CN117337286A - Methylsulfonyl urea compound and application thereof - Google Patents

Methylsulfonyl urea compound and application thereof Download PDF

Info

Publication number
CN117337286A
CN117337286A CN202280033410.9A CN202280033410A CN117337286A CN 117337286 A CN117337286 A CN 117337286A CN 202280033410 A CN202280033410 A CN 202280033410A CN 117337286 A CN117337286 A CN 117337286A
Authority
CN
China
Prior art keywords
compound
mol
added
water
stirred
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280033410.9A
Other languages
Chinese (zh)
Inventor
夏建华
贺海鹰
江志赶
张晓�
黎健
陈曙辉
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medshine Discovery Inc
Original Assignee
Medshine Discovery Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medshine Discovery Inc filed Critical Medshine Discovery Inc
Publication of CN117337286A publication Critical patent/CN117337286A/en
Pending legal-status Critical Current

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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic 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
    • C07D417/02Heterocyclic 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 two hetero rings
    • C07D417/04Heterocyclic 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 two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic 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
    • C07D417/14Heterocyclic 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The invention discloses a methylsulfonyl urea compound. Specifically disclosed are compounds represented by formula (I) and pharmaceutically acceptable salts thereof.

Description

Methylsulfonyl urea compound and application thereof
The present application claims priority as follows:
CN2021104946968, filing date: 2021, 5 and 7.
Technical Field
The invention relates to a methylsulfonyl urea compound. In particular to a compound shown in a formula (I) and pharmaceutically acceptable salts thereof.
Background
Hepatitis b virus belongs to the family of hepatiridae. It may cause acute and/or persistent progressive chronic diseases. Hepatitis b virus can also cause many other clinical manifestations in pathological forms, especially chronic inflammation of the liver, cirrhosis and cancer of hepatocytes. In addition, co-infection with hepatitis D can have adverse effects during the course of the disease.
Hepatitis B is a worldwide medical problem, no specific medicine for treating hepatitis B exists worldwide at present, the current first-line medicine for treating hepatitis B is mainly nucleoside medicines and interferon medicines, but the medicines cannot be thoroughly cured, all the medicines need to be taken for a long time, and the problems of renal insufficiency, lactic acidosis and the like exist, so that the development of a novel anti-hepatitis B medicine which is more effective and safer for patients is imperative.
Disclosure of Invention
The invention provides a compound shown as a formula (I) or pharmaceutically acceptable salt thereof,
wherein,
R 1 are each independently selected from halogen, OH, CN, NH 2 、C 1-3 Alkyl and C 1-3 Alkoxy group, the C 1-3 Alkyl groupC 1-3 Alkoxy groups are each independently optionally substituted with 1, 2 or 3 halogens;
m is selected from 0, 1, 2, 3 and 4;
T 1 and T 2 Each independently selected from CH and N;
is a single bond or a double bond;
R 2 selected from H, methyl and F;
ring A is selected from phenyl and C 4-10 Cycloalkyl, the phenyl and C 4-10 Cycloalkyl groups are each independently optionally substituted with 1, 2 or 3R a Substitution;
R a are each independently selected from halogen, OH, CN, NH 2 、C 1-3 Alkyl and C 1-3 Alkoxy group, the C 1-3 Alkyl and C 1-3 Alkoxy groups are each independently optionally substituted with 1, 2 or 3 halogens;
L 1 selected from single bond, -C.ident.C-and-CR 2 =;
L 2 Selected from single bonds and methylene groups.
In some aspects of the invention, R is as defined above 1 Are respectively and independently selected from F, cl, br, CN and-CH 3 and-OCH 3 the-CH 3 and-OCH 3 Each independently optionally substituted with one, 2 or 3F, and the other variables are as defined herein.
In some aspects of the invention, R is as defined above 1 Each independently selected from F, cl, br and CN, with the other variables being as defined herein.
In some aspects of the invention, L as described above 1 Selected from-c≡c-, -ch=, -C (F) =, and-C (CH) 3 ) =, other variables are defined as in the present invention.
In some aspects of the inventionR is as described above a Are respectively and independently selected from F, cl, br, CH 3 And OCH 3 The CH is 3 And OCH 3 Each independently optionally substituted with 1, 2 or 3F, and the other variables are as defined herein.
In some aspects of the invention, the structural units described aboveSelected from the group consisting ofThe other variables are as defined herein.
In some aspects of the invention, the structural units described aboveSelected from the group consisting of The other variables are as defined herein.
In some embodiments of the invention, the ring A is selected from phenyl, cyclobutyl, cyclohexyl,The phenyl, cyclobutyl, cyclohexyl,Are each independently optionally substituted with 1, 2 or 3R a Instead, the other variables are as defined herein.
In some embodiments of the invention, the ring A is selected fromThe other variables are as defined herein.
In some aspects of the invention, the structural units described aboveSelected from the group consisting of The other variables are as defined herein.
In some aspects of the invention, the structural units described aboveSelected from the group consisting of The other variables are as defined herein.
In some embodiments of the invention, the above compound, or a pharmaceutically acceptable salt thereof, is selected from the group consisting of:
wherein R is 1 、R 2 、L 2 And m is as defined in any one of the present inventions.
In some embodiments of the invention, the above compound, or a pharmaceutically acceptable salt thereof, is selected from the group consisting of:
wherein R is 1 、R 2 、L 2 And m is as defined in any one of the present inventions.
In some embodiments of the invention, the above compound, or a pharmaceutically acceptable salt thereof, is selected from the group consisting of:
wherein R is 1 、R 2 、L 2 And m is as defined in any one of the present inventions.
Still other embodiments of the present invention are derived from any combination of the variables described above.
The present invention also provides the following compounds, or pharmaceutically acceptable salts thereof, selected from:
the present invention also provides the following compounds, or pharmaceutically acceptable salts thereof, selected from:
the present invention also provides the following compounds, or pharmaceutically acceptable salts thereof, selected from:
the present invention also provides the following compounds, or pharmaceutically acceptable salts thereof, selected from:
the invention also provides application of the compound or the pharmaceutically acceptable salt thereof in preparing medicaments for treating diseases related to hepatitis B virus infection.
In some aspects of the invention, the hepatitis B virus infection-associated disease is chronic hepatitis B.
Definition and description
The following terms and phrases used herein are intended to have the following meanings unless otherwise indicated. A particular term or phrase, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense. When trade names are presented herein, it is intended to refer to their corresponding commercial products or active ingredients thereof.
The term "pharmaceutically acceptable" as used herein refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salts" refers to salts of the compounds of the present application prepared from the compounds found herein to have a particular substituent with a relatively non-toxic acid or base. When relatively acidic functionalities are included in the compounds of the present application, base addition salts may be obtained by contacting such compounds with a sufficient amount of base in pure solution or in a suitable inert solvent. When the compounds of the present invention contain relatively basic functional groups, the acid addition salts may be obtained by contacting such compounds with a sufficient amount of acid in pure solution or in a suitable inert solvent. Certain specific compounds of the invention contain basic and acidic functionalities that can be converted to either base or acid addition salts.
Pharmaceutically acceptable salts of the invention can be synthesized from the parent compound containing an acid or base by conventional chemical methods. In general, the preparation of such salts is as follows: prepared via reaction of these compounds in free acid or base form with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of both.
Unless otherwise indicated, the term "isomer" is intended to include geometric isomers, cis-trans isomers, stereoisomers, enantiomers, optical isomers, diastereomers and tautomers.
The compounds of the invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, and racemic mixtures and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers and mixtures thereof are included within the scope of the present invention.
Unless otherwise indicated, the term "enantiomer" or "optical isomer" refers to stereoisomers that are mirror images of each other.
Unless otherwise indicated, the term "cis-trans isomer" or "geometric isomer" is caused by the inability of a double bond or a single bond of a ring-forming carbon atom to rotate freely.
Unless otherwise indicated, the term "diastereoisomer" refers to stereoisomers of a molecule having two or more chiral centers and having a non-mirror relationship between the molecules.
Unless otherwise indicated, "(+)" means dextrorotation, "(-)" means levorotatory, "(±)" means racemization.
Unless otherwise indicated, with solid wedge bondsAnd a wedge-shaped dotted bondRepresenting the absolute configuration of a solid centre by straight solid keysAnd straight dotted line keyRepresenting the relative configuration of the stereogenic centers, e.g.Indicating that Rx and Ry are co-directional and comprisingTwo configurations.
Unless otherwise specified, when a group has one or more bondable sites, any one or more of the sites of the group may be bonded to other groups by chemical bonds. When the connection mode of the chemical bond is not positioned and the H atoms exist in the connectable site, the number of the H atoms of the site can be correspondingly reduced to be changed into the corresponding valence group along with the number of the connected chemical bond when the chemical bond is connected. The chemical bond of the site and other groups can be a straight solid line bondStraight dotted line keyOr wave linesAnd (3) representing. For example, a straight solid bond in-OCH 3 indicates that it is attached to other groups through an oxygen atom in that group;the straight dashed bonds in (a) represent the attachment to other groups through both ends of the nitrogen atom in the group; by straight broken double bonds Representing the linkage between groups by double bonds, e.g.The double bond of the straight-line indicates that the cyclohexyl group is doubly bonded to other groups.
The compounds of the invention may be present in particular. Unless otherwise indicated, the term "tautomer" or "tautomeric form" refers to the fact that at room temperature, different functional group isomers are in dynamic equilibrium and are capable of rapid interconversion. If tautomers are possible (e.g., in solution), chemical equilibrium of the tautomers can be reached. For example, proton tautomers (also known as proton tautomers) (prototropic tautomer) include interconversions by proton transfer, such as keto-enol isomerisation and imine-enamine isomerisation. Valence isomer (valance tautomer) includes the interconversion by recombination of some of the bond-forming electrons. A specific example of where keto-enol tautomerization is the interconversion between two tautomers of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one.
Unless otherwise indicated, the terms "enriched in one isomer", "enriched in one enantiomer" or "enantiomerically enriched" mean that the content of one isomer or enantiomer is less than 100% and the content of the isomer or enantiomer is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or greater than or equal to 96%, or greater than or equal to 97%, or greater than or equal to 98%, or greater than or equal to 99%, or greater than or equal to 99.5%, or greater than or equal to 99.6%, or greater than or equal to 99.7%, or greater than or equal to 99.8%, or greater than or equal to 99.9%.
Unless otherwise indicated, the term "isomer excess" or "enantiomeric excess" refers to the difference between the relative percentages of two isomers or enantiomers. For example, where one isomer or enantiomer is present in an amount of 90% and the other isomer or enantiomer is present in an amount of 10%, the isomer or enantiomer excess (ee value) is 80%.
Optically active (R) -and (S) -isomers and D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one enantiomer of a compound of the invention is desired, it may be prepared by asymmetric synthesis or derivatization with chiral auxiliary wherein the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomer. Alternatively, when the molecule contains a basic functional group (e.g., amino) or an acidic functional group (e.g., carboxyl), a diastereomeric salt is formed with an appropriate optically active acid or base, and then the diastereomeric resolution is carried out by conventional methods well known in the art, and then the pure enantiomer is recovered. Furthermore, the separation of enantiomers and diastereomers is usually accomplished by using chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization), for example, to form carbamates from amines.
The compounds of the present invention may contain non-natural proportions of atomic isotopes on one or more of the atoms comprising the compounds. For example, compounds can be labeled with radioisotopes, such as tritium @, for example 3 H) Iodine-125% 125 I) Or C-14% 14 C) A. The invention relates to a method for producing a fibre-reinforced plastic composite For example, deuterium can be substituted for hydrogen to form a deuterated drug, and the bond between deuterium and carbon is stronger than the bond between normal hydrogen and carbon, so that the deuterated drug has the advantages of reducing toxic and side effects, increasing the stability of the drug, enhancing the curative effect, prolonging the biological half-life of the drug and the like compared with the non-deuterated drug. All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
Unless otherwise specified, the term "C 1-3 Alkyl "is used to denote a straight or branched saturated hydrocarbon group consisting of 1 to 3 carbon atoms. The C is 1-3 Alkyl includes C 1-2 And C 2-3 Alkyl groups, etc.; it may be monovalent (e.g., methyl), divalent (e.g., methylene), or multivalent (e.g., methine). C (C) 1-3 Examples of alkyl groups include, but are not limited to, methyl (Me), ethylRadical (Et), propyl (including n-propyl and isopropyl), and the like.
Unless otherwise specified, the term "C 1-3 Alkoxy "means those alkyl groups containing 1 to 3 carbon atoms that are attached to the remainder of the molecule through one oxygen atom. The C is 1-3 Alkoxy includes C 1-2 、C 2-3 、C 3 And C 2 Alkoxy groups, and the like. C (C) 1-3 Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy and isopropoxy), and the like.
Unless otherwise specified, "C 4-10 Cycloalkyl "means a saturated cyclic hydrocarbon group consisting of 4 to 10 carbon atoms, which includes monocyclic, bicyclic, and tricyclic ring systems, wherein bicyclic and tricyclic ring systems include spiro, fused, and bridged rings. The C is 4-10 Cycloalkyl includes C 4-5 、C 5-6 、C 6-7 、C 6-8 、C 6-9 、C 6 、C 7 、C 8 、C 9 Or C 10 Etc.; it may be monovalent, divalent or multivalent. C (C) 4-10 Examples of cycloalkyl groups include, but are not limited to, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, [ 2.2.2.2]Bicyclo octane, [4.4.0 ]]Bicyclodecane, etc.
Unless otherwise specified, the term "halo" or "halogen" by itself or as part of another substituent means a fluorine, chlorine, bromine or iodine atom.
The term "optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is substituted with a substituent, which may include deuterium and variants of hydrogen, provided that the valence of the particular atom is normal and the substituted compound is stable. When the substituent is oxygen (i.e., =o), it means that two hydrogen atoms are substituted. Oxygen substitution does not occur on the aromatic group. The term "optionally substituted" means that the substituents may or may not be substituted, and the types and numbers of substituents may be arbitrary on the basis that they can be chemically achieved unless otherwise specified.
When any variable (e.g., R) occurs more than once in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 0 to 2R, the group may optionally be substituted with up to two R's, and R's in each case have independent options. Furthermore, combinations of substituents and/or variants thereof are only permissible if such combinations result in stable compounds.
When the number of one linking group is 0, such as- (CRR) 0 -it is meant that the linking group is a single bond.
When one of the variables is selected from a single bond, the two groups to which it is attached are indicated as being directly linked, e.g., when L in A-L-Z represents a single bond, it is indicated that the structure is actually A-Z.
When a substituent is absent, it is meant that the substituent is absent, e.g., X in A-X is absent, meaning that the structure is actually A. When the exemplified linking group does not indicate its linking direction, its linking direction is arbitrary, for example,the linking group L is-M-W-, in which case-M-W-may be a group formed by linking the rings A and B in the same direction as the reading order from left to rightThe ring A and the ring B may be connected in a direction opposite to the reading order from left to right The linking group, the substitutionCombinations of bases and/or variants thereof are permissible only if such combinations result in stable compounds.
The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, embodiments formed by combining with other chemical synthetic methods, and equivalent alternatives well known to those skilled in the art, preferred embodiments including but not limited to the examples of the present invention.
The compounds of the present invention may be structured by conventional methods well known to those skilled in the art, and if the present invention relates to the absolute configuration of a compound, the absolute configuration may be confirmed by conventional means in the art. For example, single crystal X-ray diffraction (SXRD), the grown single crystal is collected from diffraction intensity data using a Bruker D8 vent diffractometer, and the light source is cukα radiation, scanning:after scanning and collecting the relevant data, the absolute configuration can be confirmed by further analyzing the crystal structure by a direct method (Shellxs 97).
The solvent used in the present invention is commercially available. The invention adopts the following abbreviations: eq represents equivalent, equivalent; m represents mol/L; DMF represents N, N-dimethylformamide; DMSO represents dimethylsulfoxide; etOH stands for ethanol; meOH represents methanol; CBz represents benzyloxycarbonyl, which is a protecting group for an amine; boc represents that t-butoxycarbonyl is an amine protecting group; IPA represents isopropanol and dppf represents 1,1' -bisdiphenylphosphino ferrocene.
Compounds are either prepared according to the general nomenclature of the art or are usedSoftware naming, commercial compounds are referred to by vendor catalog names. Technical effects
As a new type of anti-hepatitis B medicine, the compound has obvious inhibition effect on HBV, has no obvious cytotoxicity in various liver cells, has high bioavailability of oral administration in mice, has high liver-blood ratio of the medicine, and has obvious antiviral effect in an HDI drug effect model.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limiting in any way. The present invention has been described in detail herein, and specific embodiments thereof are also disclosed, it will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the invention without departing from the spirit and scope of the invention.
Example 1
The synthetic route is as follows:
step 1: synthesis of Compound 1-b
To a round bottom flask was added compound 1-a (300 g,2.36mol,243.90mL,1 eq), ethylene glycol (292.85 g,4.72mol,263.83mL,2 eq) and toluene (1.5L) under nitrogen followed by p-toluene sulfonic acid (40.63 g,235.92mmol,0.1 eq) and refluxed at 120℃for 12 hours while water was separated by a water separator. The reaction mixture was washed with water (300 mL. Times.2). The aqueous phase was extracted with ethyl acetate (100 mL). The combined organic phases were washed with saturated brine (300 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The concentrate was separated by column chromatography on silica gel (petroleum ether: ethyl acetate=100:0 to 50:50) to give compound 1-b.
1 H NMR(400MHz,CDCl 3 )δppm7.72(d,J=3.1Hz,1H),7.30-7.08(m,1H),4.11-4.02(m,2H),3.99-3.92(m,2H),1.77(s,3H).
Step 2: synthesis of Compound 1-c
Compound 1-b (20 g,116.81mmol,1 eq) was dissolved in tetrahydrofuran (360 mL), cooled to-78deg.C, and butyllithium (2.5M, 51.40mL,1.1 eq) was slowly added dropwise. The reaction solution was stirred at-78℃for 1 hour, then a solution of carbon tetrabromide (42.61 g,128.49mmol,1.1 eq) in tetrahydrofuran (40 mL) was slowly added dropwise, the reaction solution was stirred at-78℃for 0.5 hour, and the temperature was raised to 0℃and stirred for 0.5 hour. To the reaction mixture was added dropwise a saturated ammonium chloride solution (300 mL), followed by extraction with ethyl acetate (300 mL. Times.3). The combined organic phases were washed with saturated brine (300 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/1) to give compound 1-c.
MS m/z(ESI):249.8[M+1] + .
1 H NMR(400MHz,CDCl 3 )δppm 7.65(s,1H),4.12-4.07(m,2H),4.03-3.98(m,2H),1.79(s,3H).
Step 3: synthesis of Compound 1-d
In a three-necked flask, compound 1-c (49.1 g,196.31mmol,1 eq) and acetone (280 mL) were added, and the mixture was reacted at 25℃with stirring for 2 hours while adding an aqueous hydrochloric acid solution (6M, 196.31mL,6 eq) at 0 ℃. Water (200 mL) was added to the reaction mixture, the mixture was extracted with ethyl acetate (200 mL. Times.3), and the combined organic phases were washed with saturated brine (200 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound 1-d.
1 H NMR(400MHz,CDCl 3 )δppm 7.87(s,1H),2.65(d,J=0.8Hz,3H).
Step 4: synthesis of Compound 1-e
In a three-necked flask, compound 1-d (22 g,106.76mmol,1 eq) and methanol (300 mL) were added followed by slow addition of sodium methoxide in methanol at 0deg.CSolution (5M, 42.71mL,2 eq) and dimethyl oxalate (18.91 g,160.15mmol,1.5 eq). The mixture was stirred at 70℃for 1 hour. The reaction mixture was cooled to 0℃and water (300 mL) was added thereto, followed by pH=2 to 3 with 2mol/L dilute hydrochloric acid, and the mixture was stirred at 0℃for 30 minutes and then filtered. The cake was washed with methanol (30 mL. Times.3) and dried under reduced pressure. Compound 1-e is obtained. MS m/z (ESI): 291.9[ M+1 ]] + .
1 H NMR(400MHz,DMSO-d 6 )δppm 7.92(s,1H),6.55(s,1H),3.69(s,3H).
Step 5: synthesis of Compound 1-f
Into a round bottom flask was charged compound 1-e (65 g,222.52mmol,1 eq), sulfonamide (32.08 g,333.78mmol,19.93mL,1.5 eq) and hydrochloric acid/methanol solution (4M, 500mL,8.99 eq). The mixture was stirred at 70℃for 2 hours. The reaction solution was cooled to 0 ℃, stirred for 1 hour and then filtered. Water (300 mL) was added to the filter cake, stirred for 30 minutes and filtered. To the filter cake was added tert-butyl methyl ether (200 mL), stirred for 30 minutes and filtered. And (5) drying the filter cake under reduced pressure to obtain the compound 1-f. MS m/z (ESI): 351.9[ M+1 ]] + .
1 H NMR(400MHz,DMSO-d 6 )δppm 8.05(s,1H),6.84(s,1H),3.81(s,3H).
Step 6: synthesis of Compound 1-g
Compounds 1-f (20 g,56.79mmol,1 eq) and DMF (200 mL) were added under nitrogen to a three-necked flask, dissolved by stirring, and sodium hydrogen (2.95 g,73.82mmol,60% purity, 1.3 eq) was added in portions at 0deg.C. The reaction mixture was stirred at 0deg.C for 30 min, then methyl iodide (24.18 g,170.37mmo1, 10.61mL,3 eq) was added. The mixture was stirred at 50℃for 12 hours. The reaction solution was slowly poured into ice water (200 mL), ph=2 to 3 was adjusted with 1M diluted hydrochloric acid, and the mixture was stirred for 15 minutes and then filtered. The cake was washed with water (50 mL) and methanol (20 mL. Times.2), and dried under reduced pressure. Compound 1-g was obtained.
MS m/z(ESI):365.7[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δppm 8.35(s,1H),7.37(s,1H),3.95(s,3H),3.60(s,3H).
Step 7: synthesis of Compounds 1-h
Compound 1-g (50 g,136.53mmol,1 eq) and triethylamine (69.08 g,682.67mmol,95.02mL,5 eq) were added to acetonitrile (250 mL) and water (250 mL), and the mixture was stirred at 50℃for 1 hour. Water (200 mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (200 mL. Times.2). The combined organic phases were washed successively with water (100 mL) and saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Compound 1-h was obtained.
MS m/z(ESI):351.8[M+1] + .
Step 8: synthesis of Compound 1-j
Compound 1-i (15 g,77.51mmol,1 eq) was dissolved in methanol (450 mL) and acetic acid (18 mL), reduced iron powder (15.00 g,268.60mmol,3.47 eq) was added in portions, and the reaction mixture was warmed to 65℃and stirred for 2 hours. The reaction solution was cooled to 25 ℃ and filtered, and the filtrate was concentrated under reduced pressure to give a crude product. To the crude product, ethyl acetate (300 mL) was added, followed by washing with 1N aqueous sodium hydroxide (300 mL. Times.2) and saturated brine (300 mL. Times.2), and the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give compound 1-j.
1 H NMR(400MHz,CDCl 3 )δ=6.47-6.41(m,1H),6.41-6.33(m,1H),3.71(br s,2H).
Step 9: synthesis of Compound 1-k
Compound 1-h (15 g,42.59mmol,1 eq), compound 1-j (6.97 g,42.59mmol,1 eq), N, N-diisopropylethylamine (16.51 g,127.77mmol,22.26mL,3 eq) and tri-N-propylcyclophosphatemic anhydride in 50% ethyl acetate (40.66 g,63.89mmol,38.00mL,1.5 eq) were dissolved in dichloromethane (450 mL) at 0deg.C, the reaction was replaced three times with nitrogen, and then warmed to 25deg.C and stirred for reaction for 12 hours. Water (500 mL) was added to the reaction mixture, the mixture was extracted with methylene chloride (500 mL. Times.2), and the organic phases were combined, washed with saturated brine (500 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/3) to give compound 1-k.
MS m/z(ESI):498.7[M+1] + .
Step 10: synthesis of Compound 1-l
Compound 1-k (6.87 g,13.80mmol,1 eq) was dissolved in ethanol (100 mL), the reaction solution was cooled to 0deg.C, then sodium borohydride (1.04 g,27.61mmol,2 eq) was added slowly in portions, and the reaction was stirred for 2 hours. To the reaction solution, 1M aqueous hydrochloric acid solution was slowly added to adjust ph=1 to 2, water (100 mL) was further added, and the mixture was stirred for 5 minutes and then filtered. Methanol (20 mL) was added to the filter cake and the mixture was slurried for 30 minutes, filtered, and the filter cake was dried in vacuo to give Compound 1-l.
MS m/z(ESI):502.8[M+1] + .
Step 11: synthesis of Compound 1-o
2, 6-tetramethylpiperidine (498.33 mg,3.53mmol, 598.95. Mu.L, 1.2 eq) was dissolved in tetrahydrofuran (6 mL), the system was cooled to-30℃and n-butyllithium (2.5M, 1.4mL,1.19 eq) was added dropwise and the reaction stirred at-30℃for 0.5 hours. Then the reaction was cooled to-78℃and a solution of compound 1-n (787.81 mg,2.94mmol,1 eq) in tetrahydrofuran (6 mL) was added dropwise, and stirring was carried out at-78℃for 30min, followed by addition of compound 1-m (0.5 g,2.94mmol, 467.29. Mu.L, 1 eq) in tetrahydrofuran (6 mL). The reaction was slowly warmed to 25℃and stirred at 25℃for 12 hours. The reaction system was cooled to 0℃and saturated ammonium chloride solution (5 mL) was added dropwise, the reaction solution was stirred at 0℃for 1 hour, the filtrate was concentrated under reduced pressure after filtration, water (20 mL) and ethyl acetate (20 mL) were added for extraction, and the organic phase was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product. The crude product was purified by flash column chromatography (eluent: ethyl acetate/petroleum ether, gradient: 0-20%). Compound 1-o is obtained.
1 H NMR(400MHz,CDCl 3 )δ=5.08(s,1H),4.13(q,J=7.1Hz,2H),3.18-3.09(m,1H),2.49(tt,J=3.7,10.9Hz,1H),2.40-2.33(m,1H),2.20(dt,J=3.9,12.6Hz,1H),2.12-1.99(m,3H),1.72-1.58(m,2H),1.30-1.23(m, 15H).
Step 12: synthesis of Compound 1-p
Under nitrogen, 1-l (100 mg, 199.30. Mu. Mol,1 eq), 1-o (87.95 mg, 298.95. Mu. Mol,1.5 eq), sodium carbonate (63.37 mg, 597.90. Mu. Mol,3 eq), dioxane (3 mL) and water (0.5 mL) were added to a thumb flask, followed by tetrakis (triphenylphosphine) palladium (23.03 mg, 19.93. Mu. Mol,0.1 eq). The reaction was stirred at 90℃for 3 hours. Water (10 mL) and ethyl acetate (10 mL) were added to the reaction mixture to extract, and the organic phase was concentrated under reduced pressure to give a crude product. The crude product is purified by column chromatography (eluent: ethyl acetate/petroleum ether: 0-50%). Compound 1-p is obtained.
MS m/z(ESI):589.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.71-7.61(m,2H),7.57(s,1H),6.37(s,1H),5.04-4.98(m,1H),4.48(dd,J=3.3,11.8Hz,1H),4.18-4.12(m,2H),2.99-2.92(m,1H),2.75(s,3H),2.60-2.20(m,6H),2.07(td,J=3.6,12.5Hz,2H),1.67-1.56(m,2H),1.27-1.24(m,3H).
Step 13: synthesis of Compound 1
Compound 1-p (50 mg, 84.88. Mu. Mol,1 eq) was dissolved in methanol (1 mL), and a solution of sodium hydroxide (10.18 mg, 254.64. Mu. Mol,3 eq) in water (1 mL) was added. The reaction was stirred at 25℃for 1 hour. The reaction solution was added with 2M diluted hydrochloric acid to adjust ph=1 to 3, extracted with ethyl acetate (20 mL), and the organic phase was concentrated under reduced pressure to obtain a crude product. The crude product is purified by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to obtain compound 1.
MS m/z(ESI):561.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.72-7.61(m,2H),7.59-7.56(m,1H),7.58(s,1H),6.38(s,1H),5.02(br dd,J=3.2,11.7Hz,1H),4.48(dd,J=3.1,11.8Hz,1H),2.97(br d,J=13.9Hz,1H),2.76(s,3H),2.64-2.55(m,1H),2.52-2.45(m,1H),2.43-2.29(m,3H),2.28-2.18(m,1H),2.14-2.06(m,2H),1.72-1.54(m,2H).
Step 14: synthesis of Compounds 2,3,4 and 5
Compound 1 (90 mg, 160.42. Mu. Mol,1 eq) was subjected to SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% NH) 3 H 2 O IPA]:50% -50%) to give a mixture of compounds 3 and 4, compound 2 and compound 5. The mixture of compounds 3 and 4 was subjected to a second SFC (column: DAICEL CHIRALPAK IG (250 mm. Times.30 mm,10 μm): mobile phase: [0.1% NH 3 H 2 O IPA]:60% -60%) to give compound 3 and compound 4.
Compound 2: MS m/z (ESI): 560.9[ M+1 ]] + SFC analysis method (column: chiralpak IG-3.50 x 4.6mmI.D.,3 μm; mobile phase: isopropanol (0.05% diethylamine; flow rate: 4 mL/min), SFC retention time: 0.513 min, ee% = 100%.
1 H NMR(400MHz,CD 3 OD)δ=7.59-7.50(m,2H),7.46(s,1H),6.26(s,1H),4.90(br dd,J=3.3,11.7Hz,1H),4.35(dd,J=3.3,11.8Hz,1H),2.85(br d,J=14.0Hz,1H),2.64(s,3H),2.46(br t,J=10.6Hz,1H),2.36(br d,J=13.4Hz,1H),2.31-2.17(m,3H),2.14-2.05(m,1H),2.01-1.93(m,2H),1.60-1.47(m,2H).
Compound 3: MS m/z (ESI): 560.9[ M+1 ]] + SFC retention time: 0.861 min, ee% = 100%.
1 H NMR(400MHz,CD 3 OD)δ=7.59-7.50(m,2H),7.46(s,1H),6.26(s,1H),4.90(dd,J=3.3,11.7Hz,1H),4.36(dd,J=3.3,11.8Hz,1H),2.85(br d,J=13.9Hz,1H),2.64(s,3H),2.50-2.42(m,1H),2.40-2.32(m,1H),2.29-2.16(m,3H),2.14-2.05(m,1H),2.02-1.94(m,2H),1.59-1.46(m,2H).
Compound 4: MS m/z (ESI): 561.0[ M+1 ]] + SFC retention time: 2.802 minutes, ee% = 100%.
1 H NMR(400MHz,CD 3 OD)δ=7.59-7.51(m,2H),7.46(s,1H),6.25(s,1H),4.90(dd,J=3.3,11.8Hz,1H),4.36(dd,J=3.1,11.7Hz,1H),2.86(br d,J=14.1Hz,1H),2.64(s,3H),2.47-2.41(m,1H),2.40-2.33(m,1H),2.29-2.17(m,3H),2.13-2.03(m,1H),2.00-1.93(m,2H),1.61-1.43(m,2H).
Compound 5: MS m/z (ESI): 560.9[ M+1 ]] + SFC retention time: 3.495 minutes, ee% = 100%.
1 H NMR(400MHz,CD 3 OD)δ=7.71-7.62(m,2H),7.58(s,1H),6.38(s,1H),5.02(dd,J=3.4,11.8Hz,1H),4.47(dd,J=3.2,11.7Hz,1H),2.97(br d,J=13.8Hz,1H),2.76(s,3H),2.62-2.54(m,1H),2.48(br d,J=13.5Hz,1H),2.43-2.29(m,3H),2.25-2.15(m,1H),2.13-2.05(m,2H),1.71-1.58(m,2H).
Example 2
The synthetic route is as follows:
step 1: synthesis of Compound 6-b
Compound 1-l (50 mg, 99.65. Mu. Mol,1 eq) was dissolved in N, N-dimethylformamide (1.4 mL) and triethylamine (1.4 mL) under nitrogen, and Compound 6-a (86.79 mg, 498.25. Mu. Mol,5 eq) and bis (triphenylphosphine) palladium dichloride (3.50 mg, 4.98. Mu. Mol,0.05 eq) and cuprous iodide (3.80 mg, 19.93. Mu. Mol,0.2 eq) were further added thereto, and the reaction mixture was heated to 80℃and stirred for 12 hours. To the reaction solution was added water (10 mL) and ethyl acetate (10 mL. Times.3) for extraction, and the organic phase was washed with 1M diluted hydrochloric acid (10 mL. Times.2) and saturated brine (10 mL. Times.2) in this order, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 6-b.
MS m/z(ESI):594.9[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.05(d,J=8.5Hz,2H),7.98(s,1H),7.70-7.62(m,4H),5.05(dd,J=3.1,11.9Hz,1H),4.48(dd,J=3.0,12.1Hz,1H),4.39(d,J=7.1Hz,2H),2.75(s,3H),2.45(td,J=3.2,14.3Hz,1H),2.36-2.27(m,1H),1.42-1.38(m,3H).
Step 2: synthesis of Compound 6
Compound 6-b (40 mg, 67.22. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (13.44 mg, 336.11. Mu. Mol,5 eq) was added thereto and the mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NXC18X30mmX3μm; mobile phase: water (0.225% formic acid) -acetonitrile;: acetonitrile: 45% -75%,7 min) to give compound 6.
MS m/z(ESI):567.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.04(d,J=8.5Hz,2H),7.97(s,1H),7.70-7.60(m,4H),5.04(dd,J=3.0,12.0Hz,1H),4.48(dd,J=3.0,12.0Hz,1H),2.74(s,3H),2.48-2.24(m,2H).
Example 3
The synthetic route is as follows:
step 1: synthesis of Compound 7-c:
compound 1-l (9.3 g,18.54mmol,1 eq) by SFC (column type: REGIS (s, s) WHELK-O1 (250 mm. Times.50 mm,10 μm); mobile phase: [0.1% NH 3 H 2 O,EtOH]Percent: 25% -25%) and collecting the second peak to give compound 7-c.
MS m/z(ESI):500.8[M+1] + SFC analysis method (column type (S, S) -Whelk-O1 100X 4.6mm I.D.,3 μm; mobile phase: ethanol (0.05% diethylamine; flow rate: 2.8 mL/min); retention time: 2.990 minutes; ee% = 100%.
Step 2: synthesis of Compound 7-a
To the reaction flask were added compound 1-m (2 g,11.75mmol,1.87mL,1 eq), tribromofluoromethane (3.82 g,14.10mmol,1.2 eq), triphenylphosphine (3.70 g,14.10mmol,1.2 eq) and tetrahydrofuran (60 mL). Diethyl zinc (1M, 14.10mL,1.2 eq) was added dropwise at 20deg.C. The reaction solution was stirred at 20℃for 1 hour. Methanol (50 mL) was added to the reaction mixture, and the mixture was stirred for 20 minutes and then concentrated under reduced pressure. The residue was separated by automatic column chromatography COMBI-FLASH (eluent: petroleum ether: ethyl acetate=100:0 to 10:1) to give compound 7-a.
1 H NMR(400MHz,CDCl 3 )δ=4.08-4.02(m,2H),2.72-2.68(m,1H),2.39-2.34(m,2H),1.95-1.81(m,4H),1.52-1.49(m,2H),1.21-1.18(m,3H).
Step 3: synthesis of Compound 7-b
Compound 7-a (2.3 g,8.68mmol,1 eq) was dissolved in 1, 4-dioxane (30 mL) and then pinacol borate (4.41 g,17.35mmol,2 eq), potassium acetate (2.55 g,26.03mmol,3 eq) and Pd (dppf) Cl were added 2 ·CH 2 Cl 2 (708.46 mg, 867.54. Mu. Mol,0.1 eq). After three nitrogen substitutions, the mixture was heated to 90℃and stirred for 16 hours. To the reaction solution was added water (50 mL), extracted with ethyl acetate (30 mL. Times.2), and the combined organic phases were washed with saturated aqueous sodium chloride solution (50 mL. Times.1), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was separated by automatic column chromatography COMBI-FLASH (eluent: petroleum ether: ethyl acetate=100:0 to 10:1) to give compound 7-b.
1 H NMR(400MHz,CDCl 3 )δ=4.08-4.02(m,2H),2.92-2.88(m,2H),2.43-2.37(m,1H),1.97-1.92(m,2H),1.97-1.92(m,2H),1.52-1.49(m,2H),1.21-1.18(m,15H).
Step 4: synthesis of Compound 7-d
In a thumb flask under nitrogen, compound 7-c (150.00 mg, 298.95. Mu. Mol,1 eq), 7-b (466.64 mg,1.49mmol,5 eq), sodium carbonate (158.43 mg,1.49mmol,5 eq), 1, 4-dioxane (8 mL) and water (1.5 mL) were added followed by Pd (PPh) 3 ) 4 (34.55 mg, 29.90. Mu. Mol,0.1 eq). The mixture was stirred at 100℃for 16 hours. After the reaction was cooled to about 25 ℃, water (10 mL) was added to the reaction mixture, the pH was adjusted to 1 to 2 with 1M diluted hydrochloric acid, and extraction was performed with ethyl acetate (15 ml×3). The combined organic phases were washed with saturated brine (30 mL) and concentrated under reduced pressure. The residue was isolated by automatic column chromatography COMBI-FLASH (gradient elution: petroleum ether: ethyl acetate=100:0 to 70:30) to give compound 7-d.
MS m/z(ESI):607.0[M+1] + .
1H NMR(400MHz,DMSO-d 6 )δ=7.97-7.90(m,2H),7.73(q,J=5.5Hz,2H),4.97(br t,J=9.8Hz,1H),4.43(dd,J=2.5,12.0Hz,1H),4.10-4.06(m,2H),2.78(br d,J=14.1Hz,1H),2.63(s,3H),2.57(td,J=3.8,10.4Hz,1H),2.42-2.35(m,1H),2.24-2.14(m,2H),2.13-2.05(m,1H),1.87-1.77(m,1H),1.76-1.66(m,1H),1.64-1.57(m,1H),1.56-1.43(m,3H),1.39-1.27(m,1H),1.21-1.18(m,3H).
Step 5: synthesis of Compound 7
Compounds 7-d (100 mg, 164.73. Mu. Mol,1 eq) and tetrahydrofuran (0.5 mL) were added to the thumb flask followed by aqueous sodium hydroxide (2M, 0.5mL,6.07 eq). The mixture was stirred at 25℃for 1 hour. Water (5 mL) was added to the reaction mixture, pH=1 to 2 was adjusted with 1M diluted hydrochloric acid, and extraction was performed with ethyl acetate (15 mL. Times.3). The combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was separated by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to give compound 7.
MS m/z(ESI):579.0[M+1] + .
1H NMR(400MHz,DMSO-d 6 )δ=7.99-7.88(m,2H),7.78-7.68(m,2H),4.98(br t,J=9.6Hz,1H),4.43(dd,J=2.5,12.0Hz,1H),2.77(br d,J=13.8Hz,1H),2.63(s,3H),2.55(br s,1H),2.49-2.45(m,1H),2.43-2.35(m,1H),2.24-2.11(m,2H),2.08-2.04(m,1H),1.98(br dd,J=4.1,8.1Hz,2H),1.49(q,J=11.6Hz,2H).
Example 4
The synthetic route is as follows:
step 1: synthesis of Compound 8-c
Compound 8-a (500 mg,2.97mmol,1 eq) was dissolved in tetrahydrofuran (15 mL), the reaction mixture was cooled to-78℃and then LiHMDS (1M, 2.97mL,1 eq) was slowly added dropwise thereto, the reaction was stirred at-78℃for 0.5 hours, then a solution of compound 8-b (1.06 g,2.97mmol,1 eq) in tetrahydrofuran (15 mL) was slowly added dropwise thereto, the reaction was stirred at-78℃for 0.5 hours and then the temperature was raised to 25℃for 1 hour. To the reaction mixture was slowly added water (20 mL), followed by extraction with ethyl acetate (20 mL. Times.3), and the combined organic phases were washed with saturated brine (10 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/1) to give compound 8-c.
1 H NMR(400MHz,CDCl 3 )δ=5.62-5.39(m,1H),3.71(d,J=3.5Hz,3H),3.16-3.02(m,1H),2.92(d,J=16.6Hz,2H),2.62-2.50(m,2H),2.48-2.38(m,2H).
Step 2: synthesis of Compound 8-d
Compound 8-c (400 mg,1.33mmol,1 eq), bis-pinacolato borate (405.96 mg,1.60mmol,1.2 eq), potassium acetate (392.24 mg,4.00mmol,3 eq) and 1, 1-bis (diphenylphosphine) ferrocene palladium chloride (97.48 mg, 133.22. Mu. Mol,0.1 eq) were dissolved in 1, 4-dioxane (7 mL) under nitrogen, and the reaction system was replaced three times with nitrogen and then warmed to 90℃and stirred for 3 hours. The reaction solution was concentrated directly under reduced pressure, and the crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/1) to give compound 8-d.
1 H NMR(400MHz,CDCl 3 )δ=7.12-6.70(m,1H),3.68(s,3H),3.10-2.99(m,1H),2.68-2.53(m,2H),2.53-2.32(m,4H),1.27(s,12H).
Step 3: synthesis of Compound 8-e
Under nitrogen atmosphere, compound 7-c (100 mg, 199.30. Mu. Mol,1 eq), compound 8-d (166.31 mg, 597.90. Mu. Mol,3 eq), potassium phosphate (126.91 mg, 597.90. Mu. Mol,3 eq) and 1, 1-bis (diphenylphosphine) ferrocene palladium chloride (14.58 mg, 19.93. Mu. Mol,0.1 eq) were dissolved in 1, 4-dioxane (1.4 mL) and water (0.4 mL), and the reaction system was replaced three times with nitrogen and then warmed to 80℃and stirred for 3 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 8-e.
MS m/z(ESI):573.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.70-7.60(m,5H),6.43-6.22(m,1H),5.01-4.97(m,1H),4.47-4.44(m,1H),3.68(s,3H),2.79(s,1H),2.73(s,3H),2.49-2.44(m,3H),2.42-2.36(m,2H),2.28(br d,J=14.3Hz,2H).
Step 4: synthesis of Compound 8-f
Compound 8-e (50.00 mg, 87.26. Mu. Mol,1 eq) was dissolved in methanol (3 mL), the reaction system was replaced with nitrogen three times, wet palladium on carbon (92.86 mg, 87.26. Mu. Mol,10% purity, 1 eq) was added, and after replacing with hydrogen three times, the reaction mixture was stirred at 25℃under a hydrogen atmosphere (15 Psi) for 3 hours. The reaction solution was filtered through celite, and concentrated under reduced pressure to give compound 8-f.
MS m/z(ESI):575.2[M+1] + .
Step 5: synthesis of Compound 8
Compound 8-f (15.00 mg, 26.08. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (5.22 mg, 130.42. Mu. Mol,5 eq) was added thereto and the mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is purified by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to obtain compound 8.
MS m/z(ESI):561.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.70-7.58(m,2H),7.44(s,1H),4.94(br d,J=3.3Hz,1H),4.51-4.39(m,1H),3.02(quin,J=8.3Hz,1H),2.73(s,3H),2.66-2.58(m,1H),2.52-2.09(m,10H).
Example 5
The synthetic route is as follows:
step 1: synthesis of Compound 9-b
2, 6-tetramethylpiperidine (2.38 g,16.88mmol,2.87mL,1.2 eq) was dissolved in tetrahydrofuran (24 mL), the system was cooled to-30deg.C and n-butyllithium (2.5M, 6.70mL,1.19 eq) was added. The reaction was stirred at-30℃for 0.5 hour, the reaction was cooled to-78℃and a solution of compound 1-n (3.77 g,14.07mmol,1 eq) in tetrahydrofuran (24 mL) was added, and stirring was carried out at-78℃for 30 minutes and compound 9-a (2 g,14.07mmol,1.87mL,1 eq) in tetrahydrofuran (24 mL) was added. The reaction was slowly warmed to 25℃and stirred at 25℃for 12 hours. The system was cooled to 0deg.C, saturated ammonium chloride solution (20 mL) was added dropwise, the reaction solution was stirred at 0deg.C for 1 hour, the filtrate was concentrated under reduced pressure after the reaction solution was filtered, water (20 mL), ethyl acetate (20 mL. Times.2) was added for extraction, and the organic phase was washed with saturated brine (20 mL) and concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=100:0-90:10) to give compound 9-b.
1 H NMR(400MHz,CDCl 3 )δ=5.25-5.15(m,1H),4.27-4.08(m,2H),3.23-3.08(m,4H),3.00-2.93(m,1H),1.30-1.28(m,3H),1.25(d,J=1.5Hz,12H).
Step 2: synthesis of Compound 9-c
Compound 7-c (100.00 mg, 199.30. Mu. Mol,1 eq) and compound 9-b (159.13 mg, 597.90. Mu. Mol,3 eq), sodium carbonate (63.37 mg, 597.90. Mu. Mol,3 eq), 1, 4-dioxane (2 mL) and water (0.3 mL) were added to a thumb flask under nitrogen, followed by tetrakis (triphenylphosphine) palladium (23.03 mg, 19.93. Mu. Mol,0.1 eq). The reaction was stirred at 90℃for 12 hours. The reaction solution was concentrated under reduced pressure to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=100:0-50:50) to give compound 9-c.
MS m/z(ESI):561.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.71-7.62(m,2H),7.55(s,1H),6.47(br s,1H),5.01(br d,J=9.3Hz,1H),4.47(dd,J=3.0,11.8Hz,1H),4.19(q,J=7.1Hz,2H),3.33(s,1H),3.22-3.10(m,4H),2.75(s,3H),2.43-2.30(m,2H),1.29(t,J=7.0Hz,3H).
Step 3: synthesis of Compound 9
Compound 9-c (25.00 mg, 44.56. Mu. Mol,1 eq) was dissolved in methanol (2 mL) and a solution of sodium hydroxide (5.35 mg, 133.68. Mu. Mol,3 eq) in water (2 mL) was added. The reaction was stirred at 25℃for 30 minutes. The reaction solution was added with 2M diluted hydrochloric acid to adjust ph=2 to 3, extracted with ethyl acetate (10 ml×3), and the organic phase was concentrated under reduced pressure to obtain a crude product. The crude product was purified by preparative high performance liquid chromatography (column: welch Xtimate C18X 40mm X3 μm; mobile phase [ water (0.075% trifluoroacetic acid) -acetonitrile ]; acetonitrile%: 45% -75%,8 min) to give compound 9.
MS m/z(ESI):533.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.72-7.62(m,2H),7.56(s,1H),6.49(t,J=2.3Hz,1H),5.01(br d,J=9.3Hz,1H),4.47(dd,J=3.4,11.9Hz,1H),3.30(s,1H),3.20(br s,2H),3.14(br d,J=8.5Hz,2H),2.75(s,3H),2.44-2.25(m,2H).
Example 6
The synthesis method comprises the following steps:
step 1: synthesis of Compound 10-b
2, 6-tetramethylpiperidine (9.20 g,65.14mmol,11.06mL,1.2 eq) was dissolved in tetrahydrofuran (120 mL) under nitrogen protection, after the reaction system was cooled to-30 ℃, n-butyllithium (2.5M, 26.05mL,1.2 eq) was slowly added dropwise, the reaction solution was stirred at-30℃for 0.5 hours and then cooled to-78℃and then a tetrahydrofuran (120 mL) solution of compound 1-n (14.54 g,54.28mmol,1 eq) was slowly added dropwise, the reaction solution was stirred at-78℃for 0.5 hours and then a tetrahydrofuran (120 mL) solution of compound 10-a (10 g,54.28mmol,1 eq) was added, and the reaction solution was heated to 25℃and stirred for 12 hours. After the completion of the reaction, the reaction mixture was cooled to 0℃and then a saturated ammonium chloride solution (100 mL) was slowly added dropwise thereto, and the reaction mixture was stirred at 0℃for 10 minutes. The reaction mixture was filtered and dried by spin-drying, water (60 mL) was added, the mixture was extracted with ethyl acetate (60 mL. Times.3), and the organic phase was washed with saturated brine (60 mL. Times.3), dried over anhydrous sodium sulfate, and filtered. Concentrating the filtrate under reduced pressure to obtain crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/1) to give compound 10-b.
1 H NMR(400MHz,CDCl 3 )δ=5.05(s,1H),4.18-4.08(m,2H),3.14(br dd,J=1.4,13.4Hz,1H),2.34-2.18(m,4H),2.05-1.95(m,2H),1.91-1.82(m,2H),1.28-1.24(m,15H),1.20-1.09(m,2H).
Step 2: synthesis of Compound 10-d and Compound 10-e
Compound 7-c (200 mg, 398.60. Mu. Mol,1 eq), compound 10-b (614.29 mg,1.99mmol,5 eq) and N, N-diisopropylethylamine (154.55 mg,1.20mmol, 208.29. Mu.L, 3 eq) were dissolved in 1, 4-dioxane (1.5 mL) and water (1.5 mL) under nitrogen, and bis (triphenylphosphine) palladium dichloride (27.98 mg, 39.86. Mu. Mol,0.1 eq) was added thereto, and the reaction mixture was heated to 100℃and stirred for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 10-c. Then SFC (column type: DAICEL CHIRALPAK IG (250 mm. Times.30 mm,10 μm), mobile phase: [0.1% NH ] 3 H 2 O,EtOH]Percent: 60% -60%) to give compound 10-d and compound 10-e.
Compound 10-c: MS m/z (ESI): 603.1[ M+1 ]] + .
1 H NMR(400MHz,CD 3 OD)δ=7.71-7.59(m,2H),7.54(s,1H),6.33(s,1H),4.99(dd,J=3.5,11.5Hz,1H),4.46(dd,J=3.4,11.7Hz,1H),4.14-4.11(m,2H),2.99(br d,J=13.8Hz,1H),2.74(s,3H),2.44-2.24(m,6H),2.15-2.02(m,2H),1.93(br d,J=12.8Hz,2H),1.25(d,J=1.0Hz,3H),1.17-1.08(m,2H).
SFC analysis method: column type: chiralpak IG-3.6mm I.D.,3 μm; mobile phase: ethanol (0.05% diethylamine); flow rate: 4mL/min.
Compound 10-d: MS m/z (ESI): 603.1[ M+1 ]] + SFC retention time: 1.982 minutes; ee% = 100%.
Compound 10-e: MS m/z (ESI): 603.1[ M+1 ]] + SFC retention time: 4.655 minutes; ee% = 99.5%.
Step 3: synthesis of Compound 10
Compound 10-c (70 mg, 116.07. Mu. Mol,1 eq), sodium hydroxide (2M, 290.17. Mu.L, 5 eq), tetrahydrofuran (3 mL) and water (1 mL) were added to a eggplant-shaped bottle, and the reaction solution was stirred at 20℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile;: 25% -95% acetonitrile, 7 min) to obtain compound 10.
MS m/z(ESI):575.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.78-7.48(m,3H),6.34(br s,1H),5.01(br d,J=9.5Hz,1H),4.47(br d, J=9.0Hz,1H),3.01(br d,J=14.1Hz,1H),2.75(s,3H),2.51-1.84(m,10H),1.42-1.05(m,2H).
Step 4: synthesis of Compound 11
Compound 10-d (260 mg, 431.11. Mu. Mol,1 eq) was dissolved in methanol (2 mL) and water (2 mL), and sodium hydroxide (86.21 mg,2.16mmol,5 eq) was added thereto and the mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile;: acetonitrile%: 40% -70%,7 min) to obtain compound 11.
MS m/z(ESI):575.1[M+1] + .
SFC analysis method (column: chiralPak AS-3X 4.6mm I.D.,3 μm; mobile phase: isopropanol (0.05% diethylamine; flow rate: 2.5 mL/min); SFC retention time: 5.559 minutes; ee% = 100%.
1 H NMR(400MHz,CD 3 OD)δ=7.70-7.60(m,2H),7.54(s,1H),6.33(s,1H),4.99(br dd,J=3.3,11.8Hz,1H),4.46(dd,J=3.3,11.8Hz,1H),3.00(br d,J=13.8Hz,1H),2.74(s,3H),2.46-2.28(m,4H),2.22(d,J=7.0Hz,2H),2.15-2.00(m,2H),1.96(br d,J=12.8Hz,2H),1.24-1.08(m,2H).
Step 5: synthesis of Compound 12
Compound 10-e (210.00 mg, 348.20. Mu. Mol,1 eq) was dissolved in methanol (2 mL) and water (2 mL), and sodium hydroxide (69.64 mg,1.74mmol,5 eq) was added thereto and the mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is purified by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to obtain compound 12.
MS m/z(ESI):575.1[M+1] + SFC retention time: 5.847 minutes; ee% = 98.5%.
1 H NMR(400MHz,CD 3 OD)δ=7.70-7.60(m,2H),7.54(s,1H),6.33(s,1H),4.99(br dd,J=3.3,11.8Hz,1H),4.46(dd,J=3.3,11.8Hz,1H),3.00(br d,J=13.8Hz,1H),2.74(s,3H),2.46-2.28(m,4H),2.22(d,J=7.0Hz,2H),2.15-2.00(m,2H),1.96(br d,J=12.8Hz,2H),1.24-1.08(m,2H).
Example 7
The synthetic route is as follows:
step 1: synthesis of Compound 13-a
2, 6-tetramethylpiperidine (2.90 g,20.53mmol,3.48mL,1.1 eq) was added to tetrahydrofuran (50 mL), cooled to-78 ℃, n-butyllithium (2.5M, 9.70mL,1.3 eq) was added dropwise, the reaction was stirred at-78℃for 30 minutes, the temperature was raised to 0 ℃, a solution of compound 1-n in tetrahydrofuran (15 mL) was added dropwise, the reaction was stirred at 0℃for 30 minutes, and a solution of methyl iodide (3.97 g,27.99mmol,1.74mL,1.5 eq) in tetrahydrofuran (15 mL) was added dropwise. The reaction was stirred at 0deg.C for 10 min, warmed to 25deg.C and stirred for 12 hr. The temperature of the reaction mixture was lowered to 0℃and a saturated ammonium chloride solution (30 mL) was added thereto, followed by stirring for 20 minutes and extraction with ethyl acetate (20 mL. Times.2). The combined organic phases were concentrated under reduced pressure to give the crude product. Purifying the crude product with column chromatography (eluent: petroleum ether/ethyl acetate, gradient: 0-10%) to obtain compound 13-a.
1 H NMR(400MHz,CDCl 3 )δ=1.23(s,24H),1.07(s,3H),0.90-0.84(m,1H).
Step 2: synthesis of Compound 13-b
2, 6-tetramethylpiperidine (2.49 g,17.63mmol,2.99mL,1.2 eq) was dissolved in tetrahydrofuran (30 mL), the system was cooled to-30deg.C and n-butyllithium (2.5M, 6.99mL,1.19 eq) was added dropwise. The reaction was stirred at-30℃for 0.5 hour, the reaction was cooled to-78℃and a solution of compound 13-a (4.14 g,14.69mmol,1 eq) in tetrahydrofuran (30 mL) was added dropwise, and stirring was carried out at-78℃for 30 minutes and compound 1-m (2.5 g,14.69mmol,2.34mL,1 eq) in tetrahydrofuran (30 mL) was added dropwise. The reaction was slowly warmed to 25℃and stirred at 25℃for 12 hours. The system was cooled to 0deg.C, saturated ammonium chloride solution (30 mL) was added dropwise, the reaction solution was stirred at 0deg.C for 20 minutes, and the filtrate was concentrated under reduced pressure after the reaction solution was filtered. Water (20 mL) and ethyl acetate (20 mL) were added for extraction. The combined organic phases were washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. Purifying the crude product with column chromatography (eluent: petroleum ether/ethyl acetate, gradient: 0-10%) to obtain compound 13-b.
MS m/z(ESI):309.0[M+1] + .
1 H NMR(400MHz,CDCl 3 )δ=4.13(q,J=7.1Hz,2H),3.04(br d,J=13.6Hz,1H),2.74(br d,J=13.8Hz,1H),2.56-2.45(m,1H),2.10-1.96(m,3H),1.90-1.81(m,1H),1.70(s,3H),1.62-1.55(m,2H),1.28(s,12H),1.26(br s,3H).
Step 3: synthesis of Compound 13-c
Compound 13-b (100 mg, 199.30. Mu. Mol,1 eq) and compound 7-c (307.14 mg, 996.51. Mu. Mol,5 eq) were dissolved in 1, 4-dioxane (3 mL), water (0.6 mL) and sodium carbonate (105.62 mg, 996.51. Mu. Mol,5 eq) were added, and 1, 1-bis (diphenylphosphine) ferrocene palladium chloride (29.17 mg, 39.86. Mu. Mol,0.2 eq) was added after nitrogen substitution. The reaction was stirred at 100℃for 12 hours. The reaction solution was concentrated under reduced pressure to obtain a crude product. Purifying the crude product with column chromatography (eluent: petroleum ether/ethyl acetate, gradient: 0-40%) to obtain compound 13-c.
MS m/z(ESI):603.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.73-7.62(m,2H),7.43(s,1H),5.02(dd,J=3.5,11.8Hz,1H),4.48(dd,J=3.3,11.8Hz,1H),4.18-4.11(m,2H),2.86-2.79(m,1H),2.76(s,3H),2.64-2.55(m,2H),2.44-2.28(m,2H),2.16-2.07(m,2H),2.04(s,3H),2.02-1.90(m,2H),1.69-1.60(m,1H),1.56-1.46(m,1H),1.28-1.25(m,3H).
Step 4: synthesis of Compound 13
Compound 13-c (30 mg, 49.74. Mu. Mol,1 eq) was dissolved in methanol (2 mL), and a solution of sodium hydroxide (1.99 mg, 49.74. Mu. Mol,1 eq) in water (2 mL) was added. The reaction was stirred at 25℃for 0.5 h. To the reaction mixture was added 2M diluted hydrochloric acid to adjust ph=2 to 3, and ethyl acetate (10 ml×3) was added for extraction. The combined organic phases were concentrated under reduced pressure. The crude product is purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile;: acetonitrile%: 45% -75%,7 min) to obtain compound 13.
MS m/z(ESI):575.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.73-7.60(m,2H),7.43(s,1H),5.02(dd,J=3.4,11.8Hz,1H),4.48(dd,J=3.2,11.8Hz,1H),2.83(br d,J=14.5Hz,1H),2.76(s,3H),2.67-2.51(m,2H),2.44-2.27(m,2H),2.16-2.07(m,2H),2.05(s,3H),1.98(br t,J=11.4Hz,2H),1.68-1.46(m,2H).
Example 8
The synthetic route is as follows:
step 1: synthesis of Compound 14-a
Methyl triphenylphosphine bromide (23.09 g,64.63mmol,1.1 eq) was dissolved in tetrahydrofuran (160 mL), after cooling the reaction solution to-5 ℃, butyllithium (2.5M, 25.85mL,1.1 eq) was slowly added dropwise under nitrogen atmosphere, then the reaction solution was stirred at 0℃for 1 hour, then compound 1-m (10 g,58.75mmol,9.35mL,1 eq) was slowly added dropwise, and the reaction solution was stirred at 25℃for 12 hours. After the reaction, the reaction mixture was cooled to 0℃and then a saturated ammonium chloride solution (100 mL) was slowly added to the reaction mixture, followed by stirring for 10 minutes, extraction with ethyl acetate (200 mL. Times.3) was further carried out, and the organic phase was washed with saturated brine (100 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 20/1) to give compound 14-a.
1 H NMR(400MHz,CDCl 3 )δ=4.65(s,2H),4.13(q,J=7.1Hz,2H),2.51-2.25(m,3H),2.14-1.94(m,4H),1.65-1.54(m,2H),1.32-1.19(m,3H).
Step 2: synthesis of Compound 14-b
Compound 14-a (3 g,17.83mmol,1 eq) was dissolved in tetrahydrofuran (150 mL), zinc-copper (20.69 g,160.49mmol,9 eq) was added, trichloroacetyl chloride (16.21 g,89.16mmol,9.95mL,5 eq) was slowly added dropwise, and the reaction was stirred at 25℃for 12 hours. The reaction solution was slowly poured into ice water, ethyl acetate (200 mL. Times.3) was added thereto to extract, and the combined organic phases were washed with saturated brine (100 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/1) to give compound 14-b.
1 H NMR(400MHz,CDCl 3 ) δ=4.16 (dd, j=7.2, 13.4hz, 2H), 3.04 (d, j=14.3 hz, 2H), 2.57-2.46 (m, 1H), 2.39-2.26 (m, 1H), 2.19-2.07 (m, 3H), 1.92 (br d, j=13.3 hz, 1H), 1.81-1.61 (m, 3H), 1.28 (d, j=7.0 hz, 3H),. Step 3: synthesis of Compound 14-c
Compound 14-b (2.3 g,8.24mmol,1 eq) was dissolved in glacial acetic acid (20 mL) and zinc powder (2.16 g,32.96mmol,4 eq) was added. The reaction was stirred at 50℃for 2 hours. The reaction solution was filtered, and the filter cake was quenched with 4M diluted hydrochloric acid and pH was adjusted to neutral with saturated aqueous sodium hydroxide solution. The filtrate was poured into water (50 mL), extracted with ethyl acetate (30 mL. Times.2), washed with a saturated aqueous sodium chloride solution (50 mL. Times.1) and the combined organic phases dried over anhydrous sodium sulfate, filtered and evaporated. The residue was purified by automatic column chromatography COMBI-FLASH separation (eluent: petroleum ether: ethyl acetate=100:0 to 10:1) to give compound 14-c.
1 H NMR(400MHz,CDCl 3 )δ=4.16-1.33(m,2H),2.77-2.76(d,J=8.0Hz,,4H),2.37-2.31(m,1H),1.97-1.93(m,2H),1.81-1.77(m,2H),1.65-1.55(m,4H),1.26(t,J=7.2Hz,3H).
Step 4: synthesis of Compound 14-d
Compound 14-c (300 mg,1.43mmol,1 eq) was dissolved in tetrahydrofuran (10 mL) and lithium bis (trimethylsilyl) amide (1.0M, 1.57mL,1.1 eq) was added dropwise at-78 ℃. After completion of the dropwise addition, the mixture was stirred at-78℃for 0.5 hour, and then a mixture of N-phenylbis (trifluoromethanesulfonyl) imide (560.67 mg,1.57mmol,1.1 eq) and tetrahydrofuran (10 mL) was added dropwise. The reaction was stirred at-78℃for 0.5 hours, then warmed to-78℃and stirred for 1 hour. Water (10 mL) was added to the reaction solution, the mixture was extracted with ethyl acetate (10 mL. Times.2), and the organic phases were combined, washed with saturated aqueous sodium chloride (20 mL. Times.1), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by automatic column chromatography COMBI-FLASH separation (eluent: petroleum ether: ethyl acetate=100:0 to 10:1) to give compound 14-d.
1 H NMR(400MHz,CDCl 3 )δ=5.69(s,0.5H),5.42(s,0.5H),4.09-4.03(m,2H),2.51-2.50(m,,2H),2.26-2.19(m,1H),1.89-1.86(m,2H),1.59-1.51(m,6H),1.18(t,J=6.0Hz,,3H),0.81-0.79((m,1H).
Step 5: synthesis of Compound 14-e
Compound 14-d (400 mg,1.17mmol,1 eq), pinacol borate (356.06 mg,1.40mmol,1.2 eq), potassium acetate (344.03 mg,3.51mmol,3 eq) and [1, 1-bis (diphenylphosphine) ferrocene ] palladium dichloride dichloromethane (95.42 mg, 116.85. Mu. Mol,0.1 eq) were added to 1, 4-dioxane (7 mL). The mixture was purged three times with nitrogen and then warmed to-78 ℃ and stirred for 3 hours. Water (10 mL) was added to the reaction solution, the mixture was extracted with ethyl acetate (10 mL), and the organic phases were combined, washed with saturated aqueous sodium chloride (10 mL. Times.1), dried over anhydrous sodium sulfate, and filtered and distilled off. The residue was purified by automatic column chromatography COMBI-FLASH separation (eluent: petroleum ether: ethyl acetate=100:0 to 10:1) to give compound 14-e.
1 H NMR(400MHz,CDCl 3 )δ=7.15(s,0.5H),6.86(s,0.5H),4.08-4.02(m,2H),2.21(d,J=6.0Hz,,2H),1.82-1.79(m,3H),1.69-1.56(m,2H),1.49-1.39(m,4H),1.23-1.18(m,15H).
Step 6: synthesis of Compound 14-f
Compound 1-l (100 mg, 199.30. Mu. Mol,1 eq), compound 14-e (191.47 mg, 597.90. Mu. Mol,3 eq), potassium phosphate (126.91 mg, 597.90. Mu. Mol,3 eq) and [1, 1-bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane (16.28 mg, 19.93. Mu. Mol,0.1 eq) were added to a mixture of 1, 4-dioxane (1.4 mL) and water (0.4 mL). The reaction was warmed to 80 ℃ and stirred for 3 hours. The reaction solution was poured into water (10 mL), extracted with ethyl acetate (10 ml×3), and the combined organic phases were washed with saturated aqueous sodium chloride (10 mL), dried over anhydrous sodium sulfate and filtered and distilled. The residue was purified by automatic column chromatography COMBI-FLASH separation (eluent: petroleum ether: ethyl acetate=100:0 to 2:1) to give compound 14-f.
MS m/z(ESI):615.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.64-7.61(m,3H),6.63(s,0.5H),6.33(s,0.5H),5.03(dd,J=3.2Hz,1H),4.49 (dd,J=3.2Hz,1H),4.16-4.11(m,2H),2.75(s,3H),2.53(d,J=3.2Hz,1H),2.39-2.32(m,3H),1.70-1.69(m,2H),1.69-1.49(m,7H),1.29-1.25(m,3H).
Step 7: synthesis of Compound 14-g
Compound 14-f (90 mg, 146.32. Mu. Mol,1 eq) was dissolved in methanol (3 mL) and three nitrogen substitutions were made to add wet palladium on carbon (90 mg, 146.32. Mu. Mol,10% purity, 1.00 eq). The reaction was then replaced three times with hydrogen and stirred at 20℃under hydrogen (15 Psi) for 12 hours. The reaction solution was directly filtered, and the filtrate was concentrated under reduced pressure and dried to obtain 14-g of the compound.
MS m/z(ESI):617.2[M+1] + .
Step 8: synthesis of Compound 14
Compound 14-g (30 mg, 48.61. Mu. Mol,1 eq) was dissolved in tetrahydrofuran (2 mL), and a mixture of sodium hydroxide (9.72 mg, 243.06. Mu. Mol,5 eq) and water (1 mL) was added. The reaction was stirred at 20℃for 1 hour. The organic solvent was directly distilled off, the pH was adjusted to 3-4 with 4N aqueous hydrochloric acid, and dichloromethane (10 ml×2) was used for extraction, and the combined organic phases were concentrated under reduced pressure and dried. The crude product is separated by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile;: acetonitrile%: 45% -75%,7 min) to obtain compound 14.
MS m/z(ESI):589.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.68-7.63(m,3H),7.62-7.47(m,1H),4.97-4.96(m,1H),4.47(dd,J=3.2Hz,1H),3.79-3.70(m,1H),2.75(s,3H),2.47-2.37(m,1H),2.32-2.29(m,1H),2.25-2.06(m,3H),1.93-1.89(m,4H),1.69-1.51(m,2H),1.49-1.44(m,4H).
Example 9
The synthetic route is as follows:
step 1: synthesis of Compound 15-a
Compound 1-h (152 g,431.59mmol,1 eq) was added to ethanol (1520 mL), the reaction cooled to 0deg.C, sodium borohydride (24.49 g,647.39mmol,1.5 eq) was slowly added, and the reaction stirred at 25deg.C for 0.5 hours. The reaction mixture was quenched slowly with saturated aqueous ammonium chloride (500 mL) and the pH was adjusted to neutral with 0.5M dilute hydrochloric acid. Most of the ethanol was removed by concentration under reduced pressure, 500mL of water was added thereto, ph=4 to 6 was adjusted with 0.5M diluted hydrochloric acid, and extraction was performed with ethyl acetate (1000 ml×2). The combined organic phases were washed successively with water (1000 mL) and saturated brine (1000 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product is pulped for 1 hour at room temperature by 500mL methyl tertiary butyl ether, filtered, and the filter cake is collected and dried under reduced pressure to obtain the compound 15-a.
MS m/z(ESI):355.8[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.98-7.54(m,1H),4.99(dd,J=3.0,12.0Hz,1H),4.41(dd,J=2.9,12.4Hz,1H),2.81-2.71(m,3H),2.57-2.44(m,1H),2.27-2.12(m,1H),2.27-2.12(m,1H)。
Step 2: synthesis of Compound 15-b
Compound 15-a (2 g,5.61mmol,1 eq), compound 1-o (8.26 g,28.07mmol,5 eq), N, N-diisopropylethylamine (3.63 g,28.07mmol,4.89mL,5 eq) and bis (triphenylphosphine) palladium dichloride (394.08 mg, 561.46. Mu. Mol,0.1 eq) were dissolved in 1, 4-dioxane (20 mL) and water (20 mL) under nitrogen and stirred for 12 hours at 80 ℃. To the reaction solution, 1M diluted hydrochloric acid was added to adjust ph=1 to 2, water (50 mL) was further added, extraction was performed with ethyl acetate (50 ml×3), and the combined organic phases were washed with saturated brine (50 ml×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (dichloromethane/methanol=1/0 to 5/1) to give compound 15-b.
MS m/z(ESI):444.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.57(dd,J=3.3,7.5Hz,1H),6.36(s,1H),4.98-4.93(m,1H),4.27(dd,J=2.9,12.4Hz,1H),4.14-4.07(m,1H),2.94(br d,J=14.3Hz,1H),2.75(s,3H),2.60(tt,J=4.0,10.7Hz,1H),2.50-2.41(m,2H),2.31(dt,J=3.9,12.7Hz,2H),2.23-2.14(m,2H),2.10-2.03(m,2H),1.68-1.54(m,2H),1.27-1.23(m,3H).
Step 3: synthesis of Compound 15-d
Compound 15-b (0.1 g, 225.46. Mu. Mol,1 eq) and compound 15-c (70.34 mg, 338.19. Mu. Mol,1.5 eq) were dissolved in dichloromethane (5 mL), the system was cooled to 0deg.C, and tri-N-propylcyclic phosphoric anhydride (215.21 mg, 338.19. Mu. Mol, 201.13. Mu.L, concentration: 50%,1.5 eq) and N, N-diisopropylethylamine (87.42 mg, 676.38. Mu. Mol, 117.81. Mu.L, 3 eq) were added. The reaction was stirred at 25℃for 16 hours. The reaction solution was concentrated under reduced pressure, and the crude product was purified by column chromatography (eluent: petroleum ether/ethyl acetate, gradient: 0-50%). Compound 15-d is obtained.
MS m/z(ESI):632.7[M+1] + .
Step 4: synthesis of Compound 15
Compound 15-d (50 mg, 78.92. Mu. Mol,1 eq) was dissolved in methanol (2 mL), a solution of sodium hydroxide (9.47 mg, 236.77. Mu. Mol,3 eq) in water (2 mL) was added, and the reaction was stirred at 25℃for 30 minutes. To the reaction mixture was added 2M diluted hydrochloric acid to adjust ph=3 to 4, and ethyl acetate (10 ml×3) was added for extraction. The combined organic phases were concentrated under reduced pressure. The crude product is purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile;: acetonitrile%: 40% -70%,7 min) to obtain compound 15.
MS m/z(ESI):605.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.78-7.68(m,2H),7.58(s,1H),6.38(s,1H),5.04-4.98(m,1H),4.47(dd,J=3.0,11.8Hz,1H),2.97(br d,J=14.1Hz,1H),2.76(s,3H),2.59(br s,1H),2.51-2.29(m,4H),2.28-2.19(m,1H),2.09(br d,J=12.0Hz,2H),1.71-1.55(m,2H).
Example 10
The synthetic route is as follows:
Step 1: synthesis of Compound 16-b
Compound 15-b (100 mg, 225.46. Mu. Mol,1 eq) and compound 16-a (49.75 mg, 338.19. Mu. Mol,1.5 eq) were dissolved in methylene chloride (5 mL), the reaction mixture was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (215.21 mg, 338.19. Mu. Mol, 201.13. Mu.L, concentration: 50%,1.5 eq) and N, N-diisopropylethylamine (87.42 mg, 676.38. Mu. Mol, 117.81. Mu.L, 3 eq) were added thereto, and the reaction mixture was heated to 25℃and stirred for reaction for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 16-b.
MS m/z(ESI):573.2[M+1] + .
Step 2: synthesis of Compound 16
Compound 16-b (50 mg, 87.32. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (17.46 mg, 436.59. Mu. Mol,5 eq) was added thereto, and the reaction was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18X 30mm X3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 35% -65%,7 min) to give compound 16.
MS m/z(ESI):544.5[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.60-7.42(m,3H),6.37(s,1H),5.00(br dd,J=3.3,11.8Hz,1H),4.45(br dd,J=3.3,11.8Hz,1H),2.95(br d,J=13.1Hz,1H),2.74(s,3H),2.57(brt,J=10.8Hz,1H),2.46(br d,J=13.3Hz,1H),2.41-2.26(m,3H),2.21(br t,J=11.4Hz,1H),2.07(br d,J=12.3Hz,2H),1.69-1.55(m,2H).
Example 11
The synthetic route is as follows:
step 1: synthesis of Compound 17-b
In a round bottom flask, compound 17-a (44.25 mg, 270.55. Mu. Mol,1.2 eq), 15-b (100 mg, 225.46. Mu. Mol,1 eq), tri-n-propyl cyclic phosphoric anhydride (215.21 mg, 338.19. Mu. Mol, 201.13. Mu.L, concentration: 50%,1.5 eq) and diisopropylethylamine (87.42 mg, 676.38. Mu. Mol, 117.81. Mu.L, 3 eq) were added sequentially to dichloromethane (5 mL) and the reaction stirred under nitrogen at 25℃for 2 hours. To the reaction solution was added water (50 mL), extracted with methylene chloride (30 mL. Times.2), and the combined organic phases were washed successively with water (30 mL) and saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/3) to give compound 17-b.
MS m/z(ESI):589.1[M+1] + .
Step 2: synthesis of Compound 17
Compound 17-b (60 mg, 101.85. Mu. Mol,1 eq) and sodium hydroxide (20.37 mg, 509.27. Mu. Mol,5 eq) were added to methanol (0.5 mL) and water (0.5 mL), and the reaction was stirred at 25℃for 30 minutes. The reaction solution was adjusted to ph=4 to 5 with 4M diluted hydrochloric acid, water (30 mL) was added, ethyl acetate (30 ml×2) was extracted, and the combined organic phases were washed successively with water (20 mL) and saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product is purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile;: 35% -65% acetonitrile, 7 min) to obtain compound 17.
MS m/z(ESI):561.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ=12.16(br s,1H),10.38(s,1H),7.91(br d,J=10.0Hz,1H),7.68-7.59(m,1H),7.68-7.59(m,1H),7.41-7.27(m,1H),6.41(s,1H),4.92(br t,J=10.0Hz,1H),4.46(br d,J=10.3Hz,1H),2.83(br d,J=14.1Hz,1H),2.67(s,3H),2.43-2.33(m,3H),2.31-2.21(m,1H),2.17(br d,J=12.8Hz,2H),1.98(br d,J=9.0Hz,2H),1.58-1.36(m,2H).
Example 12
The synthetic route is as follows:
step 1: synthesis of Compound 18-b
To a thumb flask was added compound 15-b (200 mg, 450.92. Mu. Mol,1 eq), compound 18-a (140.69 mg, 676.38. Mu. Mol,1.5 eq), diisopropylethylamine (174.83 mg,1.35mmol, 235.62. Mu.L, 3 eq) and tetrahydrofuran (5 mL) under nitrogen, followed by tri-n-propylcyclic phosphoric anhydride (573.90 mg, 901.84. Mu. Mol, 536.35. Mu.L, 50% purity, 2 eq). The mixture was stirred at 50℃for 12 hours. The reaction mixture was cooled to room temperature of 25℃and water (15 mL) was added thereto, pH=1 to 2 was adjusted with dilute hydrochloric acid (2M), and extraction was performed with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was isolated by automatic column chromatography COMBI-FLASH (gradient elution: petroleum ether: ethyl acetate=100:0 to 80:20) to give compound 18-b.
MS m/z(ESI):632.8[M+1] + .
Step 2: synthesis of Compound 18
A thumb flask was charged with compound 18-b (250 mg, 394.62. Mu. Mol,1 eq), methanol (5 mL), and then a solution of sodium hydroxide (78.92 mg,1.97mmol,5 eq) in water (1 mL). The mixture was stirred at 25℃for 1 hour. Water (5 mL) was added to the reaction mixture, pH=1 to 2 was adjusted with 1M diluted hydrochloric acid, and extraction was performed with ethyl acetate (15 mL. Times.3). The combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was separated by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 35% -65%,7 min) to give compound 18.
MS m/z(ESI):605.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ=12.16(br s,1H),10.35(s,1H),7.91(br d,J=9.8Hz,1H),7.72-7.61(m,2H),7.34-7.26(m,1H),6.41(s,1H),4.92(brt,J=9.9Hz,1H),4.45(br d,J=10.5Hz,1H),2.83(br d,J=14.1Hz,1H),2.67(s,3H),2.44-2.33(m,3H),2.27(br d,J=11.8Hz,1H),2.22-2.12(m,2H),2.04-1.92(m,2H),1.57-1.37(m,2H).
Example 13
The synthetic route is as follows:
step 1: synthesis of Compound 19-b
Compound 15-b (100 mg, 225.46. Mu. Mol,1 eq) and compound 19-a (60.88 mg, 338.19. Mu. Mol,1.5 eq) were dissolved in methylene chloride (5 mL), the reaction mixture was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (215.21 mg, 338.19. Mu. Mol, 201.13. Mu.L, concentration: 50%,1.5 eq) and N, N-diisopropylethylamine (87.42 mg, 676.38. Mu. Mol, 117.81. Mu.L, 3 eq) were added thereto, and the reaction mixture was heated to 25℃and stirred for reaction for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 19-b.
MS m/z(ESI):605.1[M+1] + .
Step 2: synthesis of Compound 19
Compound 19-b (50 mg, 82.57. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (16.51 mg, 412.86. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column: phenomenexGemini-NX C18 75 x 30mm x 3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile:% 40% -70% acetonitrile, 7 min) to give compound 19.
MS m/z(ESI):577.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.79(d,J=6.0Hz,2H),7.56(s,1H),6.36(s,1H),5.00(br dd,J=3.4,11.7Hz,1H),4.46(br dd,J=3.1,11.9Hz,1H),2.95(br d,J=14.1Hz,1H),2.74(s,3H),2.57(br t,J=10.8Hz,1H),2.51-2.43(m,1H),2.41-2.27(m,3H),2.20(br t,J=11.4Hz,1H),2.12-2.04(m,2H),1.69-1.55(m,2H).
Example 14
The synthetic route is as follows:
step 1: synthesis of Compound 20-a
Compound 15-a (10 g,28.07mmol,1 eq) was purified by SFC (column: DAICEL CHIRALPAKAD (250 mm. Times.50 mm,10 μm); mobile: [0.1% NH) 3 H 2 O,MeOH]Percent: 45% -45%) to give compound 20-a.
MS m/z(ESI):357.7[M+1] + SFC analysis method (column: chiralpak AD-3.150 x 4.6mm I.D.,3 μm; mobile phase: carbon dioxide 40% methanol (0.05% diethylamine; flow rate: 2.5 mL/min); SFC retention time: 3.030 minutes; ee% = 99.96%.
1 H NMR(400MHz,DMSO-d 6 )δ=7.84(s,1H),4.76(dd,J=2.6,11.9Hz,1H),3.73(br dd,J=2.3,12.3Hz,1H),3.17(s,1H),2.61(s,3H),2.30(br d,J=14.1Hz,1H),1.95-1.86(m,1H).
Step 2: synthesis of Compound 20-b
Compound 20-a (800.00 mg,2.25mmol,1 eq), compound 10-b (2.08 g,6.74mmol,3 eq), N, N-diisopropylethylamine (1.45 g,11.23mmol,1.96mL,5 eq) and bis (triphenylphosphine) palladium dichloride (157.63 mg, 224.58. Mu. Mol,0.1 eq) were dissolved in 1, 4-dioxane (10 mL) and water (10 mL) under nitrogen, and the reaction mixture was warmed to 80℃and stirred for 4 hours. To the reaction mixture was added 1M diluted hydrochloric acid to adjust the ph=1 to 2, followed by addition of water (50 mL) and extraction with ethyl acetate (50 ml×3). The combined organic phases were washed with saturated brine (50 mL ×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by C18 reverse phase column (water/acetonitrile=1/0 to 2/1) to give compound 20-b.
MS m/z(ESI):458.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.54(s,1H),6.33(s,1H),4.97(br dd,J=2.9,11.9Hz,1H),4.39(dd,J=2.8,12.4Hz,1H),4.16-4.10(m,2H),2.99(br d,J=12.0Hz,1H),2.76(s,3H),2.48-2.39(m,2H),2.35-2.20(m,4H),2.15-2.03(m,2H),1.94(br d,J=12.5Hz,2H),1.28-1.24(m,3H),1.21-1.09(m,2H).
Step 3: synthesis of Compound 20-d
Into a thumb flask, under nitrogen, 20-b (75 mg, 163.91. Mu. Mol,1 eq), 20-c (40.21 mg, 245.87. Mu. Mol,1.5 eq), diisopropylethylamine (63.55 mg, 491.74. Mu. Mol, 85.65. Mu. L,3 eq) and tetrahydrofuran (3 mL) were added, followed by tri-n-propylcyclic phosphoric anhydride (208.61 mg, 327.82. Mu. Mol, concentration: 50%,2 eq). The mixture was stirred at 50℃for 12 hours. The reaction solution was concentrated under reduced pressure. The residue was isolated by automatic column chromatography COMBI-FLASH (gradient elution: petroleum ether: ethyl acetate=100:0 to 70:30) to give compound 20-d.
MS m/z(ESI):603.1[M+1] + .
Step 4: synthesis of Compound 20
Into a thumb flask were added compound 20-d (80 mg, 132.65. Mu. Mol,1 eq) and methanol (1 mL), followed by aqueous sodium hydroxide (2M, 331.62. Mu.L, 5 eq). The mixture was stirred at 25℃for 1 hour. Water (5 mL) was added to the reaction mixture, pH=1 to 2 was adjusted with 1M diluted hydrochloric acid, and extraction was performed with ethyl acetate (15 mL. Times.2). The combined organic phases were washed with saturated brine (30 mL) and concentrated under reduced pressure. The residue was purified by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to give compound 20.
MS m/z(ESI):575.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.00(t,J=7.8Hz,1H),7.56(s,1H),7.37-7.24(m,1H),6.35(s,1H),5.03(dd,J=3.1,11.9Hz,1H),4.52(dd,J=3.0,12.0Hz,1H),3.08-2.94(m,1H),2.80(s,3H),2.52-2.40(m,2H),2.38-2.28(m,2H),2.24(d,J=7.0Hz,2H),2.18-2.02(m,2H),1.98(br d,J=12.3Hz,2H),1.27-1.08(m,2H).
Example 15
The synthetic route is as follows:
step 1: synthesis of Compound 21-b
Compound 20-b (75.00 mg, 163.91. Mu. Mol,1 eq) and compound 21-a (40.21 mg, 245.87. Mu. Mol,1.5 eq) were dissolved in tetrahydrofuran (3 mL), the reaction solution was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (208.61 mg, 327.82. Mu. Mol, concentration: 50%,2 eq) and N, N-diisopropylethylamine (63.55 mg, 491.74. Mu. Mol, 85.65. Mu.L, 3 eq) were added thereto, and the reaction solution was heated to 50℃and stirred for reaction for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 21-b.
MS m/z(ESI):603.1[M+1] + .
Step 2: synthesis of Compound 21
Compound 21-b (50 mg, 82.90. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (16.58 mg, 414.52. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column: phenomenexGemini-NX C18 75 x 30mm x 3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile:% 40% -70% acetonitrile, 7 min) to give compound 21.
MS m/z(ESI):575.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.68(dt,J=5.6,8.7Hz,1H),7.55(s,1H),7.15(dt,J=2.0,8.9Hz,1H),6.33(s,1H),5.02(dd,J=3.1,12.2Hz,1H),4.51(dd,J=3.1,11.9Hz,1H),3.00(br d,J=13.8Hz,1H),2.80(s,3H),2.49-2.39(m,2H),2.37-2.26(m,2H),2.22(d,J=7.0Hz,2H),2.16-2.00(m,2H),1.97(br d,J=11.5Hz,2H),1.25-1.09(m,2H).
Example 16
The synthetic route is as follows:
step 1: synthesis of Compound 22-b
Compound 20-b (75 mg, 163.91. Mu. Mol,1 eq) and compound 22-a (40.21 mg, 245.87. Mu. Mol,1.5 eq) were dissolved in tetrahydrofuran (3 mL) under nitrogen, the reaction mixture was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (208.61 mg, 327.82. Mu. Mol, 194.97. Mu.L, concentration: 50%,2 eq) and N, N-diisopropylethylamine (63.55 mg, 491.74. Mu. Mol, 85.65. Mu.L, 3 eq) were added thereto, and the reaction mixture was heated to 50℃and stirred for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 22-b.
MS m/z(ESI):603.1[M+1] + .
Step 2: synthesis of Compound 22
Compound 22-b (10 mg, 16.58. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (3.32 mg, 82.90. Mu. Mol,5 eq) was added thereto and the mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is purified by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile: acetonitrile%: 40% -70%,7 min) to obtain compound 22.
MS m/z(ESI):575.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.84(dd,J=8.0,11.8Hz,1H),7.58-7.50(m,2H),6.33(s,1H),5.02(dd,J=3.0,12.0Hz,1H),4.52(dd,J=3.3,12.0Hz,1H),3.00(br d,J=14.1Hz,1H),2.82(s,3H),2.54-2.40(m,2H),2.37-2.27(m,2H),2.23(d,J=7.0Hz,2H),2.16-2.00(m,2H),1.97(br d,J=11.8Hz,2H),1.23-1.09(m,2H).
Example 17
The synthetic route is as follows:
step 1: synthesis of Compound 23-b
23-a (500 mg,3.12mmol,1 eq) and wet palladium on carbon (67.47 mg, 31.23. Mu. Mol,5% purity, 0.01 eq) were added to methanol (50 mL) in a round bottom flask under nitrogen, then replaced three times with hydrogen and the reaction stirred at 40℃for 1 hour under H2 (25 psi) atmosphere. The reaction solution was filtered and concentrated under reduced pressure to give compound 23-b.
MS m/z(ESI):130.8[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ) δ=7.28 (t, j=2.6 hz, 1H), 7.06 (ddd, j= 2.4,8.6, 11.9hz, 1H), 5.54 (br s, 2H) ·step 2: synthesis of Compound 23-c
20-b (50 mg, 109.27. Mu. Mol,1 eq), 23-b (17.06 mg, 131.13. Mu. Mol,1.2 eq), tri-n-propyl cyclic phosphoric anhydride (139.08 mg, 218.55. Mu. Mol, concentration: 50%,2 eq) and diisopropylethylamine (42.37 mg, 327.82. Mu. Mol, 57.10. Mu.L, 3 eq) were added to tetrahydrofuran (5 mL), and the reaction solution was stirred at 50℃for 0.5 hours under nitrogen. Water (50 mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (30 mL. Times.2). The combined organic phases were washed successively with water (30 mL) and saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/3) to give compound 23-c.
MS m/z(ESI):570.0[M+1] + .
Step 3: synthesis of Compound 23
Compound 23-c (40 mg, 70.22. Mu. Mol,1 eq) and sodium hydroxide (28.09 mg, 702.19. Mu. Mol,10 eq) were added to methanol (1 mL) and water (1 mL), and the reaction solution was stirred at 25℃for 1 hour. The reaction mixture was adjusted to ph=4 to 5 with 4M diluted hydrochloric acid, water (30 mL) was added thereto, and extraction was performed with ethyl acetate (30 ml×2). The combined organic phases were washed successively with water (20 mL) and saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product is purified by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile: acetonitrile%: 35% -55%,7 min) to obtain compound 23.
MS m/z(ESI):541.9[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.29(ddd,J=2.3,8.7,11.2Hz,1H),8.21(t,J=2.0Hz,1H),7.57(s,1H),6.35(s,1H),5.02(br dd,J=3.0,11.8Hz,1H),4.54(dd,J=3.1,11.7Hz,1H),3.09-2.94(m,1H),2.77(s,3H),2.49-2.20(m,6H),2.19-1.92(m,4H),1.29-1.04(m,2H).
Example 18
The synthetic route is as follows:
step 1: synthesis of Compound 24-b
To a thumb flask, compound 20-b (75 mg, 163.91. Mu. Mol,1 eq), compound 24-a (31.61 mg, 245.87. Mu. Mol,1.5 eq), diisopropylethylamine (63.55 mg, 491.73. Mu. Mol, 85.65. Mu. L,3 eq) and tetrahydrofuran (3 mL) were added under nitrogen, followed by tri-n-propylcyclic phosphoric anhydride (208.61 mg, 327.82. Mu. Mol, 194.97. Mu.L, concentration: 50%,2 eq). The mixture was stirred at 50℃for 12 hours. The reaction solution was concentrated under reduced pressure. The residue was isolated by automatic column chromatography COMBI-FLASH (gradient elution: petroleum ether: ethyl acetate=100:0 to 50:50) to give compound 24-b.
MS m/z(ESI):568.0[M+1] + .
Step 2: synthesis of Compound 24
In a thumb flask was added compound 24-b (40 mg, 70.41. Mu. Mol,1 eq) and methanol (1 mL), followed by sodium hydroxide (2M, 0.5mL,14.20 eq). The mixture was stirred at 25℃for 1 hour. Water (5 mL) was added to the reaction mixture, pH=5 to 6 was adjusted with 1M diluted hydrochloric acid, and extraction was performed with ethyl acetate (15 mL. Times.3). The combined organic phases were washed with saturated brine (30 mL) and concentrated under reduced pressure. The residue was purified by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile:% 40% -70%,7 min) to give compound 24.
MS m/z(ESI):540.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.72(br s,1H),8.41-8.23(m,2H),7.57(s,1H),6.35(s,1H),5.02(br d,J=8.8Hz,1H),4.56(dd,J=3.0,11.8Hz,1H),3.11-2.94(m,1H),2.78(s,3H),2.50-2.38(m,2H),2.37-2.28(m,2H),2.25(d,J=7.0Hz,2H),2.17-1.95(m,4H),1.27-1.06(m,2H).
Example 19
The synthetic route is as follows:
step 1: synthesis of Compound 25-b
To a thumb flask, under nitrogen, was added compound 20-b (50 mg, 109.27. Mu. Mol,1 eq), compound 25-a (21.07 mg, 163.91. Mu. Mol,1.5 eq) and dichloromethane (1.5 mL), followed by pyridine (43.22 mg, 546.37. Mu. Mol, 44.10. Mu.L, 5 eq) and phosphorus oxychloride (20.11 mg, 131.13. Mu. Mol, 12.19. Mu.L, 1.2 eq) and the mixture was stirred at 20℃for 1 hour. The reaction solution was concentrated under reduced pressure to give crude compound 25-b.
MS m/z(ESI):568.1[M+1] + .
Step 2: synthesis of Compound 25
In a thumb flask were added compound 25-b (100 mg, 176.02. Mu. Mol,1 eq) and methanol (2 mL), followed by aqueous sodium hydroxide (2M, 1mL,11.36 eq). The mixture was stirred at 25℃for 1 hour. To the reaction mixture was added water (5 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (15 ml×3). The combined organic phases were washed with saturated brine (30 mL) and concentrated under reduced pressure. The residue was separated by preparative high performance liquid chromatography (column form: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile:% 30% -60%,7 min) to give compound 25.
MS m/z(ESI):540.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.26(d,J=5.5Hz,1H),7.89(d,J=1.8Hz,1H),7.63(dd,J=1.8,5.8Hz,1H),7.57(s,1H),6.35(s,1H),5.01(dd,J=3.8,11.5Hz,1H),4.58(dd,J=3.5,11.5Hz,1H),3.02(br d,J=13.6Hz,1H),2.75(s,3H),2.47-2.29(m,4H),2.25(d,J=6.8Hz,2H),2.17-2.02(m,2H),1.98(br d,J=12.0Hz,2H),1.31-1.02(m,2H).
Example 20
The synthetic route is as follows:
step 1: synthesis of Compound 26-b
Compound 20-b (50 mg, 109.27. Mu. Mol,1 eq) and compound 26-a (22.31 mg, 163.91. Mu. Mol,1.5 eq) were dissolved in tetrahydrofuran (2 mL) under nitrogen, the reaction mixture was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (139.08 mg, 218.55. Mu. Mol, 129.98. Mu.L, 50% purity, 2 eq) and N, N-diisopropylethylamine (42.37 mg, 327.82. Mu. Mol, 57.10. Mu.L, 3 eq) were slowly added thereto, and the reaction mixture was heated to 50℃and stirred for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 26-b.
MS m/z(ESI):576.1[M+1] + .
Step 2: synthesis of Compound 26
Compound 26-b (20 mg, 34.74. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (6.95 mg, 173.71. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is separated by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile: acetonitrile%: 35% -65%,7 min) to obtain compound 26.
MS m/z(ESI):548.2[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.89-7.83(m,2H),7.55(s,1H),7.34-7.28(m,1H),6.33(s,1H),5.00(dd,J=3.6,11.7Hz,1H),4.52(dd,J=3.4,11.7Hz,1H),3.00(br d,J=14.6Hz,1H),2.75(s,3H),2.44-2.36(m,2H),2.34-2.25(m,2H),2.22(d,J=7.0Hz,2H),2.15-2.03(m,2H),1.97(br d,J=12.5Hz,2H),1.21-1.10(m,2H).
Example 21
The synthetic route is as follows:
step 1: synthesis of Compound 27-b
In a round bottom flask, compound 20-b (60 mg, 131.13. Mu. Mol,1 eq), 27-a (24.01 mg, 157.36. Mu. Mol,1.2 eq), tri-n-propyl cyclic phosphoric anhydride (166.89 mg, 262.26. Mu. Mol, purity: 50%,2 eq) and diisopropylethylamine (50.84 mg, 393.39. Mu. Mol, 68.52. Mu.L, 3 eq) were added to tetrahydrofuran (6 mL) and the reaction stirred at 50℃for 0.5 h. To the reaction solution was added water (50 mL), extracted with ethyl acetate (30 mL. Times.2), and the organic phases were combined, washed with water (30 mL) and saturated brine (30 mL) in this order, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/3) to give compound 27-b.
MS m/z(ESI):591.9[M+1] + .
Step 2: synthesis of Compound 27
Compound 27-b (40 mg, 67.55. Mu. Mol,1 eq) and sodium hydroxide (27.02 mg, 675.53. Mu. Mol,10 eq) were added to methanol (1 mL) and water (1 mL), and the reaction was stirred at 25℃for 1 hour. The reaction mixture was adjusted to ph=4 to 5 with 4M hydrochloric acid, water (30 mL) was added, and extraction was performed with ethyl acetate (30 ml×2). The combined organic phases were washed successively with water (20 mL) and saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product is purified by preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile: acetonitrile%: 45% -65%,7 min) to obtain compound 27.
MS m/z(ESI):564.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=8.18-7.87(m,2H),7.69-7.40(m,2H),6.35(s,1H),5.02(dd,J=3.4,11.7Hz,1H),4.54(dd,J=3.4,11.7Hz,1H),3.15-2.90(m,1H),2.77(s,3H),2.55-2.20(m,6H),2.15-1.88(m,4H),1.26-1.06(m,2H).
Example 22
The synthetic route is as follows:
step 1: synthesis of Compound 28-b
Compound 28-a (500.00 mg,2.37mmol,1 eq) was dissolved in t-butanol (10 mL), diphenyl azide phosphate (784.28 mg,2.85mmol, 617.54. Mu.L, 1.2 eq) and triethylamine (288.37 mg,2.85mmol, 396.66. Mu.L, 1.2 eq) were added, and the reaction mixture was heated to 90℃and stirred for 6 hours. Water (15 mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (15 mL. Times.3). The combined organic phases were washed with saturated brine (15 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 28-b.
1 H NMR(400MHz,CDCl 3 )δ=8.04(br d,J=11.5Hz,1H),6.73-6.56(m,1H),1.54(s,9H)
Step 2: synthesis of Compound 28-c
Compound 28-b (400.00 mg,1.42mmol,1 eq) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added, and the reaction was stirred at 25℃for 0.5 h. The reaction solution was concentrated under reduced pressure to give crude compound 28-c.
1 H NMR(400MHz,CDCl 3 )δ=6.54(td,J=7.6,11.2Hz,1H),3.75(br s,2H).
Step 3: synthesis of Compound 28-d
Compound 20-b (50 mg, 109.27. Mu. Mol,1 eq) and compound 28-c (29.76 mg, 163.9L. Mu. Mol,1.5 eq) were dissolved in tetrahydrofuran (2 mL) under nitrogen, the reaction solution was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (139.08 mg, 218.54. Mu. Mol,50% purity, 2 eq) and N, N-diisopropylethylamine (42.37 mg, 327.81. Mu. Mol, 57.10. Mu.L, 3 eq) were slowly added thereto, and the reaction solution was heated to 50℃and stirred for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 28-d.
MS m/z(ESI):621.1[M+1] + .
Step 4: synthesis of Compound 28
Compound 28-d (20.00 mg, 32.20. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (6.44 mg, 161.01. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is separated by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile: acetonitrile%: 40% -70%,7 min) to obtain compound 28.
MS m/z(ESI):593.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.92-7.85(m,1H),7.55(s,1H),6.33(s,1H),5.01(br d,J=8.5Hz,1H),4.55-4.50(m,1H),3.00(br d,J=13.3Hz,1H),2.78(s,3H),2.45-2.39(m,2H),2.35-2.26(m,2H),2.22(d,J=7.0Hz,2H),2.15-2.03(m,2H),1.96(br d,J=12.0Hz,2H),1.15(br d,J=12.5Hz,2H).
Example 23
The synthetic route is as follows:
step 1: synthesis of Compound 29-a
In a round bottom flask, 20-b (70 mg, 152.98. Mu. Mol,1 eq), 3-chloro-4-fluoroaniline (26.72 mg, 183.58. Mu. Mol,1.2 eq), tri-n-propylcyclic phosphoric anhydride (194.71 mg, 305.97. Mu. Mol, 181.97. Mu.L, concentration: 50%,2 eq) and diisopropylethylamine (79.09 mg, 611.94. Mu. Mol, 106.59. Mu.L, 4 eq) were added to tetrahydrofuran (5 mL), and the reaction mixture was stirred under nitrogen at 50℃for 1 hour. Water (50 mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (30 mL. Times.2). The combined organic phases were washed successively with water (30 mL) and saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 10/3) to give compound 29-a.
MS m/z(ESI):585.1[M+1] + .
Step 2: synthesis of Compound 29
Compound 29-a (50 mg, 85.45. Mu. Mol,1 eq) and sodium hydroxide (17.09 mg, 427.27. Mu. Mol,5 eq) were added to methanol (0.5 mL) and water (0.5 mL), and the reaction was stirred at 25℃for 60 min. The reaction mixture was adjusted to ph=4 to 5 with 4M diluted hydrochloric acid, water (30 mL) was added thereto, and extraction was performed with ethyl acetate (30 ml×2). The combined organic phases were washed successively with water (20 mL) and saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column form: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile: [ acetonitrile%: 40% -70%,7 min.) to give compound 29.
MS m/z(ESI):556.8[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.91(dd,J=2.6,6.7Hz,1H),7.58-7.49(m,2H),7.23(t,J=8.9Hz,1H),6.35(s,1H),5.02(dd,J=3.3,11.8Hz,1H),4.44(dd,J=3.3,11.8Hz,1H),3.11-2.91(m,1H),2.77(s,3H),2.51-2.19(m,6H),2.19-1.92(m,4H),1.29-1.01(m,2H).
Step 3: synthesis of Compounds 30 and 31
Compound 29 (20 mg, 35.90. Mu. Mol) was purified by SFC (column: DAICEL CH)IRALCEL OJ (250 mm x 30mm,10 μm); mobile phase: [0.1% NH 3 H 2 O,MeOH]Percent: 40% -40%) to give compounds 30 and 31.
SFC analysis method: column type: chiralCel OJ-H150X 4.6mm I.D.,5 μm; mobile phase: methanol (0.05% diethylamine); flow rate: 2.5mL/min.
Compound 30: MS m/z (ESI): 556.9[ M+1 ]] + SFC retention time: 7.427 minutes.
1H NMR(400MHz,CD 3 OD)δ=7.91(dd,J=2.6,6.7Hz,1H),7.68-7.44(m,2H),7.24(t,J=8.9Hz,1H),6.35(s,1H),5.02(dd,J=3.4,11.7Hz,1H),4.44(dd,J=3.3,11.8Hz,1H),3.07-2.93(m,1H),2.77(s,3H),2.51-2.26(m,4H),2.22(d,J=7.0Hz,2H),2.16-1.93(m,4H),1.28-1.08(m,2H).
Compound 31: MS m/z (ESI): 556.9[ M+1 ] ] + SFC retention time: 7.808 minutes.
1H NMR(400MHz,CD 3 OD)δ=7.91(dd,J=2.6,6.7Hz,1H),7.69-7.47(m,2H),7.24(t,J=9.0Hz,1H),6.35(s,1H),5.10-4.95(m,1H),4.44(dd,J=3.0,11.5Hz,1H),3.02(br d,J=14.1Hz,1H),2.77(s,3H),2.52-2.26(m,4H),2.22(br d,J=6.8Hz,2H),2.17-1.93(m,4H),1.27-1.08(m,2H).
Example 24
The synthetic route is as follows:
step 1: synthesis of Compound 32-a
Compound 20-b (50 mg, 109.27. Mu. Mol,1 eq) and compound 15-c (22.73 mg, 109.27. Mu. Mol,1 eq) were dissolved in tetrahydrofuran (2 mL) under nitrogen, the reaction mixture was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (139.08 mg, 218.55. Mu. Mol, 129.98. Mu.L, concentration: 50%,2 eq) and N, N-diisopropylethylamine (42.37 mg, 327.82. Mu. Mol, 57.10. Mu.L, 3 eq) were slowly added thereto, and the mixture was stirred for reaction for 12 hours while heating to 50 ℃. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 32-a.
MS m/z(ESI):647.0[M+1] + .
Step 2: synthesis of Compound 32
Compound 32-a (40 mg, 61.77. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (12.35 mg, 308.86. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is separated by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to obtain compound 32.
MS m/z(ESI):618.9[M+1] + .
Step 3: synthesis of Compounds 33 and 34
Compound 32 (25 mg, 40.36. Mu. Mol,1 eq) was purified by SFC (column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% NH) 3 H 2 O,EtOH]Percent: 35% -35%) to give compounds 33 and 34.
SFC analysis method: column type: chiralPak AS-3X 4.6mm I.D.,3 μm; mobile phase: isopropanol (0.05% diethylamine); flow rate: 2.5mL/min.
Compound 33: MS m/z (ESI): 619.0[ M+1 ]] + SFC retention time: 5.872 minutes, ee% = 92.5%.
1 H NMR(400MHz,CD 3 OD)δ=7.80-7.66(m,2H),7.55(s,1H),6.33(s,1H),4.99(dd,J=3.4,11.7Hz,1H),4.46(dd,J=3.5,11.8Hz,1H),3.00(br d,J=13.1Hz,1H),2.74(s,3H),2.45-2.27(m,4H),2.26-2.18(m,2H),2.15-2.00(m,2H),1.97(br d,J=11.5Hz,2H),1.22-1.10(m,2H).
Compound 34: MS m/z (ESI): 619.0[ M+1 ]] + SFC analysis method (column: chiralpak AD-3.150 x 4.6mm I.D.,3 μm; mobile phase: isopropanol (0.05% diethylamine; flow rate: 2.5 mL/min), retention time: 6.150 min, ee% = 84.8%.
1 H NMR(400MHz,CD 3 OD)δ=7.79-7.65(m,2H),7.55(s,1H),6.33(s,1H),4.99(dd,J=3.4,11.7Hz,1H),4.46(dd,J=3.5,11.8Hz,1H),3.00(br d,J=13.8Hz,1H),2.74(s,3H),2.45-2.27(m,4H),2.22(d,J=6.8Hz,2H),2.15-2.00(m,2H),1.99-1.93(m,2H),1.21-1.09(m,2H).
Example 25
The synthetic route is as follows:
step 1: synthesis of Compound 35-a
Compound 20-b (325 mg, 710.28. Mu. Mol,1 eq) and compound 17-a (174.25 mg,1.07mmol,1.5 eq) were dissolved in methylene chloride (15 mL) under nitrogen, and after cooling the reaction solution to 0℃phosphorus oxychloride (163.36 mg,1.07mmol, 99.01. Mu.L, 1.5 eq) and pyridine (280.92 mg,3.55mmol, 286.65. Mu.L, 5 eq) were slowly added thereto, and the reaction solution was stirred at 0℃for 1 hour. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 35-a.
MS m/z(ESI):603.1[M+1] + .
Step 2: synthesis of Compounds 35-b and 35-c
Compound 35-a (220 mg, 364.78. Mu. Mol,1 eq) was purified by SFC (column: DAICEL CHIRALCEL OJ (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% NH) 3 H 2 O,EtOH]Isolation of%40% -40%) gives compounds 35-b and 35-c.
Compound 35-b: SFC analysis method (column: chiralCel OJ-H150X 4.6mm I.D.,5um; mobile phase: ethanol (0.05% diethylamine; flow rate: 2.5 mL/min); MS m/z (ESI): 603.1[ M+1 ]] + SFC retention time: 5.635 minutes, ee% = 100%.
Compound 35-b: MS m/z (ESI): 603.1[ M+1 ]] + SFC retention time: for 6.192 min, ee% = 98.8%.
Step 3: synthesis of Compound 35
Compound 35-b (90 mg, 149.23. Mu. Mol,1 eq) was dissolved in methanol (3 mL) and water (3 mL), and sodium hydroxide (29.84 mg, 746.14. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to give compound 35.
SFC analysis method: column type: chiralCel OJ-H150X 4.6mm I.D.,5 μm; mobile phase: ethanol (0.05% diethylamine); flow rate: MS m/z (ESI) at 2.5 mL/min: 575.1[ M+1 ]] + SFC retention time: 5.950 min, ee% = 98.3%.
1 H NMR(400MHz,CD 3 OD)δ=7.68(dt,J=5.8,8.7Hz,1H),7.55(s,1H),7.15(dt,J=2.3,8.9Hz,1H),6.34(s,1H),5.02(dd,J=3.1,12.2Hz,1H),4.51(dd,J=3.0,12.0Hz,1H),3.00(br d,J=13.6Hz,1H),2.80(s,3H),2.47-2.40(m,2H),2.37-2.27(m,2H),2.22(d,J=7.0Hz,2H),2.16-2.01(m,2H),1.97(br d,J=12.3Hz,2H),1.23-1.09(m,2H).
Step 4: synthesis of Compound 36
Into a thumb flask were added compound 35-c (100 mg, 165.81. Mu. Mol,1 eq) and methanol (2 mL), followed by aqueous sodium hydroxide (2M, 1mL,12.06 eq) and stirring at 25℃for 30 min. The reaction mixture was adjusted to ph=1 to 2 with 1M diluted hydrochloric acid and extracted with ethyl acetate (15 ml×3). The combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was separated by preparative high performance liquid chromatography (column form: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 35% -65%,7 min). Compound 36 was obtained.
MS m/z(ESI):575.1[M+1] + SFC retention time: 6.251 minutes, ee% = 100%.
1 H NMR(400MHz,CD 3 OD)δ=7.74-7.65(m,1H),7.57(s,1H),7.17(dt,J=2.0,8.9Hz,1H),6.35(s,1H),5.04(dd,J=3.3,12.0Hz,1H),4.53(dd,J=3.0,12.0Hz,1H),3.02(br d,J=14.8Hz,1H),2.82(s,3H),2.51-2.40(m,2H),2.39-2.29(m,2H),2.24(d,J=6.8Hz,2H),2.18-1.94(m,4H),1.28-1.04(m,2H).
Example 26
The synthetic route is as follows:
step 1: synthesis of Compound 37-a
Compound 20-b (45 mg, 98.35. Mu. Mol,1 eq) and compound 18-a (30.68 mg, 147.52. Mu. Mol,1.5 eq) were dissolved in tetrahydrofuran (2 mL) under nitrogen, the reaction mixture was cooled to 0℃and then tri-N-propylcyclic phosphoric anhydride (125.17 mg, 196.69. Mu. Mol, 116.98. Mu.L, concentration: 50%,2 eq) and N, N-diisopropylethylamine (38.13 mg, 295.04. Mu. Mol, 51.39. Mu.L, 3 eq) were added thereto, and the reaction mixture was stirred at 50℃for 12 hours. The reaction solution was concentrated directly under reduced pressure. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/0 to 2/1) to give compound 37-a.
MS m/z(ESI):647.1[M+1] + .
Step 2: synthesis of Compound 37
Compound 37-a (20 mg, 30.89. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (6.18 mg, 154.43. Mu. Mol,5 eq) was added thereto, and the reaction mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to give compound 37.
MS m/z(ESI):618.8[M+1] + .
1 H NMR(400MHz,DMSO-d6)δ=7.71-7.60(m,2H),7.37-7.23(m,1H),6.37(s,1H),4.91(br d,J=10.0Hz,1H),4.44(dd,J=2.5,12.0Hz,1H),2.87(br d,J=14.1Hz,1H),2.66(s,3H),2.42-2.33(m,2H),2.26-2.12(m,4H),2.09-1.84(m,4H),1.17-1.05(m,1H).
Example 27
The synthetic route is as follows:
step 1: synthesis of Compound 38-a
Compound 20-a (0.3 g, 842.18. Mu. Mol,1 eq), compound 1-o (371.65 mg,1.26mmol,1.5 eq), N, N-diisopropylethylamine (544.22 mg,4.21mmol, 733.45. Mu.L, 5 eq) and dichlorobis (triphenylphosphine) palladium (59.11 mg, 84.22. Mu. Mol,0.1 eq) were dissolved in 1, 4-dioxane (4 mL) and water (4 mL) under nitrogen and the mixture was stirred at 80℃for 12 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give compound 38-a, the crude product was used directly in the next reaction.
MS m/z(ESI):444.1[M+1] + .
Step 2: synthesis of Compound 38-b
Compound 38-a (100 mg, 225.46. Mu. Mol,1 eq) and compound 17-a (55.31 mg, 338.19. Mu. Mol,1.5 eq) were dissolved in methylene chloride (3 mL), the reaction solution was cooled to 0℃and phosphorus oxychloride (69.14 mg, 450.92. Mu. Mol, 41.90. Mu.L, 2 eq) and pyridine (89.17 mg,1.13mmol, 90.99. Mu.L, 5 eq) were slowly added and stirred at 0℃for 1 hour. The reaction solution was concentrated under reduced pressure to give crude compound 38-b.
MS m/z(ESI):589.1[M+1] + .
Step 3: synthesis of Compound 38
Compound 38-b (30 mg, 50.93. Mu. Mol,1 eq) was dissolved in methanol (1 mL) and water (1 mL), and sodium hydroxide (10.19 mg, 254.65. Mu. Mol,5 eq) was added thereto and the mixture was stirred at 25℃for 2 hours. To the reaction mixture was added water (10 mL), which was then adjusted to ph=1-2 with 1M diluted hydrochloric acid, followed by extraction with ethyl acetate (10 ml×3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is separated by a preparative high performance liquid chromatography (column type: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 40% -70%,7 min) to obtain compound 38.
MS m/z(ESI):561.1[M+1] + .
1H NMR(400MHz,CD 3 OD)δ=7.68(ddd,J=5.6,8.2,9.2Hz,1H),7.57(s,1H),7.15(dt,J=2.0,8.9Hz,1H),6.37(s,1H),5.02(dd,J=3.0,12.0Hz,1H),4.51(dd,J=3.0,12.0Hz,1H),2.96(br d,J=13.8Hz,1H),2.83-2.76(m,3H),2.63-2.52(m,1H),2.45(td,J=3.4,13.9Hz,2H),2.37-2.17(m,3H),2.12-2.04(m,2H),1.70-1.54(m,2H).
Example 28
The synthetic route is as follows:
step 1: synthesis of Compound 39-a
The compound 20-b (50 mg, 109.27. Mu. Mol,1 eq), 16-a (24.11 mg, 163.91. Mu. Mol, 17.35. Mu. L,1.5 eq) and methylene chloride (1.5 mL) were added to a thumb flask under nitrogen, and then pyridine (43.22 mg, 546.37. Mu. Mol, 44.10. Mu.L, 5 eq) and phosphorus oxychloride (20.11 mg, 131.13. Mu. Mol, 12.19. Mu.L, 1.2 eq) were added, and the mixture was stirred at 0℃for 1 hour. The reaction solution was concentrated under reduced pressure to give crude compound 39-a.
MS m/z(ESI):587.1[M+1] + .
Step 2: synthesis of Compound 39
In a thumb flask were added compound 39-a (60 mg, 102.28. Mu. Mol,1 eq) and methanol (1 mL), followed by sodium hydroxide (2M, 0.5mL,9.78 eq) and then stirred at 25℃for 1 hour. The reaction mixture was adjusted to ph=1 to 2 with 1M diluted hydrochloric acid and extracted with ethyl acetate (15 ml×3). The combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was separated by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 35% -65%,7 min) to give compound 39.
MS m/z(ESI):559.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.56(s,1H),7.54-7.47(m,1H),7.22-7.09(m,1H),6.35(s,1H),5.03(dd,J=3.1,11.9Hz,1H),4.53(dd,J=3.0,12.0Hz,1H),3.02(br d,J=14.1Hz,1H),2.81(s,3H),2.51-2.40(m,2H),2.38-2.28(m,2H),2.24(d,J=7.0Hz,2H),2.17-1.95(m,4H),1.29-1.07(m,2H).
Step 3: synthesis of Compounds 40 and 41
Compound 39 (25 mg, 44.76. Mu. Mol,1 eq) was purified by chiral SFC (column: DAICEL CHIRALCEL OJ-H (250 mm. Times.30 mm,5 μm); mobile phase: [0.1% NH) 3 H 2 O,MeOH]Percent: 40% -40%) to give compounds 40 and 41.
SFC analysis method: column type: chiralcel OJ-3.150 x 4.6mm I.D.,3 μm; mobile phase: methanol (0.05% diethylamine); flow rate: 2.5mL/min.
Compound 40: MS m/z (ESI): 558.9[ M+1 ]] + SFC retention time: 1.864 minutes, ee% = 96.2%.
1 H NMR(400MHz,CD 3 OD)δ=7.57(s,1H),7.54-7.47(m,1H),7.21-7.12(m,1H),6.35(s,1H),5.04(dd,J=3.1,11.9Hz,1H),4.53(dd,J=3.0,12.0Hz,1H),3.02(br d,J=13.8Hz,1H),2.81(s,3H),2.49-2.41(m,2H),2.39-2.27(m,2H),2.24(d,J=7.0Hz,2H),2.18-2.03(m,2H),1.99(br d,J=12.8Hz,2H),1.27-1.07(m,2H).
Compound 41: MS m/z (ESI): 558.9[ M+1 ] ] + SFC retention time: 2.307 minutes, ee% = 80.5%.
1 H NMR(400MHz,CD 3 OD)δ=7.56(s,1H),7.55-7.47(m,1H),7.22-7.12(m,1H),6.35(s,1H),5.04(dd,J=3.1,11.9Hz,1H),4.53(dd,J=3.0,12.0Hz,1H),3.02(br d,J=13.3Hz,1H),2.81(s,3H),2.50-2.40(m,2H),2.39-2.27(m,2H),2.22(br d,J=7.0Hz,2H),2.17-2.02(m,2H),1.99(br d,J=11.8Hz,2H),1.24-1.10(m,2H)
Example 29
The synthetic route is as follows:
step 1: synthesis of Compound 42-a
To a thumb flask were added 20-b (50 mg, 109.27. Mu. Mol,1 eq), 2-fluoro-3-chloroaniline (23.86 mg, 163.91. Mu. Mol, 26.02. Mu. L,1.5 eq) and dichloromethane (1.5 mL) under nitrogen, and then pyridine (43.22 mg, 546.37. Mu. Mol, 44.10. Mu. L,5 eq) and phosphorus oxychloride (20.11 mg, 131.13. Mu. Mol, 12.19. Mu. L,1.2 eq) and the mixture was stirred at 0℃for 1 hour. The reaction solution was concentrated under reduced pressure to obtain compound 42-a.
MS m/z(ESI):585.1[M+1] + .
Step 2: synthesis of Compound 42
In a thumb flask were added compound 42-a (60 mg, 102.54. Mu. Mol,1 eq) and methanol (1 mL), followed by sodium hydroxide (2M, 0.5mL,9.75 eq) and then stirred at 25℃for 1 hour. The reaction mixture was adjusted to ph=1 to 2 with 1M diluted hydrochloric acid and extracted with ethyl acetate (15 ml×3). The combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was separated by preparative high performance liquid chromatography (column form Phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ water (0.225% formic acid) -acetonitrile ]; acetonitrile%: 35% -65%,7 min) to give compound 42.
MS m/z(ESI):557.0[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ=7.75(ddd,J=1.6,6.8,8.2Hz,1H),7.57(s,1H),7.35(ddd,J=1.5,6.8,8.3Hz,1H),7.19(dt,J=1.6,8.2Hz,1H),6.35(s,1H),5.04(dd,J=3.0,12.0Hz,1H),4.53(dd,J=3.0,12.0Hz,1H),3.02(br d,J=13.6Hz,1H),2.82(s,3H),2.53-2.29(m,4H),2.25(d,J=6.8Hz,2H),2.18-1.96(m,4H),1.27-1.07(m,2H).
Step 3: synthesis of Compounds 43 and 44
Compound 42 (25 mg, 44.88. Mu. Mol,1 eq) was purified by SFC (column: DAICEL CHIRALCEL OJ (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% NH) 3 H 2 O,MeOH]Percent: 40% -40%) to give compounds 43 and 44.
SFC analysis method: column type: chiralcel OJ-3.150 x 4.6mm I.D.,3 μm; mobile phase: methanol (0.05% diethylamine); flow rate: 2.5mL/min.
Compound 43: MS m/z (ESI): 556.9[ M+1 ]] + SFC retention time: 3.996 min, ee% = 98.3%.
1 H NMR(400MHz,CD 3 OD)δ=7.69-7.60(m,1H),7.45(s,1H),7.23(dt,J=1.5,7.4Hz,1H),7.07(dt,J=1.6,8.2Hz,1H),6.23(s,1H),4.92(dd,J=3.3,12.0Hz,1H),4.41(dd,J=3.0,12.0Hz,1H),2.90(br d,J=13.8Hz,1H),2.82(s,3H),2.38-2.16(m,4H),2.12(d,J=7.0Hz,2H),2.05-1.83(m,4H),1.14-0.92(m,2H).
Compound 44: MS m/z (ESI): 557.0[ M+1 ]] + SFC retention time: 5.221 min, ee% = 98.4%.
1 H NMR(400MHz,CD 3 OD)δ=7.75(t,J=7.4Hz,1H),7.56(s,1H),7.35(dt,J=1.4,7.5Hz,1H),7.19(dt,J=1.5,8.2Hz,1H),6.35(s,1H),5.04(br d,J=9.3Hz,1H),4.53(dd,J=2.8,12.0Hz,1H),3.02(br d,J=14.3Hz,1H),2.82(s,3H),2.51-2.39(m,2H),2.39-2.28(m,2H),2.24(d,J=7.0Hz,2H),2.18-2.08(m,1H),2.05(dt,J=3.5,7.3Hz,1H),1.98(br d,J=12.3Hz,2H),1.26-1.06(m,2H).
Biological Activity test
Experimental example 1: quantitative qPCR assay for HBV in vitro test
1 purpose of experiment:
detection of HBV DNA content in HepG2.2.15 cells by real time quantitative qPCR assay (real time-qPCR) to compounds EC 50 The inhibition of HBV by the compounds was evaluated using the values as indicators.
2 experimental materials:
2.1 cell lines: hepG2.2.15 cells
HepG2.2.15 cell culture medium (DMEM/F12, invitrogen-11330057;10% serum, invitrogen-10099141;100units/ml penicillin and 10. Mu.g/ml streptomycin, invitrogen-15140122;1% nonessential amino acids, invitrogen-11140076;2mM L-glutamine, invitrogen-25030081; 300. Mu.g/ml geneticin, invitrogen-10131027).
2.2 reagents:
pancreatin (Invitrogen-25300062)
DPBS(Hyclone-SH30028.01B)
DMSO(Sigma-D2650-100ML)
High flux DNA purification kit (QIAamp 96 DNA Blood Kit,Qiagen-51162)
Quantitative quick-start universal probe reagent (FastStart Universal Probe Master, roche-04914058001)
2.3 consumables and instrumentation:
96-well cell culture plate (Coming-3599)
CO 2 Incubator (HERA-CELL-240)
Optical close plate film (ABI-4311971)
Quantitative PCR 96-well plate (Applied Biosystems-4306737)
Fluorescent quantitative PCR instrument (Applied Biosystems-7500 real time PCR system)
3. Experimental procedure and method:
3.1 HepG2.2.15 cells (4X 104 cells/well) to 96 well plates at 37℃with 5% CO 2 Culturing overnight.
3.2 the next day, the compound was diluted, total 8 concentrations, 3-fold gradient dilution. Adding different concentrations of compounds into the culture wells, and doubling the wells. The final concentration of DMSO in the culture was 1%.1 μM GLS4 served as 100% inhibition control; 1% DMSO was used as a 0% inhibition control.
3.3 day five, fresh medium containing the compound was changed.
3.4 culture broth from culture wells was harvested on day eight and DNA was extracted using a high throughput DNA purification kit (Qiagen-51162) for specific procedures with reference to the product instructions.
3.5 preparation of PCR reaction solution is shown in Table 1:
TABLE 1 preparation of PCR reaction solution
Upstream primer sequence: GTGTCTGCGGCGTTTTATCA
Downstream primer sequence: GACAAACGGGCAACATACCTT
Probe sequence: 5'+FAM+ CCTCTKCATCCTGCTGCTATGCCTCATC +TAMRA-3'
3.6 in 96 well PCR plate each well to 15. Mu.L of reaction mixture, then each well to 10. Mu.L of sample DNA or HBV DNA standard.
3.7 The reaction conditions for PCR were: heating at 95 ℃ for 10 minutes; then denatured at 95℃for 15 seconds and extended at 60℃for 1 minute for 40 cycles.
3.8 data analysis:
3.8.1 percent inhibition was calculated: % inh= [ 1- (copy number of DNA in sample-1 μm copy number of DNA in GLS 4)/(copy number of DNA in DMSO control-1 μm copy number of DNA in GLS 4) ]x100.
3.8.2 calculation of EC 50 : the 50% inhibitory concentration (EC) of compounds against HBV was calculated using GraphPad Prism software 50 ) Values.
4 experimental results are shown in table 2:
TABLE 2 qPCR assay to detect EC 50 Test results
Conclusion: the compound has remarkable inhibition effect on HBV.
Experimental example 2: hepatotoxicity test experiment
2.1: PHH cytotoxicity test experiment
1. Compounds were diluted in DMSO (dimethyl sulfoxide) at 3-fold gradient at 8 spots, double-multiplexed wells, and added to 96-well plates. The compound concentration was 200 times the final test concentration.
2. The cryopreserved human primary hepatocytes (PHH) were recovered, the cell density was counted with a cytometer, and the cell density was adjusted to the desired density with the medium.
3. Cells were added to 96-well plates to which the compounds had been added, with a final concentration of DMSO in each well of 0.5%. Cell wells containing 0.5% dmso were used as non-toxic negative controls, and cell broth wells were used as 100% cytotoxicity controls. The cell plates were then placed at 37℃in 5% CO 2 The cells were cultured in the incubator for 3 days.
4. The chemiluminescent signal (RLU, relative chemiluminescent unit) of each well in the cell plate was detected with a cell viability detection kit CellTiter-Glo, using a multifunctional enzyme-labeled instrument Synegy2-BioTek according to kit instructions.
5. The cell viability (cell viability%) of each well tested was calculated by substituting the raw data (RLU) into the following formula:
cell viability% = (RLU Sample -AverageRLU Mediumcontrol /(AverageRLU Cellcontrol -AverageRLU Mediumcontrol )×100%
RLU Sample Signal values for sample wells; averageRLU Cellcontrol Mean value of cell control well signal; averageRLU Mediumcontrol Mean value of medium control well signals.
6. Using GraphPad Prism software, non-linear fitting of cell viability data to a dose-response curve, and deriving the half-cytotoxicity concentration (CC 50 ) Values, results are shown in Table 3.
TABLE 3 half-cell toxicity concentration (CC 50 ) Value test results
Compounds of formula (I) CC 50 (μM)
Compound 11 >100
Conclusion: cytotoxic CC of the inventive Compounds in Primary hepatocytes 50 The value is relatively high.
2.2: huh7 cytotoxicity test experiments
1. Compounds were diluted in DMSO (dimethyl sulfoxide) at 3-fold gradient at 8 spots, double-multiplexed wells, and added to 96-well plates. The compound concentration was 200 times the final test concentration.
2. Washing the cells with PBS (phosphate buffer), adding 0.25% pancreatin at 37deg.C, 5% CO 2 After digestion for about 2-5 minutes in the incubator, the digestion was stopped with cell culture medium and the cells were dispersed into single cells by blowing with a gun.
3. Cell densities were counted with a cytometer and adjusted to the desired densities with medium.
4. Cells were added to 96-well plates to which the compounds had been added, with a final concentration of DMSO in each well of 0.5%. Cell holes containing 0.5% DMSO are used as nontoxic negative control, and cell culture solution holes are used as 100%Cytotoxicity control. The cell plates were then placed at 37℃in 5% CO 2 The cells were cultured in the incubator for 3 days.
5. The chemiluminescent signal (RLU, relative chemiluminescent unit) of each well in the cell plate was detected with a cell viability detection kit CellTiter-Glo, using a multifunctional enzyme-labeled instrument Synegy2-BioTek according to kit instructions.
6. The cell viability (cell viability%) of each well tested was calculated by substituting the raw data (RLU) into the following formula:
Cell viability% = (RLU Sample -AverageRLU Mediumcontrol /(AverageRLU Cellcontrol -AverageRLU Mediumcontrol )×100%
RLU Sample Signal values for sample wells; averageRLU Cellcontrol Mean value of cell control well signal; averageRLU Mediumcontrol Mean value of medium control well signals.
7. Non-linear fitting of cell viability data to dose-response curves using GraphPad Prism software and derivation of half-cytotoxicity concentration of compounds (CC 50 ) Values, results are shown in table 4.
TABLE 4 half-cell toxicity concentration (CC 50 ) Value test results
Compounds of formula (I) CC 50 (μM)
Compound 11 86.71
Conclusion: cytotoxic CC of the compound in liver cancer cell Huh7 50 The value is relatively high.
2.3: hepG2 cytotoxicity test experiment
1. Compounds were diluted in DMSO (dimethyl sulfoxide) at 3-fold gradient at 8 spots, double-multiplexed wells, and added to 96-well plates. The compound concentration was 200 times the final test concentration.
2. Washing the cells with PBS (phosphate buffer), adding 0.25% pancreatin at 37deg.C, 5% CO 2 After digestion for about 2-5 minutes in the incubator, the digestion was stopped with cell culture medium and the cells were dispersed into single cells by blowing with a gun.
3. Cell densities were counted with a cytometer and adjusted to the desired densities with medium.
4. Cells were added to 96-well plates to which the compounds had been added, with a final concentration of DMSO in each well of 0.5%. Cell wells containing 0.5% dmso were used as non-toxic negative controls, and cell broth wells were used as 100% cytotoxicity controls. The cell plates were then placed at 37℃in 5% CO 2 The cells were cultured in the incubator for 3 days.
5. The chemiluminescent signal (RLU, relative chemiluminescent unit) of each well in the cell plate was detected with a cell viability detection kit CellTiter-Glo, using a multifunctional enzyme-labeled instrument Synegy2-BioTek according to kit instructions.
6. The cell viability (cell viability%) of each well tested was calculated by substituting the raw data (RLU) into the following formula:
cell viability% = (RLU Sample -AverageRLU Mediumcontrol /(AverageRLU Cellcontrol -AverageRLU Mediumcontrol )×100%
RLU Sample Signal values for sample wells; averageRLU Cellcontrol Mean value of cell control well signal; averageRLU Mediumcontrol Mean value of medium control well signals.
7. Nonlinear fitting of cell viability data to dose-response curves using GraphPad Prism softwareLine, and resulting in half-cytotoxicity concentration (CC 50 ) Values, results are shown in Table 5.
TABLE 5 half-cell toxicity concentration (CC 50 ) Value test results
Compounds of formula (I) CC 50 (μM)
Compound 11 79.35
Conclusion: cytotoxic CC of the compound in liver cancer cell HepG2 50 The value is relatively high.
Experimental example 3: pharmacokinetic studies
Pharmacokinetic study of Balb/c mice orally and intravenously injected test compounds:
the tested compound 11 is mixed with 10 percent polyethylene glycol-15 hydroxystearate/20 percent polyethylene glycol 400/70 percent aqueous solution, vortexed and sonicated to prepare 0.2mg/mL clear solution, and the clear solution is filtered by a microporous filter membrane for later use. Balb/c female mice 7 to 10 weeks old were selected and given a solution of the candidate compound by intravenous injection at a dose of 1mg/kg.
The tested compound 30 is mixed with 10% dimethyl sulfoxide/10% polyethylene glycol-15 hydroxystearate/80% aqueous solution, vortexed and sonicated to prepare a clear solution of 0.2mg/mL, and the clear solution is filtered through a microporous filter membrane for later use. Balb/c female mice 7 to 10 weeks old were selected and given a solution of the candidate compound by intravenous injection at a dose of 1mg/kg.
Test compound 36 was mixed with 10% dimethyl sulfoxide/10% polyethylene glycol-15 hydroxystearate/80% aqueous solution, vortexed and sonicated to prepare a clear solution of 0.2mg/mL, and filtered through a microfiltration membrane for use. Balb/c female mice 7 to 10 weeks old were selected and given a solution of the candidate compound by intravenous injection at a dose of 1mg/kg.
Test compounds 11, 30 and 36 were each mixed with 10% aqueous polyethylene glycol-15 hydroxystearate, vortexed and sonicated to prepare 1mg/mL homogeneous suspensions for use. Balb/c female mice 7 to 10 weeks old were selected and the candidate compound solution was orally administered at a dose of 10mg/kg.
Whole blood was collected for a certain period of time, plasma was prepared, drug concentration was analyzed by LC-MS/MS method, and drug substitution parameters were calculated by Phoenix WinNonlin software (Pharsight Co., USA) and the results are shown in Table 6.
TABLE 6 pharmacokinetic results of test compounds
Conclusion: the compound of the invention has high oral bioavailability.
Experimental example 4: in vivo liver-blood ratio study of mice
Liver-to-blood ratio study of oral test compounds in Balb/c mice
The compound is mixed with 10% polyethylene glycol-15 hydroxystearate aqueous solution, vortexed and sonicated to prepare 1mg/mL uniform suspension for use. Balb/c female mice 7 to 10 weeks old were selected and the candidate compound solution was orally administered at a dose of 10mg/kg.
Whole blood was collected for a certain period of time, plasma was prepared, liver tissue was collected for a corresponding period of time, tissue homogenate was prepared, drug concentration was analyzed by LC-MS/MS method, and drug substitution parameters were calculated by Phoenix WinNonlin software (Pharsight, usa). The results are shown in Table 7.
TABLE 7 liver to blood ratio results for test compounds
Conclusion: the ratio of the compounds of the present invention in liver and plasma is relatively high.
Experimental example 5: in vivo efficacy study
HDI/HBV model
The purpose of the experiment is as follows: the anti-hepatitis B virus effect of the compounds in mice was tested by the HDI/HBV mouse model.
Compound preparation: the solvent is 10% polyethylene glycol-15 hydroxystearate; a certain amount of test compounds 11, 30 and 36 were dissolved in 10% aqueous polyethylene glycol-15 hydroxystearate, respectively, vortexed and sonicated to prepare a uniform suspension, which was stored at 4 ℃ for use.
Mice were injected with HBV plasmid DNA solution at high pressure at tail vein: the day of plasmid injection was designated as day 0, day 1 after injection was designated as day 1, and so on. All animals were injected by tail vein with a physiological saline solution containing 10. Mu.g of plasmid DNA at 8% of the body weight on day 0, and the injection was completed within 5 seconds.
Administration: all animals were dosed twice daily by gavage (8/16 hours apart) on days 1-6, once on day 7, and all animals were euthanized at afternoon on day 7. The mice were monitored daily for body weight, which remained stable throughout the experiment.
Sample collection: all animals were collected on day 5 in the morning four hours following the first dose by submaxillary vein blood collection and plasma, all blood samples were collected on K 2 In EDTA anticoagulant tube, at 4℃7000g is centrifuged for 10 min to prepare about 40. Mu.L of plasma. Day 7 all animals were CO-dosed four hours after morning dosing 2 Euthanasia, heart blood sampling and plasma preparation method are the same. Two liver tissues, 70-100mg each, were collected and snap frozen with liquid nitrogen. After all samples were collected, they were stored in a-80℃refrigerator until dry ice conditions were reached and sent to the Ming Kangde department of biology in vitro laboratory for HBV DNA content detection.
Sample analysis: all plasma and liver samples were assayed for HBV DNA by qPCR.
Experimental results: the experimental results are shown in table 8.
TABLE 8 results of HDI efficacy experiments
Note that: deltaLog 10 copies represents the difference in viral load of the dosed group and the viral load of the vehicle group
Conclusion: the compound has remarkable effect of reducing HBV DNA in an HDI model.

Claims (19)

  1. A compound represented by the formula (I) or a pharmaceutically acceptable salt thereof,
    wherein,
    R 1 are each independently selected from halogen, OH, CN, NH 2 、C 1-3 Alkyl and C 1-3 Alkoxy group, the C 1-3 Alkyl and C 1-3 Alkoxy groups are each independently optionally substituted with 1, 2 or 3 halogens;
    m is selected from 0, 1, 2, 3 and 4;
    T 1 and T 2 Each independently selected from CH and N;
    is a single bond or a double bond;
    ring A is selected from phenyl, C 4-10 Cycloalkyl, the phenyl and C 4-10 Cycloalkyl groups are each independently optionally substituted with 1, 2Or 3R a Substitution;
    R a are each independently selected from halogen, OH, CN, NH 2 、C 1-3 Alkyl and C 1-3 Alkoxy group, the C 1-3 Alkyl and C 1-3 Alkoxy groups are each independently optionally substituted with 1, 2 or 3 halogens;
    L 1 selected from single bond, -C.ident.C-and-CR 2 =;
    R 2 Selected from H, methyl and F;
    L 2 selected from single bonds and methylene groups.
  2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 1 Are respectively and independently selected from F, cl, br, CN and-CH 3 and-OCH 3 the-CH 3 and-OCH 3 Each independently optionally substituted with 1, 2 or 3F.
  3. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R 1 Each independently selected from F, cl, br and CN.
  4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein L 1 Selected from-c≡c-, -ch=, -C (F) =, and-C (CH) 3 )=。
  5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R a Are respectively and independently selected from F, cl, br, CH 3 And OCH 3 The CH is 3 And OCH 3 Each independently optionally substituted with 1, 2 or 3F.
  6. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, whereinStructural unitSelected from the group consisting of
  7. The compound of claim 6, or a pharmaceutically acceptable salt thereof, wherein the structural unitSelected from the group consisting of
  8. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from phenyl, cyclobutyl, cyclohexyl, The phenyl, cyclobutyl, cyclohexyl,Are each independently optionally substituted with 1, 2 or 3R a And (3) substitution.
  9. The compound of claim 8, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from
  10. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the structural unitSelected from the group consisting of
  11. A compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
    Wherein R is 1 、R 2 、L 2 And m is as defined in any one of claims 1 to 5.
  12. The compound of claim 11, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
    wherein R is 1 、R 2 、L 2 And m is as defined in claim 11.
  13. The compound of claim 12, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
    wherein R is 1 、R 2 、L 2 And m is as defined in claim 12.
  14. A compound of the formula:
  15. the compound of claim 14, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
  16. the compound of claim 15, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
  17. the compound of claim 16, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
  18. the use of a compound according to any one of claims 1 to 17 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a disease associated with hepatitis b virus infection.
  19. The use according to claim 18, wherein the disease associated with hepatitis b virus infection is chronic hepatitis b.
CN202280033410.9A 2021-05-07 2022-05-05 Methylsulfonyl urea compound and application thereof Pending CN117337286A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202110494696 2021-05-07
CN202110494696.8 2021-05-07
PCT/CN2022/091012 WO2022233305A1 (en) 2021-05-07 2022-05-05 Methylsulfonylurea compound and application thereof

Publications (1)

Publication Number Publication Date
CN117337286A true CN117337286A (en) 2024-01-02

Family

ID=83932008

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280033410.9A Pending CN117337286A (en) 2021-05-07 2022-05-05 Methylsulfonyl urea compound and application thereof

Country Status (2)

Country Link
CN (1) CN117337286A (en)
WO (1) WO2022233305A1 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2019010404A (en) * 2017-03-02 2019-11-21 Assembly Biosciences Inc Cyclic sulfamide compounds and methods of using same.
WO2019206072A1 (en) * 2018-04-24 2019-10-31 浙江海正药业股份有限公司 Sulfamide aryl formamide derivative and preparation method therefor and uses thereof
CN113166086A (en) * 2018-09-05 2021-07-23 组装生物科学股份有限公司 Cyclic sulfonamide compounds for the treatment of HBV
WO2020051320A1 (en) * 2018-09-05 2020-03-12 Assembly Biosciences, Inc. Cyclic sulfamide compounds for treatment of hbv

Also Published As

Publication number Publication date
WO2022233305A1 (en) 2022-11-10

Similar Documents

Publication Publication Date Title
EP4166142A1 (en) Methyl-substituted benzobisoxazole compound and use thereof
US7910745B2 (en) Organic compound and method for producing radioactive halogen-labeled organic compound using the same
WO1992012991A1 (en) Triterpene derivative
RO111847B1 (en) Optical izomer for the //4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)-phenyl/hydr azono/propane dinitrile, preparation process thereof and intermediaries for the process making thereof
JP7287978B2 (en) two 4-{[(2S)-2-{4-[5-chloro-2-(1H-1,2,3-triazol-1-yl)phenyl]-5-methoxy-2-oxopyridine-1 Method for preparing (2H)-yl}butanoyl]amino}-2-fluorobenzamide derivatives
CN116348466A (en) Pyrazine sulfur biphenyl compound and application thereof
WO1995022521A1 (en) Aminoalkylcyclopropane derivative
WO2020052647A1 (en) Spiro-heterocyclic compound acting as lsd1 inhibitor and use thereof
TWI807697B (en) Furan fused ring substituted glutarimide compounds
CN117337278A (en) Alkyl carboxylic acid compounds and uses thereof
TW202317097A (en) Glutarimide compounds and applications thereof
CN114846008B (en) Pyrimidine compound with fructokinase (KHK) inhibiting effect
EP4105214A1 (en) Use of jak inhibitors in preparation of drugs for treating jak kinase-related diseases
CN117337286A (en) Methylsulfonyl urea compound and application thereof
CN116425770A (en) Tetrafused ring compounds as Cdc7 inhibitors
TWI823274B (en) Substituted pyridine-2,4-dione derivatives
JP7340680B2 (en) Oxazole compounds as IRAK4 and BTK multi-target inhibitors
CN115003672A (en) Quinoline imidazole compound and application thereof
PT92874A (en) METHOD FOR PREPARING {(5-OXO-2-PYRROLIDINYL) -METHYL} -CYCLOHEXANOACETAMIDES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
CN111777555B (en) Aliphatic cycloalkane derivative, preparation method thereof, pharmaceutical composition and application thereof
CN116082259B (en) Carbamate or carbamoyl substituted 5-HT2B antagonists
CN111057069B (en) Cyclic compound, application and composition thereof
TW202246279A (en) Pyrazinethiobiphenyl compounds and application thereof
CN117279923A (en) Derivative of six-membered heteroaromatic urea ring and application thereof
WO2022143773A1 (en) Benzimidazole compound and application thereof

Legal Events

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