CN108948147B - Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis - Google Patents

Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis Download PDF

Info

Publication number
CN108948147B
CN108948147B CN201710363824.9A CN201710363824A CN108948147B CN 108948147 B CN108948147 B CN 108948147B CN 201710363824 A CN201710363824 A CN 201710363824A CN 108948147 B CN108948147 B CN 108948147B
Authority
CN
China
Prior art keywords
formula
compound shown
compound
carrying
condensation reaction
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
CN201710363824.9A
Other languages
Chinese (zh)
Other versions
CN108948147A (en
Inventor
饶燏
宗昱
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.)
Tsinghua University
Original Assignee
Tsinghua University
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 Tsinghua University filed Critical Tsinghua University
Priority to CN201710363824.9A priority Critical patent/CN108948147B/en
Priority to PCT/CN2017/114644 priority patent/WO2018214463A1/en
Publication of CN108948147A publication Critical patent/CN108948147A/en
Application granted granted Critical
Publication of CN108948147B publication Critical patent/CN108948147B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/06General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K11/00Depsipeptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K11/02Depsipeptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof cyclic, e.g. valinomycins ; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention discloses a novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis. The structural formula of the novel antibiotic is shown as a formula I, wherein a group R1Hydrogen, alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl with 2-15 carbon atoms, aryl or acyl; radical R2Is hydrogen, halogen, alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl, aryl, substituted aryl or acyl with 2-15 carbon atoms, R4Is alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl or aryl with 2-15 carbon atoms; radical R5Is amino, guanidino or ureido; m is a natural number between 1 and 4; x is O, S or NH. The invention synthesizes a series of teixobactin analogues by a solid-phase and liquid-phase combination method for the first time, and a convergent synthesis strategy is adopted to be beneficial to synthesizing a plurality of compounds. The compound with the activity equivalent to that of teixobactin is prepared by the invention.

Description

Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis
Technical Field
The invention relates to a novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis.
Background
Gram-positive bacterial infection is a common disease and frequently encountered disease, and harms human health. In recent years, gram-positive coccal infections have increased, the detection rate of methicillin-resistant staphylococcus aureus (MRSA) has increased, penicillin-resistant streptococcus pneumoniae (PRSP) has spread in many countries and regions, vancomycin-resistant enterococci (VRE) resistant to glycopeptides and other various antibiotics have appeared, and multidrug-resistant mycobacterium tuberculosis has increased or decreased. In order to effectively control drug-resistant bacterial infection of these existing antibiotics and antibacterial agents, research and development of drugs for treating gram-positive drug-resistant bacterial infection have been the point of attention.
At present, the bacteria can be mutated to acquire drug resistance when being contacted with antibacterial drugs, and the mechanisms of the bacteria are mainly 4 types: firstly, generating antibiotic enzyme and inactivating antibiotic; variation of action target position and no response to medicine; ③ the permeability of the outer membrane is changed, and the medicine is blocked; enhancing discharge and accelerating the pumping out of the medicine entering the bacteria. In the 'meet with war' of the antibacterial drug, the surviving bacteria accumulate abundant 'fighting experience' and become drug resistant varieties, and then 'super bacteria' such as cross drug resistance, multiple drug resistance, universal drug resistance, full drug resistance and the like are evolved. Recently, a research which is carried out jointly by Chinese and foreign researchers and has a result published in a new period of 'lancet-infectious disease' magazine attracts attention. The study showed that: the existence of a specific gene MCR-1, the bacterium carrying the gene shows strong resistance to polymyxin, and the resistance can also be transferred to other strains rapidly, which means that a new 'super bacterium' is discovered.
In 2015, the detection rate of the escherichia coli resistant to the third-generation cephalosporin in China is 59%, and the detection rate of the escherichia coli resistant to the quinolones is 53.5%. The detection rate of the drug-resistant bacteria of the klebsiella pneumoniae to the third-generation cephalosporin is 36.5 percent, the detection rate of the methicillin-resistant staphylococcus aureus is 35.8 percent, and the detection rates of the representative drug-resistant bacteria are all at a higher level. It should be noted that vancomycin-resistant enterococcus faecium has become a major challenge in developed countries, and the vancomycin resistance rate of enterococcus faecium infected by the bloodstream of american ICU patients is as high as 80.7%. Globally, the average detection rate of vancomycin resistance of enterococcus faecium in north america is up to 66.8%, and the detection rate of vancomycin resistance in latin america is 39.4%. Some multi-drug resistant and multi-drug resistant bacteria are clinically driven to the desperate state, and the research and development of novel antibacterial drugs are urgently needed.
At present, carbapenem antibiotics are well-known bacteria 'ultimate defense lines', and are commonly used for treating severe infection caused by multi-drug resistant bacteria. In recent years, carbapenem-resistant bacteria have posed serious bacterial resistance problems through inter-plasmid transmission, including recently discovered colistin resistance. These life-saving drugs are often limited in clinical use due to their high toxicity. Therefore, it is desirable to provide an antibacterial compound having a novel structure.
Disclosure of Invention
The invention aims to provide a novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis, namely a novel compound.
The structural formula of the compound provided by the invention is shown as a formula I,
Figure BDA0001300987850000021
in the formula I, the radical R1Hydrogen, alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl with 2-15 carbon atoms, aryl or acyl;
radical R2Hydrogen, halogen, alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl with 2-15 carbon atoms, aryl, substituted aryl or acyl, wherein the substituted aryl is halogen substituted aryl, alkyl substituted aryl or trifluoromethyl substituted aryl;
radical R3Is amino, guanidino, ureido or carboxamido;
radical R4Is alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl or aryl with 2-15 carbon atoms;
radical R5Is amino, guanidino or ureido;
m is a natural number between 1 and 4;
x is O, S or NH.
In the compounds of formula I provided by the invention, R2The phenyl group may be a phenyl group, a halogen-substituted phenyl group or a phenyl group substituted with an alkyl group having 1 to 15 carbon atoms, and the alkyl-substituted phenyl group having 1 to 15 carbon atoms may be a phenyl group substituted with an alkyl group having 1 to 6 carbon atoms, and is preferably a methyl-substituted phenyl group, an ethyl-substituted phenyl group, a propyl-substituted phenyl group, an isopropyl-substituted phenyl group, an n-butyl-substituted phenyl group, an isobutyl-substituted phenyl group, a tert-butyl-substituted phenyl group, a pentyl-substituted phenyl group or a hexyl-substituted phenyl group.
In the compounds of formula I provided by the invention, R1、R2And R4The alkyl group having 1 to 15 carbon atoms is preferably an alkyl group having 1 to 6 carbon atoms, for example, a methyl group, an ethyl group, a propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a tert-butyl group, a pentyl group, or a hexyl group;
R1、R2and R4The alkenyl group having 2 to 15 carbon atoms is preferably an alkenyl group having 2 to 6 carbon atoms, such as vinyl, propenyl, butanylAlkenyl, isobutenyl, pentenyl or hexenyl groups, and the like;
R1、R2and R4The alkynyl group having 2 to 15 carbon atoms is preferably an alkynyl group having 2 to 6 carbon atoms, for example, an ethynyl group, propynyl group, butynyl group, isobutynyl group, pentynyl group or hexynyl group.
In the compounds of formula I provided by the invention, R3In particular formyl or guanidino;
R5specifically, it may be an amino group;
m may be 1,2,3 or 4;
x may be O or NH.
The compound shown in the formula I is shown in a formula I-1, a formula I-2 or a formula I-3,
Figure BDA0001300987850000031
the invention further provides a preparation method of the compound shown in the formula I, which comprises the following steps:
(1) carrying out azide reduction on the compound shown in the formula 3 or directly carrying out condensation reaction on the compound and Fmoc-L-isoleucine to obtain a compound shown in a formula 4;
Figure BDA0001300987850000032
in the formula 3, Bn represents benzyl, t-Bu represents tert-butyl, and Boc represents tert-butyloxycarbonyl; y represents-N3-OH or-SH, i.e. when Y represents-N3Firstly, carrying out azide reduction and then carrying out the condensation reaction, and when Y represents-OH or-SH, directly carrying out the condensation reaction;
in the formula 4, Fmoc represents 9-fluorenylmethyloxycarbonyl, and Bn, t-Bu and Boc are defined in the formula 3; x is O, S or NH;
(2) under the alkaline condition, carrying out deprotection reaction on the compound shown in the formula 4 to obtain a compound shown in a formula 5;
Figure BDA0001300987850000041
in formula 5, Bn, t-Bu, Boc and X are as defined in formula 3;
(3) carrying out condensation reaction on the compound shown in the formula 6 and benzyloxycarbonyl-L-alanine to obtain a compound shown in a formula 7;
Figure BDA0001300987850000042
in the formulas 6 and 7, the definition of Fmoc is the same as that in the formula 4, and m is a natural number between 1 and 4;
in the formula 6, R6represents-NH-, -NH-C (═ O) -NH-, or-NH-C (═ NH) -NH-;
in formula 7, Cbz represents benzyloxycarbonyl;
(4) carrying out reduction reaction on the compound shown in the formula 7 to obtain a compound shown in a formula 8;
Figure BDA0001300987850000043
in the formula 8, Fmoc is as defined in the formula 4, Cbz is as defined in the formula 7, and m is as defined in the formula 6;
(5) carrying out condensation reaction on the compound shown in the formula 5 and the compound shown in the formula 8 to obtain a compound shown in a formula 9;
Figure BDA0001300987850000044
in formula 9, Bn, t-Bu, Boc and X are as defined in formula 3, Fmoc is as defined in formula 4, Cbz is as defined in formula 7, m and R6Is as defined in formula 6;
(6) carrying out reduction reaction on the compound shown in the formula 9, and then carrying out intramolecular condensation reaction to obtain a compound shown in a formula 10;
Figure BDA0001300987850000051
of formula 10, of t-Bu, Boc and XIn the same formula 3, m and R6Is as defined in formula 6;
(7) under an acidic condition, carrying out deprotection reaction on the compound shown in the formula 10 to obtain a compound shown in a formula 11;
Figure BDA0001300987850000052
in formula 11, t-Bu and X are as defined in formula 3, m and R6Is as defined in formula 6;
(8) carrying out condensation reaction on the compound shown as the formula 11 and the compound shown as the formula 12 to obtain a compound shown as a formula 13;
Figure BDA0001300987850000053
in the formulae 12 and 13, the group R1Hydrogen, alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl with 2-15 carbon atoms, aryl or acyl;
radical R2Hydrogen, halogen, alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl with 2-15 carbon atoms, aryl, substituted aryl or acyl, wherein the substituted aryl is halogen substituted aryl or trifluoromethyl substituted aryl;
radical R4Is alkyl with 1-15 carbon atoms, alkenyl with 2-15 carbon atoms, alkynyl or aryl with 2-15 carbon atoms;
t-Bu and Boc are as defined in formula 3;
in formula 13, X is as defined in formula 3, m and R6Is as defined in formula 6;
(9) and carrying out deprotection reaction on the compound shown as the formula 13 under alkaline conditions and acidic conditions in sequence to obtain the compound shown as the formula I in the claim 1.
In the preparation method, in the step (1), the compound represented by the formula 3 can be prepared by the following method:
carrying out condensation reaction on N-Boc-O-tert-butyl-L-serine and a compound shown in a formula 1 to obtain a compound shown in a formula 3;
Figure BDA0001300987850000062
in the formula 1, Bn represents benzyl, t-Bu represents tert-butyl, and Boc represents tert-butyloxycarbonyl; y is as defined in formula 3;
the condensation reaction is carried out under the catalysis of 6-chlorobenzotriazole-1, 1,3, 3-tetramethylurea hexafluorophosphate and N, N-diisopropylethylamine;
the molar ratio of the compound shown in the formula 1 to the N-Boc-O-tert-butyl-L-serine is 1: 1.0 to 1.5;
the molar ratio of the N-Boc-O-tert-butyl-L-serine to the 6-chlorobenzotriazole-1, 1,3, 3-tetramethylurea hexafluorophosphate to the N, N-diisopropylethylamine is 1: 1.0-1.5: 1.0 to 1.5;
the condensation reaction is carried out at the temperature of 20-25 ℃ for 2-3 hours.
In the preparation method, in the step (1), azide reduction is carried out under the catalysis of triphenylphosphine;
in the step (1) and the step (3), the condensation reaction is carried out under the catalysis of carbodiimide and 4-dimethylaminopyridine;
the molar ratio of Fmoc-L-isoleucine to the compound represented by formula 3 is 1: 0.5 to 1.0;
the molar ratio of Fmoc-L-isoleucine, carbodiimide to 4-dimethylaminopyridine is 1: 1.0-1.5: 0.1 to 0.5;
the molar ratio of the benzyloxycarbonyl-L-alanine to the compound represented by formula 6 was 1: 0.5 to 1.0;
the molar ratio of the benzyloxycarbonyl-L-alanine, the carbodiimide to the 4-dimethylaminopyridine is 1: 1.0-1.5: 0.1 to 0.5;
the condensation reaction is carried out at the temperature of 20-25 ℃ for 2-3 hours.
In the preparation method, in the step (2), the alkaline condition is obtained by preparing a diethylamine acetonitrile solution with the mass percentage of 33%;
in the step (4), the reduction reaction is carried out under the catalysis of lithium hydroxide;
the molar ratio of the compound of formula 7 to the lithium hydroxide is 1: 1-5;
in the step (5), the condensation reaction is carried out under the catalysis of 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate and N, N-diisopropylethylamine;
the molar ratio of the compound represented by formula 8 to the compound represented by formula 5 is 1: 1.0 to 1.5;
the molar ratio of the compound shown in the formula 8, the 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate and the N, N-diisopropylethylamine is 1: 1.0-1.5: 0.1 to 0.5;
the condensation reaction is carried out at the temperature of 20-25 ℃ for 2-3 hours.
In the preparation method, in the step (7), the reduction reaction is carried out under the catalysis of palladium hydroxide/carbon;
the intramolecular condensation reaction is carried out under the catalysis of 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate, 1-hydroxy-7-azobenzotriazole and N, N-diisopropylethylamine;
in the step (7), the deprotection reaction is carried out in the presence of hydrochloric acid;
in the step (8), the condensation reaction is carried out under the catalysis of 3-diethoxyphosphoryl-1, 2, 3-benzene azole 4(3H) -ketone and N, N-diisopropylethylamine;
the molar ratio of the compound shown in the formula 11, the 3-diethoxyphosphoryl-1, 2, 3-benzoxazole 4(3H) -one and the N, N-diisopropylethylamine is 1: 1-2: 1 to 2.
In the step (9), the alkaline condition is obtained by modulating 33% diethylamine in acetonitrile;
the acidic condition is obtained by preparing a mixed aqueous solution of trifluoroacetic acid and triisopropylsilane.
The compound shown in the formula I provided by the invention can be used for inhibiting the growth and/or reproduction of gram-positive bacteria.
The gram-positive bacterium can be at least one of streptococcus, staphylococcus, enterococcus, corynebacterium, listeria, bacillus, erysipelothrix, actinomycetes, helicobacter pylori, legionella pneumophila, mycobacterium tuberculosis, mycobacterium avium, mycobacterium intracellulare, staphylococcus aureus, staphylococcus epidermidis, neisseria gonorrhoeae, neisseria meningitidis, streptococcus pyogenes, streptococcus faecalis, streptococcus bovis, streptococcus pneumoniae, haemophilus, pseudomonas aeruginosa, bacillus anthracis, and bacillus subtilis.
The invention also provides an inhibitor of gram-positive bacteria, the active ingredient of which is the compound shown in the formula I.
The invention has the following advantages:
based on the fact that the existing gram-positive bacteria have serious drug resistance, the invention provides a novel antibacterial compound. The invention synthesizes a series of teixobactin analogues by a solid-phase and liquid-phase combination method for the first time, and a convergent synthesis strategy is adopted to be beneficial to synthesizing a plurality of compounds. The compound with the activity equivalent to that of teixobactin is prepared by the invention.
Detailed Description
The experimental procedures used in the following examples are all conventional procedures unless otherwise specified.
Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
Example 1 preparation of Compounds of formula I-1, formula I-2 and formula I-3
(1) Preparation of Compound represented by formula 3
The reaction equation is as follows:
Figure BDA0001300987850000081
placing N-Boc-O-tert-butyl-L-serine (11mmol) in a round-bottom flask, adding dichloromethane and N, N-dimethylformamide as a mixed solvent of 50ml, adding HCTU (6-chlorobenzotriazole-1, 1,3, 3-tetramethylurea hexafluorophosphate) (11mmol) and DIEA (N, N-diisopropylethylamine) (11mmol) in the reaction solution, adding a compound shown in formula 1-1 (10mmol), stirring at room temperature for 3 hours, and adding diluted hydrochloric acid to quench the reaction. Adding 100ml dichloromethane to dilute the reaction solution, washing with saturated aqueous solution of sodium bicarbonate and sodium chloride, distilling off dichloromethane under reduced pressure, and separating the obtained product with silica gel column (petroleum ether: ethyl acetate 2: 1) to obtain the compound shown in formula 3.
(2) Preparation of Compound represented by formula 4
The reaction equation is as follows:
Figure BDA0001300987850000082
the compound represented by the formula 3 (9mmol) was dissolved in 30ml of anhydrous tetrahydrofuran, and after adding trimethylphosphorus (20mmol) and stirring at room temperature for 24 hours, excess solvent and reagent were suspended and then dissolved in 10ml of dichloromethane to be disintegrated. Fmoc-L-isoleucine (10mmol) was placed in a round-bottomed flask, EDCI (carbodiimide) (10mmol) and DMAP (4-dimethylaminopyridine) (1mmol) were added to the reaction mixture and stirred for 20min, then an intermediate dissolved in dichloromethane was added, and after stirring overnight at room temperature, the reaction was quenched by addition of dilute hydrochloric acid. Adding 100ml dichloromethane to dilute the reaction solution, washing with saturated aqueous solution of sodium bicarbonate and sodium chloride, distilling off dichloromethane under reduced pressure, and separating the obtained product with silica gel column (petroleum ether: ethyl acetate 2: 1) to obtain the compound shown in formula 4.
(3) Preparation of Compound represented by formula 5
The reaction equation is as follows:
Figure BDA0001300987850000091
placing the compound shown in the formula 4 (10mmol) in a round-bottom flask, adding 33 wt% diethylamine acetonitrile solution, stirring at room temperature for 15min, evaporating the solvent under reduced pressure, and separating the obtained product by silica gel column (petroleum ether: ethyl acetate 2: 1) to obtain the compound shown in the formula 5.
(4) Preparation of Compound represented by formula 6
The reaction equation is as follows:
Figure BDA0001300987850000092
thionyl chloride (20mmol) was added dropwise to the methanol solution at zero degrees, N-Boc-N' -Fmoc-L-lysine (10mmol) was added thereto, and after stirring overnight at room temperature, the methanol solution was distilled off under reduced pressure. The reaction mixture was diluted with 100ml of dichloromethane, washed with saturated aqueous solutions of sodium hydrogencarbonate and sodium chloride, respectively, and the dichloromethane was distilled off under reduced pressure, and the obtained product was separated by means of a silica gel column (dichloromethane: methanol 20: 1) to obtain a compound represented by formula 6.
(5) Preparation of Compound represented by formula 7
The reaction equation is as follows:
Figure BDA0001300987850000093
benzyloxycarbonyl-L-alanine (11mmol) was placed in a round-bottomed flask, methylene chloride and N, N-dimethylformamide (50 ml) as a mixed solvent, HCTU (11mmol) and DIEA (11mmol) were added to the reaction mixture, and then compound 6(10mmol) was added thereto, and after stirring at room temperature for three hours, the reaction was quenched by addition of dilute hydrochloric acid. The reaction mixture was diluted with 100ml of dichloromethane, washed with saturated aqueous solutions of sodium hydrogencarbonate and sodium chloride, respectively, and dichloromethane was distilled off under reduced pressure, and the obtained product was separated by means of a silica gel column (petroleum ether: ethyl acetate 2: 1) to obtain a compound represented by formula 7.
(6) Preparation of Compound represented by formula 8
The reaction equation is as follows:
Figure BDA0001300987850000101
placing the compound (8mmol) shown in the formula 7 in a round-bottom flask, adding tetrahydrofuran 20ml, dissolving lithium hydroxide (16mmol) in water (7ml), adding the solution into the reaction solution, stirring at room temperature for 5min, and adding diluted hydrochloric acid to quench the reaction. Diluting the reaction solution with 100ml of ethyl acetate, washing with saturated aqueous solution of sodium chloride, removing ethyl acetate by evaporation under reduced pressure, and separating the obtained product with silica gel column (dichloromethane: methanol 10: 1) to obtain the compound represented by formula 8.
(7) Preparation of Compound represented by formula 9
The reaction equation is as follows:
Figure BDA0001300987850000102
the compound represented by the formula 8 (7mmol) was placed in a round-bottomed flask, 50ml of a mixed solvent of dichloromethane and N, N-dimethylformamide was added, HATU (7mmol) and DIEA (8mmol) were added to the reaction solution, and then the compound 5(7mmol) was added thereto, and after stirring at room temperature for three hours, the reaction was quenched by addition of dilute hydrochloric acid. The reaction mixture was diluted with 100ml of dichloromethane, washed with saturated aqueous solutions of sodium hydrogencarbonate and sodium chloride, respectively, and dichloromethane was distilled off under reduced pressure, and the obtained product was separated by means of a silica gel column (petroleum ether: ethyl acetate 1: 1) to obtain a compound represented by formula 9.
(8) Preparation of Compound represented by formula 10
The reaction equation is as follows:
Figure BDA0001300987850000103
the compound represented by the formula 9 (6mmol) and palladium hydroxide/carbon (1mmol) were placed in a round-bottom flask, 50ml of methanol was added, and the mixture was stirred at room temperature for 15min under 1 atm of hydrogen gas, and the palladium hydroxide/carbon was removed by filtration, and the remaining methanol solution was spin-dried. The crude compound was obtained. Putting the crude product (1mmol) into a 500ml round-bottom flask, adding dichloromethane and N, N-dimethylformamide as a mixed solvent (300 ml), adding HATU (2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate-condensing agent (4mmol), HOAT (1-hydroxy-7-azobenzotriazole) (4mmol) and DIEA (8mmol) into the reaction solution, stirring at room temperature for 48 hours, adding dilute hydrochloric acid for quenching reaction, washing with a dilute hydrochloric acid aqueous solution and a saturated sodium chloride aqueous solution respectively, evaporating dichloromethane under reduced pressure, and separating the obtained product by a silica gel column (dichloromethane: methanol 10: 1) to obtain the compound shown in the formula 10.
(9) Preparation of Compound represented by formula 11
The reaction equation is as follows:
Figure BDA0001300987850000111
placing the compound shown in the formula 10 (0.5mmol) in a round-bottom flask, adding 5ml of 2M ethyl acetate hydrochloride solution, stirring at room temperature for 20min, adding 10ml of ethyl acetate for dilution, adding saturated sodium bicarbonate water solution for washing, drying an organic phase by using anhydrous sodium sulfate, and evaporating the ethyl acetate solvent under reduced pressure to obtain a crude compound shown in the formula 11.
(10) Preparation of Compound represented by formula 12
The reaction equation is as follows:
Figure BDA0001300987850000112
according to the method for polypeptide solid phase synthesis, 2-Cl resin (0.5mmol) is placed in a solid phase synthesis tube, dichloromethane and N, N-dimethylformamide are added for activation for 20min, the solvent is extracted, Fmoc-L-isoleucine (5mmol), DIEA (5mmol) are dissolved in DMF and added to a reaction instrument, stirring is carried out for 2h, the solvent is extracted, Fmoc is removed by 20% piperidine solution for 15min, Fmoc-D-isoleucine (1.5mmol), HATU (1.5mmol), DIEA (3mmol) are added to a reaction vessel and stirred at room temperature for 50 min. Next, the terminal amino acid of N-Fmoc-N '-trityl-D-glutamine, Fmoc-L-serine, Fmoc-L-isoleucine, and Boc-D-4,4' -diphenylaniline or N-t-butoxycarbonyl-N-methyl-D-phenylalanine was attached under the same conditions. Finally, 25 wt% of trifluoroethanol (diluted with dichloromethane) was added to the reaction tube, stirred at room temperature for 4 hours, and the solvent was spin-dried to obtain the compound represented by formula 12.
(11) Preparation of Compound represented by formula 13
The reaction equation is as follows:
Figure BDA0001300987850000121
a compound represented by formula 11 (0.5mmol) and a compound represented by formula 12 (0.5mmol) were placed in a round-bottomed flask, tetrahydrofuran and N, N-dimethylformamide were added to a mixed solvent (10 ml), placed in an ice-water bath, followed by addition of DEPBT (1mmol) and DIEA (1mmol), reacted for 1 hour, and then stirred at room temperature overnight. The reaction was quenched by addition of dilute hydrochloric acid. Diluting the reaction mixture with 100ml of ethyl acetate, washing with saturated aqueous solution of sodium chloride, removing ethyl acetate by evaporation under reduced pressure, and separating the obtained product with silica gel column (dichloromethane: methanol 10: 1) to obtain the compound represented by formula 13.
(12) Preparation of the target Compound
The reaction equation is as follows:
Figure BDA0001300987850000131
the compound represented by formula 13 (0.2mmol) was placed in a round-bottom flask, 33 wt% diethylamine acetonitrile solvent was added to the round-bottom flask, and stirred at room temperature for 15 minutes, after Fmoc was completely removed, the solvent was dried by spinning, and the crude product was not passed through a column. Reacting trifluoroacetic acid: triisopropylsilane: adding 5ml of water (95: 2.5: 2.5, volume ratio) into a flask containing the crude product, stirring at room temperature for 1 hour, blowing the solvent with nitrogen, adding glacial ethyl ether for precipitation, centrifuging to obtain the crude product, separating by reverse phase HPLC, and freeze-drying to obtain the final pure compound of formula I-1, wherein the group R is1Is hydrogen, R2Is phenyl, R3Is formyl, R4Is ethyl, R5Is n-butylamino.
The characterization data for the compounds of formula I-1 are as follows:
1H-NMR(400MHz,CD3OD)(ppm)7.70-7.55(m,4H),7.46-7.30(m,5H),5.60-5.50(m,1H),4.55-4.45(m,1H),4.45-4,28(m,4H),4.28-4.02(m,5H),3.98-3.70(m,4H),3.20-3.10(m,2H),2.95-2.85(m,2H),2.38-2.28(m,2H),2.20-1.90(m,3H),1.90-1.60(m,8H),1.60-1.40(m,7H),1.40-1.10(m,8H),1.10-0.85(m,18H),0.85-0.70(m,6H)HRMS(ESI)[M+H]+:1277.58,found 1277.74.
from the above data, the compounds prepared have the correct structure.
Obtaining a compound represented by the formula I-2 by the above method, wherein the compound represented by the formula 1-1 is replaced with the compound represented by the formula 1-2 in the step (1);
formula I-2, radical R1Is hydrogen, R2Is phenyl, R3Is guanidino, R4Is ethyl, R5Is n-butylamino.
Figure BDA0001300987850000132
The characterization data for the compounds of formula I-2 are as follows:
1H-NMR(400MHz,CD3OD)(ppm)7.40-7.20(m,5H),5.60-5.50(m,1H),4.55-4.45(m,1H),4.45-4,28(m,4H),4.28-4.02(m,5H),3.98-3.70(m,4H),3.20-3.10(m,2H),2.95-2.85(m,2H),2.69(s,1H)2.38-2.28(m,2H),2.20-1.90(m,3H),1.90-1.60(m,8H),1.60-1.40(m,7H),1.40-1.10(m,8H),1.10-0.85(m,18H),0.85-0.70(m,6H)HRMS(ESI)[M+H]+:1305.73,found 1305.72.
from the above data, the compounds prepared have the correct structure.
Obtaining a compound represented by the formula I-3 by the above method, wherein the compound represented by the formula 1-1 is replaced with the compound represented by the formula 1-2 in the step (1);
formula I-3, group R1Is hydrogen, R2Is p-methylphenyl, R3Is formyl, R4Is ethyl, R5Is n-butylamino;
the characterization data for the compounds of formula I-3 are as follows:
1H-NMR(400MHz,CD3OD)(ppm)7.70-7.55(m,4H),7.46-7.30(m,5H),5.60-5.50(m,1H),4.55-4.45(m,1H),4.45-4,28(m,4H),4.28-4.02(m,5H),3.98-3.70(m,4H),3.20-3.10(m,2H),2.95-2.85(m,2H),2.47(s,3H),2.38-2.28(m,2H),2.20-1.90(m,3H),1.90-1.60(m,8H),1.60-1.40(m,5H),1.40-1.10(m,8H),1.10-0.85(m,18H),0.85-0.70(m,6H)HRMS(ESI)[M+H]+:1291.72,found 1291.74.
from the above data, the compounds prepared have the correct structure.
Example 2 Activity test of Compounds of formula I-1, formula I-2 and formula I-3
Compounds were tested for MIC according to CLSI guidelines.
Culturing bacteria and selecting THY culture medium, and testing and selecting MHB culture medium. The medium used was supplemented with 0.002% tween 80 to inhibit drug adherence. Cell concentration was adjusted to 5X 105Each per milliliter. The concentration of the compound is 8 mug/ml, 4 mug/ml, 2 mug/ml, 1 mug/ml, 0.5 mug/ml and 0.25 mug/ml from high to low in sequence, and after the compound is cultured for 20 hours at 37 ℃, the drug concentration of a clear hole of a culture medium is the minimum inhibitory concentration.
The test results are shown in table 1.
TABLE 1 MIC (μ g/ml) for each compound
Figure BDA0001300987850000141
Figure BDA0001300987850000151
As can be seen from the data in Table 1, the antibacterial activity of the compound prepared according to the present invention is comparable to that of teixobactin, and the molecular structural formula and synthesis cost are low.

Claims (7)

1. A compound of the formula I,
Figure FDA0002710946180000011
in the formula I, the radical R1Is hydrogen;
radical R2Is phenyl or C1-C6 alkyl substituted phenyl;
radical R3Is formyl or guanidino;
radical R4Is C1-C6 alkyl;
radical R5Is amino; m is 4;
x is NH.
2. A process for the preparation of a compound of formula I comprising the steps of:
(1) carrying out azide reduction on the compound shown in the formula 3, and carrying out condensation reaction on the compound and Fmoc-L-isoleucine to obtain a compound shown in a formula 4;
Figure FDA0002710946180000012
in the formula 3, Bn represents benzyl, t-Bu represents tert-butyl, and Boc represents tert-butyloxycarbonyl; y represents-N3Firstly, carrying out azide reduction and then carrying out the condensation reaction;
in the formula 4, Fmoc represents 9-fluorenylmethyloxycarbonyl, and Bn, t-Bu and Boc are defined in the formula 3; x is NH;
(2) under the alkaline condition, carrying out deprotection reaction on the compound shown in the formula 4 to obtain a compound shown in a formula 5;
Figure FDA0002710946180000013
in formula 5, Bn, t-Bu and Boc are as defined in formula 3, and X is as defined in formula 4;
(3) carrying out condensation reaction on the compound shown in the formula 6 and benzyloxycarbonyl-L-alanine to obtain a compound shown in a formula 7;
Figure FDA0002710946180000021
in the formulas 6 and 7, Fmoc is as defined in the formula 4, and m is 4;
in the formula 6, R6represents-NH-;
in formula 7, Cbz represents benzyloxycarbonyl;
(4) carrying out reduction reaction on the compound shown in the formula 7 to obtain a compound shown in a formula 8;
Figure FDA0002710946180000022
in formula 8, Fmoc is as defined in formula 4, Cbz is as defined in formula 7, m and R6Is as defined in formula 6;
(5) carrying out condensation reaction on the compound shown in the formula 5 and the compound shown in the formula 8 to obtain a compound shown in a formula 9;
Figure FDA0002710946180000023
in formula 9, Bn, t-Bu and Boc are as defined in formula 3, X and Fmoc are as defined in formula 4, Cbz is as defined in formula 7, m and R6Is as defined in formula 6;
(6) carrying out reduction reaction on the compound shown in the formula 9, and then carrying out intramolecular condensation reaction to obtain a compound shown in a formula 10;
Figure FDA0002710946180000024
in formula 10, t-Bu and Boc are as defined in formula 3, X is as defined in formula 4, m and R6Is as defined in formula 6;
(7) under an acidic condition, carrying out deprotection reaction on the compound shown in the formula 10 to obtain a compound shown in a formula 11;
Figure FDA0002710946180000031
in formula 11, t-Bu is as defined in formula 3, X is as defined in formula 4, m and R6Is as defined in formula 6;
(8) carrying out condensation reaction on the compound shown as the formula 11 and the compound shown as the formula 12 to obtain a compound shown as a formula 13;
Figure FDA0002710946180000032
in the formulae 12 and 13, the group R1Is hydrogen;
radical R2Is phenyl or C1-C6 alkyl substituted phenyl;
radical R4Is C1-C6 alkyl;
t-Bu and Boc are as defined in formula 3;
in formula 13, X is as defined in formula 4, m and R6Is as defined in formula 6;
(9) and carrying out deprotection reaction on the compound shown as the formula 13 under alkaline conditions and acidic conditions in sequence to obtain the compound shown as the formula I in the claim 1.
3. The method of claim 2, wherein: in the step (1), the compound represented by the formula 3 is prepared by the following method:
carrying out condensation reaction on N-Boc-O-tert-butyl-L-serine and a compound shown in a formula 1 to obtain a compound shown in a formula 3;
Figure FDA0002710946180000041
in formula 1, Bn represents a benzyl group;
the condensation reaction is carried out under the catalysis of 6-chlorobenzotriazole-1, 1,3, 3-tetramethylurea hexafluorophosphate and N, N-diisopropylethylamine;
the molar ratio of the compound shown in the formula 1 to the N-Boc-O-tert-butyl-L-serine is 1: 1.0 to 1.5;
the molar ratio of the N-Boc-O-tert-butyl-L-serine to the 6-chlorobenzotriazole-1, 1,3, 3-tetramethylurea hexafluorophosphate to the N, N-diisopropylethylamine is 1: 1.0-1.5: 1.0 to 1.5;
the condensation reaction is carried out at the temperature of 20-25 ℃ for 2-3 hours.
4. The production method according to claim 2 or 3, characterized in that: in the step (1), azide reduction is carried out under the catalysis of triphenylphosphine;
in the step (1) and the step (3), the condensation reaction is carried out under the catalysis of carbodiimide and 4-dimethylaminopyridine;
the molar ratio of Fmoc-L-isoleucine to the compound represented by formula 3 is 1: 0.5 to 1.0;
the molar ratio of Fmoc-L-isoleucine, carbodiimide to 4-dimethylaminopyridine is 1: 1.0-1.5: 0.1 to 0.5;
the molar ratio of the benzyloxycarbonyl-L-alanine to the compound represented by formula 6 was 1: 0.5 to 1.0;
the molar ratio of the benzyloxycarbonyl-L-alanine, the carbodiimide to the 4-dimethylaminopyridine is 1: 1.0-1.5: 0.1 to 0.5;
the condensation reaction is carried out at the temperature of 20-25 ℃ for 2-3 hours.
5. The method of claim 4, wherein: in the step (2), the alkaline condition is obtained by modulating 33% diethylamine in acetonitrile;
in the step (4), the reduction reaction is carried out under the catalysis of lithium hydroxide;
the molar ratio of the compound of formula 7 to the lithium hydroxide is 1: 1-5;
in the step (5), the condensation reaction is carried out under the catalysis of 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate and N, N-diisopropylethylamine;
the molar ratio of the compound represented by formula 8 to the compound represented by formula 5 is 1: 1.0 to 1.5;
the molar ratio of the compound shown in the formula 8, the 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate and the N, N-diisopropylethylamine is 1: 1.0-1.5: 0.1 to 0.5;
the condensation reaction is carried out at the temperature of 20-25 ℃ for 2-3 hours.
6. The method of claim 5, wherein: in the step (6), the reduction reaction is carried out under the catalysis of palladium hydroxide/carbon;
the intramolecular condensation reaction is carried out under the catalysis of 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate, 1-hydroxy-7-azobenzotriazole and N, N-diisopropylethylamine;
in the step (7), the deprotection reaction is carried out in the presence of hydrochloric acid;
in the step (8), the condensation reaction is carried out under the catalysis of 3-diethoxyphosphoryl-1, 2, 3-benzene azole 4(3H) -ketone and N, N-diisopropylethylamine;
the molar ratio of the compound shown in the formula 11, the 3-diethoxyphosphoryl-1, 2, 3-benzoxazole 4(3H) -one and the N, N-diisopropylethylamine is 1: 1-2: 1-2;
in the step (9), the alkaline condition is obtained by modulating 33% diethylamine in acetonitrile;
the acidic condition is obtained by preparing a mixed aqueous solution of trifluoroacetic acid and triisopropylsilane.
7. Use of a compound according to claim 1 for the preparation of a product for inhibiting the growth and/or reproduction of gram-positive bacteria;
the gram-positive bacteria is at least one of enterococcus, corynebacterium, listeria, erysipelothrix, actinomycetes, mycobacterium tuberculosis, mycobacterium avium, mycobacterium intracellulare, staphylococcus aureus, staphylococcus epidermidis, streptococcus pyogenes, streptococcus faecalis, streptococcus bovis, streptococcus pneumoniae, bacillus anthracis and bacillus subtilis.
CN201710363824.9A 2017-05-22 2017-05-22 Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis Active CN108948147B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201710363824.9A CN108948147B (en) 2017-05-22 2017-05-22 Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis
PCT/CN2017/114644 WO2018214463A1 (en) 2017-05-22 2017-12-05 Novel antibiotic and preparation method, use, and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710363824.9A CN108948147B (en) 2017-05-22 2017-05-22 Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis

Publications (2)

Publication Number Publication Date
CN108948147A CN108948147A (en) 2018-12-07
CN108948147B true CN108948147B (en) 2020-11-17

Family

ID=64396121

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710363824.9A Active CN108948147B (en) 2017-05-22 2017-05-22 Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis

Country Status (2)

Country Link
CN (1) CN108948147B (en)
WO (1) WO2018214463A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112300247B (en) * 2019-07-24 2022-12-27 广东东阳光药业有限公司 Teixobactin analogue and application thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1814614A (en) * 2005-02-06 2006-08-09 中国人民解放军军事医学科学院毒物药物研究所 Nucleic acid, peptide nucleicacid derivatives and their use
EP2925772B1 (en) * 2012-12-03 2018-02-07 Novobiotic Pharmaceuticals, LLC Novel depsipeptide and uses thereof
EP3443078A4 (en) * 2016-04-15 2020-04-15 The Regents of The University of California Antimicrobial compositions
CN106632604B (en) * 2016-12-22 2021-03-19 深圳先进技术研究院 Teixobatin analogue and preparation method and application thereof

Also Published As

Publication number Publication date
CN108948147A (en) 2018-12-07
WO2018214463A1 (en) 2018-11-29

Similar Documents

Publication Publication Date Title
Socha et al. Diversity-oriented synthesis of cyclic acyldepsipeptides leads to the discovery of a potent antibacterial agent
CN107778352B (en) Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis
CA2145826C (en) Lipopeptide derivatives, a process for their preparation and their use
AU2009330245B2 (en) Novel antibacterial agents for the treatment of gram positive infections
JP2003517004A (en) Daptomycin analogs as antibacterial agents
JP2003517480A (en) Lipopeptides as antibacterial agents
JP6452698B2 (en) Vancomycin derivatives, production methods and applications thereof
TW201026699A (en) 5-amino-2-(1-hydroxy-ethyl)-tetrahydropyran derivatives
EP2238102A1 (en) Novel semi-synthetic glycopeptides as antibacterial agents
CN108948147B (en) Novel antibiotic for treating drug-resistant gram-positive bacteria and tuberculosis
CN108137574A (en) Hydroxyalkyl thiadiazoles derivative
EP2367820B1 (en) Novel antimicrobial agents
JP2020500943A (en) Antimicrobial peptide
CA2831799A1 (en) Oxazolidinone derivative and pharmaceutical composition including the same
JP5391373B2 (en) New antibacterial compounds
CN109651352B (en) Dimeric indole alkaloid compounds, preparation method thereof and application thereof in preparation of antibacterial drugs
CN112321580B (en) Oxazole linked triazole medicine molecule for sterilization and disinfection and preparation method and application thereof
CN1427846A (en) Uridine derivatives as antibiotics
AU2018321845B2 (en) Metabolically stable N-acylaminooxadiazoles useful as antibacterial agents
US11180501B2 (en) Crystal form of β-lactamase inhibitor and preparation method therefor
Guan et al. Single Modification at the N‐Terminus of Norvancomycin to Combat Drug‐Resistant Gram‐Positive Bacteria
WO2021205372A2 (en) Antibacterial lipopeptides, pharmaceutical composition and cosmetic composition comprising them, and uses thereof
US20190345199A1 (en) Lipolanthipeptides and their uses as antimicrobial agents
Koteva et al. Synthetic Biology Facilitates Semisynthetic Development of Type V Glycopeptide Antibiotics Targeting Vancomycin-Resistant Enterococcus
WO2015172047A1 (en) Cyclic peptide compounds and related methods, salts and compositions

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