CN116514795A - Process for preparing 3CLpro protease inhibitors - Google Patents

Process for preparing 3CLpro protease inhibitors Download PDF

Info

Publication number
CN116514795A
CN116514795A CN202310029933.2A CN202310029933A CN116514795A CN 116514795 A CN116514795 A CN 116514795A CN 202310029933 A CN202310029933 A CN 202310029933A CN 116514795 A CN116514795 A CN 116514795A
Authority
CN
China
Prior art keywords
compound
formula
salt
reaction
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310029933.2A
Other languages
Chinese (zh)
Inventor
王玉兵
冷传新
房玺
刘培元
刘涛
张永晖
孔祥金
刘润来
高艺
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.)
Qilu Pharmaceutical Co Ltd
Original Assignee
Qilu Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Qilu Pharmaceutical Co Ltd filed Critical Qilu Pharmaceutical Co Ltd
Publication of CN116514795A publication Critical patent/CN116514795A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The invention provides a preparation method of a 3CLpro inhibitor, which is prepared through an amide condensation reaction and an alkylation reaction. The route has higher yield, short reaction period and simple post-treatment, and is suitable for industrial production.

Description

Process for preparing 3CLpro protease inhibitors
The present application claims priority from chinese patent application 202210111456.X, having application date 2022, 1, 29, which is incorporated herein by reference in its entirety.
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and in particular relates to a preparation method of a 3CLpro protease inhibitor.
Background
3CLPro (3C-like protease, also known as 3C-like protease) is the primary protease produced by the novel coronavirus (COVID-19, SARS-CoV-2), most of its functional proteins (non-structural proteins) are encoded by the ORF1ab gene, translated into a polyprotein (7096 aa) which is then cleaved by 3CLPro into a plurality of active proteins such as the viral replication protein RdRp. In addition, the protein may cleave the intracellular protein NEMO thereby inhibiting the activation of the interferon signaling pathway. Thus, inhibition of 3CLPro is effective in inhibiting viral infection and replication.
PCT/CN2022/117336 describes a 3CLPro protease inhibitor, wherein the compound (5-bromo-2- ((R) -1- (5- (methylamino) nicotinyl) piperidin-3-yl) amino) -3-nitrophenyl ((2R, 6S) -2, 6-dimethylmorpholinyl) methanone has good inhibition effect on SARS-CoV-23CLpro/Mpro protease, and has higher exposure and bioavailability in mice, and is expected to be developed into clinical medicines, the structure of which is shown as follows:
disclosure of Invention
The invention relates to a novel preparation method of a compound shown as a formula (I) and an intermediate thereof, which has the advantages of high reaction yield, high product purity, short reaction period, simple and convenient post-treatment and industrial application prospect.
The invention provides a preparation method of a compound shown in a formula (I) and a stereoisomer thereof, which is characterized in that,
a compound of formula (II) or a salt thereof in the presence of an acid-binding agent,
alkylation reaction with a compound of formula (III) to produce a compound of formula (I);
wherein R is as follows 1 Selected from H or C 1-4 An alkyl group;
R 2 is nitro;
R 3 and R is 4 Independently halogen.
Certain embodiments of the present invention, said R 1 Selected from H or methyl, R 2 Is nitro, R 3 And R is 4 Each independently selected from F or Br.
In certain embodiments of the present invention, the acid-binding agent is selected from one or more of triethylamine, N-diisopropylethylamine, N-methylmorpholine, pyridine, potassium carbonate, or sodium carbonate in any combination.
In certain embodiments of the present invention, the solvent used in the nucleophilic substitution reaction is selected from one or more of acetonitrile, ethanol, isopropanol, N-propanol, acetone, butanone, water, N-dimethylformamide or N, N-dimethylacetamide in any combination.
In certain embodiments of the present invention, the compound of formula (II): the molar ratio of the compound of formula (III) is 1:0.9-1: 2.
in certain embodiments of the present invention, the compound of formula (II): the molar ratio of the compound of formula (III) is 1:0.9-1:1.5.
In certain embodiments of the present invention, the compound of formula (II): the mol ratio of the acid binding agent is 1:1-1:5.
In certain embodiments of the present invention, the compound of formula (II): the mol ratio of the acid binding agent is 1:3-1:4.
In another aspect of the invention, a process for the preparation of a compound of formula (II) or a salt thereof,
the preparation method is characterized by comprising the steps of:
R 1 selected from H or C 1-4 Alkyl, preferably R 1 Selected from H or methyl.
In certain embodiments of the present invention, a process for the preparation of a compound of formula (II) or a salt thereof, characterized by comprising the steps of:
(iii) The compound of the formula (IV) and the formula (V) are subjected to condensation reaction under the condition of condensing agent and organic base to obtain a compound of the formula (VI);
(iv) The compound of formula (VI) is subjected to the action of a deprotection reagent to obtain a compound of formula (II) or a salt thereof;
the R is 5 Is an amino protecting group, R 1 Selected from H or C 1-4 Alkyl, preferably R 1 Selected from H or methyl.
Certain embodiments of the present invention, the R 5 Selected from the group consisting of benzyloxycarbonyl (Cbz), t-butyloxycarbonyl (Boc), benzyloxycarbonyl (Fmoc), p-methoxybenzyl (PMB), benzyl (Bn), trityl (Trt), p-toluenesulfonyl (Tos), phthaloyl (Pht).
In certain embodiments of the present invention, the condensing agent is selected from 2- (7-azobenzene benzotriazole) -N, N, N ', N' -tetramethyluronium Hexafluorophosphate (HATU), O-benzotriazol-tetramethyl uronium Hexafluorophosphate (HBTU), 6-chlorobenzotriazol-1, 3-tetramethyl uronium Hexafluorophosphate (HCTU), benzotriazol-1-yl-oxy-tripyrrolidinyl phosphate (PyBOP) or 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI).
In certain embodiments of the present invention, the organic base of step (i) is selected from one or more of triethylamine, N-diisopropylethylamine, or N-methylmorpholine in any combination.
In certain embodiments of the present invention, step (i) further comprises a catalyst selected from the group consisting of 4-N, N-lutidine (DMAP) and 1-hydroxybenzotriazole (HOBt).
In certain embodiments of the present invention, the compound of formula (IV) of step (i): the molar ratio of the compounds of formula (V) is 1:0.9 to 1:2.
In certain embodiments of the present invention, the compound of formula (IV) of step (i): the molar ratio of the compounds of the formula (V) is 1:0.9 to 1:1.5.
In certain embodiments of the present invention, the compound of formula (IV) of step (i): the molar ratio of the organic base is 1:2 to 1:4, preferably 1:3.
In certain embodiments of the present invention, the compound of formula (IV) of step (i): the molar ratio of the condensing agent is 1:1-1:3, preferably 1:1.5-1:3.
In certain embodiments of the present invention, the deprotecting reagent of step (ii) is selected from the group consisting of HCl/1, 4-dioxane, HCl/methanol, HCl/ethanol, trifluoroacetic acid, hydrazine hydrate, aqueous ammonia, aqueous methylamine solution, and R 5 Is tert-butoxycarbonyl (Boc) or phthaloyl (Pht), and the salt is hydrochloride or trifluoroacetate.
In another aspect of the present invention, a process for preparing a compound represented by formula (I) and stereoisomers thereof, characterized by comprising the steps of:
(a) The compound of the formula (IV) and the formula (V) are subjected to condensation reaction under the condition of condensing agent and organic base to obtain a compound of the formula (VI);
(b) The compound of formula (VI) is subjected to the action of a deprotection reagent to obtain a compound of formula (II) or a salt thereof;
(c) The compound of formula (II) or salt thereof and the compound of formula (III) are subjected to alkylation reaction in the presence of an acid binding agent to obtain the compound of formula (I).
In certain embodiments of the present invention, the process for preparing a compound of formula (III) comprises the steps of:
and (3) carrying out condensation reaction on the compound of the formula (VII) and the compound of the formula (VIII) under the conditions of a condensing agent and an organic base to obtain the compound of the formula (III).
Certain embodiments of the present invention, wherein the condensing agent is selected from 2- (7-azobenzene benzotriazole) -N, N' -tetramethyluronium Hexafluorophosphate (HATU), O-benzotriazol-tetramethyl uronium Hexafluorophosphate (HBTU), 6-chlorobenzotriazol-1, 3-tetramethyl uronium Hexafluorophosphate (HCTU), benzotriazol-1-yl-oxy-tripyrrolidinyl phosphate (PyBOP), or 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI).
Certain embodiments of the present invention, wherein the organic base is selected from one or more of triethylamine, N-diisopropylethylamine, or N-methylmorpholine or pyridine, in any combination.
Certain embodiments of the present invention, wherein the deprotecting reagent is selected from the group consisting of HCl/1, 4-dioxane, HCl/methanol, HCl/ethanol, trifluoroacetic acid, hydrazine hydrate, aqueous ammonia, and aqueous methylamine, and the salt is a hydrochloride salt or a trifluoroacetate salt.
In certain embodiments of the present invention, step (a) is performed in a solvent selected from the group consisting of one or more of N, N-dimethylformamide, N-dimethylacetamide, and N-methylpyrrolidone in any combination.
In certain embodiments of the present invention, step (a) is carried out at a reaction temperature of from-10℃to 40 ℃.
In certain embodiments of the present invention, the reaction solvent of step (b) is selected from one or more of 1, 4-dioxane, methanol, ethanol, or trifluoroacetic acid in any combination.
In certain embodiments of the present invention, step (b) is carried out at a reaction temperature of from 10℃to 40 ℃.
In certain embodiments of the present invention, step (c) is performed in a solvent selected from one or more of acetonitrile, ethanol, isopropanol, N-propanol, acetone, butanone, water, N-dimethylformamide, or N, N-dimethylacetamide in any combination.
In certain embodiments of the present invention, step (c) is carried out at a reaction temperature of 20 to 110 ℃, preferably 50 to 90 ℃.
In certain embodiments of the present invention, the step of preparing the compound of formula (III) is performed in a reaction solvent selected from any combination of one or more of N, N-dimethylformamide, N-dimethylacetamide or N-methylpyrrolidone.
In certain embodiments of the present invention, the reaction temperature for the preparation step of the compound of formula (III) is from-10℃to 40 ℃.
In certain embodiments of the present invention, the compound of formula (II) or salt thereof is selected from:
in certain embodiments of the present invention, the compound of formula (VI) is selected from:
interpretation and definition
The following terms and phrases used herein are intended to have the following meanings unless otherwise indicated. A particular term or phrase, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense.
"alkyl" refers to a straight or branched saturated hydrocarbon group. Preferably C 1-6 More preferably C 1-4 Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, and the like.
"halogen" means a fluorine, chlorine, bromine or iodine atom.
In the present invention, the amide reaction uses a condensing agent and an organic base, and the condensing agent includes, but is not limited to, carbodiimide condensing agents, carbonium salt condensing agents, phosphonium salt condensing agents, organophosphorus condensing agents, and the like.
The carbodiimide condensing agent is selected from Dicyclohexylcarbodiimide (DCC), diisopropylcarbodiimide (DIC) and 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI);
when the carbodiimide condensing agent is used, a catalyst or an activator is generally required to be added, wherein the catalyst or the activator is selected from 4-N, N-lutidine (DMAP), 1-hydroxybenzotriazole (HOBt) and the like;
the condensing agent of carbonium salts includes, but is not limited to, 2- (7-azobenzene triazole) -N, N' -tetramethyluronium Hexafluorophosphate (HATU), O-benzotriazol-tetramethyluronium Hexafluorophosphate (HBTU), 6-chlorobenzotriazol-1, 3-tetramethyluronium Hexafluorophosphate (HCTU), O- (7-azabenzotriazol-1-yl) -bis (tetrahydropyrrolyl) carbonium hexafluorophosphate (HAPyU), O- (benzotriazol-1-yl) -bis (tetrahydropyrrolyl) carbonium hexafluorophosphate (HBPyU), O- (benzotriazol-1-yl) -bis (dimethylamino) carbonium tetrafluoroborate (TBTU), O- (N-succinimidyl) -bis (dimethylamino) carbonium tetrafluoroborate (TSTU), and the like;
phosphonium salt condensing agents include, but are not limited to, benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate (PyBOP), 7-azobenzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate (PyAOP), benzotriazol-1-yloxy-tris (dimethylamino) phosphonium hexafluorophosphate (BOP);
organophosphorus condensing agents include, but are not limited to, diphenylphosphoryl chloride (DPP-Cl), diethyl cyanophosphate (DECP), diphenyl azide phosphate (DPPA), thiodimethylphosphoryl azide (MPTA), bis (2-oxo-3-oxazolidinyl) phosphoryl chloride (BOP-Cl), and the like.
In the present invention, the amino protecting group includes, but is not limited to, benzyloxycarbonyl (Cbz), t-butyloxycarbonyl (Boc), benzyloxycarbonyl (Fmoc), p-methoxybenzyl (PMB), benzyl (Bn), trityl (Trt), p-toluenesulfonyl (Tos), phthaloyl (Pht), and the like.
In the present invention, deprotectionRefers to the deprotection of amino protecting groups, and the deprotection reagents include, but are not limited to Pd-C/H 2 、PdCl 2 Na/ammonia, trifluoroacetic acid (TFA), trifluoroacetic acid/dichloromethane, HCl/1, 4-dioxane, HCl/methanol, HCl/ethanol, concentrated hydrochloric acid, sulfuric acid, hydrobromic acid, hydrazine hydrate, ammonia, aqueous methylamine solution, and the like.
In the present invention, the acid-binding agent includes an organic base or an inorganic base, wherein the inorganic base includes, but is not limited to, potassium carbonate, sodium carbonate, potassium hydroxide, sodium hydroxide, lithium hydroxide, calcium hydroxide, sodium bicarbonate, and the like.
In the present invention, the organic base includes, but is not limited to, triethylamine (TEA), N-Diisopropylethylamine (DIPEA), N-methylmorpholine (NMM), pyridine (Py), ethylenediamine (EDA), monoethanolamine (MEA), 4-N, N-Dimethylaminopyridine (DMAP), 1, 8-diazabicyclo undec-7-ene (DBU), potassium tert-butoxide, sodium tert-butoxide, N-butyllithium, tert-butyllithium, sodium bis (trimethylsilyl) amide (NaHMDS), lithium Diisopropylamide (LDA), and the like.
The isomers of the present invention include geometric and stereoisomers such as cis-trans isomers, enantiomers, diastereomers, and racemic and other mixtures thereof, all of which are within the scope of the present invention.
The term "enantiomer" refers to stereoisomers that are mirror images of each other.
The term "diastereoisomer" refers to a stereoisomer of a molecule having two or more chiral centers and having a non-mirror image relationship between the molecules.
Unless otherwise indicated, with solid wedge bondsAnd wedge-shaped dotted bond->Representing the absolute configuration of a stereogenic center.
M in the 4M hydrogen chloride-1, 4-dioxane solution is the concentration unit, represents mol/L, and the hydrogen chloride is HCl.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limiting in any way. The present invention has been described in detail herein, and specific embodiments thereof are also disclosed, it will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the invention without departing from the spirit and scope of the invention.
Summary of the laboratory instruments:
the structure of the compounds of the present invention is determined by Nuclear Magnetic Resonance (NMR) or/and liquid chromatography-mass spectrometry (LC-MS), or ultra-efficient liquid chromatography-mass spectrometry (UPLC-MS). NMR chemical shifts (δ) are given in parts per million (ppm). NMR was performed using Bruker Neo 400M or Bruker Ascend 400 nuclear magnetic instruments with deuterated dimethyl sulfoxide (DMSO-d 6) as the solvent, deuterated methanol (CD) 3 OD) and deuterated chloroform (CDCl) 3 ) Heavy water (D) 2 O), internal standard is Tetramethylsilane (TMS).
LC-MS was performed using Agilent 1260-6125B single quadrupole mass spectrometer for determination of LC-MS, column Welch Biomate column (C18, 2.7 μm, 4.6X150 mm) or waters H-Class SQD2, column Welch Ultimate column (XB-C18, 1.8 μm, 2.1X150 mm) mass spectrometer (ion source electrospray ionization).
Ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) was performed using a Waters UPLC H-class SQD mass spectrometer (electrospray ionization as the ion source).
HPLC determinations used Waters e2695-2998 or Waters ARC and Agilent 1260 or Agilent Poroshell HPH high performance liquid chromatography.
The thin layer chromatography silica gel plate uses smoke table Jiang You silica gel to develop GF254 silica gel plate of the limited company or GF254 silica gel plate of the new material of the limited company on the opal market, the specification adopted by TLC is 0.15 mm-0.20 mm, the preparation is 20 multiplied by 20cm, and column chromatography is generally used for forming 200-300 mesh silica gel as a carrier.
The starting materials in the examples of the present invention are known and commercially available or may be synthesized using or according to methods known in the art.
All reactions of the invention were carried out under continuous magnetic stirring under dry nitrogen or argon atmosphere, with the solvent being a dry solvent, and the reaction temperature being in degrees celsius or in degrees celsius, without specific description. Room temperature refers to 25±5 ℃ unless otherwise specified.
Example 1 preparation of the compound (5-bromo-2- ((R) -1- (5- (methylamino) nicotinyl) piperidin-3-yl) amino) -3-nitrophenyl ((2R, 6S) -2, 6-dimethylmorpholinyl) methanone of formula (I)
The first step: preparation of Compound I-2
Method 1:
compound I-1 (5 g,32.9 mmol) was dissolved in N, N-dimethylformamide (50 mL) at room temperature under nitrogen, cooled to 0deg.C, and N, N-diisopropylethylamine (12.7 g,98.4 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (abbreviation HATU,18.7g,49.2 mmol) and (R) -3-Boc-aminopiperidine (9.9 g,49.4 mmol) were added sequentially. The reaction was stirred for a further 15 minutes at 0 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction with water (150 mL), extract with ethyl acetate (50 ml×3), combine the organic phases, wash the organic phases with saturated brine (50 ml×2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue obtained was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-2, (8.8 g, yield 80.1%, R 5 Boc).
MS(ESI)M/Z:335.3[M+H] +
1 H NMR(400MHz,DMSO):δ=8.09~8.47(m,4H);7.22~7.35(m,1H);3.60~3.75(m,4H);3.29~3.38(m,1H);2.80~3.07(m,4H);1.53~1.79(m,2H)。
Method 2:
under the condition of room temperature and nitrogen protection, the compound I-1 (5 g,32.9 mmol) is dissolved in N, N-dimethylacetamide (50 mL), the temperature is reduced to 10 ℃, and triethylamine (10.0 g,98.8 mmol) and O-benzotriazole-tetramethylurea hexafluorophosphoric acid are added in sequenceEsters (abbreviated HBTU,18.7g,49.3 mmol) and (R) -3-Boc-aminopiperidine (9.9 g,49.4 mmol). The reaction was stirred for a further 15 minutes at 10 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction with water (150 mL), extract with ethyl acetate (50 ml×3), combine the organic phases, wash the organic phases with saturated brine (50 ml×2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue obtained was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-2, (8.5 g, yield 79.2%, R 5 Boc).
Method 3:
compound I-1 (1 g,6.6 mmol) was dissolved in N-methylpyrrolidone (10 mL) at room temperature under nitrogen, followed by N-methylmorpholine (2.0 g,19.8 mmol), benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate (abbreviation PyBOP,5.1g,9.8 mmol) and (R) -3-Boc-aminopiperidine (1.6 g,8.0 mmol). The reaction was stirred for a further 15 minutes at 30 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction with water (30 mL), extract with ethyl acetate (10 ml×3), combine the organic phases, wash the organic phases with saturated brine (10 ml×2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue obtained was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-2, (1.7 g, yield 77.3%, R 5 Boc).
Method 4:
compound I-1 (0.5 g,3.3 mmol) was dissolved in N, N-dimethylformamide (5 mL) at room temperature and cooled to-10deg.C, N-diisopropylethylamine (1.3 g,10.1 mmol), 1-hydroxybenzotriazole (abbreviated HOBt,0.7g,5.2 mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (abbreviated EDCI,1.0g,5.2 mmol) and (R) -3-Boc-aminopiperidine (0.6 g,3.0 mmol) were added sequentially under nitrogen. The reaction was stirred for a further 15 minutes at-10 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction with water (15 mL), extract with ethyl acetate (10 ml×3), combine the organic phases, wash the organic phases with saturated brine (10 ml×2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue obtained is purified by column chromatography on silica gel (eluent: petroleum ether/acetic acid)Ethyl ester=4/1, volume ratio) to give compound I-2, (0.8 g, yield 72.8%, R 5 Boc).
Method 5:
compound I-1 (1.0 g,6.6 mmol) was dissolved in N, N-dimethylformamide (10 mL) at room temperature under nitrogen, cooled to 0deg.C, and N, N-diisopropylethylamine (2.6 g,20 mmol), 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (abbreviated HATU,3.8g,10 mmol) and 3- (R) -piperidylphthalimide (2.2 g,9.6 mmol) were added sequentially. The reaction was stirred for a further 15 minutes at 0 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction with water (30 mL), extract with ethyl acetate (10 ml×3), combine the organic phases, wash the organic phases with saturated brine (10 ml×2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue obtained was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-2 (1.9 g, yield 79.3%, R) 5 Pht).
And a second step of: preparation of Compound I-3
Method 1:
compound I-2 (8 g,23.9mmol, R) is purged with nitrogen at room temperature 5 Boc) was dissolved in 4M hydrogen chloride-1, 4-dioxane solution (80 mL) and stirred for 1 hour. After LCMS monitoring shows the disappearance of the starting material, the reaction solution was concentrated by distillation under reduced pressure, methyl tert-butyl ether (120 mL) was added, stirred for crystallization, suction filtration to give a filter cake, and oven-drying to give the hydrochloride salt of compound I-3 (5.8 g, 89.5% yield).
MS(ESI)M/Z:235.2[M+H-HCl] +
1 H NMR(400MHz,DMSO):δ=7.96(s,2H);7.18(s,1H);6.96~6.99(d,1H);3.50~3.65(m,2H);2.81~2.98(m,4H);1.99(s,3H);1.45(s,9H);1.38(s,4H)。
Method 2:
compound I-2 (1 g,3.0mmol, R) is reacted under nitrogen 5 Boc) was dissolved in methanol hydrogen chloride (10 mL) and stirred at 30℃for 1 hour. After LCMS monitoring shows that the raw materials disappear, the reaction solution is distilled and concentrated under reduced pressure, methyl tert-butyl ether (20 mL) is added, stirred for crystallization and suction filtration is carried out to obtainTo the cake, the hydrochloride salt of compound I-3 (0.70 g, 86.5% yield) was obtained by drying.
Method 3:
compounds of formula I-2 (1 g,3.0mmol, R) 5 Boc) was dissolved in ethanol hydrogen chloride (10 mL) and stirred at 10deg.C for 1 hour. After LCMS monitoring shows the disappearance of the starting material, the reaction solution was concentrated by distillation under reduced pressure, methyl tert-butyl ether (20 mL) was added, stirred for crystallization, suction filtration to give a filter cake, and oven-drying to give the hydrochloride salt of compound I-3 (0.72 g, 89.0% yield).
Method 4:
compound I-2 (1 g,3.0mmol, R) is reacted under nitrogen 5 Boc) was dissolved in trifluoroacetic acid (10 mL) and stirred at 40℃for 1 hour. After LCMS monitoring shows the disappearance of the starting material, the reaction solution was concentrated by distillation under reduced pressure, methyl tert-butyl ether (20 mL) was added, stirred for crystallization, suction filtration to give a filter cake, which was dried to give the trifluoroacetate salt of Compound I-3 (0.84 g, yield 80.6%).
Method 5:
compound I-2 (1 g,2.6mmol, R) is reacted under nitrogen 5 Pht), 10mL of methanol was added, hydrazine hydrate (0.3 mL) was added, and the mixture was stirred at 10 ℃ for 3 hours. After the disappearance of the starting material by LCMS monitoring, the reaction solution was concentrated by distillation under reduced pressure, ethyl acetate (20 mL) was added, and dissolved by stirring, the organic phase was washed with saturated brine (10 ml×2), dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and distilled under reduced pressure to give compound I-3 (0.54 g, yield 88.1%).
Method 6:
compounds of formula I-2 (1 g,2.6mmol, R) 5 Pht), 10mL of aqueous ammonia was added thereto, and the mixture was stirred at 30℃for 3 hours. After the disappearance of the starting material by LCMS, the reaction solution was concentrated by distillation under reduced pressure, ethyl acetate (20 mL) was added, the solution was separated, the organic phase was washed with saturated brine (10 ml×2), dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and distilled under reduced pressure to give compound I-3 (0.50 g, yield 81.6%).
Method 7:
compound I-2 (1 g,2.6mmol, R) is reacted under nitrogen 5 Pht), 10mL of aqueous methylamine solution was added thereto, and stirred at 40℃for 3 hoursWhen (1). After LCMS monitoring showed the disappearance of starting material, the reaction solution was concentrated by distillation under reduced pressure, ethyl acetate (20 mL) was added, the solution was separated, the organic phase was washed with saturated brine (10 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Distillation under the reduced pressure gave compound I-3 (0.52 g, yield 84.9%).
And a third step of: preparation of Compound I-5
Method 1:
5-bromo-2-fluoro-3-nitrobenzoic acid (compound I-4,9g,34.1 mmol) was dissolved in N, N-dimethylformamide (90 mL) at room temperature under nitrogen, cooled to 0 ℃, N-diisopropylethylamine (13.2 g,102.1 mmol), (2- (7-azobenzotriazol) -N, N' -tetramethylurea hexafluorophosphate (abbreviation HATU,18g,47.3 mmol) and (2 r,6 s) -2, 6-dimethylmorpholine (4.7 g,40.8 mmol) were added in sequence, the reaction system was stirred continuously at 0 ℃ for 15 minutes, LCMS monitoring showed that the starting material disappeared, water (200 mL) was added to the reaction mixture was quenched, the mixture was extracted with ethyl acetate (100 ml×3), the organic phase was combined, washed with saturated brine (100 ml×2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, the resulting residue was purified by column chromatography (eluent: ethyl acetate=4.7/1 vol.% (9.9.9% yield of compound I).
MS(ESI)M/Z:361.0[M+H] +
1H NMR(400MHz,CDCl3):δ=8.25~8.27(m,1H);7.82(s,1H);4.55~4.60(m,1H);3.67(s,2H);3.20~3.24(m,1H);2.95(s,1H);2.58~2.64(t,1H);1.28~1.30(d,3H);1.16~1.18(d,2H)。
Method 2:
5-bromo-2-fluoro-3-nitrobenzoic acid (Compound I-4,1.0g,3.8 mmol) was dissolved in N, N-dimethylacetamide (10 mL) at room temperature under nitrogen, cooled to 10℃and triethylamine (1.1 g,10.9 mmol), O-benzotriazol-tetramethylurea hexafluorophosphate (abbreviated HBTU,2.2g,5.8 mmol) and (2R, 6S) -2, 6-dimethylmorpholine (0.5 g,4.3 mmol) were added sequentially. The reaction was stirred for a further 15 minutes at 10 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction mixture by adding water (20 mL), extract the mixture with ethyl acetate (10 mL. Times.3), combine the organic phases, wash the organic phases with saturated brine (10 mL. Times.2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-5 (1.0 g, yield 73.1%).
Method 3:
5-bromo-2-fluoro-3-nitrobenzoic acid (Compound I-4,1.0g,3.8 mmol) was dissolved in N-methylpyrrolidinone (10 mL) at room temperature under nitrogen, cooled to 20℃and N-methylmorpholine (1.1 g,10.9 mmol), benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate (abbreviation PyBOP,3.0g,5.8 mmol) and (2R, 6S) -2, 6-dimethylmorpholine (0.5 g,4.3 mmol) were added sequentially. The reaction was stirred for a further 15 minutes at 20 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction mixture by adding water (20 mL), extract the mixture with ethyl acetate (10 mL. Times.3), combine the organic phases, wash the organic phases with saturated brine (10 mL. Times.2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-5 (1.1 g, yield 80.4%).
Method 4:
5-bromo-2-fluoro-3-nitrobenzoic acid (Compound I-4,1g,3.8 mmol) was dissolved in N, N-dimethylformamide (10 mL) under nitrogen, and N, N-diisopropylethylamine (1.5 g,11.6 mmol), 1-hydroxybenzotriazole (abbreviated HOBt,0.8g,5.9 mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (abbreviated EDCI,1.1g,5.7 mmol) and (2R, 6S) -2, 6-dimethylmorpholine (0.5 g,4.3 mmol) were added sequentially. The reaction was stirred for a further 15 minutes at 30 ℃. After LCMS monitoring showed the disappearance of starting material, quench the reaction mixture by adding water (20 mL), extract the mixture with ethyl acetate (10 mL. Times.3), combine the organic phases, wash the organic phases with saturated brine (10 mL. Times.2), dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure. The residue was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-5 (1.0 g, yield 73.1%).
Fourth step: preparation of Compound (I)
Method 1:
compound I-3 (5.8 g,21.4 mmol) was dissolved in acetonitrile (60 mL) at room temperature under nitrogen protection, compound I-5 (9.3 g,25.7 mmol) and N, N-diisopropylethylamine (8.3 g,64.2 mmol) were added sequentially, the temperature was raised to 80 ℃, the reaction was stirred for 1h, after LCMS monitoring showed that compound I-3 disappeared, water (200 mL) was added to the reaction solution to quench, the mixture was extracted with ethyl acetate (100 mL. Times.3 times), the organic phases were combined, and the organic phase was washed with saturated brine (100 mL. Times.2 times). Then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30/1, volume ratio) to obtain compound (I) (11.0 g, yield 89.2%).
MS(ESI)M/Z:575.2[M+H] +
1H NMR(400MHz,DMSO-d6)δ=8.27(s,1H),7.67~8.00(m,4H),6.81(s,1H),6.27(s,1H),4.39(s,1H),3.08~3.91(m,10H),2.50~2.86(m,3H),1.34~1.93(m,4H),1.11~1.25(m,3H),1.03(s,3H)。
Method 2:
compound I-3 (1.0 g,3.7 mmol) was dissolved in ethanol (10 mL) at room temperature under nitrogen protection, compound I-5 (2.0 g,5.5 mmol) and triethylamine (1.1 g,10.9 mmol) were added sequentially, the temperature was raised to 50℃and the reaction was stirred for 1h, after LCMS monitoring showed that compound I-3 disappeared, water (20 mL) was added to the reaction solution to quench, the mixture was extracted with ethyl acetate (10 mL. Times.3 times), the organic phases were combined, and the organic phase was washed with saturated brine (10 mL. Times.2 times). Then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30/1, volume ratio) to give compound (I) (1.6 g, yield 75.3%).
Method 3:
compound I-3 (1.0 g,3.7 mmol) was dissolved in acetone (10 mL) at room temperature under nitrogen protection, compound I-5 (1.6 g,4.4 mmol) and N-methylmorpholine (1.1 g,10.9 mmol) were added sequentially, the temperature was raised to 90℃and the reaction was stirred for 1h, LCMS monitoring showed that compound I-3 disappeared, then water (20 mL) was added to the reaction solution to quench, the mixture was extracted with ethyl acetate (10 mL. Times.3 times), the organic phases were combined, and the organic phase was washed with saturated brine (10 mL. Times.2 times). Then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30/1, volume ratio) to obtain compound (I) (1.9 g, yield 89.4%).
Method 4:
compound I-3 (trifluoroacetate salt, 1.0g,3.0 mmol) was added to purified water (10 mL) at room temperature under nitrogen, compound I-5 (1.6 g,4.4 mmol) and N, N-diisopropylethylamine (1.4 g,10.8 mmol) were added sequentially, the temperature was raised to 80 ℃, the reaction was stirred for 1h, after LCMS monitoring showed that compound I-3 disappeared, water (20 mL) was added to the reaction solution, extracted with ethyl acetate (10 mL. Times.3 times), the organic phases were combined, and the organic phase was washed with saturated brine (10 mL. Times.2 times). Then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30/1, volume ratio) to give compound (I) (1.2 g, yield 72.6%).
Method 5:
compound I-3 (1.0 g,3.7 mmol) was added to N, N-dimethylformamide (10 mL) under nitrogen at room temperature, compound I-5 (1.2 g,3.3 mmol) and N, N-diisopropylethylamine (1.4 g,10.8 mmol) were added sequentially, the temperature was raised to 80℃and the reaction was stirred for 1h, after LCMS monitoring showed that compound I-3 disappeared, water (20 mL) was added to the reaction solution, extracted with ethyl acetate (10 mL. Times.3 times), the organic phases were combined, and the organic phase was washed with saturated brine (10 mL. Times.2 times). Then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30/1, volume ratio) to give compound (I) (1.5 g, yield 70.6%).
Comparative example
Preparation of Compound I-2
5-methylamino nicotinic acid (compound I-1,1.0g,6.6 mmol) was added to 10ml of thionyl chloride, reacted at 50℃for 3h, and then reduced to give 5-methylamino nicotinoyl chloride; 5-Methylaminonicotinoyl chloride was added to 5mL of tetrahydrofuran, a mixed solution of (R) -3-Boc-aminopiperidine (2.0 g,10.0 mmol)/triethylamine (2.0 g,19.8 mmol)/5 mL of tetrahydrofuran was added dropwise, the TLC detection reaction was not performed after 3 hours, the reaction solution was added to 20mL of purified water, extracted with ethyl acetate (10 mL. Times.3 times), the organic phases were combined, the organic phases were washed with saturated brine (10 mL. Times.2 times), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-2, (0.24 g, yield 10.9%).
Preparation of Compound I-5
5-bromo-2-fluoro-3-nitrobenzoic acid (compound I-4,1.0g,3.8 mmol) was added to 10mL thionyl chloride, reacted at 50℃for 3h, and then reduced to give 5-bromo-2-fluoro-3-nitrobenzoyl chloride; 5-bromo-2-fluoro-3-nitrobenzoyl chloride was added to 5mL of tetrahydrofuran, and (2R, 6S) -2, 6-dimethylmorpholine (0.5 g,4.3 mmol)/triethylamine (1.2 g,11.8 mmol)/5 mL of a tetrahydrofuran mixed solution was added dropwise, after 3 hours of reaction, the TLC detection reaction was no longer carried out, the reaction solution was added to 20mL of purified water, extracted with ethyl acetate (10 mL. Times.3 times), the organic phases were combined, the organic phases were washed with saturated brine (10 mL. Times.2 times), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=4/1, volume ratio) to give compound I-5 (0.12 g, yield 8.8%).
Biological test evaluation
Test example 1: evaluation of the inhibitory Effect of Compounds of formula (I) on SARS-CoV-23CLpro/Mpro target
The experiment adopts fluorescence resonance energy transfer method to detect SARS-CoV-23CLpro/Mpro protease activity, and obtains half inhibition concentration IC of compound to SARS-CoV-23CLpro/Mpro protease 50
A novel coronavirus Mpro/3CLpro inhibitor screening kit (P0315M) was purchased from Beyotime company.
Preparing enzyme solution by using reaction buffer solution, adding 4 in each well9.5. Mu.L of enzyme solution; 49.5. Mu.L of reaction buffer was added to Min wells. Compound detection IC 50 The final concentration of the test was 10. Mu.M, 3-fold dilution, 10 concentrations, each concentration set up a multiplex well test. The test compound was diluted to 200-fold final concentration and added to 384-well reaction plates using a D300e (TECAN) ultra microscale sampler with a 250nL gradient of diluted test compound. 250nL of 100% DMSO was transferred in both Max wells and Min wells. Incubate on ice for 10 minutes. 250nL of substrate solution was added to each well using D300e (TECAN). The reaction plate was centrifuged at 1000rpm for 1min and the fluorescent signal was continuously read for 30min using an Envision microplate reader (PerkinElmer). Data analysis using GraphPad Prism 8 software, calculation of IC for compounds 50
IC of the compound of formula (I) as determined experimentally 50 The value is 26nM, and has good inhibition activity on SARS-CoV-23CLpro/Mpro protease.
Test example 2: in vivo pharmacokinetic experiments in CD1 mice
The in vivo pharmacokinetic behavior of the compounds of the invention on male CD1 mice was studied using male CD1 mice as test animals.
Test article: control compounds and compounds of formula (I).
Test animals
Species: male CD1 mice (3/group)
Grade: SPF stage
Weight of: about 20 g to 30g
Age: for 6 to 8 weeks
The source is as follows: weitong Lihua (Chinese character)
The test solution was administered to male CD1 mice by gavage at a dose of 10mg/kg and a volume of 10mL/kg.
Preparing a test solution: the test sample is dissolved by solvent (10% DMSO+50% PEG400+40% purified water), and DMSO, PEG400 and purified water are sequentially added during preparation.
Control group: the structural formula of the CN 113072497A patent compound 2 is as follows:
experimental group: a compound of formula (I).
About 0.025 to 0.03mL of whole blood (anticoagulated with EDTA-K2) was collected from the dorsum veins of the mice at the set blood collection time points before (0 h) and after (0.25 h), 0.5h,1h,2h,4h,8h,24h, respectively, and placed under wet ice conditions, and centrifuged at 4000g centrifugal force at 4℃for 5min within 30min after collection, and plasma was separated and transferred to a test tube.
The pharmacokinetic parameters were calculated using WinNonlin (Phoenix. TM., version 6.1) and are shown in tables 1 and 2.
TABLE 1 pharmacokinetic parameters for intravenous administration in CD1 mice
Table 2 pharmacokinetic parameters for oral administration in CD1 mice
Remarks: NA represents no corresponding data.
The results show that: compared with a control group, the compound of the formula (I) has higher exposure and bioavailability in mice and good pharmacokinetic property.
Test example 3: in vivo pharmacokinetic experiments in cynomolgus monkeys
The pharmacokinetic behavior of the compounds of formula (I) in vivo plasma of cynomolgus monkeys was studied by oral administration at 10mg/kg in male cynomolgus monkeys as the subject animals.
1. Test protocol
1.1 test article:
a compound of formula (I).
1.2 test animals
Basic information of animals
Species: macaca fascicularis monkey
Grade: common grade
Quantity: experiment with 18 cynomolgus monkeys (Male) for administration
Weight of: about 2.5 kg to about 4.5kg
Age: about 3 to 5 years old
The source is as follows: from Huazhen animal farm (common partner)
1.3 administration:
9 male cynomolgus monkeys, 3 animals/group, 3 total groups. The animals were fasted for 12 hours before administration, and the test solution (10 mg/kg) was administered by gavage, with a volume of administration of 2.5mL/kg.
Preparing a test solution: the test sample is dissolved by 10% DMSO+50% PEG400+40% purified water, and DMSO, PEG400 and purified water are sequentially added during preparation.
1.4 sample collection:
before (0 min), after (11) 0.25h,0.5h,1h,2h,4h,6h,8h,12h,24h and 48h, respectively.
Collecting blood from the vein of monkey hind limb at about 1.5mL according to the set blood collection time point, placing under wet ice condition, and collecting
Centrifuging at 1700g for 1.5h, and centrifuging at 4deg.C for 10min. After centrifugation, plasma was taken and added to the test tube. The plasma samples can be stored in a refrigerator below-20 ℃ within 2 hours after centrifugation.
2. Experimental results and analysis
The main pharmacokinetic parameters were calculated using WinNonlin 8.1.0.3530, and the results showed that the compound of formula (I) had good pharmacokinetic properties.
Table 3 pharmacokinetic parameters for oral administration of cynomolgus monkeys
/>

Claims (23)

1. A process for producing a compound represented by the formula (I) and stereoisomers thereof, which comprises the steps of,
a compound of formula (II) or a salt thereof in the presence of an acid-binding agent,
alkylation reaction with a compound of formula (III) to produce a compound of formula (I);
wherein R is as follows 1 Selected from H or C 1-4 An alkyl group;
R 2 is nitro;
R 3 and R is 4 Independently halogen.
2. The method of claim 1, wherein R 1 Selected from H or methyl, R 2 Is nitro, R 3 And R is 4 Independently selected from F or Br.
3. The method according to claim 1 or 2, wherein the acid-binding agent is selected from one or more of triethylamine, N-diisopropylethylamine, N-methylmorpholine, pyridine, potassium carbonate or sodium carbonate.
4. The process according to claim 1 or 2, wherein the solvent used in the alkylation reaction is selected from one or more of acetonitrile, ethanol, isopropanol, N-propanol, acetone, butanone, water, N-dimethylformamide or N, N-dimethylacetamide.
5. The method according to claim 1 or 2, wherein the compound of formula (II): the molar ratio of the compound of formula (III) is 1:0.9-1: 2, preferably, said compound of formula (II): the molar ratio of the compound of formula (III) is 1:0.9-1:1.5.
6. A process for the preparation of a compound of formula (II) or a salt thereof,
the preparation method is characterized by comprising the steps of:
R 1 the definition of claim 1 or 2.
7. The method according to claim 6, characterized by the steps of:
(i) The compound of the formula (IV) and the formula (V) are subjected to condensation reaction under the condition of condensing agent and organic base to obtain a compound of the formula (VI);
(ii) The compound of formula (VI) is subjected to the action of a deprotection reagent to obtain a compound of formula (II) or a salt thereof;
the R is 5 Is an amino protecting group, R 1 The definition of claim 1 or 2.
8. The process of claim 7 wherein the condensing agent is selected from 2- (7-azobenzene triazole) -N, N' -tetramethyl urea Hexafluorophosphate (HATU), O-benzotriazole-tetramethyl urea Hexafluorophosphate (HBTU), 6-chlorobenzotriazole-1, 3-tetramethyl urea Hexafluorophosphate (HCTU), benzotriazol-1-yl-oxy-tripyrrolidinylphosphine (PyBOP), or 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI).
9. The process of claim 7, wherein the organic base of step (i) is selected from one or more of triethylamine, N-diisopropylethylamine, or N-methylmorpholine in any combination.
10. The process of claim 7 wherein step (i) further comprises a catalyst selected from the group consisting of 4-N, N-lutidine (DMAP) and 1-hydroxybenzotriazole (HOBt).
11. The method of claim 7, wherein the compound of formula (IV) of step (i): the molar ratio of the compounds of the formula (V) is from 1:0.9 to 1:2, preferably,
a compound of formula (IV) of step (i): the molar ratio of the compounds of formula (V) is: 1:0.9-1:1.5.
12. The process according to claim 7, wherein the deprotecting reagent of step (ii) is selected from the group consisting of HCl/1, 4-dioxane, HCl/methanol, HCl/ethanol, trifluoroacetic acid, hydrazine hydrate, aqueous ammonia or aqueous methylamine solution, and R is 5 Selected from tert-butoxycarbonyl (Boc) or phthaloyl (Pht), said salt being a hydrochloride or trifluoroacetate salt.
13. The preparation method of the compound shown in the formula (I) and the stereoisomer thereof is characterized by comprising the following steps:
(a) The compound of the formula (IV) and the formula (V) are subjected to condensation reaction under the condition of condensing agent and organic base to obtain a compound of the formula (VI);
(b) The compound of formula (VI) is subjected to the action of a deprotection reagent to obtain a compound of formula (II) or a salt thereof;
(c) The compound of formula (II) or salt thereof and the compound of formula (III) are subjected to alkylation reaction in the presence of an acid binding agent to obtain the compound of formula (I).
14. The process according to claim 13, the process for the preparation of the compound of formula (III) comprising the steps of:
and (3) carrying out condensation reaction on the compound of the formula (VII) and the compound of the formula (VIII) under the conditions of a condensing agent and an organic base to obtain the compound of the formula (III).
15. The process according to claim 13 or 14, wherein the condensing agent is selected from 2- (7-azobenzotriazole) -N, N' -tetramethylurea Hexafluorophosphate (HATU), O-benzotriazol-tetramethylurea Hexafluorophosphate (HBTU), 6-chlorobenzotriazol-1, 3-tetramethylurea Hexafluorophosphate (HCTU), benzotriazol-1-yl-oxy-tripyrrolidinylphosphine (PyBOP) or 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI).
16. The process according to claim 13 or 14, wherein the organic base is selected from one or more of triethylamine, N-diisopropylethylamine or N-methylmorpholine or pyridine in any combination.
17. The method of claim 13, wherein the deprotecting reagent is selected from the group consisting of HCl/1, 4-dioxane, HCl/methanol, HCl/ethanol, trifluoroacetic acid, hydrazine hydrate, aqueous ammonia, aqueous methylamine, and the salt is a hydrochloride salt or a trifluoroacetate salt.
18. The process of claim 15, wherein step (a) is performed in a solvent selected from the group consisting of N, N-dimethylformamide, N-dimethylacetamide, and N-methylpyrrolidone, in any combination thereof.
19. The process of claim 13, wherein step (b) is performed in a solvent selected from the group consisting of 1, 4-dioxane, methanol, ethanol, and trifluoroacetic acid.
20. The process of claim 13, wherein step (c) is performed in a reaction solvent selected from one or more of acetonitrile, ethanol, isopropanol, N-propanol, acetone, butanone, water, N-dimethylformamide, or N, N-dimethylacetamide in any combination.
21. The process according to claim 14, wherein the reaction solvent is selected from the group consisting of N, N-dimethylformamide, N-dimethylacetamide and N-methylpyrrolidone.
22. The compound of formula (II) or salt thereof is selected from:
23. the compound of formula (VI) is selected from:
CN202310029933.2A 2022-01-29 2023-01-10 Process for preparing 3CLpro protease inhibitors Pending CN116514795A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210111456X 2022-01-29
CN202210111456 2022-01-29

Publications (1)

Publication Number Publication Date
CN116514795A true CN116514795A (en) 2023-08-01

Family

ID=87405257

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310029933.2A Pending CN116514795A (en) 2022-01-29 2023-01-10 Process for preparing 3CLpro protease inhibitors

Country Status (1)

Country Link
CN (1) CN116514795A (en)

Similar Documents

Publication Publication Date Title
JP6861858B2 (en) SSAO inhibitor
US9193758B2 (en) Conformationally-preorganized, miniPEG-containing γ-peptide nucleic acids
JP5727937B2 (en) Method for synthesizing benzoacepine derivatives
PT2873665T (en) Antiviral compounds
JP6986653B2 (en) Disubstituted pyrazole compound as a ketohexokinase inhibitor
WO2024051720A1 (en) 5-pyridine-1h-indazole compound for targeted inhibition of clk2 and use of compound
JP4012145B2 (en) Solid phase synthesis of pyrrole-imidazole polyamide
Amant et al. Synthesis and oligomerization of Fmoc/Boc-protected PNA monomers of 2, 6-diaminopurine, 2-aminopurine and thymine
RU2715233C2 (en) Method of producing nitrogen mustard derivatives
JPH07112969A (en) Nucleic-acid-combinable oligomers with c-branch for treatment and diagnosis
CN114276333B (en) Dihydroquinoxaline bromodomain bivalent inhibitors
JP5476541B2 (en) Telomerase inhibitor
CN116514795A (en) Process for preparing 3CLpro protease inhibitors
TW202022117A (en) Method for producing hairpin single-stranded rna molecules
JP2019505495A (en) Substituted naphthalene diimide and use thereof
EP1923397B1 (en) Fluorinated amino acids and peptides
WO2018113277A1 (en) Method for preparing ledipasvir and intermediate for preparing ledipasvir
CN117486786A (en) Preparation method of novel morpholine derivative
CN117486871A (en) Process for preparing morpholine derivatives
CN114573567B (en) Indazole cyclotriazole compound and preparation method and application thereof
CN114573562B (en) Niacin-containing triazole compound and preparation method and application thereof
CN115340499B (en) BCL-XL inhibitors and uses thereof
US12006291B2 (en) Processes for the preparation of 4,6,7-trifluoro-1H-indole-2-carboxylic acid
CN112961081B (en) Bibenzamide urea compound and preparation method and application thereof
WO2023088493A1 (en) Furo-pyridone compound and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication