Proteolysis targeting chimera compound with oxido bicycloheptene compound as estrogen receptor ligand, preparation method and application

文档序号:1094427 发布日期:2020-09-25 浏览:11次 中文

阅读说明:本技术 以氧桥双环庚烯类化合物为***受体配体的蛋白水解靶向嵌合体化合物及制备方法与应用 (Proteolysis targeting chimera compound with oxido bicycloheptene compound as estrogen receptor ligand, preparation method and application ) 是由 周海兵 胡志烨 宁文涛 黄健 董春娥 于 2020-06-23 设计创作,主要内容包括:本发明公开了以氧桥双环庚烯类化合物为雌激素受体配体的蛋白水解靶向嵌合体化合物及制备方法与应用。采用两种合理的合成方式,以VHL配体或CRBN配体作为E3连接酶配体部分,通过不同长度烷基侧链连接氧桥双环庚烯类磺酸酯或磺酰胺雌激素受体配体,合成得到一系列目标产物Protac分子。这类Protac分子与现有的抗乳腺癌药物他莫昔芬的作用方式有所不同,是一类靶向雌激素下调剂。此类化合物不仅保留一定与雌激素受体结合的能力,而且具有与氟维司群相当的良好的雌激素受体α下调活性,可以实现事件驱动型靶向雌激素受体降解,有望通过该法来克服ER阳性乳腺癌传统内分泌治疗带来的耐药。(The invention discloses a proteolytic targeting chimeric compound taking an oxido-bicycloheptene compound as an estrogen receptor ligand, a preparation method and application thereof. Two reasonable synthesis modes are adopted, a VHL ligand or a CRBN ligand is used as a ligand part of an E3 ligase, alkyl side chains with different lengths are connected with oxygen bridge bicycloheptene sulfonate or sulfonamide estrogen receptor ligands, and a series of Protac molecules serving as target products are synthesized. The Protac molecules have different action modes from the existing anti-breast cancer medicament tamoxifen, and are targeted estrogen down-regulating agents. The compound not only retains certain capacity of being combined with estrogen receptors, but also has good estrogen receptor alpha down-regulation activity equivalent to fulvestrant, can realize event-driven targeted estrogen receptor degradation, and is expected to overcome the drug resistance brought by the traditional endocrine therapy of ER positive breast cancer by the method.)

1. A proteolysis targeting chimera compound with an oxido bicycloheptene compound as an estrogen receptor ligand is characterized by having the following general structure:

2. the proteolytic targeting chimeric compound using oxido-bicycloheptene as estrogen receptor ligand according to claim 1, wherein the compound is selected from any one of the following:

(2S,2R) -1- ((2S) -2- (6- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) hexanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- (4- (4-methylthiazol-5-yl) benzyl) pyrrolidine-2-carboxamide (26b),

(2S,2R) -1- ((2S) -2- (7- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) heptanamide) -3, 3-dimethylbutanoyl) -4-hydroxy-N- (4- (4-methylthiazol-5-yl) benzyl) pyrrolidine-2-carboxamide (26c),

(2S,2R) -1- ((2S) -2- (5- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) pentanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1- (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26g),

(2S,2R) -1- ((2S) -2- (6- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) hexanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1- (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26h),

(2S,2R) -1- ((2S) -2- (7- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) heptanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1- (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26i),

(2S,2R) -1- ((2S) -2- (6- (4- ((5, 6-bis (4-hydroxyphenyl) -N- (2,2, 2-trifluoroethyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonamide) phenoxy) hexanamino) -3, 3-dimethylbutyryl) -4-hydroxy-N- (4- (4-methylthiazol-5-yl) benzyl) pyrrolidine-2-carboxamide (26k),

(2S,2R) -1- ((2S) -2- (6- (4- ((5, 6-bis (4-hydroxyphenyl) -N- (2,2, 2-trifluoroethyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonamide) phenoxy) hexanamino) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1-4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26l),

2-chloro-4- ((6- (((S) -1- ((2S,4R) -4-hydroxy-2- (((4- (4-methylthiazol-5-yl) benzyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxybutan-2-yl) amino) -6-oxyhexyl) oxy) -5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene-2-sulfonate (26m),

2-chloro-4- ((6- (((S) -1- ((2S,4R) -4-hydroxy-2- (((4- (4-methylthiazol-5-yl) phenyl) ethyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxybutan-2-yl) amino) -6-oxyhexyl) oxy) -5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene-2-sulfonate (26 n).

3. The use of the proteolytic targeting chimeric compound as claimed in claim 1 or 2, wherein said compound is an estrogen receptor ligand, in the manufacture of a medicament against breast cancer.

4. The method of preparing the proteolytic targeting chimeric compound using the oxidocycloheptene compound as estrogen receptor ligand according to claim 1 or 2, wherein the Boc-tert-leucine hydroxyproline thiazole derivative 9, the E3 ligands 12a-i and 15 with long side chains, and the ER ligand portion 18,21a-b and 24 with caproic acid side chains are synthesized by the following reaction, comprising the following steps:

1) tert-butyl ((S) -1- ((2S,4R) -4-hydroxy-2- ((4- (4-methylthiazol-5-yl) benzyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxobutan-2-yl) carbamate 9a and tert-butyl ((S) -1- ((2S, synthesis of 4R) -4-hydroxy-2- (((S) -1- (4- (4-methylthiazol-5-yl) phenethyl) carbamoyl) pyrrol-idin-1-yl) -3, 3-dimethyl-1-oxybutynin-2-yl) carbamate 9 b:

reagents and reaction conditions: (a) di-tert-butyl dicarbonate, dichloromethane and triethylamine are carried out for 12 hours at 25 ℃; (b) dimethyl acetamide, methyl acetate, dimethyl acetamide, reflux at 150 deg.c and reaction overnight; (c) trifluoroacetic acid, dichloromethane, 25 ℃, 0.5 hours; (d)2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, Boc-L-hydroxyproline, N, N-dimethylformamide, at 25 ℃ for 1 hour; (e) trifluoroacetic acid, dichloromethane, 25 ℃, 0.5 hour; 2.2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, Boc-L-tert-leucine, N, N-dimethylformamide, at 25 ℃ for 0.5 hour;

(1) synthesis of tert-butyl (4-bromobenzyl) carbamate 4a and (R) - (1- (4-bromophenylethyl) carbamate 4 b:

respectively weighing 1.99g of compound 4-bromobenzylamine 3a or 2.14g of analogue (R) -1- (4-bromophenyl) ethyl-1-amine 3b thereof, placing the compound or the analogue in two 50mL double-mouth bottles, vacuumizing, introducing argon, sequentially adding 10mL of dichloromethane and 2.26mL of triethylamine, slowly adding 10mL of dichloromethane solution dissolved with 2.82g of di-tert-butyl dicarbonate after ice-cooling for 10 minutes, finishing the addition after 5 minutes, and reacting at room temperature overnight; after TLC confirms that the reaction is complete, adding water for quenching, extracting by dichloromethane, drying by organic phase anhydrous sodium sulfate, filtering, concentrating, drying by spinning, and separating and purifying by silica gel column chromatography to obtain a white solid compound 4a or 4 b;

(2) synthesis of tert-butyl (4- (4-methylthiazol-5-yl) benzyl) carbamate 6a and (R) - (1- (4- (4- (4-methylthiazol-5-yl) phenyl) ethyl) carbamate tert-butyl 6 b:

under the protection of argon, respectively weighing 3.04g of 4a or 3.18g of 4b, 1.98g of 4-methylthiazole, 225mg of palladium acetate and 3.93g of potassium acetate, putting the obtained mixture into a 100mL round-bottom flask, adding 25mL of solvent N, N-dimethylacetamide, reacting at 150 ℃ overnight, confirming the reaction by TLC, adding water for quenching, extracting by dichloromethane, washing an organic phase by water, drying by anhydrous sodium sulfate, filtering, concentrating, spin-drying, and separating and purifying by silica gel column chromatography to obtain a white solid compound 6a or 6 b;

putting the compound into a single-mouth bottle with proper volume, adding dichloromethane to dissolve the compound, dropwise adding trifluoroacetic acid with the same volume as the dichloromethane under ice bath, reacting for 1 hour at the temperature, and adding the dichloromethane for many times after spin-drying to remove the trifluoroacetic acid;

general procedure for the condensation of amino compounds and carboxylic acid compounds to amides: weighing an amino compound in a two-mouth bottle under the protection of argon, dissolving a small amount of anhydrous N, N-dimethylformamide, sequentially adding a carboxylic acid compound, slowly dropwise adding N, N-diisopropylethylamine, stirring at room temperature for 5 minutes, adding 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea Hexafluorophosphate (HATU), detecting by TLC, adding water for quenching after complete reaction, extracting with ethyl acetate (3X 30mL), washing an organic layer with saturated salt water for more than three times to remove a reaction solvent N, N-dimethylformamide, drying the organic layer with anhydrous sodium sulfate, distilling under reduced pressure to remove the solvent, and purifying by column chromatography to obtain a product;

(3) after the compound 6a or 6b is subjected to Boc removal reaction to obtain a compound 7a or 7b, the amino part of the compound is condensed with carboxyl in Boc-L-hydroxyproline to obtain a compound 8a or 8b, and then the compound is subjected to one-step Boc removal reaction and is subjected to condensation reaction with N-Boc-L-tertiary leucine to obtain a yellow solid compound 9a or 9 b;

2) synthesis of E3 ligands 12a-i and 15 with long side chains:

Figure FDA0002552570130000041

reagents and reaction conditions: (a) trifluoroacetic acid, dichloromethane, 25 ℃, 0.5 hours; (b)2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, N, N-dimethylformamide, at 25 deg.C for 0.5 hr; (c) refluxing thionyl chloride and dichloromethane for 4 hours; (d) tetrahydrofuran, reflux, overnight;

(1) synthesis of 6- ((1- ((2S,4R) -4-hydroxy-2- ((4- (4-methylthiazol-5-yl) benzyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxybutan-2-yl) amino) hexanoic acid analog 12:

compound 1.0equiv.9a or 9b was weighed out in TFA: in a mixed solution of 1:1 of DCM, after reacting for 30min, the solvent is distilled off under reduced pressure to obtain deprotected intermediate compound 10a or 10b, under the protection of argon, dissolving an intermediate compound 10a or 10b in a 25mL two-necked bottle by using anhydrous DMF, adding 1.0equiv. bromo alkyl acid 11a-f with different lengths, dropwise adding N, N-diisopropylethylamine (8.0equiv.), stirring at room temperature for 5min, adding 1.1equiv.2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate, reacting at room temperature for 30min, adding water for quenching, extracting by using ethyl acetate, washing an organic layer by using saturated common salt water, drying by using sodium sulfate, distilling under reduced pressure to remove a solvent, and purifying by using column chromatography to obtain a yellow solid compound 12a-i without water;

(2) synthesis of 5-bromo-N- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-4-yl-pentyl) pentanamide 15:

181.0mg of 5-bromohexanoic acid 11b was weighed into a 25mL single-neck flask, dissolved by adding 5mL of dichloromethane, slowly added dropwise with 0.5mL of thionyl chloride, refluxed for 4 hours, the solvent was dried by spinning, then added in the order of 5mL of anhydrous tetrahydrofuran, 136.6mg of pomalidomide 14, refluxed for 17 hours, and after TLC confirmed completion of the reaction, spun dry with dichloromethane: purifying the methanol volume ratio of 50:1 mobile phase by a silica gel column to obtain a compound 15; 3) synthesis of ER ligand moieties 18,21a-b,24 with hexanoic acid side chains

Figure FDA0002552570130000061

Reagents and reaction conditions: (a) 6-bromoethyl hexanoate, potassium carbonate and acetone at 50 ℃ for 8 hours; (b)1.22, tetrahydrofuran, 90 ℃,8 hours; 2. lithium hydroxide, 50 ℃, 10 hours; (c) n- (4-methoxyphenyl) -N- (2,2, 2-trifluoroethyl) ethanesulfonamide, tetrahydrofuran, 90 ℃ for 8 hours; 2. lithium hydroxide, 50 ℃, 10 hours;

general procedure for synthesis of Diels-Alder reaction: under the protection of Ar, putting different furan derivatives and proper dienophiles into a round-bottom flask, and then adding anhydrous THF (2mL) to be used as a cosolvent; the reaction was stirred at 90 ℃ for 8 hours, then water was added to quench the reaction and extracted with ethyl acetate; the combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated, followed by purification of the product by silica gel column chromatography;

(1) and 3- (4- ((((5, 6-bis (4-hydroxyphenyl)) -N- (2,2, 2-trifluoroethyl)) -7-oxacyclo [2.2.1] hept-5-ene) -2-sulfonylamino) phenoxy) hexanoic acid compound 18:

the phenolic hydroxyl of the compound 16 and 6-bromoethyl hexanoate are subjected to substitution reaction under the alkaline condition to form a compound 17 containing an ester side chain, and then the compound 17 and 22 are subjected to Diels-Alder reaction, and the obtained DA product is hydrolyzed under the action of lithium hydroxide to obtain a compound 18;

(2) 4- (3-Oxopropoxy) phenyl 5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] hept-5-ene-2-sulfonate analogue 21 a-b:

the phenolic hydroxyl of the compound 19a or 19b and 6-bromoethyl hexanoate are subjected to substitution reaction under the alkaline condition to form a compound 20a or 20b containing an ester side chain, and then the compound 20a or 20b and 22 are subjected to Diels-Alder reaction, and the obtained DA product is hydrolyzed under the action of lithium hydroxide to obtain a compound 21a or 21 b;

(3) synthesis of N- (4-methoxyphenyl) -N- (2,2, 2-trifluoroethyl) methanesulfonamide 3- (4- (3- (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] hept-2-en-2-yl) phenoxy) propionate Compound 24:

one side of phenolic hydroxyl of the compound 22 and 6-bromoethyl hexanoate are subjected to substitution reaction under an alkaline condition to form a compound 23 containing an ester side chain, and then the compound 23 and the dienophile 22 are subjected to Diels-Alder reaction, and the obtained DA product is hydrolyzed under the action of lithium hydroxide to obtain a compound 24;

4) synthesis of target compounds 26 a-j:

Figure FDA0002552570130000071

reagents and reaction conditions: (a) N-ethyl-N- (4-hydroxyphenyl) ethanesulfonamide, potassium carbonate, N, N-dimethylformamide, 85 ℃, 12 hours; (b)22, tetrahydrofuran, 90 ℃ and 8 hours; (c) N-ethyl-N- (4-hydroxyphenyl) ethanesulfonamide, potassium iodide, potassium carbonate, acetonitrile, 80 ℃, 12 hours;

(1) synthesis of (2S,4R) -1- ((S) -2- (6- (4- (N-ethylvinylsulfonamido) phenoxy) hexaamino) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S-1) - (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide analog 25 a-i:

carrying out nucleophilic substitution reaction on the compound 12a-i and the hydroxyl of phenol of an equimolar amount of dienophile under alkaline conditions to obtain a compound 25 a-i;

(2) synthesis of target compounds 26 a-i:

prepared by the Diels-Alder reaction of compounds 25a-i and 22;

(3) synthesis of N- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-4-yl) -3- (4- (N-ethylvinylsulfonamido) phenoxy) propionamide Compound 25 j:

under the protection of argon, 393.2mg of compound 15, 199.2mg of potassium iodide, 207.3mg of potassium carbonate and 272.4mg of N-ethyl-N- (4-hydroxyphenyl) ethanesulfonamide are weighed into a 50mL round-bottom flask, 20mL of anhydrous acetonitrile is added, the mixture is stirred and reacted overnight at 80 ℃, after TLC confirms that the reaction is complete, water is added for quenching, ethyl acetate is added for extraction, an organic phase is dried by anhydrous sodium sulfate, filtered, concentrated and dried, and then the mixture is separated and purified by silica gel column chromatography to obtain a compound 25 j;

(4) synthesis of target compound 26 j:

prepared by the Diels-Alder reaction of compounds 25j and 22;

5) synthesis of target Compound 26 k-r:

reagents and reaction conditions: (a)2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, N, N-dimethylformamide, at 25 ℃ for 4 hours;

general reaction method for condensing amino compound and carboxylic acid compound into amide final product 26 k-r: weighing an amino compound in a two-mouth bottle under the protection of argon, dissolving a small amount of anhydrous N, N-dimethylformamide, sequentially adding a carboxylic acid compound, slowly dropwise adding N, N-diisopropylethylamine, stirring at room temperature for 5 minutes, adding 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea Hexafluorophosphate (HATU), detecting by TLC, adding water for quenching after complete reaction, extracting with ethyl acetate (3X 30mL), washing an organic layer with saturated saline for more than three times to remove a reaction solvent N, N-dimethylformamide, drying the organic layer with anhydrous sodium sulfate, filtering, distilling under reduced pressure to remove the solvent, and purifying by column chromatography to obtain a product 26 k-r.

5. A pharmaceutical composition comprising a compound of claim 1 or 2, one or more pharmaceutically acceptable carriers or excipients.

6. Use of the pharmaceutical composition of claim 5 for the preparation of an anti-breast cancer medicament.

Technical Field

The invention belongs to the technical field of medicines, and relates to a targeted protein hydrolysis chimera using an oxido bicycloheptene compound as an estrogen receptor ligand, a preparation method thereof and application thereof in targeted estrogen receptor treatment of breast cancer.

Background

Breast cancer is the most common cancer in women worldwide, and its high morbidity and mortality severely threatens women's health. Among them, Estrogen Receptor (ER) positive breast cancers account for about 70% of all breast cancers, and endocrine therapy for ER signaling pathway is one of the main means of clinical treatment at present. However, endocrine therapy, for example, which represents the drug tamoxifen having both agonistic and antagonistic activity at estrogen receptors, causes drug resistance in long-term use. Drug resistance in endocrine therapy becomes an urgent problem to be solved in ER positive breast cancer therapy, however, in recent years, development of a PROTAC technology for targeted protein degradation provides a new idea for drug resistance therapy. The key point of the PROTAC technology is that two ends of the molecule respectively recognize a target protein and E3 ubiquitin ligase and form a ternary complex of the target protein-PROTAC-E3. The target protein is then labeled with ubiquitin and eventually degraded via the ubiquitin-proteasome pathway. The strategy of directly degrading the target protein does not depend on the combination with the active pocket of the target protein to play an inhibiting role, so that the drug resistance caused by the point mutation of the existing target protein can be overcome.

Protein degradation does not only depend on the affinity of a binary target protein and a ligand, but also depends on the activity of a triplet, and the activity of the triplet is influenced by the connection conformation and site of the PROTAC, the length and composition modification and concentration of a connecting chain and the like, so that the regulation is more difficult. The problems to be explored by the PROTAC technology at present mainly include optimizing its design, synthesis and evaluation, expanding the range of available E3 ubiquitin ligase, selecting and verifying target points and finally targeting non-drug-forming proteins. The development of PROTAC in the last two decades has seen the spanning progression of the E3 ligase ligand moiety from polypeptide to small molecule with further knowledge of the various E3 ligases. The progress also opens up a new idea for the diversity design of the PROTAC and lays a solid foundation for the clinical druggability of the PROTAC.

Therefore, in order to overcome the drug resistance brought by the traditional endocrine therapy of Estrogen Receptor (ER) positive breast cancer, a novel active compound which has better protein degradation activity and can be subjected to patterned design and synthesis and target degradation of ER needs to be obtained based on the protein targeted degradation PROTAC technology, and the drug resistance brought by the traditional endocrine therapy of ER positive breast cancer is overcome by the method.

Disclosure of Invention

In order to overcome the defects in the prior art, the invention mainly aims to provide a proteolytic targeting chimeric compound taking an oxido bicycloheptene compound as an estrogen receptor ligand. In the previous research of the research team of the inventor, the OBHSA compound (shown in formulas 1 and 2) with an oxygen bridge bicyclo- [2.2.1] -heptene sulfonamide structure and a special three-dimensional stereo structure is found to have certain ER alpha down-regulation activity. More than 600E 3 ligases are currently found, which express differently in different tissues and cells, and this application focuses on CRBN and VHL ligases possessing specific small molecule ligands in combination with the PROTAC analysis of a few targeted ER proteins found in previous studies by research teams. Therefore, based on the advantage skeleton of the OBHSA targeting ER discovered by research team for many years, two E3 ligase ligands are selected as another component of the ternary complex, and the two components are connected by simple alkyl side chains to synthesize the target compound.

The second purpose of the invention is to provide a preparation method of the proteolytic targeting chimeric compound, which utilizes different routes to efficiently obtain the target compound in the synthesis process, thereby summarizing a synthesis route with strong applicability and high yield.

The third purpose of the invention is to provide the application of the proteolytic targeting chimeric compound. In contrast to the use of estradiol or selective estrogen receptor modulators as the ER ligand moiety reported in the prior literature, the resulting compounds of the present invention demonstrate for the first time the positive effect of the proteolytic targeting chimera (Protac) molecule designed to have estrogen receptor down-modulators as the ER ligand moiety on the ability of the ER to degrade. The novel active compound which has better protein degradation activity and can be subjected to patterned design and synthesis and target degradation of ER is obtained, and the method is expected to overcome the drug resistance brought by the traditional endocrine therapy of ER positive breast cancer.

In order to achieve the purpose, the invention adopts the following technical scheme:

in a first aspect, a proteolytic targeting chimeric compound using an oxido bicycloheptene compound as an estrogen receptor ligand is provided, which has the following general structure:

preferably, the proteolytic targeting chimeric compound taking the oxido-bicycloheptene compound as the estrogen receptor ligand is selected from any one of the following compounds:

(2S,2R) -1- ((2S) -2- (6- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) hexanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- (4- (4-methylthiazol-5-yl) benzyl) pyrrolidine-2-carboxamide (26b),

(2S,2R) -1- ((2S) -2- (7- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) heptanamide) -3, 3-dimethylbutanoyl) -4-hydroxy-N- (4- (4-methylthiazol-5-yl) benzyl) pyrrolidine-2-carboxamide (26c),

(2S,2R) -1- ((2S) -2- (5- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) pentanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1- (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26g),

(2S,2R) -1- ((2S) -2- (6- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) hexanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1- (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26h),

(2S,2R) -1- ((2S) -2- (7- (4- ((N-ethyl-5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonylamino) phenoxy) heptanamide) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1- (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26i),

(2S,2R) -1- ((2S) -2- (6- (4- ((5, 6-bis (4-hydroxyphenyl) -N- (2,2, 2-trifluoroethyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonamide) phenoxy) hexanamino) -3, 3-dimethylbutyryl) -4-hydroxy-N- (4- (4-methylthiazol-5-yl) benzyl) pyrrolidine-2-carboxamide (26k),

(2S,2R) -1- ((2S) -2- (6- (4- ((5, 6-bis (4-hydroxyphenyl) -N- (2,2, 2-trifluoroethyl) -7-oxabicyclo [2.2.1] -5-heptene) -2-sulfonamide) phenoxy) hexanamino) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S) -1-4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide (26l),

2-chloro-4- ((6- (((S) -1- ((2S,4R) -4-hydroxy-2- (((4- (4-methylthiazol-5-yl) benzyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxybutan-2-yl) amino) -6-oxyhexyl) oxy) -5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene-2-sulfonate (26m),

2-chloro-4- ((6- (((S) -1- ((2S,4R) -4-hydroxy-2- (((4- (4-methylthiazol-5-yl) phenyl) ethyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxybutan-2-yl) amino) -6-oxyhexyl) oxy) -5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] -5-heptene-2-sulfonate (26 n).

In a second aspect, the application of the proteolytic targeting chimeric compound taking the oxido-bicycloheptene compound as an estrogen receptor ligand in preparing anti-breast cancer drugs is provided.

In a third aspect, a method for preparing the proteolytic targeting chimeric compound using the oxido-bicycloheptene compound as the estrogen receptor ligand is provided, wherein the Boc-tert-leucine hydroxyproline thiazole derivative 9, the E3 ligands 12a-i and 15 with long side chains and the ER ligand part 18,21a-b and 24 with caproic acid side chains are synthesized by the following reactions, and the method specifically comprises the following steps:

1) tert-butyl ((S) -1- ((2S,4R) -4-hydroxy-2- ((4- (4-methylthiazol-5-yl) benzyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxobutan-2-yl) carbamate 9a and tert-butyl ((S) -1- ((2S, synthesis of 4R) -4-hydroxy-2- (((S) -1- (4- (4-methylthiazol-5-yl) phenethyl) carbamoyl) pyrrol-idin-1-yl) -3, 3-dimethyl-1-oxybutynin-2-yl) carbamate 9 b:

reagents and reaction conditions: (a) di-tert-butyl dicarbonate, dichloromethane and triethylamine are carried out for 12 hours at 25 ℃; (b) dimethyl acetamide, methyl acetate, dimethyl acetamide, reflux at 150 deg.c and reaction overnight; (c) trifluoroacetic acid, dichloromethane, 25 ℃, 0.5 hours; (d)2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, Boc-L-hydroxyproline, N, N-dimethylformamide, at 25 ℃ for 1 hour; (e) trifluoroacetic acid, dichloromethane, 25 ℃, 0.5 hour; 2.2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, Boc-L-tert-leucine, N, N-dimethylformamide, at 25 deg.C for 0.5 h.

(1) Synthesis of tert-butyl (4-bromobenzyl) carbamate 4a and (R) - (1- (4-bromophenylethyl) carbamate 4 b:

the compound 4-bromobenzylamine 3a (1.99g, 10.75mmol) or its analogue (R) -1- (4-bromophenyl) ethan-1-amine 3b (2.14g, 10.75mmol) was weighed out and placed in two 50mL double-neck bottles, and after 10 minutes in ice bath, 10mL of dichloromethane and triethylamine (2.26mL, 16.13mmol) were added in sequence, and after 10 minutes in ice bath, a dichloromethane (10mL) solution containing di-tert-butyl dicarbonate (2.82g, 12.9mmol) was slowly added, and after 5 minutes addition, the reaction was carried out overnight at room temperature. After TLC confirms that the reaction is complete, water is added for quenching, dichloromethane is used for extraction, an organic phase is dried by anhydrous sodium sulfate, filtered, concentrated and dried, and then white solid compound 4a (the yield is 97%) or 4b (the yield is 93%) is obtained through silica gel column chromatography separation and purification;

4a:1H NMR(400MHz,CDCl3)7.43(d,J=6.9Hz,2H),7.14(d,J=7.6Hz,2H), 4.93(s,1H),4.24(d,J=5.2Hz,2H),1.44(s,9H).

4b:1H NMR(400MHz,CDCl3)7.52(d,J=8.1Hz,2H),7.33(d,J=8.2Hz,2H), 4.94(s,1H),4.70(q,J=6.5Hz,1H),1.45(s,9H),1.36(d,J=6.6Hz,3H).

(2) synthesis of tert-butyl (4- (4-methylthiazol-5-yl) benzyl) carbamate 6a and (R) - (1- (4- (4- (4-methylthiazol-5-yl) phenyl) ethyl) carbamate tert-butyl 6 b:

under the protection of argon, 4a (3.04g, 10.0mmol) or 4b (3.18g, 10.0mmol), 4-methylthiazole (1.98g, 20.0mmol), palladium acetate (225mg, 1.0mmol) and potassium acetate (3.93g, 40.0mmol) were weighed respectively into a 100mL round-bottomed flask, and 25mL of solvent N, N-dimethylacetamide was added to react at 150 ℃ overnight. TLC confirmed the reaction was complete, quenched with water, extracted with dichloromethane, washed the organic phase with water, dried over anhydrous sodium sulfate, filtered, concentrated and spun-dried, and purified by silica gel column chromatography to afford compound 6a (93% yield) or 6b (95% yield) as a white solid.

6a:1H NMR(400MHz,CDCl3)8.63(s,1H),7.37(d,J=8.1Hz,2H),7.31(d,J=8.0Hz,2H),5.17(s,1H),4.32(d,J=5.7Hz,2H),2.49(s,3H),1.44(s,9H).

6b:1H NMR(400MHz,CDCl3)8.59(s,1H),7.51(d,J=8.1Hz,2H),7.43(d,J=8.2Hz,2H),4.94(s,1H),4.70(q,J=6.5Hz,1H),2.48(s,3H),1.45(s,9H),1.33(d, J=6.6Hz,3H).

The Boc removal reaction is carried out by placing the compound in a single-neck flask with a suitable volume, adding dichloromethane to dissolve the compound, dropwise adding trifluoroacetic acid with the same volume as that of dichloromethane under ice bath, and reacting at the temperature for 1 hour. After spin-drying, dichloromethane is added for several times to spin-dry and remove trifluoroacetic acid.

General procedure for the condensation of amino compounds and carboxylic acid compounds to amides: weighing an amino compound in a two-mouth bottle under the protection of argon, dissolving a small amount of anhydrous N, N-dimethylformamide, sequentially adding a carboxylic acid compound, slowly dropwise adding N, N-diisopropylethylamine, stirring at room temperature for 5 minutes, and then adding 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea Hexafluorophosphate (HATU). And (3) after TLC detection reaction is completed, adding water for quenching, extracting by ethyl acetate (3X 30mL), washing an organic layer by saturated salt water for more than three times to remove a reaction solvent N, N-dimethylformamide, drying the organic layer by anhydrous sodium sulfate, distilling under reduced pressure to remove the solvent, and purifying by column chromatography to obtain the product.

(3) And after the compound 6a or 6b is subjected to Boc removal reaction to obtain a compound 7a or 7b, the amino part of the compound is condensed with carboxyl in Boc-L-hydroxyproline to obtain a compound 8a or 8 b. Subsequently, after a further Boc removal reaction, condensation reaction with N-Boc-L-tert-leucine gave compound 9a (58% yield) or 9b (55% yield) as a yellow solid.

9a:1H NMR(400MHz,MeOD)8.87(s,1H),7.45(d,J=8.1Hz,2H),7.40(d,J=8.2Hz,2H),4.60(t,J=8.3Hz,1H),4.53(d,J=15.5Hz,2H),4.35(d,J=15.5Hz, 1H),4.30(s,1H),3.92–3.74(m,2H),2.47(s,3H),2.28–2.17(m,1H),2.12–2.05 (m,1H),1.43(s,9H),1.01(s,9H).

9b:1H NMR(400MHz,CDCl3)8.66(s,1H),7.36(s,4H),5.09–5.02(m,1H), 4.75(t,J=7.9Hz,1H),4.43(s,1H),3.74(d,J=11.0Hz,1H),3.58(d,J=8.3Hz, 2H),2.50(s,3H),2.24(s,1H),2.16–2.09(m,1H),1.48(d,J=6.8Hz,3H),1.35(s, 9H),1.00(s,9H).

2) Synthesis of E3 ligands 12a-i and 15 with long side chains:

reagents and reaction conditions: (a) trifluoroacetic acid, dichloromethane, 25 ℃, 0.5 hours; (b)2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, N, N-dimethylformamide, at 25 deg.C for 0.5 hr; (c) refluxing thionyl chloride and dichloromethane for 4 hours; (d) tetrahydrofuran, reflux, overnight.

(1) Synthesis of 6- ((1- ((2S,4R) -4-hydroxy-2- ((4- (4-methylthiazol-5-yl) benzyl) carbamoyl) pyrrolidin-1-yl) -3, 3-dimethyl-1-oxybutan-2-yl) amino) hexanoic acid analog 12:

compound 9a or 9b (1.0equiv.) was weighed out and dissolved in a mixed solution of TFA and DCM ═ 1:1, and after 30min of reaction, the solvent was distilled off under reduced pressure to give deprotected intermediate compound 10a or 10 b. Then, under the protection of argon, taking the intermediate compound 10a or 10b, dissolving the intermediate compound in a 25mL two-necked bottle by using anhydrous DMF, adding bromo alkyl acids 11a-f (1.0equiv.) with different lengths, dropwise adding N, N-diisopropylethylamine (8.0equiv.), stirring at room temperature for 5min, adding 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (1.1equiv.), reacting at room temperature for 30min, adding water for quenching, extracting by using ethyl acetate, washing an organic layer by using saturated common salt water, drying by using anhydrous sodium sulfate, distilling under reduced pressure to remove a solvent, and purifying by using column chromatography to obtain a yellow solid compound 12a-i (the yield is 54% -63%).

12b:1H NMR(400MHz,MeOD)8.87(s,1H),7.45(d,J=8.2Hz,2H),7.38(d,J=8.1Hz,2H),4.63(dd,J=8.0,4.9Hz,1H),4.59(d,J=8.5Hz,1H),4.53(d,J=15.7 Hz,2H),4.36(d,J=15.5Hz,1H),3.92(d,J=11.0Hz,1H),3.80(dd,J=10.9,3.6 Hz,1H),3.41(t,J=6.7Hz,2H),2.46(s,3H),2.36–2.28(m,1H),2.29–2.19(m, 2H),2.13–2.04(m,1H),1.88–1.78(m,2H),1.67–1.56(m,2H),1.50–1.40(m, 2H),1.04(s,9H).

12c:1H NMR(400MHz,MeOD)8.86(s,1H),7.45(d,J=8.1Hz,2H),7.38(d,J=8.1Hz,2H),4.64(d,J=3.9Hz,1H),4.60(d,J=8.0Hz,1H),4.53(d,J=15.7Hz, 2H),4.36(d,J=15.6Hz,1H),3.93(d,J=11.0Hz,1H),3.80(dd,J=10.9,3.5Hz, 1H),3.40(t,J=6.7Hz,2H),2.45(s,3H),2.36–2.28(m,1H),2.27–2.18(m,2H), 2.13–2.04(m,1H),1.85–1.75(m,2H),1.67–1.54(m,2H),1.47–1.38(m,2H), 1.33(dd,J=14.7,7.2Hz,2H),1.04(s,9H).

12d:1H NMR(400MHz,MeOD)8.88(s,1H),7.46(d,J=7.8Hz,2H),7.40(d,J=8.0Hz,2H),4.70–4.63(m,1H),4.63–4.54(m,1H),4.50(t,J=15.9Hz,2H),4.37 (dd,J=15.4,4.7Hz,1H),3.92(d,J=10.9Hz,1H),3.81(dd,J=10.7,3.5Hz,1H), 3.42(t,J=6.7Hz,2H),2.46(s,3H),2.36–2.29(m,1H),2.27–2.18(m,2H),2.13– 2.02(m,1H),1.84–1.77(m,2H),1.65–1.56(m,2H),1.44–1.38(m,2H),1.34– 1.29(m,4H),1.04(s,9H).

12e:1H NMR(400MHz,MeOD)8.86(s,1H),7.44(d,J=7.1Hz,2H),7.37(d,J=6.9Hz,2H),4.64(s,1H),4.60(d,J=8.3Hz,1H),4.53(d,J=16.0Hz,2H),4.36(d, J=15.4Hz,1H),3.93(d,J=10.4Hz,1H),3.81(d,J=8.4Hz,1H),3.39(t,J=6.0 Hz,2H),2.45(s,3H),2.33–2.27(m,1H),2.26–2.18(m,2H),2.13–2.05(m,1H), 1.84–1.74(m,2H),1.62–1.53(m,2H),1.37(s,2H),1.29(s,6H),1.04(s,9H).

12f:1H NMR(400MHz,MeOD)8.87(s,1H),7.45(d,J=8.0Hz,2H),7.37(d,J=8.0Hz,2H),4.65(s,1H),4.61(d,J=8.0Hz,1H),4.54(d,J=15.9Hz,2H),4.36(d, J=15.5Hz,1H),3.93(d,J=10.8Hz,1H),3.85–3.76(m,1H),3.39(t,J=6.7Hz, 2H),2.45(s,3H),2.35–2.27(m,1H),2.27–2.18(m,2H),2.14–2.03(m,1H),1.85 –1.73(m,2H),1.64–1.53(m,2H),1.42–1.36(m,2H),1.28(s,8H),1.04(s,9H).

(2) Synthesis of 5-bromo-N- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-4-yl-pentyl) pentanamide 15:

5-Bromohexanoic acid 11b (181.0mg, 1.0mmol) was weighed into a 25mL single-neck flask, dissolved by adding 5mL of dichloromethane, thionyl chloride (0.5mL, excess) was slowly added dropwise, after refluxing for 4 hours, the solvent was spun dry, then anhydrous tetrahydrofuran 5mL, pomalidomide 14(136.6mg, 0.5 mmol) were added in that order, and refluxed for 17 hours. TLC confirmed the reaction was complete, dried and purified through silica gel column using mobile phase ratio (dichloromethane: methanol 50:1) to give compound 15 (74% yield).

1H NMR(400MHz,DMSO-d6)11.18(s,1H),9.73(s,1H),8.47(t,J=7.0Hz,1H),7.83(t,J=7.9Hz,1H),7.62(d,J=7.3Hz,1H),5.16(dd,J=12.7,5.3Hz,1H),3.54 (t,J=6.7Hz,2H),2.68–2.55(m,2H),2.50(q,J=7.5Hz,4H),1.89–1.79(m,2H), 1.70–1.60(m,2H),1.50–1.40(m,2H).

3) Synthesis of ER ligand moieties 18,21a-b,24 with hexanoic acid side chains

Reagents and reaction conditions: (a) 6-bromoethyl hexanoate, potassium carbonate and acetone at 50 ℃ for 8 hours; (b)1.22, tetrahydrofuran, 90 ℃,8 hours; 2. lithium hydroxide, 50 ℃, 10 hours; (c) n- (4-methoxyphenyl) -N- (2,2, 2-trifluoroethyl) ethanesulfonamide, tetrahydrofuran, 90 ℃ for 8 hours; 2. lithium hydroxide, 50 ℃, 10 hours

General procedure for synthesis of Diels-Alder reaction: under the protection of Ar, a solution with various furan derivatives and appropriate dienophiles was placed in a round-bottom flask, and then anhydrous THF (2mL) was added as a co-solvent. The reaction was stirred at 90 ℃ for 8 hours under heating, then water was added to quench the reaction, and extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated, followed by purification of the product by silica gel column chromatography.

(1) And 3- (4- ((((5, 6-bis (4-hydroxyphenyl)) -N- (2,2, 2-trifluoroethyl)) -7-oxacyclo [2.2.1] hept-5-ene) -2-sulfonylamino) phenoxy) hexanoic acid compound 18:

the phenolic hydroxyl group of the compound 16 and 6-bromoethyl hexanoate undergo substitution reaction under alkaline conditions to form a compound 17 containing an ester side chain, and then undergo Diels-Alder reaction with 22, and the obtained DA product is hydrolyzed under the action of lithium hydroxide to obtain a compound 18 (the yield is 84%).

(2) 4- (3-Oxopropoxy) phenyl 5, 6-bis (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] hept-5-ene-2-sulfonate analogue 21 a-b:

the phenolic hydroxyl group of the compound 19a or 19b and 6-bromoethyl hexanoate are subjected to substitution reaction under alkaline conditions to obtain a compound 20a or 20b containing an ester side chain, and then are subjected to Diels-Alder reaction with 22, and the obtained DA product is hydrolyzed under the action of lithium hydroxide to obtain a compound 21a (the yield is 73%) or 21b (the yield is 77%).

(3) Synthesis of N- (4-methoxyphenyl) -N- (2,2, 2-trifluoroethyl) methanesulfonamide 3- (4- (3- (4-hydroxyphenyl) -7-oxabicyclo [2.2.1] hept-2-en-2-yl) phenoxy) propionate Compound 24:

one side of phenolic hydroxyl of the compound 22 and 6-bromoethyl hexanoate are subjected to substitution reaction under alkaline conditions to obtain a compound 23 containing an ester side chain, and then the compound is subjected to Diels-Alder reaction with a dienophile 22, and the obtained DA product is hydrolyzed under the action of lithium hydroxide to obtain a compound 24 (the yield is 78%).

4) Synthesis of target compounds 26 a-j:

Figure RE-GDA0002612956640000111

reagents and reaction conditions: (a) N-ethyl-N- (4-hydroxyphenyl) ethanesulfonamide, potassium carbonate, N, N-dimethylformamide, 85 ℃, 12 hours; (b)22, tetrahydrofuran, 90 ℃ and 8 hours; (c) N-ethyl-N- (4-hydroxyphenyl) ethanesulfonamide, potassium iodide, potassium carbonate, acetonitrile, 80 ℃ for 12 hours

(1) Synthesis of (2S,4R) -1- ((S) -2- (6- (4- (N-ethylvinylsulfonamido) phenoxy) hexaamino) -3, 3-dimethylbutyryl) -4-hydroxy-N- ((S-1) - (4- (4-methylthiazol-5-yl) phenyl) ethyl) pyrrolidine-2-carboxamide analog 25 a-i:

nucleophilic substitution of compound 12a-i with an equimolar amount of the dienophile phenol hydroxyl under basic conditions yields compound 25a-i (41-63% yield).

(2) Synthesis of target compounds 26 a-i:

prepared by Diels-Alder reaction of compounds 25a-i and 22 (61-72% yield).

(3) Synthesis of N- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-4-yl) -3- (4- (N-ethylvinylsulfonamido) phenoxy) propionamide Compound 25 j:

compound 15(393.2mg,1.0mmol), potassium iodide (199.2mg, 1.2mmol), potassium carbonate (207.3mg,1.5mmol), N-ethyl-N- (4-hydroxyphenyl) ethanesulfonamide (272.4mg,1.2mmol) were weighed into a 50mL round-bottomed flask under argon protection, 20mL of anhydrous acetonitrile was added, and the reaction was stirred at 80 ℃ overnight. After TLC confirmed the reaction was complete, water was added and quenched, extracted with ethyl acetate, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated and dried, and purified by silica gel column chromatography to give compound 25j (30% yield).1H NMR(400MHz,Acetone-d6)10.02(s,1H),9.53(s,1H),8.82(d,J=8.5 Hz,1H),7.87–7.82(m,1H),7.58(d,J=7.2Hz,1H),7.27–7.22(m,2H),6.97– 6.93(m,2H),6.77(dd,J=16.5,9.9Hz,1H),6.05(d,J=1.6Hz,1H),6.02(d,J= 4.9Hz,1H),5.18(dd,J=12.7,5.5Hz,1H),4.05(t,J=6.4Hz,2H),3.61(q,J=7.1 Hz,2H),2.83–2.73(m,4H),2.63(t,J=7.4Hz,2H),1.92–1.80(m,2H),1.68– 1.58(m,2H),1.31(s,2H),1.07(t,J=7.1Hz,3H).

(4) Synthesis of target compound 26 j:

prepared by the Diels-Alder reaction of compounds 25j and 22 (67% yield).

5) Synthesis of target Compound 26 k-r:

reagents and reaction conditions: (a)2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, N, N-diisopropylethylamine, N, N-dimethylformamide, at 25 deg.C for 4 hours.

General reaction method for condensing amino compound and carboxylic acid compound into amide final product 26 k-r: weighing an amino compound in a two-mouth bottle under the protection of argon, dissolving a small amount of anhydrous N, N-dimethylformamide, sequentially adding a carboxylic acid compound, slowly dropwise adding N, N-diisopropylethylamine, stirring at room temperature for 5 minutes, and then adding 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea Hexafluorophosphate (HATU). And (3) after TLC detection reaction is completed, adding water for quenching, extracting by ethyl acetate (3X 30mL), washing an organic layer by saturated salt water for more than three times to remove a reaction solvent N, N-dimethylformamide, drying the organic layer by anhydrous sodium sulfate, filtering, distilling under reduced pressure to remove the solvent, and purifying by column chromatography to obtain a product 26 k-r.

In a fourth aspect, a pharmaceutical composition is provided, which comprises the proteolytic targeting chimera compound with the oxido-bicycloheptene compound as the estrogen receptor ligand, and one or more pharmaceutically acceptable carriers or excipients.

In a fifth aspect, an application of the pharmaceutical composition in preparing an anti-breast cancer medicament is provided.

The invention provides a method for synthesizing a proteolysis targeting chimera compound by taking an oxido bicycloheptene compound as an estrogen receptor ligand, which adopts two reasonable synthesis modes, takes a VHL ligand or a CRBN ligand as an E3 ligase ligand part, and connects an oxido bicycloheptene sulfonate or a sulfonamide estrogen receptor ligand through alkyl side chains with different lengths to synthesize a series of target product Protac molecules. The Protac molecules have different action modes from the existing anti-breast cancer medicament tamoxifen (4-OHT), and are targeted estrogen receptor down-regulators. Such compounds not only retain some ability to bind to estrogen receptors, but also have good estrogen receptor alpha downregulation activity comparable to fulvestrant (Ful), e.g. 26b is comparable to fulvestrant degradation activity at 1 μ M. Among them, most compounds have obvious down-regulation activity to ER alpha, such as degradation effect of 26h,26i,26k,26l and 26m is quite good. Different from the traditional estrogen receptor down-regulating agent, the compound can realize the degradation of an event-driven targeted estrogen receptor, and is expected to overcome the drug resistance brought by the traditional endocrine therapy of ER positive breast cancer by the method. In addition, the method introduces the estrogen receptor down-regulator as an ER ligand for the first time, and compared with a compound taking an estrogen receptor modulator OBHS compound as an ER ligand, the introduction of the OBHSA estrogen receptor down-regulator has a certain promotion effect on the degradation of the obtained Protac molecule to ER.

Drawings

FIG. 1 comparison of ER α degradation in MCF-7 cells treated with Compounds 26a-26i

A) Western blot analysis of ER α in MCF-7 cells treated with 1 μ M of compound Ful,26a-26 i;

B) comparison of the level of ER α degradation by Compounds 26a-26 i;

FIG. 2 comparison of ER α degradation in MCF-7 cells treated with Compounds 26j-26r

A) Western blot analysis of ER α in MCF-7 cells treated with 1 μ M of compound 4-OHT, 26j-26 r;

B) comparison of the level of ER α degradation by Compounds 26j-26 r;

Detailed Description

The features and advantages of the present invention will be further understood from the following detailed description taken in conjunction with the accompanying drawings. The examples provided are merely illustrative of the method of the present invention and do not limit the remainder of the disclosure in any way.

35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:纽甜中粘性物质的去除方法

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