WO2025190151A1 - Benzoselenazole compounds as well as preparation method therefor and use thereof - Google Patents
Benzoselenazole compounds as well as preparation method therefor and use thereofInfo
- Publication number
- WO2025190151A1 WO2025190151A1 PCT/CN2025/081112 CN2025081112W WO2025190151A1 WO 2025190151 A1 WO2025190151 A1 WO 2025190151A1 CN 2025081112 W CN2025081112 W CN 2025081112W WO 2025190151 A1 WO2025190151 A1 WO 2025190151A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- acid
- formula
- alkyl
- benzoselenazepine
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/55—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D205/00—Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D243/00—Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
- C07D243/06—Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
- C07D243/10—Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D281/00—Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one sulfur atom as the only ring hetero atoms
- C07D281/02—Seven-membered rings
- C07D281/04—Seven-membered rings having the hetero atoms in positions 1 and 4
- C07D281/08—Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
- C07D281/10—Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with one six-membered ring
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D305/00—Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms
- C07D305/02—Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings
- C07D305/04—Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
- C07D305/08—Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D517/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having selenium, tellurium, or halogen atoms as ring hetero atoms
Definitions
- the present invention relates to a benzoselenazepine Compounds and their preparation methods and uses.
- Respiratory syncytial virus is one of the most contagious human respiratory viruses. It's aptly named because RSV-infected cells fuse together, forming large cell structures resembling syncytia. It infects 3-10% of the world's population annually and is the most common cause of acute lower respiratory tract illness in infants and young children. It is also the leading cause of hospitalization for lower respiratory tract infections, bronchiolitis, and even death in children under five years of age. Globally, it causes approximately 34 million cases of lower respiratory tract infections each year, including up to 200,000 deaths in children under five. RSV can also cause severe lung infections and death in the elderly.
- RSV is an enveloped virus belonging to the family Streptococcus pneumoniae.
- the RSV genome consists of 10 genes encoding 11 proteins: nonstructural proteins 1 and 2 (NS1 and NS2), nucleoprotein (N), phosphoprotein (P), matrix protein (M), small hydrophobic protein (SH), fusion protein (F), attachment glycoprotein (G), human RNA-dependent RNA polymerase (L), and transcription antibody terminator protein (M2-1) and M2-2 proteins.
- RSV is a major cause of acute lower respiratory tract illness and hospitalization in young children. Globally, RSV causes 33.8 million children under the age of 5 years old each year, of which 3.4 million children require hospitalization for acute lower respiratory tract illness (Ramagopal G., et al, Journal of Clinical and Diagnostic Research, 2016, 10(8): SC05-SC08).
- RSV virus particle inactivators RSV virus particle inactivators
- RSV replication/protein synthesis inhibitors RSV cell binding inhibitors
- RSV cell invasion inhibitors RSV cell invasion inhibitors
- host cell regulators of apoptosis only a few have entered Phase I or II clinical trials.
- Arrow Therapeutics completed a 5-year Phase II clinical trial of the nucleocapsid (N) protein inhibitor RSV-604 in stem cell transplant patients in February 2010 (www.clinicaltriaIs.gov), but has not yet announced the final results. Most other small molecules have suspended their trials for various reasons.
- N nucleocapsid
- RSV-F respiratory syncytial virus fusion protein
- RNAi therapeutics against RSV are also currently under investigation.
- ALN-RSV01 AInyIam Pharmaceuticals, MA, LSA
- ALN-RSV01 is a siRNA targeting the RSV gene.
- Administration of ALN-RSV01 as a nasal spray 2 days before and 3 days after RSV inoculation reduced infection rates in adult volunteers (DeVincenzo J. et al., Proc Natl Acad Sci U S A. 2010 May 11;107(19):8800-5).
- results were insufficient to conclude on antiviral efficacy, although some product efficacy was observed (Zamora MR et al., Am T Respir Crit Care Med. 2011 Feb 15;183(4):531-8).
- Alios's nucleic acid analog ALS-8176 can terminate RNA chain synthesis, inhibit L protein polymerization, and reduce respiratory syncytial virus load in more than 85% of volunteers: Gilead's RSV fusion inhibitor GS-5806 oral treatment can reduce viral load, mucus quality, and symptom scores, etc.
- the technical problem to be solved by the present invention is to overcome the problems of single structure and poor pharmacokinetic properties of RSV inhibitors in the prior art, and to provide a benzoselenazepine Compounds and their preparation methods and uses. These compounds have good RSV inhibitory activity and good in vitro and in vivo pharmacokinetic properties.
- the present invention provides a benzoselenazepine as shown in formula I a compound or a pharmaceutically acceptable salt thereof,
- R 1 , R 2 , R 3 and R 4 are independently hydrogen or deuterium
- R 5 is hydrogen, halogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more Ra ;
- R a is independently deuterium, halogen or hydroxy
- R 6 is hydrogen, —C( ⁇ O)C 1-6 alkyl, —C( ⁇ O)OC 1-6 alkyl, —C( ⁇ O)C 1-6 alkyl substituted by 1 to 3 halogens, or —C( ⁇ O)OC 1-6 alkyl substituted by 1 to 3 halogens.
- the halogen is independently fluorine, chlorine, bromine or iodine, such as fluorine.
- the C 1-6 alkyl group and the C 1-6 alkyl group substituted by one or more Ra are independently C 1-4 alkyl groups, such as methyl.
- the halogen in the -C( ⁇ O)C 1-6 alkyl substituted by 1 to 3 halogens and the -C( ⁇ O)OC 1-6 alkyl substituted by 1 to 3 halogens is independently fluorine, chlorine, bromine or iodine, such as fluorine.
- R 1 , R 2 , R 3 and R 4 are independently hydrogen.
- R 5 is C 1-6 alkyl.
- R 6 is hydrogen, —C( ⁇ O)OC 1-6 alkyl, or —C ( ⁇ O)C 1-6 alkyl substituted by 1 to 3 halogens.
- R 5 is hydrogen, halogen, methyl, difluoromethyl, trifluoromethyl, hydroxymethyl, or methyl substituted with 1 to 3 deuteriums.
- R5 is methyl, -CD3 , -F, -Cl, -Br, -I, -CF3 , -CF2H , or -CH2OH .
- R 5 is methyl
- the benzoselenazepine shown in Formula I is any of the following compounds:
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising the benzoselenazepine as shown in formula I as described above.
- the selection of the pharmaceutical excipients varies depending on the route of administration and the characteristics of the action, and can generally be fillers, diluents, binders, wetting agents, disintegrants, lubricants, emulsifiers, suspending agents, etc. conventional in the art.
- the pharmaceutical composition is prepared by adding the benzoselenazepine of formula I
- the compounds are prepared with one or more pharmaceutical excipients into dosage forms suitable for human or animal use, such as tablets, coated tablets, dragees, suppositories, hard and soft gelatin capsules, solutions, emulsions or suspensions.
- the benzoselenazepine shown in Formula I is administered to the subject by an enteral or parenteral route, such as oral administration, intravenous injection, intramuscular injection, subcutaneous injection or rectal administration.
- the present invention also provides a benzoselenazepine as shown in formula I Use of a compound or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition as described above, in the preparation of a drug for treating or preventing RSV infection.
- the present invention also provides a benzoselenazepine as shown in formula I as described above.
- the preparation method of the compound comprises the following steps: in an organic solvent, in the presence of an acidic reagent, subjecting a compound as shown in Formula II to a substitution reaction as shown below with a benzopyrimidine compound as shown in Formula III to obtain the benzoselenazepine compound as shown in Formula I.
- R 6 is hydrogen, -C( ⁇ O)C 1-6 alkyl, -C( ⁇ O)OC 1-6 alkyl, -C( ⁇ O)C 1-6 alkyl substituted by 1 to 3 halogens, or -C( ⁇ O)OC 1-6 alkyl substituted by 1 to 3 halogens; and R 1 , R 2 , R 3 , R 4 and R 5 are as defined above.
- the operations and conditions in the method 1 can be conventional operations and conditions in this type of reaction in the art. Specifically, they are as follows:
- the substitution reaction can be carried out under gas protection, which can be nitrogen and/or argon.
- the organic solvent may be an alcohol solvent, such as ethanol, isopropanol or n-butanol, for example ethanol.
- the acidic reagent may be ammonium chloride.
- the molar ratio of the benzopyrimidine compound represented by Formula III to the compound represented by Formula II may be 1:(1-2), for example, 1:1.2 or 1:1.3.
- the molar ratio of the acidic reagent to the benzopyrimidine compound represented by Formula III may be (0.02-1):1, for example, 0.05:1 or 0.1:1.
- the reaction temperature of the substitution reaction may be 50-100°C, for example 80°C.
- the progress of the substitution reaction can be monitored by conventional monitoring methods in the art (eg, TLC, HPLC, or NMR).
- the reaction endpoint is generally taken as the disappearance or no-reaction of the compound III, for example, 12-16 hours.
- the present invention provides a compound as shown in Formula II,
- R 1 , R 2 , R 3 and R 4 are as defined above.
- the present invention provides a method for preparing a compound represented by Formula IIa, comprising the following steps:
- Step 1 substitution reaction: In the presence of a base and a catalyst, phenylselenol undergoes a nucleophilic substitution reaction with (2-bromoethyl)benzylcarbamate to produce intermediate II-1;
- Step 2 (cyclization reaction): In the presence of an acid, intermediate II-1 undergoes a cyclization reaction with paraformaldehyde to obtain intermediate II-2;
- Step 3 (Cbz protection removal): Intermediate II-2 is hydrolyzed with acid or subjected to reductive hydrogenation to remove the Cbz protecting group to obtain benzoselenazepine Intermediate IIa.
- step 1, step 2 and step 3 may be conventional operations and conditions for such reactions in the art.
- the base is sodium hydroxide, potassium hydroxide, potassium tert-butoxide, sodium hydride, sodium carbonate, potassium carbonate, cesium carbonate, etc., preferably potassium carbonate.
- the catalyst in step 1, is sodium iodide, potassium iodide, tetrabutylammonium iodide, tetrabutylammonium bromide, triethylammonium benzyl bromide or a crown ether (such as 18-crown-6), etc., preferably sodium iodide.
- the acid in step 2, is a hydrohalic acid (such as hydrochloric acid), sulfuric acid, phosphoric acid, trifluoroacetic acid, methanesulfonic acid, trifluoromethanesulfonic acid, perfluorosulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, etc., preferably p-toluenesulfonic acid.
- hydrohalic acid such as hydrochloric acid
- sulfuric acid sulfuric acid
- phosphoric acid trifluoroacetic acid
- methanesulfonic acid trifluoromethanesulfonic acid
- perfluorosulfonic acid perfluorosulfonic acid
- benzenesulfonic acid p-toluenesulfonic acid, etc., preferably p-toluenesulfonic acid.
- the acid in step 3, is a hydrohalic acid, preferably a hydrobromic acid-acetic acid solution.
- the catalyst in step 3, when reductive hydrogenation is used to remove the Cbz protecting group, is palladium carbon, palladium hydroxide, platinum carbon or Raney nickel, and preferably 1-5% palladium carbon.
- the present invention also provides the following compounds:
- pharmaceutically acceptable means that salts, solvents, excipients, etc. are generally non-toxic, safe, and suitable for use by patients.
- the "patient” is preferably a mammal, more preferably a human.
- pharmaceutically acceptable salt refers to a salt prepared from a compound of the present invention with a relatively non-toxic, pharmaceutically acceptable acid or base.
- a base addition salt can be obtained by contacting the prototype of such compound with a sufficient amount of a pharmaceutically acceptable base in a suitable inert solvent.
- Pharmaceutically acceptable base addition salts include, but are not limited to, lithium salts, sodium salts, potassium salts, calcium salts, aluminum salts, magnesium salts, zinc salts, bismuth salts, ammonium salts, and diethanolamine salts.
- an acid addition salt can be obtained by contacting the prototype of such compound with a sufficient amount of a pharmaceutically acceptable acid in a suitable inert solvent.
- the pharmaceutically acceptable acid includes inorganic acids, including, but not limited to, hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, carbonic acid, phosphoric acid, phosphorous acid, sulfuric acid, and the like.
- the pharmaceutically acceptable acid includes organic acids, including but not limited to acetic acid, propionic acid, oxalic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, salicylic acid, tartaric acid, methanesulfonic acid, isonicotinic acid, acid citric acid, oleic acid, tannic acid, pantothenic acid, bitartrate, ascorbic acid, gentisic acid, fumaric acid, gluconic acid, sugar acid, formic acid, ethanesulfonic acid, pamoic acid (i.e., 4,4'-methylene-bis(3-hydroxy-2-naphthoic acid)), amino acids (e.g., glutamic acid, arginine), etc
- the compounds of the present invention When the compounds of the present invention contain relatively acidic and relatively basic functional groups, they can be converted into base addition salts or acid addition salts.
- base addition salts For details, see Berge et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science 66:1-19 (1977), or Handbook of Pha rmaceutical Salts: Properties, Selection, and Use (P. Heinrich Stahl and Camille G. Wermuth, ed., Wiley-VCH, 2002).
- treatment refers to therapeutic treatment.
- treatment means: (1) alleviating the disease or one or more biological manifestations of the condition, (2) interfering with one or more points in the biological cascade that leads to or causes the condition, or one or more biological manifestations of the condition, (3) ameliorating one or more symptoms, effects, or side effects associated with the condition, or one or more symptoms, effects, or side effects associated with the condition or its treatment, or (4) slowing the progression of the condition or one or more biological manifestations of the condition.
- terapéuticaally effective amount refers to an amount of a compound that, when administered to a patient, is sufficient to effectively treat a disease or condition described herein.
- the “therapeutically effective amount” will vary depending on the compound, the condition and its severity, and the age of the patient to be treated, and can be adjusted as needed by those skilled in the art.
- the reagents and raw materials used in the present invention are commercially available.
- the positive progress of the present invention is that compared with the clinically investigated RSV inhibitor ziresovir, the compound of the present invention has stronger inhibitory activity against respiratory syncytial virus (RSV) and better in vitro and in vivo pharmacokinetic properties. Therefore, the present invention has very good prospects for preparing drugs for treating diseases related to respiratory syncytial virus (RSV) infection.
- RSV respiratory syncytial virus
- II-2 (20 g, 57.75 mmol) obtained in the previous step and 5% palladium on carbon (6.2 g, 2.89 mmol) were dissolved in tetrahydrofuran (150 mL) and reacted at room temperature under a hydrogen atmosphere for 5 hours. After completion, the reaction was filtered and the filtrate was concentrated under reduced pressure to yield the title compound IIa (11.5 g).
- Example 6 N-((3-aminooxetan-3-yl)methyl)-2-(2,3-dihydrobenzo[f][1,4]selenazepine -4(5H)-yl)-6-methylquinazoline-4-amino (I-2) and 4-(((2-(2,3-dihydrobenzo[f][1,4]selenazepine Synthesis of 4-(5H)-6-methylquinazolin-4-yl)amino)methyl)-4-(hydroxymethyl)oxazolin-2-one (I-3)
- 96-well plates were seeded at a density of 6 ⁇ 10 3 cells per well in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS). The next day, cells were infected with sufficient RSV LongStrain (ATCC) to produce approximately 80-90% cytopathic effects after 6 days in the presence of serial half-log dilutions of the compounds in a total volume of 200 ⁇ L per well. After 6 days, cell viability was determined using a Cell Counting Kit-8. The absorbance at 450 nm and the reference value at 630 nm were measured to determine the 50% effective concentration (EC 50 ).
- DMEM Dulbecco's modified Eagle's medium
- FBS fetal bovine serum
- Raw material Human liver microsomes (supplier: Corning, Cat No. 452117, containing approximately 45 mg of liver microsome protein per gram of liver)
- each plate was centrifuged at 4000 rpm and 4°C for 20 minutes.
- Caco-2 cells source American type culture collection (ATCC).
- the transport buffer used in this study was HBSS containing 10.0 mM HEPES, pH 7.40 ⁇ 0.05. Bidirectional transport experiments were performed at a concentration of 2.00 ⁇ M for the test compound, with duplicates per group. Digoxin was tested at a concentration of 10.0 ⁇ M for bidirectional transport, with duplicates per group. Both propranolol and metoprolol were tested at a concentration of 2.00 ⁇ M in the A-to-B direction. The final DMSO concentration was controlled to less than 1%. The samples were incubated for 2 hours in a CO2 incubator at 37.0°C, 5% CO2 , and high humidity (no shaking).
- test compound and control compound in the starting, donor, and receiver solutions were determined by LC-MS/MS, and quantification was performed using the peak area ratio of the test compound to the internal standard. After the transport experiment, the integrity of the Caco-2 cell monolayer was assessed using a lucifer yellow exclusion assay.
- dCr /dt represents the cumulative concentration change rate of the receiving side over time
- Vr is the volume of the solution in the receiving chamber (0.0750 mL for the apical side and 0.250 mL for the basolateral side)
- A is the surface area of transmission (the monolayer cell area is 0.143 cm2 );
- C0 is the initial concentration on the administration side.
- % Solution Recovery 100 ⁇ [(V r ⁇ C r ) + (V d ⁇ C d )] / (V d ⁇ C 0 );
- Vd represents the volume of the solution in the dosing chamber (0.0750 mL on the apical side and 0.250 mL on the basal side);
- Cd and Cr are the concentrations of the compound in the donating chamber (donor) and the receiving chamber (receiver) at the end of the experiment, respectively.
- 0.2 ml of blood was collected from the rat jugular vein at 0.033 h, 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 8 h and 24 h after administration, anticoagulated with EDTA-K2, and placed on ice.
- 0.2 ml of blood was collected from the rat jugular vein at 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after administration, anticoagulated with EDTA-K2, and placed on ice after collection.
- the pharmacokinetic parameters were calculated using the non-compartmental model using Phoenix WinNonlin 7.0 software using the plasma drug concentration data at different time points, providing parameters such as AUC 0-int , C max , T max , T 1/2 , and oral bioavailability (F%). The results are shown in Table 9 below.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Virology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
本申请要求申请日为2024/3/13的中国专利申请2024102876682的优先权。本申请引用上述中国专利申请的全文。This application claims priority to Chinese Patent Application No. 2024102876682, filed on March 13, 2024. This application incorporates the entirety of the aforementioned Chinese Patent Application.
本发明涉及一种苯并硒氮杂类化合物及其制备方法与用途。The present invention relates to a benzoselenazepine Compounds and their preparation methods and uses.
呼吸道合胞病毒(RSV)是最具传染性的人类易感呼吸道病毒之一,由于受RSV感染的细胞会融合在一起,形成类似“合胞体”的大细胞结构,因此被形象地赋予了呼吸道“合胞”病毒的名称。每年感染全世界3-10%的人口,是婴幼儿急性下呼吸道疾病的最常见病因。它也是5岁以下儿童因下呼吸道感染、细支气管炎而住院治疗,甚至死亡的主要原因。每年在全球引起约3400万例的下呼吸道感染病例,其中5岁以下儿童死亡人数高达20万人。此外,呼吸道合胞病毒还可引起老年人严重的肺部感染和死亡。呼吸道合胞病毒感染的其它高风险患者人群包括免疫功能低下的成人,以及患有慢性肺病、先天性心脏病和慢性阻塞性肺病(COPD)的患者,例如慢性支气管炎和肺气肿。呼吸道合胞病毒(RSV)是属于肺炎链球菌科的包膜病毒。RSV基因组由编码11种蛋白的10个基因组成:非结构蛋白1和2(NS1和NS2)、核蛋白(N)、磷蛋白(P)、基质蛋白(M、小疏水蛋白(SH)、融合蛋白(F)、附着糖蛋白(G)、人RNA依赖性RNA聚合酶(L)和转录抗体终止子蛋白(M2-1)和M2-2蛋白。RSV是导致幼儿急性下呼吸道疾病和住院的主要因素。全球范围内,每年RSV导致3380万5岁以下的儿童患病,其中因急性下呼吸道疾病的340万患儿需要住院治疗(Ramagopal G.,et al,Journal of Clinical and Diagnostic Research,2016,10(8):SC05-SC08)。Respiratory syncytial virus (RSV) is one of the most contagious human respiratory viruses. It's aptly named because RSV-infected cells fuse together, forming large cell structures resembling syncytia. It infects 3-10% of the world's population annually and is the most common cause of acute lower respiratory tract illness in infants and young children. It is also the leading cause of hospitalization for lower respiratory tract infections, bronchiolitis, and even death in children under five years of age. Globally, it causes approximately 34 million cases of lower respiratory tract infections each year, including up to 200,000 deaths in children under five. RSV can also cause severe lung infections and death in the elderly. Other high-risk patient populations for RSV infection include immunocompromised adults and those with chronic lung diseases, congenital heart disease, and chronic obstructive pulmonary disease (COPD), such as chronic bronchitis and emphysema. RSV is an enveloped virus belonging to the family Streptococcus pneumoniae. The RSV genome consists of 10 genes encoding 11 proteins: nonstructural proteins 1 and 2 (NS1 and NS2), nucleoprotein (N), phosphoprotein (P), matrix protein (M), small hydrophobic protein (SH), fusion protein (F), attachment glycoprotein (G), human RNA-dependent RNA polymerase (L), and transcription antibody terminator protein (M2-1) and M2-2 proteins. RSV is a major cause of acute lower respiratory tract illness and hospitalization in young children. Globally, RSV causes 33.8 million children under the age of 5 years old each year, of which 3.4 million children require hospitalization for acute lower respiratory tract illness (Ramagopal G., et al, Journal of Clinical and Diagnostic Research, 2016, 10(8): SC05-SC08).
尽管针对RSV复制、发病机制和传播进行了深入的研究,但至今尚无可用于预防呼吸道合胞病毒感染的疫苗,现有临床治疗手段仅包括对症治疗或支持治疗。批准用于预防和治疗的药物是帕利珠单抗和利巴韦林。临床推荐使用这两种药物用于RSV感染的高风险患者,但其效果存在争议(Glick A.F.,et al.,Hospital Pediatrics,2017,7(5):271-278)。Despite extensive research into RSV replication, pathogenesis, and transmission, there is no vaccine available to prevent RSV infection, and current clinical treatment options include only symptomatic or supportive care. Approved drugs for prevention and treatment are palivizumab and ribavirin. These two drugs are clinically recommended for patients at high risk of RSV infection, but their effectiveness remains controversial (Glick A.F., et al., Hospital Pediatrics, 2017, 7(5):271-278).
目前已有多种针对RSV的小分子抑制剂被报道,依照药物同病毒及宿主相互作用机制的不同,这些抑制剂可以分为RSV病毒颗粒失活剂、RSV复制/蛋白质合成抑制剂、RSV细胞结合抑制剂、RSV入侵细胞抑制剂和细胞凋亡的宿主细胞调节剂等。其中,只有少数的进入了I或II期临床试验。阿罗医疗公司(Arrow Therapeutics于2010年2月在干细胞移植患者中完成了核壳体(N)蛋白抑制剂RSV-604的5年的II期临床试验(www.clinicaltriaIs.gov),但是还没有公布最终的结果。多数其它小分子由于各种原因暂停了试验。Ziresovir(AK0529;RO-0529)是一种苯并硫氮杂类小分子RSV抑制剂,口服有效,能选择性抑制呼吸道合胞病毒融合蛋白(RSV-F)(EC50=3nM),具有良好的安全性和药代动力学,同时也是抗呼吸道合胞病毒药物首次在婴儿和自然获得的RSV疾病中显示出临床疗效。Currently, a variety of small molecule inhibitors for RSV have been reported. According to the different mechanisms of drug interaction with the virus and the host, these inhibitors can be divided into RSV virus particle inactivators, RSV replication/protein synthesis inhibitors, RSV cell binding inhibitors, RSV cell invasion inhibitors and host cell regulators of apoptosis. Among them, only a few have entered Phase I or II clinical trials. Arrow Therapeutics completed a 5-year Phase II clinical trial of the nucleocapsid (N) protein inhibitor RSV-604 in stem cell transplant patients in February 2010 (www.clinicaltriaIs.gov), but has not yet announced the final results. Most other small molecules have suspended their trials for various reasons. Ziresovir (AK0529; RO-0529) is a benzothiazepine It is a small molecule RSV inhibitor that is orally effective and can selectively inhibit the respiratory syncytial virus fusion protein (RSV-F) (EC50=3nM). It has good safety and pharmacokinetics. It is also the first time that an anti-respiratory syncytial virus drug has shown clinical efficacy in infants and naturally acquired RSV disease.
目前人们也充分研究了对抗RSV的RNAi疗法。ALN-RSV01(AInyIam PharmaceuticaIs,MA,LSA)是靶向RSV基因的siRNA。RSV接种前2天和后3天鼻喷雾给药在成人志愿者中降低了感染率(DeVincenzo J.等,ProcNatI Acad Sci U S A.2010年5月11日;107(19):8800-5)。在白然感染的肺移植患者中进行的其它II期临床试验中,结果不足以形成抗病毒疗效的结论,尽管观察到了某些产品功效(Zamora MR等,Am TRespir Crit Care Med.2011年2月15日;183(4):531-8)。在类似患者群中进行的ALN-RSV01的另外的IIb期临床试验正在进行中(www.cIinicaItrials.gov)。例如,Alios的核酸类似物ALS-8176可终止RNA链合成,抑制L蛋白聚合,并且可以降低85%上志愿者的呼吸道合胞病毒载量:Gilead的RSV融合抑制剂GS-5806口服治疗可以降低病毒载量、黏液质量,并降低症状评分等。RNAi therapeutics against RSV are also currently under investigation. ALN-RSV01 (AInyIam Pharmaceuticals, MA, LSA) is a siRNA targeting the RSV gene. Administration of ALN-RSV01 as a nasal spray 2 days before and 3 days after RSV inoculation reduced infection rates in adult volunteers (DeVincenzo J. et al., Proc Natl Acad Sci U S A. 2010 May 11;107(19):8800-5). In another Phase II clinical trial in naturally infected lung transplant patients, results were insufficient to conclude on antiviral efficacy, although some product efficacy was observed (Zamora MR et al., Am T Respir Crit Care Med. 2011 Feb 15;183(4):531-8). Another Phase IIb clinical trial of ALN-RSV01 in a similar patient population is ongoing (www.clinicaltrials.gov). For example, Alios's nucleic acid analog ALS-8176 can terminate RNA chain synthesis, inhibit L protein polymerization, and reduce respiratory syncytial virus load in more than 85% of volunteers: Gilead's RSV fusion inhibitor GS-5806 oral treatment can reduce viral load, mucus quality, and symptom scores, etc.
虽然不同作用机制的几种抑制RSV复制的药物和单克隆抗体已经取得实质性进展,但现在临床使用的两种抗病毒药物仍不足以预防和治疗RSV感染,也没有任何小分子药物被批准上市,并且现有的处于临床阶段的药物抑制RSV复制的活性不强,体内暴露量不高。无论如何,人们迫切需要安全有效的RSV疾病的治疗方法。因此RSV抑制剂的市场前景广阔,具有巨大的治疗空间,仍需要开发新的活性强、体内暴露量高的RSV抑制剂。Although substantial progress has been made in the development of several drugs and monoclonal antibodies that inhibit RSV replication with different mechanisms of action, the two antiviral drugs currently in clinical use remain insufficient for the prevention and treatment of RSV infection. No small molecule drugs have been approved for marketing, and existing clinical-stage drugs have weak activity against RSV replication and low in vivo exposure. Regardless, there is an urgent need for safe and effective treatments for RSV disease. Therefore, the market for RSV inhibitors is promising, with enormous therapeutic potential, and the development of new RSV inhibitors with strong activity and high in vivo exposure is still needed.
本发明所要解决的技术问题是为了克服现有技术中RSV抑制剂结构单一、药代性质不佳的问题,而提供了一种苯并硒氮杂类化合物及其制备方法与用途。该苯并硒氮杂类化合物具有较好的RSV抑制活性和较好的体内外药代性质。The technical problem to be solved by the present invention is to overcome the problems of single structure and poor pharmacokinetic properties of RSV inhibitors in the prior art, and to provide a benzoselenazepine Compounds and their preparation methods and uses. These compounds have good RSV inhibitory activity and good in vitro and in vivo pharmacokinetic properties.
本发明提供了一种如式I所示的苯并硒氮杂类化合物或其药学上可接受的盐,
The present invention provides a benzoselenazepine as shown in formula I a compound or a pharmaceutically acceptable salt thereof,
其中,in,
A为-Se-或-Se(=O)-;A is -Se- or -Se(=O)-;
R1、R2、R3和R4独立地为氢或氘;R 1 , R 2 , R 3 and R 4 are independently hydrogen or deuterium;
R5为氢、卤素、C1-6烷基、或被一个或多个Ra取代的C1-6烷基;R 5 is hydrogen, halogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more Ra ;
Ra独立地为氘、卤素或羟基;R a is independently deuterium, halogen or hydroxy;
R6为氢、-C(=O)C1-6烷基、-C(=O)OC1-6烷基、被1至3个卤素取代的-C(=O)C1-6烷基、或被1至3个卤素取代的-C(=O)OC1-6烷基。R 6 is hydrogen, —C(═O)C 1-6 alkyl, —C(═O)OC 1-6 alkyl, —C(═O)C 1-6 alkyl substituted by 1 to 3 halogens, or —C(═O)OC 1-6 alkyl substituted by 1 to 3 halogens.
在本发明的某些优选实施方案中,R5和Ra中,所述卤素独立地为氟、氯、溴或碘,例如氟。In certain preferred embodiments of the present invention, in R 5 and Ra , the halogen is independently fluorine, chlorine, bromine or iodine, such as fluorine.
在本发明的某些优选实施方案中,R5中,所述C1-6烷基与被一个或多个Ra取代的C1-6烷基中的C1-6烷基独立地为C1-4烷基,例如甲基。In certain preferred embodiments of the present invention, in R 5 , the C 1-6 alkyl group and the C 1-6 alkyl group substituted by one or more Ra are independently C 1-4 alkyl groups, such as methyl.
在本发明的某些优选实施方案中,R6中,所述-C(=O)C1-6烷基、-C(=O)OC1-6烷基、被1至3个卤素取代的-C(=O)C1-6烷基、与被1至3个卤素取代的-C(=O)OC1-6烷基中的C1-6烷基独立地为C1-4烷基,例如甲基或叔丁基。In certain preferred embodiments of the present invention, in R6 , the -C(=O) C1-6 alkyl, -C(=O) OC1-6 alkyl, -C(=O)C1-6 alkyl substituted by 1 to 3 halogens, and the C1-6 alkyl in -C(=O) OC1-6 alkyl substituted by 1 to 3 halogens are independently C1-4 alkyl , such as methyl or tert-butyl.
在本发明的某些优选实施方案中,R6中,所述被1至3个卤素取代的-C(=O)C1-6烷基与被1至3个卤素取代的-C(=O)OC1-6烷基中的卤素独立地为氟、氯、溴或碘,例如氟。In certain preferred embodiments of the present invention, in R 6 , the halogen in the -C(═O)C 1-6 alkyl substituted by 1 to 3 halogens and the -C(═O)OC 1-6 alkyl substituted by 1 to 3 halogens is independently fluorine, chlorine, bromine or iodine, such as fluorine.
在本发明的某些优选实施方案中,R1、R2、R3和R4独立地为氢。In certain preferred embodiments of the present invention, R 1 , R 2 , R 3 and R 4 are independently hydrogen.
在本发明的某些优选实施方案中,R5为C1-6烷基。In certain preferred embodiments of the present invention, R 5 is C 1-6 alkyl.
在本发明的某些优选实施方案中,R6为氢、-C(=O)OC1-6烷基、或被1至3个卤素取代的-C(=O)C1- 6烷基。In certain preferred embodiments of the present invention, R 6 is hydrogen, —C(═O)OC 1-6 alkyl, or —C (═O)C 1-6 alkyl substituted by 1 to 3 halogens.
在本发明的某些优选实施方案中,R5为氢、卤素、甲基、二氟甲基、三氟甲基、羟甲基或1至3个氘取代的甲基。In certain preferred embodiments of the present invention, R 5 is hydrogen, halogen, methyl, difluoromethyl, trifluoromethyl, hydroxymethyl, or methyl substituted with 1 to 3 deuteriums.
在本发明的某些优选实施方案中,R5为甲基、-CD3、-F、-Cl、-Br、-I、-CF3、-CF2H或-CH2OH。In certain preferred embodiments of the present invention, R5 is methyl, -CD3 , -F, -Cl, -Br, -I, -CF3 , -CF2H , or -CH2OH .
在本发明的某些优选实施方案中,R5为甲基。In certain preferred embodiments of the present invention, R 5 is methyl.
在本发明的某些优选实施方案中,R6为H、-C(=O)CF3或 In certain preferred embodiments of the present invention, R 6 is H, -C(=O)CF 3 or
在本发明的某些优选实施方案中,所述的如式I所示的苯并硒氮杂类化合物为如下任一化合物:
In certain preferred embodiments of the present invention, the benzoselenazepine shown in Formula I The compound is any of the following compounds:
本发明提供了一种药物组合物,其包括如上所述的如式I所示的苯并硒氮杂类化合物或其药学上可接受的盐,以及至少一种药用辅料。The present invention provides a pharmaceutical composition comprising the benzoselenazepine as shown in formula I as described above. A compound or a pharmaceutically acceptable salt thereof, and at least one pharmaceutical excipient.
所述的药用辅料的选择因施用途径和作用特点而异,通常可为本领域常规的填充剂、稀释剂、粘合剂、湿润剂、崩解剂、润滑剂、乳化剂、助悬剂等。The selection of the pharmaceutical excipients varies depending on the route of administration and the characteristics of the action, and can generally be fillers, diluents, binders, wetting agents, disintegrants, lubricants, emulsifiers, suspending agents, etc. conventional in the art.
在本发明的某些优选实施方案中,所述的药物组合物通过将所述的如式I所示的苯并硒氮杂类化合物与一种或多种药用辅料制成适于人或动物使用的剂型,例如片剂、包衣片剂、糖锭剂、栓剂、硬和软明胶胶囊、溶液剂、乳剂或混悬液剂。In certain preferred embodiments of the present invention, the pharmaceutical composition is prepared by adding the benzoselenazepine of formula I The compounds are prepared with one or more pharmaceutical excipients into dosage forms suitable for human or animal use, such as tablets, coated tablets, dragees, suppositories, hard and soft gelatin capsules, solutions, emulsions or suspensions.
在本发明的某些优选实施方案中,所述的如式I所示的苯并硒氮杂类化合物或其药学上可接受的盐、或如上所述的药物组合物以肠道或非肠道的给药途径给与受试者,例如口服、静脉注射、肌肉注射、皮下注射或直肠途径施用。In certain preferred embodiments of the present invention, the benzoselenazepine shown in Formula I The compound or pharmaceutically acceptable salt thereof, or the pharmaceutical composition as described above is administered to the subject by an enteral or parenteral route, such as oral administration, intravenous injection, intramuscular injection, subcutaneous injection or rectal administration.
本发明还提供了一种如上所述的如式I所示的苯并硒氮杂类化合物或其药学上可接受的盐、或如上所述的药物组合物在制备用于治疗或预防RSV感染的药物中的应用。The present invention also provides a benzoselenazepine as shown in formula I Use of a compound or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition as described above, in the preparation of a drug for treating or preventing RSV infection.
本发明还提供了一种如上所述的如式I所示的苯并硒氮杂类化合物的制备方法,其包括如下步骤:在有机溶剂中,在酸性试剂存在下,将如式II所示的化合物与如式III所式的苯并嘧啶化合物进行如下所示的取代反应,得到所述的如式I所示的苯并硒氮杂类化合物,
The present invention also provides a benzoselenazepine as shown in formula I as described above. The preparation method of the compound comprises the following steps: in an organic solvent, in the presence of an acidic reagent, subjecting a compound as shown in Formula II to a substitution reaction as shown below with a benzopyrimidine compound as shown in Formula III to obtain the benzoselenazepine compound as shown in Formula I. Compounds,
其中,R6为氢、-C(=O)C1-6烷基、-C(=O)OC1-6烷基、被1至3个卤素取代的-C(=O)C1-6烷基、或被1至3个卤素取代的-C(=O)OC1-6烷基;R1、R2、R3、R4和R5的定义如上所述。wherein R 6 is hydrogen, -C(═O)C 1-6 alkyl, -C(═O)OC 1-6 alkyl, -C(═O)C 1-6 alkyl substituted by 1 to 3 halogens, or -C(═O)OC 1-6 alkyl substituted by 1 to 3 halogens; and R 1 , R 2 , R 3 , R 4 and R 5 are as defined above.
所述方法一中的操作和条件可为本领域中该类反应中常规的操作和条件。具体如下:The operations and conditions in the method 1 can be conventional operations and conditions in this type of reaction in the art. Specifically, they are as follows:
所述取代反应可在气体保护下进行。所述气体可为氮气和/或氩气。The substitution reaction can be carried out under gas protection, which can be nitrogen and/or argon.
所述取代反应中,所述有机溶剂可为醇类溶剂,例如乙醇、异丙醇或正丁醇,又例如乙醇。In the substitution reaction, the organic solvent may be an alcohol solvent, such as ethanol, isopropanol or n-butanol, for example ethanol.
所述取代反应中,所述酸性试剂可为氯化铵。In the substitution reaction, the acidic reagent may be ammonium chloride.
所述取代反应中,所述的如式III所示的苯并嘧啶化合物与所述的如式II所示的化合物摩尔比可为1:(1-2),例如1:1.2或1:1.3。In the substitution reaction, the molar ratio of the benzopyrimidine compound represented by Formula III to the compound represented by Formula II may be 1:(1-2), for example, 1:1.2 or 1:1.3.
所述取代反应中,所述酸性试剂与所述的如式III所示的苯并嘧啶化合物摩尔比可为(0.02-1):1,例如0.05:1或0.1:1。In the substitution reaction, the molar ratio of the acidic reagent to the benzopyrimidine compound represented by Formula III may be (0.02-1):1, for example, 0.05:1 or 0.1:1.
所述取代反应的反应温度可为50-100℃,例如80℃。The reaction temperature of the substitution reaction may be 50-100°C, for example 80°C.
所述取代反应的进程可采用本领域中的常规监测方法(例如TLC、HPLC或NMR)进行检测,一般以所述的化合物III消失或不再反应时作为反应终点,例如12-16小时。The progress of the substitution reaction can be monitored by conventional monitoring methods in the art (eg, TLC, HPLC, or NMR). The reaction endpoint is generally taken as the disappearance or no-reaction of the compound III, for example, 12-16 hours.
本发明提供了一种如式II所示的化合物,
The present invention provides a compound as shown in Formula II,
其中,R1、R2、R3和R4的定义如上所述。wherein R 1 , R 2 , R 3 and R 4 are as defined above.
在本发明的某些优选实施方案中,所述如式II所示的化合物为 In certain preferred embodiments of the present invention, the compound shown in Formula II is
本发明提供了一种如式IIa所示的化合物的制备方法,其包括以下步骤:
The present invention provides a method for preparing a compound represented by Formula IIa, comprising the following steps:
步骤1(取代反应):在碱和催化剂存在下,苯硒酚与(2-溴乙基)氨基甲酸苄酯进行亲核取代反应,制得中间体II-1;Step 1 (substitution reaction): In the presence of a base and a catalyst, phenylselenol undergoes a nucleophilic substitution reaction with (2-bromoethyl)benzylcarbamate to produce intermediate II-1;
步骤2(环化反应):在酸存在下,中间体II-1与多聚甲醛进行环化反应,得到中间体II-2;Step 2 (cyclization reaction): In the presence of an acid, intermediate II-1 undergoes a cyclization reaction with paraformaldehyde to obtain intermediate II-2;
步骤3(脱Cbz保护):中间体II-2用酸水解或还原氢化脱Cbz保护基,得苯并硒氮杂中间体IIa。Step 3 (Cbz protection removal): Intermediate II-2 is hydrolyzed with acid or subjected to reductive hydrogenation to remove the Cbz protecting group to obtain benzoselenazepine Intermediate IIa.
所述步骤1、步骤2和步骤3中的操作和条件可为本领域中该类反应常规的操作和条件。The operations and conditions in step 1, step 2 and step 3 may be conventional operations and conditions for such reactions in the art.
在本发明的某些优选实施方案中,步骤1中,所述碱为氢氧化钠、氢氧化钾、叔丁醇钾、氢化钠、碳酸钠、碳酸钾、碳酸铯等,优选碳酸钾。In certain preferred embodiments of the present invention, in step 1, the base is sodium hydroxide, potassium hydroxide, potassium tert-butoxide, sodium hydride, sodium carbonate, potassium carbonate, cesium carbonate, etc., preferably potassium carbonate.
在本发明的某些优选实施方案中,步骤1中,所述催化剂为碘化钠、碘化钾、四丁基碘化铵、四丁基溴化铵、三乙胺溴化苄或冠醚(例如18-冠-6)等,优选碘化钠。In certain preferred embodiments of the present invention, in step 1, the catalyst is sodium iodide, potassium iodide, tetrabutylammonium iodide, tetrabutylammonium bromide, triethylammonium benzyl bromide or a crown ether (such as 18-crown-6), etc., preferably sodium iodide.
在本发明的某些优选实施方案中,步骤2中,所述酸为氢卤酸(例如盐酸)、硫酸、磷酸、三氟乙酸、甲磺酸、三氟甲磺酸、全氟磺酸、苯磺酸、对甲苯磺酸等,优选对甲苯磺酸。In certain preferred embodiments of the present invention, in step 2, the acid is a hydrohalic acid (such as hydrochloric acid), sulfuric acid, phosphoric acid, trifluoroacetic acid, methanesulfonic acid, trifluoromethanesulfonic acid, perfluorosulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, etc., preferably p-toluenesulfonic acid.
在本发明的某些优选实施方案中,步骤3中,所述酸为氢卤酸,优选氢溴酸乙酸溶液。In certain preferred embodiments of the present invention, in step 3, the acid is a hydrohalic acid, preferably a hydrobromic acid-acetic acid solution.
在本发明的某些优选实施方案中,步骤3中,采用还原氢化脱Cbz保护基时,催化剂为钯碳、氢氧化钯、铂碳或雷尼镍等,优选为1-5%钯碳。In certain preferred embodiments of the present invention, in step 3, when reductive hydrogenation is used to remove the Cbz protecting group, the catalyst is palladium carbon, palladium hydroxide, platinum carbon or Raney nickel, and preferably 1-5% palladium carbon.
本发明还提供了如下所示的化合物,
The present invention also provides the following compounds:
除非另有说明,在本发明说明书和权利要求书中出现的以下术语具有下述含义:Unless otherwise specified, the following terms appearing in the present specification and claims have the following meanings:
术语“药学上可接受的”是指盐、溶剂、辅料等一般无毒、安全,并且适合于患者使用。所述的“患者”优选哺乳动物,更优选为人类。The term "pharmaceutically acceptable" means that salts, solvents, excipients, etc. are generally non-toxic, safe, and suitable for use by patients. The "patient" is preferably a mammal, more preferably a human.
术语“药学上可接受的盐”是指本发明化合物与相对无毒的、药学上可接受的酸或碱制备得到的盐。当本发明的化合物中含有相对酸性的功能团时,可以通过在合适的惰性溶剂中用足够量的药学上可接受的碱与这类化合物的原型接触的方式获得碱加成盐。药学上可接受的碱加成盐包括但不限于:锂盐、钠盐、钾盐、钙盐、铝盐、镁盐、锌盐、铋盐、铵盐、二乙醇胺盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在合适的惰性溶剂中用足够量的药学上可接受的酸与这类化合物的原型接触的方式获得酸加成盐。所述的药学上可接受的酸包括无机酸,所述无机酸包括但不限于:盐酸、氢溴酸、氢碘酸、硝酸、碳酸、磷酸、亚磷酸、硫酸等。所述的药学上可接受的酸包括有机酸,所述有机酸包括但不限于:乙酸、丙酸、草酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、水杨酸、酒石酸、甲磺酸、异烟酸、酸式柠檬酸、油酸、单宁酸、泛酸、酒石酸氢、抗坏血酸、龙胆酸、富马酸、葡糖酸、糖酸、甲酸、乙磺酸、双羟萘酸(即4,4’-亚甲基-双(3-羟基-2-萘甲酸))、氨基酸(例如谷氨酸、精氨酸)等。当本发明的化合物中含有相对酸性和相对碱性的官能团时,可以被转换成碱加成盐或酸加成盐。具体可参见Berge et al.,"Pharmaceutical Salts",Journal of Pharmaceutical Science 66:1-19(1977)、或、Handbook of Pharmaceutical Salts:Properties,Selection,and Use(P.Heinrich Stahl and Camille G.Wermuth,ed.,Wiley-VCH,2002)。The term "pharmaceutically acceptable salt" refers to a salt prepared from a compound of the present invention with a relatively non-toxic, pharmaceutically acceptable acid or base. When the compound of the present invention contains a relatively acidic functional group, a base addition salt can be obtained by contacting the prototype of such compound with a sufficient amount of a pharmaceutically acceptable base in a suitable inert solvent. Pharmaceutically acceptable base addition salts include, but are not limited to, lithium salts, sodium salts, potassium salts, calcium salts, aluminum salts, magnesium salts, zinc salts, bismuth salts, ammonium salts, and diethanolamine salts. When the compound of the present invention contains a relatively basic functional group, an acid addition salt can be obtained by contacting the prototype of such compound with a sufficient amount of a pharmaceutically acceptable acid in a suitable inert solvent. The pharmaceutically acceptable acid includes inorganic acids, including, but not limited to, hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, carbonic acid, phosphoric acid, phosphorous acid, sulfuric acid, and the like. The pharmaceutically acceptable acid includes organic acids, including but not limited to acetic acid, propionic acid, oxalic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, salicylic acid, tartaric acid, methanesulfonic acid, isonicotinic acid, acid citric acid, oleic acid, tannic acid, pantothenic acid, bitartrate, ascorbic acid, gentisic acid, fumaric acid, gluconic acid, sugar acid, formic acid, ethanesulfonic acid, pamoic acid (i.e., 4,4'-methylene-bis(3-hydroxy-2-naphthoic acid)), amino acids (e.g., glutamic acid, arginine), etc. When the compounds of the present invention contain relatively acidic and relatively basic functional groups, they can be converted into base addition salts or acid addition salts. For details, see Berge et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science 66:1-19 (1977), or Handbook of Pha rmaceutical Salts: Properties, Selection, and Use (P. Heinrich Stahl and Camille G. Wermuth, ed., Wiley-VCH, 2002).
术语“治疗”指治疗性疗法。涉及具体病症时,治疗指:(1)缓解疾病或者病症的一种或多种生物学表现,(2)干扰导致或引起病症的生物级联中的一个或多个点,或病症的一种或多种生物学表现,(3)改善与病症相关的一种或多种症状、影响或副作用,或者与病症或其治疗相关的一种或多种症状、影响或副作用,(4)减缓病症或者病症的一种或多种生物学表现发展。The term "treat" refers to therapeutic treatment. When referring to a specific condition, treatment means: (1) alleviating the disease or one or more biological manifestations of the condition, (2) interfering with one or more points in the biological cascade that leads to or causes the condition, or one or more biological manifestations of the condition, (3) ameliorating one or more symptoms, effects, or side effects associated with the condition, or one or more symptoms, effects, or side effects associated with the condition or its treatment, or (4) slowing the progression of the condition or one or more biological manifestations of the condition.
术语“治疗有效量”是指在给予患者时,足以有效治疗本文所述的疾病或病症的化合物的量。“治疗有效量”将根据化合物、病症及其严重度、以及欲治疗患者的年龄而变化,可由本领域技术人员根据需要进行调整。The term "therapeutically effective amount" refers to an amount of a compound that, when administered to a patient, is sufficient to effectively treat a disease or condition described herein. The "therapeutically effective amount" will vary depending on the compound, the condition and its severity, and the age of the patient to be treated, and can be adjusted as needed by those skilled in the art.
在符合本领域常识的基础上,上述各优选条件,可任意组合,即得本发明各较佳实例。On the basis of conforming to the common sense in this field, the above-mentioned preferred conditions can be arbitrarily combined to obtain the preferred embodiments of the present invention.
本发明所用试剂和原料均市售可得。The reagents and raw materials used in the present invention are commercially available.
本发明的积极进步效果在于:与临床在研RSV抑制剂Ziresovir相比,本发明化合物具有更强的抑制呼吸道合胞病毒(RSV)活性和更优的体内外药代动力学性质,因此,本发明用于制备治疗呼吸道合胞病毒(RSV)感染相关疾病的药物具有非常好的前景。The positive progress of the present invention is that compared with the clinically investigated RSV inhibitor ziresovir, the compound of the present invention has stronger inhibitory activity against respiratory syncytial virus (RSV) and better in vitro and in vivo pharmacokinetic properties. Therefore, the present invention has very good prospects for preparing drugs for treating diseases related to respiratory syncytial virus (RSV) infection.
下面通过实施例的方式进一步说明本发明,但并不因此将本发明限制在所述的实施例范围之中。下列实施例中未注明具体条件的实验方法,按照常规方法和条件,或按照商品说明书选择。The present invention is further illustrated by way of examples below, but the present invention is not limited to the scope of the examples. Experimental methods in the following examples where specific conditions are not specified were performed according to conventional methods and conditions, or selected according to the product specifications.
实施例1:2,3,4,5-四氢苯并[f][1,4]硒氮杂(IIa)的合成
Example 1: 2,3,4,5-tetrahydrobenzo[f][1,4]selenazepine Synthesis of (IIa)
步骤1:(2-(苯基苯丙)乙基)氨基甲酸苄酯(II-1)Step 1: Benzyl (2-(phenylphenylpropyl)ethyl)carbamate (II-1)
将苯硒酚(50g,318.30mmol),(2-溴乙基)氨基甲酸苄酯(90.4g,350.13mmol),碘化钠(9.5g,63.66mmol)和碳酸钾(66.0g,477.53mmol)溶解于N,N-二甲甲酰胺(600mL)。在氮气保护下,升温至110℃搅拌反应24小时。将反应液加水淬灭,用二氯甲烷萃取,合并有机相,用无水硫酸钠干燥,过滤浓缩,得中间体II-1(96.5g),不经进一步纯化而在下一步骤中使用。LC-MS(ESI,m/z)336[M+H]+;1H NMR(500MHz,DMSO-d6)δ7.55–7.44(m,3H),7.40–7.21(m,8H),5.02(s,2H),3.28–3.23(m,2H),2.99(t,2H).Dissolve phenylselenol (50 g, 318.30 mmol), benzyl (2-bromoethyl)carbamate (90.4 g, 350.13 mmol), sodium iodide (9.5 g, 63.66 mmol), and potassium carbonate (66.0 g, 477.53 mmol) in N,N-dimethylformamide (600 mL). Under nitrogen, heat to 110°C and stir for 24 hours. The reaction mixture is quenched with water and extracted with dichloromethane. The organic phases are combined, dried over anhydrous sodium sulfate, filtered, and concentrated to afford Intermediate II-1 (96.5 g), which is used in the next step without further purification. LC-MS (ESI, m/z) 336[M+H] + ; 1 H NMR (500MHz, DMSO-d 6 ) δ7.55–7.44(m,3H),7.40–7.21(m,8H),5.02(s,2H),3.28–3.23(m,2H),2.99(t,2H).
步骤2:2,3-二氢苯并[f][1,4]硒氮杂卓-4(5H)-羧酸苄酯(II-2)Step 2: Benzyl 2,3-dihydrobenzo[f][1,4]selenazepine-4(5H)-carboxylate (II-2)
将化合物II-1(60g,179.49mmol)溶解于甲苯(500mL)中,加入对甲苯磺酸(9.3g,53.85mmol)。加热至70℃,分批加入多聚甲醛(53.9g,1.79mol),搅拌反应16小时,再升温回流反应1小时。冷却至室温,向反应液中加入500mL水和500mL乙酸乙酯,搅拌30min,分液,有机相依次用饱和碳酸氢钠水溶液和食盐水洗涤。减压浓缩,粗品用硅胶柱层析纯化,得标题化合物II-2(48.6g)。LC-MS(ESI,m/z)348[M+H]+;1H NMR(500MHz,DMSO-d6)δ7.63(dd,1H),7.42–7.14(m,8H),5.01(d,2H),4.57(s,1H),4.54(s,1H),4.11–4.03(m,2H),2.88(dt,2H).Dissolve compound II-1 (60 g, 179.49 mmol) in toluene (500 mL) and add p-toluenesulfonic acid (9.3 g, 53.85 mmol). Heat to 70°C and add paraformaldehyde (53.9 g, 1.79 mol) in portions. Stir and react for 16 hours, then reflux for 1 hour. Cool to room temperature, add 500 mL of water and 500 mL of ethyl acetate to the reaction mixture, stir for 30 minutes, separate the layers, and wash the organic phase with saturated sodium bicarbonate solution and brine. Concentrate under reduced pressure, and purify the crude product by silica gel column chromatography to obtain the title compound II-2 (48.6 g). LC-MS(ESI,m/z)348[M+H] + ; 1 H NMR(500MHz,DMSO-d 6 )δ7.63(dd,1H),7.42–7.14(m,8H),5.01(d,2H),4.57(s,1H),4.54(s,1H),4.11–4.03(m,2H),2.88(dt,2H).
步骤3:2,3,4,5-四氢苯并[f][1,4]硒氮杂卓氢溴酸盐(IIa)Step 3: 2,3,4,5-Tetrahydrobenzo[f][1,4]selenazepine hydrobromide (IIa)
将上一步得到的II-2(40.0g,115.51mmol)溶解于氢溴酸乙酸(33%,250mL)溶液,室温反应3小时。减压除去溶剂,加入碳酸氢钠中和,用二氯甲烷萃取,合并有机相,无水硫酸钠干燥,过滤浓缩,用硅胶柱层析纯化,得标题化合物IIa(20.6g)。LC-MS(ESI,m/z)214[M+H]+;1H NMR(400MHz,Chloroform-d)δ7.65–7.58(m,1H),7.25(d,1H),7.17–7.13(m,2H),7.04(ddd,1H),4.11(s,2H),3.54–3.49(m,2H),2.77–2.72(m,2H).II-2 (40.0 g, 115.51 mmol) obtained in the previous step was dissolved in hydrobromic acid/acetic acid (33%, 250 mL) and allowed to react at room temperature for 3 hours. The solvent was removed under reduced pressure, and the mixture was neutralized with sodium bicarbonate. The mixture was extracted with dichloromethane, and the combined organic phases were dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography to obtain the title compound IIa (20.6 g). LC-MS (ESI, m/z) 214 [M+H] + ; 1 H NMR (400 MHz, Chloroform-d) δ 7.65–7.58 (m, 1H), 7.25 (d, 1H), 7.17–7.13 (m, 2H), 7.04 (ddd, 1H), 4.11 (s, 2H), 3.54–3.49 (m, 2H), 2.77–2.72 (m, 2H).
或,将上一步得到的II-2(20g,57.75mmol)和5%钯碳(6.2g,2.89mmol)溶解于四氢呋喃(150mL)中,在氢气氛围下,室温反应5小时,反应完全,过滤,滤液减压浓缩,得到标题化合物IIa(11.5g)。LC-MS(ESI,m/z)214[M+H]+;1H NMR(400MHz,Chloroform-d)δ7.65–7.58(m,1H),7.25(d,1H),7.17–7.13(m,2H),7.04(ddd,1H),4.11(s,2H),3.54–3.49(m,2H),2.77–2.72(m,2H).Alternatively, II-2 (20 g, 57.75 mmol) obtained in the previous step and 5% palladium on carbon (6.2 g, 2.89 mmol) were dissolved in tetrahydrofuran (150 mL) and reacted at room temperature under a hydrogen atmosphere for 5 hours. After completion, the reaction was filtered and the filtrate was concentrated under reduced pressure to yield the title compound IIa (11.5 g). LC-MS (ESI, m/z) 214 [M+H] + ; 1 H NMR (400 MHz, Chloroform-d) δ 7.65–7.58 (m, 1H), 7.25 (d, 1H), 7.17–7.13 (m, 2H), 7.04 (ddd, 1H), 4.11 (s, 2H), 3.54–3.49 (m, 2H), 2.77–2.72 (m, 2H).
实施例2:N-((3-氨基氧杂环丁-3-基)甲基)-2-氯-6-甲基喹唑啉-4-胺(IIIa)的合成
Example 2: Synthesis of N-((3-aminooxetan-3-yl)methyl)-2-chloro-6-methylquinazolin-4-amine (IIIa)
步骤1:3-(氨基甲基)氧杂环丁-3-胺(III-1)Step 1: 3-(Aminomethyl)oxetan-3-amine (III-1)
向3-(氨甲基)-N,N-二苄基氧杂环丁-3-胺(10g,35mmol)的乙醇(150mL)溶液中加入10%Pd/C(1.13g),反应混合液在氢气氛围下80℃下搅拌反应16小时。过滤溶液,收集滤液,在40℃下减压浓缩。得标题化合物III-1(3.72g,黄色油状物)。LC-MS(ESI):103[M+H]+;1H NMR(400MHz,CD3OD)δ4.45(s,4H),2.91(s,2H).To a solution of 3-(aminomethyl)-N,N-dibenzyloxetan-3-amine (10 g, 35 mmol) in ethanol (150 mL) was added 10% Pd/C (1.13 g). The reaction mixture was stirred at 80°C under a hydrogen atmosphere for 16 hours. The solution was filtered, the filtrate collected, and concentrated under reduced pressure at 40°C. This afforded the title compound III-1 (3.72 g, yellow oil). LC-MS (ESI): 103 [M+H] + ; 1 H NMR (400 MHz, CD 3 OD) δ 4.45 (s, 4H), 2.91 (s, 2H).
步骤2:N-((3-氨氧杂环丁-3-基)甲基)-2-氯-6-甲基喹唑啉-4-胺(IIIa)Step 2: N-((3-aminooxetan-3-yl)methyl)-2-chloro-6-methylquinazolin-4-amine (IIIa)
将2,4-二氯-6-甲基喹唑林(3.6g,17mmol)、化合物III-1(1.90g,19mmol)和三乙胺(0.36g,3.5mmol)溶解于甲醇(40mL)中,在室温下搅拌反应12小时。减压浓缩至干,将残余物用硅胶柱层析纯化,得标题化合物IIIa(2.5g,黄色固体)。LC-MS(ESI,m/z)279[M+H]+;1H NMR(400MHz,DMSO-d6)δ8.68(s,1H),8.22(s,1H),7.67(dd,1H),7.56(d,1H),4.57(d,2H),4.40(d,2H),3.91(s,2H),3.20(s,2H),2.49(s,3H).2,4-Dichloro-6-methylquinazoline (3.6 g, 17 mmol), compound III-1 (1.90 g, 19 mmol), and triethylamine (0.36 g, 3.5 mmol) were dissolved in methanol (40 mL) and stirred at room temperature for 12 hours. The mixture was concentrated to dryness under reduced pressure, and the residue was purified by silica gel column chromatography to obtain the title compound IIIa (2.5 g, yellow solid). LC-MS (ESI, m/z) 279 [M+H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.68 (s, 1H), 8.22 (s, 1H), 7.67 (dd, 1H), 7.56 (d, 1H), 4.57 (d, 2H), 4.40 (d, 2H), 3.91 (s, 2H), 3.20 (s, 2H), 2.49 (s, 3H).
实施例3:(3-(((2-氯-6-甲基喹唑啉-4-基)氨基)甲基)氧杂环丁-3-基)氨基甲酸叔丁酯(IIIb)的合成
Example 3: Synthesis of tert-butyl (3-(((2-chloro-6-methylquinazolin-4-yl)amino)methyl)oxetan-3-yl)carbamate (IIIb)
将化合物IIIa(4.2g,15mmol)溶解于二氯甲烷中,加入三乙胺(2.1g,21mmol)和碳酸二叔丁酯(3.39g,15.5mmol).室温搅拌反应2小时,反应液用二氯甲烷(100mL)稀释,依次用饱和氯化铵(100mL)和食盐水(100mL)洗涤,无水硫酸镁干燥,过滤浓缩,硅胶柱层析纯化(PE:EtOAc=3:1至2:1),得标题化合物(4.25g,白色固体)。LC-MS(ESI,m/z)379[M+H]+;1H NMR(400MHz,DMSO-d6)δ8.57(t,1H),8.06(s,1H),7.65(dd,1H),7.53(d,1H),7.48(br s,1H),4.59-4.58(m,2H),4.55-4.53(m,2H),4.02-4.01(m,2H),2.46(s,3H),1.35(s,9H).Dissolve compound IIIa (4.2 g, 15 mmol) in dichloromethane, add triethylamine (2.1 g, 21 mmol) and di-tert-butyl carbonate (3.39 g, 15.5 mmol). Stir the reaction at room temperature for 2 hours. The reaction solution is diluted with dichloromethane (100 mL), washed with saturated ammonium chloride (100 mL) and brine (100 mL), dried over anhydrous magnesium sulfate, filtered, concentrated, and purified by silica gel column chromatography (PE:EtOAc = 3:1 to 2:1) to obtain the title compound (4.25 g, white solid). LC-MS (ESI, m/z) 379[M+H] + ; 1 H NMR (400MHz, DMSO-d 6 ) δ8.57(t,1H),8.06(s,1H),7.65(dd,1H),7.53(d,1H),7.48(br s,1H),4.59-4.58(m,2H),4.55-4.53(m,2H),4.02-4.01(m,2H),2.46(s,3H),1.35(s,9H).
实施例4:N-(3-(((2-氯-6-甲基喹唑啉-4-基)氨基)甲基)氧杂环丁-3-基)-2,2,2-三氟乙酰胺(IIIc)的合成
Example 4: Synthesis of N-(3-(((2-chloro-6-methylquinazolin-4-yl)amino)methyl)oxetan-3-yl)-2,2,2-trifluoroacetamide (IIIc)
将化合物IIIa(1.0g,3.59mmol)溶解于二氯甲烷(50mL)中,加入三乙胺(892mg,8.81mmol)和三氟乙酸酐(1.36g,6.46mmol),室温搅拌2小时,将反应液倒入水中,用二氯甲烷萃取2次,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,得标题化合物IIIc(1.2g,白色固体)。LC-MS(ESI,m/z)375[M+H]+;1HNMR(400MHz,DMSO-d6)δ9.89(s,1H),8.78(t,1H),8.06(s,1H),7.65(d,1H),7.54(d,1H),4.72(d,2H),4.60(d,2H),4.16(d,2H),2.47(s,3H).Dissolve compound IIIa (1.0 g, 3.59 mmol) in dichloromethane (50 mL), add triethylamine (892 mg, 8.81 mmol) and trifluoroacetic anhydride (1.36 g, 6.46 mmol), and stir at room temperature for 2 hours. Pour the reaction solution into water and extract twice with dichloromethane. Combine the organic phases, wash with saturated brine, and dry over anhydrous sodium sulfate. Filter and concentrate to obtain the title compound IIIc (1.2 g, white solid). LC-MS(ESI,m/z)375[M+H] + ; 1 HNMR(400MHz,DMSO-d 6 )δ9.89(s,1H),8.78(t,1H),8.06(s,1H),7.65(d,1H),7.54(d,1H),4.72(d,2H),4.60(d,2H),4.16(d,2H),2.47(s,3H).
实施例5:(3-(((2-(2,3-二氢苯并[f][1,4]硒氮杂-4(5H)-基)-6-甲基喹唑啉-4-基)氨基)甲基)氧杂环丁-3-基)氨基甲酸叔丁酯(I-1)的合成
Example 5: (3-(((2-(2,3-dihydrobenzo[f][1,4]selenazepine Synthesis of tert-butyl (4(5H)-yl)-6-methylquinazolin-4-yl)amino)methyl)oxetan-3-yl)carbamate (I-1)
将化合物IIIb(4.2g,11.1mmol)和化合物IIa(3.06g,14.4mmol)溶解于乙醇中(75mL),加入氯化铵(60mg,1.1mmol)。氮气保护下,升温至80℃搅拌反应16小时。冷却至室温,减压浓缩,硅胶柱层析纯化(DCM:MeOH=50:1至15:1),得标题化合物(4.7g,白色固体)。LC-MS(ESI,m/z)556[M+H]+;1H NMR(400MHz,CD3OD)δ7.72(dd,1H),7.68(br s,1H),7.58(dd,1H),7.43(dd,1H),7.38(d,1H),7.22(td,1H),7.07(td,1H),5.04(s,2H),4.80-4.78(m,2H),4.67-4.65(m,2H),4.53-4.51(m,2H),4.20(s,2H),3.03-3.01(m,2H),2.38(s,3H),1.40(s,9H).Dissolve compound IIIb (4.2 g, 11.1 mmol) and compound IIa (3.06 g, 14.4 mmol) in ethanol (75 mL) and add ammonium chloride (60 mg, 1.1 mmol). Under nitrogen, heat to 80°C and stir for 16 hours. Cool to room temperature, concentrate under reduced pressure, and purify by silica gel column chromatography (DCM:MeOH = 50:1 to 15:1) to obtain the title compound (4.7 g, white solid). LC-MS (ESI, m/z) 556 [M+H] + ; 1 H NMR (400MHz, CD 3 OD) δ7.72 (dd, 1H), 7.68 (br s,1H),7.58(dd,1H),7.43(dd,1H),7.38(d,1H),7.22(td,1H),7.07(td,1H),5.04(s,2H),4.80-4.78( m,2H),4.67-4.65(m,2H),4.53-4.51(m,2H),4.20(s,2H),3.03-3.01(m,2H),2.38(s,3H),1.40(s,9H).
实施例6:N-((3-氨基氧杂环丁-3-基)甲基)-2-(2,3-二氢苯并[f][1,4]硒氮杂-4(5H)-基)-6-甲基喹唑啉-4-氨基(I-2)和4-(((2-(2,3-二氢苯并[f][1,4]硒氮杂-4(5H)-基)-6-甲基喹唑啉-4-基)氨基)甲基)-4-(羟甲基)恶唑啉-2-酮(I-3)的合成
Example 6: N-((3-aminooxetan-3-yl)methyl)-2-(2,3-dihydrobenzo[f][1,4]selenazepine -4(5H)-yl)-6-methylquinazoline-4-amino (I-2) and 4-(((2-(2,3-dihydrobenzo[f][1,4]selenazepine Synthesis of 4-(5H)-6-methylquinazolin-4-yl)amino)methyl)-4-(hydroxymethyl)oxazolin-2-one (I-3)
将化合物I-1(4.6g,8.3mmol)溶解于三氟乙酸(20mL)中,室温搅拌反应15分钟,然后将反应液倒入饱和碳酸钠水溶液(150mL),用二氯甲烷萃取(50mLx5),合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,硅胶柱层析分离纯化的标题化合物I-2(1.2g,白色固体)和I-3(1.2g,白色固体)。Compound I-1 (4.6 g, 8.3 mmol) was dissolved in trifluoroacetic acid (20 mL), stirred at room temperature for 15 minutes, and then the reaction solution was poured into a saturated aqueous sodium carbonate solution (150 mL), extracted with dichloromethane (50 mL x 5), and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and separated and purified by silica gel column chromatography to obtain the title compounds I-2 (1.2 g, white solid) and I-3 (1.2 g, white solid).
I-2:LCMS(ES,m/z)456[M+H]+;1H NMR(400MHz,CD3OD)δ7.71(dd,1H),7.68(br s,1H),7.58(dd,1H),7.37(dd,1H),7.32(d,1H),7.20(td,1H),7.05(td,1H),5.02(s,2H),4.65(d,2H),4.55(d,2H),4.50-4.52(m,2H),4.05(s,2H),3.03-2.97(m,2H),2.38(s,3H).I-2: LCMS (ES, m/z) 456 [M+H] + ; 1 H NMR (400MHz, CD 3 OD) δ7.71 (dd, 1H), 7.68 (br s,1H),7.58(dd,1H),7.37(dd,1H),7.32(d,1H),7.20(td,1H),7.05(td,1H),5.02(s,2H), 4.65(d,2H),4.55(d,2H),4.50-4.52(m,2H),4.05(s,2H),3.03-2.97(m,2H),2.38(s,3H).
I-3:LCMS(ES,m/z)500[M+H]+;1H NMR(400MHz,CD3OD)δ7.69-7.65(m,2H),7.58(d,1H),7.42-7.40(m,1H),7.35(d,1H),7.22(t,1H),7.07(t,1H),5.07-4.97(m,2H),4.58-4.54(m,2H),4.34(d,1H),4.27(d,1H),4.05(d,1H),3.83(d,1H),3.70-3.65(m,2H),3.02-3.01(m,2H),2.39(s,3H).I-3: LCMS (ES, m/z) 500 [M+H] + ; 1 H NMR (400MHz, CD 3 OD)δ7.69-7.65(m,2H),7.58(d,1H),7.42-7.40(m,1H),7.35(d,1H),7.22(t,1H),7.07(t,1H),5.07-4.97(m,2H),4.5 8-4.54(m,2H),4.34(d,1H),4.27(d,1H),4.05(d,1H),3.83(d,1H),3.70-3.65(m,2H),3.02-3.01(m,2H),2.39(s,3H).
实施例7:N-((3-氨基氧杂环丁-3-基)甲基)-2-(2,3-二氢苯并[f][1,4]硒氮杂-4(5H)-基)-6-甲基喹唑啉-4-氨基(I-2)的合成
Example 7: N-((3-aminooxetan-3-yl)methyl)-2-(2,3-dihydrobenzo[f][1,4]selenazepine Synthesis of 4-(5H)-yl)-6-methylquinazoline-4-amino (I-2)
将化合物IIIa(2.78g,10.0mmol)和化合物IIa(3.06g,14.4mmol)溶解于乙醇中(75mL),加入氯化铵(60mg,1.1mmol)。氮气保护下,升温至80℃搅拌反应16小时。冷却至室温,减压浓缩,用二氯甲烷重新溶解,饱和碳酸氢钠洗涤,无水硫酸钠干燥,过滤浓缩,硅胶柱层析纯化(DCM:MeOH=50:1至15:1),得标题化合物(3.12g,白色固体)。LCMS(ES,m/z)456[M+H]+;1H NMR(400MHz,CD3OD)δ7.71(dd,1H),7.68(br s,1H),7.58(dd,1H),7.37(dd,1H),7.32(d,1H),7.20(td,1H),7.05(td,1H),5.02(s,2H),4.65(d,2H),4.55(d,2H),4.50 -4.52(m,2H),4.05(s,2H),3.03-2.97(m,2H),2.38(s,3H).Dissolve compound IIIa (2.78 g, 10.0 mmol) and compound IIa (3.06 g, 14.4 mmol) in ethanol (75 mL) and add ammonium chloride (60 mg, 1.1 mmol). Under nitrogen, heat to 80°C and stir for 16 hours. Cool to room temperature, concentrate under reduced pressure, redissolve in dichloromethane, wash with saturated sodium bicarbonate, dry over anhydrous sodium sulfate, filter, concentrate, and purify by silica gel column chromatography (DCM:MeOH = 50:1 to 15:1) to obtain the title compound (3.12 g, white solid). LCMS (ES, m/z) 456 [M+H] + ; 1 H NMR (400MHz, CD 3 OD) δ7.71 (dd, 1H), 7.68 (br s,1H),7.58(dd,1H),7.37(dd,1H),7.32(d,1H),7.20(td,1H),7.05(td,1H),5.02(s,2H),4.65(d,2H),4.55(d,2H),4.50 -4.52(m,2H),4.05(s,2H),3.03-2.97(m,2H),2.38(s,3H).
实施例8:N-(3-(((2-(2,3-二氢苯并[f][1,4]硒氮杂-4(5H)-基)-6-甲基喹唑啉-4-基)氨基)甲基)氧杂环丁-3-基)-2,2,2-三氟乙酰胺(I-4)的合成
Example 8: N-(3-(((2-(2,3-dihydrobenzo[f][1,4]selenazepine Synthesis of 4-(5H)-6-methylquinazolin-4-yl)amino)methyl)oxetan-3-yl)-2,2,2-trifluoroacetamide (I-4)
将化合物IIIc(1.2g,3.2mmol)和化合物IIa(810mg,3.80mmol)溶解于乙醇(24mL)中,升温至80℃搅拌反应16小时,冷却至室温,减压浓缩至干,硅胶柱层析纯化(DCM:MeOH=80:1至40:1)得标题化合物I-4(1.3g,白色固体)。LCMS(ES,m/z)552[M+H]+;1HNMR(400MHz,DMSO-d6)δ10.08(brs,1H),8.10-7.71(m,3H),7.57(s,1H),7.43-7.26(m,3H),7.10(s,1H),4.93(s,2H),4.75-4.60(m,4H),4.49(s,2H),4.29-4.12(s,2H),3.09-2.89(m,2H),2.36(s,3H).Compound IIIc (1.2 g, 3.2 mmol) and compound IIa (810 mg, 3.80 mmol) were dissolved in ethanol (24 mL), heated to 80°C and stirred for 16 hours. The mixture was cooled to room temperature and concentrated to dryness under reduced pressure. The mixture was purified by silica gel column chromatography (DCM:MeOH = 80:1 to 40:1) to obtain the title compound I-4 (1.3 g, white solid). LCMS(ES,m/z)552[M+H] + ; 1 HNMR(400MHz,DMSO-d 6 )δ10.08(brs,1H),8.10-7.71(m,3H),7.57(s,1H),7.43-7.26(m,3H),7.10(s,1H),4.93( s,2H),4.75-4.60(m,4H),4.49(s,2H),4.29-4.12(s,2H),3.09-2.89(m,2H),2.36(s,3H).
实施例9:2,2,2-三氟-N-(3-(((6-甲基-2-(1-氧化-2,3-二氢苯并[f][1,4]硒氮杂-4(5H)-基)喹唑啉-4-基)氨基)甲基)氧杂环丁-3-基)乙酰胺(I-5)的合成
Example 9: 2,2,2-trifluoro-N-(3-(((6-methyl-2-(1-oxido-2,3-dihydrobenzo[f][1,4]selenazepine Synthesis of 4-(5H)-quinazolin-4-yl)amino)methyl)oxetan-3-yl)acetamide (I-5)
将化合物I-4(200mg,0.363mmol)溶解于二氯甲烷(10mL)中,加入3-氯过氧苯甲酸(188mg,1.09mmol)于0℃,然后在0℃搅拌2小时。向反应中加入饱和的亚硫酸钠水溶液(20mL)和饱和的碳酸氢钠水溶液(20mL),然后用二氯甲烷(30mL)萃取两次。合并有机相,用饱和食盐水溶液(30mL)洗涤,干燥过无水硫酸钠,减压浓缩,得标题化合物I-5(200mg,黄色固体)。LCMS(ES,m/z)568[M+H]+;1HNMR(400MHz,DMSO-d6)δ10.01(s,1H),8.15(s,1H),7.98-7.89(m,1H),7.82(s,1H),7.74-7.72(m,1H),7.68(d,1H),7.44-7.41(m,1H),7.39-7.38(m,1H),7.27(d,1H),5.07(s,2H),4.75-4.67(m,3H),4.63-4.61(m,2H),4.36-4.21(m,2H),4.16-4.09(m,1H),3.46-3.42(m,1H),3.15-3.06(m,1H),2.35(s,3H).Compound I-4 (200 mg, 0.363 mmol) was dissolved in dichloromethane (10 mL), and 3-chloroperoxybenzoic acid (188 mg, 1.09 mmol) was added at 0°C, followed by stirring at 0°C for 2 hours. Saturated aqueous sodium sulfite solution (20 mL) and saturated aqueous sodium bicarbonate solution (20 mL) were added to the reaction, followed by extraction twice with dichloromethane (30 mL). The organic phases were combined, washed with saturated aqueous sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the title compound I-5 (200 mg, yellow solid). LCMS (ES, m/z) 568 [M+H] + ; 1 HNMR (400 MHz, DMSO-d 6 )δ10.01(s,1H),8.15(s,1H),7.98-7.89(m,1H),7.82(s,1H),7.74-7.7 2(m,1H),7.68(d,1H),7.44-7.41(m,1H),7.39-7.38(m,1H),7.27(d,1H ),5.07(s,2H),4.75-4.67(m,3H),4.63-4.61(m,2H),4.36-4.21(m,2H) ,4.16-4.09(m,1H),3.46-3.42(m,1H),3.15-3.06(m,1H),2.35(s,3H).
实施例10:4-(4-(((3-氨基氧杂环丁-3-基)甲基)氨基)-6-甲基喹唑啉-2-基)-2,3,4,5-四氢苯并[f][1,4]硒氮杂-1-氧化物(I-6)的合成
Example 10: 4-(4-(((3-aminooxetan-3-yl)methyl)amino)-6-methylquinazolin-2-yl)-2,3,4,5-tetrahydrobenzo[f][1,4]selenazepine Synthesis of -1-oxide (I-6)
将化合物I-5(200mg,0.353mmol)溶解于甲醇(4mL)和水(4mL)中,加入氢氧化钠(71mg,1.8mmol)于0℃,然后在60℃搅拌2小时。反应混合物倒入水(10mL)中,再用乙酸乙酯(10mL)萃取。有机层用食盐水洗涤,干燥硫酸钠,过滤。滤液经减压浓缩得到残渣,经C18柱层析纯化(甲醇:水(0.1% TFA)=5%至65%)得到目标化合物I-6(45mg,白色固体)。LCMS(ES,m/z)472[M+H]+;1HNMR(400MHz,CD3OD)δ7.84(d,J=9.2Hz,1H),7.74(s,1H),7.68(d,1H),7.55-7.49(m,2H),7.47-7.42(m,1H),7.39-7.37(m,1H),5.58-5.54(m,1H),4.99-4.93(m,1H),4.92-4.88(m,1H),4.61(d,2H),4.49(d,J=6.4Hz,1H),4.45(d,1H),4.12-4.07(m,1H),4.06-3.97(m,2H),3.96-3.91(m,1H),3.54-3.49(m,1H),2.41(s,3H).Compound I-5 (200 mg, 0.353 mmol) was dissolved in methanol (4 mL) and water (4 mL). Sodium hydroxide (71 mg, 1.8 mmol) was added at 0°C, followed by stirring at 60°C for 2 hours. The reaction mixture was poured into water (10 mL) and extracted with ethyl acetate (10 mL). The organic layer was washed with brine, dried over sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain a residue, which was purified by C18 column chromatography (methanol:water (0.1% TFA) = 5% to 65%) to obtain the target compound I-6 (45 mg, white solid). LCMS (ES, m/z) 472 [M+H] + ; 1 H NMR (400 MHz, CD 3 OD)δ7.84(d,J=9.2Hz,1H),7.74(s,1H),7.68(d,1H),7.55-7.49(m,2H),7.4 7-7.42(m,1H),7.39-7.37(m,1H),5.58-5.54(m,1H),4.99-4.93(m,1H),4.9 2-4.88(m,1H),4.61(d,2H),4.49(d,J=6.4Hz,1H),4.45(d,1H),4.12-4.07( m,1H),4.06-3.97(m,2H),3.96-3.91(m,1H),3.54-3.49(m,1H),2.41(s,3H).
效果实施例1:病毒细胞病理效应(CPE)实验Effect Example 1: Virus Cytopathic Effect (CPE) Experiment
为了测定化合物的抗-RSV活性,96-孔板以每孔6×103个细胞的密度接种,细胞在含有10%胎牛血清(FBS)的Dulbecco改良伊戈尔培养基((DMEM)中。第二天,在系列半对数稀释的化合物存在下,采用充足的RSV长链菌株(LongStrain)(ATCC)感染细胞以在6天后产生约80-90%胞病理效应,每孔的总体积为200μL。6天后,采用细胞计数试剂盒-8(Cell Counting kit-8)测定细胞的存活情况。测定450nm处的吸光度和630nm处的参比值以确定50%的有效浓度(EC50)。To determine the anti-RSV activity of the compounds, 96-well plates were seeded at a density of 6×10 3 cells per well in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS). The next day, cells were infected with sufficient RSV LongStrain (ATCC) to produce approximately 80-90% cytopathic effects after 6 days in the presence of serial half-log dilutions of the compounds in a total volume of 200 μL per well. After 6 days, cell viability was determined using a Cell Counting Kit-8. The absorbance at 450 nm and the reference value at 630 nm were measured to determine the 50% effective concentration (EC 50 ).
表1
Table 1
实验结果表明,在同等的实验条件下,本实施例化合物具有低纳摩尔级的抗RSV病毒活性,其中I-2和I-6的抗RSV病毒活性略优于阳性对照药Ziresovir。The experimental results showed that under the same experimental conditions, the compounds of this example had low nanomolar anti-RSV viral activity, among which I-2 and I-6 had slightly better anti-RSV viral activity than the positive control drug Ziresovir.
效果实施例2:本发明部分化合物的肝微粒代谢研究Effect Example 2: Study on the liver microsome metabolism of some compounds of the present invention
1.测试化合物和对照工作溶液的制备:1. Preparation of test compound and control working solutions:
将待测化合物用DMSO溶解,得10mM的储备液,取5μL化合物储备液(10mM in DMSO)用495μL乙腈稀释,得浓度为100μM中间溶液,99%乙腈。Dissolve the test compound in DMSO to obtain a 10 mM stock solution. Take 5 μL of the compound stock solution (10 mM in DMSO) and dilute it with 495 μL of acetonitrile to obtain a 100 μM intermediate solution with 99% acetonitrile.
2.NADPH辅酶的制备:2. Preparation of NADPH coenzyme:
1)原材料:NADPH·4Na(供应商:BONTAC,Cat.No.BT04)1) Raw materials: NADPH·4Na (supplier: BONTAC, Cat. No. BT04)
2)制备程序:称取适量的NADPH粉末并稀释到2mM的MgCl2溶液中(工作溶液浓度:2mM;在反应系统中的最终浓度:1mM)。2) Preparation procedure: Weigh an appropriate amount of NADPH powder and dilute it into 2 mM MgCl2 solution (working solution concentration: 2 mM; final concentration in the reaction system: 1 mM).
3.肝微粒体的制备3. Preparation of Liver Microsomes
1)原料:人肝微粒体(供应商:Coring,CatNo.452117,每g肝中含约45mg肝微粒蛋白)1) Raw material: Human liver microsomes (supplier: Corning, Cat No. 452117, containing approximately 45 mg of liver microsome protein per gram of liver)
2)制备程序:在100mM磷酸钾缓冲液中制备适当浓度的肝微粒体工作溶液。2) Preparation procedure: Prepare liver microsome working solution of appropriate concentration in 100 mM potassium phosphate buffer.
4.停止溶液的制备:4. Preparation of stopping solution:
使用含有250nM托布利特胺(tolbutamide)和250nM拉贝洛尔(labetalol)作为内标(IS)的冷(4℃)乙腈(ACN)作为停止溶液。Cold (4°C) acetonitrile (ACN) containing 250 nM tolbutamide and 250 nM labetalol as internal standard (IS) was used as the stop solution.
5.实验步骤:5. Experimental steps:
1)使用Apricot自动化工作站,向所有96孔反应板中加入2μL/孔的化合物工作溶液,除了空白(T0,T60和NCF60)。1) Using an Apricot automated workstation, add 2 μL/well of compound working solution to all 96-well reaction plates, except for the blanks (T0, T60, and NCF60).
2)使用Apricot自动化工作站向所有反应板(空白,T0,T60和NCF60)中加入100μL/孔的微粒体溶液。2) Add 100 μL/well of microsomal solution to all reaction plates (blank, T0, T60, and NCF60) using an Apricot automated workstation.
3)所有含有化合物和微粒体混合物的反应板在37℃预孵育10分钟。3) All reaction plates containing compound and microsome mixtures were pre-incubated at 37°C for 10 minutes.
4)使用Aprico自动化工作站向NCF60反应板中加入98μL/孔的100mM磷酸钾缓冲液;4) Add 98 μL/well of 100 mM potassium phosphate buffer to the NCF60 reaction plate using an Aprico automated workstation;
5)NCF60反应板在37℃孵育,并启动计时器1。5) Incubate the NCF60 reaction plate at 37°C and start timer 1.
表2 NCF60孵化
Table 2 NCF60 incubation
6)预孵育后,使用Aprico自动化工作站向每个反应板中加入98μL/孔的NADPH,除了NCF60(空白,T0和T60),以启动反应。6) After pre-incubation, 98 μL/well of NADPH was added to each reaction plate except NCF60 (blank, T0, and T60) using an Aprico automated workstation to start the reaction.
表3培养基中各成分的最终浓度
Table 3 Final concentration of each component in the culture medium
7)反应板在37℃孵育,并启动计时器2。7) Incubate the reaction plate at 37°C and start Timer 2.
表4反应板孵化
Table 4 Reaction plate incubation
8)使用Aprico自动化工作站在适当的终点时间点向每个反应板中加入600μL/孔的停止溶液,以终止反应。8) Use the Aprico automated workstation to add 600 μL/well of stop solution to each reaction plate at the appropriate endpoint time point to terminate the reaction.
9)每个板被密封并摇动10分钟。9) Each plate was sealed and shaken for 10 minutes.
10)摇动后,每个板以4000rpm和4℃的速度离心20分钟。10) After shaking, each plate was centrifuged at 4000 rpm and 4°C for 20 minutes.
11)离心后,使用Aprico自动化工作站将每个反应板的300μL上清液转移至八个新的96孔板中,以进行液相色谱-质谱分析。11) After centrifugation, 300 μL of supernatant from each reaction plate was transferred to eight new 96-well plates using an Aprico automated workstation for liquid chromatography-mass spectrometry analysis.
3.数据分析3. Data Analysis
采用Phoenix WinNonlin7.0计算半衰期(T1/2,单位:h)和肝微粒代谢固有清除率(CLint(mic),单位:μL/min/mg)和肝清除率(CLint(liver),单位:mL/min/kg),结果如下表5。Phoenix WinNonlin 7.0 was used to calculate the half-life (T 1/2 , unit: h), intrinsic clearance of hepatic microsomes (CL int(mic) , unit: μL/min/mg), and hepatic clearance (CL int(liver) , unit: mL/min/kg). The results are shown in Table 5 below.
表5
Table 5
注:如不添加辅酶NADPH(则是用缓冲液替代)。Note: If the coenzyme NADPH is not added (it is replaced by buffer).
实验结果表明,与阳性对照药Ziresovir相比,本实施例化合物具有更长的肝微粒代谢半衰期和更低的代谢清除率。The experimental results show that compared with the positive control drug Ziresovir, the compound of this example has a longer hepatic microsome metabolic half-life and a lower metabolic clearance rate.
效果实施例3本发明部分实施例的体内药代动力学研究Effect Example 3 In vivo pharmacokinetic study of some embodiments of the present invention
1.Caco-2细胞培养1. Caco-2 cell culture
Caco-2细胞来源:American type culture collection(ATCC)。Caco-2 cells source: American type culture collection (ATCC).
培养操作:将Caco-2细胞接种于孔径为0.4μm的聚碳酸酯(PC)膜上,使用96孔Corning插板,接种密度为3.5×104个细胞/cm2。在培养期间每4~5天更换一次培养基,直到第21至28天形成密集的细胞单层膜。Culture Procedure: Caco-2 cells were seeded on polycarbonate (PC) membranes with a pore size of 0.4 μm using 96-well Corning plates at a seeding density of 3.5 × 10 4 cells/cm 2 . The culture medium was changed every 4–5 days until a dense cell monolayer was formed between days 21 and 28.
2.转运方法2.Transfer method
本研究中所用的转运缓冲液为含10.0mM HEPES、pH 7.40±0.05的HBSS。待测化合物以2.00μM浓度进行双向转运实验,每组重复两次;地高辛以10.0μM浓度进行双向转运实验,每组重复两次;而心得安(nadolol)和美托洛尔(metoprolol)均以2.00μM浓度在A至B方向进行重复测试。终浓度DMSO控制在小于1%。放置于37.0℃、5% CO2并保持高湿度(无震荡)条件下的CO2培养箱中孵育2小时。所有样品在加入含有内标的乙腈混合后,以3220xg离心10分钟。起始溶液、供体溶液和接受溶液中的受试化合物及对照化合物浓度,采用LC-MS/MS测定,利用检测物与内标峰面积比(peak area ratio)进行量化。在转运实验结束后,利用荧光黄(lucifer yellow)排阻实验,用以评估Caco-2细胞单层的完整性。The transport buffer used in this study was HBSS containing 10.0 mM HEPES, pH 7.40 ± 0.05. Bidirectional transport experiments were performed at a concentration of 2.00 μM for the test compound, with duplicates per group. Digoxin was tested at a concentration of 10.0 μM for bidirectional transport, with duplicates per group. Both propranolol and metoprolol were tested at a concentration of 2.00 μM in the A-to-B direction. The final DMSO concentration was controlled to less than 1%. The samples were incubated for 2 hours in a CO2 incubator at 37.0°C, 5% CO2 , and high humidity (no shaking). All samples were mixed with acetonitrile containing an internal standard and centrifuged at 3220 x g for 10 minutes. The concentrations of the test compound and control compound in the starting, donor, and receiver solutions were determined by LC-MS/MS, and quantification was performed using the peak area ratio of the test compound to the internal standard. After the transport experiment, the integrity of the Caco-2 cell monolayer was assessed using a lucifer yellow exclusion assay.
3.数据分析3. Data Analysis
使用下式计算表观渗透系数(Papp,cm/s):Papp=(dCr/dt)×Vr/(A×C0)The apparent permeability coefficient (Papp, cm/s) was calculated using the following formula: Papp = ( dCr /dt) x Vr /(A x C0 )
其中:dCr/dt表示随时间变化的接受侧累积浓度变化速率;Vr为接受室中的溶液体积(顶侧为0.0750mL,基侧为0.250mL);A为传输的表面积(单层细胞面积为0.143cm2);C0为给药侧的初始浓度。Where: dCr /dt represents the cumulative concentration change rate of the receiving side over time; Vr is the volume of the solution in the receiving chamber (0.0750 mL for the apical side and 0.250 mL for the basolateral side); A is the surface area of transmission (the monolayer cell area is 0.143 cm2 ); C0 is the initial concentration on the administration side.
外排比(Efflux Ratio)通过下式计算:Efflux Ratio=Papp(BA)/Papp(AB)。The efflux ratio (Efflux Ratio) was calculated by the following formula: Efflux Ratio = P app (BA) / P app (AB).
溶液回收率(%Solution Recovery)计算方式如下:%Solution Recovery=100×[(Vr×Cr)+(Vd×Cd)]/(Vd×C0);The solution recovery rate (% Solution Recovery) was calculated as follows: % Solution Recovery = 100 × [(V r × C r ) + (V d × C d )] / (V d × C 0 );
其中:Vd表示给药室的溶液体积(顶侧为0.0750mL,基侧为0.250mL);Cd、Cr分别为试验结束时给药室(donor)和接受室(receiver)中的化合物浓度。Wherein: Vd represents the volume of the solution in the dosing chamber (0.0750 mL on the apical side and 0.250 mL on the basal side); Cd and Cr are the concentrations of the compound in the donating chamber (donor) and the receiving chamber (receiver) at the end of the experiment, respectively.
结果如下表6所示。
The results are shown in Table 6 below.
实验结果表明,Ziresovir的Papp(BA)与外排比(Efflux Ratio)都远高于化合物I-2和I-6。表明Ziresovir很可能被某些主动外排转运蛋白(如P-gp)强烈外排,因而实际净吸收Papp(AB)易受到影响。与Ziresovir相比,化合物I-2和I-6的Papp(BA)和外排比都显著低于Ziresovir,因此整体上看化合物I-2和I-6具有其外排程度更低,跨膜净吸收受到的外排影响相对更小的优势。Experimental results showed that the P app (BA) and efflux ratio (Efflux Ratio) of ziresovir were significantly higher than those of compounds I-2 and I-6. This suggests that ziresovir is likely strongly effluxed by certain active efflux transporters (such as P-gp), thus easily affecting the actual net absorption P app (AB). Compared with ziresovir, compounds I-2 and I-6 both had significantly lower P app (BA) and efflux ratios. Overall, compounds I-2 and I-6 have the advantage of lower efflux levels and relatively less efflux-induced net transmembrane absorption.
效果实施例4本发明部分实施例的体内药代动力学研究Effect Example 4 In vivo pharmacokinetic study of some embodiments of the present invention
1、供试品配制1. Preparation of test samples
表7
Table 7
2、实验动物2. Experimental Animals
种属:健康雄性SD(Sprague Dawley)大鼠(SPF级),体重180~220g。Species: Healthy male Sprague Dawley rats (SPF grade), weighing 180-220 g.
来源:上海市计划生育科学研究所实验动物经营部,动物转移自实验机构动物储备库(999M-017)。Source: Experimental Animal Management Department of Shanghai Institute of Family Planning Sciences. Animals were transferred from the experimental institution animal reserve (999M-017).
数量:雄性18只;动物挑选:随机分组,每组3只Number: 18 males; Animal selection: random grouping, 3 animals per group
3、给药方式与采血时间点3. Dosage method and blood collection time
给药前称重,根据体重,计算给药量。通过静脉注射或灌胃口服给药。Weigh the patient before administration and calculate the dosage based on body weight. Administer the drug by intravenous injection or oral gavage.
静脉注射给药(2mg/kg)后,于给药药后0.033h,0.25h,0.5h,1h,2h,4h,8h和24h经大鼠颈静脉取血0.2ml,EDTA-K2抗凝,采集后放置冰上。After intravenous administration (2 mg/kg), 0.2 ml of blood was collected from the rat jugular vein at 0.033 h, 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 8 h and 24 h after administration, anticoagulated with EDTA-K2, and placed on ice.
口服灌胃给药(10mg/kg)后,于给药药后0.25h,0.5h,1h,2h,4h,6h,8h和24h经大鼠颈静脉取血0.2ml,EDTA-K2抗凝,采集后放置冰上。After oral gavage administration (10 mg/kg), 0.2 ml of blood was collected from the rat jugular vein at 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after administration, anticoagulated with EDTA-K2, and placed on ice after collection.
4、血浆样品处理4. Plasma sample processing
1)分别向96孔板中加入15μL未知样品、校准标准、质控品和稀释质控品(如果有)、单空白和双空白样品;1) Add 15 μL of unknown sample, calibration standard, quality control and diluted quality control (if any), single blank and double blank samples to a 96-well plate;
2)每个样品(双空白除外)分别用150μL的IS1淬灭(双空白样品用150μL的乙腈淬灭),然后混合液在800rpm搅拌10分钟,然后在3220×g、4℃离心15分钟;2) Each sample (except the double blank) was quenched with 150 μL of IS1 (the double blank sample was quenched with 150 μL of acetonitrile), and the mixture was stirred at 800 rpm for 10 minutes and then centrifuged at 3220 × g and 4°C for 15 minutes;
3)转移55μL上清液到另一个干净的96孔板中,再次在3220×g、4℃离心5分钟,然后上清液直接进行LC-MS/MS分析。3) Transfer 55 μL of the supernatant to another clean 96-well plate and centrifuge again at 3220 × g and 4°C for 5 minutes. The supernatant is then directly subjected to LC-MS/MS analysis.
该过程在湿冰上进行。The procedure was performed on wet ice.
5、样品分析5. Sample analysis
1)UPLC-MS/MS分析方法:1) UPLC-MS/MS analysis method:
表8
Table 8
6、数据处理6. Data processing
通过不同时间点的血药浓度数据,运用Phoenix WinNonlin7.0软件非房室模型计算药代动力学参数,提供AUC0-int、Cmax、Tmax、T1/2和口服生物利用度(F%)等参数。结果如下表9所示。The pharmacokinetic parameters were calculated using the non-compartmental model using Phoenix WinNonlin 7.0 software using the plasma drug concentration data at different time points, providing parameters such as AUC 0-int , C max , T max , T 1/2 , and oral bioavailability (F%). The results are shown in Table 9 below.
表9
Table 9
实验结果表明,与Ziresovir相比,化合物I-2具有更好的口服生物利用度,无论是注射给药还是口服给药,其AUC显著提高,具有代谢清除率更低、平均驻留时间(MRT)更长的优势。因此,本发明用于制作治疗呼吸道合胞病毒毒感染相关疾病的药物具有非常好的前景。The experimental results showed that compared with ziresovir, compound I-2 has better oral bioavailability, with a significantly improved AUC for both injection and oral administration, and has the advantages of lower metabolic clearance and longer mean residence time (MRT). Therefore, the present invention has very good prospects for the preparation of drugs for treating diseases related to respiratory syncytial virus infection.
上述实施例仅是为清楚地说明所作的举例,而并非对实施方式的限定。对于所属领域的普通技术人员来说,在上述说明的基础上还可以做出其它不同形式的变化或变动。这里无需也无法对所有的实施方式予以穷举。而由此所引伸出的显而易见的变化或变动仍处于本发明创造的保护范围之中。The above embodiments are provided for illustrative purposes only and are not intended to limit the scope of implementation. Those skilled in the art will readily appreciate that other variations or modifications based on the above descriptions are possible. It is not necessary and impossible to provide an exhaustive list of all implementations. Obvious variations or modifications arising therefrom remain within the scope of protection of the present invention.
Claims (11)
A benzoselenazepine as shown in formula I a compound or a pharmaceutically acceptable salt thereof,
The benzoselenazepine of formula I as claimed in claim 1 The compound or a pharmaceutically acceptable salt thereof is characterized in that the benzoselenazepine shown in formula I The compound is any of the following compounds:
A benzoselenazepine as shown in formula I according to any one of claims 1 to 5 The preparation method of the compound is characterized in that it comprises the following steps: in an organic solvent, in the presence of an acidic reagent, subjecting the compound represented by formula II to a substitution reaction as shown below with a benzopyrimidine compound represented by formula III to obtain the benzoselenazepine represented by formula I. Compounds,
A compound as shown in formula II,
A method for preparing a compound as shown in Formula IIa, characterized in that it comprises the following steps:
A compound shown below:
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202410287668 | 2024-03-13 | ||
| CN202410287668.2 | 2024-03-13 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025190151A1 true WO2025190151A1 (en) | 2025-09-18 |
Family
ID=97062801
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2025/081112 Pending WO2025190151A1 (en) | 2024-03-13 | 2025-03-06 | Benzoselenazole compounds as well as preparation method therefor and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025190151A1 (en) |
-
2025
- 2025-03-06 WO PCT/CN2025/081112 patent/WO2025190151A1/en active Pending
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| DK2776416T3 (en) | Pharmaceutical compounds | |
| TW202115085A (en) | Antiviral heterocyclic compounds | |
| CN109641042B (en) | Heterocyclic derivatives for the treatment of RSV | |
| JP2017165751A (en) | Inhibitors of hepatitis b virus covalently closed circular dna formation and their method of use | |
| WO2019011323A1 (en) | Endocyclic thiamidinoamide-arylamide compound and use thereof for treating hepatitis b | |
| TW201036959A (en) | Novel compounds 660 | |
| WO2018121689A1 (en) | Sulfonamide-aryl amide compound and use thereof as drug for treating hepatitis b | |
| TW202345818A (en) | Antiviral heterocyclic compounds | |
| CA3173354A1 (en) | A 2,3-dihydrofuro[2,3-c]pyridine-3-carboxamide compound and its antiviral use | |
| CN113321694A (en) | N4-hydroxycytidine derivative and preparation method and application thereof | |
| WO2022211812A1 (en) | Antiviral heterocyclic compounds | |
| CN110483487B (en) | 2-thiomethylpyrazole pyrimidone compound, preparation method thereof, pharmaceutical composition and application | |
| WO2025190151A1 (en) | Benzoselenazole compounds as well as preparation method therefor and use thereof | |
| US20250090491A1 (en) | Antiviral 1,3-di-oxo-indene compounds | |
| CN114728973A (en) | Crystal form of nucleoprotein inhibitor and application thereof | |
| BR112021013730A2 (en) | INTERNAL CYCLIC SULFAMIDINE AMIDE-ARILAMIDE COMPOUND AND USE OF IT FOR HEPATITIS B TREATMENT | |
| WO2024078618A1 (en) | Crystal form of cyano-substituted polypeptide compound and preparation method therefor | |
| US9790213B2 (en) | Imidazoles for the treatment and prophylaxis of respiratory syncytial virus infection | |
| CN110041273B (en) | 2-(2-Chloro-4-methylphenyl)quinazoline-4(3H)-one compounds and their medicinal uses | |
| CN115197164B (en) | Novel thiazole compound and its preparation method and use | |
| WO2024131886A1 (en) | Benzothiazepine compound, and preparation method therefor and use thereof | |
| AU2019218150B2 (en) | Crystal form of 3,4-dihydrothieno[3,2-d]pyrimidine compound and preparation method therefor | |
| WO2024098856A1 (en) | Anti-influenza-virus derivatives and use thereof | |
| WO2025214429A1 (en) | Diaminopyrimidine compound and use thereof | |
| TW202214572A (en) | Synthesis of substituted arylmethylureas, analogues, and crystalline forms thereof and methods of using same |
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: 25769322 Country of ref document: EP Kind code of ref document: A1 |