CN107428762B - Phthalazinone derivatives, preparation method and use thereof - Google Patents

Phthalazinone derivatives, preparation method and use thereof Download PDF

Info

Publication number
CN107428762B
CN107428762B CN201680007634.7A CN201680007634A CN107428762B CN 107428762 B CN107428762 B CN 107428762B CN 201680007634 A CN201680007634 A CN 201680007634A CN 107428762 B CN107428762 B CN 107428762B
Authority
CN
China
Prior art keywords
dihydro
pyrrolo
pyrimidine
phthalazin
carbonyl
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.)
Active
Application number
CN201680007634.7A
Other languages
Chinese (zh)
Other versions
CN107428762A (en
Inventor
刘钢
于华
任云
杜静
杨定菊
李晓勇
王坤建
刘伟
唐建川
吴勇勇
曾宏
卿燕
宋宏梅
李少华
谢一
葛勇
王利春
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech 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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN107428762A publication Critical patent/CN107428762A/en
Application granted granted Critical
Publication of CN107428762B publication Critical patent/CN107428762B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

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
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The application provides a phthalazinone derivative, a preparation method and application thereof, and specifically provides a compound shown as formula I, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, a hydrate, a solvate or a crystal form thereof, a preparation method and application thereof, wherein the compound can obviously improve the proliferation inhibition effect on tumor cells; can increase the stability of the molecule in vivo and reduce the possibility of generating toxic metabolites; or the structure of the phthalazinone compound can be modified, the oxidative metabolism capability of the compound under the action of a P450 cytochrome enzyme system in vivo is reduced, the bioavailability is improved,

Description

Phthalazinone derivatives, preparation method and use thereof
Technical Field
The application relates to the field of medicines, in particular to a phthalazinone derivative, and a preparation method and application thereof.
Background
Poly (ADP-Ribose) Polymerase (PARP) is a 113kDa multidomain protein that participates in the signaling of DNA damage by recognizing and rapidly binding to broken DNA single or double strands (D' Amurs D.et al, Poly (ADP-ribosyl) interaction in the regulation of nuclear functions, biochem. J.342: 249-268 (1999)). The PARP family now includes about 18 proteins, of which PARP-1 (the earliest discovered members) and PARP-2 are the only enzymes currently found to activate their catalytic activity by undergoing DNA cleavage, making them very unique within this family. PARP-1, which is the most typical and most studied structure in the PARP family, has a molecular weight of 114KDa and synthesizes poly (ribose-adenine diphosphate) (PAR) on receptor Proteins (including themselves) using ADP as a substrate (Sakamoto-Hojo E T, Balajee A S. targeting Poly (ADP) ribose polymerase I (PARP-I) and PARP-I interacting Proteins for cancer procedure. anticancer genes Med. Chem, 2008, 8 (4): 402-.
PARP-1 is involved in DNA damage repair and transcriptional regulation and is considered to be an important regulator of cell survival and death, and also in the regulation of several transcription factors in tumorigenesis and inflammatory responses (Peralta-Leal A, Rodriguez-Vargas J M, Aguilar-Quesada R, et al. PARP inhibitors: New ligands in the therapy of cancer and inflammatory diseases. free radial Biol. Med, 2009, 47 (l): 13-26). PARP-1 has been found to be highly expressed in various human malignancies, such as malignant lymphoma, breast cancer, Ewing's sarcoma, hepatocellular carcinoma, etc.
Since PARP-1 is involved in DNA damage repair, inhibitors of PARP-1 activity alone or in combination with DNA damaging agents may promote cancer cell death. Numerous studies have demonstrated that drug inhibition or gene knock-out of PARP-1 not only avoids tissue damage caused by oxidative stress related diseases, but also improves the prognosis of tumor patients. The PARP-1 inhibitor alone also has killing effect on tumors (mainly breast cancer) with DNA damage repair defects. In addition, the relation between PARP-1 inhibitors and angiogenesis is reported in the literature, and at least five PARP inhibitors can inhibit the proliferation and migration of human umbilical vein endothelial cells and the angiogenesis induced by vascular endothelial cell growth factor (VEGF) in vitro at present. Since the brain is susceptible to oxidative stress (Sriram C S, et al. multiple faces of poly (ADP-ribose) polymerase-1-endogenous diseases. neural Regen Res.2015; 10 (1): 49-51), neuropathological studies of PARP-1 have also been gaining increasing importance.
Therefore, the development of novel PARP inhibitors has higher clinical application value.
Disclosure of Invention
The present inventors have systematically studied the structure-activity relationship of phthalazinone compounds and have found a class of compounds having excellent PARP inhibitory activity, and have completed the present application based on the above findings.
Accordingly, a first aspect of the present application provides a compound of formula I, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing,
Figure GPA0000242631730000041
wherein the content of the first and second substances,
a and B together with the atoms to which they are attached form an alicyclic, aromatic or heteroaromatic ring, optionally wherein the alicyclic, aromatic or heteroaromatic ring is each independently substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxyl, amino, cyano, carboxyl, nitro, alkyl, alkenylAlkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, roc (O) -, rc (O), RR 'N-, rc (O) NH-, and RR' nc (O) -, optionally wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl are each independently selected by one or more (e.g., 1, 2, 3, or 4) groups from halogen, hydroxy, C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl or 5-10 membered heteroaryl;
d and E are each independently selected from C and N and together with the linking atoms form a 5-to 10-membered (e.g. 5, 6,7, 8, 9, 10) ring X, wherein the ring X is selected from a heterocyclo, aromatic and heteroaromatic ring, preferably a heteroaromatic ring, such as a pyridine, pyridazine, pyrimidine, pyrazine or triazine ring;
m and n are each independently selected from 0, 1 and 2, and m + n is 2;
y is independently selected from C, O, S and N, provided that when Y ═ O or S, R6And R7Absent, when Y ═ N, R7Is absent;
R5,R6,R7and R8Each independently selected from hydrogen, hydroxy, cyano, nitro, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RO-, RC (O), O-, RR ' N-, RC (O) NH-, and RR ' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, heteroaryl, cycloalkyl-alkyl, or heterocycloalkyl-alkyl is further independently selected from one or more (e.g., 1, 2, 3, or 4) groups selected from halogen, hydroxy, cyano, amino, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, RR ' N-, RO-C1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2; or
R5And R6Together with the Y atom, form an alicyclic, aromatic, alicyclic or heteroaromatic ring, optionally wherein each of the alicyclic, aromatic, alicyclic or heteroaromatic ringsIndependently further substituted by one or more (e.g. 1, 2, 3 or 4) groups selected from halogen, hydroxy, cyano, RS (O)a-, alkyl, RR' N-, and RO-, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) groups1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the compounds of formula I are provided with the proviso that,
when A and B together with the atoms to which they are attached form a phenyl ring, R1、R2、R3Are each hydrogen, R4Is fluorine, and m and n are both 1,
1) ring X is not a 5-membered ring, a six-membered aliphatic heterocyclic ring with heteroatom N, and a six-membered aliphatic heterocyclic ring with heteroatom N and O; and the number of the first and second electrodes,
2) the above-mentioned
Figure GPA0000242631730000051
Not a group:
Figure GPA0000242631730000052
in certain preferred embodiments of the present application, the compounds have the structure shown in formula Ia,
Figure GPA0000242631730000053
wherein the content of the first and second substances,
a and B together with the atoms to which they are attached form an alicyclic, aromatic or heteroaromatic ring, optionally wherein the alicyclic, aromatic or heteroaromatic ringEach independently substituted by one or more (e.g., 1, 2, 3, or 4) groups selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl is each independently selected from halogen, hydroxy, C (e.g., 1, 2, 3, or 4) by one or more (e.g., 1, 2, 3, or 4)1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl;
D. e, G, J, K and L are each independently selected from C and N and form a six membered aromatic ring system;
m and n are each independently selected from 0, 1 and 2, and m + n is 2;
y is selected from C, O, S and N, with the proviso that when Y ═ O or S, R6And R7Absent, when Y ═ N, R7Is absent;
R5,R6,R7and R8Each independently selected from hydrogen, hydroxy, cyano, nitro, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RO-, RC- (O) -, RC (O), O-, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, heteroaryl, cycloalkyl-alkyl, or heterocycloalkyl-alkyl is further independently substituted with one or more (e.g., 1, 2, 3, or 4) groups selected from halo, hydroxy, cyano, amino, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, nitro, haloalkyl, optionally,RO-、RR’N-、RO-C1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2NR' -wherein a is 0, 1 or 2; or
R5And R6Together with the Y atom, form an alicyclic, aromatic, alicyclic or heteroaromatic ring, optionally wherein said alicyclic, aromatic, alicyclic or heteroaromatic ring is further optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from the group consisting of halogen, hydroxy, alkyl, cyano, RS (O)a-, RR' N-, and RO-, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) groups1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the limiting conditions for the compound of formula Ia are:
when A and B together with the atoms to which they are attached form a phenyl ring, R1、R2、R3Are all hydrogen, and R4When it is fluorine, the
Figure GPA0000242631730000071
Not a group:
Figure GPA0000242631730000072
in certain preferred embodiments of the present application, the compound is of formula Iaa,
Figure GPA0000242631730000073
wherein the content of the first and second substances,
a and B together with the atoms to which they are attached form an alicyclic, aromatic or heteroaromatic ring, optionally wherein the alicyclic, aromatic or heteroaromatic ring is each independently substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl is each independently selected from halogen, hydroxy, C (e.g., 1, 2, 3, or 4) by one or more (e.g., 1, 2, 3, or 4)1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl;
D. e, G, J, K and L are each independently selected from C and N and form a six membered aromatic ring system;
y is selected from C, O, S and N, with the proviso that when Y ═ O or S, R6And R7Absent, when Y ═ N, R7Is absent;
R5,R6,R7and R8Each independently selected from hydrogen, hydroxy, cyano, nitro, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RO-, RC- (O) -, RC (O), O-, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, heteroarylcycloalkyl-alkyl, or heterocycloalkyl-alkyl is further independently selected from halogen, hydroxy, cyano, amino, carboxy, nitro, or heterocycloalkyl-alkyl by one or more (e.g., 1, 2, 3, or 4) groupsAlkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, RR' N-, RO-C1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2NR' -, wherein a is 0, 1 or 2; or
R5And R6Together with the Y atom, form an alicyclic, aromatic, alicyclic or heteroaromatic ring, optionally wherein said alicyclic, aromatic, alicyclic or heteroaromatic ring is further optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from the group consisting of halogen, hydroxy, alkyl, cyano, RS (O)a-, RR' N-, and RO-, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) groups1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the limitations of the compound of formula Iaa are: when A and B together with the atoms to which they are attached form a phenyl ring, R1、R2、R3Are all hydrogen, and R4When it is fluorine, the
Figure GPA0000242631730000081
Not a group:
Figure GPA0000242631730000082
in certain preferred embodiments of the present application, the compound is represented by formula Iaa-1,
Figure GPA0000242631730000083
wherein the content of the first and second substances,
a and B together with the atoms to which they are attached form an alicyclic, aromatic or heteroaromatic ring, optionally wherein the alicyclic, aromatic or heteroaromatic ring is each independently substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RNSO2N (R') -, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl is each independently selected from halogen, hydroxy, C (e.g., 1, 2, 3, or 4) by one or more (e.g., 1, 2, 3, or 4)1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl;
D. e, G, J, K and L are each independently selected from C and N and form a six membered aromatic ring system;
R5、R6and R8Each independently selected from hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RC (O) -, RC (O) O-, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, or heterocycloalkyl-alkyl, each independently further selected from halogen, hydroxy, cyano, amino, carboxy, nitro, C, or 4, by one or more (e.g., 1, 2, 3, or 4)1-4Alkyl, halo C1-4Alkyl radical, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, RR' N-, RO-C1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2; or
R5And R6Together with the attached N atom, form an aliphatic heterocycle or heteroaromatic ring, optionally wherein said aliphatic heterocycle or heteroaromatic ring is further optionally substituted with one or more (e.g., 1, 2, 3, or 4) substituents selected from the group consisting of halogen, hydroxy, cyano, RS (O)a-、C1-4Alkyl radical, C1-4Alkoxy and RR' N-, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) groups1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the compound is represented by formula Iaa-2,
Figure GPA0000242631730000101
wherein the content of the first and second substances,
a and B together with the atoms to which they are attached form an alicyclic, aromatic or heteroaromatic ring, optionally wherein the alicyclic, aromatic or heteroaromatic ring is each independently substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RSO2Taking of N (R') -)Substituted, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl is each independently selected from halogen, hydroxy, C (e.g., 1, 2, 3, or 4) by one or more (e.g., 1, 2, 3, or 4)1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl;
D. e, G, J, K and L are each independently selected from C and N and form a six membered aromatic ring system;
R5and R8Each independently selected from hydrogen, alkyl, cycloalkyl, cyano, nitro, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RO-, RC (O) O-, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, or heterocycloalkyl-alkyl is each independently further selected from halogen, hydroxy, amino, cyano, carboxy, nitro, C, or 41-4Alkyl, halo C1-4Alkyl radical, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, RR' N-, RO-C1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently substituted with one or more (e.g.E.g. 1, 2, 3 or 4) selected from halogen, hydroxy, C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the limitations of the compound of formula Iaa-2 are:
when A and B together with the atoms to which they are attached form a phenyl ring, R1、R2、R3Are all hydrogen, and R4When it is fluorine, the
Figure GPA0000242631730000111
Not a group:
Figure GPA0000242631730000112
in certain preferred embodiments of the present application, the compound is represented by formula Iaa-3,
Figure GPA0000242631730000113
wherein the content of the first and second substances,
a and B together with the atoms to which they are attached form an alicyclic, aromatic or heteroaromatic ring, optionally wherein the alicyclic, aromatic or heteroaromatic ring is each independently substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally, itWherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl is each independently selected from halogen, hydroxy, C, by one or more (e.g., 1, 2, 3 or 4)1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl;
D. e, G, J, K and L are each independently selected from C and N and form a six membered aromatic ring system;
R5、R6、R7and R8Each independently selected from hydrogen, hydroxy, cyano, nitro, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RO-, RC- (O) -, RC (O), O-, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, heteroarylcycloalkyl-alkyl, or heterocycloalkyl-alkyl is further independently selected from halogen, hydroxy, cyano, carboxy, nitro, amino, C, and C, by one or more (e.g., 1, 2, 3, or 4) groups1-4Alkyl, halo C1-4Alkyl radical, C1-4Alkoxy radical, C2-4Alkenyl radical, C2-4Alkynyl, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, RR' N-, RO-C1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2; or
R5And R6Together with the C atom to which they are attached form an alicyclic, aromatic, alicyclic or heteroaromatic ring, optionally wherein said alicyclic, aromatic, alicyclic or heteroaromatic ring is further optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents selected from halogen, hydroxy, cyano, RS (O)a-、C1-4Alkyl radical, C1-4Alkoxy and RR' N-, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl,5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4)1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the limitations of the compound of formula Iaa-3 are:
when A and B together with the atoms to which they are attached form a phenyl ring, R1、R2、R3Are all hydrogen, and R4When it is fluorine, the
Figure GPA0000242631730000121
Not a group:
Figure GPA0000242631730000122
in certain preferred embodiments of the present application, in the compounds of formula I
Figure GPA0000242631730000123
Selected from the following structures:
Figure GPA0000242631730000124
in certain preferred embodiments of the present application, the compound of formula I is represented by formula Iaaa,
Figure GPA0000242631730000131
wherein the content of the first and second substances,
a and B together with the linking atoms form a 3-8 membered alicyclic, 5-8 membered alicyclic, 6-10 membered aromatic or 5-10 membered heteroaromatic ring, optionally wherein the 3-8 membered alicyclic, 5-8 membered alicyclic, 6-10 membered aromatic or 5-10 membered heteroaromatic ring are each independently substituted with one or more (e.g., 1, 2, 3 or 4) groups selected from halogen, hydroxy, amino, cyano, carboxy, nitro, C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O), NH-, RR' NC (O) -, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
R1、R2、R3and R4Each independently selected from hydrogen, halogen, hydroxyl, amino, cyano, carboxyl, nitro, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, 3-10 membered cycloalkyl, 5-10 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally wherein said C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, 3-10 membered cycloalkyl, 5-10 membered heterocycloalkyl, 6-10 membered aryl or 5-10 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) substituents selected from halogen, hydroxy, C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl;
l and J are each independently selected from C and N;
y is selected from C, O, S and N, with the proviso that when Y ═ O or S, R6And R7Absent, when Y ═ N, R7Is absent;
R5,R6,R7and R8Each independently selected from hydrogen, hydroxy, cyano, nitro, C1-10Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl, 3-8 membered heterocycloalkyl-C1-4Alkyl, RO-, RC (O), O-, RR 'N-, RC (O) NH-, and RR' NC (O) -, optionally, wherein C1-10Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl or 3-8 membered heterocycloalkyl-C1-4Each alkyl group is further independently selected from halogen, hydroxy, cyano by one or more (e.g. 1, 2, 3 or 4)Amino, carboxyl, nitro, C1-4Alkyl, halo C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, RO-, RR' N-, RO-C1-4Alkyl, RR' N-C1-4Alkyl, RS (O)a-、RR’NSO2-and RSO2NR' -wherein a is 0, 1 or 2; or;
R5and R6Together with the Y atom, form a 3-8 membered alicyclic, 6-10 membered aromatic ring, 5-8 membered alicyclic ring, or 5-10 membered heteroaromatic ring, optionally wherein the 3-8 membered alicyclic, 6-10 membered aromatic ring, 5-8 membered alicyclic ring, or 5-10 membered heteroaromatic ring are each independently further substituted with one or more (e.g., 1, 2, 3, or 4) groups selected from halogen, hydroxy, cyano, RS (O)a-、C1-4Alkyl radical, C1-4Alkoxy and RR' N-, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) groups1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, the limitations of the compound of formula Iaaa are:
when A and B together with the atoms to which they are attached form a phenyl ring, R1、R2、R3Are all hydrogen, and R4When it is fluorine, the
Figure GPA0000242631730000141
Not a group:
Figure GPA0000242631730000142
in certain preferred embodiments of the present application, Y in said formula I, Ia, Iaa or Iaaa is N.
In certain preferred embodiments of the present application, said R5、R6、R7And R8Each independently selected from hydrogen, hydroxy, cyano, nitro, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanoyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 6-10 membered aryl (e.g. phenyl, naphthyl), 5-10 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl, 3-8 membered heterocycloalkyl-C1-4Alkyl, optionally, wherein said C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanoyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl or 3-8 membered heterocycloalkyl-C1-4Each alkyl group is further independently selected from halogen (e.g. fluorine), hydroxy, C by one or more (e.g. 1, 2, 3 or 4)1-4Alkyl, halo C1-4Alkyl radical, C1-4Alkoxy, 3-to 8-membered cycloalkyl, 3-to 8-membered heterocycloalkyl, 6-to 10-membered aryl, 5-to 10-membered heteroaryl, RR' N-, RO-C1-4Alkyl, RR' N-C1-4Alkyl, RR' NSO2-and RSO2-is substituted with a substituent; or
R5And R6Together with the attached Y atom form a 3-8 membered alicyclic, 6-10 membered aromatic or 5-10 membered heteroaromatic ring, optionally wherein the 3-8 membered alicyclic, 6-10 membered aromatic or 5-10 membered heteroaromatic ring are each independently further substituted with one or more (e.g., 1, 2, 3 or 4) groups selected from halogen, hydroxy, cyano, RS (O)a-、C1-4Alkyl and C1-4Alkoxy, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen and C1-4An alkyl group.
In certain preferred embodiments of the present application, said R5、R6、R7And R8Each independently selected from hydrogen, methyl, ethyl, cyclopropylmethyl, oxocyclobutylmethyl, tetrahydrofuranYlmethyl, methoxyethyl, 2-ethoxyethyl, 2-hydroxyethyl, trifluoroethyl, difluoroethyl, fluoroethyl, N-dimethylaminoethyl, (R) -1-methoxy-2-propyl, (S) -1-methoxy-2-propyl, (R) -2-methoxypropyl, (S) -2-methoxypropyl, methylsulfonylethyl, dimethylaminosulfonylethyl, 2-hydroxy-isobutyl, cyclopropyl, 1-methylcyclopropyl, 1-methoxymethylcyclopropyl, 2-dimethylcyclopropyl, 1-trifluoromethylcyclopropyl, (R) -2-fluorocyclopropyl, (S) -2-fluorocyclopropyl, 2-difluorocyclopropyl, difluorobenzyl, trifluoromethyl, oxocyclobutyl, 4-hydroxycyclohexyl, 4-difluorocyclohexyl, tetrahydropyranyl, 2-dimethyltetrahydropyranyl, 4-methyl-tetrahydropyranyl, N-methylpiperidinyl, N-dimethylaminopiperidinyl, N-methylpiperazinyl, piperidinyl, N-methylpyrazolyl and 3-oxo-bicyclo [3.1.0]Hexyl; alternatively, the first and second electrodes may be,
R5and R6Together with the attached Y atom, form a tetrahydrofuran, piperidine or piperazine ring.
In certain preferred embodiments herein, a and B together with the atoms to which they are attached form an aromatic ring.
In certain preferred embodiments herein, A and B together with the atoms to which they are attached form a 6-14 membered aromatic ring, wherein the 6-14 membered aromatic ring is unsubstituted or substituted with one or two members selected from halogen, hydroxy, amino, cyano, carboxy, nitro and C1-10Alkyl substituents.
In certain preferred embodiments of the present application, a and B together with the atoms to which they are attached form an unsubstituted phenyl ring.
In certain preferred embodiments herein, a and B together with the linking atoms form a phenyl ring, wherein the phenyl ring is substituted with one or two substituents selected from halogen.
In certain preferred embodiments herein, a and B together with the linking atoms form a phenyl ring, wherein the phenyl ring is substituted with one fluorine.
In certain preferred embodiments of the present application, R1、R2And R3Is hydrogen.
In certain preferred embodiments of the present application, R4Is halogen.
In certain preferred embodiments of the present application, R4Is fluorine.
In certain preferred embodiments of the present application, R5Selected from hydroxy, cyano, nitro, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl-alkyl, heterocycloalkyl-alkyl, RO-, RC (O), O-, RR ' N-, RC (O) NH-, and RR ' NC (O) -, optionally wherein said alkyl, cycloalkyl, heterocycloalkyl, heteroaryl, cycloalkyl-alkyl, or heterocycloalkyl-alkyl are each independently further selected from halogen, hydroxy, cyano, amino, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, RO-, RR ' N-, RO-C, by one or more (e.g., 1, 2, 3, or 4) groups1-10Alkyl, RR' N-C1-10Alkyl, RS (O)a-、RR’NSO2-and RSO2N (R') -, wherein a is 0, 1 or 2;
r and R' are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, optionally said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl are each independently selected from halogen, hydroxy, C, by one or more (e.g. 1, 2, 3 or 4) groups1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl.
In certain preferred embodiments of the present application, R5Selected from hydroxy, cyano, nitro, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, 3-20 membered cycloalkyl-C1-10Alkyl, 3-20 membered heterocycloalkyl-C1-10Alkyl, RO-, RC (O) O-, RR 'N-, RC (O) NH-, and RR' NC (O) -; optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, 3-20 membered cycloalkyl-C1-10Alkyl and 3-20 membered heterocycloalkyl-C1-10Alkyl radicals eachIndependently by one or more (e.g. 1, 2, 3 or 4) selected from halogen, hydroxy, cyano, amino, carboxy, nitro, C1-10Alkyl, halo C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, 3-20 membered cycloalkyl, 3-20 membered heterocycloalkyl, 6-20 membered aryl, 5-20 membered heteroaryl, RO-, RR' N-, RO-C1-10Alkyl, RR' N-C1-10Alkyl, RSO2-and RR' NSO2-is substituted with a substituent;
r and R' are each independently selected from hydrogen and C1-10An alkyl group; optionally, wherein said C1-10Alkyl is selected by one or more (e.g. 1, 2, 3 or 4) from halogen, hydroxy, C1-4Alkyl and C1-4Substituent of alkoxy.
In certain preferred embodiments of the present application, R5Selected from hydroxy, cyano, nitro, C1-6Alkyl, 3-12 membered cycloalkyl, 3-8 membered heterocycloalkyl, 6-14 membered aryl, 5-10 membered heteroaryl, 3-12 membered cycloalkyl-C1-6Alkyl and 3-8 membered heterocycloalkyl-C1-6An alkyl group; optionally, wherein said C1-6Alkyl, 3-12 membered cycloalkyl, 3-8 membered heterocycloalkyl, 6-14 membered heteroaryl, 5-10 membered heteroaryl, 3-12 membered cycloalkyl-C1-6Alkyl and 3-8 membered heterocycloalkyl-C1-6Each alkyl group is independently selected from halogen, hydroxy, cyano, amino, carboxy, nitro, C, by one or more (e.g., 1, 2, 3, or 4)1-6Alkyl, halo C1-6Alkyl, RO-, RR' N-, RO-C1-6Alkyl, RR' N-C1-6Alkyl, RSO2-and RR' NSO2-is substituted with a substituent;
r and R' are each independently selected from hydrogen, C1-6Alkyl and halo C1-6An alkyl group.
In certain preferred embodiments of the present application, R5Selected from hydroxy, C1-4Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-6 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl and 3-8 membered heterocycloalkyl-C1-4An alkyl group; optionally, wherein said C1-4Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-6 membered heterocycloalkylMembered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl and 3-8 membered heterocycloalkyl-C1-4Each alkyl group is independently selected from halogen, hydroxy, cyano, amino, carboxy, nitro, C, by one or more (e.g., 1, 2, 3, or 4)1-4Alkyl, halo C1-4Alkyl, RO-, RR' N-, RO-C1-4Alkyl, RR' N-C1-4Alkyl, RSO2-and RR' NSO2-is substituted with a substituent;
r and R' are each independently selected from hydrogen, C1-4Alkyl and halo C1-4An alkyl group.
In certain preferred embodiments of the present application, R5Selected from the group consisting of hydroxy, methyl, ethyl, cyclopropylmethyl, oxocyclobutylmethyl, tetrahydrofurylmethyl, methoxymethyl, methoxyethyl, 2-ethoxyethyl, 2-hydroxyethyl, trifluoroethyl, difluoroethyl, fluoroethyl, trifluoromethoxyethyl, N-dimethylaminoethyl, (R) -1-methoxy-2-propyl, (S) -1-methoxy-2-propyl, (R) -2-methoxypropyl, (S) -2-methoxypropyl, methylsulfonylethyl, dimethylaminosulfonylethyl, 2-hydroxy-isobutyl, cyclopropyl, 1-methylcyclopropyl, (R) -2-methylcyclopropyl, (S) -2-methylcyclopropyl, 1-methoxymethylcyclopropyl, oxocyclobutylmethyl, tetrahydrofurylmethyl, methoxymethyl, methoxyethyl, 2-ethoxyethyl, 2-methoxy-2-propyl, and mixtures thereof, 2, 2-dimethylcyclopropyl, 1-trifluoromethylcyclopropyl, (R) -2-fluorocyclopropyl, (S) -2-fluorocyclopropyl, 2-difluorocyclopropyl, cyclobutyl, oxocyclobutyl, tetrahydrofuranyl, 4-hydroxycyclohexyl, 4-difluorocyclohexyl, 4-methoxycyclohexyl, tetrahydropyranyl, 2-dimethyltetrahydropyranyl, 4-methyl-tetrahydropyranyl, N-methylpiperidinyl, N, n-dimethylaminopiperidinyl, N-methylpiperazinyl, piperidinyl, N-methylpyrazolyl, 2-dimethylaminomethyl-1, 3-dioxane, 5-dimethylaminomethyl-1, 3-dioxane and 3-oxo-bicyclo [3.1.0 ].]And hexyl.
In certain preferred embodiments of the present application, R5Is selected from C1-4Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-6 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl, optionally, said C1-4Alkyl, 3-8 membered cycloalkyl and 5-6 membered heteroaryl are substituted by a substituent selected from the group consisting of hydroxy, RO-and RR' N-)Substitution;
r and R' are each independently selected from C1-4An alkyl group.
In certain preferred embodiments of the present application, R5Selected from the group consisting of ethyl, 2-hydroxyethyl, dimethylaminoethyl, methoxyethyl, cyclopropyl, 1-methylcyclopropyl, 4-hydroxycyclohexyl, cyclopropylmethyl, oxocyclobutyl, tetrahydrofuranyl, tetrahydropyranyl and N-methylpyrazolyl.
In certain preferred embodiments of the present application, R6Selected from hydrogen and C1-10An alkyl group.
In certain preferred embodiments of the present application, R6Is hydrogen.
In certain preferred embodiments of the present application, R6Is selected from C1-6An alkyl group.
In certain preferred embodiments of the present application, R6Is selected from C1-4An alkyl group.
In certain preferred embodiments of the present application, R6Is methyl.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a 3-20 membered heterocyclic ring, optionally said 3-20 membered heterocyclic ring is substituted with one or more (e.g. 1, 2, 3 or 4) groups selected from halogen, hydroxy, cyano, C1-10Alkyl, RR' N-, and RO-substituents;
r and R' are each independently selected from hydrogen and C1-10An alkyl group;
y is C or N.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a 5-8 membered heterocyclic ring, optionally said 5-8 membered heterocyclic ring is substituted with one or more (e.g. 1, 2, 3 or 4) groups selected from halogen, hydroxy, cyano, C1-6Alkyl, RR' N-, and RO-substituents;
r and R' are each independently selected from hydrogen and C1-6An alkyl group;
y is C or N.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a 6-membered heterocyclic aliphatic ring, optionally substituted by one or more (e.g. 1, 2, 3 or 4) groups selected from halogen, hydroxy, cyano, C1-4Alkyl, RR' N-, and RO-substituents;
r and R' are each independently selected from hydrogen and C1-4An alkyl group;
y is N.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a piperidine or piperazine ring, optionally substituted by one or two groups selected from C1-4Alkyl and RR' N-substituent;
r and R' are each independently selected from hydrogen and C1-4An alkyl group;
y is N.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a piperidine ring and a piperazine ring, optionally substituted with one methyl or dimethylamino group;
y is N.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a 5-6 membered heterocyclic ring, optionally said 5-6 membered heterocyclic ring is substituted with one or more (e.g. 1, 2, 3 or 4) groups selected from halogen, hydroxy, cyano, C1-4Alkyl, RR' N-, and RO-substituents;
r and R' are each independently selected from hydrogen and C1-4An alkyl group;
y is C.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a tetrahydrofuran or piperidine ring, optionally substituted with one or two substituents selected from C1-4Alkyl substituent substitution;
y is C.
In certain preferred embodiments of the present application, R5And R6Together with the Y atom, form a tetrahydrofuran ring or a piperidine ring, optionally substituted with one methyl group;
y is C.
In certain preferred embodiments of the present application, R7Is hydrogen.
In certain preferred embodiments of the present application, R8Selected from hydrogen, halogen, hydroxy, cyano, nitro, C1-10Alkyl, optionally, wherein said C1-10Alkyl is substituted with one or more (e.g., 1, 2, 3, or 4) substituents selected from the group consisting of halogen, hydroxy, cyano, amino, carboxy, and nitro.
In certain preferred embodiments of the present application, R8Selected from hydrogen, hydroxy, C1-6Alkyl and halo C1-6An alkyl group.
In certain preferred embodiments of the present application, R8Selected from hydrogen, hydroxy, C1-4Alkyl and halo C1-4An alkyl group.
In certain preferred embodiments of the present application, R8Selected from hydrogen, hydroxy, methyl and trifluoromethyl.
In certain preferred embodiments of the present application,
a and B together with the linking atoms form a benzene ring;
R1、R2and R3Is hydrogen; and the number of the first and second electrodes,
R4is fluorine.
In certain preferred embodiments of the present application,
a and B together with the linking atoms form a phenyl ring, wherein said phenyl ring is substituted with one fluorine;
R1、R2and R3Is hydrogen; and the number of the first and second electrodes,
R4is fluorine.
In certain preferred embodiments of the present application,
a and B together with the linking atoms form a benzene ring;
R1、R2and R3Is hydrogen; and the number of the first and second electrodes,
R4is fluorine;
y is a nitrogen atom.
In certain preferred embodiments of the present application, the compound has the structure of formula Iaa-1, wherein,
a and B together with the linking atoms form a benzene ring;
R1、R2and R3Are all hydrogen;
R4is fluorine;
D. e, G and K are both C;
l and J are N;
R5is selected from C1-4Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-6 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl, optionally, said C1-4Alkyl, 3-8 membered cycloalkyl and 5-6 membered heteroaryl substituted with a substituent selected from the group consisting of hydroxy, RO-and RR' N-; wherein R and R' are each independently selected from C1-4An alkyl group;
R6is hydrogen;
R8selected from hydrogen and C1-4An alkyl group.
In certain preferred embodiments of the present application, the compound has the structure of formula Iaa-1, wherein,
a and B together with the linking atoms form a benzene ring;
R1、R2and R3Are all hydrogen;
R4is fluorine;
D. e, G and K are both C;
l and J are N;
R5selected from the group consisting of ethyl, 2-hydroxyethyl, dimethylaminoethyl, methoxyethyl, cyclopropyl, 1-methylcyclopropyl, 4-hydroxycyclohexyl, cyclopropylmethyl, oxocyclobutyl, tetrahydrofuranyl, tetrahydropyranyl and N-methylpyrazolyl;
R6is hydrogen;
R8selected from hydrogen and methyl.
In certain preferred embodiments herein, the hydrogen described herein is protium (H) or deuterium (D).
A compound according to the first aspect of the present application, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, wherein the compound is selected from:
4- [3- (2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (2-cyclopropylmethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-hydroxyethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (2-dimethylaminoethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- [3- (2-dimethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-methoxyethylamine) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-hydroxy-2-methylpropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- {3- [2- (cyclopropylmethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluoro-benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (3-oxetanylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-hydroxycyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydrofuran-3-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (4-dimethylaminopiperidin-1-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-methylpiperazin-1-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methylpiperidin-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (4, 4-difluorocyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methyl-1H-pyrazol-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methyl-1H-pyrazol-3-amino) -5, 7-dihydro-pyrazolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (methylmethoxyethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- [3- (2-ethoxyethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
(S) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(R) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(S) -4- { 4-fluoro-3- [2- (2-methoxypropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(R) -4- { 4-fluoro-3- [2- (2-methoxypropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydrofuran-3-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydrofuran-2-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methylpiperidin-4-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (piperidin-4-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- [3- (2-cyclopropylmethyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2R) -2-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclobutylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclopropylmethoxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-hydroxypropan-2-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methoxymethylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2S) -2-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclopropylamino-4-methyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-trifluoromethylcyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (2, 2-difluorocyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- {3- [2- (2, 2-dimethylcyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2S) -2-fluorocyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2R) -2-fluorocyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-methylsulfonylethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
n, N-dimethyl-2- {6- [ 2-fluoro-5- (4-oxo-3, 4-dihydrophthalazin-1-methyl) benzoyl ] -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amino } -ethanesulfonamide;
4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-b ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (6-cyclopropylamino-1, 3-dihydro-pyrrolo [3,4-c ] pyridine-2-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-b ] pyrazine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- {3- [2- (2, 2-dimethyl-tetrahydropyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-methyl-tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (3-azabicyclo [3.1.0] hexyl-6-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2,2, 2-trifluoroethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (2, 2-difluoroethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-fluoroethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (oxetanyl-2-methyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (oxetanyl-3-methyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydrofuran-2-methyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (cyclopropylamino) -4-trifluoromethyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-trifluoromethoxyethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -7-fluoro-2H-phthalazin-1-one;
7-fluoro-4- { 4-fluoro-3- [2- (2-methoxyethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
7-fluoro-4- { 4-fluoro-3- [2- (tetrahydropyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-methoxycyclohexyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(R) -4- {3- [2- (3-tetrahydrofuryl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one;
(S) -4- {3- [2- (3-tetrahydrofuryl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one;
4- {3- [2- (((2R,5R) -5-dimethylamino-1, 3-dioxan-2-yl) methyl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one;
4- {3- [2- ((2R,5R) -2- (dimethylamino) methyl-1, 3-dioxan-5-yl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one; and the combination of (a) and (b),
4- {3- [ 4-methyl-2- (4-tetrahydrofuryl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one.
In another aspect, the present application provides a process for the preparation of said compound selected from the following synthetic routes:
route one:
Figure GPA0000242631730000241
carrying out condensation reaction on the compound A and the compound B to obtain a compound shown in a formula I; or
And a second route:
Figure GPA0000242631730000242
carrying out condensation reaction on the compound A and the compound B 'to obtain a compound C'; reacting compound C' with a compound
Figure GPA0000242631730000243
Carrying out nucleophilic substitution reaction to prepare a compound shown in a formula I;
wherein Lg represents a leaving group for nucleophilic substitution, e.g. halogen, -OCOR, -OTs, -SO2R, etc., for example, is methylsulfonyl, and the remaining atoms and substituents are as defined above.
In certain preferred embodiments herein, the condensation reaction is conducted as follows: dissolving 0.8-1.2 equivalents of compound A and 1.0 equivalent of compound B or compound B '(free amine or hydrochloride) in a solvent (such as DMF or THF), adding 0.8-1.2 equivalents of a condensing agent (such as HATU, EDCI, T3P, etc.) and 2.0-5.0 equivalents of a base (such as DIPEA, Et3N, pyridine, etc.) at room temperature, adding water and an organic solvent (such as ethyl acetate or methyl tert-butyl ether) to the reaction solution after the reaction is finished, separating, extracting, drying and spin-drying, and purifying the crude product by preparative thin layer chromatography or column chromatography to obtain the target compound of formula I or compound C'.
In certain preferred embodiments of the present application, the nucleophilic substitution reaction is conducted as follows: dissolving the compound C' (1.0eq.) in an organic solvent (such as acetonitrile, etc.), adding a nucleophilic reagent (1.0-1.5eq.) in the presence of a base (such as potassium carbonate, sodium carbonate, triethylamine, DIPEA, etc., 1.0-3.0eq.), and reacting at room temperature or under heating (40-120 ℃) to obtain the compound of the formula I.
In certain preferred embodiments of the present application, the method comprises the steps of:
Figure GPA0000242631730000251
condensation of compound a with compound 2B gives compounds of formula Ia, wherein the atoms and substituents are as defined above.
In certain preferred embodiments of the present application, the method comprises the steps of:
Figure GPA0000242631730000252
condensation reaction of compound a with compound 3B gives the compound of formula Iaa, wherein the atoms and substituents are as defined above.
In certain preferred embodiments of the present application, the method comprises the steps of:
Figure GPA0000242631730000253
condensation reaction of compound a with compound 4B gives a compound of formula Iaaa wherein the atoms and substituents are as defined above.
Compounds B or B' are commercially available or can be prepared by routine experimentation in the art. In certain preferred embodiments of the present application, as an example of the synthesis of 2-pyrimidine derivatives, the method of synthesis of compound B or B' includes, but is not limited to, the following methods:
the method comprises the following steps:
Figure GPA0000242631730000261
reacting 3-pyrrolidone with amino protected by P group with DMF-DMA under heating (such as 60-120 deg.C), concentrating under reduced pressure to remove DMF-DMA, and washing the obtained solid with solvent (such as methyl tert-butyl ether, tetrahydrofuran, diethyl ether or acetonitrile) to obtain the first step reaction product; the product is reacted with methylthioamidine sulfate in a solvent (such as ethanol, methanol or isopropanol) to obtain a ring-closing product; oxidizing the ring-closing product with an oxidant (such as m-CPBA, water peroxide or peroxyacetic acid) in a solvent (such as dichloromethane, acetonitrile or DMF) to obtain a sulfone intermediate; the sulfone intermediate and amine react at 20-100 ℃ to obtain the derivative of 2-aminopyrimidine (the reaction can use no solvent, and can also use solvents such as DMF or DMSO); finally removing the protecting group on N (such as trifluoroacetic acid or hydrochloric acid) to obtain the 2-pyrimidine derivative;
in certain preferred embodiments herein, the P group is an amino protecting group.
The second method comprises the following steps:
Figure GPA0000242631730000262
reacting 3-pyrrolidone with amino protected by P group with DMF-DMA under heating (such as 60-120 deg.C), concentrating under reduced pressure to remove DMF-DMA, and washing the obtained solid with solvent (such as methyl tert-butyl ether, tetrahydrofuran, diethyl ether or acetonitrile) to obtain the first step reaction product; reacting the product with urea at an alkaline condition at room temperature or under heating to obtain a 2-hydroxypyrimidine derivative intermediate, and reacting the intermediate with phosphorus oxychloride to obtain a 2-chloropyrimidine derivative; the resulting product and amine are then reacted at room temperature or elevated temperature using a palladium-containing catalyst (e.g., Pd (dppf) Cl) to provide the 2-aminopyrimidine derivative2Dichloromethane complex or Pd (OAc)2) Ligands (e.g. BINAP, Xantphos or Brettphospate) and bases (e.g. DIPEA, Cs)2CO3Or NaOt-Bu) to promote the reaction (the reaction may be carried out without a solvent, or with, for example, toluene, dioxaneSolvents such as cyclic, DMF, or DMSO). Finally removing the protecting group (such as trifluoroacetic acid or hydrochloric acid) on N to obtain the 2-pyrimidine derivative.
The third method comprises the following steps:
Figure GPA0000242631730000271
reacting 3-pyrrolidone with amino protected by P group with DMF-DMA under heating (such as 60-120 deg.C), concentrating under reduced pressure to remove DMF-DMA, and washing the obtained solid with solvent (such as methyl tert-butyl ether, tetrahydrofuran, diethyl ether or acetonitrile) to obtain the first step reaction product; reacting the product with urea under alkaline conditions to obtain a 2-hydroxypyrimidine derivative intermediate; reacting this intermediate with trifluoromethanesulfonic anhydride to give a trifluoromethanesulfonate intermediate, reacting this intermediate with an amine to give a 2-aminopyrimidine derivative, using a palladium-containing catalyst (e.g. Pd (dppf) Cl2 dichloromethane complex or Pd (OAc))2) Ligands (e.g. xanthphos or brettphospate) and bases (e.g. Cs)2CO3Or NaOt-Bu) to promote the reaction (the reaction may be carried out without a solvent, or with a solvent such as toluene, dioxane, DMF or DMSO), and then removing the protecting group from N to obtain the 2-pyrimidine derivative.
Or, the method four:
Figure GPA0000242631730000272
reacting 3-pyrrolidone with amino group protected by P group with DMF-DMA under heating (such as 60-120 deg.C), and concentrating under reduced pressure to remove DMF-DMA; washing the obtained solid with a solvent (such as methyl tert-butyl ether, tetrahydrofuran, diethyl ether or acetonitrile) to obtain the reaction product of the first step; the 2-pyrimidine derivatives can also be obtained by reacting this intermediate with substituted guanidines under basic conditions.
In another aspect, the present application provides a pharmaceutical composition comprising a compound described herein, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, optionally further comprising a pharmaceutically acceptable carrier or excipient.
In certain preferred embodiments of the present application, the compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or isomer, hydrate, solvate or crystal form of the foregoing may also be used in combination with one or more drugs. Thus, in certain preferred embodiments of the present application, the pharmaceutical composition further comprises one or more drugs. In certain preferred embodiments of the present application, the drug is an anti-tumor drug. In certain preferred embodiments of the present application, the antineoplastic agent is selected from the group consisting of temozolomide, doxorubicin, paclitaxel, cisplatin, carboplatin, dacarbazine, topotecan, irinotecan, gemcitabine, bevacizumab, anti-CTLA-4 mab Iplilimumab, anti-PD-1 mab pembrolizumab and Nivolumab, and anti-PD-L1 mab atezolizumab.
In certain preferred embodiments of the present application, the vector includes, but is not limited to: aluminium oxide, aluminium stearate, lecithin, serum proteins such as human serum albumin, phosphates, glycerol, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, protamine sulphate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulosic substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, beeswax, lanolin. The excipient refers to an additive in the medicinal preparation except the main medicament. The composition has stable properties, no incompatibility with main drug, no side effect, no influence on curative effect, no deformation at room temperature, no crack, mildew, moth-eaten feeling, no harm to human body, no physiological effect, no chemical or physical effect with main drug, no influence on content determination of main drug, etc. Such as binders, fillers, disintegrants, lubricants in tablets; wine, vinegar, medicinal juice, etc. in the Chinese medicinal pill; base portion in semisolid formulations ointments, creams; preservatives, antioxidants, flavoring agents, fragrances, solubilizers, emulsifiers, solubilizers, tonicity adjusting agents, colorants and the like in liquid preparations can all be referred to as excipients.
The compounds of the present application, their esters, prodrugs, isomers, hydrates, solvates, crystal forms, pharmaceutically acceptable salts, their metabolite forms, or any combination or mixture of the above, or pharmaceutical compositions thereof, may be administered by the following routes: parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular routes, or as inhalants.
The compounds of the present application, their esters, prodrugs, isomers, hydrates, solvates, crystal forms, pharmaceutically acceptable salts, their metabolite forms, or any combinations or mixtures of the foregoing, or pharmaceutical compositions thereof, may be formulated into various suitable dosage forms depending on the route of administration.
When administered orally, the compounds of the present application may be formulated in any orally acceptable dosage form, including but not limited to tablets, capsules, aqueous solutions or suspensions. Among these, carriers for tablets generally include lactose and corn starch, and additionally, lubricating agents such as magnesium stearate may be added. Diluents used in capsule formulations generally include lactose and dried corn starch. Aqueous suspension formulations are generally prepared by mixing the active ingredient with suitable emulsifying and suspending agents. Optionally, some sweetener, aromatic or colorant may be added into the above oral preparation.
When applied topically to the skin, the compounds of the present application can be formulated in a suitable ointment, lotion, cream, or like formulation in which the active ingredient is suspended or dissolved in one or more carriers. Carriers that may be used in ointment formulations include, but are not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyethylene oxide, polypropylene oxide, emulsifying wax and water; carriers that can be used in lotions or creams include, but are not limited to: mineral oil, sorbitan monostearate, tween 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
The compounds of the present application may also be administered in the form of sterile injectable preparations, including sterile injectable aqueous or oleaginous suspensions or solutions, and may also be in lyophilized form. Among the carriers and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, the sterilized fixed oil may also be employed as a solvent or suspending medium, such as a monoglyceride or diglyceride.
The pharmaceutical preparation of the present application includes any preparation that can be pharmaceutically implemented, such as an oral preparation, a parenteral preparation, and the like.
The pharmaceutical compositions and preparations of the present application may contain 0.01 to 2000mg of a compound of the present application, preferably 0.1 to 1000mg of a compound of the present application, preferably 1 to 800mg of a compound of the present application, more preferably 10 to 600mg of a compound of the present application, and particularly preferably 50 to 500mg of a compound of the present application.
In embodiments of the present application, suitable in vitro or in vivo assays are performed to determine the effectiveness of the compositions of the present application and whether administration is suitable for treating a disease or medical condition in an individual. Examples of such assays are described below in the non-limiting examples in connection with specific diseases or medical treatments. Generally, an effective amount of a composition of the present application sufficient to achieve a prophylactic or therapeutic effect is from about 0.001 mg/kg body weight/day to about 10,000 mg/kg body weight/day. Suitably, the dose is from about 0.01 mg/kg body weight/day to about 1000mg/kg body weight/day. The dosage range may be about 0.01 to 1000mg/kg of host body weight per day, every second day, or every third day, more usually 0.1 to 500mg/kg of host body weight. Exemplary treatment regimens are once every two days or once a week or once a month. The agent is typically administered multiple times, and the interval between single doses may be daily, weekly, monthly or yearly. Alternatively, the agent may be administered in a sustained release formulation, in which case less frequency of administration is required. The dose and frequency will vary depending on the half-life of the agent in the subject. It may also vary depending on whether prophylactic or therapeutic treatment is carried out. In prophylactic applications, relatively low doses are administered chronically at relatively infrequent intervals. In therapeutic applications, it is sometimes desirable to administer relatively high doses at relatively short intervals until the progression of the disease is delayed or halted, and preferably until the individual exhibits a partial or complete improvement in the symptoms of the disease, after which a prophylactic regimen can be administered to the patient.
In another aspect, the present application provides the use of the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein, in the manufacture of an agent that inhibits PARP.
In certain preferred embodiments of the present application, the agent is an agent that inhibits PARP-1.
In another aspect, the present application also provides a method of inhibiting PARP activity comprising administering to a cell in need thereof an effective amount of a compound described herein, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In certain preferred embodiments of the present application, the methods are used to inhibit PARP-1 activity.
In certain preferred embodiments of the present application, the methods are used to inhibit PARP-1 activity in a cell.
In certain preferred embodiments of the present application, the cell is a cell line or a cell from a subject.
In another aspect, the present application provides the use of the compound, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein, in the manufacture of an agent for the adjuvant treatment of a tumor or a medicament for enhancing the effect of radiation or chemotherapy.
In another aspect, the present application provides the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein for use in the adjuvant treatment of a tumor or for use in enhancing a radiation or chemotherapy effect.
In another aspect, the present application provides a method of adjunctive treatment of a tumor or for enhancing the effects of radiation or chemotherapy, comprising administering to a subject in need thereof an effective amount of a compound described herein, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In another aspect, the present application provides the use of the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein, in the manufacture of a medicament for the treatment of a tumor.
In another aspect, the present application provides the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, for use in the treatment of a tumor.
In another aspect, the present application provides a method of treating a tumor, the method comprising providing to a subject in need thereof an effective amount of a compound described herein, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In another aspect, the present application provides the use of the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein, in the manufacture of a medicament for the treatment of a vascular disease, a neurodegenerative disease, or nervous system inflammation.
In another aspect, the present application provides the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein for use in the treatment of a vascular disease, a neurodegenerative disease, or nervous system inflammation.
In another aspect, the present application provides a method of treating a vascular disease, a neurodegenerative disease, or nervous system inflammation, the method comprising administering to a subject in need thereof an effective amount of a compound described herein, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In another aspect, the present application provides the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, for use in inhibiting PARP activity in a cell.
In certain preferred embodiments of the present application, the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein is used to inhibit PARP-1 activity in a cell.
In certain preferred embodiments of the present application, the cell is a cell line or a cell from a subject.
In certain preferred embodiments of the present application, the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, is used in an in vivo method.
In certain preferred embodiments of the present application, the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, is used in an in vitro method.
In another aspect, the present application also provides a method of inhibiting PARP activity in a cell, comprising administering to the cell an effective amount of a compound described herein, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In certain preferred embodiments of the present application, the methods are used to inhibit PARP-1 activity.
In certain preferred embodiments of the present application, the cell is a cell line or a cell from a subject.
In certain preferred embodiments of the present application, the method is performed in vivo.
In certain preferred embodiments of the present application, the method is performed in vitro.
In another aspect, the present application also provides the use of the compound, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein, for the manufacture of an agent as an agent that assists or enhances the effect of a compound, prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or isomer, hydrate, solvate or crystal form of the foregoing in inhibiting tumor cell proliferation.
In certain preferred embodiments of the present application, the tumor cell is a tumor cell line or a tumor cell from a subject.
In certain preferred embodiments of the present application, the agent is used in an in vivo method.
In certain preferred embodiments of the present application, the reagents are used in vitro methods.
In another aspect, the present application also provides the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, or a pharmaceutical composition described herein for use in assisting in inhibiting tumor cell proliferation or for enhancing the effect of inhibiting tumor cell proliferation by radiation or chemical means.
In certain preferred embodiments of the present application, the tumor cell is a tumor cell line or a tumor cell from a subject.
In certain preferred embodiments of the present application, the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition of the present application, is used in an in vivo method.
In certain preferred embodiments of the present application, the compound, ester, prodrug, isomer, hydrate, solvate, crystalline form, pharmaceutically acceptable salt thereof, metabolite form thereof, or any combination or mixture of the foregoing, or a medicament as described herein, is used in an in vitro method.
In another aspect, the present application also provides a method of aiding in the inhibition of tumor cell proliferation or enhancing the effect of radiation or chemical means on the inhibition of tumor cell proliferation, the method comprising administering to a tumor cell an effective amount of a compound described herein, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In certain preferred embodiments of the present application, the tumor cell is a tumor cell line or a tumor cell from a subject.
In certain preferred embodiments of the present application, the method is performed in vitro.
In certain preferred embodiments of the present application, the method is performed in vivo.
In another aspect, the present application also provides the use of the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, in the manufacture of an agent for inhibiting tumor cell proliferation.
In certain preferred embodiments of the present application, the agent is used in an in vivo method.
In certain preferred embodiments of the present application, the reagents are used in vitro methods.
In another aspect, the present application also provides the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, for use in inhibiting tumor cell proliferation.
In certain preferred embodiments of the present application, the tumor cell is a tumor cell line or a tumor cell from a subject.
In certain preferred embodiments of the present application, the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, is used in an in vivo method.
In certain preferred embodiments of the present application, the compound, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein, is used in an in vitro method.
In another aspect, the present application also provides a method of inhibiting tumor cell proliferation, comprising administering to a cell an effective amount of a compound described herein, a prodrug, metabolite form, pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form of the foregoing, or a pharmaceutical composition described herein.
In certain preferred embodiments of the present application, the tumor is selected from the group consisting of breast cancer, ovarian cancer, colorectal cancer, melanoma, lung cancer, gastrointestinal stromal tumor, brain cancer, cervical cancer, pancreatic cancer, prostate cancer, stomach cancer, chronic myeloid leukemia, liver cancer, lymphoma, peritoneal cancer, soft tissue sarcoma, neuroendocrine tumors, and glioblastoma.
In certain preferred embodiments of the present application, the tumor cell is selected from the group consisting of a breast cancer cell, an ovarian cancer cell, a colorectal cancer cell, a melanoma cell, a lung cancer cell, a gastrointestinal stromal tumor cell, a brain cancer cell, a cervical cancer cell, a pancreatic cancer cell, a prostate cancer cell, a stomach cancer cell, a chronic myeloid leukocyte cell, a liver cancer cell, a lymphoma cell, a peritoneal cancer cell, a soft tissue sarcoma cell, a neuroendocrine tumor cell, and a glioblastoma cell.
In certain preferred embodiments of the present application, the tumor cell is a tumor cell line or a tumor cell from a subject.
Tumors described herein include both malignant and benign tumors, and correspondingly, tumor cells include both malignant and benign tumor cells.
The subject described herein is a mammal, e.g., bovine, equine, ovine, porcine, canine, feline, rodent, primate; among these, particularly preferred subjects are humans.
The terms of the present application are explained below, and for specific terms, if the meaning in the present application is inconsistent with the meaning commonly understood by those skilled in the art, the meaning in the present application controls; if not defined in the present application, have the meaning commonly understood by a person skilled in the art. Unless stated to the contrary, terms used in the present application have the following meanings.
As used herein, the term "hydrogen" and hydrogen in each of the groups described includes protium (H), deuterium (D), tritium (T). In certain preferred embodiments herein, the hydrogen is protium (H).
The term "alkyl" as used herein refers to a straight or branched chain saturated hydrocarbon group, e.g. C1-10Alkyl radical, C1-6Alkyl or C1-4Non-limiting examples of alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl and the like.
The alkyl is unsubstituted or may be further substituted with a substituent to form a substituted alkyl, which substituent may be selected from halo, cycloalkyl, heterocycloalkyl, RO-or RR 'N-, wherein R and R' are as defined herein.
When the substituent is halogen (e.g., fluorine), the substituted alkyl is a haloalkyl as described herein, including halo C1-6Alkyl (e.g. fluoro C)1-6Alkyl) and halo C1-4Alkyl (e.g. fluoro C)1-4Alkyl), and the like.
When the substituent is cycloalkyl, the substituted alkyl is cycloalkyl-alkyl as described herein, including 3-20 membered cycloalkyl-C1-10Alkyl, 3-12 membered cycloalkyl-C1-6Alkyl, 3-8 membered cycloalkyl-C1-4Alkyl, 3-membered cycloalkyl-methyl, 4-memberedCycloalkyl-methyl, 5-membered cycloalkyl-methyl, 6-membered cycloalkyl-methyl, 7-membered cycloalkyl-methyl, 8-membered cycloalkyl-methyl and the like.
When the substituent is heterocycloalkyl, the substituted alkyl is heterocycloalkyl-alkyl as described herein, including 3-20 membered heterocycloalkyl-C1-10Alkyl, 3-8 membered heterocycloalkyl-C1-6Alkyl, 3-8 membered heterocycloalkyl-C1-4Alkyl, 3-membered heterocycloalkyl-methyl, 4-membered heterocycloalkyl-methyl, 5-membered heterocycloalkyl-methyl, 6-membered heterocycloalkyl-methyl, 7-membered heterocycloalkyl-methyl, 8-membered heterocycloalkyl-methyl and the like.
When the substituent is RO-, the substituted alkyl group is RO-alkyl, including, for example, RO-C1-10Alkyl, RO-C1-6Alkyl, RO-C1-4Alkyl, RO-methyl, RO-ethyl, and the like, wherein the R groups are as described herein. In certain preferred embodiments of the present application, R is hydrogen or C1-10Alkyl, thus, the RO-alkyl group may be a hydroxyl-substituted alkyl group or C1-10An alkoxyalkyl group. The hydroxy-substituted alkyl group includes hydroxy C1-10Alkyl, hydroxy C1-6Alkyl and hydroxy C1-4Alkyl groups, and the like. The C is1-10Alkoxyalkyl radicals including C1-6alkoxy-C1-6Alkyl and C1-4alkoxy-C1-4Alkyl groups, and the like.
When the substituent is RR ' N-, the substituted alkyl is RR ' N-alkyl, including, for example, RR ' N-C1-10Alkyl, RR' N-C1-6Alkyl, RR' N-C1-4Alkyl, RR ' N-methyl, and RR ' N-ethyl, and the like, wherein the R and R ' groups are as described herein. In certain preferred embodiments of the present application, R and R' are hydrogen or C1-10Alkyl, thus, the RR' N-alkyl may be amino-substituted alkyl or C1-10Alkylaminoalkyl or di-C1-10An alkylaminoalkyl group. The RR' N-alkyl group is, for example, amino-substituted C1-10Alkyl, amino substituted C1-6Alkyl, amino substituted C1-4Alkyl radical, C1-6alkylamino-C1-6Alkyl radical, C1-4alkylamino-C1-4Alkyl, di-C1-6alkylamino-C1-6Alkyl and di-C1-4alkylamino-C1-4Alkyl groups, and the like. The term "alkenyl" as used herein refers to a straight or branched chain hydrocarbon group containing at least one carbon-carbon double bond, such as C2-10Alkenyl radical, C2-6Alkenyl or C2-4Alkenyl, non-limiting examples of alkenyl include ethenyl, propenyl, butenyl, 2-methylpropenyl, pentenyl, 2-methylbutenyl, 3-methylbutenyl, hexenyl, 2-methylpentene, 3-methylpentene, 4-methylpentene, 2-ethylbutenyl, and the like. The term "alkynyl" as used herein refers to a straight or branched chain hydrocarbon radical containing at least one carbon-carbon triple bond, e.g. C2-10Alkynyl, C2-6Alkynyl or C2-4Non-limiting examples of alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, 3-methylbutynyl, hexynyl, 3-methylpentylynyl, and the like. The term "alkoxy" as used herein refers to a group having the structure "alkyl-O-", e.g., C1-10Alkoxy radical, C1-6Alkoxy or C1-4Alkoxy, non-limiting examples of alkoxy include methoxy, ethoxy, propoxy, isopropoxy, tert-butoxy and the like.
The formula "ROC (O) -" as used herein refers to a substituted oxycarbonyl group, such as an alkoxycarbonyl group. Non-limiting examples of R-OC (O) -include methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, tert-butoxycarbonyl, benzyloxyformyl, and the like.
As used herein, the formula "RC (O) O-" refers to a substituted acyloxy group, such as an alkanoyloxy group. Non-limiting examples of R-C (O) O-include formyloxy, acetyloxy, propionyloxy, benzoyloxy and the like.
The formula "RC (O) -" as used herein refers to a substituted acyl group, such as alkanoyl. Non-limiting examples of R-C (O) -include acetyl, propionyl, butyryl, benzoyl, and the like.
The formula "RR' NH-" as used herein refers to a substituted amino group, such as alkylamino, dialkylamino, and the like. Non-limiting examples of RR' NH-include methylamino, ethylamino, propylamino, N-dimethylamino, N-diethylamino, and the like.
As used herein, the formula "RC (O) NH-" refers to substituted amido groups, such as alkanoylamino and arylamido groups, and the like. Non-limiting examples of R-C (O) NH-include formylamino, acetylamino, benzoylamino and the like.
The formula "RR' NHC (O) -" as used herein refers to substituted aminoacyl groups such as alkylaminoacyl and dialkylaminoacyl groups and the like. Non-limiting examples of RR' NHC (O) -include methyl aminoacyl, ethyl aminoacyl, N-two methyl aminoacyl, N-two acetyl aminoacyl etc..
As used herein, the formula "RS (O)a- ", where a ═ 0, 1 or 2, denotes substituted mercapto, sulfonyl or sulfinyl radicals, RS (O)aNon-limiting examples of (A-B) include mercapto, methylsulfonyl, ethylsulfonyl, phenylsulfonyl, p-methylphenylsulfonyl and the like.
The chemical formula "RR' NSO as used herein2- "denotes a substituted aminosulfonyl group, RR' NSO2Non-limiting examples of (A) - (B) include dimethylaminosulfonyl and the like.
Chemical formula "RSO" as used herein2N (R') - "refers to substituted sulfonamide groups, such as alkylsulfonamide groups, arylsulfonamide groups, and the like. RSO2Non-limiting examples of N (R') -, include methanesulfonamide group, ethanesulfonamido group, benzenesulfonamide group, p-toluenesulfonamide group and the like.
As used herein, "R" and "R'" are each independently selected from hydrogen, hydroxy, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocycloalkyl, 6-20 membered aryl and 5-20 membered heteroaryl. In certain preferred embodiments of the present application, said R and R' are hydrogen or C1-10Alkyl when a substituent referred to herein contains R or R ', it includes all R or R' as defined above in accordance with the valence rules.
The term "cycloalkyl" as used herein refers to monocyclic or polycyclic cyclic alkyl groups, such as cycloalkyl groups containing 3 to 20 carbon atoms, such as cycloalkyl groups containing 3 to 12 carbon atoms, such as cycloalkyl groups containing 3 to 10 carbon atoms, such as cycloalkyl groups containing 3 to 8 carbon atoms, such as cycloalkyl groups containing 5 to 8 carbon atoms. Non-limiting examples of monocycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl cyclooctyl, and the like. Polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups. The term "heterocycloalkyl" as used herein refers to a cycloalkyl group as described hereinbefore comprising at least one heteroatom selected from N, O and S, such as a heterocycloalkyl containing one or two N atoms, a heterocycloalkyl containing one O atom, a heterocycloalkyl containing one S atom, such as comprising 3-20 membered heterocycloalkyl, such as comprising 5-20 membered heterocycloalkyl, such as comprising 3-8 membered heterocycloalkyl, such as comprising 5-8 membered heterocycloalkyl. Non-limiting examples of heterocycloalkyl groups include glycidylalkyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like. The term "aryl" as used herein refers to groups having aromatic character, preferably 6-14 membered aryl, more preferably 6-10 membered aryl, which are non-limiting examples including, but not limited to, phenyl and naphthyl, and the like, either 6-20 membered monocyclic or fused polycyclic (e.g., bicyclic or tricyclic). The term "heteroaryl" as used herein refers to a 5-14 membered aromatic heterocyclic group, preferably 5-10 membered, substituted with at least one heteroatom selected from N, O or S. For example a heteroaryl group containing one or two N atoms, a heteroaryl group containing one O atom or a heteroaryl group containing one S atom. Non-limiting examples of heteroaryl groups include furyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, triazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl and the like.
The term "alicyclic" as used herein refers to a saturated or partially saturated carbocyclic ring having no aromatic character. Examples include 3-20 membered alicyclic ring, 3-12 membered alicyclic ring, 3-8 membered alicyclic ring, 4 membered alicyclic ring, 5 membered alicyclic ring, 6 membered alicyclic ring, 7 membered alicyclic ring, etc.
The term "lipoheterocycle" as used herein refers to an aliphatic cyclic group wherein at least one ring member is a heteroatom selected from N, O and S. For example, an aliphatic heterocyclic ring containing 1 or 2N atoms, for example, an aliphatic heterocyclic ring containing only one O atom, for example, an aliphatic heterocyclic ring containing only one S atom, and the like. For example, 3-20 membered, 3-12 membered, 3-8 membered, 4 membered, 5 membered, 6 membered, 7 membered, etc. are included.
The term "aromatic ring" as used herein refers to an aromatic ring in which all ring members are carbon atoms. For example, 6-20 membered aromatic rings, 6-14 membered aromatic rings, 6-10 membered aromatic rings, and the like are included.
The term "heteroaromatic ring" as used herein refers to a cyclic group having aromatic character with at least one ring member being a heteroatom selected from N, O and S. For example, heteroaromatic rings containing 1 or 2N atoms, such as heteroaromatic rings containing only one O atom, such as heteroaromatic rings containing only one S atom, and the like. For example, including 5-20 membered heteroaromatic rings, 5-14 membered heteroaromatic rings, 5-10 membered heteroaromatic rings, 5-6 membered heteroaromatic rings, and the like.
The term "halogen" as used herein includes fluorine, chlorine, bromine and iodine.
In the compounds of formula I as described herein, wherein
Figure GPA0000242631730000371
Non-limiting examples of (a) include:
Figure GPA0000242631730000372
Figure GPA0000242631730000381
the condensed ring may be substituted by R5R6N-group substitution, the substitution position being selected in accordance with the valence bond theory.
It will be understood by those skilled in the art that resonant structures according to valence theory are also within the scope of the present application when a conjugated system is present in the molecular structure of the compounds described herein, for example,
Figure GPA0000242631730000382
formula a, formula b and formula c are all within the scope of protection of the present application.
The term "isomer" as used herein includes all possible isomeric forms of the compounds of general formula I herein, such as enantiomers, diastereomers, epimers, cis-trans isomers, conformers, and the like. For example, enantiomers in the R and S configurations, cis and trans isomers in the Z and E configurations, and the like are within the scope of the present application.
The compounds of formula I or pharmaceutically acceptable salts thereof herein may also form solvates, such as hydrates, alcoholates and the like.
The compounds of formula I of the present application may also be prodrugs or forms which release the active ingredient after metabolic changes in the body. The selection and preparation of suitable prodrugs is well within the skill of those in the art.
The compound of formula I or a pharmaceutically acceptable salt thereof may also exist as crystals, which means an arrangement in which molecules, atoms, or ions constituting the compound are spatially repeated at a regular period, and the arrangement has a periodicity of three-dimensional space, and is repeated at a certain distance. The compounds can exist in two or more crystalline states, and molecules with the same structure are crystallized into different solid forms, which are called polymorph, i.e. polymorphic forms, polymorphic forms and the like. When referring to a particular crystalline form or polymorph, collectively referred to as "crystal form", the term "crystalline form" is used herein to encompass any crystalline form of the compound of formula I, or a pharmaceutically acceptable salt thereof.
The term "effective amount" as used herein refers to an amount sufficient to achieve a desired prophylactic and/or therapeutic effect, e.g., an amount that achieves prevention or alleviation of symptoms associated with the disease to be treated.
The term "treatment" as used herein refers to both therapeutic treatment and prophylactic measures, the purpose of which is to prevent or delay (lessen) the disease state or condition being addressed. A subject is successfully "treated" if the subject receives a therapeutic amount of a compound, or an isomer, solvate, pharmaceutically acceptable salt or pharmaceutical composition thereof, according to the methods described herein, and the subject exhibits an observable and/or detectable decrease or improvement in one or more of the indications and symptoms of the subject. It is also understood that the prevention or treatment of a disease state or condition as described includes not only complete prevention or treatment, but also less than complete prevention or treatment, but also achievement of some biologically or medically relevant result.
The term "vascular disease" as used herein refers primarily to myocardial ischemia/reperfusion injury, various forms of heart failure following injury, cardiomyopathy, circulatory shock, cardiovascular aging, cardiovascular complications of diabetes, myocardial hypertrophy, atherosclerosis, vascular remodeling, angiogenesis.
The terms "neurodegenerative diseases and nervous system inflammation" as used herein mainly refer to stroke, brain trauma, neurodegenerative diseases (Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis) and nervous system inflammation such as multiple sclerosis and the like due to the harmful effects of oxidative, nitrosative stress.
Advantageous effects of the invention
The application provides a phthalazinone compound through intensive research on phthalazinone PARP inhibitors, wherein the compound can realize at least one of the following technical effects:
(1) can obviously improve the proliferation inhibition effect on tumor cells or/and cancer cells;
(2) can increase the stability of the molecule in vivo and reduce the possibility of producing toxic metabolites;
(3) the toxicity of drug molecules can be reduced, so that the safety of the drug molecules in the aspect of disease treatment is further improved, and the applicable disease group range of the drug is expanded;
(4) through the structural modification of the phthalazinone compound, the oxidative metabolism capability of the compound under the action of a P450 cytochrome enzyme system in vivo is reduced, and the bioavailability is improved; in addition, the first and second substrates are,
(6) the compound of the application has excellent long-acting property, so that the administration frequency can be reduced, and the compliance of patients can be improved.
The good physicochemical properties of the compounds of the present application make them of great potential in the development of PARP inhibitors with excellent bioavailability, inhibitory effectiveness and safety.
Drawings
FIG. 1 shows the proliferation inhibitory effect of the compounds of the present application on breast cancer MDA-MB-453.
FIGS. 2-1 and 2-2 show the proliferation inhibitory effect of the compounds of the present application on breast cancer MDA-MB-468.
FIG. 3 shows the proliferation inhibitory effect of the compounds of the present application on pancreatic cancer, Capan-1.
FIG. 4 shows the proliferation inhibitory effect of the compounds of the present application on colorectal cancer HCT 116.
Detailed Description
Embodiments of the present invention will be described in detail below with reference to examples, but those skilled in the art will appreciate that the following examples are only illustrative of the present invention and should not be construed as limiting the scope of the present invention. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
The structure of the compound is determined by nuclear magnetic resonance1HNMR) or Mass Spectrometry (MS).
Abbreviations in Nuclear Magnetic Resonance (NMR) spectra used in the examples are shown below:
s: unimodal (singlet), d: doublet (doublt), t: triplet (triplet), q: quartet (quartz), AB: doublet (doubledoubledoublet), m: multiplet (multiplex), br: broad peak (broad).
1HNMR was measured by JEOL Eclipse 400 NMR spectrometer using deuterated methanol (CD) as the solvent3OD), deuterated chloroform (CDCl)3) Hexadeuterio dimethyl sulfoxide (DMSO-d6), internal standard Tetramethylsilane (TMS), chemical shift is 10-6(ppm) is given as a unit.
Determination of MS was performed using an agilent (esi) mass spectrometer, manufacturer: agilent, model: agilent 6120B.
Preparation of high performance liquid chromatography (YMC, ODS, 250X 20mml column) was prepared using Shimadzu LC-8A.
Thin layer chromatography silica gel plate (TLC) an aluminum plate (20X 20cm) from Merck was used, and the specification for separation and purification by thin layer chromatography was GF254(0.4-0.5nm) from Nicotiana.
The reaction was monitored by Thin Layer Chromatography (TLC) or LCMS using the following developer systems: dichloromethane and methanol system, n-hexane and ethyl acetate system, petroleum ether and ethyl acetate system, and volume ratio of solvent is regulated according to different polarities of the compounds or by adding triethylamine and the like.
The microwave reaction used a Biotage Initiator + (400W, RT-300 ℃) microwave reactor.
Column chromatography generally uses Qingdao ocean 200-mesh and 300-mesh silica gel as a carrier. The system of eluents comprises: the volume ratio of the solvent is adjusted according to different polarities of the compounds, and a small amount of triethylamine can be added for adjustment.
In the examples, the reaction temperature is, unless otherwise specified, room temperature (20 ℃ C. -30 ℃ C.).
Reagents used herein were purchased from Acros Organics, Aldrich Chemical Company, Texas Chemical, and others.
In the conventional syntheses as well as in the examples, and intermediate syntheses, the meanings of the abbreviations are as follows:
DCM: dichloromethane;
DIPEA: n, N-diisopropylethylamine;
DMAP: 4-dimethylaminopyridine;
MeOH: methanol;
THF: tetrahydrofuran;
EtOH: ethanol;
DMF: n, N-dimethylformamide;
DMF-DMA: n, N-dimethylformamide dimethyl acetal;
HATU: 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate;
NMP: n-methyl pyrrolidone;
LCMS: a liquid phase mass spectrometer;
TLC: thin layer chromatography.
Example 1:
4- [3- (2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 1)
Figure GPA0000242631730000411
The reaction route is as follows:
Figure GPA0000242631730000421
the first step is as follows: 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidinone
To N-Boc-3-pyrrolidone (56.2g) was added 300mL of DMF-DMA, and the mixture was stirred at 65 ℃ overnight. TLC showed the starting material reaction was complete. After concentration to remove DMF-DMA, the resulting solid was washed with methyl t-butyl ether, filtered, and the solid was concentrated to dryness to give the objective product (47.2g, yellow solid, yield: 64%).
MS m/z(ESI):241.1[M+H]+
The second step is that: 2-Methylthio-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To 250mL of an ethanol solution of sodium ethoxide (6.6g, 93mmol) was added methylthioamidine sulfate (7.8g, 41.6mmol) at room temperature, and after stirring for about 30 minutes, the first step of 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidone (5g, 20.8mmol) was added and stirred at reflux for 6 hours. TLC monitored the starting material for incomplete reaction, supplemented with methylthioamidine sulfate (7.8g, 41.6mmol) and sodium ethoxide (6.6g, 93mmol) and refluxed overnight. TLC was used to monitor the disappearance of the starting material, and after quenching the reaction system with water, ethanol was removed by concentration. The aqueous phase was extracted with ethyl acetate (50 mL. times.4), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin-dried and washed with methyl tert-butyl ether to give the title molecule (3.3g, pale yellow solid, yield: 59%).
MS m/z(ESI):268.1[M+H]+.
The third step: 2-Methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To a 50mL solution of 2-methylsulfanyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester (3.3g, 12.3mmol) in methylene chloride at 0 deg.C was added m-chloroperoxybenzoic acid (6.3g, 37.1mmol), allowed to spontaneously warm to room temperature, and monitored by TLC follow-up. The reaction of the starting materials was complete after 5 hours. Adding a 10% sodium sulfite solution into the reaction solution, stirring for 30 minutes, then adding a 10% sodium carbonate solution, stirring for a while, extracting with dichloromethane (50 mL. times.4), combining organic phases, drying with anhydrous sodium sulfate, spin-drying, washing the obtained residue with methyl tert-butyl ether to obtain the target molecule, wherein the yield is as follows: 86.7 percent.
MS m/z(ESI):300.3[M+H]+.
The fourth step: 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
Tert-butyl 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate (300mg, 1.00mmol) from the third step was dissolved in 5mL of acetonitrile, potassium carbonate (207mg, 1.50mmol) and ethylamine (132.6mg, 2mmol) were added, and the resulting mixture was reacted at 70 ℃ for 6 hours, quenched with water, and extracted with dichloromethane. The dichloromethane layer was dried over anhydrous sodium sulfate, and concentrated to give a residue which was purified by preparative thin layer chromatography to give the objective product (110mg, yield: 38%).
MS m/z(ESI):265[M+H]+.
The fifth step: n-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride
Tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate (10mg, 0.45mmol) from the fourth step was dissolved in a solution of hydrogen chloride in ethyl acetate (3mL) and stirred at room temperature overnight. The reaction solution was concentrated to obtain a residue, which was used in the next reaction.
MS m/z(ESI):165[M+H]+.
And a sixth step: 4- [3- (2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one
5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (134.2mg, 0.45mmol), N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride from the fifth step (74.5mg, 0.45mmol), HATU (188.2mg, 0.50mmol) and DIPEA (348.3mg, 2.7mmol) were dissolved in DMF and stirred at room temperature overnight. After completion of the reaction, water and ethyl acetate were added to the reaction mixture, followed by liquid separation, and the ethyl acetate layer was dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative thin layer chromatography to give the objective product (15mg, yield: 7.5%).
MS m/z(ESI):445[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.33-8.17(m,1H),8.02-7.95(m,1H),7.93-7.80(m,2H),7.53-7.42(m,2H),7.32-7.18(m,2H),4.69-4.53(m,2H),4.40-4.30(m,4H),3.57-3.21(m,2H),1.09(q,3H,J=8Hz).
Example 2:
4- [3- (2-Cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 2)
Figure GPA0000242631730000431
The reaction route is as follows:
Figure GPA0000242631730000441
the first step is as follows: 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidinone
To N-Boc-3-pyrrolidone (56.2g) was added 300mL of DMF-DMA, and the mixture was stirred at 120 ℃ overnight. TLC showed the starting material reaction was complete. After concentration to remove DMF-DMA, the resulting solid was washed with methyl t-butyl ether, filtered, and the solid was concentrated to dryness to give the objective product (43g, yellow solid, yield: 58%).
MS m/z(ESI):241.1[M+H]+
The second step is that: 2-Methylthio-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To 250mL of an ethanol solution of sodium ethoxide (6.6g, 93mmol) was added methylthioamidine sulfate (15.6g, 83.2mmol) at room temperature, and after stirring for about 40 minutes, the first step of 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidone (5g, 20.8mmol) was added and stirred at reflux overnight. TLC was used to monitor the disappearance of starting material, quenched with water and concentrated to remove ethanol. The aqueous phase was extracted with ethyl acetate (50 mL. times.4), and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and washed with tert-butyl methyl ether to give the objective molecule (10g, pale yellow solid, yield: 89%).
MS m/z(ESI):268.1[M+H]+.
The third step: 2- (methylsulfonyl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To a 50mL solution of 2-methylsulfanyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester (3.3g, 12.3mmol) in methylene chloride at 0 ℃ was added m-chloroperoxybenzoic acid (6.3g, 37.1mmol), and the mixture was allowed to warm to room temperature naturally, after completion of the reaction. Adding a 10% sodium sulfite solution into the reaction solution, stirring for 30 minutes, then adding a 10% sodium carbonate solution, stirring for a while, extracting with dichloromethane (50 mL. times.4), combining organic phases, drying with anhydrous sodium sulfate, concentrating under reduced pressure, and washing with methyl tert-butyl ether to obtain a target molecule, wherein the yield is as follows: 86.7%).
MS m/z(ESI):300.3[M+H]+.
The fourth step: 2-Cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
Tert-butyl 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate (300mg, 1.00mmol) from the third step was dissolved in 5mL of acetonitrile, potassium carbonate (207mg, 1.50mmol) and cyclopropylamine (2mmol) were added, and the resulting mixture was reacted at 75 ℃ for 8 hours, quenched with water, and extracted with dichloromethane. The dichloromethane layer was dried over anhydrous sodium sulfate, and concentrated to give a residue which was purified by preparative thin layer chromatography to give the objective product (400mg, yield: 69%).
MS m/z(ESI):265[M+H]+.
The fifth step: n-cyclopropyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride the tert-butyl 2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate (400mg, 18mmol) from the fourth step was dissolved in a solution of hydrogen chloride in ethyl acetate (3mL) and stirred at room temperature overnight. The reaction solution was concentrated to obtain a residue, which was used in the next reaction.
MS m/z(ESI):165[M+H]+.
And a sixth step: 4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl) -2H-phthalazin-1-one
5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (134.2mg, 0.45mmol), N-cyclopropylamino-6, 7-dihydro-5H-pyrrolo [3,4, -d ] pyrimidin-2-amine hydrochloride from step five (95mg, 0.45mmol), HATU (188.2mg, 0.50mmol) and DIPEA (348.3mg, 2.7mmol) were dissolved in DMF and stirred at room temperature overnight. After completion of the reaction, water and ethyl acetate were added to the reaction mixture, followed by liquid separation, and the ethyl acetate layer was dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative thin layer chromatography to give the objective product (76mg, yield: 37%).
MS m/z(ESI):457[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.30-8.20(m,1H),8.00-7.95(m,1H),7.96-7.84(m,2H),7.56-7.45(m,2H),7.28-7.18(m,2H),4.69-4.53(m,2H),4.39(s,2H),3.80-3.74(m,1H),3.53-3.25(m,2H),1.23-1.12(m,4H).
Example 3:
4- [3- (2-Cyclopropylmethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 3)
Figure GPA0000242631730000451
The reaction route is as follows:
Figure GPA0000242631730000452
Figure GPA0000242631730000461
the first step is as follows: 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidinone
To N-Boc-3-pyrrolidone (18.5g) was added 300mL of DMF-DMA and the mixture was stirred at 60 ℃ overnight. TLC showed the starting material reaction was complete. After concentration to remove DMF-DMA, the resulting solid was washed with methyl t-butyl ether, filtered, and the solid was concentrated to dryness to give the objective product (20g, yellow solid, yield: 83%).
MS m/z(ESI):241.1[M+H]+
The second step is that: 2-hydroxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To 250mL of sodium ethoxide (13.2g, 186mmol) in ethanol was added urea (10g, 166.4mmol) at room temperature, and after stirring for about 40 minutes, the first step of 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidone (10g, 41.6mmol) was added and stirred at reflux overnight. TLC was used to monitor the disappearance of starting material, quenched with water and concentrated to remove ethanol. The aqueous phase was extracted with ethyl acetate (50 mL. times.4), and the organic phases were combined and dried over anhydrous sodium sulfate. After spin-drying, washing with methyl tert-butyl ether gave the title molecule (6.2g, pale yellow solid, yield: 63%).
MS m/z(ESI):268.1[M+H]+.
The third step: 2-chloro-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
At 0 ℃, a solution obtained by dissolving 2-hydroxy-5, 7-dihydro-pyrrole [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester (6.2g, 26.2mmol) in phosphorus oxychloride (10mL) is naturally heated to room temperature for reaction until the raw material disappears, and residues obtained after the phosphorus oxychloride is evaporated are directly used for the next reaction.
MS m/z(ESI):156[M+H]+.
The fourth step: n- (cyclopropylmethyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine
2-chloro-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride (385mg, 2mmol) was added to cyclopropylmethylamine (10mL) with heating, and after completion of the TLC monitoring reaction, preparative TLC purification gave 40mg of the desired product in 10.5% yield.
The fifth step: 4- [3- (2-cyclopropylmethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one
5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (63mg, 0.21mmol), N- (cyclopropylmethyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine (40mg, 0.21mmol) from step four, HATU (95mg, 0.25mmol) and DIPEA (38.7mg, 0.3mmol) were dissolved in DMF and stirred at room temperature overnight. After completion of the reaction, water and ethyl acetate were added to the reaction mixture, followed by liquid separation, and the ethyl acetate layer was dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative thin layer chromatography to give the objective product (32.6mg, yield: 33%).
MS m/z(ESI):471[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.31-8.13(m,2H),8.01-7.94(m,1H),7.94-7.80(m,2H),7.51-7.42(m,2H),7.38-7.22(m,2H),4.69-4.54(m,2H),4.40-4.30(m,4H),3.18-3.07(m,2H),1.09-1.00(m,1H),0.45-0.34(m,2H),0.24-0.15(m,2H).
Example 4:
4- { 4-fluoro-3- [2- (2-hydroxyethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 4)
Figure GPA0000242631730000471
The reaction route is as follows:
Figure GPA0000242631730000472
the first step is as follows: 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidinone
To N-Boc-3-pyrrolidone (37g) was added 300mL of DMF-DMA and the mixture was stirred at 60 ℃ overnight. TLC showed the starting material reaction was complete. After concentration to remove DMF-DMA, the resulting solid was washed with tetrahydrofuran, filtered, and the solid was concentrated to dryness to obtain the objective product (35g, yellow solid, yield: 72.6%).
MS m/z(ESI):241.1[M+H]+
The second step is that: 2-hydroxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To 250mL of sodium ethoxide (26.4g, 372mmol) in ethanol was added urea (20g, 332.8mmol) at room temperature, and after stirring for about 40 minutes, the first step of 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidone (20g, 83.2mmol) was added and the mixture was stirred at reflux overnight. TLC was used to monitor the disappearance of starting material, quenched with water and concentrated to remove ethanol. The aqueous phase was extracted with ethyl acetate (50 mL. times.5), and the organic phases were combined and dried over anhydrous sodium sulfate. After spin-drying, washing with methyl tert-butyl ether gave the expected molecule (11g, pale yellow solid, yield: 55.9%).
MS m/z(ESI):268.1[M+H]+.
The third step: 2-chloro-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
At the temperature of 0 ℃, the solution obtained by dissolving 2-hydroxy-5, 7-dihydro-pyrrole [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester (10g, 42mmol) in phosphorus oxychloride (40mL) is naturally heated to room temperature for reaction until the raw material disappears, and the residue obtained after the phosphorus oxychloride is removed by evaporation is directly used for the next reaction.
MS m/z(ESI):156[M+H]+.
The fourth step: 2- ((6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-yl) amino) ethanol
2-chloro-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride (385mg, 2mmol) was added to ethanolamine (50mL) with heating, and after completion of the TLC monitoring reaction, preparative TLC purification yielded 29.6mg of the objective product in 7.8% yield.
The fifth step: 4- { 4-fluoro-3- [2- (2-hydroxyethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one
5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (63mg, 0.21mmol), 2- ((6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-yl) amino) ethanol (29.6mg, 0.16mmol) from step four, HATU (95mg, 0.25mmol) and DIPEA (38.7mg, 0.3mmol) were dissolved in DMF and stirred at room temperature overnight. After completion of the reaction, water and ethyl acetate were added to the reaction solution, followed by liquid separation, and the methyl tert-butyl ether layer was dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative thin layer chromatography to give the objective product (20.6mg, yield: 28%).
MS m/z(ESI):461[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.33-8.13(m,2H),8.01-7.80(m,3H),7.51-7.43(m,2H),7.33-7.22(m,2H),7.18-7.06(m,1H),4.73-4.54(m,2H),4.42-4.30(m,3H),3.66-3.23(m,5H).
Example 5:
4- {3- [2- (2-Dimethylaminoethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one (Compound 5)
Figure GPA0000242631730000481
Figure GPA0000242631730000491
The reaction route is as follows:
Figure GPA0000242631730000492
the first step is as follows: 2- (2-Dimethylaminoethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
The title compound was synthesized by the method of the fourth step of example 1 by substituting N, N-dimethylethylenediamine for ethylamine used in the fourth step of example 1.
The second step is that: n is a radical of1- (6, 7-dihydro-5H-pyrrolo [3, 4-d)]Pyrimidin-2-yl) -N, N-dimethylethyl-1, 2-diamine hydrochloride.
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2- (2-dimethylaminoethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate for tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate.
The third step: 4- {3- [2- (2-dimethylaminoethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one
Synthesized using an experimental procedure analogous to the sixth step of example 1, using N1- (6, 7-dihydro-5H-pyrrolo [3, 4-d)]Pyrimidin-2-yl) -N, N-dimethylethyl-1, 2-diamine hydrochloride in place of N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] used in example 1]Pyrimidine-2-amine hydrochloride.
MS m/z(ESI):488[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.32-8.24(m,2H),8.02-7.96(m,1H),7.94-7.79(m,2H),7.52-7.44(m,2H),7.32-7.23(m,1H),7.11-7.04(m,1H),4.67(s,1H),4.58(s,1H),4.40-4.31(m,4H),3.40-3.29(m,2H),2.48-2.40(m,2H),2.24-2.16(m,3H).
Example 6
4- { 4-fluoro-3- [2- (tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 6)
Figure GPA0000242631730000501
The reaction route is as follows:
Figure GPA0000242631730000502
the first step is as follows: 2- (tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
The title compound was synthesized by the method of the fourth step of example 1 substituting 4-amino-tetrahydro-pyran for the ethylamine used in the fourth step of example 1.
The second step is that: n- (tetrahydro-pyran-4-yl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride.
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate with tert-butyl 2- (tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate.
The third step: 4- { 4-fluoro-3- [2- (tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one
The synthesis was carried out using an experimental procedure analogous to the sixth step of example 1, substituting N- (tetrahydro-pyran-4-yl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride for the N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1.
MS m/z(ESI):501[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.32-8.15(m,2H),8.02-7.96(m,1H),7.94-7.81(m,2H),7.52-7.42(m,2H),7.32-7.24(m,2H),4.67(s,1H),4.58(s,1H),4.39-4.31(m,4H),3.96-3.82(m,3H),3.43-3.35(m,1H),1.84-1.75(m,2H),1.55-1.43(m,2H),1.26-1.21(m,1H).
Example 7
4- [3- (2-dimethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 7)
Figure GPA0000242631730000511
Reaction scheme
Figure GPA0000242631730000512
The first step is as follows: 2-dimethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
The title compound was synthesized by the fourth step of example 1 by substituting dimethylamine in methanol for the ethylamine used in the fourth step of example 1.
The second step is that: n, N-dimethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride.
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate with tert-butyl 2-dimethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate.
The third step:
4- [3- (2-dimethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one was synthesized using an experimental procedure similar to the sixth step of example 1, substituting N, N-dimethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride for the N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1.
MS m/z(ESI):445[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.42-8.20(m,2H),8.03-7.80(m,3H),7.52-7.20(m,3H),4.70-4.55(m,2H),4.43-4.30(m,4H),3.15-3.06(m,6H).
Example 8
4- { 4-fluoro-3- [2- (2-methoxyethylamine) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 8)
Figure GPA0000242631730000521
The reaction route is as follows:
Figure GPA0000242631730000522
the first step is as follows: 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidinone
To N-Boc-3-pyrrolidone (37g) was added 300mL of DMF-DMA and the mixture was stirred at 80 ℃ overnight. TLC showed the starting material reaction was complete. After concentration to remove DMF-DMA, the resulting solid was washed with diethyl ether, filtered, and the solid was concentrated to dryness to give the objective product (35g, yellow solid, yield: 72.6%).
MS m/z(ESI):241.1[M+H]+
The second step is that: 2-hydroxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To 250mL of sodium ethoxide (26.4g, 372mmol) in ethanol was added urea (20g, 332.8mmol) at room temperature, and after stirring for about 40 minutes, the first step of 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidone (20g, 83.2mmol) was added and the mixture was stirred at reflux overnight. TLC was used to monitor the disappearance of starting material, quenched with water and concentrated to remove ethanol. The aqueous phase was extracted with ethyl acetate (50 mL. times.5), and the organic phases were combined and dried over anhydrous sodium sulfate. After concentration under reduced pressure and washing with methyl t-butyl ether, the objective molecule (11g, pale yellow solid, yield: 55.9%) was obtained.
MS m/z(ESI):268.1[M+H]+.
The third step: 2- (((trifluoromethyl) sulfonyl) oxy) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonic acid tert-butyl ester
Tert-butyl 2-hydroxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate (237mg, 1mmol) and N-methylmorpholine (253mg, 2.5mmol) were dissolved in THF (10mL) and a solution of trifluoromethanesulfonic anhydride (338.4mg, 1.2mmol) in THF (5mL) was added dropwise over an ice bath. After TLC monitoring reaction, THF is evaporated to dryness, water and ethyl acetate are added, an ethyl acetate layer is dried by anhydrous sodium sulfate, and a crude product of a target product is obtained after rotary drying and is directly used for the next reaction.
The fourth step: 2- ((2-methoxyethyl) amino) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonic acid tert-butyl ester
Tert-butyl 2- (((trifluoromethyl) sulfonyl) oxy) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonate (370mg, 1mmol) and 2-methoxyethylamine (75mg, 1mmol) were dissolved in THF, followed by addition of Pd (dppf) Cl2 dichloromethane complex (30mg), DIPEA (142mg, 1.1mmol) and Brettphos (60 mg). After the reaction of the resulting mixture was completed under reflux, THF was evaporated to dryness, and extracted with water and ethyl acetate (10mLx 8). The ethyl acetate was evaporated to dryness and the residue was purified by preparative TLC to give 110mg of the target product in 37% yield.
The fifth step: n- (2-methoxyethyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride
Tert-butyl 2- ((2-methoxyethyl) amino) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonate from step four (110mg, 0.37mmol) was dissolved in a solution of hydrogen chloride in ethyl acetate (3mL) and stirred at room temperature overnight. The reaction solution was concentrated to obtain a residue, which was used in the next reaction.
MS m/z(ESI):194[M+H]+.
And a sixth step: 4- { 4-fluoro-3- [2- (2-methoxyethylamine) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one
5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (89.4mg, 0.3mmol), N- (2-methoxyethyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride (65mg, 0.28mmol) from the fifth step, HATU (114mg, 0.30mmol) and DIPEA (77.4mg, 0.6mmol) were dissolved in DMF and stirred at room temperature overnight. After completion of the reaction, water and ethyl acetate were added to the reaction mixture, followed by liquid separation, and the ethyl acetate layer was dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative thin layer chromatography to give the objective product (32mg, yield: 22.5%).
MS m/z(ESI):475[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.32-8.15(m,2H),8.01-7.96(m,1H),7.94-7.80(m,2H),7.50-7.43(m,2H),7.31-7.20(m,2H),4.67(s,1H),4.58(s,1H),4.40-4.31(m,4H),3.45-3.38(m,4H),3.25and3.23(s,3H).
Example 9
4- { 4-fluoro-3- [2- (2-hydroxy-2-methylpropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 9)
Figure GPA0000242631730000541
The reaction route is as follows:
Figure GPA0000242631730000542
the first step is as follows: 2- (2-hydroxy-2-methylpropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
The title compound was synthesized by the method of the fourth step of example 1 by substituting 1-amino-2-methylpropyl-2-ol for ethylamine used in the fourth step of example 1.
The second step is that: 1- (6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amino) -2-methylpropyl-2-ol hydrochloride.
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate with tert-butyl 2- (2-hydroxy-2-methylpropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate.
The third step: 4- { 4-fluoro-3- [2- (2-hydroxy-2-methylpropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one
The synthesis was carried out using an experimental procedure analogous to the sixth step of example 1, substituting 1- (6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amino) -2-methylpropyl-2-ol hydrochloride for the N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1.
MS m/z(ESI):489[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(brs,1H),8.30-8.14(m,2H),8.01-7.70(m,3H),7.50-7.42(m,2H),7.31-7.23(m,1H),4.68-4.50(m,3H),4.40-4.31(m,4H),3.32-3.20(m,2H),1.09and 1.07(s,6H).
Example 10
4- {3- [2- (cyclopropylmethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluoro-benzyl } -2H-phthalazin-1-one (Compound 10)
Figure GPA0000242631730000551
Reaction scheme
Figure GPA0000242631730000552
The first step is as follows: 2- (cyclopropylmethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
The title compound was synthesized by the method of the fourth step of example 1 substituting N-methylcyclopropylamine for ethylamine used in the fourth step of example 1.
The second step is that: N-cyclopropyl-N-methyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride.
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate with tert-butyl 2- (cyclopropylmethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate.
The third step: 4- {3- [2- (cyclopropylmethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluoro-benzyl } -2H-phthalazin-1-one
The synthesis was carried out using an experimental procedure analogous to the sixth step of example 1, substituting N-cyclopropyl-N-methyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride for the N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1.
MS m/z(ESI):471[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.42-8.24(m,2H),8.01-7.97(m,1H),7.94-7.80(m,2H),7.52-7.43(m,2H),7.31-7.23(m,1H),4.66(d,2H,J=3.6Hz),4.42-4.32(m,4H),3.06(d,3H,J=8Hz),2.80-2.69(m,1H),0.87-0.74(m,2H),0.66-0.55(m,2H).
Example 11
4- { 4-fluoro-3- [2- (1-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 11)
Figure GPA0000242631730000561
Reaction scheme
Figure GPA0000242631730000562
The first step is as follows: 2- (1-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
The title compound was synthesized by the method of the fourth step of example 1 substituting 1-methylcyclopropylamine for ethylamine used in the fourth step of example 1.
The second step is that: n- (1-methylcyclopropyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride.
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate with tert-butyl 2- (1-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate.
The third step: 4- { 4-fluoro-3- [2- (1-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one
The synthesis was carried out using an experimental procedure analogous to the sixth step of example 1, substituting N- (1-methylcyclopropyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride for the N-ethyl-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1.
MS m/z(ESI):471[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.44-8.16(m,2H),8.05-7.81(m,3H),7.64-7.40(m,3H),7.38-7.15(m,1H),4.62(d,J=4.4Hz,2H),4.46-4.29(m,4H),2.60-2.40(s,3H),1.41-1.25(m,2H),0.73-0.51(m,2H).
Example 12
4- { 4-fluoro-3- [2- (3-oxetanylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 12)
Figure GPA0000242631730000571
Reaction scheme
Figure GPA0000242631730000572
The first step is as follows: 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidinone
To N-Boc-3-pyrrolidone (37g) was added 300mL of DMF-DMA, and the mixture was stirred at 100 ℃ overnight. TLC showed the starting material reaction was complete. After concentration to remove DMF-DMA, the resulting solid was washed with methyl t-butyl ether, filtered, and the solid was concentrated to dryness to give the objective product (35g, yellow solid, yield: 72.6%).
MS m/z(ESI):241.1[M+H]+
The second step is that: 2-hydroxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylic acid tert-butyl ester
To 250mL of sodium ethoxide (26.4g, 372mmol) in ethanol was added urea (20g, 332.8mmol) at room temperature, and after stirring for about 50 minutes, the first step of 1-tert-butoxycarbonyl-3-dimethylaminomethyl-4-pyrrolidone (20g, 83.2mmol) was added and the mixture was stirred at reflux overnight. TLC was used to monitor the disappearance of starting material, quenched with water and concentrated to remove ethanol. The aqueous phase was extracted with ethyl acetate (50 mL. times.5), and the organic phases were combined and dried over anhydrous sodium sulfate. After concentration under reduced pressure and washing with methyl t-butyl ether, the objective molecule (11g, pale yellow solid, yield: 55.9%) was obtained.
MS m/z(ESI):268.1[M+H]+.
The third step: 2- (((trifluoromethyl) sulfonyl) oxy) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonic acid tert-butyl ester
Tert-butyl 2-hydroxy-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate (237mg, 1mmol) and N-methylmorpholine (253mg, 2.5mmol) were dissolved in THF (10mL) and a solution of trifluoromethanesulfonic anhydride (338.4mg, 1.2mmol) in THF (5mL) was added dropwise over an ice bath. After TLC monitoring reaction, THF is evaporated to dryness, water and ethyl acetate are added, an ethyl acetate layer is dried by anhydrous sodium sulfate, and a crude product of a target product is obtained after rotary drying and is directly used for the next reaction.
The fourth step: 2- (Oxetanyl-3-ylamino) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonic acid tert-butyl ester
Tert-butyl 2- (((trifluoromethyl) sulfonyl) oxy) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonate (370mg, 1mmol) and oxetan-3 amine (73mg, 1mmol) were dissolved in THF, followed by addition of [1, 1 ' -bis (diphenylphosphino) ferrocene ] dichloropalladium/dichloromethane complex (30mg), DIPEA (142mg, 1.1mmol) and dicyclohexyl [3, 6-dimethoxy-2 ', 4 ', 6 ' -triisopropyl [1, 1 ' -biphenyl ] -2-yl ] phosphine (60 mg). After the reaction of the resulting mixture was completed under reflux, THF was removed by concentration, and the resulting residue was extracted with water and ethyl acetate (10mLx 8). Concentration removed ethyl acetate and purification of the residue by preparative TLC afforded 53mg of the title product in 18% yield.
The fifth step: n- (Oxetanyl-3-yl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride
Tert-butyl 2- (oxetan-3-ylamino) -5H-pyrrolo [3,4-d ] pyrimidine-6 (7H) -carbonate (53mg, 0.18mmol) from the fourth step was dissolved in a solution of hydrogen chloride in ethyl acetate (3mL) and stirred at room temperature overnight. The reaction solution was concentrated to obtain a residue, which was used in the next reaction.
MS m/z(ESI):193[M+H]+.
And a sixth step: 4- (4-fluoro-3- (2- (oxetan-3-ylamino) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one
5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (89.4mg, 0.3mmol), N- (oxetan-3-yl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride (34mg, 0.15mmol) from the fifth step, HATU (114mg, 0.30mmol) and DIPEA (77.4mg, 0.6mmol) were dissolved in DMF and stirred at room temperature overnight. After completion of the reaction, water and ethyl acetate were added to the reaction mixture, followed by liquid separation, and the ethyl acetate layer was dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative thin layer chromatography to give the objective product (12mg, yield: 2.5%).
MS m/z(ESI):473[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.60(brs,1H),8.33-8.18(m,2H),8.07-7.95(m,2H),7.93-7.81(m,2H),7.51-7.42(m,2H),7.30-7.24(m,1H),4.98-4.80(m,1H),4.78-4.70(m,2H),4.67-4.59(m,2H),4.52-4.45(m,2H),4.40-4.32(m,4H).
Example 13
4- { 4-fluoro-3- [2- (4-hydroxycyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 13)
Reaction scheme
Figure GPA0000242631730000591
The first step is as follows: 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine hydrochloride
The title compound was synthesized by the fifth step of example 1 substituting tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate with tert-butyl 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate.
The second step is that: 4- [ 4-fluoro-3- (2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl ] -2H-phthalazin-1-one.
Synthesized by the sixth procedure of example 1, substituting 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine hydrochloride for the N-ethyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1.
The third step: 4- { 4-fluoro-3- [2- (4-hydroxycyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one
The title compound was synthesized by the method of the fourth step of example 1 substituting ethylamine used in the fourth step of example 1 with 4-aminocyclohexanol and tert-butyl 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl ] -2H-phthalazin-1-one used in the fourth step of example 1 with 4- [ 4-fluoro-3- (2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl ] -2H-phthalazin-1-one.
MS m/z(ESI):515[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.31-8.11(m,2H),8.00-7.95(m,1H),7.93-7.81(m,2H),7.51-7.42(m,2H),7.31-7.22(m,1H),7.15-7.04(m,1H),4.68-4.52(m,3H),4.39-4.29(m,4H),3.69-3.52(m,1H),3.43-3.35(m,1H),1.93-1.78(m,4H),1.31-1.12(m,4H).
Example 14
4- { 4-fluoro-3- [2- (tetrahydrofuran-3-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 14)
Figure GPA0000242631730000601
Reaction scheme
Figure GPA0000242631730000602
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (4-fluoro-3- (2- ((tetrahydrofuran-3-yl) amino) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the method of the third step of example 14 by substituting tetrahydrofuran-3-amine for the 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):487[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.32-8.20(m,2H),8.05-7.94(m,2H),7.92-7.80(m,2H),7.53-7.43(m,2H),7.33-7.24(m,1H),4.98-4.80(m,1H),4.78-4.70(m,2H),4.67-4.59(m,2H),4.52-4.45(m,2H),4.40-4.32(m,4H),2.33-2.22(m,1H),1.92-1.83(m,1H).
Example 15
4- {3- [2- (4-Dimethylaminopiperidin-1-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one (Compound 15)
Figure GPA0000242631730000611
Reaction scheme
Figure GPA0000242631730000612
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (3- (2- (4- (dimethylamino) piperidin-1-yl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the method of the third step of example 13 by substituting 4-dimethylaminopiperidine for 4-aminocyclohexanol used in the third step of example 14.
MS m/z(ESI):528[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.43-8.20(m,2H),8.02-7.77(m,3H),7.52-7.40(m,2H),7.32-7.20(m,1H),4.72-4.55(m,3H),4.42-4.29(m,4H),2.95-2.82(m,2H),2.45-2.30(m,1H),2.19(s,6H),1.86-1.71(m,2H),1.31-1.18(m,3H)
Example 16
4- { 4-fluoro-3- [2- (4-methylpiperazin-1-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 16)
Figure GPA0000242631730000613
Figure GPA0000242631730000621
Reaction scheme
Figure GPA0000242631730000622
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (4-fluoro-3- (2- (4-methylpiperazin-1-yl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the method of the third step of example 13 by substituting 1-methylpiperazine for the 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):514[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.41-8.21(m,2H),8.00-7.95(m,1H),7.94-7.78(m,2H),7.51-7.43(m,2H),7.32-7.23(m,1H),4.64(d,J=3.2Hz,2H),4.41-4.32(m,4H),3.76-3.65(m,4H),2.38-2.29(m,4H),2.20(d,J=4.0Hz,3H).
Example 17
4- { 4-fluoro-3- [2- (1-methylpiperidin-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 17)
Figure GPA0000242631730000623
Reaction scheme
Figure GPA0000242631730000631
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (4-fluoro-3- (2- ((1-methylpiperidin-4-yl) amino) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the method of the third step of example 13 by substituting 1-methylpiperidin-4-amine for 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):514[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.31-8.13(m,2H),8.02-7.95(m,1H),7.93-7.80(m,2H),7.51-7.42(m,2H),7.32-7.12(m,2H),4.61(d,J=3.6Hz,2H),4.39-4.30(m,4H),3.76-3.57(m,1H),2.85-2.73(m,2H),2.24-2.17(m,3H),2.11-1.94(m,2H),1.86-1.75(m,2H),1.60-1.43(m,2H).
Example 18
4- {3- [2- (4, 4-Difluorocyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one (Compound 18)
Figure GPA0000242631730000632
Reaction scheme
Figure GPA0000242631730000641
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (3- (2- ((4, 4-difluorocyclohexyl) amino) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the method of the third step of example 13 by substituting 4, 4-difluorocyclohexylamine for the 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):535[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),8.35-8.14(m,2H),8.02-7.79(m,3H),7.51-7.42(m,2H),7.37-7.22(m,2H),4.63(d,J=4.0Hz,2H),4.40-4.32(m,4H),3.96-3.81(m,1H),2.11-1.83(m,6H),1.64-1.50(m,2H).
Example 19
4- { 4-fluoro-3- [2- (1-methyl-1H-pyrazol-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 19)
Figure GPA0000242631730000642
Reaction scheme
Figure GPA0000242631730000651
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (4-fluoro-3- (2- ((1-methyl-1H-pyrazol-4-yl) amino) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the procedure of the third step of example 13 by substituting 1-methyl-1H-pyrazol-4-amine for 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):497[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),9.58-9.51(m,1H),8.46-8.23(m,2H),8.04-7.80(m,4H),7.54-7.40(m,3H),7.32-7.25(m,1H),4.70(d,J=2.4Hz,2H),4.48-4.33(m,4H),3.79(d,J=8.0Hz,1H).
Example 20
4- { 4-fluoro-3- [2- (1-methyl-1H-pyrazol-3-amino) -5, 7-dihydro-pyrazolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (compound 20)
Figure GPA0000242631730000652
Reaction scheme
Figure GPA0000242631730000661
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
See example 13 for the first step.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
See example 13 for the second step.
The third step: 4- (4-fluoro-3- (2- ((1-methyl-1H-pyrazol-3-yl) amino) -6, 7-dihydro-5H-pyrazolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one
The title compound was synthesized by the procedure of the third step of example 13 by substituting 1-methyl-1H-pyrazol-3-amine for 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):497[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.61(brs,1H),9.83-9.79(m,1H),8.47-8.24(m,2H),8.02-7.80(m,3H),7.55-7.45(m,3H),7.32-7.23(m,1H),6.56-6.49(m,1H),4.70(d,J=2.8Hz,2H),4.46-4.32(m,4H),3.73(s,3H).
Example 21
(S) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 23)
Figure GPA0000242631730000662
Reaction scheme
Figure GPA0000242631730000671
The first step is as follows: 2- (methylsulfonyl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine hydrochloride
Using a procedure similar to the fifth step of example 1, tert-butyl 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate was used instead of tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate, the title compound was obtained.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
By following a procedure analogous to the sixth step of example 1, the N-ethyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1 was replaced with 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine hydrochloride to give the title compound.
The third step: (S) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one
The title compound was synthesized by a similar procedure to the third step of example 13 except that (S) -1-methoxy-2-propylamine was used instead of 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):489[M+H]+
1HNMR(400MHz,CDCl3)δ:8.45-8.42(m,1H),7.81-7.77(m,3H),7.76-7.67(m,2H),7.66-7.61(m,1H),7.31-7.27(m,1H),5.15-5.04(m,1H),4.90-4.83(m,1H),4.40(AB,1H),4.38-4.28(m,1H),3.96(AB,1H),3.93-3.80(m,1H),3.58-3.52(m,1H),3.41-3.35(m,4H),1.96(s,1H),1.14(d,J=4Hz,3H).
Example 22
(R) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one (Compound 24)
Figure GPA0000242631730000672
Reaction scheme
Figure GPA0000242631730000681
The first step is as follows: 2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine hydrochloride
Using a similar procedure as in the fifth step of example 1, tert-butyl 2-methanesulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonate was used instead of tert-butyl 2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carboxylate, the title compound was obtained.
The second step is that: 4- (4-fluoro-3- (2- (methylsulfonyl) -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) benzyl) phthalazin-1 (2H) -one.
The title compound was obtained by substituting 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidin-2-amine hydrochloride used in example 1 with 2-methylsulfonyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine hydrochloride in a similar manner to the sixth step of example 1.
The third step: (R) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one
The title compound was synthesized by a similar procedure to the third step of example 13 except that (R) -1-methoxy-2-propylamine was used instead of 4-aminocyclohexanol used in the third step of example 13.
MS m/z(ESI):489[M+H]+
1HNMR(400MHz,CDCl3)δ:8.45-8.42(m,1H),7.81-7.77(m,1H),7.76-7.67(m,2H),7.66-7.61(m,1H),7.31-7.27(m,1H),5.15-5.04(m,1H),4.90-4.83(m,1H),4.40(AB,1H),4.38-4.28(m,1H),3.96(AB,1H),3.93-3.80(m,1H),3.58-3.52(m,1H),3.41-3.35(m,4H),1.96(s,1H),1.14(d,J=4Hz,3H).
The following compounds were also synthesized by the methods described in the examples herein:
TABLE 1
Figure GPA0000242631730000682
Figure GPA0000242631730000691
Figure GPA0000242631730000701
Figure GPA0000242631730000711
Figure GPA0000242631730000721
Figure GPA0000242631730000731
Figure GPA0000242631730000741
Biological activity
Experimental example 1PARP1 kinase Activity Screen
The enzyme activity screening of PARP-1 inhibitors was performed using the PARP1 chemiluminescence kit from BPS Bioscience.
Test compounds: compounds of the examples of the present application;
the kit comprises: PARP1 Chemimecent Assay Kit, manufacturer: BPS Bioscience.
Experimental methods
Pretreatment of the kit: adding 50 mu L/hole 1 XPARP buffer diluted 1 Xhistone, and incubating overnight at 4 ℃; the next day, the liquid in the microplate was discarded and the plate was washed with 200. mu.l/well PBST (1 XPBS and 1% Triton X-100) to remove all the wash solution; then adding 200 mul of termination buffer solution into each hole, incubating for 90min at room temperature, and then discarding the liquid; and the plate was washed with 200. mu.l/well PBST, and finally all the wash was removed.
Reaction: the substrate, enzyme and test compound were added sequentially according to the following procedure and amounts and incubated for 1h at room temperature.
TABLE 2 reaction System
Figure GPA0000242631730000751
And (3) detection: after the reaction was completed, the well plate was rinsed with PBST (200. mu.l/well), and finally the rinsing solution was completely removed; adding 50 μ l diluted Strep-HRP to each well, and culturing at room temperature for 30 min; after rinsing the well plate with PBST (200. mu.L/well), the rinsing solution was removed completely; mix HRP substrate A and B on an ice bath, then add 100. mu.l per well (50. mu.l colorimetric substrate A and 50. mu.l colorimetric substrate B per well); finally, the high-sensitivity mode of a chemiluminescence method (Luminometric Measurement) is used for detecting the PARP1 kinase inhibition activity of the compound to be detected, and the result is shown in Table 3.
As a result:
TABLE 3 Single Point concentration inhibition of tested Compounds
Compound ID Single point concentration inhibition%
1 99.3%(5nM)
2 73.0%(5nM)
3 84.8%(5nM)
4 97.8%(5nM)
5 98.9%(5nM)
6 97.7%(5nM)
8 84.7%(5nM)
11 80.1%(5nM)
As can be seen from table 3, the compounds of the present application have a significant inhibitory effect on PARP1 kinase activity.
Other compounds of the present application have similar inhibitory effects on PARP1 kinase activity, with an inhibition rate in the range of 70-99%.
Experimental example 2Cell proliferation inhibition assay
The CCK-8 method was used to determine the effect of compounds on cell proliferation.
Test compounds: compounds of the examples of the present application;
positive control compound: olaparib, laboratory self-synthesis;
the kit comprises: CCK-8 Kit (Cell Counting Kit-8), manufacturer: beyond the Beyotime.
Experimental methods
Cell culture: cells were cultured according to the following conditions, counted using a cell counter after digestion of the cells, adjusted to the desired concentration according to the following requirements, and then seeded with 100. mu.l of cells per well, and administered 24 hours after seeding.
TABLE 4 cell culture conditions
Cell name Culture solution Conditions of the incubator
Breast cancer MDA-MB-453 DMEM/F12+10%FBS 5%CO2,37℃
Breast cancer MDA-MB-468 DMEM/F12+10%FBS 5%CO2,37℃
Colorectal cancer HCT116 1640+10%FBS 5%CO2,37℃
Pancreatic cancer Capan-1 IMEM+20%FBS 5%CO2,37℃
Compound preparation: after culturing for one day, dissolving the compound to be detected and the olaparib in DMSO to prepare a mother solution, sucking a proper amount of the mother solution into the culture solution, and uniformly mixing, wherein the medicine solution is prepared into a corresponding incubation concentration.
Incubation time: incubation was continued in the incubator for 7 days after dosing.
And (3) detection: after the incubation is finished, 200 mu l/hole of fresh culture solution is replaced and placed in the culture medium for 2 hours of stabilization, 20 mu l/hole of CCK8 is added for incubation for more than 3 hours, and the absorbance is measured at the wavelength of 450nm and the reference wavelength of 650nm by double wavelengths. IC (integrated circuit)50Values were calculated using GraphPad, and the results are detailed in tables 5-1 to 5-4 and figures 1 to 4:
TABLE 5-1 Effect of compounds of the present application on inhibiting breast cancer MDA-MB-453
Figure GPA0000242631730000761
Figure GPA0000242631730000771
Table 5-1 and FIG. 1 suggest that the compounds of the present application have significant proliferation inhibitory effects on breast cancer MDA-MB-453 cells. In particular, compounds 1, 2, 3,4, 6, 8, and compounds 64, 65, and 68 were significantly superior to olaparib in proliferation inhibition of breast cancer cells MDA-MB-453.
TABLE 5-2 Effect of the Compounds of the present application in inhibiting breast cancer MDA-MB-468
Figure GPA0000242631730000772
Table 5-2 and FIGS. 2-1 and 2-2 suggest that the compounds of the present application have significant proliferation inhibitory effects on breast cancer cells MDA-MB-468. In particular, compounds 1, 2, 3,4, 6, 8, 12, 13, 19, 20, and compounds 64, 65, and 68 significantly outperformed olaparib in the proliferation inhibition of breast cancer cells MDA-MB-468.
TABLE 5-3 Effect of Compounds of the present application in inhibiting pancreatic cancer Capan-1
Figure GPA0000242631730000773
Figure GPA0000242631730000781
It is suggested from tables 5-3 and FIG. 3 that the compounds of the present application have a significant proliferation inhibitory effect on pancreatic cancer cells, Capan-1. In particular, compounds 1, 2, 3,4, 5, 6, 8, and compounds 64, 65, and 68 significantly outperformed olaparib in the proliferation inhibition of pancreatic cancer cells, Capan-1.
TABLE 5-4 inhibitory Effect of Compounds of the present application on colorectal cancer HCT116
Figure GPA0000242631730000782
Tables 5 to 4 and FIG. 4 suggest that the compounds of the present invention have a significant proliferation inhibitory effect on HCT116, a colorectal cancer cell. In particular, the proliferation inhibitory effects of compounds 1, 2, 3,4, 6, 13, 19 and 20 on colorectal cancer cell HCT116 were significantly superior to olaparib.
Other compounds of the present application have similar proliferation inhibitory activity against cancer cells, such as breast cancer cells, colorectal cancer cells, and pancreatic cancer cells.
In summary, the compounds of the present application have proliferation inhibitory activity against cancer cells such as breast cancer cells, colorectal cancer cells, and pancreatic cancer cells.
Experimental example 3Pharmacokinetics of the compound(PK) study
3.1 rat PK Studies
Male SD rats were given the compound of the present application and olaparib by intravenous and gavage, respectively, to investigate pharmacokinetic characteristics. The Intravenous (IV) and oral (PO) doses were 1 and 5mg/kg, respectively. IV and PO administration, collecting blood at different time points, anticoagulating the blood with heparin sodium, centrifuging to obtain plasma sample, and performing LC-MS/MS analysis after the plasma sample is treated with precipitated protein.
LC-MS/MS, the chromatographic column is a Waters X-Bridge C18 column (21mM * 50mM, 3.5 mu m), the mobile phase A phase is water +2mM ammonium acetate, the mobile phase B phase is methanol +2mM ammonium acetate, the flow rate is 0.4mL/min, the column temperature is 40 ℃, an ESI source positive ion mode is adopted as an ion source, and the scanning mode is Multiple Reaction Monitoring (MRM).
Pharmacokinetic parameters were calculated using WinNonlin 6.3 software using a non-compartmental model, and the results are shown in table 6.
TABLE 6 IV pharmacokinetic parameters in rats
Figure GPA0000242631730000791
The results of the intravenous administration mode suggest that the compounds of the present application have good pharmacokinetic properties. In particular, compounds 1, 2, 3, 6 and 8 have pharmacokinetic parameters in rats superior to that of olaparib.
The PO results also showed that the plasma exposure of compound 1, compound 2, compound 3, and compound 6 was higher than that of olaparib at the same orally administered dose, with AUC values in the range of about 305-1200h * ng/ml.
In addition, it was unexpectedly found in the present pharmacokinetic experiment that the half-life of olaparib was 1.31 hours and the half-life of compound 2 of the present application was 2.3 hours, thus indicating that the duration of the pharmacodynamic action of compound 2 of the present application was longer.
3.2 dog PK Studies
The test compound was administered to male Beagle dogs intravenously and intragastrically, respectively, to investigate pharmacokinetic characteristics. The doses for Intravenous (IV) and oral (PO) administration were 1 and 5mg/kg, respectively, and the vehicle systems were 10% DMSO: 10% solutol: 80% normal saline. IV and PO were administered at different time points and blood anticoagulated with heparin sodium and plasma samples obtained after centrifugation were stored at-80 ℃. Plasma samples were analyzed by LC-MS/MS after they had been treated with precipitated protein.
The compounds of the present application have superior PK properties in dogs over olaparib, given the same route and dose of administration as 3.1 rat PK.
Other compounds of the present application have similar pharmacokinetic properties in rats and dogs.
Experimental example 4Safety test
4.1 hERG assay
In cardiomyocytes, the potassium channel encoded by human Ether-a-go-go Related Gene (hERG) mediates a delayed rectifier potassium current (IKr). IKr inhibition is the most important mechanism by which drugs cause prolongation of the QT interval. In the hERG assay, the criterion is if the test compound IC50If the concentration is more than 30 mu M, the tested compound is judged to have no inhibition effect on hERG.
Using PredictorTMhERG Fluorescence Polarization Assay, the compounds of the present application were tested for their effect on hERG potassium channels at concentrations of 3, 10, 30. mu.M. The results of the experiments showed 50% Inhibitory Concentration (IC) of compound 1, compound 2, compound 3, compound 6 against hERG50Values) are all greater than 30 μ M. The compounds of the present application are suggested to have no inhibitory effect on hERG, indicating that the compounds of the present application do not have the potential safety hazard of cardiac QT interval prolongation.
4.2 acute toxicity test in mice
The test compound is administered to KM mice by gavage, and the toxicity reaction of single administration of the test compound is examined to preliminarily judge the Maximum Tolerated Dose (MTD).
The dosage is set as 1: 300mg/kg of compound; compound 2, 3, 6: 200, 300mg/kg, single gavage administration, 10% DMSO and 50% PEG as vehicle and physiological saline as balance, observed 7 days after administration.
In this experiment, the test results are as follows:
compound 1: a Maximum Tolerated Dose (MTD) of greater than 300 mg/kg;
compound 2: MTD is less than 200 mg/kg;
compound 3: MTD is more than 300 mg/kg;
compound 6: MTD is 200 mg/kg;
in conclusion, the compounds of the present application are well tolerated under single administration of high doses.
Other compounds of the present application have similar safety and high dose tolerability.
Example 5Tumor inhibition experiment in vivo
This example is used to evaluate the effectiveness of the compounds of the present application on inhibition of tumor proliferation by different routes of administration.
5.1 human pancreatic cancer cell line Capan-1 tumor-bearing mouse model in vivo tumor suppression experiment
In this example, the effect of each compound on mice bearing tumors of pancreatic cancer, Capan-1, was evaluated by measuring the change in tumor volume in mice bearing subcutaneous transplantable tumors of human pancreatic cancer cell line, Capan-1, following administration of the compound of the present application via the PO route.
Selection of tumor volume 100-3The tumor-bearing mice were randomly divided into groups, and the administration volume was 10ml/kg, 1 administration per day, for about two weeks. Tumor volume was measured 2 times a week after dosing. The formula for tumor volume is: v is 0.5a × b2And a and b represent the major and minor diameters of the tumor, respectively. The antitumor therapeutic effect of the test compound was evaluated by the tumor growth inhibition ratio TGI (%).
TGI(%)=[1-(VTPowder-VTStarting point)/(VCPowder-VCStarting point)]*100%
Wherein VTPowder: mean tumor volume at the end of treatment group experiment
VTStarting point: mean tumor volume at the beginning of treatment group dosing
VCPowder: mean tumor volume at the end of vehicle control experiment
VCStarting point: mean tumor volume at the beginning of vehicle control group administration
The tumor inhibition rates for each group of compounds against pancreatic cancer are shown in table 7 below.
Table 7: tumor inhibition rate in pancreatic cancer cell strain Capan-1 model
Group of Compound (I) Route of administration Tumor inhibition Rate (%)
1 Vehicle control p.o. /
2 Olapari (100mg/kg) p.o. 9.2
3 Compound 2(3mg/kg) p.o. 109.7
4 Compound 2(0.3mg/kg) p.o. 44.3
5 Compound 3(10mg/kg) p.o. 48.6
6 Compound 3(3mg/kg) p.o. 36.0
7 Compound 6(10mg/kg) p.o. 56.8
8 Compound 6(3mg/kg) p.o. 20.2
Under the test conditions, the compound 2 has surprising drug effect, the tumor inhibition rate of the compound reaches 109.7 percent at 3mg/kg, the tumor is partially regressed, and the tumor inhibition rate is 44.3 percent at low dose (0.3 mg/kg). The drug effects of the compound 3 and the compound 6 are better than those of the positive control group.
5.2 in vivo tumor suppression experiment of human breast cancer cell line MX-1 tumor-bearing mouse model
In the embodiment, the drug effect of each compound on MX-1 tumor-bearing mice with breast cancer is evaluated by measuring the change of the tumor volume of mice with subcutaneous transplantation tumor of human breast cancer cell strains MX-1 after the compound is applied by a PO administration way.
Selection of tumor volume 100-3The tumor-bearing mice were randomly divided into groups, and the administration volume was 10ml/kg, 1 administration per day, for about 30 days. Tumor volume was measured 2 times a week after dosing. The formula for tumor volume is: v is 0.5 × a × b2And a and b represent the major and minor diameters of the tumor, respectively. The tumor-inhibiting therapeutic effect of the test compound is relative to the tumor proliferation rate T/C (%).
T/C(%)=(VEnd of T/VT start)/(VC powder/VC start)*100%
Wherein VTPowder: mean tumor volume at the end of treatment group experiment
VTStarting point: mean tumor volume at the beginning of treatment group dosing
VCPowder: mean tumor volume at the end of vehicle control experiment
VCStarting point: mean tumor volume at the beginning of vehicle control group administration
The evaluation criteria were: T/C (%) > 40% is ineffective; T/C (%) < 40%, and P < 0.05 is effective by statistical treatment.
Experimental results show that the compound has an in vivo relative tumor proliferation rate of less than 55% in a mouse model with tumor bearing human breast cancer cell strain MX-1 under the dosage of 5mg/kg and 10mg/kg of PO administration, and has an excellent tumor inhibition effect.
Other compounds of the present application have similar anti-tumor effects.
Formulation example 1Tablet formulation
The composition of the immediate release tablets is shown in table 8:
TABLE 8 composition of immediate release tablets
Composition (I) mg/tablet In% by weight of the tablet core
Compound
1 100.00 25.00
Lactose 238.00 50.00
Microcrystalline cellulose 40.00 10.00
Croscarmellose sodium 16.00 4.00
Sodium lauryl sulfate 2.00 0.50
Magnesium stearate 4.00 1.00
Tablet core weight 400.00 /
Preparation method
Standard immediate release tablets were made using the direct compression method. Compound 1 and lactose, microcrystalline cellulose, croscarmellose sodium and sodium lauryl sulfate were weighed into a glass vial, the above mixture occupied approximately 75% of the vial volume, and then mixed together in a tumbler mixer for 30 minutes to give a blended material. The blended material was sieved through a 40 mesh (425 μm) screen and then tumble mixed for an additional 15 minutes. Magnesium stearate was then added and the blend shaken for approximately 20 seconds. The resulting mixture was then dispensed into 400mg aliquots and compressed into tablet cores using a hand press equipped with a 10 mm die with a target compression of 0.5 tons.
Preparation example 2Capsule preparation
The composition of the capsule formulation is shown in table 9:
TABLE 9 composition of capsule formulations
Figure GPA0000242631730000831
Preparation method
Lauroyl is melted at about 50-70 ℃ and subsequently weighed into a stainless steel container. Compound 2 was added and the contents were mixed to suspend homogeneously. Mixing was continued while dispensing the mixture into capsules using a thermostatically controlled automatic capsule filling machine to make 500 mg/granule capsule formulation.
While specific embodiments of the invention have been described in detail, those skilled in the art will understand that: various modifications and alterations of detail may be made in light of the overall teachings of the disclosure, and such variations are within the scope of the invention. The full scope of the invention is given by the appended claims and any equivalents thereof.

Claims (19)

1. A compound of formula Iaa-1 or a pharmaceutically acceptable salt thereof;
Figure FDA0002288870290000011
wherein the content of the first and second substances,
a and B together with the linking atoms form a benzene ring;
R1、R2and R3Are all hydrogen;
R4is fluorine;
d and E are both C;
G. j, K and L are each independently selected from C and N and form a six membered aromatic ring system with D and E;
R5is selected from C1-6Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-6 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl and 3-8 membered heterocycloalkyl-C1-4Alkyl, optionally, wherein said C1-6Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-6 membered heteroaryl, 3-8 membered cycloalkyl-C1-4Alkyl or 3-8 membered heterocycloalkyl-C1-4Each alkyl is further independently substituted by one or more groups selected from halogen, hydroxy, C1-4Alkyl, halo C1-4Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, C1-4Alkoxy, RR' N-, RO-C1-4Alkyl, RR' N-C1-4Alkyl, RS (O)2-and RR' NSO2-is substituted with a substituent;
R6is hydrogen or C1-6An alkyl group;
alternatively, the first and second electrodes may be,
R5and R6Together with the N atom to which they are attached form a 3-8 membered alicyclic, 6-10 membered aromatic ring, 3-8 membered alicyclic ring or 5-10 membered heteroaromatic ring, optionally wherein the 3-8 membered alicyclic, 6-10 membered aromatic ring, 3-8 membered alicyclic ring or 5-10 membered heteroaromatic ring are each independently further substituted with one or more groups selected from halogen, hydroxy, cyano, RS (O)a-、C1-4Alkyl or C1-4Alkoxy, wherein a is 0, 1 or 2; r and R' are each independently selected from hydrogen, C1-4Alkyl and halo C1-4An alkyl group;
R8selected from hydrogen and C1-6An alkyl group.
2. The compound of claim 1 or a pharmaceutically acceptable salt thereof,
the R is5Selected from the group consisting of methyl, ethyl, cyclopropylmethyl, oxocyclobutylmethyl, tetrahydrofurylmethyl, methoxyethyl, 2-ethoxyethyl, 2-hydroxyethyl, trifluoroethyl, difluoroethyl, fluoroethyl, N-dimethylaminoethyl, (R) -2-methoxy-1-methylethyl, (S) -2-methoxy-1-methylethyl, (R) -2-methoxypropyl, (S) -2-methoxypropyl, methylsulfonylethyl, dimethylaminosulfonylethyl, 2-hydroxy-isobutyl, cyclopropyl, 1-methylcyclopropyl, 1-methoxymethylcyclopropyl, 2-dimethylcyclopropyl, 1-trifluoromethylcyclopropyl, (R) -2-fluorocyclopropyl, (S) -2-fluorocyclopropyl, 2-difluorocyclopropyl, oxocyclobutyl, 4-hydroxycyclohexyl, 4-difluorocyclohexyl, tetrahydropyranyl, 2-dimethyltetrahydropyranyl, 4-methyl-tetrahydropyranyl, N-methylpiperidinyl, N-dimethylaminopiperidinyl, N-methylpiperazinyl, piperidinyl, N-methylpyrazolyl or 3-oxo-bicyclo [3.1.0]Hexyl;
R6selected from hydrogen and methyl;
alternatively, the first and second electrodes may be,
R5and R6Together with the attached N atom form a piperidinyl or piperazinyl group.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
a and B together with the linking atoms form a benzene ring;
R1、R2and R3Are all hydrogen;
R4is fluorine;
D. e, G and K are both C;
l and J are N;
R5selected from the group consisting of ethyl, 2-hydroxyethyl, dimethylaminoethyl, methoxyethyl, cyclopropyl, 1-methylcyclopropyl, 4-hydroxycyclohexyl, cyclopropylmethyl, oxocyclobutyl, tetrahydrofuranyl, tetrahydropyranyl and N-methylpyrazolyl;
R6is hydrogen;
R8selected from hydrogen and methyl.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof:
4- [3- (2-ethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (2-cyclopropylmethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-hydroxyethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (2-dimethylaminoethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- [3- (2-dimethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-methoxyethylamine) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-hydroxy-2-methylpropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- {3- [2- (cyclopropylmethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluoro-benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (3-oxetanylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-hydroxycyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydrofuran-3-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (4-dimethylaminopiperidin-1-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-methylpiperazin-1-yl) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methylpiperidin-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (4, 4-difluorocyclohexylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methyl-1H-pyrazol-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methyl-1H-pyrazol-3-amino) -5, 7-dihydro-pyrazolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (methylmethoxyethylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- [3- (2-ethoxyethylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
(S) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(R) -4- { 4-fluoro-3- [2- (methoxypropyl-2-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(S) -4- { 4-fluoro-3- [2- (2-methoxypropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(R) -4- { 4-fluoro-3- [2- (2-methoxypropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2R) -2-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclobutylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-methoxymethylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2S) -2-methylcyclopropylamino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclopropylamino-4-methyl-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (1-trifluoromethylcyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (2, 2-difluorocyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- {3- [2- (2, 2-dimethylcyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2S) -2-fluorocyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- ((1R,2R) -2-fluorocyclopropyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-methylsulfonylethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
n, N-dimethyl-2- {6- [ 2-fluoro-5- (4-oxo-3, 4-dihydrophthalazin-1-methyl) benzoyl ] -6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidin-2-amino } -ethanesulfonamide;
4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-b ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (6-cyclopropylamino-1, 3-dihydro-pyrrolo [3,4-c ] pyridine-2-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- [3- (2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-b ] pyrazine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one;
4- {3- [2- (2, 2-dimethyl-tetrahydropyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-methyl-tetrahydro-pyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (3-azabicyclo [3.1.0] hexyl-6-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2,2, 2-trifluoroethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [2- (2, 2-difluoroethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-fluoroethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (oxetanyl-2-methyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (oxetanyl-3-methyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (tetrahydrofuran-2-methyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (2-trifluoromethoxyethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- {3- [ 2-cyclopropylamino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -7-fluoro-2H-phthalazin-1-one;
7-fluoro-4- { 4-fluoro-3- [2- (2-methoxyethyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
7-fluoro-4- { 4-fluoro-3- [2- (tetrahydropyran-4-amino) -5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
4- { 4-fluoro-3- [2- (4-methoxycyclohexyl) amino-5, 7-dihydro-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] benzyl } -2H-phthalazin-1-one;
(R) -4- {3- [2- (3-tetrahydrofuryl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one;
(S) -4- {3- [2- (3-tetrahydrofuryl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one;
4- {3- [2- (((2R,5R) -5-dimethylamino-1, 3-dioxan-2-yl) methyl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one;
4- {3- [2- ((2R,5R) -2- (dimethylamino) methyl-1, 3-dioxan-5-yl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one; and the combination of (a) and (b),
4- {3- [ 4-methyl-2- (4-tetrahydrofuryl) amino-6, 7-dihydro-5H-pyrrolo [3,4-d ] pyrimidine-6-carbonyl ] -4-fluorobenzyl } -2H phthalazin-1-one.
5. A process for the preparation of a compound of formula Iaa or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure FDA0002288870290000081
carrying out condensation reaction on the compound A and the compound 3B to obtain a compound of a formula Iaa;
wherein Y in the compound of formula Iaa and the compound 3B is N, R7The atoms and substituents in the remaining compounds, which are not present, are as defined in claim 1.
6. A pharmaceutical composition comprising a compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, optionally further comprising a pharmaceutically acceptable carrier or excipient.
7. The pharmaceutical composition according to claim 6, wherein the composition comprises the compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof in an amount of 0.01 to 2000 mg.
8. The pharmaceutical composition according to claim 6, wherein the composition comprises the compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof in an amount of 0.1 to 1000 mg.
9. The pharmaceutical composition according to claim 6, wherein the composition comprises the compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof in an amount of 1 to 800 mg.
10. The pharmaceutical composition according to claim 6, wherein the composition comprises the compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof in an amount of 10 to 600 mg.
11. The pharmaceutical composition according to claim 6, wherein the composition comprises the compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof in an amount of 50 to 500 mg.
12. The pharmaceutical composition of any one of claims 6-11, further comprising one or more anti-tumor drugs.
13. The pharmaceutical composition of claim 12, wherein the antineoplastic agent is selected from the group consisting of temozolomide, doxorubicin, paclitaxel, cisplatin, carboplatin, dacarbazine, topotecan, irinotecan, gemcitabine and bevacizumab, anti-CTLA-4 mab Ipilimumab, anti-PD-1 mab pembrolizumab and Nivolumab, and anti-PD-L1 mab atezolizumab.
14. Use of a compound of any one of claims 1-4 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 6-13, in the manufacture of an agent that inhibits PARP.
15. The use of claim 14, wherein the agent is a PARP-1 inhibitor.
16. Use of a compound according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 6 to 13, for the manufacture of an agent for the adjuvant treatment of tumours or for the enhancement of the effect of radiotherapy or chemotherapy of tumours.
17. Use of a compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 6 to 13, in the manufacture of a medicament for the treatment of a tumour.
18. The use of claim 16 or 17, wherein the tumor is selected from the group consisting of breast cancer, ovarian cancer, colorectal cancer, melanoma, lung cancer, gastrointestinal stromal tumor, brain cancer, cervical cancer, pancreatic cancer, prostate cancer, stomach cancer, chronic myeloid leukemia, liver cancer, lymphoma, peritoneal cancer, soft tissue sarcoma, neuroendocrine tumors, and glioblastoma.
19. Use of a compound of any one of claims 1-4 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 6-13, for the manufacture of a medicament for the treatment of a vascular disease, a neurodegenerative disease, or nervous system inflammation.
CN201680007634.7A 2015-10-30 2016-10-28 Phthalazinone derivatives, preparation method and use thereof Active CN107428762B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201510724885 2015-10-30
CN2015107248854 2015-10-30
PCT/CN2016/103735 WO2017071636A1 (en) 2015-10-30 2016-10-28 Phthalazine ketone derivative, and preparation method and use thereof

Publications (2)

Publication Number Publication Date
CN107428762A CN107428762A (en) 2017-12-01
CN107428762B true CN107428762B (en) 2020-03-10

Family

ID=58629896

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201680007634.7A Active CN107428762B (en) 2015-10-30 2016-10-28 Phthalazinone derivatives, preparation method and use thereof

Country Status (2)

Country Link
CN (1) CN107428762B (en)
WO (1) WO2017071636A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI716107B (en) 2018-09-26 2021-01-11 美商美國禮來大藥廠 6-fluoro-2-methylbenzo[d]thiazol-5-yl compounds
CN110845425B (en) * 2019-11-07 2023-09-08 合肥工业大学 Phthalazine derivative and preparation method and application thereof
CN115515939A (en) * 2020-05-08 2022-12-23 谛希诺生物科技有限公司 Novel phthalazine derivatives having an inhibitory activity on ectonucleotide pyrophosphatase-phosphodiesterase and their use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1788000A (en) * 2003-03-12 2006-06-14 库多斯药物有限公司 Phthalazinone derivatives
WO2012019426A1 (en) * 2010-08-09 2012-02-16 上海恒瑞医药有限公司 Phthalazinone derivative, and preparation method and pharmaceutical use thereof
CN102898377A (en) * 2012-02-14 2013-01-30 南京圣和药业有限公司 Novel phthalazinone derivatives and uses thereof
CN103570722A (en) * 2012-07-19 2014-02-12 中国科学院上海药物研究所 Fused ring pyridazinone compounds, preparation method and uses thereof
CN104918917A (en) * 2012-12-31 2015-09-16 卡迪拉保健有限公司 Substituted phthalazin-1 (2h)-one derivatives as selective inhibitors of poly (adp-ribose) polymerase-1

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1788000A (en) * 2003-03-12 2006-06-14 库多斯药物有限公司 Phthalazinone derivatives
WO2012019426A1 (en) * 2010-08-09 2012-02-16 上海恒瑞医药有限公司 Phthalazinone derivative, and preparation method and pharmaceutical use thereof
CN102898377A (en) * 2012-02-14 2013-01-30 南京圣和药业有限公司 Novel phthalazinone derivatives and uses thereof
CN103570722A (en) * 2012-07-19 2014-02-12 中国科学院上海药物研究所 Fused ring pyridazinone compounds, preparation method and uses thereof
CN104918917A (en) * 2012-12-31 2015-09-16 卡迪拉保健有限公司 Substituted phthalazin-1 (2h)-one derivatives as selective inhibitors of poly (adp-ribose) polymerase-1

Also Published As

Publication number Publication date
WO2017071636A1 (en) 2017-05-04
CN107428762A (en) 2017-12-01

Similar Documents

Publication Publication Date Title
WO2021129820A1 (en) Spiro ring-containing quinazoline compound
EP3184521B1 (en) Indazole compounds as fgfr kinase inhibitors, preparation and use thereof
ES2928169T3 (en) 4-(3H-indol-5-yl)-N-(pyridin-2-yl)pyrimidin-2-amine derivatives as protein kinase inhibitors, and method of preparation and medical use thereof
EP4077326A1 (en) Kras mutant protein inhibitors
JP6026427B2 (en) Substituted 6,6-fused nitrogen heterocyclic compounds and uses thereof
TW202144345A (en) Kras mutant protein inhibitors
WO2021088945A1 (en) Compound as shp2 inhibitor and use thereof
WO2021027911A1 (en) Novel spirocyclic k-ras g12c inhibitor
EP2990405A1 (en) Deuterated diaminopyrimidine compounds and pharmaceutical compositions comprising such compounds
WO2015158310A1 (en) Tyrosine kinase inhibitor and uses thereof
JP2016517859A (en) 3- (2-Aminopyrimidin-4-yl) -5- (3-hydroxypropynyl) -1H-pyrrolo [2,3-C] pyridine derivatives as NIK inhibitors for the treatment of cancer
CN110914277B (en) Imidazo [1,2-b ] pyrimido [4,5-d ] pyridazin-5 (6H) -one compounds and application thereof
WO2021249563A1 (en) Aryl or heteroaryl pyridone or pyrimidone derivative, preparation method therefor and application thereof
CN107207504B (en) Phthalazinone derivatives, preparation method and use thereof
CN112771049B (en) FGFR4 inhibitor and application thereof
WO2022135432A1 (en) Macrocyclic heterocyclic compounds as egfr inhibitors, and use thereof
TW202102505A (en) A pyrroloheterocyclic derivative and preparation method and medical use thereof
EP3560921A1 (en) Quinazoline compound and preparation method, application, and pharmaceutical compostion thereof
CN107428762B (en) Phthalazinone derivatives, preparation method and use thereof
CN112457306A (en) 3, 5-disubstituted pyrazole compounds as kinase inhibitors and application thereof
WO2021129584A1 (en) Pd-l1 antagonist compound
CN105541792B (en) Polycyclic class PI3K inhibitor
JP2019518032A (en) Bicyclic pyridine, pyrazine and pyrimidine derivatives as PI3K BETA inhibitors
CN113336774A (en) Substituted chiral diaryl macrocycles as TRK inhibitors
TWI828289B (en) Compounds as TYK2/JAK1 pseudokinase domain inhibitors and methods of synthesis and use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant