WO2017063509A1 - 氧杂螺环类衍生物、其制备方法及其在医药上的应用 - Google Patents

氧杂螺环类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2017063509A1
WO2017063509A1 PCT/CN2016/101064 CN2016101064W WO2017063509A1 WO 2017063509 A1 WO2017063509 A1 WO 2017063509A1 CN 2016101064 W CN2016101064 W CN 2016101064W WO 2017063509 A1 WO2017063509 A1 WO 2017063509A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cycloalkyl
compound
alkyl
aryl
Prior art date
Application number
PCT/CN2016/101064
Other languages
English (en)
French (fr)
Inventor
李心
冯斌强
陈阳
刘涛
贺峰
何明勋
陶维康
孙飘扬
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to CA3000761A priority Critical patent/CA3000761C/en
Priority to ES16854888T priority patent/ES2772689T3/es
Priority to US15/766,985 priority patent/US10442793B2/en
Priority to MX2018004175A priority patent/MX2018004175A/es
Priority to JP2018515975A priority patent/JP6824502B2/ja
Priority to AU2016339404A priority patent/AU2016339404B2/en
Priority to CN201680003945.6A priority patent/CN107001347B/zh
Priority to KR1020187012600A priority patent/KR20180063251A/ko
Priority to DK16854888.1T priority patent/DK3354649T3/da
Priority to BR112018006448-3A priority patent/BR112018006448A2/zh
Priority to EP16854888.1A priority patent/EP3354649B1/en
Priority to RU2018115569A priority patent/RU2733373C2/ru
Priority to PL16854888T priority patent/PL3354649T3/pl
Publication of WO2017063509A1 publication Critical patent/WO2017063509A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4402Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 2, e.g. pheniramine, bisacodyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the invention belongs to the field of medicine and relates to an oxaspirocyclic derivative, a preparation method thereof and application thereof in medicine.
  • the present invention relates to an oxaspirocyclic derivative represented by the general formula (I), a process for producing the same, and a pharmaceutical composition containing the same, and the use thereof as a MOR receptor agonist and preparation thereof And/or use in drugs for the prevention of pain and pain related diseases.
  • Opioid receptors are an important class of G protein-coupled receptors (GPCRs), which are targets for the binding of endogenous opioid peptides and opioids.
  • GPCRs G protein-coupled receptors
  • the opioid receptors activate the nervous system and the endocrine system. It has a regulating effect, and opioids are the strongest and commonly used central analgesics.
  • Endogenous opioid peptides are naturally occurring opioid active substances in mammals.
  • endogenous opioid peptides are broadly classified into enkephalins, endorphins, dynorphins and neomorphins (Pharmacol Rev 2007). ;59:88–123).
  • opioid receptors in the central nervous system namely ⁇ (MOR), ⁇ (DOR), ⁇ (KOR) receptors and the like.
  • MOR opioid receptor
  • DOR
  • KOR
  • Opioid receptors that mediate analgesic effects (science, 2001, 293: 311-315; Biochem Biophys Res Commun 235: 567-570; Life Sci 61: PL409-PL415).
  • GPCR mediates and regulates physiological functions mainly through two pathways: the G protein pathway and the ⁇ -arrestin pathway.
  • Traditional GPCR agonists bind to receptors and activate G protein signaling pathways, including second messenger systems such as calcium ions, adenyl cyclase (AC), and mitogen-activated protein (mitogen-activated protein).
  • Second messenger systems such as calcium ions, adenyl cyclase (AC), and mitogen-activated protein (mitogen-activated protein).
  • MAPK mitogen-activated protein
  • ⁇ -arrestin-preferred ligands primarily activate the ⁇ -arrestin pathway.
  • the ⁇ -arrestin-mediated GPCR reaction mainly includes three aspects: 1) As a negative regulator, it interacts with G protein-coupled receptor kinase (GRK) to cause receptor desensitization in GPCRs and stop G protein signal transduction.
  • GRK G protein-coupled receptor kinase
  • MOR is a target for endogenous enkephalins and opioid analgesics such as morphine.
  • opioid analgesics such as morphine.
  • endogenous enkephalin and a class of drugs etomorphine can agonize G protein and trigger receptor endocytosis, but morphine does not induce endocytosis of the receptor at all, because morphine is agonistic of MOR phosphorylation.
  • the ability is too weak to recruit only a small amount of beta-arrestin on the membrane (Zhang et al, Proc Natl Acad Sci USA, 1998, 95(12): 7157-7162).
  • beta-arrestin Such ligands exert their physiological functions entirely through the G protein signaling pathway rather than the beta-arrestin pathway.
  • the presently disclosed MOR agonist patent applications include WO2014022733, WO2008009415, WO2009018169, WO2012129495, WO2001049650, WO2002020481, WO2010051476 and WO2013087589, and the like.
  • the object of the present invention is to provide a compound of the formula (IA) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer thereof, or a mixture form, or a pharmaceutically acceptable salt thereof:
  • Ring A is selected from the group consisting of a cycloalkyl group and a heterocyclic group
  • R is selected from the group consisting of aryl and heteroaryl, wherein the aryl and heteroaryl are optionally selected from the group consisting of alkyl, haloalkyl, halo, amino, nitro, cyano, alkoxy, haloalkoxy, hydroxy Alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -OR 3 , -C(O)R 3 , -C(O)OR 3 , -S(O) m R 3 and -NR 4 Substituting one or more substituents in R 5 ;
  • R 1 is the same or different and is each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogen, an amino group, a nitro group, a hydroxyl group, a cyano group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group.
  • alkyl group, haloalkyl group, cycloalkyl group And a heterocyclic group, an aryl group and a heteroaryl group are optionally selected from the group consisting of alkyl, haloalkyl, halogen, amino, nitro, cyano, hydroxy, alkoxy, haloalkoxy, hydroxyalkyl, cycloalkyl, Substituting one or more substituents in a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 2 are the same or different and are each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogen group, an amino group, a nitro group, a hydroxyl group, a cyano group, an oxo group, an alkenyl group, a cycloalkyl group, a heterocyclic ring.
  • alkyl group Alkoxy, alkenyl, haloalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally selected from the group consisting of a halogen atom, an alkyl group, a halogenated alkyl group, a halogen, an amino group, a nitro group, a cyano group, and a hydroxyl group. Substituted with one or more substituents of alkoxy, haloalkoxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • the two R 2 together form a cycloalkyl or heterocyclic group, wherein the cycloalkyl or heterocyclic group is optionally selected from the group consisting of alkyl, haloalkyl, halogen, amino, nitro, cyano, hydroxy, Substituted with one or more substituents of alkoxy, haloalkoxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 3 is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, an amino group, an alkoxy group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group, wherein the alkyl group, the cycloalkyl group And a heterocyclic group, an aryl group and a heteroaryl group are optionally selected from the group consisting of alkyl, halogen, hydroxy, amino, nitro, cyano, alkoxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and Substituting one or more substituents in the heteroaryl;
  • R 4 and R 5 are each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, a hydroxyalkyl group, a hydroxyl group, an amino group, a carboxylate group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group.
  • alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups are optionally selected from the group consisting of alkyl, halogen, hydroxy, amino, carboxylate, nitro, cyano, alkoxy, hydroxyalkane Substituted by one or more substituents of a group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • p, q are each independently 0, 1, 2, 3 or 4;
  • n 0, 1, or 2.
  • the compound of the formula (IA) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound of the formula (I):
  • Rings A, R, R 1 , R 2 , p and q are as defined in formula (IA).
  • the compound of the formula (I)) or formula (IA) or a tautomer, a mesogen, a racemate, an enantiomer thereof, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 2 is the same or different and is each independently selected from the group consisting of a hydrogen atom, an alkyl group, an oxo group, an alkoxy group, a hydroxyl group, a halogen, and OR 3 , wherein the alkyl group and the alkoxy group are optionally selected from the group consisting of a halogen atom, an alkyl group, a halogen, a hydroxyl group, an amino group, a carboxylate group, a nitro group, a cyano group, an alkoxy group, a hydroxyalkyl group, and a ring.
  • R 3 is selected from a hydrogen atom, an alkyl group optionally substituted by a halogen or a cycloalkyl group, and a cycloalkyl group.
  • the compound of the formula (IA) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound of the formula (II-A):
  • R a and R b are each independently selected from a hydrogen atom, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogen, an amino group, a nitro group, a hydroxyl group, a cyano group, a cycloalkyl group, a heterocyclic group, an aryl group, a heteroaryl group, -OR 3 , -C(O)R 3 , -C(O)OR 3 , -S(O) m R 3 and -NR 4 R 5 , wherein the alkyl group, haloalkyl group, cycloalkyl group, hetero
  • the cyclo, aryl and heteroaryl are optionally selected from alkyl, haloalkyl, halo, amino, nitro, cyano, hydroxy, alkoxy, haloalkoxy, hydroxyalkyl, cycloalkyl, heterocycle. Substituting one or more substituent
  • R a and R b together form a cycloalkyl or heterocyclic group, wherein said cycloalkyl or heterocyclic group is optionally selected from the group consisting of alkyl, haloalkyl, halogen, amino, nitro, cyano, hydroxy Substituted with one or more substituents of alkoxy, haloalkoxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 1 to R 5 , p, m and q are as defined in the formula (IA).
  • the compound of the formula (II-A) or a tautomer, a mesogen, a racemate, an enantiomer or a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof which is selected from the group consisting of the compounds of the formula (II-B):
  • R 1 , R 2 , R 4 , R a , R b , p and q are as defined in the formula (II-A).
  • R a , R b , R 1 , R 2 , R 4 , p and q are as defined in the formula (II-A).
  • the compound of the formula (II-A) or a tautomer, a mesogen, a racemate, an enantiomer or a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound of the formula (IV-A):
  • R 1 , R 2 and p are as defined in the formula (II-A).
  • R 1 , R 2 and p are as defined in the formula (II).
  • Typical compounds of formula (I-A) include, but are not limited to:
  • a tautomer a meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof.
  • the invention further provides a preparation according to the compound of the formula (IA) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer, or a mixture thereof, Or a method thereof, wherein the method comprises:
  • Rings A, R, R 1 , R 2 , p and q are as defined in the formula (IA).
  • the invention further provides a preparation according to the compound of the formula (IA) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer, or a mixture thereof, Or a method thereof, wherein the method comprises:
  • Rings A, R, R 1 , R 2 , p and q are as defined in formula (IA).
  • the invention further provides a preparation according to the compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer, or a mixture thereof, Or a method thereof, wherein the method comprises:
  • Rings A, R, R 1 , R 2 , p and q are as defined in formula (I).
  • Another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the above formula or a tautomer, a mesogen, a racemate, an enantiomer thereof , diastereomers, or mixtures thereof, or pharmaceutically acceptable salts, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present invention also relates to a process for the preparation of the above pharmaceutical composition which comprises the compounds of the formulas or their tautomers, meso, racemates, enantiomers, non-pairs
  • the conjugate, or a mixture thereof, or a pharmaceutically acceptable salt thereof is admixed with a pharmaceutically acceptable carrier, diluent or excipient.
  • the invention further relates to the formulae, in particular the compounds of the formula (I) or the tautomers, meso, racemates, enantiomers, diastereomers or A mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in the manufacture of a medicament for agonizing or antagonizing a MOR receptor.
  • the invention further relates to the formulae, in particular the compounds of the formula (I) or the tautomers, meso, racemates, enantiomers, diastereomers or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a use thereof, for use in the manufacture of a medicament for preventing and/or treating a MOR agonist receptor-mediated related disease, wherein the disease is selected from the group consisting of pain , immune dysfunction, inflammation, esophageal reflux, neurological and psychiatric disorders, urinary and reproductive diseases, cardiovascular diseases and respiratory diseases.
  • the invention further relates to the formulae, in particular the compounds of the formula (I) or the tautomers, meso, racemates, enantiomers, diastereomers or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a medicament comprising the pharmaceutical composition thereof for the preparation of a medicament for preventing or treating pain and pain-related diseases in a mammal, wherein the pain may be postoperative pain, Cancer-induced pain, neuropathic pain, traumatic pain, pain caused by inflammation, etc.; wherein the cancer may be selected from the group consisting of breast cancer, endometrial cancer, cervical cancer, skin cancer, prostate cancer, ovarian cancer, fallopian tube tumor, Ovarian tumors, hemophilia and leukemia.
  • the invention further relates to a method of preventing or treating a MOR agonist receptor mediated related disorder, the method comprising administering to a patient in need thereof a therapeutically effective amount of the formulae of the invention, in particular the formula (I) A compound, or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof.
  • This method shows outstanding efficacy and fewer side effects.
  • the diseases described therein are selected from the group consisting of pain, immune dysfunction, inflammation, esophageal reflux, neurological and psychiatric diseases, urinary and reproductive diseases, cardiovascular diseases and respiratory diseases; preferably pain.
  • Another aspect of the invention relates to a method for preventing or treating pain and pain related diseases in a mammal, the method comprising administering to a patient in need thereof a therapeutically effective amount of the formulas of the invention, in particular the formula (I) a compound or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof; Outstanding efficacy and less side effects, wherein the pain may be pain caused by postoperative pain, cancer-induced pain, neuropathic pain, traumatic pain, and inflammation; wherein the cancer may be selected from breast cancer, Endometrial cancer, cervical cancer, skin cancer, prostate cancer, ovarian cancer, fallopian tube tumor, ovarian tumor, hemophilia and leukemia.
  • the invention relates to the general formula, in particular the compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or
  • the mixture form, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof can also be used for the treatment of immune dysfunction, inflammation, esophageal reflux, nerve and Mental disorders, urinary and reproductive disorders, agents for drug and alcohol abuse, agents for treating gastritis and diarrhea, cardiovascular drugs, and agents for treating respiratory diseases and coughs.
  • the active ingredient-containing pharmaceutical composition may be in a form suitable for oral administration, such as tablets, dragees, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or Tincture.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions, such compositions may contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents, and preservatives, To provide a pleasing and tasty pharmaceutical preparation. Tablets contain the active ingredient and non-toxic pharmaceutically acceptable excipients suitable for the preparation of a tablet for admixture.
  • hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin, or in which the active ingredient is mixed with a water-soluble carrier such as polyethylene glycol or an oil vehicle such as peanut oil, liquid paraffin or olive oil.
  • Soft gelatin capsules provide oral preparations.
  • the aqueous suspension contains the active substance and excipients suitable for the preparation of the aqueous suspension for mixing.
  • the dispersible powders and granules suitable for the preparation of aqueous suspensions can be provided by the addition of water to provide the active ingredient and dispersing or wetting agents, suspending agents or one or more preservatives. Suitable dispersing or wetting agents and suspending agents can be used to illustrate the above examples. Other excipients such as sweetening, flavoring, and coloring agents may also be added. These compositions are preserved by the addition of an anti-oxidant such as ascorbic acid.
  • compositions of the invention may also be in the form of an oil-in-water emulsion.
  • compositions of the invention may be in the form of a sterile injectable aqueous solution.
  • acceptable vehicles or solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • compositions of the invention may be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • the dosage of the drug to be administered depends on a variety of factors including, but not limited to, the following factors: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, and the patient's behavior.
  • the dosage, the patient's diet, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, etc.; in addition, the optimal treatment mode such as the mode of treatment, the daily dosage of the compound of formula (I) or a pharmaceutically acceptable salt The type can be verified according to traditional treatment options.
  • alkyl refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • the alkyl group of the atom is a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 - dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -methylhexyl, 3-methylhexyl, 4-methylhexyl,
  • lower alkyl groups having from 1 to 6 carbon atoms, non-limiting examples including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Base, 2,3-dimethylbutyl and the like.
  • the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups independently selected from the group consisting of an alkane Base, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, naphthenic An oxy group, a heterocycloalkoxy group, a cycloalkylthio group, a heterocycloalkylthio group, an oxo group, a carboxyl group or a carboxylate group.
  • an alkane Base alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, hetero
  • alkenyl refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond.
  • a fork group a vinyl group, a 1-propenyl group, a 2-propenyl group, a 1-, 2- or 3-butenyl group, and the like.
  • the alkenyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkane.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 6 carbon atoms. One carbon atom, most preferably from 5 to 6 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
  • a polycycloalkyl group includes a spiro ring, a fused ring, and a cycloalkyl group.
  • spirocycloalkyl refers to a polycyclic group that shares a carbon atom (referred to as a spiro atom) between 5 to 20 members of a single ring, which may contain one or more double bonds, but none of the rings have a fully conjugated ⁇ electronic system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spirocycloalkyl group is classified into a monospirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a monospirocycloalkyl group and a bispirocycloalkyl group, depending on the number of common spiro atoms between the rings.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to 5 to 20 members, and each ring in the system shares an all-carbon polycyclic group of an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have complete Conjugate ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members. Depending on the number of constituent rings, it may be classified into a bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl group, preferably a bicyclic ring, a tricyclic ring or a tetracyclic ring, and more preferably a bicyclic ring or a tricyclic ring.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the ring to which the parent structure is attached is a cycloalkyl group, non-limiting examples include indanyl, tetrahydronaphthalene A benzocycloheptyl group or the like; preferably a phenylcyclopentyl group or a tetrahydronaphthyl group.
  • the cycloalkyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio group, alkylamino group, halogen, fluorenyl group, hydroxy group, nitro group, cyano group, cycloalkyl group, heterocycloalkyl group, Aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxy or carboxylate.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O).
  • a hetero atom of m (where m is an integer of 0 to 2), but excluding the ring moiety of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • Non-limiting examples of monocyclic heterocyclic groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiophenyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, Hydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl and the like are preferably tetrahydropyranyl, piperidinyl or pyrrolidinyl.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group in which one atom (called a spiro atom) is shared between 5 to 20 members of a single ring, wherein one or more ring atoms are selected from nitrogen, oxygen or S (O). ) m (where m is an integer 0 to 2) heteroatoms, and the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spiroheterocyclyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of shared spiro atoms between the ring and the ring, and is preferably a monospiroheterocyclic group and a dispiroheterocyclic group. More preferably, it is 4 yuan / 4 yuan, 4 yuan / 5 yuan, 4 yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6-membered monospiroheterocyclic group.
  • Non-limiting examples of spiroheterocyclyl groups include:
  • fused heterocyclyl refers to 5 to 20 members, and each ring in the system shares an adjacent pair of atomic polycyclic heterocyclic groups with other rings in the system, and one or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining rings
  • the atom is carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused heterocyclic groups include:
  • bridge heterocyclyl refers to a polycyclic heterocyclic group of 5 to 14 members, any two rings sharing two atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have a total A ⁇ -electron system of a yoke in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), the remaining ring atoms being carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • bridge heterocyclic groups include:
  • the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heterocyclic group, non-limiting examples of which include:
  • the heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, an oxo group, a carboxyl group or a carboxylate group.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic ring (i.e., a ring sharing a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably 6 to 10 members, more preferably 5 to 6 yuan, such as phenyl and naphthyl.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples of which include:
  • the aryl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle An alkylthio group, a carboxyl group or a carboxylate group.
  • heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 members, and has 1 to 3 hetero atoms; more preferably 5 or 6 members, and 1 to 2 hetero atoms; preferably, for example, imidazolyl, furyl, thienyl, thiazolyl, pyridyl Azolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc., preferably imidazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more pyrazolyl .
  • the heteroaryl ring may be fused to an aryl, heterocyclic or cycloalkyl ring, wherein the
  • the heteroaryl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, a carboxyl group or a carboxylate group.
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, a carboxyl group or a carboxylate group.
  • haloalkyl refers to an alkyl group substituted with a deuterium atom. Wherein alkyl is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
  • hydroxy refers to an -OH group.
  • halogen means fluoro, chloro, bromo or iodo.
  • amino means -NH 2.
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • carboxylate group refers to -C(O)O(alkyl) or -C(O)O(cycloalkyl), wherein alkyl, cycloalkyl are as defined above.
  • acyl halide refers to a compound containing a -C(O)-halogen group.
  • X is selected from A, B, or C
  • X is selected from A, B, and C
  • X is A, B, or C
  • X is A, B, and C
  • X is A, B, and C
  • Optional or “optionally” means that the subsequently described event or environment may, but need not, occur, including where the event or environment occurs or does not occur.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
  • Substituted refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiological/pharmaceutically acceptable carriers. And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the invention which is safe and effective for use in a mammal and which possesses the desired biological activity.
  • Rings A, R, R 1 , R 2 , p and q are as defined in formula (IA).
  • the compound of the formula (I-A) of the present invention can also be produced as follows:
  • Rings A, R, R 1 , R 2 , p and q are as defined in formula (IA).
  • Rings A, R, R 1 , R 2 , p and q are as defined in formula (I).
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • NMR was measured using a Bruker AVANCE-400 nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), internal standard was four.
  • DMSO-d 6 dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS Methyl silane
  • the measurement of the MS was carried out using a FINNIGAN LCQAd (ESI) mass spectrometer (manufacturer: Thermo, model: Finnigan LCQ advantage MAX).
  • ESI FINNIGAN LCQAd
  • the HPLC was measured using an Agilent 1200 DAD high pressure liquid chromatograph (Sunfire C18 150 x 4.6 mm column) and a Waters 2695-2996 high pressure liquid chromatograph (Gimini C18 150 x 4.6 mm column).
  • Thin layer chromatography silica gel plate uses Yantai Yellow Sea HSGF254 or Qingdao GF254 silica gel plate.
  • the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm.
  • the specification for thin layer chromatography separation and purification is 0.4mm. ⁇ 0.5mm.
  • the known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Companies such as Dare Chemicals.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the pressurized hydrogenation reaction was carried out using a Parr Model 3916EKX hydrogenation apparatus and a clear blue QL-500 type hydrogen generator or a HC2-SS type hydrogenation apparatus.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • the microwave reaction used a CEM Discover-S Model 908860 microwave reactor.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature and is 20 ° C to 30 ° C.
  • the progress of the reaction in the examples was monitored by thin layer chromatography (TLC).
  • TLC thin layer chromatography
  • the system used for the reaction was: A: dichloromethane and methanol system, B: n-hexane and ethyl acetate system, C: dichloro
  • the volume ratio of the solvent is adjusted depending on the polarity of the compound.
  • Purification compounds using column chromatography eluent systems and thin layer chromatography developer systems include: A: dichloromethane and methanol systems, B: n-hexane and ethyl acetate systems, C: dichloromethane and acetone
  • A dichloromethane and methanol systems
  • B n-hexane and ethyl acetate systems
  • C dichloromethane and acetone
  • the volume ratio of the solvent is adjusted depending on the polarity of the compound, and a small amount of an alkaline or acidic reagent such as triethylamine or acetic acid may be added for adjustment.
  • the reaction mixture was washed with EtOAcqqqqqqHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHH
  • the crude compound 14b (30 mg, 0.11 mmol) was dissolved in 0.5 mL of dichloromethane, and 1 mL of 4M hydrogen chloride in 1,4-dioxane solution was added, and the reaction was stirred for 2.5 hours. The reaction mixture was concentrated to dryness crystals crystals crystals crystals crystals
  • the crude compound 19a (698 mg, 2.4 mmol) was dissolved in 4 mL of dichloromethane, and 8 mL of 4M hydrogen chloride in 1,4-dioxane solution was added, and the reaction was stirred for 2 hours.
  • Methyltriphenylphosphonium bromide (2.95 g, 11.5 mmol) was dissolved in 20 mL of tetrahydrofuran, cooled to 0 ° C, potassium tert-butoxide (1.29 g, 11.5 mmol) was added, the reaction was stirred for 30 minutes, and 11b (1 g, 7.66 mmol), warmed to room temperature and stirred for 12 hours. The reaction mixture was concentrated under reduced pressure.
  • the crude product 22c (20 mg, 0.07 mmol) was dissolved in 10 mL of dichloromethane, and 10 mL of 4M hydrogen chloride in 1,4-dioxane solution was added, and the reaction was stirred for 2 hours. The reaction mixture was concentrated to dryness crystals crystals crystals crystals crystals
  • the reaction was quenched by adding 100 mL of methanol at 15 ° C for 1 hour, concentrated under reduced pressure, and added with 200 mL of ethyl acetate and 5 g of activated carbon, and stirred for 30 minutes, filtered, and washed with ethyl acetate (100 mL ⁇ 3).
  • the title product 25a (10.5 g, colorless oil) was obtained.
  • reaction solution was poured into a mixed solvent of 50 mL of water, 50 mL of n-hexane and 5 mL of ethyl acetate, and stirred for 10 minutes, and filtered to remove insoluble materials.
  • the crude product 29a (50 mg, 0.1 mmol) was dissolved in 10 mL of dichloromethane, and 0.1 mL of 4M hydrogen chloride in dioxane solution was added, and the reaction was stirred for 0.5 hour. The reaction mixture was added with aq. EtOAc EtOAc (EtOAc m.
  • Diethyl cyanomethylphosphonate (200 mg, 0.76 mmol) was dissolved in 20 mL of tetrahydrofuran, and a sodium hydrogen hydride (61 mg, 1.52 mmol) was added, and the mixture was stirred for 30 minutes under ice-water bath, and the pre-formed 11b (200 mg) was added dropwise. A solution of 0.76 mmol of tetrahydrofuran was stirred at 25 ° C for 16 hours. The reaction mixture was poured into ice water, and the mixture was evaporated, evaporated, evaporated, evaporated, evaporated, 150 mg, colorless viscous), yield 69%.
  • the compounds of the invention can activate the mu-opioid receptor (MOR).
  • MOR mu-opioid receptor
  • Activated MOR regulates the level of intracellular cAMP, and cAMP enters the nucleus to bind to the CRE region of the reporter luciferase to initiate expression of the reporter gene.
  • the luciferase reacts with its substrate to fluoresce and reflects the agonistic activity of the compound by measuring the fluorescent signal.
  • MOR/pcDNA3.1(+) and CRE/pGL4.29 were transferred to HEK293 cell line, and G418 and Hygromycin were added to the medium, and HEK293/MOR/ was screened in a 96-well cell culture plate. CRE monoclonal cell line.
  • HEK293/MOR/CRE monoclonal cell lines were cultured in DMEM/high glucose medium (10% FBS, 1 mg/ml G418, 200 ug/ml hygromycin, mixed) and passaged every 3 days.
  • DMEM/high glucose medium 10% FBS, 1 mg/ml G418, 200 ug/ml hygromycin, mixed
  • a cell suspension was prepared from fresh cell culture medium, and 20,000 cells/well was plated in a 96-well plate (BD, #356692), and cultured at 5% carbon dioxide at 37 °C.
  • the compound was dissolved in pure DMSO at a concentration of 20 mM, and then DMSO was used to prepare the first concentration of 4 mM, and diluted to 10 concentrations in 10 folds.
  • the wells were set to blank and control wells and added to 90 ⁇ l of DMSO; 2.5 ⁇ l of the compound solution prepared in DMSO in a gradient concentration was added to 97.5 ⁇ l of fresh cell culture medium containing 5 ⁇ M foscolin; 10 ⁇ l of the prepared compound was added to the cell culture plate to make the final concentration of the compound 10000, 1000, 100, 10, 1, 0.1, 0.01 nM, 5% carbon dioxide was incubated at 37 ° C for 5 hours.
  • the compounds of the present invention agonistically affect the change of downstream cAMP levels by the above test, the measured EC 50 values are shown in Table 1.1, and Emax is the maximum effect of the example compound activating MOR on the cAMP signaling pathway (maximum effect of TRV-130) 100%).
  • HEK293/KOR/CRE or HEK293/DOR/CRE (CRE cDNA purchased from Promega, Cat. No. E8471, KOR and DOR cDNA was constructed) was seeded at a density of 1 ⁇ 10 4 cells/well. Incubate overnight at 37 ° C, 5% CO 2 .
  • the drug was configured as a 20 mM stock solution, diluted to a 200x concentration gradient with a 100% DMSO gradient, and diluted 20-fold with DMEM/high glucose (SH30243.01B, Hyclone) medium.
  • the cell culture plates inoculated on the first day were taken out, and 10 ⁇ l of the diluted drug or control (0.5% DMSO) was added to each well, and the mixture was gently shaken, placed in a 37 ° C, 5% CO 2 incubator for 4 hours, and finally added for detection.
  • the reagent ONE-Glo (E6120, Promega) was 100 ⁇ l/well, and allowed to stand at room temperature for 5 minutes, and the absorption value was measured by a cold light mode of a microplate reader (PE, Victor 3).
  • EC 50 values of the compound calculated using Graphpad Prism software corresponding to each concentration of compound according to the signal value. Emax is the maximum effect of a compound causing a change in cAMP levels.
  • the change in cAMP level downstream of the compound agonistic KOR receptor or DOR receptor in the present invention is determined by the above test, and the measured EC 50 value is shown in Table 1.2, and Emax is the maximum effect of the compound causing the change of cAMP level (the maximum effect of morphine is 100%).
  • the activity of the compounds of the invention to activate the ⁇ -arrestin signaling pathway of the MOR receptor was determined.
  • CHO-K1OPRM1 ⁇ -Arrestin 93-0213C2, DiscoveRX
  • 93-0213C2 DiscoveRX cells were seeded in a 96-well plate, seeded at a density of 1 ⁇ 10 4 cells/well, and the cells were cultured overnight at 37° C. under 5% CO 2 .
  • the activity of the compounds of the present invention to activate the ⁇ -arrestin signaling pathway was determined by the above assay.
  • the measured EC 50 values are shown in Table 2.
  • Emax is the maximum effect of the compound causing the ⁇ -arrestin signaling pathway (the maximum effect of morphine is 100%).
  • Table 2 EC 50 of the compounds of the invention activating the activity of the ⁇ -arrestin signaling pathway
  • the compounds of the invention have little activation effect on the ⁇ -arrestin signaling pathway. .
  • the compound of the present invention and the positive compound TRV-130 (Journal of Pharmacology and Experimental Therapeutics, Volume 344, Issue 3, Pages 708-717, 2013) were tested on hERG using a fully automated patch clamp on a stable cell line transfected with hERG potassium channels. Blocking effect of potassium current.
  • the HEK293-hERG stable cell line was passaged at a density of 1:4 in MEM/EBSS medium (10% FBS, 400 ⁇ g/ml G418, 1% MEM non-essential amino acid solution (100 ⁇ ), 1% sodium pyruvate solution). Culture, culture within 48-72 hours for automated patch-clamp experiments.
  • the cells were digested with 0.25% trypsin, and the cells were collected by centrifugation and resuspended with extracellular fluid (140 mM NaCl, 4 mM KCl, 1 mM MgCl 2 , 2 mM CaCl 2 , 5 mM MD-water glucose, 10 mM Hepes, pH 7.4, 298 mOsmol).
  • extracellular fluid 140 mM NaCl, 4 mM KCl, 1 mM MgCl 2 , 2 mM CaCl 2 , 5 mM MD-water glucose, 10 mM Hepes, pH 7.4, 298 mOsmol.
  • the cells are made into a cell suspension.
  • the cell suspension was placed on the cell bank of the Patchliner instrument.
  • the Patchliner instrument used a negative pressure controller to apply the cells to the chip (NPC-16), and the negative pressure attracted individual cells to the wells of the chip.
  • the instrument When the whole cell mode is formed, the instrument will get the hERG current according to the set hERG current voltage program, and then the instrument will automatically perfuse the compound from low to high concentration.
  • the currents at each concentration of the compounds and the blank control current were analyzed by HEAK Patchmaster, HEAKEPC10 patch clamp amplifiers (Nanion) and Pathlinersoftware and data analysis software provided by Pathcontrol HTsoftware.
  • the compounds of the present invention have a weaker inhibitory effect on hERG than the positive compounds, and there are significant differences.

Landscapes

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

Abstract

本发明涉及氧杂螺环类衍生物、其制备方法及其在医药上的应用。特别地,本发明涉及通式(I)所示的氧杂螺环类衍生物、其制备方法及含有该衍生物的药物组合物,以及其作为MOR受体激动剂的用途,和其在制备治疗和/或预防疼痛和疼痛相关疾病的药物中的用途。其中通式(I)的各取代基同说明书中的定义相同。

Description

氧杂螺环类衍生物、其制备方法及其在医药上的应用 技术领域
本发明属于医药领域,涉及一种氧杂螺环类衍生物、其制备方法及其在医药上的应用。特别地,本发明涉及通式(I)所示的氧杂螺环类衍生物、其制备方法及含有该衍生物的药物组合物,以及其作为MOR受体激动剂的用途和其在制备治疗和/或预防疼痛和疼痛相关疾病的药物中的用途。
背景技术
阿片受体是一类重要的G蛋白偶联受体(G protein coupled receptor,GPCR),是内源性阿片肽及阿片类药物结合的靶点,阿片受体激活后对神经系统免疫及内分泌系统具有调节作用,阿片类药物是目前最强且常用的中枢镇痛药。内源性阿片肽是哺乳动物体内天然生成的阿片样活性物质,目前已知的内源性阿片肽大致分为脑啡肽、内啡肽、强啡肽和新啡肽几类(Pharmacol Rev 2007;59:88–123)。中枢神经系统中存在其相应的阿片受体,即μ(MOR)、δ(DOR)、κ(KOR)受体等。研究发现,内源性阿片肽镇痛作用的强弱,主要取决于阿片受体表达的多少,阿片受体是阿片类药物以及内源性阿片肽镇痛作用的靶点。Zadina等研究发现MOR受体与吗啡肽1的结合能力最强(360pM),是DOR受体与吗啡肽1结合力的4000倍,KOR受体与吗啡肽1结合力的15000倍,是最重要的介导镇痛作用的阿片受体(science,2001,293:311-315;Biochem Biophys Res Commun 235:567–570;Life Sci 61:PL409-PL415)。
目前的研究认为,GPCR介导及调控生理功能,主要经由两条途径:G蛋白途径和β-arrestin途径。传统的GPCR激动剂与受体结合后,激活G蛋白信号途径,包括钙离子等第二信使系统、腺苷酸环化酶(adenyl cyclase,AC)、丝裂原活化蛋白激酶(mitogen-activated protein kinases,MAPK)等,而β-arrestin偏爱性配体则主要激活β-arrestin途径。而β-arrestin介导的GPCR反应主要包括3个方面:1)作为负性调控因子,与G蛋白偶联受体激酶(GRK)作用,使GPCRs发生受体脱敏反应,中止G蛋白信号转导;2)作为支架蛋白(scaffold protein),募集胞吞蛋白,诱导GPCR内吞;3)作为接头蛋白,与GPCR下游信号分子形成复合物,以G蛋白非依赖的方式激活信号转导分子,如MAPK、Src蛋白酪氨酸激酶和Akt等。配体刺激G蛋白信号和/或β-arrestin信号的差异,最终决定了GPCR的配体特异性细胞生物学效应。
MOR是内源性脑啡肽和吗啡等阿片类镇痛药物的作用靶点。早期研究显示,内源性脑啡肽和阿类药物埃托啡可以激动G蛋白并引发受体内吞,但是吗啡却完全不会引发受体的内吞,这是因为吗啡激动MOR磷酸化的能力太弱,仅能募集微量的β-arrestin于膜上(Zhang等,Proc Natl Acad Sci USA,1998,95(12):7157-7162)。 此类配体完全通过G蛋白信号通路而不是β-arrestin途径来发挥其生理功能。研究发现给β-arrestin2基因敲除小鼠注射吗啡后,由G蛋白信号介导的镇痛效果更强且维持时间更长(Bohn等,Science,1999年)。可以预见,如果此类配体的负性β-arrestin偏爱性更强,甚至可以逃脱β-arrestin介导的受体脱敏,则可导致G蛋白信号传递时间延长,产生更强大的镇痛作用。
目前公开的MOR激动剂专利申请包括WO2014022733、WO2008009415、WO2009018169、WO2012129495、WO2001049650、WO2002020481、WO2010051476和WO2013087589等。
阿片类药物长期使用会产生耐受以及呼吸抑制、便秘等副作用,而这些副作用被证明与β-arrestin的功能密切相关。为了减小阿片类药物的副作用,可基于MOR的负性β-arrestin偏爱性配体设计药物,使β-arrestin介导的副作用降低,增强治疗效果,对于本发明的氧杂螺环类衍生物在作为MOR选择性药物的研究中,Trevena Inc公司研究发现芳基苄位取代时活性较差(J.Med.Chem.2013,56,8019-8031),但本发明在进行一系列研究后发现,芳基苄位成环后却表现出高活性,Emax也显著提高,hERG也有明显改善,更进一步的研究发现单一构型的化合物对MOR的选择性更高。
发明内容
本发明的目的在于提供一种通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐:
Figure PCTCN2016101064-appb-000001
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐,
其中:
环A选自环烷基和杂环基;
R选自芳基和杂芳基,其中所述的芳基和杂芳基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、-S(O)mR3和-NR4R5中的一个或多个取代基所取代;
R1相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤素、氨基、硝基、羟基、氰基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、 -S(O)mR3和-NR4R5,其中所述的烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R2相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤素、氨基、硝基、羟基、氰基、氧代基、烯基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、-S(O)mR3和-NR4R5,其中所述的烷基、烷氧基、烯基、卤代烷基、环烷基、杂环基、芳基和杂芳基任选被选自氘原子、烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
或者两个R2一起形成环烷基基或杂环基,其中所述的环烷基或杂环基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R3选自氢原子、烷基、氘代烷基、氨基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤素、羟基、氨基、硝基、氰基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R4和R5各自独立地选自氢原子、烷基、烷氧基、羟烷基、羟基、氨基、羧酸酯基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤素、羟基、氨基、羧酸酯基、硝基、氰基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
p、q各自独立地为0、1、2、3或4;且
m为0、1或2。
在本发明一个优选的实施方案中,通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其为通式(I)所示的化合物:
Figure PCTCN2016101064-appb-000002
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
环A、R、R1、R2、p和q如通式(I-A)中所定义。
在本发明一个优选的实施方案中,通式(I)或通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其中A环选自5~6元杂环基和5~6元环烷基。
在本发明一个优选的实施方案中,通式(I))或通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其中R为吡啶基。
在本发明一个优选的实施方案中,通式(I))或通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其中R1相同或不同,且各自独立地选自氢原子和卤素。
在本发明一个优选的实施方案中,通式(I))或通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其中R2相同或不同,且各自独立地选自氢原子、烷基、氧代基、烷氧基、羟基、卤素和-OR3,其中所述的烷基和烷氧基任选被选自氘原子、烷基、卤素、羟基、氨基、羧酸酯基、硝基、氰基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;R3选自氢原子、任选被卤素或环烷基取代的烷基和环烷基。
在本发明一个优选的实施方案中,通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其为通式(II-A)所示的化合物:
Figure PCTCN2016101064-appb-000003
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
G选自一个键、CRaRb、C=O、NR4和氧原子;
Ra和Rb各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤素、氨基、硝基、羟基、氰基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、 -S(O)mR3和-NR4R5,其中所述的烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代,优选羟基或-OR3
或者Ra和Rb一起形成环烷基基或杂环基,其中所述的环烷基或杂环基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R1~R5、p、m和q如通式(I-A)中所定义。
在本发明一个优选的实施方案中,通式(II-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其选自通式(II-B)所示的化合物:
Figure PCTCN2016101064-appb-000004
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
G选自一个键、CRaRb、C=O、NR4和氧原子;
R1、R2、R4、Ra、Rb、p和q通式(II-A)中所定义。
在本发明一个优选的实施方案中,通式(Ⅱ-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其选自通式(II)所示的化合物:
Figure PCTCN2016101064-appb-000005
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
G选自一个键、CRaRb、C=O、NR4和氧原子;
Ra、Rb、R1、R2、R4、p和q如通式(II-A)中所定义。
在本发明一个优选的实施方案中,通式(II-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其为通式(IV-A)所示的化合物:
Figure PCTCN2016101064-appb-000006
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
R1、R2和p如通式(II-A)中所定义。
在本发明一个优选的实施方案中,通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,其选自通式(IV)所示的化合物:
Figure PCTCN2016101064-appb-000007
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
R1、R2和p如通式(II)中所定义。
通式(I-A)的典型化合物,包括但不限于:
Figure PCTCN2016101064-appb-000008
Figure PCTCN2016101064-appb-000009
Figure PCTCN2016101064-appb-000010
Figure PCTCN2016101064-appb-000011
Figure PCTCN2016101064-appb-000012
Figure PCTCN2016101064-appb-000013
Figure PCTCN2016101064-appb-000014
Figure PCTCN2016101064-appb-000015
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐。
本发明另外提供一种制备根据通式(I-A)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2016101064-appb-000016
通式(VA)化合物或其盐酸盐和通式(VIA)化合物发生还原胺化反应,得到通式(I-A)化合物;
其中:
环A、R、R1、R2、p和q如权通式(I-A)中所定义。
本发明另外提供一种制备根据通式(I-A)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2016101064-appb-000017
通式(VB-A)化合物和通式(VIB-A)化合物或其盐酸盐发生还原胺化反应,得到通式(I-A)化合物;
其中:
环A、R、R1、R2、p和q如通式(I-A)中所定义。
本发明另外提供一种制备根据通式(I)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2016101064-appb-000018
通式(VB)化合物和通式(VIB)化合物或其盐酸盐发生还原胺化反应,得到通式(I)化合物;
其中:
环A、R、R1、R2、p和q如通式(I)中所定义。
本发明的另一方面涉及一种药物组合物,其含有治疗有效剂量的上述各通式所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。本发明还涉及一种制备上述药物组合物的方法,其包括将各通式所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐与药学上可接受的载体、稀释剂或赋形剂相混合。
本发明进一步涉及各通式,特别是通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其药物组合物在制备激动或拮抗MOR受体的药物中用途。
本发明进一步涉及各通式,特别是通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其药物组合物在制备用于预防和/或治疗MOR激动剂受体介导的相关疾病的药物中的用途,其中所述的疾病选自疼痛、免疫功能障碍、炎症、食管回流、神经和精神疾病、泌尿和生殖疾病、心血管疾病和呼吸道疾病。
本发明进一步涉及各通式,特别是通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其药物组合物在制备用于预防或治疗哺乳动物体内的疼痛和疼痛相关疾病的药物中的用途,其中所述的疼痛可以是术后疼痛、癌症引起的疼痛、神经性疼痛、创伤性疼痛和炎症引起的疼痛等;其中所述的癌症可以选自乳腺癌、子宫内膜癌、宫颈癌、皮肤癌、前列腺癌、卵巢癌、输卵管肿瘤、卵巢瘤、血友病和白血病等。
本发明进一步涉及一种预防或治疗MOR激动剂受体介导的相关疾病的方法,该方法包括向需要其的患者施用治疗有效剂量的本发明的各通式,特别是通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐。该方法表现出突出的疗效和较少的副作用。其中所述的疾病选自疼痛、免疫功能障碍、炎症、食管回流、神经和精神疾病、泌尿和生殖疾病、心血管疾病和呼吸道疾病;优选疼痛。
本发明另一方面涉及一种作为预防或治疗哺乳动物体内的疼痛和疼痛相关疾病的方法,该方法包括向需要其的患者施用治疗有效剂量的本发明的各通式,特别是通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐;该方法表现出突出的疗效和较少的副作用,其中所述的疼痛可以是术后疼痛、癌症引起的疼痛、神经性疼痛、创伤性疼痛和炎症引起的疼痛;其中所述的癌症可以选自乳腺癌、子宫内膜癌、宫颈癌、皮肤癌、前列腺癌、卵巢癌、输卵管肿瘤、卵巢瘤、血友病和白血病等。
本发明涉及的各通式,特别是通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其药物组合物,还可以用于治疗免疫功能障碍、炎症、食道回流、神经和 精神病症、泌尿和生殖病症、用于药物和酒精滥用的药剂、用于治疗胃炎和腹泻的药剂、心血管药物和用于治疗呼吸道疾病和咳嗽的药剂等。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。
也可用其中活性成分与惰性固体稀释剂例如碳酸钙、磷酸钙或高岭土混合的硬明胶胶囊,或其中活性成分与水溶性载体例如聚乙二醇或油溶媒例如花生油、液体石蜡或橄榄油混合的软明胶胶囊提供口服制剂。
水悬浮液含有活性物质和用于混合的适宜制备水悬浮液的赋形剂。
通过加入水可使适用于制备水混悬也的可分散粉末和颗粒提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂可说明上述的例子。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
本发明的药物组合物也可以是水包油乳剂的形式。
本发明的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。
本发明的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行被、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合等;另外,最佳的治疗方式如治疗的模式、通式化合物(I)的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“烯基”指由至少两个碳原子和至少一个碳-碳双键组成的如上述定义的烷基。例如叉基、乙烯基、1-丙烯基、2-丙烯基、1-,2-或3-丁烯基等。烯基可以是取代的或未取代的,当被取代时,取代基优选为一个或多个以下基团,独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至6个碳原子,最优选包含5至6个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2016101064-appb-000019
术语“稠环烷基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2016101064-appb-000020
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2016101064-appb-000021
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等;优选苯基并环戊基、四氢萘基。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、 芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;最优选包含3至8个环原子,其中1~3个是杂原子;最优选包含5至6个环原子,其中1~2或1~3个是杂原子。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢吡喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等,优选四氢吡喃基、哌啶基、吡咯烷基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2016101064-appb-000022
术语“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2016101064-appb-000023
Figure PCTCN2016101064-appb-000024
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2016101064-appb-000025
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2016101064-appb-000026
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,更优选5至6元,例如苯基和萘基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2016101064-appb-000027
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至2个杂原子;优选例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等,优选为咪唑基、吡唑基或嘧啶基、噻唑基;更有选吡唑基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2016101064-appb-000028
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“氘代烷基”指被氘原子取代的烷基。其中烷基如上所定义。
术语“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH2
术语“氰基”指-CN。
术语“硝基”指-NO2
术语“羧基”指-C(O)OH。
术语“羧酸酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基、环烷基如上所定义。
术语“酰卤”指含有-C(O)-卤素的基团的化合物。
“X选自A、B、或C”、“X选自A、B和C”、“X为A、B或C”、“X为A、B和C”等不同用语均表达了相同的意义,即表示X可以是A、B、C中任意一种或几种。“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
本发明化合物的合成方法
为了完成本发明的目的,本发明采用如下技术方案:
本发明通式(I-A)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
方案一
Figure PCTCN2016101064-appb-000029
通式(VA)化合物或其盐酸盐和通式(VIA)化合物发生还原胺化反应,得到通式(I-A)化合物;
其中:
环A、R、R1、R2、p和q如通式(I-A)中所定义。
本发明通式(I-A)所示的化合物还可以如下制备:
方案二
Figure PCTCN2016101064-appb-000030
通式(VB-A)化合物和通式(VIB-A)化合物或其盐酸盐发生还原胺化反应,得到通式(I-A)化合物;
其中:
环A、R、R1、R2、p和q如通式(I-A)中所定义。
本发明通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
方案一
Figure PCTCN2016101064-appb-000031
通式(VB)化合物和通式(VIB)化合物或其盐酸盐发生还原胺化反应,得到通式(I)化合物;
其中:
环A、R、R1、R2、p和q如通式(I)中所定义。
具体实施方式
以下结合实施例进一步描述本发明,但这些实施例并非限制着本发明的范围。
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。
MS的测定用FINNIGAN LCQAd(ESI)质谱仪(生产商:Thermo,型号:FinniganLCQ advantage MAX)。
HPLC的测定使用安捷伦1200DAD高压液相色谱仪(Sunfire C18 150×4.6mm色谱柱)和Waters 2695-2996高压液相色谱仪(Gimini C18 150×4.6mm色谱柱)。
激酶平均抑制率及IC50值的测定用NovoStar酶标仪(德国BMG公司)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
柱层析一般使用烟台黄海硅胶200~300目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH & Co.KG,Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)、达瑞化学品等公司。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂的体系有:A:二氯甲烷和甲醇体系,B:正己烷和乙酸乙酯体系,C:二氯甲烷和丙酮体系,溶剂的体积比根据化合物的极性不同而进行调节。
纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷和甲醇体系,B:正己烷和乙酸乙酯体系,C:二氯甲烷和丙酮体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1,2
(S)-1-乙基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢喹啉-1-胺1
(R)-1-乙基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢喹啉-1-胺2
Figure PCTCN2016101064-appb-000032
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4,5]癸烷-9-基)乙醛1a(294mg,1.135mmol,采用专利申请“WO2012129495”公开的方法制备而得)和1-乙基-1,2,3,4-四氢喹啉-4-胺1b(200mg,1.135mmol,采用专利申请“WO2014078454”公开的方法制备而)溶解于15mL二氯甲烷中,搅拌反应1小时,加入三乙酰氧基硼氢化钠(1.203g,5.675mmol),搅拌反应16小时。加入20mL水,用二氯甲烷萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相色谱法纯化所得残余物,得到标题产物1-乙基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢喹啉-1-胺,进行手性制备(分离条件:手性制备柱Superchiral S-AD(Chiralway),2cm I.D.*25cm,5um;流动相:CO2:甲醇:二乙醇胺=75:25:0.05,流速:50g/min),收集其相应组分,减压浓缩,得到标题产物1(98mg,棕色油状物)和2(95mg,黄色固体)。
实施例1:
MS m/z(ESI):420.3[M+1];
手性HPLC分析:保留时间4.028分钟,手性纯度:99.7%(色谱柱:Superchiral S-AD(Chiralway),0.46cm I.D.*15cm,5um;流动相:CO2:甲醇:二乙醇胺=75:25:0.05(v/v/v))
1H NMR(400MHz,DMSO-d6)δ8.54(s,1H),7.72(s,1H),7.45(d,1H),7.20(s,1H),6.95(s,1H),6.78(d,1H),6.52(d,1H),6.37(s,1H),3.60(br,2H),3.18-3.43(m,3H),2.99(m,1H),2.33-2.45(m,3H),1.77-1.99(m,3H),1.19-1.60(m,12H),1.00-1.06(m,4H),0.63(m,1H).
实施例2:
MS m/z(ESI):420.3[M+1];
手性HPLC分析:保留时间3.725分钟,手性纯度:99.8%(色谱柱:Superchiral S-AD(Chiralway),0.46cm I.D.*15cm,5um;流动相:CO2:甲醇:二乙醇胺=75:25:0.05(v/v/v))
1H NMR(400MHz,DMSO-d6)δ8.53(s,1H),7.72(s,1H),7.46(d,1H),7.20(s,1H),6.97(s,1H),6.85(d,1H),6.54(d,1H),6.40(s,1H),3.61(br,2H),3.17-3.25(m,3H),3.00-3.01(m,1H),2.33-2.46(m,3H),1.78-1.97(m,3H),1.24-1.65(m,12H),1.01-1.06(m,4H),0.61(m,1H).
实施例3
(1R,2R)-1-((2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-2,3-二氢-1H-茚-2-醇3
Figure PCTCN2016101064-appb-000033
将1a(50mg,0.193mmol),(1R,2R)-1-氨基-2,3-二氢-1H-茚-2-醇3a(31.6mg,0.212mmol,采用专利申请“WO2010148191”公开的方法制备而得)溶解于15mL二氯甲烷中,加入适量甲醇助溶,室温搅拌1小时,加入三乙酰氧基硼氢化钠(200mg,0.965mmol),搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物3(50mg,白色固体),产率:66%。
MS m/z(ESI):393.5[M+1]
1H NMR(400MHz,DMSO-d6)δ8.51(d,1H),7.73-7.66(m,1H),7.37(d,1H),7.28-7.20(m,3H),7.19-7.12(m,2H),4.75(d,1H),4.61(d,1H),3.82-3.71(m,4H),3.41-3.31(m,2H),2.30-2.89(m,2H),2.41-2.25(m,2H),1.96-1.90(m,2H),1.85-1.61(m,4H),1.61-1.25(m,6H).
实施例4
(1R,2R)-2-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-2,3-二氢-1H-茚-1-胺4
Figure PCTCN2016101064-appb-000034
第一步
((1R,2R)-2-甲氧基-2,3-二氢-1H-茚-1-基)氨基甲酸叔丁酯4b
将((1R,2R)-2-羟基-2,3-二氢-1H-茚-1-基)氨基甲酸叔丁酯4a(350mg,1.34mmol,采用公知的方法“Angewandte Chemie-International Edition,2012,51(34),8495-8499”制备而得)溶于15mL二氯甲烷中,加入氧化银(930mg,4.02mmol),碘甲烷(0.25mL,4.02mmol)以及少量活化的
Figure PCTCN2016101064-appb-000035
分子筛,室温搅拌反应16小时。过滤,滤液减压浓缩,用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物4b(200mg,白色固体),产率:57%。
MS m/z(ESI):208.2[M-56+1]
第二步
(1R,2R)-2-甲氧基-2,3-二氢-1H-茚-1-胺2,2,2-三氟乙酸盐4c
将4b(60mg,0.228mmol)溶解于5mL二氯甲烷中,加入0.5mL三氟乙酸,搅拌反应2小时。反应液减压浓缩,得到粗品标题产物4c(66mg,黄色油状物)。产物不经纯化直接下一步反应。
MS m/z(ESI):164.2[M+1]
第三步
(1R,2R)-2-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-2,3-二氢-1H-茚-1-胺4
将1a(50mg,0.193mmol),粗品4c(66mg,0.228mmol)溶解于15mL二氯甲烷中,室温搅拌反应30分钟,加入三乙酰氧基硼氢化钠(200mg,0.965mmol),搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物4(25mg,浅黄色油状物),产率:32%。
MS m/z(ESI):407.3[M+1]
1H NMR(400MHz,DMSO-d6)δ8.55(d,1H),7.71(d,1H),7.58(d,1H),7.40(d,1H),7.28(d,1H),7.25-7.10(m,3H),4.39(d,1H),4.26(d,1H),3.82-3.70(m,5H),3.30(s, 3H),2.88-2.30(m,2H),2.40-2.26(m,2H),1.96-1.91(m,2H),1.85-1.62(m,4H),1.61-1.24(m,6H).
实施例5
N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-苯并二氢吡喃-4-胺
Figure PCTCN2016101064-appb-000036
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4,5]癸烷-9-基)乙醛5a(20mg,0.08mmol,采用专利申请“WO2012129495”公开的方法制备而得),苯并二氢吡喃-4-胺5b(23mg,0.15mmol,采用“Bioorganic&Medicinal Chemistry Letters,2011,21(5),1338-1341”公开的方法制备而得)溶解于10mL二氯甲烷中,搅拌2小时,加入三乙酰氧基硼氢化钠(65mg,0.31mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物5(6mg,黄色油状物),产率:20%。
MS m/z(ESI):393.5[M+1]
1H NMR(400MHz,Methanol-d4)δ8.55(s,1H),7.78(t,1H),7.52(d,1H),7.27(d,1H),7.01-7.12(m,2H),6.66-6.85(m,2H),4.05-4.23(m,2H),3.71-3.86(m,2H),3.59-3.69(m,1H),2.51-2.65(m,2H),2.37-2.47(m,1H),1.98-2.17(m,2H),1.84-1.96(m,2H),1.37-1.83(m,9H),1.24-1.35(m,1H),1.05-1.17(m,1H),0.65-0.71(m,1H).
实施例6
(S)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-苯并二氢吡喃-4-胺
Figure PCTCN2016101064-appb-000037
Figure PCTCN2016101064-appb-000038
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(80mg,0.31mmol),S-苯并二氢吡喃-4-胺盐酸盐6a(86mg,0.46mmol,采用“ACS Catalysis,3(4),555-559;2013”公开的方法制备而得)溶解于10mL二氯甲烷和甲醇(V:V=5:1)混合溶剂中,搅拌1小时,加入三乙酰氧基硼氢化钠(263mg,1.24mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物6(36mg,白色粘稠固体),产率:32.1%。
MS m/z(ESI):393.5[M+1]
1H NMR(400MHz,Methanol-d4)δ8.55(d,1H),7.80-7.76(m,1H),7.53(d,1H),7.26-7.25(m,1H),7.05-7.01(m,2H),6.78-6.70(m,2H),4.17-4.10(m,2H),3.79-3.63(m,3H),2.56-2.42(m,3H),2.19-2.10(m,2H),1.92-1.82(m,2H),1.80-1.44(m,12H).
实施例7
(R)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-苯并二氢吡喃-4-胺
Figure PCTCN2016101064-appb-000039
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(80mg,0.31mmol,采用专利申请“WO2012129495”公开的方法制备而得),R-苯并二氢吡喃-4-胺盐酸盐7a(115mg,0.62mmol,采用“European Journal of Organic Chemistry,2014(31),7034-7038,2014”公开的方法制备而得),三乙酰氧基硼氢化钠(197mg,0.93mmol)溶解于10mL二氯甲烷和甲醇(V:V=5:1)混合溶剂中,搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物7(30mg,浅黄色油状物),产率:24.8%。
MS m/z(ESI):393.5[M+1]
1H NMR(400MHz,Methanol-d4)δ8.63(d,1H),7.93(t,1H),7.64(d,1H),7.39(t,1H),7.29(t,1H),7.19(d,1H),6.81-6.97(m,2H),4.25-4.35(m,1H),4.14-4.24(m,1H),3.79 (d,2H),2.47-2.65(m,3H),2.13-2.32(m,3H),1.87-2.03(m,2H),1.72-1.85(m,2H),1.40-1.71(m,5H),1.25-1.35(m,2H),1.06-1.15(m,1H),0.66-0.75(m,1H).
实施例8
6-氟-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-苯并二氢吡喃-4-胺
Figure PCTCN2016101064-appb-000040
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(30mg,0.12mmol,采用专利申请“WO2012129495”公开的方法制备而得),6-氟苯并二氢吡喃-4-胺8a(39mg,0.23mmol,采用“Bioorganic & Medicinal Chemistry Letters,2011,21(5),1338-1341”公开的方法制备而得)溶解于20mL二氯甲烷中,加入三乙酰氧基硼氢化钠(74mg,0.35mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物8(10mg,淡黄色固体),产率:20.4%。MS m/z(ESI):411.2[M+1]
1H NMR(400MHz,CDCl3)δ8.56(d,1H),7.67-7.64(m,1H),7.34-7.31(m,1H),7.16-7.14(m,1H),6.84-6.74(m,2H),6.73-6.7(m,1H),4.02-4.08(m,2H),3.78-3.75(m,3H),2.66-2.12(m,6H),2.1-1.59(m,9H),1.35-1.18(m,4H).
实施例9
(R)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺
Figure PCTCN2016101064-appb-000041
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(35mg,0.14mmol,采用专利申请“WO2012129495”公开的方法制备而得),(R)-1,2,3,4-四氢-1-萘胺9a(40mg,0.27mmol,采用“Angewandte Chemie-International Edition,45(28),4641-4644,2006”公开的方法制备而得)溶解于5mL二氯乙烷中,搅拌反应1小时,加入三乙酰氧基硼氢化钠(144mg,0.68mmol),搅拌反应1小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物9(15mg,黄色固体),产率:27.5%。
MS m/z(ESI):391.2[M+1]
1H NMR(400MHz,CDCl3)δ8.57(d,1H),7.65(t,1H),7.32(d,1H),7.16(d,1H),7.11-7.07(m,3H),7.05(d,1H),3.77(d,2H),3.60-3.57(br,1H),2.73-2.70(m,3H),2.45(d,1H),2.34(d,1H),2.15-2.08(m,1H),2.05-2.02(m,1H),1.91(d,1H),1.75-1.70(m,12H),1.50-1.44(m,3H),
实施例10
(S)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺
Figure PCTCN2016101064-appb-000042
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(20mg,0.14mmol,采用专利申请“WO2012129495”公开的方法制备而得),(S)-1,2,3,4-四氢-1-萘胺10a(50mg,0.272mmol,采用“Angewandte Chemie-International Edition,45(28),4641-4644,2006”公开的方法制备而得)溶解于20mL二氯乙烷中,搅拌反应1小时,加入三乙酰氧基硼氢化钠(144mg,0.68mmol),搅拌反应1小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物10(15mg,黄色固体),产率:28.3%。
MS m/z(ESI):391.2[M+1]
1H NMR(400MHz,Methanol-d4)δ8.77(d,1H),8.28(t,1H),7.92(d,1H),7.71(t,1H),7.33-7.19(m,4H),4.38(t,1H),3.80-3.74(m,2H),3.23-3.11(m,1H),3.08-2.98(m,1H), 2.87-2.82(m,2H),2.56-2.48(m,3H),2.26-2.0 4(m,5H),1.85-1.81(m,3H),1.56-1.32(m,5H),1.34-1.31(m,1H),0.82-0.79(m,1H).
实施例11
(S)-4-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-3,4-二氢萘-1(2H)-酮
Figure PCTCN2016101064-appb-000043
第一步
(S)-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯11a
将(S)-1,2,3,4-四氢-1-萘胺10a(3g,20.41mmol,采用“Angewandte Chemie-International Edition,45(28),4641-4644,2006”公开的方法制备而得)溶解于100mL二氯甲烷中,加入三乙胺(5.7mL,40.82mmol),加入二叔丁基二碳酸酯(4.9g,22.45mmol),搅拌反应12小时。反应液依次用水(100mL),饱和碳酸氢钠溶液洗涤(100mL),有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题产物11a(5.6g,淡黄色油状物),产物不经纯化直接进行下步反应。
MS m/z(ESI):248.3[M+1]
第二步
(S)-4-羰基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯11b
将粗品(S)-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯11a(5.6g,20.41mmol)溶解于90mL丙酮和水(V/V=2:1)混合溶剂中,加入硫酸镁(5.5g,45.66mmol),搅拌下缓慢加入高锰酸钾(7.22g,45.66mmol),搅拌反应12小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题产物11b(3.1g,类白色固体),产率:52%。
MS m/z(ESI):262.3[M+1]
第三步
(S)-4-氨基-3,4-二氢萘-1(2H)-酮11c
将(S)-4-羰基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯11b(1g,3.83mmol)溶解于20mL二氯甲烷中,加入8mL 4M氯化氢的1,4-二氧六环溶液,搅拌反应2小时。反应液减压浓缩,所得残余物加入10mL乙醇,滴加30%浓度氨水至反应液pH为8,混合液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物11c(400mg,绿色粘稠物),产率:64.8%。
MS m/z(ESI):162.3[M+1]
第四步
(S)-4-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-3,4-二氢萘-1(2H)-酮11
将(S)-4-氨基-3,4-二氢萘-1(2H)-酮11c(200mg,1.24mmol),(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(268mg,1.04mmol,采用专利申请“WO2012129495”公开的方法制备而得)溶解于20mL二氯乙烷中,搅拌反应1小时,加入三乙酰氧基硼氢化钠(1.1g,5.18mmol),搅拌反应2小时。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物11(136mg,白色固体),产率:32.4%。
MS m/z(ESI):405.6[M+1]
1H NMR(400MHz,Methanol-d4)δ8.73(d,1H),8.15-8.09(m,2H),7.83(d,1H),7.81-7.69(m,3H),7.47(d,1H),4.45(t,1H),3.77-3.74(m,2H),3.03-2.98(m,1H),2.75-2.68(m,3H),2.51-2.44(m,5H),2.05-2.01(m,2H),1.57-1.48(m,7H),1.20-1.05(m,1H),0.80-0.77(m,1H).
实施例12和实施例13
(1S,4S)-4-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-1,2,3,4-四氢-1-萘酚12
(1R,4S)-4-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-1,2,3,4-四氢-1-萘酚13
Figure PCTCN2016101064-appb-000044
Figure PCTCN2016101064-appb-000045
将(S)-4-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-3,4-二氢萘-1(2H)-酮11(50mg,0.12mmol)溶解于10mL二氯甲烷中,-78℃下滴加0.29ml 1M的二异丁基氢化铝溶液,-78℃下搅拌反应2小时。加入5ml甲醇淬灭反应,升至室温,反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物12(18mg,灰白色粘稠固体),产率:35.3%;13(20mg,灰白色粘稠固体),产率:39.2%。
12:MS m/z(ESI):407.6[M+1],
1H NMR(400MHz,CDCl3)δ8.51(d,1H),7.50(t,1H),7.36(d,1H),7.33-7.30(m,3H),7.21-7.18(m,2H),4.83(t,1H),4.25(t,1H),3.81-3.75(m,2H),2.85-2.83(m,1H),2.36-2.30(m,5H),1.98-1.80(m,2H),1.78-1.60(m,9H),1.48-1.25(m,5H).
13:MS m/z(ESI):407.6[M+1],
1H NMR(400MHz,CDCl3)δ8.51(d,1H),7.50(t,1H),7.36(d,1H),7.33-7.30(m,3H),7.21-7.18(m,2H),4.83(t,1H),4.25(t,1H),3.81-3.75(m,2H),2.85-2.83(m,1H),2.36-2.30(m,5H),1.98-1.80(m,2H),1.78-1.60(m,9H),1.48-1.25(m,5H).
实施例14
(1S,4S)-4-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺
Figure PCTCN2016101064-appb-000046
Figure PCTCN2016101064-appb-000047
第一步
(1S,4S)-4-羟基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯14a
将(S)-4-羰基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯11b(100mg,0.883mmol)溶解于5mL甲苯中,降温至0℃,加入(R)-2-甲基-CBS-恶唑硼烷(0.1ml,0.076mmol),搅拌5分钟,加入硼烷甲基硫醚(0.88ml,0.76mmol),搅拌反应2小时。加入50ml饱和氯化钠溶液淬灭反应,用乙酸乙酯萃取(30mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物14a(60mg,白色固体),产率60%。
MS m/z(ESI):208.3[M-55]
第二步
(1S,4S)-4-甲氧基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯14b
将粗品化合物14a(30mg,0.11mmol)溶解于4mL二氯甲烷中,加入氧化银(76mg,0.33mmol),碘甲烷(62mg,0.44mmol),搅拌反应48小时。过滤,滤液减压浓缩,得到粗品标题产物14b(30mg,黄色油状物),产物不经纯化直接进行下步反应。
MS m/z(ESI):278.4[M+1]
第三步
(1S,4S)-4-甲氧基-1,2,3,4-四氢萘-1-胺盐酸盐14c
将粗品化合物14b(30mg,0.11mmol)溶解于0.5mL二氯甲烷中,加入1mL 4M氯化氢的1,4-二氧六环溶液,搅拌反应2.5小时。反应液减压浓缩,得到粗品标题产物14c(24mg,白色固体),产物不经纯化直接进行下步反应。
MS m/z(ESI):178.4[M+1]
第四步
(1S,4S)-4-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺14
将化合物5a(29mg,0.11mmol),粗品化合物14c(24mg,0.11mmol),硫酸钠(60mg,0.42mmol)溶解于4mL甲醇中,搅拌反应12小时,加入硼氢化钠(8mg,0.22mmol),搅拌反应15分钟。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物14(4mg,白色固体),产率:8.7%。
MS m/z(ESI):407.6[M+1]
1H NMR(400MHz,CDCl3)δ8.56(d,1H),7.66(t,1H),7.33(d,1H),7.15(d,1H),7.08-7.06(m,3H),7.04(d,1H),3.76(d,2H),3.61-3.58(br,1H),3.41(s,3H),2.74-2.72(m,3H),2.46(d,1H),2.32(d,1H),2.13-2.08(m,1H),2.03-2.00(m,1H),1.90(d,1H),1.75-1.72(m,11H),1.51-1.46(m,3H),
实施例15
(R)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-2,3-二氢-1H-茚-1-胺
Figure PCTCN2016101064-appb-000048
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(20mg,0.08mmol),(R)-2,3-二氢-1H-茚-1-胺盐酸盐15a(27mg,0.16mmol,采用“Synthesis,(14),2283-2287,2008”公开的方法制备而得)溶解于10mL二氯甲烷中,搅拌反应2小时,加入三乙酰氧基硼氢化钠(51mg,0.24mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物15(5mg,黄色油状物),产率:16.7%。
MS m/z(ESI):377.5[M+1]
1H NMR(400MHz,Methanol-d4)δ8.62(d,1H),7.91(t,1H),7.60(d,1H),7.37(s,4H),7.35(d,1H),4.64-4.70(m,1H),3.76(d,2H),2.91-3.15(m,2H),2.41-2.60(m,4H),1.85-2.11(m,4H),1.70-1.81(m,2H),1.41-1.69(m,5H),1.31-1.39(m,1H),1.10-1.20(m,1H),0.71-0.80(m,1H).
实施例16
(S)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-2,3-二氢-1H-茚-1-胺
Figure PCTCN2016101064-appb-000049
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4,5]癸烷-9-基)乙醛5a(20mg,0.08mmol),(S)-2,3-二氢-1H-茚-1-胺盐酸盐16a(26mg,0.15mmol,采用“Tetrahedron Asymmetry,14(22),3479-3485;2003”公开的方法制备而得)溶解于10mL二氯甲烷中,搅拌2小时,加入三乙酰氧基硼氢化钠(49mg,0.23mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物16(5mg,黄色油状物),产率:17%。
MS m/z(ESI):377.5[M+1]
1H NMR(400MHz,Methanol-d4)δ8.63(d,1H),7.90(t,1H),7.60(d,1H),7.38(s,4H),7.35(d,1H),4.65-4.70(m,1H),3.76(d,2H),2.90-3.16(m,2H),2.40-2.60(m,4H),1.85-2.10(m,4H),1.70-1.80(m,2H),1.40-1.69(m,5H),1.30-1.39(m,1H),1.10-1.20(m,1H),0.70-0.80(m,1H).
实施例17
(1S,2S)-2-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-2,3-二氢-1H-茚-1-胺
Figure PCTCN2016101064-appb-000050
第一步
(1S,2S)-2-甲氧基-2,3-二氢-1H-茚-1-胺2,2,2-三氟乙酸盐17b
将(1S,2S)-2-甲氧基-2,3-二氢-1H-茚-1-氨基甲酸叔丁酯17a(110mg,0.42mmol,采用专利申请“WO2014078454”公开的方法制备而得)溶解于5mL二氯甲烷中,加入1mL三氟乙酸,搅拌反应2小时。反应液减压浓缩,用硅胶色谱法以洗脱剂体系A纯化所得残余物,得到粗品标题产物17b(70mg,黄色油状物),产率:60.3%。MS m/z(ESI):164.1[M+1]
第二步
(1S,2S)-2-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-2,3-二氢-1H-茚-1-胺17
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(25mg,0.96mmol),(1S,2S)-2-甲氧基-2,3-二氢-1H-茚-1-胺2,2,2-三氟乙酸盐17b(54mg,0.19mmol)溶解于10mL二氯甲烷中,加入三乙酰氧基硼氢化钠(61mg,0.29mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物17(10mg,黄色油状物),产率:25.5%。
MS m/z(ESI):407.6[M+1]
1H NMR(400MHz,CDCl3)δ8.58(d,1H),7.71(t,1H),7.57(d,1H),7.40(d,1H),7.29(d,1H),7.16-7.24(m,3H),3.77(d,3H),3.31(s,3H),2.87-3.05(m,2H),2.24-2.50(m,4H),2.14-2.24(m,1H),1.61-1.84(m,4H),1.35-1.51(m,5H),1.24-1.35(m,2H),1.11-1.20(m,1H),0.65-0.75(m,1H).
实施例18
(1S,2S)-1-(2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-2,3-二氢-1H-茚-2-醇
Figure PCTCN2016101064-appb-000051
第一步
2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛18b
将2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙腈18a(500mg,1.95mmol,采用专利申请“WO2012129495”公开的方法制备而得)溶解于20mL甲苯中,-78℃下缓慢滴加4.2mL 1M的二异丁基氢化铝溶液,搅拌反应1.5小时。加入18mL 2M 盐酸,搅拌30分钟,滴加5M氢氧化钠溶液至反应液pH为9~10,升至室温,用乙酸乙酯萃取(30mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物18b(270mg,黄色油状物),产率:53.4%。
MS m/z(ESI):260.5[M+1]
第二步
(1S,2S)-1-(2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙氨基)-2,3-二氢-1H-茚-2-醇18
将化合物18b(20mg,0.08mmol),(1S,2S)-1-氨基-2,3-二氢-1H-茚-2-醇18c(23mg,0.15mmol,采用“Advanced Synthesis&Catalysis,350(14+15),2250-2260;2008”公开的方法制备而得)溶解于15mL二氯甲烷和甲醇(V:V=5:1)混合溶剂中,搅拌2小时,加入三乙酰氧基硼氢化钠(49mg,0.23mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物18(10mg,黄色油状物),产率:33%。
MS m/z(ESI):393.5[M+1]
1H NMR(400MHz,CDCl3)δ8.50(d,1H),7.70(t,1H),7.37(d,1H),7.20-7.26(m,3H),7.11-7.19(m,2H),3.76(d,3H),3.36(d,1H),2.88-3.05(m,2H),2.25-2.50(m,4H),2.15-2.24(m,1H),1.60-1.84(m,4H),1.36-1.51(m,5H),1.25-1.35(m,2H),1.10-1.20(m,1H),0.65-0.75(m,1H).
实施例19
(1S,4S)-4-乙氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺
Figure PCTCN2016101064-appb-000052
第二步
(1S,4S)-4-乙氧基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯19a
将粗品(1S)-4-羟基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯14a(850mg,3.23mmol),氧化银(76mg,0.33mmol)和碘乙烷(1.3mL,16.15mmol)溶解于30mL二氯甲烷中, 搅拌反应48小时。过滤,滤液减压浓缩,得到粗品标题产物19a(800mg,黄色油状物),产物不经纯化直接进行下步反应。
MS m/z(ESI):236.1[M-55]
第三步
(1S,4S)-4-乙氧基-1,2,3,4-四氢萘-1-胺19b
将粗品化合物19a(698mg,2.4mmol)溶解于4mL二氯甲烷中,加入8mL 4M氯化氢的1,4-二氧六环溶液,搅拌反应2小时。反应液减压浓缩,乙酸乙酯打浆(30mL),过滤,滤饼溶于20mL二氯甲烷和甲醇(V:V=5:1)的混合溶剂中,用饱和碳酸氢钠溶液调节反应液pH为7~8,反应液减压浓缩,用二氯甲烷和甲醇(V:V=5:1)的混合溶剂洗涤(30mL×2),过滤,滤液减压浓缩,得到粗品标题产物19b(310mg,黄色液体),产物不经纯化直接进行下步反应。
MS m/z(ESI):191.1[M+1]
第四步
(1S,4S)-4-乙氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺19
将(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙醛5a(500mg,1.85mmol),粗品化合物19b(310mg,1.85mmol)溶解于30mL二氯乙烷中,搅拌反应40分钟,加入三乙酰氧基硼氢化钠(980mg,4.63mmol),搅拌反应2小时。依次用饱和碳酸氢钠溶液(30mL×3),用饱和氯化钠溶液洗涤(30mL×3),有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物19(280mg,黄色粘稠固体),产率:35%。
MS m/z(ESI):435.3[M+1]
1H NMR(400MHz,CDCl3)δ9.74(d,1H),9.58(d,1H),8.94(d,1H),8.37(d,1H),7.94(d,1H),7.67(d,1H),7.52(d,1H),7.47(t,1H),4.46-4.49(m,1H),4.30-4.33(m,1H),3.84-3.87(m,1H),3.66-3.70(m,2H),3.53-3.56(m,2H),2.82-2.85(d,2H),2.67(s,2H),2.39-2.41(m,4H),2.30-2.33(m,4H),1.85(s,2H),1.48-1.52(m,6H),1.27(m,3H).
实施例20
(1S,4S)-4-(环丙基甲氧基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺
Figure PCTCN2016101064-appb-000053
Figure PCTCN2016101064-appb-000054
第一步
(S)-4-羰基-1,2,3,4-四氢萘-1-基(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)氨基甲酸叔丁酯20a
将化合物11(220mg,0.54mmol),二碳酸二叔丁酯(173mg,0.82mmol)和三乙胺(0.15mL,1.08mmol)溶解于20mL二氯甲烷中,搅拌反应12小时。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物20a(100mg,浅黄色粘稠固体),产率:37%。
MS m/z(ESI):505.3[M+1]
第二步
(1S,4S)-4-羟基-1,2,3,4-四氢萘-1-基(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)氨基甲酸叔丁酯20b
将化合物20a(100mg,0.2mmol)和1M(R)-2-甲基-CBS-恶唑硼烷(0.04mL,0.04mmol)溶解于10mL甲苯中,降温至0℃,加入2M硼烷甲基硫醚(0.02mL,0.4mmol),升至室温,搅拌反应3小时。加入10ml饱和氯化钠溶液淬灭反应,用乙酸乙酯萃取(10mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(10mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物20b(10mg,白色固体),产率10%。
MS m/z(ESI):507.3[M+1]
第三步
(1S,4S)-4-(环丙基甲氧基)-1,2,3,4-四氢萘-1-基(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)氨基甲酸叔丁酯20c
将化合物20b(10mg,0.02mmol)溶解于5mL N,N-二甲基甲酰胺中,加入氢化钠(2.2mg,0.06mmol),搅拌30分钟,加入环丙基溴甲烷(6.7mg,0.05mmol),搅拌反应3小时。加入20ml水淬灭反应,用乙酸乙酯萃取(10mL×3),合并有机相, 有机相用饱和氯化钠溶液洗涤(10mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题产物20c(5mg,白色固体),产物不经纯化直接进行下步反应。
MS m/z(ESI):561.0[M+1]
第四步
(1S,4S)-4-(环丙基甲氧基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺20d
将粗品化合物20c(5mg,0.0089mmol)溶解于5mL二氯甲烷中,加入4M 0.1mL盐酸的二氧六环溶液,搅拌反应2小时。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物20d(3mg,白色固体),产率73.2%。MS m/z(ESI):461.3[M+1]
1H NMR(400MHz,CD3OD)δ8.59(d,1H),7.84-7.81(m,1H),7.55(d,1H),7.53(d,1H),7.47-7.40(m,1H),7.39-7.29(m,2H),7.25(d,1H),4.48-4.46(m,1H),4.28-4.25(m,1H),3.77-3.75(m,2H),3.45-3.43(m,2H),3.35-3.30(m,2H),2.93-2.92(m,1H),2.53-2.50(m,2H),2.49-2.48(m,1H),2.25-2.13(m,2H),1.95-1.31(m,11H),1.10-1.08(m,2H),0.76-0.73(m,1H),0.55-0.53(m,2H),0.25-0.23(m,2H).
实施例21
(1S,4S)-4-(2-氟乙氧基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺
Figure PCTCN2016101064-appb-000055
第一步
(1S,4S)-4-(2-氟乙氧基)-1,2,3,4-四氢萘-1-基(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)氨基甲酸叔丁酯21a
将20b(45mg,0.088mmol)溶解于5mL N,N-二甲基甲酰胺中,加入氢化钠(20mg,0.44mmol),搅拌20分钟,加入1-溴-2-氟乙烷(23mg,0.176mmol),搅拌反应16小时。加入5ml水淬灭反应,用乙酸乙酯萃取(10mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩,用 薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物21a(30mg,黄色油状物),产率:61.1%。
MS m/z(ESI):553.4[M+1]
第二步
(1S,4S)-4-(2-氟乙氧基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺21
将化合物21a(30mg,0.543mmol)溶解于10mL二氯甲烷中,加入4M 0.3mL盐酸的二氧六环溶液,搅拌反应1小时。加入10mg碳酸钠,反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物21(10mg,白色粘稠物),产率40.7%。
MS m/z(ESI):453.4[M+1]
1H NMR(400MHz,CDCl3)δ9.76(d,1H),9.61(d,1H),8.89(d,1H),8.34(d,1H),7.94(d,1H),7.69(d,1H),7.58(d,1H),7.44(t,1H),4.43-4.49(m,2H),4.28-4.33(m,2H),3.81-3.87(m,1H),3.61-3.71(m,2H),3.51-3.56(m,2H),2.81-2.89(d,2H),2.67(s,2H),2.39-2.43(m,4H),2.30-2.36(m,4H),1.85(s,2H),1.48-1.61(m,6H).
实施例22,23
(1S,4S)-4-(甲氧基甲基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺22
(1S,4R)-4-(甲氧基甲基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺23
Figure PCTCN2016101064-appb-000056
第一步
(S)-4-亚甲基-1,2,3,4-四氢萘-1-基氨基甲酸叔丁酯22a
将甲基三苯基溴化磷(2.95g,11.5mmol)溶解于20mL四氢呋喃中,降温至0℃,加入叔丁醇钾(1.29g,11.5mmol),搅拌反应30分钟,加入11b(1g,7.66mmol),升至室温,搅拌反应12小时。反应液减压浓缩,加入甲醇溶解,用薄层色谱法以洗脱剂体系B纯化所得残余物,得到标题产物22a(200mg,白色固体),产率:22.2%。MS m/z(ESI):204.2[M-55]
第二步
(1S)-4-(羟基甲基)-1,2,3,4-四氢萘-1-基氨基甲酸叔丁酯22b
将22a(780mg,3mmol)溶解于20mL四氢呋喃中,降温至0℃,加入6mL 1M硼烷四氢呋喃溶液,搅拌反应5小时,加入12mL 3M氢氧化钠溶液,搅拌反应30分钟,加入12mL 30%的双氧水,升至室温,搅拌反应12小时。反应液减压浓缩,用二氯甲烷萃取,合并有机相,有机相减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物22b(740mg,白色固体),产率:89.2%。
MS m/z(ESI):222.1[M-55]
第三步
(1S)-4-(甲氧基甲基)-1,2,3,4-四氢萘-1-基氨基甲酸叔丁酯22c
将22b(200mg,0.72mmol)溶解于10mL四氢呋喃中,加入氢化钠(60mg,1.4mmol),搅拌反应1小时,加入碘甲烷(123mg,0.86mmol),搅拌反应12小时。反应液减压浓缩,加入甲醇溶解,用薄层色谱法以洗脱剂体系B纯化所得残余物,得到标题产物22c(20mg,白色固体),产率:9.5%。
MS m/z(ESI):236.2[M-55]
第四步
(1S)-4-(甲氧基甲基)-1,2,3,4-四氢萘-1-胺22d
将粗品22c(20mg,0.07mmol)溶解于10mL二氯甲烷中,加入10mL 4M氯化氢的1,4-二氧六环溶液,搅拌反应2小时。反应液减压浓缩,得到粗品标题产物22d(13mg,黄色油状物),产物不经纯化直接进行下步反应。
MS m/z(ESI):192.2[M-55]
第五步
(1S,4S)-4-(甲氧基甲基)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺22
将5a(30mg,0.116mmol),粗品22d(22mg,0.116mmol)溶解于20mL二氯乙烷和甲醇(V:V=1:1)的混合溶剂中,加入氰基硼氢化钠(15mg,0.23mmol),搅拌反应12小时。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物22(10mg,白色固体),产率:20%和标题产物23(8mg,白色固体),产率:16%。
MS m/z(ESI):435.3[M+1]
实施例22
1H NMR(400MHz,CDCl3)δ8.26-8.25(d,1H),7.76-7.72(t,1H),7.42-7.40(d,1H),7.34-7.29(m,3H),7.29-7.27(m,1H),7.18-7.16(m,1H),4.16(s,1H),3.75-3.70(m,2H),3.47-3.45(m,2H),3.41(s,3H),3.35-3.33(m,1H),3.18-3.17(m,1H),2.80-2.70(m,1H),2.4-2.33(m,1H),2.28-1.95(m,7H),1.81-1.62(m,5H),1.59-1.51(m,1H),1.46-1.20(m,4H),1.22-1.1(m,1H).
实施例23
1H NMR(400MHz,CDCl3)δ8.47-8.46(d,1H),7.72-7.68(t,1H),7.39-7.37(d,1H),7.37-7.33(m,1H),7.24-7.21(m,1H),7.15-7.05(m,2H),6.93-6.91(d,1H),,3.94(s,1H),3.68-3.60(m,2H),3.59-3.57(m,2H),3.22(s,3H),3.22-3.19(m,1H),2.71-2.70(m,1H),2.34-2.30(m,5H),2.28-2.25(m,1H),1.84-1.81(m,1H),1.81-1.71(m,5H),1.69-1.51(m,2H),1.45-1.4(m,2H),1.32-1.26(m,2H),1.23-1.15(m,1H),1.1-0.95(m,1H).
实施例24
(S)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-3',4'-二氢-2'H-螺[[1,3]二硫戊环-2,1'-萘]-4'-胺24
Figure PCTCN2016101064-appb-000057
将11(35mg,0.0865mmol),乙烷-1,2-二硫醇24a(82mg,0.865mmol),吡啶对甲苯磺酸盐(240mg,0.952mmol)溶解于15mL甲苯中,升温至110℃,搅拌反应12小时。反应液减压浓缩,用薄层色谱法以洗脱剂体系A纯化所得残余物,得到标题产物24(40mg,浅黄色固体),产率96%。
MS m/z(ESI):481.2[M+1]
1H NMR(400MHz,CD3OD):δ8.60(d,1H),7.60(t,1H),7.25-7.31(m,2H),7.15-7.20(m,4H),4.26-4.30(m,1H),3.76(d,2H),2.81-3.01(m,4H),2.41-2.60(m,2H),2.21-2.30(m,2H),1.86-2.13(m,4H),1.70-1.81(m,2H),1.41-1.69(m,5H),1.31-1.39(m,2H),1.10-1.20(m,2H),0.71-0.80(m,2H).
实施例25
(1S,4R)-4-乙氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺25
Figure PCTCN2016101064-appb-000058
第一步
(1S,4R)-4-羟基-1,2,3,4-四氢萘-1-氨基甲酸叔丁酯25a
将(S)-2-甲基-CBS-恶唑硼烷(221.8mg,0.8mmol)溶于140mL四氢呋喃中,氩气保护下,加入硼烷甲基硫醚(2.4mL,48mmol),升温至30℃,30分钟内滴加80mL预制的11b(10.5g,40mmol)四氢呋喃溶液,30℃搅拌1小时。15℃下加入100mL甲醇搅拌1小时淬灭反应,减压浓缩,加入200mL乙酸乙酯和5g活性炭,微沸搅拌30分钟,过滤,乙酸乙酯洗涤滤饼(100mL×3),滤液减压浓缩,得到粗品标题产物25a(10.5g,无色油状物)。
MS m/z(ESI):264.4[M+1]
第二-四步
(1S,4R)-4-乙氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-1,2,3,4-四氢萘-1-胺25
采用实施例19的合成路线,将原料14a替换为25a,制得标题产物25(7g,淡红色油状物)。
MS m/z(ESI):435.5[M+1]
1H NMR(400MHz,DMSO-d6)δ0.62(dt,1H),0.92-1.03(m,1H),1.12(t,3H),1.34(td,2H),1.41-1.69(m,9H),1.79(d,1H),1.82-1.92(m,2H),2.02(td,1H),2.26-2.38(m,2H),2.43(d,1H),3.37-3.48(m,2H),3.52-3.66(m,3H),4.25(t,1H),7.11-7.16 (m,2H),7.16-7.20(m,1H),7.21-7.28(m,2H),7.45(d,1H),7.71(td,1H),8.52(dd,1H)
实施例26
(1S,4S)-4-(乙氧基-d5)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺26
Figure PCTCN2016101064-appb-000059
第一步
((1S,4S)-4-(乙氧基-d5)-1,2,3,4-四氢萘-1-基)氨基甲酸叔丁酯26a
将14a(3.3g,12.5mmol)溶解于50mL N,N-二甲基甲酰胺中,加入活化的分子筛,氩气保护下,冷却至0℃,加入氢氧化钠(0.75g,18.75mmol),0℃搅拌反应0.5小时,再加入氘代碘乙烷-d5(0.8mL,10mmol),0℃密封反应16小时。反应结束后,反应液倒入50mL水、50mL正己烷和5mL乙酸乙酯混合溶剂中,搅拌10分钟,过滤,除去不溶物。滤液分液,水相用正己烷和乙酸乙酯(V:V=10:1)混合溶剂萃取(33mL×2),合并有机相,用饱和氯化钠溶液洗涤(30mL×2),无水硫酸钠干燥。过滤,减压浓缩,用CombiFlash快速制备仪以洗脱剂体系B纯化所得残余物,得到标题化合物26a(1.89g,白色固体),产率:64%。
MS m/z(ESI):241.4[M-56+1]
第二步
(1S,4S)-4-(乙氧基-d5)-1,2,3,4-四氢萘-1-胺26b
将26a(1.89g,6.38mmol)中加入8mL 4M氯化氢的1,4-二氧六环溶液,搅拌反应1小时。反应液减压浓缩,加入30mL乙酸乙酯,减压浓缩。所得残余物中加入1mL饱和碳酸钠溶液,搅拌5分钟,加入30mL乙酸乙酯,2g碳酸钠固体和10g硫酸钠,搅拌30分钟至溶液不再浑浊。过滤,滤液减压浓缩,得到粗品标题产物26b(1.21g,淡黄褐色液体),产物不经纯化直接进行下一步反应。
MS m/z(ESI):197.4[M+1]
第三步
(1S,4S)-4-(乙氧基-d5)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺26
将1a(1.37g,5.31mmol),粗品26b(1.21g,6.16mmol)溶解于50mL二氯乙烷中,加入一滴醋酸,搅拌反应1小时,加入三乙酰氧基硼氢化钠(2.81g,13.27mmol),搅拌反应16小时。反应液中加入10mL饱和碳酸钠溶液,搅拌5分钟,依次滴加入10mL 15%氢氧化钠溶液,30mL水,30mL二氯甲烷,搅拌5分钟,分液,水相用二氯甲烷萃取(50mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物26(1.7g,淡黄色液体),产率:73%。
MS m/z(ESI):440.5[M+1]
1H NMR(400MHz,CDCl3)δ0.70(dt,1H),1.09-1.16(m,1H),1.45-1.55(m,4H),1.62-1.84(m,6H),1.86-2.04(m,4H),2.23(td,1H)2.34(dd,1H),2.44(dd,1H),2.53(td,1H),3.68(br.s.,1H),3.72-3.81(m,2H),4.34(t,1H),7.11(ddd,1H),7.17(t,2H),7.18-7.23(m,1H),7.31(t,2H),7.62(td,1H),8.55(dd,1H)
实施例27
(S)-4-乙基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2-二氢萘-1-胺27
Figure PCTCN2016101064-appb-000060
第一步
(S,E)-(4-乙亚基-1,2,3,4-四氢萘-1-基)氨基甲酸叔丁酯27a
将乙基三苯基溴化膦(2.1g,5.75mmol)溶于20mL四氢呋喃中,冰水浴下,加入叔丁醇钾(643mg,5.75mmol),冰水浴下搅拌反应30分钟,滴加入预制的11b(1g,3.83mmol)四氢呋喃溶液,25℃下搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物27a(530mg,浅黄色油状物),产率:51%。
第二步
(S)-4-乙基-1,2-二氢萘-1-胺盐酸盐27b
将27a(273mg,1mmol)溶解于5mL二氯甲烷中,加入2mL 1M氯化氢的1,4-二氧六环溶液,搅拌反应1小时。反应液减压浓缩,得到粗品标题产物27b(173mg,褐色油状物)。产品不经纯化直接进行下一步反应。
第三步
(S)-4-乙基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2-二氢萘-1-胺27
将1a(150mg,0.58mmol),粗品27b(158mg,0.58mmol)溶解于30mL二氯甲烷和甲醇(V:V=10:1)混合溶剂中,加入三乙酰氧基硼氢化钠(369mg,1.74mmol),搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物27(40mg,浅黄色固体),产率:17%。
MS m/z(ESI):417.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.58(d,1H),7.83-7.78(m,1H),7.53-7.48(m,3H),7.33-7.29(m,2H),7.21(d,1H),5.84(t,1H),4.25(t,1H),3.73-3.72(m,3H),3.41-3.31(m,2H),2.81-2.80(m,2H),2.41-2.25(m,3H),1.96-1.90(m,3H),1.85-1.61(m,8H),1.25(t,3H),1.23-1.21(m,1H),0.68-0.65(m,1H).
实施例28
(S)-4-亚甲基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺28
Figure PCTCN2016101064-appb-000061
采用实施例17的合成路线,将原料17a替换为原料22a,制得标题产物28(20mg,褐色固体)。
MS m/z(ESI):403.5[M+1]
1H NMR(400MHz,DMSO-d6)δ8.50(d,1H),7.72(t,1H),7.68-7.21(m,6H),5.95(d,1H),4.09(d,1H),3.71-3.69(m,3H),3.01-2.80(m,2H),2.67-2.63(m,2H),2.11(d,1H),1.74-1.21(m,14H),0.99-0.98(m,1H),0.45-0.42(m,1H).
实施例29
2-(((1S,4S)-4-((2-((R)-9-吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-1,2,3,4-四氢萘-1-基)氧基)乙腈29
Figure PCTCN2016101064-appb-000062
第一步
((1S,4S)-4-(氰基甲氧基)-1,2,3,4-四氢萘-1-基)(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)氨基甲酸叔丁酯29a
将20b(40mg,0.08mmol)溶解于10mL四氢呋喃中,加入叔丁醇钾(45mg,0.4mmol),加入溴乙腈(20mg,0.16mmol),搅拌反应16小时。加入20ml水和20mL乙酸乙酯,搅拌,静置,分液,乙酸乙酯萃取(30mL×2),合并有机相,减压浓缩,得到粗品标题产物29a(50mg,油状物)。产物不经纯化直接进行下一步反应。
第二步
2-(((1S,4S)-4-((2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-1,2,3,4-四氢萘-1-基)氧基)乙腈29
将粗品29a(50mg,0.1mmol)溶解于10mL二氯甲烷中,加入0.1mL 4M氯化氢的二氧六环溶液,搅拌反应0.5小时。反应液加入氨水至碱性,减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物29(10mg,白色蜡状物),产率8%。
MS m/z(ESI):446.3[M+1]
1H NMR(400MHz,DMSO-d6)δ8.54(d,1H),7.75-7.72(m,1H),7.43(d,1H),7.37-7.32(m,2H),7.28-7.15(m,3H),4.67(d,1H),4.40(d,2H),4.31(d,1H),3.97(d,1H),3.63-3.51(m,2H),2.41-2.25(m,2H),2.16-2.06(m,2H),2.04-1.87(m,2H),1.86-1.72(m,4H),1.62-1.21(m,8H),1.04-0.94(m,1H),0.68-0.61(m,1H).
实施例30
(1S,4R)-4-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺30
Figure PCTCN2016101064-appb-000063
第一步
((1S,4R)-4-羟基-1,2,3,4-四氢萘-1-基)(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基甲酸叔丁酯30a
将化合物13(46mg,0.11mmol),二碳酸二叔丁酯(27mg,0.121mmol)和三乙胺(23mg,0.22mmol)溶解于15mL二氯甲烷中,搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物30a(46mg,白色固体),产率:82%。
MS m/z(ESI):507.3[M+1]
第二步
((1S,4R)-4-甲氧基-1,2,3,4-四氢萘-1-基)(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基甲酸叔丁酯30b
将化合物30a(46mg,0.091mmol)溶解于10mL四氢呋喃中,加入氢化钠(8mg,0.182mmol),室温搅拌30分钟,加入碘甲烷(16mg,0.11mmol),室温搅拌反应16小时。加入50mL水和50mL乙酸乙酯,分液,有机相减压浓缩,得到粗品标题产物30b(47mg,褐色固体),产物不经纯化直接进行下一步反应。
MS m/z(ESI):521.3[M+1]
第四步
(1S,4R)-4-甲氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺30
将粗品30b(47mg,0.091mmol)溶解于10mL二氯甲烷中,加入0.1mL 4M氯化氢的1,4-二氧六环溶液,搅拌反应1小时。反应液减压浓缩,残余物中加入乙 醇,氨水调节pH至8,减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物30(36mg,黄色粘稠物),产率95%。
MS m/z(ESI):421.3[M+1]
1H NMR(400MHz,DMSO-d6)δ8.55(d,1H),7.75-7.72(m,1H),7.46(d,1H),7.37-7.32(m,2H),7.28-7.15(m,3H),4.67(d,1H),4.30(d,1H),3.97(d,1H),3.64-3.50(m,2H),3.35(s,3H),2.41-2.26(m,2H),2.16-2.06(m,2H),2.04-1.87(m,2H),1.86-1.72(m,4H),1.62-1.21(m,8H),1.04-0.94(m,1H),0.68-0.61(m,1H).
实施例31
2-((S,E)-4-((2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-3,4-二氢萘-1(2H)-亚基)乙腈31
Figure PCTCN2016101064-appb-000064
第一步
(S,E)-(4-(氰基亚甲基)-1,2,3,4-四氢萘-1-基)氨基甲酸叔丁酯31a
将氰甲基膦酸二乙酯(200mg,0.76mmol)溶于20mL四氢呋喃中,冰水浴下,加入氢化钠(61mg,1.52mmol),冰水浴下搅拌反应30分钟,滴加入预制的11b(200mg,0.76mmol)四氢呋喃溶液,25℃下搅拌反应16小时。反应液倒入冰水中,乙酸乙酯萃取三次,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物31a(150mg,无色粘稠物),产率69%。
MS m/z(ESI):285.1[M+1]
第二步
(S,E)-2-(4-氨基-3,4-二氢萘-1(2H)-亚基)乙腈盐酸盐31b
将31a(150mg,0.52mmol)溶解于5mL二氯甲烷中,加入2mL 1M氯化氢的1,4-二氧六环溶液,搅拌反应3小时。反应液减压浓缩,得到粗品标题产物31b(110mg,白色固体)。产品不经纯化直接进行下一步反应。
第三步
2-((S,E)-4-((2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-3,4-二氢萘-1(2H)-亚基)乙腈31
将5a(100mg,0.39mmol),粗品31b(85mg,0.39mmol)溶解于10mL二氯甲烷和甲醇(V:V=10:1)混合溶剂中,加入三乙酰氧基硼氢化钠(165mg,0.78mmol),搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物31(30mg,浅黄色粘稠物),产率:18%。
MS m/z(ESI):428.0[M+1]
1H NMR(400MHz,DMSO-d6)δ8.57(d,1H),7.86-7.78(m,1H),7.76-7.74(m,1H),7.39-7.22(m,3H),7.26-7.23(m,2H),6.36-6.35(m,1H),3.65-3.54(m,3H),2.90-2.60(m,2H),2.42-2.37(m,3H),2.03-1.90(m,4H),1.82-1.78(m,2H),1.51-1.24(m,10H).
实施例32
2-((4S)-4-((2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-1,2,3,4-四氢萘-1-基)乙腈32
Figure PCTCN2016101064-appb-000065
第一步
2-((4S)-4-氨基-1,2,3,4-四氢萘-1-基)乙腈盐酸盐32a
将31b(50mg,0.227mmol)溶于5mL乙醇中,加入5mg钯碳,氢气置换三次,氢气氛下室温下反应16小时。过滤除去不溶物,滤液减压浓缩,得到粗品标题产物32a(45mg,无色粘稠物)。产品不经纯化直接下一步反应。
第二步
2-((4S)-4-((2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)氨基)-1,2,3,4-四氢萘-1-基)乙腈32
将5a(53mg,0.2mmol),粗品32a(25mg,0.2mmol)溶解于10mL二氯甲烷和甲醇(V:V=10:1)混合溶剂中,加入三乙酰氧基硼氢化钠(80mg,0.4mmol),搅拌反应16小时。反应液减压浓缩,用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物32(5mg,浅黄色粘稠物),产率:5.8%。
MS m/z(ESI):430.3[M+1]
1H NMR(400MHz,DMSO-d6)δ8.55(d,1H),7.85-7.78(m,1H),7.75-7.72(m,1H),7.35-7.20(m,3H),7.25-7.21(m,2H),3.75-3.60(m,3H),2.95-2.80(m,2H),2.70-2.65(m,4H),2.41-2.30(m,4H),1.95-1.89(m,4H),1.85-1.60(m,4H),1.55-1.21(m,6H).
实施例33
(S)-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸烷-9-基)乙基)-3,4-二氢-4H-螺[萘-1,2'-[1,3]二氧戊环]-4-胺33
Figure PCTCN2016101064-appb-000066
采用实施例27的合成路线,将原料2替换为原料11,制得标题产物33(5mg,黄色油状物)。
MS m/z(ESI):449.0[M+1]
1H NMR(400MHz,DMSO-d6):δ8.58(d,1H),7.81(s,1H),7.77-7.70(m,1H),7.51(d,1H),7.27-7.11(m,4H),3.85(s,1H),3.66-3.50(m,5H),3.51-3.42(m,1H),3.42-3.33(m,1H),2.48-2.35(m,2H),2.38-2.32(m,1H),2.20-2.08(m,2H),2.01-1.88(m,2H),1.85-1.75(m,3H),1.71-1.31(m,8H),1.00-0.96(m,1H),0.70-0.62(m,1H).
实施例34
(1S,4S)-4-丙氧基-N-(2-((R)-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-9-基)乙基)-1,2,3,4-四氢萘-1-胺34
Figure PCTCN2016101064-appb-000067
Figure PCTCN2016101064-appb-000068
采用实施例21的合成路线,将原料1-溴-2-氟乙烷替换为碘丙烷,制得标题产物34(8mg,黄色固体)。
MS m/z(ESI):449.3[M+1]
1H NMR(400MHz,DMSO-d6)δ8.53(d,1H),7.75-7.70(m,1H),7.45(d,1H),7.37-7.33(m,2H),7.28-7.17(m,3H),4.65(d,1H),4.32(d,1H),3.98(d,1H),3.64-3.52(m,3H),3.49-3.40(m,1H),2.62-2.52(m,1H),2.41-2.27(m,2H),2.16-2.06(m,1H),2.04-1.87(m,2H),1.86-1.71(m,5H),1.67(d,1H),1.60-1.20(m,8H),1.13(t,3H),1.03-0.95(m,1H),0.68-0.60(m,1H).
生物学评价
以下结合测试例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。
测试例1
1、实验目的
本实验的目的是为了测试本发明化合物对MOR、KOR、DOR的激动作用,根据EC50和Emax大小评价化合物的体外活性。
2、MOR活性的测试
2.1实验目的
本发明的化合物可以激活μ-阿片受体(MOR)。激活的MOR可以调节细胞内cAMP的水平,cAMP进入细胞核与报告基因荧光素酶(Luciferase)的CRE区结合,启动报告基因的表达。,荧光素酶与其底物反应可发出荧光,通过测定荧光信号反映化合物的激动活性。
2.2实验方法
实施例化合物激动MOR影响下游cAMP水平变化的活性通过以下的方法进行测试。
2.2.1实验材料
Figure PCTCN2016101064-appb-000069
Figure PCTCN2016101064-appb-000070
2.2.2实验步骤
1)HEK293/MOR/CRE单克隆细胞株的获得
将MOR/pcDNA3.1(+)和CRE/pGL4.29转入HEK293细胞株,通过在培养基中加入G418和潮霉素(Hygromycin),并在96孔细胞培养板中筛选出HEK293/MOR/CRE单克隆细胞株。
2)实施例化合物对MOR激动作用实验
HEK293/MOR/CRE单克隆细胞株在DMEM/高葡萄糖培养基(10%FBS,1mg/ml G418,200ug/ml潮霉素,混匀)中培养,每3天传代一次。实验当天以新鲜细胞培养基制取细胞悬液,20,000细胞/孔铺96孔板(BD,#356692),5%二氧化碳37℃培养。第二天,先将化合物溶解在纯DMSO中浓度为20mM,再用DMSO配制成首个浓度4mM,并以10倍依次稀释成6个浓度,设置成空白和对照的孔加入90μl DMSO;然后再取2.5μl配制在DMSO中成梯度浓度的化合物溶液加入到97.5μl含5μM foscolin的新鲜细胞培养基;取10μl配制好的化合物加入到细胞培养板中使得化合物的终浓度为10000、1000、100、10、1、0.1、0.01nM,5%二氧化碳37℃培养5小时。在96孔细胞培养板中,每孔加入100μl荧光素酶检测液(Promega,#E6110),室温避光放置10-15min,吹吸10次,吸100μl到96孔白色板。在酶标仪(PE,Victor3)中读取化学发光信号值,读取的数据使用软件处理。
2.3测试结果
本发明的化合物激动MOR影响下游cAMP水平的变化通过以上的试验进行测定,测得的EC50值见表1.1,Emax为实施例化合物激活MOR影响cAMP信号通路的最大效应(TRV-130的最大效应为100%)。
3、KOR和DOR活性的测试
3.1实验目的
用来测定本发明化合物对于KOR受体和DOR受体下游cAMP水平影响的活性。
3.2实验步骤
在96孔板中分别接HEK293/KOR/CRE或HEK293/DOR/CRE(CRE cDNA购于Promega,货号E8471,KOR和DOR cDNA为公司构建)细胞90μl,接种密度1×104细胞/孔,细胞在37℃,5%CO2条件下培养过夜。将药物配置成20mM的储存液,用100%DMSO梯度稀释成200×浓度梯度,再用DMEM/高葡萄糖(SH30243.01B,Hyclone)培养基稀释20倍。取出第一天接种的细胞培养板,每孔 分别加入10μl稀释后药物或对照(0.5%DMSO),轻轻振荡混匀,放置37℃、5%CO2培养箱中培养4小时,最后加入检测试剂ONE-Glo(E6120,Promega)100μl/孔,室温放置5分钟,采用酶标仪(PE,Victor3)的冷光模式测吸收值。用Graphpad Prism软件根据化合物各浓度与相应的信号值计算化合物的EC50值。Emax为化合物引起cAMP水平变化的最大效应。
3.3测试结果
本发明中化合物激动KOR受体或DOR受体下游cAMP水平的变化通过以上的试验进行测定,测得的EC50值见表1.2,Emax为化合物引起cAMP水平变化的最大效应(吗啡的最大效应为100%)。
表1.1 本发明化合物激动MOR受体影响cAMP水平的EC50和Emax
Figure PCTCN2016101064-appb-000071
Figure PCTCN2016101064-appb-000072
表1.2 本发明化合物对KOR受体和DOR受体下游cAMP水平影响的EC50和Emax
Figure PCTCN2016101064-appb-000073
结论:本发明化合物对KOR受体或DOR受体的激动活性明显很弱;对MOR受体有高的选择性。
测试例2
1、实验目的
测定本发明化合物激活MOR受体的β-arrestin信号通路的活性。
2、实验方法
2.1实验步骤:
在96孔板中分别接CHO-K1OPRM1β-Arrestin(93-0213C2,DiscoveRX)细胞90μl,接种密度1×104细胞/孔,细胞在37℃,5%CO2条件下培养过夜。将药物配置成20mM的储存液,用100%DMSO梯度稀释成200×浓度梯度,再用AssayCompleteTM Cell Plating 2 Reagent(93-0563R2B,DiscoveRX)培养基稀释20倍。取出第一天接种的细胞培养板,每孔分别加入10μl稀释后药物或对照(0.5%DMSO),轻轻振荡混匀,放置37℃、5%CO2培养箱中培养90分钟,最后加入检测试剂(93-0001,DiscoveRX)50μl/孔,室温放置60分钟,采用酶标仪(PE,Victor3)的冷光模式测吸收值;用Graphpad Prism软件根据化合物各浓度与相应的信号值计算化合物的EC50值。
2.2测试结果
本发明化合物激活β-arrestin信号通路的活性通过以上的试验进行测定,测得的EC50值见表2,Emax为化合物引起β-arrestin信号通路的最大效应(吗啡的最大效应为100%)。
表2 本发明化合物激活β-arrestin信号通路活性的EC50
实施例编号 EC50(nM) Emax
1 4 12%
2 >10000 4%
11 305 37%
14 26 24%
16 94 9%
19 24 13%
20 4 9%
22 41 18%
24 6 30%
28 15 22%
29 33 32%
33 73 27%
34 16 10%
结论:本发明化合物对β-arrestin信号通路几乎无激活作用。。
测试例3
1、实验目的
应用全自动膜片钳在转染hERG钾通道的稳定细胞株上测试本发明化合物和阳性化合物TRV-130(Journal of Pharmacology and Experimental Therapeutics,Volume 344,Issue 3,Pages 708-717,2013)对hERG钾电流的阻断作用。
2、实验方法
2.1实验材料与仪器
2.1.1实验材料:
Figure PCTCN2016101064-appb-000074
2.1.2实验仪器:
Figure PCTCN2016101064-appb-000075
Figure PCTCN2016101064-appb-000076
2.2全自动膜片钳实验步骤
HEK293-hERG稳定细胞株按照1:4的密度在MEM/EBSS培养基(10%FBS,400μg/ml G418,1%MEM非必需氨基酸溶液(100×),1%丙酮酸钠溶液)中进行传代培养,培养48-72小时之内进行全自动膜片钳实验。实验当天将细胞用0.25%胰酶消化后,离心收集细胞,用细胞外液(140mM NaCl,4mM KCl,1mM MgCl2,2mM CaCl2,5mMD一水葡萄糖,10mM Hepes,pH7.4,298mOsmol)重悬细胞制成细胞悬液。将细胞悬液放置在Patchliner仪器的细胞库上,Patchliner仪器利用负压控制器将细胞加到芯片(NPC-16)上,负压将单个细胞吸引在芯片的小孔上。当形成全细胞模式后,仪器将按照设定的hERG电流电压程序得到hERG电流,然后仪器自动的由低浓度到高浓度,进行化合物灌流。通过HEAK Patchmaster,HEAKEPC10膜片钳放大器(Nanion)和Pathlinersoftware以及Pathcontrol HTsoftware提供的数据分析软件,对化合物各浓度下的电流以及空白对照电流进行分析。
2.3测试结果
本发明化合物对hERG钾电流的阻断作用通过以上的试验进行测定,测得的IC50值见表3。
表3 本发明化合物对hERG钾电流的阻断作用的IC50
实施例编号 IC50(μM)
TRV-130 1.6
1 13
3 >30
5 6.2
6 3.8
11 4.1
12 10.2
13 13.5
14 8.6
16 10
19 5.9
21 4.2
22 3.8
24 2.5
结论:本发明化合物相比阳性化合物对hERG的抑制作用更弱,有显著差异。

Claims (16)

  1. 一种通式(I)所示的化合物:
    Figure PCTCN2016101064-appb-100001
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐,
    其中:
    环A选自环烷基和杂环基;
    R选自芳基和杂芳基,其中所述的芳基和杂芳基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、-S(O)mR3和-NR4R5中的一个或多个取代基所取代;
    R1相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤素、氨基、硝基、羟基、氰基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、-S(O)mR3和-NR4R5,其中所述的烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R2相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤素、氨基、硝基、羟基、氰基、氧代基、烯基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、-S(O)mR3和-NR4R5,其中所述的烷基、烷氧基、烯基、卤代烷基、环烷基、杂环基、芳基和杂芳基任选被选自氘原子、烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    或者两个R2一起形成环烷基或杂环基,其中所述的环烷基或杂环基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R3选自氢原子、烷基、氘代烷基、氨基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤素、羟基、氨基、硝基、氰基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R4和R5各自独立地选自氢原子、烷基、烷氧基、羟烷基、羟基、氨基、羧酸酯基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤素、羟基、氨基、羧酸酯基、硝基、氰基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    p、q各自独立地为0、1、2、3或4;且
    m为0、1或2。
  2. 根据权利要求1所述的通式(I)所示的化合物,其中A环选自5~6元杂环基和5~6元环烷基。
  3. 根据权利要求1或2所述的通式(I)所示的化合物,其中R为吡啶基。
  4. 根据权利要求1所述的通式(I)所示的化合物,其为通式(II)所示的化合物:
    Figure PCTCN2016101064-appb-100002
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
    其中:
    G选自一个键、CRaRb、C=O、NR4和氧原子;
    Ra和Rb各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤素、氨基、硝基、羟基、氰基、环烷基、杂环基、芳基、杂芳基、-OR3、-C(O)R3、-C(O)OR3、-S(O)mR3和-NR4R5,其中所述的烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    或者Ra和Rb一起形成环烷基基或杂环基,其中所述的环烷基或杂环基任选被选自烷基、卤代烷基、卤素、氨基、硝基、氰基、羟基、烷氧基、卤代烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R1~R5、p、m和q如权利要求1中所定义。
  5. 根据权利要求1~4任一项所述的通式(I)所示的化合物,其中R1为氢原子或卤素。
  6. 根据权利要求1~5任一项所述的通式(I)所示的化合物,其中R2选自氢原子、烷基、氧代基、烷氧基、羟基、卤素和-OR3,其中所述的烷基和烷氧基任选被选自氘原子、烷基、卤素、羟基、氨基、羧酸酯基、硝基、氰基、烷氧基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;R3选自氢原子、任选被卤素或环烷基取代的烷基和环烷基;R2优选烷基,其中所述的烷基任选进一步被选自烷氧基、环烷基或卤代烷基;R3优选氢原子、烷基、卤代烷基或环烷基。
  7. 根据权利要求4所述的通式(I)所示的化合物,其为通式(IV)所示的化合物:
    Figure PCTCN2016101064-appb-100003
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
    其中:
    R1、R2和p如权利要求1中所定义。
  8. 根据权利要求1~7任一项所述的通式(I)所示的化合物,其中所述化合物选自:
    Figure PCTCN2016101064-appb-100004
    Figure PCTCN2016101064-appb-100005
  9. 一种制备根据权利要求1所述的通式(I)所示的化合物的方法,该方法包括:
    Figure PCTCN2016101064-appb-100006
    通式(VB)化合物和通式(VIB)化合物或其盐酸盐发生还原胺化反应,得到通式(I)化合物;
    其中:
    环A、R、R1、R2、p和q如权利要求1中所定义。
  10. 一种药物组合物,其含有治疗有效量的根据权利要求1~8任一项所述的通式(I)所示的化合物,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  11. 根据权利要求1~8任一项所述的通式(I)所示的化合物或根据权利要求10所述的药物组合物在制备预防和/或治疗MOR受体激动剂介导的相关疾病的药物中的用途。
  12. 根据权利要求11所述的用途,其中所述的MOR受体激动剂介导的相关疾病选自疼痛、免疫功能障碍、炎症、食管回流、神经和精神疾病、泌尿和生殖疾病、心血管疾病和呼吸道疾病,优选疼痛。
  13. 根据权利要求1~8任一项所述的通式(I)所示的化合物或根据权利要求10所述的药物组合物在制备预防或治疗疼痛和疼痛相关疾病的药物中的用途。
  14. 根据权利要求13所述的用途,其中所述的疼痛选自术后疼痛、癌症引起的疼痛、神经性疼痛、创伤性疼痛和炎症引起的疼痛。
  15. 根据权利要求14所述的用途,其中所述的癌症选自乳腺癌、子宫内膜癌、宫颈癌、皮肤癌、前列腺癌、卵巢癌、输卵管肿瘤、卵巢瘤、血友病和白血病。
  16. 根据权利要求1~8任一项所述的通式(I)所示的化合物或根据权利要求10所述的药物组合物在制备激动或拮抗MOR受体的药物中用途。
PCT/CN2016/101064 2015-10-15 2016-09-30 氧杂螺环类衍生物、其制备方法及其在医药上的应用 WO2017063509A1 (zh)

Priority Applications (13)

Application Number Priority Date Filing Date Title
CA3000761A CA3000761C (en) 2015-10-15 2016-09-30 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines
ES16854888T ES2772689T3 (es) 2015-10-15 2016-09-30 Derivado de oxa espiro, método de preparación del mismo y aplicaciones del mismo en medicamentos
US15/766,985 US10442793B2 (en) 2015-10-15 2016-09-30 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines
MX2018004175A MX2018004175A (es) 2015-10-15 2016-09-30 Derivado oxa espiro, su metodo de preparacion, y sus aplicaciones en medicinas.
JP2018515975A JP6824502B2 (ja) 2015-10-15 2016-09-30 オキサスピロ誘導体、その製造方法、及び医薬におけるその適用
AU2016339404A AU2016339404B2 (en) 2015-10-15 2016-09-30 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines
CN201680003945.6A CN107001347B (zh) 2015-10-15 2016-09-30 氧杂螺环类衍生物、其制备方法及其在医药上的应用
KR1020187012600A KR20180063251A (ko) 2015-10-15 2016-09-30 옥사 스파이로 유도체, 이의 제조방법, 및 의약에서 이의 응용
DK16854888.1T DK3354649T3 (da) 2015-10-15 2016-09-30 Oxaspiroderivat, fremstillingsmetode derfor og anvendelser deraf i lægemidler
BR112018006448-3A BR112018006448A2 (zh) 2015-10-15 2016-09-30 Oxically spirocyclic derivatives, methods for their preparation and their use in medicine
EP16854888.1A EP3354649B1 (en) 2015-10-15 2016-09-30 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines
RU2018115569A RU2733373C2 (ru) 2015-10-15 2016-09-30 Оксаспиропроизводное, способ его получения и его применения в лекарственных средствах
PL16854888T PL3354649T3 (pl) 2015-10-15 2016-09-30 Pochodna oksaspiranu, sposób jej otrzymywania oraz jej zastosowanie w lekach

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201510665328 2015-10-15
CN201510665328.X 2015-10-15
CN201511032876 2015-12-31
CN201511032876.5 2015-12-31

Publications (1)

Publication Number Publication Date
WO2017063509A1 true WO2017063509A1 (zh) 2017-04-20

Family

ID=58517778

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/101064 WO2017063509A1 (zh) 2015-10-15 2016-09-30 氧杂螺环类衍生物、其制备方法及其在医药上的应用

Country Status (17)

Country Link
US (1) US10442793B2 (zh)
EP (1) EP3354649B1 (zh)
JP (1) JP6824502B2 (zh)
KR (1) KR20180063251A (zh)
CN (1) CN107001347B (zh)
AU (1) AU2016339404B2 (zh)
BR (1) BR112018006448A2 (zh)
CA (1) CA3000761C (zh)
DK (1) DK3354649T3 (zh)
ES (1) ES2772689T3 (zh)
HU (1) HUE048032T2 (zh)
MX (1) MX2018004175A (zh)
PL (1) PL3354649T3 (zh)
PT (1) PT3354649T (zh)
RU (1) RU2733373C2 (zh)
TW (1) TWI727981B (zh)
WO (1) WO2017063509A1 (zh)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108424372A (zh) * 2018-05-11 2018-08-21 浙江华贝药业有限责任公司 2,2,2-三氟-n-[(s)-4-羰基四氢萘-1-基]-乙酰胺的纯化工艺
WO2018188641A1 (zh) 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 一种mor激动剂与kor激动剂的药物组合物及其用途
WO2018188643A1 (zh) 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的盐、其富马酸盐i晶型及制备方法
CN108707086A (zh) * 2018-05-15 2018-10-26 浙江华贝药业有限责任公司 一种(1s,4s)-n-(4-羟基四氢萘-1-基)叔丁氧基碳酰胺的纯化工艺
CN108727347A (zh) * 2017-04-14 2018-11-02 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的晶型及其制备方法
CN109206417A (zh) * 2017-07-04 2019-01-15 四川海思科制药有限公司 阿片受体激动剂及其应用
WO2019052557A1 (zh) 2017-09-18 2019-03-21 上海华汇拓医药科技有限公司 μ-阿片受体激动剂及其制备方法和在医药领域的应用
WO2019062804A1 (zh) 2017-09-28 2019-04-04 江苏恒瑞医药股份有限公司 一种氧杂螺环类衍生物的制备方法及其中间体
WO2019072235A1 (zh) * 2017-10-13 2019-04-18 南京明德新药研发股份有限公司 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
WO2019109937A1 (zh) 2017-12-06 2019-06-13 江苏恒瑞医药股份有限公司 Kor激动剂与mor激动剂联合在制备治疗疼痛的药物中的用途
WO2019205983A1 (zh) * 2018-04-28 2019-10-31 四川科伦博泰生物医药股份有限公司 氧杂螺环类化合物及其制备方法和用途
WO2019218953A1 (zh) * 2018-05-14 2019-11-21 江苏恒瑞医药股份有限公司 一种mor受体激动剂药物组合物
WO2020073985A1 (zh) * 2018-10-12 2020-04-16 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
WO2020073984A1 (zh) * 2018-10-12 2020-04-16 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
WO2020147848A1 (zh) * 2019-01-17 2020-07-23 上海海雁医药科技有限公司 三环取代的氧杂螺环衍生物、其制法与医药上的用途
CN111662284A (zh) * 2019-03-06 2020-09-15 上海海雁医药科技有限公司 双杂环取代的氧杂螺环衍生物、其制法与医药上的用途
WO2021027304A1 (zh) * 2019-08-14 2021-02-18 上海海雁医药科技有限公司 镇痛化合物、其制法与医药上的用途
WO2021143801A1 (zh) 2020-01-17 2021-07-22 上海海雁医药科技有限公司 氮杂双环取代的氧杂螺环衍生物、其制法与医药上的用途
WO2021143803A1 (zh) 2020-01-17 2021-07-22 上海海雁医药科技有限公司 光学纯的氧杂螺环取代的吡咯并吡唑衍生物、其制法与医药上的用途
WO2021147993A1 (zh) 2020-01-22 2021-07-29 江苏恒瑞医药股份有限公司 抗trop-2抗体-依喜替康类似物偶联物及其医药用途
CN113214264A (zh) * 2020-01-21 2021-08-06 上海海雁医药科技有限公司 二氢吡咯并五元杂芳基取代的氧杂螺环衍生物、其制法与医药上的用途
WO2022143715A1 (zh) 2020-12-29 2022-07-07 上海海雁医药科技有限公司 氧杂螺环取代的吡咯并吡唑衍生物及其中间体和制备方法
RU2776842C2 (ru) * 2017-12-06 2022-07-27 Цзянсу Хэнжуй Медсин Ко., Лтд. Применение агониста kor в комбинации с агонистом mor для получения лекарственного средства для лечения боли
WO2023284788A1 (zh) 2021-07-13 2023-01-19 上海海雁医药科技有限公司 Mor受体激动剂的药学上可接受的盐、其多晶型物及其用途
WO2023138603A1 (zh) * 2022-01-19 2023-07-27 天地恒一制药股份有限公司 阿片受体激动剂及其制备方法和用途

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114539257B (zh) * 2020-11-24 2023-08-11 江苏恒瑞医药股份有限公司 嘧啶二酮类螺环衍生物、其制备方法及其在医药上的应用
TW202321258A (zh) * 2021-08-02 2023-06-01 大陸商上海樞境生物科技有限公司 氧雜螺環類衍生物、製備方法及其用途
CN116589451A (zh) * 2022-02-11 2023-08-15 成都苑东生物制药股份有限公司 一种6-氧杂螺[4,5]癸烷类化合物的新晶型、用途及其制备方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702561A (zh) * 2011-03-23 2014-04-02 特维娜有限公司 阿片样物质受体配体以及使用和制备其的方法

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2160534B1 (es) 1999-12-30 2002-04-16 Vita Invest Sa Nuevos esteres derivados de (rr,ss)-2-hidroxibenzoato de 3-(2-dimetilaminometil-1-hidroxiciclohexil) fenilo.
DE10044649A1 (de) 2000-09-08 2002-07-04 Gruenenthal Gmbh Substituierte 4-Phenyl-1-(1-phenyl-cyclohexyl)-1,2,3,6-tetrahydropyridine
DE102006033109A1 (de) * 2006-07-18 2008-01-31 Grünenthal GmbH Substituierte Heteroaryl-Derivate
WO2009018169A1 (en) 2007-07-27 2009-02-05 Auspex Pharmaceuticals, Inc. Substituted cyclohexanols
WO2010051476A1 (en) 2008-10-31 2010-05-06 Pain Therapeutics, Inc. Filamin a-binding anti-inflammatory analgesic
WO2010148191A2 (en) 2009-06-17 2010-12-23 Board Of Regents, The University Of Texas System Compositions and methods for cyclofructans as separation agents
PL2797925T3 (pl) 2011-12-12 2016-07-29 Gruenenthal Gmbh Sposób wytwarzania (1R,4R)-6'-fluoro-(N,N-dimetylo- i n-metylo)-4-fenylo-4',9'-dihydro-3'H-spiro[cykloheksano-1,1-pirano-[3,4,b]indolo]-4-aminy
US8916707B2 (en) 2012-08-03 2014-12-23 Johnson Matthey Public Limited Company Process
HUE031557T2 (en) 2012-11-13 2017-07-28 Array Biopharma Inc Bicyclic urea, thiourea, guanidine, and cyanoguadinine compounds used to treat pain

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702561A (zh) * 2011-03-23 2014-04-02 特维娜有限公司 阿片样物质受体配体以及使用和制备其的方法

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHEN, XIAOTAI ET AL.: "Structure Activity Relationships and Discovery of a G Protein Biased u Opioid Receptor Ligand,[(3-Methoxythiophen-2-yl)methyl]( 2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro-[4.5]decan-9- yl]ethyl} )amine(TRV130), for the treatment of Acute Severe Pain", JOURNAL OF MEDICINAL CHEMISTRY, vol. 56, no. 20, 24 September 2013 (2013-09-24), pages 8019 - 8031, XP055375432 *

Cited By (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018188641A1 (zh) 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 一种mor激动剂与kor激动剂的药物组合物及其用途
WO2018188643A1 (zh) 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的盐、其富马酸盐i晶型及制备方法
CN110049980B (zh) * 2017-04-14 2020-10-20 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的盐、其富马酸盐i晶型及制备方法
CN108727347A (zh) * 2017-04-14 2018-11-02 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的晶型及其制备方法
US11014914B2 (en) 2017-04-14 2021-05-25 Jiangsu Hengrui Medicine Co., Ltd. Opioid receptor (MOR) agonist salt, fumarate salt crystal form I thereof and preparation method thereof
AU2018251119B2 (en) * 2017-04-14 2021-07-29 Jiangsu Hengrui Medicine Co., Ltd. Opioid receptor (MOR) agonist salt, fumarate salt I crystal form thereof and preparation method thereof
CN109982699A (zh) * 2017-04-14 2019-07-05 江苏恒瑞医药股份有限公司 一种mor激动剂与kor激动剂的药物组合物及其用途
CN110049980A (zh) * 2017-04-14 2019-07-23 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的盐、其富马酸盐i晶型及制备方法
CN109206417A (zh) * 2017-07-04 2019-01-15 四川海思科制药有限公司 阿片受体激动剂及其应用
CN109206417B (zh) * 2017-07-04 2023-01-03 四川海思科制药有限公司 阿片受体激动剂及其应用
US11072601B2 (en) 2017-09-18 2021-07-27 Shanghai Synergy Pharmaceutical Sciences Co., Ltd. μ-opioid receptor agonist and preparation method therefor and use thereof in field of medicine
WO2019052557A1 (zh) 2017-09-18 2019-03-21 上海华汇拓医药科技有限公司 μ-阿片受体激动剂及其制备方法和在医药领域的应用
US11111236B2 (en) 2017-09-28 2021-09-07 Jiangsu Hengrui Medicine Co., Ltd. Method for preparing oxaspirocycle derivative, and intermediate thereof
WO2019062804A1 (zh) 2017-09-28 2019-04-04 江苏恒瑞医药股份有限公司 一种氧杂螺环类衍生物的制备方法及其中间体
CN110678453A (zh) * 2017-09-28 2020-01-10 江苏恒瑞医药股份有限公司 一种氧杂螺环类衍生物的制备方法及其中间体
TWI690524B (zh) * 2017-09-28 2020-04-11 大陸商江蘇恒瑞醫藥股份有限公司 氧雜螺環類衍生物的製備方法及其中間產物
JP2020535177A (ja) * 2017-09-28 2020-12-03 江▲蘇▼恒瑞医▲薬▼股▲フン▼有限公司Jiangsu Hengrui Medicine Co., Ltd. オキサスピロサイクル誘導体の調製方法およびその中間体
CN110678453B (zh) * 2017-09-28 2023-03-10 江苏恒瑞医药股份有限公司 一种氧杂螺环类衍生物的制备方法及其中间体
RU2777983C2 (ru) * 2017-09-28 2022-08-12 Цзянсу Хэнжуй Медисин Ко., Лтд. Способ получения оксаспироциклического производного и его промежуточного соединения
EP3689859A4 (en) * 2017-09-28 2021-08-25 Jiangsu Hengrui Medicine Co., Ltd. PROCESS FOR MANUFACTURING OXASPIROCYCLE DERIVATIVES AND INTERMEDIATE THEREOF
CN115093428B (zh) * 2017-10-13 2023-09-22 山东绿叶制药有限公司 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
CN111148515B (zh) * 2017-10-13 2022-07-19 山东绿叶制药有限公司 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
WO2019072235A1 (zh) * 2017-10-13 2019-04-18 南京明德新药研发股份有限公司 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
CN111148515A (zh) * 2017-10-13 2020-05-12 山东绿叶制药有限公司 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
US11124523B2 (en) 2017-10-13 2021-09-21 Shandong Luye Pharmaceutical Co., Ltd. 2,6-dioxaspiro [4,5] decane derivatives and preparation method therefor and pharmaceutical applications thereof
CN115093428A (zh) * 2017-10-13 2022-09-23 山东绿叶制药有限公司 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
RU2776842C2 (ru) * 2017-12-06 2022-07-27 Цзянсу Хэнжуй Медсин Ко., Лтд. Применение агониста kor в комбинации с агонистом mor для получения лекарственного средства для лечения боли
US11471503B2 (en) 2017-12-06 2022-10-18 Jiangsu Hengrui Medicine Co., Ltd. Use of KOR agonist in combination with MOR agonist in preparing drug for treating pain
JP2021505547A (ja) * 2017-12-06 2021-02-18 江蘇恒瑞医薬股▲ふん▼有限公司 疼痛を治療するための薬物の製造におけるmorアゴニストと組み合わせたkorアゴニストの使用
WO2019109937A1 (zh) 2017-12-06 2019-06-13 江苏恒瑞医药股份有限公司 Kor激动剂与mor激动剂联合在制备治疗疼痛的药物中的用途
CN111065390A (zh) * 2017-12-06 2020-04-24 江苏恒瑞医药股份有限公司 Kor激动剂与mor激动剂联合在制备治疗疼痛的药物中的用途
CN111836807A (zh) * 2018-04-28 2020-10-27 四川科伦博泰生物医药股份有限公司 氧杂螺环类化合物及其制备方法和用途
WO2019205983A1 (zh) * 2018-04-28 2019-10-31 四川科伦博泰生物医药股份有限公司 氧杂螺环类化合物及其制备方法和用途
CN108424372A (zh) * 2018-05-11 2018-08-21 浙江华贝药业有限责任公司 2,2,2-三氟-n-[(s)-4-羰基四氢萘-1-基]-乙酰胺的纯化工艺
CN108424372B (zh) * 2018-05-11 2021-08-10 浙江华贝药业有限责任公司 2,2,2-三氟-n-[(s)-4-羰基四氢萘-1-基]-乙酰胺的纯化工艺
CN111163778A (zh) * 2018-05-14 2020-05-15 江苏恒瑞医药股份有限公司 一种mor受体激动剂药物组合物
WO2019218953A1 (zh) * 2018-05-14 2019-11-21 江苏恒瑞医药股份有限公司 一种mor受体激动剂药物组合物
CN111163778B (zh) * 2018-05-14 2023-01-24 江苏恒瑞医药股份有限公司 一种mor受体激动剂药物组合物
CN108707086B (zh) * 2018-05-15 2021-08-10 浙江华贝药业有限责任公司 一种(1s,4s)-n-(4-羟基四氢萘-1-基)叔丁氧基碳酰胺的纯化工艺
CN108707086A (zh) * 2018-05-15 2018-10-26 浙江华贝药业有限责任公司 一种(1s,4s)-n-(4-羟基四氢萘-1-基)叔丁氧基碳酰胺的纯化工艺
CN112638906B (zh) * 2018-10-12 2022-05-27 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
CN112638906A (zh) * 2018-10-12 2021-04-09 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
WO2020073985A1 (zh) * 2018-10-12 2020-04-16 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
WO2020073984A1 (zh) * 2018-10-12 2020-04-16 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
CN112638907B (zh) * 2018-10-12 2022-04-12 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
CN112638907A (zh) * 2018-10-12 2021-04-09 江苏恒瑞医药股份有限公司 一种阿片样物质受体(mor)激动剂的结晶形式及制备方法
CN112334465B (zh) * 2019-01-17 2024-06-11 上海海雁医药科技有限公司 三环取代的氧杂螺环衍生物、其制法与医药上的用途
WO2020147848A1 (zh) * 2019-01-17 2020-07-23 上海海雁医药科技有限公司 三环取代的氧杂螺环衍生物、其制法与医药上的用途
CN112334465A (zh) * 2019-01-17 2021-02-05 上海海雁医药科技有限公司 三环取代的氧杂螺环衍生物、其制法与医药上的用途
CN111662284A (zh) * 2019-03-06 2020-09-15 上海海雁医药科技有限公司 双杂环取代的氧杂螺环衍生物、其制法与医药上的用途
CN111662284B (zh) * 2019-03-06 2021-08-10 上海海雁医药科技有限公司 双杂环取代的氧杂螺环衍生物、其制法与医药上的用途
WO2021027304A1 (zh) * 2019-08-14 2021-02-18 上海海雁医药科技有限公司 镇痛化合物、其制法与医药上的用途
CN112789276A (zh) * 2019-08-14 2021-05-11 上海海雁医药科技有限公司 镇痛化合物、其制法与医药上的用途
CN113412263A (zh) * 2020-01-17 2021-09-17 上海海雁医药科技有限公司 氮杂双环取代的氧杂螺环衍生物、其制法与医药上的用途
RU2800295C1 (ru) * 2020-01-17 2023-07-19 Шанхай Хайян Фармасьютикал Текнолоджи Ко., Лтд. Оптически чистое оксаспиро-замещенное производное пирролопиразола, способ его получения и его фармацевтическое применение
CN114269751A (zh) * 2020-01-17 2022-04-01 上海海雁医药科技有限公司 光学纯的氧杂螺环取代的吡咯并吡唑衍生物、其制法与医药上的用途
JP7481458B2 (ja) 2020-01-17 2024-05-10 上海海雁医薬科技有限公司 光学的に純粋なオキサスピロ置換ピロロピラゾール誘導体、その調製方法及び医学的使用
WO2021143803A1 (zh) 2020-01-17 2021-07-22 上海海雁医药科技有限公司 光学纯的氧杂螺环取代的吡咯并吡唑衍生物、其制法与医药上的用途
CN114269751B (zh) * 2020-01-17 2024-04-16 上海海雁医药科技有限公司 光学纯的氧杂螺环取代的吡咯并吡唑衍生物、其制法与医药上的用途
RU2800296C1 (ru) * 2020-01-17 2023-07-19 Шанхай Хайян Фармасьютикал Текнолоджи Ко., Лтд. Оксаспиро-производное, замещенное по азабициклическому кольцу, способ его получения и его медицинское применение
WO2021143801A1 (zh) 2020-01-17 2021-07-22 上海海雁医药科技有限公司 氮杂双环取代的氧杂螺环衍生物、其制法与医药上的用途
AU2021208113B2 (en) * 2020-01-17 2023-11-30 Shanghai Haiyan Pharmaceutical Technology Co., Ltd. Azabicyclic substituted oxaspiro derivative, preparation method therefor and medical use thereof
CN113214264A (zh) * 2020-01-21 2021-08-06 上海海雁医药科技有限公司 二氢吡咯并五元杂芳基取代的氧杂螺环衍生物、其制法与医药上的用途
WO2021147993A1 (zh) 2020-01-22 2021-07-29 江苏恒瑞医药股份有限公司 抗trop-2抗体-依喜替康类似物偶联物及其医药用途
WO2022143715A1 (zh) 2020-12-29 2022-07-07 上海海雁医药科技有限公司 氧杂螺环取代的吡咯并吡唑衍生物及其中间体和制备方法
WO2023284788A1 (zh) 2021-07-13 2023-01-19 上海海雁医药科技有限公司 Mor受体激动剂的药学上可接受的盐、其多晶型物及其用途
WO2023138603A1 (zh) * 2022-01-19 2023-07-27 天地恒一制药股份有限公司 阿片受体激动剂及其制备方法和用途

Also Published As

Publication number Publication date
US10442793B2 (en) 2019-10-15
HUE048032T2 (hu) 2020-05-28
MX2018004175A (es) 2018-06-06
AU2016339404B2 (en) 2020-08-27
CN107001347A (zh) 2017-08-01
PL3354649T3 (pl) 2020-07-27
ES2772689T3 (es) 2020-07-08
TWI727981B (zh) 2021-05-21
KR20180063251A (ko) 2018-06-11
CN107001347B (zh) 2019-09-24
JP6824502B2 (ja) 2021-02-03
BR112018006448A2 (zh) 2018-10-16
AU2016339404A1 (en) 2018-05-24
CA3000761C (en) 2024-02-13
EP3354649B1 (en) 2019-12-04
JP2018534257A (ja) 2018-11-22
PT3354649T (pt) 2020-02-03
RU2018115569A (ru) 2019-11-18
DK3354649T3 (da) 2020-02-24
RU2733373C2 (ru) 2020-10-01
TW201718558A (zh) 2017-06-01
CA3000761A1 (en) 2017-04-20
RU2018115569A3 (zh) 2020-01-20
EP3354649A4 (en) 2018-08-08
US20180297988A1 (en) 2018-10-18
EP3354649A1 (en) 2018-08-01

Similar Documents

Publication Publication Date Title
TWI727981B (zh) 氧雜螺環類衍生物、其製備方法及其在醫藥上的應用
TWI601728B (zh) 吡咯并六員雜芳環類衍生物、其製備方法及其在醫藥上的應用
KR102204804B1 (ko) 디히드로피라졸 gpr40 조절제
KR101623357B1 (ko) 5-ht6 길항제로서 아릴설포닐 피라졸린 카복스아미딘 유도체
CN113164409A (zh) 具有雌激素受体α降解活性的新型化合物及其用途
CN106715435B (zh) 作为nr2b nmda受体拮抗剂的吡咯并嘧啶衍生物
CN108495618B (zh) 新的化合物及其用途
CN101602741A (zh) 作为γ-分泌酶抑制剂的取代的N-芳基磺酰基杂环胺
WO2022268230A1 (zh) 作为kif18a抑制剂的化合物
TW200914442A (en) Gamma secretase modulators
CN108602778A (zh) 8-氨基-2-氧代-1,3-二氮杂-螺-[4.5]-癸烷衍生物
TW201605838A (zh) 晶形
TW202140495A (zh) 三環四氫異喹啉類衍生物、其製備方法及其在醫藥上的應用
KR20120123615A (ko) 퀴놀론 화합물 및 제약 조성물
CN107207476A (zh) 吲哚和氮杂吲哚衍生物及其用于神经退化性疾病中的用途
Kopp et al. Novel σ1 antagonists designed for tumor therapy: Structure–activity relationships of aminoethyl substituted cyclohexanes
JP2022537330A (ja) インダゾール誘導体、その製造方法及び医薬上のその使用
JP2018515499A (ja) Gpr55受容体の活性の選択的モジュレーター:クロメノピラゾール誘導体
JP5251885B2 (ja) 縮合インダン化合物
JP2017533929A (ja) プロキネチシン介在消化器疾患を治療するためのスルホニルピペリジン誘導体及びその使用
WO2023046214A1 (zh) 一种受体相互作用蛋白激酶1的抑制剂及其制备方法、应用
EP2338485A1 (de) Substituierte 1,3-Dioxoisoindoline als Arzneimittel
WO2024002375A1 (zh) 一种用作smarca2/4抑制剂的化合物及其应用
WO2024067819A1 (zh) 含哌啶多环类衍生物调节剂、其制备方法和应用
WO2013058361A1 (ja) アンドロゲン受容体拮抗化合物の結晶

Legal Events

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

Ref document number: 16854888

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2018515975

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 3000761

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2018/004175

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 15766985

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018006448

Country of ref document: BR

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2016854888

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20187012600

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2018115569

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2016339404

Country of ref document: AU

Date of ref document: 20160930

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112018006448

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180329