WO2022073470A1 - 杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用 - Google Patents

杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用 Download PDF

Info

Publication number
WO2022073470A1
WO2022073470A1 PCT/CN2021/122546 CN2021122546W WO2022073470A1 WO 2022073470 A1 WO2022073470 A1 WO 2022073470A1 CN 2021122546 W CN2021122546 W CN 2021122546W WO 2022073470 A1 WO2022073470 A1 WO 2022073470A1
Authority
WO
WIPO (PCT)
Prior art keywords
general formula
independently
compound
pharmaceutically acceptable
compound represented
Prior art date
Application number
PCT/CN2021/122546
Other languages
English (en)
French (fr)
Inventor
万泽红
冯加权
吴永奇
秦俊
周炳城
胡志京
姜新剑
冯子晋
Original Assignee
上海枢境生物科技有限公司
江苏恩华药业股份有限公司
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 上海枢境生物科技有限公司, 江苏恩华药业股份有限公司 filed Critical 上海枢境生物科技有限公司
Priority to IL301847A priority Critical patent/IL301847A/en
Priority to JP2023546376A priority patent/JP2023544653A/ja
Priority to BR112023005443A priority patent/BR112023005443A2/pt
Priority to US18/027,226 priority patent/US20230339949A1/en
Priority to KR1020237009354A priority patent/KR20230054417A/ko
Priority to EP21876988.3A priority patent/EP4227306A4/en
Priority to CA3194682A priority patent/CA3194682A1/en
Priority to CN202180068356.7A priority patent/CN116568302A/zh
Priority to MX2023004102A priority patent/MX2023004102A/es
Priority to AU2021356875A priority patent/AU2021356875B2/en
Publication of WO2022073470A1 publication Critical patent/WO2022073470A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • 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
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/16Peri-condensed 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
    • A61K31/495Heterocyclic 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
    • A61K31/4965Non-condensed pyrazines
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs 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
    • A61P25/18Antipsychotics, 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence

Definitions

  • the invention belongs to the field of medicinal chemistry, and in particular relates to a heterocyclic substituted fused ⁇ -carboline derivative, synthesis and application thereof. More specifically, the present invention relates to a heterocyclic substituted fused ⁇ -carboline derivative, a preparation method thereof, an intermediate, a pharmaceutical composition comprising the heterocyclic substituted fused ⁇ -carboline derivative, and Use of the heterocyclic substituted fused ⁇ -carboline derivative and its pharmaceutical composition in the preparation of a medicament for preventing and/or treating neuropsychiatric diseases.
  • Schizophrenia is a disorder characterized by deep cognitive and emotional divisions, manifested in the impact of the most basic human behaviors, such as language, thought, perception, and self-perception.
  • the symptoms of the disease cover a wide range, the most common of which are mental disorders, such as hallucinations, paranoia, and delusions.
  • schizophrenia often leads to comorbidities such as anxiety disorders, depression, or psychotropic substance abuse.
  • Antipsychotics that act pharmacologically by blocking dopamine D2 receptors have traditionally been called first-generation antipsychotics, or "classic” antipsychotics (eg, haloperidol), which treat positive symptoms of schizophrenia including: Breakthrough but failed to treat negative symptoms and cognitive impairment.
  • Typical antipsychotics generally have severe EPS side effects and are ineffective in one-third of patients with schizophrenia.
  • the serotonin system plays an important role in regulating the functions of the prefrontal cortex (PFC), including emotional control, cognitive behavior, and working memory.
  • PFC prefrontal cortex
  • Pyramidal neurons and GABA interneurons of the PFC contain several serotonin receptor subtypes 5-HT1A and 5-HT2A with particularly high densities.
  • PFC and NMDA receptor channels have recently been shown to be targets of 5-HT1AR, and these two receptors modulate excitatory neurons in the cerebral cortex, thereby affecting cognitive function. Indeed, various preclinical data suggest that the 5-HT1AR may be a new target for antipsychotic drug development.
  • atypical antipsychotics such as olanzapine, aripiprazole, etc.
  • 5-HT1AR atypical antipsychotics
  • their low EPS side effects suggest that the serotonin system plays an important role in regulating the functions of the prefrontal cortex (PFC), including mood Control, cognitive behavior, and working memory.
  • Pyramidal neurons and GABA interneurons of the PFC contain several serotonin receptor subtypes 5-HT1A and 5-HT2A with particularly high densities. Recent studies have shown that 5-HT1A agonists are associated with atypical antipsychotic treatment, improving negative symptoms and cognitive impairment.
  • 5-HT2A plays an important role in all aspects of perception, emotion regulation, and motor control. Blocking the 5-HT2A receptor normalizes dopamine release, which acts as an antipsychotic. In addition, 5-HT2C receptors are closely related to weight gain.
  • D3 receptors in the brain are mainly selectively distributed in the limbic system.
  • There are two main DA neural pathways in the brain one is the nigrostriatal pathway to regulate motor function, and the other is the ventral tegmental area of the midbrain.
  • the DA pathway in the septal prefrontal cortex is closely related to learning, cognitive and emotional activities, and its dysfunction will lead to schizophrenia.
  • This DA pathway is also the main pathway for reward effects in the brain.
  • D3R is in the two DA neural pathways Both are distributed and have complex interactions with other DA receptor subtypes, and may be a target for antipsychotic drug therapy.
  • Selective D3 receptor antagonism can reduce negative and cognitive symptoms in schizophrenia.
  • Patent PCT/US2017/015178 discloses Markush formula compounds, which act on receptors 5-HT2A, D2, D1 and SERT (serotonin reuptake transporter) and other receptors, and have potential for schizophrenia and Parkinson's disease therapeutic activity,
  • Refractory schizophrenia refers to a class of patients who are treated according to general methods and cannot obtain ideal curative effects. Such patients have been treated with three antipsychotic drugs with different active ingredients, but the treatment response is poor or unable to be treated. Adverse reactions of antipsychotic drugs are tolerated, or the disease recurs or deteriorates even after adequate maintenance or preventive treatment. Therefore, anti-refractory schizophrenia treatment drugs have always been a difficult problem in current clinical drug research, and it has always been an urgent need to overcome. .
  • the present invention aims to provide a new anti-schizophrenia drug acting on serotonin receptors and/or dopamine receptors, which has good antagonistic activity on 5-HT2A receptors and/or dopamine D2 receptors, and can effectively Treat and improve schizophrenia.
  • the purpose of the present invention is to provide a heterocyclic substituted fused ⁇ -carboline derivative having the activity of treating mental and neurological diseases, its pharmaceutical composition and its application in the medical field.
  • the present invention provides a compound represented by general formula (I), its stereoisomer or its pharmaceutically acceptable salt:
  • R 1 is independently optionally substituted by halogen, C1-C3 alkoxy, cyano, amino, nitro, hydroxy, C2-C4 alkenyl, C2-C4 alkynyl, C3-C5 cycloalkyl
  • R 2 is independently -R 7 -R 8 -R 9 -;
  • R 3 is independently hydrogen, or any one of C1-C3 alkyl groups
  • R 7 is independently a C1-C5 hydrocarbylene
  • R 9 is independently any one of C1-C3 hydrocarbylene or absent
  • A is phenyl optionally substituted with one or more halogens.
  • the R 1 is independently halogen, C1-C3 alkoxy, cyano, amino, nitro, hydroxyl, C2-C4 alkenyl, C2-C4 alkynyl , C3-C5 cycloalkyl optionally substituted C1-C3 alkyl.
  • the R 3 is independently hydrogen, or any one of C1-C3 alkyl groups.
  • R 4 is -CH 2 -.
  • the R 5 and R 6 are each independently hydrogen.
  • R 4 is -CH 2 -, and R 5 and R 6 are each independently hydrogen.
  • A is independently phenyl substituted with one or more halogens.
  • the R 1 is independently an unsubstituted C1-C3 alkyl group.
  • the R 3 is independently hydrogen, or any one of unsubstituted C1-C3 alkyl groups.
  • the C1-C3 alkyl group in the optionally substituted C1-C3 alkyl group is selected from any one of methyl, ethyl or propyl.
  • the unsubstituted C1-C3 alkyl group is selected from any one of methyl, ethyl or propyl.
  • the C1-C5 hydrocarbylene group is selected from the C3-C5 hydrocarbylene group.
  • the C1-C3 hydrocarbylene group is selected from the C1-C2 hydrocarbylene group.
  • the halogen is any one of fluorine, chlorine, bromine or iodine.
  • the compound represented by the general formula (I) is selected from the compounds represented by the general formula (I-A) shown below:
  • X is halogen; the X is substituted at any substitutable position of the benzene ring; X is any one of mono-substitution or polysubstitution, preferably mono-substitution;
  • R 1 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 and R 9 are as defined above.
  • the halogen is any one of fluorine, chlorine, bromine or iodine, preferably fluorine.
  • the compound represented by the general formula (I) is selected from the compounds represented by the general formula (I-B) shown below:
  • R 1 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and X are as defined above.
  • the compound represented by the general formula (I) is selected from the compounds represented by the general formula (I-B) shown below:
  • R 1 is independently any one of methyl, ethyl or propyl
  • R is independently any one of hydrogen, methyl, ethyl, or propyl
  • R 7 is independently any one of -CH 2 -CH 2 -, -CH 2 -CH 2 -CH 2 - or -CH 2 -CH 2 -CH 2 -CH 2 -;
  • R 9 is independently any one of -CH 2 -CH 2 -, -CH 2 - or absent;
  • X is independently any one of fluorine or chlorine.
  • the C2-C4 alkynyl group includes but is not limited to -C ⁇ C, -CH 2 -C ⁇ C, -C ⁇ C-CH 2 , -C ⁇ C-CH 2 - CH 2 , -CH 2 -C ⁇ C-CH 2 , -CH 2 -CH-C ⁇ C, etc.
  • the C2-C4 alkynyl group is preferably a C2-C3 alkynyl group, selected from any of -C ⁇ C, -CH 2 -C ⁇ C or -C ⁇ C-CH 2 A sort of.
  • the C1-C3 alkoxy group is selected from -O-CH 3 , -O-CH 2 -CH 3 , -O-CH 2 -CH 2 -CH 3 , -O- Any one of CH(CH 3 )-CH 3 , preferably any one of -O-CH 3 or -O-CH 2 -CH 3 .
  • the hydrocarbylene group is a straight-chain saturated aliphatic hydrocarbon group having 1-20 carbon atoms
  • the C1-C5 hydrocarbylene group of the present invention is -CH 2 -, -CH 2 -CH2- , -CH2 - CH2 -CH2-, -CH2 - CH2 - CH2 , -CH2 - CH2 - CH2 - CH2 , -CH2 - CH2 - CH2 - CH2 - CH2- or -CH2- Any one of CH 2 -CH 2 -CH 2 -CH 2 -;
  • the C1-C3 alkylene group is -CH 2 -, -CH 2 -CH 2 - or -CH 2 -CH 2 -CH 2 Any one of -;
  • the C1-C2 hydrocarbylene group is any one of -CH 2 - or -CH 2 -CH 2 -CH 2 -.
  • the compound represented by the general formula (I) is selected from the compounds represented by the general formula (I-C) shown below:
  • R 1 is independently any one of methyl, ethyl or propyl
  • R is independently any one of methyl, ethyl or propyl
  • the compound represented by the general formula (I), its stereoisomer or its pharmaceutically acceptable salt is selected from any one of the following specific compounds:
  • the pharmaceutically acceptable salt of any of the above-mentioned compounds represented by general formula (I) is selected from fumarate, maleate, and phosphate, Nitrates, sulfates, besylate, or oxalates.
  • the present invention also provides a compound represented by general formula (I-D), its stereoisomer or its pharmaceutically acceptable salt:
  • R 1 , R 3 , R 4 , R 5 and R 6 are as defined above.
  • R 1 is independently any one of methyl, ethyl or propyl
  • R is independently any one of methyl, ethyl or propyl
  • the present invention provides a method for preparing a compound represented by general formula (I), general formula (I-A), general formula (I-B) or general formula (I-C), its stereoisomer or its pharmaceutically acceptable salt, It uses a compound represented by general formula (I-D) as a starting material or an intermediate; it may include the following steps:
  • the compound of formula (I) is prepared by nucleophilic substitution reaction between compound of general formula (I-D) and compound of general formula (I-E);
  • X 1 is halogen, selected from fluorine, chlorine, bromine or iodine, preferably chlorine;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 and A are as defined above.
  • the method for preparing the compound represented by the general formula (I-C), its stereoisomer or its pharmaceutically acceptable salt comprises the following steps:
  • the compound of formula (I-C) is prepared by nucleophilic substitution reaction between compound of general formula (I-D) and compound of general formula (I-E');
  • X 1 is halogen, selected from fluorine, chlorine, bromine or iodine, preferably chlorine;
  • R 1 , R 3 , R 4 , R 5 and R 6 are as defined above.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the compound represented by the general formula (I), its stereoisomer or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable amount thereof.
  • the pharmaceutical composition may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers.
  • the active compounds of the present invention may be formulated for oral, buccal, intranasal, parenteral (eg intravenous, intramuscular or subcutaneous) or rectal administration, or suitable for administration by inhalation or insufflation
  • the dosage form of the medicine may also be formulated in sustained release dosage forms.
  • an effective dose of a compound of the present invention or a pharmaceutically acceptable salt thereof may be administered orally together with, for example, an inert diluent or some carrier.
  • the compounds of the present invention may be encapsulated in gelatin capsules or compressed into tablets.
  • the compounds of the present invention can be used with excipients and in the form of tablets, troches, capsules, suspensions, syrups and the like.
  • the above formulations should contain at least 0.5% (w/w) of the active compound of the invention, but may vary depending on the particular dosage form, where from 4% to about 70% by weight per unit is convenient. The amount of active compound in such pharmaceutical compositions should result in an appropriate dosage.
  • the active compound of the present invention can be formulated into tablets or capsules by conventional means, for example, with pharmaceutically acceptable excipients, such as binders, fillers , lubricants, disintegrants or wetting agents. Tablets can be coated by methods well known in the art.
  • Liquid preparations for oral administration such as solutions, syrups or suspensions, or volatilized as dry products, can be reconstituted with water or other suitable carriers before use. Such liquid preparations can be prepared by conventional means using pharmaceutically acceptable additives such as suspending agents, emulsifying agents, non-aqueous carriers and preservatives.
  • the compounds provided herein can be combined with sterile aqueous or organic vehicles to form injectable solutions or suspensions.
  • the active compounds of the present invention may be formulated in rectal compositions such as suppositories or retention enemas, eg, containing conventional suppository bases such as cocoa butter or other glycerides.
  • the present invention also provides a compound represented by the general formula (I), its stereoisomer or its pharmaceutically acceptable salt or its pharmaceutical composition in the preparation involving or regulating serotonin receptor, serotonin transporter (SERT) and/or the purposes in the medicine of the dopamine receptor, preferably in the preparation of the medicine involving or regulating the 5-HT2A receptor, the serotonin transporter, the dopamine D1 receptor and/or the dopamine D2 receptor in the medicine, More preferred is the use in the manufacture of a medicament involving or modulating 5-HT2A receptors and/or dopamine D2 receptors. wherein the medicament optionally comprises another one or more active agents that modulate the nervous system of the mammal or alleviate psychiatric disorders.
  • SERT serotonin transporter
  • the modulation includes, but is not limited to, inhibitory activity or antagonistic activity on the receptor.
  • the present invention also provides the use of a compound represented by general formula (I), a stereoisomer or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of neuropsychiatric diseases .
  • the neuropsychiatric disease is selected from depression (such as major depressive disorder (MDD)), anxiety disorders, dementia, schizophrenia, sleep disorders, movement disorders, dementia behavioral disorders, Parkinson's disease, Alzheimer's disease, migraine, ADHD (eg ADHD), obsessive-compulsive disorder, social phobia, neurodegenerative disorders, bipolar disorder, post-traumatic stress syndrome one or more of symptoms, addictive disorders, withdrawal syndrome, or attention deficit.
  • depression such as major depressive disorder (MDD)
  • anxiety disorders dementia
  • schizophrenia sleep disorders
  • movement disorders dementia behavioral disorders
  • Parkinson's disease dementia behavioral disorders
  • Parkinson's disease Alzheimer's disease
  • Alzheimer's disease migraine
  • ADHD eg ADHD
  • obsessive-compulsive disorder e.g., obsessive-compulsive disorder
  • social phobia e.g., phobia
  • neurodegenerative disorders e.g., bipolar disorder, post-traumatic stress syndrome one or more of symptoms, addictive disorders, withdrawal syndrome, or attention deficit.
  • the neuropsychiatric diseases described in the present invention are preferably depression (such as major depressive disorder MDD), anxiety disorders, dementia, schizophrenia, sleep disorders, movement disorders, and behavior of patients with dementia Any one or more of a disorder, neurodegenerative disease or bipolar disorder, more preferably one or more of depression, anxiety, schizophrenia or neurodegenerative disease.
  • depression such as major depressive disorder MDD
  • anxiety disorders such as depression, dementia, schizophrenia, sleep disorders, movement disorders, and behavior of patients with dementia
  • a disorder, neurodegenerative disease or bipolar disorder more preferably one or more of depression, anxiety, schizophrenia or neurodegenerative disease.
  • the neuropsychiatric disease is preferably schizophrenia.
  • the present invention further relates to a method of treating, alleviating and/or preventing diseases involving serotonin receptors, serotonin transporters (SERTs) and/or dopamine receptors, the method comprising administering to a patient in need thereof a therapeutically effective Dosage of the compound of the present invention represented by the aforementioned general formula or a pharmaceutically acceptable salt thereof; preferably relates to a treatment, remission or prevention involving 5-HT2A receptor, serotonin transporter, dopamine D1 receptor and/or dopamine A method for a disease of the D2 receptor, the method comprising administering to a patient in need thereof a therapeutically effective dose of a compound represented by the aforementioned general formula of the present invention or a pharmaceutically acceptable salt thereof.
  • the serotonin receptor is preferably a 5-HT2A receptor
  • the dopamine receptor is preferably a dopamine D2 receptor. This method shows outstanding efficacy and fewer side effects.
  • the present invention relates to a method of treating, alleviating and/or preventing neuropsychiatric diseases, the method comprising administering to a patient in need thereof a therapeutically effective dose of the aforementioned general formula of the present invention A compound or a pharmaceutically acceptable salt thereof.
  • the neuropsychiatric disease is selected from depression (such as major depressive disorder), anxiety disorders, dementia, schizophrenia, sleep disorders, movement disorders, behavioral disorders in patients with dementia , Parkinson's disease, Alzheimer's disease, migraine, ADHD (eg ADHD), obsessive-compulsive disorder, social phobia, neurodegenerative disorders, bipolar disorder, post-traumatic stress syndrome, addiction
  • depression eg major depressive disorder
  • anxiety disorder dementia
  • schizophrenia sleep disorders
  • movement disorders behavioral disorders in patients with dementia
  • Parkinson's disease Alzheimer's disease
  • ADHD eg ADHD
  • obsessive-compulsive disorder e.g., obsessive-compulsive disorder
  • social phobia e.g., social phobia
  • neurodegenerative disorders e.g., a a a disorder, neurodegenerative disease, or bipolar disorder.
  • compounds of the present invention may be optionally substituted with one or more substituents, such as compounds of the general formula above or as specific examples, subclasses in the Examples.
  • substituents such as compounds of the general formula above or as specific examples, subclasses in the Examples.
  • substituents such as compounds of the general formula above or as specific examples, subclasses in the Examples.
  • substituents such as compounds of the general formula above or as specific examples, subclasses in the Examples.
  • substituents such as compounds of the general formula above or as specific examples, subclasses in the Examples.
  • variable eg R
  • labeled variables eg R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , etc.
  • pharmaceutically acceptable refers to a substance which, within the scope of normal medical judgment, is suitable for use in contact with a patient's tissue without undue toxicity, irritation, allergic reaction, etc., with a reasonable benefit to harm ratio, and can be effectively used for its intended purpose.
  • pharmaceutically acceptable salts refers to salts of the compounds of the present invention which are safe and effective when used in mammals and possess the desired biological activity.
  • pharmaceutically acceptable carrier refers to those substances which are not appreciably irritating to the organism and which do not impair the biological activity and properties of the active compound.
  • “Pharmaceutically acceptable carrier” includes, but is not limited to, glidants, sweeteners, diluents, preservatives, dyes/colorants, flavoring agents, surfactants, wetting agents, dispersing agents, disintegrating agents, Stabilizers, Solvents or Emulsifiers.
  • administration refers to methods by which a compound or composition can be delivered to the desired site of biological action. These methods include, but are not limited to, oral or parenteral (including intracerebroventricular, intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular injection or infusion), topical, rectal administration, and the like. Especially by injection or by mouth.
  • treating includes alleviating, alleviating or ameliorating a disease or symptom, preventing other symptoms, ameliorating or preventing the underlying metabolic factors of the symptom, inhibiting the disease or symptom, eg, preventing the progression of the disease or symptom, alleviating the disease or symptom, promoting the Relief of a disease or symptom, or cessation of the symptoms of a disease or symptom, and by extension to include prevention.
  • Treatment also includes achieving a therapeutic benefit and/or a prophylactic benefit. Therapeutic benefit refers to eradication or amelioration of the condition being treated.
  • therapeutic benefit is achieved by eradicating or ameliorating one or more physiological signs associated with the underlying disease, although the patient may still have the underlying disease, but improvement in the patient's disease may be observed.
  • a prophylactic benefit refers to a patient's use of a composition to prevent a risk of a disease, or when a patient develops one or more physiological conditions of the disease, even though the disease has not been diagnosed.
  • active ingredient refers to a chemical entity that is effective in treating or preventing a target disorder, disease, or condition.
  • neuropsychiatric diseases refers to the general term of neurological diseases and mental diseases, including neurological diseases and/or mental diseases.
  • an effective amount refers to a sufficient amount of the drug or medicament to achieve the desired effect with acceptable side effects.
  • the determination of the effective amount varies from person to person, depends on the age and general condition of the individual, and also depends on the specific active substance, and the appropriate effective amount in individual cases can be determined by those skilled in the art based on routine experiments.
  • an “individual” as used herein includes a human or non-human animal.
  • exemplary human subjects include human subjects (referred to as patients) or normal subjects with a disease (eg, a disease described herein).
  • Non-human animals in the present invention include all vertebrates such as non-mammals (eg birds, amphibians, reptiles) and mammals such as non-human primates, livestock and/or domesticated animals (eg sheep, dogs) , cats, cows, pigs, etc.).
  • C1-C6 alkyl specifically refers to the independently disclosed methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl and C6 alkyl groups.
  • alkyl groups include, but are not limited to, methyl (Me, -CH3 ), ethyl (Et, -CH2CH3), n - propyl (n - Pr, -CH2CH2CH3 ) , isopropyl Propyl (i-Pr, -CH( CH3 ) 2 ), n-butyl (n - Bu, -CH2CH2CH2CH3 ), isobutyl (i - Bu, -CH2CH ( CH3 ) ) 2 ), sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu, -C(CH 3 ) 3 ), n-pentyl (-CH 2 CH 2 ) CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl (-CH(CH 2 CH 3 ) 2 ), 2-methyl-2-
  • hydrox (H) refers to a single hydrogen atom. Such radicals may be attached to other groups, such as oxygen atoms, to form hydroxyl groups.
  • halogen refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
  • aryl refers to monocyclic, bicyclic and tricyclic carbocyclic ring systems containing 6-14 ring atoms, or 6-12 ring atoms, or 6-10 ring atoms, wherein at least one ring is aromatic .
  • the aryl group is usually, but not necessarily, attached to the parent molecule through the aromatic ring of the aryl group. Examples of aryl groups may include phenyl, naphthyl, and anthracene.
  • the aryl group is optionally substituted with one or more substituents described herein.
  • alkoxy groups include, but are not limited to, methoxy (MeO, -OCH 3 ), ethoxy (EtO, -OCH 2 CH 3 ), 1-propoxy (n-PrO, n-propoxy group, -OCH 2 CH 2 CH 3 ), 2-propoxy (i-PrO, i-propoxy, -OCH(CH 3 ) 2 ), 1-butoxy (n-BuO, n-butoxy group, -OCH 2 CH 2 CH 2 CH 3 ), 2-methyl-1-propoxy (i-BuO, i-butoxy, -OCH 2 CH(CH 3 ) 2 ), 2-butoxy (s-BuO, s-butoxy, -OCH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC(CH 3 ) 3 ), 1-pentyloxy (n-pentyloxy, -OCH 2 CH 2 CH 2 CH 2 CH 3 ), 2-pent
  • the compound provided by the present invention is an antagonist acting on 5-HT 2A receptor and/or D 2 receptor, and has good effect on 5-HT 2A receptor and/or D2 (D 2L , D 2s ) receptor Antagonism; and/or high D2/5-HT2A ratio, good selectivity; and/or good pharmacokinetic properties; and/or good in vivo pharmacodynamic effect; and/or not only It is as effective as traditional antipsychotics for positive symptoms, and has stronger ameliorating effects on negative symptoms and cognitive deficit symptoms, with fewer side effects (such as reducing the possibility of inducing EPS responses, etc.).
  • Example 1-A (6bR,10aS)-8-(4-(4-fluorophenyl)-4-oxobutyl)-3,6b-dimethyl-6b,7,8,9,10 Preparation of ,10a-hexahydro-1H-pyridine[3',4':4,5]pyrro[1,2,3-de]quinoxalin-2(3H)-one (Compound 1-A)
  • Step 2 3-ethyl-6b-methyl-6b,7,8,9,10,10a-hexahydro-1H-pyridine[3',4':4,5]pyrrole[1,2,3 Preparation of -De]quinoxalin-2(3H)-one (Intermediate 3-2)
  • the fourth step 8-(4-(4-fluorophenyl)-4-oxobutyl)-1,1,3,6b-tetramethyl-6b,7,8,9,10,10a-hexa Hydro-1H-pyridin[3',4':4,5]pyrrole[1,2,3-de]quinoxalin-2(3H)-one (Compound 4)
  • the third step 8-(4-(4-Fluorophenyl)-4-oxobutyl)-3,6b-dimethyl-6b, 7,8,9,10,10a-hexahydro-1H-
  • pyridine[3',4':4,5]pyrrole[1,2,3-de]quinoxaline-1,2(3H)-dione (Compound 5)
  • intermediate 1-8 150 mg, 0.75 mmol
  • potassium carbonate 37 mg, 0.27 mmol
  • LCMS m/z(M+H) + 436.2.
  • Intermediate D1-2 (1 g, 31 mmol) was dissolved in trifluoroacetic acid (10 mL), sodium cyanoborohydride (388 mg, 6.2 mmol) was added in an ice bath, and the reaction was carried out in an ice bath for 2 hours. Pour into ice water (50 mL), adjust pH to 7, extract with ethyl acetate (15 mL), dry over anhydrous sodium sulfate, and concentrate to dryness under reduced pressure to obtain intermediate D1-3, 700 mg, yield: 70%.
  • the fifth step 8-(4-(4-fluorophenyl)-4-carbonylbutyl)-3-methyl-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3 Synthesis of ',4':4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one
  • ITI-007 of the present invention is as follows, prepared with reference to the method of Example 261 of Patent PCT/US2000/016498,
  • Test Example 1 Functional activity test of each compound on receptors (cryopreserved cells)
  • test EC80 Dopamine and Serotonin were diluted 3-fold in compound plates (Greiner- 781280 ) using assay buffer for 10 concentration points starting at 1.2 ⁇ M, 30 ⁇ L per well, The DMSO concentration is not higher than 3%.
  • Antagonist-positive compound and test compound plate preparation The test compound was diluted 3-fold at 10 concentration points in the compound plate (Greiner-781280) using experimental buffer, the initial concentration was 3 ⁇ M, and each well was 30 ⁇ L; antagonist positive Compounds Spiperone and Ketanserin were diluted 3-fold at 10 concentration points using assay buffer, starting at 1 ⁇ M and 30 ⁇ L per well. The DMSO concentration is not higher than 3%.
  • the compounds of the present invention have good functional activity on 5-HT2A and/or D2, and the ratio of D2/5-HT2A is high, suggesting that the compounds of the present invention have a good therapeutic effect on schizophrenia, Reduce the possibility of inducing EPS.
  • the antagonism of the compounds of the present invention to 5-HT 2A receptor was detected by the calcium ion influx assay method at the cellular level.
  • Test compound compound of the example of the present invention, self-made.
  • Control compounds Serotonin hydrochloride serotonin (Sigma, H9523), Ketanserin (Targetmol, T1066).
  • test compounds The compounds of the examples of the present invention were respectively dissolved in DMSO to prepare a 10 mM mother solution, which was stored in a nitrogen cabinet for future use.
  • Experimental buffer HBSS Mix HBSS and HEPES at a ratio of 50:1, store in a refrigerator at 4°C, and heat at 37°C for use.
  • the cell suspension was transferred to a 15 mL centrifuge tube of 4 mL of preheated plating medium, and centrifuged at 1000 rpm for 5 minutes.
  • the cells were diluted to 1 ⁇ 10 6 cells/mL, and 20 ⁇ L of cell suspension was added to each well of the cell plate (Greiner-781946) so that the density of each well was 2 ⁇ 10 4 cells/well.
  • the cell plate was placed in an incubator with 5% CO 2 and 37° C. for 16-24 hours.
  • test EC80 10 concentration point 4-fold dilutions of agonists in compound plates (Greiner- 781280 ) using assay buffer, 30 ⁇ L per well.
  • test compound and positive compound plate Both the test compound and positive compound were diluted in compound plate (Greiner-781280) at 10 concentration points and 4-fold dilution, each well was 30 ⁇ L.
  • Example number 5-HT 2A IC 50 (nM) Example 1 3.72
  • Example 1-B 107.60
  • Example 4 19.69
  • Example 7 3.50 Comparative
  • Example 1 44.36 Comparative Example 1-A 9.11 ITI-007 3.35
  • the compound of the present invention has a good antagonistic effect on 5-HT 2A receptor.
  • Test Example 3 Functional activity test of the compounds of the present invention on D 2L and D 2s receptors
  • the antagonism of the compounds of the present invention to D 2L and D 2s receptors was detected by the detection method of cyclic adenosine monophosphate (cAMP) at the cellular level.
  • cAMP cyclic adenosine monophosphate
  • Test compound compound of the example of the present invention, self-made.
  • test compounds The compounds of the examples of the present invention were respectively dissolved in DMSO to prepare a 10 mM mother solution, which was stored in a nitrogen cabinet for future use.
  • IBMX was dissolved in DMSO to prepare a 0.5M stock solution and stored at -80°C.
  • Experimental buffer dilute 5 ⁇ stimulation buffer to 1 ⁇ with ddH 2 O, add IBMX with a final concentration of 0.5 mM, and mix well for use.
  • test compound preparation The test compound and the positive control compound (+)-Butaclamol hydrochloride were serially diluted in the compound plate (Greiner-781280) using Bravo. The initial concentration of the working solution was 20 ⁇ M, and the experimental buffer was used to press 4 times Dilute to 10 concentrations and centrifuge at 1000 rpm for 1 minute.
  • Cultured D 2S cells after aspirating the medium, wash the cells with 3 mL of PBS, exhaust them, digest with 0.05% Trypsin for 3 minutes, stop with an equal volume of medium, transfer to a centrifuge tube, and centrifuge at 750 rpm for 5 minutes. After draining the liquid, add HBSS-containing buffer to resuspend, and centrifuge at 750 rpm for 5 minutes.
  • the cell plate was centrifuged at 1000 rpm for 1 minute, then sealed, and incubated at room temperature for 45 minutes.
  • mice Male ICR mice were given oral gavage, the blood concentration of the compound of the present invention in the mice was determined, the PK parameters were calculated, and the pharmacokinetics of the compound of the present invention was evaluated.
  • Test drug Compounds of the examples of the present invention, self-made.
  • Drug preparation take the test compound, add normal saline and perform ultrasound.
  • Administration route and dosage oral gavage administration, administration dosage: 5 mg/kg, administration volume: 10 mL/kg.
  • Dosing frequency and duration single dose.
  • the ICR mice were stratified by body weight and then randomly divided into groups of 3 mice, which were fasted overnight before the experiment. Oral gavage, respectively, at 0, 0.033, 0.083, 0.5, 1, 2, 4, 6 and 8 hours, 250 ⁇ L of blood was collected from the mandibular vein or saphenous vein of mice into a sample tube containing anticoagulant heparin sodium. Placed in wet ice, centrifuged at 4000 r ⁇ min -1 for 10 min, separated plasma, and stored in -80°C freezer for testing.
  • the plasma concentration-time data of the compounds of the present invention measured by gavage were substituted into the Winnonlin 8.2 program to calculate the main pharmacokinetic parameters.
  • the measured values of T max and C max were used, AUC 0-t value and AUC 0- ⁇ value were calculated by trapezoidal method, and t 1/2 was calculated from the concentration point at the end of the elimination phase by semi-logarithmic plotting method. The specific results are shown in Table 4.
  • Test Example 5 Test of the effect of the compounds of the present invention on the hyperactivity behavior of mice induced by MK-801
  • the efficacy of the compounds of the present invention was evaluated by intraperitoneal injection of MK-801 in a mouse hyperactivity model.
  • Test compound compound of the example of the present invention, self-made.
  • MK-801 didrozepine maleate, (+)-MK-801 hydrogen maleate
  • SIGMA M107-250MG.
  • Solvent pure water, Guangzhou Watsons Food and Beverage Co., Ltd., 20200928C;
  • mice ICR mice, male, 8 mice/group, Speifu (Beijing) Biotechnology Co., Ltd.
  • Drug preparation take the test compound, add pure water and perform sonication.
  • Administration route and method gavage administration, 10mL/kg body weight.
  • Dosing frequency and duration single dose.
  • the animals were randomly divided into blank group, model group, and administration group after being stratified by body weight.
  • the detailed administration information is shown in the following table:
  • mice were randomly divided into model group, blank group and each administration group after being stratified according to body weight. 1h after the vehicle or drug was administered by gavage, 0.3 mg/kg of MK-801 was intraperitoneally injected (the blank group was injected with an equal volume of normal saline), and then the mice were placed in an autonomous activity box (29cm ⁇ 29cm ⁇ 30cm black polyethylene box) for video recording, the video recording time was 60 min, and video analysis was performed at the end of the video recording to evaluate the activity of the mice.
  • an autonomous activity box 29cm ⁇ 29cm ⁇ 30cm black polyethylene box
  • ED 50 is the half effective dose
  • MED is the lowest effective dose
  • the compounds of the present invention can significantly inhibit the hyperactivity induced by MK-801, and compared with the comparative example, the compounds of the present invention have a lower minimum effective dose and stronger inhibitory effect.
  • Test Example 6 Test of the effect of the compounds of the present invention on DOI-induced head shaking behavior in mice
  • the efficacy of the compounds of the present invention was evaluated by inducing head shaking behavior in mice by intraperitoneal injection of ( ⁇ ) DOI (a hallucinogen commonly used to replicate anti-schizophrenia animal models).
  • Test compound compound of the example of the present invention, self-made.
  • Solvent pure water, Guangzhou Watsons Food and Beverage Co., Ltd., 20200928C;
  • mice ICR mice, male, 8 mice/group, Speifu (Beijing) Biotechnology Co., Ltd.
  • Drug preparation Take the test compound, add the vehicle and sonicate.
  • Administration route and method oral gavage administration, 10mL/kg body weight.
  • Dosing frequency and duration single dose.
  • the animals were randomly divided into blank group, model group, and administration group after being stratified by body weight.
  • the detailed administration information is shown in the following table:
  • mice were randomly divided into model group, blank group and each administration group after being stratified according to body weight. 1h after the animals were given the vehicle or drug by gavage, the animals were placed in a beaker (13 cm in diameter, 19 cm in height) with fresh bedding, and the DOI of the modeling drug was injected intraperitoneally at a dose of 1 mg/kg, and the DOI injected by the mice intraperitoneally was recorded. The number of head shakes in the last 0-20 minutes.
  • Head shaking is defined as a rapid rotational twitch or wet dog shaking of the mouse's head, which is distinct from normal grooming or exploratory behavior.
  • ED 50 is half the effective dose.

Landscapes

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

Abstract

涉及一种杂环取代的稠合γ-咔啉类衍生物、及其制备方法、中间体及其应用,其具有如式(I)所示化合物结构,该类化合物可用于制备治疗神经精神疾病的药物。

Description

杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用
本申请要求申请日为2020/10/9的中国专利申请202011075840.6的优先权,要求申请日为2020/10/9的中国专利申请202011074344.9的优先权。本申请引用上述中国专利申请的全文。
技术领域
本发明属于药物化学领域,具体涉及一种杂环取代的稠合γ-咔啉类衍生物、合成及其应用。更具体地,本发明涉及杂环取代的稠合γ-咔啉类衍生物、及其制备方法、中间体、包含该杂环取代的稠合γ-咔啉类衍生物的药物组合物,以及该杂环取代的稠合γ-咔啉类衍生物和其药物组合物在制备预防和/或治疗神经精神类疾病药物中的用途。
背景技术
精神分裂症是以认知力和情感深度分裂为特征的一种疾病,表现为最基本的人类行为受到影响,例如语言、思想、知觉和自我感知等。该疾病的症状所包含的范围较广,最常见的为精神方面的障碍,比如产生幻觉、妄想症和错觉等。
全球范围内约有1%的人患精神分裂症,而在所有接受治疗的患者中只有5%最终能够得以完全康复。此外,由于精神分裂症通常会引发合并症,例如焦虑障碍、抑郁或精神性药物滥用等。
传统上习惯把通过阻断多巴胺D2受体发挥药理作用的抗精神病药物称为第一代抗精神病药物,即“典型”抗精神病药物(如氟哌啶醇),它们治疗精神分裂症阳性症状有突破性,但未能治疗阴性症状和认知障碍。典型抗精神病药物一般有严重的EPS副作用,并且对三分之一的精神分裂症病人无效。
20世纪60年代以后,又陆续开发了一系列新一代抗精神病药,包括齐拉西酮(Ziprasidone)、利培酮(Risperidone)等,被称为第二代抗精神病药物,即新型抗精神病药,虽然它们各自的药理作用不完全一致,但却具有共同的药理特征,即对5-羟色胺(5-HT)受体(5-HT1A、2A、2c)和去甲肾上腺素(NA)受体(α1、α2)的亲和力远比对D2受体的要高,导致D2/5-HT2A的比值较高。其临床效果与第一代抗精神病药物相比有更多优势,不但对阳性症状与传统抗精神病药同样有效,而且对阴性症状、认知缺陷症状有效,作用谱更广,但是这些药物有QT间隙延长,高泌乳素血症和体重增加等不良反应。因此寻找能对精神分裂症阳性、阴性症状和认知障碍有效,而且副作用小的药物是现在研究的热点。
5-羟色胺系统在调节前额叶皮层(PFC)的功能中起着重要作用,包括情绪控制,认知行为和工作记忆。PFC的锥体神经元和GABA中间神经元包含了几个具有特别高密度的5-羟色胺受体亚型5-HT1A和5-HT2A。最近得到证明PFC和NMDA受体通道是5-HT1AR的目标,这两个受体调节大脑皮层兴奋性神经元,从而影响认知功能。实际上,各种临床前数据表明5-HT1AR可能是抗精神病药发展药物的新目标。非典型抗精神药物(如olanzapine,aripiprazole等)对5-HT1AR的高亲和力及其低的EPS副作用均说明5-羟色胺系统在调节的前额叶皮层(PFC)的功能中起着重要作用,包括情绪控制、认知行为和工作记忆。PFC的锥体神经元和GABA中间神经元包含了几个具有特别高密度5-羟色胺受体亚型5-HT1A和5-HT2A。最近研究表明5-HT1A激动剂与非典型抗精神病药物治疗相关,能改善阴性症状和认知障碍。在应用非典型抗精神病药物氯氮平治疗精神分裂症中,人们发现5-HT2A在其中起着很重要的作用,涉及到感知、情绪调节以及运动控制的各个方面。阻断5-HT2A受体可使多巴胺的释放正常化,而起到抗精神病作用。另外,5-HT2C受体与体重增加密切相关。
D3受体在脑内的分布情况主要选择性分布于边缘系统,脑内有两条主要DA神经通路,一条是黑质纹状体通路调控运动功能,另一条是中脑腹侧被盖区伏隔核前额叶皮层DA通路与学习认知和情感活动密切相关,其功能异常将导致精神分裂症,该DA通路也是脑内奖赏效应(reward efects)的主要通路,D3R在两条DA神经通路中都有分布,并和其他DA受体亚型间存在着复杂相互作用,可能作为抗精神病药物治疗的一个目标,选择性D3受体的拮抗作用能减少精神分裂症的消极和认知症状,此外能阻止锥体外系副作用,包括迟发性运动障碍,帕金森病。因此,寻找一个多受体结合副作用小的抗精神分裂症药物对临床治疗具有重要意义。
2019年FDA新批准抗精神分裂症药物Lumateperone(研发代号ITI-007),其作为5-HT2A受体的拮抗剂并拮抗几种多巴胺受体亚型(D1,D2和D4)。它具有适度的5-HT转运蛋白再摄取抑制作用。它对α-1受体具有额外的脱靶拮抗作用,没有明显的抗毒蕈碱或抗组胺能特性,其具体结构如下所示:
Figure PCTCN2021122546-appb-000001
专利PCT/US2017/015178公开马库什通式化合物,其作用于受体5-HT2A,D2,D1以及SERT(血清素再摄取转运蛋白)等受体,具有潜在的精神分裂症,帕金森症的治疗活性,
Figure PCTCN2021122546-appb-000002
尽管用于抗精神分裂症的治疗药物较多,但目前临床应用的精神分裂症药物依然存在着多种多样的不良反应,如在目前应用非常广泛的非典型抗精神分裂症药物阿立哌唑的用药患者中,有超过10%的患者会发生包括体重增加,头痛,静坐不能,失眠和胃肠道不适等不良反应,导致患者停药使病情反复。此外,虽然目前的抗精神分裂症阴性症状(指存在正常情绪反应和其他思维过程的缺陷)药物已经应用于临床,使部分患者的阴性症状得到改善,但总体而言效果有限,仍然有许多患者因阴性症状而无法痊愈和修复正常的社交功能,导致难以恢复正常的社会劳动。另外,认知障碍治疗也是目前精神分裂症治疗的一个重点,影响大多数精神分裂症患者的言语记忆、语义处理能力和注意力功能,而目前在研或上市的抗精神分裂症药物对认知功能的改善也非常有限。
目前的抗精神分裂药物除了上述问题,难治性精神分裂症的治疗依然处于困境之中。难治性精神分裂症指按照通用方法进行治疗而不能获得理想疗效的一类患者,该类患者已经经过了三种不同活性成分的抗精神病药物治疗,足量足疗程但治疗反应不佳或者无法耐受抗精神病药物的不良反应,或者即便得到充分的维持或预防治疗依然病情反复或恶化,因此抗难治性精神分裂症治疗药物一直是当下临床药物研究的难题,也是一直亟需攻克的方向。
综上所述,具备良好且持续有效的阴性症状治疗效果,改善患者认知功能,能够有效治疗难治性精神分裂症,此外还需具备较低的药物不良反应(如椎体外系反应,体重增加,恶心呕吐等药物不良反应),且作用于多靶点的抗精神分裂症药物仍然是中枢神经领域的热点研究方向。
发明内容
本发明旨在提供一种全新的作用于5-羟色胺受体和/或多巴胺受体的抗精神分裂症药物,对5-HT2A受体和/或多巴胺D2受体有良好的拮抗活性,能够有效治疗和改善精神分裂症。
本发明的目的在于提供一种具有治疗精神神经疾病活性的杂环取代的稠合γ-咔啉类衍生物,其药物组合物及其在医疗领域的应用。
本发明提供一种如通式(I)所示的化合物、其立体异构体或其可药用的盐:
Figure PCTCN2021122546-appb-000003
其中,
R 1独立地为被卤素,C1-C3的烷氧基,氰基,氨基,硝基,羟基,C2-C4的烯基,C2-C4的炔基,C3-C5的环烷基任选取代的C1-C6的烷基;
R 2独立地为-R 7-R 8-R 9-;
R 3独立地为氢,或C1-C3的烷基中的任意一种;
R 4独立地为-C(=O)-或-CH 2-中的任意一种;
R 5和R 6各自独立地为氢,或被卤素、C1-C3的烷氧基、氰基、氨基、硝基、羟基、C2-C4的烯基、C2-C4的炔基任选取代的C1-C3的烷基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-;
R 7独立地为C1-C5的亚烃基;
R 8独立地为-C(=O)-,-CH 2-或-O-中的任意一种;
R 9独立地为C1-C3的亚烃基或不存在中的任意一种;
A为被一个或多个卤素任选取代的苯基。
本发明的一个实施例方案中,所述R 1独立地为被卤素,C1-C3的烷氧基,氰基,氨基,硝基,羟基,C2-C4的烯基,C2-C4的炔基,C3-C5的环烷基任选取代的C1-C3的烷基。
本发明的一个实施例方案中,所述R 3独立地为氢,或C1-C3的烷基中的任意一种。
本发明的一个实施例方案中,所述通式(I)所示的化合物中R 4为-CH 2-。
本发明的一个实施例方案中,所述R 4为-C(=O)-。
本发明的一个实施例方案中,所述R 5和R 6各自独立地为氢,或非取代的C1-C3的烷基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-。
本发明的一个实施例方案中,所述R 5和R 6各自独立地为氢。
本发明的一个实施例方案中,所述通式(I)所示的化合物中R 4为-CH 2-,所述R 5和R 6各自独立地为氢。
本发明的一个实施例方案中,A独立地为被一个或多个卤素取代的苯基。
本发明的一个实施例方案中,所述R 1独立地为非取代的C1-C3的烷基。
本发明的一个实施例方案中,所述R 3独立地为氢,或非取代的C1-C3的烷基中的任意一种。
本发明的一个实施例方案中,所述任选取代的C1-C3的烷基中的C1-C3的烷基选自甲基、乙基或丙基中的任意一种。
本发明的一个实施例方案中,所述非取代的C1-C3的烷基选自甲基、乙基或丙基中的任意一种。
本发明的一个实施例方案中,所述C1-C5的亚烃基选自C3-C5的亚烃基。
本发明的一个实施例方案中,所述C1-C3的亚烃基选自C1-C2的亚烃基。
本发明的一个实施例方案中,所述卤素为氟,氯,溴或碘中的任意一种。
本发明的一个实施例方案中,所述通式(I)所示的化合物选自如下所示通式(I-A)所示的化合物:
Figure PCTCN2021122546-appb-000004
其中,
X为卤素;所述X在苯环的任意可取代位置进行取代;X为单取代或多取代的任意一种情形,优选单取代;
R 1,R 3,R 4,R 5,R 6,R 7,R 8和R 9如上述所定义。
本发明的一个实施例方案中,所述卤素为氟,氯,溴或碘中的任意一种,优选氟。
本发明的一个实施例方案中,所述通式(I)所示的化合物选自如下所示通式(I-B)所示的化合物:
Figure PCTCN2021122546-appb-000005
R 1,R 3,R 4,R 5,R 6,R 7,R 8,R 9和X均如上述所定义。
本发明的一个实施例方案中,所述通式(I)所示的化合物选自如下所示通式(I-B)所示的化合物:
Figure PCTCN2021122546-appb-000006
其中,
R 1独立地为甲基,乙基或丙基中的任意一种;
R 3独立地为氢,甲基,乙基,或丙基中的任意一种;
R 4独立地为-C(=O)-,或-CH 2-中的任意一种;
R 5和R 6各自独立地为氢,甲基,或乙基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-;
R 7独立地为-CH 2-CH 2-,-CH 2-CH 2-CH 2-或-CH 2-CH 2-CH 2-CH 2-中的任意一种;
R 8独立地为-C(=O)-,-CH 2-或-O-中的任意一种;
R 9独立地为-CH 2-CH 2-,-CH 2-或不存在中的任意一种;
X独立地为氟或氯中的任意一种。
本发明的一个实施例方案中,所述C2-C4的烯基包括但不限于-CH 2=CH 2,-CH 2-CH=CH 2,-CH=CH-CH 2,-CH=CH-CH 2-CH 2,-CH 2-CH=CH-CH 2,-CH 2-CH-CH=CH等。
本发明的一个实施例方案中,所述C2-C4的烯基优选C2-C3的烯基,选自-CH 2=CH 2,-CH 2-CH=CH 2或-CH=CH-CH 2中的任意一种。
本发明的一个实施例方案中,所述C2-C4的炔基包括但不限于-C≡C,-CH 2-C≡C,-C≡C-CH 2,-C≡C-CH 2-CH 2,-CH 2-C≡C-CH 2,-CH 2-CH-C≡C等。
本发明的一个实施例方案中,所述C2-C4的炔基优选C2-C3的炔基,选自-C≡C,-CH 2-C≡C或-C≡C-CH 2中的任意一种。
本发明的一个实施例方案中,所述C1-C3的烷氧基选自-O-CH 3,-O-CH 2-CH 3,-O-CH 2-CH 2-CH 3,-O-CH(CH 3)-CH 3中的任意一种,优选-O-CH 3或-O-CH 2-CH 3中的任意一种。
本发明的一个实施例方案中,所述亚烃基为具有1-20个碳原子的饱和脂肪烃直链基团,本发明所述的C1-C5的亚烃基为-CH 2-,-CH 2-CH 2-,-CH 2-CH 2-CH 2-,-CH 2-CH 2-CH 2- CH 2,-CH 2-CH 2-CH 2-CH 2-CH 2-或-CH 2-CH 2-CH 2-CH 2-CH 2-中的任意一种;所述的C1-C3的亚烃基为-CH 2-,-CH 2-CH 2-或-CH 2-CH 2-CH 2-中的任意一种;所述的C1-C2的亚烃基为-CH 2-或-CH 2-CH 2-中的任意一种。
本发明的一个实施例方案中,所述通式(I)所示的化合物选自如下所示通式(I-C)所示的化合物:
Figure PCTCN2021122546-appb-000007
其中,
R 1独立地为甲基,乙基或丙基中的任意一种;
R 3独立地为甲基,乙基或丙基中的任意一种;
R 4独立地为-C(=O)-,或-CH 2-中的任意一种;
R 5和R 6各自独立地为氢,甲基,或乙基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-。
本发明的一个实施例方案中,所述的通式(I)所示的化合物、其立体异构体或其可药用的盐选自如下所示具体化合物的任意一个:
Figure PCTCN2021122546-appb-000008
Figure PCTCN2021122546-appb-000009
本发明的一个实施例方案中,如上所述的任一如通式(I)所示的化合物的可药用的盐,所述盐选自富马酸盐,马来酸盐,磷酸盐,硝酸盐,硫酸盐,苯磺酸盐,或草酸盐。
本发明还提供了一种如通式(I-D)所示的化合物、其立体异构体或其可药用的盐:
Figure PCTCN2021122546-appb-000010
其中:
R 1,R 3,R 4,R 5和R 6如上所定义。
在本发明的一个优选的实施方式中,如通式(I-D)所示的化合物中,
R 1独立地为甲基,乙基或丙基中的任意一种;
R 3独立地为甲基,乙基或丙基中的任意一种;
R 4独立地为-C(=O)-,或-CH 2-中的任意一种;
R 5和R 6各自独立地为氢,甲基,或乙基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-。
本发明提供了一种制备如通式(I)、通式(I-A)、通式(I-B)或通式(I-C)所示的化合物、其立体异构体或其可药用盐的方法,其使用如通式(I-D)所示的化合物作为起始原料或中间体;其可包括如下步骤:
Figure PCTCN2021122546-appb-000011
通式化合物(I-D)与通式化合物(I-E)通过亲核取代反应制备得到式(I)所示的化合物;
其中:
X 1为卤素,选自氟、氯、溴或碘,优选氯;
R 1,R 2,R 3,R 4,R 5,R 6和A如上述所定义。
本发明的一个实施例方案中,制备通式(I-C)所示的化合物、其立体异构体或其可药用的盐的方法,包括如下步骤:
Figure PCTCN2021122546-appb-000012
通式化合物(I-D)与通式化合物(I-E’)通过亲核取代反应制备得到式(I-C)所示的化合物;
其中:
X 1为卤素,选自氟、氯、溴或碘,优选氯;
R 1,R 3,R 4,R 5和R 6如上述所定义。
本发明还提供了一种药物组合物,其包含治疗有效量的、如上所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,和药学上可接受的载体。
本发明的一个实施例方案中,药物组合物可以利用一种或多种可药用的载体按照常规的方式加以配制。因此,本发明的活性化合物可以被配制成口服、口腔含化给药、鼻内、肠胃外(例如静脉内、肌内或皮下)或直肠给药的剂型,或者适用于通过吸入或吹入给药的剂型。本发明的化合物或其可药用的盐也可以被配制成持续释放的剂型。
本发明的一个实施例方案中,有效剂量的本发明的化合物或其可药用的盐可与如惰性稀释剂或某种载体一起口服。根据本发明的一些实施例,可将本发明的化合物包裹于明胶胶囊中或压制成片。为口服治疗的目的,本发明化合物可与赋形剂一起使用并以片剂、锭剂、胶囊、混悬剂、糖浆剂等形式使用。根据本发明的实施例,上述制剂应含有至少0.5%(w/w)的本发明的活性化合物,但可根据特定的剂型变化,其中占单位重量的4%至约70%是便利的。在这样的药物组合物中活性化合物的量应达到适当的剂量。
本发明的一个实施例方案中,关于口服给药,本发明的活性化合物例如可通过常规手段与可药用的赋形剂加以配制成片剂或胶囊,赋形剂例如粘合剂,填充剂,润滑剂,崩解剂或润湿剂。片剂可以通过本领域熟知的方法加以包衣。用于口服给药的液体制剂,如可以采用溶液、糖浆或悬液,或挥发为干燥产物,使用前用水或其他合适的载体再生。这类液体制剂可利用药用的添加剂通过常规手段加以制备,添加剂例如悬浮剂,乳化剂,非水性载体和防腐剂。
本发明的一个实施例方案中,当本发明的活性化合物用于胃肠外施用时,可将本发明提供的化合物与无菌水或有机介质组合形成可注射的溶液或悬液。
本发明的一个实施例方案中,本发明的活性化合物可以被配制成直肠组合物,例如栓剂或保留灌肠剂,例如含有常规的栓剂基质,例如可可脂或其他甘油酯。
本发明还提供一种通式(I)所示的的化合物、其立体异构体或其可药用的盐或其药物组合物在制备涉及或调节5-羟色胺受体、5-羟色胺转运蛋白(SERT)和/或多巴胺受体的药物中的用途,优选在制备涉及或调节5-HT2A受体、5-羟色胺转运蛋白、多巴胺D1受体和/或多巴胺D2受体的药物中的用途,更优选在制备涉及或调节5-HT2A受体和/或多巴胺D2受体的药物中的用途。其中所述药物任选包含另外一种或多种调节哺乳动物神经系统或缓解精神疾病的活性剂。
本发明的一个实施例方案中,所述调控(调节)包括但不限于对受体的抑制活性或拮抗活性。
本发明的一个实施例方案中,本发明还提供一种通式(I)所示的的化合物、其立体异 构体或其可药用的盐在制备治疗神经精神类疾病的药物中的用途。
本发明的一个实施例方案中,所述神经精神类疾病选自抑郁症(例如重度抑郁症(major depressive disorder,MDD))、焦虑症、痴呆症、精神分裂症、睡眠障碍、运动障碍、痴呆症患者的行为障碍、帕金森病、阿尔茨海默病、偏头痛、多动症(例如注意力缺陷多动症)、强迫症、社交恐惧症、神经退行性疾病、双相情感障碍、创伤后应激综合征、成瘾性疾病、戒断综合征或注意力缺陷中的一种或多种。
本发明的一个实施例方案中,本发明所述的神经精神类疾病优选抑郁症(例如重度抑郁症MDD)、焦虑症、痴呆症、精神分裂症、睡眠障碍、运动障碍、痴呆症患者的行为障碍、神经退行性疾病或双相情感障碍中的任意一种或多种,更优选抑郁症、焦虑症、精神分裂症或神经退行性疾病中的一种或多种。
本发明的一个实施例方案中,所述神经精神类疾病优选精神分裂症。
本发明进一步涉及一种治疗、缓解和/或预防涉及5-羟色胺受体、5-羟色胺转运蛋白(SERT)和/或多巴胺受体的疾病的方法,该方法包括向需要其的患者施用治疗有效剂量的本发明的前述通式所示的化合物或其可药用的盐;优选涉及一种治疗、缓解或预防涉及5-HT2A受体、5-羟色胺转运蛋白、多巴胺D1受体和/或多巴胺D2受体的疾病的方法,该方法包括向需要其的患者施用治疗有效剂量的本发明的前述通式所示的化合物或其可药用的盐。在本发明的一些实施方式中,所述5-羟色胺受体优选5-HT2A受体,所述多巴胺受体优选多巴胺D2受体。该方法表现出突出的疗效和较少的副作用。
在本发明的一些实施方式中,本发明涉及一种治疗、缓解和/或预防神经精神类疾病的方法,该方法包括向需要其的患者施用治疗有效剂量的本发明的前述通式所示的化合物或其可药用的盐。
在本发明的一些优选的实施方式中,所述神经精神类疾病选自抑郁症(例如重度抑郁症)、焦虑症、痴呆症、精神分裂症、睡眠障碍、运动障碍、痴呆症患者的行为障碍、帕金森病、阿尔茨海默病、偏头痛、多动症(例如注意力缺陷多动症)、强迫症、社交恐惧症、神经退行性疾病、双相情感障碍、创伤后应激综合征、成瘾性疾病、戒断综合征、或注意力缺陷中的一种或多种,优选抑郁症(例如重度抑郁症)、焦虑症、痴呆症、精神分裂症、睡眠障碍、运动障碍、痴呆症患者的行为障碍、神经退行性疾病或双相情感障碍中的任意一种或多种。
除非另有定义,本文所用所有技术和科学术语与本发明所属领域的普通技术人员通常理解的含义相同。若存在矛盾,则以本申请提供的定义为准。当本文中出现商品名时,意在指代其对应的商品或其活性成分。本文引用的所有专利、已经公开的专利申请和出 版物均通过引用并入到本文中。
术语“任选”、“任选地”或“任选存在”是指随后描述的事件或情形可以但不一定出现,并且该描述包括其中所述事件或情形出现的情况和不出现的情况。例如,“任选存在的键”是指该键可以存在或可以不存在,并且该描述包括单键、双键或三键等。
术语“包含”、“包括”、“具有”、“含有”或“涉及”为开放式表达,即包括本发明所指明的内容,但并不排除其他方面的内容。应当理解上述术语例如“包含”可以涵盖封闭式的含义,即“由…组成”。
像本发明所描述的,本发明的化合物可以任选地被一个或多个取代基所取代,如上面的通式化合物或者像实施例中特定的实例、子类。应了解术语“任选取代的”与术语“取代或未取代的”可以交换使用。一般而言,术语“取代的”表示所给结构中的一个或多个氢原子被特定取代基所取代。除非其他方面表明,任选取代的基团可以在该基团各个可取代的位置进行取代。当所给出的结构式中不只一个位置能被选自特定基团的一个或多个取代基所取代时,那么取代基可以相同或不同地在各个位置取代。
另外,需要说明的是,除非以其他方式明确指出,在本发明中所采用的描述方式“分别独立地为”应做广义理解,其既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响,也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
在本文中,“Z”和“-Z-”均表示为同一特定的基团,其可以互换使用。
本文所用的表述“X选自A、B或C”、“X选自A、B和C”、“X为A、B或C”、“X为A、B和C”等不同用语均表达了相同的意义,即表示X可以是A、B、C中的任意一种或几种。
当任何变量(例如R),以及带有标记的变量(例如R 1、R 2、R 3、R 4、R 5、R 6、R 7等)在化合物的组成或结构中出现一次以上时,其在每次出现时在每一种情况下的定义都是独立的。例如,如果一个基团被0、1、2、3或4个R取代基所取代,则所述基团可以任选地至多被四个R取代基所取代,并且每种情况下的每个R取代基的选项都是相互独立的。
术语“药学上可接受”的物质指这样的物质,其在正常的医学判断范围内适用于与患者的组织接触而不会有不适当毒性、刺激性、过敏反应等,具有合理的利弊比,且能有效用于其目的用途。
术语“可药用盐”指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
术语“药学上可接受的载体”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些物质。“药学上可接受的载体”包括但不限于助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味剂、表面活性剂、润湿剂、分散剂、崩解剂、稳定剂、溶剂或乳化剂。
术语“给药”或“给予”等指可以使化合物或组合物能够递送至期望的生物作用位点的方法。这些方法包括但不限于口服或肠胃外(包括脑室内、静脉内、皮下、腹膜内、肌内、血管内注射或输注)、局部、直肠给药等。特别是注射或口服。
如本文所用,术语“治疗”包括缓解、减轻或改善疾病或症状,预防其他症状,改善或预防症状的潜在代谢因素,抑制疾病或症状,例如,阻止疾病或症状发展,减轻疾病或症状,促进疾病或症状缓解,或使疾病或症状的病征停止,和延伸至包括预防。“治疗”还包括实现治疗性获益和/或预防性获益。治疗性获益是指根除或改善所治疗的病症。此外,治疗性获益通过根除或改善一个或多个与潜在疾病相关的生理病征达到,尽管患者可能仍患有潜在疾病,但可观察到患者疾病的改善。预防性获益是指,患者为预防某种疾病风险而使用组合物,或患者出现一个或多个疾病生理病症时服用,尽管尚未诊断此疾病。
术语“活性成分”、“治疗剂”、“活性物质”或“活性剂”是指一种化学实体,其可以有效地治疗或预防目标紊乱、疾病或病症。
术语“神经精神类疾病”是指神经类疾病与精神类疾病的总称,包含神经类疾病和/或精神类疾病。
针对药物、药物单元或活性成分而言,术语“有效量”、“治疗有效量”或“预防有效量”是指副作用可接受的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于个体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
如本文所使用的“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
在本说明书的各部分,本发明公开化合物的取代基按照基团种类或范围公开。特别指出,本发明包括这些基团种类和范围的各个成员的每一个独立的次级组合。例如,术语“C1-C6烷基”特别指独立公开的甲基、乙基、C3烷基、C4烷基、C5烷基和C6烷基。烷基基团的实例包括但不限于甲基(Me、-CH 3),乙基(Et、-CH 2CH 3),正丙基(n-Pr、-CH 2CH 2CH 3)、异丙基(i-Pr、-CH(CH 3) 2),正丁基(n-Bu、-CH 2CH 2CH 2CH 3)、异丁基(i-Bu、 -CH 2CH(CH 3) 2)、仲丁基(s-Bu、-CH(CH 3)CH 2CH 3)、叔丁基(t-Bu、-C(CH 3) 3)、正戊基(-CH 2CH 2CH 2CH 2CH 3)、2-戊基(-CH(CH 3)CH 2CH 2CH 3)、3-戊基(-CH(CH 2CH 3) 2)、2-甲基-2-丁基(-C(CH 3) 2CH 2CH 3)、3-甲基-2-丁基(-CH(CH 3)CH(CH 3) 2)、3-甲基-1-丁基(-CH 2CH 2CH(CH 3) 2)、2-甲基-1-丁基(-CH 2CH(CH 3)CH 2CH 3)、正己基(-CH 2CH 2CH 2CH 2CH 2CH 3)、2-己基(-CH(CH 3)CH 2CH 2CH 2CH 3)、3-己基(-CH(CH 2CH 3)(CH 2CH 2CH 3))、2-甲基-2-戊基(-C(CH 3) 2CH 2CH 2CH 3)、3-甲基-2-戊基(-CH(CH 3)CH(CH 3)CH 2CH 3)、4-甲基-2-戊基(-CH(CH 3)CH 2CH(CH 3) 2)、3-甲基-3-戊基(-C(CH 3)(CH 2CH 3) 2)、2-甲基-3-戊基(-CH(CH 2CH 3)CH(CH 3) 2)、2,3-二甲基-2-丁基(-C(CH 3) 2CH(CH 3) 2)、3,3-二甲基-2-丁基(-CH(CH 3)C(CH 3) 3)、正庚基、正辛基,等等。
术语“氢(H)”表示单个氢原子。这样的原子团可以与其他基团连接,譬如与氧原子相连,形成羟基基团。
术语“卤素”是指氟(F)、氯(Cl)、溴(Br)或碘(I)。
术语“芳基”表示含有6-14个环原子、或6-12个环原子、或6-10个环原子的单环、双环和三环的碳环体系,其中至少一个环是芳香族的。芳基基团通常,但不必须地通过芳基基团的芳香性环与母体分子连接。芳基基团的实例可以包括苯基、萘基和蒽。所述芳基基团任选地被一个或多个本发明所描述的取代基所取代。
烷氧基基团的实例包括但并不限于甲氧基(MeO、-OCH 3)、乙氧基(EtO、-OCH 2CH 3)、1-丙氧基(n-PrO、n-丙氧基、-OCH 2CH 2CH 3)、2-丙氧基(i-PrO、i-丙氧基、-OCH(CH 3) 2)、1-丁氧基(n-BuO、n-丁氧基、-OCH 2CH 2CH 2CH 3)、2-甲基-l-丙氧基(i-BuO、i-丁氧基、-OCH 2CH(CH 3) 2)、2-丁氧基(s-BuO、s-丁氧基、-OCH(CH 3)CH 2CH 3)、2-甲基-2-丙氧基(t-BuO、t-丁氧基、-OC(CH 3) 3)、1-戊氧基(n-戊氧基、-OCH 2CH 2CH 2CH 2CH 3)、2-戊氧基(-OCH(CH 3)CH 2CH 2CH 3)、3-戊氧基(-OCH(CH 2CH 3) 2)。
术语“-C(=O)-”表示羰基。
下述发明详述旨在举例说明非限制性实施方案,使本领域其它技术人员更充分地理解本发明的技术方案、其原理及其实际应用,以便本领域其它技术人员可以以许多形式修改和实施本发明,使其可最佳地适应特定用途的要求。
本发明有益的技术效果
本发明提供的化合物为一种作用于5-HT 2A受体和/或D 2受体的拮抗剂,对5-HT 2A受体和/或D2(D 2L、D 2s)受体具有良好的拮抗作用;和/或具有较高的D2/5-HT2A比值,具有良好的选择性;和/或具有良好的药代动力学性质;和/或具有良好的体内药效作用;和/或不但对阳性症状与传统抗精神病药同样有效,而且对阴性症状、认知缺陷症状具有 更强的改善作用,副作用更低(例如降低诱发EPS反应的可能性等)。
具体实施方式
下面详细描述本发明的实施例。下面描述的实施例是示例性的,仅用于解释本发明,而不能理解为对本发明的限制。除非另外指明,本文所指的比例、百分比等均以重量计。
合成实施例
实施例1、8-(4-(4-氟苯基)-4-氧代丁基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮(化合物1)
第一步:4-亚硝基-3,4-二氢喹喔啉-2(1H)-酮的制备(中间体1-2)
Figure PCTCN2021122546-appb-000013
将原料3,4-二氢喹喔啉-2(1H)-酮(2.3g,16mmol)溶于乙酸和水(25/12mL)的混合溶剂中,冰浴条件下缓慢滴加亚硝酸钠(1.1g,16mmol)的水溶液(12mL),保温反应2h后,反应液抽滤,滤饼水洗(12mL)后,干燥得到中间体1-2,黄色固体2.2g,收率:81%。LCMS m/z(M+H) +:178.1。
第二步:4-氨基-3,4-二氢喹喔啉-2(1H)-酮盐酸盐的制备(中间体1-3)
Figure PCTCN2021122546-appb-000014
将中间体1-2(1.5g,8.47mmol)溶于冰乙酸和水(25/25mL)的混合溶剂中,冰浴条件下缓慢加入锌粉(3.0g,46.1mmol),保温反应30min后,移至室温继续搅拌反应2h,反应液过滤浓缩后,浓缩物重新溶解于乙酸乙酯(100mL)后搅拌30min,反应液再次过滤,滤液干燥后加入浓度为4N的氯化氢二氧六环溶液(3mL),搅拌30min后,反应液浓缩得到中间体1-3,黄色固体1.5g粗品,无需进一步纯化直接用于下一步原料使用。
第三步:6b-甲基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-去]喹噁啉-2(3H)-酮的制备(中间体1-5)
Figure PCTCN2021122546-appb-000015
将中间体1-3(1.5g,7.54mmol)和中间体1-4(1.4g,6.78mmol)加入至异丙醇(50mL)中,于110℃条件下反应15h,反应液冷却至室温后浓缩,产物经柱层析(DCM/MeOH=1/20)分离纯化得到中间体1-5,黄色固体620mg,收率:27%。LCMS m/z(M+H) +:340.1。
第四步:3,6b-二甲基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹噁啉-2(3H)-酮的制备(中间体1-6)
Figure PCTCN2021122546-appb-000016
将中间体1-5(0.62g,1.83mmol)溶于DMF(10mL)中,冰浴条件下加入氢钠(110mg,2.75mmol),保温反应30min后,加入碘甲烷(390mg,2.75mmol),然后将反应液移至室温搅拌2h,反应完毕后,将反应液重新置于冰浴中,反应液加水(5mL)淬灭,反应液用乙酸乙酯(20mL x 3)萃取后,有机相用硫酸钠干燥,产物经柱层析分离纯化得到目标中间体1-6,黄色固体500mg,收率:77%。LCMS m/z(M+H) +:354.1。
第五步:3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹噁啉-2(3H)-酮的制备(中间体1-7)
Figure PCTCN2021122546-appb-000017
将中间体1-6(0.5g,1.41mmol)溶于四氢呋喃和水(50/5mL)的混合溶剂中,氢氧化钠(113mg,2.82mmol)加入至上述溶液中,反应液于40℃条件下反应15h后,浓缩去除四氢呋喃。残余物用二氯甲烷和甲醇的混合溶剂萃取(10/1,20mL x 3),有机相经硫酸钠干燥,柱层析(DCM/MeOH=10/1-5/1)纯化后得到目标中间体1-7,黄色固体200mg,收率:55%。LCMS m/z(M+H) +:258.2。
第六步:8-(4-(4-氟苯基)-4-氧代丁基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物1)
Figure PCTCN2021122546-appb-000018
将中间体1-7(1.1g,4.28mmol),中间体1-8(1.7g,8.56mmol),碘化钾(1.4g,8.56mmol)和DIEA(1.1g,8.56mmol)加入至DMF(20mL)中,反应液于78℃反应3h后冷却至室温,反应液直接浓缩,产物经制备液相(CH 3CN:H 2O(0.1%NH 4HCO 3)=10-70%,UV:214nm,flow rate 15mL/min)制备得到化合物1,类白色固体245mg,收率:13%。LRMS m/z(M+H) +:422.2。
1H NMR(CDCl 3,400MHz):δ8.06-8.03(m,2H),7.19-7.15(m,2H),6.88-6.76(m,3H),4.45(d,J=14.4Hz,1H),3.41-3.37(m,4H),3.05-3.02(m,2H),2.95(s,1H),2.71-2.28(m,5H),1.99-1.87(m,4H),1.67-1.50(m,4H)。
实施例1-A、(6bR,10aS)-8-(4-(4-氟苯基)-4-氧代丁基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物1-A)
Figure PCTCN2021122546-appb-000019
化合物1(1.6g)通过手性拆分,得到化合物1-A(427.7mg)。手性拆分条件如下:
仪器 Gilson GX-281
柱型 Chiralpak IE 250mm*30mm 5μm
进样量 1500μL
流动相 Hex:EtOH=40:60
流速 25mL/min
检测波长 254nm
柱温 30℃
t R=6.968min
LCMS m/z(M+H) +:422.2.
1H NMR(400MHz,CDCl 3):δ8.02-7.99(m,2H),7.16-7.10(m,2H),6.83-6.73(m,3H),4.01(d,J=14.0Hz,1H),3.35(d,J=14.0Hz,1H),3.33(s,3H),3.00-2.91(m,3H),2.67-2.55(m,2H),2.40-2.19(m,3H),1.98-1.81(m,4H),1.55-1.46(m,4H).
实施例1-B、(6bS,10aR)-8-(4-(4-氟苯基)-4-氧代丁基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物1-B)
Figure PCTCN2021122546-appb-000020
化合物1(1.6g)通过手性拆分,得到化合物1-B(467.8mg)。手性拆分条件如下:
仪器 Gilson GX-281
柱型 Chiralpak IE 250mm*30mm 5μm
进样量 1500μL
流动相 Hex:EtOH=40:60
流速 25mL/min
检测波长 254nm
柱温 30℃
t R=8.030min
LCMS m/z(M+H) +:422.2.
1H NMR(400MHz,CDCl 3):δ8.02-7.99(m,2H),7.16-7.10(m,2H),6.84-6.73(m,3H),4.01(d,J=14.0Hz,1H),3.36(d,J=14.0Hz,1H),3.33(s,3H),3.00-2.91(m,3H),2.67-2.55(m,2H),2.40-2.19(m,3H),1.98-1.81(m,4H),1.55-1.46(m,4H).
实施例2、6b-乙基-8-(4-(4-氟苯基)-4-氧代丁基)-3-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物2)
第一步:6b-乙基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-二]喹噁啉-2(3H)-酮的制备(中间体2-2)
Figure PCTCN2021122546-appb-000021
中间体2-1(800mg,3.58mmol)和中间体1-3(1.5g,9.2mmol)溶于异丙醇(50mL)中,反应液于110℃反应15h,反应液冷却至室温后减压旋蒸得到粗品,产物经柱层析(EA/PE=1/2)分离纯化得到中间体2-2,黄色固体360mg,收率28.4%。LRMS m/z(M+H) +:354.2。
第二步:6b-乙基-3-甲基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(中间体2-3)
Figure PCTCN2021122546-appb-000022
冰浴条件下向中间体2-2(400mg,1.13mmol)的DMF(15mL)溶液中加入NaH(68mg,1.70mmol),保温反应30min,然后向反应体系中加入CH 3I(241mg,1.70mmol),加毕,反应液移至室温反应16h,冰浴条件下用水(15mL)淬灭,反应液用乙酸乙酯(20mL x 3)萃取。合并有机相用无水硫酸钠干燥,过滤,滤液经减压浓缩,粗品经柱层析(PE/EA=5:1,v/v)得到中间体2-3,黄色固体200mg,收率48%。LCMS m/z(M+H) +:368.3。
第三步:6b-乙基-3-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-二]喹恶啉-2(3H)-酮(中间体2-4)
Figure PCTCN2021122546-appb-000023
室温条件下中间体2-3(200mg,0.54mmol)的MeOH/H 2O(10mL/1mL)溶液中加入K 2CO 3(150mg,1.09mmol),加毕后反应液在70℃反应4h,反应液降温至室温后加水(10mL),然后用二氯甲烷(20mL x 3)萃取,合并的有机相经无水硫酸钠干燥,过滤,滤液经减压旋蒸得到粗品,经柱层析(DCM/MeOH=20:1,v/v)得到中间体2-4,黄色油状物120mg,收率:65%。LCMS m/z(M+H) +:272.2。
第四步:6b-乙基-8-(4-(4-氟苯基)-4-氧代丁基)-3-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物2)
Figure PCTCN2021122546-appb-000024
中间体2-4(100mg,0.37mmol),中间体1-8(590mg,2.95mmol),KI(245mg,1.48mmol)和DIEA(190mg,1.48mmol)的DMF(10mL)溶液,在78℃搅拌反应16h,反应液降温至室温后加水10mL,用乙酸乙酯萃取(20mL x 3),合并的有机相经无水硫酸钠干燥,过滤,滤液减压旋蒸得到粗品,经制备液相分离纯化(CH 3CN:H 2O(0.1%NH 4HCO 3)=40-80%,UV:214nm,flow rate 15mL/min)得到化合物2,45.0mg,收率23%。LCMS m/z(M+H) +:436.2。
1H NMR(CDCl 3,400MHz):δ8.03-7.95(m,2H),7.13(t,J=8.4Hz,2H),6.83-6.74(m,2H),6.74-6.68(m,1H),4.01(d,J=14.0Hz,1H),3.37(d,J=14.4Hz,1H),3.32(s,3H),3.10-3.04(m,1H),2.98(t,J=7.2Hz,2H),2.69-2.51(m,2H),2.43-2.31(m,1H),2.30-2.15(m,2H),2.11-1.99(m,1H),1.97-1.76(m,6H),0.88(t,J=7.6Hz,3H)。
实施例3、3-乙基-8-(4-(4-氟苯基)-4-氧代丁基)-6b-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物3)
第一步:3-乙基-6b-甲基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-德]喹噁啉-2(3H)-酮的制备(中间体3-1)
Figure PCTCN2021122546-appb-000025
冰浴条件下,向中间体1-5(0.2g,0.59mmol,参照实施例1第三步所述方法制备)的DMF(10mL)溶液中,加入氢化钠(28mg,0.71mmol),保温反应30分钟后加入碘乙烷(148mg,0.89mmol),加毕,移至室温搅拌1小时,反应液再次移至冰浴中加水(5mL)淬灭,反应液用乙酸乙酯(10mL x 3)萃取,合并的有机相用无水硫酸钠干燥后浓缩,粗 品柱层析(DCM/MeOH=20/1)得到中间体3-1,黄色固体150mg,收率:69%。LCMS m/z(M+H) +:368.2。
第二步:3-乙基-6b-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(中间体3-2)
Figure PCTCN2021122546-appb-000026
向中间体3-1(150mg,0.41mmol)的四氢呋喃/水(10mL/2mL)溶液中加入氢氧化钠(32mg,0.82mmol),加毕后,反应液于40℃条件下反应2h,减压旋干溶剂后,粗品经柱层析(DCM/MeOH/NH 3H 2O=100/10/1)分离纯化得到中间体3-2,黄色油状物91mg,收率82%。LCMS m/z(M+H) +:272.2。
第三步:3-乙基-8-(4-(4-氟苯基)-4-氧代丁基)-6b-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物3)
Figure PCTCN2021122546-appb-000027
向中间体3-2(91mg,0.34mmol)的DMF(10mL)溶液中加入中间体1-8(136mg,0.68mmol),碘化钾(10mg)和N,N-二异丙基乙胺(88mg,0.68mmol),加毕后反应液于78℃反应3h,反应液降至室温后浓缩得到粗品,粗品用制备液相(CH 3CN:H 2O(0.1%NH 4HCO 3)=10-60%,UV:214nm,flow rate 15mL/min)分离纯化得到化合物3,42.3mg,收率:28%。LCMS m/z(M+H) +:436.2。
1H NMR(CDCl 3,400MHz):δ8.04-8.01(m,2H),7.17-7.13(m,2H),6.86-6.76(m,3H),4.06-3.98(m,2H),3.90-3.84(m,1H),3.37(d,J=14.4Hz,1H),3.03-2.99(m,2H),2.91(s,1H),2.68-2.56(m,2H),2.43-2.21(m,3H),1.97-1.84(m,5H),1.47(s,3H),1.30-1.27(m,3H)。
实施例4、8-(4-(4-氟苯基)-4-氧代丁基)-1,1,3,6b-四甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物4)
第一步:3,6b-二甲基-2-氧代-2,3,6b,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱] 喹喔啉-8(7H)-羧酸酯的制备(中间体4-1)
Figure PCTCN2021122546-appb-000028
向中间体1-7(1.2g,4.6mmol,参照实施例1第五步制备方法制备)的DCM(20mL)溶液中依次加入三乙胺(1.3g,13.2mmol),二碳酸二叔丁酯(4.3g,19.8mmol),室温反应2h后加入水(15mL)淬灭,分液得到有机相后干燥浓缩得到的粗品经柱层析(EA/PE=1/3)分离纯化得到中间体4-1,白色固体1.6g,收率:68%。LCMS m/z(M-56+H) +:302.2。
第二步:1,1,3,6b-四甲基-2-氧代-2,3,6b,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-8(7H)-羧酸盐的制备(中间体4-2)
Figure PCTCN2021122546-appb-000029
将中间体4-1(0.66g,1.85mmol)溶于四氢呋喃(20mL)中,置于-78℃的低温反应器中,氩气保护条件下缓慢滴加LDA(二异丙基氨基锂,2.0M,3.7mL,7.4mmol),滴毕保温反应1h。然后缓慢滴加碘甲烷(2.1g,14.8mmol),加毕保温反应2h,反应液移至室温后加入水(20mL)淬灭,反应液用乙酸乙酯萃取(15mL x 3),有机相干燥后浓缩得到的粗品经柱层析(EA/PE=1/2)分离纯化中间体4-2,黄色油状410mg,收率:58%。LCMS m/z(M+H) +:386.2。
第三步:1,1,3,6b-四甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮的制备(中间体4-3)
Figure PCTCN2021122546-appb-000030
冰浴条件下向中间体4-2(0.44g,1.14mmol)和1,6-Lutidine(0.24g,2.28mmol)的DCM(50mL)溶液中加入TMSOTf(三氟甲磺酸三甲基硅酯,380mg,1.71mmol),保温反应30min后,反应液用氯化铵水溶液(15mL)淬灭,分液后有机相经干燥浓缩得到 粗品,产物经柱层析(DCM/MeOH=10/1)分离纯化得到中间体4-3,黄色油200mg,收率:61%。LCMS m/z(M+H) +:286.2。
第四步:8-(4-(4-氟苯基)-4-氧代丁基)-1,1,3,6b-四甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)-酮(化合物4)
Figure PCTCN2021122546-appb-000031
将中间体4-3(180mg,0.63mmol),中间体1-8(252mg,1.26mmol),碘化钾(20mg,0.12mmol)和DIEA(244mg,1.89mmol)依次加入DMF(10mL),于78℃反应5h,反应完毕后反应液减压浓缩直接用制备液相(CH 3CN:H 2O(0.1%NH 4HCO 3)=10-70%,UV:214nm,flow rate 15mL/min)分离纯化得到化合物4,100mg,收率:38%。LCMS m/z(M+H) +:450.2。
1H NMR(CDCl 3,400MHz):δ8.02-7.95(m,2H),7.16-7.12(m,2H),6.79-6.70(m,3H),3.52(s,1H),3.31(s,3H),2.95-2.94(m,2H),2.78-1.92(m,10H),1.72(s,3H),1.45(s,3H),1.15(s,3H)。
实施例5、8-(4-(4-氟苯基)-4-氧代丁基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-1,2(3H)-二酮(化合物5)
第一步:3,6b-二甲基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹噁啉-1,2(3H)-二酮的制备(中间体5-1)
Figure PCTCN2021122546-appb-000032
向中间体1-6(200mg,0.50mmol)的四氯化碳(20mL)溶液中加入RuO 2(37mg,0.28mmol),NaIO 4(300mg,1.4mmol),在室温下反应48h后,反应液直接浓缩得到粗品。产物经用柱层析(EA/PE=1/1)得到中间体5-1,黄色固体114mg,收率:54%。LCMS m/z(M+H) +:368.2。
第二步:3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-1,2(3H)-二酮(中间体5-2)
Figure PCTCN2021122546-appb-000033
向中间体5-1(180mg,0.49mmol)的甲醇/水(2mL,v/v=10/1)溶液中加入碳酸钾(136mg,0.98mmol),反应液于70℃反应3h,反应完毕后,浓缩得到中间体5-2,黄色油状物150mg,产物无需进一步纯化直接用于下一步原料。LCMS m/z(M+H) +:272.2。
第三步:8-(4-(4-氟苯基)-4-氧代丁基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-1,2(3H)-二酮的制备(化合物5)
Figure PCTCN2021122546-appb-000034
向中间体5-2(150mg,0.55mmol)的DMF(3mL)溶液中,依次加入中间体1-8(150mg,0.75mmol),碳酸钾(37mg,0.27mmol),加毕后反应液于78℃反应3小时,反应完毕后反应液直接减压浓缩得到的残余物经制备液相(CH 3CN:H 2O(0.1%NH 4HCO 3)=10-70%,UV:214nm,flow rate 15mL/min)分离纯化得到化合物5,35mg,收率:15%。LCMS m/z(M+H) +:436.2。
1H NMR(CDCl 3,400MHz):δ8.02-7.98(m,2H),7.15-7.10(t,J=8.4Hz,1H),6.59-6.56(m,3H),4.32-4.29(m,2H),4.32-4.29(d,J=11.2Hz,2H),3.07-2.99(m,5H),2.66-2.45(m,5H),2.12(t,J=7.6Hz,2H),2.03-1.97(m,4H)。
实施例6、8-(3-(4-氟苯氧基)丙基)-3,6b-二甲基-6b,7,8,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-2(3H)-酮的制备(化合物6)
Figure PCTCN2021122546-appb-000035
将中间体1-7(2.0g,7.78mmol,参照实施例1第五步所述方法制备),中间体6-1(2.9g,15.56mmol),碘化钾(2.0g,15.56mmol)和DIEA(2.0g,15.56mmol)依次加入至DMF(40mL)中,于78℃反应3h。反应完毕后反应液直接浓缩,得到粗品经制备液相分离纯化得到化合物6,1.5g,收率:47%。LCMS m/z(M+H) +:410.2。
1H NMR(CDCl 3,400MHz):δ6.98-6.93(m,2H),6.84-6.28(m,5H),4.03-3.96(m,3H),3.38(s,1H),3.33(s,3H),2.92(s,1H),2.68-2.22(m,5H),1.96-1.89(m,5H),1.49(s,3H)。
实施例7、4-(3,6b-二甲基-2,3,6b,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-8(7H)-基)-1-(4-氟苯基)丁烷-1-酮的制备(化合物7)
第一步:3,6b-二甲基-2,3,6b,7,8,9,10,10a-八氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉的制备(中间体7-1)
Figure PCTCN2021122546-appb-000036
将中间体1-7(0.2g,0.78mmol)溶于THF(10mL)中,缓慢加入浓度为1M的硼烷的四氢呋喃溶液(1.56mL,1.56mmol),反应液室温搅拌15h后,冰浴条件下滴加1N盐酸水溶液(2mL)淬灭,淬灭后的反应液直接浓缩柱层析得到中间体7-1,80mg,收率:42%。LCMS m/z(M+H) +:244.2。
第二步:4-(3,6b-二甲基-2,3,6b,9,10,10a-六氢-1H-吡啶[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-8(7H)-基)-1-(4-氟苯基)丁烷-1-酮的制备(化合物7)
Figure PCTCN2021122546-appb-000037
将中间体7-1(75mg,0.308mmol),中间体1-8(184mg,0.924mmol),碘化钾(101mg,0.616mmol),DIEA(119mg,0.924mmol)加入至DMF(3mL)中,反应液于78℃反应3h。反应液冷却至室温后直接浓缩去除DMF,产物经制备液相(CH 3CN:H 2O(0.1%NH 4HCO 3)=10-70%,UV:214nm,flow rate 15mL/min)分离纯化得到目标化合物7,32mg,收率:25%。LCMS m/z(M+H) +:407.9。
1H NMR(CDCl 3,400MHz):δ8.06-8.03(m,2H),7.19-7.15(m,2H),6.73-6.71(m,1H),6.49-6.44(m,2H),3.67-3.61(m,1H),3.36-3.27(m,2H),3.07-3.04(m,2H),2.90-2.83(m,5H),2.71-2.07(m,10H),1.30(s,3H).
对比例1、8-(4-(4-氟苯基)-4-羰基丁基)-3-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-2(3H)-酮
第一步:8-(2,2,2-三氟乙酰基)-7,8,9,10-四氢-1H-吡啶并[3',4':4,5]吡咯[1,2,3-脱]喹喔啉-2(3H)酮的合成(中间体D1-2)
Figure PCTCN2021122546-appb-000038
将中间体1-3(3g,15mmol),中间体D1-1(2.9g,15mmol),异丙醇(30mL)依次加入100mL单口瓶中,升温至回流反应过夜,反应完全后,将反应液倒入冰水(5mL)中,乙酸乙酯(15mL)萃取,无水硫酸钠干燥,减压浓缩用硅胶柱层析(石油醚:乙酸乙酯=2:1)得到中间体D1-2,1.8g,收率:30%。LCMS m/z(M+H) +:324.2。
第二步:8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-2(3H)-酮的合成(中间体D1-3)
Figure PCTCN2021122546-appb-000039
将中间体D1-2(1g,31mmol)溶于三氟乙酸(10mL)中,冰浴下加入氰基硼氢化钠(388mg,6.2mmol),冰浴反应2小时,反应完全后,将反应液倒入冰水(50mL)中,调节PH~7,乙酸乙酯(15mL)萃取,无水硫酸钠干燥,减压浓缩至干得到中间体D1-3,700mg,收率:70%。
第三步:3-甲基-8-(2,2,2-三氟乙酰基)-6b,7,8,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-2(3H)-酮的合成(中间体D1-4)
Figure PCTCN2021122546-appb-000040
将中间体D1-3(700mg,2.15mmol)溶于DMF(10mL)中,冰浴下加入氢化钠(100mg,2.58mmol)和氘代碘甲烷(340mg,2.37mmol),冰浴反应1小时,反应完全后,将反应液倒入冰水(5mL)中,乙酸乙酯(15mL)萃取,无水硫酸钠干燥,减压浓缩至干得到中间体D1-4,500mg,收率:69%。LCMS m/z(M+H) +:340.1。
第四步:3-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-2(3H)-酮的合成(中间体D1-5)
Figure PCTCN2021122546-appb-000041
将中间体D1-4(300mg,0.88mmol)溶于甲醇(5mL)中,加入碳酸钾(235mg,1.7mmol),80℃反应2小时,反应完全后,减压浓缩,加入水(15mL),二氯甲烷(10mL×3)萃取,无水硫酸钠干燥,减压浓缩得到中间体D1-5,200mg,收率:93%。LCMS m/z(M+H) +:244.0。
第五步:8-(4-(4-氟苯基)-4-羰基丁基)-3-甲基-6b,7,8,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-2(3H)-酮的合成
Figure PCTCN2021122546-appb-000042
将中间体D1-5(200mg,0.82mmol)和中间体D1-6(324mg,1.23mmol)溶于乙腈(5mL)中,加入碳酸铯(400mg,1.23mmol),室温反应过夜,反应结束后,抽滤,减压浓缩至干,通过Pre HPLC(MeCN:H 2O=40:60)制备得到终产物对比例1,56mg, 收率:18%。LCMS m/z(M+H) +:408.2。
1H NMR(600MHz,CD 3OD):δ8.08–8.06(m,2H),7.24–7.21(m,2H),6.97–6.85(m,3H),4.03(d,J=13.8Hz,1H),3.47–3.38(m,4H),3.32(s,3H),3.22–3.20(m,1H),3.13–3.08(m,2H),2.87–2.80(m,3H),2.30–2.20(m,2H),2.11–2.05(m,3H)。
对比例2、1-(4-氟苯基)-4-(3-甲基-2,3,6b,9,10,10a-六氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉-8(7H)-基)丁烷-1-酮
Figure PCTCN2021122546-appb-000043
将3-甲基-2,3,6b,7,8,9,10,10a-八氢-1H-吡啶并[3',4':4,5]吡咯并[1,2,3-脱]喹喔啉(500mg,2.18mmol)溶解于15mL无水乙腈中,然后加入碳酸铯(1.42g,4.36mmol)和1-(4-氟苯基)-4-碘丁烷-1-酮(955.27mg,3.27mmol),反应液在室温条件下搅拌4个小时,LCMS监测产物生成。将反应液溶于水中(50mL),乙酸乙酯(50mL×2)萃取两次,将收集到的有机相经过无水硫酸钠干燥,过滤和浓缩,粗品经过快速制备过柱机纯化(石油醚:乙酸乙酯=10:1~1:4)得到对比例2,600mg,2.18mmol,收率:69.93%。
1H NMR(600MHz,DMSO-d 6)δ:8.06-8.01(m,2H),7.33(t,J=9.0Hz,2H),6.49(t,J=7.2Hz,1H),6.39(d,J=7.2Hz,1H),6.31(d,J=7.8Hz,1H),3.45-3.22(m,3H),3.03(br.s.,1H),2.98(t,J=6.6Hz,2H),2.90-2.84(m,1H),2.77(s,3H),2.73-2.68(m,1H),2.65(dt,J=9.6,3.0Hz,1H),2.55-2.50(m,1H),2.32-2.20(m,2H),2.06(dt,J=11.4,2.4Hz,1H),1.85-1.76(m,3H),1.73(t,J=10.8Hz,1H),1.67-1.59(m,1H)。
生物测试例
本发明下述ITI-007如下所示,参照专利PCT/US2000/016498实施例261的方法进行制备,
Figure PCTCN2021122546-appb-000044
本发明下述对比例1-A如下所示,参照专利PCT/US2017/015178说明书[0339]段的方案1的方法进行制备,
Figure PCTCN2021122546-appb-000045
测试例1、各化合物对受体的功能活性测试(冻存细胞)
1、材料和设备
细胞
细胞名称 供应商
CHO-K1/D2L/Gα15 金斯瑞生物科技股份有限公司
CHO-K1/5HT2A 珀金埃尔默股份有限公司
实验试剂耗材
Figure PCTCN2021122546-appb-000046
仪器设备
Figure PCTCN2021122546-appb-000047
Figure PCTCN2021122546-appb-000048
测试药物
名称 供应商 型号
Dopamine Sigma H8502
Serotonin Sigma H9523
Spiperone Targetmol T0280
Ketanserin Targetmol T1066
2、实验方法
第一天:细胞铺板
(1)从液氮罐中取出细胞,拧松管盖并倒置使液氮从管中流出,然后再拧紧冻存管。
(2)冻存管在37℃迅速晃动,待冰全部融化之后,用75%酒精擦拭冻存管表面,放于生物安全柜中。
(3)将细胞悬液转移到预先加入10mL预热的生长培养基(不含筛选抗生素)的50mL离心管中。
(4)1000rpm离心5分钟。
(5)弃去上清,加入8mL生长培养基(不含筛选抗生素),轻轻吹散。取出20μL使用细胞计数仪进行计数。
(6)按照指定条件进行稀释:1×10 6个/mL,细胞板每孔加入20μL细胞悬液,最终每孔浓度依次为20000个/孔
(7)将细胞板置于5%CO 2、37℃的培养箱培养16-24小时。
第二天:实验检测
检测试剂准备:
(1)250mM的丙磺舒溶液配制:取1mL的实验缓冲液加入到含有77mg粉末的管中,涡旋振荡溶解。
(2)2×荧光探针溶液配制:取1瓶荧光探针干粉平衡至室温,加入10mL的实验缓冲液和200μL的250mM丙磺舒溶液,涡旋后静置5分钟确保完全溶解,然后再涡旋。
实验操作步骤:
(1)化合物准备。
a)激动剂化合物板准备(测试EC 80):将Dopamine和Serotonin在化合物板(Greiner- 781280)中使用实验缓冲液进行10个浓度点3倍稀释,起始浓度为1.2μM,每孔30μL,DMSO浓度不高于3%。
b)拮抗剂阳性化合物及待测化合物板准备:待测化合物在化合物板(Greiner-781280)中使用实验缓冲液进行10个浓度点3倍稀释,起始浓度为3μM,每孔30μL;拮抗阳性化合物Spiperone和Ketanserin使用实验缓冲液进行10个浓度点3倍稀释,起始浓度为1μM,每孔30μL。DMSO浓度不高于3%。
(2)细胞准备:从培养箱中取出细胞,加入20μL配制好的2×荧光探针溶液,37℃孵育50分钟然后室温平衡10分钟。
(3)激动剂EC 80测试
a)将细胞板和激动剂化合物板放置到仪器内,运行读板的程序。
b)从激动剂化合物板中转移10μL到细胞板中,并读取荧光信号。
c)使用Screenworks软件计算激动剂的EC 80,配制6×EC 80浓度备用。
(4)化合物EC 50和IC 50测试
a)将细胞板、待测化合物板放入仪器内,运行读板的程序。
b)从待测化合物板中转移10μL到细胞板中,并读取荧光信号。
c)然后再放入6×EC 80激动剂板,转移10μL到细胞板中,读取荧光信号。
d)导出从1-最大次数的“Max-Min”值作为原始数据进行分析,计算化合物的EC 50和IC 50值。
3、实验结果:具体结果见表1。
表1 各化合物体外功能活性测试结果
Figure PCTCN2021122546-appb-000049
4、实验结论:
目前研究发现,对5-HT2A受体的高活性可提高非典型和典型抗精神病药的临床疗效;另外,对D2/5-HT2A具有高选择性(D2/5-HT2A比值越大越具有高选择性)可降低诱发EPS反应的可能性,也是抗精神分裂症药的一个重要设计目标。
根据上表1的结果可知,本发明的化合物对5-HT2A和/或D2具有较好的功能活性,D2/5-HT2A比值较高,提示本发明的化合物具有良好的精神分裂症治疗效果,减少诱导产生EPS的可能性。
测试例2、对5-HT 2A受体的功能性检测实验(培养细胞)
1、实验目的:
通过细胞水平的钙离子内流检测方法检测本发明化合物对5-HT 2A受体的拮抗作用。
2、实验方案:
2.1实验材料:
(1)受试化合物:本发明实施例化合物,自制。
(2)对照化合物:Serotonin hydrochloride5-羟色胺(Sigma,H9523)、Ketanserin(Targetmol,T1066)。
(3)稳转细胞系:
Figure PCTCN2021122546-appb-000050
(4)实验试剂:
Figure PCTCN2021122546-appb-000051
Figure PCTCN2021122546-appb-000052
(5)实验耗材:
耗材名称 品牌 货号
细胞计数板 Countstar IC1000
Poly-D-Lysine coated 384 well plate Greiner 781946
384-well-PP microplate,V-Form Greiner 781280
Tera Pipette Tip Black,384 Molecular Device 9000-0764
(6)实验仪器:
仪器名称 品牌 型号
电子天平 Sartorius SQP
FLIPR Molecular Device TETRA
Bravo Agilent Bravo
细胞计数仪 Countstar BioTech
超纯水仪 Millipore IQ 7000
离心机 Cence TDZ5-WS
Multifuge X4R Pro离心机 Thermo Fisher 75009915
2.2溶液配制:
(1)受试化合物配制:将本发明实施例化合物分别用DMSO溶解配成10mM母液,储存至氮气柜备用。
(2)对照化合物配制:将对照化合物用DMSO溶解配成10mM母液,分装储存至-80℃冰箱备用。
(3)250mM的Probenecid溶液配制:取1mL的实验缓冲液加入到含有77mg粉末的管中,涡旋振荡溶解。
(4)2×荧光探针溶液配制:取1瓶荧光探针干粉平衡至室温,加入10mL的实验缓冲液和200μL的250mM probenecid溶液,涡旋后静置5分钟确保完全溶解。
(5)实验缓冲液HBSS:HBSS与HEPES以50:1进行混匀,置于4℃冰箱保存,用时37℃加热即可。
2.3实验方法:
细胞复苏:
(1)从液氮罐中取出细胞,于37℃水浴锅中融化之后,用75%酒精擦拭冻存管表面,置于生物安全柜中。
(2)将细胞悬液转移至4mL预热的铺板培养基的15mL离心管中,1000rpm离心5分钟。
(3)弃去上清,加入10mL铺板培养基,轻轻吹散,转移至培养皿中生长。待细胞24h贴壁后更换为生长培养基继续培养。
细胞铺板:
(1)取对数期生长的细胞,生长至85%-90%左右,进行如下操作:细胞清洗、消化、终止,随后转移至15mL离心管,在室温条件下,1000rpm离心5min。
(2)弃去上清,加入一定的铺板培养基,轻轻吹散。取出20μL细胞悬液进行计数。
(3)将细胞稀释到1×10 6个/mL,细胞板(Greiner-781946)中每孔加入20μL细胞悬液,使得每孔的密度为2×10 4个/孔。
(4)细胞板置于5%CO 2、37℃的培养箱培养16-24小时。
实验检测步骤:
(1)化合物准备
a)激动剂化合物板准备(测试EC 80):将激动剂在化合物板(Greiner-781280)中使用实验缓冲液进行10个浓度点4倍稀释,每孔30μL。
b)待测化合物及阳性化合物板准备:待测化合物和阳性化合物均在化合物板(Greiner-781280)中使用实验缓冲液进行10个浓度点4倍稀释,每孔30μL。
(2)细胞准备:从培养箱中取出细胞,加入20μL/well的2×荧光探针溶液,37℃孵育50分钟,室温平衡10分钟。
(3)激动剂的EC 80测试
a)将细胞板和激动剂化合物板放置到仪器内,运行程序。
b)从激动剂化合物板中转移10μL到细胞板中,并读取荧光信号。
c)使用Screenworks软件计算激动剂的EC 80,配制6×EC 80浓度备用。
(4)化合物的IC 50测试
a)将细胞板和待测化合物板放入仪器内,运行相应程序。
b)从待测化合物板中转移10μL到细胞板中,并读取荧光信号。
c)取出化合物板,放入6×EC 80激动剂板,从6×EC 80激动剂板中转移10μL到细胞板中,读取荧光信号。
d)导出从1~最大次数读数的“Max-Min”值作为原始数据进行分析,计算化合物的IC 50值。
3、实验结果:如表2所示。
表2 本发明化合物对5-HT 2A受体的功能活性测试结果
实施例编号 5-HT 2A IC 50(nM)
实施例1 3.72
实施例1-A 0.78
实施例1-B 107.60
实施例4 19.69
实施例7 3.50
对比例1 44.36
对比例1-A 9.11
ITI-007 3.35
4、实验结论:
通过以上方案得出,本发明化合物对5-HT 2A受体具有较好的拮抗作用。
测试例3、本发明化合物对D 2L、D 2s受体的功能活性测试
1、实验目的:
通过细胞水平的环磷腺苷(cAMP)检测方法检测本发明化合物对D 2L、D 2s受体的拮抗作用。
2、实验方案
2.1实验材料:
(1)受试化合物:本发明实施例化合物,自制。
(2)对照化合物:Dopamine hydrochloride多巴胺盐酸盐(Sigma,H8502)、(+)-Butaclamol hydrochloride(+)-布他拉莫盐酸盐(Sigma,D033)。
(3)稳转细胞系:
Figure PCTCN2021122546-appb-000053
Figure PCTCN2021122546-appb-000054
(4)实验试剂:
试剂名称 品牌 货号
DMEM Gibco C11965500CP
PBS Gibco C10010500CP
0.05%Trypsin Invitrogen 25300062
DMSO Sigma D8418
cAMP Gi Assay Kit Cisbio 62AM9PEJ
IBMX Sigma I5879
HBSS Gibco 14175103
Forskolin Sigma F6886
(5)实验耗材:
Figure PCTCN2021122546-appb-000055
(6)实验仪器:
仪器名称 品牌 型号
电子天平 Sartorius SQP
EnVision PerkinElmer 2104
皮升级微量加样系统 Tecan D300e
Bravo Agilent Bravo
细胞计数仪 Countstar BioTech
超纯水仪 Millipore IQ 7000
离心机 Cence TDZ5-WS
2.2溶液配制:
(1)受试化合物配制:将本发明实施例化合物分别用DMSO溶解配成10mM母液,储存至氮气柜备用。
(2)对照化合物配制:将对照化合物用DMSO溶解配成10mM母液,分装储存至-80℃冰箱备用。
(3)IBMX使用DMSO溶解配制成0.5M储备液,冻存于-80℃。
(4)Forskolin使用DMSO溶解配制成1mM储备液,冻存于-80℃备用。
(5)实验缓冲液:用ddH 2O将5×stimulation buffer稀释成1×,并加入终浓度为0.5mM的IBMX,混匀备用。
2.3实验方法(Gi拮抗剂测试):
(1)化合物准备:受试化合物和阳性对照化合物(+)-Butaclamol hydrochloride在化合物板(Greiner-781280)中使用Bravo进行梯度稀释,工作液的起始浓度为20μM,采用实验缓冲液按4倍稀释10个浓度,1000rpm离心1分钟备用。
(2)冻存的D 2L细胞:从液氮罐中取出后于37℃水浴锅中解冻,将细胞悬液转移至含HBSS缓冲液的离心管中,750rpm离心5分钟。
培养的D 2S细胞:吸取培养基后用3mL的PBS清洗细胞,吸尽,用0.05%Trypsin进行消化3min,等体积的培养基终止,转移至离心管中,750rpm离心5分钟。吸尽液体后,再加入含HBSS缓冲液重悬,750rpm离心5分钟。
(3)弃去上清,沉淀用适量的实验缓冲液重悬,取20μL用于细胞计数。
(4)移取适量的细胞悬液,用实验缓冲液稀释至2×10 5个/mL,在细胞板(PerkinElmer-6008280)中加入5μL/well的细胞悬液,1000rpm离心1分钟。
(5)用Bravo转移5μL/well到细胞板中,1000rpm离心1分钟,细胞板封闭后室温孵育15分钟。
(6)用Tecan-D300e转移Forskolin到细胞板中,并转移终浓度为EC 90的Dopamine hydrochloride到细胞板中。
(7)细胞板于1000rpm条件下离心1分钟后封板,室温孵育45分钟。
(8)标准曲线:储备液浓度为2848nM的cAMP按起始最高浓度为712nM,8个点4倍连续稀释,取10μL/well加到细胞板中。
(9)加10μL/well含cAMP-d2和Anti-cAMP-Cryptate的检测溶液(按1:20用裂解缓冲液稀释)到细胞板中,1000rpm离心1分钟,室温避光孵育1小时。
(10)检测:细胞板于1000rpm离心1分钟,使用Envision进行读板(激发光340nm, 发射光620nm和665nm)。以两个通道信号的比值(665nm/620nm)乘以10000作为最后的原始数据进行分析,计算化合物的IC 50值。
3、实验结果:如表3所示。
表3 本发明化合物对D 2L、D 2s受体的功能活性测试结果
实施例编号 D 2L IC 50(nM) D 2s IC 50(nM)
实施例1 20.28 95.44
实施例1-A 30.86 261.00
实施例1-B 15.53 63.07
实施例3 51.42 207.10
实施例4 13.40 82.83
实施例5 13.11 93.37
实施例7 40.84 294.60
对比例1 199.70 363.60
对比例1-A 88.77 264.30
对比例2 35.77 622.40
ITI-007 131.30 343.70
4、实验结论:
通过以上方案得出,本发明化合物对D 2L、D 2s受体均具有较好的拮抗作用。
测试例4、本发明化合物在ICR小鼠上的PK试验
1、试验目的:
对雄性ICR小鼠进行经口灌胃给药,测定本发明化合物在小鼠中的血药浓度,计算PK参数,对本发明化合物进行药代动力学评价。
2、试验材料:
(1)试验药品:本发明实施例化合物,自制。
(2)试验动物:ICR小鼠,SPF级,雄性,上海斯莱克实验动物有限责任公司。
(3)主要试验仪器:
Figure PCTCN2021122546-appb-000056
Figure PCTCN2021122546-appb-000057
(4)主要试验试剂:
Figure PCTCN2021122546-appb-000058
3、试验方案:
(1)给药信息:
药物配制:取受试化合物,加入生理盐水并进行超声。
给药途径及剂量:经口灌胃给药,给药剂量:5mg/kg,给药体积:10mL/kg。
给药频率及期限:单次给药。
(2)试验方法:
将ICR小鼠按体重分层后随机分组,每组3只小鼠,试验前过夜禁食。分别经口灌胃给药,在0、0.033、0.083、0.5、1、2、4、6和8小时,于小鼠下颌静脉或隐静脉取血250μL至含有抗凝剂肝素钠的样品管并置于湿冰中,4000r·min -1离心10min,分离血浆,冷冻保存在-80℃冰箱中待测。
4、试验结果与分析:
将灌胃所测得的本发明化合物的血药浓度-时间数据代入Winnonlin 8.2程序计算主要药动学参数。T max和C max采用实测值,采用梯形法计算AUC 0-t值和AUC 0-∞值,以半对数作图法,由消除相末端浓度点计算t 1/2。具体结果如表4所示。
表4 小鼠药代实验结果
Figure PCTCN2021122546-appb-000059
Figure PCTCN2021122546-appb-000060
5、试验结论:
从表中小鼠药代实验结果可以看出,本发明化合物给药后能迅速吸收,表现出良好的代谢性质,暴露量AUC和最大血药浓度C max都表现良好。
测试例5、本发明化合物对MK-801诱导小鼠高活动行为影响的试验
1、实验目的:
通过腹腔注射MK-801导致小鼠高活动模型,对本发明化合物进行药效评价。
2、实验方案
(1)实验材料:
受试化合物:本发明实施例化合物,自制。
MK-801(地卓西平马来酸盐,(+)-MK-801 hydrogen maleate),SIGMA,M107-250MG。
溶媒:纯水,广州屈臣氏食品饮料有限公司,20200928C;
生理盐水,山东华鲁制药有限公司,SD20080806。
(2)实验主要仪器:
Figure PCTCN2021122546-appb-000061
(3)实验动物:
实验动物:ICR小鼠,雄性,8只/组,斯贝福(北京)生物科技有限公司。
(4)给药信息:
药物配制:取受试化合物,加入纯水并进行超声。
给药途径及方法:灌胃给药,10mL/kg体重。
给药频率及期限:单次给药。
将动物按体重分层后随机分为空白组,模型组,及给药组,详细给药信息见下表:
Figure PCTCN2021122546-appb-000062
Figure PCTCN2021122546-appb-000063
(5)实验方法:
小鼠按照体重分层后随机分为模型组、空白组及各给药组。在灌胃给予溶媒或药物后1h,腹腔注射0.3mg/kg的MK-801(空白组注射等体积的生理盐水),再将小鼠放入自主活动箱(规格为29cm×29cm×30cm的黑色聚乙烯箱)进行录像,录像时间为60min,录像结束进行视频分析,评价小鼠活动情况。
(6)数据处理和统计:
实验数据用
Figure PCTCN2021122546-appb-000064
表示,采用SPSS统计软件,先进行方差齐性检验,若方差齐则进行单因素方差分析,两两比较采用Dunnett t检验。用GraphPad Primis5软件以非线性拟合的方法计算ED 50
3、实验结果:如表5所示。
表5 本发明化合物对MK-801诱导小鼠高活动行为影响的试验结果
Figure PCTCN2021122546-appb-000065
注:ED 50为半数有效量,MED为最低起效剂量。
4、实验结论:
通过以上方案得出,本发明的化合物可明显抑制MK-801诱导的高活动,且相对于对比例,本发明的化合物最低起效剂量更低,抑制效应更强。
测试例6、本发明化合物对DOI诱导小鼠甩头行为影响的试验
1、实验目的:
通过腹腔注射(±)DOI(一种致幻剂,常用于复制抗精神分裂症动物模型)诱导小鼠甩头行为,对本发明化合物进行药效评价。
2、实验方案:
(1)实验材料:
受试化合物:本发明实施例化合物,自制。
(±)-DOI hydrochloride,SIGMA,D101-100MG。
溶媒:纯水,广州屈臣氏食品饮料有限公司,20200928C;
生理盐水,山东华鲁制药有限公司,SD20080806。
(2)实验主要仪器:
Figure PCTCN2021122546-appb-000066
(3)实验动物:
实验动物:ICR小鼠,雄性,8只/组,斯贝福(北京)生物科技有限公司。
(4)给药信息:
药物配制:取受试化合物,加入溶媒并进行超声。
给药途径及方法:经口灌胃给药,10mL/kg体重。
给药频率及期限:单次给药。
将动物按体重分层后随机分为空白组,模型组,及给药组,详细给药信息见下表:
Figure PCTCN2021122546-appb-000067
(5)实验方法:
小鼠按照体重分层后随机分为模型组、空白组及各给药组。动物灌胃给予溶媒或药 物后1h,将动物放入铺有新鲜垫料的烧杯内(直径为13cm,高19cm),按照1mg/kg的剂量腹腔注射造模药DOI,记录小鼠腹腔注射DOI后第0-20分钟内甩头的次数。
甩头行为定义为小鼠头部迅速的旋转性抽动或湿狗样抖动,该动作要区别于正常的理毛或探究行为。
(6)数据处理和统计:
实验数据用
Figure PCTCN2021122546-appb-000068
表示,采用SPSS统计软件,先进行方差齐性检验,若方差齐则进行单因素方差分析,两两比较采用Dunnett t检验。用GraphPad Primis5软件以非线性拟合的方法计算ED 50
3、实验结果:如表6所示。
表6 本发明化合物对DOI诱导小鼠甩头行为影响的试验结果
Figure PCTCN2021122546-appb-000069
注:ED 50为半数有效量。
4、实验结论:
通过以上方案得出,本发明的化合物可明显抑制DOI诱导的甩头行为。
尽管上面已经示出和描述了本发明的实施例,可以理解的是,上述实施例是示例性的,不能理解为对本发明的限制,本领域的普通技术人员在本发明的范围内可以对上述实施例进行变化、修改、替换和变型。

Claims (15)

  1. 一种如通式(I)所示的化合物、其立体异构体或其可药用的盐:
    Figure PCTCN2021122546-appb-100001
    其中,
    R 1独立地为被卤素,C1-C3的烷氧基,氰基,氨基,硝基,羟基,C2-C4的烯基,C2-C4的炔基,C3-C5的环烷基任选取代的C1-C6的烷基;
    R 2独立地为-R 7-R 8-R 9-;
    R 3独立地为氢,或C1-C3的烷基中的任意一种;
    R 4独立地为-C(=O)-或-CH 2-中的任意一种;
    R 5和R 6各自独立地为氢,或被卤素、C1-C3的烷氧基、氰基、氨基、硝基、羟基、C2-C4的烯基、C2-C4的炔基任选取代的C1-C3的烷基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-;
    R 7独立地为C1-C5的亚烃基;
    R 8独立地为-C(=O)-,-CH 2-或-O-中的任意一种;
    R 9独立地为C1-C3的亚烃基或不存在中的任意一种;
    A为被一个或多个卤素任选取代的苯基。
  2. 如权利要求1所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,其满足如下条件中的一种或多种:
    (1)所述R 1独立地为被卤素,C1-C3的烷氧基,氰基,氨基,硝基,羟基,C2-C4的烯基,C2-C4的炔基,C3-C5的环烷基任选取代的C1-C3的烷基;
    (2)所述R 3独立地为氢,或C1-C3的烷基中的任意一种;
    (3)所述R 5和R 6各自独立地为氢,或非取代的C1-C3的烷基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-;
    (4)A独立地为被一个或多个卤素取代的苯基;
    (5)R 4为-C(=O)-。
  3. 如权利要求2所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,其满足如下条件中的一种或多种:
    (1)所述R 1独立地为非取代的C1-C3的烷基;
    (2)所述R 3独立地为氢,或非取代的C1-C3的烷基中的任意一种。
  4. 如权利要求3所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,其满足如下条件中的一种或多种:
    (1)所述非取代的C1-C3的烷基选自甲基、乙基或丙基中的任意一种;
    (2)所述C1-C5的亚烃基选自C3-C5的亚烃基;
    (3)所述C1-C3的亚烃基选自C1-C2的亚烃基;
    (4)所述任选取代的C1-C3的烷基中的C1-C3的烷基选自甲基、乙基或丙基中的任意一种;
    (5)所述卤素为氟,氯,溴或碘中的任意一种。
  5. 如权利要求1-4任一项所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,所述通式(I)所示的化合物选自如下所示通式(I-A)所示的化合物:
    Figure PCTCN2021122546-appb-100002
    其中,
    X为卤素;所述X在苯环的任意可取代位置进行取代;X为单取代或多取代的任意一种情形,优选单取代;
    所述卤素为氟,氯,溴或碘中的任意一种;
    R 1,R 3,R 4,R 5,R 6,R 7,R 8和R 9如权利要求1-4任一项所定义。
  6. 如权利要求5所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,所述通式(I)所示的化合物选自如下所示通式(I-B)所示的化合物:
    Figure PCTCN2021122546-appb-100003
  7. 如权利要求6所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,所述通式(I)所示的化合物选自如下所示通式(I-B)所示的化合物:
    Figure PCTCN2021122546-appb-100004
    其中,
    R 1独立地为甲基,乙基或丙基中的任意一种;
    R 3独立地为氢,甲基,乙基,或丙基中的任意一种;
    R 4独立地为-C(=O)-,或-CH 2-中的任意一种;
    R 5和R 6各自独立地为氢,甲基,或乙基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-;
    R 7独立地为-CH 2-CH 2-,-CH 2-CH 2-CH 2-或-CH 2-CH 2-CH 2-CH 2-中的任意一种;
    R 8独立地为-C(=O)-,-CH 2-或-O-中的任意一种;
    R 9独立地为-CH 2-CH 2-,-CH 2-或不存在中的任意一种;
    X独立地为氟或氯中的任意一种。
  8. 如权利要求1-7任一项所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,所述通式(I)所示的化合物选自如下所示通式(I-C)所示的化合物:
    Figure PCTCN2021122546-appb-100005
    其中,
    R 1独立地为甲基,乙基或丙基中的任意一种;
    R 3独立地为甲基,乙基或丙基中的任意一种;
    R 4独立地为-C(=O)-,或-CH 2-中的任意一种;
    R 5和R 6各自独立地为氢,甲基,或乙基中的任意一种,或R 5、R 6和与其直接相连的碳原子共同形成-C(=O)-。
  9. 如权利要求1-8所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,其特征在于,其满足如下条件中的一种或多种:
    (1)所述的通式(I)所示的化合物选自如下所示具体化合物的任意一个:
    Figure PCTCN2021122546-appb-100006
    Figure PCTCN2021122546-appb-100007
    (2)所述盐选自富马酸盐,马来酸盐,磷酸盐,硝酸盐,硫酸盐,苯磺酸盐,或草酸盐。
  10. 一种如通式(I-D)所示的化合物、其立体异构体或其可药用的盐:
    Figure PCTCN2021122546-appb-100008
    其中:
    R 1,R 3,R 4,R 5和R 6如权利要求1-9任一项或所定义。
  11. 一种制备如权利要求1-9所述的通式(I)所示的化合物、其立体异构体或其可药用的盐的方法,其特征在于,包括如下步骤:
    Figure PCTCN2021122546-appb-100009
    通式化合物(I-D)与通式化合物(I-E)通过亲核取代反应制备得到式(I)所示的化合物;
    其中:
    X 1为卤素,选自氟、氯、溴或碘,优选氯;
    R 1,R 2,R 3,R 4,R 5,R 6和A如权利要求1-9中任一项所定义。
  12. 一种药物组合物,其特征在于,包含治疗有效量的、如权利要求1-9中任一项所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,和药学上可接受的载体。
  13. 如权利要求1-9中任一项所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,或如权利要求12所述的药物组合物在制备涉及或调节5-羟色胺受体、5-羟色胺转运蛋白和/或多巴胺受体的药物中的用途;优选在制备涉及或调节5-HT2A受体、5-羟色胺转运蛋白、多巴胺D1受体和/或多巴胺D2受体的药物中的用途,更优选在制备涉及或调节5-HT2A受体和/或多巴胺D2受体的药物中的用途。
  14. 如权利要求1-9中任一项所述的通式(I)所示的化合物、其立体异构体或其可药用的盐,或如权利要求12所述的药物组合物在制备治疗神经精神类疾病的药物中的用途。
  15. 如权利要求14所述的用途,其特征在于,所述神经精神类疾病选自抑郁症、焦虑症、痴呆症、精神分裂症、睡眠障碍、运动障碍、痴呆症患者的行为障碍、帕金森病、阿尔茨海默病、偏头痛、多动症、强迫症、社交恐惧症、神经退行性疾病、双相情感障碍、创伤后应激综合征、成瘾性疾病、戒断综合征或注意力缺陷中的一种或多种,优选抑郁症、焦虑症、痴呆症、精神分裂症、睡眠障碍、运动障碍、痴呆症患者的行为障碍、神经退行性疾病或双相情感障碍中的任意一种或多种;所述的抑郁症例如重度抑郁症,所述的多动症例如注意力缺陷多动症。
PCT/CN2021/122546 2020-10-09 2021-10-08 杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用 WO2022073470A1 (zh)

Priority Applications (10)

Application Number Priority Date Filing Date Title
IL301847A IL301847A (en) 2020-10-09 2021-10-08 A modified heterocyclic γ-carboline derivative, a method of preparation thereof, its intermediate form and its use
JP2023546376A JP2023544653A (ja) 2020-10-09 2021-10-08 複素環置換の縮合γ-カルボリン誘導体、その製造方法、中間体及び使用
BR112023005443A BR112023005443A2 (pt) 2020-10-09 2021-10-08 Derivado de y-carbolina fundido substituído heterocíclico, método de preparação do mesmo, intermediário do mesmo e uso do mesmo
US18/027,226 US20230339949A1 (en) 2020-10-09 2021-10-08 HETEROCYCLIC SUBSTITUTED FUSED y-CARBOLINE DERIVATIVE, PREPARATION METHOD THEREFOR, INTERMEDIATE THEREOF AND USE THEREOF
KR1020237009354A KR20230054417A (ko) 2020-10-09 2021-10-08 헤테로사이클로로 치환된 융합 γ-카르볼린 유도체, 이의 제조 방법, 중간체 및 용도
EP21876988.3A EP4227306A4 (en) 2020-10-09 2021-10-08 HETEROCYCLIC SUBSTITUTED CONDENSED GAMMA-CARBOLINE DERIVATIVE, PREPARATION METHOD THEREFOR, RELATED INTERMEDIATE AND USE THEREOF
CA3194682A CA3194682A1 (en) 2020-10-09 2021-10-08 Heterocyclic substituted fused .gamma.-carboline derivative, preparationmethod therefor, intermediate thereof and use thereof
CN202180068356.7A CN116568302A (zh) 2020-10-09 2021-10-08 杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用
MX2023004102A MX2023004102A (es) 2020-10-09 2021-10-08 Derivado de gamma-carbolina fusionado sustituido con heterociclo, metodo de preparacion del mismo, intermediarios del mismo y uso del mismo.
AU2021356875A AU2021356875B2 (en) 2020-10-09 2021-10-08 HETEROCYCLIC SUBSTITUTED FUSED γ-CARBOLINE DERIVATIVE, PREPARATION METHOD THEREFOR, INTERMEDIATE THEREOF AND USE THEREOF

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202011075840.6 2020-10-09
CN202011075840 2020-10-09
CN202011074344 2020-10-09
CN202011074344.9 2020-10-09

Publications (1)

Publication Number Publication Date
WO2022073470A1 true WO2022073470A1 (zh) 2022-04-14

Family

ID=81125599

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/122546 WO2022073470A1 (zh) 2020-10-09 2021-10-08 杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用

Country Status (11)

Country Link
US (1) US20230339949A1 (zh)
EP (1) EP4227306A4 (zh)
JP (1) JP2023544653A (zh)
KR (1) KR20230054417A (zh)
CN (1) CN116568302A (zh)
AU (1) AU2021356875B2 (zh)
BR (1) BR112023005443A2 (zh)
CA (1) CA3194682A1 (zh)
IL (1) IL301847A (zh)
MX (1) MX2023004102A (zh)
WO (1) WO2022073470A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024088285A1 (zh) * 2022-10-26 2024-05-02 上海枢境生物科技有限公司 一种杂环取代的稠合γ-咔啉类衍生物的甲磺酸盐、晶型及其制备方法和应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105168219A (zh) * 2008-05-27 2015-12-23 细胞内治疗公司 用于睡眠障碍和其他疾病的方法和组合物
CN106535898A (zh) * 2014-04-04 2017-03-22 细胞内治疗公司 有机化合物
WO2017117514A1 (en) * 2015-12-31 2017-07-06 Tung Roger D Deuterated iti-007
CN110430879A (zh) * 2017-03-24 2019-11-08 细胞内治疗公司 新组合物和方法

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6552017B1 (en) * 1999-06-15 2003-04-22 Bristol-Myers Squibb Pharma Company Substituted heterocycle fused gamma-carbolines
BR112018015283B1 (pt) * 2016-01-26 2023-09-26 Intra-Cellular Therapies, Inc Compostos orgânicos, composição farmacêutica compreendendo os ditos compostos e uso dos mesmos para o tratamento ou profilaxia de um distúrbio do sistema nervoso central

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105168219A (zh) * 2008-05-27 2015-12-23 细胞内治疗公司 用于睡眠障碍和其他疾病的方法和组合物
CN106535898A (zh) * 2014-04-04 2017-03-22 细胞内治疗公司 有机化合物
WO2017117514A1 (en) * 2015-12-31 2017-07-06 Tung Roger D Deuterated iti-007
CN110430879A (zh) * 2017-03-24 2019-11-08 细胞内治疗公司 新组合物和方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LI, PENG ET AL.: "Discovery of a Tetracyclic Quinoxaline Derivative as a Potent and Orally Active Multifunctional Drug Candidate for the Treatment of Neuropsychiatric and Neurological Disorders", JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 6, 21 February 2014 (2014-02-21), pages 2670 - 2682, XP055545362, DOI: 10.1021/jm401958n *
See also references of EP4227306A4

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024088285A1 (zh) * 2022-10-26 2024-05-02 上海枢境生物科技有限公司 一种杂环取代的稠合γ-咔啉类衍生物的甲磺酸盐、晶型及其制备方法和应用

Also Published As

Publication number Publication date
CN116568302A (zh) 2023-08-08
AU2021356875A1 (en) 2023-05-25
AU2021356875B2 (en) 2023-11-23
BR112023005443A2 (pt) 2023-05-09
US20230339949A1 (en) 2023-10-26
KR20230054417A (ko) 2023-04-24
JP2023544653A (ja) 2023-10-24
IL301847A (en) 2023-06-01
EP4227306A1 (en) 2023-08-16
CA3194682A1 (en) 2022-04-14
EP4227306A4 (en) 2024-02-28
AU2021356875A9 (en) 2024-02-08
MX2023004102A (es) 2023-04-27

Similar Documents

Publication Publication Date Title
TWI542590B (zh) 1,2-雙取代雜環化合物
JP5102397B2 (ja) アリールメチルベンゾキナゾリノンm1レセプターポジティブアロステリックモジュレーター
TWI527800B (zh) 作為聚(二磷酸腺苷酸-核醣)聚合酶(parp)之抑制劑之1-(芳基甲基)喹唑啉-2,4(1h,3h)-二酮及其應用
TW202043212A (zh) Shp2抑制劑及其應用
WO2020156242A1 (zh) Shp2抑制剂及其应用
US20120178742A1 (en) Metabotropic glutamate receptor modulators
WO2022206723A1 (zh) 杂环类衍生物、其制备方法及其医药上的用途
CN114430739A (zh) Egfr抑制剂、组合物及其制备方法
CN102952118B (zh) 聚(adp-核糖)聚合酶抑制剂、制备方法及其用途
CN103619841A (zh) 杂芳基化合物及其使用方法
WO2019161803A1 (zh) 肽酰精氨酸脱亚胺酶抑制剂及其用途
WO2019076336A1 (zh) 含吡唑基的三并环类衍生物、其制备方法和应用
TWI444376B (zh) 脯胺醯胺吡啶化合物、其藥學組成物及醫藥用途
CN108341819B (zh) 磷酸二酯酶抑制剂及其用途
WO2022073470A1 (zh) 杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用
JP6345651B2 (ja) ヘテロアリール化合物及びその使用方法
TWI789217B (zh) 雜環取代的稠合γ-咔啉類衍生物、其製備方法、中間體及應用
TW202214634A (zh) 雜環化合物及其衍生物
WO2018205938A1 (zh) Parp抑制剂、其药物组合物、制备方法和应用
WO2019154329A1 (zh) 具有bet抑制活性的化合物及其制备方法和用途
WO2020200161A1 (zh) 含吲唑基的三并环类衍生物的盐及其晶型
CN117263944A (zh) 泛素特异性蛋白酶1抑制剂及其用途
WO2020083089A1 (zh) 五元或六元杂环并嘧啶类化合物及其用途
WO2022206724A1 (zh) 杂环类衍生物及其制备方法和用途
WO2024093956A1 (zh) 多环类聚(adp核糖)聚合酶选择性抑制剂

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: 21876988

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 20237009354

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3194682

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023005443

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 202180068356.7

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2021356875

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2023546376

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112023005443

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20230323

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021876988

Country of ref document: EP

Effective date: 20230509

ENP Entry into the national phase

Ref document number: 2021356875

Country of ref document: AU

Date of ref document: 20211008

Kind code of ref document: A