WO2023103906A1 - Kras g12d抑制剂及其在医药上的应用 - Google Patents

Kras g12d抑制剂及其在医药上的应用 Download PDF

Info

Publication number
WO2023103906A1
WO2023103906A1 PCT/CN2022/136197 CN2022136197W WO2023103906A1 WO 2023103906 A1 WO2023103906 A1 WO 2023103906A1 CN 2022136197 W CN2022136197 W CN 2022136197W WO 2023103906 A1 WO2023103906 A1 WO 2023103906A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
halogen
alkyl
substituted
cycloalkyl
Prior art date
Application number
PCT/CN2022/136197
Other languages
English (en)
French (fr)
Inventor
吴颢
陈小平
路渊
徐人奇
匡翠文
李波燕
何将旗
杨晓峰
赵志昌
杨翔
杜亚军
李腾飞
湛波
张运来
张洪波
王冬
王国建
沈少聪
黄传龙
周全
王家炳
兰宏
丁列明
Original Assignee
贝达药业股份有限公司
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 贝达药业股份有限公司 filed Critical 贝达药业股份有限公司
Publication of WO2023103906A1 publication Critical patent/WO2023103906A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/16Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings

Definitions

  • the present invention relates to a novel compound which has KRAS inhibitory activity, especially KRAS G12D inhibitory activity.
  • the invention also relates to processes for the preparation of these compounds and pharmaceutical compositions containing them.
  • RAS is the gene with the highest mutation rate in human tumors. About 20-30% of all tumors have RAS mutations, about 98% of pancreatic cancer, 52% of colon cancer, 43% of multiple myeloma, And 32% of lung adenocarcinomas have RAS gene mutations.
  • the most common mutation of RAS is point mutation, which often occurs at codons 12, 13, and 61, and the mutation at codon 12 is the most common, such as G12C, G12D or G12V.
  • KRAS G12C inhibitors AMG510 (WO2018217651A1) and MRTX849 (WO2019099524A1) have entered late-stage clinical trials.
  • KRAS has always been a target of interest for drug developers. Despite the progress made in this field, there remains a need in the art for improved KRAS G12D mutein inhibitors.
  • the invention provides a small molecular KRAS G12D inhibitor with a novel structure, which has good antitumor activity.
  • the present invention provides a compound represented by general formula (I), its stereoisomer, tautomer, deuterated substance or pharmaceutically acceptable salt:
  • X 1 is selected from CR 4 or N;
  • R 4 is selected from H, amino, substituted amino, cyano, C 1-6 alkyl, substituted C 1-6 alkyl, halogen, C 2-6 alkenyl, substituted C 2-6 alkenyl, C 3-6 cycloalkyl or substituted C 3-6 cycloalkyl;
  • X 2 is selected from CR 5 or N;
  • R 5 is selected from H, amino, substituted amino, cyano, C 1-6 alkyl, substituted C 1-6 alkyl, halogen, C 2-6 alkenyl, substituted C 2-6 alkenyl, C 3-6 cycloalkyl or substituted C 3-6 cycloalkyl;
  • X 3 is selected from CR 6 or N;
  • R 6 is selected from H, amino, substituted amino, cyano, C 1-6 alkyl, substituted C 1-6 alkyl, halogen, C 2-6 alkenyl, substituted C 2-6 alkenyl, C 3-6 cycloalkyl or substituted C 3-6 cycloalkyl;
  • X 4 is selected from a bond, O, NR 7 , C(R 7 ) 2 or C(O); each R 7 is independently selected from H, amino, substituted amino, cyano, C 1-6 alkyl, substituted C 1-6 alkyl, halogen, C 2-6 alkenyl, substituted C 2-6 alkenyl, C 3-6 cycloalkyl or substituted C 3-6 cycloalkyl;
  • X 5 is selected from a bond, O, NR 8 , C(R 8 ) 2 or C(O); each R 8 is independently selected from H, amino, substituted amino, cyano, C 1-6 alkyl, substituted C 1-6 alkyl, halogen, C 2-6 alkenyl, substituted C 2-6 alkenyl, C 3-6 cycloalkyl or substituted C 3-6 cycloalkyl;
  • L is selected from a bond, -OC 0-4 alkylene-, -SC 0-4 alkylene-, -NR 9 -C 0-4 alkylene- or C 2-4 alkynylene, the -OC 0- 4 alkylene-, -SC 0-4 alkylene-, -NR 9 -C 0-4 alkylene- or C 2-4 alkynylene are optionally substituted by one or more R 9 ;
  • R 9 is selected from H, halogen, cyano, hydroxyl or C 1-6 alkyl, or two R 9 on the same carbon atom together with the atoms they are connected to form a C 3-6 cycloalkyl or 3-6 membered Heterocyclyl;
  • R is selected from H, hydroxyl, cyano, halogen or C 1-3 alkyl, and the C 1-3 alkyl is optionally substituted by one or more substituents selected from halogen, hydroxyl or cyano;
  • R 2 is selected from -OR a , -OC(O)N(R a ) 2 , -N(R a ) 2 , -NR a C(O)R a , -NR a C(O)N(R a ) 2.
  • R 10 is selected from H, cyano, Halogen, C 1-6 alkyl, -C 0-6 alkylene-OR a , -C 0-6 alkylene-OC(O)N(R a ) 2 , -C 0-6 alkylene- N(R a ) 2 , -C 0-6 al
  • R 3 is selected from C 6-14 aryl or 5-14 membered heteroaryl, said aryl or heteroaryl is optionally further substituted by one or more R 11 ;
  • R a is each independently selected from H, halogen, hydroxyl, amino, oxo, nitro, cyano, carboxyl, C 1-6 alkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 heteroalkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6- 14 aryl or 5-14 membered heteroaryl;
  • n is selected from 0, 1, 2, 3 or 4.
  • R a is each independently selected from H, halogen, hydroxyl, amino, oxo, nitro, cyano, carboxyl, C 1-6 alkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 heteroalkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6- 14 aryl or 5-14 membered heteroaryl.
  • a compound represented by general formula (I), its tautomer, a deuterated substance or a pharmaceutically acceptable salt thereof is selected from compounds represented by formula (IA)-formula (ID) Compounds, Stereoisomers, Tautomers, Deuterated Substances or Pharmaceutically Acceptable Salts thereof:
  • R 2 , R 3 , R 4 , R 5 , R 6 , and L are as defined above.
  • a compound represented by general formula (I), its stereoisomer, tautomer, deuterated substance or pharmaceutically acceptable salt wherein, the R 2 is 3-14 Member condensed heterocyclic group, the 3-14 membered condensed heterocyclic group is optionally further substituted by one or more R 10 , the R 10 is selected from halogen or C 2-6 alkenyl, the alkenyl is optionally Optionally, it can also be substituted by 1 or more R a ; each of the R a is independently selected from H or halogen.
  • a compound represented by general formula (I), its stereoisomer, tautomer, deuterium or pharmaceutically acceptable salt wherein the fused heterocyclic group is selected from The fused heterocyclic group is optionally further substituted by one or more R 10 , the R 10 is selected from H, halogen or C 2-6 alkenyl, and the alkenyl is optionally substituted by 1 or Substituted by a plurality of R a ; each of the R a is independently selected from H or halogen.
  • a compound described in general formula (I), its stereoisomer, tautomer, deuterated substance or pharmaceutically acceptable salt thereof wherein, the R3 is selected from said Optionally further substituted by one or more R 11 selected from H, cyano, halogen, -C 0-6 alkylene-OR a , C 1-6 alkyl, C 1-6 haloalkane C 2-6 alkenyl, C 2-6 alkynyl or -C 0-6 alkylene-C 3-14 cycloalkyl, the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene-C 3-14 cycloalkyl is optionally further substituted by one or more R a ; each of the R a is independently selected from H or halogen.
  • a compound represented by general formula (I), its stereoisomer, tautomer, deuterated substance or pharmaceutically acceptable salt is selected from (IA-1), ( Compounds represented by IA-2), (IB-1), (IB-2), (IC-1), (IC-2), (ID-1) or (ID-2):
  • each of the R a is independently selected from H or halogen, and the halogen is preferably F;
  • the R 4 are each independently selected from H, cyano or halogen, and the halogen is preferably F;
  • the R 5 are each independently selected from H, C 1-6 alkyl or halogen, the halogen is preferably F;
  • the R 6 are each independently selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 3-6 cycloalkyl or halogen, the halogen is preferably F;
  • the R 3 is selected from C 6-14 aryl or 5-14 membered heteroaryl, and the C 6-14 aryl or C 6-14 heteroaryl is optionally further substituted by one or more R 11 ;
  • the C 6-14 aryl or 5-14 membered heteroaryl is preferably selected from The R 11 is selected from H, cyano, halogen, -C 0-6 alkylene-OR a , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 Alkynyl or -C 0-6 alkylene -C 3-14 cycloalkyl, said C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene
  • the base-C 3-14 cycloalkyl is optionally further substituted by 1 or more R a ; each of the R a is independently selected from H or halogen;
  • R 10 is selected from H, halogen or C 1-6 alkyl.
  • the present invention also provides a pharmaceutical composition, wherein the pharmaceutical composition comprises a therapeutically effective amount of at least one compound represented by formula (I), its stereoisomer, tautomer or pharmaceutically acceptable salt .
  • the present invention provides the application of the compound represented by the structural formula (I) or the pharmaceutical composition in the preparation of medicine.
  • the present invention further provides the preferred technical scheme of said application:
  • the application is in the preparation of medicines for treating and/or preventing cancer.
  • the application is the application of preparing medicines for treating diseases mediated by KRAS G12D.
  • said disease is cancer.
  • the cancer is selected from breast cancer, multiple myeloma, bladder cancer, endometrial cancer, gastric cancer, cervical cancer, rhabdomyosarcoma, non-small cell lung cancer, small cell lung cancer, pleomorphic lung cancer, ovarian cancer, esophagus Carcinoma, melanoma, colorectal cancer, hepatoma, head and neck tumors, hepatocholangiocarcinoma, myelodysplastic syndrome, glioblastoma, prostate cancer, thyroid cancer, Schwann cell tumor, lung squamous cell Carcinoma, lichenoid keratosis, synovial sarcoma, skin cancer, pancreatic cancer, testicular cancer, or liposarcoma.
  • the present invention also provides a method for treating and/or preventing diseases, comprising administering a therapeutically effective amount of at least one compound represented by the structural formula (I) or a pharmaceutical composition containing it to a subject.
  • the present invention also provides a method for treating and/or preventing diseases mediated by KRAS G12D, comprising administering a therapeutically effective amount of at least one compound represented by structural formula (I) or a pharmaceutical composition containing it to a subject.
  • the present invention also provides a method for treating cancer, comprising administering a therapeutically effective amount of at least one compound represented by structural formula (I) or a pharmaceutical composition containing it to a subject.
  • the KRAS G12D-mediated disease is cancer.
  • the cancer is selected from breast cancer, multiple myeloma, bladder cancer, endometrial cancer, gastric cancer, cervical cancer, rhabdomyosarcoma, non-small cell lung cancer, small cell lung cancer, pleomorphic Lung cancer, ovarian cancer, esophageal cancer, melanoma, colorectal cancer, hepatocellular carcinoma, head and neck cancer, hepatocholangiocarcinoma, myelodysplastic syndrome, malignant glioma, prostate cancer, thyroid cancer, Schwann cell tumor, squamous cell carcinoma of the lung, lichenoid keratosis, synovial sarcoma, skin cancer, pancreatic cancer, testicular cancer, or liposarcoma.
  • halogen refers to fluorine, chlorine, bromine or iodine, unless otherwise specified.
  • alkyl includes linear or branched monovalent saturated hydrocarbon groups.
  • alkyl includes methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 3-(2-methyl)butyl, 2 -pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl, 2-methylpentyl and the like.
  • “ 1-6 " in "C 1-6 alkyl” refers to a group comprising 1, 2, 3, 4, 5 or 6 carbon atoms arranged in a linear or branched chain.
  • alkylene refers to a divalent alkyl linking group. Alkylene formally refers to an alkane in which the two CH bonds are replaced by the point of attachment of the alkylene to the rest of the compound. Similarly, “C 1-3 " in C 1-3 alkylene refers to an alkylene group containing 1, 2 or 3 carbon atoms, including but not limited to methylene, 1,2-ethylene, 1,3-propylene or 1,2-isopropylidene.
  • Alkoxy refers to the oxygen ether form of the aforementioned linear or branched alkyl, ie -O-alkyl.
  • haloalkyl refers to an alkyl group in which one or more H has been replaced by a halogen atom.
  • haloalkoxy refers to the group -O-haloalkyl.
  • oxo or "oxo” refers to an oxygen atom in the form of a divalent substituent which, when attached to C, forms a carbonyl group, which, when attached to a heteroatom, forms a sulfoxide or sulfone or N-oxide group group.
  • aromatic ring refers to an aromatic character (having (4n+2) delocalized ⁇ electrons, where n is integer) carbocyclic or heterocyclic polyunsaturated ring.
  • aryl in the present invention, unless otherwise stated, refers to an unsubstituted or substituted monocyclic or condensed ring aromatic group including carbocyclic atoms. Preferably C 6-12 aryl, more preferably aryl is C 6-10 monocyclic or bicyclic aromatic ring group. Preferred are phenyl and naphthyl.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl, or cycloalkyl, where the ring bonded to the parent structure is an aryl ring, non-limiting examples include, but are not limited to, benzocyclopentyl.
  • heterocyclyl refers to a ring system having at least one cyclized alkyl or cyclized alkenyl group containing a heteroatom selected from N, O and/or S.
  • the heterocyclic group may comprise a monocyclic or polycyclic ring (eg, having 2, 3 or 4 fused rings, spiro rings, bridged rings, etc.).
  • a heterocyclyl group can be attached to the rest of the compound via a ring-forming carbon atom or a ring-forming heteroatom.
  • a 3-14-membered heterocyclic group is preferred, and "3-14-membered" in a 3-14-membered heterocyclic group refers to a heterocyclic group composed of 3-14 C, N, O or S ring-forming atoms; more preferably 3-8 membered heterocyclic group. Wherein nitrogen or sulfur heteroatoms can be selectively oxidized, and nitrogen heteroatoms can be selectively quaternized.
  • heterocyclic groups include, but are not limited to Azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, tetrahydrofuranyl, dioxolanyl, tetrahydroimidazolyl, tetrahydrothiazolyl, Tetrahydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone and tetrahydrooxadiazolyl.
  • the heterocyclyl may be fused to an aryl, heteroaryl or cycloalkyl ring, where the ring bonded to the parent structure is the heterocyclyl.
  • heteroaryl in the present invention, unless otherwise stated, refers to a monocyclic or polycyclic (eg fused bicyclic) aromatic heterocycle having at least one heteroatom selected from N, O and/or S, and wherein the nitrogen or sulfur heteroatom can be optionally oxidized, the nitrogen heteroatom can be optionally quaternized.
  • a 5-14 membered heteroaryl group is preferred, wherein “5-14 membered” in the 5-14 membered heteroaryl group refers to a heteroaryl group consisting of 5-14 C, N, O or S ring atoms. More preferred are 5-10 membered heteroaryl groups.
  • heteroaryl groups include, but are not limited to, thienyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, pyridyl, pyridyl, Azinyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, benzofuryl, benzothienyl, benzisoxazolyl, benzothiazolyl, benzothiazolyl, benzene Thiadiazolyl, benzotriazolyladenine, quinolinyl or isoquinolinyl.
  • the heteroaryl group may be fused to an aryl, heterocyclyl or cycloalkyl ring where the ring attached to the parent structure is a heteroaryl ring.
  • cycloalkyl refers to a ring system having at least one cycloalkyl group.
  • a C 3-12 cycloalkyl group is preferred, wherein “C 3-12 " means that the cycloalkyl group can have 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 ring atoms.
  • Cycloalkyl groups can include monocyclic and polycyclic rings (eg, having 2, 3 or 4 fused rings, spiro rings, bridged rings, etc.).
  • Cycloalkyl groups in some embodiments include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, etc.; said cycloalkyl groups can also be fused to aryl, heterocyclyl or heteroaryl rings, where the parent structure The rings joined together are cycloalkyl groups.
  • substituted means that one or more hydrogen atoms in the group are respectively replaced by the same or different substituents.
  • the substituents are independently selected from the group consisting of -F, -Cl, -Br, -I, -OH, trifluoromethoxy, ethoxy, propoxy, isopropoxy, n-butoxy Butyl, isobutoxy, tert-butoxy, -SCH 3 , -SC 2 H 5 , formaldehyde, -C(OCH 3 ), cyano, nitro, -CF 3 , -OCF 3 , amino, dimethyl Amino, methylthio, sulfonyl and acetyl groups.
  • linking group When the number of a linking group is 0, such as -(CH 2 ) 0 -, it means that the linking group is a bond.
  • pharmaceutically acceptable salt refers to a salt prepared from a pharmaceutically acceptable non-toxic base or acid.
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper (superior and subvalent), ferric, ferrous, lithium, magnesium, manganese (superior and subvalent), potassium, sodium, zinc, and the like. Particular preference is given to the ammonium, calcium, magnesium, potassium and sodium salts.
  • Nontoxic organic bases from which pharmaceutically acceptable salts can be derived include primary, secondary and tertiary amines, also cyclic amines and substituted amines, such as naturally occurring and synthetic substituted amines.
  • non-toxic organic bases capable of forming salts, including ion exchange resins and arginine, betaine, caffeine, choline, N',N'-dibenzylethylenediamine, diethylamine, 2 -Diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, reduced glucosamine, glucosamine, histidine, isopropylamine, lysine acid, methylglucamine, morpholine, piperazine, piperidine, polyamine resin, procaine, chloroprocaine, purine, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine wait.
  • ion exchange resins and arginine betaine, caffeine, choline, N',N'-dibenzylethylenediamine, diethylamine, 2 -Diethylamino
  • the compound provided by the present invention is a base
  • its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids and organic acids.
  • acids include, for example, acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, formic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, hydrochloric acid, isethionic acid , lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, pamoic acid, pantothenic acid, phosphoric acid, succinic acid, sulfuric acid, oxalic acid, propionic acid, glycolic acid, hydroiodic acid, perchloric acid, Cyclohexanesulfonic acid, salicylic acid, 2-naphthalenesulfonic acid, saccharinic acid, trifluoro
  • prodrugs of the compounds of the present invention are included in the protection scope of the present invention.
  • the prodrugs refer to functional derivatives that are readily converted in vivo into the desired compound.
  • any pharmaceutically acceptable salt, ester, ester salt or other derivative of the compound of the present application which can directly or indirectly provide the compound of the present application or its pharmaceutically active metabolite or Residues.
  • the compounds of the present invention may contain one or more asymmetric centers and may thereby give rise to diastereoisomers and optical isomers.
  • the present invention includes all possible diastereoisomers and their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers and their pharmaceutically acceptable salts.
  • the present invention includes any possible tautomers and their pharmaceutically acceptable salts, and their mixtures.
  • substitution of compounds of formula (I) with heavier isotopes may afford certain therapeutic advantages due to greater metabolic stability, eg increased in vivo half-life or reduced dosage requirements.
  • pharmaceutical composition refers to a mixture of one or more compounds of the present application or their pharmaceutically acceptable salts and pharmaceutically acceptable auxiliary materials.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound of the present application to an organism.
  • compositions comprising "a” pharmaceutically acceptable excipient may be interpreted to mean that the pharmaceutical composition includes “one or more” pharmaceutically acceptable excipients.
  • pharmaceutically acceptable excipients refers to those excipients that have no obvious stimulating effect on the organism and will not impair the biological activity and performance of the active compound. Suitable excipients are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the pharmaceutical composition of the present invention can be prepared by combining the compound of the present application with suitable pharmaceutically acceptable auxiliary materials, for example, it can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, powders , granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols, etc.
  • Typical routes of administration of a compound of the invention, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, Subcutaneous, intravenous administration.
  • treatment generally refers to obtaining a desired pharmacological and/or physiological effect.
  • the effect may be therapeutic in terms of partial or complete stabilization or cure of the disease and/or side effects due to the disease.
  • treatment encompasses any treatment of a disease in a patient, including: (a) inhibiting the symptoms of the disease, ie arresting its development; or (b) relieving the symptoms of the disease, ie causing regression of the disease or symptoms.
  • the term "effective amount” means (i) treating or preventing a particular disease, condition or disorder, (ii) alleviating, ameliorating or eliminating one or more symptoms of a particular disease, condition or disorder, or (iii) preventing or delaying the The amount of a compound of the application for the onset of one or more symptoms of a particular disease, condition or disorder described in .
  • the amount of a compound of the present application that constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by a person skilled in the art according to its own knowledge and this disclosure.
  • DBU 1,8-diazabicycloundec-7-ene
  • DIEA N,N-Diisopropylethylamine
  • Dioxane dioxane
  • THF Tetrahydrofuran
  • TFA trifluoroacetic acid
  • TEA triethylamine
  • Pre-TLC preparation of thin-layer plates
  • CDI N,N'-carbonyldiimidazole
  • NCS N-chlorosuccinimide
  • Pd(DPEPhos)Cl 2 bis(diphenylphosphine phenyl ether) dichloropalladium(II);
  • CPME cyclopentyl methyl ether
  • TBDPSCl tert-butyldiphenylchlorosilane
  • TBSCl tert-butyldimethylsilyl chloride
  • DABCO triethylenediamine
  • m-CPBA m-chloroperoxybenzoic acid
  • LAH lithium aluminum hydride
  • POCl 3 phosphorus oxychloride
  • DIPEA N,N-Diisopropylethylamine
  • CsF cesium fluoride
  • compound M3-4 (187g) and difluoromethyl (2-pyridyl) sulfone (184g) were dissolved in anhydrous DMF (1.4L), and potassium tert-butoxide (107g) was added dropwise at -50°C DMF (460mL) solution, after the dropwise addition, control the temperature and react at -40°C for 2 hours. After the reaction, add saturated ammonium chloride solution dropwise at -50°C until the solution becomes weakly acidic, then naturally warm to room temperature and react for 18 hours. , filtered to obtain the filtrate, added EA (1.4 L) to dilute, then filtered to obtain the filtrate, and concentrated.
  • EA 1.4 L
  • reaction solution was lowered to 0°C, quenched with saturated ammonium chloride, then adjusted to pH 5 with dilute hydrochloric acid, concentrated tetrahydrofuran in vacuo, a large amount of solids would precipitate, filtered, and washed with EA (200ml) and PE (200ml) was beaten, filtered, and the filter cake was dried to obtain compound M4-5 (43.5g, yield 85.34%).
  • Example 97 Compound 2-amino-4-((6S,9R)-3-chloro-1-fluoro-13-((2R,7aS)-2-fluorotetrahydro-1H-pyrroline-7a(5H) -yl)methoxy)-5a,6,7,8,9,10-hexahydro-5H-6,9-epiaminoazacyclo[2',1':3,4][1,4] Synthesis of oxazol[5,6,7D]quinazolin-2-yl)-7-fluorobenzo[b]thiophene-3-carbonitrile
  • Example 98 Compound 5-ethynyl-6-fluoro-4-((6S, 9R)-1-fluoro-12-((2R, 7aS)-2-fluorotetrahydro-1H-pyrroline-7a(5H )-yl)methoxy)-5a,6,7,8,9,10-hexahydro-5H-4-oxo-3,10a,11,13,14-pentose-6,9-methylnonyl Synthesis of [1,8-ab]hepten-2-yl)naphthalene-2-ol
  • Example 99 Compound 2-Amino-4-((6S,9R)-3-chloro-13-((2-(difluoromethylene)tetrahydro-1H-pyrrolizin-7a(5H)-yl )methoxy)-1-fluoro-5a,6,7,8,9,10-hexahydro-5H-6,9-epiaminoazacyclo[2',1':3,4][1, 4] Synthesis of oxazine[5,6,7D]quinazolin-2-yl)-7-fluorobenzo[b]thiophene-3-carbonitrile
  • Example 100 Compound 2-amino-4-((6R, 9S)-3-chloro-1-fluoro-13-((2R, 7aS)-2-fluorotetrahydro-1H-pyrrolizine-7a(5H )-yl)methoxy)-5a,6,7,8,9,10-hexahydro-5H-6,9-epiaminoazacyclo[2',1':3,4][1,4 Synthesis of ]oxazol[5,6,7D]quinazolin-2-yl)-7-fluorobenzo[b]thiophene-3-carbonitrile
  • the synthesis of compound 100-1 refers to the synthesis of Example 97.
  • the KRas-G12D mutant tumor cell AGS ( CRL-1739 TM ) were spread in a low-adsorption 96-well plate at a cell density of 1 ⁇ 10 3 /well, and placed in a cell culture incubator for overnight culture. After the cells adhered to the wall, the compounds to be tested were added to the 96-well plate according to the final concentration of 10000, 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.025, and 0 nM (the final concentration of DMSO was 0.5%), and incubated at 37°C After 96 hours, add 50 ⁇ L of Cell-titer GLO working solution to each well, shake and mix well, and incubate at room temperature for 10 minutes. Read the Luminescence luminescence value on a multi-functional microplate reader, and convert the luminescence value data into inhibition percentage. And according to the following formula, calculate the percentage of inhibition of cell proliferation:
  • Inhibition percentage (maximum value - measured value) / (maximum value - Blank) ⁇ 100
  • Curve fitting was performed using GraphPad Prism software and IC50 values were obtained.
  • test results show that the compound of the present invention has good activity.
  • the KRas-G12D mutant tumor cell AGS ( CRL-1739 TM ) were plated in a 96-well plate at a cell density of 4 ⁇ 10 4 /well, and placed in a cell culture incubator for overnight culture. After the cells adhered to the wall, the compounds to be tested were added to the 96-well plate at the final concentration of 3000nM, 600nM, 120nM, 24nM, 4.8nM, 0.96nM, 0.19nM, and 0.1% DMSO. The lysate in lysate each treated cell sample (40ul/well) in the 96-well plate, and add 16ul/well protein solution and 4ul premixed pERK antibody to the HTRF detection 96-well plate after complete lysis. After overnight incubation at 4°C, the ratio signal value of 665nM/620nM was read on a multifunctional microplate reader to collect raw data. And according to the following formula, calculate the p-ERK inhibition percentage:
  • Inhibition percentage (maximum value - measured value) / (maximum value - Blank) ⁇ 100
  • Curve fitting was performed using GraphPad Prism software and IC50 values were obtained.

Abstract

提供了式(I)所述的化合物,其立体异构体、互变异构体、氘代物或药用盐,其制备方法,含有该化合物的药用组合物,及其在作为治疗和/或预防由KRAS介导的疾病的药物中的应用。

Description

KRAS G12D抑制剂及其在医药上的应用 技术领域
本发明涉及一种新型化合物,其具有KRAS抑制活性,特别是KRAS G12D抑制活性。本发明还涉及这些化合物的制备方法以及包含其的药物组合物。
背景技术
临床数据显示,RAS是人类肿瘤中发生突变率最高的基因,所有肿瘤中,约20-30%有RAS突变,大约98%的胰腺癌,52%的结肠癌,43%的多发性骨髓瘤,及32%的肺腺癌中存在RAS基因突变。RAS最常见的突变方式是点突变,经常发生在12、13、61密码子,其中又以第12位密码子突变最常见,例如G12C、G12D或G12V。
目前针对KRAS突变的药物研发是当前新药研究热点之一。KRAS G12C抑制剂AMG510(WO2018217651A1)和MRTX849(WO2019099524A1)已进入后期临床阶段。
基于KRAS异常激活在癌症进展中的重要性和KRAS基因突变在人类癌症中的普遍性,KRAS一直是药物研发人员关注的靶点。尽管己在这个领域中取得进展,但在本领域中仍需要改进的KRAS G12D突变蛋白抑制剂。
发明内容
本发明提供了一种新型结构的小分子KRAS G12D抑制剂,具有良好的抗肿瘤活性。
本发明提供一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐:
Figure PCTCN2022136197-appb-000001
其中,
X 1选自CR 4或N;R 4选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
X 2选自CR 5或N;R 5选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
X 3选自CR 6或N;R 6选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
X 4选自键、O、NR 7、C(R 7) 2或C(O);R 7各自独立地选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
X 5选自键、O、NR 8、C(R 8) 2或C(O);R 8各自独立地选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
L选自键、-O-C 0-4亚烷基-、-S-C 0-4亚烷基-、-NR 9-C 0-4亚烷基-或C 2-4亚炔基,所述-O-C 0- 4亚烷基-、-S-C 0-4亚烷基-、-NR 9-C 0-4亚烷基-或C 2-4亚炔基任选地被一个或多个R 9取代;R 9选自H、卤素、氰基、羟基或C 1-6烷基,或者同一个碳原子上的两个R 9连同其连接的原子共同形成C 3-6环烷基或3-6元杂环基;
R 1选自H、羟基、氰基、卤素或C 1-3烷基,所述C 1-3烷基任选地被一个多个选自卤素、羟基或氰基的取代基所取代;
R 2选自-OR a、-OC(O)N(R a) 2、-N(R a) 2、-NR aC(O)R a、-NR aC(O)N(R a) 2、-NR aS(O)R a、-NR aS(O) 2R a、-S(=O)R a、-S(=O) 2R a、-SR a、-S(R a) 5、-C(=O)R a、-C(=O)OR a、-C(=O)N(R a) 2、环烷基、杂环基、芳基或杂芳基,所述环烷基、杂环基、芳基或杂芳基任选地进一步被一个或多个R 10取代;R 10选自H、氰基、
Figure PCTCN2022136197-appb-000002
卤素、C 1-6烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-OC(O)N(R a) 2、-C 0-6亚烷基-N(R a) 2、-C 0-6亚烷基-NR aC(O)R a、-C 0-6亚烷基-NR aC(O)N(R a) 2、-C 0-6亚烷基-NR aS(O)R a、-C 0-6亚烷基-NR aS(O) 2R a、-C 0-6亚烷基-S(=O)R a、-C 0-6亚烷基-S(=O) 2R a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-S(R a) 5、-C 0-6亚烷基-C(=O)R a、-C 0-6亚烷基-C(=O)OR a、-C 0-6亚烷基-C(=O)N(R a) 2、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0- 6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基),所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基)任选地还可被1个或多个R a所取代;
R 3选自C 6-14芳基或5-14元杂芳基,所述芳基或杂芳基任选地进一步被一个或多个R 11取代;R 11选自H、氰基、卤素、C 1-6烷基、C 1-6卤代烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-OC(O)N(R a) 2、-C 0-6亚烷基-N(R a) 2、-C 0-6亚烷基-NR aC(O)R a、-C 0-6亚烷基-NR aC(O)N(R a) 2、-C 0-6亚烷基-NR aS(O)R a、-C 0-6亚烷基-NR aS(O) 2R a、-C 0-6亚烷基-S(=O)R a、-C 0-6亚烷基- S(=O) 2R a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-S(R a) 5、-C 0-6亚烷基-C(=O)R a、-C 0-6亚烷基-C(=O)OR a、-C 0-6亚烷基-C(=O)N(R a) 2、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基),所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基)任选地还可被1个或多个R a所取代;
R a各自独立地选自H、卤素、羟基、氨基、氧代基、硝基、氰基、羧基、C 1-6烷基、C 1-6羟基烷基、C 1-6氨基烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6杂烷基、C 3-8环烷基、3-8元杂环基、C 6-14芳基或5-14元杂芳基;
m选自0、1、2、3或4。
在本发明的一些实施方式中,所述R 10选自H、氰基、卤素、C 1-6烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-OC(O)N(R a) 2、-C 0-6亚烷基-N(R a) 2、-C 0-6亚烷基-NR aC(O)R a、-C 0-6亚烷基-NR aC(O)N(R a) 2、-C 0-6亚烷基-NR aS(O)R a、-C 0-6亚烷基-NR aS(O) 2R a、-C 0-6亚烷基-S(=O)R a、-C 0-6亚烷基-S(=O) 2R a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-S(R a) 5、-C 0-6亚烷基-C(=O)R a、-C 0-6亚烷基-C(=O)OR a、-C 0-6亚烷基-C(=O)N(R a) 2、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基),所述C 1- 6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基)任选地还可被1个或多个R a所取代;
R a各自独立地选自H、卤素、羟基、氨基、氧代基、硝基、氰基、羧基、C 1-6烷基、C 1-6羟基烷基、C 1-6氨基烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6杂烷基、C 3-8环烷基、3-8元杂环基、C 6-14芳基或5-14元杂芳基。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其互变异构体、氘代物或药用盐,其选自式(IA)-式(ID)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐:
Figure PCTCN2022136197-appb-000003
其中,R 2,R 3,R 4,R 5,R 6,L如上述所定义。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述L为-O-C 0-4亚烷基-,所述R 2为3-14元杂环基,所述3-14元杂环基任选地进一步被一个或多个R 10取代,所述R 10选自H、氰基、卤素、C 1-6烷基、C 2-6烯基、C 2-6炔基,所述C 1-6烷基、C 2-6烯基、C 2-6炔基任选地还可被1个或多个R a所取代;所述R a各自独立地选自H、卤素、羟基、氨基、氧代基、硝基、氰基或羧基。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 2为3-14元稠杂环基,所述3-14元稠杂环基任选地进一步被一个或多个R 10取代,所述R 10选自卤素或C 2-6烯基,所述烯基被任选地还可被1个或多个R a所取代;所述R a各自独立地选自H或卤素。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述稠杂环基选自
Figure PCTCN2022136197-appb-000004
Figure PCTCN2022136197-appb-000005
所述稠杂环基任选地进一步被一个或多个R 10取代,所述R 10选自H、卤素或C 2-6烯基,所述烯基被任选地还可被1个或多个R a所取代;所述R a各自独立地选自H或卤素。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 10
Figure PCTCN2022136197-appb-000006
所述R a各自独立地为卤素,优选为F。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 10
Figure PCTCN2022136197-appb-000007
所述R a为H。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 10为卤素,优选为F。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述
Figure PCTCN2022136197-appb-000008
选自
Figure PCTCN2022136197-appb-000009
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述L选自键、-O-C 0-4亚烷基-或-NR 9-C 0-4亚烷基,所述R 2选自-N(R a) 2、-C(=O)R a、-C(=O)N(R a) 2、C 3-12环烷基,所述C 3-12环烷基、任选地进一步被一个或多个R 10取代,所述R 10选自H、氰基、卤素、C 1-6烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-(3-14元杂环基),所述C 1-6烷基、-C 0-6亚烷基-(3-14元杂环基)任选地还可被1个或多个R a所取代,所述R a选自H、氰基、卤素或C 1-6烷基。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述
Figure PCTCN2022136197-appb-000010
选自
Figure PCTCN2022136197-appb-000011
Figure PCTCN2022136197-appb-000012
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 4各自独立选自H、氰基或卤素,所述卤素优选为F。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 5各自独立选自H、C 1-6烷基或卤素,所述卤素优选为F。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 6各自独立选自H、C 1-6烷基、C 2-6烯基、C 3-6环烷基或卤素,所述卤素优选为F。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述X 4选自键、NR 7、C(R 7) 2或C(O),所述R 7各自独立地选自H或甲基。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述X 5选自键或C(O)。
在本发明的一些实施方式中,一种通式(I)所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 1选自H。
在本发明的一些实施方式中,一种通式(I)所述的化合物、其立体异构体、互变异构体、 氘代物或药用盐,其中,所述R 3选自
Figure PCTCN2022136197-appb-000013
所述
Figure PCTCN2022136197-appb-000014
任选地进一步被一个或多个R 11取代,所述R 11选自H、氰基、卤素、-C 0-6亚烷基-OR a、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基或-C 0-6亚烷基-C 3-14环烷基,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基任选地进一步被1个或多个R a所取代;所述R a各自独立地选自H或卤素。
在本发明的一些实施方式中,一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其选自(IA-1)、(IA-2)、(IB-1)、(IB-2)、(IC-1)、(IC-2)、(ID-1)或(ID-2)所示化合物:
Figure PCTCN2022136197-appb-000015
Figure PCTCN2022136197-appb-000016
其中,所述R a各自独立地选自H或卤素,所述卤素优选为F;
所述R 4各自独立选自H、氰基或卤素,所述卤素优选为F;
所述R 5各自独立选自H、C 1-6烷基或卤素,所述卤素优选为F;
所述R 6各自独立选自H、C 1-6烷基、C 2-6烯基、C 3-6环烷基或卤素,所述卤素优选为F;
所述R 3选自C 6-14芳基或5-14元杂芳基,所述C 6-14芳基或C 6-14杂芳基任选地进一步被一个或多个R 11取代;所述C 6-14芳基或5-14元杂芳基优选自
Figure PCTCN2022136197-appb-000017
Figure PCTCN2022136197-appb-000018
所述R 11选自H、氰基、卤素、-C 0-6亚烷基-OR a、C 1-6烷基、C 1- 6卤代烷基、C 2-6烯基、C 2-6炔基或-C 0-6亚烷基-C 3-14环烷基,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基任选地进一步被1个或多个R a所取代;所述R a各自独立地选自H或卤素;
R 10选自H、卤素或C 1-6烷基。
在本发明的一些实施方式中,一种通式(I)化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,所述R 3选自
Figure PCTCN2022136197-appb-000019
Figure PCTCN2022136197-appb-000020
Figure PCTCN2022136197-appb-000021
在本发明的一些实施方式中,一种通式(I)化合物、其立体异构体、互变异构体、氘代物或药用盐,其中,通式(I)所示的化合物选自下式化合物:
Figure PCTCN2022136197-appb-000022
Figure PCTCN2022136197-appb-000023
本发明还提供了一种药物组合物,其中,所述药物组合物包含治疗有效量的至少一种式(I)所示的化合物、其立体异构体、互变异构体或药用盐。
本发明提供了结构式(I)所示化合物或药物组合物在制备药物中的应用。
本发明进一步提供了所述应用的优选技术方案:
作为优选,所述应用为制备治疗和/或预防癌症药物中的应用。
作为优选,所述应用为制备用于治疗由KRAS G12D介导的疾病的药物的应用。作为优选,所述疾病是癌症。
作为优选,所述癌症选自乳腺癌、多发性骨髓瘤、膀胱癌、子宫内膜癌、胃癌、宫颈癌、横纹肌肉瘤、非小细胞肺癌、小细胞肺癌、多形性肺癌、卵巢癌、食管癌、黑色素瘤、结肠直肠癌、肝细胞瘤、头颈部肿瘤、肝胆管细胞癌、骨髓增生异常综合征、恶性胶质瘤、前列腺癌、甲状腺癌、徐旺氏细胞瘤、肺鳞状细胞癌、苔藓样角化病、滑膜肉瘤、皮肤癌、胰腺癌、睾丸癌或脂肪肉瘤。
本发明还提供了一种治疗和/或预防疾病的方法,包括向治疗对象施用治疗有效量的至少任意一种结构式(I)所示化合物或含其的药物组合物。
本发明还提供了一种治疗和/或预防由KRAS G12D介导的疾病的方法,包括向治疗对象施用治疗有效量的至少任意一种结构式(I)所示化合物或含其的药物组合物。
本发明还提供了一种治疗癌症的方法,包括向治疗对象施用治疗有效量的至少任意一种结构式(I)所示化合物或含其的药物组合物。
作为优选,在上述方法中,所述KRAS G12D介导的疾病是癌症。
作为优选,在上述方法中,所述的癌症选自乳腺癌、多发性骨髓瘤、膀胱癌、子宫内膜癌、胃癌、宫颈癌、横纹肌肉瘤、非小细胞肺癌、小细胞肺癌、多形性肺癌、卵巢癌、食管癌、黑色素瘤、结肠直肠癌、肝细胞瘤、头颈部肿瘤、肝胆管细胞癌、骨髓增生异常综合征、恶性胶质瘤、前列腺癌、甲状腺癌、徐旺氏细胞瘤、肺鳞状细胞癌、苔藓样角化病、滑膜肉瘤、皮肤癌、胰腺癌、睾丸癌或脂肪肉瘤。
除非另有说明,所述结构通式中使用的一般化学术语具有通常的含义。
例如,除非另有说明,本发明所用的术语“卤素”是指氟、氯、溴或碘。
在本发明中,除非另有说明,“烷基”包括直链或支链的一价饱和烃基。例如,烷基包括甲基、乙基、丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、3-(2-甲基)丁基、2-戊基、2-甲基丁基、新戊基、正己基、2-己基、2-甲基戊基等。类似的,“C 1-6烷基”中的“ 1-6”是指包含有1、2、3、4、5或6个碳原子的直链或支链形式排列的基团。术语“亚烷基”是指二价烷基连接基团。亚烷基在形式上是指两个C-H键替换为亚烷基与化合物其余部分的连接点的烷烃。类似的,C 1-3亚烷基中的“C 1-3”是指含有1、2或3个碳原子的亚烷基,包括但不限于亚甲基、1,2-亚乙基、1,3-亚丙基或1,2-亚异丙基。
“烷氧基”是指前述的直链或支链烷基的氧醚形式,即-O-烷基。
术语“卤代烷基”是指一个或多个H已经被卤素原子置换的烷基。
术语“卤代烷氧基”是指-O-卤代烷基的基团。
术语“氧代”或“氧代基”是指呈二价取代基形式的氧原子,其与C连接时形成羰基, 其与杂原子连接时形成亚砜基或砜基或N-氧化物基团。
本发明中,除另有说明,术语“芳香环”、“芳香族环”或“芳香族杂环”即为具有芳香族特征(具有(4n+2)个非定域π电子,其中n为整数)的多不饱和环的碳环或杂环。
术语“芳基”,在本发明中,除非另有说明,是指未取代或取代的包括碳环的原子的单环或稠环芳香基团。优选C 6-12芳基,更优选芳基为C 6-10的单环或双环的芳香环基团。优选为苯基、萘基。所述芳基环可以稠合于杂芳基、杂环基或环烷基上,其中与母体结构连接在一起的环为芳基环,非限制性实例包括但不限于苯并环戊基。
术语“杂环基”是指具有至少一个含有杂环子的环化烷基或环化烯基的环系统,所述杂原子选自N、O和/或S。所述杂环基可以包括单环或多环(例如具有2、3或4个稠合环、螺环、桥环等)。杂环基可以经由成环碳原子或成环杂原子与化合物其他部分相连接。优选3-14元杂环基,3-14元杂环基中的“3-14元”是指含有3-14个C、N、O或S的成环原子组成的杂环基;更优选3-8元杂环基。其中氮或硫杂原子可以选择性地被氧化,并且氮杂原子可以选择性地被季铵化。这些杂环基的实例包括但不限于
Figure PCTCN2022136197-appb-000024
氮杂环丁烷基、吡咯烷基、哌啶基、哌嗪基、氧代哌嗪基、氧代哌啶基、四氢呋喃基、二氧戊环基、四氢咪唑基、四氢噻唑基、四氢恶唑基、四氢吡喃基、吗啉基、硫代吗啉基、硫代吗啉基亚砜、硫代吗啉基砜基和四氢恶二唑基。所述杂环基可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基。
术语“杂芳基”,在本发明中,除非另有说明,是指具有至少一个杂原子的单环或多环(例如稠合双环)芳香族杂环,所述杂原子选自N、O和/或S,并且其中所述氮或硫杂原子可以选择性地被氧化,所述氮杂原子可以选择性地被季铵化。优选5-14元杂芳基,其中5-14元杂芳基中的“5-14元”是指含有5-14个C、N、O或S的成环原子组成的杂芳基。更优选的是5-10元杂芳基。杂芳基的实例包括但不限于噻吩基、呋喃基、咪唑基、异恶唑基、恶唑基、吡唑基、吡咯基、噻唑基、噻二唑基、三唑基、吡啶基、哒嗪基、吲哚基、氮杂吲哚基、吲唑基、苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并异恶唑基、苯并噻唑基、苯并噻唑基、苯并噻二唑基、苯并三唑基腺嘌呤、喹啉基或异喹啉基。所述杂芳基可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环。
术语“环烷基”是指具有至少一个环化烷基的环系统。优选C 3-12环烷基,其中的“C 3-12” 是指环烷基可以具有3、4、5、6、7、8、9、10、11或12个成环原子。环烷基可以包括单环和多环(例如具有2、3或4个稠合环、螺环、桥环等)。一些实施例中环烷基包括但不限于环丙基、环丁基、环戊基等;所述环烷基还可以稠合于芳基、杂环基或杂芳基环上,其中与母体结构连接在一起的环为环烷基。
术语“取代的”是指基团中的一个或多个氢原子分别被相同的或者不同的取代基所取代。典型的取代基包括但不限于卤素(F、Cl、Br或I)、C 1-8烷基、C 3-12环烷基、-OR 1、-SR 1、=O、=S、-C(O)R 1、-C(S)R 1、=NR 1、-C(O)OR 1、-C(S)OR 1、-NR 1R 2、-C(O)NR 1R 2、氰基、硝基、-S(O) 2R 1、-O-S(O 2)OR 1、-O-S(O) 2R 1、-OP(O)(OR 1)(OR 2);其中R 1和R 2独立地选自-H、C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基。在一些实施例中,取代基独立地选自包含-F、-Cl、-Br、-I、-OH、三氟甲氧基、乙氧基、丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基、-SCH 3、-SC 2H 5、甲醛基、-C(OCH 3)、氰基、硝基、-CF 3、-OCF 3、氨基、二甲基氨基、甲硫基、磺酰基和乙酰基的基团。
当一个连接基团的数量为0时,比如-(CH 2) 0-表示该连接基团为键。
术语“药用盐”是指从药学上可接受的无毒的碱或酸制备的盐。
当本发明提供的化合物是酸时,可以从药学上可接受的无毒的碱,包括无机碱和有机碱,方便地制得其相应的盐。从无机碱衍生的盐包括铝、铵、钙、铜(高价和低价)、三价铁、亚铁、锂、镁、锰(高价和低价)、钾、钠、锌之类的盐。特别优选铵、钙、镁、钾和钠的盐。能够衍生成药学上可接受的盐的无毒有机碱包括伯胺、仲胺和叔胺,也包括环胺及含有取代基的胺,如天然存在的和合成的含取代基的胺。能够成盐的其他药学上可接受的无毒有机碱,包括离子交换树脂以及精氨酸、甜菜碱、咖啡因、胆碱、N',N'-二苄乙二胺、二乙胺、2-二乙氨基乙醇、2-二甲胺基乙醇、乙醇胺、乙二胺、N-乙基吗啉、N-乙基哌啶、还原葡萄糖胺、氨基葡萄糖、组氨酸、异丙胺、赖氨酸,甲基葡萄糖胺、吗啉、哌嗪、哌啶、多胺树脂、普鲁卡因、氯普鲁卡因、嘌呤、可可碱、三乙胺、三甲胺、三丙胺、氨丁三醇等。
当本发明提供的化合物是碱时,可以从药学上可接受的无毒的酸,包括无机酸和有机酸,方便制得其相应的盐。这样的酸包括,如,醋酸、苯磺酸、苯甲酸、樟脑磺酸、柠檬酸、乙磺酸、甲酸、富马酸、葡萄糖酸、谷氨酸、氢溴酸、盐酸、羟乙磺酸、乳酸、马来酸、苹果酸、扁桃酸、甲磺酸、黏酸、硝酸、扑酸、泛酸、磷酸、琥珀酸、硫酸、草酸、丙酸、乙醇酸、氢碘酸、高氯酸、环己氨磺酸、水杨酸、2-萘磺酸、糖精酸、三氟乙酸、酒石酸和对甲苯磺酸等。较优地,柠檬酸、氢溴酸、甲酸、盐酸、马来酸、磷酸、硫酸和酒石酸。更优地,甲酸和盐酸。
本发明化合物的药物前体包含在本发明的保护范围内。通常,所述药物前体是指很容易在体内转化成所需化合物的功能性衍生物。例如,本申请化合物的任何药学上可接受的盐、酯、酯的盐或其它衍生物,其在向受体施用后能够直接或间接地提供本申请的化合物或其具有药学活性的代谢物或残基。
本发明所述化合物可能含有一个或多个不对称中心,并可能由此产生非对映异构体和光学异构体。本发明包括所有可能的非对映异构体及其外消旋混合物、其基本上纯的拆分对映异构体、所有可能的几何异构体及其药用盐。
当式(I)所示化合物存在互变异构体时,除非特别声明,本发明包括任何可能的互变异构体和其药用盐,及它们的混合物。
当式(I)所示化合物用较重的同位素(例如氘)替代可能提供某些治疗优势,这是由于更大的代谢稳定性,例如增加体内半衰期或减少剂量要求。
术语“药物组合物”是指一种或多种本申请的化合物或其药用盐与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本申请的化合物。
在本发明中,“一”、“一个”、“该”、“至少一个”和“一个或多个”可互换使用。因此,例如,包含“一种”药学上可接受的辅料组成的混合物可以被解释为表示该药物组合物包括“一种或多种”药学上可接受的辅料。
术语“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。合适的辅料是本领域技术人员熟知的,例如碳水化合物、蜡、水溶性和/或水可膨胀的聚合物、亲水性或疏水性材料、明胶、油、溶剂、水等。
本发明的药物组合物可通过将本申请的化合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本发明化合物或其药用盐或其药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。
术语“治疗”一般是指获得需要的药理和/或生理效应。该效应根据部分或完全稳定或治愈疾病和/或由于疾病产生的副作用,可以是治疗性的。本文使用的“治疗”涵盖了对患者疾病的任何治疗,包括:(a)抑制疾病的症状,即阻止其发展;或(b)缓解疾病的症状,即,导致疾病或症状退化。
术语“有效量”意指(i)治疗或预防特定疾病、病况或障碍,(ii)减轻、改善或消除特定疾病、病况或障碍的一种或多种症状,或(iii)预防或延迟本文中所述的特定疾病、病况或障碍的一种 或多种症状发作的本申请化合物的用量。构成“治疗有效量”的本申请化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。
具体实施方式
为使上述内容更清楚、明确,本发明将用以下实施例来进一步阐述本发明的技术方案。以下实施例仅用于说明本发明的具体实施方式,以使本领域的技术人员能够理解本发明,但不用于限制本发明的保护范围。本发明的具体实施方式中,未作特别说明的技术手段或方法等为本领域的常规技术手段或方法等。
除非另有说明,本发明所有温度均指摄氏度。
实施例中使用了下列缩略语:
DBU:1,8-二氮杂二环十一碳-7-烯;
DIEA:N,N-二异丙基乙胺;
DCM:二氯甲烷;
Dioxane:二氧六环;
ESI-MS:电喷雾电离质谱;
MeOH:甲醇;
THF:四氢呋喃;
TFA:三氟乙酸;
TEA:三乙胺;
Pre-TLC:制备薄层板;
CDI:N,N'-羰基二咪唑;
NaH:氢化钠;
NCS:N-氯代丁二酰亚胺;
PyBOP:1H-苯并三唑-1-基氧三吡咯烷基六氟磷酸盐;
CBZCl:氯甲酸苄酯;
BH 3.THF:硼烷-四氢呋喃溶液;
Pd(DPEPhos)Cl 2:双(二苯基膦苯基醚)二氯化钯(II);
CPME:环戊基甲醚;
DMAP:4-二甲氨基吡啶;
TBDPSCl:叔丁基二苯基氯硅烷;
TBSCl:叔丁基二甲基氯硅烷;
DABCO:三乙烯二胺;
m-CPBA:间氯过氧苯甲酸;
PdCl 2(dppf):[1,1'-双(二苯基膦)二茂铁]二氯化钯;
Pre-TLC:薄层色谱分析法;
LAH:四氢铝锂;
EA:乙酸乙酯;
POCl 3:三氯氧磷;
DIPEA:N,N-二异丙基乙胺;
CsF:氟化铯;
BF 3·Et 2O:三氟化硼乙醚络合物。
中间体M1的合成:
Figure PCTCN2022136197-appb-000025
步骤1:化合物M1-1的合成
在室温下,将化合物5-氧吡咯烷-2-羧酸甲酯(25.00g)溶于二氯甲烷(250.00mL),加入三甲基氧鎓四氟硼酸盐(28.42g),在室温下反应3h。用饱和碳酸氢钠水溶液将pH调至8,分液,用DCM萃取一遍,再用饱和食盐水洗一遍,有机相干燥,过滤,浓缩即可得到化合物M1-1的粗品(20.30g,产率73.95%)。
步骤2:化合物M1-2的合成
在室温下,将化合物M1-1(42.40g)和2-硝基乙酸乙酯(107.72g)混合后,在60℃过夜反应。反应完成后,直接将反应液浓缩,经层析柱分离纯化得到黄色油状目标化合物M1-2(20.60g,产率29.57%)。
步骤3:化合物M1-3的合成
在室温下,将化合物M1-2(2.00g)溶于MeOH(20.00mL),加入湿Pd/C(1.65g),氢气置换,在室温下反应。待反应物反应完全,直接过滤反应液,得到滤液,将滤液浓缩,浓缩物经柱层析进行分离纯化得到黄色液体状化合物M1-3(0.89g,产率57.97%)。
步骤4:化合物M1-4的合成
在室温下,将化合物M1-3(7.05g)溶于DCM(70.00mL)中,加入DIEA(17.63mL),然后加入CbzCl(5.01mL),在0℃反应1h。直接将反应液浓缩后,经层析柱分离纯化得到目标化合物M1-4(7.86g,产率66.45%)。ESI-MS m/z:333[M+H] +
步骤5:化合物M1-5的合成
在室温下,将化合物M1-4(5.46g)溶于THF(50.00mL),MeOH(50.00mL)和水(50.00mL)的混合溶液中,加入NaOH(0.99g),在室温下反应1h。将反应液真空浓缩,用水稀释后,EA萃取,保留水相,加入稀盐酸调至pH=1,使用EA萃取两遍,饱和食盐水洗一遍,有机相干燥,过滤,浓缩即可得到白色固体状目标化合物M1-5(3.23g,产率64.56%)。ESI-MS m/z:305[M+H] +
步骤6:化合物M1的合成
在0℃下,将化合物M1-5(2.01g)溶于BH 3.THF(20.00mL)中,加完后将反应混合物移至40℃反应。待反应物完全反应,将反应液降至0℃,用甲醇淬灭反应液,淬灭完在50℃搅拌0.5h。将反应液浓缩,用DCM溶解后,饱和食盐水洗一遍,有机相干燥浓缩,浓缩物经柱层析进行分离纯化得到无色油状的目标中间体M1(0.65g,产率35.61%)。ESI-MS m/z:277[M+H] +
中间体M2的合成:
Figure PCTCN2022136197-appb-000026
步骤1:化合物M2-1的合成
将1-溴-2,5-二氟-3-硝基苯溶于乙醇(400ml)和水(80ml),加入氯化铵(28.1g),在搅拌的情况下加入铁粉(20.53g),在75℃下反应2h。反应完全后,将反应液冷却至室温,垫硅藻土过滤,用DCM洗涤滤渣,滤液,用水洗一遍,饱和食盐水洗一遍,有机相干燥,过 滤,浓缩得到化合物M2-1(21.13g收率96.70%)。粗品直接用于下一步。ESI-MS m/z:208.13[M+H] +
步骤2:化合物M2-2的合成
将盐酸羟胺(24.71g)和水合氯醛(25.2g)溶于水(435ml),在搅拌的情况下加入无水硫酸钠(115.43g)。将M2-1(21.13g)溶于乙醇(61ml)和水(35ml),加入浓盐酸(14.7ml),将其加入到反应瓶中,在60℃反应过夜。反应完全后,将反应液热过滤,得到滤饼,烘干得到化合物M2-2(25.10g收率88.55%)。粗品直接用于下一步。
步骤3:化合物M2-3的合成
将化合物M2-2(25.10g)加入到60℃的浓硫酸(250ml)中,加完将温度升至90℃。反应1h。反应完全后,将反应液冷却,缓慢加入到冰水中,大量固体析出,过滤得到固体,用EA溶解,饱和食盐水洗两遍,干燥,过滤,旋干得到化合物M2-3(16.07g收率68.19%)。粗品直接用于下一步。
步骤4:化合物M2-4的合成
将氢氧化钠(22.08g)溶于水(280ml),加入到装有化合物M2-3(16.07g)的反应瓶中,加完将温度降至0℃,加入双氧水(31ml),移至室温反应过夜。反应完全后,使用浓盐酸将pH调至7,过滤,得到滤液,继续使用浓盐酸将pH调至1,过滤得到固体,使用EA溶解,饱和食盐水洗一遍,干燥,过滤,浓缩得到化合物M 2-4(12.54g收率81.13%)。ESI-MS m/z:252.14[M+H] +
步骤5:化合物M2-5的合成
将化合物M2-4(12.34g)溶于DMF(120ml),加入N-氯代丁二酰亚胺(7.85g),在70℃反应0.5h。反应完全后,将反应液降至0℃,缓慢滴加水(480ml),产生大量固体,过滤得到固体,用EA溶解,饱和食盐水洗一遍,干燥,过滤,浓缩得到化合物M2-5(12.95g收率92.32%)。ESI-MS m/z:286.1[M+H] +
步骤6:化合物M2-6的合成
将化合物M2-5(12.46g)溶于THF(120ml),加入CDI(10.58g),在50℃反应0.5h,将反应液降至室温,缓慢滴加到冰的氨水(120ml)中,继续室温反应0.5h。反应完全后,用EA和水稀释反应液,再用EA萃取一遍,饱和食盐水洗一遍,干燥过滤浓缩,浓缩物通过柱层析进行纯化得到化合物M2-6(9.64g收率77.63%)。ESI-MS m/z:285.12[M+H] +
步骤7:化合物M2-7的合成
将化合物M2-6(4g)溶于THF(50ml)中,将温度升至40℃,分批加入NaH(1.4g),在40℃ 搅拌10分钟,分批加入N,N'-硫羰基二咪唑(3.75g),加完将温度升至60℃反应0.5h。反应完全后,使用饱和氯化铵淬灭反应液,使用稀盐酸将pH调至5-6,真空浓缩除掉四氢呋喃,会有大量固体析出,过滤得到固体,烘干即可得到化合物M2-7(3.79g收率82.58%)。
步骤8:化合物M2的合成
将化合物M2-7(3.59g)溶于甲醇(60ml),加入甲醇钠(0.89g)和碘甲烷(1.36ml),在室温下反应0.5h。反应完全后,往反应液中加入水(10ml),搅拌10分钟,过滤得到固体,烘干即可得到化合物M2(2.79g收率74.53%)。ESI-MS m/z:341.09[M+H] +
中间体M3的合成:
Figure PCTCN2022136197-appb-000027
步骤1:化合物M3-1的合成
室温下,将化合物M3-0(208g)溶解在无水MeOH(2L)中,在0℃滴加氯化亚砜(286mL),控制温度在5℃反应1小时,反应结束后,浓缩反应液,加入无水DCM(1L)稀释,0℃下,将稀释液体滴加到饱和碳酸氢钠溶液中,分液,有机层用饱和食盐水(500mL)洗涤,无水硫酸钠干燥,浓缩。浓缩物经柱色谱纯化(EA:DCM=0-50%)得到产物M3-1(240g,95%产率)。ESI-MS m/z=258.1[M+H] +
步骤2:化合物M3-2的合成
在室温下,将化合物M3-1(235g)溶于无水THF(2.4L),在0℃分批次加入四氢铝锂(69.4g),加完后60℃搅拌30min。反应结束后,冷却反应液,冰浴下滴加水(69.4mL),再滴加15%的氢氧化钠水溶液(69.4mL),最后滴加水(208.2mL),加入无水硫酸钠干燥,过滤得到滤液浓缩即得到产物M3-2(165g,90%产率)直接用于下一步。ESI-MS m/z=202.1[M+H] +
步骤3:化合物M3-3的合成
室温下,将化合物M3-2(160g)溶于三氟乙酸(500mL),加入水(67mL),60℃下反应过夜,浓缩反应液体得到粗品M3-3(320g,259%)直接用于下一步。ESI-MS m/z=156.1[M+H] +
步骤4:化合物M3-4的合成
在室温下,将化合物M3-3(308g)溶于DMF(350mL)中,0℃加入咪唑(540g)后滴加TBDPSCl(170mL),加完后室温搅拌1小时。反应结束后,加入水和EA稀释,用EA萃取水相3遍。合并有机相,有机相用饱和食盐水洗三遍,用无水硫酸钠干燥,过滤,浓缩。浓缩物经柱色谱纯化(EA:PE=0-15%)得到产物M3-4(192g,25%产率)。ESI-MS m/z=394.1[M+H] +
步骤5:化合物M3的合成
室温下,将化合物M3-4(187g)和二氟甲基(2-吡啶基)砜(184g)溶解在无水DMF(1.4L)中,在-50℃滴加叔丁醇钾(107g)的DMF(460mL)溶液,滴加完毕后控制温度在-40℃反应2小时,反应结束后,在-50℃下滴加饱和氯化铵溶液直至溶液成弱酸性,自然升温至室温反应18小时,过滤得到滤液,加入EA(1.4L)稀释,再过滤得到滤液,浓缩。浓缩物经柱色谱纯化(MeOH:DCM=0-10%)得到产物M3(60g,67%产率)。 1H NMR(500MHz,DMSO-d6)δ3.95-3.92(m,1H),3.70-3.67(m,1H),3.32-3.27(m,2H),2.94-2.89(m,1H),2.69-2.66(m,1H),2.50-3.45(m,1H),1.99-1.92(m,2H),1.88-1.75(m,2H)。ESI-MS m/z=190.1[M+H] +
中间体M4的合成:
Figure PCTCN2022136197-appb-000028
步骤1:化合物M4-1的合成
将2,6-二氯吡啶-4-胺(93.00g)溶于乙腈(700.00mL)和水(350.00mL),加入1-氯甲基-4-氟-1,4-重氮化二环2.2.2辛烷双(四氟硼酸)盐(242.54g),在55℃反应0.5h。反应完全后, 使用饱和亚硫酸钠淬灭,将反应液浓缩,用EA萃取两遍,饱和食盐水洗一遍,有机相干燥过滤浓缩,浓缩物经层析柱进行分离纯化得到化合物M4-1(52.2g收率50.55%)。ESI-MS m/z:181[M+H] +
步骤2:化合物M4-2的合成
将M4-1(52.2g)溶于乙腈(500.00mL),加入N-碘代丁二酰亚胺(77.86g)和对甲苯磺酸(4.97g),在70℃反应1小时。反应完全后,将反应液降至0℃,向反应液中缓慢滴加水(2500ml),有大量固体析出,过滤得到固体,用EA溶解固体,饱和食盐水洗一遍,有机相干燥过滤浓缩得到化合物M4-2(71.4g收率80.67%)。粗品直接用于下一步。ESI-MS m/z:307[M+H] +
步骤3:化合物M4-3的合成
将化合物M4-2(71.40g)溶于N,N-二甲基甲酰胺(400.00mL),加入氰化亚铜(25.00g),在125℃反应3h。反应完全后,将反应液冷却至0℃,往反应液中缓慢滴加水(2000ml),有大量固体析出,过滤得到固体,使用EA溶解固体,饱和食盐水洗一遍,过硅胶垫,滤液真空浓缩得到化合物M4-3(47g收率98.06%)。粗品直接用于下一步。
步骤4:化合物M4-4的合成
将化合物M4-3(47.00g)分批加入到浓硫酸(61.12mL),然后加入水(4.11mL),在60℃反应6小时。反应完全后,将反应液冷却,缓慢滴加到冰水(1000ml)中,有大量固体析出,过滤得到的固体,用EA溶解,饱和碳酸氢钠洗一遍,有机相干燥,过滤浓缩得到化合物M4-4(43.3g,收率84.72%)。粗品直接用于下一步。ESI-MS m/z:224[M+H] +
步骤5:化合物M4-5的合成
将化合物M4-4(42.91g)溶于四氢呋喃(500.00mL),将温度升至40℃,分批加入NaH(31.93g),加完在40℃搅拌10分钟,分批加入N,N'-硫羰基二咪唑(46.59g),加完升至60℃反应0.5小时。反应完全后,将反应液降至0℃,使用饱和氯化铵淬灭反应液,然后使用稀盐酸将pH调至5,真空浓缩四氢呋喃,会有大量固体析出,过滤,用EA(200ml)和PE(200ml)打浆,过滤,滤饼烘干得到化合物M4-5(43.5g收率85.34%)。
步骤6:化合物M4-6的合成
将化合物M4-5(43.50g)溶于乙腈(400.00mL),加入碘甲烷(15.27mL),最后加入甲醇钠(15.90g)的水(80.00mL)溶液。反应完全后,往反应液中缓慢加入水(1.6L),用稀盐酸将pH调至5,有大量固体析出,过滤,滤饼烘干得到化合物M4-6(39.3g收率85.82%)。ESI-MS m/z:280[M+H] +
步骤7:化合物M4-7的合成:
将化合物M4-6(1.2g)溶于POCl 3(15mL)中,然后将DIPEA(1.62mL)加入,105℃反应1.5h。反应完毕后,将POCl 3减压浓缩,然后将浓缩液慢慢加入到冷水中,DCM和水萃取2遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱层析分离纯化得目标化合物M4-7(760mg)。
步骤8:化合物M4-8的合成:
将化合物M4-7(300mg)和中间体M6(430mg)溶于ACN(5mL)中,然后将DIPEA(0.35mL)加入,室温反应30min。反应完毕后,加入DCM和水,DCM萃取2遍,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱层析分离纯化得目标化合物M4-8(563mg)。
步骤9:化合物M4-9的合成:
将化合物M4-8(560mg)溶于THF(5mL)中,然后将3HF·TEA(2.95mL)加入,室温反应30min。反应完毕后,加入饱和碳酸氢钠水溶液中和反应液,加入EA和水,EA萃取2遍,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱层析分离纯化得目标化合物M4-9(389mg)。
步骤10:化合物M4的合成:
将化合物M4-9(320mg)、DABCO(8mg)、Cs 2CO 3(621mg)溶于DMF(3mL)和THF(3mL)中,室温反应6h。反应完毕后,加入EA和水,EA萃取2遍,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱层析分离纯化得目标化合物M4(220mg)。
中间体M5的合成:
Figure PCTCN2022136197-appb-000029
在室温下,在反应瓶中将2-亚甲基-5-氧代-1,3,6,7-四氢吡咯利嗪-8-羧酸乙酯(10.00g)溶于THF(150.00mL),缓慢加入LAH(2.5M/THF)(3.63g),控制温度在60℃以下,加完搅拌0.2h。降温至0℃,加3.6ml水淬灭反应,再加15%的氢氧化钠水溶液3.6ml,最后加10.8ml水,搅拌10min后加入无水硫酸镁干燥,搅拌10min后过滤,滤饼用EA洗三次,母液浓缩即可得到目标中间体M5(6.5g,89.34%产率)。
中间体M6的合成:
Figure PCTCN2022136197-appb-000030
在室温下,将化合物(1S,5R)-2-(羟甲基)-3,8-二氮杂环[3.2.1]辛烷-8-羧酸叔丁酯(600mg)、TBSCl(448mg)和DIPEA(641mg)溶于DCM(15mL)中,室温反应过夜。反应完毕后,加入DCM和水,DCM萃取2遍,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物M6(740mg)。
实施例97:化合物2-氨基-4-((6S,9R)-3-氯-1-氟-13-((2R,7aS)-2-氟四氢-1H-吡咯啉-7a(5H)-基)甲氧基)-5a,6,7,8,9,10-六氢-5H-6,9-表氨基氮杂环[2’,1’:3,4][1,4]恶唑[5,6,7D]喹唑啉-2-基)-7-氟苯并[b]噻吩-3-腈的合成
Figure PCTCN2022136197-appb-000031
步骤1:化合物97-1的合成
将化合物(1R,5S)-2-(羟甲基)-3,8-二氮杂环[3.2.1]辛烷-8-羧酸叔丁酯(312mg)溶于5mL无水THF中,然后将NaH(141mg)加入,室温反应20min,然后将中间体M2慢慢加入,40℃反应30min。加入饱和氯化铵淬灭,加入EA和水,EA萃取反应液,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物97-1(330mg)。ESI-MS m/z:563[M+H] +
步骤2:化合物97-2的合成
将化合物97-1(330mg)溶于无水ACN(5mL)中,然后将DBU(445mg)和PyBOP(610mg)依次加入,室温反应1h。加入EA和水,EA萃取反应液,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物97-2(220mg)。ESI-MS m/z:545[M+H] +
步骤3:化合物97-3的合成
将化合物97-2(150mg)、中间体(3-氰基-4-(5,5-二甲基-1,3,2-二氧苄啉-2-基)-7-氟苯并噻吩-2-基特丁基)氨基甲酸酯(150mg)、Pd(DPEPhos)Cl 2(40mg)、K 3PO 4(175mg)和KF(64mg)溶于5mL的CPME中,氮气置换5次,105℃反应3h。用EA和水萃取反应液,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物97-3的粗产物230mg,直接用于下一步反应。ESI-MS m/z:757[M+H] +
步骤4:化合物97-4的合成
将化合物97-3(230mg)溶于DCM(5mL)中,然后将m-CPBA(210mg)加入,室温反应30min。加适量饱和亚硫酸钠溶液搅拌20min淬灭,加入DCM和水,DCM萃取反应液,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物97-4(138mg)。ESI-MS m/z:789[M+H] +
步骤5:化合物97-5的合成
将化合物((2R,7aS)-2-氟四氢-1H-吡咯利嗪-7a(5H)-基)甲醇(26mg)溶于2mL的无水THF中,然后将NaH(15mg)加入,室温反应20min,然后将化合物97-4(65mg)加入,室温反应30min。加适量饱和氯化铵溶液淬灭,加入EA和水,EA萃取反应液,有机相用无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物97-5(68mg)。ESI-MS m/z:868[M+H] +
步骤6:化合物97的合成
将化合物97-5(68mg)溶于2mL的DCM中,然后将TFA(1mL)加入,室温反应20min。减压浓缩,浓缩物经Pre-TLC(DCM:7M氨甲醇=8:1)分离纯化得到目标化合物97(8.7mg)。ESI-MS m/z:668[M+H] +1H NMR(500MHz,DMSO-d 6)δ8.10(s,2H),7.23(dd,J=8.4,5.2Hz,1H),7.14(t,J=8.9Hz,1H),4.82(dd,J=13.4,2.6Hz,1H),4.66(dd,J=13.2,3.1Hz,1H),4.31(dd,J=13.2,6.8Hz,1H),4.17(q,J=2.9Hz,1H),4.10(dd,J=14.7,10.3Hz,1H),4.00(t,J=10.7Hz,1H),3.88-3.72(m,2H),3.11(d,J=8.2Hz,2H),3.05(s,1H),2.84(td,J=9.0,5.8Hz,1H),2.19-2.11(m,1H),2.08(s,1H),2.03(dt,J=7.3,3.6Hz,1H),1.88-1.66(m,7H),1.24(d,J=7.2Hz,3H)。
实施例98:化合物5-乙炔基-6-氟-4-((6S,9R)-1-氟-12-((2R,7aS)-2-氟四氢-1H-吡咯啉-7a(5H)-基)甲氧基)-5a,6,7,8,9,10-六氢-5H-4-氧代-3,10a,11,13,14-戊糖-6,9-甲壬基[1,8-ab]庚烯-2-基)萘-2-醇的合成
Figure PCTCN2022136197-appb-000032
步骤1:化合物98-1的合成
在室温下,将化合物((2-氟-6-(甲氧基甲氧基)-8-(4,4,5,5-四甲基-1,3,2-二氧苯并呋喃-2-基)萘-1-基)乙炔基)三异丙基硅烷(10g)溶于HCl/Dioxane(4mol/L,25mL)中,室温搅拌反应30min。向反应混合物中加入饱和NaHCO 3溶液淬灭,在向其中加入EA萃取三次,合并有机相,浓缩即可得到目标化合物98-1(9.6g)。ESI-MS m/z:468[M+H] +
步骤2:化合物98-2的合成
将中间体M4(200mg)、PdCl 2(dppf)(70mg)、98-1(220mg)和K 2CO 3溶于1,4-二氧六环(4mL)和水(1mL)中,氮气保护下,130℃反应2h。待反应完毕后,加入EA和水,EA萃取2遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱纯化得目标化合物98-2(95mg)。ESI-MS m/z:774[M+H] +
步骤3:化合物98-3的合成
将化合物98-2(95mg)、TBSCl(74mg)和咪唑(84mg)溶于DCM(3mL)中,室温反应30min。加入DCM和水,DCM萃取2遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱分离纯化得目标化合物98-3(80mg)。ESI-MS m/z:888[M+H] +
步骤4:化合物98-4的合成
将化合物98-3(80mg)溶于DCM(2mL)中,然后将m-CPBA(68mg)加入,室温反应30min。加入饱和亚硫酸钠水溶液(2mL)并搅拌20min淬灭,然后DCM和水萃取2遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱分离纯化得到目标化合物98-4(58mg)。ESI-MS m/z:920[M+H] +
步骤5:化合物98-5的合成
将化合物((2R,7aS)-2-氟四氢-1H-吡咯利嗪-7a(5H)-基)甲醇(18mg)溶于无水 THF(2mL)中,然后将NaH(8mg)加入,室温反应20min。然后将化合物98-4(50mg)加入,室温反应30min,反应完毕后,加入饱和氯化铵淬灭,加入EA和水,EA萃取2遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱分离纯化得目标化合物98-5(49mg)。ESI-MS m/z:885[M+H] +
步骤6:化合物98-6的合成
将化合物98-5(49mg)溶于DMF(1mL)中,然后将CsF(169mg)加入,室温反应2h。反应完毕后,加入EA和水,EA萃取3遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱分离纯化得目标化合物98-6(14mg)。ESI-MS m/z:729[M+H] +
步骤7:化合物98的合成
将化合物98-6(14mg)溶于DCM(2mL)中,然后将BF 3·Et 2O(47mg)加入,室温反应30min。反应完毕后,将反应液加入到冷的饱和碳酸钠水溶液中,并用DCM:MeOH=10:1的混合溶剂萃取两遍,饱和食盐水萃取1遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱分离纯化得目标化合物98(5mg)。ESI-MS m/z:629[M+H] +1H NMR(500MHz,DMSO-d 6)δ10.14(d,J=2.2Hz,1H),7.96(ddd,J=8.9,5.9,2.6Hz,1H),7.46(td,J=9.0,5.2Hz,1H),7.37(t,J=2.2Hz,1H),7.18(d,J=2.6Hz,1H),5.42–5.20(m,2H),4.87(dd,J=25.8,13.2Hz,1H),4.56(dd,J=22.1,12.7Hz,1H),4.39(ddd,J=31.8,13.3,7.4Hz,1H),4.14–3.97(m,4H),3.11(s,3H),2.84(d,J=7.7Hz,1H),2.18–2.10(m,1H),2.07(s,1H),2.00(dt,J=9.5,6.6Hz,4H),1.79(q,J=13.9Hz,5H),1.50–1.44(m,1H),0.85(t,J=6.7Hz,2H)。
实施例99:化合物2-氨基-4-((6S,9R)-3-氯-13-((2-(二氟亚甲基)四氢-1H-吡咯利嗪-7a(5H)-基)甲氧基)-1-氟-5a,6,7,8,9,10-六氢-5H-6,9-表氨基氮杂环[2',1':3,4][1,4]恶嗪[5,6,7D]喹唑啉-2-基)-7-氟苯并[b]噻吩-3-腈的合成
Figure PCTCN2022136197-appb-000033
步骤1:化合物99-1的合成
将中间体M3(26mg)溶于2mL的无水THF中,然后将NaH(15mg)加入,室温反应20min,然后将化合物97-4(65mg)加入,室温反应30min。加适量饱和氯化铵溶液淬灭, 用EA和水萃取反应液,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经柱色谱分离纯化得目标化合物99-1(72mg)。ESI-MS m/z:898[M+H] +
步骤2:化合物99的合成
将化合物99-1(72mg)溶于2mL的DCM中,然后将TFA(1mL)加入,室温反应20min。减压浓缩,加少量DCM溶解,然后加入饱和碳酸氢钠溶液中和,DCM:MeOH=10:1(体积比)萃取两遍,再用饱和食盐水萃取1遍,保留有机相,无水硫酸钠干燥,过滤,减压浓缩,浓缩物经Pre-TLC(DCM:MeOH(7M氨)=8:1)分离纯化得目标化合物99(12.8mg)。ESI-MS m/z:698[M+H] +1H NMR(500MHz,DMSO-d 6)δ8.10(s,2H),7.21(ddd,J=8.0,5.2,2.5Hz,1H),7.14(t,J=8.9Hz,1H),4.85(dt,J=13.4,3.1Hz,1H),4.56(dd,J=13.2,3.3Hz,1H),4.43(dd,J=13.2,7.4Hz,1H),4.24-4.00(m,3H),3.78(s,1H),3.70-3.62(m,2H),3.20(s,1H),3.02-2.98(m,1H),2.66-2.61(m,1H),2.58-2.54(m,1H),2.44-2.38(m,1H),2.00-1.96(m,1H),1.86(dd,J=7.7,3.9Hz,1H),1.79-1.74(m,3H),1.72-1.59(m,2H),1.25-1.23(m,2H)。
实施例100:化合物2-氨基-4-((6R,9S)-3-氯-1-氟-13-((2R,7aS)-2-氟四氢-1H-吡咯利嗪-7a(5H)-基)甲氧基)-5a,6,7,8,9,10-六氢-5H-6,9-表氨基氮杂环[2',1':3,4][1,4]恶唑[5,6,7D]喹唑啉-2-基)-7-氟苯并[b]噻吩-3-碳腈的合成
Figure PCTCN2022136197-appb-000034
化合物100-1的合成参考实施例97的合成。
将100-1(71.00mg)溶于DCM(2.00mL)中,再加入TFA(2.00mL)。室温搅拌0.5小时。将反应液直接减压浓缩,经饱和NaHCO 3调至pH=8,再用体积比DCM/MeOH=10/1(15mL)萃取2次,有机层用饱和食盐水洗涤再用无水硫酸钠干燥,浓缩。浓缩物经Pre-TLC分离纯化DCM/MeOH(9%7M NH 3/MeOH)得化合物100(9.4mg,17.2%收率)。ESI-MS m/z:668[M+H] +1H NMR(500MHz,DMSO-d 6)δ8.08(s,2H),7.20-7.10(m,2H),5.39-5.23(m,2H),4.83(t,J=10.3Hz,1H),4.58(dd,J=10.7,2.5Hz,1H),4.48(d,J=12.8Hz,1H),4.03(d,J=58.4Hz,3H),3.82-3.71(m,2H),2.89(s,2H),2.24-1.94(m,5H),1.92-1.68(m,7H),1.62(d,J=10.1Hz,1H)。
下述的实施例采用上述方法合成,或使用相应中间体的类似方法合成。
Figure PCTCN2022136197-appb-000035
Figure PCTCN2022136197-appb-000036
Figure PCTCN2022136197-appb-000037
Figure PCTCN2022136197-appb-000038
Figure PCTCN2022136197-appb-000039
实施例102: 1H NMR(500MHz,MeOD)δ7.88-7.79(m,1H),7.36-7.26(m,2H),7.20(dd,J=33.3,2.5Hz,1H),5.07(d,J=10.6Hz,1H),4.62(t,J=11.5Hz,1H),4.52-4.43(m,1H),4.42-4.35(m,1H),4.34-4.25(m,1H),4.19(d,J=6.3Hz,1H),3.92-3.81(m,2H),3.76(s,1H),3.59-3.46(m,2H),3.22(dd,J=12.2,8.0Hz,1H),2.87-2.72(m,2H),2.55(d,J=15.9Hz,1H),2.22-2.10(m,1H),2.06-1.82(m,8H)。
生物学测试
药理实验1:细胞增殖实验(AGS)
将KRas-G12D突变型肿瘤细胞AGS(
Figure PCTCN2022136197-appb-000040
CRL-1739 TM)按1×10 3/孔的细胞密度铺于低吸附96孔板中,置于细胞培养箱隔夜培养。待细胞贴壁后,将待测化合物按照终浓度10000、 2000、400、80、16、3.2、0.64、0.128、0.025、0nM(DMSO终浓度均为0.5%)加入96孔板中,37℃培养96h后向各孔加入50μL Cell-titer GLO工作液,震荡混匀后室温孵育10min,在多功能酶标仪读取Luminescence发光值,将发光值数据计算转换为抑制百分数。并根据以下公式,计算细胞增殖抑制百分数:
抑制百分数=(最大值-所测值)/(最大值-Blank)×100
(“最大值”来自0.1%DMSO对照孔,“Blank”来自空白对照孔,“所测值”来自化合物处理孔)。
利用GraphPad Prism软件进行曲线拟合并获取IC 50值。
检测结果表明本发明的化合物具有良好活性。
表1
化合物名称 AGS IC 50(nM)
97 6.0
98 14.1
99 10.0
100 12.0
101 12.0
药理实验2:细胞p-ERK检测试验
将KRas-G12D突变型肿瘤细胞AGS(
Figure PCTCN2022136197-appb-000041
CRL-1739 TM)按4×10 4/孔的细胞密度铺于96孔板中,置于细胞培养箱隔夜培养。待细胞贴壁后,将待测化合物按照终浓度3000nM、600nM、120nM、24nM、4.8nM、0.96nM、0.19nM、0.1%DMSO加入96孔板中,培养3h后,利用pERK HTRF Kit(Cisbio)中的裂解液裂解96孔板中各处理细胞样品(40ul/孔),充分裂解完成后分别向HTRF检测96孔板中加入16ul/孔的蛋白液以及4ul预混好的pERK抗体。4℃过夜孵育后在多功能酶标仪读取665nM/620nM的ratio信号值,采集原始数据。并根据以下公式,计算p-ERK抑制百分数:
抑制百分数=(最大值-所测值)/(最大值-Blank)×100
(“最大值”来自0.1%DMSO对照孔,“Blank”来自空白对照孔,“所测值”来自化合物处理孔)。
利用GraphPad Prism软件进行曲线拟合并获取IC 50值。
表2
Figure PCTCN2022136197-appb-000042

Claims (26)

  1. 一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐:
    Figure PCTCN2022136197-appb-100001
    其中,
    X 1选自CR 4或N;R 4选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
    X 2选自CR 5或N;R 5选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
    X 3选自CR 6或N;R 6选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
    X 4选自键、O、NR 7、C(R 7) 2或C(O);R 7各自独立地选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
    X 5选自键、O、NR 8、C(R 8) 2或C(O);R 8各自独立地选自H、氨基、取代的氨基、氰基、C 1-6烷基、取代的C 1-6烷基、卤素、C 2-6烯基、取代的C 2-6烯基、C 3-6环烷基或取代的C 3-6环烷基;
    L选自键、-O-C 0-4亚烷基-、-S-C 0-4亚烷基-、-NR 9-C 0-4亚烷基-或C 2-4亚炔基,所述-O-C 0- 4亚烷基-、-S-C 0-4亚烷基-、-NR 9-C 0-4亚烷基-或C 2-4亚炔基任选地被一个或多个R 9取代;R 9选自H、卤素、氰基、羟基或C 1-6烷基,或者同一个碳原子上的两个R 9连同其连接的原子共同形成C 3-6环烷基或3-6元杂环基;
    R 1选自H、羟基、氰基、卤素或C 1-3烷基,所述C 1-3烷基任选地被一个多个选自卤素、羟基或氰基的取代基所取代;
    R 2选自-OR a、-OC(O)N(R a) 2、-N(R a) 2、-NR aC(O)R a、-NR aC(O)N(R a) 2、-NR aS(O)R a、-NR aS(O) 2R a、-S(=O)R a、-S(=O) 2R a、-SR a、-S(R a) 5、-C(=O)R a、-C(=O)OR a、-C(=O)N(R a) 2、环烷基、杂环基、芳基或杂芳基,所述环烷基、杂环基、芳基或杂芳基任选地进一步被一个 或多个R 10取代;R 10选自H、氰基、
    Figure PCTCN2022136197-appb-100002
    卤素、C 1-6烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-OC(O)N(R a) 2、-C 0-6亚烷基-N(R a) 2、-C 0-6亚烷基-NR aC(O)R a、-C 0-6亚烷基-NR aC(O)N(R a) 2、-C 0-6亚烷基-NR aS(O)R a、-C 0-6亚烷基-NR aS(O) 2R a、-C 0-6亚烷基-S(=O)R a、-C 0-6亚烷基-S(=O) 2R a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-S(R a) 5、-C 0-6亚烷基-C(=O)R a、-C 0-6亚烷基-C(=O)OR a、-C 0-6亚烷基-C(=O)N(R a) 2、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0- 6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基),所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基)任选地还可被1个或多个R a所取代;
    R 3选自C 6-14芳基或5-14元杂芳基,所述芳基或杂芳基任选地进一步被一个或多个R 11取代;R 11选自H、氰基、卤素、C 1-6烷基、C 1-6卤代烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-OC(O)N(R a) 2、-C 0-6亚烷基-N(R a) 2、-C 0-6亚烷基-NR aC(O)R a、-C 0-6亚烷基-NR aC(O)N(R a) 2、-C 0-6亚烷基-NR aS(O)R a、-C 0-6亚烷基-NR aS(O) 2R a、-C 0-6亚烷基-S(=O)R a、-C 0-6亚烷基-S(=O) 2R a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-S(R a) 5、-C 0-6亚烷基-C(=O)R a、-C 0-6亚烷基-C(=O)OR a、-C 0-6亚烷基-C(=O)N(R a) 2、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基),所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基)任选地还可被1个或多个R a所取代;
    R a各自独立地选自H、卤素、羟基、氨基、氧代基、硝基、氰基、羧基、C 1-6烷基、C 1-6羟基烷基、C 1-6氨基烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6杂烷基、C 3-8环烷基、3-8元杂环基、C 6-14芳基或5-14元杂芳基;
    m选自0、1、2、3或4。
  2. 根据权利要求1所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,R 10选自H、氰基、卤素、C 1-6烷基、-C 0-6亚烷基-OR a、-C 0-6亚烷基-OC(O)N(R a) 2、-C 0-6亚烷基-N(R a) 2、-C 0-6亚烷基-NR aC(O)R a、-C 0-6亚烷基-NR aC(O)N(R a) 2、-C 0-6亚烷基-NR aS(O)R a、-C 0-6亚烷基-NR aS(O) 2R a、-C 0-6亚烷基-S(=O)R a、-C 0-6亚烷基-S(=O) 2R a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-S(R a) 5、-C 0-6亚烷基-C(=O)R a、-C 0-6亚烷基-C(=O)OR a、-C 0-6亚烷基-C(=O)N(R a) 2、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基),所述C 1- 6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基、-C 0-6亚烷基-(3-14元杂环基)、-C 0-6亚烷基-C 6-14芳基或-C 0-6亚烷基-(5-14元杂芳基)任选地还可被1个或多个R a所取代;
    R a各自独立地选自H、卤素、羟基、氨基、氧代基、硝基、氰基、羧基、C 1-6烷基、C 1-6羟基烷基、C 1-6氨基烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6杂烷基、C 3-8环烷基、3-8元杂环基、C 6-14芳基或5-14元杂芳基。
  3. 一种通式(I)所示的化合物、其互变异构体、氘代物或药用盐,其选自式(IA)-式(ID)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐:
    Figure PCTCN2022136197-appb-100003
    其中,R 2,R 3,R 4,R 5,R 6,L如权利要求1或2中所示。
  4. 根据权利要求1-3中任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述L为-O-C 0-4亚烷基-,所述R 2为3-14元杂环基,所述3-14元杂环基任选地进一步被一个或多个R 10取代,所述R 10选自H、氰基、卤素、C 1-6烷基、C 2-6烯基、C 2-6炔基,所述C 1-6烷基、C 2-6烯基、C 2-6炔基任选地还可被1个或多个R a所取代;所述R a各自独立地选自H、卤素、羟基、氨基、氧代基、硝基、氰基或羧基。
  5. 根据权利要求4所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 2为3-14元稠杂环基,所述3-14元稠杂环基任选地进一步被一个或多个R 10取代,所述R 10选自卤素或C 2-6烯基,所述烯基被任选地还可被1个或多个R a所取代;所述R a各自独立地选自H或卤素。
  6. 根据权利要求5所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述稠杂环基选自
    Figure PCTCN2022136197-appb-100004
    Figure PCTCN2022136197-appb-100005
    所述稠杂环基任选地进一步被一个或多个R 10取 代,所述R 10选自H、卤素或C 2-6烯基,所述烯基被任选地还可被1个或多个R a所取代;所述R a各自独立地选自H或卤素。
  7. 根据权利要求1-6任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 10
    Figure PCTCN2022136197-appb-100006
    所述R a各自独立地为卤素,优选为F。
  8. 根据权利要求1-6任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 10
    Figure PCTCN2022136197-appb-100007
    所述R a为H。
  9. 根据权利要求6所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 10为卤素,优选为F。
  10. 根据权利要求1-9任一项所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述
    Figure PCTCN2022136197-appb-100008
    选自
    Figure PCTCN2022136197-appb-100009
  11. 根据权利要求1-3中任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于所述L选自键、-O-C 0-4亚烷基-或-NR 9-C 0-4亚烷基,所述R 2选自-N(R a) 2、-C(=O)R a、-C(=O)N(R a) 2、C 3-12环烷基,所述C 3-12环烷基、任选地进一步被一个或多个R 10取代,所述R 10选自H、氰基、卤素、C 1-6烷基、-C 0-6亚烷基-OR a、-C 0- 6亚烷基-(3-14元杂环基),所述C 1-6烷基、-C 0-6亚烷基-(3-14元杂环基)任选地还可被1个或多个R a所取代,所述R a选自H、氰基、卤素或C 1-6烷基。
  12. 根据权利要求11所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述
    Figure PCTCN2022136197-appb-100010
    选自
    Figure PCTCN2022136197-appb-100011
    Figure PCTCN2022136197-appb-100012
  13. 根据权利要求1-12中任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于所述R 4各自独立选自H、氰基或卤素,所述卤素优选为F。
  14. 根据权利要求1-13中任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于所述R 5各自独立选自H、C 1-6烷基或卤素,所述卤素优选为F。
  15. 根据权利要求1-14中任一项所述一种通式(I)所示的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于所述R 6各自独立选自H、C 1-6烷基、C 2-6烯基、C 3-6环烷基或卤素,所述卤素优选为F。
  16. 根据权利要求1所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述X 4选自键、NR 7、C(R 7) 2或C(O),所述R 7各自独立地选自H或甲基。
  17. 根据权利要求1或16所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述X 5选自键或C(O)。
  18. 根据权利要求1、3-17任一项所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 1选自H。
  19. 根据权利要求1-18任一项所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 3选自
    Figure PCTCN2022136197-appb-100013
    所述
    Figure PCTCN2022136197-appb-100014
    任选地进一步被一个或多个R 11取代,所述R 11选自H、氰基、卤素、-C 0-6亚烷基-OR a、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基或-C 0-6亚烷基-C 3-14环烷基,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基任选地进一步被1个或多个R a所取代;所述R a各自独立地选自H或卤素。
  20. 根据权利要求1-10,13-15,18-19中任一项所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述式(I)选自(IA-1)、(IA-2)、(IB-1)、(IB-2)、(IC-1)、(IC-2)、(ID-1)或(ID-2):
    Figure PCTCN2022136197-appb-100015
    Figure PCTCN2022136197-appb-100016
    其中,所述R a各自独立地选自H或卤素,所述卤素优选为F;
    所述R 4各自独立选自H、氰基或卤素,所述卤素优选为F;
    所述R 5各自独立选自H、C 1-6烷基或卤素,所述卤素优选为F;
    所述R 6各自独立选自H、C 1-6烷基、C 2-6烯基、C 3-6环烷基或卤素,所述卤素优选为F;
    所述R 3选自C 6-14芳基或5-14元杂芳基,所述C 6-14芳基或C 6-14杂芳基任选地进一步被一个或多个R 11取代;所述C 6-14芳基或5-14元杂芳基优选自
    Figure PCTCN2022136197-appb-100017
    Figure PCTCN2022136197-appb-100018
    所述R 11选自H、氰基、卤素、-C 0-6亚烷基-OR a、C 1-6烷基、C 1- 6卤代烷基、C 2-6烯基、C 2-6炔基或-C 0-6亚烷基-C 3-14环烷基,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-14环烷基任选地进一步被1个或多个R a所取代;所述R a各自独立地选自H或卤素;
    R 10选自H、卤素或C 1-6烷基。
  21. 根据权利要求19或20所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,其特征在于,所述R 3选自
    Figure PCTCN2022136197-appb-100019
    Figure PCTCN2022136197-appb-100020
  22. 根据权利要求1-21任一项所述的化合物、其立体异构体、互变异构体、氘代物或药用盐,通式(I)所示的化合物选自下式化合物:
    Figure PCTCN2022136197-appb-100021
    Figure PCTCN2022136197-appb-100022
  23. 一种药物组合物,其特征在于,所述药物组合物含有治疗有效量的权利要求1-22中任一项所述的化合物或其立体异构体、互变异构体、氘代物或药用盐。
  24. 权利要求1-22中任一项所述的化合物或权利要求23所述的药物组合物在制备药物中的应用。
  25. 根据权利要求24所述的应用,其特征在于,所述制备药物为在制备治疗和/或预防由KRAS G12D介导的疾病的药物中的应用。
  26. 一种治疗和/或预防疾病的方法,包括向治疗对象施用治疗有效量的权利要求1-21中任一项所述的化合物或权利要求22所述的药物组合物。
PCT/CN2022/136197 2021-12-07 2022-12-02 Kras g12d抑制剂及其在医药上的应用 WO2023103906A1 (zh)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202111485412 2021-12-07
CN202111485412.5 2021-12-07
CN202210016082.3 2022-01-07
CN202210016082 2022-01-07
CN202210997326.0 2022-08-19
CN202210997326 2022-08-19

Publications (1)

Publication Number Publication Date
WO2023103906A1 true WO2023103906A1 (zh) 2023-06-15

Family

ID=86729634

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/136197 WO2023103906A1 (zh) 2021-12-07 2022-12-02 Kras g12d抑制剂及其在医药上的应用

Country Status (1)

Country Link
WO (1) WO2023103906A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024022444A1 (zh) * 2022-07-27 2024-02-01 江苏恒瑞医药股份有限公司 稠环类化合物、其制备方法及其在医药上的应用
WO2024032704A1 (en) * 2022-08-11 2024-02-15 Beigene, Ltd. Heterocyclic compounds, compositions thereof, and methods of treatment therewith
US11912723B2 (en) 2022-02-09 2024-02-27 Quanta Therapeutics, Inc. KRAS modulators and uses thereof
WO2024041573A1 (en) * 2022-08-25 2024-02-29 Zai Lab (Shanghai) Co., Ltd. Fused multi-heterocyclic compounds as kras g12d modulators and uses thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018206539A1 (en) * 2017-05-11 2018-11-15 Astrazeneca Ab Heteroaryl compounds that inhibit g12c mutant ras proteins
WO2019215203A1 (en) * 2018-05-08 2019-11-14 Astrazeneca Ab Tetracyclic heteroaryl compounds
WO2021023154A1 (zh) * 2019-08-02 2021-02-11 上海济煜医药科技有限公司 四并环类化合物及其制备方法和应用
WO2021041671A1 (en) * 2019-08-29 2021-03-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
WO2021108683A1 (en) * 2019-11-27 2021-06-03 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022173678A1 (en) * 2021-02-09 2022-08-18 Genentech, Inc. Tetracyclic oxazepine compounds and uses thereof
WO2022188729A1 (en) * 2021-03-07 2022-09-15 Jacobio Pharmaceuticals Co., Ltd. Fused ring derivatives useful as kras g12d inhibitors
WO2022194245A1 (zh) * 2021-03-17 2022-09-22 劲方医药科技(上海)有限公司 嘧啶并环类化合物及其制法和用途
WO2022199587A1 (zh) * 2021-03-24 2022-09-29 南京明德新药研发有限公司 嘧啶并杂环类化合物及其应用
WO2022206723A1 (zh) * 2021-03-30 2022-10-06 浙江海正药业股份有限公司 杂环类衍生物、其制备方法及其医药上的用途

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018206539A1 (en) * 2017-05-11 2018-11-15 Astrazeneca Ab Heteroaryl compounds that inhibit g12c mutant ras proteins
WO2019215203A1 (en) * 2018-05-08 2019-11-14 Astrazeneca Ab Tetracyclic heteroaryl compounds
WO2021023154A1 (zh) * 2019-08-02 2021-02-11 上海济煜医药科技有限公司 四并环类化合物及其制备方法和应用
WO2021041671A1 (en) * 2019-08-29 2021-03-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
WO2021108683A1 (en) * 2019-11-27 2021-06-03 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022173678A1 (en) * 2021-02-09 2022-08-18 Genentech, Inc. Tetracyclic oxazepine compounds and uses thereof
WO2022188729A1 (en) * 2021-03-07 2022-09-15 Jacobio Pharmaceuticals Co., Ltd. Fused ring derivatives useful as kras g12d inhibitors
WO2022194245A1 (zh) * 2021-03-17 2022-09-22 劲方医药科技(上海)有限公司 嘧啶并环类化合物及其制法和用途
WO2022199587A1 (zh) * 2021-03-24 2022-09-29 南京明德新药研发有限公司 嘧啶并杂环类化合物及其应用
WO2022206723A1 (zh) * 2021-03-30 2022-10-06 浙江海正药业股份有限公司 杂环类衍生物、其制备方法及其医药上的用途

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11912723B2 (en) 2022-02-09 2024-02-27 Quanta Therapeutics, Inc. KRAS modulators and uses thereof
WO2024022444A1 (zh) * 2022-07-27 2024-02-01 江苏恒瑞医药股份有限公司 稠环类化合物、其制备方法及其在医药上的应用
WO2024032704A1 (en) * 2022-08-11 2024-02-15 Beigene, Ltd. Heterocyclic compounds, compositions thereof, and methods of treatment therewith
WO2024041573A1 (en) * 2022-08-25 2024-02-29 Zai Lab (Shanghai) Co., Ltd. Fused multi-heterocyclic compounds as kras g12d modulators and uses thereof

Similar Documents

Publication Publication Date Title
WO2023103906A1 (zh) Kras g12d抑制剂及其在医药上的应用
CN114761408B (zh) Kras g12c抑制剂及其在医药上的应用
JP6877407B2 (ja) Ntrk関連障害の治療に有用な化合物および組成物
AU2014256635B2 (en) Deuterated diaminopyrimidine compounds and pharmaceutical compositions comprising such compounds
CN113748114B (zh) 一种喹唑啉化合物及其在医药上的应用
WO2023072188A1 (zh) Kras g12d抑制剂及其在医药上的应用
CN111499634B (zh) 一种喹唑啉化合物及其在医药上的应用
ES2739150T3 (es) Inhibidores de pirazolopirimidinilo de la enzima activadora de ubiquitina
CN112851663B (zh) 一种并杂环化合物及其用途
CN112745335A (zh) 一种三并杂环化合物及其用途
CN110950876B (zh) 一类呋喃并内酰胺类化合物、制备方法和用途
CN114685460A (zh) Kras g12c抑制剂及其在医药上的应用
JP2021501215A (ja) アミノ置換窒素含有縮合環化合物、その調製方法及び使用
CN112707905A (zh) 一种三并杂环化合物及其制备方法和用途
WO2021104488A1 (zh) 作为jak抑制剂的三并杂环类化合物及其应用
CN116964058A (zh) Kras g12d抑制剂及其在医药上的应用
AU2021341258A1 (en) CD73 inhibitor and application thereof in medicine
TWI638825B (zh) 抑制癌症及病毒之化合物
WO2024082872A1 (zh) 一种pak4激酶抑制剂及其制备方法和用途
CN108456214B (zh) 含噁唑或咪唑结构的喹唑啉类化合物及其应用
WO2023104043A1 (zh) 一种抗凋亡蛋白bcl-2抑制剂、药物组合物及其应用
AU2021309147B2 (en) Codrug that disintegrates in intestine, preparation therefor, and use thereof
CN115368382A (zh) Kras g12d抑制剂及其在医药上的应用
CN114380827A (zh) Kras g12c抑制剂及其在医药上的应用
CN114599656A (zh) 咪唑烷酮类化合物及其制备方法与应用

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: 22903328

Country of ref document: EP

Kind code of ref document: A1