CN107531613B - Benzo-aliphatic ring-substituted alkylamine compound and application thereof - Google Patents

Benzo-aliphatic ring-substituted alkylamine compound and application thereof Download PDF

Info

Publication number
CN107531613B
CN107531613B CN201680022877.8A CN201680022877A CN107531613B CN 107531613 B CN107531613 B CN 107531613B CN 201680022877 A CN201680022877 A CN 201680022877A CN 107531613 B CN107531613 B CN 107531613B
Authority
CN
China
Prior art keywords
methyl
compound
reaction
amine
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201680022877.8A
Other languages
Chinese (zh)
Other versions
CN107531613A (en
Inventor
李剑
蓝乐夫
王友鑫
陈菲菲
魏汉文
蒋华良
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
East China University of Science and Technology
Shanghai Institute of Materia Medica of CAS
Original Assignee
East China University of Science and Technology
Shanghai Institute of Materia Medica of CAS
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 East China University of Science and Technology, Shanghai Institute of Materia Medica of CAS filed Critical East China University of Science and Technology
Publication of CN107531613A publication Critical patent/CN107531613A/en
Application granted granted Critical
Publication of CN107531613B publication Critical patent/CN107531613B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C209/00Preparation of compounds containing amino groups bound to a carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/01Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms
    • C07C211/26Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms of an unsaturated carbon skeleton containing at least one six-membered aromatic ring
    • C07C211/30Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms of an unsaturated carbon skeleton containing at least one six-membered aromatic ring the six-membered aromatic ring being part of a condensed ring system formed by two rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton

Abstract

The invention provides a benzo-aliphatic ring-substituted alkylamine compound and application thereof, and particularly relates to a compound shown as a formula I, or pharmaceutically acceptable salt thereof, a preparation method thereof, and application thereof (I) in preparation of staphylococcus aureus aureole synthesis inhibitor antibacterial drugs

Description

Benzo-aliphatic ring-substituted alkylamine compound and application thereof
Technical Field
The invention relates to the field of medicinal chemistry and medicinal treatment, in particular to a benzo-aliphatic ring-substituted alkylamine compound, a preparation method thereof and application thereof in preparing staphylococcus aureus aureola synthesis inhibitor antibacterial medicaments.
Background
Staphylococcus Aureus (SA) is the most common pathogen worldwide causing health care related infections. The gram-positive bacteria are the most common pathogenic bacteria causing human pyogenic infection, and can directly cause systemic infection such as local pyogenic infection, pneumonia, pseudomembranous enteritis, pericarditis, meningitis, septicemia, sepsis and the like. Infection with SA can be classified into hospital-acquired infection and community-acquired infection, the latter finding further increasing the potential biohazard of this pathogen and the possibility of causing an infectious outbreak.
At present, not only Methicillin-resistant Staphylococcus aureus (MRSA) but also MRSA with extremely high resistance (XDR) and complete resistance (TDR) have been found; even Vancomycin intermediate-resistant Staphylococcus aureus (VISA), Glycopeptide antibiotic intermediate-resistant Staphylococcus aureus (GISA), and Vancomycin-resistant Staphylococcus aureus (VRSA) have appeared. Because MRSA is resistant to various antibacterial drugs, the treatment of infection is difficult, the fatality rate is high, and serious difficulty is caused to clinical treatment due to the imminent 'no drug can be rescued'. According to the statistics of the Centers for Disease Control (CDC), about one hundred thousand people are hospitalized every year due to MRSA infection, and development of new antibacterial agents against MRSA infection is not always feasible.
In view of the above, there is an urgent need in the art to develop novel antibacterial drug action targets and novel antibacterial drugs against bacterial infections, especially antibacterial drugs against the synthesis of golden yellow pigments.
Disclosure of Invention
The invention aims to provide a benzo-aliphatic ring-substituted alkylamine compound, a preparation method thereof and application thereof in preparing staphylococcus aureus aureole synthesis inhibitor antibacterial drugs.
In a first aspect of the invention, there is provided a compound of formula I, or a pharmaceutically acceptable salt thereof:
Figure GPA0000247630360000031
in the formula:
R1is H or substituted or unsubstituted C1-C4A linear or branched alkyl group;
R2selected from the group consisting of: substituted or unsubstituted C1-C3Straight chain alkyl, substituted or unsubstituted C2-C6Straight or branched alkenyl, and substituted or unsubstituted C3-C6An alkynyl group;
R3selected from the group consisting of: substituted or unsubstituted C4-C7Cycloalkyl, substituted or unsubstituted C5-C6Heteroaryl ring radical, and substituted or unsubstituted C6-C10An aromatic ring group;
n is a positive integer of 1 to 3, and when n is 1, the substituent is located at the 1-position or the 2-position; when n is 2, the substituent is in the 2-position; when n is 3, the substituent is located at position 1;
m is an integer of 0 to 2;
p is an integer of 0 to 2;
wherein the substituents are selected from: c1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Perfluoroalkyl, halogen, nitro, and substituted or unsubstituted C1-C4Alkoxy, and the number of substituents is an integer of 1 to 4.
In another preferred embodiment, C is6-C10The aromatic ring group is phenyl or naphthalene ring group.
In another preferred embodiment, the compound is a compound of formula I-1:
Figure GPA0000247630360000032
wherein R is1、R2、R3N, m are as defined above.
In another preferred embodiment, R is1Is substituted or unsubstituted C1An alkyl group.
In another preferred embodiment, R is2Is substituted or unsubstituted C2-C6A linear alkenyl group.
In another preferred embodiment, R is2Is substituted or unsubstituted C3-C6An alkenyl group.
In another preferred embodiment, R is3Is substituted C4-C7Cycloalkyl, substituted C5-C6Heteroaromatic ring radical, substituted C6-C10An aromatic ring group, wherein the substituents are selected from: c1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Perfluoroalkyl, halogen, nitro, and substituted or unsubstituted C1-C4Alkoxy, and the number of substituents is an integer of 1 to 4.
In another preferred embodiment, the substituents are selected from the group consisting of: c1-C3Alkyl radical, C1-C3Perfluoroalkyl group, C1-C3Alkoxy, halogen, and nitro, and the number of substituents is an integer of 1 to 4.
In another preferred embodiment, the substituents are selected from the group consisting of: chlorine, bromine, methoxy, trifluoromethyl, and nitro, and the number of substituents is an integer of 1 to 2.
In another preferred embodiment, R is3Is a substituted phenyl group, wherein said substitution refers to having 1, 2 or 3 substituents selected from the group consisting of: c1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Perfluoroalkyl groups and halogens.
In another preferred embodiment, R is3Is substituted phenyl, wherein the substitution denotes halogen, -CF3
In another preferred embodiment, R is3Is monosubstituted phenyl。
In another preferred embodiment, R is3Is phenyl substituted by bromine or trifluoromethyl.
In another preferred embodiment, the compound of formula I is selected from the group consisting of:
Figure GPA0000247630360000041
wherein R is3Is as defined above.
In another preferred embodiment, the compound is selected from the group consisting of:
(E) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methyl-3-phenylprop-2-en-1-amine;
(E) -3- (4-bromophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-chlorophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-methoxyphenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-nitrophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-trifluoromethylphenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -N- [ (2, 3-dihydro-1H-inden-4-yl) methyl ] -N-methyl-3-phenylprop-2-en-1-amine;
(E) -N-methyl-3-phenyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-bromophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-chlorophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-methoxyphenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-nitrophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-bromophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -N-methyl-3-phenyl-N- [ (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methyl ] prop-2-en-1-amine;
(E) -N-methyl-3- (4-bromophenyl) -N- ((6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methyl) prop-2-en-1-amine; and
(E) -N-methyl-3- (4-chlorophenyl) -N- [ (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methyl ] prop-2-en-1-amine.
In a second aspect of the invention, there is provided a process for the preparation of a compound of formula I, comprising the steps of:
(i) reacting a compound of formula Z with a compound of formula Y in an inert solvent to obtain a compound of formula I,
Figure GPA0000247630360000054
in the formula, R1、R2、R3N, m, p are as defined above.
In another preferred embodiment, the compound of formula Z is selected from the group consisting of:
Figure GPA0000247630360000051
Figure GPA0000247630360000052
in another preferred embodiment, the compound of formula Y is
Figure GPA0000247630360000053
In another preferred embodiment, in the step (i), the solvent is selected from the group consisting of: n, N-dimethylformamide, acetonitrile, N-dimethylacetamide, or a combination thereof.
In another preferred embodiment, in step (i), the compounds Z and Y are reacted with a reagent selected from the group consisting of: potassium carbonate, sodium hydroxide, or combinations thereof.
In another preferred embodiment, in step (i), the reaction temperature is 10 to 50 ℃, preferably 20 to 30 ℃.
In another preferred embodiment, in the step (i), the reaction time is 5 to 20 hours, preferably 10 to 15 hours.
In another preferred example, after step (i-4), the method further comprises step (i-5): extracting the reaction system with an extractant selected from the group consisting of: ethyl acetate, dichloromethane, diethyl ether, or a combination thereof, washed with a detergent selected from the group consisting of: saturated brine, water, or a combination thereof, dried with a desiccant selected from the group consisting of: anhydrous sodium sulfate, anhydrous magnesium sulfate, or a combination thereof, filtering, concentrating, and chromatography.
In a third aspect of the present invention, there is provided a pharmaceutical composition comprising:
(1) a compound according to the first aspect of the invention, or a pharmaceutically acceptable salt thereof, as active ingredient; and
(2) a pharmaceutically acceptable carrier.
In another preferred embodiment, the pharmaceutical composition further comprises an additional antibiotic.
In a fourth aspect of the present invention, there is provided an antibacterial agent comprising:
(1) a compound according to the first aspect of the invention, or a pharmaceutically acceptable salt thereof;
(2) an additional antibiotic; and
(3) a pharmaceutically acceptable carrier.
In another preferred embodiment, the additional antibiotic is selected from the group consisting of: penicillins, cephalosporins, beta-lactamase inhibitors, aminoglycosides, glycopeptides, amides, macrolides, tetracyclines, sulfonamides, quinolones and nitroimidazoles.
In another preferred embodiment, the penicillins are selected from the group consisting of: penicillin, methicillin, amoxicillin, ampicillin, and meropenem.
In another preferred embodiment, the cephalosporins are selected from the group consisting of: cephalexin, cefotiam, cefadroxil, cefoxitin, cefazolin, cefradine, cefaclor, cefuroxime, cefpiramide, cefathiamidine.
In another preferred embodiment, the beta-lactamase inhibitor is selected from the group consisting of: amoxicillin and clavulanate potassium and amoxicillin and sulbactam.
In another preferred embodiment, the aminoglycoside is selected from the group consisting of: amikacin, gentamicin, amikacin, and streptomycin.
In another preferred embodiment, the amide is chloramphenicol.
In another preferred embodiment, the glycopeptide is selected from the group consisting of: vancomycin, norvancomycin, teicoplanin, and daptomycin.
In another preferred embodiment, the macrolide is selected from the group consisting of: erythromycin, erythromycin ethylsuccinate, kitasamycin, acetylkitasamycin azithromycin, roxithromycin and clarithromycin.
In another preferred embodiment, the tetracycline is selected from the group consisting of: tetracycline, doxycycline, oxytetracycline minocycline, and tigecycline.
In another preferred embodiment, the sulfanilamide is sulfamethoxazole or trimethoprim.
In another preferred embodiment, the quinolone is selected from the group consisting of: ciprofloxacin, ofloxacin, norfloxacin, levofloxacin, fleroxacin, enoxacin, lomefloxacin, gatifloxacin, sparfloxacin, moxifloxacin and pazufloxacin.
In another preferred embodiment, the nitroimidazoles are selected from the group consisting of: metronidazole, metronidazole benzoate, tinidazole and ornidazole.
In another preferred embodiment, the oxazolidinone is linezolid.
In a fifth aspect of the invention, there is provided a use of a compound according to the first aspect of the invention, or a pharmaceutically acceptable salt thereof, for the manufacture of a formulation or medicament for:
(1) inhibiting the synthesis of staphylococcus aureus and golden yellow pigment; and/or
(2) Inhibiting a key enzyme CrtN in the synthesis process of staphylococcus aureus and golden yellow pigment; and/or
(3) Inhibiting or killing staphylococcus aureus; and/or
(4) Treating infectious diseases caused by Staphylococcus aureus.
In another preferred embodiment, the staphylococcus aureus comprises MRSA.
In a sixth aspect of the invention, there is provided a non-therapeutic, in vitro method of inhibiting the synthesis of a staphylococcus aureus aureochrome or inhibiting staphylococcus aureus, comprising the steps of: contacting a compound according to the first aspect of the invention, or a pharmaceutically acceptable salt thereof, with staphylococcus aureus, thereby inhibiting synthesis of the s.
In another preferred embodiment, the concentration of the compound or pharmaceutically acceptable salt thereof is 0.5-10000nM, preferably 1-1000nM, more preferably 1-200nM, most preferably 1-100 nM.
In a seventh aspect of the present invention, there is provided a method for the prevention and/or treatment of a disease associated with staphylococcus aureus in a mammal, comprising administering to a mammal in need thereof a therapeutically effective amount of a compound according to the first aspect of the present invention, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to the second aspect of the present invention.
In another preferred embodiment, the staphylococcus aureus-related disease is selected from the group consisting of: a local pyogenic infection, pneumonia, pseudomembranous enteritis, pericarditis, sepsis, or sepsis.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1 shows a compound I of the present invention A6 data on the inhibitory activity (IC) against the resistant bacteria USA400MW 2(A), USA300LAC (B) and Mu50(C) golden yellow pigment synthesis50,nM)。
FIG. 2 shows Compound IA-2、IA-6、IC-2 and IC-6 IC for CrtN inhibitory activity of key enzyme in synthetic process of golden yellow pigment50Activity data (IC)50,μM)。
FIG. 3 shows Compound IAResults of enhanced Newman (A, survivval 36.2% vs 0.7%), USA400MW 2(B, survivval 11.7% vs 0.9%), USA300LAC (C, survivval 14.2% vs 0.4%) and Mu50(D, survivval 25.3% vs 2.1%) hydrogen peroxide killing experiments.
FIG. 4 shows Compound IAResults of human blood killing experiments on pairs-6 enhanced Newman (A, survivval 26.7% vs 1.3%), USA400MW 2(B, survivval 10.2% vs 0.9%), USA300LAC (C, survivval 12.1% vs 0.4%) and Mu50(D, survivval 16.1% vs 1.4%).
FIG. 5 shows Compound I of the present inventionA-6 results of decreasing the survival rate of staphylococcus aureus Newman in mouse kidney and heart.
FIG. 6 shows Compound I of the present inventionA-6 survival results for reduction of staphylococcus aureus USA400MW2 in mouse liver and kidney.
FIG. 7 shows Compound I of the present inventionA-6 results of decreasing survival of Staphylococcus aureus Mu50 in mouse liver and kidney.
FIG. 8 shows Compound I of the present inventionA-6 Final photograph of inhibition of the synthesis of golden yellow pigment, from left to right IAThe concentration of-6 was 50. mu.M, 10. mu.M, 5. mu.M, 2.5. mu.M, 1.25. mu.M, 0.625. mu.M, 0.3125. mu.M, 0. mu.M in this order.
FIG. 9 shows Compound I of the present inventionC-2 Final photograph of inhibition of the synthesis of golden yellow pigments, from left to right ICThe concentration of-2 was 50. mu.M, 10. mu.M, 5. mu.M, 2.5. mu.M, 1.25. mu.M, 0.625. mu.M, 0.3125. mu.M, 0. mu.M in this order.
Detailed Description
The present inventors have unexpectedly found for the first time through extensive and intensive studies. The present invention has been completed based on this finding.
Description of the terms
As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
Unless defined otherwise, the following terms used in the specification and claims have the meanings that are commonly understood by those skilled in the art. All patents, patent applications, and publications cited herein are incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular also includes the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or", "or" means "and/or" unless stated otherwise. Furthermore, the terms "comprising" or "including" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of, or" consisting of.
Definitions for the terms of the standardization sector can be found in the literature references including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4TH ED." Vols.A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terms used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH2O-is equivalent to-OCH2-。
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. For example, C1-C6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" refers to fluorine, chlorine, bromine or iodine.
"hydroxy" means an-OH group.
"nitro" means-NO2
In this application, the term "alkyl" as a group or as part of another group (such as used in halo-substituted alkyl and the like groups) refers to a fully saturated straight or branched hydrocarbon chain radical consisting only of carbon and hydrogen atoms, having, for example, 1 to 7 carbon atoms, and attached to the rest of the molecule by a single bond, including, for example, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, and the like.
In the present application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon and hydrogen atoms, containing at least one double bond, having, for example, 2 to 8 (preferably 2 to 6) carbon atoms, and being connected to the rest of the molecule by a single bond, such as, but not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In this application, the term "alkynyl" as a group or part of another group refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, optionally containing at least one double bond, having, for example, 2 to 6 carbon atoms, and being attached to the rest of the molecule by a single bond, such as, but not limited to, ethynyl, prop-1-ynyl, but-1-ynyl, pent-1-en-4-ynyl, and the like.
In this application, the term "aryl" as a group or as part of another group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, an aryl group may be a monocyclic, bicyclic, tricyclic or higher polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the aryl group is attached to the remainder of the molecule by a single bond via an atom on the aromatic ring. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthryl, phenanthryl, fluorenyl, and the like.
In this application, the term "heteroaryl" as a group or part of another group means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring. Unless otherwise specifically indicated in the specification, a heteroaryl group may be a monocyclic, bicyclic, tricyclic or higher ring system, and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl group is attached to the rest of the molecule by a single bond via an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. For the purposes of the present invention, heteroaryl is preferably a stable 5-to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or a 5-to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindolyl, purinyl, quinolyl, isoquinolyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, orthophenanthrolidinyl, isoxazolyl, phenoxazinyl, phenothiazinyl, 4, 5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, pyridinyl, and the like, [1, 2, 4] triazolo [4, 3-b ] pyridazine, [1, 2, 4] triazolo [4, 3-a ] pyrazine, [1, 2, 4] triazolo [4, 3-c ] pyrimidine, [1, 2, 4] triazolo [4, 3-a ] pyridine, imidazo [1, 2-b ] pyridazine, imidazo [1, 2-a ] pyrazine and the like.
In this application, "optionally" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted and unsubstituted aryl groups. The "optionally" substituents described in the claims and the description section of the present invention are selected from alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, cyano, nitro, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl.
The terms "moiety," "structural moiety," "chemical moiety," "group," "chemical group" as used herein refer to a specific fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded in or attached to a molecule.
When the compounds of the present invention contain olefinic double bonds, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
In the present application, the term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts with inorganic or organic acids which retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formates, acetates, 2-dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
In the present application, a "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
As used herein, "pharmaceutically acceptable excipient" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizing agent, isotonic agent, solvent, or emulsifying agent that is approved by the relevant governmental regulatory agency for human or livestock use.
The terms "preventing," "prevention," and "prevention" as used herein include reducing the likelihood of occurrence or worsening of a disease or disorder in a patient.
As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
The terms "administering," "administration," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, The pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The terms "drug combination", "administering other treatment", "administering other therapeutic agent" and the like as used herein refer to a drug treatment obtained by mixing or combining more than one active ingredient, including fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities.
A compound of formula I
The invention relates to a compound shown in formula I, or a pharmaceutically acceptable salt thereof:
Figure GPA0000247630360000131
in the formula:
R1is H or substituted or unsubstituted C1-C4A linear or branched alkyl group;
R2selected from the group consisting of: substituted or unsubstituted C1-C3Straight chain alkyl, substituted or unsubstituted C2-C6Straight or branched alkenyl, and substituted or unsubstituted C3-C6An alkynyl group;
R3selected from the group consisting of: substituted or unsubstituted C4-C7Cycloalkyl, substituted or unsubstituted C5-C6Heteroaryl ring radical, and substituted or unsubstituted C6-C10An aromatic ring group;
n is a positive integer of 1 to 3, and when n is 1, the substituent is located at the 1-position or the 2-position; when n is 2, the substituent is in the 2-position; when n is 3, the substituent is located at position 1;
m is an integer of 0 to 2;
p is an integer of 0 to 2;
wherein the substituents are selected from: c1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Perfluoroalkyl, halogen, nitro, and substituted or unsubstituted C1-C4Alkoxy, and the number of substituents is an integer of 1 to 4.
In another preferred embodiment, C is6-C10The aromatic ring radical being benzeneA phenyl group or a naphthyl group.
In another preferred embodiment, the compound is a compound of formula I-1:
Figure GPA0000247630360000132
wherein R is1、R2、R3N, m are as defined above.
In another preferred embodiment, R is1Is substituted or unsubstituted C1An alkyl group.
In another preferred embodiment, R is2Is substituted or unsubstituted C2-C6A linear alkenyl group.
In another preferred embodiment, R is2Is substituted or unsubstituted C3-C6An alkenyl group.
In another preferred embodiment, R is3Is substituted C4-C7Cycloalkyl, substituted C5-C6Heteroaromatic ring radical, substituted C6-C10An aromatic ring group, wherein the substituents are selected from: c1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Perfluoroalkyl, halogen, nitro, and substituted or unsubstituted C1-C4Alkoxy, and the number of substituents is an integer of 1 to 4.
In another preferred embodiment, the substituents are selected from the group consisting of: c1-C3Alkyl radical, C1-C3Perfluoroalkyl group, C1-C3Alkoxy, halogen, and nitro, and the number of substituents is an integer of 1 to 4.
In another preferred embodiment, the substituents are selected from the group consisting of: chlorine, bromine, methoxy, trifluoromethyl, and nitro, and the number of substituents is an integer of 1 to 2.
In another preferred embodiment, R is3Is a substituted phenyl group, wherein said substitution refers to having 1, 2 or 3 substituents selected from the group consisting of: c1-C6Alkyl radical, C1-C6Haloalkyl, C1-C6Perfluoroalkyl groups and halogens.
In another preferred embodiment, R is3Is substituted phenyl, wherein the substitution denotes halogen, -CF3
In another preferred embodiment, R is3Is monosubstituted phenyl.
In another preferred embodiment, R is3Is phenyl substituted by bromine or trifluoromethyl.
In another preferred embodiment, the compound of formula I is selected from the group consisting of:
Figure GPA0000247630360000141
wherein R is3Is as defined above.
Preparation method
The process of the present invention for preparing a compound of formula I, comprising the steps of:
(i) reacting a compound of formula Z with a compound of formula Y in an inert solvent to obtain a compound of formula I,
Figure GPA0000247630360000142
in the formula, R1、R2、R3N, m, p are as defined above.
In another preferred embodiment, the compound of formula Z is selected from the group consisting of:
Figure GPA0000247630360000151
Figure GPA0000247630360000152
in another preferred embodiment, the compound of formula Y is
Figure GPA0000247630360000153
In another preferred embodiment, in the step (i), the solvent is selected from the group consisting of: n, N-dimethylformamide, acetonitrile, N-dimethylacetamide, or a combination thereof.
In another preferred embodiment, in step (i), the compounds Z and Y are reacted with a reagent selected from the group consisting of: potassium carbonate, sodium hydroxide, or combinations thereof.
In another preferred embodiment, in step (i), the reaction temperature is 10 to 50 ℃, preferably 20 to 30 ℃,
in another preferred embodiment, in the step (i), the reaction time is 5 to 20 hours, preferably 10 to 15 hours.
In another preferred example, after step (i-4), the method further comprises step (i-5): extracting the reaction system with an extractant selected from the group consisting of: ethyl acetate, dichloromethane, diethyl ether, or a combination thereof, washed with a detergent selected from the group consisting of: saturated brine, water, or a combination thereof, dried with a desiccant selected from the group consisting of: anhydrous sodium sulfate, anhydrous magnesium sulfate, or a combination thereof, filtering, concentrating, and chromatography.
Formula IAPreparation of the Compounds
Figure GPA0000247630360000154
In the formula R3The meaning of (a) is the same as previously described.
1) Slowly dropwise adding an anhydrous tetrahydrofuran solution of the indane-5-formic acid into an anhydrous tetrahydrofuran suspension of lithium aluminum hydride at-78 ℃ under the protection of nitrogen, and reacting at 20-30 ℃ overnight after dropwise adding. And sequentially adding water and a 15% sodium hydroxide aqueous solution into the reaction system, quenching the mixture with water for reaction, directly adding anhydrous sodium sulfate into the reaction system, drying, filtering and concentrating to obtain the indan-5-methanol (intermediate II).
2) Dissolving the intermediate II in anhydrous ether, adding phosphorus tribromide under nitrogen protection and ice bath, and reacting at 20-30 ℃ for 10-20 hours. After the reaction is finished, pouring the reaction system into an ice saturated sodium bicarbonate solution, extracting with ethyl acetate for three times, washing with saturated saline, drying with anhydrous magnesium sulfate, filtering, and concentrating at 30 ℃ to obtain the 5-bromomethyl indane (intermediate III).
3) And slowly adding the ethanol solution of the intermediate III into the ethanol solution of methylamine, and reacting for 10-20 hours at the temperature of 20-30 ℃. After the reaction is finished, concentrating to obtain the N-methyl-indanyl-5-methylamine (intermediate IV).
4) Reacting (E) -3-R3Acrolein is dissolved in methanol, sodium borohydride is added in portions under ice bath, and the reaction is carried out for 10 to 30 minutes at room temperature. Concentrating, extracting the residue with ethyl acetate for three times, washing with saturated brine, drying over anhydrous magnesium sulfate, filtering, and concentrating to obtain (E) -3-R3-propenol (intermediate V).
5) Dissolving the intermediate V in anhydrous ether, adding phosphorus tribromide under nitrogen protection and ice bath, and reacting at 20-30 ℃ for 10-20 hours. After the reaction, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to give (E) -3-R3-propylene bromide (intermediate VI).
6) Adding the intermediate IV, the intermediate VI and potassium carbonate into N, N-dimethylformamide, and reacting for 10-20 hours at 20-30 ℃. After the reaction is finished, adding water into the reaction system, extracting for three times by using ethyl acetate, washing by using saturated salt water, drying by using anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography separation on residues to obtain the compound shown in the formula IAThe compound (E) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methyl-3-R3-prop-2-en-1-amine.
Formula IBPreparation of the Compounds
Figure GPA0000247630360000161
In the formula R3The meaning of (a) is the same as previously described.
1) Dissolving 4-aminoindane in concentrated hydrochloric acid, dropwise adding ammonium nitrite aqueous solution at 0 ℃, stirring for 15 minutes, slowly adding potassium iodide aqueous solution, and reacting at 20-30 ℃ overnight. After the reaction, the mixture is extracted three times by ethyl acetate, washed by saturated saline solution, dried by anhydrous magnesium sulfate, filtered, concentrated and the residue is separated by column chromatography to obtain the 4-iodine-2, 3-dihydro-1H-indene (intermediate VII).
2) Adding the intermediate VII and cuprous cyanide into N, N-dimethylformamide, and heating and refluxing for 10-20 hours. Cooling to room temperature, adding concentrated ammonia water into the system, extracting with ethyl acetate for three times, washing with saturated salt water, drying with anhydrous magnesium sulfate, filtering, concentrating, and separating the residue by column chromatography to obtain 4-cyano-2, 3-dihydro-1H-indene (intermediate VIII).
3) And slowly dropwise adding the anhydrous tetrahydrofuran solution of the intermediate VIII into the anhydrous tetrahydrofuran suspension of the lithium aluminum hydride at the temperature of minus 78 ℃ under the protection of nitrogen, and reacting at the temperature of 20-30 ℃ overnight after dropwise adding. Adding water and 15% sodium hydroxide aqueous solution into the reaction system in sequence, quenching the reaction with water, directly adding anhydrous sodium sulfate into the reaction system, drying, filtering and concentrating to obtain (2, 3-dihydro-1H-inden-4-yl) methylamine (intermediate IX).
4) Dissolving the intermediate IX in tetrahydrofuran, adding sodium hydroxide, stirring for 5-10 min, and slowly adding a tetrahydrofuran solution of di-tert-butyl dicarbonate under ice bath. Stirring and reacting for 1-3 hours at the temperature of 0-30 ℃. Filtration, concentration and column chromatography of the residue gave tert-butyl ((2, 3-dihydro-1H-inden-4-yl) methyl) carbamate (intermediate X).
5) And slowly dripping the anhydrous tetrahydrofuran solution of the intermediate X into the anhydrous tetrahydrofuran suspension of the lithium aluminum hydride at the temperature of 0 ℃ under the protection of nitrogen, and heating and refluxing for reaction for 10-20 hours after the dripping is finished. After the reaction is finished, water and 15% sodium hydroxide aqueous solution are sequentially added into the reaction system, water quenching reaction is carried out, anhydrous sodium sulfate is directly added into the reaction system for drying, filtering and concentrating to obtain the 1- (2, 3-dihydro-1H-indene-4-yl) -N-methyl methylamine (an intermediate XI).
6) Reacting (E) -3-R3Acrolein is dissolved in methanol, sodium borohydride is added in portions under ice bath, and the reaction is carried out for 10 to 30 minutes at room temperature. Concentrating, extracting the residue with ethyl acetate for three times, washing with saturated saline, drying over anhydrous magnesium sulfate, filtering, and concentrating to obtain (E) -3-R3-propenol (intermediate V).
7) Dissolving the intermediate V in anhydrous ether, adding phosphorus tribromide under nitrogen protection and ice bath, and reacting at 20-30 ℃ for 10-20 hours. After the reaction is finished, pouring ice saturated hydrogen carbonate into the reaction systemExtracting with ethyl acetate for three times, washing with saturated salt water, drying with anhydrous magnesium sulfate, filtering, and concentrating at 30 deg.C to obtain (E) -3-R3-propylene bromide (intermediate VI).
8) Adding the intermediate XI, the intermediate VI and potassium carbonate into N, N-dimethylformamide, and reacting at 20-30 ℃ for 10-20 hours. After the reaction is finished, adding water into the reaction system, extracting for three times by using ethyl acetate, washing by using saturated salt water, drying by using anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography separation on residues to obtain the compound shown in the formula IBThe compound (E) -N- [ (2, 3-dihydro-1H-inden-4-yl) methyl]-N-methyl-3-R3-prop-2-en-1-amine.
Formula ICPreparation of the Compounds
Figure GPA0000247630360000181
In the formula R3The meaning of (a) is the same as previously described.
1) Slowly dripping an anhydrous tetrahydrofuran solution of 5,6,7, 8-tetrahydro-2-naphthoic acid into an anhydrous tetrahydrofuran suspension of lithium aluminum hydride at the temperature of-78 ℃ under the protection of nitrogen, and reacting at the temperature of 20-30 ℃ overnight after dripping. Adding water and 15% sodium hydroxide aqueous solution into the reaction system in sequence, quenching the reaction with water, directly adding anhydrous sodium sulfate into the reaction system, drying, filtering and concentrating to obtain (5,6,7, 8-tetrahydronaphthalene-2-yl) methanol (intermediate XII).
2) Dissolving the intermediate XII in anhydrous ether, adding phosphorus tribromide in nitrogen protection ice bath, and reacting at 20-30 deg.C for 10-20 hr. After the reaction, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to obtain 6- (bromomethyl) -1, 2,3, 4-tetrahydronaphthalene (intermediate XIII).
3) And slowly adding the ethanol solution of the intermediate XIII into the ethanol solution of methylamine, and reacting for 10-20 hours at 20-30 ℃. After the reaction is finished, N-methyl-1- (5,6,7, 8-tetrahydronaphthalene-2-yl) methylamine (intermediate XIV) is obtained by concentration.
4) Reacting (E) -3-R3-CThe olefine aldehyde is dissolved in methanol, sodium borohydride is added in batches under ice bath, and the reaction lasts for 10 to 30 minutes at room temperature. Concentrating, extracting the residue with ethyl acetate for three times, washing with saturated saline, drying over anhydrous magnesium sulfate, filtering, and concentrating to obtain (E) -3-R3-propenol (intermediate V).
5) Dissolving the intermediate V in anhydrous ether, adding phosphorus tribromide under nitrogen protection and ice bath, and reacting at 20-30 ℃ for 10-20 hours. After the reaction, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to give (E) -3-R3-propylene bromide (intermediate VI).
6) Adding the intermediate XIV, the intermediate VI and potassium carbonate into N, N-dimethylformamide, and reacting at 20-30 ℃ for 10-20 hours. After the reaction is finished, adding water into the reaction system, extracting for three times by using ethyl acetate, washing by using saturated salt water, drying by using anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography separation on residues to obtain the compound shown in the formula ICCompound (E) -N-methyl-3-R3-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-amine.
Formula IDPreparation of the Compounds
Figure GPA0000247630360000191
In the formula R3The meaning of (a) is the same as previously described.
1) Dissolving 2-hydroxy-5-methylbenzaldehyde in N, N-dimethylformamide, adding potassium carbonate, stirring for 15 minutes under the protection of nitrogen, adding iodoethane, and stirring for 12 hours at room temperature. After the reaction, water was added to the reaction system, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was separated by column chromatography to give 2-ethoxy-5-methylbenzaldehyde (intermediate XV).
2) Adding (4-methoxy-4-oxobutyl) triphenyl phosphine bromide and hexamethyl phosphoric triamide into anhydrous tetrahydrofuran, adding lithium bis (trimethylsilyl) amide under the condition of argon protection at 0 ℃, reacting for 2 hours, then placing the system at minus 78 ℃, adding an intermediate XV, and reacting for 1 hour. After the reaction is finished, saturated ammonium chloride is used for quenching, dilute hydrochloric acid is used for adjusting the system to be acidic, ethyl acetate is used for extraction for 4 times, anhydrous magnesium sulfate is used for drying, filtration and concentration are carried out, and the residue is separated by column chromatography to obtain (E) -methyl-5- (2-ethoxy-5-methylphenyl) pent-4-enoic acid ethyl ester (intermediate XVI).
3) And dissolving the intermediate XVI in tetrahydrofuran, adding an aqueous solution dissolved with lithium hydroxide, and reacting at 20-30 ℃ for 6-10 hours. After the reaction is finished, dilute hydrochloric acid is added to adjust the system to be acidic, then ethyl acetate is used for extraction for three times, anhydrous magnesium sulfate is used for drying, filtration and concentration are carried out, and the residue is separated by column chromatography to obtain (E) -5- (2-ethoxy-5-methylphenyl) pent-4-enoic acid (intermediate XVII).
4) And dissolving the intermediate XVII in methanol, adding a palladium-carbon catalyst, and carrying out hydrogen reduction reaction at the temperature of 20-30 ℃ for 8-12 hours. After the reaction, the palladium carbon is removed by suction filtration, the filtrate is retained and concentrated to obtain 5- (2-ethoxy-5-methylphenyl) pentanoic acid (intermediate XVIII).
5) The intermediate XVIII was dissolved in Eton's reagent (7.5% solution of phosphorus pentoxide in methanesulfonic acid) under ice-bath, and then reacted at 20-30 ℃ for 10-14 hours. After the reaction, the system was poured into an ice-water mixture, followed by extraction with dichloromethane three times, drying over anhydrous magnesium sulfate, filtration, concentration, and column chromatography of the residue to give 1-ethoxy-4-methyl-6, 7,8, 9-tetrahydro-5H-benzo [7] annulen-5-one (intermediate XIX).
6) Dissolving the intermediate XIX, sodium cyanoborohydride and zinc iodide in 1, 2-dichloroethane, heating and refluxing for 8-12 hours, pouring the system into a sand core funnel containing diatomite for suction filtration after the reaction is finished, concentrating the filtrate, and carrying out column chromatography separation on the residue to obtain the 1-ethoxy-4-methyl-6, 7,8, 9-tetrahydro-5H-benzo [7] annulene (intermediate XX).
7) Adding the intermediate XX and pyridinium chlorochromate into anhydrous acetonitrile, and carrying out reflux reaction for 8-12 hours. After the reaction, the system was concentrated, and then water and methylene chloride were added to the residue to extract, the organic layer was collected, dried over anhydrous magnesium sulfate, filtered, concentrated, and the residue was subjected to column chromatography to obtain 4-ethoxy-6, 7,8, 9-tetrahydro-5H-benzo [7] annulene-1-carbaldehyde (intermediate XXI).
8) Adding the intermediate XXI and aluminum chloride into dichloromethane, and reacting at room temperature for 12-36 h. After the reaction, a little water and a saturated sodium chloride solution were added to the system, followed by extraction with dichloromethane three times, collection of the organic layer, drying over anhydrous magnesium sulfate, filtration, concentration, and column chromatography of the residue to give 4-hydroxy-6, 7,8, 9-tetrahydro-5H-benzo [7] annulene-1-carbaldehyde (intermediate XXII).
9) Intermediate XXII and pyridine are dissolved in dichloromethane, trifluoromethanesulfonic anhydride is added dropwise in an ice bath, the mixture is stirred for 2 hours at 20-30 ℃, washed with water 2 times, washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated to give 4-formyl-6, 7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl trifluoromethanesulfonate (intermediate XXIII).
10) Triethylamine was dissolved in N, N-dimethylformamide, 98% formic acid was added dropwise, followed by intermediate XXIII, palladium acetate and 1, 1' -bis (diphenylphosphino) ferrocene. Reacting at 80 deg.C for 15 min, cooling to room temperature, filtering to remove solid, adding water to the filtrate, extracting with ethyl acetate three times, drying over anhydrous magnesium sulfate, filtering, concentrating, and subjecting the residue to column chromatography to obtain 6,7,8, 9-tetrahydro-5H-benzo [7] annulene-1-carbaldehyde (intermediate XXIV).
11) Dissolving the intermediate XXIV in methanol, adding sodium borohydride in batches under ice bath, and reacting for 10-30 minutes at room temperature. Concentration, extraction of the residue with ethyl acetate three times, washing with saturated brine, drying over anhydrous magnesium sulfate, filtration and concentration gave (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methanol (intermediate XXV).
12) Dissolving the intermediate XXV in anhydrous ether, adding phosphorus tribromide under nitrogen protection and ice bath, and reacting at 20-30 ℃ for 10-20 hours. After the reaction, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to give 1- (bromomethyl) -6, 7,8, 9-tetrahydro-5H-benzo [7] annulene (intermediate XXVI).
13) And slowly adding the ethanol solution of the intermediate XXVI into the ethanol solution of methylamine, and reacting for 10-20 hours at the temperature of 20-30 ℃. After the reaction is finished, the reaction product is concentrated to obtain N-methyl-1- (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methylamine (an intermediate XXVII).
14) Reacting (E) -3-R3Acrolein is dissolved in methanol, sodium borohydride is added in portions under ice bath, and the reaction is carried out for 10 to 30 minutes at room temperature. Concentrating, extracting the residue with ethyl acetate for three times, washing with saturated saline, drying over anhydrous magnesium sulfate, filtering, and concentrating to obtain (E) -3-R3-propenol (intermediate V).
15) Dissolving the intermediate V in anhydrous ether, adding phosphorus tribromide under nitrogen protection and ice bath, and reacting at 20-30 ℃ for 10-20 hours. After the reaction, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to give (E) -3-R3-propylene bromide (intermediate VI).
16) Adding the intermediate XXVII, the intermediate VI and potassium carbonate into N, N-dimethylformamide, and reacting at 20-30 ℃ for 10-20 hours. After the reaction is finished, adding water into the reaction system, extracting for three times by using ethyl acetate, washing by using saturated salt water, drying by using anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography separation on residues to obtain the compound shown in the formula IDCompound (E) -N-methyl-3-R3-N- [ (6,7,8, 9-tetrahydro-5H-benzo [7]]Rotalen-1-yl) methyl]Prop-2-en-1-amine.
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.
As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of, or" consisting of.
The main advantages of the invention are:
1. provides a compound shown as a formula I.
2. The inhibitor has high inhibitory activity to golden yellow pigment, and takes a key enzyme CrtN in the synthetic process of the golden yellow pigment as an action target.
3. Pharmaceutical compositions for treating diseases associated with staphylococcus aureus activity are provided.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are percentages and parts by weight. Unless otherwise indicated, are in units of mass (grams).
The test materials and reagents used in the following examples are commercially available without specific reference.
The strain is as follows:
the test strains in the examples of the present invention were:
the experimental strains belong to the conventional strains,
the Newman genus is an antibiotic-sensitive strain,
USA300Lac USA400MW2 Mu50 belongs to MRSA.
EXAMPLE 1 preparation of indan-5-methanol (intermediate II)
Figure GPA0000247630360000221
250 mg of indan-5-formic acid in 10ml of anhydrous tetrahydrofuran is slowly and dropwise added into 120 mg of lithium aluminum hydride in 1ml of anhydrous tetrahydrofuran suspension at minus 78 ℃ under the protection of nitrogen, and the reaction is carried out overnight at 25 ℃ after the dropwise addition. To the reaction system were added 0.25 ml of water, 0.25 ml of 15% aqueous sodium hydroxide solution and 0.25 ml of water in this order to quench the reaction, and anhydrous sodium sulfate was directly added to the reaction system to dry, filter and concentrate to obtain the title compound in a yield of 93% as 212 mg of a white solid.
1H-NMR(400MHz,CDCl3)δ7.24(dd,J=14.3,6.5Hz,2H),7.13(d,J=7.6Hz,1H),4.65(s,2H),2.91(td,J=7.5,2.4Hz,4H),2.17-1.98(m,2H).
EXAMPLE 2 preparation of 5-bromomethylindane (intermediate III)
Figure GPA0000247630360000222
212 mg of intermediate II are dissolved in 20ml of anhydrous ether, 50. mu.l of phosphorus tribromide are added under nitrogen protection in ice bath, and the reaction is carried out overnight at 25 ℃. After the reaction was completed, the reaction system was poured into 10ml of ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to obtain the title compound in 269 mg of white solid with a yield of 89%.
1H-NMR(400MHz,CDCl3)δ7.26(s,1H),7.17(q,J=7.8Hz,2H),4.51(s,2H),2.89(t,J=6.5Hz,4H),2.17-1.99(m,2H).
EXAMPLE 3 preparation of N-methyl-indanyl-5-methanamine (intermediate IV)
Figure GPA0000247630360000231
269 mg of intermediate III in 10ml of ethanol are slowly added to 10ml of 30% methylamine in ethanol and reacted at 25 ℃ for 1 overnight. After the reaction was complete, concentration afforded the title compound in 90% yield as 184 mg of a colorless oil.
1H-NMR(400MHz,CDCl3)δ7.26-7.16(m,2H),7.12(d,J=7.7Hz,1H),4.03(s,2H),2.90-2.77(m,4H),2.59(s,3H),2.00(p,J=7.4Hz,2H).
EXAMPLE 4 preparation of (E) -3-phenylprop-2-en-1-ol (intermediate V-1)
Figure GPA0000247630360000232
100 mg of (E) -3-phenylacrolein are dissolved in 10ml of methanol, 28 mg of sodium borohydride are added in portions in ice bath and reacted for 15 minutes at room temperature. Concentration, water was added to the residue, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered and concentrated to give the title compound, 99 mg of oil, which was directly used for the next reaction in 98% yield.
EXAMPLE 5 preparation of (E) -1-phenyl-3-bromo-propene (intermediate VI-1)
Figure GPA0000247630360000233
370 mg of intermediate V-1 are dissolved in 20ml of anhydrous ether, 85. mu.l of phosphorus tribromide are added under nitrogen protection in ice bath, and the reaction is carried out overnight at room temperature. After the reaction was completed, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to obtain the title compound as 460 mg of a white solid with a yield of 85%.
1H-NMR(400MHz,CDCl3)δ7.40-7.26(m,5H),6.64(d,J=15.6Hz,1H),6.40(dt,J=15.6,7.8Hz,1H),4.15(dd,J=0.9,7.8Hz,2H)
EXAMPLE 6 preparation of (E) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methyl-3-phenylprop-2-en-1-amine (Compound I)A-1)
Figure GPA0000247630360000234
178 mg of intermediate IV, 197 mg of intermediate VI-1 and 167 mg of potassium carbonate were added to 20ml of N, N-dimethylformamide and reacted overnight at room temperature. After the reaction was completed, water was added to the reaction system, and extraction was performed three times with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was separated by column chromatography to give the title compound as a colorless oil of 120 mg in 43% yield. For purification, the compound is dissolved in 1ml of ethyl acetate, hydrogen chloride gas is introduced for one minute to prepare hydrochloride, the solvent is evaporated, 1/100 petroleum ether/ethyl acetate mixed solvent is added, white hydrochloride solid is separated out, and the compound IA-1 hydrochloride is obtained by suction filtration and washing. 1H-NMR is data on the hydrochloride form.
1H-NMR(400MHz,MeOD)δ7.60-7.48(m,2H),7.48-7.32(m,5H),7.28(d,J=7.8Hz,1H),6.93(d,J=15.8Hz,1H),6.41-6.30(m,1H),4.46(d,J=13.0Hz,1H),4.24(d,J=12.9Hz,1H),4.05(dd,J=13.2,7.2Hz,1H),3.89(dd,J=13.0,7.8Hz,1H),2.97(dd,J=13.2,7.0Hz,4H),2.81(s,3H),2.21-2.08(m,2H);HRMS(ESI)m/z calcd for C20H24N(M+H)+278.1909,found 278.1909。
EXAMPLE 7 preparation of (E) -3- (4-bromophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methylprop-2-en-1-amine (Compound I)A-2)
Figure GPA0000247630360000241
The required starting materials, reagents and preparation were the same as in example 4-6, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-bromophenyl) -acrolein, and 185 mg of the title compound was obtained in 52% yield as an oil. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.56(d,J=8.3Hz,2H),7.46(d,J=8.3Hz,2H),7.40(s,1H),7.36(d,J=7.7Hz,1H),7.29(d,J=7.5Hz,1H),6.89(d,J=15.8Hz,1H),6.46-6.31(m,1H),4.46(d,J=12.9Hz,1H),4.25(d,J=13.1Hz,1H),4.04(dd,J=13.1,6.9Hz,1H),3.97-3.84(m,1H),2.98(td,J=13.9,7.5Hz,4H),2.87-2.72(m,3H),2.22-2.07(m,2H).HRMS(ESI)m/z calcd for C20H23BrN(M+H)+356.1014,found 356.1017。
EXAMPLE 8 preparation of (E) -3- (4-chlorophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methylprop-2-en-1-amine (Compound I)A-3)
Figure GPA0000247630360000251
The required starting materials, reagents and preparation were the same as in example 4-6, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-chlorophenyl) -acrolein, and 171 mg of the title compound was obtained as a colorless oil in a yield of 55%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.53(d,J=8.3Hz,2H),7.49-7.32(m,4H),7.30(s,1H),6.91(d,J=15.7Hz,1H),6.50-6.29(m,1H),4.46(d,J=12.7Hz,1H),4.25(d,J=12.4Hz,1H),4.16-4.03(m,1H),3.95-3.84(m,1H),2.97(dd,J=13.5,6.8Hz,4H),2.81(s,3H),2.23-2.05(m,2H);HRMS(ESI)m/z calcd for C20H23ClN(M+H)+312.1519,found 312.1507。
EXAMPLE 9 preparation of (E) -3- (4-methoxyphenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methylprop-2-en-1-amine (Compound I)A-4)
Figure GPA0000247630360000252
The required starting materials, reagents and preparation were the same as in example 4-6, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-methoxyphenyl) -acrolein, and 122 mg of the title compound was obtained in an oily state with a yield of 40%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.47(d,J=8.4Hz,2H),7.44-7.32(m,3H),7.29(d,J=7.1Hz,1H),7.25(d,J=7.6Hz,1H),6.86(d,J=15.6Hz,1H),6.21(dt,J=15.3,7.5Hz,1H),4.44(dd,J=16.6,9.8Hz,1H),4.22(d,J=13.0Hz,1H),4.01(dd,J=12.9,6.9Hz,1H),3.87(d,J=7.4Hz,1H),3.84-3.80(m,3H),2.97(dd,J=12.9,6.7Hz,4H),2.79(s,3H),2.12(dd,J=14.6,7.3Hz,2H).;HRMS(ESI)m/z calcd for C21H26NO(M+H)+308.2014,found 308.2017。
Example 10 preparation of (E) -3- (4-Nitrophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methylprop-2-en-1-amine (Compound I)A-5)
Figure GPA0000247630360000261
The required starting materials, reagents and preparation were the same as in example 4-6, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-nitrophenyl) -acrolein, and 206 mg of the title compound was obtained as an oil in a yield of 64%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ8.27(d,J=8.4Hz,2H),7.78(d,J=8.4Hz,2H),7.42(d,J=7.6Hz,1H),7.35(t,J=9.2Hz,1H),7.30(d,J=7.7Hz,1H),7.04(d,J=15.7Hz,1H),6.66-6.55(m,1H),4.48(d,J=12.9Hz,1H),4.28(d,J=13.0Hz,1H),4.11(dd,J=12.9,7.0Hz,1H),3.96(dd,J=13.4,7.7Hz,1H),2.97(q,J=7.3Hz,4H),2.83(d,J=7.4Hz,3H),2.13(p,J=7.3Hz,2H).;HRMS(ESI)m/z calcd for C20H23N2O2(M+H)+323.1760,found 323.1758。
EXAMPLE 11 preparation of (E) -3- (4-trifluoromethylphenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl]-N-methylprop-2-en-1-amine (Compound I)A-6)
Figure GPA0000247630360000262
The required starting materials, reagents and preparation were the same as in example 4-6, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-trifluoromethylphenyl) -acrolein, to give 210 mg of the title compound as a colorless oil in a yield of 61%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.89-7.62(m,4H),7.48-7.24(m,3H),7.00(d,J=15.8Hz,1H),6.61-6.43(m,1H),4.47(d,J=13.2Hz,1H),4.27(d,J=12.9Hz,1H),4.09(dd,J=13.0,6.9Hz,1H),4.00-3.87(m,1H),2.97(dd,J=14.2,7.0Hz,4H),2.83(s,3H),2.22-2.02(m,2H);HRMS(ESI)m/z calcd for C21H23F3N(M+H)+346.1783,found 346.1783。
EXAMPLE 12 preparation of 4-iodo-2, 3-dihydro-1H-indene (intermediate VII)
Figure GPA0000247630360000263
2 g of 4-aminoindane is dissolved in 15ml of concentrated hydrochloric acid, 15ml of aqueous solution of 1.5 g of sodium nitrite is added dropwise at 0 ℃, stirred for 15 minutes, and then 90 ml of aqueous solution of 25 g of potassium iodide is slowly added for reaction at 20-30 ℃ overnight. After the reaction was completed, extraction was performed three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, concentrated, and the residue was separated by column chromatography to give the title compound, 2.5 g of colorless oil, yield 70%.
1H-NMR(400MHz,CDCl3)δ7.53(d,J=7.7Hz,1H),7.17(t,J=7.2Hz,1H),6.85(t,J=7.5Hz,1H),3.06(dd,J=17.6,10.2Hz,2H),2.93(dd,J=15.6,8.4Hz,2H),2.22-1.94(m,2H).
EXAMPLE 13 preparation of 4-cyano-2, 3-dihydro-1H-indene (intermediate VIII)
Figure GPA0000247630360000271
1 g of intermediate VII, 551 mg of cuprous cyanide were added to 50ml of N, N-dimethylformamide and heated under reflux overnight. After completion of the reaction of intermediate VII, it was cooled to room temperature, concentrated aqueous ammonia was added to the system until the solution was clear, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, concentrated, and the residue was separated by column chromatography to give the title compound in 417 mg of pale yellow solid in 71% yield.
1H-NMR(400MHz,CDCl3)δ7.35(t,J=6.6Hz,2H),7.14(t,J=7.6Hz,1H),3.04(dd,J=14.6,7.1Hz,2H),2.95-2.86(m,2H),2.15-2.01(m,2H).
EXAMPLE 14 preparation of (2, 3-dihydro-1H-inden-4-yl) methylamine (intermediate IX)
Figure GPA0000247630360000272
500 mg of intermediate VIII in 20ml of anhydrous tetrahydrofuran are slowly added dropwise to a suspension of 300 mg of lithium aluminum hydride in 30 ml of anhydrous tetrahydrofuran at-78 ℃ under the protection of nitrogen, and after the dropwise addition, the reaction is carried out at room temperature overnight. 0.3 ml of water, 0.3 ml of 15% aqueous sodium hydroxide solution and 0.3 ml of water were sequentially added to the reaction system to quench the reaction, and anhydrous sodium sulfate was directly added to the reaction system to dry, filter and concentrate the reaction mixture to obtain 465 mg of the title compound as a pale yellow oil with a yield of 90%.
1H-NMR(500MHz,MeOD)δ7.27-7.13(m,3H),3.87(s,2H),3.04-2.90(m,4H),2.21-2.07(m,2H).
EXAMPLE 15 preparation of tert-butyl [ (2, 3-dihydro-1H-inden-4-yl) methyl ] carbamate (intermediate X)
Figure GPA0000247630360000273
1.5 g of intermediate IX are dissolved in 30 ml of tetrahydrofuran solution, 500 mg of sodium hydroxide are added and stirred for 5 minutes, and a solution of 2.7 g of di-tert-butyl dicarbonate in 20ml of tetrahydrofuran is slowly added in ice bath. The reaction was stirred at room temperature for 1 hour. Filtration, concentration and column chromatography of the residue gave the title compound in 87% yield as a pale yellow solid, 2.1 g.
1H-NMR(400MHz,CDCl3)δ7.14(dt,J=14.6,7.2Hz,2H),7.05(d,J=7.0Hz,1H),4.29(s,2H),2.90(dt,J=12.7,7.5Hz,4H),2.18-1.95(m,3H),1.46(s,9H).
EXAMPLE 16 preparation of 1- (2, 3-dihydro-1H-inden-4-yl) -N-methylmethanamine (intermediate XI)
Figure GPA0000247630360000281
100 mg of intermediate X in 10ml of anhydrous tetrahydrofuran is slowly added dropwise to a suspension of 50mg of lithium aluminum hydride in 10ml of anhydrous tetrahydrofuran at 0 ℃ under the protection of nitrogen, and after the dropwise addition is finished, the reaction is carried out under heating and reflux for 12 hours. After the reaction, 0.1 ml of water, 0.1 ml of 15% aqueous sodium hydroxide solution and 0.1 ml of water were sequentially added to the reaction system to quench the reaction, anhydrous sodium sulfate was directly added to the reaction system to dry, filter and concentrate the reaction product to obtain the title compound, 59 mg of yellow oily substance with a yield of 90%.
1H-NMR(400MHz,CDCl3)δ7.17-7.08(m,3H),3.72(s,2H),2.91(dd,J=16.3,7.8Hz,4H),2.47(s,3H),2.08(ddd,J=14.0,11.1,5.5Hz,2H).
EXAMPLE 17 preparation of (E) -N- [ (2, 3-dihydro-1H-inden-4-yl) methyl]-N-methyl-3-phenylprop-2-en-1-amine (Compound I)B)
Figure GPA0000247630360000282
The required starting materials, reagents and procedures were the same as in example 6 except that intermediate IV was changed to intermediate XI to give 160 mg of the title compound as an oil in 58% yield. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.53(d,J=7.5Hz,2H),7.36(dt,J=19.5,6.4Hz,6H),6.96(d,J=16.0Hz,1H),6.46-6.29(m,1H),4.52(d,J=11.1Hz,1H),4.26(d,J=11.8Hz,1H),4.03(d,J=32.8Hz,2H),3.00(dd,J=14.1,6.7Hz,4H),2.87(s,3H),2.15(dd,J=14.8,7.4Hz,2H);HRMS(ESI)m/z calcd for C20H24N(M+H)+278.1909,found 278.1903。
EXAMPLE 18 preparation of (5,6,7, 8-tetrahydronaphthalen-2-yl) methanol (intermediate XII)
Figure GPA0000247630360000291
1.7 g of 5,6,7, 8-tetrahydro-2-naphthoic acid in 20ml of anhydrous tetrahydrofuran is slowly dripped into 759 mg of lithium aluminum hydride in 50ml of anhydrous tetrahydrofuran suspension at minus 78 ℃ under the protection of nitrogen, and the reaction is carried out for 12 hours at 20-30 ℃ after the dripping is finished. 0.7 ml of water, 0.7 ml of 15% aqueous sodium hydroxide solution and 0.7 ml of water were sequentially added to the reaction system to quench the reaction, and anhydrous sodium sulfate was directly added to the reaction system to dry, filter and concentrate the reaction product to obtain the title compound, 1.4 g of colorless oil, with a yield of 89%.
1H-NMR(400MHz,CDCl3)δ7.13-7.02(m,3H),4.61(s,2H),2.76(d,J=2.5Hz,4H),1.87-1.70(m,4H).
EXAMPLE 19 preparation of 6- (bromomethyl) -1, 2,3, 4-tetrahydronaphthalene (intermediate XIII)
Figure GPA0000247630360000292
Dissolving intermediate 1.6 g of intermediate XII in 30 ml of anhydrous ether, adding 0.32 ml of phosphorus tribromide under nitrogen protection and ice-cooling, and reacting at 20-30 ℃ for 12 hours. After the reaction was completed, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to give the title compound as a colorless oil 1.8 g, yield 81%.
1H-NMR(400MHz,CDCl3):δ7.15-6.99(m,3H),4.46(s,2H),2.75(s,4H),1.79(dd,J=7.6,5.3Hz,4H).
EXAMPLE 20 preparation of N-methyl-1- (5,6,7, 8-tetrahydronaphthalen-2-yl) methylamine (intermediate XIV)
Figure GPA0000247630360000293
1 g of intermediate XIII in 20ml of ethanol is slowly added to 20ml of methylamine in 33% ethanol and reacted at 25 ℃ for 12 hours. After the reaction was complete, concentration gave the title compound as a yellow oil in a yield of 700 mg (90%).
1H-NMR(400MHz,CDCl3)δ7.16(t,J=6.0Hz,3H),4.11(s,2H),2.81(d,J=5.8Hz,4H),2.71(s,3H),1.91-1.76(m,4H).
EXAMPLE 21 preparation of (E) -N-methyl-3-phenyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-ylamine (Compound I)C-1)
Figure GPA0000247630360000301
The required starting materials, reagents and procedures were the same as in example 6 except that intermediate IV was changed to intermediate XIV to give 209 mg of the title compound as an oil in a yield of 72%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.53(d,J=6.9Hz,2H),7.42-7.31(m,3H),7.28-7.13(m,3H),6.92(d,J=15.8Hz,1H),6.42-6.28(m,1H),4.31(d,J=64.0Hz,2H),3.96(d,J=38.4Hz,2H),2.82(d,J=7.7Hz,7H),1.94-1.71(m,4H);HRMS(ESI)m/z calcd for C21H26N(M+H)+292.2065,found 292.2067。
EXAMPLE 22 preparation of (E) -3- (4-bromophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-ylamine (Compound I)C-2)
Figure GPA0000247630360000302
The required starting materials, reagents and preparation were the same as in examples 4, 5 and 21 except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-bromophenyl) -acrolein, and 244 mg of the title compound was obtained in 66% yield as a colorless oil. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.61-7.51(m,2H),7.49-7.41(m,2H),7.27-7.15(m,3H),6.88(d,J=15.8Hz,1H),6.46-6.28(m,1H),4.41(d,J=13.0Hz,1H),4.21(d,J=13.0Hz,1H),4.03(dd,J=13.4,6.7Hz,1H),3.88(dd,J=13.3,7.8Hz,1H),2.79(d,J=14.7Hz,7H),1.92-1.75(m,4H);HRMS(ESI)m/z calcd for C21H25BrN(M+H)+370.1170,found 370.1161。
EXAMPLE 23 preparation of (E) -3- (4-chlorophenyl)) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-ylamine (Compound I)C-3)
Figure GPA0000247630360000311
The required starting materials, reagents and preparation were the same as in examples 4, 5 and 21 except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-chlorophenyl) -acrolein, and 225 mg of the title compound was obtained in 69% yield as a colorless oil. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.90(s,1H),7.79(d,J=7.7Hz,1H),7.50(d,J=8.3Hz,3H),7.38(d,J=7.1Hz,3H),6.96(d,J=14.9Hz,1H),6.91(d,J=15.7Hz,1H),6.47-6.32(m,1H),4.81(d,J=13.2Hz,1H),4.61(d,J=13.4Hz,1H),4.18-4.04(m,1H),4.03-3.89(m,1H),2.84(d,J=23.3Hz,3H);HRMS(ESI)m/z calcd for C21H25ClN(M+H)+326.1676,found 326.1671。
EXAMPLE 24 preparation of (E) -3- (4-methoxyphenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-ylamine (Compound I)C-4)
Figure GPA0000247630360000312
The required starting materials, reagents and preparation were the same as in examples 4, 5 and 21, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-methoxyphenyl) -acrolein, and 158 mg of the title compound was obtained in 49% yield as a colorless oil. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.47(d,J=8.4Hz,2H),7.28-7.16(m,3H),6.95(d,J=8.5Hz,2H),6.86(d,J=15.7Hz,1H),6.19(dt,J=15.4,7.7Hz,1H),4.40(d,J=13.0Hz,1H),4.18(d,J=12.9Hz,1H),4.00(dd,J=13.0,7.2Hz,1H),3.90-3.76(m,4H),2.94-2.69(m,7H),1.84(s,4H);HRMS(ESI)m/z calcd for C22H28NO(M+H)+322.2171,found 322.2170。
EXAMPLE 25 preparation of (E) -3- (4-Nitrophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-ylamine (Compound I)C-5)
Figure GPA0000247630360000321
The required starting materials, reagents and preparation were the same as in examples 4, 5 and 21 except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-nitrophenyl) -acrolein, whereby 236 mg of the title compound was obtained in the form of yellow oil with a yield of 70%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ8.28(d,J=8.6Hz,2H),7.77(d,J=8.5Hz,2H),7.34-7.11(m,3H),7.04(d,J=15.9Hz,1H),6.67-6.50(m,1H),4.44(d,J=13.0Hz,1H),4.25(d,J=12.7Hz,1H),4.10(dd,J=13.4,6.9Hz,1H),4.03-3.87(m,1H),2.83(d,J=10.2Hz,7H),1.84(s,4H).HRMS(ESI)m/z calcd for C21H25N202(M+H)+337.1916,found 337.1917。
EXAMPLE 26 preparation of (E) -3- (4-bromophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl]Prop-2-en-1-ylamine (Compound I)C-6)
Figure GPA0000247630360000322
The required raw materials, reagents and preparation methods were the same as in examples 4, 5 and 21 except that (E) -3- (4-methylphenyl) -acrolein was changed to (E) -3- (4-trifluoromethylphenyl) -acrolein, and 258 mg of the title compound was obtained as a yellow oil in a yield of 72%. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.89-7.55(m,4H),7.36-7.09(m,3H),6.99(d,J=15.8Hz,1H),6.53(d,J=3.2Hz,1H),4.40(t,J=19.1Hz,1H),4.23(d,J=12.1Hz,1H),4.07(s,1H),3.94(s,1H),2.77(d,J=50.6Hz,7H),1.84(s,4H).;HRMS(ESI)m/z calcd for C22H25F3N(M+H)+360.1939,found 360.1938。
EXAMPLE 27 preparation of 2-ethoxy-5-methylbenzaldehyde (intermediate XV)
Figure GPA0000247630360000323
4 g of 2-hydroxy-5-methylbenzaldehyde is dissolved in 40 ml of N, N-dimethylformamide, 8 g of potassium carbonate is then added under nitrogen protection, and after stirring for 15 minutes, 3.6 ml of iodoethane are added and stirred at room temperature for 12 hours. After the reaction was completed, water and ethyl acetate were added to the system for extraction, and the organic phase was collected, washed once with water and saturated sodium chloride, dried over anhydrous magnesium sulfate, filtered, concentrated, and the residue was separated by column chromatography to give the title compound, 4.6 g of a pale yellow block solid, yield 95%.
1H-NMR(400MHz,CDCl3)δ10.48(s,1H);7.63(d,J=2.1Hz,1H);7.33(dd,J=8.5,2.1Hz,1H);6.87(d,J=8.5Hz,1H);4.12(q,J=7.0Hz,2H);2.31(s,3H);1.46(t,J=7.0Hz,3H)。
EXAMPLE 28 preparation of (E) -methyl-5- (2-ethoxy-5-methylphenyl) pent-4-enoic acid ethyl ester (intermediate XVI)
Figure GPA0000247630360000331
19 g of (4-methoxy-4-oxobutyl) triphenyl phosphine bromide and 51 ml of hexamethyl phosphoric triamide are added into 102 ml of anhydrous tetrahydrofuran, 41 ml of lithium bistrimethylsilyl amide is added under the condition of argon protection at 0 ℃ for reaction for 2 hours, then the reaction system is placed at minus 78 ℃, and finally 4 g of intermediate XV is added for reaction for 1 hour. After the reaction was completed, the system was quenched with saturated ammonium chloride, made acidic with dilute hydrochloric acid, extracted 4 times with ethyl acetate, dried over anhydrous magnesium sulfate, filtered, concentrated, and the residue was separated by column chromatography to give the title compound as a pale yellow oil 5.5 g, 91% yield.
1H-NMR(400MHz,CDCl3)δ7.00(dd,J=14.1,5.8Hz,2H);6.76(d,J=8.2Hz,1H);6.55(d,J=11.6Hz,1H);5.63(dt,J=11.6,7.2Hz,1H);4.01(q,J=7.0Hz,2H);3.67(s,3H);2.58(q,J=8.0Hz,2H);2.42(dd,J=14.2,7.2Hz,2H);2.29(s,3H);1.39(t,J=7.0Hz,3H)。
EXAMPLE 29 preparation of (E) -5- (2-ethoxy-5-methylphenyl) pent-4-enoic acid (intermediate XVII)
Figure GPA0000247630360000332
5.3 g of intermediate XVI was dissolved in 35 ml of tetrahydrofuran, and then 20ml of an aqueous solution containing 1.5 g of lithium hydroxide was added to the solution, followed by reaction at room temperature for 8 hours. After the reaction was completed, diluted hydrochloric acid was added to adjust the system to acidity, followed by extraction with ethyl acetate 3 times, drying over anhydrous magnesium sulfate, filtration, concentration, and separation of the residue by column chromatography to give the title compound, 4.6 g of a colorless oil, in a yield of 92%.
1H-NMR(400MHz,CDCl3)δ7.01(d,J=8.3Hz,2H);6.76(d,J=8.0Hz,1H);6.56(d,J=11.6Hz,1H);5.64(dt,J=11.6,7.1Hz,1H);4.07-3.95(m,2H);2.66-2.53(m,2H);2.47(t,J=7.5Hz,2H);2.29(s,3H);1.39(t,J=7.0Hz,3H)。
EXAMPLE 30 preparation of 5- (2-ethoxy-5-methylphenyl) pentanoic acid (intermediate XVIII)
Figure GPA0000247630360000341
4.6 g of the intermediate XVII is dissolved in 30 ml of methanol, and then 0.67 g of palladium-carbon catalyst is added, and reduction reaction is carried out for 10 hours under hydrogen at room temperature. After the reaction was complete, palladium on carbon was removed by suction filtration, the filtrate was retained and concentrated to give the title compound as a colorless oil in 97% yield.
1H-NMR(400MHz,CDCl3)δ6.93(d,J=6.9Hz,2H);6.71(d,J=8.9Hz,1H);3.99(q,J=7.0Hz,2H);2.60(t,J=7.2Hz,2H);2.39(t,J=7.2Hz,2H);2.26(s,3H);1.75-1.56(m,4H);1.39(t,J=7.0Hz,3H)。
EXAMPLE 31 preparation of 1-ethoxy-4-methyl-6, 7,8, 9-tetrahydro-5H-benzo [7] annulen-5-one (intermediate XIX)
Figure GPA0000247630360000342
4.3 g of intermediate XVIII was dissolved in 55 ml of Eton's reagent (7.5% solution of phosphorus pentoxide in methanesulfonic acid) under ice-bath, and then reacted at 25 ℃ for 12 hours. After completion of the reaction, the system was poured into an ice-water mixture, followed by extraction with dichloromethane 4 times, drying over anhydrous magnesium sulfate, filtration, concentration, and column chromatography of the residue to give the title compound as a pale yellow oil 1.2 g, yield 31%.
1H-NMR(400MHz,CDCl3)δ7.01(d,J=8.3Hz,1H);6.82(d,J=8.3Hz,1H);4.00(q,J=7.0Hz,2H);2.82(t,J=6.2Hz,2H);2.57(t,J=6.2Hz,2H);2.22(s,3H);1.80-1.68(m,4H);1.40(t,J=7.0Hz,3H)。
EXAMPLE 32 preparation of 1-ethoxy-4-methyl-6, 7,8, 9-tetrahydro-5H-benzo [7] annulene (intermediate XX)
Figure GPA0000247630360000343
Dissolving 1.2 g of intermediate XIX, 1 g of sodium cyanoborohydride and 5.3 g of zinc iodide in 30 ml of 1, 2-dichloroethane, heating and refluxing for 6 hours, pouring the system into a sand core funnel containing kieselguhr for suction filtration while the system is hot after the reaction is finished, concentrating the filtrate, and separating the residue by column chromatography to obtain 0.9 g of colorless oily matter with the yield of 80%.
1H-NMR(400MHz,CDCl3)δ6.86(d,J=8.3Hz,1H);6.58(d,J=8.3Hz,1H);3.92(q,J=7.0Hz,2H);2.91-2.83(m,2H);2.79-2.71(m,2H);2.20(s,3H);1.78(dt,J=11.8,6.0Hz,2H);1.54(dq,J=11.3,5.6Hz,4H);1.34(t,J=7.0Hz,3H)。
Example 33 preparation of 4-ethoxy-6, 7,8, 9-tetrahydro-5H-benzo [7] annulene-1-carbaldehyde (intermediate XXI)
Figure GPA0000247630360000351
0.9 g of intermediate XX and 1 g of pyridinium chlorochromate were added to 30 ml of anhydrous acetonitrile and reacted under reflux for 10 hours. After the reaction was completed, the system was concentrated, and then water and methylene chloride were added to the residue to extract, and the organic layer was collected, dried over anhydrous magnesium sulfate, filtered, concentrated, and the residue was subjected to column chromatography to obtain the title compound, 0.26 g of a pale yellow oil, yield 27%.
1H-NMR(400MHz,CDCl3)δ10.18(s,1H);7.64(d,J=8.7Hz,1H);6.81(d,J=8.7Hz,1H);4.09(q,J=7.0Hz,2H);3.37-3.28(m,2H);3.01-2.89(m,2H);1.84(dt,J=11.8,6.0Hz,2H);1.72-1.62(m,2H);1.62-1.52(m,2H);1.45(t,J=7.0Hz,3H)。
EXAMPLE 34 preparation of 4-hydroxy-6, 7,8, 9-tetrahydro-5H-benzo [7] annulene-1-carbaldehyde (intermediate XXII)
Figure GPA0000247630360000352
250 mg of intermediate XXI and 460 mg of aluminum chloride are added to 15ml of dichloromethane and reacted at room temperature for 24 hours. After the reaction was completed, a little water and saturated sodium chloride were added to the system, followed by extraction with dichloromethane 3 times, collection of the organic layer, drying over anhydrous magnesium sulfate, filtration, concentration, and column chromatography of the residue to give 150 mg of a yellow solid in a yield of 69%.
1H-NMR(400MHz,MeOD)δ10.18(s,1H);7.58(d,J=8.5Hz,1H);6.73(d,J=8.5Hz,1H);3.39-3.24(m,2H);2.99-2.84(m,2H);1.86(dt,J=11.9,6.1Hz,2H);1.74-1.56(m,4H)。
EXAMPLE 35 preparation of 4-formyl-6, 7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl trifluoromethanesulfonate (intermediate XXIII)
Figure GPA0000247630360000361
114 mg of intermediate XXII and 0.15 ml of pyridine were dissolved in 10ml of dichloromethane, 0.21 ml of trifluoromethanesulfonic anhydride was added dropwise in ice bath, stirred at 25 ℃ for 2 hours, the mixture was washed with water 2 times, brine, dried over anhydrous magnesium sulfate, filtered and concentrated to give the title compound in 189 mg of yellow solid with 98% yield. Directly putting into the next step without separation and purification.
EXAMPLE 36 preparation of 6,7,8, 9-tetrahydro-5H-benzo [7] annulene-1-carbaldehyde (intermediate XXIV)
Figure GPA0000247630360000362
0.72 ml of triethylamine was dissolved in 5ml of N, N-dimethylformamide, 0.18 ml of 98% formic acid was added dropwise, followed by 189 mg of intermediate XXIII, 54 mg of palladium acetate and 27 mg of 1, 1' -bis (diphenylphosphino) ferrocene. The reaction was carried out at 80 ℃ for 15 minutes, cooled to room temperature, the solid was filtered off, water was added to the filtrate, extraction was carried out three times with ethyl acetate, drying was carried out over anhydrous magnesium sulfate, filtration and concentration were carried out, and the residue was subjected to column chromatography to give the title compound in 97 mg of a pale yellow oil in a yield of 95%.
1H-NMR(400MHz,CDCl3)δ10.36(s,1H),7.64(d,J=7.6Hz,1H),7.34(d,J=7.1Hz,1H),7.22(d,J=7.5Hz,1H),3.29(d,J=4.9Hz,2H),2.93-2.84(m,2H),1.86(d,J=5.5Hz,2H),1.72-1.63(m,4H).
EXAMPLE 37 preparation of (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methanol (intermediate XXV)
Figure GPA0000247630360000363
348 mg of intermediate XXIV is dissolved in 10ml of methanol, and 80mg of sodium borohydride are added in portions in ice bath and reacted for 10 to 30 minutes at room temperature. Concentration, water was added to the residue, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered and concentrated to give the title compound, 352 mg of a colorless oil, 100% yield. Directly putting into the next step without separation and purification.
EXAMPLE 38 preparation of 1- (bromomethyl) -6, 7,8, 9-tetrahydro-5H-benzo [7] annulene (intermediate XXVI)
Figure GPA0000247630360000364
352 mg of intermediate XXV are dissolved in 20ml of anhydrous ether, 0.06 ml of phosphorus tribromide is added under nitrogen protection and ice bath, and the reaction is carried out at 25 ℃ for 12 hours. After the reaction was completed, the reaction system was poured into an ice saturated sodium bicarbonate solution, extracted three times with ethyl acetate, washed with saturated brine, dried over anhydrous magnesium sulfate, filtered, and concentrated at 30 ℃ to obtain the title compound in 473 mg of a colorless oil in a yield of 99%. Directly putting into the next step without separation and purification.
EXAMPLE 39 preparation of N-methyl-1- (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methylamine (intermediate XXVII)
Figure GPA0000247630360000371
473 mg of intermediate XXVI in 10ml of ethanol was slowly added to 10ml of methylamine in 33% ethanol and reacted at 25 ℃ for 12 hours. After the reaction was complete, concentration afforded the title compound as a pale yellow oil in 95% yield. Directly putting into the next step without separation and purification.
EXAMPLE 40 preparation of (E) -N-methyl-3-phenyl-N- [ (6,7,8, 9-tetrahydro-5H-benzo [7]]Rotalen-1-yl) methyl]Prop-2-en-1-ylamine (Compound I)D-1)
Figure GPA0000247630360000372
The required starting materials, reagents and procedures were the same as in example 6 except that intermediate IV was replaced with intermediate XXVII to give 134 mg of the title compound as an oil in 44% yield. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.54(d,J=7.2Hz,2H),7.44-7.34(m,3H),7.35-7.26(m,2H),7.22(t,J=7.5Hz,1H),6.98(d,J=15.8Hz,1H),6.45-6.33(m,1H),4.60(s,1H),4.35(s,1H),4.03(s,2H),3.03-2.88(m,4H),2.84(s,3H),1.88(s,2H),1.65(s,4H);HRMS(ESI)m/z calcd for C22H28N(M+H)+306.2222,found 306.2220。
EXAMPLE 41 preparation of (E) -N-methyl-3- (4-bromophenyl) -N- ((6,7,8, 9-tetrahydro-5H-benzo [7]]Rotan-1-yl) methyl) prop-2-en-1-amine (Compound I)D-2)
Figure GPA0000247630360000381
The required starting materials, reagents and preparation were the same as in examples 4, 5 and 40, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-bromophenyl) -acrolein, and 215 mg of the title compound was obtained in 56% yield as a colorless oil. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.57(d,J=8.4Hz,2H),7.47(d,J=8.4Hz,2H),7.34-7.25(m,2H),7.22(t,J=7.5Hz,1H),6.94(d,J=15.8Hz,1H),6.49-6.37(m,1H),4.62(d,J=13.4Hz,1H),4.35(d,J=13.4Hz,1H),4.15-3.90(m,2H),3.06-2.86(m,4H),2.84(s,3H),1.88(dd,J=11.3,5.1Hz,2H),1.82-1.55(m,4H);HRMS(ESI)m/z calcd for C22H27BrN(M+H)+384.1327,found 384.1324。
EXAMPLE 42 preparation of (E) -N-methyl-3- (4-chlorophenyl) -N- [ (6,7,8, 9-tetrahydro-5H-benzo [7]]Rotalen-1-yl) methyl]Prop-2-en-1-ylamine (Compound I)D-3)
Figure GPA0000247630360000382
The required starting materials, reagents and preparation were the same as in examples 4, 5 and 40, except that (E) -3-phenyl-acrolein was changed to (E) -3- (4-chlorophenyl) -acrolein, and 214 mg of the title compound was obtained in 63% yield as a colorless oil. The hydrochloride salt of this compound was a white solid.
1H-NMR(400MHz,MeOD)δ7.53(d,J=8.4Hz,2H),7.41(d,J=8.4Hz,2H),7.30(dd,J=13.1,7.5Hz,2H),7.25-7.15(m,1H),6.95(d,J=15.8Hz,1H),6.47-6.30(m,1H),4.61(d,J=12.3Hz,1H),4.35(d,J=12.5Hz,1H),4.12-3.92(m,2H),3.00-2.89(m,4H),2.88-2.76(m,3H),1.88(d,J=5.4Hz,2H),1.79-1.52(m,4H).;HRMS(ESI)m/z calcd for C22H27C1N(M+H)+340.1832,found 340.1826。
EXAMPLE 43 preliminary screening experiment for inhibitory Activity of Compounds of the present invention on the Synthesis of golden yellow pigment
Strains for experiments: a freshly activated Staphylococcus aureus Newman wild strain (Staphylococcus aureus subsp. aureus str. Newman) and its homologous crtN-inserted mutant (no synthesis of aureochrome).
Culture medium for experiments: tryptone Soy Broth (TSB), a product of Oxid, UK, was sterilized at 121 ℃ for 15 minutes in distilled water.
The primary screening experiment method comprises the following steps:
(1) preparation of the compound: the compound of the present invention was dissolved in dimethyl sulfoxide (DMSO) to prepare a mother liquor having a concentration of 10 mM. mu.L of the mother liquor was diluted to a concentration of 2mM with 400. mu.L of DMSO, and after mixing, 250. mu.L (2mM) of the solution was further diluted 2-fold with an equal amount of DMSO until the concentration of the solution became 0.0625mM, and was ready for use.
(2) Culturing the strain: a single clone of the Newman strain was picked from the TSA plate, transferred to a tube containing 4mL of sterile TSB medium, and cultured at 37 ℃ and 250rpm for 12 hours, and then kept ready.
(3) The compound of the invention is used for primary screening of the ability of inhibiting the synthesis of the yellow pigment in staphylococcus aureus: sterile tubes were taken and fresh sterile TSB medium 3980. mu.L was added to each tube. Subsequently, 20. mu.L of each of the compound solutions prepared at concentrations of 10mM, 2mM, 1mM, 0.5mM, 0.25mM, 0.125mM, and 0.0625mM was added to the test tube to give final concentrations of 50. mu.M, 10. mu.M, 5. mu.M, 2.5. mu.M, 1.25. mu.M, 0.625. mu.M, and 0.3125. mu.M, respectively, of the compound of the present invention. Meanwhile, to another tube, 20. mu.L of DMSO solution (final concentration of 0.5%) was added as a negative control without compound. To each test tube, 40. mu.L of a cell suspension (inoculum size: 1: 100) cultured for 12 hours was added, and after culturing at 37 ℃ and 250rpm for 24 hours, 1.5mL of the cell suspension was taken out, 14000g was centrifuged for 2 minutes, and the supernatant was removed to observe whether or not the amount of the synthesized golden yellow pigment was significantly decreased as compared with the negative control after the addition of the compound of the present invention at a specific concentration to the strain.
EXAMPLE 44 Compound I of the present inventionA-6 and IC-2 preliminary screening test results for inhibitory activity against golden yellow pigment synthesis
Compounds I of the inventionA-6 and IC-2 final photographs of inhibition of synthesis of golden yellow pigment as shown in FIGS. 8 and 9, respectively, at concentrations of 50. mu.M, 10. mu.M, 5. mu.M, 2.5. mu.M, 1.25. mu.M, 0.625. mu.M, 0.3125. mu.M, 0. mu.M (control) from left to right.
The results show that the compounds I according to the invention are present at concentrations as low as 0.3125. mu.MA-6 and ICAnd 2 can obviously inhibit the synthesis of golden yellow pigment.
EXAMPLE 45 IC inhibition of golden yellow pigment Synthesis Activity by Compounds of the invention50Measurement experiment method
Selection of compound concentration: and determining the capability of each compound for inhibiting the synthesis of the golden yellow pigment according to the primary screening result. If the compound with stronger activity can still strongly inhibit the generation of the pigment at the lowest concentration of the primary screening, the experiment can be continued according to a similar method of the primary screening until the compound can not substantially inhibit the generation of the golden yellow pigment. According to the experimental results, 11 different concentration gradients were designed for each compound, such that its ability to inhibit pigment synthesis was comprised substantially between 0% and 100%. Culturing the strain: the Newman strain and the crtN mutant were picked from the TSA plate, and the monoclonal antibodies were cultured in a tube containing 4mL of sterile TSB medium at 37 ℃ and 250rpm for 12 hours. IC (integrated circuit)50The determination of (1): sterile tubes were taken and fresh sterile TSB medium 3980. mu.L was added to each tube. Subsequently, 20. mu.L of each of the 11 concentration gradients of the formulated compound of the present invention was added to the test tube. At the same time, toIn two other tubes, 20. mu.L of DMSO solution (final concentration: 0.5%) was added as a control without compound. To each of the two tubes to which the DMSO solution was added, 40. mu.L of Newman (negative control) and crtN mutant (positive control) cultured for 12 hours were added. The remaining tubes containing the compound were filled with 40. mu.L of Newman strain cultured for 12 hours. All tubes were incubated at 37 ℃ for 12 hours at 250rpm, then switched to 30 ℃ and incubated at 250rpm for an additional 36 hours to increase pigment accumulation. After completion of the culture, 2mL of the cell suspension was placed in a 2mL EP tube, centrifuged at 14000g for 2 minutes, the supernatant was removed, washed twice with PBS buffer (1 mL each), added with 300. mu.L of methanol solution, vortexed, and heated in a 55 ℃ water bath for 3 minutes to extract the pigment. After centrifugation at 14000g for 2 minutes, the methanol extract was aspirated into a 1.5mL EP tube, an equal amount of methanol solution was added, extraction was repeated twice, and the three extracted pigments were combined. The methanol extract in the crtN mutant was used as a blank control, and the absorbance of each sample at a wavelength of 450nm was measured, and the absorbance of the no-compound negative control was measured. At each concentration, the relative level of pigment synthesis was 100% a450 (sample)/a 450 (negative control). Fitting an inhibitor concentration-inhibition ratio (log (inhibitor) vs. response) curve in Graphpad prism 5.0 software by using the molar concentration of the compound as a horizontal coordinate and the relative level of the dye synthesis as a vertical coordinate, and calculating the IC of the compound for inhibiting the dye synthesis according to the fitting result by the software50
EXAMPLE 46 IC inhibition of the synthetic Activity of golden yellow pigment by Compounds of the invention50Determination of the results of the experiments
Selecting staphylococcus aureus Newman for inhibiting synthetic activity of golden yellow pigment on synthesized benzo-aliphatic ring-substituted alkylamine compound50In the tests, the activity data are shown in Table 1, and a total of 14 compounds of the invention are found to have strong activity for inhibiting the synthesis of the golden yellow pigment, wherein half of the effective inhibitory concentration IC50The active compounds with < 10nM are 5, half the effective inhibitory concentration 10nM < IC50The active compounds with < 100nM are 7, half the effective inhibitory concentration 100nM < IC50There were 2 active compounds < 1000 nM.
TABLE 1 inhibitory Activity data (IC) of Compounds of formula I on the Synthesis of golden yellow pigment50,nM)
Figure GPA0000247630360000401
Figure GPA0000247630360000411
Figure GPA0000247630360000421
Figure GPA0000247630360000431
As can be seen from Table 1, most of the compounds of the formula I have strong inhibitory activity on the synthesis of golden yellow pigment, which indicates that the compounds of the invention can be developed into novel antibacterial drugs targeting the synthesis of golden yellow pigment of staphylococcus virulence factor.
Example 47 Compound I of the present inventionA-6 IC50 determination experimental method and result for inhibiting synthesis activity of drug-resistant bacteria golden yellow pigment
The procedure is as in examples 43 and 45 except that the Staphylococcus aureus Newman is replaced by USA400MW2, USA300LAC and Mu 50.
The results are shown in FIG. 1, and the compounds I of the present invention A6, the synthesis inhibition of the staphylococcus aureus golden pigment is not limited to Newman strain, and has strong inhibition effect on drug-resistant strains USA400MW2, USA300LAC and Mu 50.
EXAMPLE 48 Compounds I of the present inventionA-2、IA-6、IC-2 and IC-6 IC for CrtN inhibitory activity of key enzyme in synthetic process of golden yellow pigment50Measurement test method and results
1) Preparation of substrate diaphylene emulsion
Overnight cultured pet28 a: : coli (DE3) was transferred to 50ml of fresh LB + kanamycin kanamycine (final concentration: 50. mu.g/ml) medium at a ratio of 1: 100 (broth: medium), cultured at 37 ℃ and 250rpm for 24 hours, and then the cells were collected by centrifugation at 8000g for 4min and washed twice with PBS buffer. Adding 20ml of acetone solution into the thalli, carrying out vortex mixing to extract the pigment and the intermediate product thereof, then adding 10ml of normal hexane and 10ml of NaCl (10% by mass/volume) solution into the extracting solution, carrying out vigorous oscillation to remove the grease component in the extracting solution, then collecting a hexane layer containing the pigment and the intermediate product thereof, adding 10ml of normal hexane, and repeating the extraction process once. The two hexane extracts were combined, and dried by adding anhydrous MgSO4 and weighed. The resulting diamphyloene and phosphatidylcholine were dissolved in 200. mu.l chloroform at a ratio of 1: 3 and concentrated to dryness in vacuo. 2ml of 0.02M HEPES buffer (20mM HEPES, pH 7.5; 500mM NaCl) was added to a mixture of 8mg of diaphylene and 24mg of phosphatidylcholine, followed by sonication in ice water until a homogeneous emulsion was formed.
2) Analysis of enzyme Activity of CrtN
Preparing related component mother liquor in a reaction system: FAD 10mM, glucose 200mM, glucose oxidase 2000U/ml, hydro-peroxidase, dissolved with diaphysoene emulsion to 20000U/ml. The above solutions were all prepared with 0.02M HEPES buffer.
The entire reaction system was 700. mu.L, and was carried out in a 2ml EP tube. The reaction system comprises the following components: 50 mul of diaphylene emulsion (containing hydro-peroxidase), 70 mul of compounds with different concentrations (prepared by distilled water) or distilled water, 262.5 mul of 0.02M HEPES buffer, 3.5 mul of FAD solution, 7 mul of glucose oxidase solution, and finally 300 mul of pet28 a: : coli (DE3) Whole cell lysate (-1.41mg of CrtN protein). The reaction was carried out in a shaker at 37 ℃ for 14 hours, the rotation of the shaker being 250 rpm/min.
3) Reaction product extraction and detection
After completion of the reaction, 500. mu.L of methanol was added to terminate the reaction, and the reaction solution was transferred to a 15ml centrifuge tube. The reaction solution was added with 700. mu.L of chloroform, vortexed sufficiently to extract the reacted pigment, and then centrifuged at 7000rpm for 3 minutesThe chloroform layer was carefully aspirated. The reaction product was extracted with 500. mu.L of chloroform in the residual reaction solution, and the combined reaction extracts were concentrated to dryness in vacuo. The concentrated product was dissolved by adding 200. mu.L of chloroform and sucked into a 96-well microplate, followed by measuring the absorbance at 450nm for the quantification of the CryN product, diaponeurosporine. IC (integrated circuit)50The concentration of the compound corresponding to half of the activity of inhibiting CrtN under the experimental condition is defined, and the CrtN enzyme activity-effect relation curve is drawn in Graphpad 5.0.
As shown in FIG. 2, the compound I of the present inventionA-2、IA-6、IC-2 and IC-6 is a potent inhibitor of the key enzyme CrtN in the synthesis of golden yellow pigment.
EXAMPLE 49 Compound I of the present invention A6 pairs of experiments to enhance the Hydrogen peroxide killing of four Staphylococcus aureus (Newman, USA400MW2, USA300LAC and Mu50) and results
To a sterile test tube was added a compound at a specified concentration to give a final concentration of 1. mu.M, and four S.aureus solutions were added in an overnight culture in an inoculum to medium ratio of 1: 100. After culturing at 37 ℃ and 250rpm for about 24 hours, 500. mu.L of the culture broth was aspirated and centrifuged, and the cells were collected and washed twice with PBS buffer. Then, 500. mu.L of PBS solution was added and vortexed thoroughly to resuspend the cells, and 15. mu.L of the cell suspension was pipetted and added to 1500. mu.L of PBS buffer and vortexed thoroughly (OD ═ 0.1). 250 mu L of the uniformly mixed bacterial liquid is taken and put into a 2ml EP tube, and 10 mu L of about 37 percent hydro-peroxide solution is added to ensure that the final concentration of hydrogen peroxide in the bacterial liquid is 1.5 percent. After addition of hydrogen peroxide, the EP tube was capped with a sealing film and incubated at 37 ℃ and 250rpm for 30 minutes for killing. Another 250. mu.L of the mixture was mixed and 10. mu.L of sterile PBS buffer was added to the mixture as a control. After completion of the reaction, 5. mu.L of a prepared catalase (stock solution: 20000U/ml, PBS buffer preparation) solution was added and vortexed to decompose the residual hydrogen peroxide. And 100. mu.L of the reaction solution was put into 900. mu.L of sterile PBS buffer, and diluted 10 times, and so on until the dilution was 106 times. The dilutions were individually spotted in 10. mu.L onto TSA plates and incubated overnight in a 37 ℃ incubator to count the number of surviving colonies. The survival rate of bacteria after hydrogen peroxide killing was calculated as (number of bacteria grown after hydrogen peroxide killing of sample × dilution factor)/(number of bacteria grown by control group × dilution factor) × 100%.
As shown in FIG. 3, Compound I of the present inventionAAnd 6, the killing of four staphylococcus aureus by hydrogen peroxide can be obviously enhanced, and the survival rate is greatly reduced.
EXAMPLE 50 Compounds I of the present invention A6 pairs of experiments to enhance the blood killing of four species of Staphylococcus aureus (Newman, USA400MW2, USA300LAC and Mu50) human and results
To a sterile test tube was added a compound at a specified concentration to give a final concentration of 1. mu.M, and four S.aureus solutions were added in an overnight culture in an inoculum to medium ratio of 1: 100. After culturing at 37 ℃ and 250rpm for about 24 hours, 500. mu.L of the culture broth was aspirated and centrifuged, and the cells were collected and washed twice with PBS buffer. Then, 500. mu.L of PBS solution was added and vortexed thoroughly to resuspend the cells, and 15. mu.L of the cell suspension was pipetted and added to 1500. mu.L of PBS buffer and vortexed thoroughly (OD ═ 0.1). Then, 150. mu.L of the bacterial solution with OD equal to 0.1 was added to 850. mu.L of sterile PBS buffer so that OD equal to-0.015 was obtained for use. Collecting fresh venous blood of healthy human body with BD VACUTAINER PT tube, taking one sterile glass test tube, adding 360 μ L fresh blood and 40 μ L bacteria solution with OD ═ 0.015 successively, then incubating at 37 deg.C and 250rpm for 6 hr, taking 50 μ L reaction solution to 450 μ L sterile PBS buffer, diluting 10 times, and so on until diluting 10 times6And (4) doubling. Another bacterial solution with OD-0.015 is taken as a reference, and diluted by 10 times until the dilution is 10 times6And (4) doubling. The dilutions were individually spotted in 10. mu.L onto TSA plates and incubated overnight in a 37 ℃ incubator to count the number of surviving colonies. The bacterial survival rate after blood killing was calculated as (number of bacteria grown after blood killing of the sample × dilution factor)/(number of bacteria grown by control group × dilution factor/10) × 100%.
As shown in FIG. 4, Compound I of the present inventionAAnd 6, the killing of human blood to four staphylococcus aureus can be remarkably enhanced, and the survival rate is greatly reduced.
EXAMPLE 51 Compounds I of the inventionA-6 is inMethod for testing activity of mice against three staphylococcus aureus (Newman, USA400MW2 and Mu50) and results
SPF-grade female BALB/c mice (purchased from Shanghai Sphere-BikKa laboratory animals Co., Ltd.) were raised aseptically to 6-8 weeks of age.
The overnight cultured S.aureus strain was transferred to fresh sterile Tryptone Soy Broth (TSB) and cultured at 37 deg.C, 250rpm for 3 hours per minute to exponential phase. After washing twice with PBS buffer, the suspension was suspended in PBS.
In the mouse infection experiment, the mice were randomly grouped into 15 mice each.
All mice were anesthetized by intraperitoneal injection of sodium pentobarbital (80mg/kg) and then infected retroorbitally with 100 μ L of different bacterial loads (see below for details).
For the compound treatment groups, two I of 200mg/kg and 50mg/kg were setA6 doses of control group, the first drug injection was 12 hours before bacterial infection and 8 total injections (2 times daily, 12 hours apart, 9 times total) within 4 days after infection. After the experiment was completed, mice were allowed to inhale CO2Is sacrificed. The heart, kidney and liver of the mice were removed and uniformly crushed in 1mL of sterile PBS buffer (containing 0.01% triton X-100). The disruption solution was serially diluted, 10. mu.L of each dilution was dropped onto a TSA plate, and the bacterial CFU count was determined. The number of bacteria in different organs is the number of bacteria at a specific dilution times. And statistically analyzed in Graphpad 5.0 software using Mann-Whitney Test (two-tailed). The infection dose: newman: 1X 107CFU;USA400 MW:4×107CFU;Mu50:1.6×108And (4) CFU. BPH-652 was set as a positive control in a drug-resistant bacteria (USA400 MW2 and Mu50) infection model.
As shown in FIG. 5, Compound I of the present inventionA-6 significantly reduced colonization of the kidney and liver of mice by the Newman strain. In the kidney, both low and high dose groups showed good activity with bacterial clearance rates of 92.9% and 93.0%. In the heart, the clearance rate of bacteria in a high-dose group reaches 97.6 percent, and the bacteria in a low-dose groupThe clearance rate reaches 92.4 percent.
As shown in FIG. 6, Compound I of the present invention A6 can obviously reduce the colonization of USA400MW2 strain in mouse liver and kidney, and the treatment effect is better than that of positive control BPH-652. In the liver, the bacterial clearance rate of the high-dose group is up to 99.95%, and the bacterial clearance rate of the low-dose group is up to 97.6%. In the kidney, the bacterial clearance rate of the high-dose group is up to 99.9 percent, and the bacterial clearance rate of the low-dose group is up to 99.7 percent.
As shown in FIG. 7, Compound I of the present inventionA-6 can obviously reduce the colonization of the Mu50 strain in the liver and kidney of the mouse, and the treatment effect is better than that of the positive control BPH-652
Figure GPA0000247630360000471
In the liver, the bacterial clearance rate of the high-dose group reaches 99.997 percent, and the bacterial clearance rate of the low-dose group reaches 99.6 percent. In the kidney, the bacterial clearance rate of the high-dose group is up to 99.1 percent, and the bacterial clearance rate of the low-dose group is up to 98.8 percent.
The benzo-aliphatic ring-substituted alkylamine compound has the advantages of simple molecular structure, simple preparation process and low production cost, and shows strong inhibitory activity in a golden yellow pigment synthesis inhibition experiment which has close relation with pathogenic links of bacteria.
The compounds of the invention simultaneously exhibit potent CrtN inhibitory activity, especially Compound IA-6:
(1) In vitro: can effectively inhibit the synthesis of golden yellow pigments of drug-resistant bacteria USA400MW2, USA300LAC and Mu50, and can obviously enhance the killing capability of hydrogen peroxide and human blood to four kinds of golden yellow staphylococcus.
(2) In vivo: can remarkably reduce the colonization of staphylococcus aureus Newman, USA400MW2 and Mu50 in the kidney, heart and liver of a mouse.
Therefore, the compound of the invention is expected to be developed into a novel antibacterial drug with a single administration mode, and can also be developed into an antibacterial drug with a combined administration mode of the existing antibiotics.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (5)

1. A compound selected from the group consisting of:
(E) -3- (4-bromophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-chlorophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-methoxyphenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-nitrophenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-trifluoromethylphenyl) -N- [ (2, 3-dihydro-1H-inden-5-yl) methyl ] -N-methylprop-2-en-1-amine;
(E) -3- (4-bromophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-chlorophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-methoxyphenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-nitrophenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -3- (4-trifluoromethylphenyl) -N-methyl-N- [ (5,6,7, 8-tetrahydronaphthalen-2-yl) methyl ] prop-2-en-1-amine;
(E) -N-methyl-3- (4-bromophenyl) -N- ((6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methyl) prop-2-en-1-amine; and
(E) -N-methyl-3- (4-chlorophenyl) -N- [ (6,7,8, 9-tetrahydro-5H-benzo [7] annulen-1-yl) methyl ] prop-2-en-1-amine.
2. A pharmaceutical composition, comprising:
(1) a compound according to claim 1, or a pharmaceutically acceptable salt thereof, as an active ingredient; and
(2) a pharmaceutically acceptable carrier.
3. An antibacterial agent, comprising:
(1) a compound of claim 1, or a pharmaceutically acceptable salt thereof;
(2) an additional antibiotic; and
(3) a pharmaceutically acceptable carrier.
4. Use of a compound as claimed in claim 1, or a pharmaceutically acceptable salt thereof, for the manufacture of a formulation or medicament for:
(1) inhibiting the synthesis of staphylococcus aureus and golden yellow pigment; and/or
(2) Inhibiting a key enzyme CrtN in the synthesis process of staphylococcus aureus and golden yellow pigment; and/or
(3) Inhibiting or killing staphylococcus aureus; and/or
(4) Treating infectious diseases caused by Staphylococcus aureus.
5. A non-therapeutic, in vitro method of inhibiting the synthesis of s.aureus or inhibiting s.aureus, comprising the steps of: contacting a compound of claim 1, or a pharmaceutically acceptable salt thereof, with staphylococcus aureus, thereby inhibiting synthesis of the staphylococcus aureus or inhibiting the staphylococcus aureus.
CN201680022877.8A 2016-02-03 2016-02-03 Benzo-aliphatic ring-substituted alkylamine compound and application thereof Active CN107531613B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2016/073384 WO2017132912A1 (en) 2016-02-03 2016-02-03 Alkylamine with benzoalicyclic substituent and application thereof

Publications (2)

Publication Number Publication Date
CN107531613A CN107531613A (en) 2018-01-02
CN107531613B true CN107531613B (en) 2021-02-05

Family

ID=59500528

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201680022877.8A Active CN107531613B (en) 2016-02-03 2016-02-03 Benzo-aliphatic ring-substituted alkylamine compound and application thereof

Country Status (2)

Country Link
CN (1) CN107531613B (en)
WO (1) WO2017132912A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108117534B (en) * 2016-11-29 2021-02-05 华东理工大学 Benzo-oxygenated aliphatic cyclomethylamines
CN108997163A (en) * 2018-07-18 2018-12-14 华东理工大学 Glycyl amine compound of the aliphatic ring structure containing benzo and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0000896B1 (en) * 1977-08-19 1982-10-13 Sandoz Ag Propenyl amines, processes for their production and pharmaceutical compositions containing them
CN1007975B (en) * 1984-06-09 1990-05-16 科研制药株式会社 Process for preparing amino derivatives and fungicides containing the same
CA2666762C (en) * 2006-10-19 2014-12-30 F. Hoffmann-La Roche Ag Aminomethyl-4-imidazoles
CN104055756B (en) * 2014-05-07 2016-10-05 中国科学院上海药物研究所 The derivant of naftifine hydrochloride, Preparation method and use
CN106414392B (en) * 2014-06-10 2019-05-14 华东理工大学 6,7,8,9- tetrahydro -5H- benzo [7] annulene -2- alkyl amine compound and application thereof

Also Published As

Publication number Publication date
WO2017132912A1 (en) 2017-08-10
CN107531613A (en) 2018-01-02

Similar Documents

Publication Publication Date Title
JP2022043078A (en) Antibacterial compounds
KR20130140868A (en) Isoxazole derivatives useful as antibaterial agents
JP2002505689A (en) Quinoline-indole antimicrobial agents, uses and compositions related thereto
JP2006500403A (en) Quinolylpropylpiperidine derivatives and their use as antibacterial substances
KR101636431B1 (en) Tricyclic benzoxaborole compound, process preparing thereof and use thereof
AU2016275764B8 (en) Efflux-pump inhibitors and therapeutic uses thereof
CN101835777A (en) Carbacephem beta-lactam antibiotics
CN107531613B (en) Benzo-aliphatic ring-substituted alkylamine compound and application thereof
JP7371838B2 (en) Ring-fused thiazolino 2-pyridones, methods of their preparation and their use in the treatment and/or prevention of diseases involving Gram-positive bacteria
US10961196B2 (en) Compounds for use as an anti-bacterial or anti-fungal agent and as a zinc sensor
CN107151264B (en) Novel glycopeptide antibiotic derivative, pharmaceutical composition, preparation method and application thereof
JP2645091B2 (en) 7- (2-Methyl-4-aminopyrrolidinyl) naphthyridine and quinoline compounds
CN108117534B (en) Benzo-oxygenated aliphatic cyclomethylamines
US8288410B2 (en) 3-substituted quinolinium and 7H-indolo[2,3-c]quinolinium salts as new antiinfectives
CN101547906B (en) Antibacterial quinoline derivatives
CN108101826B (en) Indole-substituted alkylamine compound and application thereof
CN108084125B (en) Benzoheterocyclylalkylamines and their use
WO2023224892A1 (en) Inhibitors of msba as antibiotics, pharmaceutical compositions, and uses thereof
KR20030027094A (en) Novel Ester or Amide Derivatives
KR102493756B1 (en) Novel 5-oxo-1,2,4-oxadiazole derivatives, preparation method thereof, and use thereof
WO2021224501A1 (en) Ring-fused 2-pyridone compounds, methods for preparation thereof and their use in the treatment and/or prevention of a disease involving gram-positive bacteria
WO2016123146A1 (en) Bridged compounds for the treatment of bacterial infections
WO1996000570A1 (en) Antibacterial agent
CN115124487A (en) Preparation method and application of arylimino thiazole compounds
NO315939B1 (en) N1- (halocyclopropyl) -substituted pyridonecarboxylic acid derivative, antibacterial composition comprising such a compound, use of a compound-like process for the preparation thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant