WO2013078771A1 - 一种多聚(adp-核糖)聚合酶抑制剂 - Google Patents

一种多聚(adp-核糖)聚合酶抑制剂 Download PDF

Info

Publication number
WO2013078771A1
WO2013078771A1 PCT/CN2012/001611 CN2012001611W WO2013078771A1 WO 2013078771 A1 WO2013078771 A1 WO 2013078771A1 CN 2012001611 W CN2012001611 W CN 2012001611W WO 2013078771 A1 WO2013078771 A1 WO 2013078771A1
Authority
WO
WIPO (PCT)
Prior art keywords
carbonyl
piperazine
fluoro
phenoxy
pyridazine
Prior art date
Application number
PCT/CN2012/001611
Other languages
English (en)
French (fr)
Inventor
姬建新
郭娜
薛婷
康兵强
叶鑫发
陈欣
张涛
Original Assignee
成都地奥制药集团有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 成都地奥制药集团有限公司 filed Critical 成都地奥制药集团有限公司
Priority to EP12852659.7A priority Critical patent/EP2799435B1/en
Priority to JP2014543747A priority patent/JP5820081B2/ja
Priority to US14/361,698 priority patent/US9187430B2/en
Priority to CA2857405A priority patent/CA2857405C/en
Publication of WO2013078771A1 publication Critical patent/WO2013078771A1/zh
Priority to US14/826,181 priority patent/US9718787B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • C07D237/30Phthalazines
    • C07D237/32Phthalazines with oxygen atoms directly attached to carbon atoms of the nitrogen-containing ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates to a poly(ADP-ribose) polymerase inhibitor having a compound related to phthalic acid hydrazide (pyridazinone), a composition thereof, and a medicament for preparing a disease associated with PARP enzyme activity Application.
  • a poly(ADP-ribose) polymerase inhibitor having a compound related to phthalic acid hydrazide (pyridazinone), a composition thereof, and a medicament for preparing a disease associated with PARP enzyme activity Application.
  • PARP Poly(ADP-ribose) polymerase
  • the PARP family has at least: PARP-1, PARP-2, PARP-3, PARP-4/VPARP, and terminal anchorase (Tankyrases, TANK-1, TANK-2 and TANK-3).
  • PARP superfamily members There are 16 different genes in the human genome that encode PARP superfamily members. These members have highly conserved PARP catalytically active regions composed of 50 amino acid residues. In addition, they have different primary structures and intracellular localization. And specific substrates. This indicates that poly ADP ribose is a type of post-transcriptional regulatory protein with different biological functions.
  • PARP-1 is a polypeptide chain consisting of 1014 amino acid residues with a molecular weight of 113 kDa and an isoelectric point between 8.0 and 9.8. It is the first member of the PARP family to be discovered and whose characteristics are best understood. There are three functional domains in the PARP-1 peptide chain. 1 DNA binding domain (DBD, 46kDa), located between the N-terminal 1 - 372 amino acid residues, comprising a nuclear localization sequence (NLS) and two zinc finger structures (Zn finger ).
  • DBD DNA binding domain
  • NLS nuclear localization sequence
  • Zn finger zinc finger structures
  • the 859-908 amino acid residue segment is 100% conserved in the ridge pusher.
  • the catalytic domain consists of two parts, wherein the amino acid residues near the N-terminal part are 662-784 to form six ot-helices of A, B, C, D, E, F.
  • This structure is unique to PARP, the helical region Through the combination of F oi -helix and active region, ie NAD binding site, it may be related to the activation signal Hand in relation.
  • studies have found that PARP also has a leucine zipper structure, presumably may play a role in the formation of dimers between PARP and other proteins or PARP itself.
  • PARP-1 is involved in the recognition of DNA gaps, and PARP-1 activated by DNA gaps forms homodimers and catalyzes the breakdown of NAD+ into nicotinamide and ADP ribose. The latter is used as a raw material to nucleate the nuclear receptor protein poly ADP, PARP.
  • the activation of -1 is positively correlated with the degree of DNA damage, and in the DNA damage-induced poly ADP glycosylation reaction, PARP-1 can negatively feedback the PARP-1 activity through self-repair to form DNA damage-inducing PARP. -1 active cycle. Inhibition studies of PARP-1 or studies in genetically deficient mice confirm that PARP-1 plays a key role in maintaining genomic stability.
  • PARP is quite abundant, especially in immune cells and germ cells. Poly ADP ribosylation occurs in many physiological processes. The multiple roles of PRAP include chromatin degradation, DNA replication, DNA repair, gene expression, cell division and differentiation, and cell apoptosis.
  • the PARP-1 enzyme When DNA is damaged, the PARP-1 enzyme is activated, binds to DNA, and catalyzes polyadenylation of ribosylation, thereby initiating DNA damage control and repair. At the same time, its overreaction will lead to the depletion of NAD+/ATP energy, which will eventually lead to cell necrosis.
  • This seemingly contradictory dual role of PARP-1 has attracted widespread interest from biologists and medicinal chemists. Therefore, small molecule compounds that inhibit PARP-1 activity have been widely used as drug research for anti-tumor therapy, inhibition of nerve damage and inflammatory damage. The discovery and search for inhibitors of the PARP-1 enzyme are of great significance.
  • a variety of PARP-1 inhibitors have been used in clinical drug research, such as BSI-201, AZD-2281, ABT-888, NU1025, GP 115427, AGO 14699, CEP-6800, AG14361 and INO1001.
  • AZD-2281 olaparib / KU-59436
  • AstraZeneca is a small molecule oral PARP-1 inhibitor, mainly used for the treatment of ovarian cancer, breast cancer and solid tumors.
  • AstraZeneca reported that AZD-2281 was the first to be used as a single agent in phase I clinically to inhibit gastric cancer and was well tolerated.
  • the drug is combined with cisplatin, carboplatin, paclitaxel and other drugs to treat solid tumors in the phase II clinical stage.
  • Clinical studies focusing on hereditary breast and ovarian cancer have entered Phase III clinical research.
  • a new PARP-1 inhibitor AG014699 developed by Pfizer is expected to be combined with the alkylating agent TMZ to treat malignant melanoma, which is in Phase II clinical research.
  • PARP-1 inhibitors Whether at the molecular level, at the cellular level, or at the overall level, these PARP-1 inhibitors have made some significant advances in therapeutic research in combination with chemotherapy, radiation therapy, or chemotherapy alone. Many studies have found that PARP-1 inhibitors can Enhance the efficacy of a variety of anti-tumor drugs (such as alkylating agents, topoisomerase inhibitors). Tentori et al (Blood 2002, vol. 99: 2241) found that NU1025 prolongs the survival of brain lymphoma-bearing mice. Delaney et al (Clin Cancer Res 2000, vol.
  • TMZ temozolomide
  • TP topotecan
  • GPI15427 developed by Guilford enhances the action of TMZ and significantly prolongs the survival of glioblastoma multiforme, brain lymphoma or intracranial malignant melanoma-bearing mice.
  • PARP-1 inhibitors have a neuroprotective effect after focal cerebral ischemia or stroke.
  • PARP-1 inhibitors are resistant to NMDA- or NO-induced neurotoxicity in rat cerebral cortex culture (Zhang et al, Science 1994, vol. 263: 687; Eliasson et al, Nature Med 1997, vol. 3: 1089), observation A series of compounds with PARP-1 inhibitory activity have neuroprotective effects.
  • the weaker PARP-1 inhibitor 3-aminobenzamide (3-AB) was administered intracerebroventricularly, and the infarct volume was reduced by 47% in mice (Endres et al., J Cereb Blood Flow Metab 1997, vol. 17: 1143).
  • Treatment with another PARP-1 inhibitor, 3-AB after 24 hours of ischemia slowed the reduction of NAD+ levels in ischemic tissue and reduced the synthesis of poly ADP-ribose multimers.
  • 3-AB (10 mg/kg) significantly reduced infarct volume in a suture occlusion model of focal ischemia in rats (Lo et al, Stroke 1998, vol. 29: 830).
  • PARP knockout mice can be used to help confirm the role of PARP in neurodegeneration.
  • ischemic mouse suture model an 80% reduction in infarct volume was observed in PARP-bialle knockout mice, whereas in PARP single allele knockout mice (PARP +/ - small The mouse) was reduced by 65%.
  • Endres et al (1997) reduced the infarct volume by 35% in PARP and rats, and by 31% in PARP +/ - animals.
  • experiments in PARP mice showed improvement in neurological scores. It also shows an increase in NAD+ levels after ischemia.
  • PARP_ mice compared with PARP + _ mice showed that PARP inhibitors significantly increased the motor's motor and recognition functions (Whalen et al, J Cereb Blood Flow Metab 1999, Vol. 19: 835). Production of peroxynitrite and activation of PARP have been confirmed in rats with spinal cord injury (Scott et al, Ann Neurol 1999, vol. 45: 120). These results suggest that inhibitors of PARP can provide protection against loss of function after trauma to the head or spine.
  • PARP activation is also implicated in cellular damage that occurs in various inflammatory diseases. Activation of macrophages by pro-inflammatory stimuli can result in the production of nitric oxide and superoxide anions, which in combination produce peroxynitrite, resulting in DNA single strand breaks and activation of PARP. Experiments using PARP inhibitors in numerous animal models support the hypothesis that PARP is involved in inflammatory diseases. The PARP inhibitor 5- ⁇ -6-amino-1,2-benzopyrone reduced the incidence and severity of arthritis in these animals, reducing the severity of necrosis and synovial hyperplasia.
  • PARP inhibitors can also be used to treat diabetes.
  • blood glucose levels are at a high level for a long time, which destroys the stability of endothelial cells.
  • hyperglycemia triggers the release of oxidative mediators from the mitochondrial electron transport chain (such as NADH/NADPH oxidase) and increases iNOS expression levels, resulting in excessive release of iNOS from the vascular endothelium.
  • the peroxides and superoxides produced by these factors cause DNA strand damage, activate PARP, and deplete the cell's NAD+, which leads to a series of pathological processes leading to dysfunction of the whole cell, which leads to cell death.
  • a compound described as a PARP inhibitor is disclosed in WO 99/11645, WO 00/32579, WO 02/36599 > WO 02/36599, WO 03/103666, WO 03/063874, WO 2004/096779, WO 2005/023246 WO 2005/054210, WO 2006/003148, WO 2007/138355, WO 2008/017883 US 2004/0248931, US 2006/0063926 and US 2007/0093489, and the like. Summary of the invention
  • An object of the present invention is to provide a phthalic acid (pyridazinone)-based compound, a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof as novel poly(ADP-ribose) Polymerase inhibitor.
  • Another object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof.
  • the diseases include cancer, neurodegenerative diseases, cardiovascular diseases, diabetes, and inflammation.
  • the invention further provides a method of treating a disease associated with PARP enzyme activity using a compound of the invention, a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof, or a combination thereof.
  • the invention provides a compound of formula (I), and pharmaceutically acceptable salts, hydrates, solvates and stereoisomers thereof:
  • R is selected from the group consisting of hydrogen, chlorine, fluorine, bromine, iodine, nitro, hydroxy, amino, branched or linear dC 6 alkyl, haloalkyl, CF 3 , CN, NRUR 12 , NH-CO-R 13 , OQ- An alkyl group, wherein R 11 and R 12 are each independently hydrogen or alkyl, and R 13 is hydrogen, dC 4 alkyl, dC 4 alkyl-phenyl or benzene;
  • Xi, X 2 are each independently selected from -CH or -N, but the two are not -N at the same time;
  • X 3 is selected from the group consisting of hydrogen, chlorine, fluorine, bromine, and iodine;
  • Y 1 is selected from CH and N
  • Y 2 is selected from CH and N
  • Y 3 is selected from the group consisting of CH, CF and N, wherein Y 1 , Y 2 and ⁇ 3 may be ⁇ at the same time, or only one or two may be ⁇ ;
  • is a 4 to 9 membered saturated or unsaturated monocyclic heterocyclic ring
  • R' is selected from -, -C(0)NR!-, -CC ⁇ Ri-, alkyl, cycloalkyl, cycloalkenyl, alkoxy, alkenyloxy, saturated or unsaturated monocyclic heterocyclic ring, aromatic Base, heteroaromatic group;
  • R1 is selected from the group consisting of alkyl, cycloalkyl, cycloalkenyl, alkoxy, decyloxy, saturated or unsaturated monocyclic heterocycle, aryl, heteroaryl, tert-butyl;
  • n is selected from 0 to 12;
  • the alkyl, aryl, cycloalkyl, heteroaryl, saturated or unsaturated monocyclic heterocycle, tert-butyl group present in the structure of the compound may be one or more halogen, hydroxy, cyano, amino, imino, Ether group, nitro group, nitroso group, mercapto group, thioether, sulfoxide, sulfonyl group, thioamido group, sulfonamide group, carboxylic acid, aryl group, heteroaryl group, C ⁇ o straight or branched alkyl group , Cwo straight or branched alkenyl, acyl, acyloxy, amide, acylamido, ureido, alkylamino, ester, phenoxy, benzyloxy, C 3 ⁇ cycloalkyl, C 5-2Q cycloalkenyl, C 1-2 . Alkoxy, C 1-20 olefin Aldooxy, C
  • hetero atom in the above-mentioned heteroaromatic group and the saturated or unsaturated monocyclic hetero ring includes N, 0, and S.
  • R is selected from the group consisting of hydrogen, chlorine, fluorine, bromine, iodine, nitro, hydroxy, amino, branched or linear dC 6 alkyl, 13 ⁇ 4 alkyl, CF 3 , CN, NRUR 12 , NH-CO -R 13 and Od-alkyl, wherein R 11 and R 12 are each independently hydrogen or C-C 4 alkyl, R 13 is hydrogen, dC 4 alkyl, C,-C 4 alkyl-phenyl or benzene ;
  • XX 2 are each independently selected from -CH or -N, but the two are not -N at the same time;
  • X 3 is selected from the group consisting of hydrogen, chlorine, fluorine, bromine and iodine;
  • H ring is:
  • ⁇ 1 is selected from CH and ⁇
  • ⁇ 2 is selected from CH and ⁇
  • ⁇ 3 is selected from CH, CF and N;
  • Ring A is a 4 to 9 membered and or unsaturated monocyclic heterocyclic ring
  • R' is selected from the group consisting of: -H, -C(0)Rj, -C(0)NR!, -CC R ⁇ alkyl, cycloalkyl, cycloalkenyl, alkoxy, alkenyloxy, saturated Or an unsaturated monocyclic heterocyclic ring, an aromatic group and a heteroaryl group;
  • R1 is selected from the group consisting of alkyl, cycloalkyl, cycloalkenyl, alkoxy, alkenyloxy, saturated or unsaturated monocyclic heterocyclic, aryl and heteroaryl;
  • n is an integer selected from 0 to 12;
  • any one of the alkyl group, the aryl group, the cycloalkyl group, the heteroaryl group, the saturated or unsaturated monocyclic heterocyclic ring and the t-butyl group is optionally substituted by one or more substituents selected from the group consisting of : halogen, hydroxy, aryl, aryl, imino, fluorenyl, nitro, nitroso, fluorenyl, thioether, sulfoxide, sulfonyl, thioamido, sulfonamide, carboxylic acid, aryl, Heteroaryl, straight or branched alkyl, Cwo straight or branched fluorenyl, acyl, acyloxy, amide, acylamido, ureido, alkylamino, ester, phenoxy, benzyloxy Base, C 3-2 .
  • a cycloalkyl group a C 5-20 cycloalkenyl group, a C 1-20 decyloxy group, a C 1-20 alkenyloxy group, a C 1-20 alkylthio group, and a C 1-20 haloalkyl group;
  • the hetero atom in the above-mentioned heteroaromatic group and the saturated or unsaturated monocyclic heterocyclic ring is selected from N, 0 and 5.
  • the substituent of the amino group such as the above-mentioned C NR, an alkylamino group or the like may be either a mono- or di-substituted group, and the two substituents may be the same or different.
  • the H-ring is:
  • the ring A is a substituted or unsubstituted 4-9 membered saturated or unsaturated monocyclic hetero ring containing 1 to 3 hetero atoms selected from N, 0 and S; and R' is selected from the following In the group: -H, -C(0)Ri , -C(0)NRi , -C0 2 R!
  • substituted or unsubstituted Cwo alkyl substituted or unsubstituted C 2 4o alkenyl, substituted or unsubstituted C 3-10 cycloalkyl, substituted or unsubstituted Cwo cycloalkenyl, substituted or unsubstituted CWQ alkoxy, substituted or unsubstituted C 2-1Q alkenyloxy, substituted or unsubstituted 1- 3 3-10 membered saturated or unsaturated monocyclic heterocyclic ring selected from heteroatoms of N, 0 and S, substituted or unsubstituted C 6-1 .
  • substitution is substituted by at least one group selected from the group consisting of halogen, hydroxy, cyano, amino, carboxy, C 2-10 hydrocarbyl ether, nitro, Cwo hydrocarbyloxy, C w .
  • An aromatic group a C 5-10 heteroaryl group having 1-3 hetero atoms selected from N, O and S, a C 1-1 () straight or branched alkyl group, a C 1-1 () straight chain or Branched alkenyl group, C 1-1Q hydrocarbyl acyl group, C 1-1Q hydrocarbyl acyloxy group, C 1-1Q hydrocarbyl amide group, C 1-K) hydrocarbyl acyl amide group, ureido group, (C 1-K ) alkyl group 1-2 amino, C 1-1Q hydrocarbyl ester, phenoxy, benzyloxy, C 3-1G cycloalkyl, C 5-1Q cycloalkenyl, C 1-K) alkoxy, C 1- H ) anthraceneoxy, C 1-10 alkylthio and Cwo haloalkyl.
  • the A ring may be selected from the group consisting of piperazine, homopiperazine, piperidine, tetrahydropyridine, 4-hydroxypiperidine, 2-mercaptopiperazine and 3-mercaptopiperazine.
  • R' is selected from the group consisting of: -H, -C(0)Rj, -C(0)NR1 ; -C0 2 Rj , substituted or unsubstituted Cw.
  • Alkyl substituted or unsubstituted Cw.
  • selected from the group consisting of substituted or unsubstituted C M Q alkyl, substituted or unsubstituted C 2 -K ) alkenyl, substituted or unsubstituted C 3-7 cycloalkyl, substituted or unsubstituted Cw a cycloalkenyl group, a substituted or unsubstituted Cwo alkoxy group, a substituted or unsubstituted 5-7 membered saturated or unsaturated monocyclic heterocyclic ring containing 1-3 hetero atoms selected from N, O and S, substituted or Unsubstituted C 6-1 .
  • substitution means to be At least one selected from the group of radicals substituted with: halo, hydroxy, cyano, amino, nitro, C 6 10 aryl,
  • the compound is represented by the following formula (II) or ( ⁇ ):
  • R ' is -C (0) R, wherein 1 ⁇ is selected from the group consisting of:!.
  • R is a group selected from the group consisting of hydrogen, fluorine, chlorine, bromine, nitro and amino; and X 2 is -CH, respectively.
  • R' is a group selected from the group consisting of: one or two selected from the group consisting of fluorine, chlorine, nitro, amino, decylamino, dimethylamino, decyl and phenyl.
  • the substituent is substituted with ethyl, propyl, butyl, pentyl, cyclopropyl, cyclobutane, cyclohexane, phenyl, pyrrolidinyl and piperidinyl.
  • propyl, butyl and pentyl groups may be linear or branched, and more preferably branched.
  • the propyl group is preferably an isopropyl group
  • the butyl group is preferably a tert-butyl group
  • the pentyl group is preferably a 1-ethylpropyl group or the like.
  • the compound of the formula (I) of the present invention may specifically be the following compound 1-130:
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of the above compound, a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof, and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier is a conventional pharmaceutical carrier in the art and will not be described herein.
  • the above listed compounds, and pharmaceutically acceptable salts, hydrates, solvates and stereoisomers thereof, and pharmaceutical compositions thereof, can be used as a medicament for the treatment of PARP-mediated disorders, including anti-cancer, neurodegenerative diseases , cardiovascular disease, diabetes and inflammation.
  • the invention therefore also provides the use of said compound, a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof, for the manufacture of a medicament for the treatment of a condition mediated by PARP.
  • the PARP mediated condition is selected from the group consisting of cancer, neurodegenerative diseases, cardiovascular diseases, diabetes, and inflammation.
  • the cancer is specifically histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, gastric cancer, colon cancer, rectal cancer, ovarian cancer, Cervical cancer, brain cancer, esophageal cancer, bone cancer, testicular cancer, melanoma, skin cancer, epithelial cell carcinoma, prostate cancer, nasopharyngeal carcinoma, oral cancer, leukemia, and brain, reproductive system, lymphatic system, digestive system tumor Any of a respiratory tumor and a skin tumor.
  • the onset or progression of a disease or condition associated with PARP activity is specifically histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, gastric cancer, colon cancer, rectal cancer, ovarian
  • symptoms include traumatic damage to the central nervous system, such as brain damage, and neuronal degeneration associated with traumatic damage to the central nervous system.
  • Related symptoms and diseases treatable by the methods of the invention include vascular stroke, cardiac ischemia, cerebral ischemia, cerebral vascular disorders such as multiple sclerosis, and neurodegenerative diseases such as Alzheimer's disease, Huntington's dance. Disease and Parkinson's disease.
  • the compounds of the present invention can treat or prevent tissue damage caused by cell damage or death caused by necrosis or apoptosis; and can alleviate nerve or cardiovascular tissue damage, including subsequent ischemia, myocardial infarction, and reperfusion injury. It can also be used to treat or prevent other cardiovascular diseases such as angina pectoris and cardiovascular ischemia.
  • PARP-related symptoms or diseases treatable by the compounds described herein include inflammation such as pleurisy and colitis, endotoxic shock, diabetes, arthritis, cardiac ischemia, retinal ischemia, skin aging, chronic and acute pain , hemorrhagic shock, etc.
  • inflammation such as pleurisy and colitis
  • endotoxic shock diabetes, arthritis, cardiac ischemia, retinal ischemia, skin aging, chronic and acute pain , hemorrhagic shock, etc.
  • one or more compounds of the invention are administered by the patient to prevent damage or injury to the brain from being minimized.
  • the present invention also provides a method of treating a PARP-mediated disorder, comprising administering to a patient an effective amount of a compound according to any one of claims 1 to 10, a pharmaceutically acceptable salt, hydrate, solvate or stereo thereof. Isomer, or the step of the pharmaceutical composition of claim 14.
  • compositions generally include an inert diluent carrier or an edible carrier.
  • Pharmaceutically compatible binders and/or adjuvant materials can be included as part of the composition.
  • Tablets, pills, capsules, tablets and the like may contain any of the following ingredients or compounds of similar nature: binders such as microcrystalline cellulose, tragacanth or gelatin; excipients such as starch or lactose; dispersing agents such as alginic acid, Primogel or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silica; a sweetener such as sucrose or saccharin; or a flavoring agent such as peppermint oil, methyl salicylate or orange flavor.
  • binders such as microcrystalline cellulose, tragacanth or gelatin
  • excipients such as starch or lactose
  • dispersing agents such as alginic acid, Primogel or corn starch
  • a lubricant such as magnesium stearate
  • a glidant such as colloidal silica
  • a sweetener such as sucrose or saccharin
  • a flavoring agent such as peppermin
  • the dosage unit form can contain various other materials which modify the physical form of the dosage unit, for example, a sugar coating, shellac or enteric solvent.
  • the syrup may contain sucrose as a sweetening agent as well as certain preservatives, dyes, colors and flavors.
  • Selective formulations for administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions.
  • the nonaqueous solvent are dimethyl sulfoxide, alcohol, propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include mixtures of alcohols and water, buffered media and saline.
  • Intravenous vehicles include fluids and nutrient supplements, electrolyte supplements such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as antibacterial agents, antioxidants, chelating agents, inert gases, and the like.
  • the compounds of the invention may also take the form of pharmaceutically acceptable salts, hydrates, solvates or metabolites.
  • Pharmaceutically acceptable salts include the basic salts of inorganic and organic acids including, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, malic acid, acetic acid, oxalic acid, tartaric acid, citric acid, lactic acid. , fumaric acid, succinic acid, mala Acid, salicylic acid, benzoic acid, phenylacetic acid, mandelic acid, and the like.
  • carboxyl groups When the compounds of the present invention include acidic functional groups such as carboxyl groups, suitable pharmaceutically acceptable carboxyl cations are well known to those skilled in the art, including alkali, alkaline earth, ammonium, quaternary ammonium cations and the like.
  • Fig. 1 is a graph showing the relationship between the peak area measured by HPLC and the concentration of a standard in the test example 3 using the compound AZD-2281 as a standard.
  • O-phthalic anhydride (la) reacts with hydrazine hydrate under the action of glacial acetic acid to form phthalic acid hydrazide (lb), and is reacted with sodium 3-chloromercaptobenzoate (lc) under the action of sodium hydride.
  • the reaction produces 3-((4-oxo-3,4-dihydro-indole.
  • O-phthalic acid hydrazide ( lb) forms 1, 4-dichloropyrrole (2a) under the action of phosphorus oxychloride, and then reacts with ethyl 3-hydroxybenzoate (2b) to obtain 3- (4) - Crude product of ethyl chloronaphthyridin-1-yloxy)benzoate (2c), crude in glacial acetic acid, sodium acetate to give 3-(4-oxo-3,4-dihydro- Ethyl pyridazine-1-yloxy)benzoate (2d), which is then hydrolyzed under basic conditions to give the intermediate 3-(4-oxo-3,4-dihydro-pyridazine-1- Benzyl)benzoic acid (2e), and finally with R, substituted nitrogen heterocycles such as piperazine, piperidine, 4-hydroxypiperidine, tetrahydropyridine, 1,4-diazepane, etc.
  • R substituted nitrogen
  • the ortho-dicarboxylic acid anhydride (3a) containing a substituent R is added to the hydrazine/glacial acetic acid to form an R-substituted phthalic acid hydrazide (3b), followed by the formation of a substituted dichloro group under the action of phosphorus pentachloride.
  • Pyridazine compound (3c) 2-fluoro-5-nonylphenylbenzonitrile (3d) is hydrolyzed to form 2-fluoro-5-hydroxybenzoic acid (3e), which is then esterified with decyl alcohol to give 2- Ethyl fluoro-5-hydroxybenzoate (3f).
  • 2-fluoro-5-nonylphenylbenzonitrile (3d) is hydrolyzed to form 2-fluoro-5-hydroxybenzoic acid (3e), which is then esterified with decyl alcohol to give 2- Ethyl fluoro-5-hydroxybenzoate (3f).
  • a substituted nitrogen heterocycle such as piper.
  • piperidine, 4-hydroxypiperidine, tetrahydropyridine, 1, 4-diazepane, etc. in 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDCI), triethylamine, 1-hydroxy-7-azabenzotriazole (HOAt) conditions condensed to the target compound.
  • EDCI 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride
  • HOAt 1-hydroxy-7-azabenzotriazole
  • R a substituted nitrogen heterocycle such as piper.
  • piperidine 4-hydroxypiperidine, tetrahydropyridine, 1, 4-diazepane, etc., in 1-ethyl-(3-didecylaminopropyl)carbodiimide hydrochloride (EDCI), triethylamine, 1-hydroxy-7-azabenzotriazole (HOAt) conditions condensed to the target compound.
  • EDCI 1-ethyl-(3-didecylaminopropyl)carbodiimide hydrochloride
  • HOAt 1-hydroxy-7-azabenzotriazole
  • the structure of the compound in the following examples was determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (ESI).
  • NMR nuclear magnetic resonance
  • ESI mass spectrometry
  • the NMR shift ( ⁇ 5) is given in units of ⁇ ppm ).
  • 1H-NMR optical transmission data was determined by Varian Mercury-600 MHz NMR.
  • the solvent was deuterated dimercaptosulfoxide ( ), deuterated chloroform (CDC1 3 ), and the internal standard was tetramethylsilane (TMS).
  • the ESI-MS assay was determined using a Finnigan LCQ Deca grammar.
  • HPLC High performance liquid chromatography
  • Thin layer chromatography silica gel plate uses Yantai Huanghai HSGF254 silica gel plate.
  • the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm.
  • the specification of thin layer chromatography separation and purification product is 0.4mm ⁇ 0.5mm. .
  • the column is generally used as a carrier for Yantai Yellow Sea 300 ⁇ 400 mesh silica gel.
  • Known starting materials of the invention may be synthesized by or according to methods known in the art, or may be purchased from Alfa Aesar, Lancaster, TCI, Suiyuan Chemical Technology and Ester .
  • the reactions were all carried out under an argon or nitrogen atmosphere.
  • An argon or nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 liter.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times. There is no particular description in the examples, and the reaction temperature is room temperature. Room temperature is the most suitable reaction temperature, 20 ° C ⁇ 30 ° C fatigue
  • TLC thin layer chromatography
  • the elution system for column chromatography and the developer system for thin layer chromatography using purified compounds include A: dichloromethane and decyl alcohol systems, B: n-hexane and ethyl acetate systems, C: petroleum ether and ethyl acetate systems
  • C petroleum ether and ethyl acetate systems
  • the volume ratio of the solvent is adjusted depending on the polarity of the compound, and may be adjusted by adding a small amount of an alkaline or acidic reagent such as triethylamine or acetic acid.
  • phthalic acid hydrazide ( lb) 10 g, 61.7 mmol was dissolved in a round bottom flask with dry N,N-didecylcarboxamide (100 mL), and sodium hydride (.63) was added at 0 °C. g, 67.9 mmol), stirred for 0.5 h at the end of the addition.
  • ethyl 3-chloromercaptobenzoate (lc, purchased from Suiyuan Chemical Technology) (13.49 g, 67.9 mmol) of N, N was slowly added dropwise.
  • p-Fluorobenzoic acid (6b) (7 g, 50 mmol) was dissolved in dichloromethane (100 mL), followed by 1-ethyl-(3-didecylaminopropyl)carbodiimide hydrochloride (EDCI) (10.5 g, 55 mmol), triethylamine (7.7 mL, 55 mmol), 1-hydroxy-7-azabenzotriazole (HOAt) (7.5 g, 55 mmol), stirring at room temperature for half an hour Further, 1-tert-butoxycarbonylpiperazine (6a) (10. 2 g, 55 mmol) was added, and the mixture was stirred at room temperature overnight.
  • EDCI 1-ethyl-(3-didecylaminopropyl)carbodiimide hydrochloride
  • HOAt 1-hydroxy-7-azabenzotriazole
  • N,N'-carbonyldiimidazole (CDI) (1.62 g, 10 mmol) was added to 1-tert-butoxycarbonylpiperazine (6a) (186 g, 10) under ice-cooling (0 ° C).
  • 6a 1-tert-butoxycarbonylpiperazine
  • the organic phase was washed three times with 50 mL of brine, and the organic phase was dried over anhydrous sodium sulfate.
  • the compound of the present invention 1 was prepared according to the above scheme 1:
  • the compound 2 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. -Phenylcarbonylpiperidine. A white solid was obtained.
  • the compound 3 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. -propionylpiperazine. A white solid was obtained.
  • the compound 4 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that the N-(cyclopropanecarbonyl)piperazine is substituted, and the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (cyclohexanecarbonyl)piperazine. A white solid was obtained.
  • the compound 5 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the example 1, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the compound (I) corresponding to R, the substituted ring A is prepared. - (cyclopentanecarbonyl) piperazine. A white solid was obtained.
  • the compound of the present invention 6 was prepared according to the scheme 1, and the synthesis method was referred to the example 1, except that N-(cyclopropane) was substituted.
  • Qin firstly prepared 4-acetylpiperazine corresponding to R, substituted A ring in the formula (I) in the compound 6. A white solid was obtained.
  • the compound of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example 7, except that the N-(cyclopropanecarbonyl)pyridazine is substituted, and the compound 4 in the formula (I) corresponding to R, the substituted ring A is first prepared. -propionylpiperazine. A white solid was obtained.
  • the compound 9 of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the corresponding formula (I) corresponding to R, substituted A ring is prepared. - benzoylpiperazine. A white solid was obtained.
  • the compound 10 of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the 4 ring of the A ring corresponding to the R' in the formula (I) is first prepared. - (cyclohexanecarbonyl)piperazine. A white solid was obtained.
  • the compound 11 of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (cyclopentanecarbonyl) piperazine. A white solid was obtained.
  • the compound 12 of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example of Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the corresponding formula (I) corresponding to R, substituted A ring 4 is prepared. -Acetylpiperazine. A white solid was obtained.
  • the compound of the present invention 13 is prepared according to the scheme 2, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (4-Benzyloxycarbonyl-piperidine) diffpiperazine. A white solid was obtained.
  • the compound of the present invention 14 was prepared according to the scheme 1, and the synthesis method was referred to the example 1, except that N-(cyclopropionyl)piperidin was replaced.
  • Qin firstly prepared 4-(n-nitrostylpiperazine) corresponding to R in the formula (I), substituted A ring in the compound 14. A pale yellow solid was obtained.
  • the compound of the present invention 15 was prepared according to the scheme 1, and the synthesis method was carried out in the same manner as in Example 1, except that instead of N-(cyclopropanecarbonyl)piperazine, the 4 ring corresponding to the R'-substituted A ring in the formula (I) was first prepared. - p-Nitrobenzoyl piperazine. A pale yellow solid was obtained.
  • the compound I 6 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the ring A corresponding to the R' substituted in the formula (I) is first prepared. 4-benzyl-1,4-diazepane. A white solid was obtained.
  • the compound of the present invention 17 is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that N-(cyclopropanecarbonyl)piperazine is substituted, and the corresponding compound R in the compound (I) and the substituted ring A are prepared first.
  • N-(cyclopropanecarbonyl)piperazine is substituted, and the corresponding compound R in the compound (I) and the substituted ring A are prepared first.
  • the compound 19 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that N-(cyclopropanecarbonyl)piperazine is substituted, and the compound 4 corresponding to R in the formula (I) and the substituted ring A are first prepared. - (pyrimidin-2-yl) piperazine. A white solid was obtained.
  • the compound 20 of the present invention was prepared according to the scheme 1, and the synthesis method was referred to the example 1, except that N-(cyclopropanecarbonyl)anthracene was replaced.
  • Qin first prepared 4-tert-butoxycarbonyl-1,4-diazepane corresponding to the R ring substituted by R' in the formula (I) in the compound 20. A white solid was obtained.
  • the compound 21 of the present invention was prepared according to the scheme 1, and the synthesis method was referred to the example 1, except that the N-(cyclopropanecarbonyl) group was replaced.
  • Qin 4-(3-chloro-phenyl). 4-hydroxypiperidine corresponding to R in the formula (I), substituted A ring, is first prepared. A white solid was obtained.
  • the compound 22 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared. - (4-Chloro-stupyl)- 4-hydroxypiperidine. A white solid was obtained.
  • the compound 23 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the first embodiment except that the N-(cyclopropanecarbonyl)piperone is substituted, and the compound 4 corresponding to the R in the formula (I) and the substituted ring A are prepared first.
  • -Phenylpiperazine A white solid was obtained.
  • the compound 24 of the present invention is prepared according to the scheme 1, and the synthesis method is the same as in the example 1, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the corresponding compound R in the formula (I) is substituted. - Phenyl-1,2,3,6-tetrahydropyridine. A white solid was obtained.
  • the compound 25 of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example 7 except that instead of the N-(cyclopropanecarbonyl)piperone, the 4 ring corresponding to the R'-substituted A ring in the compound (I) is first prepared. - (4-Fluoro-phenyl)-1,2,3,6-tetrahydropyridine. A white solid was obtained.
  • the compound 26 of the present invention is prepared according to the scheme 2, and the synthesis method is the same as in the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the corresponding compound R of the formula (I) and the substituted ring A are prepared. - Isonicotinic acid piperazine. A white solid was obtained.
  • a ring 27 Preparation of compounds of the invention according to Scheme 2, synthesis of Reference Example 7, except that instead of N- (carboxy-cyclopropanecarbonyl) piperazine, 27 corresponding to the general formula (I), R compound is first prepared, the 4-substituted - (4-Fluorobenzoyl) piperazine.
  • the compound of the present invention 28 is prepared according to the scheme 3, and the synthesis method is the same as in the example 1, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (4-Nitrobenzoyl) piperazine. A pale yellow solid was obtained.
  • the compound 29 of the present invention is prepared according to the scheme 3, and the synthesis method is the same as in the example 1, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (4-Benzyloxycarbonyl-piperidine)carbonylpiperazine. A white solid was obtained.
  • the compound 30 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that the N-(cyclopropanecarbonyl) group was replaced.
  • Qin 4-(4-benzyloxycarbonyl-piperidine)carbonylpiperazine corresponding to R in the formula (I), substituted A ring, is first prepared. A white solid was obtained.
  • the compound 31 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)pyrazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared.
  • - (4-Nitrobenzoyl) is given as a white solid as a yellow solid.
  • the compound 33 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the N corresponding to the ring of the formula (I) and the substituted ring A of the compound 33 are first prepared. -Phenylpiperazine-1-indoleamide. A white solid was obtained.
  • the compound 34 of the present invention was prepared according to Scheme 3, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin first prepared 4-(4-fluorobenzoyl)hypo in the compound 34 corresponding to the R'-substituted A ring of the formula (I). A white solid was obtained.
  • the compound 35 of the present invention is prepared according to the scheme 3, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the compound (I) corresponding to R, the substituted ring A is prepared. - Stupyl formyl piperazine. A white solid was obtained.
  • the compound of the present invention 37 is prepared according to Scheme 3, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the corresponding compound R in the formula (I) corresponding to R, substituted A ring is prepared. 4-(4-Fluorophenyl)piperazine. A pale yellow solid was obtained.
  • the compound of the present invention 38 is prepared according to the scheme 3, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the compound (I) corresponding to R, the substituted ring A is prepared. - Fluorobenzoyl. A white solid was obtained to give a white solid.
  • the compound 39 of the present invention was prepared according to Scheme 3, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin, 4-(4-carbamoylpiperazine) corresponding to R in the formula (I) and substituted A ring in the compound 39 was first prepared. A pale yellow solid was obtained.
  • the compound of the present invention 40 is prepared according to the scheme 3, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)pyrazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (1-Benzyloxycarbonyl-piperidine)carbonylpiperazine. A yellow solid was obtained.
  • the compound 41 of the present invention was prepared according to the scheme 3, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)methane, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - ( 4 -Nitrobenzoyl) piperazine. A yellow solid was obtained.
  • the compound 42 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin, N-cyclohexylpiperazine-1 -carboxamide corresponding to R in the formula (I) and substituted A ring in the compound 42 is first prepared. A white solid was obtained.
  • the compound 43 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7, except that N-(cyclopropanecarbonyl)piperazine is substituted, and the compound 4 in the compound (I) corresponding to R and the substituted ring A is prepared first. - Acetylpiperazine. A white solid was obtained.
  • the compound 45 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of the N-(cyclopropanecarbonyl)piperone, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (2-Benzyloxycarbonyl-piperidine)carbonylpiperazine. A white solid was obtained.
  • the compound 46 of the present invention is prepared according to Scheme 4, and the synthesis method is referred to Example 7, except that N-(cyclopropane is substituted). Carbonyl) piperazine.
  • Qin 4-(3-benzyloxycarbonyl-piperidine)carbonylpiperazine corresponding to the R ring substituted by R' in the formula (I) is first prepared. A white solid was obtained.
  • the compound 47 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7, except that the N-(cyclopropanecarbonyl)piperone is substituted, and the compound 4 corresponding to R in the formula (I) and the substituted ring A are first prepared. - (4-oxocyclohexanecarbonyl)piperazine. A white solid was obtained.
  • the compound 48 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin, 4-(4-cyanobenzoyl)piperazine corresponding to the R ring substituted by the R' in the formula (I) is first prepared. A white solid was obtained.
  • the compound 49 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound R in the corresponding formula (I) corresponding to the substituted ring A is prepared. R) 4 - (2-phenylpropionyl) piperazine. A white solid was obtained.
  • the compound 50 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the 4 ring corresponding to the R'-substituted A ring in the formula (I) is first prepared. - (cyclopropane Carbonyl) piperazine. A white solid was obtained.
  • the compound 51 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 1 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (Taste. Qin-1 -yl)propane-1,2-dione. A white solid was obtained.
  • the compound 52 of the present invention is prepared according to the scheme 4, and the synthesis method is as described in Example 7, except that instead of N-(cyclopropanecarbonyl)pyrazine, the 4 ring of the ring A corresponding to the R' substituted in the formula (I) is first prepared. - (5-Mercapthy Isoxazole-3-carbonyl)piperazine. A white solid was obtained.
  • the compound 53 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (2,2,2-trifluoroacetyl)piperazine. A white solid was obtained.
  • the compound of the present invention is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanedecyl)piperazine, the compound R in the corresponding formula (I) corresponding to R, substituted A ring is first prepared. 4-(3,3,3-trifluoropropionyl)piperazine. A white solid was obtained.
  • the compound of the present invention 56 is prepared according to the scheme 4, and the synthesis method is as described in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared. - (2-Hydroxyacetyl) piperazine. A white solid was obtained.
  • the compound of the present invention 57 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (2-cyanobenzoyl) piperazine. A white solid was obtained.
  • the compound of the present invention 60 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of the N-(cyclopropanecarbonyl)piperazine, the compound 4 in the compound 60 corresponding to R in the formula (I), substituted anthracene ring is prepared. - Isonicotinic acid piperazine. A white solid was obtained.
  • the compound 61 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (2-ethylbutyryl) piperazine. A white solid was obtained.
  • the compound 62 of the present invention is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the 4 ring corresponding to the R'-substituted A ring in the compound (I) is first prepared. - (4,4-difluoropiperidine-1-carbonyl)piperazine. A white solid was obtained.
  • the compound of the present invention 64 is prepared according to the scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared. - (3-Hydroxypropyl) piperazine. A white solid was obtained.
  • the compound of the present invention 65 is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of the N-(cyclopropanecarbonyl)-benzine, the 4 ring of the compound A corresponding to the R-substituted A ring in the formula (I) is first prepared. -carbonitrile piperidine. A white solid was obtained.
  • the compound 66 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the corresponding compound R of the formula (I), the substituted ring A is prepared. - Isobutyrylpiperazine. A white solid was obtained.
  • the compound of the present invention 67 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the N corresponding to the R in the formula (I) and the substituted ring A are prepared in the compound 67. , N-Dimethylpiperazine- 1 -nonanamide. A white solid was obtained.
  • the inventive compound 68 was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropane was replaced).
  • the compound 70 of the present invention is prepared according to the scheme 70, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanediyl)piperazine, the compound R in the compound (I) corresponding to the substituted ring A is prepared. 4-(2-hydroxypropionyl)piperazine. A white solid was obtained.
  • the compound 71 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (4,4-Difluoropiperidin-1 -carbonyl)piperidine. A white solid was obtained.
  • the compound 72 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin, 4-(piperidin- 1 ⁇ -carbonyl)piperidine corresponding to the R ring substituted by R' in the formula (I) is first prepared. A white solid was obtained.
  • the compound 73 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the N corresponding to the ring of the formula (I) and the substituted ring A of the compound 73 are first prepared. -Phenylpiperidine-4 -nonanamide. A white solid was obtained.
  • Compound 74 of the present invention is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the N in the compound 74 corresponding to R in the formula (I), substituted A ring is first prepared. -ethyl piperidine-4 -carboxamide. A white solid was obtained.
  • the compound of the present invention 75 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (4-(Dimercaptoamino)benzoyl) piperazine. A white solid was obtained.
  • the compound of the present invention is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the ring A corresponding to the R' substituted in the formula (I) is first prepared. 4-(3-(Dimercaptoamino)benzoyl) piperazine. A white solid was obtained.
  • the compound 77 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. -tert-butylpiperazine. A white solid was obtained.
  • the compound 78 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that the N-(cyclopropanecarbonyl) group was replaced.
  • Qin firstly prepared 4-(furan-2-carbonyl)piperazine corresponding to R in the formula (I) and substituted A ring in the compound 78. A white solid was obtained.
  • the compound 79 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)pyrazine, the 4 ring corresponding to the R-substituted A ring in the compound (I) was first prepared. - Isopropylpiperazine.
  • the compound of the present invention 80 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the 4 ring corresponding to the R'-substituted A ring in the compound (I) is first prepared. - Allyl piperazine. A white solid was obtained.
  • the compound 81 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - 3-cyanobenzoylpiperazine. A white solid was obtained.
  • the compound 82 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (1H-pyrazole-1-carbonyl) piperazine. A white solid was obtained.
  • the compound 83 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropionyl)piperidin was replaced.
  • Qin, 4-(morpholine-4-carbonyl)piperazine corresponding to the R ring substituted by R' in the formula (I) is first prepared. A white solid was obtained.
  • the compound 84 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (Pyrrolidin-1 -carbonyl) piperazine. A white solid was obtained.
  • the compound of the present invention 85 was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin firstly prepared 4-(piperidine-1-carbonyl)piperazine corresponding to R in the formula (I) and substituted A ring in the compound 85.
  • a white solid was obtained.
  • the compound 86 of the present invention is prepared according to the scheme 4, and the synthesis method is as described in Example 7, except that instead of N-(cyclopropanecarbonyl)piperole, the N corresponding to the ring of the formula (I) and the substituted ring A are prepared first.
  • - (3 - (Trifluoromethyl)phenyl) piperidine.
  • Qin-1 - amide A white solid was obtained.
  • the compound of the present invention 87 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (3-Nitrobenzoyl) piperazine. A white solid was obtained.
  • the compound 88 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin firstly prepared 4-(2-nitrobenzoyl) piperazine corresponding to R in the formula (I), substituted A ring, in the compound 88.
  • a white solid was obtained.
  • the compound 89 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - Butyr-2-isoylpiperazine. A white solid was obtained.
  • the compound 90 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - Mercaptopiperazine. A white solid was obtained.
  • the compound 91 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (oxazole-4-carbonyl) piperazine. A white solid was obtained.
  • the compound 92 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (2-Hydroxyethyl)piperazine. A white solid was obtained.
  • the compound 93 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of the N-(cyclopropanecarbonyl)pyridazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (1H-1,2,4-triazol-1-carbonyl)piperazine. A white solid was obtained.
  • the compound 94 of the present invention is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (tetrahydrogen Furan-2-2-carbonyl) piperazine. A white solid was obtained.
  • the compound 95 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared. - (Tetrahydrofuran-2-carbonyl) piperazine. A white solid was obtained.
  • the compound of the present invention 96 is prepared according to the scheme 4, and the synthesis method is as described in Example 7, except that instead of N-(cyclopropanecarbonyl)pyrazine, the N corresponding to the R in the formula (I) and the substituted ring A in the compound 96 are first prepared. - (4,4-difluorocyclohexyl)piperazine-1 -carboxamide. A white solid was obtained.
  • the compound 97 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the N of the compound A corresponding to the R ring substituted by the R' in the formula (I) is first prepared. -cyclopropylpiperazine-1 amide. A white solid was obtained.
  • the compound 98 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (1H-imidazolium-1-carbonyl)piperazine. A white solid was obtained.
  • the compound 99 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (cyclohexanecarbonyl)piperazine. A white solid was obtained.
  • the compound 100 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - Propionyl piperazine. A white solid was obtained.
  • the compound 101 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropane)piperazine, the compound A in the corresponding compound of the formula (I), the substituted A ring, was first prepared. 4-(Cyclopentylcarbonyl)piperazine. A white solid was obtained.
  • the compound 102 of the present invention was prepared according to Scheme 3, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl) was replaced. bundle.
  • Qin firstly the preparation of compound 102 corresponding to the general formula (I), R '4 A ring substituted - (4-benzyloxycarbonyl - piperidinyl) carbonyl piperidine ⁇ Qin. A white solid was obtained.
  • the compound 103 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula 103 corresponding to R in the formula (I), substituted ring A is prepared. - benzoylpiperazine. A white solid was obtained.
  • the compound 104 of the present invention was prepared according to the scheme 3, and the synthesis method was carried out in the same manner as in Example 7, except that instead of ⁇ -(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (4-Fluorophenyl) piperazine. A white solid was obtained.
  • the compound of the present invention 105 is prepared according to the scheme 3, and the synthesis method is the same as in the example 1, except that N-(cyclopropanecarbonyl)azine is substituted, and in the compound 105, the corresponding ring of the formula (I) and the substituted ring of the ring A are prepared. (4-Benzyloxycarbonyl-piperidine)carbonylpiperazine. A white solid was obtained.
  • Compound 106 of the present invention was prepared according to Scheme 3, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)pyridazine, the 4 ring corresponding to the R-substituted A ring in the compound (I) was first prepared. - (4-Nitrobenzoyl) piperazine. A white solid was obtained.
  • the compound 107 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)methane, the 4 ring of the compound A corresponding to the R ring substituted in the formula (I) is prepared. - ( 4,4 -difluorocyclohexanecarbonyl) ⁇ - beamazine. A white solid was obtained.
  • the compound 108 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (4-Benzyloxycarbonyl-pyrrolidine)carbonylpiperazine. A white solid was obtained.
  • the compound 109 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (Pyrrolidin-2-carbonyl) piperazine. A white solid was obtained.
  • the compound 110 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of the N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, the substituted ring A is prepared. - (4-Fluorophenyl) piperazine. A white solid was obtained.
  • the compound 111 of the present invention was prepared according to the scheme 4, and the synthesis method was carried out in the same manner as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A were prepared. - (Tetrahydrofuran-3-carbonyl)piperazine.
  • the compound 116 of the present invention is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that N-(cyclopropanecarbonyl)piperidinyl 11 is substituted, and the compound R in the corresponding formula (I) corresponding to R, substituted A ring is first prepared. 4-(4-Fluorobenzoyl) piperazine. A white solid was obtained.
  • the compound 117 of the present invention is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanecarbonyl)azine, the compound 117 corresponding to R in the formula (I), the substituted ring of the ring A is prepared. (4-fluorobenzyl) piperazine. A white solid was obtained.
  • the compound of the present invention 118 is prepared according to the scheme 4, and the synthesis method is referred to the example 7, except that instead of N-(cyclopropanecarbonyl)azine, the piperazine corresponding to the R in the formula (I) and the substituted ring A is prepared first. . A white solid was obtained.
  • the compound 119 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin, 4 - pyridinyl piperazine corresponding to R in the formula (I), substituted A ring, in the compound 119 is first prepared. A white solid was obtained.
  • the compound of the present invention 120 was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropanecarbonyl)piperidin was replaced.
  • Qin, 4-(tetrahydrofuran-2-carbonyl)piperazine corresponding to R in the formula (I) and substituted A ring is first prepared in the compound 120. A white solid was obtained.
  • the compound 125 of the present invention was prepared according to Scheme 4, and the synthesis method was referred to Example 7, except that N-(cyclopropane tracing) was replaced.
  • N-(cyclopropane tracing) was replaced.
  • 4-(1-aminocyclopropanecarbonyl)piperazine corresponding to the R ring substituted by R' in the formula (I) is first prepared. A white solid was obtained.
  • the compound of the present invention 126 is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropionyl)piperazine, the corresponding compound R in the formula (126) corresponding to R, substituted A ring is prepared. 4-(1-(Didecylamino)cyclobutanecarbonyl)piperazine. A white solid was obtained.
  • the compound of the present invention 127 is prepared according to the scheme 4, and the synthesis method is the same as in the example 7 except that instead of N-(cyclopropanedecyl)piperazine, the compound R in the compound 127 corresponding to R in the formula (I) is first prepared. 4-(1-(decylamino)cyclobutanecarbonyl)piperazine. A white solid was obtained.
  • the compound of the present invention 128 is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperole, the compound 4 in the corresponding formula (I) corresponding to R, substituted A ring 4 is prepared. - (1 -Aminocyclobutanecarbonyl)piperazine. A white solid was obtained.
  • the compound of the present invention 129 is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 corresponding to R in the formula (I) and the substituted ring A are prepared first. - (1,2-Methylpyrrolidine-2-carbonyl)piperazine. A white solid was obtained.
  • the compound of the present invention 130 is prepared according to Scheme 4, and the synthesis method is the same as in Example 7, except that instead of N-(cyclopropanecarbonyl)piperazine, the compound 4 in the formula (I) corresponding to R, substituted A ring is prepared. - (2-Methylpyrrolidine-2-carbonyl) piperidine. Qin. A white solid was obtained.
  • PARP-1 catalyzes the transfer of ADP-ribose by NAD+ to adjacent nuclear proteins such as histones.
  • the activity of PARP was judged by chemiluminescence using a HT Universal Chemiluminescent PARP Assay Kit With Histone-coated Strip Wells kit purchased from TREVIGEN to detect the poly ADP-ribose of biotin bound to histone.
  • the positive reference compound used was AZD-2281 from SelleckChem.
  • the kit adopts the ELISA method, that is, adding ⁇ /well sample, ⁇ /well PARP, PARP cocktail 25 ⁇ 1 ⁇ 11 in a 96-well plate coated with histone, the total volume is 50 ul/well, and a blank control (excluding Enzyme And sample) and negative control (without sample), incubate for 60 min at room temperature; wash twice with l x PBS (+0.1% Triton X-100), add Strep-HRP 5 ( ⁇ l/well, incubate for 60 min at room temperature; Wash twice with PBS (+0.1% Triton X-100) and add a chemiluminescent substrate of 100 ⁇ /well.
  • inhibition rate [i- ( RLU sample) -RLU blank) / ( RLU negative - RLU blank) ]> ⁇ 100%.
  • RLU relative luminescence intensity
  • the well-preserved breast cancer cell line MDA-MB-436 was inoculated into the corresponding 96-well culture plate at 5000 cells/well and 3000 cells/well, and incubated overnight in a 37-inch, 5% CO 2 and saturated humidity incubator. . On the next day, after adding a series of concentration gradient samples, the culture was continued for 6 days. The inhibition of cell growth was detected by SRB method, and the growth inhibition rate of the samples was calculated. IC 5 was calculated by 4 Parameter Logistic Model in Xlfit software. The results are shown in Table 2 below.
  • the solid or suspension dosage form In order for the compound after oral administration to be absorbed in the intestine, the solid or suspension dosage form must disintegrate, dissolve and diffuse to the surface of the intestinal epithelial cells before entering the body, and the more soluble compound can diffuse more into the intestinal epithelial cells.
  • the surface is such that it can absorb a larger amount of drug per unit time, unit surface area, incomplete absorption of insoluble compounds, and thus oral bioavailability is low (see Drug Metabolism and Disposition, 1998, 26, 152-163; Journal of Medicinal Chemistry, 2001, 44, 1313-1333).
  • an excess sample to be tested is added to a phosphate buffer solution having a pH of 7.4, and the drug is no longer dissolved after sonication for 30 minutes, and the sample is continuously shaken at a constant temperature of 25 ° C for 24 hours to dissolve the sample in the solution.
  • the composition and the undissolved fraction were equilibrated in both thermodynamics and kinetics, and then filtered to obtain a filtrate.
  • the content of the sample to be tested is determined, so as to determine the balance of the sample to be tested in the citrate buffer solution with a pH of 7.4. Solubility of S.
  • test solution was injected 3 times according to the above chromatographic conditions, the peak area was measured, and the corresponding concentration was calculated.
  • the preparation method and activity and solubility characteristics of representative compounds of the present invention are described in detail above by way of Preparation Examples and Test Examples.
  • the compound of the present invention can be synthesized by a conventional method and has better activity and solubility than the currently best-acting compound AZD-2281.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Diabetes (AREA)
  • Neurology (AREA)
  • Hematology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Cardiology (AREA)
  • Emergency Medicine (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

本发明公开了一种邻苯二甲酰肼(酞嗪酮)类化合物,以及包括该类化合物的药物组合物。它们作为DNA修复酶多聚(ADP-核酶)聚合酶的抑制剂,可有效治疗涉及PARP酶活性的疾病,包括癌症、神经变性疾病、炎症等。

Description

一种多聚(ADP-核糖)聚合痺抑制剂
技术领域
本发明涉及一种多聚(ADP-核糖) 聚合酶抑制剂, 具有涉及邻苯二曱酰肼 (舦嗪 酮)类化合物, 其组合物, 以及在制备治疗与 PARP酶活性相关的疾病的药物中应用。 背景技术
多聚(ADP-核糖) 聚合酶(poly (ADP-ribose) polymerase, PARP )是存在于真核 细胞中催化聚 ADP核糖化的细胞核酶, 它参与的聚 ADP核糖化是真核细胞中蛋白质 翻译后的重要修饰方式之一。 PARP发现 40多年来, 其在 DNA损伤修复和维持基因组 稳定性方面的重要作用引起广大学者的关注,尤其是近年来对 PARP与肿瘤发生的关系 及利用调节 PARP来提高肿瘤治疗效果的研究取得了令人鼓舞的成绩。
目前研究发现, PARP家族至少有: PARP-1 , PARP-2, PARP-3 , PARP-4/ VPARP , 端锚聚合酶(Tankyrases, TANK-1 , TANK-2和 TANK-3 )等亚型。 在人类基因组中共 有 16个不同基因编码 PARP超家族成员这些成员都具有高度保守的由 50个氨基酸残基 组成的 PARP催化活性区, 除此之外, 它们具有不同的一级结构、 细胞内定位和特异底 物。 这说明聚 ADP核糖是一类具有不同生物功能的转录后调节蛋白。
PARP-1是由 1014个氨基酸残基组成的一条多肽链, 分子量为 113kDa, 等电点在 8.0 ~ 9.8之间。它是 PARP家族中最早被发现,而且特性被了解得最清楚的成员。 PARP-1 肽链中有 3个功能性结构域。① DNA结合结构域(DNA binding domain, DBD, 46kDa ), 位于 N-末端第 1 - 372 氨基酸残基之间, 包含一个核定位序列 (nuclear localization sequence, NLS )和两个锌指结构 ( Zn finger )。 这两个锌指结构参与识别 DNA缺口, 其中第 1个锌指结构识别 DNA单链和双链断裂, 它的突变会大大降低 PARP的激活; 第 2 个锌指结构只参与 DNA单链断裂的识别。 ②自我修饰结构域( automodifieation domain, 22kDa ), 位于第 374-525氨基酸残基之间。 PARP-1通过该结构域与 ADP糖 基结合, 发生 PARP-1 自身糖基化, 并且该结构域还可以使 PARP-1形成二聚体。 ③ C- 末端第 524-1,014氨基酸残基之间为催化结构域( catalytic domain, 54 kDa )。 该结构域 是把 NAD 转化为 ADP核糖的基础, 此序列高度保守。 尤其第 859-908氨基酸残基区 段在脊推动物中的保守性达 100 %。 催化结构域由两部分组成, 其中靠近 N-末端部分 的氨基酸残基第 662-784位形成 A、 B、 C、 D、 E、 F六个 ot-螺旋, 此结构是 PARP特 有的, 螺旋区通过 F oi -螺旋与活性区, 即 NAD 结合位点结合, 可能与激活信号的转 递有关。 另外, 有研究发现 PARP还具有亮氨酸拉链结构, 推测可能在 PARP与其它蛋 白质或 PARP自身形成二聚体时发挥作用。
PARP-1参与 DNA缺口的识别, 识别 DNA缺口后活化的 PARP-1形成同型二聚体 并催化 NAD+分解成尼克酰胺和 ADP核糖,利用后者为原料使核受体蛋白聚 ADP核糖 化, PARP-1的激活与 DNA的损伤程度呈正相关, 并且在 DNA损伤诱发的聚 ADP糖 基化反应中, PARP- 1可通过自身修复作用对 PARP- 1活性进行负反馈调节, 形成 DNA 损伤激发的 PARP-1 活性循环。 PARP-1 的抑制研究或基因缺陷小鼠的研究可以肯定 PARP-1在维持基因组稳定性方面起着关键的作用。
在人体细胞中, PARP是相当丰富的, 尤其是在免疫细胞和生殖细胞中。 很多生理 过程中都有聚 ADP核糖化发生, PRAP的多重作用包括染色质的降解、 DNA的复制、 DNA修复、 基因的表达、 细胞的分裂和分化以及细胞的凋亡。
当 DNA损伤时, PARP-1酶被激活, 与 DNA结合, 催化聚腺苷二磷酸核糖基化, 从而启动了 DNA的损伤控制和修复过程。 同时它的过度反应将导致 NAD+/ATP能量耗 尽, 最终导致细胞坏死。 PARP-1的这种看似矛盾的双重作用引起生物学家和药物化学 家的广泛兴趣。因此,抑制 PARP-1活性的小分子化合物已经被广泛地作为抗肿瘤治疗、 抑制神经损伤和炎症损伤的药物研究。 发现和寻找 PARP-1酶的抑制剂具有重要意义。
从分子水平而言, 恶性肿瘤的治疗策略多数是利用损伤肿瘤细胞 DNA的方法, 如 放疗和化疗等, 来杀伤肿瘤细胞。 因此针对参与 DNA损伤识别、 反应和修复的分子靶 向治疗探索一直是近年来研究的热点。 PARP- 1在 DNA修复、 细胞死亡、 增殖分化等 方面发挥着重要作用, 推测通过抑制 PARP-1活性可抑制 PARP-1介导的 DNA修复机 制, 提高放疗和化疗对肿瘤细胞 DNA的损伤, 因而对肿瘤可能具有潜在治疗价值。
目前己有多种 PARP-1 抑制剂应用于临床药物研究, 例如已开发的 BSI-201、 AZD-2281、 ABT-888、 NU1025、 GP 115427, AGO 14699. CEP-6800、 AG14361和 INO1001 等。 其中, 由阿斯利康公司开发的 AZD-2281 ( olaparib /KU-59436)是小分子口服 PARP -1抑制剂, 主要用于治疗卵巢癌、 乳腺癌和实体瘤。 在 2009 年 4 月, 阿斯利康公司 报道了 AZD-2281 首次作为单药在 I期临床中对胃癌表现出明显的抑制作用, 且耐受 性良好。 目前该药物与顺铂、 卡铂、 紫杉醇等药物联用治疗实体瘤的研究正处于 II期 临床阶段。 主要针对遗传性乳腺癌和卵巢癌的临床研究已进入 III期临床研究阶段。 此 外, 辉瑞公司开发的一种新型 PARP-1抑制剂 AG014699有望与烷化剂 TMZ联合治疗 恶性黑色素瘤, 该药正处于 II期临床研究阶段。
无论从分子水平、 细胞水平, 还是从整体水平, 这些 PARP-1抑制剂在联合化疗、 放疗或单独化疗方面的治疗研究都取得一些重要进展。许多研究发现 PARP-1抑制剂可 加强多种抗肿瘤药(如烷化剂、拓朴异构酶抑制剂)的疗效。 Tentori 等( Blood 2002, vol. 99: 2241 )发现 NU1025可延长脑淋巴瘤荷瘤小鼠的生存期。 Delaney等( Clin Cancer Res 2000, vol. 6: 2860 )采用 12个人类肿瘤细胞株(包括肺、 结肠、 卵巢、 乳腺的肿瘤细胞 系各 3个),发现 PARP-1抑制剂 NU1025或 NU10285与抗肿瘤药烷化剂 ( temozolomide, TMZ )或喜树碱类( topotecan, TP )联用可增强后二者对瘤细胞的生长抑制作用。 NU1025 和 NU10285 的抑瘤增强作用与二者对 PARP-1 的抑制强度有关。 Guilford公司开发的 PARP-1抑制剂 GPI15427可增强 TMZ的作用, 明显延长多形性胶质母细胞瘤、 脑淋巴 瘤或颅内恶性黑色素瘤荷瘤小鼠的生存期。 Calabrese等( J Natl Cancer Inst 2004, vol. 96: 56 )研究皮下移植结肠癌细胞的小鼠发现, AG14361 会增强 TMZ 的抗肿瘤作用。 Miknyoczki等( Mol Cancer Ther 2003, vol. 2: 371 )用 PARP-1抑制剂 CEP-6800分别与 TMZ, CPT-11 (喜树碱类)或顺铂联用治疗临床相应肿瘤 (即, U251MG人类胶质母 细胞瘤, HT29 人类结肠腺癌和 Calu-6 非小细胞肺癌) 的异种移植模型, 研究表明 CEP-6800可分别使 TMZ、 CPT-11或顺铂处理后的肿瘤细胞停滞于 G2/M期的时间和 / 或比例增加, 并可增强 TMZ、 CPT-11和顺铂对荷瘤棵鼠的治疗效果。
尽管 PARP-1抑制剂联合化疗的研究表明抗肿瘤作用显著增强,但其安全性和可行 性仍然是人们担忧的一个问题。 Miknyoczki等的研究发现, 治疗剂量的 CEP-6800并未 增加这三种化疗药对离体的人类肠上皮和肾脏细胞造成损害, 也不增加活体小鼠 TMZ 的胃肠毒性和顺铂的肾脏毒性, 这预示着 CEP-6800具有联合化疗的价值和可行性。 另 外 PARP-1抑制剂 GPI15427由于半衰期长、 生物利用度高, 通过口服给药局部可达到 有效药物浓度, 在与 TMZ联用治疗的中枢神经系统肿瘤模型取得较好疗效。
已有报道, 活化的 PARP在大量病态的细胞死亡中发挥作用, 暗示 PARP抑制剂可 能在这些状况中具有治疗功效。 比如中风大鼠的病灶性大脑局部缺血之后, 已观察到 增强的多腺苷二磷酸核糖基化, 表明 PARP有所活化( Tokime等, J Cereb Blood Flow Metab 1998, vol. 18: 991 )。 公布的药理学和遗传学数据支持了 PARP-1抑制剂在大脑局 部缺血或中风之后具有神经保护性作用的这种假设。 PARP-1抑制剂能抵御大鼠大脑皮 层培养中 NMDA-或 NO-诱导的神经毒性( Zhang等, Science 1994, vol. 263: 687; Eliasson 等, Nature Med 1997, vol. 3: 1089 ), 观察到具有 PARP-1抑制活性的系列化合物对神经 有着保护作用。
Suto等(美国专利 No. 5,177,075 )研究发现, 在病灶性大脑局部缺血的大鼠模型 中,在局部缺血起始前 2小时和起始后 2小时腹膜给药( 10mg/kg )之后,强有力的 PARP-1 抑制剂 DPQ( 3,4-二氢 -5-[4-(1-哌啶基)丁氧基 ]-1-(2Η)-异喹啉酮)使梗塞体积减少了 54% (永久的 MCAo和 90分钟两边的普通颈动脉闭塞)( Takahashi等, Brain Res 1997, vol. 829: 46 )。 在采用缝合线方法 MCA闭塞 2小时之后, 向脑室内施用较弱的 PARP-1抑 制剂 3-氨基苯曱酰胺( 3-AB ),小鼠中的梗塞体积降低了 47% ( Endres等, J Cereb Blood Flow Metab 1997, vol. 17: 1143 )。 局部缺血 24小时后用另一种 PARP-1抑制剂 3-AB处 理, 减緩了局部缺血组织中 NAD+水平的降低, 并且降低了多腺苷二磷酸核糖多聚体的 合成。 相类似, 3-AB ( 10mg/kg )显著减少了大鼠病灶性局部缺血的缝合闭塞模型中的 梗塞体积(Lo等, Stroke 1998, vol. 29: 830 )。 在大鼠局部缺血的永久性脑动脉闭塞模 型中也观察到了 3-AB( 3-30mg/kg,脑室内( i.c.v. ) )的神经保护作用( Tokime等, J Cereb Blood Flow Metab 1998, vol. 18: 991 )。
此外 PARP基因敲除小鼠( Wang, Genes Dev 1995, vol. 9: 509 )可用于帮助确认 PARP 在神经变性中的作用。 在局部缺血的小鼠缝合线模型中, 观察到 PARP -双等位基因敲 除小鼠中的梗塞体积减少了 80%, 而在 PARP单等位基因敲除小鼠(PARP+/-小鼠) 中 减少了 65%。 Endres等( 1997 道了在 PARP 、鼠中梗塞体积减少了 35%,而在 PARP+/- 动物中减少了 31%。 除了神经保护作用外, PARP 小鼠的实验表明在神经学得分上的 改进并且显示在局部缺血后 NAD+水平有所增加。
PARP的活化也牵涉到功能欠缺, 这些欠缺可从外伤性脑损伤和脊髓损伤中反映出 来。 在外伤性脑损伤的受控皮层影响模型中, PARP_小鼠与 PARP+ _小鼠比较显示, PARP抑制剂显著提高了小鼠的运动和识别功能( Whalen等, J Cereb Blood Flow Metab 1999, vol. 19: 835 )。 过亚硝酸盐( Peroxynitrite )的产生和 PARP的活化已在脊髓损伤大 鼠中得到证实 ( Scott等, Ann Neurol 1999, vol. 45: 120 )。 这些结果暗示 PARP的抑制 剂可提供保护以避免在头部或脊柱创伤后功能丧失。
在局部缺血和再灌注之后,作为细胞中的死亡底物, PARP的作用不限于神经系统。 就此而论, 近期的出版物报道了各种结构不同的 PARP抑制剂, 包括 3-AB和相关化合 物,在兔子心脏局部缺血和再灌注之后减少了梗塞大小( Thiemermann等, Proc Nat Acad Sci 1997, vol. 94: 679 )。在分离的灌注兔子心脏模型中, 在全身局部缺血和再灌注之后, 骨骼肌的坏死也由 PARP抑制剂减少。 据 Zingarelli和其同事 ( Cardiovascular Research 1997, vol. 36: 205 )报道, 在大鼠心肌局部缺血 /再灌注模型中, 3-AB具有相似的心脏 保护作用。这些体内结果得到了培养的大鼠心肌细胞实验数据的进一步支持( Gilad等, J Mol Cell Cardiol 1997, vol. 29: 2585 )。 在用氧化剂如过氧化氢、 过亚硝酸盐和氧化氮 供体处理之后, 观察到 PARP抑制剂( 3-AB和尼克酰胺 )保护了心肌细胞。 近来证实, 在心肌局部缺血和再灌注之后,小鼠中 PARP表达的破坏提供了延迟的细胞损伤保护以 及炎性介体的产生 (Yang等, Shock 2000, vol. 13: 60 )。 这些数据支持了在心肌梗塞之 后施用 PARP抑制剂能有助于减緩病情的假设。 PARP活化也牵连到各种炎性疾病中发生的细胞损伤。通过促炎刺激物的巨噬细胞 的活化可导致产生氧化氮和超氧阴离子, 它们组合生成过亚硝酸盐, 导致 DNA单链断 裂和 PARP的活化。在众多动物模型中使用 PARP抑制剂的实验支持了 PARP参与炎性 疾病的假设。 PARP抑制剂 5-吲哚 -6-氨基 -1,2-苯并吡喃酮在这些动物中减少了关节炎的 发生率和严重性, 降低了坏死的严重性和滑膜增生。 在急性局部炎症的角叉藻聚糖诱 导的胸膜炎模型中, 3-AB抑制了炎性过程中的组织学损伤、 胸膜形成和单核细胞渗入 特性( Cuzzocrea等, Eur J Pharmacology 1998, vol. 342: 67 )。
此外, PARP抑制剂还可用于治疗糖尿病。 在糖尿病病人体内, 血糖浓度长期处于 较高水平, 破坏了内皮细胞的稳定。 如: 高血糖会引发氧化介质从线粒体的电子传递 链中释放 (如 NADH/NADPH氧化酶),并能提高 iNOS的表达水平,导致血管内皮的 iNOS 过度释放。 这些因素产生的过氧化物、 超氧化物导致 DNA链的损伤, 激活了 PARP, 耗竭了细胞的 NAD+, 由此引发的一系列病理过程导致整个细胞的功能紊乱, 进而导致 细胞的死亡。 Heller等人发表在《生物化学杂志》中的研究论文 "在胰岛细胞中,聚(ADP- 核糖) 聚合酶基因的失活影响氧基团和一氧化氮毒性" ( J Biol Chem 1995, vol. 270: 11176 ) 中讨论了 PARP消耗细胞 NAD+并诱导胰岛细胞死亡的倾向。 Heller等人使用 PARP基因失活的小鼠细胞, 置于破坏 DNA 的试剂中后, 这些突变细胞没有表现出 NAD+耗尽。 还发现这些突变细胞抗 NO毒性的能力更强了。
新近的对现有技术的全面综述已经由 Jagtap和 Szab6在 Nature 2005, vol. 4: 421中 发表。被描述为 PARP抑制剂的各种化合物已在下列文献中报道, Hattori等, J Med Chem 2004, vol. 47: 4151和 Menear等, J Med Chem 2008, vol. 51 : 6581。 许多其他 PARP抑制 化合物已是多项专利的主题。例如,描述为 PARP抑制剂的化合物公开在 WO 99/11645、 WO 00/32579、 WO 02/36599 > WO 02/36599、 WO 03/103666、 WO 03/063874、 WO 2004/096779、 WO 2005/023246、 WO 2005/054210、 WO 2006/003148、 WO 2007/138355, WO 2008/017883 US 2004/0248931、 US 2006/0063926和 US 2007/0093489等中。 发明内容
本发明的一个目的在于提供一种邻苯二曱跣肼 (酞嗪酮) 类化合物、 其药学上可 接受的盐、 水合物、 溶剂化物或立体异构体作为新型多聚(ADP-核糖)聚合酶抑制剂。
本发明的另一个目的在于提供含有本发明的化合物、 其药学上可接受的盐、 水合 物、 溶剂化物或立体异构体的药物组合物。
本发明的再一个目的在于提供本发明化合物在制备治疗与 PARP 酶活性相关的疾 病的药物中应用。 所述疾病包括癌症、 神经变性型疾病、 心血管疾病、 糖尿病和炎症。 本发明进一步的 的在于提供一种用本发明的化合物、 其药学上可接受的盐、 水 合物、 溶剂化物或立体异构体, 或者其组合物治疗与 PARP酶活性相关的疾病的方法。
因此根据本发明的一个方面, 本发明提供一种式(I )表示的化合物及其药学上可 接受的盐、 水合物、 溶剂化物和立体异构体:
Figure imgf000007_0001
R选自氢、 氯、 氟、 溴、 碘、 硝基、 羟基、 氨基、 支链或直链 d-C6烷基、 卤代烷 基、 CF3、 CN、 NRUR12、 NH-CO-R13、 O-Q- 烷基, 其中 R11和 R12彼此独立地为氢 或^-^烷基, 而 R13是氢、 d-C4烷基、 d-C4烷基 -苯基或苯;
Xi , X2分别独立地选自 -CH或 -N, 但两者不同时为 -N;
X3选自氢、 氯、 氟、 溴、 碘;
Het :
Figure imgf000007_0002
其中 Y1选自 CH和 N, Y2选自 CH和 N, Y3选自 CH, CF和 N, 其中 Y1 , Y2、 Υ3可以同时为 Ν, 也可以只有一个或两个为 Ν;
Α为 4 ~ 9元饱和或不饱和单环杂环;
R'选自 -、 -C(0)NR!-, -CC^Ri -、 烷基、 环烷基、 环烯基、 烷氧基、 烯氧基、 饱和或不饱和单环杂环、 芳香基、 杂芳香基;
R1选自烷基, 环烷基、 环烯基、 烷氧基、 埽氧基、 饱和或不饱和单环杂环、 芳香 基、 杂芳香基、 叔丁基;
n选自 0 ~ 12;
化合物结构中出现的烷基、 芳香基、 环烷基、 杂芳香基、 饱和或不饱和单环杂环、 叔丁基均可被一个或多个卤素、 羟基、 氰基、 氨基、 亚氨基、 醚基、 硝基、 亚硝基、 巯基、 硫醚、 亚砜、 磺酰基、 硫代酰胺基、 磺酰胺基、 羧酸、 芳香基、 杂芳香基、 C^o 直链或支链烷基、 Cwo直链或支链烯基、 酰基、 酰氧基、 酰胺基、 酰基酰胺基、 脲基、 烷基胺基、 酯基、 苯氧基、 苄氧基、 C3 ^环烷基、 C5-2Q环烯基、 C1-2。烷氧基、 C1-20烯 氧基、 Cwo烷基硫基或 Cwo卤代烷基取代;
且上述提到的杂芳香基和饱和或不饱和单环杂环中的杂原子包括 N、 0、 S。 根据本发明的再一个方面提供一种化合物、 其药学上可接受的盐、 水合物、 溶剂 化物或立体异构体,
Figure imgf000008_0001
其中:
R选自如下组中: 氢、 氯、 氟、 溴、 碘、 硝基、 羟基、 氨基、 支链或直链 d-C6烷 基、 1¾代烷基、 CF3、 CN、 NRUR12、 NH-CO-R13和 O-d- 烷基, 其中 R11和 R12彼此 独立地为氢或 C广 C4烷基, R13是氢、 d-C4烷基、 C,-C4烷基 -苯基或苯;
X X2分别独立地选自 -CH或 -N, 但两者不同时为 -N;
X3选自氢、 氯、 氟、 溴和碘;
H环为:
W 其中 Υ1选自 CH和 Ν, Υ2选自 CH和 Ν, 且 Υ3选自 CH、 CF和 N;
A环为 4 ~ 9元包和或不饱和单环杂环;
R'选自如下组中: -H, -C(0)Rj , -C(0)NR! , -CC R^ 烷基、 环烷基、 环烯基、 烷 氧基、 烯氧基、 饱和或不饱和单环杂环、 芳香基和杂芳香基;
R1选自如下组中: 烷基, 环烷基、 环烯基、 烷氧基、 烯氧基、 饱和或不饱和单环 杂环、 芳香基和杂芳香基;
n为选自 0 ~ 12中的整数;
所述烷基、 芳香基、 环烷基、 杂芳香基、 饱和或不饱和单环杂环和叔丁基中的任 何一个任选地被一个或多个选自如下组中的取代基所取代: 鹵素、 羟基、 氛基、 氛基、 亚氨基、 瞇基、 硝基、 亚硝基、 巯基、 硫醚、 亚砜、 磺酰基、 硫代酰胺基、 磺酰胺基、 羧酸、 芳香基、 杂芳香基、 直链或支链烷基、 Cwo直链或支链浠基、 酰基、 酰氧 基、 酰胺基、 酰基酰胺基、 脲基、 烷基氨基、 酯基、 苯氧基、 苄氧基、 C3-2。环烷基、 C5-20环烯基、 C1-20垸氧基、 C1-20烯氧基、 C1-20烷基硫基和 C1-20卤代烷基; 和 上述提到的杂芳香基和饱和 /或不饱和单环杂环中的杂原子选自 N、 0和5。
上述 C NR,、 烷基氨基等包含氨基的取代基中 N取代的 或烷基可为单取代, 也可为双取代, 且双取代时两个取代基可以相同也可以不相同。
根据 发明的优选实施方式, H环为:
Figure imgf000009_0001
在这种情况下, 优选 A环为取代或未取代的包含 1-3个选自 N、 0和 S的杂原子 的 4-9元饱和或不饱和单环杂环;和 R'选自如下组中: -H, -C(0)Ri , -C(0)NRi , -C02R! , 取代或未取代的 Cwo烷基, 取代或未取代的 C24o烯基, 取代或未取代的 C3-10环烷基, 取代或未取代的 Cwo环烯基,取代或未取代的 CWQ烷氧基,取代或未取代的 C2-1Q烯氧 基, 取代或未取代的含 1-3个选自 N、 0和 S的杂原子的 3-10元饱和或不饱和单环杂 环,取代或未取代 C6-1。芳香基和取代或未取代含 1-3个选自 N、 0和 S的杂原子的 C5-10 杂芳香基。 其中, !^选自如下组中: 取代或未取代的 C1-10烷基, 取代或未取代的 C2-10 烯基, 取代或未取代的 C3-1o环烷基, 取代或未取代的 C5-1Q环烯基, 取代或未取代的 Cwo烷氧基, 取代或未取代的 Cwo烯氧基, 取代或未取代的含 1-3个选自 N、 0和 S 的杂原子的 3-10元饱和或不饱和单环杂环, 取代或未取代的 C6-1Q芳香基和含 1-3个选 自 N、 0和 S的杂原子的 C5-10杂芳香基。 其中所述取代是指被至少一个选自如下组中 的基团所取代: 卤素、 羟基、 氰基、 氨基、 羧基、 C2-10烃基醚基、 硝基、 Cwo烃基磅 酰基、 Cw。烃基磺酰胺基、 C6-1。芳香基、 含 1-3个选自 N、 O和 S的杂原子的 C5-10杂 芳香基、 C1-1()直链或支链烷基、 C1-1()直链或支链烯基、 C1-1Q烃基酰基、 C1-1Q烃基酰氧 基、 C1-1Q烃基酰胺基、 C1-K)烃基酰基酰胺基、 脲基、 (C1-K)烷基) 1-2氨基、 C1-1Q烃基酯 基、 苯氧基、 苄氧基、 C3-1G环烷基、 C5-1Q环烯基、 C1-K)烷氧基、 C1-H)浠氧基、 C1-10烷 基硫基和 Cwo鹵代烷基。
根据进一步优选的实施方式, A环可选自如下组中: 哌嗪、 高哌嗪、 哌啶、 四氢 吡啶、 4-羟基哌啶、 2-曱基哌嗪和 3-曱基哌嗪。
在这种情况下, R'选自如下组中: -H, -C(0)Rj , -C(0)NR1 ; -C02Rj , 取代或未取 代的 Cw。烷基, 取代或未取代的 Cw。烯基, 取代或未取代 C6-U)芳香基, 和取代或未取 代含 1-3个选自 N、 0和 S的杂原子的 Cwo杂芳香基。 其中!^选自如下组中: 取代或 未取代的 CMQ烷基, 取代或未取代的 C2-K)烯基, 取代或未取代的 C3-7环烷基, 取代或 未取代的 Cw环烯基, 取代或未取代的 Cwo烷氧基, 取代或未取代的含 1-3个选自 N、 O和 S的杂原子的 5-7元饱和或不饱和单环杂环, 取代或未取代的 C6-1。芳香基, 和取 代或未取代的含 1-3个选自 N、 0和 S的杂原子的 C ^杂芳香基。 所述取代是指被至 少一个选自如下组中的基团所取代: 卤素、 羟基、 氰基、 氨基、 硝基、 C6.10芳香基、
C1-10直链或支链烷基、 C1-1Q烃基酰氧基、(C1-1G烷基) 1-2氨基、 c1-1()烃基酯基、 苯氧基、 苄氧基、 Cw环烷基、 Cwo烷氧基和 Cwo卤代烷基。
根据本发明更有优选的实施方式, 所述化合物由下式(II )或(ΙΠ )表示:
Figure imgf000010_0001
在以上式(II )或(III ) 中, 优选 R'为 -C(0)R! , 其中 1^选自如下组中: 取代或 未取代的 C3.6烷基, 取代或未取代的 Cw环烷基, 取代或未取代的含 1-2个选自 N、 0 和 S的杂原子的 5-6元饱和或不饱和单环杂环和取代或未取代的苯基,所述取代是指被 至少一个选自如下组中的基团所取代: 鹵素、 羟基、 氣基、 氛基、 硝基、 笨基、 Cl.3直 链或支链烷基、 (C1-3烷基) 1-2氨基。
式(II )或(III ) 中进一步优选 R为选自如下组中的基团: 氢、 氟、 氯、 溴、 硝基 和氨基; 且 X2分别为 -CH。
更进一步优选 R'为选自如下组中的基团: 任选地被选自氟、 氯、 硝基、 氨基、 曱 基氨基、 二甲基氨基、 曱基和苯基中的一个或两个取代基所取代的乙基、 丙基、 丁基、 戊基、 环丙烷基、 环丁烷基、 环己烷基、 苯基、 吡咯烷基和哌啶基。
上述丙基、 丁基和戊基可以是直链的也可以是支链的, 支链更为优选。 例如, 丙 基优选异丙基, 丁基优选叔丁基、 戊基优选 1-乙基丙基等。
此外, 本文中提及的烷基、 烯基、 烃基, 如未特别指明则均指直链或支链的烷基、 烯基、 烃基。
本发明式(I ) 的化合物具体地可为以下化合物 1-130:
化合物
结构式与命名
编号
0
1
0
4- ( 3- ( 4- (环丙烷羰基)哌嗪小叛基)苄氧基) -2Η-酞。秦小酮 0
0
— (3- (4-苯曱酰基 1-酮
Figure imgf000011_0001
- (3- ( 4-丙酰基哌 氧基) -2H-酞嗪 -1-酮
0
0
- ( 3- ( 4 - (环己烷羰基)哌。秦小羰基)苄氧基) -2Η-酞嗪 -1-酮
0
0
- (3- (4- (环戊烷羰基)派。秦小羰基)苄氧基) -2H-酞嗪小酮
Figure imgf000011_0002
- (3- (4 -乙酰基哌咯 -1 -羰基)苄氧基) -2H-酞嗪 -1-酮
0
0
- ( 3- ( 4 - (环丙烷羰基)哌嗪 -1 -羰基)苯氧基) -2Η-酞嗪小酮
Figure imgf000012_0001
0
4- (3- (4- (1-苄 ) -2H-酞嗪 -1-酮
\工 _
Figure imgf000013_0001
4 - (3- (4-对硝基苯基哌嗪 -1-羰基)苄氧基) -2H-酞嗪小酮
4 - (3- ( 4-对硝基苯曱酰基哌嗪小羰基)苄氧基) -2Η- Ο舦嗪 -1-酮
4- (3- (4-苄基 -1, 嗪 -1-S同
Figure imgf000013_0002
4- (3- (4- (3-氯 -4-三氟曱基-苯基) -4-羟基哌啶 -1-羰基)苄氧基) -2Η- 狀嗪 -1-酮
0
0
4- ( 3- ( 4 - (4-氟苯基) -1,2,3,6-四氢吡啶小羰基)苄氧基 -2H-酞嗪小酮
Figure imgf000014_0001
- ( 3- ( 4- (嘧啶 -2-基)哌嗪小羰基)苄氧基) -2H-酞嗪 -1-S同
0 ^ ο 〇= η 。
\工 0
- (3- (4-叔丁氧羰基 -1,4-二氮杂环庚小羰基)苄氧基) -2Η-酞嗪小酮
0
〕Η
0
- (3- (4- (3-氯-苯基) -4-羟基哌啶小羰基)苄氧基) -2Η-酞嗪小酮
- ( 3- ( 4- (4-氯-苯基) -4-羟基哌啶 -1-羰基)苄氧基) -2Η-狀咯 -1-酮
0
0
- (3- (4-苯基哌 -1 - 羰基)苄氧基) -2Η-酞嗪小酮
Figure imgf000014_0002
- (3- ( 4-苯基 -1,2,3,6- 四氢吡啶 -1-羰基)苄氧基) -2Η-酞嗪小酮
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
4 - ( 4 -氟 -3 - ( 4 - ( 4 - 氟苯曱酰基)哌嗪 -1 -羰基)苯氧基 ) -5 -硝基 -2H- 酞嗪 -1-酮
Figure imgf000018_0001
4- (4- 氟 -3- (4-苯甲酰基哌嗪小羰基)苯氧基) -5-硝基 -2H-酞嗪 -1-酮
Figure imgf000018_0002
8— 基—4- (2-氟— (4- ( 1-苄氧羰基-哌啶)羰基哌嗪 -1-羰基)苯氧基) -2H- 酞嗪 -1-酮
Figure imgf000018_0003
4- (4-氟 -3- (4 - (4-硝基笨甲酰基)哌嗪 -1 -羰基)苯氧基) -5-硝基 -2H- 酞嗪 -1-酮
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
0
0
4- (2- 氟 -5- (4- 氧代 -3,4-2H- 酞嗪 -1 - 基氧基)苯曱酰基) -N, N- 二曱基哌嗪 -1 - 甲酰胺
0
4- (4-氟 -3- (4- (3-三氟甲基 -4曱基 -苯曱酰基)哌嗪-卜 羰基)苯氧基) -2H-酞嗪 -1-酮 θ Η
0
4- (3 - (4 -丙烯酰基哌嗪 -1 -羰基 ) -4 -氟苯氧基 ) -2H-酞嗪小酮
0
0
4- (4- 氟 -3- (4- (2-羟基丙酰基)哌嗪 -1 -羰基)苯氧基) -2H-酞嗪 -1-酮 0
4- (3- (4- (4, 4-二氟哌啶 -1-羰基 )哌啶 -1 -羰基 ) -4 -氟苯氧基 ) -2H-酞 嗪 -1-酮
0
0
4- (4-氟 -3- (4- (哌啶 -1 -羰基)哌啶 -1-羰基)苯氧基) -2H-狀嗪 -1-S同
0
0
1 - (2- 氟 -5- (4- 氧代 -3,4-2Η- 酞嗪 -1 -基氧基)苯曱酰基) -Ν-苯基哌 啶 -4- 曱酰胺
0
0
N-乙基小(2-氟 -5- (4-氧代 -1,4-二氢-酞嗪小基氧基)苯甲酰基)哌啶 -4- 曱酰胺 0
4- (3- (4- ( 4- (二曱基氨基)苯曱酰基)哌嗪 -1 -羰基) -4 -氟苯氧基 ) -2Η- 酞嗪 -1-酮
0
0 1
4- (3- (4- (3- (二曱基氨基)苯甲酰基)哌嗪 -1-羰基) -4- 氟苯氧基) -2H-酞嗪 -1-酮
0
4 - (3- (4-叔丁基哌嗪小羰基) -4-氟苯氧基) -2Η-酞嗪小酮
0
4- ( 4 -氟 -3 - ( 4- (呋喃 -2 -羰基 )哌嗪 -1 -羰基 )笨氧基 ) -2Η-酞嗪 -1-酮
0
4- (4- 氟 -3- (4- 异丙基小羰基)苯氧基) -2H-酞嗪 -1-S同
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
0
0
1-苄氧羰基 -4- (4- (3- ((7-氟-4-氧代-3-1,4-二氢-酞嗪小基氧基) 甲基)苯 甲醜基) ^ -1 -羧基) ^
0
\ /工
0 u-
Figure imgf000032_0001
5—氯— 4- (4-氟 -3- (4- (4-硝基苯曱酰基)哌嗪 -1-羰基)苯氧基) 2H-酞嗪 -1- 酮
Figure imgf000032_0002
4 _ ( 3 _ ( 4 - ( 4,4—二氟环己烷羰基)哌嗪 -1 -羰基) -4 -氟苯氧基 ) 2H-酞。秦 -1-酮
Figure imgf000032_0003
1-苄氧羰基 -2 - (4- (2 -氟 -5 - ( 4 -氧代 -3,4 -二氢二氮杂萘 -1 -基氧基 )苯曱 酰基)哌嗪 -1-羰基)吡咯烷
Figure imgf000032_0004
4— ( 4—氟— 3- ( 4- (吡咯烷 -2 -羰基)哌咯 -1 -羰基)苯氧基 ) 2H-酞嗪 -1 -酮
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
本发明进一步提供一种药用组合物, 包括有效量的上述化合物、 其药学上可接受 的盐、 水合物、 溶剂化物或立体异构体, 以及药学上可接受的载体。 所述药学上可接 受的载体是本领域的常规药用载体, 在此不再赘述。
以上列出的化合物及其药学上可接受的盐、 水合物、 溶剂化物和立体异构体, 以 及其药用组合物可作为药物治疗由 PARP介导的病症, 包括抗癌、神经变性型疾病、 心 血管疾病、 糖尿病和炎症。
因此本发明还提供所述化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立体 异构体, 在制备治疗由 PARP介导的病症的药物中的应用。 所述由 PARP介导的病症选 自癌症、 神经变性型疾病、 心血管疾病、 糖尿病和炎症。
所述癌症具体为组织细胞性淋巴癌、 非小细胞肺癌、 小细胞肺癌、 肺腺癌、 肺鳞 癌、 胰腺癌、 乳腺癌、 前列腺癌、 肝癌、 胃癌、 结肠癌、 直肠癌、 卵巢癌、 宫茎癌、 脑癌、 食道癌、 骨癌、 睾丸癌、 黑色素癌、 皮肤癌、 上皮细胞癌、 前列腺癌、 鼻咽癌、 口腔癌、 白血病, 以及脑、 生殖系统、 淋巴系统、 消化系统肿瘤、 呼吸系统肿瘤和皮 肤肿瘤中的任一种。 PARP活性相关的疾病或症状的发作或发展。 这些症状包括对中枢神经系统的创伤性损 伤, 如脑损伤, 以及与中枢神经系统的创伤性损伤相关的神经元变性。 本发明方法可 治疗的相关症状和疾病包括血管中风、 心脏局部缺血、 大脑局部缺血、 脑血管失调如 多发性硬化症, 以及神经变性疾病如阿尔茨海默氏病、 享廷顿氏舞蹈病以及帕金森病。
本发明所述的化合物可以治疗或预防坏死或凋亡引起的细胞损伤或死亡导致的组 织损伤; 可以緩解神经或心血管组织损伤, 包括随后的局部缺血、 心肌梗死和再灌注 损伤。 还可以用于治疗或预防其他的心血管疾病, 例如, 心绞痛、 心血管缺血。
本发明所述的化合物可治疗的其他 PARP相关的症状或疾病包括炎症如胸膜炎和 结肠炎、 内毒性休克、 糖尿病、 关节炎、 心脏局部缺血、 视网膜局部缺血、 皮肤老化、 慢性和急性疼痛、 出血性休克等。 例如, 在中风症状之后, 经患者施用一种或多种本 发明所述的化合物以防止对脑的损伤或合损伤降到最低。
本发明还提供上述由 PARP介导的病症的治疗方法,包括对患者施用有效量的权利 要求 1-10中任一项所述化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立体异构 体, 或者权利要求 14所述的药用组合物的步骤。
本发明所述的化合物可以纯的形式、 与其他活性成分组合或与可药用非毒性赋形 剂或载体组合施用。 口服组合物一般包括惰性稀释载体或可食用载体。 药学上相容的 粘合剂和 /或佐剂物质可作为组合物的一部分。 片剂、 药丸、 胶嚢和药片等可含有任意 下列成分或相似性质的化合物: 粘合剂如微晶纤维素、 黄蓍胶或明胶; 赋形剂如淀粉 或乳糖; 分散剂如褐藻酸、 Primogel或玉米淀粉; 润滑剂如硬脂酸镁; 助流剂如胶状二 氧化硅; 甜味剂如蔗糖或糖精; 或调味剂如薄荷油、 水杨酸甲酯或橙味剂。 当剂量单 位形式是胶嚢时, 除上述种类的物盾以外, 其可含有液体载体如脂肪油。 此外, 剂量 单位形式可含有各种改进剂量单位的物理形式的其他物质, 例如, 糖衣、 虫胶或肠溶 剂。 进一步, 除活性化合物以外, 糖浆可含有蔗糖作为甜味剂以及某些防腐剂、 染料、 色素和香料。
用于施用的选择性制剂包括无菌水或非水溶液、 悬浮液和乳状液。 非水溶剂的例 子是二曱基亚砜、 醇、 丙二醇、 聚乙二醇、 植物油如橄榄油和可注射的有机酯如油酸 乙酯。 水性载体包括醇和水的混合物, 緩冲的介质和盐水。 静脉内栽体包括流体和营 养补充物、 电解质补充物, 如基于林格右旋糖的那些载体等。 防腐剂和其他添加剂也 可存在, 如抗菌剂、 抗氧化剂、 螯合剂、 惰性气体等。
本发明的化合物也可采用可药用盐、 水合物、 溶剂化物或代谢物的形式。 可药用 盐包括无机和有机酸的碱盐, 这些酸包括但不限于盐酸、 氢溴酸、 硫酸、 磷酸、 甲磺 酸、 乙磺酸、 苹果酸、 乙酸、 草酸、 酒石酸、 柠檬酸、 乳酸、 富马酸、 琥珀酸、 马来 酸、 水杨酸、 苯甲酸、 苯乙酸、 扁桃酸等。 当本发明的化合物包括酸性官能团如羧基 时, 则合适的可药用的羧基阳离子对本领域那些专业技术人员来说是熟知的, 包括碱、 碱土、 铵、 季铵阳离子等。 附图说明
图 1是测试例 3中以化合物 AZD-2281为标准品通过 HPLC测定得到的峰面积与标 准品浓度的关系曲线图。 具体实施方式
以下进一步解释本发明的一般方法。 本发明的化合物可以通过本领域中公知的方 法来制备。 下面以本发明的优选化合物的制备方法为例详细进行说明, 但本发明化合 物的制备方法并不局限于此。 方案 1
Figure imgf000039_0001
Figure imgf000039_0002
邻苯二曱酸酐(la)在冰醋酸的作用下与水合肼反应生成邻苯二曱酰肼(lb)后, 在氢化钠的作用下与 3-氯曱基苯曱酸乙酯(lc)反应生成 3- ((4-氧代 -3,4-二氢 -酞。秦小 基氧基) 曱基)苯曱酸乙酯 (ld), 然后在碱性条件下水解生成 3- ((4-氧代 -3,4-二氢- 酞嗪 -1-基氧基) 甲基) 苯甲酸(le), 最后再与 R,取代的氮杂环, 如哌嗪、 哌啶、 4- 羟基哌啶、 四氢吡啶、 1,4-二氮杂环庚烷(高哌嗪)等, 在 1-乙基 -(3-二曱基氨基丙基) 碳二亚胺盐酸盐 (EDCI), 三乙胺, 1-羟基 -7-氮杂苯并三唑(HOAt)条件下缩合成目 标化合物。 其中 R,的定义如上所述。 方案 2
Figure imgf000040_0001
邻苯二曱酰肼( lb)在三氯氧磷的作用下生成 1, 4-二氯酞咯 (2a), 再与 3-羟基苯 曱酸乙酯(2b)反应, 得到 3- (4-氯二氮杂萘 -1-基氧基)苯曱酸乙酯 (2c) 的粗产品, 粗品在冰醋酸中,加入醋酸钠生成 3-( 4-氧代 -3,4 -二氢 -酞嗪 -1-基氧基)苯曱酸乙酯( 2d ), 然后在碱性条件下水解就可以得到中间体 3- (4-氧代 -3,4-二氢-酞嗪-1-基氧基)苯曱酸 (2e), 最后再与 R,取代的氮杂环, 如哌嗪、 哌啶、 4-羟基哌啶、 四氢吡啶、 1, 4-二氮 杂环庚烷等, 在 1-乙基- (3-二甲基氨基丙基)碳二亚胺盐酸盐 (EDCI), 三乙胺, 1- 羟基 -7-氮杂苯并三唑(HOAt)条件下缩合成目标化合物。 其中 R,的定义如上所述。
方案 3
Figure imgf000041_0001
含取代基 R的邻笨二曱酸酐(3a)加水合肼 /冰醋酸条件下生成 R取代的邻苯二曱 酰肼(3b),接着在五氯化磷的作用下生成带取代的双氯酞嗪类化合物(3c )。另一方面, 将 2-氟 -5-曱氧基苯曱腈( 3d )在氢溴酸的作用下生成 2-氟 -5-羟基苯甲酸( 3e ), 再与曱 醇成酯得到 2-氟 -5-羟基苯曱酸曱酯(3f)。 将合成好的含取代基 R的双氯酞嗪类化合物
(3c)在氢化钠作用下与 2-氟 -5羟基苯甲酸曱酯( 3f)反应就可以得到含取代基 R的 5 - (4- 氯二氮杂萘 -1 -基氧基) -2- 氟苯曱酸甲酯(3g), 该化合物在醋 ¾ /醋酸钠条件 下回流就得到 2-氟 -5- ( 4 -氧代 -3,4 -二氢 -酞嗪 -1 -基氧基 )苯曱酸甲酯( 3h ), 然后在碱 性条件下水解就可以得到含取代基的 2-氟 -5- (4-氧代 -3,4-二氢 -酞嗪 -1-基氧基)苯曱酸
(3i), 最后再与 R,取代的氮杂环, 如哌。秦、 哌啶、 4-羟基哌啶、 四氢吡啶、 1, 4-二氮 杂环庚烷等, 在 1-乙基 - (3-二甲基氨基丙基)碳二亚胺盐酸盐 (EDCI), 三乙胺, 1- 羟基 -7-氮杂苯并三唑(HOAt)条件下缩合成目标化合物。 其中 R和 R,的定义如上所 述。 方案 4
Figure imgf000042_0001
4c 双氯酞嗪(2a )在氢化钠作用下与 2-氟 -5-羟基苯曱酸曱酯 (3f)反应就可以得到 5- ( 4-氯二氮杂萘小基氧基) -2-氟苯曱酸曱酯 (4a ) 所得化合物 (4a )在醋^ /醋酸钠 条件下回流就得到 2-氟 -5- ( 4-氧代-3,4 -二氢 -舦嗪 -1-基氧基)苯甲酸曱酯(4b ), 再在 碱性条件下水解就可以得到 2-氟 -5- ( 4-氧代 -3,4-二氢-酞嗪小基氧基)苯甲酸(4c ), 最 后再与 R,取代的氮杂环, 如哌。秦、 哌啶、 4-羟基哌啶、 四氢吡啶、 1, 4-二氮杂环庚烷 等, 在 1-乙基- ( 3-二曱基氨基丙基)碳二亚胺盐酸盐(EDCI ), 三乙胺, 1-羟基 -7-氮 杂苯并三唑(HOAt )条件下缩合成目标化合物。 其中 R,的定义如上所述。
下面通过具体实施例来进一步说明本发明, 但本领域的技术人员应知, 本发明并 不仅限于这些具体实施例。 实施例
以下实施例中的化合物结构是通过核磁共振(NMR )或 /和质谱 (ESI)来确定的。 NMR位移( <5 )以 ΙθΛ ppm )的单位给出。 1H-NMR光傳数据测定使用 Varian Mercury-600 MHz核磁共振测定溶剂为氘代二曱基亚砜( ), 氘代氯仿( CDC13 ), 内标为 四曱基硅烷(TMS )。
ESI-MS的测定使用 Finnigan LCQ Deca质语仪测定。
高效液相 ( HPLC ) 的测定使用 Agilent 1200 LC , Phenomen CI 8 柱 ( 4.6mm* 150ηιηι,0.4μηι )。
IC50值的测定 Envision 2104酶标仪( Perkin Elmer公司)。
薄层层析硅胶板使用烟台黄海 HSGF254硅胶板, 薄层色谱法( TLC )使用的硅胶 板采用的规格是 0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是 0.4mm~0.5mm。
层析柱一般使用烟台黄海 300~400目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成, 或可自阿法 埃莎、 兰卡斯特、 梯希爱、 韶远化学科技和爱斯特公司购得。
实施例中无特殊说明, 反应均在氩气或者氮气气氛下进行。 氩气或者氮气气氛是 指反应瓶连接一个约 1L容积的氩气或者氮气气球。 氢化反应通常抽真空, 充入氢气, 反复操作 3 次。 实施例中无特殊说明, 反应温度为室温。 室温为最适宜的反应温度, 为 20°C~30°C„
实施例中的反应进程的监测采用薄层色谱法( TLC ),反应所使用的展开剂体系有: A: 二氯甲烷和曱醇体系, B: 正己烷和乙酸乙酯体系, C: 石油醚和乙酸乙酯体系, 溶剂的体积比根据化合物的极性不同而进行调节。
纯化化合物采用的柱层析的洗脱体系和薄层色谱法的展开剂体系包括 A:二氯甲烷 和曱醇体系, B: 正己烷和乙酸乙酯体系, C: 石油醚和乙酸乙酯体系, 溶剂的体积比 根据化合物的极性不同而进行调节, 也可以加入少量的三乙胺和醋酸等碱性或酸性试 剂进行调节。 中间体制^^ 1、 2 -氡 -5-羟基苯甲酸甲酴的合成
Figure imgf000043_0001
将 2-氟 -5曱氧基苯曱腈(3d, 购自阿法埃莎)( 5 g, 33 mmol )用氢溴酸( 50 mL )、 冰醋酸( 30 mL )溶解后, 140°C回流 28小时后, 将反应液用乙酸乙酯 100 mL稀释后, 反应液依次用 50 mL水、 饱和氯化钠溶液洗三次,。 合并有机层浓缩干, 得到 2-氟 -5羟 基苯甲酸( 3e )粗品, 白色固体 2.88 g (产率 56% )。 Ή NMR (600 MHz, OMSO-d6) δ 7.19-7.17 (m, 1H), 7.08-7.04 (m, 1H), 6.95-6.92 (m, 1H); ESI-MS m/z: 计算值 156.02, 实 测值 154.91 [M-H]+。
将上面反应得到的 2-氟 -5-羟基苯甲酸( 3e ) ( 780 mg, 5 mmol )用 100 mL曱醇溶解 后, 在冰浴(0°C )搅拌条件下逐滴滴加二氯亚砜(0.8 mL, 10 mmol ), 滴加完后回流 2小时, 反应液浓缩至干后, 柱层析(石油醚: 乙酸乙酯 = 2: 1 )分离得到 2-氟 -5-羟 基笨曱酸曱酯(3f),白色固体 833 mg (产率 98%)。 1H NMR (600 MHz, OMSO-d6) δ 9.73 (s, 1H), 7.20-7.18 (m, 1H), 7.13-7.09 (m, 1H), 6.99- 6.97 (m, 1H), 3.80 (s, 3H); ESI-MS m/z: 计算值 170.04, 实测值 171.25 [M+H]+。 中间体制^^ 2、 3- ((4-氧代-3,4-二氢-8^1-基 ^) 甲基)苯甲酸(le)的制备
Figure imgf000044_0001
邻苯二曱酸酑 ( la, 购自韶远化学科技) ( 10g, 67.6 mmol )溶于冰醋酸( 100 mL) 中, 于冰浴(0°C )搅拌条件下逐滴滴加水合肼(20mL), 滴完后于 125Γ下回流 2h。 TLC监测反应。 冷却至室温, 抽滤, 用水洗至滤液 pH为 6.0, 烘千得到邻苯二甲 酰肼 (lb), 白色固体 10.5g (产率 96%)。 ESI-MS m/z计算值: 162.04, 实测值: 173.15 [M+H]+。
将邻苯二甲酰肼( lb) ( 10 g, 61.7 mmol)用干燥的 N, N-二曱基甲酰胺( 100 mL ) 溶于圆底烧瓶中, 于 0°C下加入氢化钠 ( 1.63 g, 67.9 mmol), 加完后搅拌 0.5h, 于 室温下, 缓慢滴加 3-氯曱基苯甲酸乙酯(lc, 购自韶远化学科技)(13.49 g, 67.9 mmol ) 的 N, N-二曱基甲酰胺(20 mL)溶液, 反应 24h后加入乙酸乙酯,反应液依次用 50 mL 饱和碳酸氢钠、 饱和氯化钠溶液洗三次, 有机层干燥浓缩, 得到的产品用石油醚: 乙 酸乙酯 =10:1柱层析, 得到 3- ((4-氧代 -3- 1,4-二氢-舦嗪小基氧基) 甲基)苯曱酸乙酯 ( Id), 白色固体 6 g (产率 30%)。 1H NMR (600 MHz, OMSO-d6): δ 11.94 (s, 1H), 8.24 (d, 1H,J= 7.50 Hz), 7.97-7.95 (m, 1Η),7.93-7.87 (m, 3H), 7.85-7.83 (m, 1H), 7.57-7.56 (m, 1H), 7.48-7.45 (m, 1H), 5.23 (s, 2H), 4.29-4.25 (m, 2H), 1.28-1.25 (m, 3H); ESI-MS m/z: 计算值 324.1, 实测值 346.9 [ M+Na]
将 3- ((4-氧代 -3- 1,4-二氢-酞嗪小基氧基)甲基)苯甲酸乙酯(Id) (lg, 3.1 mmol) 用 1M的氢氧化钾水溶液完全溶解后, 滴加浓盐酸至白色固体完全析出, 搅拌 lh后过 滤-, 水洗至弱酸性。 得到 3- ( (4-氧代 -3,4-二氢 -酞咯 -1-基氧基) 曱基)苯甲酸(le), 白色固体粗品 0.9 g (产率 98%)。 ESI-MS m/z: 计算值 269.08, 实测值 270.02 [M+H]+。 中间体制^ 3、 3- (4-氣代-3,4-二氢-舦 >1-基 ^)苯甲酸(2e)的合成
Figure imgf000045_0001
2d 2e
将邻苯二曱酰肼 ( lb) ( lg, 6.2 mmol ), 三氯氧磷(2.8 g, 18.6 mmol ), N, N-二 甲基曱酰胺, 依次加入到反应瓶中, 回流两小时后, 将反应液小心緩慢倒入水水中, 然后回升至室温, 过滤, 滤除固体, 滤液减压浓缩, 用二氯曱烷快速柱层析得到 1, 4 二氯酞嗪(2a), 淡黄色固体 281 mg (产率 22.8%)。
室温下, 依次加入 1, 4二氯酞。秦( 2a ) ( 277 mg, 1.4 mmol), N, N-二曱基曱酰胺 (10 mL),碳酸钾(192 mg, 1.4 mmol), 间甲酸乙酯苯酚(2b, 购自韶远化学科技)( 231 mg, 1.4 mol), 120°C回流 2h, 用乙酸乙酯 50mL稀释后, 反应液依次用 50 mL饱和碳 酸氢钠溶液、 饱和氯化钠溶液洗三次, 合并有机相浓缩, 得到 3- (4-氯二氮杂萘小基 氧基)苯曱酸乙酯 (2c)粗品。, 白色固体 340 mg (产率 74%)。 ( lomui zi 'S ςτ ) 、( i0丽 org '§ ι )(qs) ^匸 匸 -ε 'ζ-^-ς #
°(%£6 ) §8θοι ^ta^c^
Figure imgf000046_0001
土: )。Sn ·^ ¾1< '(Ί"ΐΟϋ) 令^。 ¾IL〈??土:)。0 ^ 'ψ 001 ) ¾§« -¾ ( louiui S6 g '§01 ) (¥J i| ) ^ 玄^
ζΙΙΐα 'ΗΒ
Figure imgf000046_0002
°+[H-W] ΙΊ8Ζ '1X83 ?^ : z/w SW.IS3 Ηζ ' ) ςς' L- L '(HZ '冚) 18·/τΖ8·Ζ·
'(HI 'ui) WL-L&L '(Hl'ui) 66H8 '(ZH 6'Z. = * 'HI 'Ρ) Π·8 '(ZH 6L =f 'HI 'Ρ) 9Γ8 '(HI 's) 96ΊΪ 9 :( -OS雨 'z丽 009)爾 N H, 。 °( %66吉^ ) Sui £68 ^gl ^Z)
Figure imgf000046_0003
re 'S i) (ρζ) ιι^^^^ ( «¾--ι-^-ι^'ε- ¾-^ ) -ε —丁^
°+[H- V] t^'60£?H«^ ToieT- : SW-ISH '(Ηε 'ZH I'L =f 'ϊ) 0 I '(HZ I'L =C 'b) LZ^ '(HZ '« 8S'Z 09'乙 '(Η3 'm) 0S'L-£^L '(HI 'ω) &L-LGL '(ΗΙ' ) 00·8-10·8 '(ZH " = " 'HI 'Ρ) Π·8 '(ZH UL =Γ 'HI 'Ρ) 9 8 '(HI 's) ς6ΊΙ 9 :(9P-OSWa 'zHW 009)腦 N H, '( %S6 ^r^)Sui 685^0^^
' 。碑^9 (pz)
Figure imgf000046_0004
W ' ί^'— ^ ' ^ [^α¥ ^ ( IomuI ε '§ui ) '( 03 ) ¾g '( louiui z 'Sui o/,9 ) ¾。T3^9 (^) ^ 玄 ( W -I-^^l^t^ ) 'ί
St'
n9l00/ZT0ZN3/X3d L8.0/CT0Z; OAV 依次加入到反应瓶中, 逐渐升温到 150°C, 待固体融化后, 于 150°C加热回流 2小时, 然后冷却到室温, 加入 200 mL干燥的二氯曱烷, 过滤除去不溶物, 滤液浓缩, 用二氯 曱烷快速柱层析,蒸干溶剂得到 1 , 4-二氯 -5-氯狀嗪( 5c ),白色固体 307 mg (产率 26% )。
将 2-氟 -5-羟基苯曱酸曱酯( 3f )( 940 mg, 5.5 mmol )溶于 N, N-二甲基曱酖胺( 2 mL ) 中, 緩慢的加入氢化钠 (140 mg, 6 mmol), 半小时后, 将反应液緩慢滴加到 1, 4-二 氯 -5-氯酞嗪(5c) (1000 mg, 4.3 mmol)的 N, N-二曱基甲酰胺( 4 mL )溶液中, 继续 搅拌一小时, 加入乙酸乙酯 250 mL稀幹, 反应液依次用 50 mL5饱和碳酸氢钠溶液、 饱和氯化钠溶液洗三次, 合并有机相浓缩干, 得到 2-氟 -5- (8-氯 -4-氧代 -3,4-二氢 -舦。秦 -1-基氧基)苯曱酸甲酯 (5d), 白色固体 1.8 g (产率 95%)。
将 2-氟 -5- ( 8-氯 -4-氧代 -3,4-二氢-酞嗪小基氧基) 苯曱酸曱酯 (5d) ( 700mg, 1.9mmol), 醋酸(20mL), 醋酸钠 (420 mg, 3 mmol )依次加到反应瓶中, 回流一小 时后, 用 50 mL乙酸乙酯稀释, 反应液依次用 50 mL饱和碳酸氢钠溶液、 饱和氯化钠 溶液洗三次,合并有机层浓缩至千,得到 2-氟 -5- (8-氯 -4-氧代 -3 ,4-二氢-酞嗪小基氧基) 苯甲酸曱酯(5e)白色固体 650 mg (产率 95%)。 Ή NMR(600MHZ, DMSO- ): δ 12.12 (s, 1H), 8.13 (d, J= 8.0 Hz, 1H), 7.95-8.00 (m, 1H), 7.83-7.87 (m, 1H),7.74- 7.77 (m, 1H), 7.61-7.64 (m, 1H), 7.42-7.47 (m, 1H), 3.86 (s, 3H); ESI-MS m/z; 计算值 348.0, 实测值 371.6 [M+Na]+。
将上步得到的 2-氟 -5-(8-氯 -4-氧代 -3,4-二氢 -酞嗪 -1-基氧基)苯曱酸曱酯(5e)( lg, 2.87 mmol)用 1M 氢氧化钾溶液溶解,室温搅拌两小时后,再逐滴滴加浓盐酸调 pH=3, 室温搅拌两小时,过滤,得到 2-氟 -5-( 8-氯 -4-氧代 -3,4-二氢 -舦嗪 -1-基氧基)笨曱酸( 5f ), 白色固体 900mg (产率 90% ), ESI-MS m/z: 计算值 334.02, 实测值 357.10 [M+Na] +。
5-硝基、 5-氟取代的 2-氟 -5- ( 4-氧代 -3,4-二氢 -酞嗪 -1-基氧基) 苯甲酸合成与本中 间体制备例 4中 2-氟 -5- (8-氯 -4-氧代 -3,4-二氢 -酞嗪 -1-基氧基) 苯甲酸的合成类似。 中间体制^^ 5、 2- L-5- (4-氧代-3,4-二氢-6^1-基|1^)苯曱酸(4d)的合成
Figure imgf000048_0001
4d
2-氟 -5- (4-氧代 -3,4-二氢 -酞嗪 -1-基氧基) 苯曱酸(4d) 的合成与中间体制备例 4 中 2-氟 -5- ( 8-氯 -4-氧代 -3,4-二氢 -酞。秦 -1-基氧基)苯曱酸(5f) 的合成类似, 只需将起 始化合物 1, 4-二氯 -5-氯酞嗪 (5c)替换为 1,4-二氯 -酞嗪(2a)。
2-氟 -5-( 4-氧代 -3,4-二氢 -酞。秦 -1-基氧基)苯曱酸( 4d ),白色固体。 1H NMR (600 MHz, DMSO) δ 11.93 (s, 1H), 8.24 (d,J=7.8 Hz, 1H), 8.11 (d, J= 7.8 Hz, 1H), 7.99-7.96 (m, 1H), 7.95-7.92 (m, 1H), 7.73-7.58 (m, 1H), 7.60-7.58 (m, 1H), 7.40-7.36 (m, 1H); ESI-MS m/z计 算值 300.1, 实测值 301.24 [M+H]+。 中间体制备例 6、 1-叔丁氧 (4-氟苯甲 S¾基)哌,秦(6c)的合成
Figure imgf000048_0002
对氟苯曱酸(6b) (7 g, 50 mmol)溶于二氯曱烷(100 mL), 依次加入 1-乙基- (3-二曱基氨基丙基)碳二亚胺盐酸盐(EDCI) ( 10.5 g, 55 mmol), 三乙胺(7.7mL, 55 mmol), 1-羟基 -7-氮杂苯并三唑(HOAt) (7.5 g, 55 mmol), 室温搅拌半小时后, 再加入 1-叔丁氧羰基哌嗪(6a) ( 10.2 g, 55 mmol ), 室温搅拌过夜, 次日加水淬灭反 应, 反应液依次用 50 mL饱和碳酸氢钠溶液、 饱和食盐水洗三次, 浓缩有机相得到 1- 叔丁氧羰基 -4- ( 4-氟苯曱酰基 )哌嗪( 6c ), 白色固体 14.8 g (产率 96% )。 ESI-MS m/z 计算值: 308.15, 实测值: 309.25 [M+H]+。 中间体制^ 7、 1-叔丁 (环丙基曱基)哌豢的合成
Figure imgf000049_0001
1-叔丁氧羰基哌嗪(6a ) ( 1.86 g, 10 mmol )溶于二氯曱烷(20 mL ) 中, 加入无 水碳酸钾 ( 829 mg, 6 mmol ), 緩慢滴加氯曱基环丙烷( 7a ) ( 1.1 mL, 12 mmol ) 的二 氯曱烷溶液, 滴完后于室温下反应过夜后, 用水洗, 水层用二氯曱烷洗萃取 2 次, 合 并有机相, 依次用 50 mL饱和碳酸氢钠溶液、 饱和食盐水洗三次, 无水硫酸钠干燥, 柱层析 (石油醚: 乙酸乙酯 =2: 1)分离得白色固体粉末 1-叔丁氧羰基 -4- (环丙基曱基) 哌嗪 ( 7b ), 白色固体 2.01 g (产率 79.0% )。 ESI-MS m/z计算值: 240.18, 实测值: 241.05 [M+H]+。 中间体制备例 8、 1-叔丁氧 (异丙基赛基甲酰基)哌嚎的合成
Figure imgf000049_0002
氮气保护下, 将 N, N'-羰基二咪唑( CDI ) ( 1.62 g, 10 mmol )于冰浴( 0°C )下加 到 1-叔丁氧羰基哌嗪 ( 6a ) ( 1.86 g, 10 mmol )的四氢呋喃溶液( 20 mL ) /三乙胺( 1.36 mL, 10 mmol )溶液中, 搅拌 1小时后, 加入异丙胺( 8a ) ( 0.71 g, 12 mmol ), 室温 反应过夜, 加水稀释反应液, 用乙酸乙酯萃取三次, 合并有机相用 50 mL饱和食盐水 洗 3次, 有机相用无水硫酸钠干燥, 浓缩有机相得到 1-叔丁氧羰基 -4- (异丙基氨基曱 醜基)哌嗪 ( 8b ), 淡黄色固体 1.68 g (产率 62% )。 ESI-MS m/z计算值: 271.19, 实测 值: 294.28 [M+Na]+
叔丁氧羰基哌嗪的 4位上取代基为芳基、烷烃或氨基甲酰基的情况, 以及 1,4-二氮 杂环庚的 4位上取代基为芳基或烷烃的情况, 其合成方法与以上中间体制备例 6、 7、 8 类似。 在此不再重复说明。
实施例 1
本发明化合物 1按照上述方案 1制备:
将 1-叔丁氧羰基 -4- (环丙烷羰基)哌嗪( 190 mg, 0.75 mmol )溶于二氯曱烷后加 入三氟醋酸(l mL ), 室温搅拌至原料反应完后, 用饱和碳酸氢钠溶液洗三次, 浓缩有 机相得到的 N- (环丙烷羰基)哌嗪淡黄色固体 112 mg (产率 97% )备用。 将 3- ( ( 4-氧代 -3,4-二氢-舦嗪小基氧基) 曱基)苯甲酸 ( 148 mg, 0.5 mmol )溶于 N, N-二甲基曱酰胺, 依次加入 1-乙基- ( 3-二曱基氨基丙基)碳二亚胺盐酸盐(EDCI ) ( 115 mg, 0.63麵 ol )、 三乙胺(83.6 μί, 0.63 mmol )、 1-羟基 -7-氮杂笨并三唑( HOAt ) ( 81.6 mg, 0.63 mmol ), 室温搅拌半小时后加入上步所得的 N- (环丙烷羰基)哌嗪, 室温反应过夜后, 加入水淬灭反应, 用乙酸乙酯萃取, 水洗 2 次, 合并有机相, 饱和 食盐水洗, 无水石克酸钠干燥, 浓缩后柱层析分离 (二氯曱烷: 甲醇 =20: 1 )得到白色 固体, 173 mg (产率 88% )。 1H NMR (600 MHz, OM , -d6):d 11.94 (s, 1H), 8.21 (d, 1H, J = 7.68 Hz), 7.99 (d, 1H, J= 7.86 Hz), 7.94-7.92 (m, 1H), 7.90-7.88 (m, 1H), 7.61 (d, 1H, J = 7.62 Hz), 7.55 (s, 1H), 7.49 (t, 1H, J = 7.62 Hz), 7.39 (d, 1H, J = 7.62 Hz), 5.38 (s, 2H), 3.87-3.47 (m, 8H), 1.95 (brs, 1H), 0.69-0.72 (m, 4H); ESI-MS m/z: 计算值 432.1, 实测值 455.1 [M+Na] +。
实施例 2
本发明化合物 2按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 2中相应于通式( I )中的 R,取代的 A环的 4-苯基羰基哌 啶。 得到白色固体。 nH NMR (600 MHz, DMSO-^)^ 11.93 (s, 1H), 8.20 (d, 1H, J = 8.64Hz), 8.00-7.98 (m, 1H), 7.89-7.87 (m, 2H), 7.60 (d, 1H, J = 7.14 Hz), 7.54 (br, 1H), 7.49-7.40 (m, 7H), 5.37 (s, 2H), 3.64-3.34 (m, 8H); ESI-MS w/z:计算值 468.1, 实测值 490.9 [M+Na] +。
实施例 3
本发明化合物 3按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基 )哌嗪,首先制备化合物 3中相应于通式( I )中的 R,取代的 A环的 4-丙酰基哌嗪。 得到白色固体。 Ή NMR (600 MHz, DMSO-^):^ 11.94 (s, 1H), 8.21 (d, 1H, J = 7.80 Hz), 7.99 (d, 1H, J = 7.74 Hz), 7.94-7.92 (m, 1H), 7.90-7.87 (m, 1H), 7.60 (d, 1H, J = 7.68 Hz), 7.54 (s, 1H), 7.49 (t, 1H, J= 7.62 Hz), 7.38 (d, 1H, J= 7.56 Hz), 5.38 (m, 2H), 3.63-3.41 (m, 8H), 2.30 (br, 2H), 0.97 (t, 3H, J = 7.26 Hz); ESI-MS m/z: 计算值 420.1, 实测值 443.1 [M+Na] +
实施例 4
本发明化合物 4按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 4中相应于通式( I ) 中的 R,取代的 A环的 4- (环己烷羰 基 )哌嗪。得到白色固体。 Ή NMR (600 MHz , OMSO-d6):S 11.94 (s, 1H), 8.21 (d, 1H, J = 7.86 Hz), 7.99 (d, 1H, J = 7.86 Hz), 7.94-7.88 (m, 1H), 7.90-7.88 (m, 1H), 7.60 (d, 1H, J = 7.26 Hz), 7.54 (s, 1H), 7.49 (t, 1H, J = 7.68 Hz), 7.38 (d, 1H, J = 7.50 Hz), 5.38 (s, 2H), 3.64-3.40 (m, 8H), 2.09-2.06 (m, 3H), 1.60-1.67 (m, 5H), 1.29-1.30 (m, 3H); ESI-MS m/z: 计算值 474.2, 实测值 497.2[M+Na] +。
实施例 5
本发明化合物 5按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 5中相应于通式( I ) 中的 R,取代的 A环的 4- (环戊烷羰 基)哌嗪。 得到白色固体。 1H NMR (600 MHz, ΌΜΈ,0-ά6):δ 11.94 (s, 1H), 8.21 (d, 1H, J = 7.86 Hz), 7.99 (d, 1H, J = 7.80 Hz), 7.94-7.92 (m, 1H), 7.90-7.87 (m, 1H), 7.60 (d, 1H, J = 7.62 Hz), 7.54 (s, 1H), 7.48 ( t, 1H, J = 7.62 Hz), 7.38 (d, 1H, J = 7.62 Hz), 5.38 (s, 2H), 3.56-3.49 (m, 8H), 2.96-2.90 (m, 1H), 1.49-1.73 (m, 8H); ESI-MS m/z: 计算值 460.2, 实测 值 483.2 [M+Na] +
实施例 6
本发明化合物 6按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 叛基 )哌。秦, 首先制备化合物 6中相应于通式( I ) 中的 R, 取代的 A环的 4 -乙酰基哌 嗪。 得到白色固体。 1H NMR (600 MHz, DMSO-^):^ 11.94 (s, 1H), 8.21 (d, 1H, J = 7.68 Hz), 7.99 (d, 1H, J = 7.92 Hz), 7.95-7.92 (m, 1H), 7.90-7.88 (m, 1H), 7.61 (d, 1H, J = 7.74 Hz), 7.54 (s, 1H), 7.48 (m, 1H), 7.39 (d, 1H, J = 7.62 Hz), 5.38 (s, 2H), 3.63-3.45 (m, 8H), 2.07 (s, 3H); ESI-MS m/z: 计算值 406.1, 实测值 429.1 [M+Na] +。
实施例 7
本发明化合物 7按照方案 2制备,
4 - ( 3- ( 4 - (环丙烷羰基)哌嗪 -1-羰基)苯氧基 ) -2H-酞嗪 -1 -酮, 本发明化合物 7 的制备:
将 1-叔丁氧羰基 -4- (环丙烷羰基)哌嗪 ( 254 mg, lmmol )溶于二氯曱烷后加入 三氟醋酸(1.5 mL ), 室温搅拌至原料反应完后, 用饱和碳酸氢钠溶液洗三次, 浓缩有 机相只是代替 N- (环丙烷羰基)哌嗪, 首先制备化合物 2中相应于通式(I ) 中的 R' 取代的 A环的 4-环己烷羰基。 得到 N- (环丙烷羰基)哌嗪备用。
将 3- ( 4-氧代 -3, 4-二氢 -酞嗪 -1-基氧基)苯曱酸(54 mg, 0.3 mmol ) 溶于 N, N- 二曱基曱酰胺, 依次加入 1-乙基- ( 3-二甲基氨基丙基)碳二亚胺盐酸盐(EDCI ) ( 220 mg, 1.2 mmol )、三乙胺(150 μί, 1.2 mmol ), 1-羟基 -7-氮杂苯并三唑( HOAt ) ( 165 mg, 1.2 mmol ) , 室温搅拌半 ' j、时后加入上步所得的 N- (环丙烷羰基 )哌。秦, 室温反应搅拌 过夜后, 加入水淬灭反应, 用乙酸乙酯萃取, 水洗 3次, 合并有机相, 饱和食盐水洗, 无水硫酸钠干燥, 浓缩后柱层析(二氯曱烷: 甲醇 =20: 1 )分离得到派。秦, 首先制备 化合物 2中相应于通式( I ) 中的 R, 取代的 A环的 4-环己烷羰基。 得到白色固体。 1H NMR (600 MHz, DMSO- 6): δ 11.98 (s, 1H), 8.27 (d, IH, J= 7.38 Hz), 8.10 (d, 1H, J= 7.74 Hz), 7.99-8.01 (m, IH), 7.94-7.96 (m, IH), 7.52 (t, IH, J= 7.86 Hz), 7.39-7.38 (m, 2H), 7.28 (d, 1H, J = 7.62 Hz), 3.33-3.81 (m, 8H), 1.96 (brs, 1H), 0.69-0.74 (m, 4H); ESI-MSm/z: 计 算值 418.16, 实测值 417.89 [M-H] +。
实施例 8
本发明化合物 8按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )唳嗪, 首先制备化合物 8中相应于通式( I ) 中的 R, 取代的 A环的 4 -丙酰基哌 嗪。 得到白色固体。 1H NMR (600 MHz, DMSO-i/6): δ 11.98 (s, IH), 8.27 (d, 1H, J = 7.62 Hz), 8.10 (d, IH, J = 7.74 Hz), 7.99-8.01 (m, IH), 7.94-7.96 (m, IH), 7.51 (t, IH, J = 7.83 Hz), 7.39-7.36 (m, 2H), 7.28 (d, J = 7.62Hz, IH), 3.60-3.48 (m, 8H), 2.33-2.29 (brs, 2H), 0.97 (t, 3H, J = 7.38 Hz); ESI-MS m/z: 计算值 406.16, 实测值 407.09 [M+H] +。
实施例 9
本发明化合物 9按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 9中相应于通式( I ) 中的 R, 取代的 A环的 4 -苯曱酰基 哌嗪。得到白色固体。 1 H NMR (600 MHz, DMSO-rf6): S 11.98 (s, IH), 8.26 (d, 1H, J= 7.80 Hz), 8.09-8.10 (m, IH), 7.98-8.00 (m, IH), 7.96-7.93 (m, 1H), 7.49-7.52 (m, 1H), 7.43-7.44 (m, 3H), 7.40-7.36 (m, 4H), 7.28-7.27 (m, IH), 3.64-3.40 (m, 8H); ESI-MS m/z: 计算值 454.16, 实测值 453.31 [M-H] +。
实旄例 10
本发明化合物 10按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 10中相应于通式(I ) 中的 R'取代的 A环的 4 - (环己烷 羰基)哌嗪。 得到白色固体。 1H NMR (600 MHz, O SO-d6) δ 11.98 (s, IH), 8.26 (d, IH, J = 7.56 Hz), 8.10 (d, 1H, J = 7.80 Hz), 7.98-8.01 (m, IH), 7.93-7.96 (m, IH), 7.51 (t, IH, J = 7.86 Hz), 7.36-7.39 (m, 2H), 7.27 (d, 1H, J = 7.56 Hz), 3.33-3.65 (m, 8H), 2.50-2.64 (m, 1H), 1.68-1.67 (m, 2H), 1.62-1.60 (m, 3H), 1.21-1.33 (m, 5H)。 ESI-MS m/z: 计算值 460.20, 实测值 460.98 [M+H] +
实施例 11
本发明化合物 11按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 11中相应于通式( I ) 中的 R,取代的 A环的 4 - (环戊烷 羰基)哌嗪。 得到白色固体。 1H NMR (600 MHz, OMSO-d6): δ 11.98 (s, IH), 8.26 (d, IH, J = 7.68Hz), 8.10 (d, 1H, J = 7.98Hz), 7.98-8.02 (m, IH), 7.92-7.97 (m, 1H), 7.51 (m, 1H), 7.33-7.40 (m, 2H), 7.27 (d, 1H, J= 7.80Hz), 3.33-3.65 (m, 8H), 2.87-3.02 (m, 1H), 1.68-1.80 (m, 2H), 1.53-1.67 (m, 6H); ESI-MS m/z: 计算值 446.20, 实测值 446.64 [M+H] +。
实施例 12
本发明化合物 12按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 12中相应于通式( I ) 中的 R,取代的 A环的 4 -乙酰基哌 嗪。 得到白色固体。 Ή NMR (600 MHz, DMSO- 6): δ 11.98 (s, 1H), 8.26 (d, 1H, J = 7.74 Hz), 8.10 (d, 1H, J = 7.98 Hz), 7.99-8.01 (m, 1H), 7.94-7.96 (m, 1H), 7.51 (t, 1H, J = 7.83 Hz), 7.39-7.36 (m, 2H), 7.27 (d, 1H, J = 7.62 Hz), 3.33-3.65 (m, 8H), 2.51 (s, 3H); ESI-MS m/z: 计算值 392.15, 实测值 393.26 [M+H] +。
实施例 13
本发明化合物 13按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 13中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4-苄氧 羰基-哌啶)叛基哌嗪。 得到白色固体。 1H NMR (600 MHz, DMSO- 6): δ 11.18 (s, 1H), 7.47-7.46 (d, 1H, J = 7.2 Hz), 7.30-7.29 (d, lH, J = 7.8 Hz), 7.21-7.18 (m, 1H), 7.16-7.13 (m, 1H), 6.72-6.70 (m, 1H), 6.59-6.47 (m, 8H), 4.25 (s, 2H), 3.20-3.18 (m, 2H), 2.77-2.54 (m, 8H), 2.10-2.0 l(brs, 3H), 0.83-0.80 (m, 2H), 0.64 -0.58(m, 2H); ESI-MS m/z: 计算值 595.2, 实测值 617.9 [M+Na] +。
实旄例 14
本发明化合物 14按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 叛基 )哌。秦, 首先制备化合物 14中相应于通式( I ) 中的 R,取代的 A环的 4 -对硝基笨 基哌嗪。得到淡黄色固体。 1H NMR (600 MHz, CDC13): δ 9.52 (s, 1H), 8.41 (d, 1H, J= 7.32 Hz), 8.11-8.09 (m, 2H), 8.04 (d, 1H, J= 7.08 Hz), 7.84-7.80 (m, 2H), 7.62 (brs, 1H), 7.59 (d, 1H, J = 7.68 Hz), 7.49 (t, 1H, J = 7.62 Hz), 7.44-7.43 (m, 1H), 6.79-6.78 (m, 2H) 5.40 (s, 2H), 3.94 (brs, 2H), 3.69 (brs, 2H), 3.46 (brs, 4H); ESI-MS m/z: 计算值 485.17, 实测值 508.05 [M+Na] +。
实施例 15
本发明化合物 15按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 15中相应于通式( I ) 中的 R'取代的 A环的 4 -对硝基苯 曱酰基哌嗪。 得到淡黄色固体。 1H NMR (600 MHz, CDC13): δ 9.52 (s, 1H), 8.42-8.40 (m, 1H), 8.30-8.29 (m, 2H), 8.04-8.03 (m, 1H), 7.85-7.81 (m, 2H), 7.59-7.57 (m, 4H), 7.48 (brs, 1H), 7.40-7.38 (m, 1H), 5.38 (s, 2H), 3.81-3.46 (m, 8H); ESI-MS m/z: 计算值 513.16, 实测 值 536.17 [M+Na] +。 实施例 16
本发明化合物 I6按照方案 1制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 16中相应于通式(I ) 中的 R'取代的 A环的 4-苄基 -1 ,4 - 二氮杂环庚烷。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.51 (s, IH), 8.41-8.39 (m, 1H), 8.04-8.02 (m, IH), 7.81-7.80 (m, 2H), 7.53-7.49 (m, 2H), 7.45-7.40 (m, IH), 7.39-7.32 (m, 3H), 7.28-7.27 (m, 2H), 7.24-7.20 (m, IH), 5.36 (d, 2H, J = 13.2 Hz), 3.81-3.79 (m, 2H), 3.67 (brs, IH), 3.60 (brs, IH), 3.47-3.45 (m, 2H), 2.83 (brs, IH), 2.73-2.72 (m,lH), 2.63-2.58 (m, 2H), 1.99 (brs, IH), 1.78 (brs, IH); ESI-MS m/z: 计算值 468.22, 实测值 491.36 [M+Na]+
实施例 17
本发明化合物 17按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 17中相应于通式( I ) 中的 R,取代的 A环的 4- ( 3 -氯 -4- 三氟甲基 -笨基 )- 4-羟基哌啶。得到白色固体。 1H NMR (600 MHz, CDC13): δ 11.92 (s, IH), 8.18 (m, IH), 8.00 (d, 1H, J = 7.26 Hz), 7.96 (brs, 1H), 7.88-7.83 (m, 2H), 7.80 (d, 1H, J = 8.28 Hz), 7.65 (d, 1H, J = 8.52 Hz), 7.59-7.58 (m, 2H), 7.50-7.47 (m, IH), 7.45-7.44 (d, IH, J = 7.50 Hz), 5.49 (s, 1H), 5.39 (s, 1H), 4.46 (brs,lH), 3.46-3.43 (m, 2H), 3.14 (brs, 1H),
2.10- 1.94 (m, 2H), 1.68 (brs,lH), 1.46 (brs, IH); ESI-MS m/z: 计算值 557.13, 实测值 580.2 [M+Na] + 0
实施例 18
本发明化合物 18按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 18中相应于通式(I ) 中的 R,取代的 A环的 4 - ( 4-氟苯 基) -1,2,3,6-四氢吡啶。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.65 (s, IH), 8.40 (d, IH, J = 9.0Hz), 8.05 (s, IH), 7.81-7.79 (m, 2H), 7.602-7.55 (m, 2H), 7.50-7.45 (m, 2H), 7.32-7.30 (m, 2H), 7.02 (d, 2H, J = 8.4Hz), 6.09-5.80 (m, IH), 5.40 (s, 2H), 4.41 (brs, IH),
4.11- 4.10 (m, 2H), 3.66 (brs, IH), 2.50-2.37 (m, 2H); ESI-MS m/z: 计算值 455.16, 实测值 478.43 [M+Na] +。
实施例 19
本发明化合物 19按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 19中相应于通式( I ) 中的 R,取代的 A环的 4- (嘧啶 -2- 基)哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.95 (s, IH), 8.41 (d, IH, J = 7.68Hz), 8.30 (d, 2H, J = 4.68 Hz), 8.04 (d, 1H, J = 7.92 Hz), 7.85-7.83 (m, IH), 7.82-7.79 (m, 1H), 7.61 (s, IH), 7.57 (d, 1H, J = 7.20 Hz), 7.48 (t, 1H, J = 7.56 Hz), 7.42 (d, IH, J = 7.56 Hz), 6.52 (m, 1H), 5.39 (s, 2H), 3.94-3.79 (m, 6H), 3.53 (brs, 2H); ESI-MS m/z: 计算 值 442.19, 实测值 465.40 [M+Na] +。
实施例 20
本发明化合物 20按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基)嗷。秦, 首先制备化合物 20中相应于通式( I ) 中的 R'取代的 A环的 4-叔丁氧羰 基 -1,4 -二氮杂环庚烷。得到白色固体。 1H NMR (600 Hz, CDC13): δ 10.44 (s, 1H), 8.41 (d, 1H, J = 6.48 Hz), 8.03 (d, 1H, J= 7.50 Hz), 7.83-7.79 (m, 2H), 7.53-7.50 (m, 2H), 7.43 (brs, 1H), 7.34 (brs, 1H), 5.36 (d, 2H, J = 9.72 Hz), 3.81 (brs, 1H), 3.71 (brs, 1H), 3.64 (brs, 1H), 3.49-3.35 (m, 5H), 2.02-1.97 (m, 1H), 1.64-1.60 (m, 1H), 1.47 (s, 9H); ESI-MS m/z: 计算值 478.54, 实测值 501.44 [M+Na] +。
实施例 21
本发明化合物 21按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基)派。秦, 首先制备化合物 21 中相应于通式(I ) 中的 R,取代的 A环的 4- ( 3 -氯- 苯基 ). 4-羟基哌啶。得到白色固体。 1H NMR (600 Hz, CDC13): δ 9.87 (s, 1H), 8.39-8.38 (m, 1H), 8.05-8.03 (m, 1H), 7.82-7.77 (m, 2H), 7.57 (s, 1H), 7.54 (d, 1H, J = 7.50 Hz), 7.47-7.44 (m, 2H), 7.42 (d, 1H, J = 7.62 Hz), 7.32 (d, 1H, J = 7.62 Hz), 7.28 (t, 1H, J = 7.74 Hz), 7.25-7.24 (m, 1H), 5.39 (s, 2H), 4.69 (brs, 1H), 3.68 (m, 1H), 3.58 (m, 1H), 3.32 (brs, 1H), 2.11 (brs, 2H), 1.87 (brs, 2H); ESI-MS m/z: 计算值 489.95, 实测值 512.28 [M+Na] +
实施例 22
本发明化合物 22按照方案 1制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 22中相应于通式(I ) 中的 R,取代的 A环的 4- ( 4 -氯- 笨基 )- 4-羟基哌啶。得到白色固体。 1H NMR (600 Hz, CDC13): δ 9.50 (s, 1H), 8.40-8.38 (m, 1H), 8.05-8.03 (m, 1H), 7.81 -7.79 (m, 2H), 7.56 (s, 1H), 7.54 (d, 1H, J = 7.50 Hz), 7.46 (t, 1H, J = 7.56 Hz), 7.42-7.39 (m, 3H), 7.34-7.32 (m, 2H), 5.39 (s, 2H), 4.70 (brs, 1H), 3.68-3.58 (m, 2H), 3.33 (brs, 1H), 2.14-2.12 (m, 1H), 1.86 (brs, 2H), 1.70 (brs, 1H); ESI-MS m/z: 计算值 489.95, 实测值 512.30 [M+Na] +
实施例 23
本发明化合物 23按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基 )哌咯, 首先制备化合物 23中相应于通式( I )中的 R,取代的 A环的 4-苯基哌嗪。 得到白色固体。 1H NMR (600 Hz, CDC13): δ 9.86 (s, 1H), 8.42-8.40 (m, 1H), 8.06-8.04 (m, 1H), 7.84-7.79 (m, 2H), 7.59 (s, 1H), 7.56 (d, 1H, J = 7.56 Hz), 7.47 (t, 1H, J = 7.56 Hz), 7.42 (d, 1H, J = 7.56 Hz), 7.29-7.28 (m, 2H), 6.92-6.90 (m, 3H), 5.40 (s, 2H), 3.95-3.61 (m, 4H), 3.26-3.13 (m, 4H); ESI-MS m/z: 计算值 440.49, 实测值 463.26 [M+Na] 实施例 24
本发明化合物 24按照方案 1制备, 合成方法参考实施例 1 , 只是代替 N- (环丙烷 羰基)哌嗪,首先制备化合物 24中相应于通式( I )中的 R,取代的 A环的 4 - 苯基 -1,2,3,6 - 四氢吡啶。 得到白色固体。 Ή NMR (600 Hz, CDC13): δ 9.67 (s, IH), 8.41 -8.40 (m, IH), 8.06-8.05 (m, IH), 7.82-7.78 (m, 2H), 7.6 l(s, IH), 7.56-7.55 (m, IH), 7.48-7.43 (m, 2H), 7.36-7.32 (m, 4H), 7.28-7.27 (m, IH), 6.16-6.15 (m, IH), 5.40 (s, 2H), 4.41 (brs, IH), 4.12 (brs, IH), 4.00 (brs, IH), 3.64 (brs, IH), 2.68-2.55 (m, 2H); ESI-MS m/z: 计算值 437.49, 实 测值 460.32 [M+Na] +。
实施例 25
本发明化合物 25按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌咯, 首先制备化合物 25中相应于通式(I ) 中的 R'取代的 A环的 4 - ( 4-氟- 苯基 ) -1 ,2,3,6 - 四氢吡啶。 得到白色固体。 1H NMR (600 MHz, DMSO): Ή NMR (600 MHz, DMSO-i/6): δ 11.97 (s, IH), 8.28 (d, IH, J= 7.8Hz), 8.13 (d, IH, J= 8.4Hz), 8.03-8.00 (m, IH), 7.80-7.95 (m, IH), 7.54 (m, 1H), 7.48 (m, 2H), 7.42-7.40 (m, 2H), 7.33-7.32 (d, IH, J= 7.8Hz), 7.19-7.16 (d, 2H, J= 7.8Hz), 6.21 (m, IH), 4.24 (m, IH), 4.10 (m, IH), 4.04-4.02 (m, IH), 3.83 (m, IH), 3.54 (m, 1H), 1.99 (brs, IH); ESI-MS m/z: 计算值 441.15, 实测值 442.03 [M+H] +o
实施例 26
本发明化合物 26按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 26中相应于通式(I ) 中的 R,取代的 A环的 4 - 异烟酰 基哌嗪。得到白色固体。 1H NMR (600 MHz, DMSO-d6): δ 11.99 (s, IH), 8.66-8.65 (m, 2H), 8.27-8.26 (m, IH), 8.10-8.08 (m, IH), 8.01-7.98 (m, IH), 7.96-7.94 (m, IH), 7.53-7.49 (m, 1H), 7.39-7.35 (m, 4H), 7.29-7.25 (m, IH), 3.73-3.44 (m, 4H), 3.26-3.21 (m, 4H); ESI-MS m/z: 计算值 455.47, 实测值 454.40 [M-H] +。
实施例 27
本发明化合物 27按照方案 2制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羧基 )哌嗪, 首先制备化合物 27中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4-氟苯 甲酰基)哌嗪。得到白色固体 1H NMR (600 MHz, DMSO-^): δ 11.98 (s, IH), 8.27 (d, IH, J = 7.2Hz), 8.10-8.09 (m, 1H), 8.01-7.98 (m, IH), 7.96-7.93 (m, IH), 7.53-7.46 (m, 3H), 7.39-7.36 (m, 2H), 7.28-7.25 (m, 3H), 3.63-3.41 (m, 8H); ESI-MS m/z 计算值 472.47, 实 测值 471.28 [M-H] +。 2012/001611
56 实施例 28
本发明化合物 28按照方案 3制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 28中相应于通式(I ) 中的 R,取代的 A环的 4- ( 4-硝基 苯曱酰基)哌嗪。得到淡黄色色固体。 'H NMR (600 MHZ, OMSO-d6):S 11.89 (s, 1 H), 8.27 (brs, 2H), 7.90 (brs, 1H), 7.80 (brs, 1H), 7.67-7.58 (m, 7H), 5.36 (s, 2H), 3.68-3.42 (m, 8H); ESI-MS m/z: 计算值 531.1 , 实测值 554.0 [M+Na] +。
实施例 29
本发明化合物 29按照方案 3制备, 合成方法参考实施例 1, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 29中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4-苄氧 羰基-哌啶)羰基哌嗪。 得到白色固体。 1H NMR (600 MHZ, DMSO-^):^ 11.90 (s, 1H), 7.93-7.91 (m, 1H), 7.82-7.80 (m, 1H), 7.67-7.58 (m, 2H), 7.54 (brs, 1H), 7.50-7.48 (m, 1H), 7.40-7.39 (m, 1H), 7.37-7.34 (m, 4H), 7.31 -7.28 (m, 1H), 5.36 (s, 2H), 5.06 (s, 2H), 4.00-3.98 (m, 2H), 3.64-3.33 (m, 8H), 2.86 (brs, 3H), 1.61 (brs, 2H), 1.42 (m, 2H); ESI-MS m/z: 计算值 627.2, 实测值 650.0 [M+Na] +。
实施例 30
本发明化合物 30按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )派。秦, 首先制备化合物 30中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4-苄氧 羰基 -哌啶 )羰基哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.90 (s, 1H), 8.43 (d, J = 7.80 Hz, 1 H), 8.13 (d, J = 7.8 Hz, 1H), 7.93-7.91 (m, 2H), 7.89-7.87 (m, 1H), 7.35-7.30 (m, 7H), 7.18-7.15 (m, 1H), 5.12 (s, 2H), 4.22 (brs, 2H), 3.83-3.75 (m, 2H), 3.67-3.56 (m, 4H), 3.50-3.38 (m, 2H), 2.88 (brs, 2H), 2.70-2.63 (m, 1H), 1.80-1.66 (m, 4H); ESI-MS m/z: 计算值 613.23, 实测值 612.41 [M-H] +。
实施例 31
本发明化合物 31按照方案 4制备, 合成方法参考实施例 7 , 只是代替 N- (环丙烷 羰基)派嗪, 首先制备化合物 31 中相应于通式 (I ) 中的 R,取代的 A环的 4- ( 4-硝基 苯甲酰基)派 "秦。得到黄色固体 白色固体。1H NMR (600 MHz, DMSO-i 6) δ 11.97 (s, 1Η), 8.27-8.26 (m, 3H), 8.12-8.08 (m, 1H), 8.01 -7.98 (m, 1H), 7.96-7.93 (m, 1 H), 7.72-7.67 (m, 2H), 7.47-7.34 (m, 3H), 3.76-3.63 (m, 4H), 3.40-3.28 (m, 4H). ESI-MS /z:计算值 517.14, 实测值 516.37[M-H〗+。
实施例 32
本发明化合物 32按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 叛基)哌 σ秦, 首先制备化合物 32中相应于通式( I ) 中的 R,取代的 Α环的 4- ( 5-甲基 异恶唑 -4-叛基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.63 (s, IH), 8.44 (d, J= 7.74 Hz, IH), 8.14 (d, J = 8.04 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.33-7.31 (m, 2H 7.29-7.27 (m, IH), 7.20-7.17 (m, IH), 3.95-3.65 (m, 4H 3.59-3.41 (m, 4H), 1.65 (s 3H)。 ESI-MS m/ 计算值 477.14, 实测值 478.25 [M+H] +。
实施例 33
本发明化合物 33按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 33中相应于通式( I ) 中的 R,取代的 A环的 N-苯基哌嗪 -1-曱酰胺。得到白色固体。 1 H NMR (600 MHz, DMSO- ): δ 9.79 (s, 1H 8.43 (d, J= 7.62 Hz, IH), 8.14 (d, J= 7.74 Hz, IH), 7.93-7.90 (m, 1H 7.88-7.86 (m, IH), 7.33 - 7.27 (m, 6H), 7.18 (t, IH, J = 8.52 Hz), 7.06-7.03 (m, IH), 6.57 (s, 1H 3.70-3.57 (m, 5H), 3.54-3.47 (m, 3H); ESI-MS m/z: 计算值 487.2, 实测值 510.1 [M+Na] +
实施例 34
本发明化合物 34按照方案 3制备, 合成方法参考实施例 7 , 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 34中相应于通式(I ) 中的 R'取代的 A环的 4- ( 4-氟苯 甲酰基 ) 秦。 得到白色固体。 Ή NMR (600 MHz, DMSO-^): δ 12.22 (s, IH), 8.28-8.26 (m, IH), 8.05 (d, J = 8.04 Hz, 1H 7.88 (t, IH, J = 7.95 Hz), 7.49-7.48 (m, 2H), 7.36-7.34 (m, 3H), 7.26 (t, 2H, J = 8.46 Hz), 3.67-3.39 (m, 8H); ESI-MS m/z: 计算值 524.0, 实测值 523.0 [M-H] +。
实施例 35
本发明化合物 35按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 35中相应于通式(I ) 中的 R,取代的 A环的 4 - 笨甲酰 基哌嗪。 得到白色固体。 1H NMR (600 MHz, OMSO-d6): δ 12.22 (s, IH), 8.27-8.26 (d, J = 7.92 Hz, IH), 8.05 (d, J = 7.38 Hz, IH), 7.88 (t, IH, J = 7.89 Hz), 7.44-7.34 (m, 8H), 3.66-3.36 (m, 8H); ESI-MS m/z: 计算值 506.0, 实测值 505.1 [M-H] +
实施例 36
本发明化合物 36按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 锬基 )哌 σ秦, 首先制备化合物 36中相应于通式( I ) 中的 R,取代的 Α环的 4 - ( 2 - 曱 基 p塞 —4 -羰基)哌嗪。得到白色固体得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.17 (s, IH), 8.43 (s, 1H), 8.13 (d, J = 7.62 Hz, IH), 7.94-7.83 (m, 3H), 7.33-7.30 (m, 2H), 7.17-7.16 (m, IH), 4.07-3.83 (m, 6H), 3.51 (brs, 2H), 2.73 (s, 3H); ESI-MS m/z: 计算值 493.12, 实测值 493.95 [M+H] +
实施例 37 N2012/001611
58 本发明化合物 37按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 37中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4 - 氟 苯基)哌嗪。得到淡黄色固体。 1H NMR (600 MHz, DMSO-rf6):6 12.31 (s, IH), 8.32 (d, J= 7.92 Hz, IH), 8.24 (d, J= 7.74 Hz, IH), 8.18 (t, IH, J= 7.95 Hz), 7.50-7.48 (m, 2H), 7.42 (t, 1H, J = 8.67 Hz), 7.08-7.05 (m, 2H), 7.00-6.97 (m, 2H), 3.78 (brs, 2H), 3.40-3.39 (m, 2H), 3.16-3.14 (m, 2H), 3.07-3.05 (m, 2H); ESI-MS m/z: 计算值 507.1, 实测值 506.2 [M+H] +
实施例 38
本发明化合物 38按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 38中相应于通式(I ) 中的 R,取代的 A环的 4 - 氟苯曱 酰基。 得到白色固体得到白色固体。 1H NMR (600 MHz, DMSO-^): δ 12.30 (s, IH), 8.32 (d, J= 8.04 Hz, IH), 8.24 (d, J= 7.68 Hz, IH), 8.16 (t, 1H, J = 7.92 Hz), 7.50-7.45 (m, 4H), 7.41-7.37 (m, IH), 7.28-7.25 (m, 2H), 3.67-3.49 (m, 4H), 3.29-3.16 (m, 4H); ESI-MS m/z: 计算值 535.1, 实测值 534.3 [M-H] +
实施例 39
本发明化合物 39按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 39中相应于通式( I ) 中的 R,取代的 A环的 4 -笨甲跣基 哌嗪。得到淡黄色固体。 1H NMR (600 MHz, DMSO-i/6): δ 12.32 (s, 1H), 8.34-8.33 (m, IH), 8.26 (d, J = 7.74 Hz, IH), 8.18 (t, IH, J = 7.86 Hz), 7.46-7.42 (m, 8H), 3.68-3.42 (m, 8H); ESI-MS m/z: 计算值 517.1 , 实测值 516.1 [M-H] + 。
实施例 40
本发明化合物 40按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)派嗪, 首先制备化合物 40中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 1-苄氧 羰基-哌啶)羰基哌嗪。 得到 黄色固体。 1H NMR (600 MHz, DMSO-i 6): 5 12.32 (s, 1H), 8.33 (d, J = 7.74 Hz, IH), 8.24 (d, J = 7.74 Hz, IH), 8.18 (t, 1H, J = 7.92 Hz), 7.48-7.47 (m, 2H), 7.42-7.40 (m, 1H), 7.38-7.34 (m, 4H), 7.32-7.30 (m, 1H), 5.07 (s, 2H), 4.01-3.99 (m, 2H), 3.66-3.47 (m, 6H), 3.29-3.24 (m, 2H), 2.90-2.78 (m, 3H), 1.65-1.62 (m, 2H), 1.46-1.40 (m, 2H); ESI-MS m/z: 计算值 658.2, 实测值 657.3 [M-H] +
实施例 41
本发明化合物 41按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )味嗪, 首先制备化合物 41中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4-硝基 苯甲酰基)哌嗪。得到 黄色固体。 Ή NMR (600 MHz, DMSO-i/6): 12.29 (s, IH), 8.34-8.15 (m, 4H), 8.18 (s, IH), 7.71 (brs, 2H), 7.49-7.39 (m, 3H), 3.76-3.65 (m, 4H), 3.44-3.36 (m, 2H), 3.31-3.25 (m, 2H); ESI-MS m/z: 计算值 562.1, 实测值 561.1 [M-H] +。
实施例 42
本发明化合物 42按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 42中相应于通式( I ) 中的 R,取代的 A环的 N-环己基哌 嗪 -1 - 甲酰胺。得到白色固体。 1H NMR (600 MHz, DMSO-^): δ 9.27 (s, 1H), 8.44 (d, J = 7.6 Hz, 1H), 8.14 (d, J= 7.4 Hz, 1H), 7.90 (m, 2H), 7.30 (m, 2H), 7.17 (m, 1H), 5.35 (s, 1H), 4.29 (m, 1H), 3.33 (m, 8H), 1.98 (m, 4H), 1.69 (s, 3H), 1.59 (s, 3H); ESI-MS m/z: 计算值 493.5, 实测值 516.1 [M+Na] +
实施例 43
本发明化合物 43按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 43中相应于通式(I ) 中的 R,取代的 A环的 4 - 乙酰基 哌嗪。 得到白色固体。 1H NMR (600 MHz, DMSO- 6): δ 9.71 (s, 1H), 8.44 (d, 1H, J= 7.74 Hz), 8.13 (d, 1H, J = 7.80 Hz), 7.93-7.91 (m, 1H), 7.89-7.87 (m, 1H), 7.33-7.31 (m, 2H), 7.19-7.16 (m, 1H), 3.82 - 3.65 (m, 4H), 3.57-3.41 (m, 4H), 2.16 (s, 3H); ESI-MS m/z: 计算 值 410.1, 实测值 433.1 [M+Na] +。
实施例 44
本发明化合物 44按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 44中相应于通式( I ) 中的 R,取代的 A环的 4- (噻唑 -4 - 羰基)哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.57 (s, 1H), 8.81-8.79 (m, 1H), 8.44 (d, J = 7.68 Hz, 1H), 8.14 (d, J = 7.92 Hz, 1H), 8.09 (brs, 1H), 7.93-7.91 (m, 1H), 7.89-7.86 (m, 1H), 7.35-7.31 (m, 2H), 7.18 (brs, 1H), 4.08-4.03 (m, 2H), 3.91-3.83 (m, 4H), 3.52 (brs, 2H); ESI-MS m/z: 计算值 479.11, 实测值 480.24 [M+H] +。
实施例 45
本发明化合物 45按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌咯, 首先制备化合物 45中相应于通式( I ) 中的 R,取代的 A环的 4- ( 2-苄氧 羰基-哌啶)羰基哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.50 (s, 1H), 8.44 (d, J = 7.86 Hz, 1H), 8.15-8.13 (m, 1H), 7.93-7.91 (m, 1H), 7.89-7.87 (m, 1H), 7.36-7.31 (m, 7H), 7.20-7.18 (m, 1H), 5.15 (s, 2H), 4.22-4.14 (m, 2H), 3.95-3.39 (m, 9H), 2.99-2.77 (m, 2H), 1.91-1.87 (m, 1H), 1.77-1.74 (m, 2H), 1.52-1.42 (m, 1H); ESI-MS m/z: 计算值 613.13, 实测值 636.29 [M+Na] + 。
实施例 46
本发明化合物 46按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌。秦, 首先制备化合物 46中相应于通式( I ) 中的 R'取代的 A环的 4 - ( 3-苄氧 羰基 -哌啶 )羰基哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 10.72 (s, IH), 8.42 (d, J = 7.26 Hz, 1H), 8.12 (d, J = 7.44 Hz, IH), 7.91-7.87 (m, IH), 7.87-7.84 (m, IH), 7.30 (brs, 6H), 7.16-7.14 (m, 2H), 5.10 (s, 2H), 3.99-3.65 (m, 4H), 3.50-3.33 (m, 6H), 1.96 (brs, 1H), 1.88-1.63 (m, 5H), 1.42 (brs, IH); ESI-MS m/z: 计算值 613.13, 实测值 636.48 [M+Na] +
实施例 47
本发明化合物 47按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌咯, 首先制备化合物 47中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4 - 氧 代环己烷羰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.50 (s, IH), 8.42 (d, J= 7.74 Hz, IH), 8.12 (d, J = 7.86 Hz, IH), 7.92-7.90 (m, 1H), 7.88-7.85 (m, 1H), 7.32-7.31 (m, 2H), 7.18-7.15 (m, 1H), 3.84-3.62 (m, 6H), 3.50-3.43 (m, 2H), 2.97-2.86 (m, 1H), 2.53 (brs, 2H), 2.37-2.25 (m, 2H), 2.04 (brs, 4H); ESI-MS m/z: 计算值 492.18, 实测值 493.28 [M+H] +
实施例 48
本发明化合物 48按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 48中相应于通式( I )中的 R'取代的 A环的 4 - ( 4 -氰 基苯曱酰基)哌嗪。 得到白色固体。 1H NMR (600 MHz, DMSO- 6): δ 11.98 (s, IH), 8.28 (d, IH, J = 7.80 Hz), 8.12 (brs, IH), 8.03-8.00 (m, 1H), 7.97-7.94 (m, 3H), 7.62 (brs, 2H), 7.49-7.36 (m, 3H), 3.76-3.65 (m, 4H), 3.44-3.37 (m, 4H); ESI-MS m/z: 计算值 497.15, 实 测值 520.12 [M+Na] +
实施例 49
本发明化合物 49按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 49中相应于通式( I )中的 R,取代的 A环的( R ) 4 - ( 2 - 笨基丙酰基)哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.73 (s, IH), 8.41 (d, J = 7.74 Hz, 1H), 8.10-8.09 (m, IH), 7.90-7.88 (m, IH), 7.86-7.84 (m, IH), 7.31-7.27 (m, 3H), 7.23-7.20 (m, 4H), 7.13-7.08 (m, IH), 3.98-3.76 (m, 3H), 3.48-3.40 (m, 4H), 3.22 (brs, 1H), 2.92-2.74 (m, 1H), 1.45-1.44 (m, 3H); ESI-MS m/z: 计算值 500.19, 实测值 523.03 [M+Na] +。
实施例 50
本发明化合物 50按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 50中相应于通式( I ) 中的 R'取代的 A环的 4- (环丙烷 羰基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, DMSO-i/6): 610.43 (s, IH), 8.43 (d, J = 7.86 Hz, IH), 8.13 (d, J= 7.86 Hz, IH), 7.92-7.90 (m, IH), 7.88-7.85 (m, IH), 7.32-7.30 (m, 2H), 7.16 (t, IH, J = 8.67 Hz), 3.85-3.68 (m, 6H), 3.49-3.42 (m, 2H), 1.76-1.69 (m, IH), 1.01 (brs, 2H), 0.81-0.75 (m, 2H); ESI-MS m/z: 计算值 436.1, 实测值 459.1 [M+Na] +。
实施例 51
本发明化合物 51按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 51中相应于通式( I )中的 R,取代的 A环的 1 - (味。秦 -1 -基 )丙烷 -1 ,2 -二酮。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.97 (s, 1H), 8.44 (d, J= 7.62 Hz, IH), 8.13 (d, J= 7.74 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.33-7.32 (m, 2H), 7.20-7.15 (m, 1H), 3.86 (brs, 2H), 3.74 (brs, IH), 3.68 (brs, IH), 3.62 (brs, IH), 3.57 (brs, IH), 3.49 (brs, 2H), 1.66 (s, 3H); SI-MS m/z: 计算值 438.13, 实测值 460.91 [M+Na] +。
实施例 52
本发明化合物 52按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )派嗪, 首先制备化合物 52中相应于通式( I ) 中的 R'取代的 A环的 4 - ( 5-曱基 异恶唑 -3-羰基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.5 l(s, IH), 8.44 (d, J= 7.62 Hz, IH), 8.14 (d, J= 6.90 Hz, 1H), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.34-7.30 (m, 2H), 7.21-7.15 (m, IH), 6.33 (d, J = 4.86 Hz, IH), 4.01-3.82 (m, 6H), 3.51 (brs, 2H), 2.48 (s, 3H); ESI-MS m/z: 计算值 477.14, 实测值 475.82 [M-H] + 。
实施例 53
本发明化合物 53按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 53中相应于通式( I )中的 R,取代的 A环的 4- ( 2,2,2 - 三 氟乙酰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.70 (s, IH), 8.42 (d, IH, J = 7.80 Hz), 8.13 (d, 1H, J = 7.80 Hz), 7.90-7.93 (m, IH), 7.88-7.86 (m, IH), 7.34-7.33 (m, 2H), 7.19-7.17 (m, IH), 3.87-3.66 (m, 6H), 3.52 (brs, 2H); ESI-MS m/z: 计算值 464.11, 实 测值 486.93 [M+Na] + 。
实施例 54
本发明化合物 54按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 薮基 )哌嗪,首先制备化合物 54中相应于通式( I )中的 R,取代的 A环的 4 - ( 3,3,3 - 三 氟丙酰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.18 (s, 1H), 8.43 (d, IH, J = 7.62 Hz), 8.13 (d, 1H, J = 7.62 Hz), 7.93-7.87 (m, 2H), 7.33-7.32 (m, 2H), 7.19-7.16 (m, IH), 3.85-3.71 (m, 4H), 3.59-3.45 (m, 4H), 3.33-3.23 (m, 2H); ESI-MS m/z: 计算值 478.13 , 实测值 500.96 [M+Na] + 。
实施例 55
本发明化合物 55按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 55中相应于通式(I ) 中的 R,取代的 A环的 4 -新戊酰 基哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 10.01 (s, 1H), 8.43 (d, J= 7.86 Hz, 1H), 8.13 (d, J = 7.74 Hz, 1H), 7.93-7.90 (m, 1H), 7.89-7.86 (m, 1H), 7.32-7.28 (m, 2H), 7.18-7.14 (m, 1H), 3.79-3.75 (m, 4H), 3.68-3.67 (m, 2H), 3.42 (brs, 2H), 1.29 (s, 9H); ESI— MS m/z: 计算值 452.19, 实测值 475.05 [ +Na]
实施例 56
本发明化合物 56按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 56中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 2 - 曱 氧基乙酰基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.85 (s, 1H), 8.44 (d, J = 7.62 Hz, 1H), 8.13 (d, J = 7.86 Hz, 1H), 7.93-7.92 (m, 1H), 7.89-7.92 (m, 1H), 7.32-7.31 (m, 2H), 7.18-7.16 (m, 1H), 4.12 (d, 2H, J= 26.4 Hz), 3.81-3.57 (m, 6H), 3.44-3.40 (m, 5H); ESI- MS m/z: 计算值 440.15, 实测值 462.85 [M+Na] +
实施例 57
本发明化合物 57按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 57中相应于通式( I )中的 R,取代的 A环的 4 - ( 2-氰基 苯甲酰基 )哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.61 (s, 1H), 8.44 (d, J= 7.20 Hz, 1H), 8.14 (brs, 1H), 7.93-7.90 (m, 1H), 7.89-7.87 (m, 1H), 7.76-7.67 (m, 2H), 7.57-7.48 (m, 2H), 7.33-7.31 (m, 2H), 7.22-7.11 (m, 1H), 3.96-3.88 (m, 4H), 3.58-3.36 (m, 4H); ESI— MS m/z: 计算值 497.15 , 实测值 520.00 [M+Na] +
实施例 58
本发明化合物 58按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 58中相应于通式( I ) 中的 R'取代的 A环的 N-异丙基哌 嗪- 甲酰胺。 得到白色固体。 Ή NMR (600 MHz, DMSO- ): δ 11.96 (s, 1H), 8.26 (d, J = 7.80 Hz, 1H),8.10 (d, J = 7.80 Hz, 1H), 8.01-7.99 (m, 1H), 7.96-7.93 (m, 1H), 7.45-7.43 (m, 1H), 7.41-7.36 (m, 2H), 6.22 (d, J = 7.38 Hz, 1H), 3.75-3.70 (m, 1H), 3.58 (brs, 2H), 3.52-3.34 (m, 2H), 3.28-3.26 (m, 2H), 3.214-3.207 (m, 2H), 1.03 (d, J = 6.54 Hz, 6H); ESI— MS m z: 计算值 453.18, 实测值 476.12 [M+Na] +。
实施例 59
本发明化合物 59按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 59中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 2 - 乙 氧基乙酰基 )哌11秦。得到白色固体。 1H NMR (600 MHz, CDC13) δ 10.38 (s, 1H), 8.43 (d, J = 7.74 Hz, 1H), 8.12 (d, J = 7.92 Hz, 1H), 7.92-7.89 (m, 1H), 7.88-7.85 (m, 1H), 7.32-7.30 (m, 2H), 7.17-7.14 (m, 1H), 4.17-4.13 (m, 2H), 3.81 (brs, 2H), 3.71-3.53 (m, 6H), 3.48-3.39 (brs, 2H), 1.24-1.19 (m, 3H). ESI-MS /z:计算值 454.17, 实测值 453·18[Μ-Η] +。
实施例 60
本发明化合物 60按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 60中相应于通式(I ) 中的 R,取代的 Α环的 4 - 异烟酰 基哌嗪。得到白色固体。 Ή NMR (600 MHz, DMSO- 6): δ 9.25 (s, 1H), 8.77-8.71 (m, 2H), 8.45 (d, J= 6.5 Hz, 1H), 8.14-8.13 (m, 1H), 7.94-7.92 (m, 1H), 7.90-7.88 (m, 1H), 7.35-7.30 (m, 4H), 7.21-7.11 (m, 1H), 3.93-3.75 (m, 4H), 3.56-3.39 (m, 4H); ESI-MS m/z: 计算值 473.1, 实测值 474.02 [M+H] +。
实施例 61
本发明化合物 61按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 61中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 2 - 乙 基丁酰基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13) δ 9.45 (s, 1H), 8.44 (d, J = 7.98 Hz, 1H), 8.14 (d, J= 7.68 Hz, 1H), 7.94-7.91 (m, 1H), 7.90-7.87 (m, 1H), 7.33-7.31 (m, 2H), 7.19-7.17 (m, 1H), 3.80-3.60 (m, 6H), 3.44-3.41 (m, 2H), 2.55-2.45 (m, 1H), 1.70-1.67 (m, 2H), 1.51-1.50 (m, 2H), 0.90-0.87 (m, 6H). ESI-MS m/z:计算值 466.20, 实测值 465.23 [M-H] +。
实施例 62
本发明化合物 62按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 62中相应于通式( I )中的 R'取代的 A环的 4 - ( 4, 4 - 二氟哌啶 -1 -羰基)哌嗪。得到白色固体。 1H NMR(600 MHz, CDC13): δ 10.61 (s, 1Η), 8.41 (d, J = 7.74 Hz, 1H), 8.11 (d, J = 7.80 Hz, 1H), 7.91-7.88 (m, 1H), 7.86-7.84 (m, 1H), 7.29-7.28 (m, 2H), 7.15-7.10 (m, 1H), 3.84-3.74 (m, 2H), 3.43 (brs, 2H), 3.38-3.35 (m, 6H), 3.30 (brs, 2H), 1.99-1.94 (m, 4H); ESI-MS m/z: 计算值 515.1, 实测值 537.95 [M+Na] + 。
实施例 63
本发明化合物 63按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 叛基 )哌嗪, 首先制备化合物 632中相应于通式( I ) 中的 R'取代的 A环的 4- (噻吩 -2 -羰基 )哌嗪。得到白色固体。 1H NMR(600 MHz, CDC13): δ 9.58 (s, 1H), 8.44 (d, J= 7.92 Hz, 1H), 8.13 (d, J= 7.74 Hz, 1H), 7.93-7.91 (m, 1H), 7.89-7.87 (m, 1H), 7.49-7.48 (m, 1H), 7.34-7.31 (m, 3H), 7.18 (t, IH, J= 8.70 Hz), 7.07-7.06 (m, IH), 3.87 (brs, 3H), 3.80-3.79 (m, 3H), 3.50 (brs, 2H); ESI-MS m/z: 计算值 478.1, 实测值 477 .2[M-H] +
实施例 64
本发明化合物 64按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 64中相应于通式(I )中的 R,取代的 A环的 4- ( 3 - 羟丙 基 )哌嗪。 得到白色固体。 Ή NMR(600 MHz, CDC13): δ 9.50 (s, IH), 8.44 (d, J= 7.74 Hz, IH), 8.14 (d, J = 7.62 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, 1H), 7.30-7.29 (m, 2H),
7.17- 7.14 (m, IH), 3.82 (t, 2H, J = 5.22 Hz), 3.45 (brs, 2H), 2.69-2.66 (m, 6H), 2.56 (brs, 2H), 1.78-1.75 (m, 2H); ESI-MS m/z: 计算值 426.1, 实测值 427.0 [M+H] + 。
实施例 65
本发明化合物 65按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)旅嗪, 首先制备化合物 65中相应于通式( I )中的 R'取代的 A环的 4-甲腈哌啶。 得到白色固体。 1H NM (600 MHz, CDC13): δ 9.28 (s, IH), 8.45 (d, J= 7.68 Hz, IH), 8.14 (d, J= 7.92 Hz, IH), 7.93-7.91 (m, IH), 7.90-7.87 (m, IH), 7.32-7.30 (m, 2H), 7.19-7.16 (m, IH), 3.94-3.82 (m, 2H), 3.59 (brs, IH), 3.40 (brs, IH), 2.95 (brs, IH), 2.06-1.88 (m, 4H); ESI-MS m/z: 计算值 393.1, 实测值 394.1 [M+H] +
实施例 66
本发明化合物 66按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 66中相应于通式(I ) 中的 R,取代的 A环的 4 - 异丁酰 基哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.15 (s, IH), 8.43 (d, J= 7.74 Hz, 1H), 8.13 (d, J = 7.92 Hz, IH), 7.93-7.90 (m, IH), 7.89-7.86 (m, IH), 7.32-7.31 (m, 2H),
7.18- 7.15 (m, IH), 3.81-3.56 (m, 6H), 3.46-3.41 (m, 2H), 2.82-2.74 (m, IH), 1.14-1.13 (m, 6H); ESI-MS m/z: 计算值 438.1 , 实测值 461.07 [M+Na] +
实施例 67
本发明化合物 67按照方案 4制备, 合成方法参考实施例 7 , 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 67中相应于通式(I ) 中的 R,取代的 A环的 N, N-二甲 基哌嗪 - 1 - 曱酰胺。 得到白色固体。 1H NMR (600 MHz, CDCI3): δ 9.87 (s, IH), 8.44 (d, J = 7.80 Hz, IH), 8.13 (d, J = 7.74 Hz, IH), 7.92-7.90 (m, IH), 7.88-7.86 (m, IH), 7.31-7.28 (m, 2H), 7.17-7.14 (m, IH), 3.80 (brs, 2H), 3.43 (brs, 2H), 3.33-3.31 (m, 2H), 3.26 (m, 2H), 2.85 (s, 6H); ESI-MS m/z: 计算值 439.1, 实测值 439.95 [M+H] +
实施例 68
本发明化合物 68按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 68中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 3-三 氟曱基 -4曱基-苯甲酰基)哌嗪。得到白色固体。1 H NMR (600 MHz, CDC13): δ 9.60 (s, IH), 8.44 (d, J = 7.86 Hz, IH), 8.13 (d, J = 7.62 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, 1H), 7.69 (s, IH), 7.48 (d, J= 7.56 Hz, IH), 7.37-7.35 (m, IH), 7.32-7.30 (m, 2H), 7.17 (brs, IH), 3.81-3.48 (m, 8H), 2.52 (s, 3H); ESI-MS m/z: 计算值 554.1, 实测值 576.98 [M+Na] +。
实施例 69
本发明化合物 69按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 69中相应于通式(I ) 中的 R,取代的 A环的 4 - 丙烯酰 基哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 10.25 (s, IH), 8.43 (d, J= 7.80 Hz, IH), 8.13 (d, J = 7.98 Hz, IH), 7.93-7.90 (m, IH), 7.89-7.86 (m, IH), 7.33-7.31 (m, 2H), 7.18-7.15 (m, IH), 6.56 (brs, IH), 6.32 (d, 1H, J = 16.86 Hz), 5.75 (brs, IH), 3.81-3.62 (m, 6H), 3.46 (brs, 2H); ESI-MS m/z: 计算值 422.1, 实测值 444.97 [M+Na] + 。
实施例 70
本发明化合物 70按照方案 70制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷復基 )哌嗪, 首先制备化合物 2中相应于通式( I )中的 R,取代的 A环的 4 - ( 2 - 羟 基丙酰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.09 (s, IH), 8.43 (d, J= 7.68 Hz, IH), 8.13 (d, J= 7.62 Hz, IH), 7.92-7.90 (m, IH), 7.88-7.86 (m, IH), 7.31 (brs, 2H), 7.17 (t, 1H, J= 8.58 Hz), 5.48-5.43 (m, IH), 4.36-3.35 (m, IH), 3.96-3.78 (m, 2H), 3.66-3.44 (m, 6H), 1.60-1.57 (m, 3H); ESI-MS m/z: 计算值 440.0, 实测值 439.0 [M-H] +。
实施例 71
本发明化合物 71按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 71 中相应于通式(I ) 中的 R,取代的 A环的 4- ( 4, 4- 二氟哌啶 -1 -羰基 )哌啶。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.40 (s, 1H), 8.44 (d, J = 7.74 Hz, IH), 8.14 (d, J = 7.98 Hz, 1H), 7.94-7.91 (m, IH), 7.89-7.86 (m, IH), 7.32-7.28 (m, 2H), 7.16 (t, 1H, J= 8.7 Hz), 4.72-4.70 (m, IH), 3.74 (brs, 3H), 3.62 (brs, 2H), 3.17 (brs, IH), 2.97 (brs, IH), 2.80 (brs, IH), 2.00 (m, 4H), 1.84 (brs, 3H), 1.71-1.69 (m, IH); ESI-MS m/z: 计算值 514.1, 实测值 515.1 [M+H] +
实施例 72
本发明化合物 72按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 72中相应于通式( I )中的 R'取代的 A环的 4 - (哌1 ^-1 -羰基 )哌啶。得到白色固体。 1H NMR(600 MHz, CDC13): δ 9.51 (s, IH), 8.44 (d, J= 7.80 Hz, 1H), 8.14 (d, J= 7.98 Hz, IH), 7.93-7.90 (m, IH), 7.88-7.86 (m, IH), 7.32-7.28 (m, 2H), 7.15 (t, 1H, J = 8.70 Hz), 4.71-4.69 (m, 1H), 3.75-3.72 (m, 1H), 3.55 (brs, 2H), 3.45 (brs, 2H), 3.15 (brs, 1H), 2.97-2.93 (m, 1H), 2.78 (brs, 1H), 1.83 (brs, 2H), 1.70-1.66 (m, 4H), 1.58-1.54 (m, 4H); ESI-MS m/z: 计算值 478.2, 实测值 501.1 [M+Na] +。
实施例 73
本发明化合物 73按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 73中相应于通式( I ) 中的 R,取代的 A环的 N-苯基哌啶 -4 - 曱酰胺。 得到白色固体。 1H NMR (600 MHz,CDCl3): S 9.82 (s, 1H), 8.43 (d,J= 7.86 Hz, 1H), 8.13 (d, J = 7.86 Hz, 1H), 7.92-7.90 (m, 1H), 7.87-7.85 (m, 1H), 7.54 (s, 1H), 7.51 (d, 2H, J = 7.38 Hz), 7.31-7.29 (m, 3H), 7.15 (t, 1H, J = 8.70 Hz), 7.10 (t, 1H, J = 7.23 Hz), 4.74-4.72 (m, 1H), 3.78-3.76 (m, 1H), 3.16 (brs, 1H), 2.97-2.93 (m, 1H), 2.56-2.51 (m, 1H), 2.05-2.03 (m, 1H), 1.95-1.84 (m, 3H); ESI-MS m/z: 计算值 486.1, 实测值 485.1 [M-H] +。
实施例 74
本发明化合物 74按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 74中相应于通式( I ) 中的 R,取代的 A环的 N-乙基哌啶 -4 - 甲酰胺。 得到白色固体。 1H NMR (600 MHz, CDC13): 5 9.60 (s, 1H), 8.44 (d, J= 7.86 Hz, 1H), 8.13 (d, J= 7.86 Hz, 1H), 7.92-7.90 (m, 1H), 7.88-7.86 (m, 1H), 7.29-7.28 (m, 2H), 7.15 (t, 1H, J= 8.70 Hz), 5.54 (brs, 1H), 4.72-4.70 (m, 1H), 3.74-3.72 (m, 1H), 3.31-3.27 (m, 2H), 3.12 (brs, 1H), 2.92-2.88 (m, 1H), 2.34-2.31 (m, 1H), 1.96-1.94 (m, 1H), 1.85-1.83 (m, 1H), 1.77-1.76 (m, 2H), 1.13 (t, J= 7.23 Hz, 3H); ESI-MS m/z: 计算值 438.1, 实测值 461.1 [M+Na] +。
实施例 75
本发明化合物 75按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 75中相应于通式( I )中的 R,取代的 A环的 4 - ( 4 - (二 曱基氨基 )苯曱酰基 )哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.50 (s, 1H), 8.44 (d, J = 7.68 Hz, 1H), 8.13 (d, J = 7.62 Hz, 1H), 7.93-7.91 (m, 1H), 7.89-7.86 (m, 1H), 7.37 (d, J = 8.76 Hz, 2H), 7.33-7.29 (m, 2H), 7.17 (t, 1H, J= 8.61 Hz), 6.68 (d, J = 8.64 Hz, 2H), 3.82-3.69 (m, 6H), 3.45 (brs, 2H), 3.01 (s, 6H); ESI-MS m/z: 计算值 515.1, 实测值 538.1 [M+Na] +
实施例 76
本发明化合^ 76按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 76中相应于通式( I )中的 R'取代的 A环的 4 - ( 3- (二 曱基氨基 )苯曱酰基 )哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.39 (s, 1H), 8.44 (d, J = 7.62 Hz, IH), 8.13 (d, J = 7.68 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.36-7.29 (m, 3H), 7.18 (brs, IH), 6.76-6.76 (m, 2H), 6.70-6.69 (m, IH), 3.79 (brs, 4H), 3.49 (brs, 4H), 2.98 (s, 6H); ESI-MS m/z: 计算值 515.1, 实测值 538.1 [M+Na] +。
实施例 77
本发明化合物 77按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 77中相应于通式(I ) 中的 R,取代的 A环的 4 -叔丁基 哌嗪。得到白色固体。 Ή NMR (600 MHz, CDC13): δ 8.53-8.48 (m, 1H), 8.13-8.12 (m, IH), 8.08 (d, J = 6.90 Hz, IH), 7.99-7.97 (m, IH), 7.91-7.87 (m, 3H), 3.84-3.79 (m, 2H), 3.68-3.62 (m, 2H), 3.50-3.41 (m, 4H), 1.59 (s, 9H); ESI-MS m/z: 计算值 424.1, 实测值 447.2 [M+Na] +
实施例 78
本发明化合物 78按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)派。秦, 首先制备化合物 78中相应于通式( I )中的 R,取代的 A环的 4 - (呋喃 -2 -羰基 )哌嗪。得到白色固体。 1 H NMR (600 MHz, CDC13): δ 9.64 (s, IH), 8.44 (d, J= 7.74 Hz, IH), 8.14 (d, J = 7.92 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.49 (s IH), 7.34-7.31 (m, 2H), 7.18 (t, 1H, J = 8.64 Hz), 7.07 (d, J = 3.36 Hz, 1H), 6.51-6,50 (m, 1H), 3.92-3.84 (m, 6H), 3.51 (br, 2H); ESI-MS m/z: 计算值 462.13, 实测值 463.08 [M+H] +。
实施例 79
本发明化合物 79按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)派嗪, 首先制备化合物 79中相应于通式(I ) 中的 R'取代的 A环的 4-异丙基哌 嗪。 得到白色固体 Ή NMR (600 MHz, CDC13): δ 9.07 (s, IH), 8.44 (d, J = 7.80 Hz, IH), 8.14 (d, J = 7.56 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.30-7.28 (m, 2H), 7.15 (t, IH, J= 8.52 Hz), 3.81 (brs, 2H), 3.41 (brs, 2H), 2.75 (brs, IH), 2.60 (brs, 2H), 2.50 (brs, 2H), 1.06 (d, 6H, J= 6.00 Hz); ESI-MS m/z: 计算值 410.18, 实测值 411.14 [M+H] +。
实施例 80
本发明化合物 80按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 80中相应于通式( I ) 中的 R'取代的 A环的 4-烯丙基哌 嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.21 (s, 1H), 8.44 (d, J= 7.86 Hz, 1H), 8.14 (d, J = 7.98 Hz, IH), 7.93-7.91 (m, IH), 7.89-7.87 (m, IH), 7.29-7.28 (m, 2H), 7.17-7.14 (m, IH), 5.89-5.81 (m, IH), 5.22-5.20 (m, 2H), 3.82 (brs, 2H), 3.42 (brs, 2H), 3.04 (d, 2H, J = 6.36 Hz), 2.54-2.53 (m, 2H), 2.44 (brs, 2H); ESI-MS m/z: 计算值 408.16, 实测 值 409.15 [M+H] +。 实施例 81
本发明化合物 81按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 81中相应于通式( I ) 中的 R,取代的 A环的 4 - 3-氰基苯 曱酰基哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.72 (s, IH), 8.45 (d, J= 7.86 Hz, IH), 8.15 (d, J= 7.86 Hz, IH), 7.95-7.93 (m, 1H), 7.91-7.88 (m, IH), 7.75-7.68 (m, 2H), 7.66 (d, J= 7.68 Hz, IH), 7.58-7.56 (m, 1H), 7.34-7.32 (m, 2H), 7.21-7.14 (m, IH), 3.83 (brs, 4H), 3.49 (brs, 4H); ESI-MS m/z: 计算值 497.15, 实测值 496.22 [M-H] +。
实施例 82
本发明化合物 82按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 82中相应于通式( I )中的 R,取代的 A环的 4- ( 1H-吡唑 -1 -羰基)哌嗪。得到白色固体。 1H NMR (600 MHz, DMSO- 6): δ 9.21 (s, IH), 8.45 (d, J = 6.72 Hz, IH), 8.16-8.13 (m, 2H), 7.94-7.91 (m, IH), 7.90-7.87 (m, IH), 7.65 (brs, IH), 7.35-7.28 (m, 2H), 7.20-7.17 (m, IH), 6.39-6.38 (m, IH), 4.03-3.94 (m, 4H), 3.56 (brs, 2H); ESI-MS m/z: 计算值 462.1, 实测值 485.1 [M+Na] +。
实施例 83
本发明化合物 83按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 叛基 )哌。秦, 首先制备化合物 83中相应于通式( I ) 中的 R'取代的 A环的 4- (吗啉 -4 - 羰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.52 (s, IH), 8.42 (d, J= 7.74 Hz, IH), 8.11 (d, J= 7.80 Hz, IH), 7.91-7.89 (m, IH), 7.87-7.84 (m, 1H), 7.29-7.28 (m, 2H), 7.15-7.12 (m, IH), 3.78 (brs, 2H), 3.67-3.65 (m, 4H), 3.43 (brs, 2H), 3.35-3.34 (m, 2H), 3.28-3.27 (m, 6H); ESI-MS m/z: 计算值 481.18, 实测值 480.22 [M+H] +。
实施例 84
本发明化合物 84按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 84中相应于通式( I ) 中的 R,取代的 A环的 4- (吡咯烷 -1 -羰基 )哌嗪。得到白色固体。 Ή NMR (600 MHz, CDC13): δ 10.14 (s, IH), 8.43 (d, J = 7.86 Hz, IH), 8.12 (d, J= 7.86 Hz, IH), 7.92-7.89 (m, IH), 7.87-7.85 (m, IH), 7.30-7.29 (m, 2H), 7.16-7.14 (m, IH), 3.80 (brs, 2H), 3.43-3.41 (m, 2H), 340-3.37 (m, 6H), 3.31-3.29 (m, 2H), 1.83 (brs, 4H); ESI-MS m/z: 计算值 465.18, 实测值 464.30 [M-H] +
实施例 85
本发明化合物 85按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 85中相应于通式( I )中的 R,取代的 A环的 4 - (哌啶 -1 -羰基)哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.19 (s, IH), 8.44 (d, J= 7.74 Hz, IH), 8.13 (d, J = 7.62 Hz, IH), 7.94-7.91 (m, IH), 7.89-7.87 (m, IH), 7.31-7.28 (m, 2H),
7.18- 7.15 (m, IH), 3.82-3.79 (m, 2H), 3.45-3.40 (m, 2H), 3.33-3.31 (m, 2H), 3.26-3.25 (m, 2H), 3.24-3.22 (m, 4H), 1.62-1.56 (m, 6H); ESI-MS m/z: 计算值 479.20, 实测值 478.30 [M-H] +o
实旄例 86
本发明化合物 86按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌咯, 首先制备化合物 86中相应于通式( I )中的 R,取代的 A环的 N- ( 3 - (三 氟曱基)苯基)哌。秦-1 - 曱酰胺。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.52 (s, IH), 8.43 (d, J = 7.8 Ηζ,ΙΗ), 8.13 (d, J = 7.8 Hz, IH), 7.92-7.86 (m, 2H), 7.64-7.62 (m, IH), 7.57-7.56 (m, 2H), 7.42-7.38 (m, 2H), 7.42-7.38 (m, 2H), 3.62-3.59 (m, 4H), 3.53-3.48 (m, 4H); ESI-MS m/z: 计算值 555.5, 实测值 554.0[M-H] +。
实施例 87
本发明化合物 87按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 87中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 3 -硝 基苯曱酰基 )哌嗪。得到白色固体。 Ή NMR (600 MHz, DMSO-J6): δ 11.96 (m, IH), 8.29 (d, 1H,J= 6.96 Hz), 8.27-8.24 (m, 2H), 8.10 (brs, 1H), 8.01-7.99 (m, 1H), 7.96-7.93 (m 1H), 7.87-7.86 (m, IH), 7.74 (brs, IH), 7.43 (brs, 3H), 3.75-3.65 (m, 4H), 3.41-3.32 (m, 4H); ESI-MS m/z: 计算值 517.14, 实测值 515.99 [M-H] +。
实施例 88
本发明化合物 88按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌。秦, 首先制备化合物 88中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 2 -硝 基苯曱酰基 )哌嗪。得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.30 (m, IH), 8.46-8.43 (m, 1H), 8.23-8.18 (m, IH), 8.16-8.11 (m, IH), 7.94-7.88 (m, 2H), 7.77-7.71 (m, IH), 7.64-7.57 (m, IH), 7.42-7.40 (m, IH), 7.36-7.34 (m IH), 7.31-7.30 (m, IH), 7.23-7.20 (m, IH), 4.24-3.68 (m, 4H), 3.59-3.28 (m, 4H); ESI-MS m/z: 计算值 517.14, 实测值 516.10 [M-H] +。
实施例 89
本发明化合物 89按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 89中相应于通式( I )中的 R,取代的 A环的 4 - 丁 -2 - 烯 酰基哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13) δ 9.59 (s, IH), 8.44 (d, J= 7.68 Hz, IH), 8.14 (d, J = 7.80 Hz, IH), 7.94-7.91 (m, 1H), 7.89-7.87 (m, IH), 7.33-7.31 (m, 2H),
7.19- 7.16 (m, IH), 6.96-6.90 (m, IH), 6.26 (brs, IH), 3.82-3.60 (m, 6H), 3.44 (brs, 2H), 1.90 (brs, 3H); ESI-MS m/z: 计算值 436.15, 实测值 435.06 [M-H] +。
实施例 90
本发明化合物 90按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 90中相应于通式(I ) 中的 R,取代的 A环的 4 - 曱基哌 嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.16 (s, 1H), 8.43 (d, 1H, J = 7.74 Hz), 8.13 (d, 1H, J = 7.80 Hz), 7.92-7.90 (m, 1H), 7.88-7.85 (m, 1H), 7.29-7.27 (m, 2H), 7.16-7.13 (m, 1H), 3.84 (brs, 2H), 3.45 (brs, 2H), 2.53 (brs, 2H), 2.45 (brs, 2H), 2.35 (s, 3H); ESI-MS m/z: 计算值 382.14, 实测值 381.17 [M-H] +。
实施例 91
本发明化合物 91按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 91中相应于通式( I )中的 R,取代的 A环的 4 - (恶唑 -4 -羰基)哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.73 (s, 1H), 8.44 (d, J= 7.74 Hz, 1H), 8.26 (s, 1H), 8.14 (d, J = 7.80 Hz, 1H), 7.93-7.86 (m, 3H), 7.35-7.30 (m, 2H), 7.19-7.16 (m, 1H), 4.23-4.09 (m, 2H), 3.89-3.79 (m, 4H), 3.51 (brs, 2H); ESI-MS m/z: 计算 值 463.13, 实测值 463.92 [M+H] +。
实施例 92
本发明化合物 92按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌嗪, 首先制备化合物 92中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 2 - 羟 乙基 )哌嗪。得到白色固体。 Ή NMR (600 MHz, CDCI3): δ 10.14 (s, 1H), 8.43 (d, J= 7.80 Hz, 1H), 8.13 (d, J= 7.92 Hz, 1H), 7.93-7.92 (m, 1H), 7.89-7.86 (m, 1H), 7.29-7.26 (m, 2H), 7.16-7.14 (m, 1H), 3.82 (brs, 2H), 3.67-3.65 (m, 2H), 3.43 (brs, 2H), 2.63-2.59 (m, 4H), 2.53 (brs, 2H); ESI-MS m/z: 计算值 412.15, 实测值 411.30 [M-H] +。
实施例 93
本发明化合物 93按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哝嗪,首先制备化合物 93中相应于通式( I )中的 R,取代的 A环的 4 - ( 1H-1,2,4 - 三唑 -1 -羰基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, DMSO-i/6): δ 9.25 (s, 1H), 8.46-8.42 (m, 2H), 8.14 (d, J = 7.86 Hz, 1H), 7.94-7.92 (m, 1H), 7.90-7.88 (m, 1H), 7.46-7.44 (m, 1H), 7.38-7.35 (m, 2H), 7.22-7.19 (m, 1H), 4.02-3.99 (m, 4H), 3.73-3.59 (m, 4H); ESI-MS m/z: 计算值 463.1 , 实测值 486.2 [M+Na] +
实施例 94
本发明化合物 94按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 94中相应于通式( I ) 中的 R,取代的 A环的 4 - (四氢 呋喃 -2 - 羰基)哌嗪。得到白色固体。 'Η NMR (600 MHz, DMSO-^): δ 9.22 (s, IH), 8.45 (d, J = 7.56 Hz, 1H), 8.14 (d, J = 7.44 Hz, IH), 7.94-7.91 (m, IH), 7.90-7.87 (m, IH), 7.32-7.31 (m, 2H), 7.19-7.16 (m, IH), 4.62-4.55 (m, IH), 4.00-3.81 (m, 5H), 3.72-3.50 (m, 5H), 2.39-2.38 (m, IH), 2.07-2.00 (m, 2H), 1.94-1.91 (m, IH); ESI-MS m/z: 计算值 466.1, 实测值 488.9 [M+Na]+
实施例 95
本发明化合物 95按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 95中相应于通式( I ) 中的 R,取代的 A环的 4 - (四氢 呋喃 -2 -羰基)哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.00 (s, IH), 8.44 (d, J= 7.80 Hz, IH), 8.13 (d, J = 7.86 Hz, IH), 7.93-7.90 (m, IH), 7.89-7.86 (m, 1H), 7.32-7.31 (m, 2H), 7.18-7.15 (m, IH), 4.62-4.55 (m, IH), 3.97-3.81 (m, 5H), 3.69-3.39 (m, 5H), 2.38-2.34 (m, 1H), 2.06-1.99 (m, 2H), 1.94-1.89 (m, IH); ESI-MS m/z: 计算值 466.17,实测 值 465.27 [M-H] +。
实施例 96
本发明化合物 96按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )派嗪, 首先制备化合物 96中相应于通式( I ) 中的 R,取代的 A环的 N- ( 4,4 - 二 氟环己基 )哌嗪-1 - 甲酰胺。得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.96 (d, J=7.8 Hz, IH), 8.43 (d, J = 7.68 Hz, IH), 8.14 (d, J = 7.80 Hz, IH), 7.99-7.91 (m, 1H), 7.89-7.87 (m, IH), 7.31-7.30 (m, 2H), 7.18-7.15 (m, IH), 4.53-4.48 (m, IH), 3.80 (brs, 2H), 3.44 (brs, 6H), 2.44-2.34 (m, IH), 2.23-2.19 (m, IH), 2.11-1.72 (m, 5H), 1.50-1.42 (m, IH); ESI-MS m/z: 计算值 529.19, 实测值 528.09 [M-H] +。
实施例 97
本发明化合物 97按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 97中相应于通式( I ) 中的 R'取代的 A环的 N-环丙基哌 嗪— 1 曱酰胺。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.14 (s, IH), 8.43 (d, J = 7.74 Hz, IH), 8.13 (d, J= 7.68 Hz, IH), 7.91-7.90 (m, IH), 7.89-7.87 (m, 1H), 7.31-7.30 (m, 2H), 7.17-7.14 (m, IH), 4.93 (brs, IH), 3.78 (brs, 2H), 3.44-3.42 (m, 6H), 0.71-0.69 (m, 2H), 0.45 (brs, 2H); ESI-MS m/z: 计算值 451.17, 实测值 450.00 [M-H] +。
实施例 98
本发明化合物 98按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 98中相应于通式(I ) 中的 R,取代的 A环的 4 - ( 1H- 咪唑 -1 -羰基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.69 (s, IH), 8.40 (d, J = 7.62 Hz, IH), 8.34 (s, 1H), 8.12 (d, J = 7.86 Hz, 1H), 7.93-7.90 (m, IH), 7.88-7.86 (m, 1H), 7.34-7.28 (m, 3H), 7.22-7.21 (m, IH), 7.19-7.16 (m, IH), 3.90 (brs, 2H), 3.74 (brs, 2H), 3.68 (brs, 2H) 3.55 (brs, 2H); ESI-MS m/z: 计算值 462.43, 实测值 461.11 [M-H] +。
实施例 99
本发明化合物 99按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌嗪, 首先制备化合物 99中相应于通式( I ) 中的 R,取代的 A环的 4 - (环己 烷羰基)哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.78 (s, IH), 8.41 (d, J = 7.74 Hz, IH), 8.11 (d, J= 7.80 Hz, 1H), 7.91-7.88 (m, IH), 7.86-7.84 (m, IH), 7.30-7.29 (m, 2H), 7.15-7.12 (m, IH), 3.80-3.54 (m, 6H), 3.44-3.39 (m, 2H), 2.48-2.41 (m, IH), 1.78-1.69 (m, 5H), 1.54-1.48 (m, 2H), 1.29-1.21 (m, 3H); ESI-MS m/z: 计算值 478.52, 实测值 477.22 [M-H] +。
实施例 100
本发明化合物 100按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌嗪, 首先制备化合物 100中相应于通式( I ) 中的 R,取代的 A环的 4 - 丙酰 基哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.69 (s, IH), 8.41 (d, IH, J= 7.80 Hz), 8.11 (d, 1H, J= 7.86 Hz), 7.90 (t, 1H, J= 7.41 Hz), 7.86 (t, IH, J= 7.38 Hz), 7.31-7.29 (m, 2H), 7.15 (t, 1H, J = 8.88 Hz ), 3.79-3.65 (m, 4H), 3.56-3.40 (m, 4H), 2.40-2.37 (m, IH), 2.32-2.31 (m, IH), 1.16-1.11 (m, 3H); ESI-MS m/z: 计算值 424.42, 实测值 423.13 [M-H]+。
实施例 101
本发明化合物 101按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷欺基 )哌嗪, 首先制备化合物 101中相应于通式( I ) 中的 R,取代的 A环的 4 - (环 戊基羰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.14 (s, IH), 8.43 (d, J = 7.80 Hz, IH), 8.13 (d, J= 7.86 Hz, IH), 7.95-7.90 (m, IH), 7.89-7.86 (m, IH), 7.31-7.30 (m, 2H), 7.18-7.15 (m, IH), 3.80-3.57 (m, 6H), 3.44-3.40 (m, 2H), 2.19 (brs, IH), 1.83-1.74 (m, 8H); ESI-MS m/z: 计算值 464.49, 实测值 463.21 [M-H] +。
实施例 102
本发明化合物 102按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)。束。秦, 首先制备化合物 102中相应于通式(I ) 中的 R'取代的 A环的 4 - ( 4- 苄氧羰基 -哌啶)羰基哌 σ秦。得到白色固体。 1H NMR (600 MHz, DMSO-^): δ 12.23 (s, IH), 8.27 (d, J = 7.56 Hz, 1H), 8.06-8.05 (m, IH), 7.88 (t, IH, J = 7.92 Hz), 7.37-7.32 (m, 7H), 7.31-7.28 (m, 1H), 5.05 (s, 2H), 3.99-3.98 (m, 2H), 3.63-3.57 (m, 3H), 3.51-3.44 (m, 5H), 2.88-2.79 (m, 3H), 1.62 (brs, 2H), 1.44-1.37 (m, 2H); ESI-MS m/z: 计算值 647.2, 实测值 646.3 [M-H] +。
实施例 103
本发明化合物 103按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌嗪, 首先制备化合物 103中相应于通式(I ) 中的 R,取代的 A环的 4 - 苯曱 酰基哌嗪。得到白色固体。 1H NMR (600 MHz, OMSO-d6): δ 11.98 (s, IH), 8.28 (d, J= 7.80 Hz, 1H), 8.12 (d, J= 8.22 Hz, IH), 8.03-8.00 (m, IH), 7.98-7.95 (m, IH), 7.51-7.49 (m, 2H), 7.47-7.40 (m, 4H), 7.28 (t, 2H,J= 8.64 Hz), 3.74-3.36 (m, 8H); ESI-MS m/z: 计算值 472.15: 实测值 471 ·27[Μ-Η] +。
实施例 104
本发明化合物 104按照方案 3制备, 合成方法参考实施例 7, 只是代替 Ν- (环丙 烷羰基)哌嗪, 首先制备化合物 104中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4 - 氟苯基)哌嗪。 得到白色固体。 1H NMR (600 MHz, DMSO-^): δ 12.22 (s, IH), 8.27-8.26 (m, IH), 8.06 (d, J = 7.74 Hz, IH), 7.88 (t, 1H, J = 7.95 Hz), 7.37-7.34 (m, 3H), 7.06-7.03 (m, 2H), 6.97-6.94 (m, 2H), 3.76-3.75 (m, 2H), 3.39-3.37 (m, 2H), 3.14-3.12 (m, 2H), 3.05-3.03 (m, 2H); ESI-MS m/z: 计算值 496.0, 实测值 497.0 [M+H] +。
实施例 105
本发明化合物 105按照方案 3制备, 合成方法参考实施例 1 , 只是代替 N- (环丙 烷羰基) 嗪, 首先制备化合物 105中相应于通式(I ) 中的 R,取代的 A环的 4 - ( 4- 苄氧羰基-哌啶)羰基哌嗪。得到白色固体。 1H NMR (600 MHz, DMSO-^): δ 12.06 (s, IH), 8.10-8.08 (m, IH), 7.90-7.89 (m, IH), 7.80-7.77 (m, IH), 7.61 (d, J = 7.50 Hz, 1H), 7.54 (s, IH), 7.49 (t, J = 7.56 Hz, IH), 7.39 (d, J = 7.62 Hz, IH), 7.37-7.28 (m, 5H), 5.38 (s, 2H), 5.06 (s, 2H), 3.40-3.98 (m, 2H), 3.62-3.38 (m, 8H), 2.87 (brs, 3H), 1.61 (brs, 2H), 1.44-1.39 (m, 2H); ESI-MS m/z: 计算值 627.3, 实测值 650.6 [M+Na] +。
实施例 106
本发明化合物 106按照方案 3制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基 )哝嗪, 首先制备化合物 106中相应于通式( I ) 中的 R'取代的 A环的 4 - ( 4 - 硝基苯曱酰基)哌嗪。 得到白色固体。 'Η NMR (600 MHz, DMSO- 6): δ 12.24 (s, IH), 8.27-8.26 (m, 3H), 8.05 (brs, IH), 7.90-7.87 (m, IH), 7.69 (brs, 2H), 7.36 (brs, 3H), 3.72-3.64 (m, 4H), 3.28-3.26 (m, 4H); ESI-MS m/z: 计算值 551.1, 实测值 550.1 [M+H] +。
实施例 107
本发明化合物 107按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基 )味嗪, 首先制备化合物 107中相应于通式( I )中的 R'取代的 A环的 4 - ( 4,4 -二氟环己烷羰基)σ束嗪。得到白色固体。1 H NMR (600 MHz, CDC13): δ 9.71 (s, 1H), 8.44 (d, J = 7.74 Hz, 1H), 8.13 (d, J = 7.80 Hz, IH), 7.94-7.91 (m , IH), 7.90-7.87 (m, IH), 7.33-7.31 (m, 2H), 7.19-7.16 (m, IH), 3.82-3.66 (m, 4H), 3.61-3.42 (m, 4H), 2.61-2.54 (m, IH), 2.23-2.19 (m, 2H), 1.95-1.89 (m, 2H), 1.83-1.72 (m, 4H); ESI-MS m/z: 计算值 514.20, 实测值 537.06 [M+ Na] +。
实施例 108
本发明化合物 108按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌嗪, 首先制备化合物 108中相应于通式(I ) 中的 R,取代的 A环的 4 - ( 4- 苄氧羰基-吡咯烷)羰基哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 9.27 (s, IH), 8.44 (d, J = 8.3 Hz, IH), 8.13 (br, IH), 7.90 (m, 2H), 7.33 (m, 7H), 7.18 (s, IH), 5.15 (m, 2H), 3.84 (br, IH), 3.50 (m, 8H), 2.19 (m, 4H), 1.91 (br, 2H); ESI-MS m/z: 计算值 599.21, 实测值 622.26 [M+ Na] +。
实施例 109
本发明化合物 109按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌嗪, 首先制备化合物 109中相应于通式( I ) 中的 R,取代的 A环的 4 - (吡 咯烷 -2 -羰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 8.44 (s, 1H), 8.15 (d, J = 7.7 Hz, IH), 7.88 (m, 3H), 7.43 (m, 2H), 7.19 (s, IH), 4.06 (m, 2H), 3.74 (m, 2H), 3.53 (m, 8H), 2.63 (brs, IH), 2.26-2.19 (m, 2H); ESI-MS m/z: 计算值 465.18, 实测值 465.98 [M+H] +
实施例 110
本发明化合物 110按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基 )哌嗪, 首先制备化合物 110中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 4 - 氟苯基)哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13) δ 10.20 (s, IH), 8.42 (d, J= 7.86 Hz, IH), 8.14 (d, J= 7.92 Hz, IH), 7.93-7.90 (m, IH), 7.86-7.84 (m, IH), 7.34-7.30 (m, 2H), 7.17 (t, IH, J = 8.67 Hz), 6.97-6.94 (m, 2H), 6.90-6.88 (m, 2H), 3.96 (brs, 2H), 3.58 (brs, 2H), 3.18-3.17 (m, 2H), 3,10 (brs, 2H). ESI-MS m/z:计算值 462.45,实测值 461.16 [M-H] +。
实施例 111
本发明化合物 111按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基 )哌嗪, 首先制备化合物 111中相应于通式( I ) 中的 R,取代的 A环的 4 - (四 氢呋喃 -3 -羰基 )哌嗪。 得到白色固体 1H NMR (600 MHz, CDC13): δ 10.16 (m, IH), 8.43 (d, J = 7.80 Hz, IH), 8.13 (d, J = 7.74 Hz, 1H), 7.93-7.90 (m, IH), 7.89-7.86 (m, IH), i nou/uosldi/-8/-oi IS(Hs £ 96£0Hl6 £ 9s£6£ e9H—-l ,....
+養 XrH Χ orsr£ΗΐzS( SMI3Η1199P88Nw. + ίs z¾-. 。.
Figure imgf000076_0001
本发明化合物 116按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基)哌11秦, 首先制备化合物 116中相应于通式( I )中的 R,取代的 A环的 4 - ( 4 - 氟 苯曱酰基 )哌嗪。得到白色固体。 1H NMR (600 MHz, CDC13): δ 10.21 (s, 1H), 8.42 (d, J= 7.74 Hz, 1H), 8.13 (d, J = 7.74 Hz, 1H), 7.93-7.90 (m, 1H), 7.88-7.86 (m, 1H), 7.44-7.42 (m, 2H), 7.32-7.30 (m, 2H), 7.17-7.09 (m, 3H), 3.81-3.27 (m, 8H); ESI-MS; m/z: 计算值 490.15, 实测值 491.01 [M+H] +。
实施例 117
本发明化合物 117按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基) 嗪, 首先制备化合物 117中相应于通式( I )中的 R,取代的 A环的 4 - ( 4 - 氟 苄基 )哌嗪。 得到白色固体。 1H NMR (600 MHz, CDCI3): δ 9.33 (s, 1H), 8.44 (d, J= 7.92 Hz, 1H), 8.13 (d, J= 7.92 Hz, 1H), 7.93-7.91 (m, 1H), 7.89-7.87 (m, 1H), 7.28-7.27 (m, 4H), 7.16-7.13 (m, 1H), 7.01 (t, 2H, J = 8.58 Hz), 3.81 (brs, 2H), 3.51 (brs, 2H), 3.45-3.38 (m, 2H), 2.52 (brs, 2H), 2.42 (brs, 2H); ESI-MS m/z: 计算值 476.17, 实测值 475.32 [M-H] +。
实施例 118
本发明化合物 118按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基) 嗪, 首先制备化合物 118中相应于通式(I ) 中的 R,取代的 A环的哌嗪。 得到 白色固体。 1H NMR (600MHz, CDC13): δ 9.51 (s, 1H), 8.44 (d, J = 7.8 Hz, 1H), 8.13 (d, J - 7.8 Hz, 1H), 7.91-7.87 (m, 2H), 7.31-7.30 (m, 2H), 7.16-7.15 (m, 1H), 3.77 (brs, 2H), 3.53 (brs, 2H), 3.00 (brs, 2H), 2.95 (brs, 2H); ESI-MS m/z 计算值: 368.13, 实测值: 367.15 [M-H]+。
实施例 119
本发明化合物 119按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷 羰基 )哌。秦, 首先制备化合物 119中相应于通式( I ) 中的 R,取代的 A环的 4 - 吡啶曱 酰基哌嗪。 得到白色固体。 1H NMR (600 MHz, CDC13) δ 9.26 (s, 1H), 8.62-8.56 (m, 1H), 8.45-8.44 (m, 1H), 8.15-8.12 (m, 1H), 7.93-7.91 (m, 1H), 7.89-7.87 (m, 1H), 7.83-7.81 (m, 1H), 7.73 (brs, 1H), 7.39-7.28 (m, 3H), 7.21-7.13 (m, 1H), 3.93-3.72 (m, 6H), 3.55-3.49 (m, 2H); ESI-MS m/z计算值:437.15, 实测值: 437.96 [M+H] +。
实施例 120
本发明化合物 120按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌。秦, 首先制备化合物 120中相应于通式( I ) 中的 R,取代的 A环的 4 - (四 氢呋喃 -2 -羰基 )哌嗪。 得到白色固体。 Ή NMR (600 MHz, CDC13): δ 9.36 (s, 1H), 8.44 (d, J = 7.56 Hz, 1H), 8.14 (d, J = 7.56 Hz, 1H), 7.94-7.91 (m, 1H), 7.90-7.87 (m, 1H), 'ui) WL-9&L '(HI 'ui) 66·/τΐΟ·8 '(HI 08 =f 'Ρ) 0Γ8 '(HI Z&L =Γ 'P) 91 '(HI 's) 96 ll?-(9P-OSVia 'ΖΗΝ009)ΉΙΜΝ Ht ~T ) • I ) - 1/ i V (I)
Figure imgf000078_0001
¾i
、上 i) -N^W¾r 'z, i^^^^-^ ^ '号 ηι ^ ^
°+[BN+W] 88fr 聊^ 'H ^ -zw SIM-IS3'(H3 ' ) £90-590 '(Η ' ) 08Ό-Ζ8Ό '(Η£ 's) 6VZ '(ΗΖ 'ω) 9 Ζ-ίΖ'ί '(Η9 'ui) "'ε-£9·ε '(ζΗ 88·8 'Ηΐ Ί) L '(ΗΖ \YL~9YL '(ΗΙ 'ω) 6·Α-96·Λ '(HI 'ui) 667τΐΟ·8 '(HI Z L =Γ 'Ρ) Π·8 '(Η1 ' 08·乙 =Γ 'Ρ) 9Γ8 '(HI 'S)
Figure imgf000078_0002
'ZHWOO9)¾WMH, 。^囟 ^ °拳 (^¾¾¾>、上 ί (节" ) - 1 ) -
Figure imgf000078_0003
'拳 (
、上 ί ) -N ¥s 'L ^^^^^ ' ίΦ ίζι ^^ ^
tzi
^ r: z/ui SW-ISH '(HZ. '冚) UZ 's-iq) 981 '(H3 ' ) i'Z-lVZ UZ OVi'LVi
'(m 'sjq) '(HI 'sjq) 6ς·ε '(HI 's-iq) 99 ε '(HE '冚) ε//ε 8·ε '(HI '^) 9VL-6VL '(HZ: ' )
\ί· L-τί· L '(Η1 '^) L^L-62'L '(HI '冚) ΐ6·Α-ε6·乙 '(ΖΗ 08· =f'm 'Ρ) ZV '(HI 'ZH 9 L =Γ 'Ρ) 8 '(HI 's) Z 6 δ :(£ΚΧ W 009)丽 N 。 HI r? 。拳 ( f^^ i
-z) - ^ iV ^ ¾t"a Ψ (i) ii^- ^ zz\ ^^ ^^- ' 。( - W¥i- ^ϊί ) -M 、L H ^^ ^^^ '号^ Zl\ ^^ ^
III f'W^ 扉^ 'ίνος ' sw-iS3 -( '冚) ·ο-6ΐ·ο ηζ '^) SSO-6SO '(HI 'sjq) wn Xnz ' ) ίζτ-ίζτ ' ) ζνζ-ίςτ 'm)乙 9·ε-08·ε '(HI ' ) 9YL- VL X '^)
\ί· L-Ζί· L '(HI '" 98'/.-68' '(HI ' ) O&L-i&L '(HI 98'Z. = "P) £Γ8 '(HI 89Ά =f 'P)
-z
Figure imgf000078_0004
Figure imgf000078_0005
Ζ.Γ9917 T^4 :z/ui SWISH - (HI 'ω) 06' '(HZ '^) 002-90 '(HI WZ-^Z '(HS 'm) 9t7-e-0,-£ '(HS 'ω) 38-e-00 '(HI ' ) 9S -^9 '(HI '冚) 9ΓΑ'6Γ '(ίΚ 'ω) \ί' L_li' L
LL
Π9ΐ00/ΖΪ0ΖΝ3/Χ3Λ /.8.0/CT0Z: OAV IH), 7.46-7.41 (m, 2H), 7.38 (t, IH, J= 8.85 Hz), 3.62-3.55 (m, 6H), 3.28-3.25 (m, 2H), 2.14 (s, 6H), 0.84-0.83 (m, 2H), 0.72-0.70 (m, 2H);ESI-MS m/z: 计算值 479.2, 实测值 502.12 [M+Na] +。
实施例 125
本发明化合物 125按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷叛基 )。糸嗪, 首先制备化合物 125中相应于通式( I ) 中的 R'取代的 A环的 4 - ( 1 - 氨基环丙烷羰基)哌嗪。得到白色固体。 1H NMR(600MHZ,
Figure imgf000079_0001
(s, IH), 8.26 (d, J = 7.62 Hz, IH), 8.11 (d, J = 8.10 Hz, 1H), 8.01-7.99 (m, IH), 7.96-7.94 (m, 1H), 7.45-7.36 (m, 3H), 3.64-3.56 (m, 6H), 3.26-3.20 (m, 2H), 0.85-0.81 (m, 2H), 0.76 (brs, IH), 0.63-0.62 (m, IH); ESI-MS w/z:计算值 451.45, 实测值 450.56 [M-H]
实施例 126
本发明化合物 126按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷叛基 )哌嗪, 首先制备化合物 126中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 1 - (二曱基氨基)环丁烷羰基)哌嗪。得到白色固体。 1H NM (600MHz, DMSO-i/6):^ 11.97 (s, IH), 8.26 (d, J = 7.8 Hz, 1H), 8.11-8.09 (m, IH), 8.01-7.98 (m, IH), 7.96-7.93 (m, IH), 7.45-7.40 (m, 2H), 7.37-7.35 (m, IH), 3.62-3.55 (m, 4H), 3.52-3.47 (m, 2H), 3.27-3.19 (m, 2H), 2.36-2.21 (m, 4H), 2.15 (s, 3H), 2.11 (s, 3H), 1.62-1.57 (m, IH), 1.53-1.48 (m, IH); ESI-MS m/z: 计算值 493.2, 实测值 515.98 [M+Na] +
实施例 127
本发明化合物 127按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷狻基 )哌嗪, 首先制备化合物 127中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 1 - (曱基氨基)环丁烷羰基 )哌嗪。得到白色固体。 1H NMR (600MHz, OMSO-d6):S 8.36 (d, J= 7.8 Hz, IH), 8.20 (d, J= 7.8 Hz, 1H), 8.02-8.00 (m, 1H), 7.97-7.94 (m, IH), 7.44-7.41 (m, IH), 7.40-7.38 (m, 2H), 3.82-3.80 (m, 2H), 3.68-3.66 (m, 2H), 3.60-3.58 (m, 2H), 3.46-2.44 (m, 2H), 3.45 (s, 3H), 2.68-2.63 (m, 2H), 2.05-2.02 (m, 3H), 1.73-1.68 (m, IH); ESI-MS m/z: 计算值 493.2, 实测值 515.98 [M+Na] +。 ESI-MS m/z: 计算值 479.20, 实测值 480.25 [M+H] +。
实施例 128
本发明化合物 128按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌咯, 首先制备化合物 128中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 1 - 氨基环丁烷羰基 )哌嗪。 得到白色固体。 Ή NMR (600MHz, OMSO-d6):S 11.97 (s, IH), 8.26 (d, J = 7.8 Hz, IH), 8.11-8.09 (m, IH), 8.01-7.98 (m, IH), 7.96-7.93 (m, IH), 7.45-7.43 (m, 1H), 7.41-7.36 (m, 2H), 3.63-3.52 (m, 3H), 3.51-3.41 (m, 4H), 3.36-3.32 (m, 1H), 2.73-2.54 (m, 1H), 2.23-2.10 (m, 1H), 1.83-1.70 (m, 2H), 1.64-1.57 (m, 1H), 1.54-1.43 (m, 1H); ESI-MS m/z: 计算值 465.2, 实测值 487.77 [M+Na] +。
实施例 129
本发明化合物 129按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙 烷羰基)哌嗪, 首先制备化合物 129中相应于通式( I )中的 R,取代的 A环的 4 - ( 1,2 -二甲基吡咯烷 -2 -羰基)哌嗪。 得到白色固体。 1H NMR (600MHz, DMSO-^)^ 8.36 (d, J= 7.2 Hz, 1H), 8.20 (d, J= 7.2 Hz, 1H), 8.02-7.99 (m, 1H), 7.96-7.94 (m, 1H), 7.44-7.43 (m, 1H), 7.40-7.38 (m, 1H), 7.32-7.29 (m, 1H), 3.82 (brs, 4H), 3.73 (brs, 2H), 3.47 (brs, 2H), 3.42 (brs, 1H), 3.02 (brs, 1H), 2.70 (s, 3H), 2.38-2.34 (m, 1H), 2.23 (brs, 1H), 2.10 (brs, 1H), 2.02-1.97 (m, 1H), 1.52 (s, 3H); ESI-MS m/z: 计算值 493.2, 实测值 491.96 [M-H] +。
实施例 130
本发明化合物 130按照方案 4制备, 合成方法参考实施例 7, 只是代替 N- (环丙烷羰 基 )哌嗪, 首先制备化合物 130中相应于通式( I ) 中的 R,取代的 A环的 4 - ( 2 - 甲 基吡咯烷 -2 -羰基)哌。秦。 得到白色固体。 1H NMR (600MHz, DMSO- 6):^ 8.37 (d, J = 7.2 Hz, 1H), 8.21 (d, J = 7.2 Hz, 1H), 8.03-8.00 (m, 1H), 7.97-7.95 (m, 1H), 7.46-7.42 (m, 1H), 7.41-7.38 (m, 1H), 7.33-7.29 (m, 1H), 3.88-3.82 (m, 2H), 3.81-3.77 (m, 2H), 3.75-3.69 (m, 2H), 3.55-3.50 (m, 2H), 2.35-2.22 (m, 2H), 2.08-2.00 (m, 2H), 1.65 (s, 3H), 1.61-1.58 (m; 2H); ESI-MS m/z: 计算值 479.2, 实测值 479.93 [M+H] +。 生物学评价
测试例 1: 聚膝苷二磷酸核糖聚合蘇 (PARP-1)抑制剂筛选试验
下面的体外筛选试验是用来测定本发明化合物对于 PARP-酶活性的抑制作用。
实验原理
PARP-1 能催化 NAD+转移 ADP-核糖到邻近的核蛋白如组蛋白上。 本实验用购自 TREVIGEN的 HT Universal Chemiluminescent PARP Assay Kit With Histone-coated Strip Wells试剂盒利用化学发光的方法检测结合到组蛋白上的生物素的聚腺苷二磷酸核糖来 判断 PARP的活性。 采用的阳性参照化合物为 AZD-2281 购自 SelleckChem。
实验方法
本试剂盒采用 ELISA的方法, 即在包被了组蛋白的 96孔板中加入 Χμΐ/well样品、 ΥμΙ/well PARP, PARP cocktail 25μ1Λνε11,总体积为 50ul/well, 同时,设空白对照(不含酶 和样品)和阴性对照 (不含样品), 室温孵育 60min; 用 l xPBS(+0.1%TritonX-100)洗涤 两次,加入 Strep-HRP 5(^l/well,室温孵育 60min; 再用 l <PBS(+0.1%TritonX-100)洗涤两 次,加入化学发光底物 100 μΐ/well,测值。 根据化学发光强度 RLU (相对发光单位)值 计算抑制率, 抑制率 =[i- ( RLU样品 -RLU空白) / ( RLU阴性 -RLU空白) ]><100%。 测 定时每个样品梯度稀释为八个浓度, 设置每个浓度单孔,根据样品抑制率, 应用 XLfit 软件中的 4 Parameter Logi odel计算 IC50, 结果如下表 1所示。
Figure imgf000081_0001
Figure imgf000081_0002
测试例 2、 细胞增殖抑制实验
下面的实验用于在体外条件下测定本发明所述化合物对三阴性表型的乳腺癌细胞 株 MDA-MB-436的的增殖抑制活性。
实验原理
单独应用 PARP-1抑制剂对肿瘤抑制的研究。乳腺癌易感基因 BRCA1和 BRCA2 在 DNA同源重组修复中发挥重要作用。 BRCA缺失细胞具有同源重组修复缺陷, 如果同 时抑制 PARP介导的 DNA修复, 可使细胞 DNA损伤产生增多, 导致染色质失常甚至 细胞死亡。
本实验选择 BRCA缺失的乳腺癌细胞株 MDA-MB-436, 单独应用 PARP抑制剂, 观察其对细胞的损伤作用。 化合物的抑制活性可以用 IC5。值来表示。
实验方法:
分别以 5000 个细胞 /孔和 3000 个细胞 /孔接种生长状态良好的乳腺癌细胞株 MDA-MB-436至相应的 96孔培养板中, 在 37Ό、 5%C02和饱和湿度培养箱孵育过夜。 次日, 加入一系列浓度梯度的样品后, 继续培养 6天, 用 SRB法检测样品对细胞生长 抑制的情况, 计算样品的生长抑制率, 应用 Xlfit软件中的 4 Parameter Logistic Model 计算 IC5。, 结果如下表 2所示。
表 2:
Figure imgf000082_0001
测试例 3、 溶解度测试
为使口服给药后的化合物得以在肠道吸收, 固体或悬浮液剂型在进入体内循环之 前必须崩解、 溶解并扩散至小肠上皮细胞表面, 溶解度大的化合物能更多的扩散到小 肠上皮细胞表面, 使得其在单位时间、 单位表面积可吸收更多量的药物, 不溶性化合 物吸收不完全,因此口服生物利用度低(参见 Drug Metabolism and Disposition, 1998, 26, 152-163; Journal of Medicinal Chemistry, 2001, 44, 1313-1333 )。
实验原理:
本方法将过量的待测样品加入 pH值为 7.4的磷酸盐緩沖溶液,超声 30分钟使药物 不再溶解, 继续在 25 °C的恒温条件下连续振摇 24小时, 使溶液中已溶解的样品成分与 未溶解部分在热力学与动力学方面均达到平衡, 然后过滤得到滤液。 根据" 2010版药典 第二部附录 V D高效液相色谱法 "中的外标法测定滤液中的待测样品的含量, 从而达到 测定待测样品在 pH值为 7.4的嶙酸盐緩沖溶液中平衡溶解度的 S的。
实验方法:
AZD-2281的溶解度测定
色谱条件:
Agilent 1200 LC , Phenomen C18柱( 4.6mm* 150mm, 0.4μηι ), 流动相: 曱醇: 水 ( 60:40 ), 流速: lmL/min,柱温: 25°C , 波长: 230nm, 进样量: ΙΟμί 供试品溶液制备:
取待测化合物样品约 30 mg于 10 mL容量瓶中,加 ρΗ=7.4緩冲液(磷酸盐)约 8 mL, 超声 30 min, 恒温 25 °C摇床振摇 24小时, 25°C环境下, 静置半小时, 用 0.45 μπι水性 滤头过滤, 滤液作为供试品。
标准品溶液制备:
取标准品化合物 AZD-2281于容量瓶中, 用流动相溶解; 分别配制一下表 2中示出 的不同浓度的标准储备液, 进样测得峰面积。 保留时间 8.5 min左右。 以峰面积对浓度 回归, 得回归方程 y = 29.66 x + 302.82 (r = 0.9997), 线性范围 52.45~83.92 g/mL (参见 图 1 )。
表 3
Figure imgf000083_0001
将供试品溶液按以上色谱条件进样 3次, 测得峰面积, 计算相应的浓度。
其余化合物的溶解度测定实验操作参考化合物 AZD-2281的测定。得到的溶解度结 果如下表 4所示。
表 4:
Figure imgf000083_0002
以上通过制备例实施例和测试例详细说明了本发明的代表性化合物的制备方法和 活性及溶解特性。 本发明的化合物可以通过常规的方法合成, 并且与目前同类效果最 佳的化合物 AZD-2281相比, 具有更好的活性和溶解度。

Claims

权 利 要 求 书
1、 一种化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立体异构体, 其中所 述化合物由下式(I )表示:
Figure imgf000084_0001
其中:
R选自如下组中: 氢、 氯、 氟、 溴、 碘、 硝基、 羟基、 氨基、 支链或直链 -。6烷 基、 卤代烷基、 CF3、 CN、 NR''R12、 NH-CO-R13和 O-d- 烷基, 其中 R11和 R12彼此 独立地为氢或 -C4烷基, R13是氢、 CrC4烷基、 -C4垸基 -苯基或苯;
X!、 X2分别独立地选自 -CH或 -N, 但两者不同时为 -N;
X3选自氢、 氯、 氟、 溴和碘;
H环为:
Figure imgf000084_0002
其中 Υ1选自 CH和 Ν, Υ2选自 CH和 Ν, 且 Υ3选自 CH、 CF和 N;
A环为 4 ~ 9元饱和或不饱和单环杂环;
R'选自如下组中: -H, -。(0)1 !、 -C(0)NRi > -C02R, , 烷基、 环烷基、 环烯基、 烷 氧基、 烯氧基、 饱和或不饱和单环杂环、 芳香基和杂芳香基;
R1选自如下组中: 烷基, 环烷基、 环烯基、 烷氧基、 烯氧基、 饱和或不饱和单环 杂环、 芳香基和杂芳香基;
n为选自 0 - 12中的整数;
化合物结构中出现的烷基、 芳香基、 环烷基、 杂芳香基、 饱和或不饱和单环杂环 和叔丁基中的任何一个任选地被一个或多个选自如下组中的取代基所取代: 1¾素、 羟 基、 氰基、 氨基、 亚氨基、 醚基、 硝基、 亚硝基、 巯基、 硫醚、 亚砜、 磺酰基、 硫代 酰胺基、 磺酰胺基、 羧酸、 芳香基、 杂芳香基、 CwQ直链或支链烷基、 Cw。直链或支 链埽基、 酰基、 酰氧基、 酰胺基、 酰基酰胺基、 脲基、 烷基氨基、 酯基、 苯氧基、 苄 氧基、 C3-2Q环烷基、 C5-2G环烯基、 C1- 烷氧基、 C1-2。烯氧基、 C1-2Q烷基破基和 C1-20卤 代烷基; 和
上述杂芳香基和饱和 /或不饱和单环杂环中的杂原子选自 N、 0和8。
2、 根据权利要求 1所述的化合物、 其药学上可接受的盐、 7 合物、 溶剂化物或立 体异构体, 其中, H环为:
Figure imgf000085_0001
3、 根据权利要求 2所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, A环为取代或未取代的包含 1-3个选自 N、 O和 S的杂原子的 4-9元 饱和或不饱和单环杂环; 和
R'选自如下组中: -H, -C(0)R! , -C(0)NR! , -CC^R 取代或未取代的 Cw。烷基, 取代或未取代的 烯基,取代或未取代的 环烷基,取代或未取代的 C5.1G环烯基, 取代或未取代的 Cwo烷氧基, 取代或未取代的 C o烯氧基, 取代或未取代的含 1-3个 选自 N、 0和 S的杂原子的 3-10元饱和或不饱和单环杂环, 取代或未取代 C^。芳香基 和取代或未取代含 1-3个选自 N、 0和 S的杂原子的 C5.1Q杂芳香基, 且
其中 选自如下组中: 取代或未取代的 Cwo烷基, 取代或未取代的 C2-1o烯基, 取代或未取代的 C3_1G环烷基,取代或未取代的 ^.10环烯基,取代或未取代的 Cwo烷氧 基, 取代或未取代的 C2_1()烯氧基, 取代或未取代的含 1-3个选自 N、 0和 S的杂原子 的 3-10元饱和或不饱和单环杂环, 取代或未取代的 C6-1Q芳香基和含 1-3个选自 N、 0 和 S的杂原子的 C5_1Q杂芳香基,
其中所述取代是指被至少一个选自如下组中的基团所取代: !¾素、 羟基、 氰基、 氨基、 羧基、 C2.1G烃基醚基、 硝基、 Cw。烃基磺酰基、 Cwo烃基磺酰胺基、 C6-1G芳香 基、 含 1-3个选自 N、 0和 S的杂原子的 C5-1o杂芳香基、 C1-10直链或支链烷基、 C1-10 直链或支链烯基、 d. 烃基酰基、 C1-1G烃基酰氧基、 C1-1Q烃基酰胺基、 CI-1Q烃基酰基 酰胺基、 脲基、 (C1-H)烷基) 1-2氨基、 。烃基酯基、 苯氧基、 苄氧基、 C3-1。环烷基、 C5-1。环烯基、 C U)烷氧基、 d-u)烯氧基、 .K)烷基硫基和 CWG卤代烷基。
4、 根据权利要求 2所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, A环选自如下组中: 哌嗪、 高哌嗪、 哌啶、 四氢吡啶、 4-羟基哌啶、 2-甲基哌嗪和 3-曱基哌嗪。
5、 根据权利要求 4所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, R'选自如下组中: -H, -C(0)Ri , -C(0)NRl 5 -C02R1 ; 取代或未取代 的 C1-K)烷基, 取代或未取代的 Cwo烯基, 取代或未取代 C o芳香基, 和取代或未取代 含 1-3个选自 N、 0和 S的杂原子的 CWQ杂芳香基, 且
其中!^选自如下组中: 取代或未取代的 C1-10烷基, 取代或未取代的 Cwo烯基, 取代或未取代的 C3-7环烷基, 取代或未取代的 C5-7环烯基, 取代或未取代的 Cwo烷氧 基,取代或未取代的含 1-3个选自 N、 0和 S的杂原子的 5-7元饱和或不饱和单环杂环, 取代或未取代的 Cwo芳香基, 和取代或未取代的含 1-3个选自 N、 0和 S的杂原子的 Cw。杂芳香基,
所述取代是指被至少一个选自如下组中的基团所取代: 卤素、 羟基、 氰基、 氨基、 硝基、 Cw。芳香基、 C1-1Q直链或支链烷基、 C1-1Q烃基酰氧基、(Cw。烷基) 1-2氨基、 d.io 烃基酯基、 苯氧基、 苄氧基、 C3.7环烷基、 Cwo烷氧基和 Cwo卤代烷基。
6、 根据权利要求 4所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, 所述化合物由下式(Π )或 (III )表示:
Figure imgf000086_0001
7、 根据权利要求 6所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, R'为 - C R 其中 !^选自如下组中: 取代或未取代的 C3-6烷基, 取代或未取代的 C3.6环烷基, 取代或未取代的含 1-2个选自 N、 0和 S的杂原子的 5-6 元饱和或不饱和单环杂环, 和取代或未取代的苯基,
所述取代是指被至少一个选自如下组中的基团所取代: 鹵素、 羟基、 氰基、 氨基、 硝基、 苯基、 Cw直链或支链烷基和(d_3烷基) 1-2氨基。
8、 根据权利要求 6所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, R选自如下组中: 氢、 氟、 氯、 溴、 硝基和氨基; 且 X X2分别为 -CH。
9、 根据权利要求 8所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或立 体异构体, 其中, R'为选自如下组中的基团: 任选地被选自氟、 氯、 硝基、 氨基、 甲基 氨基、 二甲基氨基、 甲基和苯基中的一个或两个取代基所取代的乙基、 丙基、 丁基、 戊基、 环丙烷基、 环丁烷基、 环己烷基、 笨基、 吡咯烷基和哌啶基。
10、 根据权利要求 1 所述的化合物、 其药学上可接受的盐、 水合物、 溶剂化物或 立体异构体, 其中所述化合物选自以下式 1-130所示的化合物:
4- (3- (4- (环丙烷羰基)哌。秦小羰基)苄氧基) -2H-舦嗪小酮,
4- (3- (4-苯曱跣基哌啶小羰基)苄基氧基) 2H-酞嗪小酮,
4- (3- ( 4-丙酰基哌嗪小羰基)苄氧基) -2H-酞嗪小酮,
4- (3- (4- (环己烷羰基)哌嗪小羰基)苄氧基) -2H-酞嗪小酮,
4- (3- (4- (环戊烷羰基)派嗪 -1-羰基)苄氧基) -2H-舦嗪小酮,
4 - (3- ( 4-乙酰基哌嗪 -1 -羰基)苄氧基) -2H-酞嗪 -1-酮,
4- (3- (4- (环丙烷羰基)哌嗪 -1 -羰基) 苯氧基) -2H-舦嗪小酮,
4- (3- ( 4-丙酰基哌嗪 -1-羰基)苯氧基) -2H-酞嗪小酮,
4 - (3- ( 4-苯曱酰基哌嗪 -1-羰基)苯氧基) -2H-舦。秦小酮,
4- ( 3- ( 4 - (环己烷羰基)哌嗪 -1-羰基 )笨氧基 ) -2H-酞嗪小酮,
4- (3- (4 - (环戊垸羰基) 嗪小羰基) 苯氧基) -2H-酞嗪 -1-酮,
4- ( 3- ( 4 -乙酰基哌嗪小羰基)苯氧基 ) -2H-酞嗪小酮,
4- (3- (4- (1-苄氧羰基-哌啶)羰基哌嗪 -1-羰基) 苯氧基) -2H-酞嗪 -1-酮,
4 - (3- ( 4-对硝基苯基哌嗪 -1-羰基)苄氧基) -2H-酞嗪小酮,
4 - (3- (4-对硝基苯曱酰基哌嗪小羰基)苄氧基) -2H-舦嗪小酮,
4- (3- (4-苄基 -1,4-二氮杂环庚小羰基)苄氧基) -2H-舦嗪小酮,
4- ( 3- ( 4- (3-氯 -4-三氟甲基-苯基) -4-羟基哌啶 -1-羰基)苄氧基) -2H- 嗪小酮,
4- (3- (4 - (4-氟笨基) -1,2,3,6-四氢吡啶小羰基)苄氧基 -2H-酞嗪小酮,
4— (3- (4- (嘧啶 -2-基)哌嗪小羰基)苄氧基) -2H-酞嗪小酮,
4- (3- (4-叔丁氧羰基 -1,4-二氮杂环庚小羰基)苄氧基) -2H-酞嗪小酮,
4- (3- (4- (3-氯-苯基) -4-羟基哌啶 -1-羰基)苄氧基) -2H-酞嗪小酮,
4- (3- (4- (4-氯-苯基) -4-羟基哌啶小羰基)苄氧基) -2H-酞。秦小酮,
4- (3- (4-笨基哌嗪 -1 -羰基)苄氧基) -2H-酞嗪 -1-酮,
4- (3- (4-苯基 -1,2,3,6- 四氢吡啶 -1 -羰基)苄氧基) -2H-酞咯小酮,
4- ( 3 - ( 4- (4-氟-苯基) -1,2,3,6- 四氢吡啶 -1-羰基)苯氧基) -2H-酞嗪小酮,
4- ( 3 - ( 4 - 异烟酰基哌嗪 -1 -羰基)苯氧基) -2H-舦嗪小酮, 4- (3- (4- (4-氟苯曱酰基)哌嗪小羰基)苯氧基) -2H-酞嗪小酮,
5- 氟 (3- (4- (4-硝基苯甲酰基)哌嗪小羰基)苄氧基) -2H-舦嗪小酮,
8-氟 -4- (3- (4- (1-苄氧羰基-哌啶)羰基哌喚小羰基)苯氧基) -2H-酞嗪小酮, 4- (2-氟- (4- (1-苄氧羰基-哌啶)羰基哌嗪小羰基)苯氧基) -2H-酞嗪小酮, 4- (4-氟 -3- (4- (4-硝基苯曱酰基)哌嗪 -1-羰基)苯氧基) -2H-酞嗪小酮,
4- (4-氟 -3- (4- (5- 曱基异恶唑 -4-羰基)哌喚 -1- 羰基)苯氧基 -2H-H1-酮 ,
4- (2- 氟 -5- (4- 氧代 -3,4-2H- 酞嗪小基氧基)苯曱酰基) -N-苯基哌嗪小甲酰胺,
5-氯斗(4-氟 -3- (4- (4-氟苯曱酰基)派嗪 -1-羰基)苯氧基) -2H-舦嗪小酮, 5-氯 -4- (4-氟 -3- (4- 苯曱酰基哌嗪 -1-羰基)苯氧基) -2H-酞嗪 -1-酮,
4 - ( 4 -氟 -3 - ( 4 - ( 2 -曱基噻唑 -4 -羰基)唳嗪 -1 -羰基)苯氧基 -2H-酞。秦小酮, 4 - (4-氟 -3- (4- (4-氟苯基)哌。秦-卜羰基)苯氧基) -5-硝基 -2H-舦嗪小酮, 4- (4-氟 -3- (4- (4-氟笨甲酰基)哌嗪小羰基)苯氧基) -5-硝基 -2H-酞嗪小酮, 4- (4- 氟 -3- (4-苯曱酰基哌嗪 -1-羰基) 苯氧基) -5-硝基 -2H-酞嗪小酮,
8 -硝基 4- (2-氟- (4- ( 1-苄氧羰基-哌啶)羰基哌嗪小羰基)苯氧基) -2H-酞嗪 -1-酮, 4- (4-氟 -3- (4- (4-硝基苯曱酰基)哌嗪 -1-羰基)苯氧基) -5-硝基 -2H-酞嗪小酮,
N-环己基 -4 - ( 2 - 氟 -5 - (4- 氧代 -3,4 - 2H- 酞嗪 -1 -基氧基)苯曱酰基)哌嗪 -1 - 曱酰胺,
4- (3- ( 4-乙酰基哌嗪 -1 -羰基) -4-氟苯氧基) -2H-酞嗪小酮,
4 - ( 4 -氟 -3 - ( 4 - (噻唑 -4 -羰基) 嗪 -1 -羰基)笨氧基 ) -2H-酞。秦 -1-S同,
4- (2-氟- (4- (2-苄氧羰基-哌啶)羰基哌嗪小羰基)苯氧基) -2H-酞嗪小酮, 4- (2-氟- (4- (3-苄氧羰基-哌啶)羰基哌嗪 -1-羰基)笨氧基) -2H-酞嗪小酮, 4- (4- 氟 -3- (4- (4-氧代环己烷羰基) 嗪 -1 -羰基) 苯氧基) -2H-酞。秦小酮, 4 - ( 4 -氟 -3 - ( 4 - ( 4 -氰基苯曱酰基) 嗪 -1 -羰基)苯氧基) -2H-舦嗪 -1-S同, ( R ) -4- ( 4 - 氟 -3- (4- (2-苯基丙酰基)哌嗪 -卜羰基)苯氧基) -2H-酞嗪小酮, 4 - ( 3- ( 4- (环丙烷羰基)哌嗪 -1 -羰基) -4- 氟苯氧基) -2H-酞嗪 -1-酮,
1 - (4- (2- 氟 -5- (4- 氧代 -3-1,4-二氢二氮杂萘 -1 - 基氧基)苯甲酰基)哌嗪 -1- 基) 丙烷 -1,2- 二酮酞嗪,
4- (4- 氟 -3- (4- (5-曱基异恶唑 -3-羰基)哝嗪小羰基)苯氧基) -2H-舦。秦小酮, 4- (4- 氟 -3- (4- (2,2,2- 三氟乙酰基)哌嗪 -1 -羰基)苯氧基) -2H-酞。秦小酮, 4- (4- 氟 -3- (4- (3,3,3- 三氟丙酰基)。泉嗪-卜 羰基)苯氧基) -2H-酞嗪 -1-酮, 4- (4-氟 -3- (4-新戊酰基哌嗪 -1 -羰基)苯氧基) -2H-酞嗪 -1-酮,
4- (4- 氟 -3- (4 - (2- 曱氧基乙酰基)哌嗪 -1 - 羰基)苯氧基 -2H-酞嗪 -1-酮, 4- ( 4 - 氟 -3 - (4- ( 2-氰基苯甲酰基 )哌嗪 -1 -羰基)苯氧基 ) -2H-酞嗪小酮, 4- (2-氟 -5- (4-氧代-3,4-211-舦。秦-1 -基氧基)苯曱酰基) -N-异丙基 -1 -甲酰胺, 4- ( 3- ( 4- (2-乙氧基乙酰基)哌。秦 -1 -羰基) -4-氟苯氧基) -2H-酞嗪 -1-酮,
4- (4- 氟 -3- (4- 异烟酰基哌嗪- 羰基)苯氧基) -2H-酞嗪小酮,
4- ( 3- ( 4- (2-乙基丁耽基)哌喚 -1 -羰基) -4-氟苯氧基) -2H-酞嗪 -1-酮, 4- (4- 氟 -3 - (4- (4, 4—二氟哌啶 -1 -羰基)哌嗪 -1 -羰基)苯氧基) -2H-酞嗪 -1-酮,
4- (4-氟 -3- (4- (噻吩 -2-羰基) p/艮嗪 -1 -羰基)苯氧基) -2H-酞嗪小酮,
4- (4- 氟 -3- (4 - (3-羟丙基)哌嗪 -卜羰基)苯氧基) -2H-酞嗪小酮,
1- (2-氟 -5- (4-氧代-3,4-2 酞嗪 -卜基氧基)苯曱酰基)哌啶 -4-曱腈,
4- (4- 氟 -3- (4- 异丁酰基哌。秦-卜 狻基) 苯氧基) -2H-酞嗪小酮,
4- (2- 氟 -5- (4- 氧代 -3,4-2H- 酞嗪 -1 -基氧基)苯曱酰基) -N, N-二曱基派。秦 -1 - 曱酰胺,
4-(4-氟 _3- ( 4- ( 3-三氟曱基 -4-甲基-苯曱酰基 )哌嗪 -1-羰基)苯氧基 ) -2H-舦嗪 -1-S同,
4- (3- (4 -丙烯酰基哌嗪 -1 -羰基) -4 -氟苯氧基 ) -2H-酞嗪 -1 -酮,
4- (4- 氟 -3- (4- (2-羟基丙酰基)哌嗪 -1 -羰基)苯氧基) -2H-酞嗪小酮,
4- (3- (4- (4, 4-二氟哌啶小羰基)哌啶 -1 -羰基) -4-氟苯氧基) -2H-酞。秦小酮,
4- (4-氟 -3- (4- (哌啶 -1 -羰基)哌啶小羰基)苯氧基) -2H-酞嗪 -1 -酮,
1- (2-氟-- (4- 氧代 -3,4-2H- 狀嗪小基氧基)苯甲酰基) -N-苯基派啶 -4-曱酰胺,
N-乙基小(2-氟 -5- (4-氧代 -1,4-二氢 -酞嗪小基氧基)苯曱酰基)哌啶 -4-曱酰胺,
4-(3- ( 4- ( 4- (二甲基氨基)笨曱酰基)哌嗪 -1 -羰基) -4 -氟苯氧基 ) -2H-酞嗪 -1-S同,
4 - (3- (4 -叔丁基哌嗪 -1-羰基) -4-氟苯氧基) -2H-酞嗪小酮,
4- (4-氟 -3- (4- (呋喃 -2-羰基)哌嗪 -1 -羰基)苯氧基) -2H-酞。秦 -1-酮,
4- (4- 氟 -3- (4- 异 基小羰基)苯氧基) -2H-酞嗪 -1-酮,
4- (3- (4-烯丙基哌。秦 -1 -羰基) -4-氟苯氧基) -2H-酞嗪小酮,
4- (3- (4- 3-氰基苯曱酰基哌嗪 -1 -羰基 ) -4 - 氟苯氧基 ) -2H-酞嗪小酮,
4- (3- (4- ( 1H-吡唑 -1 -羰基 )哌嗪 -1 -羰基 ) -4 - 氟苯氧基 ) -2H-酞嗪 -1-酮,
4- (4-氟 -3- (4- (吗啉 -4-羰基)哌嗪 -1-羰基)苯氧基) -2H-酞嗪小酮,
4- (4-氟 -3- (4- (吡咯烷 -1 -羰基)哌嗪 -1 -羰基) 苯氧基) -2H-酞喙小酮,
4- (4-氟 -3- (4- (哌啶 -1-羰基)哌嗪 -1 -羰基)苯氧基) -2H-酞嗪小酮,
4- (3- (4- 氧代 -3,4- 二氢 -舦嗪小基氧基)苯曱酰基) -N- (3- (三氟曱基)苯基) 哌嗪-卜 曱酰胺,
4- (4-氟 -3- (4- (3-硝基苯甲酰基)哝嗪 -1-羰基)苯氧基) -2H-酞嗪小酮,
4- (4-氟 -3- (4- (2-硝基苯曱酰基)哌嗪 -1-羰基)苯氧基) -2H-酞嗪小酮,
( E ) -4 - ( 3- (4-丁 -2-烯酰基 嗪小羰基) -4-氟苯氧基-) 2H-酞嗪 -1-酮,
4- (4-氟 -3- (4- 曱基哌嗪 -1-羰基)苯氧基) -2H-酞咯小酮,
4- (4-氟 -3- (4- (恶唑 -4-羰基)哌嗪 -1-羰基)苯氧基) 2H-酞嗪小酮,
4- (4-氟 -3- (4- (2-羟乙基)哌嗪 -1 -羰基)苯氧基) 2H-酞嗪小酮,
4- (3- (4- ( 1H-1,2,4-三唑 -1 -羰基)哌嗪 -1 -羰基) -4-氟苯氧基) 2H-酞嗪小酮,
( R ) -4- (4-氟 -3- (4- (四氢呋喃 -2-羰基)哌嗪 -1-羰基)苯氧基) 2H-酞。秦 -1-酮, 4- (4-氟 -3- (4- (四氢呋喃 -2-羰基)哌嗪 -1 -羰基) 笨氧基) 2H-酞嗪 -1-酮,
N- ( 4,4 -二氟环己基) -4 - ( 2 - 氟 -5 - (4- 氧代 -3,4 - 二氢 - 酞嗪 -1 -基氧基 ) 苯甲酰基)哌嗪 -1 - 甲酰胺,
N—环丙基 4- (2-氟 -5- ( 4-氧代 -3,4-二氢-酞嗪小基氧基)苯曱酰基)哌嗪小曱酰胺,
4- ( 3- ( 4- ( 1H-咪唑 -1-羰基)哌嗪 -1 -羰基) -4-氟苯氧基) 2H-舦嗪小酮,
4- (3- (4- (环己烷羰基)哌嗪 -1 -羰基 -4-氟苯氧基) 2H-酞嗪 -1-酮,
4- (4喜 3- ( 4-丙酰基哌嗪 -1 -羰基)苯氧基) 2H-酞嗪小酮,
4- ( 3- ( 4 - (环戊基羰基)哌嗪 -1-羰基) -4 -氟苯氧基 ) 2H-酞嗪小酮,
8-氯 4- (2-氟- (4- (4-苄氧羰基-哌啶)羰基哌嗪小羰基)苯氧基 -2H-酞嗪小酮,
4- (3- (4- 苯曱酰基哌嗪 -1 -羰基) -4-氟苯氧基) 2H-酞唤小酮,
5—氯—4- (4- 氟 -3- (4- (4-氟苯基)哌嗪 -1-羰基)苯氧基) 2H-耿嗪 -1-酮,
1-苄氧羰基 -4- (4- (3- ((7-氟 -4-氧代 -3-1,4-二氢 -酞。秦小基氧基) 曱基)苯曱酰基) 哌嗪 -1 -羰基)哌啶,
5-氯 -4- (4-氟 -3- (4- (4-硝基苯甲酰基)哌嗪 -1-羰基)苯氧基) 2H-酞嗪 -1-酮, 4 - ( 3 - ( 4 - ( 4,4 -二氟环己烷薮基 )哌嗪 -1 -羰基 ) -4 -氟苯氧基 ) 2H-酞嗪 -1-酮, 1-苄氧羰基 -2- (4- (2-氟 -5- ( 4-氧代 -3,4-二氢二氮杂萘 -1 -基氧基)笨甲酰基)哌 嗪 -1-羰基)吡咯烷,
4- (4-氟 -3- (4- (吡咯烷 -2- 羰基)哌嗪 -1 -羰基)苯氧基) 2H-舦。秦 -1-酮,
4- (4-氟 -3- (4- (4-氟苯基)哌嗪 -1 -羰基)苯氧基) 2H-酞嗪 -1-酮,
4- (4-氟 -3- (4- (四氢呋喃 -3-羰基)哌咯小羰基)苯氧基) 2H-酞0秦小酮,
N- (4- 氯苯基) -4- (2-氟 -5- (4-氧代 -3,4-二氢二氮杂萘 -1-基氧基)苯甲酰基)派咯 -1 -曱酰胺,
N, N-二乙基 -4- (2- 氟 -5- (4-氧代-3,4 -二氢 -狀嗪小基氧基) 苯曱酰基)哌。秦小曱 酰胺,
4- (4-氟 -3- (4- (四氢吡喃 -4-羰基)哌嗪 -1 -羰基)苯氧基) 2H-酞嗪小酮,
4- (4-氟 -3- (4- (4- (三氟曱基)苯曱酰基)哌嗪 -1 -羰基)苯氧基) 2H-酞。秦 -1-S同,
4- (4- 氟 -3- (4- (4-氟苯曱酰基)哌嗪 -1 -羰基)苯氧基) 2H-舦嗪 -1-酮,
4- (4-氟 -3- (4- (4-氟苄基)哌嗪小羰基)苯氧基) -2H-酞嗪 -1 -酮,
4- (4-氟 -3- (哌嗪小羰基)苯氧基) 2H-酞嗪 -1-酮,
4- (4-氟 -3- (4-吡啶甲酖基哌嗪小羰基)苯氧基) 2H-舦嗪 -1-酮,
( S ) -4 - (4-氟 -3- (4- (四氢呋喃 -2-羰基)哌嗪 -1-羰基)苯氧基) 2H-酞。秦小酮,
4- ( 3- ( 4- ( 2- 环丙基乙酰基) 嗪 -1-羰基) -4-氟苯氧基) 2H-酞嗪小酮,
4- (3 - (4- (2- 环戊基乙酖基)哌嗪 -1 -羰基) -4-氟苯氧基) 2H-酞嗪 -1-酮,
4- ( 4 - 氟 -3 - (4- ( 1 - (曱基氨基 )环丙烷羰基)派嗪 -1 -羰基)苯氧基) 2H- 酞嗪 -1-酮,
4-(3- (4- (1- (二甲基氨基)环丙烷羰基)哌嗪 -1-羰基) -4-氟苯氧基) 2H-酞嗪小 S同, 4- (3- (4- ( 1-氨基环丙烷羰基)哌嗪 -1 -羰基) -4-氟苯氧基) 2H-酞嗪小酮, 4-(3- (4- ( 1- (二曱基氨基)环丁烷羰基)哌嗪小羰基) -4-氟苯氧基) 2H-酜嗪 -1-S同, 4- (4-氟 -3- (4- ( 1- (曱基氛基)环丁烷羰基)哌嗪 -1-羰基)苯氧基) 2H-舦嗪 -1-S同, 4- (3- (4- ( 1 -氨基环丁烷羰基 )哌嗪 -1 -羰基) -4 -氟苯氧基 ) 2H-酞嗪 -1 -酮, ( R ) .4 - ( 3 - ( 4 - ( 1,2 -二曱基吡咯烷 -2 -羰基 )哌嗪 -1 -羰基 ) -4 -氟苯氧基 ) 2H- 酞嗪 -1-酮, 和
( R ) -4-(4-氟 -3- ( 4- ( 2-曱基吡咯烷 -2-羰基)哌嗪小羰基)苯氧基 )-2H-酞嗪小酮。
11、 根据权利要求 1-10中任一项所述化合物、 其药学上可接受的盐、 水合物、 溶 剂化物或立体异构体, 在制备治疗由 PARP介导的病症的药物中的应用。
12、 根据权利要求 11所述的应用, 其中所述由 PARP介导的病症选自癌症、 神经 变性型疾病、 心血管疾病、 糖尿病和炎症。
13、根据权利要求 12所述的应用, 所述癌症为组织细胞性淋巴癌、 非小细胞肺癌、 小细胞肺癌、 肺腺癌、 肺鳞癌、 胰腺癌、 乳腺癌、 前列腺癌、 肝癌、 胃癌、 结肠癌、 直肠癌、 卵巢癌、 宫茎癌、 脑癌、 食道癌、 骨癌、 睾丸癌、 黑色素癌、 皮肤癌、 上皮 细胞癌、 前列腺癌、 鼻咽癌、 口腔癌、 白血病, 以及脑、 生殖系统、 淋巴系统、 消化 系统肿瘤、 呼吸系统肿瘤和皮肤肿瘤中的任一种。
14、 一种药用组合物, 包括有效量的根据权利要求 1-10中任一项所述化合物、 其 药学上可接受的盐、 水合物、 溶剂化物或立体异构体, 以及药学上可接受的载体。
PCT/CN2012/001611 2011-11-30 2012-12-03 一种多聚(adp-核糖)聚合酶抑制剂 WO2013078771A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP12852659.7A EP2799435B1 (en) 2011-11-30 2012-12-03 Poly (adp-ribose) polymerase inhibitor
JP2014543747A JP5820081B2 (ja) 2011-11-30 2012-12-03 ポリ(adp−リボース)ポリメラーゼ阻害剤
US14/361,698 US9187430B2 (en) 2011-11-30 2012-12-03 Poly (ADP-ribose) polymerase inhibitor
CA2857405A CA2857405C (en) 2011-11-30 2012-12-03 Phthalic hydrazide (phthalazine ketone) compounds as parp inhibitors and composition thereof
US14/826,181 US9718787B2 (en) 2011-11-30 2015-08-13 Poly (ADP-ribose) polymerase inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201110390364 2011-11-30
CN201110390364.1 2011-11-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/361,698 A-371-Of-International US9187430B2 (en) 2011-11-30 2012-12-03 Poly (ADP-ribose) polymerase inhibitor
US14/826,181 Continuation-In-Part US9718787B2 (en) 2011-11-30 2015-08-13 Poly (ADP-ribose) polymerase inhibitor

Publications (1)

Publication Number Publication Date
WO2013078771A1 true WO2013078771A1 (zh) 2013-06-06

Family

ID=48491227

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/001611 WO2013078771A1 (zh) 2011-11-30 2012-12-03 一种多聚(adp-核糖)聚合酶抑制剂

Country Status (6)

Country Link
US (2) US9187430B2 (zh)
EP (1) EP2799435B1 (zh)
JP (1) JP5820081B2 (zh)
CN (1) CN103130723B (zh)
CA (1) CA2857405C (zh)
WO (1) WO2013078771A1 (zh)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015179559A3 (en) * 2014-05-21 2016-06-09 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
EP3030557A4 (en) * 2013-09-13 2017-03-22 Ildong Pharm Co., Ltd. A novel phtalazinone derivatives and manufacturing process thereof
CN107082764A (zh) * 2017-05-23 2017-08-22 上海锐聚恩新药研发有限公司 一类多羟基酞嗪酮化合物、其制备方法及应用
US10266497B2 (en) 2017-05-23 2019-04-23 Abide Therapeutics, Inc. Pyrazole MAGL inhibitors
CN109843885A (zh) * 2016-10-14 2019-06-04 上海汇伦生命科技有限公司 抗肿瘤杂环并咪唑类化合物的药用盐
US10323038B2 (en) 2015-11-20 2019-06-18 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
US10385057B2 (en) 2015-11-20 2019-08-20 Lundbeck La Jolla Research Center, Inc. Pyrazole compounds and methods of making and using same
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10519134B2 (en) 2015-11-20 2019-12-31 Lundbeck La Jolla Research Center, Inc. Pyrazole compounds and methods of making and using same
US10583137B2 (en) 2015-12-02 2020-03-10 The Scripps Research Institute Triazole DAGLα inhibitors
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US10927105B1 (en) 2017-05-23 2021-02-23 Lundbeck La Jolla Research Center, Inc. Pyrazole MAGL inhibitors
US11149037B2 (en) 2017-05-23 2021-10-19 H. Lundbeck A/S Pyrazole MAGL inhibitors
US11390608B2 (en) 2020-04-21 2022-07-19 Idience Co., Ltd. Crystalline forms of phthalazinone compound

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103130723B (zh) * 2011-11-30 2015-01-14 成都地奥制药集团有限公司 一种多聚(adp-核糖)聚合酶抑制剂
CN102898377B (zh) * 2012-02-14 2016-01-20 南京圣和药业股份有限公司 一类酞嗪酮衍生物及其用途
CN105017140B (zh) * 2014-04-28 2017-12-29 复旦大学 邻氨基苯甲酰胺化合物及其制备方法和用途
CN105777651A (zh) * 2015-01-13 2016-07-20 江苏豪森药业集团有限公司 聚腺苷酸二磷酸核糖转移酶抑制剂的晶型及其制备方法和医药用途
CN106946792B (zh) * 2017-03-29 2019-07-26 深圳市坤健创新药物研究院 一种酞嗪酮的异羟肟酸衍生物及其制备方法与应用
WO2020077123A1 (en) 2018-10-12 2020-04-16 Terns, Inc. Thyroid hormone receptor beta agonist compounds
CN111320609A (zh) 2018-12-13 2020-06-23 拓臻股份有限公司 一种THRβ受体激动剂化合物及其制备方法和用途
AU2020346057A1 (en) * 2019-09-12 2022-04-21 Terns Pharmaceuticals, Inc. Thyroid hormone receptor beta agonist compounds
CN111732844A (zh) * 2020-07-08 2020-10-02 高雪梅 一种家居防火涂料及其制备方法
CN117897380A (zh) * 2021-08-17 2024-04-16 益方生物科技(上海)股份有限公司 哒嗪酮或吡啶酮化合物、其制备方法和用途
WO2023192864A2 (en) * 2022-03-28 2023-10-05 Oregon Health & Science University Covalent parp16 inhibitors
WO2023192989A2 (en) * 2022-04-01 2023-10-05 Azkarra Therapeutics, Inc. Phthalazinone compounds as parp7 inhibitors
WO2024146631A1 (zh) * 2023-01-06 2024-07-11 齐鲁制药有限公司 一种parp7抑制剂的制备方法

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5177075A (en) 1988-08-19 1993-01-05 Warner-Lambert Company Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process
WO1999011645A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Poly(adp-ribose) polymerase ('parp') inhibitors, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
WO2000032579A1 (de) 1998-11-27 2000-06-08 Basf Aktiengesellschaft Substituierte benzimidazole und ihre verwendung als parp inhibitoren
WO2002036599A1 (en) 2000-10-31 2002-05-10 Smithkline Beecham P.L.C. Thieno[2,3-c]isoquinolines for use as inhibitors of parp
WO2003063874A1 (en) 2002-01-29 2003-08-07 Fujisawa Pharmaceutical Co., Ltd. Condensed heterocyclic compounds
WO2003103666A2 (en) 2002-06-07 2003-12-18 Altana Pharma Ag Novel 4,5-dihydro-imidazo[4,5,1-ij]quinolin-6-ones
WO2004096779A1 (en) 2003-04-30 2004-11-11 Suemegi Balazs Quinazolinone-derivatives and their use for preparation of pharmaceutical compositions having parp enzyme inhibitory effect
US20040248931A1 (en) 2002-10-01 2004-12-09 Mitsubishi Pharma Corporation Isoquinoline compound and pharmaceutical use thereof
WO2005023246A1 (en) 2003-09-04 2005-03-17 Aventis Pharmaceuticals Inc. Substituted indoles as inhibitors of poly (adp-ribose) polymerase (parp)
WO2005054210A1 (en) 2003-12-05 2005-06-16 Janssen Pharmaceutica N.V. 6-substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
WO2006003148A1 (en) 2004-06-30 2006-01-12 Janssen Pharmaceutica N.V. Quinazolinedione derivatives as parp inhibitors
US20060063926A1 (en) 2004-09-22 2006-03-23 Agouron Pharmaceuticals, Inc. Method of preparing poly(ADP-ribose) polymerases inhibitors
US20070093489A1 (en) 2005-10-19 2007-04-26 Kudos Pharmaceuticals Limited Phthalazinone derivatives
CN101048399A (zh) * 2004-08-26 2007-10-03 库多斯药物有限公司 4-杂芳基甲基取代的酞嗪酮衍生物
WO2007138355A1 (en) 2006-05-31 2007-12-06 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Pyrrolo[1,2-a]pyrazin-1(2h)-one and pyrrolo[1,2-d][1,2,4]triazin-1(2h)-one derivatives as inhibitors of poly(adp-ribose)polymerase(parp)
WO2008017883A2 (en) 2006-08-09 2008-02-14 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa 4-oxo-4,5-dihydropyrrolo[1,2-a] quinoxaline derivatives as inhibitors of poly(adp-ribose)polymerase(parp)
CN101925595A (zh) * 2008-01-23 2010-12-22 阿斯利康(瑞典)有限公司 酞嗪酮衍生物

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA000999B1 (ru) * 1995-08-02 2000-08-28 Ньюкастл Юниверсити Венчерз Лимитед Соединение бензимидазола
DE19921567A1 (de) 1999-05-11 2000-11-16 Basf Ag Verwendung von Phthalazine-Derivaten
HU228960B1 (hu) 2000-10-30 2013-07-29 Kudos Pharm Ltd Ftalazinon-származékok
US7151102B2 (en) 2000-10-30 2006-12-19 Kudos Pharmaceuticals Limited Phthalazinone derivatives
GB0026505D0 (en) * 2000-10-30 2000-12-13 Kudos Pharm Ltd Phthalazinone derivatives
KR100957516B1 (ko) * 2001-08-15 2010-05-14 이코스 코포레이션 2h-프탈라진-1-온 및 이것의 사용 방법
CA2482806A1 (en) * 2002-04-30 2003-11-13 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7449464B2 (en) 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
BRPI0408284B8 (pt) * 2003-03-12 2021-05-25 Kudos Pharm Ltd compostos derivados de ftalazinona, seu uso e composição farmacêutica compreendendo os mesmos
GB0305681D0 (en) 2003-03-12 2003-04-16 Kudos Pharm Ltd Phthalazinone derivatives
CN1905864B (zh) 2003-12-01 2011-04-06 库多斯药物有限公司 用于治疗癌症的dna损伤修复抑制剂
ES2545613T3 (es) 2003-12-01 2015-09-14 Kudos Pharmaceuticals Limited Inhibidores de reparación de daño en el ADN para el tratamiento del cáncer
BRPI0514632A (pt) 2004-08-26 2008-06-17 Kudos Pharm Ltd derivados de ftalazinona 4-heteroarilmetila substituìdos
JP2013525303A (ja) * 2010-04-16 2013-06-20 アッヴィ・インコーポレイテッド フタラジン−(2h)−オン系キナーゼ阻害薬
JP2013532683A (ja) 2010-07-27 2013-08-19 カディラ ヘルスケア リミティド ポリ(adpリボース)ポリメラーゼ−1阻害剤としての、置換4−(4−フルオロ−3−(ピペラジン−1−カルボニル)ベンジル)フタラジン−1(2h)−オン誘導体
CN102485721B (zh) 2010-12-03 2015-12-09 曹亚 取代的2,3-二氮杂萘酮化合物及其用途
CN103130723B (zh) * 2011-11-30 2015-01-14 成都地奥制药集团有限公司 一种多聚(adp-核糖)聚合酶抑制剂
CN102898377B (zh) * 2012-02-14 2016-01-20 南京圣和药业股份有限公司 一类酞嗪酮衍生物及其用途

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5177075A (en) 1988-08-19 1993-01-05 Warner-Lambert Company Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process
WO1999011645A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Poly(adp-ribose) polymerase ('parp') inhibitors, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
WO2000032579A1 (de) 1998-11-27 2000-06-08 Basf Aktiengesellschaft Substituierte benzimidazole und ihre verwendung als parp inhibitoren
WO2002036599A1 (en) 2000-10-31 2002-05-10 Smithkline Beecham P.L.C. Thieno[2,3-c]isoquinolines for use as inhibitors of parp
WO2003063874A1 (en) 2002-01-29 2003-08-07 Fujisawa Pharmaceutical Co., Ltd. Condensed heterocyclic compounds
WO2003103666A2 (en) 2002-06-07 2003-12-18 Altana Pharma Ag Novel 4,5-dihydro-imidazo[4,5,1-ij]quinolin-6-ones
US20040248931A1 (en) 2002-10-01 2004-12-09 Mitsubishi Pharma Corporation Isoquinoline compound and pharmaceutical use thereof
WO2004096779A1 (en) 2003-04-30 2004-11-11 Suemegi Balazs Quinazolinone-derivatives and their use for preparation of pharmaceutical compositions having parp enzyme inhibitory effect
WO2005023246A1 (en) 2003-09-04 2005-03-17 Aventis Pharmaceuticals Inc. Substituted indoles as inhibitors of poly (adp-ribose) polymerase (parp)
WO2005054210A1 (en) 2003-12-05 2005-06-16 Janssen Pharmaceutica N.V. 6-substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
WO2006003148A1 (en) 2004-06-30 2006-01-12 Janssen Pharmaceutica N.V. Quinazolinedione derivatives as parp inhibitors
CN101048399A (zh) * 2004-08-26 2007-10-03 库多斯药物有限公司 4-杂芳基甲基取代的酞嗪酮衍生物
US20060063926A1 (en) 2004-09-22 2006-03-23 Agouron Pharmaceuticals, Inc. Method of preparing poly(ADP-ribose) polymerases inhibitors
US20070093489A1 (en) 2005-10-19 2007-04-26 Kudos Pharmaceuticals Limited Phthalazinone derivatives
WO2007138355A1 (en) 2006-05-31 2007-12-06 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Pyrrolo[1,2-a]pyrazin-1(2h)-one and pyrrolo[1,2-d][1,2,4]triazin-1(2h)-one derivatives as inhibitors of poly(adp-ribose)polymerase(parp)
WO2008017883A2 (en) 2006-08-09 2008-02-14 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa 4-oxo-4,5-dihydropyrrolo[1,2-a] quinoxaline derivatives as inhibitors of poly(adp-ribose)polymerase(parp)
CN101925595A (zh) * 2008-01-23 2010-12-22 阿斯利康(瑞典)有限公司 酞嗪酮衍生物

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
"Appendix VD High Performance Liquid Chromatography", PART II OF PHARMACOPEIA, 2010
CALABRESE ET AL., J NATL CANCER INST, vol. 96, 2004, pages 56
CUZZOCREA ET AL., EUR. J. PHARMACOLOGY, vol. 342, 1998, pages 67
DELANEY ET AL., CLIN CANCER RES, vol. 6, 2000, pages 2860
DRUG METABOLISM AND DISPOSITION, vol. 26, 1998, pages 152 - 163
ELIASSON ET AL., NATURE MED., vol. 3, 1997, pages 1089
ENDRES ET AL., J. CEREB. BLOOD FLOW METAB., vol. 17, 1997, pages 1143
GILAD ET AL., J. MOL. CELL CARDIOL., vol. 29, 1997, pages 2585
HATTORI ET AL., J MED CHEM, vol. 47, 2004, pages 4151
HELLER ET AL.: "Inactivation of the Poly(ADP-Ribose)Polymerase Gene Affects Oxygen Radical and Nitric Oxide Toxici in Islet Cells", J. BIOL. CHEM., vol. 270, no. 19, May 1995 (1995-05-01), pages 11176 - 80, XP002924678, DOI: doi:10.1074/jbc.270.19.11176
JAGTAP; SZAB6, NATURE, vol. 4, 2005, pages 421
JOURNAL OFMEDICINAL CHEMISTRY, vol. 44, 2001, pages 1313 - 1333
LO ET AL., STROKE, vol. 29, 1998, pages 830
MENEAR ET AL., J MED CHEM, vol. 51, 2008, pages 6581
SCOTT ET AL., ANN. NEUROL., vol. 45, 1999, pages 120
TAKAHASHI ET AL., BRAIN RES., vol. 829, 1997, pages 46
TENTORI ET AL., BLOOD, vol. 99, 2002, pages 2241
THIEMERMANN ET AL., PROC. NAT. ACAD. SCI., vol. 94, 1997, pages 679
TMZ. MIKNYOCZKI ET AL., MOL CANCER THER, vol. 2, 2003, pages 371
TOKIME ET AL., J. CEREB. BLOOD FLOW METAB., vol. 18, 1998, pages 991
WANG, GENES DEV., vol. 9, 1995, pages 509
WHALEN ET AL., J. CEREB. BLOOD FLOW METAB., vol. 19, 1999, pages 835
YANG ET AL., SHOCK, vol. 13, 2000, pages 60
ZHANG ET AL., SCIENCE, vol. 263, 1994, pages 687
ZINGARELLI ET AL., CARDIOVASCULAR RESEARCH, vol. 36, 1997, pages 205

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10995078B2 (en) 2013-03-13 2021-05-04 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10472342B2 (en) 2013-03-13 2019-11-12 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10800750B2 (en) 2013-03-13 2020-10-13 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10457655B2 (en) 2013-03-13 2019-10-29 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10450286B2 (en) 2013-03-13 2019-10-22 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US9682973B2 (en) 2013-09-13 2017-06-20 Ildong Pharm Co., Ltd Phtalazinone derivatives and manufacturing process thereof
US9844550B2 (en) 2013-09-13 2017-12-19 Ildong Pharm Co., Ltd Phtalazinone derivatives and manufacturing process thereof
USRE49338E1 (en) 2013-09-13 2022-12-20 Idience Co., Ltd. Phthalazinone derivatives and manufacturing process thereof
EP3030557A4 (en) * 2013-09-13 2017-03-22 Ildong Pharm Co., Ltd. A novel phtalazinone derivatives and manufacturing process thereof
WO2015179559A3 (en) * 2014-05-21 2016-06-09 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
US10093630B2 (en) 2014-05-21 2018-10-09 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
US10385057B2 (en) 2015-11-20 2019-08-20 Lundbeck La Jolla Research Center, Inc. Pyrazole compounds and methods of making and using same
US10323038B2 (en) 2015-11-20 2019-06-18 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
US10519134B2 (en) 2015-11-20 2019-12-31 Lundbeck La Jolla Research Center, Inc. Pyrazole compounds and methods of making and using same
US10583137B2 (en) 2015-12-02 2020-03-10 The Scripps Research Institute Triazole DAGLα inhibitors
CN109843885A (zh) * 2016-10-14 2019-06-04 上海汇伦生命科技有限公司 抗肿瘤杂环并咪唑类化合物的药用盐
CN109843885B (zh) * 2016-10-14 2022-03-29 上海汇伦生物科技有限公司 抗肿瘤杂环并咪唑类化合物的药用盐
CN107082764B (zh) * 2017-05-23 2018-11-02 上海锐聚恩新药研发有限公司 一类多羟基酞嗪酮化合物、其制备方法及应用
CN107082764A (zh) * 2017-05-23 2017-08-22 上海锐聚恩新药研发有限公司 一类多羟基酞嗪酮化合物、其制备方法及应用
US10927105B1 (en) 2017-05-23 2021-02-23 Lundbeck La Jolla Research Center, Inc. Pyrazole MAGL inhibitors
US10266497B2 (en) 2017-05-23 2019-04-23 Abide Therapeutics, Inc. Pyrazole MAGL inhibitors
US11149037B2 (en) 2017-05-23 2021-10-19 H. Lundbeck A/S Pyrazole MAGL inhibitors
US11655217B2 (en) 2017-05-23 2023-05-23 H. Lundbeck A/S Pyrazole MAGL inhibitors
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11299484B2 (en) 2018-10-10 2022-04-12 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11267805B2 (en) 2018-10-29 2022-03-08 Forma Therapeutics, Inc. Solid forms of (4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl) piperazine-1-yl)(1-hydroxycyclopropyl)methanone
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
US11390608B2 (en) 2020-04-21 2022-07-19 Idience Co., Ltd. Crystalline forms of phthalazinone compound
US11691964B2 (en) 2020-04-21 2023-07-04 Ildong Pharmaceutical Co., Ltd. Crystalline forms of phthalazinone compound

Also Published As

Publication number Publication date
EP2799435A4 (en) 2016-06-15
EP2799435A1 (en) 2014-11-05
US20150051211A1 (en) 2015-02-19
US9718787B2 (en) 2017-08-01
CN103130723A (zh) 2013-06-05
JP5820081B2 (ja) 2015-11-24
JP2015500212A (ja) 2015-01-05
CN103130723B (zh) 2015-01-14
CA2857405C (en) 2015-10-06
US9187430B2 (en) 2015-11-17
EP2799435B1 (en) 2018-03-28
US20150368205A1 (en) 2015-12-24
CA2857405A1 (en) 2013-06-06

Similar Documents

Publication Publication Date Title
WO2013078771A1 (zh) 一种多聚(adp-核糖)聚合酶抑制剂
TWI553005B (zh) 經取代之酞-1(2h)-酮衍生物
RU2719428C2 (ru) Индазольные соединения в качестве ингибиторов киназы fgfr, их получение и применение
KR102318727B1 (ko) 1,4-이치환된 피리다진 유사체 및 smn-결핍-관련 상태를 치료하기 위한 그의 용도
JP6466171B2 (ja) 新規アミン誘導体またはその塩
CN102264727B (zh) 哒嗪酮衍生物
CN103153980B (zh) 吡唑并苯基苯磺酰胺化合物的衍生物及其作为抗肿瘤药的用途
CN109790169A (zh) 具有作为usp30抑制剂活性的氰基吡咯烷衍生物
CA2993096C (en) Fused ring pyrimidine compound, intermediate, and preparation method, composition and use thereof
HUE030752T2 (en) Therapeutic compounds and related application procedures
WO2016169504A1 (zh) 稠环嘧啶氨基衍生物﹑其制备方法、中间体、药物组合物及应用
EP3626718A1 (en) Five- and six-membered aza-aromatic compound, preparation method therefor, pharmaceutical composition, and application
ES2564961T3 (es) Compuestos de 1H-indazol-3-carboxamida como inhibidores de la glucógeno sintasa cinasa 3 beta
TW201315727A (zh) 尿嘧啶衍生物及其醫藥用途
TW201443010A (zh) 環烷基甲酸類衍生物、其製備方法及其在醫藥上的應用
CN113454081A (zh) 咪唑并吡啶基化合物及其用于治疗增生性疾病的用途
WO2005023782A1 (ja) 置換された縮環ピリミジン-4(3h)-オン化合物
KR102286526B1 (ko) 헤테로시클릭-이미다졸계 화합물, 그 약물 조성물 및 그 제조방법과 용도
WO2004009556A1 (ja) 4−置換アリール−5−ヒドロキシイソキノリノン誘導体
RU2606635C2 (ru) Новое соединение, проявляющее ингибиторную активность в отношении parp
JP5812541B2 (ja) 8−ヒドロキシキノリン誘導体
WO2018045182A1 (en) Disubstituted and trisubtituted 1,2,3-triazoles as wnt inhibitors
JP2018087173A (ja) 悪性脳腫瘍治療薬
KR101778938B1 (ko) 신규 퀴놀린 화합물 및 이를 포함하는 암의 예방 또는 치료용 약학 조성물
WO2008074253A1 (fr) Dérivés de pyridazinoindole: préparation et usage thérapeutique

Legal Events

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

Ref document number: 12852659

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2014543747

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2857405

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 14361698

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012852659

Country of ref document: EP