CN107207504B - Phthalazinone derivatives, preparation method and use thereof - Google Patents
Phthalazinone derivatives, preparation method and use thereof Download PDFInfo
- Publication number
- CN107207504B CN107207504B CN201680007644.0A CN201680007644A CN107207504B CN 107207504 B CN107207504 B CN 107207504B CN 201680007644 A CN201680007644 A CN 201680007644A CN 107207504 B CN107207504 B CN 107207504B
- Authority
- CN
- China
- Prior art keywords
- compound
- phthalazin
- dihydro
- fluoro
- methyl
- 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
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic 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
- A61K31/50—Pyridazines; Hydrogenated pyridazines
- A61K31/502—Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/55—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
- A61K31/551—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic 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
- C07D471/02—Heterocyclic 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
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic 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
- C07D471/12—Heterocyclic 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 three hetero rings
- C07D471/14—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic 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
- C07D487/04—Ortho-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)
- Nitrogen Condensed Heterocyclic Rings (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The application relates to phthalazinone derivatives, methods of preparation and uses thereof; in particular to a compound of 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 producing toxic metabolites; or the oxidative metabolism capability of the compound under the action of in vivo P450 cytochrome enzyme system can be reduced by reducing the structural modification of the phthalazinone compound, the bioavailability is improved,
Description
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 that stimulate 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-adenylate-ribose diphosphate (PRA) on receptor Proteins (including itself) using ADP as a substrate (Sakamoto-Hojo E T, Balajee A S. targeting Polymer (ADP) ribose polymerase I (PARP-I) and PARP-I interacting Proteins for cancer patients. anticancer genes Med. Chem,2008,8(4): 402. 416).
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. FreeRadiationbiol. 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 tumor (mainly breast cancer) with DNA damage repair defect. 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, the neuropathological study of PARP-1 has also been gaining increasing attention.
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.
A first aspect of the 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,
wherein,
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 halo, hydroxy, amino, cyano, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, alicyclic, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR ' N-, RC (O) N (R ' -, RR ' NC (O) -, and RS (O))a-wherein a is 0, 1 or 2;
d and E are each independently selected from C and N and together with the atoms to which they are attached form a 5-10 membered ring X, wherein the ring X is an alicyclic, aromatic or heteroaromatic ring; preferably, the ring X is a nitrogen-containing aliphatic heterocycle or heteroaromatic ring; preferably, the ring X is a five-or six-membered nitrogen-containing aliphatic heterocycle or heteroaromatic ring; preferably, the ring X is a five or six membered nitrogen containing heteroaromatic ring; the ring X is, for example, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring, a pyridone ring or a piperidine ring;
m and n are each independently selected from 0, 1,2 and 3, and m + n is 3 or 4;
R1、R2、R3and R4Each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, lipoheterocyclyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR' N-, RS (O)a-RC (O) N (R ') -and RR' NC (O) -; optionally, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo-aliphatic, aryl or heteroaryl 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, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-wherein a is 0, 1 or 2;
R5and R6Each independently selected from the group consisting of absent, hydrogen, oxy, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloaliphatic, aryl, heteroaryl, RO-, RC (O) -, ROC (O) -, RC (O), O-, RR ' N-, RC (O) N (R) -, RR ' NC (O) -, ROC (O) N (R ') -, RR ' NC (O) N (R ' -, RS (O))a-、RR’NSO2And RS (O)2N (R') -; optionally, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo-aliphatic, aryl or heteroaryl 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, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-wherein a is 0, 1 or 2; or,
R5or R6And form a polycyclic ring system on the ring X to which it is attached, wherein R is5Or R6Selected from the group consisting of 3-8 membered alicyclic rings, 5-8 membered alicyclic heterocyclic rings, 6-10 membered aromatic rings and 5-10 membered heteroaromatic rings;
r, R 'and R'Each independently selected from hydrogen and C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocyclylalkyl, 6-20 membered aryl, and 5-20 membered heteroaryl, optionally wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocyclylalkyl, 6-20 membered aryl or 5-20 membered heteroaryl 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, halo C1-4Alkyl, hydroxy C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-is substituted.
In certain preferred embodiments of the present application, the compounds of formula I are provided with the proviso that,
when A and B form an unsubstituted phenyl ring with the carbon atom to which they are attached, R1、R2And R3Are all H, R4In the case of F, the ratio of the total of the components is,
1) and in formula IIs composed ofWhen R is6Is absent, R5Is not H, hydroxy, methyl, isopropyl, cyclopropyl, trifluoromethyl, difluoromethyl, or trifluoroethyl;
2) and in formula IIs composed ofWhen R is5And R6Not all are H, and when R5Or R6One is H, the other is not trifluoromethyl or ethoxycarbonyl;
4) Ring X is not pyrrolidine, 2-pyrrolidone, morpholine, 3-morpholinone, tetrahydrofuran, 2-oxazolidinone, tetrahydrothiophene, and thietanesulfone; and,
5) the compound of formula I is not:
4- { [3- (3, 4-dihydro-1H-pyrrolo [1,2-a ] pyrazine-2-carbonyl) -4-fluoro-phenyl ] methyl } -2H-phthalazin-1-one;
4- { [3- (6, 8-dihydro-5H- [1,2,4] triazolo [1,5-a ] pyrazine-7-carbonyl) -4-fluoro-phenyl ] methyl } -2H-phthalazin-1-one;
4- { [ 4-fluoro-3- (2-trifluoromethyl-6, 8-dihydro-5H- [1,2,4] triazolo [1,5-a ] pyrazine-7-carbonyl) -phenyl ] methyl } -2H-phthalazin-1-one; and
4- [ 4-fluoro-3- (5, 6-dihydro-8H-imidazo [1,5-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one.
In certain preferred embodiments of the present application, in the compounds of formula I, A and B together with the atoms to which they are attached form an aromatic ring (e.g., a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, or a 6-10 membered aromatic ring), wherein the aromatic ring is unsubstituted or each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (e.g., fluorine, chlorine, bromine, or iodine), hydroxy, amino, cyano, carboxy, nitro, and C1-10Alkyl substituents.
In certain preferred embodiments of the present application, the compounds of formula I wherein a and B together with the atoms to which they are attached form a phenyl ring, wherein the phenyl ring is unsubstituted or each independently substituted with one or more (e.g. 1,2,3 or 4) substituents selected from halogen (e.g. fluorine).
In certain preferred embodiments of the present application, R in the compound of formula I1、R2、R3And R4Each independently selected from hydrogen, halogen, hydroxyl, amino, cyano, carboxyl, nitro, C1-10Alkyl and halo C1-10An alkyl group.
In certain preferred embodiments of the present application, R in the compound of formula I1、R2、R3And R4Each independently of the otherIs selected from hydrogen, halogen, hydroxy, amino, cyano, C1-6Alkyl (e.g. methyl) and halo C1-6Alkyl (e.g., trifluoromethyl).
In certain preferred embodiments of the present application, R in the compound of formula I1、R2And R3Is a hydrogen atom.
In certain preferred embodiments of the present application, R in the compound of formula I4Is halogen.
In certain preferred embodiments of the present application, R in the compound of formula I4Is fluorine.
In certain preferred embodiments of the present application, the compounds of formula I wherein A and B together with the atoms to which they are attached form a phenyl ring, R1、R2And R3Is hydrogen, and R4Is fluorine.
In certain preferred embodiments of the present application, R in the compound of formula I5And R6Each independently selected from absent, hydrogen, oxy, halogen, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclo or 5-10 membered heteroaryl are each independently selected by one or more (e.g. 1,2,3 or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy, 3-8 membered cycloalkyl, 3-8 membered lipoheterocyclyl, 6-10 membered aryl and 5-10 membered heteroaryl, wherein a is 0, 1 or 2;
and,
r and R' are each independently selected from hydrogen, C1-6Alkyl, 3-8 membered cycloalkyl, 5-8 membered heterocyclylalkyl, 6-10 membered aryl, and 5-10 membered heteroaryl; optionally, wherein said C1-6Alkyl, 3-8 membered cycloalkyl, 5-8 membered heterocyclylalkyl, 6-10 membered aryl, 5-10 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen, hydroxy, amino, cyano, C1-4Alkyl, halo C1-4Alkyl, hydroxy C1-4Alkyl, aryl, heteroaryl, and heteroaryl,RS(O)2-、C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered lipoheterocyclyl, 6-10 membered aryl and 5-10 membered heteroaryl; or, R and R' together with the N atom to which they are attached form a 4-10 membered heterocyclylalkyl group; or,
R5or R6And form a polycyclic ring system on the ring X to which it is attached, wherein R is5Or R6Selected from the group consisting of an aliphatic heterocycle (e.g., a 5-8 membered aliphatic heterocycle) and a heteroaromatic ring (e.g., a 5-10 membered heteroaromatic ring); preferably, the aliphatic heterocyclic or heteroaromatic ring is a five-or six-membered nitrogen-containing aliphatic heterocyclic or heteroaromatic ring; for example, a pyrrolidine ring, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring, an oxazole ring, a thiazole ring, a piperidine ring, a piperazine ring, a pyrazine ring, a morpholine ring, a thiomorpholine ring, a pyridine ring, a pyrimidine ring or a pyridazine ring.
In certain preferred embodiments of the present application, R in the compound of formula I5And R6Each independently selected from the group consisting of absent, hydrogen, oxy, halogen (e.g., F, Cl, Br or I), C1-6Alkyl and substituted C1-6Alkyl (e.g., methyl, ethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1-difluoroethyl, cyclopropyl-difluoromethyl, hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, 1-aminoethyl, cyclopropylmethyl, 2-hydroxy-2-propyl), C1-6Alkoxy (e.g., methoxy, ethoxy, propoxy, t-butoxy, cyclopropyloxy, 3-8 membered cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl), substituted acyl (e.g., cyclopropylformyl, acetyl), amino and substituted amino (e.g., cyclopropylamino), aminoacyl and substituted aminoacyl (aminoacyl, carbamyl, N, n-dimethylaminoyl, ethamoyl, 2-hydroxyethylaminoacyl, 2-methoxyethylamino, 2-hydroxyisobutyryl, 2-methanesulfonylethylamyl), substituted oxycarbonyl (e.g., methoxycarbonyl, ethoxycarbonyl), heteroaryl and substituted heteroaryl (e.g., pyrazolyl, 1-methylpyrazolyl), lipoheterocyclyl and substituted lipoheterocyclyl (e.g., 2-methyl- [ 1.3.4).]Oxo-oxadiazol-5-yl, 3-methyl- [1.2.4 ]]Oxo-oxadiazol-5-yl), methylthio and methylsulfonyl.
In certain preferred embodiments of the present applicationIn the formula I, R5Selected from hydrogen, oxy, halogen, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclo or 5-10 membered heteroaryl are each independently selected by one or more (e.g. 1,2,3 or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy, 3-8 membered cycloalkyl, 3-8 membered lipoheterocyclyl, 6-10 membered aryl and 5-10 membered heteroaryl, wherein a is 0, 1 or 2; or,
R5and form a polycyclic ring system on the ring X to which it is attached, wherein R is5Selected from the group consisting of an aliphatic heterocycle (e.g., a 5-8 membered aliphatic heterocycle) and a heteroaromatic ring (e.g., a 5-10 membered heteroaromatic ring); preferably, the aliphatic heterocyclic or heteroaromatic ring is a five-or six-membered nitrogen-containing aliphatic heterocyclic or heteroaromatic ring; for example, a pyrrolidine ring, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring, an oxazole ring, a thiazole ring, a piperidine ring, a piperazine ring, a pyrazine ring, a morpholine ring, a thiomorpholine ring, a pyridine ring, a pyrimidine ring or a pyridazine ring;
the R is6Absent or selected from hydrogen, oxy, halogen, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclo or 5-10 membered heteroaryl are each independently selected by one or more (e.g. 1,2,3 or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy, 3-8 membered cycloalkyl, 3-8 membered lipoheterocyclyl, 6-10 membered aryl and 5-10 membered heteroaryl, wherein a is 0, 1 or 2;
and,
r and R' are each independently selected from hydrogen, C1-6Alkyl, 3-8 membered cycloalkyl, 5-8 membered heterocyclylalkyl, 6-10 membered aryl, and 5-10 membered heteroaryl; optionally, wherein said C1-6Alkyl, 3-8 membered cycloalkyl, 5-8 membered heterocyclylalkyl, 6-10 membered aryl, 5-10 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen, hydroxy, amino, cyano, C1-4Alkyl, halo C1-4Alkyl, hydroxy C1-4Alkyl, RS (O)2-、C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered lipoheterocyclyl, 6-10 membered aryl and 5-10 membered heteroaryl, or R and R' together with the N atom to which they are attached form a 4-10 membered lipoheterocyclyl.
In certain preferred embodiments of the present application, R in the compound of formula I5Selected from hydrogen, oxy, halogen, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy and 3-8 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-6Alkyl and 3-8 membered cycloalkyl; optionally, wherein said C1-6Alkyl and 3-8 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-4Alkyl radical, C1-4Alkoxy and C1-4A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compound of formula I5Selected from hydrogen, oxy, halogen, C1-6Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocyclylalkyl, 5-6 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-6Alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocyclylalkyl, 5-6 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-4Alkyl radical, C1-4Alkoxy and 3-6 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-4Alkyl and 3-6 membered cycloalkyl; optionally, wherein said C1-4Alkyl and 3-6 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-2Alkyl radical, C1-2Alkoxy and C1-2A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compound of formula I5Selected from hydrogen, oxy, halogen, C1-4Alkyl, 3-6 membered cycloalkyl, 3-6 membered heterocyclylalkyl, 5-6 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-4Alkyl, 3-6 membered cycloalkyl, 3-6 membered lipoheterocyclyl, 5-6 membered heteroaryl are each independently selected by one or more (e.g. 1,2,3 or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-2Alkyl radical, C1-2Alkoxy and 3-6 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-2Alkyl and 3-6 membered cycloalkyl; optionally, wherein said C1-2Alkyl and 3-6 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-2Alkyl radical, C1-2Alkoxy and C1-2A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compound of formula I5Selected from the group consisting of hydrogen, bromine, hydroxymethyl, 1-hydroxyethyl, 2-hydroxy-2-propyl, difluoromethyl, trifluoromethyl, 1-difluoroethyl, pentafluoroethyl, cyclopropyl-difluoromethyl, 1-aminoethyl, cyclopropylmethyl, cyclopropyl, 1-methyl-4-pyrazolyl, 1-methyl-3-pyrazolyl, 2-methyl- [1.3.4]Oxo-oxadiazol-5-yl, 3-methyl- [1.2.4 ]]Oxo-oxadiazol-5-yl, methoxy, cyclopropyloxy, acetyl, ethoxycarbonyl, cyclopropylamino, aminoacyl,Methyl aminoacyl, N-dimethylaminoyl, ethyl aminoacyl, 2-hydroxy ethyl aminoacyl, 2-methoxy ethyl aminoacyl, 2-hydroxy isobutyl aminoacyl, 2-methyl sulfonyl ethyl aminoacyl, methylthio and methyl sulfonyl.
In certain preferred embodiments of the present application, R in the compound of formula I6Absent or selected from hydrogen and halo C1-10Alkyl (e.g. halo C)1-6An alkyl group; for example halo C1-4An alkyl group; such as trifluoromethyl).
In certain preferred embodiments of the present application, R in the compound of formula I6Absent or selected from hydrogen and trifluoromethyl.
In certain preferred embodiments of the present application, m ═ 1, n ═ 2; alternatively, m is 2 and n is 2.
In certain preferred embodiments of the present application, the compounds have the structure shown in formula I-A,
wherein,
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) groups selected from halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR ' N-, RC (O) N (R ') -, RR ' NC (O) -and RS (O)aWherein a is 0, 1 or 2;
ring X is an alicyclic ring, an alicyclic heterocyclic ring, an aromatic ring or a heteroaromatic ring, preferably, the ring X is a nitrogen-containing alicyclic heterocyclic ring or a heteroaromatic ring; preferably, the ring X is a five-or six-membered nitrogen-containing aliphatic heterocycle or heteroaromatic ring; preferably, the ring X is a five or six membered nitrogen containing heteroaromatic ring; the ring X is, for example, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring or a piperidine ring;
m and n are each independently selected from 0, 1,2 and 3, and m + n is 3;
R1、R2、R3and R4Each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, lipoheterocyclyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR' N-, RS (O)aRC (O) N (R ') -, and RR' NC (O) -; optionally, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo-aliphatic, aryl, heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-is substituted with a substituent;
R5and R6Each independently selected from the group consisting of absent, hydrogen, oxy, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloaliphatic, aryl, heteroaryl, RO-, RC (O) -, ROC (O) -, RC (O), O-, RR ' N-, RC (O) N (R) -, RR ' NC (O) -, ROC (O) N (R ') -, RR ' NC (O) N (R ' -, RS (O))a-、RR’NSO2And RS (O)2N (R') -; optionally, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo-aliphatic, aryl or heteroaryl 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, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-wherein a is 0, 1 or 2; or,
R5or R6And form a polycyclic ring system on the ring X to which it is attached, wherein R is5Or R6Selected from the group consisting of 3-8 membered alicyclic rings, 5-8 membered alicyclic heterocyclic rings, 6-10 membered aromatic rings and 5-10 membered heteroaromatic rings;
r, R 'and R' are each independently selected from hydrogen, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocyclo, 6-20 membered aryl and 5-20 membered heteroaryl; optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered cycloalkylThe aliphatic heterocyclic group, 6-20 membered aryl group, 5-20 membered heteroaryl group are each independently selected from one or more (e.g., 1,2,3 or 4) halogen, hydroxy, amino, cyano, carboxy, nitro, C1-4Alkyl, halo C1-4Alkyl, hydroxy C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-is substituted.
In certain preferred embodiments of the present application, the compound of formula I-A is limited with the proviso that ring X is not a pyrrolidine ring, a 2-pyrrolidinone ring, a morpholine ring, a 3-morpholinone ring, a tetrahydrofuran ring, a 2-oxazolidone ring, a tetrahydrothiophene ring, and a thietanesulfone ring.
In certain preferred embodiments of the present application, the compounds of formula I-A wherein A and B together with the atoms to which they are attached form an aromatic ring (e.g., a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, or a 6-10 membered aromatic ring), wherein the aromatic ring is unsubstituted or each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (e.g., fluorine, chlorine, bromine, or iodine), hydroxy, amino, cyano, carboxy, nitro and C1-10Alkyl substituents.
In certain preferred embodiments of the present application, the compounds of formula I-a wherein a and B together with the atoms to which they are attached form a phenyl ring, wherein the phenyl ring is unsubstituted or each independently substituted with one or more (e.g., 1,2,3 or 4) substituents selected from halogen (e.g., fluorine).
In certain preferred embodiments of the present application, R in the compound of formula I-A1、R2、R3And R4Each independently selected from hydrogen, halogen, hydroxyl, amino, cyano, carboxyl, nitro, C1-10Alkyl and halo C1-10An alkyl group.
In certain preferred embodiments of the present application, R in the compound of formula I-A1、R2、R3And R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, C1-6Alkyl (e.g. methyl) and halo C1-6Alkyl (e.g., trifluoromethyl).
In the present applicationIn certain preferred embodiments, R in the compound of formula I-A1、R2And R3Is a hydrogen atom.
In certain preferred embodiments of the present application, R in the compound of formula I-A4Is halogen.
In certain preferred embodiments of the present application, R in the compound of formula I-A4Is fluorine.
In certain preferred embodiments of the present application, the compounds of formula I-A wherein A and B together with the atoms to which they are attached form a phenyl ring, R1、R2And R3Is hydrogen, and R4Is fluorine.
In certain preferred embodiments of the present application, R in the compound of formula I-A5Selected from hydrogen, oxy, halogen, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-、RR’NSO2-, optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy and 3-8 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-6Alkyl and 3-8 membered cycloalkyl, optionally, wherein said C1-6Alkyl and 3-8 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-4Alkyl radical, C1-4Alkoxy and C1-4A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compound of formula I-A5Selected from hydrogen, halogen, C1-6Alkyl, 3-12 membered cycloalkyl, RC (O) -and RR' NC (O) -, optionally, wherein said C1-6Alkyl and 3-12 membered cycloalkyl are each independently selected from halogen (preferably fluoro), hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-4Alkyl, aryl, heteroaryl, and heteroaryl,C1-4Alkoxy and 3-8 membered cycloalkyl;
wherein R and R' are each independently selected from hydrogen, C1-6Alkyl and 3-8 membered cycloalkyl.
In certain preferred embodiments of the present application, R in the compound of formula I-A5Selected from hydrogen, halogen, C1-4Alkyl, 3-6 membered cycloalkyl, RC (O) -and RR' NC (O) -, optionally, wherein said C1-4Each alkyl group is independently substituted with one or more (e.g. 1,2,3 or 4) substituents selected from halo (preferably fluoro) and hydroxy;
wherein R and R' are each independently selected from hydrogen, C1-4Alkyl and 3-6 membered cycloalkyl.
In certain preferred embodiments of the present application, R in the compound of formula I-A5Selected from the group consisting of hydrogen, methyl, trifluoromethyl, cyclopropylmethyl, 2-hydroxy-2-propyl, cyclopropyl, cyclopropylformyl, aminoacyl, carbamyl and dimethylaminyl.
In certain preferred embodiments of the present application, R in the compound of formula I-A6Absent or selected from hydrogen and halo C1-10An alkyl group.
In certain preferred embodiments of the present application, R in the compound of formula I-A6Absent or hydrogen.
In certain preferred embodiments of the present application, R in the compound of formula I-A6Is absent.
In certain preferred embodiments of the present application, R in the compound of formula I-A6Is hydrogen.
In certain preferred embodiments of the present application, ring X in the compound of formula I-a is a five-membered heteroaromatic ring and the heteroatom is nitrogen, or a six-membered nitrogen-containing aliphatic heterocycle; such as a pyrazole ring or a piperidine ring.
In certain preferred embodiments of the present application, m-1, n-2; alternatively, m is 2 and n is 2.
In certain preferred embodiments of the present application, among the compounds of formula I-ASelected from the following structures:
in certain preferred embodiments of the present application, the polycyclic ring system in the compound of formula I-a is selected from the following structures:
in certain embodiments herein, the polycyclic ring system may have two chiral centers (chiral atoms are indicated by the symbol), and it will be understood by those skilled in the art that the compounds herein encompass all configurations thereof, for example, the (R, R), (R, S), (S, S) or (S, R) configuration.
In certain embodiments of the present application, the compounds have the structure shown in formula I-B,
wherein,
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 halo, hydroxy, amino, cyano, carboxy, nitro, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, alicyclic, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR ' N-, RC (O) N (R ' -, RR ' NC (O) -, and RS (O))a-wherein a is 0, 1 or 2;
ring X is an alicyclic ring, an alicyclic heterocyclic ring, an aromatic ring or a heteroaromatic ring; preferably, the ring X is a nitrogen-containing aliphatic heterocycle or heteroaromatic ring; preferably, the ring X is a five-or six-membered nitrogen-containing aliphatic heterocycle or heteroaromatic ring; more preferably, the ring X is a five or six membered nitrogen containing heteroaromatic ring; for example, a pyrrole ring, an imidazole ring, a pyrazole ring, a triazole ring or a pyridone ring;
m and n are each independently selected from 0, 1,2 and 3, and m + n is 3 or 4;
R1、R2、R3and R4Each independently selected from the group consisting of hydrogen, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, lipoheterocyclyl, aryl, heteroaryl, RO-, ROC (O) -, RC (O), O-, RC (O) -, RR' N-, RS (O)a-RC (O) N (R ') -and RR' NC (O) -; optionally, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo-aliphatic, aryl, heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) substituents selected from halogen, hydroxy, amino, cyano, carboxy, nitro, C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-wherein a is 0, 1 or 2;
R5and R6Each independently selected from the group consisting of absent, hydrogen, oxy, halogen, hydroxy, amino, cyano, carboxy, nitro, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloaliphatic, aryl, heteroaryl, RO-, RC (O) -, ROC (O) -, RC (O), O-, RR ' N-, RC (O) N (R) -, RR ' NC (O) -, ROC (O) N (R ') -, RR ' NC (O) N (R ' -, RS (O))a-、RR’NSO2And RS (O)2N (R') -; optionally, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo-aliphatic, aryl or heteroaryl 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, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-wherein a is 0, 1 or 2; or,
R5or R6And form a polycyclic ring system on the ring X to which it is attached, wherein R is5Or R6Selected from the group consisting of 3-8 membered alicyclic rings, 5-8 membered alicyclic heterocyclic rings, 6-10 membered aromatic rings and 5-10 membered heteroaromatic rings;
r, R 'and R' are each independently selected from hydrogen, C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocyclo, 6-20 membered aryl and 5-20 membered heteroaryl; renOptionally, wherein the C1-10Alkyl, 3-20 membered cycloalkyl, 5-20 membered heterocyclylalkyl, 6-20 membered aryl or 5-20 membered heteroaryl 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, halo C1-4Alkyl, hydroxy C1-4Alkyl radical, C1-4Alkoxy, 3-8 membered cycloalkyl, 5-8 membered heterocyclo, 6-10 membered aryl, 5-10 membered heteroaryl and RSO2-is substituted.
In certain preferred embodiments of the present application, the compounds of formula I-B are provided with the proviso that,
when A and B form an unsubstituted phenyl ring with the carbon atom to which they are attached, R1、R2And R3Are all H, R4In the case of F, the ratio of the total of the components is,
1) and in formula I-BIs composed ofWhen R is6Is absent, R5Is not H, hydroxy, methyl, isopropyl, cyclopropyl, trifluoromethyl, difluoromethyl, or trifluoroethyl;
2) and in formula I-BIs composed ofWhen R is5And R6Not all are H, and when R5Or R6One is H, the other is not trifluoromethyl or ethoxycarbonyl;
4) The ring X is not a pyrrolidine ring, a 2-pyrrolidinone ring, a morpholino ring, a 3-morpholino ring, and a 2-oxazolidone ring; and,
5) the compound of formula I-B is not: 4- { [3- (3, 4-dihydro-1H-pyrrolo [1,2-a ] pyrazine-2-carbonyl) -4-fluoro-phenyl ] methyl } -2H-phthalazin-1-one, 4- { [3- (6, 8-dihydro-5H- [1,2,4] triazolo [1,5-a ] pyrazine-7-carbonyl) -4-fluoro-phenyl ] methyl } -2H-phthalazin-1-one, 4- { [ 4-fluoro-3- (2-trifluoromethyl-6, 8-dihydro-5H- [1,2,4] triazolo [1,5-a ] pyrazine-7-carbonyl) -phenyl ] methyl } -2H-phthalazin-1-one, And 4- [ 4-fluoro-3- (5, 6-dihydro-8H-imidazo [1,5-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one.
In certain preferred embodiments of the present application, the compounds of formula I-B wherein A and B together with the atoms to which they are attached form an aromatic ring (e.g., a 6-20 membered aromatic ring, a 6-14 membered aromatic ring, or a 6-10 membered aromatic ring), wherein the aromatic ring is unsubstituted or each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (e.g., fluorine, chlorine, bromine, or iodine), hydroxy, amino, cyano, carboxy, nitro and C1-10Alkyl substituents.
In certain preferred embodiments of the present application, the compounds of formula I-B wherein a and B together with the atoms to which they are attached form a phenyl ring, wherein the phenyl ring is unsubstituted or each independently substituted with one or more (e.g., 1,2,3 or 4) substituents selected from halo (e.g., fluoro).
In certain preferred embodiments of the present application, R in the compounds of formula I-B1、R2、R3And R4Each independently selected from hydrogen, halogen, hydroxyl, amino, cyano, carboxyl, nitro, C1-10Alkyl and halo C1-10An alkyl group.
In certain preferred embodiments of the present application, R in the compounds of formula I-B1、R2、R3And R4Each independently selected from hydrogen, halogen, hydroxy, amino, cyano, C1-6Alkyl (e.g. methyl) and halo C1-6Alkyl (e.g., trifluoromethyl).
In certain preferred embodiments of the present application, R in the compounds of formula I-B1、R2And R3Is a hydrogen atom.
In certain preferred embodiments of the present application, R in the compounds of formula I-B4Is halogen.
In certain preferred embodiments of the present application, R in the compounds of formula I-B4Is fluorine.
In certain preferred embodiments of the present application, the compounds of formula I-B wherein A and B together with the atoms to which they are attached form a phenyl ring, R1、R2And R3Is hydrogen, and R4Is fluorine.
In certain preferred embodiments of the present application, R in the compounds of formula I-B5Selected from hydrogen, oxy, halogen, C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-、RR’NSO2-, optionally, wherein said C1-10Alkyl, 3-20 membered cycloalkyl, 3-20 membered heterocyclylalkyl, 5-10 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (preferably fluorine), hydroxy, amino, cyano, C1-6Alkyl radical, C1-6Alkoxy and 3-8 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-6Alkyl and 3-8 membered cycloalkyl, optionally, wherein said C1-6Alkyl and 3-8 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-4Alkyl radical, C1-4Alkoxy and C1-4A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compounds of formula I-B5Selected from hydrogen, oxy, halogen, C1-6Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocyclylalkyl, 5-6 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, and RS (O)a-, optionally, wherein said C1-6Alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocyclylalkyl, 5-6 membered heteroaryl are each independently substituted with one or more (e.g., 1,2,3, or 4) groups selected from halogen (preferably fluorine), hydroxy, ammoniaRadical, cyano radical, C1-4Alkyl radical, C1-4Alkoxy and 3-8 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-4Alkyl and 3-6 membered cycloalkyl, optionally, wherein said C1-4Alkyl and 3-6 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-4Alkyl radical, C1-4Alkoxy and C1-4A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compounds of formula I-B5Selected from hydrogen, oxy, halogen, C1-4Alkyl, 3-6 membered cycloalkyl, 3-6 membered heterocyclylalkyl, 5-6 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, and RS (O)a-, optionally, wherein said C1-4Alkyl, 3-6 membered cycloalkyl, 3-6 membered lipoheterocyclyl, 5-6 membered heteroaryl are each independently substituted with one or more (e.g. 1,2,3 or 4) substituents selected from halo (preferably fluoro), hydroxy, amino and 3-6 membered cycloalkyl wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-4Alkyl and 3-6 membered cycloalkyl, optionally, wherein said C1-4Alkyl and 3-6 membered cycloalkyl are each independently selected from halogen, hydroxy, amino, cyano, C by one or more (e.g. 1,2,3 or 4)1-4Alkyl radical, C1-4Alkoxy and C1-4A substituent of alkylsulfonyl.
In certain preferred embodiments of the present application, R in the compounds of formula I-B5Selected from the group consisting of hydrogen, bromine, hydroxymethyl, 2-hydroxy-2-propyl, 1-hydroxyethyl, difluoromethyl, trifluoromethyl, 1-difluoroethyl, pentafluoroethyl, 1-aminoethyl, cyclopropylmethyl, cyclopropyldifluoromethyl, cyclopropyl, 1-methyl-1H-pyrazol-4-yl, 1-methyl-1H-pyrazol-3-yl, 3-methyl- [1,2,4]Oxadiazol-5-yl, 5-methyl- [1,3,4]]Oxadiazol-2-yl, methoxy, cyclopropyloxy, ethoxycarbonyl, cyclopropylamino, aminoacyl, methionyl, ethylacyl, N-dimethylaminoyl, 2-hydroxyethylaminoacyl2-methoxyacetamido, 2-hydroxyisobutylaminoacyl, 2-methanesulfonylacetamido, methylthio and methanesulfonyl.
In certain preferred embodiments of the present application, R in the compounds of formula I-B6Absent or selected from hydrogen and halo C1-10An alkyl group.
In certain preferred embodiments of the present application, R in the compounds of formula I-B6Absent or selected from hydrogen and halo C1-6An alkyl group.
In certain preferred embodiments of the present application, R in the compounds of formula I-B6Is absent.
In certain preferred embodiments of the present application, R in the compounds of formula I-B6Selected from hydrogen and halogeno C1-4Alkyl (e.g., trifluoromethyl).
In certain preferred embodiments of the present application, m-1, n-2; alternatively, m is 2 and n is 2.
In certain preferred embodiments of the present application, ring X in formula I-B is a five-membered heteroaromatic ring and the heteroatom is nitrogen, or a six-membered nitrogen-containing aliphatic heterocycle, preferablySelected from the following structures:
in certain preferred embodiments herein, the hydrogen described in any of the first aspects of the present application is protium (H) or deuterium (D).
In certain preferred embodiments of the present application, the polycyclic ring system is selected from the following structures:
a compound, a prodrug, a metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystal form of the foregoing, according to any one of the first aspects of the present application, wherein the compound is selected from:
4- [ 4-fluoro-3- (2,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 1);
4- [ 4-fluoro-3- (2,4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 2);
4- [ 4-fluoro-3- (6, 7-dihydro-4H- [1,2,3] triazolo [1,5-a ] pyrazine-5-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 3);
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxamide (compound 4);
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (compound 5);
4- [3- (2-Cyclopropylformyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 6);
4- [3- (2-cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 7);
4- [ 4-fluoro-3- (2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 8);
4- [3- (2-Cyclopropylformyl-2, 4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 9);
cis-4- [ 4-fluoro-3- (4,5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 10);
trans-4- [ 4-fluoro-3- (4,5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 11);
4- [3- (1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 12);
7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 13);
n, N-dimethyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 14);
ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinylmethyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate (compound 15);
4- [ 4-fluoro-3- (3-trifluoromethyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 16);
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxamide (compound 17);
ethyl 7- [ 2-fluoro-5- (4-oxa-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylate (compound 18);
4- [3- (2-cyclopropyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorophenyl ] -2H-phthalazin-1-one (compound 19);
n-methyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 20);
6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 21);
4- [ 4-fluoro-3- (2-hydroxymethyl-6, 7-dihydro-4H-pyrazolo [1,5-a ] pyrazine-5-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 22);
4- [3- (3-bromo-6, 7-dihydro-4H- [1,2,3] triazolo [1,5-a ] pyrazine-5-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 23);
n- (2-hydroxyethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (compound 24);
n, N-dimethyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 25);
4- [ 4-fluoro-3- (6-oxo-1, 3,4, 6-tetrahydro-pyrido [1,2-a ] pyrazine-2-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 26);
4- [3- (3-bromo-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 27);
4- [3- (3-bromo-6, 7,8, 9-tetrahydro-5H- [1,2,4] triazolo [4,3-d ] [1,4] diazepine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 28);
4- { 4-fluoro-3- [3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 29);
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxamide (compound 30);
n-ethyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxamide (compound 31);
4- { 4-fluoro-3- [3- (1-hydroxy-1-methyl-ethyl) -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 32);
4- [ 4-fluoro-3- (3-methoxy-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 33);
n- (2-methoxyethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (compound 34);
n- (2-hydroxy-2-methylpropyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (compound 35);
n- (2-methylsulfonylethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (compound 36);
4- [3- (3-acetyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 37);
4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 38);
4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 39);
4- [ 4-fluoro-3- (3-methylthio-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 40);
4- [ 4-fluoro-3- (3-methylsulfonyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 41);
4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 42);
4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -1,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 43);
4- { 4-fluoro-3- [3- (1-hydroxyethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 44);
4- { 4-fluoro- [ (3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 45);
4- (3- (3- (1, 1-difluoroethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl) phthalazin-1 (2H) -one (compound 46);
4- [3- (3-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 47);
4- [3- (3-cyclopropyl-5, 6-dihydro-8H-imidazo [1,5-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 48);
4- [3- (3-cyclopropyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 49);
4- [3- (3-cyclopropyl-2-trifluoromethyl-5, 6 dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 50);
4- { 4-fluoro-3- [3- (3-methyl- [1,2,4] -oxadiazol-5-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 51);
4- { 4-fluoro-3- [3- (5-methyl- [1,3,4] -oxadiazol-2-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 52);
4- [3- (3-cyclopropylamino-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 53);
4- [3- (3-cyclopropylmethyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 54);
4- {3- [3- (cyclopropyldifluoromethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 55);
4- [3- (3-cyclopropyl-6, 7-dihydro-4H- [1,2,3] triazolo [1,5-a ] pyrazine-5-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 56);
4- [3- (3-cyclopropoxy-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 57);
4- {3- [3- (1-aminoethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one (compound 58);
4- [3- (3-cyclopropyl-1-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,5-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 59);
4- {3- [3- (1, 1-difluoroethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -4-fluorobenzyl } -7-fluoro-2H-phthalazin-1-one (compound 60);
4- [3- (3-cyclopropyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -7-fluoro-2H-phthalazin-1-one (compound 61);
4- [3- (3-difluoromethyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -7-fluoro-2H-phthalazin-1-one (compound 62);
4- [ 4-fluoro-3- (3-perfluoroethyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 63);
7-fluoro-4- [ 4-fluoro-3- (3-pentafluoroethyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one (Compound 64); and
4- [ 4-fluoro-3- (6-methyl-3-pentafluoroethyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 65).
A second aspect of the present application provides a process for the preparation of a compound as described in any one of the first aspects 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, selected from the following processes:
(1) when R in formula I5To RR' NC (O) -the compounds can be synthesized by the following route:
carrying out condensation reaction on the compound A and the compound B' to obtain a compound C; carrying out urethane exchange on the compound C and a compound RR' NH to obtain a compound D;
(2) when R in formula I5To rc (o) -the compound can be synthesized by the following route:
carrying out condensation reaction on the compound A and the compound B 'to obtain a compound C'; performing acylation reaction on the compound C 'and a compound RCO-Lg to obtain a compound D'; or,
(3) when R in formula I5And R6As with the other groups described in the first aspect of the application, the compounds may be synthesized by the following route:
carrying out condensation reaction on the compound A and the compound B to obtain a compound shown in a formula I;
wherein Lg in RCO-Lg represents a leaving group, e.g., halogen, -OTs, -OCOR, etc., and the remaining atoms and substituents are as defined in the first aspect of the application.
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, compound B' or compound B "(free amine or salt, e.g. hydrochloride) in a solvent (e.g. DMF or THF), adding 0.8-1.2 equivalents of a condensing agent (e.g. HATU, EDCI, T3P, etc.) and 2.0-5.0 equivalents of a base (e.g. DIPEA, Et) at room temperature3N, pyridine and the like), adding water and an organic solvent (such as ethyl acetate or methyl tert-butyl ether) into 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 product.
Compound B is commercially available or prepared by routine experimentation in the art. In certain preferred embodiments of the present application, the synthesis of compound B includes, but is not limited to, the following methods:
(1) reacting compound B-1 with Lg-R5Or-R5Nucleophilic substitution reaction is carried out to obtain a compound B-2; compound B-2 deaminating the protecting group to give compound B:
(2) carrying out Suzuki reaction on the compound B-3 and the compound B-4 to obtain a compound B-2; compound B-2 deaminating the protecting group to give compound B:
(3) carrying out addition/reduction reaction on the compound B-5 to obtain a compound B-2; compound B-2 deaminating the protecting group to give compound B:
wherein Lg is a leaving group for nucleophilic substitution reaction, such as halogen, -OTs, -OCOR, etc.; p is a common amino protecting group such as Boc, Fmoc, Cbz, Alloc, Pht, Tos, Dmb, Bn, etc.; r5' is a compound which can be subjected to a one-step addition/reduction reaction to give R as described herein5Such as ester group, acyl group, etc., and a reagent for the addition/reduction reaction such as a Grignard reagent, NaBH4、NaBH3CN, etc., and the remaining atoms or substituents are as defined in the first aspect of the application.
In certain preferred embodiments of the present application, deprotection of the amino group can be achieved by methods known in the art, and the present application is not limited thereto.
In certain preferred embodiments of the present application, the compound B may be synthesized by:
the method comprises the following steps:
dissolving the intermediate 1 and a base (such as triethylamine, DIPEA, potassium carbonate and the like) in a solvent (such as dichloromethane, tetrahydrofuran, DMF and the like), dropwise adding an acyl chloride solution (the solvent can adopt dichloromethane, tetrahydrofuran, acetonitrile and the like) at low temperature or room temperature for reaction, extracting, separating liquid, purifying to obtain intermediates 2-1 and 2-2, and removing protecting groups of the obtained intermediates to obtain target intermediates;
the second method comprises the following steps:
the intermediate 1 is dissolved in a solvent (e.g., diethyl ether, tetrahydrofuran, etc.), and then after adding a base (e.g., NaH, t-BuOK, LDA, LiHMDS, etc.) at low temperature or room temperature (-80 ℃ to room temperature) and reacting at this temperature for 10 to 30 minutes, a solution of a halogenated hydrocarbon (e.g., diethyl ether, tetrahydrofuran, etc.) is added dropwise. After the reaction is finished, extracting, drying and purifying to obtain intermediates 3-1 and 3-2, and removing protecting groups of the obtained intermediates to obtain target intermediates;
the third method comprises the following steps:
dissolving intermediate 1 in solvent (such as toluene, dioxane, DMF, etc.) and adding base (such as sodium carbonate, potassium phosphate, potassium carbonate, etc.), adding palladium catalyst (such as Pd) into the obtained mixture under inert gas protection2(dba)3,Pd(OAc)2,Pd(dppf)Cl2Etc.) and a ligand (e.g., dppf, xanthphos, x-phos, etc.), the reaction can be carried out at room temperature or under heating (60-150 ℃) or under heating in a microwave reactor, after the reaction is finished, the intermediates 4-1 and 4-2 can be obtained through liquid separation, extraction and purification, and the target intermediates can be obtained after the protecting groups of the obtained intermediates are removed.
The method four comprises the following steps:
dissolving SM-1 in a solvent (e.g., carbon tetrachloride, chloroform, dichloromethane, etc.) and then adding a brominating agent (e.g., bromine, NBS, etc.) at room temperature or low temperature to give brominated compound Int-1. The compound is reacted with 2-aminopyrazine in a solvent (such as THF, methanol, ethanol, etc.) by adding a base (such as potassium carbonate, triethylamine, etc.) at 60-120 ℃. After the reaction is finished, the compound Int-2 can be obtained through post-treatment and purification. The intermediate is dissolved in a solvent (such as methanol, ethanol, isopropanol and the like) for catalytic hydrogenation to obtain the target product with ester groups, and acid (such as acetic acid, hydrochloric acid and the like) can be added to accelerate or promote the reaction;
the compound Int-2 and amine are dissolved in a solvent (such as THF, methanol, ethanol, DMF, DMSO, and the like) to carry out aminolysis at room temperature or at the temperature of 60-150 ℃ with or without adding a base (such as triethylamine, DIPEA, and the like), and after purification, the compound Int-3 can be obtained, the reaction can be carried out in a solvent-free mode or a microwave reactor, the Int-3 can be subjected to catalytic hydrogenation in the solvent (such as methanol, ethanol, isopropanol, and the like) to obtain an amide target intermediate, and the reaction can be accelerated or promoted by adding an acid (such as acetic acid, hydrochloric acid, and the like);
the method five comprises the following steps:
dissolving pyrazine-2-hydrazine and a base (such as triethylamine, DIPEA and the like) which are used as starting materials in a solvent (such as dichloromethane, THF, acetonitrile and the like), adding oxalyl chloride monoester at low temperature or room temperature, carrying out aftertreatment and purification after the reaction is finished to obtain Int-4, dissolving the compound in a solvent (methanol, ethanol, DMF and the like) and reacting under heating (40-150 ℃), wherein the base (such as sodium methoxide, sodium ethoxide, sodium tert-butoxide and the like) can be added or not added. After the reaction is finished, carrying out post-treatment and purification to obtain a ring-closed product, carrying out catalytic hydrogenation on the product in a solvent (such as methanol, ethanol, isopropanol and the like) to obtain a target product with an ester group, and adding an acid (such as acetic acid, hydrochloric acid and the like) to accelerate or promote the reaction;
int-5 is subjected to aminolysis with various amines in solvents (such as THF, methanol, ethanol, DMF, DMSO, etc.) at room temperature or elevated temperature (60-150 deg.C), optionally with a base (such as triethylamine, DIPEA, etc.) or without a base, and purified to give Int-6. The reaction can also be carried out in a solvent-free manner or in a microwave reactor, and Int-6 can be subjected to catalytic hydrogenation in a solvent (such as methanol, ethanol, isopropanol and the like) to obtain a target amide intermediate, and an acid (such as acetic acid, hydrochloric acid and the like) can also be added to accelerate or promote the reaction.
The method six:
dissolving N-protected 3-piperidone in a solvent (such as THF, diethyl ether and the like), adding a base (such as KHMDS, LiHMDS, LDA and the like) at low temperature (-90-0 ℃) or room temperature, reacting for 30 minutes at the temperature, adding an ester, carrying out post-treatment and purification after the reaction is finished to obtain a 1, 3-diketone derivative, dissolving the derivative in a solvent (such as DCM, THF, ethanol and the like) and an aqueous solution, an alcoholic solution or a hydrochloride of hydrazine to carry out ring closure to obtain an intermediate pyrazole derivative, and removing a protecting group from the intermediate to obtain the target intermediate.
The method comprises the following steps:
when R in formula I5And form a polycyclic ring system on the ring X to which it is attached, Compound B can be synthesized by:
dissolving the starting material in a solvent (e.g., toluene, THF, etc.) and then adding a thioreagent (e.g., Lawson's reagent, P)2S5Etc.) reacting at normal temperature or at elevated temperature, purifying to obtain an intermediate with thio carbonyl, dissolving the intermediate and acylhydrazine in a proper solvent (such as methanol, ethanol, etc.), adding alkali (such as triethylamine, DIPEA, sodium methoxide, etc.) to react at room temperature or elevated temperature (40-120 ℃) or under microwave, adding equivalent mercury acetate, mercury chloride mercury salt or silver carbonate silver salt to accelerate or promote the reaction, after the reaction is completed, carrying out post-treatment and purification to obtain a ring-closing triazole intermediate, and removing a protecting group to obtain the target intermediate.
The method eight:
dissolving pyrazine-2-hydrazine and substituted difluoroacetic acid (SM-2) which are starting materials in strong acid (such as polyphosphoric acid and the like) and then performing cyclization under heating conditions (such as 90-150 ℃), performing post-treatment and purification after the reaction is completed to obtain an intermediate INT-1, dissolving the intermediate INT-2 in a solvent (such as methanol, ethanol, isopropanol and the like), and performing catalytic hydrogenation in the presence of hydrogen by using a catalyst (such as palladium carbon, palladium hydroxide carbon and the like) to obtain a target intermediate INT-2, wherein acid (such as acetic acid, hydrochloric acid and the like) can be added to accelerate or promote the reaction;
alternatively, pyrazine-2-hydrazine starting material and substituted difluoroacetic anhydride (SM-3) may be condensed in a solvent (e.g., dichloromethane, tetrahydrofuran, etc.) to provide the hydrazide intermediate INT-3. The product is dissolved in strong acid (such as polyphosphoric acid/polyphosphoric acid and the like) and then subjected to ring closure under heating (such as 90-150 ℃), and after the reaction is finished, the intermediate INT-1 is obtained through post-treatment and purification.
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, prodrugs, metabolite forms, pharmaceutically acceptable salts or esters thereof, or isomers, hydrates, solvates or crystal forms of the foregoing, 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 prodrugs, metabolite forms, pharmaceutically acceptable salts or esters, or the aforementioned isomers, hydrates, solvates or crystal forms, 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 certain preferred 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, a prodrug, metabolite form, a pharmaceutically acceptable salt or ester thereof, or an isomer, hydrate, solvate or crystalline form 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).
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 group being unsubstituted or further substituted by a substituentOr substituted alkyl, said substituent being selected from the group consisting of halogen, hydroxy, alkoxy, cycloalkyl, lipoheterocyclyl, aryl or heteroaryl. Non-limiting examples of substituted alkyls include halo C1-4Alkyl, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropan-2-yl, 3-hydroxypropyl, methoxymethyl, 2-methoxyethyl, 3-methoxypropyl, ethoxymethyl, 2-ethoxyethyl, 3-ethoxypropyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2-cyclopentylethyl, 2-cyclohexylethyl, 3-cyclopropylpropyl, 3-cyclobutylpropyl, 3-cyclopentylpropyl, 3-cyclohexylpropyl, benzyl, pyridine-2-methyl 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 groups, non-limiting examples of which 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 C2-4Alkynyl groups, non-limiting examples include ethynyl, propynyl, butynyl, pentynyl, 3-methylbutynyl, hexynyl, 3-methylpentylenyl and the like.
The term "cycloalkyl" as used herein refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon group, such as a cycloalkyl group containing 3 to 20 carbon atoms, such as a cycloalkyl group containing 3 to 12 carbon atoms, such as a cycloalkyl group containing 3 to 10 carbon atoms, such as a cycloalkyl group containing 3 to 8 carbon atoms, such as a cycloalkyl group containing 3 to 6 carbon atoms, such as a cycloalkyl group containing 5 to 8 carbon atoms. Non-limiting examples of monocycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like. Polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups.
The term "lipoheterocyclyl" as used herein refers to a saturated or partially saturated cyclic hydrocarbon group containing at least one heteroatom selected from N, O and S. For example, a heterocyclolipidyl group containing 3-20, 3-12, 3-8, 3-6, 5-20, 5-12, 5-8, or 5-6 ring members. Non-limiting examples of lipoheterocyclyl groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like.
The term "aryl" as used herein refers to a monocyclic or fused polycyclic (e.g., bicyclic or tricyclic) group having aromatic character, preferably 6-20 membered aryl, preferably 6-14 membered aryl, more preferably 6-10 membered aryl, non-limiting examples of which include, but are not limited to, phenyl, naphthyl and the like.
The term "heteroaryl" as used herein refers to a 5-20 membered aromatic heterocyclic group, preferably a 5-10 membered, preferably a 5-6 membered heteroaryl group, substituted with at least one heteroatom selected from N, O or S. Non-limiting examples of heteroaryl groups include furyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, 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.
Non-limiting examples of "RO-" herein include methoxy, ethoxy, propoxy, and cyclopropoxy groups, and the like.
Non-limiting examples of "ROC (O) -" herein include methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, and tert-butoxycarbonyl, and the like.
Non-limiting examples of "RC (O) O-" herein include formyloxy, acetoxy, propionyloxy, and the like.
Non-limiting examples of "RC (O) -" herein include formyl, acetyl, propionyl, and cyclopropaneformyl, and the like.
Non-limiting examples of "RR' N" herein include methylamino, dimethylamino, ethylamino, diethylamino, and the like.
Non-limiting examples of "RC (O) N (R") - "herein include carboxamido, acetylamino, propionamido and the like.
Non-limiting examples of "RR' NC (O) -" herein include methionyl, N-dimethycinyl, ethylacyl, 2-hydroxyethylaminoacyl, 2-methoxyethylacyl, 2-hydroxyisobutylaminoacyl, 2-methanesulfonylethylacyl, and the like.
In this text "RS (O)a- "includes substituted sulfonyl, substituted sulfinyl, and substituted mercapto, where non-limiting examples of substituted sulfonyl include methanesulfonyl, ethanesulfonyl, benzenesulfonyl, p-toluenesulfonyl, and the like. Non-limiting examples of substituted sulfinyl groups include methylsulfinyl, ethylsulfinyl, phenylsulfinyl, p-tolylsulfinyl, and the like. Non-limiting examples of substituted mercapto groups include methylmercapto, phenylmercapto, and the like.
Here, "RR' NS (O)2- "denotes substituted aminosulfonyl, RR' NS (O)2Non-limiting examples of (A) includeMethylaminosulfonyl and the like.
In this text "RS (O)2N (R') - "refers to substituted sulfonamide groups, such as alkylsulfonamide groups, arylsulfonamide groups, and the like. RS (O)2Non-limiting examples of N (R') -, include methanesulfonamide group, ethanesulfonamido group, benzenesulfonamide group, p-toluenesulfonamide group and the like.
The term "oxy" as used herein means especiallyThe oxy group may be substituted on a carbon atom of a cycloalkyl, an alicyclic, an aryl, a heteroaryl group or the like to form a carbonyl group or may exist in an enol form, for example, to form a structure of pyrrolidone, pyridin-2-one, phthalazin-1-one or the like.
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,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 term "room temperature" as used herein, also referred to as ambient or general temperature, generally refers to room temperature, preferably from 20 ℃ to 38 ℃, e.g., from 20 ℃ to 30 ℃, e.g., from 20 ℃ to 25 ℃.
The compounds of formula i or pharmaceutically acceptable salts thereof herein may also form solvates, such as hydrates, alcoholates and the like. The compound of formula i or a pharmaceutically acceptable salt thereof herein may also exist as crystals, which means an arrangement in which molecules, atoms or ions constituting the compound are spatially repeated at regular intervals, and the arrangement has a periodicity in 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 the structure-activity relationship of phthalazinone PARP inhibitors, and the compound can realize at least one of the following technical effects:
(1) can obviously improve the proliferation inhibition effect on tumor 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;
(5) the compound has good bioavailability, inhibition effectiveness and safety;
(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.
Drawings
FIG. 1 is a NOE spectrum of the product of example 19 herein.
Detailed Description
Embodiments of the present application 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 application and should not be construed as limiting the scope of the present application.
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 (DMS)O-d6), internal standard Tetramethylsilane (TMS), chemical shift is 10-6(ppm) is given as a unit.
MS was determined 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 eluent system comprises a dichloromethane and methanol system and a normal hexane and ethyl acetate system, 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.
The reaction temperature is room temperature (20 ℃ C. -30 ℃ C.) unless otherwise specified in the examples, and is carried out according to 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.
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;
NOE: nuclear Overhauser Effect, orfrassel Nuclear Effect;
MeOH: methanol;
THF: tetrahydrofuran;
EtOH: ethanol;
DMF: n, N-dimethylformamide;
NMP: n-methyl pyrrolidone;
HATU: 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate;
LCMS: a liquid phase mass spectrometer;
NaH: sodium hydrogen;
TLC: thin layer chromatography;
T3P propyl phosphoric anhydride.
Example 1:
4- [ 4-fluoro-3- (2,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) benzyl ] -2H-phthalazin-1-one (Compound 1)
The reaction route is as follows:
the compound 5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (50mg,0.17mmol), HATU (70mg,0.18mmol) and diisopropylethylamine (0.58mL,3.5mmol) were dissolved in DMF (5mL), stirred at room temperature for 30min and then 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine (22mg,0.18mmol) was added and stirred at room temperature until LCMS indicated complete consumption of starting material. The reaction solution was poured into a saturated aqueous ammonium chloride solution, and extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate, concentrated, and the residue was purified by thin layer silica gel plate to obtain the objective product (59mg, yield: 86.1%).
MS m/z(ESI):404[M+H]+
1HNMR(400MHz,CD3OD)δ:8.38-8.34(m,1H),7.97-7.93(m,1H),7.93-7.81(m,2H),7.52-7.36(m,3H),7.20-7.13(m,1H),4.90(m,1H),4.39-4.37(m,3H),3.49-3.48(t,1H,J=2Hz),3.30(m,1H),2.78-2.75(t,1H,J=6Hz),2.53(br s,1H).
Example 2:
4- [ 4-fluoro-3- (2,4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) benzyl ] -2H-phthalazin-1-one (Compound 2)
The title compound was synthesized by the method of example 1 substituting 4,5,6, 7-tetrahydro-2H-pyrrolo [4,3-c ] pyridine for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):404[M+H]+.
1HNMR(400MHz,CD3OD)δ:8.39-8.34(m,1H),8.00-7.78(m,3H),7.55-7.31(m,2H),7.28-7.10(m,2H),5.00-4.82(s,1H),4.78(s,1H),4.44-4.30(m,2H),3.85-3.51(m,1H),3.40-3.23(m,1H),2.82-2.79(m,1H),2.72-2.53(m,1H).
Example 3:
4- [ 4-fluoro-3- (6, 7-dihydro-4H- [1,2,3] triazolo [1,5-a ] pyrazine-5-carbonyl) benzyl ] -2H-phthalazin-1-one (Compound 3)
The title compound was synthesized by the method of example 1 substituting 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1 with 4,5,6, 7-tetrahydro- [1,2,3] triazolo [1,5-a ] pyrazine.
MS m/z(ESI):405[M+H]+.
1HNMR(400MHz,CDCl3)δ:8.47-8.41(m,1H),7.82-7.70(m,4H),7.67-7.65(m,1H),7.57(s,1H),7.32-7.25(m,1H),4.72(s,2H),4.34(s,2H),3.93(t,2H,J=4Hz),3.72(t,2H,J=4Hz).
Example 4:
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxamide (Compound 4)
The first step is as follows: the compound 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylic acid methyl ester
Synthesized by the procedure of example 1,4,5, 6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1 was replaced with methyl 5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazine-2-carboxylate.
The second step is that: the compound N-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydrophthalazine-1-methyl) benzoyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxamide
The compound of the first step, methyl 7- [ 2-fluoro-5- (4-oxo-3, 4 dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate (100mg,0.21mmol) was dissolved in a solution of methylamine in methanol (30%, 5mL) and the resulting solution was reacted at 85 ℃ overnight. After completion of the reaction monitored by TLC, the reaction solution was concentrated, and the residue was poured into water and extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate, concentrated and purified by preparative thin layer chromatography to give 20mg of the target product in yield: 20 percent.
MS m/z(ESI):461[M+H]+。
1HNMR(400MHz,CD3OD)δ:8.38-8.34(m,1H),8.00-7.78(m,3H),7.59-7.42(m,3H),7.23-7.15(m,1H),4.91(s,1H),4.58(s,1H),4.44-4.35(d,2H,J=4Hz),4.10-3.98(m,1H),3.80-3.68(m,1H),3.40-3.25(m,2H),2.96-2.78(m,3H).
Example 5:
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 5)
The reaction route is as follows:
the first step is as follows: compound 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester
Synthesized by the method of example 1,4,5, 6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1 was replaced with ethyl 5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] piperazine-3-carboxylate.
The second step is that: compound N-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-formamide
The second step of the synthesis of example 4 was used, replacing the methyl 7- [ 2-fluoro-5 (4-oxo-3, 4-dihydro-phthalazine 1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 with the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,4] triazolo [4,3-a ] pyrazine-3-carboxylate of the first step.
MS m/z(ESI):462[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.97-8.82(m,1H),8.34-8.19(m,1H),8.04-7.78(m,3H),7.57-7.42(m,2H),7.35-7.22(m,1H),5.13-4.84(br s,1H),4.66(s,1H),4.41-4.23(m,4H),3.70-3.58(m,1H),3.56-3.40(m,1H),2.78-2.76(d,3H,J=8Hz).
Example 6:
4- [3- (2-Cyclopropylformyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 6)
The reaction route is as follows:
the first step is as follows: 4- [ 4-fluoro-3- (2,4,5, 6-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -benzyl ] -2H-phthalazinyl-1-one
The procedure of example 1 was followed, substituting 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
The second step is that: 4- [3- (2-Cyclopropylformyl-2, 4,5, 6-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one
4- [ 4-fluoro-3- (2,4,5, 6-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -benzyl ] -2H-phthalazinyl-1-one (30mg,0.074mmol) and triethylamine (22.42mg,0.222mmol) of the first step were dissolved in tetrahydrofuran (2mL), and cyclopropylcarbonyl chloride (11.6mg,0.111mmol) was added dropwise under ice bath. After 10 minutes the reaction was complete and quenched with ice water. The aqueous phase was extracted with ethyl acetate, and the organic layer was dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography to give the desired product in yield: 57 percent.
MS m/z(ESI):472[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.66(br s,1H),8.29-8.19(m,2H),8.02-7.77(m,3H),7.50-7.19(m,3H),5.03-4.29(m,4H),3.48-3.36(m,2H),3.14-2.95(m,1H),2.77-2.53(m,2H),1.20-1.03(m,4H).
Example 7:
4- [3- (2-Cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 7)
The reaction route is as follows:
the first step is as follows: 2-Cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carboxylic acid tert-butyl ester
NaH (256.8mg,6.42mmol) was added to a solution of tert-butyl 2,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonate (250mg,1.07mmol) in N, N-dimethylformamide (5mL), and the resulting mixture was reacted at room temperature for 1 hour, followed by addition of cyclopropylmethyl bromide (288.9mg,2.14 mmol). After 6 hours at room temperature, the reaction was quenched with water and extracted with dichloromethane. The dichloromethane layer was dried over anhydrous sodium sulfate and concentrated to give the title compound (150mg crude) which was used directly in the next reaction.
The second step is that: 2-cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine hydrochloride
The first step of tert-butyl 2-cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonate was placed in a reaction flask and then a solution of hydrogen chloride in ethyl acetate (4N,3mL) was added. The mixture was stirred at room temperature overnight and the product obtained by concentration was used directly in the next reaction.
The third step: 4- [3- (2-cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one
5- [ (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl ] -2-fluorobenzoic acid (129.3mg,0.43mmol), the second step of 2-cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine hydrochloride (76.7mg,0.43mmol), HATU (179.8mg,0.47mmol) and DIPEA (332.8mg,2.58mmol) 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 (5mg, yield: 2.5%).
MS m/z(ESI):458[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.65(br s,1H),8.30-8.18(m,2H),8.02-7.75(m,3H),7.50-7.20(m,3H),5.05-4.31(m,4H),3.48-3.36(m,4H),3.15(m,2H),1.43-1.38(m,1H),1.20-1.03(m,4H).
Example 8:
4- [ 4-fluoro-3- (2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 8)
Reaction scheme
The first step is as follows: 2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carboxylic acid tert-butyl ester
NaH (108mg,2.70mmol) was added to a solution of tert-butyl 2,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonate (100mg,0.45mmol) in N, N-dimethylformamide (5mL), and the resulting mixture was reacted at room temperature for 1 hour, followed by addition of methyl iodide (127.8mg,0.90 mmol). After 7 hours at room temperature, the reaction was quenched with water and extracted with dichloromethane. The dichloromethane layer was dried over anhydrous sodium sulfate, and the concentrated residue was purified by preparative thin layer chromatography to give the product (75mg, yield: 70%).
The second step is that: 2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine hydrochloride
The first step of tert-butyl 2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonate was placed in a reaction flask and then a solution of hydrogen chloride in ethyl acetate (4N,3mL) was added. The mixture was stirred at room temperature overnight and the product obtained by concentration was used directly in the next reaction.
The third step: 4- [ 4-fluoro-3- (2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -benzyl ] -2H-phthalazin-1-one
5- [ (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl ] -2-fluorobenzoic acid (95.4mg,0.32mmol), the second step of 2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine hydrochloride (43mg,0.32mmol), HATU (146mg,0.38mmol) and DIPEA (206.4mg,1.60mmol) 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 (30mg, yield: 23%).
The nmr spectrum showed that the sample was a mixture of two isomers in a ratio of 3: 1.
MS m/z(ESI):418[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.71(br s,1H),8.27-8.25(d,1H,J=8Hz),8.02-7.95(m,1H),7.93-7.80(m,2H),7.51-7.32(m,3H),7.31-7.18(m,1H),4.82-4.60(m,1H),4.41-4.22(m,3H),3.90-3.64(m,3H),3.39-3.29(m,2H),2.70-2.53(m,1H),2.46-2.38(m,1H).
Example 9:
4- [3- (2-Cyclopropylformyl-2, 4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 9)
Synthesized by the method of the second step of example 6 from 4- [ 4-fluoro-3- (2,4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) -benzyl ] -2H-phthalazin-1-one and cyclopropanecarbonyl chloride.
MS m/z(ESI):472[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.69(br s,1H),8.30-8.25(m,1H),8.00-7.98(m,1H),7.96-7.83(m,2H),7.49-7.30(m,3H),7.29-7.18(m,1H),4.85-4.58(m,1H),4.40-4.25(m,3H),3.90-3.64(m,2H),3.39-3.29(m,2H),2.58-2.47(m,1H),1.15-1.00(m,4H).
Example 10:
cis-4- [ 4-fluoro-3- (4,5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 10)
The reaction route is as follows:
the procedure of example 1 was followed to replace the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1 with cis-4, 5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine (see ACS Med. chem. Lett. (DOI: 10.1021/ml500503n) for synthesis).
MS m/z(ESI):459[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.68-12.55(m,1H),8.53-8.44(m,1H),8.32-8.20(m,1H),8.05-7.73(m,3H),7.63-7.16(m,3H),4.58-4.43(m,1H),4.40-4.27(m,3H),3.28-2.92(m,5H),2.87-2.72(m,1H),2.13-1.64(m,4H).
Example 11:
trans-4- [ 4-fluoro-3- (4,5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (Compound 11)
The reaction route is as follows:
synthesized using the experimental procedure of example 1, using trans-4, 5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine) as described in ACS med. 10.1021/ml500503n)) in place of the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):459[M+H]+。
1HNMR(400MHz,DMSO-d6)δ:12.61(br s,1H),8.53(d,1H,J=24Hz),8.31-8.23(m,1H),8.01-7.80(m,3H),7.53-7.30(m,2H),7.28-7.20(m,1H),4.75-4.59(m,1H),4.38(s,2H),3.96-3.81(m,1H),3.57-3.14(m,3H),3.11-2.58(m,4H),1.82-1.34(m,3H)。
Example 12:
4- [3- (1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 12)
The reaction route is as follows:
the first step is as follows: 1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carboxylic acid tert-butyl ester
The compound 1,4,5, 7-tetrahydro-pyrazolo [3,4-c]Pyridine-6-carbonic acid tert-butyl ester (200mg,2.32mmol) was completely dissolved in 5mL1, 4-dioxane, cyclopropylboronic acid (250mg,1.12mmol), copper acetate (200mg,1.10mmol), pyridine (87mg,1.10mmol) and DMAP (403.15mg,3.30mmol) were added and the resulting reaction mixture was stirred at 100 ℃ overnight until the consumption of the starting material was complete, followed by extraction with dichloromethane, back washing with saturated brine, drying over sodium sulfate and concentration under reduced pressure to give a brown oil, and the residue was purified by preparative thin layer chromatography to give the desired product (160mg, yield: 26.2%). Regioisomeric chemistry of cyclopropyl by example 191NOE experiments with H NMR confirmed (no correlation peak between hydrogen in cyclopropyl and pyrazoline hydrogen).
The second step is that: 1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine hydrochloride
Tert-butyl 1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonate (160mg,0.61mmol) 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.
The third step: 4- [3- (1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one
The synthesis was performed as described in example 1, substituting the second step of 1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine hydrochloride for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):444[M+H]+。
1HNMR(400MHz,CD3OD)δ:8.39-8.32(m,1H),8.02-7.95(m,1H),7.92-7.81(m,2H),7.64-7.55(m,2H),7.35-7.22(m,1H),5.21-5.02(m,1H),4.84-4.76(m,2H),4.43(s,2H),4.17-3.79(m,6H),2.28-2.09(m,2H),2.01-1.90(m,2H)。
Example 13:
7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 13)
The title compound was synthesized by the method of example 4 substituting ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate used in example 4 for methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 and 7mol/L of methanolic ammonia for 30% methanolic methylamine used in example 9.
MS m/z(ESI):448[M+H]+。
1HNMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.31-8.23(m,2H),8.01-7.80(m,4H),7.55-7.45(m,2H),7.32-7.24(m,1H),5.00(br s,1H),4.40-4.24(m,4H),4.13-4.05(m,1H),3.67-3.60(m,1H),3.17(d,2H,J=4Hz)。
Example 14:
n, N-dimethyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] - -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 14)
The title compound was synthesized by the method of example 4 substituting ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate used in example 4 for methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methyl-benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 and dimethylamine solution for the 30% methylamine methanol solution used in example 4.
MS m/z(ESI):476[M+H]+。
1HNMR(400MHz,CDCl3)δ:8.45-8.41(m,1H),7.87-7.81(m,1H),7.75-7.69(m,3H),7.65-7.61(m,1H),7.28-7.22(m,1H),4.46(s,2H),4.34(s,2H),4.27(t,2H,J=4Hz),3.72(t,2H,J=4Hz),3.09(s,6H)。
Example 15:
7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinylmethyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester (Compound 15)
The title compound was synthesized by the method of example 1 substituting ethyl 5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):477[M+H]+。
1HNMR(400MHz,CD3OD)δ:8.39-8.32(m,1H),7.98-7.79(m,3H),7.56-7.43(m,2H),7.25-7.17(m,1H),5.12(s,1H),4.79(s,1H),4.51-4.30(m,6H),3.80-3.73(m,1H),1.45-1.34(m,3H).
Example 16:
4- [ 4-fluoro-3- (3-trifluoromethyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 16)
The title compound was synthesized by the method of example 1 substituting 3-trifluoromethyl-4, 5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] piperidine for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):476[M+H]+。
1HNMR(400MHz,CDCl3)δ:8.49-8.46(m,1H),7.78-7.71(m,3H),7.65-7.61(m,1H),7.59-7.56(m,1H),7.31-7.25(m,1H),4.72(s,2H),4.34(s,2H),3.94(t,2H,J=4Hz),3.16(t,2H,J=4Hz)。
Example 17:
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxamide (Compound 17)
The title compound was synthesized by the procedure of example 4 substituting the intermediate ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylate used in example 4 for methyl 7- [ 2-fluoro-5 (4-oxo-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate.
MS m/z(ESI):461[M+H]+。
1HNMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.29-8.20(m,2H),8.03-7.80(m,3H),7.59-7.43(m,3H),7.33-7.23(m,1H),4.83(brs,1H),4.48(br s,1H),4.37-4.29(m,3H),4.28-4.22(m,1H),3.66-3.59(m,1H),2.72(d,2H,J=4Hz)。
Example 18:
7- [ 2-fluoro-5- (4-oxa-3, 4-dihydro-1-phthalazinyl) methylbenzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylic acid ethyl ester (Compound 18)
The title compound was synthesized using the experimental procedure of example 1 substituting 2-trifluoromethyl-5, 6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazine-3-carboxylic acid ethyl ester for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):490[M+H]+。
1HNMR(400MHz,CD3OD)δ:8.40-8.31(m,1H),8.00-7.78(m,3H),7.57-7.43(m,2H),7.26-7.13(m,1H),4.97(s,1H),4.64(s,1H),4.52-4.28(m,6H),3.80-3.67(m,2H),3.27-3.19(m,1H),1.38-1.33(m,3H)。
Example 19:
4- [3- (2-cyclopropyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorophenyl ] -2H-phthalazin-1-one (compound 19)
The title compound was synthesized using the procedure of example 12. Regioisomeric chemical passage of cyclopropyl1NOE of H NMR was experimentally determined (see FIG. 1 for results).
MS m/z(ESI):444[M+H]+。
1HNMR(400MHz,DMSO-d6)δ:12.58(brs,1H),8.27-8.25(m,1H),7.99-7.96(m,1H),7.91-7.81(m,2H),7.55(s,1H),7.44-7.43(m,1H),7.37-7.35(m,1H),7.27-7.21(m,1H),4.64-4.61(m,1H),4.33-4.26(m,3H),3.84-3.57(m,2H),2.60-2.55(m,1H),2.45-2.40(m,2H),1.23(s,1H),1.00-0.85(m,4H)。
Example 20:
n-methyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 20)
The first step is as follows: 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrrolo [3,4-c ] pyridine-3-carboxylic acid ethyl ester
The synthesis was carried out using an experimental procedure analogous to that of example 1, substituting 4H-pyrrole-3-carboxylic acid ethyl ester for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
The second step is that: n-methyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine-3-carboxamide
Synthesized in analogy to the experimental procedure of example 4, the methyl 7- [ 2-fluoro-5 (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrrolo [3,4-c ] pyridine-3-carboxylate used in example 4 was replaced with the first step ethyl 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate.
MS m/z(ESI):461[M+H]+
1H NMR(400MHz,DMSO-d6)δ:13.02(br s,1H),12.60(br s,1H),8.27-8.25(m,1H),8.02-7.82(m,3H),7.46-7.41(m,2H),7.29-7.24(m,1H),4.82-4.70(m,1H),4.40-4.30(m,2H),3.45-3.38(m,3H),2.78-2.76(m,2H),2.71(d,2H,J=4Hz).
Example 21:
6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 21)
The first step is as follows: 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrrolo [3,4-c ] pyridine-3-carboxylic acid ethyl ester
See example 20 for synthesis.
The second step is that: 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide
Synthesized in analogy to the experimental procedure of example 4, the methyl 7- [ 2-fluoro-5 (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrrolo [3,4-c ] pyridine-3-carboxylate used in example 4 was replaced with the ethyl 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate of the first step and the methylamine solution used in example 4 was replaced with aqueous ammonia.
MS m/z(ESI):447[M+H]+
1HNMR(400MHz,CD3OD)δ:8.38-8.35(m,1H),7.99-7.81(m,3H),7.50-7.47(m,1H),7.40-7.38(m,1H),7.20-7.16(m,1H),5.35-5.33(m,1H),4.40-4.36(m,2H),3.57-3.28(m,3H),2.79-2.76(m,2H).
Example 22:
4- [ 4-fluoro-3- (2-hydroxymethyl-6, 7-dihydro-4H-pyrazolo [1,5-a ] pyrazine-5-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 22)
The title compound was synthesized by the method of example 1 substituting 4,5,6, 7-tetrahydro-pyrrolo [1,5-a ] pyrazin-2-yl) -methanol for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):434[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.27(d,1H,J=8Hz),7.98(t,1H,J=8Hz),7.92-7.82(m,2H),7.50-7.38(m,2H),7.30-7.24(m,1H),6.12(s,0.6H),5.94(s,0.4H),5.02-4.97(m,1H),4.84(br s,1H),4.47(br s,1H),4.37-4.33(m,4H),4.10(br s,1H),3.99-3.96(m,1H),3.67-3.65(m,1H).
Example 23:
4- [3- (3-bromo-6, 7-dihydro-4H- [1,2,3] triazolo [1,5-a ] pyrazine-5-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 23)
The title compound was synthesized by the method of example 1 substituting 3-bromo-4, 5,6, 7-tetrahydro- [1,2,3] triazolo [1,5-a ] pyrazine for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):483[M+H]+
1HNMR(400MHz,DMSO-d6)δ:8.38-8.34(m,1H),7.98-7.94(m,1H),7.90-7.80(m,2H),7.56-7.52(m,1H),7.48-7.44(m,1H),7.21(t,1H,J=9Hz),4.89(m,1H),4.54-4.50(m,1H),4.41-4.36(m,3H),3.83-3.79(m,1H),2.99(s,1H),2.86(s,1H).
Example 24:
n- (2-hydroxyethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 24)
The first step is as follows: 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester
See example 5 for a first step procedure for synthesis.
The second step is that: n- (2-hydroxyethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide
Synthesized by the procedure of example 4, using the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate of the first step instead of the methyl 7- [ 2-fluoro-5- (4-oxo-3, 4 dihydro-1-phthalazin-1-yl) -methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 and ethanolamine instead of the methylamine solution used in example 4.
MS m/z(ESI):492[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.79-8.73(m,1H),8.26(d,1H,J=8Hz),7.97(t,1H,J=8Hz),7.93(t,1H,J=7Hz),7.84(t,1H,J=7Hz),7.53-7.46(m,2H),5.00(br s,1H),4.77(t,1H,J=6Hz),4.68-4.60(m,1H),4.40-4.25(m,4H),3.68-3.62(m,1H),3.54-3.48(m,2H),4.41-4.36(m,3H),3.83-3.79(m,1H),3.35-3.28(m,2H).
Example 25:
n, N-dimethyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 25)
The first step is as follows: 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrrolo [3,4-c ] pyridine-3-carboxylic acid ethyl ester
See example 20 for synthesis.
The second step is that: n, N-dimethyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide
Synthesized by the procedure of example 4, using the first step of ethyl 6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrrolo [3,4-c ] pyridine-3-carboxylate instead of the methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazin-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4, and dimethylamine instead of the methylamine solution used in example 4.
MS m/z(ESI):475[M+H]+
1HNMR(400MHz,DMSO-d6)δ:13.02(br s,1H),12.59(br s,1H),8.28-8.25(m,1H),8.02-7.82(m,3H),7.46-7.41(m,2H),7.29-7.24(m,1H),4.82-4.70(m,1H),4.40-4.32(m,2H),3.55-3.46(m,6H),2.78-2.76(m,2H),2.71(d,2H,J=4Hz).
Example 26:
4- [ 4-fluoro-3- (6-oxo-1, 3,4, 6-tetrahydro-pyrido [1,2-a ] pyrazine-2-carbonyl) benzyl ] -2H-phthalazin-1-one (Compound 26)
The title compound was synthesized by the method of example 1 substituting 1,2,3, 4-tetrahydro-pyrido [1,2-a ] pyrazin-6-one for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):431[M+H]+
1H NMR(400MHz,DMSO-d6)δ:8.39-8.35(m,1H),7.98-7.94(m,2H),7.90-7.80(m,2H),7.58-7.37(m,3H),7.24-7.16(m,1H),6.50(d,1H,J=9Hz),6.46-6.02(m,1H),4.85(brs,1.2H),4.44(br s,0.8H),4.42-4.37(m,3H),4.21(t,1H,J=5Hz),3.91(t,1H,J=6Hz),3.62(t,1.2H,J=5Hz).
Example 27:
4- [3- (3-bromo-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 27)
The title compound was synthesized by the method of example 1 substituting 3-bromo-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):483[M+H]+
1HNMR(400MHz,CD3OD)δ:8.36(t,1H,J=8Hz),7.99-7.80(m,3H),7.57-7.50(m,1H),7.46-7.41(m,1H),7.24-7.17(m,1H),5.10-5.00(m,1H),4.76-4.59(m,1H),4.42-4.35(m,2H),4.26-3.93(m,4H).
Example 28:
4- [3- (3-bromo-6, 7,8, 9-tetrahydro-5H- [1,2,4] triazolo [4,3-d ] [1,4] diazepine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 28)
The title compound was synthesized by the method of example 1 substituting 3-bromo-6, 7,8, 9-tetrahydro-5H- [1,2,4] triazolo [4,3-d ] [1,4] diazepine for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):497[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.34(m,1H),7.99-7.80(m,3H),7.55-7.49(m,1H),7.43-7.39(m,1H),7.23-7.16(m,1H),4.42-4.33(m,4H),4.10-3.95(m,2H),3.38-3.54(m,2H),3.30-2.25(m,1H),3.04-3.00(m,1H).
Example 29:
4- { 4-fluoro-3- [3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 29)
The reaction route is as follows:
the first step is as follows: 3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carboxylic acid tert-butyl ester
The compound, methyl 7-tert-butoxycarbonyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carbonate (200mg, 0.71mmol), was completely dissolved in 5mL of dry THF, 3mol/L methylmagnesium bromide (2.37mL, 7.1mmol) was slowly added under nitrogen at room temperature, after the addition was complete, the mixture was stirred to 45 ℃ by LCMS to monitor the completion of the reaction. After cooling to room temperature, adding saturated ammonium chloride solution to quench the reaction, extracting with dichloromethane, backwashing with saturated saline solution, drying with sodium sulfate, and concentrating under reduced pressure to obtain brown oily substance which can be directly used for the next reaction without purification.
The second step is that: 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol
Completely dissolving the compound obtained in the previous step in 10mL of dichloromethane, adding 1mL of trifluoroacetic acid, turning to 35 ℃, stirring until the reaction is completely monitored by LCMS, concentrating under reduced pressure to remove dichloromethane and trifluoroacetic acid, and directly using the crude product in the next reaction.
The third step: 4- { 4-fluoro-3- [3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
Synthesized using the procedure of example 1, substituting 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):462[M+H]+
1HNMR(400MHz,CD3OD)δ:8.39-8.34(m,1H),7.98-7.94(m,1H),7.92-7.80(m,2H),7.54-7.41(m,2H),7.23-7.16(m,1H),6.83-6.76(m,1H),4.90-4.87(m,1H),4.57-4.54(m,1H),4.43-4.36(m,3H),4.27-4.14(m,2H),3.73-3.68(m,1H),1.57(s,6H).
Example 30:
n-methyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxamide (compound 30)
The first step is as follows: 7- [ 2-fluoro-5- (4-oxa-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylic acid ethyl ester
See example 18 for synthesis.
The second step is that: synthesized using the method of the second step of example 4. The methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylate used in the second step of example 4 was replaced by the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate of the first step.
MS m/z(ESI):529[M+H]+
1HNMR(400MHz,CD3OD)δ:8.39-8.35(m,1H),7.98-7.94(m,1H),7.91-7.80(m,2H),7.55-7.49(m,1H),7.46-7.43(m,1H),7.23-7.17(m,1H),4.99-4.95(m,1H),4.62-4.59(m,1H),4.42-4.37(m,2H),4.22-3.70(m,4H).
Example 31:
n-ethyl-7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxamide (compound 31)
The first step is as follows: 7- [ 2-fluoro-5- (4-oxa-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylic acid ethyl ester
See example 18 for synthesis.
The second step is that: synthesized using the method of the second step of example 4. The methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylate used in the second step of example 4 was replaced by the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate of the first step.
MS m/z(ESI):543[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.34(m,1H),7.99-7.80(m,3H),7.56-7.42(m,2H),7.23-7.17(m,1H),4.99-4.95(m,1H),4.63-4.58(m,1H),4.40(br s,2H),4.25-3.72(m,4H),3.44-3.35(q,2H,J=7Hz),1.20(t,3H,J=7Hz).
Example 32:
4- { 4-fluoro-3- [3- (1-hydroxy-1-methyl-ethyl) -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 32)
The reaction route is as follows:
the first step is as follows: 3- (1-hydroxy-1-methyl-ethyl) -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carboxylic acid tert-butyl ester
Synthesized using the procedure of the first step of example 29.
The second step is that: 2- (2-trifluoromethyl-5, 6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol
Synthesized using the method of the second step of example 29.
The third step: 4- { 4-fluoro-3- [3- (1-hydroxy-1-methyl-ethyl) -2-trifluoromethyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
Synthesized using the procedure of example 1, substituting 2- (2-trifluoromethyl-5, 6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol for the 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine used in example 1.
MS m/z(ESI):529[M+H]+
1HNMR(400MHz,CD3OD)δ:8.40-8.34(m,1H),7.99-7.79(m,3H),7.56-7.42(m,2H),7.23-7.16(m,1H),4.99-4.95(m,1H),4.63-4.58(m,1H),4.44-4.38(m,2H),4.25-3.72(m,4H),1.30(s,6H).
Example 33:
4- [ 4-fluoro-3- (3-methoxy-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (Compound 33)
The reaction route is as follows:
the first step is as follows: 3-methoxy-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate the compound 3-bromo-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate (100mg,0.33mmol) was completely dissolved in 5mM My OH, sodium methoxide (54mg,1mmol) was added and stirred at room temperature until LCMS monitors the reaction completion. Extraction with dichloromethane, washing with saturated brine, drying over sodium sulfate and concentration under reduced pressure gave a white solid (84mg, yield 99%).
The second step is that: 3-methoxy-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine
Synthesized using the method of the second step of example 32.
The third step: 4- [ 4-fluoro-3- (3-methoxy-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one
The procedure of example 1 was followed, using 3-methoxy-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine instead of 4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine as used in example 1.
MS m/z(ESI):435[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.30-8.24(m,1H),7.99-7.80(m,3H),7.52-7.40(m,2H),7.32-7.24(m,1H),4.95-4.70(m,1H),4.33(s,3H),4.15-3.98(m,4H),3.82-3.55(m,3H).
Example 34:
n- (2-methoxyethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 34)
The first step is as follows: 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester
See example 5 for synthesis.
The second step is that: n- (2-methoxyethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide
The second step of the synthesis of example 4 was carried out by substituting the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate used in example 4 for the methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazin-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 and methoxyethylamine for the methylamine solution used in example 4.
MS m/z(ESI):506[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.89-8.82(m,1H),8.29-8.24(m,1H),8.00-7.80(m,3H),7.53-7.45(m,2H),7.31-7.25(m,1H),5.08-4.64(m,2H),4.40-4.04(m,6H),3.50-3.38(m,2H),3.26(s,3H).
Example 35:
n- (2-hydroxy-2-methylpropyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 35)
The first step is as follows: 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester
See example 5 for synthesis.
The second step is that: n- (2-hydroxy-2-methylpropyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide
The second step of the synthesis of example 4 was carried out by substituting the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate used in example 4 for the methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazin-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 and 1-amino-2-methylpropyl-2-ol for the methylamine solution used in example 4.
MS m/z(ESI):520[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.47-8.38(m,1H),8.28-8.23(m,1H),8.00-7.80(m,3H),7.52-7.45(m,2H),7.31-7.24(m,1H),5.08-4.40(m,1H),4.70-4.24(m,6H),3.67-3.60(m,1H),3.26(s,3H),3.24(d,2H,J=6Hz),1.11(s,6H).
Example 36:
n- (2-methylsulfonylethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide (Compound 36)
The first step is as follows: 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester
See example 5 for synthesis.
The second step is that: n- (2-methylsulfonylethyl) -7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazine-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxamide
The second step of the synthesis of example 4 was carried out by substituting the ethyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylate used in example 4 for the methyl 7- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-1-phthalazin-methyl) -benzoyl ] -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-2-carboxylate used in example 4 and 2- (methylsulfonyl) ethylamine for the methylamine solution used in example 4.
MS m/z(ESI):554[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),9.16-9.08(m,1H),8.26(d,1H,J=8Hz),8.00-7.80(m,3H),7.52-7.45(m,2H),7.32-7.25(m,1H),5.08-4.64(m,2H),4.40-4.04(m,4H),3.72-3.35(m,6H),3.04(s,3H).
Example 37:
4- [3- (3-acetyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 37)
The reaction route is as follows:
the first step is as follows: 3-acetyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester
Synthetic procedure referring to the first step of example 29, 7-tert-butoxycarbonyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylic acid methyl ester of the first step of example 29 was substituted with 7-tert-butyl-3-ethyl-5, 6-dihydro- [1,2,4] triazolo [4,3-a ] pyrazine-3, 7(8H) -dicarboxylate and the amount of methyl Grignard reagent used was reduced to 2 equivalents.
The second step is that: 1- (5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine-3-yl) ethanone trifluoroacetate, a compound 3-acetyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate obtained in the first step is dissolved in dichloromethane, and then trifluoroacetic acid is added to be heated until the reaction is complete. The reaction mixture was concentrated and the resulting mixture was used directly in the next reaction.
The third step: 4- [3- (3-acetyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one
See example 29 for synthesis procedure in the third step, 1- (5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazin-3-yl) ethanone trifluoroacetate was used in place of 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate used in the third step of example 29.
MS m/z(ESI):447[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.29-8.24(m,2H),7.99-7.81(m,3H),7.52-7.43(m,2H),7.32-7.25(m,1H),5.03-4.70(m,2H),4.39-4.15(m,4H),3.97-3.58(m,2H),2.64(s,3H).
Example 38:
4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 38)
The reaction route is as follows:
the first step is as follows: 3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carboxylic acid tert-butyl ester
1-methyl-1H-pyrazole-4-boronic acid (265mg,2.1mmol), 3-bromo-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carboxylic acid tert-butyl ester (604mg, 2mmol) were dissolved in DME (5mL), degassed under ultrasound and then replaced with nitrogen and triphenylphosphine palladium chloride (30mg) and 2N aqueous sodium carbonate solution (1mL) were added. The mixture was reacted at 85 ℃ for 5 hours, after completion of the reaction water (3mL) was added and extracted with dichloromethane (10mLx 4). The dichloromethane layer was dried over anhydrous magnesium sulfate, concentrated and purified by thin layer TLC to give 200mg of the target product, yield: 66 percent.
The second step is that: 3- (1-methyl-1H-pyrazol-4-yl) -5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazine hydrochloride
The product obtained in the first step was dissolved in 2mL of methanol, then a saturated solution of hydrogen chloride in 1, 4-dioxane (2mL,4N) was added and the reaction was completed at room temperature, the solvent was distilled off under reduced pressure, and the residue was used directly in the next reaction.
The third step: 4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
For synthesis see the third step of example 29, 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate in example 29 is replaced with 3- (1-methyl-1H-pyrazol-4-yl) -5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazine hydrochloride.
MS m/z(ESI):485[M+H]+
1H NMR(400MHz,CD3OD)δ:8.36-8.30(m,1H),7.96-7.77(m,4H),7.68-7.59(m,1H),7.55-7.41(m,2H),7.24-7.14(m,1H),7.03-6.95(m,1H),4.96-4.90(m,1H),4.63-4.35(m,3H),4.27-4.09(m,2H),3.99-3.88(m,4H),3.77-3.70(m,1H)。
Example 39:
4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 39)
The reaction route is as follows:
the first step is as follows: 3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate
The synthesis method is that 3-bromo-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate in example 38 is replaced by 3-bromo-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-tert-butyl carbonate in the first step of example 38.
The second step is that: 3- (1-methyl-1H-pyrazol-4-yl) -5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine hydrochloride
For synthesis, referring to the second step of example 38, tert-butyl 3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonate was used in place of tert-butyl 3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonate of example 38.
The third step is 4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
For synthesis, see the third step of example 29, 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate in example 29 is replaced with 3- (1-methyl-1H-pyrazol-4-yl) -5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine hydrochloride.
MS m/z(ESI):486[M+H]+
1H NMR(400MHz,CD3OD)δ:8.37-8.30(m,1H),8.20-8.10(m,1H),7.97-7.77(m,4H),7.58-7.44(m,2H),7.25-7.17(m,1H),5.13-5.04(m,1H),4.95-4.90(m,1H),4.79-4.70(m,1H),4.41-4.36(m,2H),4.32-4.21(m,1H),4.12-4.04(m,1H),4.00-3.97(m,3H),3.83-3.74(m,1H)。
Example 40:
4- [ 4-fluoro-3- (3-methylthio-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 40)
The reaction route is as follows:
the first step is as follows: 3-Methylthio-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester
The synthesis is described in example 33 in the first step, sodium methoxide in example 33 is replaced by sodium methyl mercaptide and the solvent is DMF.
The second step is that: 3-methylmercapto-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine trifluoroacetate
See example 33 for synthesis in the second step.
The third step: 4- [ 4-fluoro-3- (3-methylmercapto-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one
See example 33 for procedure for synthesis.
MS m/z(ESI):451[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.59(br s,1H),8.29-8.24(m,1H),7.99-7.80(m,3H),7.52-7.43(m,2H),7.34-7.23(m,1H),4.60-4.39(m,3H),3.96-3.91(m,1H),3.75-3.58(m,3H),3.19-3.15(m,1H),2.61-2.56(s,3H)。
Example 41:
4- [ 4-fluoro-3- (3-methylsulfonyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (Compound 41)
The reaction route is as follows:
the first step is as follows: 3-Methylthio-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester
The synthesis is described in example 33 in the first step, sodium methoxide in example 33 is replaced by sodium methyl mercaptide and the solvent is DMF.
The second step is that: 3-methylsulfonyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester
At 0 ℃, 10mL of 3-methylmercapto-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate (270mg, 1mmol) dichloromethane solution is added with m-chloroperoxybenzoic acid (519mg, 3mmol), naturally warmed to room temperature, monitored by TLC tracking, and the raw material is completely reacted after 5 hours. Adding a 10% sodium sulfite solution into the reaction solution, stirring for 30 minutes, then adding a 10% sodium carbonate solution, continuing stirring for 10 minutes, extracting with dichloromethane (50 mL. times.4), combining organic phases, drying with anhydrous sodium sulfate, spin-drying, washing with methyl tert-butyl ether to obtain the target molecule, wherein the yield is as follows: 85%).
The third step: 3-methylsulfonyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine trifluoroacetate salt
See example 33 for synthesis in the second step.
The fourth step: 4- [ 4-fluoro-3- (3-methylsulfonyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one
See example 33 for procedure for synthesis.
MS m/z(ESI):483[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.28-8.24(m,1H),7.98-7.80(m,3H),7.55-7.42(m,2H),7.34-7.26(m,1H),4.75-4.69(m,1H),4.31(s,3H),4.20-4.07(m,2H),3.73-3.45(m,5H)。
Example 42:
4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 42)
The reaction route is as follows:
the first step is as follows: 3- (2-hydroxypropyl-2-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester
The synthesis procedure is as in the first step of example 29, and 7-tert-butoxycarbonyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-3-carboxylic acid ethyl ester was used instead of 7-tert-butoxycarbonyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylic acid methyl ester in example 29.
The second step is that: 2- (5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate salt
The synthesis procedure is as in the second step of example 29, 3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carboxylic acid tert-butyl ester from example 29 is replaced by 3- (2-hydroxypropyl-2-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester.
The third step: 4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
See example 29 for procedure for synthesis, 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate in example 29 was replaced with 2- (5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate.
MS m/z(ESI):463[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.28-8.24(m,1H),7.99-7.81(m,3H),7.53-7.43(m,2H),7.33-7.24(m,1H),4.95-4.54(m,2H),4.38-4.24(m,3H),4.16-4.00(m,2H),3.66-3.56(m,1H),1.52(s,6H).
Example 43:
4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -1,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 43)
The reaction route is as follows:
the first step is as follows: 4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxylic acid methyl ester hydrochloride
6-tert-Butoxycarbonyl-4, 5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxylic acid (267mg, 1mmol) is dissolved in 5mL of methanol and 1mL of thionyl chloride is slowly added. The resulting solution was refluxed overnight, after completion of the reaction monitored by LCMS, methanol was removed and the residue was used directly in the next reaction.
The second step is that: 6-Boc-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-3-carboxylic acid methyl ester
To the residue obtained in the first step (250mg) were added dichloromethane (5mL), triethylamine (404mg,4mmol) and di-tert-butyl dicarbonate (516mg,2mmol) in this order, and the mixture was reacted at room temperature overnight. Then, water was added to separate the reaction solution, and the dichloromethane layer was concentrated and purified by thin layer TLC to obtain the objective product (220mg, yield 78.3%).
The third step: 3- (2-hydroxypropyl-2-yl) -1,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carboxylic acid tert-butyl ester
For synthesis see the first step of example 29, methyl 7-tert-butoxycarbonyl-5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-3-carboxylate in example 29 was replaced with methyl 6-tert-butoxycarbonyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-3-carboxylate.
The fourth step: 2- (4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridin-3-yl) propyl-2-ol trifluoroacetate salt
Synthesis see the second step of example 29, substituting 3- (2-hydroxypropyl-2-yl) -1,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonate tert-butyl ester for 3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonate in example 29.
The fifth step: 4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -1,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one
For synthesis see the third step of example 29, 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate in example 29 is replaced by 2- (4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridin-3-yl) propyl-2-ol trifluoroacetate.
MS m/z(ESI):462[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.62(br s,1H),8.27-8.24(m,1H),8.00-7.96(m,1H),7.93-7.80(m,2H),7.49-7.20(m,3H),5.25-4.94(m,1H),4.71-4.63(m,1H),4.37-4.26(m,3H),4.01-3.73(m,1H),3.72-3.66(m,1H),3.53-3.39(m,1H),3.04(m,6H).
Example 44:
4- { 4-fluoro-3- [3- (1-hydroxyethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 44)
The reaction route is as follows:
the first step is as follows: 3- (1-hydroxyethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carboxylic acid tert-butyl ester
Dissolving 3-acetyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate (266mg,1mmol) in methanol (5mL) under ice bath, adding sodium cyanoborohydride (315mg,5mmol), naturally heating to room temperature, and stirring overnight. The reaction mixture was concentrated, water and dichloromethane were added, and the dichloromethane layer was dried over anhydrous magnesium sulfate and concentrated to obtain the objective product (250mg, yield 93%).
The second step is that: 1- (5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazin-3-yl) -ethanol trifluoroacetate salt
For synthesis, referring to the second step of example 29, tert-butyl 3- (1-hydroxy-1-methyl-ethyl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonate from example 29 was replaced with tert-butyl 3- (1-hydroxyethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonate.
The third step: 4- { 4-fluoro-3- [3- (1-hydroxyethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
For synthesis, see the third step of example 29, 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate from example 29 is replaced by 1- (5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazin-3-yl) ethanol trifluoroacetate.
MS m/z(ESI):449[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.60(br s,1H),8.30-8.23(m,1H),8.01-7.79(m,3H),7.53-7.40(m,2H),7.33-7.24(m,1H),5.65-5.54(m,1H),5.01-4.79(m,2H),4.34(s,2H),4.22-4.09(m,1H),4.05-3.92(m,2H),3.71-3.55(m,1H),1.50(d,3H,J=4Hz)。
Example 45:
4- { 4-fluoro- [ (3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (Compound 45)
The reaction route is as follows:
the first step is as follows: 1-methyl-1H-pyrazole-3-boronic acid
3-bromo-1-methyl-1H-pyrazole (322mg,2mmol), pinacol ester diboron (1.016g,4mmol) was dissolved in 1, 4-dioxane (10mL), followed by addition of potassium acetate (392mg,4mmol), degassing the resulting solution under ultrasonic waves, nitrogen substitution was performed, and palladium triphenylphosphine chloride (32mg) was added. After 5 hours of reaction at 110 degrees, the solvent was evaporated under reduced pressure, water (10mL) was added and then extracted with dichloromethane (10 mL. times.4), and the dichloromethane layer was dried over anhydrous magnesium sulfate and then spin-dried. The residue was purified by thin layer TLC to give 179mg of the title product in 71% yield.
The second step is that: 3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-tert-butyl carbonate
For synthesis see example 38 first step 1-methyl-1H-pyrazole-4-boronic acid was substituted for 1-methyl-1H-pyrazole-3-boronic acid in example 38.
The third step is 3- (1-methyl-1H-pyrazol-3-yl) -5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine hydrochloride
For synthesis, referring to the second step of example 38, tert-butyl 3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonate in example 38 was substituted with tert-butyl 3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonate.
The fourth step: 4{ 4-fluoro-3- [3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one
For synthesis, see the third step of example 29, 2- (5,6,7, 8-tetrahydro-imidazo [1,2-a ] pyrazin-3-yl) -propyl-2-ol trifluoroacetate from example 29 is replaced with 3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine hydrochloride.
MS m/z(ESI):485[M+H]+
1H NMR(400MHz,CD3OD)δ:8.42-8.30(m,1H),8.11-7.47(m,7H),7.31-7.20(m,1H),5.16-4.72(m,2H),4.49-4.35(m,4H),4.11-3.91(m,2H),3.51-3.40(m,3H)。
Example 46:
4- {3- [3- (1, 1-difluoroethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one (Compound 46)
The reaction route is as follows:
the first step is as follows: 3- (1, 1-difluoroethyl) - [1,2,4] triazolo [4,3-a ] pyrazine
The compound 2-hydrazinopyrazine (1g, 1eq) and 2, 2-difluoropropionic acid (3g, 3eq) were added to a round-bottom flask, and then PPA was added thereto, stirred well and reacted at 180 ℃ for 18 hours. LCMS monitoring was done until the reaction was complete. The reaction mixture was poured into a saturated aqueous sodium hydrogencarbonate solution, extracted with dichloromethane, and the organic layer was dried over anhydrous sodium sulfate. The organic solvent is removed by evaporation, and the residue is purified by column chromatography to obtain 450mg of 3- (1, 1-difluoroethyl) - [1,2,4] triazolo [4,3-a ] pyrazine with the yield of 27%.
The second step is that: 3- (1, 1-difluoroethyl) -5,6,7,8- [1,2,4] triazolo [4,3-a ] pyrazines
The compound 3- (1, 1-difluoroethyl) - [1,2,4] triazolo [4,3-a ] pyrazine (450mg) was dissolved in ethanol, and Pd/C (50mg) was added thereto. The reaction was carried out overnight under a hydrogen atmosphere. And (3) monitoring by LCMS (liquid crystal display system), and evaporating ethanol to dryness after the reaction is finished to obtain 462mg of 3- (1, 1-difluoroethyl) -5,6,7,8- [1,2,4] triazolo [4,3-a ] pyrazine.
The third step: 4- {3- [3- (1, 1-difluoroethyl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -4-fluorobenzyl } -2H-phthalazin-1-one (Compound 46)
The compound 3- (1, 1-difluoroethyl) -5,6,7,8- [1,2,4]Triazole [4,3-a ]]Pyrazine (462mg,1eq), 2-fluoro-5- ((4-oxo-3, 4-dihydrophthalazin-1-yl) methyl) benzoic acid (806mg, 1.1eq) was dissolved in DMF and T was added3Adding P (1.2eq), DIEA (1.27g,4eq), stirring at room temperature, LCMSDetecting the reaction to finish. The reaction mixture was poured into a saturated aqueous sodium bicarbonate solution, dichloromethane was added to extract, and the organic layer was dried over anhydrous sodium sulfate. The organic solvent was evaporated to dryness and the residue was purified by column chromatography to give 800mg of the title compound in 69.6% yield.
MS m/z(ESI):469[M+H]+
1HNMR(400MHz,DMSO-d6)δ:12.59(s,1H),8.29-8.22(m,1H),7.98-7.80(m,3H),7.54-7.45(m,2H),7.35-7.24(m,1H),5.10-4.61(m,2H),4.34(s,2H),4.27-3.96(m,3H),3.71-3.62(m,1H),2.16(t,3H,J=16Hz).
Example 47:
4- [ 4-fluoro-3- (3-pentafluoroethyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one (Compound 63)
The reaction route is as follows:
the first step is as follows: 3-pentafluoroethyl- [1,2,4] triazolo [4,3-a ] pyrazines
80mL of PPA and pentafluoropropionic acid (37.2g,227.0mmol) are added into pyrazine-2-hydrazine (10.0g,90.8mmol) at room temperature, and the temperature is raised to 110 ℃ for reaction until the pyrazine-2-hydrazine is completely reacted. The reaction mixture was slowly poured into 1L of ice water with stirring, the pH was adjusted to weak alkalinity with sodium bicarbonate, extraction was performed with ethyl acetate (150mL × 4), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and the residue obtained after concentration was purified by column chromatography (PE/EA ═ 4/1) to obtain the objective product (17.8g, yield: 82.4%).
MS m/z(ESI):239.1[M+H]+.
The second step is that: 3-pentafluoroethyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine
150ml of absolute ethanol was added to 3-pentafluoroethyl- [1,2,4] triazolo [4,3-a ] pyrazine (15.0g, 63.0mmol) at room temperature, and after stirring and clarification, 2.0g of 10% Pd/C was added, and after hydrogen substitution, the reaction was carried out at room temperature until the starting material disappeared completely. The filter cake was washed with 100ml of absolute ethanol by Celite pad filtration, and concentrated under reduced pressure to give the objective product (13.6g, yield: 88.9%) which was used in the next step without purification.
MS m/z(ESI):243.1[M+H]+.
The third step: 4- [ 4-fluoro-3- (3-pentafluoroethyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) benzyl ] -2H-phthalazin-1-one
At room temperature, 5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid (1.0g,3.35mmol), HATU (1.40g,3.69mmol) and DIPEA (649.4mg,5.03mmol) were mixed in 10mL of dmf, stirred at room temperature for 30min, then 3-pentafluoroethyl-5, 6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyrazine (0.89g,3.69mmol) of the second step was added, the reaction mixture was reacted at room temperature until 5- (3, 4-dihydro-4-oxo-1-phthalazinyl) methyl-2-fluorobenzoic acid completely disappeared, 150mL of water was added to the reaction mixture, extracted with ethyl acetate (30mL × 4), the organic phases were combined, washed with saturated water, dried over anhydrous sodium sulfate, and the concentrated residue was purified by column chromatography (DCM/Acetone ═ 3/1) to obtain the desired product (1.18g, yield: 67.4%).
MS m/z(ESI):523.2[M+H]+.
1HNMR(400MHz,DMSO-d6)δ:12.60(s,1H),8.27-8.24(m,1H),7.97-7.81(m,3H),7.53-7.44(m,2H),7.33-7.27(m,1H),5.04-4.95(m,1H),4.72-4.65(m,1H),4.35(s,2H),4.29-4.15(m,2H),4.02-3.98(m,1H),3.68-3.62(m,1H).
Referring to the synthetic methods described in the examples, the present application also synthesized the following compounds:
TABLE 1
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.
The kit comprises: PARP1 Chemilmescent Assay Kit;
the manufacturer: BPS Bioscience;
experimental methods
Enzyme label plate pretreatment: the plate was removed from the package and 50. mu.L/well of 1 XPARP buffered diluted 1 XPARN was added and incubated overnight at 4 ℃; discarding the liquid in the microplate the next day, and washing the plate with 200. mu.L/well PBST (1 XPBS and 1% Triton X-100) to remove all the washing liquid; then adding 200 mu L 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 μ L/well PBST, and finally all the wash was removed.
Reaction: the substrate, enzyme, test compound, etc. were added sequentially according to the procedure and amounts listed in the table below and incubated for 1h at room temperature.
TABLE 2 reaction System
And (3) detection: after the reaction is finished, 50 mu L of diluted Strep-HRP is added into each hole, and the mixture is cultured at room temperature; washing the well plate with PBST and removing the washing solution; HRP substrates A and B were mixed and then added to each well at 100. mu.L; and finally, detecting the PARP1 kinase inhibition activity of the compound to be detected by a chemiluminescence method (Luminometric Measurement).
As a result:
TABLE 3 Single Point concentration inhibition of tested Compounds
Compound ID | Single point concentration inhibition% |
Olaparib | 77.47%(5nM) |
Compound 1 | 88.77%(5nM) |
Compound 5 | 92.78%(5nM) |
Compound 13 | 99.70%(5nM) |
Compound 14 | 99.46%(5nM) |
Compound 15 | 99.50%(5nM) |
Compound 16 | 96.65%(5nM) |
Compound 17 | 97.88%(5nM) |
Compound 18 | 86.10%(5nM) |
Compound 19 | 90.90%(5nM) |
Compound 20 | 99.65%(5nM) |
Compound 21 | 98.97%(5nM) |
Compound 22 | 86.27%(5nM) |
Compound 23 | 78.15%(5nM) |
Compound 24 | 99.48%(5nM) |
Compound 25 | 98.13%(5nM) |
Compound 26 | 89.37%(5nM) |
Compound 27 | 92.66%(5nM) |
Compound 28 | 78.07%(5nM) |
Compound 29 | 81.90%(5nM) |
Compound 33 | 88.84%(5nM) |
Compound 34 | 88.12%(5nM) |
|
83.08%(5nM) |
As can be seen from Table 3, the compound of the application has obvious inhibitory effect on the PARP1 kinase activity, and the inhibitory activity of the compound is obviously superior to that of a positive drug.
Experimental example 2 cell proliferation experiment
The CCK-8 method was used to determine the effect of compounds on cell proliferation.
The kit comprises: CCK-8 Kit (Cell Counting Kit-8)
The manufacturer: biyuntian (Beyotime)
Experimental methods
Cell culture: cells were cultured according to the following conditions, counted using a cell counter after cell refinement, and adjusted to the desired concentration according to the following requirements, and then 100. mu.L of cells were seeded per well and administered 24 hours after seeding.
TABLE 4 cell culture conditions and plate Density
Compound preparation: after culturing for one day, dissolving the experimental drug 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 drug solution is prepared into a corresponding incubation concentration.
Incubation time: incubation will continue 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.
The results are detailed in tables 5-1, 5-2, 5-3:
table 5-1: inhibiting effect on breast cancer MDA-MB-468
The compounds in Table 5-1 all have better inhibitory effect on MDA-MB-468 tumor of breast cancer than Olaparib. The compounds of the present application have substantially similar inhibitory effects on breast cancer MDA-MB-468 tumors as described above. The compound of the application has similar inhibition effect on breast cancer MDA-MB-436 and MDA-MB-453 tumors.
Tables 5-2: inhibition of pancreatic cancer Capan-1
The compounds in the table 5-2 have better inhibiting effect on the tumor of the pancreatic cancer Capan-1 than the Olaparib. The compounds of the present application have substantially similar inhibitory effects on the tumor of the pancreatic cancer, Capan-1, as the above-mentioned compounds.
Tables 5 to 3: inhibition of HCT116 in colorectal cancer
The compounds in the table 5-3 have better inhibiting effect on colorectal cancer HCT116 tumor than Olaparib. The compounds of the present application have substantially similar inhibitory effects on colorectal cancer HCT116 tumors as the above-mentioned compounds.
In conclusion, the compound disclosed by the application has excellent inhibitory activity on different cancer cells such as breast cancer, colorectal cancer and pancreatic cancer, and shows good antitumor effect.
Experimental example 3 Pharmacokinetic (PK) study of Compounds
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 dosages of IV and PO were 1 and 5mg/kg, respectively, and the vehicle system was 10% DMSO: 10% solutol: 80% normal saline. IV and PO administration, collecting blood at different time points, anticoagulating with heparin sodium, centrifuging to obtain plasma sample, and storing at-80 deg.C. Plasma samples were analyzed by LC-MS/MS after they had been treated with precipitated protein.
LC-MS/MS, the chromatographic column is a Waters X-Bridge C18 column (21mm by 50mm,3.5 μm); the mobile phase A is water +2mM ammonium acetate, the phase B is methanol +2mM ammonium acetate, the flow rate is 0.4mL/min, and the column temperature is 40 ℃. The ion source is used as an ESI source positive ion mode, 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 tables 6-1,6-2.
Table 6-1: pharmacokinetic parameters of IV dosing in rats
According to the results of the IV administration regimen, the compounds of the present application showed superior pharmacokinetic parameters in rats compared to olaparib.
Table 6-2: pharmacokinetic parameters of PO administration in rats
According to the results of the PO dosing regimen, the compounds of the present application showed superior pharmacokinetic parameters in rats compared to olaparib.
In addition, it was unexpectedly found in the present pharmacokinetic experiment that the half-life of olaparib was 1.31 hours, the half-life of compound 1 of the present application was 2.56 hours, the half-life of compound 5 was 2.52 hours, and the half-life of compound 46 was 2.8 hours, thus indicating that the duration of the pharmacodynamic action of compound 2, compound 5, and compound 46 of the present application was longer. Other compounds of the present application have substantially similar pharmacodynamic properties.
Experimental example 4 safety 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 test, the determination criterion is that if the test compound IC50 is greater than 30 μ M, the test compound is determined 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 compounds of the present application were shown to have 50% inhibitory concentration (IC50) values for hERG that were greater than 30 μ M. Thus, there was no inhibition of hERG, suggesting that the compounds of the present application do not lead to the safety risk of prolongation of cardiac QT interval.
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 dose settings were compound 1: 300 mg/kg; compound 2,3, 6: 200. 300mg/kg, single gavage, 10% DMSO in vehicle plus 50% PEG, observed 7 days after dosing.
Tolerance at high doses in a single administration the compounds of the present application are well tolerated.
Example 5 in vivo tumor inhibition experiment
This example is used to evaluate the effectiveness of different routes of administration of the compounds of the present application in inhibiting tumor proliferation.
This example examines the efficacy of each sample on mice bearing tumors of pancreatic cancer, Capan-1, after administration of the compound samples of the present application via the PO route to mice bearing subcutaneous transplants of human pancreatic cancer cell line, Capan-1.
Selection of tumor volume 100-3The tumor-bearing mice were randomly divided into 8 groups of 7 mice per group, 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- (VT end-VT start)/(VC end-VC start) ]. 100%
Wherein VT is last: mean tumor volume at the end of treatment group experiment
VT begins: mean tumor volume at the beginning of treatment group dosing
VC end: mean tumor volume at the end of vehicle control experiment
VC is started: mean tumor volume at the beginning of vehicle control group administration
Under the test condition, the compound has excellent drug effect, the tumor part regresses under the administration dosage of 10ml/kg and the like, and the drug effect is superior to that of a positive control group.
Formulation example 1Tablet formulation
The composition of the immediate release tablets is shown in table 7:
TABLE 7 composition of immediate release tablets
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 to occupy approximately 75% of the vial volume and then mixed together in a tumbler mixer for 30 minutes. 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 by hand for approximately 20 seconds. The resulting mixture was then dispensed into 400 mg aliquots and compressed into tablet cores using a hand press equipped with a 10mm die with a target compression of 0.5 tons.
Preparation example 2Capsule preparation
The composition of the capsule formulation is shown in table 8:
TABLE 8 composition of capsule formulations
The preparation method comprises the following steps:
(1) sieving the compound 5, lactose, microcrystalline cellulose, aerosil and magnesium stearate in parts by mass with a 80-mesh sieve, and drying at 85-90 ℃ for about 4 hours;
(2) uniformly mixing lactose, microcrystalline cellulose and aerosil, then mixing the compound 5 with the mixture, carrying out dry granulation, sieving the obtained granules with a 100-mesh sieve, and collecting 20-60-mesh dry granules;
(3) and (3) crushing the materials with the particle size of more than 20 meshes, performing secondary dry granulation on the crushed materials and the materials with the particle size of less than 60 meshes, adding the magnesium stearate in parts by mass into the granules, uniformly mixing to obtain mixed granules, and putting the mixed granules on a capsule filling machine to prepare capsules.
Although specific embodiments of the application have been described in detail, it will be appreciated by those skilled in the art that various modifications and alternatives to those details could be developed in light of the overall teachings of the disclosure, and that such modifications are intended to be within the scope of the application. The full scope of the application is given by the appended claims and any equivalents thereof.
Claims (21)
1. A compound of formula I-A or a pharmaceutically acceptable salt thereof,
wherein,
a and B together with the linking atoms form a benzene ring;
the ring X is a five-membered nitrogen-containing heteroaromatic ring;
m and n are each independently selected from 0, 1,2 and 3, and m + n is 3 or 4;
R1、R2and R3Is a hydrogen atom;
R4is halogen;
R5selected from hydrogen, oxy, halogen, C1-6Alkyl, 3-8 membered cycloalkyl, 5-6 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-6Alkyl, 3-8 membered cycloalkyl, 5-6 membered heteroaryl are each independently substituted by 1,2,3 or 4 substituents selected from halogen, hydroxy, amino, cyano, C1-4Alkyl radical, C1-4Alkoxy and 3-6 membered cycloalkyl, wherein a is 0, 1 or 2; or,
R5and form a polycyclic ring system on the ring X connected thereto, said polycyclic ring system being
R6Absent or selected from hydrogen and trifluoromethyl;
r and R' are each independently selected from hydrogen, C1-4Alkyl and 3-6 membered cycloalkyl, optionally, wherein said C1-4Alkyl and 3-6 membered cycloalkyl each independently substituted by 1,2,3 or 4 substituents selected from halogen, hydroxy, amino, cyano, C1-2Alkyl radical, C1-2Alkoxy and C1-2A substituent of alkylsulfonyl.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein ring X in the compound of formula I-a is an imidazole ring, a pyrazole ring, or a triazole ring.
4. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof,
wherein R is5Selected from hydrogen, oxy, halogen, C1-4Alkyl, 3-6 membered cycloalkyl, 5-6 membered heteroaryl, RO-, RC (O) -, ROC (O) -, RR 'N-, RR' NC (O) -, RS (O)a-and RR' NSO2-; optionally, wherein said C1-4Alkyl, 3-6 membered cycloalkyl, 5-6 membered heteroaryl are each independently substituted by 1,2,3 or 4 substituents selected from halogen, hydroxy, amino, cyano, C1-2Alkyl radical, C1-2Alkoxy and 3-6 membered cycloalkyl, wherein a is 0, 1 or 2;
wherein R and R' are each independently selected from hydrogen, C1-2Alkyl and 3-6 membered cycloalkyl; optionally, wherein said C1-2Alkyl and 3-6 membered cycloalkyl each independently substituted by 1,2,3 or 4 substituents selected from halogen, hydroxy, amino, cyano, C1-2Alkyl radical, C1-2Alkoxy and C1-2A substituent of alkylsulfonyl.
5. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof,
wherein R is5Selected from hydrogen, C1-4Alkyl, RC (O) -and RR' NC (O) -; optionally, said C1-4Alkyl is substituted by 1,2 or 3 substituents selected from fluoro, hydroxy, amino, cyano, C1-2Alkyl radical, C1-2Alkoxy and 3-6 membered cycloalkyl;
r and R' are each independently selected from hydrogen, C1-2Alkyl and 3-6 membered cycloalkyl; optionally, wherein said C1-2Alkyl is substituted by 1,2 or 3 substituents selected from halogen, hydroxy, amino, cyano, C1-2Alkyl radical, C1-2Alkoxy and C1-2A substituent of alkylsulfonyl.
6. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof,
wherein R is5Selected from hydrogen, bromine, hydroxymethyl, 1-hydroxyethyl, 2-hydroxy-2-propyl, difluoromethyl, trifluoromethyl, 1-difluoroethyl, pentakisFluoroethyl, cyclopropylmethyl, cyclopropyl, 1-methyl-4-pyrazolyl, 1-methyl-3-pyrazolyl, 2-methyl- [ 1.3.4%]Oxo-oxadiazol-5-yl, 3-methyl- [1.2.4 ]]Oxo-oxadiazol-5-yl, methoxy, cyclopropyloxy, aminoacyl, carbamyl, N-dimethylaminoyl, ethamoyl, 2-hydroxyethylaminoacyl, 2-methoxyethylalkyl, 2-hydroxyisobutyryl, 2-methanesulfonylethylamoyl, methylthio and methanesulfonyl.
7. A compound of claims 1-3, or a pharmaceutically acceptable salt thereof, wherein R4Is fluorine.
8. A compound, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from,
4- [ 4-fluoro-3- (2,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 1);
4- [ 4-fluoro-3- (2,4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 2);
4- [3- (2-Cyclopropylformyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 6);
4- [3- (2-cyclopropylmethyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 7);
4- [ 4-fluoro-3- (2-methyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 8);
4- [3- (2-Cyclopropylformyl-2, 4,6, 7-tetrahydro-pyrazolo [4,3-c ] pyridine-5-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (Compound 9);
cis-4- [ 4-fluoro-3- (4,5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 10);
trans-4- [ 4-fluoro-3- (4,5,5a,6,7,8,9,9 a-octahydro- [1,2,4] triazolo [4,3-a ] [1,6] naphthyridine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 11);
4- [3- (1-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 12);
4- [ 4-fluoro-3- (3-trifluoromethyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 16);
4- [3- (2-cyclopropyl-2, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorophenyl ] -2H-phthalazin-1-one (compound 19);
n-methyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) benzoyl ] -4,5,6, 7-tetrahydro-2H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 20);
6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 21);
n, N-dimethyl-6- [ 2-fluoro-5- (4-oxo-3, 4-dihydro-phthalazin-1-methyl) -benzoyl ] -4,5,6, 7-tetrahydro-1H-pyrazolo [3,4-c ] pyridine-3-carboxamide (compound 25);
4- [ 4-fluoro-3- (6-oxo-1, 3,4, 6-tetrahydro-pyrido [1,2-a ] pyrazine-2-carbonyl) benzyl ] -2H-phthalazin-1-one (compound 26);
4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H-imidazo [1,2-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 38);
4- { 4-fluoro-3- [3- (1-methyl-1H-pyrazol-4-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 39);
4- [ 4-fluoro-3- (3-methylsulfonyl-5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl) -benzyl ] -2H-phthalazin-1-one (compound 41);
4- { 4-fluoro-3- [3- (2-hydroxypropyl-2-yl) -1,4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 43);
4- { 4-fluoro- [ (3- (1-methyl-1H-pyrazol-3-yl) -5, 6-dihydro-8H- [1,2,4] triazolo [4,3-a ] pyrazine-7-carbonyl ] -benzyl } -2H-phthalazin-1-one (compound 45) and
4- [3- (3-cyclopropyl-1, 4,5, 7-tetrahydro-pyrazolo [3,4-c ] pyridine-6-carbonyl) -4-fluorobenzyl ] -2H-phthalazin-1-one (compound 47).
9. A process for the preparation of a compound of claim 1, or a pharmaceutically acceptable salt thereof, selected from the following processes:
(1) when R in formula I5To RR' NC (O) -the compounds can be synthesized by the following route:
carrying out condensation reaction on the compound A and the compound B' to obtain a compound C; carrying out urethane exchange on the compound C and a compound RR' NH to obtain a compound D;
(2) when R in formula I5To rc (o) -the compound can be synthesized by the following route:
carrying out condensation reaction on the compound A and the compound B 'to obtain a compound C'; performing acylation reaction on the compound C 'and a compound RCO-Lg to obtain a compound D'; or,
(3) when R in formula I5And R6When the other group described in claim 1, the compound can be synthesized by the following route:
carrying out condensation reaction on the compound A and the compound B to obtain a compound shown in a formula I;
wherein Lg in RCO-Lg represents a leaving group selected from halogen, -OTs and-OCOR, D and E in each structural formula are both C, and the remaining atoms and substituents are as defined in claim 1.
10. A pharmaceutical composition comprising a compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof; optionally, it further comprises a pharmaceutically acceptable carrier or excipient.
11. The pharmaceutical composition of claim 10, wherein the pharmaceutical composition contains 0.01-2000mg of the compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof.
12. The pharmaceutical composition of claim 10, wherein the pharmaceutical composition contains 0.1-1000mg of the compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof.
13. The pharmaceutical composition of claim 10, wherein the pharmaceutical composition contains 1-800mg of the compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof.
14. The pharmaceutical composition of claim 10, wherein the pharmaceutical composition contains 10-600mg of the compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof.
15. The pharmaceutical composition of claim 10, wherein the pharmaceutical composition contains 50-500mg of the compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof.
16. The pharmaceutical composition of any one of claims 10-15, further comprising one or more antineoplastic agents selected from the group consisting of temozolomide, doxorubicin, paclitaxel, cisplatin, carboplatin, dacarbazine, topotecan, irinotecan, gemcitabine, bevacizumab, anti-CTLA-4 mab Ipilimumab, anti-PD-1 mab pembrolizumab and Nivolumab, and anti-PD-L1 mab atezolizumab.
17. Use of a compound of any one of claims 1-8 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition of any one of claims 10-16 for the preparation of a PARP inhibitor.
18. The use of claim 17 wherein said PARP inhibitor is a PARP-1 inhibitor.
19. Use of a compound according to any one of claims 1 to 8 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to any one of claims 10 to 16 for the manufacture of an adjuvant for the treatment of tumours or for the enhancement of the effect of radiotherapy or chemotherapy of tumours.
20. Use of a compound according to any one of claims 1 to 8 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to any one of claims 10 to 16 for the manufacture of a medicament for the treatment of a tumour selected from the group consisting of breast cancer, ovarian cancer, colorectal cancer, melanoma, lung cancer, gastrointestinal stromal tumour, brain cancer, cervical cancer, pancreatic cancer, prostate cancer, gastric cancer, chronic myeloid leukaemia, liver cancer, lymphoma, peritoneal cancer, soft tissue sarcoma, neuroendocrine tumours and glioblastoma.
21. Use of a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 10 to 16, for the manufacture of a medicament for the treatment of a vascular disease, a neurodegenerative disease or nervous system inflammation.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201510939891 | 2015-12-16 | ||
CN2015109398911 | 2015-12-16 | ||
PCT/CN2016/110007 WO2017101796A1 (en) | 2015-12-16 | 2016-12-15 | Phthalazinone derivative, and preparation method and use thereof |
Publications (2)
Publication Number | Publication Date |
---|---|
CN107207504A CN107207504A (en) | 2017-09-26 |
CN107207504B true CN107207504B (en) | 2020-03-10 |
Family
ID=59055859
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN201680007644.0A Active CN107207504B (en) | 2015-12-16 | 2016-12-15 | Phthalazinone derivatives, preparation method and use thereof |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN107207504B (en) |
WO (1) | WO2017101796A1 (en) |
Families Citing this family (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN109251204A (en) * | 2017-07-13 | 2019-01-22 | 中国药科大学 | PARP inhibitor, its preparation method and medical usage containing phthalazines -1 (2H) -one structure |
CN109748923A (en) * | 2019-01-31 | 2019-05-14 | 中国药科大学 | Benzo[4,5]imidazo[1,2-a]pyrazinone derivatives and preparation method and use thereof |
CN113087724B (en) * | 2020-01-08 | 2024-01-19 | 四川科伦博泰生物医药股份有限公司 | Isothiazolopyrimidinone compounds, pharmaceutical compositions containing the same and uses thereof |
CN113754635B (en) * | 2020-06-02 | 2024-06-21 | 成都康弘药业集团股份有限公司 | Condensed ring compound and preparation method and application thereof |
CN112724148B (en) * | 2020-12-29 | 2022-04-12 | 上海药明康德新药开发有限公司 | Imidazopyridazine compound, modified amphiphilic functional molecule and application thereof |
WO2023288002A1 (en) * | 2021-07-16 | 2023-01-19 | Oregon Health & Science University | Phthalazinone-based parp-1 inhibitors |
CN118324766A (en) * | 2023-01-12 | 2024-07-12 | 中国科学院上海药物研究所 | Phthalic acid ketone or quinazolinone derivatives, pharmaceutical compositions and uses thereof |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102372716A (en) * | 2010-08-09 | 2012-03-14 | 江苏恒瑞医药股份有限公司 | Phthalazone derivative, its preparation method and application in medicine thereof |
CN102372706A (en) * | 2010-08-09 | 2012-03-14 | 江苏恒瑞医药股份有限公司 | Phthalazinone derivative, its preparation method and application in medicament |
CN102485721B (en) * | 2010-12-03 | 2015-12-09 | 曹亚 | 2,3-benzodiazine ketone compound replaced and uses thereof |
CN102898377B (en) * | 2012-02-14 | 2016-01-20 | 南京圣和药业股份有限公司 | One class phthalazinone derivatives and uses thereof |
CN103570725B (en) * | 2012-08-01 | 2017-03-22 | 中国科学院上海药物研究所 | Piperazidinoltriazole compound as well as preparation method and application thereof |
MA38080A1 (en) * | 2012-12-31 | 2018-02-28 | Cadila Healthcare Ltd | Substituted Phthalazin-1 (2h) -one Derivatives as Selective Inhibitors of Poly (adp-Ribose) Polymerase-1 |
-
2016
- 2016-12-15 CN CN201680007644.0A patent/CN107207504B/en active Active
- 2016-12-15 WO PCT/CN2016/110007 patent/WO2017101796A1/en active Application Filing
Also Published As
Publication number | Publication date |
---|---|
WO2017101796A1 (en) | 2017-06-22 |
CN107207504A (en) | 2017-09-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN107207504B (en) | Phthalazinone derivatives, preparation method and use thereof | |
JP7487421B2 (en) | PRMT5 inhibitors | |
CN111566104B (en) | Spiro aromatic ring compound and application thereof | |
JP7311228B2 (en) | RHO-related protein kinase inhibitors, pharmaceutical compositions containing same and methods for preparation and use thereof | |
CN117043167A (en) | Pyrimidine ring compound and preparation method and application thereof | |
JP2021521194A (en) | KRas G12C inhibitor and method of using it | |
CN111936475A (en) | Immunomodulator, composition and preparation method thereof | |
JP2020519589A (en) | Heteroaryl compounds that inhibit G12C mutant RAS protein | |
CN111499634B (en) | Quinazoline compound and application thereof in medicine | |
JP6564406B2 (en) | Imidazo-pyridazine derivatives as casein kinase 1 delta / epsilon inhibitors | |
AU2019280356B2 (en) | ERK inhibitor and use thereof | |
EP3868750A1 (en) | Modulators of ror-gamma | |
JP2023538060A (en) | BICYCLIC COMPOUNDS, COMPOSITIONS CONTAINING SAME, AND THEIR USE | |
CN106029076A (en) | Benzopiperazine compositions as BET bromodomain inhibitors | |
WO2021115457A9 (en) | Pyrazolo[1,5-a]pyridine compound, preparation method therefor and use thereof | |
WO2020021024A1 (en) | Substituted bicyclic compounds as modulators of the aryl hydrocarbon receptor (ahr) | |
IL311024A (en) | Substituted tricyclic compounds as parp inhibitors and use thereof | |
WO2016106266A1 (en) | TGFβ RECEPTOR ANTAGONISTS | |
JP2022551299A (en) | COMPOUND HAVING BRD4 INHIBITORY ACTIVITY, PREPARATION AND USE THEREOF | |
WO2019031990A1 (en) | Novel heterocyclic compounds as cdk8/19 inhibitors | |
CN116143806A (en) | Nitrogen-containing heterocyclic compound, preparation method and application | |
JP2022517085A (en) | Halogenated allylamine compounds and their applications | |
WO2023098880A1 (en) | Fused ring derivative, preparation method therefor, and application thereof | |
WO2016192630A1 (en) | Compound having kinase inhibiting activity, method of preparing same, and use of same | |
CN111032643A (en) | Quinolinone compounds |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant | ||
GR01 | Patent grant |