Naphthalenesulfonyl compounds, preparation method and application thereof

文档序号:220626 发布日期:2021-11-09 浏览:30次 中文

阅读说明:本技术 萘磺酰类化合物、其制备方法和应用 (Naphthalenesulfonyl compounds, preparation method and application thereof ) 是由 唐惠儒 李鹏程 于 2021-08-11 设计创作,主要内容包括:本发明公开了萘磺酰类化合物、其制备方法和应用。具体公开了一种如式(I)所示的化合物或其盐,其作为一类可与羟基和氨基反应的特异性衍生化试剂,合成简单、反应活性高、廉价易得,可以改善目标化合物的色谱分离行为,并可以增强这些化合物的检测灵敏度。(The invention discloses a naphthalene sulfonyl compound, a preparation method and application thereof. Specifically discloses a compound shown as a formula (I) or a salt thereof, which is used as a specific derivatization reagent capable of reacting with hydroxyl and amino, has simple synthesis, high reaction activity, low price and easy obtaining, can improve the chromatographic separation behavior of a target compound, and can enhance the detection sensitivity of the compoundsAnd (4) degree.)

1. A compound of formula (I) or a salt thereof:

wherein R is1And R1' independently selected from C1-7An alkyl group;

R2is selected from H, C1-7Alkyl or benzyl;

x is selected from OH or halogen.

2. A compound of formula (I) or a salt thereof according to claim 1,

R1and R1' same;

and/or, R1And R1' independently is C1-7An alkyl group;

and/or, R2Is C1-7An alkyl group;

and/or, R1Or R1' of the above formula C1-7Alkyl is C1-4Alkyl radicals, e.g. C2-4Alkyl, such as ethyl;

and/or, R2In (A), the C1-7Alkyl is C1-4Alkyl groups such as isobutyl;

and/or, in X, the halogen is Cl.

3. The compound of formula (I) or a salt thereof according to claim 1, wherein the compound of formula (I) is

4. The compound of formula (I) or a salt thereof as claimed in claim 1, wherein the salt of the compound of formula (I) is a salt prepared from the compound of formula (I) and an acid, wherein the acid is an inorganic acid or an organic acid, preferably an organic acid.

5. A preparation method of a compound shown as a formula (I) is characterized by comprising the following one or two methods:

the first method comprises the following steps: in a solvent, in the presence of an activating agent and alkali, carrying out condensation reaction on a compound shown as a formula (III) and a compound shown as a formula (IV) to obtain a compound shown as a formula (I),

in the first method, X is OH, R1、R1' and R2As defined in any one of claims 1 to 3;

the second method comprises the following steps:

(1) in a solvent, in the presence of an activating agent and alkali, carrying out condensation reaction on a compound shown as a formula (III) and a compound shown as a formula (IV) to obtain a compound shown as a formula (V),

(2) in a solvent, carrying out acyl chlorination reaction on a compound shown as a formula (V) and a chlorinating agent to obtain a compound shown as a formula (I),

in the second method, X is Cl and R1、R1' and R2Is as defined in any one of claims 1 to 3.

6. A process for the preparation of a compound of formula (I) according to claim 5,

in the condensation reaction, the solvent is N, N-dimethylformamide;

and/or, in the condensation reaction, the activating agent is one or more of 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride and 1-hydroxybenzotriazole, such as 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride or "a combination of 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride and 1-hydroxybenzotriazole";

and/or in the condensation reaction, the base is an organic base, and is further preferably N-methylmorpholine and/or pyridine;

and/or, in the acyl chlorination reaction, the solvent is tetrahydrofuran and/or toluene;

and/or, in the acyl chlorination reaction, the chlorinating agent is phosphorus pentachloride and/or oxalyl chloride.

7. The process for producing a compound represented by the formula (I) according to claim 5, which further comprises the following process (1-1) or process (1-2):

the method (1-1) comprises the steps of: in a solvent, in the presence of a reducing agent, carrying out reductive amination reaction on a compound shown as a formula (VI), a compound shown as a formula (A-1) and a compound shown as a formula (A-2) to obtain a compound shown as a formula (III);

the method (1-2) comprises the steps of: in a solvent, in the presence of alkali, carrying out alkylation reaction on a compound shown as a formula (VI), a compound shown as a formula (B-1) and a compound shown as a formula (B-2) to obtain a compound shown as a formula (III);

X1and X2Independently halogen (e.g., I);

in the methods (1-1) and (1-2), R1、R1' and R2Is as defined in any one of claims 1 to 3.

8. A process for the preparation of a compound of formula (I) according to claim 7,

in the reductive amination reaction, the solvent is methanol, acetonitrile or sodium acetate or phosphate buffer solution with the pH value of 2-12;

and/or, in the reductive amination reaction, the reducing agent is sodium cyanoborohydride and/or 2-methylpyridine borane;

and/or, in the alkylation reaction, the solvent is acetonitrile;

and/or, in the alkylation reaction, the base is a carbonate or bicarbonate, preferably a carbonate, more preferably potassium carbonate.

9. An isotopically-labeled compound represented by the formula (II) or a salt thereof,

y is

Wherein R is1、R1’、R2And X is as defined in any one of claims 1 to 3,

at least one atom of Y is substituted with its heavier isotope.

10. The isotopically labeled compound of formula (II) or a salt thereof of claim 9,

at least one of Y1H by its heavier isotope2H is substituted;

and/or at least one of Y12C by its heavier isotope13C is substituted;

and/or at least one of Y14N by its heavier isotopes15N is substituted;

and/or at least one of Y16O by its heavier isotope18And (4) O substitution.

11. The isotopically-labeled compound of formula (II) of claim 10, which is any one of:

R0is composed ofX is OH or Cl.

12. Use of a compound of formula (I) or a salt thereof according to any one of claims 1 to 4 or an isotopically labeled compound of formula (II) or a salt thereof according to any one of claims 9 to 11 as a derivatizing agent for detecting and/or separating a compound containing a hydroxyl group and/or an amino group, wherein the compound containing a hydroxyl group and/or an amino group can also be a nucleoside metabolite.

Technical Field

The invention relates to a naphthalene sulfonyl compound, a preparation method and application thereof.

Background

Chemical labeling, i.e., isotope labeled derivatization (ICD), is a technique that introduces mass difference labels in the form of light-label and heavy-label isotopes into a target for relative quantitative analysis. The labeling technique is suitable for the quantitative analysis of target components in complex matrix samples, and when the concentration of one group of samples is known, the method can be used for absolute quantification of analytes in the samples.

The chemical labeling technology is applied to the quantitative analysis of proteome in the early stage, and along with the development of metabonomics, the stable isotope labeling technology is also gradually applied to the high-sensitivity detection of important small molecule metabolites such as amines, aldehydes and ketones, carboxylic acid metabolites and the like.

The selection of a reasonable derivatizing reagent needs to meet the following requirements: (1) the derivatization reagent is easy to synthesize, and isotope labeling in the derivatization reagent can be realized at lower cost; (2) specific derivatization labeling can be realized for target functional groups, and the reaction efficiency is stable; (3) the derivatization reaction condition is mild, and the existence form of endogenous target compounds in a system is not damaged; (4) the derivatization product can be effectively ionized to realize MS detection; (5) the isotope effect is small, and the retention time drift phenomenon basically does not exist.

In 1999, Gygi et al developed the mass difference tag-isotope affinity tag (ICAT) technology, and this reagent consisted of three major parts: an affinity tag consisting of biotin, a linking group for introducing a stable isotope, and a reactive group for specifically binding to a thiol group of a cysteine residue in the peptide fragment. In 2005, Che et al designed 4-Trimethylbutanamide (TMAB), a labeling reagent for all amino group-containing substances, and labeled with a D-labeled reagent and an H-labeled reagent, respectively, to perform quantitative analysis. The plurality of deuterated labeling sites on the TMAB and ICAT reagents cause serious isotope effect, and the retention time of the labeled substance to be detected on the chromatographic column is influenced.

Thompson equals 2003 the synthesis of equal quality tags (isobaric tags) TMT tags (tandem mass tags). The TMT is composed of a mass reporting area, a cleavable connecting area, a mass balancing area and an amino reactive group. The unique structure of the TMT reagent enables different isotopically labeled forms of the target molecule to have the same chromatographic behavior and primary MS characteristics. Through secondary mass spectrum scanning, the amino compounds in different labeling forms are fragmented in a cleavable region,different reporter ions are formed, and the relative content change of the sample can be determined by comparing the intensities of the reporter ions. TMT reagent mainly adopts13C, labeling, complex synthesis, high price and low yield, so that the use of the reagent is greatly limited.

In 2004, Applied Biosystems proposed an equal mass labeling technique (iTRAQ) labeling technique that is the same as the TMT labeling strategy. By changing the quantity and the types of isotopes of the balance report groups and the balance groups, which are designed into 4 labeling modes with the same molecular weight but different report groups, 4 groups of biological samples can be labeled by isotopes at the same time, and the target substances in multiple samples can be quantitatively analyzed by utilizing the report ion response of the report groups in MS/MS. The iTRAQ technology has been developed to eight-fold labeled reagents, but is expensive and susceptible to interference from amino-bearing species in the sample.

Based on a stable isotope labeling technology of chemical derivatization, a functional group with isotope mass difference can be labeled on different biological samples, so that light mark/heavy mark isotope labeling reflecting sample information is obtained, and then the mass spectrum response difference of target components labeled by the light mark and the heavy mark isotope is compared by utilizing a liquid chromatography-mass spectrum coupling technology to obtain quantitative information of different metabolites. The technology is widely applied to common metabolites such as ammonia, hydroxyl, phenolic hydroxyl, carboxylic acids and aldehydes and ketones, and provides a novel idea and strategy for derivatization-assisted mass spectrometry of nucleoside metabolites.

Disclosure of Invention

The invention aims to solve the technical problems of high price, serious isotope effect, poor detection sensitivity, complicated synthesis steps and the like of the existing specific derivatization reagent, and provides a naphthalene sulfonyl compound, a preparation method and application thereof.

The invention solves the technical problems through the following technical scheme.

The present invention provides a compound represented by the formula (I):

wherein R is1And R1' independently selected from C1-7An alkyl group;

R2is selected from H, C1-7Alkyl or benzyl;

x is selected from OH or halogen.

In a preferred embodiment of the present invention, some groups in the compound represented by the formula (I) or a salt thereof are defined as follows, and the groups which are not mentioned are the same as those described in any of the embodiments (abbreviated as "in one embodiment of the present invention"), R1Or R1' of the above formula C1-7Alkyl is C1-4Alkyl radicals, e.g. C2-4Alkyl, for example ethyl.

In one embodiment of the invention, R2In (A), the C1-7Alkyl is C1-4Alkyl groups, such as isobutyl.

In one embodiment of the present invention, in X, the halogen is Cl.

In one embodiment of the invention, R1And R1' same.

In one embodiment of the invention, R2Is C1-7An alkyl group.

In one embodiment of the present invention, the compound represented by formula (I) is

In a certain embodiment of the present invention, the salt of the compound represented by formula (I) is a salt prepared from the compound represented by formula (I) and an acid, and the acid is an inorganic acid or an organic acid, preferably an organic acid.

The invention also provides a preparation method of the compound shown in the formula (I), which comprises the following first method or second method:

the first method comprises the following steps: in a solvent, in the presence of an activating agent and alkali, carrying out condensation reaction on a compound shown as a formula (III) and a compound shown as a formula (IV) to obtain a compound shown as a formula (I),

in the first method, X is OH, R1、R1' and R2Is as defined in any of the preceding items;

the second method comprises the following steps:

(1) in a solvent, in the presence of an activating agent and alkali, carrying out condensation reaction on a compound shown as a formula (III) and a compound shown as a formula (IV) to obtain a compound shown as a formula (V),

(2) in a solvent, carrying out acyl chlorination reaction on a compound shown as a formula (V) and a chlorinating agent to obtain a compound shown as a formula (I),

in the second method, X is Cl and R1、R1' and R2Is as defined in any of the preceding claims.

In a certain embodiment of the preparation method of the present invention, the solvent in the condensation reaction may be a solvent conventional in such reactions in the art, and is preferably N, N-Dimethylformamide (DMF).

In a certain embodiment of the preparation process of the present invention, the activator in the condensation reaction may be an activator conventional in the art for such reactions, preferably one or more of 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride (DMTMM), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (EDC) and 1-hydroxybenzotriazole (HOBt), such as 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride or "combination of 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride and 1-hydroxybenzotriazole".

In a certain embodiment of the preparation method of the present invention, the base in the condensation reaction may be a base conventional in such reactions in the art, preferably an organic base, and more preferably N-methylmorpholine (NMM) and/or pyridine (Py).

In certain embodiments of the preparation methods of the present invention, the condensation reaction may be at a temperature conventional in the art for such reactions, such as room temperature.

In a certain embodiment of the preparation method of the present invention, the progress of the condensation reaction can be detected by a monitoring method (e.g., TLC, HPLC or NMR) which is conventional in the art, and a compound represented by the formula (III) is generally disappeared or no longer reacted as a reaction end point. The time for the condensation reaction may be 8 to 24 hours.

In a certain embodiment of the preparation method of the present invention, the solvent in the acid chlorination reaction may be a solvent conventional in such reactions in the art, and is preferably Tetrahydrofuran (THF) and/or toluene.

In a certain embodiment of the preparation method of the present invention, in the acyl chlorination reaction, the chlorinating agent may be a chlorinating agent conventional in the art for such reactions, preferably phosphorus pentachloride and/or oxalyl chloride.

In certain embodiments of the preparation methods of the present invention, the temperature of the acid chlorination reaction may be a temperature conventional in the art for such reactions, such as room temperature.

In one embodiment of the preparation method of the present invention, the progress of the acyl chlorination reaction can be monitored by a monitoring method (e.g., TLC, HPLC or NMR) conventionally used in the art, and the disappearance or no longer reaction of the compound represented by the formula (V) is generally used as the reaction end point. The time for the acyl chlorination reaction may be from 5 minutes to 4 hours.

The preparation method of the compound shown in the formula (I) can further comprise the following method (1-1) or method (1-2):

the method (1-1) comprises the steps of: in a solvent, in the presence of a reducing agent, carrying out reductive amination reaction on a compound shown as a formula (VI), a compound shown as a formula (A-1) and a compound shown as a formula (A-2) to obtain a compound shown as a formula (III);

the method (1-2) comprises the steps of: in a solvent, in the presence of alkali, carrying out alkylation reaction on a compound shown as a formula (VI), a compound shown as a formula (B-1) and a compound shown as a formula (B-2) to obtain a compound shown as a formula (III);

X1and X2Independently halogen (e.g., I);

in the methods (1-1) and (1-2), R1、R1' and R2Is as defined in any of the preceding claims.

In a certain variant of the preparation process according to the invention, the solvent in the reductive amination reaction may be a solvent customary in such reactions in the art, preferably methanol, acetonitrile or sodium acetate or phosphate buffer at a pH of 2 to 12.

In a certain variant of the preparation process according to the invention, the reducing agent in the reductive amination reaction may be a reducing agent customary in such reactions in the art, preferably sodium cyanoborohydride and/or 2-methylpyridine borane.

In a variant of the preparation process according to the invention, the temperature of the reductive amination may be a temperature customary in the art for such reactions, preferably from 30 to 40 ℃.

In one embodiment of the preparation process of the present invention, the progress of the reductive amination reaction can be monitored by monitoring methods conventional in the art (e.g., TLC, HPLC or NMR), and the disappearance or no longer reaction of the compound represented by formula (VI) is generally used as the reaction endpoint. The time for the reductive amination reaction may be 20 to 28 hours.

In a certain embodiment of the preparation process of the present invention, the solvent in the alkylation reaction may be a solvent conventional in such reactions in the art, preferably acetonitrile.

In a certain embodiment of the preparation process of the present invention, the base in the alkylation reaction may be a base conventional in such reactions in the art, preferably a carbonate or bicarbonate, preferably a carbonate, more preferably potassium carbonate.

In a variant of the preparation process according to the invention, the temperature of the alkylation may be that customary in the art for such reactions, preferably from 70 to 90 ℃.

In one embodiment of the preparation method of the present invention, the progress of the alkylation reaction can be monitored by a monitoring method (e.g., TLC, HPLC or NMR) conventionally used in the art, and the disappearance or no longer reaction of the compound represented by the formula (VI) is generally used as the reaction end point. The time for the alkylation reaction may be 20 to 28 hours.

The invention also provides an isotope labeling compound shown as a formula (II) or a salt thereof,

y is

Wherein R is1、R1’、R2And X is as defined in any of the preceding items,

at least one atom of Y is substituted with its heavier isotope.

In one embodiment of the present invention, at least one of Y1H by its heavier isotope2And H is substituted.

In one embodiment of the present invention, at least one of Y12C by its heavier isotope13C is substituted.

In one embodiment of the present invention, at least one of Y14N by its heavier isotopes15And (4) N substitution.

In one embodiment of the present invention, at least one of Y16O by its heavier isotope18And (4) O substitution.

In a certain embodiment of the present invention, the isotopically labeled compound represented by formula (II) is any one of the following compounds:

R0is composed ofX is OH or Cl.

The isotopically labeled compound of formula (II) or a salt thereof can be prepared by methods conventional in the art, for example2H、13C、15The N-labeled isotope-labeled compound is prepared by the current commercialized isotope-labeled acetaldehyde and isotope-labeled sodium cyanoborohydride,18the O-labeled isotopically-labeled compound is obtained by16O-18Obtained by oxygen exchange reaction of O.

The invention also provides application of the compound shown in the formula (I) or the salt thereof or the isotopic labeled compound shown in the formula (II) or the salt thereof as a derivatization reagent, wherein the derivatization reagent is used for detecting and/or separating compounds containing hydroxyl and/or amino, and the compounds containing hydroxyl and/or amino can be nucleoside metabolites.

Unless otherwise defined, the terms used in the present invention have the following meanings:

it will be appreciated by those skilled in the art that, in accordance with common practice used in the art, the present invention describes the structural formulae used in the structural formulae of the radicalsMeans that the corresponding group is linked to other fragments, groups in the compound through this site.

The term "halogen" refers to fluorine, chlorine, bromine or iodine.

The term "alkyl" refers to a straight or branched chain alkyl group having the indicated number of carbon atoms. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, and the like.

The above preferred conditions can be arbitrarily combined to obtain preferred embodiments of the present invention without departing from the common general knowledge in the art.

The reagents and starting materials used in the present invention are commercially available.

The positive progress effects of the invention are as follows: the invention provides several types of compounds with N, N-dialkyl amino acetyl amino naphthalene sulfonic acid and sulfonylation substance structures and a synthesis method thereof, wherein the compounds are used as specific derivatization reagents capable of reacting with hydroxyl and amino, have high reaction activity, are cheap and easy to obtain, can improve the chromatographic separation behavior of target compounds, and can enhance the detection sensitivity of the compounds.

Detailed Description

The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.

In the following examples, sources of experimental reagents are shown in table 1 below:

TABLE 1 Experimental reagents and sources

The qualitative analysis of the various starting materials and products was carried out by an AB Sciex ExionLC UHPLC system comprising modules of PDA detector, autosampler, binary gradient pump, temperature control unit, equipped with an ACQUITY UPLC HSS T3C 18 reversed phase chromatography column (1.8 μm, 2.1 mm. times.100 mm). Experiments such as molecular weight and mass spectrometry fragmentation of each raw material and product were performed on AB Sciex X500R TOF. The fine purification of the N, N-diethyl leucyl amino naphthalene sulfonic acid is realized by an Agilent 1100series LC system which comprises a VWD detector, an autosampler, a binary gradient pump, a temperature control unit and the like, and a chromatographic column is provided with a YMC Pack ODS-A C18 chromatographic column (5 mu m, 10mm multiplied by 25 mm). Structure and purity information for the starting materials and products in each synthetic step is provided by Bruker Ascend 600MHz NMR. Anke N-1001D-OSB2100 rotary evaporator for removing organic solvent, Christ ALPHA 1-2LD plus freeze dryer for removing water, glass instrument such as chromatographic column (Beijing Xin Weier instruments Co.) for completing each step of synthesis reaction.

Example 1 Synthesis of N, N-diethyl-L-leucine

L-leucine powder (800mg, 6mmol) is weighed into a 100mL round-bottom flask, 40mL sodium acetate or phosphate buffer (0.2M, pH 2-12) is added and dissolved under stirring at 37 ℃, sodium cyanoborohydride powder (1.6g, 24mmol) is added, then acetaldehyde solution (3.4mL, 60mmol) is added dropwise, the reaction mixture is stirred at 30-40 ℃ for 20-28 hours, and finally 6mol/L HCl solution (4mL, 24mmol) is added and stirred for 10min to terminate the reaction. Removing the organic reagent by using a rotary evaporator, freeze-drying the reactant by using a freeze dryer, and purifying by adopting a reverse phase column chromatography mode to obtain the pure N, N-diethylleucine.

The structure of the pure N, N-diethyl L-leucine was confirmed by NMR and mass spectrometry.

1H NMR(600MHz,D2O buffer, ph 7.4): δ 0.971(dd, 6H), 1.298(t, 6H), 1.650(m, 2H), 1.760(m, 1H), 3.247(m, 4H), 3.668(dd, 1H); MS + (TOF) m/z 188.1645.

Example 2 Synthesis of N, N-diethyl-L-leucine

L-leucine powder (800mg, 6mmol) is weighed into a 100mL round-bottom flask, 40mL sodium acetate or phosphate buffer (0.2M, pH 2-12) is added and dissolved with stirring at 37 ℃, 2-methylpyridine borane (1.3g, 12mmol) is added, then an acetaldehyde solution (3.4mL, 60mmol) is added dropwise, the reaction mixture is stirred at 30-40 ℃ for 20-28 hours, and finally 6mol/L HCl solution (4mL, 24mmol) is added and stirred for 10min to terminate the reaction. Removing the organic reagent by using a rotary evaporator, freeze-drying the reactant by using a freeze dryer, and purifying by adopting a reverse phase column chromatography mode to obtain the pure N, N-diethylleucine.

Example 3 Synthesis of N, N-diethyl L-leucine

Leucine powder (800mg, 6mmol) is weighed into a 100mL round-bottom flask, ground potassium carbonate powder (4.8g, 6mmol) and 40mL acetonitrile are added, and an iodoethane solution (9.6mL, 120mmol) is added dropwise with stirring to react for 20-28 hours at 90 ℃ under reflux. Excess potassium carbonate was removed by filtration and the solvent was removed by rotary evaporation. Adding ether into the crude product for filtration, washing the precipitate with ether for multiple times, and finally redissolving with acetonitrile for recrystallization to obtain a purified product.

Example 4 Synthesis of N, N-diethylleucylaminonaphthalenesulfonic acid

N, N-diethylleucine (800. mu.L, 16mmol) was dissolved in DMF, DMTMM (6.9mg, 24mmol) was added dropwise, NMM (43.3. mu.L, 320mmol) was added thereto by vortexing, 5-aminonaphthalenesulfonic acid powder (71.6mg, 640mmol) was added thereto by vortexing, and the mixture was reacted at room temperature for 8 to 24 hours in a metal shaker without vortexing to obtain 12 groups. The product was purified by extraction, 192mL of methylene chloride and 19.2mL of double distilled water were added to extract impurities, and the supernatant was obtained.

EXAMPLE 5 Synthesis of N, N-diethylleucylaminonaphthalenesulfonic acid

N, N-diethylleucine (35.7mg, 192mmol) was dissolved in DMF and activated with 1.2-fold equivalent of EDC (44.2mg, 230mmol) and HOBt (31.1mg, 230 mmol). 1.5 times equivalent of 5-aminonaphthalenesulfonic acid (64.4mg, 287.5mmol) was added thereto, and 2mL of pyridine was added dropwise thereto, and reacted overnight at ordinary temperature with stirring.

The organic reagent was removed using a rotary evaporator and the crude product was purified by reverse phase column chromatography with the packing being ODS C18 to yield about 26mg of a yellow-brown powder. And (3) performing fine purification and rotary evaporation by using a semi-preparative liquid chromatography Agilent 1100LC-VWD combined instrument to remove the solvent to obtain a pure product.

The structure was confirmed using NMR and mass spectrometry.

1H NMR(600MHz,MeOD):δ1.232、1.070(dd,6H),1.435(t,6H),1.832(m,2H),2.050(m,1H),3.411、3.502(q,4H),4.359(dd,1H),7.213(m,1H),7.402(m,1H),7.457(m,1H),7.692(m,1H),8.037(m,1H),8.722(m,1H);MS+(TOF)m/z 393.1845。

Example 6 Synthesis of N, N-diethylleucylaminonaphthalenesulfonyl chloride

Weighing N, N-diethyl leucyl amino naphthalene sulfonic acid (26.1mg, 0.07mmol) to be dissolved in 5mL of solvent toluene, carrying out ultrasonic dissolution promotion, weighing excess phosphorus pentachloride (0.7g, 3.33mmol) to be added into a reaction bottle according to the reaction molar ratio of 1:50, and carrying out reaction for 1-3 hours at normal temperature. Adding ethyl acetate for extraction, gradually dropwise adding ice saturated sodium bicarbonate solution for quenching reaction, adjusting the pH to 7, taking supernatant, and evaporating to dryness to obtain a crude product of the N, N-diethyl leucyl amino naphthalene sulfonyl chloride.

And (3) purifying the crude product by using normal phase column chromatography with silica gel as a filler, using petroleum ether, ethyl acetate and acetonitrile as eluent, combining elution components of 1:1 (acetonitrile/ethyl acetate) and 1:2 (acetonitrile/ethyl acetate), and performing rotary evaporation to evaporate the solvent to obtain a pure product of the N, N-diethyl leucyl amino naphthalene sulfonyl chloride.

The structure was confirmed using NMR.

1H NMR(600MHz,CD3CN):δ1.203(dd,6H),1.558(t,6H),1.958(m,2H),7.892(m,1H),8.026(m,1H),8.135(m,1H),8.593(m,1H),8.816(m,1H),8.964(m,1H);MS+(TOF)m/z 411.1495。

Example 7 Synthesis of N, N-diethylleucylaminonaphthalenesulfonyl chloride (DELANS-Cl)

N, N-diethylleucylaminonaphthalenesulfonic acid (26.1mg, 0.067mmol) was weighed and dissolved in 4mL of THF, 20 equivalents of oxalyl chloride (112. mu.L, 1.33mmol) was diluted to 500. mu.L of THF in an ice bath and added to a reaction flask, 3 drops of DMF were added dropwise and the reaction was stirred at room temperature for 10-30 minutes, and THF and oxalyl chloride were removed by rotary evaporation.

And (3) purifying the crude product by using normal phase column chromatography with silica gel as a filler, using petroleum ether, ethyl acetate and acetonitrile as eluent, combining elution components of 1:1 (acetonitrile/ethyl acetate) and 1:2 (acetonitrile/ethyl acetate), and performing rotary evaporation to evaporate the solvent to obtain a pure product of the N, N-diethyl leucyl amino naphthalene sulfonyl chloride.

Example 8d2Synthesis of (E) -N, N-diethyl L-leucine

d2-N, N-diethyl L-leucine:

d2the synthesis steps of the (E) -N, N-diethyl L-leucine are the same as those of the N, N-diethyl L-leucine, and the difference is that the (E) -N, N-diethyl L-leucine is synthesized by using deuterated sodium cyanoborohydride as a raw material. ([ M + H)]+=190.1771)

Example 9d2-N, N-diethylleucylaminonaphthalenesulfonic acid (d)2-DELANS-Cl)

d2-N, N-diethylleucylaminonaphthalenesulfonic acid:

d2the synthesis of (E) -N, N-diethylleucylaminonaphthalene sulfonic acid is carried out by the same steps as N, N-diethylleucylaminonaphthalene sulfonic acid except that d is2-N, N-diethyl L-leucine is synthesized by taking the raw material as a raw material. ([ M + H)]+=395.1968)

Example 10d2Synthesis of (E) -N, N-diethylleucyl aminonaphthalenesulfonyl chloride

d2-N, N-diethylleucylaminonaphthalenesulfonyl chloride:

d2the synthesis of (E) -N, N-diethylleucylaminonaphthalenesulfonyl chloride is carried out in the same manner as N, N-diethylleucylaminonaphthalenesulfonyl chloride except that d2The (E) -N, N-diethyl leucyl amino naphthalene sulfonic acid is synthesized by taking the (E) -N, N-diethyl leucyl amino naphthalene sulfonic acid as a raw material.

The structure was confirmed using NMR and mass spectrometry.

1H NMR(600MHz,CD3CN):δ1.203(dd,6H),1.518(d,3H),1.592(d,3H),1.959(m,2H),3.479(m,2H),4.388(m,1H),7.875(m,1H),8.038(m,1H),8.132(m,1H),8.584(m,1H),8.831(m,1H),8.959(m,1H);MS+(TOF)m/z 413.1602。

Effect example 1 derivatization reaction of derivatization reagent DELANS-Cl for nucleoside metabolites

N, N-Diethylleucylaminonaphthalenesulfonyl chloride (DELANS-Cl):d2-N, N-diethylleucylaminonaphthalenesulfonyl chloride (d)2-DELANS-Cl):

The above-mentioned derivatization reagent can be used for derivatization of compounds containing amino groups and hydroxyl groups like the classical dansyl chloride, and can also be used for derivatization of nucleoside compounds and is specifically as follows.

Preparing a working solution:

the nucleoside metabolite powders shown in table 2 were respectively and accurately weighed, dissolved in 250mM sodium carbonate/sodium bicarbonate buffer solution with pH 9.4 to obtain each metabolite standard stock solution with the concentration shown in table 2, and 50 μ L of each single standard was mixed to obtain a mixed stock solution of 65 nucleoside metabolites, which was the initial mixed stock solution. The initial mixed stock solution was diluted to give a working solution S having a total concentration of about 4mM1And then, according to the dilution ratio of 1: 2: 4: 10: 20: 40: 100: 200: 400: 1000: 2000: diluting 4000 stages to obtain S1、S2、S3、S4、S5、S6、S7、S8、S9、S10、S11、S12There were 12 concentration gradients of the working solution.

TABLE 2 Standard stock solution, initial Mixed stock solution, working solution S1And S1DerivatisationConcentration of each substance in the reaction solution

Preparing an internal standard solution:

isotopically labelled derivatizing reagents d2The solvent of-DELANS-Cl is dry acetonitrile solution, and d with the concentration of 5mmol/L is prepared2DELANS-Cl solution. Pipetting 50. mu.L of S using a pipette2The working solution was pipetted into a 500. mu.L EP tube at 400. mu.L d2To this was added a DELANS-Cl solution (5mM in acetonitrile), and the reaction EP tube was set on a metal shaker at 37 ℃ for 5 hours with shaking at 900 rpm. The reaction mixture solution was immediately transferred to ice to cool and quench the reaction. And (3) taking out 100 mu L of reaction solution after the derivatization reaction is finished, diluting by 5 times by using acetonitrile solution, uniformly mixing to obtain an internal standard solution, and storing at the low temperature of minus 20 ℃ or minus 80 ℃ in a sealed manner.

And (3) establishing a derivatization reaction and a linear curve of a derivatization reagent DELANS-Cl and a standard solution:

the solvent of the derivatization reagent DELANS-Cl is a dry acetonitrile solution, and a DELANS-Cl solution with the concentration of 5mmol/L is prepared. First, 5. mu.L of each sample solution (i.e., working solution of each concentration gradient) (dissolved in 250mM of pH 9.4 sodium bicarbonate buffer solution) was pipetted into a 500. mu.L EP tube, and then 40. mu.L of DELANS-Cl solution (5mM, acetonitrile solution) was added thereto by pipetting, and the reaction EP tube was placed on a metal shaker at a reaction temperature of 37 ℃ for 5 hours under an oscillation frequency of 900 rpm. The reaction mixture solution was then immediately transferred to ice to cool and quench the reaction. After the derivatization reaction, 8. mu.L of the reaction solution was taken out, diluted 5 times with acetonitrile solution, and 10. mu.L of the internal standard solution (volume ratio 4: 1) was added and mixed well. The treated samples were stored hermetically at-20 ℃ or-80 ℃ at low temperature before entering the UHPLC-MS system for analysis.

Derivatization reaction of derivatization reagent DELANS-Cl with the sample:

derivatization of nucleoside metabolites in the sample is as follows. mu.L of the sample solution (urine sample, serum sample, tissue sample and lung cancer cell sample, respectively) was taken out into a 1.5mL EP tube, 40. mu.L of DELANS-Cl solution (5mM in acetonitrile) was added thereto, and the reaction EP tube was set on a metal shaker at a reaction temperature of 37 ℃ for 5 hours at an oscillation frequency of 900 rpm. The reaction mixture solution was immediately transferred to ice to cool and quench the reaction. Finally, 8. mu.L of the reaction solution was taken out after the derivatization reaction was completed, diluted 5 times with acetonitrile solution, and 10. mu.L of the internal standard solution (volume ratio 4: 1) was added and mixed well. And preparing to enter a UHPLC-MS system for analysis.

Urine samples were collected from adult male morning urine. The serum sample is collected from healthy adults and meets the related requirements of scientific research ethics of the university of Compound denier and the national legal provisions. Tissue samples were taken from rabbit liver. Lung cancer cell samples: the cell sample selected in the experiment is a non-small cell lung adenocarcinoma cell line A549.

Derivatization of nucleoside metabolites with the derivatization reagents N, N-dimethylaminonaphthalenesulfonyl chloride (DNS-Cl) and N, N-diethylaminonaphthalenesulfonyl chloride (DENS-Cl) is referred to as DELANS-Cl.

N, N-dimethylaminonaphthalenesulfonyl chloride (DNS-Cl):n, N-diethylaminonaphthalenesulfonyl chloride (DENS-Cl):

the test method comprises the following steps:

the liquid phase equipped column was a Waters acquitty UPLC HSST 3C 18 reverse phase column (Waters Technologies, Milford, USA). The column temperature was 40 ℃ and the autosampler temperature was 4 ℃. Mobile phase a was 0.1% formic acid in water (MilliQ ultrapure water) and mobile phase B was 0.1% formic acid in acetonitrile. Elution gradient (B%) was as follows, 0-0.5 min: 2-25%, 0.5-3.6 min: 25%, 3.6-3.7 min: 25-30%, 3.7-4.5 min: 30%, 4.5-6 min: 30-40%, 6-7 min: 40-90%, 7-8 min: 95 percent. The flow rate was 0.5mL/min and the amount of sample was 1. mu.L.

Mass Spectrometry AB Sciex 6500plus QTRAP (ESI-MS/MS) using positive ion mode, ion source (chamber) conditions were as follows: the air pressure of an air curtain is 35psi, the air flow of the collision pool is selected to be medium, the voltage of the ion spraying is 4500V, the pressure of the spraying air is 55psi, the temperature of the spraying air is 400 ℃, and the pressure of the auxiliary heater is 50 psi. The scanning mode is an sMRM (scheduled multiple interaction monitoring) mode, the common daughter ion of the light standard derivatization product is m/z 142.2, the common daughter ion of the heavy standard derivatization product is m/z 144.2, and the Collision Energy (CE) of each derivatization product is respectively obtained by optimizing each derivatization standard product after derivatization.

Test results the linear range, correlation coefficient and minimum quantitative limit of 165 nucleoside metabolites

DELANS-Cl reacts with the working solution with each concentration gradient to obtain the linear range, the linear correlation coefficient and the lowest quantitative limit of 65 nucleoside metabolites.

TABLE 365 nucleoside metabolites Linear Range, Linear correlation coefficient and minimum quantitation Limit

Test results 2

The results of comparing the sensitivities of DELANS-Cl derivatization reactions with DNS-Cl and DENS-Cl are shown in Table 4:

TABLE 4DELANS-Cl vs DNS-Cl, DENS-Cl derivatization sensitivity comparison

Comparing the sensitivity of DELANS-Cl as a derivatizing reagent of the present invention with that of DNS-Cl or DNS-Cl as a commercially available derivatizing reagent, the sensitivity of over 58 metabolites in the nucleoside metabolite library (65) was improved. Compared with DNS-Cl, the derivatization reagent DELANS-Cl can improve the sensitivity by 541 times to the maximum extent; compared with DENS-Cl, DELANS-Cl of the derivatization reagent can improve the sensitivity by 225 times at most.

Test results 3

The derivatization reagent DELANS-Cl can be used for derivatizing metabolites containing amino and hydroxyl groups (namely nucleoside metabolites) in urine, serum, tissues and cell samples, and the derivatized samples can be quantitatively detected by using an ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS/MS) technology to respectively detect 58, 55, 59 and 60 nucleoside metabolites. The analysis results are shown in Table 5.

TABLE 5 detection results of nucleoside metabolites in urine, serum, tissue and cell samples

Remarking: "ND" means not detected or below the limit of quantitation.

Test results 4

TABLE 6S2Working solution and DELANS-Cl vs S2Retention time after derivatization of metabolites in working solution

By contrast, the retention time on a part of non-derivatized metabolite columns is poor (the retention time is close to the dead volume of a chromatographic column to 0.5min), and the metabolite which is poor in retention time on the columns after derivatization is improved, and the retention time is within 3-6 min.

40页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种硫酚与邻二碘苯的反应方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类