CN114276333A - Dihydroquinoxalines bromodomain bivalent inhibitors - Google Patents

Dihydroquinoxalines bromodomain bivalent inhibitors Download PDF

Info

Publication number
CN114276333A
CN114276333A CN202011039854.2A CN202011039854A CN114276333A CN 114276333 A CN114276333 A CN 114276333A CN 202011039854 A CN202011039854 A CN 202011039854A CN 114276333 A CN114276333 A CN 114276333A
Authority
CN
China
Prior art keywords
compound
formula
substituted
same
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202011039854.2A
Other languages
Chinese (zh)
Other versions
CN114276333B (en
Inventor
熊兵
缪泽鸿
陈丹琦
王迎庆
沈竞康
宦霞娟
吕铠铠
胡剑萍
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.)
Shanghai Institute of Materia Medica of CAS
Original Assignee
Shanghai Institute of Materia Medica of CAS
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 Shanghai Institute of Materia Medica of CAS filed Critical Shanghai Institute of Materia Medica of CAS
Priority to CN202011039854.2A priority Critical patent/CN114276333B/en
Publication of CN114276333A publication Critical patent/CN114276333A/en
Application granted granted Critical
Publication of CN114276333B publication Critical patent/CN114276333B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

The invention relates to a dihydroquinoxaline bromodomain bivalent inhibitor with a structure shown as a general formula (I), a preparation method thereof, a composition containing the same and application thereof. The bivalent inhibitor can act on two bromodomains simultaneously, has strong inhibition effect on bromodomain protein, can be used for preparing medicaments for treating a series of diseases and symptoms mediated by the bromodomain protein, and has good treatment effect on tumors.

Description

Dihydroquinoxalines bromodomain bivalent inhibitors
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a dihydroquinoxaline bromodomain (bromodomain) bivalent inhibitor, a preparation method thereof, a composition containing the same, and application of the compound in treating a series of diseases mediated by bromodomain proteins.
Background
In 2009, the Bradner topic group of Dana-Farber cancer institute reported the first small molecule inhibitor (+) -JQ1 acting on BET bromodomain proteins, and several BET family small molecule inhibitors have been reported to date. The bromodomain protein inhibitor shows certain treatment potential in preclinical evaluation and clinical research, and the experimental result greatly encourages the enthusiasm of researchers, but the experimental result faces a plurality of troubles such as low selectivity and drug resistance. Scientists have therefore continually explored new research directions in recent years, and desire to address the problems therein.
There remains a need in the art to develop novel, more clinically valuable bromodomain inhibitors that would be expected to have increased activity to effectively overcome the resistance problems of existing bromodomain protein inhibitors. The inventor of the invention synthesizes a bivalent inhibitor by a proper connection mode on the basis of the work of a previously synthesized monovalent bromodomain inhibitor, and unexpectedly finds that the activity of the bivalent inhibitor is remarkably improved.
Disclosure of Invention
The present invention provides a novel bromodomain bivalent inhibitor and a pharmaceutically usable salt thereof, a pharmaceutical composition comprising the same. The bivalent inhibitor has strong inhibition effect on bromodomain protein, and has good treatment effect on tumor.
In a first aspect of the present invention, there is provided a compound having a structure represented by general formula (i), or a pharmaceutically acceptable salt thereof:
Figure BDA0002706278290000011
wherein:
k is a bromodomain monovalent inhibitor group, which may be selected, for example, from the following groups:
Figure BDA0002706278290000021
L1is-C (═ O) -or
Figure BDA0002706278290000022
G1And G2Each independently selected from: absent, -C (═ O) -, substituted or unsubstituted C1-C4 alkylene; the substituted substituents are selected from: hydrogen, halogen, C1-C4 alkyl;
R19and R20Each independently selected from: hydrogen, substituted or unsubstituted C1-C4 alkyl, said substituted substituents being selected from the group consisting of: hydrogen, C1-C4 alkyl;
L4is selected from the group consisting ofSubstituted or unsubstituted C1-C5 alkylene, substituted or unsubstituted C3-C10 cycloalkylene, the substituted substituents being selected from: fluorine, chlorine, bromine, hydroxyl, amino, nitro, cyano, C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, etc.), C1-C6 alkoxy (e.g., methoxy, ethoxy, n-propoxy, isopropoxy, etc.);
or, L4,R19,R20And the nitrogen atom to which they are attached, together form a substituted or unsubstituted 5-10 membered heterocyclic group, said substitution being with 1-3 substituents each independently selected from: halogen, hydroxy, nitro, cyano, C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl), C1-C6 alkoxy (e.g., methoxy, ethoxy, n-propoxy, isopropoxy);
x and Y are each independently selected from: c or N; preferably C;
R1and R1' may be the same or different from each other, and each is independently selected from: hydrogen, C1-C6 alkyl (e.g., methyl, ethyl, isopropyl), C3-C8 cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl), phenyl substituted C1-C2 alkyl (e.g., benzyl);
R2and R2' may be the same or different from each other, and each is independently selected from: hydrogen, C1-C4 alkyl (e.g., methyl, ethyl), C2-C4 alkenyl-substituted C1-C4 alkyl (e.g., methyl, ethyl)
Figure BDA0002706278290000023
);
R3And R3' may be the same or different from each other, and each is independently selected from: substituted or unsubstituted phenyl, substituted or unsubstituted naphthyl, substituted or unsubstituted benzyl, said substituted substituents being selected from the group consisting of: halogen (e.g., fluorine, chlorine, bromine), hydroxy, amino, nitro, cyano, C1-C4 alkyl (e.g., methyl, ethyl, propyl, isopropyl), C1-C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy);
"" indicates that the substituent is attached thereto.
In particular, L4Selected from unsubstituted C1-C5 alkylene, unsubstituted C3-C10 cycloalkylene, or
L4,R19,R20And the nitrogen atoms to which they are attached form together
Figure BDA0002706278290000031
Figure BDA0002706278290000032
Wherein n4 and n5 are each independently selected from any integer between 0 and 4 (e.g., 0, 1,2,3, 4), which may be the same or different from each other;
n10, n11, n12 and n13 are each independently selected from any integer between 1 and 3 (e.g., 1,2, 3), which may be the same or different from each other.
More particularly, L4Is composed of
Figure BDA0002706278290000033
Or
L4,R19,R20And the nitrogen atoms to which they are attached form together
Figure BDA0002706278290000034
Figure BDA0002706278290000035
Preferably, G1And G2At least one, or G, is present1And G2Are all-C (═ O) -.
In one embodiment, L in the above general formula (I)1Selected from the following groups:
Figure BDA0002706278290000036
wherein the content of the first and second substances,
n1, n2 and n3 are each independently selected from any integer between 1 and 5 (e.g., 1,2,3,4, 5), which may be the same or different from each other;
n4, n5, n6, n7, n8 and n9 are each independently selected from any integer between 0 and 4 (e.g., 0, 1,2,3, 4), which may be the same or different from each other;
n10, n11, n12 and n13 are each independently selected from any integer between 1 and 3 (e.g., 1,2, 3), which may be the same or different from each other;
L4、R19and R20The definition of (A) is the same as in the general formula (I).
In one embodiment, the compound of formula (i) is selected from the group consisting of:
Figure BDA0002706278290000041
wherein the content of the first and second substances,
L5is selected from
Figure BDA0002706278290000042
Figure BDA0002706278290000043
Z1Is selected from-C (═ O) -or- (CH)2) n-, n is an integer of 1 to 5;
Z2is selected from-C (═ O) -or-CH2-,
R1And R1' may be the same or different from each other and are each independently selected from hydrogen, C1-C4 alkyl (e.g., methyl, ethyl, isopropyl), C3-C8 cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl), phenyl-substituted C1-C2 alkyl (e.g., benzyl); preferably selected from cyclopropyl, cyclobutyl, cyclopentyl, methyl, benzyl; preferably R1And R1' same;
R2and R2' may be the same or different from each other and are each independently selected from hydrogen, C1-C4 alkyl (e.g., methyl, ethyl, isopropyl), C2-C4 alkenyl-substituted C1-C2 alkyl (e.g., vinyl ethyl)Radical); preferably selected from methyl, vinyl ethyl; preferably R2And R2' same;
R3and R3' may be the same as or different from each other, and each is independently selected from substituted or unsubstituted phenyl, substituted or unsubstituted naphthyl, substituted or unsubstituted benzyl, preferably selected from substituted or unsubstituted phenyl, substituted or unsubstituted naphthyl; the substituted substituents are selected from halogen (e.g. fluoro, chloro, bromo), hydroxy, amino, nitro, cyano, C1-C4 alkyl (e.g. methyl, ethyl, propyl, isopropyl), C1-C4 alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy), preferably from halogen and methyl;
"" indicates that the substituent is attached thereto.
In one embodiment, in formula II, R1And R1' is cyclopentyl, R2And R2' is methyl, R3And R3' is phenyl or p-methylphenyl, Z1And Z2Is selected from-C (═ O) -or-CH2-。
In another embodiment, the compound of formula (i) is selected from the group consisting of:
Figure BDA0002706278290000051
wherein the content of the first and second substances,
L5、Z1、Z2、R1、R2and R3The same as defined in formula II.
In one embodiment, in formula III, R1Is cyclopentyl, R2Is methyl, R3Is phenyl or p-methylphenyl, Z1Is- (CH)2) n-, n is an integer of 1 to 5, Z2Is selected from-C (═ O) -or-CH2-。
In another embodiment, the compound of formula (i) is selected from the group consisting of:
Figure BDA0002706278290000052
wherein the content of the first and second substances,
L5、Z1、Z2、R1、R2、R3and R3' same as defined in formula II, R is selected from halogen.
In one embodiment, in formula IV, R1Is cyclopentyl, R2Is methyl, R3And R3' is phenyl or p-methylphenyl, Z1And Z2Is selected from-C (═ O) -or-CH2-。
In another embodiment, the compound of formula (i) is selected from the group consisting of:
Figure BDA0002706278290000053
wherein R is1、R2And R3The same as defined in formula II;
L5the same as defined in formula II, or absent;
Z1as defined in formula II, or is absent or-CH2-C(=O)-,
Z2The same as defined in formula II.
In one embodiment, in formula V, R1Is cyclopentyl, R2Is methyl, R3Is phenyl, L5Is absent or is
Figure BDA0002706278290000061
Z1Is absent or is-CH2-C(=O)-,Z2is-C (═ O) -.
In another embodiment, the compound of formula (i) is selected from the group consisting of:
Figure BDA0002706278290000062
wherein the content of the first and second substances,
R1、R2and R3The same as defined in formula II;
L5the same as defined in formula II, or absent;
Z1as defined in formula II, or is absent or-CH2-C(=O)-,
Z2The same as defined in formula II.
In one embodiment, in formula VI, R1Is cyclopentyl, R2Is methyl, R3Is phenyl, L5Is absent or is
Figure BDA0002706278290000063
Z1Is absent or is-CH2-C(=O)-,Z2is-C (═ O) -.
In one embodiment, the compound of formula (i) is selected from:
Figure BDA0002706278290000064
Figure BDA0002706278290000071
Figure BDA0002706278290000081
in the present invention, the pharmaceutically acceptable salt may be, for example, a sulfate, phosphate, hydrochloride, hydrobromide, acetate, oxalate, citrate, succinate, gluconate, tartrate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, benzoate, lactate, maleate, lithium salt, sodium salt, potassium salt or calcium salt.
In another aspect of the present invention, there is provided a pharmaceutical composition comprising one or more selected from the above-mentioned compounds described in the present invention and pharmaceutically acceptable salts thereof. The pharmaceutical composition optionally includes one or more pharmaceutical excipients. The pharmaceutical excipients include, for example, carriers, fillers, excipients, diluents, solvents, surfactants, binders, flavoring agents, sweeteners, sustained-release agents, boosters, lubricants, coating agents, antioxidants, preservatives, flavors, and the like, but are not limited thereto, and can be appropriately selected by those skilled in the art according to needs, such as formulation type, function, and the like. The pharmaceutical composition of the present invention may be prepared into various dosage forms such as tablets, pills, powders, injections, solutions, syrups, tinctures, capsules, sustained-release preparations, gels, drops, sprays, aerosols, etc. as required, but is not limited thereto.
A further aspect of the invention provides the use of a compound according to the invention, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to the invention, for the preparation of an inhibitor of a bromodomain recognition protein; or for the preparation of a medicament for the prevention and/or treatment of a disease associated with a bromodomain recognition protein mediated disease.
A further aspect of the invention provides a compound according to the invention, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to the invention, for use in the preparation of an inhibitor of a bromodomain recognition protein; or for the prevention and/or treatment of related diseases mediated by bromodomain recognition proteins.
In a further aspect, the present invention provides a method of inhibiting a bromodomain recognition protein, or a method of preventing and/or treating a related disease mediated by a bromodomain recognition protein, comprising administering to a subject in need thereof a compound according to the present invention or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to the present invention.
In one embodiment, the disease associated mediated by a bromodomain recognition protein is selected from the group consisting of: malignant tumor, immunological disease, cardiovascular system disease, virus infection, neurodegenerative disease or inflammation.
In another embodiment, the malignancy is selected from: acute lymphocytic leukemia, acute myelogenous leukemia, B-cell chronic lymphocytic leukemia, chronic myelomonocytic leukemia, testicular nucleoprotein midline cancer, small cell lung cancer, non-small cell lung cancer, B-cell lymphoma, prostate cancer, gastric cancer, colorectal cancer, renal cancer, liver cancer, breast cancer, pancreatic cancer.
Definition of terms:
unless otherwise indicated, the following terms used in the present patent specification and claims have the meanings discussed below.
C1-C6 means having 1-6 carbon atoms, C3-C10 means having 3-10 carbon atoms, and so on.
5-8 ring has 5-8 atoms, 5-10 ring has 5-10 atoms, and so on.
"alkyl" refers to a saturated aliphatic hydrocarbon group, which may be branched or straight chain alkyl.
"cycloalkyl" refers to an all-carbon monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
"alkoxy" means-O- (alkyl), such as methoxy, ethoxy;
"aryl" refers to an all-carbon monocyclic or fused ring polycyclic group having an intact conjugated pi-electron system, examples of aryl being, but not limited to, phenyl, naphthyl, and the like;
"heterocyclyl" means a monocyclic, spiro, bridged or fused ring containing one, two, three, four or five ring heteroatoms selected from N, O, S, the remaining ring atoms, if present as C, such rings may also have one or more double bonds, but such rings do not have a fully conjugated pi-electron system;
"heteroaryl" refers to a monocyclic or fused ring containing one, two, three or four ring heteroatoms selected from N, O, S, the remaining ring atoms, if present, being C, and, in addition, having an intact conjugated pi-electron system.
A compound of formula I
In the present invention, the compound represented by the general formula (I), the compound represented by the formula I, and the compound represented by the formula I all refer to a bromodomain protein bivalent inhibitor having the following structure:
Figure BDA0002706278290000091
the compounds of the present invention of the above structure may exist as stereoisomers (including enantiomers and diastereomers), solvates, hydrates, and crystalline forms, for example
Figure BDA0002706278290000092
Can be in R configuration, S configuration or racemic form. Such stereoisomers, prodrugs, solvates, hydrates and crystal forms are included within the scope of the compounds of the present invention.
The compounds of the invention may contain asymmetric or chiral centers and may therefore exist in different stereoisomeric forms. All stereoisomeric forms of the compounds of the present invention, including, but not limited to, optical isomers (including diastereomers and enantiomers), atropisomers, geometric isomers (cis-trans isomers), conformational isomers, and mixtures thereof (e.g., racemic mixtures), are included within the scope of the present invention.
The compounds of the invention may also exist in different tautomeric forms, all of which are included within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that are interconverted via a low energy barrier.
The compounds of the present invention may exist in unsolvated forms as well as solvated forms containing pharmaceutically acceptable solvents such as water, ethanol, and the like, including solvated as well as unsolvated forms.
Prodrugs may also be present in the compounds of the present invention which are converted in vivo to the compounds of the present invention and, therefore, such prodrugs are also included within the scope of the compounds of the present invention.
The compounds of the present invention may also form protein-targeted degradation conjugates (PROTACs) or antibody-drug conjugates (ADCs), and thus these PROTACs and ADCs are also included in the scope of the compounds of the present invention.
The compound shown in the general formula (I) has basic groups, so that the compound can form pharmaceutically acceptable salts (namely pharmaceutically acceptable salts) with inorganic acids or organic acids, including pharmaceutically acceptable acid addition salts. Pharmaceutically acceptable salts can be obtained by treating the free base of the compound of formula (I) with an inorganic or organic acid. Such as hydrochloric acid, hydrobromic acid, phosphoric acid and sulfuric acid, and such organic acids as ascorbic acid, nicotinic acid, citric acid, tartaric acid, lactic acid, maleic acid, malonic acid, fumaric acid, oxalic acid, malic acid, glycolic acid, succinic acid, propionic acid, acetic acid, methanesulfonic acid, trifluoromethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid and the like, but are not limited thereto.
The compound of the invention, pharmaceutically acceptable salts, solvates, prodrugs, protein targeted degradation conjugates or antibody drug conjugates thereof may also exist in one or more crystal forms having similar or improved properties, and therefore, these crystal forms are also included in the scope of the compound of the invention.
The invention also encompasses isotopically-labeled compounds of the invention, which are identical to those recited herein, except for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine and chlorine, such as:2hydrogen, hydrogen,3Hydrogen, hydrogen,11Carbon, carbon,13Carbon, carbon,14Carbon, carbon,13Nitrogen, nitrogen,15Nitrogen, nitrogen,15Oxygen, oxygen,17Oxygen, oxygen,18Oxygen, oxygen,31Phosphorus, phosphorus,32Phosphorus, phosphorus,35Sulfur, sulfur,18Fluorine,123Iodine,125Iodine and36chlorine.
Certain isotopically-labelled compounds of the invention (e.g. with3H and14c-labeled those) for compound and/or substrate tissue distribution assays. Particular preference is given to fluorination (i.e.3H) And carbon-14 (i.e.14C) Isotopes because they are easy to prepare and detect. Also, heavier isotopes such as deuterium (i.e., deuterium)2H) Substitution may provide for some of the resulting greater metabolic stabilityTherapeutic advantages (e.g., increased in vivo half-life or reduced dosage requirements) may be preferred in some circumstances. Positron emitting isotopes, e.g.15O、13N、11C and18f was used in Positron Emission Tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the schemes and/or in the examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred embodiments and materials described herein are intended to be exemplary only.
Preparation method
For purposes of illustration, the reaction schemes shown below provide possible routes for the synthesis of the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the examples section below. The compounds of the invention may be synthesized by methods including those well known in the chemical arts, particularly in light of the description of the invention. The starting materials may be obtained from commercial sources or prepared by methods known in the art or according to the methods described herein.
For example, the compounds of the present invention may be prepared by one of the following reaction schemes:
the reaction scheme I:
Figure BDA0002706278290000111
step a: carrying out nucleophilic substitution reaction (for example, in the presence of potassium carbonate) on the compound A and alanine to obtain a compound B;
step b: reducing compound B (for example, in the presence of sodium hydrosulfite and potassium carbonate) and closing the ring in the molecule to obtain compound C;
step c: subjecting the compound C and ketone or aldehyde to reductive amination (for example, in the presence of phenylsilane and dibutyltin dichloride) to obtain a compound D;
step d: compounds D and R2I or R2Br reaction (e.g. in the presence of sodium hydride) to give compound E;
step e: reacting compound E with pinacol bisboronate (e.g., in the presence of potassium acetate and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex) to give compound F;
step f: compound F and
Figure BDA0002706278290000112
by coupling reaction (e.g. in sodium bicarbonate and [1, 1' -bis (diphenylphosphino) ferrocene)]Palladium dichloride dichloromethane complex) to obtain a compound G;
step g: hydrolysis of compound G (e.g., in the presence of lithium hydroxide monohydrate) affords compound H;
step h: compound H and
Figure BDA0002706278290000121
condensation reaction (e.g. in the presence of HATU) gives compound I.
Reaction scheme two:
Figure BDA0002706278290000122
step a: compound A and diamine linker protected by unilateral tert-butyloxycarbonyl group
Figure BDA0002706278290000123
Condensing under HATU condition to obtain a compound B;
step b: removing tert-butyloxycarbonyl (Boc) from the compound B in HCl/dioxane solution to obtain a compound C;
step c: condensing the compound C and an optical isomer of the compound A under the HATU condition to obtain a compound D;
the reaction route is three:
Figure BDA0002706278290000124
step a: compound A in LiAlH4Reducing under the action of (1) to obtain a compound B;
step b: compound B and
Figure BDA0002706278290000125
in sodium bicarbonate and [1, 1' -bis (diphenylphosphino) ferrocene]Performing coupling reaction on palladium dichloride and dichloromethane complex to obtain a compound C;
step c: reacting the compound C with MsCl to obtain a compound D;
step d: compound D and
Figure BDA0002706278290000131
nucleophilic substitution under the action of DIPEA to obtain a compound E;
the reaction route is four:
Figure BDA0002706278290000132
step a: hydrolyzing the compound A to obtain a compound B;
step b: compound B and
Figure BDA0002706278290000133
condensing under HATU condition to obtain a compound C;
step c: removing tert-butyloxycarbonyl (Boc) from the compound C in HCl/dioxane solution to obtain a compound D;
step d: compound D and
Figure BDA0002706278290000134
obtaining a compound E through nucleophilic substitution reaction;
step e: compound E with
Figure BDA0002706278290000135
In the presence of sodium bicarbonate and [11' -bis (diphenylphosphino) ferrocene]Performing coupling reaction on palladium dichloride and dichloromethane complex to obtain a compound F;
reaction scheme five:
Figure BDA0002706278290000136
step a: and carrying out nucleophilic substitution reaction on the compound A and the compound B to obtain a compound C.
In the above reaction scheme, each substituent is defined as the same as the corresponding substituent in the aforementioned general formula (I).
Detailed Description
The invention is further illustrated by the following examples. The following examples are intended only to illustrate embodiments of the present invention. It is to be understood that the embodiments of the invention are not limited to the specific details of the following examples, since other variations will be apparent to those of ordinary skill in the art in view of the present disclosure.
The structure of the compound is determined by nuclear magnetic resonance1H-NMR) and/or Mass Spectrometry (MS). NMR was measured using a Mercury-400 NMR spectrometer manufactured by Varian corporation, and the solvent used was deuterated chloroform (CDC 1)3) Deuterated methanol (CD)3OD), deuterated dimethyl sulfoxide (DMSO-d)6) Or deuterated acetonitrile (CD)3CN), TMS as internal standard. MS was measured using a Thermo Finnigan LCQ-Deca XP model (ESI) liquid chromatography-mass spectrometer.
Example 1
Figure BDA0002706278290000141
Step a 2-fluoro-4-bromonitrobenzene (30g, 136.4mmol), D-alanine (15.8g, 177.3mmol), potassium carbonate (24.5g, 177.3mmol) were dissolved in 500mL ethanol: water 3: 1 at 80 ℃ for 8 hours, monitoring the reaction by a TLC plate, cooling to room temperature after the reaction is finished, evaporating the solvent to dryness, dissolving in water, adjusting the pH to 1-2 by 1N HCl, separating out a large amount of yellow solid, filtering, washing the solid by 500mL of Petroleum Ether (PE), and drying in a vacuum drying oven to obtain 34.7g of yellow solid, namely the compound 1B with the yield of 88%.
1H NMR(400MHz,CDC13))δ8.35(d,J=6.9Hz,1H),8.06(d,J=9.1Hz,1H),6.90(s,1H),6.85(d,J=9.2Hz,1H),4.33(p,J=7.0Hz,1H),1.67(d,J=7.0Hz,3H).
And step B, dissolving the compound 1B (34.7g, 120.0mmol) and potassium carbonate (16.6g, 120.0mmol) in 500mL of water, gradually adding sodium hydrosulfite (105g, 600.0mmol) in batches, reacting at 60 ℃ for 8h, generating a large amount of solid in the reaction liquid, monitoring the reaction by using a TLC plate, cooling to room temperature after the reaction is finished, filtering, washing the solid with 500mL of water, and drying in a vacuum drying oven to obtain 11g of white solid, namely the compound 1C, wherein the yield is 38%.
1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),6.82(d,J=2.1Hz,1H),6.74(dd,J=8.2,2.1Hz,1H),6.65(d,J=8.3Hz,1H),6.29(s,1H),3.87–3.77(m,1H),1.25(d,J=6.6Hz,3H).
Step C compound 1C (5.7g, 23.64mmol), phenylsilane (8.5mL, 70.93mmol), cyclopentanone (6.3mL, 70.93mmol) and dibutyltin dichloride (11g, 35.46mmol) were dissolved in 100mL Tetrahydrofuran (THF), reacted overnight at room temperature, monitored by TLC plates, the solvent evaporated after the reaction was complete, silica gel purified by flash chromatography with a gradient of Ethyl Acetate (EA)/Petroleum Ether (PE) of 10-30% to give 6.8g of a colorless oily liquid, compound 1D, 93.2% yield.
1H NMR(400MHz,CDC13)δ9.68(s,1H),6.92(d,J=1.9Hz,1H),6.88(dd,J=8.3,2.0Hz,1H),6.69(d,J=8.2Hz,1H),4.10(q,J=6.8Hz,1H),3.88–3.75(m,1H),2.08-1.94(m,2H),1.78-1.55(m,6H),1.14(d,J=6.8Hz,3H).
Step D, dissolving compound 1D (7.2g, 23.3mmol) in anhydrous 20mL of Dimethylformamide (DMF), adding NaH (1.9g, 46.6mmol) in portions in an ice-water bath, stirring at 0 ℃ for 30min, slowly adding methyl iodide (2.2mL, 34.9mmol), reacting at room temperature for 2h, monitoring the reaction with a TLC plate, after the reaction is finished, pouring the reaction solution into 200mL of ice water for quenching, extracting with 200mL of 2 × Dichloromethane (DCM), combining the organic layers, washing with 350mL of saturated common salt for 1 time, drying with anhydrous sodium sulfate, evaporating the solvent, purifying the organic phase silica gel sample by a flash chromatography column, and eluting with a gradient of EA/PE of 10-20% to obtain 5.8g of colorless oily liquid, namely compound 1E, with a yield of 76.8%.
1H NMR(400MHz,DMSO-d6)δ6.97(dd,J=8.4,2.1Hz,1H),6.92(d,J=2.1Hz,1H),6.78(d,J=8.5Hz,1H),4.17(d,J=6.8Hz,1H),3.81-3.72(m,1H),3.33(s,3H),2.08-1.96(m,2H),1.84-1.74(m,1H),1.72-1.58(m,5H),1.05(d,J=6.8Hz,3H).
Step E Compound 1E (5g, 15.5mmol), pinacol ester of bisboronic acid (4.4g, 17.0mmol), potassium acetate (3g, 31.0mmol) were dissolved in 100mL of 1.4-dioxane, purged with N2 for 1min, Pa (dppf) was added2Cl2(700mg, 0.775mmol), with N again2After 1min of aeration, reaction was carried out for 8h at 100 ℃, the reaction was monitored by TLC plates, after the reaction was completed the solvent was evaporated to dryness, the silica gel sample was purified by flash chromatography using a gradient EA/PE of 10-33% to give 4.9g of a colorless oily liquid, compound 1F, in 85.4% yield.
1H NMR(400MHz,CDC13)δ7.38(dd,J=7.9,1.3Hz,1H),7.30(d,J=1.2Hz,1H),6.97(d,J=8.0Hz,1H),4.17–4.09(m,1H),3.92(p,J=7.5Hz,1H),3.39(s,3H),2.08–1.99(m,2H),1.83–1.75(m,1H),1.75–1.50(m,5H),1.37(s,12H),1.04(d,J=6.9Hz,3H).
Step F Compound 1F (5.9g, 15.93mmol), ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate (5.4g, 17.41mmol), aqueous sodium bicarbonate (2.7g, 32.14mmol) were dissolved in 100mL THF, purged with N2 for 1min, Pa (dppf) was added2Cl2(1.3g, 1.59mmol), and N2Ventilating for 1min, reacting at 80 deg.C for 8h, monitoring reaction with TLC plate, and after reaction, 200mLH2O/200mL × 2DCM extraction, combined organic layers, washed 1 time with 200mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a gradient EA/PE of 10-33% to give 1.4G of a white solid, compound 1G, 18.6% yield.
1H NMR(400MHz,CDC13)δ7.33–7.24(m,4H),7.20(dd,J=8.3,1.9Hz,1H),6.92(d,J=8.4Hz,1H),6.87(d,J=1.9Hz,1H),4.55(qd,J=7.2,1.0Hz,2H),4.15(q,J=6.8Hz,1H),3.47(q,J=7.3Hz,1H),3.37(s,3H),2.42(s,3H),1.85–1.76(m,3H),1.73–1.66(m,1H),1.64–1.51(m,4H),1.47(t,J=7.1Hz,3H),0.99(d,J=6.8Hz,3H).
Step G Compound 1G (800mg, 1.69mmol), lithium hydroxide monohydrate (280mg, 6.76mmol) was dissolved in 50mL THF: h2O is 4: 1 at room temperature for 4h, monitoring the reaction with TLC plate, and after the reaction is finished, adjusting pH to 1-2 with 1N HCl, 50mLH2O/50mL × 2EA extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purified by flash chromatography on silica gel with organic phase eluting with MeOH/DCM ═ 1-10% gradient to afford 750mg of white solid, compound 1H, yield 99%.
1H NMR(400MHz,CDC13)δ8.81(s,1H),7.30(dd,J=8.2Hz,2H),7.26–7.19(m,3H),6.93(d,J=8.2Hz,1H),6.84(s,1H),4.19(q,J=6.7Hz,1H),3.50–3.40(m,1H),3.35(s,3H),2.39(s,3H),1.84–1.74(m,1H),1.71–1.62(m,1H),1.61–1.42(m,5H),1.36–1.24(m,1H),0.98(d,J=6.8Hz,3H).8。
Step H Compound 1H (200mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) dissolved in 10mL DMF, HATU (186.3mg, 0.49mmol) added, reaction at room temperature for 2H, piperazine (25mg, 0.2mmol) added, reaction at room temperature for 8H, reaction monitored by TLC plate, after completion of reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a MeOH/DCM ═ 1-10% gradient to give 50mg of white solid, compound 1, yield 20%.
1H NMR(400MHz,DMSO-d6)δ7.43–7.33(m,8H),7.25–7.11(m,4H),6.73(dd,J=14.7,1.8Hz,2H),4.06(p,J=6.8Hz,2H),3.95–3.72(m,8H),3.40–3.30(m,2H),3.27(d,J=8.1Hz,6H),2.39(d,J=8.3Hz,6H),1.75–1.58(m,8H),1.58–1.34(m,8H),0.88(dd,J=9.9,6.7Hz,6H).
LC-MS(ESI)[M+H]+941.68; retention time 6.019min, HPLC purity 98.927%.
Example 2
Figure BDA0002706278290000161
The compound of example 2 was prepared in the same manner as in example 1 except that ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate was replaced with ethyl 5-bromo-1- (2, 4-dimethylphenyl) -1,2, 4-triazole-3-carboxylate in step f of example 1, and the product was a white solid with a final yield of 33%.
1H NMR(400MHz,DMSO-d6)δ7.38(dd,J=14.6,8.0Hz,2H),7.33–7.08(m,8H),6.73(d,J=11.7Hz,2H),4.11–4.00(m,2H),3.85(m,8H),3.33(m,2H),3.26(d,J=8.1Hz,6H),2.37(d,J=8.1Hz,6H),1.93(s,3H),1.92(s,3H),1.68(dd,J=27.9,13.6Hz,2H),1.46(d,J=16.1Hz,10H),1.22(d,J=17.8Hz,4H),0.87(dd,J=9.6,6.6Hz,6H).
LC-MS(ESI)[M+H]+969.6; retention time 14.599min, HPLC purity 97.9%.
Example 3
Figure BDA0002706278290000162
The compound of example 3 was prepared in the same manner as in example 1 except that ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate was replaced with ethyl 5-bromo-1- (β -naphthyl) -1,2, 4-triazole-3-carboxylate in step f of example 1, and the product was a white solid with a final yield of 53%.
1H NMR(400MHz,DMSO-d6)δ8.23–7.94(m,8H),7.70–7.53(m,6H),7.36–7.24(m,2H),7.17(dd,J=16.0,8.4Hz,2H),6.70–6.58(m,2H),4.03–3.89(m,6H),3.89–3.68(m,4H),3.26(d,J=10.3Hz,6H),3.04(dt,J=15.8,8.0Hz,2H),1.48–1.12(m,4H),1.10–0.89(m,4H),0.81(dd,J=12.2,6.7Hz,6H).
LC-MS(ESI)[M+H]+1013.4, and the like; retention time 13.903min, HPLC purity 98.9%.
Example 4
Figure BDA0002706278290000171
The compound of example 4 was prepared in the same manner as in example 1 except that benzaldehyde was used instead of cyclopentanone in step c of example 1, and the product was a white solid with a final yield of 10%.
1H NMR(400MHz,DMSO-d6)δ7.40–7.15(m,18H),7.10(dd,J=14.6,8.5Hz,2H),6.99–6.80(m,4H),4.29–4.08(m,4H),3.99–3.86(m,3H),3.85–3.68(m,3H),3.57(s,4H),3.29(d,J=8.2Hz,6H),2.37(d,J=8.3Hz,6H),0.99(dd,J=11.6,6.7Hz,6H).
LC-MS(ESI)[M+H]+985.4; retention time 9.516min, HPLC purity 92.35%.
Example 5
Figure BDA0002706278290000172
The compound of example 5 was prepared in the same manner as in example 1 except that N, N' -dimethylethylenediamine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 21%.
1H NMR(400MHz,DMSO-d6)δ7.41–7.32(m,7H),7.30–7.21(m,3H),7.11–7.00(m,2H),6.73–6.61(m,2H),4.04(dd,J=9.6,6.7Hz,2H),3.30(m,2H),3.27(d,J=4.1Hz,3H),3.22(d,J=7.2Hz,3H),3.15(d,J=8.7Hz,3H),3.00(m,3H),2.39–2.31(m,6H),2.01(q,J=6.9,6.4Hz,2H),1.63(d,J=28.8Hz,4H),1.24(m,4H),1.39(d,J=22.8Hz,10H),0.93–0.75(m,6H).
LC-MS(ESI)[M+H]+943.68; retention time 5.780min, HPLC purity 100%.
Example 6
Figure BDA0002706278290000181
The compound of example 6 was prepared in the same manner as in example 1 except that homopiperazine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 26%.
1H NMR(400MHz,DMSO-d6)δ7.43–7.32(m,10H),7.26(d,J=8.2Hz,2H),6.78–6.71(m,2H),4.11–4.02(m,2H),3.96–3.87(m,2H),3.85–3.70(m,8H),3.30–3.24(m,6H),2.38(d,J=6.4Hz,6H),1.66(dd,J=35.7,15.4Hz,5H),1.52–1.33(m,11H),0.93–0.80(m,8H).
LC-MS(ESI)[M+H]+955.75; retention time 5.939min, HPLC purity 97.435%.
Example 7
Figure BDA0002706278290000182
The compound of example 7 was prepared in the same manner as in example 1 except that trans-2, 5-dimethylpiperazine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 24%.
1H NMR(400MHz,DMSO-d6)δ7.39(ddd,J=13.7,3.7,2.6Hz,8H),7.24–7.11(m,4H),6.80–6.69(m,2H),4.08(q,J=7.8,7.0Hz,4H),3.33(m,18H),2.43–2.35(m,6H),1.67(d,J=41.2Hz,5H),1.55–1.15(m,17H),0.94–0.77(m,6H).
LC-MS(ESI)[M+H]+969.76; retention time 6.167min, HPLC purity 95.513%.
Example 8
Figure BDA0002706278290000183
The compound of example 8 was prepared in the same manner as in example 7 except that ethyl 5-bromo-1- (2, 4-dimethylphenyl) -1,2, 4-triazole-3-carboxylate was replaced with ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate in step f of example 7, and the product was a white solid with a final yield of 69%.
1H NMR(400MHz,DMSO-d6)δ7.42–7.09(m,10H),6.79–6.69(m,2H),4.89(d,J=65.6Hz,2H),4.05(h,J=7.7,7.2Hz,4H),3.62(dd,J=25.0,14.3Hz,2H),3.33(m,2H),3.29–3.18(m,6H),2.38(d,J=7.4Hz,6H),1.96(dt,J=29.0,8.9Hz,7H),1.78–1.61(m,2H),1.46(d,J=14.7Hz,8H),1.35(t,J=7.5Hz,2H),1.31–1.20(m,9H),0.92–0.74(m,6H).
LC-MS(ESI)[M+H]+997.6; retention time 17.534min, HPLC purity 97.6%.
Example 9
Figure BDA0002706278290000191
The compound of example 9 was prepared in the same manner as in example 7 except that cyclopentanone was replaced with cyclobutanone in step c of example 7, and the product was a white solid with a final yield of 35%.
1H NMR(400MHz,DMSO-d6)δ7.39(ddd,J=14.4,4.0,2.6Hz,8H),7.18–7.09(m,4H),6.70–6.53(m,2H),3.96(s,6H),3.33(s,12H),3.28(s,6H),2.51(p,J=1.8Hz,6H),2.39(dd,J=7.3,2.8Hz,4H),1.58(q,J=12.7,8.6Hz,3H),1.46–1.13(m,6H),0.93–0.71(m,3H).
Example 10
Figure BDA0002706278290000192
The compound of example 10 was prepared in the same manner as in example 9 except that ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate was replaced with ethyl 5-bromo-1- (m-chlorophenyl) -1,2, 4-triazole-3-carboxylate in step f of example 9, and the product was a white solid with a final yield of 51%.
1H NMR(400MHz,DMSO-d6)δ7.76–7.55(m,5H),7.49(d,J=17.4Hz,3H),7.23–7.12(m,4H),6.62–6.52(m,2H),4.16–3.82(m,3H),3.58–3.40(m,3H),3.29(d,J=7.7Hz,6H),2.13(m,2H),1.96(dq,J=19.1,9.5,8.6Hz,2H),1.76(m,3H),1.59(d,J=11.4Hz,2H),1.31(ddt,J=31.7,13.6,6.8Hz,9H),0.84(dd,J=10.3,6.9Hz,6H).
LC-MS(ESI)[M+H]+981.4; retention time 11.954min, HPLC purity 98.5%.
Example 11
Figure BDA0002706278290000193
The compound of example 11 was prepared in the same manner as in example 1 except that 1, 3-propanediamine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 37%.
1H NMR(400MHz,DMSO-d6)δ8.77(t,J=6.1Hz,2H),7.37(d,J=1.6Hz,8H),7.23(dd,J=8.3,1.8Hz,2H),7.16(d,J=8.5Hz,2H),6.73(d,J=1.8Hz,2H),4.06(q,J=6.7Hz,2H),3.33(m,6H),3.27(s,6H),2.38(s,6H),1.48(dd,J=25.5,15.4Hz,9H),1.24(d,J=4.0Hz,9H),0.93–0.75(m,8H).
LC-MS(ESI)[M+H]+929.40; retention time 5.873min, HPLC purity 99.686%.
Example 12
Figure BDA0002706278290000201
The compound of example 12 was prepared in the same manner as in example 1 except that trans-1, 2-cyclohexanediamine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 42%.
1H NMR(400MHz,DMSO-d6)δ8.37(t,J=8.9Hz,2H),7.42–7.29(m,8H),7.20(dd,J=8.4,1.7Hz,2H),7.13(dd,J=8.5,5.5Hz,2H),6.69(t,J=1.4Hz,2H),4.13–3.89(m,2H),3.33(m,4H),2.36(s,6H),1.78–1.54(m,6H),1.53–1.32(m,16H),1.32–1.07(m,8H),0.86(dt,J=9.9,6.9Hz,6H).
LC-MS(ESI)[M+H]+969.69; retention time 6.611min, HPLC purity 95.199%.
Example 13
Figure BDA0002706278290000202
The compound of example 13 was prepared in the same manner as in example 1 except that ethylenediamine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 29%.
1H NMR(400MHz,DMSO-d6)δ8.77(s,2H),7.42–7.29(m,8H),7.20(dd,J=8.3,1.8Hz,2H),7.15(d,J=8.4Hz,2H),6.73(d,J=1.8Hz,2H),4.05(q,J=6.7Hz,2H),3.51(d,J=3.6Hz,4H),3.33(m,2H),3.27(s,6H),2.38(s,6H),1.76–1.56(m,4H),1.56–1.33(m,8H),1.21(d,J=23.9Hz,4H),0.87(d,J=6.7Hz,6H).
LC-MS(ESI)[M+H]+915.67; retention time 5.829min, HPLC purity 98.751%.
Example 14
Figure BDA0002706278290000203
The compound of example 14 was prepared in the same manner as in example 1 except that (R) -5-methyl-homopiperazine was used instead of piperazine in step h of example 1, and the product was a white solid with a final yield of 17%.
1H NMR(400MHz,DMSO-d6)δ7.39–7.33(m,8H),7.17(dp,J=8.2,2.7,2.3Hz,4H),6.70(q,J=2.4Hz,2H),4.93–4.70(m,1H),4.68–4.47(m,2H),4.37(ddd,J=26.1,13.9,6.0Hz,1H),4.24–4.11(m,1H),4.10–3.99(m,4H),3.27(q,J=3.9Hz,4H),3.23(m,6H),2.42–2.35(m,6H),1.65(d,J=43.7Hz,2H),1.28–1.12(m,16H),0.94–0.82(m,9H).
LC-MS(ESI)[M+H]+969.81; retention time 5.897min, HPLC purity 99.089%.
Example 15
Figure BDA0002706278290000211
The compound of example 15 was prepared in the same manner as in example 1 except that (R) - (-) -2-methylpiperazine was used in place of piperazine in step h of example 1, and the product was a white solid with a final yield of 28%.
1H NMR(400MHz,DMSO-d6)δ7.44–7.30(m,8H),7.28–7.09(m,4H),6.81–6.66(m,2H),4.86(d,J=61.7Hz,1H),4.68–4.14(m,3H),4.12–4.00(m,2H),3.71–3.45(m,1H),3.27(d,J=7.6Hz,6H),3.23–3.12(m,2H),3.02(t,J=12.5Hz,2H),2.38(d,J=7.0Hz,6H),1.66(dd,J=37.5,10.3Hz,2H),1.56–1.32(m,10H),1.32–1.09(m,7H),0.89(tt,J=6.9,2.5Hz,6H).
LC-MS(ESI)[M+H]+955.72; retention time 6.073min, HPLC purity 98.653%.
Example 16
Figure BDA0002706278290000212
The compound of example 16 was prepared in the same manner as in example 1 except that 2, 6-diazaspiro [3.3] heptane was used in place of piperazine in step h of example 1, and the product was a white solid with a final yield of 71%.
1H NMR(400MHz,DMSO-d6)δ7.37(s,8H),7.25–7.11(m,4H),6.71(s,2H),4.78(s,4H),4.32(s,4H),4.06(d,J=7.8Hz,4H),3.57(s,6H),2.39(s,6H),1.45(d,J=11.3Hz,8H),1.32–1.11(m,8H),0.89(d,J=6.9Hz,6H).
LC-MS(ESI)[M+H]+953.76; retention time 9.989min, HPLC purity 98.35%.
Example 17
Figure BDA0002706278290000213
The compound of example 17 was prepared in the same manner as in example 16 except that ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate was replaced with ethyl 5-bromo-1- (β -naphthyl) -1,2, 4-triazole-3-carboxylate in step f of example 16, and the product was a white solid with a final yield of 24%.
1H NMR(400MHz,DMSO-d6)δ8.17–8.09(m,3H),8.03(dd,J=24.6,7.7Hz,4H),7.65(t,J=6.5Hz,5H),7.55(dt,J=8.6,1.9Hz,2H),7.32(d,J=8.3Hz,2H),7.17(d,J=8.4Hz,2H),6.61(s,2H),4.84(s,4H),4.35(d,J=2.8Hz,4H),3.96(q,J=6.6Hz,2H),3.26(s,6H),3.02(t,J=7.8Hz,2H),1.44–1.27(m,5H),1.27–1.12(m,6H),1.11–0.84(m,7H),0.81(d,J=6.7Hz,6H).
LC-MS(ESI)[M+H]+1025.5; retention time 12.582min, HPLC purity 98.3%.
Example 18
Figure BDA0002706278290000221
The compound of example 18 was prepared in the same manner as in example 1 except that piperazine was replaced with 2, 7-diazaspiro [4.4] nonane in step h of example 1, and the product was a white solid with a final yield of 17%.
1H NMR(400MHz,DMSO-d6)δ7.37(d,J=9.8Hz,8H),7.25–7.17(m,2H),7.14(td,J=9.2,8.6,3.2Hz,2H),6.77–6.68(m,2H),4.11–3.94(m,2H),3.96–3.82(m,2H),3.75–3.48(m,4H),3.29–3.21(m,10H),2.44–2.31(m,6H),2.09–1.88(m,4H),1.75–1.56(m,4H),1.44(d,J=11.6Hz,8H),1.31–1.06(m,4H),0.94–0.83(m,6H).
LC-MS(ESI)[M+H]+981.92; retention time 10.833min, HPLC purity 98.5%.
Example 19
Figure BDA0002706278290000222
The compound of example 19 was prepared in the same manner as in example 1 except that in step c of example 1, aniline N and amide N were simultaneously methylated with 2 equivalents of methyl iodide to give compound 19A and step D was not performed, and compound 19A was used in place of compound 1D in step e, and the product was a white solid with a final yield of 70%.
1H NMR(400MHz,DMSO-d6)δ7.43–7.30(m,8H),7.07(dd,J=11.4,8.4Hz,2H),6.99–6.90(m,2H),6.74(dd,J=16.8,1.9Hz,2H),4.02(p,J=6.9Hz,2H),3.96–3.72(m,8H),3.26(d,J=7.5Hz,6H),2.60(d,J=14.2Hz,6H),2.39(d,J=8.7Hz,6H),0.96(dd,J=10.2,6.7Hz,6H).
LC-MS(ESI)[M+H]+833.4; retention time 4.448min, HPLC purity 97.4%.
Example 20
Figure BDA0002706278290000231
The compound of example 20 was prepared in the same manner as in example 19 except that trans-2, 5-dimethylpiperazine was used instead of piperazine in step h of example 19, and the product was a white solid with a final yield of 27%.
1H NMR(400MHz,DMSO-d6)δ7.44–7.27(m,8H),7.12–7.02(m,2H),6.95(dddd,J=15.1,10.8,6.0,2.2Hz,2H),6.79–6.69(m,2H),4.04(q,J=7.1Hz,4H),3.33(m,7H),3.27(dd,J=7.0,1.9Hz,6H),2.64–2.56(m,6H),2.40(d,J=7.7Hz,6H),1.42–1.22(m,6H),0.96(ddd,J=8.2,6.7,1.7Hz,6H).
LC-MS(ESI)[M+H]+861.5; retention time 5.379min, HPLC purity 98.9%.
Example 21
Figure BDA0002706278290000232
The compound of example 21 was prepared in the same manner as in example 19 except that piperazine was replaced with 2, 7-diazaspiro [4.4] nonane in step h of example 19, and the product was a white solid with a final yield of 81%.
1H NMR(400MHz,DMSO-d6)δ7.41–7.28(m,8H),7.09–7.01(m,2H),6.93(tt,J=8.3,1.6Hz,2H),6.77–6.67(m,2H),4.08–3.93(m,4H),3.92–3.81(m,2H),3.74–3.51(m,4H),3.26(dd,J=5.9,3.2Hz,6H),2.65–2.55(m,6H),2.43–2.34(m,6H),2.00(q,J=8.2,7.8Hz,4H),1.00–0.87(m,6H).
LC-MS(ESI)[M+H]+873.5; retention time 4.437min, HPLC purity 98.58%.
Example 22
Figure BDA0002706278290000233
The compound of example 22 was prepared in the same manner as in example 7 except that the product of step c was used as the starting material in step e without conducting step d of example 7 (methyl group was not introduced on amide N), and the product was a white solid with a final yield of 39%.
LC-MS(ESI)[M+H]+941.52; retention time 5.436min, HPLC purity 98.54%.
Example 23
Figure BDA0002706278290000241
Intermediate 23C was synthesized in the same manner as in 1H in example one, except that D-alanine in step a in example was replaced with L-alanine.
Step a Compound 1H (1.53g, 3.43mmol), DIPEA (0.63mL, 3.78mmol) dissolved in 20mL of DMF, HATU (1.5g, 3.78mmol) added, reaction at room temperature for 2H, N-Boc piperazine (704mg, 3.78mmol) added, reaction at room temperature for 8H, monitoring the reaction with TLC plate, after completion of the reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase on silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 600mg of white solid, compound 23A, 28.5% yield.
1H NMR(400MHz,CDC13)δ7.51–7.46(m,2H),7.45–7.41(m,2H),7.15(dd,J=8.3,1.9Hz,1H),6.93(d,J=8.3Hz,1H),6.89(d,J=1.8Hz,1H),4.17(q,J=6.8Hz,1H),4.01–3.93(m,2H),3.84(d,J=5.8Hz,2H),3.56(q,J=6.5,5.9Hz,5H),3.38(s,3H),2.43(s,3H),1.83(dt,J=10.9,5.7Hz,3H),1.69(td,J=11.3,10.2,5.0Hz,1H),1.62–1.53(m,4H),1.50(s,9H),1.02(d,J=6.8Hz,3H).
In step B, compound 23A (600mg, 0.98mmol) was dissolved in 10mL of 4N HCl/dioxane, reacted at room temperature for 8h, monitored by TLC plates, after the reaction was completed, the reaction solution was poured into 20mL of water, the sodium hydroxide solid was neutralized to pH7, 50mL × 2DCM was extracted, the organic layers were combined, washed with 50mL of saturated brine 1 time, dried over anhydrous sodium sulfate, the solvent was evaporated, the organic phase silica gel sample was purified by flash chromatography using MeOH/DCM in a 1-10% gradient to give 340mg of a red solid, compound 23B, 67.3% yield.
1H NMR(400MHz,DMSO-d6) δ 7.37(s,4H), 7.23-7.13 (m,2H),6.72(d, J ═ 1.8Hz,1H),4.06(q, J ═ 6.7Hz,1H),3.68(dt, J ═ 15.5,5.3Hz,4H),3.35(td, J ═ 15.2,7.8Hz,1H),3.27(s,3H),2.83(dt, J ═ 20.4,5.1Hz,4H),2.38(s,3H), 1.75-1.37 (m,6H), 1.31-1.12 (m,2H),0.88(d, J ═ 6.7Hz,3H), no NH at the piperazine end was detected.
LC-MS(ESI)[M+H]+514.52; retention time 2.006min, HPLC purity 99.75%.
Step C Compound 23C (200mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) dissolved in 10mL DMF, HATU (186.3mg, 0.49mmol) added, reaction at room temperature for 2h, Compound 23B (231mg, 0.45mmol) added, reaction at room temperature for 8h, reaction monitored by TLC plate, after completion of reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 60mg of white solid, compound 23, yield 14.2%.
1H NMR(400MHz,DMSO-d6)δ7.44–7.31(m,8H),7.25–7.10(m,4H),6.77–6.68(m,2H),4.06(p,J=6.8Hz,2H),3.97–3.69(m,8H),3.33(m,2H),3.27(d,J=8.1Hz,6H),2.39(d,J=8.2Hz,6H),1.72(d,J=16.5Hz,4H),1.65–1.34(m,8H),1.22(d,J=15.9Hz,4H),0.88(dd,J=10.0,6.7Hz,6H).
Example 24
Figure BDA0002706278290000251
The procedure for the synthesis of compound 24A was the same as that for compound 23C in example 23, except that cyclopentanone was replaced with cyclobutanone. The synthetic procedure for compound 24D was the same as for compound 1H in example 1, except that cyclopentanone was replaced with cyclobutanone in step c. This example, steps a-c, was carried out in the same manner as example 23 to prepare the compound of example 24 except substituting N-Boc piperazine in step a of example 23 with N-Boc- (2S, 5R) -2, 5-dimethylpiperazine. Compound 24 was a white solid with a final step yield of 45%.
1H NMR(400MHz,DMSO-d6)δ7.41(d,J=4.3Hz,4H),7.37(d,J=2.5Hz,4H),7.16–7.14(m,2H),7.11(dd,J=4.9,1.9Hz,2H),6.66–6.58(m,2H),5.06–4.48(m,2H),4.33(dd,J=25.0,13.5Hz,2H),4.04(dq,J=9.9,7.2Hz,2H),3.94(q,J=7.1Hz,2H),3.48(dt,J=15.4,7.8Hz,2H),3.35(s,6H),3.27(dd,J=7.8,1.7Hz,6H),2.51(p,J=1.8Hz,6H),2.44–2.34(m,6H),1.40–1.32(m,3H),1.31–1.22(m,3H),0.84(ddd,J=10.0,6.7,1.5Hz,6H).
LC-MS(ESI)[M+H]+941.73; retention time 5.990min, HPLC purity 97.808%.
Example 25
Figure BDA0002706278290000261
Step a Compound 25A (1.6g, 5.2mmol) was dissolved in 50mL dry THF and LiAlH was added slowly in portions in an ice water bath4(208mg, 5.2mmol) and reacted at room temperature for 4h, the reaction was monitored by TLC plates, after completion of the reaction, the reaction was quenched by pouring into saturated ammonium chloride solution, extracted 100mL x 2EA, the organic layers were combined, washed 1 time with 150mL saturated brine, dried over anhydrous sodium sulfate, the solvent was evaporated, and the organic phase silica gel was purified by flash chromatography with a gradient of MeOH/DCM 1-10% to give 860mg of a white solid, compound 25B, 62% yield.
LC-MS(ESI)[M+H]+309.97; retention time 3.045min, HPLC purity 99.105%.
Step B Compound 25B (439mg, 1.59mmol), (R) -6-bromo-4-cyclobutyl-1, 3-dimethyl-3, 4-dihydroquinoxalin-2 (1H) -one (513.5mg, 1.74mmol), aqueous sodium bicarbonate (270mg, 3.21mmol) was dissolved in 10mL THF using N2After 1min of aeration, Pa (dppf) was added2Cl2(130mg, 0.159mmol), with N again2Ventilating for 1min, reacting at 80 deg.C for 8h, monitoring reaction with TLC plate, and after reaction, 50mLH2O/50mL × 2DCM extraction, combining the organic layers, washing with 100mL saturated brine 1 time, drying over anhydrous sodium sulfate, evaporating the solvent to dryness, and purification of the organic phase silica gel sample by flash chromatography using a EA/PE ═ 10-33% gradient to afford 571mg of white solid, compound 25C, yield 86%.
1H NMR(400MHz,CDC13)δ7.28(d,J=1.0Hz,1H),7.10(dd,J=8.3,1.8Hz,2H),6.92(d,J=8.3Hz,2H),6.67(d,J=1.8Hz,2H),4.87(d,J=6.0Hz,2H),4.07(q,J=6.8Hz,1H),3.67–3.45(m,1H),3.37(d,J=1.0Hz,3H),2.41(s,3H),1.67(qq,J=18.8,9.4,8.6Hz,6H),0.95(d,J=6.8Hz,3H).
Step C Compound 25C (2.2g, 5.2mmol), DMAP (636mg, 5.2mmol) was dissolved in 100mL dry DCM, MsCl (1.2g, 10.4mmol) was added slowly under ice water bath, the reaction was monitored by TLC plate for 4h at room temperature and after completion of the reaction 100mLH2O/100mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 954mg of white solid, compound 25D, yield 37%.
Step D Compound 25D (793mg, 1.6mmol), DIPEA (207mg, 1.6mmol) dissolved in 10mL anhydrous DMF, trans-2, 5-dimethylpiperazine (92mg, 0.8mmol) added, reacted for 8h at room temperature, monitored by TLC plate and after completion of the reaction, 100mLH2Extracting with O/100mL of 2DCM, mixing organic layers, washing with 100mL of saturated saline solution for 1 time, drying with anhydrous sodium sulfate, evaporating to remove solvent, mixing organic phase silica gel, purifying with flash chromatography column, and eluting with MeOH/DCM-1-10% gradient to obtainTo a white solid of 1.17g, compound 25, yield 80%.
1H NMR(400MHz,DMSO-d6)δ7.41–7.29(m,8H),7.16–7.10(m,4H),6.57(d,J=7.0Hz,2H),3.95(q,J=6.7Hz,2H),3.74(m,9H),3.54–3.32(m,3H),3.32–3.24(m,6H),2.51(p,J=1.8Hz,6H),2.39(d,J=1.7Hz,6H),2.10(d,J=8.3Hz,3H),2.06–1.85(m,3H),1.57(ddt,J=32.2,16.6,9.3Hz,6H),0.83(d,J=6.8Hz,6H).
Example 26
Figure BDA0002706278290000271
Step a Compound 25A (525mg, 1.69mmol), lithium hydroxide monohydrate (284mg, 6.76mmol) was dissolved in 50mL THF: h2O is 4: 1 at room temperature for 4h, monitoring the reaction with TLC plate, and after the reaction is finished, adjusting pH to 1-2 with 1N HCl, 50mLH2O/50mL × 2EA extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purified by flash chromatography on silica gel with MeOH/DCM as a 1-10% gradient to give 472mg of white solid, compound 26A, 99% yield.
Step b Compound 26A (968mg, 3.43mmol), DIPEA (489mg, 3.78mmol) dissolved in 20mL DMF, HATU (1.44g, 3.78mmol) added, reaction at room temperature for 2h, N-Boc piperazine (704mg, 3.78mmol) added, reaction at room temperature for 8h, reaction monitored by TLC plate, after completion of reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 468mg of white solid, compound 26B, 28.5% yield.
LC-MS(ESI)[M+H]+477.96; retention time 3.586min, HPLC purity 89.329%.
Step C, compound 26B (469mg, 0.98mmol) was dissolved in 10mL of 4N HCl/dioxane, reacted at room temperature for 8h, monitored by TLC plates, after the reaction was completed, the reaction solution was poured into 20mL of water, the sodium hydroxide solid was neutralized to pH7, 50mL of 2DCM was extracted, the organic layers were combined, washed with 50mL of saturated brine 1 time, dried over anhydrous sodium sulfate, the solvent was evaporated, the organic phase silica gel sample was purified by flash chromatography using MeOH/DCM in a gradient of 1-10% to give 250mg of red solid, compound 26C, 67.3% yield.
LC-MS(ESI)[M+H]+334.17; retention time 2.524min, HPLC purity 91.755%.
Step d Compound 26C (605mg, 1.6mmol), (5-bromo-1- (p-methylphenyl) -1H-1,2, 4-triazol-3-yl) methyl methanesulfonate (554mg, 1.6mmol), DIPEA (207mg, 1.6mmol) was dissolved in 10mL anhydrous DMF, reacted at room temperature for 8H, monitored by TLC plates, and after completion of the reaction, 100mLH was added2O/100mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 747mg of a white solid, compound 26D, yield 80%.
LC-MS(ESI)[M+H]+585.15; retention time 3.109min, HPLC purity 99.898%.
Step e Compound 26D (929mg, 1.59mmol), (R) -6-bromo-4-cyclobutyl-1, 3-dimethyl-3, 4-dihydroquinoxalin-2 (1H) -one (538mg, 1.74mmol), aqueous sodium bicarbonate (270mg, 3.21mmol) was dissolved in 10mL of THF using N2After 1min of aeration, Pa (dppf) was added2Cl2(130mg, 0.159mmol), with N again2Ventilating for 1min, reacting at 80 deg.C for 8h, monitoring reaction with TLC plate, and after reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a gradient EA/PE of 10-33% to give 1.27g of a white solid, compound 26, 86% yield.
1H NMR(400MHz,DMSO-d6)δ7.39–7.26(m,8H),7.12(d,J=11.4Hz,4H),6.59(dd,J=11.4,1.4Hz,2H),3.95(qd,J=6.7,3.2Hz,1H),3.73(d,J=13.6Hz,2H),3.65(m,3H),3.53–3.42(m,2H),3.40–3.35(m,4H),3.27(d,J=2.0Hz,6H),2.38(d,J=3.5Hz,6H),2.17–2.06(m,5H),1.64–1.48(m,8H),1.44–1.28(m,5H),0.84(d,J=6.7Hz,6H).
LC-MS(ESI)[M+H]+927.62; retention time 3.742min, HPLC purity 99.262%.
Example 27
Figure BDA0002706278290000281
Step a, dissolving compound 27A (2g, 7.11mmol) in 30mL of anhydrous DMF, adding NaH (580mg, 14.5mmol) in portions in an ice-water bath, stirring at 0 ℃ for 30min, slowly adding 4-bromo-1-butene (1.5g, 10.67mmol), reacting at room temperature for 2h, monitoring the reaction by using a TLC plate, after the reaction is finished, pouring the reaction solution into 200mL of ice water for quenching, extracting 100mL of x 2DCM, combining organic layers, washing with 100mL of saturated saline for 1 time, drying with anhydrous sodium sulfate, evaporating the solvent, mixing with organic phase silica gel, purifying by using a flash chromatography column, and eluting with a gradient of 0-25% EA/PE to obtain 2g of white solid, namely compound 27B, with the yield of 86%.
Step B Compound 27B (5.2g, 15.5mmol), pinacol bisboronic acid ester (4.4g, 17.0mmol), potassium acetate (3g, 31.0mmol) were dissolved in 100mL 1.4-dioxane with N2After 1min of aeration, Pa (dppf) was added2Cl2(700mg, 0.775mmol), with N again2After 1min of aeration, reaction was carried out for 8h at 100 ℃, the reaction was monitored by TLC plates, after the reaction was completed, the solvent was evaporated to dryness, the silica gel sample was purified by flash chromatography using a gradient of EA/PE 10-33% to give 5g of colorless oily liquid, compound 27C, in 85.4% yield.
Step C Compound 27C (6.1g, 15.93mmol), ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate (5.4g, 17.41mmol), aqueous sodium bicarbonate (2.7g, 32.14mmol) were dissolved in 100mL THF using N2After 1min of aeration, Pa (dppf) was added2Cl2(1.3g, 1.59mmol), and N2Ventilating for 1min, reacting at 80 deg.C for 8h, monitoring reaction with TLC plate, and after reaction, 200mLH2O/200mL of 2DCM extraction, organic layer combination, 200mL of saturated salt water washing 1 times, anhydrous sodium sulfate drying, evaporation of solvent, organic phase silica gel sample through flash chromatographyColumn purification, eluting with a gradient of EA/PE 10-33% gave 1.4g of a white solid, compound 27D, in 18.6% yield.
1H NMR(400MHz,CDCl3)δ7.35–7.22(m,4H),7.21–7.08(m,2H),6.92(dd,J=8.6,3.6Hz,1H),5.32(s,3H),4.56(q,J=7.1Hz,2H),4.06(dt,J=13.2,6.7Hz,2H),3.88–3.72(m,3H),2.43(d,J=12.0Hz,3H),1.47(q,J=6.7Hz,3H),0.70(d,J=5.6Hz,1H),0.58–0.43(m,2H),0.17(d,J=10.2Hz,1H).
Step D Compound 27D (821mg, 1.69mmol), lithium hydroxide monohydrate (280mg, 6.76mmol) was dissolved in 50mL THF: h2O is 4: 1 at room temperature for 4h, monitoring the reaction with TLC plate, and after the reaction is finished, adjusting pH to 1-2 with 1N HCl, 50mLH2O/50mL × 2EA extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purified by flash chromatography on silica gel with MeOH/DCM ═ 1-10% gradient to afford 765mg of white solid, compound 27E, yield 99%.
Step E Compound 27E (206mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) dissolved in 10mL DMF, HATU (186.3mg, 0.49mmol) added, reacted at room temperature for 2h, trans-2.5-dimethylpiperazine (23mg, 0.2mmol) added, reacted at room temperature for 8h, monitored by TLC plate, after completion of reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 88mg of a white solid, compound 27, yield 20%.
1H NMR(400MHz,CDCl3-d)δ7.38–7.24(m,7H),7.19(dt,J=17.3,5.0Hz,4H),6.98–6.88(m,2H),5.90–5.77(m,2H),5.68–5.43(m,1H),5.15–5.02(m,4H),4.66–4.43(m,2H),4.08(dq,J=13.5,6.8Hz,4H),3.92–3.60(m,6H),1.70(m,2H),1.60–1.41(m,6H),1.21–1.04(m,8H),0.70(m,6H),0.55(d,J=11.0Hz,6H),0.19(d,J=11.8Hz,4H).
LC-MS(ESI)[M+Na]+1015.7; retention time 4.388min, HPLC purity 99.7%.
Example 28
Figure BDA0002706278290000291
Step a Compound 28A (4.6g, 15.5mmol), pinacol bisboronic acid ester (4.4g, 17.0mmol), potassium acetate (3g, 31.0mmol) were dissolved in 100mL of 1.4-dioxane using N2After 1min of aeration, Pa (dppf) was added2Cl2(700mg, 0.775mmol), with N again2After 1min of aeration, reaction was carried out for 8h at 100 ℃, the reaction was monitored by TLC plates, after the reaction was completed, the solvent was evaporated to dryness, the silica gel sample was purified by flash chromatography using a gradient of EA/PE 10-33% to give 4.5g of colorless oily liquid, compound 28B, in 85.4% yield.
Step B Compound 28B (5.5g, 15.93mmol), Ethyl 5-bromo-1- (p-methylphenyl) -1,2, 4-triazole-3-carboxylate (5.4g, 17.41mmol), aqueous sodium bicarbonate (2.7g, 32.14mmol) were dissolved in 100mL THF using N2After 1min of aeration, Pa (dppf) was added2Cl2(1.3g, 1.59mmol), and N2Ventilating for 1min, reacting at 80 deg.C for 8h, monitoring reaction with TLC plate, and after reaction, 200mLH2O/200mL × 2DCM extraction, combined organic layers, washed 1 time with 200mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a gradient EA/PE of 10-33% to give 1.3g of a white solid, compound 28C, 18.6% yield.
Step C Compound 28C (753mg, 1.69mmol), lithium hydroxide monohydrate (280mg, 6.76mmol) was dissolved in 50mL THF: h2O is 4: 1 at room temperature for 4h, monitoring the reaction with TLC plate, and after the reaction is finished, adjusting pH to 1-2 with 1N HCl, 50mLH2O/50mL × 2EA extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purified by flash chromatography on silica gel in organic phase eluting with MeOH/DCM ═ 1-10% gradient to afford 698mg of white solid, compound 28D, in 99% yield.
Step d: compound 28D (1.44g, 3.43mmol), DIPEA (489mg, 3.78mmol) was dissolved in 20mL DMF,HATU (1.44g, 3.78mmol) was added and reacted at room temperature for 2h, N-Boc piperazine (704mg, 3.78mmol) was added and reacted at room temperature for 8h, the reaction was monitored by TLC plate, and after completion of the reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase on silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 600mg of white solid, compound 28E, yield 28.5%.
Step E compound 28E (601mg, 0.98mmol) was dissolved in 10mL 4N HCl/dioxane, reacted for 8h at room temperature, monitored by TLC plate, after the reaction was completed, the reaction was poured into 20mL water, solid sodium hydroxide was neutralized to pH7, 50mL 2DCM was extracted, the organic layers were combined, washed with 50mL saturated brine 1 time, dried over anhydrous sodium sulfate, solvent evaporated, and the organic phase silica gel sample was purified by flash chromatography eluting with MeOH/DCM 1-10% gradient to give 363mg red solid, compound 28F, 67.3% yield.
Step F Compound 27E (206mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) dissolved in 10mL DMF, HATU (186.3mg, 0.49mmol) added, reaction at room temperature for 2h, Compound 28F (248mg, 0.45mmol) added, reaction at room temperature for 8h, reaction monitored by TLC plate, after completion of reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 858mg of white solid, compound 28, in 20% yield.
1H NMR(400MHz,CDCl3-d)δ7.38–7.24(m,10H),7.19(dd,J=17.2,3.3Hz,2H),6.98–6.83(m,2H),5.92–5.77(m,1H),5.56(m,1H),5.30(m,1H),5.14–5.05(m,2H),4.96(m,2H),4.68–4.43(m,2H),4.08(dq,J=13.9,6.7Hz,3H),3.94–3.70(m,4H),3.45(d,J=14.1Hz,2H),3.36(dd,J=6.2,1.6Hz,3H),2.45(d,J=6.0Hz,7H),1.59–1.40(m,7H),1.19–1.06(m,5H),0.71(s,3H),0.54(s,3H).
LC-MS(ESI)[M+Na]+975.6; retention time 4.177min, HPLC purity 97.788%.
Example 29
Figure BDA0002706278290000311
Compound 29A (519mg, 1.6mmol), compound 29B (843mg, 1.6mmol), DIPEA (207mg, 1.6mmol) were dissolved in 10mL anhydrous DMF, reacted at room temperature for 8h, the reaction was monitored by TLC plates and, after completion of the reaction, 100mLH2O/100mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 1g of white solid, compound 29, yield 81%.
LC-MS(ESI)[M+H]+785.5; retention time 18.158min, HPLC purity 94.961%.
Example 30
Figure BDA0002706278290000312
Compound 30A (474mg, 1.6mmol), compound 29B (843mg, 1.6mmol), DIPEA (207mg, 1.6mmol) were dissolved in 10mL anhydrous DMF, reacted at room temperature for 8h, the reaction was monitored by TLC plates and, after completion of the reaction, 100mLH2O/100mL × 2DCM extraction, combined organic layers, washed 1 time with 100mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 823mg of a white solid, compound 30, in 68% yield.
1H NMR(400MHz,CDCl3-d)δ7.29(d,J=4.0Hz,3H),7.17(d,J=8.2Hz,1H),7.11(s,1H),7.07(d,J=7.6Hz,1H),7.00(d,J=7.7Hz,1H),6.95–6.87(m,3H),4.97(m,2H),4.16(t,J=6.8Hz,1H),3.83(s,3H),3.76(s,2H),3.58(m,4H),3.48(q,J=8.3,7.6Hz,1H),3.38(s,3H),2.43(s,3H),2.33(s,3H),2.19(s,3H),1.65(m,6H),1.32(d,J=7.7Hz,2H),1.28(d,J=3.6Hz,4H),1.17(m,2H),1.01(d,J=6.8Hz,3H).
LC-MS(ESI)[M+H]+757.52; retention time 17.981min, HPLC purity 83.48%.
Example 31
Figure BDA0002706278290000321
Compound 26A (127mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) were dissolved in 10mL of DMF, HATU (186.3mg, 0.49mmol) was added and the reaction was allowed to proceed at room temperature for 2h, compound 29B (244mg, 0.45mmol) was added and the reaction was allowed to proceed at room temperature for 8h, monitored by TLC plate and after completion of the reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase on silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 56mg of white solid, compound 31, 14.2% yield.
LC-MS(ESI)[M+H]+805.32; retention time 9.095min, HPLC purity 94.877%.
Example 32
Figure BDA0002706278290000322
(R) -5- (4-cyclopentyl-1, 3-dimethyl-2-oxo-1, 2,3, 4-tetrahydroquinoxalin-6-yl) -1-phenyl-1, 2, 4-triazole-3-carboxylic acid (195mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) dissolved in 10mL DMF, HATU (186.3mg, 0.49mmol) added, reaction at room temperature for 2h, compound 32A (189mg, 0.45mmol) added, reaction at room temperature for 8h, reaction monitored by TLC plate, after reaction completion, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 53mg of white solid, compound 32, 14.2% yield.
1H NMR(400MHz,CDCl3-d)δ7.71(s,1H),7.54(s,1H),7.51–7.42(m,3H),7.38(d,J=8.0Hz,2H),7.18(m,3H),7.04(m,2H),6.93(d,J=8.3Hz,1H),6.89(s,1H),4.89(m,2H),4.56–4.35(m,1H),4.35–4.09(m,2H),3.94(s,4H),3.77(d,J=5.8Hz,1H),3.37(s,3H),3.16–2.92(m,1H),2.78(m,2H),2.21(m,2H),2.13(s,2H),1.65–1.48(m,6H),1.39–1.27(m,7H),1.21(m,1H),1.01(d,J=6.8Hz,3H).
LC-MS(ESI)[M+H]+833.47; retention time 3.456min, HPLC purity 100%.
Example 33
Figure BDA0002706278290000331
Compound 33A (215mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) were dissolved in 10mL of dmf, HATU (186.3mg, 0.49mmol) was added and reacted at room temperature for 2h, (R) -4-cyclopentyl-1, 3-dimethyl-6- (1-phenyl-3- (piperazinyl-1-carbonyl) -1,2, 4-triazole) -3, 4-dihydroquinoxalin-2-one (225mg, 0.45mmol) was added and reacted at room temperature for 8h, the reaction was monitored by TLC plate and after completion of the reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase on silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 78mg of white solid, compound 33, yield 17.9%.
1H NMR(400MHz,CDCl3-d)δ7.73–7.57(m,2H),7.47(d,J=15.5Hz,5H),7.39–7.30(m,2H),7.16(t,J=11.0Hz,2H),7.01(dd,J=17.1,8.2Hz,1H),6.96–6.81(m,3H),4.42(d,J=12.6Hz,1H),4.23(d,J=14.5Hz,2H),4.16(t,J=7.0Hz,1H),4.06(d,J=20.4Hz,2H),3.92(d,J=6.4Hz,6H),3.77(d,J=14.4Hz,7H),3.37(d,J=7.2Hz,5H),3.10(s,3H),2.75(m,2H),2.22(dd,J=11.1,6.0Hz,2H),2.10(d,J=9.3Hz,1H),1.91(m,3H),1.54(d,J=18.1Hz,5H),1.29(d,J=12.9Hz,3H),1.00(t,J=7.6Hz,4H).
LC-MS(ESI)[M+H]+959.62; retention time 3.067min, HPLC purity 100%.
Example 34
Figure BDA0002706278290000332
Compound 34A (150mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) was dissolved in 10mLAdding HATU (186.3mg, 0.49mmol) into DMF, reacting at room temperature for 2h, adding (R) -4-cyclopentyl-1, 3-dimethyl-6- (1-phenyl-3- (piperazinyl-1-carbonyl) -1,2, 4-triazole) -3, 4-dihydroquinoxalin-2-one (225mg, 0.45mmol), reacting at room temperature for 8h, monitoring the reaction by using a TLC plate, and after the reaction is finished, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase silica gel sample by flash chromatography using a MeOH/DCM ═ 1-10% gradient to afford 150mg of white solid, compound 34, yield 41.3%.
1H NMR(400MHz,CDCl3-d)δ10.41(s,1H),8.13(s,1H),7.62(d,J=14.4Hz,3H),7.46(d,J=20.9Hz,6H),7.15(d,J=8.4Hz,1H),6.95–6.80(m,2H),6.47(s,1H),5.88(td,J=17.1,6.9Hz,3H),5.19–5.06(m,2H),4.19(dt,J=27.8,7.2Hz,4H),3.97(s,3H),3.37(d,J=1.9Hz,3H),3.23(s,1H),2.62(q,J=7.4Hz,2H),1.53(m,4H),1.39–1.21(m,4H),1.21(s,4H),1.00(d,J=6.7Hz,3H),0.90(t,J=6.7Hz,1H).
LC-MS(ESI)[M+H]+844.62; retention time 3.07min, HPLC purity 95.338%.
Example 35
Figure BDA0002706278290000341
Compound 35A (202mg, 0.45mmol), DIPEA (0.81mL, 0.49mmol) were dissolved in 10mL of DMF, HATU (186.3mg, 0.49mmol) was added and reacted at room temperature for 2h, then (R) -4-cyclopentyl-1, 3-dimethyl-6- (1-phenyl-3- (piperazinyl-1-carbonyl) -1,2, 4-triazole) -3, 4-dihydroquinoxalin-2-one (225mg, 0.45mmol) was added and reacted at room temperature for 8h, the reaction was monitored by TLC plate, after completion of the reaction, 50mLH2O/50mL × 2DCM extraction, combined organic layers, washed 1 time with 50mL saturated brine, dried over anhydrous sodium sulfate, evaporated to dryness, and purification of the organic phase on silica gel by flash chromatography eluting with a MeOH/DCM ═ 1-10% gradient to give 94mg of white solid, compound 35, 22.3% yield.
1H NMR(400MHz,CDCl3-d)δ10.02(s,1H),8.03(s,1H),7.63(m,2H),7.55(d,J=6.7Hz,2H),7.49(s,2H),7.44(d,J=3.5Hz,1H),7.15(d,J=7.8Hz,1H),6.94(d,J=8.6Hz,1H),6.87(s,1H),6.49(s,1H),5.88(dt,J=16.5,8.1Hz,1H),5.47–5.31(m,2H),5.11(dd,J=24.2,13.6Hz,2H),4.23(t,J=7.4Hz,2H),4.17(q,J=6.8Hz,1H),4.06(d,J=24.5Hz,2H),3.95(s,3H),3.89(d,J=22.3Hz,3H),3.70(d,J=28.3Hz,5H),3.46(m,5H),3.38(s,3H),2.62(d,J=7.5Hz,2H),2.24(t,J=7.7Hz,3H),1.82(s,3H),1.32–1.24(m,4H),1.01(d,J=6.8Hz,3H),0.90(t,J=6.5Hz,2H).
LC-MS(ESI)[M+H]+970.5; retention time 2.815min, HPLC purity 97.631%.
Experimental examples
Method for testing molecular activity
The binding activity of the compound to the bromodomain1 domain of BRD4 protein (hereinafter referred to as BRD4 BD1) was tested using the Fluorescence Anisotropy assay (fluoroscience Anisotropy). Based on the principle that the fluorescence polarization values in the horizontal direction and the vertical direction are calculated for correlation analysis by detecting the molecular weight change before and after the interaction between the fluorescein labeled small molecule and other molecules. If the binding equilibrium between the fluorescently labeled small molecules and the large molecules is established, the fluorescence is excited and moves slowly, and the measured fluorescence polarization value is increased. If the combination between the fluorescence labeling micromolecule and the macromolecule is replaced by other ligands, the rotation or overturning speed of the fluorescence labeling micromolecule in a free state is increased, the emitted light is depolarized relative to an excitation light plane, the measured polarized light value is reduced, and the fluorescence anisotropy of the sample is calculated.
The experimental reaction system was 40ul, each of which was equipped with multiple wells, and the buffer was 50mM HEPES PH7.4,150mM NaCl,0.5mM CHAPS. The fluorescent substrate is a positive compound JQ1 connected with fluorescent molecules, the working concentration is 5nM, BRD4 protein is expressed by escherichia coli, the working concentration is 10nM, the primary screening concentration of the compound is 1uM, the final concentration of DMSO is two thousandth, all the components are mixed in a 384-well plate (corning product number: CLS3575), and the mixture is kept overnight at 4 ℃ for 16 hours. Measuring anisotpy value after reaction, calculating inhibition rate by taking average value of multiple wells, and measuring IC of compound with inhibition rate more than 50% under the condition50The value is obtained.
And (3) testing results: some of the test compounds showed higher molecular level inhibitory activity against either the BRD4 BD1 domain or the BRD4 full-length protein.
TABLE 1 inhibitory Activity IC of partial Compounds of the present invention against BRD4 BD1 bromodomain protein50Value (nM)
Examples BRD4 BD1 IC50(nM)
9 23.13
24 59.24
25 27.28
32 2.91
33 8.75
34 5.27
35 4.38
TABLE 2 inhibitory Activity IC of partial Compounds of the present invention against full-Length BRD4 protein50Value (nM)
Examples BRD4 IC50(nM)
1 6.2
5 6.5
6 9.7
23 66.6
Second, testing method of cell activity
(1) The experimental principle is as follows:
CCK8 method: the CCK8 kit contains WST-8, and the compound can be reduced into a yellow formazan product by dehydrogenase in mitochondria of living cells under the action of an electron carrier 1-methoxy-5-methylphenazinium dimethyl sulfate. The number of formazan-generating products is directly proportional to the number of viable cells, and the more rapid the cell proliferation is, the darker the color is; the more toxic the compound, the less colored the inhibition of cell proliferation. The light absorption value of the enzyme linked immunosorbent assay device is measured at the wavelength of 450nM, and the number of living cells can be indirectly reflected.
(2) Experimental materials: various tumor cell cultures (Gibco), CCK8(Sigma), trichloroacetic acid (national drug), glacial acetic acid (national drug), Tris base unbuffer (national drug).
(3) The experimental steps are as follows:
cells in logarithmic growth phase were seeded at 100. mu.l/well in 96-well plates and cultured at 37 ℃ for 24 hours until the cells were attached.
(II) Add 10. mu.l of drug at a dilution concentration per well, with triplicates per concentration. And setting physiological saline solvent control and cell-free zero setting holes with corresponding concentrations, and making cell-free zero setting holes with corresponding drug concentrations if the drugs are colored.
(III) all cell lines were cultured in complete medium supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin for optimal growth of the cells and placed at 37 ℃ with 5% CO2Cultured in an incubator.
(IV) if the cell activity is tested by a CCK8 method, adding 10 mu l/hole of CCK8 reagent, continuing culturing for 4 hours, and then measuring the light absorption value by an enzyme-labeling instrument. If the SRB method is used to test the cell activity, the culture solution is discarded, 100 mul/well of 10% TCA precooled at 4 ℃ is added, the mixture is fixed for 1 hour at 4 ℃, then the mixture is washed 5 times by distilled water and naturally dried in the air.
(V) Add 100. mu.l/well of SRB (4mg/ml) solution prepared in 1% glacial acetic acid and stain for 15min at room temperature.
(VI) the supernatant was removed, washed 5 times with 1% acetic acid and air dried.
(VII) 150. mu.l/well Tris (10mM) solution was added thereto and allowed to stand at room temperature for 5 minutes.
(VIII) light absorption (OD value) was measured at 560nM microplate reader.
(4) And (3) data analysis:
(a) calculating the formula: inhibition rate (OD value)Control groupOD valueAdministration setvalue)/OD valueControl groupA
(b)IC50The values were calculated using the Logit method.
(5) And (3) testing results:
some of the compounds tested showed strong proliferation inhibitory activity against TY82 thymus carcinoma cells and mm.1s myeloma cells, which was more than 10000-fold higher than the monovalent inhibitor (HJP-178) obtained in the previous work of the inventors.
Figure BDA0002706278290000361
TABLE 3 proliferation inhibitory Activity of some of the Compounds of the invention on tumor cell lines IC50Value (nM)
Figure BDA0002706278290000362
Figure BDA0002706278290000371
Third, the experimental method of oral metabolism experiment of mice: dissolving in dimethyl acetamide: test compound (10mg/kg) in 0.5% HPMC (hydroxypropylmethylcellulose) (5: 95, v/v) was formulated to a concentration of 1mg/mL and administered to ICR mice (male, 18-22g, n ═ 3) by gavage administration. Blood samples (anticoagulant: EDTA-Na2) were collected at 0.25, 0.5, 1,2,4, 8 and 24 hours post-dose. 100 μ L of methanol with internal standard: acetonitrile (1:1, v/v) solvent was added to 10. mu.L of plasma and vortexed thoroughly. Centrifuge for 5 minutes, then mix 20 μ L supernatant with 20 μ L water for analysis. Samples were analyzed by a Xevo TQ-S triple quadrupole mass spectrometer (Waters, USA). Analysis was performed using ACQUITY UPLC BEH C18(1.7 μm, 2.0 mm. times.50 mm, Waters, USA). The elution was performed in a gradient consisting of 5mM ammonium acetate in water containing 0.1% formic acid and acetonitrile containing 0.1% formic acid. After analyzing the concentration of the test compound, values of AUClast, AUCINF _ obs and MRTINF _ obs were calculated from the time-concentration curve of each animal using Phoenix WinNonlin (CERTARA, USA). Cmax was determined as the maximum plasma concentration and Tmax was the time to reach the maximum concentration.
The experimental results are as follows: the test compounds 7, 10 and 24 all showed excellent oral absorption capacity.
TABLE 4 oral metabolism of mice with partial compounds of the invention (10mg/kg)
7 10 24 OTX-015
T1/2(h) 4.89 4.09 4.29 0.99
Tmax(h) 1.67 1.67 1.00 0.25
Cmax(ng/mL) 2535.5 1784.8 2456.5 1507
AUClast(h*ng/mL) 19251.2 11756.0 18631.8 3078
AUCINF_pred(h*ng/mL) 19880.6 11932.9 19031.8 3088
MRTlast(h) 5.80 5.21 5.60 1.97
Method for testing pharmacodynamics in vivo
(1) Experimental materials:
DMAC (20160316, Shanghai Lingfeng Chemicals Co., Ltd.), MC (methyl cellulose M20, national drug group Chemicals Co., Ltd.), ultrapure water, experimental animals (female, BALB/c nude mice SPF grade, Beijing Wintolite laboratory animal technologies Co., Ltd.), and corresponding transplantation tumor cell lines.
(2) The experimental method comprises the following steps:
cell culture: the corresponding tumor cells were routinely cultured in IMDM medium containing 10% fetal bovine serum under 5% CO 237 ℃ culture conditions. And (4) carrying out passage 2-3 times per week according to the growth condition of the cells.
(II) establishing a model: tumor cells in logarithmic growth phase were harvested by digestion with 0.25% pancreatin and centrifugation at 800 g. Resuspended in physiological saline and cell counted. The cells were collected by centrifugation, suspended in a physiological saline + Matrigel (1:1) mixture and adjusted to 6X 10 cell concentration7cells/mL. The cells are sucked by a 1mL syringe, injected to the axilla of the front right limb of a nude mouse subcutaneously, and 0.1 mL/mouse, and the growth conditions of the animals and the transplanted tumors are observed regularly.
(III) animal group administration: after 13 days of cell inoculation (Day 13), the tumor size of the animals was measured and the tumor volume was calculated. Eliminating animals with over-small and over-large tumor volume or irregular tumor shape, selecting 112.39mm tumor volume3~185.83mm3The tumor-bearing mice are divided by a random block method according to the tumor volume, and the solvent is used12 controls and 6 others. Dosing was started according to a group schedule with a dosing period of 21 days or 28 days. During the administration period, tumor size was measured 2 times per week, animal body weight was weighed, animal life state was observed, and abnormal conditions were recorded. The last dose was added on the day of test completion, and each group was treated with CO 1 hour and 8 hours after the last dose, respectively23 animals were sacrificed at random (6 per time point in the solvent control group) and blood was collected from the heart and approximately 200. mu.L/animal of EDTA-K2 anticoagulated plasma was collected; and rapidly stripping subcutaneous tumor tissues, photographing, weighing, subpackaging in a homogenate tube according to requirements, quickly freezing by liquid nitrogen, and transferring to a refrigerator at-80 ℃ for storage to be tested. The animals were roughly dissected and observed for abnormalities in the major organs.
(3) And (3) data analysis:
(a) calculating the formula: tumor Volume (TV) 1/2 × a × b2… … … … … … … … … … (equation 5.6)
Wherein a represents the tumor major axis; b represents the tumor minor axis.
(b) Calculating the formula: relative Tumor Volume (RTV) ═ Vt/VinitialX100 (%) … … … … (formula 5.7)
Wherein VinitialMeasurement of the resulting tumor volume, V, for group administrationtFor the tumor volume at each measurement.
(c) Calculating the formula: relative tumor proliferation rate (T/C) ═ TRTV/CRTV) X 100%.... (equation 5.8)
Wherein T isRTVRelative tumor volume, C, in the treatment groupsRTVRelative tumor volumes of the solvent control groups are indicated.
(d) Calculating the formula: tumor inhibition rate (GI) ═ 1- (TV)t-TVinitial)/(CVt-CTinitial)]X 100% … … … … … … … … … … … … … … … … … … … … (formula 5.9)
Wherein TVtRepresents the tumor volume at each measurement of the treatment group; TV (television)initialRepresents the tumor volume of the treatment group when administered in groups; CV oftRepresents the tumor volume at each measurement of the control group; CTinitialTumor body of control group in divided administrationAnd (4) accumulating.
(e) Calculating the formula: weight loss rate (BW)initial-BWfinal)/BWinitialX 100%. 9
Wherein BWinitialRepresents the body weight of the animals at the time of the group administration; BW (Bandwidth)finalIndicating the animal body weight at the end of the test.
(4) The statistical analysis method comprises the following steps:
experimental data were calculated and statistically correlated using Microsoft Office Excel 2007 software, and the comparison between the two groups was performed using the t-test.
(5) And (3) testing results:
some of the tested compounds showed in vivo antitumor activity in mice superior to that of the positive compounds (OTX-015, ABBV-075 and AZD 5153). Wherein OTX-015 and ABBV-075 are monovalent inhibitors and AZD5153 is a divalent inhibitor.
Figure BDA0002706278290000391
TABLE 5 EXAMPLE 15 results of in vivo pharmacodynamic evaluation in MV4-11 (human myelomonocytic leukemia cells) nude mouse subcutaneous tumor-inhibiting model
Figure BDA0002706278290000392
TABLE 6 in vivo pharmacodynamic evaluation results of examples 9 and 10 in MV4-11 nude mouse subcutaneous tumor suppressor model
Figure BDA0002706278290000401
The technical features of the embodiments described above may be arbitrarily combined, and for the sake of brevity, all possible combinations of the technical features in the embodiments described above are not described, but should be considered as being within the scope of the present specification as long as there is no contradiction between the combinations of the technical features. The above-mentioned embodiments only express several embodiments of the present invention, and the description thereof is more specific and detailed, but not construed as limiting the scope of the invention. It should be noted that, for a person skilled in the art, several variations and modifications can be made without departing from the inventive concept, which falls within the scope of the present invention. Therefore, the protection scope of the present patent shall be subject to the appended claims.

Claims (10)

1. A compound having a structure represented by general formula (I), or a pharmaceutically acceptable salt thereof:
Figure FDA0002706278280000011
wherein:
k is selected from the following groups:
Figure FDA0002706278280000012
L1is-C (═ O) -or
Figure FDA0002706278280000013
G1And G2Each independently selected from: absent, -C (═ O) -, substituted or unsubstituted C1-C4 alkylene; the substituted substituents are selected from: hydrogen, halogen, C1-C4 alkyl;
R19and R20Each independently selected from: hydrogen, substituted or unsubstituted C1-C4 alkyl, said substituted substituents being selected from the group consisting of: hydrogen, C1-C4 alkyl;
L4selected from the group consisting of substituted or unsubstituted C1-C5 alkylene, substituted or unsubstituted C3-C10 cycloalkylene, said substituted substituents being selected from the group consisting of: fluorine, chlorine, bromine, hydroxyl, amino, nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy;
or, L4,R19,R20And the nitrogen atoms to which they are attached form together a ring systemSubstituted or unsubstituted 5-10 membered heterocyclyl, said substitution referring to having 1-3 substituents, each substituent independently selected from: halogen, hydroxy, nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy;
x and Y are each independently selected from: c or N; preferably C;
R1and R1' are the same or different from each other, and are each independently selected from: hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, phenyl-substituted C1-C2 alkyl;
R2and R2' are the same or different from each other, and are each independently selected from: hydrogen, C1-C4 alkyl, C2-C4 alkenyl-substituted C1-C4 alkyl;
R3and R3' are the same or different from each other, and are each independently selected from: substituted or unsubstituted phenyl, substituted or unsubstituted naphthyl, substituted or unsubstituted benzyl, said substituted substituents being selected from the group consisting of: halogen, hydroxy, amino, nitro, cyano, C1-C4 alkyl, C1-C4 alkoxy;
"" indicates that the substituent is attached thereto.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
L4selected from unsubstituted C1-C5 alkylene, unsubstituted C3-C10 cycloalkylene, or
L4,R19,R20And the nitrogen atoms to which they are attached form together
Figure FDA0002706278280000021
Figure FDA0002706278280000022
Wherein n4 and n5 are each independently selected from any integer between 0 and 4, which may be the same or different from each other;
n10, n11, n12 and n13 are each independently selected from any integer between 1 and 3, which are the same or different from each other;
or
L4Is composed of
Figure FDA0002706278280000023
Or
L4,R19,R20And the nitrogen atoms to which they are attached form together
Figure FDA0002706278280000024
Figure FDA0002706278280000025
Preferably, G1And G2At least one, or G, is present1And G2Are all-C (═ O) -.
3. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof,
L1selected from the following groups:
Figure FDA0002706278280000031
wherein the content of the first and second substances,
n1, n2 and n3 are each independently selected from any integer between 1 and 5, which are the same or different from each other;
n4, n5, n6, n7, n8 and n9 are each independently selected from any integer between 0 and 4, equal to or different from each other;
n10, n11, n12 and n13 are each independently selected from any integer between 1 and 3, which are the same or different from each other;
L4、R19and R20Are as defined in the appended claims.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (i) is selected from the group consisting of:
Figure FDA0002706278280000032
wherein the content of the first and second substances,
L5is selected from
Figure FDA0002706278280000033
Figure FDA0002706278280000034
Z1Is selected from-C (═ O) -or- (CH)2) n-, n is an integer of 1 to 5;
Z2is selected from-C (═ O) -or-CH2-,
R1And R1' are the same or different from each other and are each independently selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, phenyl-substituted C1-C2 alkyl; preferably selected from cyclopropyl, cyclobutyl, cyclopentyl, methyl, benzyl; preferably R1And R1' same;
R2and R2' are the same or different from each other and are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl substituted C1-C2 alkyl; preferably selected from methyl, vinyl ethyl; preferably R2And R2' same;
R3and R3' are the same or different from each other and are each independently selected from substituted or unsubstituted phenyl, substituted or unsubstituted naphthyl, substituted or unsubstituted benzyl, preferably selected from substituted or unsubstituted phenyl, substituted or unsubstituted naphthyl; the substituted substituent is selected from halogen, hydroxyl, amino, nitro, cyano, C1-C4 alkyl, C1-C4 alkoxy, preferably selected from halogen and methyl;
"" indicates that the substituent is attached at that point;
alternatively, the compound of formula (i) is selected from the following compounds:
Figure FDA0002706278280000041
wherein the content of the first and second substances,
L5、Z1、Z2、R1、R2and R3The same as defined in formula II;
alternatively, the compound of formula (i) is selected from the following compounds:
Figure FDA0002706278280000042
wherein the content of the first and second substances,
L5、Z1、Z2、R1、R2、R3and R3' As defined in formula II, R is selected from halogen;
alternatively, the compound of formula (i) is selected from the following compounds:
Figure FDA0002706278280000051
wherein R is1、R2And R3The same as defined in formula II;
L5the same as defined in formula II, or absent;
Z1as defined in formula II, or is absent or-CH2-C(=O)-,
Z2The same as defined in formula II;
alternatively, the compound of formula (i) is selected from the following compounds:
Figure FDA0002706278280000052
wherein the content of the first and second substances,
R1、R2and R3The same as defined in formula II;
L5the same as defined in formula II, or absent;
Z1as defined in formula II, or is absent or-CH2-C(=O)-,
Z2The same as defined in formula II.
5. The compound of claim 4, or a pharmaceutically acceptable salt thereof,
in the general formula II, R1And R1' is cyclopentyl, R2And R2' is methyl, R3And R3' is phenyl or p-methylphenyl, Z1And Z2Is selected from-C (═ O) -or-CH2-;
In the general formula III, R1Is cyclopentyl, R2Is methyl, R3Is phenyl or p-methylphenyl, Z1Is- (CH)2) n-, n is an integer of 1 to 5, Z2Is selected from-C (═ O) -or-CH2-;
In the formula IV, R1Is cyclopentyl, R2Is methyl, R3And R3' is phenyl or p-methylphenyl, Z1And Z2Is selected from-C (═ O) -or-CH2-;
In the formula V, R1Is cyclopentyl, R2Is methyl, R3Is phenyl, L5Is absent or is
Figure FDA0002706278280000061
Z1Is absent or is-CH2-C(=O)-,Z2is-C (═ O) -;
in the formula VI, R1Is cyclopentyl, R2Is methyl, R3Is phenyl, L5Is absent or is
Figure FDA0002706278280000062
Z1Is absent or is-CH2-C(=O)-,Z2is-C (═ O) -.
6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (i) is selected from the group consisting of:
Figure FDA0002706278280000063
Figure FDA0002706278280000071
7. a pharmaceutical composition comprising one or more selected from the compounds of any one of claims 1-6 and pharmaceutically acceptable salts thereof, optionally comprising one or more pharmaceutical excipients.
8. Use of a compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 7, for the manufacture of an inhibitor of a bromodomain recognition protein; or for the preparation of a medicament for the prevention and/or treatment of a disease associated with a bromodomain recognition protein mediated disease.
9. The use according to claim 8, wherein the relevant disease mediated by a bromodomain recognition protein is selected from: malignant tumor, immunological disease, cardiovascular system disease, virus infection, neurodegenerative disease or inflammation.
10. The use of claim 9, wherein the malignancy is selected from: acute lymphocytic leukemia, acute myelogenous leukemia, B-cell chronic lymphocytic leukemia, chronic myelomonocytic leukemia, testicular nucleoprotein midline cancer, small cell lung cancer, non-small cell lung cancer, B-cell lymphoma, prostate cancer, gastric cancer, colorectal cancer, renal cancer, liver cancer, breast cancer, pancreatic cancer.
CN202011039854.2A 2020-09-28 2020-09-28 Dihydroquinoxaline bromodomain bivalent inhibitors Active CN114276333B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011039854.2A CN114276333B (en) 2020-09-28 2020-09-28 Dihydroquinoxaline bromodomain bivalent inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011039854.2A CN114276333B (en) 2020-09-28 2020-09-28 Dihydroquinoxaline bromodomain bivalent inhibitors

Publications (2)

Publication Number Publication Date
CN114276333A true CN114276333A (en) 2022-04-05
CN114276333B CN114276333B (en) 2023-05-09

Family

ID=80867917

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011039854.2A Active CN114276333B (en) 2020-09-28 2020-09-28 Dihydroquinoxaline bromodomain bivalent inhibitors

Country Status (1)

Country Link
CN (1) CN114276333B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114907323A (en) * 2022-05-31 2022-08-16 内蒙古民族大学 Quinoxalinone compounds and preparation method and application thereof
CN114957222A (en) * 2022-05-31 2022-08-30 内蒙古民族大学 Compound and preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107056771A (en) * 2017-01-23 2017-08-18 中国药科大学 Bromodomain albumen bivalent inhibitors and its preparation method and application
CN107793371A (en) * 2016-09-05 2018-03-13 中国科学院上海药物研究所 A kind of bromine domain identification protein inhibitor and its production and use
CN108290856A (en) * 2015-08-11 2018-07-17 尼奥迈德研究所 The substituted dihydro-quinolone of aryl-, their preparation and their purposes as drug
CN108472295A (en) * 2015-11-25 2018-08-31 达纳-法伯癌症研究所股份有限公司 Divalent bromine structural domain inhibitor and application thereof
CN108570038A (en) * 2017-03-08 2018-09-25 中国科学院上海药物研究所 Dihydro-quinoxaline class bromine structural domain identifies protein inhibitor and preparation method and purposes

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108290856A (en) * 2015-08-11 2018-07-17 尼奥迈德研究所 The substituted dihydro-quinolone of aryl-, their preparation and their purposes as drug
CN108472295A (en) * 2015-11-25 2018-08-31 达纳-法伯癌症研究所股份有限公司 Divalent bromine structural domain inhibitor and application thereof
CN107793371A (en) * 2016-09-05 2018-03-13 中国科学院上海药物研究所 A kind of bromine domain identification protein inhibitor and its production and use
CN107056771A (en) * 2017-01-23 2017-08-18 中国药科大学 Bromodomain albumen bivalent inhibitors and its preparation method and application
CN108570038A (en) * 2017-03-08 2018-09-25 中国科学院上海药物研究所 Dihydro-quinoxaline class bromine structural domain identifies protein inhibitor and preparation method and purposes

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114907323A (en) * 2022-05-31 2022-08-16 内蒙古民族大学 Quinoxalinone compounds and preparation method and application thereof
CN114957222A (en) * 2022-05-31 2022-08-30 内蒙古民族大学 Compound and preparation method and application thereof
CN114957222B (en) * 2022-05-31 2023-08-15 内蒙古民族大学 Compound and preparation method and application thereof
CN114907323B (en) * 2022-05-31 2023-08-15 内蒙古民族大学 Quinoxalinone compounds, preparation method and application thereof

Also Published As

Publication number Publication date
CN114276333B (en) 2023-05-09

Similar Documents

Publication Publication Date Title
CN105228982B (en) For treating 3 acetyl-amino 1 (phenyl heteroarylaminocarbonyl or phenyl heteroarylcarbonyl-amino) benzene derivatives of hyperproliferative disorders
CN104910161B (en) Pyrazolopyrimidine compound and method as JAK inhibitor
WO2021068898A1 (en) Novel kras g12c protein inhibitor, preparation method therefor, and use thereof
CN103384670B (en) Imidazo [1, the 2-b] pyridazine replacing
CN104870440B (en) Bicyclic heterocycles and its therapeutical uses
CN104803989B (en) Application as the endocyclic compound of hepatitis c inhibitor and its in medicine
TW200936591A (en) Tricyclic compounds and use thereof
TW201041856A (en) Resorcinol derivatives
CN111670183A (en) Novel heterocyclic compounds as CDK8/19 inhibitors
CN107849049A (en) Urea derivative or its pharmaceutical salts
CN114276333B (en) Dihydroquinoxaline bromodomain bivalent inhibitors
BR112016001333B1 (en) INDOL AND PYRROL DERIVATIVES AND THEIR USE, PROCESSES FOR THE PRODUCTION THEREOF, PHARMACEUTICAL COMPOSITIONS AND THEIR USE AND COMBINATIONS AND THEIR USE
CN109311846A (en) 6 yuan of new heteroaromatic substituted cyanoindole derivatives as NIK inhibitor
CN109071565A (en) Tricyclic heterocyclic derivatives
WO2023138524A1 (en) Kras g12d degradation agent and medical use thereof
CN109641890A (en) Isocitric dehydrogenase (IDH) inhibitor
WO2022194221A1 (en) Furan fused ring-substituted glutarimide compound
WO2020238776A1 (en) Substituted fused bicyclic derivative, preparation method therefor, and application thereof in medicines
BR112016001080B1 (en) ISOINDOLINE OR ISOQUINOLINE COMPOUNDS, A PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
CN112174940A (en) 3- (6, 7-bis (2-methoxyethoxy) -quinazoline-4-amido) phenyl-1H-triazole derivative
EP3476848A1 (en) Benzofuran pyrazole amine protein kinase inhibitor
CN116375707A (en) Menin inhibitors and uses thereof
CN107250120A (en) It is used as the yl-benzamide derivatives of 1,3,4 thiadiazoles 2 of Wnt signal path inhibitor
CN104066736B (en) Substituted pyrazolopyrimidine as AKT kinase inhibitor
CN106892859A (en) (H) ketone polyamines conjugate of benzo [c, d] indoles 2 and its preparation method and application

Legal Events

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