for preparing the compound, the pharmaceutically acceptable salt or the isomer thereof, and application of the compound, the pharmaceutically acceptable salt or the isomer thereof.
Background
There are many relevant targets in the development of tumors, and the family of Poly ADP-ribose polymerase (Poly-ADP-ribose polymerases, PARPs) is responsible for transferring single or multiple ADP ribose subunits from NAD+ to a substrate target, changing the function of the target protein, performing post-translational modification, regulating basic processes such as cellular gene expression, protein degradation, cellular stress, and the like, and participating in tumor formation. Based on the homology of the human genomic catalytic domain, the PARP family can be divided into 17 members, each of which is highly expressed in different cancers.
The family of PARPs can be divided into three classes according to catalytic activity, the first class belongs to the polyprps, and comprises 4 members of PARP 1, PARP 2, PARP 5a and PARP 5b, and the 4 members catalyze the transfer of a plurality of ADP-ribose subunits to a substrate, and a plurality of inhibitor drugs of the family are marketed and play a role in DNA-induced damage; the second class belongs to monoprop and comprises 12 members of PARP 3, PARP 4, PARP 6, PARP7, PARP 8, PARP 9, PARP 10, PARP 11, PARP 12, PARP 14, PARP 15, PARP 16, catalyzing the transfer of a single ADP-ribose subunit to a substrate; the third class comprises PARP 13, which is not currently found to be catalytically active.
TIPARP (TCDD-Industable Poly-ADP-Ribose Polymerase) is 2,3,7,8-tetrachlorodibenzo p-dioxin (2, 3,7, 8-tetrahydrodibenzo-p-dioxin (dioxin), TCDD) induced Poly ADP-ribose polymerase, also known as PARP7 or ARTD14, involved in processes such as tumor formation, cell proliferation and immunosuppression. TIPARP (PARP 7 or ARTD 14) belongs to monopPARPs, can transfer a single ADP-ribose subunit to a target protein, changes the function of the target protein, belongs to posttranslational modification, participates in the regulation of vital activities such as cell proliferation and the like, and has multiple cell functions.
The transcription factor aromatic hydrocarbon receptor (Aryl hydrocarbon receptor, AHR) belongs to the transcription regulator bHLH-PAS family, mainly regulates and controls the development and physiological functions of organisms, including neurogenesis, tracheal and salivary duct formation, toxin metabolism, circadian rhythm, hypoxia response, hormone receptor functions and the like, and can be activated by environmental pollutants such as exogenous ligands such as TCDD and the like, toxic metabolites such as endogenous ligands such as kynurenine and micro-organisms and ligand small molecules from food sources, and plays different biological effects in different cells. After AHR activation, the transcription and expression of the downstream genes such as TIPARP (PARP 7 or ARTD 14), CYP1A1 and the like are promoted, wherein the TIPARP can play a role of negative regulation part of proteins through transferring single ADP-ribose subunit. The TIPARP may also be regulated by other transcription factors or signaling pathways.
In tumors or after infection of cells with virus, cytoplasmic free RNA or DNA is increased and TBK1 kinase (TANK binding kinase 1) plays a major role in this pathway in activating type I interferon responses and antiviral immunity. The TIPARP enables the kinase region of TBK1 to generate ADP-ribosylation, the TBK1 is converted into an inactive state from a free activation state, and the I-type interferon response and the subsequent innate immune response are inhibited, so that T cell mediated adaptive anti-tumor immunity is inhibited, and finally the immune activation of the whole organism is influenced. Inhibiting TIPARP can restore type I interferon response and further improve immune response.
The TIPARP is over-expressed in cancers such as lung cancer, esophagus cancer, ovarian cancer, head and neck cancer, cervical cancer and the like, especially in lung squamous cell carcinoma, and the inhibition of the TIPARP can inhibit cell proliferation. Therefore, the TIPARP inhibitor can not only directly inhibit the growth of tumor cells, but also repair I-type interferon signals, and relieve the immune escape of the tumor cells and the inhibition of an immune system.
Therefore, the TIPARP inhibitor has great potential in the aspect of treating cancers or other related diseases, no small molecular inhibitor with the target point is marketed at present, and the development of the high-efficiency low-toxicity TIPARP inhibitor has important clinical significance.
Disclosure of Invention
The invention aims to solve the technical problem of providing a polycyclic compound with a novel structure and good inhibition effect on PARP 7. Furthermore, the compounds can be used for preventing and/or treating diseases related to PARP7 over-expression. Further, the compounds are useful in the prevention and/or treatment of cancers, including carcinoma in situ and metastatic cancers.
The technical scheme of the invention is as follows:
in one aspect, the present invention provides a compound represented by the following general formula (I'), a pharmaceutically acceptable salt thereof or an isomer thereof,
wherein,
ring B is a three-ring structure; the ring B is optionally substituted with 1 to 3Q 1 s and is also optionally substituted with 1 or more R 8 Substitution; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group; each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1- 6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1- 6 Alkoxy, cyano C 1-6 An alkoxy group;
l is selected from
X、X 1 、X 2 Are each independently selected from-CH (R) 6 )-、-N(R 7 ) -, -O-, -S-; -C (O) -, -S (O) -or-S (O) 2 -;
R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
each R 4 Each R 4 ', each R 5 Each R 5 ', each R 6 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1- 6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1- 6 Alkoxy, cyano C 1-6 Alkoxy, 3-8 membered cycloalkyl-C 1-6 Alkyl, 3-8 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-7 membered heteroaryl or 5-7 membered heteroaryl-C 1-6 An alkyl group;
each R 7 Are respectively and independently selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
m, n are each independently selected from 1, 2, 3 or 4;
virtual keyIs a chemical bond or is absent, and X is a chemical bond 3 Is C.
In one aspect, the present invention provides a compound of the following formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof,
Wherein,
l is selected from
X、X 1 、X 2 Are each independently selected from-CH (R) 6 )-、-N(R 7 ) -, -O-, -S-; -C (O) -, -S (O) -or-S (O) 2 -;
X 3 Selected from C, CH or N;
X 4 selected from N or C (R) 8 );
R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together form an 8-11 membered fused ring cycloalkyl, 8-11 membered fused ring heterocyclyl, 8-11 membered fused ring aryl or 8-11 membered fused ring heteroaryl optionally substituted with 1-3Q 1's; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
each R 4 Each R 4 ', each R 5 Each R 5 ', each R 6 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1- 6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1- 6 Alkoxy, cyano C 1-6 Alkoxy, 3-8 membered cycloalkyl-C 1-6 Alkyl, 3-8 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-7 membered heteroaryl or 5-7 membered heteroaryl-C 1-6 An alkyl group;
each R 7 Are respectively and independently selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyanoRadical C 1-6 An alkoxy group;
m, n are each independently selected from 1, 2, 3 or 4;
virtual keyIs a chemical bond or is absent, and X is a chemical bond 3 Is C.
In certain embodiments, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together form an 8-11 membered fused ring heterocyclyl or an 8-11 membered fused ring heteroaryl optionally substituted with 1-3Q 1; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together form a 9-10 membered fused ring heterocyclyl or 9-10 membered fused ring heteroaryl optionally substituted with 1-3Q 1; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together form a 9-10 membered fused ring heterocyclyl optionally substituted with 1-3Q 1, a 9-10 membered fused ring heteroaryl group containing at least one nitrogen or one oxygen atom; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
In certain embodiments described above, R 2 、R 3 And (3) andR 2 attached carbon atoms, X 3 Together form the following groups optionally substituted with 1 to 3Q 1:
each X 3 Each X 8 、X 9 Each independently selected from CH or N;
each X 5 Each independently selected from CH, C or N;
each X 6 Each X 7 Are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
Ring A is selected from 5-7 membered cycloalkyl, 5-7 membered heterocyclyl, 5-7 membered aryl or 5-7 membered heteroaryl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
Each X 3 Each X 8 、X 9 Each independently selected from CH or N;
each X 5 Each independently selected from CH, C or N;
each X 6 、X 7 Are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
Ring A is selected from 5-7 membered cycloalkyl, 5-7 membered heterocyclyl, 5-7 membered aryl or 5-7 membered heteroaryl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
p is selected from 0, 1,2 or 3.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
each X 3 Each X 8 、X 9 Each independently selected from CH or N;
each X 5 Each independently selected from CH, C or N;
each X 6 、X 7 Are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
Ring a is selected from pyrrolyl, pyrazolyl, imidazolyl, furanyl, thienyl, oxazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, phenyl, pyridyl, pyrimidinyl, dihydropyrimidinyl, pyrazinyl, pyridazinyl, pyranyl, thiopyranyl, oxazinyl, azepinyl or diazaperyl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
p is selected from 0, 1,2 or 3.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
ring A is selected from 5-7 membered cycloalkyl, 5-7 membered heterocyclyl, 5-7 membered aryl or 5-7 membered heteroaryl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
X 3 selected from CH or N;
X 5 selected from CH, C or N;
p is selected from 0, 1,2 or 3.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
ring a is selected from 5-7 membered aryl or 5-7 membered heteroaryl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
p is selected from 0, 1,2 or 3.
In certain embodiments described above, ring a is selected from pyrrolyl, pyrazolyl, imidazolyl, furanyl, thienyl, oxazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, phenyl, pyridyl, pyrimidinyl, dihydropyrimidinyl, pyrazinyl, pyridazinyl, pyranyl, thiopyranyl, oxazinyl, azepinyl, or diazaperyl.
In certain embodiments described above, ring a is selected from 5-7 membered N-containing heteroaryl groups.
In certain embodiments described above, ring a is selected from pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, pyridyl, pyrimidinyl, dihydropyrimidinyl, pyrazinyl, pyridazinyl, oxazinyl, azepinyl, or diazaperyl.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
p is selected from 0, 1,2 or 3.
In certain embodiments described above, each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered cycloalkyl-C 1-6 Alkyl, 3-6 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-6 membered heteroaryl or 5-6 membered heteroaryl-C 1-6 An alkyl group.
In certain embodiments above, L is selected from
X is selected from-Ch (R) 6 )-、-N(R 7 )-、-O-、-S-;
Each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered cycloalkyl-C 1-6 Alkyl, 3-6 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-6 membered heteroaryl or 5-6 membered heteroaryl-C 1-6 An alkyl group;
each R 6 Each R 7 Are respectively and independently selected from H, C 1-6 Alkyl or halogen radicalsSubstitute C 1-6 An alkyl group;
m, n are each independently selected from 1, 2 or 3.
In certain embodiments above, L is selected from
XX 1 Select X to be from 2 -C is H (R is independently 6 ) -vertical, ground-N selection (R self 7 )--C、H-(OR- 6 、)-、-S--;N(R 7 ) -, -O-, -S-; -C (O) -, -S (O) -or-S (O) 2 -;
X 3 Selected from C, CH or N;
X 4 selected from N or C (R) 8 );
R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, fluoro C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy or fluoro C 1-6 An alkoxy group;
each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered cycloalkyl-C 1-6 Alkyl, 3-6 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-6 membered heteroaryl or 5-6 membered heteroaryl-C 1-6 An alkyl group;
each R 6 Each R 7 Are respectively and independently selected from H, C 1-6 Alkyl or halo C 1-6 An alkyl group;
each R 8 Independently selected from H, halogen, hydroxy, amino, cyano, C 1-6 Alkyl, fluoro C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy or fluoro C 1-6 An alkoxy group;
m, n are each independently selected from 1, 2 or 3;
q is selected from 1 or 2;
virtual keyIs a chemical bond or is absent, and X is a chemical bond 3 Is C.
In certain embodiments above, L is selected from
X is selected from-CH (R) 6 )-、-N(R 7 )-、-O-、-S-;
Each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered cycloalkyl-C 1-6 Alkyl, 3-6 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-6 membered heteroaryl or 5-6 membered heteroaryl-C 1-6 An alkyl group;
each R 6 Each R 7 Are respectively and independently selected from H, C 1-6 Alkyl or halo C 1-6 An alkyl group.
In certain embodiments described above, each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered saturated cycloalkyl-C 1-6 Alkyl, 5-6 membered saturated heterocyclic group-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-6 membered heteroaryl or 5-6 membered heteroaryl-C 1-6 An alkyl group.
In certain embodiments described above, each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered saturated cycloalkyl-methyl, 3-6 membered saturated cycloalkyl-ethyl, 5-6 membered saturated heterocyclyl-methyl, 5-6 membered saturated heterocyclyl-ethyl, phenyl-methyl, phenyl-ethyl, 5-6 membered heteroaryl-methyl or 5-6 membered heteroaryl-ethyl.
In certain embodiments described above, each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
In certain embodiments described above, each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1-trifluoroethyl, hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, hydroxyisopropyl, aminomethyl, 1-aminoethyl, 2-aminoethyl, cyanomethyl, 1-cyanoethyl, 2-cyanoethyl, methoxymethyl, methoxyethyl, ethoxymethyl, ethoxyethyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy or 1, 1-trifluoroethyl.
In certain embodiments described above, X 1 、X 2 Are each independently selected from-CH (R) 6 )-、-N(R 7 ) -, -O-, -S-; -C (O) -, -S (O) -or-S (O) 2 -;
Each R 6 Each R 7 Are respectively and independently selected from H, C 1-6 Alkyl or halo C 1-6 An alkyl group.
In certain embodiments described above, each R 6 Each R 7 Each independently selected from H, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1-trifluoroethyl.
In certain embodiments described above, X 1 、X 2 Are each independently selected from-CH (R) 6 )-、-N(R 7 ) -, -O-, -S-or-C (O) -;
each R 6 Each R 7 Independently selected from H or C 1-6 An alkyl group.
The above-mentionedIn certain embodiments, each R 6 Each R 7 Each independently selected from H, methyl, ethyl, propyl or isopropyl.
In certain embodiments described above, R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, fluoro C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy or fluoro C 1-6 An alkoxy group.
In certain embodiments described above, R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, methyl, ethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, cyanomethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy or trifluoromethoxy.
In certain embodiments described above, X 4 Selected from N or CH.
In certain embodiments described above, each R 8 Independently selected from H, halogen, hydroxy, amino, cyano, C 1-6 Alkyl, fluoro C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy or fluoro C 1-6 An alkoxy group.
In certain embodiments described above, each R 8 Each independently selected from H, halogen, hydroxy, amino, cyano, methyl, ethyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, cyanomethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy or trifluoromethoxy.
In certain embodiments of the above, q is selected from 1 or 2.
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
l isX is selected from-CH 2 -, -NH-; -O-or-S-;
X 1 selected from-N (R) 7 ) -, -O-or-S-;
X 2 selected from-CH (R) 6 ) -or-C (O) -;
X 4 selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
X 3 selected from CH or N;
X 5 selected from CH, C or N;
X 6 、X 7 are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
X 8 、X 9 Are each independently selected from C (R) 9 ) Or N;
ring a is selected from phenyl or a 5-6 membered nitrogen containing heteroaryl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
R 4 、R 4 ’、R 6 independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenSubstitute C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy or cyano C 1-6 An alkoxy group;
R 7 selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
each R 9 Independently selected from hydrogen, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
p is selected from 0, 1 or 2;
q is selected from 1 or 2.
In certain embodiments above, wherein R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
X 3 selected from CH or N; x is X 5 Selected from CH, C or N.
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
L isX is selected from-O-or-S-;
X 1 selected from-N (R) 7 )-;X 2 Selected from-CH (R) 6 ) -or-C (O) -;
X 4 selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
ring A is selected from phenyl or a 5-6 membered heteroaryl group containing 1-3 nitrogens;
q1 is selected from halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
R 4 、R 4 ’、R 6 independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy or cyano C 1-6 An alkoxy group;
R 7 selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
each R 9 Independently selected from hydrogen, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
p is selected from 0 or 1; q is selected from 1 or 2.
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
l isX is selected from-O-or-S-;
X 1 selected from-N (R) 7 )-;X 2 Selected from-CH (R) 6 ) -or-C (O) -;
X 4 selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Alkylthio, halo C 1-4 Alkoxy or halo C 1-4 Alkylthio;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
ring a is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrazolyl, imidazolyl or triazolyl;
q1 is selected from halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
R 4 、R 4 ' are independently selected from H, halogen, hydroxy, amino, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, C 1-4 alkoxy-C 1-4 Alkyl, C 1-4 Alkoxy, halo C 1-4 Alkoxy, hydroxy C 1-4 Alkoxy or amino C 1-4 An alkoxy group;
R 6 is H;
R 7 selected from H, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxyC 1-4 Alkyl or amino C 1-4 An alkyl group;
each R 8 Are each independently selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 alkoxy-C 1-4 Alkyl, C 1-4 Alkoxy, halo C 1-4 Alkoxy, hydroxy C 1-4 Alkoxy, amino C 1-4 Alkoxy, cyano C 1-4 An alkoxy group;
each R 9 Are each independently selected from hydrogen, halogen, hydroxy, amino, cyano, and C 1-4 Alkyl, halogenated C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
p is 1; q is selected from 1 or 2.
In certain embodiments described above, ring a is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, imidazolyl, or triazolyl.
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
l isX is selected from-O-or-S-;
X 1 selected from-N (R) 7 )-;X 2 Selected from-CH (R) 6 ) -or-C (O) -;
X 4 selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Alkylthio, halo C 1-4 Alkoxy or halo C 1-4 Alkylthio;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
q1 is selected from halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
R 4 、R 4 ' are independently selected from H, halogen, hydroxy, amino, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, C 1-4 alkoxy-C 1-4 Alkyl, C 1-4 Alkoxy, halo C 1-4 Alkoxy, hydroxy C 1-4 Alkoxy or amino C 1-4 An alkoxy group;
R 6 is H;
R 7 selected from H, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxy C 1-4 Alkyl or amino C 1-4 An alkyl group;
each R 8 Are each independently selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 alkoxy-C 1-4 Alkyl, C 1-4 Alkoxy, halo C 1-4 Alkoxy, hydroxy C 1-4 Alkoxy, amino C 1-4 Alkoxy, cyano C 1-4 An alkoxy group;
each R 9 Are each independently selected from hydrogen, halogen, hydroxy, amino, cyano, and C 1-4 Alkyl, halogenated C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
p is 1; q is selected from 1 or 2.
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
l isX is selected from-O-or-S-;
X 1 selected from-N (R) 7 )-;X 2 Selected from-CH (R) 6 ) -or-C (O) -;
X 4 Selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, fluoro C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 Alkoxy or fluoro C 1-4 An alkoxy group;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
q1 is selected from halogen, hydroxy, amino, cyano, C 1-4 Alkyl, fluoro C 1-4 Alkyl, C 1-4 Alkoxy or fluoro C 1-4 An alkoxy group;
R 4 、R 4 ' are independently selected from H, halogen, hydroxy, amino, C 1-4 Alkyl, fluoro C 1-4 Alkyl, C 1-4 Alkoxy or fluoro C 1-4 An alkoxy group;
R 6 is H;
R 7 selected from H or C 1-4 An alkyl group;
each R 8 Are each independently selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, fluoro C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 alkoxy-C 1-4 Alkyl, C 1-4 Alkoxy or fluoro C 1-4 An alkoxy group;
each R 9 Are each independently selected from hydrogen, halogen, hydroxy, amino, cyano, and C 1-4 Alkyl, fluoro C 1-4 Alkyl, C 1-4 Alkoxy or fluoro C 1-4 An alkoxy group;
p is 1; q is selected from 1 or 2.
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
l isX is selected from-O-or-S-;
X 1 selected from-N (R) 7 )-;X 2 Selected from-CH (R) 6 ) -or-C (O) -;
X 4 selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, cyano, methyl, ethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, cyanomethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy or trifluoromethoxy;
R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
q1 is selected from halogen, hydroxy, amino, cyano, methyl, ethyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy or trifluoromethoxy;
R 4 、R 4 ' each independently selected from H, halogen, hydroxy, amino, methyl, ethyl, isopropyl, trifluoromethyl, methoxy, ethoxy, or trifluoromethoxy;
R 6 is H;
R 7 selected from H, methyl, ethyl or isopropyl;
each R 8 Each independently selected from H, halogen, hydroxy, amino, cyano, methyl, ethyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy, hydroxymethyl, aminomethyl, cyanomethyl or methoxymethyl;
each R 9 Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, methyl, ethyl, isopropyl, trifluoromethyl, methoxy, ethoxy, or trifluoromethoxy;
p is 1; q is selected from 1 or 2.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
in one aspect, the present invention provides a compound represented by the following general formula (II), a pharmaceutically acceptable salt thereof or an isomer thereof,
Wherein R is 1 、R 2 、R 3 、R 7 、R 8 、L、X 3 、X 4 、Q1、X、R 4 、R 4’ 、R 5 、R 5’ M, n, q, virtual bondIs as defined in any one of the preceding schemes.
In certain embodiments, a compound of formula (II), a pharmaceutically acceptable salt or isomer thereof, wherein,
l isX is selected from-O-or-S-;
X 4 selected from N or CH;
R 1 selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, cyano C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
ring a is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, imidazolyl or 1,2, 4-triazolyl;
q1 is selected from halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
R 4 、R 4 ' are independently selected from H, halogen, hydroxy, amino, C 1-4 Alkyl, halogenated C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl or C 1-4 alkoxy-C 1-4 An alkyl group;
R 7 selected from H or C 1-4 An alkyl group;
R 8 selected from H, halogen, hydroxy, amino, cyano, C 1-4 Alkyl, halogenated C 1-4 Alkyl, C 1-4 Alkoxy or halo C 1-4 An alkoxy group;
each R 9 Are each independently selected from hydrogen, halogen, hydroxy, amino, cyano, and C 1-4 Alkyl or halo C 1-4 An alkyl group; p is 1; q is 1.
In certain embodiments described above, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
in one aspect, the present invention provides a compound represented by the following general formula (III-1) or general formula (III-2), a pharmaceutically acceptable salt thereof or an isomer thereof,
wherein R is 1 、R 2 、R 3 、R 8 、X 3 、X 4 、R 5 、R 5’ Q1, Q, virtual bondIs as defined in any one of the preceding schemes.
In one aspect, the present invention provides a compound represented by the following general formula (IV-1) or general formula (IV-2), a pharmaceutically acceptable salt thereof or an isomer thereof,
wherein R is 1 、R 8 、X 4 The definition of Q1, Q is as described in any of the previous schemes.
In one aspect, the present invention provides a compound represented by the following general formula (IV-3), a pharmaceutically acceptable salt thereof or an isomer thereof,
wherein R is 1 、R 8 、X 4 The definition of Q1, Q is as described in any of the previous schemes.
In certain embodiments, a compound of formula (IV-3), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
X 4 is N;
q1 is selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
q is selected from 1 or 2.
In one aspect, the present invention provides a compound represented by the following general formula (IV-4), a pharmaceutically acceptable salt thereof or an isomer thereof,
wherein R is 1 、R 8 、R 9 、X 4 Q1, p, Q are as defined in any one of the above schemes;
ring a is selected from phenyl, pyridyl, pyrimidinyl or pyrazinyl.
In one aspect, the present invention provides a compound represented by the following general formula (IV-5), a pharmaceutically acceptable salt thereof or an isomer thereof,
wherein R is 1 、R 8 、R 9 、X 4 Q1, p, Q are as defined in any of the above schemes.
In certain embodiments, a compound of formula (IV-5), a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein,
R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
R 9 selected from hydrogen, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkoxy C 1-6 Alkyl, halogenated C 1-6 Alkoxy, 3-6 membered cycloalkyl or 3-6 membered heterocyclyl;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
X 4 is N;
p is selected from 1 or 2;
q is selected from 1 or 2.
The technical schemes in the invention can be mutually combined to form new technical schemes, and the formed new technical schemes are also included in the scope of the invention.
In certain embodiments, the compound of formula (I'), formula (I), formula (II), formula (III-1), formula (III-2), formula (IV-1), formula (IV-2), formula (IV-3), formula (IV-4) or formula (IV-5), a pharmaceutically acceptable salt or isomer thereof, is selected from the compounds shown in Table 1 below:
TABLE 1
In certain embodiments, a compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof,
Wherein L isX is selected from-O-or-S-;
X 1 selected from-N (R) 7 )-;
X 2 is-C (O) -;
X 4 is N;
R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
R 4 、R 4 ' are independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy or cyano C 1-6 An alkoxy group;
R 7 selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
R 9 selected from hydrogen, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkoxy C 1-6 Alkyl, halogenated C 1-6 Alkoxy, 3-6 membered cycloalkyl or 3-6 membered heterocyclyl;
p is selected from 0, 1 or 2;
q is selected from 1 or 2.
The present invention also provides a compound as shown below, a pharmaceutically acceptable salt thereof or an isomer thereof,
in another aspect, the present invention also provides a pharmaceutical composition comprising the compound of formula (I'), formula (I), formula (II), formula (III-1), formula (III-2), formula (IV-1), formula (IV-2), formula (IV-3), formula (IV-4) or formula (IV-5), a pharmaceutically acceptable salt or isomer thereof, and one or more pharmaceutically acceptable excipients, wherein the pharmaceutical composition may be in any pharmaceutically acceptable dosage form. Pharmaceutically acceptable excipients are non-toxic, compatible with the active ingredient and otherwise biologically compatible substances for use in the organism. The choice of a particular excipient will depend on the mode of administration or type and state of disease used to treat a particular patient.
In certain embodiments, the above pharmaceutical compositions may be administered orally, parenterally, rectally, or pulmonary, etc., to a patient or subject in need of such treatment. For oral administration, the pharmaceutical composition may be formulated into oral preparations, for example, into conventional oral solid preparations such as tablets, capsules, pills, granules, etc.; can also be made into oral liquid preparation such as oral solution, oral suspension, syrup, etc. For parenteral administration, the pharmaceutical compositions described above may also be formulated as injections, including injectable solutions, injectable sterile powders, and injectable concentrated solutions. For rectal administration, the pharmaceutical composition may be formulated as suppositories and the like. For pulmonary administration, the pharmaceutical composition may be formulated as an inhalation, aerosol, powder spray or spray.
In a further aspect, the present invention also relates to the use of a compound of the aforementioned general formula (I'), general formula (I), general formula (II), general formula (III-1), general formula (III-2), general formula (IV-1), general formula (IV-2), general formula (IV-3), general formula (IV-4) or general formula (IV-5), a pharmaceutically acceptable salt or an isomer thereof for the manufacture of a medicament for the prevention and/or treatment of a disease associated with excessive activation of PARP 7.
In a further aspect, the invention also relates to the use of a compound of the aforementioned general formula (I'), general formula (I), general formula (II), general formula (III-1), general formula (III-2), general formula (IV-1), general formula (IV-2), general formula (IV-3), general formula (IV-4) or general formula (IV-5), a pharmaceutically acceptable salt or an isomer thereof, for the manufacture of a medicament for the prevention and/or treatment of cancer, including cancer in situ and metastatic cancer.
Furthermore, the invention also relates to the use of a pharmaceutical composition containing the compounds of the general formula (I'), the general formula (I), the general formula (II), the general formula (III-1), the general formula (III-2), the general formula (IV-1), the general formula (IV-2), the general formula (IV-3), the general formula (IV-4) or the general formula (IV-5), pharmaceutically acceptable salts or isomers thereof for preparing a medicament for preventing and/or treating diseases related to the excessive activation of PARP 7.
Furthermore, the invention also relates to the use of the compounds of the general formula (I'), the general formula (I), the general formula (II), the general formula (III-1), the general formula (III-2), the general formula (IV-1), the general formula (IV-2), the general formula (IV-3), the general formula (IV-4) or the general formula (IV-5), pharmaceutically acceptable salts or isomers thereof for preparing medicaments for preventing and/or treating cancers, including in-situ cancers and metastatic cancers.
In another aspect, the pharmaceutical composition of the present invention comprises the compound of formula (I'), formula (I), formula (II), formula (III-1), formula (III-2), formula (IV-1), formula (IV-2), formula (IV-3), formula (IV-4) or formula (IV-5), a pharmaceutically acceptable salt or isomer thereof, and one or more second therapeutically active agents selected from anticancer agents, agents that reduce or reduce one or more side effects of the compound of the present invention when used in treating a disease in a subject, or agents that enhance the efficacy of the compound of the present invention.
In another aspect, the present invention also provides a method of treating a disease associated with excessive activation of PARP7 comprising administering to a patient in need thereof an effective amount of the compound of formula (I'), formula (I), formula (II), formula (III-1), formula (III-2), formula (IV-1), formula (IV-2), formula (IV-3), formula (IV-4) or formula (IV-5), a pharmaceutically acceptable salt or isomer thereof, and a pharmaceutical composition as described above.
Further, the present invention also provides a method of treating cancer, which comprises administering to a patient in need thereof an effective amount of the compound of the aforementioned general formula (I'), general formula (I), general formula (II), general formula (III-1), general formula (III-2), general formula (IV-1), general formula (IV-2), general formula (IV-3), general formula (IV-4) or general formula (IV-5), a pharmaceutically acceptable salt or isomer thereof, and the aforementioned pharmaceutical composition.
In another aspect, the invention also provides a kit comprising an effective amount of one or more of the compounds of formula (I'), formula (I), formula (II), formula (III-1), formula (III-2), formula (IV-1) or formula (IV-2), formula (IV-3), formula (IV-4) or formula (IV-5), pharmaceutically acceptable salts or isomers thereof.
In another aspect, the invention also provides a kit comprising:
(a) An effective amount of one or more of the compounds of the aforementioned formula (I'), formula (I), formula (II), formula (III-1), formula (III-2), formula (IV-1), formula (IV-2), formula (IV-3), formula (IV-4) or formula (IV-5), pharmaceutically acceptable salts or isomers thereof,
and (b) an effective amount of one or more anticancer agents.
The term "anticancer agent" as used herein refers to agents having a therapeutic effect on tumors, including, but not limited to, mitotic inhibitors, alkylating agents, antimetabolites, DNA intercalators, antitumor antibiotics, growth factor inhibitors, signaling inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, hormonal agents, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors, protein prenyl transferase inhibitors, and the like. By "effective amount" is meant an amount of a drug that is capable of preventing, alleviating, delaying, inhibiting or curing a condition in a subject. The size of the dose administered is related to the mode of administration of the drug, the pharmacokinetics of the agent, the severity of the disease, the individual sign (sex, weight, height, age) of the subject, etc.
In the present invention, unless otherwise indicated, scientific and technical terms used herein have the meanings commonly understood by one of ordinary skill in the art, however, for a better understanding of the present invention, the following definitions of some terms are provided. When the definition and interpretation of terms provided by the present invention are not identical to the meanings commonly understood by those skilled in the art, the definition and interpretation of terms provided by the present invention is in control.
"halogen" as used herein refers to a fluorine atom, a chlorine atom, a bromine atom or an iodine atom.
"C" as described in the present invention 1-6 Alkyl "means a straight or branched chain alkyl group having 1 to 6 carbon atoms and includes, for example," C 1-4 Alkyl "," C 1-3 Alkyl "," C 1-2 Alkyl "," C 2-6 Alkyl "," C 2-5 Alkyl "," C 2-4 Alkyl "," C 2-3 Alkyl ", and the like, specific examples include, but are not limited to: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, 2-methylbutyl, neopentyl, 1-ethylpropyl, n-hexyl, isohexyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 1, 2-dimethylpropyl, and the like. "C" as described in the present invention 1-4 Alkyl "means C 1-6 Specific examples of the alkyl group include 1 to 4 carbon atoms.
"C" as described in the present invention 1-6 Alkoxy "means" C 1-6 alkyl-O- ", said" C 1-6 Alkyl "is as defined above. "C" as described in the present invention 1-4 Alkoxy "means" C 1-4 alkyl-O- ", said" C 1-4 Alkyl "is as defined above.
"C" as described in the present invention 1-6 Alkylthio "means" C 1-6 alkyl-S- ", described as" C 1-6 Alkyl "is as defined above. "C" as described in the present invention 1-4 Alkylthio "means" C 1-4 alkyl-S- ", described as" C 1-4 Alkyl "is as defined above.
The invention relates to a hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy C 1-6 Alkyl "means C 1-6 One or more hydrogens in the alkyl group are each replaced with one or more hydroxy groups,Amino, halogen, cyano or C 1-6 Alkoxy groups are substituted. C (C) 1-6 Alkyl, C 1-6 Alkoxy is as defined above.
The invention relates to the hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, halo C 1-6 Alkoxy, cyano C 1-6 Alkoxy "means" C 1-6 One or more hydrogens in the alkoxy "are substituted with one or more hydroxy, amino, halogen, or cyano groups.
The invention relates to the hydroxy C 1-6 Alkylthio, amino C 1-6 Alkylthio, halo C 1-6 Alkylthio "means" C 1-6 One or more hydrogens in the alkylthio group "are substituted with one or more hydroxy, amino, or halogen groups.
The invention relates to a fluoro C 1-6 Alkyl "," fluoro C 1-6 Alkoxy groups "refer to" C ", respectively 1-6 Alkyl "," C 1-6 One or more hydrogens in the alkoxy "are replaced with one or more fluorine atoms.
"3-8 membered cycloalkyl" as used herein refers to a saturated or partially saturated and non-aromatic monocyclic ring group containing 3-8 ring atoms, and "3-8 membered cycloalkyl" as used herein includes "3-8 membered saturated cycloalkyl" and "3-8 membered partially saturated cycloalkyl", such as "3-6 membered cycloalkyl", "3-6 membered saturated cycloalkyl", "5-7 membered saturated cycloalkyl", "5-6 membered saturated cycloalkyl", and the like. Examples include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, and the like.
"3-8 membered heterocyclic group" as used herein refers to a saturated or partially saturated and non-aromatic monocyclic ring group containing at least one (e.g., 1, 2, 3, 4 or 5) heteroatom(s) and having 3-8 ring atoms, the heteroatom being a nitrogen atom, an oxygen atom and/or a sulfur atom, optionally a ring atom in the ring structure (e.g., a carbon atom, a nitrogen atom or a sulfur atom) may be oxo. The "3-8 membered heterocyclic group" as used herein includes "3-8 membered saturated heterocyclic group" and "3-8 membered partially saturated heterocyclic group". The "3-8 membered heterocyclic group" is, for example, "3-6 membered heterocyclic group", "3-6 membered saturated heterocyclic group", "3-7 membered saturated heterocyclic group", "5-7 membered saturated heterocyclic group", "5-6 membered saturated heterocyclic group", etc. Specific examples thereof include, but are not limited to: aziridinyl, 2H-aziridinyl, diazinoalkyl, 3H-diazinopropenyl, azetidinyl, oxetanyl, 1, 4-dioxanyl, 1, 3-dioxanyl, 1, 4-dioxadienyl, tetrahydrofuranyl, dihydropyrrolyl, tetrahydropyrrolyl, tetrahydropyrazolidinyl, tetrahydroimidazolyl, 4, 5-dihydroimidazolyl, pyrazolidinyl, 4, 5-dihydropyrazolyl, 2, 5-dihydrothienyl, tetrahydrothienyl, 4, 5-dihydrothiazolyl, thiazolidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1-dioxotetrahydrothiopyranyl, piperidinyl, tetrahydropyridinyl, piperidonyl, tetrahydropyridonyl, dihydropyridonyl, piperazinyl, hexahydropyrimidinyl, morpholinyl and the like.
The invention relates to a 3-8 membered cycloalkyl-C 1-6 Alkyl "," 3-8 membered heterocyclyl-C 1-6 Alkyl "refers to 3-8 membered cycloalkyl-C, respectively 1-6 Alkyl-, "3-8 membered heterocyclyl-C 1-6 Alkyl- ", wherein" 3-8 membered cycloalkyl "," 3-8 membered heterocyclyl "," C 1-6 Alkyl "is as defined above.
The term "8-11 membered fused ring cycloalkyl" as used herein refers to a saturated or partially saturated, non-aromatic cyclic group containing 8 to 11 ring carbon atoms, formed by two or more cyclic structures sharing two adjacent atoms with each other, wherein one ring of the fused rings may be an aromatic ring, but the fused ring as a whole does not have aromaticity, examples include, but are not limited to:etc.
The term "8-11 membered condensed ring heterocyclic group" as used herein refers to a saturated or partially saturated, non-aromatic cyclic group containing 8-11 ring atoms, at least one of which is a heteroatom, which is formed by sharing two or more adjacent atoms with each other by two or more cyclic structures, wherein one of the condensed rings may be an aromatic ring, but the whole condensed ring does not have aromaticity, and the heteroatom is a nitrogen atom, an oxygen atom and/or a sulfur atom. Wherein the ring atoms (e.g., carbon, nitrogen, or sulfur atoms) in the cyclic structure may optionally be oxo. Specific examples thereof include, but are not limited to: dihydrofuropyridines, 3, 4-dihydro-2H-pyranopyridines, 3, 4-dihydro-2H-oxazinopyridines, dihydrooxazinopyrimidines, benzodihydrofuranyl groups, and the like.
As used herein, "8-11 membered fused ring aryl" refers to an unsaturated, aromatic cyclic group containing 8-11 ring carbon atoms formed by two or more cyclic structures sharing two adjacent atoms with each other, including "9-10 membered fused ring aryl", and specific examples include, but are not limited to, naphthyl.
The term "8-11 membered fused ring heteroaryl group" as used herein refers to an unsaturated aromatic ring structure having 8 to 11 ring atoms (at least one of which is a heteroatom such as a nitrogen atom, an oxygen atom or a sulfur atom) formed by sharing two or more adjacent atoms with each other. Wherein the ring atoms (e.g., carbon, nitrogen, or sulfur atoms) in the cyclic structure may optionally be oxo. Including "9-10 membered fused heteroaryl", "8-9 membered fused heteroaryl", and the like, which may be fused in such a manner that the fused moiety is a 5-membered nitrogen-containing heteroaryl and 5-6 membered nitrogen-containing heteroaryl, a 6-membered nitrogen-containing heteroaryl and 5-6 membered nitrogen-containing heteroaryl, a 5-6 membered aryl and 5-6 heteroaryl, and the like; specific examples include, but are not limited to: pyrrolopyrroles, pyrrolofurans, pyrrolopyrimidines, pyrrolopyrroles, pyrazolopyrimidines, pyrazolothiophenes, pyrazolooxazoles, benzofuranyl, benzisofuranyl, benzothienyl, indolyl, isoindolyl, benzoxazolyl, benzimidazolyl, and the like.
"5-7 membered heteroaryl" as used herein refers to a monocyclic cyclic group having aromaticity which contains 5-7 ring atoms, at least one of which is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom. Wherein the ring atoms (e.g., carbon, nitrogen, or sulfur atoms) in the cyclic structure may optionally be oxo. Including, for example, "5-6 membered heteroaryl", "5-6 membered nitrogen containing heteroaryl", "6 membered nitrogen containing heteroaryl", and the like, wherein the heteroatom in the "nitrogen containing heteroaryl" contains at least one nitrogen atom, e.g., contains only 1 or 2 nitrogen atoms, or contains one nitrogen atom and 1 or 2 other heteroatoms (e.g., oxygen and/or sulfur atoms), or contains 2 nitrogen atoms and 1 or 2 other heteroatoms (e.g., oxygen and/or sulfur atoms). Specific examples of "5-7 membered heteroaryl" include, but are not limited to, furyl, thienyl, pyrrolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, pyridyl, 2-pyridonyl, 4-pyridonyl, pyrimidinyl, pyridazinyl, pyrazinyl, 1,2, 3-triazinyl, 1,3, 5-triazinyl, 1,2,4, 5-tetrazinyl, azepanyl, 1, 3-diazepinyl, and the like.
The invention relates to a C which is of the following structure:
the term "CH" as used herein refers to the following structure:
the invention relates to CR 8 "means the following structure:
the "N" of the invention refers to the following structure:
the invention is described as "each R 4 "means that when m is 2, 3 or 4, a plurality of R 4 Each R of (a) 4 Independently selected from the groups described in the above schemes.
The invention is described as "each R 4 ' means that when m is 2, 3 or 4, a plurality of R 4 ' each R in 4 ' independently selected from the groups described in the above schemes.
The invention is described as "each R 5 "means that when n is 2, 3 or 4, a plurality of R 5 Each R of (a) 5 Independently selected from the groups described in the above schemes.
The invention is described as "each R 5 ' means that when n is 2, 3 or 4, a plurality of R 5 ' each R in 5 ' independently selected from the groups described in the above schemes.
The invention is described as "each R 6 "when X, X 1 And X 2 At the same time be "-CH (R) 6 ) - "when, a plurality of R 6 Each R of (a) 6 Independently selected from the groups described in the above schemes.
The invention is described as "each R 7 "when X, X 1 And X 2 At the same time be "-N (R) 7 ) - "when, a plurality of R 7 Each R of (a) 7 Independently selected from the groups described in the above schemes.
"optionally substituted" as used herein refers to both cases where one or more hydrogen atoms on the substituted group are "substituted" or "unsubstituted" with one or more substituents.
The invention is described inR of (C) 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 In the groups that together form a "the bond between ring A and the adjacent ring may be either a single bond or a double bond, depending mainly on the structure of ring A. For example, the group:if ring A is a saturated heterocycle, the bond between ring A and the adjacent ring is a single bond; if ring A is an aromatic ring, the bond between ring A and the adjacent ring is a single bond or a double bond.
The term "pharmaceutically acceptable salt" as used herein refers to the acidic functional groups present in the compound (e.g., -COOH, -OH, -SO) 3 H, etc.) with suitable inorganic or organic cations (bases), including salts with alkali metals or alkaline earth metals, ammonium salts, salts with nitrogen-containing organic bases; and basic functional groups present in the compounds (e.g. -NH 2 Etc.) with suitable inorganic or organic anions (acids), including salts with inorganic or organic acids (e.g., carboxylic acids, etc.).
"isomers" as used herein means that the compounds of the present invention contain one or more asymmetric centers and are therefore useful as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The compounds of the present invention may have asymmetric centers that each independently produce two optical isomers. The scope of the present invention includes all possible optical isomers and mixtures thereof. The compounds of the present invention, if they contain olefinic double bonds, include cis-isomers and trans-isomers unless specified otherwise. The compounds described herein may exist in tautomeric (one of the functional group isomers) forms having different points of attachment of hydrogen through displacement of one or more double bonds, for example, the keto and enol forms thereof are keto-enol tautomers. The compounds of the invention contain spiro structures, which are affected by the steric structure of the ring, and substituents on the ring may be present on both sides of the ring to form the opposite cis (cis) and trans (trans) isomers. Each tautomer and mixtures thereof are included within the scope of the present invention. Enantiomers, diastereomers, racemates, meso, cis-trans isomers, tautomers, geometric isomers, epimers, mixtures thereof and the like of all compounds are included within the scope of the present invention.
The compounds of the invention may be prepared by enantiospecific synthesis or resolution from mixtures of enantiomers to give the individual enantiomers. Conventional resolution techniques include resolution of mixtures of enantiomers of the starting material or final product using various well-known chromatographic methods.
When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60 wt%, 70 wt%, 80 wt%, 90 wt%, 99 wt%, or 99.9 wt% pure relative to the other stereoisomers. When a single isomer is named or depicted by structure, the depicted or named enantiomer is at least 60% by weight, 70% by weight, 80% by weight, 90% by weight, 99% by weight, or 99.9% by weight pure. Optical purity wt% is the ratio of the weight of an enantiomer to the weight of the enantiomer plus the weight of its optical isomer.
Advantageous effects of the invention
1. The compound, the pharmaceutically acceptable salt or the isomer thereof has excellent PARP7 inhibition effect, good pharmacokinetic property in organisms, lasting effect and high bioavailability.
2. The compound, the pharmaceutically acceptable salt or the isomer thereof has better treatment effect on cancers and high stability of liver microsomes.
3. The compound has the advantages of simple preparation process, high purity of the medicine, stable quality and easy mass industrial production.
Detailed description of the preferred embodiments
The technical scheme of the present invention will be described in detail below with reference to specific embodiments, but the scope of the subject matter of the present invention should not be construed as being limited to the following examples. All techniques implemented based on the above description of the invention are within the scope of the invention.
Abbreviations:
DIPEA: n, N-diisopropylethylamine; DMSO: dimethyl sulfoxide; KOtBu: potassium tert-butoxide; TFA, trifluoroacetic acid; DCM: dichloromethane; HATU: urea N, N, N ', N' -tetramethyl-O- (7-azabenzotriazol-1-yl) hexafluorophosphate; tfOH: trifluoromethanesulfonic acid; DABCO: triethylene diamine; DMF: n, N-dimethylformamide; DMA: n, N-dimethylacetamide; pd (tBu) 3 P) 2 : bis (tri-t-butylphosphine) palladium; TMSCF (TMSCF) 3 (trifluoromethyl) trimethylsilane; phI (OAc) 2 : iodobenzene diacetate; NBS, N-bromosuccinimide; NMP: n-methylpyrrolidone; TBDPSCl: t-butyldiphenylchlorosilane; TBAF: tetrabutylammonium fluoride; msCl: methanesulfonyl chloride.
Preparation example one: preparation of 5- ((((2S) -1- (3-oxo-3- (3- (trifluoromethyl)) -6a,7,9, 10-tetrahydropyrazin [1,2-d ] pyridinyl [3,2-b ] [1,4] oxazin-8 (6H) yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 1)
1. Preparation of tert-butyl 4- (3-fluoro-5- (trifluoromethyl) pyridin-2-yl) -3- (hydroxymethyl) piperazine-1-carboxylate
2, 3-difluoro-5- (trifluoromethyl) pyridine (3.66 g,20.0 mmol), tert-butyl 3- (hydroxymethyl) piperazine-1-carboxylate (4.6 g,21.0 mmol) and DIPEA (4 mL,24.0 mmol) were added to DMSO (40 mL). The reaction was carried out at 80℃for 3.0h under nitrogen protection, cooled to room temperature, the reaction solution was poured into water, ethyl acetate was extracted 3 times, the combined ethyl acetate layers were washed with water, saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (ethyl acetate: petroleum ether=8:1) to give the objective compound (7.4 g, yield: 97.5%).
2. Preparation of tert-butyl 3- (trifluoromethyl) -6a,7,9, 10-tetrahydropyrazino [1,2-d ] pyrido [3,2-b ] [1,4] oxazine-8 (6H) -carboxylate
Tert-butyl 4- (3-fluoro-5- (trifluoromethyl) pyridin-2-yl) -3- (hydroxymethyl) piperazine-1-carboxylate (4.22 g,8.4 mmol) and KOtBu (2.8 g,25.2 mmol) were added to DMSO (50 mL), reacted at 110℃for 10 hours, the reaction solution was poured into water, ethyl acetate was extracted 3 times, the combined ethyl acetate layers were washed with water, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (ethyl acetate: petroleum ether=10:1) to give the title compound (630 mg, yield: 15.8%).
3. Preparation of 3- (trifluoromethyl) -6,6a,7,8,9, 10-hexahydropyrazino [1,2-d ] pyrido [3,2-b ] [1,4] oxazine
TFA (3 mL) was slowly added dropwise to 3- (trifluoromethyl) -6a,7,9, 10-tetrahydropyrazino [1,2-d ] pyrido [3,2-b ] [1,4] oxazine-8 (6H) -carboxylic acid tert-butyl ester (550 mg,1.53 mmol) in dichloromethane (9 mL) at 16 ℃ for 3 hours, and the reaction solution was directly dried under reduced pressure to give crude title compound (900 mg) which was directly used in the next reaction without further purification.
4. Preparation of 2- (4-methoxybenzyl) -5- (((2S) -1- (3-oxo-3- (3- (trifluoromethyl) -6a,7,9, 10-tetrahydropyrazino [1,2-d ] pyrido [3,2-b ] [1,4] oxazin-8 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
(S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (200 mg,0.46 mmol), 3- (trifluoromethyl) -6,6a,7,8,9, 10-hexahydropyrazino [1,2-d ] pyrido [3,2-b ] [1,4] oxazine (182 mg,0.7 mmol), HATU (266 mg,0.7 mmol) and DIPEA (178 mg,1.38 mmol) were added to dichloromethane (4 mL) and the reaction solution was dried under reduced pressure and the residue was purified by silica gel column chromatography (dichloromethane: methanol=30:1) to give the title compound (140 mg, yield: 45.4%).
5. Preparation of 5- ((((2S) -1- (3-oxo-3- (3- (trifluoromethyl)) -6a,7,9,10, tetrahydropyrazin [1,2-d ] pyridinyl [3,2-b ] [1,4] oxazin-8 (6H) yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
Trifluoromethanesulfonic acid (0.1 mL) was slowly added dropwise to TFA (2 mL) containing 2- (4-methoxybenzyl) -5- (((2S) -1- (3-oxo-3- (3- (trifluoromethyl) -6a,7,9, 10-tetrahydropyrazino [1,2-d ] pyrido [3,2-b ] [1,4] oxazin-8 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (90 mg,0.13 mmol) at 18 ℃ and the reaction was continued for 1 hour. After removing most of TFA in the reaction solution under reduced pressure, the remaining reaction solution was diluted with ethyl acetate, the pH was adjusted to 8 by washing with saturated sodium bicarbonate solution, the ethyl acetate layer was washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol=40:1) and medium pressure reverse phase column chromatography (methanol: water=9:1) to give the title compound (25 mg, yield: 33.8%).
Molecular formula C 22 H 24 F 6 N 6 O 4 Molecular weight 550.5 LC-MS (M/e): 551.0 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.47(s,1H),8.31(s,1H),7.91(s,1H),7.55(s,1H),6.27(brs,1H),4.16-4.04(m,2H),3.99-3.88(m,2H),3.65(m,2H),3.52(m,2H),3.48(m,4H),3.33-3.07(m,2H),2.65(m,1H),2.44(m,1H),1.42(m,3H).
Preparation example II: preparation of (S) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -8, 9) dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 4)
1. Preparation of 3-iodo-5- (trifluoromethyl) pyridin-2-amine
5- (trifluoromethyl) pyridin-2-amine (9.6 g,59.2 mmol), silver sulfate (18.5 g,59.2 mmol) and iodine (7.5 g,29.6 mmol) were added to absolute ethanol (50 mL). The reaction is carried out for 8 hours at 25 ℃ under the protection of nitrogen. The reaction solution was filtered, the filtrate was concentrated under reduced pressure, the residue was dissolved in ethyl acetate, then washed with a saturated sodium sulfite solution, separated, the ethyl acetate layer was washed with water, washed with a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=3:1) to give the objective compound (2.8 g, yield: 16.4%).
2. Preparation of 5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid
Into a 100 mL-sealed tube was added 3-iodo-5- (trifluoromethyl) pyridin-2-amine (2.7 g,9.4 mmol), pyruvic acid (2.48 g,28.2 mmol), DABCO (3.16 g,28.2 mmol) and DMF (25 mL), and after 10 minutes at 25℃under nitrogen, pd (OAc) was added 2 (2.74 g,12.2 mmol) was reacted at 110℃for 3h under nitrogen. Removing DMF under reduced pressure, adding ethyl acetate and water, separating, extracting ethyl acetate layer with 2N sodium hydroxide solution twice, regulating pH of the combined water phase to 3 with 12N hydrochloric acid solution, extracting ethyl acetate for 3 times, washing the combined ethyl acetate layer with water, washing with saturated sodium chloride solution, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate under reduced pressure to obtain crude product (0.6 g) of the target compound, and directly using in the next reaction 。
3. Preparation of 5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester
Concentrated sulfuric acid (0.5 mL) was added to absolute ethanol (10 mL) containing 5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid (0.6 g crude), reacted at 85 ℃ for 16 hours, the reaction solution was directly dried under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=3:1) to give the objective compound (316 mg).
4. Preparation of ethyl 1- (cyanomethyl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate
Ethyl 5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (316 mg,1.22 mmol) was added to DMF (15 mL) containing NaH (58 mg,1.46 mmol) at 0deg.C, and after continuing the reaction for 1 hour chloroacetonitrile (110 mg,1.46 mmol) was added, and after reacting at 0deg.C for 10 minutes, the temperature was raised to 60deg.C for 3 hours. The reaction solution was poured into water, extracted with ethyl acetate 3 times, the combined ethyl acetate layers were washed with water, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and dried under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=3:1) to give the title compound (295 mg, yield: 81.3%).
5. Preparation of 3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine
1- (cyanomethyl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] at 0deg.C]Pyridine-2-carboxylic acid ethyl ester (295 mg,0.99 mmol) was added to a solution containing LiAlH 4 (94 mg,2.48 mmol) in dry diethyl ether (40 mL) at 40℃for 8 hours. The reaction solution was dried under reduced pressure, and the residue was subjected to silica gel column chromatography (dichloromethane: methanol=20:1) to give the objective compound (40 mg, yield: 16.7%).
6. Preparation of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
A solution of 3- (trifluoromethyl) -6,7,8,9 tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (35 mg,0.15 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (97 mg,0.23 mmol), HATU (87 mg,0.23 mmol) and DIPEA (58 mg,0.45 mmol) in dichloromethane (10 mL) was reacted at 25℃for 1 h. The reaction solution was dried under reduced pressure, and the residue was purified by reverse phase column (water: methanol=1:1) to give crude title compound (45 mg), which was used directly in the next reaction without further purification.
7. (S) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -8, 9) dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.4 mL) was added dropwise to trifluoroacetic acid (4 mL) containing (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (45 mg crude) at 0 ℃ for 1 hour. The reaction solution was poured into a saturated sodium hydrogencarbonate solution, extracted with methylene chloride 3 times, the methylene chloride layers were combined, the solvent was removed under reduced pressure, and the residue was purified by reverse phase column (70% methanol/water system) and purified by silica gel preparation plate (ethyl acetate as a developing agent) to give the objective compound (25 mg, two-step yield: 32.4%).
Molecular formula C 22 H 22 F 6 N 6 O 3 Molecular weight 532.4 LC-MS (M/e): 532.9 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.43(s,1H),8.52(s,1H),8.32(s,1H),7.87(m,1H),6.50(m,1H),6.23(s,1H),4.99(s,1H),4.90(s,1H),4.27(m,1H),4.19(m,1H),4.09(m,1H),3.97(m,2H),3.69(m,2H),3.47(m,2H),2.71(m,2H),1.11(m,3H).
Preparation example III: preparation of (S) -5- ((1- (3- (5-fluoro-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 26)
1. Preparation of 5-fluoro-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one
To the microwave tube was added 3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one (310 mg,1.21 mmol), select F (425 mg,1.2 mmol), acetonitrile (2.0 mL), and water (0.2 mL). The reaction was carried out at 100℃for 2 hours with microwaves. The reaction solution was concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=1:4) to give the objective compound (35 mg, yield: 10.5%).
2. Preparation of 5-fluoro-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazines
Lithium aluminum hydride (20 mg,0.52 mmol) was added to methyl tert-butyl ether (5 mL) containing 5-fluoro-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one (35 mg,0.13 mmol) at 0deg.C, and reacted at 60deg.C for 4 hours. After the reaction, the reaction was quenched by addition of water (0.5 mL), and the C18 powder was directly stirred and purified by reverse phase column (methanol: water=4:1) to give the objective compound (18 mg, yield: 54.2%).
3. Preparation of (S) -5- ((1- (3- (5-fluoro-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one
5-fluoro-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (18 mg,0.069 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl)) amino) propoxy) propionic acid (46 mg,0.11 mmol), HATU (40 mg,0.11 mmol) and DIPEA (28 mg,0.22 mmol) were added to dichloromethane (5 mL) and reacted at 29℃for 1 hour. C18 powder was directly stirred and purified by reverse phase column (methanol: water=4:1) to give the title compound (28 mg, yield: 60.1%).
4. Preparation of (S) -5- ((1- (3- (5-fluoro-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.2 mL) was added dropwise to trifluoroacetic acid (2 mL) containing (S) -5- ((1- (3- (5-fluoro-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (28 mg,0.042 mmol) at 0 ℃ for 1 hour. The reaction solution was poured into a saturated sodium hydrogencarbonate solution, extracted with methylene chloride 3 times, the methylene chloride layers were combined, the solvent was removed by distillation under the reduced pressure, and the residue was purified by reverse phase column (70% methanol/water system) and silica gel preparation plate (ethyl acetate as a developing agent) to give the title compound (12 mg, yield: 52.2%).
Molecular formula C 22 H 21 F 7 N 6 O 3 Molecular weight 550.4 LC-MS (M/e): 551.0 (M+H) + )
1 H-NMR(400MHz,DMSO-d 6 )δ:12.46(s,1H),8.63(s,1H),8.47(s,1H),7.89(s,1H),6.26(brs,1H),5.04-4.93(m,2H),4.29-4.00(m,5H),3.71(m,2H),3.64(m,2H),2.75(m,2H),1.14(m,3H).
Preparation example four: preparation of (S) -5- ((1- (3- (5-methyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 27)
1. Preparation of 3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid
3-bromo-5- (trifluoromethyl) pyridin-2-amine (1 g,4.2 mmol), butanoic acid (0.43 g,4.2 mmol), anhydrous potassium phosphate (1.8 g,8.4 mmol), magnesium sulfate (1 g,8.4 mmol), glacial acetic acid (0.25 g,4.2 mmol) and bis (tri-tert-butylphosphine) palladium (0.87 g,1.7 mmol) were added to DMA (10 mL) and reacted at 140℃for 5h under nitrogen. The reaction solution was dissolved with ethyl acetate and extracted in separate liquid, the aqueous phase was taken, ph=5-6 was adjusted with dilute hydrochloric acid, ethyl acetate was added, the extraction was performed three times in separate liquid, the organic phase was collected, dried, spin-dried, DCM was added until solid was precipitated, suction filtration was performed, and the filter cake was dried under reduced pressure to give the objective compound (330 mg, yield: 32.4%).
2. Preparation of 3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester
Concentrated sulfuric acid (0.5 mL) was added to absolute ethanol (10 mL) containing 3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid (300 mg,1.2 mmol), reacted at 85 ℃ for 16 hours, the reaction solution was adjusted to ph=7-8 by saturated sodium bicarbonate solution, then ethyl acetate was added and extracted three times with water, the organic phase was dried by spin, and the target compound (320 mg, yield 95.7%) was obtained by silica gel column chromatography (petroleum ether: ethyl acetate=5:1).
3. Preparation of ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate
Ethyl 3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (290 mg,1.1 mmol) was dissolved in DMF (5 mL), potassium tert-butoxide (134 mg,1.2 mmol) was added at 0deg.C for 30 min, and tert-butyl-1, 2, 3-oxathiazole-3-carboxylate 2, 2-dioxide (268 mg,1.2 mmol) was added and reacted at 30deg.C for 4H. After the reaction, ethyl acetate and water were added to conduct extraction, the organic phase was dried by spinning, and the objective compound (152 mg, yield 34.4%) was obtained by forward separation (petroleum ether: ethyl acetate=5:1).
4. Preparation of ethyl 1- (2-aminoethyl) -3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate hydrochloride
Ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (130 mg,0.31 mmol) was dissolved in ethyl acetate (3 mL), and ethyl acetate hydrochloride solution (6 mL) was added to react for 1H at 30 ℃. After the reaction, the mixture was dried by spin-drying to give a crude product (152 mg).
5. Preparation of 5-methyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one
1- (2-aminoethyl) -3-methyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester hydrochloride was dissolved in methanol (5 mL), and potassium carbonate (178 mg,1.3 mmol) was added and reacted at 60℃for 2H. Ethyl acetate and water were added and extracted three times, the organic phase was dried by spin-drying, and the target compound (150 mg) was obtained by forward separation (petroleum ether: ethyl acetate=1:3).
6. Preparation of 5-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine
5-methyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one (140 mg,0.52 mmol) was dissolved in methyl tert-butyl ether (6 mL), lithium aluminum hydride (119 mg,3.1 mmol) was added and reacted at 30℃for 1H. Quenched with water, dried by spin-drying, and isolated by reverse C18 (water: methanol=1:2) to give the title compound (20 mg, 14.6% yield).
7. Preparation of (S) -2- (4-methoxybenzyl) -5- ((1- (3- (5-methyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
Containing 5-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (15 mg,0.06 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (26 mg,0.06 mmol), HATU (34 mg,0.09 mmol) and DIPEA (23 mg,0.18 mmol) in dichloromethane (5 mL) were reacted at 25℃for 1h. The reaction solution was dried under reduced pressure, and the residue was purified by normal phase column (dichloromethane: methanol=20:1) to give a product (20 mg, yield 52.9%).
8. Preparation of (S) -5- ((1- (3- (5-methyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.5 mL) was added dropwise to trifluoroacetic acid (3 mL) containing (S) -2- (4-methoxybenzyl) -5- ((1- (3- (5-methyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (15 mg,0.023 mmol) at 0 ℃ for 1 hour. The reaction solution was poured into saturated sodium hydrogencarbonate solution, extracted with ethyl acetate 3 times, the solvent was removed under reduced pressure, and the residue was purified by reverse phase column (water: methanol=1:2) to give the objective compound (3 mg, yield 24.6%).
Molecular formula C 23 H 24 F 6 N 6 O 3 Molecular weight 546.5 LC-MS (M/e): 547.2 (M+H) + )
1 H-NMR(400MHz,CDCl 3 )δ:10.7(s,1H),8.5(s,1H),8.0(s,1H),7.6(s,1H),5.79(s, 1H),4.8-5.0(d,2H),4.3-4.5(d,2H),4.1(s,1H),3.8-4.0(d,4H),3.65(s,1H),3.5(s,1H),2.7(s,2H),2.3(s,3H),1.2(s,3H).
Preparation example five: preparation of 5- (((2S) -1- (3-oxo-3- (3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 28)
1. Preparation of 3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine-7 (6H) -carboxylic acid tert-butyl ester
DMAP (122 mg,1.0 mmol) was added to a mixture containing 3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5]]Pyrrolo [1,2-a ]]Pyrazine (125 mg,0.52 mmol) and (Boc) 2 O (227 mg,1.0 mmol) in dichloromethane (14 mL) was reacted at 30℃for 20 hours. After the completion of the reaction, the reaction mixture was directly dried under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=3:1) to give the objective compound (110 mg, yield: 62.1%).
2. Preparation of tert-butyl 3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine-7 (5H) -carboxylate
Sodium cyanoborohydride (90 mg,1.4 mmol) was added in portions to acetic acid (10 mL) containing 3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine-7 (6H) -carboxylic acid tert-butyl ester (100 mg,0.29 mmol) and reacted at 30℃for 40 hours. After the completion of the reaction, acetic acid was removed under reduced pressure, the residue was dissolved in ethyl acetate, the pH was adjusted to be alkaline with saturated sodium hydrogencarbonate solution, the ethyl acetate layer was directly dried under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=2:1) to give the objective compound (80 mg, yield: 79.5%).
3. Preparation of 3- (trifluoromethyl) -5,5a,6,7,8, 9-hexahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine hydrochloride
Ethyl acetate solution (4 mL) containing hydrochloric acid was added to ethyl acetate (4 mL) containing tert-butyl 3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine-7 (5H) -carboxylate (80 mg,0.23 mmol) at 30℃for 3H. After the reaction is finished, the reaction solution is directly decompressed and evaporated to dryness to obtain a crude product, which is directly used for the next reaction.
4. Preparation of 2- (4-methoxybenzyl) -5- (((2S) -1- (3-oxo-3- (3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
3- (trifluoromethyl) -5,5a,6,7,8, 9-hexahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine hydrochloride (crude from above), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propionic acid (148 mg,1.14 mmol), HATU (433 mg,1.14 mmol) and DIPEA (500 mg,3.87 mmol) were added to dichloromethane (10 mL) and reacted at 28℃for 1 hour. After the completion of the reaction, the target compound (45 mg, yield: 29.5%) was obtained by purification on a silica gel preparation plate (pure ethyl acetate as a developing solvent).
5. Preparation of 5- (((2S) -1- (3-oxo-3- (3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.2 mL) was added dropwise to trifluoroacetic acid (2 mL) containing 2- (4-methoxybenzyl) -5- (((2S) -1- (3-oxo-3- (3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (45 mg,0.069 mmol) at 0 ℃ for 1 hour. The reaction solution was poured into a saturated sodium hydrogencarbonate solution, extracted with methylene chloride 3 times, the methylene chloride layers were combined, the solvent was removed under reduced pressure, and the residue was purified by large-plate silica gel (ethyl acetate as a developing agent) and high-pressure HPLC (90% methanol/water system) to give the title compound (8 mg, yield: 21.8%).
Molecular formula C 22 H 24 F 6 N 6 O 3 Molecular weight 534.5 LC-MS (M/e): 535.2 (M+H) + )
1 H-NMR(400MHz,DMSO-d 6 )δ:12.44(brs,1H),8.11(s,1H),7.90(s,1H),7.52(s,1H),6.24(s,1H),4.51-4.42(m,1H),4.15(m,1H),4.11-3.98(m,1H),3.92-3.85(m,1H),3.71(m,2H),3.51(m,2H),3.19-3.11(m,1H),2.99-2.85(m,2H),2.62-2.52(m,4H),2.01(m,1H),1.16-1.14(m,3H).
Preparation example six: preparation of (S) -5- (methyl (1- (3-oxo-3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin) -7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (preparation of Compound 29)
1、(S)-3-(2-(λ 2 -aza) propoxy) benzyl propionate hydrochloride
To benzyl (S) -3- (2- ((tert-butoxycarbonyl) amino) propoxy) propionate (3.1 g,9.2 mmol) was added ethyl acetate hydrochloride solution (30 mL) at 25℃and the reaction was continued for 1 hour. The reaction solution was dried under reduced pressure to give a crude product (3.1 g) of the objective compound, which was directly used in the next reaction.
2. Preparation of benzyl (S) -3- (2- (methylamino) propoxy) propionate
At 25℃in the presence of (S) -3- (2- (lambda) 2 To a solution of benzyl azapropoxy) propionate hydrochloride (3.1 g crude, 9.2 mmol) in methanol (32 mL) was added aqueous formaldehyde (36%) (389 mg,4.7 mmol) and the reaction continued for 30 min. The reaction solution was dried under reduced pressureMethanol (15 mL) and sodium cyanoborohydride (1.7 g,27.6 mmol) were added and the reaction was continued for 30 min, the system was quenched with water, the organic phase was extracted with dichloromethane, dried over anhydrous sodium sulfate to give crude title compound (2.0 g) which was used directly in the next reaction.
3. Preparation of benzyl (S) -3- (2- ((tert-butoxycarbonyl) (methyl) amino) propoxy) propionate
To a solution of benzyl (S) -3- (2- (methylamino) propoxy) propionate (2.0 g crude, 9.2 mmol) in methylene chloride (50 mL) at 25℃were added di-tert-butyl dicarbonate (3.0 g,13.7 mmol) and N, N-diisopropylethylamine (2.4 g,18.6 mmol) and the reaction was continued for 16 hours. The reaction solution was dried under reduced pressure, the system was quenched with water, the organic phase was extracted with dichloromethane, dried over anhydrous sodium sulfate, the organic phase was dried under reduced pressure, the residue was purified by normal phase column (ethyl acetate: petroleum ether=1:5), the organic phase was dried under reduced pressure, and the residue was further purified by high pressure reverse phase column (acetonitrile: water=3:5) to give the objective compound (160 mg, three-step yield 4.7%).
4. Preparation of benzyl (S) -3- (2- (methylamino) propoxy) propionate hydrochloride
To benzyl (S) -3- (2- ((tert-butoxycarbonyl) (methyl) amino) propoxy) propionate (150 mg,0.43 mmol) was added ethyl acetate solution containing hydrochloric acid (10 mL) at 25℃and the reaction was continued for 1 hour. The reaction solution was dried under reduced pressure and directly used for the next reaction.
5. Preparation of benzyl (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) (methyl) amino) propoxy) propanoate
To an acetonitrile solution (10 mL) containing benzyl (S) -3- (2- (methylamino) propoxy) propionate hydrochloride (crude, 0.43 mmol) at 25℃were added 5-chloro-2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (137 mg,0.43 mmol) and N, N-diisopropylethylamine (278 mg,2.2 mmol), and the mixture was reacted at 25℃for 16 hours. The system was quenched with water, the organic phase was extracted with ethyl acetate, dried over anhydrous sodium sulfate, the organic phase was dried under reduced pressure, and the residue was purified by normal phase column (ethyl acetate: petroleum ether=2:3), and the organic phase was concentrated and dried to give the objective compound (94 mg, yield 41.3%).
6. Preparation of (S) -3- (2- ((1- (4-methoxybenzyl) -5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) (methyl) amino) propoxy) propanoic acid
To a methanol solution (5 mL) of benzyl (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) (methyl) amino) propoxy) propionate (94 mg,0.18 mmol) at 25℃was added palladium on carbon (30 mg), and the mixture was reacted under a hydrogen atmosphere for 1 hour. The filtrate was filtered, concentrated and spun dry and used directly in the next step.
7. Preparation of (S) -2- (4-methoxybenzyl) -5- (methyl (1- (3-oxo-3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of (S) -3- (2- ((1- (4-methoxybenzyl) -5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) (methyl) amino) propoxy) propanoic acid (crude, 0.18 mmol) in dichloromethane (5 mL) was added 3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (53 mg,0.22 mmol), HATU (103 mg,0.27 mmol) and DIEA (70 mg,0.54 mmol) and the mixture was reacted at 25℃for 1 hour. The system was quenched with water, the organic phase was extracted with dichloromethane, dried over anhydrous sodium sulfate, and the organic phase was dried under reduced pressure, and the residue was purified by normal phase column (ethyl acetate: petroleum ether=2:1) to give the objective compound (100 mg, yield 85.2%).
8. Preparation of (S) -5- (methyl (1- (3-oxo-3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin) -7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To (S) -2- (4-methoxybenzyl) -5- (methyl (1- (3-oxo-3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (20 mg,0.030 mmol) in trifluoroacetic acid (2 mL) was added trifluoromethanesulfonic acid (0.2 mL) at 25℃and the reaction was continued for 15 minutes. The system was quenched with saturated sodium bicarbonate solution, extracted with a mixed solvent of dichloromethane/methanol (10:1), the organic phase was dried over anhydrous sodium sulfate, the organic phase was dried under reduced pressure, and the residue was purified by high pressure reverse phase column (acetonitrile/water=3:2) to give the title compound (3.9 mg, yield 23.8%).
Molecular formula C 23 H 24 F 6 N 6 O 3 Molecular weight 546.5 LC-MS (M/e): 547.2 (M+H) + )
1 H-NMR(400MHz,CDCl 3 )δ:10.35-10.15(m,1H),8.55-8.52(m,1H),8.11(s,1H),7.76(m,1H),6.39(s,1H),5.02(s,1H),4.90(s,1H),4.38-4.25(m,2H),4.10-3.90(m,1H),3.85-3.75(m,2H),3.65-3.40(m,4H),2.91(s,3H),3.65-3.55(m,2H),1.40-1.20(m,3H).
Preparation example seven: preparation of (S) -4- (trifluoromethyl) -5- ((1- (3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ]) pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) pyridazin-3 (2H) -one (Compound 30)
1. Preparation of (S) -5- ((1- (3-hydroxypropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one
Benzyl (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propionate (450 mg,0.87 mmol) was dissolved in ethanol (10 mL), sodium borohydride (131 mg,3.5 mmol), calcium chloride (284 mg,3.5 mmol) was added and reacted at 25℃for 4h. The reaction solution was dried under reduced pressure, and the residue was purified by column chromatography (PE/ea=0 to 70%) to give the objective compound (320 mg, yield 88.9%).
2. (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanal-dehyde
(S) -5- ((1- (3-hydroxypropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (160 mg,0.38 mmol) was dissolved in dichloromethane (10 mL), DMP (245 mg,0.58 mmol) was added and reacted at 25℃for 4H. The reaction solution was dried under reduced pressure, and the residue was purified by column chromatography (PE/ea=0 to 70%) to give the objective compound (140 mg, yield 87.9%).
3. (S) -2- (4-methoxybenzyl) -4- (trifluoromethyl) -5- ((1- (3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) pyridazin-3 (2H) -one
(S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanal (140 mg,0.34 mmol) was dissolved in dichloromethane (5 mL) and 3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (82 mg,0.34 mmol), sodium triacetoxyborohydride (143 mg,0.68 mmol), acetic acid (42 mg,0.68 mmol) was added for 1h at 25 ℃. The reaction solution was washed with water, and dichloromethane was extracted to give the objective compound (120 mg), which was used directly in the next step.
4. (S) -4- (trifluoromethyl) -5- ((1- (3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ]) pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) pyridazin-3 (2H) -one
To trifluoroacetic acid (5 mL) of (S) -2- (4-methoxybenzyl) -4- (trifluoromethyl) -5- ((1- (3- (3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5 ]) pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) pyridazin-3 (2H) -one (110 mg,0.17 mmol) was added TfOH (0.5 mL) at 25℃and the reaction was carried out for 15 minutes. The reaction solution was poured into saturated sodium hydrogencarbonate solution, extracted with dichloromethane 3 times, the dichloromethane layers were combined, the solvent was removed under reduced pressure, and the residue was purified by reverse phase column (50% acetonitrile/water system) to give the objective compound (7 mg, yield 6.8%).
Molecular formula C 22 H 24 F 6 N 6 O 2 Molecular weight 518.5 LC-MS (M/e): 519.2 (M+H) + )
1 H-NMR(400MHz,DMSO-d 6 )δ:12.45(s,1H),8.49(s,1H),8.29(s,1H),7.94(s,1H),6.39(s,1H),6.27-6.30(m,1H),4.14-4.17(m,3H),3.77(s,2H),3.54-3.48(m,4H),3.48-3.41(m,2H)2.90-2.87(m,2H),1.70-1.74(m,2H),1.11-1.22(m,3H).
Preparation example eight: preparation of (S) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrazino [1,2-a ] indol-2 (1H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 31)
1. Preparation of 5- (trifluoromethyl) -1H-indole-2-carboxylic acid ethyl ester
Concentrated sulfuric acid (0.5 mL) was added to absolute ethanol (15 mL) containing 5- (trifluoromethyl) -1H-indole-2-carboxylic acid (500 mg,2.2 mmol), reacted at 85 ℃ for 16 hours, the reaction solution was adjusted to ph=7-8 by saturated sodium bicarbonate solution, extracted three more times with ethyl acetate and aqueous solution, the organic phase was dried by spin, and the target compound (540 mg, yield 96.2%) was obtained by silica gel column chromatography (petroleum ether: ethyl acetate=6:1).
2. Preparation of ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -5- (trifluoromethyl) -1H-indole-2-carboxylate
Ethyl 5- (trifluoromethyl) -1H-indole-2-carboxylate (524 mg,2.0 mmol) was dissolved in DMF (15 mL), potassium tert-butoxide (336 g,3.0 mmol) was added at zero to react for 30 min, and tert-butyl-1, 2, 3-oxathiazole-3-carboxylate 2, 2-dioxide (580 mg,2.6 mmol) was added and reacted at 25℃for 1H. After the reaction, ethyl acetate and water were added to extract, the organic phase was dried by spinning, and the product (780 mg, yield 95.6%) was obtained by forward separation (PE: ea=3:1).
3. Preparation of ethyl 1- (2-aminoethyl) -5- (trifluoromethyl) -1H-indole-2-carboxylate hydrochloride
1- (2- ((tert-Butoxycarbonyl) amino) ethyl) -5- (trifluoromethyl) -1H-indole-2-carboxylic acid ethyl ester (780 mg,1.9 mmol) was dissolved in EA (6 mL), and ethyl acetate hydrochloride solution (30 mL) was added and reacted at 25℃for 1H. And spin-drying after the reaction is finished to obtain the product which is directly used for the next reaction.
4. Preparation of 8- (trifluoromethyl) -3, 4-dihydropyrazino [1,2-a ] indol-1 (2H) -one
1- (2-aminoethyl) -5- (trifluoromethyl) -1H-indole-2-carboxylic acid ethyl ester hydrochloride was dissolved in methanol (20 mL), and potassium carbonate (787 mg,5.7 mmol) was added and reacted at 50℃for 2H. EA and water were added to the mixture to extract three times, and the organic phase was dried by spin-drying, and the desired compound (470 mg, two-step yield: 94.9%) was obtained by forward separation (ea=100%).
5. Preparation of 8- (trifluoromethyl) -1,2,3, 4-tetrahydropyrazino [1,2-a ] indole
8- (trifluoromethyl) -3, 4-dihydropyrazino [1,2-a ] indol-1 (2H) -one (200 mg,0.79 mmol) is dissolved in methyl tert-butyl ether (10 mL), lithium aluminum hydride (180 mg,4.7 mmol) is added and reacted at 60℃for 4H. Quenched with water, dried by spin-drying, and isolated by reverse C18 (water: methanol=1:4) to give the title compound (140 mg, yield 74.1%).
6. Preparation of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrazino [1,2-a ] indol-2 (1H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
8- (trifluoromethyl) -1,2,3, 4-tetrahydropyrazino [1,2-a ] indole (73 mg,0.30 mmol), (S) -3- (2- (((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (142 mg,0.33 mmol), HATU (171 mg,0.45 mmol) and DIPEA (116 mg,0.90 mmol) were added to dichloromethane (10 mL), reacted at 25℃for 30 min, the system quenched with water, extracted with ethyl acetate, the organic phases combined, dried over anhydrous sodium sulfate, and the residue was purified forward (ethyl acetate: petroleum ether=7:3) to give the title compound (130 mg, 65.6% yield).
7. Preparation of (S) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrazino [1,2-a ] indol-2 (1H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To trifluoroacetic acid (2 mL) of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrazino [1,2-a ] indol-2 (1H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (130 mg,0.20 mmol) at 25 ℃ was added dropwise TfOH (0.5 mL) and the mixture was reacted at 25 ℃ for 15 minutes. The reaction solution was poured into saturated sodium bicarbonate solution, extracted with dichloromethane 3 times, the dichloromethane layers were combined, the solvent was removed under reduced pressure, and the residue was purified by normal phase column (ethyl acetate: petroleum ether=9:13) to give the objective compound (11.2 mg, yield 10.6%).
Molecular formula C 23 H 23 F 6 N 5 O 3 Molecular weight 531.5 LC-MS (M/e): 532.2 (M+H) + )
1 H-NMR(400MHz,DMSO-d 6 )δ:12.48(s,1H),7.91-7.88(m,2H),7.60(d,J=8.0Hz,1H),7.40(d,J=8.0Hz,1H),6.49(s,1H),6.35-6.26(m,1H),4.96-4.88(m,2H),4.30-3.90(m,5H),3.80-3.65(m,2H),3.55-3.40(m,2H),2.80-2.5(m,2H),1.20-1.05(m,3H).
Preparation example nine: preparation of (S) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 32) preparation of 1, 4-methyl-5-nitro-2- (trifluoromethyl) pyridine
2-bromo-4-methyl-5-nitropyridine (10 g,46.1 mmol), methyl 2, 2-difluoro-2- (fluorosulfonyl) acetate (13.3 g,69.2 mmol) and cuprous iodide (8.8 g,46.2 mmol) were added to N, N-dimethylformamide (100 mL). The reaction is carried out for 16h at 115 ℃ under the protection of nitrogen. The system was quenched by adding aqueous ammonium chloride, adding aqueous ammonia, extracting with ethyl acetate, drying the organic phase over anhydrous sodium sulfate, filtering, concentrating the filtrate under reduced pressure, and subjecting the residue to silica gel column chromatography (petroleum ether: ethyl acetate=24:1) to give the objective compound (5.2 g, yield: 54.8%).
2. Preparation of 5- (trifluoromethyl) -1H-pyrrolo [2,3-c ] pyridine-2-carboxylic acid ethyl ester
Diethyl oxalate (8.3 g,56.8 mmol) and 1, 8-diazabicyclo [5.4.0] undec-7-ene (4.4 g,28.9 mmol) were added to 4-methyl-5-nitro-2- (trifluoromethyl) pyridine (2.5 g,12.1 mmol), the mixture was stirred at 25℃for 4 hours, acetic acid (50 mL) was added after concentrating and spin-drying, iron powder (1.4 g,25.1 mmol) was added to the mixture, and the mixture was heated to 70℃for 16 hours. The system was quenched with water, filtered and the filter cake was washed three times with water. Then the filter cake is dissolved in ethyl acetate, suction filtration is carried out, the filtrate is concentrated and dried to obtain crude product (2.9 g) of the target compound, which is directly used for the next reaction.
3. Preparation of ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-c ] pyridine-2-carboxylate
Ethyl 5- (trifluoromethyl) -1H-pyrrolo [2,3-c ] pyridine-2-carboxylate (2.9 g,11.2 mmol) was dissolved in DMF (80 mL), potassium tert-butoxide (1.9 g,17.0 mmol) was added at zero, reacted for 30 min, and tert-butyl-1, 2, 3-oxathiazole-3-carboxylate 2, 2-dioxide (3.2 g,14.3 mmol) was added and reacted for 1H at 25 ℃. After the reaction, ethyl acetate and water were added for extraction, the organic phase was dried by spinning, and the desired compound (2.9 g, two-step yield 59.6%) was obtained by separation with a normal phase column (petroleum ether: ethyl acetate=3:1).
4. Preparation of ethyl 1- (2-aminoethyl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-c ] pyridine-2-carboxylate hydrochloride
Ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-c ] pyridine-2-carboxylate (1.0 g,2.5 mmol) was dissolved in ethyl acetate (5 mL), and ethyl acetate hydrochloride solution (20 mL) was added and reacted at 25 ℃ for 1H. After the reaction is finished, spin-drying is carried out, and the obtained product is directly used for the next reaction.
5. Preparation of 8- (trifluoromethyl) -3, 4-dihydropyrido [4',3':4,5] pyrrolo [1,2-a ] pyrazin-1 (2H) -one
Ethyl 1- (2-aminoethyl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-c ] pyridine-2-carboxylate hydrochloride was dissolved in methanol (20 mL), and potassium carbonate (1.0 g,7.2 mmol) was added and reacted at 25℃for 16H. Ethyl acetate and water were added, the mixture was extracted three times with liquid separation, and the organic phase was dried by spin-drying and separated by normal phase column (dichloromethane: methanol=10:1) to give the objective compound (590 mg, two-step yield: 92.8%).
6. Preparation of 8- (trifluoromethyl) -1,2,3, 4-tetrahydropyrido [4',3':4,5] pyrrolo [1,2-a ] pyrazine
8- (trifluoromethyl) -3, 4-dihydropyrido [4',3':4,5] pyrrolo [1,2-a ] pyrazin-1 (2H) -one (100 mg,0.39 mmol) was dissolved in methyl tert-butyl ether (10 mL), lithium aluminum hydride (89 mg,2.3 mmol) was added and reacted at 25℃for 4H. Water quenching, spin drying and isolation via reverse phase C18 (water: methanol=1:1) afforded the title compound (30 mg, 31.7% yield).
7. Preparation of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrido [4',3':4,5] pyrrolo [1,2-a ] pyrazin-2 (1H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
8- (trifluoromethyl) -1,2,3, 4-tetrahydropyrido [4',3':4,5] pyrrolo [1,2-a ] pyrazine (30 mg,0.12 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (52 mg,0.12 mmol), HATU (68 mg,0.18 mmol) and DIPEA (47 mg,0.36 mmol) were added to dichloromethane (10 mL) and reacted at 25℃for 30 min. The reaction solution was dried under reduced pressure, and the residue was purified by TLC (ethyl acetate=100%) to give the objective compound (30 mg, yield 38.6%).
8. (S) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of ((S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (8- (trifluoromethyl) -3, 4-dihydropyrido [4',3':4,5] pyrrolo [1,2-a ] pyrazin-2 (1H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (30 mg,0.046 mmol) in trifluoroacetic acid (2 mL) at 25 ℃ was added dropwise TfOH (0.2 mL), reacted for 15 min at 25 ℃, the reaction solution was poured into saturated sodium bicarbonate solution, dichloromethane extracted 3 times, the dichloromethane layers were combined, the solvent was removed under reduced pressure, and the residue was purified via a plate preparation (ethyl acetate as a developing solvent) and a reverse phase column (50% acetonitrile/water system) to give the title compound (4.4 mg, yield 18.0%).
Molecular formula C 22 H 22 F 6 N 6 O 3 Molecular weight 532.4 LC-MS (M/e): 532.9 (M+H) + )
1 H-NMR(400MHz,CD 3 OD)δ:8.82-8.80(m,1H),7.99-7.87(m,2H),6.60(s,1H),5.11(s,1H),5.06-4.95(m,2H),4.44-4.37(m,3H),4.20-4.05(m,3H),3.86-3.82(m,2H),3.70-3.50(m,2H),2.84-2.81(m,2H),1.31-1.16(m,3H).
Preparation example ten: preparation of (S) -5- ((1- (3-oxo-3- (2- (trifluoromethyl) -6, 7-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-8 (9H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 33)
1. Preparation of 2-iodo-6- (trifluoromethyl) pyridin-3-amine
6- (trifluoromethyl) pyridin-3-amine (9.0 g,55.5 mmol), ag 2 SO 4 (17.3 g,55.5 mmol) and iodine (28.2 g,111.1 mmol) were added to ethanol (100 mL). The reaction was carried out at 25℃for 2 hours, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=4:1) to give the objective compound (13.0 g, yield: 81.3%).
2. Preparation of 5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylic acid
2-iodo-6- (trifluoromethyl) pyridin-3-amine (6.0 g,20.8 mmol) was dissolved in DMF (100 mL), palladium acetate (2.4 g,10.4 mmol), pyruvic acid (5.5 g,62.5 mmol) and triethylenediamine (7.0 g,62.5 mmol) were added, the mixture was stirred at 130℃for 4 hours, cooled, the reaction solution quenched with 1M NaOH aq. Ethyl acetate, the organic phase was discarded, the aqueous phase was pH adjusted to 1 with 1M HCl aq, ethyl acetate extracted, the organic phase dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure to give crude title compound (3.2 g) which was used directly in the next step.
3. Preparation of 5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylic acid ethyl ester
5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylic acid (3.0 g,13.0 mmol) was dissolved in EtOH (20 mL), concentrated sulfuric acid (1 mL) was added, and after the reaction was completed, ethyl acetate was extracted, concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=4:1) to give the objective compound (2.1 g, yield: 62.4%)
4. Preparation of ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylate
Ethyl 5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylate (2.4 g,9.3 mmol) was dissolved in DMF (20 mL), potassium tert-butoxide (1.6 g,13.9 mmol) was added at zero to react for 30 min, and tert-butyl-1, 2, 3-oxathiazole-3-carboxylate 2, 2-dioxide (2.5 g,11.1 mmol) was added and reacted at 25℃for 1H. After the reaction, EA and water were added to separate the organic phase, which was dried by spin-drying and separated by a normal phase column (PE: ea=3:1) to give the objective compound (2.4 g, yield 71.3%).
5. Preparation of ethyl 1- (2-aminoethyl) -5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylate hydrochloride
Ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylate (2.2 g,5.5 mmol) was dissolved in EA (5 mL), and ethyl acetate hydrochloride solution (20 mL) was added and reacted at 25 ℃ for 1H. And after the reaction is finished, spin-drying to obtain a crude product of the target compound, which is directly used for the next reaction.
Preparation of 2- (trifluoromethyl) -7, 8-dihydropyrido [2',3':4,5] pyrrolo [1,2-a ] pyrazin-9 (6H) -one
1- (2-aminoethyl) -5- (trifluoromethyl) -1H-pyrrolo [3,2-b ] pyridine-2-carboxylic acid ethyl ester hydrochloride (1.2 g,3.6 mmol) was dissolved in methanol (20 mL), and potassium carbonate (1.4 g,10.7 mmol) was added and reacted at 25℃for 2H. EA was added and extracted three times with water, the organic phase was dried by spin, and the desired compound (240 mg, two-step yield: 26.4%) was obtained by separation on a normal phase column (PE: ea=0-70%).
7. Preparation of 2- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [2',3':4,5] pyrrolo [1,2-a ] pyrazine
2- (trifluoromethyl) -7, 8-dihydropyrido [2',3':4,5] pyrrolo [1,2-a ] pyrazin-9 (6H) -one (104 mg,0.41 mmol) was dissolved in methyl tert-butyl ether (10 mL), lithium aluminum hydride (77 mg,2.0 mmol) was added and reacted at 25℃for 6H. Quenched with water, dried by spin-drying, and separated by reverse phase C18 (water: methanol=0-70%) to give the title compound (42 mg, yield 42.7%).
8. Preparation of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (2- (trifluoromethyl) -6, 7-dihydropyrido [2',3':4,5] pyrrolo [1,2-a ] pyrazin-8 (9H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
2- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [2',3':4,5] pyrrolo [1,2-a ] pyrazine (30 mg,0.12 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (57 mg,0.13 mmol), HATU (57 mg,0.15 mmol) and DIPEA (48 mg,0.37 mmol) were added to dichloromethane (10 mL) and reacted at 25℃for 30 min. The reaction solution was dried under reduced pressure, and the residue was purified by TLC (ethyl acetate=100%) to give the objective compound (40 mg, yield 49.3%).
9. (S) -5- ((1- (3-oxo-3- (2- (trifluoromethyl) -6, 7-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-8 (9H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
(S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (2- (trifluoromethyl) -6, 7-dihydropyrido [2',3':4,5] pyrrol [1,2-a ] pyrazin-8 (9H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (30 mg,0.046 mmol) was dissolved in trifluoroacetic acid (3 mL), tfOH (0.3 mL) was added and the mixture was reacted at 25℃for 15 minutes. The reaction solution was poured into saturated sodium hydrogencarbonate solution, extracted with dichloromethane 3 times, the dichloromethane layers were combined, the solvent was removed under reduced pressure, and the residue was purified by reverse phase column (50% acetonitrile/water system) to give the objective compound (22 mg, yield 89.9%).
Molecular formula C 22 H 22 F 6 N 6 O 3 Molecular weight 532.4 LC-MS (M/e): 532.9 (M+H) + )
1 H-NMR(400MHz,CD 3 OD)δ:8.82-8.80(m,1H),7.99-7.87(m,2H),6.60(s,1H),5.11(s,1H),5.06-4.95(m,2H),4.44-4.37(m,3H),4.20-4.05(m,3H),3.86-3.82(m,2H),3.70-3.50 (m,2H),2.84-2.81(m,2H),1.31-1.16(m,3H).
Preparation eleven: preparation of (S) -5- ((1- (3- (5-ethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 36)
1. Preparation of 3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid
N 2 Under protection, 3-bromo-5- (trifluoromethyl) pyridin-2-amine (1.0 g,4.1 mmol) and Pd (tBu) 3 P) 2 (2.1 g,4.1 mmol), pentanoic acid (1.4 g,12.1 mmol), potassium phosphate (2.6 g,12.2 mmol), magnesium sulfate (2.5 g,20.8 mmol) and acetic acid (378 mg,12.3 mmol) DMA (40 mL), and reacted at 140℃for 3 hours. After the reaction was completed, cooling to room temperature, filtering, pouring the filtrate into water, extracting with ethyl acetate 3 times, combining ethyl acetate layers, washing with water, washing with saturated sodium chloride solution, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate under reduced pressure, and purifying the residue by a reverse phase column (water: methanol=1:5) to give the objective compound (710 mg, yield: 66.1%).
2. Preparation of 3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester
Contains 3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid (710 mg,2.7 mmol) and absolute ethanol (10 mL) of concentrated sulfuric acid (2 mL), and is reacted at 80℃for 8 hours. After the completion of the reaction, most of the solvent was removed under reduced pressure, the remaining reaction solution was diluted with ethyl acetate, the pH was adjusted to about 8 with saturated sodium bicarbonate solution, extraction was performed 3 times with ethyl acetate, the ethyl acetate layers were combined, washed with water, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by a reverse phase column (water: methanol=1:6) to give the objective compound (400 mg, yield: 50.9%).
3. Preparation of ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate
Potassium tert-butoxide (191 mg,1.7 mmol) was added in portions to DMF (20 mL) containing ethyl 3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (400 mg,1.4 mmol) at 0deg.C, after stirring for 30 min, tert-butyl 1, 2-oxathiazolidine-3-carboxylate 2, 2-dioxide (335 mg,1.5 mmol) was added and the reaction was carried out at 30deg.C for 1 hour. After the completion of the reaction, the reaction solution was poured into water, extracted with ethyl acetate 3 times, the ethyl acetate layers were combined, washed with water, washed with a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=1:1) to give the objective compound (445 mg, yield: 74.2%).
4. Preparation of ethyl 1- (2-aminoethyl) -3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate hydrochloride
Ethyl acetate hydrochloride solution (4 mL) was added to ethyl acetate (4 mL) containing ethyl 1- (2- ((tert-butoxycarbonyl) amino) ethyl) -3-ethyl-5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (150 mg,0.35 mmol) at 30℃and reacted for 3H at 30 ℃. After the reaction is finished, the reaction solution is directly decompressed and evaporated to dryness to obtain a crude product, which is directly used for the next reaction.
5. Preparation of 5-ethyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one
After the crude reaction product of the above step was dissolved in methanol (5 mL), potassium carbonate (276 mg,2.0 mmol) was added and the mixture was reacted at 30℃for 2 hours. After the reaction was completed, the solvent was removed under reduced pressure, and the residue was purified by a reverse phase column (methanol: water=4:1) to give the objective compound (98 mg, combined yield in two steps: 99.0%).
6. Preparation of 5-ethyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine
Lithium aluminum hydride (53 mg,1.4 mmol) was added to methyl tert-butyl ether (10 mL) containing 5-ethyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-6 (7H) -one (98 mg,0.35 mmol) at 0deg.C, and reacted at 60deg.C for 8 hours. After the completion of the reaction, the reaction was quenched by addition of water (0.5 mL), and the C18 powder was directly stirred and purified by reverse phase column (methanol: water=2:1) to give the objective compound (60 mg, yield: 64.4%).
7. Preparation of (S) -5- ((1- (3- (5-ethyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one
Containing 5-ethyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (60 mg,0.22 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (112 mg,0.26 mmol), HATU (125 mg,0.33 mmol) and DIPEA (85 mg,0.66 mmol) in dichloromethane (10 mL) was reacted at 29℃for 1 h. C18 powder was directly stirred and purified by reverse phase column (methanol: water=4:1) to give crude target compound (110 mg).
8. Preparation of (S) -5- ((1- (3- (5-ethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.4 mL) was added dropwise to trifluoroacetic acid (4 mL) containing (S) -5- ((1- (3- (5-ethyl-3- (trifluoromethyl) -8, 9-dihydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (110 mg crude) at 0 ℃ for 1 hour. Pouring the reaction solution into a saturated sodium bicarbonate solution, extracting with dichloromethane for 3 times, combining the dichloromethane layers, removing the solvent under reduced pressure, purifying the residue by reverse phase column (70% methanol/water system) and purifying the residue by silica gel preparation plate (ethyl acetate as developing agent) to obtain the target compound (40 mg, combined yield in two steps: 32.0%)
Molecular formula C 24 H 26 F 6 N 6 O 3 Molecular weight 560.5 LC-MS (M/e) 561.3 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.45(s,1H),8.52(s,1H),8.35(s,1H),7.90(s,1H),6.23(brs,1H),4.95-4.87(m,2H),4.26(m,1H),4.19-4.05(m,2H),3.98(m,2H),3.72(m,2H),3.50(m,2H),2.73(m,4H),1.23-1.18(m,3H),1.16-0.86(m,3H).
Twelve preparation examples: preparation of 5- (((S) -1- (3- ((R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 45)
1. Preparation of (R) -1- (1- ((tert-butoxycarbonyl) amino) propan-2-yl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester
Ethyl 5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (1.0 g,3.9 mmol) was dissolved in DMF (10 mL), potassium tert-butoxide (652 mg,5.8 mmol) was added at 25℃and reacted for 30 min, tert-butyl (S) -5-methyl-1, 2, 3-oxazolidine-3-carboxylate 2, 2-dioxide (919 mg,3.9 mmol) was added and reacted for 2H at 25 ℃. After the reaction, EA and water were added to extract, the organic phase was dried by spin-drying, and the desired compound (910 mg, yield 56.7%) was obtained by normal phase column chromatography (PE: ea=0-30%).
2. Preparation of (R) -1- (1-aminopropane-2-yl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester
Ethyl (R) -1- (1- ((tert-butoxycarbonyl) amino) propan-2-yl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (910 mg,2.2 mmol) was added to an ethyl acetate hydrochloride solution (20 mL) and reacted at 25℃for 2H. The reaction was completed and dried by spin-drying to give the objective compound (750 mg). .
3. Preparation of (R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-6 (7H) -one
Ethyl (R) -1- (1-aminopropane-2-yl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (750 mg,2.1 mmol) was dissolved in methanol (10 mL), and potassium carbonate (780 mg,6.4 mmol) was added and reacted at 25℃for 2H. DCM and water were added and extracted three times, and the organic phase was dried by spinning to give the title compound (500 mg, 86.9% yield).
4. Preparation of (R) -9-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine
(R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-6 (7H) -one (500 mg,1.9 mmol) was dissolved in THF (5 mL), lithium aluminum hydride (212 mg,5.6 mmol) was added and reacted at 25℃for 1H. Quenched with water and dried by spin to give the title compound (400 mg, yield 84.4%).
5. Preparation of 2- (4-methoxybenzyl) -5- (((S) -1- (3- ((R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
A solution of (R) -9-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine (100 mg,0.39 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (168 mg,0.39 mmol), HATU (447 mg,0.24 mmol) and DIPEA (151 mg,1.2 mmol) in acetonitrile (5 mL) was reacted at 25℃for 4 hours. Purification by silica gel column chromatography (PE: ea=0-100%) gave the title compound (70 mg, yield: 26.7%).
6. Preparation of 5- (((S) -1- (3- ((R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.5 mL) was added dropwise to trifluoroacetic acid (5 mL) containing 2- (4-methoxybenzyl) -5- (((S) -1- (3- ((R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (70 mg,0.10 mmol) and reacted at 25℃for 0.5H. The reaction solution was poured into saturated sodium bicarbonate solution, extracted 3 times with dichloromethane/isopropanol, the dichloromethane layers were combined, the solvent was removed under reduced pressure, and the residue was purified by silica gel column chromatography (PE: ea=0-100%) to give the objective compound (51 mg, yield: 88.9%).
Molecular formula C 23 H 24 F 6 N 6 O 3 Molecular weight 546.5 LC-MS (M/e): 547.2 (M+H) + )
1 H-NMR(400MHz,CD 3 Cl)δ:11.15(s,1H),8.54(s,1H),8.10(s,1H),7.67-7.64(m,1H),6.36(s,1H),5.81(s,1H),5.58-5.54(m,1H),5.00(s,1H),4.81-4.73(m,1H),4.52-4.41(m,1H),3.90-3.82(m,3H),3.72-3.68(m,2H),3.39-3.36(m,1H),2.82-2.73(m,2H),1.55-1.48 (m,3H),1.35-1.22(m,3H).
Preparation example thirteen: preparation of 5- (((S) -1- (3- ((R) -5-ethyl-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxypropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 47)
1. (R) -1- (1- ((tert-Butoxycarbonyl) -lambda 2 -nitroxyl) propan-2-yl) -5- (trifluoromethyl) -1H-pyrrole [2,3-b]Preparation of pyridine-2-carboxylic acid ethyl ester
Ethyl 5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (5 g, crude) was dissolved in DMF (50 mL), potassium tert-butoxide (3.3 g,29.1 mmol) was added at 0 ℃ and after 0.5H tert-butyl (S) -5-methyl-1, 2, 3-oxazolidine-3-carboxylate 2, 2-dioxide (4.6 g,19.4 mmol) was added, the reaction was allowed to react for 4H at 25 ℃ and lcms detection was completed, water quenching was added to the reaction solution, EA extraction, organic phase spin-drying, silica gel column chromatography (EA: pe=1:4) gave the title compound (4.9 g).
2. Preparation of (R) -1- (1-aminopropane-2-yl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester hydrochloride
(R) -1- (1- ((tert-Butoxycarbonyl) -lambda 2 -nitroxyl) propan-2-yl) -5- (trifluoromethyl) -1H-pyrrole [2,3-b]Preparation of pyridine-2-carboxylic acid ethyl ester (4.9 g,11.8 mmol) was dissolved in HCl/dioxane (4M) solution (100 mL), reacted at 25℃for 4h, and the reaction was detected by LCMS and was dried by spin and used directly in the next reaction.
3. Preparation of (R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-6 (7H) -one
(R) -1- (1-aminopropane-2-yl) -5- (trifluoromethyl) -1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid ethyl ester hydrochloride (crude product, 11.8 mmol) was dissolved in methanol (100 mL), potassium carbonate (6.5 g,47.2 mmol) was added, the reaction was completed by LCMS detection at 25℃and the reaction solution was dried by spin-drying with dichloromethane, diluted with water, and the organic phase was dried by spin-drying to give the objective compound (2.8 g, yield 88.1%).
4. Preparation of (R) -9-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine
(R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrol [1,2-a ] pyrazin-6 (7H) -one (2.7 g,10.0 mmol) was dissolved in methyl tert-butyl ether (50 mL), lithium aluminum hydride (1.5 g,40.0 mmol) was added at 0deg.C, LCMS detection reaction was complete, quenched with water and 15% aqueous sodium hydroxide solution, filtered, and the filtrate was dried to give the title compound (2.5 g, 98.0% yield).
5. Tert-butyl (R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
(R) -9-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5]Pyrrole [1,2-a ]]Pyrazine (2.4 g,9.40 mmol) was dissolved in dichloromethane (40 mL) and added (Boc) 2 O (3.1 g,14.1 mmol) and triethylamine (1.9 g,18.8 mmol) were reacted at 25℃for 2 hours, after the completion of the reaction, the reaction solution was dried by spin, and the objective compound (3 g, yield 89.8%) was isolated by silica gel column chromatography (EA: PE=1:4).
6. Tert-butyl (R) -5-bromo-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
Tert-butyl (R) -9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (2.9 g,8.16 mmol) was dissolved in DMF (50 mL), NBS (1.9 g,10.6 mmol) was added, reacted at 25℃for 2H, after completion of LCMS detection reaction, quenched with water, filtered and the filter cake was isolated by silica gel column chromatography (EA: PE=1:4) to give the title compound (2.8 g, yield 79.0%).
7. Preparation of tert-butyl (R) -9-methyl-3- (trifluoromethyl) -5-vinyl-8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To a solution of tert-butyl (R) -5-bromo-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (1.6 g,3.68 mmol) in dioxane (40 mL) was added pinacol vinylborate (1.7 g,11.0 mmol), tetrakis (triphenylphosphine) palladium (855 mg,0.74 mmol), potassium carbonate (1.5 g,11.0 mmol) and water (10 mL), reacted at 90 ℃ for 16H, lcms detection was completed, quenched with water, extracted with ethyl acetate, the organic phase was spin-dried and isolated the title compound (820 mg, 58.4%) by silica gel column chromatography (EA: pe=1:3).
8. Preparation of tert-butyl (R) -5-ethyl-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To a solution of tert-butyl (R) -9-methyl-3- (trifluoromethyl) -5-vinyl-8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (790 mg,2.07 mmol) in methanol (10 mL) was added palladium on carbon (790 mg), and the mixture was stirred at 25℃for 4 hours, filtered through celite after completion of the reaction, and the reaction mixture was dried by spin-drying to give the objective compound (740 mg, yield: 93.2%).
9. Preparation of (R) -5-ethyl-9-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine trifluoroacetate salt
To tert-butyl (R) -5-ethyl-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2': trifluoroacetic acid (4 mL) was added dropwise to a solution of 4, 5-pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (740 mg,1.9 mmol) in dichloromethane (8 mL), and the reaction mixture was stirred at 25℃for 2H, after which the reaction mixture was dried by spinning to give a crude product (1.3 g) which was directly used in the next reaction.
10. Preparation of 5- (((S) -1- (3- ((R) -5-ethyl-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxypropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of 5-ethyl-9-methyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine trifluoroacetate (1.25 g, crude, 1.83 mmol) in dichloromethane (20 mL) was added (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propionic acid (749 mg,1.74 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 1.0g,2.63 mmol), N, N-diisopropylethylamine (946 mg,7.32 mmol). Stirring is carried out for 1h at 25 ℃. After the reaction was completed, the reaction solution was dried by spin drying. The title compound was isolated by silica gel column chromatography (EA: pe=4:1) (750 mg, yield: 60.0%).
11. Preparation of 5- (((S) -1- (3- ((R) -5-ethyl-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxypropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of 5- (((S) -1- (3- ((R) -5-ethyl-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrol [1,2-a ] pyrazin-7 (6H) -yl) -3-oxypropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (700 mg,1.0 mmol) in trifluoroacetic acid (10 mL) was added dropwise trifluoromethanesulfonic acid (2 mL). The reaction was carried out at 25℃for 1h. After removing most of TFA from the reaction solution under reduced pressure, the remaining reaction solution was diluted with ethyl acetate, the pH was adjusted to 8 with saturated sodium bicarbonate solution, the ethyl acetate layer was washed with water, with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (methanol: dichloromethane=1:10) and medium pressure reverse phase column chromatography (methanol: water=0-80%) to give the title compound (314 mg, yield: 54.0%)
Molecular formula C 25 H 28 F 6 N 6 O 3 Molecular weight 574.2 LC-MS (M/e): 575.2 (M+H) + )
1 H-NMR(400MHz,CDCl 3 )δ:12.43(s,1H),8.51(s,1H),8.32(s,1H),7.82-7.90(m,H),6.25(s,1H),5.10-5.30(m,1H),4.75-4.85(m,1H),4.35-4.50(m,1H),4.00-4.20(m,2H),3.60-3.78(m,3H),3.40-3.50(m,2H),2.60-2.81(m,4H),1.25-1.35(m,3H),1.05-1.24(m,6H).
Preparation example fourteen: preparation of 5- (((S) -1- (3- ((R) -5, 9-dimethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 49)
1. Preparation of tert-butyl (R) -5, 9-dimethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To a solution of tert-butyl (R) -5-bromo-9-methyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (800 mg,1.84 mmol) in 1, 4-dioxane (20 mL) was added methylboronic acid (661 mg,11.04 mmol), tetrakis triphenylphosphine palladium (427 mg,0.37 mmol), potassium carbonate (1.0 g,7.36 mmol) and water (4 mL), reacted at 100deg.C for 16H, LCMS was detected, quenched with water, extracted with ethyl acetate, and the organic phase was spin-dried and the product isolated by silica gel column chromatography (EA: PE=1:4) (280 mg, yield 41.3%).
2. Preparation of (R) -5, 9-dimethyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine trifluoroacetate salt
To a solution of tert-butyl (R) -5, 9-dimethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (280 mg,0.76 mmol) in methylene chloride (10 mL), trifluoroacetic acid (2 mL) was added dropwise, and the mixture was stirred at 25℃for 2 hours, after the reaction was completed, the reaction solution was dried by spin-drying to obtain a crude product, which was directly used for the next reaction.
3. Preparation of 5- (((S) -1- (3- ((R) -5, 9-dimethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of (R) -5, 9-dimethyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine trifluoroacetate (crude) in dichloromethane (10 mL) was added N, N-diisopropylethylamine (393 mg,3.04 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (326 mg,0.76 mmol) and 2- (7-azabenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 578mg,1.52 mmol). Stirring is carried out for 1h at 25 ℃. After completion of the reaction, quench with water, extract with DCM and spin dry the organic phase. The product was isolated by silica gel column chromatography (EA: pe=9:1) (300 mg, two step yield: 57.9%).
4. Preparation of 5- (((S) -1- (3- ((R) -5, 9-dimethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of 5- (((S) -1- (3- ((R) -5, 9-dimethyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2'4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (300 mg,0.44 mmol) in trifluoroacetic acid (TFA, 8 mL) was added dropwise trifluoromethanesulfonic acid (1 mL). The reaction was carried out at 25℃for 1h. After removing most of TFA from the reaction solution under reduced pressure, the remaining reaction solution was diluted with dichloromethane, the pH was adjusted to 8 by washing with saturated sodium bicarbonate solution, the organic phase was washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (methanol: dichloromethane=1:20) to give the objective compound (133 mg, yield: 53.9%)
Molecular formula C 24 H 26 F 6 N 6 O 3 Molecular weight 560.5 LC-MS (M/e): 561.0 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.42(s,1H),8.51(s,1H),8.30(s,1H),7.88-7.84(m,1H),6.24(s,1H),5.10-5.30(m,1H),4.80-4.90(m,1H),4.30-4.70(m,1H),4.00-4.20(m,2H),3.62-3.82(m,3H),3.40–3.50(m,2H),2.60-2.80(m,2H),2.21(s,3H),1.20–1.40(m,3H),1.10(s,3H).
Preparation example fifteen: preparation of (S) -5- ((1- (3-oxo-3- (5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 56)
1. Preparation of tert-butyl 5-bromo-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylic acid tert-butyl ester (1.2 g,3.5 mmol) was dissolved in DMF (35 mL), NBS (814 mg,10.6 mmol) was added, reaction was carried out at 25℃for 2H, after completion of LCMS detection reaction, water quench was added, ethyl acetate extracted the organic phase, and silica gel column chromatography (EA: PE=1:9) was dried to give the title compound (1.0 g, yield 66.7%).
2. Preparation of tert-butyl 5- (3, 6-dihydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To tert-butyl 5-bromo-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (900 mg,2.1 mmol) in dioxane (40 mL) and water (8 mL) was added 2- (3, 6-dihydro-2H-pyran-4-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane (1.3 g,6.2 mmol), tetrakis (triphenylphosphine) palladium (481 mg,0.42 mmol), potassium carbonate (871 mg,6.3 mmol), nitrogen protection at 100℃for 3H, LCMS detection was completed, quenched with water, extracted with ethyl acetate, dried over organic phase, and silica gel column chromatography (EA: PE=1:5) was isolated as the title compound (850 mg, 93.7%).
3. Preparation of tert-butyl 5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To a solution of tert-butyl 5- (3, 6-dihydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (850 mg,2.0 mmol) in methanol (25 mL) was added palladium on carbon (1.3 g), stirred at 30℃for 2H, and after completion of the reaction, celite was filtered, the filtrate was dried, and silica gel column chromatography (EA: PE=1:6) to give the title compound (750 mg, yield 87.8%).
4. Preparation of 5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine trifluoroacetate salt
To a solution of tert-butyl 5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (750 mg,1.8 mmol) in methylene chloride (4 mL) was added dropwise trifluoroacetic acid (13 mL), and the mixture was stirred at 25℃for 1H, after the completion of the reaction, the reaction was dried by spin-drying to give a crude product which was directly used for the next reaction.
5. Preparation of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of 5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine trifluoroacetate (crude, 1.8 mmol) in dichloromethane (30 mL) was added (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propionic acid (773 mg,1.8 mmol), 2- (7-azabenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (1.0 g,2.6 mmol), N, N-diisopropylethylamine (930 mg,7.2 mmol). Stirring is carried out for 1h at 25 ℃. The system was quenched with water, extracted with dichloromethane, the organic phase was spin-dried and the target compound was isolated by silica gel column chromatography (ea=100%) (650 mg, yield (two steps) 50%).
6. Preparation of (S) -5- ((1- (3-oxo-3- (5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of (S) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (5- (tetrahydro-2H-pyran-4-yl) -3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (620 mg,0.84 mmol) in trifluoroacetic acid (8 mL) was added dropwise trifluoromethanesulfonic acid (1 mL). The reaction was carried out at 25℃for 15 minutes. After removing most of TFA from the reaction solution under reduced pressure, the remaining reaction solution was diluted with dichloromethane, saturated sodium bicarbonate solution was adjusted to be basic, the organic phase was extracted with dichloromethane, concentrated, and the residue was subjected to silica gel column chromatography (methanol: dichloromethane=1:10) to give the objective compound (306 mg, yield: 59.0%)
Molecular formula C 27 H 30 F 6 N 6 O 4 Molecular weight 616.6 LC-MS (M/e): 617.2 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.43(s,1H),8.57(s,1H),8.35(s,1H),7.90(s,1H),6.25(s,1H),5.05-4.90(m,2H),4.30-4.10(m,4H),4.08-3.90(m,4H),3.80-3.65(m,2H),3.60-3.42(m,4H),2.85-2.70(m,2H),2.10-1.90(m,2H),1.75-1.60(m,2H),1.25(s,3H).
Preparation example sixteen: preparation of (S) -5- ((1- (3- (5-cyclopropyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 57)
1. Preparation of tert-butyl 5-bromo-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To 3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5]Pyrrole [1,2-a ]]To a solution of pyrazine-7 (6H) -carboxylic acid tert-butyl ester (2.5 g,7.3 mmol) in N, N-dimethylformamide (30 mL) was added N-bromosuccinimide (NBS, 1.6g,8.8 mmol). After the reaction was completed at 70℃for 1 hour, the reaction was quenched with water, the reaction mixture was extracted with ethyl acetate, the collected organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, dried over spin-on-column chromatography (SiO 2 Petroleum ether: ethyl acetate=6:1) to give the objective compound (1.6 g, yield 52.1%).
2. Preparation of tert-butyl 5-cyclopropyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate
To 5-bromo-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5]Pyrrole [1,2-a ]]To a solution of pyrazine-7 (6H) -carboxylic acid tert-butyl ester (720 mg,1.7 mmol) in 1, 4-dioxane (20 mL) was added cyclopropylboronic acid (178 mg,8.5 mmol), tetrakis triphenylphosphine palladium (196 mg,0.17 mmol), potassium carbonate (704 mg,5.1 mmol), water (5 mL). Stirring at 100deg.C for 12 hr, spin drying, column chromatography (SiO 2 Petroleum ether ethyl acetate=6:1) to give the target compound (92 mg, yield 14.1%).
3. Preparation of 5-cyclopropyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine
To a solution of tert-butyl 5-cyclopropyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazine-7 (6H) -carboxylate (92 mg,0.24 mmol) in methylene chloride (5 mL), trifluoroacetic acid (2 mL) was added dropwise, and the reaction mixture was stirred at 20℃for 3 hours, and the reaction mixture was dried to give a crude product (125 mg) which was directly used for the next reaction.
4. Preparation of (S) -5- ((1- (3- (5-cyclopropyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To 5-cyclopropyl-3- (trifluoromethyl) -6,7,8, 9-tetrahydropyridine [3',2':4,5]Pyrrole [1,2-a ]]To a solution of pyrazine (125 mg, crude) in dichloromethane (4 mL) was added (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (94 mg,0.22 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 91mg,0.24 mmol), N, N-diisopropylethylamine (62 mg,0.48 mmol). At 25 DEG C Stirring for 12h. After the reaction was completed, the reaction solution was dried by spin drying. Column chromatography (SiO) 2 Dichloromethane: methanol=15:1) to give crude (250 mg) which was used directly in the next reaction.
5. (S) -5- ((1- (3- (5-cyclopropyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxopropoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
To a solution of (S) -5- ((1- (3- (5-cyclopropyl-3- (trifluoromethyl) -8, 9-dihydropyridine [3',2':4,5] pyrrole [1,2-a ] pyrazin-7 (6H) -yl) -3-oxypropoxy) propan-2-yl) amino) -2- (4-methoxybenzyl) -4- (trifluoromethyl) pyridazin-3 (2H) -one (250 mg, crude) in trifluoroacetic acid (2 mL) was added dropwise trifluoromethanesulfonic acid (0.1 mL). The reaction was carried out at 20℃for 2h. After removing most of TFA in the reaction solution under reduced pressure, the remaining reaction solution was diluted with ethyl acetate, the pH was adjusted to 8 with saturated sodium bicarbonate solution, the ethyl acetate layer was washed with water, with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol=15:1) and medium pressure reverse phase column chromatography (methanol: water=9:1) to give the title compound (20 mg, yield: 14.6%).
Molecular formula C 25 H 26 F 6 N 6 O 3 Molecular weight 572.2 LC-MS (M/e): 573.2 (M+H) + )
1 H-NMR(400MHz,CDCl 3 )δ:10.65(brs,1H),8.51(d,J=10.0Hz,1H),8.17(s,1H),7.66(s,1H),5.82(s,1H),5.05(s,1H),4.95(s,1H),4.43-4.25(m,2H),4.21-4.05(m,1H),3.98-3.79(m,4H),3.73-3.58(m,1H),3.58-3.40(m,1H),2.81-2.68(m,2H),1.88-1.68(m,1H),1.38-1.20(m,3H),0.83-0.69(m,2H).
Seventeenth preparation example: preparation of(s) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -9, 10-dihydroimidazo [2,1-a ] [2,6] naphthyridin-8 (7H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 7)
1. Preparation of 8-benzyl-7, 8,9, 10-tetrahydroimidazo [2,1-a ] [2,6] naphthyridine
2-bromo-1, 1-dimethoxyethane (5.5 g,32.7 mmol) was dissolved in ethanol (230 mL), diluted HCl (1M, 200mL,200 mmol) was added and reacted at 90℃for 1h, cooled to 25℃and saturated NaHCO 3 The solution was adjusted to ph=7-8, 6-benzyl-5, 6,7, 8-tetrahydro-2, 6-naphthyridin-1-amine (4.7 g,20 mmol) was added and reacted at 90 ℃ for 2h. After the reaction was completed, it was dried and isolated by forward preparation (DCM: meoh=20:1) to give the title compound (3.1 g, 60% yield).
2. Preparation of 8-benzyl-3- (trifluoromethyl) -7,8,9, 10-tetrahydroimidazo [2,1-a ] [2,6] naphthyridine
8-benzyl-7, 8,9, 10-tetrahydroimidazole [2,1-a ]][2,6]Naphthyridine (2.6 g,10 mmol) was dissolved in acetonitrile (130 mL) and TMSCF was added 3 (5.7g,40mmol),PhI(OAc) 2 (6.4 g,20 mmol) CsF (6.1 g,40 mmol), after the addition, the reaction is carried out for 4 hours at 30 ℃, the reaction is completed and the filtration is carried out, the filtrate is dried by spin, crude products are obtained by forward preparation and separation (PE: EA=3:1), and crude products (200 mg) are obtained by reverse C18 preparation and separation (water: methanol=1:2).
3. Preparation of 3- (trifluoromethyl) -7,8,9, 10-tetrahydroimidazo [2,1-a ] [2,6] naphthyridine
8-benzyl-3- (trifluoromethyl) -7,8,9, 10-tetrahydroimidazo [2,1-a][2,6]Naphthyridine (200 mg, crude) was dissolved in methanol (5 mL) and Pd/C (20 mg), H was added 2 The reaction was carried out for 2 hours. After the reaction is finished, the solution is filtered,the filtrate was dried by spin-drying and separated by reverse C18 (water: methanol=1:2) to give crude (35 mg).
4. Preparation of(s) -2- (4-methoxybenzyl) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -9, 10-dihydroimidazo [2,1-a ] [2,6] naphthyridin-8 (7H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
3- (trifluoromethyl) -7,8,9, 10-tetrahydroimidazo [2,1-a ] [2,6] naphthyridine (35 mg crude) was dissolved in DCM (3 mL), and (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (62 mg,0.15 mmol), HATU (83 mg,0.2 mmol), DIEA (56 mg,0.4 mmol) was added and reacted at 30℃for 1h. After the reaction, the mixture was dried by spinning and separated by forward preparation (PE: EA=1:1) to give crude (40 mg).
5. Preparation of(s) -5- ((1- (3-oxo-3- (3- (trifluoromethyl) -9, 10-dihydroimidazo [2,1-a ] [2,6] naphthyridin-8 (7H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
The crude product of the above step was dissolved in TFA (3 mL), and trifluoromethanesulfonic acid (0.3 mL) was added dropwise at 0deg.C, and reacted at 30deg.C for 30min. After the reaction is completed, pouring saturated NaHCO 3 To the solution, EA and water were added for three extractions, the organic phase was dried by spinning, and the target compound (10 mg) was obtained by reverse preparative separation (water: methanol=1:1).
Molecular formula C 22 H 22 F 6 N 6 O 3 Molecular weight 532.4 LC-MS (M/e): 533.1 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.43(s,1H),8.35-8.39(t,1H),8.10(s,1H),7.88-7.90(d,J=8Hz,1H),7.00-7.07(t,1H),6.25(s,1H),4.60-4.70(m,2H),4.20(s,1H),3.60-3.80(m,4H),3.50(m,2H),2.85-3.11(m,2H),2.60-2.70(m,2H),1.05-1.2(m,3H)。
Preparation example eighteen: preparation of 5- (((S) -1- (3-oxo-3- ((S) -3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (Compound 28-1)
1. Preparation of 2- (benzyloxy) -3-bromo-5- (trifluoromethyl) pyridine
3-bromo-2-chloro-5- (trifluoromethyl) pyridine (20 g,77 mmol) was dissolved in NMP (200 mL), benzyl alcohol (9.9 g,92 mmol) and cesium carbonate (75.1 g,230 mmol) were added, the mixture was then left to react at 90℃for 16h, LCMS detection was complete, the temperature was reduced to 25℃and the reaction solution was poured into () water (1.4L) to precipitate a solid, which was filtered and the filter cake dried to give the title compound (19.5 g, 76.8% yield).
2. Preparation of methyl (S) -3- (2- (benzyloxy) -5- (trifluoromethyl) pyridin-3-yl) -2- ((tert-butoxycarbonyl) amino) propanoate
Zinc powder (11.9 g,181 mmol) was added to N, N-dimethylformamide (50 mL), nitrogen was replaced three times, a solution of iodine (3.2 g,13 mmol) in DMF (5 mL) was added, reaction was performed at 25℃for 10min, a solution of (R) -2- ((tert-butoxycarbonyl) amino) -3-iodopropionic acid methyl ester (19.9 g,60 mmol) in DMF (20 mL) and a solution of iodine (3.2 g,13 mmol) in DMF (5 mL) were added, reaction was performed at 25℃for 30min, a solution of 2- (benzyloxy) -3-bromo-5- (trifluoromethyl) pyridine (10.0 g,30 mmol) and ditriphenylphosphoric palladium dichloride (1.1 g,1.5 mmol) in DMF (20 mL) was added, the mixture was reacted at 120℃for 2h, LCMS detection was completed, the solvent was dried to 100mL, suction filtration was performed, and the filter cake was purified by a column of dichloromethane-concentrated silica gel (ethyl acetate/petroleum ether=1:5) to give the target compound (2.5 g, 18.2%).
3. Preparation of methyl (S) -2- ((tert-Butoxycarbonyl) amino) -3- (2-hydroxy-5- (trifluoromethyl) pyridin-3-yl) propanoate
Methyl (S) -3- (2- (benzyloxy) -5- (trifluoromethyl) pyridin-3-yl) -2- ((tert-butoxycarbonyl) amino) propionate (2.7 g,6 mmol) was dissolved in dichloromethane (16 mL) and methanol (4 mL), wet palladium on carbon (270 mg) was added, nitrogen was replaced three times, hydrogen was replaced three times, the reaction system was reacted at 25℃for 3h, LCMS detection was complete, the reaction solution was filtered, and the organic phase was dried by spin to give the title compound (2.1 g, yield 95.5%).
4. (S) -2- ((tert-Butoxycarbonyl) amino) -3- (5- (trifluoromethyl) -2- (((trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) propanoic acid methyl ester
Methyl (S) -2- ((tert-butoxycarbonyl) amino) -3- (2-hydroxy-5- (trifluoromethyl) pyridin-3-yl) propionate (2.1 g,6 mmol) was dissolved in dichloromethane (20 mL), pyridine (1.8 g,23 mmol) was added at 0deg.C, trifluoromethanesulfonic anhydride (2.4 g,8.6 mmol) was added dropwise at 0deg.C, the reaction was carried out for 3h, after completion of LCMS detection, the reaction solution was concentrated, pH was adjusted to alkaline with saturated sodium bicarbonate solution, the aqueous phase was extracted once with DCM, and the combined organic phases were concentrated to give the title compound (2.6 g, yield 90.3%).
5. Preparation of methyl (S) -2-amino-3- (2-chloro-5- (trifluoromethyl) pyridin-3-yl) propionate
Methyl (S) -2- ((tert-butoxycarbonyl) amino) -3- (5- (trifluoromethyl) -2- (((trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) propionate (2.6 g,5.2 mmol) was dissolved in EA (20 mL), HCl/EA solution (4 n,10 mL) was added dropwise, the mixture was reacted at 20 ℃ for 18h, the reaction solution was concentrated to dryness, dichloromethane (20 mL) was added, ph=8 was adjusted with saturated sodium bicarbonate solution, the organic phase was washed with water, and concentrated to give the desired product (crude product, 1.8g, which was used directly in the next reaction).
6. Preparation of (S) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridine-2-carboxylic acid methyl ester
To a microwave tube were added methyl (S) -2-amino-3- (2-chloro-5- (trifluoromethyl) pyridin-3-yl) propionate (1.8 g, crude product of the above step), DIPEA (4.1 g,31.8 mmol) and DMSO (6 mL) and reacted at 140℃for 1 hour. After the reaction, water was added to conduct extraction and deactivation reaction, ethyl acetate was extracted three times, ethyl acetate layers were combined, washed with water and saturated sodium chloride solution, the organic phase was removed under reduced pressure, and the residue was subjected to silica gel column chromatography (petroleum ether: ethyl acetate=3:1) to give the objective compound (800 mg, two-step yield: 62.5%).
7. Preparation of (S) - (5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-2-yl) methanol
Sodium borohydride (500 mg,13.2 mmol) was added to absolute ethanol (40 mL) containing methyl (S) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridine-2-carboxylate (650 mg,2.64 mmol), and reacted at 20℃for 3H under nitrogen. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=1:1) to give the objective compound (550 mg, yield: 95.5%).
8. Preparation of (S) -2- (((tert-butyldiphenylsilyl) oxy) methyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridine
TBDPSCl (1.0 g,3.6 mmol) was added to methylene chloride (30 mL) containing (S) - (5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-2-yl) methanol (550 mg,2.5 mmol) and imidazole (511 mg,7.5 mmol) and reacted at 20℃for 1H under nitrogen. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=2:1) to give the objective compound (720 mg, yield: 62.6%).
9. Preparation of tert-butyl (S) -2- (2- (((tert-butyldiphenylsilyl) oxy) methyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-1-yl) acetate
Sodium hydride (300 mg,7.5 mmol) was added to DMF (20 mL) containing (S) -2- (((tert-butyldiphenylsilyl) oxy) methyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridine (700 mg,1.5 mmol) at 0deg.C, and after 5 minutes the reaction was continued, tert-butyl bromoacetate (719 mg,2.2 mmol) was added. After the reaction, water was added to conduct extraction and deactivation, ethyl acetate was extracted three times, ethyl acetate layers were combined, washed with water and saturated sodium chloride solution, the organic phase was removed under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=3:1) to give the objective compound (810 mg, yield: 92.6%).
10. Preparation of (S) -2- (2- (((tert-butyldiphenylsilyl) oxy) methyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-1-yl) ethan-1-ol
Sodium borohydride (265 mg,7.0 mmol) was added to anhydrous ethanol (40 mL) containing (S) -tert-butyl 2- (2- (((tert-butyldiphenylsilyl) oxy) methyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-1-yl) acetate (800 mg,1.4 mmol) and anhydrous calcium chloride (777 mg,7.0 mmol) and reacted at 20℃for 15H under nitrogen. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was directly purified by silica gel column chromatography (petroleum ether: ethyl acetate=1:1) to give the objective compound (500 mg, yield: 71.2%).
11. Preparation of (S) -2- (2- (hydroxymethyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-1-yl) ethan-1-ol
TBAF (2.3 mL,2.3 mmol) was added to THF (5 mL) containing (S) -2- (2- (((tert-butyldiphenylsilyl) oxy) methyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-1-yl) ethan-1-ol (230 mg,0.46 mmol) and reacted at 20deg.C under nitrogen for 15H. After the completion of the reaction, the reaction solution was concentrated under reduced pressure, and the residue was directly purified on a silica gel plate (dichloromethane: methanol=10:1) to give the objective compound (120 mg, yield: 99.6%).
12. Preparation of methyl (S) - (1- (2- ((methylsulfonyl) oxy) ethyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-2-yl) methylsulfonate
MsCl (79 mg,0.69 mmol) was added to dichloromethane (5 mL) containing (S) -2- (2- (hydroxymethyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-1-yl) ethan-1-ol (60 mg,0.23 mmol) and triethylamine (121 mg,1.2 mmol) at 0deg.C, and the reaction was continued for 20 min. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was directly purified on a silica gel plate (petroleum ether: ethyl acetate=1:1) to give the objective compound (70 mg, yield: 73.1%).
13. Preparation of (S) -3- (trifluoromethyl) -5,5a,6,7,8, 9-hexahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine
To the tube was added methyl (S) - (1- (2- ((methylsulfonyl) oxy) ethyl) -5- (trifluoromethyl) -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-2-yl) methylsulfonate (70 mg,0.17 mmol), aqueous ammonia (4 mL) and acetonitrile (2 mL), and the mixture was reacted at 90℃for 2 hours. After the completion of the reaction, the reaction solution was extracted with ethyl acetate three times, and the ethyl acetate layers were combined, washed with water and saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the objective product (40 mg, yield: 98.3%).
14. Preparation of 2- (4-methoxybenzyl) -5- (((S) -1- (3-oxo-3- ((S) -3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
Containing (S) -3- (trifluoromethyl) -5,5a,6,7,8, 9-hexahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazine (40 mg,0.16 mmol), (S) -3- (2- ((1- (4-methoxybenzyl) -6-oxo-5- (trifluoromethyl) -1, 6-dihydropyridazin-4-yl) amino) propoxy) propanoic acid (69 mg,0.16 mmol), HATU (91 mg,0.24 mmol) and DIPEA (62 mg,0.48 mmol) in dichloromethane (2 mL) was reacted at 25℃for 1 hour. After the completion of the reaction, the reaction mixture was dried under reduced pressure, and the residue was purified on a silica gel plate (ethyl acetate as a developing solvent) to give the objective compound (65 mg, yield: 60.4%).
15. Preparation of 5- (((S) -1- (3-oxo-3- ((S) -3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one
TfOH (0.2 mL) was added dropwise to trifluoroacetic acid (2 mL) containing 2- (4-methoxybenzyl) -5- (((S) -1- (3-oxo-3- ((S) -3- (trifluoromethyl) -5a,6,8, 9-tetrahydropyrido [3',2':4,5] pyrrolo [1,2-a ] pyrazin-7 (5H) -yl) propoxy) propan-2-yl) amino) -4- (trifluoromethyl) pyridazin-3 (2H) -one (65 mg,0.10 mmol) at 0 ℃ for 0.5 hours. The reaction solution was poured into a saturated sodium hydrogencarbonate solution, extracted with methylene chloride 3 times, the methylene chloride layers were combined, the solvent was removed under reduced pressure, and the residue was purified on a silica gel preparation plate (ethyl acetate as a developing agent) to give the title compound (25 mg, yield: 47.1%).
Molecular formula C 22 H 24 F 6 N 6 O 3 Molecular weight 534.5 LC-MS (M/e): 535.2 (M+H) + )
1 H-NMR(400MHz,DMSO)δ:12.43(brs,1H),8.09(s,1H),7.90(s,1H),7.50(s,1H),6.24(m,1H),4.51-4.42(m,1H),4.15(m,1H),4.11-3.98(m,2H),3.92-3.85(m,1H),3.71(m,3H),3.51(m,2H),3.19-3.11(m,1H),2.99-2.85(m,2H),2.62-2.52(m,3H),1.16-1.14(m,3H).
Using the same or similar methods as the preparation examples described above, the compounds shown in the following tables were prepared:
experimental protocol
Exemplary protocols for some of the compounds of the present invention are provided below to demonstrate the advantageous activity and beneficial technical effects of the compounds of the present invention. It should be understood that the following experimental schemes are merely illustrative of the present disclosure and are not intended to limit the scope of the present disclosure.
Experimental example 1 in vitro enzymatic Activity of Compounds of the invention
Test article: the structural formula and the preparation method of the compounds shown in the table 1 are shown in the preparation example of the invention;
compound a, prepared according to prior art methods, has the structure;
abbreviations used in the following experiments represent the following meanings:
DMSO: dimethyl sulfoxide;
PBS: phosphate buffer;
PBST: phosphate buffer containing tween 20;
HRP: horseradish peroxidase
The experimental method comprises the following steps: compounds were enzymatically evaluated in vitro on PARP7 using the PARP7 chemiluminescence method from BPS Bioscience.
The experimental steps are as follows:
1. reagent preparation
1.1 histone solutions: diluting the 5 Xhistone mixture into a 1 Xworking fluid using PBS;
1.2 main mixture: diluting 10×parp buffer and 10×parp mixture with water to prepare a main mixture;
1.3 PARP7 enzyme: preparing PARP7 enzyme working solution by using 1 XPARP buffer solution;
1.4 streptavidin-HRP solution: diluting 50 Xstreptavidin-HRP into 1 Xworking solution by using a blocking buffer;
1.5 detection solution: the substrate A solution and the substrate B solution are mixed in a ratio of 1:1, prepared in situ and mixed on ice.
2. Preparing a test compound:
2.1 preparing a drug solution with a final concentration of 1000 times: a suitable amount of 10mM stock solution of the drug was taken, diluted 10-fold with DMSO to 1mM, and then diluted 10-fold with DMSO at a gradient of 6 drug concentrations.
2.2 preparing a drug solution with a final concentration of 10 times, and diluting the drug-DMSO solution with a gradient of 100 times with ultrapure water. The concentration of test compound in the final system was 1000, 100, 10, 1, 0.1, 0.01nM;
3. the test steps are as follows:
3.1 coating 96-well plates, 50 μl histone solution working fluid/well, incubating overnight at 4 ℃;
3.2 washing the plate: 200 μl PBST/hole washing 3 times, and tapping and spin-drying;
3.3 closing: 200 μl blocking buffer 3/well, incubated at room temperature for 60 min;
3.4 washing the plate: 200 μl PBST/hole washing 3 times, and tapping and spin-drying;
3.5 adding the main mixture: 25 μl/well (2.5 μl 10 XPAR buffer+2.5 μl 10 XPAR mixture+20 μl water);
adding the medicine: 5 μl/well of test compound; positive control wells and blank wells were added 5 μl of 0.1% DMSO;
Adding enzyme: positive control wells and test compound wells were incubated with 20. Mu.l/well PARP7 enzyme, blank wells with 20. Mu.l 1 XPAR buffer for 1 hour at room temperature;
3.6 washing the plate: 200 μl PBST/hole washing 3 times, and tapping and spin-drying;
3.7 adding 50 μl/well of streptavidin-HRP solution, incubating for 30 minutes at room temperature;
3.8 washing the plate: 200 μl PBST/hole washing 3 times, and tapping and spin-drying;
3.9 detection: the chemiluminescent value was measured immediately by adding 100. Mu.l/well of the test solution.
4. Data processing
Fitting the data using nonlinear S-curve regression to derive a curve and calculating therefrom the relative IC 50 Values.
Activity (%) = (test substance well reading-blank well reading)/(positive control well reading mean-blank well reading) ×100%
Experimental results
TABLE 2 in vitro enzymatic inhibitory Activity of the Compounds of the invention
Conclusion of the experiment
The compounds of the invention in Table 1 have better PARP7 inhibition activity and IC 50 Values were all less than 1. Mu.M, wherein the IC representing the compound was tested 50 The values are all less than 100nM, and the inhibition activity is better.
Experimental example 2 inhibition experiment of in vitro cell Activity by the Compounds of the present invention
Test article: the structural formula and the preparation method of the compounds shown in the table 1 are shown in the preparation example of the invention;
compound a, prepared according to prior art methods, has the structure;
Abbreviations used in the following experiments represent the following meanings:
DMSO: dimethyl sulfoxide;
GI 50 : concentration of compound corresponding to 50% inhibition of cell growth
The experimental method comprises the following steps: by usingThe luminecent method evaluates compounds for inhibition of cell proliferation on NCI-H1373 cells in vitro.
The experimental steps are as follows:
1. preparing a test compound:
preparing a mother solution of a test compound: stock solution at 10mM was prepared with 100% DMSO.
Preparing a test compound gradient dilution solution: 10mM stock of test compound was taken and serially diluted 3-fold in DMSO. mu.L of DMSO-diluted compound was added to 198. Mu.L of 10% FBS-containing culture medium to prepare 10-fold test substance, the highest concentration of the test substance was 100. Mu.M, and the DMSO concentration was 1%, and 8 concentration gradients were added.
2. Test procedure
2.1 pancreatin digestion of cells grown to 80% NCI-H1373 cells, and cells were collected by centrifugation. Resuspension cells with complete medium, count and adjust seeding 96-well plate, cell seed 4000/hole/90 μl, and culture in cell incubator at 37deg.C;
2.2 day 0 (D0) plates were incubated for 24 hours for CTG detection.
2.3 simultaneously adding 10. Mu.L of 10 times of test substances with different concentrations into each well of the drug adding plate, and culturing at 37 ℃ for 6 days (D6) for CTG detection after 2-3 compound wells of each concentration.
3. CTG detection
3.1 the plates were left at room temperature in advance.
3.2 60. Mu.L of CTG solution was added per well, protected from light.
3.3 cells were lysed by shaking on an orbital shaker for 2 minutes.
3.4 cell plates were left at room temperature for 20 min.
3.5, reading the fluorescence value by using a multifunctional enzyme label instrument.
4. Data processing
Fitting the data using nonlinear S-curve regression to derive a dose-response curve, and calculating the absolute GI therefrom 50 Values.
Cell growth (%) = (D6 test object well reading-D0 reading mean)/(D6 solvent control well reading mean-D0 reading mean) ×100%
Experimental results and conclusions:
TABLE 3 in vitro cytological inhibitory Activity of the Compounds of the invention
TABLE 4 in vitro cytological inhibitory Activity of the Compounds of the invention
Test article GI 50 (nM)
H1373 cells
Compound 36 59
Compound A 43
TABLE 5 in vitro cytological inhibitory Activity of the Compounds of the invention
TABLE 6 in vitro cytological inhibitory Activity of the Compounds of the invention
Through test, the compounds in the table 1 have better inhibition effect on NCI-H1373 cell proliferation, wherein part of the compounds inhibit NCI-H1373 cell proliferationGI 50 The value was less than 100nM.

Claims (13)

  1. A compound represented by the general formula (I), a pharmaceutically acceptable salt thereof or an isomer thereof,
    wherein,
    l is selected from
    X、X 1 、X 2 Are each independently selected from-CH (R) 6 )-、-N(R 7 ) -, -O-, -S-; -C (O) -, -S (O) -or-S (O) 2 -;
    X 3 Selected from C, CH or N;
    X 4 selected from N or C (R) 8 );
    R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
    R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together form an 8-11 membered fused ring cycloalkyl, 8-11 membered fused ring heterocyclyl, 8-11 membered fused ring aryl or 8-11 membered fused ring heteroaryl optionally substituted with 1-3Q 1's; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1- 6 An alkoxy group;
    each R 4 Each R 4 ', each R 5 Each R 5 ', each R 6 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 Alkoxy, 3-8 membered cycloalkyl-C 1-6 Alkyl, 3-8 membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-7 membered heteroaryl or 5-7 membered heteroaryl-C 1-6 An alkyl group;
    each R 7 Are respectively and independently selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
    each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
    m, n, q are each independently selected from 1, 2, 3 or 4;
    virtual bond- -is a chemical bond or is absent, and X is a chemical bond 3 Is C.
  2. The compound of claim 1, a pharmaceutically acceptable salt or isomer thereof, wherein,
    R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together form an 8-11 membered fused ring heterocyclyl or an 8-11 membered fused ring heteroaryl optionally substituted with 1-3Q 1; each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
    preferably, R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together form a 9-10 membered fused ring heterocyclyl or 9-10 membered fused ring heteroaryl optionally substituted with 1-3Q 1 s.
  3. The compound of claim 1, a pharmaceutically acceptable salt or isomer thereof, wherein,
    R 2 、R 3 And R is as follows 2 Attached carbon atoms, X 3 Together form the following groups optionally substituted with 1 to 3Q 1:
    each X 3 Each X 8 、X 9 Each independently selected from CH or N;
    each X 5 Each independently selected from CH, C or N;
    each X 6 Each X 7 Are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
    Ring A is selected from 5-7 membered cycloalkyl, 5-7 membered heterocyclyl, 5-7 membered aryl or 5-7 membered heteroaryl;
    each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1- 6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
  4. The compound of claim 1, a pharmaceutically acceptable salt or isomer thereof, wherein,
    R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
    each X 3 Each X 8 、X 9 Each independently selected from CH or N;
    each X 5 Each independently selected from CH, C or N;
    each X 6 、X 7 Are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
    Ring A is selected from 5-7 membered cycloalkyl, 5-7 membered heterocyclyl, 5-7 membered aryl or 5-7 membered heteroaryl; preferably, ring a is selected from pyrrolyl, pyrazolyl, imidazolyl, furanyl, thienyl, oxazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, phenyl, pyridyl, pyrimidinyl, dihydropyrimidinyl, pyrazinyl, pyridazinyl, pyranyl, thiopyranyl, oxazinyl, azepinyl or diazaperyl;
    Each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carbonyl, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
    p is selected from 0, 1, 2 or 3.
  5. The compound, pharmaceutically acceptable salt or isomer thereof according to any of claim 1-4, wherein,
    l is selected from
    X is selected from-CH (R) 6 )-、-N(R 7 )-、-O-、-S-;
    X 1 、X 2 Are each independently selected from-CH (R) 6 )-、-N(R 7 ) -, -O-, -S-; -C (O) -, -S (O) -or-S (O) 2 -;
    X 3 Selected from C, CH or N;
    X 4 selected from N or C (R) 8 );
    R 1 Selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, fluoro C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy or fluoro C 1-6 An alkoxy group;
    each R 4 Each R 4 ', each R 5 Each R 5 ' each independently selected from H, hydroxy, amino, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1- 6 Alkoxy, halo C 1-6 Alkoxy, 3-6 membered cycloalkyl-C 1-6 Alkyl, 3-6 membered heterocyclyl, 3-6Membered heterocyclyl-C 1-6 Alkyl, phenyl-C 1-6 Alkyl, 5-6 membered heteroaryl or 5-6 membered heteroaryl-C 1-6 An alkyl group;
    each R 6 Each R 7 Are respectively and independently selected from H, C 1-6 Alkyl or halo C 1-6 An alkyl group;
    each R 8 Independently selected from H, halogen, hydroxy, amino, cyano, C 1-6 Alkyl, fluoro C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy or fluoro C 1-6 An alkoxy group;
    m, n are each independently selected from 1, 2 or 3;
    q is selected from 1 or 2;
    virtual bond- -is a chemical bond or is absent, and X is a chemical bond 3 Is C.
  6. The compound of claim 1, a pharmaceutically acceptable salt or isomer thereof, wherein,
    l isX is selected from-CH 2 -, -NH-; -O-or-S-;
    X 1 selected from-N (R) 7 ) -, -O-or-S-;
    X 2 selected from-CH (R) 6 ) -or-C (O) -;
    X 4 selected from N or CH;
    R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
    R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
    X 3 selected from CH or N;
    X 5 selected from CH, C or N;
    X 6 、X 7 are each independently selected from-CH 2 -, -NH-, -O-; -S-, -C (O) -, -S (O) -or-S (O) 2 -;
    X 8 、X 9 Are each independently selected from C (R) 9 ) Or N;
    ring a is selected from phenyl or a 5-6 membered nitrogen containing heteroaryl;
    each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
    R 4 、R 4 ’、R 6 independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy or cyano C 1-6 An alkoxy group;
    R 7 selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino groupsC 1-6 An alkyl group;
    each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
    each R 9 Independently selected from hydrogen, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
    p is selected from 0, 1 or 2;
    q is selected from 1 or 2.
  7. The compound of claim 1, a pharmaceutically acceptable salt thereof, or an isomer thereof, selected from the group consisting of:
  8. a compound represented by the general formula (I), a pharmaceutically acceptable salt thereof or an isomer thereof,
    wherein L isX is selected from-O-or-S-;
    X 1 selected from-N (R) 7 )-;
    X 2 is-C (O) -;
    X 4 is N;
    R 1 selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 Alkylthio;
    R 2 、R 3 and R is as follows 2 Attached carbon atoms, X 3 Together the following groups are formed:
    each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
    R 4 、R 4 ' are independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy or cyano C 1-6 An alkoxy group;
    R 7 selected from H, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl or amino C 1-6 An alkyl group;
    each R 8 Independently selected from H, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, C 1-6 alkoxy-C 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, cyano C 1-6 An alkoxy group;
    R 9 selected from hydrogen, halogen, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, halogenated C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkoxy C 1-6 Alkyl, halogenated C 1-6 Alkoxy, 3-6 membered cycloalkyl or 3-6 membered heterocyclyl;
    p is selected from 0, 1 or 2;
    q is selected from 1 or 2.
  9. The compound of claim 8, a pharmaceutically acceptable salt thereof, or an isomer thereof, selected from the group consisting of:
  10. a pharmaceutical composition comprising a compound of any one of claims 1-9, a pharmaceutically acceptable salt or isomer thereof, and one or more pharmaceutically acceptable excipients; preferably, the pharmaceutical composition may further comprise one or more second therapeutically active agents selected from anticancer agents, agents which reduce or decrease one or more side effects of the above-mentioned compounds, pharmaceutically acceptable salts or isomers thereof, or agents which enhance the efficacy of the above-mentioned compounds, pharmaceutically acceptable salts or isomers thereof, when used to treat a disease in a subject.
  11. Use of a compound according to any one of claims 1-9, a pharmaceutically acceptable salt or isomer thereof, a pharmaceutical composition according to claim 8 for the manufacture of a medicament for the prevention and/or treatment of diseases associated with excessive PARP7 activation.
  12. Use of a compound according to any one of claims 1-9, a pharmaceutically acceptable salt or isomer thereof, a pharmaceutical composition according to claim 8 for the manufacture of a medicament for the prevention and/or treatment of cancer, including carcinoma in situ and metastatic cancer.
  13. A kit comprising an effective amount of one or more compounds of any one of claims 1-9, pharmaceutically acceptable salts or isomers thereof; preferably, the kit may further comprise an effective amount of one or more anticancer agents.
CN202280037061.8A 2021-05-25 2022-05-25 PARP7 inhibitors Pending CN117396485A (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
CN202110569630 2021-05-25
CN2021105696300 2021-05-25
CN2021111604768 2021-09-30
CN202111160476 2021-09-30
CN202111533850 2021-12-15
CN2021115338504 2021-12-15
PCT/CN2022/094808 WO2022247839A1 (en) 2021-05-25 2022-05-25 Parp7 inhibitor

Publications (1)

Publication Number Publication Date
CN117396485A true CN117396485A (en) 2024-01-12

Family

ID=84229495

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280037061.8A Pending CN117396485A (en) 2021-05-25 2022-05-25 PARP7 inhibitors

Country Status (2)

Country Link
CN (1) CN117396485A (en)
WO (1) WO2022247839A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024037558A1 (en) * 2022-08-17 2024-02-22 Jacobio Pharmaceuticals Co., Ltd. Solid forms of compound i or salts thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3075727A1 (en) * 2017-09-18 2019-03-21 Goldfinch Bio, Inc. Pyridazinones and methods of use thereof
CA3098585A1 (en) * 2018-04-30 2019-11-07 Ribon Therapeutics Inc. Pyridazinones as parp7 inhibitors
US20220162196A1 (en) * 2019-04-29 2022-05-26 Ribon Therapeutics, Inc. Solid forms of a parp7 inhibitor
WO2021087018A1 (en) * 2019-10-30 2021-05-06 Ribon Therapeutics, Inc. Pyridazinones as parp7 inhibitors
TW202128653A (en) * 2019-10-30 2021-08-01 美商律幫治療股份有限公司 Pyridazinones as parp7 inhibitors

Also Published As

Publication number Publication date
WO2022247839A1 (en) 2022-12-01

Similar Documents

Publication Publication Date Title
CA2932425E (en) Pyrrolo[2,3-d]pyrimidinyl, pyrrolo[2,3-b]pyrazinyl and pyrrolo[2,3-d]pyridinyl acrylamides
KR102021159B1 (en) Tricyclic heterocyclic compounds and jak inhibitors
EP2629777A2 (en) Bicyclic diamines as janus kinase inhibitors
EP3068784A1 (en) Substituted 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine derivatives as casein kinase 1 d/e inhibitors
WO2013085802A1 (en) Pyrrolopyrimidines as janus kinase inhibitors
TW201011026A (en) Therapeutic agents 414
WO2021190616A1 (en) Methods for inhibiting casein kinases
WO2021190615A1 (en) Compounds as casein kinase inhibitors
CA3085460A1 (en) Aryl-bipyridine amine derivatives as phosphatidylinositol phosphate kinase inhibitors
WO2015195880A1 (en) Imidazo-pyridazne derivatives as casein kinase 1 delta/epsilon inhibitors
US20230219986A1 (en) Novel aminopyrimidine egfr inhibitor
JP7405468B2 (en) Compounds with BRD4 inhibitory activity, preparation methods and uses thereof
TWI508968B (en) Compounds for treating respiratory syncytial virus infections
TW202204358A (en) Pyrrolopyrimidine amines as complement inhibitors
KR20220063228A (en) Azole-fused pyridazin-3(2H)-one derivatives
CN115279756A (en) METTL3 modulators
WO2016045598A1 (en) 4-substituted pyrrolo[2,3-d]pyrimidine compound and use thereof
CN117396485A (en) PARP7 inhibitors
CN112584898A (en) P2X3Receptor antagonists
KR102351782B1 (en) Pyrido 5-membered aromatic ring compound, preparation method and use thereof
CN109761986B (en) Tricyclic derivative inhibitor, preparation method and application thereof
EP4313308A1 (en) Tead inhibitors and uses thereof
CN116744926A (en) Compounds as inhibitors of casein kinase
TW202214634A (en) Heterocyclic compound and derivative thereof
CN117430606A (en) Sulfonamide PARP7 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination