WO2015131856A1 - 一类杂环化合物、其制备方法和用途 - Google Patents

一类杂环化合物、其制备方法和用途 Download PDF

Info

Publication number
WO2015131856A1
WO2015131856A1 PCT/CN2015/073854 CN2015073854W WO2015131856A1 WO 2015131856 A1 WO2015131856 A1 WO 2015131856A1 CN 2015073854 W CN2015073854 W CN 2015073854W WO 2015131856 A1 WO2015131856 A1 WO 2015131856A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
piperazin
ethyl
alkyl
alkoxy
Prior art date
Application number
PCT/CN2015/073854
Other languages
English (en)
French (fr)
Inventor
蒋华良
王震
李剑峰
张容霞
何洋
柳永建
毕明浩
刘正
田广辉
陈伟铭
杨飞瀑
吴春晖
王瑜
蒋翔锐
尹敬敬
王贯
沈敬山
Original Assignee
中国科学院上海药物研究所
苏州旺山旺水生物医药有限公司
山东特珐曼药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 中国科学院上海药物研究所, 苏州旺山旺水生物医药有限公司, 山东特珐曼药业有限公司 filed Critical 中国科学院上海药物研究所
Priority to JP2016556278A priority Critical patent/JP6395850B2/ja
Priority to CA2941771A priority patent/CA2941771C/en
Priority to RU2016137169A priority patent/RU2667498C2/ru
Priority to CN201580008897.5A priority patent/CN106132956B/zh
Priority to AU2015226578A priority patent/AU2015226578B2/en
Priority to EP15757771.9A priority patent/EP3115361B1/en
Priority to KR1020167027945A priority patent/KR101840249B1/ko
Priority to US15/124,264 priority patent/US10174011B2/en
Publication of WO2015131856A1 publication Critical patent/WO2015131856A1/zh

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/5381,4-Oxazines, e.g. morpholine ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • 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
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • C07D417/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention belongs to the field of medicinal chemistry.
  • the present invention relates to a heterocyclic compound represented by the formula (I), a stereoisomer thereof or a pharmaceutically acceptable salt thereof, a process for the preparation thereof, a pharmaceutical composition and a medicament for the preparation of a central nervous system disorder Application in medicine.
  • Psychosis is mainly a group of neurological diseases mainly characterized by disorders in behavior and psychological activities.
  • the clinical manifestations are abnormal mental activities, which are manifested as perception, thinking, attention, memory, emotion, behavior and will intelligence and consciousness. Aspects of varying degrees of barriers.
  • the dopamine system regulates physiological processes such as exercise, emotion, reward, and cognition.
  • 5-HT 5-HT
  • 5-HT is also involved in the regulation of various physiological functions, such as thermoregulation, emotional activity, pain, and sleep-wake. Therefore, most of the existing drugs in the central nervous system are related to the regulation of central nervous system transmitters such as dopamine and 5-HT.
  • D 2 antagonists are classic antipsychotics and are also used in the treatment of insomnia; 5-HT 2A receptor antagonism reduces EPS, improves negative symptoms, improves cognitive impairment, improves depressive symptoms, improves anxiety and improves insomnia Role (European Journal of Pharmacology, 2000, 407: 39-46). However, simple D 2 antagonists or D 2 antagonist/5-HT 2A antagonists still have varying degrees of side effects.
  • 5-HT 1A receptor agonists have shown good clinical application prospects in the treatment of major depression, anxiety, depression, and improvement of negative symptoms and cognitive function in patients with schizophrenia (CNS Drugs, 2013 Sep, 27: 703- 16).
  • the 5-HT 1A receptor agonist BAY-3702 has neuroprotective, anxiolytic and antidepressant effects in animal models (European Journal of Pharmacology, 1998, 357: 1-8); gepirone can be used to alleviate certain Primary depressive disease (US4771053), such as major depression, endogenous depression, and atypical depression; buprenorphone can treat various symptoms associated with attention deficit and hyperactivity disorder (ADHD), and D
  • the combination of 2 receptor agonists and 5-HT 1A receptor agonists can effectively treat ADHD and Parkinson's disease (WO200016777A); ixabepilone can effectively treat Alzheimer's disease or Parkinson's disease by improving memory. Cognitive decline in the disease (US 5,824,680).
  • Dopamine receptor partial agonists can improve positive symptoms of schizophrenia, negative symptoms, anxiety and depression, cognitive deficits, and rarely cause an increase in serum prolactin like D 2 antagonist, D 2 antagonist/5-HT 2A antagonism Adverse reactions such as weight gain and metabolic abnormalities caused by the agent are safe and well tolerated.
  • drugs with DA/5-HT multi-target effect are beneficial for better regulating the balance of multiple receptors in the brain and regulating the DA/5-HT system to treat diseases in the central nervous system.
  • the present invention provides a class of compounds having 5-HT 2A and/or D 2 receptor and/or 5-HT 1A multi-target action activity, which can be used for the treatment of central nervous system diseases, especially depression and bipolar disorder. , schizophrenia, anxiety, phobia, autism, Alzheimer's disease, bipolar disorder, snoring, obsessive-compulsive disorder, ADHD and other diseases.
  • the object of the present invention is to provide a class of antagonistic effects against serotonin 2A (5-HT 2A ) receptors and/or good for dopamine D 2 receptors and/or serotonin 1A (5-HT 1A ) receptors. Active compound.
  • An object of the present invention is to provide a heterocyclic compound represented by the formula (I), a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
  • Another object of the invention is to provide a process for the preparation of the compounds of the invention.
  • Still another object of the present invention is to provide a use of the heterocyclic compound represented by the general formula (I), a stereoisomer thereof or a pharmaceutically acceptable salt thereof for the preparation of a medicament for preventing and/or treating a central nervous system disease.
  • a further object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the formula (I) according to the present invention, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
  • A, B and D are each independently C or N, and when A is N, D is also N;
  • E is CH, N or C; when E is CH or N, it is connected to E Represents a single key; when E is C, it is connected to E Represents a double bond;
  • a carbamoyl group (-CONH 2 ), a carbamoyl group substituted by a C1
  • R 2 is absent, or 1 to 3 are each independently selected from the group consisting of halogen, hydroxy, decyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C1-C6 alkylthio, C1-C6 alkyl , halogenated C1-C6 alkyl, nitro, amino, amino substituted by C1-C6 alkyl, cyano, carboxyl, aldehyde, amino C1-C6 alkyl, hydroxy C1-C6 alkyl, cyano C1 ⁇ C6 alkyl group, C1-C6 alkanoyl group, halogenated C1-C6 alkanoyl group, sulfonic acid group (-SO 2 OH), sulfonylamino group (-SO 2 NH 2 ), carbamoyl group (-CONH 2 ), C1 -C6 alkyl-substituted carbamoyl group, carboxy C1-
  • R 3 is hydrogen or 1 to 4 substituents each independently selected from a hydroxyl group or a C1 to C6 alkyl group; preferably hydrogen or 1 to 4 substituents each independently selected from a hydroxyl group or a C1 to C4 alkyl group; Preferred is hydrogen or from 1 to 4 substituents each independently selected from hydroxy, methyl or ethyl;
  • the G ring is a monocyclic heterocyclic group or a bicyclic heterocyclic group
  • the bicyclic heterocyclic group is a benzomonocyclic heterocyclic group, a cycloalkyl-heterocyclic heterocyclic group or a monocyclic heterocyclic-monocyclic heterocyclic group.
  • the monocyclic heterocyclic group contains at least one hetero atom selected from N, S and O; preferably a 3- to 10-membered monocyclic heterocyclic group, a benzo[3- to 10-membered monocyclic heterocyclic group], [C3 -C10 cycloalkyl]][3 to 10 membered monocyclic heterocyclic group] and [3 to 10 membered monocyclic heterocyclic group] [3 to 10 membered monocyclic heterocyclic group]; more preferably 5 to 7 membered single Cycloheterocyclyl, benzo[5-7-membered monocyclic heterocyclyl], [C5-C7 cycloalkyl]-[5-7-membered monocyclic heterocyclyl] and [5-7-membered monocyclic heterocyclyl] And [5 to 7-membered monocyclic heterocyclic group]; more preferably furyl, dihydrofuranyl, tetrahydrofuranyl, thienyl, dihydrothien
  • the G ring is connected to L by a carbon atom on the G ring and is connected to L;
  • G ring is optionally substituted with one or more identical or different substituents
  • the substituent on the G ring is halogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, nitro, cyano, hydroxy, decyl Amino group, amino group substituted by C1-C6 alkyl group, azide group, C1-C6 alkanoyl group, halogenated C1-C6 alkanoyl group, C2-C6 alkenyl group, C2-C6 alkynyl group, carboxy C1-C6 alkyl group a cyano C1-C6 alkyl group, a C2-C6 alkenyloxy group, a C2-C6 alkynyloxy group, a carbamoyl group (-CONH 2 ), a carbamoyl group substituted with a C1 to C6 alkyl group, a carboxyl group, and a hydroxyl group C1 to C6
  • heterocyclic compound represented by the formula (I) of the present invention is selected from the following compounds:
  • heterocyclic compound represented by the formula (I) of the present invention is selected from the compounds shown below:
  • R 1 , R 2 , R 3 , L and G rings are as defined and preferred above.
  • heterocyclic compounds represented by the formula (I) stereoisomers thereof or pharmaceutically acceptable salts thereof, the following compounds or pharmaceutically acceptable salts thereof are most preferred:
  • the present invention provides a process for producing a heterocyclic compound represented by the general formula (I), which can be carried out by one of the following methods 1-5.
  • Process 1 The compound of the formula (II) or a salt thereof is subjected to an N-alkylation reaction with a compound of the formula (III) or a salt thereof, as shown in the reaction formula 1:
  • G ring, L, A, B, D, E, R 1 , R 2 and R 3 are as defined above and preferred;
  • X represents a leaving group such as halogen, C1-C6 alkylsulfonyloxy, benzenesulfonyloxy, naphthalenesulfonyloxy, the above C1-C6 alkylsulfonyloxy, benzenesulfonyloxy, naphthalene
  • the sulfonyloxy group may be optionally further substituted with one or more groups selected from the group consisting of halogen, C1-C6 alkyl, C1-C6 alkoxy, nitro, hydroxy, amino and C1-C6 alkanoyl;
  • the N-alkylation reaction between the compound of the formula (II) or a salt thereof and the compound of the formula (III) or a salt thereof is carried out in the absence of a solvent, or in the presence or absence of a base In the solvent.
  • the solvent includes water; ethers such as dioxane, tetrahydrofuran, diethyl ether, methyl tert-butyl ether, diisopropyl ether, diglyme, ethylene glycol dimethyl ether, etc.; aromatic hydrocarbons, Such as: benzene, toluene, xylene, nitrobenzene, chlorobenzene, etc.; alcohols, such as: methanol, ethanol, isopropanol, butanol, tert-butanol, ethylene glycol; ketones, such as acetone, methyl ethyl ketone, 4 -methyl-2-pentanone, etc.; amides such as N,N-d
  • the base may be selected from an inorganic base or an organic base, and the inorganic base includes an alkali metal hydroxide such as sodium hydroxide, potassium hydroxide, barium hydroxide, lithium hydroxide; an alkali metal carbonate such as sodium carbonate, Potassium carbonate, cesium carbonate, lithium carbonate; alkali metal hydrogen carbonate, such as: sodium hydrogencarbonate, potassium hydrogencarbonate, lithium hydrogencarbonate; alkali metals, such as: potassium, sodium; others, such as: sodium amide, potassium amide, sodium hydride Potassium hydride; organic bases include sodium methoxide, sodium ethoxide, potassium methoxide, potassium ethoxide, sodium acetate, triethylamine, pyridine, diisopropylamine, diisopropylethylamine, tripropylamine, diethylamine, pyrimidine, Quinoline, piperidine, piperazine, imidazole, dimethylaminopyridine, trimethylamine
  • the above reaction can be carried out by adding an alkali metal iodide such as potassium iodide and sodium iodide as a reaction accelerator.
  • an alkali metal iodide such as potassium iodide and sodium iodide
  • the above reaction is usually carried out at room temperature to 200 ° C, preferably at room temperature to 150 ° C, and the reaction is usually carried out in about 1 to 30 hours, preferably within 5 to 20 hours.
  • Process 2 A compound represented by the formula (IV) or a salt thereof is subjected to a coupling reaction with a compound represented by the formula (V) or a salt thereof, as shown in the reaction formula 2:
  • G ring, L, A, B, D, R 1 , R 2 and R 3 are as defined and preferred above, and E 1 represents a nitrogen atom;
  • X 1 represents a halogen or a trifluoromethanesulfonyloxy group, preferably bromine, Iodine, chlorine or trifluoromethanesulfonyloxy;
  • the coupling reaction is carried out in the presence of palladium catalysis and a base, and the obtained compound of the formula (Ia) is a specific example of the compound of the formula (I).
  • the palladium catalyst is palladium acetate (Pd(OAc) 2 ), bis(triphenylphosphine)palladium dichloride ((Ph 3 P) 2 PdCl 2 ), bis(benzonitrile)palladium chloride ((PhCN)) 2 PdCl 2 ), tetrakis(triphenylphosphine)palladium (Pd(PPh 3 ) 4 ), bis(triphenylphosphine)palladium acetate ((Ph 3 P) 2 Pd(OAc) 2 ), 1,2-di (diphenylphosphino)ethane palladium dichloride ((PdCl 2 (dppe) 2 )), bis(1,2-bis(diphenylphosphino)ethane)palladium (Pd(dppe) 2 ), double (dibenzylideneacetone) palladium (Pd(dba) 2 ), tris(dibenzylideneacetone
  • the reaction solvent is not particularly limited as long as it does not interfere with the reaction, including water; ethers such as dioxane, tetrahydrofuran, etc.; aromatic hydrocarbons such as toluene, xylene, etc.; alcohols such as: uncle Butanol or the like; ketones such as acetone; amides such as N,N-dimethylformamide; others such as dimethyl sulfoxide, acetonitrile, etc.; or a mixture of the above solvents; if necessary
  • the above reaction can be carried out by adding a suitable ligand as a reaction accelerator.
  • Suitable ligands are 2,2'-diphenylphosphino-1,1'-binaphthyl (BINAP), tri-tert-butyl (P(t-Bu) 3 ), 1,1'-di-(diphenyl) Phosphyl)ferrocene (dppf), 2-dicyclohexylphosphine-2,4,6-triisopropylbiphenyl (x-phos), 4,5-bisdiphenylphosphine-9,9-di Xantphos, tri-tert-butylphosphine tetrafluoroborate and tris(2-methylphenyl)phosphine (P(o-tolyl) 3 ).
  • BINAP 2,2'-diphenylphosphino-1,1'-binaphthyl
  • P(t-Bu) 3 tri-tert-butyl
  • dppf 1,1'-di-(diphenyl) Phosphyl
  • Process 3 Amidation reaction of a compound of the formula (VI) or a salt thereof with a compound of the formula (III) or a salt thereof to obtain a formula (VII) A compound or a salt thereof; a compound of the formula (VII) or a salt thereof treated with a reducing agent to give a compound of the formula (I), as shown in the reaction formula 3:
  • G ring, L, A, B, D, E, R 1 , R 2 and R 3 are as defined above and preferred.
  • the amidation reaction can be carried out in two ways.
  • the first amidation process is the first activation of the carboxyl group of the compound of formula (VI) with or without a catalyst followed by an aminolysis reaction with a compound of formula (III).
  • the activator may be selected from the group consisting of thionyl chloride, oxalyl chloride, brominated sulfoxide, phosphorus oxychloride, phosphorus pentachloride, pivaloyl chloride, ethyl chloroformate, isobutyl chloroformate, carbonyl diimidazole.
  • the catalyst may be one or more selected from the group consisting of N,N-dimethylformamide (DMF), diethylaniline, dimethylaniline, N-methylmorpholine and the like.
  • DMF N,N-dimethylformamide
  • the reaction solvent for activating the carboxyl group is not particularly limited as long as it does not interfere with the reaction, and may be, for example, selected from the group consisting of dichloromethane, dichloroethane, dimethyl sulfoxide, tetrahydrofuran, benzene, toluene, chloroform, carbon tetrachloride.
  • the aminolysis reaction is carried out in the presence of a base in a suitable solvent.
  • the base may be selected from the group consisting of pyridine, piperidine, pyrrolidine, morpholine, N-methylmorpholine, quinoline, 4-dimethylaminopyridine (DMAP), triethylamine, diethylamine, tri-n-butyl Amine, tripropylamine, diisopropylamine, diisopropylethylamine, sodium methoxide, sodium ethoxide, potassium t-butoxide, butyl lithium, 1,8-diaza heterocycle [5,4,0] Undecene-7 (DBU), N-methylmorpholine, quinoline, 4-dimethylaminopyridine (DMAP), sodium hydrogen, sodium hydroxide, potassium hydroxide, lithium hydroxide, sodium hydrogencarbonate, hydrogencarbonate One or more of potassium, sodium carbonate and potassium carbonate.
  • DMAP dimethylaminopyridine
  • the reaction solvent of the aminolysis reaction may be selected from the group consisting of an aromatic hydrocarbon solvent, an ether solvent, a halogenated hydrocarbon solvent, other solvents, and a combination thereof.
  • the aromatic hydrocarbon solvent may be, for example, one or more selected from the group consisting of benzene, toluene, xylene, and the like; and the ether solvent may be, for example, selected from tetrahydrofuran (THF), diethyl ether, and methyl tert-butyl ether.
  • the halogenated hydrocarbon solvent may be, for example, selected from dichloro One or more of methane, chloroform, carbon tetrachloride, dichloroethane, etc.; the other solvent may be, for example, selected from the group consisting of methanol, ethanol, ethylene glycol, n-hexane, cyclohexane, N, N- One or more of dimethylformamide (DMF), N,N-dimethylacetamide, dimethyl sulfoxide (DMSO), N-methylpyrrolidone, acetone, acetonitrile, ethyl acetate, etc., but The present invention is not limited to the above solvents.
  • the reaction temperature of the aminolysis reaction is not particularly limited, but is preferably between -30 ° C and 150 ° C, more preferably between -10 ° C and 120 ° C.
  • the reaction time of the aminolysis reaction is not particularly limited, but is preferably between 10 minutes and 24 hours.
  • the second amidation process is carried out using a condensing agent, with or without a catalyst, in the presence or absence of a base.
  • the condensing agent may be selected from the group consisting of N,N'-dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCl), N , N'-diisopropylcarbodiimide (DIC), O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate (TBTU), O-( 7-azobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU), benzotriazole-N,N,N',N'-four Methylurea hexafluorophosphate (HBTU), benzotriazol-1-yloxytris(di
  • the catalyst may be one or more selected from the group consisting of 1-hydroxy-benzotriazole (HOBt), 4-dimethylaminopyridine (DMAP) and the like.
  • the base may be selected from the group consisting of triethylamine, diethylamine, tri-n-butylamine, tripropylamine, diisopropylamine, diisopropylethylamine, trimethylamine, pyridine, 2,6-dimethyl Pyridine, 4-dimethylaminopyridine, piperidine, pyrrolidine, quinoline, morpholine, N-methylmorpholine, N-ethylmorpholine, diisopropylamine, diisopropylethylamine, 1, One of 8-diazahetero[5,4,0]undecene-7 (DBU) or 1,5-diazabicyclo[4.3.0]-indol-5-ene (DBN) Or several.
  • DBU 8-diazahetero
  • the reaction solvent of the second amidation method may be selected from the group consisting of hydrocarbons such as benzene, xylene, toluene, dichloromethane or chloroform; ethers such as tetrahydrofuran, diethyl ether, propyl ether or 1,4-dioxane.
  • hydrocarbons such as benzene, xylene, toluene, dichloromethane or chloroform
  • ethers such as tetrahydrofuran, diethyl ether, propyl ether or 1,4-dioxane.
  • the reaction temperature of the second amidation method is not particularly limited as long as it does not affect the progress of the reaction, but is preferably between -20 ° C and 80 ° C, more preferably between 0 ° C and 50 ° C.
  • the reducing agent is borane, hydrogen/palladium carbon, lithium tetrahydrogen aluminum, sodium triacetoxyborohydride, diisobutylaluminum hydride, boron trifluoride, boron tribromide, sodium borohydride and potassium borohydride. One or several of them.
  • G ring, L, A, B, D, E, R 1 , R 2 and R 3 are as defined above and preferred.
  • the reductive amination reaction is carried out in the presence of a reducing agent including, but not limited to, sodium borohydride, potassium borohydride, sodium triacetoxyborohydride (NaBH(OAc) 3 ), tetramethyltriacetyl Ammonium borohydride, sodium cyanoborohydride, and the like.
  • a reducing agent including, but not limited to, sodium borohydride, potassium borohydride, sodium triacetoxyborohydride (NaBH(OAc) 3 ), tetramethyltriacetyl Ammonium borohydride, sodium cyanoborohydride, and the like.
  • the reaction solvent is selected from the group consisting of waters; ethers such as dioxane, tetrahydrofuran, diethyl ether, methyl tert-butyl ether, Diisopropyl ether, diglyme, ethylene glycol dimethyl ether, etc.; aromatic hydrocarbons, such as: benzene, toluene, xylene, nitrobenzene, chlorobenzene, etc.; alcohols, such as: methanol, ethanol, Isopropanol, butanol, tert-butanol, ethylene glycol; ketones such as acetone, methyl ethyl ketone, 4-methyl-2-pentanone, etc.; amides such as N,N-dimethylformamide, N,N-dimethylacetamide, 1-methyl-2-pyrrolidone, etc.; halogenated hydrocarbons, such as: chloroform, dichloromethane, dichloroethane,
  • the compound of the formula (I) obtained by the methods 1 to 4 is subjected to functional group conversion, such as by an oxidation reaction, a Grignard reaction, a hydrolysis reaction, a fluorination reaction, a chlorination reaction or a thio reaction.
  • functional group conversion such as by an oxidation reaction, a Grignard reaction, a hydrolysis reaction, a fluorination reaction, a chlorination reaction or a thio reaction.
  • the oxidation reaction is carried out in the presence of an oxidizing agent including, but not limited to, Dess-Martin oxidant, Jones reagent, Swern reagent (DMSO and oxalyl chloride), and 2,3-dichloro-5,6-dicyanide pair. Benzoquinone (DDQ), etc.
  • an oxidizing agent including, but not limited to, Dess-Martin oxidant, Jones reagent, Swern reagent (DMSO and oxalyl chloride), and 2,3-dichloro-5,6-dicyanide pair.
  • DDQ Benzoquinone
  • the Grignard reaction is carried out in the presence of a Grignard reagent including, but not limited to, methyl magnesium chloride, methyl magnesium bromide, methyl magnesium iodide, and ethyl magnesium bromide.
  • a Grignard reagent including, but not limited to, methyl magnesium chloride, methyl magnesium bromide, methyl magnesium iodide, and ethyl magnesium bromide.
  • the hydrolysis reaction can be carried out in the presence of an acid or a base including, but not limited to, hydrochloric acid, sulfuric acid, sodium hydroxide, potassium hydroxide, and the like.
  • the fluorination reaction is carried out in the presence of a fluorinating agent including, but not limited to, diethylaminosulfur trifluoride (DAST), sulfur tetrafluoride, and iodine pentafluoride.
  • a fluorinating agent including, but not limited to, diethylaminosulfur trifluoride (DAST), sulfur tetrafluoride, and iodine pentafluoride.
  • the chlorination reaction is carried out in the presence of a chlorinating agent including, but not limited to, thionyl chloride, phosphorus pentachloride, and N-chlorosuccinimide (NCS).
  • a chlorinating agent including, but not limited to, thionyl chloride, phosphorus pentachloride, and N-chlorosuccinimide (NCS).
  • the thiolation reaction is carried out in the presence of a thiol reagent including, but not limited to, phosphorus pentasulfide and Lawson's reagent, and the like.
  • a thiol reagent including, but not limited to, phosphorus pentasulfide and Lawson's reagent, and the like.
  • the compound of formula (II), formula (III), formula (IV), formula (V), formula (VI), formula (VIII) is a commercially available compound or is prepared according to methods known in the art or similar Preparation of a synthetic method of the compound.
  • the starting compound used in each of the above reaction formulas may be a suitable salt
  • the suitable salts include alkali metal salts and alkaline earth metal salts such as sodium salts, potassium salts, calcium salts, magnesium salts and the like; and organic base salts such as pyridinium salts.
  • inorganic acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate, nitrate, phosphate, etc.
  • organic acid salts such as formate, acetate, propionate , glycolate, oxalate, malonate, succinate, fumarate, maleate, lactate, malate, citrate, tartrate, picrate, Glutamate, methanesulfonate and besylate;
  • the starting compounds used in the above respective reaction formulas may include a solvate form such as a hydrate, an alcoholate or the like.
  • heterocyclic compound represented by the formula (I) of the present invention and stereoisomers thereof also include solvate forms thereof such as hydrates, alcoholates and the like, and the solvates are included in the scope of the present invention. Inside.
  • heterocyclic compound represented by the formula (I) of the present invention and a pharmaceutically acceptable salt thereof as the stereoisomer thereof mean that the heterocyclic compound represented by the formula (I) or a stereoisomer thereof is treated with a suitable acid They are converted into therapeutically active non-toxic addition salt forms.
  • the salt such as hydrochloride, hydrobromide, hydroiodide, sulfate or hydrogen sulfate, nitrate, phosphate or acid phosphate, perchlorate, formate, acetate, Trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, succinate, glutarate, maleate, fumarate, milk Acid salt, malate, citrate, tartrate, picrate, glutamate, benzoate, methanesulfonate, ethanesulfonate, besylate, p-toluenesulfonate, water Salicylate, ascorbate, camphorate or camphor sulfonate.
  • solvates are included in the scope of the present invention.
  • examples of the solvate are, for example, hydrates, alcoholates and the like.
  • Each of the target compounds obtained from the respective reaction formulas can be isolated and purified from the reaction mixture by, for example, the reaction mixture is subjected to cooling, followed by filtration, extraction or concentration to separate the crude product, followed by a conventional column. Purification is carried out by chromatography, beating or recrystallization.
  • the present invention also provides the use of the heterocyclic compound of the formula (I) according to the present invention, a stereoisomer thereof or a pharmaceutically acceptable salt thereof for the preparation of a medicament for preventing and/or treating a central nervous system disease.
  • the present invention also provides a method for treating and/or preventing a central nervous system disease, which comprises administering to a human or an animal a heterocyclic compound represented by the above formula (I) of the present invention, a stereoisomer thereof or A pharmaceutically acceptable salt thereof.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the heterocyclic compound represented by the above formula (I), a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, and optionally a pharmaceutically acceptable carrier .
  • the pharmaceutical composition can be used to treat or prevent diseases of the central nervous system.
  • the present invention also provides a process for preparing a pharmaceutical composition
  • a process for preparing a pharmaceutical composition comprising mixing a heterocyclic compound represented by the above formula (I), a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier.
  • a plurality of pharmaceutical preparation forms can be selected according to the purpose of treatment, and generally include: tablets, pills, capsules, granules, suspensions, solutions, creams, ointments, powders, suppositories, and qi. Inhalation, injections, etc.
  • the above central nervous system diseases are selected from: schizophrenia; difficult to control, refractory or chronic schizophrenia; emotional disorders; mental disorders; emotional disorders; type I bipolar affective disorder; type II bipolar affective disorder; Intrinsic depression; major depression; uncontrollable depression; emotional disorders; circulatory affective disorder; panic attacks; panic disorder; social phobia; obsessive-compulsive and behavioral disorders; impulsive disorders; Post-stress disorder; anxiety disorder; acute stress disorder; rickets; anorexia nervosa; sleep disorders; adaptive disorders; cognitive impairment; autism; neuropathic headache; mania; Parkinson's disease; Huntington's disease Alzheimer's disease; various dementias; memory disorders; ADHD; attention deficit/hyperthyroidism and tic disorder.
  • halogen generally means fluorine, chlorine, bromine and iodine; preferably fluorine, chlorine or bromine; more preferably fluorine or chlorine;
  • the C1-C6 alkyl group means a straight or branched saturated hydrocarbon group having 1 to 6 carbon atoms, for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl. , sec-butyl, n-pentyl, 1-ethylpropyl, isopentyl, neopentyl, isohexyl, 3-methylpentyl or n-hexyl, etc., preferably methyl, ethyl, n-propyl, Isopropyl, butyl, isobutyl or tert-butyl;
  • the halogenated C1-C6 alkyl group means that a hydrogen atom of a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms is substituted by one or more halogen atoms which may be the same or different, for example, a trifluoromethyl group or a fluorine group.
  • Base difluoromethyl, chloromethyl, bromomethyl, dichlorofluoromethyl, chloroethyl, bromopropyl, 2-chlorobutyl or pentafluoroethyl;
  • C1-C6 alkoxy refers to a straight or branched alkoxy group having 1 to 6 carbon atoms, for example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso Butoxy, tert-butoxy, sec-butoxy, n-pentyloxy, isopentyloxy, neopentyloxy, isohexyloxy, 3-methylpentyloxy or n-hexyloxy, etc., preferably methoxy Base, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy or tert-butoxy;
  • Halogenated C1-C6 alkoxy means that a hydrogen atom of a straight or branched alkoxy group having 1 to 6 carbon atoms is substituted by one or more of the same or different halogen atoms, for example, -OCF 3 , -OCH 2 CH 2 Cl, -OCHBrCH 2 Cl or -OCF 2 CF 3 ;
  • the C1-C6 alkylthio group means a straight-chain or branched alkylthio group having 1 to 6 carbon atoms, for example, methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, and iso Butylthio, tert-butylthio, sec-butylthio, n-pentylthio, isopentylthio, neopentylthio or n-hexylthio, etc., preferably methylthio, ethylthio, n-propylthio, iso Propylthio, n-butylthio, isobutylthio or tert-butylthio;
  • the C2-C6 alkenyl group means a linear or branched unsaturated hydrocarbon group having 1 to 3 double bonds and 2 to 6 carbon atoms, and includes both a cis configuration and a trans configuration, for example, a vinyl group, 1-propenyl, 2-propenyl, 1-methyl-1-propenyl, 2-methyl-1-propenyl, 2-methyl-2-propenyl, 1-butenyl, 2-butene , 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1,3-butadienyl, 1,3-pentadienyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 3,3-dimethyl-1-propenyl or 2-ethyl-1- Propylene or the like;
  • C2-C6 alkynyl refers to a straight or branched alkynyl group having 2 to 6 carbon atoms, for example, ethynyl, 2-propynyl, 2-butynyl, 3-butynyl, 1-methyl- 2-propynyl, 2-pentynyl, 2-pentynyl or 2-hexynyl;
  • the phenyl C1-C6 alkyl group means that one carbon atom of a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms is bonded to a carbon atom of a phenyl group such as a benzyl group, a phenethyl group or a phenylpropyl group. ;
  • Phenyl C1-C6 alkoxy means that one carbon atom of a straight or branched alkoxy group having 1 to 6 carbon atoms is bonded to a carbon atom of a phenyl group such as a benzyloxy group, -OCH(CH 3 )Ph , phenylethoxy or phenylpropoxy, etc.;
  • phenyl C1 - C6 alkanoyl means that one carbon atom of a straight or branched alkanoyl group having 1 to 6 carbon atoms is bonded to a carbon atom of a phenyl group, such as benzene Formyl, phenylacetyl or phenylpropionyl;
  • the C2-C6 alkenyloxy group means a linear or branched alkenyloxy group having 1 to 3 double bonds and 2 to 6 carbon atoms, such as B. Alkenyloxy, 1-propenyloxy, 1-methyl-1-propenyloxy, 2-methyl-1-propenyloxy, 1-pentenyloxy, 1,3-pentadienyloxy or 2 -pentenyloxy group;
  • C2-C6 alkynyloxy means a straight-chain or branched alkynyloxy group having 2 to 6 carbon atoms, for example, ethynyloxy, 2-propynyloxy, 2-butynyloxy, 3-butynyloxy , 1-methyl-2-propynyloxy, 2-pentynyloxy or 2-hexynyloxy;
  • the C1-C6 alkanoyl group means a straight or branched alkanoyl group having 1 to 6 carbon atoms, such as formyl, acetyl, propionyl, butyryl, isobutyryl, valeryl, t-butyryl or hexanoyl;
  • halogenated C1 to C6 alkanoyl group means that a hydrogen atom in a straight or branched alkanoyl group having 1 to 6 carbon atoms is substituted by one or more halogen atoms which may be the same or different, for example, a trifluoroacetyl group;
  • a carbamoyl group substituted by a C1-C6 alkyl group means that the hydrogen atom on the carbamoyl group is substituted by one or two C1 to C6 alkyl groups which may be the same or different, for example, -CONHMe, -CONHEt, -CON(Me)Et , -CONEt 2 or -CONMe 2, etc.;
  • the hydroxy C1-C6 alkyl group means that one carbon atom of a linear or branched alkyl group having 1 to 6 carbon atoms is bonded to a hydroxyl group such as -CH 2 OH, -CH 2 CH 2 OH, -CH(OH)CH 3 , -CH 2 CH 2 CH 2 OH, -CH 2 CH 2 CH 2 CH 2 OH or -CH 2 CH(CH 3 )CH 2 OH;
  • Amino C1-C6 alkyl refers to a carbon atom bonded to an amino group of a straight or branched alkyl group having 1 to 6 carbon atoms, such as -CH 2 NH 2 , -CH 2 CH 2 NH 2 , -CH(NH 2 ) CH 3 , -CH 2 CH 2 CH 2 NH 2 or -CH 2 CH 2 CH 2 CH 2 NH 2 , etc.; an amino C1-C6 alkyl group substituted by a C1-C6 alkyl group means that a hydrogen atom on the amino group is 1 or 2 identical or different C1-C6 alkyl groups, such as -CH 2 NHMe or -CH 2 CH 2 NEt 2 ;
  • the carbamoyl C1-C6 alkyl group means that one carbon atom of a linear or branched alkyl group having 1 to 6 carbon atoms is bonded to a carbonyl carbon of a carbamoyl group, for example, -CH 2 CONH 2 , -CH 2 CH 2 CONH 2 , -CH(CONH 2 )CH 3 or -CH 2 CH 2 CH 2 CONH 2 ;
  • the carbamoyl C1 to C6 alkyl group substituted by a C1 to C6 alkyl group means that the amino hydrogen atom on the C1 to C6 alkyl group of the carbamoyl group is substituted by one or two C1 to C6 alkyl groups which are the same or different, for example, CH 2 CONHMe, -CH 2 CH 2 CONHEt, -CH 2 CH 2 CONMe 2 -CH 2 CONEt 2, or the like;
  • the cyano C1-C6 alkyl group means that one carbon atom of a linear or branched alkyl group having 1 to 6 carbon atoms is bonded to a cyano group such as a cyanomethyl group, a 2-cyanoethyl group or a 1-cyano group. Ethyl, 3-cyanopropyl, 4-cyanobutyl or 5-cyanopentyl;
  • the carboxy C1-C6 alkyl group means a carbon atom bonded to a carboxyl group with a straight or branched alkyl group having 1 to 6 carbon atoms, such as a carboxymethyl group, a 2-carboxyethyl group, a 1-carboxyethyl group, or a 3- Carboxypropyl, 4-carboxybutyl or 5-carboxypentyl, etc.;
  • a C1-C6 alkanesulfonyl group means a straight-chain or branched alkylsulfonyl group having 1 to 6 carbon atoms, such as a methylsulfonyl group, an ethylsulfonyl group or a propylsulfonyl group;
  • the halogenated C1 to C6 alkylsulfonyl group means that a hydrogen atom on a linear or branched alkanesulfonyl group having 1 to 6 carbon atoms is substituted by one or more halogen atoms which may be the same or different, for example, a trifluoromethanesulfonyl group. Wait;
  • the amino group substituted by a C1 to C6 alkyl group means that the hydrogen atom on the amino group is substituted by one or two C1 to C6 alkyl groups or C1 to C6 alkanoyl groups which are the same or different, for example, -NHMe or -NEt 2 ;
  • the C3-C10 cycloalkyl group means a saturated or unsaturated cyclic hydrocarbon group having 3 to 10 carbon atoms, such as a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group, a cycloheptyl group, a cyclooctyl group, a cyclobutene group. , cyclohexene, cyclohexadiene, cyclopentene or cyclopentadiene;
  • the 3-10 membered monocyclic heterocyclic group means a monocyclic group containing at least one hetero atom selected from N, S and O, such as ethylene oxide, azetidinyl, furyl, dihydrofuranyl, tetrahydrofuran.
  • the compounds of the invention have good activity against the serotonin 1A (5-HT 1A ) receptor and/or the dopamine D 2 receptor and/or the serotonin 2A (5-HT 2A ) receptor; especially for 5- The HT 1A receptor has high activity, and the EC 50 of some compounds for 5-HT 1A receptor agonism even reaches the level of 1 to 0.1 nM.
  • the compound of the present invention has multi-target action characteristics and acts on multiple subtypes of dopamine/5-hydroxytryptamine receptor, i.e., simultaneously to dopamine D 2 receptor, 5-HT 1A receptor, 5-HT 2A At least three targets such as receptors have high activity; as shown by pharmacological experiments, most of the compounds have D 2 antagonist/5-HT 1A agonistic/5-HT 2A antagonism, or have D 2 partial agonism/5-HT.
  • the compound of the present invention is a central nervous system receptor modulator, thereby reducing or avoiding side effects caused by a single D 2 antagonist or D 2 antagonist/5-HT 2A antagonist, for example, Extrapyramidal adverse reactions (EPS), side effects of metabolic disorders, etc.
  • EPS Extrapyramidal adverse reactions
  • the compound of the present invention is not only active, but also effective orally, has low pharmacological dose, small toxic and side effects, and has curative effect on diseases in the central nervous system, especially for major depression (MDD), anxiety, and schizophrenia. Negative symptoms, cognitive dysfunction, Parkinson's disease, ADHD and other diseases are effective.
  • the compound of the present invention has the advantages of multiple target effects, lower pharmacodynamic dose, less toxic side effects, better safety and tolerance than the existing antipsychotic drugs, and has good clinical application. prospect.
  • step 1
  • step 1
  • step 1
  • the compound 6-c (1.5 g, 7.85 mmol) was dissolved in dichloromethane (60 ml), triethylamine (3 ml, 23.55 mmol) was added with stirring, and methanesulfonyl chloride (0.92 ml, 11.77 mmol) was added dropwise under ice bath. After the addition was completed, the reaction was carried out to room temperature for 3 hours. An appropriate amount of water was added, and the mixture was extracted with dichloromethane.
  • step 1
  • step 1
  • the compound 1-e (900 mg, 2.83 mmol) was placed in a dry three-necked flask, dissolved in tetrahydrofuran (3 ml) under nitrogen atmosphere, cooled to -78 ° C, and poured into a solution of 2.5 M n-butyllithium in n-hexane (1.47 ml). , 3.67mmol), maintain -78 ° C anti It should be 3 hours.
  • N-chlorosuccinimide (677 mg, 5.09 mmol) was dissolved in tetrahydrofuran (3 ml), and slowly poured into the reaction system. After half an hour, the mixture was stirred at room temperature overnight, and the reaction was quenched by adding a saturated ammonium chloride solution. The mixture was extracted with EtOAc (EtOAc m.
  • step 1
  • step 1
  • step 1
  • Lithium tetrahydroaluminum (4.6 g, 121 mmol) was added to a flask containing dry tetrahydrofuran (20 ml), and a solution of compound 12-a (10 g, 53.2 mmol) in tetrahydrofuran (100 ml) was added dropwise in an ice bath.
  • the mixture was stirred at room temperature for 20 minutes, and then heated to reflux. After 3 hours, the reaction was stopped, and sodium sulfate decahydrate was slowly added until the bubbles were no longer discharged, and the filtrate was filtered, dried, and then subjected to column chromatography.
  • the product 12-b (7.8 g, yield: 85%) was obtained.
  • step 1
  • Methyl 2-aminothiophene-3-carboxylate (2.0 g, 12.7 mmol) was dissolved in formamide (60 ml), stirred and heated to 190 ° C for 4 hours. After cooling, it was poured into water (200 ml), and extracted with n-butanol (50 ml ⁇ 4). After concentration, the residue was subjected to EtOAc mjjjjjj ESI-MS (m/z): 153.0 [M+H] + .
  • 1 H-NMR 300 Hz, DMSO-d 6 ): ⁇ ppm 7.39 (d, 1H), 7.57 (d, 1H), 8.12 (s, 1H), 12.49 (brs, 1H).
  • the compound 13-d (900 mg) was dissolved in dichloromethane (10 ml), cooled to 0 ° C, and trifluoroacetic acid (1 ml) was added. The reaction mixture was concentrated to dryness EtOAc.
  • step 1
  • N-N-dimethylformamide (250 ml) and potassium carbonate (51.5 g, 373 mmol) were added to a three-necked flask, and protected with N 2 , and ethyl acetate (13.7 ml) was added dropwise at room temperature.
  • 14-b (25.2g, 124mmol)
  • the reaction was completed overnight
  • the reaction was completed
  • filtered the filtrate was washed once with water, then washed once with saturated sodium chloride solution, dried
  • the organic phase was concentrated to give a crude material. m.j.
  • step 1
  • step 1
  • step 1
  • the benzothiazole-2(3H)-one 20-a (500 mg, 3.307 mmol) was suspended in carbon disulfide (8 ml), and anhydrous aluminum chloride (2.65 g, 19.841 mmol) was added portionwise in an ice bath, and then slowly dripped. Chloroacetyl chloride (324 ul, 4.299 mmol) was added and allowed to stir at room temperature for 10 minutes and then refluxed for 1.5 hours. The reaction mixture was quenched with a small amount of ice water, and the mixture was evaporated to dryness, and the mixture was evaporated. The mixture was evaporated. The mixture was evaporated. Two times, the filter cake was dried to give a pink solid 20-b (740 mg, yield: 98%).
  • step 1
  • 2H-1,4-benzoxazine-3(4H)-one 21-a (500 mg, 3.356 mmol) was suspended in dichloromethane (8 ml), and anhydrous aluminum chloride (895 mg, 6.712mmol), slowly add dropwise chloroacetyl chloride (330ul, 4.363mmol), add it to the mixture at room temperature for 10 minutes, reflux for 5 hours, add a small amount of ice water to the reaction solution to quench the reaction, concentrate to remove dichloromethane , the residue was added with ice water (10 ml), 4N hydrochloric acid (5 ml) was added, and the mixture was stirred at room temperature for 2 hours, filtered, and the filter cake was washed twice with ice water to give a pale yellow solid 21-b (690 mg, yield: 91%) ).
  • step 1
  • step 1
  • Triethylamine (125 ul, 0.87 mmol) was added to a solution of compound 23-a (100 mg, 0.43 mmol) in dichloromethane (5 ml), and methanesulfonyl chloride (51 ul, 0.65 mmol) was added to the above reaction system under ice bath. The reaction was allowed to stand at room temperature for 2 hours, and the reaction mixture was diluted with methylene chloride, washed with saturated aqueous ammonium chloride, and the organic layer was dried, and evaporated to give a white solid (71 mg, yield: 53%).
  • ESI-MS (m/z): 311.1 [M+H] + .
  • Ethyl anthranilate 24-a (1.0 g, 6.62 mmol), acrylonitrile (0.88 ml, 13.24 mmol) was dissolved in dioxane, and hydrogen chloride / dioxane (10 ml) was slowly added in an ice bath. Stir for 4 hours, heat the tube to 80 ° C overnight, then evaporate the solvent, add water, dichloromethane, adjust the pH to 7-8 with ammonia water, filter, solid dry, beat with methyl tert-butyl ether, filter Dry to pale yellow solid (660 mg, yield: 52%).
  • ESI-MS (m/z): 209.1 [M+H] + .
  • step 1
  • Triethylsilane (900 ul, 5.58 mmol) was added dropwise to a solution of compound 25-a (500 mg, 2.32 mmol) in trifluoroacetic acid and allowed to react at 30 ° C overnight.
  • the reaction solution was poured into ice water, extracted with dichloromethane three times, the organic phase was combined, dried, concentrated, and beaten in a mixture of petroleum ether: acetone (30:1, volume ratio), filtered, and the filter cake was dried. Pale pink solid (430 mg, yield: 92%).
  • step 1
  • the diisopropylamine (2.4 g, 24.0 mmol) was dissolved in anhydrous tetrahydrofuran (20 ml), and the mixture was stirred under nitrogen, and a n-hexane solution of n-butyllithium (9.6 ml, 24.0 mm) was added dropwise at 0 ° C. After reacting for 30 minutes, anhydrous tetrahydrofuran (30 ml) was added, and the mixture was cooled to -78 ° C, and a solution of 3,5-dibromopyridine 26-a (4.7 g, 20 mm) in anhydrous tetrahydrofuran (50 ml) was added dropwise.
  • step 1
  • step 1
  • Chloroacetyl chloride (470 ul, 6.20 mmol) was added dropwise to an anhydrous aluminum chloride (2.48 g, 18.60 mmol) in a carbon disulfide suspension under ice bath, stirred for 10 minutes, and 4,5-dihydro-1H-benzene was added.
  • Azazao-2(3H)-one 29-a 500 mg, 3.10 mmol
  • Triethylsilane (840 uL, 5.25 mmol) was added dropwise to a solution of compound 29-b (500 mg, 2.10 mmol) in trifluoroacetic acid (5 ml).
  • the reaction solution was poured into ice water, extracted with dichloromethane three times, and the organic phase was combined, dried, concentrated, and the residue was pulverized in isopropyl ether, filtered, and dried to give a pale yellow solid (430 mg, yield: 91.4% ).
  • step 1
  • step 1
  • Diisopropylamine (5.14g, 50.8mmol) was dissolved in anhydrous tetrahydrofuran (50ml), protected with nitrogen, stirred, and n-hexane solution of n-butyllithium (20.3ml, 50.8mml) was added dropwise at 0 °C.
  • the reaction solution was cooled to -78 ° C for 30 minutes, and a solution of 3-bromofluorobenzene 31-a (10.0 g, 42.4 mm) in anhydrous tetrahydrofuran (100 ml) was added dropwise, and the reaction was continued for 30 minutes after the dropwise addition, and methyl formate was added dropwise.
  • the compound 31-f (3.24 g, 8.3 mmol) was dissolved in tetrahydrofuran (60 ml), and lithium hydroxide monohydrate (698 mg, 16.6 mmol) and methanol (10 ml) were added thereto with stirring, and reacted at room temperature for 5 hours, and added to the reaction mixture.
  • the pH was adjusted to 4 to 5 with 1N hydrochloric acid, and the solid was separated, filtered, and the filter cake was dried in vacuo to give a white solid 31-g (2.8 g, yield: 96.5%).
  • the product of Preparation 16 was obtained as 2-(6-chloro-2-oxo-1,2,3,4-tetrahydroquinolin-7-yl)ethyl methanesulfonate (330 mg, 1.09 mmol). Hexacyclic (5 ml) and 2,3-dichloro-5,6-dicyanyl p-benzoquinone (DDQ, 740 mg, 3.26 mmol) were heated to reflux overnight.
  • DDQ 2,3-dichloro-5,6-dicyanyl p-benzoquinone
  • step 1
  • Chloroacetyl chloride (460 uL, 6.16 mmol) was added dropwise to a suspension of anhydrous aluminum chloride (1.03 g, 7.7 mmol) in 1,2-dichloroethane under ice bath, stirred for 10 minutes, and 3-methyl was added.
  • Triethylsilane (587 ul, 3.68 mmol) was added dropwise to a solution of compound 33-b (350 mg, 1.47 mmol) in trifluoroacetic acid (4 ml), and reacted at 35 ° C for 2.5 hours, and the reaction mixture was poured into ice water to precipitate. Solid, filtered, filter cake washed with ice water 3 The mixture was dried to give a pale yellow solid (320 mg, yield: 97%).
  • step 1
  • Chloroacetyl chloride (640 ul, 8.49 mmol) was added dropwise to a suspension of anhydrous aluminum chloride (2.72 g, 20.38 mmol) in carbon disulfide (10 ml) under ice bath, stirred for 10 minutes, and 3,4-dihydroquinoline was added.
  • Triethylsilane (900 uL, 5.58 mmol) was added dropwise to a solution of compound 35-b (500 mg, 2.32 mmol) in trifluoroacetic acid (5 ml) and allowed to react at 30 ° C overnight.
  • the reaction solution was poured into ice water, extracted with dichloromethane (15 ml ⁇ 3), the organic phase was combined, dried, filtered, dried, concentrated, concentrated, and the residue was added with petroleum ether, filtered, and the filter cake was dried to give a tan solid. (417 mg, yield: 89%).
  • step 1
  • the object was prepared by the procedure of Preparation 37, using ethyl propiolate instead of ethyl 2-ethyl-3-oxopentanoate.
  • Example 5 The product of Example 5 (120 mg, 0.28 mmol) was dissolved in dry tetrahydrofuran (3 ml), and methyl brunium bromide (2 ml, 1.99 mmol, 1 M in THF) was added dropwise to the above solution at room temperature under ice bath. After stirring for 3 hours, it is quenched by the addition of saturated ammonium chloride, and the residue is evaporated to dryness. It was beaten to a pale yellow solid (40 mg, yield: 32%).
  • Example 17 The product of Example 17 (180 mg, 0.35 mmol) was. The reaction solution was cooled, and then added dropwise to isopropyl alcohol (5 ml), and solid was evaporated to give a pale yellow solid (130 mg, yield: 87%).
  • Example 5 The product of Example 5 (120 mg, 0.28 mmol) was dissolved in dichloromethane (3 ml), and dichloromethane (3. It was added dropwise to the above solution and stirred at room temperature overnight. Add saturated sodium bicarbonate solution, extract with methylene chloride, wash the organic phase with saturated brine, dry over anhydrous sodium sulfate, and then remove the desiccant, concentrate, and the residue is obtained by column chromatography to give a crude salt in a hydrogen chloride-ethanol solution A pale yellow solid (50 mg, yield: 39%).
  • Example 8 The product of Example 8 (500 mg, 1.325 mmol) was suspended in toluene (15 ml), and the solvent was added to the solvent (642 mg, 1.59 mmol), and the mixture was heated to 80 ° C overnight, and the reaction mixture was concentrated. The salt was formed in a hydrogen chloride-methanol solution, filtered, and the cake was then pulverized with a mixed solvent of isopropyl ether/methanol to give a yellow solid (40 mg, yield: 7.6%).
  • the target product was prepared by following the procedure of Example 55 using the product of Preparation 38 and the product of Preparation 1.
  • 1 H-NMR 300 Hz, DMSO-d 6 ): ⁇ ppm 9.45 (brs, 1H), 7.96 (d, 1H), 7.77 (d, 1H), 7.70 (d, 1H), 7.50 (d, 1H), 7.31 (t, 1H), 6.96 (d, 1H), 6.36 (d, 1H), 3.63 (t, 2H), 3.17-3.57 (m, 10H).
  • ESI-MS (m/z): 341.0 [M+H ] + .
  • Use LANCE TM cAMP 384 Kit (U.S. PerkinElmer Inc.) agonism compound tested expressing recombinant human 5-HT 1A receptors HEK293 cells 5-HT 1A receptor.
  • the 5-HT 1A agonism of the compounds was assessed by testing the inhibitory effect of the compounds on cAMP production in HEK293 cells.
  • the cAMP concentration test was carried out according to the method in the kit instructions. The test concentration of the compound was 0.1 nM-10000 nM, 8-OH-DPAT was used as a positive control, and the EC 50 was calculated by Excelfit software. The results are shown in Table 1.
  • Use LANCE TM cAMP 384 Kit (U.S. PerkinElmer Inc.) antagonistic compounds tested recombinant D 2 receptors HEK293 cells expressing human D 2 receptor. Antagonism of dopamine inhibition by the test compound in HEK293 cells cAMP production to assess the compound D 2 antagonism.
  • the cAMP concentration test was carried out according to the method in the kit instructions. The test concentration of the compound was 0.1 nM-10000 nM, risperidone was used as a positive control, and the IC 50 was calculated by Excelfit software. The results are shown in Table 1.
  • Calcium 5 Assay Kit (Molecular Devices USA Inc.) antagonistic compounds tested recombinant 5-HT 2A receptor in CHO-K1 cells expressing human 5-HT 2A receptor.
  • the test was carried out according to the method in the kit instructions, and the test concentration of the compound was 0.1 nM to 10000 nM, and risperidone was used as a positive control.
  • the test method was as follows: On the first day, the seed cells were placed at a density of 14 million per bottle in a 25 ml growth medium (F-12 nutrient mixture + 10% FBS + 1% penicillin / streptomycin + 1.2% 50 mg / ml Geneticin).
  • the cells were cultured for 24 hours at 37 ° C, 5% CO 2 , humidified; the next day, seed cells were seeded into 384-well cell culture plates, and each well was inoculated with 20,000 cells.
  • 50 ⁇ L of the test medium F-12 nutrient mixture + 1.5% activated carbon-treated FBS
  • the test medium was used to replace the growth medium, and the cells were cultured for 16 hours at 37 ° C, 5% CO 2 , humidified; the medium was removed on the third day.
  • Use LANCE TM cAMP 384 Kit U.S. PerkinElmer Inc.
  • D 2 agonism was assessed by compound inhibition of the test compound on the cAMP production in HEK293 cells.
  • the cAMP concentration test was carried out according to the method in the kit instructions. The test concentration of the compound was 0.1 nM to 10000 nM, and dopamine was used as a positive control. The IC 50 was calculated by Excelfit software, and the results are shown in Table 2.
  • PCP was dissolved in physiological saline to prepare a solution at a dose of 7 mg/kg.
  • Aripiprazole and the test compound are formulated into a solution of a suitable concentration with a 0.5% CMC-Na solution, which is ready for use.
  • the spontaneous sputum video analysis system was used to record the trajectory of the mice after 45 minutes of administration of the test drug or physiological saline, and then the trajectory of the mice within 75 minutes after administration of PCP was recorded.
  • the activity trajectories of the mice were analyzed by a spontaneous and open-field video analysis system, and the total distance of activities of each group of mice was counted, and the results were expressed by mean ⁇ SD. Results were statistically analyzed using one-way analysis of variance.
  • mice After PCP modeling, the spontaneous activity of the mice was significantly increased compared with the saline group.
  • the test compound at the following doses (Table 3) significantly reduced the high spontaneous activity of PCP-induced mice, which was significantly different from the model group. .

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Pain & Pain Management (AREA)
  • Hospice & Palliative Care (AREA)
  • Anesthesiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

本发明提供了一种由通式(I)表示的杂环化合物、其立体异构体或其药学上可接受的盐、其药物组合物、以及其在制备预防和/或治疗中枢神经系统疾病的药物中的应用。

Description

一类杂环化合物、其制备方法和用途 技术领域
本发明属于药物化学领域。具体而言,本发明涉及一类通式(I)所示的杂环化合物、其立体异构体或其药学上可接受的盐,其制备方法,药物组合物及其在制备中枢神经系统疾病药物中的应用。
背景技术
精神病主要是一组以表现在行为、心理活动上的紊乱为主的神经系统疾病,临床表现为精神活动异常,具体表现为感知觉、思维、注意、记忆、情感、行为和意志智能以及意识等方面不同程度的障碍。
由于中枢神经系统的复杂性,现有抗精神病药物仍有各自局限。例如,临床上治疗重度抑郁症(MDD)、焦虑的药物起效慢,药效差;现有抗精神分裂症药物仍不能很好地改善阴性症状及认知功能损伤,或者有锥体外系反应(EPS)、代谢紊乱等副作用。因此,仍有必要寻找具有疗效好、副作用低、治疗谱宽的新型抗精神病药。
多巴胺系统调控运动、情感、奖赏、认知等生理过程,5-羟色胺(5-HT)也参与多种生理功能的调节,如体温调节、情绪活动、痛觉、睡眠-觉醒等。因此,现有中枢神经系统领域药物多数与多巴胺、5-HT等中枢神经系统递质的调节有关。
D2拮抗剂是经典的抗精神病药物,也应用于失眠的治疗;5-HT2A受体拮抗作用减少了EPS,具有改善阴性症状、改善认知缺损、改善抑郁症状、改善焦虑和改善失眠的作用(European Journal of Pharmacology,2000,407:39-46)。然而,单纯的D2拮抗剂或D2拮抗/5-HT2A拮抗剂仍有不同程度的副作用。
5-HT1A受体激动剂在治疗重度抑郁症、焦虑、抑郁,改善精神分裂症患者的阴性症状及认知功能等方面表现出良好的临床应用前景(CNS Drugs,2013 Sep,27:703-16)。例如,5-HT1A受体激动剂BAY-3702在动物模型中具有神经保护、抗焦虑和抗抑郁作用(European Journal of Pharmacology,1998,357:1-8);吉哌隆可用于缓解某种原发性抑郁性疾病(US4771053),如重度抑郁症、内源性抑郁症和非典型抑郁症;丁螺旋酮可治疗与注意力缺乏与多动障碍(ADHD)相关的各种症状,且D2受体激动剂和5-HT1A受体激动剂的联合应用可有效地治疗ADHD和帕金森氏病(WO200016777A);伊沙匹隆通过改善记忆力可有效地治疗阿尔茨海默病或帕金森氏病的认知减退(US5824680)。
多巴胺受体部分激动剂可以改善精神分裂症阳性症状、阴性症状、焦虑抑郁、认知缺陷,而很少引起像D2拮抗剂所致的血清催乳素增高,D2拮抗/5-HT2A拮抗剂所致的体重增加、代谢异常等不良反应,其安全、耐受性良好。
因此,具有DA/5-HT多靶点作用的药物有利于更好地调节脑内多种受体的平衡,调节DA/5-HT系统,从而治疗中枢神经系统领域的疾病。本发明提供了一类具有5-HT2A和/或D2受体和/或5-HT1A多靶点作用活性的化合物,可用于中枢神经系统疾病的治疗,尤其是忧郁症、躁郁症、精神分裂症、焦虑症、恐怖症、自闭症、阿尔茨海默(Alzheimer)病、双极情感障碍、癔症、强迫症、多动症等疾病。
发明内容
发明目的
本发明目的是提供一类对5-羟色胺2A(5-HT2A)受体具有拮抗作用和/或对多巴胺D2受体和/或5-羟色胺1A(5-HT1A)受体均有良好活性的化合物。
本发明的一个目的是提供一种通式(I)所示杂环化合物、其立体异构体或其药学上可接受的盐。
本发明的另一个目的是提供制备本发明化合物的方法。
本发明的又一个目的是提供通式(I)所示杂环化合物、其立体异构体或其药学上可接受的盐在制备预防和/或治疗中枢神经系统疾病的药物中的应用。
本发明的再一个目的是提供包含治疗有效量的本发明所述的通式(I)所示化合物、其立体异构体或其药学上可接受的盐的药物组合物。
技术方案
根据本发明的一个方面,提供了一种由通式(I)表示的杂环化合物、其立体异构体或其药学上可接受的盐:
Figure PCTCN2015073854-appb-000001
其中:
A、B和D各自独立地为C或N,且当A为N时,D同时也为N;
Figure PCTCN2015073854-appb-000002
表示单键或双键;
E为CH,N或C;当E为CH或N时,与E相连的
Figure PCTCN2015073854-appb-000003
表示单键;当E为C时,与E相连的
Figure PCTCN2015073854-appb-000004
表示双键;
R1为氢或1至4个各自独立地选自卤素、羟基、巯基、氧代(=O)、硫代(=S)、C1~C6 烷氧基、卤代C1~C6烷氧基、C1~C6烷硫基、C1~C6烷基、卤代C1~C6烷基、硝基、氨基、被C1~C6烷基取代的氨基、氰基、羧基、醛基、氨基C1~C6烷基、羟基C1~C6烷基、氰基C1~C6烷基、C1~C6烷酰基、卤代C1~C6烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C6烷基取代的氨基甲酰基、羧基C1~C6烷基、C1~C6烷磺酰基、卤代C1~C6烷磺酰基、被C1~C6烷基取代的氨基C1~C6烷基、氨基甲酰基C1~C6烷基或被C1~C6烷基取代的氨基甲酰基C1~C6烷基;R1优选为氢或1至4个各自独立地选自卤素、羟基、巯基、氧代(=O)、硫代(=S)、C1~C4烷氧基、卤代C1~C4烷氧基、C1~C4烷硫基、C1~C4烷基、卤代C1~C4烷基、硝基、氨基、被C1~C4烷基取代的氨基、氰基、羧基、醛基、氨基C1~C4烷基、羟基C1~C4烷基、氰基C1~C4烷基、C1~C4烷酰基、卤代C1~C4烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C4烷基取代的氨基甲酰基、羧基C1~C4烷基、C1~C4烷磺酰基、卤代C1~C4烷磺酰基、被C1~C4烷基取代的氨基C1~C4烷基、氨基甲酰基C1~C4烷基或被C1~C4烷基取代的氨基甲酰基C1~C4烷基;R1更优选为氢或1至4个各自独立地选自氟、氯、溴、羟基、巯基、氧代(=O)、硫代(=S)、甲氧基、乙氧基、三氟甲氧基、-SCH3、-SCH2CH3、甲基、乙基、丙基、异丙基、叔丁基、三氟甲基、溴甲基、氯甲基、硝基、氨基、N-甲基氨基、N-乙基氨基、N,N-二甲基氨基、N,N-二乙基氨基、氰基、羧基、醛基、-CH2NH2、-CH2CH2NH2、-CH2OH、-CH2CH2OH、-CH2CN、-CH2CH2CN、甲酰基、乙酰基、丙酰基、三氟乙酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基、N-甲基氨基甲酰基、N,N-二甲基氨基甲酰基、N-乙基氨基甲酰基、N,N-二乙基氨基甲酰基、-CH2CO2H、-CH2CH2CO2H、-SO2CH3、-SO2CF3、-CH2NHMe、-CH2NMe2、-CH2CONH2、-CH2CONHMe或-CH2CONMe2
R2不存在,或者为1至3个各自独立地选自卤素、羟基、巯基、C1~C6烷氧基、卤代C1~C6烷氧基、C1~C6烷硫基、C1~C6烷基、卤代C1~C6烷基、硝基、氨基、被C1~C6烷基取代的氨基、氰基、羧基、醛基、氨基C1~C6烷基、羟基C1~C6烷基、氰基C1~C6烷基、C1~C6烷酰基、卤代C1~C6烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C6烷基取代的氨基甲酰基、羧基C1~C6烷基、C1~C6烷磺酰基、卤代C1~C6烷磺酰基、被C1~C6烷基取代的氨基C1~C6烷基、氨基甲酰基C1~C6烷基或被C1~C6烷基取代的氨基甲酰基C1~C6烷基;R2优选为不存在,或者为1至3个各自独立地选自卤素、羟基、巯基、C1~C4烷氧基、卤代C1~C4烷氧基、C1~C4烷硫基、C1~C4烷基、卤代C1~C4烷基、硝基、氨基、被C1~C4烷基取代的氨基、氰基、羧基、醛基、羟基C1~C4烷基、氰基C1~C4烷基、C1~C4烷酰基、卤代C1~C4烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C4烷基取代的氨基甲酰基、羧基C1~C4烷基、C1~C4烷磺酰基、卤代C1~C4烷磺酰基、氨基C1~C4烷基、 被C1~C4烷基取代的氨基C1~C4烷基、氨基甲酰基C1~C4烷基或被C1~C4烷基取代的氨基甲酰基C1~C4烷基;R2更优选为不存在,或者为1至3个各自独立地选自氟、氯、溴、羟基、巯基、甲氧基、乙氧基、三氟甲氧基、-SCH3、-SCH2CH3、甲基、乙基、丙基、异丙基、叔丁基、三氟甲基、溴甲基、氯甲基、硝基、氨基、N-甲基氨基、N-乙基氨基、N,N-二甲基氨基、N,N-二乙基氨基、氰基、羧基、醛基、-CH2OH、-CH2CH2OH、-CH2CN、-CH2CH2CN、甲酰基、乙酰基、丙酰基、三氟乙酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基、N-甲基氨基甲酰基、N,N-二甲基氨基甲酰基、N-乙基氨基甲酰基、N,N-二乙基氨基甲酰基、-CH2CO2H、-CH2CH2CO2H、-SO2CH3、-SO2CF3、-CH2NH2、-CH2CH2NH2、-CH2NHMe、-CH2NMe2、-CH2CONH2、-CH2CONHMe或-CH2CONMe2
R3为氢或1至4个各自独立地选自羟基或C1~C6烷基的取代基;优选为氢或1至4个各自独立地选自羟基或C1~C4烷基的取代基;更优选为氢或1至4个各自独立地选自羟基、甲基或乙基的取代基;
L不存在,或者为C1~C5亚烷基(例如,C1亚烷基、C2亚烷基、C3亚烷基、C4亚烷基、C5亚烷基),且当L为C1~C5亚烷基时,该亚烷基非必须地被一个或多个选自羟基、C1~C6烷氧基和氧代基团(O=)中的取代基所取代;优选地,L不存在,或为C1~C4亚烷基,且该亚烷基非必须地被一个或多个选自羟基、C1~C6烷氧基和氧代基团(O=)中的取代基所取代;
G环为单环杂环基或双环杂环基,所述双环杂环基为苯并单环杂环基、环烃基并单环杂环基或单环杂环基并单环杂环基,其中所述单环杂环基含有至少一个选自N、S和O的杂原子;优选为3~10元单环杂环基、苯并[3~10元单环杂环基]、[C3-C10环烃基]并[3~10元单环杂环基]和[3~10元单环杂环基]并[3~10元单环杂环基];再优选为5~7元单环杂环基、苯并[5~7元单环杂环基]、[C5-C7环烃基]并[5~7元单环杂环基]和[5~7元单环杂环基]并[5~7元单环杂环基];更优选为呋喃基、二氢呋喃基、四氢呋喃基、噻吩基、二氢噻吩基、四氢噻吩基、吡咯基、二氢吡咯基、吡咯烷基、吡唑基、二氢吡唑基、吡唑烷基、三唑基、二氢三唑基、三唑烷基、噻唑基、二氢噻唑基、噻唑烷基、异噻唑基、二氢异噻唑基、异噻唑烷基、噁唑基、二氢噁唑基、噁唑烷基、异噁唑基、二氢异噁唑基、异噁唑烷基、吡喃基、二氢吡喃基、四氢吡喃基、吡嗪基、二氢吡嗪基、四氢吡嗪基、哌嗪基、哒嗪基、二氢哒嗪基、四氢哒嗪基、
Figure PCTCN2015073854-appb-000005
Figure PCTCN2015073854-appb-000006
Figure PCTCN2015073854-appb-000007
Figure PCTCN2015073854-appb-000008
Figure PCTCN2015073854-appb-000009
Figure PCTCN2015073854-appb-000010
G环与L的连接方式为通过G环上的碳原子与L相连;
并且,G环非必须地被一个或多个相同或不同取代基取代;
所述G环上的取代基为卤素、C1~C6烷基、卤代C1~C6烷基、C1~C6烷氧基、卤代C1~C6烷氧基、硝基、氰基、羟基、巯基、氨基、被C1~C6烷基取代的氨基、叠氮基、C1~C6烷酰基、卤代C1~C6烷酰基、C2~C6链烯基、C2~C6炔基、羧基C1~C6烷基、氰基C1~C6烷基、C2~C6链烯氧基、C2~C6炔氧基、氨基甲酰基(-CONH2)、被C1~C6烷基取代的氨基甲酰基、羧基、羟基C1~C6烷基、氧代(=O)、硫代(=S)、磺酰氨基(-SO2NH2)、C1~C6烷硫基、C1~C6烷磺酰基、卤代C1~C6烷磺酰基、磺酸基(-SO2OH)、醛基、氨基C1~C6烷基、被C1~C6烷基取代的氨基C1~C6烷基、氨基甲酰基C1~C6烷基、被C1~C6烷基取代的氨基甲酰基C1~C6烷基、C3-C10环烃基、C3-C10环烃基C1~C6烷基、C3-C10环烃基甲酰胺基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C10环烃基C1~C6烷氧基、呋喃基C1~C6烷基、呋喃基C1~C6烷氧基、噻吩基C1~C6烷基、噻吩基C1~C6烷氧基、吡咯基C1~C6烷基、吡咯基C1~C6烷氧基、吡咯烷基C1~C6烷基、吡咯烷基C1~C6烷氧基、吡唑基C1~C6烷基、吡唑基C1~C6烷氧基、三唑基C1~C6烷基、三唑基C1~C6烷氧基、噻唑基C1~C6烷基、噻唑基C1~C6烷氧基、异噻唑基C1~C6烷基、异噻唑基C1~C6烷氧基、噁唑基C1~C6烷基、噁唑基C1~C6烷氧基、异噁唑基C1~C6烷基、异噁唑基C1~C6烷氧基、吡嗪基C1~C6烷基、吡嗪基C1~C6烷氧基、哒嗪基C1~C6烷基、哒嗪基C1~C6烷氧基、吡啶基C1~C6烷基、吡啶基C1~C6烷氧基、嘧啶基C1~C6烷基、嘧啶基C1~C6烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C6烷基、苯基C1~C6烷氧基、苯基C1~C6烷酰基或苯基C1~C6烷酰氧基;上述C3-C10环烃基、C3-C10环烃基C1~C6烷基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C10环烃基C1~C6烷氧基、呋喃基C1~C6烷基、呋喃基C1~C6烷氧基、噻吩基C1~C6烷基、噻吩基C1~C6烷氧基、吡咯基C1~C6烷基、吡咯基C1~C6烷氧基、吡咯烷基C1~C6烷基、吡咯烷基C1~C6烷氧基、吡唑基C1~C6 烷基、吡唑基C1~C6烷氧基、三唑基C1~C6烷基、三唑基C1~C6烷氧基、噻唑基C1~C6烷基、噻唑基C1~C6烷氧基、异噻唑基C1~C6烷基、异噻唑基C1~C6烷氧基、噁唑基C1~C6烷基、噁唑基C1~C6烷氧基、异噁唑基C1~C6烷基、异噁唑基C1~C6烷氧基、吡嗪基C1~C6烷基、吡嗪基C1~C6烷氧基、哒嗪基C1~C6烷基、哒嗪基C1~C6烷氧基、吡啶基C1~C6烷基、吡啶基C1~C6烷氧基、嘧啶基C1~C6烷基、嘧啶基C1~C6烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C6烷基、苯基C1~C6烷氧基、苯基C1~C6烷酰基和苯基C1~C6烷酰氧基非必须地被一个或多个选自如下基团的取代基所取代:卤素、C1~C6烷基、卤代C1~C6烷基、C1~C6烷氧基、C1~C6烷氧酰基、卤代C1~C6烷氧基、硝基、氰基、羟基、氨基、C1~C6烷酰基、卤代C1~C6烷酰基、氨基甲酰基或羧基;优选地,所述G环上的取代基为卤素、C1~C4烷基、卤代C1~C4烷基、C1~C4烷氧基、卤代C1~C4烷氧基、硝基、氰基、羟基、巯基、氨基、被C1~C4烷基取代的氨基、叠氮基、C1~C4烷酰基、卤代C1~C4烷酰基、C2~C4链烯基、C2~C4炔基、羧基C1~C4烷基、氰基C1~C4烷基、C2~C4链烯氧基、C2~C4炔氧基、氨基甲酰基(-CONH2)、被C1~C4烷基取代的氨基甲酰基、羧基、羟基C1~C4烷基、氧代(=O)、硫代(=S)、磺酰氨基(-SO2NH2)、C1~C4烷硫基、C1~C4烷磺酰基、卤代C1~C4烷磺酰基、磺酸基(-SO2OH)、醛基、氨基C1~C4烷基、被C1~C4烷基取代的氨基C1~C4烷基、氨基甲酰基C1~C4烷基、被C1~C4烷基取代的氨基甲酰基C1~C4烷基、C3-C7环烃基、C3-C7环烃基C1~C4烷基、C3-C7环烃基甲酰胺基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C7环烃基C1~C4烷氧基、呋喃基C1~C4烷基、呋喃基C1~C4烷氧基、噻吩基C1~C4烷基、噻吩基C1~C4烷氧基、吡咯基C1~C4烷基、吡咯基C1~C4烷氧基、吡咯烷基C1~C4烷基、吡咯烷基C1~C4烷氧基、吡唑基C1~C4烷基、吡唑基C1~C4烷氧基、三唑基C1~C4烷基、三唑基C1~C4烷氧基、噻唑基C1~C4烷基、噻唑基C1~C4烷氧基、异噻唑基C1~C4烷基、异噻唑基C1~C4烷氧基、噁唑基C1~C4烷基、噁唑基C1~C4烷氧基、异噁唑基C1~C4烷基、异噁唑基C1~C4烷氧基、吡嗪基C1~C4烷基、吡嗪基C1~4烷氧基、哒嗪基C1~C4烷基、哒嗪基C1~C4烷氧基、吡啶基C1~C4烷基、吡啶基C1~C4烷氧基、嘧啶基C1~C4烷基、嘧啶基C1~C4烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C4烷基、苯基C1~C4烷氧基、苯基C1~C4烷酰基或苯基C1~C4烷酰氧基;上述C3-C7环烃基、C3-C7环烃基C1~C4烷基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C7环烃基C1~C4烷氧基、呋喃基C1~C4烷基、呋喃基C1~C4 烷氧基、噻吩基C1~C4烷基、噻吩基C1~C4烷氧基、吡咯基C1~C4烷基、吡咯基C1~C4烷氧基、吡咯烷基C1~C4烷基、吡咯烷基C1~C4烷氧基、吡唑基C1~C4烷基、吡唑基C1~C4烷氧基、三唑基C1~C4烷基、三唑基C1~C4烷氧基、噻唑基C1~C4烷基、噻唑基C1~C4烷氧基、异噻唑基C1~C4烷基、异噻唑基C1~C4烷氧基、噁唑基C1~C4烷基、噁唑基C1~C4烷氧基、异噁唑基C1~C4烷基、异噁唑基C1~C4烷氧基、吡嗪基C1~C4烷基、吡嗪基C1~4烷氧基、哒嗪基C1~C4烷基、哒嗪基C1~C4烷氧基、吡啶基C1~C4烷基、吡啶基C1~C4烷氧基、嘧啶基C1~C4烷基、嘧啶基C1~C4烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C4烷基、苯基C1~C4烷氧基、苯基C1~C4烷酰基和苯基C1~C4烷酰氧基非必须地被一个或多个选自如下基团的取代基所取代:卤素、C1~C4烷基、卤代C1~C4烷基、C1~C4烷氧基、C1~C4烷氧酰基、卤代C1~C4烷氧基、硝基、氰基、羟基、氨基、C1~C4烷酰基、卤代C1~C4烷酰基、氨基甲酰基或羧基;更优选地,所述G环上的取代基为氟、氯、溴、甲基、乙基、丙基、异丙基、叔丁基、三氟甲基、甲氧基、乙氧基、三氟甲氧基、硝基、氰基、羟基、巯基、氨基、N-甲基氨基、N-乙基氨基、N,N-二甲基氨基、N,N-二乙基氨基、叠氮基、甲酰基、乙酰基、丙酰基、三氟乙酰基、-CH2CO2H、-CH2CH2CO2H、-CH2CN、-CH2CH2CN、氨基甲酰基、N-甲基氨基甲酰基、N,N-二甲基氨基甲酰基、N-乙基氨基甲酰基、N,N-二乙基氨基甲酰基、羧基、-CH2OH、-CH2CH2OH、氧代(=O)、硫代(=S)、磺酰氨基(-SO2NH2)、-SCH3、-SCH2CH3、-SO2CH3、-SO2CF3、磺酸基(-SO2OH)、醛基、-CH2NH2、-CH2CH2NH2、-CH2NHMe、-CH2NMe2、-CH2NHEt、-CH2NEt2、-CH2CH2NHMe、-CH2CH2NHEt、-CH2CH2NMe2、-CH2CH2NEt2、-CH2CONH2、-CH2CONHMe、-CH2CONMe2、-CH2CONHEt、-CH2CONEt2、-CH2CH2CONH2、-CH2CH2CONHMe、-CH2CH2CONMe2、-CH2CH2CONHEt、-CH2CH2CONEt2、苯基、苯氧基、苯磺酰基、-CH2Ph、-CH2CH2Ph、-OCH2Ph、-OCH2CH2Ph、-COPh、-COCH2Ph或-CH2Ph(OMe)2
条件是不包括以下化合物:
1)1-[2-[4-(2-氰基-7-氟苯并噻吩-4-基)-2-甲基-1-哌嗪基]乙基]-3,4-二氢-1H-2-苯并吡喃-6-甲酰胺;
2)1-[2-[4-(7-氟苯并噻吩-4-基)-2-甲基-1-哌嗪基]乙基]-3,4-二氢-1H-2-苯并吡喃-6-甲酰胺;
3)6-乙基-4-[4-[2-(2-噻吩基)乙基]-1-哌嗪基]-噻吩并[2,3-d]嘧啶;
4)6-[2-[4-(5-甲基噻吩并[2,3-d]嘧啶-4-基)-1-哌嗪基]乙基]-2H-1,4-苯并噁嗪-3(4H)-酮;
5)6-乙基-4-[4-[3-[3-(2-噻吩基)-1,2,4-噁二唑-5-基]丙基]-1-哌嗪基]-噻吩并[2,3-d]嘧啶;
6)5,6-二甲基-4-[4-[3-[3-(4-甲基苯基)-1,2,4-噁二唑-5-基]丙基]-1-哌嗪基]-噻吩并[2,3-d]嘧啶。
在优选实施方式中,本发明所述通式(I)表示的杂环化合物中:
Figure PCTCN2015073854-appb-000011
优选地,
Figure PCTCN2015073854-appb-000012
在优选实施方式中,本发明所述通式(I)表示的杂环化合物选自下列化合物:
Figure PCTCN2015073854-appb-000013
Figure PCTCN2015073854-appb-000014
更优选地,本发明所述通式(I)表示的杂环化合物选自如下所示化合物:
Figure PCTCN2015073854-appb-000015
其中,R1、R2、R3、L和G环如前所定义和优选。
在通式(I)表示的杂环化合物、其立体异构体或其药学上可接受的盐中,最优选如下化合物或其药学上可接受的盐:
(1)6-氯-5-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(2)3-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(3)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(4)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(4a)(+)-3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(4b)(-)-3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(5)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-7,8-二氢-6H-吡啶并[1,2-a]嘧啶-4,9-二酮;
(6)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2,9-二甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(7)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(8)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(9)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(10)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)-3,4-二氢喹啉-2(1H)-酮;
(11)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
(12)7-(5-(4-(2-氯苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
(13)7-(5-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
(14)7-(5-(4-(苯并噻吩-4-基)-5,6-二氢吡啶-1(2H)-基)戊基)喹啉-2(1H)-酮;
(15)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯二氢吲哚-2-酮;
(16)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7-二氢吡啶并[1,2-a]嘧啶-4-酮;
(17)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-(苄氧基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮;
(18)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮;
(19)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(20)9-羟基-2-甲基-3-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(21)2-甲基-3-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(22)7-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
(23)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯-3,4-二氢喹啉-2(1H)-酮;
(24)6-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)-2-甲基喹唑啉-4(3H)-酮;
(25)7-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(26)1-(苯并噻吩-4-基)-4-(2-(4-甲基噻唑-5-基)乙基)哌嗪;
(27)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二氢苯并咪唑-2-酮;
(28)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9,9-二氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(29)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基-2(3H)-苯并噻唑酮;
(30)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基-2H-1,4-苯并噁嗪-3(4H)-酮;
(31)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基-2H-1,4-苯并噻嗪-3(4H)-酮;
(32)7-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(33)7-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)喹啉-2(1H)-酮;
(34)6-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)喹唑啉-4(3H)-酮;
(35)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹唑啉-4(3H)-酮;
(36)3-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(37)5-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-6-氯-二氢吲哚-2-酮;
(38)4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(39)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(40)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯喹啉-2(1H)-酮;
(41)9-羟基-2-甲基-3-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并 [1,2-a]嘧啶-4-酮;
(42)6-氯-5-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(43)7-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(44)7-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)喹啉-2(1H)-酮;
(45)7-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基-3,4-二氢喹啉-2(1H)-酮;
(46)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-吲哚;
(47)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(48)5-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)二氢吲哚-2-酮;
(49)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-4,5-二氢-1H-苯并氮杂卓-2(3H)-酮;
(50)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮;
(51)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3-甲基-3,4-二氢喹唑啉-2(1H)-酮;
(52)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-氯-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
(53)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-硫酮;
(54)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-硫酮;
(55)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5,6-二乙基嘧啶-4(3H)-酮;
(56)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)嘧啶-4(3H)-酮;
(57)7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(58)2-((4-(苯并噻吩-4-基)哌嗪-1-基)甲基)-1H-苯并咪唑;
(59)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑;
(60)7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
(61)N-(7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(62)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
(63)N-(5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(64)7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(65)6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(66)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-7-氟-2H-苯并[1,4]噁嗪-3(4H)-酮;
(67)7-(2-(4-(3-甲基苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(68)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噁唑-2(3H)-酮;
(69)4-((4-(苯并噻吩-4-基)哌嗪-1-基)甲基)喹啉-2(1H)-酮;
(70)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-1H-吲哚-5-甲腈;
(71)7-(5-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
(72)6-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-8-氟-2H-苯并[1,4]噁嗪-3(4H)-酮;
(73)1-(苯并噻吩-4-基)-4-((2,3-二氢苯并[1,4]二氧杂环己烯-2-基)甲基)哌嗪;
(74)6-(4-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
(75)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-苯并咪唑;
(76)N-(6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(77)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-苯并咪唑-2(3H)-酮;
(78)6-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(79)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(80)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基喹唑啉-4(3H)-酮;
(81)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,1-二氧代-3,4-二氢-2H-苯并[1,2]噻嗪;
(82)5-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)二氢吲哚-2-酮;
(83)7-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(84)5-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(85)7-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(86)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-甲基-1H-苯并咪唑-2(3H)-酮;
(87)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二甲基-1H-苯并咪唑-2(3H)-酮;
(88)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-甲基-1H-苯并咪唑-2(3H)-酮;
(89)6-(2-(4-(2-甲氧基苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(90)3-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-4-酮;
(91)7-(2-(4-(2-氧代-2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(92)6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(93)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二甲基-1H-苯并咪唑-2(3H)-酮;
(94)6-氟-5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(95)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氟二氢吲哚-2-酮;
(96)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-苄基-3-甲基喹唑啉-2,4(1H,3H)-二酮;
(97)6-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(98)6-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(99)3-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-2-甲基-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-4-酮;
(100)6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(101)5-(4-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)丁基)二氢吲哚-2-酮;
(102)7-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
(103)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-6,7-二甲氧基-4H-色烯-4-酮;
(104)6-(4-(4-(苯并噻吩-4-基)哌啶-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
(105)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5-甲氧基-1H-吲哚;
(106)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-5-甲氧基-1H-吲哚;
(107)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-5-甲氧基-1H-吲哚;
(108)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-1H-吲哚-5-甲腈;
(109)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-吲哚-5-甲腈;
(110)1-乙酰基-3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-1H-吲哚-5-甲腈;
(111)6-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
(112)5-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)二氢吲哚-2-酮;
(113)6-氯-5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
(114)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-4-甲基-2H-苯并[1,4]噁嗪-3(4H)-酮;
(115)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-甲基-3,4-二氢喹啉-2(1H)-酮;
(116)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
(117)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
(118)N-(5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(119)N-(7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(120)4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
(121)N-(4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(122)7-(2-(4-(2-甲基苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(123)3-((4-(苯并噻吩-4-基)哌嗪-1-基)甲基)-1-甲基-1H-吲哚;
(124)1-(3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-1-基)乙酮;
(125)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-对甲苯磺酰基-1H-吲哚-5-甲腈;
(126)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-1-对甲苯磺酰基-1H-吲哚-5-甲腈;
(127)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-5-甲氧基-1-对甲苯磺酰基-1H-吲哚;
(128)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-5-甲氧基-1-对甲苯磺酰基-1H-吲哚;
(129)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5-甲氧基-1-对甲苯磺酰基-1H-吲哚;
(130)6-(2-(4-(2-氧代-2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
(131)3-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-4-酮;
(132)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(133)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-硫酮;
(134)(3aR,4R,6aS)-4-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)四氢-1H-噻吩并[3,4-d]咪唑-2(3H)-酮;
(135)(6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)氨基甲酸戊酯;
(136)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-4-氧代-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-9-基苯甲酸酯;
(137)6-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(138)1-(苯并噻吩-4-基)-4-(4-(1-环己基-1H-四唑-5-基)丁基)哌嗪;
(139)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-8-氟-2H-苯并[1,4]噁嗪-3(4H)-酮;
(140)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟乙基)喹啉-2(1H)-酮;
(141)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(142)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基乙基)-2H-苯并[1,4]噻嗪-3(4H)-酮;
(143)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)二氢吲哚-2-酮;
(144)8-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-甲氧基乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
(145)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基乙基)二氢吲哚-2-酮;
(146)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)苯并噻唑-2(3H)-酮;
(147)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)-1H-苯并咪唑-2(3H)-酮;
(148)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)-3,4-二氢喹啉-2(1H)-酮;
(149)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊酰基)喹啉-2(1H)-酮;
(150)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基戊基)-3,4-二氢喹啉-2(1H)-酮;
(151)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊酰基)-3,4-二氢喹啉-2(1H)-酮;
(152)N-(6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)环戊甲酰胺;
(153)N-(6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
(154)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺。
本发明提供一类通式(I)所示的杂环化合物的制备方法,所述制备方法可通过如下方法1-5之一进行。
方法1:式(II)所示化合物或其盐与式(III)所示化合物或其盐进行N-烷基化反应,如反应式1所示:
Figure PCTCN2015073854-appb-000016
其中,G环、L、A、B、D、E、R1、R2和R3同上文所定义和优选;
X代表离去基团,如卤素、C1~C6烷基磺酰氧基、苯磺酰氧基、萘磺酰氧基,上述C1~C6烷基磺酰氧基、苯磺酰氧基、萘磺酰氧基可非必须地被一个或多个选自卤素、C1~C6烷基、C1~C6烷氧基、硝基、羟基、氨基和C1~C6烷酰基的基团进一步取代;X优选为卤素、C1~C4烷基磺酰氧基、苯磺酰氧基、萘磺酰氧基,上述C1~C4烷基磺酰氧基、苯磺酰氧基、萘磺酰氧基可非必须地被一个或多个选自卤素、C1~C4烷基、C1~C4烷氧基、硝基、羟基、氨基和C1~C4烷酰基进一步取代;X最优选为氯、溴、甲磺酰氧基、三氟甲磺酰氧基、苯磺酰氧基、萘磺酰氧基、甲基苯磺酰氧基、硝基苯磺酰氧基、氨基苯磺酰氧基、氯苯磺酰氧基、溴苯磺酰氧基和甲氧基苯磺酰氧基。
式(II)所示的化合物或其盐与式(III)所示的化合物或其盐之间的N-烷基化反应是在没有溶剂的条件下进行的,或者是在有碱或没有碱的溶剂中进行的。所述溶剂包括水;醚类,如:二噁烷、四氢呋喃、乙醚、甲基叔丁基醚、二异丙醚、二甘醇二甲醚、乙二醇二甲醚等;芳香烃类,如:苯、甲苯、二甲苯、硝基苯、氯苯等;醇类,如:甲醇、乙醇、异丙醇、丁醇、叔丁醇、乙二醇;酮类,如丙酮、甲乙酮、4-甲基-2-戊酮等;酰胺类,如:N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、1-甲基-2-吡咯烷酮等;卤代烃类,如:氯仿、二氯甲烷、二氯乙烷、四氯化碳;酯类,如:乙酸乙酯、甲酸乙酯、乙酸甲酯、乙酸异丙酯;其它类,如:二甲基亚砜、乙腈等;或上述溶剂的混合物。
所述碱可选自无机碱或有机碱,无机碱包括碱金属氢氧化物,如:氢氧化钠、氢氧化钾、氢氧化铯、氢氧化锂;碱金属碳酸盐,如:碳酸钠、碳酸钾、碳酸铯、碳酸锂;碱金属碳酸氢化物,如:碳酸氢钠、碳酸氢钾、碳酸氢锂;碱金属,如:钾、钠;其它,如:氨基钠、氨基钾、氢化钠、氢化钾;有机碱包括甲醇钠、乙醇钠、甲醇钾、乙醇钾、醋酸钠、三乙胺、吡啶、二异丙基胺、二异丙基乙胺、三丙胺、二乙胺、嘧啶、喹啉、哌啶、哌嗪、咪唑、二甲氨基吡啶、三甲胺、N-乙基二异丙胺、N-甲基吗啉、二甲基苯胺、1,8-二氮杂二环[5.4.0]十一碳-7-烯(DBU)、1,5-二氮杂二环[4.3.0]壬-5-烯(DBN)、1,4-二氮杂二环[2.2.2]辛烷(DABCO)。这些碱可以一种单独使用,或者两种或多种联合使用。
如果有必要,可以加入碱金属碘化物如碘化钾和碘化钠作为反应促进剂进行上述反应。
上述反应通常在室温至200℃下进行,优选室温至150℃下进行,反应通常在约1~30小时,优选在5-20小时内完成。
方法2:式(IV)所示的化合物或其盐与式(V)所示的化合物或其盐进行偶联反应,如反应式2所示:
Figure PCTCN2015073854-appb-000017
其中,G环、L、A、B、D、R1、R2和R3同上文所定义和优选,E1代表氮原子;X1代表卤素或三氟甲磺酰氧基,优选溴、碘、氯或三氟甲磺酰氧基;
所述偶联反应在钯催化和碱存在下进行,得到的式(Ia)化合物为式(I)化合物的一个特例。
所述钯催化剂为醋酸钯(Pd(OAc)2)、二(三苯基膦)二氯化钯((Ph3P)2PdCl2)、双(苯甲腈)氯化钯((PhCN)2PdCl2)、四(三苯基膦)钯(Pd(PPh3)4)、双(三苯基膦)醋酸钯((Ph3P)2Pd(OAc)2)、1,2-二(二苯基膦基)乙烷二氯化钯((PdCl2(dppe)2))、双(1,2-双(二苯基膦)乙烷)钯(Pd(dppe)2)、双(二亚苄基丙酮)钯(Pd(dba)2)、三(二亚苄基丙酮)二钯(Pd2(dba)3)、[1,3-双(二苯基膦基)丙烷]二氯化钯(PdCl2(dippp))和[1,1′-双(二苯基膦)二茂铁]二氯化钯(Pd(dppf)Cl2);所述碱为双(三甲硅基)氨基钠、叔丁醇钾、叔丁醇钠、碳酸铯、磷酸钾、磷酸钠、甲醇钠、乙醇钠、氢氧化钾、氢氧化钠、氟化钾、氟化钠、氟化四丁基铵(TBAF)、醋酸钠、醋酸钾、碳酸铯、碳酸钾和碳酸钠中的一种或多种。所述反应溶剂没有特殊限制,只要其不干扰反应即可,包括水;醚类,如:二氧六环、四氢呋喃等;芳香烃类,如:甲苯、二甲苯等;醇类,如:叔丁醇等;酮类,如丙酮等;酰胺类,如:N,N-二甲基甲酰胺等;其它类,如:二甲基亚砜、乙腈等;或上述溶剂的混合物;如果有必要,可以加入合适的配体作为反应促进剂进行上述反应。合适的配体为2,2′-二苯膦基-1,1′-联萘(BINAP)、三叔丁基(P(t-Bu)3)、1,1′-二-(二苯膦基)二茂铁(dppf)、2-二环己基磷-2,4,6-三异丙基联苯(x-phos)、4,5-双二苯基膦-9,9-二甲基氧杂蒽(Xantphos)、三叔丁基膦四氟硼酸盐和三(2-甲基苯基)膦(P(o-tolyl)3)。
方法3:式(VI)所示化合物或其盐与式(III)所示化合物或其盐经酰胺化反应得到式(VII) 化合物或其盐;式(VII)化合物或其盐用还原剂处理,得到式(I)化合物,如反应式3所示:
Figure PCTCN2015073854-appb-000018
其中,G环、L、A、B、D、E、R1、R2和R3同上文所定义和优选。
所述的酰胺化反应可采用两种方法进行。
第一种酰胺化方法是在有或无催化剂存在下,式(VI)化合物的羧基先活化再与式(III)所示化合物进行氨解反应。所述活化剂可以为选自氯化亚砜、草酰氯、溴化亚砜、三氯氧磷、五氯化磷、特戊酰氯、氯甲酸乙酯、氯甲酸异丁酯、碳酰二咪唑(CDI)、甲磺酰氯、对甲苯磺酰氯、对硝基苯磺酰氯等中的一种或几种。所述的催化剂可以为选自N,N-二甲基甲酰胺(DMF)、二乙基苯胺、二甲基苯胺、N-甲基吗啉等中的一种或几种。所述活化羧基的反应溶剂没有特殊限制,只要其不干扰反应即可,例如可以为选自二氯甲烷、二氯乙烷、二甲亚砜、四氢呋喃、苯、甲苯、氯仿、四氯化碳、二甲苯、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺等中的一种或几种。所述的氨解反应在碱存在下,在适当的溶剂中进行。所述的碱可以为选自吡啶、哌啶、吡咯烷、吗啉、N-甲基吗啉、喹啉、4-二甲氨基吡啶(DMAP)、三乙胺、二乙胺、三正丁胺、三丙基胺、二异丙基胺、二异丙基乙胺、甲醇钠、乙醇钠、叔丁醇钾、丁基锂、1,8-二氮杂环[5,4,0]十一烯-7(DBU)、N-甲基吗啉、喹啉、4-二甲氨基吡啶(DMAP)、钠氢、氢氧化钠、氢氧化钾、氢氧化锂、碳酸氢钠、碳酸氢钾、碳酸钠和碳酸钾中的一种或几种。所述氨解反应的反应溶剂可选自芳烃类溶剂、醚类溶剂、卤代烃类溶剂、其他溶剂和其组合。其中,所述芳烃类溶剂例如可以为选自苯、甲苯、二甲苯等中的一种或几种;所述醚类溶剂例如可以为选自四氢呋喃(THF)、乙醚、甲基叔丁基醚、乙二醇二甲醚、二乙二醇二甲醚、乙二醇单甲醚、二氧六环等中的一种或几种;所述卤代烃类溶剂例如可以为选自二氯甲烷、氯仿、四氯化碳、二氯乙烷等中的一种或几种;所述其他溶剂例如可以为选自甲醇、乙醇、乙二醇、正己烷、环己烷、N,N-二甲基甲酰胺(DMF)、N,N-二甲基乙酰胺、二甲亚砜(DMSO)、N-甲基吡咯烷酮、丙酮、乙腈、乙酸乙酯等中的一种或几种,但本发明不局限于上述溶剂。所述氨解反应的反应温度没有特殊限制,但是优选在-30℃~150℃,更优选-10℃~120℃之间。所述氨解反应的反应时间没有特殊限制,但是优选在10分钟~24小时之间。
第二种酰胺化方法是使用缩合剂,在有或无催化剂条件下,在有或无碱存在下进行。所述的缩合剂可以为选自N,N′-二环己基碳二亚胺(DCC)、1-乙基-3-(3-二甲氨基丙基)碳二亚胺(EDCl)、N,N′-二异丙基碳二亚胺(DIC)、O-苯并三氮唑-N,N,N′,N′-四甲基脲四氟硼酸酯(TBTU)、O-(7-偶氮苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(HATU)、苯并三氮唑-N,N,N′,N′-四甲基脲六氟磷酸酯(HBTU)、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸(BOP)苯并三氮唑-1-基氧-三(四氢吡咯基)鏻鎓六氟磷酸盐(PyBOP)中的一种或几种。所述的催化剂可以为选自1-羟基-苯并三氮唑(HOBt)、4-二甲氨基吡啶(DMAP)等中的一种或几种。所述碱可以为选自三乙胺、二乙胺、三正丁胺、三丙基胺、二异丙基胺、二异丙基乙胺、三甲胺、吡啶、2,6-二甲基吡啶、4-二甲氨基吡啶、哌啶、吡咯烷、喹啉、吗啉、N-甲基吗啉、N-乙基吗啉、二异丙基胺、二异丙基乙胺、1,8-二氮杂环[5,4,0]十一烯-7(DBU)或1,5-二氮杂二环[4.3.0]-壬-5-烯(DBN)等中的一种或几种。所述第二种酰胺化方法的反应溶剂可以为选自:烃类,如苯、二甲苯、甲苯、二氯甲烷或氯仿;醚类,如四氢呋喃、乙醚、丙醚或1,4-二氧六环;酰胺类,如N,N-二甲基甲酰胺、N,N-二乙基甲酰胺或N,N-二甲基乙酰胺;腈类,如乙腈;其它,如二甲亚砜等;以及上述溶剂的混合物,其中,优选的反应溶剂为四氢呋喃、乙腈、二氯甲烷、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺或二甲亚砜。所述第二种酰胺化方法的反应温度没有特殊限制,只要不影响反应进行即可,但是优选在-20℃~80℃,更优选0℃~50℃之间。
所述还原剂为硼烷、氢气/钯炭、四氢铝锂、三乙酰氧基硼氢化钠、二异丁基氢化铝、三氟化硼、三溴化硼、硼氢化钠和硼氢化钾中的一种或几种。
方法4:式(VIII)所示化合物或其盐与式(III)所示化合物或其盐经还原胺化反应得到式(I)化合物,如反应式4所示:
Figure PCTCN2015073854-appb-000019
其中,G环、L、A、B、D、E、R1、R2和R3同上文所定义和优选。所述还原胺化反应在还原剂存在下进行,所述还原剂包括但不限于:硼氢化钠、硼氢化钾、三乙酰氧基硼氢化钠(NaBH(OAc)3)、四甲基三乙酰氧硼氢化铵和氰基硼氢化钠等。
反应溶剂选自所述溶剂包括水;醚类,如:二噁烷、四氢呋喃、乙醚、甲基叔丁基醚、 二异丙醚、二甘醇二甲醚、乙二醇二甲醚等;芳香烃类,如:苯、甲苯、二甲苯、硝基苯、氯苯等;醇类,如:甲醇、乙醇、异丙醇、丁醇、叔丁醇、乙二醇;酮类,如丙酮、甲乙酮、4-甲基-2-戊酮、等;酰胺类,如:N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、1-甲基-2-吡咯烷酮等;卤代烃类,如:氯仿、二氯甲烷、二氯乙烷、四氯化碳;酯类,如:乙酸乙酯、甲酸乙酯、乙酸甲酯、乙酸异丙酯;腈类,如乙腈;其它,如二甲亚砜等;或上述溶剂的混合物;反应温度没有特别限制,通常为10~100℃,优选20~50℃;反应时间没有特别限制,通常为1~30小时。
方法5:
由方法1~4得到的式(I)化合物进行官能团转化得到,如通过氧化反应、格氏反应、水解反应、氟代反应、氯代反应或硫代反应等。
所述氧化反应在氧化剂存在下进行,所述氧化剂包括但不限于:戴斯-马丁氧化剂、琼斯试剂、Swern试剂(DMSO和草酰氯)和2,3-二氯-5,6-二氰对苯醌(DDQ)等。
所述格氏反应在格氏试剂存在下进行,所述格氏试剂包括但不限于:甲基氯化镁、甲基溴化镁,甲基碘化镁和乙基溴化镁等。
所述水解反应可在酸或碱存在下进行,所述酸或碱包括但不限于:盐酸、硫酸、氢氧化钠和氢氧化钾等。
所述氟代反应在氟代试剂存在下进行,所述氟代试剂包括但不限于:二乙胺基三氟化硫(DAST)、四氟化硫和五氟化碘等。
所述氯代反应在氯代试剂存在下进行,所述氯代试剂包括但不限于:氯化亚砜、五氯化磷和N-氯代丁二酰亚胺(NCS)等。
所述硫代反应在硫代试剂存在下进行,所述硫代试剂包括但不限于:五硫化二磷和劳森试剂等。
式(II)、式(III)、式(IV)、式(V)、式(VI)、式(VIII)化合物是商业上可得到的化合物或者是按照技术上已知的方法制备或按照相似化合物的合成方法制备。
上述各反应式中使用的起始化合物可以是适合的盐,所述合适的盐包括碱金属盐和碱土金属盐,如钠盐、钾盐、钙盐、镁盐等;有机碱盐如吡啶盐、三乙胺盐等;无机酸盐如盐酸盐、氢溴酸盐、氢碘酸盐、硫酸盐、硝酸盐、磷酸盐等;有机酸盐如甲酸盐、乙酸盐、丙酸盐、羟乙酸盐、乙二酸盐、丙二酸盐、丁二酸盐、富马酸盐、马来酸盐、乳酸盐、苹果酸盐、柠檬酸盐、酒石酸盐、苦味酸盐、谷氨酸盐、甲磺酸盐和苯磺酸盐等;
此外,上述各反应式中使用的起始化合物可以包括溶剂合物形式,如水合物、醇合物等。
本发明通式(I)所示的杂环化合物及其立体异构体也同样包括其溶剂合物形式,如水合物、醇合物等,并且所述的溶剂合物包括在本发明的范围内。
本发明通式(I)所示的杂环化合物及其立体异构体的药学上可接受的盐是指通式(I)所示的杂环化合物或其立体异构体用适当的酸处理,把它们转化成治疗活性的非毒性的加成盐形式。所述的盐例如盐酸盐、氢溴酸盐、氢碘酸盐、硫酸盐或硫酸氢盐、硝酸盐、磷酸盐或酸式磷酸盐、高氯酸盐、甲酸盐、乙酸盐、三氟乙酸盐、丙酸盐、丙酮酸盐、羟乙酸盐、乙二酸盐、丙二酸盐、丁二酸盐、戊二酸盐、马来酸盐、富马酸盐、乳酸盐、苹果酸盐、柠檬酸盐、酒石酸盐、苦味酸盐、谷氨酸盐、苯甲酸盐、甲磺酸盐、乙磺酸盐、苯磺酸盐、对甲苯磺酸盐、水杨酸盐、抗坏血酸盐、樟脑酸盐或樟脑磺酸盐等。相反,也可以用碱处理把盐形式转化成游离碱形式。
上述所用的术语“药学上可接受的盐”也包括它们的溶剂合物,并且所述的溶剂合物包括在本发明的范围内。溶剂合物的例子有,例如,水合物、醇合物等。
由各反应式获得的各目标化合物可以通过以下方法将其从反应混合物中分离和纯化,例如:反应混合物在冷却后经过如过滤、萃取或浓缩等方法分离出粗产物,接着通过常规的如柱层析法、打浆法或重结晶法来进行纯化。
本发明还提供根据本发明的通式(I)所示杂环化合物、其立体异构体或其药学上可接受的盐在制备预防和/或治疗中枢神经系统疾病的药物中的应用。
本发明还提供一种用于治疗和/或预防中枢神经系统疾病的方法,这种方法包括向人或动物施用上述本发明的通式(I)表示的杂环化合物、其立体异构体或其药学上可接受的盐。
本发明还提供一种药物组合物,其包含治疗有效量的上述通式(I)所示的杂环化合物、其立体异构体或其药学上可接受的盐和任选的可药用载体。所述药物组合物可用于治疗或者预防中枢神经系统疾病。
本发明还提供一种制备药物组合物的方法,包括将上述通式(I)所示的杂环化合物、其立体异构体或其药学上可接受的盐与可药用载体混合。
在本发明的药物组合物中,根据治疗目的可以选择多种药物制剂形式,一般包括:片剂、丸剂、胶囊剂、颗粒剂、混悬液、溶液、霜剂、软膏、粉剂、栓剂、气雾剂和注射剂等。
上述中枢神经系统疾病选自:精神分裂症;难控制的、难处理的或慢性精神分裂症;情感紊乱;精神紊乱;情绪紊乱;I型双极情感障碍;II型双极情感障碍;抑郁症;内因性抑郁症;重性抑郁症;难控制的抑郁症;情绪恶劣性障碍;循环情感性障碍;恐慌发作;惊恐性障碍;社交恐惧症;强迫性观念与行为病症;冲动性病症;创伤后精神紧张性障碍;焦虑症;急性应激障碍;癔病;神经性厌食症;睡眠障碍;适应性障碍;认知障碍;自闭症;神经性头痛;狂躁症;帕金森症;亨廷顿舞蹈症;阿尔茨海默症;各种痴呆症;记忆障碍;多动症;注意力缺乏/亢进类疾病和抽动症等。
在通式(I)中的各基团的定义如下:
术语卤素通常是指氟、氯、溴及碘;优选为氟、氯或溴;更优选为氟或氯;
C1~C6烷基指含有1-6个碳原子的直链或支链的饱和烃基,例如,甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1-乙基丙基、异戊基、新戊基、异己基、3-甲基戊基或正己基等,优选为甲基、乙基、正丙基、异丙基、丁基、异丁基或叔丁基;
卤代C1~C6烷基是指含有1-6个碳原子的直链或支链的饱和烃基的氢原子被1个或多个相同或不同的卤原子取代,例如三氟甲基、氟甲基、二氟甲基、氯甲基、溴甲基、二氯氟甲基、氯乙基、溴丙基、2-氯丁基或五氟乙基等;
C1~C6烷氧基指含有1-6个碳原子的直链或支链烷氧基,例如,甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基、仲丁氧基、正戊氧基、异戊氧基、新戊氧基、异己氧基、3-甲基戊氧基或正己氧基等,优选为甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基;
卤代C1~C6烷氧基是指含有1-6个碳原子的直链或支链烷氧基的氢原子被1个或多个相同或不同的卤原子取代,例如-OCF3、-OCH2CH2Cl、-OCHBrCH2Cl或-OCF2CF3等;
C1~C6烷硫基指含有1-6个碳原子的直链或支链烷硫基,例如,甲硫基、乙硫基、正丙硫基、异丙硫基、正丁硫基、异丁硫基、叔丁硫基、仲丁硫基、正戊硫基、异戊硫基、新戊硫基或正己硫基等,优选为甲硫基、乙硫基、正丙硫基、异丙硫基、正丁硫基、异丁硫基或叔丁硫基;
C2~C6链烯基指含有1~3个双键和2-6个碳原子的直链或支链的不饱和烃基,既包括顺式构型也包括反式构型,例如,乙烯基、1-丙烯基、2-丙烯基、1-甲基-1-丙烯基、2-甲基-1-丙烯基、2-甲基-2-丙烯基、1-丁烯基、2-丁烯基、3-丁烯基、1-戊烯基、2-戊烯基、3-戊烯基、4-戊烯基、1,3-丁二烯基、1,3-戊二烯基、1-己烯基、2-己烯基、3-己烯基、4-己烯基、5-己烯基、3,3-二甲基-1-丙烯基或2-乙基-1-丙烯基等;
C2~C6炔基指含有2-6个碳原子的直链或支链炔基,例如,乙炔基、2-丙炔基、2-丁炔基、3-丁炔基、1-甲基-2-丙炔基、2-戊炔基、2-戊炔基或2-己炔基等;
苯基C1~C6烷基是指含有1-6个碳原子的直链或支链的饱和烃基的一个碳原子与苯基的碳原子连接,如苯甲基、苯乙基或苯丙基等;
苯基C1~C6烷氧基是指含有1-6个碳原子的直链或支链烷氧基的一个碳原子与苯基的碳原子连接,如苄氧基、-OCH(CH3)Ph、苯乙氧基或苯丙氧基等;苯基C1~C6烷酰基是指含有1-6个碳原子的直链或支链烷酰基的一个碳原子与苯基的碳原子连接,如苯甲酰基、苯乙酰基或苯丙酰基等;
C2~C6链烯氧基是指含有1~3个双键和2-6个碳原子的直链或支链的链烯氧基,如乙 烯氧基、1-丙烯氧基、1-甲基-1-丙烯氧基、2-甲基-1-丙烯氧基、1-戊烯氧基、1,3-戊二烯氧基或2-戊烯氧基等;
C2~C6炔氧基指含有2-6个碳原子的直链或支链炔氧基,例如,乙炔氧基、2-丙炔氧基、2-丁炔氧基、3-丁炔氧基、1-甲基-2-丙炔氧基、2-戊炔氧基或2-己炔氧基等;
C1~C6烷酰基指含有1-6个碳原子的直连或支链烷酰基,如甲酰基、乙酰基、丙酰基、丁酰基、异丁酰基、戊酰基、叔丁酰基或己酰基等;
卤代C1~C6烷酰基是指含有1-6个碳原子的直连或支链烷酰基中的氢原子被1个或多个相同或不同的卤原子取代,例如三氟乙酰基等;
被C1~C6烷基取代的氨基甲酰基是指氨基甲酰基上的氢原子被1个或2个相同或不同的C1~C6烷基取代,例如-CONHMe、-CONHEt、-CON(Me)Et、-CONEt2或-CONMe2等;
羟基C1~C6烷基指含有1-6个碳原子的直链或支链烷基的一个碳原子与羟基连接,如-CH2OH、-CH2CH2OH、-CH(OH)CH3、-CH2CH2CH2OH、-CH2CH2CH2CH2OH或-CH2CH(CH3)CH2OH等;
氨基C1-C6烷基指与含有1-6个碳原子的直链或支链烷基的一个碳原子与氨基连接,如-CH2NH2、-CH2CH2NH2、-CH(NH2)CH3、-CH2CH2CH2NH2或-CH2CH2CH2CH2NH2等;被C1~C6烷基取代的氨基C1~C6烷基是指氨基上的氢原子被1个或2个相同或不同的C1~C6烷基取代,例如-CH2NHMe或-CH2CH2NEt2等;
氨基甲酰基C1~C6烷基是指含有1-6个碳原子的直链或支链烷基的一个碳原子与氨基甲酰基的羰基碳连接,例如-CH2CONH2、-CH2CH2CONH2、-CH(CONH2)CH3或-CH2CH2CH2CONH2等;
被C1~C6烷基取代的氨基甲酰基C1~C6烷基是指氨基甲酰基C1~C6烷基上的氨基氢原子被1个或2个相同或不同的C1~C6烷基取代,例如-CH2CONHMe、-CH2CH2CONHEt、-CH2CH2CONMe2或-CH2CONEt2等;
氰基C1~C6烷基是指含有1-6个碳原子的直链或支链烷基的一个碳原子与氰基连接,如氰基甲基、2-氰基乙基、1-氰基乙基、3-氰基丙基、4-氰基丁基或5-氰基戊基等;
羧基C1~C6烷基指与含有1-6个碳原子的直链或支链烷基的一个碳原子与羧基连接,如羧基甲基、2-羧基乙基、1-羧基乙基、3-羧基丙基、4-羧基丁基或5-羧基戊基等;
或C1~C6烷磺酰基是指含有1-6个碳原子的直链或支链烷磺酰基,如甲磺酰基、乙磺酰基或丙磺酰基等;
卤代C1~C6烷磺酰基是指含有1-6个碳原子的直链或支链烷磺酰基上的氢原子被1个或多个相同或不同的卤原子取代,例如三氟甲磺酰基等;
被C1~C6烷基取代的氨基是指氨基上的氢原子被1个或2个相同或不同的C1~C6烷 基或C1~C6烷酰基取代,例如-NHMe或-NEt2等;
C3-C10环烃基是指含有3-10个碳原子的饱和或不饱和的环烃基,如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环丁烯、环己烯、环己二烯、环戊烯或环戊二烯等;
3-10元单环杂环基指至少包含一个选自N、S和O的杂原子的单环基,如环氧乙烷、氮杂环丁烷基、呋喃基、二氢呋喃基、四氢呋喃基、噻吩基、二氢噻吩基、四氢噻吩基、吡咯基、二氢吡咯基、吡咯烷基、吡唑基、二氢吡唑基、吡唑烷基、三唑基、二氢三唑基、三唑烷基、噻唑基、二氢噻唑基、噻唑烷基、异噻唑基、二氢异噻唑基、异噻唑烷基、噁唑基、二氢噁唑基、噁唑烷基、异噁唑基、二氢异噁唑基、异噁唑烷基、吡喃基、二氢吡喃基、四氢吡喃基、吡嗪基、二氢吡嗪基、四氢吡嗪基、哌嗪基、哒嗪基、二氢哒嗪基、四氢哒嗪基、
Figure PCTCN2015073854-appb-000020
Figure PCTCN2015073854-appb-000021
Figure PCTCN2015073854-appb-000022
Figure PCTCN2015073854-appb-000023
等。
本发明化合物具有以下有益效果:
1)本发明化合物对5-羟色胺1A(5-HT1A)受体和/或多巴胺D2受体和/或5-羟色胺2A(5-HT2A)受体具有良好的活性;尤其对5-HT1A受体具有高活性,部分化合物对5-HT1A受体激动作用的EC50甚至达到1~0.1nM水平。
2)本发明化合物具有多靶点作用的特性,同时作用于多巴胺/5-羟色胺受体的多个亚 型,即,同时对多巴胺D2受体、5-HT1A受体、5-HT2A受体等至少三个靶点具有高活性;由药理实验结果可见,大部分化合物同时具有D2拮抗/5-HT1A激动/5-HT2A拮抗作用,或者具有D2部分激动/5-HT1A激动/5-HT2A拮抗作用;特别地,部分化合物具有极高的D2拮抗/5-HT1A激动/5-HT2A拮抗作用,对各受体的活性甚至达到10-9~10-10M水平;该多靶点作用的特性有利于调节脑内受体平衡,尤其是平衡拮抗5-HT及DA受体,调节DA/5-HT系统,对中枢神经系统类疾病具有较好疗效。
3)由于本发明化合物具有多靶点作用的特性,为中枢神经系统受体调节剂,从而减少或避免了单一的D2拮抗剂或者D2拮抗/5-HT2A拮抗剂导致的副作用,例如锥体外系不良反应(EPS)、代谢紊乱副作用等。
4)本发明化合物不仅活性强,而且口服有效,具有药效剂量低、毒副作用小等特点,对中枢神经系统领域的疾病具有疗效,尤其对重度抑郁症(MDD)、焦虑症、精神分裂症阴性症状、认知功能障碍、帕金森症、多动症等疾病疗效好。
总之,本发明化合物与现有抗精神病药物相比,具有多靶点作用、更低的药效剂量、更少的毒副作用、更好的安全性和耐受性等优点,具有良好的临床应用前景。
具体实施方式
下列制备例、实施例和药理活性测试实施例进一步阐明本发明,但并不限制本发明的范围。
制备例1 1-(苯并噻吩-4-基)哌嗪盐酸盐的制备
Figure PCTCN2015073854-appb-000024
步骤1:
将2-氯-6-氟苯甲醛1-a(500mg,3.15mmol),N-叔丁氧羰基哌嗪(646mg,3.47mmol)溶解到N,N-二甲基甲酰胺(5ml)中,氮气保护下,室温下加入碳酸钾(2.18g,15.77mmol),混合物在80℃搅拌4小时,冷却过滤,加入水(20ml),乙酸乙酯(5ml×3)萃取,用1N盐酸(3ml)洗涤,饱和碳酸氢钠水溶液洗涤,无水硫酸钠干燥,浓缩得固体,用石油醚(50ml)打浆1小时后,过滤得淡黄色固体1-b(1.0g,收率:90%)。1H-NMR(300Hz,DMSO-d6):δppm 10.19(s,1H),7.52(t,1H),7.18(d,2H),3.46(t,4H),2.94(t,4H),1.39(s,9H).
步骤2:
在N2保护下,室温下向N,N-二甲基甲酰胺(50ml)内加入化合物1-b(5g,15.3mmol),巯基乙酸乙酯(1.8g,15.3mmol),碳酸钾(6.2g,44.9mmol),混合物在80℃搅拌5小时,冷却,加入乙酸乙酯(50ml),过滤,滤液中加入水(200ml),用乙酸乙酯萃取,干燥有机相,浓缩得红棕色油状物,放置成固体,用石油醚打浆,过滤得固体1-c(5.9g,收率:98%).ESI-MS(m/z):391.4[M+H]+1H-NMR(300Hz,CDCl3):δppm 8.10(s,1H),7.52(d,1H),7.37(t,1H),6.88(d,1H),4.40(q,2H),3.68(t,4H),3.10(t,4H),1.49(s,9H),1.41(t,3H).
步骤3:
化合物1-c(1.0g,2.5mmol)溶解到1,4-二氧六环(5ml)内,加入4N氢氧化钠水溶液(1.8ml,7.2mmol),混合物在80℃搅拌3小时,冷却到室温,加入水(5ml)和乙酸乙酯(10ml),收集水相,用1N盐酸调节pH至4.0,固体过滤,烘干,得淡黄色固体1-d(0.83g,收率:90%)。1H-NMR(300Hz,DMSO-d6):δppm 13.51(brs,1H),7.96(s,1H),7.65(d,1H),7.40(t,1H),6.95(d,1H),3.55(s,4H),3.00(s,4H),1.41(s,9H).
步骤4:
将化合物1-d(20g,54mmol),氧化亚铜(1g)溶解到喹啉(50ml)内,加热到140℃反应过夜,冷却后过滤,加入水,用乙酸乙酯萃取,加入1N盐酸洗涤至弱酸性,饱和碳酸氢钠水溶液洗涤,无水硫酸钠干燥,滤除干燥剂,浓缩,残留物柱层析,浓缩得到的固体用石油醚打浆,得到类白色固体1-e(10g)。ESI-MS(m/z):319.2[M+H]+1H-NMR(300Hz,CDCl3):δppm 7.57(d,1H),7.41(s,2H),7.27(t,1H),6.88(d,1H),3.66(t,4H),3.09(t,4H),1.50(s,9H).
步骤5:
取化合物1-e(1g,3.14mmol)加入盐酸-甲醇溶液(5ml),50℃反应半小时,将反应液浓缩干,加入乙腈打浆,过滤,得类白色固体(800mg,收率:100%)。ESI-MS(m/z):219.2[M+H]+
制备例2 5-(2-氧代-1,2,3,4-四氢喹啉-7-基)戊基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000025
步骤1:
反应瓶中投入7-羟基-3,4-二氢喹啉-2(1H)-酮2-a(10g,61.3mmol),加入三氯甲烷(100ml)和吡啶(10.6g,134mmol),室温搅拌10分钟,降温到0℃以下,慢慢滴入三氟甲磺酸酐(17.2g,60.99mmol),搅拌30分钟,撤去冰浴,室温反应1小时,过滤出固体,分别用1M的硫 酸氢钾水溶液和水洗涤两次,无水硫酸钠干燥,浓缩,柱层析得淡黄色固体2-b(12g,收率:67%)。
步骤2:
反应瓶中投入化合物2-b(18g,61.0mmol),双(三苯基磷)二氯化钯(3.6g,5.12mmol)和碘化亚铜(3.96g,20.8mmol),氮气置换三次,注入4-戊炔-1-醇(5g,59.5mmol),三乙胺(26g,25.7mmol)和N,N-二甲基甲酰胺(100ml),80℃过夜反应。反应液降温到室温,加入乙酸乙酯(300ml),过滤出固体,滤液用1N盐酸洗涤2次,水洗2次,无水硫酸钠干燥,浓缩,残留物柱层析得白色固体2-c(5.5g,收率:39%)。
步骤3:
250mL单口瓶中投入化合物2-c(5.5g,24.0mmol),加入甲醇(55ml)溶解,再投入10%的Pd/C(1.2g),氢气置换3次,升温到55℃反应15小时,降温,过滤,甲醇淋洗,浓缩,柱层析得白色固体2-d(3.3g,收率:60%)。ESI-MS(m/z):234.3[M+H]+1H-NMR(300Hz,CDCl3):δppm 1.43(m,2H),1.64(m,4H),2.59(t,2H),2.65(t,2H),2.95(t,2H),3.67(t,2H),6.60(s,1H),6.83(d,1H),7.08(d,1H),8.01(brs,1H).
步骤4:
取化合物2-d(0.5g,2.14mmol),加入二氯甲烷(5ml),三乙胺(0.6ml,4.28mmol),冰浴下缓慢注入甲磺酰氯(0.2ml,2.56mmol),15分钟后移至室温,2小时后将反应液中加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,浓缩得到白色固体(0.6g,收率:90%)。ESI-MS(m/z):312.1[M+H]+
制备例3 5-(2-氧代-1,2-二氢喹啉-7-基)戊基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000026
取制备例2产物(0.8g,2.57mmol)加入1,4-二氧六环(5ml),2,3-二氯-5,6-二氰基-1,4-苯醌(0.875mg,3.85mmol),95℃加热反应3小时,将反应液冷却,过滤除去不溶物,滤液加入二氯甲烷,依次用饱和碳酸氢钠溶液,硫代硫酸钠溶液和饱和食盐水洗涤,无水硫酸钠干燥,浓缩,柱层析得到白色固体(0.6g,收率:75%)。ESI-MS(m/z):310.1[M+H]+
制备例4 4-(2-氧代-1,2,3,4-四氢喹啉-7-基)丁基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000027
参照制备例2的方法,由3-丁炔-1-醇替代4-戊炔-1-醇,制备得到淡黄色油状物,收率:34%。ESI-MS(m/z):298.1[M+H]+1H-NMR(300Hz,CDCl3):δppm 8.43(s,1H),7.07(d,1H),6.79(d,1H),6.60(s,1H),4.23(t,2H),3.00(s,3H),2.93(t,2H),2.62(m,4H),1.75(m,4H).
制备例5 4-(2-氧代-1,2-二氢喹啉-7-基)丁基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000028
参照制备例3的方法,以制备例4产物为原料,制备得到灰色固体,收率:61%。ESI-MS(m/z):296.1[M+H]+1H-NMR(300Hz,CDCl3):δppm 12.37(s,1H),7.85(d,1H),7.52(d,1H),7.26(s,1H),7.09(d,1H),6.72(d,1H),4.25(t,2H),3.00(s,3H),2.78(t,2H),1.79(m,4H).
制备例6 2-(2-氧代-1,2,3,4-四氢喹啉-7-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000029
步骤1:
取化合物2-b(20g,67.7mmol),加入氯化锂(8.7g,203.1mmol),双(三苯基膦)二氯化钯(5.7g,8.12mmol),N,N-二甲基甲酰胺(200ml)作溶剂。氮气置换后注入三丁基乙烯基锡(21ml,74.4mmol),100℃加热过夜。过滤不溶物,滤液加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,干燥,浓缩,残留物经柱层析得化合物6-a(8.0g,收率:68%)。
步骤2:
取化合物6-a(8g,46.2mmol),加入过硫酸氢钾复合物(Oxone,43.6g,69.3mmol),碳酸氢钠(29.2g),丙酮-水(400ml∶400ml)做溶剂,室温搅拌2小时,过滤除去不溶物,滤液中加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,干燥,浓缩,残留物经柱层析得目标物6-b(5.7g,收率:65%)。ESI-MS(m/z):190.1[M+H]+1H-NMR(300Hz,CDCl3):δppm8.42(brs,1H),7.16(d,1H),6.94(dd,1H),6.71(d,1H),3.85(dd,1H),3.15(dd,1H),2.98(t,2H),2.78(dd,1H),2.66(t,2H).
步骤3:
反应瓶中加入化合物6-b(3.6g,19.0mmol),甲酸铵(3.0g,47.6mmol),乙酸乙酯(100ml),甲醇(100ml)和10%Pd/C(460mg),氮气保护下加热回流过夜。过滤,滤液浓缩干,柱层析得化合物6-c(1.5g,收率:41%)。ESI-MS(m/z):192.0[M+H]+1H-NMR(300Hz,CDCl3):δppm 8.87(brs,1H),7.07(d,1H),6.84(d,1H),6.69(s,1H),3.84(t,2H),3.78(s,1H),2.90(t,2H),2.80(t,2H),2.59(t,2H).
步骤4:
取化合物6-c(1.5g,7.85mmol)溶于二氯甲烷(60ml),搅拌下加入三乙胺(3ml,23.55mmol),冰浴下滴加甲磺酰氯(0.92ml,11.77mmol),加完移至室温反应3小时。加入适量水,二氯甲烷萃取,有机相干燥浓缩,所得粗品经柱层析得白色固体(1.3g,收率:61%)。1H-NMR(300Hz,CDCl3):δppm 8.97(brs,1H),7.11(d,1H),6.86(dd,1H),6.69(d.1H),4.39(t, 2H),3.00(t,2H),2.94(t,2H),2.93(s,3H),2.63(t,2H).
制备例7 2-(2-氧代-1,2-二氢喹啉-7-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000030
参照制备例3的方法,以制备例6产物为原料,制备得到白色固体,收率:75%。ESI-MS(m/z):268.1[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 11.74(brs,1H),7.87(d,1H),7.61(d,1H),7.18(s,1H),7.12(d,1H),6.45(d,1H),4.44(t,2H),3.13(s,3H),3.07(t,2H).
制备例8 1-(2-氟苯并噻吩-4-基)哌嗪的制备
Figure PCTCN2015073854-appb-000031
步骤1:
将化合物1-e(1g,3.14mmol)加入干燥的四氢呋喃溶解,氮气置换后置于-78℃搅拌,30分钟后滴入2.5M正丁基锂的正己烷溶液(1.65ml,4.08mmol),搅拌3小时,将氟代双苯磺酰胺(1.5g,4.71mmol)用四氢呋喃(5ml)溶解,缓慢滴入反应体系,维持-78℃反应1小时后自然升温,搅拌反应过夜,加入饱和氯化铵溶液淬灭反应,加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,浓缩,残留物经柱层析得油状物8-a(600mg,收率:57%)。
步骤2:
取化合物8-a(600mg),加入二氯甲烷(1ml)和三氟醋酸(1ml),室温下搅拌1小时,浓缩反应液,加入饱和碳酸氢钠溶液(5ml),析出黄色固体,过滤,烘干得淡黄色固体(400mg,收率95%)。ESI-MS(m/z):237.1[M+H]+
制备例9 1-(2-氯苯并噻吩-4-基)哌嗪的制备
Figure PCTCN2015073854-appb-000032
步骤1:
取化合物1-e(900mg,2.83mmol)置于干燥的三口瓶中,氮气保护下加入四氢呋喃(3ml)溶清,降温至-78℃,注入2.5M正丁基锂的正己烷溶液(1.47ml,3.67mmol),维持-78℃反 应3小时。取N-氯代丁二酰亚胺(677mg,5.09mmol)溶于四氢呋喃(3ml),缓慢注入反应体系中,半小时后移至室温下搅拌过夜,加入饱和氯化铵溶液淬灭反应,加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析得油状物9-a(540mg,收率:54%)。
步骤2:
取化合物9-a(540mg,1.53mmol),加入二氯甲烷(1ml)和三氟乙酸(1ml),室温下搅拌3小时。浓缩溶剂,加入饱和碳酸氢钠溶液(3ml),析出固体,过滤,乙腈打浆得到白色固体(210mg,收率:54%)。ESI-MS(m/z):253.2[M+H]+1HNMR(300MHz,DMSO-d6):δppm8.19(brs,1H),7.46-7.68(m,2H),7.32(t,1H),6.98(d,1H),3.25(s,4H),3.16(s,4H).
制备例10 3-(2-氯乙基)-2-甲基-6,7-二氢吡啶并[1,2-a]嘧啶-4-酮的制备
Figure PCTCN2015073854-appb-000033
步骤1:
取化合物10-a(1g,4.13mmol),加入三乙胺(0.85ml,6.19mmol)和二氯甲烷(10ml),冰浴下缓慢滴加甲磺酰氯(0.38ml,4.95mmol),加完后室温下反应30分钟,加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,干燥,浓缩得油状物10-b(1.23g,收率:93%)。
步骤2:
取化合物10-b(520mg,1.62mmol),加入溴化锂(418mg,4.86mmol),碳酸锂(358mg,4.86mmol)和N,N-二甲基甲酰胺(5ml),115℃加热2小时,加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,干燥,浓缩得黄色固体(400mg,收率:100%)。ESI-MS(m/z):225.1[M+H]+1H-NMR(300Hz,CDCl3):δppm 2.35(s,3H),2.54(m,2H),3.01(t,2H),3.76(t,2H),4.14(t,2H),6.35(dt,1H),6.64(m,1H).
制备例11 6-(5-氯正戊基)-2-甲基喹唑啉-4(3H)-酮的制备
Figure PCTCN2015073854-appb-000034
步骤1:
取5-氯-2-硝基苯甲酸11-a(5g,24.8mmol),加入氢氧化钾(13.8g,248mmol)的水溶液 (40ml),加热回流24小时。冰浴下滴加浓盐酸调pH至2,析出固体,过滤,烘干得到白色固体11-b(4.1g,收率:90%)。
步骤2:
取化合物11-b(4.1g,22.4mmol),加入氯化亚砜(26.8mmol)和甲醇(10ml),加热回流5小时。浓缩反应液,加入乙酸乙酯,水萃取,饱和食盐水洗,干燥,浓缩得目标物11-c(3.19g,收率:72%)。
步骤3:
取化合物11-c(2.66g,13.5mmol),加入吡啶(67.5mmol)和二氯甲烷(10ml)。冰浴下缓慢滴入三氟甲磺酸酐(20.2mmol),滴完室温下搅拌6小时。加入二氯甲烷(100ml)稀释,1N盐酸(70ml)洗两次,饱和食盐水洗,有机相用无水硫酸钠干燥,浓缩,得到棕黄色固体11-d(3.86g,收率:87%)。
步骤4:
取化合物11-d(2.83g,8.6mmol),4-戊炔-1-醇(789mg,9.4mmol),双(三苯基膦)二氯化钯(603mg,0.86mmol),碘化亚铜(245mg,1.29mmol),三乙胺(5.9ml,43mmol)和N,N-二甲基甲酰胺(10ml)混合,室温下搅拌,反应过夜。将反应液用二氯甲烷稀释,稀盐酸洗两次,有机相干燥浓缩,柱层析得油状物11-e(2.2g,收率:97%)。1H-NMR(300Hz,CDCl3):δppm7.88(d,1H),7.67(d,1H),7.57(dd,1H),3.92(s,3H),3.80(q,2H),2.58(t,2H),1.87(m,2H),1.49(t,1H).
步骤5:
取化合物11-e(1.04g,3.95mmol),加入钯碳(150mg)和乙醇(5ml),50℃下搅拌反应24小时,过滤除去不溶物,滤饼用甲醇洗几次,浓缩滤液,加入氯化氢-乙醇溶液成盐,在丙酮/甲基叔丁基醚中打浆,过滤,滤饼烘干,得黄色固体11-f(840mg,收率:89%)。ESI-MS(m/z):238.1[M+H]+
步骤6:
取化合物11-f(455mg,1.91mmol),加入乙腈(3ml)和氯化氢/二氧六环溶液(3ml),封管70℃反应过夜。浓缩反应液,用饱和碳酸氢钠溶液调pH至中性,乙酸乙酯萃取,有机相干燥,浓缩,硅胶柱层析,得目标化合物(280mg,收率:55%)。ESI-MS(m/z):265.1[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 12.11(brs,1H),7.86(d,1H),7.60(dd,1H),7.48(d,1H),3.61(t,2H),2.69(t,2H),2.32(s,3H),1.73(m,2H),1.62(m,2H),1.39(m,2H).
制备例12 1-(2,3-二氢苯并噻吩-4-基)哌嗪盐酸盐的制备
Figure PCTCN2015073854-appb-000035
步骤1:
将四氢铝锂(4.6g,121mmol)加入到含干燥四氢呋喃(20ml)的烧瓶中,在冰浴条件下,将化合物12-a(10g,53.2mmol)溶于四氢呋喃(100ml)的溶液滴加到上述反应液中,滴加完后,室温搅拌20分钟,然后加热回流,3小时后停反应,缓慢加入十水硫酸钠,直至气泡不再放出,抽滤,干燥滤液,浓缩后柱层析得产物12-b(7.8g,收率:85%)。
步骤2:
将化合物12-b(6.7g,38.5mmol),三乙胺(4.66g,46.2mmol)和二氯甲烷(80ml)加入烧瓶中,室温条件下,将对甲苯磺酰氯(7.34g,38.5mmol)溶于二氯甲烷(20ml)滴加到上述溶液中,反应完后,加1N盐酸和水,用二氯甲烷萃取,有机相用水和饱和碳酸氢钠溶液洗涤,干燥,浓缩,得产物12-c(10g,收率:80%)。
步骤3:
将化合物12-c(10g,30.4mmol),九水硫化钠(8.75g,36.4mmol)和N-甲基吡咯烷酮(50ml)加入烧瓶中,150℃加热4小时,加水(100ml),用甲基叔丁基醚(160ml)分2次萃取,有机相用饱和食盐水(200ml)分两次洗,干燥,浓缩,柱层析得产物12-d(1.8g,收率:35%)。
步骤4:
将化合物12-d(500mg,2.94mmol),无水哌嗪(379mg,4.41mmol),叔丁醇钠(423mg,4.41mmol),2,2′-双二苯膦基-1,1′-联萘(83mg,0.13mmol),醋酸钯(10mg,0.044mmol)和干燥的甲苯(10ml)加入烧瓶中,置换3次氮气,115℃回流24小时,浓缩反应液,残留物经柱层析得油状物,加入氯化氢-乙醇溶液成盐,用乙腈打浆得白色固体(280mg,收率:43%)。ESI-MS(m/z):221.1[M+H]+
制备例13 4-(哌嗪-1-基)噻吩并[2,3-d]嘧啶三氟乙酸盐的制备
Figure PCTCN2015073854-appb-000036
步骤1:
将2-氨基噻吩-3-羧酸甲酯(2.0g,12.7mmol)溶于甲酰胺(60ml)中,搅拌,加热至190℃,反应4小时。冷却,倒入水(200ml)中,正丁醇(50ml×4)萃取,合并有机相,无水硫酸钠干燥。浓缩,残留物经柱层析得淡黄色固体13-b(1.2g,收率:63%)。ESI-MS(m/z):153.0[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 7.39(d,1H),7.57(d,1H),8.12(s,1H),12.49(brs,1H).
步骤2:
将三氯氧磷(10ml)加入到化合物13-b(500mg,3.23mmol)中,搅拌,回流过夜。将反应液冷却至室温,倒入强烈搅拌的冰水混合物中,继续搅拌30分钟,二氯甲烷(50ml×3)萃取,合并有机相,饱和食盐水洗一次,无水硫酸钠干燥,浓缩,得到黄色固体13-c(600mg,收率:100%)。ESI-MS(m/z):170.9[M+H]+1H-NMR(300Hz,CDCl3):δppm 7.46(d,1H),7.64(d,1H),8.87(s,1H).
步骤3:
将化合物13-c(600mg,3.53mmol)溶于四氢呋喃(20ml)中,搅拌,加入N-叔丁氧羰基哌嗪(984mg,5.3mmol)和N,N-二异丙基乙胺(910mg,7.06mmol),回流2小时,将反应液冷却至室温,浓缩,柱层析得白色固体13-d(900m g,收率:79.4%)。ESI-MS(m/z):321.2[M+H]+1H-NMR(300Hz,CDCl3):δppm 1.48(s,9H),3.61(m,4H),3.92(m,4H),7.32(m,2H),8.50(s,1H).
步骤4:
取化合物13-d(900mg)溶于二氯甲烷(10ml)中,冷至0℃,加入三氟乙酸(1ml),加完后室温搅拌反应过夜。将反应液减压浓缩至干,残余油状物用甲基叔丁基醚/甲醇(15ml/1ml)混合溶剂打浆,过滤,烘干得黄色固体(606mg,收率:98%)。
制备例14 4-(苯并噻吩-4-基)-1,2,3,6-四氢吡啶盐酸盐的制备
Figure PCTCN2015073854-appb-000037
步骤1:
500mL的三口瓶中加入四氢呋喃(250ml)和二异丙基胺(28ml),N2保护,-10℃~0℃下滴加正丁基锂(78ml),滴加完毕,0℃下继续反应30分钟,冷却至-78℃,滴加间氟溴苯 (21ml),滴完后此温度下继续反应1小时,滴加N,N-二甲基甲酰胺(17ml),滴完后-78℃下反应20分钟,然后快速加入冰醋酸(28ml)和水(200ml),升温至15℃,用乙酸乙酯(500ml×2)萃取,有机相用水(300ml×2)洗,后用饱和食盐水(200ml×1)洗,无水硫酸钠干燥,浓缩得粗品,经柱层析得目标物14-b(32g,收率:81%)。
步骤2:
三口瓶中加入N,N-二甲基甲酰胺(250ml)和碳酸钾(51.5g,373mmol),N2保护,室温下滴加巯基乙酸乙酯(13.7ml),滴加完毕,快速加入化合物14-b(25.2g,124mmol),后加热至60℃,反应过夜,反应完毕加入乙酸乙酯(300ml)稀释,过滤,滤液用水洗1次,再用饱和氯化钠溶液洗1次,干燥有机相,浓缩得粗品,经柱层析得化合物14-c(23g,收率:66%)。
步骤3:
500mL的单口瓶中加入化合物14-c(20g,70.4mmol),四氢呋喃(120ml)和水(100ml),搅拌下加入4N氢氧化钠水溶液(40ml),然后加热至70℃反应1小时。用乙酸乙酯(70ml×2)萃取反应液,水相用浓盐酸酸化至pH 1~2,有大量固体析出,过滤,滤饼用水洗3次,所得固体烘干得化合物14-d(16g,收率:88%)。
步骤4:
25mL的单口瓶中加入化合物14-d(14g,54.6mmol),二甲基亚砜(140ml),搅拌下加入碳酸银(15g,54.3mmol)和醋酸(163mg),然后加热至120℃,反应过夜,冷却至室温,加入乙酸乙酯(100ml),过滤,滤液加入适量水,乙酸乙酯(200ml×3)萃取,干燥有机相,浓缩,柱层析得无色透明油状物14-e(11g,收率:95%)。
步骤5:
在氮气保护下,将镁屑(113mg,4.7mmol)加入到四氢呋喃(5ml)内,搅拌5min后,加热到60℃。将化合物14-e(1.0g,4.7mmol)的四氢呋喃(5ml)溶液中的(2ml)滴加入上述体系中,保温60℃搅拌,当颜色变淡青蓝色时,表明反应引发开始。保持微回流条件下,滴加剩余的化合物14-e的四氢呋喃溶液,反应颜色变淡黄色,继续回流反应2小时,至镁屑消失,或者近乎消失,表明格氏试剂14-f制备完成。
步骤6:
在0℃下,向新制备的格氏试剂14-f内滴加N-叔丁氧羰基哌啶酮(1.0g,5mmol)的四氢呋喃(5ml)溶液。滴加完毕后渐渐升温至室温,室温搅拌1小时后,冷却至0℃,加入饱和氯化铵溶液,乙酸乙酯萃取,饱和食盐水洗涤,干燥,浓缩,残留物经柱层析得化合物14-g(700mg,两步收率44%)。
步骤7:
将化合物14-g(120mg,0.36mmol)溶解到甲苯(5ml)和6N盐酸(5ml)内,加热到100℃,反应过夜,冷却到室温,收集水相,0℃下用10%的氢氧化钠水溶液调节pH至9.0,用二 氯甲烷萃取,干燥,浓缩,在乙酸乙酯中用氯化氢-乙醇溶液成盐,过滤,得目标物(50mg,收率:65%)。ESI-MS(m/z):216.1[M+H]+1H-NMR(300Hz,MeOH-d4):δppm 7.89(d,1H),7.64(d,1H),7.56(dd,1H),7.37(t,1H),7.28(dd,1H),5.96(m,1H),3.93(m,2H),3.55(t,2H),2.85(m,2H).
制备例15 5-(2-氯乙基)二氢吲哚-2-酮的制备
Figure PCTCN2015073854-appb-000038
步骤1:
取二氢吲哚-2-酮15-a(2g,15.03mmol),加入二氯甲烷(10ml),三氯化铝(7g,52.60mmol),冰浴下将氯乙酰氯(2.26ml,30.06mmol)的二氯甲烷(10ml)溶液缓慢滴入反应液中,1小时后撤除冰浴,加热回流2小时,冷却,将反应液倒入冰水中搅拌,调节pH至强酸性,析出大量固体,抽滤,烘干得到浅灰白色固体(2.811g,收率:90%)。
步骤2:
取化合物15-b(1g,4.78mmol),加入三氟乙酸(10ml),冰浴下加入三乙基硅烷(1.5ml),10分钟后撤除冰浴,室温下搅拌,2小时后将反应液倒入水中,加入乙酸乙酯萃取2次,弃去水相,有机相用饱和碳酸氢钠溶液洗涤,饱和食盐水洗涤,干燥,浓缩得到土黄色固体(0.9g,收率:96%)。ESI-MS(m/z):195.9[M+H]+1H-NMR(300Hz,CDCl3):δppm 7.69(brs,1H),7.10(s,1H),7.07(d,1H),6.80(d,1H),3.68(t,2H),3.52(s,2H),3.02(t,2H).
制备例16 2-(6-氯-2-氧代-1,2,3,4-四氢喹啉-7-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000039
取制备例6产物(440mg,1.63mmol)加入N,N-二甲基甲酰胺(8ml),N-氯代丁二酰亚胺(228mg,1.71mmol),100℃下加热4小时,加入适量水,二氯甲烷萃取,有机相用饱和食盐水洗1次,无水硫酸钠干燥,浓缩,柱层析得白色固体(330mg,收率:67%)。ESI-MS(m/z):304.0[M+H]+
制备例17 3-(2-氯乙基)-9-氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮的制备
Figure PCTCN2015073854-appb-000040
将3-(2-氯乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮10-a(500mg,2.06mmol)加入二氯甲烷(10ml)中,在冰浴条件下将含有二乙氨基三氟化硫(0.32ml, 2.47mmol)的二氯甲烷溶液(8ml)滴加入上述溶液中,室温下搅拌3小时,加水淬灭,加入饱和碳酸氢钠溶液调节pH至中性,用二氯甲烷萃取,无水硫酸钠干燥,浓缩,柱层析得白色晶体(370mg,收率:74%)。ESI-MS(m/z):245.1[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 1.92(m,2H),2.15(m,2H),2.29(s,3H),2.90(t,2H),3.55(m,1H),3.73(t,2H),4.00(dt,1H),5.26-5.47(dt,1H).
制备例18 5-(2-氯乙基)-4-甲基噻唑的制备
Figure PCTCN2015073854-appb-000041
将2-(4-甲基噻唑-5-基)乙醇18-a(500mg,3.49mmol)加入二氯亚砜(4ml)中,回流反应3小时,减压蒸除溶剂,残液用甲苯带两次,得淡黄色固体(680mg,收率:98%)。ESI-MS(m/z):162.0[M+H]+
制备例19 5-(2-氯乙基)-1H-苯并咪唑-2(3H)-酮的制备
Figure PCTCN2015073854-appb-000042
步骤1:
取1H-苯并咪唑-2(3H)-酮19-a(500mg,3.73mmol)加入无水氯化铝(1.98g,14.9mmol)和四氯乙烷(3ml)中。另取氯乙酰氯(843mg,7.46mmol)溶于四氯乙烷(3ml)中,冰浴下将其滴加入反应体系中,加完100℃加热1小时,冷却至室温,有大量黑色胶状物,加冰块,有淡黄色固体析出,加入4N盐酸(20ml),搅拌6小时,过滤,滤饼在异丙醇中打浆,过滤,滤饼烘干得灰白色固体19-b(730mg,收率:93%)。EI-MS(m/z):210。
步骤2:
将化合物19-b(350mg,1.67mmol)加入三氟乙酸(5ml)中,冰浴下滴加三乙基硅烷(0.66ml,4.17mmol),室温下反应过夜。浓缩反应液,加入饱和碳酸氢钠水溶液,有固体析出,过滤,滤饼用冰水洗三次,烘干得淡黄色固体(310mg,收率:95%)。EI-MS(m/z):196。
制备例20 6-(2-氯乙基)苯并噻唑-2(3H)-酮的制备
Figure PCTCN2015073854-appb-000043
步骤1:
将苯并噻唑-2(3H)-酮20-a(500mg,3.307mmol)悬浮于二硫化碳(8ml)中,冰浴下分批加 入无水氯化铝(2.65g,19.841mmol),再缓慢滴加氯乙酰氯(324ul,4.299mmol),加毕让其在室温下搅拌10分钟,再回流1.5小时。向反应液加少量冰水淬灭反应,浓缩除去二硫化碳,残液加冰水(15ml),大量粉红色固体析出,加入4N盐酸(10ml),室温下搅拌2小时,过滤,滤饼用冰水洗两遍,滤饼烘干得粉红色固体20-b(740mg,收率:98%)。ESI-MS(m/z):227.9[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 12.35(s,1H),8.25(d,1H),7.89(dd,1H),7.20(d,1H),5.13(s,2H).
步骤2:
将化合物20-b(360mg,1.586mmol)加到三氟乙酸(5ml)中,冰浴下滴加三乙基硅烷(630ul,3.965mmol),加毕在10℃下反应过夜。浓缩反应液,残液加入水,有固体析出,用二氯甲烷萃取,有机相用无水硫酸钠干燥,过滤,浓缩得粗品385mg,柱层析得目标物(280mg,收率:82.5%)。
制备例21 6-(2-氯乙基)-2H-1,4-苯并噁嗪-3(4H)-酮的制备
Figure PCTCN2015073854-appb-000044
步骤1:
2H-1,4-苯并噁嗪-3(4H)-酮21-a(500mg,3.356mmol)悬浮于二氯甲烷(8ml)中,冰浴下分批加入无水氯化铝(895mg,6.712mmol),再缓慢滴加氯乙酰氯(330ul,4.363mmol),加毕让其在室温下搅拌10分钟,再回流5小时,向反应液加少量冰水淬灭反应,浓缩除去二氯甲烷,残液加冰水(10ml),加入4N盐酸(5ml),室温下搅拌2小时,过滤,滤饼用冰水洗2次,烘干得淡黄色固体21-b(690mg,收率:91%)。
步骤2:
将化合物21-b(370mg,1.637mmol)加到三氟乙酸(4ml)中,冰浴下滴加三乙基硅烷(640ul,4.093mmol),加毕在10℃下反应过夜。浓缩反应液,残液中加入水,用二氯甲烷萃取,有机相用无水硫酸钠干燥,过滤,浓缩得粗品375mg,在石油醚中打浆,过滤,烘干得淡黄色固体(316mg,收率:91%)。1H-NMR(300Hz,DMSO-d6):δppm 10.68(brs,1H),6.86(d,1H),6.81(dd,1H),6.74(d,1H),4.51(s,2H),3.75(t,2H),2.90(t,2H).
制备例22 6-(2-氯乙基)-2H-1,4-苯并噻嗪-3(4H)-酮的制备
Figure PCTCN2015073854-appb-000045
步骤1:
冰浴下将无水氯化铝(1.62g,12.15mmol)分批加入到氯乙酰氯(912ul,12.11mmol)的二氯甲烷溶液(6ml)中,加毕让其在冰浴下搅拌10分钟,再分批加入2H-1,4-苯并噻嗪-3(4H)-酮22-a(500mg,3.027mmol),加毕,反应液继续在冰浴下搅拌4小时,移至室温下反应过夜。向反应液中加冰水(10ml),大量淡黄色固体析出,加入4N盐酸(10ml),室温下搅拌2小时,过滤,滤饼用冰水洗2次,烘干得淡黄色固体22-b(700mg,收率:95.6%)。1H-NMR(300Hz,DMSO-d6):δppm 10.76(brs,1H),7.57(dd,1H),7.50(m,2H),5.11(s,2H),3.54(s,2H).
步骤2:
将化合物22-b(350mg,1.446mmol)加到三氟乙酸(4ml)中,冰浴下滴加三乙基硅烷(578ul,3.615mmol),加毕,反应液在30℃下反应过夜。将反应液倒入冰水中,用二氯甲烷萃取,有机相用无水硫酸钠干燥,过滤,浓缩,柱层析得淡黄色固体(285mg,收率:86.8%)。EI-MS(m/z):227。
制备例23 5-(4-氧代-3,4-二氢喹唑啉-6-基)戊基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000046
步骤1:
取制备例11的中间体11-f(395mg,1.66mmol),加入甲酸铵(1.04g,16.6mmol)和甲酰胺(5ml),135℃加热60小时,将反应液倒入冰水中,用乙酸乙酯萃取3次,合并有机相,水洗1次,无水硫酸钠干燥,浓缩,柱层析得目标物23-a(150mg,收率:38%)。ESI-MS(m/z):233.2[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 12.17(brs,1H),8.03(s,1H),7.90(d,1H),7.65(dd,1H),7.57(d,1H),4.36(t,1H),3.37(m,2H),2.70(t,2H),1.60(m,2H),1.43(m,2H),1.30(m,2H).
步骤2:
将三乙胺(125ul,0.87mmol)加入到化合物23-a(100mg,0.43mmol)的二氯甲烷(5ml)溶液中,冰浴下将甲磺酰氯(51ul,0.65mmol)加入上述反应体系中,室温下反应2小时,加入二氯甲烷稀释反应液,用饱和氯化铵水溶液洗涤,干燥有机相,浓缩,柱层析得白色固体(71mg,收率:53%)。ESI-MS(m/z):311.1[M+H]+
制备例24 2-(2-氯乙基)喹唑啉-4(3H)-酮的制备
Figure PCTCN2015073854-appb-000047
取邻氨基苯甲酸乙酯24-a(1.0g,6.62mmol),丙烯腈(0.88ml,13.24mmol)溶于二氧六环中,冰浴下缓慢加入氯化氢/二氧六环(10ml),搅拌4小时,封管中升温至80℃过夜,将溶剂蒸干,加入水,二氯甲烷,用氨水调节pH至7~8,过滤,固体烘干后用甲基叔丁基醚打浆,过滤,烘干得淡黄色固体(660mg,收率:52%)。ESI-MS(m/z):209.1[M+H]+
制备例25 5-(3-氯丙基)二氢吲哚-2-酮的制备
Figure PCTCN2015073854-appb-000048
步骤1:
冰浴下将3-氯丙酰氯(538ul,5.63mmol)滴加到无水氯化铝(2g,15.02mmol)的二硫化碳悬浮液中,搅拌10分钟,加入二氢吲哚-2-酮15-a(500mg,3.75mmol),室温下搅拌15分钟后回流反应3小时,除去二硫化碳,反应液中加入冰块,析出大量固体,加入4N盐酸(5ml),搅拌,过滤,滤饼用冰水洗3次,烘干得粗品,在乙酸乙酯中打浆,过滤,烘干得淡粉红色固体25-a(777mg,收率:92.4%)。1HNMR(300MHz,DMSO-d6):δppm 10.77(s,1H),7.86(d,1H),7.81(s,1H),6.89(d,1H),3.89(t,2H),3.54(s,2H),3.44(t,2H).
步骤2:
将三乙基硅烷(900ul,5.58mmol)滴加到化合物25-a(500mg,2.32mmol)的三氟乙酸溶液中,30℃下反应过夜。将反应液倒入冰水中,用二氯甲烷萃取3次,合并有机相,干燥,浓缩,在石油醚:丙酮(30∶1,体积比)的混合液中打浆,过滤,滤饼烘干得淡粉红色固体(430mg,收率:92%)。
制备例26 4-(哌嗪-1-基)噻吩并[2,3-c]吡啶三氟乙酸盐的制备
Figure PCTCN2015073854-appb-000049
步骤1:
将二异丙胺(2.4g,24.0mmol)溶于无水四氢呋喃(20ml)中,氮气保护,搅拌下在0℃滴加正丁基锂的正己烷溶液(9.6ml,24.0mml),滴完后反应30分钟,加入无水四氢呋喃 (30ml),冷却至-78℃,滴加3,5-二溴吡啶26-a(4.7g,20mml)的无水四氢呋喃(50ml)溶液,滴完后继续反应30分钟,滴加甲酸甲酯(2.4g,40mml),30分钟后升温至室温,倒入饱和碳酸氢钠水溶液(100ml)中,乙酸乙酯(50ml×3)萃取,合并有机相,无水硫酸钠干燥,浓缩,残留物经柱层析得淡黄色固体26-b(4.0g,收率:75%)。1HNMR(300MHz,DMSO-d6):δppm 10.06(s,1H),8.87(s,2H).
步骤2:
将化合物26-b(2.0g,7.5mmol)加入四氢呋喃(10ml)中,在搅拌下0℃加入巯基乙酸乙酯(0.68ml,7.5mmol),继续搅拌反应1小时后,加入碳酸铯(2.46g,7.5mmol),室温搅拌过夜,过滤反应液,滤液浓缩,残留物经柱层析得白色固体26-c(1.6g,收率:74%)。ESI-MS(m/z):286.1[M+H]+1HNMR(300MHz,DMSO-d6):δppm 9.36(s,1H),8.71(s,1H),8.01(s,1H),4.39(q,2H),1.34(t,3H).
步骤3:
将化合物26-c(2.0g,7.0mmol)溶于四氢呋喃(40ml)中,搅拌下加入一水合氢氧化锂(588mg,14.0mmol)和甲醇(5ml),加毕在室温下继续反应2小时,用2N盐酸调节pH至4,浓缩除去四氢呋喃和甲醇,过滤,滤饼真空干燥得白色固体26-d(1.0g,收率:55%)。1HNMR(300MHz,DMSO-d6):δppm 9.33(s,1H),8.69(s,1H),7.94(s,1H).
步骤4:
将化合物26-d(500mg,1.9mol)加入二苯醚(6ml)中,加热至230℃,搅拌反应2小时,冷却至室温,加入二氯甲烷稀释,加入硅胶拌样,柱层析得白色固体26-e(300mg,收率:73%)。ESI-MS(m/z):213.8[M+H]+1HNMR(300MHz,CDCl3):δppm 9.10(s,1H),8.60(s,1H),7.89(d,1H),7.54(d,1H).
步骤5:
将化合物26-e(1.5g,7.0mmol),N-叔丁氧羰基哌嗪(2.6g,14.0mml),2,2′-双二苯膦基-1,1′-联萘(436mg,0.7mml),三(二亚苄基丙酮)二钯(320.3mg,0.35mmol)和叔丁醇钠(1.34g,14.0mml)分散在无水甲苯(50ml)中,氮气置换三次,100℃搅拌反应过夜,反应液降温至室温,加入硅胶拌样,柱层析得淡黄色固体26-f(1.7g,收率:77%)。ESI-MS(m/z):320.3[M+H]+
步骤6:
取化合物26-f(687mg)溶于二氯甲烷(10ml)中,冷至0℃,加入三氟乙酸(3.0ml),加完后室温搅拌反应过夜。将反应液减压浓缩至干,残余油状物用甲基叔丁基醚/甲醇(15ml/1ml)混合溶剂打浆,过滤,烘干得黄色固体(941mg,收率:98%)。1HNMR(300MHz,CDCl3):δppm 8.90(s,1H),8.06(s,1H),7.89(d,1H),7.51(d,1H),3.68(t,4H),3.21(t,4H),1.49(s,9H).
制备例27 3-(2-氧代-1,2,3,4-四氢喹啉-7-基)丙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000050
步骤1:
将化合物2-b(10.0g,33.9mmol),三苯基膦二氯化钯(1.2g,1.7mmol),三苯基膦(888.2mg,3.39mmol),碘化亚酮(644mg,3.39mol)和二异丙胺(17.17g,170.0mmol)分散在无水N,N-二甲基甲酰胺(100ml)中,搅拌,氮气置换三次,加热至80℃,滴加丙炔醇(9.5g,170.0mmol),加毕继续反应4小时,冷却,浓缩除去N,N-二甲基甲酰胺,残留物经柱层析得淡黄色固体(4.0g,收率:58%)。ESI-MS(m/z):202.1[M+H]+1HNMR(300MHz,DMSO-d6):δppm10.14(s,1H),7.14(d,1H),6.93(dd,1H),6.85(d,1H),5.30(t,1H),4.25(d,2H),2.85(t,2H),2.42(t,2H).
步骤2:
将化合物27-a(2.0g,10.0mmol)溶于甲醇(50ml)中,搅拌,加入10%Pd/C(200mg),氢气置换三次,加氢搅拌过夜,冷却至室温,过滤,浓缩滤液,残留物经柱层析得到白色固体27-b(1.2g,收率:58%)。ESI-MS(m/z):206.2[M+H]+1HNMR(300MHz,DMSO-d6):δppm 9.98(s,1H),7.02(d,1H),6.71(dd,1H),6.64(d,1H),4.44(t,1H),3.37(q,2H),2.78(t,2H),2.49(t,2H),2.39(t,2H),1.63(m,2H).
步骤3:
将化合物27-b(600mg,2.93mmol)溶于二氯甲烷(10ml)中,搅拌,加入三乙胺(443.9mg,4.4mmol),冰浴下滴加甲磺酰氯(404mg,3.51mmol),加毕继续在室温反应2小时,反应液中加入二氯甲烷(100ml),分出有机相,依次用1N盐酸和饱和碳酸氢钠水溶液洗涤,无水硫酸钠干燥,浓缩,残留物经柱层析得白色固体(745mg,收率:90.5%)。ESI-MS(m/z):284.1[M+H]+1HNMR(300MHz,CDCl3):δppm 8.76(s,1H),7.08(d,1H),6.81(d,1H),6.63(s,1H),4.21(t,2H),3.01(s,3H),2.93(t,2H),2.70(t,2H),2.63(t,2H),2.04(m,2H).
制备例28 3-(2-氧代-1,2-二氢喹啉-7-基)丙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000051
参照制备例3的方法,以制备例27的产物为原料,得到灰色固体,收率37.5%。ESI-MS(m/z):282.2[M+H]+1HNMR(300MHz,DMSO-d6):δppm 11.67(s,1H),7.83(d,1H),7.56(d,1H),7.10(s,1H),7.04(d,1H),6.40(d,1H),4.19(t,2H),3.16(s,3H),2.71(t,2H),1.96(m,2H).
制备例29 7-(2-氯乙基)-4,5-二氢-1H-苯并氮杂卓-2(3H)-酮的制备
Figure PCTCN2015073854-appb-000052
步骤1:
冰浴下将氯乙酰氯(470ul,6.20mmol)滴加到无水氯化铝(2.48g,18.60mmol)的二硫化碳悬浮液中,搅拌10分钟,加入4,5-二氢-1H-苯并氮杂卓-2(3H)-酮29-a(500mg,3.10mmol),室温下搅拌15分钟,回流3小时,除去二硫化碳,反应液中加入冰块,析出大量淡黄色固体,加入4N盐酸(5ml),搅拌,过滤,滤饼用冰水洗3次,烘干得粗品780mg,用水(11ml)-甲醇(11ml)重结晶,得黄褐色针状晶体(640mg,收率:86.7%)。1HNMR(300MHz,CDCl3):δppm 8.31(s,1H),7.86(d,1H),7.84(dd,1H),7.07(d,1H),4.67(s,2H),2.88(t,2H),2.41(t,2H),2.29(m,2H).
步骤2:
将三乙基硅烷(840uL,5.25mmol)滴加到化合物29-b(500mg,2.10mmol)的三氟乙酸(5ml)溶液中,50℃下反应过夜。将反应液倒入冰水中,二氯甲烷萃取3次,合并有机相,干燥,浓缩,残留物在异丙醚中打浆,过滤,滤饼烘干得淡黄色固体(430mg,收率:91.4%)。
制备例30 2-(2-吲哚酮-6-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000053
步骤1:
反应瓶中加入对苯二乙酸30-a(9.2g,47.4mmol)和乙醇(50ml),搅拌下缓慢滴加浓硫酸(5ml,94.8mmol),升温至80℃回流过夜反应,减压浓缩除去乙醇,加入二氯甲烷溶解,用饱和碳酸氢钠水溶液调pH至8,分液,水相用二氯甲烷再萃取1次,合并有机相,干燥,浓缩,得到固体30-b(10.3g,收率:87%)。ESI-MS(m/z):251.2[M+H]+
步骤2:
反应瓶中加入化合物30-b(10g,40mmol)和浓硫酸(30ml),搅拌下在0℃滴加发烟硝酸(1.7ml,40.4mmol),加毕在室温继续反应1小时,将反应液倒入冰水中,析出固体,过滤,滤饼用水洗至中性,烘干,得到固体30-c(11g,收率:93%)。
步骤3:
反应瓶中依次加入化合物30-c(4.7g,15.9mmol),乙醇(25ml),10%Pd/C(850mg),40℃加氢反应过夜,40℃热过滤,滤饼用乙醇洗涤,浓缩滤液得粗品3.1g,用石油醚∶乙酸乙酯(6∶1,体积比)打浆,得到目标物30-d(2.4g,收率:71%)。EI-MS(m/z):219.1HNMR(300MHz,CDCl3):δppm 8.80(brs,1H),7.17(d,1H),6.93(d,1H),6.86(s,1H),4.17(q,2H),3.60(s,2H),3.52(s,2H),1.28(t,3H).
步骤4:
反应瓶中加入化合物30-d(2.86g,13.0mmol)和四氢呋喃(200ml),在0℃分批加入氢化铝锂(2.5g,65.3mmol),30分钟加完,在0℃继续反应30分钟,往反应液中依次缓慢滴加水(1.24ml)、15%氢氧化钠水溶液(1.24ml)、水(3.72ml),过滤,滤饼用甲醇洗涤,浓缩滤液,加入二氯甲烷打浆,抽滤,得粗品1.28g,用石油醚∶二氯甲烷打浆,抽滤,烘干得白色固体30-e(1.17g,收率:50%)。ESI-MS(m/z):178.1[M+H]+1HNMR(300MHz,DMSO-d6):δppm 10.31(brs,1H),7.07(d,1H),6.76(d,1H),6.68(s,1H),4.64(t,1H),3.57(q,2H),3.39(s,2H),2.67(t,2H).
步骤5:
取化合物30-e(200mg,1.1mmol),吡啶(0.24,3.3mmol),悬浮于二氯甲烷中,冰浴下缓慢滴入甲磺酰氯(0.12ml,1.21mmol),加毕,移去冰浴,室温下搅拌反应4小时,将反应液倒入水中,依次用1N盐酸和饱和盐水洗涤,干燥有机相,滤除干燥剂,浓缩得目标物(220mg,收率:76%)。ESI-MS(m/z):256.0[M+H]+
制备例31 4-(苯并噻吩-4-基)哌啶三氟乙酸盐的制备
Figure PCTCN2015073854-appb-000054
步骤1:
将二异丙胺(5.14g,50.8mmol)溶于无水四氢呋喃(50ml)中,氮气保护,搅拌,在0℃滴加正丁基锂的正己烷溶液(20.3ml,50.8mml),滴完反应30分钟,反应液冷却至-78℃,滴加3-溴氟苯31-a(10.0g,42.4mml)的无水四氢呋喃(100ml)溶液,滴完后继续反应30分钟,滴加甲酸甲酯(7.63g,127.2mml),滴完后反应30分钟,升温至室温,将反应液倒入1N盐 酸(100ml),用乙酸乙酯(100ml×3)萃取,合并有机相,无水硫酸钠干燥,滤除干燥剂,浓缩,残留物经柱层析得白色固体31-b(8.0g,收率:93%)。
步骤2:
将化合物31-b(4.1g,20.4mmol),硼酸酯(6.3g,20.4mmol),[1,1′-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(Pd(dppf)Cl2.CH2Cl2,1.7g,2.04mmol)和碳酸钾(7.04g,51.0mmol)分散在无水N,N-二甲基甲酰胺(80ml)中,搅拌,氮气置换3次,80℃反应过夜,冷却至室温,倒入水(200ml)中,用乙酸乙酯(100ml×3)萃取,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,滤除干燥剂,浓缩,残留物经柱层析得到无色油状物31-c(3.5g,收率:55.5%)。
1HNMR(300MHz,CDCl3):δppm 10.26(s,1H),7.50(m,1H),7.09(t,1H),7.03(d,1H),5.55(s,1H),4.05(m,2H),3.67(m,2H),2.33(s,2H),1.49(s,9H).
步骤3:
将化合物31-c(3.5g,11.5mmol)溶于乙酸乙酯(50ml)中,搅拌,加入10%Pd/C(1.0g),氢气置换3次,室温下反应5小时,过滤,浓缩滤液得无色油状物31-d(3.5g,100%)。ESI-MS(m/z):310.1[M+H]+1HNMR(300MHz,DMSO-d6):δppm 7.26(dd,1H),7.04(d,1H),6.93(t,1H),4.80(s,2H),4.24(d,2H),3.08(t,1H),2.82(t,2H),1.78(d,2H),1.72-1.53(m,4H),1.47(s,9H).
步骤4:
将化合物31-d(3.5g,11.5mmol)溶于二氯甲烷(50ml)中,搅拌,室温下加入氯铬酸吡啶(PCC,3.0g,13.8mmol),反应1小时后,过滤,浓缩滤液,残留物经柱层析得白色固体31-e(3.0g,收率:85%)。ESI-MS(m/z):308.1[M+H]+1HNMR(300MHz,CDCl3):δppm10.54(s,1H),7.51(dd,1H),7.16(d,1H),7.02(t,1H),4.23(d,2H),3.78(t,1H),2.86(t,2H),1.78(d,2H),1.57(m,2H),1.47(s,9H).
步骤5:
将化合物31-e(3.0g,9.4mmol)溶于乙腈(50ml)中,搅拌下加入碳酸钾(1.95g,14.1mmol)和巯基乙酸乙酯(0.85ml,9.4mmol),80℃反应过夜,过滤,浓缩滤液,残留物经柱层析得白色固体31-f(3.3g,收率:90.1%)。ESI-MS(m/z):390.0[M+H]+1HNMR(300MHz,CDCl3):δppm 8.20(s,1H),7.71(d,1H),7.41(t,1H),7.22(d,1H),4.41(q,2H),4.30(d,2H),3.22(m,1H),2.90(t,2H),1.91(d,2H),1.75(m,2H),1.49(s,9H),1.42(t,3H).
步骤6:
将化合物31-f(3.24g,8.3mmol)溶于四氢呋喃(60ml)中,搅拌下加入一水合氢氧化锂(698mg,16.6mmol)和甲醇(10ml),室温反应5小时,在反应液中加入1N盐酸调节pH至4~5,析出固体,过滤,滤饼真空干燥,得白色固体31-g(2.8g,收率:96.5%)。
步骤7:
将化合物31-g(2.44g,6.76mmol)分散在喹啉(30ml)中,搅拌,加入氧化亚铜(966mg,0.676mml),140℃反应5小时,降温至室温,用1N盐酸调pH至4~5,用乙酸乙酯(100ml×3)萃取,合并有机相,无水硫酸钠干燥,滤除干燥剂,浓缩,残留物经柱层析得白色固体31-h(1.0g,收率:46.7%)。ESI-MS(m/z):318.2[M+H]+1HNMR(300MHz,CDCl3):δppm7.76(d,1H),7.47(s,2H),7.32(t,1H),7.19(d,1H),4.31(s,2H),3.20(t,1H),2.89(t,2H),1.93(d,2H),1.77(m,2H),1.49(s,9H).
步骤8:
取化合物31-h(400mg,1.26mmol)溶于二氯甲烷(1ml),加入三氟乙酸(1ml),室温下搅拌过夜,将反应液减压浓缩干,残余油状物用甲基叔丁基醚/甲醇(15ml/1ml)混合溶剂打浆,过滤,烘干得黄色固体(219mg,收率:80%)。ESI-MS(m/z):218.0[M+H]+
制备例32 2-(6-氯-2-氧代-1,2-二氢喹啉-7-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000055
取制备例16产物2-(6-氯-2-氧代-1,2,3,4-四氢喹啉-7-基)乙基甲磺酸酯(330mg,1.09mmol),加入二氧六环(5ml)和2,3-二氯-5,6-二氰对苯醌(DDQ,740mg,3.26mmol),加热回流过夜。反应完毕,浓缩除去二氧六环,加入二氯甲烷(25ml),依次用饱和碳酸氢钠水溶液,硫代硫酸钠水溶液和饱和食盐水洗,干燥有机相,滤除干燥剂,浓缩,残留物经柱层析得目标物(180mg,收率:55%)。ESI-MS(m/z):301.9[M+H]+
制备例33 6-(2-氯乙基)-3-甲基-3,4-二氢喹唑啉-2(1H)-酮的制备
Figure PCTCN2015073854-appb-000056
步骤1:
冰浴下将氯乙酰氯(460uL,6.16mmol)滴加到无水氯化铝(1.03g,7.7mmol)的1,2-二氯乙烷悬浮液中,搅拌10分钟,加入3-甲基-3,4-二氢喹唑啉-2(1H)-酮33-a(500mg,3.08mmol),室温下继续搅拌15分钟,升温至48℃反应3小时,停止加热,待反应液冷至室温后,将反应液倾入冰中,析出淡黄色固体,过滤,滤饼用冰水洗3次,烘干,得淡黄色固体33-b(710mg,收率:97%)。1HNMR(300MHz,DMSO-d6):δppm 9.72(s,1H),7.82-7.73(m,2H),6.84(d,1H),5.06(s,2H),4.47(s,2H),2.87(s,3H).
步骤2:
将三乙基硅烷(587ul,3.68mmol)滴加到化合物33-b(350mg,1.47mmol)的三氟乙酸(4ml)溶液中,35℃下反应2.5小时,将反应液倾入冰水中,析出固体,过滤,滤饼用冰水洗3 次,烘干,得淡黄色固体(320mg,收率:97%)。
制备例34 2-(1H-吲哚-3-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000057
取化合物34-a(200mg,1.24mmol)溶于二氯甲烷(5ml)中,加入三乙胺(0.206ml,1.49mmol),冰浴条件下缓慢滴入甲磺酰氯(0.105ml,1.36mmol),滴完室温下搅拌反应3小时,加入水,用二氯甲烷萃取,有机相用饱和食盐水洗,干燥,滤除干燥剂,浓缩得目标物(260mg,收率:87%)。ESI-MS(m/z):240.0[M+H]+
制备例35 6-(2-氯乙基)-3,4-二氢喹啉-2(1H)-酮的制备
Figure PCTCN2015073854-appb-000058
步骤1:
冰浴下将氯乙酰氯(640ul,8.49mmol)滴加到无水氯化铝(2.72g,20.38mmol)的二硫化碳(10ml)悬浮液中,搅拌10分钟,加入3,4-二氢喹啉-2(1H)-酮35-a(500mg,3.40mmol),室温下先搅拌15分钟,后加热回流9小时,将二硫化碳倒掉,加入冰块,析出大量淡黄色固体,加入4N盐酸(5ml),搅拌,过滤,滤饼用冰水洗3次,烘干得粗品730mg,加入乙酸乙酯打浆,过滤,烘干得灰白色固体(710mg,收率:93%)。1HNMR(300MHz,DMSO-d6):δppm 10.46(s,1H),7.81(d,1H),7.78(dd,1H),6.92(d,1H),5.07(s,2H),2.93(t,2H),2.48(t,2H).
步骤2:
将三乙基硅烷(900uL,5.58mmol)滴加到化合物35-b(500mg,2.32mmol)的三氟乙酸(5ml)溶液中,30℃下反应过夜。将反应液倒入冰水中,用二氯甲烷(15ml×3)萃取,合并有机相,干燥,滤除干燥剂,浓缩,残留物中加入石油醚打浆,过滤,滤饼烘干得黄褐色固体(417mg,收率:89%)。
制备例36 2-(2-吲哚酮-4-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000059
将4-(2-羟基乙基)二氢吲哚-2-酮36-a(500mg,2.822mmol)和二氯甲烷(15ml)混合,加入吡啶(335mg,4.233mmol),冰浴冷却至0℃,滴加甲磺酰氯(356mg,3.104mmol),加完后撤去冰浴,室温搅拌下反应过夜。反应液中加入二氯甲烷(15ml),依次用1N盐酸,水和饱和食盐水各洗1次,用无水硫酸钠干燥有机相,滤除干燥剂,滤液减压浓缩至干,残留物经柱层析得黄色固体(460mg,收率:64%)。ESI-MS(m/z):256.0[M+H]+
制备例37 2-(1-(叔丁氧基羰基)-4,5-二乙基-6-氧代-1,6-二氢嘧啶-2-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000060
步骤1:
取化合物37-a(2.0g,16.3mmol),2-乙基-3-氧代戊酸乙酯37-b(6.0g,19.5mmol),碳酸钾(9.4g,48.9mmol)与乙醇(20ml)混合,加热回流15小时,冷却,过滤固体,柱层析得目标物(198mg)。ESI-MS(m/z):197.3[M+H]+1H-NMR(300Hz,DMSO-d6):δppm 12.09(brs,1H),4.75(brs,1H),3.72(t,2H),2.61(t,2H),2.45(q,2H),2.37(q,2H),1.11(t,3H),0.98(t,3H).
步骤2:
将化合物37-c(250mg,1.32mmol)悬于四氢呋喃(15ml)中,加入二碳酸二叔丁酯(317mg,1.45mmol),反应液在25~30℃反应过夜,反应液减压浓缩至干,残余物经柱层析得浅黄色油状物(494mg)。
步骤3:
取化合物37-d(494mg,1.667mmol)溶于二氯甲烷(20ml)中,加入三乙胺(253mg,2.5mmol),冷至0~5℃,加入甲磺酰氯(210mg,1.834mmol),加完后室温反应1小时。反应液水洗,饱和食盐水洗,无水硫酸钠干燥,过滤,滤液浓缩至干,残余物经柱层析得目标物(520mg)。ESI-MS(m/z):374.3[M+H]+
制备例38 2-(1-(叔丁氧基羰基)-6-氧代-1,6-二氢嘧啶-2-基)乙基甲磺酸酯的制备
Figure PCTCN2015073854-appb-000061
参照制备例37的方法,用丙炔酸乙酯代替2-乙基-3-氧代戊酸乙酯制备目标物。
ESI-MS(m/z):319.1[M+H]+
实施例
实施例1 6-氯-5-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮
将6-氯-5-(2-氯乙基)二氢吲哚-2-酮(120mg,0.52mmol),制备例12产物(115mg,0.52mmol),碳酸钾(215mg,1.56mmol),碘化钾(86mg,0.52mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,柱层析得白色固体(120mg,收率:56%)。1HNMR(300MHz,DMSO-d6):δppm 10.42(brs,1H),7.21(s,1H),7.07(t,1H),6.90(d,1H),6.80(s,1H),6.69(d,1H),3.45(s,2H),3.30(t,2H),3.14(t,2H),2.74-2.95(m,6H),2.59(brs,4H),2.50(t,2H).ESI-MS(m/z):414.2[M+H]+
实施例2 3-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将3-(2-氯乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮10-a(120mg,0.49mmol),制备例12产物(109mg,0.49mmol),碳酸钾(202mg,1.47mmol),碘化钾(81mg,0.49mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,柱层析得淡黄色固体(120mg,收率:56%)。1HNMR(300MHz,DMSO-d6):δppm 7.07(t,1H),6.89(d,1H),6.69(d,1H),5.68(d,1H),4.44(m,1H),3.89(m,1H),3.66(m,1H),3.30(t,2H),3.14(t,2H),2.87(brt,4H),2.53-2.67(m,6H),2.39(t,2H),2.26(s,3H),1.73-2.06(m,4H).ESI-MS(m/z):427.2[M+H]+
实施例3 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将3-(2-氯乙基)-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮(120mg,0.53mmol),制备例1产物(116mg,0.53mmol),碳酸钾(219mg,1.59mmol),碘化钾(88mg,0.53mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,柱层析得白色固体(120mg,收率:55%)。
1HNMR(300MHz,DMSO-d6):δppm 7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),6.89(d,1H),3.78(t,2H),3.07(brs,4H),2.75(t,2H),2.68(brs,4H),2.63(t,2H),2.42(t,2H),2.22(s,3H),1.85(m,2H),1.76(m,2H).ESI-MS(m/z):409.1[M+H]+
实施例4 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将3-(2-氯乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮10-a(1g,4.13mmol),制备例1产物(1.05g,4.13mmol),碳酸钾(1.7g,12.3mmol),碘化钾(0.68g,4.13mmol)以及乙腈(10ml)加入烧瓶中,85℃回流过夜,浓缩反应液,柱层析得淡黄色固体(1g,收率:58%)。1HNMR(300MHz,DMSO-d6):δppm 7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),6.90(d,1H),5.69(d,1H),4.44(q,1H),3.91(m,1H),3.68(m,1H),3.08(brs,4H),2.69(brs,4H),2.66(t,2H),2.45(t,2H),2.28(s,3H),1.74-2.04(m,4H).ESI-MS(m/z): 425.3[M+H]+
用HPLC方法进行手性拆分,分别得到单一异构体4a和4b,保留时间分别为13.2分钟和16.3分钟。色谱柱型号:AY-H4.6×250mm;流动相:乙醇∶正己烷=30∶70(v/v);流速:1.0ml/min;检测波长:277nm。
化合物4a:(+)-3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮,比旋光度:+8.82°(C=0.17,CH2Cl2)。
化合物4b:(-)-3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮,比旋光度:-7.98°(C=0.17,CH2Cl2)。
实施例5 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-7,8-二氢-6H-吡啶并[1,2-a]嘧啶-4,9-二酮
将实施例4产物(120mg,0.28mmol)溶于二氯甲烷(5ml)中,室温下分批加入Dess-Martin试剂(359mg,0.84mmol),室温搅拌5h,加入饱和碳酸氢钠溶液(2ml)和硫代硫酸钠溶液(1ml),搅拌10分钟,用二氯甲烷萃取,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析得淡黄色固体(70mg,收率:58%)。1HNMR(300MHz,CDCl3):δppm 7.54(d,1H),7.39(m,2H),7.27(t,1H),6.89(d,1H),4.20(t,2H),3.20(brs,4H),2.92-2.74(m,8H),2.62(t,2H),2.48(s,3H),2.33(m,2H).ESI-MS(m/z):423.2[M+H]+
实施例6 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2,9-二甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮盐酸盐
将实施例5产物(120mg,0.28mmol)溶于干燥的四氢呋喃(3ml),在冰浴条件下,将甲基溴化镁(2ml,1.99mmol,1M in THF)滴加到上述溶液中,室温搅拌3小时,加入饱和氯化铵淬灭,浓缩除去四氢呋喃,用二氯甲烷萃取,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析得油状物,加入氯化氢-乙醇溶液成盐,乙腈打浆得淡黄色固体(40mg,收率:32%)。1HNMR(300MHz,MeOH-d6):δppm 7.65(d,1H),7.58(d,1H),7.48(d,1H),7.31(t,1H),7.02(d,1H),4.37(d,1H),3.83(m,1H),3.66(m,3H),3.41(m,2H),3.19(m,4H),2.93-2.49(m,4H),2.22-1.60(m,6H),1.52(s,3H).
实施例7 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将制备例17产物(125mg,0.51mmol),制备例1产物(112mg,0.51mmol),碳酸钾(213mg,1.54mmol),碘化钾(85mg,0.51mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,柱层析得淡黄色固体(150mg,收率:68%)。1HNMR(300MHz,CDCl3):δppm7.54(d,1H),7.36-7.43(m,2H),7.27(t,1H),6.90(d,1H),5.25-5.46(dt,1H),4.18(dt,1H),3.75(m,1H),3.21(brt,4H),2.82(m,6H),2.60(t,2H),2.30-2.46(m,1H),2.39(s,3H),1.96-2.25(m,3H).ESI-MS(m/z):427.2[M+H]+
实施例8 5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮
将制备例15产物(79mg,0.40mmol),制备例1产物(104mg,0.40mmol),碳酸钠(108mg,0.81mmol),碘化钠(60mg,0.40mmol)以及水(3ml)加入烧瓶中,100℃回流过夜,加入水,用二氯甲烷萃取,有机相用无水硫酸钠干燥,浓缩,柱层析,乙腈打浆得白色粉末(90mg,收率:58%)。1HNMR(300MHz,DMSO-d6):δppm 10.28(brs,1H),7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),7.10(s,1H),7.04(d,1H),6.90(d,1H),6.72(d,1H),3.44(s,2H),3.07(brs,4H),2.72(t,2H),2.70(brs,4H),2.57(t,2H).ESI-MS(m/z):378.3[M+H]+
实施例9 7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮
将制备例7产物(440mg,1.64mmol)和制备例1产物(418mg,1.64mmol),碳酸钾(682mg,4.94mmol),碘化钾(272mg,1.64mmol)溶于乙腈(10ml)中,加热回流过夜。将反应液倒入冰水中,加入乙酸乙酯萃取2次;合并有机相,水洗2次,饱和食盐水洗1次,无水硫酸钠干燥,过滤,滤液浓缩至于得粗品,经柱层析得类白色固体(200mg,收率:31%)。
1HNMR(400MHz,DMSO-d6):δppm 11.70(brs,1H),7.86(d,1H),7.70(d,1H),7.61(d,1H),7.57(d,1H),7.40(d,1H),7.27(t,1H),7.17(s,1H),7.09(dd,1H),6.90(d,1H),6.42(dd,1H),3.08(brs,4H),2.85(t,2H),2.71(brs,4H),2.64(t,2H).ESI-MS(m/z):390.2[M+H]+
实施例10 7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)-3,4-二氢喹啉-2(1H)-酮
将制备例2产物(250mg,0.80mmol),制备例1产物(140mg,0.80mmol),碳酸钾(221mg,1.6mmol),碘化钾(132mg,0.80mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,残留物经柱层析得白色粉末(270mg,收率:77%)。1HNMR(300MHz,DMSO-d6):δppm 9.98(brs,1H),7.68(d,1H),7.60(d,1H),7.38(d,1H),7.26(t,1H),7.04(d,1H),6.88(d,1H),6.74(d,1H),6.67(s,1H),3.05(brs,4H),2.80(t,2H),2.58(brs,4H),2.49(t,2H),2.40(t,2H),2.34(t,2H),1.51(m,4H),1.30(m,2H).ESI-MS(m/z):434.3[M+H]+
实施例11 7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮
将制备例3产物(180mg,0.58mmol),制备例1产物(148g,0.58mmol),无水碳酸钾(241mg,1.74mmol),碘化钾(96mg,0.58mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,残留物经柱层析得白色粉末(200mg,收率:79%)。1HNMR(300MHz,DMSO-d6):δppm 11.65(s,1H),7.84(d,1H),7.68(d,1H),7.60(d,1H),7.55(d,1H),7.38(d,1H),7.26(t,1H),7.11(s,1H),7.03(dd,1H),6.87(d,1H),6.41(d,1H),3.03(brs,4H),2.65(t,2H),2.57(brs,4H),2.34(t,2H),1.62(m,2H),1.49(m,2H),1.33(m,2H).ESI-MS(m/z):432.3[M+H]+
实施例12 7-(5-(4-(2-氯苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮
将制备例3产物(108mg,0.35mmol),制备例9产物(90mg,0.35mmol),碳酸钾(147mg,1.07mmol),碘化钾(58mg,0.35mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,残留物经柱层析得白色粉末(75mg,收率:45%)。1HNMR(500MHz,DMSO-d6):δppm 11.66(brs,1H),7.84(d,1H),7.55(m,2H),7.36(s,1H),7.29(t,1H),7.10(s,1H),7.03(d,1H),6.91(d,1H),6.41(d,1H),3.00(brs,4H),2.64(t,2H),2.56(brs,4H),2.33(t,2H),1.61(m,2H),1.49(m,2H),1.32(m,2H).ESI-MS(m/z):466.2[M+H]+
实施例13 7-(5-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮盐酸盐
将制备例3产物(104mg,0.33mmol),制备例8产物(80mg,0.33mmol),碳酸钾(140mg,1.01mmol),碘化钾(56mg,0.33mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,残留物经柱层析得油状物,加入氯化氢-乙醇溶液成盐,得白色固体(95mg,收率:62%)。1HNMR(400MHz,DMSO-d6):δppm 11.70(s,1H),10.67(brs,1H),7.85(d,1H),7.61(d,1H),7.57(d,1H),7.31(t,1H),7.15(d,1H),7.12(s,1H),7.05(d,1H),7.01(d,1H),6.42(d,1H),3.55(d,1H),3.44(d,1H),3.05-3.29(m,6H),2.67(t,2H),1.77(m,2H),1.64(m,2H),1.35(m,2H).ESI-MS(m/z):450.2[M+H]+
实施例14 7-(5-(4-(苯并噻吩-4-基)-5,6-二氢吡啶-1(2H)-基)戊基)喹啉-2(1H)-酮
将制备例3产物(114mg,0.37mmol),制备例14产物(80mg,0.37mmol),碳酸钾(154mg,1.11mmol),碘化钾(61mg,0.37mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜,浓缩反应液,残留物经柱层析得白色粉末(66mg,收率:60%)。1HNMR(500MHz,DMSO-d6):δppm11.65(brs,1H),7.88(d,1H),7.84(d,1H),7.73(d,1H),7.55(d,1H),7.48(d,1H),7.32(t,1H),7.21(d,1H),7.11(s,1H),7.04(d,1H),6.41(d,1H),5.86(s,1H),3.09(s,2H),2.65(m,4H),2.49(t,2H),2.40(t,2H),1.63(m,2H),1.53(m,2H),1.35(m,2H).ESI-MS(m/z):429.3[M+H]+
实施例15 5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯二氢吲哚-2-酮盐酸盐
将6-氯-5-(2-氯乙基)二氢吲哚-2-酮(332mg,1.45mmol),制备例1产物(370mg,1.45mmol),碳酸钠(461mg,4.35mmol),碘化钠(216mg,1.45mmol)以及水(5ml)加入烧瓶中,回流反应24小时。反应液冷至室温,加入水,用二氯甲烷萃取,无水硫酸钠干燥有机相,滤除干燥剂,浓缩,残留物经柱层析得油状物,加入氯化氢-乙醇溶液成盐,异丙醇打浆,过滤得淡黄色固体(308mg,收率:51%)。1HNMR(400MHz,DMSO-d6):δppm 11.04(brs,1H),10.55(s,1H),7.77(d,1H),7.71(d,1H),7.50(d,1H),7.32(t,1H),7.29(s,1H),6.98(d,1H),6.88(s,1H),3.71(d,2H),3.57(d,2H),3.50(s,2H),3.38(m,4H),3.21(m,4H).ESI-MS(m/z):412.2[M+H]+
实施例16 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7-二氢吡啶并[1,2-a]嘧啶-4-酮盐酸盐
将制备例10产物(100mg,0.44mmol),制备例1产物(113mg,0.44mmol),碳酸钾(184mg,1.33mmol),碘化钾(74mg,0.44mmol)以及乙腈(5ml)加入烧瓶中,85℃回流过夜。反应液冷至室温,加入水,用二氯甲烷萃取,无水硫酸钠干燥有机相,滤除干燥剂,浓缩,残留物经柱层析得油状物,加入氯化氢-乙醇溶液成盐,异丙醇打浆,过滤得白色粉末(60mg,收率:33%)。1HNMR(300MHz,DMSO-d6):δppm 11.34(brs,1H),7.77(d,1H),7.70(d,1H), 7.50(d,1H),7.32(t,1H),7.00(m,2H),6.60(d,1H),4.04(t,2H),3.71(d,2H),3.56(d,2H),3.14-3.47(m,6H),3.02(m,2H),2.61(m,2H),2.42(s,3H).ESI-MS(m/z):407.1[M+H]+
实施例17 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-(苄氧基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮盐酸盐
将9-(苄氧基)-3-(2-氯乙基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮(200mg,0.60mmol),制备例1产物(155mg,0.60mmol),碳酸钾(673mg,4.87mmol),碘化钾(101mg,0.60mmol)以及乙腈(5ml)加入烧瓶中,85℃回流24小时。反应液冷至室温,加入水,用二氯甲烷萃取,无水硫酸钠干燥,滤除干燥剂,浓缩,残留物经柱层析得油状物,加入氯化氢-乙醇溶液成盐,过滤得白色粉末(200mg,收率:64%)。1HNMR(300MHz,CDCl3):δppm 8.61(dd,1H),7.54(d,1H),7.24-7.49(m,8H),6.86-6.93(m,3H),5.39(s,2H),3.23(brs,4H),3.01(t,2H),2.87(brs,4H),2.69(t,2H),2.64(s,3H).ESI-MS(m/z):511.3[M+H]+
实施例18 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮
将实施例17产物(180mg,0.35mmol)加入浓盐酸(5ml)中,80℃加热2小时。将反应液冷却后滴加入异丙醇(5ml)中,析出固体,过滤得到淡黄色固体(130mg,收率87%)。
1HNMR(300MHz,DMSO-d6):δppm 11.64(brs,1H),8.63(d,1H),7.88(m,1H),7.77(d,1H),7.69(d,1H),7.57(td,1H),7.50(d,1H),7.31(t,1H),6.96(d,1H),3.75(d,1H),3.56(d,1H),3.50-3.12(m,8H),2.70(s,3H).ESI-MS(m/z):421.2[M+H]+
实施例19 7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮
将制备例6产物(100mg,0.37mmol)和制备例1产物(95mg,0.37mmol),碳酸钾(153mg,1.11mmol),碘化钾(61mg,0.37mmol)溶于乙腈(10ml)中,加热回流过夜。浓缩反应液,柱层析得白色固体(70mg,收率48%)。1HNMR(400MHz,DMSO-d6):δppm 10.02(brs,1H),7.70(d,1H),7.62(d,1H),7.41(d,1H),7.28(t,1H),7.07(d,1H),6.91(d,1H),6.80(dd,1H),6.74(d,1H),3.08(brs,4H),2.82(t,2H),2.69(m,6H),2.58(t,2H),2.43(t,2H).ESI-MS(m/z):392.1[M+H]+
实施例20 9-羟基-2-甲基-3-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将3-(2-氯乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮10-a(73mg,0.30mmol)和制备例13产物(77mg,0.30mmol),碳酸钾(145mg,1.05mmol),碘化钾(50mg,0.30mmol)溶于乙腈(7.5ml)中,加热回流过夜。浓缩反应液,残留物经柱层析得类白色固体(53mg,收率:41%)。1HNMR(300MHz,DMSO-d6):δppm 8.40(s,1H),7.63(s,2H),5.71(d,1H),4.44(m,1H),3.87(brs,5H),3.66(m,1H),2.53-2.70(m,6H),2.39(t,2H),2.26(s,3H),1.73-2.06(m,4H).ESI-MS(m/z):427.2[M+H]+
实施例21 2-甲基-3-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并 [1,2-a]嘧啶-4-酮
将3-(2-氯乙基)-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮(82mg,0.36mmol)和制备例13产物(93mg,0.36mmol),碳酸钾(173mg,1.26mmol),碘化钾(60mg,0.36mmol)溶于乙腈(7.5ml)中,加热回流过夜。浓缩反应液,残留物经柱层析得类白色固体(80mg,收率:54%)。1HNMR(300MHz,DMSO-d6):δpmm 8.40(s,1H),7.62(s,2H),3.87(brs,4H),3.77(t,2H),2.74(t,2H),2.58(brs,6H),2.38(t,2H),1.84(m,2H),1.74(m,2H).ESI-MS(m/z):411.1[M+H]+
实施例22 7-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮
将制备例4产物(154mg,0.52mmol),制备例1产物(133mg,0.52mmol),碳酸钾(215mg,1.56mmol)和碘化钾(86mg,0.52mmol)溶于乙腈(7.5ml)中,加热回流过夜。浓缩反应液,残留物经柱层析得黄色固体(110mg,收率:50%)。1HNMR(300MHz,DMSO-d6):δppm 10.02(brs,1H),7.69(d,1H),7.61(d,1H),7.39(d,1H),7.27(t,1H),7.05(d,1H),6.88(d,1H),6.74(d,1H),6.68(s,1H),3.05(brs,4H),2.81(t,2H),2.58(brs,4H),2.50(t,2H),2.41(t,4H),1.39-1.64(m,4H).ESI-MS(m/z):420.2[M+H]+
实施例23 7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯-3,4-二氢喹啉-2(1H)-酮
将制备例16产物(100mg,0.33mmol),制备例1产物(84mg,0.33mmol),碳酸钾(114mg,0.82mmol),碘化钾(55mg,0.33mmol)溶于乙腈(10ml)中,加热回流过夜。浓缩,残留物经柱层析得类白色固体(45mg,收率:32%)。1HNMR(300MHz,DMSO-d6):δppm 10.17(brs,1H),7.69(d,1H),7.62(d,1H),7.41(d,1H),7.20-7.33(m,2H),6.90(d,1H),6.84(s,1H),3.08(brs,4H),2.83(m,4H),2.70(brs,4H),2.56(t,2H),2.42(t,2H).ESI-MS(m/z):426.3[M+H]+
实施例24 6-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)-2-甲基喹唑啉-4(3H)-酮
取制备例11产物(77mg,0.29mmol),制备例1产物(74mg,0.29mmol),碳酸钾(120mg,0.87mmol),碘化钾(48mg,0.29mmol),加入乙腈(3ml)作溶剂,回流50小时,浓缩反应液,残留物经柱层析得淡黄色固体(35mg,收率:27%)。1H-NMR(300Hz,CDCl3):δppm10.84(brs,1H),8.05(s,1H),7.51-7.63(m,3H),7.38(m,2H),7.27(t,1H),6.91(d,1H),3.27(brs,4H),2.83(brs,4H),2.77(t,2H),2.57(t,2H),2.54(s,3H),1.72(m,4H),1.41(m,2H).ESI-MS(m/z):447.3[M+H]+
实施例25 7-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮
将制备例12产物(85mg,0.38mmol),制备例7产物(103mg,0.38mmol),碳酸钾(157mg,1.14mmol),碘化钾(63mg,0.38mmol)和乙腈(5ml)加入烧瓶中,85℃回流过夜。浓缩反应液,残留物经柱层析得粗品,再用乙腈打浆得白色固体(70mg,收率:46%)。1H-NMR(300Hz,DMSO-d6):δppm 11.67(brs,1H),7.85(d,1H),7.56(d,1H),7.16(s,1H),7.07(m,2H),6.90(d,1H),6.69(d,1H),6.42(d,1H),3.30(t,2H),3.14(t,2H),2.88(brs,4H),2.83(t,2H),2.61(m,6H).ESI-MS(m/z):392.2[M+H]+
实施例26 1-(苯并噻吩-4-基)-4-(2-(4-甲基噻唑-5-基)乙基)哌嗪盐酸盐
将制备例18产物(150mg,0.757mmol)、制备例1产物(154mg,0.529mmol),碳酸钾(836mg,6.058mmol),碘化钾(87mg,0.529mmol)加入到乙腈(6ml)中,回流过夜。浓缩反应液,加水和二氯甲烷,分出有机层,水层用二氯甲烷再萃取2次,干燥有机相,滤除干燥剂,浓缩,残留物经柱层析得油状物140mg,在氯化氢-乙醇溶液中成盐,过滤,滤饼烘干得白色固体(78mg,收率:39%)。1H-NMR(300Hz,DMSO-d6):δppm 8.79(s,1H),7.70(d,1H),7.62(d,1H),7.40(d,1H),7.28(t,1H),6.91(d,1H),3.09(brs,4H),2.95(t,2H),2.70(brs,4H),2.60(t,2H),2.33(s,3H).ESI-MS(m/z):344.2[M+H]+
实施例27 5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二氢苯并咪唑-2-酮
将制备例19产物(150mg,0.76mmol),制备例1产物(178mg,0.61mmol),碳酸钾(42mg,3.04mmol),碘化钾(126mg,0.76mmol)加入到N,N-二甲基甲酰胺(6ml)中,105℃加热过夜。将反应液倒入水中,用二氯甲烷萃取,干燥有机相,滤除干燥剂,浓缩,残留物经柱层析得淡黄色固体(59mg,收率:20%)。1H-NMR(300Hz,DMSO-d6):δppm 10.53(s,1H),10.49(s,1H),7.70(d,1H),7.62(d,1H),7.40(d,1H),7.28(t,1H),6.90(d,1H),6.81(m,3H),3.08(brs,4H),2.75(t,2H),2.69(brs,4H),2.58(t,2H).ESI-MS(m/z):379.2[M+H]+
实施例28 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9,9-二氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮盐酸盐
将实施例5产物(120mg,0.28mmol)溶于二氯甲烷(3ml)中,在冰浴条件下将含二乙胺基三氟化硫(0.079ml,0.59mmol)的二氯甲烷溶液(3ml)滴加到上述溶液中,室温搅拌过夜。加入饱和碳酸氢钠溶液,用二氯甲烷萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,滤除干燥剂,浓缩,残留物经柱层析得粗品,在氯化氢-乙醇溶液中成盐,得淡黄色固体(50mg,收率:39%)。1HNMR(300MHz,DMSO-d6):δppm 10.91(brs,1H),7.77(d,1H),7.71(d,1H),7.51(d,1H),7.32(t,1H),6.98(d,1H),3.88(t,2H),3.73(d,2H),3.58(d,2H),3.40(m,2H),3.23(m,4H),3.02(m,2H),2.43(m,2H),2.40(s,3H),2.07(m,2H).ESI-MS(m/z):445.3[M+H]+
实施例29 6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2(3H)-苯并噻唑酮盐酸盐
将制备例1产物(228mg,0.784mmol),制备例20产物(186mg,0.869mmol),碳酸钠(333mg,3.142mmol),碘化钠(5mg)加入到4-甲基-2-戊酮(MIBK,6ml)中,氮气保护,回流24h。浓缩反应液,残留物经柱层析得黄色油状物,在氯化氢-乙醇溶液中成盐,在丙酮∶异丙醚(1∶1,体积比)混合液中打浆,过滤,滤饼烘干得淡黄色固体(58mg,收率:17%)。1H-NMR(400Hz,DMSO-d6):δppm 11.98(s,1H),11.05(brs,1H),7.78(d,1H),7.72(d,1H),7.53(s,1H),7.50(d,1H),7.33(t,1H),7.23(dd,1H),7.12(d,1H),6.99(d,1H),3.68(d,2H),3.57(d,2H),3.39(m,4H),3.24(t,2H),3.14(m,2H).ESI-MS(m/z):396.2[M+H]+
实施例30 6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-1,4-苯并噁嗪-3(4H)-酮盐酸盐
将制备例1产物(224mg,0.77mmol),制备例21产物(196mg,0.925mmol),碳酸钠(328mg,3.094mmol),碘化钠(5mg)加入到N-甲基吡咯烷酮(NMP,6ml)中,氮气保护,120℃加热11小时,反应液倒入冰水中搅拌,大量红褐色固体析出,过滤,滤饼用冰水洗三次,烘干,用二氯甲烷/甲醇溶解,硅胶拌样柱层析,得粗品112mg,在氯化氢-乙醇溶液中成盐,丙酮打浆,过滤,滤饼烘干得淡黄色固体(105mg,收率:31.7%)。1H-NMR(300Hz,DMSO-d6):δppm 10.67(s,1H),7.69(d,1H),7.62(d,1H),7.40(d,1H),7.28(t,1H),6.90(d,1H),6.88-6.76(m,3H),4.52(s,2H),3.08(brt,4H),2.69(m,6H),2.56(t,2H).ESI-MS(m/z):394.3[M+H]+
实施例31 6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-1,4-苯并噻嗪-3(4H)-酮
将制备例1产物(204mg,0.701mmol),制备例22产物(200mg,0.881mmol),碳酸钠(300mg,2.83mmol),碘化钠(132mg,0.881mmol)加入到N-甲基吡咯烷酮(NMP,6ml)中,氮气保护,120℃加热过夜。将反应液倒入冰水中搅拌,大量黄褐色固体析出,过滤,滤饼用冰水洗三次,烘干,用二氯甲烷/甲醇溶解,硅胶拌样柱层析,得油状物,乙酸乙酯中打浆,过滤,滤饼烘干得淡黄色固体(43mg,收率:15%)。1H-NMR(300Hz,DMSO-d6):δppm10.54(brs,1H),7.71(d,1H),7.64(d,1H),7.43(d,1H),7.27(m,2H),6.90(m,3H),3.44(s,2H),3.11(brt,4H),2.74(m,6H),2.50(t,2H).ESI-MS(m/z):410.2[M+H]+
实施例32 7-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮
将制备例7产物(40mg,0.15mmol)和制备例13产物(50mg,0.15mmol),碳酸钾(41mg,0.3mmol),碘化钾(25mg,0.15mmol)悬于水(3ml)中,加热至80℃反应过夜。反应液冷至室温,过滤,滤饼水洗,烘干得灰色固体(30mg,收率:51%)。1H-NMR(300Hz,DMSO-d6):δppm 11.68(s,1H),8.41(s,1H),7.85(d,1H),7.63(m,2H),7.56(d,1H),7.17(s,1H),7.08(d,1H),6.42(d,1H),3.89(brt,4H),2.84(t,2H),2.60(m,6H).ESI-MS(m/z):392.3[M+H]+
实施例33 7-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)喹啉-2(1H)-酮
将制备例5产物(100mg,0.34mmol),制备例1产物(87mg,0.34mmol),碳酸钾(117mg,0.85mmol),碘化钾(56mg,0.34mmol)悬于乙腈(7.5ml)中,加热至80℃回流反应过夜。将反应液冷至室温,加硅胶拌样,柱层析得到的粗品用甲基叔丁基醚打浆,过滤,收集滤饼,烘干得产品类白色固体(40mg,收率:28%)。1H-NMR(DMSO-d6):δppm 11.70(brs,1H),7.86(d,1H),7.73(d,1H),7.66(d,1H),7.58(d,1H),7.44(d,1H),7.29(t,1H),7.12(s,1H),7.06(d,1H),6.93(d,1H),6.43(d,1H),3.33(t,2H),3.07(m,6H),2.70(brs,4H),1.64(m,4H).ESI-MS(m/z):418.4[M+H]+
实施例34 6-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)喹唑啉-4(3H)-酮盐酸盐
将制备例1产物(60mg,0.206mmol),制备例23产物(71mg,0.229mmol),碳酸钾(95mg,0.688mmol),加入到乙腈(3ml)中,回流过夜。反应液硅胶拌样柱层析,得油状物60mg,在氯化氢-乙醇溶液中成盐酸盐,用丙酮打浆,过滤,滤饼烘干得淡黄色固体(55mg,收 率:56.9%)。1H-NMR(300Hz,DMSO-d6):δppm 12.15(brs,1H),8.03(s,1H),7.93(d,1H),7.68(d,1H),7.66(d,1H),7.61(d,1H),7.58(d,1H),7.38(d,1H),7.27(t,1H),6.87(d,1H),3.03(brs,4H),2.74(t,2H),2.58(brs,4H),2.35(t,2H),1.65(m,2H),1.51(m,2H),1.33(m,2H).ESI-MS(m/z):433.4[M+H]+
实施例35 2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹唑啉-4(3H)-酮
将制备例24产物(120mg,0.57mmol),制备例1产物(145mg,0.57mmol),碳酸钾(236mg,1.71mmol),碘化钾(95mg,0.57mmol)和水(10ml)加入烧瓶中,回流反应过夜,浓缩反应液,残留物经柱层析得产物(100mg,收率:44%)。1H-NMR(400Hz,DMSO-d6):δppm12.29(s,1H),8.09(dd,1H),7.79(td,1H),7.70(d,1H),7.62(d,2H),7.47(t,1H),7.41(d,1H),7.27(t,1H),6.90(d,1H),3.07(brs,4H),2.87(m,4H),2.73(brs,4H).ESI-MS(m/z):391.3[M+H]+
实施例36 3-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将3-(2-氯乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮10-a(133mg,0.55mmol),制备例31产物(120mg,0.55mmol),碳酸钾(229mg,1.65mmol),碘化钾(92mg,0.55mmol)和乙腈(5ml)加入烧瓶中,回流12小时,将反应液浓缩,残留物经柱层析得白色固体(115mg,收率:49%)。1H-NMR(400Hz,DMSO-d6):δppm 7.84(d,1H),7.76(d,1H),7.62(d,1H),7.33(t,1H),7.26(d,1H),5.71(d,1H),4.45(q,1H),3.91(m,1H),3.68(m,1H),3.08(m,3H),2.65(m,2H),2.41(t,2H),2.28(s,3H),2.21(t,2H),2.06-1.70(m,8H).ESI-MS(m/z):424.4[M+H]+
实施例37 5-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-6-氯-二氢吲哚-2-酮
将制备例31产物(120mg,0.553mmol),6-氯-5-(2-氯乙基)二氢吲哚-2-酮(150mg,0.663mmol),碳酸钾(230mg,1.66mmol),碘化钾(90mg,0.553mmol)和水(5ml)加入反应瓶中,105℃回流12小时,冷却反应液,抽滤,滤饼用乙腈打浆得产物(155mg,收率:58%)。1H-NMR(400Hz,DMSO-d6):δppm 10.43(s,1H),7.84(d,1H),7.76(d,1H),7.62(d,1H),7.33(t,1H),7.26(d,1H),7.23(s,1H),6.81(s,1H),3.47(s,2H),3.08(d,3H),2.84(t,2H),2.51(m,2H),2.23(t,2H),1.79(m,4H).ESI-MS(m/z):411.3[M+H]+
实施例38 4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮
将制备例36产物(180mg,0.706mmol)和制备例1产物(180mg,0.706mmol),碳酸钾(243mg,1.77mmol),碘化钾(176mg,1.06mmol)加入乙腈(7.5ml)中,加热至80℃反应过夜,浓缩反应液,残留物经柱层析得黄色固体(126mg,收率:47%)。1H-NMR(300Hz,DMSO-d6):δppm 10.35(s,1H),7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),7.10(t,1H),6.89(d,1H),6.82(d,1H),6.66(d,1H),3.49(s,2H),3.07(brs,4H),2.70(m,6H),2.60(t,2H).ESI-MS(m/z):378.4[M+H]+
实施例39 6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮盐酸盐
将制备例30产物(110mg,0.412mmol),制备例1产物(100mg,0.344mmol),碳酸钾(200mg,1.376mmol),碘化钾(73mg,0.344mmol)悬浮于乙腈(5ml)中,加热回流过夜,浓缩反应液,残留物柱层析得粗品,在氯化氢-乙醇溶液中成盐得灰白色固体(100mg,收率:61%)。1H-NMR(400Hz,DMSO-d6):δppm 11.05(brs,1H),10.52(s,1H),7.79(d,1H),7.72(d,1H),7.50(d,1H),7.33(t,1H),7.19(d,1H),6.99(d,1H),6.87(dd,1H),6.77(d,1H),3.69(d,2H),3.58(d,2H),3.46(s,2H),3.38(m,4H),3.24(t,2H),3.09(m,2H).ESI-MS(m/z):378.2[M+H]+
实施例40 7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯喹啉-2(1H)-酮
将制备例32产物(24mg,0.079mmol)和制备例1产物(20mg,0.079mmol),碳酸钾(22mg,0.158mmol),碘化钾(13mg)悬于水(3ml)中,加热至80℃反应过夜。反应液冷至室温,过滤,滤饼用水洗,烘干得灰色固体(23mg,收率:69%)。1H-NMR(300Hz,DMSO-d6):δppm11.77(brs,1H),7.80(m,2H),7.67(d,1H),7.59(d,1H),7.39(d,1H),7.26(m,2H),6.88(d,1H),6.48(d,1H),3.08(brs,4H),2.94(t,2H),2.71(brs,4H),2.63(t,2H).ESI-MS(m/z):424.2[M+H]+
实施例41 9-羟基-2-甲基-3-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
将制备例26产物(200mg,0.632mmol)和3-(2-氯乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮10-a(158mg,0.695mmol),碳酸钾(218mg,1.58mmol,碘化钾(157mg,0.948mmol)加入乙腈(7.5ml)中,加热至80℃反应过夜。浓缩反应液,残留物经柱层析得黄色固体(125mg,收率:46.6%)。1H-NMR(300Hz,DMSO-d6):δppm 8.91(s,1H),8.06(m,2H),7.53(d,1H),5.71(d,1H),4.45(q,1H),3.91(m,1H),3.68(m,1H),3.21(brs,4H),2.95-2.61(m,6H),2.50(t,2H),2.29(s,3H),2.10-1.70(m,4H).ESI-MS(m/z):426.3[M+H]+
实施例42 6-氯-5-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮
取制备例26产物(200mg,0.632mmol)和6-氯-5-(2-氯乙基)二氢吲哚-2-酮(160mg,0.695mmol),碳酸钾(218mg,1.58mmol),碘化钾(157mg,0.948mmol)加入乙腈(7.5ml)中,加热至80℃反应过夜。浓缩反应液,残留物经柱层析得白色固体(25mg,收率:8.7%)。1H-NMR(300Hz,DMSO-d6):δppm 10.41(s,1H),8.89(s,1H),8.04(m,2H),7.51(d,1H),7.23(s,1H),6.81(s,1H),3.46(s,2H),3.18(brs,4H),2.85(t,2H),2.71(brs,4H),2.56(t,2H).ESI-MS(m/z):413.3[M+H]+
实施例43 7-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮
取制备例26产物(177mg,0.56mmol)和制备例7产物(150mg,0.56mmol),碳酸钾(271mg,1.96mmol),碘化钾(140mg,0.84mmol)加入乙腈(7.5ml)中,加热至80℃反应过夜。浓缩反应液,残留物经柱层析得白色固体(75mg,收率:34.4%)。1H-NMR(DMSO-d6):δppm 11.68(s,1H),8.90(s,1H),8.05(s,2H),7.85(d,1H),7.54(m,2H),7.18(s,1H),7.09(m,1H),6.42(d,1H),3.19(brs,4H),2.86(t,2H),2.74(brs,6H).ESI-MS(m/z):391.3[M+H]+
实施例44 7-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)喹啉-2(1H)-酮
取制备例28产物(100mg,0.355mmol)和制备例1产物(91mg,0.355mmol),碳酸钾(172mg,1.24mmol),碘化钾(88mg,0.53mmol)加入乙腈(7.5ml)中,加热至80℃反应过夜。将反应液冷至室温,倒入水(30ml)中,析出固体,过滤,滤饼水洗,烘干得粗品。粗品用乙酸乙酯(2.0ml)打浆,过滤,烘干得类白色固体(88mg,收率:61.5%)。1H-NMR(DMSO-d6):δppm 11.68(s,1H),7.85(d,1H),7.69(d,1H),7.61(d,1H),7.56(d,1H),7.38(d,1H),7.27(t,1H),7.14(s,1H),7.06(d,1H),6.89(d,1H),6.42(d,1H),3.07(brs,4H),2.69(t,2H),2.60(brs,4H),2.38(t,2H),1.79(m,2H).ESI-MS(m/z):404.4[M+H]+
实施例45 7-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基-3,4-二氢喹啉-2(1H)-酮
取制备例27产物(120mg,0.424mmol)和制备例1产物(108mg,0.424mmol),碳酸钾(205mg,1.484mmol),碘化钾(106mg,0.636mmol)加入乙腈(7.5ml)中,加热至80℃反应过夜。将反应液冷至室温,倒入水(30ml)中,析出固体,过滤,滤饼水洗,烘干得粗品。粗品用石油醚-乙酸乙酯打浆,过滤,烘干得类白色固体(58mg,收率:34%)。1H-NMR(DMSO-d6):δppm 10.01(s,1H),7.69(d,1H),7.61(d,1H),7.39(d,1H),7.27(t,1H),7.05(d,1H),6.89(d,1H),6.76(d,1H),6.70(s,1H),3.07(brs,4H),2.81(t,2H),2.59(brs,4H),2.54(t,2H),2.39(m,4H),1.72(m,2H).ESI-MS(m/z):406.3[M+H]+
实施例46 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-吲哚
取制备例34产物(200mg,0.83mmol)和制备例1产物(182mg,0.83mmol),碳酸钾(347mg,2.5mmol),碘化钾(139mg,0.83mmol)加入乙腈(5ml)中,加热至80℃反应过夜,浓缩反应液,残留物经柱层析得白色固体(120mg,收率:39%)。1H-NMR(DMSO-d6):δppm10.85(s,1H),7.71(d,1H),7.63(d,1H),7.56(d,1H),7.43(d,1H),7.35(d,1H),7.29(t,1H),7.20(s,1H),7.07(t,1H),6.98(t,1H),6.92(d,1H),3.16(brs,4H),3.04-2.63(m,8H).ESI-MS(m/z):362.1[M+H]+
实施例47 6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮盐酸盐
将制备例35产物(120mg,0.57mmol),制备例1产物(163mg,0.56mmol),碳酸钠(238mg,2.23mmol),碘化钠(2mg)加入到N,N-二甲基甲酰胺(3ml)中,氮气保护,120℃反应9小时。将反应液倒入冰水中,析出淡黄色固体,过滤,滤饼用冰水洗三次,烘干,柱层析得粗品,在氯化氢-乙醇溶液中成盐酸盐,乙腈/甲醇中打浆,过滤,滤饼用乙腈洗,烘干得淡黄色固体(80mg,收率:33%)。1H-NMR(300Hz,DMSO-d6):δppm 9.99(s,1H),7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),7.05(d,1H),7.01(d,1H),6.90(d,1H),6.76(d,1H),3.07(brt,4H),2.83(t,2H),2.69(m,6H),2.56(t,2H),2.42(t,2H).ESI-MS(m/z):392.1[M+H]+
实施例48 5-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)二氢吲哚-2-酮盐酸盐
将制备例25产物(100mg,0.48mmol),制备例1产物(136mg,0.47mmol),碳酸钠(198mg,1.87mmol),碘化钠(71mg,0.48mmol)加入到水(4ml)中,氮气保护,回流9小时,向反应液中加入二氯甲烷萃取两次,合并有机相,干燥,浓缩,残留物经柱层析得粗品,在氯化氢-乙醇溶液中成盐酸盐,乙酸乙酯/甲醇中打浆,过滤,滤饼用乙酸乙酯洗,烘干得白色固体(135mg,收率:68%)。1H-NMR(300Hz,DMSO-d6):δppm 10.27(s,1H),7.69(d,1H),7.61(d,1H),7.39(d,1H),7.27(t,1H),7.07(s,1H),7.00(d,1H),6.89(d,1H),6.71(d,1H),3.43(s,2H),3.07(brs,4H),2.59(brs,4H),2.55(t,2H),2.37(t,2H),1.73(m,2H).ESI-MS(m/z):392.2[M+H]+
实施例49 7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-4,5-二氢-1H-苯并氮杂卓-2(3H)-酮盐酸盐
将制备例29产物(100mg,0.45mmol),制备例1产物(130mg,0.45mmol),碳酸钾(246mg,1.79mmol),碘化钾(74mg,0.45mmol)加入到乙腈(4ml)中,回流20小时,反应液中加入水,用二氯甲烷萃取2次,合并有机相,干燥,浓缩,残留物经柱层析得粗品,在氯化氢-乙醇溶液中成盐酸盐,乙酸乙酯中打浆,过滤,滤饼用乙酸乙酯洗,烘干得淡黄色固体(90mg,收率:45.6%)。1H-NMR(300Hz,DMSO-d6):δppm 9.45(s,1H),7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),7.11(m,2H),6.89(m,2H),3.08(brs,4H),2.80-2.54(m,10H),2.11(m,4H).ESI-MS(m/z):406.3[M+H]+
实施例50 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮盐酸盐
将3-(2-氯乙基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮(40mg,0.18mmol),制备例1产物(50mg,0.17mmol),碳酸钾(82mg,0.60mmol),碘化钾(2mg)加入到乙腈(2ml)中,回流过夜。反应液中加入水,用二氯甲烷萃取2次,合并有机相,干燥,浓缩,残留物经柱层析得粗品,在氯化氢-乙醇溶液中成盐酸盐,乙酸乙酯-正己烷中打浆,过滤,滤饼用乙酸乙酯洗,烘干得白色固体(40mg,收率:49%)。1H-NMR(300Hz,DMSO-d6):δppm 8.89(d,1H),7.85(t,1H),7.69(d,1H),7.60(t,2H),7.42(d,1H),7.28(t,2H),6.91(d,1H),3.09(brs,4H),2.85(t,2H),2.73(brs,4H),2.54(t,2H),2.50(s,3H).ESI-MS(m/z):405.3[M+H]+
实施例51 6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3-甲基-3,4-二氢喹唑啉-2(1H)-酮
将制备例33产物(180mg,0.80mmol),制备例1产物(230mg,0.79mmol),碳酸钠(335mg,3.16mmol),碘化钠(4mg)加入到N,N-二甲基甲酰胺(4ml)中,氮气保护,120℃反应16小时,将反应液倒入冰水中,析出淡黄色固体,过滤,滤饼用冰水洗3次,烘干,经柱层析得粗品110mg,在乙酸乙酯中打浆,过滤,滤饼用乙酸乙酯洗,烘干得淡黄色固体(80mg,收率:25%)。1H-NMR(300Hz,DMSO-d6):δppm 9.12(s,1H),7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),7.00(d,1H),6.96(s,1H),6.89(d,1H),6.68(d,1H),4.36(s,2H),3.07(brs,4H),2.84(s,3H),2.67(m,6H),2.55(t,2H).ESI-MS(m/z):407.3[M+H]+
实施例52 3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-氯-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮
冰浴下将二氯亚砜(6.8ul,0.094mmol)的二氯甲烷溶液滴加到实施例4产物(20mg,0.047mmol)的二氯甲烷溶液中,10℃下反应1小时,用二氯甲烷稀释反应液,饱和碳酸氢钠水溶液洗,干燥有机相,浓缩,残留物经柱层析得目标物(19mg,收率:91%)。1H-NMR(300Hz,MeOH-d4):δppm 7.66(d,1H),7.60(d,1H),7.50(d,1H),7.33(t,1H),7.03(d,1H),5.18(m,1H),4.30(m,1H),3.71(m,1H),3.49(brs,4H),3.41(brs,4H),3.21(t,2H),3.03(m,2H),2.43(s,3H),2.35(m,3H),2.14(m,1H).ESI-MS(m/z):443.3[M+H]+
实施例53 5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-硫酮盐酸盐
取实施例8产物(500mg,1.325mmol)悬于甲苯(15ml)中,加入劳森试剂(642mg,1.59mmol),加热至80℃反应过夜,浓缩反应液,残留物经柱层析得粗品,在氯化氢-甲醇溶液中成盐,过滤,滤饼再用异丙醚/甲醇的混合溶剂打浆,得黄色固体(40mg,收率:7.6%)。1H-NMR(300Hz,DMSO-d6):δppm 12.66(s,1H),10.96(brs,1H),7.77(d,1H),7.71(d,1H),7.49(d,1H),7.32(t,1H),7.25(s,1H),7.19(d,1H),6.99(d,1H),6.96(d,1H),4.06(s,2H),3.02-3.74(m,12H).
实施例54 7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-硫酮
取实施例9产物(200mg,0.51mmol)悬于甲苯(20ml)中,加入劳森试剂(416mg,1.02mmol),加热至80℃反应14小时,浓缩反应液,残留物经柱层析得黄色固体(60mg,收率:28.8%)。1H-NMR(300Hz,DMSO-d6):δppm 13.64(s,1H),7.80(d,1H),7.72(m,2H),7.63(d,1H),7.51(s,1H),7.43(d,1H),7.28(m,2H),7.21(d,1H),6.92(d,1H),3.14(brt,4H),2.64-3.06(m,8H).ESI-MS(m/z):406.2[M+H]+
实施例55 2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5,6-二乙基嘧啶-4(3H)-酮盐酸盐
取制备例37产物(270mg,0.72mmol)和制备例1产物(314mg,1.44mmol)溶于乙腈(10ml)中,氮气保护,加热回流过夜。反应液冷至室温,浓缩,残留物经柱层析得粗品160mg,溶于甲醇(5ml)中,加入氯化氢-甲醇溶液(1.0ml),室温搅拌3小时,将反应液浓缩至干,残留物经柱层析纯化得到目标物(35mg,收率:12.2%)。1H-NMR(300Hz,DMSO-d6):δppm12.23(brs,1H),7.69(d,1H),7.61(d,1H),7.40(d,1H),7.27(t,1H),6.89(d,1H),3.05(brt,4H),2.61-2.86(m,8H),2.31-2.57(m,4H),1.13(t,3H),0.99(t,3H).ESI-MS(m/z):397.2[M+H]+
实施例56 2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)嘧啶-4(3H)-酮盐酸盐
参照实施例55的方法,以制备例38产物和制备例1产物为原料制备目标物。1H-NMR(300Hz,DMSO-d6):δppm 9.45(brs,1H),7.96(d,1H),7.77(d,1H),7.70(d,1H),7.50(d,1H),7.31(t,1H),6.96(d,1H),6.36(d,1H),3.63(t,2H),3.17-3.57(m,10H).ESI-MS(m/z):341.0[M+H]+
在下表中显示的是实施例57~154中的化合物,除了使用与最终产物相对应的起始原料和中间体以外,使用与以上实施例中相同的方法,就可以制备出这些化合物。
Figure PCTCN2015073854-appb-000062
Figure PCTCN2015073854-appb-000063
Figure PCTCN2015073854-appb-000064
Figure PCTCN2015073854-appb-000065
Figure PCTCN2015073854-appb-000066
Figure PCTCN2015073854-appb-000067
Figure PCTCN2015073854-appb-000068
Figure PCTCN2015073854-appb-000069
Figure PCTCN2015073854-appb-000070
Figure PCTCN2015073854-appb-000071
Figure PCTCN2015073854-appb-000072
Figure PCTCN2015073854-appb-000073
Figure PCTCN2015073854-appb-000074
Figure PCTCN2015073854-appb-000075
Figure PCTCN2015073854-appb-000076
Figure PCTCN2015073854-appb-000077
Figure PCTCN2015073854-appb-000078
Figure PCTCN2015073854-appb-000079
Figure PCTCN2015073854-appb-000080
药理实验
1)5-HT1A激动活性测试
使用LANCETM cAMP 384 Kit(美国PerkinElmer公司产品)测试了化合物对表达人重组5-HT1A受体HEK293细胞的5-HT1A受体的激动作用。通过测试化合物对HEK293细胞中cAMP生成的抑制作用来评估化合物的5-HT1A激动作用。cAMP浓度测试按照试剂盒说明书中的方法进行,化合物的测试浓度为0.1nM-10000nM,采用8-OH-DPAT作为阳性对照,EC50由Excelfit软件计算得到,结果见表1。
2)D2拮抗活性测试
使用LANCETM cAMP 384 Kit(美国PerkinElmer公司产品)测试了化合物对表达人重组D2受体HEK293细胞的D2受体的拮抗作用。通过测试化合物拮抗多巴胺对HEK293细胞中cAMP生成的抑制作用来评估化合物的D2拮抗作用。cAMP浓度测试按照试剂盒说明书中的方法进行,化合物的测试浓度为0.1nM-10000nM,采用利培酮作为阳性对照,IC50由Excelfit软件计算得到,结果见表1。
3)5-HT2A拮抗活性测试
使用
Figure PCTCN2015073854-appb-000081
Calcium 5 Assay Kit(美国Molecular Devices公司产品)测试了化合物对表达人重组5-HT2A受体CHO-K1细胞的5-HT2A受体的拮抗作用。测试按照试剂盒说明书中的方法进行,化合物的测试浓度为0.1nM-10000nM,采用利培酮作为阳性对照。测试方法如下:第一天种子细胞以1400万每瓶的密度置于含25ml生长培养液(F-12营养混合物+10%FBS+1%青霉素/链霉素+1.2%50mg/ml Geneticin)的T-175瓶中,在37℃,5% CO2,加湿的条件下,细胞培养24小时;第二天将种子细胞接种至384孔细胞培养板中,每孔接种含2万个细胞,用50μL检测培养液(F-12营养混合物+1.5%活性碳处理过的FBS)替换生长培养液,在37℃,5%CO2,加湿的条件下,细胞培养16小时;第三天除去培养基,细胞培养板每孔加入24μL新配的上样染料溶液(按说明书方法配制),将板置于培养箱中,37℃,5%CO2湿化条件下,孵化120分钟;转移6μL配好的待测化合物溶液到分析板中,轻轻震摇1分钟,37℃,5%CO2湿化条件下,孵化30分钟;分析板每孔加入10μL新配制的1.2μM的α-甲基-5-羟色胺溶液(α-甲基-5-羟色胺的最终浓度是300nM),用Flipr(美国Molecular Devices公司产品)进行检测分析。计算不同浓度化合物的抑制率,IC50由Excelfit软件计算得到,结果见表1。
表1:
Figure PCTCN2015073854-appb-000082
Figure PCTCN2015073854-appb-000083
Figure PCTCN2015073854-appb-000084
Figure PCTCN2015073854-appb-000085
4)D2激动活性测试
使用LANCETM cAMP 384 Kit(美国PerkinElmer公司产品)测试了化合物对表达人重组D2受体HEK293细胞的D2受体的激动作用。通过测试化合物对HEK293细胞中cAMP生成的抑制作用来评估化合物的D2激动作用。cAMP浓度测试按照试剂盒说明书中的方法进行,化合物的测试浓度为0.1nM-10000nM,采用多巴胺作为阳性对照,IC50由Excelfit软件计算得到,结果见表2。
表2:
Figure PCTCN2015073854-appb-000086
Figure PCTCN2015073854-appb-000087
5)体内药效试验(PCP诱导的小鼠高自发活动试验)
PCP溶于生理盐水中,配制成7mg/kg剂量的溶液。阿立哌唑用及受试化合物用0.5%CMC-Na溶液配制成合适浓度的溶液,现配现用。雄性ICR小鼠,18-22g.试验时小鼠随机分为溶剂对照组、模型对照组、阳性对照组、及各受试药组。每组8只小鼠。各组小鼠分别灌胃给予各受试药。给受试药后45分钟,分别给予小鼠腹腔注射PCP(7mg/kg)溶液。用自发、旷场视频分析系统记录给予受试药或生理盐水45min后小鼠活动轨迹,然后记录给予PCP后75min内小鼠的活动轨迹。用自发、旷场视频分析系统分析小鼠的活动轨迹,统计各组小鼠的活动总路程,其结果用mean±SD表示。结果采用单因素方差分析进行统计。
PCP造模后,小鼠自发活动与生理盐水组相比显著增加,受试化合物在以下剂量(表3)均可显著降低PCP诱导的小鼠高自发活动,与模型组相比具有显著性差异。
表3:
Figure PCTCN2015073854-appb-000088
Figure PCTCN2015073854-appb-000089
Figure PCTCN2015073854-appb-000090

Claims (14)

  1. 一种由通式(I)表示的杂环化合物、其立体异构体或其药学上可接受的盐:
    Figure PCTCN2015073854-appb-100001
    其中:
    A、B和D各自独立地为C或N,且当A为N时,D同时也为N;
    Figure PCTCN2015073854-appb-100002
    表示单键或双键;
    E为CH,N或C;当E为CH或N时,与E相连的
    Figure PCTCN2015073854-appb-100003
    表示单键;当E为C时,与E相连的
    Figure PCTCN2015073854-appb-100004
    表示双键;
    R1为氢或1至4个各自独立地选自卤素、羟基、巯基、氧代(=O)、硫代(=S)、C1~C6烷氧基、卤代C1~C6烷氧基、C1~C6烷硫基、C1~C6烷基、卤代C1~C6烷基、硝基、氨基、被C1~C6烷基取代的氨基、氰基、羧基、醛基、氨基C1~C6烷基、羟基C1~C6烷基、氰基C1~C6烷基、C1~C6烷酰基、卤代C1~C6烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C6烷基取代的氨基甲酰基、羧基C1~C6烷基、C1~C6烷磺酰基、卤代C1~C6烷磺酰基、被C1~C6烷基取代的氨基C1~C6烷基、氨基甲酰基C1~C6烷基或被C1~C6烷基取代的氨基甲酰基C1~C6烷基;
    R2不存在,或者为1至3个各自独立地选自卤素、羟基、巯基、C1~C6烷氧基、卤代C1~C6烷氧基、C1~C6烷硫基、C1~C6烷基、卤代C1~C6烷基、硝基、氨基、被C1~C6烷基取代的氨基、氰基、羧基、醛基、氨基C1~C6烷基、羟基C1~C6烷基、氰基C1~C6烷基、C1~C6烷酰基、卤代C1~C6烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C6烷基取代的氨基甲酰基、羧基C1~C6烷基、C1~C6烷磺酰基、卤代C1~C6烷磺酰基、被C1~C6烷基取代的氨基C1~C6烷基、氨基甲酰基C1~C6烷基或被C1~C6烷基取代的氨基甲酰基C1~C6烷基;
    R3为氢或1至4个各自独立地选自羟基或C1~C6烷基的取代基;
    L不存在,或者为C1~C5亚烷基,且当L为C1~C5亚烷基时,该亚烷基非必须地被一个或多个选自羟基、C1~C6烷氧基和氧代基团(O=)中的取代基所取代;
    G环为单环杂环基或双环杂环基,所述双环杂环基为苯并单环杂环基、环烃基并单环杂环基或单环杂环基并单环杂环基,其中所述单环杂环基含有至少一个选自N、S和O的杂原子;
    G环与L的连接方式为通过G环上的碳原子与L相连;
    并且,G环非必须地被一个或多个相同或不同取代基取代;
    所述G环上的取代基为卤素、C1~C6烷基、卤代C1~C6烷基、C1~C6烷氧基、卤代C1~C6烷氧基、硝基、氰基、羟基、巯基、氨基、被C1~C6烷基取代的氨基、叠氮基、C1~C6烷酰基、卤代C1~C6烷酰基、C2~C6链烯基、C2~C6炔基、羧基C1~C6烷基、氰基C1~C6烷基、C2~C6链烯氧基、C2~C6炔氧基、氨基甲酰基(-CONH2)、被C1~C6烷基取代的氨基甲酰基、羧基、羟基C1~C6烷基、氧代(=O)、硫代(=S)、磺酰氨基(-SO2NH2)、C1~C6烷硫基、C1~C6烷磺酰基、卤代C1~C6烷磺酰基、磺酸基(-SO2OH)、醛基、氨基C1~C6烷基、被C1~C6烷基取代的氨基C1~C6烷基、氨基甲酰基C1~C6烷基、被C1~C6烷基取代的氨基甲酰基C1~C6烷基、C3-C10环烃基、C3-C10环烃基C1~C6烷基、C3-C10环烃基甲酰胺基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C10环烃基C1~C6烷氧基、呋喃基C1~C6烷基、呋喃基C1~C6烷氧基、噻吩基C1~C6烷基、噻吩基C1~C6烷氧基、吡咯基C1~C6烷基、吡咯基C1~C6烷氧基、吡咯烷基C1~C6烷基、吡咯烷基C1~C6烷氧基、吡唑基C1~C6烷基、吡唑基C1~C6烷氧基、三唑基C1~C6烷基、三唑基C1~C6烷氧基、噻唑基C1~C6烷基、噻唑基C1~C6烷氧基、异噻唑基C1~C6烷基、异噻唑基C1~C6烷氧基、噁唑基C1~C6烷基、噁唑基C1~C6烷氧基、异噁唑基C1~C6烷基、异噁唑基C1~C6烷氧基、吡嗪基C1~C6烷基、吡嗪基C1~C6烷氧基、哒嗪基C1~C6烷基、哒嗪基C1~C6烷氧基、吡啶基C1~C6烷基、吡啶基C1~C6烷氧基、嘧啶基C1~C6烷基、嘧啶基C1~C6烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C6烷基、苯基C1~C6烷氧基、苯基C1~C6烷酰基或苯基C1~C6烷酰氧基;上述C3-C10环烃基、C3-C10环烃基C1~C6烷基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C10环烃基C1~C6烷氧基、呋喃基C1~C6烷基、呋喃基C1~C6烷氧基、噻吩基C1~C6烷基、噻吩基C1~C6烷氧基、吡咯基C1~C6烷基、吡咯基C1~C6烷氧基、吡咯烷基C1~C6烷基、吡咯烷基C1~C6烷氧基、吡唑基C1~C6烷基、吡唑基C1~C6烷氧基、三唑基C1~C6烷基、三唑基C1~C6烷氧基、噻唑基C1~C6 烷基、噻唑基C1~C6烷氧基、异噻唑基C1~C6烷基、异噻唑基C1~C6烷氧基、噁唑基C1~C6烷基、噁唑基C1~C6烷氧基、异噁唑基C1~C6烷基、异噁唑基C1~C6烷氧基、吡嗪基C1~C6烷基、吡嗪基C1~C6烷氧基、哒嗪基C1~C6烷基、哒嗪基C1~C6烷氧基、吡啶基C1~C6烷基、吡啶基C1~C6烷氧基、嘧啶基C1~C6烷基、嘧啶基C1~C6烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C6烷基、苯基C1~C6烷氧基、苯基C1~C6烷酰基和苯基C1~C6烷酰氧基非必须地被一个或多个选自如下基团的取代基所取代:卤素、C1~C6烷基、卤代C1~C6烷基、C1~C6烷氧基、C1~C6烷氧酰基、卤代C1~C6烷氧基、硝基、氰基、羟基、氨基、C1~C6烷酰基、卤代C1~C6烷酰基、氨基甲酰基或羧基;
    条件是不包括以下化合物:
    1)1-[2-[4-(2-氰基-7-氟苯并噻吩-4-基)-2-甲基-1-哌嗪基]乙基]-3,4-二氢-1H-2-苯并吡喃-6-甲酰胺;
    2)1-[2-[4-(7-氟苯并噻吩-4-基)-2-甲基-1-哌嗪基]乙基]-3,4-二氢-1H-2-苯并吡喃-6-甲酰胺;
    3)6-乙基-4-[4-[2-(2-噻吩基)乙基]-1-哌嗪基]-噻吩并[2,3-d]嘧啶;
    4)6-[2-[4-(5-甲基噻吩并[2,3-d]嘧啶-4-基)-1-哌嗪基]乙基]-2H-1,4-苯并噁嗪-3(4H)-酮;
    5)6-乙基-4-[4-[3-[3-(2-噻吩基)-1,2,4-噁二唑-5-基]丙基]-1-哌嗪基]-噻吩并[2,3-d]嘧啶;
    6)5,6-二甲基-4-[4-[3-[3-(4-甲基苯基)-1,2,4-噁二唑-5-基]丙基]-1-哌嗪基]-噻吩并[2,3-d]嘧啶。
  2. 根据权利要求1所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,G环为3~10元单环杂环基、苯并[3~10元单环杂环基]、[C3-C10环烃基]并[3~10元单环杂环基]和[3~10元单环杂环基]并[3~10元单环杂环基]。
  3. 根据权利要求2所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,G环为5~7元单环杂环基、苯并[5~7元单环杂环基]、[C5-C7环烃基]并[5~7元单环杂环基]和[5~7元单环杂环基]并[5~7元单环杂环基]。
  4. 根据权利要求3所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,G环为呋喃基、二氢呋喃基、四氢呋喃基、噻吩基、二氢噻吩基、四氢噻吩基、吡咯基、二氢吡咯基、吡咯烷基、吡唑基、二氢吡唑基、吡唑烷基、三唑基、二氢三唑基、三唑烷基、噻唑基、二氢噻唑基、噻唑烷基、异噻唑基、二氢异噻唑基、异噻唑烷基、噁唑基、二氢噁唑基、噁唑烷基、异噁唑基、二氢异噁唑基、异噁唑烷基、吡喃基、二氢吡喃基、 四氢吡喃基、吡嗪基、二氢吡嗪基、四氢吡嗪基、哌嗪基、哒嗪基、二氢哒嗪基、四氢哒嗪基、
    Figure PCTCN2015073854-appb-100005
    Figure PCTCN2015073854-appb-100006
    Figure PCTCN2015073854-appb-100007
    Figure PCTCN2015073854-appb-100008
    Figure PCTCN2015073854-appb-100009
  5. 根据权利要求1-4所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,
    R1为氢或1至4个各自独立地选自卤素、羟基、巯基、氧代(=O)、硫代(=S)、C1~C4烷氧基、卤代C1~C4烷氧基、C1~C4烷硫基、C1~C4烷基、卤代C1~C4烷基、硝基、氨基、被C1~C4烷基取代的氨基、氰基、羧基、醛基、氨基C1~C4烷基、羟基C1~C4烷基、氰基C1~C4烷基、C1~C4烷酰基、卤代C1~C4烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C4烷基取代的氨基甲酰基、羧基C1~C4烷基、C1~C4烷磺酰基、卤代C1~C4烷磺酰基、被C1~C4烷基取代的氨基C1~C4烷基、氨基 甲酰基C1~C4烷基或被C1~C4烷基取代的氨基甲酰基C1~C4烷基;
    R2为不存在,或者为1至3个各自独立地选自卤素、羟基、巯基、C1~C4烷氧基、卤代C1~C4烷氧基、C1~C4烷硫基、C1~C4烷基、卤代C1~C4烷基、硝基、氨基、被C1~C4烷基取代的氨基、氰基、羧基、醛基、羟基C1~C4烷基、氰基C1~C4烷基、C1~C4烷酰基、卤代C1~C4烷酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基(-CONH2)、被C1~C4烷基取代的氨基甲酰基、羧基C1~C4烷基、C1~C4烷磺酰基、卤代C1~C4烷磺酰基、氨基C1~C4烷基、被C1~C4烷基取代的氨基C1~C4烷基、氨基甲酰基C1~C4烷基或被C1~C4烷基取代的氨基甲酰基C1~C4烷基;
    R3为氢或1至4个各自独立地选自羟基或C1~C4烷基的取代基;
    L不存在,或者为C1~C4亚烷基,且该亚烷基非必须地被一个或多个选自羟基、C1~C6烷氧基和氧代基团(O=)中的取代基所取代;
    G环与L的连接方式为通过G环上的碳原子与L相连;并且,G环非必须地被一个或多个相同或不同取代基取代;
    所述G环上的取代基为卤素、C1~C4烷基、卤代C1~C4烷基、C1~C4烷氧基、卤代C1~C4烷氧基、硝基、氰基、羟基、巯基、氨基、被C1~C4烷基取代的氨基、叠氮基、C1~C4烷酰基、卤代C1~C4烷酰基、C2~C4链烯基、C2~C4炔基、羧基C1~C4烷基、氰基C1~C4烷基、C2~C4链烯氧基、C2~C4炔氧基、氨基甲酰基(-CONH2)、被C1~C4烷基取代的氨基甲酰基、羧基、羟基C1~C4烷基、氧代(=O)、硫代(=S)、磺酰氨基(-SO2NH2)、C1~C4烷硫基、C1~C4烷磺酰基、卤代C1~C4烷磺酰基、磺酸基(-SO2OH)、醛基、氨基C1~C4烷基、被C1~C4烷基取代的氨基C1~C4烷基、氨基甲酰基C1~C4烷基、被C1~C4烷基取代的氨基甲酰基C1~C4烷基、C3-C7环烃基、C3-C7环烃基C1~C4烷基、C3-C7环烃基甲酰胺基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C7环烃基C1~C4烷氧基、呋喃基C1~C4烷基、呋喃基C1~C4烷氧基、噻吩基C1~C4烷基、噻吩基C1~C4烷氧基、吡咯基C1~C4烷基、吡咯基C1~C4烷氧基、吡咯烷基C1~C4烷基、吡咯烷基C1~C4烷氧基、吡唑基C1~C4烷基、吡唑基C1~C4烷氧基、三唑基C1~C4烷基、三唑基C1~C4烷氧基、噻唑基C1~C4烷基、噻唑基C1~C4烷氧基、异噻唑基C1~C4烷基、异噻唑基C1~C4烷氧基、噁唑基C1~C4烷基、噁唑基C1~C4烷氧基、异噁唑基C1~C4烷基、异噁唑基C1~C4烷氧基、吡嗪基C1~C4烷基、吡嗪基C1~4烷氧基、哒嗪基C1~C4烷基、哒嗪基C1~C4烷氧基、吡啶基C1~C4烷基、吡啶基C1~C4烷氧基、嘧啶基C1~C4烷基、嘧啶基C1~C4烷氧基、苯基、苯氧基、苯磺酰基、 苯基C1~C4烷基、苯基C1~C4烷氧基、苯基C1~C4烷酰基或苯基C1~C4烷酰氧基;上述C3-C7环烃基、C3-C7环烃基C1~C4烷基、呋喃基、噻吩基、吡咯基、吡咯烷基、吡唑基、吡唑烷基、三唑基、三唑烷基、噻唑基、噻唑烷基、异噻唑基、异噻唑烷基、噁唑基、噁唑烷基、异噁唑基、异噁唑烷基、吡喃基、吡嗪基、哌嗪基、哒嗪基、吡啶基、哌啶基、嘧啶基、咪唑基、C3-C7环烃基C1~C4烷氧基、呋喃基C1~C4烷基、呋喃基C1~C4烷氧基、噻吩基C1~C4烷基、噻吩基C1~C4烷氧基、吡咯基C1~C4烷基、吡咯基C1~C4烷氧基、吡咯烷基C1~C4烷基、吡咯烷基C1~C4烷氧基、吡唑基C1~C4烷基、吡唑基C1~C4烷氧基、三唑基C1~C4烷基、三唑基C1~C4烷氧基、噻唑基C1~C4烷基、噻唑基C1~C4烷氧基、异噻唑基C1~C4烷基、异噻唑基C1~C4烷氧基、噁唑基C1~C4烷基、噁唑基C1~C4烷氧基、异噁唑基C1~C4烷基、异噁唑基C1~C4烷氧基、吡嗪基C1~C4烷基、吡嗪基C1~4烷氧基、哒嗪基C1~C4烷基、哒嗪基C1~C4烷氧基、吡啶基C1~C4烷基、吡啶基C1~C4烷氧基、嘧啶基C1~C4烷基、嘧啶基C1~C4烷氧基、苯基、苯氧基、苯磺酰基、苯基C1~C4烷基、苯基C1~C4烷氧基、苯基C1~C4烷酰基和苯基C1~C4烷酰氧基非必须地被一个或多个选自如下基团的取代基所取代:卤素、C1~C4烷基、卤代C1~C4烷基、C1~C4烷氧基、C1~C4烷氧酰基、卤代C1~C4烷氧基、硝基、氰基、羟基、氨基、C1~C4烷酰基、卤代C1~C4烷酰基、氨基甲酰基或羧基。
  6. 根据权利要求5所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,
    R1为氢或1至4个各自独立地选自氟、氯、溴、羟基、巯基、氧代(=O)、硫代(=S)、甲氧基、乙氧基、三氟甲氧基、-SCH3、-SCH2CH3、甲基、乙基、丙基、异丙基、叔丁基、三氟甲基、溴甲基、氯甲基、硝基、氨基、N-甲基氨基、N-乙基氨基、N,N-二甲基氨基、N,N-二乙基氨基、氰基、羧基、醛基、-CH2NH2、-CH2CH2NH2、-CH2OH、-CH2CH2OH、-CH2CN、-CH2CH2CN、甲酰基、乙酰基、丙酰基、三氟乙酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基、N-甲基氨基甲酰基、N,N-二甲基氨基甲酰基、N-乙基氨基甲酰基、N,N-二乙基氨基甲酰基、-CH2CO2H、-CH2CH2CO2H、-SO2CH3、-SO2CF3、-CH2NHMe、-CH2NMe2、-CH2CONH2、-CH2CONHMe或-CH2CONMe2
    R2不存在,或者为1至3个各自独立地选自氟、氯、溴、羟基、巯基、甲氧基、乙氧基、三氟甲氧基、-SCH3、-SCH2CH3、甲基、乙基、丙基、异丙基、叔丁基、三氟甲基、溴甲基、氯甲基、硝基、氨基、N-甲基氨基、N-乙基氨基、N,N-二甲基氨基、N,N-二乙基氨基、氰基、羧基、醛基、-CH2OH、-CH2CH2OH、-CH2CN、-CH2CH2CN、甲酰基、乙酰基、丙酰基、三氟乙酰基、磺酸基(-SO2OH)、磺酰氨基(-SO2NH2)、氨基甲酰基、N-甲基氨基甲酰基、N,N-二甲基氨基甲酰基、N-乙基氨基甲酰基、N,N-二乙基氨基甲酰 基、-CH2CO2H、-CH2CH2CO2H、-SO2CH3、-SO2CF3、-CH2NH2、-CH2CH2NH2、-CH2NHMe、-CH2NMe2、-CH2CONH2、-CH2CONHMe或-CH2CONMe2
    R3为氢或1至4个各自独立地选自羟基、甲基或乙基的取代基;
    所述G环上的取代基为氟、氯、溴、甲基、乙基、丙基、异丙基、叔丁基、三氟甲基、甲氧基、乙氧基、三氟甲氧基、硝基、氰基、羟基、巯基、氨基、N-甲基氨基、N-乙基氨基、N,N-二甲基氨基、N,N-二乙基氨基、叠氮基、甲酰基、乙酰基、丙酰基、三氟乙酰基、-CH2CO2H、-CH2CH2CO2H、-CH2CN、-CH2CH2CN、氨基甲酰基、N-甲基氨基甲酰基、N,N-二甲基氨基甲酰基、N-乙基氨基甲酰基、N,N-二乙基氨基甲酰基、羧基、-CH2OH、-CH2CH2OH、氧代(=O)、硫代(=S)、磺酰氨基(-SO2NH2)、-SCH3、-SCH2CH3、-SO2CH3、-SO2CF3、磺酸基(-SO2OH)、醛基、-CH2NH2、-CH2CH2NH2、-CH2NHMe、-CH2NMe2、-CH2NHEt、-CH2NEt2、-CH2CH2NHMe、-CH2CH2NHEt、-CH2CH2NMe2、-CH2CH2NEt2、-CH2CONH2、-CH2CONHMe、-CH2CONMe2、-CH2CONHEt、-CH2CONEt2、-CH2CH2CONH2、-CH2CH2CONHMe、-CH2CH2CONMe2、-CH2CH2CONHEt、-CH2CH2CONEt2、苯基、苯氧基、苯磺酰基、-CH2Ph、-CH2CH2Ph、-OCH2Ph、-OCH2CH2Ph、-COPh、-COCH2Ph或-CH2Ph(OMe)2
  7. 权利要求1~6中任一项所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,
    Figure PCTCN2015073854-appb-100010
    Figure PCTCN2015073854-appb-100011
    优选地,
    Figure PCTCN2015073854-appb-100012
  8. 权利要求1~6中任一项所述的杂环化合物、其立体异构体或其药学上可接受的盐,其中,通式(I)表示的杂环化合物选自下列通式所示的化合物:
    Figure PCTCN2015073854-appb-100013
    Figure PCTCN2015073854-appb-100014
    优选地,通式(I)表示的杂环化合物选自如下通式所示的化合物:
    Figure PCTCN2015073854-appb-100015
    其中,R1、R2、R3、L和G环与相应权利要求中的定义相同。
  9. 权利要求1-8中任一项所述的杂环化合物、其立体异构体或其药学上可接受的盐,其选自如下化合物:
    (1)6-氯-5-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (2)3-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (3)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (4)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (4a)(+)-3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (4b)(-)-3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (5)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-7,8-二氢-6H-吡啶并[1,2-a]嘧啶-4,9-二酮;
    (6)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2,9-二甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (7)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (8)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (9)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (10)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)-3,4-二氢喹啉-2(1H)-酮;
    (11)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
    (12)7-(5-(4-(2-氯苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
    (13)7-(5-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
    (14)7-(5-(4-(苯并噻吩-4-基)-5,6-二氢吡啶-1(2H)-基)戊基)喹啉-2(1H)-酮;
    (15)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯二氢吲哚-2-酮;
    (16)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7-二氢吡啶并[1,2-a]嘧啶-4-酮;
    (17)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-(苄氧基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮;
    (18)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮;
    (19)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (20)9-羟基-2-甲基-3-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (21)2-甲基-3-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (22)7-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
    (23)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯-3,4-二氢喹啉-2(1H)-酮;
    (24)6-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)-2-甲基喹唑啉-4(3H)-酮;
    (25)7-(2-(4-(2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (26)1-(苯并噻吩-4-基)-4-(2-(4-甲基噻唑-5-基)乙基)哌嗪;
    (27)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二氢苯并咪唑-2-酮;
    (28)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9,9-二氟-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (29)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基-2(3H)-苯并噻唑酮;
    (30)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基-2H-1,4-苯并噁嗪-3(4H)-酮;
    (31)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基-2H-1,4-苯并噻嗪-3(4H)-酮;
    (32)7-(2-(4-(噻吩并[2,3-d]嘧啶-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (33)7-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)喹啉-2(1H)-酮;
    (34)6-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)喹唑啉-4(3H)-酮;
    (35)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹唑啉-4(3H)-酮;
    (36)3-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (37)5-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-6-氯-二氢吲哚-2-酮;
    (38)4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (39)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (40)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氯喹啉-2(1H)-酮;
    (41)9-羟基-2-甲基-3-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (42)6-氯-5-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (43)7-(2-(4-(噻吩并[2,3-c]吡啶-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (44)7-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)喹啉-2(1H)-酮;
    (45)7-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基-3,4-二氢喹啉-2(1H)-酮;
    (46)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-吲哚;
    (47)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (48)5-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)二氢吲哚-2-酮;
    (49)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-4,5-二氢-1H-苯并氮杂卓-2(3H)-酮;
    (50)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-4H-吡啶并[1,2-a]嘧啶-4-酮;
    (51)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3-甲基-3,4-二氢喹唑啉-2(1H)-酮;
    (52)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-9-氯-2-甲基-6,7,8,9-四氢吡啶并[1,2-a]嘧啶-4-酮;
    (53)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-硫酮;
    (54)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-硫酮;
    (55)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5,6-二乙基嘧啶-4(3H)-酮;
    (56)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)嘧啶-4(3H)-酮;
    (57)7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (58)2-((4-(苯并噻吩-4-基)哌嗪-1-基)甲基)-1H-苯并咪唑;
    (59)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑;
    (60)7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
    (61)N-(7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (62)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
    (63)N-(5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (64)7-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (65)6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (66)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-7-氟-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (67)7-(2-(4-(3-甲基苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (68)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噁唑-2(3H)-酮;
    (69)4-((4-(苯并噻吩-4-基)哌嗪-1-基)甲基)喹啉-2(1H)-酮;
    (70)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-1H-吲哚-5-甲腈;
    (71)7-(5-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)戊基)喹啉-2(1H)-酮;
    (72)6-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-8-氟-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (73)1-(苯并噻吩-4-基)-4-((2,3-二氢苯并[1,4]二氧杂环己烯-2-基)甲基)哌嗪;
    (74)6-(4-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
    (75)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-苯并咪唑;
    (76)N-(6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (77)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-苯并咪唑-2(3H)-酮;
    (78)6-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (79)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (80)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基喹唑啉-4(3H)-酮;
    (81)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,1-二氧代-3,4-二氢-2H-苯并[1,2]噻嗪;
    (82)5-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)二氢吲哚-2-酮;
    (83)7-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (84)5-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (85)7-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (86)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-甲基-1H-苯并咪唑-2(3H)-酮;
    (87)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二甲基-1H-苯并咪唑-2(3H)-酮;
    (88)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-甲基-1H-苯并咪唑-2(3H)-酮;
    (89)6-(2-(4-(2-甲氧基苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (90)3-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-4-酮;
    (91)7-(2-(4-(2-氧代-2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (92)6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (93)5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-1,3-二甲基-1H-苯并咪唑-2(3H)-酮;
    (94)6-氟-5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (95)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-6-氟二氢吲哚-2-酮;
    (96)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-苄基-3-甲基喹唑啉-2,4(1H,3H)-二酮;
    (97)6-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (98)6-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (99)3-(2-(4-(苯并噻吩-4-基)哌啶-1-基)乙基)-2-甲基-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-4-酮;
    (100)6-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (101)5-(4-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)丁基)二氢吲哚-2-酮;
    (102)7-(2-(4-(6-氟苯并噻吩-4-基)哌嗪-1-基)乙基)喹啉-2(1H)-酮;
    (103)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-6,7-二甲氧基-4H-色烯-4-酮;
    (104)6-(4-(4-(苯并噻吩-4-基)哌啶-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
    (105)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5-甲氧基-1H-吲哚;
    (106)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-5-甲氧基-1H-吲哚;
    (107)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-5-甲氧基-1H-吲哚;
    (108)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-1H-吲哚-5-甲腈;
    (109)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1H-吲哚-5-甲腈;
    (110)1-乙酰基-3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-1H-吲哚-5-甲腈;
    (111)6-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-3,4-二氢喹啉-2(1H)-酮;
    (112)5-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)二氢吲哚-2-酮;
    (113)6-氯-5-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-2-酮;
    (114)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-4-甲基-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (115)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-甲基-3,4-二氢喹啉-2(1H)-酮;
    (116)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
    (117)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
    (118)N-(5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (119)N-(7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (120)4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺;
    (121)N-(4-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (122)7-(2-(4-(2-甲基苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (123)3-((4-(苯并噻吩-4-基)哌嗪-1-基)甲基)-1-甲基-1H-吲哚;
    (124)1-(3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)二氢吲哚-1-基)乙酮;
    (125)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-1-对甲苯磺酰基-1H-吲哚-5-甲腈;
    (126)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-1-对甲苯磺酰基-1H-吲哚-5-甲腈;
    (127)3-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-5-甲氧基-1-对甲苯磺酰基-1H-吲哚;
    (128)3-(3-(4-(苯并噻吩-4-基)哌嗪-1-基)丙基)-5-甲氧基-1-对甲苯磺酰基-1H-吲哚;
    (129)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-5-甲氧基-1-对甲苯磺酰基-1H-吲哚;
    (130)6-(2-(4-(2-氧代-2,3-二氢苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-酮;
    (131)3-(2-(4-(2-氟苯并噻吩-4-基)哌嗪-1-基)乙基)-9-羟基-2-甲基-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-4-酮;
    (132)2-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (133)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-3,4-二氢喹啉-2(1H)-硫酮;
    (134)(3aR,4R,6aS)-4-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊基)四氢-1H-噻吩并[3,4-d]咪唑-2(3H)-酮;
    (135)(6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)氨基甲酸戊酯;
    (136)3-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-2-甲基-4-氧代-6,7,8,9-四氢-4H-吡啶并[1,2-a]嘧啶-9-基苯甲酸酯;
    (137)6-(4-(4-(苯并噻吩-4-基)哌嗪-1-基)丁基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (138)1-(苯并噻吩-4-基)-4-(4-(1-环己基-1H-四唑-5-基)丁基)哌嗪;
    (139)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)-8-氟-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (140)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟乙基)喹啉-2(1H)-酮;
    (141)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (142)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基乙基)-2H-苯并[1,4]噻嗪-3(4H)-酮;
    (143)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)二氢吲哚-2-酮;
    (144)8-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-甲氧基乙基)-2H-苯并[1,4]噁嗪-3(4H)-酮;
    (145)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基乙基)二氢吲哚-2-酮;
    (146)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)苯并噻唑-2(3H)-酮;
    (147)5-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)-1H-苯并咪唑-2(3H)-酮;
    (148)7-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙酰基)-3,4-二氢喹啉-2(1H)-酮;
    (149)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊酰基)喹啉-2(1H)-酮;
    (150)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)-1-羟基戊基)-3,4-二氢喹啉-2(1H)-酮;
    (151)7-(5-(4-(苯并噻吩-4-基)哌嗪-1-基)戊酰基)-3,4-二氢喹啉-2(1H)-酮;
    (152)N-(6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)环戊甲酰胺;
    (153)N-(6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-基)乙酰胺;
    (154)6-(2-(4-(苯并噻吩-4-基)哌嗪-1-基)乙基)苯并噻唑-2-胺。
  10. 一种药物组合物,其包含治疗有效量的选自权利要求1~9中任一项所述的杂环化合物、其立体异构体或其药学上可接受的盐和任选的可药用载体。
  11. 一种制备药物组合物的方法,包括将权利要求1~9中任一项所述的杂环化合物、其立体异构体或其药学上可接受的盐与可药用载体混合。
  12. 权利要求1~9中任一项所述的杂环化合物、其立体异构体或其药学上可接受的盐在制备预防和/或治疗中枢神经系统疾病的药物中的应用。
  13. 权利要求12所述的应用,其中所述枢神经系统疾病选自:精神分裂症;难控制的、难处理的或慢性精神分裂症;情感紊乱;精神紊乱;情绪紊乱;I型双极情感障碍;II型双极情感障碍;抑郁症;内因性抑郁症;重性抑郁症;难控制的抑郁症;情绪恶劣性障碍;循环情感性障碍;恐慌发作;惊恐性障碍;社交恐惧症;强迫性观念与行为病症;冲动性病症;创伤后精神紧张性障碍;焦虑症;急性应激障碍;癔病;神经性厌食症;睡眠障碍;适应性障碍;认知障碍;自闭症;神经性头痛;狂躁症;帕金森症;亨廷顿舞蹈症;阿尔茨海默症;各种痴呆症;记忆障碍;多动症;注意力缺乏/亢进类疾病和抽动症。
  14. 制备权利要求1~9中任一项所述的的杂环化合物、其立体异构体或其药学上可接受的盐的方法,其可按如下方法1~5中的一种进行:
    方法1:由式(II)所示化合物或其盐与式(III)所示化合物或其盐进行反应,如反应式1所示:
    Figure PCTCN2015073854-appb-100016
    其中,G环、L、A、B、D、E、R1、R2和R3的定义同相应权利要求中的定义相同;
    X代表离去基团,如卤素、C1~C6烷基磺酰氧基、苯磺酰氧基、萘磺酰氧基,上述C1~C6烷基磺酰氧基、苯磺酰氧基、萘磺酰氧基可非必须地被一个或多个选自卤素、C1~C6烷基、C1~C6烷氧基、硝基、羟基、氨基和C1~C6烷酰基的基团进一步取代;
    方法2:式(IV)所示的化合物或其盐与式(V)所示的化合物或其盐进行反应,如反应式2所示:
    Figure PCTCN2015073854-appb-100017
    其中,G环、L、A、B、D、R1、R2和R3的定义与相应权利要求中的定义相同,E1代表氮原子;
    X1代表卤素或三氟甲磺酰氧基;
    方法3:式(VI)所示化合物或其盐与式(III)所示化合物或其盐经酰胺化反应得到式(VII)化合物或其盐;式(VII)化合物或其盐用还原剂处理,得到式(I)化合物,如反应式3所示:
    Figure PCTCN2015073854-appb-100018
    其中,G环、L、A、B、D、E、R1、R2和R3的定义与相应权利要求中的定义相同;
    方法4:式(VIII)所示化合物或其盐与式(III)所示化合物或其盐经还原胺化反应得到式(I)化合物,如反应式4所示:
    Figure PCTCN2015073854-appb-100019
    其中,G环、L、A、B、D、E、R1、R2和R3的定义与相应权利要求中的定义相同;
    方法5:由方法1~4得到的式(I)化合物进行官能团转化得到。
PCT/CN2015/073854 2014-03-07 2015-03-09 一类杂环化合物、其制备方法和用途 WO2015131856A1 (zh)

Priority Applications (8)

Application Number Priority Date Filing Date Title
JP2016556278A JP6395850B2 (ja) 2014-03-07 2015-03-09 複素環式化合物、その製造方法および使用
CA2941771A CA2941771C (en) 2014-03-07 2015-03-09 Heterocyclic compounds, process for preparation of the same and use thereof
RU2016137169A RU2667498C2 (ru) 2014-03-07 2015-03-09 Гетероциклические соединения, способ их получения и их применение
CN201580008897.5A CN106132956B (zh) 2014-03-07 2015-03-09 一类杂环化合物、其制备方法和用途
AU2015226578A AU2015226578B2 (en) 2014-03-07 2015-03-09 Heterocyclic compounds, process for preparation of the same and use thereof
EP15757771.9A EP3115361B1 (en) 2014-03-07 2015-03-09 Heterocyclic compounds, and preparation method and use thereof
KR1020167027945A KR101840249B1 (ko) 2014-03-07 2015-03-09 헤테로고리 화합물 및 이의 제조방법과 용도
US15/124,264 US10174011B2 (en) 2014-03-07 2015-03-09 Heterocyclic compounds, process for preparation of the same and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201410083602 2014-03-07
CN201410083602.8 2014-03-07
CN201410853950.9A CN104892589A (zh) 2014-03-07 2014-12-31 一类杂环化合物、其制备方法和用途
CN201410853950.9 2014-12-31

Publications (1)

Publication Number Publication Date
WO2015131856A1 true WO2015131856A1 (zh) 2015-09-11

Family

ID=54025602

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/073854 WO2015131856A1 (zh) 2014-03-07 2015-03-09 一类杂环化合物、其制备方法和用途

Country Status (9)

Country Link
US (1) US10174011B2 (zh)
EP (1) EP3115361B1 (zh)
JP (1) JP6395850B2 (zh)
KR (1) KR101840249B1 (zh)
CN (2) CN104892589A (zh)
AU (1) AU2015226578B2 (zh)
CA (1) CA2941771C (zh)
RU (1) RU2667498C2 (zh)
WO (1) WO2015131856A1 (zh)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3244898A4 (en) * 2015-01-12 2018-08-22 Reviva Pharmaceuticals, Inc. Methods for treating psychosis associated with parkinson's disease
EP3244897A4 (en) * 2015-01-12 2018-08-22 Reviva Pharmaceuticals, Inc. Methods for treating alzheimer's disease
EP3244896A4 (en) * 2015-01-12 2018-08-29 Reviva Pharmaceuticals, Inc. Methods for treating pulmonary hypertension
JP2018529727A (ja) * 2015-10-02 2018-10-11 クリスタル ファルマ、エセ、ア、ウCrystal Pharma,S.A.U. ベンゾ[b]チオフェン化合物を製造するための方法および中間体
US10100044B2 (en) * 2013-10-18 2018-10-16 Suzhou Vigonvita Life Sciences Co., Ltd. Methods for preparing brexpiprazole, key intermediates thereof and salts thereof
WO2019242717A1 (zh) 2018-06-21 2019-12-26 中国科学院上海药物研究所 一种苯并噻吩化合物的马来酸盐、其结晶形式及其用途
WO2023186023A1 (zh) * 2022-03-30 2023-10-05 苏州旺山旺水生物医药股份有限公司 一类n-取代的喹啉酮化合物、其制备方法和用途

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105461608B (zh) * 2015-11-23 2017-11-28 东南大学 二氢吲哚‑2‑酮类d3受体配体及其制备方法和用途
US10464931B2 (en) 2015-12-28 2019-11-05 Honour (R&D) Process for the preparation of Quinolin-2(1H)-one derivatives
CN106478548A (zh) * 2016-07-19 2017-03-08 贵州大学 一种6‑羟基灭草松的制备工艺
CN107098855A (zh) * 2017-04-05 2017-08-29 上海诺星医药科技有限公司 一种制备7‑羟基‑2‑喹啉酮的方法
CN111004211B (zh) * 2019-12-29 2021-04-02 苏州诚和医药化学有限公司 一种依匹哌唑中间体4-溴苯并[b]噻吩的合成方法
US11851435B2 (en) * 2022-03-07 2023-12-26 National Health Research Institutes PTGR2 inhibitors and their use

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101754964A (zh) * 2007-07-19 2010-06-23 库多斯药物有限公司 Dna pk抑制剂
WO2013024291A2 (en) * 2011-08-18 2013-02-21 Ucb Pharma S.A. Therapeutically active fused pyrimidine derivatives

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4771053A (en) 1987-03-02 1988-09-13 Bristol-Myers Company Method for alleviation of primary depressive disorders
US5824680A (en) 1991-08-31 1998-10-20 Bayer Aktiengesellschaft Ipsapirone for the treatment of alzheimer's disease by improving memory
US5137894A (en) * 1991-12-05 1992-08-11 New James S 4-(4-Piperidinyl-thieno[3,2-c]pyridine derivatives of n-alkylglutarimides
HN1999000146A (es) 1998-09-21 2000-11-11 Pfizer Prod Inc Agentes farmaceuticos para el tratamiento de la enfermedad de parkinson, adhd y microadenomas.
GB2370270A (en) * 2000-12-20 2002-06-26 Lilly Co Eli Pharmaceutical compounds
GB0227240D0 (en) * 2002-11-21 2002-12-31 Glaxo Group Ltd Compounds
UA84566C2 (ru) * 2004-06-09 2008-11-10 Солвей Фармасьютикалс Б.В. Производные тиофенпиримидинонов и их применение в терапии, фармацевтическая композиция на их основе
JP4315393B2 (ja) * 2005-04-14 2009-08-19 大塚製薬株式会社 複素環化合物
TWI320783B (en) * 2005-04-14 2010-02-21 Otsuka Pharma Co Ltd Heterocyclic compound
JP2008037850A (ja) * 2006-08-10 2008-02-21 Mitsubishi Tanabe Pharma Corp 新規置換ピペリジン誘導体
WO2008047883A1 (en) * 2006-10-13 2008-04-24 Otsuka Pharmaceutical Co., Ltd. Piperazine-substituted benzothiophenes for treatment of mental disorders
JP2008115175A (ja) * 2006-10-13 2008-05-22 Otsuka Pharmaceut Co Ltd 複素環化合物
JP4540700B2 (ja) 2006-10-13 2010-09-08 大塚製薬株式会社 医薬
AU2009206580A1 (en) * 2008-01-25 2009-07-30 Merck Sharp & Dohme Corp. Quinolizidinone M1 receptor positive allosteric modulators
WO2010030757A2 (en) 2008-09-10 2010-03-18 Kalypsys Inc. Aminopyrimidine inhibitors of histamine receptors for the treatment of disease
CA2987743A1 (en) * 2009-03-13 2010-09-16 Katholieke Universiteit Leuven, K.U.Leuven R&D Purine derivatives and their use as immunosuppressive agents
CA2773131C (en) * 2009-09-04 2015-07-14 The Regents Of The University Of Michigan Compositions and methods for treatment of leukemia
KR101756495B1 (ko) * 2010-03-11 2017-07-10 다이닛본 스미토모 세이야꾸 가부시끼가이샤 N-아실 시클릭 아민 유도체 또는 이의 의약상 허용되는 염
GB201012889D0 (en) * 2010-08-02 2010-09-15 Univ Leuven Kath Antiviral activity of novel bicyclic heterocycles
JP2014162781A (ja) * 2013-02-27 2014-09-08 Otsuka Pharmaceut Co Ltd 複素環化合物
WO2016003296A1 (en) * 2014-07-04 2016-01-07 Instytut Farmakologii Polskiej Akademii Nauk (quinoline or isoquinoline)sulfonamides of cyclic amines as antipsychotic drugs

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101754964A (zh) * 2007-07-19 2010-06-23 库多斯药物有限公司 Dna pk抑制剂
WO2013024291A2 (en) * 2011-08-18 2013-02-21 Ucb Pharma S.A. Therapeutically active fused pyrimidine derivatives

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10100044B2 (en) * 2013-10-18 2018-10-16 Suzhou Vigonvita Life Sciences Co., Ltd. Methods for preparing brexpiprazole, key intermediates thereof and salts thereof
EP3244898A4 (en) * 2015-01-12 2018-08-22 Reviva Pharmaceuticals, Inc. Methods for treating psychosis associated with parkinson's disease
EP3244897A4 (en) * 2015-01-12 2018-08-22 Reviva Pharmaceuticals, Inc. Methods for treating alzheimer's disease
EP3244896A4 (en) * 2015-01-12 2018-08-29 Reviva Pharmaceuticals, Inc. Methods for treating pulmonary hypertension
US10441590B2 (en) 2015-01-12 2019-10-15 Reviva Pharmaceuticals, Inc. Methods for treating pulmonary hypertension
JP2018529727A (ja) * 2015-10-02 2018-10-11 クリスタル ファルマ、エセ、ア、ウCrystal Pharma,S.A.U. ベンゾ[b]チオフェン化合物を製造するための方法および中間体
WO2019242717A1 (zh) 2018-06-21 2019-12-26 中国科学院上海药物研究所 一种苯并噻吩化合物的马来酸盐、其结晶形式及其用途
US11384073B2 (en) * 2018-06-21 2022-07-12 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Maleate salt of benzothiophene compound, crystalline form thereof, and use thereof
WO2023186023A1 (zh) * 2022-03-30 2023-10-05 苏州旺山旺水生物医药股份有限公司 一类n-取代的喹啉酮化合物、其制备方法和用途

Also Published As

Publication number Publication date
CN104892589A (zh) 2015-09-09
RU2667498C2 (ru) 2018-09-21
CA2941771A1 (en) 2015-09-11
RU2016137169A (ru) 2018-03-21
RU2016137169A3 (zh) 2018-03-21
KR101840249B1 (ko) 2018-03-20
EP3115361B1 (en) 2019-10-23
AU2015226578B2 (en) 2017-11-23
KR20160122269A (ko) 2016-10-21
AU2015226578A1 (en) 2016-10-20
CN106132956B (zh) 2019-12-10
CN106132956A (zh) 2016-11-16
JP2017508756A (ja) 2017-03-30
JP6395850B2 (ja) 2018-09-26
CA2941771C (en) 2020-02-18
EP3115361A1 (en) 2017-01-11
US10174011B2 (en) 2019-01-08
EP3115361A4 (en) 2017-04-26
US20170158680A1 (en) 2017-06-08

Similar Documents

Publication Publication Date Title
CN106132956B (zh) 一类杂环化合物、其制备方法和用途
US10556908B2 (en) Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
CN104470902B (zh) N‑(3‑杂芳基芳基)‑4‑芳基芳基甲酰胺和类似物作为Hedgehog通路抑制剂及其应用
CN101048414B (zh) 用于治疗精神障碍的稠合的三环衍生物
CN105315285B (zh) 2,4‑二取代7H‑吡咯并[2,3‑d]嘧啶衍生物、其制法与医药上的用途
WO2021088945A1 (zh) 作为shp2抑制剂的化合物及其应用
TWI413522B (zh) 苯并二氮呯化合物及藥學組成物
US20120178742A1 (en) Metabotropic glutamate receptor modulators
WO2016007731A1 (en) Imidazopyridines and imidazopyrazines as lsd1 inhibitors
WO2002032872A1 (en) Nitrogenous aromatic ring compounds
TW200843777A (en) Inhibitors of the hedgehog pathway
AU2013339167A1 (en) Novel amine derivative or salt thereof
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
WO2017114512A1 (zh) 含氮稠杂环化合物、制备方法、中间体、组合物和应用
EP3426244B1 (en) 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
JPWO2019208812A1 (ja) ベンゾイソオキサゾール化合物
WO2019034031A1 (zh) 一类四氢原小檗碱类化合物,其制备方法、用途及药物组合物
WO2017101796A1 (zh) 酞嗪酮衍生物、其制备方法及用途
WO2018019252A1 (en) Novel fused pyridine derivatives useful as fak/aurora kinase inhibitors
CN103087050A (zh) 芳基激酶抑制剂
CN107690434B (zh) 作为tnf活性调节剂的稠合三环咪唑并吡嗪衍生物
WO2019076336A1 (zh) 含吡唑基的三并环类衍生物、其制备方法和应用
WO2018228275A1 (zh) 作为mnk抑制剂的杂环化合物
CA2937012A1 (en) Fused pyrazole derivative
TW201607941A (zh) T型鈣通道阻礙劑

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: 15757771

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2016556278

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2941771

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15124264

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2016137169

Country of ref document: RU

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2015757771

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015757771

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20167027945

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015226578

Country of ref document: AU

Date of ref document: 20150309

Kind code of ref document: A