WO2018006801A1 - 孤儿核受体Nur77的配体及其用途 - Google Patents

孤儿核受体Nur77的配体及其用途 Download PDF

Info

Publication number
WO2018006801A1
WO2018006801A1 PCT/CN2017/091716 CN2017091716W WO2018006801A1 WO 2018006801 A1 WO2018006801 A1 WO 2018006801A1 CN 2017091716 W CN2017091716 W CN 2017091716W WO 2018006801 A1 WO2018006801 A1 WO 2018006801A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cell
alkyl
nur77
cancer cell
Prior art date
Application number
PCT/CN2017/091716
Other languages
English (en)
French (fr)
Inventor
张晓坤
林祥志
苏迎
曾志平
胡梦婕
罗强
朱怡
古丽米然阿里同别克
Original Assignee
厦门大学
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 厦门大学 filed Critical 厦门大学
Priority to CN201780002455.9A priority Critical patent/CN108026142B/zh
Publication of WO2018006801A1 publication Critical patent/WO2018006801A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • C07J63/008Expansion of ring D by one atom, e.g. D homo steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)

Definitions

  • This application relates to the field of medicine and biology.
  • the present application relates to a novel ligand for Nur77 and its use.
  • the present application also relates to the use of a compound of formula I-formula V as a ligand for the orphan nuclear receptor Nur77.
  • the present application also relates to the use of a compound of formula I-formula V for the prevention or treatment of an inflammatory disease associated with the orphan nuclear receptor Nur77.
  • the present application also relates to a screening method for a medicament having anti-inflammatory and/or anti-obesity activity.
  • the orphan nuclear receptor Nur77 also known as NGFIB (nerve growth factor IB) or the orphan nuclear receptor TR3, is a key regulator of the development of cancer, metabolism and inflammatory diseases.
  • NGFIB ner growth factor IB
  • TR3 tumor growth factor 3
  • cytokinins hormones, stress, metabolism, and apoptotic signals.
  • Nur77 has been shown to be abnormally expressed in inflamed human synovial tissue, cancer cells, psoriasis patients, atherosclerotic patients, multiple sclerosis patients, and its expression can be rapidly and efficiently induced by some cytokines. Genetic studies have revealed a key role for Nur77 in controlling inflammatory responses, particularly in atherosclerosis (Hamers, AA, et al. Bone marrow-specific deficiency of nuclear receptor Nur77 enhances atherosclerosis. Circulation research 110, 428-438, 2012; Hanna, RN, et al.
  • NR4A1 (Nur77) deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis. Circulation research 110, 416-427, 2012), obesity (Perez-Sieira, S., et al.Female Nur77 -deficient mice show increased susceptibility to diet-induced obesity. PloS one 8, e53836, 2013), diabetes (Chao, LC, et al. Insulin resistance and altered systemic glucose metabolism in mice lacking Nur77. Diabetes 58, 2788-2796, 2009 ) Surge (Kurakula, K., et al.
  • Nuclear Receptor Nur77 Attenuates Airway Inflammation in Mice by Suppressing NF-kappaB Activity in Lung Epithelial Cells. Journal of immunology 195, 1388-1398, 2015), Arthritis (De Silva, S., Et al.Reduction of the incidence and severity of collagen-induced arthritis by constitutive Nur77 expression in the T cell lineage.Arthritis and rheumatism 52,333-338,2005) and inflammatory bowel disease (Hamers, AA, et al.Deficiency of Nuclear Receptor Nur77 Aggravates Mouse Experimental Colitis by Increased NFkappaB Activity in Macrophages.PloS one 10,e0133598,2015;Wu,H.,et al.NUR77 exerts a protective effect against inflammatory bowel disease by negatively regulating the TRAF6/TLR-IL-1R Signalling axis. The Journal of pathology 238, 457-469, 2016) The role of patients.
  • Nur77 in a variety of cellular processes and a variety of disease processes (eg, inflammation (such as inflammation associated with atherosclerosis, inflammation associated with obesity, and inflammation associated with diabetes) and cancer (such as triple-negative breast cancer)
  • inflammation such as inflammation associated with atherosclerosis, inflammation associated with obesity, and inflammation associated with diabetes
  • cancer such as triple-negative breast cancer
  • the inventors of the present application found that certain compounds are capable of inhibiting inflammation and autophagy by binding to Nur77 and inducing Nur77-dependent inhibition.
  • the compound interacts with tumor necrosis factor receptor-associated factor 2 (TRAF2, an important scaffold protein in the inflammatory signaling pathway and a key ubiquitin ligase) by facilitating the transport of Nur77 from the nucleus to the mitochondria.
  • TRAF2 tumor necrosis factor receptor-associated factor 2
  • This interaction not only inhibits ubiquitination of TRAF2 but also induces ubiquitination of Nur77 at K63.
  • ubiquitinated Nur77 interacts with the p62/SQSTM1 ubiquitin assembly domain on the mitochondria to enhance mitochondrial autophagy sensitivity.
  • LC3 an autophagy-related protein, specifically interacts with the modified Nur77 to ensure that the damaged mitochondria are selectively cleared.
  • the inventors of the present application established a method for screening Nur77-dependent compounds that inhibit inflammation and induce autophagy by the mechanism set forth above, and analyzed the biological activities of the selected compounds.
  • terpenoids and steroidal compounds have good antitumor and immunomodulatory activities.
  • the inventors of the present application have discovered through a large number of experimental studies a new class of pentacyclic triterpenoids YXY101 and its derivatives, which are capable of binding Nur77 to regulate mitochondrial activity and exert Nur77-dependent inhibition of inflammation and promotion of autophagy (especially It is the role of mitochondrial autophagy.
  • the present application provides promising compounds useful for the development of new drugs for the treatment of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, and inflammation associated with diabetes) and obesity or New therapy.
  • tautomer refers to a functional group isomer that is produced by the rapid movement of an atom in a molecule at two positions.
  • a typical example of such a tautomer is a keto-enol tautomer.
  • the compounds described herein may exist in tautomeric forms and thus encompass all possible tautomers, and any combination or any mixture thereof.
  • stereoisomer refers to an isomer that is caused by the same order of attachment of atoms or groups of atoms in a molecule, but differs in spatial arrangement.
  • “stereoisomerization” of a compound is classified into conformational and conformational isomerization, and configurational isomerism is also classified into cis-trans isomerization and optical isomerism.
  • “stereoisomer” includes all possible optical isomers and diastereomers, as well as any combination thereof, such as racemates (racemic mixtures), single enantiomeric A conformation, a mixture of diastereomers, a single diastereomer.
  • the compound of the present invention contains an olefinic double bond, it includes a cis isomer and a trans isomer, and any combination thereof, unless otherwise specified.
  • the term "pharmaceutically acceptable salts” refers to (1) a compound of the present invention, in the presence of acidic functional group (e.g. -COOH, -OH, -SO 3 H, etc.) with a suitable inorganic or organic a salt formed by a cation (base), for example, a salt of a compound of the invention with an alkali metal or alkaline earth metal, an ammonium salt of a compound of the invention, and a salt of a compound of the invention with a nitrogen-containing organic base; and (2) a compound of the invention A salt of a basic functional group (for example, -NH 2 or the like) which is formed with a suitable inorganic or organic anion (acid), for example, a salt of a compound of the present invention with an inorganic acid or an organic carboxylic acid.
  • acidic functional group e.g. -COOH, -OH, -SO 3 H, etc.
  • bases for example, a salt of a compound of the invention with
  • salts of the compounds of the invention include, but are not limited to, alkali metal salts such as sodium, potassium, lithium, and the like; alkaline earth metal salts such as calcium, magnesium, and the like; other metal salts, Such as aluminum salt, iron salt, zinc salt, copper salt, nickel salt, cobalt salt, etc.; inorganic alkali salt, such as ammonium salt; organic alkali salt, such as t-octylamine salt, dibenzylamine salt, morpholine salt, Portuguese Glycosylamine salt, phenylglycine alkyl ester salt, ethylenediamine salt, N-methylglucamine salt, sulfonium salt, diethylamine salt, triethylamine salt, dicyclohexylamine salt, N, N'- Dibenzylethylenediamine salt, chloroprocaine salt, procaine salt, diethanolamine salt, N-benzyl-phenethylamine salt, piperaz
  • the term "pharmaceutically acceptable ester” refers to an ester which is formed by esterification of an alcohol when a compound of the invention is present; when the compound of the invention has a hydroxyl group, it is organic An ester formed by an esterification reaction of an acid, an inorganic acid, an organic acid salt or the like. The ester can be hydrolyzed to form the corresponding acid or alcohol in the presence of an acid or a base.
  • C1-6 alkyl means a straight or branched alkyl group having 1 to 6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, or Butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, 2-methylbutyl, neopentyl, 1-ethylpropyl, n-hexyl, isohexyl, 3-methyl Pentyl, 2-methylpentyl, 1-methylpentyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 1, 2-dimethylbutyl, 1,3-dimethylbutyl, 2,3-dimethylbutyl, 2-ethylbutyl, 1,2-dimethylpropyl, and the like.
  • C 1-6 alkyl group Preferable examples of the C 1-6 alkyl group include a C 1-5 alkyl group, a C 1-4 alkyl group, and a C 1-3 alkyl group.
  • the "C 1-4 alkyl group” as used in the present invention means a linear or branched alkyl group having 1 to 4 carbon atoms, which includes, but is not limited to, a specific example having 1 to 4 carbon atoms in the above examples. .
  • alkenyl refers to a straight or branched chain hydrocarbon radical containing at least one carbon to carbon double bond, a typical example of which is a C 2-10 alkenyl group, such as a C 2-6 alkene. Base or C 2-4 alkenyl.
  • Specific examples include, but are not limited to, vinyl, propenyl, 2-propenyl, butenyl, 2-butenyl, butadienyl, pentenyl, 2-methyl-butenyl, 3-methyl -butenyl, 1,3-pentadienyl, 1,4-pentadienyl, hexenyl, 2-ethyl-butenyl, 3-methyl-pentenyl, 4-methyl a pentenyl group, a 1,3-hexadienyl group, a 1,4-hexadienyl group, a 1,5-hexadienyl group or the like.
  • alkynyl refers to a straight or branched chain hydrocarbon radical containing at least one carbon to carbon triple bond, a typical example of which is a C 2-10 alkynyl group, such as a C 2-6 alkynyl group or a C. 2-4 alkynyl.
  • Specific examples include, but are not limited to, ethynyl, propynyl, 2-propynyl, butynyl, 2-butynyl, 2-methyl-propynyl, butadiynyl, pentynyl, 2 -methyl-butynyl, 3-methyl-butynyl, 1,3-pentadiynyl, 1,4-pentadiynyl, hexynyl, 2-ethyl-butynyl, 3- Methyl-pentynyl, 4-methyl-pentynyl, 1,3-hexadiynyl, 1,4-hexadiynyl, 1,5-hexadiynyl and the like.
  • cycloalkyl refers to a monocyclic saturated alkyl group, a typical example of which is a 3-8 membered cycloalkyl group, such as 3, 4, 5, 6, 7 or 7 or 8-membered cycloalkyl.
  • 3-8 membered cycloalkyl refers to a cycloalkyl group containing from 3 to 8 carbon atoms. Specific examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like.
  • heterocycloalkyl refers to a cycloalkyl group containing at least 1 up to 4 (eg 1, 2, 3 or 4) heteroatoms selected from N, O and S, wherein
  • cycloalkyl is as described above, and a typical example thereof is a 3-8 membered heterocycloalkyl group such as a 3-, 4-, 5-, 6-, 7- or 8-membered heterocycloalkyl group.
  • 3-8 membered heterocycloalkyl refers to a heterocycloalkyl group containing from 3 to 8 carbon atoms.
  • Oxo 3-8 membered cycloalkyl refers to a 3-8 membered heterocycloalkyl group as defined above, wherein the hetero atom is O.
  • Specific examples include, but are not limited to, epoxyethyl, oxocyclobutyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl and the like.
  • aryl refers to an aromatic group, a typical example of which is a 6-14 membered aryl group, such as a 6-10 membered aryl group.
  • 6-14 membered aryl refers to a monocyclic, bicyclic or polycyclic aromatic group containing from 6 to 14 carbon atoms, including, for example, 6-8 membered aryl groups and 8-14 A fused ring aryl group.
  • the 6-8 membered aryl group means an aryl group having 6 to 8 carbon atoms, such as a phenyl group.
  • the 8- to 14-membered fused ring aryl group means an unsaturated aromatic fused ring formed by having two or more ring-shaped carbon atoms and two or more ring-shaped structures sharing two adjacent carbon atoms. Specific examples include, but are not limited to, naphthalene, anthracene, phenanthrene, and the like.
  • the term "6-10 membered aryl” means an aromatic group having 6 to 10 carbon atoms, which includes, but is not limited to, an aromatic group having 6 to 10 ring atoms in the above examples.
  • aryl-C 1-6 alkyl refers to a group formed in the manner of an aryl-C 1-6 alkyl group, wherein “aryl” and “C 1-6 alkane”
  • base The definitions of "base” are as described above.
  • C 1-6 alkoxy refers to a group formed in the C 1-6 alkyl-O- form, wherein “C 1-6 alkyl” is as defined above. .
  • C 1-6 alkylamino refers to a group formed in the C 1-6 alkyl-NH- form, wherein “C 1-6 alkyl” is as defined above.
  • C1-6 alkylthio refers to a group formed in the C1-6 alkyl-S- mode, wherein “ C1-6 alkyl” is as defined above .
  • C 1-6 alkanoyl refers to a group formed in the C 1-5 alkyl-C(O)-form, wherein "C 1-5 alkyl” is as defined above Said.
  • C1-6 alkoxycarbonyl refers to a group formed in the C1-6 alkyl-OC(O)- form, wherein “ C1-6 alkyl” is as defined As mentioned above.
  • C 1-6 alkoxycarbonyl-C 1-6 alkyl refers to a group formed by a C 1-6 alkyl-OC(O)-C 1-6 alkyl group.
  • 3-8 membered cycloalkyl-aminoacyl refers to a group formed in the form of a 3-8 membered cycloalkyl-NHC(O)-, wherein “3-8 membered cycloalkane”
  • base is as described above.
  • halogen includes, for example, a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
  • 6-15 membered heteroaryl refers to an aromatic group containing from 6 to 15 ring atoms and at least one of which is a hetero atom.
  • the 6-15 membered heteroaryl group includes a "5-8 membered heteroaryl group” such as "5-7 membered heteroaryl group", "5-6 membered heteroaryl group” and the like.
  • 5-8 membered heteroaryl include, but are not limited to, furyl, thienyl, pyrrolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl , imidazolyl, pyrazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, pyridyl, 2-pyridone, 4-pyridone, pyrimidinyl, 1,4-dioxadienyl 2H-1,2-oxazinyl, 4H-1,2-oxazinyl, 6H-1,2-oxazinyl, 4H-1,3-oxazinyl, 6H-1,3-oxazinyl 4H-1,
  • the 6-15 membered heteroaryl group also includes "9-15 membered fused heteroaryl group" (for example, 9-15 membered benzofused heteroaryl group), and specific examples thereof include, but are not limited to, benzofuranyl group, benzisofuran , benzothienyl, fluorenyl, isoindole, benzoxazolyl, benzimidazolyl, oxazolyl, benzotriazolyl, quinolinyl, 2-quinolinone, 4-quinoline Ketone, 1-isoquinolinone, isoquinolyl, acridinyl, phenanthryl, benzoxazinyl, pyridazinyl, quinazolinyl, quinoxalinyl, phenolzinyl, acridinyl, Anthracenyl, naphthyridinyl, phenazine, phenothiazine, and the like.
  • the term "cell” particularly preferably refers to a cell that expresses Nur77.
  • the compounds of the present invention are capable of specifically binding to Nur77 and functioning as a ligand thereof. Therefore, the invention is particularly advantageous
  • the compound is capable of acting on cells expressing Nur77.
  • the cell is an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell, an endothelial cell, an epithelial cell, a neural cell, a stem cell, a lymphocyte, etc.).
  • orphan nuclear receptor Nur77 refers to nerve growth factor IB (NGFIB), which is encoded by the NR4A1 gene (Chang C et al. (1989), “Isolation and characterization of human TR3”).
  • Receptor a member of steroid receptor superfamily", J. Steroid Biochem. 34(1-6): 391-5).
  • Nur77 is involved in processes such as cell cycle, inflammation, and apoptosis, and its subcellular localization is associated with cell survival and death (Pei L et al. (2006), “Regulation of macrophage inflammatory gene expression by the orphan nuclear receptor Nur77", Mol . Endocrinol. 20(4): 786-94; Zhang XK (2007), “Targeting Nur77 translocation", Expert Opin. Ther. Targets 11(1): 69-79).
  • Nur77-associated disease refers to a disease whose occurrence and/or progression is associated with the Nur77 signaling pathway. Studies have shown that Nur77 is involved in processes such as cell cycle, inflammation, and apoptosis, and its subcellular localization is associated with cell survival and death (ibid.). In addition, it has been reported that Nur77 can be induced by a variety of stimuli, including physiological stimuli such as fatty acids, prostaglandins, growth factors, inflammatory cytokines, peptide hormones, etc.; and physical stimuli such as magnetic fields, mechanical agitation (shearing forces) , membrane depolarization, etc.
  • physiological stimuli such as fatty acids, prostaglandins, growth factors, inflammatory cytokines, peptide hormones, etc.
  • physical stimuli such as magnetic fields, mechanical agitation (shearing forces) , membrane depolarization, etc.
  • Nur77 has also been shown to be involved in the metastasis of some solid tumors (Ramaswamy S, Ross KN, Lander ES, Golub TR (2003), "A molecular signature of metastasis in primary solid tumors", Nat. Genet. 33(1): 49-54). In addition, Nur77 has been shown to be abnormally expressed in inflamed human synovial tissue, cancer cells, psoriasis patients, atherosclerotic patients, and multiple sclerosis patients.
  • Neur77-associated diseases includes, but is not limited to, inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis, and inflammation) Enteropathy), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • inflammation eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis, and inflammation
  • Enteropathy eg, atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis
  • cancer eg, triple-negative breast cancer
  • the term "subject" refers to an animal, particularly a mammal, preferably a human.
  • high fat diet means that the amount of fat in a daily intake of an animal (eg, a mammal, such as a human) exceeds the amount of fat required for normal physiological activity of the animal.
  • the term "effective amount" refers to an amount sufficient to achieve, or at least partially achieve, a desired effect.
  • a prophylactically effective amount refers to an amount sufficient to prevent, arrest, or delay the onset of a disease
  • a therapeutically effective amount refers to an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. Determination of such an effective amount is well within the capabilities of those skilled in the art.
  • the amount effective for therapeutic use will depend on the severity of the disease to be treated, the overall state of the patient's own immune system, the general condition of the patient such as age, weight and sex, the mode of administration of the drug, and other treatments administered simultaneously. and many more.
  • immunological method refers to a detection method that utilizes specific interaction/binding affinity between antigen-antibodies, which can generally be used to detect the presence of a particular antigen or antibody in a sample or Level.
  • immunological assays are well known to those skilled in the art and include, but are not limited to, ELISA assays, Elispot assays, Western blots, surface plasmon resonance assays, and the like.
  • ELISA assays ELISA assays
  • Elispot assays Elispot assays
  • Western blots Western blots
  • surface plasmon resonance assays and the like.
  • a compound represented by Formula I-Formula V can target the orphan nuclear receptor Nur77 and function as a ligand thereof.
  • such compounds can inhibit the inflammatory response by binding to Nur77 to regulate mitochondrial activity, and prevent and treat diseases associated with the orphan nuclear receptor Nur77, such as inflammation (eg, inflammation associated with obesity, and atherosclerosis).
  • inflammation eg, inflammation associated with obesity, and atherosclerosis.
  • Inflammation inflammation associated with diabetes, hepatitis, pneumonia, arthritis, enteritis
  • atherosclerosis obesity, diabetes, psoriasis, multiple sclerosis and cancer.
  • the present application relates to the use of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, of formula I, for use as an orphan nuclear receptor Nur77 a ligand, or a drug used to prepare a ligand for use as an orphan nuclear receptor Nur77:
  • X represents -NH-, -N(R)-, -O-, -CH 2 - or halogen; wherein, when X is a halogen, R 1 is absent;
  • Y When Y is a single bond between the carbon atom to which it is attached, Y represents H, halogen, -OR, -SR or -NRR'; when Y is a double bond between the carbon atom to which it is attached, Y represents O, S or NR;
  • R 1 is absent or represents H, -PO(OR) 2 , C 1-6 alkyl, glycosyl, C 1-6 alkoxycarbonyl-C 1-6 alkyl, 3-8 membered cycloalkyl-ammonia
  • R 2 represents H, D, -PO(OR) 2 , -CONH 2 , -NH 2 , -NHR, -NRR', -NHCOR, -NRCOR, -NHCOOR, -NHCONHR, -NHCONRR', -NRCONHR, -NRCONRR ', -OH, -OR, -OCONHR, -OCONRR', -SH, -SR, -SOR, -SOOR, -SO 2 NHR", nitro, halogen, glycosyl, cyano, trifluoromethyl, C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, aryl, C 1-6 alkyl substituted aryl, 6-15 membered heteroaryl, alkenyl, alkyne a sulfinyl group, a sulfonic acid or a sulfonate; wherein the C 1-6 al
  • substituents selected from the group consisting of amino, halogen. , hydroxy, oxy, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1-6 alkanoyl, 3-8 membered cycloalkyl (eg cyclopropyl), Oxy 3-8 membered cycloalkyl (eg oxocyclobutyl), cyano, trifluoromethyl, C 1-6 alkoxycarbonyl, C 1-6 alkylamido, ureido, carbamate Base, carboxyl group and aryl group;
  • R 3 and R 4 each independently represent a non-existent or represent H, C 1-6 alkyl, C 1-6 alkanoyl, C 1-6 alkoxycarbonyl, glycosyl, aryl-C 1-6 alkyl Or an aryl group, wherein the C 1-6 alkyl group, the C 1-6 alkanoyl group, the C 1-6 alkoxycarbonyl group, the glycosyl group, the aryl-C 1-6 alkyl group, and the aryl group are unsubstituted or Or a plurality (for example 1, 2, 3 or 4) of substituents selected from the group consisting of halogen, hydroxy, amino, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylamino And a C 1-6 alkanoyl group; preferably, the aryl group is a 6-14 membered aryl group, such as a 6-10 membered aryl group; more preferably a phenyl or naphthyl group;
  • R and R' are each independently selected from H, C 1-6 alkyl, 3-8 membered cycloalkyl, aryl-C 1-6 alkyl or aryl, wherein said C 1-6 alkyl, 3
  • the 8- to 8-membered cycloalkyl, aryl-C 1-6 alkyl and aryl groups are unsubstituted or substituted by one or more (eg 1, 2, 3 or 4) substituents selected from the group consisting of halogens, a hydroxyl group, an amino group, a C 1-6 alkyl group, a C 1-6 alkoxy group, and a C 1-6 alkylamino group;
  • R" represents a C 1-6 alkyl or aryl group (for example a 6-10 membered aryl group, preferably a phenyl group);
  • Virtual real double bond in formula (I) Represents a single bond or a double bond; preferably, ring A contains 0, 1, 2 or 3 carbon-carbon double bonds; ring B contains 0, 1 or 2 carbon-carbon double bonds.
  • X represents NH, O or CH 2 ;
  • Y When Y is a single bond between the carbon atom to which it is attached, Y represents H, halogen, -OR, -SR or -NRR'; when Y is a double bond between the carbon atom to which it is attached, Y represents O, S or NR;
  • R 1 represents H, C 1-6 alkyl, aryl-C 1-6 alkyl or aryl, wherein the C 1-6 alkyl, aryl-C 1-6 alkyl and aryl are unsubstituted Or substituted by one or more (for example 1, 2, 3 or 4) substituents selected from the group consisting of halogen, hydroxy, amino, C 1-6 alkyl, C 1-6 alkoxy and C 1- 6 alkylamino; preferably, the aryl group is a 6-14 membered aryl group, such as a 6-10 membered aryl group; more preferably a phenyl or naphthyl group;
  • R 2 represents H, -CONH 2 , -NH 2 , -NHR, -NRR', -NHCOR, -NRCOR, -NHCOOR, -NHCONHR, -NHCONRR', -NRCONHR, -NRCONRR', -OH, -OR,- OCONHR,-OCONRR',-SH,-SR,-SOR,-SOOR,-SO 2 NHR",halogen,cyano,-CF 3 ,C 1-6 alkyl, 3-8 membered cycloalkyl, 3- 8-membered heterocycloalkyl, aryl, C 1-6 alkyl substituted aryl, 6-15 heteroaryl, alkenyl, alkynyl, sulfinyl, sulfonic acid or sulfonate; wherein said C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, aryl, C 1-6 alkyl
  • R 3 and R 4 each independently represent a non-existent or represent H, C 1-6 alkyl, aryl-C 1-6 alkyl or aryl, wherein said C 1-6 alkyl, aryl-C 1-6 alkyl and aryl are unsubstituted or substituted by one or more (eg 1, 2, 3 or 4) substituents selected from halogen, hydroxy, amino, C 1-6 alkyl, a C 1-6 alkoxy group and a C 1-6 alkylamino group; preferably, the aryl group is a 6-14 membered aryl group, for example a 6-10 membered aryl group; more preferably a phenyl group or a naphthyl group;
  • R and R' are each independently selected from H, C 1-6 alkyl, aryl-C 1-6 alkyl or aryl, wherein said C 1-6 alkyl, aryl-C 1-6 alkyl And the aryl group is unsubstituted or substituted by one or more (for example 1, 2, 3 or 4) substituents selected from the group consisting of halogen, hydroxy, amino, C 1-6 alkyl, C 1-6 alkane Oxyl and C 1-6 alkylamino;
  • R" represents a C 1-6 alkyl or aryl group (for example a 6-10 membered aryl group, preferably a phenyl group);
  • Virtual real double bond in formula (I) Represents a single bond or a double bond; preferably, ring A contains 0, 1, 2 or 3 carbon-carbon double bonds; and/or ring B contains 0, 1 or 2 carbon-carbon double bonds.
  • the sulfonate is selected from the group consisting of sodium sulfonate, potassium sulfonate, calcium sulfonate, and magnesium sulfonate.
  • the 6-15 membered heteroaryl is selected from the group consisting of 9-15 membered fused heteroaryl; more preferably, the 6-15 membered heteroaryl is selected from the group consisting of 9-15 membered benzo thick Heteroaryl, for example, fluorenyl, benzofuranyl, benzothienyl, benzimidazolyl or quinolyl.
  • Y is a double bond between the carbon atom to which it is attached, and Y represents O.
  • X represents NH
  • Y is a double bond between the carbon atom to which it is attached, and Y represents O
  • R 1 represents one or more (eg 1, 2, 3 or 4)
  • R 2 represents H.
  • X represents O; Y is a double bond between the carbon atom to which it is attached, and Y represents O; R 1 represents H; and R 2 represents H, sulfonate or 6-15.
  • a heteroaryl group preferably, the sulfonate is selected from the group consisting of sodium sulfonate, potassium sulfonate, calcium sulfonate and magnesium sulfonate; preferably, the 6-15 membered heteroaryl is selected from the group consisting of 9-15 yuan thick More preferably, the 6-15 membered heteroaryl group is selected from a 9-15 membered benzofused heteroaryl group, such as an indenyl group, a benzofuranyl group, a benzothienyl group, a benzimidazole or a quinoline. base.
  • X represents O
  • Y is a double bond between the carbon atom to which it is attached, Y represents O
  • R 1 represents a C 1-6 alkyl or aryl-C 1-6 alkyl group. (preferably benzyl); R 2 represents H.
  • R 3 is absent and R 4 represents H. In certain preferred embodiments, R 3 represents H and R 4 is absent. In certain preferred embodiments, both R 3 and R 4 are H.
  • R 3 is absent, R 4 represents H, and ring A and ring B each have two carbon-carbon double bonds.
  • R 3 represents H
  • R 4 is absent, and there are 0, 1 or 2 carbon-carbon double bonds in ring A; preferably, there are 0, 1 or 2 carbons in ring B Double key.
  • R 3 and R 4 are both H and there are 3 carbon-carbon double bonds in ring A (ie, ring A is a benzene ring); preferably, 0 or 1 in ring B A carbon-carbon double bond; more preferably, a carbon-carbon double bond is present between the 7- and 8-position carbon atoms in the ring B.
  • neither R 3 nor R 4 is present.
  • the carbon atom at position 2 in ring A is a carbon-oxygen double bond between the O atom to which it is attached, and the carbon atom at the 3-position carbon atom is bonded to the O atom to which it is attached.
  • a carbon-carbon double bond is between the 7 and 8 carbon atoms. In certain preferred embodiments, a carbon-carbon single bond is between the 7 and 8 carbon atoms.
  • X represents -NH -, - N (R) -, - O -, - CH 2 - or halogen;
  • R represents a C 1-6 alkyl group or a 3-8 membered cycloalkyl group (preferably Cyclohexyl); wherein, when X is a halogen, R 1 is absent;
  • X represents -NH-, -N(R)-, -O- or fluoro; R represents cyclohexyl.
  • R 1 is absent or represents hydrogen, C 1-4 alkyl, -PO(OR) 2 , monosaccharide, C 1-4 alkoxycarbonyl-C 1-4 alkyl, a 3-6 membered cycloalkyl-aminoacyl group, an aryl-C 1-4 alkyl group or an aryl group; wherein the C 1-4 alkyl group, monosaccharide group, C 1-4 alkoxycarbonyl group-C 1- 4- alkyl, 3-6 membered cycloalkyl-aminoacyl, aryl-C 1-4 alkyl and aryl are unsubstituted or are selected from one or more (eg 1, 2, 3 or 4) Substituted substituents: halogen, hydroxy, amino, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylamino and C 1-4 alkanoyl; preferably, the aryl is 6 a 14-membered aryl group,
  • R represents a C 1-4 alkyl group.
  • R 1 is absent or represents hydrogen, C 1-4 alkyl, -PO(OR) 2 , glucosyl, C 1-2 alkoxycarbonyl-C 1-2 alkyl, ring Hexyl-aminoacyl, phenyl-C 1-2 alkyl, naphthyl-C 1-2 alkyl, phenyl or naphthyl; wherein the methyl, ethyl, glucosyl, C 1-2 alkoxy Carbonyl-C 1-2 alkyl, cyclohexyl-aminoacyl, phenyl-C 1-2 alkyl, naphthyl-C 1-2 alkyl, phenyl or naphthyl are unsubstituted or one or more For example, 1, 2, 3 or 4) are substituted with a substituent selected from the group consisting of C 1-2 alkyl, C 1-2 alkoxy and C 1-2 alkanoyl;
  • R represents a C 1-4 alkyl group.
  • R 1 represents hydrogen, C 1-4 alkyl, -PO(OR) 2 or C 1-2 alkoxycarbonyl-C 1-2 alkyl;
  • R represents a C 1-3 alkyl group.
  • R 1 is absent or represents hydrogen, methyl, ethyl, -PO(OMe) 2 , -PO(OEt) 2 , -PO(O i Pr) 2 , 2,3, 4,6-Tetraacetoxy- ⁇ -D-glucopyranosyl, EtOCOCH 2 -, cyclohexyl-aminoacyl, benzyl, methoxyphenyl or tert-butylphenyl.
  • R 2 represents H, D, -OH, -PO(OR) 2 , C 1-6 alkyl, 9-15 membered fused heteroaryl or sulfonate; wherein said C 1 -6 alkyl or 6-15 membered heteroaryl is unsubstituted or substituted by one or more (eg 1, 2, 3 or 4) substituents selected from the group consisting of amino, halo, hydroxy, oxy, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkanoyl, cyano, trifluoromethyl and carboxyl;
  • R represents H, C 1-6 alkyl or aryl.
  • R 2 represents H, D, -PO(OR) 2 , C 1-4 alkyl, 9-15 membered benzofused heteroaryl or sulfonate; wherein said C 1 -4 alkyl or 9-15 membered benzoheteroaryl is unsubstituted or substituted by one or more (eg 1, 2, 3 or 4) substituents selected from the group consisting of amino, halogen, hydroxy, An oxy group, a C 1-4 alkyl group, a C 1-4 alkoxy group, a C 1-4 alkanoyl group, a cyano group, a trifluoromethyl group and a carboxyl group;
  • R represents H, C 1-4 alkyl or phenyl.
  • R 2 represents H, D or a sulfonate
  • R 2 represents H, D, -PO(OR) 2 , 2-oxophenyl, decyl or sodium sulfonate; wherein the thiol is unsubstituted or Or a plurality (for example 1, 2, 3 or 4) of substituents selected from the group consisting of amino, fluorine, chlorine, bromine, hydroxy, methyl, methoxy, formyl, cyano, trifluoromethyl And carboxyl groups;
  • R represents H, methyl, ethyl, isopropyl or phenyl.
  • the carbon and carbon double bonds between the 7 and 8 carbon atoms of the compound are preferred embodiments.
  • the compound Y and the carbon atom to which it is attached are carbon-carbon double bonds.
  • the compound Y and the carbon atom to which it is attached are a carbon-carbon single bond.
  • the compound has the structure:
  • R 3 and R 4 each independently represent H, C 1-6 alkyl or C 1-6 alkanoyl
  • R 3 and R 4 each independently represent H, C 1-4 alkyl or C 1-4 alkanoyl.
  • R 3 and R 4 each independently represent H, methyl or butanoyl.
  • the compound has the structure:
  • R 4 represents H, C 1-6 alkanoyl, C 1-6 alkoxycarbonyl or a monosaccharide group substituted by one or more (for example 1, 2, 3 or 4) C 1-6 alkanoyl;
  • R 4 represents H, C 1-4 alkanoyl, C 1-4 alkoxycarbonyl or by one or more (eg 1, 2, 3 or 4) C 1-4 alkanoyl Substituted glucose glycosyl;
  • R 4 represents H or C 1-2 alkoxycarbonyl
  • R 4 represents H, butyryl, ethoxycarbonyl or 2,3,4,6-tetraacetoxy- ⁇ -D-glucopyranose.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the transcriptional activity of Nur77.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit transcription of Nur77 in vivo, in vitro or ex vivo active.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit the transcriptional activity of Nur77 in a cell.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, Cervical cancer cells, lung cancer cells, breast cancer cells, colorectal cancer cells or prostate cancer cells) or inflammatory sensitive cells (such as fat cells, inflammatory cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, Cervical cancer cells, lung cancer cells, breast cancer cells, colorectal cancer cells or prostate cancer cells
  • inflammatory sensitive cells such as fat cells, inflammatory cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.) in cells.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other in vivo, in vitro or ex vivo Biological effects of inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells.
  • the biological effect of TNF ⁇ or other inflammatory factors is ⁇ B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / ⁇ ) Phosphorylation, degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B inhibitory protein
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ ) , IL-6 or IL-8, etc.) induced degradation of I ⁇ B ⁇ .
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Nur77, TRAF2 Mitochondrial transport of p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondrial activity. In certain preferred embodiments, the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondria activity in cells.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, A disease associated with Nur77 is prevented or treated in a subject in need thereof.
  • the disease associated with the orphan nuclear receptor Nur77 is selected from the group consisting of inflammation (eg, inflammation associated with obesity, inflammation associated with atherosclerosis, inflammation associated with diabetes, hepatitis, Pneumonia, arthritis, enteritis), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis and cancer.
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or The medicament is for inhibiting the level of a cytokine such as IL-1 ⁇ and/or IL-6 (for example, in serum and/or liver) in a subject having inflammation.
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application relates to a method of inhibiting the transcriptional activity of the orphan nuclear receptor Nur77, comprising: Nur77 with a compound of formula I, a tautomer, a stereoisomer or a pharmaceutically thereof Acceptable salt or ester phase contact steps:
  • the methods are for inhibiting the transcriptional activity of Nur77 in vivo, in vitro or ex vivo. In certain preferred embodiments, the method is for inhibiting the transcriptional activity of Nur77 in a cell. In certain preferred embodiments, the methods comprise administering to a cell in need thereof an effective amount of the compound to inhibit transcriptional activity of Nur77 in the cell. In certain preferred embodiments, the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inhibiting the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in a cell, including, to a cell in need thereof Administration of an effective amount of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, as shown in Formula I:
  • the method inhibits the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells in vivo, in vitro, or ex vivo.
  • TNF ⁇ or other inflammatory factors eg, IL-1 ⁇ , IL-6, or IL-8, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester binds to Nur77 in the cell and thereby inhibits TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL) -6 or IL-8, etc.) Biological effects in cells.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • the method is for inhibiting I ⁇ B ⁇ degradation induced by TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.).
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inducing interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub in a cell, comprising administering to a cell in need thereof an effective amount of Formula I a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the present application relates to a method of inducing mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub in a cell, comprising: Nur77 and a compound of formula I, a tautomer thereof , the step of contacting the stereoisomer or the pharmaceutically acceptable salt or ester:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the compounds of the invention, their tautomers, stereoisomers or pharmaceutically acceptable salts or esters or agents are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention, their tautomers, stereoisomers or pharmaceutically acceptable salts or esters or agents are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the present application relates to a method of modulating mitochondrial activity in a cell comprising administering to a cell in need thereof an effective amount of a compound of formula I, a tautomer thereof, a stereoisomer or A pharmaceutically acceptable salt or ester or the drug:
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the present application relates to a method of preventing or treating a disease associated with Nur77 comprising administering to a subject in need thereof an effective amount of a compound of formula I, a tautomer thereof , a stereoisomer or a pharmaceutically acceptable salt or ester:
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the method comprises administering to a subject having inflammation an effective amount of the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, thereby The level of cytokines such as IL-1 ⁇ and/or IL-6 (for example, levels in serum and/or liver) is inhibited in the tester.
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the method comprises administering to a subject having obesity an effective amount of the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt thereof or The ester thereby inhibiting weight gain and/or fat increase in the subject.
  • the method comprises administering to a subject having obesity an effective amount of the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt thereof or Ester, thereby inhibiting obesity complications in the subject, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the method comprises administering to a subject having cancer an effective amount of the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof. Thereby inhibiting proliferation and/or metastasis of cancer cells and/or promoting apoptosis of cancer cells in the subject.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the method further comprises administering to the subject having the cancer an effective amount of TNF[alpha].
  • the application relates to the use of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, according to formula IV, for use as a ligand for the orphan nuclear receptor Nur77 Or a drug for the preparation of a ligand for use as an orphan nuclear receptor Nur77:
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the transcriptional activity of Nur77.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit transcription of Nur77 in vivo, in vitro or ex vivo active.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit the transcriptional activity of Nur77 in a cell.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or is sensitive to inflammation.
  • Cells eg, adipocytes, inflammatory cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.) in cells.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other in vivo, in vitro or ex vivo Biological effects of inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B kappa B inhibitory protein
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ ) , IL-6 or IL-8, etc.) induced degradation of I ⁇ B ⁇ .
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its mutual variation A construct, a stereoisomer or a pharmaceutically acceptable salt or ester or the drug is used to induce mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub in a cell.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the compounds of the invention, their tautomers, stereoisomers or pharmaceutically acceptable salts or esters or agents are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention, their tautomers, stereoisomers or pharmaceutically acceptable salts or esters or agents are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondrial activity. In certain preferred embodiments, the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondria activity in cells.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, A disease associated with Nur77 is prevented or treated in a subject in need thereof.
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as IL-1 ⁇ and/or IL-6 levels (eg, at Levels in serum and / or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application relates to a method of inhibiting the transcriptional activity of the orphan nuclear receptor Nur77, comprising: Nur77 with a compound of formula IV, a tautomer, a stereoisomer or a pharmaceutically thereof Acceptable salt or ester phase contact steps:
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the methods are for inhibiting the transcriptional activity of Nur77 in vivo, in vitro or ex vivo. In certain preferred embodiments, the method is for inhibiting the transcriptional activity of Nur77 in a cell. In certain preferred embodiments, the methods comprise administering to a cell in need thereof an effective amount of the compound to inhibit transcriptional activity of Nur77 in the cell. In certain preferred embodiments, the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inhibiting the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in a cell, including, to a cell in need thereof Administration of an effective amount of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, as shown in Formula IV:
  • TNF ⁇ or other inflammatory factors eg, IL-1 ⁇ , IL-6, or IL-8, etc.
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the method inhibits the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells in vivo, in vitro, or ex vivo.
  • TNF ⁇ or other inflammatory factors eg, IL-1 ⁇ , IL-6, or IL-8, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester binds to Nur77 in the cell and thereby inhibits TNF ⁇ or other inflammatory factors Biological effects in cells such as IL-1 ⁇ , IL-6 or IL-8.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B kappa B inhibitory protein
  • the method is for inhibiting I ⁇ B ⁇ degradation induced by TNF ⁇ or other inflammatory factors such as IL-1 ⁇ , IL-6 or IL-8, and the like.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inducing interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub in a cell, comprising administering to a cell in need thereof an effective amount of Formula IV a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the present application relates to a method of inducing mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub in a cell comprising the step of formulating Nur77 with a compound of formula IV, tautomer thereof , the step of contacting the stereoisomer or the pharmaceutically acceptable salt or ester:
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the compounds of the invention, their tautomers, stereoisomers or pharmaceutically acceptable salts or esters or agents are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention, their tautomers, stereoisomers or pharmaceutically acceptable salts or esters or agents are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the present application relates to a method of modulating mitochondrial activity in a cell comprising administering to a cell in need thereof an effective amount of a compound of formula IV, a tautomer thereof, a stereoisomer or A pharmaceutically acceptable salt or ester or the drug:
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the present application relates to a method of preventing or treating a disease associated with Nur77 comprising administering to a subject in need thereof an effective amount of a compound of formula IV, a tautomer thereof, Steps of a stereoisomer or a pharmaceutically acceptable salt or ester:
  • X represents NH, O or CH 2 ;
  • Y represents O, S or NR
  • R 5 and R 6 each independently represent a H or C 1-6 alkyl group
  • R 7 When R 7 and a carbon atom to which it is bonded are a single bond, R 7 represents OH, and when R 7 and the carbon atom to which it is bonded are a double bond, R 7 represents O;
  • R 8 represents H or C 1-6 alkyl
  • Virtual real double bond in formula (IV) Represents a single button or a double button.
  • the compound is selected from the group consisting of:
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as the level of IL-1 ⁇ and/or IL-6 (eg, levels in serum and/or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or The medicament is for inhibiting weight gain and/or fat gain in a subject having obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the application relates to the use of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, which is used as a ligand for the orphan nuclear receptor Nur77, or for preparation Drugs used as ligands for the orphan nuclear receptor Nur77:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the transcriptional activity of Nur77.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit transcription of Nur77 in vivo, in vitro or ex vivo active.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit the transcriptional activity of Nur77 in a cell.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, Cervical cancer cells, lung cancer cells, breast cancer cells, colorectal cancer cells or prostate cancer cells) or inflammatory sensitive cells (such as fat cells, inflammatory cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, Cervical cancer cells, lung cancer cells, breast cancer cells, colorectal cancer cells or prostate cancer cells
  • inflammatory sensitive cells such as fat cells, inflammatory cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.) in cells.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other in vivo, in vitro or ex vivo Biological effects of inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B kappa B inhibitory protein
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ ) , IL-6 or IL-8, etc.) induced degradation of I ⁇ B ⁇ .
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Nur77, TRAF2 Mitochondrial transport of p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondrial activity. In certain preferred embodiments, the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondria activity in cells.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, A disease associated with Nur77 is prevented or treated in a subject in need thereof.
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cytokines such as IL-1 ⁇ and in a subject having inflammation / or the level of IL-6 (for example, levels in serum and / or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application relates to a method of inhibiting the transcriptional activity of the orphan nuclear receptor Nur77, comprising: Nur77 with the following compounds, tautomers, stereoisomers or pharmaceutically acceptable salts thereof or The steps of contacting the ester phase:
  • the methods are for inhibiting the transcriptional activity of Nur77 in vivo, in vitro or ex vivo. In certain preferred embodiments, the method is for inhibiting the transcriptional activity of Nur77 in a cell. In certain preferred embodiments, the methods comprise administering to a cell in need thereof an effective amount of the compound to inhibit transcriptional activity of Nur77 in the cell. In certain preferred embodiments, the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inhibiting the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in a cell, including, to a cell in need thereof Administration of an effective amount of a compound shown below, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • the method inhibits the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells in vivo, in vitro, or ex vivo.
  • TNF ⁇ or other inflammatory factors eg, IL-1 ⁇ , IL-6, or IL-8, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester binds to Nur77 in the cell and thereby inhibits TNF ⁇ or other inflammatory factors Biological effects in cells such as IL-1 ⁇ , IL-6 or IL-8.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • the method is for inhibiting I ⁇ B ⁇ degradation induced by TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.).
  • the cell expresses Nur77.
  • the cells are cancer cells (eg, liver cancer cells, cervical cancer cells, lung cancer cells, breast cancer cells, colorectal cancer cells, or prostate cancer cells) or inflammatory sensitive cells (eg, fat cells, inflammatory cells, endothelial cells, epithelial cells, nerves). Cells, stem cells, lymphocytes, etc.).
  • cancer cells eg, liver cancer cells, cervical cancer cells, lung cancer cells, breast cancer cells, colorectal cancer cells, or prostate cancer cells
  • inflammatory sensitive cells eg, fat cells, inflammatory cells, endothelial cells, epithelial cells, nerves. Cells, stem cells, lymphocytes, etc.
  • the present application relates to a method of inducing interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub in a cell, comprising administering to a cell in need thereof an effective amount of the following a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the present application relates to a method of inducing mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub in a cell comprising: Nur77 with a compound shown below, a tautomer thereof, a stereo
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable is used as a ligand for the orphan nuclear receptor Nur77 for inducing mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the present application relates to a method of modulating mitochondrial activity in a cell, comprising administering to a cell in need thereof an effective amount of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable compound thereof Accepted salt or ester or the drug:
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the present application relates to a method of preventing or treating a disease associated with Nur77 comprising administering to a subject in need thereof an effective amount of a compound shown below, a tautomer thereof, a stereoisomer Structure or Steps for pharmaceutically acceptable salts or esters:
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as the level of IL-1 ⁇ and/or IL-6 (eg, levels in serum and/or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application relates to a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof as shown below:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the present application relates to the use of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, according to Formula V, for use as a ligand for the orphan nuclear receptor Nur77 Or a drug for the preparation of a ligand for use as an orphan nuclear receptor Nur77:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the transcriptional activity of Nur77.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit transcription of Nur77 in vivo, in vitro or ex vivo active.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit the transcriptional activity of Nur77 in a cell.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of TNF ⁇ or other inflammation
  • the biological effects of disease factors such as IL-1 ⁇ , IL-6 or IL-8, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other in vivo, in vitro or ex vivo Biological effects of inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B kappa B inhibitory protein
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ ) , IL-6 or IL-8, etc.) induced degradation of I ⁇ B ⁇ .
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, a thin fat) Cells, inflammatory cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, a thin fat
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondrial activity. In certain preferred embodiments, the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondria activity in cells.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, A disease associated with Nur77 is prevented or treated in a subject in need thereof.
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as the level of IL-1 ⁇ and/or IL-6 (eg, levels in serum and/or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN)-induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application relates to a method of inhibiting the transcriptional activity of the orphan nuclear receptor Nur77, comprising: Nur77 and a compound of Formula V, a tautomer, a stereoisomer thereof or a pharmaceutically acceptable The steps of contacting the accepted salt or ester:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the methods are for inhibiting the transcriptional activity of Nur77 in vivo, in vitro or ex vivo. In certain preferred embodiments, the method is for inhibiting the transcriptional activity of Nur77 in a cell. In certain preferred embodiments, the methods comprise administering to a cell in need thereof an effective amount of the compound to inhibit transcriptional activity of Nur77 in the cell. In certain preferred embodiments, the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inhibiting the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in a cell, including, to a cell in need thereof Administration of an effective amount of a compound of formula V, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the method inhibits the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells in vivo, in vitro, or ex vivo.
  • TNF ⁇ or other inflammatory factors eg, IL-1 ⁇ , IL-6, or IL-8, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester binds to Nur77 in the cell and thereby inhibits TNF ⁇ or other inflammatory factors Biological effects in cells such as IL-1 ⁇ , IL-6 or IL-8.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • the method is for inhibiting I ⁇ B ⁇ degradation induced by TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.).
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inducing interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub in a cell, comprising administering to a cell in need thereof an effective amount of Formula V a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the present application relates to a method of inducing mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub in a cell, comprising: Nur77 and a compound represented by Formula V, a tautomer thereof, The step of contacting the stereoisomer or the pharmaceutically acceptable salt or ester:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the present application relates to a method of modulating mitochondrial activity in a cell comprising administering to a cell in need thereof an effective amount of a compound of formula V, a tautomer thereof, a stereoisomer or A pharmaceutically acceptable salt or ester or the drug:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the present application relates to a method of preventing or treating a disease associated with Nur77 comprising administering to a subject in need thereof an effective amount of a compound of formula V, a tautomer thereof, a stereo Steps for isomers or pharmaceutically acceptable salts or esters:
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-6 alkyl and aryl;
  • R 9 and R 10 are each independently selected from the group consisting of hydrogen, C 1-4 alkyl, phenyl and naphthyl;
  • R 9 and R 10 are each independently selected from the group consisting of methyl, ethyl and isopropyl.
  • the compound is selected from the group consisting of:
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • inflammation eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis
  • inflammatory bowel disease eg, atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis
  • cancer eg, triple-negative breast cancer
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as the level of IL-1 ⁇ and/or IL-6 (eg, levels in serum and/or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the application relates to the following compounds, tautomers, stereoisomers or pharmaceutically acceptable salts or esters thereof:
  • the application relates to the use of a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, which is used as a ligand for the orphan nuclear receptor Nur77, or for preparation Drugs used as ligands for the orphan nuclear receptor Nur77:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the transcriptional activity of Nur77.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit transcription of Nur77 in vivo, in vitro or ex vivo active.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit the transcriptional activity of Nur77 in a cell.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, Inhibition of the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6 or IL-8, etc.) in cells.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other in vivo, in vitro or ex vivo Biological effects of inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B kappa B inhibitory protein
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ ) , IL-6 or IL-8, etc.) induced degradation of I ⁇ B ⁇ .
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymph Cells, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymph Cells, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondrial activity. In certain preferred embodiments, the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for regulation Mitochondria activity in cells.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77, A disease associated with Nur77 is prevented or treated in a subject in need thereof.
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis and cancer (eg Such as Sanyin breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as the level of IL-1 ⁇ and/or IL-6 (eg, levels in serum and/or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer.
  • the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application relates to a method of inhibiting the transcriptional activity of the orphan nuclear receptor Nur77, comprising: Nur77 with the following compounds, tautomers, stereoisomers or pharmaceutically acceptable salts thereof or The steps of contacting the ester phase:
  • the methods are for inhibiting the transcriptional activity of Nur77 in vivo, in vitro or ex vivo. In certain preferred embodiments, the method is for inhibiting the transcriptional activity of Nur77 in a cell. In certain preferred embodiments, the methods comprise administering to a cell in need thereof an effective amount of the compound to inhibit transcriptional activity of Nur77 in the cell. In certain preferred embodiments, the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inhibiting the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in a cell, including, to a cell in need thereof Administration of an effective amount of a compound shown below, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • the method inhibits the biological effects of TNF ⁇ or other inflammatory factors (eg, IL-1 ⁇ , IL-6, or IL-8, etc.) in cells in vivo, in vitro, or ex vivo.
  • TNF ⁇ or other inflammatory factors eg, IL-1 ⁇ , IL-6, or IL-8, etc.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester binds to Nur77 in the cell and thereby inhibits TNF ⁇ or other inflammatory factors Biological effects in cells such as IL-1 ⁇ , IL-6 or IL-8.
  • biological effects of TNF ⁇ or other inflammatory factors include, but are not limited to, kappa B inhibitory protein (I ⁇ B) kinase ⁇ / ⁇ (IKK ⁇ / Phosphorylation of ⁇ ), degradation of I ⁇ B ⁇ , nuclear translocation of NF- ⁇ B subunit p65, and/or activation of NF- ⁇ B.
  • I ⁇ B kappa B inhibitory protein
  • the method is for inhibiting I ⁇ B ⁇ degradation induced by TNF ⁇ or other inflammatory factors such as IL-1 ⁇ , IL-6 or IL-8, and the like.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, inflammation) Cells, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, inflammation
  • the present application relates to a method of inducing interaction or colocalization of Nur77 with TRAF2, p62, LC3 and/or Ub in a cell, comprising administering to a cell in need thereof an effective amount of the following a compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof:
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77 in cells with TRAF2, p62, LC3 and / or Ub interaction or co-localization.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • the present application relates to a method of inducing mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub in a cell comprising: Nur77 with a compound shown below, a tautomer thereof, a stereo
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used as a ligand for the orphan nuclear receptor Nur77 for induction Mitochondrial transport of Nur77, TRAF2, p62, LC3 and/or Ub.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to induce Nur77, TRAF2, p62, LC3 and/ in cells Or mitochondrial transport of Ub.
  • the cell expresses Nur77.
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with LC3 and mitochondrial localization, Promote the process of autophagy.
  • the compounds of the invention are capable of promoting the interaction of Nur77 with p62 and mitochondrial localization, Promotes the removal of damaged mitochondria.
  • the present application relates to a method of modulating mitochondrial activity in a cell, comprising administering to a cell in need thereof an effective amount of a compound, tautomer, stereoisomer or pharmaceutically thereof as shown below
  • a compound, tautomer, stereoisomer or pharmaceutically thereof as shown below
  • the cell is a cancer cell (eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell) or an inflammatory sensitive cell (eg, an adipocyte, an inflammatory cell) , endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.).
  • a cancer cell eg, a liver cancer cell, a cervical cancer cell, a lung cancer cell, a breast cancer cell, a colorectal cancer cell, or a prostate cancer cell
  • an inflammatory sensitive cell eg, an adipocyte, an inflammatory cell
  • endothelial cells e.g, endothelial cells, epithelial cells, nerve cells, stem cells, lymphocytes, etc.
  • the present application relates to a method of preventing or treating a disease associated with Nur77 comprising administering to a subject in need thereof an effective amount of a compound shown below, a tautomer thereof, a stereoisomer Structure or Steps for pharmaceutically acceptable salts or esters:
  • the Nur77-associated disease is selected from the group consisting of inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis, pneumonia, arthritis) Or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis, and cancer (eg, triple-negative breast cancer).
  • the Nur77-related disease is selected from the group consisting of inflammation, obesity, and cancer.
  • the disease associated with Nur77 is inflammation.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting cells in a subject having inflammation Factors such as the level of IL-1 ⁇ and/or IL-6 (eg, levels in serum and/or liver).
  • the inflammation is an inflammation associated with TNF[alpha] (eg, acute inflammation or chronic inflammation).
  • the inflammation is hepatitis or pneumonia, such as acute hepatitis or pneumonia (eg, lipopolysaccharide (LPS) and/or D-galactosamine (D-GalN) induced acute hepatitis or pneumonia).
  • the inflammation is chronic inflammation, such as chronic inflammation in a subject having obesity.
  • the disease associated with Nur77 is obesity.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the medicament is for inhibiting the body weight of a subject suffering from obesity Growth and / or fat increase.
  • the compound, its tautomer, stereoisomer or pharmaceutically acceptable salt or ester or the drug is used to inhibit concurrency in a subject having obesity Symptoms, such as chronic inflammation.
  • the obesity is caused by a high fat diet.
  • the disease associated with Nur77 is cancer.
  • the cancer is selected from the group consisting of liver cancer, cervical cancer, lung cancer, and breast cancer. In some excellent In selected embodiments, the cancer is triple negative breast cancer (ie, breast cancer with negative estrogen receptor (ER), progesterone receptor (PR), and proto-oncogene Her-2).
  • the medicament is for use in treating cancer and comprises the compound, a tautomer, a stereoisomer or a pharmaceutically acceptable salt or ester thereof, and TNF ⁇ .
  • the present application is directed to a screening method for a medicament having anti-inflammatory and/or anti-obesity activity, comprising the steps of:
  • the a-j term can be detected by immunological methods.
  • the immunological method is selected from the group consisting of an ELISA assay, an Elispot assay, a Western blot, a surface plasmon resonance method, an immunofluorescence stain, an immunohistochemical stain, and an immunoprecipitation.
  • the compound transports Nur77 from the nucleus to the mitochondria, and the tumor necrosis factor receptor-associated factor 2 (TRAF2, an important scaffold protein and a key pan in an inflammatory signaling pathway) Ligase) interaction.
  • the interaction is mediated by the LXXLL motif on TRAF2. This interaction not only inhibits ubiquitination of TRAF2 but also induces ubiquitination of Nur77 at K63.
  • ubiquitinated Nur77 interacts with the p62/SQSTM1 ubiquitin assembly domain on the mitochondria to enhance mitochondrial autophagy sensitivity.
  • LC3 an autophagy-related protein, specifically interacts with the modified Nur77 to ensure that the damaged mitochondria are selectively cleared.
  • the present inventors screened a series of compounds having anti-inflammatory and/or therapeutic obesity activity by detecting the interaction of Nur77 with TRAF2, LC3, p62 and/or Ub and mitochondrial transport. Thus, the inventors have established a method of screening for anti-inflammatory and/or anti-obesity drugs.
  • the compounds of the present application are also useful for the prevention and treatment of diseases associated with the orphan nuclear receptor Nur77, such as inflammation (e.g., inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis) , pneumonia, arthritis or inflammatory bowel disease), atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis and cancer (eg triple-negative breast cancer).
  • inflammation e.g., inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes, hepatitis
  • pneumonia e.g., arthritis or inflammatory bowel disease
  • the present invention provides novel, potent, and specific binding to Nur77 ligands that can be used to develop treatments for inflammation (eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes) New treatments for hepatitis, pneumonia, arthritis or inflammatory bowel disease, atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis and cancer (eg triple-negative breast cancer).
  • inflammation eg, inflammation associated with atherosclerosis, inflammation associated with obesity, inflammation associated with diabetes
  • New treatments for hepatitis, pneumonia, arthritis or inflammatory bowel disease, atherosclerosis, obesity, diabetes, psoriasis, multiple sclerosis and cancer eg triple-negative breast cancer.
  • Figure 1 is a graph showing the mechanism by which the inventors of the present application discovered that Nur77 is involved in the inhibition of inflammation and induction of autophagy.
  • Figure 2 shows the screening of compounds that bind to Nur77 using the Biacore T200 instrument.
  • the figure shows the experimental results of detecting the combination of YXY101 and Nur77-LBD with a Biacore T200 instrument, where the red dot represents Compound YXY101; blue dots represent the control compound. The results showed that the compound YXY101 was able to bind to Nur77-LBD.
  • Figure 3A shows the chemical structural formula of tripterine (compound YXY101).
  • Figure 3B shows the results of experiments with further binding of different concentrations of YXY101 (0.04 ⁇ M, 0.08 ⁇ M, 0.16 ⁇ M, 0.32 ⁇ M, 0.64 ⁇ M) to Nur77-LBD.
  • the results showed that the dissociation constant (Kd) of the compound YXY101 combined with Nur77-LBD was 292 nM;
  • Fig. 3C shows the experimental results of detecting the binding of YXY101 to Nur77-LBD by circular dichroism spectroscopy, wherein the red curve represents YXY101+Nur77-LBD; the blue curve represents Nur77-LBD.
  • the results showed that the compound YXY101 could change the CD spectrum of Nur77-LBD. This indicates that the compound YXY101 can bind to Nur77-LBD.
  • Figure 3D shows the experimental results of the detection of the binding of YXY101 to Nur77-LBD by HPLC, wherein the red curve represents YXY101+Nur77-LBD; the purple curve represents YXY101+RXR ⁇ -LBD. The results showed that the compound YXY101 was able to combine with Nur77-LBD to form a complex, but did not bind to RXR ⁇ -LBD.
  • Figure 3E shows the results of an experiment for detecting the binding of YXY101 to Nur77-LBD using the dual luciferase reporter system.
  • the results showed that the compound YXY101 inhibited the transcriptional activation of Nur77, but had no significant effect on the transcriptional activation of the glucocorticoid receptor (GR).
  • GR glucocorticoid receptor
  • Figure 3F shows the molecular docking of YXY101 with Nur77.
  • Molecular docking results show that YXY101 binds to the known hydrophobic grooves on the surface of the Nur77 protein mainly by hydrophobic interaction.
  • Figures 4A-4B show the results of immunoblot analysis of I ⁇ B ⁇ and phosphorylated IKK ⁇ / ⁇ in cells treated with different concentrations of YXY101 and TNF ⁇ .
  • Figure 4C shows the results of immunofluorescence staining of cells treated with YXY101 and TNF ⁇ (Scale bar: 20 ⁇ m).
  • Figure 4D shows the results of analysis of NF- ⁇ B activity of cells treated with YXY101 and TNF ⁇ , wherein **P < 0.01, ***P < 0.001 (T test).
  • Figure 4E shows I ⁇ B ⁇ and phosphoric acid in different cancer cell lines treated with different concentrations of YXY101 and TNF ⁇ . The results of immunoblot analysis of IKK ⁇ / ⁇ .
  • Figure 4F shows the results of immunoblot analysis of I ⁇ B ⁇ in HepG2 cells treated with different compounds and stimulated with TNF ⁇ .
  • Figures 5A-5B show the results of immunoblot analysis of Nur77, RXR ⁇ and I ⁇ B ⁇ in HepG2 cells transfected with different siRNAs and treated with different concentrations of YXY101 and TNF ⁇ .
  • Figure 5C shows the results of immunoblot analysis of I ⁇ B ⁇ in MEF cells and Nur77-/-MEF cells treated with different concentrations of YXY101 and TNF ⁇ .
  • Figure 5D shows the results of immunofluorescence staining of MEF cells and Nur77-/-MEF cells treated with YXY101 and TNF ⁇ (Scale bar: 10 ⁇ m).
  • Figure 6A shows serum ALT and serum AST levels in mice of each treatment group.
  • Figure 6B shows the levels of serum IL-1 ⁇ and serum IL-6 in mice of each treatment group.
  • Figure 6C shows mRNA levels of liver IL-1 ⁇ and liver IL-6 in mice of each treatment group.
  • the data represent the mean ⁇ SEM of three independent experiments; ns indicates no significant difference; * P ⁇ 0.05, ** P ⁇ 0.01, *** P ⁇ 0.001 (Student's t-test).
  • Figure 6D shows the protein levels of liver I ⁇ B ⁇ in mice of each treatment group.
  • Figure 7A shows the H&E staining results (scale bar, 100 ⁇ m) of the liver of each treatment group of mice.
  • Figure 7B shows the p65 immunohistochemical staining results (scale bar, 20 ⁇ m) of the liver of mice in each treatment group.
  • Figure 8A shows the body type, body weight, and status of adipose tissue blocks of mice in each treatment group.
  • Figure 8B shows mRNA levels of liver IL-1 ⁇ and liver IL-6 in mice of each treatment group.
  • Figure 8C shows the protein levels of liver I ⁇ B ⁇ in mice of each treatment group.
  • Fig. 8D shows the results of H&E staining of the liver of each treatment group, the results of p65 immunohistochemical staining, and the staining results of hepatic neutrophils.
  • Figure 9A shows that wild-type and Nur77-deficient MEF cells were treated with TNF ⁇ (20 ng/ml, 30 min) and YXY101 (2 ⁇ M, 1 h), and the localization of Nur77 and TRAF2 in mitochondria was observed by immunofluorescence technique.
  • Figure 9B shows the statistical results of Nur77, TRAF2, mitochondria colocalization in the above treated cells.
  • Figure 9C shows that HepG2 cells were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M, 1 h/9 h), mitochondria were isolated, and Nur77, TRAF2, p62, LC3 in whole cell proteins and mitochondrial proteins were detected by immunoblotting. PARP, Hsp60 levels.
  • FIG. 10A shows that HepG2 cells, IP:TRAF2, were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and Nur77 interacting with TRAF2 was detected by immunoblotting.
  • Figure 10B shows that His-Nur77-LBD protein bound to GST-TRAF2 protein was detected by immunoblotting using YXY101 in combination with GST-TRAF2, His-Nur77-LBD protein.
  • Figure 1C shows that the Flag-TRAF2 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and the co-localization of endogenous Nur77 and exogenous Flag-TRAF2 was detected by immunostaining.
  • Figure 10D shows that the Flag-TRAF2, GFP-Nur77 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M). The co-localization of Flag-TRAF2, GFP-Nur77 and mitochondria was detected by immunostaining. .
  • Figure 11A shows that the Flag-TRAF2, GFP-LC3 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M). The co-localization of Flag-TRAF2, GFP-LC3 and mitochondria was detected by immunostaining. .
  • Figure 11B shows the transfection of RFP-LC3, GFP-Nur77 plasmid in HepG2 cells, and treatment with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), using immunostaining to detect colocalization of RFP-LC3, GFP-Nur77 and mitochondria .
  • Figure 11C shows that wild-type and Nur77-deficient MEF cells were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of Nur77, LC3 and mitochondria was detected by immunostaining.
  • Figure 11D shows the statistical results of Nur77, LC3, mitochondria colocalization in the above treated cells.
  • Figure 12A shows that the Myc-Nur77, Flag-p62 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), IP: Flag, and Myc interacting with Flag-p62 was detected by immunoblotting. -Nur77.
  • Figure 12B shows that HepG2 cells were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of Nur77 and p62 was detected by immunostaining.
  • Figure 12C shows that the GFP-LC3 plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of GFP-Nur77 and p62 was detected by immunostaining.
  • Figure 13A shows the transfection of Flag-TRAF2, Myc-Nur77, HA-Ub plasmids in HepG2 cells, treatment with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), IP: Myc, detection of ubiquitination of Nur77 by immunoblotting happening.
  • FIG. 13B shows that Myc-Nur77, HA-Ub plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), mitochondria were isolated, IP: Myc, and Nur77 pan in mitochondria was detected by immunoblotting. The situation of vegetarianization.
  • Figure 13C shows that GFP-Nur77, HA-Ub plasmids were transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of GFP-Nur77 and HA-Ub was detected by immunostaining.
  • Figure 13D shows that GFP-Nur77 plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of GFP-Nur77 and Ub was detected by immunostaining.
  • Figure 14A shows the results of immunoblot analysis of I ⁇ B ⁇ in MEF cells treated with different concentrations of XS0284 and TNF ⁇ and Nur77-/-MEF.
  • Figure 14B shows the results of immunoblot analysis of I ⁇ B ⁇ in MEF cells treated with different concentrations of XS0284 and TNF ⁇ and Nur77-/-MEF.
  • Figure 14C shows the results of immunoblot analysis of I ⁇ B ⁇ in MEF cells treated with different concentrations of XS0474, XS0503, XS0419, XS0486 and TNF ⁇ and Nuk77-/-MEF.
  • Figure 15A shows the initial screening of YXY101 derivatives that bind to Nur77 using a Biacore T200 instrument with YXY101 as a control.
  • Figure 15B shows the results of experiments with further binding of different concentrations of YXY101 derivative (0.3125 ⁇ M, 0.625 ⁇ M, 1.25 ⁇ M, 2.5 ⁇ M, 5 ⁇ M, 10 ⁇ M) to Nur77-LBD.
  • the dissociation constant (Kd) of the compound XS0284 and Nur77-LBD was 386 nM; the combination of XS0394 and Nur77-LBD
  • the dissociation constant (Kd) was 1.26 ⁇ M; the dissociation constant (Kd) of the compound XS0418 and Nur77-LBD was 3.67 ⁇ M; the dissociation constant (Kd) of the compound XS0462 and Nur77-LBD was 2.07 ⁇ M; the compounds XS0474 and Nur77
  • the dissociation constant (Kd) of -LBD binding was 2.60 ⁇ M; the dissociation constant (Kd) of compound XS0486 and Nur77-LBD was 1.20 ⁇ M; the dissociation constant (Kd) of compound XS0419 and Nur77-LBD was 404 nM;
  • the dissociation constant (Kd) of XS0503 in combination with Nur77-LBD was 1.63 ⁇ M.
  • Figure 16A shows I ⁇ B ⁇ and PARP in MDA-MB-231 cells treated with YXY101 and different YXY101 derivatives XS0284, XS0285, XS0286, XS0287, XS0335, XS0366, XS0260, XS0394, XS0395, XS0419, XS0420, XS0421 and TNF ⁇ .
  • Figure 16B shows MDA treated with YXY101 and different YXY101 derivatives XS0284-4, XS0284, XS0077, XS0503, XS0486, XS0419, XS0474, XS0462, XS0285, XS0394, XS0454, XS0455, XS0462, XS0473, XS0474, XS0480, and TNF ⁇ .
  • Figure 16C shows MDA-MB treated with YXY101 and different YXY101 derivatives XS0284, XS0285, XS0335, XS0394, XS0418, XS0419, XS0454, XS0455, XS0462, XS0502, XS0503, XS0504, XS0506, XS0507, XS0508, XS0077 and TNF ⁇ .
  • Figure 17A shows the transfer of Flag-p62, Myc-Nur77 plasmid in HepG2 cells, treated with TNF ⁇ (20 ng/ml), YXY101 (4 ⁇ M), XS0284 (2 ⁇ M, 4 ⁇ M), IP: Flag, using immunoblotting and Flag- P62 interacts with Myc-Nur77.
  • Figure 17B shows the transfer of Flag-TRAF2, HA-Ub plasmid in HepG2 cells, treatment with TNF ⁇ (20 ng/ml), YXY101 (4 ⁇ M), XS0284 (4 ⁇ M), IP: Flag, detection of Flag-TRAF2 pan by immunoblotting The situation of vegetarianization.
  • Figure 17C shows the transfection of GFP-Nur77, Flag-TRAF2 plasmid in HepG2 cells, treatment with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), XS0284 (4 ⁇ M), and detection of GFP-Nur77 and Flag-TRAF2 by immunostaining technique. Colocalization of mitochondria.
  • Figure 17D shows the transfection of GFP-Nur77, RFP-LC3 plasmid in HepG2 cells, treatment with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), XS0284 (4 ⁇ M), and detection of GFP-Nur77 by immunostaining technique. Co-localization with RFP-LC3.
  • Figure 17E shows that GFP-Nur77, Flag-p62 plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), XS0284 (4 ⁇ M), and GFP-Nur77 and Flag-p62 were detected by immunostaining. Colocation of Ub.
  • Figure 18A shows the results of immunofluorescence staining of Hela cells treated with XS0077 and TNF ⁇ .
  • HepG2 cells were treated with XS0077 (2 ⁇ M) for 3 hours and then treated with TNF ⁇ (20 ng/mL) for 30 minutes. Subsequently, the colocalization of Nur77 and Traf2 in the cells was detected by immunofluorescence staining.
  • Figure 18B shows the results of immunofluorescence staining of Hela cells treated with XS0503 and TNF ⁇ .
  • Hela cells were treated with XS0503 (2 ⁇ M) for 3 hours and then treated with TNF ⁇ (20 ng/mL) for 30 minutes. Subsequently, the colocalization of Nur77 and Traf2 in the cells was detected by immunofluorescence staining.
  • Figure 19A shows the results of immunoblot analysis of PARP in MDA-MB-231 cells treated with YXY101 and XS0284 and TNF ⁇ .
  • Figure 19B shows the results of immunoblot analysis of PARP in NCL-H292 cells treated with YXY101 and XS0284 and TNF ⁇ .
  • Figure 19C shows the inhibition rate of proliferation of HepG2 cells by compounds YXY101 and XS0284.
  • Figure 19D shows changes in mitochondrial membrane potential of compounds YXY101 and XS0284 and TNF ⁇ treated MDA-MB-231 by flow cytometry.
  • Fig. 20A shows the tissue morphology of heart, liver, small intestine and white fat of each group of mice after intragastric administration of 6 mg C57BL/6 mice to 200 mg/kg of YXY101 and its derivative XS0284, respectively. And the results of HE staining.
  • Fig. 20B shows the tissue morphology of heart, liver, white fat and kidney of each group of mice after intraperitoneal injection of 200 mg/kg of YXY101 and its derivative XS0284 in 6 weeks of C57BL/6 mice. And the results of HE staining.
  • Figure 21 uses high fat-induced obese mice, intraperitoneally injected with YXY101 (0.1 mg/kg) and XS0284 (0.1 mg/kg) for one week.
  • Figure 21A shows the external morphological characteristics of the mouse and the morphological characteristics of the liver;
  • Figure 21B shows the statistical analysis of the change in body weight of the above mouse;
  • Figure 21C shows the change in body weight of the above mouse within 7 days of administration.
  • Figure 21D shows a graph of percent body weight loss in mice.
  • Figure 22 shows a schematic diagram of the effect of computer-assisted simulation of the binding of Nur77 to its ligand.
  • the molecular biology experimental methods and immunoassays used in the present invention are basically referred to J. Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, 1989, and The method described in FMAusubel et al., Guide to Molecular Biology, 3rd Edition, John Wiley & Sons, Inc., 1995; the use of restriction enzymes according to the conditions recommended by the product manufacturer.
  • the reagents or instruments used are not indicated by the manufacturer, and are conventional products that can be obtained commercially.
  • Fig. 1 is a view showing the mechanism of the first discovery of Nur77 involved in the inhibition of inflammation and induction of autophagy by the inventors of the present application.
  • the inventors of the present application found that certain compounds are capable of inhibiting inflammation and autophagy by binding to Nur77 and inducing Nur77-dependent inhibition.
  • the compound interacts with tumor necrosis factor receptor-associated factor 2 (TRAF2, an important scaffold protein in the inflammatory signaling pathway and a key ubiquitin ligase) by facilitating the transport of Nur77 from the nucleus to the mitochondria.
  • TRAF2 tumor necrosis factor receptor-associated factor 2
  • the interaction is mediated by the LXXLL motif on TRAF2. This interaction not only inhibits ubiquitination of TRAF2 but also induces ubiquitination of Nur77 at K63.
  • ubiquitinated Nur77 interacts with the p62/SQSTM1 ubiquitin assembly domain on the mitochondria to enhance mitochondrial autophagy sensitivity.
  • LC3 an autophagy-related protein, specifically interacts with the modified Nur77 to ensure that the damaged mitochondria are selectively cleared.
  • the inventors of the present application established a method for screening Nur77-dependent compounds that inhibit inflammation and induce autophagy by the mechanism described above, and analyzed the biological activity of the selected compounds, which will be Further explanation is given below.
  • YXY101 The binding of YXY101 to Nur77 was determined by surface plasmon resonance. Briefly, 50 [mu]g of purified Nur77 ligand binding domain (Nur77-LBD) protein was coupled to Biacore's CM5 chip; binding of YXY101 (20 [mu]m) to Nur77-LBD was then determined by a Biacore T200 instrument. Unrelated compounds were used as controls. The measurement results are shown in Fig. 2 .
  • Figure 2 shows the results of an experiment in which YXY101 was combined with Nur77-LBD using a Biacore T200 instrument, wherein red dots represent compound YXY101; blue dots represent control compounds. The results showed that the compound YXY101 was able to bind to Nur77-LBD.
  • the dissociation constant (Kd) of YXY101 and Nur77 was determined by surface plasmon resonance. Briefly, binding of different concentrations of YXY101 (0.04 ⁇ M, 0.08 ⁇ M, 0.16 ⁇ M, 0.32 ⁇ M, 0.64 ⁇ M) to Nur77-LBD was determined using a Biacore T200 instrument. The measurement results are shown in Fig. 3B.
  • YXY101 and Nur77-LBD were further analyzed using circular dichroism spectroscopy (CD). Briefly, YXY101 (1 ml, 1 mg/ml) was added to a phosphate buffer (10 ⁇ m, pH 7.4) of Nur77-LBD protein (1 ml, 1 mg/ml) and incubated at 4 ° C for 3 h. Subsequently, 0.7 ml of the solution was taken and detected using a Jasco J-810 spectropolarimeter. CD spectra from 190 nm to 260 nm were recorded. A separate Nur77-LBD solution (i.e., no compound YXY101 was added) was used as a control. The results of the detection are shown in Figure 3C.
  • YXY101 and Nur77-LBD were further analyzed using high performance liquid chromatography (HPLC). Briefly, YXY101 (600 uL, 0.1 mg/ml) was incubated with purified Nur77-LBD protein (5 ml, 1 mg/ml). After incubation for 3 h at 4 °C, the complex of YXY101 and Nur77-LBD was captured with Ni beads. Subsequently, the complex was dissociated using chloroform, and YXY101 in the dissociated product was extracted.
  • HPLC high performance liquid chromatography
  • YXY101 and Nur77-LBD were further analyzed using a dual luciferase reporter assay.
  • the experiment was repeated using YXY101 with a glucocorticoid receptor (GR) for use as a control.
  • GR glucocorticoid receptor
  • the compound YXY101 inhibited the transcriptional activation of Nur77, but had no significant effect on the transcriptional activation of the glucocorticoid receptor (GR). This indicates that the compound YXY101 is capable of binding to Nur77-LBD and inhibiting its transcriptional activity; and, the compound YXY101 does not bind to GR.
  • YXY101 and Nur77 (PDB code: 4JGV).
  • the conformation of YXY101 was constructed by the Lamarck genetic algorithm.
  • the lattice center is selected in the reported THPN coordinates (-12.08, 18.29, -4.233), and the grid size is set to 40*40*40 (X, Y, Z) grid points, each grid The dot spacing is 0.375A.
  • HepG2 cells were treated with different concentrations of YXY101 (0 ⁇ M, 0.25 ⁇ M, 0.5 ⁇ M, 1 ⁇ M, 2 ⁇ M or 4 ⁇ M) was treated for 1 hour and treated with TNF ⁇ (0 ng/mL or 20 ng/mL) for 30 minutes. Subsequently, I ⁇ B ⁇ and phosphorylated IKK ⁇ / ⁇ in the cells were detected by Western Blotting. The results are shown in Figures 4A-4B.
  • HepG2 cells were treated with YXY101 (0 or 1 ⁇ M) for 1 hour and then treated with TNF ⁇ (20 ng/mL) for 30 minutes. Subsequently, the NF- ⁇ B subunit p65 in the cells was detected by immunofluorescence staining. Untreated cells were used as controls. The results are shown in Figure 4C.
  • Figure 4C shows the results of immunofluorescence staining of HepG2 cells treated with YXY101 and TNF ⁇ (Scalebar: 20 ⁇ m).
  • the results in Figure 3C show that TNF ⁇ is able to induce nuclear translocation of the NF- ⁇ B subunit p65 in cells; whereas YXY101 is able to inhibit TNF ⁇ -induced p65 nuclear translocation.
  • the NF- ⁇ B reporter gene was transfected into HEK-293T cells and then treated with YXY101 (0 or 1 ⁇ M) and TNF ⁇ (20 ng/mL). Subsequently, NF- ⁇ B activity in the cells was examined. Untreated cells were used as controls. The results are shown in Figure 4D.
  • Figure 4D shows the results of analysis of NF- ⁇ B activity of cells treated with YXY101 and TNF ⁇ , wherein **P < 0.01, ***P < 0.001 (T test).
  • the results in Figure 4D show that TNF ⁇ is able to induce transcriptional activation of NF- ⁇ B in cells; whereas YXY101 is able to inhibit TNF ⁇ -induced NF- ⁇ B transcriptional activation.
  • a variety of cancer cell lines (LO2, SMMC-7721, QGY-7703, HeLa, H460) were treated with different concentrations of YXY101 (0 ⁇ M, 1 ⁇ M or 4 ⁇ M) for 1 hour and then treated with TNF ⁇ (0 ng/mL or 20 ng/mL). minute. Subsequently, I ⁇ B ⁇ and phosphorylated IKK ⁇ / ⁇ in the cells were detected by Western Blotting. The results are shown in Figure 4E.
  • TNF ⁇ was able to induce phosphorylation of IKK ⁇ / ⁇ and degradation of I ⁇ B ⁇ in various cell lines, while YXY101 was able to inhibit TNF ⁇ -induced phosphorylation of IKK ⁇ / ⁇ and degradation of I ⁇ B ⁇ .
  • HepG2 cells were also used for the experiments. Briefly, HepG2 cells were treated with various concentrations of various compounds (YXY101, XS0284, and XS0287) for the indicated times and then treated with TNF ⁇ (0 ng/mL or 20 ng/mL) for 30 minutes. Subsequently, I ⁇ B ⁇ in the cells was detected by Western Blotting. The results are shown in Figure 4F.
  • Figure 4F shows that YXY101 and its derivatives XS0284 and XS0287 are capable of inhibiting TNF ⁇ -induced degradation of I ⁇ B ⁇ in HepG2 cells.
  • YXY101 and its derivatives XS0284 and XS0287 are capable of inhibiting various biological effects of TNF ⁇ in cells, including phosphorylation of IKK ⁇ / ⁇ phosphorylation, degradation of I ⁇ B ⁇ , and NF- ⁇ B subunit p65 Nuclear transport, and transcriptional activation of NF- ⁇ B.
  • Control siRNA, Nur77 siRNA or RXR ⁇ siRNA were transfected into HepG2 cells. Subsequently, HepG2 cells were treated with different concentrations of YXY101 (0 ⁇ M, 1 ⁇ M or 4 ⁇ M) for 1 hour and then treated with TNF ⁇ (0 ng/mL or 20 ng/mL) for 30 minutes. Subsequently, Nur77, RXR ⁇ and I ⁇ B ⁇ in the cells were detected by Western Blotting. The results are shown in Figures 5A-5B.
  • Figures 5A-5B show the results of immunoblot analysis of Nur77, RXR ⁇ and I ⁇ B ⁇ in HepG2 cells transfected with different siRNAs and treated with different concentrations of YXY101 and TNF ⁇ .
  • the results showed that Nur77 siRNA effectively inhibited/knockout the expression of Nur77 in cells; and, RXR ⁇ siRNA effectively inhibited/knockout the expression of RXR ⁇ in cells; control siRNA did not affect the normal expression of Nur77 and RXR ⁇ .
  • MEF cells and Nur77-/-MEF cells i.e., MEF cells not expressing Nur77.
  • MEF cells and Nur77-/-MEF cells were treated with different concentrations of YXY101 (0 ⁇ M or 1 ⁇ M) for 1 hour and treated with TNF ⁇ (0 ng/mL or 20 ng/mL) for 30 minutes.
  • TNF ⁇ 0. ng/mL or 20 ng/mL
  • I ⁇ B ⁇ in the cells was detected by Western Blotting. The results are shown in Figure 5C.
  • MEF cells and Nur77-/-MEF cells were treated with YXY101 (0 or 1 ⁇ M) for 1 hour and then treated with TNF ⁇ (20 ng/mL) for 30 minutes. Subsequently, immunofluorescence staining was used to detect NF- ⁇ B in cells. Subunit p65. Untreated cells were used as controls. The results are shown in Figure 5D.
  • Figure 5D shows the results of immunofluorescence staining of MEF cells and Nur77-/-MEF cells treated with YXY101 and TNF ⁇ (Scale bar: 10 ⁇ m).
  • the results showed that YXY101 was able to inhibit TNF ⁇ -induced p65 nuclear translocation in MEF cells expressing Nur77; however, in Nur77-/-MEF cells, YXY101 lost the ability to inhibit p65 entry into the nucleus.
  • Example 5 YXY101 is capable of inhibiting acute inflammation in animals
  • a mouse acute hepatitis model induced by intraperitoneal injection of 80 ug/kg LPS and 200 ug/kg D-GalN for 6 hours was used.
  • ALT and AST levels in the liver, serum levels of IL-1 ⁇ and IL-6, and liver IL-1 ⁇ and IL- were observed after intraperitoneal injection of YXY101 (0.5 mg/kg) for 12 hours. 6 mRNA levels.
  • mice There were 18 wild-type mice and Nur77-knockout mice, male and weighing 18-22 g. SPF level of Experimental Animal Center of Xiamen University
  • Wild-type and Nur77 knockout mice were housed at a temperature of 23 ⁇ 1 ° C, humidity: 40-60%, natural light, free access to water, and free access to normal feed; small wild-type mice and Nur77 knocked out
  • the rats were randomly divided into 3 groups (6 in each group), which were normal control group, LPS/D-GalN model group, and YXY101 (0.5 mg/kg) group.
  • Drug configuration method YXY101-DMSO saturated solution +5% (v / v) Tween-80 + normal saline, configured to 0.05mg / ml
  • Normal control group and LPS/D-GalN-induced acute hepatitis model group Normal saline was administered intraperitoneally before the feed at 20 pm.
  • Group YXY101 The drug was administered intraperitoneally once before the feed at 20 pm, and the dose was 0.5 mg/kg.
  • the acute hepatitis model group and the YXY101 group were given 80 ug/kg LPS and 200 mg/kg D-GalN to induce acute hepatitis.
  • Figure 6A shows serum ALT and serum AST levels in mice of each treatment group.
  • Figure 6B shows the levels of serum IL-1 ⁇ and serum IL-6 in mice of each treatment group.
  • Figure 6C shows mRNA levels of liver IL-1 ⁇ and liver IL-6 in mice of each treatment group.
  • the data represent the mean ⁇ SEM of three independent experiments; ns indicates no significant difference; * P ⁇ 0.05, ** P ⁇ 0.01, *** P ⁇ 0.001 (Student's t-test).
  • Figure 6D shows the protein levels of liver I ⁇ B ⁇ in mice of each treatment group.
  • Figure 7A shows the H&E staining results (scale bar, 100 ⁇ m) of the liver of each treatment group of mice.
  • Figure 7B shows the p65 immunohistochemical staining results (scale bar, 20 ⁇ m) of the liver of mice in each treatment group.
  • YXY101 lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced hepatitis damage in wild-type and Nur77-deficient mice.
  • LPS lipopolysaccharide
  • D-GalN D-galactosamine
  • YXY101 has a Nur77-dependent resistance/inhibition effect on LPS/D-GalN-induced pneumonia and neutrophil infiltration (data not provided). Therefore, the inhibitory effect of YXY101 on acute inflammation induced by LPS/D-GalN is dependent on Nur77.
  • Example 6 YXY101 is capable of inhibiting chronic inflammation in animals
  • a mouse obesity model induced by 60% high fat diet was used.
  • serum ALT and AST levels, liver ALT and AST levels, serum IL-1 ⁇ and IL-6 levels, and liver IL-1 ⁇ and IL-6 mRNA levels were observed in YXY101 versus obese model mice. influences.
  • Wild-type and Nur77 knockout mice were housed under the conditions of temperature 23 ⁇ 1°C, humidity: 40-60%, natural light, free drinking water, and free eating; except for the normal control group, the other groups were fed as normal: 60% high-fat diet (5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5) was fed in an adaptive manner for 9 days to Reduce gastrointestinal damage caused by direct high-fat feeding. After the end of the transition period, all the groups except the normal control group were fed with a complete 60% high-fat diet. Wild type mice and Nur77 knockout mice were randomly divided into 3 groups (8 in each group), which were normal control group, obese model group, and YXY101 (0.1 mg/kg) group. Except for the normal control group, the other groups were given high-fat diet. Feed for 17 weeks.
  • Normal control group and obesity model group Normal saline was given before the feed was given at 7:00 pm, in the form of intraperitoneal injection.
  • YXY101 group daily before the feed at 7:00, once a dose, 0.1mg/kg, way: belly Cavity injection.
  • Figure 8A shows the body type, body weight, and status of adipose tissue blocks of mice in each treatment group.
  • Figure 8B shows mRNA levels of liver IL-1 ⁇ and liver IL-6 in mice of each treatment group.
  • Figure 8C shows the protein levels of liver I ⁇ B ⁇ in mice of each treatment group.
  • Fig. 8D shows the results of H&E staining of the liver of each treatment group, the results of p65 immunohistochemical staining, and the staining results of hepatic neutrophils.
  • YXY101 was able to reduce body weight by 22% in wild-type mice, while only 4% in Nur77-/- mice (Fig. 8A).
  • YXY101 significantly reduces the ability of HFD-induced obesity and elevated ALT and AST in serum in Nur77-/- mice compared to wild-type mice.
  • YXY101 administration greatly inhibited the expression and production of IL-1 and IL-6 in wild-type mice, whereas Nur77-/- mice did not.
  • YXY101 administration inhibited downregulation of I ⁇ B ⁇ protein levels in wild-type mice (Fig. 8C), inhibited p65 entry into the nucleus (Fig. 8D), inhibited hepatitis (Fig. 8D), inhibited hepatic neutrophils (Fig. 8D) and giant The accumulation of phagocytes. Nur77-/- mice otherwise. Therefore, YXY101 inhibits chronic inflammation in obese animals and is also dependent on Nur77.
  • Example 7 The anti-inflammatory and slimming activity of YXY101 is achieved by inducing the transport of TRAF2 to the mitochondria, which is also dependent on Nur77.
  • YXY101 which specifically binds to Nur77, was able to significantly reverse the degradation of I ⁇ B ⁇ caused by TNF ⁇ in cells, so MEF, HepG2 cells were used, and treatment with TNF ⁇ and YXY101 was performed. Wild-type and Nur77-deficient MEF cells were treated with TNF ⁇ (20 ng/ml, 30 min) and YXY101 (2 ⁇ M, 1 h), and the localization of Nur77 and TRAF2 in mitochondria was observed by immunofluorescence technique.
  • Fig. 9A shows the results of immunostaining.
  • TRAF2 was not significantly aggregated under the condition of TNF ⁇ alone, and TRAF2 was aggregated in a small amount under the condition of YXY101 alone, but when TNF ⁇ and YXY101 were used together, TRAF2 was clearly aggregated and localized at the mitochondria, indicating that the anti-inflammatory and weight-loss functions induced by YXY101 may be induced by promoting the transport of TRAF2 to mitochondria. Under the same treatment conditions, the transport of TRAF2 in Nur77 knockout MEF cells disappeared.
  • Fig. 9B shows the statistical results of the above phenomenon.
  • the co-localization of the experimental group using YXY101 alone was different from that of the control group, and when TNF ⁇ was added, the co-localization was greatly increased. There was a significant difference, and in the MEF cells in which Nur77 was deleted, there was no significant difference between the control group and the experimental group.
  • HepG2 cells were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M, 1 h/9h), mitochondria were isolated, and Nur77, TRAF2, p62, LC3, PARP, Hsp60 levels in whole cell proteins and mitochondrial proteins were detected by immunoblotting. .
  • Figure 9C shows that YXY101 can significantly increase the content of Nur77, p62, TRAF2, and LC3 on mitochondria, further demonstrating that YXY101 can promote the transport of Nur77, p62/TRAF2, and LC3 to mitochondria.
  • Example 8 YXY101 exerts anti-inflammatory and slimming activity by inducing interaction between Nur77 and TRAF2 mitochondria
  • FIG. 10A shows that HepG2 cells, IP:TRAF2, were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and whether Nur77 interacts with TRAF2 was detected by immunoblotting.
  • TNF ⁇ 20 ng/ml
  • YXY101 2 ⁇ M
  • the results showed that at the cellular level, after treatment with TNF ⁇ and YXY101, the interaction of TNF ⁇ alone could not induce the interaction between the two, and when TNF ⁇ and YXY101 were added, the interaction intensity of Nur77 and TRAF2 was significantly enhanced.
  • Figure 10B shows that the Flag-TRAF2 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and the co-localization of endogenous Nur77 and exogenous Flag-TRAF2 was detected by immunostaining.
  • TNF ⁇ (20 ng/ml
  • YXY101 (2 ⁇ M)
  • the results showed that the in vitro purified GST-TRAF2 and His-Nur77-LBD proteins exhibited strong interactions under the action of YXY101.
  • Figure 1C shows that the Flag-TRAF2 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and the co-localization of endogenous Nur77 and exogenous Flag-TRAF2 was detected by immunostaining. The results showed that under the conditions of TNF ⁇ and YXY101 co-treatment, exogenous Flag-TRAF2 and endogenous Nur77 could form a prominent co-localization structure outside the nucleus. Further, Fig.
  • 10D shows that the Flag-TRAF2, GFP-Nur77 plasmid was transfected in HepG2 cells, and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and the total of Flag-TRAF2, GFP-Nur77, and mitochondria were detected by immunostaining technique. Positioning situation. The results showed that this extranuclear colocalization structure colocalized with mitochondria, indicating that the interaction between Nur77 and TRAF2 induced by YXY101 is mitochondria.
  • the anti-inflammatory and weight-reducing mechanisms of YXY101 are not only related to the transport of TRAF2, but also induce the interaction between Nur77 and TRAF2 on the mitochondria. Based on this, we can screen for drug molecules with potential anti-inflammatory and weight-loss activities by detecting the interaction between Nur77 and TRAF2 and their mitochondrial localization.
  • Example 9 The anti-inflammatory and slimming activities of YXY101 are closely related to the Nur77-dependent autophagy process.
  • the anti-inflammatory mechanism may be related to the inhibition of TNF ⁇ -induced over-activation of the NF- ⁇ B pathway, or may be achieved by autophagy to clear damaged mitochondria. Accordingly, we examined LC3 proteins that are closely related to the autophagy process.
  • Figure 11A shows that the Flag-TRAF2, GFP-LC3 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M). The co-localization of Flag-TRAF2, GFP-LC3 and mitochondria was detected by immunostaining.
  • Figure 11B shows the transfection of RFP-LC3, GFP-Nur77 plasmid in HepG2 cells, and treatment with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), using immunostaining to detect colocalization of RFP-LC3, GFP-Nur77 and mitochondria .
  • FIG. 11C shows that wild-type and Nur77-deficient MEF cells were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and Nur77, LC3 and mitochondria were detected by immunostaining. Co-location situation. The results showed that the phenomenon of LC3 localization to mitochondria disappeared after knocking out Nur77.
  • Figure 11D shows the statistical results of Nur77, LC3, mitochondria colocalization in the above treated cells.
  • YXY101 can promote the interaction between Nur77 and LC3, promote the process of autophagy, and then achieve anti-inflammatory and weight loss effects. Based on this, we can screen for drug molecules with potential anti-inflammatory and weight-loss activities by detecting the interaction of Nur77 with LC3 and its mitochondrial localization, or detecting autophagy.
  • Example 10 YXY101 promotes the interaction between Nur77 and p62, and removes damaged mitochondria, thereby achieving anti-inflammatory and weight-reducing effects.
  • Figure 12A shows that the Myc-Nur77, Flag-p62 plasmid was transfected in HepG2 cells and treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), IP: Flag, and Myc interacting with Flag-p62 was detected by immunoblotting. -Nur77. The results showed that YXY101 significantly induced the interaction between Nur77 and p62.
  • Figure 12B shows that HepG2 cells were treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of Nur77 and p62 was detected by immunostaining.
  • Figure 12C shows that the GFP-LC3 plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of GFP-Nur77 and p62 was detected by immunostaining.
  • Figures 12B-C demonstrate the validation of endogenous or exogenous Nur77, p62 colocalization under the induction of YXY101.
  • YXY101 can promote the interaction of Nur77 with LC3 and P62, respectively, thereby promoting autophagy, promoting the damage of damaged mitochondria, and finally achieving anti-inflammatory and weight-reducing effects. Based on this, we can screen the drug molecules with potential anti-inflammatory and weight-loss activities by detecting the interaction of Nur77 with LC3 and p62.
  • Example 11 YXY101 promotes ubiquitination of Nur77, thereby achieving anti-inflammatory and weight-reducing effects
  • Figure 13A shows the transfection of Flag-TRAF2, Myc-Nur77, HA-Ub plasmids in HepG2 cells, treatment with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), IP: Myc, detection of ubiquitination of Nur77 by immunoblotting happening.
  • TNF ⁇ 20 ng/ml
  • YXY101 2 ⁇ M
  • IP Myc
  • Figure 13B shows that Myc-Nur77, HA-Ub plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), mitochondria were isolated, IP: Myc, and Nur77 pan in mitochondria was detected by immunoblotting. The situation of vegetarianization.
  • Figure 13B demonstrates that YXY101 induced ubiquitination modification is performed on mitochondria.
  • Figure 13C shows that GFP-Nur77, HA-Ub plasmids were transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of GFP-Nur77 and HA-Ub was detected by immunostaining.
  • Figure 13D shows that GFP-Nur77 plasmid was transfected in HepG2 cells, treated with TNF ⁇ (20 ng/ml) and YXY101 (2 ⁇ M), and co-localization of GFP-Nur77 and Ub was detected by immunostaining.
  • Figures 13C-13D illustrate that YXY101 can induce colocalization of Nur77 and Ub onto mitochondria.
  • Related data indicate that ubiquitination on the mitochondria of Nur77 is associated with autophagy.
  • YXY101 can induce ubiquitination of Nur77, interaction of Nur77 with p62, and interaction of Nur77 with LC3, ultimately eliciting autophagy, clearing damaged mitochondria, and inhibiting inflammation.
  • Example 12 Screening for new active molecules by detecting the interaction of Nur77 with p62, TRAF2, LC3, Ub
  • the active molecule XS0284 was screened according to the inventive method described in Examples 1-6. First, as shown in Figure 14A, using Biacore T200 instrument, XS0284 was initially screened by SPR to specifically bind Nur77 protein. Figure 15B was further tested with XS0284 with gradient concentration (0.15 ⁇ M, 0.3 ⁇ M, 0.625 ⁇ M, 1.25 ⁇ M, 2.5 ⁇ M) and Nur77. The binding strength, Figure 15B shows that the dissociation constant (Kd) of XS0284 in combination with Nur77-LBD is 386 nM. Figure 16A shows that XS0284 is able to significantly reverse TNF ⁇ down-regulation of I ⁇ B ⁇ with significant anti-inflammatory activity.
  • Kd dissociation constant
  • Figure 17A shows that XS0284 is capable of inducing the interaction of Nur77 and p62.
  • Figure 17B shows that XS0284 is similar to YXY101 in that it can attenuate the ubiquitination of TRAF2.
  • Figure 17C shows that XS0284 can induce colocalization of GFP-Nur77 with Flag-TRAF2 on mitochondria.
  • Figure 17D shows that XS0284 is capable of inducing colocalization of GFP-Nur77 with RFP-LC3.
  • Figure 17E shows that XS0284 is capable of inducing colocalization of GFP-Nur77, Flag-p62 and Ub.
  • Examples 1-6 we used the method of Examples 1-6 to detect the specific binding of the compound to Nur77; the detection compound reversed the activity of TNF ⁇ to down-regulate I ⁇ B ⁇ ; and whether the compound can induce the interaction of Nur77 with TRAF2, p62, LC3, Ub or Positioning; detecting whether the compound induces mitochondrial transport of Nur77, TRAF2, p62, LC3, Ub; and detecting whether the compound induces autophagy and clears damaged mitochondria; thereby screening compound XS0284; it has good activity, is potential anti-inflammatory, and loses weight Active drug molecule.
  • YXY101 derivative has a stronger and more specific function of inhibiting the biological effect of TNF ⁇ than YXY101
  • Figure 16A shows I ⁇ B ⁇ and PARP in MDA-MB-231 cells treated with YXY101 and different YXY101 derivatives XS0284, XS0285, XS0286, XS0287, XS0335, XS0366, XS0260, XS0394, XS0395, XS0419, XS0420, XS0421 and TNF ⁇ .
  • Figure 16B MDA-MB treated with YXY101 and different YXY101 derivatives XS0284-4, XS0284, XS0077, XS0503, XS0486, XS0419, XS0474, XS0462, XS0285, XS0394, XS0454, XS0455, XS0462, XS0473, XS0474, XS0480 and TNF ⁇ are shown. Results of immunoblot analysis of I ⁇ B ⁇ and PARP in -231 cells.
  • Figure 16C shows MDA-MB treated with YXY101 and different YXY101 derivatives XS0284, XS0285, XS0335, XS0394, XS0418, XS0419, XS0454, XS0455, XS0462, XS0502, XS0503, XS0504, XS0506, XS0507, XS0508, XS0077 and TNF ⁇ .
  • Figures 16A-16C show the results of immunoblot analysis of I ⁇ B ⁇ and PARP in cells treated with the same concentration of YXY101 and TNF ⁇ .
  • TNF ⁇ was able to induce degradation of I ⁇ B ⁇ in cells; YXY101 was able to inhibit TNF ⁇ -induced degradation of I ⁇ B ⁇ .
  • the red font represents a derivative having a stronger anti-inflammatory effect than YXY101, including XS0284, XS0394, XS0077, XS0503, XS0486, XS0462, XS0474, XS0418, XS0419 and the like.
  • YXY101 induces apoptosis by inducing strong PARP cleavage, making YXY101 difficult to use as an anti-inflammatory and anti-obesity drug with no toxic side effects or low side effects.
  • the YXY101 derivative obtained by our method has stronger anti-inflammatory effect and less toxic side effects than YXY101.
  • XS0284 is a typical example, which inhibits TNF ⁇ -induced degradation of I ⁇ B ⁇ more significantly than YXY101. At the same time, it does not cause PARP cleavage, has weak toxic side effects, and has better drug-forming properties.
  • Compounds similar to XS0284 include XS0394, XS0503, XS0486, XS0462, XS0474, XS0418, XS0419. These results indicate that compounds with greater activity than YXY101 and lower cytotoxicity can be screened using our method.
  • Example 14 Application of a method for detecting the colocalization of Nur77 with TRAF2 or P62 to further screen a reliable anti-inflammatory compound targeting Nur77
  • Figure 18A shows the results of immunofluorescence staining of Hela cells treated with XS0077 and TNF ⁇ . HepG2 cells were treated with XS0077 (2 ⁇ M) for 3 hours and then treated with TNF ⁇ (20 ng/mL) for 30 minutes. Subsequently, the colocalization of Nur77 and Traf2 in the cells was detected by immunofluorescence staining.
  • Figure 18B shows the results of immunofluorescence staining of Hela cells treated with XS0503 and TNF ⁇ . Hela cells were treated with XS0503 (2 ⁇ M) for 3 hours and then treated with TNF ⁇ (20 ng/mL) for 30 minutes.
  • Figure 15 shows the results of experiments in which the binding of different compounds (XS0284, XS0394, XS0503, XS0486, XS0462, XS0474, XS0418, XS0419) to Nur77-LBD was detected using a Biacore T200 instrument.
  • the dissociation constant (Kd) of the compound XS0284 and Nur77-LBD was 386 nM; the dissociation constant (Kd) of the binding of XS0394 to Nur77-LBD was 1.26 ⁇ M; the dissociation constant of the compound XS0418 combined with Nur77-LBD (Kd) ) is 3.67 ⁇ M; the dissociation constant (Kd) of compound XS0462 and Nur77-LBD is 2.07 ⁇ M; the dissociation constant (Kd) of compound XS0474 and Nur77-LBD is 2.60 ⁇ M; the solution of compound XS0486 and Nur77-LBD The off-constant (Kd) was 1.20 ⁇ M; the dissociation constant (Kd) of the compound XS0419 bound to Nur77-LBD was 404 nM; the dissociation constant (Kd) of the compound XS0503 bound to Nur77-LBD was 1.63 ⁇ M.
  • the method of detecting compounds that specifically bind to Nur77 by SPR further screening for compounds having anti-inflammatory-promoting autophagy activity is reliable, and the selected compounds have strong target specificity and specificity. Sex.
  • Example 17 XS0284 has a less toxic side effect on cells than YXY101
  • FIG. 19A shows the results of immunoblot analysis of PARP in MDA-MB-231 cells treated with YXY101 and XS0284 and TNF ⁇ .
  • Figure 19B shows the results of immunoblot analysis of PARP in NCL-H292 cells treated with YXY101 and XS0284 and TNF ⁇ .
  • Figure 19C shows the inhibition rate of proliferation of HepG2 cells by compounds YXY101 and XS0284.
  • JC-1 can accumulate in the matrix of mitochondria, and the formed polymer can produce red fluorescence; when the mitochondrial membrane potential is low, JC-1 can not accumulate in the matrix of mitochondria, but The monomer form is present, producing green fluorescence. Therefore, the change in the mitochondrial membrane potential can be detected by detecting the transition of the fluorescent color.
  • MDA-MB231 cells after treatment with 5 ⁇ M of drug for 24 hours, the mitochondrial membrane potential was stained with JC-1, and the expression of FITC and PE was detected by flow cytometry to detect the mitochondrial membrane potential of the cells. It was found that YXY101 caused The mitochondrial membrane potential is reduced, while XS0284 has almost no effect.
  • Example 18 The acute side effects of the drug of XS0284 are lower than YXY101
  • mice were observed for food intake, drinking water, spontaneous activity, mental state, limb activity, bowel quality, hair gloss, etc., and detailed records of possible toxicity Should be changed and started and disappeared at the time. Histopathological observation of the tissue morphology of the heart, liver, small intestine, white fat and kidney.
  • Figure 20A shows the gavage group: 1) The cardiomyocytes of the blank group have clear boundaries, many nuclei and obvious nuclei, and the myocardial fibers are arranged neatly and clearly; the number of hepatic sinus in the liver tissue is clear and clear, and the hepatic lobule boundary is obvious; The intestinal tissue is clearly defined, the cells are full, the intestinal villi are arranged neatly and the boundaries are obvious; the adipose tissue cells are clearly defined and arranged neatly.
  • the myocardial cells and myocardial fibers of the YXY101 group were disorderly arranged; the hepatic lobule boundary was blurred, the cytoplasm was reduced, and most of the hepatocytes died; the intestinal tissue was swollen, the cell death increased, the inflammatory reaction was obvious, and the intestinal villi were found.
  • the arrangement is disordered; the adipose tissue cells are disorderly arranged and individual fibers are broken.
  • the XS0284 group had relatively clear cardiac cardiomyocyte boundaries, and the myocardial fibers were relatively neatly arranged with obvious boundaries.
  • the hepatocytes in the liver tissue had obvious boundaries and clear structure; the intestinal tissue was clear, the cells were full, and the intestinal villi were arranged. It is neat and has obvious boundaries; the adipose tissue cells have clear boundaries, neatly arranged, and reduced fiber breakage. It can be concluded that the derivative XS0284 group showed a weaker toxic effect on the liver and small intestine than YXY101.
  • Figure 20B shows the intraperitoneal injection group.
  • the cardiomyocytes of the blank group have clear boundaries, many nuclei and obvious nuclei, and the myocardial fibers are arranged neatly and clearly; the number of hepatic sinus in the liver tissue is clear and clear, and the hepatic lobule is clearly defined; The cell boundaries are clear and well-arranged; the glomeruli of the kidney tissue are clearly defined, the cells are full, the cells are neatly arranged, and the boundaries are obvious.
  • the myocardial cells and myocardial fibers of the YXY101 group were disorderly arranged; the hepatic lobule boundary was basically obvious, and the hepatic cells were normal; but the adipose tissue cells were disorderly arranged and a large number of inflammatory cells infiltrated; The boundary of the ball is blurred, and there are a large number of inflammatory cells infiltrating, and the arrangement is not neat.
  • the cardiomyocytes of XS0284 group have relatively clear boundary, and the myocardial fibers are relatively neatly arranged and the boundaries are obvious.
  • the liver cells have obvious boundary and clear structure; the adipose tissue cells have clear boundaries, neatly arranged, and fiber breakage. Reduced; renal tissue glomerular boundaries are clear, cells are full, with a small amount of inflammatory cell infiltration. It can be concluded that the XS0284 group of the intraperitoneal injection group showed a weaker toxicity to kidney and fat than YXY101.
  • Example 19 XS0284 has a milder effect of inhibiting obesity than YXY101
  • YXY101 0.1 mg/kg
  • XS0284 0.1 mg/kg high fat-induced obese mice were intraperitoneally injected for one week.
  • the results showed that YXY101 can significantly reduce body weight and improve HFD-induced fatty liver, but the body weight loss caused by excessive weight loss, and XS0284 also has a significant weight loss effect, but its weight loss effect is more mild, the mouse did not show Significantly thinning, as can be seen from the body weight curve in Figure 21C, XS0284 is more gentle and reduces body weight at a slightly slower rate. On the basis of this, it can be seen from Fig. 21A that XS0284 can also effectively improve fatty liver.
  • Nur77 is the target. Virtual screening of marine natural products resulted in some well-bound compounds, as shown below.
  • the steps of computer-assisted virtual screening in this example are as follows: download protein eutectic structure PDB ID: 3V3Q, protein pretreatment, hydrogenation, removal of crystal water molecules and small molecules of glycerol in crystal structure; ligands selected, ligands Center, 12angstrom size to build the dock grid file; run Glide docking, select the pre-built grid grid file and the processed Seaweed Metabolism Database for docking; according to the docking score and The interaction between the compound and the protein receptor is a comprehensive evaluation of the docking results.
  • An example of a schematic diagram of the simulated combination is shown in Figure 22.
  • Example 24 Preparation of Compounds XS0366, XS0434-XS0438, XS0440, XS0441, XS0443, XS0463 and XS0464
  • the present invention also synthesizes the following compounds:
  • the compound YXY101 (50 mg, 0.11 mmol) was weighed into a 25 ml reaction flask, and 4 ml of acetone was added thereto to stir and dissolve, and then a drop of concentrated hydrochloric acid was added as a catalyst, and the reaction was carried out at room temperature for 12 hours. The reaction was quenched, and the solvent was evaporated, evaporated, evaporated, evaporated
  • the compound YXY101 (50 mg, 0.11 mmol) was stirred and dissolved in 2 mL of deuterated methanol solvent, followed by addition.
  • Sodium borohydride (44 mg, 1.1 mmol) was added, and the mixture was stirred at room temperature for 30 min. 1 mol/L HCl (1 mL) was quenched, 9 mL of pure water was added, and extracted with dichloromethane (3 mL each time). The organic layer was collected and dried over anhydrous sodium sulfate.
  • the compound XS0419 (50.1 mg) was obtained as a white solid.
  • tripterine 135.2 mg, 0.3 mmol was stirred and dissolved in 2 mL of DMF, followed by sodium hydrogencarbonate (138.6 mg, 1.65 mmol), and ethyl bromide (234 ⁇ L, 0.15 mmol) was stirred at room temperature for 12 hours.
  • the reaction was quenched with 1 mol/L HCl (1 mL), 9 mL of pure water was added, and extracted with ethyl acetate three times (15 mL each time), the organic layer was collected, dried over anhydrous sodium sulfate, and the organic solvent ethyl acetate was removed by distillation under reduced pressure.
  • the ester was obtained as an orange-red solid mixture.
  • scutellarin 250 mg, 0.54 mmol was dissolved in 20 mL of tetrahydrofuran, followed by LiAlH 4 (1.2 mL, 1.1 mmol), stirred at room temperature for 2 h, and quenched with 10 mL of deionized water, 1 mol/L HCl (5 mL) The mixture was acidified, and extracted with ethyl acetate (3 mL). The organic layer was collected and dried over anhydrous sodium sulfate.
  • the present invention also synthesizes the following compounds:
  • the preparation method is exemplified by XS0439: Compound YXY101 (100 mg, 0.22 mmol) was dissolved in dichloromethane (4 mL) under stirring. 7-Methoxy substituted hydrazine (65.3 mg, 0.44 mmol) was added, and then aluminum trichloride hexahydrate (5.3 mg, 0.022 mmol) was added, and the reaction was stirred at room temperature for 5 hours. The reaction was stopped, and deionized water (15 mL) was added to the reaction mixture and extracted three times with ethyl acetate.
  • compound XS0486 Taking compound XS0486 as an example: firstly, compound YXY101 (50 mg, 0.11 mmol) was dissolved in dioxane (600 ⁇ L), triethylamine (150 ⁇ L, 0.33 mmol) was added, and then the reaction bottle was added with dioxane (100 ⁇ L).
  • the compound YXY101 (50 mg, 0.11 mmol) was weighed into a 50 ml round bottom flask, dissolved in 2 ml of DCM, transferred to -78 ° C, stirred, and then subjected to DAST (150 ul, 10 eq), and reacted at -78 ° C for 1 h.
  • the system was separated and purified by silica gel column chromatography to give an orange-red solid.
  • the compound YXY101 (50 mg, 0.11 mmol) was weighed into a 25 ml thick-walled pressure tube, and tetrabutylammonium bromide (TBAB, 17.5 mg, 0.05 mmol) was added thereto, and dissolved in 2 ml of dichloromethane (DCM). Then, 5% NaOH (180 ⁇ l) was added dropwise, and the reaction was carried out for 30 min at room temperature, and then transferred to an oil bath at 50 ° C, and 2,3,4,6-tetraacetoxy- ⁇ -D-glucopyran bromide was added dropwise.
  • TBAB tetrabutylammonium bromide
  • the compound-dichloromethane solution (57 mg-1 ml, 0.138 mmol) was reacted at 50 ° C for 12 h, the reaction was stopped, a large amount of water and saturated brine were added, and the mixture was extracted with DCM three times, and the organic phases were combined and dried over anhydrous Na 2 SO 4 The organic phase was concentrated under reduced pressure and purified with silicagel eluting
  • reaction solution was poured into a large amount of ice directly stop the reaction, the aqueous phase extracted with DCM three times, the combined organic phases were washed with saturated NaHCO 3 The organic phase was washed, dried over anhydrous Na 2 SO 4 the organic phase, the organic phase was concentrated under reduced pressure, neutralized with acetic acid
  • XS0077 50 mg, 0.11 mmol was weighed into a 25 ml thick-walled pressure tube, and tetrabutylammonium bromide (TBAB, 17.5 mg, 0.05 mmol) was added thereto, and dissolved in 2 ml of dichloromethane (DCM). Then add 5% NaOH (180 ⁇ L) dropwise, react at room temperature for 30 min, transfer to 50 ° C oil bath, add 2,3,4,6-tetraacetoxy- ⁇ -D-glucopyran bromide dropwise.
  • TBAB tetrabutylammonium bromide

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Steroid Compounds (AREA)

Abstract

本申请涉及孤儿核受体Nur77的配体及其用途。特别地,本申请涉及如式I所示的化合物用作孤儿核受体Nur77的配体的用途。此外,本申请还涉及如式I所示的化合物用于预防或治疗与孤儿核受体Nur77相关的疾病的用途,

Description

孤儿核受体Nur77的配体及其用途 技术领域
本申请涉及医药领域和生物领域。具体而言,本申请涉及一种新的Nur77的配体及其用途。本申请还涉及如式I-式V所示的化合物用作孤儿核受体Nur77的配体的用途。本申请还涉及如式I-式V所示的化合物用于预防或治疗与孤儿核受体Nur77相关的炎症疾病的用途。本申请还涉及一种具有抗炎和/或治疗肥胖症活性的药物的筛选方法。
背景技术
孤儿核受体Nur77,其也被称为NGFIB(神经生长因子IB)或孤儿核受体TR3,是癌症、新陈代谢和炎症性疾病发生发展过程中一个的关键调节子。作为一类即刻早期应答基因,Nur77在大量细胞过程中起着重要作用,包括由细胞分裂素、激素、压力、代谢和凋亡信号等不同刺激引起的细胞生存、凋亡、炎症和自噬过程(Pei L等人(2006),″Regulation of macrophage inflammatory gene expression by the orphan nuclear receptor Nur77″,Mol.Endocrinol.20(4):786-94;Zhang XK(2007),″Targeting Nur77 translocation″,Expert Opin.Ther.Targets 11(1):69-79)。近来,研究主要关注于它在炎性疾病和癌症中的强效抗炎作用。已显示,Nur77在发炎的人体滑膜组织、癌细胞、牛皮癣患者、动脉粥样硬化患者、多发性硬化患者中异常表达,并且其表达能够被一些细胞因子快速有效地诱导。遗传研究已揭示了Nur77在控制炎症反应中的关键作用,尤其是在动脉粥样硬化(Hamers,A.A.,et al.Bone marrow-specific deficiency of nuclear receptor Nur77 enhances atherosclerosis.Circulation research 110,428-438,2012;Hanna,R.N.,et al.NR4A1(Nur77)deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis.Circulation research 110,416-427,2012)、肥胖症(Perez-Sieira,S.,et al.Female Nur77-deficient mice show increased susceptibility to diet-induced obesity.PloS one 8,e53836,2013)、糖尿病(Chao,L.C.,et al.Insulin resistance and altered systemic glucose metabolism in mice lacking Nur77.Diabetes 58,2788-2796,2009)、哮 喘(Kurakula,K.,et al.Nuclear Receptor Nur77 Attenuates Airway Inflammation in Mice by Suppressing NF-kappaB Activity in Lung Epithelial Cells.Journal of immunology 195,1388-1398,2015)、关节炎(De Silva,S.,et al.Reduction of the incidence and severity of collagen-induced arthritis by constitutive Nur77 expression in the T cell lineage.Arthritis and rheumatism 52,333-338,2005)和炎症性肠病(Hamers,A.A.,et al.Deficiency of Nuclear Receptor Nur77 Aggravates Mouse Experimental Colitis by Increased NFkappaB Activity in Macrophages.PloS one 10,e0133598,2015;Wu,H.,et al.NUR77 exerts a protective effect against inflammatory bowel disease by negatively regulating the TRAF6/TLR-IL-1R signalling axis.The Journal of pathology 238,457-469,2016)患者中的作用。
鉴于Nur77在多种细胞过程以及多种疾病过程(例如,炎症(如与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,和与糖尿病相关的炎症)和癌症(如三阴乳腺癌))中的重要作用,其已成为开发此类疾病的新疗法的重要靶点。因此,本领域需要寻找有效的、特异性结合Nur77的配体,以开发用于治疗炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,和与糖尿病相关的炎症)和癌症(如三阴乳腺癌)的新疗法。本申请的发明人首次发现的Nur77参与抑制炎症和诱导自噬作用的机理。本申请的发明人发现某些化合物能通过结合Nur77,并诱导Nur77依赖的抑制炎症和自噬作用。所述化合物通过促进Nur77从核内转运到线粒体上,与肿瘤坏死因子受体相关因子2(TRAF2,一种炎症信号通路中重要的支架蛋白和关键的泛素连接酶)相互作用。所述相互作用是通过TRAF2上的LXXLL基序介导的。该相互作用不仅抑制了TRAF2的泛素化还诱导了Nur77在K63位的泛素化。在炎症状态下,泛素化的Nur77在线粒体上与p62/SQSTM1泛素集合域相互作用来增强线粒体自噬的敏感性。LC3(一种自噬相关蛋白)从而与修饰后的Nur77特异的相互作用,保证受损的线粒体能被选择性清除。本申请的发明人通过上述所阐明的机理,建立一套筛选Nur77依赖的抑制炎症和诱导自噬作用的化合物的方法,并对筛选得到的化合物的生物学活性进行了分析。
海洋生态环境由于与陆地差异较大,因此,导致海洋生物体内小分子代谢物的生物合成途径也迥然不同。因此,海洋天然产物拥有大量结构独特的化合物并具有许多各种各样的生物学活性。比如,萜类和甾体化合物等具有较好的抗肿瘤以及免疫调节活性等。 本申请的发明人通过大量的实验研究发现了一类新的五环三萜类化合物YXY101及其衍生物,其能够结合Nur77从而调控线粒体活性,并发挥Nur77依赖的抑制炎症和促进自噬(特别是线粒体自噬)作用。从而,本申请提供了有前景的化合物,其可用于开发治疗炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,和与糖尿病相关的炎症)和肥胖症的新药物或新疗法。
发明内容
在本发明中,除非另有说明,否则本文中使用的科学和技术名词具有本领域技术人员所通常理解的含义。并且,本文中所用的细胞培养、分子遗传学、核酸化学、免疫学实验室操作步骤均为相应领域内广泛使用的常规步骤。同时,为了更好地理解本发明,下面提供相关术语的定义和解释。
如本文中所使用的,术语“互变异构体”是指,因分子中某一原子在两个位置迅速移动而产生的官能团异构体。此类互变异构体的典型实例为酮-烯醇互变异构体。本发明所述的化合物可以以互变异构体形式存在,并因此涵盖所有可能的互变异构体,及其任何组合或任何混合物。
如本文中所使用的,术语“立体异构体”是指,分子中原子或原子团的连接次序相同,但空间排列不同而引起的异构体。在本申请中,化合物的“立体异构”分为构象和构型异构,而构型异构还分为顺反异构和旋光异构。因此,在本申请中,“立体异构体”包括所有可能的光学异构体和非对映异构体,以及其任何组合,例如外消旋体(外消旋混合物),单一对映异构体,非对映异构体混合物,单一非对映异构体。例如,当本发明所述的化合物含有烯烃双键时,除非特别说明,否则其包括顺式异构体和反式异构体,以及其任何组合。
如本文中所使用的,术语“药学上可接受的盐”是指,(1)本发明化合物中存在的酸性官能团(例如-COOH、-OH、-SO3H等)与适当的无机或者有机阳离子(碱)形成的盐,例如本发明化合物与碱金属或碱土金属形成的盐、本发明化合物的铵盐,和本发明化合物与含氮有机碱形成的盐;以及(2)本发明化合物中存在的碱性官能团(例如-NH2等)与适当的无机或者有机阴离子(酸)形成的盐,例如本发明化合物与无机酸或有机羧酸形成的盐。
因此,本发明化合物的“药学上可接受的盐”包括但不限于,碱金属盐,如钠盐、钾盐、锂盐等;碱土金属盐,如钙盐、镁盐等;其他金属盐,如铝盐、铁盐、锌盐、铜盐、镍盐、钴盐等;无机碱盐,如铵盐;有机碱盐,如叔辛基胺盐、二苄基胺盐、吗啉盐、葡糖胺盐、苯基甘氨酸烷基酯盐、乙二胺盐、N-甲基葡糖胺盐、胍盐、二乙胺盐、三乙胺盐、二环己基胺盐、N,N’-二苄基乙二胺盐、氯普鲁卡因盐、普鲁卡因盐、二乙醇胺盐、N-苄基-苯乙基胺盐、哌嗪盐、四甲基胺盐、三(羟甲基)氨基甲烷盐;氢卤酸盐,如氢氟酸盐、盐酸盐、氢溴酸盐、氢碘酸盐等;无机酸盐,如硝酸盐、高氯酸盐、硫酸盐、磷酸盐等;低级烷磺酸盐,如甲磺酸盐、三氟甲磺酸盐、乙磺酸盐等;芳基磺酸盐,如苯磺酸盐、对苯磺酸盐等;有机酸盐,如醋酸盐、苹果酸盐、富马酸盐、琥珀酸盐、柠檬酸盐、酒石酸盐、草酸盐、马来酸盐等;氨基酸盐,如甘氨酸盐、三甲基甘氨酸盐、精氨酸盐、鸟氨酸盐、谷氨酸盐、天冬氨酸盐等。
如本文中所使用的,术语“药学上可接受的酯”是指,当本发明化合物存在羧基时,其与醇发生酯化反应而形成的酯;当本发明化合物存在羟基时,其与有机酸、无机酸、有机酸盐等发生酯化反应而形成的酯。酯在酸或者碱存在的条件下,可以发生水解反应生成相应的酸或醇。
如本文中所使用的,术语“C1-6烷基”表示直链或支链的含有1-6个碳原子的烷基,如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、异戊基、2-甲基丁基、新戊基、1-乙基丙基、正己基、异己基、3-甲基戊基、2-甲基戊基、1-甲基戊基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、1,2-二甲基丁基、1,3-二甲基丁基、2,3-二甲基丁基、2-乙基丁基、1,2-二甲基丙基等。C1-6烷基的优选实例包括C1-5烷基、C1-4烷基、C1-3烷基。本发明所述的“C1-4烷基”表示直链或支链的含有1-4个碳原子的烷基,其包括但不限于上述实例中的含有1-4个碳原子的具体实例。
如本文中所使用的,术语“链烯基”是指含有至少一个碳碳双键的直链或支链烃基,其典型的实例为C2-10链烯基,例如C2-6链烯基或C2-4链烯基。具体的实例包括但不限于:乙烯基、丙烯基、2-丙烯基、丁烯基、2-丁烯基、丁二烯基、戊烯基、2-甲基-丁烯基、3-甲基-丁烯基、1,3-戊二烯基、1,4-戊二烯基、己烯基、2-乙基-丁烯基、3-甲基-戊烯基、4-甲基-戊烯基、1,3-己二烯基、1,4-己二烯基、1,5-己二烯基等。
如本文中所使用的,术语“炔基”是指至少含有一个碳碳三键的直链或支链烃基, 其典型的实例为C2-10炔基,例如C2-6炔基或C2-4炔基。具体的实例包括但不限于:乙炔基、丙炔基、2-丙炔基、丁炔基、2-丁炔基、2-甲基-丙炔基、丁二炔基、戊炔基、2-甲基-丁炔基、3-甲基-丁炔基、1,3-戊二炔基、1,4-戊二炔基、己炔基、2-乙基-丁炔基、3-甲基-戊炔基、4-甲基-戊炔基、1,3-己二炔基、1,4-己二炔基、1,5-己二炔基等。
如本文中所使用的,术语“环烷基”是指单环饱和烷基,其典型的实例为3-8元环烷基,例如3元、4元、5元、6元、7元或8元环烷基。如本文中所使用的,术语“3-8元环烷基”是指含有3-8个碳原子的环烷基。具体的实例包括但不限于:环丙基、环丁基、环戊基、环己基、环庚基、环辛基等。
如本文中所使用的,术语“杂环烷基”是指含有至少1个至多4个(例如1、2、3或4个)选自N、O和S的杂原子的环烷基,其中“环烷基”的定义如前文所述,其典型的实例为3-8元杂环烷基,例如3元、4元、5元、6元、7元或8元杂环烷基。如本文中所使用的,术语“3-8元杂环烷基”是指含有3-8个碳原子的杂环烷基。如本文中所使用的“氧代3-8元环烷基”是指所述杂原子为O的如前文所定义的3-8元杂环烷基。具体的实例包括但不限于:环氧乙基、氧代环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、硫吗啉基等。
如本文中所使用的,术语“芳基”是指芳香族基团,其典型实例为6-14元芳基,例如6-10元芳基。如本文中所使用的,术语“6-14元芳基”是指含有6-14个碳原子的单环、双环或多环芳香族基团,包括例如6-8元芳基和8-14元稠环芳基。6-8元芳基是指含有6-8个碳原子的芳基,例如苯基等。8-14元稠环芳基是指含有8-14个环碳原子、由两个或两个以上环状结构彼此共用两个相邻的碳原子所形成的不饱和的具有芳香性的稠环基团,具体实例包括但不仅限于:萘、蒽、菲等。术语“6-10元芳基”是指含有6-10个碳原子的芳香族基团,其包括但不限于上述实例中环原子个数为6-10个的芳香族基团。
如本文中所使用的,术语“芳基-C1-6烷基”是指以芳基-C1-6烷基-方式形成的基团,其中“芳基”和“C1-6烷基”的定义各自如前文所述。
如本文中所使用的,术语“C1-6烷氧基”是指以C1-6烷基-O-方式形成的基团,其中“C1-6烷基”的定义如前文所述。
如本文中所使用的,术语“C1-6烷氨基”是指以C1-6烷基-NH-方式形成的基团,其 中“C1-6烷基”的定义如前文所述。
如本文中所使用的,术语“C1-6烷硫基”是指以C1-6烷基-S-方式形成的基团,其中“C1-6烷基”的定义如前文所述。
如本文中所使用的,术语“C1-6烷酰基”是指以C1-5烷基-C(O)-方式形成的基团,其中“C1-5烷基”的定义如前文所述。
如本文中所使用的,术语“C1-6烷氧羰基”是指以C1-6烷基-O-C(O)-方式形成的基团,其中“C1-6烷基”的定义如前文所述。
如本文中所使用的,术语“C1-6烷氧羰基-C1-6烷基”是指以C1-6烷基-O-C(O)-C1-6烷基-方式形成的基团,其中“C1-6烷基”的定义如前文所述。
如本文中所使用的,术语“3-8元环烷基-氨酰基”是指以3-8元环烷基-NHC(O)-方式形成的基团,其中“3-8元环烷基”的定义如前文所述。
如本文中所使用的,术语“卤素”包括例如氟原子、氯原子、溴原子和碘原子。
如本文中所使用的,术语“6-15元杂芳基”是指含有6-15个环原子且其中至少一个为杂原子的、具有芳香性的基团。6-15元杂芳基包括“5-8元杂芳基”,例如“5-7元杂芳基”、“5-6元杂芳基”等。“5-8元杂芳基”具体实例包括但不限于,呋喃基、噻吩基、吡咯基、噻唑基、异噻唑基、噻二唑基、噁唑基、异噁唑基、噁二唑基、咪唑基、吡唑基、1,2,3-三唑基、1,2,4-三唑基、1,2,3-噁二唑基、1,2,4-噁二唑基、1,2,5-噁二唑基、1,3,4-噁二唑基、吡啶基、2-吡啶酮、4-吡啶酮、嘧啶基、1,4-二氧杂环己二烯基、2H-1,2-噁嗪基、4H-1,2-噁嗪基、6H-1,2-噁嗪基、4H-1,3-噁嗪基、6H-1,3-噁嗪基、4H-1,4-噁嗪基、哒嗪基、吡嗪基、1,2,3-三嗪基、1,3,5-三嗪基、1,2,4,5-四嗪基、氮杂环庚三烯基、1,3-二氮杂环庚三烯基、氮杂环辛四烯基等。6-15元杂芳基还包括“9-15元稠杂芳基”(例如9-15元苯并稠杂芳基),其具体实例包括但不限于:苯并呋喃基、苯并异呋喃基、苯并噻吩基、吲哚基、异吲哚、苯并噁唑基、苯并咪唑基、吲唑基、苯并三唑基、喹啉基、2-喹啉酮、4-喹啉酮、1-异喹啉酮、异喹啉基、吖啶基、菲啶基、苯并哒嗪基、酞嗪基、喹唑啉基、喹喔啉基、酚嗪基、喋啶基、嘌呤基、萘啶基、吩嗪、吩噻嗪等。
如本文中所使用的,术语“细胞”特别优选地是指,表达Nur77的细胞。本发明化合物能够与Nur77特异性结合,并作为其配体发挥作用。因此,特别有利地,本发明 化合物能够作用于表达Nur77的细胞。在某些优选的实施方案中,所述细胞为炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
如本文中所使用的,术语“孤儿核受体Nur77”或“Nur77”是指,神经生长因子IB(NGFIB),其由NR4A1基因编码(Chang C等(1989),″Isolation and characterization of human TR3 receptor:a member of steroid receptor superfamily″,J.Steroid Biochem.34(1-6):391-5)。Nur77参与细胞周期、炎症和细胞凋亡等过程,并且其亚细胞定位与细胞的存活和死亡相关(Pei L等人(2006),″Regulation of macrophage inflammatory gene expression by the orphan nuclear receptor Nur77″,Mol.Endocrinol.20(4):786-94;Zhang XK(2007),″Targeting Nur77 translocation″,Expert Opin.Ther.Targets 11(1):69-79)。
如本文中所使用的,术语“与Nur77相关的疾病”是指,其发生和/或进展与Nur77信号通路相关的疾病。研究已显示,Nur77参与细胞周期、炎症和细胞凋亡等过程,并且其亚细胞定位与细胞的存活和死亡相关(同上)。此外,还已报道,Nur77可被多种刺激诱导,包括生理刺激,例如脂肪酸,前列腺素,生长因子,炎性细胞因子,肽激素等;以及物理刺激,例如磁场,机械搅拌(剪切力),膜去极化等(Maxwell MA,Muscat GE(2006),″The NR4A subgroup:immediate early response genes with pleiotropic physiological roles″,Nucl Recept Signal 4:e002)。另外,还已显示,Nur77参与一些实体肿瘤的转移(Ramaswamy S,Ross KN,Lander ES,Golub TR(2003),″A molecular signature of metastasis in primary solid tumors″,Nat.Genet.33(1):49-54)。此外,还已显示,Nur77在发炎的人体滑膜组织、癌细胞、牛皮癣患者、动脉粥样硬化患者、多发性硬化患者中异常表达。因此,术语“与Nur77相关的疾病”包括但不限于,炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎和炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
如本文中所使用的,术语“受试者”是指动物,特别是哺乳动物,优选人。
如本文中所使用的,术语“高脂肪膳食”是指,动物体(例如哺乳动物,例如人)日常摄入的膳食中的脂肪含量超出了动物体正常生理活动所需的脂肪量。
如本文中所使用的,术语“有效量”是指,足以获得或至少部分获得期望的效果的量。例如,预防有效量是指,足以预防,阻止,或延迟疾病的发生的量;治疗有效量是指,足以治愈或至少部分阻止已患有疾病的患者的疾病和其并发症的量。测定这样的有效量完全在本领域技术人员的能力范围之内。例如,对于治疗用途有效的量将取决于待治疗的疾病的严重度,患者自己的免疫系统的总体状态,患者的一般情况例如年龄、体重和性别,药物的施用方式,以及同时施用的其他治疗等等。
如本文中所使用的,术语“免疫学方法”是指利用抗原-抗体之间的特异性相互作用/结合亲和力来进行的检测方法,其一般可用于检测特定抗原或者抗体在样品中的存在或水平。此类免疫学测定是本领域技术人员公知的,包括但不限于,ELISA检测,Elispot检测,Western印迹,表面等离子共振法等。关于免疫学测定的详细描述,可参见例如,Fundamental Immunology,Ch.7 Paul,W.,ed.,第2版,Raven Press,N.Y.(1989)。
在本申请中,发明人通过大量的研究发现,如式I-式V所示的化合物(例如化合物YXY101)能够靶向孤儿核受体Nur77,并作为其配体发挥作用。例如,此类化合物可通过结合Nur77调控线粒体活性从而抑制炎症反应,达到预防和治疗与孤儿核受体Nur77相关的疾病,例如炎症(例如,与肥胖症相关的炎症、与动脉粥样硬化相关的炎症、与糖尿病相关的炎症、肝炎、肺炎、关节炎、肠炎)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症。
因此,在第一个方面,本申请涉及如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
Figure PCTCN2017091716-appb-000001
其中,
X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和 C1-6烷氨基;
R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
式(I)中的虚实双键
Figure PCTCN2017091716-appb-000002
代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
在某些优选实施方案中,X代表NH、O或CH2
当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
R1代表H、C1-6烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
R2代表H,-CONH2,-NH2,-NHR,-NRR’,-NHCOR,-NRCOR,-NHCOOR,-NHCONHR,-NHCONRR’,-NRCONHR,-NRCONRR’,-OH,-OR,-OCONHR,-OCONRR’,-SH,-SR,-SOR,-SOOR,-SO2NHR”,卤素,氰基,-CF3,C1-6烷基,3-8元环烷基,3-8元杂环烷基,芳基,C1-6烷基取代的芳基,6-15杂芳基,链烯基,炔基,亚磺基,磺酸或磺酸盐;其中所述C1-6烷基,3-8元环烷基,3-8元杂环烷基,芳基,C1-6烷基取代的芳基,6-15杂芳基,链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:C1-6烷基,3-8元环烷基(例如环丙基),氧代3-8元环烷基(例如氧代环丁基),氨基,卤素,羟基,C1-6烷氧基,C1-6烷硫基,氰基,-CF3,C1-6烷氧羰基,酰胺,脲,氨基甲酸酯基,羧基以及芳基;
R3和R4各自独立地表示不存在,或代表H、C1-6烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
R和R’各自独立地选自H、C1-6烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
式(I)中的虚实双键
Figure PCTCN2017091716-appb-000003
代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;和/或,环B包含0、1或2个碳碳双键。
在某些优选实施方案中,所述磺酸盐选自磺酸钠、磺酸钾、磺酸钙和磺酸镁。
在某些优选实施方案中,所述6-15元杂芳基选自9-15元稠杂芳基;更优选地,所述6-15元杂芳基选自9-15元苯并稠杂芳基,例如吲哚基、苯并呋喃基、苯并噻吩基、苯并咪唑基或喹啉基。
在某些优选的实施方案中,Y和与之相连的碳原子之间为双键,且Y代表O。
在某些优选的实施方案中,X代表NH;Y和与之相连的碳原子之间为双键,且Y代表O;R1代表被一个或多个(例如1、2、3或4个)选自下述的取代基取代的芳基:C1-6烷基和C1-6烷氧基;优选地,所述芳基为苯基或萘基;R2代表H。
在某些优选的实施方案中,X代表O;Y和与之相连的碳原子之间为双键,且Y代表O;R1代表H;R2代表H、磺酸盐或6-15元杂芳基;优选地,所述磺酸盐选自磺酸钠、磺酸钾、磺酸钙和磺酸镁;优选地,所述6-15元杂芳基选自9-15元稠杂芳基;更优选地,所述6-15元杂芳基选自9-15元苯并稠杂芳基,例如吲哚基、苯并呋喃基、苯并噻吩基、苯并咪唑或喹啉基。
在某些优选的实施方案中,X代表O;Y和与之相连的碳原子之间为双键,Y代表O;R1代表C1-6烷基或芳基-C1-6烷基(优选苄基);R2代表H。
在某些优选的实施方案中,R3不存在,且R4代表H。在某些优选的实施方案中,R3代表H,且R4不存在。在某些优选的实施方案中,R3和R4均为H。
在某些优选的实施方案中,R3不存在,R4代表H,且环A和环B中各有两个碳碳双键。
在某些优选的实施方案中,R3代表H,R4不存在,且环A中有0、1或2个碳碳双键;优选地,环B中有0、1或2个碳碳双键。
在某些优选的实施方案中,R3和R4均为H,且环A中有3个碳碳双键(即,环A为苯环);优选地,环B中有0或1个碳碳双键;更优选地,环B中7位和8位碳原子之间为碳碳双键。
在某些优选的实施方案中,R3和R4均不存在。在此类实施方案中,环A中2位碳原子与其所连接的O原子之间为碳氧双键,并且3位碳原子与其所连接的O原子之间为碳 氧双键。
在某些优选的实施方案中,7位和8位碳原子之间为碳碳双键。在某些优选的实施方案中,7位和8位碳原子之间为碳碳单键。
在某些优选实施方案中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;R代表C1-6烷基或3-8元环烷基(优选环己基);其中,当X为卤素时,R1不存在;
在某些优选实施方案中,X代表-NH-、-N(R)-、-O-或氟;R代表环己基。
在某些优选实施方案中,R1不存在,或代表氢、C1-4烷基、-PO(OR)2、单糖基、C1-4烷氧羰基-C1-4烷基、3-6元环烷基-氨酰基、芳基-C1-4烷基或芳基;其中,所述C1-4烷基、单糖基、C1-4烷氧羰基-C1-4烷基、3-6元环烷基-氨酰基、芳基-C1-4烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-4烷基、C1-4烷氧基、C1-4烷氨基和C1-4烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
R代表C1-4烷基。
在某些优选实施方案中,R1不存在,或代表氢、C1-4烷基、-PO(OR)2、葡萄糖基、C1-2烷氧羰基-C1-2烷基、环己基-氨酰基、苯基-C1-2烷基、萘基-C1-2烷基、苯基或萘基;其中,所述甲基、乙基、葡萄糖基、C1-2烷氧羰基-C1-2烷基、环己基-氨酰基、苯基-C1-2烷基、萘基-C1-2烷基、苯基或萘基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:C1-2烷基、C1-2烷氧基和C1-2烷酰基;
R代表C1-4烷基。
在某些优选实施方案中,R1代表氢、C1-4烷基、-PO(OR)2或C1-2烷氧羰基-C1-2烷基;
R代表C1-3烷基。
在某些优选实施方案中,R1不存在,或代表氢、甲基、乙基、-PO(OMe)2、-PO(OEt)2、-PO(OiPr)2、2,3,4,6-四乙酰氧基-α-D-吡喃葡萄糖基、EtOCOCH2-、环己基-氨酰基、苄基、甲氧基苯基或叔丁基苯基。
在某些优选实施方案中,R2代表H、D、-OH、-PO(OR)2、C1-6烷基、9-15元稠杂芳基或磺酸盐;其中所述C1-6烷基或6-15元杂芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、 C1-6烷酰基、氰基、三氟甲基和羧基;
R代表H、C1-6烷基或芳基。
在某些优选实施方案中,R2代表H、D、-PO(OR)2、C1-4烷基、9-15元苯并稠杂芳基或磺酸盐;其中,所述C1-4烷基或9-15元苯并稠杂芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-4烷基、C1-4烷氧基、C1-4烷酰基、氰基、三氟甲基和羧基;
R代表H、C1-4烷基或苯基。
在某些优选实施方案中,R2代表H、D或磺酸盐;
在某些优选实施方案中,R2代表H、D、-PO(OR)2、2-氧代苯基、吲哚基或磺酸钠;其中,所述吲哚基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、氟、氯、溴、羟基、甲基、甲氧基、甲酰基、氰基、三氟甲基和羧基;
R代表H、甲基、乙基、异丙基或苯基。
在某些优选实施方案中,所述化合物的7位和8位碳原子之间为碳碳双键。
在某些优选实施方案中,所述化合物Y和与之相连的碳原子之间为碳碳双键。
在某些优选实施方案中,所述化合物Y和与之相连的碳原子之间为碳碳单键。
在某些优选实施方案中,所述化合物具有如下结构:
Figure PCTCN2017091716-appb-000004
其中,R3和R4各自独立地代表H、C1-6烷基或C1-6烷酰基;
其余原子或取代基的定义如前文中所定义。
在某些优选实施方案中,R3和R4各自独立地代表H、C1-4烷基或C1-4烷酰基。
在某些优选实施方案中,R3和R4各自独立地代表H、甲基或丁酰基。
在某些优选实施方案中,所述化合物具有如下结构:
Figure PCTCN2017091716-appb-000005
其中,R4代表H、C1-6烷酰基、C1-6烷氧羰基或被一个或多个(例如1、2、3或4个)C1-6烷酰基取代的单糖基;
其余原子或取代基的定义如前文中所述。
在某些优选实施方案中,R4代表H、C1-4烷酰基、C1-4烷氧羰基或被一个或多个(例如1、2、3或4个)C1-4烷酰基取代的葡萄糖糖基;
在某些优选实施方案中,R4代表H或C1-2烷氧羰基;
在某些优选实施方案中,R4代表H、丁酰基、乙氧羰基或2,3,4,6-四乙酰氧基-α-D-吡喃葡萄糖糖基。
在某些优选的实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000006
Figure PCTCN2017091716-appb-000007
Figure PCTCN2017091716-appb-000008
Figure PCTCN2017091716-appb-000009
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞, 宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应为κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、 p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体在细胞中的活性。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病。在某些优选的实施方案中,所述与孤儿核受体Nur77相关的疾病选自炎症(例如,与肥胖症相关的炎症、与动脉粥样硬化相关的炎症、与糖尿病相关的炎症、肝炎、肺炎、关节炎、肠炎)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述 药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000010
其中,所述原子或取代基的定义如本文第一方面中所述。
在某些优选的实施方案中,所述方法用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述方法用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000011
其中,所述原子或取代基的定义如前文中所述。
在某些优选的实施方案中,所述方法在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案 中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯与细胞内的Nur77相结合,并由此抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000012
其中,各原子或取代基的定义如本文第一方面中所述。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000013
其中,各原子和取代基的定义如本文第一方面中所述。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在另一个方面,本申请涉及一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
Figure PCTCN2017091716-appb-000014
其中,各原子和取代基的定义如本文第一方面中所述。
在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种预防或治疗与Nur77相关的疾病的方法,其包括,给有此需要的受试者施用有效量的如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000015
其中,各原子和取代基的定义如本文第一方面中所述。
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施 方案中,所述方法包括,给患有炎症的受试者施用有效量的所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,从而在所述受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述方法包括,给患有肥胖症的受试者施用有效量的所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,从而抑制所述受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述方法包括,给患有肥胖症的受试者施用有效量的所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,从而抑制所述受试者的肥胖症并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述方法包括,给患有癌症的受试者施用有效量的所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,从而在所述受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述方法还包括,给患有癌症的受试者施用有效量的TNFα。
在另一个方面,本申请涉及如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
Figure PCTCN2017091716-appb-000016
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000017
代表单键或者双键。
在某些优选的实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000018
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细 胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异 构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体在细胞中的活性。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病。
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在 血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000019
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000020
代表单键或者双键。
在某些优选的实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000021
在某些优选的实施方案中,所述方法用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述方法用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000022
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000023
代表单键或者双键。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000024
在某些优选的实施方案中,所述方法在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯与细胞内的Nur77相结合,并由此抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。
在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中, 所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000025
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000026
代表单键或者双键。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000027
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000028
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000029
代表单键或者双键。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000030
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在另一个方面,本申请涉及一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
Figure PCTCN2017091716-appb-000031
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000032
代表单键或者双键。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000033
在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种预防或治疗与Nur77相关疾病的方法,其包括,向有此需要的受试者施用有效量的如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的步骤:
Figure PCTCN2017091716-appb-000034
其中,X代表NH、O或CH2
Y代表O、S或NR;
R5和R6各自独立地代表代表H或C1-6烷基;
当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
R8代表H或C1-6烷基;
式(IV)中的虚实双键
Figure PCTCN2017091716-appb-000035
代表单键或者双键。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000036
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所 述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
Figure PCTCN2017091716-appb-000037
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞, 宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、 p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体在细胞中的活性。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病。
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施 方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000038
Figure PCTCN2017091716-appb-000039
在某些优选的实施方案中,所述方法用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述方法用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000040
在某些优选的实施方案中,所述方法在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯与细胞内的Nur77相结合,并由此抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。
在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中, 所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000041
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000042
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接 受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在另一个方面,本申请涉及一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的下述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
Figure PCTCN2017091716-appb-000043
在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种预防或治疗与Nur77相关疾病的方法,其包括,向有此需要的受试者施用有效量的如下所示的化合物、其互变异构体、立体异构体或 药学上可接受的盐或酯的步骤:
Figure PCTCN2017091716-appb-000044
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。 在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及如下所示化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000045
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000046
在另一个方面,本申请涉及如式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
Figure PCTCN2017091716-appb-000047
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000048
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎 症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细 胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体在细胞中的活性。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病。
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或 肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000049
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000050
在某些优选的实施方案中,所述方法用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述方法用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000051
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000052
在某些优选的实施方案中,所述方法在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯与细胞内的Nur77相结合,并由此抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。
在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000053
Figure PCTCN2017091716-appb-000054
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000055
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000056
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000057
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在另一个方面,本申请涉及一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
Figure PCTCN2017091716-appb-000058
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000059
在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种预防或治疗与Nur77相关疾病的方法,其包括,向有此需要的受试者施用有效量的式V所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的步骤:
Figure PCTCN2017091716-appb-000060
其中,
R9和R10各自独立地选自氢、C1-6烷基和芳基;
优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
在某些优选实施方案中,所述化合物选自下列化合物:
Figure PCTCN2017091716-appb-000061
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000062
在另一个方面,本申请涉及下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
Figure PCTCN2017091716-appb-000063
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制Nur77的转录活性。在某些 优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴 细胞等)。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体在细胞中的活性。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病。
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例 如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000064
在某些优选的实施方案中,所述方法用于在体内、体外或离体抑制Nur77的转录活性。在某些优选的实施方案中,所述方法用于抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000065
在某些优选的实施方案中,所述方法在体内、体外或离体抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯与细胞内的Nur77相结合,并由此抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应。
在某些优选的实施方案中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活。在某些优选的实施方案中, 所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解。在某些优选的实施方案中,所述细胞表达Nur77。在某些优选的实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
Figure PCTCN2017091716-appb-000066
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
Figure PCTCN2017091716-appb-000067
在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运。在某些优选实施方案中,所述细胞表达Nur77。在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与LC3的相互作用及线粒体定位,促进自噬过程。
在某些优选实施方案中,本发明所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物能够促进Nur77与p62的相互作用及线粒体定位,促进损伤线粒体的清除。
在另一个方面,本申请涉及一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的如下所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
Figure PCTCN2017091716-appb-000068
在某些优选实施方案中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
在另一个方面,本申请涉及一种预防或治疗与Nur77相关疾病的方法,其包括,向有此需要的受试者施用有效量的如下所示的化合物、其互变异构体、立体异构体或 药学上可接受的盐或酯的步骤:
Figure PCTCN2017091716-appb-000069
在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。在某些优选的实施方案中,所述与Nur77相关的疾病选自炎症、肥胖症和癌症。
在某些优选的实施方案中,所述与Nur77相关的疾病为炎症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有炎症的受试者中抑制细胞因子例如IL-1β和/或IL-6的水平(例如,在血清和/或肝脏中的水平)。在某些优选的实施方案中,所述炎症为与TNFα相关的炎症(例如急性炎症或慢性炎症)。在某些优选的实施方案中,所述炎症为肝炎或肺炎,例如急性肝炎或肺炎(例如脂多糖(LPS)和/或D-半乳糖胺(D-GalN)诱导的急性肝炎或肺炎)。在某些优选的实施方案中,所述炎症为慢性炎症,例如患有肥胖症的受试者中的慢性炎症。
在某些优选的实施方案中,所述与Nur77相关的疾病为肥胖症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的体重增长和/或脂肪增加。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制患有肥胖症的受试者的并发症,例如慢性炎症。在某些优选的实施方案中,所述肥胖症是由高脂肪膳食引起的。
在某些优选的实施方案中,所述与Nur77相关的疾病为癌症。在某些优选的实施方案中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于在患有癌症的受试者中抑制癌细胞的增殖和/或转移和/或促进癌细胞的凋亡。在某些优选的实施方案中,所述癌症选自肝癌,宫颈癌,肺癌、和乳腺癌。在某些优 选的实施方案中,所述癌症为三阴性乳腺癌(即,雌激素受体(ER)、孕激素受体(PR)和原癌基因Her-2均为阴性的乳腺癌)。在某些优选的实施方案中,所述药物用于治疗癌症,并且包含所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,以及TNFα。
在另一个方面,本申请涉及一种具有抗炎和/或治疗肥胖症活性的药物的筛选方法,其包括以下步骤:
(1)提供表达孤儿核受体Nur77以及至少下述一种蛋白的细胞:TRAF2、p62、LC3和Ub;
(2)用TNFα和候选试剂处理所述细胞,设立阴性对照组(即不经候选试剂处理);
(3)检测并判断如下项:
a.所述候选试剂是否可以与Nur77结合;
b.是否可以诱导Nur77从细胞核转运至线粒体;
c.与阴性对照组比,IκBα的表达水平是否升高;
d.TRAF2的泛素化是否被抑制;
e.Nur77的泛素化是否被诱导;
f.是否发生TRAF2与Nur77在线粒体的相互作用;
g.是否发生p62的线粒体定位;
h.是否发生p62与Nur77在线粒体的相互作用;
i.是否发生LC3的线粒体定位;
j.是否发生LC3与Nur77在线粒体的相互作用;
(4)如所述步骤(3)中a项和b项判断结果均为“是”,且,c至i任一项判断结果为“是”,则判断其具有抗炎和/或治疗肥胖症活性。
在某些优选实施方案中,a-j项可通过免疫学方法进行检测。
在某些优选实施方案中,所述免疫学方法选自ELISA检测、Elispot检测、Western印迹、表面等离子共振法、免疫荧光染色、免疫组化染色和免疫共沉淀等。
发明的有益效果
(1)本发明人首次发现,如式I-式V所示的化合物能够靶向孤儿核受体Nur77,并作为其配体发挥作用。
(2)本发明人发现如式I-式V所示的化合物可诱导Nur77依赖的抑制炎症和自噬作用。
在本申请的某些实施方案中,所述化合物通过促进Nur77从核内转运到线粒体上,与肿瘤坏死因子受体相关因子2(TRAF2,一种炎症信号通路中重要的支架蛋白和关键的泛素连接酶)相互作用。所述相互作用是通过TRAF2上的LXXLL基序介导的。该相互作用不仅抑制了TRAF2的泛素化还诱导了Nur77在K63位的泛素化。在炎症状态下,泛素化的Nur77在线粒体上与p62/SQSTM1泛素集合域相互作用来增强线粒体自噬的敏感性。LC3(一种自噬相关蛋白)从而与修饰后的Nur77特异的相互作用,保证受损的线粒体能被选择性清除。
本发明人通过检测Nur77与TRAF2、LC3、p62和/或Ub的相互作用及线粒体转运,筛选得到一系列具有抗炎和/或治疗肥胖症活性的化合物。由此,本发明人建立了一套筛选抗炎和/或治疗肥胖症的药物的方法。
(3)本申请的化合物还可用于预防和治疗与孤儿核受体Nur77相关的疾病,例如炎症(例如与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。因此,本发明提供了新的、有效的、特异性结合Nur77的配体,其可用于开发治疗炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)的新疗法。
附图说明
图1显示了本申请的发明人发现的Nur77参与抑制炎症和诱导自噬作用的机理说明图。
图2显示了用Biacore T200仪器筛选能与Nur77结合的化合物。如图显示了用Biacore T200仪器检测YXY101与Nur77-LBD的结合的实验结果,其中,红色点代表 化合物YXY101;蓝色点代表对照化合物。结果显示,化合物YXY101能够与Nur77-LBD结合。
图3A显示了雷公藤红素(化合物YXY101)的化学结构式。
图3B显示了进一步用不同浓度的YXY101(0.04μM,0.08μM,0.16μM,0.32μM,0.64μM)与Nur77-LBD的结合的实验结果。结果显示,化合物YXY101与Nur77-LBD结合的解离常数(Kd)为292nM;
图3C显示了用圆二色光谱法检测YXY101与Nur77-LBD的结合的实验结果,其中,红色曲线代表YXY101+Nur77-LBD;蓝色曲线代表Nur77-LBD。结果显示,化合物YXY101能改变Nur77-LBD的CD光谱。这表明,化合物YXY101能够与Nur77-LBD结合。
图3D显示了用HPLC法检测YXY101与Nur77-LBD的结合的实验结果,其中,红色曲线代表YXY101+Nur77-LBD;紫色曲线代表YXY101+RXRα-LBD。结果显示,化合物YXY101能够与Nur77-LBD结合形成复合体,但不与RXRα-LBD结合。
图3E显示了用双荧光素酶报告基因系统检测YXY101与Nur77-LBD的结合的实验结果。结果显示,化合物YXY101能抑制Nur77的转录激活功能,但对糖皮质激素受体(GR)的转录激活功能无明显作用。这表明,化合物YXY101能够与Nur77-LBD结合,并抑制其转录活性;并且,化合物YXY101不与GR结合。
图3F显示了YXY101与Nur77的分子对接。分子对接结果显示,YXY101主要通过疏水作用结合到Nur77蛋白表面的已知的疏水凹槽。
图4A-4B显示了经不同浓度的YXY101和TNFα处理的细胞中IκBα和磷酸化的IKKα/β的免疫印迹分析结果。
图4C显示了经YXY101和TNFα处理的细胞的免疫荧光染色结果(Scale bar:20μm)。
图4D显示了经YXY101和TNFα处理的细胞的NF-κB活性的分析结果,其中,**P<0.01,***P<0.001(T检验)。
图4E显示了经不同浓度的YXY101和TNFα处理的不同癌细胞系中IκBα和磷酸 化的IKKα/β的免疫印迹分析结果。
图4F显示了经不同化合物处理、且接受TNFα刺激的HepG2细胞中IκBα的免疫印迹分析结果。
图5A-5B显示了转染了不同siRNA、且经不同浓度的YXY101和TNFα处理的HepG2细胞中Nur77、RXRα和IκBα的免疫印迹分析结果。
图5C显示了经不同浓度的YXY101和TNFα处理的MEF细胞和Nur77-/-MEF细胞中IκBα的免疫印迹分析结果。
图5D显示了经YXY101和TNFα处理的MEF细胞和Nur77-/-MEF细胞的免疫荧光染色结果(Scale bar:10μm)。
图6A显示了各处理组小鼠的血清ALT和血清AST的水平。
图6B显示了各处理组小鼠的血清IL-1β和血清IL-6的水平。
图6C显示了各处理组小鼠的肝脏IL-1β和肝脏IL-6的mRNA水平。在图7A-7C中,数据表示三次独立实验的平均值±SEM;ns表示无显著差异;*P<0.05,**P<0.01,***P<0.001(学生氏t检验)。
图6D显示了各处理组小鼠的肝脏IκBα的蛋白水平。
图7A显示了各处理组小鼠的肝脏的H&E染色结果(scale bar,100μm)。
图7B显示了各处理组小鼠的肝脏的p65免疫组化染色结果(scale bar,20μm)。
图8A显示了各处理组小鼠的体型,体重,脂肪组织块的状况。
图8B显示了各处理组小鼠的肝脏IL-1β和肝脏IL-6的mRNA水平。
图8C显示了各处理组小鼠的肝脏IκBα的蛋白水平。
图8D显示了各处理组小鼠的肝脏的H&E染色结果、p65免疫组化染色结果、以及肝嗜中性粒细胞的染色结果。
图9A显示了,用TNFα(20ng/ml,30min)和YXY101(2μM,1h)处理野生型和Nur77缺失的MEF细胞,运用免疫荧光技术手段观察Nur77、TRAF2在细胞线粒体中的定位情况。
图9B显示了上述处理的细胞中Nur77、TRAF2、线粒体共定位的统计结果。
图9C显示了用TNFα(20ng/ml)和YXY101(2μM,1h/9h)处理HepG2细胞,分离出线粒体,并用免疫印迹的方法检测全细胞蛋白和线粒体蛋白中的Nur77、TRAF2、p62、LC3、PARP、Hsp60水平。
图10A显示了用TNFα(20ng/ml)和YXY101(2μM)处理HepG2细胞,IP:TRAF2,并用免疫印迹的方法检测与TRAF2相互作用的Nur77。
图10B显示了用YXY101与GST-TRAF2、His-Nur77-LBD蛋白共孵育,并用免疫印迹的方法检测与GST-TRAF2蛋白结合的His-Nur77-LBD蛋白。
图10C显示了在HepG2细胞中外转Flag-TRAF2质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测内源的Nur77和外源的Flag-TRAF2共定位情况。
图10D显示了在HepG2细胞中外转Flag-TRAF2、GFP-Nur77质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测Flag-TRAF2、GFP-Nur77、线粒体的共定位情况。
图11A显示了在HepG2细胞中外转Flag-TRAF2、GFP-LC3质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测Flag-TRAF2、GFP-LC3和线粒体的共定位情况。
图11B显示了在HepG2细胞中外转RFP-LC3、GFP-Nur77质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测RFP-LC3、GFP-Nur77和线粒体的共定位情况。
图11C显示了用TNFα(20ng/ml)和YXY101(2μM)处理野生型和Nur77缺失的MEF细胞,运用免疫染色技术检测Nur77、LC3和线粒体的共定位情况。
图11D显示了上述处理的细胞中Nur77、LC3、线粒体共定位的统计结果。
图12A显示了在HepG2细胞中外转Myc-Nur77、Flag-p62质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,IP:Flag,并用免疫印迹的方法检测与Flag-p62相互作用的Myc-Nur77。
图12B显示了用TNFα(20ng/ml)和YXY101(2μM)处理HepG2细胞,运用免疫染色技术检测Nur77和p62的共定位情况。
图12C显示了在HepG2细胞中外转GFP-LC3质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测GFP-Nur77和p62的共定位情况。
图13A显示了在HepG2细胞中外转Flag-TRAF2、Myc-Nur77、HA-Ub质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,IP:Myc,用免疫印迹手段检测Nur77的泛素化情况。
图13B显示了在HepG2细胞中外转Myc-Nur77、HA-Ub质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,分离出线粒体,IP:Myc,用免疫印迹手段检测线粒体中的Nur77泛素化情况。
图13C显示了在HepG2细胞中外转GFP-Nur77、HA-Ub质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测GFP-Nur77和HA-Ub的共定位情况。
图13D显示了在HepG2细胞中外转GFP-Nur77质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测GFP-Nur77和Ub的共定位情况。
图14A显示了经过不同浓度的XS0284和TNFα处理的MEF细胞和Nur77-/-MEF中IκBα的免疫印迹分析结果。
图14B显示了经过不同浓度的XS0284和TNFα处理的MEF细胞和Nur77-/-MEF中IκBα的免疫印迹分析结果。
图14C显示了经过不同浓度的XS0474、XS0503、XS0419、XS0486和TNFα处理的MEF细胞和Nur77-/-MEF中IκBα的免疫印迹分析结果。
图15A显示了用Biacore T200仪器以YXY101作为对照,初步筛选能与Nur77结合的YXY101衍生物。
图15B显示了进一步用不同浓度的YXY101衍生物(0.3125μM,0.625μM,1.25μM,2.5μM,5μM,10μM)与Nur77-LBD的结合的实验结果。结果显示,化合物XS0284与Nur77-LBD结合的解离常数(Kd)为386nM;XS0394与Nur77-LBD结合的 解离常数(Kd)为1.26μM;化合物XS0418与Nur77-LBD结合的解离常数(Kd)为3.67μM;化合物XS0462与Nur77-LBD结合的解离常数(Kd)为2.07μM;化合物XS0474与Nur77-LBD结合的解离常数(Kd)为2.60μM;化合物XS0486与Nur77-LBD结合的解离常数(Kd)为1.20μM;化合物XS0419与Nur77-LBD结合的解离常数(Kd)为404nM;化合物XS0503与Nur77-LBD结合的解离常数(Kd)为1.63μM。
图16A显示了用YXY101和不同的YXY101衍生物XS0284、XS0285、XS0286、XS0287、XS0335、XS0366、XS0260、XS0394、XS0395、XS0419、XS0420、XS0421以及TNFα处理的MDA-MB-231细胞中IκBα和PARP的免疫印迹分析结果。
图16B显示了用YXY101和不同的YXY101衍生物XS0284-4、XS0284、XS0077、XS0503、XS0486、XS0419、XS0474、XS0462、XS0285、XS0394、XS0454、XS0455、XS0462、XS0473、XS0474、XS0480以及TNFα处理的MDA-MB-231细胞中IκBα和PARP的免疫印迹分析结果。
图16C显示了用YXY101和不同的YXY101衍生物XS0284、XS0285、XS0335、XS0394、XS0418、XS0419、XS0454、XS0455、XS0462、XS0502、XS0503、XS0504、XS0506、XS0507、XS0508、XS0077以及TNFα处理的MDA-MB-231细胞中IκBα和PARP的免疫印迹分析结果。
图17A显示了在HepG2细胞中外转Flag-p62、Myc-Nur77质粒,用TNFα(20ng/ml)、YXY101(4μM)、XS0284(2μM、4μM)处理,IP:Flag,运用免疫印迹检测与Flag-p62相互作用的Myc-Nur77。
图17B显示了在HepG2细胞中外转Flag-TRAF2、HA-Ub质粒,用TNFα(20ng/ml)、YXY101(4μM)、XS0284(4μM)处理,IP:Flag,运用免疫印迹检测Flag-TRAF2的泛素化情况。
图17C显示了在HepG2细胞中外转GFP-Nur77、Flag-TRAF2质粒,用TNFα(20ng/ml)和YXY101(2μM)、XS0284(4μM)处理,运用免疫染色技术检测GFP-Nur77和Flag-TRAF2和线粒体的共定位情况。
图17D显示了在HepG2细胞中外转GFP-Nur77、RFP-LC3质粒,用TNFα(20ng/ml)和YXY101(2μM)、XS0284(4μM)处理,运用免疫染色技术检测GFP-Nur77 和RFP-LC3的共定位情况。
图17E显示了在HepG2细胞中外转GFP-Nur77、Flag-p62质粒,用TNFα(20ng/ml)和YXY101(2μM)、XS0284(4μM)处理,运用免疫染色技术检测GFP-Nur77和Flag-p62、Ub的共定位情况。
图18A显示了经XS0077和TNFα处理的Hela细胞的免疫荧光染色结果。HepG2细胞用XS0077(2μM)处理3小时,然后用TNFα(20ng/mL)处理30分钟。随后,用免疫荧光染色法检测细胞中的Nur77与Traf2的共定位情况。
图18B显示了经XS0503和TNFα处理的Hela细胞的免疫荧光染色结果。Hela细胞用XS0503(2μM)处理3小时,然后用TNFα(20ng/mL)处理30分钟。随后,用免疫荧光染色法检测细胞中的Nur77与Traf2的共定位情况。
图19A显示了用YXY101和XS0284以及TNFα处理的MDA-MB-231细胞中PARP的免疫印迹分析结果。
图19B显示了用YXY101和XS0284以及TNFα处理的NCL-H292细胞中PARP的免疫印迹分析结果。
图19C显示了化合物YXY101和XS0284对HepG2细胞的增殖抑制率。
图19D显示了用流式细胞术检测化合物YXY101和XS0284以及TNFα处理的MDA-MB-231的线粒体膜电位的变化。
图20A显示了,在对6周的C57BL/6小鼠灌胃分别给药200mg/kg的YXY101及其衍生物XS0284一次后,分别观察各组小鼠心脏、肝脏、小肠、白色脂肪的组织形态以及HE染色的结果。
图20B显示了,在对6周的C57BL/6小鼠腹腔注射分别给药200mg/kg的YXY101及其衍生物XS0284一次后,分别观察各组小鼠心脏、肝脏、白色脂肪、肾脏的组织形态以及HE染色的结果。
图21使用高脂诱导的肥胖小鼠,腹腔注射YXY101(0.1mg/kg)、XS0284(0.1mg/kg)一周时间。图21A显示了小鼠的外部形态特征和肝脏的形态特征;图21B显示了上述小鼠的体重变化的统计学分析;图21C显示了上述小鼠在给药7天之内的体重变化曲 线;图21D显示了小鼠体重减少百分比统计图。
图22显示了利用计算机辅助模拟Nur77与其配体结合的作用模式图。
具体实施方式
现参照下列意在举例说明本发明(而非限定本发明)的实施例来描述本发明。
除非特别指明,本发明中所使用的分子生物学实验方法和免疫检测法,基本上参照J.Sambrook等人,分子克隆:实验室手册,第2版,冷泉港实验室出版社,1989,以及F.M.Ausubel等人,精编分子生物学实验指南,第3版,John Wiley & Sons,Inc.,1995中所述的方法进行;限制性内切酶的使用依照产品制造商推荐的条件。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。
实施例1.Nur77参与抑制炎症和诱导自噬作用的机理说明
图1显示了本申请的发明人首次发现的Nur77参与抑制炎症和诱导自噬作用的机理说明图。如图所示,本申请的发明人发现某些化合物能通过结合Nur77,并诱导Nur77依赖的抑制炎症和自噬作用。所述化合物通过促进Nur77从核内转运到线粒体上,与肿瘤坏死因子受体相关因子2(TRAF2,一种炎症信号通路中重要的支架蛋白和关键的泛素连接酶)相互作用。所述相互作用是通过TRAF2上的LXXLL基序介导的。该相互作用不仅抑制了TRAF2的泛素化还诱导了Nur77在K63位的泛素化。在炎症状态下,泛素化的Nur77在线粒体上与p62/SQSTM1泛素集合域相互作用来增强线粒体自噬的敏感性。LC3(一种自噬相关蛋白)从而与修饰后的Nur77特异的相互作用,保证受损的线粒体能被选择性清除。
综上,本申请的发明人通过上述所阐明的机理,建立一套筛选Nur77依赖的抑制炎症和诱导自噬作用的化合物的方法,并对筛选得到的化合物的生物学活性进行了分析,将在以下做进一步说明。
实施例2.化合物YXY101的表征
(1)表面等离子共振术(SPR)
通过表面等离子共振术来测定YXY101与Nur77的结合。简言之,将50μg经纯化的Nur77配体结合域(Nur77-LBD)蛋白与Biacore的CM5芯片偶联;随后通过Biacore T200仪器来测定YXY101(20μm)与Nur77-LBD的结合。将无关化合物用作对照。测定结果示于图2中。
图2显示了用Biacore T200仪器检测YXY101与Nur77-LBD的结合的实验结果,其中,红色点代表化合物YXY101;蓝色点代表对照化合物。结果显示,化合物YXY101能够与Nur77-LBD结合。
(2)YXY101与Nur77的结合动力学
通过表面等离子共振术来测定YXY101与Nur77的解离常数(Kd)。简言之,使用Biacore T200仪器来测定不同浓度的YXY101(0.04μM,0.08μM,0.16μM,0.32μM,0.64μM)与Nur77-LBD的结合。测定结果示于图3B中。
结果显示,化合物YXY101与Nur77-LBD结合的解离常数(Kd)为292nM。
(3)圆二色光谱法(CD)
使用圆二色光谱法(CD)对YXY101与Nur77-LBD的结合作进一步分析。简言之,将YXY101(1ml,1mg/ml)加入Nur77-LBD蛋白(1ml,1mg/ml)的磷酸盐缓冲液(10μm,PH7.4)中,并在4℃下孵育3h。随后,取0.7ml的溶液,并用Jasco J-810分光偏振计进行检测。记录从190nm到260nm的CD光谱。将单独的Nur77-LBD溶液(即,未添加化合物YXY101)用作对照。检测结果示于图3C中。
结果显示,化合物YXY101能改变Nur77-LBD的CD光谱。这表明,化合物YXY101能够与Nur77-LBD结合。
(4)高效液相色谱(HPLC)分析
使用高效液相色谱(HPLC)对YXY101与Nur77-LBD的结合作进一步分析。简言之,将YXY101(600uL,0.1mg/ml)与经纯化的Nur77-LBD蛋白(5ml,1mg/ml)共同孵育。在4℃条件下孵育3h后,用Ni beads捕获YXY101和Nur77-LBD的复合体。随后,使用氯仿将该复合体解离,并将解离产物中的YXY101萃取出来。然后, 使用HPLC分光仪(Shimadzu LC 20A,Japan)来检测萃取产物中的YXY101,其中,所使用的柱子为ODS柱(5um,4.6*250mm),流动相为含有0.2%H3PO4的乙腈水溶液,检测波长为425nm。另外,还使用YXY101与RXRα-LBD(维甲类X受体α的配体结合域)重复上述实验,用作对照。检测结果示于图3D中。
结果显示,化合物YXY101能够与Nur77-LBD结合形成复合体,但不与RXRα-LBD结合。
(5)双荧光素酶报告基因检测法
使用双荧光素酶报告基因检测法对YXY101与Nur77-LBD的结合作进一步分析。另外,还使用YXY101与糖皮质激素受体(GR)来重复该实验,用作对照。实验结果示于图3E中。
结果显示,化合物YXY101能抑制Nur77的转录激活功能,但对糖皮质激素受体(GR)的转录激活功能无明显作用。这表明,化合物YXY101能够与Nur77-LBD结合,并抑制其转录活性;并且,化合物YXY101不与GR结合。
(6)分子模拟
使用AutoDock V4.2.来完成YXY101与Nur77(PDB code:4JGV)的对接。YXY101的构象由拉马克遗传算法构建。在Nur77的晶体结构中,格中心被选择在已报道的THPN坐标(-12.08,18.29,-4.233),网格尺寸设置为40*40*40(X,Y,Z)格点,每个格点的间隔为0.375A。
在分子对接中,应用标准对接方案:随机放置个体数量为150;能量评估的最大数量为250万;基因突变的比率为0.02;交叉比率为0.8;个体进行局部搜索的概率为0.06;波的下界是0.01;分子可视化使用版本0.99的PyMOL。结果示于图3F中。
分子对接结果显示,YXY101主要通过疏水作用结合到Nur77蛋白表面的已知的疏水凹槽。
实施例3.YXY101抑制TNFα的生物学效应
HepG2细胞用不同浓度的YXY101(0μM,0.25μM,0.5μM,1μM,2μM或4 μM)处理1小时,再用TNFα(0ng/mL或20ng/mL)处理30分钟。随后,用免疫印迹法(Western Bloting)检测细胞中的IκBα和磷酸化的IKKα/β。结果示于图4A-4B中。
图4A-4B的结果显示,TNFα能够在细胞中诱导IKKα/β的磷酸化以及IκBα的降解;而YXY101则能够抑制TNFα诱导的IKKα/β的磷酸化和IκBα的降解。
HepG2细胞用YXY101(0或1μM)处理1小时,然后用TNFα(20ng/mL)处理30分钟。随后,用免疫荧光染色法检测细胞中的NF-κB亚基p65。未经处理的细胞用作对照。结果示于图4C中。
图4C显示了经YXY101和TNFα处理的HepG2细胞的免疫荧光染色结果(Scalebar:20μm)。图3C的结果显示,TNFα能够在细胞中诱导NF-κB亚基p65的入核转运;而YXY101则能够抑制TNFα诱导的p65入核转运。
将NF-κB的报告基因转染入HEK-293T细胞,然后用YXY101(0或1μM)和TNFα(20ng/mL)进行处理。随后,检测细胞中的NF-κB活性。未经处理的细胞用作对照。结果示于图4D中。
图4D显示了经YXY101和TNFα处理的细胞的NF-κB活性的分析结果,其中,**P<0.01,***P<0.001(T检验)。图4D的结果显示,TNFα能够在细胞中诱导NF-κB的转录激活;而YXY101则能够抑制TNFα诱导的NF-κB转录激活。
多种癌细胞系(LO2,SMMC-7721,QGY-7703,HeLa,H460)用不同浓度的YXY101(0μM,1μM或4μM)处理1小时,再用TNFα(0ng/mL或20ng/mL)处理30分钟。随后,用免疫印迹法(Western Bloting)检测细胞中的IκBα和磷酸化的IKKα/β。结果示于图4E中。
结果显示,TNFα能够在各种细胞系中诱导IKKα/β的磷酸化以及IκBα的降解;而YXY101则能够抑制TNFα诱导的IKKα/β的磷酸化和IκBα的降解。
另外,还使用HepG2细胞来进行实验。简言之,HepG2细胞用指定浓度的各种化合物(YXY101、XS0284和XS0287)处理指定的时间,然后再用TNFα(0ng/mL或20ng/mL)处理30分钟。随后,用免疫印迹法(Western Bloting)检测细胞中的IκBα。结果示于图4F中。
图4F显示在HepG2细胞中,YXY101及其衍生物XS0284和XS0287能够抑制TNFα诱导的IκBα的降解。
图4A-4F的结果表明,YXY101及其衍生物XS0284和XS0287能够抑制TNFα在细胞中的各种生物学效应,包括IKKα/β的磷酸化的磷酸化、IκBα的降解、NF-κB亚基p65的入核转运、和NF-κB的转录激活。
实施例4.YXY101对TNFα生物学效应的抑制作用由Nur77介导
将对照SiRNA、Nur77 SiRNA或RXRα SiRNA转染入HepG2细胞。随后,HepG2细胞用不同浓度的YXY101(0μM,1μM或4μM)处理1小时,再用TNFα(0ng/mL或20ng/mL)处理30分钟。随后,用免疫印迹法(Western Bloting)检测细胞中的Nur77、RXRα和IκBα。结果示于图5A-5B中。
图5A-5B显示了转染了不同siRNA、且经不同浓度的YXY101和TNFα处理的HepG2细胞中Nur77、RXRα和IκBα的免疫印迹分析结果。结果显示,Nur77 SiRNA有效抑制/敲除了Nur77在细胞中的表达;并且,RXRα SiRNA有效抑制/敲除了RXRα在细胞中的表达;对照SiRNA则不影响Nur77和RXRα的正常表达。进一步,图5A-5B的结果显示,在表达Nur77的细胞中,YXY101均能够抑制TNFα诱导的IκBα的降解;然而,当Nur77的表达被敲除时,YXY101丧失了抑制IκBα降解的能力。这些结果表明,YXY101对TNFα生物学效应的抑制作用是由Nur77介导的。
还使用MEF细胞和Nur77-/-MEF细胞(即,不表达Nur77的MEF细胞)验证了上述实验结果。简言之,MEF细胞和Nur77-/-MEF细胞用不同浓度的YXY101(0μM或1μM)处理1小时,再用TNFα(0ng/mL或20ng/mL)处理30分钟。随后,用免疫印迹法(Western Bloting)检测细胞中的IκBα。结果示于图5C中。
结果显示,在表达Nur77的MEF细胞中,YXY101均能够抑制TNFα诱导的IκBα的降解;然而,在Nur77-/-MEF细胞中,YXY101丧失了抑制IκBα降解的能力。这些结果表明,YXY101对TNFα生物学效应的抑制作用是由Nur77介导的。
另外,将MEF细胞和Nur77-/-MEF细胞用YXY101(0或1μM)处理1小时,然后用TNFα(20ng/mL)处理30分钟。随后,用免疫荧光染色法检测细胞中的NF-κB 亚基p65。未经处理的细胞用作对照。结果示于图5D中。
图5D显示了经YXY101和TNFα处理的MEF细胞和Nur77-/-MEF细胞的免疫荧光染色结果(Scale bar:10μm)。结果显示,在表达Nur77的MEF细胞中,YXY101能够抑制TNFα诱导的p65入核转运;然而,在Nur77-/-MEF细胞中,YXY101丧失了抑制p65入核的能力。这些结果表明,YXY101对TNFα生物学效应的抑制作用是由Nur77介导的。
实施例5.YXY101能够抑制动物的急性炎症
本实施例采用,腹腔注射80ug/kg LPS和200ug/kg D-GalN 6个小时诱发的小鼠急性肝炎模型。在本实施例中,观察腹腔注射YXY101(0.5mg/kg)12个小时后,小鼠肝脏的ALT和AST水平,血清中IL-1β和IL-6的水平,及肝脏IL-1β和IL-6的mRNA水平。
1、实验材料和方法
1)实验动物和试剂
野生型小鼠和Nur77敲除的小鼠各18只,雄性、均重18-22克,厦门大学实验动物中心SPF级
2)动物处理方法
野生型和Nur77敲除的小鼠在温度23±1℃,湿度:40-60%,自然光照,自由饮水,自由进食普通饲料的条件下进行饲养;将野生型小鼠和Nur77敲除的小鼠各随机分为3组(每组6只),分别为正常对照组、LPS/D-GalN模型组、YXY101(0.5mg/kg)组。
药物配置方法:YXY101-DMSO饱和溶液+5%(v/v)吐温-80+生理盐水,配置成0.05mg/ml
给药方法:
正常对照组和LPS/D-GalN诱导的急性肝炎模型组:于下午20点给予饲料前腹腔注射生理盐水。
YXY101组:于下午20点给予饲料前腹腔注射给药一次,剂量0.5mg/kg。
次日(12小时后),除正常组外,急性肝炎模型组及YXY101组给予80ug/kg LPS和200mg/kg D-GalN,以诱导急性肝炎。
6个小时后,采集样品,即,对于每一只动物,摘除眼球取血,摘取肝脏组织;测定血清中的谷丙转氨酶ALT和谷草转氨酶AST水平,以及肝脏中的谷丙转氨酶ALT和谷草转氨酶AST水平;另外,将部分肝脏组织用多聚甲醛固定,常规方法包埋,并进行免疫组化染色分析。
统计学分析:
数据采用Graphpad Prism 5软件(GraphPad Software Inc,USA)进行统计学分析。使用单因素方差分析来进行组间比较;当方差分析显示差异有显著性时,进一步用q检验进行两两比较。
实验结果实验图6-7中。图6A显示了各处理组小鼠的血清ALT和血清AST的水平。图6B显示了各处理组小鼠的血清IL-1β和血清IL-6的水平。图6C显示了各处理组小鼠的肝脏IL-1β和肝脏IL-6的mRNA水平。在图6A-6C中,数据表示三次独立实验的平均值±SEM;ns表示无显著差异;*P<0.05,**P<0.01,***P<0.001(学生氏t检验)。图6D显示了各处理组小鼠的肝脏IκBα的蛋白水平。图7A显示了各处理组小鼠的肝脏的H&E染色结果(scale bar,100μm)。图7B显示了各处理组小鼠的肝脏的p65免疫组化染色结果(scale bar,20μm)。
为确定Nur77在体内的作用,我们检查了YXY101对野生型小鼠和Nur77缺失型小鼠中的由脂多糖(LPS)/D-半乳糖胺(D-GalN)诱导的肝炎损伤的作用。当对小鼠施用0.2mg/kg的YXY101时,LPS/D-GalN诱导的血清丙氨酸转氨酶(ALT)和天冬氨酸转氨酶(AST)的水平分别降低了35%和47%(图6A)。虽然注射LPS/D-GalN后,Nur77敲除(Nur77-/-)的小鼠的ALT和AST的血清水平比野生型小鼠的高,但YXY101在Nur77-/-小鼠中的抑制作用大大减弱(图6A)。
进一步的分析表明,野生型小鼠中LPS/D-GalN诱导的血清IL-1和IL-6的水平(图6B)及其肝mRNA的表达(图6C)都被YXY101显著抑制,但这种抑制作用在Nur77-/-小鼠中大大减弱。YXY101也显著减弱了野生型小鼠内LPS/D-GalN诱导的IκBα的下 调(图6D)、肝结构的破坏(图7A)、p65的入核(图7B),但在Nur77-/-小鼠内无明显作用。
此外,肺组织的组织学分析也显示,YXY101对于LPS/D-GalN诱导的肺炎和中性粒细胞浸润的抵抗/抑制作用具有Nur77依赖性(数据未提供)。因此,YXY101对LPS/D-GalN诱导的急性炎症的抑制作用是依赖于Nur77的。
实施例6.YXY101能够抑制动物的慢性炎症
本实施例采用,喂饲60%高脂饲料诱发的小鼠肥胖模型。在本实施例中,观察YXY101对肥胖模型小鼠的血清ALT和AST水平,肝脏ALT和AST水平,血清中IL-1β和IL-6的水平及肝脏IL-1β和IL-6的mRNA水平的影响。
1、实验材料和方法
1)实验动物和试剂
野生型小鼠和Nur77敲除的小鼠,雄性、4周龄左右,厦门大学实验动物中心SPF级
2)动物处理方法
野生型和Nur77敲除的小鼠在温度23±1℃,湿度:40-60%,自然光照,自由饮水,自由进食的条件下进行饲养;除正常对照组外,其余各组按普通饲料∶60%高脂饲料(5∶1、4∶1、3∶1、2∶1、1∶1、1∶2、1∶3、1∶4、1∶5)比例适应性喂养9天,以减少直接高脂喂养带来的消化道损伤。过渡期结束后,除正常对照组外,其余各组均使用完全的60%高脂饲料进行饲养。将野生型小鼠和Nur77敲除的小鼠各随机分为3组(每组8只),分别为正常对照组、肥胖模型组、YXY101(0.1mg/kg)组。除正常对照组给予普通饲料外,其余各组均给予高脂饲料。喂养17周。
给药方法:
正常对照组和肥胖模型组:每日于晚上7点给予饲料前,给予生理盐水,方式:腹腔注射。
YXY101组:每日于晚上7点给予饲料前,给药一次,剂量0.1mg/kg,方式:腹 腔注射。
按照上述方法连续给药2周后,对于每一只动物,摘眼球取血,摘取肝脏组织;测定血清中的谷丙转氨酶ALT和谷草转氨酶AST水平,以及肝脏中的谷丙转氨酶ALT和谷草转氨酶AST水平;另外,将部分肝脏组织用多聚甲醛固定,常规方法包埋,并进行免疫组化染色分析。
统计学分析:
数据采用Graphpad Prism 5软件(GraphPad Software Inc,USA)进行统计学分析。使用单因素方差分析来进行组间比较;当方差分析显示差异有显著性时,进一步用q检验进行两两比较。
实验结果实验图8中。图8A显示了各处理组小鼠的体型,体重,脂肪组织块的状况。图8B显示了各处理组小鼠的肝脏IL-1β和肝脏IL-6的mRNA水平。图8C显示了各处理组小鼠的肝脏IκBα的蛋白水平。图8D显示了各处理组小鼠的肝脏的H&E染色结果、p65免疫组化染色结果、以及肝嗜中性粒细胞的染色结果。
我们通过能代表慢性低水平炎症状态的、由高脂膳食(HFD)诱导的肥胖动物模型来确定YXY101和Nur77的作用。结果显示,YXY101在野生型小鼠内能显著减少体重、减少脂肪组织块和脂肪细胞大小,改善HFD诱导的脂肪肝(图8A)。相比于野生型小鼠,Nur77-/-小鼠对HFD诱导的肥胖表现更敏感,具有更高的体重及更多的脂肪量,这与Nur77在代谢中的作用一致。然而,更重要的是,YXY101能在野生型小鼠中减少体重22%,而在Nur77-/-小鼠中只能减少4%(图8A)。这表明,Nur77-/-小鼠对YXY101的抗肥胖作用耐受。类似地,还已显示,与野生型小鼠相比,YXY101在Nur77-/-小鼠中抑制HFD诱导的肥胖以及血清中ALT和AST的升高的能力显著减弱。
此外,用HFD饲养野生型和Nur77-/-型小鼠后,两者的IL-1和IL-6在血清中的水平以及肝内mRNA的表达水平都增强(图8B)。YXY101给药大大抑制了野生型小鼠体内IL-1和IL-6的表达和产生,而Nur77-/-型小鼠则不然。YXY101给药能在野生型小鼠中抑制IκBα蛋白水平的下调(图8C),抑制p65入核(图8D)、抑制肝炎(图8D)、抑制肝嗜中性粒细胞(图8D)和巨噬细胞的累积。而Nur77-/-型小鼠则 不然。因此,YXY101抑制肥胖动物的慢性炎症也是依赖于Nur77的。
综上,我们开创性地以Nur77为靶点,运用SPR手段筛选得到能够特异性结合Nur77的药物分子,具有明显抗炎、减肥、预防脂肪肝的活性,具有开发为减肥药物等的前景。
接下来,我们通过对化合物抗炎、减肥等活性的进一步研究,建立筛选具有相应活性的药物分子的方法。
实施例7.YXY101的抗炎、减肥活性是通过诱导TRAF2转运至线粒体上实现的,这种转运的过程同时是依赖于Nur77的
特异性结合Nur77的YXY101在细胞中能够明显逆转TNFα导致的IκBα降解,所以使用MEF、HepG2细胞,用TNFα和YXY101进行处理。用TNFα(20ng/ml,30min)和YXY101(2μM,1h)处理野生型和Nur77缺失的MEF细胞,运用免疫荧光技术手段观察Nur77、TRAF2在细胞线粒体中的定位情况。
图9A显示了免疫染色结果,在野生型MEF中,单独用TNFα的条件下,TRAF2没有明显聚集的现象,单独使用YXY101的条件下,TRAF2有少量聚集的情况,但当合用TNFα和YXY101时,TRAF2明显聚集并定位于线粒体处,表明YXY101所诱导的抗炎、减肥功能可能通过促进TRAF2转运至线粒体所诱导;同样的处理条件下,Nur77敲除的MEF细胞中,TRAF2的转运现象基本消失。
图9B显示了上述现象的统计结果,在野生型MEF细胞中,对比于对照组,单独使用YXY101的实验组中共定位情况已有差异,而当共加了TNFα时,共定位的情况大幅增加,具有显著差异,而在Nur77缺失的MEF细胞中,对照组和多有实验组之间无明显差异。
用TNFα(20ng/ml)和YXY101(2μM,1h/9h)处理HepG2细胞,分离出线粒体,并用免疫印迹的方法检测全细胞蛋白和线粒体蛋白中的Nur77、TRAF2、p62、LC3、PARP、Hsp60水平。
图9C显示了,YXY101能够显著增加线粒体上Nur77、p62、TRAF2、LC3的含量,进一步证明YXY101能够促进Nur77、p62/TRAF2、LC3转运至线粒体。
综上,我们发现了YXY101的抗炎、减肥机制与促进TRAF2等蛋白转运至线粒体的过程息息相关,且整个过程依赖于Nur77。据此,我们可以通过检测TRAF2等蛋白的线粒体定位,来筛选具有潜在抗炎、减肥活性的药物分子。
实施例8.YXY101通过诱导Nur77与TRAF2在线粒体的相互作用,发挥出抗炎、减肥活性
在本实施例中,我们发现了YXY101能诱导TRAF2的转运,并且这种作用依赖于Nur77,这不仅说明Nur77这个靶点的关键性作用,并且提示Nur77和TRAF2这两个蛋白之间具有千丝万缕的关系。
图10A显示了用TNFα(20ng/ml)和YXY101(2μM)处理HepG2细胞,IP:TRAF2,并用免疫印迹的方法检测Nur77与TRAF2是否具有相互作用。实验结果表明,在细胞水平上,通过TNFα和YXY101处理后,单加TNFα时并不能诱导两者的相互作用,而当共加TNFα和YXY101时,Nur77与TRAF2的相互作用强度被明显增强。
图10B显示了在HepG2细胞中外转Flag-TRAF2质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测内源的Nur77和外源的Flag-TRAF2共定位情况。结果显示,在体外水平,体外提纯的GST-TRAF2、His-Nur77-LBD蛋白能够在YXY101的作用下表现出强烈的相互作用。
图10C显示了在HepG2细胞中外转Flag-TRAF2质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测内源的Nur77和外源的Flag-TRAF2共定位情况。结果显示,在TNFα和YXY101共处理条件下,外源的Flag-TRAF2与内源的Nur77能够在核外形成明显的共定位结构。进一步,图10D显示了在HepG2细胞中外转Flag-TRAF2、GFP-Nur77质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测Flag-TRAF2、GFP-Nur77、线粒体的共定位情况。结果显示,这种核外形成的共定位结构与线粒体共定位,说明YXY101所诱导的Nur77与TRAF2相互作用场所是线粒体。
综上,YXY101的抗炎、减肥机制不仅与TRAF2的转运相关,也能够诱导Nur77与TRAF2在线粒体上的相互作用。据此,我们可以通过检测Nur77与TRAF2的相互作用及其线粒体定位,来筛选具有潜在抗炎、减肥活性的药物分子。
实施例9.YXY101的抗炎、减肥活性与Nur77依赖的自噬过程息息相关
抗炎的机制,可能是通过抑制TNFα诱导NF-κB通路的过度激活相关,也可能是通过自噬作用清除损伤线粒体来实现的。据此,我们检测了与自噬过程密切相关的LC3蛋白。
图11A显示了在HepG2细胞中外转Flag-TRAF2、GFP-LC3质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测Flag-TRAF2、GFP-LC3和线粒体的共定位情况。图11B显示了在HepG2细胞中外转RFP-LC3、GFP-Nur77质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测RFP-LC3、GFP-Nur77和线粒体的共定位情况。结果表明,YXY101能诱导TRAF2与LC3的共定位,也能诱导Nur77与LC3的共定位,并且均是定位于线粒体上。进一步在野生型和敲除Nur77的MEF细胞中验证,图11C显示了用TNFα(20ng/ml)和YXY101(2μM)处理野生型和Nur77缺失的MEF细胞,运用免疫染色技术检测Nur77、LC3和线粒体的共定位情况。结果显示,敲除了Nur77之后,LC3定位于线粒体的现象消失。图11D显示了上述处理的细胞中Nur77、LC3、线粒体共定位的统计结果。图11D的结果也显示出在野生型细胞中,YXY101单独处理和YXY101与TNFα共处理均能增加Nur77、LC3和线粒体三者的共定位,但当敲除Nur77之后,YXY101的诱导作用显著降低。图11C-11D结果表明,上述的共定位过程是依赖于Nur77的。
综上,YXY101能够促进Nur77与LC3的相互作用,促进自噬过程,进而达到抗炎、减肥作用。据此,我们可以通过检测Nur77与LC3的相互作用及其线粒体定位,或者检测自噬过程,以此来筛选具有潜在抗炎、减肥活性的药物分子。
实施例10.YXY101促进Nur77与p62的相互作用,清除损伤线粒体,进而达到抗炎、减肥效果
图12A显示了在HepG2细胞中外转Myc-Nur77、Flag-p62质粒,并用TNFα(20ng/ml)和YXY101(2μM)处理,IP:Flag,并用免疫印迹的方法检测与Flag-p62相互作用的Myc-Nur77。结果表明,YXY101能显著诱导Nur77与p62的相互作用。图12B显示了用TNFα(20ng/ml)和YXY101(2μM)处理HepG2细胞,运用免疫染色技术检测Nur77和p62的共定位情况。图12C显示了在HepG2细胞中外转GFP-LC3质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测GFP-Nur77和p62的共定位情况。图12B-C验证了验证内源或外源的Nur77、p62能在YXY101的诱导下共定位。
结合实施例9,YXY101能够促进Nur77分别与LC3、P62的相互作用,进而促进自噬作用,促进损伤线粒体的清除,最终达到抗炎、减肥效果。据此,我们可以通过检测Nur77与LC3、p62的相互作用,以此来筛选具有潜在抗炎、减肥活性的药物分子。
实施例11.YXY101促进Nur77的泛素化,进而达到抗炎、减肥效果
图13A显示了在HepG2细胞中外转Flag-TRAF2、Myc-Nur77、HA-Ub质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,IP:Myc,用免疫印迹手段检测Nur77的泛素化情况。结果表明,共处理TNFα、YXY101后,不仅能诱导Nur77与TRAF2的相互作用,并且诱导了Nur77的泛素化。图13B显示了在HepG2细胞中外转Myc-Nur77、HA-Ub质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,分离出线粒体,IP:Myc,用免疫印迹手段检测线粒体中的Nur77泛素化情况。图13B验证了YXY101诱导的泛素化修饰是在线粒体上进行的。图13C显示了在HepG2细胞中外转GFP-Nur77、HA-Ub质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测GFP-Nur77和HA-Ub的共定位情况。图13D显示了在HepG2细胞中外转GFP-Nur77质粒,用TNFα(20ng/ml)和YXY101(2μM)处理,运用免疫染色技术检测GFP-Nur77和Ub的共定位情况。图13C-13D说明YXY101能诱导Nur77与Ub共定位到线粒体上。相关数据表明,Nur77在线粒体上的泛素化与自噬过程相关联。
结合实施例9-11,YXY101能诱导Nur77的泛素化、Nur77与p62的相互作用、Nur77与LC3的相互作用,最终引发自噬过程,清除损伤线粒体,抑制炎症。我们可以通过检测Nur77的泛素化,以此来筛选具有潜在抗炎、减肥活性的药物分子。
实施例12.通过检测Nur77与p62、TRAF2、LC3、Ub的相互作用,筛选出新的活性分子
根据实例1-6所述的创新性方法,筛选出了活性分子XS0284。首先如图14A使用Biacore T200仪器,通过SPR来初步筛选出XS0284能够特异性结合Nur77蛋白,图15B进一步用梯度浓度(0.15μM、0.3μM、0.625μM、1.25μM、2.5μM)的XS0284检测与Nur77的结合强度,图15B显示XS0284与Nur77-LBD结合的解离常数(Kd)为386nM。图16A显示了XS0284能够显著逆转TNFα下调IκBα具有明显的抗炎活性。图17A显示了XS0284能诱导Nur77和p62的相互作用。图17B显示了XS0284类似于YXY101均能削弱TRAF2的泛素化。图17C显示了XS0284能诱导GFP-Nur77与Flag-TRAF2共定位于线粒体上。图17D显示了XS0284能诱导GFP-Nur77与RFP-LC3的共定位。图17E显示了XS0284能诱导GFP-Nur77、Flag-p62与Ub的共定位。
综上,我们使用实施例1-6的方法,检测化合物与Nur77的特异性结合;检测化合物逆转TNFα下调IκBα的活性;检测化合物是否能够诱导Nur77与TRAF2、p62、LC3、Ub的相互作用或者共定位;检测化合物是否诱导Nur77、TRAF2、p62、LC3、Ub的线粒体转运;以及检测化合物是否诱导自噬和清除损伤线粒体;以此筛选出化合物XS0284;其具有良好的活性,是潜在抗炎、减肥活性的药物分子。
实施例13.YXY101衍生物具有比YXY101更强更特异的抑制TNFα的生物学效应的功能
进一步的,我们利用上述方法进一步筛选特异性结合Nur77并具有抗炎活性的化合物。图16A显示了用YXY101和不同的YXY101衍生物XS0284、XS0285、XS0286、XS0287、XS0335、XS0366、XS0260、XS0394、XS0395、XS0419、XS0420、XS0421以及TNFα处理的MDA-MB-231细胞中IκBα和PARP的免疫印迹分析结果。图16B 显示了用YXY101和不同的YXY101衍生物XS0284-4、XS0284、XS0077、XS0503、XS0486、XS0419、XS0474、XS0462、XS0285、XS0394、XS0454、XS0455、XS0462、XS0473、XS0474、XS0480以及TNFα处理的MDA-MB-231细胞中IκBα和PARP的免疫印迹分析结果。图16C显示了用YXY101和不同的YXY101衍生物XS0284、XS0285、XS0335、XS0394、XS0418、XS0419、XS0454、XS0455、XS0462、XS0502、XS0503、XS0504、XS0506、XS0507、XS0508、XS0077以及TNFα处理的MDA-MB-231细胞中IκBα和PARP的免疫印迹分析结果。图16A-16C显示了经同一浓度的YXY101和TNFα处理的细胞中IκBα和PARP的免疫印迹分析结果。结果显示,TNFα能够在细胞中诱导IκBα的降解;而YXY101则能够抑制TNFα诱导的IκBα的降解。红色字体代表的衍生物具有比YXY101更强的抗炎效果,包括XS0284、XS0394、XS0077、XS0503、XS0486、XS0462、XS0474、XS0418、XS0419等。更重要的是,YXY101由于诱导较强的PARP切割从而诱导细胞凋亡,使得YXY101很难作为没有毒副作用或毒副作用较低的抗炎药和减肥药用于实际应用中。而在我们的方法筛选得到的YXY101衍生物,具有比YXY101更强的抗炎效果,更小的毒副作用。其中,XS0284是一个典型的例子,比YXY101更显著的抑制TNFα诱导的IκBα的降解,同时,不引起PARP切割,具有较弱的毒副作用,具有更好的成药性。与XS0284相似作用的化合物包括XS0394、XS0503、XS0486、XS0462、XS0474、XS0418、XS0419。这些结果表明,利用我们的方法能筛选到具有比YXY101更强的活性以及更低的细胞毒性的化合物。
实施例14.应用检测Nur77与TRAF2或P62共定位的方法进一步筛选可靠的靶向Nur77的抗炎化合物
图18A显示了经XS0077和TNFα处理的Hela细胞的免疫荧光染色结果。HepG2细胞用XS0077(2μM)处理3小时,然后用TNFα(20ng/mL)处理30分钟。随后,用免疫荧光染色法检测细胞中的Nur77与Traf2的共定位情况。图18B显示了经XS0503和TNFα处理的Hela细胞的免疫荧光染色结果。Hela细胞用XS0503(2μM)处理3小时,然后用TNFα(20ng/mL)处理30分钟。随后,用免疫荧光染色法检测细胞中的Nur77与Traf2的共定位情况。未经处理的细胞用作对照,结果示于图中。 图18A-18B的结果显示,XS0077与XS0503能够促进Nur77与Traf2和p62的共定位。进一步证明可以通过检测Nur77与Traf2共定位的方法来筛选具有良好的抗炎生物活性的化合物。
实施例15.YXY101衍生物对TNFα生物学效应的抑制作用由Nur77介导
为了证明YXY101衍生物对TNFα生物学效应的抑制作用由Nur77介导,我们从图16的结果中选出XS0284、XS0474、XS0503、XS0419、XS0486进行验证。图14A-14C的结果显示,在表达Nur77的MEF细胞中,XS0284、XS0474、XS0503、XS0419、XS0486均能够抑制TNFα诱导的IκBα的降解,在不表达Nur77的MEF细胞中,XS0284、XS0474、XS0503、XS0419、XS0486丧失了抑制IκBα降解的能力。这些结果表明,YXY101衍生物,比如XS0284、XS0474、XS0503、XS0419、XS0486,对TNFα生物学效应的抑制作用是由Nur77介导的。
实施例16.表面等离子共振术(SPR)对其他化合物的表征
根据实施例2中描述的方法,使用Biacore T200仪器,通过SPR来检测多种YXY101衍生物物(XS0284、XS0394、XS0503、XS0486、XS0462、XS0474、XS0418、XS0419)与Nur77-LBD的结合。结果示于图15中。
图15显示了用Biacore T200仪器检测不同化合物(XS0284、XS0394、XS0503、XS0486、XS0462、XS0474、XS0418、XS0419)与Nur77-LBD的结合的实验结果。结果显示,化合物XS0284与Nur77-LBD结合的解离常数(Kd)为386nM;XS0394与Nur77-LBD结合的解离常数(Kd)为1.26μM;化合物XS0418与Nur77-LBD结合的解离常数(Kd)为3.67μM;化合物XS0462与Nur77-LBD结合的解离常数(Kd)为2.07μM;化合物XS0474与Nur77-LBD结合的解离常数(Kd)为2.60μM;化合物XS0486与Nur77-LBD结合的解离常数(Kd)为1.20μM;化合物XS0419与Nur77-LBD结合的解离常数(Kd)为404nM;化合物XS0503与Nur77-LBD结合的解离常数(Kd)为1.63μM。
综上所述,通过SPR检测与Nur77特异性结合的化合物的方法进一步筛选具有抗炎促进自噬活性的化合物是可靠的,筛选得到的化合物具有很强的靶点特异性和专一 性。
实施例17.XS0284的对细胞的毒副作用作用低于YXY101
为了证明YXY101衍生物对细胞的毒性低于YXY101,我们选取其中抗炎效果出色的化合物之一XS0284进行验证。图19A显示了用YXY101和XS0284以及TNFα处理的MDA-MB-231细胞中PARP的免疫印迹分析结果。图19B显示了用YXY101和XS0284以及TNFα处理的NCL-H292细胞中PARP的免疫印迹分析结果。图19C显示了化合物YXY101和XS0284对HepG2细胞的增殖抑制率。图19A-19B的结果显示,同样的浓度(4μM)和时间(9h)条件下,YXY101能明显促进PARP切割,而XS0284不能诱导PARP的切割。图19C的结果显示,用同一浓度的化合物YXY101和XS0284处理HepG2细胞,XS0284对细胞的增殖抑制率低于YXY101。
我们还通过流式细胞术检测JC-1来进一步明确YXY101和XS0284对细胞线粒体膜电位的作用。在线粒体膜电位较高时,JC-1能聚集在线粒体的基质中,所形成的聚合物能产生红色荧光;在线粒体膜电位较低时,JC-1不能聚集在线粒体的基质中,而以单体形式存在,产生绿色荧光。故而可通过检测荧光颜色的转变来检测线粒体膜电位的变化。我们在MDA-MB231细胞上,以5μM的药物处理24小时后,用JC-1染线粒体膜电位,通过流式细胞技术检测FITC与PE的表达情况来检测细胞的线粒体膜电位,发现YXY101会引起线粒体膜电位的降低,而XS0284则几乎没有影响。
这些结果表明YXY101的系列衍生物,例如XS0284具有比YXY101更低的细胞毒作用。一方面,这提示着这系列化合物具有比YXY101更强的靶向性和专一性;另一方面,这也提示着这系列YXY101衍生物作为开发成更安全的抗炎减肥药的可能。
实施例18.XS0284的药物急性毒副作用低于YXY101
本实施例采用,一次性灌胃注射200mg/kg的YXY101及其衍生物XS0284或腹腔注射20mg/kg的YXY101及其衍生物XS0284后诱发的小鼠急性毒性模型。灌胃或腹腔注射YXY101和XS0284后,分别观察各组小鼠进食量、饮水、自发活动能力、精神状态、四肢活动、大小便质量、毛发光泽等,并且详细记录可能出现的一些毒性反 应化及开始和消失时间点。组织病理学观察心脏、肝脏、小肠、白色脂肪和肾脏的组织形态。
实验结果实验图20中。图20A显示了灌胃组:1)空白组小鼠的心脏心肌细胞界限清晰,胞核多且明显,心肌纤维排列整齐,界限明显;肝组织中肝窦数量多且清楚,肝小叶界限明显;肠组织界限清晰,细胞饱满,肠绒毛排列整齐、界限明显;脂肪组织细胞界限清晰、排列整齐。2)与空白组比,YXY101组小鼠的心肌细胞和心肌纤维排列杂乱;肝小叶界限模糊,胞浆减少,肝细胞多数出现死亡;肠组织肿胀明显,细胞死亡增多,炎症反应明显,肠绒毛排列紊乱;脂肪组织细胞排列杂乱,个别纤维断裂。3)与空白组比,XS0284组小鼠的心脏心肌细胞界限相对清晰,心肌纤维相对排列整齐,界限明显;肝组织中肝细胞界限明显,结构清晰;肠组织界限清晰,细胞饱满,肠绒毛排列整齐、界限明显;脂肪组织细胞界限清晰、排列整齐,纤维断裂减少。由此可以得出衍生物XS0284组显示对肝脏和小肠的毒性作用比YXY101更弱。
图20B显示了腹腔注射组1)空白组小鼠的心脏心肌细胞界限清晰,胞核多且明显,心肌纤维排列整齐,界限明显;肝组织中肝窦数量多且清楚,肝小叶界限明显;脂肪组织细胞界限清晰、排列整齐;肾脏组织肾小球界限清晰,细胞饱满,排列整齐、界限明显。2)与空白组比,YXY101组小鼠的心肌细胞和心肌纤维排列杂乱;肝小叶界限基本明显,肝细胞饱满形态正常;但脂肪组织细胞排列杂乱,且有大量炎性细胞浸润;肾脏组织肾小球界限模糊,且有大量炎性细胞浸润,排列不整齐。3)与空白组比,XS0284组小鼠的心脏心肌细胞界限相对清晰,心肌纤维相对排列整齐,界限明显;肝组织中肝细胞界限明显,结构清晰;脂肪组织细胞界限清晰、排列整齐,纤维断裂减少;肾脏组织肾小球界限清晰,细胞饱满,有少量炎性细胞浸润。由此可以得出腹腔注射组衍生物XS0284组显示对肾脏和脂肪的毒性作用比YXY101更弱。
这些结果表明YXY101的系列衍生物,例如XS0284具有比YXY101更低的动物急性毒作用。进一步提示着这系列化合物具有比YXY101更强的靶向性和专一性,以及这系列YXY101衍生物作为开发成更安全的抗炎减肥药的可能。
实施例19.XS0284具有比YXY101更温和的抑制肥胖的效果
本实施例腹腔注射YXY101(0.1mg/kg)、XS0284(0.1mg/kg)高脂诱导的肥胖小鼠一周的时间。结果显示,YXY101能显著减少体重并改善HFD诱导的脂肪肝,但是因减肥速度过快而导致体型消瘦,而XS0284也具有明显减少体重的效果,但是其减肥效果更加温和,小鼠并未表现出明显消瘦状态,从图21C的体重曲线可以看出,XS0284能更加温和并以稍微缓慢的速度减少体重。在此基础上从图21A可以看出XS0284也能有效改善脂肪肝。
这些结果表明YXY101衍生物(例如XS0284)具有可与之比拟的减肥作用、预防脂肪肝作用,同时相比于YXY101其衍生物的药效更加温和,这一点提示着这系列化合物相比YXY101更具有开发为减肥药物的价值。
实施例20.
利用计算机辅助分子对接技术,以Nur77为靶点。对海洋天然产物进行虚拟筛选从中获得一些结合较好的化合物,具体如下所示。
Figure PCTCN2017091716-appb-000070
该实例中利用计算机辅助进行虚拟筛选的步骤如下:下载蛋白共晶结构PDB ID:3V3Q,蛋白预处理,加氢,去除结晶水分子和晶体结构中甘油小分子;选中配体,以配体为中心,12angstrom的大小构建对接grid格点文件;运行Glide docking,选中事先构建的grid格点文件和处理好的Seaweed Metabolism Database进行对接;根据对接打分和化 合物与蛋白受体之间的相互作用,对对接结果进行综合评价。模拟结合的作用模式图示例见图22。
化合物XS0077的制备
Figure PCTCN2017091716-appb-000071
首先将化合物YXY101(50mg,0.11mmol)搅拌溶解于2mL DMF中,随后加入碳酸氢钠(56mg,0.66mmol),碘甲烷(42μL,0.66mmol)室温搅拌反应12小时。1mol/L HCl(1mL)淬灭反应,加入9mL纯水,用乙酸乙酯萃取3次(每次5mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯,得到橘红色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=10∶1)为洗脱剂,300-400目硅胶填充柱层析分离,得到橘红色固体产物。
1H NMR(600MHz,DMSO-d6)δppm 0.44(s,3H),0.91(d,J=14.31Hz,1H),1.07(s,3H),1.12(s,3H),1.21(s,3H),1.30-1.35(m,1H),1.38(s,3H),1.41-1.46(m,1H),1.50-1.59(m,3H),1.61-1.72(m,4H),1.78-1.86(m,1H),1.95(td,J=13.98,3.76Hz,1H),2.06(d,J=14.12Hz,1H),2.09(s,3H),2.17-2.22(m,1H),2.31(d,J=15.77Hz,1H),3.48(s,3H),6.35(d,J=7.15Hz,1H),6.39(d,J=1.28Hz,1H),7.07(dd,J=7.15,1.10Hz,1H),8.72(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.10,17.96,21.41,28.08,29.19,29.42,30.12,30.34,31.34,32.23,32.91,34.39,36.02,37.83,38.8,39.83,41.99,43.64,44.48,51.44,117.26,118.05,120.18,126.89,133.13,146.42,162.94,167.80,177.93,177.96.
实施例21.化合物XS0284的制备
Figure PCTCN2017091716-appb-000072
在氮气环境下,在搅拌条件下,将化合物YXY101(50mg,0.11mmol)溶于甲醇(2.5mL)。加入亚硫酸氢钠水溶液(14mg,0.13mmol,溶解于1mL水中),并于室温下反应3小时。反应后,用旋转蒸发仪浓缩反应体系得无色晶体。用甲醇/水对无色晶体进行重结晶,所获得的产物经真空干燥得呈白色固体的化合物XS0284(56.6mg)。
1H NMR(600MHz,DMSO-d6)δppm 0.59(s,3H)0.86(d,J=12.10Hz,1H)1.05(s,3H)1.09(s,3H)1.18(s,3H)1.43-1.60(m,6H)1.62(s,3H)1.78-1.85(m,1H)1.93-2.04(m,3H)2.21(s,3H)2.32(d,J=15.22Hz,1H)4.48(d,J=6.24Hz,1H)5.81(d,J=6.60Hz,1H)6.58(s,1H)7.61(br.s.,1H)8.81(br.s.,1H).
13C NMR(151MHz,DMSO-d6)δppm 13.57,18.46,21.64,29.04,29.98,30.47,30.60,30.64,31.94,32.92,34.80,35.11,36.90,36.96,37.90,38.07,43.95,44.39,60.19,108.96,118.87,123.26,124.39,140.89,141.93,144.02,150.07,180.00.
实施例22.化合物XS0285的制备
Figure PCTCN2017091716-appb-000073
在搅拌条件下,将化合物YXY101(50mg,0.11mmol)溶于N,N-二甲基甲酰胺(2mL)。加入碳酸氢钠(50.4mg,0.6mmol),然后加入苄基溴(0.17mg,20μL),并于室温下搅拌反应24小时。停止反应,向反应体系中加入去离子水(15mL),并用 乙酸乙酯萃取3次。合并乙酸乙酯层,用饱和NaCl水溶液洗涤3次,用无水Na2SO4干燥,用旋转蒸发仪浓缩得粗产物(深红棕色油状物)。粗产物经快速柱层析法(乙酸乙酯∶正己烷)分离纯化,真空干燥得呈红色固体的化合物XS-0285(44mg)。
1H NMR(600MHz,CHLOROFORM-d)δppm 0.50(s,3H)0.97(d,J=13.75Hz,1H)1.09(s,3H)1.21(s,3H)1.22-1.25(m,3H)1.25-1.28(m,1H)1.41(s,3H)1.47-1.58(m,3H)1.58-1.72(m,5H)1.87(d,J=6.05Hz,1H)1.99-2.11(m,3H)2.21(d,J=1.65Hz,3H)2.24(d,J=14.12Hz,1H)2.44(d,J=15.77Hz,1H)4.93(d,J=12.29Hz,1H)5.02(d,J=12.47Hz,1H)6.32(d,J=7.15Hz,1H)6.49(s,1H)7.01(d,J=6.97Hz,1H)7.27-7.30(m,2H)7.30-7.36(m,3H).
13C NMR(151MHz,CHLOROFORM-d)δppm 10.29,18.53,21.58,28.61,29.53,29.90,30.55,30.76,31.57,32.76,33.27,34.69,36.35,38.25,39.43,40.44,42.93,44.25,45.03,66.32,117.36,118.12,119.62,127.38,128.24,128.32,128.64,134.24,135.68,146.06,164.79,170.27,177.95,178.36.
实施例23.化合物XS0335的制备
Figure PCTCN2017091716-appb-000074
在搅拌条件下,将化合物YXY101(50mg,0.11mmol)溶于二氯甲烷(1mL)。加入钯碳(5mg),再加入二氯甲烷(1mL),持续通入氢气,并在室温下搅拌反应24小时。停止反应,向反应体系中加入去离子水(15mL),并用乙酸乙酯萃取3次。合并乙酸乙酯层,然后用饱和NaCl洗3次,用无水Na2SO4干燥,用旋转蒸发仪浓缩得粗产物(无色油状物)。粗产物经快速柱层析法(乙酸乙酯∶正己烷)分离纯化,真空干燥得呈白色固体的化合物XS-0335(46mg)。
1H NMR(600MHz,METHANOL-d4)δppm 0.93-1.00(m,2H)1.03(s,3H)1.11 (s,3H)1.19(t,J=3.48Hz,4H)1.26(s,3H)1.41-1.45(m,6H)1.47-1.55(m,2H)1.57(d,J=8.25Hz,1H)1.59-1.68(m,2H)1.83-1.98(m,3H)2.06-2.11(m,4H)2.11-2.20(m,4H)2.43(d,J=15.77Hz,1H)2.68(d,J=14.67Hz,1H)6.67(s,1H).
13C NMR(151MHz,METHANOL-d4)δppm 11.94,18.51,20.54,26.82,28.66,30.74,31.15,31.57,31.61,32.02,32.09,32.32,33.63,34.65,37.09,37.56,38.53,39.45,39.85,41.69,58.12,58.48,112.24,121.74,129.36,141.47,142.39,144.19,182.95.
实施例24.化合物XS0366,XS0434-XS0438,XS0440,XS0441,XS0443,XS0463和XS0464的制备
Figure PCTCN2017091716-appb-000075
以XS0366为例,在搅拌条件下,将化合物YXY101(100mg,0.22mmol)溶于二氯甲烷(4mL)。加入吲哚(52mg,0.44mmol),再加入六水合三氯化铝(5.3mg,0.022mmol),并在室温下搅拌反应5小时。停止反应,向反应体系中加入去离子水(15mL),并用乙酸乙酯萃取3次。合并乙酸乙酯层,然后用饱和NaCl洗3次,用无水Na2SO4干燥,用旋转蒸发仪浓缩得粗产物(棕色油状物)。粗产物经快速柱层析法(乙酸乙酯∶正己烷)分离纯化,真空干燥得呈紫红色固体的化合物XS0366(122.2mg)。
1H NMR(600MHz,CHLOROFORM-d)δppm 0.73(br.s.,3H)0.87-0.90(m,1H)0.95-1.00(m,3H)1.01(br.s.,3H)1.14(br.s.,3H)1.19(t,J=7.06Hz,1H)1.25-1.27(m,1H)1.34(br.s.,3H)1.42-1.57(m,4H)1.57-1.76(m,4H)1.90(s,3H)1.99-2.07(m,2H)2.10-2.17(m,1H)2.40(d,J=15.04Hz,1H)4.90(d,J=5.69Hz,1H) 6.21(d,J=6.24Hz,1H)6.23(br.s.,1H)6.79(br.s.,1H)7.11(t,J=7.43Hz,1H)7.16(t,J=7.43Hz,1H)7.28(d,J=7.89Hz,1H)7.75(d,J=7.70Hz,1H)7.88(br.s.,1H).
13C NMR(151MHz,CHLOROFORM-d)δppm 11.53,18.84,21.93,28.89,29.63,29.75,30.40,30.54,30.72,31.55,32.84,34.62,35.50,36.74,36.94,37.76,40.35,43.62,44.28,108.93,111.30,119.09,119.28,120.23,121.55,121.58,121.67,127.10,127.84,136.49,139.92,142.17,142.83,147.48,184.30.
根据上述方法,本发明还合成了下述化合物:
Figure PCTCN2017091716-appb-000076
实施例25.化合物XS0395的制备
Figure PCTCN2017091716-appb-000077
称取化合物YXY101(50mg,0.11mmol)于25ml反应瓶中,加入4ml丙酮搅拌溶解,再加入一滴浓盐酸作催化剂,室温条件下反应12h。反应停止,直接将反应液减压浓缩溶剂,用乙酸乙酯∶正己烷=4∶1体系经硅胶柱层析分离纯化,得白色固体,产率51%。
1H NMR(600MHz,DMSO-d6)δppm 0.62(s,3H),0.82-1.86(m,1H),1.04(s,3H),1.09(s,3H),1.15(s,3H),1.25-1.38(m,4H),1.39(s,3H),1.42-1.50(m,2H),1.55-1.71(m,4H),1.77(td,J=13.9,6.1Hz,1H),1.93-2.02(m,2H),2.03(s,3H),2.09(s,3H),2.27-2.36(m,2H),2.67(dd,J=16.2,2.7Hz,1H),3.71-3.78(m,1H),5.72(d,J=6.4Hz,1H),6.63(s,1H),7.91(s,1H),8.94(s,1H),12.06(br.s.,1H).
13C NMR(151MHz,DMSO-d6)δppm 11.64,18.43,22.63,29.01,29.90,30.36,30.47,30.62,30.77,31.44,31.87,32.88,32.92,34.94,35.60,36.79,36.85,36.98,37.74,39.87,43.74,44.36,51.72,109.06,120.00,122.04,126.53,140.46,141.62,143.85,149.91,179.96.208.01.
实施例26.化合物XS0419的制备
Figure PCTCN2017091716-appb-000078
将化合物YXY101(50mg,0.11mmol)搅拌溶解于2mL氘代甲醇溶剂中,随后加 入硼氢化钠(44mg,1.1mmol),室温搅拌反应30min。1mol/L HCl(1mL)猝灭反应,加入9mL纯水,用二氯甲烷萃取三次(每次5mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏快速除去有机溶剂二氯甲烷,得到白色固体的化合物XS0419(50.1mg)。
1H NMR(600MHz,DMSO-d6)ppm 0.66(s,3H)0.85(d,J=13.9Hz,1H)1.05(s,3H)1.11(s,3H)1.17(s,3H)1.22(s,3H)1.29(ddd,J=13.7,4.4Hz,1H)1.36-1.41(m,1H)1.43-1.51(m,3H)1.53-1.59(m,1H)1.59-1.63(m,1H)1.63-1.69(m,1H)1.79(ddd,J=13.8,6.5Hz,1H)1.86(ddd,J=13.8,5.0Hz,1H)1.94-2.00(m,2H)2.01(s,3H)2.04(d,J=13.6Hz,1H)2.34(d,J=15.6Hz,1H)2.91(dd,J=20.0,1.5Hz,1H)3.18(dd,J=20.5,6.2Hz,1H)5.72(dd,J=6.1,1.8Hz,1H)6.61(s,1H)7.82(s,1H)8.80(s,1H)12.05(br.s.,1H).
13C NMR(151MHz,DMSO-d6)ppm 11.53,18.10,22.70,27.30,28.45,29.46,29.77,30.08,30.19,31.39,32.44,34.06,34.09,34.40,36.07,36.56,37.14,39.44,43.25,43.83,108.17,117.67,120.10,123.10,139.35,140.56,143.11,149.22,179.51.
实施例27.化合物XS0462的制备
Figure PCTCN2017091716-appb-000079
首先将雷公藤红素(135.2mg,0.3mmol)搅拌溶解于2mL DMF中,随后加入碳酸氢钠(138.6mg,1.65mmol),溴乙烷(234μL,0.15mmol)室温搅拌反应12小时。1mol/L HCl(1mL)淬灭反应,加入9mL纯水,用乙酸乙酯萃取3次(每次15mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯,得到橘红色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=10∶1)为洗脱剂,300-400目硅胶填充柱层析分离,得到橘红色固体产物。
1H NMR(600MHz,DMSO-d6)δppm 0.47(s,3H),0.91(d,J=14.1Hz,1H),1.07(s, 3H),1.11(s,3H),1.12-1.15(m,3H),1.21(s,3H),1.31-1.36(m,1H),1.38(s,3H),1.41-1.46(m,1H),1.52-1.58(m,3H),1.61-1.71(m,4H),1.78-1.87(m,1H),1.90-1.99(m,1H),2.03-2.08(m,1H),2.09(s,3H),2.21(d,J=11.2Hz,1H),2.34(d,J=15.6Hz,1H),3.91(m,2H),6.35(d,J=7.2Hz,1H),6.39(s,1H),7.05-7.10(m,1H),8.73(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.53,14.28,18.50,21.84,28.55,29.57,29.75,30.58,31.76,32.76,33.32,34.78,36.43,38.20,39.27,40.13,40.44,42.43,44.04,44.93,60.33,117.74,118.51,120.56,127.29,133.67,146.86,163.40,168.31,177.83,178.41.
实施例28.化合物XS0474的制备
Figure PCTCN2017091716-appb-000080
将化合物YXY101(50mg,0.11mmol)搅拌溶解于2mL氘代甲醇溶剂中,随后加入硼氘化钠(48.4mg,1.1mmol),室温搅拌反应30min。1mol/L HCl(1mL)猝灭反应,加入9mL纯水,用二氯甲烷萃取三次(每次5mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏快速除去有机溶剂二氯甲烷,得到白色固体的化合物XS0474(50.2mg)。
1H NMR(600MHz,DMSO-d6)ppm 0.66(s,3H)0.85(d,J=12.84Hz,1H)1.04(s,3H)1.10(s,3H)1.17(s,3H)1.22(s,3H)1.29(td,J=13.66,4.40Hz,1H)1.35-1.41(m,1H)1.43-1.52(m,3H)1.53-1.58(m,1H)1.58-1.63(m,1H)1.63-1.69(m,1H)1.79(td,J=13.66,6.60Hz,1H)1.86(td,J=13.71,5.04Hz,1H)1.94-1.99(m,2H)2.01(s,3H)2.04(d,J=12.10Hz,1H)2.34(d,J=15.59Hz,1H)3.16(d,J=6.05Hz,1H)5.71(d,J=6.24Hz,1H)6.61(s,1H)7.82(s,1H)8.83(s,1H)12.03(br.s.,1H).
13C NMR(151MHz,DMSO-d6)ppm 11.60,18.15,22.74,26.98,28.52,29.51,29.83,30.14,30.25,31.45,32.50,34.15,34.21,34.46,36.14,36.62,37.21,39.49,43.31,43.89, 108.23,117.68,120.20,123.11,139.45,140.60,143.18,149.32,179.59.
实施例29.化合物XS0503的制备
Figure PCTCN2017091716-appb-000081
首先将扁蒴藤素(250mg,0.54mmol)溶解于20mL四氢呋喃中,随后加入LiAlH4(1.2mL,1.1mmol),室温搅拌反应2h,加入10mL去离子水猝灭反应,1mol/L HCl(5mL)酸化,用乙酸乙酯萃取3次(每次15mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯,得到橙黄色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=2∶1)为洗脱剂,300-400目硅胶填充柱层析分离,得到橙黄色固体产物。
1H NMR(600MHz,DMSO-d6)ppm 0.76(s,3H)0.83-0.87(m,1H)0.89(s,3H)1.11(s,3H)1.21(s,3H)1.23-1.25(m,1H)1.26(s,3H)1.27-1.34(m,2H)1.48(dd,J=6.7,3.9Hz,1H)1.50-1.54(m,1H)1.55-1.59(m,1H)1.60(d,J=5.0Hz,1H)1.64(dd,J=9.2,4.6Hz,2H)1.65-1.69(m,2H)1.69-1.72(m,1H)1.91(d,J=5.3Hz,1H)1.93-1.97(m,1H)2.02(s,3H)2.92(d,J=19.4Hz,1H)2.96(dd,J=10.3,4.8Hz,1H)3.15-3.20(m,1H)3.21(t,J=6.0Hz,1H)4.44(t,J=5.0Hz,1H)5.73(dd,J=6.1,1.7Hz,1H)6.61(s,1H)7.80(s,1H)8.78(s,1H).
13C NMR(151MHz,CHLOROFORM-d)ppm 11.49,19.35,25.65,27.80,28.10,28.75,29.36,30.31,30.45,30.57,32.27,32.89,33.58,34.24,36.55,36.82,36.95,37.68,42.91,43.15,71.82,108.42,118.15,120.42,125.27,139.96,141.09,141.87,151.04.
实施例30.化合物XS0508的制备
Figure PCTCN2017091716-appb-000082
称取XS0077(50mg,0.11mmol)于50ml圆底瓶中,加入3ml甲醇溶解,置换气体,氮气保护,加入亚硫酸氢钠溶液(28mg NaHSO3-1ml H2O,0.26mmol),室温下氮气保护反应3h,停止反应,减压蒸馏浓缩溶剂,加入吡啶溶解残留固体,抽滤,减压浓缩得白色固体,产率90%。
1H NMR(600MHz,DMSO-d6)δppm 0.57(s,3H)1.07(s,3H)1.12-1.14(m,1H)1.14-1.16(m,1H)1.17(s,3H)1.21(s,3H)1.22-1.23(m,2H)1.24-1.28(m,4H)1.28-1.32(m,2H)1.38(s,3H)1.40-1.46(m,2H)1.47-1.52(m,2H)1.57(d,J=6.79Hz,1H)1.59-1.63(m,4H)1.64-1.72(m,6H)1.75-1.83(m,4H)1.92-1.96(m,1H)1.97-2.01(m,1H)2.03-2.07(m,1H)2.09(s,3H)2.18(d,J=10.82Hz,2H)2.73(d,J=15.04Hz,1H)3.40-3.49(m,1H)3.80-3.89(m,1H)6.36(d,J=7.34Hz,1H)6.40(d,J=0.92Hz,1H)7.07(dd,J=6.97,0.92Hz,1H)7.73(d,J=8.07Hz,1H)8.71(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.08,18.32,21.83,24.68,24.75,25.13,25.29,25.63,25.67,28.38,28.59,29.91,30.13,31.02,31.08,31.39,31.82,31.91,31.93,32.36,33.10,36.02,36.13,37.67,38.99,42.16,42.81,44.45,44.49,50.05,54.77,117.23,117.93,120.04,126.75,133.25,146.43,153.97,163.09,168.74,175.44,177.83.
实施例31.化合物XS0536的制备
Figure PCTCN2017091716-appb-000083
称取XS0077(150mg,0.32mmol)于厚壁耐压管中,加入碳酸钾(220mg,1.6mmol),加入4ml丙酮室温下搅拌溶解样品,再加入100μL硫酸二甲酯溶液,转移至70℃油浴加热8h。加入1mol/L HCl淬灭,加水,乙酸乙酯萃取3次,无水硫酸钠干燥,旋转蒸发仪浓缩溶剂,用乙酸乙酯∶正己烷=1∶20体系经硅胶柱层析分离纯化得到纯品,白色固体,产率为21%。
1H NMR(600MHz,CHLOROFORM-d)d ppm 0.61(s,3H)0.94(d,J=13.75Hz,1H)1.08(s,3H)1.17(s,3H)1.22(s,3H)1.34(s,3H)1.40(dd,J=14.12,4.22Hz,1H)1.44(dd,J=14.40,2.84Hz,1H)1.51-1.57(m,2H)1.57-1.63(m,1H)1.63-1.69(m,2H)1.73(d,J=11.37Hz,1H)1.85(td,J=13.71,6.33Hz,1H)2.01-2.07(m,1H)2.07-2.13(m,2H)2.17(s,3H)2.18-2.23(m,1H)2.44(d,J=15.59Hz,1H)3.02(d,J=19.99Hz,1H)3.28(dd,J=20.00,5.87Hz,1H)3.54(s,3H)3.77(s,3H)3.87(s,3H)5.77(d,J=4.58Hz,1H)6.78(s,1H).
13C NMR(151MHz,CHLOROFORM-d)d ppm 11.77,18.28,22.73,27.85,28.85,29.87,30.29,30.50,30.85,31.55,32.88,34.16,34.51,34.79,36.83,37.15,37.52,40.45,43.69,44.33,51.47,55.88,60.30,106.30,117.63,125.49,127.80,144.62,144.80,149.13,150.89,179.05.
实施例32.化合物XS0286的制备
Figure PCTCN2017091716-appb-000084
在搅拌条件下,将化合物YXY101(50mg,0.11mmol)溶于N,N-二甲基甲酰胺(2mL)。加入催化剂EDCI(85.4mg,0.55mmol)和HOBT(74.3mg,0.55mmol),搅拌使其溶解;加入对叔丁基苯胺(49.2mg,0.33mmol),并在室温下搅拌反应36小时。停止反应,向反应体系中加入去离子水(15mL),并用乙酸乙酯萃取3次。 合并乙酸乙酯层,然后用饱和NaCl洗3次,用无水Na2SO4干燥,用旋转蒸发仪浓缩得粗产物(暗红色油状物)。粗产物经快速柱层析法(乙酸乙酯∶正己烷)分离纯化,真空干燥得呈暗红色固体的化合物XS0286(10mg)。
1H NMR(600MHz,CHLOROFORM-d)δppm 0.64(s,3H)1.08(d,J=13.20Hz,1H)1.15(s,3H)1.24-1.27(m,6H)1.40-1.42(m,4H)1.48-1.57(m,3H)1.63(m,5H)1.65-1.79(m,5H)1.85(d,J=11.92Hz,1H)1.91(d,J=6.24Hz,1H)1.98-2.06(m,3H)2.08(d,J=12.10Hz,2H)2.18(d,J=1.65Hz,3H)2.54(d,J=15.77Hz,1H)6.29(d,J=6.79Hz,1H)6.45(s,1H)6.92-7.00(m,2H)7.30-7.34(m,2H)7.34-7.37(m,2H)7.37-7.41(m,1H).
13C NMR(151MHz,CHLOROFORM-d)δppm 10.21,18.49,21.75,28.58,29.63,29.69,30.41,30.83,31.33,33.24,33.73,34.35,34.98,36.32,38.06,39.38,41.10,42.96,44.47,45.11,76.81,77.02,77.23,116.99,117.95,119.53,119.81,125.89,127.38,133.95,135.11,146.01,147.26,164.68,170.02,175.83,178.38.
实施例33.化合物XS0287的制备
Figure PCTCN2017091716-appb-000085
在搅拌条件下,将化合物YXY101(50mg,0.11mmol)溶于N,N-二甲基甲酰胺(2mL)。加入催化剂EDCI(85.4mg,0.55mmol)和HOBT(74.3mg,0.55mmol),搅拌使其溶解;加入对甲氧基苯胺(40.6mg,0.33mmol),并在室温下搅拌反应36小时。停止反应,向反应体系中加入去离子水(15mL),并用乙酸乙酯萃取3次。合并乙酸乙酯层,然后用饱和NaCl洗3次,用无水Na2SO4干燥,用旋转蒸发仪浓缩得粗产物(暗红色油状物)。粗产物经快速柱层析法(乙酸乙酯∶正己烷)分离纯化,真空干燥得呈暗红色固体的化合物XS-0287(20mg)。
1H NMR(600MHz,CHLOROFORM-d)δppm 0.53(s,3H)1.05(d,J=13.94Hz,1H)1.13(s,3H)1.21(s,3H)1.24(s,3H)1.26(s,3H)1.35(s,3H)1.48-1.54(m,2H)1.58(d,J=7.34Hz,2H)1.64-1.72(m,2H)1.78-1.89(m,3H)2.03-2.11(m,2H)2.18(s,3H)2.55(d,J=15.77Hz,1H)3.79(s,3H)6.25(d,J=7.15Hz,1H)6.34(s,1H)6.91(m,J=8.99Hz,2H)6.96(d,J=7.15Hz,1H)7.40(m,J=8.80Hz,2H)7.48(s,1H).
13C NMR(151MHz,CHLOROFORM-d)δppm 10.23,18.30,21.75,28.46,29.39,29.70,30.30,30.79,31.52,32.86,33.81,34.89,36.21,37.99,39.28,40.96,42.96,44.43,45.12,55.48,114.21,117.18,117.82,119.38,121.99,127.34,130.98,134.06,146.06,156.41,164.80,170.23,175.88,178.42.
实施例34.化合物XS0394的制备
Figure PCTCN2017091716-appb-000086
在搅拌条件下,将化合物YXY101(50mg,0.11mmol)和三乙胺(37μL)溶于重蒸四氢呋喃中(3.0mL,0.26mmol),然后冷却至低温下-30℃,10分钟后加入逐滴加入氯甲酸乙酯(22μL,0.23mmol)加入至反应液中。-30℃搅拌反应12小时后,停止反应,砂芯漏斗过滤除去不溶物,四氢呋喃洗涤,得黄色滤液。向黄色滤液中加入10mL纯水,用乙酸乙酯萃取3次(每次15mL),合并有机层,将有机层用饱和食盐水洗三次(每次30mL),有机相无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂,得到橘黄色油状液体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=10∶1)为洗脱剂,300-400目硅胶填充柱层析分离,得到黄色固体产物。
1H NMR(600MHz,DMSO-d6)ppm 1.05-1.07(m,1H)1.08(s,3H)1.09-1.12(m,1H)1.18(s,3H)1.23(s,3H)1.27(t,J=7.06Hz,3H)1.28-1.31(m,1H)1.41(s,3H)1.43-1.50(m,4H)1.53(dd,J=12.10,5.69Hz,1H)1.59(d,J=7.89Hz,1H)1.65(d,J=10.64Hz, 2H)1.70-1.78(m,2H)1.79-1.85(m,1H)1.90(td,J=13.98,3.39Hz,1H)1.95-2.01(m,1H)2.01-2.06(m,1H)2.15(s,3H)2.26(d,J=15.22Hz,1H)4.21(q,J=1.00Hz,2H)6.30(d,J=7.15Hz,1H)6.40(d,J=0.73Hz,1H)7.29(dd,J=7.52,0.73Hz,1H)12.07(br.s,1H).
13C NMR(151MHz,CHLOROFORM-d)ppm 11.17,14.09,19.00,21.96,28.60,29.35,29.36,30.47,30.67,31.49,32.67,33.38,34.53,36.20,38.24,39.13,40.16,42.88,44.05,45.34,65.05,117.76,117.78,122.70,126.08,133.37,142.54,152.62,163.30,172.96,183.86,183.90.
实施例35.化合物XS0418的制备
Figure PCTCN2017091716-appb-000087
首先将化合物YXY101(50mg,0.11mmol)搅拌溶解于2mL四氢呋喃中,随后加入碳酸钾(15.2mg,0.11mmol),溴乙酸乙酯(18.37mg,0.11mmol)溶于1mL四氢呋喃中,然后逐滴加入反应液,室温搅拌反应4小时。1mol/L HCl(1mL)淬灭反应,加入9mL纯水,用乙酸乙酯萃取3次(每次15mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯,得到橘红色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=10∶1)为洗脱剂,300-400目硅胶填充柱层析分离,得到橘红色固体产物。
1H NMR(600MHz,CHLOROFORM-d)ppm 0.53(s,3H)0.99(d,J=14.31Hz,1H)1.11(s,3H)1.25(t,J=7.20Hz,3H)1.27(s,3H)1.28(s,3H)1.39-1.44(m,1H)1.45(s,3H)1.51(dd,J=14.95,4.13Hz,1H)1.54-1.58(m,1H)1.60(d,J=7.89Hz,1H)1.63-1.67(m,1H)1.67-1.71(m,1H)1.75(dd,J=15.96,8.07Hz,1H)1.80-1.82(m,1H)1.83-1.85(m,1H)1.85-1.92(m,1H)2.06(td,J=14.12,3.85Hz,1H)2.13-2.18(m,1H)2.21 (s,3H)2.25(d,J=14.31Hz,1H)2.48(d,J=15.96Hz,1H)4.19(q,J=7.15Hz,2H)4.41(d,J=15.77Hz,1H)4.54(d,J=15.96Hz,1H)6.35(d,J=7.15Hz,1H)6.53(d,J=1.10Hz,1H)7.02(dd,J=7.06,1.19Hz,1H)
13C NMR(151MHz,CHLOROFORM-d)ppm 10.23,14.04,18.57,21.61,28.59,29.57,29.74,30.47,30.63,31.55,32.56,33.50,34.67,36.31,38.24,39.39,40.40,42.91,44.19,45.01,60.52,61.28,117.15,118.13,119.52,127.37,134.09,145.98,164.74,167.70,170.03,177.53,178.31
实施例36.化合物XS0421,XS0457,XS0473和XS0493的制备
Figure PCTCN2017091716-appb-000088
以XS0421合成为例:称取化合物YXY101(50mg,0.11mmol)于50ml密封管中,加入亚磷酸二甲酯(123mg,1.1mmol)、六水合氯化铝2.7mg(0.1eq),加入2mL DCM溶解,密封,室温条件下反应6h。反应停止时,加入饱和NaCl淬灭,乙酸乙酯萃取三次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩溶剂,用乙酸乙酯∶正己烷=1∶2体系经硅胶柱层析分离纯化,得白色固体,产率55%。
1H NMR(600MHz,DMSO-d6)ppm 0.61(3H,s)0.86(1H,d,J=12.84Hz)1.06(3H,s)1.10(3H,s)1.19(3H,s)1.22-1.27(1H,m)1.27-1.32(1H,m)1.40(1H,d,J=4.95Hz)1.43(1H,d,J=5.32Hz)1.49(1H,d,J=8.07Hz)1.51-1.55(1H,m)1.57(3H,s)1.59-1.60(1H,m)1.60-1.64(1H,m)1.76-1.85(1H,m)1.93-1.97(1H,m)1.99(1H,d,J=2.20Hz)2.00-2.02(1H,m)2.02-2.06(1H,m)2.13(3H,s)2.32(1H,d,J=15.59Hz)3.49(3H,d,J=8.80Hz)3.51(3H,d,J=8.99Hz)4.19(1H,dd,J=23.66,6.24Hz)5.64(1H, dd,J=6.33,3.21Hz)6.66(1H,s)7.97(1H,s)9.04(1H,br.s.)12.04(1H,s).
13C NMR(151MHz,DMSO-d6)ppm 12.60,14.00(1C,s)18.04,21.58(1C,d,J=6.60Hz)22.11,28.78,29.48,30.02(1C,d,J=13.20Hz)30.18,31.00,31.45,32.42,33.49,33.57,34.57,36.42,37.43,37.89,38.79,43.88,52.31(1C,d,J=6.60Hz)52.81(1C,d,J=6.60Hz)109.32,114.64(1C,d,J=12.10Hz)117.90(1C,d,J=7.70Hz)121.35(1C,d,J=4.40Hz)140.50(1C,d,J=6.60Hz)141.05(1C,d,J=3.30Hz)144.08(1C,d,J=3.30Hz)150.62(1C,d,J=12.10Hz)179.51.
根据上述制备方法,本发明还合成了以下化合物:
Figure PCTCN2017091716-appb-000089
实施例37.化合物XS0439,XS0442,XS0444-XS0449,XS0478-XS0480,XS0487和XS0490的制备
Figure PCTCN2017091716-appb-000090
制备方法以XS0439为例:在搅拌条件下,将化合物YXY101(100mg,0.22mmol)溶于二氯甲烷(4mL)。加入7-甲氧基取代吲哚(65.3mg,0.44mmol),再加入六水合三氯化铝(5.3mg,0.022mmol),并在室温下搅拌反应5小时。停止反应,向反应体系中加入去离子水(15mL),并用乙酸乙酯萃取3次。合并乙酸乙酯层,然后用饱和NaCl洗3次,用无水Na2SO4干燥,用旋转蒸发仪浓缩得粗产物(棕色油状物)。粗产物经快速柱层析法(乙酸乙酯∶正己烷)分离纯化,真空干燥得呈紫红色固体的化合物。
1H-NMR(DMSO-d6)δppm 0.71(s,3H),0.86(d,J=9.9Hz,1H),0.96(s,3H),1.01(s,3H),1.10(s,3H),1.22-1.29(m,2H),1.32(s,3H),1.34-1.40(m,2H),1.45(d,J=8.1Hz,1H),1.50-1.61(m,3H),1.62-1.75(m,2H),1.79(s,3H),1.96-2.08(m,3H),2.34(d,J=15.4Hz,1H),3.88(s,3H),4.79(d,J=5.9Hz,1H),6.12(d,J=6.2Hz,1H),6.18(s,1H),6.62(d,J=7.7Hz,1H),6.73(s,1H),6.90(t,J=7.9Hz,1H),7.22(d,J=7.9Hz,1H),7.88(br.s.,1H),8.99(br.s.,1H),10.69(s,1H),12.04(br.s.,1H).
13C-NMR(DMSO-d6)δppm 11.9,18.5,22.3,29.1,29.9,30.4,30.5,30.6,31.8,32.9,34.9,35.4,35.5,35.6,36.8,36.9,37.8,43.5,44.3,55.4,101.8,108.8,112.2,119.2,119.8,121.1,121.6,122.6,126.3,126.7,128.5,140.8,141.4,144.0,146.6,147.0,180.0.
实施例38.化合物XS0486,XS0491和XS0492的制备
Figure PCTCN2017091716-appb-000091
以化合物XS0486为例:首先将化合物YXY101(50mg,0.11mmol)溶于二氧六环(600μL)中,加入三乙胺(150μL,0.33mmol),再加入二氧六环(100μL)洗涤反应瓶壁上残留,冷却至0℃,然后将亚磷酸二甲酯(110mg,1.1mmol)溶解于50μL四氯化碳中,在将溶解后的亚磷酸二甲酯慢慢滴入到溶有雷公藤红素的反应液中,0℃搅拌反应12h,加入10mL冰冷的去离子水,10mL饱和氯化铵溶液猝灭反应,用乙酸乙酯萃取3次(每次15mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯,得到红色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=1∶1)为洗脱剂,300-400目硅胶填充柱层析快速分离,得到红色固体产物。
1H NMR(600MHz,DMSO-d6)δppm 0.60(s,3H)0.96(d,J=13.57Hz,1H)1.08(s,3H)1.23(s,3H)1.24(s,3H)1.38(s,3H)1.41-1.48(m,2H)1.55-1.60(m,3H)1.61-1.67(m,2H)1.69-1.74(m,1H)1.78(dd,J=16.14,7.89Hz,1H)1.81-1.88(m,1H)1.95(td, J=14.12,3.85Hz,1H)2.02(d,J=13.94Hz,1H)2.09(s,3H)2.21-2.24(m,1H)2.27(d,J=15.96Hz,1H)3.74(dd,J=11.55,5.14Hz,6H)6.36(d,J=7.15Hz,1H)6.39(d,J=1.28Hz,1H)7.08(dd,J=6.97,1.10Hz,1H)8.75(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.07,18.53,21.43,28.03,29.09,29.24,29.71,30.07,31.21,31.46,32.76,33.92,35.85,37.71,38.78,41.23(d,J=5.50Hz,)41.97,43.37,44.45,55.15(dd,J=17.61,5.50Hz,2C)117.24,118.13,120.12,126.89,133.12,146.44,162.82,167.43,172.21(d,J=11.00Hz)177.98.
实施例39.化合物XS0488的制备
Figure PCTCN2017091716-appb-000092
首先将化合物XS0077(80mg,0.17mmol)溶于四氢呋喃(4mL)中,加入三乙胺(370μL,2.5mmol),再加入4-DMAP(24.7mg,0.22mmol),搅拌均匀,加入正丁酰氯(113μL,1.1mmol),室温搅拌反应30min,加入10mL饱和氯化铵溶液猝灭反应,用乙酸乙酯萃取3次(每次15mL),收集有机层,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯,得到红色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=4∶1)为洗脱剂,300-400目硅胶填充柱层析快速分离,得到明黄色固体产物。
1H NMR(600MHz,DMSO-d6)ppm 0.46-0.51(m,3H)0.91(d,J=1.00Hz,1H)0.95-1.01(m,3H)1.08(s,3H)1.12(s,3H)1.23(s,3H)1.36(ddd,J=13.80,4.20Hz,1H)1.42(s,3H)1.44-1.48(m,1H)1.54-1.60(m,3H)1.63(s,2H)1.66-1.68(m,2H)1.70(d,J=9.17Hz,2H)1.83(dd,J=13.57,7.70Hz,1H)1.93-1.99(m,1H)2.06(d,J=13.94Hz,1H)2.10(s,3H)2.22(d,J=7.34Hz,1H)2.32(d,J=15.59Hz,1H)2.55(t,J=7.15Hz,2H)3.49(s,3H)6.38(s,1H)6.41(d,J=7.15Hz,1H)7.31(d,J=6.97Hz,1H).
13C NMR(151MHz,DMSO-d6)ppm 13.41,18.05,21.59,28.06,29.11,29.42,30.11, 30.16,30.32,31.32,32.21,32.31,33.06,34.36,34.91,36.00,37.90,38.77,39.83,42.22,43.64,44.85,51.48,117.99,122.13,125.21,133.52,136.69,142.26,162.81,170.65,171.21,176.21,177.91.
实施例40.化合物XS0506的制备
Figure PCTCN2017091716-appb-000093
称取化合物YXY101(50mg,0.11mmol)于50ml圆底瓶中,加入二环己基碳二亚胺(23mg,0.11mmol)、葡萄糖(24mg,0.11mmol),加入2ml DCM溶解,室温反应12h。加入大量水淬灭,乙酸乙酯萃取三次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩溶剂,用乙酸乙酯∶正己烷=1∶4体系经硅胶柱层析分离纯化,得橙红色固体,产率42%。
1H NMR(600MHz,DMSO-d6)δppm 0.57(s,3H)1.07(s,3H)1.12-1.14(m,1H)1.14-1.16(m,1H)1.17(s,3H)1.21(s,3H)1.22-1.23(m,2H)1.24-1.28(m,4H)1.28-1.32(m,2H)1.38(s,3H)1.40-1.46(m,2H)1.47-1.52(m,2H)1.57(d,J=6.79Hz,1H)1.59-1.63(m,4H)1.64-1.72(m,6H)1.75-1.83(m,4H)1.92-1.96(m,1H)1.97-2.01(m,1H)2.03-2.07(m,1H)2.09(s,3H)2.18(d,J=10.82Hz,2H)2.73(d,J=15.04Hz,1H)3.40-3.49(m,1H)3.80-3.89(m,1H)6.36(d,J=7.34Hz,1H)6.40(d,J=0.92Hz,1H)7.07(dd,J=6.97,0.92Hz,1H)7.73(d,J=8.07Hz,1H)8.71(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.08,18.32,21.83,24.68,24.75,25.13,25.29,25.63,25.67,28.38,28.59,29.91,30.13,31.02,31.08,31.39,31.82,31.91,31.93,32.36,33.10,36.02,36.13,37.67,38.99,42.16,42.81,44.45,44.49,50.05,54.77,117.23,117.93, 120.04,126.75,133.25,146.43,153.97,163.09,168.74,175.44,177.83.
实施例41.化合物XS0507的制备
Figure PCTCN2017091716-appb-000094
称取化合物YXY101(50mg,0.11mmol)于50ml圆底瓶中,加入2ml DCM溶解,转移至-78℃搅拌,加入DAST(150ul,10eq),-78℃反应1h。将反应液直接倒入大量冰中停止反应,加入DCM萃取水相三次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩溶剂,用乙酸乙酯∶正己烷=1∶4体系经硅胶柱层析分离纯化,得橙红色固体,产率58%。
1H NMR(600MHz,DMSO-d6)δppm 0.56(s,3H)0.98(d,J=11.92Hz,1H)1.08(s,3H)1.22(s,3H)1.29(s,3H)1.38(s,3H)1.43-1.47(m,1H)1.48-1.52(m,1H)1.55-1.59(m,2H)1.60-1.64(m,1H)1.67(d,J=4.03Hz,1H)1.71(d,J=3.85Hz,1H)1.80(dd,J=16.41,8.16Hz,1H)1.83-1.87(m,1H)1.88-1.92(m,1H)1.96(d,J=17.61Hz,1H)1.95-1.95(m,1H)2.09(s,3H)2.20(d,J=1.83Hz,2H)6.35(d,J=7.34Hz,1H)6.38(d,J=1.28Hz,1H)7.06(dd,J=6.97,1.28Hz,1H)8.73(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.07,18.85,21.41,27.89,29.26,29.33,29.46,29.94,31.16,32.71,33.79,35.78,37.87,38.74,40.04,40.22,41.89,43.26,44.33,117.21,118.14,120.15,126.99,132.88,146.41,162.78,167.39(d,J=418.15Hz,1C),166.71,177.97.
实施例42.化合物XS0509的制备
Figure PCTCN2017091716-appb-000095
称取化合物YXY101(50mg,0.11mmol)于25ml厚壁耐压管中,加入四丁基溴化铵(Tetrabutylammonium bromide,TBAB,17.5mg,0.05mmol),加入2ml二氯甲烷(dichloromethane,DCM)溶解,再逐滴加入5%NaOH(180μl),室温条件下反应30min,再转移至50℃油浴下,逐滴加入2,3,4,6-四乙酰氧基-α-D吡喃葡萄糖溴化物-二氯甲烷溶液(57mg-1ml,0.138mmol),50℃反应12h,停止反应,加入大量水和饱和食盐水,加入DCM萃取3次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩,用乙酸乙酯∶正己烷=4∶1体系经硅胶层析柱进行纯化,得黄色固体,产率25%。
1H NMR(600MHz,DMSO-d6)δppm 0.51(s,3H)0.95(d,J=13.57Hz,1H)1.08(d,J=6.24Hz,6H)1.22(s,3H)1.23(br.s.,1H)1.38(s,3H)1.41(d,J=4.22Hz,1H)1.46(d,J=11.92Hz,1H)1.56(d,J=7.52Hz,2H)1.58(s,3H)1.60-1.63(m,1H)1.64(d,J=4.95Hz,2H)1.66-1.69(m,1H)1.81-1.87(m,1H)1.91(s,3H)1.94(d,J=3.67Hz,1H)1.97(s,3H)2.00-2.04(m,1H)2.08(s,3H)2.12(s,3H)2.19(d,J=8.80Hz,1H)2.31(d,J=15.59Hz,1H)3.85-3.90(m,1H)3.92-3.96(m,1H)4.30(dd,J=8.16,4.13Hz,1H)5.05(dd,J=10.09,8.44Hz,1H)5.23(d,J=3.48Hz,1H)5.29(dd,J=10.45,3.67Hz,1H)5.83(d,J=8.25Hz,1H)6.36(d,J=7.15Hz,1H)6.37(d,J=0.92Hz,1H)7.06(d,J=6.97Hz,1H)8.71(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.06,18.63,19.97,20.31,20.42,21.57,28.11,28.59,29.22,29.97,30.07,31.31,32.62,32.84,34.57,35.93,37.76,38.52,40.05,40.24,41.97,43.46,44.54,61.76,67.35,67.85,69.78,71.21,91.64,117.29,118.08,120.11,126.93,133.07,146.36,162.86,167.78,169.24,169.41,169.57,169.97,176.51,177.93.
实施例43.化合物XS0514,XS0515的制备
Figure PCTCN2017091716-appb-000096
以合成XS0514为例:称取XS0419(50mg,0.11mmol)于50ml圆底瓶中,加入2ml DCM溶解,转移至-78℃搅拌,加入二乙胺基三氟化硫(Diethylaminosulfurtrifluoride,DAST,150ul,10eq),-78℃反应1h。将反应液直接倒入大量冰中停止反应,加入DCM萃取水相三次,合并有机相,用饱和NaHCO3洗涤有机相,无水Na2SO4干燥有机相,将有机相减压浓缩,用乙酸乙酯∶正己烷=1∶8体系经硅胶柱层析进行分离纯化,得橙红色固体,产率58%。
1H NMR(600MHz,DMSO-d6)δppm 0.61(s,3H)0.95(d,J=14.49Hz,1H)1.06(s,3H)1.18(s,3H)1.23(s,3H)1.28(s,3H)1.41(d,J=14.86Hz,2H)1.47(dd,J=13.85,4.31Hz,2H)1.54(d,J=7.70Hz,1H)1.58-1.63(m,2H)1.74-1.84(m,2H)1.88(d,J=6.05Hz,1H)1.94(d,J=10.64Hz,1H)1.98(d,J=10.09Hz,1H)2.01(s,3H)2.07(d,J=17.97Hz,1H)2.24(d,J=15.96Hz,1H)2.89-2.94(m,1H)3.16-3.21(m,1H)5.74(d,J=4.77Hz,1H)6.61(s,1H)7.81(br.s.,1H)8.78(br.s.,1H).
13C NMR(151MHz,DMSO-d6)δppm 11.56,18.66,22.57,27.30,28.27,29.21,29.54,29.98,30.02,30.08,31.18,33.69,33.99,34.13,36.05,36.27,37.14,40.25,43.12,43.42,108.21,118.07,120.13,123.02,139.23,140.61,143.17,148.76,167.64(d,J=380.73Hz,1C).
实施例44.化合物XS0516的制备
Figure PCTCN2017091716-appb-000097
称取XS0077(50mg,0.11mmol)于25ml厚壁耐压管中,加入四丁基溴化铵(Tetrabutylammonium bromide,TBAB,17.5mg,0.05mmol),加入2ml二氯甲烷(dichloromethane,DCM)溶解,再逐滴加入5%NaOH(180μL),室温条件下反应30min,再转移至50℃油浴下,逐滴加入2,3,4,6-四乙酰氧基-α-D吡喃葡萄糖溴化物-二氯甲烷溶液(57mg-1ml,0.138mmol),50℃反应12h,停止反应,加入大量水和饱和食盐水,加入DCM萃取3次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩,用乙酸乙酯∶正己烷=4∶1体系经硅胶层析柱进行纯化,得黄色固体,产率25%。
1H NMR(600MHz,CHLOROFORM-d)δppm 0.53(s,3H)0.97(d,J=14.31Hz,1H)1.10(s,3H)1.18(s,3H)1.26(s,3H)1.38(td,J=14.03,4.58Hz,1H)1.47(s,3H)1.50(dd,J=15.59,4.58Hz,1H)1.53-1.56(m,1H)1.58(d,J=8.25Hz,1H)1.62-1.65(m,2H)1.66(d,J=5.32Hz,1H)1.67-1.72(m,2H)1.76-1.80(m,1H)1.82-1.87(m,1H)1.87-1.91(m,1H)1.98(s,3H)2.01(s,3H)2.10-2.14(m,1H)2.15(s,3H)2.18(s,3H)2.23(s,3H)2.42(d,J=15.77Hz,1H)3.55(s,3H)3.86(t,J=7.34Hz,1H)4.06(dd,J=11.10,7.61Hz,1H)4.16(dd,J=11.10,6.14Hz,1H)5.11(dd,J=10.36,3.58Hz,1H)5.31(d,J=7.89Hz,1H)5.38-5.43(m,2H)6.31(d,J=7.15Hz,1H)6.39(d,J=1.10Hz,1H)7.02(dd,J=7.06,1.01Hz,1H).
13C NMR(151MHz,CHLOROFORM-d)δppm 11.54,18.33,20.61,20.70,21.02,21.86,28.57,29.57,29.85,30.52,30.82,31.55,32.65,33.67,34.70,36.34,38.20,39.24,40.37,42.30,44.27,45.07,51.54,60.81,66.90,69.09,70.49,70.84,100.37,117.93,123.33,126.92,134.32,134.96,145.71,162.50,170.07,170.33,170.35,170.37,170.61,178.68,179.14.
实施例45.化合物XS0534的制备
Figure PCTCN2017091716-appb-000098
称取XS0077(150mg,0.32mmol)溶于4mL丙酮中,搅拌溶解,加入无水碳酸钾(180mg,1.3mmol),搅拌加入硫酸二甲酯(95μL,0.96mmol),油浴70℃反应12h;用1mol/L的HCl猝灭反应,并调节PH=7,乙酸乙酯萃取三次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩溶剂,用乙酸乙酯∶正己烷=1∶10体系经硅胶柱层析分离纯化,得白色固体,产率24.3%。
1H NMR(600MHz,DMSO-d6)d ppm 0.51(s,3H)0.87-0.92(m,1H)1.06(s,3H)1.12(s,3H)1.18(s,3H)1.27(s,3H)1.32-1.37(m,1H)1.37-1.42(m,1H)1.47-1.52(m,3H)1.55-1.61(m,2H)1.65(dd,J=15.68,8.16Hz,1H)1.80(td,J=13.43,7.24Hz,1H)1.84-1.90(m,1H)1.97(td,J=13.75,4.03Hz,1H)2.02(s,3H)2.06(d,J=13.57Hz,1H)2.12-2.18(m,1H)2.33(d,J=15.77Hz,1H)2.95(dd,J=20.17,1.40Hz,1H)3.21(dd,J=20.72,6.24Hz,1H)3.46-3.50(m,3H)3.77(s,3H)5.72(dd,J=6.42,1.28Hz,1H)6.72(s,1H)8.10(s,1H).
13C NMR(151MHz,DMSO-d6)d ppm 11.35,17.69,22.41,27.31,28.41,29.42,29.90,30.13,30.25,31.36,32.37,34.01,34.21,34.36,36.41,36.49,37.11,39.85,43.23,43.80,51.37,55.82,105.29,117.48,119.79,124.58,139.03,141.52,145.83,148.73,178.04.
实施例46.化合物XS0455,XS0504的制备
Figure PCTCN2017091716-appb-000099
以XS0455为例:首先将化合物YXY101(40mg,0.09mmol)搅拌溶解于1.2mL THF中,随后加入钐粉(70mg,0.36mmol),对甲基苯磺酸(67mg,0.36mmol),室温搅拌反应4小时。砂芯漏斗过滤催化剂钐粉以及不溶物,用乙酸乙酯洗涤3次(每次10mL),收集滤液,无水硫酸钠干燥后,用减压蒸馏法除去有机溶剂乙酸乙酯和四氢呋喃,得到黄褐色固体混合粗品。以正己烷和乙酸乙酯(hexane/ethyl acetate=2∶1)为洗脱剂,300-400目硅胶填充柱层析分离,得到黄褐色固体产物。
1H NMR(600MHz,DMSO-d6)ppm 0.82(1H,d,J=13.39Hz)0.89(3H,s)0.94(3H,br.s.)0.94(3H,br.s.)1.02(3H,s)1.13(3H,s)1.22(1H,d,J=15.96Hz)1.29-1.34(1H,m)1.36(1H,dd,J=13.39,3.12Hz)1.63-1.76(4H,m)1.92(1H,ddd,J=13.60,4.00Hz)1.96-2.00(1H,m)2.02(1H,d,J=6.97Hz)2.06(3H,s)2.12(2H,dd,J=13.66,7.06Hz)5.41(1H,d,J=5.87Hz)6.21(1H,d,J=9.90Hz)6.31(1H,d,J=9.90Hz)6.56(1H,s)8.03(1H,s)9.20(1H,s)12.03(1H,br.s).
13C NMR(151MHz,DMSO-d6)ppm 11.18,18.81,19.25,22.49,23.39,29.25,30.51,30.75,31.24,32.68,32.82,36.79,37.26,38.37,40.04,40.24,43.38,46.31,108.52,119.50,119.71,121.02,122.84,128.56,137.37,142.00,142.94,144.16,179.82.
实施例47.化合物XS0420的制备
Figure PCTCN2017091716-appb-000100
称取化合物YXY101(50mg,0.11mmol)于50ml圆底瓶中,加入PTSA(100mg,过量),加入甲苯2mL,室温搅拌6h。加入饱和NaHCO3溶液淬灭,加入乙酸乙酯萃取3次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩溶剂,用乙酸乙酯∶正己烷=1∶4体系经硅胶柱层析分离纯化,得灰白色固体,产率70%。
1H NMR(600MHz,DMSO-d6)δppm 1.03(s,3H)1.06(s,3H)1.07-1.11(m,1H)1.15(s,3H)1.18(s,3H)1.21-1.24(m,1H)1.28-1.32(m,1H)1.37(td,J=13.75,5.32Hz,1H)1.46(dd,J=8.62,4.03Hz,2H)1.48-1.50(m,2H)1.51-1.55(m,1H)1.58(dd,J=12.93,1.93Hz,1H)1.83-1.90(m,3H)2.33(t,J=1.00Hz,1H)2.36(s,3H)2.38(s,3H)2.80-2.92(m,2H)2.80-2.92(m,2H)7.06(d,J=8.62Hz,1H)7.18(s,1H)7.49(d,J=8.44Hz,1H)8.48(br.s.,1H)9.92(s,1H).
13C NMR(151MHz,DMSO-d6)δppm 10.96,19.45,21.73,21.95,23.51,24.03,25.45,30.89,30.96,34.59,35.08,36.41,37.98,38.30,40.71,41.70,43.50,99.01,102.91,115.74,120.73,126.07,127.17,127.30,129.03,132.36,143.60,146.12,175.68.
实施例48.化合物XS0502的制备
Figure PCTCN2017091716-appb-000101
称取化合物XS0077(50mg,0.11mmol)于50ml圆底瓶中,加入SeO2(200mg,过量),加入二氧六环2mL,55℃搅拌12h。加入去离子水淬灭反应,加入乙酸乙酯萃取3次,合并有机相,无水Na2SO4干燥,将有机相减压浓缩溶剂,用乙酸乙酯∶正己烷=1∶10体系经硅胶柱层析分离纯化,得灰白色固体,产率16.8%。
1H NMR(600MHz,DMSO-d6)d ppm 0.75(s,3H)0.93-0.97(m,1H)1.10(s,3H)1.21(s,3H)1.22(s,3H)1.40(td,J=14.08,4.13Hz,1H)1.49(dt,J=14.53,4.65Hz,1H)1.71-1.79(m,2H)1.79-1.82(m,1H)1.82-1.88(m,1H)2.07-2.10(m,1H)2.10-2.12(m,1H)2.13(s,3H)2.29(d,J=14.31Hz,1H)2.46(t,J=4.77Hz,1H)2.54(s,3H)3.50(s,3H)6.20(d,J=9.54Hz,1H)6.45(d,J=9.54Hz,1H)7.11(s,1H)9.44(s,1H).
13C NMR(151MHz,DMSO-d6)d ppm 11.88,17.45,19.70,21.88,26.78,29.04,30.34,30.42,30.47,31.02,34.19,36.47,40.05,40.43,41.84,46.15,51.58,122.97,125.72,125.78,129.24,136.20,139.22,142.11,142.43,144.61,146.30,176.92,178.25,181.38.
尽管本发明的具体实施方式已经得到详细的描述,根据已经公开的所有教导,本领域技术人员可以对本发明技术方案的细节进行各种修改和替换,这些改变均在本发明的保护范围之内。本发明的全部范围由所附权利要求及其任何等同物给出。

Claims (73)

  1. 如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
    Figure PCTCN2017091716-appb-100001
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取 代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100002
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  2. 权利要求1的用途,其中,Y和与之相连的碳原子之间为双键,且Y代表O。
  3. 权利要求1或2的用途,其中,所述化合物中的X代表-NH-、-N(R)-、-O-、-CH2-或卤素;R代表C1-6烷基或3-8元环烷基(优选环己基);其中,当X为卤素时,R1不存在;
    优选地,X代表-NH-、-N(R)-、-O-或氟;R代表环己基。
  4. 权利要求1-3任一项的用途,其中,所述化合物中的R1不存在,或代表氢、C1-4烷基、-PO(OR)2、单糖基、C1-4烷氧羰基-C1-4烷基、3-6元环烷基-氨酰基、芳基-C1-4烷基或芳基;其中,所述C1-4烷基、单糖基、C1-4烷氧羰基-C1-4烷基、3-6元环烷基-氨酰基、芳基-C1-4烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自 下述的取代基取代:卤素、羟基、氨基、C1-4烷基、C1-4烷氧基、C1-4烷氨基和C1-4烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R代表C1-4烷基;
    优选地,R1不存在,或代表氢、C1-4烷基、-PO(OR)2、葡萄糖基、C1-2烷氧羰基-C1-2烷基、环己基-氨酰基、苯基-C1-2烷基、萘基-C1-2烷基、苯基或萘基;其中,所述甲基、乙基、葡萄糖基、C1-2烷氧羰基-C1-2烷基、环己基-氨酰基、苯基-C1-2烷基、萘基-C1-2烷基、苯基或萘基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:C1-2烷基、C1-2烷氧基和C1-2烷酰基;
    R代表C1-4烷基;
    优选地,R1代表氢、C1-4烷基、-PO(OR)2或C1-2烷氧羰基-C1-2烷基;
    R代表C1-3烷基;
    优选地,R1不存在,或代表氢、甲基、乙基、-PO(OMe)2、-PO(OEt)2、-PO(OiPr)2、2,3,4,6-四乙酰氧基-α-D-吡喃葡萄糖基、EtOCOCH2-、环己基-氨酰基、苄基、甲氧基苯基或叔丁基苯基。
  5. 权利要求1-4任一项的用途,其中,所述化合物中的R2代表H、D、-OH、-PO(OR)2、C1-6烷基、9-15元稠杂芳基或磺酸盐;其中所述C1-6烷基或6-15元杂芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷酰基、氰基、三氟甲基和羧基;
    R代表H、C1-6烷基或芳基;
    优选地,R2代表H、D、-PO(OR)2、C1-4烷基、9-15元苯并稠杂芳基或磺酸盐;其中,所述C1-4烷基或9-15元苯并稠杂芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-4烷基、C1-4烷氧基、C1-4烷酰基、氰基、三氟甲基和羧基;
    R代表H、C1-4烷基或苯基;
    优选地,R2代表H、D、-PO(OR)2、C1-4烷基、9-15元苯并稠杂芳基或磺酸盐;其中,所述C1-4烷基和9-15元苯并稠杂芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-4烷基、C1-4烷氧基、C1-4烷 酰基、氰基、三氟甲基和羧基;
    R代表H、C1-4烷基或苯基;
    优选地,R2代表H、D、-PO(OR)2、2-氧代苯基、吲哚基或磺酸钠;其中,所述吲哚基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、氟、氯、溴、羟基、甲基、甲氧基、甲酰基、氰基、三氟甲基和羧基;
    R代表H、甲基、乙基、异丙基或苯基。
  6. 权利要求1-5任一项的用途,其中,所述化合物的7位和8位碳原子之间为碳碳双键。
  7. 权利要求1-6任一项的用途,其中,所述化合物Y和与之相连的碳原子之间为碳碳双键。
  8. 权利要求1-6任一项的用途,其中,所述化合物Y和与之相连的碳原子之间为碳碳单键。
  9. 权利要求1-8任一项的用途,其中,所述化合物具有如下结构:
    Figure PCTCN2017091716-appb-100003
    其中,R3和R4各自独立地代表H、C1-6烷基或C1-6烷酰基;
    优选地,R3和R4各自独立地代表H、C1-4烷基或C1-4烷酰基;
    优选地,R3和R4各自独立地代表H、甲基或丁酰基。
  10. 权利要求1-8任一项的用途,其中,所述化合物具有如下结构:
    Figure PCTCN2017091716-appb-100004
    其中,R4代表H、C1-6烷酰基、C1-6烷氧羰基或被一个或多个(例如1、2、3或4个)C1-6烷酰基取代的单糖基;
    优选地,R4代表H、C1-4烷酰基、C1-4烷氧羰基或被一个或多个(例如1、2、3或4个)C1-4烷酰基取代的葡萄糖糖基;
    优选地,R4代表H或C1-2烷氧羰基;
    优选地,R4代表H、丁酰基、乙氧羰基或2,3,4,6-四乙酰氧基-α-D-吡喃葡萄糖糖基。
  11. 权利要求1的用途,其中,所述化合物选自下列化合物:
    Figure PCTCN2017091716-appb-100005
    Figure PCTCN2017091716-appb-100006
    Figure PCTCN2017091716-appb-100007
    Figure PCTCN2017091716-appb-100008
  12. 权利要求1-11任一项的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  13. 权利要求1-11任一项的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应;
    优选地,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  14. 权利要求1-11任一项的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  15. 权利要求1-11任一项的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  16. 权利要求1-11任一项的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于调节线粒体在细胞中的活性;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  17. 权利要求1-11任一项的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病;
    优选地,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、 动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
  18. 一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
    Figure PCTCN2017091716-appb-100009
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、 氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100010
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  19. 权利要求18的方法,其中,式I所示的化合物如权利要求2-11任一项中所定义;
    优选地,所述方法用于抑制Nur77在细胞中的转录活性;
    优选地,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  20. 一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的如式I所示的化合物、 其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100011
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100012
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  21. 权利要求20的方法,其中,式I所示的化合物如权利要求2-11任一项中所定义;
    优选地,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活;
    优选地,所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  22. 一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100013
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、 氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100014
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  23. 权利要求22的方法,其中,式I所示的化合物如权利要求2-11任一项中所定义;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  24. 一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与如式I所示的化合物、其互变异构体、立体异构体或药学上可接 受的盐或酯相接触的步骤:
    Figure PCTCN2017091716-appb-100015
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100016
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  25. 权利要求24的方法,其中,式I所示的化合物如权利要求2-11任一项中所定义;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  26. 一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
    Figure PCTCN2017091716-appb-100017
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰 基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100018
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  27. 权利要求26的方法,其中,式I化合物如权利要求2-11中所定义;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  28. 一种预防或治疗与Nur77相关的疾病的方法,其包括,给有此需要的受试者施用有效量的如式I所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100019
    其中,X代表-NH-、-N(R)-、-O-、-CH2-或卤素;其中,当X为卤素时,R1不存 在;
    当Y和与之相连的碳原子之间为单键时,Y代表H、卤素、-OR、-SR或-NRR’;当Y和与之相连的碳原子之间为双键时,Y代表O、S或NR;
    R1不存在,或代表H、-PO(OR)2、C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、糖基、C1-6烷氧羰基-C1-6烷基、3-8元环烷基-氨酰基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R2代表H、D、-PO(OR)2、-CONH2、-NH2、-NHR、-NRR’、-NHCOR、-NRCOR、-NHCOOR、-NHCONHR、-NHCONRR’、-NRCONHR、-NRCONRR’、-OH、-OR、-OCONHR、-OCONRR’、-SH、-SR、-SOR、-SOOR、-SO2NHR”、硝基、卤素、糖基、氰基、三氟甲基、C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基、炔基、亚磺基、磺酸或磺酸盐;其中所述C1-6烷基、3-8元环烷基、3-8元杂环烷基、芳基、C1-6烷基取代的芳基、6-15元杂芳基、链烯基和炔基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:氨基、卤素、羟基、氧基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷酰基、3-8元环烷基(例如环丙基)、氧代3-8元环烷基(例如氧代环丁基)、氰基、三氟甲基、C1-6烷氧羰基、C1-6烷基酰胺基、脲基、氨基甲酸酯基、羧基以及芳基;
    R3和R4各自独立地表示不存在,或代表H、C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、C1-6烷酰基、C1-6烷氧羰基、糖基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基、C1-6烷氨基和C1-6烷酰基;优选地,所述芳基为6-14元芳基,例如6-10元芳基;更优选地为苯基或萘基;
    R和R’各自独立地选自H、C1-6烷基、3-8元环烷基、芳基-C1-6烷基或芳基,其中所述C1-6烷基、3-8元环烷基、芳基-C1-6烷基和芳基未被取代或被一个或多个(例如1、2、3或4个)选自下述的取代基取代:卤素、羟基、氨基、C1-6烷基、C1-6烷氧基和 C1-6烷氨基;
    R”代表C1-6烷基或芳基(例如6-10元芳基,优选苯基);
    式(I)中的虚实双键
    Figure PCTCN2017091716-appb-100020
    代表单键或者双键;优选地,环A包含0、1、2或3个碳碳双键;环B包含0、1或2个碳碳双键。
  29. 权利要求28的方法,其中,式I所示的化合物如权利要求2-11任一项中所定义;
    优选地,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
  30. 如式IV所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性;
    Figure PCTCN2017091716-appb-100021
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100022
    代表单键或者双键。
  31. 权利要求30的用途,其中所述化合物选自下列化合物:
    Figure PCTCN2017091716-appb-100023
  32. 权利要求30或31的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  33. 权利要求30或31的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应;
    优选地,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  34. 权利要求30或31的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  35. 权利要求30或31的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  36. 权利要求30或31的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于调节线粒体在细胞中的活性;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  37. 权利要求30或31的用途,其中,所述化合物、其互变异构体、立体异构体 或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病;
    优选地,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
  38. 一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
    Figure PCTCN2017091716-appb-100024
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100025
    代表单键或者双键。
  39. 权利要求38的方法,其中,式I所示的化合物如权利要求31中所定义;
    优选地,所述方法用于抑制Nur77在细胞中的转录活性;
    优选地,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细 胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  40. 一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100026
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100027
    代表单键或者双键。
  41. 权利要求40的方法,其中,式I所示的化合物如权利要求31中所定义;
    优选地,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活;
    优选地,所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)诱导的IκBα降解;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细 胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  42. 一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100028
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100029
    代表单键或者双键。
  43. 权利要求42的方法,其中,式I所示的化合物如权利要求31中所定义;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  44. 一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
    Figure PCTCN2017091716-appb-100030
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100031
    代表单键或者双键。
  45. 权利要求44的方法,其中,式I所示的化合物如权利要求31中所定义;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  46. 一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药 物:
    Figure PCTCN2017091716-appb-100032
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100033
    代表单键或者双键。
  47. 权利要求46的方法,其中所述化合物如权利要求31中所定义;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  48. 一种预防或治疗与Nur77相关疾病的方法,其包括,向有此需要的受试者施用有效量的如式IV所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的步骤:
    Figure PCTCN2017091716-appb-100034
    其中,X代表NH、O或CH2
    Y代表O、S或NR;
    R5和R6各自独立地代表代表H或C1-6烷基;
    当R7和与之相连的碳原子之间为单键时,R7代表OH,当R7和与之相连的碳原子之间为双键时,R7代表O;
    R8代表H或C1-6烷基;
    式(IV)中的虚实双键
    Figure PCTCN2017091716-appb-100035
    代表单键或者双键。
  49. 权利要求48的方法,其中所述化合物如权利要求31中所定义;
    优选地,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
  50. 下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的用途,其用作孤儿核受体Nur77的配体,或者用于制备用作孤儿核受体Nur77的配体的药物:
    Figure PCTCN2017091716-appb-100036
  51. 权利要求50的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制Nur77的转录活性;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于抑制Nur77在细胞中的转录活性;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  52. 权利要求51的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应;
    优选地,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  53. 权利要求51的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  54. 权利要求51的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于诱导Nur77、 TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  55. 权利要求51的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于调节线粒体的活性;
    优选地,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于调节线粒体在细胞中的活性;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  56. 权利要求51的用途,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用作孤儿核受体Nur77的配体,用于在有此需要的受试者中预防或治疗与Nur77相关的疾病;
    优选地,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
  57. 一种抑制孤儿核受体Nur77的转录活性的方法,其包括,将Nur77与下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
    Figure PCTCN2017091716-appb-100037
  58. 权利要求57的方法,其中,所述方法用于抑制Nur77在细胞中的转录活性;
    优选地,所述方法包括,给有此需要的细胞施用有效量的所述化合物,从而抑制Nur77在细胞中的转录活性;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  59. 一种抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)在细胞中的生物学效应的方法,其包括,给有此需要的细胞施用有效量的下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100038
  60. 权利要求59的方法,其中,TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等)的生物学效应包括但不限于κB抑制蛋白(IκB)激酶α/β(IKKα/β)的磷酸化,IκBα的降解,NF-κB亚基p65的入核转运,和/或NF-κB的激活;
    优选地,所述方法用于抑制TNFα或其他炎症因子(例如IL-1β、IL-6或IL-8等) 诱导的IκBα降解;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  61. 一种诱导细胞中Nur77与TRAF2、p62、LC3和/或Ub的相互作用或者共定位的方法,其包括,给有此需要的细胞施用有效量的下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100039
  62. 权利要求61的方法,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导Nur77在细胞中与TRAF2、p62、LC3和/或Ub的相互作用或者共定位;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  63. 一种诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运的方法,其包括,将Nur77与下列所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯相接触的步骤:
    Figure PCTCN2017091716-appb-100040
  64. 权利要求63的方法,其中,所述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物用于诱导细胞中Nur77、TRAF2、p62、LC3和/或Ub的线粒体转运;
    优选地,所述细胞表达Nur77;
    优选地,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  65. 一种调节细胞中线粒体活性的方法,其包括,给有此需要的细胞施用有效量的下述化合物、其互变异构体、立体异构体或药学上可接受的盐或酯或者所述药物:
    Figure PCTCN2017091716-appb-100041
  66. 权利要求65的方法,其中,所述细胞为癌细胞(例如肝癌细胞,宫颈癌细胞,肺癌细胞、乳腺癌细胞、结直肠癌细胞或前列腺癌细胞)或炎症敏感细胞(例如脂肪细胞,炎症细胞,内皮细胞,上皮细胞,神经细胞,干细胞,淋巴细胞等)。
  67. 一种预防或治疗与Nur77相关疾病的方法,其包括,向有此需要的受试者施 用有效量的如下所示的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯的步骤:
    Figure PCTCN2017091716-appb-100042
  68. 权利要求67的方法,其中,所述与Nur77相关的疾病选自炎症(例如,与动脉粥样硬化相关的炎症,与肥胖症相关的炎症,与糖尿病相关的炎症,肝炎,肺炎,关节炎或炎症性肠病)、动脉粥样硬化、肥胖症、糖尿病、牛皮癣、多发性硬化和癌症(例如三阴乳腺癌)。
  69. 如下所示化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100043
    其中,
    R9和R10各自独立地选自氢、C1-6烷基和芳基;
    优选地,R9和R10各自独立地选自氢、C1-4烷基、苯基和萘基;
    优选地,R9和R10各自独立地选自甲基、乙基和异丙基。
  70. 权利要求69的化合物、其互变异构体、立体异构体或药学上可接受的盐或酯,其中所述化合物选自下列化合物:
    Figure PCTCN2017091716-appb-100044
  71. 下列化合物、其互变异构体、立体异构体或药学上可接受的盐或酯:
    Figure PCTCN2017091716-appb-100045
  72. 一种具有抗炎和/或治疗肥胖症活性的药物的筛选方法,其包括以下步骤:
    (1)提供表达孤儿核受体Nur77以及至少下述一种蛋白的细胞:TRAF2、p62、LC3和Ub;
    (2)用TNFα和候选试剂处理所述细胞,设立阴性对照组(即不经候选试剂处理);
    (3)检测并判断如下项:
    a.所述候选试剂是否可以与Nur77结合;
    b.是否可以诱导Nur77从细胞核转运至线粒体;
    c.与阴性对照组比,IκBα的表达水平是否升高;
    d.TRAF2的泛素化是否被抑制;
    e.Nur77的泛素化是否被诱导;
    f.是否发生TRAF2与Nur77在线粒体的相互作用;
    g.是否发生p62的线粒体定位;
    h.是否发生p62与Nur77在线粒体的相互作用;
    i.是否发生LC3的线粒体定位;
    j.是否发生LC3与Nur77在线粒体的相互作用;
    (4)如所述步骤(3)中a项和b项判断结果均为“是”,且,c至i任一项判断结果为“是”,则判断其具有抗炎和/或治疗肥胖症活性。
  73. 权利要求72的方法,其中,a-j项可通过免疫学方法进行检测;优选地,所述免疫学方法包括ELISA检测、Elispot检测、Western印迹、表面等离子共振法等。
PCT/CN2017/091716 2016-07-04 2017-07-04 孤儿核受体Nur77的配体及其用途 WO2018006801A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201780002455.9A CN108026142B (zh) 2016-07-04 2017-07-04 孤儿核受体Nur77的配体及其用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610520308.8 2016-07-04
CN201610520308 2016-07-04

Publications (1)

Publication Number Publication Date
WO2018006801A1 true WO2018006801A1 (zh) 2018-01-11

Family

ID=60912349

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2017/091716 WO2018006801A1 (zh) 2016-07-04 2017-07-04 孤儿核受体Nur77的配体及其用途
PCT/CN2017/091726 WO2018006804A1 (zh) 2016-07-04 2017-07-04 孤儿核受体Nur77的配体及其用途

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/091726 WO2018006804A1 (zh) 2016-07-04 2017-07-04 孤儿核受体Nur77的配体及其用途

Country Status (5)

Country Link
US (1) US10808005B2 (zh)
EP (1) EP3480207A4 (zh)
JP (1) JP2019523245A (zh)
CN (2) CN108026141B (zh)
WO (2) WO2018006801A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109678923A (zh) * 2019-01-04 2019-04-26 中国药科大学 雷公藤红素(异)阿魏酸酯类衍生物及其制备方法与用途
CN111620924A (zh) * 2020-06-04 2020-09-04 华中农业大学 基于天然产物的药物设计方法、五环三萜类化合物、其制备方法及应用
WO2021047672A1 (en) * 2019-09-12 2021-03-18 Ixmedicine Co., Ltd Triterpenoid compounds, pharmaceutical compositions thereof, and their use for treating nuclear receptor subfamily 4 group member 1-mediated disease
CN113234116A (zh) * 2021-01-26 2021-08-10 延边大学 一种雷公藤红素衍生物及其制备方法和医用用途
CN116023426A (zh) * 2022-12-30 2023-04-28 上海海洋大学 去甲泽拉木醛衍生物及其在制备抗癌药物中的应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040220267A1 (en) * 2003-02-07 2004-11-04 Devlin J. P. Derivatives of pentacyclic nortriterpene quinone methides as compounds useful in the treatment of inflammatory, neurodegenerative, and neoplastic diseases
WO2009067891A1 (fr) * 2007-11-16 2009-06-04 Shanghai Huatuo Medical Science Co., Ltd Composés de triterpènephénol solubles dans l'eau ayant une activité anti-tumorale et leur préparation
WO2015148802A1 (en) * 2014-03-26 2015-10-01 The Children's Medical Center Corporation Celastrol and derivatives for the treatment of obesity
CN105985401A (zh) * 2015-02-16 2016-10-05 上海华拓医药科技发展有限公司 一种雷公藤红素衍生物、其制备方法及用途
WO2017070615A1 (en) * 2015-10-23 2017-04-27 Erx Pharmaceuticals, Inc. Analogs of celastrol

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0952899A (ja) * 1994-09-30 1997-02-25 Tsumura & Co ロイコトリエン拮抗剤
ES2277568B1 (es) 2005-12-30 2008-04-01 Consejo Superior De Investigaciones Cientificas Derivados de triterpenoquinona y triterpenofenoles y su aplicacion para el tratamiento de tumores y enfermedades parasitarias.
HUE025852T2 (en) * 2008-06-26 2016-04-28 Orphazyme Aps Use of HSP70 to regulate enzyme activity
CN101624415A (zh) * 2008-07-10 2010-01-13 朱永亮 雷公藤红素及其衍生物用于癌症、炎症及中枢神经系统疾病的治疗
CN101805390B (zh) 2010-04-13 2012-07-18 暨南大学 一种雷公藤红素衍生物及其用途
CN102574890B (zh) * 2010-08-23 2015-03-11 苏州润新生物科技有限公司 某些化学个体、组合物及方法
CN103642887B (zh) * 2013-11-21 2016-06-15 华侨大学 一种雷公藤红素衍生物的制备方法及其产品和应用
CN104829679B (zh) * 2014-02-10 2017-12-22 上海华拓医药科技发展有限公司 一类水溶性南蛇藤素衍生物及其用途
WO2016007535A1 (en) * 2014-07-08 2016-01-14 The Board Of Trustees Of The Leland Stanford Junior University Antagonists of hsp90/cdc37 and methods of using the same

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040220267A1 (en) * 2003-02-07 2004-11-04 Devlin J. P. Derivatives of pentacyclic nortriterpene quinone methides as compounds useful in the treatment of inflammatory, neurodegenerative, and neoplastic diseases
WO2009067891A1 (fr) * 2007-11-16 2009-06-04 Shanghai Huatuo Medical Science Co., Ltd Composés de triterpènephénol solubles dans l'eau ayant une activité anti-tumorale et leur préparation
WO2015148802A1 (en) * 2014-03-26 2015-10-01 The Children's Medical Center Corporation Celastrol and derivatives for the treatment of obesity
CN105985401A (zh) * 2015-02-16 2016-10-05 上海华拓医药科技发展有限公司 一种雷公藤红素衍生物、其制备方法及用途
WO2017070615A1 (en) * 2015-10-23 2017-04-27 Erx Pharmaceuticals, Inc. Analogs of celastrol

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HARADA, R. ET AL.: "STRUCTURE OF PRISTIWERIN, A QUINONOID TRITERPENE", TETRAHEDRON LETTERS, 31 December 1962 (1962-12-31), pages 603 - 607 *
HU , MENGJIE ET AL.: "Celastrol-Induced Nur77 Interaction with TRAF2 Alleviates Inflammation by Promoting Mitochondrial Ubiquitination and Autophagy", MOLECULAR CELL, vol. 66, no. 1, 6 April 2017 (2017-04-06), pages 141 - 153, XP029970893, DOI: 10.1016/j.molcel.2017.03.008 *
KLAIC, L. ET AL.: "Remarkable Stereospecific Conjugate Additions to the Hsp90 Inhibitor Celastrol", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 133, 16 November 2011 (2011-11-16), pages 19634 - 19637, XP055192971, DOI: doi:10.1021/ja208359a *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109678923A (zh) * 2019-01-04 2019-04-26 中国药科大学 雷公藤红素(异)阿魏酸酯类衍生物及其制备方法与用途
CN109678923B (zh) * 2019-01-04 2021-04-27 中国药科大学 雷公藤红素(异)阿魏酸酯类衍生物及其制备方法与用途
WO2021047672A1 (en) * 2019-09-12 2021-03-18 Ixmedicine Co., Ltd Triterpenoid compounds, pharmaceutical compositions thereof, and their use for treating nuclear receptor subfamily 4 group member 1-mediated disease
CN111620924A (zh) * 2020-06-04 2020-09-04 华中农业大学 基于天然产物的药物设计方法、五环三萜类化合物、其制备方法及应用
CN113234116A (zh) * 2021-01-26 2021-08-10 延边大学 一种雷公藤红素衍生物及其制备方法和医用用途
CN113234116B (zh) * 2021-01-26 2022-02-25 延边大学 一种雷公藤红素衍生物及其制备方法和医用用途
CN116023426A (zh) * 2022-12-30 2023-04-28 上海海洋大学 去甲泽拉木醛衍生物及其在制备抗癌药物中的应用

Also Published As

Publication number Publication date
CN108026141A (zh) 2018-05-11
CN108026142A (zh) 2018-05-11
JP2019523245A (ja) 2019-08-22
CN108026141B (zh) 2022-03-29
CN108026142B (zh) 2022-07-08
EP3480207A4 (en) 2020-03-11
EP3480207A1 (en) 2019-05-08
WO2018006804A1 (zh) 2018-01-11
US10808005B2 (en) 2020-10-20
US20190330261A1 (en) 2019-10-31

Similar Documents

Publication Publication Date Title
WO2018006801A1 (zh) 孤儿核受体Nur77的配体及其用途
TWI776886B (zh) Sestrin-gator2交互作用之調節劑及其用途
JP2021531325A (ja) 甲状腺ホルモン受容体アゴニスト及びその用途
JP2021513532A (ja) ステロイド系誘導体モジュレーター、その製造方法及び応用
CA2773713A1 (en) Novel trpa1 antagonists
US11866452B2 (en) Biphenyl compound as immunomodulator, preparation method therefor and application thereof
Chen et al. Discovery of novel liver X receptor inverse agonists as lipogenesis inhibitors
WO2020143640A1 (zh) 甾体类化合物、用途及其制备方法
US20090215908A1 (en) Toll like receptor (tlr) signaling antagonist
KR102206745B1 (ko) 안토시아닌 및 gabab 수용체 작용제를 유효성분으로 포함하는 신경질환 치료제
JP6808154B2 (ja) アダマンタン誘導体およびその使用
WO2017143946A1 (zh) 一类脂肪酸类化合物、其制备方法及其用途
JP7138647B2 (ja) Rorガンマモジュレーター及びその使用
JP2010526812A (ja) 炎症性疾患の治療のためのスピロ化合物
TW201922690A (zh) 環-amp反應元素結合蛋白的抑制劑
JP2020505413A (ja) Rorガンマモジュレーター及びその使用
EP3937923B1 (en) Synthetic retinoids for use in rar activation
US8143260B2 (en) Tricyclic amine compound
WO2020140960A1 (zh) 吲哚甲酰胺类衍生物的晶型及其制备方法
EP4309652A1 (en) Composition for use in the prevention and/or treatment of diseases mediated by tlr4, il1r, cox1/2 and/or rbp4
US10519179B2 (en) Highly efficient NRF2 activators-co-releasing molecule hybrids, their use in the treatment of inflammatory or cardiovascular diseases and their process of preparation
WO2024073673A1 (en) Modulators of cd1 protein binding to t cell receptors
EP4067367A1 (en) Oleanane cinnamamide derivative, preparation method therefor, and use thereof
CA3132179A1 (en) Method of inhibiting trem-1
WO2023023594A1 (en) 2‑diarylmethyl‑4‑aminotetrahydropyran derivatives and related compounds as anticancer, antiinflammatory, antifibrotic and neuroprotective agents

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: 17823613

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17823613

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17823613

Country of ref document: EP

Kind code of ref document: A1

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

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 07.08.2019)

122 Ep: pct application non-entry in european phase

Ref document number: 17823613

Country of ref document: EP

Kind code of ref document: A1