CN109535086B - Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound - Google Patents

Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound Download PDF

Info

Publication number
CN109535086B
CN109535086B CN201910083408.2A CN201910083408A CN109535086B CN 109535086 B CN109535086 B CN 109535086B CN 201910083408 A CN201910083408 A CN 201910083408A CN 109535086 B CN109535086 B CN 109535086B
Authority
CN
China
Prior art keywords
quinoxaline
ketone
ester
reaction
cdcl
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.)
Expired - Fee Related
Application number
CN201910083408.2A
Other languages
Chinese (zh)
Other versions
CN109535086A (en
Inventor
何卫民
谢龙勇
包文虎
王毅
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.)
Hunan University of Science and Engineering
Original Assignee
Hunan University of Science and Engineering
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 Hunan University of Science and Engineering filed Critical Hunan University of Science and Engineering
Priority to CN201910083408.2A priority Critical patent/CN109535086B/en
Publication of CN109535086A publication Critical patent/CN109535086A/en
Application granted granted Critical
Publication of CN109535086B publication Critical patent/CN109535086B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • C07D241/40Benzopyrazines
    • C07D241/44Benzopyrazines with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a method for synthesizing a quinoxaline-2 (1H) -ketone C-3 site carboxylate compound, which is characterized in that quinoxaline-2 (1H) -ketone derivatives, hydrazino carboxylate and persulfate react in one pot to generate the quinoxaline-2 (1H) -ketone C-3 site carboxylate compound in an air atmosphere.

Description

Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound
Technical Field
The invention relates to a method for synthesizing a quinoxaline-2 (1H) -ketone C-3 site carboxylate compound, in particular to a method for synthesizing a quinoxaline-2 (1H) -ketone C-3 site carboxylate compound with high selectivity by using hydrazino carboxylate as a carboxylate source and directly oxidizing and removing hydrazine to replace C-3 hydrogen in an air atmosphere, belonging to the technical field of synthesis of organic intermediates.
Background
The quinoxaline-2 (1H) -ketone compound is a common pharmacophore in the field of pharmaceutical research, and the derivative with the parent structure has various physiological and pharmacological activities, is widely used as an anticancer drug, an antitumor drug, an antibacterial drug and the like, is a potential multipurpose lead compound, and has wide development and application prospects. Indeed, the quinoxalinone and modified functional groups on the parent quinoxalinone structure together affect the drug and biological activity of the molecule, not just the parent quinoxalinone structure itself. Therefore, modification of various substituents on the parent quinoxalinone structure and study of the structure-activity relationship are becoming hot points of study. In the past few years, considerable progress has been made in the functionalization of quinoxaline-2 (1H) -ones with C3, including C-H bond alkylation, arylation, acylation, amination, azidation, quinolination, trifluoromethylation, phosphonylation, and the like. Quinoxaline-3-carboxylic acid esters not only have excellent biological and pharmaceutical activity, but also are valuable intermediates for the construction of a variety of functionalized quinoxaline derivatives. The conventional scheme for preparing N-substituted quinoxaline-3-carboxylic acid salts is based on a condensation-cyclization between o-phenylenediamine and a 1, 3-dicarbonyl compound and a subsequent N-substitution reaction in the presence of a base (scheme 1), however, this process is limited by the selection range of o-phenylenediamine, and the use of asymmetric diamines as starting materials inevitably leads to isomeric products. Recently, Yu and colleagues have reported for the first time the one-step synthesis of N-substituted quinoxaline-3-carboxylate compounds by free-radical cyclisation of alkyl 2- (N-substituted carboxy-ethyl) -2-chloroiminoacetates (scheme 2). However, this method still has some disadvantages: if high-activity raw materials are needed, the steps are complicated, and the reaction system is relatively complex and expensive. Given the abundance and ready availability of quinoxalin-2 (1H) -ones, the synthesis of N-substituted quinoxaline-3-carboxylic acid esters by direct introduction of an ester group is undoubtedly a simple strategy. However, at present, no relevant report is found.
Scheme 1:
Figure GDA0002407513420000011
scheme 2:
Figure GDA0002407513420000021
disclosure of Invention
Aiming at the defects of the existing synthesis method of the quinoxaline-2 (1H) -ketone C-3 site carboxylate compound, the invention aims to provide the synthesis method of the quinoxaline-2 (1H) -ketone C-3 site carboxylate compound with important physiological activity, and the method has the advantages of high yield, low cost, environmental protection and contribution to industrial production and application.
In order to realize the technical purpose, the invention provides a synthesis method of a quinoxaline-2 (1H) -ketone C-3 carboxylic ester compound, which comprises the step of carrying out one-pot reaction on a quinoxaline-2 (1H) -ketone derivative, hydrazino carboxylic ester and persulfate in an air atmosphere to generate the quinoxaline-2 (1H) -ketone C-3 carboxylic ester compound.
In a preferred embodiment, the quinoxalin-2 (1H) -one derivative has the structure of formula 1;
Figure GDA0002407513420000022
the hydrazinocarboxylic acid ester has the structure of formula 2:
Figure GDA0002407513420000023
the quinoxaline-2 (1H) -one C-3 carboxylate compound has the structure of formula 3:
Figure GDA0002407513420000024
wherein the content of the first and second substances,
R1selected from H, C1~C10The alkyl group (e.g., a straight-chain alkyl group such as methyl, ethyl, propyl, octyl, etc., and an alkyl group having 3 or more carbon atoms also includes isomers such as a branched alkyl group, specifically, isobutyl, isooctyl, etc.), C2~C10The alkylene group (may be an aliphatic chain containing one or more alkenes, such as ethylene, propylene, etc.), C2~C10Alkynyl (may be aliphatic chain containing one or more alkynyl groups, such as ethynyl, propynyl, etc.), benzyl, C2~C10Ester group (methoxyacyl group, ethoxyacyl group, butoxyacyl group and the like) of (a) or phenyl group;
R2is selected from C1~C10The alkyl group (e.g., a straight-chain alkyl group such as methyl, ethyl, propyl, octyl, etc., and an alkyl group having 3 or more carbon atoms includes isomers such as a branched alkyl group, specifically, isobutyl, isooctyl, etc.), a halogen (e.g., fluorine, chlorine, bromine, or iodine), trifluoromethyl, C2~C10Acyl (formyl, acetyl, propionyl, etc.), C2~C10An ester group (methoxyacyl group, ethoxyacyl group, butoxyacyl group and the like) or a nitro group of (1);
R3is selected from C1~C10The alkyl group of (2) includes straight-chain alkyl groups such as methyl, ethyl, propyl, and octyl, and isomers such as branched alkyl groups, specifically isobutyl, isooctyl, and the like, are also included in the alkyl group having 3 or more carbon atoms.
In a preferred embodiment, the molar ratio of quinoxaline-2 (1H) -one derivative, hydrazinocarboxylic acid ester and persulfate is 1: 1-2: 2 to 4. The most preferred molar ratio is 1: 1.5: 3.
according to the preferable scheme, the persulfate is potassium persulfate, and a large number of experiments show that other common persulfate such as sodium persulfate, ammonium persulfate and the like cannot meet the requirements.
In a preferred embodiment, the reaction conditions are: reacting for 5-15 hours at 80-100 ℃. Most preferably, the reaction is carried out at a temperature of 85 to 95 ℃ for 8 to 10 hours.
In a preferred embodiment, the reaction employs acetonitrile as a reaction medium, the acetonitrile is a benign solvent in the technical scheme of the invention, and other common solvents such as methanol, DCE and DMF are non-benign solvents.
In a preferred scheme, the molar concentration of the quinoxaline-2 (1H) -ketone derivative in an acetonitrile reaction medium is 0.1-0.3 mmol/mL. The most preferred molar concentration is 0.2 mmol/mL.
The route of the addition reaction of the quinoxaline-2 (1H) -ketone derivative, the hydrazinocarboxylic ester and the potassium persulfate is as follows:
Figure GDA0002407513420000031
the invention also provides a reasonable reaction mechanism. The reaction of methyl carbazate with quinoxalin-2 (1H) -one is illustrated. Methyl carbazate (2) with sulfate anion radical (generated in situ from persulfate anion cleavage) to form ester radical and release HSO4 -Anions and molecular nitrogen. Ester radical attack of C3 into N-methylquinoxaline-2 (1H) -one (1) forms a radical intermediate (A) which is subsequently hydrogen-abstracted/oxidized by sulfate anions to give the desired product (3).
Figure GDA0002407513420000032
Compared with the prior art, the technical scheme of the invention has the beneficial technical effects that:
1) the invention firstly synthesizes the C-3 site carboxylate compound of quinoxaline-2 (1H) -ketone by one step of addition reaction of quinoxaline-2 (1H) -ketone derivatives, hydrazino carboxylate and potassium persulfate.
2) The invention adopts the hydrazino carboxylic ester as the esterification reagent and the potassium persulfate as the oxidant, and has the advantages of easily obtained sources and low price;
3) the quinoxaline-2 (1H) -ketone derivative has wide selectivity and good functional group compatibility, and is easy to modify various groups;
4) the invention does not use transition metal and various acid-base additives, has high reaction selectivity, and the product is easy to separate and purify
Drawings
FIG. 1 is a nuclear magnetic hydrogen spectrum of 1-methyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester;
FIG. 2 is a nuclear magnetic carbon spectrum of methyl 1-methyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylate.
Detailed Description
The following specific examples are intended to further illustrate the present disclosure, but not to limit the scope of the claims.
Comparative example:
the following control experiment groups 1 to 16 all react according to the following reaction equation:
Figure GDA0002407513420000041
the specific operation steps are as follows: 1-methyl-3, 4-dihydro-2 (1H) -quinoxalinone (1 equivalent, 0.1mmol), methyl carbazinate, potassium persulfate and solvent are sequentially added into a 5mL round-bottom flask, and the obtained mixed solution is reacted in a heating stirring reaction device. The reaction progress is followed by a thin layer chromatography plate, after the reaction is finished, the reaction is cooled to room temperature, 3ml of water is added for dilution, the reactant is extracted by ethyl acetate, and the yield is analyzed by nuclear magnetic crude spectrum.
Figure GDA0002407513420000042
Figure GDA0002407513420000051
In the table, experimental groups 1-6 investigate the influence of various oxidants on the addition reaction of the quinoxaline-2 (1H) -ketone derivative and the hydrazinocarboxylic acid methyl ester, and experimental data show that the reaction is very sensitive to the types of the oxidants, and the ideal product yield can be obtained only by using potassium persulfate.
In the table, experimental groups 1 and 7-12 investigate the influence of the reaction medium on the addition reaction of the quinoxaline-2 (1H) -ketone derivative and the hydrazinocarboxylic acid methyl ester, and experiments show that acetonitrile is the optimal reaction medium for the reaction.
In the table, experiment groups 1 and 13-14 investigate the influence of the amount of esterification reagent of methyl hydrazinocarboxylate on the addition reaction of quinoxaline-2 (1H) -one derivatives and methyl hydrazinocarboxylate, and experiments show that the optimal molar amount of methyl hydrazinocarboxylate is 1.5 equivalents, the yield of the target product is not obviously increased when the molar amount is too high, and the yield of the target product is obviously reduced when the molar amount is too low.
In the table, experiment groups 1 and 15-16 investigate the influence of the dosage of potassium persulfate serving as an oxidant on the addition reaction of the quinoxaline-2 (1H) -one derivative and the hydrazinocarboxylic acid methyl ester, and experiments show that the optimal molar dosage of the potassium persulfate is 3 equivalents, the yield of a target product is not obviously increased when the dosage is too high, and the yield of the target product is obviously reduced when the dosage is too low.
In the above table, experiment groups 1 and 17-18 investigate the influence of the molar concentration of the quinoxaline-2 (1H) -ketone derivative on the addition reaction of the quinoxaline-2 (1H) -ketone derivative and the methyl carbazate, and experiments show that the molar concentration of the quinoxaline-2 (1H) -ketone derivative is 0.2mmol/mL, which is the optimum substrate molar concentration for the reaction, when the molar concentration is too high, the yield of the target product is not obviously increased, and when the molar concentration is too low, the yield of the target product is obviously reduced.
In the table, experiment groups 1 and 19-20 investigate the influence of the reaction temperature on the addition reaction of the quinoxaline-2 (1H) -ketone derivative and the hydrazinocarboxylic acid methyl ester, and experiments show that the reaction temperature is the optimum reaction temperature at 90 ℃, the yield of the target product is not obviously increased when the reaction temperature is too high, and the yield of the target product is obviously reduced when the reaction temperature is too low.
Examples 1 to 18
The following examples 1 to 18 were all reacted according to the following reaction equations:
Figure GDA0002407513420000061
the specific operation steps are as follows: quinoxaline-2 (1H) -one derivative (1 equivalent, 0.2mmol), hydrazinocarboxylic acid ester (1.5 equivalent, 0.3mmol), potassium persulfate (3 equivalent, 0.6mmol) and acetonitrile (1mL) are sequentially added into a 10mL round-bottom flask, and the obtained mixed solution is heated and stirred for reaction at 90 ℃ under the condition of air. The reaction progress is followed by thin-layer chromatography plates, generally for a reaction time of 8 to 12 hours. After the reaction is finished, cooling to room temperature, adding 3ml of water for dilution, extracting a reactant by ethyl acetate, performing nuclear magnetic crude spectrum analysis on the yield, concentrating the filtrate by using a rotary evaporator, and performing column chromatography purification by using Petroleum Ether (PE)/Ethyl Acetate (EA) as an eluent and silica gel (200-mesh and 300-mesh sieve).
Example 1
Raw materials:
Figure GDA0002407513420000071
and (3) target products:
Figure GDA0002407513420000072
1-methyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield was 84%.
1H NMR(400MHz,CDCl3):δ=7.94(dd,J1=8.0Hz,J2=1.2Hz,1H),7.69–7.64(m,1H),7.41–7.35(m,2H),4.02(s,3H),3.72(s,3H);
13C NMR(100MHz,CDCl3):δ=164.0,152.5,148.4,134.1,132.7,131.8,131.3,124.2,113.9,53.2,29.2;
HRMS(ESI)m/z calcd.for C11H11N2O3[M+H]+:219.0764,found 219.0762.
Example 2
Raw materials:
Figure GDA0002407513420000073
and (3) target products:
Figure GDA0002407513420000074
1-ethyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield was 87%.
1H NMR(400MHz,CDCl3):δ=7.95(dd,J1=8.0Hz,J2=1.2Hz,1H),7.68–7.64(m,1H),7.40–7.36(m,2H),4.34(q,J=7.2Hz,2H),4.02(s,3H),1.39(t,J=7.2Hz,3H);
13C NMR(100MHz,CDCl3):δ=164.1,152.1,148.4,133.1,132.6,132.1,131.5,124.0,113.7,53.1,37.6,12.3;
HRMS(ESI)m/z calcd.for C12H13N2O3[M+H]+:233.0921,found 233.0917.
Example 3
Raw materials:
Figure GDA0002407513420000075
and (3) target products:
Figure GDA0002407513420000076
1-pentyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield is 92%
1H NMR(400MHz,CDCl3):δ=7.93(dd,J1=8.0Hz,J2=1.6Hz,1H),7.66–7.62(m,1H),7.38–7.33(m,2H),4.24(t,J=8.0Hz,2H),4.01(s,3H),1.79–1.71(m,2H),1.45–1.35(m,4H),0.90(t,J=7.2Hz,3H);
13C NMR(100MHz,CDCl3):δ=164.1,152.2,148.3,133.3,132.5,132.1,131.4,124.0,113.8,53.1,42.5,28.9,26.8,22.2,13.8;
HRMS(ESI)m/z calcd.for C15H19N2O3[M+H]+:275.1390,found 275.1390.
Example 4
Raw materials:
Figure GDA0002407513420000081
and (3) target products:
Figure GDA0002407513420000082
1-propenyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield is 82 percent
1H NMR(400MHz,CDCl3):δ=7.96(dd,J1=8.0Hz,J2=1.6Hz,1H),7.65–7.61(m,1H),7.41–7.33(m,2H),5.97–5.88(m,1H),5.31–5.19(m,2H),4.94–4.92(m,2H),4.03(s,3H);
13C NMR(100MHz,CDCl3):δ=164.0,152.1,148.4,133.4,132.6,132.0,131.4,130.1,124.2,118.8,114.5,53.2,44.7;
HRMS(ESI)m/z calcd.for C13H13N2O3[M+H]+:245.0921,found 245.0925.
Example 5
Raw materials:
Figure GDA0002407513420000083
and (3) target products:
Figure GDA0002407513420000084
1-cinnamyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield is 78 percent
1H NMR(400MHz,CDCl3):δ=7.98(dd,J1=8.0Hz,J2=1.6Hz,1H),7.67–7.63(m,1H),7.46–7.44(m,1H),7.42–7.38(m,1H),7.34–7.27(m,4H),7.25–7.22(m,1H),6.65(d,J=16.0Hz,1H),6.30–6.23(m,1H),5.09(dd,J1=5.6Hz,J2=1.6Hz,2H),4.05(s,3H);
13C NMR(100MHz,CDCl3):δ=164.0,152.2,148.4,135.7,134.3,132.7,131.5,128.6,128.2,126.5,124.3,121.3,114.4,53.2,44.4;
HRMS(ESI)m/z calcd.for C19H17N2O3[M+H]+:321.1234,found 321.1236.
Example 6
Raw materials:
Figure GDA0002407513420000091
and (3) target products:
Figure GDA0002407513420000092
1- (2-prop-ynyl) -2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield is 83 percent
1H NMR(400MHz,CDCl3):δ=7.98(dd,J1=8.0Hz,J2=1.6Hz,1H),7.73–7.69(m,1H),7.53–7.51(m,1H),7.46–7.42(m,1H),5.08(d,J=2.4Hz,2H),4.03(s,3H),2.31(t,J=2.4Hz,1H);
13C NMR(100MHz,CDCl3):δ=163.7,151.5,148.2,132.8,132.7,132.1,124.6,121.4,114.5,76.1,73.7,53.3,31.6;
HRMS(ESI)m/z calcd.for C13H11N2O3[M+H]+:243.0764,found 243.0769.
Example 7
Raw materials:
Figure GDA0002407513420000093
and (3) target products:
Figure GDA0002407513420000094
1-benzyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid methyl ester; the yield is 86 percent
1H NMR(400MHz,CDCl3):δ=7.95(dd,J1=8.0Hz,J2=1.6Hz,1H),7.56–7.51(m,1H),7.36–7.25(m,7H),5.51(s,2H),4.04(s,3H);
13C NMR(100MHz,CDCl3):δ=164.0,152.6,148.5,134.6,133.5,132.6,132.1,131.4,129.0,127.9,127.0,124.2,114.7,53.2,46.0;
HRMS(ESI)m/z calcd.for C17H15N2O3[M+H]+:295.1077,found 295.1072.
Example 8
Raw materials:
Figure GDA0002407513420000095
and (3) target products:
Figure GDA0002407513420000096
1- (2-ethoxy-2-oxoethyl) -2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 74 percent
1H NMR(400MHz,CDCl3):δ=7.96(dd,J1=8.0Hz,J2=1.6Hz,1H),7.64–7.60(m,1H),7.41–7.37(m,1H),7.11(dd,J1=8.4Hz,J2=0.8Hz,1H),5.03(s,2H),4.23(q,J=7.2Hz,2H),4.01(s,3H),1.25(t,J=7.2Hz,3H);
13C NMR(100MHz,CDCl3):δ=166.5,163.6,152.1,147.9,133.3,132.9,131.8,131.6,124.5,113.4,62.2,53.2,43.5,14.0;
HRMS(ESI)m/z calcd.for C14H15N2O5[M+H]+:291.0975,found 291.0969.
Example 9
Raw materials:
Figure GDA0002407513420000101
and (3) target products:
Figure GDA0002407513420000102
1-phenyl-2-oxo-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 85 percent
1H NMR(400MHz,CDCl3):δ=7.98(dd,J1=8.0Hz,J2=1.6Hz,1H),7.63–7.56(m,3H),7.47–7.43(m,1H),7.38–7.34(m,1H),7.30–7.28(m,2H),6.73(dd,J1=8.4Hz,J2=1.2Hz,1H),4.02(s,3H);
13C NMR(100MHz,CDCl3):δ=163.9,152.2,149.2,135.0,134.9,132.3,131.7,130.8,130.3,129.7,128.1,124.4,115.7,53.2;
HRMS(ESI)m/z calcd.for C16H13N2O3[M+H]+:281.0921,found 281.0922.
Example 10
Raw materials:
Figure GDA0002407513420000103
and (3) target products:
Figure GDA0002407513420000104
1-methyl-2-oxo-6-fluoro-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 76%
1H NMR(400MHz,CDCl3):δ=7.65(dd,J1=8.0Hz,J2=3.2Hz,1H),7.46–7.41(m,1H),7.36–7.32(m,1H),4.03(s,3H),3.73(s,3H);
13C NMR(100MHz,CDCl3):δ=163.8,158.8(d,J=244.3Hz,1C),152.2,149.9,132.3,130.9(d,J=2.1Hz,1C),120.6(d,J=24.1Hz,1C),116.4(d,J=21.8Hz,1C),115.1(d,J=8.0Hz,1C),53.3,29.5;
19F NMR(376MHz,CDCl3):δ=-117.5;
HRMS(ESI)m/z calcd.for C11H10FN2O3[M+H]+:237.0670,found 237.0668.
Example 11
Raw materials:
Figure GDA0002407513420000111
and (3) target products:
Figure GDA0002407513420000112
1-methyl-2-oxo-6-chloro-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 92 percent
1H NMR(400MHz,CDCl3):δ=7.91(d,J=2.8Hz,1H),7.61(dd,J1=8.8Hz,J2=2.4Hz,1H),7.30(d,J=9.2Hz,1H),4.02(s,3H),3.70(s,3H);
13C NMR(100MHz,CDCl3):δ=163.6,152.1,149.7,132.8,132.6,132.2,130.3,129.6,115.1,53.2,29.4;
HRMS(ESI)m/z calcd.for C11H10ClN2O3[M+H]+:253.0374,found 253.0367.
Example 12
Raw materials:
Figure GDA0002407513420000113
and (3) target products:
Figure GDA0002407513420000114
1-methyl-2-oxo-6-bromo-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 90 percent
1H NMR(400MHz,CDCl3):δ=8.08(d,J=2.0Hz,1H),7.74(dd,J1=8.8Hz,J2=2.4Hz,1H),7.24(d,J=8.8Hz,1H),4.02(s,3H),3.70(s,3H);
13C NMR(100MHz,CDCl3):δ=163.6,152.1,149.6,135.3,133.5,133.2,132.5,116.8,115.3,53.3,29.4;
HRMS(ESI)m/z calcd.for C11H10BrN2O3[M+H]+:296.9869,found 296.9867.
Example 13
Raw materials:
Figure GDA0002407513420000115
target product
Figure GDA0002407513420000116
1-methyl-2-oxo-1, 2-dihydroquinoxaline-3, 6-dicarboxylic acid ester; the yield is 80 percent
NMR(400MHz,CDCl3):δ=8.63(d,J=1.6Hz,1H),8.31(dd,J1=8.4Hz,J2=1.6Hz,1H),7.41(d,J=8.8Hz,1H),4.04(s,3H),3.97(s,3H),3.76(s,3H);
13C NMR(100MHz,CDCl3):δ=165.5,163.6,152.4,149.3,137.2,133.2,133.1,131.2,126.3,114.0,53.3,52.5,29.5;
HRMS(ESI)m/z calcd.for C13H13N2O5[M+H]+:277.0819,found 277.0816.
Example 14
Raw materials:
Figure GDA0002407513420000121
target product
Figure GDA0002407513420000122
1-methyl-2-oxo-6-benzoyl-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 73 percent
1H NMR(400MHz,CDCl3):δ=8.35(d,J=2.0Hz,1H),8.26(dd,J1=8.8Hz,J2=2.0Hz,1H),7.82–7.79(m,2H),7.65–7.61(m,1H),7.53–7.49(m,3H),4.03(s,3H),3.80(s,3H);
13C NMR(100MHz,CDCl3):δ=194.4,163.5,152.4,149.3,137.0,133.9,133.6,133.3,132.8,130.7,129.8,129.6,128.6,114.4,53.3,29.6;
HRMS(ESI)m/z calcd.for C18H15N2O4[M+H]+:323.1026,found 323.1020.
Example 15
Raw materials:
Figure GDA0002407513420000123
target product
Figure GDA0002407513420000124
1-methyl-2-oxo-6-nitro-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 73 percent
1H NMR(400MHz,CDCl3)δ8.04(d,J=8.4Hz,1H),6.84(d,J=8.4Hz,1H),6.81(d,J=8.4Hz,1H),6.66(d,J=2.4Hz,1H),6.60(dd,J=2.4,8.4Hz,1H),6.01(s,2H).
13C NMR(100MHz,CDCl3)δ167.6,148.2,146.0,144.1,143.9,119.3,113.5,108.1,105.0,103.1,101.9.
HRMS Calcd(EI)m/z for C11H7NO4Se:[M]+296.9540,found:296.9534.
Example 16
Raw materials:
Figure GDA0002407513420000125
target product
Figure GDA0002407513420000126
1-methyl-2-oxo-7-trifluoromethyl-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 77 percent
1H NMR(400MHz,CDCl3):δ=8.06(d,J=8.4Hz,1H),7.64–7.60(m,2H),4.03(s,3H),3.76(s,3H);
13C NMR(100MHz,CDCl3):δ=163.5,152.1,150.9,134.1,134.0,133.7,132.0,123.2(q,J=271.2Hz,1C),120.6(q,J=3.7Hz,1C),111.4(q,J=4.3Hz,1C),53.4,29.4;19FNMR(376MHz,CDCl3):δ=-62.7;
HRMS(ESI)m/z calcd.for C12H10F3N2O3[M+H]+:287.0638,found 287.0644.
Example 17
Raw materials:
Figure GDA0002407513420000131
target product
Figure GDA0002407513420000132
1,6, 7-trimethyl-2-oxo-6-chloro-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 85 percent
1H NMR(400MHz,CDCl3):δ=7.67(s,1H),7.09(s,1H),4.00(s,3H),3.68(s,3H),2.43(s,3H),2.33(s,3H);
13C NMR(100MHz,CDCl3):δ=164.2,152.6,146.7,143.3,133.4,132.3,131.1,130.3,114.3,53.0,29.1,20.8,19.1;
HRMS(ESI)m/z calcd.for C13H15N2O3[M+H]+:247.1077,found 247.1076.
Example 18
Raw materials:
Figure GDA0002407513420000133
target product
Figure GDA0002407513420000134
1-methyl-2-oxo-6, 7-dichloro-1, 2-dihydroquinoxaline-3-carboxylic acid ester; the yield is 78 percent
1H NMR(400MHz,CDCl3):δ=8.00(s,1H),7.45(s,1H),4.01(s,3H),3.68(s,3H);
13C NMR(100MHz,CDCl3):δ=163.4,151.3,137.1,133.4,131.8,131.2,130.7,128.3,115.5,53.3,29.5;
HRMS(ESI)m/z calcd.for C11H9Cl2N2O3[M+H]+:286.9985,found 286.9976。

Claims (4)

1. A method for synthesizing a quinoxaline-2 (1H) -ketone C-3 site carboxylate compound is characterized by comprising the following steps: under the air atmosphere, quinoxaline-2 (1H) -ketone derivative, hydrazino carboxylic ester and persulfate react in one pot to generate quinoxaline-2 (1H) -ketone C-3 site carboxylic ester compound; the persulfate is potassium persulfate; the reaction adopts acetonitrile as a reaction medium;
the quinoxaline-2 (1H) -one derivative has a structure shown in a formula 1;
Figure FDA0002407141510000011
the hydrazinocarboxylic acid ester has the structure of formula 2:
Figure FDA0002407141510000012
the quinoxaline-2 (1H) -one C-3 carboxylate compound has the structure of formula 3:
Figure FDA0002407141510000013
wherein the content of the first and second substances,
R1selected from H, C1~C10Alkyl of (C)2~C10Alkenyl group of (C)2~C10Alkynyl, benzyl, C2~C10Ester group of (a) or phenyl group;
R2is selected from C1~C10Alkyl, halogen, trifluoromethyl, C2~C10Acyl group of (1), C2~C10Ester group or nitro group of (1);
R3is selected from C1~C10Alkyl group of (1).
2. The method for synthesizing a quinoxaline-2 (1H) -one C-3 carboxylate compound according to claim 1, wherein: the molar ratio of the quinoxaline-2 (1H) -one derivative, the hydrazino carboxylic acid ester and the persulfate is 1: 1-2: 2 to 4.
3. The method for synthesizing a C-3 carboxylate compound of quinoxaline-2 (1H) -ketone according to any one of claims 1 to 2, wherein: the reaction conditions are as follows: reacting for 5-15 hours at 80-100 ℃.
4. The method for synthesizing a quinoxaline-2 (1H) -one C-3 carboxylate compound according to claim 1, wherein: the molar concentration of the quinoxaline-2 (1H) -ketone derivative in an acetonitrile reaction medium is 0.1-0.4 mmol/mL.
CN201910083408.2A 2019-01-29 2019-01-29 Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound Expired - Fee Related CN109535086B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910083408.2A CN109535086B (en) 2019-01-29 2019-01-29 Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910083408.2A CN109535086B (en) 2019-01-29 2019-01-29 Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound

Publications (2)

Publication Number Publication Date
CN109535086A CN109535086A (en) 2019-03-29
CN109535086B true CN109535086B (en) 2020-05-05

Family

ID=65838522

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910083408.2A Expired - Fee Related CN109535086B (en) 2019-01-29 2019-01-29 Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound

Country Status (1)

Country Link
CN (1) CN109535086B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110483421B (en) * 2019-09-29 2022-06-10 沈阳师范大学 Preparation method of quinoxalinone derivative
CN114230528B (en) * 2021-11-25 2024-02-06 桂林理工大学 Method for preparing quinoxalinone derivative

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101016268B (en) * 2007-01-24 2010-08-11 华中农业大学 Chemical synthesis method for 2-formylcyanoacetylhydrazone-quinoxaline-1,4-dioxide
CN101704792B (en) * 2009-11-02 2012-10-03 山东大学 Quinoxalinone derivative with matrix metalloproteinase inhibitory activity and preparation method and application thereof
CN102250022B (en) * 2011-05-26 2013-10-16 浙江大学 Substituted quinoxalinamine compounds, and preparation method and application thereof
CN106243047B (en) * 2016-07-27 2019-07-23 江苏神华药业有限公司 The double aryl ureas of quinokysalines with VEGFR-2 and B-raf double inhibition effect and its derivative, preparation method and applications

Also Published As

Publication number Publication date
CN109535086A (en) 2019-03-29

Similar Documents

Publication Publication Date Title
CN110204486B (en) Synthesis method of quinoline derivative
CN107973779B (en) Preparation method of N- (2-pyridine/pyrimidine-based) indole derivative
CN109535086B (en) Synthetic method of quinoxaline-2 (1H) -ketone C-3 carboxylate compound
CN111675662A (en) Preparation method of 2-trifluoromethyl substituted quinazolinone compound
CN110642798B (en) Green synthesis method of N-substituted-1, 4-dihydro-2, 3-quinoxalinedione compound
CN109651271B (en) Synthetic method of 3-tert-butyl-N-methylquinoxaline-2 (1H) -ketone compound
CN111138345B (en) Method for synthesizing polysubstituted pyridine derivative based on oxime ester and unsaturated ketone under catalysis of iron salt
CN110105355B (en) Preparation method of 1,2, 3-triazole- [1,5-a ] quinoline compound
CN109535087B (en) Synthetic method of quinoxaline-2 (1H) -ketone C-3 site aroyl compound
CN112047879B (en) Method for selectively synthesizing halogenated arylamine by copper catalysis
CN114014805B (en) Preparation method of trifluoromethyl 2, 4-quinoline diketone compound
CN109651367A (en) A method of preparing 1,4- dihydroquinoline and pyrrolo- [1,2-a] quinolines
CN112174880B (en) Preparation method of 1,3,4, 6-tetra-substituted pyridone derivative
CN109535140A (en) A method of double indoles substituted-dihydro pyrrolones derivatives are constructed based on oxime ester and indoles
CN109988113B (en) Synthesis method of [60] fullerene tetrahydroquinoline derivative
KR100574350B1 (en) Process for preparation of 2-aminopyridine derivatives
CN102718694B (en) 3-cyan substituted indole compound and synthetic method thereof
CN105745210B (en) Method for preparing the ketone derivatives of 8 (4 amino-benzene oxygen) 4H pyridos [2,3 B] pyrazine 3
CN104193667A (en) Synthesis method of divergently oriented azacycles
CN111808072B (en) Synthetic method of 3-formyl indole derivative
CN114349684B (en) Synthetic method of benzo [ c, d ] indole imine derivative
CN114195726B (en) Preparation method of 1,2, 4-triazole substituted arylamine compound
Hajbi et al. General Synthetic Approach to 4-Substituted 2, 3-Dihydrofuro [2, 3-b] pyridines and 5-Substituted 3, 4-Dihydro-2H-pyrano [2, 3-b] pyridines
TW201835088A (en) Method for producing lamellarin and derivatives thereof relating to a method for synthesis of lamellarin capable of greatly shortening the synthesis path
US5344938A (en) Indole 3-carboxylic acid derivatives

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
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20200505

CF01 Termination of patent right due to non-payment of annual fee