WO2020125391A1 - 用于抑制蛋白激酶活性的氨基嘧啶类化合物 - Google Patents

用于抑制蛋白激酶活性的氨基嘧啶类化合物 Download PDF

Info

Publication number
WO2020125391A1
WO2020125391A1 PCT/CN2019/122399 CN2019122399W WO2020125391A1 WO 2020125391 A1 WO2020125391 A1 WO 2020125391A1 CN 2019122399 W CN2019122399 W CN 2019122399W WO 2020125391 A1 WO2020125391 A1 WO 2020125391A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
haloalkyl
cycloalkyl
groups
Prior art date
Application number
PCT/CN2019/122399
Other languages
English (en)
French (fr)
Inventor
王义汉
李焕银
Original Assignee
深圳市塔吉瑞生物医药有限公司
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 深圳市塔吉瑞生物医药有限公司 filed Critical 深圳市塔吉瑞生物医药有限公司
Priority to EP19899282.8A priority Critical patent/EP3885346A4/en
Priority to US17/415,492 priority patent/US20220048891A1/en
Priority to JP2021536036A priority patent/JP7240032B2/ja
Publication of WO2020125391A1 publication Critical patent/WO2020125391A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention belongs to the technical field of medicine, and particularly relates to aminopyrimidine compounds having protein tyrosine kinase inhibitory effects, pharmaceutical compositions containing them, and their preparation methods and uses.
  • disorders of protein kinases have been implicated in many diseases and disorders, such as central nervous system diseases (eg Alzheimer's disease), inflammatory and autoimmune diseases (eg asthma, rheumatoid arthritis, Crohn's disease, inflammatory Bowel syndrome and psoriasis), bone diseases (e.g. osteoporosis), metabolic disorders (e.g. diabetes), vascular proliferative diseases, eye diseases, cardiovascular diseases, cancer, restenosis, pain, transplant rejection and Infectious diseases.
  • central nervous system diseases eg Alzheimer's disease
  • inflammatory and autoimmune diseases eg asthma, rheumatoid arthritis, Crohn's disease, inflammatory Bowel syndrome and psoriasis
  • bone diseases e.g. osteoporosis
  • metabolic disorders e.g. diabetes
  • vascular proliferative diseases vascular proliferative diseases
  • eye diseases e.g. asthma, rheumatoid arthritis, Crohn's disease, inflammatory Bowel syndrome and p
  • EGFR is a member of the transmembrane protein tyrosine kinase of the erbB receptor family.
  • growth factor ligands such as epidermal growth factor (EGF)
  • the receptor can dimerize with EGFR or another family member such as erbB2 (HER2), erbB3 (HER3) and erbB4 (HER4).
  • HER2 epidermal growth factor
  • HER3 erbB3
  • HER4 erbB4
  • Dimerization of the erbB receptor leads to phosphorylation of key tyrosine residues in the intracellular domain, and in turn stimulates many intracellular signal transduction pathways involved in cell proliferation and survival.
  • Dysregulation of erbB family signal transduction promotes proliferation, invasion, metastasis, angiogenesis and tumor survival, and has been described in many human cancers such as lung cancer and breast cancer.
  • EGFR is an ideal target for the development of anticancer drugs, and a variety of compounds currently targeting EGFR are clinically available, including the first-generation inhibitors gefitinib and erlotinib.
  • the most common EGFR kinase active mutations L858R and del19 are sensitive to the treatment of gefitinib or erlotinib, but ultimately the gefitinib or erlot resulting from the mutation of the main gatekeeper residue T790M Tinib treatment resistance, which was detected in approximately half of clinically resistant patients, resulted in double mutants L858R/T790M and del19/T790M.
  • R 2 is selected from H, D, 4-7 membered heterocycloalkyl or -NR 7 R 8 , wherein the 4-7 membered heterocycloalkyl is optionally substituted with 1-10 R 9 groups;
  • R 3 is selected from H, D, halogen, -CN, -NO 2 , C 1-6 alkyl, C 1-6 haloalkyl or C 3-6 cycloalkyl, wherein the C 1-6 alkyl, C 1-6 haloalkyl or C 3-6 cycloalkyl is optionally substituted with 1-13 R 9 groups;
  • R 4 is selected from H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl or 5-10 membered Heteroaryl, wherein said C 6-10 aryl and 5-10 membered heteroaryl are optionally substituted with 1-13 R 9 groups;
  • R 5 is selected from H, D, -(CH 2 ) n OR 7 , -(CH 2 ) n NR 7 R 8 , -(CD 2 ) n OR 7 or -(CD 2 ) n NR 7 R 8 , where n From 1, 2, 3 or 4;
  • R 6 is selected from H, D or C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-13 R 9 groups;
  • R 9 is independently selected from H, D, halogen, -OH, C 1-6 alkoxy, -NH 2 , -NH(C 1-6 alkyl), -N(C 1-6 alkyl) 2 , -C(O)C 1-6 alkyl, -C(O)OC 1-6 alkyl, -C(O)NHC 1-6 alkyl, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl or 5-10 membered heteroaryl; or two R 9 groups on the same atom or adjacent atoms may be formed together C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl or 5-10 membered heteroaryl; wherein each group in the definition of R 9 is optionally substituted by one or more D substituted until completely deuterated;
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention or a pharmaceutically acceptable salt, stereoisomer, solvate or hydrate thereof, and a pharmaceutically acceptable excipient .
  • the compound of the present invention is provided in the pharmaceutical composition in an effective amount.
  • the compound of the invention is provided in a therapeutically effective amount.
  • the compounds of the present invention are provided in a prophylactically effective amount.
  • the invention provides a method of treating a protein kinase-mediated disease in a subject, comprising administering to the subject a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer thereof , Solvate or hydrate, or the pharmaceutical composition of the present invention.
  • the invention provides a compound of the invention or a pharmaceutically acceptable salt, stereoisomer, solvate or hydrate thereof, or a pharmaceutical composition of the invention for use in the treatment of diseases mediated by protein kinases .
  • the disease is mediated by at least one mutant EGFR kinase.
  • the at least one mutant EFGR is del19, L858R, or T790M.
  • the at least one mutant EGFR is at least one double mutant selected from del19/T790M or L858R/T790M.
  • the disease is mediated by wild-type and/or mutant JAK3 kinase.
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1 -2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 and C 5 -6 alkyl.
  • C 1-6 alkyl refers to a linear or branched saturated hydrocarbon group having 1-6 carbon atoms, and is also referred to herein as "lower alkyl". In some embodiments, C 1-4 alkyl is particularly preferred. Examples of the alkyl group include but are not limited to: methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), tertiary Butyl (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl (C 5 ), 3-methyl-2-butyl (C 5 ), tert-amyl (C 5 ) and n-hexyl (C 6 ).
  • each of the alkyl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, and the appropriate substituents are as follows definition.
  • C 2-6 alkenyl refers to a straight or branched chain hydrocarbon group having 2-6 carbon atoms and one or more carbon-carbon double bonds (eg, 1, 2, or 3 carbon-carbon double bonds) .
  • the one or more carbon-carbon double bonds can be internal (eg, in 2-butenyl) or terminal (eg, in 1-butenyl).
  • C 2-4 alkenyl is particularly preferred.
  • C 2-6 alkynyl means having 2-6 carbon atoms, one or more carbon-carbon triple bonds (for example, 1, 2, or 3 carbon-carbon triple bonds) and optionally one or more carbons -A linear or branched hydrocarbon group of carbon double bonds (for example, 1, 2, or 3 carbon-carbon double bonds).
  • C 2-4 alkynyl is particularly preferred.
  • the alkynyl group does not contain any double bonds.
  • the one or more carbon triple bonds can be internal (eg, in 2-butynyl) or terminal (eg, in 1-butynyl).
  • C 1-6 alkylene group refers to a divalent group formed by removing another hydrogen of the C 1-6 alkyl group, and may be a substituted or unsubstituted alkylene group. In some embodiments, C 1-4 alkylene is particularly preferred.
  • the unsubstituted alkylene groups include, but are not limited to: methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), butylene Group (-CH 2 CH 2 CH 2 CH 2 -), pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) ,and many more.
  • C 1-6 alkoxy refers to the group -OR, where R is a substituted or unsubstituted C 1-6 alkyl. In some embodiments, C 1-4 alkoxy is particularly preferred.
  • the specific alkoxy groups include but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, t-butoxy, sec-butoxy, n-pentoxy, N-hexyloxy and 1,2-dimethylbutoxy.
  • Halo or halogen refers to fluorine (F), chlorine (Cl), bromine (Br), and iodine (I).
  • the halogen group is F, Cl, or Br.
  • the halogen group is F or Cl.
  • the halogen group is F.
  • a 3-7 membered heterocycloalkyl is preferred, which is a 3-7 membered non-aromatic ring system having ring carbon atoms and 1-3 ring heteroatoms; in some embodiments, 4- 7-membered heterocycloalkyl groups are preferred, which are 4-7 membered non-aromatic ring systems with ring carbon atoms and 1-3 ring heteroatoms; in some embodiments, 3-6 membered heterocycloalkyl groups are particularly Preferably, it is a 3-6 membered non-aromatic ring system having ring carbon atoms and 1-3 ring heteroatoms; more preferably a 5-6 membered heterocycloalkyl group, which has ring carbon atoms and 1-3 rings Heteroatom 5-6 membered non-aromatic ring system.
  • Exemplary 3-membered heterocycloalkyl groups containing one heteroatom include, but are not limited to: aziridine, oxepanyl, thiorenyl.
  • Exemplary 4-membered heterocycloalkyl groups containing one heteroatom include, but are not limited to: azetidine, oxetanyl, and thietane.
  • Exemplary 5-membered heterocycloalkyl groups containing one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2 , 5-dione.
  • Exemplary 8-membered heterocycloalkyl groups containing one heteroatom include, but are not limited to: azetyl octyl, oxetanyl, and thiohexyl.
  • Exemplary 5-membered heterocyclic alkyl groups fused to a C 6 aryl ring include, but are not limited to: indoline, isoindoline Indolyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinone, and so on.
  • C 6-14 aryl refers to a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n+2 aromatic ring system having 6-14 ring carbon atoms and zero heteroatoms (e.g., having Groups of 6, 10 or 14 ⁇ electrons shared in a ring arrangement.
  • the aryl group has six ring carbon atoms ("C 6 aryl”; for example, phenyl).
  • the aryl group has ten ring carbon atoms (“C 10 aryl”; for example, naphthyl, for example, 1-naphthyl and 2-naphthyl).
  • the aryl group has fourteen ring carbon atoms (“C 14 aryl”; for example, anthracenyl). In some embodiments, C 6-10 aryl is particularly preferred, and C 6 aryl is more preferred.
  • the aryl group also includes a ring system in which the above aryl ring is condensed with one or more cycloalkyl or heterocycloalkyl groups, and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues Represents the number of carbon atoms in the aryl ring system.
  • each of the aryl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, and suitable substituents are as follows definition.
  • 5-10 membered heteroaryl refers to a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms (eg, having a shared ring arrangement 6 or 10 ⁇ electrons) wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur.
  • the point of attachment may be a carbon or nitrogen atom as long as the valency permits.
  • Heteroaryl bicyclic ring systems can include one or more heteroatoms in one or two rings.
  • Heteroaryl also includes ring systems in which the above heteroaryl ring is fused with one or more cycloalkyl or heterocycloalkyl groups, and the point of attachment is on the heteroaryl ring, in this case, the carbon atom The number of continues to indicate the number of carbon atoms in the heteroaryl ring system.
  • a 5-6 membered heteroaryl group is particularly preferred, which is a 5-6 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms.
  • each of the heteroaryl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, appropriate substitution
  • the basis is defined as follows.
  • Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furyl and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl, and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyridyl.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl.
  • Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azacyclotrienyl, oxacyclotrienyl, and thiocyclotrienyl.
  • Exemplary 5,6-bicyclic heteroaryl groups include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl , Benzisofuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, Indenazinyl and purinyl.
  • Each of R cc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocycloalkyl, aryl, and heteroaryl, or two R cc groups are combined to form a hetero Cycloalkyl or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocycloalkyl, aryl, and heteroaryl is independently substituted by 0, 1, 2, 3, 4 or 5 R dd group substitution;
  • Each R ee is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocycloalkyl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, Carbocyclyl, heterocycloalkyl, aryl and heteroaryl are independently substituted with 0, 1, 2, 3, 4 or 5 R gg groups;
  • Each R ff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocycloalkyl, aryl, and heteroaryl, or two R ff groups combine to form a heterocycle Alkyl or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocycloalkyl, aryl, and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R gg group substitution;
  • pharmaceutically acceptable salts refers to, within the scope of reliable medical judgment, suitable for contact with tissues of humans and lower animals without excessive toxicity, irritation, allergies, etc., and with reasonable benefits/hazards The proportion of those salts.
  • Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts described in detail by Berge et al. in J. Pharmaceutical Sciences (1977) 66: 1-19.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and inorganic and organic bases.
  • non-toxic acid addition salts examples include salts with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid, or salts with organic acids, such as acetic acid, oxalic acid, Maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Also included are salts formed using conventional methods in the art, for example, ion exchange methods.
  • salts include: adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphor Salt, camphor sulfonate, citrate, cyclopentapropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, gluconate, glycerin Phosphate, gluconate, hemisulfate, enanthate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate , Malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate Salt, peroxine sodium
  • Pharmaceutically acceptable salts derived from suitable bases include alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • Representative alkali metal or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like.
  • other pharmaceutically acceptable salts include non-toxic ammonium salts, quaternary ammonium salts and amine cations formed with counter ions, such as halide, hydroxide, carboxylate, sulfate, phosphate, Nitrate, lower alkyl sulfonate and aryl sulfonate.
  • Subjects administered include, but are not limited to: humans (ie, male or female of any age group, for example, pediatric subjects (eg, infants, children, adolescents) or adult subjects (eg, young Adults, middle-aged adults or older adults)) and/or non-human animals, for example, mammals, for example, primates (for example, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep , Goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "human", “patient” and “subject” are used interchangeably herein.
  • Combination and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention.
  • the compound of the present invention can be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously administered with another therapeutic agent in a single unit dosage form.
  • R 2 is selected from H, D, 4-7 membered heterocycloalkyl or -NR 7 R 8 , wherein the 4-7 membered heterocycloalkyl is optionally substituted with 1-10 R 9 groups;
  • R 6 is selected from H, D or C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-13 R 9 groups;
  • R 7 and R 8 are each independently selected from H, D, C 1-6 alkyl, C 3-7 cycloalkyl or 4-7 membered heterocycloalkyl, or R 7 , R 8 and the N to which they are attached The atoms together form a 4-7 membered heterocycloalkyl; wherein the C 1-6 alkyl, C 3-7 cycloalkyl, or 4-7 membered heterocycloalkyl is optionally substituted by 1-13 R 9 groups Group replacement
  • R 9 is independently selected from H, D, halogen, -OH, C 1-6 alkoxy, -NH 2 , -NH(C 1-6 alkyl), -N(C 1-6 alkyl) 2 , -C(O)C 1-6 alkyl, -C(O)OC 1-6 alkyl, -C(O)NHC 1-6 alkyl, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl or 5-10 membered heteroaryl; or two R 9 groups on the same atom or adjacent atoms may be formed together C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl or 5-10 membered heteroaryl; wherein each group in the definition of R 9 is optionally substituted by one or more D substituted until completely deuterated;
  • R 1 is selected from C 1-6 haloalkyl or C 1-6 haloalkoxy
  • X is CH; in another specific embodiment, X is CD; in another specific embodiment, X is N.
  • R 1 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy or- OC 3-7 cycloalkyl, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and -OC 3-7 cycloalkyl is optionally substituted with 1-13 (e.g.
  • R 9 groups In a specific embodiment, R 1 is selected from -OR, wherein R is selected from H, C 1-6 alkyl, C 1-6 haloalkyl or C 3-7 cycloalkyl, wherein the C 1- 6 alkyl, C 1-6 haloalkyl and C 3-7 cycloalkyl are optionally 1-13 (e.g.
  • R 1 is C 1-6 haloalkoxy, wherein said C 1-6 haloalkoxy is optionally substituted by 1-12 (eg 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) R 9 group substitution; in a specific embodiment, R 1 is selected from -OCH 3 , -OCH 2 CH 3 , -OCH(CH 3 ), -OCHF 2 , -OCF 3 , -OCH 2 CF 3 , -OCD 3 , -OCD 2 CH 3 , -OCD (CD 3 ), -OCDF 2 , -OCF 3 or -OCD 2 CF 3 ; In a specific embodiment, R 1 is selected from —OCHF 2 , —OCF 3 or —OCH 2 CF 3 .
  • R 3 is selected from H, D, halogen, -CN, -NO 2 , C 1-6 alkyl or C 1-6 haloalkyl, wherein the C 1-6 alkyl and C 1-6 haloalkyl is optionally substituted with 1-13 (eg 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13) R 9 groups; in one
  • R 3 is selected from H, F, Cl, —CH 3 or CD 3 .
  • R 4 is selected from C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl or 5-10 membered heteroaryl, wherein said C 6- 10 aryl and 5-10 membered heteroaryl are optionally substituted with 1-8 (eg 1, 2, 3, 4, 5, 6, 7 or 8) R 9 groups; in a specific embodiment , R 4 is selected from C 6-10 aryl or 5-10 membered heteroaryl, wherein the C 6-10 aryl and 5-10 membered heteroaryl are optionally 1-8 (eg 1, 2, 3, 4, 5, 6, 7 or 8) R 9 group substitution.
  • R 6 is H; in another specific embodiment, R 6 is D.
  • the present invention relates to compounds of the following general formula:
  • the present invention relates to the following compounds:
  • the compounds of the present invention may include one or more asymmetric centers, and thus may exist in various stereoisomeric forms, for example, enantiomers and/or diastereomers.
  • the compounds of the present invention may be individual enantiomers, diastereomers, or geometric isomers (eg, cis and trans isomers), or may be in the form of mixtures of stereoisomers, This includes racemic mixtures and mixtures rich in one or more stereoisomers.
  • Isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and formation and crystallization of chiral salts; or preferred isomers can be obtained by Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • organic compounds can form a complex with a solvent in which they react or precipitate or crystallize out of the solvent. These complexes are called “solvates”. When the solvent is water, the complex is called “hydrate”. The present invention covers all solvates of the compounds of the present invention.
  • solvate refers to the form of a compound or salt thereof that is generally formed by a solvolysis reaction and is combined with a solvent. This physical association may include hydrogen bonding.
  • Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like.
  • the compounds described herein can be prepared, for example, in crystalline form, and can be solvated.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be able to separate, for example, when one or more solvent molecules are incorporated into the crystal lattice of the crystalline solid.
  • “Solvate” includes both solvates and isolatable solvates. Representative solvates include hydrates, ethanolates and methanolates.
  • hydrate refers to a compound that binds to water. Generally, the ratio of the number of water molecules contained in the hydrate of the compound to the number of molecules of the compound in the hydrate is determined. Therefore, the hydrate of the compound can be represented by, for example, the general formula R ⁇ x H 2 O, where R is the compound, and x is a number greater than 0.
  • R is the compound
  • x is a number greater than 0.
  • a given compound can form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5 H 2 O)) and polyhydrates (x is a number greater than 1, for example, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • the compound of the present invention may be in an amorphous or crystalline form (polymorph).
  • the compounds of the present invention may exist in one or more crystalline forms. Therefore, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention.
  • polymorph refers to a crystalline form (or a salt, hydrate, or solvate) of a compound in a specific crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, photoelectric properties, stability, and solubility. Recrystallization solvent, crystallization rate, storage temperature and other factors can cause a crystalline form to dominate.
  • Various polymorphs of the compound can be prepared by crystallization under different conditions.
  • the present invention also includes isotopically labeled compounds, which are equivalent to those described in formula (I), but one or more atoms are replaced by atoms having an atomic mass or mass number different from the atomic mass or mass number common in nature.
  • isotopes that can be introduced into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18, respectively O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • Compounds of the invention, prodrugs thereof, and pharmaceutically acceptable salts of the compounds or prodrugs containing the above isotopes and/or other isotopes of other atoms are within the scope of the invention.
  • Tritium, ie 3 H, and carbon-14, ie, 14 C isotopes are particularly preferred because they are easy to prepare and detect.
  • Isotope-labeled compounds of formula (I) of the present invention and their prodrugs can generally be prepared in such a way that, when performing the processes disclosed in the following procedures and/or examples and preparation examples, non-isotopically-labeled reagents are used instead of non-isotopic reagents Labeled reagents.
  • prodrugs are also included in the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted into its active form having a medical effect in the body by, for example, hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of AC Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral delivery: delivery: limits of overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each introduced This article is for reference.
  • a prodrug is any covalently bonded compound of the invention, and when such a prodrug is administered to a patient, it releases the parent compound in the body.
  • Prodrugs are usually prepared by modifying functional groups in such a way that the modification can be produced by conventional operations or by cleavage in vivo to produce the parent compound.
  • Prodrugs include, for example, compounds of the present invention in which a hydroxyl group, amino group, or sulfhydryl group is bonded to any group, which, when administered to a patient, can be cleaved to form a hydroxyl group, amino group, or thiol group.
  • prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of the hydroxyl, mercapto, and amino functional groups of compounds of formula (I).
  • esters such as methyl ester, ethyl ester and the like can be used.
  • the ester itself can be active and/or can be hydrolyzed under human body conditions.
  • Suitable pharmaceutically acceptable in vivo hydrolyzable ester groups include those groups that are easily decomposed in the human body to release the parent acid or its salt.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of active ingredient.
  • the pharmaceutical composition comprises a therapeutically effective amount of active ingredient.
  • the pharmaceutical composition comprises a prophylactically effective amount of active ingredient.
  • the pharmaceutically acceptable excipients used in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that can be used in the compositions of the present invention include, but are not limited to, ion exchangers, aluminum oxide, aluminum stearate, lecithin, serum proteins (such as human serum albumin ), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylate, wax, polyethylene-polyoxypropylene-embedded Segment polymers, poly
  • kits eg, pharmaceutical packaging.
  • the provided kit may include the compound of the present invention and other therapeutic agents, as well as the first and second containers containing the compound of the present invention and other therapeutic agents (eg, vials, ampoules, bottles, syringes, and/or dispersible packaging or other Suitable container).
  • the provided kit may also optionally include a third container containing a pharmaceutical excipient for diluting or suspending the compound of the invention and/or other therapeutic agents.
  • the compound of the present invention and other therapeutic agents provided in the first container and the second container are combined to form a unit dosage form.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intra-articular administration, intraarterial administration, intrasynovial administration, intrasternal administration , Intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
  • an effective amount of a compound provided herein is administered.
  • the amount of compound actually administered can be determined by the doctor .
  • the compounds provided herein are administered to a subject at risk of developing the disorder, typically based on doctor's recommendations and administered under the supervision of a doctor, at dosage levels as described above.
  • Subjects at risk of developing a particular disorder usually include subjects with a family history of the disorder, or those subjects who have been determined to be particularly sensitive to the development of the disorder by genetic testing or screening.
  • the pharmaceutical compositions provided herein can also be administered chronically ("long-term administration").
  • Long-term administration refers to administration of the compound or its pharmaceutical composition over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or continuous administration indefinitely, For example, the rest of the subject's life.
  • chronic administration is intended to provide a constant level of the compound in the blood over a long period of time, for example, within a therapeutic window.
  • the pharmaceutical composition may be administered by bolus injection, for example, in order to rapidly increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic level of the active component.
  • an intramuscular or subcutaneous bolus dose allows the active component to be slowly released, while a bolus delivered directly to the vein (eg, IV IV infusion) can be more The rapid delivery allows the concentration of the active component in the blood to rapidly increase to an effective level.
  • the pharmaceutical composition may be administered as a continuous infusion, for example, by IV IV infusion, thereby providing a steady state concentration of the active component in the body of the subject.
  • the bolus dose of the pharmaceutical composition can be administered first, and then the infusion can be continued.
  • Oral compositions can take the form of bulk liquid solutions or suspensions or bulk powders. However, more generally, to facilitate precise dosing, the composition is provided in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unit dosages for human patients and other mammals, each unit containing a predetermined number of active substances suitable for producing the desired therapeutic effect and suitable pharmaceutical excipients.
  • Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of liquid compositions, or pills, tablets, capsules, etc. in the case of solid compositions.
  • the compound is generally a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), and the remainder is each useful for forming the desired form of administration A variety of carriers or excipients and processing aids.
  • a typical regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides about 0.01 to about 20 mg/kg of the compound of the present invention, and the preferred doses each provide about 0.1 to about 10 mg/kg, especially about 1 to about 5 mg/kg.
  • the transdermal dose is usually selected in a quantity of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, preferably about 0.1 To about 10% by weight, and more preferably about 0.5 to about 15% by weight.
  • the injection dose level ranges from about 0.1 mg/kg/hour to at least 10 mg/kg/hour.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more can also be given.
  • the maximum total dose cannot exceed approximately 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous carriers and buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • Solid forms may include, for example, any of the following components, or compounds with similar properties: binders, for example, microcrystalline cellulose, tragacanth or gelatin; excipients, for example, starch or lactose, disintegrating agents, For example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, for example, colloidal silicon dioxide; sweeteners, for example, sucrose or saccharin; or flavoring agents, for example, mint, water Methyl salicylate or orange flavoring agent.
  • binders for example, microcrystalline cellulose, tragacanth or gelatin
  • excipients for example, starch or lactose
  • disintegrating agents For example, alginic acid, Primogel or corn starch
  • lubricants
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound is typically a minor component, often about 0.05 to 10% by weight, and the remainder is injectable excipients and the like.
  • the transdermal composition is typically formulated as a topical ointment or cream containing the active ingredient.
  • the active ingredient When formulated as an ointment, the active ingredient is typically combined with paraffin or a water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art, and generally include other components for enhancing the stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope provided by the present invention.
  • transdermal administration can be achieved using a reservoir or porous membrane type, or a variety of solid matrix patches.
  • compositions for oral administration, injection or topical administration are only representative.
  • Other materials and processing techniques are described in Remington's Pharmaceuticals, 17th Edition, 1985, Mack Publishing Company, Easton, Pennsylvania, Section 8. This document is incorporated by reference.
  • the compounds of the present invention can also be administered in a sustained release form or from a sustained release drug delivery system.
  • sustained-release materials can be found in Remington's Pharmaceuticals.
  • the invention also relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation contains water.
  • the formulation contains a cyclodextrin derivative.
  • the most common cyclodextrins are ⁇ -, ⁇ -, and ⁇ -cyclodextrins consisting of 6, 7 and 8 ⁇ -1,4-linked glucose units, which optionally include a Or multiple substituents, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substitutions.
  • the cyclodextrin is sulfoalkyl ether ⁇ -cyclodextrin, for example, sulfobutyl ether ⁇ -cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation includes hexapropyl- ⁇ -cyclodextrin (eg, 10-50% in water).
  • cancer refers to the abnormal growth of cells that proliferate in an uncontrolled manner and in some cases metastasize.
  • Types of cancer include but are not limited to solid tumors such as bladder, intestine, brain, breast, endometrium, heart, kidney, lung, lymphoid tissue (lymphoma), ovary, pancreas or other endocrine organs (thyroid), prostate, skin (Melanoma) or hematological tumors (such as leukemia).
  • EGFR mutation refers to a mutation of T790M (resistant or carcinogenic), L858R (activated), del19 (activated), or a combination thereof.
  • the present invention selectively inhibits one activating mutation and one point mutation.
  • at least one activating mutation or deletion mutation del19.
  • at least one activating mutation is a point mutation L858R.
  • at least one resistance mutation is a point mutation T790M.
  • at least one mutation of EGFR is L858R and/or T790M.
  • mutant selective inhibition used in contrast to the inhibition of wild-type EGFR means that the present invention inhibits at least one EGFR mutation (i.e. at least one) in at least one assay described herein (eg, biochemical or cellular).
  • at least one EGFR mutation i.e. at least one
  • assay described herein eg, biochemical or cellular
  • the state of a deletion mutation, at least one activating mutation, at least one resistance mutation, or a combination of at least one deletion mutation and at least one point mutation may be used herein, the present invention inhibits at least one EGFR mutation (i.e. at least one) in at least one assay described herein (eg, biochemical or cellular).
  • selective inhibition used in contrast to the inhibition of other kinases means that the present invention undesirably inhibits at least one kinase group.
  • the term “inhibitor” refers to a compound that inhibits one or more kinases described herein.
  • EGFR mutant inhibitor refers to a compound that inhibits the receptor of the EGFR mutant or reduces the effect of signal transduction.
  • protein kinase-mediated disease refers to any disease state mediated or regulated by the protein kinase described in the text. This disease state includes but is not limited to non-small cell lung cancer.
  • EGFR mutant-mediated disease refers to any disease state mediated or regulated by the EGFR mutant kinase mechanism. This disease state includes but is not limited to non-small cell lung cancer, metastatic brain cancer, and other solid cancers.
  • JAK3 mediated disease refers to any disease state mediated or regulated by the JAK3 kinase mechanism. This disease state includes but is not limited to rheumatoid arthritis, psoriasis and organ transplant rejection, and some solid cancers.
  • the present invention provides a method for inhibiting protein kinases (such as EGFR kinase) or treating diseases (such as cancer, cell proliferative diseases, inflammation, infection, immune diseases, organ transplantation, viral diseases, cardiovascular diseases or metabolic diseases) ) Method, comprising the steps of: administering to a subject in need of treatment a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug, or isotope derivative Or the pharmaceutical composition described in the present invention.
  • protein kinases such as EGFR kinase
  • diseases such as cancer, cell proliferative diseases, inflammation, infection, immune diseases, organ transplantation, viral diseases, cardiovascular diseases or metabolic diseases
  • the compound of the present invention or a pharmaceutically acceptable salt or pharmaceutical composition thereof can be used to treat cancer caused by EGFR.
  • the compounds can be used to treat cancers caused by EGFR expressing EGFR mutants and to treat cancers caused by EGFR refractory to RTKI therapy (eg, erlotinib or gefitinib).
  • the compounds of the present invention are inhibitors of at least one mutant of EGFR and are therefore suitable for treatment with one or more EGFR mutants (such as deletion mutations, activation mutations, resistance mutations or combinations thereof, specific examples include T790M mutations, L858R mutation, del19 and del19/T790M or L858R/T790M double mutation) activity related to one or more conditions.
  • EGFR mutants such as deletion mutations, activation mutations, resistance mutations or combinations thereof, specific examples include T790M mutations, L858R mutation, del19 and del19/T790M or L858R/T790M double mutation
  • the present invention provides a method of treating a mutant EGFR-mediated disorder, which comprises administering a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer thereof to a patient in need , Solvates, hydrates, crystal forms, prodrugs or isotope derivatives, or the steps of administering the pharmaceutical composition of the present invention.
  • the compounds of the present invention are JAK3 inhibitors and are therefore suitable for the treatment of one or more conditions associated with the activity of wild-type or mutant JAK3 kinase. Therefore, in a specific embodiment, the present invention provides a method of treating wild-type or mutant JAK3 kinase-mediated disorders, which comprises administering a compound of the present invention, or a pharmaceutically acceptable salt thereof, to a patient in need, Stereoisomers, solvates, hydrates, crystal forms, prodrugs or isotopic derivatives, or the steps of administering the pharmaceutical composition of the present invention.
  • Cancers treatable by the compounds of the present invention include, but are not limited to: non-small cell lung cancer (NSCLS), small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell carcinoma, Gastrointestinal stromal tumors, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma and other hyperproliferative diseases.
  • the compounds of the present invention can also be used to maintain the prevention of cancer recurrence in patients in need of such treatment.
  • effective amount is intended to mean an amount or dose sufficient to produce the desired therapeutic benefit in the individual in need of the treatment.
  • the effective amount or dose of the compound of the present invention can be determined by conventional methods (such as modeling, dose escalation, or clinical trials) and conventional factors (such as the mode or route of drug delivery, the pharmacokinetics of the agent, the severity and course of infection, the individual Health status and weight, and the judgment of the treating physician) to determine.
  • Exemplary dosages are in the range of about 0.1 mg to 1 g per day, or about 1 mg to 50 mg per day, or about 50 mg to 250 mg per day, or about 250 mg to 1 g per day.
  • the total dose can be a single or separate dosage unit (eg, BID, TID, QID).
  • the dose can be adjusted for prophylactic or maintenance treatment. For example, depending on the symptoms, the dosage or frequency of administration or both can be reduced to an amount that maintains the desired therapeutic or preventive effect.
  • the symptoms if the symptoms have been alleviated to an appropriate level, then treatment can be stopped. However, when any of the symptoms recurs, the patient may require long-term intermittent treatment. Patients may also require long-term slow treatment.
  • the compounds of the present invention described herein can be used in pharmaceutical compositions or methods in combination with one or more other active ingredients to treat the diseases and disorders described herein.
  • additional active ingredients include other therapeutic agents or agents that alleviate the adverse effects of the therapeutic agent against the intended disease target.
  • the combination can be used to increase efficacy, improve symptoms of other diseases, reduce one or more negative effects, or reduce the required dose of the compound of the present invention.
  • the additional active ingredient may be formulated as a pharmaceutical composition separate from the compound of the present invention or may be included in a single pharmaceutical composition with the compound of the present invention.
  • the additional active ingredient may be administered at the same time, before, or after the administration of the compound of the present invention.
  • Combination agents include those additional active ingredients that are known or observed to be effective in treating the diseases and disorders described herein, including those effective against another target associated with the disease.
  • the compositions and formulations and treatment methods of the present invention may further include other drugs or medicines, such as other active agents that can be used to treat or alleviate the target disease or related symptoms or conditions.
  • kinase inhibitors such as EGFR inhibitors (e.g., erlotinib, gefitinib); Raf inhibitors (e.g., vero Vemurafenib), VEGFR inhibitors (eg sunitinib); standard chemotherapeutic agents such as alkylating agents, antimetabolites, antitumor antibiotics, topoisomerase inhibitors, platinum drugs , Mitotic inhibitors, antibodies, hormone therapy or corticosteroids.
  • suitable combination agents include anti-inflammatory agents, such as NSAIDs.
  • the pharmaceutical composition of the present invention may additionally contain one or more of the active agents, and the method of treatment may further comprise administering an effective amount of one or more of the active agents.
  • each reaction is carried out in an inert solvent at room temperature to reflux temperature (eg, 0°C to 100°C, preferably 0°C to 80°C).
  • the reaction time is usually 0.1-60 hours, preferably 0.5-24 hours.
  • N-bromosuccinimide N-bromosuccinimide (NBS, 3.6 g, 20 mmol) was added, and the reaction was stirred at room temperature for 1 h.
  • the organic layer was separated, and the aqueous layer was extracted with ethyl acetate (30 mL ⁇ 3).
  • the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the solvent was distilled off under reduced pressure.
  • WT EGFR (Carna, catalog number 08-115), EGFR [L858R] (Carna, catalog number 08-502), EGFR [L858R/T790M] (Carna, catalog number 08-510), ATP (Sigma, catalog number A7699- 1G), DMSO (Sigma, catalog number D2650), 96-well plate (Corning, catalog number 3365), 384-well plate (Greiner, catalog number 784076), HTRF Kinase TK kit (Cisbio, catalog number 62TK0PEJ), Erlotti (Selleckchem, catalog number S7787), EGFR [d746-750] (Life Technologies, catalog number PV6178), 5x kinase buffer A (Life Technologies, catalog number PV3186), kinase tracer 199 (Life Technologies, catalog number PV5830 ), Eu-anti-GST antibody (Life Technologies, catalog number PV5594).
  • test compound preparation The test compound is dissolved in DMSO to make a 20 mM stock solution. Then, dilute three times in a DMSO medium gradient and dilute ten times. When adding drugs, dilute with buffer 10 times.
  • WT EGFR and EGFR [L858R/T790M] kinase detection In 5x kinase buffer A, WT EGFR or EGFR [L858R/T790M] kinase is mixed with pre-diluted compounds of different concentrations for 10 minutes, each concentration is double-well . Add the corresponding substrate and ATP, and react at room temperature for 20 minutes (in which a negative control is set: a negative control is a blank control, and a positive is erlotinib). After the reaction, the detection reagent (the reagent in the HTRF Kinase TK kit) was added.
  • the detection reagent the reagent in the HTRF Kinase TK kit
  • the enzyme activity in the presence of each concentration of the compound of the invention was measured by Evnvision microplate reader, and the compound pairs of different concentrations were calculated.
  • the inhibitory activity of the enzyme activity then according to the four-parameter equation, according to Graphpad 5.0 software to fit the inhibitory activity of the enzyme activity under different concentrations of compounds to calculate the IC 50 value.
  • the compound of the present invention was tested in the above kinase inhibition experiment, and it was found that the compound of the present invention has strong activity on EGFR [L858R/T790M] and excellent selectivity superior to WT EGFR.
  • the results of the representative example compounds are summarized in Table 1 below.
  • the MTS method was used to detect the anti-proliferative activity of the compound of the present invention against three tumor cells cultured in vitro.
  • the experimental results show that the compound of the present invention has an inhibitory effect on the in vitro proliferation of cancer cells cultured in vitro; wherein the inhibitory effect on the in vitro proliferation of lung cancer cells is stronger than that of skin cancer cells.
  • Cell lines skin cancer cells A431 (purchased from the American Standard Biological Collection Center (ATCC)); lung cancer cells NCI-H1975 (purchased from the American Standard Biological Collection Center (ATCC)) and HCC827 (purchased from the American Standard Biological Collection Center) (ATCC)); all were cultured in RPMI1640 medium containing 10% fetal bovine serum, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin.
  • ATCC American Standard Biological Collection Center
  • ATCC827 purchased from the American Standard Biological Collection Center
  • Reagents and consumables RPMI-1640 (GIBCO, catalog number A10491-01); fetal bovine serum (GIBCO, catalog number 10099141); 0.25% trypsin-EDTA (GIBCO, catalog number 25200); penicillin-streptomycin; liquid ( GIBCO, catalog number 15140-122); DMSO (Sigma, catalog number D2650); MTS test kit (Promega, catalog number G3581), 96-well plate (Corning, catalog number 3365).
  • test compound preparation The test compound is dissolved in DMSO to prepare a 20 mM stock solution and stored at -20°C. Dilute with DMSO and other gradients 3 times and 10 times. When adding drugs, dilute with cell culture medium 4 times.
  • MTS cell viability test 0.25% trypsin-EDTA digestion of logarithmic growth phase cells, inoculate 150 ⁇ l into 96-well plates at the optimized density, add compound diluted 4 times in medium after 24 hours, 50 ⁇ l/well (generally choose ten Concentration: 100, 33.3, 11.1, 3.70, 1.23, 0.412, 0.137, 0.0457, 0.0152, 0.00508 ⁇ M). As a control, wells with the same volume of 0.5% DMSO added. After the cells were continuously cultured for 72 hours, MTS detected cell viability.
  • the compound of the present invention was tested in the above-mentioned cytotoxicity experiment, and it was found that the compound of the present invention has potent activity against lung cancer cells NCI-H1975 and HCC827 and excellent selectivity superior to skin cancer cell A431.
  • the results of the inhibition of the in vitro proliferation of cancer cells by representative examples are summarized in Table 2 below.
  • Rats are fed on standard feed and given water. Fasting began 16 hours before the test.
  • the drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the orbit. The time points for blood collection were 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after administration.
  • the rats were briefly anesthetized, and 300 ⁇ L blood samples were collected in the test tubes. There is 30 ⁇ L of 1% heparin salt solution in the test tube. Before use, the test tube was dried at 60°C overnight. After the blood samples were collected at the last time point, the rats were sacrificed after ether anesthesia.
  • the blood sample was centrifuged at 5000 rpm for 5 minutes at 4°C to separate the plasma from the red blood cells. Pipette 100 ⁇ L of plasma into a clean plastic centrifuge tube, indicating the compound name and time point. Plasma was stored at -80°C until analysis. The concentration of the compound of the present invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the blood drug concentration of each animal at different time points.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Diabetes (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Obesity (AREA)
  • Transplantation (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

本发明涉及对蛋白激酶活性具有抑制作用的氨基嘧啶类化合物,以及它们的制备和用途。具体地,本发明公开了式(I)所示的氨基嘧啶类化合物、或其药学上可接受的盐、立体异构体、溶剂合物或水合物,以及含有该化合物的药物组合物和使用其的方法,包括治疗细胞增殖性疾病和病症如癌症和免疫疾病的方法。

Description

用于抑制蛋白激酶活性的氨基嘧啶类化合物
对相关申请的引用
本申请要求了2018年12月21日提交的中国专利申请号201811571875.1的优先权,将申请的全部内容并入本文作为说明书的一部分。
技术领域
本发明属于医药技术领域,尤其涉及蛋白酪氨酸激酶具有抑制作用的氨基嘧啶类化合物,包含它们的药物组合物,以及它们的制备方法和用途。
背景技术
蛋白激酶的失调已涉及许多疾病和病症,例如中枢神经系统疾病(例如阿尔茨海默病)、炎性和自身免疫性疾病(例如,哮喘、类风湿性关节炎、克罗恩病、炎性肠综合征和银屑病)、骨疾病(例如骨质疏松症)、代谢紊乱(例如糖尿病)、血管增生性疾病、眼病疾病、心血管疾病、癌症、再狭窄、疼痛感、移植排斥反应和感染性疾病。
其中,EGFR的过表达和失调通常在乳腺、肺、胰腺、头颈以及膀胱肿瘤中出现。EGFR是erbB受体家族的跨膜蛋白酪氨酸激酶成员。在生长因子配体如表皮生长因子(EGF)结合后,该受体可以与EGFR或另一家族成员例如erbB2(HER2)、erbB3(HER3)和erbB4(HER4)二聚化。erbB受体的二聚化导致细胞内结构域中关键酪氨酸残基的磷酸化,并继而刺激参与细胞增殖和存活的许多细胞内信号转导途径。erbB家族信号转导的失调促进增殖、侵袭、转移、血管生成和肿瘤存活,并已在许多人类癌症如肺癌和乳腺癌中有所描述。
因此,EGFR是抗癌药物开发的理想靶标,并且目前靶向EGFR的多种化合物临床可用,包括第一代抑制剂吉非替尼和厄洛替尼。据报道,最常见的EGFR激酶活性突变L858R和del19对吉非替尼或厄洛替尼的治疗敏感,但是最终获得主要卡口(gatekeeper)残基T790M的突变产生的吉非替尼或厄洛替尼治疗抗性,其在大约一半的临床耐药患者中检测到,这导致双突变体L858R/T790M和del19/T790M。
EGFR突变体的生物学和临床重要性在该领域得到认可,并且例如BIBW2992、HKI-272和PF0299804的几种第二代药物有效针对T790M抗性突变,但同时表现出对野生型(WT)EGFR强烈的抑制,这导致严重的不良作用。因此,对于有效抑制EGFR单突变和双突变体并且对WT EGFR具有选择性的化合物仍然存在着强烈的需求,从而为与EGFR突变体相关或由其介导的疾病提供有效且安全的临床疗法。
另一个涉及许多疾病和病症的蛋白激酶失调的实例是Janus激酶(JAK)3。与Janus家族成员JAK1、JAK2和Tyk2相对普遍存在的表达相反,JAK3主要在造血谱系中表达,如NK细胞、T细胞和B细胞以及肠细胞中表达,因此高度选择性的JAK3抑制剂应该具有针对免疫细胞的精确作用和最小的多效缺陷。JAK3抑制剂的选择性也优于目前广泛使用的具有丰富靶标和多种副作用的免疫抑制药物。 JAK3抑制剂可用于治疗自身免疫病,以及JAK3介导的白血病和淋巴瘤。
例如,在唐氏综合症儿童和非唐氏综合症成人两者中少数急性巨核细胞白血病(AMKL)患者中,以及在急性淋巴细胞白血病患者中,也鉴定到了JAK3的体细胞突变。此外,在几种淋巴增殖性疾病中鉴定到了JAK3激活,所述疾病包括套细胞(mantle cell)淋巴瘤、伯基特淋巴瘤、人T细胞白血病/淋巴瘤、病毒1诱导的成人T细胞淋巴瘤/白血病、和间变性大细胞淋巴瘤。已经显示JAK3/STAT途径的组成型激活在白血病和淋巴瘤细胞生长和存活以及侵袭性表型中具有主要作用。因此,可由JAK3激活性突变引起的JAK3的组成型激活是几种白血病和淋巴瘤的常见特征,因而对JAK3的选择性抑制可以成为治疗靶标。
因此,对于选择性并有效抑制JAK3野生型和突变体并且对其他JAK家族成员有选择性的化合物存在着强烈的需求,从而为与JAK3相关或由其介导的疾病提供有效且安全的临床疗法。
还对用于向有此需要的患者或对象施用这些化合物、药物制剂和药物的方法存在着需求。
发明概述
本发明提供了一种新的氨基嘧啶类化合物及包含该化合物的组合物及其用途,其对某些突变形式的EGFR、野生型和突变型的JAK3、SYK和KDR等激酶具有更好地抑制活性和选择性,且具有更优异的药代动力学性质,治疗EGFR突变体、JAK3、SYK或KDR等激酶介导的疾病。
对此,本发明采用以下技术方案:
在一方面中,本发明涉及式(I)化合物:
Figure PCTCN2019122399-appb-000001
其中,
X选自CH、CD或N;
环A选自至少含有一个N原子的五元杂芳环;
R 1选自H、D、卤素、-CN、-NO 2、-OH、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基或-OC 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-OC 3-7环烷基任选地被1-13个R 9基团取代;
R 2选自H、D、4-7元杂环烷基或-NR 7R 8,其中所述的4-7元杂环烷基任选地被1-10个R 9基团取代;
R 3选自H、D、卤素、-CN、-NO 2、C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基任选地被1-13个R 9基团取代;
R 4选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基,其中所述的C 6-10芳基和5-10元杂芳基任选地被1-13个R 9基团取代;
R 5选自H、D、-(CH 2) nOR 7、-(CH 2) nNR 7R 8、-(CD 2) nOR 7或-(CD 2) nNR 7R 8,其中n选自1、2、3 或4;
R 6选自H、D或C 1-6烷基,其中所述的C 1-6烷基任选地被1-13个R 9基团取代;
R 7和R 8各自独立地选自H、D、C 1-6烷基、C 3-7环烷基或4-7元杂环烷基,或者R 7、R 8与它们所连接的N原子一起形成4-7元杂环烷基;其中所述的C 1-6烷基、C 3-7环烷基或4-7元杂环烷基任选地被1-13个R 9基团取代;
R 9独立地选自H、D、卤素、-OH、C 1-6烷氧基、-NH 2、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、-C(O)C 1-6烷基、-C(O)OC 1-6烷基、-C(O)NHC 1-6烷基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;或者相同原子或相邻原子上的两个R 9基团可以一起形成C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;其中R 9定义中的每个基团任选地被一个或多个D取代,直至完全氘代;
前提是,当X为CH,且环A为
Figure PCTCN2019122399-appb-000002
时,R 1选自C 1-6卤代烷基或C 1-6卤代烷氧基;
或其药学上可接受的盐、立体异构体、溶剂合物或水合物。
在另一方面,本发明提供了一种药物组合物,其含有本发明化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,和药学上可接受的赋形剂。在具体实施方案中,本发明化合物以有效量提供在所述药物组合物中。在具体实施方案中,本发明化合物以治疗有效量提供。在具体实施方案中,本发明化合物以预防有效量提供。
在另一方面,本发明提供了本发明化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,或本发明药物组合物在制备用于治疗蛋白激酶介导的疾病的药物中的用途。
在另一方面,本发明提供了一种治疗受试者中蛋白激酶介导的疾病的方法,包括向所述受试者给药本发明化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,或本发明药物组合物。
在另一方面,本发明提供了本发明化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,或本发明药物组合物,其用于治疗蛋白激酶介导的疾病。
在具体实施方案中,所述疾病由至少一种突变型EGFR激酶介导。在具体实施方案中,所述至少一种突变型EFGR是del19、L858R或T790M。在某些实施方案中,所述至少一种突变型EGFR是选自del19/T790M或L858R/T790M的至少一种双突变体。
在具体实施方案中,所述疾病由野生型和/或突变型JAK3激酶介导。
由随后的具体实施方式、实施例和权利要求,本发明的其他目的和优点将对于本领域技术人员显而易见。
定义
化学定义
下面更详细地描述具体官能团和化学术语的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C 1-6烷基”包括C 1、C 2、C 3、 C 4、C 5、C 6、C 1-6、C 1-5、C 1-4、C 1-3、C 1-2、C 2-6、C 2-5、C 2-4、C 2-3、C 3-6、C 3-5、C 3-4、C 4-6、C 4-5和C 5-6烷基。
“C 1-6烷基”是指具有1-6个碳原子的直链或支链饱和烃基团,本文也称为“低级烷基”。在一些实施方案中,C 1-4烷基是特别优选的。所述烷基的实例包括但不限于:甲基(C 1)、乙基(C 2)、正丙基(C 3)、异丙基(C 3)、正丁基(C 4)、叔丁基(C 4)、仲丁基(C 4)、异丁基(C 4)、正戊基(C 5)、3-戊基(C 5)、戊基(C 5)、新戊基(C 5)、3-甲基-2-丁基(C 5)、叔戊基(C 5)和正己基(C 6)。不论烷基前是否修饰有“取代的”,烷基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“C 2-6烯基”是指具有2-6个碳原子和一个或多个碳-碳双键(例如,1、2或3个碳-碳双键)的直链或支链烃基团。一个或多个碳-碳双键可以在内部(例如,在2-丁烯基中)或端部(例如,在1-丁烯基中)。在一些实施方案中,C 2-4烯基是特别优选的。所述烯基的实例包括但不限于:乙烯基(C 2)、1-丙烯基(C 3)、2-丙烯基(C 3)、1-丁烯基(C 4)、2-丁烯基(C 4)、丁二烯基(C 4)、戊烯基(C 5)、戊二烯基(C 5)、己烯基(C 6),等等。不论烯基前是否修饰有“取代的”,烯基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“C 2-6炔基”是指具有2-6个碳原子、一个或多个碳-碳叁键(例如,1、2或3个碳-碳叁键)以及任选一个或多个碳-碳双键(例如,1、2或3个碳-碳双键)的直链或支链烃基团。在一些实施方案中,C 2-4炔基是特别优选的。在一些实施方案中,炔基不含有任何双键。一个或多个碳叁键可以在内部(例如,在2-丁炔基中)或端部(例如,在1-丁炔基中)。所述炔基的实例包括但不限于:乙炔基(C 2)、1-丙炔基(C 3)、2-丙炔基(C 3)、1-丁炔基(C 4)、2-丁炔基(C 4)、戊炔基(C 5)、己炔基(C 6),等等。不论炔基前是否修饰有“取代的”,炔基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“C 1-6亚烷基”是指除去C 1-6烷基的另一个氢而形成的二价基团,并且可以是取代或未取代的亚烷基。在一些实施方案中,C 1-4亚烷基是特别优选的。未取代的所述亚烷基包括但不限于:亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-)、亚丙基(-CH 2CH 2CH 2-)、亚丁基(-CH 2CH 2CH 2CH 2-)、亚戊基(-CH 2CH 2CH 2CH 2CH 2-)、亚己基(-CH 2CH 2CH 2CH 2CH 2CH 2-),等等。示例性的取代的所述亚烷基,例如,被一个或多个烷基(甲基)取代的所述亚烷基,包括但不限于:取代的亚甲基(-CH(CH 3)-、-C(CH 3) 2-)、取代的亚乙基(-CH(CH 3)CH 2-、-CH 2CH(CH 3)-、-C(CH 3) 2CH 2-、-CH 2C(CH 3) 2-)、取代的亚丙基(-CH(CH 3)CH 2CH 2-、-CH 2CH(CH 3)CH 2-、-CH 2CH 2CH(CH 3)-、-C(CH 3) 2CH 2CH 2-、-CH 2C(CH 3) 2CH 2-、-CH 2CH 2C(CH 3) 2-),等等。
“C 0-6亚烷基”包括化学键和如上定义的C 1-6亚烷基。
“C 1-6烷氧基”是指基团-OR,其中,R为取代或未取代的C 1-6烷基。在一些实施方案中,C 1-4烷氧基是特别优选的。具体的所述烷氧基包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、叔丁氧基、仲丁氧基、正戊氧基、正己氧基和1,2-二甲基丁氧基。
“卤代”或“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。在一些实施方案中,卤素基团是F、Cl或Br。在一些实施方案中,卤素基团是F或Cl。在一些实施方案中,卤素基团是F。
因此,“C 1-6卤代烷基”和“C 1-6卤代烷氧基”是指上述“C 1-6烷基”和“C 1-6烷氧基”,其被一个或多个卤素基团取代。在一些实施方案中,C 1-4卤代烷基是特别优选的,更优选C 1-2卤代烷基。在一些实施方案中,C 1-4卤代烷氧基是特别优选的,更优选C 1-2卤代烷氧基。示例性的所述卤代烷基包括但不限于:-CF 3、-CH 2F、-CHF 2、-CHFCH 2F、-CH 2CHF 2、-CF 2CF 3、-CCl 3、-CH 2Cl、-CHCl 2、2,2,2-三氟-1,1-二甲基-乙基,等等。示例性的所述卤代烷氧基包括但不限于:-OCH 2F、-OCHF 2、-OCF 3,等等。
“C 3-10环烷基”是指具有3-10个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C 3-7环烷基是优选的,C 3-6环烷基是特别优选的,更优选C 5-6环烷基。环烷基还包括其中上述环烷基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基(C 3)、环丙烯基(C 3)、环丁基(C 4)、环丁烯基(C 4)、环戊基(C 5)、环戊烯基(C 5)、环己基(C 6)、环己烯基(C 6)、环已二烯基(C 6)、环庚基(C 7)、环庚烯基(C 7)、环庚二烯基(C 7)、环庚三烯基(C 7)、环辛基(C 8)、环辛烯基(C 8)、二环[2.2.1]庚基(C 7)、二环[2.2.2]辛基(C 8)、环壬基(C 9)、环壬烯基(C 9)、环癸基(C 10)、环癸烯基(C 10)、八氢-1H-茚基(C 9)、十氢萘基(C 10)、螺[4.5]癸基(C 10),等等。不论环烷基前是否修饰有“取代的”,环烷基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“3-10元杂环烷基”或是指具有环碳原子和1-4个环杂原子的3-10元非芳香环系的基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环烷基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,3-7元杂环烷基是优选的,其为具有环碳原子和1-3个环杂原子的3-7元非芳香环系;在一些实施方案中,4-7元杂环烷基是优选的,其为具有环碳原子和1-3个环杂原子的4-7元非芳香环系;在一些实施方案中,3-6元杂环烷基是特别优选的,其为具有环碳原子和1-3个环杂原子的3-6元非芳香环系;更优选5-6元杂环烷基,其为具有环碳原子和1-3个环杂原子的5-6元非芳香环系。杂环烷基还包括其中上述杂环烷基环与一个或多个环烷基、芳基或杂芳基稠合的环体系,其中连接点在杂环烷基环上;且在这样的情况下,环成员的数目继续表示在杂环烷基环体系中环成员的数目。不论杂环烷基前是否修饰有“取代的”,杂环烷基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
示例性的包含一个杂原子的3元杂环烷基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基(thiorenyl)。示例性的含有一个杂原子的4元杂环烷基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环烷基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环烷基包括但不限于:二氧杂环戊烷基、氧硫杂环戊烷基(oxasulfuranyl)、二硫杂环戊烷基(disulfuranyl)和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环烷基包括但不限于:三唑啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环烷基包括但不限于:哌啶基、四氢吡喃基、二氢吡啶基和硫杂环己烷基(thianyl)。示例性的包含两个杂原子的6元杂环烷基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性的包含三个杂原子的6元杂环烷基包括但不限于:六氢三嗪基(triazinanyl)。示例性的含有一个杂原子的7元杂环烷基包括但不限于:氮杂环庚烷基、 氧杂环庚烷基和硫杂环庚烷基。示例性的包含一个杂原子的8元杂环烷基包括但不限于:氮杂环辛烷基、氧杂环辛烷基和硫杂环辛烷基。示例性的与C 6芳基环稠合的5元杂环烷基(在本文中也称作5,6-双环杂环烷基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与C 6芳基环稠合的6元杂环烷基(本文还指的是6,6-双环杂环烷基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。
“C 6-14芳基”是指具有6-14个环碳原子和零个杂原子的单环或多环的(例如,双环或三环)4n+2芳族环体系(例如,具有以环状排列共享的6、10或14个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“C 6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C 10芳基”;例如,萘基,例如,1-萘基和2-萘基)。在一些实施方案中,芳基具有十四个环碳原子(“C 14芳基”;例如,蒽基)。在一些实施方案中,C 6-10芳基是特别优选的,更优选C 6芳基。芳基还包括其中上述芳基环与一个或多个环烷基或杂环烷基稠合的环系统,而且连接点在所述芳基环上,在这种情况下,碳原子的数目继续表示所述芳基环系统中的碳原子数目。不论芳基前是否修饰有“取代的”,芳基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“5-10元杂芳基”是指具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。杂芳基双环系统在一个或两个环中可以包括一个或多个杂原子。杂芳基还包括其中上述杂芳基环与一个或多个环烷基或杂环烷基稠合的环系统,而且连接点在所述杂芳基环上,在这种情况下,碳原子的数目继续表示所述杂芳基环系统中的碳原子数目。在一些实施方案中,5-6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环或双环的4n+2芳族环体系。不论杂芳基前是否修饰有“取代的”,杂芳基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
示例性的含有一个杂原子的5元杂芳基包括但不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:三唑基、噁二唑基和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括但不限于:吡啶基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并三唑基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR aa、-ON(R bb) 2、-N(R bb) 2、-N(R bb) 3 +X -、-N(OR cc)R bb、-SH、-SR aa、-SSR cc、-C(=O)R aa、-CO 2H、-CHO、-C(OR cc) 2、 -CO 2R aa、-OC(=O)R aa、-OCO 2R aa、-C(=O)N(R bb) 2、-OC(=O)N(R bb) 2、-NR bbC(=O)R aa、-NR bbCO 2R aa、-NR bbC(=O)N(R bb) 2、-C(=NR bb)R aa、-C(=NR bb)OR aa、-OC(=NR bb)R aa、-OC(=NR bb)OR aa、-C(=NR bb)N(R bb) 2、-OC(=NR bb)N(R bb) 2、-NR bbC(=NR bb)N(R bb) 2、-C(=O)NR bbSO 2R aa、-NR bbSO 2R aa、-SO 2N(R bb) 2、-SO 2R aa、-SO 2OR aa、-OSO 2R aa、-S(=O)R aa、-OS(=O)R aa、-Si(R aa) 3、-OSi(R aa) 3、-C(=S)N(R bb) 2、-C(=O)SR aa、-C(=S)SR aa、-SC(=S)SR aa、-SC(=O)SR aa、-OC(=O)SR aa、-SC(=O)OR aa、-SC(=O)R aa、-P(=O) 2R aa、-OP(=O) 2R aa、-P(=O)(R aa) 2、-OP(=O)(R aa) 2、-OP(=O)(OR cc) 2、-P(=O) 2N(R bb) 2、-OP(=O) 2N(R bb) 2、-P(=O)(NR bb) 2、-OP(=O)(NR bb) 2、-NR bbP(=O)(OR cc) 2、-NR bbP(=O)(NR bb) 2、-P(R cc) 2、-P(R cc) 3、-OP(R cc) 2、-OP(R cc) 3、-B(R aa) 2、-B(OR cc) 2、-BR aa(OR cc)、烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(R bb) 2、=NNR bbC(=O)R aa、=NNR bbC(=O)OR aa、=NNR bbS(=O) 2R aa、=NR bb或=NOR cc取代;
R aa的每个独立地选自烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基,或者两个R aa基团结合以形成杂环烷基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R bb的每个独立地选自:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基,或者两个R bb基团结合以形成杂环烷基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R cc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基,或者两个R cc基团结合以形成杂环烷基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R dd的每个独立地选自:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR ee、-ON(R ff) 2、-N(R ff) 2,、-N(R ff) 3 +X -、-N(OR ee)R ff、-SH、-SR ee、-SSR ee、-C(=O)R ee、-CO 2H、-CO 2R ee、-OC(=O)R ee、-OCO 2R ee、-C(=O)N(R ff) 2、-OC(=O)N(R ff) 2、-NR ffC(=O)R ee、-NR ffCO 2R ee、-NR ffC(=O)N(R ff) 2、-C(=NR ff)OR ee、-OC(=NR ff)R ee、-OC(=NR ff)OR ee、-C(=NR ff)N(R ff) 2、-OC(=NR ff)N(R ff) 2、-NR ffC(=NR ff)N(R ff) 2、-NR ffSO 2R ee、-SO 2N(R ff) 2、-SO 2R ee、-SO 2OR ee、-OSO 2R ee、-S(=O)R ee、-Si(R ee) 3、-OSi(R ee) 3、-C(=S)N(R ff) 2、-C(=O)SR ee、-C(=S)SR ee、-SC(=S)SR ee、-P(=O) 2R ee、-P(=O)(R ee) 2、-OP(=O)(R ee) 2、-OP(=O)(OR ee) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基、杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代,或者两个偕R dd取代基可结合以形成=O或=S;
R ee的每个独立地选自烷基、卤代烷基、烯基、炔基、碳环基、芳基、杂环烷基和杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团 取代;
R ff的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基,或者两个R ff基团结合形成杂环烷基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R gg的每个独立地是:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OC 1-6烷基、-ON(C 1-6烷基) 2、-N(C 1-6烷基) 2、-N(C 1-6烷基) 3 +X -、-NH(C 1-6烷基) 2 +X -、-NH 2(C 1-6烷基) +X -、-NH 3 +X -、-N(OC 1-6烷基)(C 1-6烷基)、-N(OH)(C 1-6烷基)、-NH(OH)、-SH、-SC 1-6烷基、-SS(C 1-6烷基)、-C(=O)(C 1-6烷基)、-CO 2H、-CO 2(C 1-6烷基)、-OC(=O)(C 1-6烷基)、-OCO 2(C 1-6烷基)、-C(=O)NH 2、-C(=O)N(C 1-6烷基) 2、-OC(=O)NH(C 1-6烷基)、-NHC(=O)(C 1-6烷基)、-N(C 1-6烷基)C(=O)(C 1-6烷基)、-NHCO 2(C 1-6烷基)、-NHC(=O)N(C 1-6烷基) 2、-NHC(=O)NH(C 1-6烷基)、-NHC(=O)NH 2、-C(=NH)O(C 1-6烷基)、-OC(=NH)(C 1-6烷基)、-OC(=NH)OC 1-6烷基、-C(=NH)N(C 1-6烷基) 2、-C(=NH)NH(C 1-6烷基)、-C(=NH)NH 2、-OC(=NH)N(C 1-6烷基) 2、-OC(NH)NH(C 1-6烷基)、-OC(NH)NH 2、-NHC(NH)N(C 1-6烷基) 2、-NHC(=NH)NH 2、-NHSO 2(C 1-6烷基)、-SO 2N(C 1-6烷基) 2、-SO 2NH(C 1-6烷基)、-SO 2NH 2、-SO 2C 1-6烷基、-SO 2OC 1-6烷基、-OSO 2C 1-6烷基、-SOC 1-6烷基、-Si(C 1-6烷基) 3、-OSi(C 1-6烷基) 3、-C(=S)N(C 1-6烷基) 2、C(=S)NH(C 1-6烷基)、C(=S)NH 2、-C(=O)S(C 1-6烷基)、-C(=S)SC 1-6烷基、-SC(=S)SC 1-6烷基、-P(=O) 2(C 1-6烷基)、-P(=O)(C 1-6烷基) 2、-OP(=O)(C 1-6烷基) 2、-OP(=O)(OC 1-6烷基) 2、C 1-6烷基、C 1-6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 7碳环基、C 6-C 10芳基、C 3-C 7杂环烷基、C 5-C 10杂芳基;或者两个偕R gg取代基可结合形成=O或=S;其中,X -为反离子。
示例性的氮原子上取代基包括但不局限于:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR bb)R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基,或者连接至氮原子的两个R cc基团结合形成杂环烷基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环烷基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代,且其中R aa、R bb、R cc和R dd如上所述。
术语“药学上可接受的盐”是指,在可靠的医学判断范围内,适合与人和低等动物的组织接触而没有过度毒性、刺激性、变态反应等等,并且与合理的益处/危险比例相称的那些盐。药学上可接受的盐在本领域是众所周知的。例如,Berge等人在J.Pharmaceutical Sciences(1977)66:1-19中详细描述的药学上可接受的盐。本发明化合物的药学上可接受的盐包括衍生自合适的无机和有机酸和无机和有机碱的盐。药学上可接受的无毒的酸加成盐的实例是与无机酸形成的盐,例如盐酸、氢溴酸、磷酸、硫酸和高氯酸,或与有机酸形成的盐,例如乙酸、草酸、马来酸、酒石酸、枸橼酸、琥珀酸或丙二酸。也包括使用本领域常规方法形成的盐,例如,离子交换方法。其它药学上可接受的盐包括:已二酸盐、海藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯甲酸盐、重硫酸盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、柠檬酸盐、环戊丙酸盐、二葡糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、富马酸盐、葡萄糖酸盐、甘油磷酸盐、葡糖酸盐、半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖酸盐、乳酸盐、月桂酸盐、月桂基硫酸盐、苹果酸盐、马来酸盐、丙二酸盐、甲磺酸盐、2-萘磺酸 盐、烟酸盐、硝酸盐、油酸盐、草酸盐、棕榈酸盐、双羟萘酸盐、果胶酯酸盐、过硫酸盐、3-苯丙酸盐、磷酸盐、苦味酸盐、特戊酸盐、丙酸盐、硬脂酸盐、琥珀酸盐、硫酸盐、酒石酸盐、硫氰酸盐、对甲苯磺酸盐、十一烷酸盐、戊酸盐,等等。衍生自合适的碱的药学上可接受的盐包括碱金属、碱土金属、铵和N +(C 1-4烷基) 4盐。代表性的碱金属或碱土金属盐包括钠、锂、钾、钙、镁盐,等等。如果合适的话,其它的药学上可接受的盐包括与反离子形成的无毒的铵盐、季铵盐和胺阳离子,反离子例如卤离子、氢氧根、羧酸根、硫酸根、磷酸根、硝酸根、低级烷基磺酸根和芳基磺酸根。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。本文可互换使用术语“人”、“患者”和“受试者”。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
“组合”以及相关术语是指同时或依次给药本发明的治疗剂。例如,本发明化合物可以与另一治疗剂以分开的单位剂型同时或依次给药,或与另一治疗剂一起呈单一单位剂型同时给药。
具体实施方式
化合物
本文中,“本发明化合物”指的是以下的式(I)化合物-式(VII)化合物(包括各式的子集,例如式(II-1)化合物),或其药学上可接受的盐、立体异构体、溶剂合物或水合物。
在一个实施方案中,本发明涉及式(I)化合物:
Figure PCTCN2019122399-appb-000003
其中,
X选自CH、CD或N;
环A选自至少含有一个N原子的五元杂芳环;
R 1选自H、D、卤素、-CN、-NO 2、-OH、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基或-OC 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-OC 3-7环烷基任选地被1-13个R 9基团取代;
R 2选自H、D、4-7元杂环烷基或-NR 7R 8,其中所述的4-7元杂环烷基任选地被1-10个R 9基团取代;
R 3选自H、D、卤素、-CN、-NO 2、C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基任选地被1-13个R 9基团取代;
R 4选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基,其中所述的C 6-10芳基和5-10元杂芳基任选地被1-13个R 9基团取代;
R 5选自H、D、-(CH 2) nOR 7、-(CH 2) nNR 7R 8、-(CD 2) nOR 7或-(CD 2) nNR 7R 8,其中n选自1、2、3或4;
R 6选自H、D或C 1-6烷基,其中所述的C 1-6烷基任选地被1-13个R 9基团取代;
R 7和R 8各自独立地选自H、D、C 1-6烷基、C 3-7环烷基或4-7元杂环烷基,或者R 7、R 8与它们所连接的N原子一起形成4-7元杂环烷基;其中所述的C 1-6烷基、C 3-7环烷基或4-7元杂环烷基任选地被1-13个R 9基团取代;
R 9独立地选自H、D、卤素、-OH、C 1-6烷氧基、-NH 2、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、-C(O)C 1-6烷基、-C(O)OC 1-6烷基、-C(O)NHC 1-6烷基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;或者相同原子或相邻原子上的两个R 9基团可以一起形成C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;其中R 9定义中的每个基团任选地被一个或多个D取代,直至完全氘代;
前提是,当X为CH,且环A为
Figure PCTCN2019122399-appb-000004
时,R 1选自C 1-6卤代烷基或C 1-6卤代烷氧基;
或其药学上可接受的盐、立体异构体、溶剂合物或水合物。
X
在一个具体实施方案中,X为CH;在另一个具体实施方案中,X为CD;在另一个具体实施方案中,X为N。
环A
在一个具体实施方案中,环A选自
Figure PCTCN2019122399-appb-000005
在另一个具体实施方案中,环A选自
Figure PCTCN2019122399-appb-000006
Figure PCTCN2019122399-appb-000007
在另一个具体实施方案中,环A选自
Figure PCTCN2019122399-appb-000008
R 1
在一个具体实施方案中,R 1选自C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷 氧基或-OC 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-OC 3-7环烷基任选地被1-13个(例如1、2、3、4、5、6、7、8、9、10、11、12或13个)R 9基团取代;在一个具体实施方案中,R 1选自-OR,其中R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个(例如1、2、3、4、5、6、7、8、9、10、11、12或13个)R 9基团取代;在一个具体实施方案中,R 1为C 1-6卤代烷氧基,其中所述的C 1-6卤代烷氧基任选地被1-12个(例如1、2、3、4、5、6、7、8、9、10、11或12个)R 9基团取代;在一个具体实施方案中,R 1选自-OCH 3、-OCH 2CH 3、-OCH(CH 3)、-OCHF 2、-OCF 3、-OCH 2CF 3、-OCD 3、-OCD 2CH 3、-OCD(CD 3)、-OCDF 2、-OCF 3或-OCD 2CF 3;在一个具体实施方案中,R 1选自-OCHF 2、-OCF 3或-OCH 2CF 3
R 2
在一个具体实施方案中,R 2选自4-7元杂环烷基或-NR 7R 8,其中所述的4-7元杂环烷基任选地被1-10个(例如1、2、3、4、5、6、7、8、9或10个)R 9基团取代;在一个具体实施方案中,R 2选自-N(CH 3) 2
Figure PCTCN2019122399-appb-000009
R 3
在一个具体实施方案中,R 3选自H、D、卤素、-CN、-NO 2、C 1-6烷基或C 1-6卤代烷基,其中所述的C 1-6烷基和C 1-6卤代烷基任选地被1-13个(例如1、2、3、4、5、6、7、8、9、10、11、12或13个)R 9基团取代;在一个具体实施方案中,R 3选自H、F、Cl、-CH 3或CD 3
R 4
在一个具体实施方案中,R 4选自C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基,其中所述的C 6-10芳基和5-10元杂芳基任选地被1-8个(例如1、2、3、4、5、6、7或8个)R 9基团取代;在一个具体实施方案中,R 4选自C 6-10芳基或5-10元杂芳基,其中所述的C 6-10芳基和5-10元杂芳基任选地被1-8个(例如1、2、3、4、5、6、7或8个)R 9基团取代。
R 5
在一个具体实施方案中,R 5选自-(CH 2) nOR 7、-(CH 2) nNR 7R 8、-(CD 2) nOR 7或-(CD 2) nNR 7R 8,其中n 选自1、2、3或4;在一个具体实施方案中,R 5为-(CH 2) nNR 7R 8或-(CD 2) nNR 7R 8,其中n选自1或2;在一个具体实施方案中,R 5选自-CH 2N(CH 3) 2、-CH 2N(CH 3)(CH 2CH 3)、
Figure PCTCN2019122399-appb-000010
Figure PCTCN2019122399-appb-000011
-CD 2N(CH 3) 2、-CH 2N(CD 3) 2或-CD 2N(CD 3) 2
R 6
在一个具体实施方案中,R 6为H;在另一个具体实施方案中,R 6为D。
在更具体的实施方案中,本发明涉及以下通式化合物:
Figure PCTCN2019122399-appb-000012
Figure PCTCN2019122399-appb-000013
在更具体实施方案中,本发明涉及以下化合物:
Figure PCTCN2019122399-appb-000014
Figure PCTCN2019122399-appb-000015
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何 异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理缔合可包括氢键键合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、乙醚等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂合物包括药学上可接受的溶剂合物且进一步包括化学计量的溶剂合物和非化学计量的溶剂合物。在一些情况下,所述溶剂合物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂合物”包括溶液状态的溶剂合物和可分离的溶剂合物。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R·x H 2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R·0.5 H 2O))和多水合物(x为大于1的数,例如,二水合物(R·2 H 2O)和六水合物(R·6 H 2O))。
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。
本发明还包括同位素标记的化合物,它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如 3H和 14C)的那些可用于药物和/或底物组织分布测定。氚、即 3H和碳-14、即 14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即 2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(I)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(I)化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
药物组合物、制剂和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的活性组分。在一些实施方案中,所述药物组合物包含治疗有效量的活性组分。在一些实施方案中,所述药物组合物包含预防有效量的活性组分。
用于本发明的药学上可接受的赋形剂是指不会破坏一起配制的化合物的药理学活性的无毒载剂、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括但不限于,离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度快速提高至有效水平。推注剂量取决于活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注(例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药物组合物,而后持续输液。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、 调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington′s Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材料的描述可在Remington′s Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。
适应症
如本文所用,术语“癌症”是指以不受控制的方式增殖并且在一些情况下转移的细胞的异常生长。癌症的类型包括但不限于实体瘤,如膀胱、肠、脑、乳腺、子宫内膜、心脏、肾、肺、淋巴组织(淋巴瘤)、卵巢、胰腺或其它内分泌器官(甲状腺)、前列腺、皮肤(黑素瘤)或血液肿瘤(如白血病)的那些。
如本文所用,术语“EGFR突变”或“突变型EGFR”是指T790M(抗性或致癌性)、L858R(激活)、del19(激活)、或其组合的突变。
在某些实施方案中,本发明选择性地抑制一个激活性突变和一个点突变。在一些实施方案中,至少一个激活性突变或缺失突变,del19。在一些实施方案中,至少一个激活性突变是点突变L858R。在一些实施方案中,至少一个抗性突变是点突变T790M。在一些实施方案中,EGFR的至少一个突变 是L858R和/或T790M。
如本文所用,与野生型EGFR的抑制相对比使用的术语“突变体选择性抑制”是指本文所述的至少一种测定中(例如生化或细胞)本发明抑制至少一种EGFR突变(即至少一个缺失突变,至少一个激活性突变,至少一个抗性突变,或至少一个缺失突变和至少一个点突变的组合)的状态。
如本文所用,与其他激酶的抑制相对比使用的术语“选择性抑制”是指本发明不良地抑制至少一个激酶组。
如本文所用,术语“抑制剂”是指抑制本文所述的一种或更多种激酶的化合物。例如,术语“EGFR突变体抑制剂”是指抑制EGFR突变体受体或降低信号转导效应的化合物。
如本文所用,术语“蛋白激酶介导的疾病”是指文本所述的蛋白激酶介导或调节的任何疾病状态。这种疾病状态包括但不限于非小细胞肺癌。
如本文所用,术语“EGFR突变体介导的疾病“是指EGFR突变体激酶机制介导或调节的任何疾病状态。这种疾病状态包括但不限于非小细胞肺癌、转移性脑癌和其他实体癌。
如本文所用,术语“JAK3介导的疾病“是指由JAK3激酶机制介导或调节的任何疾病状态。这种疾病状态包括但不限于类风湿性关节炎、银屑病和器官移植排斥反应以及一些实体癌。
本发明提供了一种抑制蛋白激酶(如EGFR激酶)的方法或治疗疾病(如癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病)的方法,它包括步骤:给需要治疗的受试者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物。
本发明化合物或其药学上可接受的盐或药物组合物可用于治疗EGFR导致的癌症。尤其是,所述化合物可用于治疗表达EGFR突变体的EGFR导致的癌症和用于治疗对RTKI疗法(例如,厄洛替尼或吉非替尼)难治的EGFR导致的癌症。
本发明化合物是EGFR的至少一种突变体的抑制剂并且因此适用于治疗与一种或一种以上EGFR突变体(例如缺失突变、活化突变、抗性突变或其组合,具体实例包括T790M突变、L858R突变、del19和del19/T790M或L858R/T790M双突变)的活性相关的一种或一种以上病症。因此,在具体实施方案中,本发明提供一种治疗突变型EGFR介导的病症的方法,其包含向有需要的患者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物的步骤。
本发明化合物是JAK3抑制剂并且因此适用于治疗与野生型或突变型JAK3激酶的活性相关的一种或一种以上病症。因此,在具体实施方案中,本发明提供一种治疗野生型或突变型JAK3激酶介导的病症的方法,其包含向有需要的患者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物的步骤。
本发明化合物可治疗的癌症包括但不限于:非小细胞肺癌(NSCLS)、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、前列腺癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌等过度增殖性疾病。此外,本发明化合物也可用于在需要此类治疗的患者中起到预防癌症复发的维持作用。
在本发明的治疗方法中,“有效量”打算指足以在需要所述治疗的个体中产生所需治疗益处的量或剂量。本发明化合物的有效量或剂量可通过常规方法(例如模型化、剂量递增或临床试验)以及常规因素(例如药物递送的模式或途径、药剂的药代动力学、感染的严重程度和过程、个体的健康状况和体重、和治疗医师的判断)来确定。示例性剂量是在每天约0.1mg到1g、或每天约1mg到50mg、或每天约50mg到250mg或每天约250mg到1g的范围内。总剂量可以单一或分开剂量单位(例如,BID、TID、QID)。
在患者的疾病发生改善后,可调整剂量以便预防性或维持性治疗。例如,可根据症状将给药剂量或给药频率或二者降低到维持所需治疗或预防效应的量。当然,如果症状已减轻到适当程度,那么可停止治疗。然而,任一症状复发时,患者可能需要长期间歇治疗。患者还可需要长期缓慢治疗。
药物组合
本文所述的本发明化合物可与一或多种其它活性成份组合用于药物组合物或方法中以治疗本文所述的疾病和病症。其它额外活性成份包括缓和治疗剂针对预期疾病靶标的不利效应的其它治疗剂或药剂。所述组合可用于增加功效,改善其它疾病症状,降低一或多种负效应,或降低本发明化合物的所需剂量。额外活性成份可调配成与本发明化合物分开的药物组合物或可与本发明化合物包括在单一药物组合物中。额外活性成份可与本发明化合物的给药同时、在其之前或在其之后给药。
组合药剂包括那些已知或观察到在治疗本文所述疾病和病症中有效的额外活性成份,包括那些有效针对与疾病相关的另一靶标。举例来说,本发明的组合物和制剂、以及治疗方法可进一步包含其它药物或医药,例如其它可用于治疗或缓解目标疾病或相关症状或状况的活性剂。对于癌症适应症来说,其它所述药剂包括(但不限于)激酶抑制剂,例如EGFR抑制剂(例如,埃罗替尼、吉非替尼(gefitinib));Raf抑制剂(例如,维罗非尼(vemurafenib))、VEGFR抑制剂(例如,舒尼替尼(sunitinib));标准化学治疗剂,例如烷基化剂、抗代谢物、抗肿瘤抗生素、拓扑异构酶抑制剂、铂药物、有丝分裂抑制剂、抗体、激素疗法或皮质类固醇。对于疼痛适应症来说,适宜组合药剂包括消炎剂,例如NSAID。本发明的药物组合物可另外包含一或多种所述活性剂,并且治疗方法可另外包含给药有效量的一或多种所述活性剂。
实施例
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则份数和百分比为重量份和重量百分比。
通常,在制备流程中,各反应在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~80℃)下进行。反应时间通常为0.1-60小时,优选地为0.5-24小时。
实施例1 N-(5-((4-(4-((二甲基氨基)甲基)-3-苯基-1H-吡唑-1-基)嘧啶-2-基)氨基)-6-甲氧基-2-吗啉 代吡啶-3-基)丙烯酰胺(化合物T-1)的制备
Figure PCTCN2019122399-appb-000016
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000017
步骤1 化合物2的合成
依次往配有磁力搅拌的250mL单口烧瓶中加入4-氯-2-甲硫基嘧啶(化合物1,7.25mL,62.3mmol)和95%乙醇(100mL),搅拌溶清,冷却到0℃,缓慢滴加入四水钼酸铵(2.18g,1.87mmol)的双氧水(30%,14.4mL,187mmol)预冷却溶液,滴完后升温到室温,氮气下搅拌反应过夜。减压蒸除大部分有机溶剂,加水(200mL),二氯甲烷萃取(70mLx3),合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩,硅胶柱层析得白色固体10.2g,收率85.7%。 1H NMR(400MHz,CDCl 3)δ8.82(d,J=5.6Hz,1H),7.63(d,J=4.8Hz,1H),3.33(s,3H)。
步骤2 化合物4的合成
向配有磁力搅拌和冷凝管的50mL单口烧瓶中加入乙醇/水混合液(80mL,2/1)和化合物3(3.0g, 15.9mmol),搅拌下加入还原铁粉(4.44g,79.6mmol)和氯化铵(4.25g,79.6mmol),氮气下升温到85℃,并保温搅拌反应1h。冷却到室温,滤除不溶性固体,减压蒸除有机溶剂,加入饱和NaHCO 3水液(5mL),乙酸乙酯萃取(50mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩得棕色固体2.47g,收率97.9%。LC-MS(APCI):m/z=159.1(M+1) +
步骤3 化合物5的合成
向配有磁力搅拌的50mL单口瓶中依次加化合物4(1.5g,9.46mmol)和甲酸(20mL),混合物氮气下升温到100℃并搅拌反应1小时。冷却到室温,减压蒸除未反应的甲酸,残留物加入乙酸乙酯(50mL)和饱和NaHCO 3(30mL),搅拌5分钟,分出有机层,水相乙酸乙酯萃取(40mLx2),合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,浓缩得棕色固体1.59g,收率90.1%。LC-MS(APCI):m/z=187.1(M+1) +.
步骤4 化合物6的合成
向配有磁力搅拌的50mL单口瓶中依次加化合物5(1.58g,8.47mmol)和三氟醋酸酐(10ml),冷却到-5℃,缓慢滴加入浓硝酸(533mg,8.47mmol)。滴毕,氮气氛下保温搅拌反应一个半小时。反应混合物小心倒入碎冰中(200g),搅拌20分钟,过滤,滤饼水洗(10mL),溶于乙酸乙酯(50mL)中,无水硫酸钠干燥,过滤,浓缩,过硅胶柱得黄色固体717mg,收率36.6%。LC-MS(APCI):m/z=232.2(M+1) +.
步骤5 化合物7的合成
向配有磁力搅拌的50mL单口瓶中依次加化合物6(710mg,3.07mmol)和乙腈(10ml),搅拌溶清,加入吗啉(401mg,4.6mmol)和三乙胺(465mg,4.6mmol),混合物氮气氛下升温到80℃并保温搅拌3小时。冷却到室温,减压蒸除溶剂,加入乙酸乙酯(20mL)和水(20mL),搅拌5分钟,过滤,滤饼水洗(10mL),烘干得黄色固体660mg,收率76.3%。LC-MS(APCI):m/z=283.2(M+1) +. 1H NMR(400MHz,DMSO-D 6)δ9.97(s,1H),9.04(s,1H),8.28(s,1H),4.03(s,3H),3.71(t,J=5.6Hz,4H),3.42(t,J=5.6Hz,4H).
步骤6 化合物8的合成
向配有磁力搅拌50mL三口瓶中加入化合物7(200mg,0.71mmol)和无水DMF(3mL),冷却到0℃,氮气氛下加入NaH(34mg,0.85mmol,60%),拆去冰浴,氮气氛下室温搅拌反应30分钟,再次冷却到0℃,缓慢滴加入DMF溶液(164mg,0.85mmol,2mL),滴毕,混合物室温继续搅拌反应2小时。加入饱和NaHCO 3水液(20mL)淬灭反应,并搅拌2小时,析出固体过滤,滤饼水洗(10mL),溶于二氯甲烷(20mL)中,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得黄色固体115mg,收率44.2%。LC-MS(APCI):m/z=367.1(M+1) +.
步骤7 化合物10的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物9(60mg,348umol)和干燥的DMF(2ml),冷却到0℃,加入NaH(60%,18mg,453umol),氮气下室温搅拌反应半小时,然后冷却到0℃,缓慢滴加入化合物8(115mg,314umol)的干燥DMF(2ml)溶液,滴毕,反应液升到室温并升温到60℃,保温搅拌反应2h。加入水(25mL)淬灭反应,搅拌2h,乙酸乙酯萃取(50mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得黄色固体130mg,收率74.2%。LC-MS(APCI):m/z =503.3(M+1) +
步骤8 化合物11的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物10(130mg,259umol)和二氯甲烷/甲醇溶液(4mL,1/1),搅拌溶清,加入二甲胺的甲醇溶液(2.59mL,2M)和冰醋酸(1滴),氮气氛下室温搅拌10分钟,缓慢加入氰基硼氢化钠(49mg,466umol),室温下搅拌反应2小时。加入水(10mL)淬灭反应,搅拌10分钟,减压蒸除有机溶剂,析出固体过滤,少量水洗,干燥得白色固体108mg,收率78.5%。LC-MS(APCI):m/z=532.3(M+1) +
步骤9 化合物12的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物11(108mg,203ummol)和乙醇/水混合液(6mL,2/1),搅拌溶清,加入还原铁粉(113mg,2.03mmol)和氯化铵(54mg,1.02mmol),升温到85℃并氮气氛下保温搅拌反应1小时。冷却到室温,过滤,滤液减压浓缩蒸除乙醇,残留液中加入饱和NaHCO 3水液(5mL),二氯甲烷萃取(15mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩得棕色固体100mg,收率98.1%。LC-MS(APCI):m/z=502.3(M+1) +.
步骤10 化合物T-1的合成
向配有磁力搅拌50mL三口烧瓶中加入干燥二氯甲烷(10mL)和化合物12(100mg,199umol),搅拌溶清,冷却到-10℃,加入三乙胺(40mg,399mmol),氮气下缓慢滴加入丙烯酰氯(217mg,199umol)的二氯甲烷(1mL)溶液,滴毕,-10℃保温搅拌反应30分钟。加入饱和Na 2CO 3水液(5mL)淬灭反应,搅拌10分钟,分出有机层,水相二氯甲烷萃取(10mLx2),合并有机相,无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得白色固体70mg,收率63.2%。LC-MS(APCI):m/z=556.3(M+1) +1H NMR(400MHz,CDCl 3)δ9.93(s,1H),9.27(s,1H),8.49(d,J=5.6Hz,1H),8.16-8.08(m,3H),7.70(s,1H),7.54-7.42(m,4H),6.60-6.56(m,1H),6.39-6.33(m,1H),5.86(d,J=10.4Hz,1H),4.04(s,3H),3.92(t,J=4.0Hz,4H),3.58(s,2H),3.06(t,J=4.0Hz,4H),2.34(s,6H).
实施例2 N-(5-((4-(4-((二甲基氨基)甲基-3-苯基-1H-吡唑-1-基-5-氟嘧啶-2-基)氨基)-6-甲氧基-2-吗 啉代吡啶-3-基)丙烯酰胺(化合物T-2)的制备
Figure PCTCN2019122399-appb-000018
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000019
步骤1 化合物14的合成
向配有磁力搅拌和冷凝管的100mL单口瓶中依次加入化合物9(860mg,5mmol)和乙腈(15mL),搅拌溶清,再加入碳酸钾(1.38g,20mmol)和化合物13(800mg,5mmol),氮气下室温搅拌反应过夜。减压蒸除溶剂,加入水(20mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得白色固体1.0g,收率64.1%。LC-MS(APCI):m/z=303.1(M+1) +1H NMR(400MHz,CDCl 3)δ/ppm:10.20(s,1H),9.21(s,1H),8.74(d,J=2.8Hz,1H),7.95-7.92(m,2H),7.59-7.57(m,3H).
步骤2 化合物15的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物14(900mg,3.0mmol)和乙醇(20mL,),搅拌溶清,加入二甲胺的四氢呋喃溶液(1.5mL,3mmol,2M)和冰醋酸(2滴),氮气氛下室温搅拌2分钟,缓慢加入氰基硼氢化钠(370mg,466umol),室温下搅拌反应2小时。减压蒸除溶剂,加入水(20mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤、浓缩并过硅胶柱得白色固体340mg,收率33.2%。LC-MS(APCI):m/z=332.1(M+1) +
步骤3 化合物16的合成
向配有磁力搅拌的50mL单口瓶中加入化合物7(2.3g,8.1mmol)和四氢呋喃(10mL),滴加入NaOH水溶液(2.4g溶于10mL,60mmol),氮气氛下室温搅拌反应2小时。二氯甲烷萃取(30mLx3),合并有机相,饱和食盐水洗涤(30mL),无水硫酸钠干燥,过滤,浓缩并过硅胶柱得棕色固体800mg,收率39.2%。LC-MS(APCI):m/z=255.1(M+1) +.
步骤4 化合物17的合成
向配有磁力搅拌的10mL微波管中依次加化合物15(166mg,0.5mmol)、化合物16(127mg,0.5mmol)和叔丁醇(5mL),搅拌溶清,再加入碳酸钾(138mg,1.0mmol)、xphos(2-二环己基磷-2,4,6-三异丙基联苯,48mg,0.1mmol)和Pd 2(dab) 3(三(二亚苄基丙酮)二钯,46mg,0.05mmol),抽真空并氮气置换3次,置于微波反应器中,升温到160℃并保温搅拌反应1小时。冷却到室温,加入乙酸乙酯(30mL)稀释,滤掉不溶性固体,滤液浓缩,残留物过硅胶柱得白色固体129mg,收率为47.2%。LC-MS(APCI):m/z=550.2(M+1) +.
步骤5 化合物18的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物17(112mg,203ummol)和乙醇/水混合液(6mL,2/1),搅拌溶清,加入还原铁粉(113mg,2.03mmol)和氯化铵(54mg,1.02mmol),升温到85℃并氮气氛下保温搅拌反应1小时。冷却到室温,过滤,滤液减压浓缩蒸除乙醇,残留液中加入饱和NaHCO 3水液(5mL),二氯甲烷萃取(15mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩得棕色固体100mg,收率98.1%。LC-MS(APCI):m/z=520.3(M+1) +.
步骤6 化合物T-2的合成
向配有磁力搅拌50mL三口烧瓶中加入干燥二氯甲烷(10mL)和化合物18(100mg,199umol),搅拌溶清,冷却到-10℃,加入三乙胺(40mg,399mmol),氮气下缓慢滴加入丙烯酰氯(217mg,199umol)的二氯甲烷(1mL)溶液,滴毕,-10℃保温搅拌反应30分钟。加入饱和Na 2CO 3水液(5mL)淬灭反应,搅拌10分钟,分出有机层,水相二氯甲烷萃取(10mLx2),合并有机相,无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得白色固体70mg,收率63.2%。LC-MS(APCI):m/z=574.3(M+1) +1H NMR(400MHz,DMSO-D 6)δ10.37(br s,1H),9.48(s,1H),9.02(br s,1H),8.68(d,J=4.0Hz,1H),8.64(s,1H),8.29(br s,1H),7.79-7.77(m,2H),7.54-7.46(m,3H),6.68-6.61(m,1H),6.28(d,J=17.6Hz,1H),5.79-5.76(m,1H),4.32(s,2H),3.90(m,3H),3.75(t,J=4.4Hz,4H),3.12(t,J=4.4Hz,4H),2.55(s,6H).
实施例3 N-(5-((4-(4-((二甲基氨基)甲基)-3-苯基-1H-吡唑-1-基)-5-甲基嘧啶-2-基)氨基)-6-甲氧基 -2-吗啉代吡啶-3-基)丙烯酰胺(化合物T-3)的制备
Figure PCTCN2019122399-appb-000020
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000021
步骤1 化合物20的合成
向配有磁力搅拌和冷凝管的100mL单口瓶中依次加入化合物9(860mg,5mmol)和乙腈(15mL),搅拌溶清,再加入碳酸钾(1.38g,20mmol)和化合物19(800mg,5mmol),氮气下室温搅拌反应过夜。减压蒸除溶剂,加入水(20mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得白色固体1.0g,收率64.1%。LC-MS(APCI):m/z=299.1(M+1) +1H NMR(300MHz,DMSO-D 6)δ10.11(s,1H),9.46(s,1H),8.89(s,1H),8.01-7.98(m,2H),7.54-7.52(m,3H),2.68(s,3H).
步骤2 化合物21的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物20(900mg,3.0mmol)和乙醇(20mL,),搅拌溶清,加入二甲胺的四氢呋喃溶液(1.5mL,3mmol,2M)和冰醋酸(2滴),氮气氛下室温搅拌2分钟,缓慢加入氰基硼氢化钠(370mg,466umol),室温下搅拌反应2小时。减压蒸除溶剂,加入水(20mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤、浓缩并过硅胶柱得白色固体340mg,收率33.2%。LC-MS(APCI):m/z=328.1(M+1) +
步骤3 化合物22的合成
向配有磁力搅拌的10mL微波管中依次加化合物21(166mg,0.5mmol)、化合物16(127mg,0.5mmol)和叔丁醇(5mL),搅拌溶清,再加入碳酸钾(138mg,1.0mmol)、xphos(48mg,0.1mmol)和Pd 2(dab) 3(46mg,0.05mmol),抽真空并氮气置换3次,置于微波反应器中,升温到160℃并保温搅拌反应1小时。冷却到室温,加入乙酸乙酯(30mL)稀释,滤掉不溶性固体,滤液浓缩,残留物过硅胶柱得白色固体129mg,收率为47.2%。LC-MS(APCI):m/z=546.2(M+1)+. 1H NMR(400MHz,DMSO-D 6)δ8.97(s,1H),8.64(s,1H),8.59(s,1H),8.46(s,1H),7.98-7.96(m,2H),7.50-7.46(m,2H),7.42-7.40(m,1H),4.00(s,3H),3.72(t,J=4.8Hz,4H),3.42(t,J=4.8Hz,4H),3.40(s,2H),2.57(s,3H),2.22(s,6H).
步骤4 化合物23的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物22(112mg,203ummol)和乙醇/水混合液(6mL,2/1),搅拌溶清,加入还原铁粉(113mg,2.03mmol)和氯化铵(54mg,1.02mmol),升温到 85℃并氮气氛下保温搅拌反应1小时。冷却到室温,过滤,滤液减压浓缩蒸除乙醇,残留液中加入饱和NaHCO 3水液(5mL),二氯甲烷萃取(15mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩得棕色固体100mg,收率98.1%。LC-MS(APCI):m/z=516.3(M+1) +.
步骤5 化合物T-3的合成
向配有磁力搅拌50mL三口烧瓶中加入干燥二氯甲烷(10mL)和化合物23(100mg,199umol),搅拌溶清,冷却到-10℃,加入三乙胺(40mg,399mmol),氮气下缓慢滴加入丙烯酰氯(217mg,199umol)的二氯甲烷(1mL)溶液,滴毕,-10℃保温搅拌反应30分钟。加入饱和Na 2CO 3水液(5mL)淬灭反应,搅拌10分钟,分出有机层,水相二氯甲烷萃取(10mLx2),合并有机相,无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得白色固体70mg,收率63.2%。LC-MS(APCI):m/z=570.3(M+1) +1H NMR(300MHz,CDCl 3)δ9.71(br s,1H),9.63(s,1H),8.39(s,1H),8.26(s,1H),7.71(d,J=6.6Hz,2H),7.66(s,1H),7.55-7.46(m,3H),6.57-6.51(m,1H),6.43-6.34(m,1H),5.97(d,J=10.2Hz,1H),4.46(s,2H),4.03(s,3H),3.90(t,J=4.5Hz,4H),3.05(t,J=4.5Hz,4H),2.70(s,3H),2.48(s,6H).
实施例4 N-(5-((4-(4-((二甲基氨基)甲基)-3-苯基-1H-吡唑-1-基)-嘧啶-2-基)氨基)-6-异丙氧基-2-吗 啉代吡啶-3-基)丙烯酰胺(化合物T-4)的制备
Figure PCTCN2019122399-appb-000022
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000023
步骤1 化合物24的合成
向配有磁力搅拌的50mL三口瓶中依次加化合物9(512mg,3.0mmol)和乙醇(20mL,),搅拌溶清,加入二甲胺的四氢呋喃溶液(1.5mL,3mmol,2M)和冰醋酸(2滴),氮气氛下室温搅拌2分钟,缓慢加入氰基硼氢化钠(370mg,466umol),室温下搅拌反应2小时。减压蒸除溶剂,加入水(20mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤、浓缩并过硅胶柱得白色固体400mg,收率66.0%。LC-MS(APCI):m/z=202.1(M+1) +1H NMR(300MHz,CDCl 3)δ7.72-7.68(m,2H),7.54(s,1H),7.45-7.28(m,3H),3.44(s,2H),2.25(s,6H).
步骤2 化合物26的合成
向配有磁力搅拌和冷凝管的50mL三口瓶中依次加入化合物24(400mg,2mmol)和无水DMF(10mL),搅拌溶清,冰水浴冷却,加入NaH(60%,96mg,2.4mmol),搅拌10分钟,缓慢滴加入化合物25(357mg,2.4mmol)的DMF溶液(2mL),拆去冰浴,室温搅拌1小时,再升温到60℃反应30分钟。冷却到室温,加入水(100mL),析出大量固体,过滤,溶于二氯甲烷(50mL)中,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得黄色固体350mg,收率55.7%。LC-MS(APCI):m/z=314.1(M+1) +
步骤3 化合物28的合成
向配有磁力搅拌的100mL单口瓶中依次加化合物27(5.8g,30.05mmol)和乙腈(60mL,),搅拌溶清,冰水浴冷却下依次滴加入吗啉(2.62g,30.05mmol)和三乙胺(4.56g,45.08mmol),氮 气氛下保温搅拌反应10分钟。加入二氯甲烷(200mL),水洗(50mLx2),饱和食盐水洗(30mL),无水硫酸钠干燥,过滤,浓缩并过硅胶柱得黄色固体5.91g,收率80.7%。LC-MS(APCI):m/z=244.1(M+1) +.
步骤4 化合物29的合成
向配有磁力搅拌和冷凝管的50mL三口瓶中依次加入NaH(490mg,12.24mmol),抽真空并氮气保护,冰水浴冷却下缓慢滴加入异丙醇(630mg,10.49mmol)的无水THF(25mL)溶液,搅拌10分钟,再次滴加入化合物28(2.13g,8.74mmol)的无水THF(5mL),滴毕,拆去冰浴,氮气氛下室温搅拌反应过夜。加入饱和水的乙酸乙酯(60mL),过滤,滤液无水硫酸钠干燥,过滤,浓缩并过硅胶柱得黄色固体1.13g,收率48.3%。LC-MS(APCI):m/z=268.2(M+1) +.
步骤5 化合物30的合成
向配有磁力搅拌的100mL单口瓶中依次加化合物29(1.12g,4.19mmol)和乙腈(20mL,),搅拌溶清,冰水浴冷却,缓慢加入NBS(N-溴代琥珀酰亚胺,746mg,4.19mmol),加完后,拆去冰浴,氮气氛下室温搅拌反应1小时。减压蒸除溶剂,残留物中加入水(20mL),二氯甲烷萃取(30mLx2),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得黄色固体1.0g,收率68.9%。LC-MS(APCI):m/z=346.2(M+1) +.
步骤6 化合物31的合成
向配有磁力搅拌的10mL微波管中依次加化合物30(200mg,0.58mmol)和DMSO(2mL),搅拌溶清,加入CuI(22mg,0.12mmol)、K 2CO 3(120mg,0.87mmol)和L-脯氨酸(26mg,0.23mmol),抽真空并氮气置换3次,加入氨水(108mg,0.87mmol),密封并置于微波反应器中,升温到150℃并保温搅拌反应1小时。冷却到室温,加入乙酸乙酯(30mL)稀释,滤掉不溶性固体,滤液浓缩,残留物过硅胶柱得白色固体52mg,收率为31.9%。LC-MS(APCI):m/z=283.2(M+1) +. 1H NMR(300MHz,CDCl 3)δ7.67(s,1H),5.39-5.31(m,1H),3.85(t,J=4.5Hz,4H),3.34(t,J=4.5Hz,4H),1.41(d,J=6.0Hz,6H).
步骤7 化合物32的合成
向配有磁力搅拌的10mL微波管中依次加化合物31(50mg,0.18mmol)、化合物26(67mg,0.21mmol)和叔丁醇(3mL),搅拌溶清,再加入碳酸钾(37mg,0.27mmol)、xphos(8mg)和Pd 2(dab) 3(8mg),抽真空并氮气置换3次,置于微波反应器中,升温到160℃并保温搅拌反应1小时。冷却到室温,加入乙酸乙酯(30mL)稀释,滤掉不溶性固体,滤液浓缩,残留物过硅胶柱得白色固体67mg,收率为47.2%。LC-MS(APCI):m/z=560.2(M+1)+. 1H NMR(300MHz,CDCl 3)δ9.65(s,1H),8.70(s,1H),8.51(d,J=5.4Hz,1H),8.03-8.00(m,2H),7.52-7.28(m,5H),5.47-5.43(m,1H),3.88(t,J=4.8Hz,4H),3.51-3.46(m,6H),2.36(s,6H),1.47(d,J=6.0Hz,6H).
步骤8 化合物33的合成
向配有磁力搅拌的50mL单口瓶中依次加化合物32(67mg,120ummol)和乙醇/水混合液(3mL,2/1),搅拌溶清,加入还原铁粉(67mg,1.2mmol)和氯化铵(32mg,0.6mmol),升温到85℃并氮气氛下保温搅拌反应1小时。冷却到室温,过滤,滤液减压浓缩蒸除乙醇,残留液中加入饱和NaHCO 3水液(5mL),二氯甲烷萃取(15mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩得棕色固体60mg,收率94.6%。LC-MS(APCI):m/z=530.3(M+1) +.
步骤9 化合物T-4的合成
向配有磁力搅拌50mL三口烧瓶中加入干燥二氯甲烷(10mL)和化合物33(60mg,199umol),搅拌溶清,冷却到-10℃,加入三乙胺(23mg,226umol),氮气下缓慢滴加入丙烯酰氯(15mg,169umol)的二氯甲烷(1mL)溶液,滴毕,-10℃保温搅拌反应30分钟。加入饱和Na 2CO 3水液(5mL)淬灭反应,搅拌10分钟,分出有机层,水相二氯甲烷萃取(10mLx2),合并有机相,无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得白色固体50mg,收率75.6%。LC-MS(APCI):m/z=584.3(M+1) +1H NMR(300MHz,CDCl 3)δ9.83(s,1H),8.51(d,J=5.4Hz,1H),8.23(s,1H),7.85-7.80(m,1H),7.74(s,1H),7.54-7.28(m,4H),6.57-6.52(m,1H),6.41-6.32(m,1H),5.94-5.39(m,1H),5.41-5.36(m,1H),3.91(t,J=4.5Hz,4H),3.04(t,J=4.5Hz,4H),2.43(s,6H),1.43(d,J=6.3Hz,6H).
实施例5 N-(5-((4-(2-((二甲基氨基)甲基)-1-苯基-1H-咪唑-4-基)嘧啶-2-基)氨基)-4-甲氧基-2-吗啉 代苯基)丙烯酰胺(化合物T-5)的制备
Figure PCTCN2019122399-appb-000024
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000025
Figure PCTCN2019122399-appb-000026
步骤1 化合物34的合成
向配有磁力搅拌和冷凝管的100mL双口烧瓶中加入化合物1(3.2g,20mmol)和甲苯(50mL),搅拌溶清,加入四(三苯基膦)钯(1.2g,1mmol),抽真空并氮气置换3次,氮气氛下加入三丁基(1-乙氧基乙烯)锡(8.7g,24mmol),反应混合物升温到回流,氮气氛下保温搅拌反应过夜。冷却到室温,加入乙酸乙酯(100mL)稀释反应液,加入氟化钾饱和水溶液(150mL),搅拌1h。滤除不溶性固体,分出有机相,水层乙酸乙酯萃取(80mLx2),合并有机相,无水硫酸钠干燥,过滤,减压蒸除溶剂,浓缩至干,直接用于下一步。
步骤2 化合物35的合成
向上述化合物34中加入四氢呋喃(20mL),搅拌溶清,加入水(15mL),冰水浴冷却,加入N-溴代丁二酰亚胺(NBS,3.6g,20mmol),室温搅拌反应1h。分出有机层,水层乙酸乙酯萃取(30mLx3),合并有机相,饱和食盐水洗涤(30mL),无水硫酸钠干燥,过滤,减压蒸除溶剂,残留物过硅胶柱得黄色固体2.4g,两步收率49.0%。LC-MS(APCI):m/z=247.0(M+1) +,UV254. 1H NMR(400MHz,CDCl 3)δ8.78(d,J=4.4Hz,1H),7.56(d,J=4.4Hz,1H),4.74(s,2H),2.62(s,3H).
步骤3 化合物36的合成
向配有磁力搅拌的100mL双口烧瓶中加入无水苯甲醇(5.4g,50mmol)和无水四氢呋喃(40mL),冰水浴冷却,加入氢化钠(2.3g,60mmol,60%),氮气氛下保温搅拌半小时。缓慢滴加入溴乙腈(6.0g,50mmol)的四氢呋喃溶液(15mL),滴毕,拆去冰浴,氮气氛下室温搅拌反应过夜。加入水(40mL)淬灭反应,加入乙酸乙酯(50mL),分出有机层,水层乙酸乙酯萃取(30mLx3),合并有机相,饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得淡黄色油状物4.0g,收率54.4%。LC-MS(APCI):m/z=148.0(M+1) +,UV254.
步骤4 化合物37的合成
向配有磁力搅拌及冷凝管的50mL双口烧瓶中加入化合物36(2.2g,15mmol)和无水甲苯(10mL),搅拌溶清,加入苯胺(1.67g,1.8mmol),抽真空并氮气置换3次,冰水浴冷却,氮气氛下缓慢滴加入三甲基铝溶液(11mL,17.6mmol,1.6M甲苯溶液),滴毕,拆去冰浴,升温回流搅拌反应2小时。冷却到室温,加入水(30mL)和乙酸乙酯(50mL)淬灭反应,滤除不溶性固体物质,乙酸乙酯洗涤(20mL),分出有机相,水相乙酸乙酯萃取(40mLx2),合并有机相,无水硫酸钠干燥,过滤,浓缩得棕色固体2.6g,收率72.2%。直接用于下一步。LC-MS(APCI):m/z=241.0(M+1) +,UV254.
步骤5 化合物38的合成
向配有磁力搅拌及冷凝管的50mL双口烧瓶中加入化合物37(845mg,3.52mmol)和异丙醇(10mL),搅拌溶清,加入化合物35(1.13g,4.57mmol)和碳酸氢钠(591mg,7.03mmol),反应液氮气氛下升温到回流,并保温搅拌反应2小时。冷却到室温,减压蒸除溶剂,残留物过硅胶柱得白色固体500mg,收率36.6%。LC-MS(APCI):m/z=389.1(M+1) +,UV254.
步骤6 化合物39的合成
向配有磁力搅拌的50mL单口烧瓶中加入化合物38(250mg,0.63mmol)和四氢呋喃(5mL),搅拌溶清,加入水(5mL),冰水浴下加入过硫酸氢钾(oxone,593mg,0.96mmol),反应液氮气氛下室温搅拌过夜。加入饱和硫代硫酸钠水液(10mL)淬灭反应,乙酸乙酯萃取(20mLx3),合并有机相,饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤,浓缩,过硅胶柱得白色固体255mg,收率96.1%。LC-MS(APCI):m/z=421.1(M+1) +,UV254. 1H NMR(400MHz,CDCl 3)δ8.84(d,J=5.6Hz,1H),8.17(d,J=5.6Hz,1H),8.14(s,1H),7.51-7.49(m,5H),7.34-7.28(m,5H),4.60(s,2H),4.54(s,2H),3.38(s,3H).
步骤7 化合物41的合成
向配有磁力搅拌、冷凝管的100mL单口瓶中依次加化合物40(9.3g,50mmol)和甲酸(50mL),混合物升温到回流,保温搅拌反应2小时。冷却到室温,减压蒸除未反应的甲酸,残留物过硅胶柱得白色固体7.8g,收率73%。LC-MS(APCI):m/z=215.1(M+1) +,UV 254.
步骤8 化合物42的合成
向配有磁力搅拌的100mL单口瓶中依次加化合物41(2.14g,10mmol)、DMF(25mL)、K 2CO 3(2.07g,15mmol)和吗啉(0.87g,10mmol),混合物氮气下室温搅拌反应过夜。加入乙酸乙酯(80mL),滤除不溶性固体,滤液浓缩,残留物过硅胶柱得黄的固体2.11g,收率70%。LC-MS(APCI):m/z=282.1(M+1) +,UV 254.
步骤9 化合物43的合成
向配有磁力搅拌的50mL双口瓶中加化合物42(140mg,0.5mmol)和无水DMF(2ml),冰水浴冷却下,加入NaH(60%,40mg,1.0mmol)。氮气下室温搅拌反应半小时,冰水浴冷却,缓慢滴加入化合物39(281mg,0.6mmol)的无水DMF(1ml)溶液,滴毕,室温搅拌反应3小时。冰水浴冷却,加入2M氢氧化钠水浴(10mL),保温搅拌反应半小时,析出大量黄色固体,过滤,水洗(10mL),并溶于二氯甲烷(20mL)中,无水硫酸钠干燥,浓缩并过硅胶柱得黄色固体240mg,收率81.2%。 LC-MS(APCI):m/z=594.2(M+1) +,UV 254.
步骤10 化合物44的合成
向配有磁力搅拌的25mL单口瓶中加化合物43(200mg,0.34mmol)和浓盐酸(3mL),氮气氛下升温到85摄氏度,保温搅拌反应1小时。冷却到室温,冰水浴冷却下缓慢滴加入2M氢氧化钠水液,调pH~9,二氯甲烷萃取(20mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得黄色固体140mg,收率82.5%。LC-MS(APCI):m/z=504.2(M+1) +,UV254. 1H NMR(300MHz,CDCl 3)δ9.66(s,1H),8.48(d,J=3.6Hz,1H),8.20(s,1H),7.67(s,1H),7.59-7.57(m,4H),7.52-7.50(m,1H),7.43-7.41(m,1H),6.60(s,1H),4.73(s,2H),4.01(s,3H),3.90-3.88(m,4H),3.08-3.06(m,4H).
步骤11 化合物45的合成
向配有磁力搅拌的50mL单口瓶中加化合物44(140mg,0.28mmol)和二氯甲烷(5mL),冰水浴下加入三乙胺(56mg,0.57mmol)和甲基磺酰氯(48mg,0.42mmol),加毕,拆去冰浴,室温搅拌反应1小时。减压浓缩至干,直接用于下一步。
步骤12 化合物46的合成
向上述包含化合物45浓缩液中加入乙腈(5mL),搅拌溶清,加入二甲胺的四氢呋喃溶液(4mL,2M),氮气氛下室温搅拌反应1小时。减压蒸除溶剂,残留物过硅胶柱得黄色固体90mg,两步收率65.8%。LC-MS(APCI):m/z=531.2(M+1) +,UV 254.
步骤13 化合物47的合成
向配有磁力搅拌的50mL单口瓶中加化合物46(90mg,0.16mmol)和乙醇(4mL),搅拌溶清,加入水(2mL),再加入还原铁粉(92mg,1.64mmol)和氯化铵(88mg,1.64mmol),氮气氛下升温到回流,并保温搅拌反应一个半小时。冷却到室温,减压蒸除有机溶剂,加入饱和碳酸氢钠水浴(5mL),二氯甲烷萃取(20mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩至干得棕色固体68mg,收率82.8%。LC-MS(APCI):m/z=501.2(M+1) +,UV 254.
步骤14 化合物T-5的合成
向配有磁力搅拌的25mL双口瓶中加化合物47(68mg,0.14mmol)和二氯甲烷(4mL),搅拌溶清,冷却到-10摄氏度,加入三乙胺(27mg,0.27mmol),缓慢滴加入丙烯酰氯(18mg,0.20mmol)的二氯甲烷(1mL)溶液,滴毕,保温搅拌反应1小时。加入饱和碳酸氢钠水液(5mL)淬灭反应,二氯甲烷萃取(10mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得淡黄色固体50mg,收率66.4%。LC-MS(APCI):m/z=555.3(M+1) +,UV254.1H NMR(300MHz,CDCl 3)δ9.60(s,1H),9.72(br s,1H),8.59(s,1H),8.42(d,J=3.6Hz,1H),7.78(s,1H),7.69(d,J=6.0Hz,1H),7.58(s,1H),7.52-7.42(m,3H),6.75(s,1H),6.18-6.11(m,1H),5.86-5.82(m,1H),5.53-5.51(m,1H),3.88(s,3H),3.87-3.81(m,4H),3.35(s,2H),2.86-2.84(m,4H),2.30(s,6H).
实施例6 N-(5-((4-(3-((二甲基氨基)甲基)-4-苯基-1H-吡唑-1-基)嘧啶-2-基)氨基)-4-甲氧基-2-吗啉 代苯基)丙烯酰胺(化合物T-6)的制备
Figure PCTCN2019122399-appb-000027
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000028
步骤1 化合物48的合成
向配有磁力搅拌的100mL三口瓶中依次加化合物42(2.81g,10mmol)和无水DMF(15ml),冷却到0℃,加入NaH(60%,480mg,12mmol)。氮气下室温搅拌反应半小时,然后冷却到0℃,缓慢滴加入化合物2(1.93g,10mmol)的干燥DMF(15ml)溶液,滴毕,室温搅拌反应3h。加入水(25mL)淬灭反应,搅拌2h,乙酸乙酯萃取(50mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩,残留物过硅胶柱得黄色固体2.83g,收率77%。LC-MS(APCI):m/z=366.1(M+1) +1H NMR(400MHz,CDCl 3) δ9.19(s,1H),8.36(d,J=5.2Hz,1H),7.70(s,1H),6.82(d,J=5.2Hz,1H),6.61(s,1H),4.00(s,3H),3.89(t,J=4.8Hz,4H,3.08(t,J=4.8Hz,4H).
步骤2 化合物50的合成
向配有磁力搅拌的20mL微波管中加入化合物49(2.0g,10mmol)、苯硼酸(1.22g,10mmol)和1,4-二氧六环(10mL),搅拌溶清,加入碳酸铯(4.87g,15mmol)、水(1mL)和四(三苯基膦)钯(1.15g,1.0mmol),抽真空并氮气置换3次,置于微波反应器中,升温到150℃,保温搅拌反应1小时。冷却到室温,加入乙酸乙酯(30mL),滤除不溶性固体,滤液减压浓缩,残留物过硅胶柱得黄色固体600mg,收率30.2%。LC-MS(APCI):m/z=203.1(M+1) +,UV 254.
步骤3 化合物51的合成
向配有磁力搅拌的50mL双口瓶中加化合物50(600mg,3.0mmol)和无水DMF(5ml),冰水浴冷却下,加入NaH(60%,160mg,4.0mmol)。氮气下室温搅拌反应半小时,冰水浴冷却,缓慢滴加入化合物48(1.1g,3.0mmol)的无水DMF(3ml)溶液,滴毕,室温搅拌反应3小时。加入水(40mL),析出大量黄色固体,过滤,水洗(10mL),并溶于二氯甲烷(40mL)中,无水硫酸钠干燥,浓缩并过硅胶柱得黄色固体600mg,收率37.7%。LC-MS(APCI):m/z=532.2(M+1) +,UV 254.
步骤4 化合物52的合成
向配有磁力搅拌的50mL单口瓶中加化合物51(530mg,1.0mmol)和无水THF(20ml),冰水浴冷却,缓慢加入氢化铝锂(38mg,1.0mmol),氮气氛下保温搅拌反应1h。加入十水硫酸钠(5g)淬灭反应,加入乙酸乙酯(20mL),滤除不溶性固体,减压浓缩并过硅胶柱得白色固体400mg,收率79.5%。LC-MS(APCI):m/z=504.2(M+1) +,UV254. 1H NMR(400MHz,CDCl 3)δ9.45(s,1H),8.89(s,1H),8.49(d,J=5.6Hz,1H),7.71(s,1H),7.66(d,J=8.0Hz,1H),7.49-7.45(m,2H),7.39(d,J=5.2Hz,2H),7.36-7.34(m,1H),6.60(s,1H),4.92(s,2H),4.01(s,3H),3.91-3.88(m,4H),3.17(br s,1H),3.10-3.06(m,4H).
步骤5 化合物53的合成
向配有磁力搅拌的50mL单口瓶中加化合物52(400mg,0.8mmol)和二氯甲烷(20mL),冰水浴下加入三乙胺(1.6g,1.6mmol)和甲基磺酰氯(MsCl,0.12g,1.0mmol),加毕,拆去冰浴,室温搅拌反应1小时。减压浓缩至干,直接用于下一步。
步骤6 化合物54的合成
向上述含化合物53的浓缩液中加入乙腈(10mL),搅拌溶清,加入二甲胺的四氢呋喃溶液(10mL,2M),氮气氛下室温搅拌反应1小时。减压蒸除溶剂,残留物过硅胶柱得黄色固体320mg,两步收率75.7%。LC-MS(APCI):m/z=531.2(M+1) +,UV 254.
步骤7 化合物55的合成
向配有磁力搅拌的50mL单口瓶中加化合物54(320mg,0.6mmol)和乙醇(10mL),搅拌溶清,加入水(5mL),再加入还原铁粉(0.34g,6.0mmol)和氯化铵(0.32g,6.0mmol),氮气氛下升温到回流,并保温搅拌反应一个半小时。冷却到室温,减压蒸除有机溶剂,加入饱和碳酸氢钠水浴(10mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩至干得棕色固体200mg,收率66.7%。LC-MS(APCI):m/z=501.2(M+1) +,UV 254.
步骤8 化合物T-6的合成
向配有磁力搅拌的25mL双口瓶中加化合物55(200mg,0.4mmol)和二氯甲烷(10mL),搅拌溶清,冷却到-10摄氏度,加入三乙胺(80mg,0.8mmol),缓慢滴加入丙烯酰氯(45mg,0.50mmol)的二氯甲烷(2mL)溶液,滴毕,保温搅拌反应1小时。加入饱和碳酸氢钠水液(10mL)淬灭反应,二氯甲烷萃取(20mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得淡黄色固体130mg,收率58.5%。LC-MS(APCI):m/z=555.3(M+1) +,UV254.1H NMR(400MHz,CDCl 3)δ9.76(s,1H),9.48(br s,1H),8.65(s,1H),8.46(d,J=4.8Hz,1H),7.89(s,1H),7.69(d,J=7.2Hz,1H),7.43-7.36(m,3H),7.33-7.31(m,1H),6.78(s,1H),6.25-6.21(m,1H),6.15-6.11(m,1H),5.68-5.66(m,1H),4.00(s,2H),3.91(s,3H),3.88-3.86(m,4H),2.88-2.86(m,4H),2.49(s,6H).
实施例7 N-(5-((4-(5-((二甲基氨基)甲基)-1-苯基-1H-吡唑-3-基)嘧啶-2-基)氨基)-4-甲氧基-2-吗啉 代苯基)丙烯酰胺(化合物T-7)的制备
Figure PCTCN2019122399-appb-000029
采用以下路线进行合成:
Figure PCTCN2019122399-appb-000030
Figure PCTCN2019122399-appb-000031
步骤1 化合物56的合成
向配有磁力搅拌的50mL单口瓶中加化合物34(2.0g,10mmol)和THF(20mL),搅拌溶清,加入4M盐酸(20mL),氮气氛下搅拌反应1小时。减压蒸除有机溶剂,乙酸乙酯萃取(30mLx3),饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得白色固体1.5g,收率89.2%。LC-MS(APCI):m/z=169.1(M+1) +,UV 254.
步骤2 化合物57的合成
向配有磁力搅拌的50mL双口瓶中加化合物56(1.5g,8.9mmol)和无水THF(10mL),搅拌溶清,加入化合物57(1.3g,8.9mmol),冰水浴冷却,滴加入叔丁醇钾的THF溶液(9.8mL,9.8mmol,1M),滴毕,拆去冰浴,氮气氛下室温搅拌反应过夜。加入水(20mL)和乙酸乙酯(40mL)淬灭反应,分出有机相,乙酸乙酯萃取水相(30mLx2),合并有机相,饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤,浓缩至干得棕色油状物2.3g,收率96.4%,直接用于下一步。LC-MS(APCI):m/z=269.1(M+1) +,UV 254.
步骤3 化合物58的合成
向配有磁力搅拌和冷凝管的50mL单口瓶中加化合物57(2.3g,8.6mmol)和乙醇(30mL),搅拌溶清,加入醋酸钠(2.33g,17.2mmol)和冰醋酸(1.0g,17.2mmol),冰水浴冷却,加入苯肼(0.83g,0.76mmol),加毕,拆去冰浴,升温回流反应2小时。冷却到室温,减压蒸除溶剂,残留物过硅胶柱得黄色固体1.5g,收率51.3%。LC-MS(APCI):m/z=341.1(M+1) +,UV 254.
步骤4 化合物59的合成
向配有磁力搅拌的50mL单口瓶中加化合物58(1.5g,4.4mmol)和无水THF(30mL),搅拌溶清,冰水浴冷却,加入氢化铝锂(0.17g,4.4mmol),氮气氛下保温搅拌反应1小时。加入十水硫酸钠(5g)淬灭反应,再加入乙酸乙酯(20mL),滤除不溶性固体,减压浓缩并过硅胶柱得白色固体1.0g,收率76.2%。LC-MS(APCI):m/z=299.1(M+1) +,UV254.1H NMR(400MHz,CDCl 3)δ8.43(d,J=5.2Hz,1H),7.45-7.39(m,3H),7.33-7.31(m,2H),6.95(s,1H),6.90(d,J=5.2Hz,1H),4.81(s,2H),2.08(s,3H).
步骤5 化合物60的合成
向配有磁力搅拌的50mL单口瓶中加化合物59(600mg,2.0mmol)和二氯甲烷(20mL),冰水浴下加入三乙胺(400mg,4.0mmol)和甲基磺酰氯(MsCl,0.27g,2.4mmol),加毕,拆去冰浴,室温搅拌反应1小时。减压浓缩至干,直接用于下一步。
步骤6 化合物61的合成
向上述浓缩物中加入乙腈(20mL),搅拌溶清,加入二甲胺的四氢呋喃溶液(20mL,2M),氮气氛下室温搅拌反应1小时。减压蒸除溶剂,残留物过硅胶柱得黄色固体520mg,两步收率80.0%。LC-MS(APCI):m/z=326.2(M+1) +,UV 254. 1H NMR(400MHz,DMSO-d 6)δ8.63(d,J=5.6Hz,1H),7.48-7.46(m,3H),7.38-7.34(m,3H),7.20(s,1H),3.86(s,2H),2.46(s,6H),1.90(s,3H).
步骤7 化合物62的合成
向配有磁力搅拌的50mL单口烧瓶中加入化合物61(520mg,1.6mmol)和四氢呋喃(10mL),搅拌溶清,加入水(10mL),冰水浴下加入过硫酸氢钾(oxone,1.0g,2.4mmol),反应液氮气氛下室温搅拌过夜。加入饱和硫代硫酸钠水液(20mL)淬灭反应,乙酸乙酯萃取(30mLx3),合并有机相,饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤,浓缩,过硅胶柱得白色固体500mg,收率87.5%。LC-MS(APCI):m/z=358.1(M+1) +,UV 254.
步骤8 化合物63的合成
向配有磁力搅拌的50mL双口瓶中加化合物42(140mg,0.5mmol)和无水DMF(2ml),冰水浴冷却下,加入NaH(60%,40mg,1.0mmol)。氮气下室温搅拌反应半小时,冰水浴冷却,缓慢滴加入化合物62(214mg,0.6mmol)的无水DMF(1ml)溶液,滴毕,室温搅拌反应3小时。冰水浴冷却,加入2M氢氧化钠水浴(10mL),保温搅拌反应半小时,析出大量黄色固体,过滤,水洗(10mL),并溶于二氯甲烷(20mL)中,无水硫酸钠干燥,浓缩并过硅胶柱得黄色固体200mg,收率75.3%。LC-MS(APCI):m/z=531.2(M+1) +,UV254.
步骤9 化合物64的合成
向配有磁力搅拌的50mL单口瓶中加化合物63(200mg,0.38mmol)和乙醇(10mL),搅拌溶清,加入水(5mL),再加入还原铁粉(211mg,3.8mmol)和氯化铵(205mg,3.8mmol),氮气氛下升温到回流,并保温搅拌反应一个半小时。冷却到室温,减压蒸除有机溶剂,加入饱和碳酸氢钠水浴(10mL),二氯甲烷萃取(30mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩至干得棕色固体150mg,收率78.9%。LC-MS(APCI):m/z=501.2(M+1) +,UV254.
步骤10 化合物T-7的合成
向配有磁力搅拌的25mL双口瓶中加化合物64(150mg,0.3mmol)和二氯甲烷(10mL),搅拌溶清,冷却到-10摄氏度,加入三乙胺(60mg,0.6mmol),缓慢滴加入丙烯酰氯(36mg,0.40mmol)的二氯甲烷(2mL)溶液,滴毕,保温搅拌反应1小时。加入饱和碳酸氢钠水液(10mL)淬灭反应,二氯甲烷萃取(20mLx3),合并有机相,无水硫酸钠干燥,过滤,浓缩并过硅胶柱得淡黄色固体90mg,收率54.0%。LC-MS(APCI):m/z=555.3(M+1) +,UV254.1H NMR(400MHz,CDCl 3)δ9.56(s,1H),8.67(br s,1H),8.26(d,J=5.2Hz,1H),7.72(br s,1H),7.67(s,1H),7.47-7.45(m,3H),7.42-7.38(m,2H),6.78(s,1H),6.45-6.41(m,1H),6.35-6.25(m,2H),5.84-5.81(m,1H),4.00(s,2H),3.90-3.86(m,7H), 3.90-3.88(m,4H),2.61(s,6H).
生物活性测试
生物实施例1:激酶抑制作用
试剂和耗材:
WT EGFR(Carna,目录号08-115),EGFR[L858R](Carna,目录号08-502),EGFR[L858R/T790M](Carna,目录号08-510),ATP(Sigma,目录号A7699-1G),DMSO(Sigma,目录号D2650),96孔板(Corning,目录号3365),384孔板(Greiner,目录号784076),HTRF Kinase TK试剂盒(Cisbio,目录号62TK0PEJ),厄洛替尼(Selleckchem,目录号S7787),EGFR[d746-750](Life Technologies,目录号PV6178),5x激酶缓冲液A(Life Technologies,目录号PV3186),激酶示踪剂199(Life Technologies,目录号PV5830),
Figure PCTCN2019122399-appb-000032
Eu-anti-GST抗体(Life Technologies,目录号PV5594)。
具体实验方法:
化合物配制:将受试化合物溶于DMSO配成20mM母液。然后,在DMSO中等梯度3倍稀释,稀释十次。加药时再用缓冲液稀释10倍。
WT EGFR及EGFR[L858R/T790M]激酶检测:在5x激酶缓冲液A中,将WT EGFR或EGFR[L858R/T790M]激酶与预先稀释配制的不同浓度的化合物混合10分钟,每个浓度双复孔。加入对应底物及ATP,室温反应20分钟(其中设置阴阳性对照:阴性为空白对照,阳性为厄洛替尼)。反应完毕加入检测试剂(HTRF Kinase TK试剂盒内的试剂),室温孵育30分钟后,通过Evnvision酶标仪检测,测定在各浓度的本发明化合物存在下的酶活力,并计算不同浓度的化合物对酶活力的抑制活性,之后根据四参数方程,根据Graphpad 5.0软件对不同浓度化合物下酶活力的抑制活性进行拟合,计算出IC 50值。
在上述激酶抑制实验中测试了本发明化合物,发现本发明化合物对EGFR[L858R/T790M]具有强效的活性以及优于WT EGFR的优异选择性。代表性实施例化合物的结果归纳于如下表1中。
表1
实施例化合物 EGFR(WT)IC 50(nM) L858R/T790M IC 50(nM)
T-1 1.29 0.40
T-2 3.33 0.44
T-3 6.29 0.60
T-4 1.98 0.81
T-7 0.55 1.15
生物实施例2:细胞毒性实验
采用MTS方法检测了本发明化合物对体外培养的3株肿瘤细胞的体外抗增殖活性。实验结果表明本发明化合物对体外培养的癌细胞的体外增殖具有抑制作用;其中对肺癌细胞的体外增殖的抑制作用比皮肤癌细胞的体外增殖的抑制作用强。
细胞系:皮肤癌细胞A431(购自美国标准生物品收藏中心(ATCC));肺癌细胞NCI-H1975(购自美国标准生物品收藏中心(ATCC))和HCC827(购自美国标准生物品收藏中心(ATCC));均用含10%胎牛血清、100U/ml青霉素、100μg/ml链霉素的RPMI1640培养基培养。
试剂和耗材:RPMI-1640(GIBCO,目录号A10491-01);胎牛血清(GIBCO,目录号10099141);0.25%胰蛋白酶-EDTA(GIBCO,目录号25200);青霉素-链霉素;液体(GIBCO,目录号15140-122);DMSO(Sigma,目录号D2650);MTS测试试剂盒(Promega,目录号G3581),96孔板(Corning,目录号3365)。
具体实验方法:
化合物配制:受试化合物溶于DMSO配成20mM母液,-20℃保存。用DMSO等梯度3倍稀释,稀释10倍。加药时再用细胞培养基稀释4倍。
MTS细胞活力检测:0.25%胰蛋白酶-EDTA消化对数生长期细胞,按已优化的密度接种150μl于96孔板,24小时后加入培养基稀释4倍的化合物,50μl/孔(一般选择十个浓度:100、33.3、11.1、3.70、1.23、0.412、0.137、0.0457、0.0152、0.00508μM)。以加入同样体积的0.5%DMSO的孔作为对照。细胞继续培养72小时后,MTS检测细胞活力。
具体操作:贴壁细胞,弃去培养基,每孔加入含20μL MTS和100μl培养基的混合液。放入培养箱继续培养1-4小时后检测OD490,以OD650值作为参考。用GraphPad Prism软件制作量效曲线并计算IC 50
在上述细胞毒性实验中测试了本发明化合物,发现本发明化合物对肺癌细胞NCI-H1975和HCC827具有强效的活性以及优于皮肤癌细胞A431的优异选择性。代表性实施例对癌细胞的体外增殖的抑制作用的结果归纳于下表2中。
表2
Figure PCTCN2019122399-appb-000033
生物实施例3:大鼠药代动力学实验
6只雄性Sprague-Dawley大鼠,7-8周龄,体重约210g,分成2组,每组3只,经静脉或口服单个剂量的化合物(口服10mg/kg),比较其药代动力学差异。
大鼠采用标准饲料饲养,给予水。试验前16小时开始禁食。药物用PEG400和二甲亚砜溶解。眼眶采血,采血的时间点为给药后0.083小时,0.25小时、0.5小时、1小时、2小时、4小时、6小时、8小时、12小时和24小时。
大鼠吸入乙醚后短暂麻醉,眼眶采集300μL血样于试管。试管内有30μL 1%肝素盐溶液。使用前,试管于60℃烘干过夜。在最后一个时间点血样采集完成之后,大鼠乙醚麻醉后处死。
血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血样在4℃ 5000rpm离心5分钟,将血浆与红细胞分离。用移液器吸出100μL血浆到干净的塑料离心管中,标明化合物的名称和时间点。血浆在进行分析前保存在-80℃。用LC-MS/MS测定血浆中本发明化合物的浓度。 药代动力学参数基于每只动物在不同时间点的血药浓度进计算。
实验表明,本发明化合物在动物体内具有更好的药代动力学性质,因此具有更好的药效学和治疗效果。
表3
Figure PCTCN2019122399-appb-000034
上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明。

Claims (21)

  1. 式(I)化合物:
    Figure PCTCN2019122399-appb-100001
    其中,
    X选自CH、CD或N;
    环A选自至少含有一个N原子的五元杂芳环;
    R 1选自H、D、卤素、-CN、-NO 2、-OH、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基或-OC 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-OC 3-7环烷基任选地被1-13个R 9基团取代;
    R2选自H、D、4-7元杂环烷基或-NR 7R 8,其中所述的4-7元杂环烷基任选地被1-10个R 9基团取代;
    R 3选自H、D、卤素、-CN、-NO 2、C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基任选地被1-13个R 9基团取代;
    R 4选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基,其中所述的C 6-10芳基和5-10元杂芳基任选地被1-8个R 9基团取代;
    R 5选自H、D、-(CH 2) nOR 7、-(CH 2) nNR 7R 8、-(CD 2) nOR 7或-(CD 2) nNR 7R 8,其中n选自1、2、3或4;
    R 6选自H、D或C 1-6烷基,其中所述的C 1-6烷基任选地被1-13个R 9基团取代;
    R 7和R 8各自独立地选自H、D、C 1-6烷基、C 3-7环烷基或4-7元杂环烷基,或者R 7、R 8与它们所连接的N原子一起形成4-7元杂环烷基;其中所述的C 1-6烷基、C 3-7环烷基或4-7元杂环烷基任选地被1-13个R 9基团取代;
    R 9独立地选自H、D、卤素、-OH、C 1-6烷氧基、-NH 2、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、-C(O)C 1-6烷基、-C(O)OC 1-6烷基、-C(O)NHC 1-6烷基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;或者相同原子或相邻原子上的两个R 9基团可以一起形成C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;其中R 9定义中的每个基团任选地被一个或多个D取代,直至完全氘代;
    前提是,当X为CH,且环A为
    Figure PCTCN2019122399-appb-100002
    时,R 1选自C 1-6卤代烷基或C 1-6卤代烷氧基;
    或其药学上可接受的盐、立体异构体、溶剂合物或水合物。
  2. 根据权利要求1所述的化合物,其为式(II)化合物:
    Figure PCTCN2019122399-appb-100003
    其中,
    X选自CH、CD或N;
    Y 1、Y 2、Y 3、Y 4所在的环为芳香环,并且它们独立地选自C、N、O或S原子;
    R 1选自H、D、卤素、-CN、-NO 2、-OH、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基或-OC 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-OC 3-7环烷基任选地被1-13个R 9基团取代;
    R 2选自H、D、4-7元杂环烷基或-NR 7R 8,其中所述的4-7元杂环烷基任选地被1-10个R 9基团取代;
    R 3选自H、D、卤素、-CN、-NO 2、C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基或C 3-6环烷基任选地被1-13个R 9基团取代;
    R 4选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基,其中所述的C 6-10芳基和5-10元杂芳基任选地被1-8个R 9基团取代;
    R 5选自H、D、-(CH 2) nOR 7、-(CH 2) nNR 7R 8、-(CD 2) nOR 7或-(CD 2) nNR 7R 8,其中n选自1、2、3或4;
    R 6选自H、D或C 1-6烷基,其中所述的C 1-6烷基任选地被1-13个R 9基团取代;
    R 7和R 8各自独立地选自H、D、C 1-6烷基、C 3-7环烷基或4-7元杂环烷基,或者R 7、R 8与它们所连接的N原子一起形成4-7元杂环烷基;其中所述的C 1-6烷基、C 3-7环烷基或4-7元杂环烷基任选地被1-13个R 9基团取代;
    R 9独立地选自H、D、卤素、-OH、C 1-6烷氧基、-NH 2、-NH(C 1-6烷基)、-N(C 1-6烷基) 2、-C(O)C 1-6烷基、-C(O)OC 1-6烷基、-C(O)NHC 1-6烷基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;或者相同原子或相邻原子上的两个R 9基团可以一起形成C 3-7环烷基、4-7元杂环烷基、C 6-10芳基或5-10元杂芳基;其中R 9定义中的每个基团任选地被一个或多个D取代,直至完全氘代;
    前提是,当X为CH,且环A为
    Figure PCTCN2019122399-appb-100004
    时,R 1选自C 1-6卤代烷基或C 1-6卤代烷氧基;
    或其药学上可接受的盐、立体异构体、溶剂合物或水合物。
  3. 根据权利要求2所述的化合物,其中:
    Figure PCTCN2019122399-appb-100005
    选自:
    Figure PCTCN2019122399-appb-100006
    Figure PCTCN2019122399-appb-100007
  4. 根据权利要求1-3中任一项所述的化合物,其为式(II-1)化合物:
    Figure PCTCN2019122399-appb-100008
    其中,
    R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个R 9基团取代;
    X、Y 1-Y 4、R 3-R 6和R 9如权利要求1所定义。
  5. 根据权利要求1-4中任一项所述的化合物,其为式(II-2)化合物:
    Figure PCTCN2019122399-appb-100009
    其中,
    R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个R 9基团取代;
    X、Y 1-Y 4、R 3和R 9如权利要求1所定义。
  6. 根据权利要求1-5中任一项所述的化合物,其为式(III)或(IV)化合物:
    Figure PCTCN2019122399-appb-100010
    其中,R 1-R 6如权利要求1所定义。
  7. 根据权利要求6所述的化合物,其为式(III-1)或(IV-1)化合物:
    Figure PCTCN2019122399-appb-100011
    其中,
    R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个R 9基团取代;
    R 3-R 6和R 9如权利要求1所定义。
  8. 根据权利要求7所述的化合物,其为式(III-2)或(IV-2)化合物:
    Figure PCTCN2019122399-appb-100012
    其中,
    R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个R 9基团取代;
    R 3选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基,其中所述的C 1-6烷基和C 1-6卤代烷基任选地被1-13个R 9基团取代;
    R 9如权利要求1所定义。
  9. 根据权利要求1-5中任一项所述的化合物,其为式(V)化合物:
    Figure PCTCN2019122399-appb-100013
    其中,X和R 1-R 6如权利要求1所定义。
  10. 根据权利要求9所述的化合物,其为式(V-1)化合物:
    Figure PCTCN2019122399-appb-100014
    其中,
    R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个R 9基团取代;
    X、R 3-R 6和R 9如权利要求1所定义。
  11. 根据权利要求9所述的化合物,其为式(V-2)化合物:
    Figure PCTCN2019122399-appb-100015
    其中,
    R选自H、C 1-6烷基、C 1-6卤代烷基或C 3-7环烷基,其中所述的C 1-6烷基、C 1-6卤代烷基和C 3-7环烷基任选地被1-13个R 9基团取代;
    X选自CH、CD或N;
    R 3选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基,其中所述的C 1-6烷基和C 1-6卤代烷基任选地被1-13个R 9基团取代;
    R 9如权利要求1所定义。
  12. 根据权利要求1-5中任一项所述的化合物,其为式(VI)或(VII)化合物:
    Figure PCTCN2019122399-appb-100016
    其中,
    R 1选自C 1-6卤代烷基或C 1-6卤代烷氧基,其中所述的C 1-6卤代烷基和C 1-6卤代烷氧基任选地被1-12个R 9基团取代;
    R 2-R 6和R 9如权利要求1所定义。
  13. 根据权利要求12所述的化合物,其为式(VI-1)或(VII-1)化合物:
    Figure PCTCN2019122399-appb-100017
    其中,
    R选自C 1-6卤代烷基,其中所述的C 1-6卤代烷基任选地被1-12个R 9基团取代;
    R 3-R 6和R 9如权利要求1所定义。
  14. 根据权利要求12所述的化合物,其为式(VI-2)或(VII-2)化合物:
    Figure PCTCN2019122399-appb-100018
    其中,
    R选自C 1-6卤代烷基,其中所述的C 1-6卤代烷基任选地被1-12个R 9基团取代;
    R 3选自H、D、卤素、C 1-6烷基或C 1-6卤代烷基,其中所述的C 1-6烷基和C 1-6卤代烷基任选地被1-13个R 9基团取代;
    R 9如权利要求1所定义。
  15. 化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,所述化合物选自:
    Figure PCTCN2019122399-appb-100019
    Figure PCTCN2019122399-appb-100020
  16. 药物组合物,其含有权利要求1-15中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,和药学上可接受的赋形剂。
  17. 如权利要求1-15中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,或如权利要求16的药物组合物在制备用于治疗蛋白激酶介导的疾病的药物中的用途。
  18. 如权利要求1-15中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,或如权利要求16的药物组合物,其用于治疗蛋白激酶介导的疾病。
  19. 一种治疗受试者中蛋白激酶介导的疾病的方法,包括向所述受试者给药如权利要求1-15中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物或水合物,或如权利要求16的药物组合物。
  20. 根据权利要求17的用途或权利要求18的化合物或药物组合物或权利要求19的方法,其中所述的蛋白激酶选自野生型和/或突变型的EGFR或野生型和/或突变型的JAK3。
  21. 根据权利要求20所述的用途或化合物或药物组合物或方法,其中,所述的突变型的EGFR选自del19、L858R、T790M、del19/T790M或L858R/T790M。
PCT/CN2019/122399 2018-12-21 2019-12-02 用于抑制蛋白激酶活性的氨基嘧啶类化合物 WO2020125391A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP19899282.8A EP3885346A4 (en) 2018-12-21 2019-12-02 AMINOPYRIMIDINE COMPOUND USED TO INHIBIT PROTEIN KINASE ACTIVITY
US17/415,492 US20220048891A1 (en) 2018-12-21 2019-12-02 Aminopyrimidine compound used for inhibiting activity of protein kinase
JP2021536036A JP7240032B2 (ja) 2018-12-21 2019-12-02 プロテインキナーゼ活性を阻害するためのアミノピリミジン系化合物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201811571875.1 2018-12-21
CN201811571875 2018-12-21

Publications (1)

Publication Number Publication Date
WO2020125391A1 true WO2020125391A1 (zh) 2020-06-25

Family

ID=71102512

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/122399 WO2020125391A1 (zh) 2018-12-21 2019-12-02 用于抑制蛋白激酶活性的氨基嘧啶类化合物

Country Status (5)

Country Link
US (1) US20220048891A1 (zh)
EP (1) EP3885346A4 (zh)
JP (1) JP7240032B2 (zh)
CN (1) CN111349084B (zh)
WO (1) WO2020125391A1 (zh)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CN104788427A (zh) * 2015-02-05 2015-07-22 上海泓博智源医药技术有限公司 3-(2-嘧啶氨基)苯基丙烯酰胺类化合物及其应用
CN104860941A (zh) * 2014-02-25 2015-08-26 上海海雁医药科技有限公司 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
CN105315259A (zh) * 2014-07-29 2016-02-10 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物、其制备方法及应用
WO2017016463A1 (zh) * 2015-07-24 2017-02-02 上海海雁医药科技有限公司 Egfr抑制剂及其药学上可接受的盐和多晶型物及其应用
CN106795144A (zh) * 2014-10-13 2017-05-31 株式会社柳韩洋行 用于调节egfr突变体激酶活性的化合物和组合物
WO2018194356A1 (en) * 2017-04-21 2018-10-25 Yuhan Corporation Salt of an aminopyridine derivative compound, a crystalline form thereof, and a process for preparing the same
CN108947974A (zh) * 2017-08-30 2018-12-07 深圳市塔吉瑞生物医药有限公司 一种氨基嘧啶类化合物及包含该化合物的组合物及其用途

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6740452B2 (ja) * 2016-07-26 2020-08-12 深▲セン▼市塔吉瑞生物医▲薬▼有限公司Shenzhen TargetRx,Inc. プロテインチロシンキナーゼの活性を阻害するためのアミノピリミジン系化合物

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CN104860941A (zh) * 2014-02-25 2015-08-26 上海海雁医药科技有限公司 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2015127872A1 (zh) * 2014-02-25 2015-09-03 上海海雁医药科技有限公司 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
CN105315259A (zh) * 2014-07-29 2016-02-10 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物、其制备方法及应用
CN106795144A (zh) * 2014-10-13 2017-05-31 株式会社柳韩洋行 用于调节egfr突变体激酶活性的化合物和组合物
CN104788427A (zh) * 2015-02-05 2015-07-22 上海泓博智源医药技术有限公司 3-(2-嘧啶氨基)苯基丙烯酰胺类化合物及其应用
WO2017016463A1 (zh) * 2015-07-24 2017-02-02 上海海雁医药科技有限公司 Egfr抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2018194356A1 (en) * 2017-04-21 2018-10-25 Yuhan Corporation Salt of an aminopyridine derivative compound, a crystalline form thereof, and a process for preparing the same
CN108947974A (zh) * 2017-08-30 2018-12-07 深圳市塔吉瑞生物医药有限公司 一种氨基嘧啶类化合物及包含该化合物的组合物及其用途

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BERGE ET AL.: "pharmaceutically acceptable salts", J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
D. FLEISHERS. RAMONH. BARBRA: "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", ADVANCED DRUG DELIVERY REVIEWS, vol. 19, no. 2, 1996, pages 115 - 130
See also references of EP3885346A4
T. HIGUCHIV. STELLA: "Bioreversible Carriers in Drug Design", vol. 14, 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS, article "Prodrugs as Novel Delivery Systems, A.C.S. Symposium Series"

Also Published As

Publication number Publication date
EP3885346A1 (en) 2021-09-29
CN111349084B (zh) 2022-11-25
US20220048891A1 (en) 2022-02-17
JP2022515196A (ja) 2022-02-17
CN111349084A (zh) 2020-06-30
JP7240032B2 (ja) 2023-03-15
EP3885346A4 (en) 2022-01-12

Similar Documents

Publication Publication Date Title
WO2021219090A1 (zh) 喹喔啉二酮衍生物作为kras g12c突变蛋白的不可逆抑制剂
CN109970745B (zh) 取代的吡咯并三嗪类化合物及其药物组合物及其用途
CN110724139B (zh) 用于抑制蛋白激酶活性的(杂)芳基酰胺类化合物
CN112047938B (zh) 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
WO2020114388A1 (zh) 取代的吡唑并[1,5-a]吡啶化合物及包含该化合物的组合物及其用途
CN109627263B (zh) 用于抑制激酶活性的二苯氨基嘧啶类化合物
CN109851638B (zh) 取代的二氨基嘧啶化合物
WO2019201131A1 (zh) 用于抑制蛋白激酶活性的二(杂)芳基大环化合物
WO2021185256A1 (zh) 取代的嘧啶或吡啶胺衍生物、其组合物及医药上的用途
WO2023134266A1 (zh) 2-哌啶基或2-吡唑基取代的嘧啶化合物作为egfr抑制剂
WO2023109751A1 (zh) 嘧啶或吡啶类衍生物及其医药用途
CN109111439B (zh) 一种酰胺类化合物及包含该化合物的组合物及其用途
CN110343092B (zh) 用于抑制蛋白激酶活性的(杂)芳基酰胺类化合物
WO2019228330A1 (zh) 取代的苯并[d]咪唑类化合物及其药物组合物
WO2019120094A1 (zh) 用于抑制激酶活性的芳基磷氧化物
WO2020125391A1 (zh) 用于抑制蛋白激酶活性的氨基嘧啶类化合物
WO2021057796A1 (zh) 取代的稠合三环衍生物及其组合物及用途
WO2017097113A1 (zh) 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
WO2022223020A1 (zh) 靶向活化与失活态kras g12d的抑制剂
WO2021164697A1 (zh) 取代的酰胺衍生物及其组合物及用途
WO2022166916A1 (zh) 取代的杂芳基衍生物及其组合物及用途
WO2023198180A1 (zh) Egfr降解剂
WO2023207556A1 (zh) Prmt5-mta抑制剂
JP2024504532A (ja) カモスタット及びニクロサミドを含む共結晶、それを含む薬学組成物、及び、その製造方法
WO2023216910A1 (zh) 取代的双环杂芳基化合物作为usp1抑制剂

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: 19899282

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021536036

Country of ref document: JP

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: 2019899282

Country of ref document: EP

Effective date: 20210621