WO2024099393A1 - 一类含芳基的胺类化合物、其制备方法和用途 - Google Patents

一类含芳基的胺类化合物、其制备方法和用途 Download PDF

Info

Publication number
WO2024099393A1
WO2024099393A1 PCT/CN2023/130755 CN2023130755W WO2024099393A1 WO 2024099393 A1 WO2024099393 A1 WO 2024099393A1 CN 2023130755 W CN2023130755 W CN 2023130755W WO 2024099393 A1 WO2024099393 A1 WO 2024099393A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
formula
membered
added
Prior art date
Application number
PCT/CN2023/130755
Other languages
English (en)
French (fr)
Inventor
沈敬山
艾萨·阿吉·艾克拜尔
何洋
纪靖
朱维良
张勇
吴春晖
田广辉
胡天文
奥布力·麦麦提艾力
Original Assignee
中国科学院上海药物研究所
苏州旺山旺水生物医药股份有限公司
中国科学院新疆理化技术研究所
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股份有限公司, 中国科学院新疆理化技术研究所 filed Critical 中国科学院上海药物研究所
Publication of WO2024099393A1 publication Critical patent/WO2024099393A1/zh

Links

Definitions

  • the present invention belongs to the field of pharmaceutical chemistry. Specifically, the present invention relates to an aromatic amine compound represented by formula (I), a preparation method and a pharmaceutical composition thereof, and an application thereof in the preparation of a drug for preventing or treating diseases associated with NMDA receptors and/or monoamine transporters and/or sigma receptors, in particular central nervous system diseases.
  • an aromatic amine compound represented by formula (I) represented by formula (I)
  • a preparation method and a pharmaceutical composition thereof and an application thereof in the preparation of a drug for preventing or treating diseases associated with NMDA receptors and/or monoamine transporters and/or sigma receptors, in particular central nervous system diseases.
  • Depression is a serious mental illness, which is mainly manifested by depression, low self-esteem, and even suicidal tendencies.
  • the main drugs for treating depression are SSRI/SSNI (Selective serotonin/norepinephrine reuptake inhibitors).
  • SSRI/SSNI Selective serotonin/norepinephrine reuptake inhibitors.
  • this type of drug has no effect on nearly one-third of patients, and generally has defects such as slow onset of effect and even increased suicidal tendencies in patients. There is a huge unmet clinical need in this field.
  • NMDA N-methyl-D-aspartate glutamate receptor
  • NMDAR N-methyl-D-aspartate glutamate receptor
  • This receptor can be activated by glutamate, the most important excitatory neurotransmitter in the central nervous system, in vivo, thereby mediating the conduction of excitatory signals between synapses.
  • the permeability to cations such as Ca2+ , K + , and Na + is enhanced, generating excitatory postsynaptic potentials and triggering a series of physiological and biochemical reactions.
  • the molecular structure of NMDAR is complex, and various subtypes are specific in spatiotemporal distribution and pharmacological properties. Its number, composition, and distribution show dynamic changes in different developmental periods and different brain regions, participating in many physiological activities, providing a molecular basis for complex neural activities, and thus ensuring the normal activity of neural networks.
  • the integration, localization, recycling, and distribution of NMDAR inside and outside the synapse depend on the regulation of neural activity.
  • the disruption of its functional homeostasis is highly related to many brain diseases, such as depression, epilepsy, and schizophrenia.
  • Monoamine transporters are a class of proteins found on the cell membranes of neurons in the central and peripheral nervous systems that are responsible for transporting common neurotransmitters in synaptic transmission.
  • Monoamine transporter inhibitors are commonly used as antidepressants to treat depression and anxiety disorders, such as bupropion, which has norepinephrine transporter and dopamine transporter inhibitory effects, and fluoxetine and citalopram, which have 5-HT transporter inhibitory effects.
  • Sigma receptors are mainly located in the central nervous system and also in the endocrine and immune systems.
  • Sigma1 receptor agonists may have antidepressant and learning and memory promoting effects, while antagonists may have antipsychotic and analgesic effects.
  • Sigma2 receptors are ideal molecular targets for the treatment of central nervous system diseases such as Alzheimer's disease, schizophrenia and traumatic brain injury, and both sigma2 receptor agonists and antagonists are potential therapies for neurodegenerative diseases.
  • NMDA receptor antagonist ketamine has been used in medicine as a good anesthetic for more than 50 years. Studies have found that intravenous subanesthetic doses of NMDAR antagonist ketamine can quickly relieve symptoms of depression within hours, and the effect can last for at least a week. Esketamine developed by Johnson & Johnson was approved by the FDA in 2019 for the treatment of refractory depression. Given that the main molecular target of ketamine is NMDAR, many people have proposed that inhibiting this target is also the reason for ketamine's antidepressant effect. This mechanism suggests that ketamine's antidepressant effect and dissociative side effects may be inseparable at the mechanistic level. However, much evidence has questioned this hypothesis (J Psychiatry Neurosci.
  • the purpose of the present invention is to provide a novel aromatic amine compound having NMDA receptor and/or monoamine transporter and/or sigma receptor regulating effect, and a preparation method and use thereof.
  • An object of the present invention is to provide an aromatic amine compound represented by formula (I), or a stereoisomer, geometric isomer, conformational isomer, tautomer, pharmaceutically acceptable salt, polymorph, solvate, hydrate or isotope-labeled compound thereof.
  • Another object of the present invention is to provide a method for preparing the aromatic amine compound represented by formula (I).
  • Another object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of one or more of a compound selected from the group consisting of formula (I), its stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds, and optionally one or more pharmaceutically acceptable carriers, diluents or excipients.
  • Another object of the present invention is to provide a compound selected from the group consisting of formula (I), its stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds, or the use of the above-mentioned pharmaceutical compositions in the preparation of drugs for regulating the activity of NMDA receptors and/or monoamine transporters and/or sigma receptors.
  • Another object of the present invention is to provide a compound selected from the group consisting of formula (I), its stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds, or the use of the above-mentioned pharmaceutical compositions in the preparation of drugs for preventing and/or treating diseases associated with NMDA receptors and/or monoamine transporters and/or sigma receptors, in particular central nervous system diseases.
  • Ring A is selected from a 4-10 membered heterocycle or a C6-C10 aromatic ring and a 4-10 membered heterocycle containing 1-3, for example 1, 2 or 3 heteroatoms selected from N, O and S;
  • the 4-10 membered heterocycle is, for example, a 5-, 6-, 7-, 8- or 9-membered heterocycle, preferably a 5-8 membered heterocycle;
  • the heterocycle in ring A contains 1 N atom and 1 S atom, or contains two N atoms, or contains 1 S atom, or contains two N atoms and 1 S atom, or contains 1 N atom and 1 O atom;
  • each R 5 is independently selected from halogen, hydroxyl, amino, cyano, carboxyl, oxo, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy, C1-C6 alkanoyl, carbamoyl (-CONH 2 ), carbamoyl substituted by C1-C6 alkyl, amino substituted by one or two C1-C6 alkyl, amino substituted by one or two halogenated C1-C6 alkyl, amino substituted by one or two C1-C6 alkanoyl, C1-C6 alkoxycarbonyl, C3-C6 cycloalkyl, 4-10 membered heterocycloalkyl, C6-14 aryl, 5-10 membered heteroaryl, C6-C14 arylC1-C6 alkyloxy or 5-10 membered heteroarylC1-C6
  • Ring B is a 3-10 membered carbocyclic ring; preferably a 5-8 membered carbocyclic ring, such as a 5-, 6-, 7- or 8-membered carbocyclic ring, more preferably a 5-, 6- or 7-membered carbocyclic ring;
  • x is an integer from 0 to 2, such as 0, 1 or 2;
  • R2 and R3 are each independently selected from hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy C1-C6 alkyl, hydroxy C1-C6 alkyl, C6-C14 aryl C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkanoyl, halogenated C1-C6 alkanoyl, C3-C6 cycloalkyl C1-C6 alkanoyl, C6-C14 aryl C1-C6 alkanoyl, C1-C6 alkylsulfonyl, C1-C6 alkylsulfoxide or C3-C6 cycloalkyl C1-C6 alkyl; preferably each independently selected from hydrogen, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy C1-C4 alkyl,
  • R 2 , R 3 and the connected nitrogen atom together form a 3-9 membered heterocycloalkyl group
  • the 3-9 membered heterocycloalkyl ring optionally contains one or more additional nitrogen atoms or oxygen atoms
  • the 3-9 membered heterocycloalkyl group is optionally substituted by one or more C1-C6 alkyl groups, preferably C1-C4 alkyl groups
  • R 2 , R 3 and the connected nitrogen atom together form an azetidinyl group, a pyrrolidinyl group, a piperidinyl group, an azepanyl group, or a morpholinyl group;
  • R 1 is selected from C6-C14 aryl, 5-10 membered heteroaryl, 4-10 membered heterocyclyl, C6-C10 aryl and 4-10 membered carbocyclic ring (e.g., indanyl) or C6-C10 aryl and 4-10 membered heterocyclic ring (e.g., 1,2-methylenedioxyphenyl, 2,3-dihydrobenzofuranyl), preferably selected from C6-C14 aryl, 5-10 membered heteroaryl, more preferably phenyl, naphthyl, quinolyl, isoquinolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, thienyl or thiazolyl, further preferably phenyl, naphthyl, pyrimidinyl, pyridyl, pyrazinyl, pyridazinyl or quinolyl, further preferably
  • the C6-C14 aryl, 5-10 membered heteroaryl, 4-10 membered heterocyclyl, C6-C10 aryl and 4-10 membered carbocyclic ring or C6-C10 aryl and 4-10 membered heterocyclic ring is optionally substituted by one or more R 6 ;
  • R 6 is each independently selected from halogen, hydroxyl, mercapto, cyano, carbamoyl (NH 2 CO-), aminosulfonyl (NH 2 SO 2 -), C1-C6 alkyl, halogenated C1-C6 alkyl, hydroxy C1-C6 alkyl, C3-C6 cycloalkyl, C3-C6 cycloalkyloxy, C3-C6 cycloalkyl substituted by C1-C3 alkyl, C3-C6 cycloalkyloxy substituted by C1-C3 alkyl, C1-C6 alkoxy, C1-C6 alkylthio, halogenated C1-6 alkoxy, C3-C6 cycloalkyl C1-C6 alkoxy, C1-C6 alkanoyloxy, C6-C14 aryl, 5-10 membered heteroaryl, C6-C14 aryl C1-C6 alkoxy, 5-10
  • n is an integer from 0 to 3, such as 0, 1, 2 or 3;
  • R4 is connected to the B ring except R1 and On any carbon atom other than the commonly connected carbon atom, and R 4 is independently selected from hydroxyl, halogen, C1-C6 alkyl.
  • the compound of formula (I) is selected from compounds of formula (IA)-(ID):
  • ring A, R 1 , m, R 4 , R 2 , R 3 and x are as defined above.
  • the A ring is selected from a 5-membered or 6-membered heterocyclic ring or a benzo 5-membered or 6-membered heterocyclic ring.
  • the A ring is selected from:
  • each R 7 is independently selected from hydrogen, halogen, cyano, hydroxyl, amino, carbamoyl (-CONH 2 ), carbamoyl substituted by C1-C6 alkyl, carboxyl, C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkyl, amino substituted by C1-C6 alkyl, amino substituted by halogenated C1-C6 alkyl, amino substituted by C1-C6 alkanoyl, C1-C6 alkanoyl, C1-C6 alkoxycarbonyl, C3-C6 cycloalkyl, 4-8 membered heterocycloalkyl, C6-10 aryl, 5-10 membered hetero
  • Ring A is selected from:
  • Each R 7 is independently selected from hydrogen, halogen, cyano, hydroxyl, amino, carbamoyl (-CONH 2 ), carbamoyl substituted by C1-C6 alkyl, carboxyl, C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkyl, amino substituted by C1-C6 alkyl, amino substituted by halogenated C1-C6 alkyl, amino substituted by C1-C6 alkanoyl, C1-C6 alkanoyl, C1-C6 alkoxycarbonyl, C3-C6 cycloalkyl, C3-C6 cycloalkyl, 4-8 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C6-C10 arylC1-C4 alkyloxy or 5-10 membered heteroarylC1-C4 alkyloxy; preferably, each R 7 is independently selected from hydrogen
  • R 1 is phenyl optionally substituted with one or more R 6 ; R 6 is as defined above.
  • R 1 is naphthyl optionally substituted with one or more R 6 ;
  • R 6 is as defined above.
  • one of R 2 and R 3 is hydrogen, or both R 2 and R 3 are hydrogen.
  • R 1 is phenyl optionally substituted with one or more R 6 , and one of R 2 and R 3 is hydrogen, or both R 2 and R 3 are hydrogen.
  • R 1 is naphthyl optionally substituted with one or more R 6
  • one of R 2 and R 3 is hydrogen, or both R 2 and R 3 are hydrogen.
  • Ring B is a 6-membered carbocyclic ring;
  • R 1 is phenyl optionally substituted with one or more R 6 ;
  • one of R 2 and R 3 is hydrogen, or both R 2 and R 3 are hydrogen.
  • Ring B is a 6-membered carbocyclic ring; R 1 is naphthyl optionally substituted with one or more R 6 ; one of R 2 and R 3 is hydrogen, or both R 2 and R 3 are hydrogen.
  • Ring B is a 5-membered carbocyclic ring; R 1 is phenyl optionally substituted with one or more R 6 ; R 2 and R 3 are both hydrogen.
  • Ring B is a 7-membered carbocyclic ring; R 1 is phenyl optionally substituted with one or more R 6 ; R 2 and R 3 are both hydrogen.
  • the compound of formula (I) is selected from the following compounds:
  • the compound of formula (I) is selected from the following compounds:
  • R 2 , R 3 , R 6 , and R 7 are as defined above;
  • phenyl, pyridyl, naphthyl, quinolyl, pyrimidyl, pyrazinyl or pyridazinyl and -NR 2 R 3 are attached to the same ring carbon atom of the carbocyclic ring to which they are attached.
  • the compound of formula (I) is selected from the following compounds:
  • R 2 , R 3 , R 6 and R 7 are as defined above.
  • halogen generally refers to fluorine, chlorine, bromine and iodine; preferably fluorine, chlorine or bromine; more preferably fluorine or chlorine.
  • alkyl refers to a straight-chain or branched saturated hydrocarbon group
  • a C1-C6 alkyl group refers to a straight-chain or branched saturated hydrocarbon group containing 1 to 6 carbon atoms, for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-ethylpropyl, isopentyl, neopentyl, isohexyl, 3-methylpentyl or n-hexyl, etc., preferably methyl, ethyl, n-propyl, isopropyl, butyl or isobutyl.
  • halogenated C1-C6 alkyl refers to a linear or branched saturated hydrocarbon group containing 1 to 6 carbon atoms, wherein the hydrogen atoms are replaced by one or more halogen atoms which are the same or different.
  • halogenated C1-C4 alkyl for example, trifluoromethane 1-fluorobutyl, ...
  • C1-C6 alkoxy refers to a straight or branched alkoxy group containing 1 to 6 carbon atoms, “C1-C4 alkoxy”, “C1-C3 alkoxy” and so on, for example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, sec-butoxy, n-pentoxy, isopentoxy, neopentoxy, isohexoxy, 3-methylpentoxy or n-hexoxy, etc., preferably methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy or isobutoxy.
  • halogenated C1-C6 alkoxy refers to a linear or branched alkoxy group containing 1 to 6 carbon atoms in which the hydrogen atoms are substituted by one or more halogen atoms which are the same or different.
  • halogenated C1- C4 alkoxy for example, -OCF3 , -OCH2CH2Cl , -OCHBrCH2Cl , or -OCF2CF3 .
  • C1-C6 alkanoyl refers to a straight chain or branched alkanoyl group of 1 to 6 carbon atoms, “C1-C4 alkanoyl”, “C1-C3 alkanoyl” and so on, for example, formyl, acetyl, propionyl, butyryl, isobutyryl, valeryl and the like, preferably formyl, acetyl and propionyl.
  • halogenated C1-C6 alkanoyl refers to a straight or branched chain alkanoyl group of 1 to 6 carbon atoms in which the hydrogen atoms are replaced by one or more same or different halogen atoms, and the same shall apply to “halogenated C1-C4 alkanoyl”, “halogenated C1-C3 alkanoyl” and so on.
  • amino group substituted by C1-C6 alkyl group means that the hydrogen atom on the amino group is substituted by 1 or 2 identical or different C1-C16 alkyl groups, such as -NHMe, -NHEt, -N(Me)Et or -NEt2 , etc.
  • amino group substituted by C1-C6 alkanoyl group means that the hydrogen atom on the amino group is substituted by 1 or 2 identical or different C1-C6 alkanoyl groups, such as -NHCHO, -NHCOCH 3 or -NHCOCH 2 CH 3 .
  • C1-C6 alkoxy C1-C6 alkyl refers to a C1-C6 alkoxy group in which the oxygen atom is connected to a C1- C6 alkyl group , such as -CH2OCH2CH3 , -CH2CH2OCH2CH3 or -CH2CH2OCH3 .
  • C3-C6 cycloalkyl refers to a saturated cyclic hydrocarbon group containing 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
  • hydroxy C1-C6 alkyl refers to a straight or branched alkyl group containing 1 to 6 carbon atoms in which one carbon atom is connected to a hydroxy group, such as -CH2OH , -CH2CH2OH , -CH ( OH ) CH3 , -CH2CH2CH2OH , -CH2CH2CH2CH2OH , or -CH2CH ( CH3 ) CH2OH .
  • C6-C14 aryl refers to a monocyclic or cyclic ring containing 6 to 14 ring atoms but no heteroatoms.
  • 3-10 membered carbocycle refers to a saturated or unsaturated monocyclic hydrocarbon group containing 3-10 carbon atoms.
  • the carbocycle preferably has 5-8 ring carbon atoms, more preferably 5-6 carbon atoms, such as cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexene, cycloheptane, cycloheptene, etc.
  • C6-C14 aryl C1-C6 alkyl refers to a C6-C14 aryl group connected to a C1-C6 alkyl group, for example, benzyl, phenethyl, phenylpropyl, and the like.
  • C3-C6 cycloalkyl C1-C6 alkanoyl refers to a C3-C6 cycloalkyl group in which the ring carbon is connected to the alkyl carbon of a C1-C6 alkanoyl group, for example, cyclopropylcarbonyl, cyclopropylacetyl, cyclobutylcarbonyl, cyclopentylcarbonyl, and the like.
  • C6-C14 aryl C1-C6 alkanoyl refers to a C6-C14 aryl group connected to the alkyl carbon of a C1-C6 alkanoyl group, for example, benzoyl, phenylacetyl, and the like.
  • C3-C6 cycloalkyl C1-C6 alkyl refers to a C3-C6 cycloalkyl group connected to a C1-C6 alkyl group, for example, cyclopropylmethyl, cyclobutylmethyl and the like.
  • heterocycle refers to a monocyclic or polycyclic group containing at least one heteroatom selected from N, O and S as a ring member, which may be an aromatic or non-aromatic group, and is preferably a monocyclic group herein;
  • a 4-10 membered heterocycle refers to a heterocyclic group containing 4 to 10 ring atoms, such as pyridyl, piperidyl, morpholinyl, furanyl, thienyl, thiazolyl, imidazolyl, pyrrolyl, pyrazine, pyridazine, and pyrimidinyl.
  • heterocycloalkyl refers to a saturated monocyclic or polycyclic group containing at least one heteroatom selected from N, O and S as a ring member.
  • 3-9 membered heterocycloalkyl groups include azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, morpholinyl and the like.
  • heteroaryl refers to a monocyclic or bicyclic aromatic ring group containing at least one heteroatom selected from nitrogen, oxygen or sulfur as a ring member; 5-10 membered heteroaryl groups include but are not limited to the following groups: pyrrolyl, imidazolyl, pyrazolyl, 1,2,3-triazolyl, pyridyl, pyridonyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, quinolyl and the like.
  • optional means that the event described subsequently may or may not occur, and the description includes both instances where the event occurs and instances where the event does not occur.
  • optionally substituted alkyl includes “unsubstituted alkyl” and “substituted alkyl” as defined herein.
  • Optionally substituted with halogen includes both instances of “substituted with halogen” and instances of "not substituted with halogen", for example, substituted with 0-3 halogens.
  • formula (I) when formula (I) is mentioned herein, the term also includes its sub-formulas, such as formula (I-1-a), (I-1-b), (I-2-a), (I-2-b) or (II-1) (II-2) and the like.
  • the aromatic amine compounds represented by the general formula (I) of the present invention and their geometric isomers, conformational isomers, and tautomers also include solvate forms, such as hydrates, alcoholates, etc., and the solvates are also included in the scope of the present invention.
  • the pharmaceutically acceptable salts of the heterocyclic compounds represented by the general formula (I) of the present invention and their geometric isomers, conformational isomers, and tautomers refer to the aromatic amine compounds represented by the general formula (I) or their stereoisomers treated with appropriate acids to convert them into therapeutically active non-toxic addition salt forms.
  • Said salts are, for example, hydrochloride, hydrobromide, hydroiodide, sulfate or bisulfate, nitrate, phosphate or acid phosphate, perchlorate, format, acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, succinate, glutarate, maleate, fumarate, lactate, malate, citrate, tartrate, picrate, glutamate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, salicylate, ascorbate, camphorate or camphorsulfonate, etc.
  • the salt form can also be converted into the free base form by treatment with alkali.
  • solvates include, for example, hydrates, alcoholates, and the like.
  • Stepoisomers refer to isomers produced by different spatial arrangements of atoms in molecules. They can be divided into cis-trans isomers and enantiomers, or they can be divided into two major categories: enantiomers and diastereomers.
  • Enantiomers are a pair of stereoisomers that are non-superimposable mirror images of each other.
  • a 1 : 1 mixture of a pair of enantiomers is a “racemic” mixture. The term is used to refer to a racemic mixture when appropriate.
  • the conventional RS system is used to designate single stereoisomers with known relative and absolute configurations of two chiral centers (e.g., (1S, 2S)); single stereoisomers with known relative configurations but unknown absolute configurations are marked with an asterisk (e.g., (1R*, 2R*)); racemates with two letters (e.g., (1RS, 2RS) is a racemic mixture of (1R, 2R) and (1S, 2S); (1RS, 2SR) is a racemic mixture of (1R, 2S) and (1S, 2R)).
  • "Diastereomers” are stereoisomers that have at least two asymmetric atoms but are not mirror images of each other.
  • Absolute stereochemistry is indicated according to the Cahn-lngold-Prelog RS system.
  • the stereochemistry at each chiral carbon can be specified by R or S.
  • Resolved compounds whose absolute configuration is unknown can be assigned (+) or (-) based on the direction (right-handed or left-handed) that they rotate plane polarized light at the wavelength of the sodium D line.
  • resolved compounds can be identified by the corresponding enantiomer.
  • the diastereomers were defined by their respective retention times on chiral HPLC.
  • Geometric isomers can occur when a compound contains a double bond or some other feature that imparts a certain amount of structural rigidity to the molecule. If the compound contains a double bond, the substituents can be in the E or Z conformation. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituents can have either the cis or trans configuration.
  • Conformational isomers are isomers that differ by rotation about one or more valence bonds.
  • Tautomer refers to an isomer formed when a proton is shifted from one atom of a molecule to another atom of the same molecule. All tautomeric forms of the compounds of the present invention are intended to be encompassed within the scope of the present invention.
  • Polymorph refers to crystalline forms having the same chemical structure/composition but different spatial arrangements of the molecules and/or ions that form the crystals.
  • the compounds of the invention may be provided as amorphous solids or crystalline solids. Freeze drying may be used to provide solid compounds of the invention.
  • Solvate refers to a physical association of a compound of the invention with one or more organic or inorganic solvent molecules. Such physical associations include hydrogen bonds. In some cases, the solvate will be able to separate, for example when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid.
  • the solvent molecules in the solvate can exist in a regular arrangement and/or a non-ordered arrangement.
  • the solvate can contain stoichiometric or non-stoichiometric amounts of solvent molecules.
  • “Solvate” includes solution phase and separable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are well known in the art.
  • the present invention also includes all suitable isotopic variants of the compound of the present invention or its pharmaceutically acceptable salt.
  • Isotopic variants of the compound of the present invention or its pharmaceutically acceptable salt are defined as those in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass often found in nature.
  • Isotopes that can be incorporated into the compound of the present invention and its pharmaceutically acceptable salt include but are not limited to isotopes of H, C, N and O, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 35 S, 18 F, 36 Cl and 125 I.
  • Isotopic variants of the compound of the present invention or its pharmaceutically acceptable salt can be prepared by conventional techniques, using appropriate isotopic variants of suitable reagents.
  • aromatic amine compound represented by formula (I) is selected from the following structures:
  • a method for preparing an aromatic amine compound represented by formula (I) is provided, wherein the method can be carried out by one of the following methods 1-3 or a combination thereof:
  • the process comprises the following steps:
  • Ring A, Ring B, and R 1 are as defined and preferred above;
  • G represents a leaving group, such as C1-C6 alkylsulfinyl, benzenesulfinyl, naphthalenesulfinyl, benzyl, and the above C1-C6 alkylsulfinyl, benzenesulfinyl, naphthalenesulfinyl, benzyl are optionally further substituted by one or more groups selected from halogen, C1-C6 alkyl, nitro, hydroxyl, amino, C1-C6 alkanoyl, C1-C6 alkoxy, phenyl; G is preferably C1-C4 alkylsulfinyl, benzenesulfinyl, naphthalenesulfinyl, benzyl, and the above C1-C4 alkylsulfinyl, benzenesulfinyl, naphthalenesulfinyl, benzyl are
  • M represents a leaving group, such as a metal element, a halogen, a metal complex, borane, silane, a diazonium salt, etc., preferably -MgBr, -MgCl, -Li;
  • Step a) can be carried out in the presence or absence of an acid in a solvent
  • the solvent can be selected from ethers, such as dioxane, tetrahydrofuran, ethyl ether, methyl tert-butyl ether, diisopropyl ether, diethylene glycol dimethyl ether, ethylene glycol dimethyl ether, etc.; aromatics, such as benzene, toluene, nitrobenzene, chlorobenzene, etc.; alcohols, such as methanol, ethanol, isopropanol, butanol, tert-butanol, ethylene glycol; halogenated hydrocarbons, such as chloroform, dichloromethane, dichloroethane, carbon tetrachloride; esters, such as ethyl acetate, ethyl formate, methyl acetate, isopropyl acetate; others, such as dimethyl sulfoxide,
  • the acid can be selected from organic acid, inorganic acid or Lewis acid.
  • the inorganic acid can include: hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, phosphoric acid, perchloric acid;
  • the organic acid can include: formate, acetic acid, trifluoroacetic acid, propionic acid, pyruvic acid, glycolic acid, oxalic acid, malonic acid, succinic acid, glutaric acid, maleic acid, fumaric acid, lactic acid, malic acid, citric acid, tartaric acid, picric acid, glutamic acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, salicylic acid, ascorbic acid, camphoric acid or camphorsulfonic acid;
  • Lewis acid includes: aluminum chloride, ferric chloride, boron trifluoride,
  • Step b) can be carried out in a solvent, and the solvent can be selected from ethers, such as dioxane, tetrahydrofuran, ethyl ether, methyl tert-butyl ether, diisopropyl ether, diethylene glycol dimethyl ether, ethylene glycol dimethyl ether, etc.; aromatics, such as benzene, toluene, nitrobenzene, chlorobenzene, etc.; 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, carbon tetrachloride; esters, such as ethyl acetate, ethyl formate, methyl
  • Ring A, Ring B, and R 1 are as defined and preferred above;
  • M represents a leaving group, such as a metal element, a halogen, a metal complex, borane, silane, a diazonium salt, etc., preferably -MgBr, -MgCl, -Li;
  • Step c) can be carried out in a solvent, and the solvent can be selected from ethers, such as dioxane, tetrahydrofuran, ethyl ether, methyl tert-butyl ether, diisopropyl ether, diethylene glycol dimethyl ether, ethylene glycol dimethyl ether, etc.; aromatics, such as benzene, toluene, nitrobenzene, chlorobenzene, etc.; amides, such as N,N-dimethylformamide, N,N-dimethylacetamide, 1-methyl-2-pyrrolidone, etc.; halogenated hydrocarbons, such as chloroform, dichloromethane, dichloroethane, carbon tetrachloride; esters, such as ethyl acetate, ethyl formate, methyl acetate, isopropyl acetate; others, such as dimethyl sulfoxide, acetonitrile,
  • Step d) can be carried out in a solvent under acid catalysis, and the acid can include: hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, phosphoric acid, trifluoroacetic acid, acetic acid, etc.; the azide includes: sodium azide, potassium azide, trimethylsilylazide, etc.; the reaction solvent can be selected from water; ethers, such as: dioxane, tetrahydrofuran, ethyl ether, methyl tert-butyl ether, diisopropyl ether, diethylene glycol dimethyl ether, ethylene glycol dimethyl ether, etc.; aromatics, such as: benzene, toluene, nitrobenzene, Chlorobenzene, etc.; ketones, such as acetone, methyl ethyl ketone, 4-methyl-2-pentanone, etc.; amides, such as N,N-di
  • Step e) can be carried out in a solvent under the condition of a reducing agent, and the reducing agent system includes: Pd/C catalyst catalytic hydrogenation, Pd/BaSO 4 catalyst catalytic hydrogenation, PtO 2 catalyst catalytic hydrogenation, Raney Ni catalyst catalytic hydrogenation, PPh 3 -THF-H 2 O system, LAH, etc.;
  • the required solvent is selected from water, ethers, such as dioxane, tetrahydrofuran, ethyl ether, methyl tert-butyl ether, diisopropyl ether, diethylene glycol dimethyl ether, ethylene glycol dimethyl ether, etc.; aromatic hydrocarbons, such as benzene, toluene, nitrobenzene, chlorobenzene, etc.; alcohols: methanol, ethanol, isopropanol, butanol, tert-butanol, ethylene glycol, etc., or a mixture of
  • the compound of formula (I-a) obtained by method 1-2 is subjected to amino functional group transformation to obtain other aromatic amine compounds represented by formula (I), such as by alkylation reaction, acylation reaction, etc.;
  • the alkylation reaction can be carried out in the presence of an alkylating agent, and the alkylating agent includes but is not limited to: methyl iodide, ethyl iodide, 2-bromopropane, cyclopropane bromide, tert-butyl bromide, etc.;
  • the acylation reaction can be carried out in the presence of an acylating agent, which includes but is not limited to acetic anhydride, acetyl chloride, benzoyl chloride, methylacetic anhydride, propionic anhydride, cyclopropanecarboxylic anhydride, etc.
  • an acylating agent which includes but is not limited to acetic anhydride, acetyl chloride, benzoyl chloride, methylacetic anhydride, propionic anhydride, cyclopropanecarboxylic anhydride, etc.
  • the starting compound used in each of the above reaction formulas can be a suitable salt
  • the suitable salt includes alkali metal salts and alkaline earth metal salts, such as sodium salt, potassium salt, calcium salt, magnesium salt, etc.; organic base salts, such as pyridinium salt, triethylamine salt, etc.; inorganic acid salts, such as hydrochloride, hydrobromide, hydroiodide, sulfate, nitrate, phosphate, etc.; organic acid salts, such as formates, acetates, propionates, glycolates, oxalates, malonates, succinates, fumarates, maleates, lactates, malates, citrates, isocyanates, picrates, glutamates, methanesulfonates, benzenesulfonates, etc.
  • the starting compounds used in the above reaction formulae may include solvate forms such as hydrates, alcoholates, and the like.
  • Each target compound obtained from each reaction formula can be separated and purified from the reaction mixture by the following methods, for example: after the reaction mixture is cooled, the crude product is separated by methods such as filtration, extraction or concentration, and then purified by conventional methods such as column chromatography, slurrying or recrystallization.
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound selected from the group consisting of a compound represented by formula (I), its stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds, and optionally one or more pharmaceutically acceptable carriers, diluents or excipients.
  • a compound represented by formula (I) its stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds, and optionally one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the compounds of the present invention have multi-target effects on NMDA receptors and/or monoamine transporters and/or sigma receptors, and can be used to treat various central nervous system diseases, especially depression, bipolar disorder, schizophrenia, anxiety, phobia, autism, Alzheimer's disease, bipolar disorder, hysteria, obsessive-compulsive disorder, hyperactivity disorder, epilepsy and the like.
  • the fourth aspect of the present invention there is provided one or more of the compounds represented by formula (I), their stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds, or the above-mentioned pharmaceutical compositions for preparing a method for regulating NMDA receptors. and/or monoamine transporter and/or sigma receptor activity, specifically, in the preparation of NMDA receptor antagonists, in the preparation of monoamine transporter inhibitors, and in the preparation of sigma receptor agonists or antagonists.
  • an aromatic group-containing amine compound selected from the group consisting of the stereoisomers, geometric isomers, conformational isomers, tautomers, their pharmaceutically acceptable salts, polymorphs, solvates, hydrates and isotope-labeled compounds represented by the above formula (I), or the use of the above pharmaceutical composition in the preparation of drugs for preventing and/or treating diseases associated with NMDA receptors and/or monoamine transporters and/or sigma receptors, in particular central nervous system diseases.
  • the central nervous system disease is selected from the group consisting of: cerebral ischemia; stroke; cerebral infarction; traumatic brain injury; anti-NMDA receptor encephalitis; epilepsy; amyotrophic lateral sclerosis; schizophrenia; refractory, intractable or chronic schizophrenia; affective disorders; mental disorders; mood disorders; bipolar I disorder; bipolar II disorder; depression; intrinsic depression; major depressive disorder; refractory depression; dysthymic disorder; cyclothymic disorder; panic attack; panic disorder; social phobia; obsessive-compulsive disorder; impulsive disorder; post-traumatic stress disorder; anxiety disorder; acute stress disorder; hysteria; anorexia nervosa; sleep disorder; adjustment disorder; cognitive disorder; autism; neuropathic pain; mania; Parkinson's disease; Huntington's disease; Alzheimer's disease; various dementias; memory disorders; ADHD; attention deficit/hyperactivity disorder; tics and other nervous system events or neurodegeneration caused by NMDA receptor activation.
  • the neuropathic pain includes peripheral diabetic neuropathy, post-herpetic neuralgia, complex regional pain syndrome, peripheral neuropathy, chemotherapy-induced neuropathic pain, cancer neuropathic pain, neuropathic low back pain, HIV neuropathic pain, trigeminal neuralgia, and central post-stroke pain.
  • the central nervous system disease is selected from: bipolar I disorder; bipolar II disorder; depression; intrinsic depression; major depressive disorder; refractory depression; dysthymic disorder; cyclothymic disorder; panic attack; panic disorder; social phobia; obsessive-compulsive disorder; impulsive disorder; post-traumatic stress disorder; anxiety disorder; acute stress disorder; Parkinson's disease; peripheral diabetic neuropathy; post-herpetic neuralgia; complex regional pain syndrome.
  • a method for treating and/or preventing diseases associated with NMDA receptors and/or monoamine transporters and/or sigma receptors, in particular central nervous system diseases comprises administering to humans or animals an aromatic amine compound represented by the above formula (I), a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
  • a method for preparing the above-mentioned pharmaceutical composition comprising mixing the aromatic amine compound represented by the above-mentioned formula (I), its stereoisomer or a pharmaceutically acceptable salt thereof with a pharmaceutically acceptable carrier.
  • compositions of the present invention a variety of pharmaceutical preparation forms can be selected according to the therapeutic purpose, including but not limited to: tablets, pills, capsules, granules, suspensions, solutions, creams, ointments, powders, suppositories, aerosols and injections (such as fat-soluble or oil-soluble injections), etc.
  • a “therapeutically effective amount” of a compound of the invention refers to an amount of a compound of the invention that can elicit a biological or medical response in an individual or improve symptoms, slow or delay progression of a disease, or prevent a disease, etc.
  • a “therapeutically effective amount” can be determined by the attending physician or veterinary practitioner and will vary with factors such as the compound, the disease state being treated, the severity of the disease being treated, the age and relevant health conditions of the individual, the route and form of administration, and the judgment of the attending physician or veterinary practitioner.
  • subject refers to an animal.
  • the animal is a mammal.
  • Subject also refers to, for example, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, etc.
  • the subject is a human.
  • inhibitor refers to a reduction or suppression of a particular condition, symptom or disorder or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • any disease or condition refers in one embodiment to ameliorating the disease or condition (i.e., arresting or slowing the development of the disease or at least one of its clinical symptoms). In another embodiment, “treating” refers to improving at least one physical parameter, which may not be perceived by the patient. In another embodiment, “treating” refers to modulating the disease or condition physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a physical parameter), or both.
  • prevention refers to the administration of one or more pharmaceutical substances, particularly compounds of the present invention and/or pharmaceutically acceptable salts thereof, to an individual with a predisposition to the disease in question, in order to prevent the individual from developing the disease.
  • the present invention provides a new NMDAR antagonist, which belongs to a channel pore blocker, and can inhibit the channel opening caused by excessive activation of NMDA under pathological conditions to avoid excessive influx of Ca2+ without affecting the normal function of NMDAR.
  • the NMDAR antagonist of the present invention is a reversible NMDAR antagonist, which dissociates very quickly after binding without affecting the normal function of NMDA receptors.
  • the compounds of the present invention can modulate NMDA receptor activity and thus can be used to treat and/or prevent diseases associated with NMDA receptors.
  • the compounds of the present invention have a good inhibitory effect on monoamine transporters (5-HT transporter and/or norepinephrine transporter and/or dopamine transporter), and can improve anxiety, depression or other central nervous system diseases such as anxiety and depression.
  • the compounds of the present invention have good binding activity to sigma receptors and can protect nerves, regulate cognition, improve drug addiction, and improve motor dysfunction by regulating sigma receptors in the central nervous system.
  • the compounds of the present invention have the characteristics of high oral bioavailability, low effective dose, small toxic and side effects, etc., and have therapeutic effects on diseases in the field of central nervous system, especially on central nervous system events caused by NMDA receptor activation.
  • the compounds of the present invention have the advantages of multi-target action, lower effective dose, fewer toxic side effects, better safety and tolerability, etc., have good comprehensive drugability, and have good clinical application prospects.
  • the raw materials, reagents, methods, etc. used in the examples and pharmacological examples are conventional raw materials, reagents, methods in the art.
  • Chiral separation of different compounds can be achieved by adjusting the mobile phase ratio or flow rate based on the above conditions.
  • A1-P1 (R configuration): HPLC purity: >99%, retention time: 6.534 min. Chiral purity: 100%, retention time: 9.186 min.
  • A1-P2 (S configuration): HPLC purity: >99%, retention time: 6.553 min. Chiral purity: 100%, retention time: 14.432 min.
  • A3-P1 (R configuration): HPLC purity: >98%, retention time: 6.914 min. Chiral purity: >95%, retention time: 6.847 min.
  • A3-P2 (S configuration): HPLC purity: >95%, retention time: 6.924 min. Chiral purity: >95%, retention time: 6.782 min.
  • reaction was quenched with 2M sodium hydroxide aqueous solution (1 mL), anhydrous magnesium sulfate was added, and dichloromethane was stirred, filtered, and concentrated by column chromatography to obtain a white solid (229 mg), ethyl acetate (3 mL) was added, 2M hydrogen chloride ethyl acetate solution (0.5 mL) was added dropwise, stirred for 30 min, and filtered to obtain compound A7 hydrochloride (240 mg).
  • A9-1 900 mg A9-1 was dissolved in DCM (10 mL), sodium azide (0.83 g, 5 eq) was added, TFA (9 mL) was added dropwise in an ice bath, and the mixture was reacted for 30 min in an ice bath, and then moved to room temperature and protected from light overnight. Ice water (50 mL) was added, and the pH was adjusted to 9 with ammonia water, and extracted with DCM. The organic phase was washed with a saturated sodium chloride aqueous solution, dried over anhydrous sodium sulfate, concentrated, and column chromatography was performed to obtain A9-2 (70 mg).
  • A9-2 (70 mg) was dissolved in methanol (70 mL), 10% palladium carbon (50 mg) was added, hydrogen was introduced, and the reaction was carried out at room temperature overnight. The mixture was filtered, the filtrate was concentrated, and the compound A9 was obtained by column chromatography as a white solid (38 mg), with a yield of 59%.
  • Step 1 triazole (12.4 g, 1.1 eq), tetrahydropyrrole (13.35 g, 1.2 eq), 1,4-cyclohexanedione monoethylene glycol acetal A12-1 (25.0 g, 160 mmol) were dissolved in toluene, refluxed for 18 h, and the temperature was lowered to room temperature to obtain A12-2 solution for use.
  • Step 2 Dissolve bromobenzene (75 g, 3 eq) in anhydrous THF, slowly add n-butyllithium (2.5 M) (150 mL, 3.3 eq) at -78°C, after the addition is complete, keep warm at -78°C for 1 h, add the A12-2 solution prepared above, after the addition is complete, keep warm and stir for 1 h, stir at room temperature for 12 h, and obtain A12-3 by column chromatography, 10.5 g.
  • n-butyllithium 2.5 M
  • Step 3 Compound A12-3 (10.5 g, 6.5 mmol) was dissolved in ethanol, concentrated hydrochloric acid (12 M, 3 eq) was added, and the mixture was stirred at room temperature overnight. The solvent was concentrated to dryness, and the pH value was adjusted to 11-12 with saturated aqueous sodium bicarbonate solution. The mixture was extracted with DCM, dried, filtered, and concentrated. Column chromatography was performed to obtain compound A12-4, 8.5 g.
  • A12-P1 (S configuration): HPLC purity: >99%, retention time: 8.786 min. Chiral purity: >99%, retention time: 7.703 min.
  • A12-P2 (R configuration): HPLC purity: >99%, retention time: 8.778 min. Chiral purity: >95%, retention time: 10.236 min.
  • A13-P1 (R configuration): HPLC purity: >95%, retention time: 10.144 min. Chiral purity: >99%, retention time: 4.501 min.
  • A13-P2 (S configuration): HPLC purity: >95%, retention time: 10.151 min. Chiral purity: >99%, Retention time: 6.418min.
  • A14-P1 (R configuration): HPLC purity: >98%, retention time: 9.514 min. Chiral purity: 100%, retention time: 7.835 min.
  • A14-P2 (S configuration): HPLC purity: >95%, retention time: 9.607 min. Chiral purity: 100%, retention time: 21.014 min.
  • Step 1 Add A15-1 (5 g, 1 eq), tert-butylsulfenamide (5.57 g, 1.4 eq), and titanic acid into the reaction flask. Tetraethyl acetate (8.24 g, 1.1 eq), THF, nitrogen protection, reflux reaction for 24 h (70 ° C). Cool to room temperature, pour the reaction solution into ice brine, add EA and stir, filter, separate and retain the organic phase, extract the aqueous phase with EA, combine the organic phases, wash with saturated brine, dry and concentrate, and obtain compound A15-2 by column chromatography, 2.69 g, yield 32.1%.
  • Step 2 Add iodobenzene (1.0 g, 2.5 eq) to DCM (10 mL), protect with nitrogen, reduce to -60 ° C, add butyl lithium (1.64 mL, 2.5 M, 2.1 eq) dropwise, keep warm for 1.5 h, record as reaction solution 2, and set aside. Add compound A15-2 (500 mg, 1 eq) to DCM (10 mL) and dissolve, record as reaction solution 1, and set aside. Control the temperature of reaction solution 1 at about -60 ° C and add dropwise to reaction solution 2, keep warm for 2 h. Quench the reaction with ammonium chloride, extract with EA, wash with saturated brine, dry with anhydrous sodium sulfate, concentrate, and obtain compound A15 by column chromatography, 322 mg solid, yield 49.3%.
  • PPA polyphosphoric acid
  • A19-P1 (R configuration): HPLC purity: >93%, retention time: 13.149 min. Chiral purity: 99%, retention time: 9.196 min.
  • methyl acetic anhydride Preparation of methyl acetic anhydride: Add acetic anhydride (0.51 mL, 28 eq) and 98% formic acid (0.21 mL, 28 eq) to a 10 mL reaction bottle, stir at 65 ° C for 1-2 h, cool to room temperature, and prepare methyl acetic anhydride for use. Add a DCM/THF mixed solution of compound A19 (45 mg, 1 eq, basic form) dropwise to the homemade methyl acetic anhydride under an ice-water bath, and stir at room temperature for 1 h.
  • reaction solution was cooled to 0-10 ° C, 0.05 mL of water and 0.05 mL of 20% sodium hydroxide solution were added dropwise, and filtered through diatomaceous earth, concentrated to the minimum volume, 1 mL of 4M hydrogen chloride ethyl acetate solution was added, stirred for 10 min, concentrated, and then 0.5 mL of EA was added for stirring and crystallization, and compound A21 in the form of hydrochloride was obtained by filtration, 23 mg of off-white solid, and the yield was 57.5%.
  • A22-1 (195 mg, 1.3 mmol) was placed in a 50 mL three-necked flask, and after replacing nitrogen, dry THF (5 mL) was added, and the temperature was lowered to 0°C, and PhMgCl (1 mL, 1.97 mmol, 1.5 eq.) was added dropwise, and the mixture was reacted at 0°C for 2 h after the addition was complete. Saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with ethyl acetate. The organic phase was washed with saturated brine, dried, filtered, concentrated to dryness, and the product A22-2 was obtained by column chromatography, as a yellow oil (115 mg).
  • A22-2 (115 mg, 0.5 mmol) was dissolved in DCM (3 mL), and NaN 3 (65 mg, 1 mmol, 2 eq.) was added. After replacing the nitrogen, the temperature was lowered to 0°C, and TFA (0.2 mL, 2.56 mmol, 5 eq.) was added dropwise. After the addition was complete, the mixture was reacted at 0°C for 2.5 h. After dilution with water under an ice-water bath, ammonia was added to quench the reaction, and then extracted with ethyl acetate. The organic phase was washed with saturated brine, dried, filtered, and concentrated to obtain A22-3.
  • A22-3 was dissolved in MeOH (10 mL), Pd/C (150 mg) was added, hydrogen was replaced, and the reaction was carried out at room temperature for 2 h.
  • the reaction solution was filtered to remove Pd/C and then concentrated, the residue was dissolved in MTBE (10 mL), 4M hydrogen chloride/ethyl acetate solution (0.1 mL) was added dropwise, a white solid precipitated, and compound A22 was obtained by filtration, a gray-white solid of 30 mg, and a three-step yield of 8.8%.
  • A22-P1 (R configuration): HPLC purity: >97%, retention time: 13.319 min. Chiral purity: 99%, retention time: 10.137 min.
  • A22-P2 (S configuration): HPLC purity: >97%, retention time: 13.321 min. Chiral purity: 98.8%, retention time: 10.871 min.
  • reaction flask In the reaction flask, add compound A23-18g, N-methylbenzylamine (6.83g, 1.1eq), 1,2,3-triazole (4.25g, 1.2eq), and 50mL of toluene, reflux and separate water overnight, and cool to room temperature. Take another reaction flask and add phenylmagnesium bromide (4eq, in THF), add the above reaction solution dropwise in an ice bath, stir for 90min, pour the reaction solution into an aqueous solution of ammonium chloride, add EA, wash the organic phase with water, dry, concentrate, and column chromatograph to obtain compound A23-2, 3.85g of light yellow oil, with a yield of 22%.
  • acetic anhydride Preparation of acetic anhydride: Add acetic anhydride (127.8 mg, 3 eq) and anhydrous formic acid (57.6 mg, 3 eq) into a 10 mL reaction bottle, stir at 65 °C for 1-2 h, cool to room temperature, and obtain acetic anhydride for use.
  • Grignard reagent Mg (3.4 g, 10 eq) was suspended in anhydrous THF, replaced with nitrogen three times, bromobenzene (29 g, 10 eq) was slowly added dropwise, heated at 50°C for 1 h, and then cooled to -10-0°C for use.
  • Grignard reagent Magnesium (345.6 mg, 4.8 eq) is suspended in dry tetrahydrofuran, bromobenzene (200 mg, 0.48 eq) is added dropwise and the temperature is raised to 50-60°C to initiate the reaction. Bromobenzene (1.8 g, 3.6 eq) is then added dropwise to maintain a slight boil. The addition is completed over 30 min, the temperature is raised to 60°C and stirred for 1 h. Most of the magnesium disappears, and the temperature is lowered to 0-10°C for standby use.
  • Triazole (12.4 g, 1.1 eq), tetrahydropyrrole (13.35 g, 1.2 eq), and 1,4-cyclohexanedione monoethylene glycol acetal (25.0 g, 160 mmol) were dissolved in toluene, refluxed for 18 h, cooled to room temperature, and a toluene solution of compound A50-1 was obtained for later use.
  • A50-P1 (S configuration): HPLC purity: >99%, retention time: 5.761 min. Chiral purity: >95%, retention time: 7.617 min.
  • A50-P2 (R configuration): HPLC purity: >99%, retention time: 7.452 min. Chiral purity: >95%, retention time: 7.623 min.
  • Step 1 At 0-10°C, add phosphorus oxychloride (126 mg, 2 eq) dropwise to a DMF (50 mg, 1.5 eq) dichloromethane solution, stir for 2 h in an ice bath, dissolve compound A50-3 (100 mg, 0.5 mmol) in dichloromethane, add to the above reaction solution, stir at room temperature overnight, slowly dropwise into a sodium acetate aqueous solution, separate the liquids, dry, filter, and concentrate the organic phase to obtain compound A52-1 as an oil.
  • DMF 50 mg, 1.5 eq dichloromethane solution
  • Step 2 Dissolve the above oily substance A52-1 in ethanol, add ethyl mercaptoacetate (74 mg, 1.5 eq), stir for 10 min, add sodium ethoxide (55 mg, 4 eq) under ice bath, react overnight at room temperature, add water, heat under reflux for 2 h. Concentrate ethanol, adjust pH to 5-6 with 1M hydrochloric acid solution, extract with dichloromethane, dry the organic phase, concentrate, and perform column chromatography to obtain compound A5255 mg as a light yellow oily substance.
  • Triazole 1.0 g, 1.2 eq
  • N-methylbenzylamine 1.8 mL, 1.2 eq
  • 1,4-cyclohexanedione monoethylene glycol acetal 2.0 g, 1.28 mmol
  • o-Chlorobromobenzene 24.83 g, 130 mmol was dissolved in dry THF (100 mL), cooled to -78 °C, and n-BuLi (48 mL, 2.5 M) was added dropwise. The addition was completed in about 1 h. After 1 h, a solution of A35-2 (5.340 g, 20 mmol) in THF (80 mL) was added dropwise, and the temperature was controlled at -78 °C and stirred for 5 h.
  • reaction was quenched with saturated aqueous ammonium chloride solution, extracted with EA, and the organic phase was washed with saturated aqueous sodium chloride solution, dried, concentrated, and column chromatography to obtain compound A60-1 as a yellow foamy solid (2.8 g).
  • phenylmagnesium chloride (3eq, 20mL) was added to a 250mL three-necked flask.
  • a tetrahydrofuran solution of A65-1 was slowly added under an ice bath. After the addition, the mixture was stirred for 20min and then the ice bath was removed and stirred at room temperature. After the reaction was completed by TLC monitoring, a saturated aqueous solution of ammonium chloride was added to quench the reaction. After filtration, the mixture was extracted with EA three times. The mixture was dried and concentrated, and A65-2 was obtained by column chromatography as a white solid (3.01g).
  • PTSA 0.5 eq, 0.705 g was added and stirred.
  • a saturated aqueous sodium bicarbonate solution was added to quench the reaction.
  • EA was extracted three times, dried and concentrated to obtain A65-3, a brown-red liquid of 1.675 g, with a yield of 77.15%.
  • A65-3 (1 eq, 1.675 g) and DMF-DMA (5 eq, 3.79 mL) were heated to reflux overnight at 110° C. After the reaction was completed, A65-4 solution was obtained, which was used directly in the next step without treatment.
  • the A65-4 solution obtained in the previous step and hydrazine hydrate (5 eq, 1.68 g) were dissolved in methanol and heated at 80°C overnight. Concentrated to dryness to obtain A65-5, 2 g of brown-red liquid.
  • A65-5 (1 eq, 500 mg) was dissolved in 2 mL of anhydrous ethanol. After stirring for ten minutes, a 2 M hydrochloric acid solution was added and stirred for 2 h. The mixture was quenched with a saturated sodium bicarbonate aqueous solution, the pH was adjusted to 8-9, and the mixture was extracted with EA three times, dried and concentrated, and chromatographed with an amino column to obtain compound A65, 340 mg of a dark red compound.
  • Step 1 Compound A48-3 (1eq, 0.7g) was dissolved in toluene, dried with anhydrous magnesium sulfate, and filtered for use; toluene, sodium tert-butoxide (2eq, 523mg) and ethyl formate (1.5eq, 303mg) were added to another reaction bottle, and the toluene solution of the above compound A48-3 was added at 0-10°C, and stirred at room temperature overnight.
  • reaction solution was extracted with toluene twice, the aqueous phase was retained and the pH was adjusted to 6-7 with 1M hydrochloric acid solution, extracted with dichloromethane three times, dried, concentrated, and column chromatographed to obtain compound A70-1, 0.72g, with a yield of 93%.
  • Step 2 Compound A70-1 (1 eq, 90 mg) was dissolved in ethanol, 85% hydrazine hydrate (3 eq, 97 mg) was added, and the temperature was raised to 85°C for 5 h. After adding anhydrous sodium sulfate to dry, the solvent was concentrated, and the residue was subjected to column chromatography to obtain compound A70 as a white solid (87 mg) with a yield of 98%.
  • A70-P1 (S configuration): HPLC purity: >95%, retention time: 10.024 min. Chiral purity: >99%, retention time: 6.319 min.
  • A70-P2 (R configuration): HPLC purity: >95%, retention time: 10.009 min. Chiral purity: >95%, retention time: 7.971 min.
  • Step 1 Add A252-1 (5.5 g, 1.0 eq), ethylene glycol (1.7 g, 1.05 eq), and toluene (100 ml) into a 250 ml three-necked flask. Heat to 120 °C and react for 2-3 h under nitrogen protection. Cool the reaction solution to room temperature, wash with 100 ml of saturated sodium bicarbonate, and collect The organic phases were collected, washed with 100 ml of saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain A252-2 as an oily product (5.2 g).
  • Step 2 Add the above oily substance A252-2 to a 250ml three-necked flask, stir and dissolve with 50ml tetrahydrofuran, replace with nitrogen, cool to about 0°C in an ice-water bath, add lithium aluminum hydride (1.5g, 1.5eq) in batches, and after the addition is complete, heat to about 70°C and stir for 2h.
  • lithium aluminum hydride 1.5g, 1.5eq
  • reaction solution is cooled to 0°C in an ice-water bath, quenched with saturated ammonium chloride solution, extracted with 100ml ethyl acetate, and the organic phase is washed with 50ml saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain A252-3, 5.5g of off-white solid.
  • Step 3 Add A252-3 (2.2 g) and 20 ml of ethyl formate to a 100 ml three-necked flask, stir and dissolve, heat to 70°C under nitrogen protection and react for about 2 hours. Concentrate under reduced pressure to obtain A252-4, 2 g of off-white solid.
  • Step 4 Add A252-4 (650 mg) to a 50 ml three-necked flask, stir and dissolve with 10 ml of dichloromethane, add tribromopyridinium (950 mg, 1.1 eq), and stir at room temperature for 4-5 h. Quench the reaction with 5 ml of saturated sodium thiosulfate solution, extract with 20 ml of dichloromethane, combine the organic phases, dry and concentrate to obtain 450 mg of off-white solid. Add 10 ml of dioxane and stir to dissolve, add thiourea (615 mg, 3.0 eq), DIPEA (700 mg, 2.0 eq), heat to 80-90 ° C and stir overnight. The reaction solution is cooled to room temperature, and 10 ml of water and dichloromethane are added for extraction. The organic phase is dried, concentrated, and column chromatography is performed to obtain A252-5, an off-white solid of 350 mg.
  • Step 5 Add A252-5 (200 mg) and 10 ml tetrahydrofuran to a 50 ml three-necked flask and stir to dissolve. Under nitrogen protection, add borane tetrahydrofuran solution (7 ml, 10 eq), heat to 60 ° C and keep warm for 2 h.
  • test compounds are represented by "rac” if they are racemates, and "R” or “S” if they are pure enantiomers.
  • NMDA receptor N-methyl-D-aspartate receptor, NR1/2A
  • Patch clamp amplifier Multiclamp 700B, Axopatch 200B, Axon, USA
  • Electrode pulling apparatus P-97, Sutter, USA
  • HEPES 4-(2-Hydroxyethyl)piperazine-1-ethanesulfonic acid, N-(2-hydroxyethyl)piperazine-N'-(2-ethanesulfonic acid)
  • Ethylene glycol bis(2-aminoethyl ether)tetraacetic acid (Sigma, Cat: E3889)
  • Lipofectamine 3000 transfection kit (Gibco, Cat: L3000015) (Contains two reagents: Lipofectamine 3000 and P3000)
  • Fetal bovine serum (FBS) (Gibco, Cat: 10099141)
  • Intracellular solution formulation 10 CsCl, 115 CsF, 10 EGTA and 10 HEPES, pH adjusted to 7.2 with CsOH.
  • HEK293 cells were cultured in DMEM medium containing 10% fetal bovine serum at 37°C and 5% carbon dioxide.
  • Cell passaging Remove the old culture medium and wash once with PBS, then add 1 mL 0.25% Trypsine-EDTA solution and incubate at room temperature for 1 minute. When the cells detach from the bottom of the dish, add 3 mL complete culture medium (90% DMEM + 10% FBS) preheated at 37°C. Gently blow the cell suspension with a pipette to separate the aggregated cells. Transfer the cell suspension to a sterile centrifuge tube and collect the cells by centrifugation at 800 rpm for 3 minutes. Inoculate the cells in a T25 cell culture flask at a ratio of 1:5 (final volume: 6 mL) for expansion or maintenance culture.
  • 3 mL complete culture medium 90% DMEM + 10% FBS
  • Transient transfection 24 h before transient transfection, HEK-293 cells with a cell density of about 80% were re-plated into a 35 mm2 cell culture dish, with an inoculation volume of 3 ⁇ 105 cells per cell culture dish.
  • HEK293 cells transiently expressing NMDA receptor channels were used to record the current induced by 100 ⁇ M Glutamic acid (containing 100 ⁇ M Glycine) at room temperature using the whole-cell patch clamp technique.
  • the glass microelectrode was pulled from a glass electrode blank (BF150-86-10, Sutter) using a puller. The tip resistance after perfusing the electrode liquid was about 2-5M ⁇ .
  • the glass microelectrode was inserted into the amplifier probe to connect to the patch clamp amplifier.
  • the clamping voltage and data recording were controlled and recorded by a computer using pClamp software, with a sampling frequency of 10kHz and a filter frequency of 2kHz.
  • the cell was clamped at -70mV, and 100 ⁇ M Glutamic acid (containing 100 ⁇ M Glycine) was used to induce channel current. After the current stabilized, 100 ⁇ M Glutamic acid (containing 100 ⁇ M Glycine) containing the compound was administered to observe the change in current amplitude. The compound was administered continuously from low concentration to high concentration, and finally 100 ⁇ M Glutamic acid (containing 100 ⁇ M Glycine) was administered again. Each test concentration of the compound was administered for at least 20s, and at least 2 cells (n ⁇ 2) were tested for each concentration.
  • Inhibition% represents the inhibition percentage of the compound on the NMDA channel current
  • I and Io represent the current amplitude induced by 100 ⁇ M Glutamic acid (containing 100 ⁇ M Glycine) before and after drug addition, respectively.
  • X is the Log value of the test sample concentration
  • Y is the inhibition percentage at the corresponding concentration
  • Bottom and Top are the minimum and maximum inhibition percentages, respectively.
  • Liver microsomes were incubated in 96-well plates. The volume of each incubation system was 200 ⁇ L. The medium was 0.1 M phosphate buffer (pH 7.4), including liver microsomes with a final concentration of 0.2 mg/mL, 1 ⁇ M of the test drug, 3.0 mM MgCl 2 , 0.01% DMSO, 0.5% acetonitrile and 2.0 mM NADPH. Using a rotating water bath constant temperature oscillator, the above incubation system without NADPH was pre-incubated at 37°C for 5 minutes, and then NADPH was added to start the reaction.
  • phosphate buffer pH 7.4
  • Sample analysis After the incubated samples are treated with organic solvent extraction and protein precipitation, the concentration of the test drug or positive control drug in the sample is semi-quantitatively determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The ratio of the analyte peak area to the internal standard peak area is used to express the concentration in the sample.
  • LC-MS/MS liquid chromatography-tandem mass spectrometry
  • Animals Male ICR mice. Animals were randomly assigned to treatment groups and fasted for 12 h before dosing.
  • the compound of the present invention is dissolved in a solvent (5% DMSO + 5% Solutol HS15 + 90% Saline).
  • the intravenous (iv) dose is 2.5 mg/kg, and the administration volume is 5 mL/kg; the oral (po) dose is 10 mg/kg, and the administration volume is 10 mL/kg body weight.
  • Plasma samples were obtained by centrifugation of whole blood, and 10 ⁇ L of plasma was taken and mixed with 190 ⁇ L of internal standard solution (20 ng/mL dissolved in acetonitrile containing 0.1% formic acid). After mixing, centrifuge at 13000 rpm for 10 min, take 120 uL of supernatant each, and then take out 0.5-10 ul (depending on the sensitivity of the compound) for drug analysis using an appropriate liquid chromatography-tandem mass spectrometry (LC-MS/MS) method, and the standard of each analyte is used for calibration and identification.
  • LC-MS/MS liquid chromatography-tandem mass spectrometry
  • the membrane proteins used in this experiment were extracted from the HEK293 cell line stably expressing the Sigma1 receptor and the HEK293 cell line stably expressing the Sigma2 receptor constructed by WuXi AppTec.
  • PEI Poly ethyleneimine
  • the cell membrane solution and isotope solution were prepared with pre-cooled detection buffer.
  • the preparation information was as follows: membrane concentration was 10 ⁇ g/well, and the final concentration of the isotope ligand [3H]-DTG was 5 nM.
  • the cell membranes were collected onto a GF/C filter plate using a cell collector, washed 4 times with pre-cooled plate washing buffer, and then the GF/C filter plate was placed in a 50° C. oven to dry for 1 hour.
  • Ki IC 50 /(1+L/K D ).
  • the compound of the present invention was mixed with 5% DMSO and then 5% Mix HS 15 and add 90% saline to prepare a solution of appropriate concentration for immediate use.
  • mice Male C57 mice, about 22 g. The animals were randomly divided into a blank control group and each test drug group, with 8 animals in each group. The mice in each group were intraperitoneally injected with the solvent or each test drug.
  • mice were subjected to forced swimming test.
  • the water level in the forced swimming device was 45 cm and the water temperature was 25°C.
  • the mice were placed in the experimental room to adapt to the environment for 1 hour before the experiment. At the beginning of the experiment, the mice were placed in the device for 6 minutes. The whole process was recorded by a camera. When analyzing the data, only the last 4 minutes of immobility time of the mice were counted.
  • Bovine Serum Albumin (Sigma, Cat: B2064-100G)
  • Neurotransmitter transporter uptake assay kit (Molecular devices, Cat: R8174)
  • Neurotransmitter transporter uptake assay kit (Molecular devices) was used to detect the inhibitory effect of the test compound on the human SERT transporter. The test was performed according to the method in the kit manual, and Citalopram was used as a positive control. The specific operation is as follows:
  • HEK-293-hSERT cells were seeded into a 384-well plate at 20,000 cells/well, and the 384-well plate was then transferred to an incubator at 37°C overnight;
  • test solutions of citalopram and the compound of the present invention were prepared with an experimental buffer (HBSS solution containing 0.1% BSA and 20 mM HEPES), the starting concentration of citalopram was 1 ⁇ M, 3X dilution, the starting concentration of the test compound was 10 ⁇ M or 100 ⁇ M, 3X dilution, and each concentration was repeated 2 times;
  • an experimental buffer HBSS solution containing 0.1% BSA and 20 mM HEPES
  • Neurotransmitter transporter uptake assay kit was used to test the transporter inhibition of HEK-293 cells expressing human DAT and NET. The test was performed according to the method in the kit manual, using Centanafadine as a positive control. The specific operation is as follows:
  • HEK 293T cells were digested with trypsin, centrifuged and resuspended in culture medium, and then inoculated in a 6cm culture dish after cell counting, with a seeding density of 3 ⁇ 106 cells/well;
  • HEK 293T cells were treated with a change of medium, and then the NET-pcDNA5/FRT plasmid to be transfected was prepared in two tubes A and B.
  • 200 ⁇ L Opti-MEM was added to tube A, and then 10 ⁇ L Lipofectamine TM 3000 was added and mixed evenly.
  • 200 ⁇ L Opti-MEM was first added to tube B, and then 5 ⁇ g NET-pcDNA5/FRT plasmid was added, mixed evenly, and then 10 ⁇ L P3000 TM was added to tube B and mixed evenly (the ratio of plasmid to transfection reagent was 1 ⁇ g: 2 ⁇ L).
  • the diluted solution in tube A was added to the diluted solution in tube B, mixed evenly, and incubated at room temperature for 15 minutes. Finally, the mixture was gently added to the cells that had been changed, gently shaken to mix, and then cultured overnight in an incubator at a temperature of 37°C and a carbon dioxide concentration of 5%. Cells were used for compound functional activity determination 18-20 hours after transfection.
  • DAT stable culture DAT-HEK cell line was cultured in DMEM medium containing 10% fetal bovine serum and 0.2 mg/mL Hygromycin B. The culture temperature was 37°C and the carbon dioxide concentration was 5%.
  • Passaging of DAT stable transfected cells Remove the old culture medium and wash once with PBS, then add 1 mL of TrypLE TM Express solution and incubate at 37°C for about 2 minutes. When the cells detach from the bottom of the dish, add about 5 mL of complete culture medium preheated at 37°C. Gently blow the cell suspension with a pipette to separate the aggregated cells. Transfer the cell suspension to a sterile centrifuge tube and centrifuge at 1000 rpm for 5 minutes. To maintain the physiological activity of the cells, the experimental cell confluence was controlled at about 80%.
  • NET cells were trypsinized after overnight transfection, resuspended in DMEM + 10% Dialyzed FBS medium, and inoculated with 20,000 cells/well in 384-well plates for overnight culture.
  • Stably transfected DAT cells were trypsinized, resuspended in DMEM + 10% Dialyzed FBS medium, and inoculated with 2,500 cells/well in 384-well plates for overnight culture.
  • step d) Take 16 ⁇ L of the compound prepared in step b) and add it to the corresponding experimental wells, where the positive control well is added with 2 ⁇ the first concentration of the positive control compound, and the negative control well is added with 0.2% DMSO buffer. After centrifugation, incubate at 37°C for 30 minutes;

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明公开了一类含芳基的胺类化合物、其制备方法和用途。所述含芳基的胺类化合物由式(I)表示,具有可调节NMDA受体和/或单胺转运体和/或sigma受体活性的功能,从而可用于制备治疗和/或预防与NMDA受体和/或单胺转运体和/或sigma受体相关联的疾病,特别是中枢神经系统疾病的药物。

Description

一类含芳基的胺类化合物、其制备方法和用途
相关申请的交叉引用
本申请要求2022年11月9日提交的申请号为202211401952.5,以及2023年9月4日提交的申请号为202311134131.4,发明名称均为“一类芳胺类化合物、其制备方法和用途”的中国专利申请的优先权,该申请的全部内容通过引用整体并入本文,其程度如同完全记载在本文中一样。
技术领域
本发明属于药物化学领域。具体而言,本发明涉及一类式(I)所示的含芳基的胺类化合物,其制备方法和药物组合物,及其在制备预防或治疗与NMDA受体和/或单胺转运体和/或sigma受体相关联的疾病,特别是中枢神经系统疾病的药物中的应用。
背景技术
抑郁症是一种严重的精神类疾病,主要表现是情绪消沉,自卑抑郁,甚至患者有自杀的倾向。目前治疗抑郁症的主要药物是SSRI/SSNI(Selective serotonin/norepinephrine reuptake inhibitors)。但此类药对近三分之一的患者没有疗效,且普遍存在起效慢、甚至增加患者自杀倾向等缺陷。该领域存在巨大的未满足的临床需求。
NMDA(N-甲基-D-天冬氨酸(N-methyl-D-aspartic acid))型谷氨酸受体(简称NMDA受体或NMDAR)是一种配体门控离子通道。该受体在体内可以被中枢神经内最重要的兴奋性神经递质谷氨酸所激活,从而介导突触间兴奋性信号的传导。NMDAR的离子通道开放后,对Ca2+、K+、Na+等阳离子的通透性增强,产生兴奋性突触后电位,引发一系列生理生化反应。NMDAR分子结构复杂,各种亚型在时空分布和药理学特性上具有特异性。其数量、构成和分布在不同发育时期、不同脑区呈现动态变化,参与诸多生理活动,为复杂的神经活动提供分子基础,从而保证了神经网络的正常活动。NMDAR的整合、定位、回收、以及突触内外的分布等过程依赖于神经活动的调节,其功能稳态被打破和众多脑疾病高度相关,比如抑郁症、癫痫、精神分裂症等。
单胺转运体是在中枢和外周神经系统神经元细胞膜上发现的一类负责转运突触传递中常用神经递质的蛋白质。单胺转运体抑制剂通常被用作抗抑郁药物来治疗抑郁和焦虑障碍,如具有去甲肾上腺素转运体和多巴胺转运体抑制作用的安非他酮,具有5-HT转运体抑制作用的氟西汀、西酞普兰。Sigma受体主要位于中枢神经系统中,也位于内分泌和免 疫系统中,与中枢神经系统中的精神病、抑郁症、认知、神经保护和运动功能紧密相关。Sigma1受体激动剂可能具有抗抑郁、促进学习记忆的作用,而拮抗剂可能具有抗精神病、镇痛作用。sigma2受体是治疗阿尔茨海默病、精神分裂症和创伤性脑损伤等中枢疾病的理想分子靶标,且sigma2受体激动剂和拮抗剂都是神经退行性疾病的潜在疗法。
NMDA受体拮抗剂氯胺酮作为一种良好的麻醉剂已被运用于医学上超过50年。研究发现静脉注射亚麻醉剂量的NMDAR拮抗剂氯胺酮能在数小时内迅速缓解抑郁症的症状,并且效果能维持至少一周。强生公司研发的艾氯胺酮已于2019年获FDA批准上市,用于难治性抑郁症的治疗。鉴于氯胺酮的主要分子靶点是NMDAR,许多人提出抑制该靶点也是氯胺酮抗抑郁作用的原因。这种机制表明,氯胺酮的抗抑郁作用和解离副作用可能在机制层面上是密不可分的。然而,许多证据对这一假设提出了质疑(J Psychiatry Neurosci.2017,42(4),222.)。首先,已发现氯胺酮的R对映异构体(R-ket)在啮齿动物模型中作为抗抑郁药比S对映异构体(S-ket)更有效且更持久,尽管R-ket对NMDAR的亲和力明显比S-ket弱(Pharmacol Biochem Behav.2014,116,137.)。类似地,氯胺酮代谢物(2R,6R)-羟基去甲氯胺酮(HNK)已被证明在啮齿动物模型中诱导抗抑郁作用,但在诱导抗抑郁作用的剂量水平下在体内不会与NMDAR结合(Nature.2016,533(7604),481.和Proc Natl Acad Sci USA.2019,116(11),5160.和Org Lett.2017,19(17),4572.)。因此,R-ket和HNK都可以诱导抗抑郁作用,同时限制氯胺酮的解离作用。然而,为减弱氯胺酮的解离副作用而提出的其他策略结果均不佳,例如,通过靶向NMDAR的NR2B亚基或利用具有低捕获特性的化合物(Nature.2016,533(7604),481.和Sci Rep.2017,7(1),15725.和Int J Neuropsychopharmacol.2019,22(2),119.和J Psychiatr Res.2017,86,55.和Psychiatry Res.2016,239,281.)。因此,支持氯胺酮抗抑郁作用的精确分子机制仍然知之甚少,可能涉及其他尚未确定的靶点目标。此外,NMDAR调节剂的抗抑郁作用及其伴随的解离作用的大小通常是高度不可预测的。这些发现提出了令人兴奋的可能性,即氯胺酮的抗抑郁作用实际上可能与其解离副作用分开。
氯胺酮和艾氯胺酮的解离性副作用和较差的口服生物利用度极大地限制了其临床应用。尽管已经开发了其他可口服的NMDAR拮抗剂,但迄今为止还没有一种药物被证明具有类似于氯胺酮的快速抗抑郁临床疗效。因此,仍然迫切需要具有强大的临床功效、低或无解离副作用、良好的口服生物利用度的新型抗抑郁药。保留类似于氯胺酮的快速抗抑郁活性,同时具有降低的解离副作用和良好的口服生物利用度的药物将提供一种新的治疗选择,由于其降低的解离作用和伴随的降低的滥用可能性,该治疗选择更易于管理并且可能在家庭使用中可行。
发明内容
发明目的
本发明的目的是提供一种具有NMDA受体和/或单胺转运体和/或sigma受体调节作用的新型含芳基的胺类化合物及其制备方法和用途。
本发明的一个目的是提供一种式(I)所示的含芳基的胺类化合物,或其立体异构体、几何异构体、构象异构体、互变异构体、药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物。
本发明的另一个目的是提供制备式(I)所示的含芳基的胺类化合物的方法。
本发明的又一个目的是提供药物组合物,包含治疗有效量的选自式(I)化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,以及可选地一种或多种可药用载体、稀释剂或赋形剂。
本发明的又一个目的是提供选自式(I)化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,或上述药物组合物在制备调节NMDA受体和/或单胺转运体和/或sigma受体活性的药物中的应用。
本发明的再一个目的是提供选自式(I)化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,或上述药物组合物在制备预防和/或治疗与NMDA受体和/或单胺转运体和/或sigma受体相关联的疾病,特别是中枢神经系统疾病的药物中的应用。
技术方案
根据本发明的一个方面,提供了一种式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物:
其中:
A环选自含有1-3个,例如1、2或3个选自N、O和S的杂原子的4-10元杂环或C6-C10芳环并4-10元杂环;所述4-10元杂环例如为5、6、7、8、9元杂环,优选为5-8元杂环;优选地,环A中杂环中含有1个N原子和1个S原子,或者含有两个N原子,或者含有1个S原子,或者含有两个N原子和1个S原子,或者含有1个N原子和1个O原子;
所述A环任选地被一个或多个R5取代,各R5独立选自卤素、羟基、氨基、氰基、羧基、氧代、C1-C6烷基、卤代C1-C6烷基、C1-C6烷氧基、C1-C6烷酰基、氨基甲酰基(-CONH2)、被C1-C6烷基取代的氨基甲酰基、被一个或两个C1-C6烷基取代的氨基、被一个或两个卤代C1-C6烷基取代的氨基、被一个或两个C1-C6烷酰基取代的氨基、C1-C6烷氧基羰基、C3-C6环烷基、4-10元杂环烷基、C6-14芳基、5-10元杂芳基、C6-C14芳基C1-C6烷基氧基或者5-10元杂芳基C1-C6烷基氧基;优选地,各R5独立选自卤素、羟基、氨基、氰基、羧基、氧代、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、C1-C4烷酰基、氨基甲酰基(-CONH2)、被C1-C4烷基取代的氨基甲酰基、被一个或两个C1-C4烷基取代的氨基、被一个或两个卤代C1-C4烷基取代的氨基、被一个或两个C1-C4烷酰基取代的氨基、C1-C4烷氧基羰基、C3-C6环烷基、4-8元杂环烷基、C6-10芳基、5-10元杂芳基、C6-C10芳基C1-C4烷基氧基或者5-10元杂芳基C1-C4烷基氧基;更优选地,各R5独立选自卤素(特别是溴)、氨基、羟基、氰基、羧基、C1-C3烷基(特别是甲基、乙基、异丙基)、卤代C1-C3烷基(特别是三氟甲基)、C1-C3烷氧基(特别是甲氧基、乙氧基)、C1-C3烷酰基(特别是甲酰基、乙酰基)、氨基甲酰基(-CONH2)、甲酰基氨基、乙酰基氨基、甲氨基、乙氨基、N,N-二甲基氨基、2,2,2-三氟乙基氨基、C1-C3烷氧基羰基(特别是甲氧基羰基、乙氧羰基)、C3-C5环烷基(特别是环丙基)、苯基、吡啶基、吡咯烷基、哌啶基、吗啉基或苄氧基;
环B为3-10元碳环;优选为5-8元碳环,例如5、6、7或8元碳环,更优选为5、6或7元碳环;
R1连接于环B中同一个环碳原子;
x为0-2的整数,例如0、1或2;
R2和R3各自独立地选自氢、C1-C6烷基、卤代C1-C6烷基、C1-C6烷氧基C1-C6烷基、羟基C1-C6烷基、C6-C14芳基C1-C6烷基、C3-C6环烷基、C1-C6烷酰基、卤代C1-C6烷酰基、C3-C6环烷基C1-C6烷酰基、C6-C14芳基C1-C6烷酰基、C1-C6烷磺酰基、C1-C6烷基亚砜基或C3-C6环烷基C1-C6烷基;优选各自独立地选自氢、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基C1-C4烷基、羟基C1-C4烷基、C6-C14芳基C1-C4烷基、C3-C6环烷基、C1-C4烷酰基、卤代C1-C4烷酰基、C3-C6环烷基C1-C4烷酰基、C6-14芳基C1-C4烷酰基、C1-C4烷磺酰基、C1-C4烷基亚砜基或C3-C6环烷基C1-C4烷基;优选选自氢、C1-C4烷基(特别是甲基、乙基、丙基、异丙基、叔丁基)、卤代C1-C4烷基(特别是1,1,1-三氟乙基)、甲氧基乙基、羟甲基、羟乙基、C3-C6环烷基甲基(特别是环丙基甲基、环丁基甲基)、苄基、C3-C6环烷基(特别是环丙基)、C1-C3烷基酰基(特别是乙酰基、丙酰基)、环丙基甲酰基、苯甲酰基、叔丁基亚砜基;
或者,R2、R3和相连的氮原子一起形成3-9元杂环烷基,所述3-9元杂环烷基环中任选地包含一个或多个额外的氮原子或氧原子,所述3-9元杂环烷基任选地被一个或多个C1-C6烷基,优选C1-C4烷基取代;优选地,R2、R3和相连的氮原子一起形成氮杂环丁烷基、吡咯烷基、哌啶基、氮杂环庚烷基、吗啉基;
R1选自C6-C14芳基、5-10元杂芳基、4-10元杂环基、C6-C10芳基并4-10元碳环(例如茚满基)或C6-C10芳基并4-10元杂环(例如1,2-甲撑二氧基苯基、2,3-二氢苯并呋喃基),优选选自C6-C14芳基、5-10元杂芳基,更优选为苯基、萘基、喹啉基、异喹啉基、吡啶基、嘧啶基、吡嗪基、哒嗪基、噻吩基或噻唑基,还优选为苯基、萘基、嘧啶基、吡啶基、吡嗪基、哒嗪基或喹啉基,进一步优选为苯基;
所述C6-C14芳基、5-10元杂芳基、4-10元杂环基、C6-C10芳基并4-10元碳环或C6-C10芳基并4-10元杂环任选地被一个或多个R6取代;
R6各自独立地选自卤素、羟基、巯基、氰基、氨基甲酰基(NH2CO-)、氨基磺酰基(NH2SO2-)、C1-C6烷基、卤代C1-C6烷基、羟基C1-C6烷基、C3-C6环烷基、C3-C6环烷氧基、被C1-C3烷基取代的C3-C6环烷基、被C1-C3烷基取代的C3-C6环烷氧基、C1-C6烷氧基、C1-C6烷硫基、卤代C1-6烷氧基、C3-C6环烷基C1-C6烷氧基、C1-C6烷酰基氧基、C6-C14芳基、5-10元杂芳基、C6-C14芳基C1-C6烷氧基、5-10元杂芳基C1-C6烷氧基;优选地,R6各自独立地选自卤素、氰基、羟基、氨基甲酰基(NH2CO-)、氨基磺酰基(NH2SO2-)、C1-C4烷基、卤代C1-C4烷基、羟基C1-C4烷基、C3-C6环烷基、C3-C6环烷氧基、被C1-C3烷基取代的C3-C6环烷基、被C1-C3烷基取代的C3-C6 环烷氧基、C1-C4烷氧基、C1-C4烷硫基、卤代C1-C4烷氧基、C3-C6环烷基C1-C3烷氧基、C1-C3烷酰基氧基、C6-C10芳基、5-6元杂芳基、C6-C10芳基C1-C4烷氧基、5-6元杂芳基C1-C4烷氧基;更优选地,R6各自独立地选自氟、氯、溴、氰基、羟基、甲基、异丙基、二氟甲基、三氟甲基、羟甲基、甲基环丙基氧基、甲氧基、乙氧基、异丙氧基、异丁氧基、甲硫基、环丙基、环丙基氧基、氨基甲酰基(NH2CO-)、氨基磺酰基(NH2SO2-)、二氟甲氧基、三氟甲氧基、1,1,1,-三氟乙氧基、氰基、环丙基甲氧基、乙酰氧基、苯基、吡啶基或苄氧基;
m为0-3的整数,例如0、1、2或3;
R4连接至B环上除R1共同连接的碳原子外的任意碳原子上,且R4各自独立地选自羟基、卤素、C1-C6烷基。
在一些实施方案中,式(I)化合物选自式(I-A)-(I-D)化合物:
其中,环A、R1、m、R4R2、R3、x如前所定义。
在一些实施方案中,A环选自5元或6元杂环或苯并5元或6元杂环,优选地,A环选自:
其中,Z1、Z2、Z4独立地选自N、CR7,各个Z3独立地选自NR7、O、S;且Z1、Z2、Z4不同时为CR7;各R7独立选自氢、卤素、氰基、羟基、氨基、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烷氧基、卤代C1-C6烷基、被C1-C6烷基取代的氨基、被卤代C1-C6烷基取代的氨基、被C1-C6烷酰基取代的氨基、C1-C6烷酰基、C1-C6烷氧羰基、C3-C6环烷基、4-8元杂环烷基、C6-10芳基、5-10元杂芳基、C6-C10芳基C1-C4烷基氧基或者5-10元杂芳基C1-C4烷基氧基;优选地,各R7独立选自氢、卤素(特别是溴)、氨基、羟基、氰基、羧基、C1-C3烷基(特别是甲基、乙基、异丙基)、卤代C1-C3烷基(特别是三氟甲基)、C1-C3烷氧基(特别是甲氧基、乙氧基)、C1-C3烷酰基(特别是甲酰基、乙酰基)、氨基甲酰基(-CONH2)、 甲酰基氨基、乙酰基氨基、甲氨基、乙氨基、N,N-二甲基氨基、2,2,2-三氟乙基氨基、C1-C3烷氧基羰基(特别是甲氧基羰基、乙氧羰基)、C3-C5环烷基(特别是环丙基)、苯基、吡啶基、吡咯烷基、哌啶基、吗啉基或苄氧基;
更优选地,A环选自:
各R7独立选自氢、卤素、氰基、羟基、氨基、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烷氧基、卤代C1-C6烷基、被C1-C6烷基取代的氨基、被卤代C1-C6烷基取代的氨基、被C1-C6烷酰基取代的氨基、C1-C6烷酰基、C1-C6烷氧羰基、C3-C6环烷基、C3-C6环烷基、4-8元杂环烷基、C6-10芳基、5-10元杂芳基、C6-C10芳基C1-C4烷基氧基或者5-10元杂芳基C1-C4烷基氧基;优选地,各R7独立选自氢、卤素(特别是溴)、氨基、羟基、氰基、羧基、C1-C3烷基(特别是甲基、乙基、异丙基)、卤代C1-C3烷基(特别是三氟甲基)、C1-C3烷氧基(特别是甲氧基、乙氧基)、C1-C3烷酰基(特别是甲酰基、乙酰基)、氨基甲酰基(-CONH2)、甲酰基氨基、乙酰基氨基、甲氨基、乙氨基、N,N-二甲基氨基、2,2,2-三氟乙基氨基、C1-C3烷氧基羰基(特别是甲氧基羰基、乙氧羰基)、C3-C5环烷基(特别是环丙基)、吗啉基、苯基、吡啶、吡咯烷基、哌啶基或苄氧基。
在一些实施方案中,R1为任选地被一个或多个R6取代的苯基;R6定义如前所述。
在一些实施方案中,R1为任选地被一个或多个R6取代的萘基;R6定义如前所述。
在一些实施方案中,R2和R3之一为氢,或者R2和R3都为氢。
在一些实施方案中,R1为任选地被一个或多个R6取代的苯基,且R2和R3之一为氢,或者R2和R3都为氢。
在一些实施方案中,R1为任选地被一个或多个R6取代的萘基,且R2和R3之一为氢,或者R2和R3都为氢。
在一些实施方案中,环B为6元碳环;R1为任选地被一个或多个R6取代的苯基;R2和R3之一为氢,或者R2和R3都为氢。
在一些实施方案中,环B为6元碳环;R1为任选地被一个或多个R6取代的萘基;R2和R3之一为氢,或者R2和R3都为氢。
在一些实施方案中,环B为5元碳环;R1为任选地被一个或多个R6取代的苯基;R2和R3都为氢。
在一些实施方案中,环B为7元碳环;R1为任选地被一个或多个R6取代的苯基;R2和R3都为氢。
在一些实施方案中,式(I)化合物选自下列化合物:
其中,x、Z1、Z2、Z3、Z4、R2、R3、R6定义如前所述。
在一些实施方案中,式(I)化合物选自下列化合物:

其中,R2、R3、R6、R7定义如前所述;
且苯基、吡啶基、萘基、喹啉基、嘧啶基、吡嗪基或哒嗪基和-NR2R3 连接于它们相连的碳环的同一个环碳原子。
在一些实施方案中,式(I)化合物选自下列化合物:

其中,R2、R3、R6、R7定义如前所述。
除非另有说明,否则本文中各基团的定义如下:
如本文所用,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。
如本文所用,“烷基”是指直链或支链的饱和烃基,例如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、丁基或异丁基。
如本文所用,“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,“卤代C1-C4烷基”依此类推,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。
如本文所用,“C1-C6烷氧基”指含有1-6个碳原子的直链或支链烷氧基,“C1-C4烷氧基”、“C1-C3烷氧基”依此类推,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、正丁氧基或异丁氧基。
如本文所用,“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一个或多个相同或不同的卤原子取代,“卤代C1-C4烷氧基”依此类推,例如-OCF3、-OCH2CH2Cl、-OCHBrCH2Cl、或-OCF2CF3等。
如本文所用,“C1-C6烷酰基”是指1-6个碳原子的直链或支链烷酰基,“C1-C4烷酰基”、“C1-C3烷酰基”依此类推,例如,甲酰基、乙酰基、丙酰基、丁酰基、异丁酰基、戊酰基等,优选为甲酰基、乙酰基、丙酰基。
如本文所用,“卤代C1-C6烷酰基”是指1-6个碳原子的直链或支链烷酰基被的氢原子被一个或多个相同或不同的卤原子取代,“卤代C1-C4烷酰基”、“卤代C1-C3烷酰基”依此类推。
如本文所用,“氧代”是指“=O”,即氧原子通过双键与其它原子连接。
如本文所用,“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-C16烷基取代,例如-NHMe、-NHEt、-N(Me)Et或-NEt2等。
如本文所用,“被C1-C6烷酰基取代的氨基”是指氨基上的氢原子被1个或2个相同或不同的C1-C6烷酰基取代,例如-NHCHO、-NHCOCH3或-NHCOCH2CH3等。
如本文所用,“C1-C6烷氧基C1-C6烷基”是指C1-C6烷氧基的氧原子与C1-C6烷基连接,如-CH2OCH2CH3、-CH2CH2OCH2CH3或-CH2CH2OCH3等。
如本文所用,“C1-C6烷氧基羰基”是指C1-C6烷氧基的氧原子与羰基连接,例如-C=OOCH2CH3、-C=OOCH2CH2CH3、-C=OOCH2CH(CH3)2等。
如本文所用,“C3-C6环烷基”是指含有3-6个碳原子的饱和环烃基,如环丙基、环丁基、环戊基或环己基等。
如本文所用,“羟基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羟基连接,如-CH2OH、-CH2CH2OH、-CH(OH)CH3、-CH2CH2CH2OH、-CH2CH2CH2CH2OH或-CH2CH(CH3)CH2OH等。
如本文所用,“C6-C14芳基”是指包含6-14个环原子但环原子中不含杂原子的单环或 多环芳香族环基,“C6-C12芳基”依此类推,例如苯基、萘基。
如本文所用,“3-10元碳环”指含3-10个碳原子的饱和或不饱和的单环烃基团,碳环优选具有5-8个环碳原子,更优选具有5-6个碳原子,例如环丙烷、环丁烷、环戊烷、环戊烯、环己烷、环己烯、环庚烷、环庚烯等。
如本文所用,“C6-C14芳基C1-C6烷基”是指C6-C14芳基与C1-C6烷基连接,例如苄基、苯乙基、苯丙基等。
如本文所用,“C3-C6环烷基C1-C6烷酰基”是指C3-C6环烷基的环碳与C1-C6烷酰基的烷基碳连接,例如环丙基甲酰基、环丙基乙酰基、环丁基甲酰基、环戊基甲酰基等。
如本文所用,“C6-C14芳基C1-C6烷酰基”是指C6-C14芳基与C1-C6烷酰基的烷基碳连接,例如苯甲酰基、苯乙酰基等。
如本文所用,“C1-C6烷磺酰基”C1-C6烷基与磺酰基-S(=O)2-连接,例如甲基磺酰基、乙基磺酰基等。
如本文所用,“C1-C6烷基亚砜基”是指C1-C6烷基与亚砜基(>S=O)连接,例如,甲基亚砜基、乙基亚砜基、丙基亚砜基、丁基亚砜基、叔丁基亚砜基等。
如本文所用,“C3-C6环烷基C1-C6烷基”是指C3-C6环烷基与C1-C6烷基连接,例如环丙基甲基、环丁基甲基等。
如本文所用,“杂环”是指含有至少一个选自N、O和S的杂原子作为环成员的单环或多环基团,其可以是芳香性或非芳香性基团,本文优选为单环基团;4-10元杂环是指包含4至10个环原子的杂环基,例如吡啶基、哌啶基、吗啉基、呋喃基、噻吩基、噻唑基、咪唑基、吡咯基、吡嗪、哒嗪、嘧啶基。
如本文所用,“杂环烷基”是指含有至少一个选自N、O和S的杂原子作为环成员饱和单环或多环基团,3-9元杂环烷基包括氮杂环丁烷基、吡咯烷基、哌啶基、氮杂环庚烷基、吗啉基等。
如本文所用,“杂芳基”是指含有至少一个选自氮、氧或硫的杂原子作为环成员的单环或双环芳香环基团;5-10元杂芳基包括但不仅限于下列基团:吡咯基、咪唑基、吡唑基、1,2,3-三唑基、吡啶基、吡啶酮基、哒嗪基、嘧啶基、吡嗪基、三嗪基、噁唑基、异噁唑基、噁二唑基、噻唑基、异噻唑基、噻二唑基、喹啉基等。
本文所用的“任选”、“任选的”或“任选地”意指随后描述的事件可以发生或可以不发生,并且该描述包括所述事件发生的情形以及所述事件不发生的情形。例如,“任选被取代的烷基”包括本文定义的“未取代的烷基”和“被取代的烷基”。“任选被卤素取代”包括“被卤素取代”的情形和“未被卤素取代”的情形,例如被0-3个卤素取代。本领域技术人员应 当理解的是,对于含有一个或多个取代基的任意基团而言,所述基团不包括任何在空间上不切实际的、化学上不正确的、合成上不可行的和/或内在不稳定的取代模式。
当本文中提及式(I)时,该称谓也包括其亚式,例如式(I-1-a)、(I-1-b)、(I-2-a)、(I-2-b)或(II-1)(II-2)等。
本发明通式(I)所示含芳基的胺类化合物及其几何异构体、构象异构体、互变异构体也包括溶剂合物形式,如水合物、醇合物等,并且所述的溶剂合物也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几何异构体、构象异构体、互变异构体的药学上可接受的盐是指通式(I)所示的含芳基的胺类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。
本领域技术人员将认可,本发明的化合物可以含有手性中心,照此可以存在不同的异构形式。如本文所用的“异构体”指具有相同分子式、但是原子的排列和构型有区别的不同化合物。
“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,它可分为顺反异构体、对映异构体两种,也可分为对映异构体和非对映异构体两大类。
“对映异构体”是相互为不可重叠的镜像的一对立体异构体。一对对映异构体的11混合物是“外消旋”混合物。合适时,该术语用于指外消旋混合物。当指示本发明的化合物的立体化学时,采用常规的RS系统指定了具有两个手性中心的已知的相对和绝对构型的单一立体异构体(例如(1S,2S));具有已知的相对构型、但是绝对构型未知的单一立体异构体标示了星号(例如(1R*,2R*));具有两个字母的外消旋物(例如(1RS,2RS)为(1R,2R)和(1S,2S)的外消旋混合物;(1RS,2SR)为(1R,2S)和(1S,2R))的外消旋混合物。“非对映异构体”是具有至少两个不对称原子、但是相互不为镜像的立体异构体。根据Cahn-lngold-Prelog R-S系统指明绝对立体化学。当化合物是纯的对映异构体时,各手性碳处的立体化学可以通过R或S说明。绝对构型未知的被拆分的化合物可以根据它们在钠D线波长处旋转平面偏振光的方向(右旋或左旋)指定为(+)或(-)。或者,被拆分的化合物可以通过相应的对映异构体 /非对映异构体经由手性HPLC的各自的保留时间来定义。
当化合物含有双键或一些其它的使得分子具有一定量结构刚性的特征时,可以发生几何异构体。如果化合物含有双键,则取代基可以是E或Z构象。如果化合物含有二取代的环烷基,则环烷基取代基可以具有顺式或反式构型。
“构象异构体”是通过有关一个或多个价键的旋转而不同的异构体。
“互变异构体”是指质子从分子的一个原子转移至相同分子的另一个原子而形成的异构体。本发明的化合物的所有互变异构形式也将包含在本发明的范围内。
“多晶型物”指具有相同的化学结构/组成、但是形成结晶的分子和/或离子的空间排列不同的结晶形式。本发明的化合物可以作为无定型固体或结晶固体来提供。冷冻干燥法可用于提供固体的本发明的化合物。
“溶剂合物”指本发明的化合物与一种或多种有机或无机溶剂分子的物理联合物。这种物理联合物包括氢键。在一些情况中,溶剂合物将能够分离,例如当一种或多种溶剂分子掺入结晶固体的晶格中时。溶剂合物中的溶剂分子可以以有规排列和/或非有序排列存在。溶剂合物可以包含化学计算量或非化学计算量的溶剂分子。“溶剂合物”包括溶液相和可分离的溶剂合物。示例性的溶剂合物包括但不限于水合物、乙醇化物、甲醇化物和异丙醇化物。溶剂化的方法是本领域公知的。
本发明还包括本发明的化合物或其药学上可接受的盐的所有适宜的同位素变体。本发明的化合物或其药学上可接受的盐的同位素变体被定义为其中至少一个原子被具有相同原子数、但原子质量与自然界经常发现的原子质量不同的原子所替换的那些。可以掺入到本发明的化合物及其药学上可接受的盐中的同位素包括但不限于H、C、N和O的同位素,例如2H、3H、11C、13C、14C、15N、17O、18O、35S、18F、36Cl和125I。本发明所述化合物或其药学上可接受的盐的同位素变体可以通过常规技术、采用适宜试剂的适当同位素变体来制备。
根据本发明,式(I)所示的含芳基的胺类化合物选自如下结构:



















其中,如本领域中常规所理解的,“Bn”表示苄基,“Bz”表示苯甲酰基,“Me”表示甲基,“Et”表示乙基,“Ph”表示苯基。
根据本发明的第二个方面,提供制备式(I)所示的含芳基的胺类化合物的方法,所述方法可通过如下方法1-3之一或其组合进行:
方法1:
如反应式1所示,包括以下步骤:
a)式(II)所示化合物与式(III)所示化合物进行缩合反应,生成式(IV)所示化合物;
b)式(IV)所示化合物和式(V)所示化合物进行亲核加成反应,生成式(I-a)所示化合物;
其中A环、B环、R1同上文所定义和优选;
G代表离去基团,如C1-C6烷基亚磺酰基、苯亚磺酰基、萘亚磺酰基、苄基,上述C1-C6烷基亚磺酰基、苯亚磺酰基、萘亚磺酰基、苄基可选地被一个或多个选自卤素、C1-C6烷基、硝基、羟基、氨基、C1-C6烷酰基、C1-C6烷氧基、苯基的基团进一步取代;G优选为C1-C4烷基亚磺酰基、苯亚磺酰基、萘亚磺酰基、苄基,上述C1-C4烷基亚磺酰基、苯亚磺酰基、萘亚磺酰基、苄基可选地被一个或多个选自卤素、C1-C4烷基、硝基、羟基、氨基、C1-C4烷酰基、C1-C4烷氧基、苯基的基团进一步取代;G更优选为叔丁基亚磺酰基、对甲苯亚磺酰基、三氟甲基亚磺酰基、对溴亚磺酰基、苄基、对甲氧基苄基或 三苯基甲基;
M代表离去基团,如金属元素、卤素、金属合物、硼烷、硅烷、重氮盐等,优选-MgBr,-MgCl,-Li;
步骤a)可以在有或无酸存在下、在溶剂中进行,所述溶剂可以选自醚类,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;芳香类,如:苯、甲苯、硝基苯、氯苯等;醇类:甲醇、乙醇、异丙醇、丁醇、叔丁醇、乙二醇;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;其他类,如:二甲亚砜、乙腈等,或上述溶剂的混合物;
所述酸可以选自有机酸、无机酸或路易斯酸,无机酸可以包括:盐酸、氢溴酸、氢碘酸、硫酸、硝酸、磷酸、高氯酸;有机酸可以包括:甲酸盐、乙酸、三氟乙酸、丙酸、丙酮酸、羟乙酸、乙二酸、丙二酸、丁二酸、戊二酸、马来酸、富马酸、乳酸、苹果酸、柠檬酸、酒石酸、苦味酸、谷氨酸、苯甲酸、甲磺酸、乙磺酸、苯磺酸、对甲苯磺酸、水杨酸、抗坏血酸、樟脑酸或樟脑磺酸;路易斯酸包括:氯化铝、氯化铁,三氟化硼,钛酸乙酯等;这些酸可以单独使用,或是两种或多种联合使用;
步骤b)可以在溶剂中进行,所述溶剂可以选自醚类,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;芳香类,如:苯、甲苯、硝基苯、氯苯等;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;其他类,如:二甲亚砜、乙腈等,或上述溶剂的混合物;
方法2如反应式2所示,包括以下步骤:
c)式(II)所示化合物与式(V)所示化合物进行亲核加成反应,生成式(VI)所示化合物;
d)式(VI)所示化合物和叠氮化物进行取代反应,生成式(VII)所示化合物;
e)式(VII)所示化合物发生还原反应生成式(I-a)所示化合物;
其中A环、B环、R1同上文所定义和优选;
M代表离去基团,如金属元素、卤素、金属合物、硼烷、硅烷、重氮盐等,优选-MgBr,-MgCl,-Li;
步骤c)可以在溶剂中进行,所述溶剂可以选自醚类,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;芳香类,如:苯、甲苯、硝基苯、氯苯等;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;其他类,如:二甲亚砜、乙腈等,或上述溶剂的混合物;
步骤d)可以在酸催化条件下在溶剂中进行,所述酸可以包括:盐酸、氢溴酸、氢碘酸、硫酸、硝酸、磷酸、三氟乙酸、乙酸等;所述叠氮化物包括:叠氮化钠、叠氮化钾、三甲基叠氮硅烷等;反应溶剂可选自水;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;芳香类,如:苯、甲苯、硝基苯、氯苯等;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;其他类,如:二甲亚砜、乙腈、乙酸、甲酸、特戊酸等,或上述溶剂的混合物;
步骤e)可以在还原剂条件下在溶剂中进行,所述还原剂体系包括:Pd/C催化剂催化加氢、Pd/BaSO4催化剂催化加氢、PtO2催化剂催化加氢、Raney Ni催化剂催化加氢、PPh3-THF-H2O体系、LAH等;所需溶剂选自水,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、硝基苯、氯苯等;醇类:甲醇、乙醇、异丙醇、丁醇、叔丁醇、乙二醇等,或上述溶剂的混合物;
方法3:
由方法1-2得到的式(I-a)化合物进行氨基的官能团转化得到其他式(I)所示含芳基的胺类化合物,如通过烷基化反应、酰化反应等;
所述烷基化反应可以在烷基化试剂存在下进行,所述烷基化试剂剂包括但不限于:碘甲烷、碘乙烷、2-溴丙烷、溴代环丙烷、叔丁基溴等;
所述酰化反应可以在酰化试剂存在下进行,所述酰化试剂包括但不限于:醋酐、乙酰氯、苯甲酰氯、甲乙酐、丙酸酐、环丙烷甲酸酐等;
式(II)、式(III)、式(IV)、式(V)、式(VI)、式(VII)化合物是商业上可以得到的化合物或是按照技术上已知的方法制备或是按照相似化合物的方法制备。
上述各反应式中使用的起始化合物可以是合适的盐,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、就是酸盐、苦味酸盐、谷氨酸盐、甲磺酸盐、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。
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。
根据本发明的第三个方面,提供一种药物组合物,包含治疗有效量的选自式(I)所示化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,以及可选地一种或多种可药用载体、稀释剂或赋形剂。
本发明的化合物具有NMDA受体和/或单胺转运体和/或sigma受体多靶点作用,可用于各种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、癫痫等疾病。
因此,根据本发明的第四个方面,提供选自式(I)所示化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,或上述药物组合物在制备调节NMDA受体 和/或单胺转运体和/或sigma受体活性的药物中的应用,具体而言,在制备NMDA受体拮抗剂中的应用,在制备单胺转运体抑制剂中的应用,在制备sigma受体激动或拮抗剂中的应用。
根据本发明的第五个方面,还提供选自上述式(I)所示的含芳基的胺类化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,或上述药物组合物在制备预防和/或治疗与NMDA受体和/或单胺转运体和/或sigma受体相关联的疾病,特别是中枢神经系统疾病的药物中的应用。
所述中枢神经系统疾病选自:脑缺血;脑卒中;脑梗塞;创伤性脑损伤;抗NMDA受体脑炎;癫痫;肌萎缩侧索硬化症;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;神经性疼痛;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;抽动症以及由NMDA受体活化引起的其他神经系统事件或神经变性。
在一些实施方案中,所述神经性疼痛包括外周糖尿病性神经病、疱疹后神经痛、复杂局部疼痛综合征、外周神经病、化学治疗诱导的神经性疼痛、癌性神经性疼痛、神经性下背痛、HIV神经性疼痛、三叉神经痛和中枢性中风后疼痛。
在一些优先的实施方案中,所述中枢神经系统疾病选自: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;帕金森症;外周糖尿病性神经病;疱疹后神经痛;复杂局部疼痛综合征。
根据本发明的第六个方面,还提供一种用于治疗和/或预防与NMDA受体和/或单胺转运体和/或sigma受体相关联的疾病,特别是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式(I)表示的含芳基的胺类化合物、其立体异构体或其药学上可接受的盐。
根据本发明的第六个方面,还提供一种制备上述药物组合物的方法,包括将上述式(I)所示的含芳基的胺类化合物、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,包括但不限于: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(例如脂溶性或油溶性注射剂)等。
本发明化合物的“治疗有效量”指可以引起个体生物学或医学反应或改善症状、减慢或延缓疾病恶化或预防疾病等的本发明化合物的量。“治疗有效量”可以由参与医师或兽医执业者来确定,并且将随着化合物、所治疗的疾病状态、所治疗的疾病的严重程度、个体的年龄和相关健康状况、施用途径和形式、主治医师或兽医执业者的判断等因素而变化。
如本文所用,“个体”指动物。优选地,动物是哺乳动物。个体还指例如灵长类(例如人类)、牛、绵羊、山羊、马、狗、猫、兔、大鼠、小鼠、鱼、鸟等。在一优选实施方案中,个体是人。
如本文所用,“抑制”指特定的病患、症状或病症或疾病的减轻或抑制,或者生物学活性或过程基线活性的显著降低。
如本文所用,在一个实施方案中术语“治疗”任何疾病或病症指改善疾病或病症(即阻止或减缓疾病或其至少一种临床症状的发展)。在另一个实施方案中,“治疗”指改善至少一种身体参数,其可能不为患者所察觉。在另一个实施方案中,“治疗″指身体上(例如稳定可察觉的症状)或生理学上(例如稳定身体的参数)或上述两方面调节疾病或病症。
如本文所用,“预防”指给具有易患所述疾病的体质的个体施用一种或多种药物物质、特别是本发明的化合物和/或其可药用盐,用以防止个体罹患该疾病。
有益效果
本发明提供了一种全新的NMDAR拮抗剂,属于通道孔阻断剂,可抑制在病理性条件下NMDA过度激活而导致的通道开放从而避免Ca2+的过多内流,而不影响NMDAR的正常功能。本发明所述的NMDAR拮抗剂是一种可逆性的NMDAR拮抗剂,结合后解离非常迅速,而不影响NMDA受体的正常功能。
本发明化合物具有以下有益效果:
1)本发明化合物具有可调节NMDA受体活性,从而可用于治疗和/或预防与NMDA受体相关联的疾病。
2)体外研究结果显示,本发明化合物与NMDAR的解离速度较快,明显优于氯胺酮,提示本发明化合物具有更低的拟精神样不良反应风险,安全窗大。
3)本发明化合物对单胺转运体(5-HT转运体和/或去甲肾上腺素转运体和/或多巴胺转运体)具有良好的抑制作用,可以改善焦虑症、抑郁症或者其他中枢神经系统类疾病的焦虑、抑郁等共病。
4)本发明化合物对sigma受体具有良好的结合活性,可以通过调节中枢神经系统内的sigma受体发挥保护神经、调节认知、改善药物成瘾、运动功能障碍等作用。
5)本发明化合物具有口服生物利用度高、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,尤其对由NMDA受体活化引起的中枢神经系统事件疗效好。
总之,本发明化合物与现有的NMDAR拮抗剂相比,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,综合成药性好,具有良好的临床应用前景。
具体实施方式
下列实施例和药理实施例进一步阐明本发明,但并不限制本发明的范围。
除特殊说明外,在实施例和药理实施例中所采用的原料、试剂、方法等均为本领域常规的原料、试剂、方法。
缩略词
以下实施例涉及到手性分离的操作,手性分离条件如下:
色谱柱:DAICEL CHIRALCEL OD-H 4.6x250mm,5um;流动相:正己烷/0.1%二乙胺异丙醇=50/50;流速:0.8ml/min;柱温:35℃;检测波长:270nm。
不同化合物的手性分离,可以在上述条件的基础上,通过调整流动相比例或流速进行分离。
实施例
实施例1 6-苯基-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A1)的制备
步骤一:
化合物A1-1(765mg)溶于二氯甲烷(20mL),加入DMAP(444mg)、二碳酸二叔丁酯(1.5g),室温反应过夜,反应液浓缩,柱层析得化合物A1-2,类白色固体250mg。
步骤二:
50mL反应瓶中加入无水THF(5mL),苯基氯化镁(4.7mL)氮气保护,冷却到0-5℃,滴加化合物A1-2(500mg)的THF(5mL)溶液,室温反应过夜。饱和氯化铵水溶液淬灭反应,反应液经乙酸乙酯萃取,干燥,浓缩,柱层析纯化,得化合物A1-3约385mg。
步骤三:
化合物A1-3(500mg)溶于DCM(10mL)中,加叠氮钠(470mg,5eq),冰浴下滴加TFA(1.32g,8eq),冰浴下反应30min后,移至室温避光过夜。反应液中加冰水,用氨水调节pH至9,转移至分液漏斗中,DCM萃取,有机相用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,浓缩,得化合物A1-4,棕色固体350mg。
步骤四:
化合物A1-4(1.0g)溶于甲醇(25mL)中,加钯碳(10%,100mg),通氢气,室温反应过夜。反应液过滤,浓缩,二氯甲烷打浆得固体434mg,母液浓缩,柱层析纯化得固体172mg,共得化合物A1约606mg。LRMS-ESI(m/z):246.16[M+H]+.
1H NMR(400MHz,DMSO-d6)δ7.51(d,J=7.3Hz,2H),7.28(t,J=7.6Hz,2H),7.18(t,J=7.3Hz,1H),6.60(s,2H),2.98(d,J=16.0Hz,1H),2.58(d,J=16.0Hz,1H),2.49(m,1H),2.10(m,2H),1.91(br,2H),1.79(m,1H).
手性柱分离得到异构体A1-P1(R构型)和A1-P2(S构型):
A1-P1(R构型):HPLC纯度:>99%,保留时间:6.534min。手性纯度:100%,保留时间:9.186min。
A1-P2(S构型):HPLC纯度:>99%,保留时间:6.553min。手性纯度:100%,保留时间:14.432min。
实施例2 N-(2-氨基-6-苯基-4,5,6,7-四氢苯并噻唑-6-基)甲酰胺(化合物A2)的制备
50mL反应瓶中加入甲酸(920mg)、乙酸酐(2.04g),氮气保护,60℃回流反应2h,冷却到室温,加入二氯甲烷(40mL),制得0.5M甲乙酐溶液备用。取化合物A1(3.8g)加入二氯甲烷(50mL),滴加上述0.5M甲乙酐溶液(35mL),室温反应1h。用饱和碳酸氢钠水溶液调pH=9,二氯甲烷萃取,干燥,滤液浓缩,柱层析得化合物A2,类白色固体3.8g。
1H NMR(400MHz,DMSO-d6)δ8.28(s,1H),7.97(s,1H),7.38(m,2H),7.30(t,J=7.6Hz,2H),7.20(t,J=7.1Hz,1H),6.65(s,2H),2.99(dd,J=29.1,16.3Hz,2H),2.56(m,1H),2.40(m,1H),2.25(m,1H),2.16(m,1H).LRMS-ESI(m/z):274.16[M+H]+.
实施例3 N6-甲基-6-苯基-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A3)及其盐酸盐的制备
取化合物A2(3.8g)加入无水THF(60mL),滴加BMS(136mL),65℃反应过夜,冷却到室温,滴加甲醇淬灭,浓缩干溶剂,加入甲醇(50mL)、水(50mL)回流2h,DCM萃取,干燥,浓缩,柱层析纯化得碱式的A3,类白色固体(2.8g),加入甲醇(30mL)溶清,滴加 4M HCl/EA(3.2mL)搅拌反应,浓缩干溶剂,MTBE打浆,过滤,得化合物A3盐酸盐(2.9g)。
1H NMR(400MHz,DMSO-d6)δ10.36(m,1H),9.82(m,1H),9.26(br,2H),7.61(m,2H),7.46(m,3H),3.64(d,J=16.5Hz,1H),3.27(d,J=16.5Hz,1H),2.66(m,2H),2.52(m,1H),2.14(t,J=4.8Hz,3H),1.90(m,1H).LRMS-ESI(m/z):246.16[M+H]+.
手性柱分离得到异构体A3-P1(R构型)和A3-P2(S构型):
A3-P1(R构型):HPLC纯度:>98%,保留时间:6.914min。手性纯度:>95%,保留时间:6.847min。
A3-P2(S构型):HPLC纯度:>95%,保留时间:6.924min。手性纯度:>95%,保留时间:6.782min。
实施例4N-(2-氨基-6-苯基-4,5,6,7-四氢苯并噻唑-6-基)乙酰胺(化合物A4)的制备
取化合物A1(400mg)加入二氯甲烷(20mL),滴加乙酸酐(167mg,1.1eq),25℃反应过夜。用饱和碳酸氢钠水溶液调pH=9,二氯甲烷萃取,干燥,浓缩,柱层析得化合物A4,类白色固体(359mg,收率76.5%)。
实施例5 N6-乙基-6-苯基-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A5)及其盐酸盐的制备
化合物A4(320mg)加入二氯甲烷(20mL),滴加TMSCl(500mg,5eq),搅拌反应30min, 加入四氢铝锂(480mg,12eq),25℃反应5h。2M氢氧化钠水溶液(1mL)淬灭反应,加入无水硫酸镁、二氯甲烷搅拌,过滤,浓缩柱层析得白色固体(247mg),加入乙酸乙酯(3mL),滴加2M氯化氢乙酸乙酯溶液(0.5mL)搅拌反应30min,过滤,得化合物A5盐酸盐(250mg)。
1H NMR(400MHz,DMSO-d6)δ9.96(s,1H),9.33(s,1H),7.57(m,J=7.4Hz,2H),7.44(m,3H),6.81(s,2H),3.61(d,J=15.8Hz,1H),3.21(d,J=15.1Hz,1H),2.78(m,1H),2.60(m,1H),2.43(m,2H),2.27(m,1H),1.71(m,1H),1.14(t,J=6.8Hz,3H).LRMS-ESI(m/z):274.17[M+H]+.
实施例6 N-(2-氨基-6-苯基-4,5,6,7-四氢苯并噻唑-6-基)丙酰胺(化合物A6)的制备
取化合物A1(400mg)加入二氯甲烷(20mL),滴加丙酸酐(234mg,1.1eq),25℃反应过夜。饱和碳酸氢钠水溶液调pH=9,二氯甲烷萃取,无水硫酸钠干燥,浓缩,柱层析得化合物A6,类白色固体(274mg,收率55.8%)。
实施例7 6-苯基-N6-丙基-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A7)及其盐酸盐的制备
取化合物A6(274mg)加入二氯甲烷(20mL),滴加TMSCl(990mg,10eq),搅拌反应30min,加入四氢铝锂(968mg,28eq),回流反应24h。2M氢氧化钠水溶液(1mL)淬灭反应,加入无水硫酸镁,二氯甲烷搅拌,过滤,浓缩柱层析得白色固体(229mg),加入乙酸乙酯(3mL),滴加2M氯化氢乙酸乙酯溶液(0.5mL)搅拌反应30min,过滤,得化合物A7盐酸盐(240mg)。
LRMS-ESI(m/z):288.19[M+H]+.1H NMR(400MHz,DMSO-d6)δ9.62(s,1H),9.14(s,1H),7.55(m,2H),7.43(m,3H),6.78(s,2H),3.64(d,J=16.3Hz,1H),3.15(d,J=16.2Hz,1H),2.71(m,1H),2.65-2.31(m,3H),2.19(m,1H),1.73(m,1H),1.56(m,2H),0.80(t,J=7.4Hz,3H).
实施例8 6-(2-氟苯基)-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A9)的制备
步骤一:
邻氟溴苯(5g,1eq)、氯化锂(0.6g,0,5eq)加入到THF(30mL)中,氮气保护下,降温至-10℃以下,加入2.0M异丙基氯化镁THF溶液(14.3mL,1eq),加入完毕,控制-5--10℃搅拌3h。
另取A1-2(1g,0.13eq)分散到10mL THF中,滴到上述体系中,控温-5--10℃反应2h。加入甲醇和水淬灭,加入EA(100mL)搅拌,过滤,滤液用水(50mL)洗涤,无水硫酸钠干燥,浓缩,柱层析纯化得A9-1,类白色固体0.95g(收率70.5%)。
步骤二:
900mg A9-1溶于DCM(10mL)中,加叠氮钠(0.83g,5eq),冰浴下滴加TFA(9mL),冰浴下反应30min后,移至室温避光过夜。加冰水(50mL),用氨水调节pH至9,DCM萃取,有机相用饱和氯化钠水溶液洗涤,无水硫酸钠干燥,浓缩,柱层析后得A9-2,70mg。
步骤三:
A9-2(70mg)溶于甲醇(70mL)中,加10%钯碳(50mg),通氢气,室温反应过夜。过滤,滤液浓缩,柱层析得化合物A9,白色固体38mg,收率59%。
1H NMR(400MHz,DMSO-d6)δ7.47(t,J=16.3Hz,1H),7.31-7.26(m,1H),7.17-7.11(m,2H),6.63(s,2H),3.10(d,J=16Hz,1H),2.62-2.50(m,2H),2.35-2.31(m,1H),2.16-2.12(m,1H),1.82-1.78(m,1H).ESI(m/z):264.13[M+H]+.HPLC纯度:>97%,保留时间:6.229min。
实施例9 5-苯基-5-(吡咯烷-1-基)-4,5,6,7-四氢-1H-吲唑(化合物A12)及其盐酸盐的制备
步骤一:三氮唑(12.4g,1.1eq)、四氢吡咯(13.35g,1.2eq)、1,4-环己二酮单乙二醇缩酮A12-1(25.0g,160mmol)溶于甲苯中,回流分水18h,温度降至室温,得到A12-2溶液备用。
步骤二:溴苯(75g,3eq)溶于无水THF中,-78℃缓慢滴入正丁基锂(2.5M)(150mL,3.3eq),滴加完毕,-78℃保温1h,滴入上述制备的A12-2溶液,滴加完毕后,保温搅拌1h,室温搅拌12h,柱层析得A12-3,10.5g。
步骤三:化合物A12-3(10.5g,6.5mmol)溶于乙醇中,加入浓盐酸(12M,3eq),室温搅拌过夜,浓缩干溶剂,饱和碳酸氢钠水溶液调节pH=11-12,DCM萃取,干燥过滤浓缩,柱层析得化合物A12-4,8.5g。
步骤四:
化合物A12-4(3.0g,11.1mmol)溶于甲苯中,加入无水硫酸镁干燥2天,过滤待用;甲苯中加入叔丁醇钠(2.46g,2eq),加入甲酸乙酯(1.38g,1.5eq),0-10℃下加入上述A12-4溶液,室温搅拌过夜。将反应液用甲苯萃取2次,水相调节pH=6-7,二氯甲烷萃取3次,干燥,过滤浓缩柱层析得化合物A12-5,2.5g。
步骤五:
化合物A12-5(200mg,7.3mmol)溶于乙醇中,加入水合肼0.5mL,加热回流7h。浓缩干溶剂,残余物柱层析得A12约60mg,将上述柱层析得到的产物溶于甲基叔丁基醚中,加入市售4M的氯化氢/二氧六环溶液(1eq,0.056mL),析出固体,过滤烘干得化合物A12盐酸盐25mg。
LRMS-ESI(m/z):268.49[M+H]+.1H NMR(400MHz,DMSO-d6)δ11.67(s,1H),7.75(m,2H),7.64(s,1H),7.42(m,3H),3.87(d,J=14.2Hz,1H),3.48(m,1H),3.32(d,J=15.3Hz,1H),3.21(m,1H),3.10(m,1H),3.00(m,2H),2.83(dd,J=16.8,5.1Hz,1H),2.55(m,1H),2.13(m,1H),1.83(m,2H),1.66(m,2H).
手性柱分离得到异构体A12-P1(S构型)和A12-P2(R构型):
A12-P1(S构型):HPLC纯度:>99%,保留时间:8.786min。手性纯度:>99%,保留时间:7.703min。
A12-P2(R构型):HPLC纯度:>99%,保留时间:8.778min。手性纯度:>95%,保留时间:10.236min。
实施例10 N-甲基-6-苯基-4,5,6,7-四氢苯并噻唑-6-胺(化合物A13)及其盐酸盐的制备
将化合物A3(450mg)加入到85%的磷酸中(25mL),降温至-10℃,加入亚硝酸钠(6eq)的水溶液,-10℃搅拌1h,然后滴加到0℃的50%的次磷酸溶液中(7mL),0℃下搅拌1.5h,饱和碳酸钠中和至pH为8-9,EA萃取三次,合并有机相,浓缩,柱层析得到的A13样品加入到50mL的EA中,加入HCl/EA溶液成盐,得到化合物A13盐酸盐,190mg。
LRMS-ESI(m/z):245.16[M+H]+.1H NMR(400MHz,DMSO-d6)δ10.32(m,1H),9.75(m,1H),8.95(s,1H),7.57(d,J=6.9Hz,2H),7.41(m,3H),4.03(d,J=16.7Hz,1H),3.48(d,J=16.7Hz,1H),2.91(d,J=16.4Hz,1H),2.72(m,1H),2.55(m,1H),2.19(t,J=5.2Hz,3H),2.10(m,1H).
手性柱分离得到异构体A13-P1(R构型)和A13-P2(S构型):
A13-P1(R构型):HPLC纯度:>95%,保留时间:10.144min。手性纯度:>99%,保留时间:4.501min。
A13-P2(S构型):HPLC纯度:>95%,保留时间:10.151min。手性纯度:>99%, 保留时间:6.418min。
实施例11 6-苯基-4,5,6,7-四氢苯并噻唑-6-胺(化合物A14)及其盐酸盐的制备
将化合物A1(300mg)加入到浓盐酸(12M,8mL)中,搅拌至溶清后,降温至-30℃,滴加1M的亚硝酸钠水溶液(1.7eq,2mL),加入后保温搅拌1h,加入次磷酸(0.2mL),冰水浴下搅拌1h后,降温至-30℃加入饱和碳酸钠水溶液中和,二氯甲烷-甲醇萃取,无水硫酸钠干燥后,浓缩至干,柱层析纯化得到60mg的A14样品,加入2mL的EA,滴加氯化氢乙酸乙酯溶液,析出固体,过滤,干燥得到化合物A14盐酸盐,35mg。
LRMS-ESI(m/z):231.12[M+H]+.1H NMR(400MHz,DMSO-d6)δ8.97(s,1H),8.93(br,3H),7.58(d,2H),7.40(m,3H),3.78(d,J=16.6Hz,1H),3.47(d,J=16.6Hz,1H),2.90(m,1H),2.59(m,1H),2.44(m,1H),2.27(m,1H).
手性柱分离得到异构体A14-P1(R构型)和A14-P2(S构型):
A14-P1(R构型):HPLC纯度:>98%,保留时间:9.514min。手性纯度:100%,保留时间:7.835min。
A14-P2(S构型):HPLC纯度:>95%,保留时间:9.607min。手性纯度:100%,保留时间:21.014min。
实施例12 2-甲基-N-(4-苯基-4,5,6,7-四氢苯并噻吩-4-基)丙烷-2-磺胺(化合物A15)的制备
步骤一:反应瓶中加入A15-1(5g,1eq)、叔丁基亚磺酰胺(5.57g,1.4eq)、钛酸 四乙脂(8.24g,1.1eq)、THF,氮气保护,回流反应24h(70℃)。冷却到室温,反应液倒入冰盐水中,加入EA搅拌,过滤,分液保留有机相,水相用EA萃取,合并有机相,饱和食盐水洗,干燥浓缩,柱层析得化合物A15-2,2.69g,收率32.1%。
步骤二:碘苯(1.0g,2.5eq)加入DCM(10mL)中,氮气保护,降至-60℃,滴加丁基锂(1.64mL,2.5M,2.1eq),保温反应1.5h,记作反应液2,待用。化合物A15-2(500mg,1eq)加入DCM(10mL)中溶清,记作反应液1,待用。反应液1控温-60℃左右滴加入反应液2中,保温反应2h。氯化铵淬灭反应,EA萃取,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析得到化合物A15,322mg固体,收率49.3%。
实施例13 4-苯基-4,5,6,7-四氢苯并噻吩-4-胺(化合物A16)的制备
取化合物A15(322mg,1eq)加入二氧六环(9.7mL)中,滴加盐酸乙酸乙酯溶液(0.73mL,4M,3eq),室温反应0.5h。加入饱和碳酸氢钠水溶液中和,EA萃取,饱和食盐水洗,干燥浓缩,柱层析得到化合物A16,192mg,收率86.7%。加入2mLDCM,滴0.3mL 4M的氯化氢乙酸乙酯溶液,浓缩,异丙醇打浆,过滤得到盐酸盐形式的A16,HPLC:96.7%。
1H NMR(400MHz,DMSO-d6)δ9.14(s,3H),7.49(d,J=5.3Hz,1H),7.44-7.33(m,3H),7.30-7.22(m,2H),7.03(d,J=5.3Hz,1H),2.85(m,2H),2.23(t,J=4.8Hz,1H),1.95(m,1H),1.49(m,1H).
实施例14 N-(4-苯基-4,5,6,7-四氢苯并噻吩-4-基)甲酰胺(化合物A17)的制备
反应瓶中加入无水甲酸(347mg,9eq)和乙酸酐(770mg,9eq)中,氮气保护升温至60℃反应2h后降至室温(25℃左右),加入化合物A16(192mg,1eq)的DCM(4mL)溶液,室温下搅拌0.5h。加入饱和碳酸氢钠水溶液淬灭反应,DCM萃取,浓缩,得到226mg油状液体,为化合物A17。
实施例15 N-(4-苯基-4,5,6,7-四氢苯并噻吩-4-基)甲酰胺(化合物A18)及其盐酸盐的制备
取化合物A17(226mg,超理论,1eq)加入THF(10mL)溶液中,氮气保护下滴加1.0M的硼烷四氢呋喃溶液(8.4mL,10eq,1.0M)升温至回流反应3h。加入甲醇淬灭反应,浓缩至干,加入甲醇和水1∶1(5mL)升温至回流,TLC检测反应完毕,降至室温DCM萃取,浓缩得到A18粗品,加入2mL DCM,滴0.3mL 4M的氯化氢乙酸乙酯溶液,浓缩至干,加入异丙醇打浆,过滤,得到盐酸盐形式的A18,类白色固体150mg,收率73.6%。HPLC:98.3%。
1H NMR(400MHz,DMSO-d6)δ10.10(s,1H),9.75(s,1H),7.57(d,J=5.3Hz,1H),7.49-7.37(m,3H),7.36-7.31(m,2H),7.10(d,J=5.3Hz,1H),2.92-2.78(m,2H),2.41(t,J=5.2Hz,3H),2.35(dd,J=12.5,2.4Hz,1H),2.25(m,1H),1.94(m,1H),1.40(m,1H).
实施例16 6-苯基-4,5,6,7-四氢苯并噻吩-6-胺(化合物A19)的制备
步骤一:
100mL反应瓶中依次加入化合物A19-1(10g,1eq)、丙二酸(13g,1.4eq)和吡啶12mL,升温至100℃,反应6h。反应液冷却后,加入3M盐酸,调pH至3,析出固体,过滤,水淋洗,正庚烷淋洗(50mL*2),得化合物A19-2,13.6g类白色固体。
步骤二:
500mL反应瓶中,加入化合物A19-2(13.6g)、甲酸铵(22.47g,4eq)、10%钯碳(10g)和异丙醇(150mL),氮气保护,升温至90℃反应36h。反应液冷却至室温,垫 硅藻土滤除钯碳,浓缩大部分异丙醇,加入2M盐酸稀释(300mL,pH=2-3),EA萃取(200mL*2),硫酸钠干燥浓缩得化合物A19-3,10.5g固体,两步收率76.09%。
步骤三:
250mL反应瓶中加入多聚磷酸(PPA)(50g),升温至120℃,待PPA能搅拌起来,分批加入化合物A19-3(5g,1eq),保温反应1h。撤出油浴,趁热且缓慢向反应体系中加冰块淬灭反应(共加入冰水100mL),待料液冷却至室温后,加入EA(100mL)搅拌,垫硅藻土滤除黑渣,静置分层,水相再用50mL EA反萃一次,合并有机相,饱和碳酸氢钠洗涤一次(100mL),水洗(100mL),干燥,垫硅胶滤过,浓缩得化合物A19-4,棕黄色固体2.5g,收率56.56%。
步骤四:
250mL三口瓶中加入甲基三苯基溴化膦(19.40g,1.5eq)和无水四氢呋喃(40mL),控温0℃左右,滴加入2.5M丁基锂(2.10mL,1.4eq),加完室温反应1h。控温0℃左右,将化合物A19-4(5g,1eq)的四氢呋喃(10mL)溶液滴加入上述反应体系中,加完,室温反应过夜。向料液中加入冰水(100mL)和EA(100mL),垫硅藻土滤过,静置分液,有机相浓缩至最小体积,柱层析得化合物A19-5,1.1g油状物,收率22.31%。
步骤五:
100mL反应瓶中加入化合物A19-5(1.3g,1eq)和甲醇-水(体积比95/5,共39mL),冰水浴下,分批加入HTIB氧化剂(3.56g,0.95eq),加完,室温反应20min。反应液浓缩除大部分甲醇(35℃),加入DCM和水(各20mL),室温搅拌20min,分层,DCM相浓缩、柱层析纯化得化合物A19-6,630mg油状物,收率43.45%。
步骤六:
25mL反应瓶中,加入化合物A19-6(358mg,1eq)和DCM(5mL),氮气保护,控温0-10℃滴加入2M苯基氯化镁(1.18mL,1eq),加完后,保温反应30min。将反应液倒入饱和氯化铵中,分出DCM,干燥,浓缩,柱层析纯化得化合物A19-7,263mg油状物,收率48.52%。
步骤七:
50mL反应瓶中加入化合物A19-7(263mg,1eq)、叠氮化钠(1.11g,15eq)和DCM(26.3mL),氮气保护,冰水浴下,滴加入TFA(13mL),加完保温反应30min。反应液倒入冰水中(20mL),用氨水调pH至9,静置分出DCM相,干燥、柱层析纯化得到化合物A19-8。
步骤八:
将化合物A19-8溶于甲醇中(13mL),加入钯碳(0.52g),氢气置换,常压反应2h。反应液滤除钯碳,浓缩得化合物A19,90mg固体,两步收率34.35%。
LRMS-ESI(m/z):230.14[M+H]+.1H NMR(400MHz,CDCl3)δ7.54(d,J=7.3Hz,1H),7.35(t,J=7.6Hz,1H),7.25(m,1H),7.11(d,J=5.1Hz,1H),6.79(d,J=5.1Hz,1H),3.34(d,J=16.6Hz,1H),2.94(d,J=16.2Hz,1H),2.87-2.74(m,1H),2.61(dt,J=16.7,5.5Hz,1H),2.32-2.17(m,1H),2.04-1.92(m,1H).
手性柱分离得到异构体A19-P1(R构型)和A19-P2(S构型):
A19-P1(R构型):HPLC纯度:>93%,保留时间:13.149min。手性纯度:99%,保留时间:9.196min。
A19-P2(S构型):HPLC纯度:>97%,保留时间:13.279min。手性纯度:98.8%,保留时间:10.021min。
实施例17 N-(6-苯基-4,5,6,7-四氢苯并噻吩-6-基)甲酰胺(化合物A20)的制备
制备甲乙酐:10mL反应瓶中加入乙酸酐(0.51mL,28eq)和98%甲酸(0.21mL,28eq),65℃搅拌1-2h,冷却至室温,制得甲乙酐备用。将化合物A19(45mg,1eq,碱式)的DCM/THF混合溶液,冰水浴下滴加入自制的甲乙酐中,室温搅拌1h。向反应液中缓慢加入饱和碳酸氢钠水溶液,调pH至8,分出DCM相,浓缩,柱层析纯化得化合物A20,37mg胶状物,收率73.27%。
实施例18 N-甲基-6-苯基-4,5,6,7-四氢苯并噻吩-6-胺(化合物A21)及其盐酸盐的制备
将化合物A20(37mg,1eq)溶于THF(3.7mL)中,氮气保护,冰水浴下分批加入四氢铝锂(55mg,10eq),升温至回流反应3h。反应液冷却至0-10℃,滴加0.05mL水和0.05mL 20%氢氧化钠溶液,垫硅藻土滤过,浓缩至最小体积,加入1mL 4M氯化氢乙酸乙酯溶液,搅拌10min,浓缩,再加0.5mL EA搅拌析晶,过滤得盐酸盐形式的化合物A21,23mg类白色固体,收率57.5%。
1H NMR(400MHz,DMSO-d6)δ10.02(s,1H),9.53(s,1H),7.56(d,J=6.9Hz,2H),7.46-7.36(m,3H),7.31(d,J=5.1Hz,1H),6.70(d,J=5.1Hz,1H),3.90(d,J=16.5Hz,1H),3.35(d,J=16.5Hz,1H),2.73(m,1H),2.62(m,1H),2.41(m,1H),1.96(m,1H).LRMS-ESI(m/z):244.18[M+H]+.
实施例19 5-苯基-4,5,6,7-四氢苯并噻吩-5-胺(化合物A22)的制备
步骤一:
取A22-1(195mg,1.3mmol)于50mL三口瓶中,置换氮气后加入干燥的THF(5mL),降温至0℃,滴加PhMgCl(1mL,1.97mmol,1.5eq.),滴加完毕后0℃下反应2h。加饱和氯化铵水溶液淬灭反应,乙酸乙酯萃取,有机相用饱和食盐水洗涤后,干燥,过滤,浓缩至干,柱层析得到产品A22-2,黄色油状物115mg。
步骤二:
取A22-2(115mg,0.5mmol)溶解于DCM(3mL),加入NaN3(65mg,1mmol,2eq.),,置换氮气后降温至0℃,滴加TFA(0.2mL,2.56mmol,5eq.),滴加完毕后0℃下反应2.5h。冰水浴下加水稀释后加氨水淬灭反应,然后乙酸乙酯萃取,有机相用饱和食盐水洗涤后,干燥,过滤,浓缩得到A22-3。
步骤三:
取A22-3溶解于MeOH(10mL),加入Pd/C(150mg),置换氢气,室温反应2h。将反应液过滤除去Pd/C后浓缩、残余物溶解于MTBE(10mL),滴加4M的氯化氢/乙酸乙酯溶液(0.1mL),有白色固体析出,过滤得到化合物A22,灰白色固体30mg,三步收率8.8%。
1H NMR(400MHz,CD3OH)δ7.52-7.50(m,2H),7.46-7.38(m,3H),7.28(d,J=4.0Hz,1H),6.92(d,J=4.0Hz,1H),3.62(d,J=16Hz,1H),3.15(d,J=16Hz,1H),2.97-2.92(m,1H),2.64-2.59(m,1H),2.55-2.47(m,1H),2.44-2.38(m,1H).LRMS-ESI(m/z):213.17[M- NH2]+.
手性柱分离得到异构体A22-P1(S构型)和A22-P2(R构型):
A22-P1(R构型):HPLC纯度:>97%,保留时间:13.319min。手性纯度:99%,保留时间:10.137min。
A22-P2(S构型):HPLC纯度:>97%,保留时间:13.321min。手性纯度:98.8%,保留时间:10.871min。
实施例20 5-(N-甲基苯甲酰胺基)-5-苯基-4,5,6,7-四氢苯并噻吩-2-羧酸(化合物A23)的制备
步骤一:
反应瓶中,加入化合物A23-18g、N-甲基苄胺(6.83g,1.1eq)、1,2,3-三氮唑(4.25g,1.2eq)、甲苯50mL,回流分水过夜,降温至室温。另取反应瓶加入苯基溴化镁(4eq,in THF),冰浴下将上述反应液滴加入其中,搅拌90min,将反应液倒入氯化铵水溶液中,加EA,有机相水洗,干燥、浓缩、柱层析得化合物A23-2,淡黄色油状物3.85g,收率22%。
步骤二:
反应瓶中加入3.85g化合物A23-2,加入Pd(OH)2/C 460mg,加入乙醇50mL和甲酸铵6.75g(10eq),氮气置换,回流50min。滤除不溶物,滤渣用乙醇淋洗,合并滤液,浓缩,残液加DCM稀释,分别用水和饱和氯化钠溶液洗涤一次,干燥浓缩得化合物A23-3, 2.49g无色油状物,收率92%。
步骤三:
反应瓶中加入2.49g化合物A23-3,加入DCM,DIPEA 1.5eq,冰浴下加入苯甲酰氯1.1eq,加毕室温下反应过夜。加入EA稀释,分别用氯化铵水溶液和饱和氯化钠水溶液洗涤一次,干燥浓缩,再用15mL正庚烷打浆,得化合物A23-4,淡黄色固体2.97g。收率84%。
步骤四:
反应瓶中加入2.9g化合物A23-4,加入80mL丙酮和40mL水,加入0.2eq对甲苯磺酸,回流5h。加入EA稀释,分别用饱和碳酸氢钠水溶液和饱和氯化钠水溶液洗涤一次,干燥、浓缩、柱层析得化合物A23-5,无色油状物2.34g,收率92%。
步骤五:
将4eq DMF溶于30mL干燥DCM中,冰浴下搅拌20min,将3eq三氯氧磷滴加入体系中,加毕,室温下搅拌1h,再次冰浴下搅拌10min,将化合物A23-5(2g,1eq)溶于15mL干燥DCM中,快速加入上述反应液。氮气保护,自然升温至室温,搅拌过夜。将反应液倒入冰水中,调pH至弱碱性,分出有机相,饱和食盐水洗,干燥、浓缩、柱层析得化合物A23-6,淡黄色油状物1.04g。收率45%。
步骤六:
将钠块(3eq)加入到冰浴下的无水乙醇中,待完全反应后,加入2eq巯基乙酸乙酯,再加入化合物A23-6(1g,1eq)的无水乙醇溶液,氮气保护,室温下搅拌3h。直接加入5N氢氧化钠水溶液(按4eq NaOH计),60℃下搅拌4h。反应结束,加水稀释,加MTBE分层,弃去有机层,分出水层,用1M的盐酸调pH至2左右,加EA萃取,有机相再用饱和食盐水洗,干燥、浓缩得化合物A23,淡黄色固体1.03g。
实施例21 N-甲基-N-(5-苯基-4,5,6,7-四氢苯并噻吩-5-基)苯甲酰胺(化合物A24)的制备
反应瓶中加入600mg化合物A23,加入氧化亚铜0.5eq,DBU 0.4eq和少量DMF, 氮气置换多次,160℃反应5h,冷却反应液,加过量EA搅拌,滤除不溶物,滤渣用EA淋洗,滤液加EA稀释,分别用氯化铵水溶液和饱和食盐水洗涤一次,干燥浓缩柱层析得化合物A24,白色固体405mg。
实施例22 N-甲基-5-苯基-4,5,6,7-四氢苯并噻吩-5-胺(化合物A25)及其盐酸盐的制备
反应瓶中加入干燥THF和360mg化合物A24搅拌溶解,冰浴降温,分批多次加入2eq LAH,加毕氮气保护,自然升温至环境温度搅拌3h。将反应液倒入含有2.7g四水合酒石酸钾钠的冰水溶液中,加入EA萃取,有机相用饱和食盐水洗,干燥浓缩,碱性三氧化二铝柱层析,得无色油状物A25约220mg,溶于甲醇中,冰浴下滴加氯化氢/甲醇溶液(HCl约1eq),加毕,搅拌30min,浓缩,往残余物中加入适量乙腈,搅拌,析出白色固体,过滤,烘干得化合物A25盐酸盐,白色固体210mg。
1H NMR(400MHz,DMSO-d6)δ10.21(q,J=6.2Hz,1H),9.67(m,1H),7.58-7.50(m,2H),7.45-7.36(m,3H),7.34(d,J=5.1Hz,1H),6.94(d,J=5.1Hz,1H),3.72(dd,J=16.1,2.0Hz,1H),3.25(dd,J=16.0,2.3Hz,1H),2.87(dq,J=16.9,2.4Hz,1H),2.67(ddt,J=12.8,5.2,2.4Hz,1H),2.47(m,1H),2.19(t,J=5.3Hz,3H),2.14-2.00(m,1H).LRMS-ESI(m/z):244.14[M+H]+.
实施例23 7-(N-甲基甲酰胺)-7-苯基-4,5,6,7-四氢苯并噻吩-2-羧酸甲酯(化合物A27)的制备
步骤一:
制备甲乙酐:10mL反应瓶中加入乙酸酐(127.8mg,3eq)和无水甲酸(57.6mg,3eq),65℃搅拌1-2h,冷却至室温,得甲乙酐备用。
化合物A27-1(100mg)悬浮于DCM中,加入5eq DIPEA,冰浴,滴加入3eq自制的甲乙酐,加毕环境温度下反应40min,TLC检测反应结束。DCM稀释,稀盐酸洗,食盐水洗,干燥浓缩得化合物A27-2,类白色固体90mg。采用此法再次制备了1.1g化合物A27-2。
步骤二:
反应瓶中加入540mg化合物A27-2与2eq三乙胺,5mL DMF-DMA混合物,95℃下反应过夜。浓缩除去溶剂,残余物加DCM稀释,分别用氯化铵水溶液、饱和食盐水洗涤,干燥,浓缩得黄色油状物;另取反应瓶中加入将2.5eq DMF(按照A27-2计算)与干燥DCM混合,置于冰浴下搅拌,滴加入2.5eq三氯氧磷,加毕,室温下搅拌40min,将之前所得的若干黄色油状物溶于少量干燥DCM中,滴加入体系中,加毕氮气保护下室温搅拌3h。DCM稀释,氢氧化钠水溶液洗,饱和食盐水洗,干燥浓缩柱层析得化合物A27-3,淡黄色油状物613mg。收率94%。
步骤三:
将钠块(2.5eq)加入到冰浴下的无水甲醇中,待完全反应后,加入1.1eq巯基乙酸乙酯,再加入化合物A27-3(610mg,1eq)的无水甲醇溶液,室温下搅拌3h。取少量反应液,与氯化铵溶液混合,加EA萃取,有机相干燥浓缩,柱层析得标题化合物A27。
1H NMR(400MHz,Chloroform-d)δ8.03(s,1H),7.51(s,1H),7.45-7.30(m,5H),3.81(s,3H),2.78(s,3H),2.75(m,2H),2.50-2.41(m,2H),1.88(m,2H).
实施例24 7-(N-甲基甲酰胺)-7-苯基-4,5,6,7-四氢苯并噻吩-2-羧酸(化合物A28)的制备
向化合物A27的反应液中加入5M氢氧化钠水溶液(按4eq NaOH计),室温下搅拌过夜。加水稀释,加MTBE,分层,弃去有机层,分出水层用4M盐酸调pH至2左右,加DCM萃取,有机相再用饱和食盐水洗,干燥浓缩得化合物A28,淡黄色油730mg,收率100%。
LRMS-ESI(m/z):314.39[M-H]-.
实施例25 N-甲基-N-(7-苯基-4,5,6,7-四氢苯并噻吩-7-基)甲酰胺(化合物A29)的制备
化合物A28投料720mg,加入氧化亚铜0.5eq、DBU 0.4eq和少量DMF,氮气置换多次,160℃反应4h,冷却反应液,加过量EA搅拌,滤除不溶物,滤渣用EA洗,滤液加EA稀释,分别用饱和氯化铵水溶液和饱和食盐水洗涤一次,干燥浓缩柱层析得化合物A29,白色固体350mg,收率60%。
1H NMR(400MHz,Chloroform-d)δ8.01(s,1H),7.44-7.30(m,5H),7.22(d,J=5.2Hz,1H),6.79(d,J=5.2Hz,1H),2.76(s,3H),2.74(m,2H),2.53-2.40(m,2H),1.93(m,1H),1.83(m,1H).
实施例26 N-甲基-7-苯基-4,5,6,7-四氢苯并噻吩-7-胺(化合物A30)及其盐酸盐的制备
化合物A29投料350mg,溶于干燥THF中,冰浴降温,分批多次加入2eq四氢铝锂,加毕氮气保护,自然升温至室温搅拌40min。将反应液倒入四水合酒石酸钾钠的冰水溶液中,加入EA萃取,分层清晰,有机相再用饱和食盐水洗,干燥浓缩,中性三氧化二铝柱层析,分离得到无色油状物210mg,油状物A30溶于甲醇中,冰浴10min,滴加氯化氢/甲醇溶液(HCl约1eq),加毕,搅拌10min,减压浓缩至干,再加入2mL乙腈,环境温度下搅拌过夜,过滤,烘干得170mg化合物A30盐酸盐,白色固体。
1H NMR(500MHz,Methanol-d4)δ7.63(d,J=5.2Hz,1H),7.51-7.41(m,3H),7.36-7.33(m,2H),7.03(d,J=5.2Hz,1H),2.88-2.73(m,2H),2.69(s,3H),2.51-2.37(m,2H),2.02-1.92(m,1H),1.70-1.59(m,1H).LRMS-ESI(m/z):213.07[M-NHMe]+.
实施例27 2-甲基-N-(4-苯基-4,5,6,7-四氢苯并噻唑-4-基)丙烷-2-磺胺(化合物A31)的制备
步骤一:
化合物A31-1(20g,1eq)加入冰醋酸(200mL),氮气保护,缓慢滴加液溴(29g,1eq)的醋酸溶液(20mL),室温反应2h。加入硫脲(27.4g,2eq),加热回流3小时,浓缩干溶剂,加入乙醇打浆,过滤得化合物A31-2(15g,50%)。
步骤二:
化合物A31-2(20.6g,1eq)加入THF,氮气保护,冷却到0-5℃,滴加亚硝酸异戊酯(17.3g,1.2eq),滴毕,升温到45℃反应2h,氯化铵淬灭,DCM萃取,饱和食盐水洗,干燥,浓缩,柱层析得化合物A31-3,深棕色固体5.7g。
步骤三:
反应瓶中加入化合物A31-3(300mg,1eq)、叔丁基亚磺酰胺(330mg,1.4eq)、钛酸四乙脂(489mg,1.1eq)、THF,氮气保护,40℃反应过夜,冷却到室温,反应液倒入冰盐水中,加入EA搅拌过滤,分液,EA萃取,饱和食盐水洗,干燥浓缩,柱层析得化合物A31-4约288mg。采用同样的方法制备克级化合物A31-4。
步骤四:
碘苯(35.4g,2.5eq)加入DCM(356mL)中,氮气保护,降至-60℃,滴加丁基锂(58.3mL,2.5M,2.1eq),保温反应3h,计作反应液2,待用。化合物A31-4(17.8g,1eq)加入DCM(338mL)中溶清,计作反应液1,待用。反应液1控温-60℃左右滴加入反应液2中,保温反应2h。氯化铵淬灭反应,EA萃取,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析得到5.5g化合物A31。
实施例28 4-苯基-4,5,6,7-四氢苯并噻唑-4-胺(化合物A32)的制备
化合物A31(5.5g,1eq)加入二氧六环(165mL)中,滴加盐酸乙酸乙酯溶液(12.4mL,4M,3eq),室温反应0.5h。加入饱和碳酸氢钠水溶液中和,EA萃取,饱和食盐水洗,干燥浓缩,柱层析得到化合物A32,2.81g。
1H NMR(400MHz,DMSO-d6)δ8.85(s,1H),7.68-6.74(m,5H),2.85(m,2H),2.27(s,2H),1.92(m,2H),1.85(m,1H),1.56(m,1H).LRMS-ESI(m/z):214.09[M-NH2]+.
实施例29 N-(4-苯基-4,5,6,7-四氢苯并噻唑-4-基)甲酰胺(化合物A33)的制备
无水甲酸(1.8g,9eq)加入乙酸酐(3.99g,9eq)中,氮气保护升温至60℃反应2h后降至室温(25℃左右),加入化合物A32(1.0g,1eq)的DCM(20mL)溶液,室温下搅拌1.5h。加入饱和碳酸氢钠水溶液淬灭反应,DCM萃取,有机相浓缩,柱层析,得到化合物A33,952mg,淡黄色固体。
实施例30 N-甲基-4-苯基-4,5,6,7-四氢苯并噻唑-4-胺(化合物A34)及其盐酸盐的制备
化合物A33(950mg,1eq)加入THF(40mL)溶液中,氮气保护下滴加硼烷四氢呋喃(36.8mL,10eq,1.0M)升温至回流反应2h,TLC显示反应完毕,降至0-5℃,冰浴下加入甲醇淬灭反应,浓缩至干,加入甲醇和水1∶1(17mL)室温搅拌,TLC检测反应完毕,DCM萃取,有机相浓缩,柱层析,得到364mg粗品,加入10mLDCM,滴氯化氢乙酸乙酯溶液(0.37mL,1.0eq,4.0M),减压浓缩至干,加入异丙醚打浆,过滤,得到330mg化合物A34。
1H NMR(400MHz,DMSO-d6)δ9.94(m,2H),9.21(s,1H),7.46-7.36(m,3H),7.29(m, 2H),2.90(m,2H),2.47(m,1H),2.43(t,J=5.2Hz,3H),2.36(td,J=13.1,2.4Hz,1H),2.00(m,1H),1.43(m,1H).LRMS-ESI(m/z):214.08[M-NHCH3]+.
实施例31 4-苯基-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A35)的制备
步骤一:
化合物A35-1(800mg)加入二氯甲烷(20mL),加入DMAP(464mg)、二碳酸二叔丁酯(1.57g),室温反应过夜。浓缩,柱层析得化合物A35-2,类白色固体250mg。
步骤二:
100mL三口瓶中加入2.7mL(26.087mmol,7eq)溴苯、6mL无水四氢呋喃,氮气保护下置于-78℃的低温反应釜中搅拌。待内温降到-60℃以下,开始滴加10.4mL(26.087mmol,7eq)2.5M的正丁基锂,控制内温不超过-60℃,滴毕,1h后,缓慢加入1g(3.726mmol,1eq)化合物A35-2的THF溶液(10mL),控制内温不超过-60℃,滴毕,-78℃下搅拌5h。反应体系中加入饱和氯化铵水溶液,加入EA萃取,EA相水洗,饱和食盐水洗,无水硫酸钠干燥,浓缩,柱层析得化合物A35-3,1.13g淡黄色固体。
LRMS-ESI(m/z):347.18[M+H]+.
步骤三:
取1.13g(3.261mmol,1eq)化合物A35-3于50mL三口瓶中,加入8mL DCM,氮气保护下置于冰浴中搅拌10min后加入2.1g(32.61mmol,10eq)NaN3,加毕,缓慢滴加4mL CF3COOH,室温搅拌过夜。体系倾入适量冰水中,缓慢滴加氨水调节pH值10左右,适量DCM萃取,DCM相水洗,饱和食盐水洗,无水硫酸钠干燥,浓缩得化合物A35-4,油状物。
LRMS-ESI(m/z):272.06[M+H]+.
步骤四:
上步所得油状物A35-4中加入2mL THF,122mg(0.2eq)六水合氯化钴,氮气保护下 搅拌。缓慢滴加390mg(4eq)NaBH4的水(10mL)溶液,室温搅拌2h。用2M盐酸调节pH值为3左右,过滤,滤去不溶物,再用饱和碳酸钠溶液调节pH值为10左右,DCM/MeOH=20∶1萃取,无水硫酸钠干燥,浓缩柱层析得化合物A35,白色固体320mg。
1H NMR(400MHz,DMSO-d6)δ7.30-7.22(m,4H),7.16-7.12(m,1H),6.67(s,2H),2.57-2.54(m,2H),2.22(s,2H),1.84-1.81(m,2H),1.78-1.72(m,1H),1.54-1.43(m,1H).LRMS-ESI(m/z):274.17[M-NH2]+.HPLC纯度:>97%,保留时间:10.848min。
实施例32-35 N-(2-氨基-4-苯基-4,5,6,7-四氢苯并噻唑-4-基)甲酰胺(化合物A36)、N,N’-(4-苯基-4,5,6,7-四氢苯并噻唑-2,4-二基)二甲酰胺(化合物A37)、N4-甲基-4-苯基-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A38)、N4-甲基-4-苯基-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A39)的制备
步骤一:
取240mg化合物A35于25mL单口瓶中,加入8mL甲酸乙酯,氮气保护下回流过夜。将反应体系浓缩干,加入适量DCM∶MeOH=20∶1,有机相用饱和食盐水洗一次,硫酸钠干燥,浓缩得黄褐色固体200mg。取少量柱层析分离得化合物A36和得化合物A37。
A36:LRMS-ESI(m/z):274.17[M+H]+.
A37:LRMS-ESI(m/z):320.20[M+H]+.
步骤二:
A36和A37的混合物加入5mL硼烷四氢呋喃溶液,氮气保护下回流过夜。将体系冷却至室温,缓慢滴加甲醇淬灭多余的硼烷,浓缩,加入6mL甲醇和2滴浓盐酸,氮气保护下回流过夜。体系冷却至室温,加入少量氨水调节pH值8-9,浓缩,柱层析得50mg化合物A39,HCl/MeOH成盐得56mg淡黄色粉末。质谱出碎片峰,LRMS-ESI(m/z):243.32[M-NHCH3]+.
1H NMR(400MHz,DMSO-d6)δ9.03(br,1H),7.57(m,1H),7.39(m,4H),7.35(m,1H),2.80(d,J=4.7Hz,3H),2.60(m,2H),2.43(s,3H),2.22(m,2H),1.85(m,1H),1.33(m,1H).
另柱层析得18mg化合物A38。
1H NMR(400MHz,DMSO-d6)δ7.25(d,J=4.0Hz,4H),7.19-7.14(m,1H),6.69(s,2H),2.56-2.54(m,2H),2.17(s,3H),2.05-1.98(m,1H),1.78-1.66(m,2H),1.49-1.39(m,1H).LRMS-ESI(m/z):260.21[M+H]+.
实施例36 N-(2-氨基-5-苯基-4,5,6,7-四氢苯并噻唑-5-基)甲酰胺(化合物A40)的制备
步骤一:
化合物A40-1(5.0g,43.1mmol)溶于DMF中,加入咪唑(6.5g,2eq),0-10℃下加入TBDPSCl(12g,1.1eq),室温搅拌过夜,加入水淬灭,EA/水萃取,有机相干燥浓缩柱层析得化合物A40-2,6.0g淡黄色油状物。
步骤二:
化合物A40-2(6.0g,16.9mmol)溶于DCM中,戴斯马丁试剂(24g,1.5eq),室温搅拌过夜。将反应液倒入水中,EA萃取,干燥浓缩、柱层析得化合物A40-3,5.0g。
步骤三:
化合物A40-3(5.0g,14.2mmol)溶于无水THF中,加入叔丁基亚磺酰胺(2.6g,1.5eq),0-10℃滴入钛酸乙酯(11mL,3eq),室温过夜。将反应液倒入水中,过滤,滤液用EA萃取,干燥浓缩柱层析得化合物A40-4,6.6g。
步骤四:
格氏试剂制备:Mg(3.4g,10eq)悬浮于无水THF中,氮气置换3次,缓慢滴入溴苯(29g,10eq),50℃加热1h,降至-10-0℃待用。
化合物A40-4(6.6g,14.2mol)溶于无水THF中,缓慢滴入上述格氏试剂,0-10℃搅拌 2h,室温搅拌1h。加入2M盐酸,室温搅拌过夜。将反应液倒入水中,调节pH=8-9,二氯甲烷萃取,干燥浓缩柱层析得化合物A40-5,3.5g。
步骤五:
化合物A40-5(3.5g)溶于甲酸乙酯中,加热回流24h。浓缩干溶剂,柱层析得化合物A40-6,3.5g。
步骤六:
化合物A40-6(3.5g)溶于THF中,加入TABF(1M)(16mL,2eq),室温搅拌过夜。浓缩干溶剂,柱层析得化合物A40-7,110mg。
步骤七:
化合物A40-7(1.1g,5.1mmol)溶于DCM中,加入戴斯马丁试剂(3.8g,2eq),室温搅拌过夜。加入硫代硫酸钠淬灭反应,二氯甲烷萃取,干燥过滤浓缩,柱层析得化合物A40-8,700mg。
步骤八:
化合物A40-8(350mg,1.6mmol)溶于DCM中,0-10℃,加入三溴吡啶嗡(700mg,1.1eq),室温搅拌3h。加入硫代硫酸钠淬灭反应,二氯甲烷萃取,干燥过滤浓缩,柱层析得溴代中间体,380mg淡黄色固体。将上述淡黄色固体溶于二氧六环中,加入硫脲(350mg,3eq)和二异丙基乙胺(400mg,2eq),80-90℃搅拌5h。加入DCM/水萃取,有机相干燥过滤浓缩柱层析得标题化合物200mg。
1H NMR(400MHz,DMSO-d6)δ8.31-8.19(m,1H),8.02-7.97(m,1H),7.42-7.19(m,5H),6.72-6.70(m,2H),2.97-2.90(m,2H),2.67-2.62(m,1H),2.52-2.47(m,1H),2.41-2.32(m,1H),2.28-2.14(m,1H).
实施例37 N-(2-氨基-7-苯基-4,5,6,7-四氢苯并噻唑-7-基)甲酰胺(化合物A41)的制备
化合物A40(200mg,0.73mmol)悬浮于THF中,加入硼烷四氢呋喃溶液(10eq),加热回流3h。加入甲醇淬灭反应,浓缩干溶剂,加入甲醇/水,回流过夜,氯仿萃取,干燥、浓缩、柱层析得化合物A41 100mg。
1H NMR(400MHz,DMSO-d6)δ7.40(m,2H),7.35(t,J=7.5Hz,2H),7.26(t,J=6.9 Hz,1H),6.69(s,2H),3.09(d,J=16.7Hz,1H),2.69(d,J=16.2Hz,1H),2.16(m,2H),2.00(m,5H).LRMS-ESI(m/z):260.19[M+H]+.
实施例38 N-(2-氨基-7-苯基-4,5,6,7-四氢苯并噻唑-7-基)甲酰胺(化合物A42)的制备
取化合物A41(350mg,1.6mmol)溶于DCM中,加热回流,加入三溴吡啶嗡(700mg,1.1eq),搅拌1h。加入硫代硫酸钠淬灭反应,二氯甲烷萃取,干燥过滤浓缩,柱层析得溴代中间体,380mg淡黄色固体。将上述淡黄色固体溶于二氧六环中,加入硫脲(350mg,3eq)和二异丙基乙胺(400mg,2eq),80-90℃加热5h。加入DCM/水萃取,有机相干燥过滤浓缩柱层析得化合物A42,150mg。
实施例39 N7-甲基-7-苯基-4,5,6,7-四氢苯并噻唑-2,7-二胺(化合物A43)的制备
化合物A42(150mg,0.56mmol)悬浮于THF中,加入硼烷四氢呋喃溶液10eq,加热回流3h。加入甲醇淬灭反应,浓缩干溶剂,加入甲醇/水等体积的混合液,回流过夜,氯仿萃取,干燥、过滤、柱层析得化合物A43,80mg。
1H NMR(400MHz,DMSO-d6)δ7.36(m,2H),7.29(t,J=7.5Hz,2H),7.19(t,J=7.1Hz,1H),6.63(s,2H),2.92(d,J=16.3Hz,1H),2.55(d,J=16.6Hz,1H),2.03(m,3H),1.92(s,3H),1.88(m,1H).LRMS-ESI(m/z):260.18[M+H]+.
实施例40 5-吗啉-5-苯基-4,5,6,7-四氢苯并噻吩-2-羧酸乙酯(化合物A44)及其盐酸盐的制备
步骤一:
将5g化合物A23-1与1.1eq吗啉,1.2eq三氮唑混合于40mL甲苯中,回流分水过夜,制得含有化合物A44-1的溶液。
步骤二:
氮气保护下,将4eq苯基溴化镁四氢呋喃溶液置于冰浴中搅拌10min,再将前述化合物A44-1的溶液滴加到反应体系中,加毕于室温下搅拌2h,将反应液倒入氯化铵水溶液中,加入EA萃取,有机相再用水洗涤两次,直接浓缩得含有化合物A44-2的粗品。
步骤三:
化合物A44-2溶于20mL四氢呋喃中,加入20mL 1M的盐酸,40℃反应过夜。调pH至弱碱性,EA萃取,有机相水洗,饱和食盐水洗,干燥浓缩柱层析得化合物A44-3,油状物4.4g,三步收率53%。于PE中打浆,过滤得淡黄色固体3.4g。
步骤四:
冰浴及氮气保护下,将2eq三氯氧磷滴加到含1.6eq DMF的二氯甲烷溶液中,加毕于室温下搅拌20min,再次冰浴降温,将200mg化合物A44-3(1eq)分批多次加入到反应体系中,加毕于室温下反应过夜。加碳酸氢钠水溶液,分层,有机相饱和食盐水洗,浓缩、柱层析得到115mg化合物A44-4。
步骤五:
将2eq金属钠加入到冰浴下的无水乙醇中,氮气保护,待钠块完全消失于溶液中后,加入1.1eq巯基乙酸乙酯,再滴加入化合物A44-4(110mg,1eq)的乙醇溶液,加毕室温下反应过夜。加氯化铵淬灭反应,DCM萃取,有机相干燥、浓缩柱层析得无色油状物105mg。于甲醇中加入氯化氢/甲醇溶液成盐酸盐,于EA中打浆,过滤得到化合物A44盐酸盐,白色固体85mg。
1H NMR(400MHz,DMSO-d6)δ11.65(s,1H),7.69(m,2H),7.57(s,1H),7.52-7.42(m, 3H),4.23(q,J=7.1Hz,2H),4.20-3.82(m,5H),3.60-3.50(m,3H),3.24(d,J=12.5Hz,1H),3.12-3.03(m,1H),2.70-2.55(m,3H),2.35(m,1H),1.25(t,J=7.1Hz,3H).LRMS-ESI(m/z):372.43[M+H]+.
实施例41 3-氨基-5-吗啉-5-苯基-4,5,6,7-四氢苯并噻吩-2-羧酸乙酯(化合物A45)及其盐酸盐的制备
将200mg化合物A44-3与1.5eq氰基乙酸乙酯,1.5eq吗啉,1.5eq单质硫混合于乙醇中,50℃搅拌过夜。反应液冷却后有固体析出,过滤,滤饼柱层析纯化后得到80mg A45固体,于甲醇中加入氯化氢/甲醇溶液成盐酸盐,于乙腈中打浆,过滤得到标题化合物A45盐酸盐,淡黄色固体65mg。
1H NMR(400MHz,DMSO-d6)δ11.30(s,1H),7.67(s,2H),7.48(m,3H),7.25(m,2H),4.16-3.82(m,7H),3.65-3.40(m,3H),3.09(d,J=12.9Hz,1H),2.99-2.89(m,1H),2.55(m,2H),2.39(m,1H),2.11(m,1H),1.17(t,J=7.1Hz,3H).LRMS-ESI(m/z):387.39[M+H]+.
实施例42 5-吗啉-5-苯基-4,5,6,7-四氢苯并噻吩-2-羧酸(化合物A46)的制备
反应瓶中加入化合物A44590mg,加入乙醇和1.27mL 5N氢氧化钠水溶液(4eq),60℃反应8h,加水稀释,加入MTBE,分出水相,调pH至中性,DCM萃取(5mL×5),合并有机相,少量饱和食盐水洗,干燥浓缩得粗品455mg,加入MTBE打浆,过滤,滤饼烘干得淡粉红色固体320mg,化合物A46。
1H NMR(400MHz,DMSO-d6)δ12.78(s,1H),7.53(s,1H),7.35-7.17(m,5H),3.50(t,J=4.5Hz,4H),3.23(d,J=16.5Hz,1H),3.00(d,J=16.4Hz,1H),2.80(dt,J=17.0,4.4Hz,1H),2.45(m,2H),2.27(m,3H),2.19-2.01(m,2H).LRMS-ESI(m/z):344.47[M+H]+.
实施例43 4-(5-苯基-4,5,6,7-四氢苯并噻吩-5-基)吗啉(化合物A47)及其盐酸盐的 制备
将235mg化合物A46与0.5eq氧化亚铜,0.4eq DBU混合于少量DMF中,氮气置换多次,160℃反应4h,冷却后加EA稀释,氯化铵水溶液洗,饱和食盐水洗,干燥浓缩柱层析得白色固体100mg的A47,于甲醇中加入氯化氢/甲醇溶液成盐酸盐,于乙腈中打浆,过滤得到白色固体100mg,为化合物A47盐酸盐。
1H NMR(400MHz,DMSO-d6)δ11.67(s,1H),7.77-7.60(m,2H),7.45(m,3H),7.28(d,J=5.1Hz,1H),6.87(d,J=5.1Hz,1H),4.17-3.81(m,5H),3.63(d,J=12.4Hz,1H),3.49(d,J=14.4Hz,2H),3.21(q,J=4.5Hz,1H),2.98(dd,J=17.2,5.4Hz,1H),2.60(m,3H),2.29(m,1H).LRMS-ESI(m/z):300.45[M+H]+.
实施例44 6-苯基-6-(哌啶-1-基)-4,5,6,7-四氢苯并噻唑-2-胺(化合物A48)及其盐酸盐的制备
步骤一:
格氏试剂制备:镁(345.6mg,4.8eq)悬浮于干燥的四氢呋喃中,滴入溴苯(200mg,0.48eq)升温至50-60℃,引发,再滴入溴苯(1.8g,3.6eq)保持微沸,30min滴加完毕,升温至60℃搅拌1h,大部分镁消失,降至0-10℃待用。
另取反应瓶中加入三氮唑(1.0g,1.2eq),哌啶(1.4mL,1.2eq),1,4-环己二酮单乙二醇缩酮A23-1(2.0g,12.8mmol)溶于甲苯中,回流分水18h,得到A48-1的甲苯溶液。降至室温,滴入上述制备的格氏试剂中(0-10℃),滴加完毕后,室温搅拌12h,柱层析得化合物A48-2,1.0g。
步骤二:
将化合物A48-2(900mg)溶于乙醇中,加入盐酸,加热至70℃搅拌12h。将反应液浓 缩至干,甲叔醚/水萃取,水相用氢氧化钠溶液调节pH=11-12,二氯甲烷萃取,干燥过滤浓缩得化合物A48-3,500mg。
步骤三:
化合物A48-3(500mg,2mmol)溶于乙酸中,加入三溴吡啶(800mg,1.2eq),加热回流3h,加入硫脲(300mg,2eq)加热回流18h,浓缩干溶剂,加入饱和碳酸钠溶液,调节pH=9-10,二氯甲烷萃取,有机相干燥浓缩柱层析得A48,白色固体200mg。加入甲醇(5mL)溶清,滴加HCl/EA溶液搅拌反应,浓缩干溶剂,得化合物A48盐酸盐,白色固体。
LRMS-ESI(m/z):314.28[M+H]+.1H NMR(400MHz,DMSO-d6)δ10.75(s,1H),7.69(br,2H),7.45(m,3H),7.15(br,2H),3.99(d,J=15.1Hz,1H),3.68(d,J=10.9Hz,1H),3.61(d,J=11.2Hz,1H),3.47(d,J=15.0Hz,1H),3.11(m,1H),2.55(m,2H),2.32(m,2H),2.10(m,1H),1.71(d,J=13.2Hz,2H),1.60(d,J=11.7Hz,1H),1.18(m,1H).
实施例45 6-(3-氯苯基)-6-(哌啶-1-基)-4,5,6,7-四氢苯并噻唑-2-胺(化合物A49)的制备
步骤一:
间氯苯碘(12g,2eq)溶于无水THF中,-78℃缓慢滴入正丁基锂(2.5M)(23mL,1.1eq),滴加完毕,-78℃保温1h,滴入新制备的A48-1甲苯溶液,滴加完毕后,保温-78℃搅拌1h,室温搅拌12h,柱层析得化合物A49-1,3.0g。
步骤二:
化合物A49-1(3.0g,1.35mmol)溶于乙醇中,加入浓盐酸(3eq,12M),室温搅拌过夜,浓缩干溶剂,饱和碳酸氢钠调节pH=11-12,DCM萃取,干燥过滤浓缩,柱层析得化合物A49-2,1.8g。
步骤三:
化合物A49-2(200mg,0.7mmol)溶于乙酸中,加入三溴吡啶嗡(1.1g,4eq),加热回流4h。继续加入硫脲(420mg,8mmol),加热回流18h,浓缩干溶剂,加入饱和碳酸 钠溶液,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得标题化合物A4925mg。
LRMS-ESI(m/z):348.38[M+H]+.
实施例46 6-苯基-6-(吡咯烷-1-基)-4,5,6,7-四氢苯并噻唑-2-胺(化合物A50)的制备
步骤一:
三氮唑(12.4g,1.1eq)、四氢吡咯(13.35g,1.2eq)、1,4-环己二酮单乙二醇缩酮(25.0g,160mmol)溶于甲苯中,回流分水18h,降至室温,得到化合物A50-1的甲苯溶液待用。
步骤二:
溴苯(75g,3eq)溶于无水THF中,-78℃缓慢滴入正丁基锂(2.5M)(150mL,3.3eq),滴加完毕,-78℃保温1h,滴入上述制备的A50-1甲苯溶液,滴加完毕后,保温-78℃搅拌1h,室温搅拌12h,柱层析得化合物A50-2,10.5g。
步骤三:
化合物A50-2(10.5g,6.5mmol)溶于乙醇中,加入浓盐酸(12M,1.6mL),室温搅拌过夜,浓缩干溶剂,饱和碳酸氢钠调节pH=11-12,DCM萃取,干燥过滤浓缩,柱层析得化合物A50-3,8.5g。
步骤四:
化合物A50-3(200mg,0.7mmol)溶于乙酸中,加入三溴吡啶嗡(1.1g,4eq),加热回流4h。继续加入硫脲(420mg,8mmol),加热回流18h,浓缩干溶剂,加入饱和碳酸钠溶液,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得标题化合物200mg。
LRMS-ESI(m/z):300.33[M+H]+.1H NMR(400MHz,DMSO-d6)δ7.37(m,2H),7.28(t,J=7.4Hz,2H),7.20(t,J=6.9Hz,1H),6.59(s,2H),3.15(d,J=14.6Hz,1H),2.89(d,J=15.7Hz,1H),2.55(br,2H),2.39(m,3H),2.16(m,2H),1.73(m,1H),1.56(br,4H).
手性柱分离得到异构体A50-P1(S构型)和A50-P2(R构型):
A50-P1(S构型):HPLC纯度:>99%,保留时间:5.761min。手性纯度:>95%,保留时间:7.617min。
A50-P2(R构型):HPLC纯度:>99%,保留时间:7.452min。手性纯度:>95%,保留时间:7.623min。
实施例47 6-(3-氯苯基)-6-(吡咯烷-1-基)-4,5,6,7-四氢苯并噻唑-2-胺(化合物A51)的制备
步骤一:
间氯碘苯(45g,1.2eq)溶于无水THF中,-78℃缓慢滴入正丁基锂(2.5M)(88mL,1.5eq),滴加完毕,-78℃保温1h,滴入新制备的A50-1甲苯溶液,滴加完毕后,保温-78℃搅拌1h,室温搅拌12h,柱层析得化合物A51-1,8.5g。
步骤二:
化合物A51-1(8.5g,6.5mmol)溶于乙醇中,加入浓盐酸(12M,1.6mL),室温搅拌过夜,浓缩干溶剂,饱和碳酸氢钠溶液调节pH=11-12,DCM萃取,干燥过滤浓缩,柱层析得化合物A51-2,5.2g。
步骤三:
化合物A51-2(300mg,9.4mmol)溶于乙酸中,加入三溴吡啶嗡(1.02g,4eq),加热回流4h,生成溴代中间体。继续加入二甲基硫脲(560mg,8eq),加热回流18h,浓缩干溶剂,加入饱和碳酸钠溶液,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得90mg化合物A51,白色固体。
LRMS-ESI(m/z):334.39[M+H]+.1H NMR(400MHz,CDCl3)δ7.38(s,1H),7.29(m,3H),6.62(s,2H),3.13(d,J=16.2Hz,1H),2.88(d,J=16.0Hz,1H),2.55(br,2H),2.37(m,3H),2.15(m,2H),1.70(m,1H),1.58(br,4H).
实施例48 5-苯基-5-(吡咯烷-1-基)-4,5,6,7-四氢苯并噻吩-2-羧酸(化合物A52)的制备
步骤一:0-10℃下,将三氯氧磷(126mg,2eq)滴加到DMF(50mg,1.5eq)二氯甲烷溶液中,冰浴下搅拌2h,化合物A50-3(100mg,0.5mmol)溶于二氯甲烷中,加入上述反应液中,室温搅拌过夜。缓慢滴入乙酸钠水溶液中,分液,有机相干燥过滤、浓缩得到化合物A52-1,油状物。
步骤二:将上述油状物A52-1溶于乙醇中,加入巯基乙酸乙酯(74mg,1.5eq),搅拌10min,冰浴下加入乙醇钠(55mg,4eq),室温下反应过夜,加入水,加热回流2h。浓缩乙醇,1M盐酸溶液调节pH=5-6,二氯甲烷萃取,有机相干燥,浓缩,柱层析得化合物A5255mg,淡黄色油状物。
实施例49 1-(5-苯基-4,5,6,7-四氢苯并噻吩-5-基)吡咯烷(化合物A53)的制备
化合物A52(55mg,0.17mmol)溶于NMP中,加入氧化亚铜(24mg,1eq),喹啉(26mg,1.1eq),升温至140℃,12h,原料基本完全反应,将反应液倒入水中,二氯甲烷萃取,干燥过滤浓缩得化合物A53,20mg。
实施例50 6-苯基-6-(吡咯烷-1-基)-4,5,6,7-四氢苯并噻二唑(化合物A54)的制备
化合物A50-3(200mg,0.9mmol)溶于甲醇中,加入氨基脲(200mg,2eq),乙酸钾(200mg,2.1eq)加热回流6h。浓缩干溶剂,二氯甲烷/水萃取,干燥过滤浓缩,得固体300mg。将上述制备的固体溶于二氯甲烷中,-20℃缓慢滴入二氯亚砜中,室温搅拌1h。将反应液倒入水中,氢氧化钠调节pH=12,二氯甲烷萃取,干燥,过滤浓缩,柱层析得 化合物A54100mg。
1H NMR(400MHz,CDCl3)δ7.29(m,5H)3.65(d,J=17.3Hz,1H),3.41(d,J=17.1Hz,1H),3.28(dt,J=16.4,4.4Hz,1H),2.64(m,3H),2.55(m,2H),2.43(m,2H),1.68(m,4H).LRMS-ESI(m/z):286.35[M+H]+.
实施例51 5-苯基-5-(吡咯烷-1-基)-4,5,6,7-四氢苯并[d]异恶唑(化合物A55)的制备
步骤一:
化合物A50-3(300mg,0.7mmol)溶于甲苯中,加入无水硫酸镁干燥2天,过滤待用;甲苯中加入叔丁醇钠(246mg,2eq),加入甲酸乙酯(138mg,1.5eq),0-10℃下加入A50-3的甲苯溶液,室温搅拌过夜。将反应液用甲苯萃取2次,保留水相并用1M盐酸溶液调节pH=6-7,二氯甲烷萃取3次,干燥,过滤浓缩柱层析得化合物A55-1,800mg。
步骤二:
化合物A55-1(110mg)溶于乙酸中,加入盐酸羟胺(70mg,2eq),升温至80℃加热5h。浓缩干溶剂,饱和碳酸钠调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩,柱层析得化合物A5566mg。
ESI-MS m/z 269.36[M+H]+.
实施例52 N,N-二甲基-6-苯基-6-(吡咯烷-1-基)-4,5,6,7-四氢苯并噻唑-2-胺(化合物A56)的制备
化合物A50-3(50mg,0.16mmol)溶于乙酸中,加入三溴吡啶嗡(300g,4eq),加热回流4h,得到溴代中间体。继续加入二甲基硫脲(100mg,8mmol),加热回流18h。浓缩干溶剂,加入饱和碳酸钠水溶液,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得化合物A5610mg。
LRMS-ESI(m/z):328.45[M+H]+.
1H NMR(400MHz,DMSO-d6)δ7.36(m,2H)7.31(t,J=7.6Hz,2H),7.21(t,J=7.1Hz,1H),2.97(d,J=16.8Hz,1H),2.94(s,3H),2.83(s,3H),2.78(d,J=16.8Hz,1H),2.50(m,2H),2.46-2.33(m,3H),2.26(m,1H),2.15(m,1H),1.68(m,1H),1.55(m,4H).
实施例53 6-(3-氯苯基)-N,N-二甲基-6-(吡咯烷-1-基)-4,5,6,7-四氢苯并噻唑-2-胺(化合物A57)及其盐酸盐的制备
化合物A51-2(300mg,9.4mmol)溶于乙酸中,加入三溴吡啶嗡(1.1g,4eq),加热回流4h,生成溴代中间体。继续加入二甲基硫脲(600mg,8eq),加热回流18h,浓缩干溶剂,加入饱和碳酸钠水溶液,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得A57,为150mg油状物,将上述油状物溶于甲基叔丁基醚中,加入12M的浓盐酸调节pH=2-3,大量固体析出,过滤烘干得化合物A57盐酸盐,95mg。
1H NMR(400MHz,DMSO-d6)δ12.28(s,1H),10.32(s,1H),7.93(s,1H),7.77(d,J=7.6Hz,1H),7.53(m,2H),4.15(d,J=16.3Hz,1H),3.57(d,J=17.9Hz,1H),3.40(s,3H),3.35(m,1H),3.17(m,1H),2.96(s,3H),2.88(m,4H),2.63(td,J=13.1,5.9Hz,1H),2.11(m,1H),1.84(m,2H),1.67(m,2H).LRMS-ESI(m/z):362.41[M+H]+.
实施例54 6-(3-氯苯基)-6-(哌啶-1-基)-4,5,6,7-四氢苯并噻二唑(化合物A58)及其马来酸盐的制备
化合物A49-2(300mg,1.3mmol)溶于甲醇中,加入氨基脲(300mg,2eq)、乙酸钾(300mg,2.1eq),加热回流6h。浓缩干溶剂,二氯甲烷/水萃取,有机相干燥过滤浓缩,得300mg固体。将上述制备的固体溶于二氯甲烷中,-20℃缓慢滴入二氯亚砜中,室温搅 拌1h。将反应液倒入水中,氢氧化钠溶液调节pH=12,二氯甲烷萃取,干燥,过滤柱层析得碱式化合物A58,100mg固体。将上述固体溶于丙酮中,加入马来酸搅拌,大量固体析出,过滤,烘干得化合物A58马来酸盐100mg,白色固体。
1H NMR(400MHz,DMSO-d6)δ7.58(br,1H),7.41(m,3H),6.16(s,2H),3.54(d,J=17.3Hz,1H),3.30(d,J=14.6Hz,1H),2.99-2.37(m,8H),1.58(br,4H),1.39(br,2H).ESI-MS m/z 334.38[M+H]+.
实施例55 6-(3-氯苯基)-N6-甲基-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A59)的制备
步骤一:
三氮唑(1.0g,1.2eq)、N-甲基苄胺(1.8mL,1.2eq)、1,4-环己二酮单乙二醇缩酮(2.0g,1.28mmol)溶于甲苯中,回流分水18h,将至室温,得到含A59-1的溶液待用。
步骤二:
间氯苯碘(6g,2eq)溶于无水THF中,-78℃缓慢滴入正丁基锂(2.5M)(6mL,1.1eq),滴加完毕,-78℃保温1h,滴入上述制备的A59-1,滴加完毕后,保温搅拌1h,室温搅拌12h,柱层析得化合物A59-2,600mg。
步骤三:
化合物A59-2(500mg,1.35mmol)溶于甲苯中,加入DIAD(275mg,1.1eq),加热回流18h,浓缩干甲苯,加入饱和氯化铵水溶液和乙醇,回流18h,浓缩除去乙醇,氨水调节pH至7-8,EA萃取,干燥浓缩,柱层析得化合物A59-3,120mg。
步骤四:
化合物A59-3(50mg)溶于乙醇中,加入浓盐酸(12M,3eq),室温搅拌12h。浓缩干溶剂,加入饱和碳酸钠,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得化合物A59-4,20mg。
步骤五:
化合物A59-4(20mg,0.09mmol)溶于乙酸中,加入三溴吡啶嗡(135mg,0.36mmol), 加热回流4h,生成溴代中间体。继续加入硫脲(54mg,0.72mmol),加热回流18h,浓缩干溶剂,加入饱和碳酸钠水溶液,调节pH=9-10,二氯甲烷萃取,干燥,过滤浓缩柱层析得化合物A595mg。
1H NMR(400MHz,DMSO-d6)δ7.64(s,1H),7.49(d,J=7.7Hz,1H),7.36(t,J=7.8Hz,1H),7.30(d,J=7.9Hz,1H),6.72(s,2H),3.10(m,3H),3.00(d,J=15.8Hz,1H),2.67(d,J=16.0Hz,1H),2.50(m,2H),2.32(d,J=10.7Hz,1H),1.90(dd,J=10.6,3.0Hz,1H).ESI-MS m/z 294.33[M+H]+.
实施例56 4-(2-氯苯基)-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A60)及其马来酸盐的制备
步骤一:
邻氯溴苯(24.83g,130mmol)溶于干燥THF(100mL)中,冷至-78℃,滴加n-BuLi(48mL,2.5M),约1h加完,1h后滴加A35-2(5.340g,20mmol)的THF(80mL)溶液,控温-78℃搅拌5h。饱和氯化铵水溶液淬灭反应,EA萃取,有机相用饱和氯化钠水溶液洗涤,干燥,浓缩,柱层析得化合物A60-1,黄色泡沫状固体(2.8g)。
步骤二:
化合物A60-1(2.3g,6.0mmol)溶于DCM(100mL)中,加叠氮钠(7.8g,120mmol),冰浴下滴加TFA(80mL),1h内加完,加毕后,冰浴下继续反应1h,室温过夜,加冰水100mL,用5M氨水调节pH至9,DCM萃取,有机相用饱和氯化钠洗涤,无水硫酸钠干燥,柱层析得化合物A60-2,白色泡沫状固体(1.050g)。
步骤三:
化合物A60-2(1.050g)溶于THF(20mL)中,加六水合氯化钴(167mg),滴加硼氢化钠(260mg)水溶液(20mL),室温反应30min,加1M盐酸淬灭反应,再调节pH=12,DCM萃取,合并有机相,无水硫酸钠干燥,浓缩,柱层析得碱式化合物A60(浅褐色固 体),参照实施例54的方法成马来酸盐得化合物A60马来酸盐(682mg)。
1H NMR(400MHz,DMSO-d6)δ8.89(br,2H),7.44(m,2H),7.38(m,1H),7.30(d,J=7.5Hz,1H),6.92(s,2H),6.02(s,2H),2.71(m,1H),2.61(m,1H),2.43(m,1H),2.04(m,2H),1.80(m,1H).ESI-MS m/z 308.11[M+H]+.
实施例57 N-(2-氨基-4-(2-氯苯基)-4,5,6,7-四氢苯并噻唑-4-基)甲酰胺(化合物A61)的制备
取化合物A60(260mg)溶于甲酸乙酯(12mL),90℃加热18h。将反应液浓缩干,柱层析得化合物A61。
实施例58 4-(2-氯苯基)-N4-甲基-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A62)及其马来酸盐的制备
取化合物A61溶于干燥的THF(20mL)中,滴加硼烷二甲硫醚溶液(3.8mL,2M),室温搅拌20min后,回流5h。冷至室温,小心加入甲醇(15mL)淬灭反应,浓缩溶剂,加甲醇(30mL)回流过夜,浓缩,柱层析得碱式化合物A62(浅褐色固体),参照实施例54的方法成马来酸盐得化合物A62马来酸盐。
1H NMR(400MHz,MeOD)δ7.58(d,J=7.2Hz,1H),7.46(t,J=7.0Hz,1H),7.36(t,J=7.7Hz,1H),6.92(d,J=7.8Hz,1H),6.31(s,3H),2.94(m,1H),2.85(s,3H),2.71(m,2H),2.20(t,J=11.9Hz,1H),1.97(m,1H),1.46(m,1H).ESI-MS m/z 294.05[M+H]+.
实施例59 4-(2-氯苯基)-N4-乙基-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A63)的制备
取化合物A60(195mg,0.7mmol)溶于MeOH(5mL)中,加醋酸(84mg,1.4mmol),乙醛(168μL,5M in THF),室温搅拌1h后,加氰基硼氢化钠(91mg),室温反应24h后,补加乙醛(42μL)、氰基硼氢化钠(23mg),继续反应20h。加2M氢氧化钠溶液调至pH=12,DCM萃取,干燥,浓缩,柱层析得化合物A63,浅黄色油状物72mg。
1H NMR(400MHz,DMSO-d6)δ7.63(s,1H),7.37-7.12(m,4H),6.61(br,2H),2.58(m,3H),2.21(m,1H),2.05(m,2H),1.67(m,2H),1.01(m,3H).ESI-MS m/z 308.11[M+H]+.
实施例60 4-(2-氯苯基)-N4-丙基-4,5,6,7-四氢苯并噻唑-2,4-二胺(化合物A64)的制备
化合物A60(20mg,0.07mmol)溶于甲醇(1mL)中,加乙酸(9mg,0.14mmol),丙醛(5mg,1.2eq),5min后加氰基硼氢化钠(9mg,2eq),搅拌5h。加水10mL,1M氢氧化钠溶液调至pH=10,DCM萃取,干燥,浓缩,柱层析得化合物A64,白色固体(13mg)。
1H NMR(400MHz,MeOD)δ7.49(d,J=7.8Hz,1H),7.36(t,J=7.2Hz,1H),7.30(t,J=7.4Hz,1H),7.06(s,1H),3.37(m,1H),2.79(m,1H),2.74-2.56(m,3H),2.09(m,1H),1.97(m,1H),1.68(m,2H),1.49(m,1H),0.97(t,J=7.4Hz,3H).
实施例61 5-苯基-4,5,6,7-四氢-1H-吲唑-5-胺(化合物A65)的制备
步骤一:
将化合物A23-1(1eq,5g)、叔丁基亚磺酰胺(1.5eq,5.82g)、钛酸四乙酯(3eq,21g)溶于超干四氢呋喃(100mL)中,氮气保护,室温下搅拌过夜。用饱和碳酸氢钠水溶液淬灭。过滤后,滤液用乙酸乙酯萃取3次。干燥浓缩,柱层析得A65-1,7.06g,产率85%。
步骤二:
氮气保护下,将苯基氯化镁(3eq,20mL)加入到250mL三口瓶中。冰浴下加入缓慢加入A65-1的四氢呋喃溶液。加毕,搅拌20min后撤去冰浴室温搅拌。TLC监测反应完毕后,加入饱和氯化铵水溶液淬灭反应。过滤后,EA萃取三次。干燥浓缩,柱层析得A65-2,白色固体3.01g。
步骤三:
将A65-2(1eq,2.5g)溶于丙酮和水的混合溶剂40mL(丙酮∶水=3∶1)。加入PTSA(0.5eq,0.705g)搅拌。TLC监测反应完毕后,加入饱和碳酸氢钠水溶液淬灭。EA萃取三次,干燥浓缩得A65-3,棕红色液体1.675g,产率77.15%。
步骤四:
将A65-3(1eq,1.675g)、DMF-DMA(5eq,3.79mL)在110℃下加热回流过夜。反应完毕后得到A65-4溶液,无需处理直接用于下一步。
步骤五:
将上一步得到的A65-4溶液和水合肼(5eq,1.68g)溶于甲醇,80℃下加热过夜。浓缩至干,得A65-5,棕红色液体2g。
步骤六:
将A65-5(1eq,500mg)溶于2mL的无水乙醇中。搅拌十分钟后加入2M的盐酸溶液搅拌2h。用饱和碳酸氢钠水溶液淬灭,调节pH为8-9,EA萃取三次,干燥浓缩,用氨基柱柱层析得化合物A65,暗红色化合物340mg。
1H NMR(500MHz,Chloroform-d)δ7.58-7.52(m,2H),7.40(s,1H),7.37(t,J=7.6Hz,2H),7.26(s,1H),3.67(d,J=2.0Hz,2H),3.17(d,J=15.6Hz,1H),2.88(dt,J=15.7,7.9Hz,1H),2.76(d,J=15.6Hz,1H),2.69-2.58(m,1H),2.31(ddd,J=14.2,8.8,6.0Hz,1H),2.06(dq,J=13.6,6.8,5.8Hz,1H).ESI-MS m/z 197.34[M+H]+.
实施例62 N-(2-氨基-6-苯基-4,5,6,7-四氢苯并噻唑-6-基)环丙烷甲酰胺(化合物A66)的制备
取A1(15mg,1eq)溶于2mL THF中,加入三乙胺(9mg,1.1eq),冰水浴降温至0℃,缓慢加入环丙烷甲酸酐(14mg,1.1eq),室温反应14小时。加饱和碳酸氢钠溶液淬灭,DCM萃取3次,有机相合并,干燥,浓缩,柱层析纯化,得18mg化合物A66。
1H NMR(400MHz,DMSO-d6)δ8.21(s,1H),7.34(d,J=7.6Hz,2H),7.28(t,J=7.6Hz,2H),7.18(t,J=7.1Hz,1H),6.63(s,2H),3.04(d,J=16.5Hz,1H),2.93(d,J=16.1Hz,1H),2.62(m,1H),2.35(m,1H),2.15(m,2H),1.69(m,1H),0.57(m,4H).ESI-MS m/z 314.22[M+H]+.
实施例63 6-苯基-N6-(环丙基甲基)-4,5,6,7-四氢苯并噻唑-2,6-二胺(化合物A67)盐酸盐的制备
取A1(150mg,1eq)溶于2mL DCM中,加入环丙甲醛(43mg,1eq),室温反应14小时,缓慢加入三乙酰氧基硼氢化钠(130mg,1eq),室温反应1小时。加水淬灭,DCM萃取3次,有机相合并,干燥,浓缩,柱层析纯化,残余物A67于甲醇中加入氯化 氢/甲醇溶液成盐酸盐,浓缩得105mg盐酸盐形式的化合物A67。
1H NMR(400MHz,DMSO-d6)δ10.42(s,1H),9.59(s,1H),9.16(br,2H),7.59(m,2H),7.44(m,3H),3.60(m,2H),2.78-2.53(m,4H),2.13(m,1H),1.76(m,1H),0.98(m,1H),0.50(d,J=8.0Hz,2H),0.24(m,1H),0.064(m,1H).
ESI-MS m/z 300.18[M+H]+.
实施例64 N-苄基-N-甲基-5-苯基-4,5,6,7-四氢-1H-吲唑-5-胺(化合物A68)的制备
步骤一:
化合物A23-2(1eq)溶于乙醇中,加入浓盐酸(12M,3eq),室温搅拌过夜,加入硫酸钠干燥后浓缩溶剂,饱和碳酸氢钠水溶液调节pH=11-12,DCM萃取,干燥过滤浓缩,柱层析得到化合物A68-1,产率83%。
步骤二:
化合物A68-1溶于甲苯中,加入无水硫酸镁干燥,过滤待用;另取反应瓶中加入甲苯、叔丁醇钠(2eq),再加入甲酸乙酯(1.5eq),0-10℃下加入上述化合物A68-1的溶液,室温搅拌过夜。将反应液用甲苯萃取2次,保留水相并用1M盐酸溶液调节pH=6-7,二氯甲烷萃取3次,干燥、浓缩、柱层析得到化合物A68-2,产率93%。
步骤三:
化合物A68-2(1eq,90mg)溶于乙醇中,加入85%水合肼(3eq,97mg),升温至85℃加热5h。加入无水硫酸钠干燥后,浓缩干溶剂,残余物柱层析得化合物A68,白色固体,产率98%。
1H NMR(500MHz,Chloroform-d)δ7.63-7.51(m,2H),7.47(s,1H),7.31(d,J=8.1Hz,3H),7.27(d,J=2.5Hz,3H),7.23(qd,J=6.9,3.7Hz,2H),3.57(d,J=13.3Hz,1H),3.43(d,J=13.3Hz,1H),3.16(s,2H),2.70(ddd,J=16.0,4.9,3.0Hz,1H),2.43(td,J=12.2,5.0Hz,1H),2.24(s,3H),2.21(s,1H),1.97(ddd,J=16.4,11.9,4.8Hz,1H).ESI-MS m/z 318.18[M+H]+.
实施例65 N-甲基-5-苯基-4,5,6,7-四氢-1H-吲唑-5-胺(化合物A69)的制备
取化合物A68(1eq,25mg)溶于硝基甲烷中,室温下加入Oxone氧化剂(3eq,81mg)。30℃下搅拌24h。加入硫代硫酸钠淬灭反应,EA萃取三次,用饱和食盐水洗涤两次,干燥、浓缩、柱层析得化合物A69。
1H NMR(500MHz,Chloroform-d)δ7.50-7.40(m,2H),7.35(s,1H),7.32(dd,J=8.4,7.0Hz,2H),7.25-7.21(m,1H),3.13(d,J=15.5Hz,1H),2.82(d,J=15.5Hz,1H),2.73(ddd,J=16.3,7.4,5.6Hz,1H),2.46(dt,J=16.9,6.6Hz,1H),2.30(q,J=6.7Hz,2H),2.13(s,3H).ESI-MS m/z 228.11[M+H]+.
实施例66 5-苯基-5-(哌啶-1-基)-4,5,6,7-四氢-1H-吲唑(化合物A70)的制备
步骤一:化合物A48-3(1eq,0.7g)溶于甲苯中,加入无水硫酸镁干燥,过滤待用;另取反应瓶中加入甲苯、叔丁醇钠(2eq,523mg),再加入甲酸乙酯(1.5eq,303mg),0-10℃下加入上述化合物A48-3的甲苯溶液,室温搅拌过夜。将反应液用甲苯萃取2次,保留水相并用1M盐酸溶液调节pH=6-7,二氯甲烷萃取3次,干燥、浓缩、柱层析得到化合物A70-1,0.72g,产率93%。
步骤二:化合物A70-1(1eq,90mg)溶于乙醇中,加入85%水合肼(3eq,97mg),升温至85℃加热5h。加入无水硫酸钠干燥后,浓缩干溶剂,残余物柱层析得化合物A70,白色固体87mg,产率98%。
1H NMR(500MHz,Chloroform-d)δ7.46-7.32(m,3H),7.19(dt,J=14.0,7.0Hz,3H),3.37-2.83(m,2H),2.61(d,J=15.9Hz,3H),2.34(s,3H),2.10-1.75(m,2H),1.68-1.21(m,6H).ESI-MS m/z 282.31[M+H]+.
手性柱分离得到异构体A70-P1(S构型)和A70-P2(R构型):
A70-P1(S构型):HPLC纯度:>95%,保留时间:10.024min。手性纯度:>99%,保留时间:6.319min。
A70-P2(R构型):HPLC纯度:>95%,保留时间:10.009min。手性纯度:>95%,保留时间:7.971min。
实施例67 6-苯基-6-(哌啶-1-基)-4,5,6,7-四氢苯并噻唑(化合物A71)的制备
取A48碱式(160mg,1.0eq)溶于THF(2mL),加入亚硝酸叔丁酯(65mg,1.1eq),60℃反应5h。加入饱和碳酸氢钠溶液淬灭,乙酸乙酯萃取,浓缩,柱层析纯化,得到化合物A7125mg,无色油状物。1H NMR(500MHz,Chloroform-d)δ8.57(s,1H),7.33(m,2H),7.22(m,3H),3.27(m,2H),2.84(d,J=15.4Hz,1H),2.60(m,2H),2.36(m,3H),2.05(m,2H),1.80-1.33(m,6H).ESI-MS m/z 299.20[M+H]+.
实施例68 6-((甲氨基)甲基)-6-苯基-4,5,6,7-四氢苯并[d]噻唑-2-胺(化合物A252)的制备
步骤一:250ml三口瓶中加入A252-1(5.5g,1.0eq),乙二醇(1.7g,1.05eq),甲苯(100ml)氮气保护下,升温至120℃反应2-3h。反应液降温至室温,100ml饱和碳酸氢钠洗涤,收 集有机相,100ml饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩得A252-2,油状物5.2g。
步骤二:250ml三口瓶中加入上述油状物A252-2,50ml四氢呋喃搅拌溶清,氮气置换,冰水浴降温至0℃左右,分批次加入氢化铝锂(1.5g,1.5eq),待加入完毕后,升温至70℃左右搅拌2h。反应液冰水浴降温至0℃,饱和氯化铵溶液淬灭,100ml乙酸乙酯萃取,有机相用50ml饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩,得A252-3,类白色固体5.5g。
步骤三:100ml三口瓶中加入A252-3(2.2g),20ml甲酸乙酯搅拌溶解,氮气保护下升温至70℃反应2h左右。减压浓缩得A252-4,类白色固体2g。
步骤四:50ml三口瓶中加入A252-4(650mg),10ml二氯甲烷搅拌溶清,加入三溴吡啶翁(950mg,1.1eq),室温搅拌4-5h。5ml饱和硫代硫酸钠溶液淬灭反应,20ml二氯甲烷萃取,合并有机相,干燥、浓缩得450mg类白色固体。加入10ml二氧六环搅拌溶清,加入硫脲(615mg,3.0eq),DIPEA(700mg,2.0eq),升温至80-90℃搅拌过夜。反应液冷却至室温,加入水、二氯甲烷各10ml萃取,有机相干燥、浓缩,柱层析,得A252-5,类白色固体350mg。
步骤五:50ml三口瓶中加入A252-5(200mg),10ml四氢呋喃搅拌溶清,氮气保护下,加入硼烷四氢呋喃溶液(7ml,10eq),升温至60℃保温反应2h。降温至0℃左右,缓慢滴加5ml甲醇淬灭反应,浓缩除去溶剂,加入水和甲醇各10ml,升温至70℃搅拌1-2h,降温至室温,加入20ml二氯甲烷萃取,有机相无水硫酸钠干燥,浓缩,柱层析得类白色固体100mg。1H NMR(400MHz,DMSO-d6)δ7.43(s,1H),7.37-7.26(m,3H),7.24-7.17(m,1H),6.63(s,2H),5.48(s,1H),3.44(d,J=16.2Hz,1H),3.05-2.94(m,2H),2.88(d,J=15.7Hz,1H),2.83-2.78(m,1H),2.20-2.06(m,2H),1.87-1.79(m,1H),1.70(d,J=5.2Hz,3H).ESI-MS m/z 274.18[M+H]+
在下表中显示的化合物A8、A10、A11、A26、A72-A396,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制备。



























































以下药理实施例中被测试化合物为外消旋体用“rac”表示,纯对映体用“R”或“S”表示。
药理实施例1 NMDA受体拮抗活性测试
采用电生理全细胞手动膜片钳方法,测试化合物对NMDA受体(N-甲基-D-天冬氨酸受体,NR1/2A)通道电流的影响。
实验仪器:
膜片钳放大器(Multiclamp 700B,Axopatch 200B,Axon,美国)
数模转换器(Digidata 1440A,Digidata 1550B,Axon,美国)
倒置显微镜(IX71,IX51,Olympus,日本)
快速给药系统(RSC-200,Bio-Logic,法国)
微操作器(MX7600R,Syskiyou,美国)
电极拉制仪(P-97,Sutter,美国)
玻璃电极(BF150-86-10,Sutter,美国)
防震台与屏蔽网(63-534,TMC,美国)
数据采集与分析软件(pClamp,Axon,美国)
二氧化碳培养箱(HERAcell 150i,Thermo,美国)
生物安全柜(MODEL 1384,Thermo,美国)
纯水仪(Milli Q,Millipore,美国)
试剂:
氯化钠(NaCl)(Sigma,Cat:S7653)
氯化钾(KCl)(Sigma,Cat:P9333)
氯化铯(CsCl)(Sigma,Cat:V900481)
氟化铯(CsF)(Sigma,Cat:289345)
氯化钙(CaCl2)(Sigma,Cat:21115)
葡萄糖(Glucose)(Sigma,Cat:G7528)
4-(2-羟乙基)哌嗪-1-乙磺酸,N-(2-羟乙基)哌嗪-N’-(2-乙磺酸)(简称HEPES)(Sigma,Cat:H3375)
乙二醇双(2-氨基乙基醚)四乙酸(简称EGTA)(Sigma,Cat:E3889)
Lipofectamine 3000转染试剂盒(Gibco,Cat:L3000015)(包含Lipofectamine 3000和P3000两种试剂)
DMEM(Gibco,Cat:C11995500BT)
胎牛血清(FBS)(Gibco,Cat:10099141)
Opti-MEM(Gibco,Cat:31985070)
氢氧化钠(NaOH)(国药,Cat:10019718)
氢氧化铯(CsOH)(Sigma,Cat:232068)
二甲亚砜(DMSO)(Sigma,Cat:276855)
谷氨酸(Glutamic acid)(Sigma,Cat:G1626-100G)
甘氨酸(Glycine)(Amresco,Cat:0167-1KG)
细胞外液配方(mM):140 NaCl,2.8 KCl,1 CaCl2,10 HEPES and 20 Sucrose,用NaOH调节pH至7.4。
细胞内液配方(mM):10 CsCl,115 CsF,10 EGTA and 10 HEPES,用CsOH调节pH至7.2。
具体操作:
a.细胞培养和处理
HEK293细胞系在含有10%胎牛血清的DMEM培养基中培养,培养温度为37℃,二氧化碳浓度为5%。
细胞传代:除去旧培养基并用PBS洗一次,然后加入1mL 0.25%Trypsine-EDTA溶液,室温孵育1分钟。当细胞从皿底脱离,加入3mL 37℃预热的完全培养基(90%DMEM+10%FBS)。将细胞悬液用吸管轻轻吹打使聚集的细胞分离。将细胞悬液转移至无菌的离心管中,800rpm离心3分钟收集细胞。按1∶5的比例将细胞接种于T25细胞培养瓶中(最终体积:6mL),用于扩增或维持培养。
瞬时转染:瞬转前24h将细胞密度80%左右的HEK-293细胞重铺至35mm2的细胞培养皿中,每个细胞培养皿接种量为3×105个细胞。
每孔按如下体积配制瞬转试剂:
1)取7.5μL Lipofectamine 3000加入至250μL Opti-MEM培养基中,轻轻吹打混匀,作为组分A,室温孵育5min;
2)取pCDNA5-FRT-TO-hNR1-T2A-2A质粒3.6μg、GFP质粒0.4μg、7.5μL P3000加入至250μL Opti-MEM培养基中,轻轻吹打混匀,作为组分B,室温孵育5min;
3)将B组分加入A中,轻轻吹打至充分混匀,室温孵育15min形成DNA脂质体混合物;
4)将DNA-脂质体混合物加入到35mm2的细胞培养皿中,每孔500μL。放入培养箱继续培养;
5)6h后换液,将35mm2的细胞培养皿中的培养基全部吸出,加入2mL/孔的完全培养基(90%DMEM+10%FBS),继续培养18h后进行膜片钳检测;
b.化合物准备:测试当天,将本发明化合物母液用DMSO稀释成中间浓度,然后用细胞外液(含100μM Glutamic acid+100μM Glycine)稀释得到需要测试的最终浓度。最终测试浓度中的DMSO含量不超过0.2%。
c.电生理记录过程
瞬时表达NMDA受体通道的HEK293细胞,在室温下用全细胞膜片钳技术记录100μM Glutamic acid(含100μM Glycine)诱发的电流。玻璃微电极由玻璃电极毛胚(BF150-86-10,Sutter)经拉制仪拉制而成,灌注电极内液后的尖端电阻为2-5MΩ左右,将玻璃微电极插入放大器探头即可连接至膜片钳放大器。钳制电压和数据记录由pClamp软件通过电脑控制和记录,采样频率为10kHz,滤波频率为2kHz。在得到全细胞记录后,细胞钳制在-70mV,用快速给药系统以重力方式给予100μM Glutamic acid(含100μM  Glycine)诱发通道电流,待电流稳定后给予含化合物的100μM Glutamic acid(含100μM Glycine)观察电流幅度变化情况,化合物从低浓度至高浓度连续给药,最后再次给予100μM Glutamic acid(含100μM Glycine)。化合物每个测试浓度至少给予20s,每个浓度至少测试2个细胞(n≥2)。
d.数据处理
数据分析处理采用pClamp,GraphPad Prism 8和Excel软件。不同化合物浓度对通道电流(-70mV下100μM Glutamic acid(含100μM Glycine)诱发的电流幅度)的抑制程度用以下公式计算:
Inhibition%=[1-(I/Io)]×100%
其中,Inhibition%代表化合物对NMDA通道电流的抑制百分率,I和Io分别表示在加药后和加药前100μM Glutamic acid(含100μM Glycine)诱发的电流幅度。
化合物IC50使用GraphPad Prism 8软件通过以下方程拟合计算得出:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
其中,X为供试品检测浓度的Log值,Y为对应浓度下抑制百分率,Bottom和Top分别为最小和最大抑制百分率。部分化合物的测试结果见表1。
表1:



以上数据显示,本发明实施例化合物对NMDA受体具有一定的拮抗活性,预期对NMDA受体相关的中枢神经系统疾病具有治疗作用。
药理实施例2:人肝微粒体实验
对照品和试剂:NADPH、氯化镁、睾酮、磷酸缓冲液等由大连美仑生物技术有限公司等提供。
肝微粒体:人肝微粒体由Corning公司提供。
实验步骤:肝微粒体孵育于96孔板中进行,每个孵育体系体积为200μL,介质为0.1M磷酸缓冲液(pH 7.4),包括终浓度为0.2mg/mL的肝微粒体、1μM的受试药物、3.0mM MgCl2、0.01%DMSO、0.5%乙腈及2.0mM NADPH。采用回旋式水浴恒温震荡器,先以不含NADPH的上述孵育体系37℃预孵5min后,加入NADPH起始反应,分别在0、5、15、30、60min混匀后从孵育体系中取出20μL样品加入到200μL含内标的乙腈终止反应。相同条件下1μM睾酮(Testosterone)作为阳性对照,用来检测反应体系的可靠性。对于阴性对照,用磷酸缓冲液代替NADPH,其他孵育条件同上,60min时混匀后取出20μL样品加入到200μL含内标的终止液终止反应。
样品分析:孵育样品经有机溶剂抽提沉淀蛋白等处理后,采用液相色谱-串联质谱(LC-MS/MS)方法,对样品中的受试药物或阳性对照药物的浓度进行半定量测定。使用分析物峰面积与内标峰面积的比值来表示样品中的浓度。
数据处理和分析:使用Excel软件,以孵育体系中药物的ln剩余率对孵育时间作图, 进行线性回归得到斜率k,按以下公式求半衰期T1/2(min)、固有清除率CLint(mL/min/kg):
T1/2=-0.693/k
实验结果见下表2:
表2
NA:无法计算。
以上数据显示,本发明实施例化合物在人肝微粒体中具有良好的代谢稳定性。
药理实施例3:小鼠体内代谢实验
动物:雄性ICR小鼠。动物被随机分配到治疗组,并在给药前12h禁食。
药物:本发明化合物用溶媒(5%DMSO+5%Solutol HS15+90%Saline)溶解。静脉注射(iv)剂量为2.5mg/kg,给药体积为5mL/kg;口服(po)剂量为10mg/kg,给药体积为10mL/kg体重。样品采集和生物分析:小鼠于0.083、0.25、0.5、1、2、4、6、8h(每个时间点n=3)从眼眶取血放入含有肝素钠的EP管中。小鼠取血后立即脱椎处死,取下对应时间点的小鼠大脑。所有样本立即收集,快速冷冻并置于-70℃备用。血浆样本是通过全血离心得到,再取10μL血浆与190μL内标溶液(20ng/mL溶于含0.1%甲酸乙腈)混匀。混匀后在13000rpm下离心10min,每个取120uL上清液,然后取出0.5-10ul(根据化合物的灵敏度决定)采用合适的液相色谱-串联质谱(LC-MS/MS)方法进行药物分析,每个分析物的标准品用于校准和鉴定。按脑组织∶提取液(50%乙腈水)=1(g)∶8(mL),在OMNI Bead Ruptor 24 Elite匀浆机中使用3mm磁珠5颗在速度3.2m/s下匀浆2min,4℃,3000g条件下,离心3min,取上清液50uL,加入200uL内标溶液(20ng/mL溶于80%乙腈水)混匀,混匀后在13000rpm下离心10min,每个取120uL上清液,然后取出0.5-10ul(根据化合物的灵敏度决定)上清液采用合适的LC-MS/MS方法进行药物分析,每个分析物的标准品用于校准和鉴定。
数据分析:低于定量下限(LLOQ)的药物浓度为零。使用非室间、快速浓注或血管外输入分析模型在PK Solver中进行药代动力学数据分析。LLOQ以下的数据点不参与分析,以提高t1/2计算的有效性。实验结果见下表3:
表3

以上数据显示,本发明实施例化合物在小鼠体内具有良好的口服生物利用度。
药理实施例4:sigma受体亲和力实验
4.1材料与试剂
细胞膜:本实验采用的膜蛋白提取于药明康德构建的稳定表达Sigma1受体的HEK293细胞株和稳定表达Sigma2受体的HEK293细胞株。
试剂:
[3H]-DTG(PerkinElmer,Cat:NET986250UC)
Haloperidol(Sigma,Cat:Sigma-H1512-5G)
Tris-HCl(Sigma,Cat:T3038-1L)
NaCl(Sigma,Cat:S5150-1L)
PEI(Poly ethyleneimine)(Sigma,Cat:P3143)
Microscint 20 cocktail(PerkinElmer,Cat:6013329)
Haloperidol(Sigma,Sigma-H1512-5G)
仪器与耗材:
Top Seal-A封板膜(Perkin Elmer,Cat#6050185)
96 well conical polypropylene plates(Agilent,Cat#50421385)
96-well细胞收集器(PerkinElmer,Cat#C961961)
Unifilter-96GF/C过滤板(Perkin Elmer,Cat#6005174)
Microbeta(PerkinElmer)
4.2方法与步骤
1)检测缓冲液:50mM Tris-HCl,pH=7.4,4℃保存;
2)Sigma 1R实验洗板缓冲液:50mM Tris-HCl,pH=7.4,4℃保存;Sigma2R实验洗 板缓冲液:50mM Tris-HCl,100mM NaCl,pH=7.4,4℃保存;
3)将化合物稀释到各工作浓度:8个浓度4倍连续梯度稀释。
用预冷的检测缓冲液配置细胞膜溶液及同位素溶液,配置信息如下:膜浓度10μg/well,同位素配体[3H]-DTG的最终浓度5nM。
5)将待测化合物和阳性对照稀释至工作浓度后,向实验板内按照每孔1μL加入。Sigma 1R实验的高信号对照孔(High control)加入1μL DMSO,低信号对照孔(Low control)中加入1μL 100μM的Haloperidol(终浓度500nM)。Sigma 2R实验的高信号对照孔(High control)加入1μL DMSO,低信号对照孔(Low control)中加入1μL 400μM的Haloperidol(终浓度2uM)。
6)按照每孔100μL细胞膜加入到实验板,然后加入100μL对应的同位素配体。
7)密封实验板,37℃条件下震荡孵育2小时。用0.3%PEI浸泡GF/C过滤板,至少30分钟。
8)孵育完毕用细胞收集器将细胞膜收集到GF/C过滤板上,用预冷的洗板缓冲液洗4次后,将GF/C过滤板置于50℃烘箱中干燥1小时。
9)把干燥好的GF/C过滤板底部封膜,每孔加入50μL闪烁液,并密封。
10)使用Microbeta读数。
11)使用GraphPad Prism分析数据,计算IC50值和Ki值。亲和力抑制常数Ki采用Cheng Prusoff公式计算:Ki=IC50/(1+L/KD)。
实验结果见下表4:
表4

以上表4数据显示,本发明实施例化合物对sigma受体具有较好的亲和力,预期对与sigma受体相关的中枢神经系统疾病具有治疗作用。
药理实施例5:强迫游泳实验(Forced Swim Test):
药物:本发明化合物用5%DMSO混匀后加入5%HS 15混匀,再加入90%生理盐水,配制成合适浓度的溶液,现配现用。
动物:雄性C57小鼠,22g左右。对动物随机分组,分为空白对照组和各受试药组,每组动物8只,各组小鼠分别腹腔注射给予溶媒处方或者各受试药。
实验步骤:给药后0.5h对小鼠进行强迫游泳测试。强迫游泳设备中水位为45cm,水温25℃,实验开始前将小鼠置于实验房间适应环境1h。实验开始时将小鼠置于设备中,时长6min,整个过程用摄像头记录,分析数据时只统计最后4min小鼠的不动时间。
实验结果见下表5:
表5

以上数据显示,药物组在1-5mg/kg低剂量下都表现出了显著的抗抑郁样作用。
药理实施例6:单胺转运体抑制活性测试
(1)5-HT转运体抑制活性测试:
主要试剂与仪器:
HEPES(Invitrogen,Cat:15630-106)
HBSS(Invitrogen,Cat:14025)
Bovine Serum Albumin(Sigma,Cat:B2064-100G)
Neurotransmitter transporter uptake assay kit(Molecular devices,Cat:R8174)
Incubator(Thermo,240)
Envision(Perkin Elmer,envision2014)
384孔板(Greiner,Cat:784075)
在过表达人SERT的HEK-293细胞中,使用Neurotransmitter transporter uptake assay kit(Molecular devices)检测待测化合物对人SERT的转运体抑制作用。测试按照试剂盒说明书中的方法进行,采用西酞普兰(Citalopram)作为阳性对照。具体操作如下:
a)将HEK-293-hSERT细胞以20000个/孔接种到384孔板中,然后将384孔板转移至培养箱中,37℃过夜;
b)次日,于384孔板中,用实验缓冲液(含0.1%BSA及20mM HEPES的HBSS溶液)配制西酞普兰和本发明化合物的测试溶液,西酞普兰的测试起始浓度为1μM,3X稀释,待测化合物测试起始浓度为10μM或100μM,3X稀释,每个浓度重复2次;
c)从培养箱中取出培养有HEK-293-hSERT细胞的384孔板,移去孔中培养基,每孔中加入25μL待测试化合物溶液;在37℃培养箱中孵育30min;
d)每孔加入25μL染料,在37℃培养箱中孵育30min;
e)在Envision上读取荧光值,使用Graphpad Prism软件分析数据,结果见表6。
(2)DAT和NET转运体抑制活性测试:
主要试剂与仪器:
使用Neurotransmitter transporter uptake assay kit对表达人DAT和NET的HEK-293细胞的转运体抑制作用。测试按照试剂盒说明书中的方法进行,采用Centanafadine作为阳性对照。具体操作如下:
NET瞬转细胞准备:
第一天细胞铺板:HEK 293T细胞经胰酶消化处理,离心后用培养基重悬,经细胞计数后接种于6em培养皿中,接种密度为3×106cells/孔;
第二天细胞转染:HEK 293T细胞进行换液处理,随后将待转染NET-pcDNA5/FRT质粒分别准备A、B两管。A管加入200μL Opti-MEM,然后加入10μL LipofectamineTM3000混合均匀。B管先加入200μL Opti-MEM,然后加入5μg NET-pcDNA5/FRT质粒,混合均匀,再在B管中加入10μL P3000TM混合均匀(质粒与转染试剂比例为1μg∶2μL)。将稀释后的A管溶液加入到稀释后的B管溶液中,混合均匀,室温孵育15分钟。最后将混合物轻轻加入已经换液的细胞中,轻轻摇动以混合,然后于温度为37℃,二氧化碳浓度为5%的培养箱中培养过夜。转染18-20小时后细胞用于化合物功能活性测定。
DAT稳转培养:DAT-HEK细胞系在含有10%胎牛血清及0.2mg/mL Hygromycin B的DMEM培养基中培养,培养温度为37℃,二氧化碳浓度为5%。
DAT稳转细胞传代:除去旧培养基并用PBS洗一次,然后加入1mL TrypLETM Express溶液,37℃孵育2min左右。当细胞从皿底脱离,加入约5mL 37℃预热的完全培养基。 将细胞悬液用吸管轻轻吹打使聚集的细胞分离。将细胞悬液转移至无菌的离心管中,1000rpm离心5min。为维持细胞的生理活性,实验细胞融合度控制在80%左右。
a)NET细胞转染过夜后,经胰酶消化处理,用DMEM+10%Dialyzed FBS培养基重悬后接种20000cells/孔细胞于384孔板中培养过夜。稳转的DAT细胞经胰酶消化处理,用DMEM+10%Dialyzed FBS培养基重悬后接种2500cells/孔细胞种于384孔板中培养过夜。
b)按照试剂盒说明书配制1×Assay Buffer待用;将阳性化合物及待测化合物用DMSO进行梯度稀释,然后用1×Assay Buffer稀释至2×;
c)离心去掉384孔板中的培养基;
d)取16μL步骤b)中配制好的化合物加入到相应实验孔中,其中阳性对照孔加入首浓度的2×阳性对照化合物,阴性对照孔加入0.2%DMSO buffer,离心后置于37℃孵育30min;
e)用1×HBSS配制检测试剂,每孔加入16μL的检测试剂,离心后置于37℃孵育60min;
f)孵育完成后,利用酶标仪检测波长425nm激发下,510nm读值,用GraphPad Prism软件非线性回归方法进行曲线拟合及IC50计算。结果见下表6。
表6



以上数据显示,本发明化合物对单胺转运体具有一定的拮抗活性,预期对单胺转运体相关联的中枢神经系统疾病具有治疗作用。

Claims (11)

  1. 一种式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物:
    其中:
    A环选自4-10元杂环或C6-C10芳环并4-10元杂环;所述4-10元杂环中含有1-3个选自N、O和S的杂原子,优选为5-8元杂环;优选地,环A中杂环中含有1个N原子和1个S原子,或者含有两个N原子,或者含有1个S原子,或者含有两个N原子和1个S原子,或者含有1个N原子和1个O原子;
    所述A环任选地被一个或多个R5取代,各R5独立选自卤素、羟基、氨基、氰基、羧基、氧代、C1-C6烷基、卤代C1-C6烷基、C1-C6烷氧基、C1-C6烷酰基、氨基甲酰基(-CONH2)、被C1-C6烷基取代的氨基甲酰基、被一个或两个C1-C6烷基取代的氨基、被一个或两个卤代C1-C6烷基取代的氨基、被一个或两个C1-C6烷酰基取代的氨基、C1-C6烷氧基羰基、C3-C6环烷基、4-10元杂环烷基、C6-14芳基、5-10元杂芳基、C6-C14芳基C1-C6烷基氧基或者5-10元杂芳基C1-C6烷基氧基;优选地,各R5独立选自卤素、羟基、氨基、氰基、羧基、氧代、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、C1-C4烷酰基、氨基甲酰基(-CONH2)、被C1-C4烷基取代的氨基甲酰基、被一个或两个C1-C4烷基取代的氨基、被一个或两个卤代C1-C4烷基取代的氨基、被一个或两个C1-C4烷酰基取代的氨基、C1-C4烷氧基羰基、C3-C6环烷基、4-8元杂环烷基、C6-10芳基、5-10元杂芳基、C6-C10芳基C1-C4烷基氧基或者5-10元杂芳基C1-C4烷基氧基;更优选地,各R5独立选自卤素(特别是溴)、氨基、羟基、氰基、羧基、C1-C3烷基(特别是甲基、乙基、异丙基)、卤代C1-C3烷基(特别是三氟甲基)、C1-C3烷氧基(特别是甲氧基、乙氧基)、C1-C3烷酰基(特别是甲酰基、乙酰基)、氨基甲酰基(-CONH2)、甲酰基氨基、乙酰基氨基、甲氨基、乙氨基、N,N-二甲基氨基、2,2,2-三氟乙基氨基、C1-C3烷氧基羰基(特别是甲氧基羰基、乙氧羰基)、C3-C5环烷基(特别是环丙基)、苯基、吡啶基、吡咯烷基、哌啶基、吗啉基或苄氧基;
    环B为3-10元碳环;优选为5-8元碳环,例如5、6、7或8元碳环,更优选为5、6 或7元碳环;
    R1连接于环B中同一个环碳原子;
    x为0-2的整数;
    R2和R3各自独立地选自氢、C1-C6烷基、卤代C1-C6烷基、C1-C6烷氧基C1-C6烷基、羟基C1-C6烷基、C6-C14芳基C1-C6烷基、C3-C6环烷基、C1-C6烷酰基、卤代C1-C6烷酰基、C3-C6环烷基C1-C6烷酰基、C6-C14芳基C1-C6烷酰基、C1-C6烷磺酰基、C1-C6烷基亚砜基或C3-C6环烷基C1-C6烷基;优选各自独立地选自氢、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基C1-C4烷基、羟基C1-C4烷基、C6-C14芳基C1-C4烷基、C3-C6环烷基、C1-C4烷酰基、卤代C1-C4烷酰基、C3-C6环烷基C1-C4烷酰基、C6-14芳基C1-C4烷酰基、C1-C4烷磺酰基、C1-C4烷基亚砜基或C3-C6环烷基C1-C4烷基;优选选自氢、C1-C4烷基(特别是甲基、乙基、丙基、异丙基、叔丁基)、卤代C1-C4烷基(特别是1,1,1-三氟乙基)、甲氧基乙基、羟甲基、羟乙基、C3-C6环烷基甲基(特别是环丙基甲基、环丁基甲基)、苄基、C3-C6环烷基(特别是环丙基)、C1-C3烷基酰基(特别是乙酰基、丙酰基)、环丙基甲酰基、苯甲酰基、叔丁基亚砜基;
    或者,R2、R3和相连的氮原子一起形成3-9元杂环烷基,所述3-9元杂环烷基环中任选地包含一个或多个额外的氮原子或氧原子,所述3-9元杂环烷基任选地被一个或多个C1-C6烷基,优选C1-C4烷基取代;优选地,R2、R3和相连的氮原子一起形成氮杂环丁烷基、吡咯烷基、哌啶基、氮杂环庚烷基、吗啉基;
    R1选自C6-C14芳基、5-10元杂芳基、4-10元杂环基、C6-C10芳基并4-10元碳环(例如茚满基)或C6-C10芳基并4-10元杂环(例如1,2-甲撑二氧基苯基、2,3-二氢苯并呋喃基),优选选自C6-C14芳基、5-10元杂芳基,更优选为苯基、萘基、喹啉基、异喹啉基、吡啶基、嘧啶基、吡嗪基、哒嗪基、噻吩基或噻唑基,还优选为苯基、萘基、嘧啶基、吡啶基、吡嗪基、哒嗪基或喹啉基,进一步优选为苯基;
    所述C6-C14芳基、5-10元杂芳基、4-10元杂环基、C6-C10芳基并4-10元碳环或C6-C10芳基并4-10元杂环任选地被一个或多个R6取代;
    R6各自独立地选自卤素、羟基、巯基、氰基、氨基甲酰基(NH2CO-)、氨基磺酰基(NH2SO2-)、C1-C6烷基、卤代C1-C6烷基、羟基C1-C6烷基、C3-C6环烷基、C3-C6环烷氧基、被C1-C3烷基取代的C3-C6环烷基、被C1-C3烷基取代的C3-C6环烷氧基、C1-C6烷氧基、C1-C6烷硫基、卤代C1-6烷氧基、C3-C6环烷基C1-C6烷氧基、C1-C6烷酰基氧基、C6-C14芳基、5-10元杂芳基、C6-C14芳基C1-C6烷氧基、5-10元杂芳基 C1-C6烷氧基;优选地,R6各自独立地选自卤素、氰基、羟基、氨基甲酰基(NH2CO-)、氨基磺酰基(NH2SO2-)、C1-C4烷基、卤代C1-C4烷基、羟基C1-C4烷基、C3-C6环烷基、C3-C6环烷氧基、被C1-C3烷基取代的C3-C6环烷基、被C1-C3烷基取代的C3-C6环烷氧基、C1-C4烷氧基、C1-C4烷硫基、卤代C1-C4烷氧基、C3-C6环烷基C1-C3烷氧基、C1-C3烷酰基氧基、C6-C10芳基、5-6元杂芳基、C6-C10芳基C1-C4烷氧基、5-6元杂芳基C1-C4烷氧基;更优选地,R6各自独立地选自氟、氯、溴、氰基、羟基、甲基、异丙基、二氟甲基、三氟甲基、羟甲基、甲基环丙基氧基、甲氧基、乙氧基、异丙氧基、异丁基氧基、甲硫基、环丙基、环丙基氧基、氨基甲酰基(NH2CO-)、氨基磺酰基(NH2SO2-)、二氟甲氧基、三氟甲氧基、1,1,1,-三氟乙氧基、氰基、环丙基甲氧基、乙酰氧基、苯基、吡啶基或苄氧基;
    m为0-3的整数;
    R4连接至B环上除R1共同连接的碳原子外的任意碳原子上,且R4各自独立地选自羟基、卤素、C1-C6烷基。
  2. 根据权利要求1所述的式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物,其特征在于,
    A环选自5元或6元杂环或苯并5元或6元杂环,
    优选地,A环选自:
    其中,Z1、Z2、Z4独立地选自N、CR7,Z3选自NR7、O、S;且Z1、Z2、Z4不同时为CR7;各R7独立选自氢、卤素、氰基、羟基、氨基、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烷氧基、卤代C1-C6烷基、被C1-C6烷基取代的氨基、被卤代C1-C6烷基取代的氨基、被C1-C6烷酰基取代的氨基、C1-C6烷酰基、C1-C6烷氧羰基、C3-C6环烷基、4-8元杂环烷基、C6-10芳基、5-10元杂芳基、C6-C10芳基C1-C4烷基氧基或者5-10元杂芳基C1-C4烷基氧基;
    更优选地,A环选自:
    其中,各R7独立选自氢、卤素(特别是溴)、氨基、羟基、氰基、羧基、C1-C3烷基(特别是甲基、乙基、异丙基)、卤代C1-C3烷基(特别是三氟甲基)、C1-C3烷氧基(特别是甲氧基、乙氧基)、C1-C3烷酰基(特别是甲酰基、乙酰基)、氨基甲酰基(-CONH2)、甲酰基氨基、乙酰基氨基、甲氨基、乙氨基、N,N-二甲基氨基、2,2,2-三氟乙基氨基、C1-C3烷氧基羰基(特别是甲氧基羰基、乙氧羰基)、C3-C5环烷基(特别是环丙基)、吗啉基、苯基、吡啶、吡咯烷基、哌啶基或苄氧基。
  3. 根据权利要求1或2所述的式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物,其中,
    R1为任选地被一个或多个R6取代的苯基或萘基,
    R6定义如权利要求1所述。
  4. 根据权利要求3所述的式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物,其特征在于,
    R2和R3之一为氢,或者
    R2和R3都为氢。
  5. 根据权利要求2所述的式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物,其特征在于,所述式(I)化合物选自下列化合物:
    其中,x、Z1、Z2、Z3、Z4、R2、R3、R6定义如权利要求2所述。
  6. 根据权利要求1所述的式(I)化合物或其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物或同位素标记的化合物,其特征在于,所述式(I)化合物选自下列化合物:

    R2、R3、R6、R7定义如权利要求1所述;
    且苯基、吡啶基、萘基、喹啉基、嘧啶基、吡嗪基或哒嗪基和-NR2R3 连接于它们相连的碳环的同一个环碳原子。
  7. 根据权利要求1-6任一项所述的通式(I)所示的含芳基的胺类化合物,及其立体异构体、几何异构体、构象异构体、互变异构体、药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物,其特征在于,所述式(I)化合物选自以下结构:




















  8. 制备权利要求1-7中任一项所述的式(I)所示含芳基的胺类化合物的方法,所述方法通过如下方法1-3之一或其组合进行:
    方法1:
    如反应式1所示,包括以下步骤:
    a)式(II)所示化合物与式(III)所示化合物进行缩合反应,生成式(IV)所示化合物;
    b)式(IV)所示化合物和式(V)所示化合物进行亲核加成反应,生成式(I-a)所示化合物;
    其中A环、B环、R1同相应权利要求所定义;
    G代表离去基团,选自C1-C6烷基亚磺酰基、苯亚磺酰基、萘亚磺酰基、苄基,所述C1-C6烷基亚磺酰基、苯亚磺酰基、萘亚磺酰基、苄基可选地被一个或多个选自卤素、C1-C6烷基、硝基、羟基、氨基、C1-C6烷酰基、C1-C6烷氧基、苯基的基团取代;
    M代表离去基团,选自金属元素、卤素、金属合物、硼烷、硅烷、重氮盐,优选为-MgBr,-MgCl,-Li;
    方法2:
    如反应式2所示,包括以下步骤:
    c)式(II)所示化合物与式(V)所示化合物进行亲核加成反应,生成式(VI)所示化合物;
    d)式(VI)所示化合物和叠氮化物进行取代反应,生成式(VII)所示化合物;
    e)式(VII)所示化合物发生还原反应生成式(I-a)所示化合物;
    其中A环、B环、R1如相应权利要求所定义;
    M代表离去基团,如金属元素、卤素、金属合物、硼烷、硅烷、重氮盐等,优选为-MgBr,-MgCl,-Li;
    方法3:
    由方法1-2得到的式(I-a)化合物进行氨基的官能团转化得到其他式(I)所示含芳基的胺类化合物。
  9. 一种药物组合物,包含治疗有效量的选自权利要求1-7任一项所述的式(I)所示含芳基的胺类化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,以及可选地一种或多种可药用载体、稀释剂或赋形剂。
  10. 权利要求1-7任一项所述的式(I)所示含芳基的胺类化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,或权利要求9所述的药物组合物在制备调节NMDA受体和/或单胺转运体和/或sigma受体活性的药物中的应用,特别是在制备以下药物中的应用:
    a)NMDA受体拮抗剂;
    b)单胺转运体抑制剂;
    c)sigma受体激动或拮抗剂。
  11. 权利要求1-7任一项所述式(I)所示含芳基的胺类化合物,其立体异构体、几何异构体、构象异构体、互变异构体,它们的药学上可接受的盐、多晶型物、溶剂合物、水合物以及同位素标记的化合物中的一种或多种,或权利要求9所述药物组合物在制备预防和/或治疗与NMDA受体和/或单胺转运体和/或sigma受体相关联的疾病,特别是中枢神经系统疾病的药物中的应用;
    优选地,所述中枢神经系统疾病选自:脑缺血;脑卒中;脑梗塞;创伤性脑损伤;抗NMDA受体脑炎;癫痫;肌萎缩侧索硬化症;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;神经性疼痛;狂躁症;帕金森症;亨廷顿舞蹈症;阿尔茨海默症;痴呆症;记忆障碍;多动症;注意力缺乏/亢进类疾病;抽动症以及由NMDA受体活化引起的其他神经系统事件或神经变性;优选地,所述神经性疼痛包括外周糖尿病性神经病、疱疹后神经痛、复杂局部疼痛综合征、外周神经病、化学治疗诱导的神经性疼痛、癌性神经性疼痛、神经性下背痛、HIV神经性疼痛、三叉神经痛和中枢性中风后疼痛;
    进一步优选地,所述中枢神经系统疾病选自: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;帕金森症;外周糖尿病性神经病;疱疹后神经痛;复杂局部疼痛综合征。
PCT/CN2023/130755 2022-11-09 2023-11-09 一类含芳基的胺类化合物、其制备方法和用途 WO2024099393A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202211401952 2022-11-09
CN202211401952.5 2022-11-09
CN202311134131.4 2023-09-04
CN202311134131 2023-09-04

Publications (1)

Publication Number Publication Date
WO2024099393A1 true WO2024099393A1 (zh) 2024-05-16

Family

ID=91032001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/130755 WO2024099393A1 (zh) 2022-11-09 2023-11-09 一类含芳基的胺类化合物、其制备方法和用途

Country Status (1)

Country Link
WO (1) WO2024099393A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4085280A (en) * 1977-04-07 1978-04-18 E. R. Squibb & Sons, Inc. 7-Aminoalkyl-3,7-diaryl-3,3a,4,5,6,7-hexahydro-2H-indazoles
US4292323A (en) * 1980-03-31 1981-09-29 Schering Corporation Phenyl-1,2,3,4-tetrahydrocarbazoles and use thereof
WO2014025651A1 (en) * 2012-08-06 2014-02-13 Amgen Inc. Chroman derivatives as trpm8 inhibitors
WO2017035366A1 (en) * 2015-08-26 2017-03-02 Incyte Corporation Pyrrolopyrimidine derivatives as tam inhibitors
WO2022140417A1 (en) * 2020-12-21 2022-06-30 Kanna Health Ltd. Methods for delay of ejaculation in human males

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4085280A (en) * 1977-04-07 1978-04-18 E. R. Squibb & Sons, Inc. 7-Aminoalkyl-3,7-diaryl-3,3a,4,5,6,7-hexahydro-2H-indazoles
US4292323A (en) * 1980-03-31 1981-09-29 Schering Corporation Phenyl-1,2,3,4-tetrahydrocarbazoles and use thereof
WO2014025651A1 (en) * 2012-08-06 2014-02-13 Amgen Inc. Chroman derivatives as trpm8 inhibitors
WO2017035366A1 (en) * 2015-08-26 2017-03-02 Incyte Corporation Pyrrolopyrimidine derivatives as tam inhibitors
WO2022140417A1 (en) * 2020-12-21 2022-06-30 Kanna Health Ltd. Methods for delay of ejaculation in human males

Similar Documents

Publication Publication Date Title
CN104024251B (zh) 苯磺酰胺化合物及其作为治疗剂的用途
JP4777974B2 (ja) ヒスタミンh3受容体作用物質、製剤および治療的使用
CN101495184A (zh) 芳基和杂芳基取代的四氢苯并氮杂及其在阻断去甲肾上腺素多巴胺和血清素的重摄取中的应用
TWI391385B (zh) An amine-based indane derivative or a salt thereof
JP5315244B2 (ja) アザビシクロヘキサン誘導体の使用
HUT61719A (en) Process for producing 2-aminotetraline derivatives and pharmaceutical compositions comprising such compounds
JPWO2008059854A1 (ja) ピペリジン誘導体またはその塩
CA2497665A1 (en) Imidazopyridine compounds as 5-ht4 receptor agonists
EP3610875A1 (en) Opioid receptor (mor) agonist salt, fumarate salt i crystal form thereof and preparation method thereof
WO2010151597A1 (en) Methods for using pyrrolo-benzo-1,4-diazines as sodium channel blockers
WO2006033318A1 (ja) 環状アミン誘導体又はその塩
US11633390B2 (en) 5-HT2A agonists for use in treatment of depression
UA125631C2 (uk) Нові модулятори 5-гідрокситриптамінового рецептора 7 і спосіб їх застосування
KR20130031323A (ko) Kv7 칼륨 채널 개방제로서의 피페리디닐 피리미딘 아미드
JP2006515618A (ja) 置換アルキルアミドピペリジン類
NO180195B (no) Analogifremgangsmåte ved fremstilling av heksahydroazepinderivater
WO2022017440A1 (zh) 5-ht 2a受体抑制剂或反向激动剂及其制备方法和应用
JP5538907B2 (ja) セロトニン5−ht6受容体の調節に応答する障害を治療する上で好適なキノリン化合物
EP2987793A1 (en) Heterocyclic acetic acid amide compound
CN111108101A (zh) 用于治疗疼痛和疼痛相关病症的新丙胺衍生物
WO2024099393A1 (zh) 一类含芳基的胺类化合物、其制备方法和用途
JP6772251B2 (ja) オレキシン受容体拮抗薬の化合物の結晶形およびその製造方法と使用
TW201141476A (en) Novel indole derivative
TWI422578B (zh) 縮合茚滿化合物
CN109689650A (zh) 取代的n-[2-(4-苯氧基哌啶-1-基)-2-(1,3-噻唑-5-基)乙基]苯甲酰胺和n-[2-(4-苄氧基哌啶-1-基)-2-(1,3-噻唑-5-基)乙基]苯甲酰胺衍生物p2x7受体拮抗剂