CN116669738A - Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof - Google Patents

Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof Download PDF

Info

Publication number
CN116669738A
CN116669738A CN202180076242.7A CN202180076242A CN116669738A CN 116669738 A CN116669738 A CN 116669738A CN 202180076242 A CN202180076242 A CN 202180076242A CN 116669738 A CN116669738 A CN 116669738A
Authority
CN
China
Prior art keywords
cancer
compound
mmol
kras
membered
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180076242.7A
Other languages
Chinese (zh)
Inventor
谢成英
郑苏欣
陆晓杰
郑明月
乔刚
叶阳亮
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.)
Suzhou Almai Biotechnology Co ltd
Original Assignee
Suzhou Almai Biotechnology 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 Suzhou Almai Biotechnology Co ltd filed Critical Suzhou Almai Biotechnology Co ltd
Publication of CN116669738A publication Critical patent/CN116669738A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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

Landscapes

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

Abstract

Pyrimidopyridinone derivatives as SOS1 inhibitors, and preparation method and application thereof are provided. The pyrimidopyridone derivative has a structure shown in a formula (I), has remarkable RAS signal pathway inhibiting effect, and is used for treating and/or preventing cancers, such as pancreatic cancer, colorectal cancer, lung cancer, hepatocellular carcinoma, renal cancer, gastric cancer, cholangiocarcinoma and the like.

Description

Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof Technical Field
The invention belongs to the technical field of pyrimidopyridone derivatives, and particularly relates to a pyrimidopyridone derivative serving as an SOS1 inhibitor, a preparation method and application thereof.
Background
RAS family proteins belong to a small GTPase comprising three subfamilies of KRAS, NRAS and HRAS. Mutated RAS gene is an important oncogene, and the discovery of RAS gene mutation is found in 20-30% of human tumors, particularly pancreatic, colorectal and lung cancers. The various isoforms of RAS proteins have a balance of GTP-bound active and GDP-bound inactive states, GTPase-activated proteins (GTPase-activating proteins, GAPs) may facilitate the conversion of GTP to GDP, thereby converting RAS proteins to inactive states, and guanine nucleotide exchange factors (guanine nucleotide exchange factor, GEFs) may facilitate the release of GDP and the binding of GTP, thereby converting RAS proteins to active states. Activation of RAS proteins promotes proliferation of cells, apoptosis escape, metabolic recombination and the like through signal pathways of RAS-RAF-MEK-ERK and RAS-PI3K-PDK1-AKT, thereby promoting occurrence and development of tumors.
SOS1 (son of sevenless 1) is a key guanine nucleotide exchange factor (GE)F) Can bind with RAS protein, promote RAS protein binding with GTP, and turn RAS protein to active state. Recent researches find that SOS1 inhibitor can inhibit the growth of various RAS mutant cells singly, can also generate synergistic effect with MEK inhibitor, and has obvious inhibition effect on KRAS-driven tumors 1-2 . The development of SOS1 inhibitors is a hotspot of research, and SOS1 inhibitors of different structural types are reported in several patents, such as WO2018172250, WO2019201848, WO2018115380, WO2019122129, WO2020173935, WO2020180768 and WO2020180770, etc.
However, there is still uncertainty in the effectiveness, safety or selectivity of these compounds and test drugs disclosed in the prior art, and thus there is a need to study and develop new selective SOS1 inhibitors.
Reference is made to:
1、Hillig et al.Discovery of poetent SOS1inhibitors that block RAS activation via disruption of the RAS-SOS1interaction.PNAS.116,2251-2560(2019).
2、Hofmann et al.BI-3406,a potent and selective SOS1::KRAS interaction inhibitor,is effective in KRAS-driven cancers through combined MEK inhibition.Cancer Discov.CD-20-0142(2020).
disclosure of Invention
In order to solve the above problems of the prior art, the present invention aims to provide a pyrimidopyridone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof, for screening out a compound used as an SOS1 inhibitor having excellent properties in terms of performance such as effectiveness, safety and selectivity.
It is another object of the present invention to provide a process for the preparation of said derivatives, pharmaceutically acceptable salts thereof, tautomers thereof or stereoisomers thereof.
In order to achieve the aim of the invention, the invention adopts the following technical scheme:
in a first aspect, the present invention provides a pyrimidopyridone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof, wherein the pyrimidopyridone derivative has a structure as shown in formula (I):
wherein: r is R 1 Selected from hydrogen or C 1 -C 3 An alkyl group; preferably hydrogen or methyl;
R 2 selected from hydrogen or C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl are optionally substituted with 1-3R 21 Substituted;
R 21 selected from C 1 -C 3 Alkyl, hydroxy, halogen, cyano, amino, C 1 -C 3 Alkoxy or = O;
l may be absent or selected from O, NH or N- (C) 1 -C 3 An alkyl group);
R 3 selected from H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl are optionally substituted with 1-3R 31 Substituted;
R 31 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, =o, -NHCOR 32 or-COR 32
R a Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
R b Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Halogenated compoundsAlkyl or 3-6 membered cycloalkyl;
R 32 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl or 4-7 membered heterocyclyl;
AR is selected from 6-10 membered aryl or 5-10 membered heteroaryl, wherein aryl or heteroaryl is optionally substituted with 1-4R 4 Substituted;
R 4 selected from H, halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy-C 1 -C 3 Alkyl, hydroxy-C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR a 、-NR a R b 6-10 membered aryl or 5-10 membered heteroaryl, wherein 6-10 membered aryl or 5-10 membered heteroaryl is optionally substituted with 1-4R c Substituted;
R c selected from H, halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy-C 1 -C 3 Alkyl, hydroxy-C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR a 、-NR a R b 、NR a R b -C 1 -C 4 Alkyl, NR a R b -C 1 -C 4 A haloalkyl group; the hetero atom in the heterocyclic group or heteroaryl group in the formula (I) is 1-3 and is selected from one or more of oxygen, nitrogen and sulfur.
Preferably, the pyrimidopyridone derivative has a structure shown in a formula (II):
wherein R is 1 、R 2 、R 3 And R is 4 Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4);
preferably, said compound of formula (II), said phenyl group being optionally substituted with 1 to 4R 4 Substituted when said R 4 When the number of R is 2-4, R is 4 May be the same or different;
and/or when said R 4 Is C 1 -C 3 In the case of haloalkyl, said R 4 1-3, when more than two R's are present 4 When said R is 4 May be the same or different;
and/or when said R 4 Is C 1 -C 3 When halogenoalkyl, the halogen atom is fluorine;
and/or when said R 4 In the case of halogen, R is 4 1-3, when two or more R's are present 4 When said R is 4 May be the same or different;
and/or when said R 4 When halogen, the halogen atom is fluorine;
and/or when said R 4 is-NR a R b When said R is 4 1-3, when two or more R's are present 4 When said R is 4 May be the same or different;
and/or when said R 4 is-NR a R b When said R is a And R is b May be the same or different;
and/or R 3 H, C of a shape of H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein 4-7 membered heterocyclyl is optionally substituted with 1-3R 31 Substituted;
and/or when R 3 Is optionally covered with 1-3R 31 Substituted 4-7 membered heterocyclyl, said R 31 When the number of the components is 2-3, R 31 The same or different;
and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
and/or, the R 3 Is optionally covered with 1-3R 31 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
And/or, the R 3 Is optionally covered with 1-3R 31 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclic group, the R 31 Selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, =o, -NHCOR 32 or-COR 32
R a Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
R b selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
R 32 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl or 4-7 membered heterocyclyl.
Preferably, the pyrimidopyridone derivative has a structure shown in a formula (III):
wherein R is 1 、R 2 、R 3 And R is 4 Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Further preferred, for compounds of formula (III), the phenyl group is optionally substituted with 1 to 4R 4 Substituted when said R 4 When the number of R is 2-4, R is 4 May be the same or different;
and/or when said R 4 Is C 1 -C 3 In the case of haloalkyl, said R 4 1-3, when more than two R's are present 4 When said R is 4 May be the same or different;
and/or when said R 4 Is C 1 -C 3 When halogenoalkyl, the halogen atom is fluorine;
And/or when said R 4 In the case of halogen, R is 4 1-2, when more than two R's are present 4 When said R is 4 May be the same or different;
and/or when said R 4 When halogen, the halogen atom is fluorine;
and/or when said R 4 is-NR a R b When said R is 4 1-3, when more than two R's are present 4 When said R is 4 May be the same or different;
and/or when said R 4 is-NR a R b When said R is a And R is b May be the same or different;
and/or R 3 H, C of a shape of H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein 4-7 membered heterocyclyl is optionally substituted with 1-3R 31 Substituted;
and/or when R 3 Is optionally covered with 1-3R 31 Substituted 4-7 membered heterocyclic group,the R is 31 When the number of the components is 2-3, R 31 The same or different;
and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
and/or, the R 3 Is optionally covered with 1-3R 31 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
and/or, the R 3 Is optionally covered with 1-3R 31 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclic group, the R 31 Selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, =o, -NHCOR 32 or-COR 32
R a Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
R b selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
R 32 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl or 4-7 membered heterocyclyl.
Preferably, the pyrimidopyridone derivative has a structure shown in a formula (IV):
wherein R is 1 、R 2 、R 3 、R c And L has the same defined range as above; m=1-4 (e.g., m=1, m=2, m=3, m=4). Further preferred, the compound of formula (IV), L is selected from O, NH or NCH 3
Further preferably, the pyrimidopyridinone derivative is selected from any one of the following structures:
in a second aspect, the present invention provides a process for the preparation of a pyrimidopyridone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof according to the first aspect, selected from one of the following two schemes:
scheme one
The preparation method of the compound shown in the general formula (I) or the stereoisomer, the tautomer or the medicinal salt thereof comprises the following steps:
Preparation of key intermediate (I-A):
step one, performing metal catalytic cross coupling on an aromatic compound of a general formula (I-1) to obtain a compound of a general formula (I-2);
secondly, reacting the compound shown in the general formula (I-2) under the condition of a catalyst to obtain a chiral sulfimide compound shown in the general formula (I-3);
thirdly, reducing the compound of the general formula (I-3) by a metal reducing agent to obtain a chiral compound of the general formula (I-4);
fourthly, the compound of the general formula (I-4) is cracked into the chiral benzylamine compound of the general formula (I-A) under the acid condition.
Preparation of key intermediate (I-B), method one:
the first step, a compound of a general formula (I-5) and tert-butyl bromoacetate undergo substitution reaction under alkaline conditions to obtain a compound of a general formula (I-6);
secondly, removing tert-butyl from the compound of the general formula (I-6) under an acidic condition to obtain a compound of the general formula (I-7);
thirdly, carrying out esterification reaction on the compound of the general formula (I-7) and alcohol under an acidic condition to obtain a compound of the general formula (I-8);
fourth, the compound of the general formula (I-9) and the amine of the general formula (I-10) are subjected to substitution reaction to obtain the compound of the general formula (I-11);
fifthly, carrying out halogenation reaction on the compound of the general formula (I-11) to obtain a compound of the general formula (I-12);
sixthly, under alkaline condition, the compound of the general formula (I-12) and boric acid or boron ester compound are subjected to Suzuki reaction in the presence of a metal catalyst and a ligand to obtain a compound of the general formula (I-13);
Seventh, oxidizing double bonds in the compound of the general formula (I-13) to obtain a compound of the general formula (I-14);
eighth, the compound of formula (I-14) and the compound of formula (I-8) are subjected to basic conditions to give the compound of formula (I-B).
Preparation of key intermediate (I-B), method II:
the first step, the compound of the general formula (I-15) and the amine of the general formula (I-10) are subjected to substitution reaction to obtain the compound of the general formula (I-16);
in the second step, the compound of the formula (I-16) and the compound of the formula (I-8) are subjected to basic conditions to obtain the compound of the formula (I-B).
Preparation of general formula (I):
in the first step, the compound of the general formula (I-B) and the compound of the general formula (I-A) are subjected to substitution reaction to obtain the compound of the general formula (I).
Therein, X, X 1 、X 2 And X 3 Is halogen; x is preferably bromine; x is X 1 、X 2 Preferably chlorine; x is X 3 Preferably iodine; r is R 1 、R 2 、R 3 AR and L have the same defined ranges as described above.
Scheme II
The preparation method of the compound shown in the general formula (I) or the stereoisomer, the tautomer or the medicinal salt thereof comprises the following steps:
firstly, a compound of a general formula (I-12) and methyl acrylate are subjected to Heck reaction under the regulation of an alkaline catalyst and a metal catalyst to obtain a compound of a general formula (I-17);
secondly, forming amide in the molecule of the compound of the general formula (I-17) under alkaline conditions to obtain a compound of the general formula (I-18);
Thirdly, carrying out halogenation reaction on the compound with the general formula (I-18) to obtain a compound with the general formula (I-19);
step four, demethylating the compound of the general formula (I-19) under an acidic condition to obtain a compound of the general formula (I-20);
fifthly, reacting the compound of the general formula (I-20) with 2,4, 6-triisopropylbenzenesulfonyl chloride or phosphorus oxychloride under alkaline conditions to obtain a compound of the general formula (I-21);
sixthly, reacting the compound shown in the general formula (I-21) with the compound shown in the general formula (I-A) under alkaline conditions to obtain a compound shown in the general formula (I-22);
seventh, the compound of the general formula (I-22) and the compound of the general formula (I-23) are subjected to Suzuki reaction under alkaline conditions in the presence of a metal catalyst and a ligand to obtain the compound of the general formula (I).
Wherein X is 1 、X 3 And X 4 Is halogen; x is X 1 Preferably chlorine; x is X 3 Preferably iodine; x is X 4 Preferably bromine; x is X 5 Is halogen orW isR 1 、R 2 、R 3 AR and L have the same defined ranges as described above.
In the case of the above-mentioned preparation method,
the reagent for providing alkaline conditions is selected from organic bases or inorganic bases, wherein the organic bases are one or more of triethylamine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, lithium bistrimethylsilylamino, sodium tert-butoxide, sodium methoxide and potassium tert-butoxide, and the inorganic bases are one or more of sodium hydride, potassium phosphate, sodium carbonate, potassium acetate, cesium carbonate, sodium hydroxide, potassium hydroxide, sodium bicarbonate and lithium hydroxide;
The reagent providing the acidic condition is one or more of hydrogen chloride, 1, 4-dioxane solution of hydrogen chloride, methanol solution of hydrogen chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid and phosphoric acid;
the metal catalyst is palladium/carbon, raney nickel, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, and [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (Pd (dppf) Cl) 2 ) [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride dichloromethane complex, ditriphenylphosphine palladium dichloride (Pd (PPh) 3 )Cl 2 ) And tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) One or more of the following;
the ligand is one or more of 2-dicyclohexylphosphine-2, 6' -dimethoxybiphenyl (SPhos), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (Xantphos), 2-dicyclohexylphosphine-2, 4, 6-triisopropylbiphenyl (XPhos), 2-dicyclohexylphosphino-2 ' - (N, N-dimethylamine) -biphenyl (DavePhos), 1' -bis (diphenylphosphine) ferrocene (Dppf) and 1,1' -binaphthyl-2, 2' -bis-diphenylphosphine (BINAP), preferably 1,1' -binaphthyl-2, 2' -bis-diphenylphosphine (BINAP);
the reducing agent is one or more of sodium borohydride, potassium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride and lithium aluminum hydride;
The oxidant is one or more of potassium permanganate, manganese dioxide, potassium dichromate, sodium dichromate and potassium osmium;
the above reaction is preferably carried out in a solvent selected from one or more of N, N-dimethylformamide, N-methylpyrrolidone, dimethylsulfoxide, 1, 4-dioxane, water, tetrahydrofuran, methylene chloride, 1, 2-dichloroethane, methanol, ethanol, toluene, petroleum ether, ethyl acetate, N-hexane and acetone.
In a third aspect, the present invention provides a pharmaceutical composition comprising a pyrimidopyridone derivative and/or a stereoisomer, tautomer, pharmaceutically acceptable salt according to the first aspect; and pharmaceutically acceptable carriers and/or excipients and/or sustained release agents.
In the present invention, the term "comprising" means that the various ingredients may be applied together in a mixture or composition of the present invention. Thus, the terms "consisting essentially of and" consisting of are encompassed by the term "containing.
In the present invention, a "pharmaceutically acceptable" ingredient is a substance that is suitable for use in humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio.
In the present invention, a "pharmaceutically acceptable carrier" is a pharmaceutically acceptable solvent, suspending agent or excipient for delivering the active substance of the present invention or a physiologically acceptable salt thereof to an animal or human. The carrier may be a liquid or a solid.
In the present invention, the pharmaceutical composition contains a safe and effective amount (e.g., 0.001 to 99.9 parts by weight, more preferably, 0.01 to 99 parts by weight, still more preferably, 0.1 to 90 parts by weight) of the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or excipient, wherein the total weight of the composition is 100 parts by weight.
Alternatively, the pharmaceutical composition of the present invention contains 0.001 to 99.9wt%, more preferably 0.01 to 99 wt%, still more preferably 0.1 to 90 wt% of the compound represented by formula (I) or a pharmaceutically acceptable salt thereof based on the total weight; and a pharmaceutically acceptable carrier or excipient, wherein the total weight of the composition is 100 wt%.
In another preferred embodiment, the preferred ratio of the compound of formula (I) to the pharmaceutically acceptable carrier, excipient or slow release agent is such that formula (I) is present as active ingredient in an amount of more than 65% by weight, the remainder being present in an amount of 0.5 to 40% by weight, or more preferably 1 to 20% by weight, or most preferably 1 to 10% by weight.
The pharmaceutical compositions of the present invention comprise in unit dose form 0.05mg to 500mg, preferably 0.5mg to 200mg, more preferably 0.1mg to 100mg of the compound of formula (I), enantiomer, racemate, pharmaceutically acceptable salt or mixture thereof per dose.
When the pharmaceutical composition contains an additional pharmaceutically active ingredient for treating or preventing cancer, the amount of the active ingredient may be generally the conventional amount or less in the art.
The pharmaceutical compositions of the invention may be in a variety of forms, such as tablets, capsules, powders, syrups, solutions, suspensions, aerosols and the like, wherein the compounds of formula (I) may be presented in a suitable solid or liquid carrier or diluent. The pharmaceutical compositions of the present invention may also be stored in a suitable injectable or instillation disinfection device. The pharmaceutical composition may also contain odorants, flavoring agents, etc.
The compounds of formula (I) or pharmaceutical compositions comprising the compounds of formula (I) of the present invention may be administered to mammals, including humans, clinically by oral, nasal, dermal, pulmonary or gastrointestinal routes of administration. The preferred route of administration is oral. The preferred daily dosage is 0.5mg-200mg/kg body weight, administered once or in divided doses. Regardless of the method of administration, the optimal dosage for an individual will depend on the particular treatment. Typically starting from a small dose, the dose is gradually increased until the most appropriate dose is found.
The effective dose of the active ingredient used may vary with the compound used, the mode of administration and the severity of the condition to be treated. However, generally, satisfactory results are obtained when the compounds of the invention are administered at a dose of about 1-300mg/kg animal body weight per day, preferably at 1-3 divided doses per day, or in a sustained release form. For most large mammals, the total daily dose is about 5-1000mg, preferably about 10-500mg. Dosage forms suitable for oral administration comprise about 1-200mg of the active compound intimately mixed with a solid or liquid pharmaceutically acceptable carrier. This dosage regimen can be adjusted to provide the optimal therapeutic response. For example, separate doses may be administered several times per day, or the dose may be proportionally reduced, as dictated by the urgent need for the treatment of the condition.
The compounds or pharmaceutically acceptable salts thereof and compositions thereof may be administered orally as well as intravenously, intramuscularly or subcutaneously. Preferred pharmaceutical compositions are solid compositions, especially tablets and solid filled or liquid filled capsules, from the standpoint of ease of preparation and administration. Oral administration of the pharmaceutical composition is preferred.
The solid support comprises: starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin, while the liquid carrier includes: sterile water, polyethylene glycols, non-ionic surfactants and edible oils (e.g., corn, peanut and sesame oils) as are appropriate to the nature of the active ingredient and the particular mode of administration desired. Adjuvants commonly used in the preparation of pharmaceutical compositions may also advantageously be included, for example flavouring agents, pigments, preservatives and antioxidants such as vitamin E, vitamin C, BHT and BHA.
The active compound or a pharmaceutically acceptable salt thereof and compositions thereof may also be administered parenterally or intraperitoneally. Solutions or suspensions of these active compounds (as the free base or pharmaceutically acceptable salt) may also be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquids, polyethylene glycols and mixtures thereof in oils. Under normal conditions of storage and use, these formulations contain preservatives to prevent microbial growth.
Pharmaceutical forms suitable for injection include: sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, these forms must be sterile and must be fluid to facilitate the discharge of the fluid from the syringe. Must be stable under the conditions of manufacture and storage and must be able to prevent the contaminating effects of microorganisms such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, alcohols (such as glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
The compounds of formula (I) or pharmaceutically acceptable salts and compositions thereof may also be administered in combination with other active ingredients or drugs for the treatment or prophylaxis of chronic pain disorders. When two or more drugs are administered in combination, there is generally an effect superior to the effect of the two drugs administered separately.
In a fourth aspect, the present invention provides the use of a pyrimidopyridone derivative according to the first aspect, a stereoisomer, a tautomer, a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to the third aspect, alone or in combination with a KRAS inhibitor, for the preparation of a medicament for the treatment of cancer or for the preparation of a SOS1 inhibitor; preferably, the cancer includes, but is not limited to, astrocyte cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, laryngeal cancer, pancreatic cancer, lung cancer, oral cancer, ovarian cancer, prostate cancer, thyroid cancer, sarcoma, renal cancer, and cholangiocarcinoma; further preferably, the cancer includes, but is not limited to: pancreatic cancer, colorectal cancer, lung cancer, hepatocellular carcinoma, renal cancer, gastric cancer, and cholangiocarcinoma; and/or the KRAS inhibitor is a KRAS G12C, KRAS G12V, KRAS G12S or KRAS G12D inhibitor; and/or the MEK signaling pathway inhibitor such as a RAF, MEK or ERK1/2 inhibitor.
Preferably, the pyrimidopyridinone derivative, stereoisomer, tautomer, pharmaceutically acceptable salt thereof according to the first aspect or the pharmaceutical composition according to the third aspect is in a dosage range selected from 10-100mg/kg, preferably 25-50mg/kg; the KRAS G12C inhibitor is selected from 3-100mg/kg, preferably 5-30mg/kg; the dosage range of MEK inhibitor is selected from 0.1-0.2mg/kg, preferably 0.125mg/kg.
Preferably, the KRAS G12C inhibitor is MRTX849 and the MEK inhibitor is Trametinib.
The use of the fourth aspect, wherein the cancer is a RAS family related cancer; preferably KRAS, HRAS or NRAS G12C-associated cancer, KRAS, HRAS or NRAS G12D-associated cancer, KRAS, HRAS or NRAS G12V-associated cancer, KRAS, HRAS or NRAS G12S-associated cancer, KRAS, HRAS or NRAS G13D-associated cancer, KRAS, HRAS or NRAS G13C-associated cancer, KRAS, HRAS or NRAS Q61L-associated cancer, KRAS, HRAS or NRAS a 146T-associated cancer, KRAS, HRAS or NRAS a 146V-associated cancer or KRAS, HRAS or NRAS a 146P-associated cancer.
In another aspect, there is provided a method of treating cancer comprising providing to a cancer patient a therapeutically effective amount of formula (I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the compound or a pharmaceutically acceptable salt thereof. In one embodiment, the cancer is a RAS family-related cancer. In one embodiment, the cancer is NF-l/NF-2-associated cancer.
The compositions and methods provided herein are useful for treating a variety of cancers, including prostate, breast, brain, skin, cervical cancer, testicular cancer, and the like. More specifically, cancers treatable by the compositions and methods of the invention include, but are not limited to, tumor types such as astrocyte, breast, cervical, colorectal, endometrial, esophageal, stomach, head-neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid cancers and sarcomas. More specifically, these compounds are useful in the treatment of: and (3) heart: sarcomas (hemangiosarcoma, fibrosarcoma, rhabdomyoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma, and teratoma; lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondrioma, mesothelioma; gastrointestinal tract: esophageal (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), gastric (carcinoma, lymphoma, leiomyosarcoma), pancreatic (ductal adenocarcinoma, insulinoma, glucagon, gastrinoma, carcinoid, VIPoma), small intestine (adenocarcinoma, lymphoma, carcinoid, kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large intestine (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); urogenital tract: kidney (adenocarcinoma, nephroblastoma (nephroblastoma), lymphoma, leukemia), bladder and urinary tract (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); liver: liver cancer (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; biliary tract: gall bladder cancer, ampulla cancer, bile duct cancer; bone: osteosarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, ewing's sarcoma, malignant lymphoma (reticuloma), multiple myeloma, malignant giant cell tumor chordoma, bone chronicity tumor (osteochondrosarcoma), benign tumor, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumor; the nervous system: skull (bone tumor, hemangioma, granuloma, xanthoma, osteomyelitis), meninges (meningioma, glioma), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germ cell tumor (pineal tumor), glioblastoma multiforme, oligodendroglioma, schwannoma, retinoblastoma, congenital tumor), spinal fibroma, meningioma, glioma, sarcoma; gynaecology: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-cancerous cervical dysplasia), ovary (ovarian carcinoma (serous cystic adenocarcinoma, myxocyst adenocarcinoma, unclassified carcinoma), granulosa sheath cell carcinoma, backing cell carcinoma, germ cell carcinoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, cystoid sarcoma (embryonal rhabdomyosarcoma), fallopian tube (carcinoma), hematology (acute and chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), hodgkin's disease, non-hodgkin's lymphoma (malignant lymphoma), skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, kaposi's sarcoma, nevus, lipoma, hemangioma, skin fibroma, lupus erythematosus, psoriasis, and adrenal gland neuroblastoma).
In one embodiment, the cancer is a RAS family-related cancer, such as KRAS, NRAS, or HRAS-related cancer. In certain embodiments, the RAS family-related cancer is non-small cell lung cancer or pancreatic cancer. In one embodiment, the cancer is a cancer associated with SOS 1. In certain embodiments, the SOS 1-associated cancer is lung adenocarcinoma, embryonal rhabdomyosarcoma, support cell testicular tumor, and skin granulosa cell tumor. In one embodiment, the cancer is NF-1-associated cancer.
Preferably, the cancer is pancreatic cancer, colorectal cancer, lung cancer, hepatocellular cancer, renal cancer, gastric cancer, or cholangiocarcinoma.
Interpretation of the terms
Unless stated to the contrary, some of the terms used in the specification and claims of the present invention are defined as follows:
as used herein, "KRAS G12C" refers to a mutant form of a mammalian KRAS protein that contains an amino acid that replaces glycine with a cysteine at amino acid position 12.
The determination of amino acid codons and residue positions for human KRAS is based on UniProtKB/SwissProt P01116: amino acid sequence of variant p.Gly12 Cys.
"KRAS G12D" refers to a mutant of a mammalian KRAS protein that contains an amino acid that replaces glycine at position 12 with aspartic acid. The amino acid codon and residue positions of human KRAS are according to UniProtKB/Swi-prot P01116: variant P.Gly12 Asp.
"KRAS G12V" refers to a mutant of a mammalian KRAS protein that contains an amino acid valine substituted for glycine at position 12. The amino acid codon and residue positions of human KRAS are according to UniProtKB/Swi-prot P01116: variant P.Gly12 Val.
"KRAS G12S" refers to a mutant of a mammalian KRAS protein comprising an amino acid having a serine substituted for glycine at amino acid position 12. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt P01116: the amino acid sequence identified by variant p.Gly12Ser.
As used herein, "KRAS G12A" refers to a mutant form of a mammalian KRAS protein that contains an amino acid with alanine in place of glycine at amino acid position 12. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt P01116: amino acid sequence identified by variant p.Gly12 Ala.
"KRAS G13D" refers to a mutant of a mammalian KRAS protein comprising an amino acid with an aspartic acid substituted for glycine at amino acid position 13. The determination of amino acid codons and residue positions for human KRAS is based on UniProtKB/SwissProt P01116: the amino acid sequence determined by variant p.gly 13 Asp.
"KRAS G13C" refers to a mutant of a mammalian KRAS protein that contains an amino acid with a cysteine at amino acid 13 in place of glycine. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt-Prot P01116: the amino acid sequence of the variant P.Gly 13Cys is determined.
As used herein, "KRAS Q61L" refers to a mutant form of a mammalian KRAS protein that contains the amino acid leucine in place of glutamine at amino acid 61. The determination of amino acid codons and residue positions of human KRAS is based on the amino acid sequence of UniProtKB/SwissProt P01116: variant p.Gln61Leu.
As used herein, "KRAS A146T" refers to a mutant form of a mammalian KRAS protein that contains an amino acid that replaces alanine with threonine at amino acid position 146. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt P01116: the amino acid sequence identified by variant p.al46thr.
As used herein, "KRAS A146V" refers to a mutant form of a mammalian KRAS protein that contains an amino acid with valine at amino acid position 146 in place of alanine. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt P01116: amino acid sequence identified by variant p.al46Val.
As used herein, "KRAS A146P" refers to a mutant form of a mammalian KRAS protein that contains an amino acid with proline at amino acid position 146 in place of alanine. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt P01116: the amino acid sequence identified by variant p.Al46Pro.
As used herein, "HRAS G12C" refers to a mutant form of mammalian HRAS protein which contains an amino acid which replaces glycine with a cysteine at amino acid position 12. According to UniProtKB/SwissProt P01112: amino acid sequences identified by variants p.Gly12 Cys, amino acid codons and residue positions of human HRAs were determined.
"HRAS G12D" refers to a mutant of a mammalian HRAS protein containing the amino acid substitution amino acid position 12 of aspartic acid-substituted glycine, the determination of the amino acid codon and residue position of human HRAS being based on UniProtKB/SwissProt P01112: amino acid sequence of variant p.gly 12 Asp.
"HRAS G12S" refers to a mutant of a mammalian HRAS protein containing an amino acid with serine substituted for glycine at 12. Amino acid codon and residue positions of human HRAS are according to UniProtKB/SwissProt P01112: variant P.Gly12Ser.
"HRAS G12A" refers to a mutant of a mammalian HRAS protein, the amino acid glycine being replaced with alanine at protein 12. The amino acid codon and residue positions of human KRAS are according to UniProtKB/SwissProt P01112: amino acid sequence identified by variant p.gly 12 Ala.
"HRAS G13D" refers to a mutant of mammalian HRAS protein, which contains an amino acid with aspartic acid substituted for glycine at position 13. Amino acid codon and residue positions of human HRAS are according to UniProtKB/Swi-prot P01112: variant p.gly 13 Asp.
"HRAS G13C" is a mutant of a mammalian HRAS protein, which contains a cysteine at amino acid 13 in place of the glycine amino acid. Determination of amino acid codon and residue positions for human HRAS is based on UniProtKB/SwissProt-Prot P01112: amino acid sequence of variant p.gly 13 Cys.
As used herein, "HRAS Q61L" refers to a mutant form of a mammalian HRAS protein comprising the amino acid leucine in place of glutamine at amino acid position 61, with amino acid position 61. The amino acid codons and residue positions of human HRAs were determined from the amino acid sequence identified by UniProtKB/SwissProt P01112 variant p.gln61leu.
As used herein, "HRAS A146T" refers to a mutant form of mammalian HRAS protein which contains an amino acid threonine at amino acid position 146 in place of alanine. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01112: the amino acid sequence identified by variant p.Al46Thr.
As used herein, "HRAS A146V" refers to a mutant form of mammalian HRAS protein which contains the amino acid valine at amino acid position 146 in place of alanine. One amino acid replaces alanine at amino acid position 146. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01112: the amino acid sequence identified by variant p.al46Val.
As used herein, "HRAS A146P" refers to a mutant form of mammalian HRAS protein which contains an amino acid with proline at amino acid position 146 in place of alanine. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01112: variant p.Al46Pro identified amino acid sequence determination.
"NRAS G12C" refers to a mutant form of a mammalian NRAS protein that contains an amino acid that has a cysteine at amino acid 12 that replaces glycine. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt-Prot P01111: variant p.gly 12 Cys.
"NRAS G12D" refers to a mutant of a mammalian NRAS protein that contains an amino acid that replaces glycine at position 12 with aspartic acid. Amino acid codon and residue positions of human NRAS are according to UniProtKB/Swi-prot P01111: variant P.Gly12 Asp.
"NRAS G12S" refers to a mutant of a mammalian NRAS protein that contains an amino acid that replaces glycine with serine at amino acid 12. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01111: the amino acid sequence identified by variant p.Gly12Ser.
"NRAS G12A" refers to a mutant form of a mammalian NRAS protein that contains an amino acid that replaces glycine with alanine at amino acid 12. The determination of amino acid codons and residue positions for human KRAS is based on UniProtKB/SwissProt P01111: amino acid sequence of variant p.Gly12 Ala.
As used herein, "NRAS G13D" refers to a mutant form of a mammalian NRAS protein that contains an amino acid substitution of aspartic acid at amino acid 13 for glycine at the amino acid position. The determination of amino acid codons and residue positions for human NRAS is based on UniProtKB/SwissProt P01111: amino acid sequence determined by variant p.Gly13 Asp.
"HNras G13C" refers to a mutant form of a mammalian NRAS protein that contains an amino acid with a cysteine substituted for glycine at amino acid 13. The determination of amino acid codons and residue positions for human NRAS is based on UniProtKB/SwissProt P01111: amino acid sequence of variant p.gly 13 Cys.
As used herein, "HRAS Q61L" refers to a mutant form of a mammalian HRAS protein comprising the amino acid leucine in place of glutamine at amino acid 61, which has an amino acid position of 61. The amino acid codons and residue positions of human HRAs were determined from the amino acid sequence identified by UniProtKB/SwissProt P01112 variant p.gln61leu.
As used herein, "NRAS A146T" refers to a mutant form of a mammalian NRAS protein that contains an amino acid threonine at amino acid 146 instead of alanine. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01111: the amino acid sequence identified by variant p.al46thr.
As used herein, "NRAS A146V" refers to a mutant form of a mammalian NRAS protein that contains an amino acid valine for alanine at amino acid 146. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01111: the amino acid sequence identified by variant p.al46Val.
As used herein, "NRAS a146P" refers to a mutant form of a mammalian NRAS protein that contains an amino acid with proline at amino acid 146 in place of alanine. Amino acid codon and residue positions of human NRAS are according to UniProtKB/SwissProt P01111: the amino acid sequence identified by variant p.Al46Pro.
As used herein, "RAS family member" or "RAS family" refers to KRAS, HRAS, NRAS and its activating mutants, including at the G12, G13, Q61 and A146 positions.
As used herein, a 'RAS family related disease or disorder' refers to a disease or disorder associated with, mediated by or having an activating RAS mutation, such as RAS at the G12, G13, Q61 or a146 position. Non-limiting examples of RAS family related diseases or disorders are KRAS, HRAS or NRAS G12C related cancer, KRAS, HRAS or NRAS G12D related cancer, KRAS, HRAS or NRAS G12V related cancer, KRAS, HRAS or NRAS G12A related cancer, KRAS, HRAS or NRAS G13D related cancer, KRAS, HRAS or NRAS G13C related cancer, KRAS, HRAS or NRAS Q61L related cancer, KRAS, HRAS or NRAS a146T related cancer, KRAS, HRAS or NRAS a146V related cancer or KRAS, HRAS or NRAS 146P related cancer.
As used herein, an "SOS 1-related disease or disorder" refers to a disease or disorder associated with, mediated by, or having an activated SOS1 mutation. Examples of activating SOS1 mutations include SOS1N233S and SOS1N233Y mutations.
As used herein, "SOS1N233S" refers to a mutant form of a mammalian SOS1 protein that contains an amino acid substitution of serine for glutamine at amino acid position 233. The determination of amino acid codon and residue positions of human SOS1 is based on UniProtKB/SwissProt Q07889: amino acid sequence identified as variant p.Gln2338er.
As used herein, "SOS1N233Y" refers to a mutant form of a mammalian SOS1 protein in which the amino acid containing tyrosine substituted glutamine has been substituted for amino acid 233. The determination of amino acid codon and residue positions of human SOS1 is based on UniProtKB/SwissProt Q07889: amino acid sequence identified by variant p.gln2335yr.
As used herein, "SOS1 inhibitor" refers to a compound of the present invention, as described herein, represented by formula (I). These compounds negatively inhibit all or part of the interaction of SOS1 with RAS family mutants or SOS1 activating mutations, thereby reducing and/or modulating the nucleotide exchange activity of RAS family member-SOS 1 complexes.
As used herein, "KRAS inhibitor" refers to inhibitors known in the art for various KRAS mutations. Inhibitors such as KRAS G12C, KRAS G12D, KRAS G12V may be targeted.
As used herein, an "MEK signaling pathway" inhibitor refers to inhibitors known in the art for RAF, MEK, and ERK 1/2.
"NF-1/NF-2 related disease or disorder" refers to a disease or disorder caused by a loss-of-function mutation in a neurocellulase (NF-1) gene or a neurocellulase 2 (NF-2) gene.
"loss-of-function mutation" as used herein refers to any point mutation, splice site mutation, fusion, nonsense mutation (amino acid mutation to stop codon), intra-frame mutation or frame transfer mutation, including insertions and deletions, as well as homozygous deletion of the gene encoding the protein in the cell of interest or cancer cell, resulting in partial or complete loss of presence, activity and/or function of the encoded protein.
"alkyl" refers to a saturated aliphatic hydrocarbon group comprising 1 to 20 carbon atoms, or 1 to 10 carbon atoms, or 1 to 6 carbon atoms, or 1 to 4 carbon atoms, or 1 to 3 carbon atoms, or 1 to 2 carbon atoms, saturated straight or branched chain monovalent hydrocarbon groups, wherein the alkyl groups may be independently optionally substituted with one or more substituents described herein. Examples of alkyl groups further include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, and the like. The alkyl group may be optionally substituted or unsubstituted.
"alkenyl" refers to a straight or branched monovalent hydrocarbon radical of 2 to 12 carbon atoms, or 2 to 8 carbon atoms, or 2 to 6 carbon atoms, or 2 to 4 carbon atoms, wherein at least one C-C is sp 2 Double bonds, wherein the alkenyl groups may be independently optionally substituted with 1 or more substituents as described herein, specific examples of which include, but are not limited to, vinyl, allyl, and allyl groups, and the like. Alkenyl groups may be optionally substituted or unsubstituted.
"cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring comprising 3 to 20 carbon atoms, preferably comprising 3 to 12 carbon atoms, more preferably comprising 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups. Cycloalkyl groups may be optionally substituted or unsubstituted.
"spirocycloalkyl" refers to a 5 to 18 membered, two or more cyclic structure, and monocyclic polycyclic groups sharing one carbon atom (called spiro atom) with each other, containing 1 or more double bonds within the ring, but no ring has a completely conjugated pi-electron aromatic system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spiro group, a double spiro group or a multiple spirocycloalkyl group according to the number of common spiro atoms between rings, preferably single spiro group and double spirocycloalkyl group, preferably 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered. Non-limiting examples of "spirocycloalkyl" include, but are not limited to:
"fused ring alkyl" refers to an all-carbon polycyclic group containing two or more cyclic structures sharing a pair of carbon atoms with each other, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system, preferably 6 to 12 members, more preferably 7 to 10 members. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group. Non-limiting examples of "fused ring alkyl" include, but are not limited to:
"bridged cycloalkyl" means an aromatic system having 5 to 18 members, containing two or more cyclic structures, sharing two all-carbon polycyclic groups with one another that are not directly attached to a carbon atom, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron, preferably 6 to 12 members, more preferably 7 to 10 members. Cycloalkyl groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged cycloalkyl" include, but are not limited to:
The cycloalkyl ring may be fused to an aryl, heteroaryl or heterocyclyl ring, wherein the ring attached to the parent structure is cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, and the like.
"heterocyclyl", "heterocycle" or "heterocyclic" are used interchangeably herein and refer to a saturated or partially unsaturated, monocyclic, bicyclic or tricyclic, non-aromatic heterocyclic group containing 3 to 12 ring atoms, at least one of which is a heteroatom such as oxygen, nitrogen, sulfur, and the like. Preferably having a 5 to 7 membered mono-or 7 to 10 membered bi-or tri-ring, which may contain 1,2 or 3 atoms selected from nitrogen, oxygen and/or sulphur. Examples of "heterocyclyl" include, but are not limited to, morpholinyl, oxetanyl, thiomorpholinyl, tetrahydropyranyl, 1-dioxo-thiomorpholinyl, piperidinyl, 2-oxo-piperidinyl, pyrrolidinyl, 2-oxo-pyrrolidinyl, piperazin-2-one, 8-oxa-3-aza-bicyclo [3.2.1] octyl, and piperazinyl. The heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is heterocyclyl. The heterocyclyl group may be optionally substituted or unsubstituted.
"spiroheterocyclyl" refers to a 5-to 18-membered, two or more cyclic structure, polycyclic group having single rings sharing one atom with each other, containing 1 or more double bonds within the ring, but no ring having a fully conjugated pi-electron aromatic system in which one or more ring atoms are selected from nitrogen, oxygen, sulfur or S (O) m The remaining ring atoms are carbon, m=1 or 2. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spiroheterocyclyl groups are classified into a single spiroheterocyclyl group, a double spiroheterocyclyl group or a multiple spiroheterocyclyl group according to the number of common spiro atoms between rings, and preferably a single spiroheterocyclyl group and a double spiroheterocyclyl group. More preferably a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered single spiro heterocyclic group. Non-limiting example package of "spiroheterocyclylIncluding but not limited to:
"fused heterocyclyl" refers to an all-carbon polycyclic group containing two or more cyclic structures sharing a common pair of atoms with each other, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system in which one or more of the ring atoms is selected from nitrogen, oxygen, sulfur or S (O) m The remaining ring atoms are carbon, m=1 or 2. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group. Non-limiting examples of "fused heterocyclyl" include, but are not limited to:
"bridged heterocyclyl" means a 5-to 18-membered, polycyclic group containing two or more cyclic structures sharing two atoms not directly attached to each other, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system in which one or more of the ring atoms is selected from nitrogen, oxygen, sulfur or S (O) m The remaining ring atoms are carbon, m=1 or 2. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Heterocyclic groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged heterocyclyl" include, but are not limited to:
"aryl" refers to a carbocyclic aromatic system containing one or two rings, wherein the rings may be linked together in a fused manner. The term "aryl" includes aromatic groups such as phenyl, naphthyl, tetrahydronaphthyl. Preferably aryl is C 6 -C 10 Aryl, more preferably aryl is phenyl and naphthyl, most preferably phenyl. Aryl groups may be substituted or unsubstituted. The "aryl" may be fused to a heteroaryl, heterocyclyl, or cycloalkyl group, wherein the aryl ring is attached to the parent structure, non-limiting examples include, but are not limited to:
"heteroaryl" refers to an aromatic 5-to 6-membered monocyclic or 9-to 10-membered bicyclic ring, which may contain 1 to 4 atoms selected from nitrogen, oxygen and/or sulfur. Examples of "heteroaryl" include, but are not limited to, furyl, pyridyl, 2-oxo-1, 2-dihydropyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thienyl, isoxazolyl, oxazolyl, oxadiazolyl, imidazolyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, 1,2, 3-thiadiazolyl, benzodioxolyl, benzimidazolyl, indolyl, isoindolyl, 1, 3-dioxo-isoindolyl, quinolinyl, indazolyl, benzisothiazolyl, benzoxazolyl and benzisoxazolyl. Heteroaryl groups may be optionally substituted or unsubstituted. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples include, but are not limited to:
"alkoxy" refers to a group of (alkyl-O-). Wherein alkyl is as defined herein. C (C) 1 -C 6 Is preferably selected. Examples include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy and the like.
"haloalkyl" refers to an alkyl group having one or more halo substituents, wherein the alkyl group has the meaning as described herein. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, perfluoroethyl, 1-dichloroethyl, 1, 2-dichloropropyl, and the like.
"hydroxy" refers to an-OH group.
"halogen" means fluorine, chlorine, bromine and iodine, preferably fluorine, chlorine and bromine.
"amino" means-NH 2
"cyano" refers to-CN.
"nitro" means-NO 2
"benzyl" means-CH 2 -phenyl.
"carboxy" means-C (O) OH.
"acetyl" means-C (O) CH 3 Or Ac.
"carboxylate" refers to-C (O) O (alkyl) or (cycloalkyl), wherein alkyl, cycloalkyl are as defined above.
"optional" means that the event described may, but need not, occur. For example, "AR is optionally 1 to multiple R c Substitution the description includes that the AR group may be substituted with 1 to a plurality of R c Substituted or not by R c Substitution case.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"substituted" or "substituted" as used herein, unless otherwise indicated, means that the group may be substituted with one or more groups selected from the group consisting of: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, alkenyl, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxyl, carboxylate, =o, -C (O) R b 、-OC(O)R b 、-NR b R b 、-C(O)NR b R b 、-NR b C(O)R b 、-S(O)NR b R b or-S (O) 2 NR b R b Wherein R is b The definition of (C) is as described in the general formula (I).
As used herein, the term "subject," "individual," or "patient" is used interchangeably to refer to any animal, including mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses, primates, and humans. In some embodiments, the patient is a human. In some embodiments, the subject has experienced and/or displayed a disease or at least one symptom of a disease to be treated and/or prevented. In some embodiments, the subject has been determined or diagnosed as a cancer having a KRAS G12 or G13 mutation (e.g., as determined by an FDA-approved regulatory agency, such as an FDA-approved, assay, or kit). In some embodiments, the subject has a tumor positive for a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12S mutation, a KRAS G12V mutation, a KRAS G12A mutation, a KRAS G13D mutation, or a KRAS G13C mutation (e.g., as determined by regulatory agency-approved assays or kits). The subject may be a tumor patient with a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12V mutation, a KRAS G12A mutation, a KRAS G13D mutation, or a KRAS G13C mutation (e.g., approved regulatory bodies-such as FDA approved, analytical, or kit). The subject may be a subject whose tumor has a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12V mutation, a KRAS G12A mutation, a KRAS G13D mutation, or a KRAS G13C mutation (e.g., the tumor is determined by an FDA-approved regulatory agency, kit, or assay). In some embodiments, the subject is suspected of having a KRAS G12 or G13 gene-related cancer. In some embodiments, the subject has a clinical record indicating that the subject has a tumor with a KRAS G12C mutation (and optionally a clinical record indicating that the subject should be treated with any of the compositions provided herein).
The term "pediatric patient" as used herein refers to a patient less than 16 years of age at the time of diagnosis or treatment. The term "child" can also be divided into the following subclasses: neonates (from birth to first month of birth); infants (1 month to two years); children (2 to 12 years); teenagers (12 to 21 years old (up to but not including 22 years of birth)). Berhman RE, kliegman R, arvin AM, nelson we. Nelson textbook, 15 th edition. Philadelphia: w.b. samanders company, 1996; rudolph AM, et al. Pediatric version 21 of rudoffer. New York: mcGrow-Hill, 2002; and Avery MD, first LR. Pediatric medicine, second edition. Balm: williams & Wilkins;1994.
as used herein, an "effective amount" of a compound refers to an amount sufficient to down-regulate or inhibit SOS 1 enzymatic activity.
As used herein, a "therapeutically effective dose" of a compound refers to an amount sufficient to ameliorate or somehow reduce symptoms, stop or reverse progression of a disease, or down regulate or inhibit SOS 1 activity. Such doses may be administered as a single dose or may be administered according to a regimen so as to be effective.
As used herein, "treating" refers to ameliorating or otherwise altering the condition, disorder, or symptom or pathology of a disease in a patient in any manner.
As used herein, "ameliorating a symptom of a particular disease by use of a particular compound or pharmaceutical composition" refers to any reduction, whether permanent or temporary, persistent or temporary, attributable to or associated with the use of the composition.
The definition and use of stereochemistry in the present invention is generally referred to in the following documents:
S.P. Parker, ed., mcGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hillbook Company, new York; and Eliel, e.and Wilen, s., "Stereochemistry of Organic Compounds", john Wiley & Sons, inc., new York,1994. The compounds of the invention may contain asymmetric or chiral centers and thus exist as different stereoisomers. All stereoisomeric forms of the compounds of the invention, including, but in no way limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, form part of the present invention. Diastereomers can be separated into the individual diastereomers by chromatography, crystallization, distillation, or sublimation, based on their physical-chemical differences. Enantiomers may be converted into diastereomeric mixtures by separation by reaction with an appropriate optically active compound (e.g., a chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers, and converting the individual diastereomers to the corresponding pure enantiomers. The intermediates and compounds of the invention may also exist in different tautomeric forms and all such forms are encompassed within the scope of the invention. Many organic compounds exist in optically active form, i.e. they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefix D, L or R, S is used to denote the absolute configuration of the chiral center of the molecule. The prefix d, l or (+), (-) is used to name the sign of the compound plane polarization rotation, where (-) or l means that the compound is left-handed and the prefix (+) or d means that the compound is right-handed. The atoms or groups of atoms of these stereoisomers are connected in the same order but in different steric structures. The particular stereoisomer may be an enantiomer, and the mixture of isomers is commonly referred to as an enantiomeric mixture. 50: mixtures of enantiomers of 50 are referred to as racemic mixtures or racemates, which may result in no stereoselectivity or stereospecificity during chemical reactions. The terms "racemic mixture" and "racemate" refer to a mixture of two enantiomers in equimolar amounts, lacking optical activity.
"tautomer" or "tautomeric form" refers to isomers of structures of different energies that can be interconverted by a low energy barrier. For example, proton tautomers (i.e., proton-shifted tautomers) include tautomerism by proton shift, such as keto-enol and imine-enamine isomerisation. Valency (valence) tautomers include tautomers that reorganize into bond electrons. Unless otherwise indicated, the structural formulae described herein include all isomeric forms (e.g., enantiomers, diastereomers, and geometric isomers): for example, R, S configuration containing asymmetric centers, the (Z), (E) isomers of double bonds, and the conformational isomers of (Z), (E). Thus, individual stereochemical isomers of the compounds of the invention, or enantiomers, diastereomers, or mixtures of geometric isomers thereof, are all within the scope of the invention.
"pharmaceutically acceptable salts" refers to salts of the compounds of the present invention which are safe and effective when used in the human or animal body. Salts of the compounds may be obtained by dissolving the corresponding addition salts in pure solution or in a suitable inert solvent with sufficient amounts of base or acid. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia, magnesium salts, and the like, and pharmaceutically acceptable acid addition salts include inorganic and organic acid salts including hydrochloric acid, hydrobromic acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, monohydrogen sulfate, acetic acid, maleic acid, malonic acid, succinic acid, fumaric acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, methanesulfonic acid, and the like (see Berge et al, "Pharmaceutical Salts", journal of Pharmaceutical Science 66:1-19 (1977)).
The invention provides an SOS1 inhibitor with a novel structure, and test results show that the pyrimidopyridone derivative shows excellent SOS1 inhibition activity and excellent safety and selectivity, and can be used for preparing medicines for treating cancers, especially pancreatic cancer, colorectal cancer, lung cancer, hepatocellular carcinoma, renal cancer, gastric cancer, cholangiocarcinoma and other diseases.
Drawings
FIG. 1 is a graph showing the effect of compounds according to the present invention on KRAS/ERK1/2 signaling pathways of K-562 cells.
Figure 2 is a graph of in vivo anti-tumor effects of SOS1 inhibitors and MRTX849 of the present invention on KRASG12C mutant miappa a-2 pancreatic xenograft tumor nude mouse models, alone or in combination.
FIG. 3 is a graph of the mouse body weight effect of SOS1 inhibitor and MRTX849 according to the present invention on a KRASG12C mutant MIAPaCa-2 pancreatic xenograft tumor nude mouse model, alone or in combination.
FIG. 4 is a graph of in vivo anti-tumor effects of SOS1 inhibitors and Trametinib (Trametinib) according to the invention, alone or in combination, on KRASG12D mutant HPAF-II pancreatic xenograft tumor nude mouse models.
FIG. 5 is a graph of the body weight effects of SOS1 inhibitors and Trametinib (Trametinib) according to the invention, alone or in combination, on a KRASG12D mutant HPAF-II pancreatic xenograft tumor nude mouse model.
Detailed Description
The method of the present invention will be described by way of specific examples, so that the technical solution of the present invention can be understood and grasped more easily, but the present invention is not limited thereto. In the following examples 1 The H NMR spectrum was determined with a Bruker instrument (400 MHz) and the chemical shifts were expressed in ppm. Tetramethylsilane internal standard (0.00 ppm) was used. 1 H NMR representation method: s=singlet, d=doublet, t=triplet, q=quartet, m=multiplet, br=broadened, dd=doublet of doublet, dt=doublet of triplet. If coupling constants are provided, they are in Hz.
The mass spectrum is measured by an LC/MS instrument, and the ionization mode is ESI.
High performance liquid chromatograph model: agilent 1260, siemens flying U3000; chromatographic column model: waters xbridge C18 (4.6. Times.150 mm,3.5 μm); mobile phase: ACN, B Water (0.1% H) 3 PO 4 ) The method comprises the steps of carrying out a first treatment on the surface of the Flow rate: 1.0mL/min; gradient: 5%A for 1min,increase to 20%A within4min,increase to 80%A within 8min,80%A for 2min,back to 5%A within 0.1min; wavelength: 220nm; column incubator: 35 ℃.
The thin layer chromatography silica gel plate is a smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification of the silica gel plate used by the Thin Layer Chromatography (TLC) is 0.2mm-0.3mm, and the specification of the thin layer chromatography separation and purification product is 0.4mm-0.5mm.
Column chromatography generally uses tobacco stage yellow sea silica gel 200-300 mesh silica gel as carrier.
In the following examples, unless otherwise indicated, all temperatures are in degrees celsius and unless otherwise indicated, various starting materials and reagents are either commercially available or synthesized according to known methods, and are used without further purification, and unless otherwise indicated, commercially available manufacturers include, but are not limited to, the national pharmaceutical community, the carbofuran technologies, the tencel (Shanghai) chemical industry development limited, the Shanghai Pico pharmaceutical technologies limited, the Shanghai Michelson chemical technologies limited, and the like.
CD 3 OD: deuterated methanol
CDCl 3 : deuterated chloroform
DMSO-d 6 : deuterated dimethyl sulfoxide
Pd 2 (dba) 3 : tris (dibenzylideneacetone) dipalladium
Pd(dppf)Cl 2 : [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
XantPhos:4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene
XPhos: 2-dicyclohexylphosphorus-2, 4, 6-triisopropylbiphenyl
HATU:2- (7-Oxybenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate
TLC: thin layer chromatography
HPLC: high performance liquid chromatography
purity: purity of
And (3) the following steps: and
the hydrogen atmosphere is defined as the reaction flask being connected to a hydrogen balloon of about 1L volume.
The examples are not particularly described, and the solution in the reaction is an aqueous solution.
The examples are not specifically described, and the reaction temperature is room temperature and is 20℃to 30 ℃.
The monitoring of the progress of the reaction in the examples employed Thin Layer Chromatography (TLC), the developing reagent used for the reaction, the system of eluent for column chromatography employed for purifying the compound or the developing reagent system of thin layer chromatography included: a: petroleum ether and ethyl acetate systems; b: methylene chloride and methanol systems; c: n-hexane: ethyl acetate; the volume ratio of the solvent is different according to the polarity of the compound, and can be adjusted by adding a small amount of acidic or alkaline reagent, such as acetic acid or triethylamine.
Preparation of intermediates
Intermediate 1
(S) -2- ((tetrahydrofuran-3-yl) oxy) acetic acid ethyl ester IN-1
First step (S) -tetrahydrofuran-3-yl 4-nitrobenzoate IN-1b
(R) -3-hydroxytetrahydrofuran IN-1a (10.0 g,0.113 mol) was dissolved IN tetrahydrofuran (130 mL) under nitrogen, nitrobenzoic acid (18.9 g,0.113 mol) and triphenylphosphine (35.6 g,0.136 mol) were added at room temperature, the temperature was lowered to 0℃and diisopropyl azodicarboxylate (27.8 g,0.137 mol) IN tetrahydrofuran (20 mL) was added dropwise, and the reaction was completed at room temperature by TLC, indicating that the starting material was complete. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-1b (29.8 g, crude product, containing a small amount of phosphorus oxychloride) as a white solid.
Second step (S) -3-hydroxytetrahydrofuran IN-1c
Compound IN-1b (29.8 g, crude) was dissolved IN ethanol (300 mL) and sodium carbonate (25.0 g,0.236 mol) was added and heated to 85℃for 2 hours, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-1c (8.1 g, two-step yield 81%).
Third step (S) -2- ((tetrahydrofuran-3-yl) oxy) acetic acid tert-butyl ester IN-1d
Compound IN-1c (3.0 g,34.05 mmol) was dissolved IN toluene (40 mL), tetrabutylammonium bromide (3.0 g,9.30 mmol) and aqueous sodium hydroxide (30 mL,360mmol,12 mol/L) were added, tert-butyl bromoacetate (9.9 g,50.75 mmol) was added with stirring at room temperature, and stirring overnight at 20℃was complete, and TLC showed complete reaction. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on a silica gel column to give the title compound IN-1d (5.5 g, yield 80%).
1 H NMR(400MHz,CDCl 3 )δ4.22-4.25(m,1H),3.97(s,2H),3.80-3.95(m,4H),1.96-2.09(m,2H),1.48(s,9H).
Fourth step (S) -2- ((tetrahydrofuran-3-yl) oxy) acetic acid IN-1e
Compound IN-1d (5.5 g,27.19 mmol) was dissolved IN dichloromethane (15 mL) and trifluoroacetic acid (30 mL) was added and heated to 45℃for 2 hours, and TLC showed substantial completion. The reaction solution was concentrated (dichloromethane with 3 times) to give the title compound IN-1e (13.5 g, crude) as a pale yellow oil, which was used directly IN the next step.
Fifth step (S) -2- ((tetrahydrofuran-3-yl) oxy) ethyl acetate IN-1
Compound IN-1e (13.5 g, crude) was dissolved IN ethanol (100 mL), concentrated sulfuric acid (30 mL) was added at room temperature, and the mixture was heated to 85℃for reaction for 5 hours. The reaction solution was cooled to room temperature, poured into brine ice, extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on silica gel to give the title compound IN-1 (2.1 g, two-step yield 44%) as a yellowish oil.
1 H NMR(400MHz,CDCl 3 )δ4.29-4.18(m,3H),4.08(s,2H),3.96-3.79(m,4H),2.09-1.96(m,2H),1.29(t,J=7.2Hz,3H).
Intermediate 2
(R) - (2- (5- (1-aminoethyl) thiophen-3-yl) benzyl) (methyl) carbamate IN-2
First step 1- (4-bromo-2-thiophen) ethanone IN-2b
2-Acetylthiophene IN-2a (10.0 g,79.26 mmol) and aluminum trichloride (23.14 g,173.54 mmol) were dispersed IN chloroform (60 mL), and bromine (13.48 g,84.36 mmol) dissolved IN carbon tetrachloride (90 mL) was slowly added dropwise. After the addition, the reaction was completed at room temperature for 4 hours, and TLC monitored the completion of the reaction of the starting material 2-acetylthiophene. The reaction solution was poured into ice water, extracted with ethyl acetate (30 ml. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on a silica gel column to give the title compound IN-2b (12.0 g, yield 74%) as a yellow oil.
1 H NMR(400MHz,CDCl 3 )δ7.58(d,J=1.2Hz,1H),7.58(d,J=1.2Hz,1H),2.54(s,3H).
Second step (R) -N- (1- (4-bromothiophen-2-yl) ethylene) -2-methylpropane-2-sulfinamide IN-2c
To a round bottom flask was added tetraethyltitanate (60 mL), compound IN-2b (6.0 g,29.26 mmol) and (R) - (+) -tert-butylsulfinamide (9.58 g,79.04 mmol) IN sequence at room temperature, and the reaction was slowly warmed to 100deg.C for 1 hour, with TLC monitoring complete reaction of starting materials. The reaction solution was cooled to room temperature, poured into ice water, extracted with ethyl acetate (50 ml. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated, and the crude product was subjected to silica gel column chromatography to give the title compound IN-2c (8.6 g, yield 95%).
Third step (R) -N- ((R) -1- (4-bromothiophen-2-yl) ethyl) -2-methylpropane-2-sulfinamide IN-2d
Compound IN-2c (6.4 g,20.76 mmol) was dissolved IN a mixed solvent of tetrahydrofuran (60 mL) and water (1.2 mL), cooled to-50℃and sodium borohydride (2.23 g,58.95 mmol) was added IN portions, and the reaction was slowly warmed to room temperature for 2 hours, and TLC showed complete reaction of the starting materials. The reaction solution was diluted with ethyl acetate, washed with saturated brine 2 times, washed 2 times with water, dried over anhydrous sodium sulfate, and concentrated, and the crude product was subjected to silica gel column chromatography to give the title compound IN-2d (5.0 g, yield 78%).
Fourth step (R) -1- (4-bromothiophen-2-yl) -1-ethylamine IN-2e
To tetrahydrofuran (50 mL) was added compound IN-2d (5.0 g,16.11 mmol) and concentrated hydrochloric acid (12N, 0.5 mL), and the mixture was stirred for 1 hour at 80℃until the reaction was complete by TLC. The reaction solution was cooled to room temperature, saturated aqueous sodium bicarbonate solution was added to adjust ph=8, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-2e (3.34 g, crude) which was used directly IN the next step.
Fifth step (R) - (1- (4-bromothiophen-2-yl) ethyl) carbamic acid tert-butyl ester IN-2f
Compound IN-2e (12.4 g, crude) and triethylamine (7.3 g,72.14 mmol) were dissolved IN dichloromethane (150 mL), di-tert-butyl dicarbonate (15.7 g,71.94 mmol) was added and stirred overnight at room temperature and TLC checked for complete reaction of starting materials. The reaction solution was washed with water, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2f (15.0 g, 82% IN two steps).
Sixth step (R) - (1- (4- (2-formylphenyl) thiophen-2-yl) ethyl) carbamic acid tert-butyl ester IN-2g
Compound IN-2f (15.0 g,48.98 mmol) and 2-formylphenylboronic acid (8.8 g,58.78 mmol) were dissolved IN 1, 4-dioxane (100 mL) and water (10 mL) under nitrogen, and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (600 mg, 0.730 mmol) and sodium carbonate (10.4 g,98.12 mmol) were added at room temperature and reacted at 90℃for 3 hours, the reaction was essentially complete by TLC. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2g (7.9 g, yield 49%).
Seventh step (R) - (1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) carbamic acid tert-butyl ester IN-2h
Compound IN-2g (7.9 g,23.84 mmol) was dissolved IN methylamine solution (25 mL,30% ethanol solution) and stirred overnight at room temperature, sodium cyanoborohydride (3.0 g,47.74 mmol) was added and stirred at room temperature for 2 hours, and TLC detects that the starting material was essentially complete. The reaction solution was quenched with water, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-2h (7.5 g, crude) which was used directly IN the next step.
LC-MS:m/z=347.2[M+H] +
Eighth step (R) - (2- (5- (1- ((tert-butoxycarbonyl) amino) ethyl) thiophen-3-yl) benzyl) (methyl) carbamic acid benzyl ester IN-2i
Compound IN-2h (7.5 g, crude) was dissolved IN dichloromethane (100 mL) and N, N-diisopropylethylamine (4.2 g,32.50 mmol) and benzyl chloroformate (4.4 g,25.79 mmol) were added sequentially and stirred at room temperature for 2h, and TLC detected complete reaction of starting materials. The reaction solution was washed with water, saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2i (10.6 g, two-step yield 92%).
Ninth step (R) - (2- (5- (1-aminoethyl) thiophen-3-yl) benzyl) (methyl) carbamate IN-2
Compound IN-2i (7.0 g,14.56 mmol) was dissolved IN dichloromethane (5 mL), trifluoroacetic acid (3 mL) was added and stirred at room temperature for 1 hour, and TLC was used to detect complete reaction of starting materials. The reaction solution was diluted with methylene chloride, washed with water, saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2 (3.2 g, yield 58%).
1 H NMR(400MHz,DMSO-d 6 )δ7.64-6.96(m,11H),6.18(br,2H),5.16-4.98(m,2H),4.60-4.42(m,3H),2.78(s,3H),1.50(s,3H).
Intermediate 3
(R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethanamine IN-3
First step 1- (3-nitro-5- (trifluoromethyl) phenyl) ethan-1-one IN-3b
3-bromo-5-nitrobenzotrifluoride IN-3a (2.0 g,7.41 mmol), tributyl (1-ethoxyethylene) tin (3.5 g,9.69 mmol) and Pd (PPh) 3 ) 2 Cl 2 (520 mg,0.74 mmol) was added sequentially to toluene (25 mL), and heated under nitrogen toThe reaction was carried out overnight at 100℃and TLC showed completion. The reaction was cooled to room temperature, hydrochloric acid (15 ml,3 n) was added, stirred for 30 minutes, filtered through celite, the filtrate was separated, the aqueous phase extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-3b (1.25 g, yield 72%) as a yellow oil.
1 H NMR(400MHz,CDCl 3 )δ8.94(s,1H),8.68(s,1H),8.53(s,1H),2.75(s,3H).
Second step (R, Z) -2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethylene) propane-2-sulfinamide IN-3c
A mixture of compound IN-3b (1.25 g,5.36 mmol), (R) - (+) -tert-butylsulfinamide (974 mg,8.04 mmol) and tetraethyl titanate (10 mL,47.70 mmol) was heated to 80℃for 3 hours and the TLC showed little starting material remaining. The reaction solution was cooled to room temperature, poured into ice water (60 mL), extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on silica gel to give the title compound IN-3c (1.01 g, yield 56%) as a yellow oil.
Third step (R) -2-methyl-N- ((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) propane-2-sulfinamide IN-3d
Compound IN-3c (260 mg,0.77 mmol) was dissolved IN tetrahydrofuran (2.5 mL) and water (0.05 mL), cooled to-60℃and sodium borohydride (74 mg,1.95 mmol) was added IN portions, and after addition, stirring was continued for 1 hour at-60℃until TLC showed complete reaction. The reaction solution was quenched with water, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-3d (150 mg, yield 58%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ8.43-8.42(m,2H),7.95(s,1H),4.75-4.69(m,1H),3.55(d,J=4.4Hz,1H),1.61(d,J=6.8Hz,3H),1.25(s,9H).
Fourth step (R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethanamine IN-3
Compound IN-3d (164 mg,0.48 mmol) was dissolved IN tetrahydrofuran (3 mL), concentrated hydrochloric acid (0.5 mL) was added dropwise, and after the addition, the reaction was completed by TLC at room temperature for 1 hour. The reaction was then added dropwise to a saturated aqueous sodium carbonate solution to adjust ph=8, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-3 (105 mg, yield 92%) as a yellow oil.
LC-MS:m/z=235.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.56(s,1H),8.32(s,1H),8.24(s,1H),4.24(q,J=6.8Hz,1H),2.22(br,2H),1.30(d,J=2.8Hz,3H).
Intermediate 4
(R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethan-1-amine IN-4
First step 1-bromo-3-difluoromethyl-2-fluorobenzene IN-4b
2-fluoro-3-bromobenzaldehyde IN-4a (10.0 g,49.26 mmol) was dissolved IN dichloromethane (200 mL), cooled to 0deg.C, diethylaminosulfur trifluoride (15.9 g,98.64 mmol) was slowly added dropwise, and after the dropwise addition, the mixture was slowly warmed to room temperature and stirred for 1 hour, and TLC showed complete reaction of the starting materials. The reaction solution was quenched by pouring into saturated aqueous sodium bicarbonate, extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-4b (8.1 g, yield 73%) as a yellow liquid.
1 H NMR(400MHz,CDCl 3 )δ7.68(t,J=7.2Hz,1H),7.55(t,J=7.2Hz,1H),7.14(t,J=8.0Hz,1H),6.89(t,J=54.8Hz,1H).
Second step 1- (3- (difluoromethyl) -2-fluorophenyl) ethan-1-one IN-4c
Compound IN-4b (8.1 g,36.0 mmol) was dissolved IN 1, 4-dioxane (80 mL), triethylamine (9.1 g,89.9 mmol) and tributyl (1-ethoxyethylene) tin (15.6 g,43.2 mmol) were added sequentially at room temperature, nitrogen was bubbled for 15 min, ditriphenylphosphine palladium dichloride (250 mg,0.36 mmol) was added, nitrogen substitution was repeated, and stirring was carried out at 100℃for 1 hour, and TLC showed complete reaction. The reaction was cooled to room temperature, diluted hydrochloric acid (14.4 mL,72.0mmol, 5M) was added and stirred at room temperature for 1 hour, TLC indicated complete reaction of starting materials. The reaction solution was diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-4c (5.6 g, yield 83%) as a yellow liquid.
1 H NMR(400MHz,CDCl 3 )δ8.00(t,J=7.2Hz,1H),7.79(t,J=6.8Hz,1H),7.34(t,J=7.6Hz,1H),6.94(t,J=54.8Hz,1H),2.67(d,J=5.2Hz,3H).
Third step 1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethan-1-one IN-4d
Potassium nitrate (45.6 g,0.45 mmol) was dissolved IN concentrated sulfuric acid (100 mL), stirred at room temperature for 30 min, cooled to about 0deg.C, and compound IN-4c (8.5 g,45.18 mmol) was slowly added dropwise, stirred at 0deg.C for 5 min, and TLC showed completion of the reaction. The reaction solution was slowly added to ice water, extracted with ethyl acetate, and the organic phases were combined, washed with saturated aqueous sodium bicarbonate, saturated brine, and concentrated, and the crude product was purified by silica gel column to give the title compound IN-4d (9.0 g, yield 86%).
1 H NMR(400MHz,CDCl 3 )δ8.88-8.86(m,1H),8.66-8.64(m,1H),7.12-6.85(m,1H),2.73(d,J=4.8Hz,3H).
Fourth step (R, Z) -N- (1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethylene) -2-methylpropane-2-sulfinamide IN-4e
Compound IN-4d (9.0 g,38.60 mmol) was dispersed IN tetraethyltitanate (90 mL) and (R) - (+) -tert-butylsulfinamide (7.0 g,57.76 mmol) was added at room temperature and slowly warmed to 100deg.C and stirred for 1 hour, TLC indicated complete reaction of starting materials. The reaction solution was cooled to room temperature, poured into vigorously stirred ice water, stirred for 10 minutes, filtered, the filter cake was repeatedly washed with ethyl acetate, the filtrate extracted with ethyl acetate, the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-4e (9.0 g, crude) as a yellow oil, which was used directly IN the next step.
LC-MS:m/z=337.1[M+H] +
Fifth step (R) -N- ((R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethyl) -2-methylpropane-2-sulfinamide IN-4f
Compound IN-4e (9.0 g, crude) was dissolved IN tetrahydrofuran (150 mL) and water (2 mL), cooled to about-60℃and sodium borohydride (3.1 g,81.95 mmol) was added IN portions, after which the reaction was continued at room temperature for 2 hours with slow warming, and TLC indicated that the starting material was consumed. The reaction solution was poured into ice water to quench, extracted with ethyl acetate, and the organic phases were combined, washed with water, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound IN-4f (2.8 g, two-step yield 21%) as a brown oil.
LC-MS:m/z=339.1[M+H] +
Sixth step (R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethan-1-amine IN-4
Compound IN-4f (2.8 g,8.28 mmol) was dissolved IN tetrahydrofuran (30 mL), concentrated hydrochloric acid (1.4 mL,16.52 mmol) was added and stirred at room temperature for 1 hour, TLC showed starting material to be consumed. The reaction solution was cooled to room temperature, saturated aqueous sodium hydrogencarbonate solution was then added to adjust the pH to alkaline, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give a brown liquid, which solidified upon standing to give the title compound IN-4 (1.8 g, yield 95%).
LC-MS:m/z=235.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.76-8.74(m,1H),8.41-8.39(m,1H),7.52-7.25(m,1H),4.42(q,J=6.4Hz,1H),2.48-2.24(m,2H),1.35(d,J=6.8Hz,3H).
Intermediate 5
(R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethanamine IN-5
First step (R, Z) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethylene) -2-methylpropane-2-sulfinamide IN-5a
Compound IN-4c (5.6 g,29.76 mmol) was dissolved IN tetraethyltitanate (50 mL) and (R) - (+) -tert-butylsulfinamide (10.8 g,89.11 mmol) was added at room temperature and slowly warmed to 100deg.C and stirred for 1 hour, TLC indicated complete reaction of starting materials. The reaction solution was cooled to room temperature, poured into vigorously stirred ice water, stirred for 10 minutes, filtered, the filter cake was repeatedly washed with ethyl acetate, the filtrate extracted with ethyl acetate, the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-5a (9.5 g, crude) as a yellow oil, which was used directly IN the next step.
LC-MS:m/z=292.1[M+H] +
Second step (R) -N- ((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methylpropane-2-sulfinamide IN-5b
Compound IN-5a (9.5 g, crude) was dissolved IN tetrahydrofuran (100 mL) and water (2 mL), cooled to about-60℃and sodium borohydride (3.7 mg,97.80 mmol) was added IN portions, after which the reaction was continued at room temperature for 2 hours with slow warming to TLC indicating depletion of the starting material. The reaction solution was quenched by pouring ice water, extracted with ethyl acetate, and the organic phases were combined, washed with water, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-5b (2.9 g, two-step yield 33%) as a yellow oil.
LC-MS:m/z=294.1[M+H] +
Third step (R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethylamine IN-5
Compound IN-5b (2.9 g,9.88 mmol) was dissolved IN tetrahydrofuran (50 mL) and concentrated hydrochloric acid (720 mg,19.8 mmol) was added at room temperature, and the mixture was stirred for 1 hour at 80℃with TLC indicating depletion of the starting material. The reaction solution was cooled to room temperature, saturated aqueous sodium hydrogencarbonate solution was then added to adjust the pH to alkaline, the mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-5 (2.0 g, yield 107%) as a brown liquid.
LC-MS:m/z=190.1[M+H] +
1 H NMR(400MHz,CDCl 3 )δ7.58(t,J=7.4Hz,1H),7.47(t,J=6.8Hz,1H),7.23(t,J=7.6Hz,1H),6.90(t,J=55.2Hz,1H),4.45(q,J=6.8Hz,1H),1.80(s,2H),1.43(d,J=6.4Hz,3H).
Intermediate 6
Benzyl (S) -3- (2-ethoxy-2-oxoethoxy) pyrrolidine-1-carboxylic acid tert-butyl ester IN-6
First step (S) -3-hydroxypyrrolidine-1-carboxylic acid benzyl ester IN-6b
(S) -1-N-Boc-3-hydroxypyrrolidine IN-6a (5.0 g,26.70 mmol) was dissolved IN methanol hydrochloride (40 mL, 4N) and reacted overnight at room temperature. The reaction solution was concentrated, tetrahydrofuran (50 mL) and water (50 mL) were added, sodium carbonate (14.1 g,0.13 mmol) and benzyl chloroformate (6.8 g,40.02 mmol) were added, and the mixture was stirred overnight at room temperature. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-6b (5.9 g, yield 100%) as a pale yellow oil.
Second step (S) -3- (2- (tert-butoxy) -2-oxoethoxy) pyrrolidine-1-carboxylic acid benzyl ester IN-6c
Compound IN-6b (5.9 g,26.67 mmol) was dissolved IN toluene (40 mL), tetrabutylammonium bromide (2.7 g,8.41 mmol) and aqueous sodium hydroxide (24 mL,0.28mol, 12M) were added, tert-butyl bromoacetate (8.2 g,42.08 mmol) was added with stirring at room temperature, and stirring overnight at room temperature, TLC indicated complete reaction. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-6c (10.7 g, crude product) as a pale yellow oil, which was used directly IN the next step.
Third step (S) -2- ((1- ((benzyloxy) carbonyl) pyrrolidin-3-yl) oxy) acetic acid IN-6d
Compound IN-6c (10.7 g, crude) was dissolved IN dichloromethane (100 mL) and trifluoroacetic acid (10 mL) was added and reacted overnight at room temperature, with TLC showing substantial completion. The reaction solution was concentrated (dichloromethane with 3 times) to give the title compound IN-6d (10.1 g, crude) as a pale yellow oil, which was used directly IN the next step.
Fourth step (S) -3- (2-ethoxy-2-oxoethoxy) pyrrolidine-1-carboxylic acid benzyl ester IN-6
Compound IN-6d (10.1 g, crude) was dissolved IN N, N-dimethylformamide (50 mL), and potassium carbonate (23.2 g,0.17 mol) and ethyl iodide (11.9 g,84.06 mmol) were added and reacted at room temperature for 5 hours. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-6 (2.0 g, three-step yield 23%) as a yellow oil.
Intermediate 7
(R) -3- (2-ethoxy-2-oxoethoxy) pyrrolidine-1-carboxylic acid benzyl ester IN-7
First step (R) -3-hydroxypyrrolidine-1-carboxylic acid benzyl ester IN-7b
(R) -1-N-Boc-3-hydroxypyrrolidine IN-7a (10.0 g,53.40 mmol) was dissolved IN methanol hydrochloride (80 mL, 4N) and reacted overnight at room temperature. The reaction solution was concentrated, tetrahydrofuran (100 mL) and water (100 mL) were added, sodium carbonate (28.2 g,0.26 mmol) and benzyl chloroformate (13.6 g,80.04 mmol) were added, and stirred at room temperature overnight. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-7b (9.2 g, yield 78%) as a pale yellow oil.
Second step (R) -3- (2- (tert-butoxy) -2-oxoethoxy) pyrrolidine-1-carboxylic acid benzyl ester IN-7c
Compound IN-7b (9.2 g,41.58 mmol) was dissolved IN toluene (60 mL), tetrabutylammonium bromide (4.0 g,12.47 mmol) and aqueous sodium hydroxide (35 mL,0.41mol, 12M) were added, tert-butyl bromoacetate (12.2 g,62.37 mmol) was added with stirring at room temperature, and stirring overnight at room temperature, TLC indicated complete reaction. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-7c (12.1 g, yield 86%) as a pale yellow oil.
Third step (R) -2- ((1- ((benzyloxy) carbonyl) pyrrolidin-3-yl) oxy) acetic acid IN-7d
Compound IN-7c (6.5 g,19.38 mmol) was dissolved IN dichloromethane (70 mL) and trifluoroacetic acid (7 mL) was added and reacted overnight at room temperature, TLC showed substantial completion. The reaction solution was concentrated (dichloromethane with 3 times) to give the title compound IN-7d (7.2 g, crude) as a pale yellow oil, which was used directly IN the next step.
Fourth step (R) -3- (2-ethoxy-2-oxoethoxy) pyrrolidine-1-carboxylic acid benzyl ester IN-7
Compound IN-7d (7.2 g, crude) was dissolved IN N, N-dimethylformamide (50 mL), and potassium carbonate (16.6 g,0.12 mol) and ethyl iodide (6.0 g,38.80 mmol) were added at room temperature and reacted at room temperature for 5 hours. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-7 (4.2 g, two-step yield 71%) as a yellow oil.
Intermediate 8
(R) -1- (3- (trifluoromethyl) phenyl) ethan-1-amine IN-8
First step (R, Z) -2-methyl-N- (1- (3- (trifluoromethyl) phenyl) ethylene) propane-2-sulfinamide IN-8b
M-trifluoromethyl acetophenone IN-8a (8.0 g,42.52 mmol) and (R) - (+) -tert-butylsulfinamide (9.3 g,76.53 mmol) were dissolved IN isopropyl titanate (60 mL), warmed to 80℃and stirred for 3 hours, and TLC detection was complete. The reaction solution was cooled to room temperature, poured into ice water (100 mL), ethyl acetate was added thereto and stirred, insoluble matters were removed by filtration, the aqueous phase was extracted with ethyl acetate, the organic phase was saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-8b (14.0 g, crude product) which was used directly IN the next step.
Second step (R) -2-methyl-N- ((R) -1- (3- (trifluoromethyl) phenyl) ethyl) propane-2-sulfinamide IN-8c
Compound IN-8b (14.0 g, crude product) was dissolved IN tetrahydrofuran (100 mL) and water (5 mL), cooled to-60℃and sodium borohydride (2.9 g,76.54 mmol) was added IN portions, and the mixture was stirred for 2 hours at room temperature after natural cooling, and the reaction was complete by TLC. The reaction mixture was quenched with water, extracted with ethyl acetate, and the organic phase was saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-8c (7.9 g, two-step yield 63%).
Third step (R) -1- (3- (trifluoromethyl) phenyl) ethan-1-amine IN-8
Compound IN-8c (1.1 g,3.75 mmol) was dissolved IN tetrahydrofuran (20 mL), concentrated hydrochloric acid (0.6 mL, 12N) was added dropwise, and the mixture was stirred at room temperature for 2 hours, and the reaction was complete by TLC. The reaction solution was neutralized with saturated aqueous sodium hydrogencarbonate, extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-8 (700 mg, yield 99%).
Example 1
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 1
First step 6-chloro-N, 2-dimethyl-4-pyrimidinamine 1b
4, 6-dichloro-2-methylpyrimidine 1a (20.0 g,0.123 mol) was added in portions to a methylamine solution (100 mL,30% ethanol solution) and reacted at room temperature for 1 hour, and TLC showed that the starting material was reacted. The reaction solution was concentrated to give the title compound 1b (28.0 g, crude) as a white solid, which was used directly in the next step.
LC-MS:m/z=158.1[M+H] +
Second step 6-chloro-5-iodo-N, 2-methyl-4-pyrimidinamine 1c
Compound 1b (28.0 g, crude) was dissolved in acetic acid (150 mL) and NIS (33.0 g,0.147 mol) was added in portions at room temperature and heated to 70℃for 6 hours, TLC indicated complete reaction of starting materials. The reaction solution was cooled to room temperature, saturated aqueous sodium hydrogencarbonate solution was then added to adjust to neutrality, the organic phases were combined, washed with saturated aqueous sodium hydrogencarbonate solution, saturated aqueous sodium sulfite solution, saturated aqueous brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 1c (37.2 g, crude product containing succinimide) as a white solid, which was used directly in the next step.
LC-MS:m/z=284.0[M+H] +
Third step 6-chloro-N, 2-dimethyl-5-vinyl-4-pyrimidinamine 1d
Compound 1c (15.0 g, crude product) was dissolved in ethylene glycol dimethyl ether (150 mL), and potassium vinyltrifluoroborate (10.6 g,79.13 mmol), potassium phosphate (28.1 g,0.132 mol) and Pd (dppf) Cl were added 2 Dichloromethane complex (400 mg,0.49 mmol), nitrogen displacement 3 times, heating to 80 ℃ and reacting overnight, TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on silica gel to give the title compound 1d (3.87 g, three-step yield 40%) as a white solid.
LC-MS:m/z=184.1[M+H] +
Fourth step 4-chloro-2-methyl-6- (methylamino) pyrimidine-5-carbaldehyde 1e
Compound 1d (3.87 g,21.07 mmol) was dissolved in acetone (40 mL) and water (10 mL), N-methylmorpholine oxide (4.94 g,42.17 mmol) was added, the solution was stirred at room temperature, potassium osmium sulfate aqueous solution (catalytic amount) was added dropwise, the reaction was carried out overnight at room temperature, TLC showed complete reaction of the starting material, sodium periodate (10.2 g,47.69 mmol) was added, and the reaction was carried out at room temperature for 1 hour, TLC showed complete reaction in intermediate state. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on silica gel to give the title compound 1e (3.7 g, yield 94%) as a white solid.
LC-MS:m/z=186.1[M+H] +
Fifth step (S) -4-chloro-2, 8-dimethyl-6- ((tetrahydrofuran-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 1f
Compound 1e (900 mg,4.85 mmol) was dissolved IN tetrahydrofuran (40 mL), intermediate IN-1 (1.1 g,6.31 mmol) was added, cooled to-60℃and lithium diisopropylamide (6.0 mL,12.0mmol, 2.0M) was added dropwise, and the reaction was completed at-60℃for 1 hour, with TLC indicating complete reaction of starting material. The reaction solution was quenched with citric acid, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the crude product, which was slurried with a mixture of petroleum ether/ethyl acetate=3/1, and filtered to give the title compound 1f (560 mg, yield 39%) as a white solid.
LC-MS:m/z=296.1[M+H] +
Sixth step 2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 1g
Compound 1f (95 mg,0.32 mmol) was dissolved IN dimethyl sulfoxide (3 mL), intermediate IN-3 (72 mg,0.31 mmol) and N, N-diisopropylethylamine (125 mg,0.97 mmol) were added at room temperature and heated to 80℃for 16 h, TLC indicated complete reaction of starting materials. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 1g (82 mg, yield 54%) as a yellow solid.
LC-MS:m/z=494.2[M+H] +
Seventh step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 1
1g (82 mg,0.17 mmol) of the compound was dissolved in methanol (5 mL), and palladium on carbon (60 mg, 10%) and concentrated hydrochloric acid (3 drops) were added to react at room temperature under a hydrogen atmosphere for 2 hours. The reaction was filtered through celite, the filtrate was concentrated, and the crude product was purified by prep-TLC to give the title compound 1 (45 mg, yield 58%) as a white solid.
LC-MS:m/z=464.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ7.54(s,1H),6.97-6.93(m,2H),6.79(s,1H),5.55(q,J=7.2Hz,1H),5.12-5.08(m,1H),4.05-3.96(m,3H),3.92-3.87(m,1H),3.71(s,3H),2.43(s,3H),2.34-2.16(m,2H),1.60(d,J=7.2Hz,3H).(99.45%purity by HPLC)
Example 2
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidine
-7 (8H) -one 2
First step 6-chloro-2-methyl-4-pyrimidinamine 2a
4, 6-dichloro-2-methylpyrimidine 1a (15.2 g,93.25 mmol) was added to an aqueous ammonia solution (150 mL, 25%) and the reaction was carried out at 65℃for 3 hours, with TLC indicating completion of the reaction of the starting material. The reaction solution was concentrated to give the title compound 2a (11.9 g, crude) as a white solid, which was used directly in the next step.
LC-MS:m/z=144.1[M+H] +
Second step 6-chloro-5-iodo-2-methyl-4-pyrimidinamine 2b
Compound 2a (11.9 g, crude) was added to acetic acid (120 mL) and N-iodosuccinimide (18.64 g,82.85 mmol) was added in portions at room temperature and heated to 70℃for 4 hours, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, saturated aqueous sodium hydrogencarbonate solution was then added to adjust to neutrality, the organic phases were combined, washed with saturated aqueous sodium hydrogencarbonate solution, aqueous sodium sulfite solution, saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 2b (11.82 g, crude product) as a white solid, which was used directly in the next step.
LC-MS:m/z=270.0[M+H] +
Third step 6-chloro-2-methyl-5-vinyl-4-pyrimidinamine 2c
Compound 2b (18.0 g, crude product) was dissolved in ethylene glycol dimethyl ether (180 mL), and potassium vinyltrifluoroborate (17.89 g,0.13 mol), potassium phosphate (35.9 g,0.17 mol) and Pd (dppf) Cl were added sequentially at room temperature 2 Dichloromethane complex (1.64 g,2.01 mmol), nitrogen displacement 3 times, temperature up to 80 ℃ for 3 hours, TLC indicated complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, the organic phases were combined, saturated brine was washed, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 2c (6.1 g, three-step yield 25%)。
Fourth step 4-amino-6-chloro-2-methylpyrimidine-5-carbaldehyde 2d
Compound 2c (5.00 g,29.48 mmol) was dissolved in acetone and water (120 mL/30 mL) and sodium periodate (14.32 g,66.95 mmol) and aqueous potassium osmium sulfate (catalytic amount) were added and reacted at room temperature for 2 hours, TLC indicated complete reaction of starting materials. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on silica gel to give the title compound 2d (2.5 g, yield 49%) as an off-white solid.
LC-MS:m/z=172.1[M+H] +
Fifth step (S) -4-chloro-2-methyl-6- ((tetrahydrofuran-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 2e
Compound 2d (2.50 g,14.57 mmol) was dissolved IN tetrahydrofuran (500 mL) under nitrogen, intermediate IN-1 (3.30 g,18.94 mmol) was added at room temperature, cooled to-60℃and lithium diisopropylamide (22 mL,43.86mmol, 2M) was added dropwise and reacted at-60℃for 1 hour, slowly warmed to room temperature overnight. The reaction solution was quenched by addition of citric acid, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 2e (1.0 g, yield 24%) as a pale yellow solid.
LC-MS:m/z=282.1[M+H] +
Sixth step 2-methyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 2f
Compound 2e (100 mg,0.35 mmol) was dissolved IN dimethyl sulfoxide (3 mL), intermediate IN-3 (83 mg,0.35 mmol) and N, N-diisopropylethylamine (160 mg,1.24 mmol) were added at room temperature, heated to 100deg.C and reacted overnight, and TLC showed completion of the reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 2f (80 mg, yield 48%) as a yellow solid.
LC-MS:m/z=480.1[M+H] +
Seventh step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 2
Compound 2f (80 mg,0.17 mmol) was dissolved in methanol (5 mL), and palladium on carbon (30 mg, 10%) and concentrated hydrochloric acid (1 drop) were added to react at room temperature under a hydrogen atmosphere for 2 hours. The reaction solution was filtered through celite, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 2 (51 mg, yield 67%) as a white solid.
LC-MS:m/z=450.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ7.57(s,1H),6.96(s,2H),6.80(s,1H),5.55(q,J=7.2Hz,1H),5.12-5.06(m,1H),4.05-3.95(m,3H),3.93-3.85(m,1H),2.39(s,3H),2.34-2.15(m,2H),1.60(d,J=7.2Hz,3H).(98.80%purity by HPLC)
Example 3
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 3
First step 4-chloro-6- ((4-methoxybenzyl) amino) pyrimidine-5-carbaldehyde 3b
4, 6-dichloro-5-pyrimidine-carbaldehyde 3a (15.00 g,84.76 mmol) was dissolved in methylene chloride (200 mL), cooled to 0deg.C, N-diisopropylethylamine (10.90 g,84.34 mmol) was added dropwise, p-methoxybenzylamine (7.54 g,54.96 mmol) was added dropwise after the addition was completed, and the reaction was completed at 0deg.C for 2 hours by TLC. The reaction solution was concentrated, and the crude product was purified by silica gel column chromatography to give a yellow solid crude product, which was slurried with methyl t-butyl ether, filtered, and washed with filter cake methyl t-butyl ether to give the title compound 3b (12.1 g, yield 51%) as a pale yellow solid.
LC-MS:m/z=278.1[M+H] +
Second step (S) -4-chloro-8- (4-methoxybenzyl) -6- ((tetrahydrofuran-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 3c
Compound 3b (2.00 g,7.20 mmol) was dissolved IN tetrahydrofuran (300 mL) under nitrogen, intermediate IN-1 (1.63 g,9.36 mmol) was added at room temperature, cooled to-10deg.C, lithium diisopropylamide (10.8 mL,21.60mmol, 2M) was added dropwise, and the reaction was allowed to proceed slowly to room temperature overnight after completion of the dropwise reaction. The reaction solution was quenched by addition of citric acid, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 3c (900 mg, yield 32%) as a tan solid.
LC-MS:m/z=388.1[M+H] +
Third step 8- (4-methoxybenzyl) -4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 3d
Compound 3c (500 mg,1.29 mmol) was dissolved IN dimethyl sulfoxide (6 mL), intermediate IN-3 (300 mg,1.28 mmol) and N, N-diisopropylethylamine (579 mg,4.48 mmol) were added sequentially at room temperature, heated to 90℃and reacted overnight, and TLC showed completion of the reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 3d (580 mg, yield 77%) as a yellow solid.
LC-MS:m/z=586.2[M+H] +
Fourth step 4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 3e
Compound 3d (580 mg,0.99 mmol) was dissolved in trifluoroacetic acid (10 mL) and heated to 75deg.C for 2 hours, TLC indicated complete reaction of starting material. The reaction solution was cooled to room temperature, saturated aqueous sodium bicarbonate was added dropwise to adjust the pH to alkaline, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 3e (430 mg, crude) as an off-brown solid, which was used directly in the next step.
LCMS:m/z=466.2[M+H] +
Fifth step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 3
Compound 3e (80 mg, crude) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (48 mg,0.86 mmol) and ammonium chloride (45 mg,0.84 mmol) were added at room temperature, and the reaction was heated to 90℃for 3 hours, and TLC showed complete reaction of the starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 3 (42 mg, two-step yield 52%) as a pale yellow solid.
LC-MS:m/z=436.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.20(s,1H),7.61(s,1H),7.05-6.77(m,3H),5.55-5.43(m,1H),5.17-5.09(m,1H),4.07-3.85(m,4H),2.37-2.16(m,2H),1.68-1.55(m,3H).(98.04%purity by HPLC)
Example 4
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 4
First step 8-methyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 4a
Compound 3e (100 mg,0.21 mmol) was dissolved in N, N-dimethylformamide (3 mL), potassium carbonate (89 mg,0.64 mmol) and methyl iodide (61 mg,0.43 mmol) were added and reacted overnight at room temperature, and TLC showed complete reaction of the starting materials. The reaction solution was extracted with water and ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 4a (91 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=480.2[M+H] +
Second step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 4
Compound 4a (91 mg, crude) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (53 mg,0.95 mmol) and ammonium chloride (51 mg,0.95 mmol) were added at room temperature, and the reaction was heated to 90℃for 3 hours, and TLC showed complete reaction of the starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 4 (42 mg, 45% yield in two steps) as a pale yellow solid.
LC-MS:m/z=450.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.31-8.25(m,1H),7.58(s,1H),6.93(s,2H),6.79(s,1H),5.49(q,J=6.8Hz,1H),5.17-5.10(m,1H),4.06-3.86(m,4H),3.77-3.66(m,3H),2.36-2.17(m,2H),1.61(d,J=6.8Hz,3H).(99.14%purity by HPLC)
Example 5
4- (((R) -1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 5
First step 4- (((R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethyl) amino) -8- (4-methoxybenzyl) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 5a
Compound 3c (120 mg,0.31 mmol) was dissolved IN dimethyl sulfoxide (3 mL), intermediate IN-4 (73 mg,0.31 mmol) and N, N-diisopropylethylamine (140 mg,1.08 mmol) were added at room temperature, and the reaction was heated to 100deg.C for 8 hours, TLC indicated that the starting material was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 5a (66 mg, yield 36%) as a yellow solid.
LC-MS:m/z=586.3[M+H] +
Second step 4- (((R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethyl) amino) -6- (((S) -tetrahydrofuran-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 5b
Compound 5a (66 mg,0.11 mmol) was dissolved in trifluoroacetic acid (2 mL) and heated to 75deg.C for 2 hours, TLC indicated that the starting material was complete. The reaction solution was cooled to room temperature, saturated aqueous sodium bicarbonate was added dropwise to adjust the pH to alkaline, the ethyl acetate extracts, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by Prep-TLC to give the title compound 5b as an off-brown solid (27 mg, yield 52%).
LC-MS:m/z=466.2[M+H] +
Third step 4- (((R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethyl) amino) -8-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 5c
Compound 5b (27 mg,0.058 mmol) was dissolved in N, N-dimethylformamide (3 mL), potassium carbonate (16 mg,0.12 mmol) and methyl iodide (25 mg,0.18 mmol) were added at room temperature, and the temperature was raised to 50℃for 1 hour, and TLC showed complete reaction of the starting material. The reaction solution was extracted with water and ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 5c (30 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=480.2[M+H] +
Fourth step 4- (((R) -1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 5
Compound 5c (30 mg, crude product) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (18 mg,0.32 mmol) and ammonium chloride (17 mg,0.32 mmol) were added at room temperature, and the reaction was heated to 90℃for 3 hours, and TLC showed complete reaction of the starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 5 (12 mg, two-step yield 46%) as a pale yellow solid.
LC-MS:m/z=450.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.26(s,1H),7.64(s,1H),7.01-6.83(m,2H),6.76-6.73(m,1H),5.64(q,J=7.2Hz,1H),5.19-5.13(m,1H),4.07-3.98(m,3H),3.94-3.88(m,1H),3.71(s,3H),2.38-2.18(m,2H),1.63(d,J =6.8Hz,3H).(96.10%purity by HPLC)
Example 6
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 6
First step (E) -3- (4-chloro-2-methyl-6- (methylamino) pyrimidin-5-yl) acrylic acid methyl ester 6a
Compound 1c (10.5 g,37.04 mmol) was dissolved in N, N-dimethylformamide (100 mL), triethylamine (8.9 g,87.95 mmol) and palladium acetate (200 mg,0.89 mmol) were added at room temperature, nitrogen was replaced 3 times, methyl acrylate (3.95 g,45.88 mmol) was added by syringe, heated to 100deg.C and reacted overnight, and TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, water was added, extraction was performed with ethyl acetate, the organic phases were combined, saturated brine was washed with water, dried over anhydrous sodium sulfate, concentrated, and the crude product was slurried with a mixture (petroleum ether/ethyl acetate=5/1), filtered, and the cake was washed and dried to give the title compound 6a (6.0 g, crude product) as a earthy yellow solid, which was directly used in the next step.
LC-MS:m/z=242.1[M+H] +
Second step 4-methoxy-2, 8-dimethylpyrido [2,3-d ] pyrimidin-7 (8H) -one 6b
Compound 6a (2.17 g, crude) was added to methanol (30 mL), cooled to 0deg.C, sodium hydride (754 mg,18.85mmol, 60%) was added in portions, and heated to 60deg.C for 6 hours, LCMS indicated complete reaction of starting materials. The reaction solution was cooled to 0 ℃, quenched with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 6b (1.80 g, crude) as a pale yellow solid, which was used directly in the next step.
LC-MS:m/z=206.1[M+H] +
Third step 6-bromo-4-methoxy-2, 8-dimethylpyrido [2,3-d ] pyrimidin-7 (8H) -one 6c
Compound 6b (1.8 g, crude) was dissolved in N, N-dimethylformamide (20 mL), N-bromosuccinimide (1.72 g,9.66 mmol) was added in portions at room temperature, and the reaction was heated to 50deg.C for 2 hours, with TLC indicating complete reaction of the starting materials. The reaction solution was cooled to room temperature, water was then added thereto, stirring was carried out for 20 minutes, filtration was carried out, and methylene chloride was added to dissolve the cake, and dried over anhydrous sodium sulfate, and concentrated to give the title compound 6c (2.2 g, crude product) as a pale yellow solid, which was used directly in the next step.
Fourth step 6-bromo-4-hydroxy-2, 8-dimethylpyrido [2,3-d ] pyrimidin-7 (8H) -one 6d
Compound 6c (2.2 g, crude) was dissolved in hydrochloric acid/1, 4-dioxane solution (20 mL, 4M), heated to 65deg.C and reacted for 2 hours, TLC indicated that the starting material was complete. The reaction was cooled to room temperature, diluted with dichloromethane, stirred for 30 min, filtered, and the filter cake washed with dichloromethane and dried to give the title compound 6d (1.78 g, crude) as a pale yellow solid, which was used directly in the next step.
LC-MS:m/z=272.0[M+H] +
Fifth step 6-bromo-2, 8-dimethyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl 2,4, 6-triisopropylbenzenesulfonate 6e
Compound 6d (1.78 g, crude) was suspended in dichloromethane (20 mL), triethylamine (2.74 g,27.08 mmol) and 4-dimethylaminopyridine (83 mg,0.68 mmol) were added at room temperature, cooled to 0deg.C, 2,4, 6-triisopropylbenzenesulfonyl chloride (2.46 g,8.12 mmol) was added in portions, and the reaction was allowed to proceed to room temperature for 0.5 h, with TLC indicating complete reaction of starting material. The reaction mixture was extracted with water and dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 6e (2.45 g, five-step yield 59%) as a white solid.
LC-MS:m/z=536.0[M+H] +
Sixth step (R) -6-bromo-2, 8-dimethyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 6f
Compound 6e (1.45 g,2.70 mmol) was dissolved IN dimethyl sulfoxide (20 mL), intermediate IN-3 (600 mg,2.56 mmol) and N, N-diisopropylethylamine (1.15 g,8.90 mmol) were added at room temperature and reacted for 3 hours with heating to 80℃and TLC indicated complete reaction of the starting materials. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was subjected to silica gel column chromatography to give the title compound 6f (1.08 g, yield 86%) as a pale yellow solid.
LC-MS:m/z=486.0[M+H] +
Seventh step (R) -5- (2, 8-dimethyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester for 6H
Under nitrogen, 6f (200 mg,0.41 mmol) of the compound 6f and 6g (190 mg,0.61 mmol) of 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester were dissolved in 1, 4-dioxane (15 mL) and water (3 mL), and Pd (dppf) Cl was added at room temperature 2 Dichloromethane complex (35 mg,0.043 mmol) and sodium carbonate (130 mg,1.23 mmol), and stirred for 1 hour at 100℃and TLC detected completion of the reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, the organic phases were combined, saturated brine was washed, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound as a yellow solid for 6h (251 mg, crude product) which was directly used in the next step.
LC-MS:m/z=589.3[M+H] +
Eighth step 3- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 6i
Compound 6h (251 mg, crude) was dissolved in ethyl acetate (10 mL), palladium on carbon (100 mg, 10%) was added at room temperature, the reaction was allowed to proceed to 50 ℃ under hydrogen atmosphere overnight, and LCMS detection was complete. The reaction was cooled to room temperature, filtered through celite, and the filter cake washed with ethyl acetate, and the filtrate was concentrated to give the title compound 6i (260 mg, crude) as a pale yellow oil, which was used directly in the next step.
LC-MS:m/z=561.3[M+H] +
Ninth step 3- (4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 6j
Compound 6i (260 mg,0.43 mmol) was dissolved in tetrahydrofuran (5 mL), acetic anhydride (66 mg,0.65 mmol) and triethylamine (113 mg,0.88 mmol) were added at room temperature, heated to 30deg.C and reacted overnight, and TLC detection was complete. The reaction solution was diluted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 6j (270 mg, crude) as a pale yellow solid, which was used directly in the next step.
LC-MS:m/z=603.3[M+H] +
Tenth step N- (3- ((1R) -1- ((2, 8-dimethyl-7-oxo-6- (piperidin-3-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 6k
Compound 6j (270 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed complete reaction of starting material. The reaction solution was adjusted to be basic by adding saturated aqueous sodium bicarbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 6k (200 mg, crude) as a yellow solid, which was used directly in the next step.
Eleventh step N- (3- ((1R) -1- ((2, 8-dimethyl-6- (1-methylpiperidin-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 6l
Compound 6k (200 mg, crude) was dissolved in methanol (5 mL), aqueous formaldehyde (100 mg,1.23mmol, 37%) and palladium on carbon (70 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, TLC showed complete reaction of starting materials. The reaction solution was filtered, and the filtrate was concentrated to give 6l (190 mg, crude) of the title compound as a yellow oil, which was used directly in the next step.
LC-MS:m/z=517.3[M+H] +
Twelfth step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 6
Compound 6l (190 mg, crude) was dissolved in methanol (6 mL), aqueous sodium hydroxide (2 mL,8.0mmol, 4N) was added at room temperature, heated to 80deg.C and reacted overnight, TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 6 (85 mg, six-step yield 44%) as a white solid.
LC-MS:m/z=475.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.10(s,1H),7.01-6.94(m,2H),6.80(s,1H),5.55(q,J=7.2Hz,1H),3.68(s,3H),3.29-3.15(m,2H),3.12-3.03(m,1H),2.43(s,6H),2.29-2.18(m,2H),2.00-1.75(m,3H),1.61(d,J=7.2Hz,3H),1.58-1.51(m,1H).(99.91%purity by HPLC)
Example 7
4- (((R) -1- (5-amino-2-fluoro-3- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 7
First step 2-fluoro-5-nitro-3- (trifluoromethyl) benzaldehyde 7b
2-fluoro-3-trifluoromethylbenzaldehyde 7a (3 g,15.62 mmol) was dissolved in concentrated sulfuric acid (6 mL) at 0deg.C, nitric acid (2.3 mL, 68%) was added dropwise, and the mixture was slowly warmed to room temperature to react for 5 hours, and TLC monitoring showed that a small amount of starting material remained. Ice water was slowly added to the reaction solution, extraction was performed with ethyl acetate, the organic phases were combined, brine was saturated, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7b (2.2 g, yield 59%).
1 H NMR(400MHz,CDCl 3 )δ10.42(s,1H),8.96-8.94(m,1H),8.77-8.75(m,1H).
Second step (2-fluoro-5-nitro-3- (trifluoromethyl) phenyl) methanol 7c
Compound 7b (940 mg,3.96 mmol) was dissolved in absolute ethanol (15 mL), cooled to 0deg.C, sodium borohydride (226 mg,5.97 mmol) was added in portions, and the reaction was continued at 0deg.C for 35 min after the addition, and TLC showed complete conversion of starting material. The reaction was quenched by dropwise addition of saturated aqueous ammonium chloride, concentrated to remove ethanol, extracted with ethyl acetate in aqueous phase, combined organic phases, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7c (792 mg, yield 84%).
Third step (5-amino-2-fluoro-3- (trifluoromethyl) phenyl) methanol 7d
Compound 7c (792 mg,3.31 mmol) was dissolved in absolute ethanol (18 mL) and palladium on carbon (400 mg, 10%) was added at room temperature overnight under a hydrogen atmosphere and TLC showed complete conversion of starting material. The reaction was diafiltered against celite, the filter cake was washed with ethanol, and the filtrate was concentrated to give the title compound 7d (656 mg, crude) which was used directly in the next step.
LC-MS:m/z=210.1[M+H] +
Fourth step benzyl (4-fluoro-3- (hydroxymethyl) -5- (trifluoromethyl) phenyl) carbamate 7e
Compound 7d (731 mg, crude), potassium carbonate (284 mg,5.24 mmol) and benzyl chloroformate (716 mg,4.20 mmol) were added sequentially to tetrahydrofuran (20 mL) and reacted at room temperature for 3 hours, and TLC showed completion of the reaction. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7e (1.06 g, 84% yield in two steps) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ7.64(t,J=5.6Hz,2H),7.40-7.33(m,5H),6.90(s,1H),5.20(s,2H),4.77(s,2H).
Fifth step (4-fluoro-3-formyl-5- (trifluoromethyl) phenyl) carbamic acid benzyl ester 7f
Compound 7e (1.06 g,3.09 mmol) was dissolved in dichloromethane (20 mL) and manganese dioxide (2.7 g,31.03 mmol) was added and reacted overnight at room temperature, TLC showed completion of the reaction. The reaction solution was filtered with celite, the filter cake was washed with dichloromethane, the filtrate was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7f (934 mg, yield 89%) as a white solid.
Sixth step (benzyl 4-fluoro-3- (1-hydroxyethyl) -5- (trifluoromethyl) phenyl) carbamate 7g
Compound 7f (823mg, 2.42 mmol) was dissolved in anhydrous tetrahydrofuran (18 mL), cooled to 0deg.C, methyl magnesium bromide (2.5 mL,7.5mmol,3M in tetrahydrofuran) was added dropwise, and the mixture was allowed to react overnight at room temperature, and TLC showed complete conversion of starting material. The reaction mixture was quenched by dropwise addition of saturated aqueous ammonium chloride, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound (7 g, 858mg, yield 99%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ7.67-7.65(m,2H),7.41-7.33(m,5H),6.74(s,1H),5.21(s,3H),1.96-1.95(m,1H),1.51(d,J=6.4Hz,3H).
Seventh step (3-acetyl-4-fluoro-5- (trifluoromethyl) phenyl) carbamic acid benzyl ester 7h
7g (858 mg,2.40 mmol) of the compound was dissolved in chloroform (20 mL), manganese dioxide (3.2 g,36.78 mmol) was added and reacted overnight at room temperature, TLC showed incomplete conversion of starting material, heating under reflux for 6 hours, TLC showed complete conversion of starting material. The reaction was cooled to room temperature, filtered with celite, the filter cake washed with dichloromethane, the filtrate concentrated and the crude purified by silica gel column chromatography to give the title compound 7h (680 mg, 80% yield).
1 H NMR(400MHz,CDCl 3 )δ8.17(s,1H),7.83-7.81(m,1H),7.40-7.36(m,5H),6.86(s,1H),5.22(s,2H),2.67(d,J=5.2Hz,3H).
Eighth step (R, E) - (3- (1- ((tert-butylsulfinyl) imino) ethyl) -4-fluoro-5- (trifluoromethyl) phenyl) carbamic acid benzyl ester 7i
Compound 7h (620 mg,1.75 mmol), (R) - (+) -tert-butylsulfinamide (317 mg,2.62 mmol) and tetraethyl titanate (995 mg,4.36 mmol) were added sequentially to tetrahydrofuran (15 mL), heated to 60℃and reacted overnight, TLC indicated complete conversion of starting material. The reaction solution was cooled to room temperature, quenched with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7i (343 mg, yield 43%).
Ninth step (3- ((R) -1- (((R) -tert-butylsulfinyl) amino) ethyl) -4-fluoro-5- (trifluoromethyl) phenyl) carbamic acid benzyl ester 7j
Compound 7i (343 mg,0.75 mmol) was dissolved in tetrahydrofuran (3 mL) and water (2 drops), cooled to-70℃and sodium borohydride (114 mg,3.01 mmol) was added in portions and the reaction was continued for 1 hour. TLC showed no conversion of starting material, additional sodium borohydride (114 mg,3.01 mmol), gradual increase to-45℃for 1 hour, and two new spots were formed (LCMS confirmed two isomers, ratio about 3:1, the desired configuration was the main ratio). The reaction was quenched by dropwise addition of saturated ammonium chloride solution, extraction with ethyl acetate, combining the organic phases, saturated brine, drying over anhydrous sodium sulfate, concentrating, and purifying the crude product by silica gel column chromatography to give the title compound 7j (122 mg, yield 35%).
LC-MS:m/z=461.1[M+H] +
Tenth step (R) - (3- (1-aminoethyl) -4-fluoro-5- (trifluoromethyl) phenyl) carbamic acid benzyl ester 7k
Compound 7j (122 mg,0.26 mmol) was dissolved in tetrahydrofuran (1 mL) and concentrated hydrochloric acid (0.3 mL) was added dropwise at room temperature, and the reaction was continued at room temperature for 40 min after the completion of the conversion of the starting material by TLC. The reaction was quenched by dropwise addition of saturated sodium carbonate solution, extraction with ethyl acetate, combining the organic phases, saturated brine, drying over anhydrous sodium sulfate, concentrating, and purifying the crude product by silica gel column chromatography to give the title compound 7k (92 mg, yield 97%).
Eleventh step (3- ((R) -1- ((2, 8-dimethyl-7-oxo-6- (((S) -tetrahydrofurane-3-yl) oxy) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -4-fluoro-5- (trifluoromethyl) phenyl) carbamic acid benzyl ester 7l
Compound 1f (88 mg,0.30 mmol) was dissolved in N-methylpyrrolidone (5 mL), compound 7k (97 mg,0.27 mmol) and sodium carbonate (86 mg,0.81 mmol) were added at room temperature, and the temperature was raised to 100℃for 5 hours, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 7l (48 mg, yield 28%) as a white solid.
Twelfth step 4- (((R) -1- (5-amino-2-fluoro-3- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 7
Compound 7l (48 mg,0.078 mmol) was dissolved in methanol (5 mL), and palladium on carbon (60 mg, 10%) was added thereto and reacted at room temperature under a hydrogen atmosphere for 2 hours. The reaction solution was filtered through celite, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 7 (14 mg, yield 37%) as a white solid.
LC-MS:m/z=482.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ7.60(s,1H),6.92-6.90(m,1H),6.79-6.77(m,1H),5.66(q,J=7.2Hz,1H),5.15-5.11(m,1H),4.07-3.98(m,3H),3.93-3.88(m,1H),3.70(s,3H),2.38(s,3H),2.36-2.19(m,2H),1.61(d,J=7.2Hz,3H).(94.15%purity by HPLC)
Example 8
N- ((R) -1- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-7-oxo 7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) acetamide 8
First step ((R) -1- (2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) carbamic acid tert-butyl ester 8b
Compound 6f (300 mg,0.62 mmol) was dissolved in toluene (30 mL), and (R) -3-t-butoxycarbonylaminopyrrolidine 8a (172 mg,0.92 mmol), pd was added at room temperature 2 (dba) 3 (56 mg,0.061 mmol), 1 '-binaphthyl-2, 2' -diphenylphosphine (57 mg,0.092 mmol) and sodium tert-butoxide (178 mg,1.85 mmol), nitrogen were replaced 3 times and heated to 100℃overnight, TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 8b (185 mg, yield 51%) as a brown solid.
LC-MS:m/z=592.3[M+H] +
Second step 6- ((R) -3-Aminopyrrolidin-1-yl) -2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 8c
Compound 8b (172 mg,0.29 mmol) was dissolved in methanol hydrochloride (3 mL, 4M) and reacted at room temperature for 1 hour, TLC showed complete reaction of the starting material. The reaction was then added dropwise with saturated aqueous sodium bicarbonate to adjust to basicity, extracted with ethyl acetate, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 8c (160 mg, crude) as a brown solid, which was used directly in the next step.
LC-MS:m/z=492.2[M+H] +
Step three N- ((R) -1- (2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) acetamide 8d
Compound 8c (160 mg, crude) was dissolved in dichloromethane (5 mL), triethylamine (98 mg,0.97 mmol) and acetic anhydride (50 mg,0.49 mmol) were added and reacted at room temperature for 1 hour, TLC showed complete reaction of starting material. The reaction solution was diluted with dichloromethane, washed with hydrochloric acid (1N), saturated brine of the organic phase, dried over anhydrous sodium sulfate, concentrated, and the crude product was slurried with a mixture (dichloromethane/petroleum ether=1/10), filtered, and the filter cake was washed and dried to give the title compound 8d (120 mg, crude product) as a yellow solid, which was directly used in the next step.
LC-MS:m/z=534.2[M+H] +
Fourth step N- ((R) -1- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-7-oxo 7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) acetamide 8
Compound 8d (120 mg, crude) was dissolved in ethyl acetate (3 mL) and methanol (1 mL), concentrated hydrochloric acid (3 drops) and palladium on carbon (80 mg, 10%) were added and reacted at room temperature under a hydrogen atmosphere for 2 hours, and TLC showed that the starting material was complete. The reaction solution was filtered through celite, and the filtrate was adjusted to basic by dropwise addition of methanolic ammonia (1 mL), concentrated, and the crude product was purified by Prep-TLC to give the title compound 8 as a white solid (50 mg, three step yield 34%).
LC-MS:m/z=504.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ6.99(s,1H),6.97(s,1H),6.95(s,1H),6.79(s,1H),5.53(q,J=6.8Hz,1H),4.44-4.38(m,1H),3.75-3.61(m,5H),3.50-3.37(m,2H),2.40(s,3H),2.28-2.19(m,1H),1.95(s,3H),1.93-1.88(m,1H),1.60(d,J=7.2Hz,3H).(98.27%purity by HPLC)
Example 9
N- ((S) -1- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) acetamide 9
First step ((S) -1- (2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) carbamic acid tert-butyl ester 9b
Compound 6f (300 mg,0.62 mmol) was dissolved in toluene (30 mL) and (S) -3-t-butoxycarbonylaminopyrrolidine 9a (172 mg,0.92 mmol), pd was added 2 (dba) 3 (56 mg,0.061 mmol), 1 '-binaphthyl-2, 2' -diphenylphosphine (57 mg,0.092 mmol) and sodium tert-butoxide (178 mg,1.85 mmol), nitrogen were replaced 3 times and heated to 100℃overnight, TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the brown solid title compound 9b (105 mg, yield 29%).
LC-MS:m/z=592.3[M+H] +
Second step 6- ((S) -3-Aminopyrrolidin-1-yl) -2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 9c
Compound 9b (105 mg,0.18 mmol) was dissolved in methanol hydrochloride (3 mL, 4M) and reacted at room temperature for 1 hour, TLC showed complete reaction of starting material. The reaction mixture was then added dropwise with saturated aqueous sodium hydrogencarbonate to adjust to basicity, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 9c (90 mg, crude) as a brown solid, which was used directly in the next step.
Step three N- ((S) -1- (2, 8-dimethyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) acetamide 9d
Compound 9c (90 mg, crude) was dissolved in dichloromethane (3 mL), triethylamine (98 mg,0.97 mmol) and acetic anhydride (50 mg,0.49 mmol) were added and reacted at room temperature for 1 hour, TLC showed complete reaction of starting material. The reaction mixture was diluted with dichloromethane, washed with 1N hydrochloric acid, saturated brine of the organic phase, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by Prep-TLC to give the title compound 9d (71 mg, 75% yield in two steps) as a yellow solid.
LC-MS:m/z=534.2[M+H] +
Fourth step N- ((S) -1- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidin-3-yl) acetamide 9
Compound 9d (71 mg,0.13 mmol) was dissolved in ethyl acetate (3 mL) and methanol (1 mL), concentrated hydrochloric acid (3 drops) and palladium on carbon (80 mg, 10%) were added and reacted at room temperature under a hydrogen atmosphere for 2 hours, and TLC showed complete reaction of the starting materials. The reaction solution was filtered through celite, and the filtrate was adjusted to basic by dropwise addition of methanolic ammonia (1 mL), concentrated, and the crude product was purified by Prep-TLC to give the title compound 9 (30 mg, yield 45%) as a white solid.
LC-MS:m/z=504.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ6.99(s,1H),6.97(s,1H),6.95(s,1H),6.79(s,1H),5.53(q,J=7.2Hz,1H),4.44-4.38(m,1H),3.75-3.61(m,5H),3.50-3.37(m,2H),2.40(s,3H),2.28-2.19(m,1H),1.95(s,3H),1.93-1.88(m,1H),1.60(d,J=6.8Hz,3H).(98.94%purity by HPLC)
Example 10
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (1-methylpiperidin-4-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 10
First step (E) -methyl 3- (4-chloro-6- ((4-methoxybenzyl) amino) pyrimidin-5-yl) acrylate 10a
Compound 3b (7.41 g,26.68 mmol) was dissolved in tetrahydrofuran (100 mL) and methoxyformyl methylene triphenylphosphine (11.60 g,34.69 mol) was added at room temperature and heated to 65℃for 2 hours, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 10a (8.61 g, yield 97%) as a pale yellow oil.
LC-MS:m/z=334.1[M+H] +
Second step 4-methoxy-8- (4-methoxybenzyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 10b
Compound 10a (8.61 g,25.80 mmol) was dissolved in methanol (100 mL), cooled to 0deg.C, sodium hydride (2.27 g,56.75mmol, 60%) was added, and the reaction was allowed to proceed overnight at 65deg.C, TLC indicated complete starting material reaction. The reaction solution was cooled to room temperature, quenched with water, stirred for 0.5 hour, filtered, washed with cake water, dissolved with dichloromethane/methanol, dried over anhydrous sodium sulfate, and concentrated to give the title compound 10b (7.67 g, crude) as a white solid, which was used directly in the next step.
Third step 4-methoxypyrido [2,3-d ] pyrimidin-7 (8H) -one 10c
Compound 10b (7.67 g, crude) was dissolved in trifluoroacetic acid (60 mL) and reacted at 75℃for 3 hours, with TLC indicating completion of the starting material. The reaction solution was concentrated, neutralized with saturated aqueous sodium hydrogencarbonate, added with methylene chloride (the product was insoluble in methylene chloride), stirred for 30 minutes, filtered, and the cake was washed with water and with methylene chloride, and dried to give the title compound 10c (3.90 g, crude product) as a white solid, which was used directly in the next step.
LC-MS:m/z=178.1[M+H] +
Fourth step 4-methoxy-8-methylpyrido [2,3-d ] pyrimidin-7 (8H) -one 10d
Compound 10c (3.90 g, crude) was dissolved in N, N-dimethylformamide (40 mL), potassium carbonate (4.56 g,32.99 mmol) and methyl iodide (6.25 g,44.03 mmol) were added at room temperature, and the reaction was heated to 50deg.C for 1 hour, and TLC showed complete reaction of the starting materials. The reaction solution was cooled to room temperature, water was added thereto, solids were precipitated, filtration was carried out, the cake was washed with water, dichloromethane was added thereto for dissolution, anhydrous sodium sulfate was dried, and the title compound 10d (3.04 g, crude product) was obtained as a white solid by concentration, and was used directly in the next step.
Fifth step 6-bromo-4-hydroxy-8-methylpyrido [2,3-d ] pyrimidin-7 (8H) -one 10e
Compound 10d (2.45 g, crude) was dissolved in N, N-dimethylformamide (25 mL), N-bromosuccinimide (2.74 g,15.39 mmol) was added at room temperature, heated to 100deg.C and reacted overnight, TLC indicated complete reaction of starting materials. The reaction mixture was cooled to room temperature, water was added, solids precipitated, stirred at room temperature for 0.5 h, filtered, the filter cake was washed with water and dried to give a white solid mixture (product and non-demethylated mixture), dissolved in hydrochloric acid/1, 4-dioxane (25 ml,4 n), heated to 70 ℃ and reacted for 2 h, TLC indicated complete reaction of the starting material. The reaction solution was cooled to room temperature, diluted with methanol, stirred for 0.5 hour, filtered, and the filter cake was washed with methanol and dried to give the title compound 10e (2.07 g, crude) as a pale yellow solid, which was used directly in the next step.
Sixth step 6-bromo-4-chloro-8-methylpyrido [2,3-d ] pyrimidin-7 (8H) -one 10f
Compound 10e (2.07 g, crude) was suspended in 1, 4-dioxane (60 mL), N-diisopropylethylamine (4.17 g,32.27 mmol) and phosphorus oxychloride (4.95 g,32.28 mmol) were added at room temperature and reacted for 4 hours with heating to 80℃and TLC indicated complete reaction of the starting materials. The reaction solution was cooled, saturated aqueous sodium hydrogencarbonate solution (Wen Dijia) was then adjusted to be basic, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 10f (1.5 g, 26% yield in five steps) as a pale yellow solid.
LC-MS:m/z=274.0[M+H] +
1 H NMR(400MHz,CDCl 3 )δ8.87(s,1H),8.46(s,1H),3.87(s,3H).
Seventh step (R) -6-bromo-8-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 10g
Compound 10f (750 mg,3.21 mmol) was dissolved IN dimethyl sulfoxide (6 mL), N-diisopropylethylamine (1.24 g,9.59 mmol) and intermediate IN-3 (600 mg,2.56 mmol) were added sequentially at room temperature and heated to 80℃for 4 hours, and TLC showed complete reaction of the starting materials. The reaction solution was cooled to room temperature, water was then added thereto, stirring was carried out for 0.5 hour, filtration was carried out, and after washing the cake with water, it was dissolved with ethyl acetate, washed with hydrochloric acid (1N), washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound (10 g, 1.17g, yield 97%) as a yellow solid.
LCMS:m/z=472.1[M+H] +
Eighth step (R) -8-methyl-6- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) -4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 10i
10g (150 mg,0.32 mmol) of the compound and 10h (106 mg,0.48 mmol) of pinacol 4-methylcyclohex-1-enyl borate were dissolved in 1, 4-dioxane (10 mL) and water (2 mL) and Pd (dppf) Cl was added at room temperature 2 Dichloromethane complex (18 mg,0.022 mmol) and sodium carbonate (148 mg,1.40 mmol), and was heated to 100 ℃ for 1 hour, and TLC detection of reaction was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 10i (120 mg, yield 77%) as a brown oil.
LC-MS:m/z=489.2[M+H] +
Ninth step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (1-methylpiperidin-4-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 14
Compound 10i (120 mg,0.25 mmol) was dissolved in methanol (5 mL), palladium on carbon (50 mg, 10%) was added and the reaction was allowed to proceed to 30℃for 2 hours under hydrogen atmosphere, and completion of the reaction was detected by LCMS. The reaction solution was filtered through celite, the filter cake was washed with methanol, the filtrate was concentrated, the crude product was purified by Prep-TLC to give a yellow solid crude product, dissolved with dilute hydrochloric acid (1N), extracted with ethyl acetate, the organic phase was discarded, aqueous saturated aqueous sodium bicarbonate was adjusted to basic, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 14 (31 mg, yield 27%) as a pale yellow solid.
LC-MS:m/z=461.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.33(s,1H),8.22(s,1H),7.00-6.96(m,1H),6.94(s,1H),6.79(s,1H),5.52(q,J=7.2Hz,1H),3.72-3.68(m,3H),3.65-3.56(m,2H),3.52-3.43(m,1H),3.24-3.08(m,2H),2.98-2.93(m,1H),2.91(s,3H),2.20-2.07(m,3H),1.63(d,J=6.8Hz,3H).(97.21%purity by HPLC)
Example 11
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (1-methylpyrrolidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 11
First step (R) -3- (8-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyridino [2,3-d ] pyrimidin-6-yl) -2, 5-dihydro-1H-pyrrole-1-carboxylic acid tert-butyl ester 11b
Compound 10g (200 mg,0.42 mmol) and 1-tert-butoxycarbonyl-2, 5-dihydro-1H-pyrrole-3-boronic acid pinacol ester 11a (187 mg,0.63 mmol) were dissolved in 1, 4-dioxane (10 mL) and water (2 mL) and Pd (dppf) Cl was added at room temperature 2 Dichloromethane complex (35 mg,0.043 mmol) and sodium carbonate (130 mg,1.23 mmol), nitrogen displacement 3 times, heating to 100 ℃ and reacting for 1 hour, TLC detection reaction was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 11b (200 mg, yield 84%) as a yellow solid.
LC-MS:m/z=561.3[M+H] +
Second step (R) -3- (4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -2, 5-dihydro-1H-pyrrole-1-carboxylic acid tert-butyl ester 11c
Compound 11b (200 mg,0.36 mmol) was dissolved in ethyl acetate (10 mL) and palladium on carbon (50 mg, 10%) was added at room temperature and reacted overnight at 50℃under hydrogen atmosphere with TLC indicating completion of the starting material reaction. The reaction was cooled to room temperature, filtered through celite, and the filter cake washed with ethyl acetate, and the filtrate was concentrated to give the title compound 11c (170 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=529.2[M+H] -
Third step (R) -3- (4- ((1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -2, 5-dihydro-1H-pyrrole-1-carboxylic acid tert-butyl ester 11d
Compound 11c (170 mg, crude) was dissolved in tetrahydrofuran (5 mL), acetic anhydride (124 mg,1.21 mmol) and N, N-diisopropylethylamine (235 mg,1.82 mmol) were added at room temperature, heated to 40℃and reacted overnight, and the reaction was complete by TLC. The reaction solution was cooled to room temperature, diluted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 11d (207 mg, crude) as a brown solid, which was used directly in the next step.
LC-MS:m/z=573.3[M+H] +
Fourth step 3- (4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 11e
Compound 11d (207 mg, crude) was dissolved in methanol (10 mL), palladium on carbon (50 mg, 10%) was added and reacted at 30℃for 2 hours under hydrogen atmosphere, TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give the title compound 11e (200 mg, crude) as a yellow solid, which was used directly in the next step.
Fifth step N- (3- ((1R) -1- ((8-methyl-7-oxo-6- (pyrrolidin-3-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 11f
Compound 11e (200 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed complete reaction of starting material. The reaction solution was adjusted to be basic by adding a saturated aqueous sodium hydrogencarbonate solution, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 11f (187 mg, crude) as a yellow solid, which was used directly in the next step.
Sixth step N- (3- ((1R) -1- ((8-methyl-6- (1-methylpyrrolidin-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 11g
Compound 11f (187 mg, crude) was dissolved in methanol (5 mL), aqueous formaldehyde (100 mg,1.23mmol, 37%) and palladium on carbon (70 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, TLC indicated complete reaction of starting materials. The reaction solution was filtered through celite, the cake was washed with methanol, and the filtrate was concentrated to give the title compound as a yellow oil (11 g, 190mg, crude) which was used directly in the next step.
Seventh step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (1-methylpyrrolidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 11
11g (190 mg, crude) of compound was dissolved in methanol (6 mL), aqueous sodium hydroxide (3 mL, 4N) was added at room temperature, and the reaction was heated to 70℃overnight, with TLC indicating completion of the starting material reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was subjected to Prep-TLC to give the title compound 11 (60 mg, 40% in six steps) as a white solid.
LC-MS:m/z=447.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.34(s,1H),8.28(s,1H),6.96(s,1H),6.93(s,1H),6.79(s,1H),5.50(q,J=6.8Hz,1H),3.79-3.71(m,1H),3.70(s,3H),3.45-3.36(m,2H),3.29-3.24(m,1H),3.23-3.15(m,1H),2.80(s,3H),2.52-2.40(m,1H),2.26-2.15(m,1H),1.62(d,J=7.2Hz,3H).(94.63%purity by HPLC)
Example 12
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- ((1-methylpyrrolidin-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 12
The compound of example 12 was synthesized by reference to the synthesis of example 13.
Example 13
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (((S) -1-methylpyrrolidin-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 13
First step 4-chloro-6- (methylamino) pyrimidine-5-carbaldehyde 13a
4, 6-dichloro-5-pyrimidine-carbaldehyde 3a (52.0 g,0.29 mol) was dissolved in methylene chloride (500 mL), cooled to 0deg.C, triethylamine (59.5 g,0.59 mol) was added dropwise, methylamine hydrochloride (16.9 g,0.25 mol) was added in portions, and the reaction was completed by TLC at 0deg.C for 2 hours. The reaction solution was filtered through celite, the filtrate was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the residue was purified by silica gel column chromatography to give a crude product, which was slurried with methyl t-butyl ether, filtered, and the cake was washed with methyl t-butyl ether to give the title compound 13a (23.5 g, yield 46%) as a pale yellow solid.
LCMS:m/z=172.1[M+H] +
Second step (S) -3- ((4-chloro-8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 13b
Compound 13a (900 mg,5.24 mmol) was dissolved IN tetrahydrofuran (15 mL), intermediate IN-6 (2.1 g,6.81 mmol) was added, cooled to-15℃and lithium diisopropylamide (6.6 mL,13.20mmol, 2.0M) was added dropwise, reacted at-10℃for 5 hours, and reacted overnight at room temperature, where TLC showed complete reaction. The reaction mixture was quenched by dropwise addition of saturated aqueous citric acid, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 13b as a yellow oil (144 mg, yield 7%).
LCMS:m/z=415.1[M+H] +
(S) -3- ((8-methyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 13c
Compound 13b (144 mg,0.35 mmol) was dissolved IN dimethyl sulfoxide (4 mL), intermediate IN-3 (82 mg,0.35 mmol) and N, N-diisopropylethylamine (135 mg,1.04 mmol) were added, the reaction was heated to 90℃overnight and TLC showed complete reaction of the starting materials. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phase was washed with 1N hydrochloric acid, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was subjected to Prep-TLC to give the title compound 13c (194 mg, yield 92%) as a yellow solid.
Fourth step (S) -3- ((4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 13d
Compound 13c (194 mg,0.32 mmol) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (89 mg,1.58 mmol) and amine chloride (86 mg,1.58 mmol) were added at room temperature, and the temperature was raised to 90℃for 5 hours, and TLC showed complete reaction. The reaction solution was cooled to room temperature, filtered through celite, the filtrate was added with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 13d (140 mg, yield 76%).
Fifth step (S) -3- ((4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 13e
Compound 13d (140 mg,0.24 mmol) was dissolved in tetrahydrofuran (5 mL), acetic anhydride (47 mg,0.47 mmol) and N, N-diisopropylethylamine (91 mg,0.70 mmol) were added, and the reaction was allowed to stand overnight at room temperature, and the reaction was complete by TLC. The reaction solution was diluted with ethyl acetate, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 13e (104 mg, yield 69%) as a pale yellow solid.
Sixth step N- (3- ((R) -1- ((8-methyl-7-oxo-6- (((S) -pyrrolidin-3-yl) oxy) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 13f
Compound 13e (104 mg,0.16 mmol) was dissolved in ethanol (5 mL) and palladium on carbon (60 mg, 10%) was added and reacted at room temperature under hydrogen atmosphere for 3 hours, TLC showed completion of the reaction. The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give the title compound 13f (90 mg, crude) as a pale yellow solid, which was used directly in the next step.
Seventh step N- (3- ((R) -1- ((8-methyl-6- (((S) -1-methylpyrrolidin-3-yl) oxy) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 13g
Compound 13f (90 mg, crude) was dissolved in methanol (5 mL), and aqueous formaldehyde (100 mg,1.23mmol, 37%) and palladium on carbon (70 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, and TLC showed completion of the reaction. The reaction solution was filtered through celite, the filter cake was washed with methanol, and the filtrate was concentrated to give the title compound as a pale yellow solid (13 g, 95mg, crude) which was used directly in the next step.
Eighth step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (((S) -1-methylpyrrolidin-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 13
Compound 13g (95 mg, crude) was dissolved in methanol (4 mL), aqueous sodium hydroxide (4 mL,16.00mmol, 4N) was added at room temperature, the reaction was allowed to proceed overnight at 80℃and TLC showed completion of the reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 13 (40 mg, three-step yield 52%) as a pale yellow solid.
LCMS:m/z=463.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.29(s,1H),7.53(s,1H),6.93(s,2H),6.79(s,1H),5.55(t,J=7.2Hz,1H),5.10-5.03(m,1H),3.74(s,3H),3.20-3.13(m,1H),3.01-2.89(m,2H),2.79-2.71(m,1H),2.51(s,3H),2.48-2.38(m,1H),2.17-2.08(m,1H),1.62(d,J=7.2Hz,3H).
Example 14
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (((R) -1-methylpyrrolidin-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 14
First step (R) -3- ((4-chloro-8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 14a
Compound 13a (700 mg,4.08 mmol) was dissolved IN tetrahydrofuran (40 mL), intermediate IN-7 (1.6 g,5.30 mmol) was added, cooled to-15℃and lithium diisopropylamide (5.1 mL,10.20mmol, 2.0M) was added dropwise, the reaction was completed at-10℃for 5 hours, and the reaction was continued at room temperature overnight, with TMC indicating completion of the reaction. The reaction mixture was quenched by dropwise addition of saturated aqueous citric acid, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 14a (176 mg, yield 10%) as a yellow oil.
LCMS:m/z=415.2[M+H] +
Second step (R) -3- ((8-methyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 14b
Compound 14a (176 mg,0.42 mmol) was dissolved IN dimethyl sulfoxide (4 mL), intermediate IN-3 (99 mg,0.42 mmol) and N, N-diisopropylethylamine (164 mg,1.27 mmol) were added at room temperature, the temperature was raised to 90℃and reacted overnight, and TLC showed complete reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phase was washed with 1N hydrochloric acid, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 14b (252 mg, yield 97%) as a brown solid.
LCMS:m/z=613.2[M+H] +
(R) -3- ((4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 14c
Compound 14b (252 mg,0.41 mmol) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (115 mg,2.06 mmol) and amine chloride (111 mg,2.06 mmol) were added at room temperature, and the temperature was raised to 90℃for 5 hours, and TLC showed complete reaction. The reaction solution was cooled to room temperature, filtered through celite, the filtrate was added with water, extracted with ethyl acetate, the phases were combined, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 14c (125 mg, yield 52%) as a tan solid.
LCMS:m/z=583.3[M+H] +
Fourth step (R) -3- ((4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) oxy) pyrrolidine-1-carboxylic acid benzyl ester 14d
Compound 14c (110 mg,0.19 mmol) was dissolved in tetrahydrofuran (5 mL), acetic anhydride (38 mg,0.38 mmol) and N, N-diisopropylethylamine (73 mg,0.57 mmol) were added and reacted overnight at room temperature and the reaction was complete by TLC. The reaction solution was diluted with ethyl acetate, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 14d (80 mg, yield 67%) as a pale yellow solid.
Fifth step N- (3- ((R) -1- ((8-methyl-7-oxo-6- (((R) -pyrrolidin-3-yl) oxy) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 14e
Compound 14d (80 mg,0.13 mmol) was dissolved in ethanol (5 mL) and palladium on carbon (40 mg, 10%) was added and reacted at room temperature under hydrogen atmosphere for 3 hours, TLC showed completion of the reaction. The reaction solution was filtered through celite, the filter cake was washed with methanol, and the filtrate was concentrated to give the title compound 14e (85 mg, crude) as a pale yellow solid, which was used directly in the next step.
Sixth step N- (3- ((R) -1- ((8-methyl-6- (((R) -1-methylpyrrolidin-3-yl) oxy) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 14f
Compound 14e (85 mg, crude) was dissolved in methanol (5 mL), aqueous formaldehyde (85 mg,1.05mmol, 37%) and palladium on carbon (30 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, and TLC showed completion of the reaction. The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give the title compound 14f (90 mg, crude) as a pale yellow solid, which was used directly in the next step.
Seventh step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (((R) -1-methylpyrrolidin-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 14
Compound 14f (90 mg, crude) was dissolved in methanol (4 mL), aqueous sodium hydroxide (4 mL,16.00mmol, 4N) was added, the temperature was raised to 80℃and the reaction was allowed to proceed overnight, and TLC showed completion. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 14 (35 mg, three-step yield 59%) as a pale yellow solid.
LCMS:m/z=463.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.29(s,1H),7.55(s,1H),6.93(s,2H),6.79(s,1H),5.50(q,J=6.8Hz,1H),5.11-5.04(m,1H),3.74(s,3H),3.23-3.16(m,1H),3.06-2.94(m,2H),2.83-2.74(m,1H),2.54(s,3H),2.50-2.38(m,1H),2.17-2.07(m,1H),1.62(d,J=6.8Hz,3H).
Example 15
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 15
First step (R) -5- (8-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 15a
Compound 10g (240 mg,0.51 mmo) and 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester 6g (236 mg,0.76 mmo) were dissolved in 1Pd (dppf) Cl was added to 4-dioxane (10 mL) and water (2 mL) at room temperature 2 Dichloromethane complex (35 mg,0.043 mmol) and sodium carbonate (135 mg,1.27 mmol), nitrogen displacement 3 times, heating to 100 ℃ and reacting for 1 hour, TLC detection reaction was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 15a (260 mg, yield 89%) as a yellow solid.
LC-MS:m/z=575.3[M+H] +
Second step (R) -5- (4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 15b
Compound 15a (260 mg,0.45 mmol) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (126 mg,2.26 mmol) and amine chloride (121 mg,2.26 mmol) were added at room temperature, and the temperature was raised to 90℃for 4 hours, and TLC showed complete reaction of the starting material. The reaction solution was cooled to room temperature, filtered through celite, the filtrate was extracted with water, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 15b (253 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=545.3[M+H] +
Third step (R) -5- (4- ((1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 15c
Compound 15b (253 mg, crude) was dissolved in tetrahydrofuran (5 mL), acetic anhydride (190 mg,1.86 mmol) and N, N-diisopropylethylamine (360 mg,2.79 mmol) were added at room temperature, heated to 40℃and reacted overnight, and TLC detected completion of the reaction. The reaction solution was cooled to room temperature, diluted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 15c (250 mg, crude) as a brown solid, which was used directly in the next step.
Fourth step 3- (4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 15d
Compound 15c (250 mg, crude) was dissolved in methanol (10 mL), palladium on carbon (80 mg, 10%) was added at room temperature, the temperature was raised to 30℃under hydrogen atmosphere and reacted for 2 hours, and TLC showed complete reaction of starting materials. The reaction was cooled to room temperature, filtered through celite, and the filter cake was washed with methanol, and the filtrate was concentrated to give the title compound 15d (240 mg, crude) as a yellow solid, which was used directly in the next step.
Fifth step N- (3- ((1R) -1- ((8-methyl-7-oxo-6- (piperidin-3-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 15e
Compound 15d (240 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed complete reaction of starting material. The reaction solution was adjusted to be basic by adding saturated aqueous sodium bicarbonate, extracted with ethyl acetate, and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound 15e (180 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=489.3[M+H] +
Sixth step N- (3- ((1R) -1- ((8-methyl-6- (1-methylpiperidin-3-yl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 15f
Compound 15e (180 mg, crude) was dissolved in methanol (5 mL), aqueous formaldehyde (100 mg,1.23mmol, 37%) and palladium on carbon (70 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, TLC showed complete reaction of starting materials. The reaction solution was filtered through celite, the filter cake was washed with methanol, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 15f (142 mg, five step yield 63%) as a pale yellow solid.
Seventh step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -8-methyl-6- (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 15
Compound 15f (142 mg,0.28 mmol) was dissolved in methanol (6 mL), aqueous sodium hydroxide (3 mL,12mmol, 4N) was added at room temperature, and the reaction was heated to 70℃overnight, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 15 (80 mg, yield 62%) as a white solid.
LC-MS:m/z=461.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.33(s,1H),8.22(s,1H),6.96(s,1H),6.93(s,1H),6.80(s,1H),5.50(q,J=7.2Hz,1H),3.69(s,3H),3.44-3.37(m,1H),3.30-3.24(m,2H),2.70-2.57(m,5H),2.06-1.97(m,2H),1.91-1.81(m,1H),1.78-1.67(m,1H),1.62(d,J=7.2Hz,3H).(99.76%purity by HPLC)
Example 16
4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 16
First step (R) -6-bromo-4- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methylpyrido [2,3-d ] pyrimidin-7 (8H) -one 16a
Compound 10f (150 mg,0.55 mmol) was dissolved IN dimethyl sulfoxide (3 mL), N-diisopropylethylamine (212 mg,1.64 mmol) and intermediate IN-5 (80 mg,0.42 mmol) were added at room temperature and heated to 80℃for 1 hour, and TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 16a (140 mg, yield 75%) as a yellow solid.
Second step (R) -5- (4- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 16b
Compound 16a (140 mg,0.33 mmol) and 6g (141 mg,0.46 mmol) of 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester were dissolved in 1, 4-dioxane (10 mL) and water (3 mL) and Pd (dppf) Cl was added at room temperature 2 Dichloromethane complex (35 mg,0.043 mmol) and sodium carbonate (97 mg,0.92 mmol) under nitrogen protectionThe temperature was raised to 100℃and stirred for 1 hour, and TLC detected the completion of the reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, the organic phases were combined, saturated brine was washed with water, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 16b (200 mg, crude product) as a brown oil, which was used directly in the next step.
LC-MS:m/z=530.3[M+H] +
Third step 3- (4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 16c
Compound 16b (200 mg, crude) was dissolved in methanol (10 mL), palladium on carbon (50 mg, 10%) was added at room temperature, the temperature was raised to 30℃under hydrogen atmosphere and reacted for 2 hours, and TLC showed complete reaction of the starting material. The reaction was cooled to room temperature, filtered through celite, and the filter cake was washed with methanol, and the filtrate was concentrated to give the title compound 16c (220 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=532.3[M+H] +
Fourth step 4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- (piperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 16d
Compound 16c (220 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed complete reaction of starting material. The reaction solution was adjusted to be basic by adding saturated aqueous sodium bicarbonate, extracted with ethyl acetate, and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound 16d (210 mg, crude) as a yellow solid, which was used directly in the next step.
Fifth step 4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 16
Compound 16d (210 mg, crude) was dissolved in methanol (5 mL), aqueous formaldehyde (125 mg,1.54mmol, 37%) and palladium on carbon (70 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, TLC indicated complete reaction of starting materials. The reaction was filtered, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 16 (70 mg, four step yield 48%) as a pale yellow solid.
LC-MS:m/z=446.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.28(s,1H),8.21(s,1H),7.57(t,J=7.2Hz,1H),7.47(t,J=6.8Hz,1H),7.23(t,J=7.6Hz,1H),7.14-6.86(m,1H),5.78(t,J=7.2Hz,1H),3.68(s,3H),3.30-3.19(m,2H),3.14-3.08(m,1H),2.48(s,3H),2.37-2.25(m,2H),2.04-1.76(m,3H),1.66(d,J=6.8Hz,3H),1.64-1.56(m,1H).(97.17%purity by HPLC)
Example 17
4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((S) -1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 17-1
4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((R) -1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 17-2
First step 3- (2-ethoxy-2-oxoethyl) piperidine-1-carboxylic acid tert-butyl ester 17b
N-Boc-3-piperidineacetic acid 17a (5.0 g,20.55 mmol) was dissolved in N, N-dimethylformamide (50 mL), and potassium carbonate (8.5 g,61.65 mmol) and ethyl iodide (4.8 g,30.82 mmol) were added to react at room temperature for 5 hours. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 17b (5.2 g, yield 93%) as a yellow oil.
Second step 3- (4-chloro-8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 17c
Compound 13a (1.13 g,6.59 mmol) was dissolved in tetrahydrofuran (70 mL), compound 17b (2.32 g,8.56 mmol) was added, cooled to-15℃and lithium diisopropylamide (6.6 mL,13.17mmol, 2.0M) was added dropwise, reacted at-10℃for 5 hours, warmed to room temperature overnight, and TLC showed complete reaction of the starting material. The reaction solution was cooled to 0 ℃, quenched with saturated aqueous citric acid, extracted with ethyl acetate, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the residue was chromatographed on a silica gel column to give a crude petroleum ether, which was slurried and filtered to give the title compound 17c as a yellow solid (840 mg, yield 34%).
LCMS:m/z=323.1[M+H-56] +
Third step (S) -3- (4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 17d-1& (R) -3- (4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 17d-2
Compound 17c (426 mg,1.11 mmol) was dissolved IN dimethyl sulfoxide (4 mL), intermediate IN-5 (200 mg,0.10 mmol) and N, N-diisopropylethylamine (430 mg,3.33 mmol) were added at room temperature, and the reaction was warmed to 100deg.C for 3 hours and TLC showed completion. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phase was washed with 1N hydrochloric acid, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 17d (310 mg, yield 52%) as a yellow solid. Chiral resolution of compound 17d (macrocelluloid AD-H,30×250mm,5 μm,30mL/min, IPA: hexane=5:95) gave compound 17d-1 (peak No. 1, RT 7.95 min) (100 mg, yield 17%) and compound 17d-2 (peak No. 2, RT 11.33 min) (114 mg, yield 20%) as pale yellow solids. The configuration and the properties of the compound need to be further detected, and the tentative 17d-1 and 17d-2 are the configurations.
LCMS:m/z=532.2[M+H]
Fourth step 4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((S) -piperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 17e-1&4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((R) -piperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 17e-2
Compound 17d-1 (100 mg,0.19 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed completion of the reaction. The reaction mixture was then added dropwise with saturated aqueous sodium hydrogencarbonate to adjust to basicity, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 17e-1 (100 mg, crude) as a yellow solid, which was used directly in the next step.
Compound 17d-2 (114 mg,0.21 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed completion of the reaction. The reaction mixture was then added dropwise with saturated aqueous sodium hydrogencarbonate to adjust to basicity, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 17e-2 (85 mg, crude) as a yellow solid, which was used directly in the next step.
The configuration and the properties of the compound need to be further detected, and the tentative 17e-1 and 17e-2 are in the above configurations.
Fifth step 4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((S) -1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 17-1&4- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((R) -1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 17-2
Compound 17e-1 (100 mg, crude) was dissolved in ethanol (5 mL), aqueous formaldehyde (100 mg,1.23mmol, 37%) and palladium on carbon (40 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, and TLC showed completion of the reaction. The reaction solution was filtered through celite, the filter cake was washed with methanol, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 17-1 (55 mg, 65% yield in two steps) as a pale yellow solid.
Compound 17e-2 (85 mg, crude) was dissolved in ethanol (5 mL), aqueous formaldehyde (85 mg,1.05mmol, 37%) and palladium on carbon (40 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, and TLC showed completion of the reaction. The reaction solution was filtered through celite, the cake was washed with methanol, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 17-2 (47 mg, 50% yield in two steps) as a pale yellow solid.
The configuration and the property of the compound need to be further detected, and the tentative 17-1 and 17-2 are in the above configurations.
17-1:
LCMS:m/z=446.2[M+H]+
1 H NMR(400MHz,CD 3 OD)δ8.29(s,1H),8.20(s,1H),7.57(t,J=7.2Hz,1H),7.47(t,J=7.2Hz,1H),7.23(t,J=7.6Hz,1H),6.99(t,J=54.8Hz,1H),5.78(q,J=6.8Hz,1H),3.68(s,3H),3.30-3.19(m,2H),3.14-3.07(m,1H),2.47(s,3H),2.36-2.25(m,2H),2.03-1.77(m,3H),1.66(d,J=7.2Hz,3H),1.65-1.56(m,1H).
17-2:
LCMS:m/z=446.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.29(s,1H),8.19(s,1H),7.57(t,J=7.6Hz,1H),7.47(t,J=7.2Hz,1H),7.23(t,J=7.6Hz,1H),6.99(t,J=54.8Hz,1H),5.78(q,J=6.8Hz,1H),3.69(s,3H),3.30-3.16(m,2H),3.11-3.04(m,1H),2.45(s,3H),2.33-2.20(m,2H),2.03-1.96(m,1H),1.95-1.76(m,2H),1.66(d,J=7.2Hz,3H),1.63-1.55(m,1H).
Example 18
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- ((R) (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 18-1
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2, 8-dimethyl-6- ((S) (1-methylpiperidin-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one 18-2
The compound of example 6 was isolated by chiral resolution (macrocelluloid AD-H,30 x 250mm,5 μm,30mL/min, IPA: hexane=5:95) to give the title compound 18-1, 18-2 as a solid. The configuration and the property of the compound need to be further detected, and the tentative 18-1 and 18-2 are in the above configurations.
18-1:
LC-MS:m/z=475.3[M+H] +
1 H NMR(400MHz,Methanol-d 4 )δ8.13(s,1H),6.96(s,2H),6.80(s,1H),5.55(q,J=7.0Hz,1H),3.68(s,3H),3.27(d,J=21.5Hz,2H),3.20(d,J=11.5Hz,1H),2.57(s,3H),2.45(d,J=13.9Hz,5H),2.03–1.94(m,2H),1.85(d,J=13.1Hz,1H),1.72–1.56(m,4H).
18-2:
LC-MS:m/z=475.3[M+H] +
1 H NMR(400MHz,Methanol-d 4 )δ8.10(s,1H),6.95(d,J=5.5Hz,2H),6.80(s,1H),5.55(d,J=7.1Hz,1H),3.69(s,3H),3.29–3.19(m,2H),3.11(d,J=11.9Hz,1H),2.48(s,3H),2.44(s,3H),2.32(q,J=11.6Hz,2H),1.94(dd,J=19.9,13.4Hz,2H),1.83(t,J=12.8Hz,1H),1.60(d,J=7.1Hz,4H).
Example 19
8-methyl-6- (1-methylpiperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 19
The compound of example 19 was synthesized by reference to the synthesis of example 20.
Example 20
8-methyl-6- ((S) -1-methylpiperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 20-1 8-methyl-6- ((R) -1-methylpiperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 20-2
First step (S) -3- (8-methyl-7-oxo-4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) -7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 20a-1& (R) -3- (8-methyl-7-oxo-4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) -7, 8-dihydropyrido [2,3-d ] pyrimidin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 20a-2
Compound 17c (3411 mg,0.90 mmol) was dissolved IN dimethyl sulfoxide (4 mL), intermediate IN-8 (153 mg,0.81 mmol) and N, N-diisopropylethylamine (348 mg,2.70 mmol) were added at room temperature, and the reaction was warmed to 100deg.C for 3 hours and TLC showed completion. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, washing was performed with dilute hydrochloric acid (1N) and saturated brine, drying was performed with anhydrous sodium sulfate, and concentration was performed, and the crude product was purified by Prep-TLC to give the title compound 20a as a yellow solid (198 mg, yield 46%). Chiral resolution of compound 20a (macrocelluloid AD-H,30 x 250mm,5 μm,30mL/min, IPA: hexane=5:95) gave compound 20a-1 (peak No. 1, RT 8.75 min) (85 mg, 20% yield) and compound 20a-2 (peak No. 2, RT 8.53 min) (94 mg, 22% yield) as pale yellow solids. The configuration and properties of the compounds need to be further detected, and the tentative 20a-1 and 20a-2 are in the above configurations.
LCMS:m/z=532.2[M+H] +
Second step 8-methyl-6- ((S) -piperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 20 b-1&8-methyl-6- ((R) -piperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 20b-2
Compound 20a-1 (85 mg,0.16 mmol) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (2 mL) was added and the reaction was allowed to react at room temperature for 2 hours, and TLC showed that the reaction was complete. The reaction mixture was then added dropwise with saturated aqueous sodium hydrogencarbonate to adjust to basicity, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 20b-1 (100 mg, crude) as a yellow solid, which was used directly in the next step.
Compound 20a-2 (94 mg,0.17 mmol) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, TLC showed completion of the reaction. The reaction mixture was then added dropwise with saturated aqueous sodium hydrogencarbonate to adjust to basicity, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 21b-2 (105 mg, crude) as a yellow solid, which was used directly in the next step.
The configuration and the properties of the compound need to be further detected, and the tentative 20b-1 and 20b-2 are in the above configurations.
Third step 8-methyl-6- ((S) -1-methylpiperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 20-1&8-methyl-6- ((R) -1-methylpiperidin-3-yl) -4- (((R) -1- (3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one 20-2
Compound 20b-1 (100 mg, crude) was dissolved in ethanol (5 mL), and aqueous formaldehyde (100 mg,1.23mmol, 37%) and palladium on carbon (40 mg, 10%) were added and reacted at room temperature under a hydrogen atmosphere for 2 hours, and TLC showed completion of the reaction. The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give the title compound 20-1,20-2 (55 mg, two-step yield 65%) as a pale yellow solid.
Compound 21b-2 (105 mg, crude) was dissolved in ethanol (5 mL), aqueous formaldehyde (105 mg,1.29mmol, 37%) and palladium on carbon (45 mg, 10%) were added and reacted at room temperature under a hydrogen atmosphere for 2 hours, and TLC showed completion of the reaction. The reaction solution was filtered through celite, the filter cake was washed with methanol, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 20-2 (50 mg, 63% yield in two steps) as a pale yellow solid.
The configuration and the property of the compound need to be further detected, and the tentative 20-1 and 20-2 are the configurations.
20-1:
LCMS:m/z=446.2[M+H]+
1 H NMR(400MHz,CD 3 OD)δ8.32(s,1H),8.19(s,1H),7.73-7.66(m,2H),7.55-7.47(m,2H),5.60(q,J=6.8Hz,1H),3.70(s,1H),3.37-3.26(m,1H),3.26-3.19(m,1H),2.59(s,3H),2.56-2.44(m,2H),2.05-1.94(m,2H),1.93-1.79(m,1H),1.76-1.67(m,1H),1.67(d,J=6.8Hz,3H).
20-2:
LCMS:m/z=446.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.32(s,1H),8.19(s,1H),7.76-7.67(m,2H),7.56-7.45(m,2H),5.60(q,J=6.8Hz,1H),3.69(s,3H),3.38-3.26(m,1H),3.26-3.19(m,1H),2.59(s,3H),2.58-2.43(m,2H),2.06-1.94(m,2H),1.92-1.80(m,1H),1.75-1.68(m,1H),1.67(d,J=7.2Hz,3H).
Example 21
4- (((R) -1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -8-methyl-6- ((1-methylpyrrolidin-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 21
Referring to example 14, synthetic example 21.
Example 22
2, 8-dimethyl-4- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 22
First step (benzyl 2- (5- ((R) -1- ((2, 8-dimethyl-7-oxo-6- (((S) -tetrahydrofuranyl-3-yl) oxy) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) thiophen-3-yl) benzyl) (methyl) carbamate 22a
Compound 1f (129 mg,0.44 mmol) was dissolved IN N-methylpyrrolidone (3 mL), intermediate IN-2 (119 mg,0.31 mmol) and triethylamine (126 mg,1.24 mmol) were added at room temperature, and the reaction was warmed to 100℃for 4 hours, and TLC showed complete reaction of starting material. The reaction mixture was diluted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by Prep-TLC to give the title compound 22a (30 mg, yield 15%) as a white solid.
Second step 2, 8-dimethyl-4- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 22
Compound 22a (30 mg,0.047 mmol) was dissolved in methanol (5 mL), palladium on carbon (30 mg, 10%) was added and reacted at room temperature under hydrogen atmosphere for 2 hours, the reaction solution was filtered through celite, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 22 (14 mg, yield 60%) as a white solid.
LC-MS:m/z=506.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ7.51(s,1H),7.48-7.42(m,1H),7.39-7.28(m,3H),7.21(d,J=1.2Hz,1H),7.12(s,1H),5.99(q,J=6.8Hz,1H),5.09-5.06(m,1H),4.03-3.94(m,3H),3.92-3.84(m,3H),3.73(s,3H),2.50(s,3H),2.35(s,3H),2.32-2.14(m,2H),1.77(d,J=7.2Hz,3H).(89.04%purity by HPLC)
Example 23
2-methyl-4- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 23
First step methyl (2- (5- ((R) -1- ((2-methyl-7-oxo-6- (((S) -tetrahydrofuranyl-3-yl) oxy) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) thiophen-3-yl) phenyl) carbamic acid benzyl ester 23a
Compound 2e (221 mg,0.78 mmol) was dissolved IN dimethyl sulfoxide (3 mL), intermediate IN-2 (300 mg,0.79 mmol) and N, N-diisopropylethylamine (356 mg,2.75 mmol) were added at room temperature, heated to 100deg.C and reacted overnight, and TLC showed completion of the reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 23a (140 mg, yield 29%) as a yellow solid.
LC-MS:m/z=626.3[M+H] +
Second step 2-methyl-4- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 23
Compound 23a (140 mg,0.22 mmol) was dissolved in methanol (5 mL), and palladium on carbon (30 mg, 10%) and concentrated hydrochloric acid (2 drops) were added to react at room temperature under a hydrogen atmosphere for 2 hours. The reaction solution was filtered through celite, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 23 (53 mg, yield 49%) as a white solid.
LC-MS:m/z=492.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ7.53(s,1H),7.49-7.44(m,1H),7.39-7.29(m,3H),7.21(s,1H),7.13(s,1H),5.98(q,J=6.8Hz,1H),5.07(s,1H),4.05-3.83(m,6H),2.46(s,3H),2.37(s,3H),2.32-2.13(m,2H),1.77(d,J=6.8Hz,3H).(91.42%purity by HPLC)
Example 24
8-methyl-4- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -6- (((S) -tetrahydrofuranyl-3 yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 24
First step (2- ((methylamino) methyl) phenyl) boronic acid 24a
O-formylboronic acid IN-2j (5.0 g,33.35 mmol) was dissolved IN methylamine ethanol (30 mL, 7M), palladium on charcoal (200 mg, 10%) was added and reacted at room temperature under hydrogen atmosphere for 5 hours, and TLC detected complete reaction of the starting materials. The reaction solution was filtered through celite, the filter cake was washed, and the filtrate was concentrated to give the title compound 24a (5.3 g, crude) as a pale yellow foamy solid, which was used directly in the next step.
Second step (2- (((tert-butoxycarbonyl) (methyl) -14-aza-yl) methyl) phenyl) boronic acid 24b
Compound 24a (5.3 g, crude product) was dissolved in tetrahydrofuran (100 mL) and water (20 mL), sodium carbonate (10.3 g,97.18 mmol) was added at room temperature, cooled to 0deg.C, di-tert-butyl dicarbonate (8.5 g,38.95 mmol) was added dropwise, and after the addition, the reaction was resumed at room temperature overnight, and TLC detection was complete. The reaction mixture was extracted with water, ethyl acetate, dried over anhydrous sodium sulfate, concentrated, slurried with crude petroleum ether and ethyl acetate (5/1), filtered, and the filter cake was washed and dried to give the title compound 24b as a white solid (7.4 g, two-step yield 84%).
1 H NMR(400MHz,DMSO-d 6 )δ8.14(s,2H),7.50(dd,J=7.2,0.8Hz,1H),7.38-7.30(m,1H),7.21(t,J=7.2Hz,1H),7.08(d,J=7.6Hz,1H),4.56(s,2H),2.74(s,3H),1.40(s,9H).
Third step (R) - (2- (5- (1-aminoethyl) thiophen-3-yl) benzyl) (methyl) carbamic acid tert-butyl ester 24c
Compound IN-2e (1.0 g,4.85 mmol) and compound 24b (1.9 g,7.17 mmol) were dissolved IN 1, 4-dioxane (20 mL) and water (5 mL) and added at room temperatureSodium carbonate (1.5 g,14.15 mmol) and Pd (dppf) Cl were added 2 Dichloromethane complex (200 mg,0.24 mmol), nitrogen displacement 3 times, heating to 100 ℃ for 4 hours, TLC detection reaction was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by column chromatography to give the title compound 24c (800 mg, yield 48%) as a red oil.
LC-MS:m/z=347.2[M+H] +
Fourth step (S) -4-chloro-8-methyl-6- ((tetrahydrofuran-3-yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 24d
Compound 13a (12.0 g,6.99 mmol) was dissolved IN tetrahydrofuran (180 mL), intermediate IN-1 (18.3 g,0.11 mol) was added under nitrogen, cooled to-60℃and lithium bis (trimethylsilyl) amide (17.5 mL,0.175mol,1.0M IN tetrahydrofuran) was added dropwise, and the reaction was completed at-10℃for 5 hours, with TLC indicating that the intermediate was complete and a small amount of starting material was not complete. The reaction mixture was quenched by dropwise addition of saturated aqueous citric acid, adjusted to acidity, extracted with ethyl acetate, the organic phases combined, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 24d (7.2 g, yield 36%).
LCMS:m/z=282.1[M+H] +
Fifth step tert-Butylmethyl (2- (5 ((R) -1- ((8-methyl-7-oxo-6- (((S) -tetrahydrofuranyl-3-yl) oxy) -7, 8-dihydropyrido [2,3-d ] pyrimidin-4-yl) amino) ethyl) thiophen-3-yl) benzyl) carbamate 24e
Compound 24d (81 mg,0.29 mmol) was dissolved in dimethyl sulfoxide (3 mL), compound 24c (100 mg,0.29 mmol) and N, N-diisopropylethylamine (111 mg,0.86 mmol) were added at room temperature, and the reaction was warmed to 90℃overnight, TLC showed completion. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 24e (80 mg, yield 47%) as a yellow solid.
LCMS:m/z=592.3[M+H] +
Sixth step 8-methyl-4- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -6- (((S) -tetrahydrofuranyl-3 yl) oxy) pyrido [2,3-d ] pyrimidin-7 (8H) -one 24
Compound 24e (80 mg,0.14 mmol) was dissolved in methanol hydrochloride (3 mL, 4N) and reacted at room temperature for 2 hours, and TLC showed completion of the reaction. The reaction was added dropwise to saturated aqueous sodium bicarbonate to adjust the pH to basicity, extracted with ethyl acetate, the organic phases combined, saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product purified by Prep-TLC to give the title compound 24 (45 mg, 67% yield) as a yellow solid.
LCMS:m/z=492.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.37(s,1H),7.53(s,1H),7.40-7.47(m,1H),7.39-7.26(m,3H),7.21(s,1H),7.13(s,1H),5.94(t,J=6.4Hz,1H),5.15-5.06(m,1H),4.07-3.94(m,3H),3.93-3.85(m,1H),3.83(s,2H),3.74(s,3H),2.32(s,3H),2.30-2.24(m,1H),2.24-2.15(m,1H),1.78(d,J=6.8Hz,3H).(97.81%purity by HPLC)
Test example 1 Compounds for SOS1 inhibitory Activity
The experimental steps are as follows:
1. treatment of the compound: the compound was formulated at 400-fold final concentration, e.g., 5uM for detection, and at 400-fold concentration, i.e., 2mM. The compound was diluted in gradient to the set number of concentration spots with an automated microwell pipette.
2. Transfer compounds to 384 well plate reaction plates: the diluted compounds were transferred from Echo 384 well plates to 384 well reaction plates using an ultrasonic nanoliter liquid treatment system, and both negative and positive controls transferred 50nL of 100% dmso.
3. 4-fold Tag1-SOS1 solution was prepared and transferred: a4-fold Tag1-SOS1 solution was prepared with the reagent provided in the kit (KRAS-G12C/SOS 1 BINGDING ASSAY KIT (Cisbio, cat. No.63 ADK000CB16PEG)), transferred into 5ul to 384 well reaction plates, and for negative control wells, transferred 5ul of reagent instead of enzyme solution, and centrifuged at 1000rpm for 1 min.
4. 4 times Tag2-KRAS G12C solution was prepared: a4-fold Tag2-KRAS G12C solution was prepared with the reagent provided in the kit, transferred to a 5ul 384 well reaction plate, and centrifuged at 1000rpm for 1 minute.
5. Transfer 2-fold detection solution: 2-fold Anti-Tag1-Tb3+ and Anti-Tag2-XL665 solutions were prepared using the Detection Buffer provided in the kit, transferred to 10ul to 384 well reaction plates, centrifuged at 1000rpm for 1 min and incubated at room temperature for 60 min.
6. Reading: the data fluorescence signal values were read with a microplate reader Envision (Ex 665/Em 615).
7. Inhibition calculation and IC50 fitting
Values were copied from the plate reader, where maximum refers to the reading of the positive control and minimum refers to the reading of the negative control. Inhibition ratio (%) = (maximum value-sample value)/(maximum value-minimum value) ×100%.
Data was imported into MS Excel and fitted to IC with XLFIT Excel add-in version5.4.0.8 50 A value; the results are shown in Table 1.
Fitting formula: y=bottom+ (Top-Bottom)/(1+ (IC 50/X)/(HillSlope)
Table 1 inhibitory Activity of Compounds provided in the examples against SOs1
Numbering of compounds IC 50 (nM) Numbering of compounds IC 50 (nM) Numbering of compounds IC 50 (nM)
1 8.5 2 9.7 3 8.0
4 6.5 5 6.1 6 5.9
7 5.3 8 12.0 9 13.0
10 5.0 13 4.6 14 5.7
17-1 5.1 17-2 9.3 18-1 4.9
18-2 11 20-1 8.3 20-2 15
22 8.0 23 5.0 24 5.8
Test example 2 Compounds for IC inhibition of K-562 cell proliferation 50 Measurement
Human chronic myelogenous leukemia cell K-562 (CCL-243) used in the present invention was purchased from American Type Culture Collection (ATCC). Cells in RPMI 1640 medium with 10% Fetal Bovine Serum (FBS) and 1% diabody at 37 ℃,5% co 2 Is grown in the environment of (a).
The inhibition of proliferation of K-562 cells cultured in vitro by the compounds was determined by the following method:
1) Cell inoculation: inoculating K-562 cells in good logarithmic phase into 96-well plate at 20000 cells/well and 90 μl, inoculating at 37deg.C with 5% CO 2 Culturing for 24 hours under the condition.
2) Adding the medicine: the compound to be tested was diluted in a gradient in complete medium and 10 μl of diluted compound was added to 90 μl of cells to give final concentrations of 10000, 3000, 1000, 300, 100, 30, 10, 3, 1nM, with corresponding vehicle controls. Placing at 37deg.C and 5% CO 2 The cells were cultured in a cell incubator for 96 hours.
3) And (3) detection: after adding 10. Mu.L of 5mg/mL MTT working solution (ABCONE, M9609) per well and allowing to act at 37℃for 4 hours, a triple solution (10% SDS,0.5% isopropyl alcohol, 0.1mol/L HCL) was added until the cell lysate was completely dissolved, and OD570 and OD690 values were read using a TECAN SPARK microplate reader.
4) And (3) calculating: the cell growth inhibition was calculated by the following formula:
inhibition = (control wells OD570nm-OD690nm -an administration orifice OD570nm-OD690nm ) Control well OD570nm-OD690nm ×100%
Calculation of IC from compound concentration and corresponding inhibition ratio using Graphpad prism 5.0 software 50 Values. The test results are shown in Table 2.
TABLE 2 IC for the proliferation inhibition of K-562 cells by the compounds of the present invention 50 (nM)
Numbering of compounds K-562 Numbering of compounds K-562
BI-3406 35.2 13 99.9
1 461.7 15 17.0
2 246.9 16 38.3
3 118.3 17-1 28
4 99.9 17-2 148.8
5 75.6 18-1 50.7
6 94.9 18-2 206.8
8 288.8 20-1 29.5
9 326.5 20-2 198.3
10 34.0 22 333
11 37.1 23 274.3
Conclusion: the compounds of the examples of the present invention have proliferation inhibitory effect on K-562 cells, and the activity of a plurality of compounds is equivalent to that of BI-3402.
Test example 3 Effect of Compounds on the level of ERK1/2 phosphorylation of the KRAS downstream Signal molecule in K-562 cells
The effect of the compounds of the invention on ERK1/2 phosphorylation levels in K-562 cells was detected by the following method:
1) Cell inoculation: taking K-562 cells with good logarithmic growth phase at 1×10 6 The cells/wells were inoculated into six well plates and incubated overnight at 37℃under 5% CO 2.
2) Adding the medicine: the compounds to be tested were added to the cells after gradient dilution in complete medium to give final concentrations of 1000, 100, 10, 1nM. Placing at 37deg.C and 5% CO 2 The cells were cultured in a cell incubator for 24 hours.
3) Protein sample preparation: cell suspensions were collected, centrifuged at 500g for 5 min, the supernatant was discarded, washed 3 times with PBS, and 100. Mu.L of cells were lysed with 1 XSDS gel loading buffer (50 mM Tris-HCl (pH 6.8), 100mM DTT,2%SDS,10% glycerol, 0.1% bromophenol blue). Cell lysates were denatured by heating at 100℃for 10 min.
4) Western blot: carrying out SDS-PAGE electrophoresis on a protein sample, transferring the protein to a PVDF membrane by a wet transfer system after the electrophoresis is finished, placing the PVDF membrane in a sealing solution (5% skimmed milk powder is diluted in TBS/T) for sealing for 1 hour at room temperature, and then resisting reaction I and II; after washing the membrane, color development was performed with Immobilon Western HRP Substrate luminal reagent reagent and a Western Blot imager (Tanon, 4600) was used to photograph. The following are the antibody information used: p-ERK1/2 (CST: 4370); ERK1/2 (CST: 9102); beta-tubulin (CST: 2146).
The effect of compounds on ERK1/2 phosphorylation in K-562 cells is shown in FIG. 1.
Conclusion: in the embodiment of the invention, the compound 5 has obvious inhibition effect on the phosphorylation of ERK1/2 of K-562 cells, the inhibition activity is concentration gradient dependent, and the activity is approximately equivalent to that of BI-3402.
Test example 4 Compounds for liver microsomal stability experiments on mice and humans
The experimental steps are as follows:
(1) Taking out liver microsomes (20 mg protein/mL) from a refrigerator at-80 ℃, placing the liver microsomes on a water bath constant temperature oscillator at 37 ℃ for pre-incubation for 3min, and melting the liver microsomes for later use.
(2) According to the above "constitution of the experimental incubation system", a mixed solution of the incubation system (without beta-NADPH) was prepared.
(3) Preparing 100 mu M test compound working solution for later use.
(4) Control group (without β -NADPH): and (3) respectively taking 25 mu L of PB solution into 75 mu L of the mixed solution of the incubation system (2), swirling for 30s, uniformly mixing, and carrying out reaction with the total volume of 100 mu L, and repeating. Incubation was performed in a 37℃water bath thermostatted shaker and timing was started with sampling time points of 0min and 60min.
(5) Sample group: mu.L of beta-NADPH solution (4 mM) was added to 75. Mu.L of the reaction system (2), and the mixture was vortexed for 30s, mixed well, and the total volume of the reaction was 100. Mu.L, and the reaction was repeated. Incubation was performed in a 37℃water bath thermostated shaker and timing was started with sampling times of 0min,5min,15min,30min,60min.
(6) The sample tube was removed at each time point and 300. Mu.L of cold stop reagent (containing internal standard) was added to stop the reaction.
(7) Vortex and centrifuge.
(8) 150 μl of the supernatant was added with 150 μl of water, vortexed and mixed well, and analyzed by LC-MS/MS.
Data analysis: half-life (t) was calculated using the following first order kinetic equation 1/2 ) And Clearance (CL)
C t =C 0 *e -kt
C t =(1/2)*C 0
t 1/2 =ln2/k=0.693/k
CL=V d *k
Vd=1/protein content in liver microsomes
CL int(liver) =CL int(mic) X liver weight to weight ratio x liver microsomal protein concentration per gram of liver
The parameters in the formula are shown in Table 3:
TABLE 3 common parameters of liver and blood in mice, rats, humans
The experimental results are shown in table 4:
liver microsomal stability of the compounds of Table 4 in different species
In the human liver microsome stability experiment, the stability of the compound 2,3,4,13 is obviously better than that of BI-3406, and the stability of the compound 5 is equivalent to that of BI-3406; in the rat liver microsome stability experiment, the stability of the compound 3,5,6,13 is obviously better than that of BI-3406, and the stability of the compound 4 is equivalent to that of BI-3406; in the mouse liver microsome stability experiment, the stability of the compound 2,3,4,5,6,13,18 is significantly better than that of BI-3406. In summary, liver microsomal stability of the various species of the compounds of the examples was significantly better than BI-3406.
CYP enzyme inhibition study of test example 5 Compounds
The experimental steps are as follows:
(1) 100 x specific inhibitors: diluting the corresponding stock solution with 50% acetonitrile-water to prepare inhibitor working solution with corresponding concentration;
(2) 100 x compound: diluting a compound stock solution into 2000 mu M working solution by using pure acetonitrile;
(3) 200 x substrate: diluting the corresponding stock solution with 50% acetonitrile-water to prepare substrate working solution with corresponding concentration;
(4) Preparing a 4mM NADPH solution using the PB solution as a solvent;
(5) Mix4in1 mixed solution: a certain amount of PB was added to the centrifuge tube, a certain amount of MgCl2-PB (6 mM) solution was added, followed by addition of human liver microsome (20 mg/mL) solution, substrate working solution (Phe: 1A2,18mM; bup:2B6,12mM; amo:2C8, 200. Mu.M; tes:3A4,4 mM), vortexing, and split charging was performed at 148. Mu.L/tube.
(6) Mix3in1 mixed solution: a certain amount of PB was taken into the centrifuge tube, a certain amount of MgCl2-PB (6 mM) solution was added, then human liver microsome (20 mg/mL) solution was added, and the substrate working solution (Dic: 2C9,1.6mM; DM:2D6, 800. Mu.M, mid:3A, 400. Mu.M) was vortexed and mixed well, and split charging was performed at 148. Mu.L/tube.
(7) Liver microsome substrate cocktail 2C19: a certain amount of PB was taken into the centrifuge tube, a certain amount of MgCl2-PB (6 mM) solution was added, then human liver microsome (20 mg/mL) solution and substrate working solution (Mep: 2C19,4 mM) were added, vortexed and mixed well, and split charging was performed at 148. Mu.L/tube.
(8) Adding 1 mu LPB into each tube of the to-be-detected object, adding 1 mu L of to-be-detected object working solution/50% acetonitrile-water, adding 2 mu L of to-be-detected object working solution/50% acetonitrile-water into each tube of the inhibitor group, then placing in a 37 ℃ water bath for pre-incubation for 5min, and simultaneously pre-incubating NADPH in the 37 ℃ water bath for 5min;
(9) Add 50. Mu.L/well NADPH working solution, incubate for 30min (S-Mephenytoin), 10min (other substrates);
(10) Adding 600 mu L/hole of ice internal standard working solution, swirling for 5min to stop the reaction, and centrifuging for 10min with 5500 g;
(11) 100. Mu.L of the supernatant was taken and 300. Mu.L of water (phenacetin Ding Hea mol of diaquine) was added, 150. Mu.L of the supernatant was taken and 150. Mu.L of water (other substrate group) was added, and the mixture was vortexed and analyzed by LC-MS/MS.
Data analysis
Calculating the inhibition rate of the test substance and the control substance to the metabolism rate of each specific probe substrate at different concentration levels by Excel, and then performing IC according to the following formula 50 The results are shown in Table 5:
*IC 50 =x (100-enzyme inhibition% at x)/enzyme inhibition, assuming hillslope=1.
x is the concentration of the inhibitor
TABLE 5 inhibitory Activity of example Compounds against CYP enzymes
In general, for each subtype of CYP450, IC 50 >10. Mu. Mol/L may be considered to be weakly inhibited by CYP; 3 mu mol/L<IC 50 <10. Mu. Mol/L can be considered to be moderate inhibition of CYP; IC (integrated circuit) 50 <3. Mu. Mol/L was considered to be strongly inhibited by CYP. BI-3406 can be seen to exhibit moderate inhibitory activity at 2C9,2C19,2D6,3A and like multiple CYP450 subtypes. However, compound 2,3,13,18,23,24 exhibited weak inhibition of the 2C9 subtype; compound 2,3,5,6,13,18,23,24 exhibited weak inhibition of subtype 2C 19; compound 2,3,4,5,6,8,13,18 exhibited weak inhibition of the 2D6 subtype; compound 3,6,13,18,23,24 exhibited weak inhibition of subtype 3A; the tested compounds all showed weak inhibitory activity against 1A2,2B6,2C8. Taken together, it can be seen that the inhibition of CYP450 by multiple compounds is weaker than BI-3406, suggesting that there may be a lower risk of drug interactions and better drug safety.
Test example 6 Compounds for in vivo anti-tumor Activity
MIAPaCa-2 or HPAF-II cells (1.0X10) were injected subcutaneously on the right side of female BALB/c mice (6-8 weeks) 7 Cell/mouse). Mice were dosed by gavage, compound 5 (50 mg/kg, bid), MRTX849 (5 mg/kg, qd), trametinib (0.125 mg/kg, bid), and combinations of compound 5 and Trametinib or MRTX 849. Mice were monitored daily and caliper measurements were started when tumors became visible. The tumor volume was calculated by measuring two perpendicular diameters using the following formula: (L x W2)/2, wherein L and W refer to the length and width of the tumor diameter. When the average tumor volume reaches 100mm 3 At that time, mice were grouped (D0, n=5/group) and given the compound. Tumor volume and mouse body weight were measured 1 time every 3 days during dosing. The results are shown in FIGS. 2-5.
Conclusion: the combination of the compound 5 and the KRAS G12C inhibitor MRTX849 has remarkable synergy on MIAPaCa-2 (KRAS G12C) nude mice transplanted tumor, and the tumor inhibition rate of the single drug group, the MRTX849 (5 mg/kg, qd) single drug group and the two drug combination group of the embodiment 5 (50 mg/kg, bid) is 71.2 percent, 60.8 percent and 98.1 percent (P < 0.001); the combination of the compound 5 and the MEK inhibitor Trametinib has remarkable synergy on HPAF-II (KRAS G12D) nude mice transplantation tumor, and the tumor inhibition rates of the single drug group, the Trametinib (0.125 mg/kg, bid) single drug group and the two drug combination group of the embodiment 5 (50 mg/kg, bid) are 61.3%, 76.2% and 105.3% (P < 0.001). In conclusion, in the mouse model, the activity of the single-use group of the compound 5 of the embodiment of the invention is approximately equivalent to that of BI-3402, and the single-use group of the compound 5 has better anti-tumor activity on KRAS mutant tumor when combined with MRTX849 or Trametinib, and is obviously superior to that of the single-use group.
The applicant states that the present invention is illustrated by the above examples as a pyrimidopyridone derivative as SOS1 inhibitor, and its preparation method and application, but the present invention is not limited to the above examples, i.e. it is not meant that the present invention must be practiced depending on the above examples. It should be apparent to those skilled in the art that any modification of the present invention, equivalent substitution of raw materials for the product of the present invention, addition of auxiliary components, selection of specific modes, etc., falls within the scope of the present invention and the scope of disclosure.
The preferred embodiments of the present invention have been described in detail above, but the present invention is not limited to the specific details of the above embodiments, and various simple modifications can be made to the technical solution of the present invention within the scope of the technical concept of the present invention, and all the simple modifications belong to the protection scope of the present invention.
In addition, the specific features described in the above embodiments may be combined in any suitable manner, and in order to avoid unnecessary repetition, various possible combinations are not described further.

Claims (11)

  1. A pyrimidopyridone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof, characterized in that the pyrimidopyridone derivative has a structure as shown in formula (I):
    wherein: r is R 1 Selected from hydrogen or C 1 -C 3 An alkyl group;
    R 2 selected from hydrogen or C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl are optionally substituted with 1-3R 21 Substituted;
    R 21 selected from C 1 -C 3 Alkyl, hydroxy, halogen, cyano, amino, C 1 -C 3 Alkoxy or = O;
    l may be absent or selected from O, NH or N- (C) 1 -C 3 An alkyl group);
    R 3 selected from H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl are optionally substituted with 1-3R 31 Substituted;
    R 31 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, =o, -NHCOR 32 or-COR 32
    R a Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
    R b selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
    R 32 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl or 4-7 membered heterocyclyl;
    AR is selected from aryl or aryl of 6-10 membered5-to 10-membered heteroaryl, wherein aryl or heteroaryl is optionally substituted with 1-4R 4 Substituted;
    R 4 selected from H, halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy-C 1 -C 3 Alkyl, hydroxy-C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR a 、-NR a R b 6-10 membered aryl or 5-10 membered heteroaryl, wherein 6-10 membered aryl or 5-10 membered heteroaryl is optionally substituted with 1-4R c Substituted;
    R c selected from H, halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy-C 1 -C 3 Alkyl, hydroxy-C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR a 、-NR a R b 、NR a R b -C 1 -C 4 Alkyl, NR a R b -C 1 -C 4 A haloalkyl group; the hetero atom in the heterocyclic group or heteroaryl group in the formula (I) is 1-3 and is selected from one or more of oxygen, nitrogen and sulfur.
  2. The pyrimidopyridone derivative of claim 1, a pharmaceutically acceptable salt thereof, a tautomer thereof, or a stereoisomer thereof, wherein the pyrimidopyridone derivative has a structure represented by formula (ii):
    wherein R is 1 、R 2 、R 3 And R is 4 Having the same limitations as claim 1; n=1 to 4;
    preferably, said compound of formula (II), said phenyl group being optionally substituted with 1 to 4R 4 Substituted when said R 4 When the number of R is 2-4, R is 4 May be the same or different;
    and/or when said R 4 Is C 1 -C 3 In the case of haloalkyl, said R 4 1-3, when two or more R's are present 4 When said R is 4 May be the same or different;
    and/or when said R 4 Is C 1 -C 3 When halogenoalkyl, the halogen atom is fluorine;
    and/or when said R 4 In the case of halogen, R is 4 1-3, when two or more R's are present 4 When said R is 4 May be the same or different;
    and/or when said R 4 When halogen, the halogen atom is fluorine;
    and/or when said R 4 is-NR a R b When said R is 4 1-3, when two or more R's are present 4 When said R is 4 May be the same or different;
    and/or when said R 4 is-NR a R b When said R is a And R is b May be the same or different;
    and/or R 3 H, C of a shape of H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein 4-7 membered heterocyclyl is optionally substituted with 1-3R 31 Substituted;
    and/or when R 3 Is optionally covered with 1-3R 31 Substituted 4-7 membered heterocyclyl, said R 31 When the number of the components is 2-3, R 31 The same or different;
    and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
    and/or, the R 3 Is optionally covered with 1-3R 31 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
    and/or, the R 3 Is optionally covered with 1-3R 31 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
    and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclic group, the R 31 Selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, =o, -NHCOR 32 or-COR 32
    R a Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
    R b selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
    R 32 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl or 4-7 membered heterocyclyl.
  3. The pyrimidopyridone derivative of claim 1, a pharmaceutically acceptable salt thereof, a tautomer thereof, or a stereoisomer thereof, wherein the pyrimidopyridone derivative has a structure represented by formula (III):
    Wherein R is 1 、R 2 、R 3 And R is 4 Having the same limitations as claim 1; n=1 to 4;
    preferably, for compounds of formula (III), the phenyl group is optionally substituted with 1 to 4R 4 Substituted when said R 4 When the number of R is 2-4, R is 4 May be the same or different;
    and/or when said R 4 Is C 1 -C 3 In the case of haloalkyl, said R 4 1-3, when more than two R's are present 4 When said R is 4 May be the same or different;
    and/or when said R 4 Is C 1 -C 3 When halogenoalkyl, the halogen atom is fluorine;
    and/or when said R 4 In the case of halogen, R is 4 1-3, when more than two R's are present 4 When said R is 4 May be the same or different;
    and/or when said R 4 When halogen, the halogen atom is fluorine;
    and/or when said R 4 is-NR a R b When said R is 4 1-3, when more than two R's are present 4 When said R is 4 May be the same or different;
    and/or when said R 4 is-NR a R b When said R is a And R is b May be the same or different;
    and/or R 3 Is H,C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein 4-7 membered heterocyclyl is optionally substituted with 1-3R 31 Substituted;
    and/or when R 3 Is optionally covered with 1-3R 31 Substituted 4-7 membered heterocyclyl, said R 31 When the number of the components is 2-3, R 31 The same or different;
    And/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
    and/or, the R 3 Is optionally covered with 1-3R 31 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
    and/or, the R 3 Is optionally covered with 1-3R 31 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
    and/or, the R 3 Is optionally covered with 1-3R 31 When substituted 4-7 membered heterocyclic group, the R 31 Selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, =o, -NHCOR 32 or-COR 32
    R a Selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
    R b selected from H, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or 3-6 membered cycloalkyl;
    R 32 selected from C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl, 3-6 membered cycloalkyl or 4-7 membered heterocyclyl.
  4. The pyrimidopyridone derivative of claim 1, a pharmaceutically acceptable salt thereof, a tautomer thereof, or a stereoisomer thereof, wherein the pyrimidopyridone derivative has a structure represented by formula (IV):
    wherein R is 1 、R 2 、R 3 、R c And L has the same definition as claim 1; m=1-4;
    Preferably, the compound of formula (IV), L is selected from O, NH or NCH 3
  5. The pyrimidopyridone derivative of claim 1, a pharmaceutically acceptable salt thereof, a tautomer thereof, or a stereoisomer thereof, wherein said pyrimidopyridone derivative is selected from any one of the following structures:
  6. a process for the preparation of a pyrimidopyridone derivative, a stereoisomer thereof, a tautomer thereof or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 5, selected from one of the following two processes:
    in the first method, the compound of the general formula (I-B) and the compound of the general formula (I-A) are subjected to substitution reaction to obtain the compound of the general formula (I):
    wherein X is 1 Halogen, preferably chlorine; r is R 1 、R 2 、R 3 AR and L have the same defined ranges as in claim 1.
    A second method,
    From compoundsAnd compound R 3 Under alkaline condition, W is subjected to Suzuki reaction in the presence of a metal catalyst and a ligand to obtain a compound shown in a general formula (I);
    wherein X is 4 Halogen, preferably bromine; w isR 1 、R 2 、R 3 AR and L have the same defined ranges as in claim 1.
  7. A pharmaceutical composition comprising a pyrimidopyridone derivative of any one of claims 1 to 5, a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof;
    Preferably, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier and/or excipient.
  8. Use of a pyrimido pyridone derivative, a stereoisomer thereof, a tautomer thereof, a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 7 alone or in combination with a KRAS inhibitor or a MEK signaling pathway inhibitor according to any one of claims 1 to 5 for the manufacture of a medicament for the treatment of cancer or for the manufacture of a SOS1 inhibitor; preferably, the cancer includes, but is not limited to, astrocyte cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, laryngeal cancer, pancreatic cancer, lung cancer, oral cancer, ovarian cancer, prostate cancer, thyroid cancer, sarcoma, renal cancer, and cholangiocarcinoma; further preferably, the cancer includes, but is not limited to: pancreatic cancer, colorectal cancer, lung cancer, hepatocellular carcinoma, renal cancer, gastric cancer, and cholangiocarcinoma; and/or the KRAS inhibitor is a KRAS G12C, KRAS G12V, KRAS G12S or KRAS G12D inhibitor; and/or the MEK signaling pathway inhibitor such as a RAF, MEK or ERK1/2 inhibitor.
  9. The use of claim 8, wherein the pyrimidopyridone derivative, stereoisomer thereof, tautomer thereof, pharmaceutically acceptable salt thereof or the pharmaceutical composition is in a dosage range of 10-100mg/kg; and/or the KRAS G12C inhibitor is selected from 3-100mg/kg; and/or the dosage range of the MEK inhibitor is selected from 0.1-0.2mg/kg; preferably, the pyrimidopyridinone derivative, stereoisomer thereof, tautomer thereof, pharmaceutically acceptable salt thereof or the pharmaceutical composition is in a dosage range of 25-50mg/kg; and/or the KRAS G12C inhibitor is in the dosage range of 5-30mg/kg; and/or the dosage range of the MEK inhibitor is selected from 0.125mg/kg; and/or the KRAS G12C inhibitor is MRTX849 and/or the MEK inhibitor is Trametinib.
  10. The use of claim 8, wherein the cancer is a RAS family-related cancer; preferably KRAS, HRAS or NRAS G12C-associated cancer, KRAS, HRAS or NRAS G12D-associated cancer, KRAS, HRAS or NRAS G12V-associated cancer, KRAS, HRAS or NRAS G12S-associated cancer, KRAS, HRAS or NRAS G13D-associated cancer, KRAS, HRAS or NRAS G13C-associated cancer, KRAS, HRAS or NRAS Q61L-associated cancer, KRAS, HRAS or NRAS a 146T-associated cancer, KRAS, HRAS or NRAS a 146V-associated cancer or KRAS, HRAS or NRAS a 146P-associated cancer.
  11. A method for preventing and/or treating cancer comprising administering to a human a therapeutically effective amount of a compound of any one of claims 1-5, or a pharmaceutically acceptable, stereoisomer, tautomer, pharmaceutically acceptable salt thereof, or pharmaceutical composition of claim 7.
CN202180076242.7A 2021-02-09 2021-11-16 Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof Pending CN116669738A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2021101812393 2021-02-09
CN202110181239 2021-02-09
PCT/CN2021/131012 WO2022170802A1 (en) 2021-02-09 2021-11-16 Pyrimido-pyridone derivative as sos1 inhibitor, preparation method therefor and use thereof

Publications (1)

Publication Number Publication Date
CN116669738A true CN116669738A (en) 2023-08-29

Family

ID=82838244

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180076242.7A Pending CN116669738A (en) 2021-02-09 2021-11-16 Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN116669738A (en)
WO (1) WO2022170802A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024074827A1 (en) 2022-10-05 2024-04-11 Sevenless Therapeutics Limited New treatments for pain
CN115819251B (en) * 2022-12-20 2024-04-09 沧州普瑞东方科技有限公司 Preparation method of (1R) -1- [3- (difluoromethyl) -2-fluorophenyl ] ethylamine

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7219218B2 (en) * 2016-12-22 2023-02-07 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Novel benzylamino-substituted quinazolines and derivatives as SOS1 inhibitors
US20220235013A1 (en) * 2017-03-21 2022-07-28 Bayer Pharma Aktiengesellschaft 2-methyl-quinazolines
AU2018390927B2 (en) * 2017-12-21 2023-01-12 Boehringer Ingelheim International Gmbh Novel benzylamino substituted pyridopyrimidinones and derivatives as SOS1 inhibitors
US20220274979A1 (en) * 2018-04-18 2022-09-01 Bayer Pharma Aktiengesellschaft 2-methyl-aza-quinazolines
US20230148450A9 (en) * 2019-03-01 2023-05-11 Revolution Medicines, Inc. Bicyclic heteroaryl compounds and uses thereof
CN113727758A (en) * 2019-03-01 2021-11-30 锐新医药公司 Bicyclic heterocyclic compounds and use thereof
BR112022008858A2 (en) * 2019-11-08 2022-09-06 Revolution Medicines Inc COMPOUND, PHARMACEUTICAL COMPOSITION AND METHODS FOR INHIBITING SOS1 IN A SUBJECT, FOR INHIBITING THE INTERACTION OF SOS1 AND A PROTEIN, TO TREAT OR PREVENT A DISEASE AND TO TREAT OR PREVENT CANCER

Also Published As

Publication number Publication date
WO2022170802A1 (en) 2022-08-18

Similar Documents

Publication Publication Date Title
US10556908B2 (en) Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
JP7038414B2 (en) Positive allosteric modulator of muscarinic acetylcholine receptor M4
RU2685234C1 (en) Condensed bicyclic heteroaromatic derivatives as modulators of tnf activity
RU2684641C1 (en) Pyrazolopyridine derivatives as modulators of tnf activity
JP5752232B2 (en) Substituted pyrrolotriazine compounds as protein kinase inhibitors
KR102650565B1 (en) PDE9 inhibitors and their uses
RU2684644C1 (en) Purine derivatives as tnf activity modulators
CA2932008A1 (en) Fused tricyclic benzimidazoles derivatives as modulators of tnf activity
US20170233395A1 (en) 2,4-disubstituted 7h-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
WO2010101230A1 (en) Quinoxaline compounds
WO2022170952A1 (en) Polycyclic pyridazinone derivative serving as sos1 inhibitor, preparation method therefor and use thereof
CN116669738A (en) Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof
CN113754682B (en) Compound having macrocyclic structure and use thereof
WO2020143763A1 (en) Haloallylamine compounds and application thereof
CA3181790A1 (en) Amidopyrimidone derivatives
TW201910331A (en) Fibroblast growth factor receptor inhibitor and use thereof
US10647727B2 (en) Substituted pyrazolo/imidazolo bicyclic compounds as PDE2 inhibitors
CN116568689A (en) Polycyclic pyrimidine derivative as SOS1 inhibitor, and preparation method and application thereof
EA036289B1 (en) Fused tricyclic imidazo pyrazines as modulators of tnf activity
KR20210105375A (en) Macrocyclic compounds as CDK inhibitors, methods for their preparation, and their use in medicine
CA3019245A1 (en) Fused pentacyclic imidazole derivatives as modulators of tnf activity
WO2023036252A1 (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical use thereof
CN114555597B (en) Isocitrate Dehydrogenase (IDH) inhibitors
EP4284797A1 (en) Pyrazoleamide derivatives
CN116600808A (en) Tetrahydronaphthyridine derivative serving as KRAS mutant G12C inhibitor, 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