WO2020259432A1 - Kras-g12c抑制剂 - Google Patents

Kras-g12c抑制剂 Download PDF

Info

Publication number
WO2020259432A1
WO2020259432A1 PCT/CN2020/097397 CN2020097397W WO2020259432A1 WO 2020259432 A1 WO2020259432 A1 WO 2020259432A1 CN 2020097397 W CN2020097397 W CN 2020097397W WO 2020259432 A1 WO2020259432 A1 WO 2020259432A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
substituted
halogen
compound
cycloalkyl
Prior art date
Application number
PCT/CN2020/097397
Other languages
English (en)
French (fr)
Inventor
谢雨礼
樊后兴
Original Assignee
微境生物医药科技(上海)有限公司
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 微境生物医药科技(上海)有限公司 filed Critical 微境生物医药科技(上海)有限公司
Priority to CN202080016738.0A priority Critical patent/CN113544128B/zh
Publication of WO2020259432A1 publication Critical patent/WO2020259432A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention belongs to the field of medicinal chemistry, and more specifically, to a new type of KRAS-G12C inhibitor, its preparation method and the use method of the compound.
  • RAS represents a group of closely related monomeric globular proteins (21 kDa molecular weight), which have 189 amino acids and are connected to the plasma membrane and bind GDP or GTP. Under normal development or physiological conditions, RAS is activated by receiving growth factors and various other extracellular signals, and is responsible for regulating cell growth, survival, migration and differentiation. RAS acts as a molecular switch. The on/off state of the RAS protein is determined by nucleotide binding. The active signaling conformation is combined with GTP, and the inactive conformation is combined with GDP.
  • RAS When RAS contains bound GDP, it is in a dormant or resting or off state, and is "inactive", in response to exposure to certain growth-promoting stimuli, inducing RAS to convert bound GDP to GTP. As GTP is bound, RAS is "on” and can interact with and activate other proteins (its “downstream targets”).
  • the RAS protein itself has a very low inherent ability to hydrolyze GTP back to GDP and thereby turn itself into a closed state. The conversion of RAS to shutdown requires exogenous proteins called GTPase activating proteins (GAPs), which interact with RAS and greatly promote the conversion of GTP to GDP.
  • GAPs GTPase activating proteins
  • KRAS non-small cell lung cancer
  • KRAS mutations including G12C
  • other known driver oncogenic mutations in NSCLC including EGFR, ALK, ROS1, RET, and BRAF
  • KRAS mutations often co-occur with certain co-mutations, such as STK11, KEAP1, and TP53, which cooperate with mutant RAS to transform cells into highly malignant and aggressive tumor cells.
  • the present invention aims to provide a class of compounds with general structural formula as shown in formula (1), or each of its optical isomers, crystal forms, pharmaceutically acceptable salts, hydrates or solvates:
  • n 0, 1 or 2;
  • A is a bivalent 4-12 membered saturated or partially saturated monocyclic, bicyclic, bridged or spiro ring containing 1-2 N atoms.
  • the monocyclic, bicyclic, bridged or spiro ring may be optionally substituted by one substituted by one or more R 4, when a plurality of substituents R 4, R 4 may be the same or different;
  • Y is a chemical bond or C1-C6 alkylene group
  • R 1 is an aryl group or a heteroaryl group, and the aryl group or heteroaryl group can be substituted by 1-3 of the following groups: halogen, hydroxyl, amino, C1-C3 alkyl, C2-C4 alkenyl, C3 -C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl or halogen substituted C1-C3 alkoxy;
  • R 2 is an aminoalkyl group, a cycloalkyl group, an alkyl substituted amido group, a heterocyclic group, an aryl group or a heteroaryl group, and the heterocyclic group, aryl group or heteroaryl group may be optionally substituted by one or more R 5 , when it is substituted by multiple R 5 , R 5 may be the same or different;
  • R 3 independently selects C1-C3 alkyl or halo C1-C3 alkyl
  • R 4 is independently selected from H, CN, C1-C3 alkyl, halogen substituted C1-C3 alkyl, or cyano substituted C1-C3 alkyl;
  • R 5 is independently selected from halogen, H, O, CN, OH, alkylhydroxyl, dialkylamino, C1-C6 alkyl, C3-C6 cycloalkyl, halogen-substituted C1-C3 alkyl, or halogen-substituted C1- C3 alkoxy;
  • E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of the K-Ras, H-Ras or N-Ras mutant protein.
  • E is a group containing an electrophilic carbon-carbon double bond or a carbon-carbon triple bond.
  • E is: Wherein, R a is H or F, R b is H, -CH 2 F, -CHF 2 ,
  • AE is: Wherein, n is 1 or 2, and R 4 is H, CN, C1-C3 alkyl, halogen substituted C1-C3 alkyl, or cyano substituted C1-C3 alkyl.
  • Y is a chemical bond, -CH 2 -, -CH(Me)- or -CH 2 CH 2 -.
  • R 1 is: Wherein R c and R d are independently halogen, hydroxyl, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl or halogen Substituted C1-C3 alkoxy.
  • R 2 is: Where n is 1 or 2, R e and R f are independently H, halogen, hydroxyl, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen Substituted C1-C3 alkyl or halogen substituted C1-C3 alkoxy, R g is C1-C3 alkyl, C3-C6 cycloalkyl, C3-C6 cycloalkylalkyl, C1-C3 alkoxyalkyl, Halogen substituted C1-C3 alkyl, halogen substituted C3-C6 cycloalkyl or
  • Another aspect of the present invention aims to provide a class of compounds whose general structural formula is represented by formula (2), or each of its optical isomers, crystal forms, pharmaceutically acceptable salts, hydrates or solvates :
  • A is a bivalent 4-12 membered saturated or partially saturated monocyclic, bicyclic, bridged or spiro ring containing 1-2 N atoms.
  • the monocyclic, bicyclic, bridged or spiro ring may be optionally substituted by one substituted by one or more R 4, when a plurality of substituents R 4, R 4 may be the same or different;
  • Y is a chemical bond or C1-C6 alkylene group
  • W is N, CR 8 or COR 6 :
  • R 2 is aminoalkyl, cycloalkyl, alkyl-substituted amido, heterocyclic, aryl or heteroaryl, the heterocyclic, aryl or heteroaryl group It may be optionally substituted by one or more R 5 , when it is substituted by more R 5 , R 5 may be the same or different;
  • R 2 is an aminoalkyl, alkyl substituted amido or heterocyclic group, and the heterocyclic group may be optionally substituted by one or more R 5 , when When one R 5 is substituted, R 5 may be the same or different;
  • R 1 is an aryl group or a heteroaryl group, and the aryl group or heteroaryl group can be substituted by 1-3 of the following groups: halogen, hydroxyl, amino, C1-C3 alkyl, C2-C4 alkenyl, C3 -C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl, halogen substituted C1-C3 alkoxy;
  • R 4 is independently selected from H, CN, C1-C3 alkyl, halogen substituted C1-C3 alkyl, or cyano substituted C1-C3 alkyl;
  • R 5 is independently selected from halogen, H, O, CN, OH, alkylhydroxyl, dialkylamino, C1-C6 alkyl, C3-C6 cycloalkyl, halogen substituted C1-C3 alkyl, or cyano substituted C1 -C3 alkyl;
  • R 6 is C1-C3 alkyl, halogen-substituted C1-C3 alkyl or C3-C6 cycloalkyl;
  • R 7 is H, halogen, C1-C3 alkyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1-C3 alkyl, halogen substituted C1-C3 alkoxy or C2-C4 alkenyl;
  • R 8 is H, halogen, C1-C3 alkyl, C3-C6 cycloalkyl, or halogen substituted C1-C3 alkyl;
  • E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of the K-Ras, H-Ras or N-Ras mutant protein.
  • E is a group containing an electrophilic carbon-carbon double bond or a carbon-carbon triple bond.
  • E is: Wherein, R a is H or F, R b is H, -CH 2 F, -CHF 2 ,
  • AE is: Wherein, n is 1 or 2, wherein R 4 is H, CN, C1-C3 alkyl, halogen substituted C1-C3 alkyl, or cyano substituted C1-C3 alkyl.
  • Y is a chemical bond, -CH 2 -, -CH(Me)- or -CH 2 -CH 2 -.
  • R 1 is: Wherein R c and R d are independently halogen, hydroxy, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen-substituted C1-C3 alkyl or halogen Substituted C1-C3 alkoxy.
  • W is COR 6 and R 2 is:
  • R e and R f are independently halogen, hydroxy, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1 -C3 alkyl or halogen substituted C1-C3 alkoxy
  • R g is C1-C3 alkyl, C3-C6 cycloalkyl, C3-C6 cycloalkylalkyl, C1-C3 alkoxyalkyl, halogen Substituted C1-C3 alkyl, halogen substituted C3-C6 cycloalkyl or
  • W is N or CR 8
  • R 2 is: Where n is 1 or 2, R e and R f are independently halogen, hydroxy, amino, C1-C3 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C1-C3 alkoxy, halogen substituted C1 -C3 alkyl or halogen substituted C1-C3 alkoxy, R g is C1-C3 alkyl, C3-C6 cycloalkyl, C3-C6 cycloalkylalkyl, C1-C3 alkoxyalkyl, halogen substituted C1-C3 alkyl, halogen substituted C3-C6 cycloalkyl or
  • the compound has one of the structures listed in Table 1 below:
  • Another object of the present invention is to provide a pharmaceutical composition, which contains a pharmacologically acceptable excipient or carrier, and the compound of the general formula (1) or general formula (2) of the present invention, or their respective optical differences Constructs, pharmaceutically acceptable inorganic or organic salts are used as active ingredients.
  • Another object of the present invention is to provide the use of the above-mentioned compound of the present invention, or each of its optical isomers, or pharmaceutically acceptable inorganic or organic salts, for preparing and treating tumor-related diseases.
  • the compounds of the general formula (1) and (2) described above can be synthesized using standard synthesis techniques or well-known techniques and methods combined in the text. In addition, the solvent, temperature and other reaction conditions mentioned here can be changed.
  • the starting materials for the synthesis of the compound can be synthesized or obtained from commercial sources such as, but not limited to, Aldrich Chemical Co. (Milwaukee, Wis.) or Sigma Chemical Co. (St. Louis, Mo.).
  • the compounds described herein and other related compounds with different substituents can be synthesized using well-known techniques and raw materials, including those found in March, ADVANCED ORGANIC CHEMISTRY 4 th Ed., (Wiley 1992); Carey and Sundberg, ADVANCED ORGANIC CHEMISTRY 4 th Ed., Vols. A and B (Plenum 2000, 2001), Green and Wuts, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS 3 rd Ed., (Wiley 1999).
  • the general method of compound preparation can be changed by using appropriate reagents and conditions for introducing different groups in the molecular formula provided herein.
  • the compounds described herein are according to methods well known in the art.
  • the conditions of the method such as reactants, solvents, bases, amounts of compounds used, reaction temperature, time required for the reaction, etc. are not limited to the following explanations.
  • the compounds of the present invention can also be conveniently prepared by combining various synthetic methods described in this specification or known in the art, and such combinations can be easily performed by those skilled in the art to which the present invention belongs.
  • the present invention also provides a method for preparing the compound represented by the general formula (1) and the general formula (2), which is prepared by the following general reaction schemes 1, 2, 3 or 4:
  • the embodiment of the compound of general formula (1) can be prepared according to general reaction scheme 1 (method A), wherein R 1 , R 2 , R 4 , A, E and Y are as defined above, and X represents I, Br, Cl, OTf , -B(OH) 2 and other groups.
  • general reaction scheme 1 the intermediate A1 (synthesized according to the method described in WO2017201161) and the fragment A produce A2 under alkaline conditions
  • the compound of structure A2 produces A3 in the presence of an oxidizing agent
  • the compound A3 produces A4 under alkaline conditions.
  • A4 reacts with R 2 -YX fragments under appropriate conditions to produce A5, A5 removes the protective group (such as Boc) to obtain A6, A6 and R1-X couple to obtain A7, A7 removes the protective group (such as Cbz) to obtain A8 , A8 reacts with acid chloride or acid anhydride compound to generate A9.
  • A5 removes the protective group (such as Boc) to obtain A6, A6 and R1-X couple to obtain A7, A7 removes the protective group (such as Cbz) to obtain A8 , A8 reacts with acid chloride or acid anhydride compound to generate A9.
  • the embodiment of the compound of general formula (2) can be prepared according to general reaction scheme 2 (method B), wherein R 1 , R 2 , R 4 , R 6 , R 7 , A, E and Y are as defined above, X represents groups such as I, Br, Cl, OTf, -B(OH) 2 and the like.
  • intermediate B1 (synthesized with reference to the method described in WO2016164675) and fragment A generate B2 under basic conditions
  • compound B2 under basic conditions produces B3, and compound B3 under basic conditions B4, B4 react with R 2 -YX fragments under appropriate conditions to generate B5, B5 and R 1 -X are coupled to obtain B6, B6 continues to be coupled with R 7 X to obtain B7, and B7 is obtained by removing the protective group (such as Cbz) B8, B8 and acid chloride or acid anhydride compound react to produce B9.
  • protective group such as Cbz
  • the embodiment of the compound of general formula (2) can be prepared according to General Reaction Scheme 3 (Method C), wherein R 1 , R 2 , R 4 , R 7 , A, E and Y are as defined above, and X represents I, Br, Cl, OTf, -B(OH) 2 and other groups.
  • the intermediate C1 (synthesized with reference to the method described in US2016164675) undergoes amidation reaction to generate C2, the structure C2 compound reacts with an appropriate reagent to generate substituted isocyanate, which further reacts to generate C3, and compound C3 is basic Under appropriate conditions, the reaction produces pyrimidinedione intermediate C4, C4 reacts with chlorinating reagents under appropriate conditions to produce C5, C5 reacts under alkaline conditions to produce C6, C6 and R 1 -X are coupled to obtain C7, C7 deprotection group ( For example, Boc) obtains C8, C8 reacts with acid chloride or acid anhydride compound to generate C9.
  • Boc Boc
  • the embodiment of the compound of general formula (2) can be prepared according to general reaction scheme 4 (method D), wherein R 1 , R 2 , R 4 , R 7 , R 8 , A, E and Y are as defined above .
  • general reaction scheme 4 intermediate D1 (synthesized with reference to the method described in WO2016164675) and fragment A generate D2 under alkaline conditions, and the compound of structure D2 generates D3 under alkaline conditions, and D3 and R 2 under appropriate conditions -YX fragments react to form D4, D4 and R 1 -X are coupled to form D5, D5 is deprotected (such as Boc) to form D6, and D6 is reacted with acid chloride or acid anhydride compound to form D7.
  • D5 is deprotected (such as Boc) to form D6
  • D6 is reacted with acid chloride or acid anhydride compound to form D7.
  • “Pharmaceutically acceptable” here refers to a substance, such as a carrier or diluent, that does not make the biological activity or properties of the compound disappear, and is relatively non-toxic, for example, when a substance is administered to an individual, it does not cause unwanted biological effects or Interacts with any components it contains in a harmful way.
  • pharmaceutically acceptable salt refers to a form of a compound that does not cause important irritation to the administered organism and does not cause the biological activity and properties of the compound to disappear.
  • pharmaceutically acceptable salts are obtained by reacting compounds of formula (1) or formula (2) with acids, such as inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid, nitric acid, phosphoric acid, etc.
  • references to pharmaceutically acceptable salts include solvent addition forms or crystalline forms, especially solvates or polymorphs.
  • Solvates contain stoichiometric or non-stoichiometric solvents, and are selectively formed during crystallization with pharmaceutically acceptable solvents such as water, ethanol, etc.
  • a hydrate is formed when the solvent is water, or an alcoholate is formed when the solvent is ethanol.
  • the solvate of the compound of formula (1) or formula (2) is conveniently prepared or formed according to the method described herein.
  • the hydrate of the compound of formula (1) or formula (2) is conveniently prepared by recrystallization from a mixed solvent of water/organic solvent.
  • the organic solvents used include, but are not limited to, dioxane and tetrahydrofuran. , Ethanol or methanol.
  • the compounds mentioned here can exist in unsolvated and solvated forms. In summary, for the purposes of the compounds and methods provided herein, the solvated form is considered equivalent to the unsolvated form.
  • the compounds of formula (1) or (2) are prepared in different forms, including, but not limited to, amorphous, pulverized, and nano-particle size forms.
  • the compound of formula (1) or formula (2) includes a crystalline form, and may also be a polymorphic form.
  • Polymorphs include different lattice arrangements of the same elemental composition of the compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal form, optical and electrical properties, stability and solubility. Different factors such as recrystallization solvent, crystallization rate and storage temperature may cause a single crystal form to dominate.
  • the compound of formula (1) or formula (2) has one or more stereocenters, and is therefore classified as racemate, racemic mixture, single enantiomer, diastereomeric compound and single diastereomer Appears in the form of enantiomers.
  • the asymmetric centers that can exist depend on the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers, and all possible mixtures of optical isomers and diastereomers and pure or partially pure compounds are included in the scope of the present invention. The present invention is meant to include all such isomeric forms of these compounds.
  • Alkyl refers to saturated aliphatic hydrocarbon groups, including straight and branched chain groups of 1 to 6 carbon atoms. Preferably, a lower alkyl group containing 1 to 4 carbon atoms, such as methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl, tert-butyl.
  • alkyl includes unsubstituted and substituted alkyl groups, especially alkyl groups substituted with one or more halogens.
  • Preferred alkyl groups are selected from CH 3 , CH 3 CH 2 , CF 3 , CHF 2 , CF 3 CH 2 , i Pr, n Pr, i Bu, c Pr, n Bu or t Bu.
  • Cycloalkyl refers to a 3- to 6-membered all-carbon monocyclic aliphatic hydrocarbon group, in which one or more rings may contain one or more double bonds, but none of the rings have a fully conjugated ⁇ -electron system.
  • cyclopropyl, cyclobutyl, cyclopentyl, cyclohexane, cyclohexadiene and the like are examples of the rings.
  • Alkoxy refers to an alkyl group bonded to the rest of the molecule through an ether oxygen atom.
  • Representative alkoxy groups are those with 1-6 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy And tert-butoxy.
  • alkoxy includes unsubstituted and substituted alkoxy, especially alkoxy substituted with one or more halogens.
  • Preferred alkoxy groups are selected from OCH 3 , OCF 3 , CHF 2 O, CF 3 CH 2 O, i PrO, n PrO, i BuO, c PrO, n BuO or t BuO.
  • Aryl refers to a group having at least one aromatic ring structure, that is, a carbocyclic aryl group having a conjugated ⁇ electron system, such as a benzene ring and a naphthalene ring.
  • Heteroaryl refers to an aromatic group containing one or more heteroatoms (O, S or N).
  • the heteroaryl group is monocyclic or polycyclic, such as a monocyclic heteroaryl ring and one or more carbocyclic rings. Aromatic groups or other monocyclic heterocyclic groups are condensed.
  • heteroaryl groups include, but are not limited to, pyridyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, furanyl , Thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, indolyl, benzimidazolyl, benzofuranyl, benzothiazolyl, benzothienyl, benzoxanyl Azolyl, benzopyridyl and pyrrolopyrimidinyl.
  • Halogen refers to fluorine, chlorine, bromine or iodine.
  • bond refers to a chemical bond between two atoms or between two fragments (when the atoms connected by a bond are considered part of a larger structure).
  • bond refers to a chemical bond between two atoms or between two fragments (when the atoms connected by a bond are considered part of a larger structure).
  • bond when the group described herein is a bond, the lack of a reference group allows a bond to be formed between the remaining defined groups.
  • membered ring includes any cyclic structure.
  • element means to indicate the number of skeletal atoms constituting the ring.
  • cyclohexyl, pyridyl, pyranyl, and thiopyranyl are six-membered rings
  • cyclopentyl, pyrrolyl, furyl, and thienyl are five-membered rings.
  • fragment refers to a specific part or functional group of a molecule. Chemical fragments are generally considered to be chemical entities contained or attached to molecules.
  • treatment refers to a prescription component or active ingredient that does not have unduly harmful effects on the health of the general treatment target.
  • treatment includes alleviating, inhibiting, or improving the symptoms or conditions of diseases; inhibiting the occurrence of complications; improving or preventing underlying metabolic syndrome; inhibiting the occurrence of diseases or symptoms, Such as controlling the development of diseases or conditions; reducing diseases or symptoms; reducing diseases or symptoms; reducing complications caused by diseases or symptoms, or preventing or treating signs caused by diseases or symptoms.
  • a certain compound or pharmaceutical composition after administration, can improve a certain disease, symptom or condition, especially its severity, delay the onset, slow the progression of the disease, or reduce the duration of the disease. Regardless of fixed administration or temporary administration, continuous administration or intermittent administration, it can be attributed to or related to the administration.
  • Active ingredient refers to a compound represented by general formula (1) or general formula (2), and a pharmaceutically acceptable inorganic or organic salt of a compound of general formula (1) or general formula (2).
  • the compounds of the present invention may contain one or more asymmetric centers, and therefore appear as racemates, racemic mixtures, single enantiomers, diastereomeric compounds, and single diastereomers.
  • the asymmetric centers that can exist depend on the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers, and all possible mixtures of optical isomers and diastereomers and pure or partially pure compounds are included within the scope of the present invention.
  • the present invention is meant to include all such isomeric forms of these compounds.
  • composition refers to when applied to an individual (human Or animal), a compound or composition that can induce the desired pharmaceutical and/or physiological response through local and/or systemic effects.
  • administered refers to the direct administration of the compound or composition, or the administration of a prodrug, derivative, or analog of the active compound Etc., and can form an equivalent amount of the active compound in the subject of administration.
  • the present invention provides methods for using the compounds or pharmaceutical compositions of the present invention to treat conditions, including but not limited to conditions involving G12C K-Ras, G12C H-Ras, and/or G12C N-Ras mutations (such as cancer).
  • a method for cancer treatment comprising administering to an individual in need an effective amount of any of the aforementioned pharmaceutical compositions comprising a compound of structure (1) or (2).
  • the cancer is mediated by K-Ras, H-Ras, and/or G12C N-Ras mutations.
  • the cancer is lung cancer, pancreatic cancer, colon cancer, MYH-related polyposis, or colorectal cancer.
  • the compound of the present invention and its pharmaceutically acceptable salt can be prepared into various preparations, which contain a safe and effective amount of the compound of the present invention or its pharmaceutically acceptable salt and a pharmacologically acceptable excipient or carrier .
  • the "safe and effective amount” refers to: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the safe and effective amount of the compound is determined according to the age, condition, and course of treatment of the subject to be treated.
  • “Pharmaceutically acceptable excipient or carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use, and must have sufficient purity and sufficiently low toxicity .
  • Compatibility here means that each component of the composition can be blended with the compound of the present invention and between them without significantly reducing the efficacy of the compound.
  • pharmacologically acceptable excipients or carriers include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers Wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with the following ingredients: (a) fillers or compatibilizers, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders such as hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) humectants, For example, glycerin; (d) disintegrants, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) Absorption accelerators, such as quaternary amine compounds; (g) wetting agents, such as cetyl alcohol and glycty
  • Solid dosage forms such as tablets, sugar pills, capsules, pills and granules can be prepared with coatings and shell materials, such as enteric coatings and other materials known in the art. They may contain opacifying agents, and the active compound or the release of the compound in such a composition may be released in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be used are polymeric substances and waxes. If necessary, the active compound can also be formed into microcapsules with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • the liquid dosage form may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-Butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances.
  • the composition may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • composition for parenteral injection may contain physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • the dosage form of the compound of the present invention for topical administration includes ointment, powder, patch, spray and inhalant.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants that may be required if necessary.
  • the compound of the present invention can be administered alone or in combination with other pharmaceutically acceptable compounds.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage is the pharmaceutically effective dosage considered to be administered.
  • the daily administration dose is usually 1 to 1000 mg, preferably 10 to 500 mg.
  • the specific dosage should also consider factors such as the route of administration, the patient's health status, etc., which are within the skill range of a skilled physician.
  • Ar stands for argon; CDCl 3 stands for deuterated chloroform; CDI stands for 1,1'-carbonyldiimidazole; CD 3 OD stands for deuterated methanol; CuI stands for cuprous iodide; Methyl chloride; DIPEA stands for diisopropylethylamine; DMF stands for dimethylformamide; DMSO stands for dimethyl sulfonate; EA stands for ethyl acetate; h stands for hours; NaOH stands for sodium hydroxide; LC-MS stands for liquid Phase-mass spectrometry; m-CPBA stands for m-chloroperoxybenzoic acid; MeOH stands for methanol; min stands for minutes; MS stands for mass spectrometry; NMR stands for nuclear magnetic resonance; Pd(dppf)2Cl2 stands for [1,1'-bis(diphenylphosphine) ) Ferrocene] dichloropalladium dichloropalladium dichloro
  • Compound 1 was prepared according to Method A as described below:
  • Compound 53 was prepared according to Method B as described below:
  • CDI (35.6g, 220mmol) was added to C1 (42g, 200mmol) in THF (400mL) in batches. The mixture was stirred for 5min, protected by Ar, heated to 50°C, reacted for 1h, monitored by LC-MS, and the raw materials disappeared. The solution was diluted with toluene (100 mL) and concentrated to half of the initial volume. The resulting mixture was cooled to 0° C., and ammonium hydroxide (55 mL, 400 mmol) was slowly added. React at room temperature for 10 minutes, dilute with EA (200 mL), and wash with water (100 mL*3). The organic layer was dried with anhydrous Na 2 SO 4 and spin-dried.
  • H358 cells were planted in an ultra-low adsorption six-well plate. After one day of growth, the compound to be tested (at a concentration of 1 ⁇ M) was added. After the compound was used for 24 hours, the cells were lysed, and GST-Raf1(1-149) was added ( millipore 14-863), after incubating at 4°C overnight, add GST beads (millipore, G0924), incubate at 4°C for 2 hours, then centrifuge, take the beads, wash the beads with IP buffer 3 times, then add SDS lysis solution, and run Gel, use Ras antibody (CST, 3339) western blot to detect the pulled Ras-GTP content. Compared with the DMSO group, the percentage of the compound inhibiting Ras-GTP activity was calculated. The results are shown in Table 2 below.
  • Compound Inhibition rate(%) Compound Inhibition rate(%) Compound Inhibition rate(%) 1 +++ 2 ++ 3 ++ 4 ++ 5 ++ 6 ++ 7 ++ 8 + 9 ++ 10 +++ 11 +++ 12 +++ 13 +++ 14 +++ 15 +++ 16 +++ 17 +++ 18 +++ 19 +++ 20 ++ twenty one +++ twenty two +++ twenty three +++ twenty four +++
  • +++ means the inhibition rate is greater than 90%
  • Compound Inhibition rate(%) Compound Inhibition rate(%) Compound Inhibition rate(%) 1 +++ 2 ++ 3 ++ 4 + 5 ++ 6 ++ 7 ++ 8 + 9 ++ 10 +++ 11 +++ 12 +++ 13 +++ 14 +++ 15 +++ 16 +++ 17 +++ 18 +++ 19 ++ 20 +++ twenty one +++ twenty two +++ twenty three +++ twenty four +++ 25 +++ 26 +++ 27 +++ 28 +++ 29 +++ 30 ++ 31 +++ 32 +++ 33 +++ 34 +++ 35 +++ 36 +++ 37 ++ 38 +++ 39 +++ 40 +++ 41 +++ 42 +++ 43 +++ 44 +++ 45 +++ 46 +++ 47 +++ 48 +++ 49 +++ 50 +++ 51 ++ 52 ++ 53 +++ 54 +++ 55 +++ 56 +++ 57 +++ 58 +++ 59 ++ 60 +++ 61 +++ 62 +++ 63 +++
  • +++ means the inhibition rate is less than 1 ⁇ M
  • the compounds of the present invention have good biological activities, including the inhibitory activity of Ras-GTP in H358 cells and the anti-proliferation activity of H358 cells.

Abstract

本发明提供了G12C突变体K-Ras蛋白不可逆抑制剂,本发明还公开了上述G12C突变体K-Ras蛋白不可逆抑制剂及其制备方法和用途。

Description

KRAS-G12C抑制剂
本申请要求申请日为2019年6月26日的中国专利申请CN2019105708974的优先权。本申请引用上述中国专利申请的全文。
技术领域
本发明属涉及药物化学领域,更具体而言,涉及一类新型KRAS-G12C抑制剂,及其制备方法和该类化合物的使用方法。
背景技术
RAS代表一组紧密相关的单体球状蛋白质(21kDa分子量),其具有189个氨基酸且与质膜相连并且结合GDP或GTP。在正常发育或生理条件下,RAS接收生长因子和各种其它细胞外信号而被激活,负责调节细胞生长、存活、迁移和分化等功能。RAS起分子开关作用,RAS蛋白的开/关状态通过核苷酸结合确定,活性信号传导构象结合GTP,非活性构象结合GDP。当RAS包含结合的GDP时,其处于休眠或静止或关闭状态,并且是“非活化的”,响应于暴露于某些促生刺激,诱导RAS将结合的GDP转换为GTP。随着GTP被结合,RAS是“开启的”,并且能够与其它蛋白相互作用且活化其它蛋白(其“下游靶标”)。RAS蛋白本身具有极低的将GTP水解回到GDP并由此将自身变为关闭状态的固有能力。将RAS转换为关闭需要称作GTP酶激活蛋白(GAPs)的外源性蛋白,其与RAS相互作用并且能大大促进GTP向GDP的转化。任何在RAS中的影响其与GAP相互作用或将GTP转化回到GDP的能力的突变,将会导致所述蛋白的延长的活化,并且因此产生到细胞的延长的信号,该信号告知其继续生长和分裂。因此这些信号会使得细胞生长和分裂,过度活化的RAS信号转导可能最终导致癌症。
在RAS家族成员中,致癌突变最常见于KRAS(85%),而NRAS(12%)和HRAS(3%)则较为少见。KRAS突变在美国三大致命癌症类型中普遍存在:胰腺癌(95%)、结肠直肠癌(45%)和肺癌(25%),而在乳腺癌、卵巢癌和脑癌中很少发现(<2%)。在非小细胞肺癌(NSCLC)中,KRAS G12C是最常见的突变,占所有KRAS突变的近一半,其次是G12V和G12D。在非小细胞肺癌中,特定等位基因突变频率的增加多来自经典的由吸烟诱导的典型突变(G:C至T:A置换),从而导致了KRAS G12C(GGT至TGT)和G12V(GGT至GTT)突变。
大型基因组学研究表明,肺癌KRAS突变,包括G12C,与NSCLC中其它已知的驱动致癌突变相互排斥,包括EGFR、ALK、ROS1、RET和BRAF,表明KRAS突变在肺癌中的独特性。而同时,KRAS突变经常与某些共突变共同发生,例如STK11、KEAP1和TP53,它们与突变的RAS合作将细胞转化为高度恶性和侵袭性的肿瘤细胞。
三种RAS癌基因构成了人类癌症中突变最频繁的基因家族。令人失望的是,尽管经过三十多年的研究努力,临床上仍然没有有效的抗RAS疗法,使用小分子靶向该基因是项挑战。因此,本领域迫切需要用于靶向RAS(例如,K-RAS,H-RAS和/或N-RAS)的小分子并且利用其治疗多种疾病,例如癌症。
发明内容
本发明旨在提供一类结构通式如式(1)所示的化合物、或其各光学异构体、各晶型、药学上可接受的盐、水合物或溶剂合物:
Figure PCTCN2020097397-appb-000001
式(1)中:
m为0、1或2;
A为一个二价含有1-2个N原子的4-12元饱和或部分饱和的单环、双环、桥环或螺环,所述单环、双环、桥环或螺环可任选被一个或多个R 4所取代,当被多个R 4取代时,R 4可以相同或不同;
Y为化学键或C1-C6亚烷基;
R 1为芳基或杂芳基,所述芳基或杂芳基可被1-3个下述基团所取代:卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基;
R 2为胺基烷基、环烷基、烷基取代酰胺基、杂环基、芳基或杂芳基,所述杂环基、芳基或杂芳基可任选被一个或多个R 5所取代,当被多个R 5取代时,R 5可以相同或不同;
R 3独立选择C1-C3烷基或卤代C1-C3烷基;
R 4独立选自H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基;
R 5独立选自卤素、H、O、CN、OH、烷基羟基、二烷基胺基、C1-C6烷基、C3-C6 环烷基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基;
E是能够与K-Ras、H-Ras或N-Ras突变体蛋白的12位的半胱氨酸残基形成共价键的亲电部分。
在另一优选例中,其中所述式(1)中,E为一个含亲电碳碳双键或碳碳叁键的基团。
在另一优选例中,其中所述式(1)中,E为:
Figure PCTCN2020097397-appb-000002
其中,R a为H或F,R b为H、-CH 2F、-CHF 2
Figure PCTCN2020097397-appb-000003
Figure PCTCN2020097397-appb-000004
在另一优选例中,其中所述式(1)中,A-E为:
Figure PCTCN2020097397-appb-000005
Figure PCTCN2020097397-appb-000006
其中,n为1或2,R 4为H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基。
在另一优选例中,其中所述式(1)中,Y为化学键、-CH 2-、-CH(Me)-或-CH 2CH 2-。
在另一优选例中,其中所述式(1)中,R 1为:
Figure PCTCN2020097397-appb-000007
Figure PCTCN2020097397-appb-000008
其中R c和R d独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基。
在另一优选例中,其中所述式(1)中,R 2为:
Figure PCTCN2020097397-appb-000009
Figure PCTCN2020097397-appb-000010
Figure PCTCN2020097397-appb-000011
其中n为1或2,R e和R f独立地为H、卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基,R g为C1-C3烷基、C3-C6环烷基、C3-C6环烷基烷基、C1-C3烷氧基烷基、卤素取代C1-C3烷基、卤素取代C3-C6环烷基或
Figure PCTCN2020097397-appb-000012
本发明的另一方面,旨在提供一类结构通式如式(2)所示的化合物、或其各光学异构体、各晶型、药学上可接受的盐、水合物或溶剂合物:
Figure PCTCN2020097397-appb-000013
式(2)中:
A为一个二价含有1-2个N原子的4-12元饱和或部分饱和的单环、双环、桥环或螺环,所述单环、双环、桥环或螺环可任选被一个或多个R 4所取代,当被多个R 4取代时,R 4可以相同或不同;
Y为化学键或C1-C6亚烷基;
W为N、C-R 8或C-O-R 6
其中,当W为C-O-R 6时,R 2为胺基烷基、环烷基、烷基取代酰胺基、杂环基、芳基或杂芳基,所述杂环基、芳基或杂芳基可任选被一个或多个R 5所取代,当被多个R 5取代时,R 5可以相同或不同;
其中,当W为N或C-R 8时,R 2为胺基烷基、烷基取代酰胺基或杂环基,所述杂环基可任选被一个或多个R 5所取代,当被多个R 5取代时,R 5可以相同或不同;
R 1为芳基或杂芳基,所述芳基或杂芳基可被1-3个下述基团所取代:卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基、卤素取代C1-C3烷氧基;
R 4独立选自H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基;
R 5独立选自卤素、H、O、CN、OH、烷基羟基、二烷基胺基、C1-C6烷基、C3-C6 环烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基;
R 6为C1-C3烷基、卤素取代C1-C3烷基或C3-C6环烷基;
R 7为H、卤素、C1-C3烷基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基、卤素取代C1-C3烷氧基或C2-C4烯基;
R 8为H、卤素、C1-C3烷基、C3-C6环烷基或卤素取代C1-C3烷基;
E是能够与K-Ras、H-Ras或N-Ras突变体蛋白的12位的半胱氨酸残基形成共价键的亲电部分。
在另一优选例中,其中所述式(2)中,E为一个含亲电碳碳双键或碳碳叁键的基团。
在另一优选例中,其中所述式(2)中,E为:
Figure PCTCN2020097397-appb-000014
其中,R a为H或F,R b为H、-CH 2F、-CHF 2
Figure PCTCN2020097397-appb-000015
Figure PCTCN2020097397-appb-000016
在另一优选例中,其中所述式(2)中,A-E为:
Figure PCTCN2020097397-appb-000017
Figure PCTCN2020097397-appb-000018
其中,n为1或2,其中,R 4为H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基。
在另一优选例中,其中所述式(2)中,Y为化学键、-CH 2-、-CH(Me)-或-CH 2-CH 2-。
在另一优选例中,其中所述式(2)中,R 1为:
Figure PCTCN2020097397-appb-000019
Figure PCTCN2020097397-appb-000020
其中R c和R d独立地为卤素、 羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基。
在另一优选例中,其中所述式(2)中,W为C-O-R 6,R 2为:
Figure PCTCN2020097397-appb-000021
Figure PCTCN2020097397-appb-000022
其中n为1或2,R e和R f独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基、或卤素取代C1-C3烷氧基,R g为C1-C3烷基、C3-C6环烷基、C3-C6环烷基烷基、C1-C3烷氧基烷基、卤素取代C1-C3烷基、卤素取代C3-C6环烷基或
Figure PCTCN2020097397-appb-000023
在另一优选例中,其中所述式(2)中,W为N或C-R 8,R 2为:
Figure PCTCN2020097397-appb-000024
Figure PCTCN2020097397-appb-000025
Figure PCTCN2020097397-appb-000026
其中n为1或2,R e和R f独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基,R g为C1-C3烷基、C3-C6环烷基、C3-C6环烷基烷基、C1-C3烷氧基烷基、卤素取代C1-C3烷基、卤素取代C3-C6环烷基或
Figure PCTCN2020097397-appb-000027
在各种不同实施方式中,化合物具有下表1中所列结构中的一个:
表1:本发明代表性化合物列表:
Figure PCTCN2020097397-appb-000028
Figure PCTCN2020097397-appb-000029
Figure PCTCN2020097397-appb-000030
Figure PCTCN2020097397-appb-000031
Figure PCTCN2020097397-appb-000032
Figure PCTCN2020097397-appb-000033
Figure PCTCN2020097397-appb-000034
Figure PCTCN2020097397-appb-000035
Figure PCTCN2020097397-appb-000036
Figure PCTCN2020097397-appb-000037
Figure PCTCN2020097397-appb-000038
Figure PCTCN2020097397-appb-000039
Figure PCTCN2020097397-appb-000040
Figure PCTCN2020097397-appb-000041
Figure PCTCN2020097397-appb-000042
Figure PCTCN2020097397-appb-000043
Figure PCTCN2020097397-appb-000044
Figure PCTCN2020097397-appb-000045
Figure PCTCN2020097397-appb-000046
Figure PCTCN2020097397-appb-000047
Figure PCTCN2020097397-appb-000048
Figure PCTCN2020097397-appb-000049
Figure PCTCN2020097397-appb-000050
Figure PCTCN2020097397-appb-000051
Figure PCTCN2020097397-appb-000052
Figure PCTCN2020097397-appb-000053
Figure PCTCN2020097397-appb-000054
Figure PCTCN2020097397-appb-000055
Figure PCTCN2020097397-appb-000056
Figure PCTCN2020097397-appb-000057
本发明的另一个目的是提供了一种药物组合物,它含有药理上可接受的赋形剂或载体,以及本发明通式(1)或通式(2)中化合物、或其各光学异构体、药学上可接受的无机或 有机盐做为活性成分。
本发明的再一个目的提供了本发明的上述化合物、或其各光学异构体、药学上可接受的无机或有机盐用于制备治疗肿瘤相关的疾病中的应用。
应理解,本发明的前述一般性描述和以下详细描述都是示例性和说明性的,旨在提供对所要求保护的本发明的进一步说明。
化合物的合成
下面具体地描述本发明通式通式(1)和通式(2)结构化合物的制备方法,但这些具体方法不对本发明构成任何限制。
以上说明的通式(1)和通式(2)化合物可使用标准的合成技术或公知的技术与文中结合的方法来合成。此外,在此提到的溶剂,温度和其他反应条件可以改变。用于化合物的合成的起始物料可以由合成或从商业来源上获得,如,但不限于Aldrich Chemical Co.(Milwaukee,Wis.)或Sigma Chemical Co.(St.Louis,Mo.)。本文所述的化合物和其他具有不同取代基的有关化合物可使用公知的技术和原料来合成,包括发现于March,ADVANCED ORGANIC CHEMISTRY 4 th Ed.,(Wiley 1992);Carey和Sundberg,ADVANCED ORGANIC CHEMISTRY 4 th Ed.,Vols.A和B(Plenum 2000,2001),Green和Wuts,PROTECTIVE GROUPS IN ORGANIC SYNTHESIS 3 rd Ed.,(Wiley 1999)中的方法。化合物制备的一般方法可通过使用适当的试剂和在此提供的分子式中引入不同基团的条件来改变。
一方面,本文所述的化合物根据工艺中公知的方法。然而方法的条件,例如反应物、溶剂、碱、所用化合物的量、反应温度、反应所需时间等不限于下面的解释。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便的制得,这样的组合可由本发明所属领域的技术人员容易的进行。一方面,本发明还提供了一种所述的通式通式(1)和通式(2)所示化合物的制备方法,其采用下列一般反应流程1、2、3或4制备:
一般反应流程1
Figure PCTCN2020097397-appb-000058
通式(1)化合物的实施方式可根据一般反应流程1(方法A)制备,其中R 1、R 2、R 4、A、E及Y如上文中所定义,X表示I、Br、Cl、OTf、-B(OH) 2等基团。如一般反应流程1所示,中间体A1(参照WO2017201161中描述的方法合成)和片段A在碱性条件下生产A2,结构A2化合物在氧化剂存在下生成A3,化合物A3在碱性条件下生成A4,A4在适当条件下和R 2-Y-X的片段反应生成A5,A5脱除保护基(例如Boc)得到A6,A6和R1-X偶联得到A7,A7脱除保护基(例如Cbz)得到A8,A8和酰氯或酸酐化合物反应生成A9。
一般反应流程2
Figure PCTCN2020097397-appb-000059
一方面,通式(2)化合物的实施方式可根据一般反应流程2(方法B)制备,其中R 1、R 2、R 4、R 6、R 7、A、E及Y如上文中所定义,X表示I、Br、Cl、OTf、-B(OH) 2等基团。如一般反应流程2所示,中间体B1(参照WO2016164675中描述的方法合成)和片段A在碱性条件下生成B2,结构B2化合物在碱性条件下生成B3,化合物B3在碱性条件下生成B4,B4在适当条件下和R 2-Y-X的片段反应生成B5,B5和R 1-X偶联得到B6,B6继续和R 7X偶联得到B7,B7脱除保护基(例如Cbz)得到B8,B8和酰氯或酸酐化合物反应生成B9。
一般反应流程3
Figure PCTCN2020097397-appb-000060
另一方面,通式(2)化合物的实施方式可根据一般反应流程3(方法C)制备,其中R 1、R 2、R 4、R 7、A、E及Y如上文中所定义,X表示I、Br、Cl、OTf、-B(OH) 2等基团。如一般反应流程3所示,中间体C1(参照US2016164675中描述的方法合成)进行酰胺化反应生成C2,结构C2化合物和适当的试剂反应生成取代异氰酸酯,其进一步反应生成C3,化合物C3在碱性条件下反应生成嘧啶二酮中间体C4,C4在适当条件下和氯化试剂反应生成C5,C5在碱性条件反应生成C6,C6和R 1-X偶联得到C7,C7脱除保护基(例如Boc)得到C8,C8和酰氯或酸酐化合物反应生成C9。
一般反应流程4
Figure PCTCN2020097397-appb-000061
再一方面,通式(2)化合物的实施方式可根据一般反应流程4(方法D)制备,其中R 1、R 2、R 4、R 7、R 8、A、E及Y如上文中所定义。如一般反应流程4所示,中间体D1(参照WO2016164675中描述的方法合成)和片段A在碱性条件下生成D2,结构D2化合物在碱性条件下生成D3,D3在适当条件下和R 2-Y-X的片段反应生成D4,D4和R 1-X偶联得到D5,D5脱除保护基(例如Boc)得到D6,D6和酰氯或酸酐化合物反应生成D7。
化合物的进一步形式
“药学上可接受”这里指一种物质,如载体或稀释液,不会使化合物的生物活性或性质消失,且相对无毒,如,给予个体某物质,不会引起不想要的生物影响或以有害的方式与任何其含有的组分相互作用。
术语“药学上可接受的盐”指一种化合物的存在形式,该形式不会引起对给药有机体的重要的刺激,且不会使化合物的生物活性和性质消失。在某些具体方面,药学上可接受的盐是通过式(1)或式(2)化合物与酸反应获得,如盐酸、氢溴酸、氢氟酸、硫酸、磷酸、硝酸、磷酸等无机酸,甲酸、乙酸、丙酸、草酸、三氟乙酸、丙二酸、琥珀酸、富马酸、马来酸、乳酸、苹果酸、酒石酸、柠檬酸、苦味酸、甲磺酸、苯磺酸、对甲苯磺酸等有机酸以及天冬氨酸、谷氨酸等酸性氨基酸。
应理解药学上可接受的盐的参考包括溶剂添加形式或结晶形式,尤其是溶剂化物或多晶型。溶剂化物含有化学计量或非化学计量的溶剂,且是在与药学上可接受溶剂如水,乙醇等,结晶化过程中选择性形成的。当溶剂是水时形成水合物,或当溶剂是乙醇时形成醇化物。式(1)或式(2)化合物的溶剂化物按照本文所述的方法,很方便的制得或形成。举例说明,式(1)或式(2)化合物的水合物从水/有机溶剂的混合溶剂中重结晶而方便的制得,使用的有机溶剂包括但不限于,二氧杂环乙烷,四氢呋喃,乙醇或甲醇。此外,在此提到的化合物能够以非溶剂化和溶剂化形式存在。总之,对于在此提供的化合物和方法为目的,溶剂化形式被认为相当于非溶剂化形式。
在其他具体实施例中,式(1)或式(2)化合物被制备成不同的形式,包括但不限于,无定形,粉碎形和毫微-粒度形式。此外,式(1)或式(2)化合物包括结晶型,也可以作为多晶型。多晶型包括化合物的相同元素组成的不同晶格排列。多晶型通常有不同的X-射线衍射图,红外光谱,熔点,密度,硬度,晶型,光和电的性质,稳定性和溶解性。不同的因素如重结晶溶剂,结晶速率和贮存温度可能引起单一晶型为主导。
在另一个方面,式(1)或式(2)化合物有一个或多个立体中心,并因此以消旋体、外消旋混合物、单一对映体、非对映异构体化合物和单一非对映体的形式出现。可以存在的不对称中心,取决于分子上各种取代基的性质。每个这种不对称中心将独立地产生两个 旋光异构体,并且所有可能的旋光异构体和非对映体混合物以及纯或部分纯的化合物包括在本发明的范围之内。本发明意味着包括这些化合物的所有这种异构形式。
术语
如果无另外说明,用于本发明申请,包括说明书和权利要求书中的术语,定义如下。必须注意,在说明书和所附的权利要求书中,如果文中无另外清楚指示,单数形式“一个”包括复数意义。如果无另外说明,使用质谱、核磁、HPLC、蛋白化学、生物化学、重组DNA技术和药理的常规方法。在本申请中,如果无另外说明,使用“或”或“和”指“和/或”。
“烷基”指饱和的脂肪烃基团,包括1至6个碳原子的直链和支链基团。优选含有1至4个碳原子的低级烷基,例如甲基、乙基、丙基、2-丙基、正丁基、异丁基、叔丁基。如本文所用,“烷基”包括未取代和取代的烷基,尤其是被一个或多个卤素所取代的烷基。优选的烷基选自CH 3,CH 3CH 2,CF 3,CHF 2,CF 3CH 2iPr, nPr, iBu, cPr, nBu或 tBu。
“环烷基”指3至6元全碳单环脂肪烃基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。例如,环丙基、环丁基、环戊基、环己烷、环己二烯等。
“烷氧基”指通过醚氧原子键合到分子其余部分的烷基。代表性的烷氧基为具有1-6个碳原子的烷氧基,如甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、异丁氧基、仲丁氧基和叔丁氧基。如本文所用,“烷氧基”包括未取代和取代的烷氧基,尤其是被一个或多个卤素所取代的烷氧基。优选的烷氧基选自OCH 3,OCF 3,CHF 2O,CF 3CH 2O, iPrO, nPrO, iBuO, cPrO, nBuO或 tBuO。
“芳基”指具有至少一个芳环结构的基团,即具有共轭的π电子系统的碳环芳基,如苯环和萘环。
“杂芳基”指含有一个或多个杂原子(O,S或N)的芳香基团,杂芳基是单环或多环的,例如单环杂芳基环与一个或多个碳环芳香基团或其它单环杂环基基团稠和。杂芳基的例子包括但不限于,吡啶基、哒嗪基、咪唑基、嘧啶基、吡唑基、三唑基、吡嗪基、喹啉基、异喹啉基、四唑基、呋喃基、噻吩基、异噁唑基、噻唑基、噁唑基、异噻唑基、吡咯基、吲哚基、苯并咪唑基、苯并呋喃基、苯并噻唑基、苯并噻吩基、苯并噁唑基、苯并吡啶基和吡咯并嘧啶基。
“卤素”指氟、氯、溴或碘。
术语“键”或“单键”指两个原子间或两个片断间(当通过键来连接的原子被认为是大结构的一部分时)的化学键。一方面,当本文所述的基团是一个键时,缺少参考基团,允许在剩余的确定基团间形成一个键。
术语“元环”包括任何环状结构。术语“元”意为表示构成环的骨架原子的数量。这样,如,环己基、吡啶基、吡喃基、噻喃基是六元环,环戊基、吡咯基、呋喃基和噻吩基是五元环。
术语“片断”指分子的具体部分或官能团。化学片断通常被认为是包含在或附在分子中的化学实体。
特定药学及医学术语
术语“可接受的”,如本文所用,指一个处方组分或活性成分对一般治疗目标的健康没有过分的有害影响。术语“治疗”、“治疗过程”或“疗法”如本文所用,包括缓和、抑制或改善疾病的症状或状况;抑制并发症的产生;改善或预防潜在代谢综合症;抑制疾病或症状的产生,如控制疾病或情况的发展;减轻疾病或症状;使疾病或症状减退;减轻由疾病或症状引起的并发症,或预防或治疗由疾病或症状引起的征兆。如本文所用,某一化合物或药物组合物,给药后,可以使某一疾病、症状或情况得到改善,尤指其严重度得到改善,延迟发病,减缓病情进展,或减少病情持续时间。无论固定给药或临时给药、持续给药或断续给药,可以归因于或与给药有关的情况。
“活性成分”指通式(1)或通式(2)所示化合物,以及通式(1)或通式(2)化合物的药学上可接受的无机或有机盐。本发明的化合物可以含有一个或多个不对称中心,并因此以消旋体、外消旋混合物、单一对映体、非对映异构体化合物和单一非对映体的形式出现。可以存在的不对称中心,取决于分子上各种取代基的性质。每个这种不对称中心将独立地产生两个旋光异构体,并且所有可能的旋光异构体和非对映体混合物以及纯或部分纯的化合物包括在本发明的范围之内。本发明意味着包括这些化合物的所有这种异构形式。
“化合物(compound)”、“组合物(composition)”、“药剂(agent)”或“医药品(medicine or medicament)”等词在此可交替使用,且都是指当施用于一个体(人类或动物)时,能够透过局部和/或全身性作用而诱发所亟求的药学和/或生理反应的一种化合物或组合物。
“施用(administered、administering或、administration)”一词在此是指直接施用所述的化合物或组合物,或施用活性化合物的前驱药(prodrug)、衍生物(derivative)、或类似物(analog)等,而可在施用个体体内形成该活性化合物的一相当用量者。
虽然用以界定本发明较广范围的数值范围与参数皆是约略的数值,此处已尽可能精确地呈现具体实施例中的相关数值。然而,任何数值本质上不可避免地含有因个别测试方法所致的标准偏差。在此处,“约”通常是指实际数值在一特定数值或范围的正负10%、5%、1%或0.5%之内。或者是,“约”一词代表实际数值落在平均值的可接受标准误差之内,视本领域技术人员的考虑而定。除了实验例之外,或除非另有明确的说明,当可理解 此处所用的所有范围、数量、数值与百分比(例如用以描述材料用量、时间长短、温度、操作条件、数量比例及其它相似者)均经过“约”的修饰。因此,除非另有相反的说明,本说明书与附随权利要求书所揭示的数值参数皆为约略的数值,且可视需求而更动。至少应将这些数值参数理解为所指出的有效位数与套用一般进位法所得到的数值。
除非本说明书另有定义,此处所用的科学与技术词汇的含义与本领域技术人员所理解与惯用的意义相同。此外,在不和上下文冲突的情形下,本说明书所用的单数名词涵盖该名词的复数型;而所用的复数名词时亦涵盖该名词的单数型。
治疗用途
本发明提供了使用本发明化合物或药物组合物治疗病况的方法,包括但不限于涉及G12C K-Ras、G12C H-Ras和/或G12C N-Ras突变的病况(例如癌症)。在一些实施方案中,提供了用于癌症治疗的方法,该方法包括给予有需要的个体有效量的任何前述的包含结构(1)或(2)化合物的药物组合物。在一些实施方案中,癌症由K-Ras、H-Ras和/或G12C N-Ras突变介导。在其它实施方案中,该癌症是肺癌、胰腺癌、结肠癌、MYH相关息肉病或结肠直肠癌。
给药途径
本发明的化合物及其药学上可接受的盐可制成各种制剂,其中包含安全、有效量范围内的本发明化合物或其药学上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全、有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。化合物的安全、有效量根据治疗对象的年龄、病情、疗程等具体情况来确定。“药学上可以接受的赋形剂或载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能与本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药理上可以接受的赋形剂或载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂
Figure PCTCN2020097397-appb-000062
润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。施用本发明化合物时,可以口服、直肠、肠胃外(静脉内、肌肉内或皮下)、局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b) 粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。本发明化合物可以单独给药,或者与其他药学上可接受的化合物联合给药。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~1000mg,优选10~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。本发明提到的上述特征,或实施例提到的特征可以任意组合。本案说明书所揭示的所有特征可与任何组合物形式并用,说明书中所揭示的各个特征,可以任何可提供相同、均等或相似目的的替代性特征取代。因 此除有特别说明,所揭示的特征仅为均等或相似特征的一般性例子。
在下面的说明中将会详细阐述上述化合物、方法、药物组合物的各个具体方面、特性和优势,使本发明的内容变得十分明了。在此应理解,下述的详细说明及实例描述了具体的实施例,仅用于参考。在阅读了本发明的说明内容后,本领域的技术人员可对本发明作各种改动或修改,这些等价形势同样落于本申请所限定的范围。所有实施例中, 1H-NMR用Vian Mercury 400核磁共振仪记录,化学位移以δ(ppm)表示;分离用硅胶未说明均为200-300目,洗脱液的配比均为体积比。本发明采用下述缩略词:Ar代表氩气;CDCl 3代表氘代氯仿;CDI代表1,1'-羰基二咪唑;CD 3OD代表氘代甲醇;CuI代表碘化亚铜;DCM代表二氯甲烷;DIPEA代表二异丙基乙基胺;DMF代表二甲基甲酰胺;DMSO代表二甲基亚风;EA代表乙酸乙酯;h代表小时;NaOH代表氢氧化钠;LC-MS代表液相-质谱;m-CPBA代表间氯过氧苯甲酸;MeOH代表甲醇;min代表分钟;MS代表质谱;NMR代表核磁共振;Pd(dppf)2Cl2代表[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物;PE代表石油醚;Ruphos-Pd-G3代表甲磺酸(2-二环己基膦基-2',6'-二异丙氧基-1,1'-联苯基)(2-氨基-1,1'-联苯-2-基)钯(II);TFA代表三氟乙酸;THF代表四氢呋喃。
具体实施方式
实施例1(S)-4-(4-丙烯酰哌嗪-1-基)-7-(5-甲基-1H-吲唑-4-基)-1-((1-甲基吡咯烷-2-基)甲基)-5,6,7,8-四氢吡啶并[3,4-d]嘧啶-2(1H)-酮(化合物1)的合成
Figure PCTCN2020097397-appb-000063
化合物1根据如下文所述的方法A制备:
4-(4-((苄氧基)羰基)哌嗪-1-基)-2-(甲巯基)-5,8-二氢吡啶并[3,4-d]嘧啶-7(6H)-甲酸叔丁酯(1-1)
往250mL单口瓶中加入A1(5.2g,12.11mmol),DIPEA(3.2g,24.22mmol),苄基-1-哌嗪碳酸酯(2.9g,13.31mmol)和DMF(50mL),Ar保护下,升温至100℃反应1小时。TLC监测(PE/EA=10/1),原料反应完全,将反应液冷却至室温后,加入水(100mL),用EA(50mL*2)萃取,合并有机相,饱和氯化钠洗,有机相浓缩,柱层析纯化(PE/EA=1/0to 2/1)得白色固体1-1(6.03g,收率99%),ESI-MS m/z:500.2[M+H] +
4-(4-((苄氧基)羰基)哌嗪-1-基)-2-(甲砜基)-5,8-二氢吡啶并[3,4-d]嘧啶-7(6H)-甲酸叔丁酯(1-2)
往250mL单口瓶中加入1-1(6.03g,12.07mmol)和DCM(60mL),Ar保护下,冰浴冷却至0-5℃,加入m-CPBA(7.29g,42.24mmol),冰浴下反应2小时。TLC监测(PE/EA=1/10),原料反应完全,往反应液中加入饱和碳酸氢钠溶液(60mL),搅拌,分液;水相再用DCM(50mL)萃取,合并有机相,饱和氯化钠洗,有机相浓缩,柱层析纯化(PE/EA=1:0 to 1/1)得白色固体1-2(4.96g,收率78%),ESI-MS m/z:532.1[M+H] +
4-(4-((苄氧基)羰基)哌嗪-1-基)-2-氧代-2,5,6,8-四氢吡啶并[3,4-d]嘧啶-7(1H)-甲酸叔丁酯(1-3)
往250mL单口瓶中加入1-2(4.694g,8.83mmol)和1,4-二氧六环(88mL),然后加入氢氧化钠溶液(44.1mL,2N),室温反应过夜,TLC监测(PE/EA=1/1),原料反应完全,用2N盐酸调pH至7,析出固体,过滤得白色固体1-3;滤液再用EA(30mL)萃取,饱和氯化钠溶液洗,无水硫酸钠干燥,过滤,浓缩,粗品加入EA(5mL)打浆搅拌,过滤得白色固体1-3,合并得到1-3(3.48g,收率79%),ESI-MS m/z:470.3[M+H] +
(S)-4-(4-((苄氧基)羰基)哌嗪-1-基)-1-((1-甲基吡咯烷-2-基)甲基)-2-氧代-2,5,6,8-四氢吡啶并[3,4-d]嘧啶-7(1H)-甲酸叔丁酯(1-4)
100mL封管反应器中加入1-3(3.2g,6.74mmol),Cs 2CO 3(4.4g,13.48mmol),(S)-2-(溴甲基)-1-甲基吡咯烷(1.8g,10.1mmol),CuI(257mg,1.35mmol)和DMSO(30mL),密封后升至100℃搅拌反应20h。体系降至室温,加入饱和氯化铵溶液(30mL)淬灭,室温搅拌30min后加EA(50mL*2)萃取,合并有机相用饱和氯化钠溶液(50mL*2)洗涤,浓缩,残留物柱层析(DCM/MeOH=30/0 to 20/1)纯化得黄棕色固体1-4(2.1g,收率55%),ESI-MS m/z:567.3[M+H] +
(S)-4-(1-((1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2,5,6,7,8-六氢吡啶并[3,4-d]嘧啶-4基)哌 嗪-1-甲酸苄酯(1-5)
250mL单口瓶中加入1-4(4.66g,8.22mmol),DCM(20mL)和HCl/Dioxane溶液(4M,21mL,82mmol),混合液室温搅拌反应4h。LC-MS监测反应完全后,体系浓缩,残留物加入DCM(50mL),碳酸氢钠溶液(50mL),室温搅拌30min后分液,水相再用DCM(50mL)萃取,合并有机相用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,浓缩至干得黄棕色油状1-5(3.96g,收率100%),ESI-MS m/z:467.3[M+H] +
4-(7-(5-甲基-1-(四氢-2H-吡喃-2-基)-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2,5,6,7,8-六氢吡啶并[3,4-d]嘧啶-4基)哌嗪-1-甲酸苄酯(1-6)
100mL单口瓶中加入1-5(379mg,0.812mmol),4-溴-5-甲基-1-(四氢-2H-吡喃-2-基)-1H-吲唑(311mg,1.056mmol),叔丁醇钠(195mg,2.03mmol),Ruphos-Pd-G3(134mg,0.16mmol)和Dioxane(20mL),Ar置换保护后升温至回流搅拌反应20h。LC-MS监测反应完成后,体系加水淬灭,EA(20mL*2)萃取,有机相用饱和氯化钠洗涤,浓干,pre-TLC制备纯化(DCM/MeOH/NH 4OH=20/1/0.02)得淡棕色油状物1-6(100mg,收率18%),ESI-MS m/z:481.3[M+H] +
7-(5-甲基-1-(四氢-2H-吡喃-2-基)-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-4-(哌嗪-1-基)-5,6,7,8-四氢吡啶并[3,4-d]嘧啶-2(1H)-酮(1-7)
100mL单口瓶中加入1-6(100mg,0.147mmol),MeOH(10mL)和10%Pd/C(20mg,wet%=50%),H 2置换三次后室温常压下搅拌20h。LC-MS监测反应完成后,体系过滤,滤液浓缩至干,得黄色固体1-7(80mg,收率100%),ESI-MS m/z:547.2[M+H] +
4-(4-丙烯酰基哌嗪-1-基)-7-(5-甲基-1-(四氢-2H-吡喃-2-基)-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-4-(哌嗪-1-基)-5,6,7,8-四氢吡啶并[3,4-d]嘧啶-2(1H)-酮(1-8)
50mL单口瓶中加入1-7(80mg,0.147mmol),DCM(5mL),DIPEA(38mg,0.294mmol),Ar保护下冰浴降温至0℃,后滴加丙烯酰氯(16mg,0.177mmol)的DCM溶液,滴毕,体系0-5℃搅拌1h。LC-MS监测反应完全后,加水淬灭,分液,水相用DCM(10mL)萃取,合并有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,浓干得淡黄色固体1-8(60mg,收率68%),ESI-MS m/z:601.2[M+H] +
(S)-4-(4-丙烯酰哌嗪-1-基)-7-(5-甲基-1H-吲唑-4-基)-1-((1-甲基吡咯烷-2-基)甲基)-5,6,7,8-四氢吡啶并[3,4-d]嘧啶-2(1H)-酮(1)
50mL单口瓶中加入1-8(60mg,0.1mmol),DCM(5mL),TFA(114mg,1mmol),Ar保护下室温搅拌2h,LC-MS监测反应完全后,体系浓缩至干,残留物pre-TLC纯化(DCM/MeOH/NH 4OH=10/1/0.1)得类白色固体化合物1(25mg,收率48%)。
1H NMR(400MHz,CDCl 3)δ:8.03(s,1H),7.17-7.23(m,2H),6.53-6.63(m,1H),6.32(dd,J=16.9,1.9Hz,1H),5.73(ddd,J=10.5,3.2,1.9Hz,1H),4.46(d,J=11.9Hz,1H),4.28(s,2H),3.74(d,J=17.5Hz,5H),3.48(t,J=5.4Hz,7H),2.85(s,2H),2.74(s,3H),2.53(s,1H),2.40(d,J=1.7Hz,3H),2.21(d,J=24.4Hz,2H),1.86-2.10(m,3H);ESI-MS m/z:517.3[M+H] +
实施例2 4-(4-丙烯酰基哌嗪-1-基)-6-环丙基-8-甲氧基-7-(5-甲基-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)喹唑啉-2(1H)-酮(化合物53)的合成
Figure PCTCN2020097397-appb-000064
化合物53根据如下文所述的方法B制备:
4-(7-溴-2,6-二氯-8-氟喹唑啉-4-基)哌嗪-1-甲酸叔丁酯(53-1)
250mL单口瓶中加入B1(5g,15.13mmol),DIPEA(4g,30.3mmol),N-Boc-哌嗪(2.96g,15.89mmol)和DMF(100mL),Ar保护下,升温至80℃反应2小时。TLC点板(PE/EA=5/1),原料反应完全,将反应液冷却至室温后,加入水(100mL),用EA(100mL*2)萃取,合并有机相,饱和氯化钠洗涤,有机相浓缩,柱层析纯化(PE/EA=1:0 to 2/1)得白色固体53-1(5.23g,收率72%),ESI-MS m/z:479.1/481.1[M+H] +
4-(7-溴-6-氯-8-氟-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-1-甲酸叔丁酯(53-2)
250mL单口瓶中加入53-1(5.23g,10.89mmol)和THF(100mL),然后加入氢氧化钠溶液(2N,10.5mL,21mmol),室温反应过夜。TLC监测(PE/EA=1/1),原料反应完全,用 2N HCl调pH至7,加EA(50mL*2)萃取,合并有机相用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,浓缩,粗品加入EA(5mL)/PE(20mL)室温打浆,过滤得白色固53-2(2.61g,收率52%),ESI-MS m/z:462.1/463.1[M+H] +
4-(7-溴-6-氯-8-甲氧基-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-1-甲酸叔丁酯(53-3)
250ml三口瓶中加入DMF(40ml),甲醇钠(485mg,8.66mmol),Ar置换保护下降温至0-5℃,后滴加53-2(2g,4.33mmol)的DMF(10mL)溶液,滴毕,体系升至室温搅拌反应。LC-MS监测反应完成后,体系加水(50mL)淬灭,1N HCl调pH至中性,EA(50mL*2)萃取,合并有机相用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,浓缩,残留物加入EA(5mL)/PE(20mL)室温打浆,过滤得白色固体53-3(1.68g,82%),ESI-MS m/z:473.1/475.1[M+H] +
(S)-4-(7-溴-6-氯-8-甲氧基-1-((1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-1-甲酸叔丁酯(53-4)
100mL封管反应器中加入53-3(840mg,1.773mmol),Cs 2CO 3(1.72g,5.32mmol),(S)-2-(氯甲基)-1-甲基吡咯烷盐酸盐(600mg,3.55mmol),CuI(67mg,0.355mmol)和DMSO(20mL),密封后升至100℃搅拌反应20h。LC-MS监测反应完成后,体系降至室温,加入饱和氯化铵溶液(20mL)淬灭,室温搅拌30min后加EA(40mL*2)萃取,合并有机相用饱和氯化钠溶液(40mL*2)洗涤,浓缩,残留物柱层析(DCM/MeOH=50/0 to 20/1)纯化得类白色固体53-4(638mg,收率63%),ESI-MS m/z:570.0/572.0[M+H] +
4-(6-氯-8-甲氧基-7-(5-甲基-1-(四氢-2H-吡喃-2-基)-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-1-甲酸叔丁酯(53-5)
100mL单口瓶中加入53-4(638mg,1.118mmol),4-溴-5-甲基-1-(四氢-2H-吡喃-2-基)-1H-吲唑(428mg,1.453mmol),叔丁醇钠(268mg,2.79mmol),Ruphos-Pd-G3(184mg,0.22mmol)和Dioxane(20mL),Ar置换保护后升温至回流搅拌反应20h。LC-MS监测反应完成后,体系加水淬灭,EA(20mL*2)萃取,有机相用饱和氯化钠洗涤,浓干,pre-TLC制备纯化(DCM/MeOH/NH 4OH=20/1/0.02)得淡棕色油状物53-5(371mg,收率47%),ESI-MS m/z:706.2[M+H] +
4-(6-环丙基-8-甲氧基-7-(5-甲基-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-1-甲酸叔丁酯(53-6)
100mL单口瓶中加入53-5(371mg,0.525mmol),环丙基硼酸(68mg,0.788mmol),无水磷酸钾(278mg,1.313mmol)和Dioxane(10mL),Ar置换保护后加入Pd(dppf) 2Cl 2(58mg,0.1mmol),后升温至回流反应20h。LC-MS监测反应完成后,混合液浓缩至干, 残留物过反相Flash制备纯化得53-6(269mg,收率72%),ESI-MS m/z:712.2[M+H] +
6-环丙基-8-甲氧基-7-(5-甲基-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-4-(哌嗪-1-基)喹唑啉-2(1H)-酮(53-7)
50mL单口瓶中加入53-6(269mg,0.378mmol),DCM(10mL)和HCl/Dioxane溶液(4M,2mL,8mmol),混合液室温搅拌反应20h。LC-MS监测反应完全后,体系浓缩至干得黄棕色固体53-7(253mg,收率100%),ESI-MS m/z:527.2[M+H] +
4-(4-丙烯酰基哌嗪-1-基)-6-环丙基-8-甲氧基-7-(5-甲基-1H-吲唑-4-基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)喹唑啉-2(1H)-酮(53)
25mL单口瓶中加入53-7(25mg,0.039mmol),DCM(5mL),DIPEA(25mg,0.196mmol),Ar保护下,冰浴降温至0℃,后滴加丙烯酰氯(2.8mg,0.031mmol)的DCM溶液,滴毕,体系0-5℃搅拌1h。LC-MS监测反应剩余少量原料,加水淬灭,分液,水相用DCM(10mL)萃取,合并有机相,浓干,粗品pre-TLC(DCM/MeOH/NH 4OH=20/1/0.02)纯化得化合物53(8mg,收率35%)。
1H NMR(400MHz,CDCl 3)δ:8.05(s,1H),7.15-7.20(m,2H),7.09(s,1H),6.47-6.61(m,1H),6.32(dd,J=16.9,1.9Hz,1H),5.73(ddd,J=10.5,3.2,1.9Hz,1H),4.42(s,3H),3.74(m,6H),3.48(t,J=5.4Hz,4H),2.55(s,3H),2.44(s,3H),2.40(d,J=1.7Hz,3H),1.86-2.10(m,5H),0.30-0.51(m,4H);ESI-MS m/z:582.3[M+H] +
实施例3 4-((S)-4-丙烯酰基-2-甲基哌嗪-1-基)-6-氟-7-(2-氟-6-羟基苯基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)吡啶并[2,3-d]嘧啶-2(1H)-酮(化合物74)的合成
Figure PCTCN2020097397-appb-000065
化合物74根据如下文所述的方法C制备:
2,6-二氯-5-氟烟酰胺(74-1)
将CDI(35.6g,220mmol)分批加入到C1(42g,200mmol)的THF(400mL)溶液中,混合物搅拌5min,Ar保护,加热到50℃,反应1h,LC-MS监测,原料消失,反应液用甲苯(100mL)稀释,浓缩到初始体积的一半,将得到的混合物冷却到0℃,缓慢加入氢氧化铵(55mL,400mmol)。室温反应10min,EA稀释(200mL),水洗(100mL*3)。有机层用无水Na 2SO 4干燥,旋干。PE/EA(10/1,200mL)打浆,过滤,将剩余母液浓缩至初始体积的一半,冷却至0℃,再次析出固体,过滤。合并两批固体,得到浅黄色固体产物74-1(22.10g,产率53%),初产物不经纯化直接用于下一步反应,ESI-MS m/z:208.9[M+H] +
(S)-2,6-二氯-5-氟-N-(((1-甲基吡咯啉-2-基)甲基)氨基甲酰基)烟酰胺(74-2)
将74-1(5.2g,25mmol)溶于THF(50mL)中,氩气保护下冷却至-78℃,通过注射器缓慢加入草酸氯(2M溶液DCM,13mL,26mmol),加毕,反应液升温60℃反应3.5h,停止加热,冷却至-78℃,加入TEA(7.6g,75mmol),(S)-(1-甲基吡咯啉-2-基)甲胺(2.85g,25mmol),加毕,升至室温搅拌1小时,加水(120mL)淬灭反应,EA萃取,合并有机相,有机相用无水Na 2SO 4干燥,浓缩。柱层析(DCM/MeOH=100/1to 20/1),得到浅黄色固体74-2(5.20g,收率60%),ESI-MS m/z:349.1[M+H] +
(S)-7-氯-6-氟-1-((1-甲基吡咯啉-2-基)甲基)吡啶并[2,3-d]嘧啶-2,4(1H,3H)-二酮(74-3)
将74-2(3.5g,10mmol)溶于THF(100mL)中,氩气保护下冰浴,缓慢加入KHMDS(1M THF溶液,20mL),加毕,搅拌10min,撤冰浴,室温下反应30min。饱和氯化铵(125mL)淬灭反应,用EA(250mL)萃取。有机相用饱和食盐水洗,无水Na 2SO 4干燥,浓缩。柱层析(DCM/MeOH=100/1to 20/1),得到浅黄色固体74-3(1.71g,产率),ESI-MS m/z:313.1[M+H] +
(S)-4,7-二氯-6-氟-1-((1-甲基吡咯啉-2-基)甲基)吡啶并[2,3-d]嘧啶-2(1H)-酮(74-4)
将74-3(1.56g,5.0mmol)溶于乙腈(10mL)中,加入DIPEA(1.9g,15mmol),POCl 3(920mg,6.0mmol),升温80℃搅拌反应30min。LC-MS监测,反应完毕,浓缩,直接投入下一步反应,ESI-MS m/z:331.1[M+H] +
(S)-4-(7-氯-6-氟-1-(((S)-1-甲基吡咯啉-2-基)甲基)-2-氧代-1,2-二氢吡啶并[2,3-d]嘧啶-4-基)-3-甲基哌嗪-1-甲酸叔丁酯(74-5)
将上步所得74-4(理论量5.0mmol)溶于DMF(15mL)中,加入DIPEA(1.9g,15mmol),(S)-3-甲基哌嗪-1-甲酸叔丁酯(1.2g,6.0mmol),室温反应30min钟,LC-MS监测,基本 反应完毕,加EA稀释,碳酸氢钠水溶液洗,干燥,浓缩,柱层析(DCM/MeOH=100/1to20/1),得到到浅黄色固体74-5(2.01g,收率20%),ESI-MS m/z:495.2[M+H] +
(S)-4-(7-(2-羟基-6-甲基苯基)-6-氟-1-(((S)-1-甲基吡咯啉-2-基)甲基)-2-氧代-1,2-二氢吡啶并[2,3-d]嘧啶-4-基)-3-甲基哌嗪-1-甲酸叔丁酯(74-6)
将74-5(1.2g,3.0mmol)、Pd(dppf)Cl 2(220mg,0.3mmol)、AcOK(2.36g,24mmol)溶于1,4-dioxane(50mL)中,氩气保护下,升温90℃反应3h,LC-MS监测反应完毕,直接浓缩,柱层析(DCM/MeOH=100/1to 20/1),得到到浅黄色固体74-6(1.10g,收率65%),ESI-MS m/z:567.3[M+H] +
4-((S)-4-丙烯酰基-2-甲基哌嗪-1-基)-6-氟-7-(2-氟-6-羟基苯基)-1-(((S)-1-甲基吡咯啉-2-基)甲基)吡啶并[2,3-d]嘧啶-2(1H)-酮(74)
将74-6(566mg,1.0mmol)溶于TFA(10mL)中,室温反应30min,LC-Ms监测,反应完毕,直接浓缩。残留物溶于DCM,加入DIPEA(650mg,5.0mmol),冰浴,滴加丙烯酰氯(100mg,1.1mmol),加毕,室温反应1h,LC-MS监测,反应完毕,反应液用DCM稀释,用水洗,浓缩,柱层析(DCM/MeOH=100/1to 20/1),得到到浅黄色固体74(320mg,收率61%)。
1H NMR(400MHz,CDCl 3)δ:7.92(s,1H),7.41-7.56(m,3H),6.41-6.55(m,1H),6.31(dd,J=16.9,1.9Hz,1H),5.73(ddd,J=10.5,3.2,1.9Hz,1H),3.74(m,5H),3.48(t,J=5.4Hz,4H),2.45(s,3H),2.40(m,3H),1.86-2.10(m,5H),1.52(d,J=5.2Hz,2H);ESI-MS m/z:525.2[M+H] +
实施例4 2-((2S)-1-丙烯酰-4-(6-氯-7-(2-氟-6-羟基苯基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-2-基)乙腈(化合物107)的合成:
Figure PCTCN2020097397-appb-000066
化合物107根据如下文所述的方法D制备:
(S)-4-(7-溴-2,6-二氯喹唑啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸叔丁酯(107-1)
往250mL单口瓶中加入7-溴-2,4,6-三氯喹唑啉(4.68g,15.0mmol),DIPEA(3.87g,30mmol),(S)-2-(氰基甲基)哌嗪-1-羧酸叔丁酯(3.55g,15.75mmol)和DMF(100mL),Ar保护下,升温至60℃反应20小时。TLC监测(PE/EA=10/1),原料反应完全,将反应液冷却至室温后,加入水(200mL),用EA(100mL*2)萃取,合并有机相,饱和氯化钠洗,有机相浓缩,柱层析纯化(PE/EA=1/0 to 2/1)得白色固体107-1(5.26g,收率70%),ESI-MS m/z:500.2/502.2[M+H] +
(S)-4-(7-溴-6-氯-2-氧代-1,2-二氢喹唑啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸叔丁酯(107-2)
往250mL单口瓶中加入107-1(5.26g,10.5mmol)和1,4-二氧六环(100mL),然后加入氢氧化钠溶液(51mL,2N),室温反应过夜,TLC监测(PE/EA=1/1),原料反应完全,用2N盐酸调pH至7,析出固体,过滤得白色固体107-2;滤液再用EA(50mL)萃取,饱和氯化钠溶液洗,无水硫酸钠干燥,过滤,浓缩,粗品加入EA(10mL)打浆搅拌,过滤得白色固体1-2,合并得到1-2(3.29g,收率65%),ESI-MS m/z:482.1/484.1[M+H] +
(S)-4-(7-溴-6-氯-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基叔丁基基)-2-(氰基甲基)哌嗪-1-羧酸叔丁酯(107-3)
100mL封管反应器中加入107-2(3.25g,6.74mmol),Cs 2CO 3(4.4g,13.48mmol),(S)-2-(溴甲基)-1-甲基吡咯烷(1.8g,10.1mmol),CuI(257mg,1.35mmol)和DMSO(30mL),密封后升至100℃搅拌反应20h。体系降至室温,加入饱和氯化铵溶液(30mL)淬灭,室温搅拌30min后加EA(50mL*2)萃取,合并有机相用饱和氯化钠溶液(50mL*2)洗涤,浓缩,残留物柱层析(DCM/MeOH=30/0 to 20/1)纯化得黄棕色固体107-3(2.58g,收率66%),ESI-MS m/z:579.1/581.1[M+H] +
(2S)-4-(6-氯-7-(2-氟-6-((四氢-2H-吡喃-2-基)氧基)苯基)-1-(((S)-1-(叔丁基甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基)-2-(氰基甲基)哌嗪-1-羧酸叔丁酯(107-4)
100mL单口瓶中加入107-3(565mg,0.974mmol),2-(2-氟-6-((四氢-2H-吡喃-2-基)氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(376mg,1.17mmol),乙酸钾(239mg,2,436mmol),Pd(dppf) 2Cl 2(81mg,0.1mmol)和Dioxane(20mL),H 2O(5ml),Ar置换保护后升温至回流搅拌反应6h。LC-MS监测反应完成后,体系加水淬灭,EA(20mL*2)萃取,有机相用饱和氯化钠洗涤,浓干,柱层析(DCM/MeOH=20/1)得淡棕色油状物107-4(508mg,收率75%),ESI-MS m/z:695.1[M+H] +
2-((2S)-4-(6-氯-7-(2-氟-6-羟基苯基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢 喹唑啉-4-基)哌嗪-2-基)乙腈(107-5)
50mL单口瓶中加入107-4(500mg,0.719mmol),EA(10mL),HCl/Dioxane(3.6mL,4M,14.4mmol),Ar保护下室温搅拌2h,LC-MS监测反应完全后,体系浓缩至干得浅棕色固体107-5(370mg,收率100%)。ESI-MS m/z:511.1[M+H] +
2-((2S)-1-丙烯酰-4-(6-氯-7-(2-氟-6-羟基苯基)-1-(((S)-1-甲基吡咯烷-2-基)甲基)-2-氧代-1,2-二氢喹唑啉-4-基)哌嗪-2-基)乙腈(化合物107)
25mL单口瓶中加入107-5(51mg,0.1mmol),DCM(5mL),DIPEA(65mg,0.5mmol),Ar保护下冰浴降温至0℃,后滴加丙烯酰氯(10mg,0.11mmol)的DCM溶液,滴毕,体系0-5℃搅拌1h。LC-MS监测反应完全后,加水淬灭,分液,水相用DCM(10mL)萃取,合并有机相用饱和氯化钠溶液(10mL)洗涤,浓干,残留物用pre-TLC纯化(DCM/MeOH/NH 4OH=20/1/0.1)得类白色固体化合物107(24mg,收率42.5%)。
1H NMR(400MHz,CDCl 3)δ:9.82(s,1H),8.33(s,1H),7.93(s,1H),7.37-7.25(m,3H),6.53-6.63(m,1H),6.32(m,1H),5.73(m,1H),3.44(m,7H),3.24(m,2H),2.69(s,3H),2.58(m,3H),2.42(m,2H),1.86-2.10(m,4H);ESI-MS m/z:565.2[M+H] +
实施例5化合物对H358细胞内Ras-GTP含量的检测
一百万H358细胞种植于超低吸附的六孔板中,生长一天后,加入待测化合物(浓度为1μM),化合物作用24小时后,裂解细胞后,加入GST-Raf1(1-149)(millipore 14-863),4度孵育过夜后,加入GST beads(millipore,G0924),4度孵育2个小时,然后离心,取beads,beads用IP buffer洗3次后,然后加入SDS裂解液,跑胶,用Ras抗体(CST,3339)western blot检测拉下的Ras-GTP含量。与DMSO组对比,计算化合物抑制Ras-GTP活性的百分比,结果见下列表2。
表2.本发明化合物对H358细胞内Ras-GTP的抑制活性
化合物 抑制率(%) 化合物 抑制率(%) 化合物 抑制率(%)
1 +++ 2 ++ 3 ++
4 ++ 5 ++ 6 ++
7 ++ 8 + 9 ++
10 +++ 11 +++ 12 +++
13 +++ 14 +++ 15 +++
16 +++ 17 +++ 18 +++
19 +++ 20 ++ 21 +++
22 +++ 23 +++ 24 +++
25 +++ 26 +++ 27 +++
28 +++ 29 +++ 30 ++
31 +++ 32 +++ 33 +++
34 +++ 35 +++ 36 ++
37 +++ 38 +++ 39 +++
40 +++ 41 +++ 42 +++
43 +++ 44 +++ 45 +++
46 +++ 47 +++ 48 +++
49 +++ 50 +++ 51 +++
52 ++ 53 ++ 54 +++
55 +++ 56 +++ 57 +++
58 +++ 59 ++ 60 +++
61 +++ 62 +++ 63 +++
64 +++ 65 +++ 66 +++
67 +++ 68 +++ 69 +++
70 +++ 71 +++ 72 +++
73 +++ 74 +++ 75 +++
76 +++ 77 +++ 78 +++
79 +++ 80 ++ 81 +++
82 +++ 83 +++ 84 ++
85 ++ 86 +++ 87 ++
88 ++ 89 +++ 90 +++
91 +++ 92 +++ 93 +++
94 +++ 95 +++ 96 +++
97 +++ 98 ++ 99 +++
100 ++ 101 ++ 102 +++
103 +++ 104 +++ 105 +++
106 +++ 107 +++ 108 +++
109 +++ 110 +++ 111 +++
112 +++
+表示抑制率至多50%
++表示抑制率为50%至90%
+++表示抑制率大于90%
实施例6化合物对H358细胞的抗增殖活性
2500个H358细胞种植于超低吸附的96孔板(corning,7007)中,生长三天后,加入梯度稀释化合物(最高30uM,5倍稀释,一共五个剂量),加入化合物四天后,加入Cell Titer Glow(Promega,G9681)评价小球的生长情况,计算IC 50值,结果见下列表3。
表3.本发明化合物对H358细胞的抗增殖活性
化合物 抑制率(%) 化合物 抑制率(%) 化合物 抑制率(%)
1 +++ 2 ++ 3 ++
4 + 5 ++ 6 ++
7 ++ 8 + 9 ++
10 +++ 11 +++ 12 +++
13 +++ 14 +++ 15 +++
16 +++ 17 +++ 18 +++
19 ++ 20 +++ 21 +++
22 +++ 23 +++ 24 +++
25 +++ 26 +++ 27 +++
28 +++ 29 +++ 30 ++
31 +++ 32 +++ 33 +++
34 +++ 35 +++ 36 +++
37 ++ 38 +++ 39 +++
40 +++ 41 +++ 42 +++
43 +++ 44 +++ 45 +++
46 +++ 47 +++ 48 +++
49 +++ 50 +++ 51 ++
52 ++ 53 +++ 54 +++
55 +++ 56 +++ 57 +++
58 +++ 59 ++ 60 +++
61 +++ 62 +++ 63 +++
64 +++ 65 +++ 66 +++
67 +++ 68 +++ 69 +++
70 +++ 71 +++ 72 +++
73 +++ 74 +++ 75 ++
76 +++ 77 +++ 78 +++
79 +++ 80 +++ 81 +++
82 +++ 83 +++ 84 ++
85 ++ 86 +++ 87 +++
88 ++ 89 +++ 90 ++
91 + 92 ++ 93 +++
94 +++ 95 +++ 96 +++
97 +++ 98 +++ 99 +++
100 +++ 101 ++ 102 +++
103 +++ 1104 +++ 105 +++
106 +++ 107 +++ 108 +++
109 +++ 110 +++ 111 +++
112 +++
+表示大于30μM
++表示抑制率为1至30μM
+++表示抑制率小于1μM
由上表2和表3数据可知,本发明化合物具有较好的生物活性,包括H358细胞内Ras-GTP的抑制活性和H358细胞抗增殖活性。
虽然以上描述了本发明的具体实施方式,但是本领域的技术人员应当理解,这些仅是举例说明,在不背离本发明的原理和实质的前提下,可以对这些实施方式做出多种变更或修改。因此,本发明的保护范围由所附权利要求书限定。

Claims (22)

  1. 一种结构如通式(1)所示的化合物或其各光学异构体、各晶型、药学上可接受的盐、水合物或溶剂合物:
    Figure PCTCN2020097397-appb-100001
    式(1)中:
    m为0、1或2;
    A为一个二价含有1-2个N原子的4-12元饱和或部分饱和的单环、双环、桥环或螺环,所述单环、双环、桥环或螺环可任选被一个或多个R 4所取代,当被多个R 4取代时,R 4可以相同或不同;
    Y为化学键或C1-C6亚烷基;
    R 1为芳基或杂芳基,所述芳基或杂芳基可被1-3个下述基团所取代:卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基;
    R 2为胺基烷基、环烷基、烷基取代酰胺基、杂环基、芳基或杂芳基,所述杂环基、芳基或杂芳基可任选被一个或多个R 5所取代,当被多个R 5取代时,R 5可以相同或不同;
    R 3独立选择C1-C3烷基或卤代C1-C3烷基;
    R 4独立选自H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基;
    R 5独立选自卤素、H、O、CN、OH、烷基羟基、二烷基胺基、C1-C6烷基、C3-C6环烷基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基;
    E是能够与K-Ras、H-Ras或N-Ras突变体蛋白的12位的半胱氨酸残基形成共价键的亲电部分。
  2. 如权利要求1所述的化合物,其中所述式(1)中,E为一个含亲电碳碳双键或碳碳叁键的基团。
  3. 如权利要求2所述的化合物,其中所述式(1)中,E为:
    Figure PCTCN2020097397-appb-100002
    其中,R a为H或F,R b为H、-CH 2F、-CHF 2
    Figure PCTCN2020097397-appb-100003
    Figure PCTCN2020097397-appb-100004
  4. 如权利要求1所述的化合物,其中所述式(1)中,A-E为:
    Figure PCTCN2020097397-appb-100005
    Figure PCTCN2020097397-appb-100006
    其中,n为1或2,R 4为H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基。
  5. 如权利要求1所述的化合物,其中所述式(1)中,Y为化学键、-CH 2-、-CH(Me)-或-CH 2CH 2-。
  6. 如权利要求1所述的化合物,其中所述式(1)中,R 1为:
    Figure PCTCN2020097397-appb-100007
    Figure PCTCN2020097397-appb-100008
    其中R c和R d独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基。
  7. 如权利要求1所述的化合物,其中所述式(1)中,R 2为:
    Figure PCTCN2020097397-appb-100009
    Figure PCTCN2020097397-appb-100010
    Figure PCTCN2020097397-appb-100011
    其中n为1或2,R e和R f独立地为H、卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基,R g为C1-C3烷基、C3-C6环烷基、C3-C6 环烷基烷基、C1-C3烷氧基烷基、卤素取代C1-C3烷基、卤素取代C3-C6环烷基或
    Figure PCTCN2020097397-appb-100012
  8. 如权利要求1-7所述的化合物,或其一种药学上可接受的盐,其中所述化合物具有以下结构之一:
    Figure PCTCN2020097397-appb-100013
    Figure PCTCN2020097397-appb-100014
    Figure PCTCN2020097397-appb-100015
  9. 一种结构如通式(2)所示的化合物或其各光学异构体、各晶型、药学上可接受的盐、水合物或溶剂合物:
    Figure PCTCN2020097397-appb-100016
    式(2)中:
    A为一个二价含有1-2个N原子的4-12元饱和或部分饱和的单环、双环、桥环或螺环,所述单环、双环、桥环或螺环可任选被一个或多个R 4所取代,当被多个R 4取代时,R 4可以相同或不同;
    Y为化学键或C1-C6亚烷基;
    W为N、C-R 8或C-O-R 6
    其中,当W为C-O-R 6时,R 2为胺基烷基、环烷基、烷基取代酰胺基、杂环基、芳基或杂芳基,所述杂环基、芳基或杂芳基可任选被一个或多个R 5所取代,当被多个R 5取代时,R 5可以相同或不同;
    其中,当W为N或C-R 8时,R 2为胺基烷基、烷基取代酰胺基或杂环基,所述杂环基可任选被一个或多个R 5所取代,当被多个R 5取代时,R 5可以相同或不同;
    R 1为芳基或杂芳基,所述芳基或杂芳基可被1-3个下述基团所取代:卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基、卤素取代C1-C3烷氧基;
    R 4独立选自H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基;
    R 5独立选自卤素、H、O、CN、OH、烷基羟基、二烷基胺基、C1-C6烷基、C3-C6环烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基;
    R 6为C1-C3烷基、卤素取代C1-C3烷基或C3-C6环烷基;
    R 7为H、卤素、C1-C3烷基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基、卤素取代C1-C3烷氧基或C2-C4烯基;
    R 8为H、卤素、C1-C3烷基、C3-C6环烷基或卤素取代C1-C3烷基;
    E是能够与K-Ras、H-Ras或N-Ras突变体蛋白的12位的半胱氨酸残基形成共价键的亲电部分。
  10. 如权利要求9所述的化合物,其中所述式(2)中,E为一个含亲电碳碳双键或碳碳叁键的基团。
  11. 如权利要求10所述的化合物,其中所述式(2)中,E为:
    Figure PCTCN2020097397-appb-100017
    其中,R a为H或F,R b为H、-CH 2F、-CHF 2
    Figure PCTCN2020097397-appb-100018
    Figure PCTCN2020097397-appb-100019
  12. 如权利要求9所述的化合物,其中所述式(2)中,A-E为:
    Figure PCTCN2020097397-appb-100020
    Figure PCTCN2020097397-appb-100021
    其中,n为1或2,其中,R 4为H、CN、C1-C3烷基、卤素取代C1-C3烷基或氰基取代C1-C3烷基。
  13. 如权利要求9所述的化合物,其中所述式(2)中,Y为化学键、-CH 2-、-CH(Me)-或-CH 2CH 2-。
  14. 如权利要求9所述的化合物,其中所述式(2)中,R 1为:
    Figure PCTCN2020097397-appb-100022
    Figure PCTCN2020097397-appb-100023
    其中R c和R d独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基。
  15. 如权利要求9所述的化合物,其中所述式(2)中,W为C-O-R 6,R 2为:
    Figure PCTCN2020097397-appb-100024
    Figure PCTCN2020097397-appb-100025
    Figure PCTCN2020097397-appb-100026
    Figure PCTCN2020097397-appb-100027
    其中n为1或2,R e和R f独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基、或卤素取代C1-C3烷氧基,R g为C1-C3烷基、C3-C6环烷基、C3-C6环烷基烷基、C1-C3烷氧基烷基、卤素取代C1-C3烷基、卤素取代C3-C6环烷基或
    Figure PCTCN2020097397-appb-100028
  16. 如权利要求9所述的化合物,其中所述式(2)中,W为N或C-R 8,R 2为:
    Figure PCTCN2020097397-appb-100029
    Figure PCTCN2020097397-appb-100030
    Figure PCTCN2020097397-appb-100031
    其中n为1或2,R e和R f独立地为卤素、羟基、氨基、C1-C3烷基、C2-C4烯基、C3-C6环烷基、C1-C3烷氧基、卤素取代C1-C3烷基或卤素取代C1-C3烷氧基,R g为C1-C3烷基、C3-C6环烷基、C3-C6环烷基烷基、C1-C3烷氧基烷基、卤素取代C1-C3烷基、卤素取代C3-C6环烷基或
    Figure PCTCN2020097397-appb-100032
  17. 如权利要求9-16所述的化合物,或其一种药学上可接受的盐,其中所述化合物具有以下结构之一:
    Figure PCTCN2020097397-appb-100033
    Figure PCTCN2020097397-appb-100034
    Figure PCTCN2020097397-appb-100035
  18. 一种用于治疗、调节和/或预防与K-RAS G12C、H-RAS G12C或N-RAS G12C突变体蛋白相关的生理学病况相关的疾病的药物组合物,其特征在于,它含有药学上可接受的赋形剂或载体,以及作为活性成分的权利要求1-17任一项所述的化合物、或其各光学异构体、药学上可接受的盐、水合物或溶剂合物。
  19. 如权利要求18所述的药物组合物,其特征在于,所述的组合物为口服剂型。
  20. 如权利要求18所述的药物组合物,其特征在于,所述的组合物为注射剂型。
  21. 一种权利要求1-17任一项所述的化合物、或其各光学异构体、各晶型、药学上可接受的盐、水合物或溶剂合物的用途,其特征是用于治疗有需要的个体由K-RAS G12C、H-RAS G12C或N-RAS G12C突变介导的疾患的方法。
  22. 如权利要求21所述的的方法,其中所述的疾患是癌症,所述癌症是血液癌和实体瘤。
PCT/CN2020/097397 2019-06-26 2020-06-22 Kras-g12c抑制剂 WO2020259432A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080016738.0A CN113544128B (zh) 2019-06-26 2020-06-22 Kras-g12c抑制剂

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910570897.4A CN110256421A (zh) 2019-06-26 2019-06-26 Kras-g12c抑制剂
CN201910570897.4 2019-06-26

Publications (1)

Publication Number Publication Date
WO2020259432A1 true WO2020259432A1 (zh) 2020-12-30

Family

ID=67922629

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/097397 WO2020259432A1 (zh) 2019-06-26 2020-06-22 Kras-g12c抑制剂

Country Status (2)

Country Link
CN (2) CN110256421A (zh)
WO (1) WO2020259432A1 (zh)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021257736A1 (en) 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
WO2022060583A1 (en) 2020-09-03 2022-03-24 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
WO2022222871A1 (en) * 2021-04-21 2022-10-27 Beijing Innocare Pharma Tech Co., Ltd. Heterocyclic compounds as kras g12c inhibitors
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
WO2023031781A1 (en) 2021-09-01 2023-03-09 Novartis Ag Pharmaceutical combinations comprising a tead inhibitor and uses thereof for the treatment of cancers
US11697657B2 (en) 2019-10-28 2023-07-11 Merck Sharp & Dohme Llc Small molecule inhibitors of KRAS G12C mutant

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020113071A1 (en) * 2018-11-29 2020-06-04 Araxes Pharma Llc Compounds and methods of use thereof for treatment of cancer
MX2021014096A (es) 2019-05-21 2022-02-11 Inventisbio Co Ltd Compuestos heterociclicos, metodos de preparacion y usos de estos.
CN112047937B (zh) * 2019-06-06 2023-04-07 劲方医药科技(上海)有限公司 四氢吡啶并[3,4-d]嘧啶-2(1H)-酮类化合物,其制法与医药上的用途
CN110256421A (zh) * 2019-06-26 2019-09-20 微境生物医药科技(上海)有限公司 Kras-g12c抑制剂
WO2021063346A1 (zh) * 2019-09-30 2021-04-08 上海迪诺医药科技有限公司 Kras g12c抑制剂及其应用
CN112851663B (zh) * 2019-11-12 2023-07-18 博瑞生物医药(苏州)股份有限公司 一种并杂环化合物及其用途
TWI760919B (zh) * 2019-11-15 2022-04-11 大陸商四川海思科製藥有限公司 一種嘧啶並環衍生物及其在醫藥上的應用
IL293206A (en) * 2019-11-27 2022-07-01 Turning Point Therapeutics Inc Combination therapy involving diaryl macrocyclic compounds
WO2021120045A1 (en) 2019-12-18 2021-06-24 InventisBio Co., Ltd. Heterocyclic compounds, preparation methods and uses thereof
CN113045565A (zh) * 2019-12-27 2021-06-29 微境生物医药科技(上海)有限公司 新型K-Ras G12C抑制剂
CN112094269B (zh) * 2020-01-01 2021-12-07 上海凌达生物医药有限公司 一类饱和六元环并杂环类化合物、制备方法和用途
WO2021169990A1 (zh) * 2020-02-24 2021-09-02 泰励生物科技(上海)有限公司 用于癌症治疗的kras抑制剂
CN115572278B (zh) * 2022-11-21 2023-09-01 北京志道生物科技有限公司 京尼平衍生物及其制备方法和应用

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018119183A2 (en) * 2016-12-22 2018-06-28 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2018140600A1 (en) * 2017-01-26 2018-08-02 Araxes Pharma Llc Fused hetero-hetero bicyclic compounds and methods of use thereof
WO2018217651A1 (en) * 2017-05-22 2018-11-29 Amgen Inc. Kras g12c inhibitors and methods of using the same
CN110256421A (zh) * 2019-06-26 2019-09-20 微境生物医药科技(上海)有限公司 Kras-g12c抑制剂
WO2019213516A1 (en) * 2018-05-04 2019-11-07 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2020035031A1 (en) * 2018-08-16 2020-02-20 Genentech, Inc. Fused ring compounds
WO2020050890A2 (en) * 2018-06-12 2020-03-12 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2020113071A1 (en) * 2018-11-29 2020-06-04 Araxes Pharma Llc Compounds and methods of use thereof for treatment of cancer

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL293443A (en) * 2017-09-08 2022-07-01 Amgen Inc kras g12c inhibitors and methods of using them
CA3139769A1 (en) * 2019-05-29 2020-12-03 Shiqiang Liu Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
CN112047937B (zh) * 2019-06-06 2023-04-07 劲方医药科技(上海)有限公司 四氢吡啶并[3,4-d]嘧啶-2(1H)-酮类化合物,其制法与医药上的用途

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018119183A2 (en) * 2016-12-22 2018-06-28 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2018140600A1 (en) * 2017-01-26 2018-08-02 Araxes Pharma Llc Fused hetero-hetero bicyclic compounds and methods of use thereof
WO2018217651A1 (en) * 2017-05-22 2018-11-29 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2019213516A1 (en) * 2018-05-04 2019-11-07 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2020050890A2 (en) * 2018-06-12 2020-03-12 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2020035031A1 (en) * 2018-08-16 2020-02-20 Genentech, Inc. Fused ring compounds
WO2020113071A1 (en) * 2018-11-29 2020-06-04 Araxes Pharma Llc Compounds and methods of use thereof for treatment of cancer
CN110256421A (zh) * 2019-06-26 2019-09-20 微境生物医药科技(上海)有限公司 Kras-g12c抑制剂

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11697657B2 (en) 2019-10-28 2023-07-11 Merck Sharp & Dohme Llc Small molecule inhibitors of KRAS G12C mutant
WO2021257736A1 (en) 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
WO2022060583A1 (en) 2020-09-03 2022-03-24 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
WO2022222871A1 (en) * 2021-04-21 2022-10-27 Beijing Innocare Pharma Tech Co., Ltd. Heterocyclic compounds as kras g12c inhibitors
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
WO2023031781A1 (en) 2021-09-01 2023-03-09 Novartis Ag Pharmaceutical combinations comprising a tead inhibitor and uses thereof for the treatment of cancers

Also Published As

Publication number Publication date
CN110256421A (zh) 2019-09-20
CN113544128B (zh) 2023-12-08
CN113544128A (zh) 2021-10-22

Similar Documents

Publication Publication Date Title
WO2020259432A1 (zh) Kras-g12c抑制剂
CN113767103B (zh) 新型螺环类K-Ras G12C抑制剂
WO2021129824A1 (zh) 新型K-Ras G12C抑制剂
CN115335379B (zh) 含螺环的喹唑啉化合物
WO2021129820A1 (zh) 含螺环的喹唑啉化合物
CN111440189B (zh) 稠环嘧啶氨基衍生物、其制备方法、中间体、药物组合物及应用
CN113527293B (zh) Kras g12c突变蛋白抑制剂及其药物组合物、制备方法和用途
WO2021213317A1 (zh) Hpk1抑制剂及其制备方法和用途
WO2020233641A1 (zh) 用作ret激酶抑制剂的化合物及其应用
WO2022188819A1 (zh) Sos1蛋白水解调节剂及其制备方法和应用
WO2022063297A1 (zh) 喹唑啉衍生物及其制备方法和用途
CN113045570A (zh) 含螺环的喹唑啉化合物
WO2022048631A1 (zh) 一种具有抗肿瘤活性的化合物及其用途
JP2023535932A (ja) 三環式のヘテロ環
CN112513041B (zh) 三环化合物
WO2023016562A1 (zh) 多环类化合物及其用途
WO2022171088A1 (zh) 吡唑并[3,4-d]嘧啶-3-酮衍生物
CN105026399A (zh) 作为钾离子通道抑制剂的吡咯并三嗪类化合物
CN113754659A (zh) 含螺环的喹唑啉化合物
JP2009073743A (ja) 新規な縮合環式ピリミジン化合物又はその塩、及びその医薬組成物
CN115322158B (zh) 作为krasg12c蛋白抑制剂的取代喹唑啉类化合物
WO2021244505A1 (zh) 新型吡嗪化合物
WO2024067744A1 (zh) 杂环取代喹唑啉及其制备方法和应用
CN115322158A (zh) 作为krasg12c蛋白抑制剂的取代喹唑啉类化合物
CN117800989A (zh) 一类含环外双键、多取代甲基醚结构的化合物及其应用

Legal Events

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

Ref document number: 20833635

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20833635

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 11/04/2022)

122 Ep: pct application non-entry in european phase

Ref document number: 20833635

Country of ref document: EP

Kind code of ref document: A1