WO2021110169A1 - 作为erk抑制剂的噻唑并内酰胺类化合物及其应用 - Google Patents

作为erk抑制剂的噻唑并内酰胺类化合物及其应用 Download PDF

Info

Publication number
WO2021110169A1
WO2021110169A1 PCT/CN2020/134286 CN2020134286W WO2021110169A1 WO 2021110169 A1 WO2021110169 A1 WO 2021110169A1 CN 2020134286 W CN2020134286 W CN 2020134286W WO 2021110169 A1 WO2021110169 A1 WO 2021110169A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
present
reaction
add
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2020/134286
Other languages
English (en)
French (fr)
Inventor
李翼
刘宁
于涛
吴成德
黎健
陈曙辉
Original Assignee
南京明德新药研发有限公司
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 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to EP20895381.0A priority Critical patent/EP4071155A4/en
Priority to US17/782,814 priority patent/US20230072937A1/en
Priority to KR1020227021797A priority patent/KR20220107249A/ko
Priority to AU2020398022A priority patent/AU2020398022B2/en
Priority to JP2022534190A priority patent/JP7409719B2/ja
Priority to CA3160903A priority patent/CA3160903A1/en
Priority to CN202080084536.XA priority patent/CN114829365B/zh
Publication of WO2021110169A1 publication Critical patent/WO2021110169A1/zh

Links

Images

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
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • the invention relates to a class of thiazololactam compounds and their application in the preparation of drugs for treating ERK-related diseases. Specifically, it relates to a compound represented by formula (III) or a pharmaceutically acceptable salt thereof.
  • Ras/Raf/MEK/ERK pathway is a classic mitogen-activated protein kinase (MAPK) signaling cascade, which participates in the activation of various growth factors, cytokines, mitogens and hormone receptors. Transduction is one of the most important signal transduction pathways that control cell growth, differentiation and survival.
  • MAPK mitogen-activated protein kinase
  • Extracellular regulated protein kinases are the main participants and key downstream nodes of the Ras/Raf/MEK/ERK pathway. They can be found in many human cancers. Excessive activation. As the terminal signal kinase of this pathway, ERK has not been found to have drug resistance mutations. Therefore, drugs targeting ERK kinase are expected to overcome the problem of drug resistance after treatment with upstream target inhibitors and become a more potential therapeutic strategy. But so far, the research on ERK inhibitors is still in the clinical stage, and no ERK inhibitor has been approved for marketing as a drug.
  • ERK Extracellular regulated protein kinases
  • the present invention provides a compound represented by formula (III) or a pharmaceutically acceptable salt thereof,
  • R 1 is selected from H, C 1-3 alkyl and C 3-5 cycloalkyl, wherein the C 1-3 alkyl and C 3-5 cycloalkyl are optionally substituted by 1, 2 or 3 R a replace;
  • R 2 and R 3 are each independently selected from a C 1-3 alkyl group, wherein the C 1-3 alkyl group is optionally substituted with 1, 2 or 3 R b ;
  • R 4 is selected from H, F, Cl, Br, I and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R c ;
  • R 5 is selected from F, Cl, Br, I, and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R e ;
  • n 0, 1 and 2;
  • n is selected from 0, 1 and 2;
  • Ring A is selected from pyrazolyl and tetrahydropyranyl, wherein the pyrazolyl and tetrahydropyranyl are optionally substituted with 1, 2 or 3 Rd ;
  • R a, R b, R c and R e are each independently selected from D, F, Cl, Br, I, OH and OCH 3;
  • R d is selected from F, Cl, Br, I, CH 3 and OCH 3 .
  • R 1 is selected from H, CH 3 and cyclopropyl, CH 3 and the cyclopropyl optionally substituted with 1, 2 or 3 R a, the other variables are as defined in the present invention.
  • R 1 is selected from H, CH 3 , CHF 2 , CD 3 , CH 2 CH 2 OCH 3 and cyclopropyl, and other variables are as defined in the present invention.
  • R 2 and R 3 are each independently selected from optionally CH 3 and CH 2 CH 3 , said CH 3 and CH 2 CH 3 are optionally substituted by 1, 2 or 3 R b , Other variables are as defined in the present invention.
  • R 2 and R 3 are each independently selected from CH 3 , and other variables are as defined in the present invention.
  • R 4 is selected from H, F, Cl, Br, I and CH 3 , and the CH 3 is optionally substituted with 1, 2 or 3 R c , and other variables are as defined in the present invention.
  • R 4 is selected from H, F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • R 5 is selected from F, Cl, Br, I, and CH 3 , and the CH 3 is optionally substituted with 1, 2 or 3 R e , and other variables are as defined in the present invention.
  • R 5 is selected from F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • R 5 is selected from H, F, Cl, Br, I and CH 3, CH 3 optionally substituted with a 2 or 3 R e, the other variables are as defined in the present invention.
  • R 5 is selected from H, F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from Said Optionally substituted by 1, 2 or 3 Rd , other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from Other variables are as defined in the present invention.
  • the present invention provides a compound represented by formula (III) or a pharmaceutically acceptable salt thereof,
  • R 1 is selected from H and C 1-3 alkyl, wherein said C 1-3 alkyl optionally substituted with 1, 2 or 3 R a;
  • R 2 and R 3 are each independently selected from a C 1-3 alkyl group, wherein the C 1-3 alkyl group is optionally substituted with 1, 2 or 3 R b ;
  • R 4 is selected from H, F, Cl, Br, I and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R c ;
  • R 5 is selected from F, Cl, Br, I, and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R e ;
  • n 0, 1 and 2;
  • n is selected from 0, 1 and 2;
  • Ring A is selected from pyrazolyl and tetrahydropyranyl, wherein the pyrazolyl and tetrahydropyranyl are optionally substituted with 1, 2 or 3 Rd ;
  • R a, R b, R c and R e are each independently selected from F, Cl, Br, I and OH;
  • R d is selected from F, Cl, Br, I and CH 3 .
  • R 1 is selected from H and CH 3, CH 3 optionally substituted by the two or three R a, the other variables are as defined in the present invention.
  • R 1 is selected from CH 3 , and other variables are as defined in the present invention.
  • R 2 and R 3 are each independently selected from optionally CH 3 and CH 2 CH 3 , said CH 3 and CH 2 CH 3 are optionally substituted by 1, 2 or 3 R b , Other variables are as defined in the present invention.
  • R 2 and R 3 are each independently selected from CH 3 , and other variables are as defined in the present invention.
  • R 4 is selected from H, F, Cl, Br, I and CH 3 , and the CH 3 is optionally substituted with 1, 2 or 3 R c , and other variables are as defined in the present invention.
  • R 4 is selected from H, F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • R 5 is selected from F, Cl, Br, I, and CH 3 , and the CH 3 is optionally substituted with 1, 2 or 3 R e , and other variables are as defined in the present invention.
  • R 5 is selected from F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • R 5 is selected from H, F, Cl, Br, I and CH 3, CH 3 optionally substituted with a 2 or 3 R e, the other variables are as defined in the present invention.
  • R 5 is selected from H, F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from Said Optionally substituted by 1, 2 or 3 Rd , other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from Other variables are as defined in the present invention.
  • the present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof,
  • R 1 is selected from H and C 1-3 alkyl, wherein said C 1-3 alkyl optionally substituted with 1, 2 or 3 R a;
  • R 2 and R 3 are each independently selected from C 1-3 alkyl, said C 1-3 alkyl optionally substituted with 1, 2 or 3 R b;
  • R 4 is selected from H, F, Cl, Br, I and C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted with 1, 2 or 3 R c ;
  • n 0, 1 and 2;
  • Ring A is selected from pyrazolyl and tetrahydropyranyl, and the pyrazolyl and tetrahydropyranyl are optionally substituted with 1, 2 or 3 R d ;
  • R a , R b and R c are each independently selected from F, Cl, Br, I and OH;
  • R d is selected from F, Cl, Br, I and CH 3 .
  • R 1 is selected from H and CH 3, CH 3 optionally substituted by the two or three R a, the other variables are as defined in the present invention.
  • R 1 is selected from CH 3 , and other variables are as defined in the present invention.
  • R 2 and R 3 are each independently selected from optionally CH 3 and CH 2 CH 3 , said CH 3 and CH 2 CH 3 are optionally substituted by 1, 2 or 3 R b , Other variables are as defined in the present invention.
  • R 2 and R 3 are each independently selected from CH 3 , and other variables are as defined in the present invention.
  • R 4 is selected from H, F, Cl, Br, I and CH 3 , and the CH 3 is optionally substituted with 1, 2 or 3 R c , and other variables are as defined in the present invention.
  • R 4 is selected from H, F, Cl, Br, I and CH 3 , and other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from Said Optionally substituted by 1, 2 or 3 Rd , other variables are as defined in the present invention.
  • the above-mentioned ring A is selected from Other variables are as defined in the present invention.
  • the above-mentioned compound or a pharmaceutically acceptable salt thereof is selected from:
  • R 1 , R 2 , R 3 , R 4 , R 5 and m are as defined in the present invention.
  • the present invention also provides a compound represented by the following formula or a pharmaceutically acceptable salt thereof,
  • the above-mentioned compound or a pharmaceutically acceptable salt thereof is used in the preparation of a medicine for treating ERK-related diseases.
  • the above application is characterized in that the ERK inhibitor-related drug is a drug for the treatment of solid tumors.
  • the compound of the present invention exhibits superior inhibitory activity on ERK2 enzyme; at the same time, it exhibits superior inhibitory activity on HT29 cell proliferation.
  • the compound of the present invention exhibits excellent oral exposure and bioavailability; the compound of the present invention can significantly inhibit tumor growth, and the body weight of the animal is not significantly reduced during the administration process, and the tolerance is good.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms that are within the scope of reliable medical judgment and are suitable for use in contact with human and animal tissues. , Without excessive toxicity, irritation, allergic reactions or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a salt of the compound of the present invention, which is prepared from a compound with specific substituents discovered in the present invention and a relatively non-toxic acid or base.
  • a base addition salt can be obtained by contacting the compound with a sufficient amount of base in a pure solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amine or magnesium salt or similar salts.
  • the acid addition salt can be obtained by contacting the compound with a sufficient amount of acid in a pure solution or a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogen carbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and organic acid salts, the organic acid includes, for example, acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid and methanesulfonic acid; also include salts of amino acids (such as arginine, etc.) , And salts of organic acids such as glucuronic acid. Certain specific compounds of the present invention contain basic and
  • the pharmaceutically acceptable salt of the present invention can be synthesized from the parent compound containing acid or base by conventional chemical methods. In general, such salts are prepared by reacting these compounds in free acid or base form with a stoichiometric amount of appropriate base or acid in water or organic solvent or a mixture of both.
  • the compounds of the present invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers Isomers, (D)-isomers, (L)-isomers, and their racemic mixtures and other mixtures, such as enantiomers or diastereomer-enriched mixtures, all of these mixtures belong to this Within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All these isomers and their mixtures are included in the scope of the present invention.
  • enantiomer or “optical isomer” refers to stereoisomers that are mirror images of each other.
  • cis-trans isomer or “geometric isomer” is caused by the inability to rotate freely because of double bonds or single bonds of ring-forming carbon atoms.
  • diastereomer refers to a stereoisomer in which the molecule has two or more chiral centers and the relationship between the molecules is non-mirror-image relationship.
  • wedge-shaped solid line keys And wedge-shaped dashed key Represents the absolute configuration of a three-dimensional center, with a straight solid line key And straight dashed key Indicates the relative configuration of the three-dimensional center, using wavy lines Represents a wedge-shaped solid line key Or wedge-shaped dashed key Or use wavy lines Represents a straight solid line key Or straight dashed key
  • the term “enriched in one isomer”, “enriched in isomers”, “enriched in one enantiomer” or “enriched in enantiomers” refers to one of the isomers or pairs of
  • the content of the enantiomer is less than 100%, and the content of the isomer or enantiomer is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or 96% or greater, or 97% or greater, or 98% or greater, or 99% or greater, or 99.5% or greater, or 99.6% or greater, or 99.7% or greater, or 99.8% or greater, or greater than or equal 99.9%.
  • the term “isomer excess” or “enantiomeric excess” refers to the difference between the relative percentages of two isomers or two enantiomers. For example, if the content of one isomer or enantiomer is 90%, and the content of the other isomer or enantiomer is 10%, the isomer or enantiomer excess (ee value) is 80% .
  • optically active (R)- and (S)-isomers and D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If you want to obtain an enantiomer of a compound of the present invention, it can be prepared by asymmetric synthesis or derivatization with chiral auxiliary agents, in which the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide pure The desired enantiomer.
  • the molecule when the molecule contains a basic functional group (such as an amino group) or an acidic functional group (such as a carboxyl group), it forms a diastereomeric salt with an appropriate optically active acid or base, and then passes through a conventional method known in the art The diastereoisomers are resolved, and then the pure enantiomers are recovered.
  • the separation of enantiomers and diastereomers is usually accomplished through the use of chromatography, which uses a chiral stationary phase and is optionally combined with chemical derivatization (for example, the formation of amino groups from amines). Formate).
  • the compound of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms constituting the compound.
  • compounds can be labeled with radioisotopes, such as tritium ( 3 H), iodine-125 ( 125 I), or C-14 ( 14 C).
  • deuterium can be substituted for hydrogen to form deuterated drugs.
  • the bond formed by deuterium and carbon is stronger than the bond formed by ordinary hydrogen and carbon.
  • deuterated drugs can reduce toxic side effects and increase drug stability. , Enhance the efficacy, extend the biological half-life of drugs and other advantages. All changes in the isotopic composition of the compounds of the present invention, whether radioactive or not, are included in the scope of the present invention.
  • substituted means that any one or more hydrogen atoms on a specific atom are replaced by substituents, and may include deuterium and hydrogen variants, as long as the valence of the specific atom is normal and the substituted compound is stable of.
  • oxygen it means that two hydrogen atoms are replaced. Oxygen substitution does not occur on aromatic groups.
  • optionally substituted means that it can be substituted or unsubstituted. Unless otherwise specified, the type and number of substituents can be arbitrary on the basis that they can be chemically realized.
  • any variable such as R
  • its definition in each case is independent.
  • the group can optionally be substituted with up to two Rs, and R has independent options in each case.
  • combinations of substituents and/or variants thereof are only permitted if such combinations result in stable compounds.
  • linking group When the number of a linking group is 0, such as -(CRR) 0 -, it means that the linking group is a single bond.
  • the substituent can be bonded with any atom on the ring, for example, a structural unit It means that the substituent R can be substituted at any position on the cyclohexyl or cyclohexadiene.
  • substituents do not indicate which atom is connected to the substituted group, such substituents can be bonded via any atom.
  • a pyridyl group can pass through any one of the pyridine ring as a substituent. The carbon atom is attached to the substituted group.
  • the middle linking group L is -MW-, at this time -MW- can be formed by connecting ring A and ring B in the same direction as the reading order from left to right It can also be formed by connecting ring A and ring B in the opposite direction to the reading order from left to right
  • Combinations of the linking groups, substituents, and/or variants thereof are only permitted if such combinations result in stable compounds.
  • any one or more sites of the group can be connected to other groups through chemical bonds.
  • the connection method of the chemical bond is not positioned, and there is a H atom at the connectable site, when the chemical bond is connected, the number of H atoms at the site will correspondingly decrease with the number of chemical bonds connected to become the corresponding valence number ⁇ The group.
  • the chemical bond between the site and other groups can be a straight solid bond Straight dashed key Or wavy line Said.
  • the straight solid bond in -OCH 3 means that it is connected to other groups through the oxygen atom in the group;
  • the straight dashed bond in indicates that the two ends of the nitrogen atom in the group are connected to other groups;
  • the wavy line in indicates that the phenyl group is connected to other groups through the 1 and 2 carbon atoms;
  • the number of atoms in a ring is generally defined as the number of ring members.
  • “5-7 membered ring” refers to a “ring” in which 5-7 atoms are arranged around.
  • C 1-3 alkyl is used to indicate a linear or branched saturated hydrocarbon group composed of 1 to 3 carbon atoms.
  • the C 1-3 alkyl group includes C 1-2 and C 2-3 alkyl groups, etc.; it can be monovalent (such as methyl), divalent (such as methylene) or multivalent (such as methine) .
  • Examples of C 1-3 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
  • C 3-5 cycloalkyl means a saturated cyclic hydrocarbon group composed of 3 to 5 carbon atoms, which is a monocyclic ring system, and the C 3-5 cycloalkyl includes C 3 -4 and C 4-5 cycloalkyl, etc.; it can be monovalent, divalent or multivalent.
  • Examples of C 3-5 cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl and the like.
  • protecting group includes, but is not limited to, "amino protecting group", “hydroxy protecting group” or “thiol protecting group”.
  • amino protecting group refers to a protecting group suitable for preventing side reactions at the amino nitrogen position.
  • Representative amino protecting groups include, but are not limited to: formyl; acyl, such as alkanoyl (such as acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, such as tert-butoxycarbonyl (Boc) ; Arylmethyloxycarbonyl, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethyloxycarbonyl (Fmoc); arylmethyl, such as benzyl (Bn), trityl (Tr), 1,1-di -(4'-Methoxyphenyl)methyl; silyl groups, such as trimethylsilyl (TMS) and tert-butyldi
  • hydroxy protecting group refers to a protecting group suitable for preventing side reactions of the hydroxyl group.
  • Representative hydroxy protecting groups include but are not limited to: alkyl groups, such as methyl, ethyl, and tert-butyl; acyl groups, such as alkanoyl groups (such as acetyl); arylmethyl groups, such as benzyl (Bn), p-methyl Oxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (diphenylmethyl, DPM); silyl groups such as trimethylsilyl (TMS) and tert-butyl Dimethylsilyl (TBS) and so on.
  • alkyl groups such as methyl, ethyl, and tert-butyl
  • acyl groups such as alkanoyl groups (such as acetyl)
  • arylmethyl groups such as benzyl (Bn), p-methyl Oxybenzyl (P
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and those well known to those skilled in the art Equivalent alternatives, preferred implementations include but are not limited to the embodiments of the present invention.
  • the structure of the compound of the present invention can be confirmed by conventional methods well known to those skilled in the art. If the present invention relates to the absolute configuration of the compound, the absolute configuration can be confirmed by conventional technical means in the art.
  • the single crystal X-ray diffraction method uses the Bruker D8 venture diffractometer to collect the diffraction intensity data of the cultured single crystal.
  • the light source is CuK ⁇ radiation
  • the scanning method After scanning and collecting relevant data, the direct method (Shelxs97) is further used to analyze the crystal structure to confirm the absolute configuration.
  • the solvent used in the present invention is commercially available.
  • aq stands for water; eq stands for equivalent, equivalent; dichloromethane stands for dichloromethane; PE stands for petroleum ether; DMSO stands for dimethyl sulfoxide; EtOAc stands for ethyl acetate; ethanol stands for ethanol; MeOH represents methanol; Cbz represents benzyloxycarbonyl, which is an amine protecting group; BOC represents tert-butoxycarbonyl, which is an amine protecting group; rt represents room temperature; O/N represents overnight; tetrahydrofuran represents tetrahydrofuran; Boc 2 O represents Di-tert-butyl dicarbonate; TFA stands for trifluoroacetic acid; DIPEA stands for diisopropylethylamine; iPrOH stands for 2-propanol; mp stands for melting point.
  • Figure 1 Tumor growth curve of human non-small cell lung cancer H358 model animals after administration of solvent and WX006.
  • Figure 2 The body weight change rate (%) of human non-small cell lung cancer H358 model animals during administration.
  • reaction solution was poured into water (200 mL), and the aqueous phase was extracted with ethyl acetate (50 mL*3).
  • the organic phases were combined, and the organic phase was washed with saturated brine (200 mL), dried over anhydrous sodium sulfate, filtered to collect the filtrate, and concentrated under reduced pressure to obtain a residue.
  • the residue was separated by flash column chromatography and purified to obtain A-1-2.
  • reaction solution was quenched with saturated aqueous ammonium chloride (100mL), and ethyl acetate (100mL*2) and dichloromethane (100mL) extraction, the organic phase was dried with anhydrous sodium sulfate, filtered and spin-dried to obtain a crude product.
  • the crude product is purified by column chromatography to obtain A-1.
  • WX001-4 (200mg, 651.12 ⁇ mol, 1eq) was added to the dry reaction flask, acetic anhydride (2mL) was pumped with nitrogen, and the reaction was carried out at 90°C for 16 hours. After the completion of the reaction, the reaction liquid was concentrated under reduced pressure with an oil pump at 45°C to obtain WX001-5.
  • WX004-1 50mg, 115.61 ⁇ mol, 1eq
  • WX004-2 11.69mg, 115.61 ⁇ mol, 1eq
  • the reaction solution is concentrated and purified by TLC plate to obtain WX004.
  • WX005-1 (379.45mg, 1.43mmol, 1eq) and dipalladium tridibenzylideneacetone (65.29mg, 71.30 ⁇ mol, 0.05eq), tris(2-furyl)phosphine (33.11mg, 142.60) were added dropwise at -25°C A solution of ⁇ mol, 0.1eq) and tetrahydrofuran (0.5mL) was reacted at 20°C for 10 hours. Add palladium tetrakistriphenylphosphine (82.5mg, 71.5 ⁇ mol, 0.05eq) and react at 65°C for 20 hours.
  • WX007-1 (30mg, 66.82 ⁇ mol, 1eq) and WX004-2 (6.76mg, 66.82 ⁇ mol, 1eq) to the pre-dried reaction flask, then dissolve it with dimethyl sulfoxide (1mL) and stir the reaction at 100°C. hour. After the reaction is complete, the reaction solution is concentrated and purified by TLC plate to obtain WX007.
  • WX009-1 (8.03g, 75.67mmol, 7.65mL, 1eq)
  • WX009-2 (10g, 75.67mmol, 1eq)
  • tetrahydrofuran 100mL
  • the reaction solution was spin-dried to obtain WX009-3.
  • 1 H NMR (DMSO-d 6 , 400MHz): ⁇ (ppm) 10.86 (br s, 1H), 8.00 (s, 1H), 7.59 (br d, J 6.6 Hz, 2H), 7.31-7.45 (m, 3H), 1.47(s, 9H).
  • WX009-5 (6.6g, 44.23mmol, 1eq) and hydrochloric acid ethyl acetate solution (4M, 33.18mL, 3eq) were added to the reaction flask, and the mixed solution was reacted at 25°C for 16 hours. The reaction solution was spin-dried to obtain WX009-6.
  • 1 H NMR 400MHz, DMSO-d 6 ) ⁇ ppm 4.95 (br s, 3H).
  • reaction solution was diluted with saturated sodium bicarbonate aqueous solution (50mL), extracted with dichloromethane (50mL*3), the organic phase was washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was kept at 45°C Concentrate under reduced pressure with a water pump, and purify the crude product by column chromatography to obtain WX009-8.
  • reaction solution was diluted with saturated aqueous ammonium chloride solution (5mL), extracted with dichloromethane (5mL*3), the organic phase was washed with saturated brine (5mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was kept at 45°C Concentrate under reduced pressure with a water pump, and purify with TLC plate to obtain WX009.
  • Extract with dichloromethane 3mL*2 combine the organic phases, wash the organic phase with 3mL saturated brine, dry with anhydrous sodium sulfate, and concentrate under reduced pressure to obtain a crude product.
  • the crude product was purified by thin layer chromatography silica gel plate to obtain WX011-2.
  • WX013-3 (520mg, 2.29mmol, 1eq) was added to the reaction flask, hydrochloric acid ethyl acetate solution (4M, 5mL, 8.74eq) was added, and the mixture was stirred at 25°C for 2 hours. After the reaction is completed, water (10 mL) is added to the reaction solution, the aqueous phase is extracted and collected, and then spin-dried to obtain a crude product. The crude product was purified by column chromatography to obtain WX013-4. 1 H NMR (400MHz, CD 3 Cl) ⁇ ppm 3.53 (s, 3H) 3.82 (s, 3H) 5.01 (s, 1H).
  • the ability of the compound to inhibit ERK2 kinase activity is measured.
  • test compound was dissolved in 100% DMSO to prepare a mother liquor with a specific concentration. Use the Integra Viaflo Assist smart pipette to serially dilute the compound in the DMSO solution.
  • the ERK2 kinase activity is calculated as the ratio of the remaining kinase activity in the test sample to the kinase activity of the control group (DMSO treatment). Using Prism (GraphPad Software) were fitted curve 50 calculated value IC.
  • the compound of the present invention exhibits excellent inhibitory activity against ERK2 enzyme.
  • the ability of the compound to inhibit the proliferation of HT29 tumor cells is measured.
  • test compound was dissolved in 100% DMSO to prepare a 10 mM mother liquor.
  • the compound of the present invention exhibits excellent inhibitory activity on the proliferation of HT29 cells.
  • the solvent is 5% DMSO+95% (20% HP- ⁇ -CD).
  • the compound to be tested is mixed with an appropriate amount of intravenous solvent, vortexed and sonicated to prepare a clear solution of 0.5 mg/mL, which is filtered by a microporous membrane. It is used later; the solvent of the oral group is 5% DMSO+95% (20% HP- ⁇ -CD), and the test compound is mixed with the solvent, vortexed and sonicated to prepare a 0.3 mg/mL solution.
  • DMSO dimethyl sulfoxide
  • HP- ⁇ -CD hydroxypropyl- ⁇ -cyclodextrin.
  • C max is the maximum concentration
  • F% is the oral bioavailability
  • DNAUC AUC PO /Dose
  • AUC PO is the oral exposure
  • Dose is the drug dose
  • Vd ss is the volume of distribution
  • Cl is the clearance rate
  • T 1/2 Is the half-life
  • NA means not detected.
  • the compound of the present invention exhibits excellent oral exposure and bioavailability.
  • Cage It is made of polycarbonate, with a volume of 300mm ⁇ 180mm ⁇ 150mm, and the bedding is corncob, which is replaced once a week;
  • Experimental animals can eat freely during the entire experimental period (irradiation sterilization, dry granular food);
  • Drinking water laboratory animals can drink sterilized water freely
  • the animal information card of each cage should indicate the number of animals in the cage, gender, strain, date of receipt, and experiment number of the dosing regimen.
  • Animal identification laboratory animals are identified by ear tags.
  • Human non-small cell lung cancer H358 cells are cultured in a monolayer in vitro, and the culture conditions are 1640 medium with 10% fetal bovine serum, 37°C and 5% CO 2 incubator. Use pancreatin-EDTA for routine digestion and passaging three times a week. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected, counted, and inoculated;
  • Tumor tissue inoculation and grouping 0.1 mL (5 ⁇ 10 5 cells) of H358 cells were subcutaneously inoculated into the right armpit of each mouse. When the average tumor volume reached 100 mm 3 , the animals were randomly divided into 2 groups and the administration medicine. The experimental grouping and dosing schedule are shown in Table 5;
  • Vehicle group 5% DMSO + 95% (20% HP- ⁇ -CD).
  • Test compound group Weigh the quantitative test compound in the dispensing bottle, add the corresponding volume of DMSO and vortex to obtain a clear solution, add the corresponding volume of 20% HP- ⁇ -CD and vortex to obtain a uniform Suspension. The compound is prepared once every three days.
  • TGI (%) The anti-tumor efficacy of the compound was evaluated by TGI (%).

Landscapes

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

Abstract

提供了一类噻唑并内酰胺类化合物,及其在制备治疗ERK相关疾病的药物中的应用。具体提供了式(III)所示化合物或其药学上可接受的盐。

Description

作为ERK抑制剂的噻唑并内酰胺类化合物及其应用
本申请主张如下优先权
CN201911244773.3,申请日:2019年12月06日;
CN201911257990.6,申请日:2019年12月10日;
CN202010107001.1,申请日:2020年02月20日;
CN202011138526.8,申请日:2020年10月22日;
CN202011402966.X,申请日:2020年12月02日。
技术领域
本发明涉及一类噻唑并内酰胺类化合物,及其在制备治疗ERK相关疾病的药物中的应用。具体涉及式(III)所示化合物或其药学上可接受的盐。
背景技术
Ras/Raf/MEK/ERK通路是一条经典的有丝分裂原活化蛋白激酶(mitogen activated protein kinase,MAPK)信号级联通路,参与各种生长因子、细胞因子、丝裂原以及激素受体活化后的信号转导,是控制细胞生长、分化和存活最重要的信号转导途径之一。
研究表明,突变或扩增引起的Ras/Raf/MEK/ERK通路异常活化是多种癌症发生的决定因素。在人类肿瘤中,RAS突变发生率约为22%,BRAF突变发生率约为7%,MEK突变发生率约为1%,因此,该通路上的关键节点蛋白已成为癌症治疗的重要靶点(Cancer Discov.2019,9,329-341)。目前,已有多个BRAF抑制剂和MEK1/2抑制剂,以及它们的联用方案,被美国FDA批准用于黑色素瘤、BRAFV600E突变型非小细胞肺癌等癌症的治疗。然而,使用这些上游节点的BRAF和MEK抑制剂后,由于突变或通路重新激活,会快速导致耐药性问题,极大地限制了它们的临床应用。
细胞外调节蛋白激酶(extracellular regulated protein kinases,ERK),特别是ERK1和ERK2激酶,是Ras/Raf/MEK/ERK通路的主要参与者和下游关键节点,在许多人类的癌症中都可发现它们的过度激活。ERK作为该通路的末端信号激酶,目前尚未发现有耐药突变,因此,靶向ERK激酶的药物有望克服上游靶点抑制剂治疗后产生的耐药性问题,成为更具潜力的治疗策略。但迄今为止,关于ERK抑制剂的研究仍处于临床阶段,还没有ERK抑制剂作为药物批准上市。
综上所述,迫切需要研发出安全、有效的ERK抑制剂药物满足肿瘤治疗的需要。
发明内容
本发明提供了式(III)所示化合物或其药学上可接受的盐,
Figure PCTCN2020134286-appb-000001
其中,
R 1选自H、C 1-3烷基和C 3-5环烷基,其中,所述C 1-3烷基和C 3-5环烷基任选被1、2或3个R a取代;
R 2和R 3分别独立地选自C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R b取代;
R 4选自H、F、Cl、Br、I和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R c取代;
R 5选自F、Cl、Br、I和和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R e取代;
m选自0、1和2;
n选自0、1和2;
环A选自吡唑基和四氢吡喃基,其中,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
R a、R b、R c和R e分别独立地选自D、F、Cl、Br、I、OH和OCH 3
R d选自F、Cl、Br、I、CH 3和OCH 3
本发明的一些方案中,上述R 1选自H、CH 3和环丙基,所述CH 3和环丙基任选被1、2或3个R a取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自H、CH 3、CHF 2、CD 3、CH 2CH 2OCH 3和环丙基,其他变量如本发明所定义。
本发明的一些方案中,上述R 2和R 3分别独立地选自任选CH 3和CH 2CH 3,所述CH 3和CH 2CH 3任选被1、2或3个R b取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 2和R 3分别独立地选自CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 4选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R c取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 4选自H、F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R e取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R e取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自H、F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2020134286-appb-000002
所述
Figure PCTCN2020134286-appb-000003
Figure PCTCN2020134286-appb-000004
任选被1、2或3个R d取代,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2020134286-appb-000005
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020134286-appb-000006
选自
Figure PCTCN2020134286-appb-000007
Figure PCTCN2020134286-appb-000008
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020134286-appb-000009
选自
Figure PCTCN2020134286-appb-000010
Figure PCTCN2020134286-appb-000011
其他变量如本发明所定义。
本发明提供了式(ⅠII)所示化合物或其药学上可接受的盐,
Figure PCTCN2020134286-appb-000012
其中,
R 1选自H和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R a取代;
R 2和R 3分别独立地选自C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R b取代;
R 4选自H、F、Cl、Br、I和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R c取代;
R 5选自F、Cl、Br、I和和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R e取代;
m选自0、1和2;
n选自0、1和2;
环A选自吡唑基和四氢吡喃基,其中,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
R a、R b、R c和R e分别独立地选自F、Cl、Br、I和OH;
R d选自F、Cl、Br、I和CH 3
本发明的一些方案中,上述R 1选自H和CH 3,所述CH 3任选被1、2或3个R a取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 2和R 3分别独立地选自任选CH 3和CH 2CH 3,所述CH 3和CH 2CH 3任选被1、2或3个R b取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 2和R 3分别独立地选自CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 4选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R c取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 4选自H、F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R e取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R e取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 5选自H、F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2020134286-appb-000013
所述
Figure PCTCN2020134286-appb-000014
Figure PCTCN2020134286-appb-000015
任选被1、2或3个R d取代,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2020134286-appb-000016
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020134286-appb-000017
选自
Figure PCTCN2020134286-appb-000018
Figure PCTCN2020134286-appb-000019
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020134286-appb-000020
选自
Figure PCTCN2020134286-appb-000021
Figure PCTCN2020134286-appb-000022
其他变量如本发明所定义。
本发明提供了式(Ⅰ)所示化合物或其药学上可接受的盐,
Figure PCTCN2020134286-appb-000023
其中,
R 1选自H和C 1-3烷基,其中所述C 1-3烷基任选被1、2或3个R a取代;
R 2和R 3分别独立地选自C 1-3烷基,所述C 1-3烷基任选被1、2或3个R b取代;
R 4选自H、F、Cl、Br、I和C 1-3烷基,其中所述C 1-3烷基任选被1、2或3个R c取代;
m选自0、1和2;
环A选自吡唑基和四氢吡喃基,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
R a、R b和R c分别独立地选自F、Cl、Br、I和OH;
R d选自F、Cl、Br、I和CH 3
本发明的一些方案中,上述R 1选自H和CH 3,所述CH 3任选被1、2或3个R a取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 2和R 3分别独立地选自任选CH 3和CH 2CH 3,所述CH 3和CH 2CH 3任选被1、2或3个R b取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 2和R 3分别独立地选自CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述R 4选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R c取代,其他变量如本发明所定义。
本发明的一些方案中,上述R 4选自H、F、Cl、Br、I和CH 3,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2020134286-appb-000024
所述
Figure PCTCN2020134286-appb-000025
Figure PCTCN2020134286-appb-000026
任选被1、2或3个R d取代,其他变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2020134286-appb-000027
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020134286-appb-000028
选自
Figure PCTCN2020134286-appb-000029
Figure PCTCN2020134286-appb-000030
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2020134286-appb-000031
选自
Figure PCTCN2020134286-appb-000032
Figure PCTCN2020134286-appb-000033
其他变量如本发明所定义。
本发明还有一些方案由上述变量任意组合而来。
本发明的一些方案中,上述述化合物或其药学上可接受的盐,其选自:
Figure PCTCN2020134286-appb-000034
其中,
R 1、R 2、R 3、R 4、R 5和m如本发明所定义。
本发明还提供了下式所示化合物或其药学上可接受的盐,
Figure PCTCN2020134286-appb-000035
Figure PCTCN2020134286-appb-000036
本发明的一些方案中,上述化合物或其药学上可接受的盐在制备治疗ERK相关疾病的药物中的应用。
本发明的一些方案中,上述应用,其特征在于,所述ERK抑制剂相关药物是用于治疗实体瘤的药物。
技术效果
本发明化合物表现出较优的对ERK2酶的抑制活性;同时,表现出较优的对HT29细胞增殖抑制活性。本发明化合物展现了优良的口服暴露量和生物利用度;本发明化合物能显著抑制肿瘤生长,给药过程中动物的体重未见明显下降,耐受性好。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与 相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机胺或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
除非另有说明,术语“异构体”意在包括几何异构体、顺反异构体、立体异构体、对映异构体、旋光异构体、非对映异构体和互变异构体。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,“(+)”表示右旋,“(-)”表示左旋,“(±)”表示外消旋。
除非另有说明,用楔形实线键
Figure PCTCN2020134286-appb-000037
和楔形虚线键
Figure PCTCN2020134286-appb-000038
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2020134286-appb-000039
和直形虚线键
Figure PCTCN2020134286-appb-000040
表示立体中心的相对构型,用波浪线
Figure PCTCN2020134286-appb-000041
表示楔形实线键
Figure PCTCN2020134286-appb-000042
或楔形虚线键
Figure PCTCN2020134286-appb-000043
或用波浪线
Figure PCTCN2020134286-appb-000044
表示直形实线键
Figure PCTCN2020134286-appb-000045
或直形虚线键
Figure PCTCN2020134286-appb-000046
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于 99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当一个取代基数量为0时,表示该取代基是不存在的,比如-A-(R) 0表示该结构实际上是-A。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基的键可以交叉连接到一个环上的两一个以上原子时,这种取代基可以与这个环上的任意原子相键合,例如,结构单元
Figure PCTCN2020134286-appb-000047
表示其取代基R可在环己基 或者环己二烯上的任意一个位置发生取代。当所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,吡啶基作为取代基可以通过吡啶环上任意一个碳原子连接到被取代的基团上。
当所列举的连接基团没有指明其连接方向,其连接方向是任意的,例如,
Figure PCTCN2020134286-appb-000048
中连接基团L为-M-W-,此时-M-W-既可以按与从左往右的读取顺序相同的方向连接环A和环B构成
Figure PCTCN2020134286-appb-000049
也可以按照与从左往右的读取顺序相反的方向连接环A和环B构成
Figure PCTCN2020134286-appb-000050
所述连接基团、取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
除非另有规定,当某一基团具有一个或多个可连接位点时,该基团的任意一个或多个位点可以通过化学键与其他基团相连。当该化学键的连接方式是不定位的,且可连接位点存在H原子时,则连接化学键时,该位点的H原子的个数会随所连接化学键的个数而对应减少变成相应价数的基团。所述位点与其他基团连接的化学键可以用直形实线键
Figure PCTCN2020134286-appb-000051
直形虚线键
Figure PCTCN2020134286-appb-000052
或波浪线
Figure PCTCN2020134286-appb-000053
表示。例如-OCH 3中的直形实线键表示通过该基团中的氧原子与其他基团相连;
Figure PCTCN2020134286-appb-000054
中的直形虚线键表示通过该基团中的氮原子的两端与其他基团相连;
Figure PCTCN2020134286-appb-000055
中的波浪线表示通过该苯基基团中的1和2位碳原子与其他基团相连;
Figure PCTCN2020134286-appb-000056
表示该哌啶基上的任意可连接位点可以通过1个化学键与其他基团相连,至少包括
Figure PCTCN2020134286-appb-000057
这4种连接方式,即使-N-上画出了H原子,但是
Figure PCTCN2020134286-appb-000058
仍包括
Figure PCTCN2020134286-appb-000059
这种连接方式的基团,只是在连接1个化学键时,该位点的的H会对应减少1个变成相应的一价哌啶基。
除非另有规定,环上原子的数目通常被定义为环的元数,例如,“5-7元环”是指环绕排列5-7个原子的“环”。
除非另有规定,术语“C 1-3烷基”用于表示直链或支链的由1至3个碳原子组成的饱和碳氢基团。所述C 1-3烷基包括C 1-2和C 2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次 甲基)。C 1-3烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)等。
除非另有规定,“C 3-5环烷基”表示由3至5个碳原子组成的饱和环状碳氢基团,其为单环体系,所述C 3-5环烷基包括C 3-4和C 4-5环烷基等;其可以是一价、二价或者多价。C 3-5环烷基的实例包括,但不限于,环丙基、环丁基、环戊基等。
术语“保护基”包括但不限于“氨基保护基”、“羟基保护基”或“巯基保护基”。术语“氨基保护基”是指适合用于阻止氨基氮位上副反应的保护基团。代表性的氨基保护基包括但不限于:甲酰基;酰基,例如链烷酰基(如乙酰基、三氯乙酰基或三氟乙酰基);烷氧基羰基,如叔丁氧基羰基(Boc);芳基甲氧羰基,如苄氧羰基(Cbz)和9-芴甲氧羰基(Fmoc);芳基甲基,如苄基(Bn)、三苯甲基(Tr)、1,1-二-(4'-甲氧基苯基)甲基;甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。术语“羟基保护基”是指适合用于阻止羟基副反应的保护基。代表性羟基保护基包括但不限于:烷基,如甲基、乙基和叔丁基;酰基,例如链烷酰基(如乙酰基);芳基甲基,如苄基(Bn),对甲氧基苄基(PMB)、9-芴基甲基(Fm)和二苯基甲基(二苯甲基,DPM);甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明的化合物可以通过本领域技术人员所熟知的常规方法来确认结构,如果本发明涉及化合物的绝对构型,则该绝对构型可以通过本领域常规技术手段予以确证。例如单晶X射线衍射法(SXRD),把培养出的单晶用Bruker D8 venture衍射仪收集衍射强度数据,光源为CuKα辐射,扫描方式:
Figure PCTCN2020134286-appb-000060
扫描,收集相关数据后,进一步采用直接法(Shelxs97)解析晶体结构,便可以确证绝对构型。
本发明所使用的溶剂可经市售获得。
本发明采用下述缩略词:aq代表水;eq代表当量、等量;二氯甲烷代表二氯甲烷;PE代表石油醚;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;乙醇代表乙醇;MeOH代表甲醇;Cbz代表苄氧羰基,是一种胺保护基团;BOC代表叔丁氧羰基是一种胺保护基团;r.t.代表室温;O/N代表过夜;四氢呋喃代表四氢呋喃;Boc 2O代表二叔丁基二碳酸酯;TFA代表三氟乙酸;DIPEA代表二异丙基乙基胺;iPrOH代表2-丙醇;mp代表熔点。
化合物依据本领域常规命名原则或者使用
Figure PCTCN2020134286-appb-000061
软件命名,市售化合物采用供应商目录名称。
说明书附图
图1:人非小细胞肺癌H358模型动物在分别给予溶剂和WX006后的肿瘤生长曲线。
图2:人非小细胞肺癌H358模型动物在给药过程中的体重变化率(%)。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
参考例1:片段A-1
Figure PCTCN2020134286-appb-000062
步骤1:化合物A-1-2的合成
在预先干燥过的单口瓶中加入乙酸钠(4.64g,56.60mmol,5eq)、单过硫酸氢钾(13.92g,22.64mmol,2eq)和水(47mL)的溶液,降温至0℃,滴加A-1-1(4.7g,11.32mmol,1eq)、溶剂四氢呋喃(47mL)和甲醇(47mL)的溶液,并在0℃搅拌1小时。29℃油浴搅拌15小时。反应结束后将该反应液倒入水(200mL)中,水相用乙酸乙酯萃取(50mL*3)。合并有机相,有机相依次用饱和食盐水洗涤(200mL),无水硫酸钠干燥,过滤收集滤液,减压浓缩得到残余物。残余物通过快速柱层析分离,纯化得到A-1-2。 1H NMR(400MHz,CDCl 3)δppm 8.67(d,J=4.9Hz,1H),7.64(d,J=4.9Hz,1H),3.37(s,3H),1.63-1.53(m,6H),1.39-1.30(m,6H),1.26-1.12(m,6H),0.90(t,J=7.3Hz,9H)。
步骤2:化合物A-1的合成
向反应瓶中加入A-1-2(3.9g,8.72mmol,1eq)、A-1-3(1.02g,10.46mmol,1.2eq)和四氢呋喃(117mL),抽换氮气后-35℃滴加入六甲基二硅基胺基锂(1M,18.31mL,2.1eq),-35℃下混合液反应10分钟,反应结束后将反应液用饱和氯化铵水溶液(100mL)淬灭,乙酸乙酯(100mL*2)和二氯甲烷(100mL)萃取,有机相用无水硫酸钠干燥,过滤旋干得粗品。粗品用柱层析纯化得到A-1。 1H NMR(400MHz,CDCl 3)δppm 8.17(d,J=4.85Hz,1H),7.46(d,J=1.76Hz,1H),6.91(d,J=4.63Hz,1H),6.60(s,1H),6.32(d,J=1.98Hz,1H),3.79(s,3H),1.52-1.61(m,6H),1.28-1.40(m,6H),1.03-1.20(m,6H),0.89(t,J=7.28Hz,9H)。
实施例1
Figure PCTCN2020134286-appb-000063
合成路线
Figure PCTCN2020134286-appb-000064
步骤1:WX001-3的合成
氮气保护,在-78℃下向反应瓶中加入二异丙基胺基锂(2M,2.88mL,2.4eq)的四氢呋喃(5mL)溶液,随后慢慢加入WX001-1(500mg,2.40mmol,1eq),四甲基乙二胺(418.94mg,3.61mmol,544.08μL,1.5eq)的四氢呋喃(1mL),-78℃反应0.5小时,然后再加入WX001-2(279.18mg,4.81mmol,353.40μL,2eq),-78℃继续反应2小时。反应完毕后,在0℃下将反应液缓慢倒入30mL饱和氯化铵水溶液中,用盐酸(2mol/L)调节pH至3-4左右,用乙酸乙酯(20mL*3)萃取,合并有机相,并用饱和食盐水(20mL*3)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩得到WX001-3。
步骤2:WX001-4的合成
向反应瓶中加入WX001-3(250mg,939.45μmol,1eq)和乙腈(8mL),抽换氮气后0℃加入浓硫酸(110.57mg,1.13mmol,60.09μL,1.2eq),25℃下混合液反应16小时。反应完毕后,向反应液中加入10mL水稀释,乙酸乙酯(30mL*3)萃取,合并有机相,并用饱和食盐水(30mL*3)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩得到WX001-4。
步骤3:WX001-5的合成
向干燥的反应瓶中加入WX001-4(200mg,651.12μmol,1eq),醋酸酐(2mL)抽换氮气,90℃反应16小时。反应完毕后,反应液用油泵在45℃下减压浓缩得到WX001-5。
步骤4:WX001-6的合成
向干燥的反应瓶中加入WX001-5(60mg,207.51μmol,1eq),盐酸(2M,2mL,19.28eq),和乙醇(2mL),50℃反应16小时。70℃反应4小时。反应完毕后,反应液用水泵在45℃下减压浓缩, 用乙酸乙酯(30mL*3)萃取合并有机相,并用饱和食盐水(30mL*3)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩,粗产品通过薄层制备色谱板纯化得到WX001-6。 1H NMR(400MHz,DMSO-d 6)δppm 8.85(br s,1H),1.48(s,6H)。
步骤5:WX001-7的合成
向干燥的反应瓶中加入WX001-6(60mg,242.80μmol,1eq)和N’N-二甲基甲酰胺(1mL),抽换氮气,0℃加入钠氢(14.57mg,364.21μmol,60%纯度,1.5eq),在0℃下反应0.5小时,然后再加入3-氯苄溴(49.89mg,242.80μmol,31.78μL,1eq),反应液慢慢升到25℃继续反应2小时。反应完毕后,反应液加入20mL水,用乙酸乙酯(10mL*3)萃取,合并有机相,并用饱和食盐水(10mL*3)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩。粗品通过薄层制备色谱板纯化得到WX001-7。 1H NMR(400MHz,CDCl 3)δppm 7.26(s,1H),7.17(s,3H),4.62(s,2H),1.37(s,6H)。
步骤6:WX001的合成
向反应瓶中加入WX001-7(35mg,94.17μmol,1eq),A-1(56.28mg,103.58μmol,1.1eq),甲苯(2mL),抽换氮气,加热到125℃,然后慢慢加入四三苯基膦钯(21.76mg,18.83μmol,0.2eq)。125℃反应48小时。反应完毕后,反应液用水泵在45℃下减压浓缩。粗产品通过薄层制备色谱板纯化得到WX001。
实施例2
Figure PCTCN2020134286-appb-000065
合成路线
Figure PCTCN2020134286-appb-000066
步骤1:WX002-2的合成
向干燥的反应瓶中加入WX001-6(50mg,202.34μmol,1eq)和二甲基甲酰胺(1mL),抽换氮气,0℃加入钠氢(12.14mg,303.51μmol,60%纯度,1.5eq),在0℃下反应0.5小时,然后再加入WX002-1(38.25mg,202.34μmol,24.84μL,1eq),反应液慢慢升到20℃继续反应2小时。反应完毕后,向反应液加入20mL水,用乙酸乙酯(10mL*3)萃取,合并有机相,并用饱和食盐水(10mL*3)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩。粗产品通过薄层制备色谱板纯化得到WX002-2。 1H NMR(400MHz,DMSO-d 6):δ(ppm)7.32-7.40(m,1H),7.15-7.23(m,2H),7.03-7.11(m,1H),4.67(s,2H),1.48(d,J=3.8Hz,6H)。
步骤2:WX002的合成
向反应瓶中加入WX002-2(60mg,168.91μmol,1eq),A-1(100.94mg,185.80μmol,1.1eq),甲苯(1mL),抽换氮气,加热到125℃,然后慢慢加入四三苯基膦钯(39.04mg,33.78μmol,0.2eq)。125℃反应48小时。反应完毕后,反应液用水泵在45℃下减压浓缩。粗品通过薄层制备色谱板纯化得到WX002。
实施例3
Figure PCTCN2020134286-appb-000067
合成路线
Figure PCTCN2020134286-appb-000068
步骤1:WX003-2的合成
向干燥的反应瓶中加入WX001-6(80mg,325.04μmol,1eq)和二甲基甲酰胺(4mL),抽换氮气,0℃加入钠氢(19.50mg,487.56μmol,60%纯度,1.5eq),在0℃下反应0.5小时,然后再加入WX003-1(67.29mg,325.04μmol,41.54μL,1eq),反应液慢慢升到25℃继续反应2小时。反应完毕后,向反应液中加入10mL水,用乙酸乙酯(20mL*3)萃取,合并有机相,并用饱和食盐水(20mL*3)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩。粗产品通过柱分离纯化得到WX003-2。 1H NMR(400MHz,DMSO-d 6):δ(ppm)7.33-7.47(m,2H),7.23(m,1H),4.65(s,2H),1.41-1.52(m,6H)。
步骤2:WX003的合成
向反应瓶中加入WX003-2(70mg,187.56μmol,1eq),A-1(112.09mg,206.32μmol,1.1eq),甲苯(2mL),抽换氮气,加热到125℃,然后慢慢加入四三苯基膦钯(43.35mg,37.51μmol,0.2eq)。125℃反应48小时。反应完毕后,反应液用水泵在45℃下减压浓缩。粗品通过薄层制备色谱板纯化得到WX003。
实施例4
Figure PCTCN2020134286-appb-000069
合成路线
Figure PCTCN2020134286-appb-000070
步骤1:WX004-1的合成
向反应瓶中加入WX002-2(70mg,197.06μmol,1eq),A-1-2(113.45mg,216.76μmol,1.1eq),甲苯(2mL),抽换氮气,加热到125℃,然后慢慢加入四三苯基膦钯(45.54mg,39.41μmol,0.2eq)。125℃反应48小时。反应完毕后,反应液用水泵在45℃下减压浓缩。粗品通过薄层制备色谱板纯化得到WX004-1。 1H NMR(400MHz,DMSO-d 6):δ(ppm)9.26(d,J=5.1Hz,1H),8.43(d,J=5.3Hz,1H), 7.33-7.42(m,1H),7.16-7.28(m,2H),7.08(td,J=8.6,2.0Hz,1H),4.73(s,2H),3.50(s,3H),1.55(s,6H)。
步骤2:WX004的合成
在预先干燥的反应瓶中加入WX004-1(50mg,115.61μmol,1eq),WX004-2(11.69mg,115.61μmol,1eq),之后用二甲基亚砜(1mL)溶解,100℃搅拌反应14小时。反应完毕,反应液浓缩后用薄层制备色谱板纯化得到WX004。
实施例5
Figure PCTCN2020134286-appb-000071
合成路线
Figure PCTCN2020134286-appb-000072
步骤1:WX005-2的合成
向反应瓶中加入WX001-7(530mg,1.43mmol,1eq)、氯化锌(0.7M,1.83mL,0.9eq)和四氢呋喃(3.5mL),抽换氮气后-25℃加入正丁基锂(2.5M,855.58μL,1.5eq),20℃下混合液反应1小时。-25℃滴加WX005-1(379.45mg,1.43mmol,1eq)和三二亚苄基丙酮二钯(65.29mg,71.30μmol,0.05eq)、三(2-呋喃基)膦(33.11mg,142.60μmol,0.1eq)和四氢呋喃(0.5mL)的溶液,20℃反应10小时。补加四三苯基膦钯(82.5mg,71.5μmol,0.05eq),65℃反应20小时。反应完毕后,反应液用甲醇淬灭,浓缩获得粗品。粗品用制备薄层色谱纯化得到WX005-2。 1H NMR(DMSO-d 6,400MHz):δ(ppm)8.75(s,1H),7.46(s,1H),7.26-7.41(m,3H),4.72(s,2H),2.65(s,3H),2.59(s,3H),1.53(s,6H)。
步骤2:WX005-3的合成
向反应瓶中加入WX005-2(140mg,324.85μmol,1eq)和二氯甲烷(6mL),抽换氮气后0℃加入间氯过氧苯甲酸(210.22mg,974.54μmol,80%纯度3eq),缓慢升温至20℃下混合液反应10小时。反应完毕后,反应液用饱和亚硫酸钠(15mL)和饱和碳酸氢钠水溶液(15mL)淬灭至pH到碱性和KI试纸不变色。二氯甲烷(3mL*3)萃取,有机相在45℃用水泵减压浓缩干得棕色固体。乙酸乙酯(3mL)打浆得到WX005-3。 1H NMR(DMSO-d 6,400MHz):δ(ppm)9.18(s,1H),7.47(s,1H),7.30-7.39(m,3H),4.73(s,2H),3.48(s,3H),2.83(s,3H),1.55(s,6H)。
步骤3:WX005的合成
向反应瓶中加入A-1-3(15.73mg,162.00μmol,1.5eq)和N’N-二甲基甲酰胺(1mL),抽换氮气后0℃加入钠氢(6.48mg,162.00μmol,60%纯度,1.5eq),0.5小时后加入WX005-3(50mg,108.00μmol,1eq)和N’N-二甲基甲酰胺(1mL)的溶液,20℃下混合液反应10小时。反应完毕后,反应液用饱和氯化铵水溶液(5mL)淬灭,二氯甲烷(1mL*3)萃取,有机相在45℃用水泵减压浓缩干。粗品用薄层层析纯化得到WX005。
实施例6
Figure PCTCN2020134286-appb-000073
合成路线
Figure PCTCN2020134286-appb-000074
步骤1:WX006-1的合成
在预先干燥的反应瓶中加入WX002-2(0.456g,1.28mmol,1eq),氯化锌(0.7M,1.65mL,0.9eq), 四氢呋喃(10.5mL),抽换氮气后,将反应体系降至-25℃,加入正丁基锂(2.5M,770.22μL,1.5eq),20℃混合液搅拌反应1小时,-25℃下加入WX005-1(341.59mg,1.28mmol,1eq),四三苯基膦钯(74.17mg,64.18μmol,0.05eq)的四氢呋喃(1.5mL)溶液,60℃搅拌反应12小时。反应完毕后,向反应液中加入甲醇3mL淬灭,直接旋干得粗品。粗产品通过薄层层析硅胶板纯化得到WX006-1。
步骤2:WX006-2的合成
在预先干燥的反应瓶中加入WX006-1(210mg,506.61μmol,1eq)和二氯甲烷(5mL,将反应体系降至0℃,加入间氯过氧苯甲酸(327.85mg,1.52mmol,80%纯度,3eq),缓慢升温至室温(20℃)搅拌反应12小时。反应完毕后,用碳酸氢钠水溶液(5mL)调节反应液pH至8左右,加入饱和的亚硫酸钠溶液至淀粉KI试纸不显蓝色,加入二氯甲烷(10mL*3)萃取,合并有机相,并用饱和食盐水(10mL*2)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩。粗产品通过薄层层析硅胶板纯化得到WX006-2。
步骤3:WX006的合成
在预先干燥的反应瓶中加入A-1-3(26.10mg,268.75μmol,1.2eq)和N’N-二甲基甲酰胺(3mL),将反应体系降至0℃,加入钠氢(13.44mg,335.93μmol,60%纯度,1.5eq),0℃搅拌反应0.5小时,加入WX006-2(100mg,223.96μmol,1eq),0℃搅拌反应0.5小时。反应完毕后,加水(5mL)淬灭反应,加入乙酸乙酯(5mL*3)萃取,合并有机相,并用饱和食盐水(5mL*4)洗涤,无水硫酸钠干燥,过滤,滤液用水泵在45℃下减压浓缩。粗产品通过薄层层析硅胶板纯化得到WX006。
实施例7
Figure PCTCN2020134286-appb-000075
合成路线
Figure PCTCN2020134286-appb-000076
步骤1:WX007-1的合成
向反应瓶中加入WX001-7(50mg,134.52μmol,1eq),A-1-2(77.45mg,147.98μmol,1.1eq),甲苯(2mL),抽换氮气,加热到125℃然后慢慢加入四三苯基膦钯(31.09mg,26.90μmol,0.2eq)。125℃反应48小时。反应完毕,反应液用水泵在45℃下减压浓缩。粗产品通过薄层层析硅胶板纯化得到WX007-1。 1H NMR(DMSO-d 6,400MHz):δ(ppm)9.26(d,J=5.1Hz,1H),8.43(d,J=5.1Hz,1H),7.47(s,1H),7.28-7.39(m,3H),4.73(s,2H),3.50(s,3H),1.55(s,6H)。
步骤2:WX007的合成
在预先干燥的反应瓶中加入WX007-1(30mg,66.82μmol,1eq),WX004-2(6.76mg,66.82μmol,1eq),之后用二甲基亚砜(1mL)溶解,100℃搅拌反应16小时。反应完毕,反应液浓缩后用薄层制备色谱板纯化得到WX007。
实施例8
Figure PCTCN2020134286-appb-000077
合成路线
Figure PCTCN2020134286-appb-000078
步骤1:WX008-2的合成
向反应瓶中加入WX008-1(9g,79.59mmol,1eq)、碳酸钾(13.20g,95.51mmol,1.2eq)和N’N-二甲基甲酰胺(100mL),抽换氮气后加热至120℃,搅拌10分钟,随后分批加入2-氯-2,2-二氟乙酸钠(24.27g,159.19mmol,2eq),120℃下混合液反应20分钟。反应完毕后,反应液用水(800mL)稀释,乙酸乙酯(200mL*3)萃取,有机相用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液用水泵减压浓缩,通过薄层层析硅胶板纯化得到WX008-2。 1H NMR(DMSO-d 6,400MHz):δ(ppm)7.96-8.28(m,2H),7.50(d,J=2.0Hz,1H)。
步骤2:WX008-3的合成
向反应瓶中加入钯碳(50mg,367.91μmol,10%纯度,1eq)和甲醇(1mL),抽换氮气后加入WX008-2(60mg,367.91μmol,1eq),抽换氢气,在氢气(741.67μg,367.91μmol,1eq)氛围下,15psi压力下,25℃下混合液反应1小时。反应完毕后,反应液用硅藻土过滤,滤液用水泵减压浓缩干得WX008-3。 1H NMR(DMSO-d 6,400MHz):δ(ppm)7.36-7.69(m,1H),7.33(s,1H),5.84(br s,2H),5.31(d,J=1.3Hz,1H)。
步骤3:WX008的合成
向反应瓶中加入WX008-3(17.88mg,134.37μmol,1.5eq)、WX006-2(40mg,89.58μmol,1eq)、二氯甲烷(0.5mL)和四氢呋喃(0.5mL),抽换氮气后降温至0℃,缓慢滴入六甲基二硅基胺基锂(1M,188.12μL,2.1eq),0℃下混合液反应0.5小时。反应完毕,反应液浓缩后用薄层制备色谱板纯化得到WX008。
实施例9
Figure PCTCN2020134286-appb-000079
合成路线
Figure PCTCN2020134286-appb-000080
步骤1:WX009-3的合成
向反应瓶中加入WX009-1(8.03g,75.67mmol,7.65mL,1eq)、WX009-2(10g,75.67mmol,1eq)和四氢呋喃(100mL),抽换氮气,25℃下混合液反应4小时。反应完毕后,反应液旋干得WX009-3。 1H NMR(DMSO-d 6,400MHz):δ(ppm)10.86(br s,1H),8.00(s,1H),7.59(br d,J=6.6Hz,2H),7.31-7.45(m,3H),1.47(s,9H)。
步骤2:WX009-4的合成
向反应瓶中加入WX009-3(10g,45.40mmol,1eq)、叔丁醇钾(6.11g,54.48mmol,1.2eq)和四氢呋喃(160mL),抽换氮气后缓慢滴入氘代碘甲烷(7.90g,54.48mmol,3.39mL,1.2eq),25℃下混合液反应16小时。反应完毕后,反应液用水(50mL)稀释,乙酸乙酯(100mL*3)萃取,有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液用水泵减压浓缩得到WX009-4。 1H NMR(DMSO-d 6,400MHz):δ(ppm)7.81(s,1H),7.64-7.72(m,2H),7.32-7.46(m,3H),1.50(s,9H)。
步骤3:WX009-5的合成
向反应瓶中加入湿钯碳(2g,10%纯度)和甲醇(100mL),抽换氩气后加入WX009-4(10.5g,44.25mmol,1eq),50psi压力下,氢气(89.19mg,44.25mmol,1eq)气氛中,50℃下混合液反应48小时。反应完毕后,反应液用硅藻土过滤,甲醇(20mL*2)洗涤,滤液用水泵减压浓缩干得到WX009-5。 1H NMR(DMSO-d 6,400MHz):δ(ppm)4.48(s,2H),1.40(s,9H)。
步骤4:WX009-6的合成
向反应瓶中加入WX009-5(6.6g,44.23mmol,1eq)和盐酸乙酸乙酯溶液(4M,33.18mL,3eq), 25℃下混合液反应16小时。反应液旋干得到WX009-6。 1H NMR(400MHz,DMSO-d 6)δppm 4.95(br s,3H)。
步骤5:WX009-8的合成
向反应瓶中加入WX009-6(1.5g,17.53mmol,1eq,HCl)、WX009-7(2.51g,17.53mmol,2.63mL,1eq)、乙酸(2.11g,35.07mmol,2.01mL,2eq)、硫酸镁(5.02g,41.73mmol,2.38eq)和乙醇(75mL),抽换氮气,90℃下混合液反应2小时。反应完毕后,反应液用饱和碳酸氢钠水溶液(50mL)稀释,二氯甲烷(50mL*3)萃取,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液在45℃用水泵减压浓缩,粗品用柱层析纯化得到WX009-8。 1H NMR(DMSO-d 6,400MHz):δ(ppm)6.98(d,J=1.6Hz,1H),5.24(d,J=1.8Hz,1H),5.09(br s,2H)。
步骤6:WX009的合成
向反应瓶中加入WX009-8(13.46mg,134.37μmol,1.5eq)、WX006-2(40mg,89.58μmol,1eq)、二氯甲烷(0.5mL)和四氢呋喃(0.5mL),抽换氮气后降温至0℃,缓慢滴入六甲基二硅基胺基锂(1M,188.12μL,2.1eq),0℃下混合液反应0.5小时。反应完毕后,反应液用饱和氯化铵水溶液(5mL)稀释,二氯甲烷(5mL*3)萃取,有机相用饱和食盐水(5mL)洗涤,无水硫酸钠干燥,过滤,滤液在45℃用水泵减压浓缩,用薄层制备色谱板纯化得到WX009。
实施例10
Figure PCTCN2020134286-appb-000081
合成路线
Figure PCTCN2020134286-appb-000082
步骤1:WX010的合成
在干躁的反应瓶中,将WX006-2(30mg,67.19μmol,1eq)、WX010-1(15.68mg,141.09μmol,2.1eq)加入到四氢呋喃(1.5mL)和二氯甲烷(1.5mL)的混合溶液,置换氮气,降温至0℃,加入六甲基二硅基胺基锂(1M,134.37μL,2eq),在0℃条件下搅拌0.5小时,升温至25℃,搅拌1.5小时。反应完毕后,加入水(10mL)稀释,加二氯甲烷(10mL*3)萃取,分液后收集有机相,依次用饱和食盐水溶液(10mL*3),无水硫酸钠干燥,减压浓缩,用薄层制备色谱板纯化得到WX010。
实施例11
Figure PCTCN2020134286-appb-000083
合成路线
Figure PCTCN2020134286-appb-000084
步骤1:WX011-2的合成
向干燥的反应瓶中加入WX009-7(226.22mg,1.58mmol,237.13μL,1eq)、硫酸镁(452.63mg,3.76mmol,2.38eq)、乙酸(189.75mg,3.16mmol,180.72μL,2eq)、乙醇(2.5mL),抽换氮气,25℃反应1小时,然后加入WX011-1(200mg,1.58mmol,1eq,HCl),80℃反应2小时,90℃反应12小时。反应完毕后,将反应液倒3mL入饱和碳酸氢钠水溶液中。用二氯甲烷3mL*2萃取,合并有机相,有机相用3mL饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到粗品。粗产品通过薄层层析硅胶板纯化得到WX011-2. 1H NMR(CH 3Cl-d,400MHz):δ(ppm)7.25(d,J=1.8Hz,1H),5.50(d,J=2.0Hz,1H),4.15-4.22(m,2H),3.83-4.11(m,2H),3.68-3.73(m,2H),3.34(s,3H)。
步骤2:WX011的合成
在干躁的反应瓶中,将WX006-2(40mg,89.58μmol,1eq)、WX011-2(26.56mg,188.12μmol,2.1eq)加入到四氢呋喃(2mL)和二氯甲烷(2mL)的混合溶液,置换氮气,降温至0℃,加入六甲基二硅基胺基锂(1M,179.16μL,2eq),在0℃条件下,搅拌0.5小时,升温至25℃,搅拌1小时。反应完毕后,加入水(5mL)稀释,二氯甲烷(5mL*3)萃取,分液后收集有机相,依次用饱和食盐水溶液(5mL*3),无水硫酸钠干燥,减压浓缩,用薄层制备色谱板纯化得到WX011。
实施例12
Figure PCTCN2020134286-appb-000085
合成路线
Figure PCTCN2020134286-appb-000086
步骤1:WX012的合成
向反应瓶中加入WX006-2(40mg,89.58μmol,1eq)、WX012-1(16.55mg,134.37μmol,1.5eq)、二氯甲烷(1mL)和四氢呋喃(1mL),抽换氮气后降温至0℃,缓慢滴入六甲基二硅基胺基锂(1M,188.12μL,2.1eq),0℃下混合液反应0.5小时。反应完毕后,反应液浓缩后用薄层制备色谱板纯化得到WX012。
实施例13
Figure PCTCN2020134286-appb-000087
合成路线
Figure PCTCN2020134286-appb-000088
步骤1:WX013-2的合成
将WX013-1(800mg,4.70mmol,1eq)与一水合氢氧化锂(986.42mg,23.51mmol,5eq)溶于水(8mL)与四氢呋喃(8mL)的混合溶液中,在25℃下搅拌2小时。反应完毕后,将反应液直接旋干,然后加入水(10mL)和二氯甲烷(10mL*3)进行萃取,收集有机相,用饱和食盐水洗,用无水硫酸钠干燥,过滤并旋干得到WX013-2。
步骤2:WX013-3的合成
将WX013-2(500mg,3.20mmol,1eq)、叠氮磷酸二苯酯(889.00mg,3.23mmol,700μL,1.01eq)、三乙胺(1.45g,14.37mmol,2mL,4.49eq)溶于叔丁醇(10mL)中,氮气置换三次后,在85℃下搅拌16小时。反应完毕后,将反应液旋干。粗品通过柱层析法纯化得到WX013-3。 1H NMR(400MHz,CD 3Cl)δppm 1.50(s,9H)3.58(s,3H)3.84(s,3H)5.61(br s,1H)。
步骤3:WX013-4的合成
将WX013-3(520mg,2.29mmol,1eq)加入到反应瓶中,加入盐酸乙酸乙酯溶液(4M,5mL,8.74eq),在25℃搅拌2小时。反应完毕后,向反应液中加入水(10mL),萃取并收集水相,然后旋干得到粗品。粗品通过柱层析法进行纯化得到WX013-4。 1H NMR(400MHz,CD 3Cl)δppm 3.53(s,3H)3.82(s,3H)5.01(s,1H)。
步骤4:WX013的合成
向干燥的反应瓶中加入WX006-2(50mg,111.98μmol,1eq),WX013-4(29.90mg,235.15μmol,2.1eq),二氯甲烷(1mL)和四氢呋喃(1mL),抽换氮气,将反应降温至0℃,滴加六甲基二硅基胺基锂(1M,223.96μL,2eq),0℃反应0.5小时,25℃反应1小时。反应完毕后,加入10mL水淬灭,加入20mL二氯甲烷萃取,分液后收集有机相,水相用二氯甲烷萃取(20mL*3)。合并有机相,依次用饱和食盐水洗涤(20mL*3),无水硫酸钠干燥,减压浓缩。浓缩后用薄层制备色谱板纯化得到WX013。
各实施例的氢谱和质谱数据如表1所示。
表1
Figure PCTCN2020134286-appb-000089
Figure PCTCN2020134286-appb-000090
Figure PCTCN2020134286-appb-000091
实验例一、体外酶活性测试:
1.实验目的:
测量化合物抑制ERK2激酶活性的能力。
2.实验缓冲液:
20mM Hepes(pH 7.5),10mM MgCl 2,1mM乙二醇双(2-氨基乙基醚)四乙酸(EGTA),0.02%Brij35,0.02mg/mL牛血清白蛋白(BSA),0.1mM Na 3VO 4,2mM二硫苏糖醇(DTT),1%DMSO。
3.化合物处理:
将测试化合物溶于100%DMSO中,配制成特定浓度的母液。利用Integra Viaflo Assist智能移液器将化合物连续稀释在DMSO溶液中。
3.实验方法
1)在新制备的反应缓冲液中配置底物MBP;
2)将ERK2激酶加入到上述MBP溶液中并轻轻混合;
3)运用超声技术(Echo550;纳升范围)将溶于100%DMSO中的化合物加入到激酶反应体系中,在室温下孵育20分钟;
4)将 33P-ATP(特定浓度10μCi/μL)加入到反应体系中,此时开始发生反应;
5)在室温下孵育2小时;
6)通过过滤--结合方法检测放射性的量;
7)ERK2激酶活性计算方式为测试样品中剩余激酶活性占对照组(二甲基亚砜处理)激酶活性的比值。使用Prism(GraphPad软件)进行曲线拟合并计算IC 50值。
4.实验结果见表2:
表2体外酶活性测试结果
Figure PCTCN2020134286-appb-000092
Figure PCTCN2020134286-appb-000093
结论:本发明化合物表现出较优的对ERK2酶抑制活性。
实验例二、体外细胞增殖抑制实验:
1.实验目的:
测量化合物抑制HT29肿瘤细胞增值的能力。
2.化合物处理:
将测试化合物溶于100%DMSO中,配制成10mM的母液。
3.实验步骤与方法:
1)开启生物安全柜紫外灯,倒计时30分钟;
2)37度水浴锅中,预热RPMI1640培养基和胰酶;
3)紫外照射完毕,开启生物安全柜,将预热培养基,胰酶,磷酸缓冲盐溶液(PBS)等用酒精擦拭并放入生物安全柜中;
4)将HT29细胞从培养箱中取出,在生物安全柜中去除旧培养基,加入10毫升PBS,轻轻摇晃,并去除PBS;
5)加入预热0.25%胰酶1.5毫升,水平晃动培养瓶,使其均匀覆盖到底部的细胞,置培养箱中2分钟;
6)用完全培养基终止细胞消化,并吹打至均匀的细胞悬液计数;
7)根据细胞计数结果,调整细胞悬液密度为1500细胞每孔,50微升每孔进行种板;
8)将化合物母液连续稀释在DMSO溶液中,并使用Tecan将化合物加入细胞板中;
9)将加过化合物的细胞板和CellTiterGlo放到室温平衡,后加CellTiterGlo 25微升至每孔,震荡1-2分钟,静置10分钟后检测信号值,并用XL-Fit分析数据,计算各化合物的IC 50
4.实验结果见表3:
表3体外细胞活性测试结果
Figure PCTCN2020134286-appb-000094
Figure PCTCN2020134286-appb-000095
结论:本发明化合物表现出较优的对HT29细胞增殖抑制活性。
实验例三、体内DMPK研究:
小鼠体内DMPK研究
1.实验目的:
以雌性BALB/c小鼠为受试动物,单次给药后测定化合物血药浓度并评估药代动力学行为。
2.实验操作:
选择健康成年雌性BALB/c小鼠8只,4只为静注组,4只为口服组。静注组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与适量静注溶媒混合,涡旋并超声,制备得到0.5mg/mL澄清溶液,微孔滤膜过滤后备用;口服组溶媒为5%DMSO+95%(20%HP-β-CD),待测化合物与溶媒混合后,涡旋并超声,制备得到0.3mg/mL溶液。小鼠1mg/kg静脉给药或3mg/kg口服给药后,收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
注DMSO:二甲基亚砜;HP-β-CD:羟丙基-β-环糊精。
3.实验结果见表4:
表4化合物PK测试结果
Figure PCTCN2020134286-appb-000096
Figure PCTCN2020134286-appb-000097
备注:C max为最大浓度;F%为口服生物利用度;DNAUC=AUC PO/Dose,AUC PO为口服暴露量,Dose为药物剂量;Vd ss为分布容积;Cl为清除率;T 1/2为半衰期;NA指未检测。
结论:本发明化合物展现了优良的口服暴露量和生物利用度。
实验例四、小鼠H358模型体内药效研究:
1.实验目的:
使用人非小细胞肺癌H358细胞皮下异种移植肿瘤裸小鼠模型,评价WX006的抗肿瘤作用。
2.实验动物:
种属:小鼠
品系:BALB/c裸小鼠
周龄:6-7周龄
性别:雌性
体重:20克
供应商:上海西普尔-必凯实验动物有限公司
动物合格证号:20180006017149
3.饲养环境:
动物在SPF级动物房以IVC(独立送风系统,恒温恒湿)笼具饲养(每笼4只),温度:20-26℃,
湿度:40-70%;
笼具:以聚碳酸酯制成,体积300mm×180mm×150mm,垫料为玉米芯,每周更换一次;
食物:实验动物在整个实验阶段中可自由进食(照射灭菌,干颗粒状食物);
饮水:实验动物可自由饮用灭菌水;
笼具标识:每笼动物信息卡应注明笼内动物数目,性别,品系,接收日期,给药方案实验编号,
组别以及实验开始日期;
动物标识:实验动物以耳标进行标识。
4.实验内容:
1)实验细胞及培养:人非小细胞肺癌H358细胞体外单层培养,培养条件为1640培养基中加10%胎牛血清,37℃5%CO 2孵箱培养。一周三次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%,数量到达要求时,收取细胞,计数,接种;
2)肿瘤组织接种及分组:0.1mL(5×10 5个)H358细胞皮下接种于每只小鼠的右侧腋下,肿瘤平均体积达到100mm 3时,将动物随机分为2组,开始给药。实验分组和给药方案见表5;
表5实验动物分组及给药方案
Figure PCTCN2020134286-appb-000098
Figure PCTCN2020134286-appb-000099
3)实验动物日常观察:本实验方案的拟定及任何修改均通过了实验动物管理与使用委员会(IACUC)的评估核准。实验动物的使用及福利遵照国际实验动物评估和认可委员会(AAALAC)的规定执行。每天监测动物的健康状况及死亡情况,例行检查包括观察肿瘤生长和药物治疗对动物日常行为表现的影响如行为活动,摄食摄水量(仅目测),体重变化(每周测量两次体重),外观体征或其它不正常情况。基于各组动物数量记录了组内动物死亡数和副作用。
4)受试物的配制
a)溶媒组:5%DMSO+95%(20%HP-β-CD)。
b)待测化合物组:称量定量的受试化合物于配药瓶内,加入相应体积的DMSO后涡旋,得到澄清溶液,在加入相应体积的20%HP-β-CD后涡旋,得到均一混悬液。化合物三天配制一次。
5)肿瘤测量和实验指标:
a)每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:TV=1/2×a×b 2,a和b分别表示肿瘤的长径和短径;
b)化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)={[1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)]/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)}×100%。
5.实验结果:
1)如表6和图1所示,在人非小细胞肺癌H358细胞皮下异种移植肿瘤裸小鼠模型上,口服给药至第28天,WX006 30mg/kg具有明显的抑制肿瘤生长作用,TGI为94%。
2)实验动物的体重作为间接测定药物毒性的参考指标。如图2所示,给药至第28天时,溶剂对照组和WX006组所有动物的体重均未有明显下降,无发病或死亡现象。
表6:小鼠H358模型体内药效实验结果
药物 TGI
WX006(30mg/kg,PO,QD) 94%
6.实验结论:WX006在给药剂量下,能显著抑制肿瘤生长;给药过程中动物的体重未见明显下降,耐受性好。

Claims (17)

  1. 式(III)所示化合物或其药学上可接受的盐,
    Figure PCTCN2020134286-appb-100001
    其中,
    R 1选自H、C 1-3烷基和C 3-5环烷基,其中,所述C 1-3烷基和C 3-5环烷基任选被1、2或3个R a取代;
    R 2和R 3分别独立地选自C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R b取代;
    R 4选自H、F、Cl、Br、I和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R c取代;
    R 5选自F、Cl、Br、I和和C 1-3烷基,其中,所述C 1-3烷基任选被1、2或3个R e取代;
    m选自0、1和2;
    n选自0、1和2;
    环A选自吡唑基和四氢吡喃基,其中,所述吡唑基和四氢吡喃基任选被1、2或3个R d取代;
    R a、R b、R c和R e分别独立地选自D、F、Cl、Br、I、OH和OCH 3
    R d选自F、Cl、Br、I、CH 3和OCH 3
  2. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R 1选自H、CH 3和环丙基,所述CH 3和环丙基任选被1、2或3个R a取代。
  3. 根据权利要求2所述化合物或其药学上可接受的盐,其中R 1选自H、CH 3、CHF 2、CD 3、CH 2CH 2OCH 3和环丙基。
  4. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R 2和R 3分别独立地选自任选CH 3和CH 2CH 3,所述CH 3和CH 2CH 3任选被1、2或3个R b取代。
  5. 根据权利要求4所述化合物或其药学上可接受的盐,其中,R 2和R 3分别独立地选自CH 3
  6. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R 4选自H、F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R c取代。
  7. 根据权利要求6所述化合物或其药学上可接受的盐,其中,R 4选自H、F、Cl、Br、I和CH 3
  8. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R 5选自F、Cl、Br、I和CH 3,所述CH 3任选被1、2或3个R e取代。
  9. 根据权利要求8所述化合物或其药学上可接受的盐,其中,R 5选自F、Cl、Br、I和CH 3
  10. 根据权利要求1所述化合物或其药学上可接受的盐,其中,环A选自
    Figure PCTCN2020134286-appb-100002
    Figure PCTCN2020134286-appb-100003
    所述
    Figure PCTCN2020134286-appb-100004
    任选被1、2或3个R d取代。
  11. 根据权利要求10所述化合物或其药学上可接受的盐,其中,环A选自
    Figure PCTCN2020134286-appb-100005
    Figure PCTCN2020134286-appb-100006
  12. 根据权利要求1所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2020134286-appb-100007
    选自
    Figure PCTCN2020134286-appb-100008
  13. 根据权利要求12所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2020134286-appb-100009
    选自
    Figure PCTCN2020134286-appb-100010
  14. 根据权利要求1~9任意一项所述化合物或其药学上可接受的盐,其选自
    Figure PCTCN2020134286-appb-100011
    其中,
    R 1如权利要求1~3任意一项所定义;
    R 2和R 3如权利要求1、4或5任意一项所定义;
    R 4如权利要求1、6或7任意一项所定义;
    R 5如权利要求1、8或9任意一项所定义;
    m如权利要求1所定义。
  15. 下式所示化合物或其药学上可接受的盐,
    Figure PCTCN2020134286-appb-100012
  16. 根据权利要求1~15任意一项所述化合物或其药学上可接受的盐在制备治疗ERK相关疾病 的药物中的应用。
  17. 根据权利要求16所述的应用,其特征在于,所述ERK抑制剂相关药物是用于治疗实体瘤的药物。
PCT/CN2020/134286 2019-12-06 2020-12-07 作为erk抑制剂的噻唑并内酰胺类化合物及其应用 WO2021110169A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP20895381.0A EP4071155A4 (en) 2019-12-06 2020-12-07 THIAZOLOLACTAM COMPOUND USED AS ERK INHIBITOR AND USE THEREOF
US17/782,814 US20230072937A1 (en) 2019-12-06 2020-12-07 Thiazololactam compound as erk inhibitor and use thereof
KR1020227021797A KR20220107249A (ko) 2019-12-06 2020-12-07 Erk 억제제로서의 티아졸로락탐 화합물 및 이의 용도
AU2020398022A AU2020398022B2 (en) 2019-12-06 2020-12-07 Thiazololactam compound as ERK inhibitor and use thereof
JP2022534190A JP7409719B2 (ja) 2019-12-06 2020-12-07 Erk阻害剤としてのチアゾロラクタム系化合物およびその使用
CA3160903A CA3160903A1 (en) 2019-12-06 2020-12-07 Thiazololactam compound as erk inhibitor and use thereof
CN202080084536.XA CN114829365B (zh) 2019-12-06 2020-12-07 作为erk抑制剂的噻唑并内酰胺类化合物及其应用

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN201911244773 2019-12-06
CN201911244773.3 2019-12-06
CN201911257990.6 2019-12-10
CN201911257990 2019-12-10
CN202010107001 2020-02-20
CN202010107001.1 2020-02-20
CN202011138526.8 2020-10-22
CN202011138526 2020-10-22
CN202011402966.X 2020-12-02
CN202011402966 2020-12-02

Publications (1)

Publication Number Publication Date
WO2021110169A1 true WO2021110169A1 (zh) 2021-06-10

Family

ID=76221475

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/134286 WO2021110169A1 (zh) 2019-12-06 2020-12-07 作为erk抑制剂的噻唑并内酰胺类化合物及其应用

Country Status (9)

Country Link
US (1) US20230072937A1 (zh)
EP (1) EP4071155A4 (zh)
JP (1) JP7409719B2 (zh)
KR (1) KR20220107249A (zh)
CN (1) CN114829365B (zh)
AU (1) AU2020398022B2 (zh)
CA (1) CA3160903A1 (zh)
TW (1) TWI758999B (zh)
WO (1) WO2021110169A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022253186A1 (zh) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 一种二甲基取代的噻唑并吡咯酮类化合物的晶型及其制备方法
WO2023274256A1 (zh) * 2021-06-28 2023-01-05 南京明德新药研发有限公司 噻唑并内酰胺并螺杂环类化合物及其应用
WO2023274088A1 (zh) * 2021-06-28 2023-01-05 南京明德新药研发有限公司 二甲基取代的噻唑并内酰胺类化合物及其应用

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108349983A (zh) * 2015-11-09 2018-07-31 阿斯利康(瑞典)有限公司 治疗癌症有用的二氢咪唑并吡嗪酮衍生物
CN109608444A (zh) * 2018-11-27 2019-04-12 中国药科大学 含异吲哚啉酮的erk抑制剂及其制备方法与用途
WO2019223632A1 (en) * 2018-05-22 2019-11-28 Js Innomed Holdings Ltd. Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
WO2019233457A1 (zh) * 2018-06-08 2019-12-12 贝达药业股份有限公司 Erk抑制剂及其应用
CN110577543A (zh) * 2018-10-10 2019-12-17 深圳市塔吉瑞生物医药有限公司 一种二氢咪唑并吡嗪酮化合物及包含该化合物的组合物及其用途
CN110950876A (zh) * 2019-12-10 2020-04-03 如东凌达生物医药科技有限公司 一类呋喃并内酰胺类化合物、制备方法和用途
WO2020200069A1 (zh) * 2019-03-29 2020-10-08 江苏恒瑞医药股份有限公司 吡咯并杂环类衍生物、其制备方法及其在医药上的应用

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101864908B1 (ko) * 2010-09-01 2018-06-05 질레드 코네티컷 인코포레이티드 피리다지논, 그의 제조 방법 및 사용 방법
US10457669B2 (en) * 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108349983A (zh) * 2015-11-09 2018-07-31 阿斯利康(瑞典)有限公司 治疗癌症有用的二氢咪唑并吡嗪酮衍生物
WO2019223632A1 (en) * 2018-05-22 2019-11-28 Js Innomed Holdings Ltd. Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
WO2019233457A1 (zh) * 2018-06-08 2019-12-12 贝达药业股份有限公司 Erk抑制剂及其应用
CN110577543A (zh) * 2018-10-10 2019-12-17 深圳市塔吉瑞生物医药有限公司 一种二氢咪唑并吡嗪酮化合物及包含该化合物的组合物及其用途
CN109608444A (zh) * 2018-11-27 2019-04-12 中国药科大学 含异吲哚啉酮的erk抑制剂及其制备方法与用途
WO2020200069A1 (zh) * 2019-03-29 2020-10-08 江苏恒瑞医药股份有限公司 吡咯并杂环类衍生物、其制备方法及其在医药上的应用
CN110950876A (zh) * 2019-12-10 2020-04-03 如东凌达生物医药科技有限公司 一类呋喃并内酰胺类化合物、制备方法和用途

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CANCER DISCOV, vol. 9, 2019, pages 329 - 341
JI DEZHONG; ZHANG LINGZHI; ZHU QIHUA; BAI YING; WU YAOYAO; XU YUNGEN: "Discovery of potent, orally bioavailable ERK1/2 inhibitors with isoindolin-1-one structure by structure-based drug design", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 164, 1 January 1900 (1900-01-01), AMSTERDAM, NL, pages 334 - 341, XP085584645, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2018.12.040 *
RICHARD A. WARD, PAUL BETHEL, CALUM COOK, EMMA DAVIES, JUDIT E. DEBRECZENI, GARY FAIRLEY, LYMAN FERON, VIKKI FLEMINGTON, MARK A. G: "Structure-Guided Discovery of Potent and Selective Inhibitors of ERK1/2 from a Modestly Active and Promiscuous Chemical Start Point", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, vol. 60, no. 8, 27 April 2017 (2017-04-27), pages 3438 - 3450, XP055763583, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.7b00267 *
See also references of EP4071155A4

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022253186A1 (zh) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 一种二甲基取代的噻唑并吡咯酮类化合物的晶型及其制备方法
WO2023274256A1 (zh) * 2021-06-28 2023-01-05 南京明德新药研发有限公司 噻唑并内酰胺并螺杂环类化合物及其应用
WO2023274088A1 (zh) * 2021-06-28 2023-01-05 南京明德新药研发有限公司 二甲基取代的噻唑并内酰胺类化合物及其应用
TWI825811B (zh) * 2021-06-28 2023-12-11 大陸商德昇濟醫藥(無錫)有限公司 噻唑並內醯胺並螺雜環類化合物及其應用
TWI836486B (zh) * 2021-06-28 2024-03-21 大陸商德昇濟醫藥(無錫)有限公司 二甲基取代的噻唑並內醯胺類化合物及其應用

Also Published As

Publication number Publication date
TWI758999B (zh) 2022-03-21
JP2023505288A (ja) 2023-02-08
US20230072937A1 (en) 2023-03-09
EP4071155A4 (en) 2024-01-17
AU2020398022B2 (en) 2023-04-27
CA3160903A1 (en) 2021-06-10
CN114829365B (zh) 2023-04-25
EP4071155A1 (en) 2022-10-12
KR20220107249A (ko) 2022-08-02
TW202128710A (zh) 2021-08-01
AU2020398022A1 (en) 2022-07-14
JP7409719B2 (ja) 2024-01-09
CN114829365A (zh) 2022-07-29

Similar Documents

Publication Publication Date Title
WO2021110169A1 (zh) 作为erk抑制剂的噻唑并内酰胺类化合物及其应用
WO2020228817A1 (zh) Erk抑制剂及其应用
WO2021110168A1 (zh) 作为erk抑制剂的螺环类化合物及其应用
CN114761407B (zh) 作为高选择性ros1抑制剂的化合物及其应用
CN113993590B (zh) 新型egfr抑制剂
WO2021057893A1 (zh) 作为选择性btk激酶抑制剂的吡唑并吡啶类化合物
WO2019201296A1 (zh) 作为rho激酶抑制剂的吡唑类化合物
RU2805569C1 (ru) Тиазололактамные соединения в качестве ингибиторов ERK и их применение
WO2023274088A1 (zh) 二甲基取代的噻唑并内酰胺类化合物及其应用
WO2023274256A1 (zh) 噻唑并内酰胺并螺杂环类化合物及其应用
WO2023083200A1 (zh) 吡唑并环化合物及其应用
CN113801120B (zh) Mdm2抑制剂的微悬浮液以及治疗应用
CN114945576B (zh) 氘代噻吩并吡啶类化合物
JP7391226B2 (ja) ピリミジン化合物の結晶
CN114957259A (zh) 氰基取代的芳香双环类化合物及其应用
WO2021098734A1 (zh) 作为atm抑制剂的有取代的喹啉吡咯酮类合物及其应用
CN113801135A (zh) 噻吩并吡唑类化合物,包含其的药物组合物及其用途

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: 20895381

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022534190

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3160903

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 20227021797

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020895381

Country of ref document: EP

Effective date: 20220706

ENP Entry into the national phase

Ref document number: 2020398022

Country of ref document: AU

Date of ref document: 20201207

Kind code of ref document: A