CN112480116B - PKB inhibitors - Google Patents

PKB inhibitors Download PDF

Info

Publication number
CN112480116B
CN112480116B CN201910858374.XA CN201910858374A CN112480116B CN 112480116 B CN112480116 B CN 112480116B CN 201910858374 A CN201910858374 A CN 201910858374A CN 112480116 B CN112480116 B CN 112480116B
Authority
CN
China
Prior art keywords
formula
methyl
compound
oxo
amino
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.)
Active
Application number
CN201910858374.XA
Other languages
Chinese (zh)
Other versions
CN112480116A (en
Inventor
马昌友
张林林
吴有智
吴舰
徐丹
朱春霞
田舟山
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.)
Nanjing Chia Tai Tianqing Pharmaceutical Co Ltd
Original Assignee
Nanjing Chia Tai Tianqing Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Chia Tai Tianqing Pharmaceutical Co Ltd filed Critical Nanjing Chia Tai Tianqing Pharmaceutical Co Ltd
Priority to CN201910858374.XA priority Critical patent/CN112480116B/en
Publication of CN112480116A publication Critical patent/CN112480116A/en
Application granted granted Critical
Publication of CN112480116B publication Critical patent/CN112480116B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

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

Abstract

The invention provides PKB inhibitors, in particular to a compound shown in a formula I or pharmaceutically acceptable salt thereof, and a preparation method thereof.

Description

PKB inhibitors
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a PKB inhibitor, a preparation method and medical application thereof.
Background
The PI3K/AKT/mTOR pathway composed of phosphatidylinositol 3-kinase (PI 3K) and its downstream proteins AKT (also known as protein kinase B, PKB) and mammalian target of rapamycin (mTOR) serves as a very important intracellular signal transduction pathway, playing an extremely important biological role in the processes of growth, survival, proliferation, apoptosis, angiogenesis, autophagy, etc. of cells. Abnormal activation of this pathway can cause a range of diseases including cancer, neuropathy, autoimmune diseases, and diseases of the blood lymphatic system.
AKT has three subtypes: AKT1, AKT2 and AKT3. As typical protein kinases, each subtype consists of an amino-terminal PH domain (Pleckstrin homology domain), a mid-ATP-binding kinase domain, and a carboxy-terminal regulatory domain. About 80% of the amino acid sequences of the 3 subtypes are homologous, varying only in the PH domain and kinase domain junction regions.
Currently, targeted drugs against the PI3K/AKT/mTOR signaling pathway are mainly PI3K inhibitors and mTOR inhibitors, whereas AKT is at the core of this signaling pathway. Inhibiting AKT activity can avoid serious side effects caused by inhibiting upstream PI3K, and can also avoid negative feedback mechanism caused by inhibiting downstream mTOR from affecting drug efficacy. Thus, the search for potent and selective AKT inhibitors is an important direction in the development of current tumor-targeted drugs.
Disclosure of Invention
In one aspect, the present invention provides a compound of formula I or a pharmaceutically acceptable salt thereof,
wherein:
R 1 and R is 2 Each independently selected from H or C1-C6 alkyl;
m is selected from 0 or 1;
a is optionally substituted with 1-5R 3 Substituted phenyl, wherein each R 3 Each independently selected from halogen or C1-C6 alkyl, wherein C1-C6 alkyl may be optionally substituted with halogen;
selected from->
G is selected from 5-6 membered heteroaryl or phenyl, wherein the heteroaryl or phenyl is optionally substituted with R 4 Substituted, and R 4 Selected from halogen or C1-C6 alkyl;
l isA group, wherein:
Y 1 selected from CY 11 Y 12 Or NY 13 ,Y 2 Selected from O, CY 21 Y 22 Or a bond, and is satisfied when Y 2 When selected from O, Y 1 Selected from CY 11 Y 12 And, in addition, the method comprises,
Y 11 and Y 21 Each independently selected from H, OH, halogen or C1-C6 alkyl, wherein C1-C6 alkyl may be optionally substituted with halogen, OH, C1-C3 alkoxy or CN,
Y 12 And Y 22 Each independently selected from H or C1-C6 alkyl;
Y 13 selected from H or C1-C6 alkyl, wherein C1-C6 alkyl may be optionally substituted with halogen, OH, C1-C3 alkoxy or CN.
In some embodiments, R 1 And R is 2 Each independently selected from H, methyl, ethyl or isopropyl; preferably, R 1 Selected from H, R 2 Selected from H, methyl, ethyl or isopropyl; more preferably, R 1 Selected from H, R 2 Selected from H or isopropyl; most preferably, R 1 Selected from H, R 2 Selected from H.
In some embodiments, m is selected from 1.
In some embodiments, A is a polypeptide comprising 1 or 2R 3 A substituted phenyl group.
In some embodiments, a is selected from the group consisting of:
in some embodiments, R 3 Independently selected from F, cl, br, I, CH 3 、C 2 H 5 Or CF (CF) 3 The method comprises the steps of carrying out a first treatment on the surface of the In some typical embodiments, R 3 Selected from F, cl or CF 3 The method comprises the steps of carrying out a first treatment on the surface of the In some more typical embodiments, R 3 Selected from F or Cl; in some most typical embodiments, R 3 Selected from F.
In some embodiments, a is selected from the group consisting of:
in some typical embodiments, a is selected from the following groups:
in some typical embodiments, a is selected from the following groups:
in some more typical embodiments, A is selected from
In some embodiments of the present invention, in some embodiments, Selected from->
In some embodiments, G is selected from: five membered heteroaryl, pyridinyl or phenyl, wherein said five membered heteroaryl, pyridinyl or phenyl is optionally substituted with R 4 Substituted, and R 4 Selected from halogen or C1-C6 alkyl.
In some embodiments, R 4 Selected from CH 3 F or Cl; in some typical embodiments, R 4 Selected from CH 3 Or F.
In some embodiments, G is selected from the group consisting of optionally R 4 Substituted with the following groups:
wherein,represents the position where G is linked to Q by a chemical bond, < >>Represents the position where G is attached to L by a chemical bond;
R 4 is as defined above.
In some typical embodiments, G is selected from the group consisting of optionally R 4 Substituted with the following groups:
wherein R is 4 Is as defined above.
In some more typical embodiments, G is selected from the group consisting of optionally R 4 Substituted with the following groups:
wherein R is 4 Is as defined above.
In some more typical embodiments, G is selected from the following groups:
in some more typical embodiments, G is selected from the following groups:
in some more typical embodiments, G is selected from the following groups:
in some embodiments, Y 1 Selected from CY 11 Y 12 Wherein Y is 11 Selected from H, OH or C1-C6 alkyl, wherein C1-C6 alkyl may be optionally substituted with F; in some exemplary embodiments, Y 11 Selected from H,CH 3 Or CF (CF) 3 The method comprises the steps of carrying out a first treatment on the surface of the In some more typical embodiments, Y 11 Selected from CH 3 Or CF (CF) 3 The method comprises the steps of carrying out a first treatment on the surface of the In some most typical embodiments, Y 11 Selected from CH 3
In some embodiments, Y 12 Selected from H or CH 3
In some embodiments, Y 1 Selected from NY 13 Wherein Y is 13 Selected from H, CH 3 、C 2 H 5 、CH 2 CH 2 CH 3 、CHCH 3 CH 3 、CH 2 OH、C 2 H 4 OH、C 3 H 6 OH、CH 2 OCH 3 、CH 2 OC 2 H 5 、C 2 H 4 OCH 3 、CH 2 CN or C 2 H 4 A CN; preferably, Y 13 Selected from CH 3 、C 2 H 4 OH or CH 2 A CN; in some more typical embodiments, Y 13 Selected from CH 3
In some embodiments, Y 2 Selected from O, CY 21 Y 22 Or a bond, wherein Y 21 And Y 22 All are H; in some exemplary embodiments, Y 2 Selected from CH 2 Or a root key.
In some embodiments, L is selected from the group consisting of:
in some typical embodiments, G is selected from the following groups:
in some more typical embodiments, G is selected from the following groups:
in another aspect, the present invention provides a compound of formula II or a pharmaceutically acceptable salt thereof,
therein, A, G, L, R 1 And R is 2 Is defined as for the compounds of formula I.
In another aspect, the present invention provides a compound of formula III-1 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 31 Compounds of the formula I Y 11 ,R 32 Compounds of the formula I Y 12
In another aspect, the present invention provides a compound of formula III-2 or a pharmaceutically acceptable salt thereof,
Therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 33 Compounds of the formula I Y 13 Is defined in (a).
In another aspect, the present invention provides a compound of formula III-3 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 34 Compounds of the formula I Y 11 ,R 35 Compounds of the formula I Y 12
In another aspect, the present invention provides a compound of formula III-4 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 36 Compounds of the formula I Y 13 Is defined in (a).
In another aspect, the invention provides the following compounds, or pharmaceutically acceptable salts thereof:
in another aspect, the present invention provides a method for preventing and/or treating AKT protein kinase mediated diseases or conditions, comprising administering to a subject in need thereof a compound of formula I of the present invention or a pharmaceutically acceptable salt thereof.
In another aspect, the present invention also provides a compound of formula I of the present invention, or a pharmaceutically acceptable salt thereof, for use in the prevention and/or treatment of AKT protein kinase mediated diseases or conditions.
In yet another aspect, the present invention provides a process for preparing a compound of formula I, including, but not limited to, the following synthetic schemes:
wherein G, A is as defined above; y is Y 1 、Y 2 Derivatives as hereinbefore described or substituted with protecting groups; x is a leaving group such as halogen; p (P) 1 Is C1-C6 alkyl, preferably methyl, ethyl or isopropyl; p (P) 2 、P 3 Selected from H orTogether with the atoms to which they are attached form a borate; by "is meant that the compound may be in optically pure form or a mixture of two or more optically pure forms.
The compounds of formula 1-1 and the compounds of formula 1-2 are mixed in a base (e.g., cesium carbonate), a catalyst (e.g., pd (dppf) Cl) 2 ) And a solvent (e.g., 1, 4-dioxane) to produce a compound of formulas 1-3; preparing the compounds of the formulas 1-4 by hydrolysis reaction of the compounds of the formulas 1-3; reacting a compound of formula 1-4 with a compound of formula 1-5 in the presence of a base (e.g., N-diisopropylethylamine), a condensing agent (e.g., BOP), and a solvent (e.g., DMF) to produce a compound of formula 1-6; the compounds of formulas 1-6 produce compounds of formulas 1-7 under hydrazine hydrate conditions.
Detailed Description
Correlation definition
Unless specifically indicated, the following terms used in the specification and claims have the following meanings:
the term "compound" as used herein includes all stereoisomers and tautomers.
The compounds of the invention may be asymmetric, e.g., have one or more stereoisomers. Unless otherwise indicated, all stereoisomers include, for example, enantiomers and diastereomers. The compounds of the invention containing asymmetric carbon atoms can be isolated in optically pure or racemic form. Optically pure forms can be resolved from the racemic mixture or synthesized by using chiral starting materials or chiral reagents. Racemates, diastereomers, and enantiomers are all included within the scope of the present invention.
The compounds of the invention also include tautomeric forms. Tautomers originate from the exchange of one single bond with an adjacent double bond and accompany the migration of one proton.
The term "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The term "selected from" means that the event or circumstance described subsequently must occur.
Numerical ranges herein refer to individual integers within a given range. For example, "C1-C6" means that the group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms.
The term "substituted" means that any one or more hydrogen atoms on a particular atom or group is substituted with a substituent, provided that the valence of the particular atom or group is normal and the substituted compound is stable. The kind and number 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 1 to 5R, the group may optionally be substituted with up to 5R, and R in each case has an independent option. Furthermore, combinations of substituents and/or variants thereof are only permissible if such combinations result in stable compounds.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group, including straight or branched chain saturated hydrocarbon groups, having the indicated number of carbon atoms. The term "C1-C6 alkyl" includes C1 alkyl, C2 alkyl, C3 alkyl, C4 alkyl, C5 alkyl, C6 alkyl, examples including, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, n-pentyl, 2-pentyl, 3-pentyl, n-hexyl, 2-hexyl, 3-hexyl, and the like. It may be divalent, e.g. methylene, ethylene.
The term "alkoxy" refers to a group having the structure of an-O-alkyl, an alkyl being a saturated monovalent hydrocarbon radical comprising straight or branched chains. For example, "C1-C3 alkoxy" includes-OCH 3 、-OC 2 H 5 、-OC 2 H 4 CH 3 and-OCH (CH) 3 ) 2
The term "5-6 membered heteroaryl" refers to an aryl group comprising at least one 5-or 6-membered ring independently selected from nitrogen, oxygen and sulfur heteroatoms. Examples of heteroaryl groups include, but are not limited to, imidazolyl, pyrazolyl, thiazolyl, isothiazole, furanyl, pyrrolyl, thienyl, pyridyl, oxazolyl, pyrimidinyl, pyridazinyl, pyrazinyl, 1,2, 4-triazinyl, 1,3, 5-triazinyl, 1,2, 3-triazinyl.
The term "meta" refers to the number of backbone atoms that make up the ring. For example, "5-6 membered" means that the number of backbone atoms constituting the ring is 5 or 6.
All refer to the chemical bond connection, and have the same chemical meaning, if no special description exists,and->Only the difference in (a) is in the location or order of the connections.
The term "pharmaceutically acceptable salt" refers to salts that retain the biological effectiveness of the free acids and bases of the particular compounds without biological adverse effects. Such as acid (including organic and inorganic acids) addition salts or base addition salts (including organic and inorganic bases).
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.
M:mol/L
mM:mmol/L
nM:nmol/L
h: hours of
min: dividing into
1 H NMR: nuclear magnetic resonance hydrogen spectrum
LC/MS: liquid phase/mass spectrum combination
DIAD: diisopropyl azodicarboxylate
DCM: dichloromethane (dichloromethane)
BOP: benzotriazol-1-yloxy tris (dimethylamino) phosphonium hexafluorophosphate
Boc: boc-group
The preparation method comprises the following steps:
the preparation methods of the compounds of the present invention are described more specifically below, but these specific preparation methods do not set any limit to the scope of the present invention. In addition, the reaction conditions such as reactants, solvents, bases, amounts of compounds used, reaction temperatures, reaction times, and the like are not limited to the following examples.
The compounds of the present invention may also be conveniently prepared by optionally combining the various synthetic methods described in this specification or known in the art, such combination being readily carried out by one of ordinary skill in the art.
Preparation example 1
Synthesis of methyl 5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-3-carboxylate
2.0g of methyl 5-bromothiophene-3-carboxylate, 3.0g of pinacol biborate, 0.34g of [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride and 1.8g of potassium acetate are dissolved in 20mL of dioxane, the mixture is heated to reflux under the protection of nitrogen, the heating is stopped after 4 hours, and the reaction solution is decompressed, evaporated to dryness and then subjected to column chromatography to obtain 1.6g of white solid. 1 H NMR(400MHz,CDCl 3 )δ8.33(d,J=1.1Hz,1H),8.04(d,J=1.1Hz,1H),3.86(s,3H),1.35(s,12H).
Preparation example 2
Synthesis of 4-bromo-5-methylthiophene-2-carboxylic acid
2.2g of liquid bromine is dissolved in 5mL of acetic acid, 30mL of acetic acid solution containing 2.0g of 5-methylthiophene-2-carboxylic acid and 0.46g of ferric trichloride is added dropwise at room temperature, the mixture is stirred for 5 hours at room temperature, ice water is poured into the mixture, solid is separated out, suction filtration is carried out, and a filter cake is dried to obtain 2.66g of yellow solid after being washed by ice water. LC/MS (ESI+): 221.0 (M+H)
Preparation example 3
Synthesis of methyl 4-bromo-5-methylthiophene-2-carboxylate
2.65g of 4-bromo-5-methylthiophene-2-carboxylic acid was dissolved in 50mL of methanol at room temperature, after addition of 3mL of concentrated sulfuric acid, heated to 50℃and after 12h the reaction was cooled to room temperature, poured into ice water, pH was adjusted to 10-11 with 2M NaOH solution, extracted with DCM, the organic phase was taken out, dried over anhydrous sodium sulfate and evaporated to dryness under reduced pressure to give 2.7g of brown solid. LC/MS (ESI+): 234.9 (M+H). 1 H NMR(400MHz,DMSO-d 6 )δ7.71(d,J=1.4Hz,1H),3.82(s,3H),2.43(s,3H).
Preparation example 4
Synthesis of methyl 5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylate
2.0g of methyl 4-bromo-5-methylthiophene-2-carboxylate and 2.8g of pinacol biborate, 0.3g of [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride and 1.7g of potassium acetate are dissolved in 30ml of dioxane, the mixture is heated to reflux under the protection of nitrogen, the heating is stopped after 4 hours, and the reaction solution is decompressed, evaporated to dryness and then subjected to column chromatography to obtain 1.9g of white solid. LC/MS (ESI+): 283.1 (M+H). 1 H NMR(400MHz,CDCl 3 )δ7.95(s,1H),3.86(s,3H),2.72(s,3H),1.34(s,12H).
Preparation example 5
Synthesis of methyl 2-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-3-carboxylate
0.9g of methyl 5-bromo-2-methylthiophene-3-carboxylate, 1.46g of pinacol biborate, 0.28g of [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride and 0.74g of potassium acetate are dissolved in 20mL of dioxane, the mixture is heated to reflux under the protection of nitrogen, the heating is stopped after 4 hours, and the reaction solution is decompressed, evaporated to dryness and then subjected to column chromatography to obtain 0.65g of white solid. 1 H NMR(400MHz,DMSO-d 6 )δ7.72(s,1H),3.78(s,3H),2.71(s,3H),1.28(s,12H).
Preparation example 6
Synthesis of methyl 3-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylate
/>
Methyl 4-bromo-3-fluorothiophene-2-carboxylate (2.0 g), pinacol biborate (2.8 g), 1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.3 g) and potassium acetate (1.7 g) were dissolved in dioxane, heated to reflux under nitrogen protection, heating was stopped after 4h, and the reaction solution was evaporated to dryness under reduced pressure and then subjected to column chromatography to give a white solid (1.7 g). LC/MS (ESI+): 287.1 (M+H).
Preparation example 7
Synthesis of methyl 3-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylate
Methyl 4-bromo-3-methylthiophene-2-carboxylate (2.1 g), pinacol biborate (2.9 g), 1' -bis (diphenylphosphino) ferrocene palladium dichloride (0.3 g) and potassium acetate (1.7 g) were dissolved in dioxane, heated to reflux under nitrogen protection, after 4h, heating was stopped, the reaction solution was evaporated to dryness under reduced pressure, and column chromatography was performed to give a white solid (2.0 g). LC/MS (ESI+): 283.1 (M+H).
Preparation example 8
Synthesis of (R) -4-chloro-5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidin-7 (6H) -one
Reaction conditions: a) Ethyl crotonate, sodium methoxide in methanol (30% wt), methanol; b) Disodium hydrogen phosphate, deionized water, hydrochloric acid, lipase (candida rugosa), sodium hydroxide; c) Formamidine acetate, sodium methoxide, methanol; d) Phosphorus oxychloride, diisopropylethylamine, acetonitrile; e) Ammonia (25-28% wt);
a) 2-methylpropane-1, 3-tricarboxylic acid trimethyl ester
Sodium methoxide methanol solution (30% wt,50.32 g) was added to methanol (900 mL) at 20℃under nitrogen, followed by heating to 70℃and allowing dimethyl malonate (461.12 g) and ethyl crotonate (349.46 g) to react at 70℃for 3 hours by dropwise addition to the sodium methoxide methanol solution. After completion of the reaction, the solvent was distilled off under reduced pressure, ethyl acetate (1L) was added, pH was adjusted to 7-8 with 4M hydrochloric acid, then 500mL of water was added, and the organic phase was distilled off under reduced pressure to give 777.68g of a yellow liquid. 1 H NMR(400MHz,DMSO-d 6 )δ(ppm)3.67(s,3H),3.65(s,3H),3.59(s,3H),3.56(d,J=6.8Hz,1H),2.45-2.58(m,2H),2.23-2.29(m,1H),0.93(d,J=6.8Hz,3H)。
b) (R) -2-methylpropane-1, 3-tricarboxylic acid trimethyl ester
Disodium hydrogen phosphate (4.5 g) was dissolved in 1.5L deionized water at 25deg.C, pH=7.05 was adjusted with 2N hydrochloric acid, trimethyl 2-methylpropane-1, 3-tricarboxylic acid (150.46 g) and lipase (Candida rugosa, 40g added in 6 days) were added, pH was adjusted to 7.0-7.6 with 2N sodium hydroxide solution, and the mixture was reacted at 35deg.C for 6 days, chiral detection ee%>98%, chiral detection conditions (chiral column AS, n-hexane: ethanol=9:1, 0.2% dimethylethylamine, volume ratio). The reaction solution was cooled to 10℃and pH was adjusted to 3-4 with 3M hydrochloric acid, 500mL of ethyl acetate was added, suction filtration was performed, the cake was washed with ethyl acetate (600 mL), the solution was separated, saturated aqueous sodium bicarbonate solution (100 mL) was added for washing, the solution was separated, and the organic phase was concentrated to give 26.89g of a pale yellow liquid. 1 H NMR(400MHz,CDCl 3 )δ(ppm)3.74(s,6H),3.68(s,3H),3.46(d,J=7.2Hz,1H),2.71-2.79(m,1H),2.54(dd,J=15.6、4.8Hz,1H),2.32(dd,J=16.0、8.4Hz,1H),1.06(d,J=6.8Hz,3H)。
c) (R) -3- (4, 6-dihydroxypyrimidin-5-yl) butanoic acid methyl ester
Formamidine acetate (11.33 g) was dissolved in methanol (200 mL) at 20℃under nitrogen protection, cooled to 0℃and then sodium methoxide methanol solution (30%, 55.62 g) was added dropwise thereto for reaction at 0℃for 60 minutes, and a solution of trimethyl (R) -2-methylpropane-1, 3-tricarboxylic acid (24.07 g) in methanol (60 mL) was added dropwise thereto and the temperature was raised naturally to 20℃for reaction for 10 hours. After the reaction was completed, the reaction solution was cooled to 0 ℃, 3N hydrochloric acid was added to adjust pH to 5-6, the solvent was distilled off under reduced pressure, then cooled to 0 ℃, 3N hydrochloric acid was added to adjust ph=3, solids were precipitated, the solids were collected by suction filtration, the filter cake was washed with ice water (100 mL), and the filter cake was dried in vacuo to give 18.79g of a white solid, which was directly used in the next step.
d) (R) -3- (4, 6-dichloropyrimidin-5-yl) butanoic acid methyl ester
Methyl (R) -3- (4, 6-dihydroxypyrimidin-5-yl) butyrate (14.63 g) was dispersed in acetonitrile (70 mL) under nitrogen protection at 22deg.C, phosphorus oxychloride (26.42 g) and diisopropylethylamine (12.51 g) were added dropwise, the system was exothermic and the temperature was then raised to 60deg.C, the solids were gradually dissolved, and the reaction was continued for 18 hours. After completion of the reaction, the reaction solution was cooled to 0℃and 100mL of ethyl acetate was added, the pH was adjusted to 7-8 with saturated sodium bicarbonate solution, extraction was performed with ethyl acetate (50 mL. Times.3), and the organic phase was distilled off under reduced pressure to obtain 13.89g of a yellow solid which was used directly in the next step.
e) (R) -4-chloro-5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidin-7 (6H) -one
Methyl (R) -3- (4, 6-dichloropyrimidin-5-yl) butyrate (13.89 g) and ammonia (25-28% wt,70 mL) were added to a 100mL autoclave at 20℃and the temperature was raised to 50℃for 18 hours. After the reaction was completed, the reaction solution was cooled to 0 ℃, filtered by suction, and the cake was slurried with 30mL (petroleum ether: ethyl acetate=10:1, volume ratio) to obtain 7.32g of pale yellow solid. LC-MS (ESI) M/z:198 (M+H). 1 H NMR(300MHz,CDCl 3 )δ(ppm)1.30(d,J=7.2Hz,3H),2.65-2.69(m,1H),2.86-2.92(m,1H),3.47-3.54(m,1H),8.64(s,1H),10.10(s,1H)。
Preparation example 9
Synthesis of 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one
Reaction conditions: a) Ethyl bromoacetate, sodium hydride, tetrabutylammonium iodide, tetrahydrofuran; b) 2, 4-dimethoxy benzyl amine, triethylamine, isopropanol; c) Methyl iodide, sodium hydride, N dimethylformamide; d) Trifluoroacetic acid;
a) 2- ((4, 6-dichloropyrimidin-5-yl) amino) acetic acid ethyl ester
To a solution of 4, 6-dichloro-5-aminopyrimidine (10.0 g) in tetrahydrofuran (100 mL) was added sodium hydride (2.93 g) at 0deg.C. The reaction mixture was stirred for 2min. After the temperature was raised to 20℃naturally, ethyl bromoacetate (12.22 g) was added thereto, and tetrabutylammonium iodide (27.03 g) was further added thereto. The reaction mixture was stirred at 20 ℃ for 16 hours. After completion of the reaction, the reaction mixture was poured into water (100 mL), stirred for 30min, and extracted with ethyl acetate (100 mL). The organic phase was washed with saturated sodium chloride solution (100 ml x 3), dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure. The product was purified by silica gel chromatography (petroleum ether: ethyl acetate=20:1, volume ratio) to give 6.5g of colorless oil, which was used directly in the next step.
b) 4-chloro-8- (2, 4-dimethoxybenzyl) -5, 8-dihydro-pteridin-7 (6H) -one
To an isopropanol solution (150 mL) dissolved with ethyl 2- ((4, 6-dichloropyrimidin-5-yl) amino) acetate (5 g) was added 2, 4-dimethoxybenzyl amine (3.67 g) followed by triethylamine (4.45 g) at 20 ℃. The reaction mixture was stirred at 80℃for 18 hours. After the reaction was completed, the reaction solution was filtered, and the filter cake was washed with ethanol and dried under reduced pressure to give 5.0g of an off-white solid, which was directly used in the next step.
c) 4-chloro-8- (2, 4-dimethoxybenzyl) -5-methyl-5, 8-dihydro-pteridin-7 (6H) -one
To a solution of 4-chloro-8- (2, 4-dimethoxybenzyl) -5, 6-dihydropteridin-7 (8H) -one (3.3 g) in N, N-dimethylformamide (30 mL) was added methyl iodide (1.68 g) at 0℃and the reaction mixture was stirred for 20min. Maintain 0℃and add sodium hydride (0.47 g). The reaction mixture was stirred for a further 3 hours at 0 ℃. After completion of the reaction, the reaction mixture was poured into water (100 mL), followed by extraction with ethyl acetate (100 mL). The organic phase was washed with saturated sodium chloride solution (100 ml x 3), dried over anhydrous sodium sulfate and the solvent was distilled off under reduced pressure. The product was purified by silica gel chromatography (petroleum ether: ethyl acetate=5:1, volume ratio) to give 1.5g of product as a white solid, which was used directly in the next step.
d) 4-chloro-5-methyl-5, 8-dihydro-pteridin-7 (6H) -one
To 4-chloro-8- (2, 4-dimethoxybenzyl) -5-methyl-5, 6-dihydropteridin-7 (8H) -one (5 g) was added trifluoroacetic acid (20 mL) at 20 ℃. The reaction mixture was stirred at 60 ℃ for 16 hours. After completion of the reaction, the solvent was distilled off under reduced pressure to give a near-violet solid (1.0 g). LC-MS (ESI) M/z 199.1 (M+H). 1 H NMR(300MHz,DMSO-d 6 )δ(ppm)11.60(s,1H),8.35(s,1H),3.77(s,2H),2.85(s,3H).
Preparation example 10
Synthesis of 2- (2-amino-3- (3, 4-difluorophenyl) propyl) isoindoline-1, 3-dione
The title compound was prepared starting from tert-butyl (1- (3, 4-difluorophenyl) -3-hydroxypropan-2-yl) carbamate by the preparation method described in WO2010093885A1, preparation example 1.
PREPARATION EXAMPLE 11
Synthesis of 5-chloro-4-methyl-1, 4-dihydro-2H-pyrimido [4,5-d ] [1,3] oxazin-2-one
a) 1- (4-amino-6-chloropyrimidin-5-yl) ethanones
1- (4, 6-dichloropyrimidin-5-yl) ethanone (2.5 g) was dissolved in tetrahydrofuran (15 mL) at 20 ℃, then ammonia water (9 g) was added, the reaction solution was stirred at 20 ℃ for 5 hours, then concentrated and diluted with a small amount of water, suction filtration was performed to obtain a white solid, and after vacuum drying, 2g of white solid was obtained, which was directly used in the next step.
b) 1- (4-amino-6-chloropyrimidin-5-yl) ethan-1-ol 1- (4-amino-6-chloropyrimidin-5-yl) ethanone (1.5 g) was dissolved in methanol (15 mL) and cooled to-10 ℃ and sodium borohydride (1 g) was added in portions, after which the reaction mixture was slowly warmed to 20 ℃ and stirred for 3 hours. After the reaction was completed, the reaction solution was quenched with a saturated aqueous ammonium chloride solution. The reaction was then concentrated and slurried with ethyl acetate (20 ml x 2). Concentrating the mother liquor to obtain an oily crude product. The crude product was isolated by column chromatography to give 400mg of product as a white oil. LC-MS (ESI+): 174.1 (M+H).
c) 5-chloro-4-methyl-1, 4-dihydro-2H-pyrimidine [4,5-d ] [1,3] oxazin-2-one
1- (4-amino-6-chloropyrimidin-5-yl) ethan-1-ol (300 mg), N-diisopropylethylamine (282 mg) was dissolved in tetrahydrofuran (3 mL) at 20℃and then cooled to-5℃and bis (trichloromethyl) carbonate (300 mg) was slowly added and stirred at-5℃for 0.5 hours. Then slowly heating to 18 ℃ and stirring for 1.5 hours. After completion of the reaction, the reaction was quenched with aqueous sodium bicarbonate and extracted with ethyl acetate (10 ml x 3), the organic phases were combined and dried over anhydrous sodium sulfate, filtered and concentrated to give crude oil. The crude product was purified by column chromatography to give 108mg of a white solid). LC/MS (esi+): 200.1 (M+H).
Preparation example 12
Synthesis of 2- (4-chloro-7-oxo-7, 8-dihydropteridin-5 (6H) -yl) acetonitrile
a) 2- (4-chloro-8- (3, 4-dimethoxybenzyl) -7-oxo-7, 8-dihydro-pteridin-5 (6H) -yl) acetonitrile
Sodium hydride (60%, 0.48 g) was added to an N, N-dimethylformamide solution (20 mL) containing 4-chloro-8- (2, 4-dimethoxybenzyl) -5, 8-dihydropteridin-7 (6H) -one (2 g) at 0℃and bromoacetonitrile (1 g) was slowly dropped after releasing no bubbles, and the reaction mixture was stirred at 0℃for 3 hours. After completion of the reaction, the reaction mixture was poured into water (100 mL), followed by extraction with ethyl acetate (100 mL). The organic phase was washed with saturated sodium chloride solution (100 ml x 3), dried over anhydrous sodium sulfate and the solvent was distilled off under reduced pressure. The product was purified by silica gel column chromatography to give 0.6g of a yellow solid which was used directly in the next step. LC/MS (ESI+): 342.1 (M+H)
b) 2- (4-chloro-7-oxo-7, 8-dihydro-pteridin-5 (6H) -yl) acetonitrile
2- (4-chloro-8- (3, 4-dimethoxybenzyl) -7-oxo-7, 8-dihydropteridin-5 (6H) -yl) acetonitrile (0.6 g) was dissolved in trifluoroacetic acid (15 ml) and reacted at 55℃for 4 hours. After the reaction, the ph=8 was adjusted with saturated sodium bicarbonate solution, extracted with ethyl acetate (15 ml×4), the organic layers were combined, dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation under reduced pressure, and the white solid (0.3 g) was purified by column chromatography. LC/MS (ESI+): 224.1 (M+H).
Preparation example 13
Synthesis of 4-chloro-5- (2-methoxyethyl) -5, 8-dihydro-pteridin-7 (6H) -one
a) 4-chloro-8- (2, 4-dimethoxybenzyl) -5- (2-methoxyethyl) -5, 8-dihydro-pteridin-7 (6H) -one
To a solution of 4-chloro-8- (2, 4-dimethoxybenzyl) -5, 8-dihydropteridin-7 (6H) -one (1 g) in N, N-dimethylformamide (10 mL) at 0deg.C was added sodium hydride (60%, 0.24 g), and after releasing no bubbles, 1-iodo-2-methoxyethane (0.8 g) was slowly added dropwise, and the reaction mixture was stirred at 0deg.C for 3 hours. After completion of the reaction, the reaction mixture was poured into water (100 mL), followed by extraction with ethyl acetate (100 mL). The organic phase was washed with saturated sodium chloride solution (100 ml x 3), dried over anhydrous sodium sulfate and the solvent was distilled off under reduced pressure. The product was purified by silica gel column chromatography to give 1.2g of yellow solid which was used directly in the next step. LC/MS (ESI+): 361.2 (M+H).
b) 4-chloro-5- (2-methoxyethyl) -5, 8-dihydro-pteridin-7 (6H) -one
4-chloro-8- (2, 4-dimethoxybenzyl) -5- (2-methoxyethyl) -5, 8-dihydropteridin-7 (6H) -one (1 g) was dissolved in trifluoroacetic acid (15 ml) and reacted at 55℃for 4 hours. After the reaction, the ph=8 was adjusted with saturated sodium bicarbonate solution, extracted with ethyl acetate (15 ml×4), the organic layers were combined, dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation under reduced pressure, and the white solid (0.5 g) was purified by column chromatography.
LC/MS(ESI+):243.2(M+H)。
Example 1 (S) -2- (4-chlorophenyl) -3- (isopropylamino) -N- (3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) propanamide
a) Tert-butyl (3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) carbamate
300mg of 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one, 540mg of (3- ((tert-butoxycarbonyl) amino) phenyl) boric acid, 105mg of [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride and 980mg of cesium carbonate are dissolved in a mixed solution of 20mL of dioxane and 2mL of water, heating is carried out under the protection of nitrogen until reflux is carried out, heating is stopped after 5 hours, and the reaction solution is dried under reduced pressure and then subjected to column chromatography to obtain 210mg of pale yellow solid;
b) 4- (3-aminophenyl) -5-methyl-5, 8-dihydro-pteridin-7 (6H) -one
Dissolving tert-butyl (3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridine-4-yl) phenyl) carbamate prepared in the previous step in 10mL of trifluoroacetic acid, refluxing for 1h, evaporating to dryness under reduced pressure, neutralizing with 10mL of saturated sodium bicarbonate solution, and evaporating to dryness to obtain 110mg of yellow solid by column chromatography;
c) Tert-butyl (S) - (2- (4-chlorophenyl) -3- ((3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) amino) -3-oxopropyl) (isopropyl) carbamate
110mg of 4- (3-aminophenyl) -5-methyl-5, 8-dihydropteridine-7 (6H) -one trifluoroacetate, 150mg of (S) -3- ((tert-butoxycarbonyl) (isopropyl) amino) -2- (4-chlorophenyl) propionic acid and 200mg of N, N-diisopropylethylamine were dissolved in 10mL of dichloromethane, 400mg of benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate were added at 25℃and reacted for 1 hour under stirring, and then evaporated to dryness under reduced pressure, followed by column chromatography to give 197mg of pale yellow solid;
d) (S) -2- (4-chlorophenyl) -3- (isopropylamino) -N- (3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) propanamide
Tert-butyl (S) - (2- (4-chlorophenyl) -3- ((3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) amino) -3-oxopropyl) (isopropyl) carbamate (197 mg) was dissolved in trifluoroacetic acid (10 ml), and after 1h of reflux, evaporated to dryness under reduced pressure to give a yellow solid (50 mg).
LC/MS(ESI+):479.2(M+H).
1 H NMR(400MHz,CDCl 3 )δ10.90(s,1H),8.59(d,J=1.9Hz,1H),8.16(s,1H),7.72(s,1H),7.56(s,1H),7.46–7.31(m,4H),4.25(s,1H),3.93(d,J=17.2Hz,1H),3.83(d,J=16.9Hz,1H),3.72(qd,J=7.1,1.1Hz,1H),3.49(d,J=1.6Hz,2H),3.16–2.93(m,2H),2.47(s,3H),1.23(ddd,J=16.4,8.6,6.1Hz,7H).
Example 2 (S) -2- (4-chlorophenyl) -3- (isopropylamino) -N- (4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) propanamide
a) Tert-butyl (4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) carbamate
By way of example 1, step a, was synthesized starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one, (4- ((tert-butoxycarbonyl) amino) phenyl) boronic acid.
b) 4- (4-aminophenyl) -5-methyl-5, 8-dihydro-pteridin-7 (6H) -one
Reference is made to the procedure of step b) of example 1 from tert-butyl (4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) carbamate.
c) Tert-butyl (S) - (2- (4-chlorophenyl) -3- ((4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) amino) -3-oxopropyl) (isopropyl) carbamate
Reference is made to example 1, step c), which is synthesized from 4- (4-aminophenyl) -5-methyl-5, 8-dihydropteridin-7 (6H) -one trifluoroacetate and (S) -3- ((tert-butoxycarbonyl) (isopropyl) amino) -2- (4-chlorophenyl) propanoic acid.
d) (S) -2- (4-chlorophenyl) -3- (isopropylamino) -N- (4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) propanamide
Reference is made to example 1, step d), which is synthesized from tert-butyl (S) - (2- (4-chlorophenyl) -3- ((4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) phenyl) amino) -3-oxopropyl) (isopropyl) carbamate.
LC/MS(ESI+):479.2(M+H).
1 H NMR(400MHz,DMSO)δ10.60(s,1H),8.51(s,1H),8.32(s,1H),8.06(d,J=8.5Hz,2H),7.72(d,J=8.5Hz,2H),7.42(s,4H),3.94(s,1H),3.81(s,2H),3.33-3.28(m,1H),2.87-2.83(m,2H),2.42(s,3H),1.03(s,3H),1.02(s,3H).
Example 3 (S) -2- (4-chlorophenyl) -3- (isopropylamino) -N- (4- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) phenyl) propanamide
a) (R) - (4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) phenyl) carbamic acid tert-butyl ester
By the method of step a of example 1, starting from (R) -4-chloro-5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidin-7 (6H) -one and (4- ((tert-butoxycarbonyl) amino) phenyl) boronic acid.
b) (R) -4- (4-aminophenyl) -5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidin-7 (6H) -one
Reference is made to the procedure of step b, example 1, from the product of step a.
c) Tert-butyl ((S) -2- (4-chlorophenyl) -3- ((4- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] ] pyrimidin-4-yl) phenyl) amino) -3-oxopropyl) (isopropyl) carbamate
Synthesized by the method of step c of example 1.
d) (S) -2- (4-chlorophenyl) -3- (isopropylamino) -N- (4- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) phenyl) propanamide
Reference is made to the procedure of step d, example 1, from the product of step c.
LC/MS(ESI+):478.2(M+H).
1 H NMR(400MHz,DMSO)δ11.17(s,1H),10.88(s,1H),9.18(s,1H),8.75(s,1H),8.58(s,1H),7.81(d,J=8.6Hz,2H),7.60–7.43(m,6H),4.41(d,J=4.6Hz,1H),3.71(m,1H),3.39(s,1H),3.38–3.24(m,2H),3.14–3.02(m,1H),2.95(dd,J=16.3,6.0Hz,1H),2.32(d,J=15.7Hz,1H),1.27(s,3H),1.26(s,3H),0.94(d,J=7.0Hz,3H).
Example 4 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-3-carboxamide
a) 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxylic acid methyl ester
Methyl 5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-3-carboxylate (485 mg), 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one (180 mg), bis triphenylphosphine palladium dichloride (63 mg) and cesium carbonate (586 mg) were dissolved in a mixed solution of dioxane (20 mL) and water (2 mL), heated to reflux under nitrogen protection, heating was stopped after 8H, and the reaction solution was evaporated to dryness under reduced pressure and then subjected to column chromatography to give a pale yellow solid (240 mg).
LC/MS(ESI+):305.1(M+H).
1 H NMR(400MHz,CDCl 3 )δ9.03(s,1H),8.64(s,1H),8.60(d,J=1.3Hz,1H),8.29(d,J=1.4Hz,1H),3.92(s,3H),3.82(m,2H),2.77(s,3H).
b) 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxylic acid
150mg of methyl 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxylate are dissolved in 10mL of methanol, 10mL of 2M lithium hydroxide solution is added, stirring is carried out at room temperature overnight, methanol is removed by rotary evaporation, 2M hydrochloric acid is added to adjust the pH to 4-5, after extraction with ethyl acetate, the organic phase is taken, anhydrous sodium sulfate is dried and column chromatography is carried out to obtain 55mg of light yellow solid.
c) (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxamide
50mg of 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxylic acid, 70mg of (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione, 113mg of benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate, 52mg of N, N-diisopropylethylamine were dissolved in 5mL of N, N-dimethylformamide and stirred overnight at room temperature. Pouring the reaction solution into water, precipitating solid, filtering, drying, and purifying by column chromatography to obtain 52mg pale yellow solid.
d) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-3-carboxamide
50mg of the product from step c was dissolved in 5mL of methanol, 44mg of hydrazine hydrate was added, the mixture was stirred at room temperature overnight, the reaction mixture was evaporated under reduced pressure, and 14mg of an off-white solid was obtained by column chromatography.
LC/MS(ESI+):441.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.57(s,1H),8.44(d,J=1.5Hz,1H),8.33(d,J=1.3Hz,1H),8.30(s,1H),7.36–7.23(m,2H),7.10(d,J=8.7Hz,2H),4.19(s,2H),3.77(m,2H),2.97–2.92(m,1H),2.79(m,2H),2.61(s,3H).
Example 5N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -4- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) thiophene-2-carboxamide
a) (R) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) thiophene-2-carboxylic acid methyl ester
Reference to the procedure of example 4, step a, was synthesized starting from methyl 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylate and (R) -4-chloro-5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidin-7 (6H) -one.
b) (R) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) thiophene-2-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -4- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) thiophene-2-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 570.2 (M+H)
d) Synthesis of N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -4- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) thiophene-2-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):440.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.72(s,1H),8.43(d,J=8.8Hz,1H),8.19(d,J=1.3Hz,1H),8.15(d,J=1.3Hz,1H),7.33–7.25(m,1H),7.12–7.03(m,2H),6.99(td,J=8.3,1.8Hz,1H),4.06(d,J=5.5Hz,1H),3.51(d,J=5.8Hz,1H),2.93(m,3H),2.79(m,1H),2.68(d,J=6.1Hz,2H),2.39(d,J=1.7Hz,1H),2.35(d,J=1.6Hz,1H),1.13(d,J=7.1Hz,3H).
Example 6 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) furan-2-carboxamide
Prepared according to the method of example 4 starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and methyl 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) furan-2-carboxylate.
LC/MS(ESI+):425.2(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.66(s,2H),8.50(d,J=8.6Hz,1H),7.90(s,1H),7.36(td,J=7.9,6.1Hz,1H),7.21–7.13(m,2H),7.05(td,J=8.9,8.4,2.1Hz,1H),4.18(d,J=6.6Hz,2H),3.79(s,3H),3.01(dd,J=13.7,5.3Hz,1H),2.95–2.70(m,3H),2.65(s,3H).
Example 7 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
a) Synthesis of methyl 4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylate
Prepared from 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylic acid methyl ester and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one as starting materials according to the method of example 4 step a.
b) Synthesis of 4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione.
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):441.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.76(m,1H),8.66(m,1H),8.60(m,1H),8.55(m,1H),7.30(m,1H),7.10(m,2H),7.05(m,1H),4.33(m,2H),3.77(s,2H),2.98–2.84(m,4H),2.68(s,3H).
Example 8 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5-methyl-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
a) Synthesis of methyl 5-methyl-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylate
Prepared by reacting 5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylic acid methyl ester synthesized in preparation example 4 with 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one according to the method of example 4 step a.
b) Synthesis of 5-methyl-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -5-methyl-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione.
d) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5-methyl-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):455.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.43(s,1H),8.30(d,J=8.4Hz,1H),7.86(s,1H),7.30(td,J=8.0,6.2Hz,1H),7.13–7.03(m,2H),7.00(td,J=8.6,2.5Hz,1H),4.13(t,J=6.9Hz,1H),3.88(d,J=1.4Hz,2H),2.93(dd,J=13.6,5.4Hz,1H),2.86–2.66(m,3H),2.40(s,3H),2.36(s,3H),1.32–1.21(m,2H).
Example 9 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-methyl-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Specific preparation method referring to example 4, starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and 3-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylic acid methyl ester.
LC/MS(ESI+):455.1(M+H)..
1 H NMR(400MHz,Chloroform-d)δ8.52(s,1H),7.57(s,1H),7.11–6.90(m,3H),6.52(d,J=8.0Hz,1H),4.34(d,J=7.0Hz,1H),3.87(s,2H),3.03(dd,J=13.8,6.5Hz,1H),2.97–2.79(m,3H),2.51(s,3H),2.38(s,3H).
Example 10 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-fluoro-4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Specific preparation method referring to example 4, starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and 3-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylic acid methyl ester.
LC/MS(ESI+):459.2(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.43(s,1H),8.36(s,1H),8.13(d,J=4.0Hz,1H),7.88(s,2H),7.32(q,J=7.5Hz,1H),7.15–6.94(m,3H),4.19(s,1H),3.86(s,2H),3.00–2.65(m,5H),2.51(s,3H).
Example 11 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
a) Synthesis of methyl 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylate
Prepared by the method of step a of example 4 from methyl 5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylate and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one.
b) Synthesis of 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 571.2 (M+H).
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):441.2(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.57(s,1H),8.49(s,1H),8.36(s,1H),8.13(d,J=3.9Hz,1H),7.83(d,J=4.1Hz,1H),7.31(q,J=7.4Hz,1H),7.10(d,J=8.1Hz,2H),7.05–6.95(m,1H),4.13(s,1H),3.77(s,2H),3.00–2.92(m,1H),2.79(dd,J=35.3,7.7Hz,3H),2.62(s,3H).
Example 12 Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-3-carboxamide
Specific preparation method referring to example 4, starting from methyl 4-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-3-carboxylate and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one.
LC/MS(ESI+):455.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.40(d,J=16.0Hz,3H),8.03(s,1H),7.33(td,J=8.0,6.2Hz,1H),7.20–6.93(m,3H),4.26(d,J=9.4Hz,1H),3.87(s,2H),3.01–2.71(m,4H),2.42(s,3H),2.12(s,3H).
Example 13 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
a) Synthesis of methyl 3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylate
Prepared from 3-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylic acid methyl ester and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one as starting materials according to the method of example 4 step a.
b) Synthesis of 3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 585.2 (M+H).
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):455.1(M+H).
1 H NMR(400MHz,CDCl 3 )δ8.43(s,1H),7.82(s,1H),7.42–7.29(m,1H),7.12–6.93(m,3H),6.66(d,J=8.1Hz,1H),4.40(q,J=6.7Hz,1H),3.89–3.57(m,2H),3.14–2.81(m,4H),2.63(s,3H),2.43(s,3H).
Example 14 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
a) Synthesis of methyl 4-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylate
Prepared from 4-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-2-carboxylic acid methyl ester and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one as starting materials according to the method of example 4 step a.
b) Synthesis of 4-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -4-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-2-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione.
d) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-2-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):455.1(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.62(s,1H),8.47(s,1H),8.39(s,1H),7.65(s,1H),7.35-7.29(m,1H),7.11-7.09(m,2H),7.01(t,J=8.2Hz,1H),4.25-4.11(m,1H),3.86(s,2H),3.01-2.90(m,2H),2.90-2.74(m,4H),2.47(s,3H),2.28(s,3H).
Example 15 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -2-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-3-carboxamide
a) Synthesis of methyl 2-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxylate
Prepared by the method of step a of example 4 from methyl 2-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) thiophene-3-carboxylate and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one.
b) 2-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxylic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -2-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) thiophene-3-carboxamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 585.2 (M+H).
d) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -2-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) thiophene-3-carboxamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):445.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.55(s,1H),8.21(s,1H),8.09(t,J=9.8Hz,1H),7.31(q,J=7.4Hz,1H),7.13–7.04(m,2H),7.01(t,J=8.5Hz,1H),6.26(s,2H),4.10(d,J=7.8Hz,1H),3.74(s,2H),3.17(s,1H),2.96(dd,J=13.8,5.1Hz,1H),2.84–2.66(m,3H),2.56(s,3H).
Example 16 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -1-methyl-3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) -1H-pyrazole-5-carboxamide
Specific preparation referring to example 4, starting from methyl 1-methyl-3- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazole-5-carboxylate and 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one.
LC/MS(ESI+):439.2(M+H)
Example 17 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
a) Synthesis of methyl 4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate
Prepared by the method of example 4, step a, from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one with (4- (methoxycarbonyl) phenyl) boronic acid.
b) Synthesis of 4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione.
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):435.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.71(d,J=8.3Hz,1H),8.53(s,1H),8.39(s,1H),8.01(d,J=8.2Hz,2H),7.91(d,J=8.2Hz,2H),7.32(dd,J=14.4,7.6Hz,1H),7.13(d,J=7.7Hz,2H),7.01(t,J=8.4Hz,1H),4.33(d,J=6.7Hz,1H),3.88(s,2H),3.00–2.84(m,4H),2.40(s,3H).
Example 18 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
a) Synthesis of methyl 3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate
Prepared by the method of step a of example 4 starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (3- (methoxycarbonyl) phenyl) boronic acid.
b) Synthesis of 3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoic acid
Reference is made to the procedure of step a, example 4, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 565.2 (M+H).
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):435.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.55(d,J=12.3Hz,2H),8.34(d,J=19.9Hz,2H),8.10(d,J=7.8Hz,1H),7.88(d,J=7.8Hz,1H),7.56(t,J=7.7Hz,1H),7.36–7.26(m,1H),7.14–7.06(m,2H),7.00(td,J=8.7,2.6Hz,1H),4.27(s,1H),3.94–3.79(m,2H),3.00–2.85(m,1H),2.84(s,2H),2.38(s,3H).
Example 19 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
a) Synthesis of methyl 3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate
Prepared by the method of step a of example 4 starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (3- (methoxycarbonyl) -5-methylphenyl) boronic acid.
b) Synthesis of 3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 579.2 (M+H).
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-methyl-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
Reference is made to example 4, procedure d, from the product of step c.
1 H NMR(400MHz,DMSO-d 6 )δ8.57(d,J=8.1Hz,1H),8.51(s,1H),8.37(s,1H),8.11(d,J=1.7Hz,1H),7.87(d,J=1.7Hz,1H),7.71(d,J=1.9Hz,1H),7.32(td,J=8.1,6.3Hz,1H),7.10(dt,J=8.7,2.3Hz,2H),7.07–6.96(m,1H),4.32(d,J=7.8Hz,2H),3.98–3.79(m,2H),3.05–2.77(m,3H),2.42(s,3H),2.38(s,3H).LC/MS(ESI+):449.1(M+H).
Example 20 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
a) Synthesis of methyl 2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate
Prepared by the method of example 4, step a, starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (4-fluoro-3- (methoxycarbonyl) phenyl) boronic acid.
b) Synthesis of 2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 583.2 (M+H).
d) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
Reference is made to example 4, procedure d, from the product of step c.
1 H NMR(400MHz,DMSO-d 6 )δ9.26(s,1H),8.58(s,1H),8.48(d,J=8Hz,1H),8.11-7.99(d,J=8Hz,1H),7.80-7.77(m,1H),7.66-7.57(m,4H),7.24-7.22(d,J=8Hz,1H),6.90-6.88(d,J=8Hz,2H),4.15-4.10(m,2H),3.20-3.10(br,3H),2.89-3.05(br,3H).LC/MS(ESI+):453.2(M+H).
Example 21 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
a) Synthesis of methyl 3-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate
Prepared by the method of step a of example 4 starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (3-fluoro-5- (methoxycarbonyl) phenyl) boronic acid.
b) Synthesis of 3-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 583.2 (M+H).
d) Synthesis of (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):453.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ9.44(s,1H),8.64(s,1H),8.21-8.19(d,J=8Hz,1H),8.10-7.99(d,J=8Hz,1H),7.80-7.77(m,1H),7.66-7.57(m,5H),7.24-7.22(d,J=8Hz,1H),6.90-6.88(d,J=8Hz,2H),4.15-4.10(m,2H),3.20-3.10(br,3H),2.89-3.05(br,3H).
Example 22 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -4-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
Specific preparation method referring to example 4, 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one was prepared by reacting (2-fluoro-5- (methoxycarbonyl) phenyl) boronic acid as starting material.
LC/MS(ESI+):453.2(M+H)
1 H NMR(400MHz,DMSO)δ8.43(s,1H),8.29(d,J=8.3Hz,1H),8.02(dt,J=9.2,4.6Hz,1H),7.97–7.89(m,1H),7.41(t,J=9.1Hz,1H),7.29(dd,J=14.3,7.8Hz,1H),7.11–7.04(m,2H),6.98(t,J=8.6Hz,1H),4.10(d,J=5.8Hz,1H),3.87(d,J=17.0Hz,2H),2.95(dd,J=13.6,5.3Hz,1H),2.84–2.74(m,1H),2.67(d,J=6.0Hz,2H),2.37(s,4H).
Example 23N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -5- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) nicotinamide
The specific preparation method is described in example 4, and (R) -4-chloro-5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidine-7 (6H) -ketone and (5- (methoxycarbonyl) pyridine-3-yl) boric acid are used as starting materials for reaction.
LC/MS(ESI+):435.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ9.05(s,1H),8.91(s,1H),8.80(s,1H),8.51(d,J=8.0Hz,1H),8.31(s,1H),7.29(q,J=7.4Hz,1H),7.16–7.04(m,2H),6.99(t,J=8.3Hz,1H),4.13(s,1H),3.29–3.09(m,3H),2.98(dq,J=13.9,4.8Hz,2H),2.80(dd,J=13.6,8.9Hz,1H),2.70(d,J=6.1Hz,2H),2.35(d,J=16.2Hz,1H),1.00(d,J=7.0Hz,3H).
Example 24N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -2-fluoro-5- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) benzamide
a) Synthesis of methyl (R) -2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) benzoate
By the method of step a, reference example 4, was synthesized starting from (4-fluoro-3- (methoxycarbonyl) phenyl) boronic acid and (R) -4-chloro-5-methyl-5, 8-dihydropyrido [2,3-d ] pyrimidin-7 (6H) -one.
b) Synthesis of (R) -2-fluoro-5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) benzoic acid
Reference is made to the procedure of example 4, step b, from the product of step a.
c) Synthesis of N- ((S) -1- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -2-fluoro-5- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) benzamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione.
d) N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -2-fluoro-5- ((R) -5-methyl-7-oxo-5, 6,7, 8-tetrahydropyrido [2,3-d ] pyrimidin-4-yl) benzamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):452.1(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.73(s,1H),8.28(d,J=8.4Hz,1H),7.75-7.68(m,1H),7.66(d,J=6.4Hz,1H),7.47-7.40(m,1H),7.33-7.26(m,1H),7.12-7.05(m,2H),7.00(t,J=8.5Hz,1H),4.35-4.17(m,1H),4.10(d,J=6.0Hz,1H),3.28-3.24(m,1H),3.02-2.96(m,1H),2.93(s,2H),2.78-2.72(m,2H),2.67(d,J=5.8Hz,2H),2.34(d,J=15.9Hz,1H),0.97(d,J=7.0Hz,3H).
Example 25 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) nicotinamide
a) Synthesis of methyl 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) nicotinate
Prepared by the method of step a of example 4 starting from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (5- (methoxycarbonyl) pyridin-3-yl) boronic acid.
b) 5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) nicotinic acid
Reference is made to the procedure of example 4, step b, from the reaction of the product of step a. LC/MS (ESI+): 286.1[ M+H ] +.
c) Synthesis of (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) nicotinamide
Prepared by the method of example 4, step c, from the product of step b by reaction with (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione. LC/MS (ESI+): 566.2[ M+H ] +.
d) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -5- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) nicotinamide
Reference is made to example 4, procedure d, from the product of step c.
LC/MS(ESI+):436.2(M+H).
Example 26 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5- (2-methoxyethyl) -7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
Specific preparation referring to example 4, the title compound was prepared starting from 4-chloro-5- (2-methoxyethyl) -5, 8-dihydropteridin-7 (6H) -one and (3- (methoxycarbonyl) phenyl) boronic acid prepared in preparation 13.
LC/MS(ESI+):479.1(M+H)
EXAMPLE 27N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (4-methyl-2-oxo-1, 4-dihydro-2H-pyrimido [4,5-d ] [1,3] oxazin-5-yl) benzamide
The title compound was prepared by the specific preparation method described in example 4 starting from 5-chloro-4-methyl-1, 4-dihydro-2H-pyrimido [4,5-d ] [1,3] oxazin-2-one prepared in preparation 11 and (3- (methoxycarbonyl) phenyl) boronic acid.
LC/MS(ESI+):436.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.85(s,1H),8.31(d,J=8.4Hz,1H),8.01–7.87(m,2H),7.79–7.68(m,1H),7.61(t,J=7.7Hz,1H),7.35–7.21(m,1H),7.15–7.03(m,2H),6.97(tt,J=7.9,3.3Hz,1H),5.74(dq,J=10.2,6.7Hz,1H),4.12(td,J=8.1,5.0Hz,1H),3.06–2.74(m,2H),2.68(dd,J=6.1,2.1Hz,2H),1.33(dd,J=6.7,4.2Hz,3H).
Example 28 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5- (2-hydroxyethyl) -7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
a) 5- (2- ((tert-Butyldimethylsilanyloxy) ethyl) -4-chloro-8- (2, 4-dimethoxybenzyl) -5, 8-dihydro-pteridin-7 (6H) -one
Sodium hydride (60%, 0.24 g) was added to an N, N-dimethylformamide solution (10 mL) containing 4-chloro-8- (2, 4-dimethoxybenzyl) -5, 8-dihydropteridin-7 (6H) -one (1 g), and (2-bromoethoxy) (tert-butyl) dimethylsilane (0.7 g) was slowly dropped after releasing no bubbles, and the reaction mixture was stirred at 0℃for 3 hours. After completion of the reaction, the reaction mixture was poured into water (100 mL), followed by extraction with ethyl acetate (100 mL). The organic phase was washed with saturated sodium chloride solution (100 ml x 3), dried over anhydrous sodium sulfate and the solvent was distilled off under reduced pressure. The product was purified by silica gel column chromatography to give 0.3g of a yellow solid which was used directly in the next step.
b) 3- (5- (2- ((tert-Butyldimethylsilanyloxy) ethyl) -8- (2, 4-dimethoxybenzyl) -7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzoic acid methyl ester
The product (0.3 g), (3- (methoxycarbonyl) phenyl) boric acid (0.2 g), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (47 mg) and cesium carbonate (0.4 g) obtained in the step a were dissolved in a mixed solution of dioxane (20 mL) and water (2 mL), heated to reflux under nitrogen protection, heating was stopped after 8 hours, and the reaction solution was evaporated to dryness under reduced pressure and then subjected to column chromatography to obtain a pale yellow solid (240 mg). .
c) 3- (5- (2-hydroxyethyl) -7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzoic acid methyl ester
The product of step b (260 mg) was dissolved in trifluoroacetic acid (5 ml) and reacted at 55℃for 4 hours. After the reaction, ph=8 was adjusted with saturated sodium bicarbonate solution, extracted with ethyl acetate (5 ml×4), the organic layers were combined, dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation under reduced pressure, and the product was purified by column chromatography to give a white solid (98 mg).
d) 3- (5- (2-hydroxyethyl) -7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzoic acid
98mg of the product obtained in step c is dissolved in 10mL of methanol, 10mL of 2M lithium hydroxide solution is added, the mixture is stirred at room temperature overnight, the methanol is removed by rotary evaporation, the pH is adjusted to 4-5 by adding 2M hydrochloric acid, EA extraction is carried out, an organic phase is taken, anhydrous sodium sulfate is dried, and then the mixture is subjected to column chromatography to obtain 55mg of light yellow solid.
e) (S) -N- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3- (5- (2-hydroxyethyl) -7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
50mg of 3- (5- (2-hydroxyethyl) -7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzoic acid, 70mg of (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione, 113mg of benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate, 52mg of N, N-diisopropylethylamine were dissolved in 5mL of N, N-dimethylformamide and stirred at room temperature overnight. Pouring the reaction solution into water, precipitating solid, filtering, drying, and purifying by column chromatography to obtain 52mg pale yellow solid.
f) (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5- (2-hydroxyethyl) -7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
50mg of the product from step e is dissolved in 5mL of methanol, 44mg of hydrazine hydrate is added, the mixture is stirred at room temperature overnight, the reaction mixture is evaporated to dryness under reduced pressure, and 14mg of off-white solid is obtained by column chromatography.
LC/MS(ESI+):465.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.53(d,J=13.5Hz,1H),8.36(d,J=6.4Hz,1H),8.28(d,J=8.3Hz,1H),8.14(t,J=7.3Hz,1H),7.86(d,J=7.8Hz,1H),7.55(t,J=7.8Hz,1H),7.30(dd,J=14.3,7.9Hz,1H),7.15–7.05(m,2H),6.99(dd,J=11.9,5.4Hz,1H),4.53(s,1H),3.89(d,J=17.4Hz,2H),3.24–3.09(m,4H),2.69(ddd,J=21.0,12.9,5.7Hz,4H).
Example 29 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5- (cyanomethyl) -7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
Specific preparation referring to example 4, the title compound was prepared starting from 2- (4-chloro-7-oxo-7, 8-dihydropteridin-5 (6H) -yl) acetonitrile and (3- (methoxycarbonyl) phenyl) boronic acid.
LC/MS(ESI+):460.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.52(s,1H),8.39(d,J=8.4Hz,1H),8.26(s,1H),8.03(d,J=7.8Hz,1H),7.90(d,J=7.8Hz,1H),7.55(t,J=7.7Hz,1H),7.30(dd,J=14.3,8.1Hz,1H),7.10(d,J=8.1Hz,2H),6.99(t,J=8.0Hz,1H),4.17(s,2H),4.06–3.80(m,3H),2.83(dd,J=26.7,12.9Hz,2H),2.72(t,J=14.3Hz,2H).
Example 30 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (7-methyl-8-oxo-8, 9-dihydro-7H-purin-6-yl) benzamide
Specific preparation referring to example 4, the title compound was prepared starting from 6-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one and (3- (methoxycarbonyl) phenyl) boronic acid.
LC/MS(ESI+):421.1(M+H)
Example 31 (S) -N- (1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5, 5-dimethyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide
The title compound was prepared according to the specific preparation method described in example 4 starting from 4-chloro-5, 5-dimethyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] pyrimidin-6-one and (3- (methoxycarbonyl) phenyl) boronic acid.
LC/MS(ESI+):434.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.73(s,1H),8.30(d,J=8.3Hz,1H),7.93(dd,J=9.4,5.9Hz,2H),7.66–7.55(m,2H),7.27(dd,J=14.4,7.8Hz,1H),7.07(t,J=6.6Hz,2H),6.97(t,J=8.6Hz,1H),4.17–4.06(m,1H),2.87–2.73(m,2H),2.65(dd,J=13.5,6.0Hz,2H),1.12(d,J=8.3Hz,6H).
Example 32N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide
Specific preparation referring to example 4, the title compound was prepared starting from 4-chloro-5-methyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] pyrimidin-6-one and (3- (methoxycarbonyl) phenyl) boronic acid.
LC/MS(ESI+):420.2(M+H)
1 H NMR(400MHz,DMSO-d 6 )δ8.81(d,J=1.0Hz,1H),8.53(dd,J=9.6,6.3Hz,2H),8.29(d,J=8.7Hz,1H),7.91(d,J=7.8Hz,1H),7.60(t,J=7.7Hz,1H),7.39–7.24(m,1H),7.10(q,J=7.6Hz,2H),7.07–6.87(m,1H),6.76(s,1H),3.03–2.88(m,2H),2.81(dd,J=13.4,9.0Hz,2H),2.68(d,J=6.1Hz,2H),1.24(s,3H).
Example 33N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5-hydroxy-5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide
a) 3- (5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzoic acid methyl ester
250mg of 4-chloro-5-methyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] pyrimidin-6-one, 347mg of (3- (methoxycarbonyl) phenyl) boric acid, 50mg of [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride and 886mg of cesium carbonate were dissolved in a mixed solution of 10mL of dioxane and 1mL of water, heated to reflux under nitrogen protection, heating was stopped after 5 hours, and the reaction solution was evaporated to dryness under reduced pressure and then subjected to column chromatography to obtain 200mg of pale yellow solid.
LC/MS(ESI+):284.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ11.63(s,1H),8.80(s,1H),8.53(s,1H),8.20(d,J=7.8Hz,1H),8.11(d,J=7.7Hz,1H),7.71(t,J=7.8Hz,1H),4.26(q,J=7.6Hz,1H),3.91(s,3H),1.12(d,J=7.6Hz,3H).
b) 3- (5-hydroxy-5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzoic acid
200mg of methyl 3- (5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzoate is dissolved in 20mL of methanol, 6mL of a 2M lithium hydroxide solution is added, stirring is carried out at room temperature for 6 hours, 2M hydrochloric acid is added to adjust the pH to 6-7, the reaction solution is evaporated to dryness under reduced pressure, and then column chromatography is carried out to obtain 160mg of off-white solid.
LC/MS(ESI+):286.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ11.73(s,1H),8.81(d,J=5.2Hz,2H),8.58(d,J=7.8Hz,1H),8.08(d,J=7.7Hz,1H),7.65(t,J=7.8Hz,1H),7.00(s,1H),3.15(s,1H),1.24(s,3H).
c) N- ((S) -1- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3- (5-hydroxy-5-methyl-6-oxo) -6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide
160mg of 3- (5-hydroxy-5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzoic acid, 240mg of (S) -2- (2-amino-3- (3-fluorophenyl) propyl) isoindoline-1, 3-dione, 391mg of benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate and 119mg of N, N-diisopropylethylamine were dissolved in 10mL of N, N-dimethylformamide and stirred overnight at room temperature. The reaction solution was poured into ethyl acetate, the organic phase was taken out after washing with water, evaporated to dryness under reduced pressure, and purified by column chromatography to obtain 180mg of off-white solid. LC/MS (ESI+): 566.1 (M+H).
d) N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -3- (5-hydroxy-5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide
180mg of N- ((S) -1- (1, 3-dioxoisoindolin-2-yl) -3- (3-fluorophenyl) propan-2-yl) -3- (5-hydroxy-5-methyl-6-oxo) -6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide was dissolved in 20mL of methanol, after adding 328mg of 80% hydrazine hydrate, stirring overnight at room temperature, evaporating the reaction solution under reduced pressure, and obtaining 80mg of an off-white solid by column chromatography.
LC/MS(ESI+):436.1(M+H).
1 H NMR(400MHz,DMSO-d 6 )δ8.82(s,1H),8.54(ddd,J=9.9,7.0,2.2Hz,2H),8.31(d,J=8.4Hz,1H),7.97–7.83(m,1H),7.60(t,J=7.7Hz,1H),7.30(ddd,J=14.4,8.0,6.5Hz,1H),7.10(ddd,J=10.1,7.6,5.5Hz,2H),6.98(tt,J=8.5,3.1Hz,1H),4.14(tt,J=10.9,8.5,4.1Hz,1H),2.96(ddd,J=13.7,5.5,2.0Hz,1H),2.90–2.75(m,1H),2.77–2.56(m,2H),2.48(s,2H),1.24(s,3H).
Example 34N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -3- ((R) -5-hydroxy-5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide and N- ((S) -1-amino-3- (3-fluorophenyl) propan-2-yl) -3- ((S) -5-hydroxy-5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-4-yl) benzamide
The product of example 33 was resolved by chiral column to give the title compound.
Chiral resolution conditions:
instrument: waters SFC; chiral column Daicel Chiralcel AD,250×30mm I.D.,5 μm; the mobile phase is that the A phase is carbon dioxide and the B phase is ethanol (containing 0.1% ammonia water); a: b=60: 40 (volume ratio); the flow rate is 60mL/min; column temperature 38 ℃. The product with retention time of 13-18min was collected as isomer 1 (24 mg) and the product with retention time of 27-41min was collected as isomer 2 (35 mg).
HPLC analysis conditions:
instrument: waters UPCC, chiral column: daicel Chiralcel AD, 2.1X105 mm I.D.,3 μm, mobile phase: phase A is CO 2 Phase B ethanol (0.1% diethanolamine) gradient elution: 0-6.5min, phase b: 5% -40% (volume ratio), flow rate: 1mL/min, column temperature: 40 ℃.
Isomer 1: RT=5.3 min, LC/MS (ESI+): 436.1 (M+H)
Isomer 2: RT=6.5 min, LC/MS (ESI+): 436.1 (M+H)
Example 35 (S) -N- (1-amino-3- (3, 5-difluorophenyl) propan-2-yl) -3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydrodishidin-4-yl) benzamide
Specific preparation method referring to example 4, methyl 3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate was prepared from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (3-methoxycarbonyl) phenyl) boronic acid as starting materials, followed by hydrolysis, reaction with (S) -2- (2-amino-3- (3, 5-difluorophenyl) propyl) isoindoline-1, 3-dione and deprotection reaction to prepare the title compound.
LC/MS(ESI+):453.2(M+H).
1 H NMR(400MHz,CDCl 3 )δ8.61(d,J=3.3Hz,1H),8.40(d,J=24.0Hz,1H),8.18(d,J=8.0Hz,1H),7.94–7.72(m,2H),7.55(dd,J=15.4,7.8Hz,2H),6.92–6.74(m,2H),6.68(t,J=9.0Hz,1H),4.36(d,J=6.6Hz,1H),3.89(s,2H),3.04(m,1H),2.96–2.79(m,2H),2.54(s,3H),2.27–2.15(m,1H),2.02(m,1H).
Example 36N- (1-amino-3- (3, 4-difluorophenyl) propan-2-yl) -3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydronaphthyridin-4-yl) benzamide
Specific preparation method referring to example 4, methyl 3- (5-methyl-7-oxo-5, 6,7, 8-tetrahydropteridin-4-yl) benzoate was prepared from 4-chloro-5-methyl-5, 8-dihydropteridin-7 (6H) -one and (3-methoxycarbonyl) phenyl) boronic acid, followed by hydrolysis, reaction with 2- (2-amino-3- (3, 5-difluorophenyl) propyl) isoindoline-1, 3-dione, and deprotection reaction to prepare the title compound.
LC/MS(ESI+):453.2(M+H)
1 H NMR(400MHz,CDCl 3 )δ8.61(d,J=3.3Hz,1H),8.40(d,J=24.0Hz,1H),8.18(d,J=8.0Hz,1H),7.94–7.72(m,2H),7.55(dd,J=15.4,7.8Hz,2H),6.92–6.74(m,2H),6.68(t,J=9.0Hz,1H),4.36(d,J=6.6Hz,1H),3.89(s,2H),3.04(m,1H),2.96–2.79(m,2H),2.54(s,3H),2.27–2.15(m,1H),2.02(m,1H).
Test example 1 in vitro enzyme Activity test
1. Experimental procedure
The method Mobility shift assay is adopted to establish AKT1, AKT2 and AKT3 kinase activity detection platforms, and compound activity determination is carried out. Compounds were subjected to 3-fold gradient dilutions (total 10 concentrations) starting at 100. Mu.M in 100% DMSO (purchased from Sigma, cat: D8418-1L), each concentration transferred to a destination plate OptiPlate-384F (purchased from Perkinelmer, cat: 6007290) using a dispenser Echo 550. Positive and negative control wells were added 250nL DMSO. Add 10. Mu.L of AKT kinase solution (purchased from Carna, AKT1 Cat:01-101, AKT2 Cat:01-102, AKT3 Cat:01-103; final concentration of AKT1 kinase reaction 0.5nM, final concentration of AKT2 kinase reaction 0.05nM, final concentration of AKT3 kinase reaction 0.25 nM) to each of the compound wells and positive control wells; mu.L of 1 XKinase buffer (containing 50mM HEPES,10mM MgCl) was added to the negative control wells 2 2mM DTT). Then centrifuged at 1000rpm for 30 seconds, mixed with shaking and incubated at room temperature for 10 minutes. Then 15. Mu.L of ATP (purchased from Sigma, cat. A7699-5G, lot. SLBT6850, final concentration of ATP detected by AKT1 of 120. Mu.M, final concentration of ATP detected by AKT2 of 500. Mu.M, final concentration of ATP detected by AKT3 of 29.3. Mu.M) and C were addedThe reaction was initiated with an aliper substrate 6 (purchased from GL, cat:116368, substrate final concentration 3. Mu.M) mixture, with a total reaction volume of 25. Mu.L. 384 well plates were centrifuged at 1000rpm for 30 seconds, and after shaking mixing, incubated at room temperature for respective times (60 min for AKT1 assay, 30min for AKT2 assay, and 10min for AKT3 assay). The kinase reaction was stopped by adding 30. Mu.L of stop detection solution (stop detection solution containing 50mM EDTA), centrifuging at 1000rpm for 30 seconds, and shaking and mixing. Conversion was read at Caliper EZ Reader II (device parameters: -1.2PSI, upstream voltage-500, downstream voltage-2250, reading interval 45 s). The IC of the compound was obtained using a GraphPad Prism 5 software log (inhibitor) vs. response-Variable slope fit 50 Values.
The inhibition rate calculation formula:wherein:
conversion% _sample: is a conversion reading of the sample;
convertion% _min: negative control Kong Junzhi, representing conversion reading without enzyme wells;
Convesion% _max: positive control Kong Junzhi, represents a conversion reading without compound inhibition wells.
2. Experimental results
The experimental results are shown in the following table:
/>
/>
/>
/>

Claims (41)

1. a compound of formula I or a pharmaceutically acceptable salt thereof,
wherein:
R 1 and R is 2 Each independently selected from H or C1-C6 alkyl;
m is selected from 0 or 1;
a is optionally substituted with 1-5R 3 Substituted phenyl, wherein each R 3 Each independently selected from halogen or C1-C6 alkyl, wherein C1-C6 alkyl is optionally substituted with halogen;
selected from->
G is selected from 5-6 membered heteroaryl or phenyl, wherein the heteroaryl or phenyl is optionally substituted with R 4 Substituted, and R 4 Selected from halogen or C1-C6 alkyl;
l isA group, wherein:
Y 1 selected from CY 11 Y 12 Or NY 13 ,Y 2 Selected from O, CY 21 Y 22 Or a bond, and is satisfied when Y 2 When selected from O, Y 1 Selected from CY 11 Y 12 And Y is 11 And Y 21 Each independently selected from H, OH, halogen or C1-C6 alkyl, wherein C1-C6 alkyl is optionally substituted with halogen, OH, C1-C3 alkoxy or CN;
Y 12 and Y 22 Each independently selected from H or C1-C6 alkyl;
Y 13 selected from H or C1-C6 alkyl, wherein C1-C6 alkyl is optionally substituted with halogen, OH, C1-C3 alkoxy or CN.
2. A compound of formula I according to claim 1, wherein:
R 1 and R is 2 Each independently selected from H, methyl, ethyl or isopropyl;
m is selected from 1;
a is represented by 1 or 2R 3 A substituted phenyl group;
R 3 independently selected from F, cl, br, I, CH 3 、C 2 H 5 Or CF (CF) 3
3. A compound of formula I according to claim 2, wherein: r is R 1 Selected from H, R 2 Selected from H, methyl, ethyl or isopropyl.
4. A compound of formula I according to claim 2, wherein: r is R 1 Selected from H, R 2 Selected from H or isopropyl.
5. A compound of formula I according to claim 2, wherein: r is R 1 Selected from H, R 2 Selected from H.
6. A compound of formula I according to claim 2, wherein: a is selected from the following groups:
7. a compound of formula I according to claim 2, wherein: r is R 3 Selected from F, cl or CF 3
8. A compound of formula I according to claim 2, wherein: r is R 3 Selected from F or Cl.
9. As claimed in claim 2The compound of formula I, wherein: r is R 3 Selected from F.
10. A compound of formula I according to claim 1, wherein:
a is selected from the following groups:
11. a compound of formula I according to claim 1, wherein:
a is selected from the following groups:
12. a compound of formula I according to claim 1, wherein:
a is selected from the following groups:
13. a compound of formula I according to claim 1, wherein:
a is selected from
14. A compound of formula I according to claim 1, wherein:selected from->
15. A compound of formula I according to claim 1, wherein:
G is selected from: five membered heteroaryl, pyridinyl or phenyl, wherein said five membered heteroaryl, pyridinyl or phenyl is optionally substituted with R 4 Substituted, and R 4 Selected from halogen or C1-C6 alkyl.
16. A compound of formula I according to claim 1, wherein:
g is selected from optionally R 4 Substituted with the following groups:
wherein,represents the position where G is linked to Q by a chemical bond, < >>Represents the position where G is attached to L by a chemical bond; r is R 4 Selected from halogen or C1-C6 alkyl.
17. A compound of formula I according to claim 15 or 16, wherein: r is R 4 Selected from CH 3 F or Cl.
18. A compound of formula I according to claim 15 or 16, wherein: r is R 4 Selected from CH 3 Or F.
19. A compound of formula I according to claim 15, wherein:
g is selected from optionally R 4 Substituted with the following groups:
20. a compound of formula I according to claim 15, wherein:
g is selected from optionally R 4 Substituted with the following groups:
21. a compound of formula I according to claim 15, wherein:
g is selected from the following groups:
22. a compound of formula I according to claim 15, wherein:
g is selected from the following groups:
23. a compound of formula I according to claim 15, wherein:
g is selected from the following groups:
24. a compound of formula I according to claim 15, wherein:
G is selected from the following groups:
25. a compound of formula I according to claim 1, wherein:
Y 1 selected from (a): CY (CY) 11 Y 12 Wherein Y is 11 Selected from H, OH or C1-C6 alkyl, wherein C1-C6 alkyl is optionally substituted with F; y is Y 12 Selected from H or CH 3
Y 1 Selected from (b): NY (NY) 13 Wherein Y is 13 Selected from H, CH 3 、C 2 H 5 、CH 2 CH 2 CH 3 、CHCH 3 CH 3 、CH 2 OH、C 2 H 4 OH、C 3 H 6 OH、CH 2 OCH 3 、CH 2 OC 2 H 5 、C 2 H 4 OCH 3 、CH 2 CN or C 2 H 4 CN;
Y 2 Selected from O, CY 21 Y 22 Or a bond, wherein Y 21 And Y 22 All are H.
26. A compound of formula I according to claim 25, wherein: y is Y 11 Selected from H, CH 3 Or CF (CF) 3
27. A compound of formula I according to claim 25, wherein: y is Y 11 Selected from CH 3 Or CF (CF) 3
28. A compound of formula I according to claim 25, wherein: y is Y 11 Selected from CH 3
29. A compound of formula I according to claim 25, wherein: y is Y 13 Selected from CH 3 、C 2 H 4 OH or CH 2 CN。
30. A compound of formula I according to claim 25, wherein: y is Y 13 Selected from CH 3
31.A compound of formula I according to claim 25, wherein: y is Y 2 Selected from CH 2 Or a root key.
32. A compound of formula I according to claim 1, L is selected from the following groups:
33. the compound of formula I according to claim 1,
l is selected from the following groups:
34. the compound of formula I according to claim 1,
l is selected from the following groups:
35. a compound of formula II or a pharmaceutically acceptable salt thereof,
therein, A, G, L, R 1 And R is 2 Is as defined for the compounds of formula I.
36. A compound of formula III-1 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 31 Compounds of the formula I Y 11 ,R 32 Compounds of the formula I Y 12 Is defined in (a).
37. A compound of formula III-2 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 33 Compounds of the formula I Y 13 Is defined in (a).
38. A compound of formula III-3 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 34 Compounds of the formula I Y 11 Definition of R 35 Compounds of the formula I Y 12 Is defined in (a).
39. A compound of formula III-4 or a pharmaceutically acceptable salt thereof,
therein, A, R 1 、R 2 And G is as defined for compounds of formula I, R 36 Compounds of the formula I Y 13 Is defined in (a).
40. The following compounds or pharmaceutically acceptable salts thereof:
41. use of a compound according to any one of claims 1 to 40, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the prevention and/or treatment of AKT protein kinase mediated diseases or conditions.
CN201910858374.XA 2019-09-11 2019-09-11 PKB inhibitors Active CN112480116B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910858374.XA CN112480116B (en) 2019-09-11 2019-09-11 PKB inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910858374.XA CN112480116B (en) 2019-09-11 2019-09-11 PKB inhibitors

Publications (2)

Publication Number Publication Date
CN112480116A CN112480116A (en) 2021-03-12
CN112480116B true CN112480116B (en) 2024-03-29

Family

ID=74920223

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910858374.XA Active CN112480116B (en) 2019-09-11 2019-09-11 PKB inhibitors

Country Status (1)

Country Link
CN (1) CN112480116B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2024517861A (en) * 2021-05-06 2024-04-23 エクセンシア・エイアイ・リミテッド PKC-θ modulators

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2014073979A (en) * 2012-10-03 2014-04-24 Daiichi Sankyo Co Ltd Triazolothiazepine derivative
CN105153164A (en) * 2014-05-30 2015-12-16 南京明德新药研发股份有限公司 Dihydropyrimidocyclic derivative taken as HBV inhibitor
TW201602107A (en) * 2014-06-17 2016-01-16 南京明德新藥研發股份有限公司 Pyrido[1,2-a]pyrimidone derivatives as a mTOR/PI3K suppressor
CN105837576A (en) * 2015-01-14 2016-08-10 湖北生物医药产业技术研究院有限公司 BTK inhibitors
CN108349977A (en) * 2015-01-20 2018-07-31 无锡福祈制药有限公司 Jak inhibitor
WO2019034128A1 (en) * 2017-08-18 2019-02-21 浙江海正药业股份有限公司 Pyrrolotriazine derivative, preparation method and use thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2014073979A (en) * 2012-10-03 2014-04-24 Daiichi Sankyo Co Ltd Triazolothiazepine derivative
CN105153164A (en) * 2014-05-30 2015-12-16 南京明德新药研发股份有限公司 Dihydropyrimidocyclic derivative taken as HBV inhibitor
TW201602107A (en) * 2014-06-17 2016-01-16 南京明德新藥研發股份有限公司 Pyrido[1,2-a]pyrimidone derivatives as a mTOR/PI3K suppressor
CN105837576A (en) * 2015-01-14 2016-08-10 湖北生物医药产业技术研究院有限公司 BTK inhibitors
CN108349977A (en) * 2015-01-20 2018-07-31 无锡福祈制药有限公司 Jak inhibitor
WO2019034128A1 (en) * 2017-08-18 2019-02-21 浙江海正药业股份有限公司 Pyrrolotriazine derivative, preparation method and use thereof
CN109400610A (en) * 2017-08-18 2019-03-01 浙江海正药业股份有限公司 Pyrrolo-triazine analog derivative, preparation method and its purposes in medicine

Also Published As

Publication number Publication date
CN112480116A (en) 2021-03-12

Similar Documents

Publication Publication Date Title
CN111138301B (en) Biphenyl compound, intermediate, preparation method, pharmaceutical composition and application
JP6549109B2 (en) Sulfoximine substituted quinazolines and their use as MNK1 and / or MNK2 kinase inhibitors
CN114008035A (en) SHP2 phosphatase allosteric inhibitor
CN101878031A (en) The inhibitors of kinases that is used for the treatment of myeloproliferative disease and other proliferative diseases
AU2005270049A1 (en) Small molecule thienopyrimidine-based protein tyrosine kinase inhibitors
FR2941948A1 (en) AZAINDOLE DERIVATIVES AS INHIBITOR OF ABL KINASE PROTEINS AND SRC
CN115991706A (en) PIM kinase inhibitors
CN115073469A (en) Preparation and application of pyrrolopyrimidine compound as kinase inhibitor
CN112480116B (en) PKB inhibitors
CA3093323C (en) Oxazino-quinazoline and oxazino-quinoline type compound, preparation method and uses thereof
CN115703761A (en) Compound as WWP1 inhibitor and application thereof
CN109384784B (en) Sulfonamide derivative, preparation method thereof and application thereof in medicine
CN116891464A (en) AT2R agonist
CN115894381A (en) 2,4,5-trisubstituted pyrimidine compound and preparation method and application thereof
WO2015110092A1 (en) 4-substituted pyrrolo[2,3-d]pyrimidine compound and use thereof
AU2014359456A1 (en) Sulfoximine substituted quinazolines for pharmaceutical compositions
JP2024520929A (en) Heterocyclic Derivatives as CAMKK2 Inhibitors
JP2023538137A (en) Aromatic ring lactam compound, its production method and use
WO2016054804A1 (en) Substituted pyrimidines as inhibitors of hif prolyl hydroxylase
AU2022305807B2 (en) Pyrimidine compounds for use as map4k1 inhibitors
CN115745975B (en) JAK kinase domain and pseudokinase domain co-inhibition prodrug, preparation method and medical application
ES2309518T3 (en) DIAMINOPIRROLOQUINAZOLINE COMPOUNDS AS INHIBITORS OF PROTEIN TIROSINA FOSFATASA.
CN114989176B (en) Imidazopyridazine derivative and application thereof
CN109305961A (en) Imatinib amine derivative with pharmaceutical activity and preparation method thereof
CN113461687B (en) 2, 8-aza- [4,5] decaspirocyclic ketone derivative and preparation method 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
GR01 Patent grant
GR01 Patent grant