[go: up one dir, main page]

HK1228391A1 - A compound as hepatitis c inhibitor and its use in medicament - Google Patents

A compound as hepatitis c inhibitor and its use in medicament Download PDF

Info

Publication number
HK1228391A1
HK1228391A1 HK17101942.8A HK17101942A HK1228391A1 HK 1228391 A1 HK1228391 A1 HK 1228391A1 HK 17101942 A HK17101942 A HK 17101942A HK 1228391 A1 HK1228391 A1 HK 1228391A1
Authority
HK
Hong Kong
Prior art keywords
compound
hcv
reaction
added
radical
Prior art date
Application number
HK17101942.8A
Other languages
Chinese (zh)
Other versions
HK1228391B (en
Inventor
张英俊
罗慧超
任青云
熊志敏
刘洋
雷义波
熊金峰
张健存
Original Assignee
广东东阳光药业股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 广东东阳光药业股份有限公司 filed Critical 广东东阳光药业股份有限公司
Publication of HK1228391A1 publication Critical patent/HK1228391A1/en
Publication of HK1228391B publication Critical patent/HK1228391B/en

Links

Abstract

The present invention relates to a compound which can act as an inhibitor of hepatitis C and its use in medicament. In Special, there are provided a compound represented by the formula (I), and stereoisomer, tautomer, enantiomer, oxynitride, hydrate, solvate, metabolite and pharmaceutically acceptable salt or prodrug of the compound represented by the formula (I), which are used to treat hepatitis C virus (HCV) infection or hepatitis C disease. The present invention also discloses a pharmaceutical composition containing such a compound and a method of treating an HCV infection or a hepatitis C disease using the compound of the present invention or a pharmaceutical composition thereof.

Description

Compound as hepatitis C inhibitor and application thereof in medicine
Technical Field
The present invention is in the field of medicine and relates to compounds useful for treating Hepatitis C Virus (HCV) infection, compositions of the compounds, and uses and methods of use thereof. In particular, the compounds of the present invention are useful for inhibiting the NS3/4A protease. More particularly, the present invention relates to compounds that can inhibit the function of the NS3/4A protein encoded by the hepatitis c virus, pharmaceutical compositions of said compounds and methods for inhibiting the function of the NS3/4A protein.
Background
HCV is the major human pathogen, estimated to infect approximately 1.7 million people worldwide, 5 times as many as people infected with human immunodeficiency virus type 1. Most of these HCV-infected individuals develop severe progressive liver disease, including cirrhosis and hepatocellular carcinoma. Thus, chronic HCV infection will be a leading cause of premature death of patients from liver disease worldwide.
Currently, the most effective HCV therapy is the use of a combination of interferon-alpha and ribavirin, which produces sustained efficacy in 40% of patients. Recent clinical results indicate that pegylated interferon-alfa is superior to unmodified interferon-alfa as a monotherapy. However, even with experimental treatment regimens involving combinations of pegylated interferon-alfa and ribavirin, most patients are unable to continue to reduce viral load, and many patients are often associated with side effects that do not allow for long-term treatment. Thus, new effective methods for treating HCV infection are currently urgently needed.
HCV is a positive-stranded RNA virus. Based on a comparison of the broad similarity of the deduced amino acid sequence and the 5' untranslated region, HCV was classified into a single genus of the Flaviviridae family (Flaviviridae family). All members of the flaviviridae family are enveloped virions containing a positive-stranded RNA genome that encodes all known virus-specific proteins via translation of a single uninterrupted Open Reading Frame (ORF).
Considerable heterogeneity exists within the nucleotide and encoded amino acid sequences of the entire HCV genome. At least 7 major genotypes have been identified and over 50 subtypes have been disclosed. In HCV-infected cells, viral RNA is translated into polyproteins and split into 10 individual proteins. At the amino terminus is the structural protein, immediately following E1 and E2. In addition, there are 6 nonstructural proteins, namely NS2, NS3, NS4A, NS4B, NS5A and NS5B, which play a very important role in the HCV life cycle (see, e.g., Lindenbach, b.d. and c.m. rice, nature.436,933-938,2005).
The major genotypes of HCV vary in their distribution throughout the world, and despite the large number of genotypes studied for pathogenesis and therapeutic role, the clinical importance of the genetic heterogeneity of HCV remains unclear.
The single-stranded HCV RNA genome is approximately 9500 nucleotides in length, has a single open reading frame, and encodes a single large polyprotein of about 3000 amino acids. In infected cells, the polyprotein is cleaved at multiple sites by cellular and viral proteases, producing structural and non-structural (NS) proteins. In the case of HCV, the formation of mature nonstructural proteins (NS2, NS3, NS4A, NS4B, NS5A and NS5B) is achieved by two viral proteases. The first is generally considered to be a metalloprotease, which cleaves at the NS2-NS3 junction; the second is a serine protease contained in the N-terminal region of NS3 (also referred to herein as NS3 protease) which mediates all subsequent cleavage downstream of NS3 in cis at the NS3-NS4A cleavage site and in trans at the remaining NS4A-NS4B, NS4B-NS5A, NS5A-NA5B sites. The NS4A protein appears to have multiple functions, acting as a cofactor for NS3 protease and possibly assisting in membrane localization of NS3 and other viral replicase components. The formation of the complex of NS3 protein with NS4A appears to be a processing event necessary to increase proteolytic efficiency at all sites. The NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities. NS5B (also referred to herein as HCV polymerase) is an RNA-dependent RNA polymerase involved in HCV replication.
Disclosure of Invention
The compounds of the present invention are useful for treating HCV infection in a patient, and selectively inhibit the replication of the HCV virus.
The invention relates to a novel macrocyclic compound, the inhibitory activity of which to HCV replicon is obviously superior to that of TMC-435(Simeprevir) drugs on the market, the compound has better inhibitory effect to various HCV genotypes (such as genotype 1a, genotype 1b, genotype 2a, genotype 3a, genotype 4a and genotype 5a), and especially has better inhibitory activity to genotype 2 a. The compound or the pharmaceutical composition has good inhibition effect on HCV infection, in particular to HCV NS3/4A protein.
In one aspect, the invention relates to a compound that is a compound of formula (I) or a stereoisomer, a tautomer, an enantiomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug of a compound of formula (I),
wherein:
R1is C6-10Aryl or C1-9A heteroaryl group;
R2and R3Each independently is H, F, Cl, Br, I, amino, hydroxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl or C1-9A heteroaryl group;
R4is H, deuterium or C1-6An alkyl group;
said C is1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl radical, C1-9Heteroaryl or amino is independently optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C1-6Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl or C1-9Heteroaryl group is substituted.
In some embodiments, the compound has the structure shown in formula (II):
or stereoisomers, tautomers, enantiomers, nitrogen oxides, hydrates, solvates, metabolites and pharmaceutically acceptable salts or prodrugs thereof.
In some embodiments, R in formula (I) or formula (II)1Is phenyl or heteroaryl of 5 to 6 ring atoms; said phenyl or heteroaryl of 5-6 ring atoms is optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3Alkenyl radical, C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy, C1-3Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclyl or C6-10Aryl group.
In still other embodiments, R in formula (I) or formula (II)1Is phenyl, furyl, thienyl, thiazolyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, oxadiazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyridyl, quinolyl, indolyl or acridinyl, wherein R is1The group may be optionally substituted with 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, trifluoromethyl, difluoroethyl, trifluoromethoxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, vinyl, ethynyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl or phenyl.
In some embodiments, R in formula (I) or formula (II)2And R3Each independentlyIs H, F, Cl, Br, I, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, trifluoromethyl, trifluoromethoxy, tert-butyl, ethenyl, propenyl, ethynyl, propynyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl, phenyl, methylamino or ethylamino.
In some embodiments, R4 in formula (I) or formula (II) is H, deuterium, methyl, deuterated methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, or tert-butyl.
In some embodiments, R in formula (I) or formula (II)1Is heteroaryl of 5 to 6 ring atoms; wherein said heteroaryl of 5 to 6 ring atoms is optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3Alkenyl radical, C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy or C1-3Substituted by alkylamino substituents; r in formula (I) or formula (II)2Is C1-6An alkyl group; r in formula (I) or formula (II)3Is C1-3An alkyl group; r in formula (I) or formula (II)4Is H or C1-3An alkyl group.
In another aspect, the present invention provides a pharmaceutical composition comprising any one of the compounds described above.
In some embodiments, the pharmaceutical composition may further comprise a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle, or combination thereof.
In some embodiments, it further comprises an additional anti-HCV agent, wherein the anti-HCV agent is an interferon, ribavirin, interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, an interfering RNA, an antisense RNA, imiqimod, an inosine 5' -monophospate dehydrogenase inhibitor, amantadine, rimantadine, bavacizumab, CivacirTMBeaprevir, telaprevir, erlotinib, daclatasvir, siThe interferon of meprevir, asunaprevir, vaniprevir, faldaprevir, paritaprevir, danoprrevir, sovaprevir, gradoprrevir, vedroprevir, BZF-961, GS-9256, narloprevir, ANA975, ombitasvir, EDP239, PPI-668, velpatasvir, samatasvir, elbasvir, MK-8325, GSK-2336805, PPI-461, BI-2013335, cilaprevir, ACH-1098655, VX-985, IDX-375, VX-500, VX-176813, PHX-2056, PHX-2054, IDX-136, IDX-316, modithcycline, VBY-376, TMC-649128, mersibiravir, PSI-61184, PSI-4024, IDX-647-136, PSI-102, PSI-369, BCI-369, BCB-344695, BCI-369, BCI-V-369, BCI-9, BCI-III-9, BCI-III, VITAB-III, BCI-III, VITAI-III, VITAB-III, VITAI-III, VITAB-III, VITAI-III, VITAI-.
In some embodiments, it further comprises at least one HCV inhibitor.
In yet other embodiments, wherein the HCV inhibitor is used to inhibit the HCV replication process and/or inhibit the function of HCV viral proteins; the HCV replication process comprises HCV entry, HCV uncoating, HCV translation, HCV replication, HCV assembly, or HCV release; the HCV viral protein is selected from metalloprotease, NS2, NS3, NS4A, NS4B, NS5A or NS5B, and an Internal Ribosome Entry Site (IRES) and inosine monophosphate dehydrogenase (IMPDH) required for HCV viral replication.
In another aspect, the compounds or pharmaceutical compositions of the present invention are useful in medicaments for inhibiting the HCV replication process and/or inhibiting the function of HCV viral proteins; the HCV replication process comprises HCV entry, HCV uncoating, HCV translation, HCV replication, HCV assembly, or HCV release; the HCV viral protein is selected from metalloprotease, NS2, NS3, NS4A, NS4B, NS5A or NS5B, and an Internal Ribosome Entry Site (IRES) and inosine monophosphate dehydrogenase (IMPDH) required for HCV viral replication.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition of the invention for the manufacture of a medicament for the prevention, treatment or amelioration of a hepatitis c disease in a patient, comprising administering to the patient an effective amount of a compound according to the invention or a pharmaceutical composition according to the invention.
Another aspect of the invention relates to methods for the preparation, isolation and purification of the compounds encompassed by formula (I) and formula (II).
The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting. These and other aspects will be more fully described below.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be found in the descriptions of "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and Jerry March, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "subject" as used herein refers to an animal. Typically the animal is a mammal. Subjects, e.g., also primates (e.g., humans, males or females), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, etc. In certain embodiments, the subject is a primate. In other embodiments, the subject is a human.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"chiral" is a molecule having the property of not overlapping its mirror image; and "achiral" refers to a molecule that can overlap with its mirror image.
"enantiomer" refers to two isomers of a compound that are not overlapping but are in mirror image relationship to each other.
"diastereomer" refers to a stereoisomer having two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow the general definitions of S.P. Parker, Ed., McGraw-Hilldictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; andEliel, E.and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
Many organic compounds exist in an optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of a molecule with respect to one or more of its chiral centers. The prefixes d and l or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or l indicates that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. A particular stereoisomer is an enantiomer and a mixture of such isomers is referred to as an enantiomeric mixture. A50: 50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process.
Any asymmetric atom (e.g., carbon, etc.) of a compound disclosed herein can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Depending on the choice of starting materials and methods, the compounds of the invention may exist as one of the possible isomers or as mixtures thereof, for example as racemates and diastereomeric mixtures (depending on the number of asymmetric carbon atoms). Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituents may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have cis or trans configuration.
Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
The racemates of any of the resulting end products or intermediates can be resolved into the optical enantiomers by known methods using methods familiar to those skilled in the art, e.g., by separation of the diastereomeric salts obtained. The racemic product can also be separated by chiral chromatography, e.g., High Performance Liquid Chromatography (HPLC) using a chiral adsorbent. In particular, Enantiomers may be prepared by asymmetric synthesis, for example, see Jacques, et al, Enantiomers, racemesand alcoholutions(Wiley Interscience,New York,1981);Principles of AsymmetricSynthesis(2ndEd.Robert E.Gawley,Jeffrey Aubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);Wilen,S.H.Tablesof Resolving Agents and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of NotreDame Press,Notre Dame,IN 1972);Chiral Separation Techniques:A PracticalApproach(Subramanian,G.Ed.,Wiley-VCH Verlag GmbH&Co.KGaA,Weinheim,Germany,2007)。
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (lowenergy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers (valenctautomers) include interconversion by recombination of some of the bonding electrons. A specific example of keto-enol tautomerism is the tautomerism of the pentan-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the tautomerism of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention. It is understood that the term "optionally substituted" may be used interchangeably with the term "substituted or unsubstituted". In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a particular substituent. Unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently. When substituents are described as "independently selected" groups, each substituent is selected independently of the other, and thus each substituent may be the same or different from the other.
In addition, unless otherwise explicitly indicated, the descriptions of the terms "… independently" and "… independently" and "… independently" used in the present invention are interchangeable and should be understood in a broad sense to mean that the specific items expressed between the same symbols do not affect each other in different groups or that the specific items expressed between the same symbols in the same groups do not affect each other.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated, straight or branched chain monovalent hydrocarbon radical containing from 1 to 20 carbon atoms, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 3 carbon atoms.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH)3) Ethyl (Et-CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) T-butyl (t-Bu, -C (CH)3)3) N-pentyl (-CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH(CH3)2) 3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) N-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2) 2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2) 3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)3) N-heptyl, n-octyl, and the like.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "trans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 8 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms; in another embodiment, the alkenyl group contains 2 to 3 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. a carbon-carbon sp triple bond, wherein said alkynyl radical may optionally be substituted with one or more substituents as described herein. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms; in another embodiment, alkynyl groups contain 2-3 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH)2C.ident.CH), 1-propynyl (-C.ident.C-CH)3) And so on.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 carbon atoms. In one embodiment, the cycloalkyl group contains 3 to 12 carbon atoms; in another embodiment, cycloalkyl groups contain 3 to 10 carbon atoms; in another embodiment, cycloalkyl contains 3 to 8 carbon atoms; in yet another embodiment, the cycloalkyl group contains 3 to 6 carbon atoms. The cycloalkyl groups may be independently unsubstituted or substituted with one or more substituents described herein.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein and refer to a saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring containing 3 to 12 ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur and oxygen atoms, but wherein at least one ring does not belong to the aromatic group. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH2The group may optionally be replaced by-c (o) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thietanyl, oxazepanyl, and the likeRadical diazaRadical, sulfur nitrogen heteroRadicals, indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1]Hept-5-yl. In heterocyclic radicals of-CH2Examples of-groups substituted by-C (O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl and pyrimidinedione. Examples of sulfur atoms in heterocyclic groups being oxidizedIncluding, but not limited to, sulfolane, 1-dioxothiomorpholinyl. The heterocyclyl group may be optionally substituted with one or more substituents as described herein.
In one embodiment, heterocyclyl is a 4-7 atom heterocyclyl and refers to a saturated or partially unsaturated monocyclic ring containing 4-7 ring atoms in which at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. Unless otherwise specified, a heterocyclic group of 4 to 7 atoms may be carbon-based or nitrogen-based, and-CH2The group may optionally be replaced by-c (o) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclic groups consisting of 4 to 7 atoms include, but are not limited to: azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thioxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepanyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, morpholinyl, thiomorpholinyl, piperazinyl, dihydromorpholinylRadical diazaRadical, sulfur nitrogen heteroAnd (4) a base. In heterocyclic radicals of-CH2Examples of-groups substituted by-C (O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl and pyrimidinedione. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group, 1-dioxothiomorpholinyl group. Said heterocyclyl group consisting of 4 to 7 atoms may optionally be substituted by one or more substituentsSubstituted by the substituents described in the invention.
In another embodiment, heterocyclyl is a 4-atom heterocyclyl and refers to a saturated or partially unsaturated monocyclic ring containing 4 ring atoms in which at least one ring atom is substituted by a member selected from the group consisting of nitrogen, sulfur, and oxygen atoms. Unless otherwise specified, a heterocyclic group consisting of 4 atoms may be carbon-based or nitrogen-based, and-CH2The group may optionally be replaced by-c (o) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclic groups consisting of 4 atoms include, but are not limited to: azetidinyl, oxetanyl, thietanyl. The 4-atom heterocyclyl group may be optionally substituted with one or more substituents described herein.
In another embodiment, heterocyclyl is a5 atom heterocyclyl and refers to a saturated or partially unsaturated monocyclic ring containing 5 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. Unless otherwise specified, a 5-atom heterocyclic group may be carbon-based or nitrogen-based, and-CH2The group may optionally be replaced by-c (o) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of 5-atom heterocyclic groups include, but are not limited to: pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl. In heterocyclic radicals of-CH2Examples of the-group substituted by-C (O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group. The 5-atom heterocyclyl group may be optionally substituted with one or more substituents described herein.
In another embodiment, heterocyclyl is a 6-atom heterocyclyl and refers to saturated or unsaturated compounds containing 6 ring atomsA partially unsaturated monocyclic ring, wherein at least one ring atom is selected from nitrogen, sulfur and oxygen atoms. Unless otherwise specified, a heterocyclic group of 6 atoms may be carbon-based or nitrogen-based, and-CH2The group may optionally be replaced by-c (o) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclic groups consisting of 6 atoms include, but are not limited to: tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl. In heterocyclic radicals of-CH2Examples of-groups substituted with-C (O) -include, but are not limited to, 2-piperidinonyl, 3, 5-dioxopiperidinyl and pyrimidinedione. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, 1, 1-dioxothiomorpholinyl. The 6-atom heterocyclyl group may be optionally substituted with one or more substituents described herein.
In yet another embodiment, heterocyclyl is a 7-12 atom heterocyclyl and refers to a saturated or partially unsaturated spiro-or fused-bicyclic ring containing 7-12 ring atoms in which at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms. Unless otherwise specified, a heterocyclic group of 7 to 12 atoms may be carbon-based or nitrogen-based, and-CH2The group may optionally be replaced by-c (o) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclic groups consisting of 7 to 12 atoms include, but are not limited to: indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1]Hept-5-yl. Said heterocyclyl group of 7 to 12 atoms may be optionally substituted by one or more substituents as described herein.
The term "heterocyclylalkyl" includes heterocyclyl-substituted alkyl groups; the term "heterocyclylalkoxy" includes heterocyclyl-substituted alkoxy groups in which an oxygen atom is attached to the remainder of the molecule; the term "heterocyclylalkylamino" includes heterocyclyl-substituted alkylamino groups in which the nitrogen atom is attached to the remainder of the molecule. Wherein heterocyclyl, alkyl, alkoxy and alkylamino have the meanings as described herein, and such examples include, but are not limited to, pyrrol-2-ylmethyl, morpholin-4-ylethyl, morpholin-4-ylethoxy, piperazin-4-ylethoxy, piperidin-4-ylethylamino and the like.
The term "n-atomic" where n is an integer typically describes the number of ring-forming atoms in a molecule in which the number of ring-forming atoms is n. For example, piperidinyl is a heterocycloalkyl group of 6 atoms, while 1,2,3, 4-tetrahydronaphthalene is a cycloalkyl group of 10 atoms.
The term "unsaturated" as used herein means that the group contains one or more unsaturations.
The term "heteroatom" refers to O, S, N, P and Si, including N, S and any oxidation state form of P; primary, secondary, tertiary amines and quaternary ammonium salt forms; or a form in which a hydrogen on a nitrogen atom in the heterocycle is substituted, for example, N (like N in 3, 4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like NR in N-substituted pyrrolidinyl).
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
The term "aryl" denotes monocyclic, bicyclic and tricyclic carbon ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring system is aromatic, wherein each ring system comprises a ring of 3 to 7 atoms with one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". Examples of the aryl group may include phenyl, naphthyl and anthracenyl. The aryl group may independently be optionally substituted with one or more substituents described herein.
The term "heteroaryl" denotes monocyclic, bicyclic and tricyclic ring systems containing 1 to 9 carbon atoms, and at least one ring system containing one or more heteroatoms selected from O, S and N, at least one ring system being aromatic, wherein each ring system contains a ring of 5 to 7 atoms with one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". The heteroaryl group is optionally substituted with one or more substituents described herein.
In some embodiments, "heteroaryl" is a monocyclic, bicyclic, and tricyclic ring system containing 5-12 ring atoms, wherein at least one ring system is aromatic and at least one ring system contains one or more heteroatoms selected from O, S, and N, wherein each ring system contains a ring of 5-7 atoms with one or more attachment points attached to the rest of the molecule. In some embodiments, a "heteroaryl" group is a monocyclic or bicyclic ring system containing 5 to 10 ring atoms, wherein at least one ring system is aromatic and at least one ring system contains one or more heteroatoms selected from O, S and N, wherein each ring system contains a ring of 5 to 7 atoms with one or more attachment points to the rest of the molecule. In yet another embodiment, a "heteroaryl" group is an aromatic monocyclic ring system of 5-6 atoms and contains 1,2,3 or 4 heteroatoms independently selected from O, S and N, and has one or more attachment points to the rest of the molecule. Examples of heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 1,2, 3-triazolyl, and 1,2, 4-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, oxadiazolyl (e.g., 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, and 1,2, 4-oxadiazolyl), 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, pyrazinyl, 1,3, 5-triazinyl; the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridyl, and the like.
The term "carboxy", whether used alone or in combination with other terms, such as "carboxyalkyl", denotes-CO2H; the term "carbonyl", whether used alone or in combination with other terms, such as "aminocarbonyl" or "acyloxy", denotes — (C ═ O).
The term "alkylamino" or "alkylamino" includes "N-alkylamino" and "N, N-dialkylamino" in which the amino groups are each independently substituted with one or two alkyl groups. In some of these embodiments, the alkylamino group is one or two C1-6Lower alkylamino groups in which the alkyl group is attached to the nitrogen atom. In other embodiments, the alkylamino group is C1-3Lower alkylamino groups of (a). Suitable alkylamino groups can be monoalkylamino or dialkylamino, and such examples include, but are not limited to, N-methylamino, N-ethylamino, N-dimethylamino, N-diethylamino, and the like.
The term "arylamino" denotes an amino group substituted with one or two aryl groups, examples of which include, but are not limited to, N-phenylamino. In some embodiments, the aromatic ring on the arylamino group may be further substituted.
The term "aminoalkyl" includes C substituted with one or more amino groups1-10A straight or branched alkyl group. In some of these embodiments, aminoalkyl is C substituted with one or more amino groups1-6"lower aminoalkyl" such examples include, but are not limited toLimited to aminomethyl, aminoethyl, aminopropyl, aminobutyl and aminohexyl.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein.
Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-PrO, n-propoxy, -OCH)2CH2CH3) 2-propoxy (i-PrO, i-propoxy, -OCH (CH)3)2) 1-butoxy (n-BuO, n-butoxy, -OCH)2CH2CH2CH3) 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH)2CH(CH3)2) 2-butoxy (s-BuO, s-butoxy, -OCH (CH)3)CH2CH3) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH)3)3) 1-pentyloxy (n-pentyloxy, -OCH)2CH2CH2CH2CH3) 2-pentyloxy (-OCH (CH)3)CH2CH2CH3) 3-pentyloxy (-OCH (CH))2CH3)2) 2-methyl-2-butoxy (-OC (CH))3)2CH2CH3) 3-methyl-2-butoxy (-OCH (CH)3)CH(CH3)2) 3-methyl-l-butoxy (-OCH)2CH2CH(CH3)2) 2-methyl-l-butoxy (-OCH)2CH(CH3)CH2CH3) And so on.
The term "alkylthio" denotes a group resulting from replacement of an oxygen atom in an alkoxy group by a sulfur atom.
The terms "haloalkyl", "haloalkenyl" or "haloalkoxy" denote alkyl, alkenyl or alkoxy groups substituted with one or more halogen atoms, examples of which include, but are not limited to, trifluoromethyl, difluoroethyl (CHF)2CH2-), trifluoromethoxy, and the like.
The term "hydroxyalkyl" or "hydroxy-substituted alkyl" means that the alkyl group is substituted with one or more hydroxy groups, wherein the alkyl group has the meaning described herein. Examples include, but are not limited to, hydroxymethyl, hydroxyethyl, 1, 2-dihydroxyethyl, and the like.
The term "amino" (alone or in combination with other terms) means-NH2
The term "protecting group" or "PG" refers to a substituent that, when reacted with other functional groups, is generally used to block or protect a particular functionality. For example, "amino protecting group" refers to a substituent attached to an amino group to block or protect the functionality of the amino group in a compound, and suitable amino protecting groups include acetyl, trifluoroacetyl, t-butyloxycarbonyl (BOC ), benzyloxycarbonyl (CBZ ) and 9-fluorenylmethylenoxycarbonyl (Fmoc). Similarly, "hydroxyl protecting group" refers to the functionality of a substituent of a hydroxyl group to block or protect the hydroxyl group, and suitable protecting groups include acetyl and silyl groups. "carboxy protecting group" refers to the functionality of a substituent of a carboxy group to block or protect the carboxy group, and typical carboxy protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General descriptions of protecting groups can be found in the literature: greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.
Used in the inventionThe term "prodrug" of (a) represents a compound which is converted in vivo to a compound of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel delivery systems, Vol.14of the A.C.S.Symphosium Series, Edward B.Roche, ed., Bioredeployers in Drug designs, American Pharmaceutical Association and PergammonPress, 1987, J.Rautio et al, Prodrug: Design and Clinical Applications, Nature review delivery, 2008,7,255 and 270, S.J.Hecker et al, Prodrugs of pharmaceuticals and phosphates, Journal of chemical Chemistry,2008,51,2328 and 2345.
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, descripberphPharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, which are formed by reaction with amino groups, or which are obtained by other methods described in the literature, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, lauryl sulfates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, bisulfates, salts of sodium, potassium, sodium, potassium, sodium, Thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
The term "hydrate" refers to an association of solvent molecules that is water.
"nitroxide" in the context of the present invention means that when a compound contains several amine functional groups, 1 or more than 1 nitrogen atom can be oxidized to form an N-oxide. Specific examples of N-oxides are N-oxides of tertiary amines or N-oxides of nitrogen-containing heterocyclic nitrogen atoms. The corresponding amines can be treated with an oxidizing agent such as hydrogen peroxide or a peracid (e.g., peroxycarboxylic acid) to form the N-oxide (see Advanced Organic Chemistry, Wiley Interscience, 4 th edition, Jerry March, pages). In particular, the N-oxide may be prepared by the method of L.W.Deady (Syn.Comm.1977,7,509-514), for example by reacting an amine compound with m-chloroperoxybenzoic acid (MCPBA) in an inert solvent such as dichloromethane.
The term "treating" or "treatment" as used herein refers, in some embodiments, to ameliorating a disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
Pharmaceutically acceptable acid addition salts may be formed with inorganic and organic acids, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheophylline, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, Polysilonolactates, propionates, stearates, succinates, sulfosalicylates, tartrates, tosylates and trifluoroacetates.
Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, sulfosalicylic acid, and the like.
Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of groups I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (cholinate), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of pharmaceutical salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable salts.
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms.
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H,3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl and125I。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined hereinSubstances, e.g. in which radioactive isotopes are present, e.g.3H、14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13C. the isotopically enriched compounds can be used for metabolic studies (use)14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stability. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is considered as a substituent of the compound of formula (I). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D2O, acetone-d6、DMSO-d6Those solvates of (a).
In another aspect, the invention relates to intermediates for the preparation of compounds encompassed by formula (I).
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds encompassed by formula (I).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention, a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle, or combination thereof. In some embodiments, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
"combination" means a fixed combination or a kit of parts for combined administration in the form of a single dosage unit, wherein a compound disclosed herein and a combination partner may be administered separately at the same time or may be administered separately within certain time intervals, in particular such that the combination partners exhibit a cooperative, e.g. synergistic, effect. The terms "co-administration" or "co-administration" and the like as used herein are intended to encompass the administration of the selected combination partners to a single individual in need thereof (e.g., a patient), and are intended to encompass treatment regimens in which the substances are not necessarily administered by the same route of administration or simultaneously. The term "pharmaceutical combination product" as used herein denotes a product obtained by mixing or combining more than one active ingredient and includes both fixed and non-fixed combinations of active ingredients. The term "fixed combination" means that the active ingredients, such as the disclosed compounds and combination partners, are administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, such as the disclosed compounds and combination partners, are both administered to a patient as separate entities simultaneously, jointly or sequentially with no specific time limits, wherein the administration provides therapeutically effective levels of both compounds in the patient. The latter is also applicable to cocktail therapy, e.g. administering 3 or more active ingredients.
It should be noted that the term "inhibiting HCV viral proteins" in the present invention is to be understood in a broad sense, and includes both the level of inhibiting expression of HCV viral proteins and the level of inhibiting activity, assembly and release of HCV viral proteins. Among them, HCV protein expression levels include, but are not limited to: the level of translation of viral protein genes, the level of post-translational modification of proteins, the level of replication of progeny genetic material, and the like.
Description of the Compounds of the invention
The invention relates to a macrocyclic compound and a pharmaceutical preparation thereof, which can effectively inhibit HCV infection, in particular can inhibit the activity of HCV NS3/4A protein.
In one aspect, the invention relates to a compound that is a compound of formula (I ') or a stereoisomer, tautomer, enantiomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt, or prodrug of a compound of formula (I'),
wherein: ring A is C6-10Aryl or C1-9A heteroaryl group; the A ring is optionally substituted with 1,2,3 or 4R2Substituted;
R1is C6-10Aryl or C1-9A heteroaryl group;
each R2And R3Independently H, F, Cl, Br, I, amino, hydroxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl or C1-9A heteroaryl group;
R4is H, deuterium or C1-6An alkyl group;
n is 0, 1,2,3 or 4;
said C is1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl radical, C1-9Heteroaryl or amino is independently optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C1-6Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl or C1-9Heteroaryl group is substituted.
In some embodiments, wherein ring a is phenyl or heteroaryl of 5 to 6 ring atoms; said phenyl or heteroaryl of 5-6 ring atoms is optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3Alkenyl radical, C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy, C1-3Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclyl or C6-10Aryl group.
In still other embodiments, ring a is phenyl, furyl, thienyl, thiazolyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyridyl, quinolinyl, indolyl, or acridinyl.
In some embodiments, the compound has the structure shown in formula (I):
or stereoisomers, tautomers, enantiomers, nitrogen oxides, hydrates, solvates, metabolites and pharmaceutically acceptable salts or prodrugs thereof.
In some embodiments, the compound has the structure shown in formula (II):
or stereoisomers, tautomers, enantiomers, nitrogen oxides, hydrates, solvates, metabolites and pharmaceutically acceptable salts or prodrugs thereof.
In some embodiments, R in formula (I'), formula (I), or formula (II)1Is phenyl or heteroaryl of 5 to 6 ring atoms; said phenyl or heteroaryl of 5-6 ring atoms is optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3Alkenyl radical, C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy, C1-3Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclyl or C6-10Aryl group.
In still other embodiments, R in formula (I'), formula (I), or formula (II)1Is phenyl, furyl, thienyl, thiazolyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, oxadiazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyridyl, quinolyl, indolyl or acridinyl, wherein R is1The group may be optionally substituted with 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, trifluoromethyl, difluoroethyl, trifluoromethoxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, vinyl, ethynyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl or phenyl.
In some embodiments, R in formula (I'), formula (I) or formula (II)2And R3Each independently is H, F, Cl, Br, I, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, trifluoromethyl, trifluoromethoxy, tert-butyl,Ethenyl, propenyl, ethynyl, propynyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl, phenyl, methylamino or ethylamino.
In some embodiments, R in formula (I'), formula (I), or formula (II)4Is H, deuterium, methyl, deuterated methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
In some embodiments, R in formula (I'), formula (I), or formula (II)1Is heteroaryl of 5 to 6 ring atoms; wherein said heteroaryl of 5 to 6 ring atoms is optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3Alkenyl radical, C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy or C1-3Substituted by alkylamino substituents; r in formula (I'), formula (I) or formula (II)2Is C1-6An alkyl group; r in formula (I'), formula (I) or formula (II)3Is C1-3An alkyl group; r in formula (I'), formula (I) or formula (II)4Is H or C1-3An alkyl group.
In some embodiments, the compound has one of the following structures or stereoisomers, tautomers, enantiomers, nitrogen oxides, hydrates, solvates, metabolites and pharmaceutically acceptable salts or prodrugs thereof:
the compounds of the present invention (in the present text, the expressions "compounds of formula (I '), formula (I) or formula (II) or stereoisomers, geometric isomers, tautomers, enantiomers, nitrogen oxides, hydrates, solvates, metabolites and pharmaceutically acceptable salts or prodrugs of compounds of formula (I'), formula (I) or formula (II)) can be used for the manufacture of a pharmaceutical product for the treatment of acute and chronic HCV infections, including those described in the present invention. Further, the compounds of the present invention may be used in the manufacture of anti-HCV preparations. Thus, the compounds of the present invention may be used in the manufacture of a medicament for alleviating, preventing, controlling or treating HCV-mediated disorders, in particular HCV NS3/4A protein-mediated diseases. Thus, the compounds of the present invention may be used as active ingredients of pharmaceutical compositions which may comprise a compound represented by formula (I'), formula (I) or formula (II), and may further comprise at least one pharmaceutically acceptable carrier, adjuvant or diluent.
In particular, the salts are pharmaceutically acceptable salts. The term "pharmaceutically acceptable" means that the substance or composition employed must be compatible chemically or toxicologically with the other ingredients comprising the formulation and the mammal being treated. The "pharmaceutically acceptable" substance or composition may be specifically selected by those skilled in the art depending on the other components employed and the subject, e.g., human, being treated.
Salts of the compounds of the present invention also include, but are not necessarily pharmaceutically acceptable salts of, intermediates used in the preparation or purification of a compound of formula (I '), formula (I) or formula (II), or isolated enantiomers of a compound of formula (I'), formula (I) or formula (II).
If the compounds of the invention are basic, the desired salts may be prepared by any suitable method provided in the literature, for example, using inorganic or organic acids. Among them, examples of the inorganic acid include, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Examples of organic acids include, but are not limited to, acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, and salicylic acid; pyranonic acids, such as glucuronic acid and galacturonic acid; alpha-hydroxy acids such as citric acid and tartaric acid; amino acids such as aspartic acid and glutamic acid; aromatic acids such as benzoic acid and cinnamic acid; sulfonic acids such as p-toluenesulfonic acid, ethanesulfonic acid, and the like.
If the compounds of the invention are acidic, the desired salts can be prepared by suitable methods, e.g., using inorganic or organic bases, such as ammonia (primary, secondary, tertiary), alkali or alkaline earth metal hydroxides, and the like. Suitable salts include, but are not limited to, organic salts derived from amino acids such as glycine and arginine, ammonia such as primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine, piperazine and the like, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
Compositions, formulations and administration of the Compounds of the invention
The pharmaceutical composition comprises any one of the compounds of the present invention. The pharmaceutical composition may further comprise a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle, or combination thereof. The pharmaceutical composition can be used for treating Hepatitis C Virus (HCV) infection or hepatitis C disease, and particularly has a good inhibition effect on HCV NS3/4A protein.
The pharmaceutical composition further comprises an anti-HCV agent. The anti-HCV agent can be any other known anti-HCV agent other than the compounds of the present invention. For example, it may be interferon, ribavirin, interleukin 2, interleukin 6, interleukin 12, a compound that promotes the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, imiqimod, an inosine 5' -monophospate dehydrogenase inhibitor, amantadine, rimantadine, a pharmaceutically acceptable salt thereof,bavin-Xib, CivacirTMThe HCV target protein is selected from a group consisting of a HCV NS-2129, a HCV target protein, a HCV target protein, a HCV target protein, a polypeptide, a protein, a HCV protein, a protein, a-protein, a protein-polypeptide, a protein, a protein, a-polypeptide-6, a-polypeptide-.
When useful in therapy, a therapeutically effective amount of a compound of the present invention, particularly a compound of formula (I) and pharmaceutically acceptable salts thereof, may be administered as the raw chemical or as the active ingredient of a pharmaceutical composition. Accordingly, the present disclosure also provides pharmaceutical compositions comprising a therapeutically effective amount of a compound of the present invention, particularly a compound of formula (I) or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients. The term "therapeutically effective amount" as used herein refers to the total amount of each active component sufficient to show meaningful patient benefit (e.g., reduction in viral load). When the active ingredient alone is used for separate administration, the term refers only to that ingredient. When used in combination, the term refers to the combined amounts of the active ingredients that, when combined, administered sequentially or simultaneously, result in a therapeutic effect. The compounds of the invention, especially the compounds of formula (I) and pharmaceutically acceptable salts thereof, are as described above. The carrier, diluent or excipient must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. According to another aspect of the present disclosure there is also provided a process for the preparation of a pharmaceutical formulation which comprises mixing a compound of the present invention, especially a compound of formula (I) or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients. The term "pharmaceutically acceptable" as used herein refers to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and which are effective for their intended use.
The pharmaceutical preparations may be in unit dosage form, each unit dosage containing a predetermined amount of the active ingredient. Dosage levels of the compounds of the present disclosure are between about 0.01 and about 250 mg/kg body weight/day, preferably between about 0.05 and about 100mg/kg body weight/day, often as monotherapy for the prevention or treatment of HCV-mediated diseases. The pharmaceutical compositions of the present disclosure may generally be administered from about 1 to about 5 times per day or as a continuous infusion. Such administration may be used as a long term or short term therapy. The amount of active ingredient mixed with a carrier material to prepare a single dosage form will vary depending on the disease to be treated, the severity of the disease, the time of administration, the route of administration, the rate of excretion of the compound used, the time of treatment and the age, sex, body weight and condition of the patient. Preferred unit dosage forms are those containing a daily or divided dose or suitable fraction thereof of the active ingredient described herein above. Treatment can be initiated with small doses, which are clearly below the optimal dose of the compound. Thereafter, the dosage is increased in smaller increments until the optimum effect is achieved in this case. In general, the compounds are most desirably administered at concentration levels that generally provide effective results in terms of antiviral efficacy without causing any harmful or toxic side effects.
When the compositions of the present disclosure comprise a combination of a compound of the present disclosure and one or more other therapeutic or prophylactic agents, the dosage level of the compound and the additional agent(s) will generally be from about 10% to about 150% of the normally administered dose, more preferably from about 10% to about 80% of the normally administered dose, in a monotherapy regimen. The pharmaceutical formulations are adapted for administration by any suitable route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intradermal, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional, intravenous or subdermal injection or infusion) route. Such formulations may be prepared by any method known in the art of pharmacy, for example by mixing the active ingredient with a carrier or excipient. Oral administration or injection administration is preferred.
Pharmaceutical formulations adapted for oral administration are provided in discrete units, such as capsules or tablets; powder or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foam or foam formulations (whip); or an oil-in-water emulsion or a water-in-oil emulsion.
For example, for oral administration in the form of a tablet or capsule, the active pharmaceutical ingredient may be mixed with a pharmaceutically acceptable oral, non-toxic inert carrier (e.g., ethanol, glycerol, water, etc.). Powders are prepared by pulverizing the compound to a suitable fine size and mixing with a pharmaceutically acceptable carrier (e.g., an edible sugar such as starch or mannitol) which is also pulverized. Flavoring, preservative, dispersing and coloring agents may also be present.
Capsules are prepared by preparing a powdered mixture as described above and filling into shaped gelatin shells. Glidants and lubricants (e.g., colloidal silicon dioxide, talc, magnesium stearate, calcium stearate, or solid polyethylene glycol) may be added to the powder mixture prior to the filling operation. Disintegrating or solubilizing agents (e.g., agar-agar, calcium carbonate or sodium carbonate) that will improve the availability of the drug when the capsule is taken can also be added.
In addition, if desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars (e.g., glucose or beta-lactose), corn sweeteners, natural and synthetic gums (e.g., gum arabic, tragacanth or sodium alginate), carboxymethylcellulose, polyethylene glycol, and the like. Lubricants used in these dosage forms include sodium oleate, sodium chloride, and the like. Disintegrants include, but are not limited to, starch, methylcellulose, agar, bentonite, xanthan gum, and the like. For example, tablets are prepared by making a powder mixture, granulating or slugging, adding a lubricant and a disintegrant, and compressing into tablets. The powdered mixture is prepared by mixing the appropriately comminuted compound with a diluent or base as described above, optionally with a binder (for example carboxymethylcellulose, alginates, gelatin or polyvinylpyrrolidone), a dissolution inhibitor (for example paraffin), an absorption accelerator (quaternary salt) and/or an absorbent (for example bentonite, kaolin or dicalcium phosphate). The powdered mixture may be granulated by wetting with a binder such as syrup, starch slurry, acacia slurry (acadia mucilage) or a solution of cellulosic or polymeric material and pressure sieving. An alternative to granulation is to pass the powder mixture through a tablet press, with the result that poorly formed agglomerates are broken up into granules. The granules may be lubricated by the addition of stearic acid, a stearate salt, talc or mineral oil to prevent sticking to the dies of the tablet press. The lubricated mixture is then compressed into tablets. The compounds of the present disclosure may also be combined with a free-flowing inert carrier and compressed into tablets without going through a granulation or pre-compression step. Transparent or opaque protective coating materials may be provided which consist of a shellac coating, a sugar coating or a coating of a polymeric material and a waxy polishing coating (wax). Dyes may be added to these coatings to distinguish different unit doses.
Oral liquid preparations such as solutions, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs can be prepared through the use of non-toxic vehicles. Solubilizing agents and emulsifiers (e.g., ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers), preservatives, flavoring additives (e.g., peppermint oil or natural sweeteners or saccharin or other artificial sweeteners), and the like may also be added.
Dosage unit formulations for oral administration may be microencapsulated, if appropriate. The formulations may also be formulated for extended or sustained release, for example by coating or embedding in a particulate material such as a polymer, wax or the like.
The compounds of the invention, particularly the compounds of formula (I) and pharmaceutically acceptable salts thereof, may also be administered in liposomal delivery systems, such as small unilamellar liposomes, large unilamellar liposomes, and multilamellar liposomes. Liposomes can be composed of a variety of phospholipids (e.g., cholesterol, octadecylamine, or phosphatidylcholine).
The compounds of the invention, especially the compounds of formula (I) and pharmaceutically acceptable salts thereof, may also be delivered by using the monoclonal antibody as a separate carrier to which the compound molecule is coupled. The compounds may also be conjugated to soluble polymers as targetable drug carriers. Such polymers may include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide phenol, polyhydroxyethylaspartamide phenol, or polyethyleneoxide polylysine substituted with palmitoyl residues. In addition, the compounds may be coupled to a class of biodegradable polymers for achieving controlled release of a drug, such as polylactic acid, poly-caprolactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and crosslinked or amphipathic block copolymers of hydrogels.
Pharmaceutical formulations adapted for transdermal administration may be presented as discrete patches (patches) to remain in intimate contact with the epidermis of the recipient for an extended period of time. For example, the active ingredient may be delivered by iontophoretic patches, as generally described in Pharmaceutical Research 1986,3(6), 318.
Pharmaceutical preparations suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols, oils or transdermal patches.
Pharmaceutical formulations adapted for rectal administration may be presented as suppositories or as enemas.
Pharmaceutical formulations suitable for nasal administration, wherein the carrier is a solid, include coarse powders having a particle size in the range of, for example, 20 to 500 microns, which are administered by nasal inhalation, i.e. by rapid inhalation through the nasal passage from a coarse powder container adjacent the nose. Suitable formulations in which the carrier is a liquid, suitable for administration as a nasal spray or nasal drops, include aqueous or oily solutions of the active ingredient.
Pharmaceutical formulations suitable for administration by inhalation include finely divided particulate powders (dust) or mists (mist), which may be prepared in different types of metered dose compressed aerosols, nebulised inhalers, insufflators or other devices adapted to deliver aerosol sprays.
Pharmaceutical formulations adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
Pharmaceutical formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions, which may contain antioxidants, buffers, bacteriostats and solutes that render the formulation isotonic with the blood of the recipient, and aqueous and non-aqueous sterile suspensions, which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed amkside and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. The injection solution and suspension can be prepared into sterile powder for injection, granule and tablet.
It will be appreciated that in addition to the ingredients particularly mentioned above, the formulations may include other ingredients conventional in the art having regard to the type of formulation in question, for example, such formulations which are suitable for oral administration may include flavouring agents.
Use of the Compounds and pharmaceutical compositions of the invention
The present invention provides the use of a compound or pharmaceutical composition of the invention in the manufacture of a medicament for inhibiting the HCV replication process and/or inhibiting the function of HCV viral proteins; the HCV replication process comprises HCV entry, HCV uncoating, HCV translation, HCV replication, HCV assembly, or HCV release; the HCV viral protein is selected from metalloprotease, NS2, NS3, NS4A, NS4B, NS5A or NS5B, and an Internal Ribosome Entry Site (IRES) and inosine monophosphate dehydrogenase (IMPDH) required for HCV viral replication. Any compound or pharmaceutical composition of the invention can be used for treating Hepatitis C Virus (HCV) infection or hepatitis C disease, and particularly has good inhibition effect on HCV NS3/4A protein.
A method of treatment comprising administering a compound or pharmaceutical composition of the invention further comprising administering to a patient an additional HCV agent, whereby a compound of the invention may be administered in combination therapy with an additional HCV agent, wherein the anti-HCV agent is interferon, ribavirin, interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, imiqimod, an inosine 5' -monophospate dehydrogenase inhibitor, amantadine, rimantadine, bavacizumab, CivacirTMPoplarvir, telaprevir, erlotinib, daclatasvir, simeprevir, asunaprevir, vanipredir, faldaprevir, paritaprevir, danoprrevir, sovaprevir, gradoprrevir, vedrorevir, BZF-961, GS-9256, narloprevir, ANA975, ombitasvir, EDP239, PPI-668, velpatasvir, dolpatasvir,The interferon of the interferon composition is a compound interferon of samatasvir, elbasvir, MK-8325, GSK-2336805, PPI-461, BI-2013335, cilaprevir, ACH-1095, VX-985, IDX-375, VX-500, VX-813, PHX-1766, PHX-2054, IDX-136, IDX-316, modithromycin, VBY-376, TMC-649128, meritibabine, sofosbuvir, INX-189, IDX-184, IDX102, R-1479, UNX-08189, PSI-6130, PSI-938, PSI-879, nesbuvir, HCV-371, VCH-916, lomibuvir, MK-3281, dasbuvir, ABT-865, filibuvir, eeuvivir, teobrevir, JVA-837093, JMAb-835, MK-3281, MK-369, GABA-369, MK-369, BCG-35, BCG-24, BCG-4695, BCG-24, BCG-35, BCG-24, BCG-35, BCG-19, BCG-24, BCG-35, BCG-24, BCG-19, BCG-27, BCG-III-27, BCG-19, BCG.
And a method of treatment comprising administering a compound or pharmaceutical composition of the invention, further comprising administering an additional anti-HCV agent, wherein the additional anti-HCV agent may be administered in combination with a compound or pharmaceutical composition of the invention as a single dosage form, or as separate compounds or pharmaceutical compositions as part of a multiple dosage form. Other anti-HCV agents may be administered with or without the compounds of the present invention. In the latter case, administration may be carried out by, for example, shifting between 6 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, or 2 months.
An "effective amount" or "effective dose" of a compound or pharmaceutically acceptable composition of the invention refers to an amount effective to treat or reduce the severity of one or more of the conditions mentioned herein. The compounds and compositions according to the methods of the present invention can be administered in any amount and by any route effective to treat or reduce the severity of the disease. The exact amount necessary will vary depending on the patient, depending on the race, age, general condition of the patient, severity of infection, particular factors, mode of administration, and the like. The compound or composition may be administered in combination with one or more other therapeutic agents, as discussed herein.
General synthetic procedure
In general, the compounds of the present invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
The examples described below, unless otherwise indicated, are all temperatures set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Inc., Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin HaoLiyu Chemicals Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaseiki chemical plant.
The anhydrous tetrahydrofuran, dioxane, toluene and ether are obtained through reflux drying of metal sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, petroleum ether, N-hexane, N-dimethylacetamide and N, N-dimethylformamide were used by being dried beforehand over anhydrous sodium sulfate.
The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. The glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants. Nuclear magnetic resonance spectroscopy with CDC13、DMSO-d6、CD3OD or acetone-d6As solvent (reported in ppm) TMS (0ppm) or chloroform (7.25ppm) was used as reference standard. When multiple peaks occur, the following abbreviations will be used: s (singleton), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad), dd (doublet of doublets), dt (doublet of triplets), and dt (doublet of triplets). Coupling constants are expressed in hertz (Hz).
Low resolution Mass Spectral (MS) data were measured by an Agilent 6320 series LC-MS spectrometer equipped with a G1312A binary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315B DAD detector were applied for analysis, and an ESI source was applied to the LC-MS spectrometer.
Low resolution Mass Spectral (MS) data were determined by Agilent 6120 series LC-MS spectrometer equipped with a G1311A quaternary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315D DAD detector were used for analysis, and an ESI source was used for the LC-MS spectrometer.
Both spectrometers were equipped with an Agilent Zorbax SB-C18 column, 2.1X 30mm, 5 μm. The injection volume is determined by the sample concentration; the flow rate is 0.6 mL/min; peaks of HPLC were recorded by UV-Vis wavelength at 210nm and 254 nm. The mobile phases were 0.1% formic acid in acetonitrile (phase a) and 0.1% formic acid in ultrapure water (phase B). Gradient elution conditions are shown in table 1:
TABLE 1
Time (min) A(CH3CN,0.1%HCOOH) B(H2O,0.1%HCOOH)
0-3 5-100 95-0
3-6 100 0
6-6.1 100-5 0-95
6.1-8 5 95
Compound purification was assessed by Agilent 1100 series High Performance Liquid Chromatography (HPLC) with UV detection at 210nm and 254nm, a Zorbax SB-C18 column, 2.1X 30mm, 4 μm, 10min, flow rate 0.6mL/min, 5-95% (0.1% formic acid in acetonitrile) in (0.1% formic acid in water), the column temperature was maintained at 40 ℃.
The following acronyms are used throughout the invention:
ac acetyl group
Ac2O acetic anhydride
BBr3Boron tribromide
Br2Bromine compound
BOC, Boc tert-butoxycarbonyl
BAST bis (2-methoxyethyl) aminosulfur trifluoride
Cs2CO3Cesium carbonate
CHCl3Chloroform
CDC13Deuterated chloroform
Cu copper
CuI cuprous iodide
CH2Cl2DCM dichloromethane
CDI N, N' -carbonyldiimidazole
DBU 1, 8-diazabicyclo [5.4.0] -undec-7-ene
DCE 1, 2-dichloroethane
DMF N, N-dimethylformamide
DMAP 4-dimethylaminopyridine
DMSO dimethyl sulfoxide
Dippa Azoic Diphenyl phosphate
DIPEA diisopropylethylamine
DME ethylene glycol dimethyl ether
DPPPy diphenyl-2-pyridylphosphine
DBAD azodicarboxylic acid di-tert-butyl ester
DIAD diisopropyl azodicarboxylate
Dess-Martin (Dess-Martin oxidant) (1,1, 1-triacetoxy) -1, 1-dihydro-1, 2-phenyliodoyl-3 (1H) -one
EDC, EDCI 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride
EtOAc ethyl acetate
EA Ethyl acetate
Et2O Ether
Et2NSF3Diethylamine sulfur trifluoride
Et3N, TEA Triethylamine
EtOH ethanol
Fe iron
HCl & EA, HCl/EA hydrogen chloride in ethyl acetate
HATU 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate
HBr hydrobromic acid
HCl hydrochloric acid
HOAt, HOAT 1-hydroxy-7-azabenzotriazole
HOBT 1-hydroxybenzotriazole
H2Hydrogen gas
H2O2Hydrogen peroxide
H2O water
HOAc acetic acid
I2Iodine
IPA isopropyl alcohol
IMPDH inosine monophosphate dehydrogenase
IRES internal ribosome entry Point
K2CO3Potassium carbonate
KOH potassium hydroxide
LDA lithium diisopropylamide
LiHMDS lithium hexamethyldisilazide
LiN(SiMe3)2Lithium bis (trimethylsilyl) amide
Lawesson's Reagent 2, 4-bis (4-methoxyphenyl) -1, 3-dithio-2, 4-phosphane-2, 4-disulfide
MTBE methyl tert-butyl ether
MsOH methanesulfonic acid
MCPBA m-chloroperoxybenzoic acid
MgSO4Magnesium sulfate
MeOH,CH3OH methanol
MeI methyl iodide
MeCN,CH3CN acetonitrile
mL of
NH3Ammonia
NH4C1 Ammonia chloride
NMP N-methylpyrrolidone
NIS N-iodosuccinimide
N2Nitrogen gas
NaHCO3Sodium bicarbonate
NaBH4Sodium borohydride
NaBH3CN Cyanoborohydride sodium salt
NaOtBu tert-butyl sodium alcoholate
NaOH sodium hydroxide
NaClO2Sodium chlorite
NaCl sodium chloride
NaH2PO4Sodium dihydrogen phosphate
NaH sodium hydride
NaI sodium iodide
Na2SO4Sodium sulfate
NADPH reduced coenzyme II
NBS N-bromosuccinimide
PPh3MeBr Bromomethyltriphenylphosphine
P(t-bu)3Tri (tert-butyl) phosphine
Pd/C Palladium/carbon
PE Petroleum ether (60-90 deg.C)
PBS phosphate buffered saline
POC13Phosphorus oxychloride
PPA polyphosphoric acid
Pd(PPh3)4Tetratriphenylphosphine palladium
Pd(dppf)Cl21,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
PhNTf2N-phenyl bis (trifluoromethanesulfonyl) imide
p-TSA-p-toluenecarboxylic acid
RT, RT Room temperature
rf reflow
Rt Retention time
SEMCl 2- (trimethylsilyl) ethoxymethyl chloride
t-BuOLi tert-Butanolate lithium
TBME methyl tert-butyl ether
TBTU O-benzotriazole-N, N, N ', N' -tetramethyluronium tetrafluoroborate
THF tetrahydrofuran
TFA trifluoroacetic acid
TBAI tetrabutylammonium iodide
TEAF Triethylamine Carboxylic acid
Tf2O-Trifluoromethanesulfonic anhydride
TFAA trifluoroacetic anhydride
TsOH p-toluenesulfonic acid
TsCl tosyl chloride
TMSA trimethylsilyl acetylene
TMSCl trimethylchlorosilane
TBDMSOTf tert-butyl dimethyl p-toluene sulfonic silane
TCCA trichloroisocyanuric acid
TEMPO 2,2,6, 6-tetramethylpiperidine-nitrogen-oxide
TEBAC benzyl triethyl ammonium chloride
Synthesis method
Synthesis method 1
Compound 8 can be prepared by synthetic method 1, wherein R2And R3Have the meaning as described in the present invention. Firstly, a compound 1 and a compound 2 are subjected to ring closure reaction to obtain a compound 3, the compound 3 is hydrolyzed under the action of alkali (such as lithium hydroxide, sodium hydroxide and the like), and a hydrolysis product is reacted with acyl chloride (such as oxalyl chloride, thionyl chloride and the like) to obtain a compound 5. The compound 5 reacts with the compound 6 to obtain a compound 7, and finally the compound 7 forms a ring under the action of alkali (such as potassium tert-butoxide, sodium tert-butoxide and the like) to form a compound 8.
Synthesis method 2
Compound 13 can be prepared by synthetic method 2, wherein R4The condensing agent has the meaning of the invention, the condensing agent is CDI, EDCI, HATU and the like, and the acid is hydrochloric acid, sulfuric acid, p-toluenesulfonic acid and the like. Firstly, compound 10 and compound 11 are subjected to the action of a condensing agent to obtain compound 12, and then compound 12 is further deprotected under an acidic condition to obtain compound 13.
The synthesis method 3:
compound 20 can be prepared by synthesis method 3. Firstly, compound 14 and compound 15 react under the action of alkali (such as lithium hydroxide, sodium hydroxide, potassium carbonate, sodium carbonate, cesium carbonate and the like) to obtain compound 16, compound 16 is firstly hydrolyzed under the action of alkali (such as lithium hydroxide, sodium hydroxide, potassium hydroxide and the like), and the hydrolysis product is decarboxylated under the action of acid (such as citric acid, hydrochloric acid, sulfuric acid, p-toluenesulfonic acid and the like) to obtain compound 18. Compound 18 is then selectively deacetylated by an enzyme (e.g., deacetylase, etc.) to give compound 19, and finally the protecting group (e.g., Boc, Fmoc, etc.) on compound 19 gives compound 20.
The synthesis method 4 comprises the following steps:
compound 28 can be prepared by synthetic method 4, wherein R1、R2、R3And R4The protective group PG is Boc, Fmoc, Cbz, Bn, PMB and the like, and the used alkali is sodium hydroxide, sodium carbonate, potassium tert-butoxide,Sodium tert-butoxide and the like, X is halogen or hydroxyl, the condensing agent used is CDI, EDCI, HATU and the like, and the macrocyclic catalysts used include Grubb's second generation catalysts, Janz 1B catalysts, Janz 1C catalysts and the like. The compound 8 and the compound 21 are subjected to Mitsunobu reaction to obtain a compound 22, the compound 22 is hydrolyzed under alkaline conditions, and the obtained hydrolysate is further reacted with the compound 13 under the action of a condensing agent to obtain a compound 23. Deprotection of compound 23 affords compound 24, and reaction of compound 24 with compound 20 under the action of a condensing agent affords compound 25. Compound 25 is reacted with a macrocyclic catalyst to form compound 26, which is further deprotected to form compound 27, and finally compound 27 is reacted with compound 38 with a condensing agent to form compound 28.
Synthesis method 5
Compound 28 can also be prepared by synthetic method 5, wherein R1、R2、R3And R4The protecting group PG has the meaning of Boc, Fmoc, Cbz, Bn, PMB and the like, the used alkali is sodium hydroxide, sodium carbonate, potassium tert-butoxide, sodium tert-butoxide and the like, and X is halogen or hydroxyl; the macrocyclic catalysts used include Grubb's second generation catalysts, Jansen 1B catalysts, Jansen 1C catalysts, and the like. The compound 29 and the compound 30 react under the action of a condensing agent to obtain a compound 31, the amino protecting group of the compound 31 is removed to obtain a compound 32, and the compound 32 and the compound 20 react under the action of the condensing agent to obtain a compound 33. Then carrying out Mitsunobu reaction on the compound 33 and the compound 8 to obtain a compound 34, carrying out Mitsunobu reaction on the compound 34 under the action of a macrocyclic catalyst to obtain a compound 35, and carrying out hydrolysis on the compound 35 under the action of alkali to obtain a compound 36. Then the compound 36 reacts with the compound 11 under the action of a condensing agent to obtainCompound 26. And compound 26 is deprotected to give compound 27. Finally, the compound 27 reacts with the compound 38 under the action of a condensing agent to obtain a compound 28.
The synthesis method 6 comprises the following steps:
compound 28 can be prepared by synthetic method 6, wherein R1、R2、R3And R4Have the meaning as described in the present invention. The protecting group PG is Boc, Fmoc, Bn, Cbz and the like. First, the amine protecting group is attached to compound 39 to provide compound 40. Compound 40 undergoes a Mitsunobu reaction to form lactone ring compound 41, and then compound 41 is deprotected to obtain compound 42. The compound 42 and the compound 20 are subjected to condensation reaction to obtain a compound 43, and the used carboxylic reagent is CDI, EDCI, HATU and the like. Compound 43 and compound 30 are reacted with a base (e.g., sodium isooctanoate, sodium carbonate, potassium carbonate, sodium hydroxide, potassium hydroxide, etc.) to form compound 44. The compound 44 reacts with sulfonyl chloride (such as methylsulfonyl chloride, p-toluenesulfonyl chloride, p-bromobenzenesulfonyl chloride, p-nitrobenzenesulfonyl chloride and the like) to obtain an intermediate with a better leaving group. Then, the obtained intermediate and a compound 8 are reacted under the action of a base (such as cesium carbonate, potassium carbonate, sodium carbonate and the like) to form a compound 45, and an amide on the compound 45 is protected to obtain a compound 46. The resulting compound 46 is subjected to olefin metathesis in the presence of a catalyst (e.g., Janz catalyst 1B, Janz catalyst 1C, Grubbs second generation catalyst, etc.) to provide compound 47. Deprotection of compound 47 affords compound 48. The resulting compound 48 is then condensed with compound 38 to provide compound 49, and compound 49 is hydrolyzed with a base (e.g., lithium hydroxide, sodium hydroxide, potassium hydroxide, etc.) to provide compound 50. Finally, compound 50 reacts with compound 11 to give compound 28.
Examples
Example 1
The synthetic route is as follows:
step 1: synthesis of Compounds 1-3
Compound 1-2(20g,150mmol) and compound 1-1(30g,170mmol) were added to ethanol (100mL), then warmed to 75 ℃ and stirred at this temperature for two hours. And after the reaction is completed, cooling to room temperature, removing the ethanol solution under reduced pressure, adding 100mL of water to quench the reaction, and adjusting the pH value of the mixture to 6-8 by using ammonia water. The reaction was then extracted twice with MTBE (100 mL. times.2), the combined organic phases were washed twice with water (50 mL. times.2), separated, and the resulting organic phase was concentrated under reduced pressure to give a crude product 1-3 as a red brown color, which was carried on to the next reaction without further purification.
MS(ESI,pos.ion)m/z:200.3[M+1]+
Step 2: synthesis of Compounds 1-4
Compound 1-3(28g,140mmol) was added to methanol (140mL), then cooled to 0 deg.C and LiOH. H was added2O (7g, 168mmol), stirred at this temperature for 30 minutes, then warmed to room temperature and stirred for an additional 3 hours. After completion of the reaction, the methanol was removed under reduced pressure, then MTBE (140mL) was added and the reaction mixture was warmed to 55 deg.C and stirred at this temperature for 3 hours, then cooled to room temperature and stirred for an additional 2 hours. Filtration was carried out, and the filter cake was washed twice with MTBE (50mL), and the resulting solid was dried under vacuum at 50 ℃ to give compound 1-4(21.2g, two-step reaction yield: 80%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:172.2[M+1]+
And step 3: synthesis of Compounds 1-5
Compound 1-4(20g,113mmol) was added to 100mL DCM, then cooled to 0 deg.C and oxalyl chloride (30g,226mmol) was added slowly dropwise, after which stirring was continued at 0 deg.C for 30min, then warmed to room temperature and stirred for 4 h. After the reaction was completed, filtration was carried out, and the obtained filtrate was concentrated under reduced pressure, and the obtained crude product 1-5 was directly subjected to the next reaction without further purification.
And 4, step 4: synthesis of Compounds 1-7
Compound 1-5(16g, 84mmol) was added to acetone (50mL), cooled to 0 deg.C, then compound 1-6(10g, 55.8mmol) was added, after which stirring was continued at 0 deg.C for 30 minutes, then warmed to room temperature and stirred for 4 hours. After the reaction was complete, the reaction solution was slowly dropped into 500mL of water, a large amount of solid precipitated during stirring, filtered, and the filter cake was washed twice with water (200 mL). The resulting solid was dried under vacuum at 55 ℃ to give compounds 1 to 7(14.8g, yield: 80%) as pale yellow solids.
MS(ESI,pos.ion)m/z:333.2[M+1]+
And 5: synthesis of Compounds 1-8
Compound 1-7(5g,15mmol) and potassium tert-butoxide (3.36g,30mmol) were added to tert-butanol (50mL), and then warmed to 100 ℃ and reacted at this temperature for 8 hours. After completion of the reaction, it was cooled to room temperature, 20mL of t-butanol was removed under reduced pressure, and then the reaction solution was poured into an aqueous HCl solution (0.5M,200mL) at 0 ℃ to produce a large amount of yellow solid during stirring, and filtration was carried out, and the obtained filter cake was washed once with 20mL of water and vacuum-dried at 50 ℃ to obtain a pale yellow solid. The resulting pale yellow solid was added to 25mL of isopropyl ether, warmed to 60 ℃ and stirred for 2 hours, then cooled to room temperature, filtered, and the filter cake was washed twice with isopropyl ether (5 mL. times.2) to give compounds 1 to 8(4.24g, yield: 90%) as white solids.
MS(ESI,pos.ion)m/z:315.2[M+1]+
Example 2
Synthetic route
Step 1: synthesis of Compounds 2-3
Benzaldehyde 2-1(95.6mL,943mmol), glycine ethyl ester hydrochloride 2-2(131g,943mmol) and Na2SO4(80g,565mmol) was added to 900 mL of TBME, then cooled to 0 deg.C, and triethylamine (197mL,1414mmol) was added slowly, after which time it was warmed to 25 deg.C and stirred at that temperature for 24 hours. The mixture was chromatographed over celite to remove solids, the organic solvent was removed from the filtrate under reduced pressure and the residue was stripped of solvent under high vacuum to give quantitative crude 2-3 which was used in the next reaction without further purification.
1H NMR(600MHz,CDCl3):8.28(s,1H),7.86–7.71(m,2H),7.50–7.32(m,3H),4.47–4.32(m,2H),4.29–4.18(m,2H),1.32–1.22(m,3H)ppm。
Step 2: synthesis of Compounds 2-4
Lithium tert-butoxide (17.60g,220mmol) was added to 250ml of toluene, and then a solution of the compound 2-3(22.00g,117mmol) in 50ml of toluene and a solution of trans-1, 4-dibromo-2-butene (20.00g,94mmol) in 50ml of toluene were simultaneously added dropwise at 0 ℃ while keeping the rates of addition of the two solutions the same and were completely added dropwise over about 1 hour. Then the temperature is raised to 30 ℃, the mixture is stirred for two hours, the reaction is quenched by 200ml of water, 200ml of TBME is used for back extraction, and organic phases are combined. Then 200ml of 1M hydrochloric acid solution are added toThe organic phase was stirred for two hours. The separated organic phase was extracted with 150ml of water, the aqueous phases were combined, sodium chloride (131.00g,2241mmol) and 200ml of TBME were added and the pH was adjusted to between 12 and 13 with 10M aqueous NaOH. The organic phase was separated, the aqueous phase was extracted with 100ml TBME, the organic phases were combined and Boc was added2O (21mL,98mmol), stirred at room temperature overnight, and warmed to 60 ℃ for two hours for completion of the reaction. After that, the mixture was cooled to room temperature, dried over anhydrous sodium sulfate, and the organic solvent was removed under reduced pressure. The resulting residue was purified by silica gel column chromatography eluting with petroleum ether and ethyl acetate (V: V) ═ 5:1, giving 2-4(12g, 50% yield) as a yellow oil.
1H NMR(400MHz,CDCl3):5.17–5.80(m,1H),5.29(s,1H),5.06–5.09(m,1H),4.12–4.19(m,2H),2.17–2.14(m,1H),1.77(s,1H),1.43(s,1H),1.41(s,9H),1.24(t,J=7.2Hz,3H)ppm。
And step 3: synthesis of Compounds 2-5
Alcalase 2.4L (52mL) of proteolytic enzyme was added to sodium phosphate buffer (0.1M,550mL, pH 8), then warmed to 39 ℃, and adjusted to pH 8.0 by the addition of 50% aqueous sodium hydroxide. At this temperature, a solution of compounds 2-4(10.58g, 42.5mmol) in 100mL DMSO was added slowly over a period of 20 minutes. Then, the temperature is raised to 40 ℃, the mixture is stirred for 24 hours, and a 50 percent sodium hydroxide aqueous solution is continuously added in the reaction process to ensure that the pH value of the reaction system is about 8.0. Thereafter, it was cooled to 30 ℃ and stirred for 48 hours, adjusted to pH 8.5 by addition of 50% aqueous sodium hydroxide solution, extracted twice with 150ml of TBME, the organic phases combined and the organic phase treated with 50ml of 5% NaHCO3The aqueous solution was washed three times, 50ml of water was washed three times, dried over anhydrous sodium sulfate, and the organic phase was removed under reduced pressure to obtain Compound 2-5(5.01g, yield: 47%) as a yellow solid.
MS(ESI,pos.ion)m/z:256.1[M+1]+
And 4, step 4: synthesis of Compounds 2-6
Compound 2-5(14.57g,57mmol) was dissolved in 100mL THF and 50mL methanol, cooled to 0 deg.C, then added with 25mL aqueous solution of lithium hydroxide (4.79g,114mmol), and after dropwise addition, warmed to 30 deg.C and stirred overnight. The organic solvent was removed under reduced pressure to give a solid, and 50ml of ethyl acetate and 50ml of water were added to dissolve the solid. The aqueous phase was adjusted to pH 4 with 1M hydrochloric acid solution, extracted three times with 50ml ethyl acetate, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 12g of crude product 2-6, which was directly subjected to the next reaction without further purification.
MS(ESI,pos.ion)m/z:228.1[M+1]+
And 5: synthesis of Compounds 2 to 8
Compound 2-6(1.00g,4.4mmol) was dissolved in 20mL of anhydrous tetrahydrofuran, CDI (0.93g,5.7mmol) was added, the mixture was refluxed for two hours at elevated temperature, then cooled to room temperature, compound 2-7(0.80g,6.6mmol) and DBU (1mL,6.6mmol) were added, and the reaction was stirred at room temperature for 16 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the resulting concentrate was diluted with ethyl acetate, washed with 1M aqueous hydrochloric acid solution and then with saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed from the filtrate under reduced pressure to obtain a crude product 2-8(0.94g, yield: 65%).
MS(ESI,pos.ion)m/z:331.1[M+1]+
Step 6: synthesis of Compounds 2-9
Dissolving the compound 2-8(2.10g,6.3mmol) in 40ml of isopropanol solution, adding p-toluenesulfonic acid (1.8g,9.4mmol), heating to 65 ℃, stirring for 4 hours, cooling to room temperature, removing the organic solvent under reduced pressure to obtain a white solid, washing the solid with ethyl acetate, filtering to obtain a solid which is the compound 2-9(1.49g, yield: 80%)
1H NMR(400MHz,CD3OD):7.73(d,J=8.0Hz,2H),7.26(d,J=7.9Hz,2H),5.81–5.65(m,1H),5.44(d,J=17.0Hz,1H),5.36(d,J=10.3Hz,1H),3.13–2.96(m,1H),2.47–2.32(m,4H),2.20(t,J=7.9Hz,1H),1.72(t,J=9.0Hz,1H),1.39–1.20(m,2H),1.20–1.02(m,2H)ppm。
Example 3:
the synthetic route is as follows:
step 1: synthesis of Compound 3-3
Compound 3-1(100g,460.36mmol) and compound 3-2(100g,564.72mmol) were dissolved in 1L of acetonitrile, cesium carbonate (90g,276.219mmol) and potassium carbonate (40g,289.427mmol) were then added, and the reaction mixture was refluxed at elevated temperature for 24 hours. Cooling to room temperature, filtering, removing the organic solvent from the filtrate under reduced pressure to obtain crude product 3-3, and directly carrying out the next reaction without further purification. MS (ESI, pos.ion) M/z 314.4[ M +1 ]]+
Step 2: synthesis of Compounds 3-5
After compound 3-3(150g,478.6mmol) was dissolved in ethanol (750mL), KOH (81g,1443.72mmol) in water (75mL) was slowly added dropwise thereto, and the reaction was refluxed at elevated temperature for 4 hours. After completion of the reaction, ethanol was removed under reduced pressure, 500mL of water was added, the pH was adjusted to about 4-5 with citric acid (103g,536.118mmol), and the resulting reaction mixture was refluxed at elevated temperature overnight. After the reaction was completed, the reaction solution was cooled to 0 ℃ and the pH was adjusted to about 2 to 3 with 1M HCl solution, a solid precipitated, filtered, and the filter cake was washed with a small amount of water and dried to obtain 3 to 5(70g, yield: 68.58%) as a yellow solid product.
MS(ESI,pos.ion)m/z:214.3[M+1]+
And step 3: synthesis of Compounds 3-6
Compound 3-5(70g,328.22mmol) was dissolved in H2To O (500mL), sodium hydroxide (13.2g,330mmol) and CoCl were added at room temperature2·6H2O (0.05g,0.3mmol) was dissolved with stirring. Then 0.1M aqueous NaOH solution was added dropwise to adjust the pH to 7.8, the temperature was raised to 38.5 ℃ and Acylase I deacetylase (350mg,0.5U/mg) was added and the reaction mixture was stirred at this temperature overnight. After the reaction was complete, the pH was adjusted to 7.6 with 1M hydrochloric acid solution, cooled to room temperature and filtered. The cake was washed with a mixed solution of 1L of water, 0.75L of water and methanol (V: V ═ 10:1) in this order, and the cake was dried at 55 ℃ under vacuum to give compound 3-6(26g, yield: 46.2%) as a white solid.
And 4, step 4: synthesis of Compounds 3-7
Compounds 3-6(250g,1.46mol) were dissolved in THF (1500mL) and H2To a mixed solvent of O (1500mL), NaOH (76g,1.9mol) was added under ice-cooling, and then Boc was added dropwise to the reaction solution2O (440mL,1900mmol), at room temperature overnight. After completion of the reaction, THF was distilled off under reduced pressure and the aqueous phase was saturated with NaHSO4The solution was adjusted to pH 2-3, then extracted with ethyl acetate (1000mL × 3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was spin-dried under reduced pressure to give a red oily liquid, then MeCN (2500mL) was added to dilute, dicyclohexylamine (340mL,1710mmol) was added dropwise, the reaction mixture was warmed to 50 ℃ to react for 30 minutes, then cooled to room temperature, and stirred for 4 hours, acetonitrile was spin-dried under reduced pressure to give a white solid, 1L petroleum ether was added to pulp, filtered, and the filter cake was vacuum-dried at 50 ℃ to give compound 3-7(500g, yield: 96.70%).
MS(ESI,pos.ion)m/z:172.3[M+1-100]+
Example 4:
the synthetic route is as follows:
step 1 Synthesis of Compound 4-2
Compound 1-8(0.30g, 1mmol), compound 4-1(0.23g, 0.94mmol), diphenyl-2-pyridylphosphine (DPPPy, 0.4g,2mmol) and di-tert-butyl azodicarboxylate (DBAD, 0.3g,2mmol) were dissolved in 15mL THF, and the reaction was stirred at room temperature under nitrogen for 24 hours. After the reaction, the solvent was dried under reduced pressure, 30mL of ethyl acetate was added, the mixture was washed with 20mL of 1M HCl solution, and the resulting organic phase was separated and washed with anhydrous Na2SO4Drying, filtering, and removing the organic solvent from the filtrate under reduced pressure. The resulting mixture was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 5:1 as eluent, to give 4-2(0.4g, yield: 80%) as a pale yellow oily product
MS(ESI,pos.ion)m/z:541.8[M+1]+
Step 2 Synthesis of Compound 4-3
Compound 4-2(0.7g, 1mmol) was dissolved in methanol (10mL), and then a 5mL aqueous solution of LiOH (0.06g,3mmol) was slowly added, followed by stirring at room temperature for 3 hours. After completion of the reaction, the organic solvent was removed under reduced pressure, and then acidified to pH 3 with 1M HCl solution, extracted twice with 20mL of ethyl acetate, the organic phases were combined, washed with 20mL of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure to obtain compound 4-3(0.7g, yield: 100%) as a white solid.
MS(ESI,pos.ion)m/z:528.3[M+1]+
Step 3 Synthesis of Compound 4-4
Compound 4-3(0.7g,1mmol), compound 2-9(0.6g,1mmol), EDCI (0.3g,2mmol) and HOAT (0.2g,1mmol) were added to a round bottom flask, 30mL of dichloromethane were added under nitrogen, then cooled to 0 deg.C and DIPEA (0.7mL,3mmol) was added slowly and the reaction mixture was warmed to 30 deg.C and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 20ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give purified compounds 4 to 4(0.54g, yield: 60%) as white solids.
MS(ESI,pos.ion)m/z:739[M+1]+
Step 4 Synthesis of Compounds 4-5
Compound 4-4(0.54g,0.73mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 20ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until the reaction was complete without gas evolution. Filtration and washing of the resulting white solid with 20ml of ethyl acetate. The resulting solid hydrochloride compound and compounds 3-7(0.32g,0.71mmol), EDCI (0.15g,0.78mmol), HOAT (0.1g,0.7mmol) were charged to a round bottom flask, 20mL of dichloromethane were added under nitrogen, then cooled to 0 deg.C and DIPEA (0.3mL,2mmol) was added. The temperature was raised to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, and then extracted twice with 10ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether ethyl acetate (V: V) ═ 2:1 as eluent, to obtain 4 to 5(0.32g, yield 61%) as a white solid after purification.
Step 5 Synthesis of Compounds 4-6
Compound 4-5(0.32g,0.35mmol) was dissolved in 300 ml of 1, 2-dichloroethane, 0.05g of Janz 1B catalyst was added under nitrogen, the temperature was then raised to 65 ℃ and the reaction mixture was stirred at this temperature for 48 hours. Cooled to room temperature, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give 4 to 6(0.18g, yield: 58%) as a white solid.
MS(ESI,pos.ion)m/z:865[M+1]+
Step 6 Synthesis of Compounds 4-9
Compound 4-6(0.18g,0.2mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 20ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until no gas was evolved. Filtration and washing of the resulting white solid with 20ml ethyl acetate afforded solid hydrochloride compound 4-7. The resulting solid hydrochloride salt compound 4-7 and compound 4-8(0.02g,0.16mmol), EDCI (0.04g,0.21mmol), HOAT (0.03g,0.22mmol) were charged to a round bottom flask, 20mL of dichloromethane were added under nitrogen, then cooled to 0 deg.C and DIPEA (0.1mL,0.6mmol) was added slowly. After the addition was complete, the reaction mixture was warmed to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether ethyl acetate (V: V) ═ 2:1 as eluent, to obtain 4 to 9(0.03g, yield 23%) as a purified white solid.
MS(ESI,pos.ion)m/z:874[M+1]+
1H NMR(400MHz,CDCl3):10.22(s,1H),7.88(d,J=9.2Hz,1H),7.77(s,1H),7.71(d,J=7.2Hz,1H),7.56(s,1H),7.10–6.95(m,3H),5.74(d,J=9.2Hz,1H),5.58(s,1H),5.04–4.97(m,1H),4.72–4.66(m,2H),4.63–4.55(m,1H),4.11–4.06(m,1H),3.93(s,3H),3.79(s,3H),3.26–3.15(m,1H),2.96–2.89(m,1H),2.67(s,3H),2.37–2.18(m,2H),1.98–1.87(m,3H),1.53–1.39(m,16H),0.93–0.87(m,4H)ppm。
Example 5:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.26g,0.30mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until no gas was evolved. Filtering, washing the obtained white solid with 20ml ethyl acetate to obtain a solid compound 4-7. The resulting solid hydrochloride compound 4-7 was added to a round-bottomed flask along with 5-methyl-1H-pyrazole-3-carboxylic acid (0.05g,0.4mmol), EDCI (0.13g,2.8mmol), and HOAT (0.1g,3.0mmol), followed by addition of 20mL of dichloromethane under nitrogen, cooling to 0 deg.C, addition of DIPEA (0.5mL,10mmol), and stirring of the reaction mixture at 30 deg.C for 6 hours. The reaction was quenched with 10ml of water, extracted twice with 20ml of dichloromethane, the organic phases were combined, washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 5-2(0.135g, yield 51.9%) as a white solid after purification.
MS(ESI,pos.ion)m/z:873.3[M+1]+
1H NMR(600MHz,CDCl3):7.99(s,1H),7.89(t,J=7.3Hz,1H),7.50(s,1H),7.35(dd,J=10.7,4.3Hz,2H),7.04(d,J=4.4Hz,1H),7.00(d,J=9.2Hz,1H),6.65(d,J=2.2Hz,1H),5.69(dd,J=18.2,8.5Hz,1H),5.49(s,1H),4.98–4.94(m,1H),4.64(t,J=7.4Hz,2H),4.48(d,J=11.4Hz,1H),4.16(dd,J=11.3,4.4Hz,1H),3.90(d,J=6.8Hz,3H),3.77(s,3H),3.23(dt,J=13.6,6.8Hz,1H),2.87(dq,J=8.1,4.9Hz,1H),2.69(dd,J=13.7,7.8Hz,1H),2.62(s,3H),2.58–2.52(m,2H),2.25(dd,J=17.4,8.7Hz,1H),2.04(s,1H),1.93–1.87(m,1H),1.82(dd,J=8.0,5.9Hz,1H),1.74–1.69(m,1H),1.65(dd,J=9.3,5.8Hz,1H),1.52–1.44(m,5H),1.35–1.25(m,6H),1.10(dddd,J=24.1,15.7,7.6,5.3Hz,3H),0.93–0.84(m,2H)ppm。
Example 6:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.21g,0.24mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until no gas was evolved. Filtering, washing the obtained white solid with 20ml ethyl acetate to obtain a solid compound 4-7. The obtained solid hydrochloride compound 4-7 and 5-methylisoxazole-3-carboxylic acid (0.05g,0.4mmol), EDCI (0.13g,2.8mmol), HOAT (0.1g,3.0mmol) were charged in a round-bottomed flask, 20mL of dichloromethane was added under nitrogen protection, then cooled to 0 ℃ and DIPEA (0.5mL,10mmol) was slowly added, after the addition, the temperature was raised to 30 ℃ and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 20ml of dichloromethane, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to obtain compound 6-1 as a white solid (0.203g, yield 88.6%).
MS(ESI,pos.ion)m/z:874.3[M+1]+
1H NMR(600MHz,CDCl3):10.44(s,1H),8.09(d,J=7.8Hz,1H),8.00(s,1H),7.84(d,J=9.1Hz,1H),7.57(s,1H),7.03(d,J=9.1Hz,2H),6.13(d,J=16.7Hz,1H),5.64(dd,J=17.9,8.8Hz,1H),5.54(s,1H),4.95–4.87(m,1H),4.74(ddd,J=23.5,13.1,5.3Hz,2H),4.63(d,J=11.5Hz,1H),4.12(dd,J=11.5,3.7Hz,1H),3.87(s,3H),3.21(dq,J=13.6,6.8Hz,1H),2.90–2.85(m,1H),2.75(dd,J=13.8,7.7Hz,1H),2.66(s,3H),2.61–2.54(m,1H),2.36(s,3H),2.24(q,J=8.5Hz,1H),2.09(dd,J=23.2,11.2Hz,1H),1.85–1.80(m,1H),1.77(dd,J=7.6,6.2Hz,1H),1.65–1.58(m,1H),1.48–1.38(m,12H),1.28–1.22(m,3H),1.14–1.09(m,1H),1.08–1.03(m,1H),0.89(dd,J=10.5,5.5Hz,1H)ppm。
Example 7:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.21g,0.24mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 6ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until no gas was evolved. The white solid obtained is filtered, washed once with 20ml of ethyl acetate and then dried in vacuum to obtain the solid compound 4-7. The solid compound 4-7 obtained by drying, compound 7-1(0.05g,0.4mmol), EDCI (0.13g,2.8mmol) and HOAT (0.1g,3.0mmol) were charged into a round-bottomed flask, 20mL of dichloromethane was added under nitrogen protection, the mixture was cooled to 0 ℃ and DIPEA (0.5mL,10mmol) was slowly added thereto, after completion of the dropwise addition, the mixture was heated to 30 ℃ and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 20ml of dichloromethane, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the filtrate was freed of the organic solvent under reduced pressure, and the resulting residue was purified by column chromatography on silica gel with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 7-2 as a white solid (0.203g, yield 88.6%).
MS(ESI,pos.ion)m/z:902.3[M+1]+
1H NMR(600MHz,CDCl3):10.44(s,1H),8.08(d,J=7.4Hz,1H),8.01(s,1H),7.86–7.82(m,2H),7.52(s,1H),7.06–7.03(m,2H),5.69(dd,J=17.9,8.8Hz,1H),5.49(s,1H),5.06(dt,J=13.1,6.5Hz,1H),4.98–4.94(m,1H),4.74(t,J=8.3Hz,2H),4.67–4.63(m,1H),4.13(dd,J=11.7,2.7Hz,1H),3.89(s,3H),3.22(dd,J=13.4,6.3Hz,1H),2.89(td,J=8.0,4.1Hz,1H),2.73(dd,J=13.8,7.4Hz,1H),2.67(s,3H),2.31(q,J=8.5Hz,1H),2.07(d,J=5.9Hz,1H),1.85(ddd,J=13.9,8.8,5.3Hz,2H),1.66(dd,J=16.5,7.2Hz,1H),1.55–1.40(m,10H),1.29(dd,J=18.7,15.0Hz,9H),1.15–1.04(m,4H),0.92–0.87(m,2H)ppm。
Example 8:
the synthetic route is as follows:
step 1 Synthesis of Compound 8-2
Ethyl acetate (10g,113.5mmol) and hydrazine hydrate (10mL,257.3mmol,25.73mol/L) were dissolved in ethanol (15mL), heated to reflux and reacted for 4 hours. After completion of the reaction, it was cooled to room temperature, and the solvent was distilled off under reduced pressure to give the product 8-2(8.2g, yield: 98%) as a white solid.
MS(ESI,pos.ion)m/z:75[M+1]+
Step 2 Synthesis of Compound 8-4
Compound 8-2(1.0g,13mmol) was dissolved in dichloromethane (20mL), triethylamine (2mL,14.3mmol) was added under ice-bath, then compound 8-3(1.8g,13mmol) was added slowly, after the addition was complete, the mixture was warmed to room temperature and stirred overnight. After completion of the reaction, 100mL of ethyl acetate was added to the reaction mixture to dilute the mixture, the resulting salt was removed by filtration, the obtained filtrate was dried under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 5:1, to obtain 8-4(2.2g,13mmol, yield: 94%) as a white solid after purification
MS(ESI,neg.ion)m/z:173[M-1]-
Step 3 Synthesis of Compounds 8-5
Compound 8-4(2.2g,13mmol) was dissolved in dichloromethane (45mL), triethylamine (2.5mL,18mmol) and p-toluenesulfonyl chloride (3.0g,16mmol) were added under ice-bath, and the mixture was warmed to room temperature and stirred for 8 hours. After completion of the reaction, 50mL of water was added to quench the reaction, and the reaction mixture was filtered, the obtained organic phase was dried over anhydrous sodium sulfate, filtered, and the organic solvent was evaporated under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1, to obtain compound 8-5(1.1g, yield: 56%) as a white solid after purification.
MS(ESI,pos.ion)m/z:157[M+1]+
Step 4 Synthesis of Compounds 8-6
Compound 8-5(1.1g,7.0mmol) was dissolved in ether (25mL), potassium trimethylsilanolate (0.9g,7mmol) was added with stirring, a white solid was produced during the reaction, and stirred at room temperature overnight. After completion of the reaction, filtration was carried out, the filter cake was washed with a small amount of ethanol, and the resulting solid was dried under vacuum to obtain the product 8-6(0.85g, yield: 73%) as a white solid.
Step 5 Synthesis of Compounds 8-7
Compounds 4-7(0.15g,0.19mmol), compounds 8-6(0.04g,0.23mmol), EDCI (0.04g,0.21mmol) and HOAT (0.03g,0.22mmol) were added to a round bottom flask, to which was added 20mL of dichloromethane under nitrogen, then cooled to 0 deg.C and DIPEA (0.1mL,0.6mmol) was added slowly. The temperature was raised to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compounds 8 to 7(0.05g, yield 30%) as white solids.
MS(ESI,pos.ion)m/z:874[M+1]+
1H NMR(600MHz,CDCl3):10.37(s,1H),8.59(s,1H),7.98–7.85(m,2H),7.62–7.54(m,1H),7.15–7.03(m,2H),5.76(dd,J=17.6,9.0Hz,1H),5.59(s,1H),5.07–4.98(m,1H),4.84(t,J=8.0Hz,1H),4.71(t,J=7.6Hz,1H),4.64(d,J=11.5Hz,1H),4.20(d,J=8.8Hz,1H),3.91(s,3H),3.30–3.18(m,1H),2.99–2.88(m,1H),2.79(s,2H),2.70(s,4H),2.53(s,3H),2.33–2.28(m,1H),2.24–2.17(m,1H),1.79–1.68(m,3H),1.42(d,J=6.9Hz,6H),1.33–1.27(m,7H),0.98–0.85(m,4H)ppm。
Example 9:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.2g,0.3mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added, warmed to room temperature and stirred until no gas was evolved. Filtering, washing the obtained white solid with 20ml ethyl acetate to obtain a solid compound 4-7. The solid compound 4-7, compound 9-1(0.06g,0.4mmol), EDCI (0.13g,0.66mmol) and HOAT (0.1g,0.7mmol) were charged into a round-bottomed flask, 10mL of dichloromethane were added under nitrogen protection, then cooled to 0 ℃ and DIPEA (0.2mL,1mmol) was added, after the addition, the temperature was raised to 30 ℃ and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, the resulting mixture was taken twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the resulting residue was purified by column chromatography on silica gel using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 9-2(0.180g, yield 80%) as a white solid.
MS(ESI,pos.ion)m/z:885.3[M+1]+
1H NMR(600MHz,CDCl3):10.38(s,1H),9.07(d,J=0.8Hz,1H),8.44(s,1H),8.15(d,J=6.7Hz,1H),7.85(d,J=9.1Hz,2H),7.47(s,1H),7.04(s,1H),6.95(d,J=9.2Hz,1H),5.73(dd,J=18.2,8.7Hz,1H),5.50(s,1H),5.02–4.97(m,1H),4.69–4.64(m,2H),4.49(d,J=11.5Hz,1H),4.10(dd,J=11.6,3.9Hz,1H),3.77(s,3H),3.22(dt,J=13.7,6.8Hz,1H),2.92–2.86(m,1H),2.71(dd,J=13.8,7.7Hz,1H),2.65(s,3H),2.60(s,4H),2.29–2.25(m,1H),2.04–2.00(m,1H),1.91(dd,J=7.8,6.1Hz,2H),1.78–1.71(m,1H),1.66(dd,J=9.4,5.9Hz,1H),1.53–1.45(m,5H),1.39(t,J=13.5Hz,7H),1.16–1.07(m,3H),0.94–0.86(m,2H)ppm。
Example 10:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.2g,0.3mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added, warmed to room temperature and stirred until no gas was evolved. Filtration and washing of the resulting white solid with 20ml of ethyl acetate. The resulting solid 4-7, compound 10-1(0.1g,0.6mmol), EDCI (0.13g,0.66mmol) and HOAT (0.1g,0.7mmol) were charged into a round-bottomed flask, 10mL of dichloromethane were added under nitrogen, then cooled to 0 deg.C, DIPEA (0.2mL,1mmol) was added, after the addition, the temperature was raised to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, taken twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether ethyl acetate (V: V) ═ 2:1 as eluent, to obtain compound 10-2(0.166g, yield 70%) as a purified white solid.
MS(ESI,pos.ion)m/z:901.3[M+1]+
1H NMR(600MHz,CDCl3):10.37(s,1H),7.98(d,J=9.1Hz,1H),7.54(s,1H),7.41(d,J=2.3Hz,1H),7.37(d,J=7.3Hz,1H),7.10(t,J=6.8Hz,1H),7.04(s,1H),6.68(d,J=2.3Hz,1H),5.71(dd,J=18.3,8.5Hz,1H),5.53(s,1H),4.98–4.94(m,1H),4.75(ddd,J=10.5,7.4,3.1Hz,1H),4.65(d,J=11.4Hz,1H),4.60(t,J=7.8Hz,1H),4.49(dd,J=13.4,6.7Hz,1H),4.19–4.11(m,3H),3.89(s,3H),3.23(dt,J=13.8,6.8Hz,1H),2.92–2.87(m,1H),2.66(d,J=10.1Hz,4H),2.61–2.55(m,2H),2.33(q,J=8.7Hz,1H),2.06(s,3H),2.04–1.90(m,4H),1.86(dd,J=8.0,6.0Hz,1H),1.75–1.69(m,1H),1.50(dt,J=10.2,5.1Hz,9H),1.41(s,3H),1.19–1.05(m,3H),0.95–0.86(m,2H)ppm。
Example 11:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.16g,0.2mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added, warmed to room temperature and stirred until the reaction was complete without gas evolution. Filtering, washing the obtained white solid with 20ml ethyl acetate to obtain a solid compound 4-7. The resulting solid compound 4-7, compound 11-1(0.1g,0.6mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were charged into a round-bottomed flask, 10mL of dichloromethane were added under nitrogen, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added, after the addition, the temperature was raised to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, the resulting mixture was extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the resulting residue was purified by column chromatography on silica gel using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 11-2 as a white solid (0.09g, 50% yield).
MS(ESI,pos.ion)m/z:902.3[M+1]+
1H NMR(600MHz,CDCl3):10.39(s,1H),8.05(d,J=7.4Hz,1H),7.89(d,J=9.1Hz,1H),7.76(s,1H),7.57(s,1H),7.09–7.04(m,2H),6.16(s,1H),5.71(dd,J=17.7,9.0Hz,1H),5.57(s,1H),4.99–4.94(m,1H),4.81(dd,J=13.3,5.3Hz,1H),4.72–4.67(m,2H),4.18–4.14(m,1H),3.91(s,3H),3.27–3.20(m,1H),3.02(dt,J=13.8,6.9Hz,1H),2.91(ddd,J=12.9,8.2,4.9Hz,1H),2.79–2.73(m,2H),2.70(s,3H),2.65(d,J=4.8Hz,1H),2.29(dd,J=17.0,8.4Hz,1H),2.14(dd,J=22.8,10.8Hz,1H),2.04(d,J=23.2Hz,1H),1.85(dd,J=14.1,7.9Hz,2H),1.71–1.65(m,1H),1.49(ddd,J=17.2,9.9,5.8Hz,5H),1.42(t,J=7.0Hz,8H),1.30(s,2H),1.13(ddt,J=21.0,15.4,6.6Hz,3H),0.92(dt,J=13.9,7.6Hz,2H)ppm。
Example 12:
the synthetic route is as follows:
the synthesis steps are as follows:
compound 4-6(0.3g,0.4mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added, warmed to room temperature and stirred until the reaction was complete without gas evolution. Filtering, washing the obtained white solid with 20ml ethyl acetate to obtain a solid compound 4-7. The solid compound 4-7, compound 12-1(0.06g,0.4mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were charged into a round-bottomed flask, 20mL of dichloromethane were added under nitrogen protection, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added, after the addition, the temperature was raised to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 20ml of water, the resulting mixture was extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the resulting residue was purified by column chromatography on silica gel using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 12-2(0.2g, yield 60%) as a white solid after purification.
MS(ESI,pos.ion)m/z:889.3[M+1]+
1H NMR(600MHz,CDCl3):10.49(s,1H),9.09(s,3H),8.37(s,1H),8.17(s,1H),7.44(s,1H),7.40(s,1H),7.26(d,J=9.0Hz,1H),7.21(s,1H),6.97(s,1H),6.65(s,1H),5.67–5.53(m,2H),4.94–4.69(m,3H),4.45(s,1H),4.04(d,J=6.8Hz,1H),3.92(d,J=38.0Hz,3H),3.31(dt,J=13.6,6.8Hz,1H),2.81(d,J=29.5Hz,2H),2.57(s,2H),2.45(d,J=10.5Hz,6H),2.18(s,1H),1.91(s,1H),1.76(s,1H),1.64(d,J=39.2Hz,3H),1.44(dd,J=6.7,2.4Hz,7H),1.29–1.21(m,3H),1.14(s,1H),1.04–0.85(m,3H)ppm。
Example 13:
the synthetic route is as follows:
step 1 Synthesis of Compound 13-1
Compound 4-6(0.3g,0.4mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until the reaction was complete without gas evolution. Filtering, washing the obtained white solid with 20ml ethyl acetate to obtain a solid compound 4-7. The resulting solid compound 4-7, compound 1-4(0.05g,0.3mmol), EDCI (0.05g,0.3mmol) and HOAT (0.040g,0.29mmol) were dissolved in DMF (20mL), DIPEA (0.1mL,0.6mmol) was added slowly over an ice bath, and the reaction was stirred at this temperature for 0.5 h, then warmed to room temperature and reacted overnight. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 13-1(164mg, yield: 60%) as a white solid.
MS(ESI,pos.ion)m/z:918.4[M+1]+
1H NMR(600MHz,CDCl3):10.32(s,1H),8.16(d,J=6Hz,1H),7.66(s,1H),7.25(d,J=12Hz,2H),7.12(s,1H),5.75(dd,J1=18Hz,J2=12Hz,1H),5.68(s,1H),5.32(s,1H),5.05(t,J1=12Hz,J2=6Hz,1H),4.73(d,J=6Hz,1H),4.69(d,J=6Hz,2H),4.33(t,J1=6Hz,J2=6Hz,1H),4.16–4.15(m,1H),3.98(s,3H),3.35–3.31(m,1H),3.14–3.08(m,1H),2.94–2.86(m,1H),2.80-2.73(m,2H),2.65(s,2H),2.37–2.33(m,1H),2.10–2.04(m,1H),1.94–1.93(m,2H),1.79–1.72(m,2H),1.57–1.47(m,3H),1.44(d,J=6Hz,6H),1.33(d,J=6Hz,6H),1.20–1.13(m,1H),1.08–1.06(m,1H),1.00–0.86(m,6H)ppm。
Example 14:
the synthetic route is as follows:
step 1 Synthesis of Compound 14-3
Na (2.63g, 0.11mmol) was added to EtOH (65mL), heated to reflux under nitrogen, then cooled to 0 deg.C, compound 14-1(10g,116.1mmol) and compound 14-2(16.98g,116.2mmol) were added, then reacted at room temperature for 1 hour, warmed to 80 deg.C for 0.75 hour, and cooled to room temperature after completion of the reaction. The pH was adjusted to 2 with 1M hydrochloric acid solution, then 100mL of water was added for dilution, ether was used for extraction (30 mL. times.3), the organic phases were combined, the resulting organic phase was washed with a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was spun dry under reduced pressure to give 14-3 as an oil (19.4g, yield: 89.7%).
MS(ESI,pos.ion)m/z:187.1[M+1]+
1H NMR(600MHz,CDCl3):6.31–6.30(m,1H),4.26–4.23(m,2H),2.58–2.55(m,2H),1.28–1.26(m,3H),1.09–1.08(m,6H)ppm。
Step 2 Synthesis of Compound 14-4
Compound 14-3(1g,5.37mmol) and hydrazine hydrate (10.7mmol,9.64mol/L) were added to ethanol (20mL), and the temperature was raised to 75 ℃ under nitrogen protection for 1 hour. After completion of the reaction, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 14-4(0.55g, yield: 56%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:183.1[M+1]+
1HNMR(600MHz,CDCl3):11.37(b,1H),6.63(s,1H),4.40–4.37(m,2H),3.07–3.05(m,1H),1.43–1.40(m,3H),1.39–1.30(m,6H)ppm。
Step 3 Synthesis of Compound 14-5
Compound 14-4(2.2g,12mmol) was added to a mixed solvent of ethanol (10mL) and water (5mL), NaOH (1.9g,48mmol) was further added, and the reaction was carried out at room temperature for 3 hours. After completion of the reaction, the pH of the mixture was adjusted to about 2 with 1M hydrochloric acid solution, ethanol was removed under reduced pressure, 50mL of water was added, followed by extraction with ethyl acetate (50 mL. times.3), the organic phases were combined, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the resulting filtrate was concentrated under reduced pressure to give compound 14-5(1.64g, yield: 88%) as a white solid.
Step 4 Synthesis of Compounds 14-6
Compound 4-6(0.3g,0.4mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 2ml of a 30% strength ethyl acetate solution of hydrogen chloride was added, warmed to room temperature and stirred until the reaction was complete without gas evolution. And filtering, and washing the obtained white solid with 20ml of ethyl acetate to obtain a white solid compound 4-7. The resulting white solid, compound 4-7, compound 14-5(0.04g,0.3mmol), EDCI (0.05g,0.3mmol) and HOAT (0.040g,0.29mmol) were dissolved in DMF (5mL), cooled to 0 ℃ under nitrogen, DIPEA (0.1mL,0.6mmol) was added, stirred at 0 ℃ for 0.5 h, then warmed to room temperature and reacted overnight. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give 14-6(150mg,0.17mmol) as a purified white solid.
MS(ESI,pos.ion)m/z:901.4[M+1]+
1H NMR(600MHz,CDCl3):10.73(s,1H),8.92(s,1H),7.90(s,1H),7.49(s,1H),7.31(s,1H),6.95(d,J=6Hz,1H),6.42(s,1H),5.81(d,J=6Hz,1H),5.60(s,1H),5.12-5.09(m,1H),4.69–4.65(m,2H),4.52(s,1H),4.20–4.12(m,1H),4.04(d,J=12Hz,1H),3.68(s,3H),3.38–3.32(m,1H),3.04–3.03(m,1H),2.68–2.63(m,1H),2.52(s,1H),2.40(s,3H),2.25–2.21(m,1H),2.07(s,1H),2.00(s,1H),1.91(s,1H),1.74–1.71(m,1H),1.59–1.56(m,1H),1.47–1.43(m,11H),1.30–1.26(m,7H),1.22–1.08(m,4H),1.02–0.97(m,1H),0.93–0.87(m,1H)ppm。
Example 15:
the synthetic route is as follows:
step 1 Synthesis of Compound 15-3
Mixing compound 15-1(1.5g,11mmol), compound 15-2(1.91mL) and K2CO3(2.46g, 17.8mmol) was added to acetonitrile (15mL) and stirred at room temperature under nitrogen for 4 days. After completion of the reaction, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give 15-3(70mg, yield: 3.6%) MS (ESI, pos.ion) M/z:184.3[ M +1]+
Step 2 Synthesis of Compound 15-4
Compound 15-3(0.2g,1mmol) was added to EtOH (4mL) and H2O (2mL), and then LiOH & H2O (84mg,2mmol) at room temperature for 12 hours after the reaction, ethanol was removed under reduced pressure, 10mL of water was added, the pH of the mixture was adjusted to about 5 with 2M aqueous HCl, and then dichloromethane (30mL × 3) was used for extraction, the organic phases were combined, the resulting organic phase was washed with saturated sodium chloride solution, and anhydrous Na was added2SO4Drying and filtration were carried out, and the obtained filtrate was freed of the organic solvent under reduced pressure to give compound 15-4(0.1g, yield: 60%) as a white solid.
MS(ESI,pos.ion)m/z:156.2[M+1]+
Step 3 Synthesis of Compound 15-5
Compound 4-7(0.2g,0.25mmol), compound 15-4(0.05g,0.3mmol), EDCI (0.05g,0.3mmol) and HOAT (0.040g,0.29mmol) were dissolved in DMF (5mL), cooled to 0 deg.C, DIPEA (97mg,0.75mmol) was added slowly, after dropwise addition, the mixture was warmed to room temperature and reacted overnight. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give 15-5(112mg, yield: 50%) as a white solid.
MS(ESI,pos.ion)m/z:901.6[M+1]+
1H NMR(600MHz,CDCl3):10.30(s,1H),7.98–7.97(m,1H),7.87(d,J=12Hz,1H),7.07–7.05(m,2H),6.85–6.76(m,1H),5.77–5.70(m,1H),5.59–5.52(m,1H),5.32(s,2H),5.13–5.06(m,1H),5.02–4.98(m,1H),4.79–4.63(m,4H),3.93(s,3H),3.27–3.20(m,1H),2.96–2.90(m,1H),2.78–2.73(m,1H),2.70(s,3H),1.94–1.89(m,2H),1.70(d,J=6Hz,6H),1.33–1.24(m,9H),1.19–1.09(m,3H),0.90–0.84(m,6H)ppm。
Example 16:
the synthetic route is as follows:
step 1: synthesis of Compound 16-2
Compound 16-1(1g,7.09mmol) and 2-iodopropane (2.4g, 14.2mmol) were added to acetonitrile (15mL) followed by K2CO3(1.955g,14.17mmol), warmed to 80 ℃ under nitrogen and stirred overnight. After the reaction is finishedThen, the mixture was cooled to room temperature, and the organic solvent was spin-dried under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether and ethyl acetate (V: V) ═ 5:1 as an eluent, to give 16-2(0.4g, yield: 30%) as a colorless oily compound.
MS(ESI,pos.ion)m/z:184.1[M+1]+
Step 2 Synthesis of Compound 16-3
Compound 16-2(0.4g,2mmol) was added to EtOH (4mL) and H2O (2mL), and then LiOH & H2O (168mg,4mmol), reaction at room temperature for 12 hours, after completion of the reaction, removing EtOH under reduced pressure, diluting with 10mL of water, adjusting the pH to about 5 with 2M hydrochloric acid solution, extracting with ethyl acetate (10mL × 3) three times, combining the organic phases, washing the organic phases with saturated sodium chloride solution, and removing anhydrous Na2SO4Drying, filtration and concentration of the filtrate under reduced pressure gave compound 16-3 as a white solid (0.29g, yield: 90%).
MS(ESI,pos.ion)m/z:156.2[M+1]+
Step 4 Synthesis of Compound 16-4
Compound 4-6(0.3g,0.4mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 4ml of a 30% strength ethyl acetate solution of hydrogen chloride was added, warmed to room temperature and stirred until the reaction was complete without gas evolution. Filtration and washing of the resulting white solid with 20ml of ethyl acetate. The resulting white solid compounds 4 to 7 and 16 to 3(0.05g,0.3mmol), EDCI (0.05g,0.3mmol) and HOAT (0.040g,0.29mmol) were dissolved in DMF (5mL), DIPEA (97mg,0.75mmol) was added slowly at 0 ℃ and after completion of the dropwise addition, the mixture was warmed to room temperature and reacted overnight. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 16-4(162mg, yield: 60%) as a white solid.
MS(ESI,pos.ion)m/z:901.6[M+1]+
1H NMR(600MHz,CDCl3):10.33(s,1H),7.96(d,J=6Hz,1H),7.92(s,1H),7.24(d,J=12Hz,1H),7.17(s,1H),7.09(d,J=12Hz,1H),7.03(s,1H),5.72–5.68(m,1H),5.52(m,1H),4.97(t,J=6Hz,J=12Hz,1H),4.86–4.81(m,1H),4.80–4.76(m,1H),4.60(t,J=6Hz,J=12Hz,2H),3.91(s,3H),3.24–3.18(m,1H),2.92–2.84(m,1H),2.73–2.69(m,1H),2.67(s,3H),2.63–2.56(m,2H),2.37–2.33(m,1H),2.07–2.01(m,2H),1.74–1.68(m,5H),1.58(d,J=6Hz,6H),1.50–1.48(m,2H),1.47-1.42(m,4H),1.40–1.39(m,6H),0.96(m,4H)ppm。
Example 17:
the synthetic route is as follows:
step 1 Synthesis of Compound 17-1
The compound 2-iodopropane (3g,17.648mmol) and the compound sodium azide (4.589g,70.59mmol) were added to DMF (20mL) and reacted at room temperature for 48 hours. After the reaction was completed, filtration was carried out to obtain compound 17-2(0.2g,2mmol) and CH3CN (20mL) was added to the filtrate, and CuSO was added thereto with stirring at room temperature4·5H2O (0.05g,0.2mmol), Cu (0.03g,0.5mmol), nitrogen protection, and room temperature reaction for 72 hours. After the reaction, 10mL of water was added to quench the reaction, 30mL of ethyl acetate was added to dilute the reaction solution, and saturated NH was used4Washing with Cl (30mL) solution, extracting with ethyl acetate (30mL × 2), combining the organic phases, washing the organic phase with saturated sodium chloride, drying over anhydrous sodium sulfate, filtering, and removing the organic phase under reduced pressureThe solvent and the residue were purified by silica gel column chromatography using petroleum ether and ethyl acetate (V: V) ═ 6:1 as eluent, to give compound 17-3(2g, yield: 85.63%) as a white solid.
MS(ESI,pos.ion)m/z:184.3[M+1]+
1H NMR(600MHz,CDCl3):8.08(s,1H),4.87–4.80(m,1H),4.39–4.31(m,2H),1.57–1.53(m,6H),1.35–1.30(m,3H)ppm。
Step 3 Synthesis of Compound 17-4
Compound 17-3(1.4g,7.6mmol) was added to EtOH (10mL) and H2O (5mL), NaOH (1.2g, 31mmol) was added, and the mixture was stirred at room temperature overnight after completion of the reaction, ethanol was removed under reduced pressure, the pH was adjusted to about 5 with 2M HCl solution, extraction was performed with methylene chloride (30mL × 3), the organic phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, and filtered, and the resulting filtrate was dried under reduced pressure to obtain compound 17-4 as a white solid (0.7g, yield: 80%).
MS(ESI,neg.ion)m/z:154.1[M-1]-
1H NMR(600MHz,CDCl3):8.47(s,1H),8.18(s,1H),4.83–4.75(m,1H),1.47(d,J=12Hz,6H)ppm。
Step 4 Synthesis of Compound 17-5
Compound 4-6(0.3g,0.4mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 4ml of 30% strength ethyl acetate hydrochloride solution was added and stirred at room temperature until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 20ml of ethyl acetate. The resulting white solid, Compound 4-7, Compound 17-4(0.04g,0.3mmol), EDCI (0.05g,0.3mmol) and HOAT (0.040g,0.29mmol) were dissolved in DMF (20mL), DIPEA (97mg,0.75mmol) was added slowly at 0 ℃ and after the addition was complete, the temperature was raised to room temperature and the reaction was allowed to proceed overnight. After completion of the reaction, the reaction was quenched with 10ml of water, taken twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1, to obtain 17-5(135mg, yield: 50%)
MS(ESI,pos.ion)m/z:901.6[M+1]+
1H NMR(600MHz,CDCl3):10.41(s,1H),8.04(d,J=12Hz,1H),7.97(s,1H),7.86(s,1H),7.68(s,1H),7.31(s,1H),7.25(d,J=12Hz,1H),6.42(s,1H),5.76–5.69(m,1H),5.05(t,J1=6Hz,J2=12Hz,1H),4.83–4.76(m,1H),4.69(d,J=12Hz,1H),4.22(d,J=6Hz,1H),3.98(s,3H),3.38–3.31(m,1H),2.96–2.90(m,1H),2.81–2.74(m,2H),2.65(s,3H),2.62–2.54(m,1H),2.37–2.32(m,1H),2.14–2.09(m,1H),1.96–1.88(m,2H),1.76–1.73(m,1H),1.56–1.54(m,6H),1.43(d,6H),1.39–1.33(m,2H),1.32–1.28(m,1H),1.26–1.17(m,2H),1.16–1.07(m,1H),0.91–0.84(m,6H)ppm。
Example 18:
the synthetic route is as follows:
step 1 Synthesis of Compound 18-2
Compound 18-1(0.7mL,8mmol) was dissolved in EtOH (40mL), then Na2CO3(1g,9.434mmol) and hydroxylamine hydrochloride (0.65g,9.4mmol) were added and the reaction refluxed for 7 hours. Then cooling to room temperature, and adding Na2CO3(1g,9.434mmol) and hydroxylamine hydrochloride (0.65g,9.4mmol), and the reaction was refluxed at elevated temperature overnight. After the reaction is completed, cooling to room temperatureFiltration, removal of ethanol under reduced pressure, dissolution of the residue in 20mL of ether, washing with 10mL of 1M HCl solution, separation of the organic phase, adjustment of the pH of the organic phase to neutrality with aqueous ammonia, separation, extraction of the aqueous phase with ether (30mL × 3), combination of the organic phases, washing of the organic phase with saturated sodium chloride solution, drying over anhydrous sodium sulfate, filtration, removal of the solvent gave compound 18-2 as a white solid (0.6g, yield: 70%).
MS(ESI,pos.ion)m/z:103.3[M+1]+
Step 2 Synthesis of Compound 18-4
Compound 18-2(4g,39.2mmol) was dissolved in pyridine (20mL,247mmol), stirred at 0 ℃ for 10 minutes, then compound 18-3(5.3mL,47mmol) was added dropwise, and after completion of the addition, the reaction was stirred at 80 ℃ for 2 hours. The reaction solution was poured into 100mL of ice water, extracted once with 30mL of dichloromethane, and the resulting organic phase was washed once with 10mL of 1M hydrochloric acid solution and once with saturated sodium chloride solution (20mL), then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound 18-4 as a white solid (6.1g, yield: 85%).
MS(ESI,pos.ion)m/z:185.1[M+1]+
1H NMR(600MHz,CDCl3):4.49–4.41(m,2H),3.18–3.05(m,1H),1.41–1.35(m,3H),1.32–1.27(m,6H)ppm。
Step 3 Synthesis of Compound 18-5
Compound 18-4(5.6g,30mmol) was dissolved in ethanol (10mL), and a solution of NaOH (1.3g,33mmol) in water (5mL) was slowly added under ice-bath, followed by warming to 60 ℃ for 1.5 hours. After completion of the reaction, the solvent was removed under reduced pressure to give compound 18-5(5.0g, yield: 92%) as a white solid.
Step 4 Synthesis of Compound 18-6
Compound 4-6(0.3g,0.4mmol) was dissolved in 2ml of ethyl acetate, cooled to 0 ℃ and then 4ml of a 30% strength ethyl acetate solution of hydrogen chloride was added and stirred at room temperature until the reaction was complete without gas evolution. Filtration and washing of the resulting white solid once with 20ml of ethyl acetate. The resulting white solid, compound 4-7, compound 18-5(0.05g,0.3mmol), EDCI (0.05g,0.3mmol) and HOAT (0.040g,0.29mmol) were dissolved in DMF (5mL), DIPEA (97mg,0.75mmol) was added slowly at 0 ℃ and after the addition was complete, the reaction was allowed to warm to room temperature overnight. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether ethyl acetate (V: V) ═ 2:1 as eluent, to give purified compound 18-6(135mg, yield: 50%)
MS(ESI,pos.ion)m/z:902.6[M+1]+
1HNMR(600MHz,CDCl3):10.15(s,1H),8.03(d,J=12Hz,1H),7.59(s,1H),7.11(s,1H),6.74(s,1H),5.80–5.76(m,1H),5.60(s,1H),5.40–5.33(m,1H),5.06–5.03(m,1H),4.82–4.80(m,1H),4.65–4.63(m,1H),4.56(d,J=12Hz,1H),4.18(d,J=12Hz,1H),4.00(s,3H),3.31–3.26(m,1H),3.21–3.15(m,1H),2.97–2.92(m,1H),2.79(s,3H),2.61–2.53(m,1H),2.37–2.31(m,1H),2.28–2.26(m,1H),2.07–2.04(m,2H),2.02–1.99(m,2H),1.86–1.81(m,1H),1.73–1.63(m,1H),1.43-1.41(d,6H),1.41-1.35(d,5H),1.20-1.15(m,2H),1.15-1.05(m,2H),1.21–1.18(m,2H),1.15–1.10(m,2H),1.02–0.95(m,2H),0.91–0.86(m,2H)ppm。
Example 19:
the synthetic route is as follows:
step 1 Synthesis of Compounds 19-6
AcOH (25mL) was placed in a reaction flask, hydroxylamine hydrochloride (4.6g,66mmol) and compound 19-3(5.0g, 50mmol) were added sequentially, AcONa (5.4g, 66mmol) was added in portions, and the reaction was stirred at room temperature for 2 hours. TLC monitored the completion of the reaction of 19-3 starting material, at which point intermediate 19-4 was formed. Adding Ac slowly into the reaction solution2O (7.2mL,76mmol), and stirring was continued for 30 min. TLC monitored compound 19-4 after completion of the reaction, at which point intermediate 19-5 was formed. The reaction solution was heated to 100 ℃ and stirred for 12 hours. After TLC monitoring of the completion of the reaction of compound 19-5, acetic acid was removed under reduced pressure, and the resulting residue was dissolved in 150mL of ethyl acetate, washed successively with 30mL of water and saturated 30mL of aqueous sodium chloride solution, the organic phase was dried over anhydrous sodium sulfate, and finally concentrated under reduced pressure to give compound 19-6(3.1g) as a white solid, which was directly subjected to the next reaction without purification.
MS(ESI,pos.ion)m/z:157[M+1]+
Step 2 Synthesis of Compound 19-1
Compound 19-6(3.1g,20mmol) was dissolved in EtOH (40mL) and aqueous potassium hydroxide (11.68mol/L, 2mL,23.36mmol) was added to precipitate a solid immediately, which was stirred at room temperature overnight. The reaction solution was filtered, and the filter cake was washed with a small amount of ethanol and dried under vacuum to give compound 19-1(2.4g,14mmol, yield: 73%) as a white solid.
Step 3 Synthesis of Compound 19-2
Compound 4-7(0.15g,0.19mmol), compound 19-1(0.04g,0.23mmol), EDCI (0.04g,0.21mmol) and HOAT (0.03g,0.22mmol) were charged into a round bottom flask, under nitrogen, 20mL of dichloromethane were added, the reaction was cooled to 0 deg.C and DIPEA (0.1mL,0.6mmol) was added. The reaction mixture was warmed to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, and then extracted twice with 10ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 19-2 as a white solid (0.07g, yield 42%).
MS(ESI,pos.ion)m/z:875[M+1]+
1H NMR(600MHz,CDCl3)10.32(s,1H),7.93(d,J=8.9Hz,1H),7.71(d,J=6.7Hz,1H),7.63(s,1H),7.50(s,1H),7.13(d,J=9.2Hz,1H),7.04(s,1H),5.69(dd,J=18.0,8.7Hz,1H),5.52(s,1H),4.94(t,J=9.4Hz,1H),4.77(s,1H),4.69(t,J=7.7Hz,1H),4.42(d,J=11.3Hz,1H),4.13(d,J=8.2Hz,1H),3.86(s,3H),3.27–3.17(m,1H),2.90–2.77(m,2H),2.64(d,J=11.7Hz,6H),2.56–2.47(m,1H),2.28–2.19(m,1H),2.06–1.93(m,2H),1.87–1.81(m,1H),1.73(s,1H),1.58–1.52(m,1H),1.46–1.44(m,2H),1.41(d,J=6.9Hz,6H),1.33–1.25(m,4H),1.14–1.04(m,2H),0.92–0.85(m,3H)ppm。
Example 20:
the synthetic route is as follows:
step 1 Synthesis of Compound 20-2
Adding the compound 20-1(50g,216mmol), triphenylphosphine (68g, 259mmol) and dichloromethane (375mL) into a reaction bottle, cooling to-10 ℃ under the protection of nitrogen, slowly adding DIAD (52.5g, 260mmol) dropwise, continuing to stir at-10 ℃ for 3 hours after dropping, adding methanesulfonic acid (62.5g, 650mmol) after reaction is completed, heating to 40 ℃ and stirring for 2 hours. After completion of the reaction, it was cooled to room temperature, filtered, the filter cake was washed once with a small amount of dichloromethane, and the resulting solid was dried under vacuum at 40 ℃ for 4 hours to give compound 20-2(40g, yield 88.4%) as a white solid.
Step 2 Synthesis of Compound 20-3
Compound 20-2(10g,47.8mmol), compound 3-7(10g, 36.8mmol), ethyl 2-oxime cyanoacetate (1.3g, 9.1mmol), DIPEA (9.0mL,54mmol) and dichloromethane (250mL) were charged to a round bottom flask, EDCI (0.85g,4.4mmol) was added under nitrogen and the reaction mixture was reacted at room temperature for 3 hours. After completion of the reaction, 250mL of aqueous solution was added to the reaction mixture to separate the solution, and the organic phase was washed once with 10% aqueous citric acid (250mL), saturated sodium bicarbonate (250mL) and saturated sodium chloride solution (250mL), dried over anhydrous sodium sulfate, and finally concentrated under reduced pressure to give compound 20-3(12.7g, yield 94.1%) as a brownish red oily liquid.
And step 3: synthesis of Compound 20-5
Compound 20-3(13.37g,36.49mmol) was added to a 250mL one-neck flask, toluene (13mL) and water (130mL) were added, and finally compound 20-4(11.66g,37.20mmol) and sodium isooctanoate (10.00g,54.85mmol) were added and reacted at room temperature overnight. After completion of the reaction, ethyl acetate (150 mL. times.2) was added to extract the separated liquid, and the organic phases were combined. The organic phase was washed with 150mL of a saturated sodium bicarbonate solution, 1mol/L of hydrochloric acid (150mL) and saturated sodium chloride, respectively, dried over anhydrous sodium sulfate, and the organic solvent was spin-dried under reduced pressure to give 20-5(18.52g, yield: 100%) as a brownish red oily liquid compound.
And 4, step 4: synthesis of Compounds 20-7
Adding 20-5(18.5g,36.4mmol) of the compound, 20-6(10.48g, 47.3mmol) of the compound and toluene (100mL) into a 250mL single-neck flask, cooling the reaction mixture to-10 ℃, slowly dripping a solution of potassium tert-butoxide (6.12g, 54.6mmol) in anhydrous tetrahydrofuran (20mL) into the reaction mixture, controlling the reaction temperature to be not higher than-5 ℃, and after dripping, continuing to stir the reaction mixture at-5 ℃ for 3 hours. After the reaction was complete, 100mL of 1mol/L hydrochloric acid solution was added, the reaction mixture was warmed to room temperature and stirred for 30 minutes, then the layers were separated, the aqueous phase was extracted once with toluene (100mL), and the organic phases were combined. The organic phase was washed once with 100mL of a saturated sodium bicarbonate solution and 1mol/L hydrochloric acid (150mL), and finally with saturated sodium chloride, dried over anhydrous sodium sulfate, and the organic solvent was spin-dried under reduced pressure to give compound 20-7(24.0g, yield 97.6%) as a brownish red oily liquid.
And 5: synthesis of Compounds 20-8
Compound 20-7(24g,35.4mmol), compound 1-8(10g, 31.8mmol), cesium carbonate (15g, 46mmol) and N-methylpyrrolidone (70mL) were added to a 250mL single-necked flask, and the mixture was heated to 50 ℃ and stirred overnight. After the reaction is completed, 100mL of water and 70mL of methyl tert-butyl ether are added, extraction and liquid separation are carried out, the water phase is extracted once by methyl tert-butyl ether (70mL), organic phases are combined, then 70mL of saturated sodium bicarbonate and saturated sodium chloride are respectively used for washing, anhydrous sodium sulfate is used for drying, organic solvents are dried in a rotary manner under reduced pressure to obtain brownish red oily liquid, then isopropanol (210 mL) is used for heating and dissolving the brownish red oily liquid, and cooling is carried out to room temperature to separate out 20-8(20.0g, yield 80%) of white solid compounds.
Step 6: synthesis of Compounds 20-9
The compound 20-8(20g,24.8mmol) and toluene (1400mL) were added to the reaction flask, warmed to 110 deg.C under reflux and stirred for one hour. The Jansen 1B catalyst (0.08g, 0.1mmol) is dissolved in toluene (200mL), slowly added dropwise into the reaction solution under the protection of nitrogen for 3 hours, and after the dropwise addition is finished, the reaction solution is continuously stirred under reflux for 2 hours. After completion of the reaction, the solvent was dried under reduced pressure to give a brown-gray oily liquid, and methyl t-butyl ether (80mL) was added thereto, and the mixture was heated to reflux to dissolve the brown-gray oily liquid, followed by cooling to room temperature to precipitate a white solid compound 20-9(15.0g, yield: 80%).
And 7: synthesis of Compounds 20-10
Compound 20-9(10g,12.89mmol), lithium hydroxide monohydrate (1.1g, 26mmol), methanol (40mL), tetrahydrofuran (40mL) and water (20mL) were added to a 250mL single-necked flask and stirred at room temperature overnight. After the reaction was completed, the organic solvent was spin-dried under reduced pressure, 50ml of 1mol/L hydrochloric acid and 50ml of ethyl acetate were added, the layers were separated by extraction, the aqueous phase was extracted once with ethyl acetate (50ml), and the organic phases were combined. The organic phase was washed with 50ml of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, and the organic solvent was spin-dried under reduced pressure to give 20-10(9.15g, yield: 93.2%) as a white solid.
And 8: synthesis of Compounds 20-12
20-10(2g,2.625mmol), CDI (0.87g, 5.3mmol) and dichloromethane (20mL) were added to a round bottom flask and stirred at room temperature for 3 hours, followed by addition of DBU (0.82g, 5.3mmol) and compound 20-11(0.72g, 5.3mmol), and the reaction mixture stirred at room temperature overnight. After completion of the reaction, 40mL of 1mol/L hydrochloric acid was added, the mixture was subjected to extraction and liquid separation, the aqueous phase was extracted once with methylene chloride (10mL), the organic phases were combined, washed with 50mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, and the organic solvent was spin-dried under reduced pressure to obtain 20-12(2.2g, yield: 94.42%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:880.8[M+1]+
1H NMR(600MHz,CDCl3):10.31(s,1H),8.03(d,J=7.5Hz,1H),7.93(s,1H),7.84–7.79(m,2H),7.54(s,1H),7.06–7.01(m,2H),5.70(dd,J=17.9,8.7Hz,1H),5.52(s,1H),5.03–4.98(m,1H),4.76–4.69(m,2H),4.65–4.60(m,1H),4.13–4.09(m,1H),3.89(s,3H),3.73(s,3H),3.26–3.19(m,2H),2.75(dd,J=13.8,7.4Hz,1H),2.65(s,3H),2.31(d,J=8.6Hz,1H),2.07–2.04(m,1H),1.85(dd,J=15.5,9.2Hz,2H),1.78(dd,J=10.5,5.1Hz,1H),1.66–1.62(m,1H),1.50(d,J=7.3Hz,4H),1.40(d,J=6.9Hz,7H),1.26(d,J=4.2Hz,3H),1.20(s,3H),0.86–0.77(m,3H)ppm。
And step 9: synthesis of target Compounds 20-15
Compound 20-12(0.2g,0.2mmol) was dissolved in 2mL of isopropanol, cooled to 0 deg.C, then 5mL of isopropanol solution containing 40% hydrogen chloride was added and stirred at room temperature until the reaction was complete when no gas was evolved. Filtration and washing of the resulting filter cake with 5ml of ethyl acetate followed by vacuum drying gave a white solid. The resulting white solid, compound 20-14(0.1g,0.6mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were added to a round bottom flask under nitrogen, 10mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, taken twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the resulting crude product was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 20-15(0.140g, yield 60%) as a white solid.
MS(ESI,pos.ion)m/z:890.8[M+1]+
1H NMR(600MHz,CDCl3):10.31(s,1H),8.03(d,J=7.5Hz,1H),7.93(s,1H),7.84–7.79(m,2H),7.54(s,1H),7.06–7.01(m,2H),5.70(dd,J=17.9,8.7Hz,1H),5.52(s,1H),5.03–4.98(m,1H),4.76–4.69(m,2H),4.65–4.60(m,1H),4.13–4.09(m,1H),3.89(s,3H),3.73(s,3H),3.26–3.19(m,2H),2.75(dd,J=13.8,7.4Hz,1H),2.65(s,3H),2.31(d,J=8.6Hz,1H),2.07–2.04(m,1H),1.85(dd,J=15.5,9.2Hz,2H),1.78(dd,J=10.5,5.1Hz,1H),1.66–1.62(m,1H),1.50(d,J=7.3Hz,4H),1.40(d,J=6.9Hz,7H),1.26(d,J=4.2Hz,3H),1.20(s,3H),0.86–0.77(m,3H).
Example 21:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(0.2g,0.2mmol) was dissolved in 2ml of isopropanol, cooled to 0 ℃ and then 5ml of a 40% strength solution of hydrogen chloride in isopropanol was added and stirred at room temperature until the reaction was complete when no gas was evolved. Filtration and washing of the white solid with 5ml of ethyl acetate and drying in vacuo at room temperature. The white solid obtained by the above reaction and compound 21-1(0.1g,0.6mmol) were added to 10mL of dichloromethane, cooled to 0 ℃ under nitrogen, DIPEA (0.5mL,3mmol) was added, and the reaction mixture was warmed to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, and then extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 21-2(0.130g, yield 60%) as a white solid.
MS(ESI,pos.ion)m/z:889.4[M+1]+
1H NMR(600MHz,CDCl3):10.31(s,1H),8.03(d,J=7.5Hz,1H),7.93(s,1H),7.84–7.79(m,2H),7.54(s,1H),7.06–7.01(m,2H),5.70(dd,J=17.9,8.7Hz,1H),5.52(s,1H),5.03–4.98(m,1H),4.76–4.69(m,2H),4.65–4.60(m,1H),4.13–4.09(m,1H),3.89(s,3H),3.73(s,3H),3.26–3.19(m,2H),2.75(dd,J=13.8,7.4Hz,1H),2.65(s,3H),2.31(d,J=8.6Hz,1H),2.07–2.04(m,1H),1.85(dd,J=15.5,9.2Hz,2H),1.78(dd,J=10.5,5.1Hz,1H),1.66–1.62(m,1H),1.50(d,J=7.3Hz,4H),1.40(d,J=6.9Hz,7H),1.26(d,J=4.2Hz,3H),1.20(s,3H),0.86–0.77(m,3H)ppm。
Example 22:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(0.2g,0.2mmol) was dissolved in 2ml of isopropanol, cooled to 0 ℃ and then 5ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 22-1(0.1g,0.7mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were charged to a round bottom flask, protected with nitrogen, 10mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added, warmed to 30 deg.C, and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, and taken twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, and the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 20-2(0.163g, yield 70%) as a white solid.
MS(ESI,pos.ion)m/z:900.4[M+1]+
1H NMR(600MHz,CDCl3):10.25(s,1H),9.07(s,1H),8.43(s,1H),8.17(d,J=6.8Hz,1H),7.86–7.78(m,2H),7.48(s,1H),7.05(s,1H),6.95(d,J=9.1Hz,1H),5.74(dd,J=18.0,8.6Hz,1H),5.51(s,1H),5.02(t,J=9.5Hz,1H),4.72–4.65(m,2H),4.51(d,J=11.4Hz,1H),4.14–4.11(m,1H),3.78(s,3H),3.26–3.19(m,2H),2.74–2.69(m,1H),2.67–2.55(m,9H),2.29(dd,J=17.3,8.6Hz,1H),2.07–2.00(m,2H),1.95–1.89(m,2H),1.78(t,J=10.0Hz,1H),1.64(dd,J=9.2,5.9Hz,1H),1.50(s,3H),1.41(d,J=6.9Hz,7H),1.27(d,J=3.5Hz,2H),1.20(s,3H),0.92–0.75(m,3H)ppm。
Example 23:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.5g,1.7mmol) was dissolved in 20ml of isopropanol, cooled to 0 ℃ and then 20ml of a 40% by weight solution of hydrogen chloride in isopropanol was added and stirred at room temperature until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 23-1(0.6g,3mmol), EDCI (0.4g,3mmol) and HOAT (0.3g,2.2mmol) were charged to a round bottom flask, protected with nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2mL,12.1mmol) was added, warmed to 30 deg.C and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, taken twice with 20ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography with petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 23-2(0.75g, yield 45%) as a white solid.
MS(ESI,pos.ion)m/z:917.4[M+1]+
1H NMR(400MHz,CDCl3):10.27(s,1H),8.04(dd,J=29.9,23.6Hz,2H),7.84(d,J=11.6Hz,2H),7.52(s,1H),7.05(d,J=12.8Hz,2H),5.70(d,J=7.9Hz,1H),5.51(s,1H),5.01(dd,J=22.2,12.9Hz,2H),4.78–4.61(m,3H),4.14(d,J=9.9Hz,1H),3.97(d,J=17.5Hz,2H),3.90(s,3H),3.22(s,1H),2.69(d,J=18.9Hz,6H),2.32(d,J=7.6Hz,2H),2.10–2.02(m,1H),1.84(s,2H),1.77(d,J=9.6Hz,1H),1.69(s,1H),1.53(d,J=6.1Hz,3H),1.49(s,3H),1.41(s,3H),1.32–1.19(m,10H),0.90–0.68(m,3H)ppm。
Example 24:
the synthetic route is as follows:
step 1 Synthesis of Compound 24-4
Compound 24-3(20g,172.18mmol), hydrazine hydrate (20mL,514.5mmol,25.73mol/L) was dissolved in ethanol (30mL) and reacted for 4 hours under reflux. After completion of the reaction, it was cooled to room temperature, and the solvent was dried by spinning to obtain the product 24-4(10.7g, yield: 61%) as a white solid.
MS(ESI,pos.ion)m/z:103[M+1]+
Step 2 Synthesis of Compound 24-6
Compound 24-4(3.0g,29mmol) was dissolved in dichloromethane (40mL), triethylamine (4mL,29mmol) was added under ice bath followed by slow addition of compound 24-5(4.0g,29mmol) which was allowed to warm to room temperature and stirred overnight. After completion of the reaction, 100mL of ethyl acetate was added to the reaction mixture to dilute the mixture, the resulting salt was removed by filtration, the filtrate was dried by spinning, and the resulting residue was purified by a silica gel column using petroleum ether, ethyl acetate (V: V) ═ 5:1, to give compound 24-6(5.8g, yield: 99%) as a white solid.
MS(ESI,pos.ion)m/z:203[M+1]+
Step 3 Synthesis of Compounds 24-7
Compound 24-6(2.0g,9.9mmol) was dissolved in dichloromethane (40mL), the resulting solution was placed in an ice bath, triethylamine (1.8mL,13mmol) and p-toluenesulfonyl chloride (2.3g,12mmol) were added to the reaction solution, and the mixture was warmed to room temperature and stirred for 8 hours. After completion of the reaction, 50mL of water was added to quench the reaction, the organic phase was dried over sodium sulfate, concentrated under reduced pressure, and the obtained residue was purified by a silica gel column, eluting with petroleum ether and ethyl acetate (V: V) ═ 5:1, to obtain 24-7(1.1g, yield: 60%) as a white solid compound
MS(ESI,pos.ion)m/z:185[M+1]+
And 4, step 4: synthesis of Compound 24-2
Compound 24-7(1.1g,6mmol) was dissolved in ethanol (25mL), potassium trimethylsilanolate (0.9g,7mmol) was added with stirring, and a white solid was formed, which was stirred at room temperature overnight. After completion of the reaction, filtration was carried out, and the filter cake was washed with a small amount of ethanol and dried to obtain the product 24-2 as a white solid (0.81g, yield: 70%).
1H NMR(400MHz,D2O)3.26–3.12(m,1H),1.31(d,J=7.0Hz,6H)ppm。
And 5: synthesis of Compound 24-1
Compounds 1-7(0.5g,0.6mmol), 24-2(0.14g,0.7mmol), EDCI (0.12g,0.63mmol) and HOAT (0.1g,0.67mmol) were added to a round bottom flask, under nitrogen, 20mL of dichloromethane were added, then cooled to 0 deg.C and DIPEA (0.3mL,1.8mmol) was added. The reaction solution was heated to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, followed by extraction twice with 10ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, and then the organic solvent was removed under reduced pressure, and the resulting residue was purified by a silica gel column, eluting with petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 24-1(0.14g, yield 20%) as a white solid.
MS(ESI,pos.ion)m/z:903[M+1]+
1H NMR(600MHz,CDCl3)10.37(s,1H),8.64(s,1H),8.03(s,1H),7.90(d,J=9.0Hz,1H),7.57(s,1H),7.11(d,J=9.1Hz,1H),7.04(s,1H),5.80–5.67(m,1H),5.57(s,1H),5.05–4.96(m,1H),4.86–4.78(m,1H),4.71–4.57(m,2H),4.22(d,1H),3.90(s,3H),3.27–3.18(m,1H),3.16–3.05(m,1H),2.94–2.87(m,1H),2.82–2.74(m,2H),2.68(s,4H),2.31–2.25(m,1H),2.22–2.15(m,1H),1.87–1.83(m,2H),1.40(d,J=6.9Hz,7H),1.31(dd,J=18.4,6.9Hz,11H),1.21–1.03(m,3H),0.97–0.82(m,3H)ppm。
Example 25:
the synthetic route is as follows:
step 1: synthesis of Compound 25-4
Ethanol (50mL) was placed in a reaction flask, hydroxylamine hydrochloride (5.3g,76mmol) and compound 25-3(5.0g,50 mmol) were added, and K was added portionwise2CO3(8.4g,61mmol), 25mL water was added with final stirring, after addition, the reaction mixture was stirred at room temperature for 2h TLC to monitor completion of the starting material, ethanol was removed by spinning, 20mL water was added for dilution, then DCM (30mL × 5) was used for extraction, the organic phase was dried over anhydrous sodium sulfate, and then dried to give compound 25-4 as a white solid (2.3g, yield: 35%).
Step 2: synthesis of Compounds 25-5
Compound 25-4(1.0g,7.6mmol) was dissolved in dichloromethane (20mL), isobutyryl chloride (1.2g,11mmol) was slowly added dropwise in an ice bath, and after completion of the addition, the mixture was warmed to room temperature and stirred for 4 hours. After the reaction, 50mL of water was added to quench the reaction, dichloromethane was used for extraction (30 mL. times.2), the organic phases were combined, the organic phase was dried over anhydrous sodium sulfate, filtered and then dried by spinning to obtain 25-5(1.5g, yield: 98%) as a colorless oily product, which was directly subjected to the next reaction without purification
1H NMR(400MHz,CDCl3)4.40(q,J=7.1Hz,2H),2.74(m,1H),1.40(t,J=7.1Hz,3H),1.27(d,J=7.0Hz,6H)ppm。
And step 3: synthesis of Compound 25-6
Compound 25-5(1.5g,7.4mmol) was dissolved in acetic acid and allowed to react overnight at 100 ℃. After the reaction, the reaction solution was evaporated under reduced pressure to remove acetic acid, the residue was dissolved in 50mL of ethyl acetate, washed with 30mL of water and 30mL of saturated sodium chloride in this order, and the organic phase was spin-dried to obtain a pale yellow oily compound 25-6, which was subjected to the next reaction without purification.
MS(ESI,pos.ion)m/z:185[M+1]+
And 4, step 4: synthesis of Compound 25-2
Compound 25-6(1.5g,20mmol) was dissolved in EtOH (40mL), followed by addition of aqueous potassium hydroxide (11.68mol/L, 2mL) and immediate precipitation of a solid, which was stirred at room temperature overnight. The reaction solution was filtered, and the filter cake was washed with a small amount of ethanol and then dried to obtain 25-2(2.4g, yield: 62%)
13C NMR(150MHz,D2O)178.82,165.05,163.27,11.60ppm。
And 5: synthesis of Compound 25-1
Compound 4-7(0.5g,0.6mmol), compound 25-2(0.1g,0.7mmol), EDCI (0.12g,0.6mmol) and HOAT (0.1g,0.7mmol) were charged to a round bottom flask, protected with nitrogen, 20mL of dichloromethane were added, then cooled to 0 deg.C and DIPEA (0.3mL,1.8mmol) was added. The reaction was warmed to 30 ℃ and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of ethyl acetate, the organic phases were combined, the organic phase was washed with 20ml of saturated brine, dried over anhydrous sodium sulfate, the organic solvent was removed under reduced pressure, and the resulting residue was purified by a silica gel column, eluting with petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 25-1(0.29g, yield 50%) as a white solid.
MS(ESI,pos.ion)m/z:903[M+1]+
1H NMR(400MHz,CDCl3)10.25(s,1H),7.98(d,J=9.1Hz,1H),7.69(d,J=7.4Hz,1H),7.53(s,1H),7.20(d,J=9.6Hz,2H),7.04(s,1H),5.77–5.63(m,1H),5.55(s,1H),4.97(t,J=9.4Hz,1H),4.86(t,J=6.9Hz,1H),4.64(t,J=7.7Hz,1H),4.51(d,J=11.3Hz,1H),4.17(dd,J=11.1,3.6Hz,1H),3.94(s,3H),3.77–3.71(m,1H),3.61–3.41(m,3H),3.31–3.16(m,2H),2.93–2.83(m,1H),2.70(s,4H),2.34–2.24(m,1H),1.91–1.84(m,2H),1.62–1.54(m,2H),1.44–1.38(m,14H),1.19–1.04(m,3H),0.97–0.87(m,4H)ppm。
Example 26:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(0.2g,0.25mmol) was dissolved in 5ml of isopropanol, cooled to 0 ℃ and then 5ml of a 40% by weight solution of hydrogen chloride in isopropanol was added and stirred at room temperature until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting solid, compound 26-1(0.067g,0.367mmol), EDCI (0.07g,0.367mmol) and HOAT (0.05g,0.367mmol) were charged to a round bottom flask, nitrogen blanketed, 10mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (0.4mL,2mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. The reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, and the organic solvent was removed under reduced pressure, and the resulting residue was purified by a silica gel column using petroleum ether, ethyl acetate (V: V) ═ 1:2, to give compound 26-2 as a white solid (0.160g, yield 70%).
MS(ESI,pos.ion)m/z:938.4[M+1]+
1H NMR(600MHz,CDCl3)10.16(s,1H),7.96(d,J=8.6Hz,1H),7.55(d,J=37.1Hz,2H),7.41(s,1H),7.20(s,1H),6.79(s,1H),6.62(s,1H),5.73(dd,J=17.9,8.6Hz,1H),5.60(s,1H),5.09–5.00(m,1H),4.68(dd,J=32.0,27.1Hz,4H),4.20–4.13(m,1H),3.94(s,2H),3.67–3.35(m,5H),3.31(dd,J=13.6,6.8Hz,1H),3.25(s,3H),2.78(s,2H),2.64(s,2H),2.57(s,1H),2.35–2.26(m,1H),1.95(dd,J=33.2,15.1Hz,2H),1.81–1.65(m,2H),1.43(d,J=6.9Hz,4H),1.38–1.31(m,2H),1.31–1.25(m,2H),1.22(s,7H),0.88–0.75(m,2H)ppm。
Example 27:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(0.2g,0.2mmol) was dissolved in 2ml of isopropanol, cooled to 0 ℃ and then 5ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 27-1(0.1g,0.7mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were added to a round bottom flask under nitrogen, 10mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added, the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. The reaction was quenched with 10ml of water, taken twice with 10ml of dichloromethane, the organic phases were combined, washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 27-2 as a white solid (0.120g, yield 60%).
MS(ESI,pos.ion)m/z:900.4[M+1]+
1H NMR(600MHz,CDCl3):10.22(s,1H),7.92(d,J=9.1Hz,1H),7.81–7.74(m,2H),7.51(s,1H),7.12(t,J=8.8Hz,1H),7.04(s,1H),5.67(dd,J=18.1,8.6Hz,1H),5.53(s,1H),4.96(t,J=9.6Hz,1H),4.83–4.78(m,1H),4.70(t,J=7.8Hz,1H),4.43(d,J=11.4Hz,1H),4.17–4.12(m,1H),3.88(s,3H),3.24–3.19(m,1H),2.80(dd,J=13.9,7.7Hz,1H),2.65(s,3H),2.61(s,3H),2.53–2.45(m,1H),2.26–2.23(m,1H),2.21–2.11(m,2H),2.01(d,J=12.1Hz,1H),1.95–1.90(m,1H),1.83(dd,J=7.7,6.2Hz,1H),1.52(t,J=4.6Hz,1H),1.48(s,4H),1.45(d,J=8.7Hz,3H),1.40(d,J=6.9Hz,6H),1.28–1.25(m,3H),0.87–0.77(m,3H)ppm。
Example 28:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(0.2g,0.2mmol) was dissolved in 2ml of isopropanol, cooled to 0 ℃ and then 5ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 28-1(0.1g,0.7mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were added to a round bottom flask under nitrogen, 10mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added, the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 28-2(0.165g, yield 80%) as a white solid.
MS(ESI,pos.ion)m/z:889.3[M+1]+
1H NMR(600MHz,CDCl3):10.30(s,1H),8.09(d,J=7.4Hz,1H),7.94(s,1H),7.82(d,J=9.1Hz,1H),7.57(s,1H),7.02(d,J=12.2Hz,2H),6.13(s,1H),5.67(dd,J=17.6,8.7Hz,1H),5.56(s,1H),4.95(t,J=9.4Hz,1H),4.78(t,J=7.8Hz,1H),4.71(t,J=7.8Hz,1H),4.65(d,J=11.4Hz,1H),4.13(dt,J=22.7,11.4Hz,1H),3.86(s,3H),3.24–3.18(m,2H),2.79–2.62(m,6H),2.35(s,3H),2.27(dd,J=16.9,8.3Hz,2H),2.11(dd,J=22.4,10.5Hz,1H),1.81–1.78(m,1H),1.63(s,1H),1.39(d,J=6.8Hz,7H),1.30–1.21(m,6H),1.19(s,3H),0.86–0.73(m,3H)ppm。
Example 29:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(0.2g,0.2mmol) was dissolved in 2ml of isopropanol, cooled to 0 ℃ and then 5ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 29-1(0.1g,0.7mmol), EDCI (0.2g,1.5mmol) and HOAT (0.15g,1.1mmol) were added to a round bottom flask under nitrogen, 10mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (0.5mL,3mmol) was added, the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 29-2(0.150g, yield 70%) as a white solid.
MS(ESI,pos.ion)m/z:874.3[M+1]+
1H NMR(600MHz,CDCl3):10.30(s,1H),8.40(s,1H),7.95(d,J=7.4Hz,1H),7.87(d,J=9.1Hz,1H),7.81(s,1H),7.55(s,1H),7.04(d,J=9.2Hz,2H),6.57(s,1H),5.63(dd,J=17.8,8.6Hz,1H),5.51(d,J=27.3Hz,1H),4.92(t,J=9.4Hz,1H),4.76(t,J=7.3Hz,1H),4.70(t,J=7.8Hz,1H),4.58(d,J=11.4Hz,1H),4.16–4.10(m,1H),3.88(s,3H),3.26–3.20(m,2H),2.75(dd,J=13.6,7.7Hz,1H),2.65(d,J=18.3Hz,4H),2.54–2.48(m,1H),2.38(s,1H),2.26(dd,J=17.2,8.5Hz,1H),2.05(dd,J=21.8,10.4Hz,1H),1.89–1.83(m,1H),1.79–1.69(m,2H),1.43(d,J=5.5Hz,2H),1.39(d,J=6.9Hz,7H),1.28–1.23(m,3H),1.19(s,3H),0.96–0.66(m,3H)ppm。
Example 30:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.5g,1.7mmol) was dissolved in 20ml of isopropanol, cooled to 0 ℃ and then 20ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 30-1(0.6g,3mmol), EDCI (0.4g,3mmol) and HOAT (0.3g,2.2mmol) were charged to a round bottom flask, protected with nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2mL,12.1mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 4 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 20ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give 30-2(0.75g, yield 45%) as a white solid.
MS(ESI,pos.ion)m/z:917.4[M+1]+
1H NMR(400MHz,CDCl3):10.27(s,1H),8.04(dd,J=29.9,23.6Hz,2H),7.84(d,J=11.6Hz,2H),7.52(s,1H),7.05(d,J=12.8Hz,2H),5.70(d,J=7.9Hz,1H),5.51(s,1H),5.01(dd,J=22.2,12.9Hz,2H),4.78–4.61(m,3H),4.14(d,J=9.9Hz,1H),3.97(d,J=17.5Hz,2H),3.90(s,3H),3.22(s,1H),2.69(d,J=18.9Hz,6H),2.32(d,J=7.6Hz,2H),2.10–2.02(m,1H),1.84(s,2H),1.77(d,J=9.6Hz,1H),1.69(s,1H),1.53(d,J=6.1Hz,3H),1.49(s,3H),1.41(s,3H),1.32–1.19(m,10H),0.90–0.68(m,3H)ppm。
Example 31:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(3g,3.7mmol) was dissolved in 20ml of isopropanol, cooled to 0 ℃ and then 20ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 31-1(1.3g,7.8mmol), EDCI (1.5g,7.8mmol) and HOAT (0.9g,6mmol) were charged to a round bottom flask, 30mL of dichloromethane were added under nitrogen, then cooled to 0 deg.C, DIPEA (3mL,17.1mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 30ml of water, extracted twice with 30ml of dichloromethane, the organic phases were combined, washed with 30ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give 31-2(1.3g, yield 39%) as a white solid.
MS(ESI,pos.ion)m/z:917.3[M+1]+
1H NMR(400MHz,CDCl3):10.32(s,1H),8.21(d,J=8.0Hz,1H),7.97(s,1H),7.86(d,J=9.1Hz,1H),7.57(s,1H),7.05(d,J=9.4Hz,2H),6.14(s,1H),5.68(dd,J=17.6,8.7Hz,1H),5.56(s,1H),5.02–4.93(m,1H),4.81(t,J=8.1Hz,1H),4.71(t,J=8.1Hz,2H),4.17(dd,J=11.2,3.3Hz,1H),3.89(s,3H),3.23(dt,J=13.6,6.8Hz,1H),2.99(dq,J=13.7,6.9Hz,1H),2.76(d,J=7.1Hz,2H),2.69(s,3H),2.29(d,J=9.5Hz,2H),2.15(dd,J=22.6,11.1Hz,1H),1.79(dd,J=17.2,9.8Hz,3H),1.66(s,1H),1.48(d,J=10.1Hz,7H),1.40(d,J=6.9Hz,7H),1.23(dd,J=11.9,7.0Hz,9H),0.86–0.76(m,2H)ppm。
Example 32:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.35g,1.66mmol) was dissolved in 15ml of isopropanol, cooled to 0 ℃ and then 15ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 32-1(0.3g,2mmol), EDCI (0.5g,4mmol) and HOAT (0.5g,4mmol) were charged to a round bottom flask under nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2.5mL,14mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 30ml of water, extracted twice with 30ml of dichloromethane, the organic phases were combined, washed with 30ml of saturated brine, dried over anhydrous sodium sulfate, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1, to give compound 32-2(0.72g, yield 49%) as a white solid.
MS(ESI,pos.ion)m/z:888.4[M+1]+
1H NMR(400MHz,CDCl3):10.35(s,1H),8.00(s,1H),7.89(d,J=9.1Hz,1H),7.49(s,1H),7.34(d,J=2.2Hz,2H),7.03(s,1H),6.98(d,J=9.3Hz,1H),6.63(d,J=2.1Hz,1H),5.65(dd,J=18.1,8.5Hz,1H),5.47(s,1H),4.94(t,J=9.5Hz,1H),4.62(t,J=7.5Hz,2H),4.46(d,J=11.2Hz,1H),4.13(dd,J=11.4,4.2Hz,1H),3.87(s,3H),3.75(s,3H),3.20(dd,J=13.0,5.9Hz,1H),2.67(t,J=10.7Hz,1H),2.61(s,3H),2.53–2.45(m,2H),2.26–2.20(m,1H),1.91(dd,J=24.3,12.3Hz,2H),1.81–1.72(m,2H),1.68–1.59(m,2H),1.46(s,3H),1.39(d,J=6.9Hz,7H),1.25(d,J=9.0Hz,5H),0.94–0.81(m,1H),0.76(s,2H)ppm。
Example 33:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.5g,1.8mmol) was dissolved in 15ml of isopropanol, cooled to 0 ℃ and then 15ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 33-1(0.3g,2mmol), EDCI (0.5g,4mmol) and HOAT (0.5g,4mmol) were charged to a round bottom flask under nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2.5mL,14mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 30ml of water, extracted twice with 30ml of dichloromethane, the organic phases were combined, washed with 30ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the resulting residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 33-2(0.8g, yield 50%) as a white solid.
MS(ESI,pos.ion)m/z:916.4[M+1]+
1H NMR(400MHz,CDCl3)10.20(s,1H),7.99(d,J=9.0Hz,1H),7.57(s,1H),7.41(d,J=2.1Hz,1H),7.28(s,2H),7.11(d,J=9.1Hz,1H),7.04(s,1H),6.69(d,J=2.1Hz,1H),5.73(dd,J=18.0,8.7Hz,1H),5.56(s,1H),5.05–4.99(m,1H),4.79(t,J=7.5Hz,1H),4.66(dd,J=16.6,9.3Hz,2H),4.49(dd,J=13.4,6.7Hz,1H),4.14(ddd,J=18.9,13.2,7.2Hz,2H),3.92(s,3H),3.23(d,J=5.0Hz,1H),2.69(s,6H),2.58(s,1H),2.34(dd,J=16.2,7.5Hz,2H),2.07–2.03(m,2H),1.94–1.84(m,2H),1.76(t,J=11.3Hz,2H),1.54(d,J=6.7Hz,4H),1.41(d,J=6.5Hz,4H),1.27(s,6H),1.21(s,1H),0.94–0.76(m,6H)ppm。
Example 34:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.5g,1.8mmol) was dissolved in 20ml of isopropanol, cooled to 0 ℃ and then 20ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 34-1(0.4g,3mmol), EDCI (0.8g,4mmol) and HOAT (0.5g,4mmol) were charged to a round bottom flask under nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2.5mL,14mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 34-2(0.34g, yield 21%) as a white solid.
MS(ESI,pos.ion)m/z:890.3[M+1]+
1H NMR(600MHz,CDCl3):10.36(s,1H),8.29–8.09(m,2H),7.74(d,J=5.3Hz,1H),7.65(s,1H),7.44(s,1H),7.37(d,J=9.1Hz,1H),5.84(s,1H),5.67(dd,J=18.0,8.5Hz,1H),5.00(t,J=9.4Hz,1H),4.83(s,1H),4.73–4.58(m,2H),4.17(d,J=10.6Hz,1H),4.02(s,3H),3.31(dq,J=13.3,6.6Hz,1H),2.86(dd,J=12.9,6.0Hz,1H),2.71(dd,J=20.4,11.6Hz,1H),2.58(d,J=30.1Hz,3H),2.52(d,J=9.7Hz,1H),2.42(s,3H),2.33(dd,J=16.5,8.1Hz,1H),2.11–2.00(m,1H),1.95–1.85(m,1H),1.79–1.69(m,2H),1.66(t,J=11.9Hz,1H),1.58(s,1H),1.50(s,1H),1.48(s,3H),1.41(t,J=22.4Hz,8H),1.36–1.26(m,4H),0.83–0.72(m,2H)ppm。
Example 35:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.5g,1.8mmol) was dissolved in 20ml of isopropanol, cooled to 0 ℃ and then 20ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 35-1(0.5g, 4mmol), EDCI (0.8g,4mmol) and HOAT (0.5g,4mmol) were added to a round bottom flask under nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2.5mL,14mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to obtain 35-2(0.640g, yield 40%) as a white solid.
MS(ESI,pos.ion)m/z:888.3[M+1]+
1H NMR(400MHz,CDCl3):10.30(s,1H),8.05(d,J=9.1Hz,1H),7.84(s,1H),7.58(s,1H),7.26(s,1H),7.18(d,J=9.3Hz,1H),6.91(d,J=6.9Hz,1H),6.80(d,J=2.9Hz,1H),6.68(s,1H),6.05(d,J=2.6Hz,1H),5.72–5.62(m,2H),5.02(t,J=9.5Hz,1H),4.77–4.60(m,3H),4.13(t,J=10.4Hz,1H),3.92(s,3H),3.30(dt,J=13.0,6.5Hz,1H),2.82–2.73(m,1H),2.70–2.63(m,1H),2.60–2.52(m,4H),2.33(d,J=8.3Hz,4H),1.97(dd,J=32.5,10.3Hz,2H),1.85–1.80(m,1H),1.70(t,J=10.9Hz,2H),1.60(d,J=5.7Hz,1H),1.49(s,5H),1.43(s,4H),1.29(dd,J=12.9,5.3Hz,4H),1.25–1.18(m,2H),0.79(s,2H)ppm。
Example 36:
the synthetic route is as follows:
synthesis procedure
Compound 20-12(1.5g,1.8mmol) was dissolved in 20ml of isopropanol, cooled to 0 ℃ and then 20ml of a 40% strength solution of hydrogen chloride in isopropanol were added until the reaction was complete when no gas was evolved. Filtration and washing of the resulting white solid with 5ml of ethyl acetate. The resulting white solid, compound 36-1(0.4g,3mmol), EDCI (0.8g,4mmol) and HOAT (0.5g,4mmol) were added to a round bottom flask under nitrogen, 30mL of dichloromethane were added, then cooled to 0 deg.C, DIPEA (2.5mL,14mmol) was added and the reaction mixture was warmed to 30 deg.C and stirred for 6 hours. After completion of the reaction, the reaction was quenched with 10ml of water, extracted twice with 10ml of dichloromethane, the organic phases were combined, the organic phase was washed with 10ml of saturated brine, dried over anhydrous sodium sulfate, filtered, the organic solvent was removed under reduced pressure, and the obtained residue was purified by silica gel column chromatography using petroleum ether, ethyl acetate (V: V) ═ 2:1 as eluent, to give compound 36-2(0.60g, yield 40%) as a white solid.
MS(ESI,pos.ion)m/z:889.4[M+1]+
1H NMR(600MHz,CDCl3):10.21(s,1H),7.98–7.84(m,2H),7.56(d,J=19.3Hz,1H),7.44(s,1H),7.26(d,J=7.2Hz,1H),7.08(d,J=9.5Hz,2H),5.72(dd,J=17.7,8.6Hz,1H),5.55(s,1H),5.02(t,J=9.4Hz,1H),4.76(d,J=6.8Hz,1H),4.67(t,J=7.7Hz,1H),4.55(d,J=11.1Hz,1H),4.20(s,3H),4.17–4.12(m,1H),3.89(s,3H),3.60(d,J=40.8Hz,2H),3.27–3.23(m,1H),2.74(dd,J=13.2,7.4Hz,1H),2.64(s,3H),2.58–2.52(m,1H),2.32(dd,J=17.2,8.6Hz,1H),2.08–1.99(m,1H),1.97–1.92(m,1H),1.91–1.87(m,1H),1.79–1.71(m,2H),1.55(dd,J=9.1,6.3Hz,1H),1.49(s,4H),1.41(d,J=6.8Hz,4H),1.36–1.26(m,4H),1.21(s,1H),0.96–0.74(m,3H)ppm。
Biological activity
1) HCV NS3/4A protease inhibition assay
Using FRET (Fluorescence Resonance Energy Transfer) based techniquesThe 520HCV protease detection kit (Anaspec) performs high-throughput screening on HCV NS3/4A protease inhibitors. At 5-FAM/QXLTMFluorescence of 5-FAM by QXL in 520FRET substrate peptideTM520 quenching. 5-FAM/QXLTMThe 520FRET substrate peptide contains a HCV NS3/4A protease cleavage site and can be cleaved into two independent fragments by HCV NS3/4A proteaseThe fragment of (4), 5-FAM, was found to have recovered fluorescence and could be detected at 490/510 nm.
The inhibitory effect of the compounds on HCV NS3/4A protease was evaluated by detecting the change in the intensity of the fluorescent signal of 5-FAM. The experimental procedure is briefly described as follows, compounds are dissolved in DMSO, and mixed well to prepare 10mM mother liquor, diluted with Assay buffer containing DTT, 1 μ M as the initial concentration of drug test, 3-fold gradient dilution, 10 dilution points, 3 μ l of the compound diluted in gradient is added into 384-well plates, each well DMSO has a final concentration of 1%. Then, 3. mu.L of HCV NS3/4A recombinant protease genotype 1a (Anaspec) or HCV NS3/4A recombinant protease genotype 1b (Anaspec) was added to the 384-well plates at a final concentration of 0.25 ng/. mu.l for each well. Positive compound control, negative control and substrate control were set. The 384 well plates were incubated at 25 ℃ for 15min for enzymatic reactions while the substrate solutions were incubated at the same temperature. After 15min, adding 50 times diluted FRET substrate peptide into each hole, and mixing by gentle shaking for 1 min. Immediately placing in a PHERAStar FS multifunctional microplate reader (BMGLAbtech), detecting fluorescence intensity under the condition of Ex/Em being 490/520nm by a kinetic method, recording data every 1min, and continuously detecting for 30 min. The results were processed using GraphPad Prism software to calculate the IC50 value of the compounds for inhibition of HCV NS3/4A enzyme. The compound has good inhibition effect on NS3/4A protease gene types 1a and 1 b.
2) HCV subgenomic replicon assay
GT1a, GT1b and GT2a replicon activity test, HCV GT1a H77 replicon, GT1b Con1b replicon and GT2a JFH1 replicon carrying G418 resistance gene NEO and luciferase reporter gene are respectively transiently transfected into Huh-7 cells by electric shock method, G418 is added for screening for 3-4 weeks to construct stably transfected cell strains, Huh7-H77 and Huh7-JFH1 stably transfected cell strains are diluted to 5 × 104PermL, 200. mu.L into a 96-well plate, and the Huh7-Con1b stable transfectant cell line was diluted to 1 × 105mL, inoculate 50 μ L to 384 well plates. After 16-24h, the compound was diluted to the appropriate concentration using a 3-fold gradient, 11 dilution points dilution method, and POD was usedTM810 microplate pretreatment system for dilutingThe released compounds were added to 96-well plates with a final DMSO concentration of 0.5% per well. At 37 ℃ 5% CO2CO of2After incubation for 72h in the isothermal culture, 40. mu.L of luciferase assay reagent (Promega Bright-Glo) was added to each well, and after 5min, detection was performed using the chemiluminescence detection system (Envision). The results of the experiment were processed using GraphPad Prism software to calculate the EC50 for compound inhibition of each HCV replicon.
GT3a, GT4a and GT5a chimeric replicon activity assay: HCVGT1b/GT3a-NS3, HCVGT1b/GT4a-NS3 and HCVGT1b/GT5a-NS3 chimeric replicon RNA were transferred into Huh7 cells by electric shock method, and then the cells were seeded into 96-well plates containing the corresponding concentration of compound at a density of 10000 per well. Compound DMSO stock solution was diluted and added to 96 well assay plates to a final DMSO concentration of 0.5%. Cells were cultured for 72 hours at 37 ℃ with 5% CO 2. Luciferase luminescent substrate Bright-Glo was added to the wells, and 5 minutes later the Luminescence signal was measured using the chemiluminescence detection System Envision and the raw data (RLU) was used to calculate compound inhibitory activity. And introducing the inhibition percentage into GraphPad Prism software to carry out nonlinear fitting calculation to obtain a curve corresponding to the compound and an inhibition activity (EC50) value of the compound on the hepatitis C virus replicon.
The results of compound EC50 for HCV genotype 1a, genotype 1b, genotype 2a, genotype 3a, genotype 4a, and genotype 5a replicons are shown in table 2.
TABLE 2
The results of IC50 and EC50 of the compound on inhibition of HCV NS3/4A protease and inhibition of HCV replicon show that the compound of the application has better inhibitory activity on genotypes 1a, 1b, 2a, 3a, 4A and 5a, especially on genotype 2a, so that the compound can specifically inhibit HCV NS3/4A protease and has good antiviral effect.
3) Liver microsome stability test
Add 30. mu.L of 1.5. mu.M dosing solution to 1.5mL EP tube and add 150mL CH immediately3CN (containing internal standard), 15. mu.L NADPH solution (6mM) was added, mixed well and placed in a 4 ℃ freezer as the initial 0 spot sample, and each drug was done in duplicate. 30 μ L of 1.5 μ M dosing solution was added to a 96-well plate at positions set to different time points, two wells per drug were made in parallel, and pre-heated at 37 ℃ for 10 min. mu.L of NADPH solution (6mM) was added to the position set as the time point of 60min, and the reaction start timing was started. At the end of the incubation, 150mL ACN (containing internal standard) was added to the positions set for all time points. Then 15. mu.L of NADPH solution (6mM) was added to the position set to 0 min. Centrifuge at 4000rpm for 10 min. Taking out the supernatant, injecting and analyzing. And (4) analyzing by LC/MS/MS to obtain the ratio of the peak area of the sample to the peak area of the internal standard, regarding the drug content at the 0min point as 100%, and calculating the relative content of the drug at each time point. In terms of "Log [ drug concentration]The rate constants were obtained by plotting the "incubation time" to calculate the half-life and intrinsic clearance of the drug. The results of the experiment are shown in table 3.
TABLE 3 stability data of the compounds in human, rat and dog liver microsomes
Infinity is infinity
And (4) conclusion: the compounds of the invention have good stability in human, rat and dog liver microsomes.
4) Rat PK screening assay:
the test method comprises the following steps: dividing 250-sample 300g male SD rats into two groups, wherein each group comprises 3 male SD rats, respectively administering a compound to be detected to the male rats through veins and gavages, collecting blood for 8-9 time points within 24h, establishing a standard curve in a proper range according to the concentration of a sample, determining the concentration of the compound to be detected in a plasma sample in an MRM mode by using AB SCIEX API4000 or Agilent 6430 type LC-MS/MS, and carrying out quantitative analysis. Pharmacokinetic parameters were calculated according to the drug concentration-time curve using the WinNonLin 6.3 software non-compartmental model method.
And (4) conclusion: the compound of the invention has more obvious advantages in exposure amount and half-life than the reported compounds.
It will be evident to those skilled in the art that the present disclosure is not limited to the foregoing illustrative embodiments, but may be embodied in other specific forms without departing from the essential characteristics thereof. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive, reference being made to the appended claims, rather than to the foregoing embodiments, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example" or "some examples" or the like are intended to mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above do not necessarily refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that those skilled in the art can make changes, modifications, substitutions and alterations to the above embodiments without departing from the principle and spirit of the present invention, the scope of which is defined by the appended claims and their equivalents.

Claims (14)

1. A compound which is a compound represented by formula (I) or a stereoisomer, a tautomer, an enantiomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug of the compound represented by formula (I),
wherein: r1Is C6-10Aryl or C1-9A heteroaryl group;
R2and R3Each independently is H, F, Cl, Br, I, amino, hydroxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl or C1-9A heteroaryl group;
R4is H, deuterium or C1-6An alkyl group;
said C is1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl radical, C1-9Heteroaryl or amino is independently optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C1-6Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclic group, C6-10Aryl or C1-9Heteroaryl group is substituted.
2. The compound of claim 1, having the structure of formula (II):
or stereoisomers, tautomers, enantiomers, nitrogen oxides, hydrates, solvates, metabolites and pharmaceutically acceptable salts or prodrugs thereof.
3. The compound of claim 1 or 2, wherein R1Is phenyl or heteroaryl of 5 to 6 ring atoms; said phenyl or heteroaryl of 5-6 ring atoms is optionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3An alkenyl group,C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy, C1-3Alkylamino radical, C3-10Cycloalkyl radical, C2-10Heterocyclyl or C6-10Aryl group.
4. A compound according to claim 1 or 2, wherein R1Is phenyl, furyl, thienyl, thiazolyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, oxadiazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyridyl, quinolyl, indolyl or acridinyl, wherein R is1The group is optionally substituted with 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, trifluoromethyl, difluoroethyl, trifluoromethoxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, vinyl, ethynyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl or phenyl.
5. A compound according to claim 1 or 2, wherein R2And R3Each independently is H, F, Cl, Br, I, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, trifluoromethyl, trifluoromethoxy, tert-butyl, ethenyl, propenyl, ethynyl, propynyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl, phenyl, methylamino or ethylamino.
6. A compound according to claim 1 or 2, wherein R4Is H, deuterium, methyl, deuterated methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl.
7. The compound according to claim 1 or 2, wherein,
R1is heteroaryl of 5 to 6 ring atoms; wherein said 5-6 ring atoms of heteroarylOptionally substituted by 1,2,3 or 4 substituents selected from deuterium, hydroxy, amino, F, Cl, Br, I, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C2-3Alkenyl radical, C2-3Alkynyl, C1-3Alkoxy radical, C1-3Haloalkoxy or C1-3Substituted by alkylamino substituents;
R2is C1-6An alkyl group;
R3is C1-3An alkyl group;
R4is H or C1-3An alkyl group.
8. The compound according to claim 1 or 2, wherein the compound has one of the following structures or a stereoisomer, tautomer, enantiomer, nitrogen oxide, hydrate, solvate, metabolite, and pharmaceutically acceptable salt or prodrug thereof:
9. a pharmaceutical composition, wherein the pharmaceutical composition comprises a compound of any one of claims 1-8, further comprising a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle, or combination thereof.
10. The pharmaceutical composition of claim 9, further comprising an additional anti-HCV agent, wherein the anti-HCV agent is an interferon, ribavirin, interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, imiqimod, an inosine 5' -monophospate dehydrogenase inhibitor, amantadine, rimantadine, bavacizumab, CivacirTMWherein the interferon is a combination of interferon of Bupriravir, Tiarervir, erlotinib, daclatasvir, simeprevir, asunapprevir, vanioprevir, faldaprevir, paritaprevir, danoprevir, sovaprevir, gradoprevir, vedroprevevir, BZF-961, GS-9256, narloprevir, ANA975, ombitasvir, EDP239, PPI-668, velpatasvir, samatasvir, elbasvir, MK-8325, GSK-2336805, PPI-461, BI-2013335, ciloprevir, ACH-1095, VX-176985, IDX-375, VX-500, VX-813, PHX-6, PHX-136, IDX-316, modhrithrin, 376-PSI-1769, IBX-402985, IDX-375, VX-500, VX-40217, PHX-40217, VIX-4029, VITAB-102, VITAB-369, VITAB-102, VIALB-102, VITAB-90, VITAB-35, VITAB-90, VITAB-9.
11. The pharmaceutical composition of any one of claims 9-10, further comprising at least one HCV inhibitor.
12. The pharmaceutical composition according to claim 11, wherein the HCV inhibitor is for inhibiting the HCV replication process and/or inhibiting the function of HCV viral proteins; the HCV replication process comprises HCV entry, HCV uncoating, HCV translation, HCV replication, HCV assembly, or HCV release; the HCV viral protein is selected from metalloprotease, NS2, NS3, NS4A, NS4B, NS5A or NS5B, and an Internal Ribosome Entry Site (IRES) and inosine monophosphate dehydrogenase (IMPDH) required for HCV viral replication.
13. Use of a compound according to any one of claims 1 to 8 or a pharmaceutical composition according to any one of claims 9 to 12 in the manufacture of a medicament for inhibiting the HCV replication process and/or inhibiting the function of HCV viral proteins; the HCV replication process comprises HCV entry, HCV uncoating, HCV translation, HCV replication, HCV assembly, or HCV release; the HCV viral protein is selected from metalloprotease, NS2, NS3, NS4A, NS4B, NS5A or NS5B, and an Internal Ribosome Entry Site (IRES) and inosine monophosphate dehydrogenase (IMPDH) required for HCV viral replication.
14. Use of a compound of any one of claims 1-8 or a pharmaceutical composition of any one of claims 9-12 in the manufacture of a medicament for preventing, treating, or ameliorating HCV infection or hepatitis c disease.
HK17101942.8A 2015-02-13 2017-02-22 A compound as hepatitis c inhibitor and its use in medicament HK1228391B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201510080564.5 2015-02-13

Publications (2)

Publication Number Publication Date
HK1228391A1 true HK1228391A1 (en) 2017-11-03
HK1228391B HK1228391B (en) 2018-11-30

Family

ID=

Similar Documents

Publication Publication Date Title
CN106065009B (en) Application as the compound of hepatitis c inhibitor and its in drug
TWI610916B (en) Bridged ring compounds as hepatitis c virus (hcv) inhibitors and pharmaceuticals applications thereof
CN105884779B (en) Application as the compound of hepatitis c inhibitor and its in drug
CN104803989B (en) Application as the endocyclic compound of hepatitis c inhibitor and its in medicine
CN102863428B (en) Spiro-compounds taken as hepatitis c virus (HCV) inhibitor
CN103848821B (en) Spiro-compound, pharmaceutical composition and their purposes as hepatitis c inhibitor
CN103420991B (en) Application as the pyrrolidin derivatives of hepatitis c inhibitor and its in medicine
TWI703141B (en) Compounds as hepatitis c virus inhibitors and pharmaceutical uses thereof
JP2014534206A (en) Hepatitis C virus inhibitor
CN104003980B (en) It is used as the endocyclic compound and its pharmaceutical composition and purposes of hepatitis c inhibitor
CN103848819B (en) Spiro compound serving as hepatitis C inhibitor, drug composition and applications of spiro compound and drug composition in drugs
CN103709151B (en) Spiro-compound as hepatitis c inhibitor and its application in medicine
CN103880823B (en) Application as the spiro-compound of hepatitis c inhibitor and its in medicine
CN103848820A (en) Spiro compound serving as hepatitis C inhibitor and applications thereof in drugs
CN105384792B (en) Compounds as Hepatitis C Inhibitors and Their Use in Medicines
TWI600653B (en) Fused ring compounds as hepatitis c virus inhibitors and uses thereof
HK1228391A1 (en) A compound as hepatitis c inhibitor and its use in medicament
CN105085493A (en) Spiro compounds as hepatitis C inhibitors and their use in medicine
CN107074876B (en) Macrocyclic heterocyclic compound for inhibiting hepatitis C virus and preparation and application thereof
HK1228391B (en) A compound as hepatitis c inhibitor and its use in medicament
CN107266529A (en) One class suppresses the Large cyclic heterocyclic compound and its preparation and use of hepatitis C virus
CN105985355A (en) Fused tricyclic hepatitis virus inhibitor and application thereof