CN112574178A - Deuterated benzimidazole compound and application thereof as EP300/CBP inhibitor - Google Patents

Deuterated benzimidazole compound and application thereof as EP300/CBP inhibitor Download PDF

Info

Publication number
CN112574178A
CN112574178A CN202011005085.4A CN202011005085A CN112574178A CN 112574178 A CN112574178 A CN 112574178A CN 202011005085 A CN202011005085 A CN 202011005085A CN 112574178 A CN112574178 A CN 112574178A
Authority
CN
China
Prior art keywords
compound
cbp
inhibitor
cancer
salt
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
CN202011005085.4A
Other languages
Chinese (zh)
Inventor
樊磊
王飞
罗童川
李兴海
陈元伟
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.)
Haichuang Pharmaceutical Co Ltd
Hinova Pharmaceuticals Inc
Original Assignee
Haichuang 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 Haichuang Pharmaceutical Co Ltd filed Critical Haichuang Pharmaceutical Co Ltd
Publication of CN112574178A publication Critical patent/CN112574178A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The invention discloses a deuterated benzimidazole compound and application thereof as an EP300/CBP inhibitor. Specifically provided is a deuterated benzimidazole compound shown in formula I, or a salt thereof, or a conformational isomer thereof, or a crystal form thereof, or a solvate thereof. The compound has high selectivity to EP300/CBP, and can effectively inhibit the activity of EP 300/CBP; meanwhile, compared with a non-deuterated compound, the compound provided by the invention has the advantages that the metabolic stability and the pharmacokinetic performance are obviously improved. In addition, the compound has excellent inhibition effect on various tumor cells including prostate cancer cells, leukemia cells, breast cancer cells and multiple myeloma cells.Therefore, the compound has good application prospect in preparing EP300/CBP inhibitors and medicines for preventing and/or treating tumors and regulating regulatory T cells.

Description

Deuterated benzimidazole compound and application thereof as EP300/CBP inhibitor
Technical Field
The invention belongs to the field of pharmaceutical synthesis industry, and particularly relates to a deuterated benzimidazole compound with a novel structure and application thereof as an EP300/CBP inhibitor.
Background
Histone Acetyltransferases (HATs) and Histone Deacetylases (HDACs) can affect histone acetylation, and recruitment and normal function of HATs and HDACs are key regulatory steps in gene expression and cell cycle, and functional defects of these enzymes may lead to a variety of diseases including tumors.
The EP300/CBP family, consisting of highly homologous adenovirus E1A-associated 300kDa protein (EP300) and the binding protein (CBP) of cyclic adenosine monophosphate response element binding protein (CREB), is one of the major members of the Histone Acetyltransferase (HAT) family. EP300/CBP is involved in cell cycle progression and cell growth, differentiation and development, is a very important class of co-activators, and can regulate the functions of a variety of key transcriptional regulators. Researches show that the EP300/CBP is highly expressed and activated in various tumors, and the EP300/CBP is closely related to various tumor diseases and is a tumor treatment target with a great application prospect. Thus, EP300/CBP inhibitors are receiving increasing attention from researchers.
Researchers at the Structural genomics society of oxford university (SGC) first designed and synthesized the benzimidazole compound 9a, but 9a had poor selectivity for CBP and BRD4 (CBP: IC50 ═ 4 μmol · L)-1,BRD4:IC50=6.3μmol·L-1). Then, compound 10a (CBP: IC50 ═ 0.12 μmol · L) was obtained by further optimization-1,BRD4:IC50=2.4μmol·L-1) And 11a (SGC-CBP30, CBP: IC50 ═ 69nmol-1、Kd=21nmol.L-1,p300:Kd=32nmol.L-1)。
Figure BDA0002695596540000011
However, the EP300/CBP inhibitors reported at present still cannot meet the clinical requirements, and therefore, it is very important to research more EP300/CBP inhibitors with novel structure, higher selectivity and better inhibitory activity.
Deuterated drugs refer to replacement of a portion of the hydrogen atoms in a drug molecule with deuterium. Because deuterium is close to hydrogen in shape and volume in a drug molecule, deuterated drugs generally retain the biological activity and selectivity of the original drug. Since the C-D bond is more stable than the C-H bond, the C-D bond is less likely to be broken during the chemical reaction of the deuterated drug, and the half-life period of the deuterated drug may be prolonged.
However, due to the complex metabolic processes of biological systems, the pharmacokinetic properties of drugs in the body are influenced by various factors and show corresponding complexity. The change in pharmacokinetic properties of deuterated drugs represents a great chance and unpredictability compared to corresponding non-deuterated drugs. Deuteration at some sites, not only does not prolong half-life, but may shorten it (Scott l. harbeson, Roger d. tung. deuterium in Drug Discovery and Development, P405-406.), deteriorating its pharmacokinetic properties; on the other hand, hydrogen at some positions on a drug molecule is also not easily deuterated due to steric hindrance and the like, so that the deuteration of the drug is not random and a site capable of deuteration is unpredictable.
Therefore, the development of the EP300/CBP inhibitor which has excellent pharmacokinetic properties and high activity simultaneously has a very good application prospect.
Disclosure of Invention
The invention aims to provide a high-activity EP300/CBP inhibitor with better metabolic stability and pharmacokinetic performance.
The invention provides a compound shown as a formula I, or a salt, a conformational isomer, a crystal form or a solvate thereof:
Figure BDA0002695596540000021
wherein R is0~R25Each independently selected from H, deuterium, C1-C4Alkyl radical, C3-C6Cycloalkyl, C substituted by more than 1 deuterium1-C4Alkyl, C substituted by more than 1 deuterium3-C6A cycloalkyl group; and R is0~R25At least one group contains deuterium.
Further, R0~R25Each independently selected from H, deuterium, methyl, CD3(ii) a And R is0~R25In which at least one group is deuterium or CD3
Further, R0、R1、R10Each independently selected from methyl or CD3,R2~R9、R11~R25Each independently selected from hydrogen or deuterium; preferably, R10Is a CD3
Further, the structure of the compound is shown as a formula II-1 or a formula II-2:
Figure BDA0002695596540000031
wherein R is0~R25As described above.
Further, the compound is one of the following compounds:
Figure BDA0002695596540000041
the invention also provides application of the compound or a deutero-compound thereof, or a salt thereof, or a conformational isomer thereof, or a crystal form thereof, or a solvate thereof in preparation of an EP300 inhibitor, a CBP inhibitor or an EP300/CBP inhibitor.
Further, the EP300/CBP inhibitor is a medicament for preventing and/or treating tumors and malignant diseases of hematopoietic stem/progenitor cells of myeloid lines and regulating regulatory T cells;
preferably, the tumor is selected from hematological malignancy, gastric cancer, intestinal cancer, cervical cancer, bladder cancer, laryngeal cancer, liver cancer, lung cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma or multiple myeloma, and the myeloid hematopoietic stem/progenitor cell malignancy is leukemia;
more preferably, the lymphoma is non-hodgkin's lymphoma, diffuse large B cell lymphoma and the leukemia is acute myeloid leukemia.
The invention also provides a medicament for treating diseases, which is a preparation prepared by taking the compound, or a deuteron, or a salt, or a conformational isomer, or a crystal form, or a solvate thereof as an active ingredient and adding pharmaceutically acceptable auxiliary materials.
The invention also provides a combined drug which contains the compound, or a deuterogen thereof, or a salt thereof, or a conformational isomer thereof, or a crystal form thereof, or a solvate thereof, which are prepared from unit preparations with the same or different specifications and used for simultaneous or separate administration, other drugs with anti-tumor effects, and a pharmaceutically acceptable carrier.
Further, the other drugs with the anti-tumor effect are chemotherapeutic drugs or radiotherapy drugs, preferably, the chemotherapeutic drugs are targeted drugs; or, the other medicine with the anti-tumor effect is selected from one or more than two of CDK4/6 inhibitor, Parp inhibitor, androgen receptor inhibitor and immune checkpoint inhibitor.
Definitions of terms used in connection with the present invention: the initial definitions provided herein for a group or term apply to that group or term throughout the specification unless otherwise indicated; for terms not specifically defined herein, the meanings that would be given to them by a person skilled in the art are to be given in light of the disclosure and the context.
"EP 300/CBP inhibitor" refers to an inhibitor that is capable of inhibiting both EP300 and CBP activity.
"EP 300" is "P300" and "EP 300/CBP inhibitor" is "P300/CBP inhibitor".
The minimum and maximum values of the carbon atom content in the hydrocarbon group are indicated by a prefix, e.g. prefix Ca-CbAlkyl represents any alkyl group containing from "a" to "b" carbon atoms. E.g. C1-C4The alkyl group is a straight-chain or branched alkyl group having 1 to 4 carbon atoms.
Cycloalkyl refers to a saturated or unsaturated cyclic hydrocarbon substituent; the cyclic hydrocarbon may be monocyclic or polycyclic. For example, "C3-C6The cycloalkyl refers to a saturated or unsaturated cycloalkyl with 3-6 carbon atoms in the ring.
By "pharmaceutically acceptable" is meant that the carrier, diluent, excipient, and/or salt formed is generally chemically or physically compatible with the other ingredients comprising a pharmaceutical dosage form and physiologically compatible with the recipient.
"salts" are acid and/or base salts of a compound or a stereoisomer thereof with inorganic and/or organic acids and/or bases, and also include zwitterionic (inner) salts, as well as quaternary ammonium salts, such as alkylammonium salts. These salts can be obtained directly in the final isolation and purification of the compounds. The compound, or a stereoisomer thereof, may be obtained by appropriately (e.g., equivalentlymixing) a certain amount of an acid or a base. These salts may form precipitates in the solution which are collected by filtration, or they may be recovered after evaporation of the solvent, or they may be prepared by reaction in an aqueous medium followed by lyophilization.
The salts described in the present invention include pharmaceutically acceptable salts. The salt in the invention can be hydrochloride, sulfate, citrate, benzene sulfonate, hydrobromide, hydrofluoride, phosphate, acetate, propionate, succinate, oxalate, malate, succinate, fumarate, maleate, tartrate or trifluoroacetate of the compound.
The experimental result shows that the compound has high selectivity to EP300/CBP and can effectively inhibit the activity of EP 300/CBP; meanwhile, compared with a non-deuterated compound, the compound provided by the invention has the advantages that the metabolic stability and the pharmacokinetic performance are obviously improved. In addition, the compound has excellent inhibition effect on various tumor cells including prostate cancer cells, leukemia cells, breast cancer cells and multiple myeloma cells. Therefore, the compound has good application prospect in preparing EP300/CBP inhibitors and medicines for preventing and/or treating tumors and regulating regulatory T cells.
Obviously, many modifications, substitutions, and variations are possible in light of the above teachings of the invention, without departing from the basic technical spirit of the invention, as defined by the following claims.
The present invention will be described in further detail with reference to the following examples. This should not be understood as limiting the scope of the above-described subject matter of the present invention to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
Detailed Description
The raw materials and equipment used in the invention are known products and are obtained by purchasing commercial products.
Example 1 Synthesis of Compound 100 of the invention
Figure BDA0002695596540000061
(1) Synthesis of Compound 4-bromo-N- ((1r,4r) -4-methoxycyclohexyl) -2-nitroaniline (Int 1)
Figure BDA0002695596540000062
SM1(22 g; 0.1mol) was dissolved in acetonitrile (110mL), potassium carbonate (16.6 g; 0.12mol) and trans-4-methoxycyclohexylamine (12.9 g; 0.1mol) were added, and the mixture was heated under reflux for 3 h. The reaction was poured into water (500mL), filtered and washed with water to give Int 1(30 g; 0.09mol) as an orange solid in 91.2% yield.
(2) Compound 4-bromo-N1Synthesis of (- (1r,4r) -4-methoxycyclohexyl) benzene-1, 2-diamine (Int 2)
Figure BDA0002695596540000071
Int 1(30 g; 0.09mol) was dissolved in tetrahydrofuran (300mL), water (100mL), aqueous ammonia (28%, 60mL) and finally sodium dithionite (62 g; 0.36mol) were added and stirred at room temperature for 16 hours. The layers were separated by settling, the aqueous phase was extracted with ethyl acetate (50 mL. times.2), the organic phases were combined, washed with saturated brine (200mL), dried over anhydrous sodium sulfate and spun dry to give Int 2(24 g; 0.08mol) in 88.2% yield.
(3) Synthesis of Compound (S) -N- (5-bromo-2- (((1r,4S) -4-methoxycyclohexyl) amino) phenyl) -6-oxopiperidine-2-carboxamide (Int 3)
Figure BDA0002695596540000072
Int 2(24 g; 0.08mol), (S) -6-oxopiperidine-2-carboxylic acid (12 g; 0.084mol) and DIPEA (12 g; 0.096mol) were added to dichloromethane (240mL), propylphosphoric anhydride (50% in EA; 61 g; 0.096mol) was added dropwise with stirring at room temperature, and after the addition was complete, stirring was continued for 3 hours at room temperature. The Int 3(27 g; 0.064mol) was obtained by column chromatography in 79% yield.
(4) Synthesis of compound (S) -6- (5-bromo-1- ((1r,4S) -4-methoxycyclohexyl) -1H-benzo [ d ] imidazol-2-yl) piperidin-2-one (Int 4)
Figure BDA0002695596540000073
To acetic acid (270mL) was added Int 3(27 g; 0.064mol) and stirred at 50 ℃ for 5 days. Removing acetic acid by spinning, adjusting the pH value to 7-8 by using saturated sodium bicarbonate aqueous solution, extracting by using dichloromethane (100mL multiplied by 3), and washing an organic phase by using saturated saline water (100 mL). After drying over anhydrous sodium sulfate, spin-drying gave Int4(20.7 g; 0.051mol) with a yield of 80%.
(5) Synthesis of compound (S) -6- (5-bromo-1- ((1r,4S) -4-methoxycyclohexyl) -1H-benzo [ d ] imidazol-2-yl) -1- (3, 4-difluorophenyl) piperidin-2-one (Int 5)
Figure BDA0002695596540000081
Pyridine (41 mL; 0.52mol), copper acetate monohydrate (12.4 g; 0.066mol) and 3, 4-difluorophenylboronic acid (29 g; 0.2mol) were added in this order to methylene chloride (200mL) in which Int4(20.7 g; 0.051mol) was dissolved, and the mixture was stirred at room temperature for 5 hours. Washing with water and washing with saturated salt water. The Int 5(20 g; 0.04mol) was obtained by column chromatography with a yield of 75.7%.
(6) Synthesis of compound (S) -1- (3, 4-difluorophenyl) -6- (5- (3, 5-dimethylisoxazol-4-yl) -1- ((1r,4S) -4-methoxycyclohexyl) -1H-benzo [ d ] imidazol-2-yl) piperidin-2-one (Int 6)
Figure BDA0002695596540000082
Int 5(20 g; 0.04mol) was added to a mixed solvent of 1, 4-dioxane (180mL) and water (30mL), followed by addition of Pd (dppf) Cl2(2.9 g; 0.004mol) and cesium carbonate (26 g; 0.08mol), under nitrogen, and stirred at 100 ℃ for 16 hours. After column chromatography, Int 6(19 g; 0.036mol) was obtained in 92.2% yield.
(7) Synthesis of compound (S) -1- (3, 4-difluorophenyl) -6- (5- (3, 5-dimethylisoxazol-4-yl) -1- ((1r,4S) -4-hydroxycyclohexyl) -1H-benzo [ d ] imidazol-2-yl) piperidin-2-one (Int 7)
Figure BDA0002695596540000091
Int 6(54 mg; 0.1mmol) was dissolved in dichloromethane (5mL), cooled to 0 ℃ in an ice bath and a solution of boron tribromide (75 mg; 0.3mmol) in dichloromethane (1mL) was added dropwise with stirring. After the addition was complete, the temperature was maintained and stirring was continued for 2 hours. The reaction was dropped into ice water. The organic phases were combined, washed with saturated brine (15mL) and dried over anhydrous sodium sulfate. Spin-dry to obtain Int 7(31 mg; 0.06mmol), 58.9% yield.
(8) Synthesis of compound (S) -1- (3, 4-difluorophenyl) -6- (5- (3, 5-dimethylisoxazol-4-yl) -1- ((1r,4S) -4-deuterated methylcyclohexyl) -1H-benzo [ d ] imidazol-2-yl) piperidin-2-one (100)
Figure BDA0002695596540000092
Int 7(31 mg; 0.06mmol) was dissolved in tetrahydrofuran (1mL), sodium hydride (60% dispersion in minor oil; 3.6 mg; 0.09mmol) was added with stirring for 2 minutes under cooling in an ice-water bath, and deuterated iodomethane (9 mg; 0.06mmol) was added. The reaction was allowed to stand at this temperature for 30 minutes, poured into ice water (5mL), extracted with methylene chloride (5 mL. times.3), washed with saturated brine (15mL), and dried over anhydrous sodium sulfate. Column chromatography gave compound 100(26 mg; 0.048mmol) in 81.2% yield. Mass (M + H) 538.3. H NMR (400MHz, DMSO) δ 7.79(d, J ═ 8.6Hz,1H),7.70(s,1H), 7.41-7.30 (m,2H),7.15(d, J ═ 8.5Hz,1H),7.06(s,1H),5.77(s,1H),4.38(s,1H),3.40(s,2H),2.56(s,2H),2.41(s,3H),2.27(d, J ═ 22.7Hz,4H),2.18(d, J ═ 13.8Hz,2H),2.01(d, J ═ 14.7Hz,3H),1.80(s,2H), 1.42-1.34 (m,2H),1.25(d, J ═ 9.2Hz,1H)
Example 2 Synthesis of Compound 101 of the invention
The synthetic route for the compound (S) -1- (3, 4-difluorophenyl) -6- (5- (3, 5-dimethylisoxazol-4-yl) -1- ((1r,4S) -4-deuterated methylcyclohexyl) -1H-benzo [ d ] imidazol-2-yl) piperidin-2-one-3, 3-didedeuterium (101) is as follows:
Figure BDA0002695596540000101
100(54 mg; 0.1mmol) was dissolved in CD3OD/D2Adding NaOD (41 mg; 1mmol) to O (3mL/0.5mL) at 10 deg.C, stirring for 12 hr, and performing rotary column chromatography to obtain compound 101(36) with yield of 70%. Mass (M + H) 540.2. H NMR (400MHz, DMSO) δ 7.79(d, J ═ 8.6Hz,1H),7.70(s,1H), 7.41-7.30 (m,2H),7.15(d, J ═ 8.5Hz,1H),7.06(s,1H),5.77(s,1H),4.38(s,1H),3.40(s,2H),2.56(s,2H),2.41(s,3H),2.01-2.27(m,7H),1.80(s,2H), 1.42-1.34 (m,2H),1.25(d, J ═ 9.2Hz,1H)
The beneficial effects of the present invention are demonstrated by the following experimental examples.
Experimental example 1 stability of hepatocyte microsome metabolism of Compound of the present invention
(1) Experimental Material
The microsomes were stored in a-80 ℃ freezer and the information is given in Table 1. Before use, the mixture is melted in a water bath at 37 ℃ and placed on ice for standby after being melted. Other reagents were purchased through local suppliers.
TABLE 1 liver microsome information
Species of species Goods number Batch number Line of Sex Supply ofBusiness support
Human being 452117 38295 N/A Mixing Corning
Mouse M1000 1910002 ICR/CD-1 Male part Xenotech
(2) Experimental procedure
Step 1: the reaction system was configured as shown in Table 2
Table 2: reaction system configuration method
Figure BDA0002695596540000102
Figure BDA0002695596540000111
Step 2: the reaction was preincubated in a water bath at 37 ℃ for 10 minutes. To the reaction system, 40. mu.L of a10 mM NADPH solution was added, and the final concentration of NADPH was 1mM each. As a negative control, 40. mu.L of ultrapure water was used in place of the NADPH solution.
And step 3: the reaction was initiated by adding 4 μ L of 200 μ M test compound or non-deuterated control compound to the reaction, with a final concentration of 2 μ M drug.
And 4, step 4: mu.L of each reaction was removed at 0, 15, 30, 45 and 60 minutes and quenched with 4-fold cold acetonitrile containing internal standards (200nM alprazolam, 200nM labetalol, 2. mu.M ketoprofen, 200nM caffeine). The samples were centrifuged at 3,220g for 45 minutes. After the centrifugation is finished, 90 mu L of supernatant and 90 mu L of ultrapure water are uniformly mixed for LC-MS/MS analysis and detection.
(3) Data analysis
All data calculations were performed by Microsoft Excel software. Peak areas were detected by extracting ion spectra. The in vitro half-life (t) of the parent drug was determined by linear fitting of the percentage disappearance of the parent drug to time1/2). Half life in vitro (t)1/2) By slope calculation:
invitrot1/2=-(0.693/k)
the in vitro half-life (t) is then determined using the following formula1/2) Conversion to in vitro clearance (in. mu.L/min/mg):
Figure BDA0002695596540000112
(4) results of the experiment
Table 3: results of stability of hepatocyte microsome metabolism by Compounds of the invention
Figure BDA0002695596540000113
Figure BDA0002695596540000121
The results show that the in vitro half-life of the compounds of the invention in mouse and human hepatocyte microsomes is significantly prolonged and the in vitro clearance rate is significantly reduced compared to non-deuterated control compounds. Indicating that the compounds of the invention have significantly improved metabolic stability over the non-deuterated control compounds.
Experimental example 2 mouse pharmacokinetics of the Compound of the present invention
(1) Experimental methods
Healthy adult ICR mice 18 (compound 9 animals per mode of administration, 3 animals per time point) were fasted overnight (free drinking water), administered by tail vein injection and gavage, respectively, blood was collected at 0.1ml and anticoagulated with EDTA-K2, plasma was separated by centrifugation at 4 ℃ for 5min, and stored at-80 ℃ for testing.
The plasma concentration-time curve is drawn and the main pharmacokinetic parameters are calculated using WinNonlin 6.3 software.
Time to peak TmaxAnd peak concentration CmaxAll adopt measured values;
area under the time curve AUCallThe value: calculating by adopting a trapezoidal method; AUCinf=AUCall+Ct/keCt is the blood concentration at the last measurable time point, keTo eliminate the rate constant;
elimination of half-life t1/2=0.693/ke
Mean residence time MRT ═ AUMC/AUC;
clearance rate CL ═ D/AUCinf(D is the administered dose);
volume V of steady state distributionss=CL x MRT。
Absolute bioavailability F ═ AUC (AUC)i.g.x Di.v.)/(AUCi.v.x Di.g.)×100%
(2) Results of the experiment
Table 4: mouse pharmacokinetic results for Compounds of the invention
Figure BDA0002695596540000131
Remarking: "IV (1 mpk)" in the table means administration by tail vein injection at a dose of 1mg per kg; PO (10mpk) indicates intragastric administration at a dose of 10 mg per kg.
As can be seen from the results in Table 4, the compounds of the present invention have a higher AUC in mice than the non-deuterated control compoundsallValues, longer elimination half-life, lower clearance, higher absolute bioavailability. Illustrative of the inventionThe substance has significantly improved pharmacokinetic properties over the non-deuterated control compound.
Experimental example 3 inhibitory Activity of the Compound of the present invention on CBP/EP300
A. Experimental methods
CBP and EP300 AlphaScreen assay:
1. preparing 1-fold concentration detection buffer
2. Preparation of the Compounds
Compounds were first diluted 1000-fold to final concentration using Precision autosampler (3-fold gradient dilution):
1) 50 μ L of compound stored at a concentration of 10mM was transferred to A2 wells of a 96-well plate using an Echo autosampler.
2) Transfer 30 μ L of DMSO to a1 well, A3 to a12 well with Precision.
3) Precision was used to transfer 15. mu.L from A2 to A3, in turn from A10 to A11. Completing 1: and 3, gradient dilution.
4) The compound plate was centrifuged at 1000 rpm for 1 minute.
3. Preparing a test board
Transfer 20nL to assay plate from each concentration of compound plate with Echo autosampler: from plate-configured a1 to plate-configured a1 and a2, and from plate-configured a2 to plate-configured A3 and a 4.
4. Bromodomain (BRD) binding detection assay
1) Preparing 2-fold protein and polypeptide solution
Proteins and polypeptides were dissolved in 1-fold concentration of assay buffer.
2) 10 μ L of protein and polypeptide solution was transferred to each well of 3 to 24 columns in the assay plate, and 10 μ L of 1-fold assay buffer was transferred to wells of 1 to 2 columns in the assay plate as a negative control.
3) The plates were centrifuged at 1000 rpm for 1 minute.
4) Incubate at room temperature for 15 minutes.
5) Acceptor and donor beads were formulated with 1-fold detection buffer to 2-fold acceptor and donor solutions.
6) Transfer 2-fold of the receptor and donor solutions to the assay plate.
Add 15. mu.L of acceptor and donor solutions and avoid light.
7) The assay plate was centrifuged at 1000 rpm for 1 minute and incubated at room temperature for 60 minutes.
5. The endpoint was read using ensspire and Alpha modes.
6. Fitting of curves
Inputting experimental data into an Excel document and obtaining the inhibition rate Inh% by using equation (1):
equation (1): inh% (Max-Signal)/(Max-Min) × 100
Wherein, Max: control with DMSO, Max: a low control with the addition of DMSO was used,
inputting the obtained data into XL-Fit software and obtaining IC by using equation (2)50The value of (c):
equation (2) Y ═ Bottom + (Top-Bottom)/(1+10^ ((Logicc))50-X)×Hill Slope))
B. Results of the experiment
Table 5: IC of each compound on CBP BRD and EP300 BRD50
Figure BDA0002695596540000141
Figure BDA0002695596540000151
The results show that the compound of the invention has obvious inhibitory activity on both CBP BRD and EP300 BRD, and the inhibitory activity on CBP BRD and EP300 BRD is obviously higher than that of the non-deuterated control compound.
Experimental example 4 inhibitory Effect of the Compound of the present invention on proliferation of prostate cancer CWR22RV1 cells
1. The experimental steps are as follows:
subculturing the prostate cancer CWR22RV1 cells with a cell culture solution, inoculating the cells with good growth state into a 96-well plate, wherein each well has 80 mu L of cells, the number of the cells in each well is 1500, and the temperature is 37 ℃ and the CO content is 5 percent2The cells were incubated overnight in a cell incubator.
② the drug is prepared into 30mM stock solution by dimethyl sulfoxide (DMSO). Immediately before use, the mixture was diluted 3 times with DMSO and then 3 times in a gradient to obtain 9 concentration gradients, and the compound was diluted 200 times with the culture medium (to ensure that the DMSO concentration in the culture system was 0.1%), and the concentration was repeated in 2 wells. mu.L of the diluted compound was added to the cell culture well (final concentration 10. mu.M, 3.3. mu.M, 1.1. mu.M …) and mixed by gentle shaking. Additional 3 negative control wells containing cells only and 3 blank control wells containing culture medium only (6 wells each containing 20. mu.L of culture medium diluted 200-fold in DMSO) were also provided.
2. The detection method comprises the following steps:
(1) after 6 days of incubation, 10. mu.L of CCK-8 was added to each well and incubation was continued for 2.5 hours at 37 ℃ in a 5% CO2 cell incubator.
(2) Absorbance (OD value) was measured at 450nm using a multifunctional microplate reader.
(3) The data were analyzed using the Dose-response-inhibition equation in the software GraphPad Prism6 to yield IC50The value is obtained.
3. Results of the experiment
IC for inhibiting activity of compound of the invention on CWR22RV1 cells50The (nM) results are shown in Table 6.
A represents IC50Less than or equal to 500 nM; b represents IC50Greater than 500nM and less than or equal to 2000 nM; c represents IC50Greater than 2000 nM.
Table 6: IC of each compound on CWR22RV1 cells50
Compound (I) IC50
Compound 100 A
Compound 101 A
As can be seen, the compound has obvious inhibitory effect, IC, on prostate cancer CWR22RV1 cells50As low as 500nM or less.
Experimental example 5 inhibitory Effect of the Compound of the present invention on proliferation of other tumor cells 1 Experimental method
Using the same method as in Experimental example 4, CWR22RV1 cells were replaced with tumor cells shown in Table 7, and IC for inhibition of activity of the compounds of the present invention on these tumor cells was tested and calculated50(nM) and the results are shown in Table 7.
2. Results of the experiment
In Table 7, A represents IC50Less than or equal to 500 nM; b represents IC50Greater than 500nM and less than or equal to 2000 nM.
Table 7: IC of Compounds on other tumor cells50
Figure BDA0002695596540000161
The compound prepared by the invention has obvious inhibition effect on the proliferation of other prostate cancer cells, leukemia cells, breast cancer cells and multiple myeloma cells. The compound of the invention has good inhibition effect on various tumors.
Experimental example 6 pharmacodynamic evaluation of the compound of the present invention in human acute myeloid leukemia cell strain MOLM-16 subcutaneous xenografted NOD
1. Cell culture
Human acute myeloid leukemia MOLM-16 cells were cultured in RPMI1640 medium containing 20% fetal bovine serum. The exponential growth phase of MOLM-16 cells were collected and PBS resuspended to appropriate concentrations for subcutaneous tumor inoculation in mice.
2. Animal model
Experimental mice were inoculated subcutaneously on the right back with 1X 107MOLM-16 cells, which are resuspended in 1:1 PBS and matrigel (0.1 ml/piece) to periodically observe the growth of tumor, until the tumor grows to the average volume of 150 (the average tumor volume is 100-3The administration was randomized and divided into groups according to the tumor size and the mouse body weight. The vehicle was given the same dose as the control group. The day of tumor cell inoculation was defined as day 0.
After the start of the administration, the body weight and tumor size of the mice were measured twice a week. Tumor volume calculation formula: tumor volume (mm)3)=1/2×(a×b2) (wherein a represents a long diameter and b represents a short diameter).
Student director (tm) (version number 3.1.399.19, supplier student system, Inc.) software was used in the experiments to collect data, including measurements of the long and short diameter of tumors and weighing of animal body weights.
3. Evaluation index
Relative tumor proliferation rate, T/C (%), i.e. the percentage value of the relative tumor volume or tumor weight of the treated and control groups at a certain time point. The calculation formula is as follows:
T/C%=TRTV/CRTV×100%
wherein, TRTV: mean Relative Tumor Volume (RTV) in treatment groups; CRTV: mean Relative Tumor Volume (RTV) of control group; RTV-Vt/V0, V0 is the tumor volume of the animal at the time of the grouping, and Vt is the tumor volume of the animal after treatment.
Or T/C% ═ TTW/CTW × 100%
Wherein, TTW: mean tumor weights at the end of treatment group experiments; CTW: mean tumor weights at the end of the control experiment.
Relative tumor inhibition, TGI (%), calculated by the formula:
TGI%=(1-T/C)×100%
where T and C are the Relative Tumor Volume (RTV) or Tumor Weight (TW) at a particular time point in the treated and control groups, respectively.
4. Results of the experiment
Table 8: efficacy of each Compound in MOLM-16 subcutaneous xenograft mouse model
Figure BDA0002695596540000171
The experimental results show that the compound has good treatment effect on human acute myeloid leukemia model animals, and the treatment effect of the compound is obviously improved compared with that of a non-deuterated compound.
In conclusion, the invention provides a deuterated benzimidazole compound with a novel structure, the compound has high selectivity on EP300/CBP, and can effectively inhibit the activity of EP300/CBP, and simultaneously, compared with a non-deuterated compound, the deuterated compound provided by the invention has remarkably improved metabolic stability and pharmacokinetic performance. In addition, the compound has excellent inhibition effect on various tumor cells including prostate cancer cells, leukemia cells, breast cancer cells and multiple myeloma cells. Therefore, the compound has good application prospect in preparing EP300/CBP inhibitors and medicines for preventing and/or treating tumors and regulating regulatory T cells.

Claims (10)

1. A compound represented by formula I, or a salt thereof, or a conformational isomer thereof, or a crystalline form thereof, or a solvate thereof:
Figure FDA0002695596530000011
wherein R is0~R25Each independently selected from H, deuterium, C1-C4Alkyl radical, C3-C6Cycloalkyl, C substituted by more than 1 deuterium1-C4Alkyl, C substituted by more than 1 deuterium3-C6A cycloalkyl group; and R is0~R25At least one group contains deuterium.
2. The compound according to claim 1, or a salt thereof, or a conformational isomer thereof, or a crystalline form thereof, or a solvate thereof, characterized in thatThe method comprises the following steps: r0~R25Each independently selected from H, deuterium, methyl, CD3(ii) a And R is0~R25In which at least one group is deuterium or CD3
3. The compound according to claim 2, or a salt thereof, or a conformational isomer thereof, or a crystalline form thereof, or a solvate thereof, characterized in that: r0、R1、R10Each independently selected from methyl or CD3,R2~R9、R11~R25Each independently selected from hydrogen or deuterium; preferably, R10Is a CD3
4. A compound according to any one of claims 1-3, or a salt thereof, or a conformational isomer thereof, or a crystalline form thereof, or a solvate thereof, characterized in that: the structure of the compound is shown as a formula II-1 or a formula II-2:
Figure FDA0002695596530000021
wherein R is0~R25The method according to any one of claims 1 to 3.
5. The compound of claim 4, or a salt thereof, or a conformational isomer thereof, or a crystalline form thereof, or a solvate thereof, wherein: the compound is one of the following compounds:
Figure FDA0002695596530000031
6. use of a compound of any one of claims 1-5, or a deuterode thereof, or a salt thereof, or a conformational isomer thereof, or a crystalline form thereof, or a solvate thereof, in the preparation of an EP300 inhibitor, a CBP inhibitor, or an EP300/CBP inhibitor.
7. Use according to claim 6, characterized in that: the EP300/CBP inhibitor is a medicament for preventing and/or treating tumors and malignant diseases of hematopoietic stem/progenitor cells of myeloid lines and regulating and controlling regulatory T cells;
preferably, the tumor is selected from hematological malignancy, gastric cancer, intestinal cancer, cervical cancer, bladder cancer, laryngeal cancer, liver cancer, lung cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma or multiple myeloma, and the myeloid hematopoietic stem/progenitor cell malignancy is leukemia;
more preferably, the lymphoma is non-hodgkin's lymphoma, diffuse large B cell lymphoma and the leukemia is acute myeloid leukemia.
8. A medicament for treating disease characterized by: the medicine is a preparation prepared by taking the compound, or a deuteron thereof, or a salt thereof, or a conformational isomer thereof, or a crystal form thereof, or a solvate thereof, as an active ingredient and adding pharmaceutically acceptable auxiliary materials.
9. A combination comprising: a compound of any one of claims 1-5, or a deutero-derivative thereof, or a salt thereof, or a conformational isomer thereof, or a crystal form thereof, or a solvate thereof, and other drugs having an anti-tumor effect, in unit preparations of the same or different specifications for simultaneous or separate administration, and a pharmaceutically acceptable carrier.
10. The combination of claim 9, wherein: the other medicines with the anti-tumor effect are chemotherapeutic medicines or radiotherapy medicines, preferably, the chemotherapeutic medicines are targeted medicines; or, the other medicine with the anti-tumor effect is selected from one or more than two of CDK4/6 inhibitor, Parp inhibitor, androgen receptor inhibitor and immune checkpoint inhibitor.
CN202011005085.4A 2019-09-27 2020-09-22 Deuterated benzimidazole compound and application thereof as EP300/CBP inhibitor Pending CN112574178A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019109257822 2019-09-27
CN201910925782 2019-09-27

Publications (1)

Publication Number Publication Date
CN112574178A true CN112574178A (en) 2021-03-30

Family

ID=75119510

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011005085.4A Pending CN112574178A (en) 2019-09-27 2020-09-22 Deuterated benzimidazole compound and application thereof as EP300/CBP inhibitor

Country Status (1)

Country Link
CN (1) CN112574178A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115919873A (en) * 2023-03-07 2023-04-07 信义核新(北京)生物科技有限公司 Application of compound and salt thereof in cancer treatment medicine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110049983A (en) * 2016-10-18 2019-07-23 细胞中心有限公司 Medical compounds
CN112351982A (en) * 2018-04-18 2021-02-09 细胞中心有限公司 Process for preparing P300 and/or CBP modulators

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110049983A (en) * 2016-10-18 2019-07-23 细胞中心有限公司 Medical compounds
CN112351982A (en) * 2018-04-18 2021-02-09 细胞中心有限公司 Process for preparing P300 and/or CBP modulators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
江文峰等: "氘代作用在药物研究中的应用", 《齐鲁药事》 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115919873A (en) * 2023-03-07 2023-04-07 信义核新(北京)生物科技有限公司 Application of compound and salt thereof in cancer treatment medicine
CN115919873B (en) * 2023-03-07 2023-09-29 信义核新(北京)生物科技有限公司 Application of compound and salt thereof in cancer treatment medicine

Similar Documents

Publication Publication Date Title
CN107428758B (en) Acrylic acid derivative, preparation method and medical application thereof
US10640512B2 (en) Imidazopyrazinamine phenyl derivative and use thereof
WO2020259683A1 (en) 2,4-disubstituted pyrimidine derivative, preparation method for same, and uses thereof
WO2021068898A1 (en) Novel kras g12c protein inhibitor, preparation method therefor, and use thereof
WO2020114482A1 (en) Isoindoline compound, and preparation method, pharmaceutical composition, and application of isoindoline compound
CN112225734B (en) KRAS G12C inhibitors and uses thereof
CN108329311B (en) Tricyclic compound as selective estrogen receptor down-regulator and application thereof
JP2021038245A (en) Pyrazole compounds as modulators of fshr and uses thereof
CN110563703B (en) Compound for inducing PARP-1 degradation based on CRBN ligand, preparation method and application
JP7128345B2 (en) Diaryl macrocyclic compound, pharmaceutical composition and use thereof
CN105246896A (en) Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
CN107814798A (en) 3 substitutional crylic acid class compounds and its production and use
EP4043458A1 (en) Ep300/cbp inhibitor
WO2017088746A1 (en) New epidermal growth factor receptor inhibitor and application thereof
CA3136224A1 (en) Condensed azines for ep300 or cbp modulation and indications therefor
CN114516832A (en) Tubulin inhibitor and preparation method and application thereof
WO2020156283A1 (en) Alkynylpyrimidine or alkynylpyridine compound, and composition and application thereof
JP2020529465A (en) Substituted pyrazolopyrimidine useful as a kinase inhibitor
CN102316738A (en) Amides as kinase inhibitors
JP2022527925A (en) Use of aromatic amine compounds in the manufacture of AR and BRD4 double inhibitors and regulators of the compounds.
WO2022206705A1 (en) Heterocyclic compound as tyk2 pseudokinase domain inhibitor, synthetic method, and use
CN112574178A (en) Deuterated benzimidazole compound and application thereof as EP300/CBP inhibitor
CN108602779A (en) The method for preparing 5,6- dihydros -6- phenyl benzo [F] isoquinolin -2- amine of substitution
CN108299420B (en) Pentacyclic compounds as selective estrogen receptor down-regulators and uses thereof
CN115477608A (en) Tubulin inhibitor and preparation method and application thereof

Legal Events

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