CN118027003A - PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent - Google Patents

PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent Download PDF

Info

Publication number
CN118027003A
CN118027003A CN202410152143.8A CN202410152143A CN118027003A CN 118027003 A CN118027003 A CN 118027003A CN 202410152143 A CN202410152143 A CN 202410152143A CN 118027003 A CN118027003 A CN 118027003A
Authority
CN
China
Prior art keywords
compound
hours
organic solvent
thalidomide
formula
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.)
Pending
Application number
CN202410152143.8A
Other languages
Chinese (zh)
Inventor
杨学谦
范培红
娄红祥
郑家晴
董旭
张建勇
刘城
孔令金
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.)
Shandong University
Shandong Qidu Pharmaceutical Co Ltd
Original Assignee
Shandong University
Shandong Qidu Pharmaceutical Co Ltd
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 Shandong University, Shandong Qidu Pharmaceutical Co Ltd filed Critical Shandong University
Priority to CN202410152143.8A priority Critical patent/CN118027003A/en
Publication of CN118027003A publication Critical patent/CN118027003A/en
Pending legal-status Critical Current

Links

Landscapes

  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses PROTAC compounds, a preparation method and application of the compounds as estrogen receptor degradation agents. The PROTAC compounds comprise a pharmaceutically acceptable salt, hydrate, solvate, polymorph or prodrug, or an estrogen receptor proteolytically targeted chimeric derivative thereof. The compound is prepared by linking 8- (2, 4-dichlorophenyl) -9- (4- (4-formylpiperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylic acid methyl ester and derivatives thereof with E3 ligase ligand consisting of lenalidomide, thalidomide, hydroxy thalidomide, pomalidomide or 4-amino-4-oxo-2- (1-oxo-isoindol-2-yl) butyric acid or derivatives and isomers thereof. In order to find novel estrogen receptor degrading agents, a series of proteolysis targeting chimera compounds are developed. The compound has good degradation effect on estrogen receptors, and can be developed into novel estrogen receptor degradation agent medicines.

Description

PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent
Technical Field
The invention relates to the technical field of medicine synthesis and application, in particular to PROTAC compounds, a preparation method and application of the compounds as estrogen receptor degrading agents.
Background
Breast cancer is one of the most common forms of cancer at present. According to the National Cancer Institute (NCI), it was estimated that about 25 ten thousand women were diagnosed with breast cancer in 2017, with more than 4 ten thousand patients being present. ER+/HER 2-breast cancer accounts for about 70% of breast cancer patients. Such patients may be treated by endocrine disruption therapy in the early stages, but as the disease progresses, the tumor develops resistance to this therapy and many mutations accumulate, causing therapeutic difficulties.
In recent years, the incidence of breast cancer is on the rise, and is now the leading one among female malignant tumors. Of breast cancer patients, about 70% appear to be estrogen receptor ER positive breast cancer. ER is therefore an important target in breast cancer drug research. To date, the primary intervention in the treatment of ER-positive breast cancer has remained by endocrine therapy inhibiting estrogen levels associated with breast cancer, mainly including selective estrogen receptor modulators (e.g. tamoxifen, raloxifene) and aromatase inhibitors (e.g. letrozole, exemestane) as well as selective ER downregulators. Although endocrine therapy achieves great results in treating breast cancer patients, it has problems of toxic and side effects, long treatment time, drug resistance, and the like.
The technology of proteolysis targeting chimera PROTAC is proposed by Craig m.crews professor team, currently on the leading edge, targeting therapeutic strategy PROTACs is a hybrid bifunctional molecule, one end of which is a ligand capable of binding to E3 ligase, the other end of which is a ligand binding to target protein (protein of interest, POI), and the middle of which is a linker. The action mechanism is that the target protein is recruited to the vicinity of E3 ligase through PROTACs molecules, so that the target protein is marked by ubiquitination, and then the target protein is degraded through ubiquitin-proteasome pathway, and finally the purpose of treating diseases is achieved. The ER in the breast cancer cells is degraded to play a role in treatment, so that the drug resistance can be effectively overcome, and the drug is a promising anti-breast cancer drug.
CN116178340a discloses a PROTAC compound. CN115124590a discloses PROTAC class compounds targeted to degrade FLT3-ITD muteins. CN114736264a discloses Tau protein visualization PROTAC degrading compounds. CN113336748a discloses protein degradation targeting chimeras (PROTAC). CN109152843a discloses PROTAC antibody conjugates. CN110612297a discloses estrogen receptor proteolytic modulators. CN110291087a discloses tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degrading agents.
Disclosure of Invention
The invention aims to find novel estrogen receptor degradation agent medicines, and designs a series of proteolysis targeting chimeric compounds. Accordingly, the present invention provides estrogen receptor proteolytically targeted chimeras (compound I) having the general formula (I).
According to a first embodiment of the present invention there is provided a compound having the general formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt, hydrate, solvate, polymorph or prodrug thereof, or an estrogen receptor proteolytically targeted chimeric derivative thereof:
In formula (I), R is one of C1-C5 alkoxy (such as methoxy, ethoxy, propoxy, butoxy), amino, C1-C5 alkylamino (such as methylamino, ethylamino, propylamino, butylamino), or OH. That is, R in structural formula (I) is CH3 (CH 2) pO-, p=0, 1,2,3,4 or 5; or an amino group; or CH3 (CH 2) qNH-, q=one of 0, 1,2,3,4 or 5, or-OH.
The E3 ligase ligand in the formula (I) has a structure of a general formula (II); preferably, the E3 ligase ligand is lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindol-1, 3-dione, or derivatives and isomers thereof:
n=0 or 1, m=0 or 1;
Wherein the linker site is one of R1, R2 and R3, and the remaining substituents (i.e., the other two of R1, R2 and R3) are each independently: one of H, OH, F, cl, br, NH2, CH3 or OCH 3. And/or, in addition, one E3 ligase ligand when n=0 or 1, m=0 or 1, R4 is hydrogen or methyl in the structural formula (II) may also be one of thalidomide or its variant 4-amino-4-oxo-2- (1-oxo-isoindol-2-yl) butyric acid.
The compounds of formula (I) are estrogen receptor proteolytically targeted chimeras (I).
Preferably, the E3 ligase ligand in formula (I) is lenalidomide (IIa):
the site (x) or manner of linkage of lenalidomide (IIa) to linker is, or the E3 ligase ligand in formula (I) is, for example, one of the following:
(wherein represents the site of attachment. The following is the same).
The NH-group in formula (IIa-1) may also be part of a linker group or a terminal group (-NH-), as follows. Namely, the following formula:
wherein-L' -NH-is (as divalent linking group) -Linker-or-L-. The following is the same.
Alternatively, the E3 ligase ligand in formula (I) is thalidomide (IIb):
The site (x) or manner of linking of thalidomide (IIb) to linker is, or the E3 ligase ligand in formula (I) is, for example, one of the following:
Or, the E3 ligase ligand in formula (I) is 4-hydroxy-thalidomide (IIc-1) or 5-hydroxy-thalidomide (IIc-2):
the site (x) or manner of linkage of the 4-hydroxy-thalidomide (IIc-1) to the linker is, or the E3 ligase ligand of formula (I) is, for example, one of the following:
the site (x) or manner of linkage of the 5-hydroxy-thalidomide (IIc-2) to the linker is, or the E3 ligase ligand of formula (I) is, for example, one of the following:
The O-group may also be part of a linker group or a terminal group (-O-), as follows. Namely, the following formula:
wherein-L' -O-is (as divalent linking group) -Linker-or-L-. The following is the same.
Alternatively, the E3 ligase ligand in formula (I) is pomalidomide (IId):
The site (x) or manner of attachment of pomalidomide (IId-1) to linker is, or the E3 ligase ligand in formula (I) is, for example, one of the following:
The NH-group may also be part of a linker group or a terminal group (-NH-), as follows. Namely, the following formula:
wherein-L' -NH-is (as divalent linking group) -Linker-or-L-. The following is the same.
The site (x) or manner of linking of pomalidomide (IId-2) to linker is, for example, one of the following:
The NH-group may also be part of a linker group or a terminal group (-NH-), as follows. Namely, the following formula:
wherein-L' -NH-is (as divalent linking group) -Linker-or-L-. The following is the same.
Or, the E3 ligase ligand of formula (I) is 3- (1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (IIe):
The site (x) or manner of linkage of the 3- (1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (IIe) to the linker is, or the E3 ligase ligand in formula (I) is, for example, one of the following:
Or, the E3 ligase ligand of formula (I) is 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butanoic acid (IIf):
The site (x) or manner of linkage of 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid (IIf) to linker is, or the E3 ligase ligand in formula (I) is, for example, one of the following:
Or, the E3 ligase ligand of formula (I) is 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione (IIg):
The site (x) or mode of linkage of the 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione (IIg) to the linker is, or the E3 ligase ligand in formula (I) is, for example, one of the following:
Linker (or L) in formula (I) is a (divalent) linking group or bridging group. Typically, a linker is one or a combination of two or more of a flexible or rigid linker or bridging group.
Preferably, the rigid linker is one selected from the following formulae:
The flexible linker is one selected from the following formulas:
the combination of the flexible linker and the rigid linker is one selected from the following formulas:
In the present application, n=0 to 5 represents n=0, 1, 2, 3, 4 or 5. Similarly, n=2-10 represents n=2, 3, 4, 5, 6, 7, 8, 9 or 10.
According to a second embodiment of the present invention, there is provided a method for preparing the estrogen receptor proteolytically targeted chimera (I), the method comprising: dissolving compound 5 in an organic solvent (e.g., N dimethylformamide, or dimethylsulfoxide), adding an E3 ligase ligand (preferably, lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid, or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione or a derivative thereof), (stirring at normal temperature (e.g., 0.5 to 3 hours, such as 1 hour), then adding a reducing agent (e.g., an alkali metal borohydride such as sodium borohydride, potassium borohydride, lithium borohydride, or sodium triacetylborohydride), (continuing stirring at normal temperature), and after completion of the reaction, isolating and purifying (e.g., extraction purification) to obtain the target compound (I), wherein compound 5 has the following structural formula:
Wherein: r is one of C1-C5 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), amino, C1-C5 alkylamino (e.g., methylamino, ethylamino, propylamino, butylamino), or OH; for example methyl 8- (2, 4-dichlorophenyl) -9- (4- (4-formylpiperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate.
More specifically, the preparation method of the estrogen receptor proteolytic targeting chimera (I) (namely the compound (I)) comprises the following steps:
preferably, the method for preparing the estrogen receptor proteolytic targeting chimera (I) (i.e. compound (I)) comprises:
step 1: allowing compound 1 to react with (4-bromophenyl) boronic acid In organic solvent in the presence of catalyst, separating to obtain compound 2,
Wherein compound 1 and compound 2 each have the following structural formula:
Wherein: r is one of C1-C5 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), amino, C1-C5 alkylamino (e.g., methylamino, ethylamino, propylamino, butylamino), or OH.
Step 2: reacting the compound 2 with 4- (diethoxymethyl) piperidine in an organic solvent in the presence of a catalyst, and separating to obtain a compound 3:
step 3: reacting the compound 3 with pyridinium tribromide in an organic solvent in the presence of a catalyst, and separating to obtain a compound 4:
step 4: reacting the compound 4 with (2, 4-dichlorophenyl) boric acid in an organic solvent in the presence of a catalyst, and separating to obtain a compound 5:
Step 5: dissolving lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxo-isoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxo-isoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione with an organic solvent (e.g., dimethyl sulfoxide, or N, N dimethylformamide), adding a linker and a tertiary amine compound (e.g., a tertiary amine containing an isopropyl group such as N, N-diisopropylethylamine), reacting (e.g., 4-12 hours or 5-10 hours such as 6-8 hours) at an elevated temperature (e.g., 80-130 ℃ or 90-110 ℃ such as 100 ℃), separating and purifying, and then removing the protecting group on the linker to obtain the combination 1 (e.g., 2- (2, 6-dioxopiperidin-3-yl) -piperazine-1, 3-dione).
Step 6: compound 5 and combination 1 are dissolved in an organic solvent (e.g., N dimethylformamide), stirred (e.g., at 10-35 degrees c) for 1 hour, then a reducing agent (e.g., an alkali metal borohydride such as sodium borohydride, potassium borohydride, lithium borohydride or sodium triacetylborohydride) is added, stirring is continued (preferably, stirring is performed at room temperature), and after the reaction, the compound of formula (I) is obtained by extraction and purification. For example, other target compounds (I) than the compounds QDE-003-C, QDE-003-D listed below, namely, the compounds QDE-003, QDE-003-A, QDE-003-B, QDE-003-E, QDE-003-F, QDE-003-G, QDE-003-H, QDE-003-I, or QDE-003-J listed below.
More preferably, the method for preparing the estrogen receptor proteolytic targeting chimera (I) (i.e. compound (I)) comprises:
Step 1: dissolving compound 1 in 1, 4-dioxane under anhydrous and anaerobic environment, adding triphenylphosphine palladium and cesium carbonate, ventilating with nitrogen for several times (for example, two, three or four times), and adding (4-bromophenyl) boric acid The reaction is carried out at elevated temperature (preferably 80-130 c, preferably 85-120 c, more preferably 90-110 c, for example 95, 100 or 105 c) (for example 6-20 hours or 8-16 hours, for example 10-12 hours), TLC monitors the progress of the reaction, filtration is carried out after completion of the reaction (preferably, filtration while hot), the filtrate is extracted (for example three times) with an organic solvent as extractant and water (for example ethyl acetate and water), the organic solvent (for example ethyl acetate) layer is separated off with a separating funnel, evaporated to dryness in vacuo, purified (for example column chromatography purification) to give compound 2,
Wherein compound 1 has the following structural formula: (e.g./>) );
Wherein compound 2 has the following structural formula: (e.g./>) )。
Step 2: dissolving compound 2 and 4- (diethoxymethyl) piperidine in an anhydrous and anaerobic environment, adding an organic solvent (e.g. 1,4 dioxane), adding dibenzylideneacetone dipalladium, 2-dicyclohexylphosphine-2, 6-diisopropyloxybiphenyl, cesium carbonate, venting with nitrogen (e.g. twice, three times or four times), reacting at elevated temperature (preferably 70-120 ℃, preferably 75-110 ℃, more preferably 80-100 ℃, e.g. 85, 90 or 95 ℃) for 6-20 hours or 8-16 hours, e.g. 10-12 hours, TLC monitoring the progress of the reaction, filtering (preferably, filtering while hot) after completion of the reaction, taking the filtrate, extracting (e.g. three times) with an organic solvent and water (e.g. ethyl acetate and water) as extractant, separating the organic solvent (e.g. ethyl acetate) layer with a separating funnel, evaporating in vacuo, purifying (e.g. column chromatography) to give compound 3:
(e.g./>) )。
Step 3: dissolving compound 3 in an anhydrous and oxygen-free environment, adding an organic solvent (e.g., anhydrous tetrahydrofuran), cooling the mixture to a temperature of2 ℃ to-2 ℃ (e.g., 1 ℃ to-1 ℃, such as 0 ℃), replacing nitrogen (e.g., two, three or four times), slowly dropping a solution of pyridinium tribromide in the organic solvent (e.g., a tetrahydrofuran solution of pyridinium tribromide using a syringe) into the reaction system, continuing the reaction at normal temperature (preferably 20 to 40 ℃, such as 25 to 35 ℃) for (e.g., 4 to 15 hours or 5 to 14 hours, such as 6 to 13 hours, 7 to 12 hours, or 8 to 10 hours), adding an inorganic base (e.g., sodium carbonate solution or sodium bicarbonate solution or potassium bicarbonate solution), quenching the reaction, extracting the reaction mixture with an organic solvent and water (e.g., ethyl acetate and water) as an extractant (e.g., three times), separating the organic solvent (e.g., ethyl acetate) layer with a separating liquid, distilling under reduced pressure (preferably 40 to 60 ℃, such as 50 ℃), purifying the compound by distillation column (e.g., purifying the purified compound under reduced pressure at 50 ℃):
(e.g./>) )。
Step 4: compound 4 is reacted with (2, 4-dichlorophenyl) boronic acid, palladium tetraphenyl phosphine, cesium carbonate, added to a reactor (e.g., a round bottom flask), an organic solvent (e.g., 1, 4-dioxane) is added to dissolve the mixture, nitrogen displacement (e.g., two, three or four times), at elevated temperature (preferably 80-130 ℃, preferably 85-120 ℃, more preferably 90-110 ℃, e.g., 95, 100 or 105 ℃) for (e.g., 6-15 hours or 7-12 hours, e.g., 8-10 hours), TLC monitors the progress of the reaction, and after completion of the reaction, filtration (preferably, hot filtration); the reaction mixture is extracted (e.g., three times) with an organic solvent such as ethyl acetate and water as extractant, and the organic solvent (e.g., ethyl acetate) layer is separated with a separating funnel, distilled under reduced pressure (preferably, distilled under reduced pressure at a temperature of 40-60 ℃, such as 50 ℃) and purified (e.g., column chromatography purification) to give compound 5 (i.e., methyl 8- (2, 4-dichlorophenyl) -9- (4- (4-formylpiperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate):
(e.g./>) )。
Step 5: dissolving lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione with an organic solvent (e.g., dimethyl sulfoxide, or N, N dimethylformamide), adding a linker with a tertiary amine compound (e.g., a tertiary amine containing an isopropyl group such as N, N-diisopropylethylamine), reacting (e.g., 95-12 hours or 5-8 hours, such as 90-110 ℃ C.) at an elevated temperature (preferably 80-120 ℃ C., more preferably 85-120 ℃ C.), separating and purifying, and then removing the protecting group on the linker to obtain the combination (e.g., 2- (2, 6-dioxopiperidin-3-yl) -1, 3-dione).
Step 6: dissolving compound 5 and combination 1 with an organic solvent (e.g., N dimethylformamide), stirring (e.g., at room temperature, e.g., 20-40deg.C, such as 25-35deg.C) for 1 hr, adding a reducing agent (e.g., an alkali metal borohydride such as sodium borohydride, potassium borohydride, lithium borohydride or sodium triacetyl borohydride), continuing stirring (preferably, stirring at room temperature, e.g., 20-40deg.C, such as 25-35deg.C), and extracting and purifying after the reaction is completed to obtain the compound of formula (I). For example, other target compounds (I) than the compounds QDE-003-C, QDE-003-D listed below, namely, the compounds QDE-003, QDE-003-A, QDE-003-B, QDE-003-E, QDE-003-F, QDE-003-G, QDE-003-H, QDE-003-I, or QDE-003-J listed below.
Wherein the compound QDE-003-C and the compound QDE-003-D are prepared by respectively adding ammonia water and sodium hydroxide solution into the compound QDE-003-A for hydrolysis, separating and purifying. Wherein the compound QDE-003-C is prepared by adding sodium hydroxide solution into the compound QDE-003-A for hydrolysis, separating and purifying. Wherein the compound QDE-003-D is prepared by adding ammonia water into the compound QDE-003-A for hydrolysis, separating and purifying.
Therefore, the preparation method further comprises the following step 6: adding sodium hydroxide solution into the compound QDE-003-A for hydrolysis, separating and purifying to obtain the compound QDE-003-C, or adding ammonia water into the compound QDE-003-A for hydrolysis, separating and purifying to obtain the compound QDE-003-D.
According to a preferred embodiment of the present invention, as compounds of formula (I), the following compounds are provided:
The invention carries out intensive chemical and pharmacological activity researches on series of proteolytic targeted chimeric compounds, discovers that more than ten compounds (QDE-003 and QDE-003-A to J) have better inhibiting effect on estrogen, has the prospect of developing new medicaments for treating breast cancer, and provides a new approach for developing new medicaments for treating breast cancer and enriching clinical medicament varieties.
The estrogen receptor proteolytic targeting chimera provided by the invention has good estrogen receptor protein degradation activity, and can be used for preparing medicaments for resisting breast cancer.
The present application also provides a pharmaceutical composition comprising a therapeutically effective amount of compound (I) of the present application, an isomer thereof or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
The application also provides application of the compound (I), an isomer or a pharmaceutically acceptable salt thereof in preparing medicaments for treating diseases related to estrogen receptor protein degradation targeting chimera.
The present application also provides a method of treating a disease associated with estrogen receptor protein degradation targeted chimeras comprising administering to a mammal (preferably a human) in need of such treatment a therapeutically effective amount of a compound of the present application, an isomer thereof, or a pharmaceutically acceptable salt thereof.
The application also provides the use of a compound of the application, an isomer thereof or a pharmaceutically acceptable salt thereof in the treatment of a disease associated with estrogen receptor protein degradation targeting chimeras.
The application also provides isomers of the compounds of the application or pharmaceutically acceptable salts thereof for use in the treatment of diseases associated with estrogen receptor protein degradation targeted chimeras.
The application also provides isomers of the compounds of the application or pharmaceutically acceptable salts thereof for use in the treatment of diseases associated with estrogen receptor protein degradation targeted chimeras.
In some embodiments of the application, the estrogen receptor protein degradation targeting chimeric related disorder is selected from a tumor or a cancer.
In some aspects of the application, the estrogen receptor protein degradation targeted chimeric related disorder is selected from the group consisting of breast cancer, endometrial cancer, ovarian cancer, uterine cancer, prostate cancer, endometriosis, lung cancer, and esophageal cancer.
In some embodiments of the application, the estrogen receptor protein degradation targeting chimeric related disorder is selected from breast cancer.
Correlation definition
The following terms and phrases used herein are intended to have the following meanings unless otherwise indicated. A particular term or phrase, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense. When trade names are presented herein, it is intended to refer to their corresponding commercial products or active ingredients thereof.
The term "pharmaceutically acceptable" as used herein is intended to refer to those compounds, materials, compositions and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation and allergic response. Or other problems or complications, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salt" refers to salts of the compounds of the present application prepared from the compounds of the present application which have the specified substituents found herein with relatively non-toxic acids or bases. When the compounds of the present application contain relatively acidic functional groups, base addition salts may be obtained by contacting such compounds with a sufficient amount of base in pure solution or in a suitable inert solvent. When the compounds of the present application contain relatively basic functional groups, the acid addition salts may be obtained by contacting such compounds with a sufficient amount of acid in pure solution or in a suitable inert solvent. Certain specific compounds of the application contain basic and acidic functionalities that can be converted to either base or acid addition salts.
Pharmaceutically acceptable salts of the application can be synthesized from the parent compound containing an acid or base by conventional chemical methods. In general, the preparation of such salts is as follows: prepared via reaction of these compounds in free acid or base form with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of both.
The compounds of the application may exist in specific geometric or stereoisomeric forms. The present application contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (2) -isomers, and racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the application. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers and mixtures thereof are included within the scope of the present application.
The compounds and intermediates of the application may also exist in different tautomeric forms and all such forms are included within the scope of the application. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can interconvert via a low energy barrier. For example, proton tautomers (also known as proton transfer tautomers) include tautomers via proton transfer, such as keto-enol and imine-enamine isomerisation. A specific example of a proton tautomer is an imidazole moiety, where a proton can migrate between two ring nitrogens. Valence tautomers include tautomers by recombination of some bond-forming electrons. The compounds of the present application may contain non-natural proportions of atomic isotopes on one or more of the atoms comprising the compounds. For example, compounds such as tritium (2H), iodine-125 (125I) or C-14 (14C) may be labeled with a radioisotope. For example, deuterium can be substituted for hydrogen to form a deuterated drug, and the bond between deuterium and carbon is stronger than the bond between normal hydrogen and carbon, so that the deuterated drug has the advantages of reducing toxic and side effects, increasing the stability of the drug, enhancing the curative effect, prolonging the biological half-life of the drug and the like compared with the non-deuterated drug. All isotopic variations of the compounds of the present application, whether radioactive or not, are intended to be encompassed within the scope of the present application.
The term "optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The term "targeting chimera" means a bifunctional molecule comprising two small molecule ligands, one with high affinity for the target protein of interest, and a second for recruiting E3 ligase that ubiquitinates the protein and targets the protein for proteolysis by the proteasome.
The compounds of the present application may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, embodiments formed by combining with other chemical synthetic methods, and equivalent alternatives well known to those skilled in the art, preferred embodiments including but not limited to the examples of the present application.
The solvent used in the present application is commercially available. Compounds are named according to the general naming principles in the art or using ChemDraw@software, and commercially available compounds are referred to by the vendor catalog name.
The beneficial technical effects of the invention
The compound provided by the application is taken as a difunctional ER PROTAC molecule with a novel structure, has good inhibition effect on estrogen-induced signal transduction and good degradation effect on ERa. The difunctional compound has good properties such as pharmacokinetics, bioavailability, in-vivo efficacy and the like, and can be developed into a novel estrogen receptor degradation agent.
Drawings
FIG. 1 shows the results of an estrogen receptor proteolytic activity assay.
Detailed Description
The following examples illustrate the technical aspects of the invention, and the scope of the invention claimed includes but is not limited to the following examples.
In order to find estrogen receptor degradants with better curative effect, a plurality of estrogen receptor degradants are synthesized according to related documents by combining the current knowledge of the types of medicines, and the obtained target compounds are subjected to estrogen receptor protein degradation activity test, ten compounds are found to have certain degradation effect on estrogen receptors, wherein the compounds QDE-003-C, QDE-003-D, QDE-003-E and QDE-003-F have better degradation effect compared with fulvestrant.
The structure of the target compound has been confirmed by high resolution mass spectrometry (HR-ESI-MS) and nuclear magnetic resonance hydrogen spectrometry (1H-NMR).
The embodiment of the invention adopts the conditions of the reagent:
In the present invention, the compound 1 is 9- (((trifluoromethyl) sulfonyl) oxy) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylic acid or 9- (((trifluoromethyl) sulfonyl) oxy) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylic acid ester, for example, 9- (((trifluoromethyl) sulfonyl) oxy) -6, 7-dihydro-5H-benzo [7] cyclo-3-carboxylic acid methyl ester (CAS 2114341-38-5), haohong biological medicine science, inc.
4- (Diethoxymethyl) piperidine: CAS144872-31-1.
Pyridinium tribromide: CAS 39416-48-3.
(2, 4-Dichlorophenyl) boronic acid: CAS 68716-47-2.
Lenalidomide: CAS191732-72-6.
Thalidomide: CAS 50-35-1.
4-Hydroxy-thalidomide: CAS 64567-60-8.
5-Hydroxy-thalidomide: CAS203450-07-1.
Pomalidomide: CAS19171-19-8.
3- (1-Oxo-isoindolin-2-yl) piperidine-2, 6-dione: CAS26581-81-7.
Example 1:
The compound QDE-003 was prepared as follows:
Step 1: 5g of methyl 9- ((trifluoromethyl) sulfonyl) oxy) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate (14.3 mmol) and 4.3g of 4-bromophenylboronic acid (21.5 mmol) are dissolved in 1,4 dioxane (50 ml), 1.65g of tetrakis (triphenylphosphine) palladium (1.43 mmol) and 9.31g of cesium carbonate (28.6 mmol) are added, the reaction is carried out three times under nitrogen ventilation at 100 ℃ for 10-12 hours, TLC monitors the progress of the reaction, the reaction is filtered off while it is still hot, the filtrate is extracted three times with ethyl acetate and water, the ethyl acetate layer is separated off by a separating funnel, dried under vacuum and purified by column chromatography to obtain 3.72g of methyl 6, 7-dihydro-9- (4-bromophenyl) -5H-benzylcycloheptene-3-carboxylate, the yield of the compound 2, 72.2%. Multiple reactions accumulate the mass of this compound.
Step 2: 2.3g of Compound 2 (6.4 mmol) and 2.4g of 4- (diethoxymethyl) piperidine (12.8 mmol) are dissolved in 30ml of 1,4 dioxane, 5.86g of dibenzylideneacetone dipalladium (0.64 mmol), 6g of 2-dicyclohexylphosphine-2, 6-diisopropyloxybiphenyl (1.28 mmol), 4.17g of cesium carbonate (12.8 mmol) are taken in nitrogen and purged three times, the reaction progress is monitored by TLC at 90℃for 10-12H, after completion of the reaction, the reaction is filtered while hot, the filtrate is extracted three times with ethyl acetate and water, the ethyl acetate layer is separated by a separating funnel, and after vacuum evaporation, 1.87g of methyl 9- (4- (4- (diethoxymethyl) piperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] piperidin-3-carboxylate is purified by column chromatography, the yield of Compound 3 is 63%. Multiple reactions accumulate the mass of this compound.
Step 3: 3g of Compound 3 (6.45 mmol) was dissolved in 50ml of tetrahydrofuran, cooled to 0℃in an ice bath, replaced with nitrogen gas three times, 2.5g of pyridinium tribromide (6.45 mmol) in 10ml of tetrahydrofuran was slowly dropped into the reaction system using a syringe, the ice bath was removed after the dropping was completed, the reaction was carried out for 8-10 hours at room temperature, after quenching with sodium bicarbonate solution, ethyl acetate was extracted with water three times, the ethyl acetate layer was separated out with a separating funnel, distilled under reduced pressure at 50℃and purified by column chromatography to give 1.28g of methyl 8-bromo-9- (4- (4-formylpiperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate in 42.5% yield, namely Compound 4. Multiple reactions accumulate the mass of this compound.
Step 4: 1.5g of Compound 4 (3.2 mmol) and 0.8g of (2, 4-dichlorophenyl) boronic acid (4.8 mmol), 0.7g of tetrakis triphenylphosphine palladium (0.32 mol), 3.1g of cesium carbonate (9.6 mmol) were added to a round-bottomed flask, 50ml of 1, 4-dioxane was dissolved, nitrogen was replaced three times, the reaction was continued at 100℃for 8 to 10 hours, TLC was monitored for the progress of the reaction, and after completion of the reaction, it was filtered while it was still hot. Ethyl acetate was extracted with water three times, ethyl acetate was separated by a separating funnel, distilled under reduced pressure, and purified by column chromatography to give 0.76g of methyl 8- (2, 4-dichlorophenyl) -9- (4- (4-formylpiperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate, yield 44.2%, compound 5.
Step 5: 5g of 5-fluorosalicylic amine (18.1 mmol) was dissolved in dimethyl sulfoxide, 6.7g of 1- (t-butoxycarbonyl) piperazine (36.2 mmol) and 4.7g of N, N-diisopropylethylamine (36.2 mmol) were added, the mixture was reacted at 90℃for 8 hours, after separation and purification by column chromatography (PE: EA=1:1), ethyl acetate hydrochloride solution was added, and after stirring at normal temperature for 4 hours, the solution was distilled off in vacuo to give 2- (2, 6-dioxopiperidin-3-yl) -5- (piperazin-1-yl) isoindoline-1, 3-dione dihydrochloride, and saturated sodium bicarbonate solution and dichloromethane 1) were added: 2, stirring at room temperature for 3 hours, separating, and spin-drying a dichloromethane layer to obtain 5.14g of 2- (2, 6-dioxopiperidin-3-yl) -5- (piperazin-1-yl) isoindoline-1, 3-dione (i.e. combination 1) with a yield of 83%.
Step 6: 1g of Compound 5 (1.8 mmol) and 0.92g of 2- (2, 6-dioxopiperidin-3-yl) -5- (piperazin-1-yl) isoindoline-1, 3-dione (2.7 mmol) were dissolved in 30ml of N, N dimethylformamide, stirred at room temperature for 1 hour, then 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with water, ethyl acetate and water, then a saturated brine wash ethyl acetate phase was added, and the plates were prepared by distillation under reduced pressure to give 0.66g of QDE-003 pure product with a yield of 41.0%.
Confirmation of QDE-003 Structure by high resolution Mass Spectrometry (HR-ESI-MS) and Nuclear magnetic resonance Hydrogen Spectrometry (1H-NMR) :ESI-MS(positive mode)m/z:861.3[M+H]+(calculated for C48H47Cl2N5O6,860.8).1H-NMR(600Hz,CDCl3):7.951-6.650(C-H,13H),5.398(N-CH,1H),3.888(CH3,3H),3.486-3.106(N-CH2,8H),3.169(C=CCH2,2H),2.916-1.253(CH2,18H),1.866(CH,1H).
Example 2:
The compound QDE-003-A is prepared as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 1g of Compound 5 (1.8 mmol) and 1.36g of (S) -3- (1-oxo-5- (piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione benzenesulfonate (2.7 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after TLC detection reaction was completed, quenched with saturated ammonium chloride solution, extracted with ethyl acetate and water, and then a saturated brine-washed ethyl acetate phase was added, and after distillation under reduced pressure, plates were prepared for separation to give QDE-003-A0.42 g, yield 27%.
The structure of QDE-003-A was confirmed by high resolution mass spectrometry (HR-ESI-MS): ESI-MS (positive mode) m/z 847.5[ M+H ] + (calculated for C48H49Cl2N5O5, 846.8).
Example 3:
the preparation of compound QDE-003-B is carried out as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 1g of Compound 5 (1.8 mmol) and 0.47g of (3S) -3- (5-amino-1, 3-dihydro-1-oxo-2H-isoindol-2-yl) -2, 6-piperidinedione (1.8 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with water, washed with saturated brine ethyl acetate phase, distilled under reduced pressure, and separated from the preparation plate to give 0.50g of pure QDE-003-B in 30.1% yield.
Confirmation of the structure of QDE-003-B by high resolution Mass Spectrometry (HR-ESI-MS) Nuclear magnetic resonance Hydrogen Spectrometry (1H-NMR) :ESI-MS(positive mode)m/z:779.3[M+H]+(calculated for C44H42Cl2N4O5,777.7),H-NMR(600Hz,CDCl3):7.945-6.618(C-H,13H),5.210-3.609(N-CH2,7H),3.630(C=C-CH2,2H),3.100(CH3,3H),2.916-1.253(CH2,14H).
Example 4:
the preparation of compound QDE-003-C is carried out as follows:
The preparation method of 5 steps from step 1 to step 5 was substantially the same as in example 2 except for the amount.
Step 6: 0.50g of QDE-003-A (0.6 mmol) is dissolved in tetrahydrofuran, 2M NaOH solution is added to adjust the PH=13, the mixture is stirred for 5 to 6 hours at normal temperature, TLC detection reaction is carried out, acetic acid is added to adjust the PH=7 after the reaction is completed, ethyl acetate and saturated saline water are extracted, and the ethyl acetate phase is taken for reduced pressure distillation and then column chromatography is carried out to obtain 0.43g of QDE-003-C pure product, and the yield is 85.1%.
The structure of QDE-003-C was confirmed by high resolution mass spectrometry (HR-ESI-MS): ESI-MS (positive mode) m/z 852.3[ M+H ] + (calculated for C47H49Cl2N5O6, 850.8).
Example 5:
The preparation of compound QDE-003-D is carried out as follows:
The preparation method of 5 steps from step 1 to step 5 was substantially the same as in example 2 except for the amount.
Step 6: 0.50g of QDE-003-A (0.6 mmol) is dissolved in tetrahydrofuran, 25% ammonia water is added to adjust the PH=8-9, the mixture is stirred for 5-6 hours at normal temperature, TLC detection reaction is carried out, acetic acid is added to adjust the PH=7 after the reaction is completed, ethyl acetate and saturated saline water are extracted, and the ethyl acetate phase is taken for reduced pressure distillation and then column chromatography is carried out to obtain 0.33g of QDE-003-D pure product, and the yield is 66%.
The structure ESI-MS (positive mode) m/z of QDE-003-D was confirmed by high resolution mass spectrometry (HR-ESI-MS) 866.4[ M+H ] + (calculated for C48H 512 Cl2N5O6, 864.8).
Example 6:
the preparation of compound QDE-003-E is carried out as follows:
The preparation method of 4 steps from step 1 to step 4 was substantially the same as in example 1 except for the amount.
Step 5: 1.5g of Compound 5 (3.2 mmol) and 0.8g of (2, 4-dichlorophenyl) boronic acid (4.8 mmol), 0.7g of tetrakis triphenylphosphine palladium (0.32 mol), 3.1g of cesium carbonate (9.6 mmol) were added to a round-bottomed flask, 50ml of 1, 4-dioxane was dissolved, nitrogen was replaced three times, the reaction was continued at 100℃for 8 to 10 hours, TLC was monitored for the progress of the reaction, and after completion of the reaction, it was filtered while it was still hot. Ethyl acetate was extracted with water three times, ethyl acetate was separated by a separating funnel, distilled under reduced pressure, and purified by column chromatography to give 1.63g of pure methyl 8- (2, 4-dichlorophenyl) -9- (4- (4- ((4- (diethoxymethyl) piperidin-1-yl) methyl) piperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate, 78.4% yield, i.e. compound 6.
Step 6: 1.5g of compound 6 is added into 10ml of hydrochloric acid/ethyl acetate solution, stirred at normal temperature for 4-5 hours, after TLC detection reaction is finished, saturated sodium bicarbonate solution is added to adjust PH=7, quenching reaction is carried out, ethyl acetate and water are extracted, the ethyl acetate phase is distilled under reduced pressure, and column chromatography is carried out to obtain 1.12g of pure 8- (2, 4-dichlorophenyl) -9- (4- (4- ((4-formylpiperidine-1-yl) methyl) piperidine-1-acyl) phenyl) -6, 7-dihydro-5H-benzo [7] cycloolefin-3-carboxylic acid methyl ester, and the yield is 83 percent, namely the compound 7.
Step 7: 1g of Compound 7 (1.5 mmol) and 0.68g of (S) -3- (1-oxo-5- (piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione benzenesulfonate (2.4 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, 0.63g of sodium triacetylborohydride (3 mmol) was added, stirred at room temperature for 8 to 10 hours, after TLC detection reaction was completed, quenched with saturated ammonium chloride solution, extracted with water, ethyl acetate and water, a saturated brine-washed ethyl acetate phase was added, and the plates were prepared by distillation under reduced pressure to obtain 0.35g of QDE-003-E in 23.6% yield.
Confirmation of the structure of QDE-003-E by high resolution Mass Spectrometry (HR-ESI-MS) Nuclear magnetic resonance Hydrogen Spectrometry (1H-NMR) :ESI-MS(positive mode)m/z:945.5[M+H]+(calculated for C54H60Cl2N5O6,944.0);H-NMR(600Hz,CDCl3):7.945-6.618(C-H,13H),5.210-3.609(N-CH2,19H),3.630(C=C-CH2,2H)3.100(CH3,3H),2.916-1.253(CH2,20H).
Example 7:
the preparation of compound QDE-003-F is carried out as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 5g of 5-fluorosalicide (18.1 mmol) was dissolved in dimethyl sulfoxide, 8.7g of tert-butyl 3- (piperidin-4-yl) azetidine-1-carboxylate (36.2 mmol) and 4.7g of N, N-diisopropylethylamine (36.2 mmol) were added, after purification by column chromatography (PE: EA=1:1) at 90℃for 8 hours, ethyl acetate hydrochloride solution was added, stirring at room temperature for 4 hours, and the solution was distilled off in vacuo to give 5- (4- (azetidin-3-yl) piperidin-1-yl) -2- (2, 6-dioxapiperidin-3-yl) isoindoline-1, 3-dione hydrochloride, and saturated sodium bicarbonate solution was added with dichloromethane 1:2, stirring at normal temperature for 3 hours, separating the solution, taking a dichloromethane layer, spin-drying, and obtaining 6.3g of 5- (4- (azetidin-3-yl) piperidine-1-yl) -2- (2, 6-dioxopiperidine-3-yl) isoindoline-1, 3-dione, wherein the yield is 77%.
Step 6: 1g of Compound 5 (1.8 mmol) was dissolved in 0.72g of 5- (4- (azetidin-3-yl) piperidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (1.8 mmol) with 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, then 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with ethyl acetate and water, and then washed with ethyl acetate phase with saturated saline, and then subjected to distillation under reduced pressure to give 0.61g of QDE-003-F as a pure product in 85.9% yield.
The structure of QDE-003-F was confirmed by high resolution mass spectrometry (HR-ESI-MS): ESI-MS (positive mode) m/z 915.6[ M+H ] + (calculated for C52H53Cl2N5O6, 914.9).
Example 8:
the preparation of compound QDE-003-G is carried out as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 5g of 5-fluorosalicide (18.1 mmol) was dissolved in dimethyl sulfoxide, 8.7g of tert-butyl 4- (azetidin-3-yl) piperidine-1-carboxylate (36.2 mmol) and 4.7g of N, N-diisopropylethylamine (36.2 mmol) were added, after purification by column chromatography (PE: EA=1:1) at 90℃for 8 hours, ethyl acetate hydrochloride solution was added, stirring at room temperature for 4 hours and then vacuum distillation was carried out to obtain 2- (2, 6-dioxapiperidin-3-yl) -5- (2, 7-diazaspiro [3.5] non-2-yl) isoindoline-1, 3-dione hydrochloride, saturated sodium bicarbonate solution and dichloromethane 1:2, stirring for 3 hours at normal temperature, separating the liquid, taking a dichloromethane layer, spin-drying to obtain 6.6g of 2- (2, 6-dioxopiperidine-3-yl) -5- (2, 7-diazaspiro [3.5] non-2-yl) isoindoline-1, 3-dione, and the yield is 79%.
Step 6: 1G of Compound 5 (1.8 mmol) and 0.69G of 2- (2, 6-dioxopiperidin-3-yl) -5- (2, 7-diazaspiro [3.5] non-2-yl) isoindoline-1, 3-dione (1.8 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, then 0.76G of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with ethyl acetate and water, washed with saturated brine ethyl acetate phase, and distilled under reduced pressure to give a plate, 0.79G of QDE-003-G as a pure product, yield 47.0%.
Confirmation of the structure of QDE-003-G by high resolution Mass Spectrometry (HR-ESI-MS) Nuclear magnetic resonance Hydrogen Spectrometry (1H-NMR) :ESI-MS(positive mode)m/z:902.4[M+H]+(calculated for C51H51Cl2N5O6,900.9)1H-NMR(600Hz,CDCl3):7.945-6.618(C-H,13H),5.210-3.609(N-CH2,11H),3.630(C=C-CH2,2H),3.100(CH3,3H),2.916-1.253(CH2,20H).
Example 9:
the preparation of compound QDE-003-H is carried out as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 5g of 5-fluorosalicide (18.1 mmol) was dissolved in dimethyl sulfoxide, 8.2g of tert-butyl 2, 7-diazaspiro [3.5] nonane-7-carboxylate (36.2 mmol) and 4.7g of N, N-diisopropylethylamine (36.2 mmol) were added, after separation and purification by column chromatography (PE: EA=1:1) at 90℃ethyl acetate solution was added, stirring at room temperature for 4 hours and then vacuum distillation was carried out to obtain 2- (2, 6-dioxapiperidin-3-yl) -5- (2, 7-diazaspiro [3.5] non-7-yl) isoindoline-1, 3-dione hydrochloride, saturated sodium bicarbonate solution and dichloromethane 1:2, stirring for 3 hours at normal temperature, separating the liquid, taking a dichloromethane layer, spin-drying to obtain 7.26g of 2- (2, 6-dioxopiperidine-3-yl) -5- (2, 7-diazaspiro [3.5] non-2-yl) isoindoline-1, 3-dione, and obtaining the yield of 82%.
Step 6: 1g of Compound 5 (1.8 mmol) and 0.69g of 2- (2, 6-dioxopiperidin-3-yl) -5- (2, 7-diazaspiro [3.5] non-7-yl) isoindoline-1, 3-dione (1.8 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, then 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with ethyl acetate and water, washed with saturated brine ethyl acetate phase, and distilled under reduced pressure to give a plate, 0.74g of QDE-003-H as a pure product, 43.7% was obtained.
The structure of QDE-003-H was confirmed by high resolution mass spectrometry (HR-ESI-MS) by nuclear magnetic resonance hydrogen spectrometry (1H-NMR):
ESI-MS(positive mode)m/z:902.4[M+H]+(calculated for C53H55Cl2N5O6,900.9);1H-NMR(600Hz,CDCl3):7.945-6.618(C-H,13H),5.210-3.609(N-CH2,11H),3.630(C=C-CH2,2H),3.100(CH3,3H),2.916-1.253(CH2,20H).
Example 10:
the preparation of compound QDE-003-I is carried out as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 5g of 5-fluorosalicylic amine (18.1 mmol) was dissolved in dimethyl sulfoxide, 9.2g of tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (36.2 mmol) and 4.7g of N, N-diisopropylethylamine (36.2 mmol) were added, after separation and purification by column chromatography (PE: EA=1:1) at 90 ℃ C.) ethyl acetate solution was added, stirring at room temperature for 4 hours and then vacuum distillation was carried out to obtain 2- (2, 6-dioxapiperidin-3-yl) -5- (3, 9-diazaspiro [5.5] undec-3-yl) isoindoline-1, 3-dione hydrochloride, saturated sodium bicarbonate solution and dichloromethane 1:2, stirring for 3 hours at normal temperature, separating the liquid, taking a dichloromethane layer, spin-drying, and obtaining 6.21g of 2- (2, 6-dioxopiperidine-3-yl) -5- (3, 9-diazaspiro [5.5] undec-3-yl) isoindoline-1, 3-dione, wherein the yield is 77%.
Step 6: 1g of Compound 5 (1.8 mmol) and 0.74g of 2- (2, 6-dioxopiperidin-3-yl) -5- (3, 9-diazaspiro [5.5] undec-3-yl) isoindoline-1, 3-dione (1.8 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, then 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with ethyl acetate and water, then ethyl acetate phase was washed with saturated saline, and a plate was prepared after distillation under reduced pressure to obtain 0.77g of pure QDE-003-I in 45.6% yield.
The structure of QDE-003-I was confirmed by high resolution mass spectrometry (HR-ESI-MS): ESI-MS (positive mode) m/z 930.6[ M+H ] + (calculated for C53H55Cl2N5O6, 929.0).
Example 11:
the preparation of compound QDE-003-J is carried out as follows:
The preparation method of the four steps from step 1 to step 4 was substantially the same as in example 1 except for the amounts.
Step 5: 5g of 5-fluorosalicylic amine (18.1 mmol) was dissolved in dimethyl sulfoxide, 9.2g of tert-butyl 2, 9-diazaspiro [5.5] undecane-9-carboxylate (36.2 mmol) and 4.7g of N, N-diisopropylethylamine (36.2 mmol) were added, after separation and purification by column chromatography (PE: EA=1:1) at 90 ℃ C.) ethyl acetate solution was added, stirring at room temperature for 4 hours and then vacuum distillation was carried out to obtain 2- (2, 6-dioxapiperidin-3-yl) -5- (3, 9-diazaspiro [5.5] undec-3-yl) isoindoline-1, 3-dione hydrochloride, saturated sodium bicarbonate solution and dichloromethane 1:2, stirring for 3 hours at normal temperature, separating the liquid, taking a dichloromethane layer, spin-drying, and obtaining 5.6g of 2- (2, 6-dioxopiperidine-3-yl) -5- (3, 9-diazaspiro [5.5] undecane-3-yl) isoindoline-1, 3-dione, wherein the yield is 70%.
Step 6: 1g of Compound 5 (1.8 mmol) and 0.74g of 2- (2, 6-dioxopiperidin-3-yl) -5- (2, 9-diazaspiro [5.5] undec-2-yl) isoindoline-1, 3-dione (1.8 mmol) were dissolved in 30ml of N, N-dimethylformamide, stirred at room temperature for 1 hour, then 0.76g of sodium triacetylborohydride (3.6 mmol) was added, stirred at room temperature for 8 to 10 hours, after completion of TLC detection reaction, quenched with saturated ammonium chloride solution, extracted with ethyl acetate and water, then ethyl acetate phase was washed with saturated saline, and a plate was prepared after distillation under reduced pressure to obtain 0.7g of QDE-003-J pure product in 41.4% yield.
The structure of QDE-003-J was confirmed by high resolution mass spectrometry (HR-ESI-MS): ESI-MS (positive mode) m/z 930.6[ M+H ] + (calculated for C53H55Cl2N5O6, 929.0).
Measurement of estrogen receptor proteolytic Activity:
the test of the proteolytic activity of the estrogen receptor was carried out as follows:
1. Taking MCF-7 cells in logarithmic growth phase, spreading the MCF-7 cells into a 6-well plate according to 1.5X106/well, and culturing overnight;
2. adding a positive medicine and a to-be-detected substance group (the DMSO proportion is 0.1%) with the final concentration of 1 mu M respectively, and incubating in an incubator for 24 hours;
3. Each group of cells was collected and washed 1 time with PBS; cell lysates containing 1X Protease Inhibitor cocktail and 1X Phosphatase Inhibitor Cocktail II were added, and the mixture was thoroughly lysed on ice, and the supernatant was collected by centrifugation at 14000rpm for 10min at 4 ℃.
After the protein concentration was determined by bca kit, protein level Estrogen Receptor alpha was detected by Western blot.
The test results are shown in fig. 1.
The foregoing is merely representative examples of the present invention and is not intended to limit the present invention in any way. Any modification, equivalent variation and modification of the above embodiments according to the technical substance of the present invention still fall within the scope of the technical solution of the present invention.

Claims (10)

1. A compound having the general formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt, hydrate, solvate, polymorph or prodrug thereof, or an estrogen receptor proteolytically targeted chimeric derivative thereof:
In the formula (I), R is one of C1-C5 alkoxy, amino, C1-C5 alkylamino or OH;
The E3 ligase ligand has a structure of a general formula (II); preferably, the E3 ligase ligand is lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindol-1, 3-dione, or derivatives and isomers thereof:
n=0 or 1, m=0 or 1;
Wherein the site connected with linker is one of R1, R2 and R3, and the other substituents, namely the other two of R1, R2 and R3, are respectively and independently: one of H, OH, F, cl, br, NH2, CH3 or OCH 3; or one E3 ligase ligand when n=0 or 1, m=0 or 1, R4 is hydrogen or methyl in formula (II) is thalidomide or one of its variants 4-amino-4-oxo-2- (1-oxo-isoindol-2-yl) butyric acid;
Linker in formula (I) is a linker.
2. A compound according to claim 1, wherein: the E3 ligase ligand in formula (I) is lenalidomide (IIa):
alternatively, the E3 ligase ligand in formula (I) is thalidomide (IIb):
Or, the E3 ligase ligand in formula (I) is 4-hydroxy-thalidomide (IIc-1) or 5-hydroxy-thalidomide (IIc-2):
alternatively, the E3 ligase ligand in formula (I) is pomalidomide (IId):
or, the E3 ligase ligand of formula (I) is 3- (1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (IIe):
Or, the E3 ligase ligand of formula (I) is 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butanoic acid (IIf):
Or, the E3 ligase ligand of formula (I) is 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione (IIg):
3. The compound of claim 1 or 2, wherein linker is one of a flexible or rigid linker, or a combination of two or more.
4. A compound according to claim 3, wherein the rigid linker is one selected from the following formulae:
and/or wherein the flexible linker is one selected from the following formulas:
n=0-5; and/or
Wherein the combination of the flexible linker and the rigid linker is one selected from the following formulas:
5. A compound having the structural formula:
6. A process for the preparation of a compound of formula (I) as claimed in any one of claims 1 to 4, which process comprises: dissolving compound 5 in an organic solvent, adding an E3 ligase ligand (preferably lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione or a derivative thereof), stirring, then adding a reducing agent (for example, an alkali metal borohydride such as sodium borohydride, potassium borohydride, lithium borohydride or sodium triacetyl borohydride), continuing stirring, and after the reaction is completed, separating and purifying to obtain the target compound (I); wherein compound 5 has the following structural formula:
wherein: r is one of C1-C5 alkoxy, amino, C1-C5 alkylamino, or OH.
7. A process for the preparation of a compound of formula (I) as claimed in any one of claims 1 to 4, which process comprises:
step 1: allowing compound 1 to react with (4-bromophenyl) boronic acid In organic solvent in the presence of catalyst, separating to obtain compound 2,
Wherein compound 1 and compound 2 each have the following structural formula:
wherein: r is one of C1-C5 alkoxy, amino, C1-C5 alkylamino or OH;
step 2: reacting the compound 2 with 4- (diethoxymethyl) piperidine in an organic solvent in the presence of a catalyst, and separating to obtain a compound 3:
step 3: reacting the compound 3 with pyridinium tribromide in an organic solvent in the presence of a catalyst, and separating to obtain a compound 4:
step 4: reacting the compound 4 with (2, 4-dichlorophenyl) boric acid in an organic solvent in the presence of a catalyst, and separating to obtain a compound 5:
Step 5: dissolving lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione with an organic solvent (e.g., dimethyl sulfoxide, or N, N dimethylformamide), adding a linker and a tertiary amine compound (e.g., a tertiary amine containing isopropyl groups such as N, N-diisopropylethylamine), reacting (e.g., 4-12 hours or 5-10 hours such as 6-8 hours) at an elevated temperature (e.g., 80-130 ℃ or 90-110 ℃ such as 100 ℃), separating and purifying, and then removing the protecting group on the linker to obtain a combination 1 (e.g., 2- (2, 6-dioxopiperidin-3-yl) -piperazine-1, 3-dione);
Step 6: compound 5 and combination 1 are dissolved in an organic solvent (e.g., N dimethylformamide), stirred (e.g., at 10-35 degrees c) for 1 hour, then a reducing agent (e.g., an alkali metal borohydride such as sodium borohydride, potassium borohydride, lithium borohydride or sodium triacetylborohydride) is added, stirring is continued (preferably, stirring is performed at room temperature), and after the reaction, the compound of formula (I) is obtained by extraction and purification.
8. The method of claim 7, wherein
Step 1 is performed as follows: dissolving compound 1 in 1, 4-dioxane under anhydrous and anaerobic environment, adding triphenylphosphine palladium and cesium carbonate, ventilating with nitrogen for several times (for example, two, three or four times), and adding (4-bromophenyl) boric acidThe reaction is carried out at elevated temperature (preferably 80-130 c, preferably 85-120 c, more preferably 90-110 c, for example 95, 100 or 105 c) (for example 6-20 hours or 8-16 hours, for example 10-12 hours), TLC monitors the progress of the reaction, filtration is carried out after completion of the reaction (preferably, filtration while hot), the filtrate is extracted (for example three times) with an organic solvent as extractant and water (for example ethyl acetate and water), the organic solvent (for example ethyl acetate) layer is separated off with a separating funnel, evaporated to dryness in vacuo, purified (for example column chromatography purification) to give compound 2,
Wherein compound 1 has the following structural formula:
wherein compound 2 has the following structural formula:
And/or
Step 2 is performed as follows: dissolving compound 2 and 4- (diethoxymethyl) piperidine in an anhydrous and anaerobic environment, adding an organic solvent (e.g. 1,4 dioxane), adding dibenzylideneacetone dipalladium, 2-dicyclohexylphosphine-2, 6-diisopropyloxybiphenyl, cesium carbonate, venting with nitrogen (e.g. twice, three times or four times), reacting at elevated temperature (preferably 70-120 ℃, preferably 75-110 ℃, more preferably 80-100 ℃, e.g. 85, 90 or 95 ℃) for 6-20 hours or 8-16 hours, e.g. 10-12 hours, TLC monitoring the progress of the reaction, filtering (preferably, filtering while hot) after completion of the reaction, taking the filtrate, extracting (e.g. three times) with an organic solvent and water (e.g. ethyl acetate and water) as extractant, separating the organic solvent (e.g. ethyl acetate) layer with a separating funnel, evaporating in vacuo, purifying (e.g. column chromatography) to give compound 3:
And/or
Step 3 is performed as follows: dissolving compound 3 in an anhydrous and oxygen-free environment, adding an organic solvent (e.g., anhydrous tetrahydrofuran), cooling the mixture to a temperature of 2 ℃ to-2 ℃ (e.g., 1 ℃ to-1 ℃, such as 0 ℃), replacing nitrogen (e.g., two, three or four times), slowly dropping a solution of pyridinium tribromide in the organic solvent (e.g., a tetrahydrofuran solution of pyridinium tribromide using a syringe) into the reaction system, continuing the reaction at normal temperature (preferably 20 to 40 ℃, such as 25 to 35 ℃) for (e.g., 4 to 15 hours or 5 to 14 hours, such as 6 to 13 hours, 7 to 12 hours, or 8 to 10 hours), adding an inorganic base (e.g., sodium carbonate solution or sodium bicarbonate solution or potassium bicarbonate solution), quenching the reaction, extracting the reaction mixture with an organic solvent and water (e.g., ethyl acetate and water) as an extractant (e.g., three times), separating the organic solvent (e.g., ethyl acetate) layer with a separating liquid, distilling under reduced pressure (preferably 40 to 60 ℃, such as 50 ℃), purifying the compound by distillation column (e.g., purifying the purified compound under reduced pressure at 50 ℃):
And/or
Step 4 is performed as follows: compound 4 is reacted with (2, 4-dichlorophenyl) boronic acid, palladium tetraphenyl phosphine, cesium carbonate, added to a reactor (e.g., a round bottom flask), an organic solvent (e.g., 1, 4-dioxane) is added to dissolve the mixture, nitrogen displacement (e.g., two, three or four times), at elevated temperature (preferably 80-130 ℃, preferably 85-120 ℃, more preferably 90-110 ℃, e.g., 95, 100 or 105 ℃) for (e.g., 6-15 hours or 7-12 hours, e.g., 8-10 hours), TLC monitors the progress of the reaction, and after completion of the reaction, filtration (preferably, hot filtration); the reaction mixture is extracted (e.g., three times) with an organic solvent such as ethyl acetate and water as extractant, and the organic solvent (e.g., ethyl acetate) layer is separated with a separating funnel, distilled under reduced pressure (preferably, distilled under reduced pressure at a temperature of 40-60 ℃, such as 50 ℃) and purified (e.g., column chromatography purification) to give compound 5, i.e., methyl 8- (2, 4-dichlorophenyl) -9- (4- (4-formylpiperidin-1-yl) phenyl) -6, 7-dihydro-5H-benzo [7] cyclo-olefin-3-carboxylate:
And/or
Step 5 is performed as follows: dissolving lenalidomide, thalidomide, 4-hydroxy-thalidomide, 5-hydroxy-thalidomide, pomalidomide, 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione, 4-amino-4-oxo-2- (1-oxoisoindol-2-yl) butyric acid or 2- (1-methyl-2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione with an organic solvent (e.g., dimethyl sulfoxide, or N, N dimethylformamide), adding a linker with a tertiary amine compound (e.g., a tertiary amine containing isopropyl groups such as N, N-diisopropylethylamine), reacting (e.g., 95-12 hours or 5-8 hours, such as 90-110 ℃ C., 100 or 105 ℃) at an elevated temperature (preferably 80-130 ℃, preferably 85-120 ℃), and separating and purifying to obtain a linker (e.g., 2- (2, 6-diisopropyl-ethylamine) and separating and purifying the linker to obtain a combination (e.g., 2- (2-1-2-dioxan-1-piperidyl) -1, 3-dione);
And/or
Step 6 is performed as follows: dissolving compound 5 and combination 1 with organic solvent (N, N dimethylformamide), stirring (at room temperature, for example, 20-40deg.C, such as 25-35deg.C) for 1 hr, adding reducing agent (for example, alkali metal borohydride such as sodium borohydride, potassium borohydride, lithium borohydride or sodium triacetyl borohydride), stirring (preferably, stirring at room temperature, for example, 20-40deg.C, such as 25-35deg.C), and extracting and purifying to obtain compound of formula (I).
9. A pharmaceutical composition, wherein the composition comprises: a therapeutically effective amount of a compound (I), isomer thereof, or pharmaceutically acceptable salt thereof of any of claims 1-4 and a pharmaceutically acceptable carrier;
Or (b)
The composition comprises: a therapeutically effective amount of a compound of claim 5, an isomer thereof, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
10. Use of a compound (I) according to any one of claims 1 to 4 or a compound according to claim 5, an isomer thereof or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a disease associated with estrogen receptor protein degradation targeted chimeras.
CN202410152143.8A 2024-02-03 2024-02-03 PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent Pending CN118027003A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410152143.8A CN118027003A (en) 2024-02-03 2024-02-03 PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410152143.8A CN118027003A (en) 2024-02-03 2024-02-03 PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent

Publications (1)

Publication Number Publication Date
CN118027003A true CN118027003A (en) 2024-05-14

Family

ID=90996359

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410152143.8A Pending CN118027003A (en) 2024-02-03 2024-02-03 PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent

Country Status (1)

Country Link
CN (1) CN118027003A (en)

Similar Documents

Publication Publication Date Title
KR102482673B1 (en) Aromatic acetylenic or aromatic ethylenic compounds, their intermediates, manufacturing methods, drug compositions and uses
KR102401841B1 (en) estrogen receptor modulators
US20180305334A1 (en) Compounds, compositions and methods of use against stress granules
US20220315578A1 (en) Brd9 bifunctional degraders and their methods of use
KR102609360B1 (en) COT modifiers and methods of using them
TW201725207A (en) Crystalline form of BTK kinase inhibitor and a preparation method thereof
JPS5943459B2 (en) N-alkylpiperidine derivative
US10626107B2 (en) Crystal form, salt type of substituted 2-hydro-pyrazole derivative and preparation method therefor
WO2021213317A1 (en) Hpk1 inhibitor, preparation method therefor and use thereof
CN111410647B (en) Mu opium receptor bias agonist and medical application thereof
CA3026074A1 (en) N-hexanoic-l-tyrosine-l-isoleucine-(6)-aminohexanoic amide compounds and their use to treat neurodegenerative diseases
JP7260718B2 (en) Diazaindole derivatives and their use as Chk1 inhibitors
AU2022250316A1 (en) Tetrahydronaphthalene compound, and preparation method therefor and use thereof in medicine
WO2020173417A1 (en) Acryloyl-containing nuclear transport regulator and uses thereof
CN118027003A (en) PROTAC compound, preparation method thereof and application of PROTAC compound as estrogen receptor degradation agent
WO2023151113A1 (en) Selective histone deacetylase 8 degrader, preparation method and application thereof in anti-tumor activity
AU2019214670A1 (en) ALDH2 activator
WO2022148439A1 (en) Heterocyclic compound as bcl-2 inhibitor
US20230128137A1 (en) Benzo five-membered cyclic compound
WO2021228216A1 (en) BIARYL COMPOUND CAPABLE OF SERVING AS RORγ MODULATOR
TW202146381A (en) Biaryl compound capable of serving as ror[gamma] regulator
TW214545B (en)
CN109384727B (en) Phthalazinone compound, preparation method, pharmaceutical composition and application thereof
JPS6183163A (en) Antitumoral
CN113582971B (en) Small molecule immunosuppressant, preparation method and application thereof

Legal Events

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