WO2018233670A1 - Benzoisoxazole spiropyrimidinetrione compound, preparation method therefor and use thereof - Google Patents

Benzoisoxazole spiropyrimidinetrione compound, preparation method therefor and use thereof Download PDF

Info

Publication number
WO2018233670A1
WO2018233670A1 PCT/CN2018/092208 CN2018092208W WO2018233670A1 WO 2018233670 A1 WO2018233670 A1 WO 2018233670A1 CN 2018092208 W CN2018092208 W CN 2018092208W WO 2018233670 A1 WO2018233670 A1 WO 2018233670A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
oxo
formula
nmr
Prior art date
Application number
PCT/CN2018/092208
Other languages
French (fr)
Chinese (zh)
Inventor
杨玉社
张银勇
石程辉
陈乾
Original Assignee
中国科学院上海药物研究所
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
Priority claimed from CN201711116329.4A external-priority patent/CN109111466B/en
Application filed by 中国科学院上海药物研究所 filed Critical 中国科学院上海药物研究所
Publication of WO2018233670A1 publication Critical patent/WO2018233670A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings

Definitions

  • the present invention relates to a benzopyrazole group-containing spiropyrimidine compound, a pharmaceutical composition thereof, a process for the preparation thereof and use thereof in an anti-infective drug.
  • Benzoisoxazole spirotriones are a new class of bacterial type II topoisomerase inhibitors. Although they interact with quinolones for bacterial type II topoisomerases, their specific regions of action and The mechanism of action is completely different.
  • the novel chemical structure and new mechanism of action of benzoisoxazole spiropyrimidines have good antibacterial activity against sensitive and resistant Gram-positive bacteria, and there is no cross-resistance with clinical antibacterial drugs. Medicinal.
  • AstraZeneca and other companies reported a series of spirotriazol compounds, and a preliminary study on its structure-activity relationship found that the methyl-substituted spiropyrimidine compounds on isoxazole have good antibacterial activity.
  • AstraZeneca continued to conduct derivatization studies on such compounds, and found AZD0914 with better activity, while eliminating the genotoxicity and bone marrow toxicity of such compounds.
  • AZD0914 is undergoing Phase II clinical studies.
  • spirotrione compounds such as AZD0914
  • AZD0914 still have the problem that the antibacterial activity is not strong and the metabolic properties are not ideal, resulting in a large amount of clinical drug overdose, which limits the possibility of treating the systemic infection drug to some extent.
  • the object of the present invention is to provide a benzoisoxazole spiropyrimidine compound having better activity and superior metabolic properties, the enantiomers, diastereomers, racemates and mixtures thereof. And its pharmaceutically acceptable salts.
  • R 1 is hydrogen, halogen or cyano
  • R 2 and R 3 are independently a hydrogen atom or a C 1 -C 3 alkyl group; or R 2 and R 3 together with a carbon atom to be bonded form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring;
  • R 4 is a hydrogen atom or a C 1 -C 3 alkyl group
  • R 1 is a halogen
  • R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
  • the general formula (I) of the present invention means that the compound contains at least three chiral centers in the presence of enantiomers and diastereomers.
  • two enantiomers can be obtained using a general chiral resolution method or an asymmetric synthesis method.
  • separation can be carried out by fractional recrystallization or chromatography.
  • the compound represented by the formula (I) of the present invention includes any one of the above isomers or a mixture thereof.
  • R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
  • R 2 and R 3 are simultaneously hydrogen or hydrogen at the same time.
  • R 1 is hydrogen, fluorine, chlorine or cyano.
  • R 1 is hydrogen, fluorine or chlorine; and R 2 and R 3 together with the attached carbon atoms form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
  • R 1 is hydrogen, fluorine or chlorine; and R 4 is hydrogen.
  • R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring; and R 4 is hydrogen.
  • R 1 is a cyano group
  • R 2 and R 3 are independently a hydrogen atom or a C 1 -C 3 alkyl group; or R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring.
  • the compound is:
  • a pharmaceutical composition comprising the compound of the first aspect or an enantiomer thereof, a diastereomer, a racemate, and a mixture thereof, Or a pharmaceutically acceptable salt thereof;
  • a pharmaceutically acceptable carrier or excipient is provided.
  • the present invention provides a novel benzisoxazole spiropyrimidine compound which can be used alone or mixed with a pharmaceutically acceptable adjuvant (for example, an excipient, a diluent, etc.) to prepare a tablet for oral administration. , capsules, granules or syrups, etc.
  • a pharmaceutically acceptable adjuvant for example, an excipient, a diluent, etc.
  • the pharmaceutical composition can be obtained by a conventional method of pharmacy.
  • a third aspect of the invention provides a process for the preparation of the compound of the first aspect, the method comprising the steps of:
  • intermediate Ib is nucleophilic substituted to form intermediate Ic
  • intermediate Ic is catalytically hydrolyzed to form intermediate Id;
  • R 1 , R 2 , R 3 and R 4 are as defined above.
  • intermediate Ia is reacted with cesium carbonate by intramolecular nucleophilic substitution to form intermediate Ib.
  • the intermediate Ib is reacted with N,N'-carbonyldiimidazole by nucleophilic substitution to form the intermediate Ic.
  • the intermediate Id is reacted with 2R,6R-dimethylmorpholine by nucleophilic substitution to form the intermediate Ie.
  • the intermediate Ic is hydrolyzed by hydrochloric acid to form an intermediate Id.
  • a fourth aspect of the invention provides the process for the preparation of the compound of the first aspect, wherein when R 1 is a cyano group, the method comprises the steps of:
  • intermediate I-13 is subjected to intramolecular nucleophilic substitution to give intermediate I-14;
  • intermediate I-14 is subjected to nucleophilic substitution to give intermediate I-15;
  • the intermediate I-12 is hydrolyzed by hydrochloric acid to form an intermediate I-13.
  • intermediate I-13 is reacted with sodium bicarbonate by intramolecular nucleophilic substitution to form intermediate I-14.
  • intermediate I-14 is reacted with 2R,6R-dimethylmorpholine by nucleophilic substitution to give intermediate I-15.
  • intermediate I-15 is reacted with N,N'-carbonyldiimidazole by nucleophilic substitution to give intermediate I-16.
  • a fifth aspect of the invention provides the compound of the first aspect, or an enantiomer, diastereomer, racemate or mixture thereof, or a pharmaceutically acceptable salt thereof, or a second
  • the use of the pharmaceutical composition of the aspect for the preparation of a medicament for the treatment of a bacterial infectious disease.
  • the infectious disease is an infectious disease caused by Gram-positive bacteria.
  • the infectious disease is an infectious disease caused by a multidrug resistant bacteria.
  • the multidrug resistant bacteria are selected from the group consisting of MRSA, MSSA, MRSE, MSSE, PRSP, and Spy.
  • an in vitro bacteriostatic method for administering a compound of the above aspect or an enantiomer thereof, a diastereomer, a racemate and a mixture thereof to a subject or the environment. Or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of the second aspect.
  • a compound of the formula I the structure of which is represented by the formula Ia, Ib, Ic, Id or Ie:
  • R 1 , R 2 , R 3 and R 4 are as defined above.
  • the compound of the invention has better in vitro and in vivo antibacterial activity, excellent in vivo metabolic properties, exposure and peak concentration are far superior to the positive control drug AZD0914, and the compound of the invention has better medicine than the positive control drug AZD0914. Sex, is expected to become a better antibacterial drug.
  • the inventors of the present application have extensively and intensively studied to carry out various structural modifications to AZD0914, especially to introduce a spiro ring structure on an oxazolidinone ring, and synthesize a series of novel spiropyrimidine compounds, which are found in vitro.
  • drug metabolism properties are much better than AZD0914, it is suitable as a new antibacterial drug for human or animal antibacterial treatment of infection.
  • the present invention has been completed.
  • C 1 -C 3 alkyl means a straight or branched alkyl group having 1 to 3 carbon atoms, and includes, without limitation, methyl, ethyl, propyl, isopropyl. .
  • the term "3-7 membered aliphatic ring” means a cyclic alkyl group having 3 to 7 carbon atoms in the ring, and includes, without limitation, a cyclopropane ring, a cyclobutane ring, a cyclopentane ring. , a cyclohexane ring, a cycloheptane ring, and the like.
  • the term "3-7 membered oxygen-containing heterocyclic ring” means a cycloalkyl ring having 3 to 7 ring atoms and containing 1, 2 or 3 O atoms, and includes, without limitation, a propylene oxide ring. , butylene oxide ring, epoxy heptane ring and the like.
  • the "pharmaceutically acceptable salt” includes pharmaceutically acceptable base addition salts. These include, but are not limited to, salts of inorganic bases such as sodium, potassium, calcium and magnesium salts. These include, but are not limited to, salts of organic bases such as ammonium salts, triethylamine salts, lysine salts, arginine salts and the like. These salts can be prepared by methods known in the art.
  • the compound of the present invention (I) can be produced by the following method, however, the conditions of the method, such as the reactant, the solvent, the base, the amount of the compound used, the reaction temperature, the time required for the reaction, and the like are not limited to the following explanations.
  • the compounds of the present invention may also be conveniently prepared by combining various synthetic methods described in the specification or known in the art, and such combinations are readily made by those skilled in the art to which the present invention pertains.
  • R 2 , R 3 and R 4 are as defined above.
  • intermediate A [J. Med. Chem, 2015, 58 (15): 6264-6282.] and N-chlorosuccinimide in a polar aprotic solvent, from room temperature to 50 ° C conditions
  • the lower reaction is carried out for 20-60 minutes (minutes) to obtain intermediate I-1.
  • the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide,
  • the optimum reaction temperature was 40 ° C and the optimum reaction time was 30 min.
  • intermediate I-1 in the presence of different primary amines in a polar aprotic solvent, 0 ° C to room temperature reaction for 20-60min, nucleophilic substitution to form the corresponding intermediate I-2
  • the primary amine is a variety of suitable primary amines
  • the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, optimum reaction temperature The optimum reaction time was 30 min at 0 °C.
  • Intermediate I-2 is reacted in a polar aprotic solvent in a polar aprotic solvent at room temperature to 70 ° C for 2-5 h (hours) in the presence of cesium carbonate to form the corresponding intermediate I-3.
  • the aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 60 ° C, and the optimum reaction time is 3 h.
  • intermediate I-3 and N,N'-carbonyldiimidazole are catalyzed by 4-dimethylaminopyridine in a polar aprotic solvent at 60-90 ° C for 3-8 h to form a corresponding Intermediate I-4
  • the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 80 ° C, the best The reaction time was 5 h.
  • the solvent may be 1,4-dioxane, tetrahydrofuran, N, N-dimethylformamide, the optimum reaction temperature is 30 ° C, and the optimum reaction time is 3 h.
  • intermediate I-5 and 2R, 6R-dimethylmorpholine in the presence of an organic base, in a polar aprotic solvent, reacted at 70-100 ° C for 12-20h to form the corresponding intermediate I -6
  • the organic base may be triethylamine or N,N-diisopropylethylamine
  • the polar aprotic solvent may be acetonitrile, 1,4-dioxane, tetrahydrofuran, N, N. - dimethylformamide
  • the optimum reaction temperature is 80 ° C
  • the optimum reaction time is 15 h.
  • the polar aprotic solvent may be tetrahydrofuran, dichloromethane, the optimum reaction temperature is -80 ° C, and the optimum reaction time is 5 min.
  • Intermediate I-7 and ethylene glycol are reacted in a toluene solvent at 100-130 ° C for 3-8 h under catalytic conditions of p-toluenesulfonic acid to form intermediate I-8.
  • the optimum reaction temperature is 120 ° C.
  • the optimal reaction time is 4h.
  • Intermediate I-8 lithium diisopropylamide and N,N-dimethylformamide are reacted in a polar aprotic solvent at -80 to -65 ° C for 20-60 min to form intermediate I.
  • the polar aprotic solvent may be tetrahydrofuran, dichloromethane, the optimum reaction temperature is -80 ° C, and the optimum reaction time is 30 min.
  • intermediate I-9 and hydroxylamine hydrochloride in an organic base, in a polar solvent, at 0 ° C to room temperature for 1-3h to form intermediate I-10
  • the organic base can be pyridine, triethyl Amine or N,N-diisopropylethylamine
  • the polar solvent may be methanol, dichloromethane, 1,4-dioxane, tetrahydrofuran, N,N-dimethylformamide, preferably
  • the reaction temperature was 0 ° C and the optimum reaction time was 2 h.
  • intermediate I-10 intermediate I-10 and N-chlorosuccinimide, reacted in a polar aprotic solvent at room temperature to 50 ° C for 20-60 min to obtain intermediate I-11, the polarity
  • the aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 40 ° C, and the optimum reaction time is 30 min.
  • intermediate I-11 and various qualified primary amines in a polar aprotic solvent, reacted at 0 ° C to room temperature for 10-60 min, nucleophilic substitution to form intermediate I-12, the polarity is not
  • the protic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 0 ° C, and the optimum reaction time is 30 min.
  • intermediate I-12 and hydrochloric acid reacted in a solvent at room temperature to 50 ° C for 2-5h to form intermediate I-13
  • the solvent may be: 1,4-dioxane, tetrahydrofuran, N, N-dimethylformamide
  • the optimum reaction temperature is 40 ° C
  • the optimum reaction time is 3 h.
  • intermediate I-13 and aqueous sodium hydrogencarbonate, reacting in a solvent at 0 ° C to 40 ° C for 1-4 h to form intermediate I-14
  • the solvent may be: 1,4-dioxane, Tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 25 ° C, and the optimum reaction time is 2 h.
  • intermediate I-14 and 2R, 6R-dimethylmorpholine in the presence of an organic base, in a polar aprotic solvent, reacted at 70-100 ° C for 12-20h to form intermediate I-15
  • the organic base may be triethylamine or N,N-diisopropylethylamine
  • the polar aprotic solvent may be acetonitrile, 1,4-dioxane, tetrahydrofuran, N,N-dimethyl
  • the optimum reaction temperature is 80 ° C and the optimum reaction time is 12 h.
  • intermediate I-15 and N, N'-carbonyldiimidazole catalyzed by 4-dimethylaminopyridine, reacted in a polar aprotic solvent at 60-90 ° C for 3-8 h, corresponding to the corresponding
  • the intermediate I-16, the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N, N-dimethylformamide, the optimum reaction temperature is 80 ° C, the most The reaction time is 5h.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an active ingredient in a safe and effective amount, together with a pharmaceutically acceptable carrier.
  • active ingredient refers to a compound of formula I as described herein.
  • the "active ingredient" and pharmaceutical composition of the present invention are useful for the preparation of a medicament for treating an infectious disease.
  • a medicament for the preparation of an infectious disease caused by a multidrug-resistant bacterium is prepared and/or treated.
  • the pharmaceutical compositions contain from 1 to 2000 mg of active ingredient per dose, more preferably from 10 to 200 mg of active ingredient per dose.
  • the "one dose” is a tablet.
  • “Pharmaceutically acceptable carrier” means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity.
  • “compatibility” it is meant herein that the components of the composition are capable of intermingling with the active ingredients of the present invention and with respect to each other without significantly reducing the efficacy of the active ingredients.
  • pharmaceutically acceptable carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid).
  • magnesium stearate magnesium stearate
  • calcium sulfate vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyol (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifier Wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
  • the administration form of the active ingredient or the pharmaceutical composition of the present invention is not particularly limited, and representative administration methods include
  • oral but not limited to: oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs.
  • the compounds of the invention may be administered alone or in combination with other therapeutic agents such as antibacterials.
  • a safe and effective amount of a compound of the invention is administered to a mammal (e.g., a human) in need of treatment wherein the dosage is a pharmaceutically effective effective dosage, for a 60 kg body weight
  • the dose to be administered is usually from 1 to 2000 mg, preferably from 20 to 500 mg.
  • specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • (2R, 4S, 4aS)-11-fluoro-2 was synthesized according to the synthesis method of compound 1, using intermediate I-6-2 (93 mg, 0.229 mmol) and barbituric acid (33 mg, 0.252 mmol) as raw materials.
  • (2R, 4S, 4aS)-11-fluoro-2 was synthesized according to the synthesis method of compound 1, using intermediate I-6-3 (92 mg, 0.228 mmol) and barbituric acid (32 mg, 0.251 mmol) as raw materials.
  • (2R, 4S, 4aS)-11-fluoro-2 was synthesized according to the synthesis method of compound 1, using intermediate I-6-4 (99 mg, 0.237 mmol) and barbituric acid (34 mg, 0.261 mmol) as raw materials.
  • intermediate I-4-1 The synthesis of intermediate I-4-1 was carried out using intermediate I-15-7 (120 mg, 0.335 mmol), carbonyldiimidazole (109 mg, 0.670 mmol) and 4-dimethylaminopyridine (41 mg, 0.335 mmol).
  • strains were clinical isolates collected in Sichuan and Beijing in December 2015.
  • the collection unit was identified by VITEK-60 automatic microbiological identification instrument and then re-identified by conventional methods.
  • Each strain of bacteria was divided into single colonies by agar plates before the experiment, and the freshly cultured cells at 37 ° C overnight were appropriately diluted for the experiment.
  • test bacterial liquid was adjusted to a bacterial suspension equivalent to 0.5 McMulster standard with physiological saline, diluted with MH broth 1:100, and added to the chemical solution to make the final concentration of the bacterial liquid about 10 4 CFU/ml; Then, 100 ⁇ l of the bacterial solution was separately added to the above wells (the total volume in each well was 200 ⁇ l), sealed, and placed in a 35-37 ° C incubator for 18-20 hours to judge the result. The OD 600 value was measured by a microplate reader to minimize the minimum drug concentration of the bacteria in the wells to be the minimum inhibitory concentration (MIC).
  • MIC minimum inhibitory concentration
  • Table 1 The results of in vitro antibacterial activity are shown in Table 1. It can be seen from Table 1 that the compound of the present invention has good in vitro antibacterial activity, and the in vitro antibacterial activity of Compound 1 on MRSA, MSSA, MRSE, MSSE, PRSP and Spy is much better than that of the control drug AZD0914, which is about 8 times that of AZD0914. The in vitro antibacterial activity of Compounds 2, 3 and 4 was comparable to that of the control drug.
  • Preparation of artificial gastric juice (without pepsin): Take 20 mg of sodium chloride, add 70 ⁇ L of concentrated hydrochloric acid and sufficient water to make up to 10 mL.
  • the pH of the artificial gastric juice is about 1.2.
  • Preparation of artificial intestinal juice (without trypsin): Take 68mg of potassium dihydrogen phosphate dissolved in 2500 ⁇ L of water, add 770 ⁇ L of 0.2N sodium hydroxide solution and 5mL of water, adjust the pH to 6.8 with 0.2N sodium hydroxide or 0.2N hydrochloric acid. ⁇ 0.1, add water to a volume of 10mL.
  • Standard curve samples (12.5, 25, 50, 100, 200, 400, 2000, 10000 nM) were prepared with acetonitrile/water (1/1, v/v) and the peak area of the analyte was quantified by Waters MassLynx (version V4.1) software.
  • a is the slope and b is the intercept.
  • the excellent metabolic properties are the key indicators of the drug-forming properties of the compounds.
  • the pharmacokinetic experiments prove that the compounds of the present invention have ideal metabolic characteristics, and the exposure and peak concentrations are far superior to the positive control drug AZD0914.
  • Compound 1 Based on the pharmacokinetic parameters of various aspects of Compound 1, it can be seen that Compound 1 has excellent metabolic properties in rats and is worthy of further research and development.
  • mice Take healthy Kunming mice, weighing 18-22 grams, randomly divided into groups, 5-10 mice in each group, male and female, absorb the above different dilutions of bacterial solution, intraperitoneal injection into mice, re-injection every 20g mouse 0.5ml, observed for 7-14 days after infection, and recorded the number of mouse deaths, the minimum amount of bacteria causing 100% death of mice as the minimum lethal bacteria amount (MLD), using the amount of bacteria as the amount of infectious bacteria in the in vivo protection test .
  • MLD minimum lethal bacteria amount
  • Liquid preparation test with medicinal 0.5% sodium carboxymethyl cellulose and dilute to the desired concentration of solution
  • Dosing capacity 0.5ml / 20g BW (body weight, body weight)
  • Group design AZD0914 (20, 10, 5, 2.5g ⁇ kg -1 ) group, compound 1 (20, 10, 5, 2.5g ⁇ kg -1 ) group, infection control group, blank control group, ABT toxicity control .
  • mice were stopped from feeding water for 18 hours before the test, and were randomly divided into groups according to their body weight. Each group of 8 mice, male and female, were injected intraperitoneally with the infection test bacterial solution. Each 20g mouse was injected with 0.5ml, 0.5h after infection, 4h. The designed dose was intragastrically administered, and 0.5 ml was perfused into the stomach every 20 g of mice; the number of deaths of the mice was observed and recorded, and the number of deaths was observed for 7-14 days. According to the number of deaths of mice, Sun Ruiyuan and other editors DAS1.0 software were used to calculate half of the Bliss method. Effective dose ED 50 and 95% confidence limit.
  • control drug AZD0914 group received oral cytochrome P450 inhibitor 1-aminobenzotriazole (ABT) 50 mg/kg two hours before infection, and 50 mg/kg ABT once orally after 12 hours. Other compounds did not require oral ABT.
  • ABT cytochrome P450 inhibitor 1-aminobenzotriazole
  • Infected control group only the infected bacteria were not administered, and the same volume of physiological saline was administered after the infection of the bacteria; the blank control group: no infection of the bacteria, intraperitoneal injection of an equal volume of physiological saline, and an equal volume of physiological saline.
  • the positive control drug AZD0914 needs to be combined with the cytochrome P450 inhibitor 1-aminobenzotriazole (ABT), and the ED 50 for S. aureus MRSA15-3 infected mice is 11.51 mg/kg, while Compound 1 does not need to be combined.
  • ABT, ED 50 for oral administration to S. aureus MRSA15-3 infected mice was 3.87 mg/kg, which was significantly better than AZD0914.
  • the compound of the present invention has better drug-forming properties than the positive control drug AZD0914, and is expected to be a better antibacterial drug.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed in the present invention are a benzoisoxazole spiropyrimidinetrione compound, a preparation method therefor and use thereof, said compound having a structure as represented by formula (I). In said formula, the definition of each substituent is as described in the description and claims. The benzoisoxazole spiropyrimidinetrione compound of the present invention has higher antibacterial activities and better metabolic properties in vitro and in vivo.

Description

苯并异噁唑螺嘧啶三酮类化合物及制备方法和用途Benzoisoxazole spiropyrimidine compound, preparation method and use thereof 技术领域Technical field
本发明涉及一种含苯并异噁唑基团的螺嘧啶三酮类化合物、其药物组合物及其制备方法和在抗感染药物中的用途。The present invention relates to a benzopyrazole group-containing spiropyrimidine compound, a pharmaceutical composition thereof, a process for the preparation thereof and use thereof in an anti-infective drug.
背景技术Background technique
随着抗菌药物的广泛使用,甚至是滥用,致病菌的多重耐药性已成为一个威胁人类健康的严重问题,以多药耐药的金黄色葡萄球菌MRSA为代表的一系列耐药病原体,给临床治疗带来巨大挑战。近年来虽然有一些新型抗菌药物如利奈唑胺、达托霉素等上市,能有效的控制MRSA等革兰氏阳性菌的感染,但随着10余年的临床应用,细菌对这些药物也开始慢慢产生耐药菌株。因此,研发作用机制新颖、结构独特、对耐药菌有效的全新型抗菌药,是急迫的科研任务,也是未满足的重大临床需求。With the widespread use and even abuse of antibacterial drugs, the multi-drug resistance of pathogenic bacteria has become a serious problem threatening human health. A series of drug-resistant pathogens represented by multidrug-resistant Staphylococcus aureus MRSA, Bringing great challenges to clinical treatment. In recent years, although some new antibacterial drugs such as linezolid and daptomycin have been marketed, it can effectively control the infection of Gram-positive bacteria such as MRSA. However, with more than 10 years of clinical application, bacteria have begun to slow down these drugs. Slowly producing resistant strains. Therefore, a new type of antibacterial drug with novel mechanism of research and development, unique structure and effective against drug-resistant bacteria is an urgent scientific research task and an unmet major clinical requirement.
苯并异噁唑螺嘧啶三酮类化合物是一类新型的细菌II型拓扑异构酶抑制剂,虽然其与喹诺酮类药物都是作用于细菌II型拓扑异构酶,但其具体作用区域和作用机制完全不同。苯并异噁唑螺嘧啶三酮类化合物的新颖化学结构和全新作用机制,使其对敏感和耐药的革兰氏阳性菌都具有良好抗菌活性,且与临床已有抗菌药物不存在交叉耐药性。Benzoisoxazole spirotriones are a new class of bacterial type II topoisomerase inhibitors. Although they interact with quinolones for bacterial type II topoisomerases, their specific regions of action and The mechanism of action is completely different. The novel chemical structure and new mechanism of action of benzoisoxazole spiropyrimidines have good antibacterial activity against sensitive and resistant Gram-positive bacteria, and there is no cross-resistance with clinical antibacterial drugs. Medicinal.
阿斯利康等公司报道了一系列螺嘧啶三酮类化合物,并对其构效关系做了初步研究后发现,异噁唑上甲基取代的螺嘧啶三酮类化合物具有良好的抗菌活性。阿斯利康继续对此类化合物进行衍生化研究,进而发现了活性更好的AZD0914,同时消除了此类化合物的基因毒性和骨髓毒性,目前,AZD0914正在进行II期临床研究。AstraZeneca and other companies reported a series of spirotriazol compounds, and a preliminary study on its structure-activity relationship found that the methyl-substituted spiropyrimidine compounds on isoxazole have good antibacterial activity. AstraZeneca continued to conduct derivatization studies on such compounds, and found AZD0914 with better activity, while eliminating the genotoxicity and bone marrow toxicity of such compounds. Currently, AZD0914 is undergoing Phase II clinical studies.
但是,这些螺嘧啶三酮化合物如AZD0914还是存在抗菌活性不强、代谢性质不理想导致临床用药过量大的问题,在一定程度上限制了其作为治疗系统性感染药物的可能性。However, these spirotrione compounds, such as AZD0914, still have the problem that the antibacterial activity is not strong and the metabolic properties are not ideal, resulting in a large amount of clinical drug overdose, which limits the possibility of treating the systemic infection drug to some extent.
因此本领域尚需开发活性更好、代谢性质更优的用于人或动物系统性感染的螺嘧啶三酮类化合物克服其上述问题。Therefore, there is still a need in the art to develop a spiropyrimidine compound for use in human or animal systemic infection with better activity and superior metabolic properties to overcome the above problems.
发明内容Summary of the invention
本发明的目的在于提供一种活性更好、代谢性质更优的苯并异恶唑螺嘧啶三酮类化合物,其对映异构体、非对映异构体、外消旋体及其混合物,以及其药学上可接受的盐。The object of the present invention is to provide a benzoisoxazole spiropyrimidine compound having better activity and superior metabolic properties, the enantiomers, diastereomers, racemates and mixtures thereof. And its pharmaceutically acceptable salts.
本发明的第一方面,提供一种通式(I)所示的化合物,或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐,In a first aspect of the invention, there is provided a compound of the formula (I), or an enantiomer, diastereomer, racemate thereof and mixtures thereof, or a pharmaceutically acceptable thereof salt,
Figure PCTCN2018092208-appb-000001
Figure PCTCN2018092208-appb-000001
式中,R 1为氢、卤素或氰基; Wherein R 1 is hydrogen, halogen or cyano;
R 2、R 3独立为氢原子或C 1-C 3烷基;或者R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环; R 2 and R 3 are independently a hydrogen atom or a C 1 -C 3 alkyl group; or R 2 and R 3 together with a carbon atom to be bonded form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring;
R 4为氢原子或C 1-C 3烷基; R 4 is a hydrogen atom or a C 1 -C 3 alkyl group;
各*独立表示消旋、S型或R型;Each * indicates the racemization, S type or R type independently;
条件是当R 1为卤素时,R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环。 Provided that when R 1 is a halogen, R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
本发明的通式(I)表示化合物中至少含有3个手性中心,其存在对映异构体和非对映异构体。对于对映异构体,采用一般的手性拆分方法或不对称合成方法可以得到两个对映体。对于非对映异构体,可以通过分步重结晶或者色谱分离等方法分离。本发明的通式(I)表示的化合物包括上述异构体中的任何一种或者它们的混合物。The general formula (I) of the present invention means that the compound contains at least three chiral centers in the presence of enantiomers and diastereomers. For enantiomers, two enantiomers can be obtained using a general chiral resolution method or an asymmetric synthesis method. For the diastereomers, separation can be carried out by fractional recrystallization or chromatography. The compound represented by the formula (I) of the present invention includes any one of the above isomers or a mixture thereof.
在另一优选例中,R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环。 In another preferred embodiment, R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
在另一优选例中,R 2、R 3同时为氢或不同时为氢。 In another preferred embodiment, R 2 and R 3 are simultaneously hydrogen or hydrogen at the same time.
在另一优选例中,R 1为氢、氟、氯或氰基。 In another preferred embodiment, R 1 is hydrogen, fluorine, chlorine or cyano.
在另一优选例中,R 1为氢、氟或氯;R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环。 In another preferred embodiment, R 1 is hydrogen, fluorine or chlorine; and R 2 and R 3 together with the attached carbon atoms form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
在另一优选例中,R 1为氢、氟或氯;R 4为氢。 In another preferred embodiment, R 1 is hydrogen, fluorine or chlorine; and R 4 is hydrogen.
在另一优选例中,R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环;R 4为氢。 In another preferred embodiment, R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring; and R 4 is hydrogen.
在另一优选例中,R 1为氰基; In another preferred embodiment, R 1 is a cyano group;
R 2、R 3独立为氢原子或C 1-C 3烷基;或者R 2、R 3与相连的碳原子共同形成3-7元脂肪环。 R 2 and R 3 are independently a hydrogen atom or a C 1 -C 3 alkyl group; or R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring.
在另一优选例中,所述化合物为:In another preferred embodiment, the compound is:
Figure PCTCN2018092208-appb-000002
Figure PCTCN2018092208-appb-000002
本发明的第二方面,提供一种药物组合物,所述药物组合物包含第一方面所述的化合物或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐;以及According to a second aspect of the present invention, there is provided a pharmaceutical composition comprising the compound of the first aspect or an enantiomer thereof, a diastereomer, a racemate, and a mixture thereof, Or a pharmaceutically acceptable salt thereof;
药学上可接受的载体或赋形剂。A pharmaceutically acceptable carrier or excipient.
本发明提供新型的苯并异恶唑螺嘧啶三酮类化合物,可以单独使用,或者将其与可药用的辅料(例如赋形剂、稀释剂等)混合,配制成口服给药的片剂、胶囊剂、颗粒剂或糖浆剂等。该药物组合物可以按照制药学上常规方法制得。The present invention provides a novel benzisoxazole spiropyrimidine compound which can be used alone or mixed with a pharmaceutically acceptable adjuvant (for example, an excipient, a diluent, etc.) to prepare a tablet for oral administration. , capsules, granules or syrups, etc. The pharmaceutical composition can be obtained by a conventional method of pharmacy.
本发明的第三方面,提供第一方面所述的化合物的制备方法,所述方法包括以下步骤:A third aspect of the invention provides a process for the preparation of the compound of the first aspect, the method comprising the steps of:
Figure PCTCN2018092208-appb-000003
Figure PCTCN2018092208-appb-000003
(i)中间体Ia经分子内亲核取代反应生成中间体Ib;(i) intermediate Ia by intramolecular nucleophilic substitution reaction to form intermediate Ib;
(ii)中间体Ib经亲核取代反应生成中间体Ic;(ii) intermediate Ib is nucleophilic substituted to form intermediate Ic;
(iii)中间体Ic经催化水解反应生成中间体Id;(iii) intermediate Ic is catalytically hydrolyzed to form intermediate Id;
(iv)中间体Id经亲核取代反应生成中间体Ie;(iv) intermediate Id by nucleophilic substitution reaction to form intermediate Ie;
(v)中间体Ie与巴比妥酸反应生成通式I所示的化合物,(v) reacting intermediate Ie with barbituric acid to form a compound of formula I,
各式中,*、R 1、R 2、R 3和R 4的定义如前所述。 In the formulas, *, R 1 , R 2 , R 3 and R 4 are as defined above.
在另一优选例中,中间体Ia与碳酸铯经分子内亲核取代反应生成中间体Ib。In another preferred embodiment, intermediate Ia is reacted with cesium carbonate by intramolecular nucleophilic substitution to form intermediate Ib.
在另一优选例中,中间体Ib与N,N'-羰基二咪唑经亲核取代反应生成中间体Ic。In another preferred embodiment, the intermediate Ib is reacted with N,N'-carbonyldiimidazole by nucleophilic substitution to form the intermediate Ic.
在另一优选例中,中间体Id与2R,6R-二甲基吗啉经亲核取代反应生成中间体Ie。In another preferred embodiment, the intermediate Id is reacted with 2R,6R-dimethylmorpholine by nucleophilic substitution to form the intermediate Ie.
在另一优选例中,中间体Ic经盐酸催化水解反应生成中间体IdIn another preferred embodiment, the intermediate Ic is hydrolyzed by hydrochloric acid to form an intermediate Id.
本发明的第四方面,提供第一方面所述的化合物的制备方法,R 1为氰基时,所述方法包括以下步骤: A fourth aspect of the invention provides the process for the preparation of the compound of the first aspect, wherein when R 1 is a cyano group, the method comprises the steps of:
Figure PCTCN2018092208-appb-000004
Figure PCTCN2018092208-appb-000004
(i’)中间体I-12经水解反应生成中间体I-13;(i') intermediate I-12 is hydrolyzed to form intermediate I-13;
(ii’)中间体I-13经分子内亲核取代反应生成中间体I-14;(ii') intermediate I-13 is subjected to intramolecular nucleophilic substitution to give intermediate I-14;
(iii’)中间体I-14经亲核取代反应生成中间体I-15;(iii') intermediate I-14 is subjected to nucleophilic substitution to give intermediate I-15;
(iv’)中间体I-15经亲核取代反应生成中间体I-16;(iv') intermediate I-15 is subjected to nucleophilic substitution to give intermediate I-16;
(v’)中间体I-16与巴比妥酸反应生成式C所示的权利要求1所述的化合物,(v') intermediate I-16 is reacted with barbituric acid to form the compound of claim 1 represented by formula C,
各式中,*、R 2、R 3和R 4的定义如前所述。 In the formulas, *, R 2 , R 3 and R 4 are as defined above.
在另一优选例中,中间体I-12经盐酸催化发生水解反应生成中间体I-13In another preferred embodiment, the intermediate I-12 is hydrolyzed by hydrochloric acid to form an intermediate I-13.
在另一优选例中,中间体I-13与碳酸氢钠经分子内亲核取代反应生成中间体I-14。In another preferred embodiment, intermediate I-13 is reacted with sodium bicarbonate by intramolecular nucleophilic substitution to form intermediate I-14.
在另一优选例中,中间体I-14与2R,6R-二甲基吗啉经亲核取代反应生成中间体I-15。In another preferred embodiment, intermediate I-14 is reacted with 2R,6R-dimethylmorpholine by nucleophilic substitution to give intermediate I-15.
在另一优选例中,中间体I-15与N,N'-羰基二咪唑经亲核取代反应生成中间体I-16。In another preferred embodiment, intermediate I-15 is reacted with N,N'-carbonyldiimidazole by nucleophilic substitution to give intermediate I-16.
本发明的第五方面,提供第一方面所述的化合物或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐,或第二方面所述的药物组合物的用途,用于制备治疗细菌感染性疾病的药物。A fifth aspect of the invention provides the compound of the first aspect, or an enantiomer, diastereomer, racemate or mixture thereof, or a pharmaceutically acceptable salt thereof, or a second The use of the pharmaceutical composition of the aspect for the preparation of a medicament for the treatment of a bacterial infectious disease.
在另一优选例中,所述感染性疾病为革兰氏阳性菌引起的感染性疾病。In another preferred embodiment, the infectious disease is an infectious disease caused by Gram-positive bacteria.
在另一优选例中,所述感染性疾病为多药耐药菌引起的感染性疾病。In another preferred embodiment, the infectious disease is an infectious disease caused by a multidrug resistant bacteria.
在另一优选例中,所述多药耐药菌选自下组:MRSA、MSSA、MRSE、MSSE、 PRSP和Spy。In another preferred embodiment, the multidrug resistant bacteria are selected from the group consisting of MRSA, MSSA, MRSE, MSSE, PRSP, and Spy.
本发明的第六方面,提供一种体外抑菌方法,向对象或环境中施用第一方面所述的化合物或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐,或第二方面所述的药物组合物。According to a sixth aspect of the present invention, there is provided an in vitro bacteriostatic method for administering a compound of the above aspect or an enantiomer thereof, a diastereomer, a racemate and a mixture thereof to a subject or the environment. Or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of the second aspect.
本发明的第七方面,提供一种式I化合物中间体,其结构如式Ia、Ib、Ic、Id或Ie所示:According to a seventh aspect of the invention, there is provided a compound of the formula I, the structure of which is represented by the formula Ia, Ib, Ic, Id or Ie:
Figure PCTCN2018092208-appb-000005
Figure PCTCN2018092208-appb-000005
各式中,*、R 1、R 2、R 3和R 4的定义如前所述。 In the formulas, *, R 1 , R 2 , R 3 and R 4 are as defined above.
本发明的化合物,具有较佳的体外、体内抗菌活性,优良的体内代谢性质,暴露量和达峰浓度远远优于阳性对照药物AZD0914,且本发明化合物比阳性对照药AZD0914具有更好的成药性,有望成为更好的抗菌药物。The compound of the invention has better in vitro and in vivo antibacterial activity, excellent in vivo metabolic properties, exposure and peak concentration are far superior to the positive control drug AZD0914, and the compound of the invention has better medicine than the positive control drug AZD0914. Sex, is expected to become a better antibacterial drug.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。说明书中所揭示的各个特征,可以被任何提供相同、均等或相似目的的替代性特征取代。限于篇幅,在此不再一一累述。It is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to form a new or preferred technical solution. Each feature disclosed in the specification can be replaced by any alternative feature that provides the same, equal or similar purpose. Due to space limitations, we will not repeat them here.
具体实施方式Detailed ways
本申请的发明人经过广泛而深入地研究,对AZD0914进行各种结构修饰,特别是噁唑烷酮环上引入螺环结构,合成了一系列结构新颖的螺嘧啶三酮类化合物,发现其体外、体内抗菌活性、药物代谢性质大大优于AZD0914,很适合作为新型抗菌药物用于人或动物的抗菌治疗感染。在此基础上,完成了本发明。The inventors of the present application have extensively and intensively studied to carry out various structural modifications to AZD0914, especially to introduce a spiro ring structure on an oxazolidinone ring, and synthesize a series of novel spiropyrimidine compounds, which are found in vitro. In vivo antibacterial activity, drug metabolism properties are much better than AZD0914, it is suitable as a new antibacterial drug for human or animal antibacterial treatment of infection. On the basis of this, the present invention has been completed.
术语the term
在本发明中,术语“C 1-C 3烷基”是指具有1至3个碳原子的直链或支链烷基,非限制性地包括甲基、乙基、丙基、异丙基。 In the present invention, the term "C 1 -C 3 alkyl" means a straight or branched alkyl group having 1 to 3 carbon atoms, and includes, without limitation, methyl, ethyl, propyl, isopropyl. .
在本发明中,术语“3-7元脂肪环”是指在环上具有3至7个碳原子的环状烷基,非限制性地包括环丙烷环、环丁烷环、环戊烷环、环己烷环、环庚烷环等。In the present invention, the term "3-7 membered aliphatic ring" means a cyclic alkyl group having 3 to 7 carbon atoms in the ring, and includes, without limitation, a cyclopropane ring, a cyclobutane ring, a cyclopentane ring. , a cyclohexane ring, a cycloheptane ring, and the like.
在本发明中,术语“3-7元含氧杂环”是指具有3-7个环原子且包含1、2或3个O原子的环烷基环,非限制性地包括环氧丙烷环、环氧丁烷环、环氧庚烷环等。In the present invention, the term "3-7 membered oxygen-containing heterocyclic ring" means a cycloalkyl ring having 3 to 7 ring atoms and containing 1, 2 or 3 O atoms, and includes, without limitation, a propylene oxide ring. , butylene oxide ring, epoxy heptane ring and the like.
所述“药学上可接受的盐”包括药学可接受的碱加成盐。包括但不限于无机碱的盐如钠盐,钾盐,钙盐和镁盐等。包括但不限于有机碱的盐,比如铵盐,三乙胺盐,赖氨酸盐,精氨酸盐等。这些盐可通过本专业已知的方法制备。The "pharmaceutically acceptable salt" includes pharmaceutically acceptable base addition salts. These include, but are not limited to, salts of inorganic bases such as sodium, potassium, calcium and magnesium salts. These include, but are not limited to, salts of organic bases such as ammonium salts, triethylamine salts, lysine salts, arginine salts and the like. These salts can be prepared by methods known in the art.
制备方法Preparation
本发明(I)所示化合物可通过如下的方法制得,然而该方法的条件,例如反应物、溶剂、碱、所用化合物的量、反应温度、反应所需时间等不限于下面的解释。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便地制得,这样的组合可由本发明所属领域的技术人员容易的进行。The compound of the present invention (I) can be produced by the following method, however, the conditions of the method, such as the reactant, the solvent, the base, the amount of the compound used, the reaction temperature, the time required for the reaction, and the like are not limited to the following explanations. The compounds of the present invention may also be conveniently prepared by combining various synthetic methods described in the specification or known in the art, and such combinations are readily made by those skilled in the art to which the present invention pertains.
路线1Route 1
在优选实施方式中,以路线1制备化合物1、化合物2、化合物3和化合物4。In a preferred embodiment, Compound 1, Compound 2, Compound 3 and Compound 4 are prepared in Scheme 1.
Figure PCTCN2018092208-appb-000006
Figure PCTCN2018092208-appb-000006
R 2、R 3和R 4定义如前。 R 2 , R 3 and R 4 are as defined above.
a、将中间体A[J.Med.Chem,2015,58(15):6264-6282.]与N-氯代丁二酰亚胺在极性非质子性溶剂中,于室温至50℃条件下反应20-60min(分钟),得到中间体I-1,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为40℃,最佳反应时间为30min。a, intermediate A [J. Med. Chem, 2015, 58 (15): 6264-6282.] and N-chlorosuccinimide in a polar aprotic solvent, from room temperature to 50 ° C conditions The lower reaction is carried out for 20-60 minutes (minutes) to obtain intermediate I-1. The polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, The optimum reaction temperature was 40 ° C and the optimum reaction time was 30 min.
b、中间体I-1分别在不同的伯胺的存在下,在极性非质子性溶剂中,于0℃到室温反应20-60min,亲核取代生成相对应的中间体I-2,所述伯胺为各种符合要求的伯胺,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为0℃,最佳反应时间为30min。b, intermediate I-1 in the presence of different primary amines in a polar aprotic solvent, 0 ° C to room temperature reaction for 20-60min, nucleophilic substitution to form the corresponding intermediate I-2, The primary amine is a variety of suitable primary amines, and the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, optimum reaction temperature The optimum reaction time was 30 min at 0 °C.
c、中间体I-2在碳酸铯的存在下,在极性非质子性溶剂中,于室温至70℃反应2-5h(小时),生成相对应的中间体I-3,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为60℃,最佳反应时间为3h。c. Intermediate I-2 is reacted in a polar aprotic solvent in a polar aprotic solvent at room temperature to 70 ° C for 2-5 h (hours) in the presence of cesium carbonate to form the corresponding intermediate I-3. The aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 60 ° C, and the optimum reaction time is 3 h.
d、中间体I-3与N,N'-羰基二咪唑在4-二甲氨基吡啶的催化下,在极性非质子性溶剂中,于60-90℃反应3-8h,生成相对应的中间体I-4,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为80℃, 最佳反应时间为5h。d, intermediate I-3 and N,N'-carbonyldiimidazole are catalyzed by 4-dimethylaminopyridine in a polar aprotic solvent at 60-90 ° C for 3-8 h to form a corresponding Intermediate I-4, the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 80 ° C, the best The reaction time was 5 h.
e、中间体I-4和盐酸,在溶剂中于室温至50℃下反应2-5h,生成相对应的中间体I-5,所述溶剂可以是1,4-二氧六环、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为30℃,最佳反应时间为3h。e, intermediate I-4 and hydrochloric acid, reacted in a solvent at room temperature to 50 ° C for 2-5 h to form the corresponding intermediate I-5, the solvent may be 1,4-dioxane, tetrahydrofuran, N, N-dimethylformamide, the optimum reaction temperature is 30 ° C, and the optimum reaction time is 3 h.
f、中间体I-5与2R,6R-二甲基吗啉,在有机碱存在下,于极性非质子性溶剂中,在70-100℃反应12-20h,生成相对应的中间体I-6,所述有机碱可以是三乙胺或N,N-二异丙基乙胺,所述的极性非质子溶剂可以为乙腈、1,4-二氧六环、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为80℃,最佳反应时间为15h。f, intermediate I-5 and 2R, 6R-dimethylmorpholine, in the presence of an organic base, in a polar aprotic solvent, reacted at 70-100 ° C for 12-20h to form the corresponding intermediate I -6, the organic base may be triethylamine or N,N-diisopropylethylamine, and the polar aprotic solvent may be acetonitrile, 1,4-dioxane, tetrahydrofuran, N, N. - dimethylformamide, the optimum reaction temperature is 80 ° C, and the optimum reaction time is 15 h.
g、中间体I-6与巴比妥酸,在醋酸和水的混合溶剂中,与100-120℃反应4-7h,生成相对应的化合物B,最佳反应温度为100℃,最佳反应时间为5h。g, intermediate I-6 and barbituric acid, in a mixed solvent of acetic acid and water, reacted with 100-120 ° C for 4-7h, the corresponding compound B is formed, the optimal reaction temperature is 100 ° C, the best reaction The time is 5h.
路线2Route 2
在优选实施方式中,以路线2制备化合物5、化合物6和化合物7。In a preferred embodiment, Compound 5, Compound 6 and Compound 7 are prepared in Scheme 2.
Figure PCTCN2018092208-appb-000007
Figure PCTCN2018092208-appb-000007
a、2,6-二氟苯腈与二异丙基氨基锂、N,N-二甲基甲酰胺,在极性非质子性溶剂中,于-80~-65℃反应3-8min,生成中间体I-7,所述极性非质子性溶剂可以是四氢呋喃、二氯甲烷,最佳反应温度为-80℃,最佳反应时间为5min。a, 2,6-difluorobenzonitrile and lithium diisopropylamide, N,N-dimethylformamide, reacted in a polar aprotic solvent at -80 ~ -65 ° C for 3-8min, resulting Intermediate I-7, the polar aprotic solvent may be tetrahydrofuran, dichloromethane, the optimum reaction temperature is -80 ° C, and the optimum reaction time is 5 min.
b、中间体I-7与乙二醇,在对甲苯磺酸催化条件下,于甲苯溶剂中,100-130℃反应3-8h,生成中间体I-8,最佳反应温度为120℃,最佳反应时间为4h。b. Intermediate I-7 and ethylene glycol are reacted in a toluene solvent at 100-130 ° C for 3-8 h under catalytic conditions of p-toluenesulfonic acid to form intermediate I-8. The optimum reaction temperature is 120 ° C. The optimal reaction time is 4h.
c、中间体I-8与二异丙基氨基锂、N,N-二甲基甲酰胺,在极性非质子性溶剂中,于-80~-65℃反应20-60min,生成中间体I-9,所述极性非质子性溶剂可以是四氢呋喃、二氯甲烷,最佳反应温度为-80℃,最佳反应时间为30min。c. Intermediate I-8, lithium diisopropylamide and N,N-dimethylformamide are reacted in a polar aprotic solvent at -80 to -65 ° C for 20-60 min to form intermediate I. -9, the polar aprotic solvent may be tetrahydrofuran, dichloromethane, the optimum reaction temperature is -80 ° C, and the optimum reaction time is 30 min.
d、中间体I-9与盐酸羟胺,在有机碱条件下,于极性溶剂中,于0℃至室温反应1-3h,生成中间体I-10,所述有机碱可以是吡啶、三乙胺或N,N-二异丙基乙胺,所述的极性溶剂可以为甲醇、二氯甲烷、1,4-二氧六环、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为0℃,最佳反应时间为2h。d, intermediate I-9 and hydroxylamine hydrochloride, in an organic base, in a polar solvent, at 0 ° C to room temperature for 1-3h to form intermediate I-10, the organic base can be pyridine, triethyl Amine or N,N-diisopropylethylamine, the polar solvent may be methanol, dichloromethane, 1,4-dioxane, tetrahydrofuran, N,N-dimethylformamide, preferably The reaction temperature was 0 ° C and the optimum reaction time was 2 h.
e、中间体I-10与N-氯代丁二酰亚胺,在极性非质子性溶剂中,于室温至50℃条件下反应20-60min,得到中间体I-11,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为40℃,最佳反应时间为30min。e, intermediate I-10 and N-chlorosuccinimide, reacted in a polar aprotic solvent at room temperature to 50 ° C for 20-60 min to obtain intermediate I-11, the polarity The aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 40 ° C, and the optimum reaction time is 30 min.
f、中间体I-11与各种符合条件的伯胺,在极性非质子性溶剂中,于0℃至室温反应10-60min,亲核取代生成中间体I-12,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为0℃,最佳反应时间为30min。f, intermediate I-11 and various qualified primary amines, in a polar aprotic solvent, reacted at 0 ° C to room temperature for 10-60 min, nucleophilic substitution to form intermediate I-12, the polarity is not The protic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 0 ° C, and the optimum reaction time is 30 min.
g、中间体I-12与盐酸,在溶剂中于室温至50℃下反应2-5h,生成中间体I-13,所述溶剂可以是:1,4-二氧六环、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为40℃,最佳反应时间为3h。g, intermediate I-12 and hydrochloric acid, reacted in a solvent at room temperature to 50 ° C for 2-5h to form intermediate I-13, the solvent may be: 1,4-dioxane, tetrahydrofuran, N, N-dimethylformamide, the optimum reaction temperature is 40 ° C, and the optimum reaction time is 3 h.
h、中间体I-13与碳酸氢钠水溶液,在溶剂中于0℃至40℃下反应1-4h,生成中间体I-14,所述溶剂可以是:1,4-二氧六环、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为25℃,最佳反应时间为2h。h, intermediate I-13 and aqueous sodium hydrogencarbonate, reacting in a solvent at 0 ° C to 40 ° C for 1-4 h to form intermediate I-14, the solvent may be: 1,4-dioxane, Tetrahydrofuran, N,N-dimethylformamide, the optimum reaction temperature is 25 ° C, and the optimum reaction time is 2 h.
i、中间体I-14与2R,6R-二甲基吗啉,在有机碱存在下,于极性非质子性溶剂中,在70-100℃反应12-20h,生成中间体I-15,所述有机碱可以是三乙胺或N,N-二异丙基乙胺,所述的极性非质子溶剂可以为乙腈、1,4-二氧六环、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为80℃,最佳反应时间为12h。i, intermediate I-14 and 2R, 6R-dimethylmorpholine, in the presence of an organic base, in a polar aprotic solvent, reacted at 70-100 ° C for 12-20h to form intermediate I-15, The organic base may be triethylamine or N,N-diisopropylethylamine, and the polar aprotic solvent may be acetonitrile, 1,4-dioxane, tetrahydrofuran, N,N-dimethyl The optimum reaction temperature is 80 ° C and the optimum reaction time is 12 h.
j、中间体I-15与N,N'-羰基二咪唑,在4-二甲氨基吡啶的催化下,在极性非质子性溶剂中,于60-90℃反应3-8h,生成相对应的中间体I-16,所述极性非质子性溶剂可以是:1,4-二氧六环、甲苯、四氢呋喃、N,N-二甲基甲酰胺,最佳反应温度为80℃,最佳反应时间为5h。j, intermediate I-15 and N, N'-carbonyldiimidazole, catalyzed by 4-dimethylaminopyridine, reacted in a polar aprotic solvent at 60-90 ° C for 3-8 h, corresponding to the corresponding The intermediate I-16, the polar aprotic solvent may be: 1,4-dioxane, toluene, tetrahydrofuran, N, N-dimethylformamide, the optimum reaction temperature is 80 ° C, the most The reaction time is 5h.
k、中间体I-16与巴比妥酸,在醋酸和水的混合溶剂中,于100-120℃反应4-7h,生成化合物C,最佳反应温度为110℃,最佳反应时间为5h。k, intermediate I-16 and barbituric acid, in a mixed solvent of acetic acid and water, reacted at 100-120 ° C for 4-7h to form compound C, the optimal reaction temperature is 110 ° C, the optimal reaction time is 5h .
药物组合物Pharmaceutical composition
本发明还提供了一种药物组合物,它包含安全有效量范围内的活性成分,以及药学上可接受的载体。The invention also provides a pharmaceutical composition comprising an active ingredient in a safe and effective amount, together with a pharmaceutically acceptable carrier.
本发明所述的“活性成分”是指本发明所述的式I化合物。As used herein, "active ingredient" refers to a compound of formula I as described herein.
本发明所述的“活性成分”和药物组合物用于制备治疗感染性疾病的药物。在另一优选例中,用于制备预防和/或治疗多药耐药菌引起的感染性疾病的药物。The "active ingredient" and pharmaceutical composition of the present invention are useful for the preparation of a medicament for treating an infectious disease. In another preferred embodiment, a medicament for the preparation of an infectious disease caused by a multidrug-resistant bacterium is prepared and/or treated.
“安全有效量”指的是:活性成分的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg活性成分/剂,更佳地,含有10-200mg活性成分/剂。较佳地,所述的“一剂”为一个药片。By "safe and effective amount" is meant that the amount of active ingredient is sufficient to significantly improve the condition without causing serious side effects. In general, the pharmaceutical compositions contain from 1 to 2000 mg of active ingredient per dose, more preferably from 10 to 200 mg of active ingredient per dose. Preferably, the "one dose" is a tablet.
“药学上可接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的活性成分以及它们之间相互掺和,而不明显降低活性成分的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂
Figure PCTCN2018092208-appb-000008
润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
"Pharmaceutically acceptable carrier" means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity. By "compatibility" it is meant herein that the components of the composition are capable of intermingling with the active ingredients of the present invention and with respect to each other without significantly reducing the efficacy of the active ingredients. Examples of pharmaceutically acceptable carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid). , magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyol (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifier
Figure PCTCN2018092208-appb-000008
Wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
本发明的活性成分或药物组合物的施用方式没有特别限制,代表性的施用方式包括The administration form of the active ingredient or the pharmaceutical composition of the present invention is not particularly limited, and representative administration methods include
(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)等。(but not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous).
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs.
本发明化合物可以单独给药,或者与其他治疗药物(如抗菌药)联合给药。The compounds of the invention may be administered alone or in combination with other therapeutic agents such as antibacterials.
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。When a pharmaceutical composition is used, a safe and effective amount of a compound of the invention is administered to a mammal (e.g., a human) in need of treatment wherein the dosage is a pharmaceutically effective effective dosage, for a 60 kg body weight, The dose to be administered is usually from 1 to 2000 mg, preferably from 20 to 500 mg. Of course, specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件(如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件)或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数是重量百分比和重量份数。The invention is further illustrated below in conjunction with specific embodiments. It is to be understood that the examples are not intended to limit the scope of the invention. The experimental methods in the following examples, which do not specify the specific conditions, are usually manufactured according to conventional conditions (such as those described in Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989)). The conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are by weight and parts by weight.
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。文中所述的较佳实施方法与材料仅作示范之用。Unless otherwise defined, all professional and scientific terms used herein have the same meaning as those skilled in the art. In addition, any methods and materials similar or equivalent to those described may be employed in the methods of the invention. The preferred embodiments and materials described herein are for illustrative purposes only.
以下所有实施例中, 1H-NMR用Varian Mercury 400核磁共振仪记录, 13C-NMR用BRUKER 500MHz核磁共振仪(低温)记录,化学位移以δ(ppm)表示;分离用硅胶,未说明均为200-300目,洗脱液的配比均为体积比。 In all of the following examples, 1 H-NMR was recorded on a Varian Mercury 400 nuclear magnetic resonance apparatus, 13 C-NMR was recorded on a BRUKER 500 MHz nuclear magnetic resonance apparatus (low temperature), and the chemical shift was expressed in δ (ppm); For the 200-300 mesh, the ratio of the eluents is a volume ratio.
实施例1:(2R,4S,4aS)-11-氟-2,4-二甲基-8-(5-氧代-4-氧-6-氮杂螺[2.4]庚烷-6-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物1)Example 1: (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(5-oxo-4-oxo-6-azaspiro[2.4]heptane-6-yl -1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5, 5'-pyrimidine]-2',4',6'(1'H,3'H)-trione (Compound 1)
(a)1-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)-1-环丙醇(I-3-1)(a) 1-(((5-(1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)amino)methyl)- 1-cyclopropanol (I-3-1)
Figure PCTCN2018092208-appb-000009
Figure PCTCN2018092208-appb-000009
将中间体A(1g,4.048mmol)加入到20ml N,N-二甲基甲酰胺中,搅拌使其溶解,室温下加入N-氯代丁二酰亚胺(650mg,4.868mmol),升至40℃反应30min,TLC(薄层色谱法)监测反应完全,将反应液将至0℃,缓慢加入1-氨甲基-1-环丙醇(1.06g,12.144mmol),恢复至室温反应1h,TLC监测反应完全,加入碳酸铯(4.567g,12.144mmol),升至60℃反应2h,TLC监测反应完全,降至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=1:1],得白色固体810mg,收率65%, 1H NMR(400MHz,CDCl 3)δ7.53(dd,J=5.4,1.5Hz,1H),6.13(s,1H),4.79(t,J=5.6Hz,1H),4.22–4.09(m,4H),3.58(d,J=5.5Hz,2H),2.65(s,1H),0.95(t,J=6.2Hz,2H),0.75(t,J=6.2Hz,2H)。 Intermediate A (1 g, 4.048 mmol) was added to 20 ml of N,N-dimethylformamide, and the mixture was stirred and dissolved. N-chlorosuccinimide (650 mg, 4.668 mmol) was added at room temperature. The reaction was carried out at 40 ° C for 30 min, and the reaction was monitored by TLC (thin-layer chromatography). The reaction mixture was taken to 0 ° C, and 1-aminomethyl-1-cyclopropanol (1.06 g, 12.144 mmol) was slowly added and returned to room temperature for 1 h. The reaction was monitored by TLC. EtOAc (EtOAc: EtOAc (EtOAc: EtOAc) dried over anhydrous sodium sulfate, and concentrated under reduced pressure and column chromatography [petroleum ether: ethyl acetate = 1: 1] to give a white solid 810 mg, yield 65%, 1 H NMR (400MHz , CDCl 3) δ7.53 (dd, J = 5.4, 1.5 Hz, 1H), 6.13 (s, 1H), 4.79 (t, J = 5.6 Hz, 1H), 4.22 - 4.09 (m, 4H), 3.58 (d, J = 5.5 Hz, 2H), 2.65 (s, 1H), 0.95 (t, J = 6.2 Hz, 2H), 0.75 (t, J = 6.2 Hz, 2H).
(b)6-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-4-氧-6-氮杂螺[2.4]庚烷-5-酮(I-4-1)(b) 6-(5-((1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)-4-oxo-6- Azaspiro[2.4]heptane-5-one (I-4-1)
Figure PCTCN2018092208-appb-000010
Figure PCTCN2018092208-appb-000010
将中间体I-3-1(344mg,1.036mmol)加入到15mlN,N-二甲基甲酰胺中,搅拌使其溶解,分别依次加入羰基二咪唑(671mg,4.414mmol)和4-二甲氨基吡啶(63mg,0.52mmol),加热到115℃反应8h,TLC监测反应完全,冷却至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=2:1],得白色固体337mg,收率96%, 1H NMR(400MHz,CDCl 3)δ8.55(dd,J=5.9,1.9Hz,1H),6.16(s,1H),4.31(s,2H),4.22–4.09(m,4H),1.44–1.39(m,2H),0.98–0.92(m,2H)。 Intermediate 1-3-1 (344 mg, 1.036 mmol) was added to 15 ml of N,N-dimethylformamide, and dissolved by stirring, and carbonyldiimidazole (671 mg, 4.414 mmol) and 4-dimethylamino group were sequentially added, respectively. The pyridine (63 mg, 0.52 mmol) was heated to 115 ° C for 8 h. The reaction was taken from EtOAc EtOAc. Column chromatography [petroleum ether: ethyl acetate = 2:1] afforded 337 mg of white solid (yield: 96%, 1 H NMR (400 MHz, CDCl 3 ) δ 8.55 (dd, J = 5.9, 1.9 Hz, 1H) , 6.16 (s, 1H), 4.31 (s, 2H), 4.22 - 4.09 (m, 4H), 1.44 - 1.39 (m, 2H), 0.98 - 0.92 (m, 2H).
(c)6,7-二氟-3-(5-氧代-4-氧-6-氮杂螺[2.4]庚烷-6-基)苯并[d]异噁唑-5-甲醛(I-5-1)(c) 6,7-Difluoro-3-(5-oxo-4-oxo-6-azaspiro[2.4]heptan-6-yl)benzo[d]isoxazole-5-carbaldehyde ( I-5-1)
Figure PCTCN2018092208-appb-000011
Figure PCTCN2018092208-appb-000011
将中间体I-4-1(330mg,0.976mmol)溶解于15ml1,4-二氧六环中,加6M盐酸(5ml),室温下搅拌2h,TLC监测反应完全,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:二氯甲烷:乙酸乙酯=5:5:1],得白色固体285mg,收率99%, 1H NMR(400MHz,CDCl 3)δ10.30(s,1H),9.00(dd,J=5.9,1.9Hz,1H),4.32(s,2H),1.44(m,2H),1.00–0.95(m,2H)。 The intermediate I-4-1 (330 mg, 0.976 mmol) was dissolved in 15 ml of 1,4-dioxane, and 6M hydrochloric acid (5 ml) was added, and the mixture was stirred at room temperature for 2 h. The organic layer was combined, washed with water, brine, dried over anhydrous sodium sulfate sulfatesssssssssssssssssss , 1 H NMR (400MHz, CDCl 3 ) δ 10.30 (s, 1H), 9.00 (dd, J = 5.9, 1.9 Hz, 1H), 4.32 (s, 2H), 1.44 (m, 2H), 1.00 - 0.95 (m, 2H).
(d)6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(5-氧代-4-氧-6-氮杂螺[2.4]庚烷-6-基)苯并[d]异噁唑-5-甲醛(I-6-1)(d) 6-((2R,6R)-2,6-dimethylmorpholine)-7-fluoro-3-(5-oxo-4-oxo-6-azaspiro[2.4]heptane- 6-yl)benzo[d]isoxazole-5-formaldehyde (I-6-1)
Figure PCTCN2018092208-appb-000012
Figure PCTCN2018092208-appb-000012
将中间体I-5-1(200mg,0..67mmol)溶于10ml乙腈中,搅拌使其溶解,分别依次加入2R,6R-二甲基吗啉(0.31ml,2.54mmol)和N,N-二异丙基乙胺(0.42ml,2.54mmol),将反应液于90℃条件下回流反应10h,TLC监测反应完全,冷却至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=5:1],得淡黄色固体80mg,收率31%, 1H NMR(400MHz,CDCl3)δ10.39(s,1H),8.83(s,1H),4.30(s,2H),4.29–4.22(m,2H),3.45(d,J=11.7Hz,2H),3.06(dd,J=11.7,5.4Hz,2H),1.45–1.40(m,2H),1.34(d,J=6.5Hz,6H),0.98–0.93(m,2H)。 Intermediate 1-5-1 (200 mg, 0..67 mmol) was dissolved in 10 ml of acetonitrile and stirred to dissolve. 2R,6R-dimethylmorpholine (0.31 ml, 2.54 mmol) and N, N were sequentially added. -diisopropylethylamine (0.42 ml, 2.54 mmol), the reaction mixture was refluxed for 10 h at 90 ° C, the reaction was completed by TLC, cooled to room temperature, extracted with water and ethyl acetate. washed with brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure and column chromatography [petroleum ether: ethyl acetate = 5: 1] to give a pale yellow solid 80mg, yield 31%, 1 H NMR (400MHz , CDCl3) δ10.39 (s, 1H), 8.83 (s, 1H), 4.30 (s, 2H), 4.29 - 4.22 (m, 2H), 3.45 (d, J = 11.7 Hz, 2H), 3.06 (dd, J = 11.7, 5.4 Hz, 2H), 1.45 - 1.40 (m, 2H), 1.34 (d, J = 6.5 Hz, 6H), 0.98 - 0.93 (m, 2H).
(e)(2R,4S,4aS)-11-氟-2,4-二甲基-8-(5-氧代-4-氧-6-氮杂螺[2.4]庚烷-6-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物1)(e) (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(5-oxo-4-oxo-6-azaspiro[2.4]heptane-6-yl) -1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5,5 '-Pyrimidine]-2',4',6'(1'H,3'H)-Trione (Compound 1)
Figure PCTCN2018092208-appb-000013
Figure PCTCN2018092208-appb-000013
将中间体I-6-1(80mg,0.206mmol)加入到4ml醋酸和1ml水的混合溶剂中,搅拌使其溶解,加入巴比妥酸(32mg,0.246mmol),升至100℃反应3h,TLC监测反应完全,冷却至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩反相柱制备[甲醇:水=50:50],得白色固体78mg,收率76%,m.p.266-267℃; 1H NMR(400MHz,DMSO)δ11.82(s,1H),11.46(s,1H),7.76(s,1H),4.29–4.21(m,2H),4.11(d,J=12.9Hz,1H),3.94(d,J=8.8Hz,1H),3.78(br s,1H),3.72–3.62(m,2H),3.17–3.06(m,1H),2.92(d,J=14.1Hz,1H),1.24-1.19(m,2H),1.15(d,J=6.3Hz,3H),1.00–0.95(m,2H),0.90(d,J=6.3Hz,3H). 13C NMR(126 MHz,DMSO)δ171.40,168.15,154.20(d,J=12.8Hz),153.79,153.17,149.98,135.30,133.76(d,J=238.8Hz),122.85,118.83,106.52,72.58,72.14,64.88,62.33,56.73(d,J=9.3Hz),53.38,50.71,39.02,18.64,18.61,10.32,10.26;MS(ESI)m/z:[(M+1) +,500.1]。 Intermediate I-6-1 (80 mg, 0.206 mmol) was added to a mixed solvent of 4 ml of acetic acid and 1 ml of water, stirred to dissolve, and barbituric acid (32 mg, 0.246 mmol) was added thereto, and the mixture was heated to 100 ° C for 3 h. The reaction was completed by TLC, and the mixture was cooled to room temperature. EtOAc (EtOAc) White solid 78 mg, yield 76%, mp 266-267 ° C; 1 H NMR (400 MHz, DMSO) δ 11.82 (s, 1H), 11.46 (s, 1H), 7.76 (s, 1H), 4.29 - 4.21. , 2H), 4.11 (d, J = 12.9 Hz, 1H), 3.94 (d, J = 8.8 Hz, 1H), 3.78 (br s, 1H), 3.72 - 3.62 (m, 2H), 3.17 - 3.06 (m , 1H), 2.92 (d, J = 14.1 Hz, 1H), 1.24-1.19 (m, 2H), 1.15 (d, J = 6.3 Hz, 3H), 1.00 - 0.95 (m, 2H), 0.90 (d, J = 6.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) δ 171.40, 168.15, 154.20 (d, J = 12.8 Hz), 153.79, 153.17, 149.98, 135.30, 133.76 (d, J = 238.8 Hz), 122.85 , 118.83, 106.52, 72.58, 72.14, 64.88, 62.33, 56.73 (d, J = 9.3 Hz), 53.38, 50.71, 39.02, 18.64, 18.61, 10.32, 10.26; MS (ESI) m/z: [(M+1) ) + , 500.1].
实施例2:(2R,4S,4aS)-11-氟-2,4-二甲基-8-(6-氧代-2,5-二氧-7-氮杂螺[3.4]辛烷-7-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物2)Example 2: (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(6-oxo-2,5-dioxo-7-azaspiro[3.4]octane- 7-yl)-1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole[4,5-g][1,4]oxazine[4,3-a]quinoline -5,5'-pyrimidine]-2',4',6'(1'H,3'H)-trione (Compound 2)
(a)3-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)氧杂环丁醇(I-3-2)(a) 3-(((5-(1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)amino)methyl)oxy Heterocyclic butanol (I-3-2)
Figure PCTCN2018092208-appb-000014
Figure PCTCN2018092208-appb-000014
按照中间体I-3-1的合成方法,以中间体A(1.2g,4.857mmol)、N-氯代丁二酰亚胺(778mg,5.828mmol)、3-(氨甲基)氧杂环丁醇(1.25g,12.142mmol)和碳酸铯(4.748g,14.571mmol)为原料,合成1-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)氧杂环丁醇949mg,白色固体,收率59.5%, 1H NMR(400MHz,DMSO)δ8.08(d,J=5.7Hz,1H),7.41(t,J=5.7Hz,1H),6.09(s,1H),6.01(s,1H),4.48(dd,J=15.1,6.6Hz,4H),4.12–3.99(m,4H),3.55(d,J=5.8Hz,2H)。 According to the synthesis method of the intermediate I-3-1, intermediate A (1.2 g, 4.857 mmol), N-chlorosuccinimide (778 mg, 5.828 mmol), 3-(aminomethyl) oxacyclohexane Butanol (1.25g, 12.142mmol) and cesium carbonate (4.748g, 14.571mmol) were used as raw materials to synthesize 1-(((5-(1,3-dioxolan-2-yl)-6,7-di) Fluorobenzo[d]isoxazol-3-yl)amino)methyl)oxybutanol 949 mg, white solid, yield 59.5%, 1 H NMR (400 MHz, DMSO) δ 8.08 (d, J = 5.7 Hz, 1H), 7.41 (t, J = 5.7 Hz, 1H), 6.09 (s, 1H), 6.01 (s, 1H), 4.48 (dd, J = 15.1, 6.6 Hz, 4H), 4.12 - 3.99 ( m, 4H), 3.55 (d, J = 5.8 Hz, 2H).
(b)7-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-2,5-二氧-7-氮杂螺[3.4]辛烷-6-酮(I-4-2)(b) 7-(5-((1,3-Diethoxylan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)-2,5-dioxo -7-azaspiro[3.4]octane-6-one (I-4-2)
Figure PCTCN2018092208-appb-000015
Figure PCTCN2018092208-appb-000015
按照中间体I-4-1的合成方法,以中间体I-3-2(940mg,2.86mmol)、N,N'-羰基二咪唑(1.857g,11.45mmol)和4-二甲基氨基吡啶(175mg,1.43mmol)为原料,合成7-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-2,5-二氧-7-氮杂螺[3.4]辛烷-6-酮872mg,白色固体,收率86%, 1H NMR(400MHz,CDCl 3)δ8.44(dd,J=5.9,1.8Hz,1H),6.12(s,1H),5.10(d,J=8.8Hz,2H),4.82(d,J=8.7Hz,2H),4.49(s,2H),4.20–4.04(m,4H)。 Intermediate I-3-2 (940 mg, 2.86 mmol), N,N'-carbonyldiimidazole (1.857 g, 11.45 mmol) and 4-dimethylaminopyridine according to the procedure of Intermediate 1-4-1. (175 mg, 1.43 mmol) as a starting material for the synthesis of 7-(5-((1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazole-3-yl) -2,5-Dioxa-7-azaspiro[3.4]octane-6-one 872 mg, white solid, yield 86%, 1 H NMR (400 MHz, CDCl 3 ) δ 8.44 (dd, J = 5.9 , 1.8 Hz, 1H), 6.12 (s, 1H), 5.10 (d, J = 8.8 Hz, 2H), 4.82 (d, J = 8.7 Hz, 2H), 4.49 (s, 2H), 4.20 - 4.04 (m , 4H).
(c)6,7-二氟-3-(6-氧代-2,5-二氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛(I-5-2)(c) 6,7-Difluoro-3-(6-oxo-2,5-dioxo-7-azaspiro[3.4]octane-7-yl)benzo[d]isoxazole-5 - Formaldehyde (I-5-2)
Figure PCTCN2018092208-appb-000016
Figure PCTCN2018092208-appb-000016
按照中间体I-5-1的合成方法,以中间体I-4-2(885mg,2.498mmol)和6M盐酸(30ml)为原料,合成6,7-二氟-3-(6-氧代-2,5-二氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛580mg,白色固体,收率75%, 1H NMR(400MHz,CDCl 3)δ10.31(s,1H),8.91(dd,J=5.8,1.8Hz,1H),5.14(d,J=9.0Hz,2H),4.85(d,J=8.7Hz,2H),4.53(s,2H)。 Synthesis of 6,7-difluoro-3-(6-oxo) from Intermediate I-4-2 (885 mg, 2.498 mmol) and 6M hydrochloric acid (30 mL). -2,5-Dioxa-7-azaspiro[3.4]octane-7-yl)benzo[d]isoxazole-5-formaldehyde 580 mg, white solid, yield 75%, 1 H NMR (400 MHz , CDCl 3 ) δ 10.31 (s, 1H), 8.91 (dd, J = 5.8, 1.8 Hz, 1H), 5.14 (d, J = 9.0 Hz, 2H), 4.85 (d, J = 8.7 Hz, 2H) , 4.53 (s, 2H).
(d)6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(6-氧代-2,5-二氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛(I-6-2)(d) 6-((2R,6R)-2,6-Dimethylmorpholine)-7-fluoro-3-(6-oxo-2,5-dioxo-7-azaspiro[3.4] Octane-7-yl)benzo[d]isoxazole-5-carbaldehyde (I-6-2)
Figure PCTCN2018092208-appb-000017
Figure PCTCN2018092208-appb-000017
按照中间体I-6-1的合成方法,以中间体I-5-2(150mg,0.484mmol)、2R,6R-二甲基吗啉(139mg,1.209mmol)和N,N-二异丙基乙胺(187mg,1.451mmol)为原料,合成6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(6-氧代-2,5-二氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛172mg,白色固体,收率87%, 1H NMR(400MHz,CDCl 3)δ10.37(s,1H),8.71(s,1H),5.14(d,J=8.3Hz,2H),4.85(d,J=8.2Hz,2H),4.50(s,2H),4.30–4.22(m,2H),3.45(d,J=12.0Hz,2H),3.06(dd,J=11.7,5.6Hz,2H),1.34(d,J=6.5Hz,6H)。 Intermediate I-5-2 (150 mg, 0.484 mmol), 2R,6R-dimethylmorpholine (139 mg, 1.209 mmol) and N,N-diisopropyl Synthesis of 6-((2R,6R)-2,6-dimethylmorpholine)-7-fluoro-3-(6-oxo-2,5-di) by using ethylamine (187 mg, 1.451 mmol) as a starting material Oxy-7-azaspiro[3.4]octane-7-yl)benzo[d]isoxazole-5-carboxaldehyde 172 mg, white solid, yield 87%, 1 H NMR (400 MHz, CDCl 3 ) δ 10 . 37 (s, 1H), 8.71 (s, 1H), 5.14 (d, J = 8.3 Hz, 2H), 4.85 (d, J = 8.2 Hz, 2H), 4.50 (s, 2H), 4.30 - 4.22 (m) , 2H), 3.45 (d, J = 12.0 Hz, 2H), 3.06 (dd, J = 11.7, 5.6 Hz, 2H), 1.34 (d, J = 6.5 Hz, 6H).
(e)(2R,4S,4aS)-11-氟-2,4-二甲基-8-(6-氧代-2,5-二氧-7-氮杂螺[3.4]辛烷-7-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物2)(e) (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(6-oxo-2,5-dioxo-7-azaspiro[3.4]octane-7 -yl)-1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline- 5,5'-pyrimidine]-2',4',6'(1'H,3'H)-trione (compound 2)
Figure PCTCN2018092208-appb-000018
Figure PCTCN2018092208-appb-000018
按照化合物1的合成方法,以中间体I-6-2(93mg,0.229mmol)和巴比妥酸(33mg,0.252mmol)为原料,合成(2R,4S,4aS)-11-氟-2,4-二甲基-8-(6-氧代-2,5-二氧-7-氮杂螺 [3.4]辛烷-7-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮90mg,白色固体,收率76%, 1H NMR(400MHz,DMSO)δ11.63(s,2H),7.73(s,1H),4.86–4.76(m,4H),4.43–4.36(m,2H),4.10(d,J=12.5Hz,1H),3.93(d,J=8.9Hz,1H),3.83–3.74(m,1H),3.72–3.60(m,2H),3.18–3.05(m,1H),2.90(d,J=13.7Hz,1H),1.14(d,J=6.2Hz,3H),0.89(d,J=6.4Hz,3H); 13C NMR(126MHz,DMSO)δ171.46,168.17,154.15(d,J=13.0Hz),152.88,152.86,150.07,135.28,133.74(d,J=238.9Hz),122.83,118.85,106.49,81.72,81.52,80.54,72.57,72.12,64.90,56.68,53.38,52.55,39.02,18.62,18.62;MS(EI)m/z:[M +,515]。 (2R, 4S, 4aS)-11-fluoro-2 was synthesized according to the synthesis method of compound 1, using intermediate I-6-2 (93 mg, 0.229 mmol) and barbituric acid (33 mg, 0.252 mmol) as raw materials. 4-Dimethyl-8-(6-oxo-2,5-dioxo-7-azaspiro[3.4]octane-7-yl)-1,2,4,4a-tetrahydro-2' H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5,5'-pyrimidine]-2',4',6' ( 1'H,3'H)-trione 90 mg, white solid, yield 76%, 1 H NMR (400 MHz, DMSO) δ 11.63 (s, 2H), 7.73 (s, 1H), 4.86 - 4.76 (m) , 4H), 4.43–4.36 (m, 2H), 4.10 (d, J = 12.5 Hz, 1H), 3.93 (d, J = 8.9 Hz, 1H), 3.83–3.74 (m, 1H), 3.72–3.60 ( m, 2H), 3.18 - 3.05 (m, 1H), 2.90 (d, J = 13.7 Hz, 1H), 1.14 (d, J = 6.2 Hz, 3H), 0.89 (d, J = 6.4 Hz, 3H); 13 C NMR (126 MHz, DMSO) δ 171.46, 168.17, 154.15 (d, J = 13.0 Hz), 152.88, 152.86, 150.07, 135.28, 133.74 (d, J = 238.9 Hz), 122.83, 118.85, 106.49, 81.72, 81.52, 80.54, 72.57, 72.12, 64.90, 56.68, 53.38, 52.55, 39.02, 18.62, 18.62; MS (EI) m/z: [M + , 515].
实施例3:(2R,4S,4aS)-11-氟-2,4-二甲基-8-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物3)Example 3: (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(6-oxo-5-oxo-7-azaspiro[3.4]octane-7-yl -1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5, 5'-pyrimidine]-2',4',6'(1'H,3'H)-trione (compound 3)
(a)1-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)环丁醇(I-3-3)(a) 1-(((5-(1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)amino)methyl)) Butanol (I-3-3)
Figure PCTCN2018092208-appb-000019
Figure PCTCN2018092208-appb-000019
按照中间体I-3-1的合成方法,以中间体A(750mg,3.036mmol)、N-氯代丁二酰亚胺(527mg,3.947mmol)、1-(氨甲基)环丁醇(675mg,6.679mmol)和碳酸铯(3.957g,12.144mmol)为原料,合成1-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)环丁醇710mg,白色固体,收率72%, 1H NMR(400MHz,DMSO)δ8.15(d,J=5.9Hz,1H),7.23(t,J=5.4Hz,1H),6.08(s,1H),5.27(s,1H),4.11–3.99(m,4H),3.34(d,J=5.6Hz,2H),2.12–1.92(m,4H),1.72–1.45(m,2H)。 According to the synthesis method of Intermediate 1-3-1, Intermediate A (750 mg, 3.036 mmol), N-chlorosuccinimide (527 mg, 3.947 mmol), 1-(aminomethyl)cyclobutanol ( 675 mg, 6.679 mmol) and cesium carbonate (3.957 g, 12.144 mmol) were used as starting materials to synthesize 1-(((5-(1,3-dioxolan-2-yl)-6,7-difluorobenzo[ d] isoxazol-3-yl)amino)methyl)cyclobutanol 710 mg, white solid, yield 72%, 1 H NMR (400 MHz, DMSO) δ 8.15 (d, J = 5.9 Hz, 1H), 7.23 (t, J = 5.4 Hz, 1H), 6.08 (s, 1H), 5.27 (s, 1H), 4.11 - 3.99 (m, 4H), 3.34 (d, J = 5.6 Hz, 2H), 2.12 - 1.92 (m, 4H), 1.72–1.45 (m, 2H).
(b)7-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-5-氧-7-氮杂螺[3.4]辛烷-6-酮(I-4-3)(b) 7-(5-((1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)-5-oxo-7- Azaspiro[3.4]octane-6-one (I-4-3)
Figure PCTCN2018092208-appb-000020
Figure PCTCN2018092208-appb-000020
按照中间体I-4-1的合成方法,以中间体I-3-3(533mg,1.63mmol)、N,N'-羰基二咪唑(1.059g,6.53mmol)和4-二甲基氨基吡啶(100mg,0.82mmol)为原料,合成7-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-5-氧-7-氮杂螺[3.4]辛烷-6-酮468mg,白色固体,收率82%, 1H NMR(400MHz,CDCl 3)δ8.51(dd,J=5.9,1.8Hz,1H),6.15(s,1H),4.26(s,2H),4.22–4.07(m,4H),2.71(ddd,J=19.3,10.1,2.9Hz,2H),2.38(ddd,J=11.4,8.5,3.7Hz,2H),2.09–1.97(m,1H),1.85–1.71(m,1H)。 Intermediate I-3-3 (533 mg, 1.63 mmol), N,N'-carbonyldiimidazole (1.059 g, 6.53 mmol) and 4-dimethylaminopyridine according to the procedure of Intermediate 1-4-1. (100 mg, 0.82 mmol) was used as a starting material to synthesize 7-(5-((1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl) -5-oxo-7-azaspiro[3.4]octane-6-one 468 mg, white solid, yield 82%, 1 H NMR (400 MHz, CDCl 3 ) δ 8.51 (dd, J = 5.9, 1.8 Hz , 1H), 6.15 (s, 1H), 4.26 (s, 2H), 4.22 - 4.07 (m, 4H), 2.71 (ddd, J = 19.3, 10.1, 2.9 Hz, 2H), 2.38 (ddd, J = 11.4) , 8.5, 3.7 Hz, 2H), 2.09 - 1.97 (m, 1H), 1.85 - 1.71 (m, 1H).
(c)6,7-二氟-3-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛(I-5-3)(c) 6,7-Difluoro-3-(6-oxo-5-oxo-7-azaspiro[3.4]octane-7-yl)benzo[d]isoxazole-5-carbaldehyde ( I-5-3)
Figure PCTCN2018092208-appb-000021
Figure PCTCN2018092208-appb-000021
按照中间体I-5-1的合成方法,以中间体I-4-3(468mg,1.328mmol)和6M盐酸(15ml)为原料,合成6,7-二氟-3-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛380mg,白色固体,收率93%, 1H NMR(400MHz,CDCl 3)δ10.29(s,1H),8.95(dd,J=5.8,1.7Hz,1H),4.27(s,2H),2.73(ddd,J=19.5,10.1,2.9Hz,2H),2.44–2.35(m,2H),2.10–2.00(m,1H),1.87–1.74(m,1H)。 The synthesis of 6,7-difluoro-3-(6-oxo) was carried out according to the synthesis of the intermediate I-5-1 using the intermediate I-4-3 (468 mg, 1.328 mmol) and 6M hydrochloric acid (15 ml). -5-oxo-7-azaspiro[3.4]octane-7-yl)benzo[d]isoxazole-5-carbaldehyde 380 mg, white solid, yield 93%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.29 (s, 1H), 8.95 (dd, J = 5.8, 1.7 Hz, 1H), 4.27 (s, 2H), 2.73 (ddd, J = 19.5, 10.1, 2.9 Hz, 2H), 2.44 - 2.35 (m, 2H), 2.10 - 2.00 (m, 1H), 1.87 - 1.74 (m, 1H).
(d)6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛(I-6-3)(d) 6-((2R,6R)-2,6-dimethylmorpholine)-7-fluoro-3-(6-oxo-5-oxo-7-azaspiro[3.4]octane- 7-yl)benzo[d]isoxazole-5-carbaldehyde (I-6-3)
Figure PCTCN2018092208-appb-000022
Figure PCTCN2018092208-appb-000022
按照中间体I-6-1的合成方法,以中间体I-5-3(100mg,0.324mmol)、2R,6R-二甲基吗啉(75mg,0.649mmol)和N,N-二异丙基乙胺(126mg,0.973mmol)为原料,合成6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)苯并[d]异噁唑-5-甲醛103mg,白色固体,收率79%, 1H NMR(400MHz,CDCl 3)δ10.38(s,1H),8.79(s,1H),4.31–4.20(m,4H),3.44(d,J=12.1Hz,2H),3.05(dd,J=11.9,5.6Hz,2H),2.72(ddd,J=19.8,10.0,3.2Hz,2H),2.43–2.34(m,2H),2.10–1.98(m,1H),1.85–1.73(m,1H),1.34(d,J=6.4Hz,6H)。 According to the synthesis of the intermediate I-6-1, intermediate I-5-3 (100 mg, 0.324 mmol), 2R, 6R-dimethylmorpholine (75 mg, 0.649 mmol) and N,N-diisopropyl Synthesis of 6-((2R,6R)-2,6-dimethylmorpholine)-7-fluoro-3-(6-oxo-5-oxo-7) with ethylamine (126 mg, 0.973 mmol) as starting material - azaspiro[3.4]octane-7-yl)benzo[d]isoxazole-5-carbaldehyde 103 mg, white solid, yield 79%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.38 (s , 1H), 8.79 (s, 1H), 4.31 - 4.20 (m, 4H), 3.44 (d, J = 12.1 Hz, 2H), 3.05 (dd, J = 11.9, 5.6 Hz, 2H), 2.72 (ddd, J=19.8, 10.0, 3.2 Hz, 2H), 2.43–2.34 (m, 2H), 2.10–1.98 (m, 1H), 1.85–1.73 (m, 1H), 1.34 (d, J=6.4 Hz, 6H) .
(e)(2R,4S,4aS)-11-氟-2,4-二甲基-8-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物3)(e) (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(6-oxo-5-oxo-7-azaspiro[3.4]octane-7-yl) -1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5,5 '-pyrimidine]-2',4',6'(1'H,3'H)-trione (compound 3)
Figure PCTCN2018092208-appb-000023
Figure PCTCN2018092208-appb-000023
按照化合物1的合成方法,以中间体I-6-3(92mg,0.228mmol)和巴比妥酸(32mg, 0.251mmol)为原料,合成(2R,4S,4aS)-11-氟-2,4-二甲基-8-(6-氧代-5-氧-7-氮杂螺[3.4]辛烷-7-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮99mg,白色固体,收率84%, 1H NMR(400MHz,DMSO)δ11.73(s,1H),11.47(s,1H),7.70(s,1H),4.23(s,2H),4.10(d,J=12.5Hz,1H),3.93(d,J=8.8Hz,1H),3.84–3.74(m,1H),3.71–3.62(m,2H),3.17–3.05(m,1H),2.91(d,J=13.9Hz,1H),2.50–2.32(m,4H),1.89–1.63(m,2H),1.15(d,J=6.3Hz,3H),0.89(d,J=6.3Hz,3H). 13C NMR(126MHz,DMSO)δ171.41,168.15,154.12(d,J=12.9Hz),153.34,153.24,150.00,135.26,133.76(d,J=238.8Hz),122.77,118.81,106.65,81.39,72.57,72.13,64.87,56.73(d,J=9.4Hz),55.57,53.37,39.01,34.95,34.79,18.64,18.61,11.96;MS(EI)m/z:[M +,513]。 (2R, 4S, 4aS)-11-fluoro-2 was synthesized according to the synthesis method of compound 1, using intermediate I-6-3 (92 mg, 0.228 mmol) and barbituric acid (32 mg, 0.251 mmol) as raw materials. 4-Dimethyl-8-(6-oxo-5-oxo-7-azaspiro[3.4]octane-7-yl)-1,2,4,4a-tetrahydro-2'H,6H - snail [isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5,5'-pyrimidine]-2',4',6'(1'H , 3'H) - 99 mg of trione, as a white solid, yield 84%, 1 H NMR (400MHz , DMSO) δ11.73 (s, 1H), 11.47 (s, 1H), 7.70 (s, 1H), 4.23 (s, 2H), 4.10 (d, J = 12.5 Hz, 1H), 3.93 (d, J = 8.8 Hz, 1H), 3.84 - 3.74 (m, 1H), 3.71 - 3.62 (m, 2H), 3.17 - 3.05 (m, 1H), 2.91 (d, J = 13.9 Hz, 1H), 2.50 - 2.32 (m, 4H), 1.89 - 1.63 (m, 2H), 1.15 (d, J = 6.3 Hz, 3H), 0.89 (d, J = 6.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) δ 171.41, 168.15, 154.12 (d, J = 12.9 Hz), 153.34, 153.24, 150.00, 135.26, 133.76 (d, J = 238.8 Hz) , 122.77, 118.81, 106.65, 81.39, 72.57, 72.13, 64.87, 56.73 (d, J = 9.4 Hz), 55.57, 53.37, 39.01, 34.95, 34.79, 18.64, 18.61, 11.96; MS (EI) m/z: [ M + , 513].
实施例4:(2R,4S,4aS)-11-氟-2,4-二甲基-8-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物4)Example 4: (2R,4S,4aS)-11-fluoro-2,4-dimethyl-8-(2-oxo-1-oxo-3-azaspiro[4.4]decane-3-yl -1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5, 5'-pyrimidine]-2',4',6'(1'H,3'H)-trione (compound 4)
(a)1-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)环戊醇(I-3-4)(a) 1-(((5-(1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)amino)methyl)) Pentanol (I-3-4)
Figure PCTCN2018092208-appb-000024
Figure PCTCN2018092208-appb-000024
按照中间体I-3-1的合成方法,以中间体A(600mg,2.430mmol)、N-氯代丁二酰亚胺(422mg,3.160mmol)、1-(氨甲基)环戊醇(615mg,5.346mmol)和碳酸铯(3.167g,9.720mmol)为原料,合成1-(((5-(1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)氨基)甲基)环戊醇582mg,白色固体,收率70%, 1H NMR(400MHz,DMSO)δ8.13(d,J=5.0Hz,1H),7.19(t,J=5.6Hz,1H),6.08(s,1H),4.51(s,1H),4.14–3.99(m,4H),3.31(d,J=5.8Hz,2H),1.78–1.50(m,8H)。 According to the synthesis method of Intermediate 1-3-1, Intermediate A (600 mg, 2.430 mmol), N-chlorosuccinimide (422 mg, 3.160 mmol), 1-(aminomethyl)cyclopentanol ( 615 mg, 5.346 mmol) and cesium carbonate (3.167 g, 9.720 mmol) were used as starting materials to synthesize 1-(((5-(1,3-dioxolan-2-yl)-6,7-difluorobenzo[ d] isoxazol-3-yl)amino)methyl)cyclopentanol 582 mg, white solid, yield 70%, 1 H NMR (400 MHz, DMSO) δ 8.13 (d, J = 5.0 Hz, 1H), 7.19 (t, J = 5.6 Hz, 1H), 6.08 (s, 1H), 4.51 (s, 1H), 4.14 - 3.99 (m, 4H), 3.31 (d, J = 5.8 Hz, 2H), 1.78 - 1.50 (m, 8H).
(b)3-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-1-氧-3-氮杂螺[4.4]壬烷-2-酮(I-4-4)(b) 3-(5-((1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazol-3-yl)-1-oxo-3- Azaspiro[4.4]decane-2-one (I-4-4)
Figure PCTCN2018092208-appb-000025
Figure PCTCN2018092208-appb-000025
按照中间体I-4-1的合成方法,以中间体I-3-4(526mg,1.546mmol)、N,N'-羰基二咪唑(1.002g,6.182mmol)和4-二甲基氨基吡啶(95mg,0.773mmol)为原料,合成3-(5-((1,3-二氧戊环-2-基)-6,7-二氟苯并[d]异噁唑-3-基)-1-氧-3-氮杂螺[4.4]壬烷-2-酮450mg,白色固体,收率79%, 1H NMR(400MHz,CDCl 3)δ8.54(dd,J=5.9,1.7 Hz,1H),6.16(s,1H),4.23–4.07(m,6H),2.32–2.21(m,2H),2.03–1.80(m,6H)。 Intermediate 1-3-4 (526 mg, 1.546 mmol), N,N'-carbonyldiimidazole (1.002 g, 6.182 mmol) and 4-dimethylaminopyridine according to the procedure of Intermediate 1-4-1. (95mg, 0.773mmol) was used as raw material to synthesize 3-(5-((1,3-dioxolan-2-yl)-6,7-difluorobenzo[d]isoxazole-3-yl) 1-oxo-3-azaspiro[4.4]nonan-2-one 450 mg, white solid, yield 79%, 1 H NMR (400 MHz, CDCl 3 ) δ 8.54 (dd, J = 5.9, 1.7 Hz , 1H), 6.16 (s, 1H), 4.23 - 4.07 (m, 6H), 2.32 - 2.21 (m, 2H), 2.03 - 1.80 (m, 6H).
(c)6,7-二氟-3-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)苯并[d]异噁唑-5-甲醛(I-5-4)(c) 6,7-Difluoro-3-(2-oxo-1-oxo-3-azaspiro[4.4]decane-3-yl)benzo[d]isoxazole-5-carbaldehyde ( I-5-4)
Figure PCTCN2018092208-appb-000026
Figure PCTCN2018092208-appb-000026
按照中间体I-5-1的合成方法,以中间体I-4-4(158mg,0.431mmol)和6M盐酸(5ml)为原料,合成6,7-二氟-3-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)苯并[d]异噁唑-5-甲醛135mg,白色固体,收率97%, 1H NMR(400MHz,CDCl 3)δ10.30(s,1H),8.98(dd,J=5.9,1.8Hz,1H),4.17(s,2H),2.33–2.22(m,2H),2.08–1.81(m,6H)。 Synthesis of 6,7-difluoro-3-(2-oxo) from the intermediate I-4-4 (158 mg, 0.431 mmol) and 6M hydrochloric acid (5 ml). -1-Oxo-3-azaspiro[4.4]decane-3-yl)benzo[d]isoxazole-5-carboxaldehyde 135 mg, white solid, yield 97%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.30 (s, 1H), 8.98 (dd, J = 5.9, 1.8 Hz, 1H), 4.17 (s, 2H), 2.33 - 2.22 (m, 2H), 2.08 - 1.81 (m, 6H).
(d)6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)苯并[d]异噁唑-5-甲醛(I-6-4)(d) 6-((2R,6R)-2,6-dimethylmorpholine)-7-fluoro-3-(2-oxo-1-oxo-3-azaspiro[4.4]decane- 3-yl)benzo[d]isoxazole-5-formaldehyde (I-6-4)
Figure PCTCN2018092208-appb-000027
Figure PCTCN2018092208-appb-000027
按照中间体I-6-1的合成方法,以中间体I-5-4(100mg,0.310mmol)、2R,6R-二甲基吗啉(72mg,0.620mmol)和N,N-二异丙基乙胺(120mg,0.931mmol)为原料,合成6-((2R,6R)-2,6-二甲基吗啉)-7-氟-3-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)苯并[d]异噁唑-5-甲醛114mg,淡白色固体,收率88%, 1H NMR(400MHz,CDCl 3)δ10.38(s,1H),8.81(s,1H),4.31–4.21(m,2H),4.15(s,2H),3.44(d,J=12.1Hz,2H),3.05(dd,J=11.8,5.5Hz,2H),2.28(m,2H),2.06–1.80(m,6H),1.34(d,J=6.4Hz,6H)。 Intermediate 1--5-4 (100 mg, 0.310 mmol), 2R,6R-dimethylmorpholine (72 mg, 0.620 mmol) and N,N-diisopropyl. Synthesis of 6-((2R,6R)-2,6-dimethylmorpholine)-7-fluoro-3-(2-oxo-1-oxo-3) by using ethylamine (120 mg, 0.931 mmol) as a starting material - azaspiro[4.4]decane-3-yl)benzo[d]isoxazole-5-formaldehyde 114 mg, pale white solid, yield 88%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.38 ( s, 1H), 8.81 (s, 1H), 4.31 - 4.21 (m, 2H), 4.15 (s, 2H), 3.44 (d, J = 12.1 Hz, 2H), 3.05 (dd, J = 11.8, 5.5 Hz , 2H), 2.28 (m, 2H), 2.06 - 1.80 (m, 6H), 1.34 (d, J = 6.4 Hz, 6H).
(e)(2R,4S,4aS)-11-氟-2,4-二甲基-8-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮(化合物4)(e) (2R, 4S, 4aS)-11-fluoro-2,4-dimethyl-8-(2-oxo-1-oxo-3-azaspiro[4.4]decane-3-yl) -1,2,4,4a-tetrahydro-2'H,6H-spiro[isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5,5 '-pyrimidine]-2',4',6'(1'H,3'H)-trione (Compound 4)
Figure PCTCN2018092208-appb-000028
Figure PCTCN2018092208-appb-000028
按照化合物1的合成方法,以中间体I-6-4(99mg,0.237mmol)和巴比妥酸(34mg,0.261mmol)为原料,合成(2R,4S,4aS)-11-氟-2,4-二甲基-8-(2-氧代-1-氧-3-氮杂螺[4.4]壬烷-3-基)-1,2,4,4a-四氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-2',4',6'(1'H,3'H)-三酮94mg,白色固体,收率75%, 1H NMR(400MHz,DMSO)δ11.82(s,1H),11.46(s,1H),7.73(s,1H),4.14–4.08(m,3H),3.93(d,J=8.8Hz,1H),3.83–3.74(m,1H),3.70–3.62(m,2H),3.15–3.07(m,1H),2.91(d,J=13.7Hz,1H),2.12–2.04(m,2H),1.96–1.87(m,2H),1.80–1.71(m,4H),1.15(d,J=6.2Hz,3H),0.89(d,J=6.4Hz,3H); 13C NMR(151MHz,DMSO)δ170.96,167.69,153.68(d,J=12.9Hz),153.06,152.90,149.54,134.78,133.29(d,J=238.8Hz),122.23,118.53,106.19,90.29,72.12,71.68,64.42,56.27(d,J=8.9Hz),53.33,52.92,38.55,37.72,37.55,23.11,23.08,18.19,18.17;MS(EI)m/z:[M +,527]。 (2R, 4S, 4aS)-11-fluoro-2 was synthesized according to the synthesis method of compound 1, using intermediate I-6-4 (99 mg, 0.237 mmol) and barbituric acid (34 mg, 0.261 mmol) as raw materials. 4-Dimethyl-8-(2-oxo-1-oxo-3-azaspiro[4.4]decane-3-yl)-1,2,4,4a-tetrahydro-2'H,6H - snail [isoxazole [4,5-g][1,4]oxazine [4,3-a]quinoline-5,5'-pyrimidine]-2',4',6'(1'H , 3'H)-trione 94 mg, white solid, yield 75%, 1 H NMR (400 MHz, DMSO) δ 11.82 (s, 1H), 11.46 (s, 1H), 7.73 (s, 1H), 4.14 –4.08(m,3H),3.93(d,J=8.8Hz,1H),3.83–3.74(m,1H), 3.70–3.62(m,2H),3.15–3.07(m,1H),2.91(d , J = 13.7 Hz, 1H), 2.12 - 2.04 (m, 2H), 1.96 - 1.87 (m, 2H), 1.80 - 1.71 (m, 4H), 1.15 (d, J = 6.2 Hz, 3H), 0.89 ( d, J = 6.4 Hz, 3H); 13 C NMR (151 MHz, DMSO) δ 170.96, 167.69, 153.68 (d, J = 12.9 Hz), 153.66, 152.90, 149.54, 134.78, 133.29 (d, J = 238.8 Hz), 122.23, 118.53, 106.19, 90.29, 72.12, 71.68, 64.42, 56.27 (d, J = 8.9 Hz), 53.33, 52.92, 38.55, 37.72, 37.55, 23.11, 23.08, 18.19, 18.17; MS (EI) m/z: [M + , 527].
实施例5:(2R,4S,4aS)-2,4-二甲基-2',4',6'-三氧代-8-(5-氧代-4-氧杂-6-氮杂螺[2.4]庚烷-6-基)-1,1',2,3',4,4a,4',6'-八氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-11-腈(化合物5)Example 5: (2R, 4S, 4aS)-2,4-dimethyl-2',4',6'-trioxo-8-(5-oxo-4-oxa-6-aza Spirulina [2.4]heptane-6-yl)-1,1',2,3',4,4a,4',6'-octahydro-2'H,6H-spiro[isoxazole [4,5 -g][1,4]oxazine [4,3-a]quinoline-5,5'-pyrimidine]-11-carbonitrile (Compound 5)
(a)2,6-二氟-3-氰基苯甲醛(I-7)(a) 2,6-difluoro-3-cyanobenzaldehyde (I-7)
Figure PCTCN2018092208-appb-000029
Figure PCTCN2018092208-appb-000029
室温下,将2M二异丙基氨基锂的四氢呋喃溶液(14ml,28.04mmol)加入到20ml干燥的四氢呋喃中,氩气保护下,降温至-80℃,缓慢滴加2,6-二氟苯腈(3.00g,21.57mmol)的四氢呋喃溶液,5min后,缓慢滴加N,N-二甲基甲酰胺(5.00ml,64.71mmol),5min后,TLC监测反应完全,加入饱和氯化铵淬灭反应,升至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=2:1],得无色油状物1.72g,收率48%, 1H NMR(400MHz,CDCl 3)δ10.32(d,J=0.6Hz,1H),8.20(ddd,J=8.9,7.8,6.2Hz,1H),7.27–7.22(m,1H)。 2M diisopropylamide lithium tetrahydrofuran solution (14 ml, 28.04 mmol) was added to 20 ml of dry tetrahydrofuran under argon atmosphere, cooled to -80 ° C, and 2,6-difluorobenzonitrile was slowly added dropwise. (3.00 g, 21.57 mmol) in tetrahydrofuran solution. After 5 min, N,N-dimethylformamide (5.00 ml, 64.71 mmol) was slowly added dropwise. After 5 min, the reaction was monitored by TLC and quenched with saturated ammonium chloride. ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 1.72g of oil, yield 48%, 1 H NMR (400MHz, CDCl 3 ) δ 10.32 (d, J = 0.6 Hz, 1H), 8.20 (ddd, J = 8.9, 7.8, 6.2 Hz, 1H), 7.27 –7.22 (m, 1H).
(b)3-(1,3-二氧戊环-2-基)-2,6-二氟苯腈(I-8)(b) 3-(1,3-dioxolan-2-yl)-2,6-difluorobenzonitrile (I-8)
Figure PCTCN2018092208-appb-000030
Figure PCTCN2018092208-appb-000030
室温下,将中间体I-7(2.00g,11.97mmol)加入到15ml甲苯中,搅拌溶解,加入乙二醇(2.00ml,35.92mmol)和对甲苯磺酸一水合物(227mg,1.197mmol),搭载分水器,升至110℃反应3h,TLC监测反应完全,降至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=4:1],得无色油状物1.92g,收率76%, 1H NMR(400MHz,CDCl 3)δ7.85–7.79(m,1H),7.12–7.06(m,1H),6.04(s,1H),4.18–4.07(m,4H)。 Intermediate I-7 (2.00 g, 11.97 mmol) was added to 15 ml of toluene at room temperature, stirred and dissolved, and ethylene glycol (2.00 ml, 35.92 mmol) and p-toluenesulfonic acid monohydrate (227 mg, 1.197 mmol) were added. , equipped with a water separator, raised to 110 ° C for 3 h, TLC monitoring reaction was completed, cooled to room temperature, extracted with water and ethyl acetate, combined organic layer, washed with water, washed with saturated brine, dried over anhydrous sodium sulfate Analysis [petroleum ether: ethyl acetate = 4:1] afforded 1.92 g of colorless oil, yield 76%, 1 H NMR (400 MHz, CDCl 3 ) δ 7.85 - 7.79 (m, 1H), 7.12 - 7.06 (m, 1H), 6.04 (s, 1H), 4.18 - 4.07 (m, 4H).
(c)3-(1,3-二氧戊环-2-基)-2,6-二氟-5-甲酰基苯腈(I-9)(c) 3-(1,3-dioxolan-2-yl)-2,6-difluoro-5-formylbenzonitrile (I-9)
Figure PCTCN2018092208-appb-000031
Figure PCTCN2018092208-appb-000031
室温下,将2M二异丙基氨基锂的四氢呋喃溶液(7.7ml,15.41mmol)加入到15ml干燥的四氢呋喃中,氩气保护下,降温至-80℃,缓慢滴加中间体I-8(2.71g,12.84mmol)的四氢呋喃溶液,30min后,缓慢滴加N,N-二甲基甲酰胺(2.97ml,38.52mmol),30min后,TLC监测反应完全,加入饱和氯化铵淬灭反应,升至室温,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=5:1],得无色油状物1.95g,收率64%, 1H NMR(400MHz,CDCl 3)δ10.31(s,1H),8.36(t,J=7.7Hz,1H),6.07(s,1H),4.21–4.06(m,4H)。 2M diisopropylamide lithium tetrahydrofuran solution (7.7 ml, 15.41 mmol) was added to 15 ml of dry tetrahydrofuran under argon atmosphere, cooled to -80 ° C, and slowly added dropwise intermediate I-8 (2.71) g, 12.84 mmol) in tetrahydrofuran solution. After 30 min, N,N-dimethylformamide (2.97 ml, 38.52 mmol) was slowly added dropwise. After 30 min, the reaction was completely monitored by TLC, and saturated ammonium chloride was added to quench the reaction. The mixture was combined with EtOAc. EtOAc. 1.95g, yield 64%, 1 H NMR (400MHz, CDCl 3 ) δ 10.31 (s, 1H), 8.36 (t, J = 7.7 Hz, 1H), 6.07 (s, 1H), 4.21 - 4.06 (m) , 4H).
(d)3-(1,3-二氧戊环-2-基)-2,6-二氟-5-((肟基)甲基)苯腈(I-10)(d) 3-(1,3-dioxolan-2-yl)-2,6-difluoro-5-((indolyl)methyl)benzonitrile (I-10)
Figure PCTCN2018092208-appb-000032
Figure PCTCN2018092208-appb-000032
冰浴条件下,将中间体I-9(700mg,2.929mmol)加入到20ml甲醇中,搅拌使其溶解,依次加入盐酸羟胺(224mg,3.222mmol)和吡啶(0.31ml,3.808mmol),升至室温搅拌1h,TLC监测反应完全,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=2:1],得白色固体720mg,收率96%, 1H NMR(400MHz,CDCl 3)δ8.28(s,1H),8.23(t,J=7.8Hz,1H),7.79(s,1H),6.02(s,1H),4.17–4.05(m,4H)。 Intermediate I-9 (700 mg, 2.929 mmol) was added to 20 ml of methanol under ice-cooling, and stirred to dissolve. Hydrochloric acid hydrochloride (224 mg, 3.222 mmol) and pyridine (0.31 ml, 3.808 mmol) were added to the mixture. After stirring at room temperature for 1 h, the reaction was completed with EtOAc EtOAc (EtOAc)EtOAc. , 720 mg of white solid, yield 96%, 1 H NMR (400 MHz, CDCl 3 ) δ 8.28 (s, 1H), 8.23 (t, J = 7.8 Hz, 1H), 7.79 (s, 1H), 6.02 ( s, 1H), 4.17 - 4.05 (m, 4H).
(e)3-氰基-5-(1,3-二氧戊环-2-基)-2,4-二氟-N'-羟基-N-((1-羟基环丙基)甲基)苯甲脒(I-12-5)(e) 3-cyano-5-(1,3-dioxolan-2-yl)-2,4-difluoro-N'-hydroxy-N-((1-hydroxycyclopropyl)methyl Benzoquinone (I-12-5)
Figure PCTCN2018092208-appb-000033
Figure PCTCN2018092208-appb-000033
室温下,将中间体I-10(400mg,1.574mmol)加入到20ml N,N-二甲基甲酰胺中,搅拌使其溶解,加入N-氯代丁二酰亚胺(252mg,1.889mmol),升至40℃反应30min,TLC监测反应完全,将反应液降至0℃,缓慢滴加1-氨甲基-1-环丙醇(343mg,3.935mmol),10min后,TLC监测反应完全,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=1:3],得淡黄色粉末150mg,收率28%, 1H NMR(400MHz,DMSO)δ10.05(s,1H),7.85(t,J=8.0Hz,1H),6.07–6.02(m,2H),5.27(s,1H),4.10–3.97(m,4H),2.90(d,J=6.4Hz,2H),0.48(dd,J=6.7,4.8Hz,2H),0.37(dd,J=6.5,4.6Hz,2H)。 Intermediate I-10 (400 mg, 1.574 mmol) was added to 20 ml of N,N-dimethylformamide at room temperature, stirred to dissolve, and N-chlorosuccinimide (252 mg, 1.889 mmol) was added. The reaction was carried out to 40 ° C for 30 min, and the reaction was completely monitored by TLC. The reaction mixture was dropped to 0 ° C, and 1-aminomethyl-1-cyclopropanol (343 mg, 3.935 mmol) was slowly added dropwise. After 10 min, the reaction was completely monitored by TLC. After adding water and ethyl acetate, the organic layer was combined, washed with water, brine, dried over anhydrous sodium sulfate 28%, 1 H NMR (400 MHz, DMSO) δ 10.05 (s, 1H), 7.85 (t, J = 8.0 Hz, 1H), 6.07 - 6.02 (m, 2H), 5.27 (s, 1H), 4.10– 3.97 (m, 4H), 2.90 (d, J = 6.4 Hz, 2H), 0.48 (dd, J = 6.7, 4.8 Hz, 2H), 0.37 (dd, J = 6.5, 4.6 Hz, 2H).
(f)6-氟-5-甲酰基-3-(((1-羟基环丙基)甲基)氨基)苯并[d]异噁唑-7-腈(I-14-5)(f) 6-Fluoro-5-formyl-3-(((1-hydroxycyclopropyl)methyl)amino)benzo[d]isoxazole-7-carbonitrile (I-14-5)
Figure PCTCN2018092208-appb-000034
Figure PCTCN2018092208-appb-000034
室温下,将中间体I-12-5(150mg,0.442mmol)加入到10ml 1,4-二氧六环中,加6M盐酸(4ml),升至35℃搅拌2h,TLC监测反应完全,将反应液降至0℃,加饱和碳酸氢钠水溶液调pH值至9,搅拌30min,TLC监测反应完全,加水加乙酸乙酯萃取,合并有机层,水洗,饱和食盐水洗,无水硫酸钠干燥,减压浓缩柱层析[石油醚:乙酸乙酯=1:3],得黄色粉末83mg,收率68%, 1H NMR(400MHz,DMSO)δ10.17(s,1H),8.97(d,J=6.6Hz,1H),7.79(t,J=5.4Hz,1H),5.49(s,1H),3.36(d,J=5.9Hz,2H),0.63(d,J=7.3Hz,4H)。 Intermediate I-12-5 (150 mg, 0.442 mmol) was added to 10 ml of 1,4-dioxane, 6M hydrochloric acid (4 ml) was added, and the mixture was stirred at 35 ° C for 2 h, and the reaction was completely monitored by TLC. The reaction mixture was cooled to 0 ° C, and the mixture was stirred and evaporated to dryness. Column chromatography under reduced pressure [petroleum ether: ethyl acetate = 1 : 3] afforded a yellow powder (yield: 68 mg, yield: 68%, 1 H NMR (400 MHz, DMSO) δ 10.17 (s, 1H), 8.97 (d, J = 6.6 Hz, 1H), 7.79 (t, J = 5.4 Hz, 1H), 5.49 (s, 1H), 3.36 (d, J = 5.9 Hz, 2H), 0.63 (d, J = 7.3 Hz, 4H) .
(g)6-((2R,6R)-2,6-二甲基吗啉)-5-甲酰基-3-(((1-羟基环丙基)甲基)氨基)苯并[d]异噁唑-7-腈(I-15-5)(g) 6-((2R,6R)-2,6-dimethylmorpholine)-5-formyl-3-(((1-hydroxycyclopropyl)methyl)amino)benzo[d] Isoxazole-7-carbonitrile (I-15-5)
Figure PCTCN2018092208-appb-000035
Figure PCTCN2018092208-appb-000035
室温下,将中间体I-14-5(83mg,0.323mmol)加入到20ml乙腈中,加入2R,6R-二甲基吗啉(0.065ml,0.484mmol)和N,N-二异丙基乙基胺(0.110ml,0.646mmol),升至40℃搅拌2h,TLC监测反应完全,降至室温,直接拌样柱层析[石油醚:乙酸乙酯=2:3],得黄色固体50mg,收率42%, 1H NMR(400MHz,CDCl 3)δ10.18(s,1H),8.22(s,1H),5.23(t,J=5.3Hz,1H),4.37–4.28(m,2H),3.76(dd,J=12.3,3.0Hz,2H),3.56(d,J=5.4Hz,2H),3.27(dd,J=12.3,5.7Hz,2H),1.34(d,J=6.5Hz,6H),0.95(dd,J=6.5,5.7Hz,2H),0.76(dd,J=6.8,5.5Hz,2H)。 Intermediate I-14-5 (83 mg, 0.323 mmol) was added to 20 ml of acetonitrile at room temperature, and 2R,6R-dimethylmorpholine (0.065 ml, 0.484 mmol) and N,N-diisopropyl The amine (0.110 ml, 0.646 mmol) was stirred at 40 ° C for 2 h, and the reaction was completed by TLC. The mixture was taken to room temperature, and the mixture was directly subjected to column chromatography [ petroleum ether: ethyl acetate = 2:3] to obtain 50 mg of yellow solid. Yield 42%, 1 H NMR (400MHz, CDCl 3 ) δ 10.18 (s, 1H), 8.22 (s, 1H), 5.23 (t, J = 5.3 Hz, 1H), 4.37 - 4.28 (m, 2H) , 3.76 (dd, J = 12.3, 3.0 Hz, 2H), 3.56 (d, J = 5.4 Hz, 2H), 3.27 (dd, J = 12.3, 5.7 Hz, 2H), 1.34 (d, J = 6.5 Hz, 6H), 0.95 (dd, J = 6.5, 5.7 Hz, 2H), 0.76 (dd, J = 6.8, 5.5 Hz, 2H).
(h)6-((2R,6R)-2,6-二甲基吗啉)-5-甲酰基-3-(5-氧代-4-氧杂-6-氮杂螺[2.4]庚烷-6-基)苯并[d]异噁唑-7-腈(I-16-5)(h) 6-((2R,6R)-2,6-dimethylmorpholine)-5-formyl-3-(5-oxo-4-oxa-6-azaspiro[2.4]g Alkan-6-yl)benzo[d]isoxazole-7-carbonitrile (I-16-5)
Figure PCTCN2018092208-appb-000036
Figure PCTCN2018092208-appb-000036
按照中间体I-4-1的合成方法,以中间体I-15-5(50mg,0.135mmol)、N,N-羰 基二咪唑(44mg,0.270mmol)、4-二甲氨基吡啶(17mg,0.135mmol)为原料,制得黄色固体20mg,收率38%, 1H NMR(400MHz,CDCl 3)δ10.06(s,1H),9.17(s,1H),4.40–4.32(m,2H),4.30(d,J=2.3Hz,2H),3.80(dd,J=12.7,2.9Hz,2H),3.23(dd,J=12.7,6.0Hz,2H),1.43(t,J=7.4Hz,2H),1.32(d,J=6.4Hz,6H),0.99–0.94(m,2H)。 Intermediate I-15-5 (50 mg, 0.135 mmol), N,N-carbonyldiimidazole (44 mg, 0.270 mmol), 4-dimethylaminopyridine (17 mg, 0.135 mmol) as a starting material, 20 mg of a yellow solid, yield 38%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.06 (s, 1H), 9.17 (s, 1H), 4.40 - 4.32 (m, 2H) , 4.30 (d, J = 2.3 Hz, 2H), 3.80 (dd, J = 12.7, 2.9 Hz, 2H), 3.23 (dd, J = 12.7, 6.0 Hz, 2H), 1.43 (t, J = 7.4 Hz, 2H), 1.32 (d, J = 6.4 Hz, 6H), 0.99 - 0.94 (m, 2H).
(i)(2R,4S,4aS)-2,4-二甲基-2',4',6'-三氧代-8-(5-氧代-4-氧杂-6-氮杂螺[2.4]庚烷-6-基)-1,1',2,3',4,4a,4',6'-八氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-11-腈(化合物5)(i) (2R, 4S, 4aS)-2,4-dimethyl-2',4',6'-trioxo-8-(5-oxo-4-oxa-6-aza snail [2.4]heptane-6-yl)-1,1',2,3',4,4a,4',6'-octahydro-2'H,6H-spiro[isoxazole [4,5- g][1,4]oxazine [4,3-a]quinoline-5,5'-pyrimidine]-11-carbonitrile (Compound 5)
Figure PCTCN2018092208-appb-000037
Figure PCTCN2018092208-appb-000037
以中间体I-16-5(20mg,0.050mmol)和巴比妥酸(8mg,0.060mmol)为原料,按照化合物1的合成方法,制得淡黄色固体15mg,收率59%, 1H NMR(400MHz,DMSO)δ11.75(s,2H),8.08(s,1H),4.56(d,J=13.4Hz,1H),4.23(s,2H),4.05(d,J=8.9Hz,1H),3.87–3.80(m,1H),3.75(d,J=14.6Hz,1H),3.70–3.63(m,1H),3.26(dd,J=14.6,10.3Hz,1H),2.86(d,J=14.3Hz,1H),1.20(t,J=7.1Hz,2H),1.18(d,J=6.2Hz,3H),0.97(t,J=7.1Hz,2H),0.92(d,J=6.3Hz,3H); 13C NMR(126MHz,DMSO)δ171.22,168.39,166.83,153.87,153.32,151.45,150.24,127.89,121.77,115.55,104.11,76.30,72.83,72.72,66.01,62.47,56.45,52.58,50.54,38.31,18.52,18.16,10.30,10.24;MS(EI)m/z:[M +,506]。 Starting from intermediate I-16-5 (20 mg, 0.050 mmol) and barbituric acid (8 mg, 0.060 mmol), according to the synthesis of compound 1, 15 mg of pale yellow solid, yield 59%, 1 H NMR (400MHz, DMSO) δ11.75 (s, 2H), 8.08 (s, 1H), 4.56 (d, J = 13.4 Hz, 1H), 4.23 (s, 2H), 4.05 (d, J = 8.9 Hz, 1H) ), 3.87–3.80 (m, 1H), 3.75 (d, J = 14.6 Hz, 1H), 3.70–3.63 (m, 1H), 3.26 (dd, J = 14.6, 10.3 Hz, 1H), 2.86 (d, J = 14.3 Hz, 1H), 1.20 (t, J = 7.1 Hz, 2H), 1.18 (d, J = 6.2 Hz, 3H), 0.97 (t, J = 7.1 Hz, 2H), 0.92 (d, J = 6.3 Hz, 3H); 13 C NMR (126MHz, DMSO) δ 171.22, 168.39, 166.83, 153.87, 153.32, 151.45, 150.24, 127.89, 121.77, 115.55, 104.11, 76.30, 72.83, 72.72, 66.01, 62.47, 56.45, 52.58, 50.54, 38.31, 18.52, 18.16, 10.30, 10.24; MS (EI) m/z: [M + , 506].
实施例6、(2R,4S,4aS)-2,4-二甲基-8-((S)-4-甲基-2-氧代噁唑-3-基)-2',4',6'-三氧代-1,1',2,3',4,4a,4',6'-八氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-11-甲腈(化合物6)Example 6, (2R, 4S, 4aS)-2,4-dimethyl-8-((S)-4-methyl-2-oxooxazol-3-yl)-2',4', 6'-Trioxo-1,1',2,3',4,4a,4',6'-octahydro-2'H,6H-spiro[isoxazole [4,5-g][1 , 4]oxazine [4,3-a]quinoline-5,5'-pyrimidine]-11-carbonitrile (compound 6)
(a)(S)-3-氰基-5-(1,3-二氧戊环-2-基)-2,4-二氟-N'-羟基-N-(1-羟基丙烷-2-基)苯甲脒(I-12-6)(a) (S)-3-cyano-5-(1,3-dioxolan-2-yl)-2,4-difluoro-N'-hydroxy-N-(1-hydroxypropane-2 -based) benzamidine (I-12-6)
Figure PCTCN2018092208-appb-000038
Figure PCTCN2018092208-appb-000038
以中间体I-10(300mg,1.181mmol)、N-氯代丁二酰亚胺(189mg,1.417mmol)和(S)-2-氨基-1-丙醇(0.200ml,2.598mmol)为原料,按照中间体I-12-5的合成方法,制得棕黄色油状物299mg,收率77%, 1H NMR(400MHz,CD 3OD)δ10.03(s,1H),7.86(t,J=7.9Hz,1H),6.05(s,1H),5.79(d,J=10.5Hz,1H),4.67(t,J=5.4Hz,1H),4.09–3.99(m,5H),3.23(t,J=5.4Hz,2H),0.99(d,J=6.5Hz,3H)。 Starting from intermediate I-10 (300 mg, 1.181 mmol), N-chlorosuccinimide (189 mg, 1.417 mmol) and (S)-2-amino-1-propanol (0.200 ml, 2.598 mmol) According to the synthesis method of the intermediate I-12-5, 299 mg (yield: 77%) of pale brown oil, 1 H NMR (400 MHz, CD 3 OD) δ 10.03 (s, 1H), 7.86 (t, J) = 7.9 Hz, 1H), 6.05 (s, 1H), 5.79 (d, J = 10.5 Hz, 1H), 4.67 (t, J = 5.4 Hz, 1H), 4.09 - 3.99 (m, 5H), 3.23 (t , J = 5.4 Hz, 2H), 0.99 (d, J = 6.5 Hz, 3H).
(b)(S)-6-氟-5-甲酰基-3-((1-羟基丙烷-2-基)氨基)苯并[d]异噁唑-7-甲腈(I-14-6)(b) (S)-6-Fluoro-5-formyl-3-((1-hydroxypropan-2-yl)amino)benzo[d]isoxazole-7-carbonitrile (I-14-6 )
Figure PCTCN2018092208-appb-000039
Figure PCTCN2018092208-appb-000039
以中间体I-12-6(200mg,0.612mmol)、6M盐酸(5ml)和碳酸氢钠饱和水溶液为原料,按照中间体I-14-5的合成方法,制得白色固体70mg,收率44%, 1H NMR(400MHz,CDCl 3)δ10.35(s,1H),8.40(d,J=6.2Hz,1H),5.00(d,J=6.4Hz,1H),3.97(dd,J=21.6,7.8Hz,2H),3.76–3.70(m,1H),1.92(t,J=4.6Hz,1H),1.40(d,J=6.6Hz,3H)。 The intermediate I-12-6 (200 mg, 0.612 mmol), 6M hydrochloric acid (5 ml) and a saturated aqueous solution of sodium hydrogencarbonate were used as a starting material to obtain a white solid 70 mg (yield: 44). %, 1 H NMR (400MHz, CDCl 3 ) δ 10.35 (s, 1H), 8.40 (d, J = 6.2 Hz, 1H), 5.00 (d, J = 6.4 Hz, 1H), 3.97 (dd, J = 21.6, 7.8 Hz, 2H), 3.76 - 3.70 (m, 1H), 1.92 (t, J = 4.6 Hz, 1H), 1.40 (d, J = 6.6 Hz, 3H).
(c)6-((2R,6R)-2,6-二甲基吗啉)-5-甲酰基-3-(((S)-1-羟基丙烷-2-基)氨基)苯并[d]异噁唑-7-甲腈(I-15-6)(c) 6-((2R,6R)-2,6-dimethylmorpholine)-5-formyl-3-(((S)-1-hydroxypropan-2-yl)amino)benzo[ d] Isoxazole-7-carbonitrile (I-15-6)
Figure PCTCN2018092208-appb-000040
Figure PCTCN2018092208-appb-000040
以中间体I-14-6(70mg,0.266mmol)、2R,6R-二甲基吗啉(0.037ml,0.277mmol)、N,N-二异丙基乙基胺(0.057ml,0.333mmol)为原料,按照中间体I-15-5的合成方法,制得黄色固体67mg,收率70%, 1H NMR(400MHz,CDCl 3)δ10.18(s,1H),8.18(s,1H),4.90(d,J=7.2Hz,1H),4.35–4.29(m,2H),4.00–3.91(m,2H),3.75(dd,J=12.9,3.3Hz,2H),3.72–3.68(m,1H),3.27(dd,J=12.4,5.8Hz,2H),1.37(d,J=6.6Hz,3H),1.34(d,J=6.5Hz,6H)。 Intermediate I-14-6 (70 mg, 0.266 mmol), 2R, 6R-dimethylmorpholine (0.037 ml, 0.277 mmol), N,N-diisopropylethylamine (0.057 ml, 0.333 mmol) As a raw material, according to the synthesis method of Intermediate I-15-5, 67 mg of a yellow solid was obtained, yield 70%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.18 (s, 1H), 8.18 (s, 1H) , 4.90 (d, J = 7.2 Hz, 1H), 4.35 - 4.29 (m, 2H), 4.00 - 3.91 (m, 2H), 3.75 (dd, J = 12.9, 3.3 Hz, 2H), 3.72 - 3.68 (m , 1H), 3.27 (dd, J = 12.4, 5.8 Hz, 2H), 1.37 (d, J = 6.6 Hz, 3H), 1.34 (d, J = 6.5 Hz, 6H).
(d)6-((2R,6R)-2,6-二甲基吗啉)-5-甲酰基-3-((S)-4-甲基-2-氧代噁唑-3-基)苯并[d]异噁唑-7-甲腈(I-16-6)(d) 6-((2R,6R)-2,6-Dimethylmorpholine)-5-formyl-3-((S)-4-methyl-2-oxooxazol-3-yl Benzo[d]isoxazole-7-carbonitrile (I-16-6)
Figure PCTCN2018092208-appb-000041
Figure PCTCN2018092208-appb-000041
以中间体I-15-6(67mg,0.187mmol)、羰基二咪唑(61mg,0.374mmol)和4-二甲氨基吡啶(23mg,0.187mmol)为原料,按照中间体I-4-1的合成方法,制得黄色固体50mg,收率70%, 1H NMR(400MHz,CDCl 3)δ10.06(s,1H),9.03(s,1H),4.79–4.73(m,2H),4.39–4.33(m,2H),4.28–4.23(m,1H),3.79(dd,J=12.6,3.1Hz,2H),3.22(dd,J=12.7,5.9Hz,2H),1.61(d,J=5.9Hz,3H),1.31(d,J=6.5Hz,6H)。 Synthesis of Intermediate I-4-1 with Intermediate I-15-6 (67 mg, 0.187 mmol), carbonyldiimidazole (61 mg, 0.374 mmol) and 4-dimethylaminopyridine (23 mg, 0.187 mmol). Method, 50 mg of yellow solid was obtained, yield 70%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.06 (s, 1H), 9.03 (s, 1H), 4.79 - 4.73 (m, 2H), 4.39 - 4.33 (m, 2H), 4.28 - 4.23 (m, 1H), 3.79 (dd, J = 12.6, 3.1 Hz, 2H), 3.22 (dd, J = 12.7, 5.9 Hz, 2H), 1.61 (d, J = 5.9) Hz, 3H), 1.31 (d, J = 6.5 Hz, 6H).
(e)(2R,4S,4aS)-2,4-二甲基-8-((S)-4-甲基-2-氧代噁唑-3-基)-2',4',6'-三氧代-1,1',2,3',4,4a,4',6'-八氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-11- 甲腈(化合物6)(e) (2R, 4S, 4aS)-2,4-dimethyl-8-((S)-4-methyl-2-oxooxazol-3-yl)-2',4',6 '-Trioxo-1,1',2,3',4,4a,4',6'-octahydro-2'H,6H-spiro[isoxazole [4,5-g][1, 4] Oxazine [4,3-a]quinoline-5,5'-pyrimidine]-11-carbonitrile (Compound 6)
Figure PCTCN2018092208-appb-000042
Figure PCTCN2018092208-appb-000042
以中间体I-16-6(50mg,0.130mmol)和巴比妥酸(18mg,0.143mmol)为原料,按照化合物1的合成方法,制得淡黄色固体35mg,收率54%, 1H NMR(500MHz,DMSO)δ11.88(s,1H),11.57(s,1H),7.93(s,1H),4.72–4.61(m,2H),4.56(dd,J=14.3,1.3Hz,1H),4.19(dd,J=8.0,5.3Hz,1H),4.05(d,J=8.9Hz,1H),3.87–3.79(m,1H),3.73(d,J=14.6Hz,1H),3.65(dq,J=12.8,6.3Hz,1H),3.27(dd,J=14.6,10.4Hz,1H),2.89(d,J=14.5Hz,1H),1.42(d,J=6.0Hz,3H),1.18(d,J=6.2Hz,3H),0.92(d,J=6.3Hz,3H); 13C NMR(126MHz,DMSO)δ170.97,168.19,166.56,154.63,152.77,151.48,149.88,127.72,121.66,115.51,104.94,76.44,72.86,72.69,70.80,65.94,56.49,52.93,52.65,38.29,18.54,18.15,17.74;MS(ESI)m/z:[(M-1) -,493.2]。 Using Intermediate I-16-6 (50 mg, 0.130 mmol) and barbituric acid (18 mg, 0.143 mmol) as a starting material, according to the synthesis of compound 1 to give a pale yellow solid, 35 mg, yield 54%, 1 H NMR (500MHz, DMSO) δ 11.88 (s, 1H), 11.57 (s, 1H), 7.93 (s, 1H), 4.72 - 4.61 (m, 2H), 4.56 (dd, J = 14.3, 1.3 Hz, 1H) , 4.19 (dd, J = 8.0, 5.3 Hz, 1H), 4.05 (d, J = 8.9 Hz, 1H), 3.87 - 3.79 (m, 1H), 3.73 (d, J = 14.6 Hz, 1H), 3.65 ( Dq, J = 12.8, 6.3 Hz, 1H), 3.27 (dd, J = 14.6, 10.4 Hz, 1H), 2.89 (d, J = 14.5 Hz, 1H), 1.42 (d, J = 6.0 Hz, 3H), 1.18 (d, J = 6.2 Hz, 3H), 0.92 (d, J = 6.3 Hz, 3H); 13 C NMR (126 MHz, DMSO) δ 170.97, 168.19, 166.56, 154.63, 152.77, 151.48, 149.88, 127.72, 121.66, 115.51, 104.94, 76.44, 72.86, 72.69, 70.80, 65.94, 56.49, 52.93, 52.65, 38.29, 18.54, 18.15, 17.74; MS (ESI) m/z: [(M-1) - , 493.2].
实施例7、(2R,4S,4aS)-2,4-二甲基-8-((S)-5-甲基-2-氧代噻唑-3-基)-2',4',6'-三氧代-1,1',2,3',4,4a,4',6'-八氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-11-腈(化合物7)Example 7, (2R, 4S, 4aS)-2,4-dimethyl-8-((S)-5-methyl-2-oxothiazol-3-yl)-2',4',6 '-Trioxo-1,1',2,3',4,4a,4',6'-octahydro-2'H,6H-spiro[isoxazole [4,5-g][1, 4] Oxazine [4,3-a]quinoline-5,5'-pyrimidine]-11-carbonitrile (Compound 7)
(a)(R)-3-氰基-5-(1,3-二氧戊环-2-基)-2,4-二氟-N'-羟基-N-(2-羟丙基)苯甲脒(I-12-7)(a) (R)-3-cyano-5-(1,3-dioxolan-2-yl)-2,4-difluoro-N'-hydroxy-N-(2-hydroxypropyl) Benzoquinone (I-12-7)
Figure PCTCN2018092208-appb-000043
Figure PCTCN2018092208-appb-000043
以中间体I-10(800mg,3.149mmol)、N-氯代丁二酰亚胺(463mg,3.464mmol)和(R)-(-)-1-氨基-2-丙醇(0.545ml,6.928mmol)为原料,按照中间体I-12-5的合成方法,制得淡黄色粉末456mg,收率44%, 1H NMR(400MHz,CDCl 3)δ7.89(t,J=7.7Hz,1H),6.05(s,1H),5.77(s,1H),4.18–4.05(m,4H),3.82–3.75(m,1H),2.97(d,J=13.1Hz,1H),2.87–2.80(m,1H),1.11(d,J=6.3Hz,3H)。 Intermediate I-10 (800 mg, 3.149 mmol), N-chlorosuccinimide (463 mg, 3.464 mmol) and (R)-(-)-1-amino-2-propanol (0.545 ml, 6.928) Methyl) 456 mg, yield 44%, 1 H NMR (400 MHz, CDCl 3 ) δ 7.89 (t, J = 7.7 Hz, 1H). ), 6.05 (s, 1H), 5.77 (s, 1H), 4.18 - 4.05 (m, 4H), 3.82 - 3.75 (m, 1H), 2.97 (d, J = 13.1 Hz, 1H), 2.87 - 2.80 ( m, 1H), 1.11 (d, J = 6.3 Hz, 3H).
(b)(R)-6-氟-5-甲酰基-3-((2-羟丙基)氨基)苯并[d]异噁唑-7-腈(I-14-7)(b) (R)-6-fluoro-5-formyl-3-((2-hydroxypropyl)amino)benzo[d]isoxazole-7-carbonitrile (I-14-7)
Figure PCTCN2018092208-appb-000044
Figure PCTCN2018092208-appb-000044
以中间体I-12-7(443mg,1.354mmol)、6M盐酸(8ml)和饱和碳酸氢钠水溶 液为原料,按照中间体I-14-5的合成方法,得淡黄色粉末150mg,收率42%, 1H NMR(400MHz,DMSO)δ10.17(s,1H),8.90(d,J=6.7Hz,1H),7.71(t,J=5.7Hz,1H),4.86(d,J=4.8Hz,1H),3.97–3.87(m,1H),3.17(td,J=6.0,2.2Hz,2H),1.13(d,J=6.2Hz,3H)。 The intermediate I-12-7 (443 mg, 1.354 mmol), 6M hydrochloric acid (8 ml) and a saturated aqueous solution of sodium hydrogencarbonate were used as a starting material. %, 1 H NMR (400 MHz, DMSO) δ 10.17 (s, 1H), 8.90 (d, J = 6.7 Hz, 1H), 7.71 (t, J = 5.7 Hz, 1H), 4.86 (d, J = 4.8) Hz, 1H), 3.97 - 3.87 (m, 1H), 3.17 (td, J = 6.0, 2.2 Hz, 2H), 1.13 (d, J = 6.2 Hz, 3H).
(c)6-((2R,6R)-2,6-二甲基吗啉)-5-甲酰基-3-(((R)-2-羟丙基)氨基)苯并[d]异噁唑-7-腈(I-15-7)(c) 6-((2R,6R)-2,6-dimethylmorpholine)-5-formyl-3-(((R)-2-hydroxypropyl)amino)benzo[d]iso Oxazole-7-carbonitrile (I-15-7)
Figure PCTCN2018092208-appb-000045
Figure PCTCN2018092208-appb-000045
以中间体I-14-7(150mg,0.570mmol)、2R,6R-二甲基吗啉(0.090ml,0.684mmol)和N,N-二异丙基乙基胺(0.200ml,1.140mmol)为原料,按照中间体I-15-5的合成方法,制得淡黄色固体162mg,收率79%, 1H NMR(400MHz,CDCl 3)δ10.18(s,1H),8.22(s,1H),5.28–5.24(m,1H),4.36–4.28(m,2H),4.24–4.16(m,1H),3.75(dd,J=12.3,3.0Hz,2H),3.60–3.54(m,1H),3.32–3.22(m,3H),1.33(d,J=6.4Hz,9H)。 Intermediates I-14-7 (150 mg, 0.570 mmol), 2R, 6R-dimethylmorpholine (0.090 ml, 0.684 mmol) and N,N-diisopropylethylamine (0.200 ml, 1.140 mmol) As a raw material, 162 mg (yield: 79%) of pale yellow solid (yield: 79%, 1 H NMR (400 MHz, CDCl 3 ) δ 10.18 (s, 1H), 8.22 (s, 1H). ), 5.28–5.24 (m, 1H), 4.36–4.28 (m, 2H), 4.24–4.16 (m, 1H), 3.75 (dd, J = 12.3, 3.0 Hz, 2H), 3.60–3.54 (m, 1H) ), 3.32–3.22 (m, 3H), 1.33 (d, J = 6.4 Hz, 9H).
(d)6-((2R,6R)-2,6-二甲基吗啉)-5-甲酰基-3-((R)-5-甲基-2-氧代噁唑-3-基)苯并[d]异噁唑-7-甲腈(I-16-7)(d) 6-((2R,6R)-2,6-Dimethylmorpholine)-5-formyl-3-((R)-5-methyl-2-oxooxazol-3-yl Benzo[d]isoxazole-7-carbonitrile (I-16-7)
Figure PCTCN2018092208-appb-000046
Figure PCTCN2018092208-appb-000046
以中间体I-15-7(120mg,0.335mmol)、羰基二咪唑(109mg,0.670mmol)和4-二甲氨基吡啶(41mg,0.335mmol)为原料,按照中间体I-4-1的合成方法,制得黄色固体82mg,收率64%, 1H NMR(400MHz,CDCl 3)δ10.05(s,1H),9.11(s,1H),5.03(dd,J=14.0,7.5Hz,1H),4.39–4.29(m,3H),3.84(dd,J=8.5,5.8Hz,1H),3.80(dd,J=12.3,2.6Hz,2H),3.23(dd,J=12.7,5.9Hz,2H),1.64(d,J=6.3Hz,3H),1.32(d,J=6.4Hz,6H)。 The synthesis of intermediate I-4-1 was carried out using intermediate I-15-7 (120 mg, 0.335 mmol), carbonyldiimidazole (109 mg, 0.670 mmol) and 4-dimethylaminopyridine (41 mg, 0.335 mmol). Method, 82 mg of a yellow solid was obtained in a yield of 64%, 1 H NMR (400 MHz, CDCl 3 ) δ10.05 (s, 1H), 9.11 (s, 1H), 5.03 (dd, J = 14.0, 7.5 Hz, 1H) ), 4.39 - 4.29 (m, 3H), 3.84 (dd, J = 8.5, 5.8 Hz, 1H), 3.80 (dd, J = 12.3, 2.6 Hz, 2H), 3.23 (dd, J = 12.7, 5.9 Hz, 2H), 1.64 (d, J = 6.3 Hz, 3H), 1.32 (d, J = 6.4 Hz, 6H).
(e)(2R,4S,4aS)-2,4-二甲基-8-((R)-5-甲基-2-氧代噁唑-3-基)-2',4',6'-三氧代-1,1',2,3',4,4a,4',6'-八氢-2'H,6H-螺[异噁唑[4,5-g][1,4]恶嗪[4,3-a]喹啉-5,5'-嘧啶]-11-甲腈(化合物7)(e) (2R, 4S, 4aS)-2,4-dimethyl-8-((R)-5-methyl-2-oxooxazol-3-yl)-2',4',6 '-Trioxo-1,1',2,3',4,4a,4',6'-octahydro-2'H,6H-spiro[isoxazole [4,5-g][1, 4] Oxazine [4,3-a]quinoline-5,5'-pyrimidine]-11-carbonitrile (Compound 7)
Figure PCTCN2018092208-appb-000047
Figure PCTCN2018092208-appb-000047
以中间体I-16-7(82mg,0.213mmol)和巴比妥酸(30mg,0.235mmol)为原料,按照化合物1的合成方法,制得白色固体34mg,收率32%, 1H NMR(400MHz,DMSO)δ11.77(s,1H),11.64(s,1H),8.08(s,1H),5.02–4.92(m,1H),4.60–4.54(m,1H),4.22(dd,J=9.5,8.3Hz,1H),4.04(d,J=8.9Hz,1H),3.88–3.81(m,1H),3.77(d,J=14.7Hz,1H),3.74–3.62(m,2H),3.27(dd,J=14.7,10.3Hz,1H),2.85(dd,J=14.5,1.2Hz,1H),1.46(d,J=6.2Hz,3H),1.19(d,J=6.3Hz,3H),0.93(d,J=6.4Hz,3H); 13C NMR(151MHz,DMSO)δ170.98,168.12,166.72,154.18,153.44,151.29,149.90,128.07,121.48,115.52,104.19,76.22,73.07,72.78,72.64,65.95,56.39,52.60,51.24,38.22,20.32,18.48,18.10;MS(ESI)m/z:[(M-1) -,493.2]。 Using Intermediate I-16-7 (82 mg, 0.213 mmol) and barbituric acid (30 mg, 0.235 mmol) as a starting material, according to the synthesis of Compound 1, a white solid, 34 mg, yield 32%, 1 H NMR ( 400 MHz, DMSO) δ 11.77 (s, 1H), 11.64 (s, 1H), 8.08 (s, 1H), 5.02 - 4.92 (m, 1H), 4.60 - 4.54 (m, 1H), 4.22 (dd, J = 9.5, 8.3 Hz, 1H), 4.04 (d, J = 8.9 Hz, 1H), 3.88 - 3.81 (m, 1H), 3.77 (d, J = 14.7 Hz, 1H), 3.74 - 3.62 (m, 2H) , 3.27 (dd, J = 14.7, 10.3 Hz, 1H), 2.85 (dd, J = 14.5, 1.2 Hz, 1H), 1.46 (d, J = 6.2 Hz, 3H), 1.19 (d, J = 6.3 Hz, 3H), 0.93 (d, J = 6.4 Hz, 3H); 13 C NMR (151 MHz, DMSO) δ 170.98, 168.12, 166.72, 154.18, 153.44, 151.29, 149.90, 128.07, 121.48, 115.52, 104.19, 76.22, 73.07, 72.78 , 72.64, 65.95, 56.39, 52.60, 51.24, 38.22, 20.32, 18.48, 18.10; MS (ESI) m/z: [(M-1) - , 493.2].
实施例8 体外抗菌活性测定Example 8 Determination of in vitro antibacterial activity
1.实验菌株Experimental strain
体外抗菌活性实验选用临床分离的3株甲氧西林耐药金黄色葡萄球菌MRSA,3株甲氧西林敏感的金黄色葡萄球菌MSSA,3株甲氧西林耐药表皮葡萄球菌MRSE,3株甲氧西林敏感表皮葡萄球菌MSSE,3株青霉素耐药肺炎链球菌PRSP,3株化脓性链球菌。In vitro antibacterial activity experiments were performed with 3 strains of methicillin-resistant Staphylococcus aureus MRSA, 3 strains of methicillin-sensitive Staphylococcus aureus MSSA, 3 strains of methicillin-resistant Staphylococcus epidermidis MRSE, and 3 strains of methoxy Xilin sensitive Staphylococcus epidermidis MSSE, 3 strains of penicillin-resistant Streptococcus pneumoniae PRSP, 3 strains of Streptococcus pyogenes.
以上菌株均为2015年12月在四川地区、北京地区收集的临床分离致病菌。在收集单位经VITEK-60自动微生物鉴定仪鉴定再经常规方法重新鉴定。每株细菌在实验前经琼脂平板划单菌落分纯,37℃隔夜新鲜培养的菌体适当稀释用于实验。The above strains were clinical isolates collected in Sichuan and Beijing in December 2015. The collection unit was identified by VITEK-60 automatic microbiological identification instrument and then re-identified by conventional methods. Each strain of bacteria was divided into single colonies by agar plates before the experiment, and the freshly cultured cells at 37 ° C overnight were appropriately diluted for the experiment.
质控菌株:金黄色葡萄球菌ATCC25923。购自中华人民共和国卫生部临床检测中心。Quality control strain: Staphylococcus aureus ATCC25923. Purchased from the Clinical Testing Center of the Ministry of Health of the People's Republic of China
2.培养基及培养条件2. Medium and culture conditions
培养基:MH(Mueller-Hinton,水解酪蛋白)肉汤培养基(OXOID);Medium: MH (Mueller-Hinton, hydrolyzed casein) broth medium (OXOID);
培养条件:35-37℃孵育16-18h。Culture conditions: incubation at 35-37 ° C for 16-18 h.
3.试验方法3. Test method
采用美国临床和实验室标准协会(Clinical and Laboratory Standards Institute,CLSI)抗菌药物敏感性试验操作规程【(Performance Standards for Antimicrobial Susceptibility Testing;Twenty-Third Informational Supplement)M02-A11、M07-A9和M11-A8,2013】推荐的微量肉汤稀释法测定各受试样品对所试菌株的最低抑菌浓度(Minimal Inhibitory Concentration,MIC)值。Adopted the Clinical and Laboratory Standards Institute (CLSI) Antimicrobial Susceptibility Testing (Twenty-Third Informational Supplement) M02-A11, M07-A9 and M11-A8 , 2013] The recommended micro-broth dilution method is used to determine the minimum inhibitory concentration (MIC) of each test sample against the tested strain.
用MH肉汤培养基分别将各受试样品倍比稀释至不同浓度后,分别吸取100μl各不同浓度的受试样品溶液于灭菌的96孔聚苯乙烯板中。第1至第12孔加药液,各孔 中药物终浓度分别为64、32、16、8、4、2、1、0.5、0.25、0.125、0.06、0.03mg/L。另设不加药和菌,作为空白对照。加菌不加药作为细菌生长对照。After diluting each test sample to different concentrations in MH broth medium, 100 μl of each test solution of different concentrations was separately aspirated into a sterilized 96-well polystyrene plate. The first to twelfth wells were filled with a drug solution, and the final concentrations of the drugs in each well were 64, 32, 16, 8, 4, 2, 1, 0.5, 0.25, 0.125, 0.06, and 0.03 mg/L, respectively. There is no additional medicine and bacteria as a blank control. Adding bacteria without dosing as a bacterial growth control.
用生理盐水将试验菌液调至相当于0.5麦氏比浊标准的菌悬液,经MH肉汤1∶100稀释后,加入药液中,使菌液最终浓度约为10 4CFU/ml;再分别吸取100μl菌液加入上述孔中(每孔中总体积为200μl),密封后置于35-37℃培养箱内培养18-20h,判断结果。用酶标仪测定OD 600值,以在小孔内完全抑制细菌生长的最低药物浓度为其最低抑菌浓度(Minimal Inhibitory Concentration,MIC)。测试肺炎链球菌时,需用哥伦比亚肉汤培养基中加入终浓度为5%的无菌脱纤维羊血,5%CO 2,35-37℃培养箱内培养18-20h。 The test bacterial liquid was adjusted to a bacterial suspension equivalent to 0.5 McMulster standard with physiological saline, diluted with MH broth 1:100, and added to the chemical solution to make the final concentration of the bacterial liquid about 10 4 CFU/ml; Then, 100 μl of the bacterial solution was separately added to the above wells (the total volume in each well was 200 μl), sealed, and placed in a 35-37 ° C incubator for 18-20 hours to judge the result. The OD 600 value was measured by a microplate reader to minimize the minimum drug concentration of the bacteria in the wells to be the minimum inhibitory concentration (MIC). When testing Streptococcus pneumoniae, it is necessary to add a final concentration of 5% sterile defibrinated sheep blood to a broth of Colombian broth, 5% CO 2 , and culture in a 35-37 ° C incubator for 18-20 h.
体外抗菌活性结果如表1所示。由表1可知,本发明化合物具有很好的体外抗菌活性,化合物1对MRSA、MSSA、MRSE、MSSE、PRSP和Spy的体外抗菌活性都要大大优于对照药AZD0914,是AZD0914的8倍左右,而化合物2、3和4的体外抗菌活性与对照药相当。The results of in vitro antibacterial activity are shown in Table 1. It can be seen from Table 1 that the compound of the present invention has good in vitro antibacterial activity, and the in vitro antibacterial activity of Compound 1 on MRSA, MSSA, MRSE, MSSE, PRSP and Spy is much better than that of the control drug AZD0914, which is about 8 times that of AZD0914. The in vitro antibacterial activity of Compounds 2, 3 and 4 was comparable to that of the control drug.
表1本发明部分化合物的体外抗菌活性实验结果Table 1 Experimental results of in vitro antibacterial activity of some compounds of the present invention
Figure PCTCN2018092208-appb-000048
Figure PCTCN2018092208-appb-000048
aMethicillin resistant S.aureus. bMethicillin sensitive S.aureus. cMethicillin resistant S.epidermidis. dMethicillin sensitive S.epidermidis. ePenicillin resistant S.pneumoniae. fS.pyogenes. a Methicillin resistant S.aureus. b Methicillin sensitive S.aureus. c Methicillin resistant S.epidermidis. d Methicillin sensitive S.epidermidis. e Penicillin resistant S.pneumoniae. f S.pyogenes.
实施例9 化合物溶解度测定Example 9 Determination of Compound Solubility
1.空白基质Blank matrix
人工胃液(不含胃蛋白酶)的配制:取20mg氯化钠,加入70μL的浓盐酸和足够的水定容到10mL。人工胃液的pH约为1.2。Preparation of artificial gastric juice (without pepsin): Take 20 mg of sodium chloride, add 70 μL of concentrated hydrochloric acid and sufficient water to make up to 10 mL. The pH of the artificial gastric juice is about 1.2.
人工肠液(不含胰酶)的配制:取68mg磷酸二氢钾溶于2500μL水中,加入770μL0.2N氢氧化钠溶液和5mL水,用0.2N的氢氧化钠或0.2N的盐酸调节pH至6.8±0.1,加水定容至10mL。Preparation of artificial intestinal juice (without trypsin): Take 68mg of potassium dihydrogen phosphate dissolved in 2500μL of water, add 770μL of 0.2N sodium hydroxide solution and 5mL of water, adjust the pH to 6.8 with 0.2N sodium hydroxide or 0.2N hydrochloric acid. ±0.1, add water to a volume of 10mL.
去离子水。Deionized water.
2.样品制备2. Sample preparation
称取约3mg化合物,分别加入500μL空白基质,于37℃环境下摇24h。孵育结束后的样品,离心30min,上清液转移至新的EP管中,继续离心30min。About 3 mg of the compound was weighed and added to 500 μL of a blank substrate, and shaken at 37 ° C for 24 hours. After the incubation, the sample was centrifuged for 30 min, and the supernatant was transferred to a new EP tube and centrifuged for 30 min.
3.色谱条件:3. Chromatographic conditions:
分析柱:Acquity BEH C18(1.5μm;2.1x 50mm,Waters)Analytical column: Acquity BEH C18 (1.5 μm; 2.1 x 50 mm, Waters)
流动相:0.1%FA+5mM NH4AC in Water:0.1%FA in ACNMobile phase: 0.1% FA + 5 mM NH4AC in Water: 0.1% FA in ACN
4.标准曲线制作4. Standard curve production
用乙腈/水(1/1,v/v)配制标准曲线样品(12.5,25,50,100,200,400,2000,10000nM),待测物峰面积由Waters MassLynx(版本V4.1)软件定量。使用外标法计算待测物浓度。以待测物的峰面积(y)为纵坐标,待测物的浓度(x)为横坐标,用加权(1/x2)最小二乘法进行线性回归,得到标准曲线方程y=ax+b,a是斜率,b是截距。Standard curve samples (12.5, 25, 50, 100, 200, 400, 2000, 10000 nM) were prepared with acetonitrile/water (1/1, v/v) and the peak area of the analyte was quantified by Waters MassLynx (version V4.1) software. The concentration of the analyte is calculated using an external standard method. Taking the peak area (y) of the object to be tested as the ordinate, the concentration (x) of the object to be tested is the abscissa, and the linear regression is performed by the weighted (1/x2) least squares method to obtain the standard curve equation y=ax+b. a is the slope and b is the intercept.
5.样品分析5. Sample analysis
每种基质的样品分别各取50μL上清液,加入450μL乙腈/水(1/1,v/v),涡流混匀后,再取20μL加入180μL乙腈/水(1/1,v/v),涡流混匀后进行LC-MS/MS分析,将所得峰面积带入标准曲线方程得到每种基质的饱和溶解度。每个样品于每种基质分别各取四次进行测量,所得结果取平均值得到最终化合物的不同基质饱和溶解度。Take 50 μL of the supernatant from each sample of the matrix, add 450 μL of acetonitrile/water (1/1, v/v), mix by vortexing, and then add 20 μL to 180 μL of acetonitrile/water (1/1, v/v). After vortex mixing, LC-MS/MS analysis was carried out, and the obtained peak area was brought into the standard curve equation to obtain the saturation solubility of each matrix. Each sample was measured four times for each matrix, and the results were averaged to obtain different matrix saturation solubility of the final compound.
实验结果见表2。溶解度是评价药物分子成药性的一个重要性质,它影响药物分子在动物和人体内的吸收、分布、药物暴露量、口服生物利用度等。目前绝大多数药物分子最终不能成功上市,溶解度差是一个关键因素。从表2可以看出,本发明化合物在不同基质中的溶解度相对于阳性对照药AZD0914有很大提高,生物药学性质大幅改善,成药性更优。The experimental results are shown in Table 2. Solubility is an important property in evaluating the drug-forming properties of drug molecules. It affects the absorption, distribution, drug exposure, oral bioavailability of drug molecules in animals and humans. At present, most drug molecules cannot be successfully marketed, and poor solubility is a key factor. It can be seen from Table 2 that the solubility of the compound of the present invention in different matrices is greatly improved relative to the positive control drug AZD0914, the biopharmaceutical properties are greatly improved, and the drug-forming property is superior.
表2不同基质下化合物的溶解度数据Table 2 Solubility data of compounds in different matrices
Figure PCTCN2018092208-appb-000049
Figure PCTCN2018092208-appb-000049
实施例10 小鼠体内药代动力学试验Example 10 Pharmacokinetic test in mice
健康雌性CD-1小鼠,随机分成两组,每组3只,口服给予被试化合物,具体安排见下表3。Healthy female CD-1 mice were randomly divided into two groups, 3 in each group, and the test compounds were orally administered. The specific arrangement is shown in Table 3 below.
表3给药方案Table 3 dosage regimen
Figure PCTCN2018092208-appb-000050
Figure PCTCN2018092208-appb-000050
每只动物每次通过眼眶取0.030mL血液,EDTAK2抗凝,采集时间点为:PO组:给予受试物后15min,30min,1h,2h,4h,6h,8h和24h。血液样本采集后置于冰上,并于30分钟之内离心分离血浆(离心条件:5000转/分钟,10分钟,室温)。分析前存放于–80℃。实验结果见表4。Each animal took 0.030 mL of blood through the eye, and EDTAK2 was anticoagulated. The time of collection was: PO group: 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after administration of the test substance. Blood samples were collected and placed on ice, and plasma was separated by centrifugation within 30 minutes (centrifugation conditions: 5000 rpm, 10 minutes, room temperature). Store at –80 °C before analysis. The experimental results are shown in Table 4.
表4小鼠口服药代动力学数据Table 4 oral pharmacokinetic data of mice
Figure PCTCN2018092208-appb-000051
Figure PCTCN2018092208-appb-000051
优良的代谢性质是化合物成药性的关键指标,药代实验证明,本发明化合物具有理想的代谢特征,暴露量和达峰浓度远远优于阳性对照药物AZD0914。The excellent metabolic properties are the key indicators of the drug-forming properties of the compounds. The pharmacokinetic experiments prove that the compounds of the present invention have ideal metabolic characteristics, and the exposure and peak concentrations are far superior to the positive control drug AZD0914.
实施例11Example 11
大鼠体内药代动力学试验Pharmacokinetic test in rats
SD大鼠9只,随机分成三组,每组3只,具体安排见表5。Nine SD rats were randomly divided into three groups of three, each of which is shown in Table 5.
表5给药方案Table 5 dosing schedule
Figure PCTCN2018092208-appb-000052
Figure PCTCN2018092208-appb-000052
每只动物每次通过眼眶取0.100mL血液,EDTAK 2抗凝,采集时间点为:(1)PO组:给予受试物后15min,30min,1h,2h,4h,6h,8h,24h;(2)IV组:给予受试物后5min,15min,30min,1h,2h,4h,6h,8h,24h。血液样本采集后置于冰上,并于30分钟之内离心分离血浆(离心条件:5000转/分钟,10分钟, 4℃)。分析前存放于–80℃。实验结果见表6。 Each animal took 0.100 mL of blood through the eye, and EDTAK 2 was anticoagulated. The time of collection was: (1) PO group: 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, 24 h after administration of the test article; 2) Group IV: 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, 24 h after administration of the test substance. Blood samples were collected and placed on ice, and plasma was centrifuged within 30 minutes (centrifugation conditions: 5000 rpm, 10 minutes, 4 ° C). Store at –80 °C before analysis. The experimental results are shown in Table 6.
表6 SD大鼠口服给药(PO)或静脉注射(IV)化合物1后的主要药动学参数Table 6 Main pharmacokinetic parameters of SD rats after oral administration (PO) or intravenous (IV) compound 1
Figure PCTCN2018092208-appb-000053
Figure PCTCN2018092208-appb-000053
从10mg/kg和100mg/kg口服给药剂量下所得结果对比中显示出二者血浆中的C max与给药量呈现明显的正相关关系,化合物1给药100mg/kg的Cmax是给药10mg/kg的15倍,表明化合物1在100mg/kg剂量下的吸收并未受到明显影响,口服生物利用度也逐渐增大,从22.6%上升至45.8%。 Displays the 10mg / kg and 100mg / kg Comparative results obtained are shown in the case of oral administration dosage and dose both C max plasma showed significant positive correlation between the administration of the compound. 1 100mg / kg was administered 10mg of Cmax 15 times of /kg, indicating that the absorption of Compound 1 at the dose of 100 mg / kg was not significantly affected, oral bioavailability also increased gradually, from 22.6% to 45.8%.
综合化合物1各方面药代动力学参数,可以看出,化合物1在大鼠体内代谢性质优良,值得进一步研究开发。Based on the pharmacokinetic parameters of various aspects of Compound 1, it can be seen that Compound 1 has excellent metabolic properties in rats and is worthy of further research and development.
实施例12Example 12
本发明化合物对MRSA的体内抗菌活性In vivo antibacterial activity of the compounds of the invention against MRSA
1.试验菌株Test strain
根据体外试验结果选取临床分离耐甲氧西林金黄色葡萄球菌MRSA15-3(见下表7)作为体内保护实验的备用受试菌株。According to the results of the in vitro test, clinical isolates of methicillin-resistant Staphylococcus aureus MRSA15-3 (see Table 7 below) were selected as spare test strains for in vivo protection experiments.
表7本发明化合物及对照药对金黄色葡萄球菌MRSA15-3的体外抗菌活性-MIC(mg/L)Table 7 In vitro antibacterial activity of the compound of the present invention and a control drug against Staphylococcus aureus MRSA15-3 - MIC (mg / L)
Figure PCTCN2018092208-appb-000054
Figure PCTCN2018092208-appb-000054
2.试验动物2. Test animals
健康4周龄左右昆明种小鼠,体重为18-22g,雌雄各半,SPF级Healthy Kunming mice around 4 weeks old, weighing 18-22g, male and female, SPF
预计小鼠使用数量:200只Estimated number of mice used: 200
3.体内保护试验方法3. In vivo protection test method
3.1菌液配备3.1 bacterial liquid equipment
将试验用菌于感染前一天,挑取2-3个单菌落接种于2ml MH肉汤,37℃培养6h, 取此菌液0.1ml转种于10ml MH肉汤中,37℃培养18h,该菌液即为原菌液。将该菌液用5%干酵母液进行倍比稀释备用(当日新鲜配制)。One day before the infection of the test bacteria, 2-3 single colonies were picked and inoculated into 2 ml of MH broth, and cultured at 37 ° C for 6 h. 0.1 ml of the bacterial solution was transferred to 10 ml of MH broth and cultured at 37 ° C for 18 h. The bacterial liquid is the original bacterial liquid. The bacterial solution was diluted with 5% dry yeast solution for use (prepared freshly on the day).
3.2最小致死菌量(MLD)试验3.2 Minimum lethal bacteria (MLD) test
取健康昆明种小白鼠,体重18~22克,随机分组,每组5-10只小鼠,雌雄各半,吸取上述不同稀释浓度菌液,分别腹腔注射入小鼠体内,每20g鼠重注射0.5ml,感染后观察7-14天,并记录小鼠死亡数,以引起小鼠100%死亡的最低菌量作为最小致死菌量(MLD),用该菌量作为体内保护试验的感染菌量。Take healthy Kunming mice, weighing 18-22 grams, randomly divided into groups, 5-10 mice in each group, male and female, absorb the above different dilutions of bacterial solution, intraperitoneal injection into mice, re-injection every 20g mouse 0.5ml, observed for 7-14 days after infection, and recorded the number of mouse deaths, the minimum amount of bacteria causing 100% death of mice as the minimum lethal bacteria amount (MLD), using the amount of bacteria as the amount of infectious bacteria in the in vivo protection test .
3.3药液配备及给药途径:3.3 drug solution and route of administration:
药液配备:试验用药用0.5%羧甲基纤维素钠配制并稀释到所需浓度溶液备用Liquid preparation: test with medicinal 0.5% sodium carboxymethyl cellulose and dilute to the desired concentration of solution
给药途径:灌胃给药Route of administration: intragastric administration
给药容量:0.5ml/20g BW(body weight,体重)Dosing capacity: 0.5ml / 20g BW (body weight, body weight)
3.4组别设计3.4 group design
拟设供试品剂量范围为20-2.5mg/kg,精确称取药物,按效价将其换算到有效药物重量,用药均用0.5%羧甲基纤维素钠配制成所需浓度溶液,剂量组间矩为1:0.5。It is proposed to set the dose range of the test sample to 20-2.5mg/kg, accurately weigh the drug, convert it to the effective drug weight according to the potency, and use the 0.5% sodium carboxymethyl cellulose to prepare the solution of the desired concentration. The moment between groups is 1:0.5.
组别设计:AZD0914(20、10、5、2.5g·kg -1)组、化合物1(20、10、5、2.5g·kg -1)组、感染对照组、空白对照组、ABT毒性对照。 Group design: AZD0914 (20, 10, 5, 2.5g·kg -1 ) group, compound 1 (20, 10, 5, 2.5g·kg -1 ) group, infection control group, blank control group, ABT toxicity control .
3.5体内保护实验方法及结果统计3.5 In vivo protection experimental methods and results statistics
将小鼠于试验前18小时停食供水,按体重随机分组,每组8只小鼠,雌雄各半,分别腹腔注射感染试验菌液,每20g鼠重注射0.5ml,感染后0.5h,4h按设计剂量灌胃给药,每20g鼠重灌胃0.5ml;观察并记录小鼠死亡数,连续观察7-14天,根据小鼠死亡数,用孙瑞元等主编DAS1.0软件按Bliss法计算半数有效剂量ED 50及95%可信限。 The mice were stopped from feeding water for 18 hours before the test, and were randomly divided into groups according to their body weight. Each group of 8 mice, male and female, were injected intraperitoneally with the infection test bacterial solution. Each 20g mouse was injected with 0.5ml, 0.5h after infection, 4h. The designed dose was intragastrically administered, and 0.5 ml was perfused into the stomach every 20 g of mice; the number of deaths of the mice was observed and recorded, and the number of deaths was observed for 7-14 days. According to the number of deaths of mice, Sun Ruiyuan and other editors DAS1.0 software were used to calculate half of the Bliss method. Effective dose ED 50 and 95% confidence limit.
对照药AZD0914组在感染前两小时,口服细胞色素P450抑制剂1-氨基苯并三唑(ABT)50mg/kg,12小时后再口服一次50mg/kg ABT,其他化合物不需要口服ABT。The control drug AZD0914 group received oral cytochrome P450 inhibitor 1-aminobenzotriazole (ABT) 50 mg/kg two hours before infection, and 50 mg/kg ABT once orally after 12 hours. Other compounds did not require oral ABT.
感染对照组:只感染细菌不给药,感染细菌后即灌胃等体积的生理盐水;空白对照组:不感染细菌,腹腔注射等体积的生理盐水,灌胃等体积的生理盐水。Infected control group: only the infected bacteria were not administered, and the same volume of physiological saline was administered after the infection of the bacteria; the blank control group: no infection of the bacteria, intraperitoneal injection of an equal volume of physiological saline, and an equal volume of physiological saline.
4.试验结果4. Test results
全身系统感染模型的体内抗菌活性实验结果见表8。The results of in vivo antibacterial activity of the systemic systemic infection model are shown in Table 8.
表8本发明化合物对金黄色葡萄球菌MRSA15-3感染小鼠的体内保护作用(ED 50) Table 8 Protective effect of compounds of the invention against S. aureus infection in mice in vivo MRSA15-3 (ED 50)
Figure PCTCN2018092208-appb-000055
Figure PCTCN2018092208-appb-000055
阳性对照药AZD0914需要与细胞色素P450抑制剂1-氨基苯并三唑(ABT)联用,对金黄色葡萄球菌MRSA15-3感染小鼠的ED 50为11.51mg/kg,而化合物1无需联用ABT,口服给药对金黄色葡萄球菌MRSA15-3感染小鼠的ED 50为3.87mg/kg,明显优于AZD0914。 The positive control drug AZD0914 needs to be combined with the cytochrome P450 inhibitor 1-aminobenzotriazole (ABT), and the ED 50 for S. aureus MRSA15-3 infected mice is 11.51 mg/kg, while Compound 1 does not need to be combined. ABT, ED 50 for oral administration to S. aureus MRSA15-3 infected mice was 3.87 mg/kg, which was significantly better than AZD0914.
综上所述,本发明化合物比阳性对照药AZD0914具有更好的成药性,有望成为更好的抗菌药物。In summary, the compound of the present invention has better drug-forming properties than the positive control drug AZD0914, and is expected to be a better antibacterial drug.
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the the In addition, it should be understood that various modifications and changes may be made by those skilled in the art in the form of the present invention.

Claims (10)

  1. 一种通式(I)所示的化合物,或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐,a compound of the formula (I), or an enantiomer, diastereomer, racemate or mixture thereof, or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2018092208-appb-100001
    Figure PCTCN2018092208-appb-100001
    式中,R 1为氢、卤素或氰基; Wherein R 1 is hydrogen, halogen or cyano;
    R 2、R 3独立为氢原子或C 1-C 3烷基;或者R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环; R 2 and R 3 are independently a hydrogen atom or a C 1 -C 3 alkyl group; or R 2 and R 3 together with a carbon atom to be bonded form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring;
    R 4为氢原子或C 1-C 3烷基; R 4 is a hydrogen atom or a C 1 -C 3 alkyl group;
    各*独立表示消旋、S型或R型;Each * indicates the racemization, S type or R type independently;
    条件是当R 1为卤素时,R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环。 Provided that when R 1 is a halogen, R 2 and R 3 together with the attached carbon atom form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
  2. 如权利要求1所述的化合物,其特征在于,R 1为氟、氯或氰基。 The compound of claim 1 wherein R 1 is fluoro, chloro or cyano.
  3. 如权利要求1所述的化合物,其特征在于,R 1为氟;R 2、R 3与相连的碳原子共同形成3-7元脂肪环或3-7元含氧杂环。 The compound according to claim 1, wherein R 1 is fluorine; and R 2 and R 3 together with the carbon atom to be bonded form a 3-7 membered aliphatic ring or a 3-7 membered oxygen-containing heterocyclic ring.
  4. 如权利要求1所述的化合物,其特征在于,R 1为氰基;R 2、R 3独立为氢原子或C 1-C 3烷基;或者R 2、R 3与相连的碳原子共同形成3-7元脂肪环。 The compound according to claim 1, wherein R 1 is a cyano group; R 2 and R 3 are independently a hydrogen atom or a C 1 - C 3 alkyl group; or R 2 and R 3 are formed together with a carbon atom to which it is bonded. 3-7 yuan fat ring.
  5. 如权利要求1所述的化合物,其特征在于,所述化合物为:The compound of claim 1 wherein said compound is:
    Figure PCTCN2018092208-appb-100002
    Figure PCTCN2018092208-appb-100002
    Figure PCTCN2018092208-appb-100003
    Figure PCTCN2018092208-appb-100003
  6. 一种药物组合物,其特征在于,所述药物组合物包含权利要求1所述的化合物或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐;以及A pharmaceutical composition comprising the compound of claim 1 or an enantiomer thereof, a diastereomer, a racemate, and a mixture thereof, or a pharmaceutical thereof Acceptable salt;
    药学上可接受的载体或赋形剂。A pharmaceutically acceptable carrier or excipient.
  7. 如权利要求1所述的化合物的制备方法,其特征在于,所述方法包括以下步骤:The method of preparing a compound according to claim 1, wherein the method comprises the steps of:
    Figure PCTCN2018092208-appb-100004
    Figure PCTCN2018092208-appb-100004
    (i)中间体Ia经分子内亲核取代反应生成中间体Ib;(i) intermediate Ia by intramolecular nucleophilic substitution reaction to form intermediate Ib;
    (ii)中间体Ib经亲核取代反应生成中间体Ic;(ii) intermediate Ib is nucleophilic substituted to form intermediate Ic;
    (iii)中间体Ic经水解反应生成中间体Id;(iii) intermediate Ic is hydrolyzed to form intermediate Id;
    (iv)中间体Id经亲核取代反应生成中间体Ie;(iv) intermediate Id by nucleophilic substitution reaction to form intermediate Ie;
    (v)中间体Ie与巴比妥酸反应生成通式I所示的化合物,(v) reacting intermediate Ie with barbituric acid to form a compound of formula I,
    各式中,*、R 1、R 2、R 3和R 4的定义如权利要求1所述。 In the formulas, *, R 1 , R 2 , R 3 and R 4 are as defined in claim 1.
  8. 如权利要求1所述的化合物的制备方法,其特征在于,R 1为氰基时,所述方法包括以下步骤: A method of producing a compound according to claim 1, wherein when R 1 is a cyano group, the method comprises the steps of:
    Figure PCTCN2018092208-appb-100005
    Figure PCTCN2018092208-appb-100005
    (i’)中间体I-12经水解反应生成中间体I-13;(i') intermediate I-12 is hydrolyzed to form intermediate I-13;
    (ii’)中间体I-13经分子内亲核取代反应生成中间体I-14;(ii') intermediate I-13 is subjected to intramolecular nucleophilic substitution to give intermediate I-14;
    (iii’)中间体I-14经亲核取代反应生成中间体I-15;(iii') intermediate I-14 is subjected to nucleophilic substitution to give intermediate I-15;
    (iv’)中间体I-15经亲核取代反应生成中间体I-16;(iv') intermediate I-15 is subjected to nucleophilic substitution to give intermediate I-16;
    (v’)中间体I-16与巴比妥酸反应生成式C所示的权利要求1所述的化合物,(v') intermediate I-16 is reacted with barbituric acid to form the compound of claim 1 represented by formula C,
    各式中,*、R 2、R 3和R 4的定义如权利要求1所述。 In the formulas, *, R 2 , R 3 and R 4 are as defined in claim 1.
  9. 如权利要求1所述的化合物或其对映异构体、非对映异构体、外消旋体及其混合物,或其药学上可接受的盐,或权利要求7所述的药物组合物的用途,其特征在于,用于制备治疗细菌感染性疾病的药物。The compound of claim 1 or an enantiomer thereof, a diastereomer, a racemate, and mixtures thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 7. Use for the preparation of a medicament for the treatment of a bacterial infectious disease.
  10. 一种式I化合物中间体,其特征在于,所述式I化合物中间体的结构如式Ia、Ib、Ic、Id或Ie所示:A compound intermediate of formula I, characterized in that the structure of the intermediate of the compound of formula I is as shown in formula Ia, Ib, Ic, Id or Ie:
    Figure PCTCN2018092208-appb-100006
    Figure PCTCN2018092208-appb-100006
    各式中,*、R 1、R 2、R 3和R 4的定义如权利要求1所述。 In the formulas, *, R 1 , R 2 , R 3 and R 4 are as defined in claim 1.
PCT/CN2018/092208 2017-06-22 2018-06-21 Benzoisoxazole spiropyrimidinetrione compound, preparation method therefor and use thereof WO2018233670A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201710482663 2017-06-22
CN201710482663.5 2017-06-22
CN201711116329.4A CN109111466B (en) 2017-06-22 2017-11-13 Benzisoxazole spiropyrimidine triones compound, preparation method and application thereof
CN201711116329.4 2017-11-13

Publications (1)

Publication Number Publication Date
WO2018233670A1 true WO2018233670A1 (en) 2018-12-27

Family

ID=64737498

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/092208 WO2018233670A1 (en) 2017-06-22 2018-06-21 Benzoisoxazole spiropyrimidinetrione compound, preparation method therefor and use thereof

Country Status (1)

Country Link
WO (1) WO2018233670A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3643719A4 (en) * 2017-06-22 2020-11-04 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Thiazolidone spiro pyrimidine trione compound, preparation method therefor and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014114929A1 (en) * 2013-01-23 2014-07-31 Astrazeneca Ab Compounds and methods for treating bacterial infections

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014114929A1 (en) * 2013-01-23 2014-07-31 Astrazeneca Ab Compounds and methods for treating bacterial infections

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BASARAB, GEORG S. ET AL.: "Discovery of Novel DNA Gyrase Inhibiting Spiropyrimidinetriones: Benzisoxazole Fusion with N-Linked Oxazolidinone Substituents Leading to a Clinical Candidate (ETX0914)", JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, no. 15, 9 July 2015 (2015-07-09), pages 6264 - 6282, XP055555089, ISSN: 022-2623, DOI: 10.1021/acs.jmedchem.5b00863 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3643719A4 (en) * 2017-06-22 2020-11-04 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Thiazolidone spiro pyrimidine trione compound, preparation method therefor and uses thereof
US11180509B2 (en) 2017-06-22 2021-11-23 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Thiazolidone spiro pyrimidine trione compound, preparation method therefor and uses thereof

Similar Documents

Publication Publication Date Title
WO2021185233A1 (en) Kras mutant protein inhibitors
JP7099824B2 (en) Antibacterial compound
EP2691396B1 (en) Enopeptins, uses thereof, and methods of synthesis thereto
US10927079B2 (en) Intermediate compound of novel tetrahydronaphthyl urea derivative
JP2010511668A (en) Antibacterial quinoline derivative
WO2013013505A1 (en) 9-aminomethyl substituted tetracycline compound
JP2016536338A (en) Antimicrobial compounds and methods for their production and use
WO2023280136A1 (en) Trideuteromethyl-substituted pyrazino pyrazino quinolinone derivative, and preparation method therefor and use thereof in medicine
WO2021123372A1 (en) Novel compounds and their use
RU2439058C2 (en) Antibacterial chinolin derivatives
CN110799513B (en) Ring-fused thiazolo-2-pyridones, method for the production thereof and use thereof for treating and/or preventing diseases which involve gram-positive bacteria
WO2018233670A1 (en) Benzoisoxazole spiropyrimidinetrione compound, preparation method therefor and use thereof
CN109111466B (en) Benzisoxazole spiropyrimidine triones compound, preparation method and application thereof
JP2010511669A (en) Antibacterial quinoline derivative
JP5466013B2 (en) Antibacterial quinoline derivative
JP2010511670A (en) Antibacterial quinoline derivative
CN109111465B (en) Thiazolidone spiropyrimidine trione compound and preparation method and application thereof
BR112015014145B1 (en) antibacterial compounds, pharmaceutical composition, their preparation and combination processes
CN115551869B (en) Condensed ring 2-pyridone compounds, process for their preparation and their use in the treatment and/or prophylaxis of diseases involving gram-positive bacteria
ES2409536B1 (en) SYNTHETIC HETEROCYCLIC KINONES WITH ANTIMICROBIAL ACTIVITY.
JP7334183B2 (en) glycosyltransferase inhibitor
ES2389070B1 (en) DERIVED FROM PRENILATED DIBENZOFURANIC POLYCETIDES AND THEIR APPLICATION FOR THE TREATMENT OF INFECTIOUS, PARASITIC DISEASES AND TUMORS.
CN115124487A (en) Preparation method and application of arylimino thiazole compounds
WO2009019616A2 (en) Isoprekinamycin analogs and syntheses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18819826

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18819826

Country of ref document: EP

Kind code of ref document: A1