WO2023138603A1 - 阿片受体激动剂及其制备方法和用途 - Google Patents

阿片受体激动剂及其制备方法和用途 Download PDF

Info

Publication number
WO2023138603A1
WO2023138603A1 PCT/CN2023/072810 CN2023072810W WO2023138603A1 WO 2023138603 A1 WO2023138603 A1 WO 2023138603A1 CN 2023072810 W CN2023072810 W CN 2023072810W WO 2023138603 A1 WO2023138603 A1 WO 2023138603A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
membered
alkyl
pharmaceutically acceptable
pain
Prior art date
Application number
PCT/CN2023/072810
Other languages
English (en)
French (fr)
Inventor
罗明
聂岳坤
李政
喻龙江
肖文喜
刘军华
Original Assignee
天地恒一制药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 天地恒一制药股份有限公司 filed Critical 天地恒一制药股份有限公司
Priority to CN202380008359.0A priority Critical patent/CN116801881A/zh
Publication of WO2023138603A1 publication Critical patent/WO2023138603A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/113Spiro-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D495/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention belongs to the field of medicinal chemistry, and specifically relates to a class of oxaspiro ring-type small molecule compounds, a preparation method thereof, a pharmaceutical composition containing the compound and its use as a therapeutic agent, especially as a MOR receptor agonist and in the preparation of medicines for treating and preventing pain and other related diseases.
  • Opioid receptor is a G protein-coupled receptor (G Protein-Coupled Receptor, GPCR), which is the binding target of endogenous opioid peptides and opioids.
  • GPCR G protein-Coupled Receptor
  • opioid receptors There are a variety of opioid receptors in the human body, mainly including three types of mu opioid receptor (MOR), delta opioid receptor (DOR) and kappa opoid receptor (Kappa opoid receptor, KOR), which are widely distributed in the central nervous system, heart, digestive tract, blood vessels, kidneys and other peripheral tissues (Nature, 2016, 537(7619): 185) .
  • MOR has the strongest binding ability to morphine, and it is the receptor protein site for the main action of analgesics such as morphine and fentanyl.
  • Zadina studies of Zadina have found that the binding ability (360pm) of MOR receptor and morphin 1 is 4000 times and 15,000 times the combination of DOR receptors, KOR receptors and morphine 1 (Science 2001 VOL.293 NO: 311-315; Life SCI 61: 409-415).
  • the G protein signaling pathway mainly includes second messenger systems such as calcium ions, adenylate cyclase, and mitogen-activated protein kinase.
  • the ⁇ -arrestin pathway has three main aspects: (1) acting as a negative regulatory factor and interacting with GPCR kinases to cause receptor desensitization of GPCRs to terminate G protein signal transduction; (2) recruiting endocytic proteins as a scaffolding protein to induce GPCR endocytosis; (3) serving as an adapter protein to form a complex with GPCR downstream signaling molecules to activate signal transduction molecules in a G protein-independent manner.
  • a "biased" MOR agonist drug that can selectively activate the G protein signaling pathway that is, the negative ⁇ -arrestin-biased ligand design drug of MOR, can reduce the side effects mediated by ⁇ -arrestin, which has significant clinical value and social significance in the field of analgesia.
  • the present invention provides a compound with novel structure that can be used as MOR receptor agonist.
  • This type of compound shows high activity, E max is also significantly improved, and has higher selectivity to MOR.
  • the first aspect of the present invention provides a compound represented by formula (IV) or (V) or (VI), its solvate, stereoisomer, deuterated compound, or a pharmaceutically acceptable salt thereof,
  • Ring B and Ring C are each independently selected from substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl;
  • Ring D is selected from cycloalkyl, heterocycloalkyl
  • each R is independently selected from deuterium atom, halogen, -OH, -C 1-6 alkyl, -C 1-6 alkyl-OC 1-6 alkyl, -OC 1-6 alkyl, 3 to 6 membered cycloalkyl, -OC 2-6 alkynyl, -OC 2-6 alkenyl, -C 2-6 alkynyl, -C 2-6 alkenyl , amino, carboxylate, nitro, cyano, hydroxyalkyl, heterocyclyl, aryl and heteroaryl;
  • the g is selected from 0, 1, 2, 3, 4, 5, 6;
  • the f is selected from 0, 1, 2;
  • p, q, L are each independently 0, 1, 2, 3 or 4;
  • heteroaryl group, the heterocycloalkyl group or the heteroatoms on the heterocyclyl group are each independently selected from O, S or N.
  • the compound is selected from formula (VII) or (VIII) or (IX) or (X) or (XI) or (XII):
  • the compound is selected from the following structures:
  • the compound is selected from the following structures:
  • the compound is selected from the following structures:
  • the compound is selected from the following structures:
  • q is selected from 1, 2, 3 or 4.
  • the compound is selected from:
  • the second aspect of the present invention relates to a pharmaceutical composition, which includes the compound shown in the first aspect of the present invention, its solvate, stereoisomer body, a deuterated compound or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the third aspect of the present invention provides the use of the compound shown in the first aspect of the present invention, its solvate, stereoisomer, deuterated compound or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition described in the second aspect of the present invention in the preparation of medicines for preventing and/or treating related diseases mediated by MOR receptor agonists.
  • the related diseases mediated by MOR receptor agonists are selected from pain, immune dysfunction, inflammation, esophageal reflux, neurological and mental diseases, urinary and reproductive diseases, cardiovascular diseases and respiratory diseases; preferably, the pain is selected from postoperative pain, pain caused by cancer, neuropathic pain, traumatic pain and pain caused by inflammation.
  • C2-6 alkynyl used in the present invention refers to a straight-chain or branched alkyne group derived from an alkyne moiety of 2 to 6 carbon atoms containing a carbon-carbon triple bond, such as ethynyl, propynyl, 2-butynyl, 2-pentynyl, 3-pentynyl, 4-methyl-2-pentynyl, 2-hexynyl, 3-hexynyl, etc.
  • cycloalkyl in the present invention includes all possible monocyclic and condensed rings (including fused in the form of parallel, spiro, and bridge); for example: "3-12 membered cycloalkyl", which can be a monocyclic, bicyclic, or polycyclic cycloalkyl system (also known as a fused ring system).
  • the monocyclic ring system is a cyclohydrocarbyl group containing 3-8 carbon atoms, examples include but are not limited to: cyclopropyl, cyclobutanyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and the like.
  • the fused ring cycloalkyl group includes parallel cycloalkyl group, bridged cycloalkyl group and spirocycloalkyl group.
  • the cyclocycloalkyl can be 6-11 membered cyclocycloalkyl, 7-10 membered cyclocycloalkyl, representative examples of which include, but are not limited to, bicyclo[3.1.1]heptane, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1]nonane, and bicyclo[4.2.1]nonane.
  • Spirocycloalkyl can be 7-12 membered spirocycloalkyl, 7-11 membered spirocycloalkyl, examples include but not limited to: group.
  • the above bridged cycloalkyl group can be 6-11 membered bridged cycloalkyl group, 7-10 membered bridged cycloalkyl group, examples of which include but are not limited to: group.
  • heterocycloalkyl refers to a monovalent monocyclic non-aromatic ring system, whose ring atoms are composed of carbon atoms and heteroatoms selected from nitrogen, oxygen, sulfur and phosphorus, and connected to the parent nucleus or other groups through a single bond; base, tetrahydro-2hydro-pyran-2-yl, tetrahydro-2hydro-pyran-4-yl, piperidin-2-yl, piperidin-4-yl, etc.
  • heterocyclyl refers to a non-aromatic cyclic group in which at least one ring carbon atom of 3-12 members is replaced by a heteroatom selected from O, S, and N, preferably 1-3 heteroatoms, including carbon atoms, nitrogen atoms and sulfur atoms that can be oxo-substituted.
  • heteroatom selected from O, S, and N, preferably 1-3 heteroatoms, including carbon atoms, nitrogen atoms and sulfur atoms that can be oxo-substituted.
  • 3-12 membered heterocyclyl refers to monocyclic heterocyclyl, bicyclic heterocyclyl system or polycyclic heterocyclyl system (also known as fused ring system), including saturated and partially saturated heterocyclyl, but excluding aromatic rings. Unless otherwise specified, all monocyclic rings, condensed rings (including fused in the form of parallel, spiro, and bridge), saturated, and partially saturated that may be formed are included.
  • Monoheterocyclyl can be 3-8 membered monoheterocyclyl, 3-6 membered monoheterocyclyl, 4-7 membered monoheterocyclyl, 5-7 membered monoheterocyclyl, 5-6 membered monoheterocyclyl, 5-6 membered oxygen-containing monoheterocyclyl, 3-8 membered nitrogen-containing monoheterocyclyl, 5-6 membered nitrogen-containing monoheterocyclyl, 5-6 membered saturated monoheterocyclyl, etc. Examples include but are not limited to: group.
  • Fused heterocycles include heterocyclyls, spiroheterocyclyls, bridged heterocyclyls and may be saturated, partially saturated or unsaturated, but are not aromatic.
  • the heterocyclyl can be 6-12 membered heterocyclyl, 7-10 membered heterocyclyl, 6-10 membered heterocyclyl, 6-12 membered saturated heterocyclyl, representative examples include but are not limited to: group.
  • the spiroheterocyclyl can be 6-12 membered spiroheterocyclyl, 7-11 membered spiroheterocyclyl, 6-12 membered saturated spiroheterocyclyl, examples include but not limited to: group;
  • the above bridged heterocyclic group can be 6-12 membered bridged heterocyclic group, 7-11 membered bridged heterocyclic group, 6-12 membered bridged heterocyclic group, examples of which include but are not limited to: group.
  • aryl refers to a monovalent monocyclic or polycyclic (including fused form) aromatic ring system, which is composed of only carbon atoms and hydrogen atoms; common aryl groups include (but are not limited to) phenyl, naphthyl, anthracenyl, phenanthrenyl, acenaphthyl, azulenyl, fluorenyl, indenyl, pyrenyl, etc.
  • the above-mentioned aryl also includes heterocyclyl and aryl, cycloalkyl and aryl;
  • heteroaryl refers to a monovalent monocyclic or polycyclic (including fused forms) aromatic ring system whose ring atoms are composed of carbon atoms and heteroatoms selected from nitrogen, oxygen, sulfur and phosphorus; common heteroaryl groups include, but are not limited to, benzopyrrolyl, benzofuryl, benzothienyl, benzimidazole, benzoxazolyl, benzothiazolyl, azetidinyl, carbazolyl, pyrrolyl, furyl , thienyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, isothiazolyl, indazolyl, indolyl, quinolinyl, isoquinolyl, phenazinyl, phenoxazinyl, phenothiazinyl, pteri
  • the "stereoisomer" of the compound shown in the present invention means that when the compound has an asymmetric carbon atom, enantiomers will be produced; when the compound has a carbon-carbon double bond or a cyclic structure, cis-trans isomers will be produced; when the compound has ketones or oximes, tautomers will be produced, and enantiomers, diastereoisomers, racemic isomers, cis-trans isomers, tautomers, geometric isomers, epimers and mixtures thereof of all compounds are included in the scope of the present invention.
  • room temperature refers to about 20-30°C; "overnight” refers to about 10h to 16h; 1M, 1N is 1mmol/L, 1 ⁇ M is 1 ⁇ mol/L, 1mM is 1mmol/L, and 1nM is 1nmol/L; eq: equivalent;
  • Step 1 Synthesis of 2-9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl)acetic acid (141-2)
  • Step 2 Synthesis of (R)-2-9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl)acetic acid and (S) phenethylamine salt (141-3)
  • Step 3 Synthesis of (R)-2-9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl)acetic acid (141-4)
  • Step 4 Synthesis of nitrogen-methyl-nitrogen-methoxy-(R)-2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decen-9-yl)acetamide (141-5)
  • Step 5 Synthesis of compound nitrogen-methyl-nitrogen-methoxy-(R)-2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decen-9-yl)acetaldehyde (141-6)
  • Step 6 Synthesis of (1R,4R)-4-ethoxy-N- ⁇ 2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-ene-9-e]ethyl ⁇ -1,2,3,4-tetrahydronaphthalene-1-amine (141)
  • Step 1 Synthesis of nitrogen-methyl-nitrogen-methoxy-(R)-2-(4'-(pyridin-2-yl)tetrahydrooxaspiro[bicyclo[3.1.0]hexane-3,2'-pyran]-4'-ylacetamide (143-1)
  • compound 141-5 (590 mg, 1.86 mmol) was dissolved in DCM (15 mL). Under nitrogen protection, 2 mol/L dimethyl zinc (3.7 mL, 7.46 mmol) and diiodomethane (5.0 g, 18.6 mmol) were added. After the addition was complete, the mixture was moved to room temperature and stirred for 16 h.
  • Step 2 Synthesis of (R)-2-(4'-(pyridin-2-yl)tetrahydrooxaspiro[bicyclo[3.1.0]hexane-3,2'-pyran]-4'-ylacetaldehyde (143-2)
  • Step 3 Synthesis of (1R,4R)-4-ethoxy-N- ⁇ 2-[4'-(pyridin-2-yl)spiro[bicyclo[3.1.0]hexane-3,2'-oxane]-4'-yl]ethyl ⁇ -1,2,3,4-tetrahydronaphthalene-1-amine (143)
  • compound A 100 mg, 0.39 mmol
  • compound 9 76 mg, 0.58 mmol
  • dichloroethane DCE, 6 mL
  • TIPT tetraisopropyl titanate
  • the compounds of the present invention can activate mu-opioid receptors (MOR).
  • MOR mu-opioid receptors
  • Activated MOR can regulate the level of intracellular cAMP, cAMP enters the nucleus and binds to the cAMP response element (cAMP response element, CRE) region of the reporter gene Luciferase, and initiates the expression of the reporter gene.
  • Luciferase can emit fluorescence when it reacts with its substrate, and the agonistic activity of the compound can be reflected by measuring the fluorescent signal.
  • Detection buffer 1 ⁇ stimulation buffer, 500 ⁇ M 1-methyl-3-isobutylxanthine (IBMX), ddH 2 O.
  • Compound preparation the compound was dissolved in dimethyl sulfoxide (DMSO) and prepared as a mother solution with a final concentration of 10 mM, diluted to a working concentration of 0.08 mM, and the compound was diluted 4 times with an Echo pipette, with an initial concentration of 0.08 mM, 10 concentration gradients, and 50 nL was added to a 384 cell plate in duplicate, with a final concentration of 0.4 ⁇ M, and then the cell plate was centrifuged at 1000 rpm for 1 min. 50 nL of Forskolin (final concentration 1 ⁇ M) was transferred to the 384 cell plate with an Echo pipette.
  • DMSO dimethyl sulfoxide
  • Cell plating Thaw the frozen cells, centrifuge at 1000 rpm for 5 min, discard the supernatant, wash the cells twice with HBSS buffer, resuspend the cells with detection buffer, adjust the cell density to 5.0 ⁇ 105 /mL, add to a 384-well plate, 10 ⁇ L per well, 5000 cells. Shake for 20s, centrifuge at 1000rpm for 1min, and place the cell plate in a 23°C incubator for 60min.
  • cAMP adenosine-3',5'-cyclic phosphate
  • Detection reagent preparation Dilute Anti cAMP-Cryptate and AMP-d2 to 1 ⁇ with lysis buffer, add 10 ⁇ L of detection reagent to each well according to the microplate layout, shake for 20 s, centrifuge at 1000 rpm for 1 min, put the cell plate in an incubator at 23°C for 60 min; read the plate on an Envision microplate reader.
  • the compound of the present invention stimulates the MOR to affect the change of the downstream cAMP level through the above experiments.
  • the experimental results show that this series of compounds exhibits strong Op-Mu agonistic effect, and the measured EC50 values of typical representative compounds are shown in Table 1.
  • 6 groups of controls are set up, which are compound 83, compound 91, compound 31, TRV130, TRV130 (racemization) and SHR8554, wherein TRV130, TRV130 (racemization) and SHR8554 have the following structural formulas respectively.
  • For the preparation method of TRV130 refer to patent CN103702561A; for the preparation method of SHR8554, refer to patent CN107001347B; Maximum potency for changes in cAMP levels.
  • the preferred compounds of the embodiments of the present invention have obvious agonistic effects on Mu opioid receptors, and the EC 50 and E max of some compounds are far better than those of the control group.
  • ForsKolin forskolin
  • K opioid receptor agonists can inhibit the cAMP release stimulated by forsKolin.
  • the agonistic activity of the compound on the human K sheet receptor can be determined. Firstly, a certain concentration of forsKolin and different concentrations of test compounds are used to incubate with human cell lines highly expressing K opioid receptors. cAMP levels in stimulated OPRK1 cells were determined using a time resolved fluorescence resonance energy transfer (TR-FRET) based cAMP immunoassay (LANCEPerKinElmer). The specific method is as follows:
  • Detection buffer 1 ⁇ stimulation buffer, 500 ⁇ M IBMX, ddH 2 O.
  • Compound preparation the compound was dissolved in DMSO to prepare a mother solution with a final concentration of 10 mM, diluted to a working concentration of 2 mM, and the compound was diluted 4-fold with Echo, with an initial concentration of 2 mM and 10 concentration gradients, and 50 nL was added to a 384 cell plate in double wells with a final concentration of 10 ⁇ M, then the cell plate was centrifuged at 1000 rpm for 1 min; 50 nL of ForsKolin (final concentration was 3 ⁇ M) was transferred to 38 4 cell plate.
  • Cell plating Thaw the frozen cells, centrifuge at 1000 rpm for 5 min, discard the supernatant, wash the cells twice with HBSS buffer, resuspend the cells with detection buffer, adjust the cell density to 3.0 ⁇ 105 cells/mL, add to a 384-well plate, 10 ⁇ L per well, 3000 cells. Shake for 20s, centrifuge at 1000rpm for 1min, and place the cell plate in a 23°C incubator for 60min. Preparation of standard curve: 4-fold gradient dilution of standard cAMP with detection buffer, a total of 8 concentration points, the highest concentration is 800nM, add 10 ⁇ L to each well according to the microplate layout.
  • Detection reagent preparation Dilute the intermediates Anti cAMP-Cryptate and AMP-d2 to 1 ⁇ with lysis buffer, add 10uL detection reagent to each well, shake for 20s, centrifuge at 1000rpm for 1min, and place the cell plate in an incubator at 23°C for 60min. Finally the plate was read on Envision.
  • the Dose-response-Stimulation—log[agonist]vs.response—Variable slope mode was selected for agonist fitting analysis, and the EC 50 value of each test sample was obtained.
  • the experimental data are shown in Table 2.
  • the activity of the compound of the embodiment of the present invention on stimulating the K opioid receptor is significantly weaker; it shows that the compound of the present invention has high selectivity to the MOR receptor, and it is speculated that the compound of the embodiment of the present invention has lower side effects.
  • This study aimed to evaluate the ⁇ -Arrestin recruitment efficiency of agonists targeting the ⁇ -opioid receptor MOR by EC50 and EMAX assays of CHO-K1/Arrestin/hMOR.
  • the CHO-K1/Arrestin/hMOR cell line expresses hMOR fused to a ⁇ -galactosidase donor fragment and ⁇ -Arrestin fused to a ⁇ -galactosidase acceptor fragment. When ⁇ -arrestin interacts with hMOR, these fragments form active ⁇ -galactosidase.
  • the compounds of the examples of the present invention have almost no activation effect on the ⁇ -arrestin signaling pathway, and the compounds of the present invention have better bias (cAMP and ⁇ -arrestin signaling pathway) than the control group. It is speculated that the compounds of the present invention have lower side effects than the control group.
  • CHO-hERG cells Chinese hamster ovary (CHO) cell line, CHO-hERG cells were used in this experiment.
  • the complete medium is F12 medium, supplemented with 10% fetal bovine serum, 1% Selective antibiotic (G418), 89 ⁇ g/mL hygromycin B (HB).
  • the recovery medium is F12 medium supplemented with 10vol% fetal bovine serum.
  • CHO-hERG cells were grown in a high humidity incubator at 37°C ( ⁇ 2°C), 5% CO 2 (4% to 8%). Cells were revived with recovery medium, passaged in complete medium, and cells used for patch clamp experiments were replaced with recovery medium at the last passage.
  • the hERG current was recorded under the whole-cell patch clamp technique, and the recording temperature was room temperature.
  • the output signal of the patch clamp amplifier is converted by digital to analog and filtered by 2.9KHz low pass. Data records were collected with Patchmaster Pro software.
  • the cell species is placed on the inverted microscope stage in the cell recording tank, and a cell in the recording tank is randomly selected for the experiment.
  • the perfusion system was fixed on the inverted microscope stage and cells were continuously perfused with ECS.
  • Microelectrodes for manual patch clamp experiments were prepared from capillary glass tubes filled with intracellular fluid. On the day of the patch clamp test, use borosilicate Electrodes were prepared from glass tubes (BF150-117-10, SUTTER INSTRUMENT USA). After the electrode is filled with ICS, the resistance is between 2-5M ⁇ .
  • the clamping voltage was -80mV, the first step depolarized to +60mV and maintained for 850ms to open the hERG channel. Then, the voltage is set to -50mV and maintained for 1275ms, resulting in rebound current or tail current, the peak value of tail current will be measured and used for analysis. Finally, the voltage returns to the clamping voltage (-80mV). During the test, this command voltage program is repeated every 15s.
  • test articles/positive controls or multiple concentrations of the same drug can be tested on one cell, and the different test articles/positive controls need to be washed with a solvent control working solution until the hERG current returns to more than 80% of the size before adding the drug.
  • the standard deviation of the inhibition rate of each recorded cell under the same concentration is not more than 15%.
  • cisapride was tested at a concentration of 0.1 [mu]M in duplicates of the cells. According to scientific literature reports, cisapride at 0.1 ⁇ M inhibits hERG current by more than 50%. (Milnes, J.T., et al.).
  • a good whole-cell recording should meet the following conditions: path resistance (Rs) less than 10M ⁇ ; membrane resistance (Rm) greater than 500M ⁇ and membrane capacitance (Cm) less than 100pF.
  • Leakage current Under the clamping voltage of -80mV, the absolute value of the leakage current should be less than 200pA. The current amplitude will be corrected for the leakage current at -80mV. Scanning curves whose absolute value of leakage current is greater than 200pA cannot be used for analysis.
  • the percentage inhibition of each concentration of the test product and the positive control is calculated from the recorded current response using the following formula: (1-tail peak current recorded after the test product/positive control perfusion/vehicle control perfusion recorded tail peak current (initial current)) ⁇ 100%.
  • the compounds of the examples of the present invention Compared with the control group, the compounds of the examples of the present invention have higher hERG IC 50 values, with significant difference, showing a weaker inhibitory effect on hERG, indicating that the compounds of the present invention have a lower risk of cardiotoxicity.
  • the compound under study was administered orally or intravenously (vehicle 5vol% DMSO + 10vol% Solutol (HS-15) + 85vol% saline) to animals (such as mice, rats, dogs or monkeys), and blood was collected at fixed time points. Immediately after blood sample collection, gently invert the tube at least 5 times to ensure thorough mixing and place on ice. The blood was anticoagulated with heparin, and then centrifuged at 8000rpm for 5 minutes to separate the serum from the red blood cells. Aspirate the serum with a pipette and transfer it to a 2mL polypropylene tube, mark the name and time point of the compound, and store it in a -40°C refrigerator before LC-MS analysis for testing.
  • animals such as mice, rats, dogs or monkeys
  • the compounds of the examples of the present invention exhibited good pharmacokinetic properties in rats; compared with the control group, the AUC (h*ng/mL) of the compounds of the present invention in plasma free base was significantly improved.
  • D0 For female SD rats, the date when the rats started training was recorded as D0. At D0, set the temperature of the analgesia instrument to 52°C (52.0 ⁇ 0.5°C), put the rat on the hot plate and time it at the same time, and record the time (s) it takes for the rat to lick the hind paw or jump to the pain threshold. If the rat did not experience thermal pain response on the hot plate for more than 30s, the rat was taken out immediately, and the pain threshold was recorded as 30s. Responsive and unresponsive rats were eliminated.
  • the pre-screened rats were taken, and the temperature of the analgesia instrument was set to 52°C (52.0 ⁇ 0.5°C), the rats were placed on a hot plate and timed, and the time (s) taken for the rats to lick their hind feet or jump the pain threshold was recorded, and measured three times in total. The average value of three times was used as the baseline pain threshold of rats.
  • rats were randomly divided into groups according to the baseline pain threshold, with 8 animals in each group.
  • the dosage group in the table give vehicle or compound by tail vein injection mode (each group is administered in equimolar dosage, The vehicle is 5vol% DMSO+10vol% Solutol (HS-15)+85vol% saline).
  • the temperature of the analgesia instrument was set to 52°C (52.0 ⁇ 0.5°C), the rats were placed on a hot plate and timed, and the time (s) taken for the rats to lick their hind feet or jump when the pain threshold took place was recorded, and each time point was measured once. If the rat did not experience thermal pain response on the hot plate for more than 30s, the rat was taken out immediately, and the pain threshold was recorded as 30s. Calculate the pain threshold %MPE at each time point of each group, and evaluate the analgesic efficacy of each test sample in vivo.
  • the experimental data were expressed as Mean ⁇ SEM, and the data among the groups were analyzed by analysis of variance (ANOVA) test (Two Way ANOVA or One-Way ANOVA) using GraphPad Prism, and P ⁇ 0.05 was considered to have a significant difference.
  • the compound of the embodiment of the present invention exhibits better analgesic effect than the control group, and shows a higher pain threshold and longer-lasting analgesic effect in the rat hot plate analgesic efficacy study.
  • a single intravenous administration of the compound (vehicle 5vol% DMSO+10vol% Solutol (HS-15)+85vol% saline) was given to SD rats (4-6 dose groups were set up for each compound, 10 rats in each dose group, half and half female), and clinical observation was performed after administration. Clinical observation twice a day on the first day, once a day from the second day, for 14 consecutive days. Including behavioral observations, autonomic and neurological behaviors and death conditions, etc., to obtain the maximum tolerance of the compound dose (MTD value) and median lethal dose ( LD50 value).
  • the experimental results show that the MTD value and LD 50 value of the compound of the present invention in a single intravenous administration to rats are significantly improved compared with the control group, indicating that the compound of the present invention has good safety.

Landscapes

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

Abstract

本发明提供了一类氧杂螺环类小分子化合物,以及含有该化合物的药物组合物及其作为治疗剂,特别是作为MOR受体激动剂和在制备治疗和/或预防疼痛等相关疾病的药物中的用途。本发明提供的一种结构新颖的MOR受体激动剂,表现出高活性,以及对MOR具有较高的选择性,并且其最大效能Emax也具有明显改善。

Description

阿片受体激动剂及其制备方法和用途
本申请要求于2022年1月19日提交中国专利局、申请号为202210057938.1发明名称为“阿片受体激动剂及其制备方法和用途”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。
技术领域
本发明属于药物化学领域,具体涉及一类氧杂螺环类小分子化合物、其制备方法及含有该化合物的药物组合物以及其作为治疗剂,特别是作为MOR受体激动剂和在制备治疗和预防疼痛等相关疾病的药物中的用途。
背景技术
阿片受体是一种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。人体内存在多种阿片受体,主要包括μ阿片受体(Mu opioid receptor,MOR)、δ阿片受体(Delta Opioid Receptor,DOR)和κ阿片受体(Kappa opoid receptor,KOR)三类,广泛分布在中枢神经系统、心脏、消化道、血管、肾脏等外周组织(Nature,2016,537(7619):185)。MOR与吗啡肽的结合能力最强,是吗啡、芬太尼等镇痛药主要作用的受体蛋白位点。Zadina等研究发现MOR受体与吗啡肽1的结合能力(360pM)是DOR受体、KOR受体与吗啡肽1结合力的4000倍和15000倍(Science 2001 Vol.293 No:311-315;Biochem Biophys Res Commun 235:567-570;Life Sci 61:409-415)。
研究发现GPCR介导及调控生理功能主要经由激活G蛋白途径和β-arrestin途径。G蛋白信号途径主要包括钙离子等第二信使系统、腺苷酸环化酶、丝裂原活化蛋白激酶等。β-arrestin途径主要有三个方面:(1)作为负性调控因子与GPCR激酶作用使GPCRs发生受体脱敏反应从而中止G蛋白信号转导;(2)作为支架蛋白募集胞吞蛋白诱导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。早期研究显示,内源性脑啡肽和阿片类药物埃托啡可以激动G蛋白并引发受体内吞,而吗啡则不引发受体内吞,这是因为吗啡通过G蛋白信号通路而不是β-arrestin途径来发挥其生理功能(Zhang等,Proc Natl Acad Sci USA,1998,95(12):7157-7162)。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)。由此可见,配体刺激G蛋白和/或β-arrestin信号的差异决定了GPCR的配体特异性细胞生物学效应,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G蛋白信号传递时间延长,镇痛作用更强。近年来研究发现,β-arrestin通路与MOR激动剂多个副作用相关,如便秘、呼吸抑制和镇痛耐受(Science 1999 Vol.286:2495-2498:J.Pharmacol.Exp.Ther.2005,314:1195-1201)。因此,研发一种可选择性激活G蛋白信号通路的“偏向性”MOR激动剂药物,即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,在镇痛领域具有显著的临床价值和社会意义。
FDA于2020年8月批准了Trevena Inc公司药物Olinvyk(WO2012129495)的上市申请,目前关于G蛋白偏向性MOR激动剂的研发报道的专利有WO2017063509A1、WO2019205983A1、CN10920641A、WO2019072235A1、CN111662284A、WO2019052557A1等,虽然这些专利已经公开了一系列G蛋白偏向性MOR激动剂,但其分子结构与本发明提供的结构具有较大区别,且其药效、安全性暂未得到证实,临床上仍需要开发新的分子结构,以获得具有更好的药效、选择性、药用安全性、药物代谢结果的MOR激动剂。
发明内容
针对现有技术的需求,本发明提供了一种结构新颖的可作为MOR受体激动剂的化合物,该类化合物表现出高活性,Emax也具有明显改善,以及对MOR较高的选择性。
本发明第一方面提供了如式(Ⅳ)或(Ⅴ)或(Ⅵ)所示的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,
其中,环B、环C各自独立的选自被取代的或未被取代的芳基、被取代的或未被取代的杂芳基;
环D选自环烷基、杂环烷基;
R4、R5各自独立的选自H、氘原子、烷基、氧代、烷氧基、羟基、卤素、氰基、炔基、烯基、-(CH2)g-O-3至12元杂环基、-(CH2)g-O-3至12元环烷基、-(CH2)g-3至12元环烷基、-(CH2)g-3至12元杂环基、5至10元杂芳基、5至10元芳基、-S(=O)f-C1-6烷基、-O-C2-6炔基、-O-C2-6烯基;其中,所述杂环基、杂芳基、芳基、烷基、炔基、烯基、烷氧基可任选进一步被1至3个R6所取代;
其中,R6各自独立的选自氘原子、卤素、-OH、-C1-6烷基、-C1-6烷基-O-C1-6烷基、-O-C1-6烷基、3至6元环烷基、-O-C2-6炔基、-O-C2-6烯基、-C2-6炔基、-C2-6烯基、氨基、羧酸酯基、硝基、氰基、羟烷基、杂环基、芳基和杂芳基;
所述g选自0、1、2、3、4、5、6;
所述f选自0、1、2;
p、q、L各自独立地为0、1、2、3或4;
所述杂芳基、所述杂环烷基或所述杂环基上的杂原子各自独立地选自O、S或N。
在本发明提供的一些实施例中,所述化合物选自式(Ⅶ)或(Ⅷ)或(Ⅸ)或(Ⅹ)或(Ⅺ)或(Ⅻ):

在本发明提供的一些实施例中,所述化合物选自下式结构:
在本发明提供的一些实施例中,所述化合物选自下式结构:
在本发明提供的一些实施例中,所述化合物选自下式结构:
优选的,所述的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,所述化合物选自下式结构:
其中,R4独立的选自氘原子、烷基、氧代、烷氧基、羟基、卤素、氰基、炔基、烯基、-(CH2)g-O-3至12元杂环基、-(CH2)g-O-3至12元环烷基、-(CH2)g-3至12元环烷基、-(CH2)g-3至12元杂环基、5至10元杂芳基、5至10元芳基、-S(=O)f-C1-6烷基、-O-C2-6炔基、-O-C2-6烯基;
q选自1、2、3或4。
本发明提供的一些优选实施例中,化合物选自:
本发明的第二方面涉及一种药物组合物,其包括本发明第一方面所示的化合物、其溶剂化物、立体异构 体、氘代化合物或其药学可接受的盐,以及药学上可接受的载体。
本发明的第三方面提供了本发明第一方面所示的化合物、其溶剂化物、立体异构体、氘代化合物或其药学可接受的盐,或本发明第二方面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。
本发明提供的一些优选实施例中,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、心血管疾病和呼吸疾病;优选地,所述疼痛选自术后疼痛、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。
术语解释
本发明所用术语“C2-6炔基”指含有碳碳叁键的2~6个碳原子的炔烃部分去除一个氢原子衍生的直链或支链的炔烃基,如乙炔基、丙炔基、2-丁炔基、2-戊炔基、3-戊炔基、4-甲基-2-戊炔基、2-己炔基、3-己炔基等。
本发明上述的“环烷基”包括可能形成的所有单环、稠环(包括以并、螺、桥的形式稠合)的情形;例如:“3-12元环烷基”,可以是单环、双环、或者多环环烷基系统(也称为稠环系统)。在不特别指明的情况下,单环系统是含3-8个碳原子的环烃基基团,实例包括但不限于:环丙烷基、环丁烷基、环戊烷基、环己烷基、环庚烷基、环辛烷基等。
稠环环烷基包括并环环烷基、桥环烷基、螺环烷基。
并环环烷基可以为6-11元并环环烷基、7-10元并环环烷基,其代表性例子包括但不限于双环[3.1.1]庚烷、双环[2.2.1]庚烷、双环[2.2.2]辛烷、双环[3.2.2]壬烷、双环[3.3.1]壬烷和双环[4.2.1]壬烷。
螺环烷基可以为7-12元螺环烷基、7-11元螺环烷基,其实例包括但不限于: 的基团。
上述的桥环烷基可以为6-11元桥环烷基、7-10元桥环烷基,其实例包括但不限于: 的基团。
本文所用术语“杂环烷基”(或“杂脂环”)是指一价的单环的非芳香族环系,其环原子由碳原子及选自氮、氧、硫和磷的杂原子构成,并且通过一个单键连接至母核或其他基团;常见的杂环烷基包括(但不限于)环氧乙烷基、氧杂环丁烷-3-基、氮杂环丁烷-3-基、四氢呋喃-2-基、吡咯烷-1-基、吡咯烷-2-基、四氢-2氢-吡喃-2-基、四氢-2氢-吡喃-4-基、哌啶-2-基、哌啶-4-基等。
本文所用术语“杂环基”指3-12元的至少一个环碳原子被选自O、S、N的杂原子替代的非芳香性的环状基团,优选1-3个杂原子,同时包括碳原子、氮原子和硫原子可以被氧代。“3-12元杂环基”,是指单环杂环基、双环杂环基系统或多环杂环基系统(也称为稠环系统),包括饱和、部分饱和的杂环基,但不包括芳环。在不特别指明的情况下,包括可能形成的所有单环、稠环(包括以并、螺、桥的形式稠合)、饱和、部分饱和的情形。
单杂环基可以为3-8元单杂环基、3-6元单杂环基、4-7元单杂环基、5-7元单杂环基、5-6元单杂环基、5-6元含氧单杂环基、3-8元含氮单杂环基、5-6元含氮单杂环基、5-6元饱和单杂环基等,其实例包括但不限于: 的基团。
稠杂环包括并杂环基、螺杂环基、桥杂环基,可以是饱和的、部分饱和的或不饱和的,但不是芳香性的。
并杂环基可以为6-12元并杂环基、7-10元并杂环基、6-10元并杂环基、6-12元饱和并杂环基,代表性实例包括但不限于:的基团。
螺杂环基可以为6-12元螺杂环基、7-11元螺杂环基、6-12元饱和螺环基,其实例包括但不限于:

的基团;
上述的桥杂环基可以为6-12元桥杂环基、7-11元桥杂环基、6-12元饱和桥环基,其实例包括但不限于:的基团。
本文所用术语“芳基”(或“芳环”)是指一价的单环或多环(包含稠合形式)的芳香族环系,其仅有碳原子和氢原子构成;常见的芳基包括(但不限于)苯基、萘基、蒽基、菲基、苊基、薁基、芴基、茚基、芘基等。上述芳基也包括杂环基并芳基、环烷基并芳基;
本文所用术语“杂芳基”(或“杂芳环”)是指一价的单环或多环(包含稠合形式)的芳香族环系,其环原子由碳原子及选自氮、氧、硫和磷的杂原子构成;常见的杂芳基包括(但不限于)苯并吡咯基、苯并呋喃基、苯并噻吩基、苯并咪唑基、苯并噁唑基、苯并噻唑基、氮杂环丁烷基、咔唑基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、噻唑基、吡唑基、异噁唑基、异噻唑基、吲唑基、吲嗪基、吲哚基、喹啉基、异喹啉基、吩嗪基、吩噁嗪基、吩噻嗪基、蝶啶基、嘌呤基、吡嗪基、嘧啶基、哒嗪基、吡啶基、三唑基、四唑基等。上述杂芳基也包括杂环基并杂芳基、环烷基并杂芳基。
本发明所示化合物的“立体异构体”是指当化合物存在不对称碳原子时,会产生对映异构体;当化合物存在碳碳双键或环状结构时,会产生顺反异构体;当化合物存在酮或肟时,会产生互变异构体,所有化合物的对映异构体、非对映异构体、消旋异构体、顺反异构体、互变异构体、几何异构体、差向异构体及其混合物,均包括在本发明范围中。
具体实施方式
为使本发明的目的、技术方案、及优点更加清楚明白,以下举实施例,对本发明进一步详细说明。显然,所描述的实施例仅仅是本发明一部分实施例,而不是全部的实施例。基于本发明中的实施例,本领域普通技术人员所获得的所有其他实施例,都属于本发明保护的范围。
如本文所用,室温是指约20-30℃;“过夜”是指约10h~16h;1M、1N为1mmol/L,1μM为1μmol/L,1mM为1mmol/L,1nM为1nmol/L;eq:当量;
收率或产率=实际合成产物质量/理论合成产物质量×100%。
实施例1
(1R,4R)-4-乙氧基-N-{2-[9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基]乙基}-1,2,3,4-四氢萘-1-胺(化合物141)的合成
步骤1:2-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基)乙酸(141-2)的合成
室温下,将化合物141-1(4.66g,18.32mmol,1.0eq)溶解于1M HCl水溶液(23mL)中,升温至100℃后,在N2保护下搅拌反应16小时。待反应结束后,待其自然冷却至室温,水相用乙酸乙酯(100mL×3)洗涤,然后将水相用饱和NaOH水溶液调节pH到~13,用乙酸乙酯(100mL×3)洗涤,将水相用1M HCl水溶液调节pH到4~6,用乙酸乙酯(100mL×5)萃取。合并萃取后的有机相用盐水(100mL×2)洗涤,用无水硫酸钠干燥,过滤,滤液真空下浓缩得到淡黄色固体化合物141-2(4.52g,产率90.22%,[M+H]+:274.17。
步骤2:(R)-2-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基)乙酸和(S)苯乙胺盐(141-3)的合成
室温下,向100mL的单口瓶中加入乙醇(EtOH,30mL)和化合物141-2(4.52g,16.46mmol),加热到50℃,在50℃下搅拌0.5小时,在50℃下缓慢滴加S-苯乙胺和乙醇(15mL)的溶液。加料完毕后,升温到80℃搅拌1小时。缓慢冷却到室温,并在室温下搅拌12小时。析出白色固体,过滤,固体用乙醇(10mL×2)洗涤2次,收集得到的白色固体加入到30mL的乙醇中,加热到80℃,待其溶清。在80℃下搅拌0.5小时,逐渐冷却到室温,待白色固体析出后,过滤,滤饼用乙醇(10mL×2)洗涤2次,收集固体,得到白色固体141-3(1.2g,e.e.%:99.6%)。
步骤3:(R)-2-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基)乙酸(141-4)的合成
将化合物141-3(1.2g)溶于20mL的水中,用1M氢氧化钠溶液调节pH>9,用二氯甲烷(50mL×3)洗涤,水相用1N盐酸溶液调节pH=4~6后用二氯甲烷(50mL×5)萃取,合并萃取有机相并用饱和食盐水(50mL×1)洗涤,无水硫酸钠干燥,过滤,旋干滤液,得到淡黄色油状化合物141-4(600mg)。
步骤4:氮-甲基-氮-甲氧基-(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烯烃-9-基)乙酰胺(141-5)的合成
室温下,将化合物141-4(600mg,2.2mmol,1.0eq)用二氯甲烷(30mL)溶解,然后依次加入甲氧基(甲基)胺盐酸盐(256.93mg,2.63mmol,1.2eq)、1-乙基-3(3-二甲基丙胺)碳二亚胺)(EDCI)(631.22mg,3.29mmol,1.5eq)和4-二甲氨基吡啶(DMAP)(26.82mg,219.51μmol,0.1eq),在N2保护下搅拌0.5h后,再加入N,N-二异丙基乙胺(DIPEA)(851.17mg,6.59mmol,3.0eq)搅拌反应过夜。待反应结束后,往反应液中加入NH4Cl饱和溶液(100mL)淬灭,再用二氯甲烷(100mL×5)萃取,合并有机相并用饱和食盐水(100mL×2)洗涤,无水Na2SO4干燥,过滤,滤液通过减压浓缩得到黄色稠状物141-5(592mg,产率:85.24%),[M+H]+:317.17。
步骤5:化合物氮-甲基-氮-甲氧基-(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烯烃-9-基)乙醛(141-6)的合成
室温下,将化合物141-5(438.6mg,1.39mmol,1.0eq)用甲苯(Tol,7mL)溶解,在-40℃、N2保护下,缓慢滴加红铝(Red-Al)(657.9mg,2.28mmol,1.05eq),加料完毕后搅拌反应4h。待反应结束后,往反应液中加入质量分数为10%的柠檬酸溶液(20mL)淬灭,再移至室温,向反应液中加入质量分数为10%柠檬酸水溶液(10mL),再移至室温,用1mol/L的HCl溶液调pH=2~3,用乙酸乙酯(30mL×1)萃取;水相再用5N NaOH溶液调pH=11~13,用二氯甲烷(50mL×3)萃取,合并有机相并用饱和食盐水(50mL×1)洗涤,用无水硫酸钠干燥,过滤,滤液通过减压浓缩得到橘红色稠状物141-6(287.2mg),收率:80.5%,[M+H]+:258.16。
步骤6:(1R,4R)-4-乙氧基-N-{2-[9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-e]乙基}-1,2,3,4-四氢萘-1-胺(141)的合成
室温下,将化合物141-6(50mg,0.19mmol),化合物141-7(37.2mg,0.19mmol)溶于二氯甲烷(DCM)(3mL),再加入MgSO4(116.9mg,0.97mmol),在氮气保护下,室温搅拌12h,再加入硼氢化钠(22.1mg,0.58mmol),并搅拌2h,再加入甲醇(1mL),并搅拌0.5h。再用硅藻土过滤,滤液室温浓缩,大板(V 油醚(PE):V乙酸乙酯(EA)=0:1)纯化得到黄色稠状物141(35mg,产率41.7%),[M+H]+:433.40。
1H NMR(400MHz,CDCl3)δ8.60-8.56(m,1H),7.70-7.64(m,1H),7.36-7.32(m,2H),7.26-7.19(m,3H),7.18-7.13(m,1H),5.66-5.61(m,1H),5.49-5.45(m,1H),4.39-4.35(m,1H),3.95-3.88(m,1H),3.87-3.82(m,1H),3.71-3.64(m,2H),3.56-3.51(m,1H),2.59-2.49(m,3H),2.47-2.38(m,2H),2.29-2.22(m,1H),2.12-1.98(m,5H),1.96-1.90(m,1H),1.87-1.77(m,3H),1.30-1.20(m,5H).
实施例2
(1R,4R)-4-乙氧基-N-{2-[4'-(吡啶-2-基)螺[双环[3.1.0]己烷-3,2'-氧烷]-4'-基]乙基}-1,2,3,4-四氢萘-1-胺(化合物143)的合成
步骤1:氮-甲基-氮-甲氧基-(R)-2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基乙酰胺(143-1)的合成
冰水浴下,将化合物141-5(590mg,1.86mmol)溶解于DCM(15mL)中,在氮气保护下,加入2mol/L的二甲基锌(3.7mL,7.46mmol)、二碘甲烷(5.0g,18.6mmol),加料完毕,移至室温搅拌反应16h。反应结束后,向反应液中加入乙酸乙酯(50mL),再用饱和NaHCO3溶液(20mL×2)洗涤,有机相用无水硫酸钠干燥,过滤,滤液旋干,柱层析(VPE:VEA=2:1)纯化得到黄色稠状物143-1(223mg,产率36.26%),[M+H]+:331.19。
步骤2:(R)-2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基乙醛(143-2)的合成
室温下,将化合物143-1(223mg,0.68mmol)溶解于四氢呋喃(THF)(10mL)中,在-40℃、氮气保护下,缓慢滴加红铝(335mg,70%,1.16mmol),加料完毕,搅拌反应4h。反应结束后,向反应液中加入质量分数为10%的柠檬酸水溶液(10mL),再移至室温,用1mol/L的HCl溶液调pH=2~3,用乙酸乙酯(30mL×1)萃取;水相再用质量分数为10%的NaOH溶液调pH=11~13,用二氯甲烷(50mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩得到棕色油状物143-2(160mg,产率87.37%),[M+H]+:272.23。
步骤3:(1R,4R)-4-乙氧基-N-{2-[4'-(吡啶-2-基)螺[双环[3.1.0]己烷-3,2'-氧烷]-4'-基]乙基}-1,2,3,4-四氢萘-1-胺(143)的合成
室温下,将化合物143-2(50mg,0.18mmol),化合物141-7(35mg,0.18mmol)溶于DCM(3mL),再加入MgSO4(111mg,0.92mmol),在氮气保护下,室温搅拌12h,再加入NaBH4(21mg,0.55mmol),并搅拌1h,再加入甲醇(1mL),并搅拌0.5h。再用硅藻土过滤,滤液室温浓缩,大板(VPE:VEA=0:1)纯化得到黄色稠状物143(32mg,产率39.0%),[M+H]+:447.43。
1H NMR(400MHz,CDCl3)δ8.56-8.49(m,1H),7.69-7.63(m,1H),7.39-7.34(m,1H),7.33-7.30(m,1H),7.26-7.18(m,3H),7.17-7.12(m,1H),4.41-4.36(m,1H),3.84-3.62(m,5H),3.59-3.48(m,2H),2.39-2.28(m,2H),2.26-2.21(m,1H),2.17-2.04(m,3H),1.89-1.80(m,3H),1.37-1.22(m,7H),1.19-1.10(m,2H),1.09-0.99(m,2H),0.59-0.54(m,1H),0.30-0.24(m,1H).
实施例3
(1R,4R)-4-乙氧基-氮-{2-[9-(吡啶-2-基)-2,6-二氧杂螺[4.5]癸烷-9]乙基}-1,2,3,4-四氢萘-1-胺(化合物156)的合成
室温下,将化合物156-1(50mg,0.19mmol),化合物141-7(37.2mg,0.19mmol)溶于DCM(3mL),再加入MgSO4(116.9mg,0.97mmol),在氮气保护下,室温搅拌12h,再加入硼氢化钠(22.1mg,0.58mmol),并搅拌2h,再加入甲醇(1mL),并搅拌0.5h。再用硅藻土过滤,滤液室温浓缩,大板(VPE:VEA=0:1)纯化 得到黄色稠状物156(35mg,产率41.7%),[M+H]+:437.27。
1H NMR(400MHz,CDCl3)δ8.60-8.56(m,1H),7.70-7.64(m,1H),7.36-7.32(m,2H),7.26-7.19(m,3H),7.18-7.13(m,1H),4.39-4.35(m,1H),3.95-3.88(m,1H),3.87-3.82(m,1H),3.71-3.64(m,2H),3.56-3.51(m,1H),2.59-2.49(m,3H),2.47-2.38(m,2H),2.29-2.22(m,1H),2.12-1.98(m,5H),1.96-1.90(m,1H),1.87-1.77(m,3H),1.45-1.39(m,1H)1.30-1.20(m,5H)。
实施例4
氮-(2-(9-(吡啶-2-基)-6-氧杂螺环[4.5]癸烷-2-烯-9-基)乙基)-2,3-二氢-1氢-茚-1-胺(化合物128)的合成
氮气保护下,将化合物A(100mg,0.39mmol),化合物9(76mg,0.58mmol),二氯乙烷(DCE,6mL)依次加入单口瓶中,室温下加入钛酸四异丙酯(TIPT,1.5mL),60℃搅拌16h后,加入硼氢化钠(44mg,1.2mmol),60℃搅拌反应2小时。反应完后,加入1.5mL水淬灭,加10mL二氯甲烷,过滤,室温浓缩过柱(V二氯甲烷:V甲醇=10:1),得黄色稠状化合物128(20mg,产率14%),[M+H]+:375.3。
1H NMR(400MHz,CDCl3)δ8.58(d,J=8.0Hz,1H),7.675(t,J=8.0Hz,1H),7.32(d,J=8.0Hz,1H),7.20-7.10(m,5H),5.61(brs,1H),5.45(brs,1H),4.08-4.04(m,1H),3.94-3.89(m,1H),3.83-3.80(m,1H),2.95-2.88(m,1H),2.77-2.69(m,1H),2.63-2.38(m,5H),2.26-2.14(m,2H),2.04-1.94(m,3H),1.81-1.76(m,2H),1.70-1.56(m,2H).
实施例5
氮-(2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基)乙基)-2,3-二氢-1氢-茚-1-胺(化合物1)的合成
制备方法参考实施例4,得黄色稠状化合物1(产率7%),[M+H]+:389.3。
1H NMR(400MHz,CDCl3)δ8.59-8.50(m,1H),7.67-7.62(m,1H),7.38-7.27(m,2H),7.20-7.19(m,2H),7.15-7.10(m,2H),4.25(brs,1H),3.80-3.68(m,2H),3.04-2.98(m,1H),2.81-2.79(m,1H),2.77-2.66(m,1H),2.32-2.10(m,5H),1.90-1.84(m,5H),1.66-1.62(m,1H),1.30-1.25(m,2H),1.11-1.01(m,3H),0.55-0.52(m,1H),0.24-0.22(m,1H)。
对比例1
氮-((3-甲氧基噻吩-2-基)甲基)-2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基)乙胺(化合物91)的合成
氮气保护下,将中间体B(100mg,0.37mmol),中间体1(63mg,0.44mmol),硫酸镁(882mg,7.4mmol),二氯甲烷(DCM,6mL)依次加入单口瓶中,室温搅拌16h,加入硼氢化钠(42mg,1.1mmol),搅拌10分钟,加入甲醇(MeOH,0.5mL),搅拌反应2小时。反应完后,加入二氯甲烷(10mL),过滤,室温浓缩,粗品柱层析纯化(V二氯甲烷:V甲醇=10:1),得到黄色稠状化合物91(12.43mg,产率8.5%),[M+H]+:399.3。
1H NMR(400MHz,氘代氯仿(CDCl3))δ8.53-8.52(m,1H),7.62(dt,J1=2.0Hz,J2=7.6Hz,1H),7.27(d,J=8.0Hz 1H),7.13-7.07(m,2H),6.77(d,J=5.2Hz 1H),3.81-3.66(m,7H),2.60-2.53(m,1H),2.32-2.28(m,1H),2.23-2.19(m,1H),2.05-1.96(m,2H),1.89-1.80(m,4H),1.69-1.60(m,2H),1.31-1.26(m,1H),1.10-1.09(m,1H),1.04-1.01(m,1H),0.55-0.52(m,1H),0.25-0.22(m,1H).
对比例2
氮-((3-甲氧基噻吩-2-基)甲基)-2-(9-(吡啶-2-基)-6-氧杂螺环[4.5]癸烷-2-烯-9-基)乙胺(化合物83)的合成
氮气保护下,将中间体A(80mg,0.31mmol),中间体1(53mg,0.37mmol),硫酸镁(744mg,6.2mmol),二氯甲烷(6mL)依次加入单口瓶中,室温搅拌16h,加入硼氢化钠(35mg,0.93mmol),搅拌10分钟,加入甲醇(0.5mL),搅拌反应2小时。反应完后,加入二氯甲烷(10mL),过滤,室温浓缩,粗品柱层析纯化(V二氯甲烷:V甲醇=10:1),得黄色稠状化合物83(35mg,产率29%),[M+H]+:385.3。
1H NMR(400MHz,CDCl3)δ8.57-8.55(m,1H),7.62(dt,J1=2.0Hz,J2=8.0Hz,1H),7.29(d,J=8.0Hz 1H),7.13-7.09(m,1H),7.04(d,J=5.6,Hz 1H),6.76(d,J=5.6Hz 1H),5.61(brs,1H),5.44(brs,1H),3.89-3.86(m,1H),3.83-3.79(m,4H),3.74-3.66(m,2H),2.54-2.37(m,5H),2.14-1.91(m,4H),1.81-1.72(m,2H),1.62-1.56(m,1H).
对比例3
氮-((3-甲氧基噻吩-2-基)甲基)-2-(9-(吡啶-2-基)-2,6-二氧杂螺[4.5]癸烷-9-基)乙胺(化合物31) 的合成
室温下,将中间体C(50mg,0.162mmol)用二氯甲烷(3mL)溶解,然后依次加入硫酸钠(136mg,0.96mmol)和中间体1(41mg,0.288mmol)。反应在氮气保护下室温过夜。反应16h后,加入硼氢化钠继续搅拌30分钟。反应完全后,用15mL水进行淬灭,加入乙酸乙酯(15×2mL)进行萃取然后用饱和碳酸钠溶液(10mL×2)洗涤,有机相用无水硫酸钠干燥,过滤,粗品用柱层析纯化(V石油醚:V乙酸乙酯=1:1)得到淡黄色油状液体31(20mg,产率22%),[M+H]+:389.2。
1H NMR(400MHz,氯仿-d(Chloroform-d))δ8.59(d,J=8.5,3.0Hz,1H),7.65(d,J=7.7,6.0,4.1,1.9Hz,1H),7.32(d,J=8.1Hz,1H),7.17–7.12(m,1H),7.05(d,J=5.5Hz,1H),6.79(d,J=5.4Hz,1H),4.87(s,1H),3.88–3.82(m,3H),3.79(s,3H),3.75(d,J=3.3Hz,1H),3.69(d,J=2.0Hz,2H),3.55(d,J=9.3Hz,1H),3.51(s,2H),3.18(d,J=10.0Hz,1H),2.86(d,J=10.0Hz,1H),2.47(dd,J=11.0,5.9Hz,2H),2.14(dd,J=10.3,4.8Hz,1H),2.03(d,J=13.7Hz,1H),1.92(d,J=9.0Hz,1H),1.77(d,J=4.8Hz,1H),1.45–1.39(m,1H),1.20–1.12(m,1H)。
生物学评价
Op-Mu激动剂cAMP测试实验
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cAMP应答元件(cAMP response element,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。
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。
1.材料与试剂

2.实验操作步骤
检测缓冲液:1×stimulation buffer,500μM 1-甲基-3-异丁基黄嘌呤(IBMX),ddH2O。
化合物配制:化合物用二甲基亚砜(DMSO)溶解后配制为终浓度为10mM的母液,稀释成0.08mM的工作浓度,用Echo移液器对化合物进行4倍梯度稀释,初始浓度为0.08mM,10个浓度梯度,并分别加50nL到384细胞板中,双复孔,终浓度为0.4μM,然后将细胞板1000rpm离心1min。用Echo移液器转移50nL毛喉素(Forskolin)(终浓度是1μM)至384细胞板中。
细胞铺板:将冻存的细胞融化,1000转离心5min,弃掉上清液,用HBSS缓冲液清洗两次细胞后,用检测缓冲液重悬细胞,将细胞密度调整到5.0×105个/mL,加入至384孔板中,每孔10μL,5000个细胞。震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min。
标准曲线的配制:用检测缓冲液对标准品腺苷-3',5'-环磷酸(cAMP)进行4倍梯度稀释,共8个浓度点,最高浓度是800nM,按照微孔板布局图每孔加入10μL。
检测试剂配制:用裂解缓冲液将Anti cAMP-Cryptate和AMP-d2稀释至1×,按照微孔板布局图,每孔加入10μL检测试剂,震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min;在Envision酶标仪上读板。
3.结果分析
运用Microsoft Excel软件计算活性百分比,对于激动剂使用公式%Effect=100×(Sample Raw Value-Low Control Average)/(High Control Average-Low Control Average),运用GraphPad Prism 5数据分析软件,对于激动剂选用Dose-response-Stimulation—log[agonist]vs.response--Variable slope模式进行拟合分析,得出各检测样品的EC50值。
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,实验结果表明此系列化合物表现出较强的Op-Mu激动效应,其中测得的具有典型代表的化合物的EC50值见表一。其中,设置6组对照,分别为化合物83、化合物91、化合物31、TRV130、TRV130(消旋)和SHR8554,其中TRV130、TRV130(消旋)和SHR8554分别具有以下结构式,TRV130制备方法参考专利CN103702561A;SHR8554制备方法参考专利CN107001347B;Emax为化合物引起cAMP水平变化的最大效能。
表一:受试化合物对MOR受体影响cAMP水平的EC50和Emax

本发明实施例的优选化合物对Mu阿片受体具有明显的激动作用,部分化合物EC50值和Emax远优于对照组。
Op-Kappa激动剂cAMP测试实验
ForsKolin(毛喉素)能够刺激人K阿片受体高表达细胞株-OPRKI细胞(DiscoveRx)cAMP的释放,而K阿片受体激动剂能够抑制forsKolin刺激的cAMP释放。通过检测受试化合物对forsKolin刺激的cAMP释放的抑制作用,能够测定化合物对人K片受体的激动活性。首先用一定浓度的forsKolin和不同浓度的受试化合物与人高表达K阿片受体细胞株一起孵育。使用基于时间分辨荧光共振能量转移(TR-FRET)的cAMP免疫测试法(LANCEPerKinElmer)来确定所激发的OPRK1细胞中的cAMP水平。具体方法如下:
检测缓冲液:1×stimulation buffer,500μM IBMX,ddH2O。化合物配制:化合物用DMSO溶解后配制为终浓度为10mM的母液,稀释成2mM的工作浓度,用Echo对化合物进行4倍梯度稀释,初始浓度为2mM,10个浓度梯度,并分别加50nL到384细胞板中,双复孔,终浓度为10μM,然后将细胞板1000rpm离心1min;用Echo转移50nL ForsKolin(终浓度是3μM)至384细胞板。
细胞铺板:将冻存的细胞融化,1000转离心5min,弃掉上清液,用HBSS缓冲液清洗两次细胞后,用检测缓冲液重悬细胞,将细胞密度调整到3.0×105个/mL,加入至384孔板中,每孔10μL,3000个细胞。震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min。标准曲线的配制:用检测缓冲液对标准品cAMP进行4倍梯度稀释,共8个浓度点,最高浓度是800nM,按照微孔板布局图每孔加入10μL。检测试剂配制:用裂解缓冲液将中间体Anti cAMP-Cryptate和AMP-d2稀释至1×,每孔加入10uL检测试剂,震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min。最后在Envision上读板。
运用Microsoft Excel软件计算活性百分比,对于激动剂使用公式%Effect=100×(Sample Raw Value-Low Control Average)/(High Control Average-Low Control Average)。运用GraphPad Prism 5数据分析软件,对于激动剂选用Dose-response-Stimulation—log[agonist]vs.response--Variable slope模式进行拟合分析,得出各检测样品的EC50值。实验数据见表二。
表二:受试化合物对K阿片受体影响
本发明实施例化合物对激动K阿片受体的活性相比对照组明显较弱;表现出本发明化合物对MOR受体有高的选择性,推测本发明实施例化合物具有更低的副作用。
Mμ阿片受体的β-arrestin信号通路的活性测试实验
本研究旨在通过CHO-K1/Arrestin/hMOR的EC50和EMAX测定来评估靶向μ-阿片受体MOR的激动剂的β-Arrestin招募效率。CHO-K1/Arrestin/hMOR细胞株表达与β-半乳糖苷酶供体片段融合的hMOR和与β-半乳糖苷酶受体片段融合的β-Arrestin。当β-arrestin与hMOR相互作用时,这些片段形成活性的β-半乳糖苷酶。准备384孔板,将60nL/孔系列稀释的化合物滴入384孔板,将20μLCHO-K1/Arrestin/hMOR细胞浮液注入测定板,细胞密度为7.5k细胞/孔。37℃,0.5%CO2(体积分数,其余为空气)孵育分析板120分钟,用蜻蜓法将10μL/孔检测试剂加入分析板中,培养皿在室温孵育60分钟,通过Envision检测化学发光信号,使用XLfit进行数据分析。实验数据见表三。
表三:受试化合物对β-arrestin信号通路的影响
本发明实施例化合物对β-arrestin信号通路几乎无激活作用,以及本发明化合物和对照组相比有更好的偏向性(cAMP与β-arrestin信号通路),推测本发明化合物相比对照组具有更低的副作用。
测试本发明化合物对hERG钾电流的阻断作用
试验系统
细胞:中国仓鼠卵巢(CHO)细胞系,CHO-hERG细胞用于本试验。
细胞培养液及培养条件:完全培养基为F12培养基,补充加入10%胎牛血清,1%选择性抗生素(G418),89μg/mL潮霉素B(HB)。复苏培养基为F12培养基补充加入10vol%胎牛血清。CHO-hERG细胞生长在37℃(±2℃)、5%CO2(4%至8%)的高湿度培养箱中。细胞用复苏培养基复苏,完全培养基传代,用于膜片钳试验的细胞在最后一次传代时换成复苏培养基。
细胞外液及内液成分:
试验方法
(1)将处于指数生长期的CHO-hERG细胞收集并重悬在ECS中备用。
(2)手动膜片钳试验
全细胞膜片钳技术下记录hERG电流,记录温度为室温。膜片钳放大器输出信号通过数模转换以及2.9KHz低通滤波。数据记录用Patchmaster Pro软件采集。
细胞种在细胞记录槽中放置在倒置显微镜载物台上,随机选择记录槽中的一个细胞进行试验。灌流系统固定在倒置显微镜载物台上用ECS持续灌流细胞。
用毛细玻璃管制备手动膜片钳试验记录微电极,其中充灌细胞內液。在膜片钳试验当天,使用硼硅酸盐 玻璃管(BF150-117-10,SUTTER INSTRUMENT USA)制备电极。电极充灌ICS后电阻在2-5MΩ之间。
钳制电压为-80mV,第一步去极化至+60mV并维持850ms开放hERG通道。然后,电压设置为-50mV并维持1275ms,产生反弹电流或者称为尾电流,尾电流的峰值将被测量并用于分析。最后,电压恢复到钳制电压(-80mV)。试验过程中,这个指令电压程序每间隔15s重复一次。
在溶媒对照工作溶液灌流的记录开始阶段,监测尾电流峰值直至稳定3条以上扫描曲线后则可以灌流待测试的供试品/阳性对照工作溶液,直到供试品/阳性对照工作溶液对hERG电流峰值的抑制作用达到稳定状态。一般以最近的连续3个电流曲线峰值基本重合作为判断是否稳定状态的标准。达到稳定态势以后继续灌流下一浓度供试品。一个细胞上可以测试一个或多个供试品/阳性对照,或者同一种药物的多个浓度,不同供试品/阳性对照之间需用溶媒对照工作液冲洗直到hERG电流回复到加药物之前80%以上的大小。同一浓度下各记录细胞抑制率的标准差不超过15%。
阳性对照西沙必利的测试浓度为0.1μM,重复测定两个细胞。根据科学文献报道,0.1μM的西沙必利抑制hERG电流超过50%。(Milnes,J.T.,et al.)。
(3)手动膜片钳数据接受标准
封接标准:全细胞模式形成后,施加钳制电压(-80mV),可以记录到细胞膜相关参数(Cm,Rm以及Ra)。一个好的的全细胞记录应该满足以下条件:路径电阻(Rs)小于10MΩ;膜电阻(Rm)大于500MΩ和膜电容(Cm)小于100pF。
电流大小:供试品/阳性对照品作用前峰电流幅度在400pA和5000pA之间。否则,放弃该细胞。
漏电流:在-80mV的钳制电压下,漏电流绝对值应该小于200pA。电流幅度将会用-80mV下的漏电流校正。漏电流绝对值大于200pA的扫描曲线不能用于分析。
数据分析
对于每个细胞,每一个浓度的供试品及阳性对照的抑制百分比由记录到的电流反应用以下公式算出:(1–供试品/阳性对照灌流后记录到的尾峰值电流/溶媒对照灌流记录到的尾峰值电流(起始电流))×100%。
对于每一个浓度记录到所有的细胞抑制百分比取均值,IC50值(半数抑制浓度)由Hill拟合的方法由浓度效应曲线中得出。
试验结果
本发明部分化合物对hERG电流的抑制结果,具体见下表四;
表四:受试化合物对hERG电流的抑制结果
注:20μM>IC50>10μM为++,10μM>IC50>1μM为+。
本发明实施例化合物相比对照组具有较高的hERG IC50值,具有显著差异,表现出对hERG抑制作用更弱,说明本发明化合物的心脏毒性风险较低。
药代动力学实验
被研究化合物单次口服或者静脉给药(溶媒5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline)于动物(例如小鼠、大鼠、犬或者猴子),在固定的时间点取血。血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血液用肝素抗凝,然后8000rpm离心5分钟,将血清与红细胞分离。用移液器吸岀血清转移至2mL的聚丙烯管,标明化合物的名称和时间点,在进行LC-MS分析前保存在-40℃冰箱,待测。髙浓度样品用空白血浆稀释测定时。样品处理后,用LCMS/MS对血浆中的物质进行定量分析。通过进行了验证的药动学计算机程序,用以这种方式获得的血浆浓度/时曲线来计算药动学参数。实验发现本发明化合物均具有较好的药代动力学性质。
SD雄性大鼠以表五组别剂量静脉给药后(各组为等摩尔剂量给药,溶媒为5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline,每组3只),在固定的时间点取血检测。本发明的部分化合物的在大鼠血浆中的原型化合物药代动力学参数如下表五;表五中,IV为静脉给药,AUC为血浆浓度-时间曲线下面积,Cmax为最大血药浓度,T1/2为消除半衰期,Variable为变量,Mean为平均数,SD为标准差。
表五:受试化合物药代动力学参数
本发明实施例化合物在大鼠体内展现出良好的药代动力学性质;与对照组相比,本发明化合物在血浆中游离碱的AUC(h*ng/mL)均有显著提高。
大鼠热板法镇痛药效实验
雌性SD大鼠,将大鼠开始训练日期记为D0。于D0,将测痛仪温度设定为52℃(52.0±0.5℃),大鼠置于热板同时计时,记录大鼠出现舔后足或跳跃时痛阈值所用的时间(s)。若大鼠于热板上超过30s没有出现热痛反应,立即取出大鼠,痛阈值记为30s。淘汰反应敏感和反应迟钝大鼠。于D1,取预筛后的大鼠,将测痛仪温度设定为52℃(52.0±0.5℃),大鼠置于热板并计时,记录大鼠出现舔后足或跳跃时痛阈值所用的时间(s),共测三次。以三次平均值作为大鼠baseline痛阈值。于D1,大鼠根据baseline痛阈值随机分组,每组8只动物。于D2,按照表中各剂量组,通过尾静脉注射方式给予溶媒或化合物(各组等摩尔剂量给药, 溶媒为5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline)。于D2,各组大鼠给药后0.5min、0.5h、1h、3h,将测痛仪温度设定为52℃(52.0±0.5℃),大鼠置于热板并计时,记录大鼠出现舔后足或跳跃时痛阈值所用的时间(s),每个时间点测1次。若大鼠于热板上超过30s没有出现热痛反应,立即取出大鼠,痛阈值记为30s。计算各组各个时间点痛阈%MPE,评价各受试样品体内镇痛药效。实验数据以Mean±SEM表示,各组间数据采用GraphPad Prism进行方差分析(ANOVA)检验(Two Way ANOVA或One-Way ANOVA),P<0.05认为是有显著性差异。最大镇痛效应百分率(即痛阈%MPE)=(给药后痛阈值-基础痛阈值)/(30-基础痛阈值)×100%,实验结果下表六;表六中,Variable为变量,Average为平均数,SEM为标准误差。
表六:受试化合物体内镇痛药效
注:与对照组(溶媒,IV)相比,***p<0.001
本发明实施例的化合物相比对照组展现出更好的镇痛效果,在大鼠热板法镇痛药效研究中表现出痛阈更高,持续时间更长久的镇痛作用。
急性毒性实验
化合物单次静脉给药(溶媒5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline)于SD大鼠(每个化合物设置4-6剂量组,每剂量组10只,雌性各半),给药后后进行临床观察。临床观察第一天两次,第二天开始一天一次,连续14天。包括行为学观察、自主活动和神经系统行为和死亡情况等,得到化合物的最大耐受 量(MTD值)和半数致死量(LD50值)。实验结果显示本发明化合物在大鼠单次静脉给药的MTD值和LD50值相比对照组有显著提高,表明本发明化合物具有良好的安全性。
以上所述仅为本发明的较佳实施例,并不用以限制本发明,凡在本发明的精神和原则之内,所做的任何修改、等同替换、改进等,均应包含在本发明保护的范围之内。

Claims (10)

  1. 式(Ⅳ)或(Ⅴ)或(Ⅵ)所示的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,
    其中,环B、环C各自独立的选自被取代的或未被取代的芳基、被取代的或未被取代的杂芳基;
    环D选自环烷基、杂环烷基;
    R4、R5各自独立的选自H、氘原子、烷基、氧代、烷氧基、羟基、卤素、氰基、炔基、烯基、-(CH2)g-O-3至12元杂环基、-(CH2)g-O-3至12元环烷基、-(CH2)g-3至12元环烷基、-(CH2)g-3至12元杂环基、5至10元杂芳基、5至10元芳基、-S(=O)f-C1-6烷基、-O-C2-6炔基、-O-C2-6烯基;其中,所述杂环基、杂芳基、芳基、烷基、炔基、烯基、烷氧基可任选进一步被1至3个R6所取代;
    所述R6各自独立的选自氘原子、卤素、-OH、-C1-6烷基、-C1-6烷基-O-C1-6烷基、-O-C1-6烷基、3至6元环烷基、-O-C2-6炔基、-O-C2-6烯基、-C2-6炔基、-C2-6烯基、氨基、羧酸酯基、硝基、氰基、羟烷基、杂环基、芳基、杂芳基;
    所述g选自0、1、2、3、4、5、6;
    所述f选自0、1、2;
    p、q、L各自独立地为0、1、2、3或4;
    所述杂芳基、所述杂环烷基或所述杂环基上的杂原子各自独立地选自O、S或N。
  2. 根据权利要求1所述的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,其特征在于,所述化合物选自式(Ⅶ)或(Ⅷ)或(Ⅸ)或(Ⅹ)或(Ⅺ)或(Ⅻ):

  3. 根据权利要求1所述的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,其特征在于,所述化合物选自下式结构:
  4. 根据权利要求1所述的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,其特征在于,所述化合物选自下式结构:
  5. 根据权利要求1所述的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,其特征在于,所述化合物选自下式结构:
    其中,R4独立的选自氘原子、烷基、氧代、烷氧基、羟基、卤素、氰基、炔基、烯基、-(CH2)g-O-3至12元杂环基、-(CH2)g-O-3至12元环烷基、-(CH2)g-3至12元环烷基、-(CH2)g-3至12元杂环基、5至10元杂芳基、5至10元芳基、-S(=O)f-C1-6烷基、-O-C2-6炔基、-O-C2-6烯基;
    q选自1、2、3或4。
  6. 根据权利要求1所述的化合物、其溶剂化物、立体异构体、氘代化合物或其药学可接受的盐,其特征在于,所述化合物选自:
  7. 一种药物组合物,其特征在于,所述药物组合物包括权利要求1至5中任一项所述的化合物、其溶剂化物、立体异构体、氘代化合物或其药学可接受的盐,以及药学上可接受的载体。
  8. 根据权利要求1至6中任一项所述的化合物、其溶剂化物、立体异构体、氘代化合物或其药学可接受的盐,或根据权利要求7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。
  9. 根据权利要求8所述的用途,其特征在于,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、心血管疾病和呼吸疾病。
  10. 根据权利要求9所述的用途,其特征在于,所述疼痛选自术后疼痛、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。
PCT/CN2023/072810 2022-01-19 2023-01-18 阿片受体激动剂及其制备方法和用途 WO2023138603A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202380008359.0A CN116801881A (zh) 2022-01-19 2023-01-18 阿片受体激动剂及其制备方法和用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210057938.1 2022-01-19
CN202210057938 2022-01-19

Publications (1)

Publication Number Publication Date
WO2023138603A1 true WO2023138603A1 (zh) 2023-07-27

Family

ID=87347858

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/072810 WO2023138603A1 (zh) 2022-01-19 2023-01-18 阿片受体激动剂及其制备方法和用途

Country Status (2)

Country Link
CN (2) CN117263956A (zh)
WO (1) WO2023138603A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017063509A1 (zh) * 2015-10-15 2017-04-20 江苏恒瑞医药股份有限公司 氧杂螺环类衍生物、其制备方法及其在医药上的应用
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
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
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

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017063509A1 (zh) * 2015-10-15 2017-04-20 江苏恒瑞医药股份有限公司 氧杂螺环类衍生物、其制备方法及其在医药上的应用
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
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
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

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
XIN LI, WEI HE, YANG CHEN, GUIMEI YANG, HONG WAN, LEI ZHANG, QIYUE HU, JUN FENG, ZHIGAO ZHANG, FENG HE, CHANG BAI, LIANSHAN ZHANG,: "Discovery of SHR9352: A Highly Potent G Protein-Biased μ-Opioid Receptor Agonist", ACS OMEGA, vol. 2, no. 12, 31 December 2017 (2017-12-31), US , pages 9261 - 9267, XP055650322, ISSN: 2470-1343, DOI: 10.1021/acsomega.7b01452 *
YANG YIFEI; WANG YONGHAI; ZUO AIXIA; LI CHUNMEI; WANG WENYAN; JIANG WANGLIN; ZHANG XIAOCHEN; CHE XIN; ZHANG YANG; WU WENTAO; CEN X: "Synthesis, biological, and structural explorations of a series of μ-opioid receptor (MOR) agonists with high G protein signaling bias", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 228, 12 November 2021 (2021-11-12), AMSTERDAM, NL , pages 1 - 17, XP086906209, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2021.113986 *

Also Published As

Publication number Publication date
CN116801881A (zh) 2023-09-22
CN117263956A (zh) 2023-12-22

Similar Documents

Publication Publication Date Title
JP6923683B2 (ja) オピオイド受容体リガンド並びにそれらの使用方法及び製造方法
TWI615393B (zh) 丙烯酸類衍生物、其製備方法及其在醫藥上的用途
CN104093716B (zh) 联芳醚磺酰胺化合物及其作为治疗剂的用途
CN103649056B (zh) 作为离子通道调节剂的稠合杂环化合物
CN109415361B (zh) 丙烯酸类衍生物及其制备方法和其在医药上的用途
EP2970277B1 (en) Imidazo(4,5-b) pyridin-2-yl amides as kv7 channel activators
US20070254903A1 (en) Novel Spiroindoline or Spiroisoquinoline Compounds, Methods of Use and Compositions Thereof
CN104024251A (zh) 苯磺酰胺化合物及其作为治疗剂的用途
JP2008536942A (ja) 2−ピロリドン誘導体および炎症状態および疼痛の治療のためのその使用
CN106573937A (zh) 作为fshr调节剂的吡唑化合物及其用途
JP2020531526A (ja) アデノシン受容体アンタゴニストとしてのキノキサリン誘導体
JP6466602B2 (ja) ムスカリンm1受容体ポジティブアロステリックモジュレーター
CN114591293A (zh) 作为Nav1.8抑制剂的并环化合物及其用途
JP2006523641A (ja) グルカゴンレセプターアンタゴニストとしての置換3−シアノチオフェンアセトアミド
KR20080018907A (ko) 수용체 티로신 키나아제 met의 억제제로서의벤조사이클로헵타피리딘
JP2020528922A (ja) アデノシン受容体アンタゴニストとしてのチアゾロピリジン誘導体
KR101618624B1 (ko) 신규한 옥타히드로티에노퀴놀린 유도체, 그것을 함유하는 의약 조성물 및 그것들의 용도
JP2020531531A (ja) アデノシン受容体アンタゴニストとしてのベンズイミダゾール誘導体
WO2023138603A1 (zh) 阿片受体激动剂及其制备方法和用途
EP3589637B1 (en) Compounds useful for inhibiting ror-gamma-t
JP6185650B2 (ja) 5−ht4受容体アゴニストとしての5−アミノ−キノリン−8−カルボキサミド誘導体
CN112851583A (zh) 新型苯并氮杂䓬类化合物、组合物及其用途
WO2023030319A1 (zh) 阿片受体激动剂及其制备方法和用途
CN118184641A (zh) 阿片受体激动剂及其制备方法和用途
CN116162093B (zh) 一种tyk2抑制剂化合物及其用途

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 202380008359.0

Country of ref document: CN

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

Ref document number: 23742918

Country of ref document: EP

Kind code of ref document: A1