CN111836807A - Oxaspiro compounds, preparation method and application thereof - Google Patents

Oxaspiro compounds, preparation method and application thereof Download PDF

Info

Publication number
CN111836807A
CN111836807A CN201980016139.6A CN201980016139A CN111836807A CN 111836807 A CN111836807 A CN 111836807A CN 201980016139 A CN201980016139 A CN 201980016139A CN 111836807 A CN111836807 A CN 111836807A
Authority
CN
China
Prior art keywords
alkyl
ring
hydroxy
halogen
group
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN201980016139.6A
Other languages
Chinese (zh)
Inventor
田强
张毅涛
宋智泉
宋立强
蔡家强
王利春
王晶翼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN111836807A publication Critical patent/CN111836807A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • 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

Abstract

An oxaspiro compound and a preparation method and application thereof, in particular to a compound shown in a formula (I), a solvate, a stereoisomer, a crystal form, a pharmaceutically acceptable salt or ester thereof, or any combination of the compounds, the preparation method thereof and the application thereof in activating opioid receptor activity and preparing analgesic drugs,

Description

Oxaspiro compounds, preparation method and application thereof
the present application is based on and claims priority from applications having CN application No. 201810402312.3, application date 2018, 28/4, and CN application No. 201811358917.3, application date 2018, 11/15, the disclosure of which is incorporated herein by reference in its entirety.
Technical Field
The application relates to the field of medicine, in particular to an oxaspiro compound, a preparation method thereof and application thereof in activating opioid receptor activity.
Background
Opioid receptors are important G Protein Coupled Receptors (GPCRs) which are widely distributed in central and peripheral nervous systems, mainly comprise three subtypes of mu, kappa and kappa, and are targets of analgesic effects of opioid drugs and endogenous opioid peptides. Conventional opioid agonists, such as morphine and its derivatives, which act primarily on the mu receptor activation, are the most effective drugs for the treatment of chronic arthritis, inflammatory neuralgia, postoperative pain, and moderate to severe pain caused by various cancers. These traditional drugs can cause side effects such as respiratory depression, gastrointestinal adverse reactions, drug addiction, confusion and tolerance problems, etc., which are closely related to the function of beta-arrestin (beta-arrestin).
Studies have found that opioid receptors mediate and regulate physiological functions primarily through the G protein pathway and the β -arrestin pathway. The stimulation of the mu receptor can not only produce the analgesic effect, but also have obvious respiratory depression and gastrointestinal adverse reactions such as constipation, nausea and the like. Animal models of gene knockout demonstrate: side effects such as analgesia and respiratory depression are achieved through two different signal pathways. Among them, the analgesic action is achieved by activating the downstream signal pathway of G protein, and promotion of recruitment of β -arrestin (β -arrestin) leads to activation of the downstream signal pathway thereof to produce side effects such as respiratory depression and constipation. After morphine is injected into a beta-arrestin 2 gene knockout mouse, the analgesic effect is stronger and the maintenance time is longer. This result is a good indication of the effectiveness and safety of the G protein pathway (Bohn et al, Science, 1999).
Therefore, there is still a need in the field of analgesic research to develop novel opioid receptor agonists that act selectively on the G protein pathway with fewer side effects.
Disclosure of Invention
The present inventors have discovered a class of oxaspiro compounds that selectively act on the G protein pathway to activate opioid receptors, and have made this application based on these findings.
Accordingly, in one aspect, the present application provides a compound of formula (I), a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination of the foregoing,
Figure PCTCN2019082841-APPB-000001
wherein R is1Selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or, R1Form a ring with ring A;
R2selected from the group consisting of hydroxy, cyano, halogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, alkenyl, haloalkenyl, alkynyl, haloalkynyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
R3Selected from the group consisting of hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; or, R3And R2Connecting to form a ring;
Raselected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
Rband RcIndependently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, and sulfonyl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, sulfonyl is optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
w and U are independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-NRf-、-(CRdRe)m1-C(=O)-、-(CRdRe)m1-S(O)q-、-C(=O)-NRf-and-C (═ O) -;
v is selected from C and N;
Rd、Reand RfIndependently selected from hydrogen, hydroxy, halogen, alkyl, alkoxy, cycloalkyl, and heterocycloalkyl; or, RdAnd ReForm a ring with the attached carbon atom;
m1selected from 1,2,3 and 4;
ring a is selected from the group consisting of aromatic, heteroaromatic, aliphatic carbocycle, and lipoheterocycle;
m and n are independently selected from 0, 1,2,3,4 and 5;
x and y are independently selected from 1,2,3 and 4;
q is selected from 0, 1 and 2.
In certain embodiments, R1Selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or, R1Form a loop with ring a.
In other embodiments, R1Is a heterocycloalkyl group.
In certain embodiments, R2Selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
In other embodiments, R2Selected from hydrogen and alkenyl.
In certain preferred embodiments, the compounds have the structure of formula (II),
Figure PCTCN2019082841-APPB-000002
wherein R is1Selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or, R1Form a ring with ring A;
R2selected from the group consisting of hydroxy, cyano, halogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, alkenyl, haloalkenyl, alkynyl, haloalkynyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
R3Selected from the group consisting of hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; or, R3And R2Connecting to form a ring;
Raselected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
Rband RcIndependently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, and sulfonyl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, sulfonyl is optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
w and U are independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-S(O)q-、-(CRdRe)m1-NRf-and-C (═ O) -;
Rd、Reand RfIndependently selected from hydrogen, hydroxy, halogen, alkyl and alkoxy; or, RdAnd ReIs connected withThe carbon atom of (a) forms a ring;
m1selected from 1,2,3 and 4;
ring a is selected from the group consisting of aromatic, heteroaromatic, aliphatic carbocycle, and lipoheterocycle;
m and n are independently selected from 0, 1,2,3,4 and 5;
x and y are independently selected from 1,2,3 and 4;
q is selected from 0, 1 and 2.
In certain embodiments, in said formula (II),
R1selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or, R1Form a ring with ring A;
R2selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
R3Selected from the group consisting of hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; or, R3And R2Connecting to form a ring;
Raselected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
Rband RcIndependently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, and sulfonyl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, sulfonyl is optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
w and U are independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-and- (CR)dRe)m1-NRf-;
Rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, alkyl and alkoxy; or, RdAnd ReForm a ring with the attached carbon atom;
m1selected from 1,2,3 and 4;
ring a is selected from the group consisting of aromatic, heteroaromatic, aliphatic carbocycle, and lipoheterocycle;
m and n are independently selected from 0, 1,2,3,4 and 5;
x and y are independently selected from 1,2,3 and 4.
In certain preferred embodiments, the compounds have the structure of formula (III),
Figure PCTCN2019082841-APPB-000003
wherein the groups and substituents are as defined above.
In certain preferred embodiments, the compounds have a structure represented by formula (III-1) or formula (III-2),
Figure PCTCN2019082841-APPB-000004
wherein the groups and substituents are as defined above.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-S(O)q-and-C (═ O) -; rdAnd ReEach independently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReTo the carbon atom to which they are attached3-7 membered lipoheterocyclic; m is1Selected from 1 and 2; q is selected from 0, 1 and 2; x is selected from 1,2 and 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-SO2-and-C (═ O) -; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl radical, C1-4Alkoxy radical, C3-6Cycloalkyl and 3-6 membered heterocycloalkyl; m is1Is 1 or 2; x is selected from 1,2 and 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-SO2-and-C (═ O) -; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; x is 1 or 2.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from-CH2-、-CH2CH2-、-CH2-O-、-CH2-SO2-and-C (═ O) -; x is 1.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-O-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; x is 1,2 or 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-O-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; x is 1,2 or 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000005
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; x is 1,2 or 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000006
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; x is 1 or 2.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000007
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; x is 1 or 2.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-NRf-、-(CRdRe)m1-C(=O)-、-(CRdRe)m1-S(O)q-, -C (═ O) -, and-C (═ O) -NRf-;Rd、ReAnd RfIndependently selectFrom hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; q is 0, 1 or 2; x is 1,2 or 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-NRf-、-(CRdRe)m1-C(=O)-、-(CRdRe)m1-S(O)q-, -C (═ O) -, and-C (═ O) -NRf-;Rd、 ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; q is 0, 1 or 2; x is 1,2 or 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000008
Figure PCTCN2019082841-APPB-000009
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; q is 0, 1 or 2; x is 1,2 or 3.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000010
Figure PCTCN2019082841-APPB-000011
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; q is 0, 1 or 2; x is 1 or 2.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000012
Figure PCTCN2019082841-APPB-000013
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rd、ReAnd RfIndependently selected from hydrogen, hydroxy, fluoro, chloro, methyl and ethyl; m is1Is 1 or 2; q is 0, 1 or 2; x is 1 or 2.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000014
Wherein the 1 position is connected with the 3 position in the general formula, and the 2 position is connected with the 4 position in the general formula; x is 1 or 2.
In certain embodiments, W in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000015
and-C (═ O) -, where the 1 position is attached to the 3 position in the formula and the 2 position is attached to the 4 position in the formula; x is 1.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-and- (CR)dRe)m1-O-;RdAnd ReEach independently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReTo the carbon atom to which it is attached to form a 3-7 membered heterocyclic ring; m is1Selected from 1,2,3 and 4; q is selected from 0, 1 and 2; y is selected from 1,2 and 3.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-and- (CR)dRe)m1-O-;RdAnd ReEach independently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-6Cycloalkyl and 3-6 membered heterocycloalkyl; or, RdAnd ReTo the carbon atom to which it is attached to form a 3-6 membered heterocyclic ring; m is1Selected from 1 and 2; y is selected from 1,2 and 3.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; y is selected from 1 or 2.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from-CH2-and-CH2CH2-; y is 1.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-O-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; m is1Is 1,2 or 3; y is 1,2 or 3.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from- (CR)dRe)m1-O-;RdAnd ReIs independently selected fromHydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1,2 or 3; y is 1,2 or 3.
In certain embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000016
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 5 in the general formula; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; y is 1,2 or 3.
In certain preferred embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000017
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 5 in the general formula; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; y is 1 or 2.
In certain preferred embodiments, U in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from
Figure PCTCN2019082841-APPB-000018
Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 5 in the general formula; y is 1 or 2.
In certain preferred embodiments, W and U in each of formula I, formula II, formula III-1, and formula III-2 are each independently selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; m is1Is 1 or 2; x and y are independently selected from1.2 and 3.
In certain preferred embodiments, W and U in each of formula I, formula II, formula III-1, and formula III-2 are each independently selected from- (CR)dRe)m1-; wherein R isdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; x and y are independently selected from 1,2 and 3.
In certain preferred embodiments, W and U in each of formula I, formula II, formula III-1, and formula III-2 are each independently selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; x and y are independently 1 or 2.
In certain preferred embodiments, W and U in each of formula I, formula II, formula III-1, and formula III-2 are each independently selected from-CH2-and-CH2CH2-; x and y are independently 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or R1Form a fused ring with ring a, said fused ring being a ring a and 5-6 membered lipoheterocyclic ring or a ring a and 5-6 membered heteroaromatic ring;
Raselected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
m is selected from 0, 1,2,3,4 and 5.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl radical, C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or R1Form a fused ring with ring a, said fused ring being a ring a and 5-6 membered lipoheterocyclic ring or a ring a and 5-6 membered heteroaromatic ring;
Raselected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-to 10-membered heteroaryl,C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
m is selected from 0, 1,2,3,4 and 5.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from hydroxy, cyano, halogen, C1-6Alkyl and-ORa(ii) a Or, R1Form a fused ring with ring A, said fused ring is ring A and 5-6 membered lipoheterocycle;
Raselected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
m is selected from 0, 1,2,3 and 4.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from hydroxy, cyano, halogen, C1-4Alkyl and-ORa(ii) a Or, R1Form a fused ring with ring A, said fused ring is ring A and 5-6 membered lipoheterocycle;
Raselected from hydrogen, C1-4Alkyl and 5-6 membered heterocycloalkyl; wherein, said C1-4Alkyl and 5-6 membered heterocycloalkyl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-4Alkyl and halo C1-4An alkyl group;
m is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from hydroxy, cyano, fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butylSec-butyl, isobutyl, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, isobutoxy and tetrahydrofuryloxy; or, R1Form a fused ring with ring A, wherein the fused ring is ring A and 1, 4-dioxane;
m is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from hydroxy, cyano, fluoro, chloro, methyl, ethyl, methoxy, ethoxy, n-propoxy and tetrahydrofuryloxy; or, R1Form a fused ring with ring A, wherein the fused ring is ring A and 1, 4-dioxane;
m is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, fluoro, methoxy and tetrahydrofuryloxy; or, R1Form a fused ring with ring A, wherein the fused ring is ring A and 1, 4-dioxane;
m is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, halogen and-ORa(ii) a Or, R1Form a fused ring with ring A, said fused ring is ring A and 5-6 membered lipoheterocycle;
Raselected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
m is selected from 0, 1,2,3 and 4;
in certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, halogen and-ORa(ii) a Or, R1Form a loop with the ring A, saidAnd the ring is ring A and 5-6 membered lipoheterocycle;
Raselected from hydrogen, C1-4Alkyl and 5-6 membered heterocycloalkyl; wherein, said C1-4Alkyl and 5-6 membered heterocycloalkyl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-4Alkyl and halo C1-4An alkyl group;
m is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, fluoro, chloro, bromo, iodo, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, isobutoxy, and tetrahydrofuranyloxy; or, R1Form a fused ring with ring A, wherein the fused ring is ring A and 1, 4-dioxane;
m is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, halogen and 3-6 membered heterocycloalkyl;
m is 0, 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, halogen and 5-6 membered heterocycloalkyl;
m is 0, 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-21Independently selected from cyano, fluoro and morpholinyl;
m is 0 or 1.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C1-6Hydroxyalkyl, -C1-4alkyl-O-C1-4Alkyl radical, C3-8Cycloalkyl, 4-8 membered heterocycloalkyl, C2-6Alkenyl, halo C2-6Alkenyl radical, C2-6Alkynyl, halo C2-6Alkynyl, C6-10Aryl, 5-to 10-membered heteroarylRadical, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
RaSelected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl radical, C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
n is selected from 0, 1,2,3,4 and 5.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C1-6Hydroxyalkyl, -C1-4alkyl-O-C1-4Alkyl radical, C3-8Cycloalkyl, 4-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
RaSelected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl radical, C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
n is selected from 0, 1,2,3,4 and 5.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen, C1-4Alkyl, halo C1-4Alkyl radical, C1-4Hydroxyalkyl radical, C3-6Cycloalkyl, -C1-4alkyl-O-C1-4Alkyl, -ORaand-CO2Ra
RaSelected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
n is selected from 0, 1,2,3 and 4.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen, C1-4Alkyl, halo C1-4Alkyl radical, C1-4Hydroxyalkyl radical, C3-6Cycloalkyl, -C1-3alkyl-O-C1-3Alkyl, -ORaand-CO2Ra
RaSelected from hydrogen, C1-4Alkyl and 5-6 membered heterocycloalkyl; wherein, said C1-4Alkyl and 5-6 membered heterocycloalkyl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-4Alkyl and halo C1-4An alkyl group;
n is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, isobutyl, fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, -CH2OCH3Cyclopropyl, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, isobutoxy, tetrahydrofuryloxy and-C (O) OCH3
n is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, fluoro, chloro, methyl, ethyl, n-propyl, isopropyl, fluoromethyl, difluoromethyl, hydroxymethyl, hydroxyethyl, -CH2OCH3Cyclopropyl, methoxy, ethoxy, tetrahydrofuryloxy and-C (O) OCH3
n is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl radical, C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
RaSelected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl radical, C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
n is selected from 0, 1,2,3,4 and 5.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen and-ORa
RaSelected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
n is selected from 0, 1,2,3 and 4.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, halogen and-ORa
RaSelected from hydrogen, C1-4Alkyl and 5-6 membered heterocycloalkyl; wherein, said C1-4Alkyl and 5-6 membered heterocycloalkyl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-4Alkyl and halo C1-4An alkyl group;
n is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Independently selected from hydroxy, cyano, fluoro, chloro, bromo, iodo, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, isobutoxy, and tetrahydrofuryloxy;
n is 1 or 2.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-22Selected from the group consisting of hydroxy, methoxy and tetrahydrofuryloxy;
n is 1 or 2.
In certain preferred embodiments, R in formula I and/or formula II and/or formula III-1 and/or formula III-22Independently selected from C2-6Alkenyl, halo C2-6Alkenyl radical, C2-6Alkynyl or halo C2-6Alkynyl.
In certain preferred embodiments, R in formula I and/or formula II and/or formula III-1 and/or formula III-22Independently selected from C2-4Alkenyl, halo C2-4Alkenyl radical, C2-4Alkynyl or halo C2-4Alkynyl.
In certain preferred embodiments, R in formula I and/or formula II and/or formula III-1 and/or formula III-22Independently selected from ethenyl, propenyl, fluoroethenyl, 1-difluoroethenyl, 2-methylpropenyl, ethynyl, propynyl, fluoroethynyl. In certain preferred embodiments, R in formula I, formula II, formula III-1, and formula III-22Independently selected from hydroxy, C1-6Alkyl radical, C1-6Hydroxyalkyl and C2-6An alkenyl group;
n is 0, 1 or 2.
In certain preferred embodiments, R in formula I, formula II, formula III-1, and formula III-22Independently selected from hydroxy, C1-4Alkyl radical, C1-4Hydroxyalkyl and C2-4An alkenyl group;
n is 0, 1 or 2.
In certain preferred embodiments, R in formula I, formula II, formula III-1, and formula III-22Independently selected from the group consisting of hydroxy, methyl, hydroxymethyl, and vinyl;
n is 0 or 1.
In certain preferred embodiments, the ring A in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from C6-10Aromatic ring, 5-10 membered heteroaromatic ring, C3-8Aliphatic carbocycles and 3-8 membered aliphatic heterocycles.
In certain preferred embodiments, the ring A in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from C6-10Aromatic rings and 5-10 membered heteroaromatic rings.
In certain preferred embodiments, ring A in each of formulas I, II, III-1, and III-2 is independently selected from the group consisting of a phenyl ring and a 5-6 membered heteroaryl ring.
In certain preferred embodiments, ring a in each of formulas I, II, III-1, and III-2 is independently selected from the group consisting of a benzene ring, a pyrrole ring, a furan ring, a thiophene ring, an oxazole ring, an imidazole ring, a thiazole ring, a pyridine ring, a pyrimidine ring, a pyrazine ring, a pyridazine ring, a 1,2, 4-1H-triazole ring, and a pyrazole ring.
In certain preferred embodiments, ring a in each of formula I, formula II, formula III-1, and formula III-2 is independently selected from the group consisting of a benzene ring, a pyrrole ring, a furan ring, a thiophene ring, an oxazole ring, an imidazole ring, a thiazole ring, a pyridine ring, a pyrimidine ring, a pyrazine ring, and a pyridazine ring.
In certain preferred embodiments, ring A in each of formulas I, II, III-1, and III-2 is independently selected from the group consisting of a benzene ring and a thiophene ring.
In certain embodiments, ring A in each of formulas I, II, III-1, and III-2 is independently a phenyl ring.
In certain preferred implementationsIn the scheme, R in each formula I, formula II, formula III-1 and formula III-23Independently selected from hydrogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-23Independently selected from hydrogen, hydroxy, C1-6Alkyl and halo C1-6An alkyl group.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-23Independently selected from hydrogen and C1-4An alkyl group.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-23Independently selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-23Independently selected from hydrogen, methyl, ethyl, n-propyl and isopropyl.
In certain preferred embodiments, R in each of formulas I, II, III-1, and III-23Is hydrogen.
In certain preferred embodiments, the compound is selected from:
Figure PCTCN2019082841-APPB-000019
Figure PCTCN2019082841-APPB-000020
Figure PCTCN2019082841-APPB-000021
Figure PCTCN2019082841-APPB-000022
Figure PCTCN2019082841-APPB-000023
in certain preferred embodiments, the compound is selected from:
Figure PCTCN2019082841-APPB-000024
Figure PCTCN2019082841-APPB-000025
Figure PCTCN2019082841-APPB-000026
Figure PCTCN2019082841-APPB-000027
Figure PCTCN2019082841-APPB-000028
in certain preferred embodiments, the compound is selected from:
Figure PCTCN2019082841-APPB-000029
Figure PCTCN2019082841-APPB-000030
Figure PCTCN2019082841-APPB-000031
Figure PCTCN2019082841-APPB-000032
Figure PCTCN2019082841-APPB-000033
in certain embodiments, the pharmaceutically acceptable salt of the compound is trifluoroacetate, formate.
In another aspect, the present application provides a pharmaceutical composition comprising a compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, as described above; optionally, it further contains one or more pharmaceutical excipients.
In embodiments of the present application, the pharmaceutical composition may be administered in any of the following ways: oral, aerosol inhalation, rectal, nasal, buccal, topical, parenteral, e.g. subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal and intracranial injection or infusion, or via an external reservoir. Among them, oral, intraperitoneal or intravenous administration is preferable.
When administered orally, the compounds of the present application may be formulated in any orally acceptable dosage form, including but not limited to tablets, capsules, aqueous solutions or suspensions. Among these, carriers for tablets generally include lactose and corn starch, and additionally, lubricating agents such as magnesium stearate may be added. Diluents used in capsule formulations generally include lactose and dried corn starch. Aqueous suspension formulations are generally prepared by mixing the active ingredient with suitable emulsifying and suspending agents. If desired, sweetening, flavoring or coloring agents may be added to the above oral dosage forms.
When the compound is used for local administration, particularly for treating affected surfaces or organs which are easy to reach by local external application, such as eyes, skin or lower intestinal tract neurogenic diseases, the compound can be prepared into different local administration preparation forms according to different affected surfaces or organs, and the specific description is as follows:
when administered topically to the eye, the compounds of the present application may be formulated as a micronized suspension or solution in sterile saline at a pH that is isotonic, with or without the addition of preservatives such as benzylalkenoxides. For ophthalmic use, the compounds may also be formulated in the form of ointments such as vaseline.
When applied topically to the skin, the compounds of the present application can be formulated in a suitable ointment, lotion, or cream formulation, in which the active ingredient is suspended or dissolved in one or more carriers. Carriers that may be used in ointment formulations include, but are not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyethylene oxide, polypropylene oxide, emulsifying wax and water; carriers that can be used in lotions or creams include, but are not limited to: mineral oil, sorbitan monostearate, tween 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
The compounds of the present application may also be administered in the form of sterile injectable preparations, including sterile injectable aqueous or oleaginous suspensions or solutions. Among the carriers and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, the sterilized fixed oil may also be employed as a solvent or suspending medium, such as a monoglyceride or diglyceride.
In another aspect, the present application provides the use of a compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition thereof, as described hereinbefore, for the manufacture of a medicament for activating the activity of an opioid receptor (e.g., a μ -opioid receptor).
In another aspect, the present application provides a compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition as described hereinbefore for use in activating opioid receptor (e.g., μ -opioid receptor) activity.
In another aspect, the present application provides a method of activating opioid receptor (e.g., μ -opioid receptor) activity in a subject, comprising the step of administering to the subject an effective amount of a compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition as described above.
In another aspect, the present application provides the use of a compound, solvate, stereoisomer, crystal form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition as described hereinbefore, in the manufacture of an analgesic medicament.
In another aspect, the present application provides a compound, solvate, stereoisomer, crystal form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition as described hereinbefore for use in analgesia.
In another aspect, the present application provides a method of treating pain, comprising the step of administering to a subject in need thereof an effective amount of a compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition as described above.
In another aspect, the present application provides a method of preparing a compound as described hereinbefore, comprising the steps of:
Figure PCTCN2019082841-APPB-000034
wherein R is1、R2、R3W, U, V, A, m, n, x, y are as previously described;
the compound of formula I is prepared by carrying out reductive amination reaction on the compound of formula I-A and the compound of formula I-B. The reductive amination reaction can be carried out with reference to experimental conditions commonly employed in the art. In certain preferred embodiments, an acid and/or a reducing agent is added to the reductive amination reaction. In certain preferred embodiments, the acid is selected from AcOH and TFA.
In certain preferred embodiments, the reducing agent is selected from NaBH4、NaCNBH3And NaBH (OAc)3
In certain preferred embodiments, the compounds are prepared by a process comprising the steps of:
Figure PCTCN2019082841-APPB-000035
wherein R is1、R2、R3W, U, A, m, n, x, y are as described above;
the compound of formula II is prepared by the reductive amination reaction of the compound of formula I-A and the compound of formula II-B. The reductive amination reaction can be carried out with reference to experimental conditions commonly employed in the art. In certain preferred embodiments, an acid and/or a reducing agent is added to the reductive amination reaction. In certain preferred embodiments, the acid is selected from AcOH and TFA. In certain preferred embodiments, the reducing agent is selected from NaBH4、NaCNBH3And NaBH (OAc)3
As used herein, the term "solvate" means a substance formed by combining a compound of the present application with a pharmaceutically acceptable solvent. Pharmaceutically acceptable solvents include water, ethanol, acetic acid and the like. Solvates include stoichiometric and non-stoichiometric amounts of solvates, preferably hydrates.
As used herein, the term "stereoisomer" includes conformational isomers and configurational isomers, wherein the configurational isomers include primarily cis-trans isomers and optical isomers. The compounds described herein may exist in stereoisomeric forms and thus encompass all possible stereoisomeric forms, and any combinations or any mixtures thereof. For example, a single enantiomer, a single diastereomer or a mixture thereof. When a compound described herein contains an olefinic double bond, it includes both cis and trans isomers, and any combination thereof, unless otherwise specified.
In addition, molecules, atoms or ions of partial natural or artificial compounds can be arranged in a certain regular periodic repetition mode in space, the arrangement has periodicity of three-dimensional space, and the molecules, atoms or ions can repeatedly appear at a certain distance. In this case, the compounds may exist in two or more crystalline states, molecules of the same structure, crystallized in different solid forms, called polymorphs or polymorphs (polymorphs). When referring to a particular crystalline form, it is often referred to as "crystal form", that is the term "crystalline form" as used in this application.
As used herein, the term "pharmaceutically acceptable salt" means a salt of a compound of the present application that is pharmaceutically acceptable and that has the pharmacological activity of the parent compound. Such salts include: acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with organic acids; such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, trifluoroacetic acid, formic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or salts formed when an acidic proton present on the parent compound is replaced by a metal ion, e.g., an alkali metal ion or an alkaline earth metal ion; or a complex compound with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, or the like.
As used herein, the term "pharmaceutically acceptable ester" refers to an ester formed by esterification of a compound of the present application with an alcohol when a carboxyl group is present; when a hydroxyl group is present in the compound of the present application, an ester is formed by an esterification reaction with an organic acid, an inorganic acid, an organic acid salt, or the like. The ester can be hydrolyzed in the presence of acid or alkali to generate corresponding acid or alcohol.
As used herein, the term "cycloalkyl" means a saturated cyclic hydrocarbon group which may be a monocyclic or polycyclic fused system, andand may be fused to an aromatic ring. E.g. C3-8Cycloalkyl radical, C3-6Cycloalkyl radical, C4-6Cycloalkyl radical, C5-6Cycloalkyl groups, and the like. Examples of such groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
As used herein, the term "heterocycloalkyl" refers to a monocyclic or bicyclic saturated or partially saturated cyclic group, such as 3-8 membered heterocycloalkyl, 3-6 membered heterocycloalkyl, 4-6 membered heterocycloalkyl, or 5-6 membered heterocycloalkyl, optionally substituted with at least one and up to four heteroatoms independently selected from N, O or S. Examples of such groups include, but are not limited to, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, piperidinyl, morpholinyl, piperazinyl, or the like.
As used herein, the term "aryl" refers to a monocyclic or bicyclic aromatic group comprising at least one aromatic ring, preferably C6-10Aryl, i.e. aryl of 6,7,8,9 or 10 carbon atoms. Examples of the aromatic group in the present application include phenyl, naphthyl, 1,2,3, 4-tetrahydronaphthyl, indenyl and the like.
As used herein, the term "heteroaryl" refers to a monocyclic or bicyclic aromatic ring group, preferably a 5-10 membered heteroaryl, a 5-6 membered heteroaryl, and the like, optionally substituted with at least one heteroatom independently selected from N, O or S. Examples of such groups include, but are not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyrimidinyl, triazinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, benzofuranyl, benzothienyl, indolyl, isoindolyl, pyridazinyl, pyrazinyl, quinolinyl, and the like.
As used herein, the term "alkyl" refers to a straight or branched chain saturated hydrocarbon group. The term "C1-6Alkyl "means a straight or branched chain alkyl group having 1 to 6, i.e. 1,2,3,4, 5 or 6 carbon atoms, typically methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, neopentyl, pentyl, hexyl and the like. Similarly, the term "C1-4Alkyl "means a straight or branched chain alkyl group having 1,2,3 or 4 carbon atoms, including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, and the like.
As used herein, the term "halogen" refers to fluorine, chlorine, bromine, and iodine.
As used herein, the term "haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein the alkyl group is as previously described. For example halo C1-6Alkyl, halo C1-4Alkyl groups, and the like. Specific examples include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, fluoroethyl, chloroethyl, and the like.
As used herein, the term "hydroxyalkyl" refers to an alkyl group substituted with one or more hydroxyl groups, wherein the alkyl group is as described previously. For example, C1-6 hydroxyalkyl, C1-4 hydroxyalkyl. Specific examples include, but are not limited to, hydroxymethyl, 1-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 1- (hydroxymethyl) -2-methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2, 3-dihydroxypropyl, 2-hydroxy-1-hydroxymethylethyl, 2, 3-dihydroxybutyl, 3, 4-dihydroxybutyl and 2- (hydroxymethyl) -3-hydroxypropyl.
As used herein, the term "alkoxy" refers to a group having the structure alkyl-O-, wherein the alkyl is as described previously. E.g. C1-6Alkoxy radical, C1-4Alkoxy, and the like. Specific examples include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy, and the like.
As used herein, the term "acyl" refers to a group having the structure alkyl-c (o) -wherein the alkyl is as described previously. E.g. C1-6Alkanoyl radical, C1-4Alkanoyl groups, and the like. Specific examples include, but are not limited to, formyl, acetyl, n-propionyl, isopropionyl, n-butyryl, isobutyryl, t-butyryl, and the like.
As used herein, the term "sulfonyl" refers to a compound having an alkyl-S (O)2-a group of structure (la) wherein the alkyl group is as described hereinbefore. E.g. C1-6Alkylsulfonyl radical, C1-4Alkylsulfonyl and the like. Specific examples include, but are not limited to, methylsulfonyl, ethylsulfonyl, n-propylsulfonyl, isopropylsulfonyl, n-butylsulfonyl, isobutylsulfonyl, sec-butylsulfonyl, tert-butylsulfonyl, and the like.
As used herein, the term "aromatic ring" refers to a monocyclic or polycyclic ring system comprising at least one aromatic ring. E.g. C6-10Aromatic rings, i.e. aromatic rings of 6,7,8,9 or 10 carbon atoms. Examples of the aromatic ring in the present application include a benzene ring, a naphthalene ring, a 1,2,3, 4-tetrahydronaphthalene ring, an indene ring and the like.
As used herein, the term "heteroaromatic ring" refers to an aromatic monocyclic or multicyclic ring system comprising 5 to 14 ring atoms, wherein 1 to 4 ring atoms are independently O, N or S, and the remaining ring atoms are carbon atoms. Such as 5-10 membered heteroaromatic rings, 5-6 membered heteroaromatic rings, and the like. Specific examples include, but are not limited to, a pyrrole ring, a furan ring, a thiophene ring, an imidazole ring, a pyrazole ring, a triazole ring, a tetrazole ring, an oxazole ring, an isoxazole ring, an oxadiazole ring, a thiazole ring, an isothiazole ring, a thiadiazole ring, a pyridine ring, a pyrimidine ring, a triazine ring, a benzimidazole ring, a benzoxazole ring, a benzothiazole ring, a benzofuran ring, a benzothiophene ring, an indole ring, an isoindole ring, a pyridazine ring, a pyrazine ring, a quinoline ring, and the like.
As used herein, the term "aliphatic carbocycle" refers to a saturated or partially saturated non-aromatic hydrocarbon ring, which may be a monocyclic or polycyclic fused system. E.g. C3-8Aliphatic carbocyclic ring, C3-6Aliphatic carbocyclic ring, C3-5Aliphatic carbocycles, and the like. Specific examples include, but are not limited to, cyclopropane rings, cyclobutane rings, cyclopentane rings, and cyclohexane rings.
As used herein, the term "aliphatic heterocycle" refers to a saturated or partially saturated, non-aromatic, monocyclic or polycyclic ring optionally substituted with at least one and up to four heteroatoms independently selected from N, O or S. For example, 3-to 8-membered aliphatic heterocyclic ring, 3-to 6-membered aliphatic heterocyclic ring, 3-to 5-membered aliphatic heterocyclic ring, etc. Specific examples include, but are not limited to, pyrrolidine ring, tetrahydrofuran ring, dihydrofuran ring, tetrahydrothiophene ring, piperidine ring, morpholine ring, piperazine ring, or the like.
When a group is described as "optionally substituted with one or more substituents selected from the group consisting of" the group may be (1) unsubstituted or (2) substituted. If a carbon on a group is described as optionally substituted with one or more substituents selected from the group consisting of those described below, one or more hydrogens on the carbon (to the extent of any hydrogens present) may be replaced individually and/or together with an independently selected optional substituent. If a nitrogen on a group is described as being optionally substituted with one or more of the following substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogen is present) may each be replaced with an independently selected optional substituent.
The invention also includes pharmaceutically acceptable isotopic compounds of the compounds which are identical in structure to the compounds of the invention, except that one or more atoms are replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g. hydrogen)2H、3H) (ii) a Isotopes of carbon (e.g. of11C、13C and14C) (ii) a Isotopes of chlorine (e.g. of chlorine)36Cl); isotopes of fluorine (e.g. of fluorine)18F) (ii) a Isotopes of iodine (e.g. of iodine)123I and125I) (ii) a Isotopes of nitrogen (e.g. of13N and15n); isotopes of oxygen (e.g. of15O、17O and18o); isotopes of phosphorus (e.g. of phosphorus)32P); and isotopes of sulfur (e.g. of35S)。
As used herein, the term "pharmaceutical excipient" refers to excipients and additives used in the manufacture of pharmaceutical products and in the formulation of pharmaceutical formulations, and refers to substances which have been reasonably evaluated in terms of safety and which are included in pharmaceutical formulations, in addition to active ingredients. The pharmaceutic adjuvant has important functions of solubilization, dissolution assistance, sustained and controlled release and the like besides excipient, carrier and stability improvement, and is an important component which may influence the quality, safety and effectiveness of the medicine. They can be classified into natural, semi-synthetic and total synthetic ones according to their origin. According to their action and use, they can be divided into: solvents, propellants, solubilizers, solubilizing agents, emulsifiers, colorants, adhesives, disintegrants, fillers, lubricants, wetting agents, osmotic pressure regulators, stabilizers, glidants, flavoring agents, preservatives, suspending agents, coating materials, fragrances, anti-adherents, antioxidants, chelating agents, permeation promoters, pH regulators, buffers, plasticizers, surfactants, foaming agents, antifoaming agents, thickeners, encapsulating agents, humectants, absorbents, diluents, flocculants and deflocculants, filter aids, release retardants, and the like; according to the administration route, the medicine can be divided into oral administration, injection, mucous membrane, percutaneous or local administration, nasal or oral inhalation administration, ocular administration and the like. The same pharmaceutic adjuvant can be used for pharmaceutic preparations of different administration routes and has different functions and purposes.
As used herein, the term "subject" refers to an animal, particularly a mammal, preferably a human.
As used herein, the term "effective amount" refers to an amount sufficient to obtain, or at least partially obtain, a desired effect. For example, a prophylactically effective amount is an amount sufficient to prevent, or delay the onset of disease; a therapeutically effective amount is an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. It is well within the ability of those skilled in the art to determine such effective amounts. For example, an amount effective for therapeutic use will depend on the severity of the disease to be treated, the general state of the patient's own immune system, the general condition of the patient, e.g., age, weight and sex, the mode of administration of the drug, and other treatments administered concurrently, and the like.
Detailed Description
Embodiments of the present invention will be described in detail below with reference to examples, but those skilled in the art will appreciate that the following examples are only illustrative of the present invention and should not be construed as limiting the scope of the present invention. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
The structures of the compounds described in the following examples were determined by nuclear magnetic resonance (1H NMR) or Mass Spectrometry (MS).
Nuclear magnetic resonance (1H NMR) was performed using a Bruker 400MHz nuclear magnetic resonance apparatus; the solvent was determined to be deuterated methanol (CD)3OD), deuterated chloroform (CDCl)3) Hexadeuterio dimethyl sulfoxide (DMSO-d)6) (ii) a The internal standard substance is Tetramethylsilane (TMS). All values are expressed in ppm.
Abbreviations in Nuclear Magnetic Resonance (NMR) spectra used in the examples are shown below:
s: unimodal (singlet), d: doublet (doublt), t: triplet (triplet), q: quartet (quartz), dd: doublet (doubledoubledoublet), qd: quartet doubtet, ddd: double doublet (double doublet), ddt: double double triplet (double double triple), dddd: double double doublet (double double doublet), m: multiplet (multiplex), br: broad (broad), J: coupling constant, Hz: hertz, DMSO-d6: deuterated dimethyl sulfoxide.
Mass Spectrometry (MS) was performed using an Agilent (ESI) mass spectrometer, model Agilent 6120B.
The compounds of the examples were purified using preparative liquid phase (Prep-HPLC) methods including methods A, B and C.
Method a for preparing liquid phase purification:
liquid chromatography instrument model: agilent 1260;
a chromatographic column: waters SunAire Prep C18 OBD (19 mm. times.150 mm. times.5.0 μm);
temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214nm and 254 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous formic acid.
Method B for preparation of liquid phase purification:
liquid chromatography instrument model: agilent 1260;
a chromatographic column: waters SunAire Prep C18 OBD (19 mm. times.150 mm. times.5.0 μm);
temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214nm and 254 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous trifluoroacetic acid.
Method C for preparation of liquid phase purification:
liquid chromatography instrument model: agilent 1260;
a chromatographic column: waters Xbridge Prep C18 OBD (19 mm. times.150 mm. times.5.0 μm);
temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214nm and 254 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous ammonium bicarbonate solution.
The first embodiment is as follows: preparation of N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-indene-2-amine
Figure PCTCN2019082841-APPB-000036
The method comprises the following steps: synthesis of 2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde
Reacting 2- (9- (pyridin-2-yl) -6-oxaspiro [4.5]]Decan-9-yl) acetonitrile (1.55g,6.05mmol) was added to dry toluene (35mL) and cooled to-78 ℃ under nitrogen. Diisobutylaluminum hydride (1.28g,9.07mmol) was added dropwise. After the completion of the dropwise addition, the mixture was stirred at-78 ℃ for 2 hours, and after completion of the reaction was monitored by LCMS, a saturated aqueous ammonium chloride solution was added to the reaction mixture, and the mixture was warmed to room temperature and stirred for 1 hour. 6N hydrochloric acid was added to the reaction solution, and the mixture was stirred for 6 hours while the temperature was raised to 85 ℃. Cooling the reaction solution to room temperature, adding sodium bicarbonate, adjusting pH to 8, separating, concentrating the aqueous phase to dryness, adding DCM and anhydrous sodium sulfate, stirring for 30 minutes, filtering, and concentrating to obtain 2- (9- (pyridin-2-yl) -6-oxaspiro [4.5]]Decan-9-yl) acetaldehyde crude 1.2 g. ESI-MS (M/z) 260.16[ M + H]+
Step two: synthesis of N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-indene-2-amine
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (80mg,0.308mmol) and 2-aminoindene hydrochloride (49.3mg,0.291mmol) were added to dichloromethane (15mL) under nitrogen, anhydrous magnesium sulfate (222.78mg,1.85mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (23.26mg,0.37mmol) and methanol (5mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. The reaction was quenched by addition of methanol (15mL), filtered, and the solvent was evaporated under reduced pressure to give 150mg of crude product. After purification from the preparative liquid phase (preparative liquid phase purification using method A) lyophilized to give the title compound as formate salt 9.53 mg.
ESI-MS(m/z):377.2[M+H]+
1H NMR(400MHz,CDCl3)8.50(m,1H),8.29(s,1H),7.67(m,1H),7.33(m,1H),7.20–7.09(m,5H),3.84–3.60(m,3H),3.13(m,2H),3.00(m,2H),2.82(m,1H),2.52–2.41(m,1H),2.40–2.26(m,2H),2.18(m,1H),2.03–1.88(m,2H),1.84–1.57(m,3H),1.56–1.30(m,4H),1.15–1.01(m,1H),0.65m,1H).
Example two: synthesis of N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -1,2,3, 4-tetrahydronaphthalen-2-amine
Figure PCTCN2019082841-APPB-000037
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (75mg,0.289mmol) and 1,2,3, 4-tetrahydronaphthalen-2-amine (75.55mg,0.289mmol) were added to dichloromethane (15mL) under nitrogen protection, anhydrous magnesium sulfate (348.09mg,2.89mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (90.87mg,1.45mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. The raw materials are not completely reacted and products are generated by LC-MS detection. Direct post-treatment, adding diatomite, filtering, concentrating the filtrate under reduced pressure, adding 100mL of water, extracting with ethyl acetate (30 mL. times.3), combining organic phases, drying with anhydrous sodium sulfate, concentrating, purifying with a preparative liquid phase (the preparative liquid phase is purified by the method B), and freeze-drying to obtain 22.9mg of the trifluoroacetate salt of the title compound.
ESI-MS(m/z):391.56[M+H]+
1H NMR(400MHz,CDCl3)9.34(d,J=66.3Hz,2H),8.66(t,J=6.8Hz,1H),8.06(d,J=7.8Hz,1H),7.63(d,J=8.0Hz,1H),7.52(t,J=6.2Hz,1H),7.18–6.94(m,4H),4.68(s,5H),3.87–3.66(m,2H),3.29(s,1H),3.05(d,J=15.9Hz,2H),2.87(s,3H),2.73–2.52(m,1H),2.20(d,J=14.8Hz,2H),2.02(d,J=13.8Hz,1H),1.93–1.74(m,3H),0.81–0.65(m,1H).
Example three: synthesis of 2- ((2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amine) -2, 3-dihydro-1H-inden-1-ol
Figure PCTCN2019082841-APPB-000038
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (75mg,0.289mmol) and 2-amino-2, 3-dihydro-1H-inden-1-ol (51.77mg,0.347mmol) were added to dichloromethane (15mL) under nitrogen protection, anhydrous magnesium sulfate (208.86mg,1.75mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (21.81mg,0.347mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Methanol (15mL) was added to the system to quench the reaction, filtered and the solvent was evaporated under reduced pressure to give the crude product. Purification by preparative liquid phase (preparative liquid phase purification using method B) followed by lyophilization afforded the trifluoroacetate salt of the title compound.
ESI-MS(m/z):393.53[M+H]+
1H NMR(400MHz,CDCl3)8.68(m,1H),8.11(m,1H),7.60(m,2H),7.37(d,J=7.2Hz,1H),7.27(m,2H),7.16(m,1H),5.16(m,1H),4.18(m,1H),3.88–3.54(m,3H),3.09(m,3H),2.67(m,1H),2.47–2.15(m,4H),2.02(d,J=13.9Hz,1H),1.85(d,J=11.4Hz,2H),1.67(m,1H),1.62-1.36(m,4H),1.15(s,3H).
Example four: synthesis of 5-fluoro-N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-indene-2-amine
Figure PCTCN2019082841-APPB-000039
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (75mg,0.289mmol) and 5-fluoro-2, 3-dihydro-1H-inden-2-amine (52.46mg,0.347mmol) were added to dichloromethane (15mL) under nitrogen, anhydrous magnesium sulfate (208.86mg,1.75mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (21.81mg,0.347mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Methanol (15mL) was added to the system to quench the reaction, filtered and the solvent was evaporated under reduced pressure to give the crude product. Purification by preparative liquid phase (preparative liquid phase purification using method B) followed by lyophilization afforded the trifluoroacetate salt of the title compound.
ESI-MS(m/z):395.52[M+H]+
1H NMR(400MHz,CDCl3)8.56(d,J=5.2Hz,1H),8.08(m,1H),7.60(d,J=8.0Hz,1H),7.52(m,1H),7.02(m,1H),6.88–6.69(m,2H),3.87–3.71(m,2H),3.64(m,1H),3.06(m,5H),2.51(m,1H),2.28(m,4H),1.97(d,J=14.0Hz,1H),1.89–1.69(m,2H),1.70–1.54(m,1H),1.56–1.31(m,4H),1.18(m,1H),1.09(m,1H),0.85–0.58(m,1H).
Example five: 2- ((2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amine) -2, 3-dihydro-1H-indene-5-carbonitrile
Figure PCTCN2019082841-APPB-000040
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (75mg,0.289mmol) and 2-amino-2, 3-dihydro-1H-indene-5-carbonitrile (54.9mg,0.347mmol) were added to dichloromethane (15ml) under nitrogen, anhydrous magnesium sulfate (208.86mg,1.75mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (21.81mg,0.347mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Methanol (15mL) was added to the system to quench the reaction, filtered and the solvent was evaporated under reduced pressure to give the crude product. Purification by preparative liquid phase (preparative liquid phase purification using method B) followed by lyophilization afforded the trifluoroacetate salt of the title compound.
ESI-MS(m/z):402.54[M+H]+
1H NMR(400MHz,CDCl3)8.69(s,1H),8.27–8.07(m,1H),7.76–7.57(m,2H),7.47(m,1H),7.41(d,J=3.9Hz,1H),7.27(m,1H),6.55(s,1H),3.95–3.77(m,2H),3.71(m,1H),3.21(m,4H),3.01(m,1H),2.55(d,J=12.0Hz,1H),2.37(m,3H),2.20(m,1H),2.02(d,J=13.5Hz,1H),1.95–1.75(m,2H),1.76–1.60(m,1H),1.60–1.36(m,4H),1.36–1.05(m,1H),0.73(m,1H).
Example six: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-inden-2-amine
Figure PCTCN2019082841-APPB-000041
The method comprises the following steps: synthesis of (R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetonitrile (1.00g,3.90mmol) was added to dry toluene (35mL), cooled to-78 ℃ under nitrogen and diisobutylaluminum hydride (0.83g,5.85mmol) was added dropwise. After the addition, the mixture was stirred at-78 ℃ for 2 hours, and after completion of the reaction was monitored by LCMS, a saturated aqueous ammonium chloride solution was added to the reaction mixture, and the mixture was warmed to room temperature and stirred for 1 hour. 6N hydrochloric acid was added to the reaction solution, and the mixture was stirred for 6 hours while the temperature was raised to 85 ℃. The reaction solution was cooled to room temperature, sodium bicarbonate was added thereto, PH was adjusted to 8, liquid separation was performed, the aqueous phase was concentrated to dryness, dichloromethane and anhydrous sodium sulfate were added thereto, stirring was performed for 30 minutes, filtration was performed, and concentration was performed to obtain 1.21g of a crude product. Pre-HPLC purification gave 0.30g of (R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde.
Step two: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-inden-2-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (88.0mg,0.34mmol) and 2-aminoindene hydrochloride (69.1mg,0.41mmol) were added to dichloromethane (15mL) under nitrogen, anhydrous magnesium sulfate (245.06mg,2.04mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (25.59mg,0.41mmol) and methanol (5mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. The reaction was quenched by addition of methanol (15mL), filtered, and the solvent was evaporated under reduced pressure to give 150mg of crude product. Purification by preparative liquid phase (preparative liquid phase purification using method B) followed by lyophilization afforded 12.6mg of the trifluoroacetate salt of the title compound.
ESI-MS(m/z):377.2[M+H]+
1H NMR(400MHz,DMSO-d6)8.62-8.58(m,1H),8.55(s,2H),7.84-7.80(m,1H),7.52(d,J=8.1Hz,1H),7.34-7.26(m,1H),7.26-7.14(m,4H),3.94-3.87(m,1H),3.72-3.54(m,2H),3.20-3.14(m,2H),2.94-2.82(m,3H),2.51-2.37(m,3H),2.35-2.23(m,2H),2.06-1.98(m,1H),1.89-1.77(m,2H),1.73-1.57(m,2H),1.56-1.42(m,2H),1.37-1.35(m,2H),1.04-0.95(m,1H),0.68-0.60(m,1H).
Example seven: synthesis of N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl) -5-morpholin-2, 3-dihydro-1H-inden-2-amine
Figure PCTCN2019082841-APPB-000042
The method comprises the following steps: synthesis of N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl) -5-morpholin-2, 3-dihydro-1H-inden-2-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (88.0mg,0.34mmol) and 5-morpholine-2, 3-dihydro-1H-inden-2-amine (89.0mg,0.41mmol) were added to dichloromethane (15mL) under nitrogen protection, anhydrous magnesium sulfate (490mg,4.07mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (107mg,1.70mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Adding methanol (15mL) into the system, performing extraction and quenching reaction, filtering, and evaporating the solvent under reduced pressure to obtain a crude product. Purification by preparative liquid phase (preparative liquid phase purification using method C) and lyophilisation gave 3.7mg of the title compound.
ESI-MS(m/z):462.2[M+H]+
1H NMR(400MHz,CD3OD)8.63(dd,J=4.9,1.7Hz,1H),7.99-7.94(m,1H),7.65(d,J=8.1Hz,1H),7.47–7.39(m,1H),7.20(dd,J=8.3,2.1Hz,1H),7.06–6.97(m,2H),3.91–3.84(m,4H),3.77(dd,J=7.7,2.8Hz,2H),3.25–3.19(m,4H),3.07–2.81(m,4H), 2.57–2.38(m,3H),2.19-2.10(m,1H),2.04–1.85(m,3H),1.82–1.69(m,2H),1.67–1.37(m,6H),1.35-1.25(m,2H),1.17-1.05(m,1H).
Example eight: synthesis of (S) -N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl-1, 2,3, 4-tetrahydronaphthalen-2-amine
Figure PCTCN2019082841-APPB-000043
The method comprises the following steps: synthesis of (S) -N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl-1, 2,3, 4-tetrahydronaphthalen-2-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (80.0mg,0.31mmol) and (S) -1,2,3, 4-tetrahydronaphthalen-2-amine (90.8mg,0.62mmol) were added to dichloromethane (15mL) under nitrogen protection, anhydrous magnesium sulfate (186mg,1.54mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (97.0mg,1.54mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Adding methanol (15mL) into the system, performing extraction and quenching reaction, filtering, and evaporating the solvent under reduced pressure to obtain a crude product. After purification of the preparative liquid phase (preparative liquid phase purification using method C), the title compound was lyophilized, dissolved in acetonitrile, added 1mol/L dilute hydrochloric acid and stirred for 30 minutes and lyophilized again to give 29.6mg of the hydrochloride salt of the title compound.
ESI-MS(m/z):391.2[M+H]+
1H NMR(400MHz,DMSO-d6)8.65–8.59(m,1H),7.94-7.90(m,1H),7.61(d,J=8.1Hz,1H),7.40-7.37(m,1H),7.18–7.03(m,4H),3.61(t,J=11.6Hz,2H),3.32-3.28(m,1H),3.04-2.91(m,2H),2.87–2.61(m,3H),2.48–2.18(m,4H),2.07-2.03(m,2H),1.95–1.80(m,2H),1.75–1.41(m,6H),1.40-1.30(m,2H),1.03-0.95(m,1H),0.68-0.60(m,1H).
Example nine: synthesis of (R) -N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl) chroman-3-amine
Figure PCTCN2019082841-APPB-000044
The method comprises the following steps: synthesis of (R) -N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl) chroman-3-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (60.0mg, 0.23mmol) and (R) -chroman-3-amine (43.0mg,0.23mmol) were added to dichloromethane (15mL) under nitrogen protection, anhydrous magnesium sulfate (139mg,1.16mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (93.1mg,1.16mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Adding methanol (15mL) into the system, performing extraction and quenching reaction, filtering, and evaporating the solvent under reduced pressure to obtain a crude product. Purification by preparative liquid phase (preparative liquid phase purification using method C), lyophilization afforded the title compound, which was dissolved in acetonitrile and stirred for 30 minutes with 1mol/L dilute hydrochloric acid and lyophilized again to afford 13.5mg of the hydrochloride salt of the title compound.
ESI-MS(m/z):393.2[M+H]+
1H NMR(400MHz,DMSO-d6)8.63(d,J=5.0Hz,1H),7.96(s,1H),7.62(d,J=8.2Hz,1H),7.42(s,1H),7.16–7.06(m,2H),6.92–6.89(m,1H),6.81(d,J=7.8Hz,1H),4.22–4.15(m,2H),3.58(t,J=11.1Hz,4H),3.17–3.04(m,1H),2.94–2.78(m,2H),2.44(d,J=13.9Hz,3H),2.16–2.06(m,1H),1.97(t,J=11.1Hz,1H),1.86(d,J=13.7Hz,1H),1.76–1.60(m,2H),1.57–1.31(m,5H),1.04–0.92(m,1H),0.73–0.64(m,1H).
Example ten: synthesis of (S) -N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl) chroman-3-amine
Figure PCTCN2019082841-APPB-000045
The method comprises the following steps: synthesis of (S) -N- (2- ((R) -9- (pyridin-2-yl-6-oxaspiro [4.5] decan-9-yl) ethyl) chroman-3-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (60.1mg,0.23mmol) and (S) -chroman-3-amine (43.0mg,0.23mmol) were added to dichloromethane (15mL) under nitrogen protection, anhydrous magnesium sulfate (139mg,1.16mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (93.2mg,1.16mmol) and methanol (3mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. Adding methanol (5mL) into the system, performing extraction and quenching reaction, filtering, and evaporating the solvent under reduced pressure to obtain a crude product. Purification by preparative liquid phase (preparative liquid phase purification using method C), lyophilization afforded the title compound which was dissolved in acetonitrile and stirred for 30 minutes with 1mol/L dilute hydrochloric acid and lyophilized again to afford 23.4mg of the hydrochloride salt of the title compound.
ESI-MS(m/z):393.2[M+H]+
1H NMR(400MHz,DMSO-d6)8.63(d,J=5.0Hz,1H),7.96(s,1H),7.62(d,J=8.2Hz,1H),7.42(s,1H),7.16–7.06(m,2H),6.92–6.89(m,1H),6.81(d,J=7.8Hz,1H),4.22–4.15(m,2H),3.58(t,J=11.1Hz,4H),3.17–3.04(m,1H),2.94–2.78(m,2H),2.44 (d,J=13.9Hz,3H),2.16–2.06(m,1H),1.97(t,J=11.1Hz,1H),1.86(d,J=13.7Hz,1H),1.76–1.60(m,2H),1.57–1.31(m,5H),1.04–0.92(m,1H),0.73–0.64(m,1H).
Example eleven: synthesis of (R) - (2- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amino) -2, 3-dihydro-1H-inden-2-yl) methanol
Figure PCTCN2019082841-APPB-000046
The method comprises the following steps: synthesis of (2-amino-2, 3-dihydro-1H-inden-2-yl) methanol
2-amino-2, 3-dihydro-1H-indene-2-carboxylic acid methyl ester hydrochloride (50.3mg,0.21mmol) was dissolved in THF (35mL) and LiAlH was added4(24.5mg, 0.64mmol), and reacted at 25 ℃ for 2 hours. After completion of the reaction was monitored by LCMS, the reaction mixture was quenched by addition of water and 15% NaOH solution, dried over anhydrous sodium sulfate, filtered and concentrated to give 30.5mg of crude (2-amino-2, 3-dihydro-1H-inden-2-yl) methanol.
ESI-MS(m/z):164.22[M+H]+
Step two: synthesis of (R) - (2- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amino) -2, 3-dihydro-1H-inden-2-yl) methanol
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (50.2mg,0.19mmol) and (2-amino-2, 3-dihydro-1H-inden-2-yl) methanol (39.7mg,0.19mmol) were added to MeOH (5mL) under nitrogen and stirred at room temperature for 0.5H. Sodium cyanoborohydride (123.6mg,1.93mmol) was added, and the reaction was continued at room temperature for 4 hours. Through LC-MS detection, the raw materials disappear and the target product is generated. Filtration, concentration of the filtrate to remove some of the solvent, and lyophilization of the residue after purification of the preparative liquid phase (preparative liquid phase purification using method C) gave 12.34mg of the title compound.
1H NMR(400MHz,CD3OD)8.48(d,J=4.0Hz,1H),7.73(t,J=8.0Hz,1H),7.42(d,J=8.0Hz,1H),7.21(dd,J=7.2,5.2Hz,1H),7.11–6.99(m,4H),3.78–3.66(m,2H),3.38(d,J=11.2Hz,1H),3.34(d,J=11.2Hz,1H),2.80(dd,J=16.4,10.8Hz,2H),2.68(dd,J=16.0,14.4Hz,2H),2.49–2.32(m,3H),1.98–1.84(m,3H),1.77–1.58(m,4H),1.57–1.36(m,4H),1.14–1.03(m,1H),0.70(dt,J=13.6,8.8Hz,1H).
Example twelve: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -6,7,8, 9-tetrahydro-5H-benzo [7] cyclopenten-7-amine
Figure PCTCN2019082841-APPB-000047
The method comprises the following steps: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -6,7,8, 9-tetrahydro-5H-benzo [7] cyclopenten-7-amine
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (44.5mg,0.17mmol) and 6,7,8, 9-tetrahydro-5H-benzo [7] cyclopenten-7-amine (35.2mg,0.17mmol) were added to MeOH (5mL) under nitrogen and stirred at room temperature for 0.5H. Sodium cyanoborohydride (39.5mg,1.72mmol) was added and the reaction was continued at room temperature for 4 hours. Through LC-MS detection, the raw materials disappear and the target product is generated. Filtration, concentration of the filtrate to remove some of the solvent, and lyophilization of the residue after purification of the preparative liquid phase (preparative liquid phase purification using method C) gave 11.2mg of the title compound.
ESI-MS(m/z):405.3[M+H]+
1H NMR(400MHz,CD3OD)8.62–8.53(m,1H),7.81(td,J=8.0,2.0Hz,1H),7.55(d,J=8.0Hz,1H),7.28(ddd,J=7.6,4.8,0.8Hz,1H),7.13–6.99(m,4H),3.85–3.72(m,2H),2.77–2.66(m,4H),2.65–2.51(m,3H),2.46(dd,J=14.0,2.0Hz,1H),2.12–1.88(m,5H),1.78–1.62(m,4H),1.61–1.39(m,4H),1.19–0.99(m,3H),0.75(dt,J=13.6,9.2Hz,1H).
Example thirteen: synthesis of (R) -2-methyl-N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-inden-2-amine
Figure PCTCN2019082841-APPB-000048
The method comprises the following steps: synthesis of (R) -2-methyl-N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-inden-2-amine
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (60.2mg,0.23mmol) and 2-methyl-2, 3-dihydro-1H-inden-2-amine (43.8mg,1.23mmol) were added to MeOH (5mL) under nitrogen and stirred at room temperature for 0.5H. Sodium cyanoborohydride (148.4mg,2.31mmol) was added and the reaction was continued at room temperature for 4 hours. Through LC-MS detection, the raw materials disappear and the target product is generated. Filtration, concentration of the filtrate to remove some of the solvent, and lyophilization of the residue after purification of the preparative liquid phase (preparative liquid phase purification using method C) gave 9.3mg of the title compound.
ESI-MS(m/z):391.3[M+H]+
1H NMR(400MHz,CD3OD)8.52–8.47(m,1H),7.75(td,J=8.0,2.0Hz,1H),7.47(d,J=8.0Hz,1H),7.23(ddd,J=7.6,4.8,0.8Hz,1H),7.10–7.03(m,4H),3.79–3.68(m,2H),2.79(dd,J=15.6,6.4Hz,2H),2.67(dd,J=15.6,11.6Hz,2H),2.52–2.38(m,3H),1.99–1.86(m,3H),1.77–1.61(m,4H),1.57–1.40(m,4H),1.13–1.08(m,1H),1.08(s,3H),0.72(dt,J=13.6,8.8Hz,1H).
Example fourteen: synthesis of 3- (2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amino) thiochroman 1, 1-dioxide
Figure PCTCN2019082841-APPB-000049
2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (30.0mg,0.12mmol) and 3-aminothiochroman 1, 1-dioxide (22.8mg,0.12mmol) were added to MeOH (5mL) under nitrogen and stirred at room temperature for 0.5 h. Sodium cyanoborohydride (74.2mg,1.16mmol) was added and the reaction was continued at room temperature for 4 hours. Through LC-MS detection, the raw materials disappear and the target product is generated. Filtration, concentration of the filtrate to remove some of the solvent, and lyophilization of the residue after purification by preparative liquid phase (preparative liquid phase purification using method C) gave 8.4mg of the title compound.
ESI-MS(m/z):441.2[M+H]+
1H NMR(400MHz,CD3OD)8.54(d,J=4.4Hz,1H),7.84–7.71(m,2H),7.58–7.48(m,2H),7.45(t,J=7.6Hz,1H),7.31(d,J=8.0Hz,1H),7.28–7.21(m,1H),3.83–3.69(m,2H),3.48(t,J=13.6Hz,1H),3.40–3.31(m,1H),3.19–3.04(m,2H),2.75(ddd,J=16.4,10.8,5.2Hz,1H),2.66–2.50(m,2H),2.43(d,J=13.2Hz,1H),2.19–2.08(m,1H),2.04–1.97(m,1H),1.91(d,J=13.3Hz,1H),1.78–1.67(m,3H),1.66–1.35(m,5H),1.15–1.01(m,1H),0.78–0.65(m,1H).
Example fifteen: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2-vinyl-2, 3-dihydro-1H-indene-2-amine
Figure PCTCN2019082841-APPB-000050
The method comprises the following steps: synthesis of 2-aminoindan-2-carboxylic acid hydrochloride
To N-tert-butoxycarbonyl-2-aminoindan-2-carboxylic acid (1.13g,4.07mmol) was added a 4M dioxane hydrochloride solution (10mL) and the mixture was reacted at room temperature for 2 hours, and after completion of the reaction was monitored by LC-MS, the mixture was concentrated to give the title compound as a white solid (0.85 g).
Step two: synthesis of 2-amino-2, 3-dihydro-1H-indene-2-methanol
2-aminoindan-2-carboxylic acid hydrochloride (0.85g, 3.98mmol) was dissolved in THF (50mL), lithium aluminum hydride (0.40mg,11.9mmol) was slowly added under ice bath conditions, and the reaction was slowly warmed to room temperature for 3 hours. After the completion of the reaction was monitored by LC-MS, the reaction was quenched with water (0.4mL), 15% aqueous NaOH (0.4mL), and water (1.2mL), and stirred for 30min with anhydrous sodium sulfate, and filtered through celite. The filtrate was concentrated and purified by silica gel column chromatography with DCM/MeOH ═ 10/1 to give the title compound 292mg as a pale yellow solid.
Step three: synthesis of tert-butyl-N- (2 (hydroxymethyl) indol-2-yl) carbamate
2-amino-2, 3-dihydro-1H-indene-2-methanol (0.15g,0.92mmol) was dissolved in DCM (10mL), and triethylamine (1.38mmol,0.19mL) and di-tert-butyl dicarbonate (1.01mmol,0.23mL) were added to react at 25 ℃ for 2 hours.
The reaction was monitored by LC-MS to endpoint and purified by DCM/MeOH 10/1pre-TLC to afford the title compound as a white solid, 207 mg.
Step four: synthesis of tert-butyl-N- (2-formylindol-2-yl) carbamate
Oxalyl chloride (0.15g,1.18mmol) was dissolved in DCM (5mL) and reacted at-78 deg.C under nitrogen protection with DMSO (0.12g,1.57mmol,0.11mL) in DCM (5mL) and at that temperature for 1 hour, tert-butyl-N- (2 (hydroxymethyl) indol-2-yl) carbamate (0.21g,0.79mmol) in DCM (5mL) was added dropwise and at that temperature for 1 hour, and finally triethylamine (0.32g,3.14mmol) was added and allowed to warm to room temperature for 2 hours. TLC monitored the reaction to endpoint. The solvent was spun dry and PE/EA 2/1pre-TLC afforded 136mg of the title compound as a colorless oily liquid.
Step five: synthesis of tert-butyl N- (2-vinyl indol-2-yl) carbamate
Methyltriphenylphosphonium iodide (556mg,1.38mmol) was dissolved in THF (10mL) to give a dark yellow solution, N-BuLi (2.5M in hexane) (1.38mmol,0.55mL) was added under ice-bath conditions, the solution was reacted at this temperature for 30 minutes to give a pale yellow solution, and a solution of tert-butyl-N- (2-formylindol-2-yl) carbamate (45mg,0.17mmol) in THF (5mL) was added dropwise. Slowly raising the temperature to room temperature for reaction for 2 hours. The reaction was detected by LC-MS to the end point. After dilution with water (20mL), liquid was separated, EA (20mL × 3) was extracted, dried over anhydrous sodium sulfate, filtered, concentrated, and PE/EA 5/1pre-TLC separated and purified to give the title compound as a colorless oily liquid, 34 mg.
Step six: synthesis of 2-vinyl indole-2-amine hydrochloride
To tert-butyl N- (2-vinylindol-2-yl) carbamate (30mg,0.12mmol) was added hydrochloric acid (4M dioxane solution, 12.0mmol, 3mL) and the mixture was reacted at room temperature for 1 hour. The reaction was monitored by LC-MS to endpoint. Concentration afforded the title compound as a pale yellow solid, 22 mg.
Step seven: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2-vinyl-2, 3-dihydro-1H-indene-2-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (32mg,0.12mmol) and 2-ethylindole-2-amine hydrochloride (22mg,0.11mmol) were dissolved in MeOH (5mL) under nitrogen, reacted at 50 ℃ for 1h, glacial acetic acid (8.74mmol,0.5mL) and sodium cyanoborohydride (25.9mg,1.12mmol) were added and the reaction was continued for 18 h. The reaction was terminated by LC-MS detection, filtered, concentrated, purified by preparative liquid phase (preparative liquid phase purification using method C), and lyophilized to give 18.4mg of the title compound.
ESI-MS(m/z):403.2[M+H]+
1H NMR(400MHz,CD3OD)8.55–8.46(m,1H),7.75(td,J=7.6,2.0Hz,1H),7.46(d,J=8.0Hz,1H),7.27-7.18(m,1H),7.13–7.01(m,4H),5.72(dd,J=17.2,10.8Hz,1H),4.94–4.91(m,2H),3.80–3.66(m,2H),3.00–2.74(m,4H),2.51–2.31(m,3H),2.01–1.83(m,3H),1.76–1.37(m,8H),1.13–1.04(m,1H),0.76–0.63(m,1H).
Example sixteen: synthesis of 5- ((2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amino) -4H-cyclopenta [ b ] thiophen-6 (5H) -one
Figure PCTCN2019082841-APPB-000051
The method comprises the following steps: (E) synthesis of (E) -5- (oximido) -4H-cyclopentano [ b ] thiophen-6 (5H) -one
4H-Cyclopenta [ b ] thiophen-6 (5H) -one (0.60g,4.34mmol) was added to methanol (7.5mL), and the mixture was stirred at 40 ℃ with warming. N-butyl nitrite (0.90g,8.68mmol) was added dropwise to the reaction system, and after completion of the addition, concentrated hydrochloric acid (1mL) was added dropwise. After further stirring at 40 ℃ for 1.5 hours, LC-MS shows that 50% of the raw material is not converted, the mixture is cooled to room temperature, and the mixture is concentrated, purified (the preparation liquid phase is purified by a method C) and lyophilized to obtain 0.38g of (E) -5- (hydroxyimino) -4H-cyclopenta [ b ] thiophene-6 (5H) -ketone.
Step two: synthesis of 5-amino-4H-cyclopenta [ b ] thiophen-6 (5H) -one
(E) -5- (hydroxyimino) -4H-cyclopenta [ b ] thiophen-6 (5H) -one (70mg,0.42mmol) was added to methanol (25mL) and HCl (0.5mL), palladium on carbon (8.8mg,0.07mmol) was added, and the mixture was stirred overnight with hydrogen substitution 3 times. LC-MS showed the raw material reaction was complete, and direct filtration and concentration gave 65 mg of hydrochloride of 5-amino-4H-cyclopenta [ B ] thiophen-6 (5H) -one. The crude product was used directly in the next step.
Step three: synthesis of 5- ((2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amino) -4H-cyclopenta [ b ] thiophen-6 (5H) -one
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (82.0mg, 0.32mmol) and hydrochloride salt of 5-amino-4H-cyclopenta [ b ] thiophen-6 (5H) -one (60.1mg,0.32mmol) were added to dichloromethane (25mL) under nitrogen, anhydrous magnesium sulfate (0.23g,1.90mmol) and sodium cyanoborohydride (23.9mg,0.38mmol) were added. Stirring for 18 hours, LC-MS tracing the reaction, product formation, incomplete conversion of the raw material, celite filtration through pad, methanol elution, reduced pressure spin-drying, preparative purification (preparative liquid phase purification using method C) lyophilization yielded 3.8mg of 5- ((2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) amino) -4H-cyclopenta [ b ] thiophen-6 (5H) -one.
ESI-MS(m/z):397.54[M+H]+
1H NMR(400MHz,CD3OD)8.54–8.50(m,1H),8.15–8.13(m,1H),7.79–7.74(m,1H),7.52–7.49(m,1H),7.25–7.22(m,1H),7.10(d,J=4.8Hz,1H),3.79–3.71(m,2H),3.26–3.18(m,1H),2.72–2.47(m,3H),2.43–2.39(m,1H),2.21–1.95(m,2H),1.92(d,J=13.8Hz,1H),1.80–1.36(m,10H),0.76–0.67(m,1H).
Example seventeen: synthesis of N- (2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-amine
Figure PCTCN2019082841-APPB-000052
The method comprises the following steps: synthesis of tert-butyl (6-oxo-5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-yl) carbamate
Di-tert-butyl dicarbonate (50mg,0.23mmol) and (E) -5- (hydroxyimino) -4H-cyclopenta [ B ] thiophen-6 (5H) -one (38.3mg,0.23mmol) were added to methanol (6mL), and after palladium on charcoal (27.8mg,0.23mmol) was added, hydrogen gas was substituted for 3 times and the mixture was stirred overnight. LC-MS showed the starting material reacted completely, filtered directly and concentrated to give 58mg of tert-butyl (6-oxo-5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-yl) carbamate. The crude product was used directly in the next step.
Step two: synthesis of tert-butyl (6-hydroxy-5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-yl) carbamate
Sodium borohydride (19.4mg,0.52mmol) was added to tetrahydrofuran (15mL), and tert-butyl (6-oxo-5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-yl) carbamate (65.2mg,0.26mmol) in methanol (5mL) was added dropwise to the reaction mixture, and stirred at room temperature overnight. LC-MS shows that the raw materials completely react, the reaction solution is concentrated to be dry, water and DCM are added for liquid separation, the water phase is extracted for 2 times by DCM, organic phases are combined, and the tert-butyl (6-hydroxy-5, 6-dihydro-4H-cyclopenta [ B ] thiophen-5-yl) carbamate is obtained by drying and concentration after 35.5mg of tert-butyl (6-hydroxy-5, 6-dihydro-4H-cyclopenta [ B ] thiophen-5-yl) carbamate is.
Step three: synthesis of 5, 6-dihydro-4H-cyclopenta [ b ] thiophene-5-amine
Tert-butyl (6-hydroxy-5, 6-dihydro-4H-cyclopenta [ b)]Thien-5-yl) carbamate (35.1mg,0.14mmol) was dissolved in dichloromethane (17mL) and trifluoroacetic acid (1.7mL,0.14mmol) and Et were added sequentially3SiH (1.7mL,0.14mmol), 0.5 h at 0 ℃. LC-MS shows that the reaction is complete, and the 5, 6-dihydro-4H-cyclopentano [ b ] is directly obtained by concentration]30.5mg of thiophene-5-amine. The crude product was used directly in the next step.
Step four: synthesis of N- (2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-amine
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (74.5mg,0.29mmol) and 5, 6-dihydro-4H-cyclopenta [ b ] thiophen-5-amine (40.2mg,0.29mmol) were added to dichloromethane (5mL) under nitrogen, anhydrous magnesium sulfate (207.5mg,1.72mmol) and sodium cyanoborohydride (21.7mg,0.34mmol) were added. Stirring for 18H, performing LC-MS tracking reaction to generate a product, incompletely converting the raw material, filtering with diatomite, eluting with methanol, performing reduced pressure spin-drying, and performing preparative purification (liquid phase purification is performed by method C) and freeze-drying to obtain 4.8mg of N- (2- ((R) -9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -5, 6-dihydro-4H-cyclopenta [ b ] thiophene-5-amine.
ESI-MS(m/z):383.2[M+H]+
1H NMR(400MHz,CD3OD)8.54–7.53(m,1H),7.80–7.76(m,1H),7.53–7.51(m,1H),7.26–7.23(m,1H),7.18–7.17(m,1H),6.73–6.72(m,1H),3.86–3.72(m,2H),3.06–3.00(m,1H),2.94–2.84(m,1H),2.61–2.48(m,3H),2.46–2.34(m,2H),2.08–1.95(m,2H),1.91(d,J=13.7Hz,1H),1.79–1.36(m,9H),1.14–1.05(m,1H),0.76–0.68(dt,J=13.6,8.8Hz,1H).
Example eighteen: synthesis of (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-inden-2-amine
Figure PCTCN2019082841-APPB-000053
The method comprises the following steps: (R) -N- (2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) ethyl) -2, 3-dihydro-1H-indene-2-amine (hydrochloride)
(R) -2- (9- (pyridin-2-yl) -6-oxaspiro [4.5] decan-9-yl) acetaldehyde (235.0mg,0.91mmol) and 2-aminoindene hydrochloride (153.7mg,0.91mmol) were added to dichloromethane (50mL) under nitrogen, anhydrous magnesium sulfate (654.4mg,5.44mmol) was added, and the mixture was stirred at room temperature for 2 hours. Sodium cyanoborohydride (68.3mg,1.09mmol) and methanol (10mL) were added, and the reaction was stirred at room temperature overnight. Through LC-MS detection, the raw materials disappear and the target product is generated. The reaction was quenched by adding methanol (15mL), filtered, and the solvent was evaporated under reduced pressure to give 377mg of crude product. The free compound (107 mg) was obtained by preparative liquid phase purification (preparative liquid phase purification using method C) followed by lyophilization. This compound was added to 35mL of dilute hydrochloric acid (1N), stirred for 30 minutes, and lyophilized again to give 106mg of the hydrochloride of the title compound.
ESI-MS(m/z):377.2[M+H]+
1H NMR(400MHz,DMSO-d6)9.35(s,1H),9.22(s,1H),8.78–8.58(m,1H),8.14(s,1H),7.78(s,1H),7.59(s,1H),7.29–7.11(m,4H),3.90–3.83(m,1H),3.73–3.68(m,1H),3.63–3.51(m,1H),3.18–3.12(m,2H),3.06–2.99(m,2H),2.85–2.83(m,1H),2.54–2.44(m,2H),2.35–2.31(m,1H),2.25–2.12(m,1H),2.06–2.02(m,1H),1.93(d,J=13.8Hz,1H),1.76–1.69(m,2H),1.63–1.30(m,5H),1.04–1.00(m,1H),0.73–0.66(m,1H).
Biological evaluation
Experimental example 1
1.1 purpose of the test
The purpose of this assay was to test compounds for agonism at the mu opioid receptor (mu OR) G protein signaling pathway.
1.2 principle of the test
Activation of the G protein signaling pathway of the μ OR can regulate intracellular cAMP levelsThe method of phase time resolved fluorescence (HTRF) measures changes in intracellular cAMP levels, which may reflect agonistic activity of the compound. Maximum Effect E on changes in cAMP levels according to the Compoundmax(maximum effect of 1. mu.M of the full agonist DAMGO (H-Tyr-D-Ala-Gly-N-MePhe-Gly-OH) is 100%) and the concentration of compound EC which causes half of the maximum effect50The compounds were evaluated for in vitro activity.
1.3 test methods
1.3.1 test materials and instruments
Figure PCTCN2019082841-APPB-000054
Figure PCTCN2019082841-APPB-000055
1.3.2 test procedure
Samples of test compounds at serial dilution concentrations were prepared on 384-well LDV plates and the sample dilution sequences were transferred to an experimental plate (Corning-3824) using an Echo machine, corresponding to 30nL per well, together with 30nL of the positive control damgo (hpe) and the negative control dmso (zpe) to the corresponding wells. Then 5. mu.L of stimulation buffer (STB) and 5. mu.L of hMOR/CHO cell suspension (10000 cells/well) were added to each well of the plate in sequence, with the final concentrations of IBMX and NKH477 compounds in the 10. mu.L system being 100. mu.M and 1.5. mu.M, respectively. The experimental plate is placed in a constant-temperature incubator at 37 ℃ for incubation for 40 min. cAMP levels were then detected according to the cAMP Dynamic 2 kit instructions.
1.4 test results
The level of agonism of the μ OR G protein signaling pathway by the test compounds is determined by the above assay, and the EC measured50And EmaxThe results are shown in Table 1 (100% maximal effect with 1. mu.M full agonist DAMGO).
TABLE 1 EC for test compounds agonizing the μ OR G protein signaling pathway affecting cAMP levels50And Emax
Examples EC 50(nM) E max(%)
Example one 1.0 83.4
Example two 7.2 68.3
EXAMPLE III 12.4 63.5
Example four 13.7 67.1
EXAMPLE six 0.1 61.7
EXAMPLE seven 97.2 100.9
Example eight 9.3 71.3
Example nine 8.5 81.1
Example ten 17.6 74.9
EXAMPLE eleven 57.5 79.4
Example twelve 15.3 73.6
EXAMPLE thirteen 21.4 63.4
Example fifteen 22.5 100.8
Example sixteen 23.0 70.3
Example seventeen 1.0 65.8
EXAMPLE eighteen 1.3 63.9
And (4) conclusion: the compounds of the application have strong agonistic activity on the mu opioid receptor (mu OR) G protein signaling pathway.
Experimental example 2
2.1 purpose of the test
The purpose of this assay is to test the activity of recruiting β arrestin2 following activation of μ opioid receptors (μ OR) by the compounds of the present application.
2.2 principle of the test
The activity of recruiting β arrestin2 after activation of μ opioid receptors (μ OR) by compounds was examined by enzyme fragment complementation method (EFC). The agonist, when bound to a mu opioid receptor (mu OR) overexpressing a coupled beta-gal fragment, recruits the beta-gal complementary fragment coupled to the beta-arrestin to form an intact catalytically active enzyme, which catalyzes the substrate to produce chemiluminescence. The maximum effect Emax (100% of the maximum effect with the 1 μ M full agonist DAMGO) on the level of β arrestin2 recruitment with the compound.
2.3 test methods
2.3.1 test materials and instruments
Figure PCTCN2019082841-APPB-000056
Figure PCTCN2019082841-APPB-000057
2.3.2 test procedure
Samples of test compounds at serial dilution concentrations were prepared on 384-well LDV plates, and the sample dilution sequences were transferred to an experimental plate (PerkinElmer) using an Echo machine, corresponding to 60nL per well, while transferring 60nL of the positive control damgo (hpe) and the negative control dmso (zpe) to the corresponding wells. 20 μ L U2OS/OPRM1 cell suspension (7500 cells/well) was then added to each well of the plate, centrifuged at 300rpm for 30s, and incubated at room temperature for 2 hours. Then 6. mu.L PathHunter detection reagent was added to each well of the plate as per PathHunter detection kit instructions and read on Envision after standing for 60 minutes at room temperature.
2.4 test results
Levels of recruitment of beta-arrestin 2 following activation of μ OR by the test Compounds were determined by the above assay, EmaxThe results are shown in Table 2 (100% maximal effect with 1. mu.M full agonist DAMGO).
TABLE 2 recruitment of beta-arrestin 2 levels of E following activation of μ OR by test Compoundsmax
Examples E max(%)
Example one <3
Example two <3
EXAMPLE six <3
Example eight <3
Example ten <3
Example fifteen <3
Example sixteen N/A
Example seventeen N/A
EXAMPLE eighteen <3
Note: "N/A" means no agonist activity.
And (4) conclusion: the level of recruitment of beta arrestin2 following activation of the mu opioid receptor (mu OR) by the compounds of the present application was shown to be weak.
Experimental example 3 Effect on respiratory System of conscious non-restraint C57 mice
3.1 purpose of the experiment
The compound of example eighteen was administered in a single subcutaneous injection to conscious unbound C57 mice in this experiment to evaluate its effect on the respiratory system of conscious unbound C57 mice.
3.2 Experimental methods
In this test, 4 groups of six animals each having half of a sex were administered with PBS (vehicle control group), eighteen examples (1.2 mg/kg), eighteen examples (2.4 mg/kg) and morphine hydrochloride injection (10 mg/kg). Respiratory system indicators (respiratory rate, RR; tidal volume, TV; minute ventilation, MV) were recorded and analyzed using a systemic plethysmography system at 0.25, 0.75, 2, and 4 hours before and after administration, respectively.
3.3 results of the experiment
And (3) displaying a respiratory index detection result: there was no abnormality at each time point after vehicle control administration compared to pre-administration, and under the present experimental conditions, eighteen examples given at doses of 1.2mg/kg and 2.4mg/kg by single subcutaneous injection had no inhibitory effect on the respiratory system of conscious non-restraint C57 mice.
In summary, the compounds of the present application have selective agonism at the mu opioid receptor (mu OR) G protein signaling pathway.
While specific embodiments of the invention have been described in detail, it will be appreciated by those skilled in the art that various modifications and alternatives to those details could be developed in light of the overall teachings of the disclosure, and that such modifications are intended to be included within the scope of the disclosure. The full scope of the invention is given by the appended claims and any equivalents thereof.

Claims (22)

  1. A compound shown in a formula (I), a solvate, a stereoisomer, a crystal form, a pharmaceutically acceptable salt or ester thereof, or any combination of the compound,
    Figure PCTCN2019082841-APPB-100001
    wherein R is1Selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or, R1Form a ring with ring A;
    R2selected from the group consisting of hydroxy, cyano, halogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, alkenyl, haloalkenyl, alkynyl, haloalkynyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
    R3Selected from the group consisting of hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; or, R3And R2Connecting to form a ring;
    Raselected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
    Rband RcIndependently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, and sulfonyl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, sulfonyl is optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
    w and U are independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-NRf-、-(CRdRe)m1-C(=O)-、-(CRdRe)m1-S(O)q-、-C(=O)-NRf-and-C (═ O) -;
    v is selected from C and N;
    Rd、Reand RfIndependently selected from hydrogen, hydroxy, halogen, alkyl, alkoxy, cycloalkyl, and heterocycloalkyl; or, RdAnd ReForm a ring with the attached carbon atom;
    m1selected from 1,2,3 and 4;
    ring a is selected from the group consisting of aromatic, heteroaromatic, aliphatic carbocycle, and lipoheterocycle;
    m and n are independently selected from 0, 1,2,3,4 and 5;
    x and y are independently selected from 1,2,3 and 4;
    q is selected from 0, 1 and 2.
  2. The compound of claim 1, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, having a structure according to formula (II),
    Figure PCTCN2019082841-APPB-100002
    wherein R is1Selected from hydroxy, cyano, halogen, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or, R1Form a ring with ring A;
    R2selected from the group consisting of hydroxy, cyano, halogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, alkenyl, haloalkenyl, alkynyl, haloalkynyl, aryl, heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
    R3Selected from the group consisting of hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; or, R3And R2Connecting to form a ring;
    Raselected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
    Rband RcIndependently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, and sulfonyl; wherein said alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, acyl, sulfonyl is optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, alkyl, and haloalkyl;
    w and U are independently selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-NRf-;
    Rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, alkyl and alkoxy; or, RdAnd ReForm a ring with the attached carbon atom;
    m1selected from 1,2,3 and 4;
    ring a is selected from the group consisting of aromatic, heteroaromatic, aliphatic carbocycle, and lipoheterocycle;
    m and n are independently selected from 0, 1,2,3,4 and 5;
    x and y are independently selected from 1,2,3 and 4.
  3. The compound of claim 1 or 2, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, having a structure represented by formula (III),
    Figure PCTCN2019082841-APPB-100003
    wherein the groups and substituents are as defined in claim 1 or 2.
  4. The compound of any one of claims 1-3, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, having a structure represented by formula (III-1) or formula (III-2),
    Figure PCTCN2019082841-APPB-100004
    wherein the groups and substituents are as defined in claim 1 or 2.
  5. The compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination of the foregoing of any one of claims 1-4, wherein,
    w is selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; m is1Is 1 or 2; x is selected from 1,2 and 3;
    preferably, W is selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; x is selected from 1,2 and 3;
    preferably, W is selected from the group consisting ofCRdRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; x is 1 or 2;
    preferably, W is selected from-CH2-and-CH2CH2-; x is 1 or 2.
  6. The compound of any one of claims 1-4, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein W is selected from- (CR)dRe)m1-O-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; x is 1,2 or 3;
    preferably, W is selected from- (CR)dRe)m1-O-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; x is 1,2 or 3;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100005
    Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; x is 1,2 or 3;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100006
    Wherein, the 1 position is connected with the 3 position in the general formula, and the 2 positionTo position 4 in the general formula; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; x is 1 or 2;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100007
    Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; x is 1 or 2.
  7. The compound of any one of claims 1-4, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein W is selected from- (CR)dRe)m1-NRf-、-(CRdRe)m1-C(=O)-、-(CRdRe)m1-S(O)q-, -C (═ O) -, and-C (═ O) -NRf-;Rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; q is 0, 1 or 2; x is 1,2 or 3;
    preferably, W is selected from- (CR)dRe)m1-NRf-、-(CRdRe)m1-C(=O)-、-(CRdRe)m1-S(O)q-, -C (═ O) -, and-C (═ O) -NRf-;Rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; or, RdAnd ReForm a 3-7 membered heterocyclic ring with the carbon atom to which it is attached; m is1Is 1 or 2; q is 0, 1 or 2; x is 1,2 or 3;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100008
    Figure PCTCN2019082841-APPB-100009
    Figure PCTCN2019082841-APPB-100010
    Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; q is 0, 1 or 2; x is 1,2 or 3;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100011
    Figure PCTCN2019082841-APPB-100012
    Figure PCTCN2019082841-APPB-100013
    Wherein, the position 1 is connected with the position 3 in the general formula, and the position 2 is connected with the position 4 in the general formula; rd、ReAnd RfIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; q is 0, 1 or 2; x is 1 or 2;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100014
    Figure PCTCN2019082841-APPB-100015
    Wherein the 1 position is connected with the 3 position in the general formula, and the 2 position is connected with the 4 position in the general formula;Rd、ReAnd RfIndependently selected from hydrogen, hydroxy, fluoro, chloro, methyl and ethyl; m is1Is 1 or 2; q is 0, 1 or 2; x is 1 or 2;
    Figure PCTCN2019082841-APPB-100016
    connecting at the position; x is 1 or 2;
    preferably, W is selected from
    Figure PCTCN2019082841-APPB-100017
    and-C (═ O) -, where the 1 position is attached to the 3 position in the formula and the 2 position is attached to the 4 position in the formula; x is 1.
  8. The compound of any one of claims 1-4, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein W is selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-S(O)q-and-C (═ O) -; rdAnd ReEach independently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; or, RdAnd ReTo the carbon atom to which it is attached to form a 3-7 membered heterocyclic ring; m is1Selected from 1 and 2; q is selected from 0, 1 and 2; x is selected from 1,2 and 3;
    preferably, W is selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-SO2-and-C (═ O) -; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl radical, C1-4Alkoxy radical, C3-6Cycloalkyl and 3-6 membered heterocycloalkyl; m is1Is 1 or 2; x is selected from 1,2 and 3;
    preferably, W is selected from- (CR)dRe)m1-、-(CRdRe)m1-O-、-(CRdRe)m1-SO2-and-C (═ O) -; rdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; x is 1 or 2;
    preferably, W is selected from-CH2-、-CH2CH2-、-CH2-O-、-CH2-SO2-and-C (═ O) -; x is 1.
  9. The compound of any one of claims 1-8, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein U is selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C3-8Cycloalkyl and 4-8 membered heterocycloalkyl; m is1Is 1 or 2; y is selected from 1,2 and 3;
    preferably, U is selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-6Alkyl and C1-6An alkoxy group; m is1Is 1 or 2; y is selected from 1,2 and 3;
    preferably, U is selected from- (CR)dRe)m1-;RdAnd ReIndependently selected from hydrogen, hydroxy, halogen, C1-4Alkyl and C1-4An alkoxy group; m is1Is 1 or 2; y is 1 or 2;
    preferably, U is selected from-CH2-and-CH2CH2-; y is 1 or 2.
  10. The compound of any one of claims 1-9, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein R is1Selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl、C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or R1Form a fused ring with ring a, said fused ring being a ring a and 5-6 membered lipoheterocyclic ring or a ring a and 5-6 membered heteroaromatic ring;
    Raselected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
    Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
    m is selected from 0, 1,2,3,4 and 5;
    preferably, R1Selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl radical, C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc(ii) a Or R1Form a fused ring with ring a, said fused ring being a ring a and 5-6 membered lipoheterocyclic ring or a ring a and 5-6 membered heteroaromatic ring;
    Raselected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10A membered heteroaryl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
    Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
    m is selected from 0, 1,2,3,4 and 5;
    preferably, R1Selected from cyano, halogen and 3-6 membered heterocycloalkyl;
    m is 0, 1 or 2;
    preferably, R1Selected from cyano, halogen and 5-6 membered heterocycloalkyl;
    m is 0, 1 or 2;
    preferably, R1Selected from cyano, fluoro and morpholinyl;
    m is 0 or 1.
  11. The compound of any one of claims 1-10, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein R is2Selected from hydroxy, cyano, halogen, C1-6Alkyl, halo C1-6Alkyl radical, C1-6Hydroxyalkyl, -C1-4alkyl-O-C1-4Alkyl radical, C3-8Cycloalkyl, 4-8 membered heterocycloalkyl, C2-6Alkenyl, halo C2-6Alkenyl radical, C2-6Alkynyl, halo C2-6Alkynyl, C6-10Aryl, 5-10 membered heteroaryl, -ORa、-NRbRc、-SRa、-CO2Raand-C (O) NRbRc
    RaSelected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
    Rband RcIndependently selected from hydrogen, C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl radical, C1-6An alkylsulfonyl group; wherein, said C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl, 5-10 membered heteroaryl, C1-6Alkanoyl and C1-6Alkylsulfonyl is optionally substituted with one or more substituents selected from: halogen, hydroxy, amino, cyano, carboxy, C1-6Alkyl and halo C1-6An alkyl group;
    n is selected from 0, 1,2,3,4 and 5;
    preferably, R2Selected from hydroxy, C1-6Alkyl radical, C1-6Hydroxyalkyl and C2-6An alkenyl group;
    n is 0, 1 or 2;
    preferably, R2Selected from hydroxy, C1-4Alkyl radical, C1-4Hydroxyalkyl and C2-4An alkenyl group;
    n is 0, 1 or 2;
    preferably, R2Selected from the group consisting of hydroxy, methyl, hydroxymethyl, and vinyl;
    n is 0 or 1.
  12. The compound of any one of claims 1-11, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein ring A is selected from C6-10Aromatic ring, 5-10 membered heteroaromatic ring, C3-8Aliphatic carbocycles and 3-8 membered aliphatic heterocycles;
    preferably, ring A is selected from C6-10Aromatic rings and 5-10 membered heteroaromatic rings;
    preferably, ring A is selected from a phenyl ring and a 5-6 membered heteroaromatic ring;
    preferably, ring a is selected from the group consisting of a benzene ring, a pyrrole ring, a furan ring, a thiophene ring, an oxazole ring, an imidazole ring, a thiazole ring, a pyridine ring, a pyrimidine ring, a pyrazine ring, a pyridazine ring, a 1,2, 4-1H-triazole ring and a pyrazole ring;
    preferably, ring a is selected from the group consisting of a benzene ring, a pyrrole ring, a furan ring, a thiophene ring, an oxazole ring, an imidazole ring, a thiazole ring, a pyridine ring, a pyrimidine ring, a pyrazine ring and a pyridazine ring.
  13. The compound of any one of claims 1-12, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof,
    wherein R is3Selected from hydrogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl radical, C3-8Cycloalkyl, 3-8 membered heterocycloalkyl, C6-10Aryl and 5-10 membered heteroaryl;
    preferably, R3Selected from hydrogen, hydroxy, C1-6Alkyl and halo C1-6An alkyl group;
    preferably, R3Selected from hydrogen and C1-4An alkyl group;
    preferably, R3Selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, and tert-butyl;
    preferably, R3Selected from hydrogen, methyl, ethyl, n-propyl and isopropyl;
    preferably, R3Is hydrogen.
  14. The compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, according to any one of claims 1 to 13, wherein the compound is selected from:
    Figure PCTCN2019082841-APPB-100018
    Figure PCTCN2019082841-APPB-100019
    Figure PCTCN2019082841-APPB-100020
    preferably, said compound is selected from:
    Figure PCTCN2019082841-APPB-100021
    Figure PCTCN2019082841-APPB-100022
    Figure PCTCN2019082841-APPB-100023
    preferably, said compound is selected from:
    Figure PCTCN2019082841-APPB-100024
    Figure PCTCN2019082841-APPB-100025
    Figure PCTCN2019082841-APPB-100026
    Figure PCTCN2019082841-APPB-100027
  15. a pharmaceutical composition comprising a compound of any one of claims 1-14, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination of the foregoing; optionally, it further contains one or more pharmaceutical excipients.
  16. Use of a compound of any one of claims 1-14, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition of claim 15, for the manufacture of a medicament for activating the activity of an opioid receptor (e.g., a μ -opioid receptor).
  17. Use of a compound of any one of claims 1-14, a solvate, stereoisomer, crystal form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition of claim 15, for the manufacture of an analgesic medicament.
  18. The compound of any one of claims 1-14, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or the pharmaceutical composition of claim 15 for use in activating opioid receptor (e.g., μ -opioid receptor) activity.
  19. A compound, solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof according to any one of claims 1 to 14 or any combination of the foregoing, or a pharmaceutical composition according to claim 15 for use in analgesia.
  20. A method of activating the activity of an opioid receptor (e.g., a μ -opioid receptor) in a subject, comprising the step of administering to the subject an effective amount of a compound of any one of claims 1-14, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or a pharmaceutical composition of claim 15.
  21. A method of analgesia comprising the step of administering to a subject in need thereof an effective amount of a compound of any one of claims 1-14, a solvate, stereoisomer, crystalline form, pharmaceutically acceptable salt or ester thereof, or any combination thereof, or the pharmaceutical composition of claim 15.
  22. A process for the preparation of a compound according to claim 1 or 2, comprising the steps of:
    Figure PCTCN2019082841-APPB-100028
    wherein R is1、R2、R3W, U, V, A, m, n, x, y are as described in claim 1;
    carrying out reductive amination reaction on a compound shown in a formula I-A and a compound shown in a formula I-B to prepare a compound shown in a formula I;
    preferably, the first and second electrodes are formed of a metal,
    Figure PCTCN2019082841-APPB-100029
    wherein R is1、R2、R3W, U, A, m, n, x, y are as described in claim 2;
    carrying out reductive amination reaction on the compound of the formula I-A and the compound of the formula II-B to prepare a compound of the formula II;
    preferably, the reductive amination reaction is carried out with addition of an acid and/or a reducing agent; preferably, the acid is selected from AcOH and TFA; preferably, the reducing agent is selected from NaBH4、NaCNBH3And NaBH (OAc)3
CN201980016139.6A 2018-04-28 2019-04-16 Oxaspiro compounds, preparation method and application thereof Pending CN111836807A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN201810402312 2018-04-28
CN2018104023123 2018-04-28
CN2018113589173 2018-11-15
CN201811358917 2018-11-15
PCT/CN2019/082841 WO2019205983A1 (en) 2018-04-28 2019-04-16 Oxa-spiro compound and preparation method therefor and uses thereof

Publications (1)

Publication Number Publication Date
CN111836807A true CN111836807A (en) 2020-10-27

Family

ID=68293799

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201980016139.6A Pending CN111836807A (en) 2018-04-28 2019-04-16 Oxaspiro compounds, preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN111836807A (en)
WO (1) WO2019205983A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112334465A (en) * 2019-01-17 2021-02-05 上海海雁医药科技有限公司 Tricyclic substituted oxaspiro derivatives, preparation method and medical application thereof
CN111662284B (en) * 2019-03-06 2021-08-10 上海海雁医药科技有限公司 Biheterocyclic substituted oxaspiro derivative, preparation method and medical application thereof
AU2021207143B2 (en) * 2020-01-17 2023-12-14 Shanghai Haiyan Pharmaceutical Technology Co., Ltd. Optically pure oxaspiro-substituted pyrrolopyrazole derivative, preparation method therefor and pharmaceutical use thereof
CA3164577A1 (en) * 2020-01-17 2021-07-22 Bin Hu Azabicyclic substituted oxaspiro derivative, preparation method therefor and medical use thereof
CN113214264B (en) * 2020-01-21 2022-08-09 上海海雁医药科技有限公司 Dihydropyrrolo five-membered heteroaryl substituted oxaspiro derivatives, preparation method and medical application thereof
CN112694485B (en) * 2020-09-10 2023-04-07 四川海品信医药科技有限公司 Opioid receptor agonist thiophene compound and preparation method thereof
JP2024501554A (en) * 2020-12-29 2024-01-12 上海海雁医薬科技有限公司 Oxyspiro ring-substituted pyrrolopyrazole derivatives and intermediates thereof and methods for their preparation
CA3227380A1 (en) * 2021-08-02 2023-02-09 Jiaquan Feng Oxaspiro derivative, and preparation method therefor and use thereof
CN117263956A (en) * 2022-01-19 2023-12-22 天地恒一制药股份有限公司 Opioid receptor agonist, preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702561A (en) * 2011-03-23 2014-04-02 特维娜有限公司 Opioid receptor ligands and methods of using and making same
WO2017063509A1 (en) * 2015-10-15 2017-04-20 江苏恒瑞医药股份有限公司 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018188641A1 (en) * 2017-04-14 2018-10-18 江苏恒瑞医药股份有限公司 Pharmaceutical composition containing mor agonist and kor agonist, and uses 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
CN108727347B (en) * 2017-04-14 2020-06-16 江苏恒瑞医药股份有限公司 Crystal form of opioid receptor (MOR) agonist and preparation method thereof
MX2020003784A (en) * 2017-09-28 2020-08-03 Jiangsu Hengrui Medicine Co Method for preparing oxaspirocycle derivative, and intermediate thereof.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702561A (en) * 2011-03-23 2014-04-02 特维娜有限公司 Opioid receptor ligands and methods of using and making same
WO2017063509A1 (en) * 2015-10-15 2017-04-20 江苏恒瑞医药股份有限公司 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEN,XIAOTAO等: "Structure−Activity Relationships and Discovery of a G Protein Biasedμ 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" *
LI,XIN等: "Discovery of SHR9352: A Highly Potent G Protein-Biased μ‑Opioid Receptor Agonist" *

Also Published As

Publication number Publication date
WO2019205983A1 (en) 2019-10-31

Similar Documents

Publication Publication Date Title
CN111836807A (en) Oxaspiro compounds, preparation method and application thereof
TWI753946B (en) Pyrazolopyridine derivatives with GLP-1 receptor agonist
JP6622824B2 (en) Kynurenin-3-monooxygenase inhibitor, pharmaceutical composition thereof, and methods of use thereof
TWI374141B (en) Spiro-oxindole compounds and their uses as therapeutic agents
AU2009331179B2 (en) Novel bicyclic heterocyclic compound
JP5658664B2 (en) 1,2-disubstituted heterocyclic compounds
KR20170082577A (en) Substituted pyrazolo(1,5-a)pyrimidines and their use in the treatment of medical disorders
RU2712163C2 (en) Activator of kcnq2-5 channels
WO2007032466A1 (en) Heterocyclic compound, and production process and use thereof
TW200838539A (en) Pyridopyrimidinone compounds useful in treating sodium channel-mediated diseases or conditions
CN107074833B (en) Having beta2Benzo ring derivatives with receptor agonistic and M3 receptor antagonistic activities and application thereof in medicine
JP2020506878A (en) Activator of TREK (TWIK related K channel) channel
KR20220141331A (en) P2X3 modifier
TW202315863A (en) Prodrugs and derivatives of psilocin and uses thereof
AU2012367780A1 (en) Derivatives of aza adamantane and uses thereof
JP2024501641A (en) Substituted Macrocycles and Related Treatment Methods
MX2007016217A (en) Alpha-(aryl-or heteroaryl-methyl)-beta piperidino propanamide compounds as orl1-receptor antagonists.
WO2007057775A1 (en) Spiropiperidine derivatives
TW479058B (en) 2,7-substituted octahydro-pyrrolo[1,2-a]pyrazine derivatives
WO2019179515A1 (en) Receptor inhibitor, pharmaceutical composition comprising same, and use thereof
DK2546255T3 (en) benzazepine
JP2012102018A (en) Amide compound
WO2021065893A1 (en) Tetralin derivative or pharmaceutically acceptable acid addition salt thereof
TW201708226A (en) Indole derivatives
CN111825595A (en) Sodium channel blockers

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20201027

WD01 Invention patent application deemed withdrawn after publication