CN117777047A - N-substituted azaspiro-decone compounds, preparation method and application thereof - Google Patents

N-substituted azaspiro-decone compounds, preparation method and application thereof Download PDF

Info

Publication number
CN117777047A
CN117777047A CN202410208065.9A CN202410208065A CN117777047A CN 117777047 A CN117777047 A CN 117777047A CN 202410208065 A CN202410208065 A CN 202410208065A CN 117777047 A CN117777047 A CN 117777047A
Authority
CN
China
Prior art keywords
compound
substituted
azaspiro
intermediate compound
decarenone
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.)
Granted
Application number
CN202410208065.9A
Other languages
Chinese (zh)
Other versions
CN117777047B (en
Inventor
何菱
葛俊
许威
吴静涵
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.)
Sichuan University
Original Assignee
Sichuan 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 Sichuan University filed Critical Sichuan University
Priority to CN202410208065.9A priority Critical patent/CN117777047B/en
Publication of CN117777047A publication Critical patent/CN117777047A/en
Application granted granted Critical
Publication of CN117777047B publication Critical patent/CN117777047B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)
  • Indole Compounds (AREA)

Abstract

The invention provides an N-substituted aza-spiro-decone compound, a preparation method and application thereof, and belongs to the technical field of spiro-decone compounds. The N-substituted azaspiro-decone compound has a structure shown in a formula I or a formula II; the invention realizes the efficient construction of the N-substituted azaspiro-decarenone compounds through nucleophilic addition reaction. The preparation method is simple, and the prepared compound has stable structure, small toxic and side effects, good water solubility and anti-tumor activity and can be used for preparing anti-tumor drugs.

Description

N-substituted azaspiro-decone compounds, preparation method and application thereof
Technical Field
The invention belongs to the technical field of spirodecenone compounds, and particularly relates to an N-substituted aza-spirodecenone compound, a preparation method and application thereof.
Background
Malignant tumor is a serious disease which endangers human health and life, so the research and development of novel antitumor drugs with low toxicity and high curative effect based on different structure and effect relations and different action targets are significant. Spirodecenone is a derivative containing a fully substituted quaternary carbon center (spiroatom) and has various biological activities such as anti-tumor, anti-inflammatory, antibacterial, antiviral and anti-degenerative diseases. Moreover, spirodecenone has a unique rigid and conformational structure, which is of great interest due to its structural specificity and versatile bioactivity.
The prior researches show that the sulfonamide has good nitrene reactivity and stability, the sulfonamide compound can be selectively enriched in tumor tissues, has the characteristics of potential of tumor targeted drug delivery carriers and the like, and the sulfonamide is adopted as a nitrogen source to design and synthesize serial sulfonamide biazaspirodecadienone H1 and H2 derivatives which have certain antitumor activity, however, the sulfonamide biazaspirodecadienone H1 and H2 derivatives can show the characteristic of enrichment at tumor sites and better antitumor activity, but have the problems of stronger toxicity, poor water solubility, low in-vitro activity and the like, and the defects limit the further application research of the sulfonamide biazaspirodecadienone H1 and H2 derivatives. The structural formula of the sulfonamide biazaspirodecadienone H1 and H2 derivatives is shown as follows:
disclosure of Invention
The invention aims to solve the problems that: provides N-substituted aza-spiro-decylenone compounds, and a preparation method and application thereof, so as to solve the problems of strong toxicity, poor water solubility and low in-vitro activity of sulfonamide biazaspiro-decylenone derivatives.
The technical scheme adopted for solving the technical problems is as follows: provided are N-substituted azaspiro-decarenones having a structure as shown in formula I or formula II:
wherein:
r is H, C 1-20 Linear or branched alkyl, halogen, alicyclic, aromatic ring, carboxyl, hydroxyl, amino, mercapto, carbo-acylated derivative, phosphorylated derivative, phosphitylated derivative or thiolated derivative;
R 2 is an aromatic sulfonyl or substituted aromatic sulfonyl, aromatic carbonyl or substituted aromatic carbonyl, aromatic phosphoryl or substituted aromatic phosphorylAn aromatic phosphoryl group, an aromatic ring or a substituted aromatic ring, an alicyclic ring or a substituted alicyclic ring, a heterocyclic ring or a substituted heterocyclic ring, a benzyl group or a substituted benzyl group;
R 3 is C 1-20 Linear or branched alkyl, halogen, alicyclic, aromatic ring, heterocyclic, carboxyl, hydroxyl, amino, mercapto, carbo-acylated derivative, phosphorylated derivative, phosphitylated derivative or thiolated derivative;
x is oxygen, sulfur, nitrogen or carbon element, and n is 0, 1,2,3, 4 or 5.
Preferably, R is H, halogen or an aromatic ring; r is R 2 Is an aromatic sulfonyl or substituted aromatic sulfonyl, aromatic ring or substituted aromatic ring, heterocycle or substituted heterocycle, benzyl or substituted benzyl; r is R 3 Is a heterocycle, carboxyl, mercapto or carboacylated derivative; x is oxygen; n is 1.
Preferably, the specific structural formula of the N-substituted azaspiro-decarenone compound is as follows:
the preparation method of the N-substituted azaspiro-decone compound comprises the following synthetic routes:
synthesis path 1: dissolving a compound A and a compound B in an organic solvent, reacting under the action of a catalyst to obtain an intermediate compound 1, adding an oxidant to react to obtain an intermediate compound 2, and adding a nucleophile to react to obtain an N-substituted azaspiro-decamonoalkone compound shown in a formula I; the synthetic route is as follows:
synthesis path 2: dissolving a compound C and a compound D in an organic solvent, reacting under the action of a dehydrating agent to obtain an intermediate compound 3, adding an oxidant to react to obtain an intermediate compound 4, adding a compound B, reacting under the action of a catalyst to obtain an intermediate compound 5, and adding a nucleophile to react to obtain the N-substituted azaspiro-decone compound shown in the formula II; the synthetic route is as follows:
synthesis path 3: adding a dehydrating agent into the compound E and the compound F to react to obtain an intermediate compound 6, adding an oxidant and a copper salt catalyst to react to obtain an intermediate compound 5, and adding a nucleophile to react to obtain the N-substituted azaspiro-decamonoalkenone compound shown in the formula II; the synthetic route is as follows:
more preferably, compound E isWhen the N-substituted azaspiro-decone compound is prepared, the preparation method comprises the following steps: dissolving 3-chlorobenzaldehyde, cyanamide and methanol in N-bromosuccinimide, reacting under the action of a catalyst to obtain an intermediate compound 7, adding phenylhydrazine and the catalyst to react to obtain an intermediate compound 8, adding 2- (4-methoxyphenoxy) acetic acid, carbodiimide and 1-hydroxybenzotriazole to react to obtain an intermediate compound 9, adding an oxidant and a copper salt catalyst to react to obtain an intermediate compound 10, and adding a nucleophile to react to obtain the N-substituted azaspiro-decamonoalkenone compound shown in formula II; the synthetic route is as follows:
the invention has the advantages that: the intermediate compound before nucleophilic reagent addition is Michael acceptor molecule, which has electrophilicity and can react with various nucleophilic bioactive molecules in vivo to express biological effect, the molecule can be covalently combined with nucleophilic amino acid residues (such as cysteine, arginine and the like) of various proteins, hydroxyl or sulfhydryl to regulate signal paths in cells such as NF-kappa B, STAT3 and the like, but proteins containing nucleophilic amino acid residues and the like are likely to be the acting targets of Michael acceptor molecules, so that toxic and side effects caused by off-target effects are easy to generate, and the intermediate compound and nucleophilic reagent have great potential risk in clinical application, and can reduce the in-vivo addition reaction sites of the Michael acceptor molecule, namely the finally obtained N-substituted azaspirodecanone compound can effectively reduce toxic and side effects and has better safety; meanwhile, the ternary ring, polar or hydrophilic group is introduced into the spirodecenone structure, so that the water solubility of the spirodecenone structure can be further improved, such as the multi-nitrogen heterocyclic structures of imidazole, triazole, piperazine and the like, and the bonding force of the compound and a target point can be improved, and the lipid-water distribution coefficient is improved, so that the pharmacological activity of the spirodecenone is improved.
More preferably, the catalysts are triethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene or potassium tert-butoxide.
More preferably, the oxidizing agents are iodobenzene trifluoroacetate or diacetoxyiodobenzene; the copper salt catalyst is tetraacetonitrile copper perchlorate.
More preferably, the dehydrating agent in synthetic pathway 2 is 1, 3-dicyclohexylcarbodiimide; the dehydrating agent in synthetic pathway 3 is a mixture of carbodiimide and 1-hydroxybenzotriazole.
More preferably, the nucleophiles are ethanethiol, dimethyl malonate, 2H-1,2, 3-triazole, imidazole or 1-methylpiperazine.
More preferably, the organic solvents are all dichloromethane, acetonitrile, methanol, ethanol, diethyl ether, 1, 2-dichloroethane, chloroform, toluene or xylene.
The invention also provides application of the N-substituted azaspiro-decone compounds in preparing antitumor drugs.
The invention has the following beneficial effects:
(1) The method for preparing the N-substituted azaspiro-decarenone compound provided by the invention has moderate reaction, and the prepared N-substituted azaspiro-decarenone compound is not easy to open the ring and has better biological activity.
(2) The N-substituted azaspiro-decarenone compound provided by the invention has good anti-tumor activity, low toxicity, good water solubility and high purity, and the structure is a novel compound.
(3) The N-substituted azaspiro-decarenone compound provided by the invention is simple in synthesis, easy in raw material acquisition and capable of being generated in a large scale.
Detailed Description
The examples given below are only intended to illustrate the invention and are not intended to limit the scope thereof. The specific conditions are not noted in the examples and are carried out according to conventional conditions or conditions recommended by the manufacturer. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention.
The ratios of millimoles (mmol) of the charges in examples 1 to 12 were uniform.
Example 1
10- (ethylsulfanyl) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dode-6-en-8-one (I-6) having the structure shown in the following formula:
the preparation of 10- (ethylsulfanyl) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dode-6-en-8-one in this example was as follows:
s1, dissolving 2-phenoxyethylamine (1.2 mmol) in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine (1.5 mmol) and p-toluenesulfonyl chloride (1 mmol) in 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline, and separating and purifying with polyamide to obtain an intermediate compound 1;
s3, mixing and adding the intermediate compound 1 (0.4 mmol), iodobenzene trifluoroacetate (1 mmol), rhodium diacetate dimer (0.04 mmol) and calcium oxide (4 mmol) into dichloromethane of 25 mL, stirring at 50 ℃ for reaction 6 h, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 2;
s4, dissolving the intermediate compound 2 (0.1 mmol) in an ethanol solution with the volume fraction of 50%, adding ethanethiol (0.2 mmol), stirring at room temperature for reaction 10 h, and separating and purifying by using polyamide after the reaction to obtain 10- (ethylthio) -4-toluenesulfonyl-1-oxygen-4-azaspiro [4.5] dodecan-6-en-8-one (yield 39%).
The nuclear magnetic data of 10- (ethylthio) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dode-6-en-8-one are specifically as follows:
1 H NMR (400MHz, CDCl 3 )δ(ppm):7.79 (d,J= 8.3 Hz, 2H), 7.36 (d,J= 8.3 Hz, 2H), 6.65 (d,J= 10.1 Hz, 1H), 6.05 (d,J= 10.1 Hz, 1H), 4.38 – 4.27 (m, 1H), 4.13 – 4.00 (m, 2H), 3.81 – 3.61 (m, 2H), 2.92 – 2.85 (m, 2H), 2.78 – 2.53 (m, 2H), 2.46 (s, 3H), 1.24 (t,J= 7.4 Hz, 3H);
13 C-NMR (400MHz, CDCl 3 )δ(ppm):196.9, 146.5, 144.3, 136.7, 130.3, 129.9, 127.5, 94.2, 66.4, 49.5, 47.9, 43.9, 25.9, 21.6, 14.9;
HRMS (ESI)m/z(%) forC 17 H 21 NNaO 4 S 2 (M+Na):Calcd. 390.0810Found. 390.0811。
example 2
Dimethyl 2- (8-oxo-4-tosyl-1-oxo-4-azaspiro [4.5] dec-9-en-6-yl) malonate (I-7) having the structure shown in the following formula:
in this example dimethyl 2- (8-oxo-4-tosyl-1-oxo-4-azaspiro [4.5] dec-9-en-6-yl) malonate was prepared as follows:
s1, dissolving 2-phenoxyethylamine in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine and p-toluenesulfonyl chloride in a 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline solution, and separating and purifying with silica gel to obtain an intermediate compound 1;
s3, mixing the intermediate compound 1, iodobenzene trifluoroacetate, rhodium diacetate dimer and calcium oxide, adding the mixture into methylene dichloride, stirring the mixture at 50 ℃ for reaction 8h, and separating and purifying the reacted reaction solution by silica gel to obtain an intermediate compound 2;
s4, dissolving the intermediate compound 2 in acetonitrile, adding 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), adding dimethyl malonate, stirring at room temperature for reaction 7 h, and separating and purifying by silica gel after the reaction to obtain dimethyl 2- (8-oxo-4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dec-9-en-6-yl) malonate (yield 67%).
The nuclear magnetic data of dimethyl 2- (8-oxo-4-tosyl-1-oxo-4-azaspiro [4.5] dec-9-en-6-yl) malonate are specifically as follows:
1 H NMR (400MHz, CDCl 3 )δ(ppm):7.74 (d,J= 8.3 Hz, 2H), 7.33 (d,J= 8.3 Hz, 2H), 6.62 (d,J= 10.2 Hz, 1H), 6.03 (d,J= 10.2 Hz, 1H), 4.06 – 3.91 (m, 2H), 3.80 (m, 2H), 3.72 (s, 3H), 3.69 (s, 3H), 3.63 (m, 1H), 3.51 (d,J= 3.6 Hz, 1H), 3.02 (m, 1H), 2.82 (m, 1H), 2.45 (s, 3H);
13 C NMR (400MHz, CDCl 3 )δ(ppm):197.2, 168.4, 167.9, 145.3, 144.5, 136.6, 130.5, 130.0, 127.4, 94.1, 64.9, 52.8, 52.3, 49.9, 47.3, 42.3, 37.9, 21.6;
HRMS (ESI)m/z(%) forC 20 H 24 NO 8 S (M+H):Calcd. 438.1223Found. 438.1214。
example 3
4-toluenesulfonyl-10- (2)H-1,2, 3-triazol-2-yl) -1-oxo-4-azaspiro [4.5]Dec-6-en-8-one (I-5) has a structure represented by the following formula:
in this example 4-toluenesulfonyl-10- (2)H-1,2, 3-triazol-2-yl) -1-oxo-4-azaspiro [4.5]The preparation process of dec-6-en-8-one is as follows:
s1, dissolving 2-phenoxyethylamine in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine and p-toluenesulfonyl chloride in a 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline solution, and then separating and purifying with polyamide to obtain an intermediate compound 1;
s3, mixing the intermediate compound 1, iodobenzene trifluoroacetate, rhodium diacetate dimer and calcium oxide, adding the mixture into methylene dichloride, stirring the mixture at 50 ℃ for reaction 8h, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 2;
s4, dissolving the intermediate compound 2 in acetonitrile, adding DBU, and then adding 2HStirring to react 8h at room temperature with 1,2, 3-triazole, and separating and purifying with polyamide after the reaction to obtain 4-toluenesulfonyl-10- (2)H-1,2, 3-triazol-2-yl) -1-oxo-4-azaspiro [4.5]Dec-6-en-8-one (47% yield).
4-toluenesulfonyl-10- (2)H-1,2, 3-triazol-2-yl) -1-oxo-4-azaspiro [4.5]The nuclear magnetic data of dec-6-en-8-one are specifically as follows:
1 H NMR (400MHz, CDCl 3 )δ(ppm):7.98 (d,J= 8.4 Hz, 2H), 7.67 (s, 2H), 7.36 (d,J= 8.4 Hz, 2H), 6.82 (d,J= 10.2 Hz, 1H), 6.27 (dd,J= 10.2, 1.1 Hz, 1H), 6.03 (m, 1H), 3.93 – 3.86 (m, 1H), 3.71 (dd,J= 16.7, 12.6 Hz, 1H), 3.47 – 3.39 (m, 1H), 3.06 (m, 1H), 2.96 – 2.87 (m, 2H), 2.46 (s, 3H);
13 C NMR (400MHz, CDCl 3 )δ(ppm):195.2, 144.7, 144.1, 135.9, 134.6, 131.3, 129.7, 128.4, 92.7, 65.7, 63.7, 46.6, 40.5, 21.7;
HRMS (ESI)m/z(%) forC 17 H 19 N 4 O 4 S(M+H):Calcd. 375.1127Found. 375.1119。
example 4
10-(1H-imidazol-1-yl) -4-tosyl-1-oxo-4-azaspiro [4.5]Twelve-6-alkene-8-ketone (I-3) has a structure shown in the following formula:
10- (1) in this exampleH-imidazol-1-yl) -4-tosyl-1-oxo-4-azaspiro [4.5]The preparation process of the dodeca-6-en-8-one is as follows:
s1, dissolving 2-phenoxyethylamine in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine and p-toluenesulfonyl chloride in a 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline solution, and separating and purifying with silica gel to obtain an intermediate compound 1;
s3, mixing the intermediate compound 1, iodobenzene trifluoroacetate, rhodium diacetate dimer and calcium oxide, adding the mixture into methylene dichloride, stirring the mixture at 50 ℃ for reaction 8h, and separating and purifying the reacted reaction solution by silica gel to obtain an intermediate compound 2;
s4, will be in the middleDissolving the compound 2 in acetonitrile, adding DBU, adding imidazole, stirring at room temperature for reaction 8h, and separating and purifying with silica gel to obtain 10- (1)H-imidazol-1-yl) -4-tosyl-1-oxo-4-azaspiro [4.5]Dod-6-en-8-one (61% yield).
10-(1H-imidazol-1-yl) -4-tosyl-1-oxo-4-azaspiro [4.5]The nuclear magnetic data of the dod-6-en-8-one are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.73 (d,J= 8.1 Hz, 2H), 7.69 (s, 1H), 7.38 (d,J= 8.0 Hz, 2H), 7.13 (d,J= 25.3 Hz, 2H), 6.54 (d,J= 10.2 Hz, 1H), 6.18 (d,J= 10.2 Hz, 1H), 5.50 (dd,J= 13.6, 4.4 Hz, 1H), 3.88 (td,J= 7.2, 4.2 Hz, 1H), 3.42 – 3.32 (m, 1H), 3.32 – 3.19 (m, 2H), 3.13 (ddd,J= 8.5, 5.9, 4.2 Hz, 1H), 2.98 – 2.89 (m, 1H), 2.47 (s, 3H);
13 C NMR (101 MHz, Chloroform-d)δ194.69, 145.07, 144.07, 137.40, 135.91, 131.21, 130.17, 129.10, 127.58, 119.27, 92.51, 65.82, 57.47, 47.00, 41.46, 21.65
HRMS(ESI)m/z: calculated for C 18 H 19 N 3 O 4 S [M+H] + 374.1173, found 374.1173。
example 5
10- (4-methylpiperazin-1-yl) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dod-6-en-8-one (I-4) having the structure shown in the following formula:
the procedure for the preparation of 10- (4-methylpiperazin-1-yl) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dod-6-en-8-one in this example is as follows:
s1, dissolving 2-phenoxyethylamine in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine and p-toluenesulfonyl chloride in a 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline solution, and then separating and purifying with polyamide to obtain an intermediate compound 1;
s3, mixing the intermediate compound 1, iodobenzene trifluoroacetate, rhodium diacetate dimer and calcium oxide, adding the mixture into methylene dichloride, stirring the mixture at 50 ℃ for reaction 8h, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 2;
s4, dissolving the intermediate compound 2 in acetonitrile, adding DBU, adding 1-methylpiperazine, reacting at room temperature for 8h, and separating and purifying by polyamide after the reaction to obtain 10- (4-methylpiperazin-1-yl) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dodeca-6-en-8-one (yield 36%).
The nuclear magnetic data of 10- (4-methylpiperazin-1-yl) -4-toluenesulfonyl-1-oxo-4-azaspiro [4.5] dod-6-en-8-one are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.90 – 7.70 (m, 2H), 7.34 (d,J= 8.1 Hz, 2H), 6.64 (d,J= 10.2 Hz, 1H), 6.04 (dd,J= 10.2, 1.3 Hz, 1H), 4.20 – 4.11 (m, 1H), 4.02 (q,J= 7.1 Hz, 1H), 3.90 – 3.81 (m, 1H), 3.75 (dd,J= 13.0, 4.2 Hz, 1H), 3.66 (ddd,J= 8.1, 6.6, 4.6 Hz, 1H), 3.00 – 2.63 (m, 4H), 2.59 – 1.76 (m, 12H);
13 C NMR (101 MHz, Chloroform-d)δ198.67, 146.88, 144.03, 137.43, 130.35, 129.83, 127.32, 95.68, 65.80, 63.58, 55.87, 48.06, 45.83, 36.89, 21.56;
HRMS(ESI)m/z: calculated for C 20 H 27 N 3 O 4 S [M+H] + 406.1788, found 406.1788。
example 6
4- ((4-fluorophenyl) sulfonyl) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]Twelve-6-alkene-8-ketone (I-2) has a structure shown in the following formula:
in this example 4- ((4-fluorophenyl) sulfonyl) -10- (1H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The preparation process of the dodeca-6-en-8-one is as follows:
s1, dissolving 2-phenoxyethylamine in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine and p-fluorobenzenesulfonyl chloride in a 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline solution, and separating and purifying with silica gel to obtain an intermediate compound 1;
s3, mixing the intermediate compound 1, iodobenzene trifluoroacetate, rhodium diacetate dimer and calcium oxide, adding the mixture into methylene dichloride, stirring the mixture at 50 ℃ for reaction 8h, and separating and purifying the reacted reaction solution by silica gel to obtain an intermediate compound 2; the nuclear magnetic data of intermediate compound 2 are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.83 (dd,J= 8.6, 5.0 Hz, 2H), 7.22 (t,J= 8.4 Hz, 2H), 6.50 (d,J= 9.8 Hz, 2H), 6.22 (d,J= 9.8 Hz, 2H), 4.19 (t,J= 6.1 Hz, 2H), 3.73 (t,J= 6.1 Hz, 2H);
13 C NMR (101 MHz, Chloroform-d)δ184.63, 165.46, 143.03, 135.04, 130.50, 129.68, 116.48, 86.29, 65.56, 47.05;
HRMS(ESI)m/z: calculated for C 14 H 12 FNO 4 S [M+H] + 310.0545, found 310.0545;
s4, dissolving the intermediate compound 2 in acetonitrile, adding DBU, adding imidazole, stirring at room temperature for reaction 8H, and separating and purifying by silica gel after the reaction to obtain 4- ((4-fluorophenyl) sulfonyl) -10- (1H-imidazole-1-yl) -1-oxo-4-azaspiro [4.5] dodeca-6-en-8-one (yield 55%).
4- ((4-fluorophenyl) sulfonyl) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]]The nuclear magnetic data of the dod-6-en-8-one are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.95 – 7.80 (m, 2H), 7.73 (s, 1H), 7.34 – 7.22 (m, 2H), 7.15 (d,J= 19.7 Hz, 2H), 6.52 (d,J= 10.2 Hz, 1H), 6.20 (dd,J= 10.2, 1.1 Hz, 1H), 5.51 (dd,J= 13.6, 4.4 Hz, 1H), 3.91 (ddd,J= 8.0, 6.5, 4.4 Hz, 1H), 3.43 – 3.21 (m, 3H), 3.19 – 3.08 (m, 1H), 2.95 (ddd,J= 15.8, 4.4, 1.2 Hz, 1H);
13 C NMR (101 MHz, Chloroform-d)δ194.43, 165.73, 143.63, 137.31, 134.92, 131.52, 130.39, 129.07, 119.26, 116.96, 92.75, 65.81, 57.50, 47.03, 41.50;
HRMS(ESI)m/z: calculated for C 17 H 16 FN 3 O 4 S [M+H] + 378.0916, found 378.0916。
example 7
4- ((4-chlorophenyl) sulfonyl) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]Twelve-6-alkene-8-ketone (I-1) has a structure shown in the following formula:
in this example 4- ((4-chlorophenyl) sulfonyl) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The preparation process of the dodeca-6-en-8-one is as follows:
s1, dissolving 2-phenoxyethylamine in dichloromethane to obtain a 2-phenoxyethylamine solution;
s2, sequentially dissolving triethylamine and p-chlorobenzenesulfonyl chloride in a 2-phenoxyethylamine solution under ice bath, stirring at room temperature for reaction 8h, washing the reacted reaction solution with saturated saline solution, and then separating and purifying with polyamide to obtain an intermediate compound 1;
s3, mixing the intermediate compound 1, iodobenzene trifluoroacetate, rhodium diacetate dimer and calcium oxide, adding the mixture into methylene dichloride, stirring the mixture at 50 ℃ for reaction 8h, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 2; the nuclear magnetic data of intermediate compound 2 are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.80 – 7.66 (m, 2H), 7.57 – 7.43 (m, 2H), 6.57 – 6.40 (m, 2H), 6.29 – 6.14 (m, 2H), 4.19 (t,J= 6.1 Hz, 2H), 3.73 (t,J= 6.1 Hz, 2H);
13 C NMR (101 MHz, Chloroform-d)δ184.60, 142.91, 140.06, 137.44, 129.75, 129.51, 129.11, 86.36, 65.56, 47.08;
HRMS(ESI)m/z: calculated for C 14 H 12 ClNO 4 S [M+H] + 326.0246, found 326.0246;
s4, dissolving the intermediate compound 2 in acetonitrile, adding DBU, adding imidazole, stirring at room temperature for reaction 8h, and separating and purifying by polyamide after the reaction to obtain 4- ((4-chlorophenyl) sulfonyl) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]Dod-6-en-8-one (yield 47%).
4- ((4-chlorophenyl) sulfonyl) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The nuclear magnetic data of the dod-6-en-8-one are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.79 (d,J= 8.2 Hz, 2H), 7.73 (s, 1H), 7.57 (d,J= 8.2 Hz, 2H), 7.15 (d,J= 19.7 Hz, 2H), 6.50 (d,J= 10.2 Hz, 1H), 6.20 (d,J= 10.2 Hz, 1H), 5.50 (dd,J= 13.6, 4.4 Hz, 1H), 3.91 (dt,J= 11.9, 5.4 Hz, 1H), 3.45 – 3.04 (m, 4H), 2.95 (dd,J= 15.8, 4.4 Hz, 1H);
13 C NMR (151 MHz, Chloroform-d)δ194.38, 143.50, 140.65, 137.28, 137.24, 131.54, 129.90, 129.07, 128.91, 119.23, 92.73, 65.78, 57.50, 47.01, 41.44;
HRMS(ESI)m/z: calculated for C 17 H 16 ClN 3 O 4 S [M+H] + 394.0626, found 394.0626。
example 8
Dimethyl-2- (4- (3- (3-chlorophenyl) -1-phenyl-1)H-1,2, 4-triazol-5-yl) -3, 8-dioxo-1-oxo-4-azaspiro [4.5]Dimethyl dodeca-9-en-6-yl malonate (II-3) having the structure shown in the following formula:
in this example dimethyl-2- (4- (3- (3-chlorophenyl) -1-phenyl-1)H-1,2, 4-triazol-5-yl) -3, 8-dioxo-1-oxo-4-azaspiro [4.5]The preparation process of the dimethyl dodecyl-9-en-6-yl) malonate is as follows:
s1, adding 3-chlorobenzaldehyde, cyanamide, methanol, potassium tert-butoxide and N-bromosuccinimide into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 7;
s2, adding the intermediate compound 7, phenylhydrazine and triethylamine into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using polyamide to obtain an intermediate compound 8;
s3, adding the intermediate compound 8, 2- (4-methoxyphenoxy) acetic acid, carbodiimide and 1-hydroxybenzotriazole into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 9;
s4, adding the intermediate compound 9, iodobenzene trifluoroacetate and copper tetraacetonitrile perchlorate into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by polyamide to obtain an intermediate compound 10;
s5, dissolving the intermediate compound 10 in acetonitrile, adding DBU, adding dimethyl malonate, stirring at 30 ℃ for reaction 8h, and separating and purifying by polyamide after the reaction to obtain dimethyl-2- (4- (3- (3-chlorophenyl) -1-phenyl-1)H-1,2, 4-triazol-5-yl) -3, 8-dioxo-1-oxo-4-nitrogenHeterospiro [4.5]]Dimethyl dodeca-9-en-6-yl malonate (yield: 59.6%).
Dimethyl-2- (4- (3- (3-chlorophenyl) -1-phenyl-1)H-1,2, 4-triazol-5-yl) -3, 8-dioxo-1-oxo-4-azaspiro [4.5]The nuclear magnetic data of dimethyl dodecyl-9-en-6-yl) malonate are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ8.05 (d,J= 1.7 Hz, 1H), 7.95 (dt,J= 7.0, 1.6 Hz, 1H), 7.63–7.51 (m, 5H), 7.44–7.33 (m, 2H), 6.52 (d,J= 10.2 Hz, 1H), 6.00 (d,J= 9.5 Hz, 1H), 4.49–4.36 (m, 2H), 3.78 (d,J= 2.7 Hz, 1H), 3.74 (s, 3H), 3.74 (s, 3H), 3.24–3.13 (m, 1H), 3.03 (dd,J= 16.7, 12.8 Hz, 1H), 2.77 (dd,J= 16.4, 4.4 Hz, 1H);
13 C NMR (100 MHz, Chloroform-d)δ196.1, 171.1, 168.1, 167.5, 160.8, 143.7, 142.8, 135.9, 134.7, 132.1, 131.6, 130.4, 123.0, 129.7, 126.6, 125.0, 124.4, 95.0, 66.3, 53.1, 52.5, 49.1, 41.3,36.3;
HRMS(ESI-TOF) m/z: [M+H] + calcd for C 27 H 24 ClN 4 O 7 , 551.1328; found, 551.1324。
example 9
4- (3- (3-chlorophenyl) -1-phenyl-1H-1,2, 4-triazol-5-yl) -10- (1H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The twelve-6-alkene-3, 8-diketone (II-2) has the structure shown in the following formula:
in this example 4- (3- (3-chlorophenyl) -1-phenyl-1H-1,2, 4-triazol-5-yl) -10- (1H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The preparation process of the twelve-6-alkene-3, 8-diketone is as follows:
s1, adding 3-chlorobenzaldehyde, cyanamide, methanol, potassium tert-butoxide and N-bromosuccinimide into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 7;
s2, adding the intermediate compound 7, phenylhydrazine and triethylamine into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 8;
s3, adding the intermediate compound 8, 2- (4-methoxyphenoxy) acetic acid, carbodiimide and 1-hydroxybenzotriazole into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 9;
s4, adding the intermediate compound 9, iodobenzene trifluoroacetate and copper tetraacetonitrile perchlorate into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 10;
s5, dissolving the intermediate compound 10 in acetonitrile, adding DBU, adding imidazole, stirring at 30 ℃ for reaction 8h, and separating and purifying by silica gel after the reaction to obtain 4- (3- (3-chlorophenyl) -1-phenyl-1)H-1,2, 4-triazol-5-yl) -10- (1H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]Twelve-6-alkene-3, 8-diketone (yield: 46.6%).
4- (3- (3-chlorophenyl) -1-phenyl-1H-1,2, 4-triazol-5-yl) -10- (1H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The nuclear magnetic data of the twelve-6-alkene-3, 8-diketone are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ8.13 (s, 1H), 8.03 (d,J= 8.9 Hz, 2H), 7.58–7.51 (m, 3H), 7.50–7.42 (m, 4H), 7.20 (s, 1H), 6.38 (d,J= 10.2 Hz, 1H), 6.25 (d,J= 10.2 Hz, 1H), 5.72 (dd,J= 13.8, 4.3 Hz, 1H), 4.25–4.06 (m, 2H), 3.50–3.38 (m, 2H), 3.06 (dd,J= 15.8, 3.8 Hz, 1H);
HRMS (ESI-TOF) m/z: [M+H] + calcd for C 25 H 20 ClN 6 O 3 , 487.1280; found, 487.1281。
example 10
4- (3- (3-chlorophenyl) -1-phenyl-1H-1,2, 4-trisOxazol-5-yl) -10- (ethylsulfanyl) -1-oxo-4-azaspiro [4.5]The twelve-6-alkene-3, 8-diketone (II-5) has the structure shown in the following formula:
in this example 4- (3- (3-chlorophenyl) -1-phenyl-1H-1,2, 4-triazol-5-yl) -10- (ethylsulfanyl) -1-oxo-4-azaspiro [4.5]The preparation process of the twelve-6-alkene-3, 8-diketone is as follows:
s1, adding 3-chlorobenzaldehyde, cyanamide, methanol, potassium tert-butoxide and N-bromosuccinimide into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 7;
s2, adding the intermediate compound 7, phenylhydrazine and triethylamine into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 8;
s3, adding the intermediate compound 8, 2- (4-methoxyphenoxy) acetic acid, carbodiimide and 1-hydroxybenzotriazole into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 9;
s4, adding the intermediate compound 9, iodobenzene trifluoroacetate and copper tetraacetonitrile perchlorate into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 10;
s5, dissolving the intermediate compound 10 in acetonitrile, adding DBU, adding ethanethiol, stirring at 30 ℃ for reaction 8h, and separating and purifying by silica gel after the reaction to obtain the 4- (3- (3-chlorophenyl) -1-phenyl-1)H-1,2, 4-triazol-5-yl) -10- (ethylsulfanyl) -1-oxo-4-azaspiro [4.5]Twelve-6-alkene-3, 8-diketone (yield: 42.3%).
4- (3- (3-chlorophenyl) -1-phenyl-1H-1,2, 4-triazol-5-yl) -10- (ethylsulfanyl) -1-oxo-4-nitrogenHeterospiro [4.5]]The nuclear magnetic data of the twelve-6-alkene-3, 8-diketone are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ8.06 (d,J= 1.8 Hz, 1H), 7.95 (td,J= 6.8, 1.7 Hz, 1H), 7.65 (dd,J= 8.2, 1.5 Hz, 2H), 7.59–7.55 (m, 1H), 7.55–7.49 (m, 2H), 7.42–7.35 (m, 2H), 6.56 (d,J= 10.1 Hz, 1H), 6.12 (dd,J= 10.1, 1.0 Hz, 1H), 4.75 (d,J= 14.4 Hz, 1H), 4.37 (d,J= 14.4 Hz, 1H), 4.23 (dd,J= 13.0, 5.0 Hz, 1H), 3.12–2.87 (m, 3H), 2.69 (dd,J= 12.5, 7.5 Hz, 1H), 1.33–1.28 (m, 3H);
13 C NMR (100 MHz, Chloroform-d)δ196.1, 169.7, 160.1, 144.4, 143.3, 137.0, 134.8, 133.4, 131.7, 130.0, 130.0, 129.7, 129.7, 126.4, 124.3, 124.3, 94.9, 68.0, 49.1, 43.1, 26.2, 15.1。
example 11
4- (2, 6-dichlorobenzene) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]Dodecyl-6-ene-3, 8-dione
(II-7) having a structure represented by the following formula:
in this example 4- (2, 6-dichlorobenzene) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The preparation process of the twelve-6-alkene-3, 8-diketone is as follows:
s1, adding para-aminophenol, glycollic acid and dimethyl dicyandiamide into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 3;
s2, adding the intermediate compound 3, diacetoxyiodobenzene, tetraacetonitrile copper perchlorate and methylene dichloride into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 4;
s3, adding the intermediate compound 4, DBU, tetrahydrofuran and 2, 6-dichlorobenzyl chloride into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 5;
s4, dissolving the intermediate compound 5 in acetonitrile, adding DBU, adding imidazole, stirring at 30 ℃ for reaction 8h, and separating and purifying by silica gel after the reaction to obtain 4- (2, 6-dichlorobenzene) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]Twelve-6-alkene-3, 8-diketone (yield: 53.3%).
4- (2, 6-dichlorobenzene) -10- (1)H-imidazol-1-yl) -1-oxo-4-azaspiro [4.5]The nuclear magnetic data of the twelve-6-alkene-3, 8-diketone are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.53 (s, 1H), 7.41–7.36 (m, 2H), 7.31 (dd,J= 9.1, 6.8 Hz, 1H), 7.15 (s, 1H), 7.00 (s, 1H), 6.06 (d,J= 10.2 Hz, 1H), 5.79 (d,J= 10.2 Hz, 1H), 5.36 (d,J= 15.2 Hz, 1H), 5.22 (dd,J= 13.4, 4.3 Hz, 1H), 4.59 (d,J= 15.2 Hz, 1H), 4.26 (d,J= 13.8 Hz, 1H), 3.70 (d,J= 13.8 Hz, 1H), 3.34 (dd,J= 15.6, 13.5 Hz, 1H), 2.91 (dd,J= 15.6, 4.3 Hz, 1H);
13 C NMR (100 MHz, Chloroform-d)δ193.9, 168.3, 144.4, 136.8, 136.5, 131.2, 131.1, 130.3, 130.1, 129.1, 118.3, 92.2, 66.4, 54.8, 40.5, 39.2;
HRMS(ESI-TOF) m/z: [M+H] + calcd for C 18 H 16 N 3 O 3 Cl 2 , 392.0563, found, 392.0577。
example 12
4- (4-chlorophenyl) -10- (ethylsulfanyl) -1-oxo-4-azaspiro [4.5] dod-6-en-3, 8-dione (II-8) has a structure represented by the following formula:
the preparation of 4- (4-chlorophenyl) -10- (ethylsulfanyl) -1-oxo-4-azaspiro [4.5] dode-6-ene-3, 8-dione in this example was as follows:
s1, adding para-aminophenol, glycollic acid and dimethyl dicyandiamide into a reaction bottle, uniformly stirring, reacting for 8 hours at room temperature, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 3;
s2, adding the intermediate compound 3, diacetoxyiodobenzene, tetraacetonitrile copper perchlorate and methylene dichloride into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 4;
s3, adding the intermediate compound 4, DBU, tetrahydrofuran and 2, 6-dichlorobenzyl chloride into a reaction bottle, uniformly stirring, reacting at room temperature for 8 hours, and separating and purifying the reacted reaction solution by using silica gel to obtain an intermediate compound 5;
s4, dissolving the intermediate compound 5 in acetonitrile, adding DBU, adding ethanethiol, stirring at 30 ℃ for reaction 8h, and separating and purifying by silica gel after the reaction to obtain 4- (4-chlorobenzene) -10- (ethylthio) -1-oxo-4-azaspiro [4.5] dodeca-6-alkene-3, 8-dione (yield: 41.1%).
The nuclear magnetic data of 4- (4-chlorobenzene) -10- (ethylthio) -1-oxo-4-azaspiro [4.5] dod-6-en-3, 8-dione are specifically as follows:
1 H NMR (400 MHz, Chloroform-d)δ7.33–7.27 (m, 4H), 6.38 (d,J= 10.1 Hz, 1H), 6.06 (dd,J= 10.1, 1.1 Hz, 1H), 4.57 (d,J= 13.5 Hz, 1H), 4.48–4.37 (m, 3H), 3.19 (dd,J= 13.0, 4.4 Hz, 1H), 3.01 (dd,J= 16.2, 13.0 Hz, 1H), 2.81 (td,J= 16.2, 4.4, 1.1 Hz, 1H), 2.34 (td,J= 11.8, 7.4 Hz, 1H), 2.11 (dd,J= 11.8, 7.4 Hz, 1H), 1.13 (t,J= 7.4 Hz, 3H);
13 C NMR (100 MHz, Chloroform-d)δ196.3, 170.7, 146.1, 134.4, 134.3, 132.5, 130.2, 129.0, 93.8, 68.0, 48.8, 43.3, 42.4, 25.2, 14.6;
HRMS(ESI-TOF) m/z: [M+H] + calcd for C 17 H 19 NO 3 SCl, 352.0769, found, 352.0785。
example 13 determination of antitumor Activity
Proliferation of adherent cells was detected using the Sulfonyl Rhodamine B (SRB) method. The positive control drugs were bendamustine and vorinostat. Human breast cancer cells MDA-MB-231 in the logarithmic growth phase were inoculated into 96-well plates (200. Mu.L/well), and cultured at 37℃and 5% carbon dioxide concentration for adherence to 24 h. Then, the sample was treated with different concentrations of the test compound, 3 duplicate wells were set for each concentration, and corresponding blank and zeroed wells were set. After incubation 72 h, 1h was fixed with 10% trichloroacetic acid (TCA) at 4 ℃, the fixed solution was poured, washed 5 times with distilled water, and naturally air-dried. After completion of fixation, the cells were stained with 0.4% (w/v) SRB at room temperature for 20 min, the solution was poured, washed 5 times with 1% glacial acetic acid, the excess unbound SRB dye was removed, and air-dried. SRB-binding proteins were solubilized with 10 mM Tris (hydroxymethyl amino) methane (Tris), shaking with a horizontal shaker for 20 min, and using equivalent amounts of DMSO as vehicle controls, bendamustine (bifunctional alkylating agent) and vorinostat (SAHA, HDAC inhibitor) as positive controls. The cell number was counted using a cell viability analyzer (Beckman-Coulter). Survival is the ratio of the number of compound-treated cells to the number of vector-treated cells, and IC is obtained from the dose-response curve for each compound 50 Values. Experiments were performed in triplicate and data are presented as average values.
In vitro antitumor activity experiments were performed on the compounds of examples 1 to 12, and the in vitro cell growth inhibition ability of the target compounds on human breast cancer cells MDA-MB-231 was tested using bendamustine (a bifunctional alkylating agent) and vorinostat (an SAHA, HDAC inhibitor) as positive drug controls, and the results are shown in Table 1. As can be seen from Table 1, after the nucleophilic site in intermediate compound 2 is added, the activity of the prepared N-substituted azaspiro-decarenone compound is significantly improved compared with that before the nucleophilic site is added except for examples 4 and 5; meanwhile, N-substituted azaspiro-decene prepared in examples 1 to 12The ketone compounds also show a certain proliferation inhibition activity (IC) on lung cancer cell line A549 and cervical cancer cell line Hela cells 50 0.13~4.66 μM)。
TABLE 1
Example 14 acute toxicity assay and analysis of Compound dissolution behavior
Acute toxicity tests were conducted on the intermediate compound 2 and the N-substituted azaspiro-decone compounds prepared in examples 1 to 7, SPF-grade Km mice were divided into two groups, and the two groups of mice were respectively administered with different doses of the intermediate compound 2 and the N-substituted azaspiro-decone compounds, the administration doses were 120 to 1000mg/kg, and 14 days of observation was conducted on the mice after administration. The results show that the mice in the group of the intermediate compound 2 die, and the mice in the group of the N-substituted azaspiro-decamonooleone compound do not die, and the mice have no obvious change in behavior, normal diet, no abnormal secretion and continuous increase of body weight; the intermediate compound 2 is shown to be more toxic than the N-substituted azaspiro-decone compounds, i.e. the toxicity can be reduced after the nucleophilic site in the intermediate compound 2 is added. Meanwhile, the in-vivo and in-vitro activity research results show that the activity of certain compounds (I-2) in the N-substituted azaspiro-decarenone compounds synthesized by eliminating the single-side alpha, beta-unsaturated ketone structure or modifying other types of the intermediate compounds 2 can be raised to the nanomolar level, and the toxicity is greatly reduced; animal toxicity experiments show that the LD of the intermediate compound 2 50 66.1-80 mg/kg, which belongs to the medium toxicity range, and the half lethal dose of the N-substituted azaspiro-decylenone compound to Km mice is more than 120-1000 mg/kg.
Carrying out an animal level antitumor activity experiment, randomly dividing 24 BALB/c tumor-bearing mice into 4 groups, wherein 6 BALB/c tumor-bearing mice are respectively a blank control group, a positive drug control-bendamustine control group, namely 10 mg/kg, a low-dose N-substituted azaspiro-decarenone compound group, namely 10 mg/kg, and a high-dose N-substituted azaspiro-decarenone compound group, namely 20 mg/kg; four groups of mice were dosed once every two days, 7 times consecutively, and mice signs were observed over 14 days. The result shows that the tumor inhibition rate of the N-substituted azaspiro-decarenone compound on the BALB/c breast cancer model mice reaches 61%; the results of histopathological section and staining show that the compounds do not cause obvious lesions of normal tissues. Namely, the N-substituted aza-spiro-decone ketone compound reduces the protein toxicity site on the parent nucleus on the premise of retaining the basic structure of the spiro-decone, thereby reducing the toxicity of the compound in vivo.
Meanwhile, as nucleophilic site addition reaction is carried out under the condition of ethanol/water or acetonitrile/organic alkali, residual transition metal salt is dissolved in the system, and the final product is further purified after the process, so that the purity of the prepared N-substituted azaspiro-decarenone compound is improved.
In addition, the lipid partition coefficient is an important index, and determines the distribution of the compound in the body. Suitable water solubility aids in dissolving the drug in the body fluid to achieve an effective concentration at the site of action, thereby producing a therapeutic effect. For the antitumor drugs, the better water solubility not only facilitates absorption and distribution, but also enables the drugs to be discharged rapidly, and reduces accumulation and toxic and side effects on kidneys. The incorporation of one or more heteroatoms in the N-substituted azaspiro-decarenone compound increases the likelihood of hydrogen bonding with water, which improves the dissolution behavior compared to intermediate 2, as is evident from the observation that 2 mg N-substituted azaspiro-decarenone compound dissolves in 200. Mu.L of DMSO and 1.8 mL olive oil solution while 1 mg intermediate 2 remains in suspension in 200. Mu.L of DMSO and 1.8 mL olive oil solution when animal level antitumor activity studies are conducted. Thus being more beneficial to improving the binding force of the compound and the target spot and improving the lipid-water distribution coefficient so as to improve the pharmacological activity of the compound.
While the invention has been described in terms of the foregoing embodiments, it will be understood that the embodiments are not intended to limit the invention in any way, but are intended to cover modifications of the invention using equivalent alternatives or modifications.

Claims (10)

  1. An N-substituted azaspiro-decarenone compound, wherein the N-substituted azaspiro-decarenone compound has a structure as shown in formula I or formula II:
    wherein:
    r is H, C 1-20 Linear or branched alkyl, halogen, alicyclic, aromatic ring, carboxyl, hydroxyl, amino, mercapto, carbo-acylated derivative, phosphorylated derivative, phosphitylated derivative or thiolated derivative;
    R 2 is an aromatic sulfonyl or substituted aromatic sulfonyl, aromatic carbonyl or substituted aromatic carbonyl, aromatic phosphoryl or substituted aromatic phosphoryl, aromatic ring or substituted aromatic ring, aliphatic ring or substituted aliphatic ring, heterocyclic ring or substituted heterocyclic ring, benzyl or substituted benzyl;
    R 3 is C 1-20 Linear or branched alkyl, halogen, alicyclic, aromatic ring, heterocyclic, carboxyl, hydroxyl, amino, mercapto, carbo-acylated derivative, phosphorylated derivative, phosphitylated derivative or thiolated derivative;
    x is oxygen, sulfur, nitrogen or carbon element, and n is 0, 1,2,3, 4 or 5.
  2. 2. The N-substituted azaspiro-decarenone compound of claim 1, wherein R is H, halogen, or an aromatic ring; the R is 2 Is an aromatic sulfonyl or substituted aromatic sulfonyl, aromatic ring or substituted aromatic ring, heterocycle or substituted heterocycle, benzyl or substituted benzyl; the R is 3 Is a heterocycle, carboxyl, mercapto or carboacylated derivative; x is oxygen; and n is 1.
  3. 3. The N-substituted azaspiro-decarenone compound of claim 2, having the specific structural formula:
  4. 4. a method for preparing an N-substituted azaspiro-decarenone compound according to any one of claims 1 to 3, comprising the following synthetic routes:
    synthesis path 1: dissolving a compound A and a compound B in an organic solvent, reacting under the action of a catalyst to obtain an intermediate compound 1, adding an oxidant to react to obtain an intermediate compound 2, and adding a nucleophile to react to obtain an N-substituted azaspiro-decamonoalkone compound shown in a formula I; the synthetic route is as follows:
    synthesis path 2: dissolving a compound C and a compound D in an organic solvent, reacting under the action of a dehydrating agent to obtain an intermediate compound 3, adding an oxidant to react to obtain an intermediate compound 4, adding a compound B, reacting under the action of a catalyst to obtain an intermediate compound 5, and adding a nucleophile to react to obtain the N-substituted azaspiro-decone compound shown in the formula II; the synthetic route is as follows:
    synthesis path 3: adding a dehydrating agent into the compound E and the compound F to react to obtain an intermediate compound 6, adding an oxidant and a copper salt catalyst to react to obtain an intermediate compound 5, and adding a nucleophile to react to obtain the N-substituted azaspiro-decamonoalkenone compound shown in the formula II; the synthetic route is as follows:
  5. 5. the method for preparing an N-substituted azaspiro-decone compound according to claim 4, wherein the catalysts are triethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene or potassium tert-butoxide.
  6. 6. The method for preparing an N-substituted azaspiro-decarenone compound of claim 4, wherein the oxidizing agents are iodobenzene trifluoroacetate or diacetoxyiodobenzene; the copper salt catalyst is tetraacetonitrile copper perchlorate.
  7. 7. The method for preparing an N-substituted azaspiro-decarenone compound according to claim 4, wherein the dehydrating agent in the synthetic pathway 2 is 1, 3-dicyclohexylcarbodiimide; the dehydrating agent in the synthetic route 3 is a mixture of carbodiimide and 1-hydroxybenzotriazole.
  8. 8. The method for preparing N-substituted azaspiro-decone ketone according to claim 4, wherein the nucleophiles are ethanethiol, dimethyl malonate, 2H-1,2, 3-triazole, imidazole or 1-methylpiperazine.
  9. 9. The method for preparing an N-substituted azaspiro-decarenone compound according to claim 4, wherein the organic solvents are dichloromethane, acetonitrile, methanol, ethanol, diethyl ether, 1, 2-dichloroethane, chloroform, toluene or xylene.
  10. 10. Use of an N-substituted azaspiro-decarenone compound according to any one of claims 1 to 3 for the preparation of an antitumor drug.
CN202410208065.9A 2024-02-26 2024-02-26 N-substituted azaspiro-decone compounds, preparation method and application thereof Active CN117777047B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410208065.9A CN117777047B (en) 2024-02-26 2024-02-26 N-substituted azaspiro-decone compounds, preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410208065.9A CN117777047B (en) 2024-02-26 2024-02-26 N-substituted azaspiro-decone compounds, preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN117777047A true CN117777047A (en) 2024-03-29
CN117777047B CN117777047B (en) 2024-04-30

Family

ID=90379982

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410208065.9A Active CN117777047B (en) 2024-02-26 2024-02-26 N-substituted azaspiro-decone compounds, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN117777047B (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3692797A (en) * 1970-04-24 1972-09-19 Hoffmann La Roche 3-(5-nitro-2-thienyl)-1-oxa-2,4-diazaspiro(4,5)-dec-2-ene
CN102285934A (en) * 2009-01-08 2011-12-21 四川大学 Spirocycle dienone derivates as well as preparation method and application thereof
CN102850337A (en) * 2011-06-30 2013-01-02 四川大学 Multi-azole linked spirorenone compound and preparation method and application thereof
US20160264522A1 (en) * 2013-10-23 2016-09-15 Janssen Sciences Ireland Uc Carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US20170002025A1 (en) * 2015-07-02 2017-01-05 Janssen Sciences Ireland Uc Cyclized sulfamoylarylamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US20170253609A1 (en) * 2016-03-07 2017-09-07 Enanta Pharmaceuticals, Inc. Hepatitis b antiviral agents
CN110183503A (en) * 2019-04-19 2019-08-30 四川大学 Sulphonyl azaspiro decadinene ketone compounds and application thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3692797A (en) * 1970-04-24 1972-09-19 Hoffmann La Roche 3-(5-nitro-2-thienyl)-1-oxa-2,4-diazaspiro(4,5)-dec-2-ene
CN102285934A (en) * 2009-01-08 2011-12-21 四川大学 Spirocycle dienone derivates as well as preparation method and application thereof
CN102850337A (en) * 2011-06-30 2013-01-02 四川大学 Multi-azole linked spirorenone compound and preparation method and application thereof
US20160264522A1 (en) * 2013-10-23 2016-09-15 Janssen Sciences Ireland Uc Carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US20170002025A1 (en) * 2015-07-02 2017-01-05 Janssen Sciences Ireland Uc Cyclized sulfamoylarylamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US20170253609A1 (en) * 2016-03-07 2017-09-07 Enanta Pharmaceuticals, Inc. Hepatitis b antiviral agents
CN110183503A (en) * 2019-04-19 2019-08-30 四川大学 Sulphonyl azaspiro decadinene ketone compounds and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HONGLU YIN,等: "Design, synthesis, and antitumor study of a series of novel 1-Oxa-4-azaspironenone derivatives", 《BIOORGANIC & MEDICINAL CHEMISTRY LETTERS》, vol. 74, 6 August 2022 (2022-08-06), pages 128925, XP087166293, DOI: 10.1016/j.bmcl.2022.128925 *

Also Published As

Publication number Publication date
CN117777047B (en) 2024-04-30

Similar Documents

Publication Publication Date Title
CA2758904C (en) Novel compounds of reverse-turn mimetics, method for manufacturing the same and use thereof
AU2002254400B2 (en) Molecular conjugates for use in treatment of cancer
AU617304B2 (en) Novel cc-1065 analogs
HUE030685T2 (en) Macrocyclic purines for the treatment of viral infections
EP2949647B1 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing same
JP2013544279A (en) Process for the preparation of morpholinyl anthracycline derivatives
TW200815456A (en) Artemisinin derivative dimers, their preparation and their therapeutic application
KR20220052918A (en) Peptide conjugates of cytotoxins as therapeutic agents
EP3539964B1 (en) Base acetamide glycerin ether molecule, chemical synthesis method therefor, and applications thereof in field of gene therapy
Rodrigues et al. Correlation of the acid-sensitivity of polyethylene glycol daunorubicin conjugates with their in vitro antiproliferative activity
CN114349738B (en) Small molecule conjugate for targeted degradation of CDK2 and application thereof
CN117777047B (en) N-substituted azaspiro-decone compounds, preparation method and application thereof
CN110183503B (en) Sulfonyl azaspirodecadienone compounds and application thereof
JP7260717B2 (en) Lipidoids and their use for nucleic acid transfection
CA3221027A1 (en) Cyclohexane lipidoids for nucleic acid transfection and use thereof
CN113234117A (en) Hederagenin C-28 polyethylene glycol modified derivative and preparation method thereof
CN113698415A (en) Novel oridonin analogue and derivative, preparation method and medical application thereof
KR101465626B1 (en) Target-specific anti-cancer prodrug comprising a biotin
EP3828196A1 (en) Method for preparing antibody-drug conjugate intermediate by means of acid method and application thereof
CN115057850B (en) Aloe-emodin derivative and preparation method and application thereof
CN114380780B (en) Novel long tannin extract A derivative, preparation method and medical application thereof
CN113527405B (en) Application of hederagenin polyethylene glycol modified derivative in preparation of tumor drug resistance reversal agent
KR102620495B1 (en) Cyclic dinucleotide prodrug molecules and their preparation methods and applications
EP4201429A1 (en) Antibody-drug conjugate intermediate comprising sn38 and preparation method therefor
CA3076714C (en) Method for producing antibody-drug conjugate intermediate by addition of acid and use thereof

Legal Events

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