WO2012025054A1 - 小檗胺的二酰亚胺化衍生物、其制备方法和应用 - Google Patents

小檗胺的二酰亚胺化衍生物、其制备方法和应用 Download PDF

Info

Publication number
WO2012025054A1
WO2012025054A1 PCT/CN2011/078905 CN2011078905W WO2012025054A1 WO 2012025054 A1 WO2012025054 A1 WO 2012025054A1 CN 2011078905 W CN2011078905 W CN 2011078905W WO 2012025054 A1 WO2012025054 A1 WO 2012025054A1
Authority
WO
WIPO (PCT)
Prior art keywords
berbamine
group
derivative
pharmaceutically acceptable
formula
Prior art date
Application number
PCT/CN2011/078905
Other languages
English (en)
French (fr)
Inventor
徐荣臻
荣风光
谢福文
赖洪喜
Original Assignee
杭州本生药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 杭州本生药业有限公司 filed Critical 杭州本生药业有限公司
Priority to JP2013525130A priority Critical patent/JP5701387B2/ja
Priority to CN201180004924.3A priority patent/CN102712655B/zh
Priority to DK11819429.9T priority patent/DK2610257T3/en
Priority to US13/819,111 priority patent/US9249137B2/en
Priority to EP11819429.9A priority patent/EP2610257B1/en
Publication of WO2012025054A1 publication Critical patent/WO2012025054A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/16Peri-condensed systems

Definitions

  • the invention belongs to the field of natural medicines and medicinal chemistry, relates to novel berbamine derivatives, in particular to diimidized derivatives of berbamine, to a method for preparing the same, a composition comprising the same, and a preparation thereof Use in anti-tumor drugs.
  • Background technique
  • Berbamine is also known as 6,6,7-trimethoxy-2,2,-dimethylberbaman-12-ol (6 5 6',7-trimethoxy-2,2' -dimethylberbaman-12-ol ) is a bisbenzylisoquinoline alkaloid extracted from Chinese herbal medicine Berberis. Xiao Yan
  • Berbamine stimulates myeloblast proliferation and increases hematopoietic stem cell colony factor
  • the content of (GCSF) promotes the proliferation of bone marrow hematopoietic stem cells and granulocyte progenitor cells, and differentiates into granulocyte cells and promotes the proliferation of leukocytes (Lin Chuanrong, et al., Clinical observation of leukocapac (pefamide) in the treatment of chemotherapy-induced leukopenia, Chinese patent medicine, 1994, 16 (7): 29).
  • Berbamine inhibits the proliferation of prostate cancer PC-3 cells by inducing apoptosis and affecting the cell cycle in a time and concentration dependent manner. (Sun Peng et al, berberine induced apoptosis and mechanism of prostate cancer PC-3 cells, Chinese Journal of Experimental Surgery, 2007, 24 (8): 957).
  • Berbamine has obvious proliferation inhibition effect and clear apoptosis-inducing effect on K562 cells in vitro, and it has a time-concentration-dependent relationship. In the tumor-bearing mice, berbamine also has a significant inhibitory effect on the growth of K562 cells. In particular, it can down-regulate the expression level of bcr/abl mRNA in tumor tissue cells. (Wu Dong et al., Experimental study on the effect of berbamine on K562 cells in vitro and in vivo, Chinese Journal of Experimental Hematology, 2005, 13 (3): 373). Berbamine inhibits cytotoxic T lymphocytes and promotes the activity of natural killer cells in vitro. It induces high levels of interleukin II in vitro and in vivo.
  • IL-2 can avoid the side effects caused by high dose IL-2 treatment of tumors.
  • berbamine has a good protective effect on the immune system of radiation-induced mice (Liu Xin et al., Immunomodulatory effects of berbamine on BALB/C mice, Journal of China Medical University, 1996, 25 (3): 229; Luo Chongnian et al. Inhibition of spleen cytotoxic sputum lymphocyte activity by berberine, Chinese Journal of Pharmacology and Toxicology, 1995, 9 (2): 159-160; Ge Mingzhu et al. Experimental study on the immunoprotective effects of mice, J. Immunol., 1998, 14 (4): 238).
  • berbamine can selectively inhibit the apoptosis of human leukemia Jurkat cells, arrest the cell cycle in the S phase, and increase the expression of caspase-3 protein.
  • concentration of the drug increased from 0.5 ug/mL to 10 ug/mL, the cell viability decreased from 93.69% to 14.85%. In this concentration range, berberine had no obvious cytotoxic effect on normal human peripheral blood leukocytes.
  • berbamine and some of its derivatives have significant inhibitory effects on brain malignant glioma cells, human cervical cancer cells, ascites cancer cells, and melanoma cells (Zhang Jinhong et al., The structure of berbamine and its derivatives for cervical cancer)
  • [0-(4-ethoxy)-butyl]- berbamine is a highly specific CaM antagonist with a specificity coefficient 6.5 times higher than that of berbamine. EBB induces apoptosis of lung cancer cells, and maintains the normal biological functions of major organ cells (Duan Jiangyan et al, [0-(4-ethoxy)-butyl]- berbamine induces apoptosis in lung cancer cells, Journal of Shanxi Normal University ( Natural Science Edition), 2001, 15 (4): 55).
  • Another berbamine derivative is 0-dansylsulfonyl berbamine (DB), which contains hydrophobicity. Fluorescent group.
  • DB inhibits the Ca 2+ + Mg 2+ ATPase activity of CaM-dependent erythrocyte membrane 25 times stronger than that of berbamine; DB has a significant inhibitory effect on intracellular granzyme phosphodiesterase activity, and there is dose and activity. relationship.
  • DB has a stronger effect on lung cancer cells than small guanamine, but has less cytotoxicity to human embryonic lung cells, and its inhibition of lung cancer cells is related to the inhibition of protooncogenes and to the control of inactivated tumor suppressor genes (Zhang Jinhong) Et al., Effect of calmodulin antagonist 0-dansyl berbamine on phosphodiesterase and lung cell proliferation, Journal of Nankai University (Natural Science), 2001, 34 (3): 64).
  • the reported small amide compounds have only transiently inhibited tumor cell growth and do not completely eliminate tumors.
  • hematological malignancies such as leukemia, multiple myeloma, lymphoma, and solid tumors such as liver cancer, lung cancer, breast cancer, prostate cancer, osteosarcoma and the like. It is obvious that the small anti-tumor activity of berbamine is still to be further studied and developed. Summary of the invention
  • R! is selected from the group consisting of H, aryl, amino, nitro, cyano, hydroxy, decyl, substituted or unsubstituted CC 6 alkyl, substituted or unsubstituted C r C 6 oxy, substituted or unsubstituted - C 6 alkyl, substituted or unsubstituted C r C 6 alkylamino, substituted or unsubstituted C 3 -C 7 cycloalkyl;
  • A is selected from linear or branched, substituted or unsubstituted alkylene-(CH 2 ) n - optionally interrupted by a hetero atom selected from 0, N and S, wherein n 1-15 Integer
  • W, X, Y and Z are independently selected from substituted or unsubstituted methine CH, fluorenylene C And a hetero atom selected from the group consisting of 0, N and S, wherein at least two of W, X, Y and Z are CH or CH 2 ;
  • the "" is substituted by a substituent selected from the group consisting of: alkene, an amine group, a nitro group, a cyano group, a hydroxyl group, and a fluorenyl group.
  • A is an unsubstituted alkylene group -(CH 2 ) n .
  • the present invention provides an phthalimide derivative of berbamine of the formula (I-a),
  • One of the groups is as defined in formula (I).
  • One of the groups is as defined in the general formula (I).
  • Another object of the invention is to provide a process for the preparation of a compound of the formula I according to the invention:
  • Compound (I) is prepared by reacting a compound of formula (m) with a compound of formula (III) wherein R l5 A, W, X, Y, Z are the same as defined above in formula (I); LG is a leaving group which may be, but not limited to, a halogen, a sulfonate group or the like.
  • Another object of the invention is to provide a pharmaceutical composition comprising a compound of the invention, said pharmaceutical composition comprising at least one compound of the invention, and optionally pharmaceutically acceptable! !Vulture agent.
  • Another object of the present invention is to provide a use of a compound of the present invention or a pharmaceutical composition comprising a compound of a compound for the preparation of a medicament, particularly an antitumor medicament.
  • the invention provides a method of treating a tumor patient comprising administering to a patient in need of treatment a therapeutically effective amount of at least one compound of the invention.
  • the tumor is particularly selected from the group consisting of leukemia, multiple myeloma, lymphoma, liver cancer, gastric cancer, breast cancer, cholangiocarcinoma, pancreatic cancer, lung cancer, colon cancer, osteosarcoma, melanoma, prostate cancer and the like.
  • the invention also relates to compounds of the invention for use in the treatment of tumors. Detailed ways
  • the present invention relates to a dioxime imidized derivative of the novel berbamine of the formula (I) or a pharmacologically acceptable thereof
  • W, X, Y and Z are independently selected from substituted or unsubstituted methine CH, fluorenylene CH 2 And a hetero atom selected from the group consisting of 0, N and S, wherein at least two of W, X, Y and Z are CH or CH 2 ;
  • substitution is substituted by a substituent selected from the group consisting of a aryl group, an amine group, a nitro group, a cyano group, a hydroxyl group and a fluorenyl group.
  • the present invention provides a phthalimide of the small guanamine of the formula (I-a). Aminated derivative.
  • the present invention provides an aromatic heterocyclic ring of a small guanamine of the formula (Ib) Dimethylimidized derivatives ⁇ ,
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 15, but when 1 is hydrogen, n is not 2.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 15, but when n is hydrogen, n is not 3.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 10.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 10, but when hydrogen is, n is not 2.
  • the invention relates to compounds of Formula Ia, wherein n is an integer from 1 to 10, but n is not 3 when 1 is hydrogen. In one embodiment, the invention relates to compounds of Formula Ia, wherein n is an integer from 1-8.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 8, but when hydrogen is, n is not 2.
  • the invention relates to compounds of formula i-a, wherein n is an integer from 1 to 8, but! When ⁇ is hydrogen, n is not 3.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 7.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 7, but n is not 2 when 1 ⁇ is hydrogen.
  • the invention relates to compounds of formula I-a, wherein n is an integer from 1 to 7, but n is not 3 when 1 is hydrogen.
  • the invention relates to compounds of formula Ia and Ib, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15, Or n is an integer in the range of any of these numbers, for example, n is 4-7, 4-8, 4-9,
  • the invention relates to a compound of formula I selected from the group consisting of H, CC 6 alkoxy, 13 ⁇ 4 generation 0 3 -: 6 alkoxy, C r C 6 alkylthio, generation ( ⁇ -( 6 alkylthio, C r C 6 alkyl, substituting ( ⁇ - C 6 alkyl, C r C 7 cycloalkyl, substituting (3 - C 7 cycloalkyl, 13 ⁇ 4, nitro, optionally substituted by one or Two C r C 6 alkyl substituted amino groups and a cyano group.
  • the invention relates to compounds of formula I, R 1 is H, C r C 6 alkoxy, C r C 6 alkylthio, C r C 6 alkyl, C 3 -C 7 ring An alkyl group, a nitro group, an amino group optionally substituted by one or two C r C 6 alkyl groups or a cyano group.
  • the present invention relates to a compound of formula I, 1 ⁇ to 11, C r C 6 alkoxy, C r C 6 alkyl, C r C 7 cycloalkyl or halo.
  • the present invention relates to a compound of formula I, 1 ⁇ to 11, C r C 3 alkoxy, CC 3 alkyl or C 5 -C 6 alkyl bad.
  • the invention relates to a compound of formula I, 1 is 11, decyloxy Or methyl.
  • the invention relates to a compound of formula I, 13 ⁇ 4 is 11.
  • the present invention relates to compounds of formula I, wherein is selected from substituted or unsubstituted alkyl c r c 6, the "substituted” is selected from the following substituents: halogen, amine , nitro, cyano, hydroxy and fluorenyl. .
  • the present invention relates to compounds of formula I, wherein is selected from substituted or unsubstituted C r C 6 alkoxy, said "substitution” is selected from the following substituents: halogen, amine Base, nitro, cyano, hydroxy and fluorenyl.
  • the invention relates to compounds of formula I, wherein! ⁇ is selected from a substituted or unsubstituted C r C 6 alkylthio group, which is substituted by a substituent selected from the group consisting of halogen, amine, nitro, cyano, hydroxy and decyl.
  • the invention relates to compounds of formula I, wherein 1 ⁇ is selected from substituted or unsubstituted CrC ⁇ amine groups, said "substituted” being substituted by a substituent selected from the group consisting of: halogen, Amine, nitro, cyano, hydroxy and fluorenyl.
  • the present invention relates to compounds of general formula I, wherein 1 ⁇ is selected from substituted or unsubstituted C r C 7 cycloalkyl, said "substitution” is selected from the following substituents: halogen, Amine, nitro, cyano, hydroxy and fluorenyl.
  • the position of the substituent in the compound of formula I of the invention is in the alpha position.
  • the compounds of formula I of the invention are in! ⁇
  • the position of the substituent is in the ⁇ position.
  • the substituents of the compounds of formula I of the invention are positioned at the ex and beta positions.
  • the invention particularly preferably comprises a compound of formula I or a pharmaceutically acceptable salt thereof:
  • C r C 6 alkyl means a straight or branched chain hydrocarbon containing 1-6 carbon atoms.
  • Examples of C r C 6 alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, t-butyl, n-pentyl, and n-hexyl.
  • C r C 6 alkoxy refers to -0- c r c 6 alkyl.
  • C r C 6 alkylthio group refers to -S- C c 6 alkyl.
  • C 3 -C 7 cycloalkyl refers to a hydrocarbon of a 3-7 membered monocyclic ring system having a saturated ring, and the C 3 -C 7 cycloalkyl group may be a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, or a ring. Hexyl, cycloheptyl.
  • substituted in the present invention may be one or more depending on the amount of hydrogen on the group having a substituent and the stability of the chemically-based combination. When a plurality of substituents are present, these substituents may be the same or different.
  • the term "pharmaceutically acceptable salts of the compounds of formula (I)” is an organic acid salt formed from an organic acid which forms a pharmaceutically acceptable anion, including but not limited to terpene sulfonate. Acid salt, sulfonate, malate, acetate, citrate, malonate, tartrate, succinate, benzoate, ascorbate, lactate, alpha-ketoglutarate Salts and alpha-glycerol phosphates; also suitable inorganic salts, including but not limited to hydrochlorides, sulfates, nitrates, bicarbonates and carbonates, phosphates, hydrobromides, hydrogenates Salt and so on.
  • compositions can be obtained using standard procedures well known in the art, for example, by reacting a sufficient amount of a basic compound with a suitable acid which provides a pharmaceutically acceptable anion.
  • polymorph refers to the solid crystalline form of a compound of the invention or a complex thereof. Different polymorphs of the same compound may exhibit different physical, chemical and/or spectral properties. Different physical properties include, but are not limited to, stability (eg, for heat or light), compressibility and density (important for formulation and product production), and dissolution rate (which can affect bioavailability).
  • Differences in stability can cause chemical reactivity (eg, differential oxidation, such that when composed of a polymorph Another polymorph constitutes a faster fading of the dosage form) or mechanical properties (eg, tablet granules that are kinetically favorable polymorphs when stored are converted to thermodynamically more stable polymorphs) or both (for example, a tablet of a polymorph is more susceptible to breakage at high humidity).
  • chemical reactivity eg, differential oxidation, such that when composed of a polymorph Another polymorph constitutes a faster fading of the dosage form
  • mechanical properties eg, tablet granules that are kinetically favorable polymorphs when stored are converted to thermodynamically more stable polymorphs
  • both for example, a tablet of a polymorph is more susceptible to breakage at high humidity.
  • the different physical properties of polymorphs can affect their processing. For example, one polymorph may be more likely to form a solvate than the other or may be more difficult to filter or wash away than the other due to, for example
  • hydrate refers to a compound of the invention or a salt thereof, which further comprises a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • prodrug refers to a derivative which can be hydrolyzed, oxidized or otherwise reacted under biological conditions (in vitro or in vivo) to provide a compound of the invention. Prodrugs undergo this reaction to become active compounds only under biological conditions, or they are active in their unreacted form. Prodrugs can generally be prepared using well-known methods, such as those described in 1 Burger's Medicinal Chemistry and Drug Discovery (1995) 172-178, 949-982 (Manfred E. Wolff, ed., 5th edition).
  • the small guanamine ring moiety of the compound of the present invention has a stereochemical structure as shown by the structural formula of Formula I.
  • the definitions and conventions for stereochemistry used herein generally follow MCGRAW- HILL DICTIONARY OF CHEMICAL TERMS (SP Parker, Ed., McGraw-Hill Book Company, New York, 1984); and ELIEL, E. and WILEN, S., STEREOCHEMISTRY OF ORGANIC COMPOUNDS (John Wiley & Sons, Inc., New York, 1994).
  • Many organic compounds exist in optically active forms, i.e., they have the ability to rotate planes of plane polarization.
  • treating generally refers to obtaining the desired pharmacological and/or physiological effects.
  • the effect may be prophylactic according to the prevention of the disease or its symptoms, in whole or in part; and/or may be therapeutic according to the partial or complete stabilization or cure of the disease and/or side effects due to the disease.
  • treatment covers any treatment for a patient's condition, including: (a) prevention of a disease that is susceptible to an infectious disease or condition but has not yet been diagnosed. Disease or symptom; (b) inhibiting the symptoms of the disease, ie preventing its development; or (c) alleviating the symptoms of the disease, ie, causing the disease or symptoms to degenerate.
  • the compounds of the present invention can be prepared according to conventional organic chemical synthesis methods.
  • the present invention relates to a process for the preparation of a compound of formula (I),
  • compound (I) is prepared by reacting a compound of formula (III) with a compound of formula ( ⁇ ), wherein ⁇ , ⁇ , W, X, Y, Z and 1 ⁇ are as defined above in formula (I)
  • LG is a leaving group, which may be, but not limited to, a halogen (e.g., chlorine, bromine, iodine), a sulfonate group (e.g., a mesylate group, a p-toluenesulfonate group), and the like.
  • the reaction is generally carried out in the presence of a base, which may be, but not limited to, sodium, sodium hydride, sodium hydroxide or potassium hydroxide.
  • a base which may be, but not limited to, sodium, sodium hydride, sodium hydroxide or potassium hydroxide.
  • the reaction is generally carried out in a solvent.
  • the solvent to be used includes, but is not limited to, an aprotic polar solvent such as dimercaptosulfoxide (DMSO), dimercaptocarboxamide (DMF) or hexamethylphosphoramide (hydrazine).
  • DMSO dimercaptosulfoxide
  • DMF dimercaptocarboxamide
  • hydrazine hexamethylphosphoramide
  • the reaction temperature of the reaction is usually from 0 ° C to room temperature, and generally varies depending on the starting materials used and the base.
  • the formula ( ⁇ ) compound is a small guanamine obtained by separating and extracting a natural product, which is commercially available.
  • N-(2-chloroethyl)-phthalimide N-(5-bromopentyl)-phthalimide, N-(7-bromoheptyl)-o-phenylene Imide, N-(3-bromopropyl)-pyridinedicarboximide, and the like.
  • the compound of the formula (III) can be synthesized by a conventional method, for example, by an optionally substituted phthalic phthalimide or a pyridine diimide and an alkylene group II! 3 ⁇ 4 (such as alkylene dichloride or dibromide) or alkylene disulfonate (such as phthalate or tosylate) is prepared by reacting in the presence of a base.
  • an alkylene group II! 3 ⁇ 4 such as alkylene dichloride or dibromide
  • alkylene disulfonate such as phthalate or tosylate
  • the reactant alkylene bis(3 ⁇ 4) or alkylene disulfonate can be obtained by I3 ⁇ 4 or esterification of an alkylene glycol.
  • Protecting groups are those which, once attached to an active moiety (eg, a hydroxyl or amino group), prevent such hydroxy protecting groups include, but are not limited to, alkyl, benzyl, allyl, triphenyl fluorenyl (ie, triphenylmethyl)
  • An acyl group for example, benzoyl, acetyl or HOOC-X"-CO-, X" is an alkylene, alkenylene, cycloalkylene or arylene group
  • a silyl group for example, Trimethylsilyl, triethylsilyl and tert-butyldimethylfluorenyl
  • alkoxycarbonyl aminocarbonyl (eg, dimethylaminoindenyl, methylethylaminoindenyl, and benzene) Aminocarbonyl), alkoxymethyl, benzyloxy and alkyl fluorenyl.
  • amino protecting group examples include, but are not limited to, an alkoxycarbonyl group, an alkanoyl group, an aryloxycarbonyl group, an aryl-substituted alkyl group, and the like. Hydroxy and amino protecting groups have been discussed in T. W. Greene and P. G.. M. Wuts, Protective Groups in Organic Synthesis, 2nd ed., John Wiley and Sons (1991). Both the hydroxy group and the amino protecting group can be removed by a conventional method after the reaction.
  • the invention also provides a pharmaceutical composition comprising a compound of formula I of the invention.
  • the invention provides a pharmaceutical composition comprising at least one of the compounds of formula I of the invention as described above, and optionally a pharmaceutically acceptable excipient.
  • the pharmaceutical preparation of the present invention is produced by a known method, including a conventional mixing, dissolving or lyophilizing method.
  • the compounds of the invention may be formulated into pharmaceutical compositions and administered to a patient in a variety of routes suitable for the chosen mode of administration, e.g., orally or parenterally (by intravenous, intramuscular, topical or subcutaneous routes).
  • the compounds of the invention may be administered systemically, e.g., orally, in combination with a pharmaceutically acceptable carrier such as an inert diluent or an assimilable edible carrier. They can be enclosed in hard or soft shell gelatin capsules and can be compressed into tablets.
  • a pharmaceutically acceptable carrier such as an inert diluent or an assimilable edible carrier. They can be enclosed in hard or soft shell gelatin capsules and can be compressed into tablets.
  • the active compound may be combined with one or more excipients and in the form of swallowable tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Form use.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the ratio of such compositions and formulations may of course vary and may range from about 1% to about 99% by weight of a given unit dosage form. In such therapeutically useful compositions, the amount of active compound enables an effective dosage level to be obtained
  • Tablets, lozenges, pills, capsules, and the like may also contain: a binder such as tragacanth, acacia, corn starch or gelatin; an excipient such as dicalcium phosphate; a disintegrant such as corn starch , potato starch, alginic acid, etc.; a lubricant such as magnesium stearate; and a sweetener such as sucrose, fructose, lactose or aspartame; or a flavoring agent such as mint, wintergreen or cherry.
  • a binder such as tragacanth, acacia, corn starch or gelatin
  • an excipient such as dicalcium phosphate
  • a disintegrant such as corn starch , potato starch, alginic acid, etc.
  • a lubricant such as magnesium stearate
  • a sweetener such as sucrose, fructose, lactose or aspartame
  • a flavoring agent such as mint, winter
  • any material used to prepare any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound can be incorporated into a sustained release preparation and a sustained release device.
  • the active compound can also be administered intravenously or intraperitoneally by infusion or injection.
  • An aqueous solution of the active compound or a salt thereof can be prepared, optionally mixed with a non-toxic surfactant.
  • Dispersants in glycerol, liquid polyethylene glycols, triacetin and mixtures thereof, and oils are prepared. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • Pharmaceutical dosage forms suitable for injection or infusion may include sterile aqueous solutions or dispersions of the active ingredient (optionally encapsulated in liposomes) containing the immediate formulation of a suitable injectable or injectable solution or dispersing agent. Or sterile powder. In all cases, the final dosage form must be sterile, liquid, and stable under the conditions of manufacture and storage.
  • the liquid carrier can be a solvent or liquid dispersion medium including, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), vegetable oils, non-toxic glycerides, and suitable mixtures thereof.
  • Appropriate fluidity can be maintained, for example, by liposome formation, by maintaining the desired particle size in the case of dispersing agents, or by the use of surfactants.
  • Microbial action can be prevented by various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol, sorbic acid, thimerosal, etc.).
  • isotonic agents such as sugars, buffers or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use of compositions that delay the absorption (e.g., aluminum monostearate and gelatin).
  • Sterile injectable solutions are prepared by combining the required active compound in a suitable solvent with the various other ingredients listed above, followed by filter sterilization.
  • the preferred preparation methods are vacuum drying and freeze drying techniques which result in the active ingredient plus any additional ingredients present in the previously sterile filtration solution. powder.
  • Useful solid carriers include comminuted solids (e.g., talc, clay, microcrystalline cellulose, silica, alumina, etc.).
  • Useful liquid carriers include water, ethanol or ethylene glycol or a water-ethanol/ethylene glycol mixture, and the compounds of the present invention may be dissolved or dispersed in an effective amount, optionally with the aid of a non-toxic surfactant.
  • Adjuvants e.g., fragrances
  • additional antimicrobial agents can be added to optimize the properties for a given use.
  • Thickeners can also be used with liquid carriers to form coatable pastes, gels, ointments. , soap, etc., used directly on the user's skin.
  • the therapeutic requirements of a compound or an active salt or derivative thereof depend not only on the particular salt selected, but also on the mode of administration, the nature of the disease to be treated, and the age and condition of the patient, ultimately depending on the attending physician or clinician decision.
  • unit dosage form is a unit dispersion unit containing a unit dosage unit suitable for administration to humans and other mammalian bodies.
  • the unit dosage form can be a capsule or tablet, or a lot of capsules or tablets.
  • the amount of unit dose of the active ingredient can vary or be adjusted between about 0.1 to about 1000 grams or more.
  • the invention provides a method of treating a tumor patient comprising administering to a patient in need of treatment a therapeutically effective amount of at least one compound of the invention.
  • the berbamine derivative of the present invention or a pharmaceutically acceptable salt thereof can be used, for example, for treating leukemia, multiple myeloma, lymphoma, liver cancer, gastric cancer, breast cancer, cholangiocarcinoma, pancreatic cancer, lung cancer, colon cancer, flesh Tumors, melanoma, human cervical cancer, glioma, nasopharyngeal carcinoma, laryngeal cancer, esophageal cancer, middle ear tumor, prostate cancer and other tumors.
  • Leukemia cell line human K562 leukemia cell line (chronic myeloid leukemia, CML), K562/adr (resistant chronic myeloid leukemia, CML), NB4 (acute promyelocytic leukemia, AML), Kasumi-1 (acute marrow) Leukemia type M2, AML-M2), Jurkat (acute lymphocytic leukemia, ALL), H9 (acute lymphocytic leukemia, ALL;).
  • BBM Berylamine
  • 6000 well-grown leukemia cells were inoculated into the wells of a 96-well cell culture plate.
  • the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum.
  • Different concentrations of berbamines were added, mixed, and placed in a carbon dioxide (5% CO 2 ) cell incubator for 72 hours at 37 °C.
  • the viable cell concentration was then determined by the MTT method.
  • the cell viability was set to 100% in the control group (without compound treatment), and cell viability (%) and 72-hour leukemia cell growth inhibition concentration (72 h IC 5 value) were calculated.
  • Table 1 shows that the berbamine derivative of the present invention can induce human chronic medulla Leukemia, acute myeloid leukemia and acute lymphocytic leukemia cell death and inhibition of growth of these leukemia cells, compared with berbamine itself, the activity of the benzamine derivative of the invention against leukemia cells is markedly enhanced, wherein the berbamine derivative of the present invention (BS-BE-003) The activity of anti-human K562 chronic myeloid leukemia was increased by more than 5 times, and the activity of anti-K562/adr (resistant chronic myeloid leukemia) was increased by more than 6 times.
  • Example 5 Determination of cell activity of berberamine derivatives against human multiple myeloma and lymphoma of the present invention
  • Myeloma and lymphoma cell lines U266 (multiple myeloma), RPMI8226 (multiple myeloma), DOHH2 (lymphoma).
  • Main instruments Cell culture incubator, microplate reader.
  • the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum. Different concentrations of berbamines were added, mixed and placed in a carbon dioxide (5% CO 2 ) cell culture incubator 37. C culture for 72 hours. The viable cell concentration was then determined by the MTT method. In the present control group (without compound treatment), the cell viability was set to 100%, and cell viability (%) and 72-hour leukemia cell growth inhibition concentration (72 h IC 5 value) were calculated.
  • Table 1 shows that the diimidized derivatives of the berbamines of the present invention are capable of inducing death of human myeloma and lymphoma cells and inhibiting the growth of these tumor cells, and the novel berbamine derivatives are antimyeloma and compared with berbamine itself. Lymphoma cell activity is markedly enhanced, wherein the berbamine derivative (BS-BE-003) of the present invention is nearly 6-fold more potent against RPMI8226 (multiple myeloma).
  • Table 1 Determination of half-growth inhibition concentration of leukemia, lymphoma and multiple myeloma cells by diimidation derivatives of berbamine (72 hours, IC 5G value)
  • Example 6 Determination of anti-human solid tumor action of diimidized derivatives of berbamine of the present invention
  • Human solid tumor cell lines HepG2 (human hepatocellular carcinoma, HCC), A549 (human lung cancer), MCF-7 (breast cancer), PANC-1 (pancreatic cancer), PC-3 (prostate cancer), MG63 (osteosarcoma) ), AGS (gastric cancer), Huh7 (human liver cancer cells), Becap37 (human breast cancer cells), Hela (human cervical cancer cells), RKO (human colon adenocarcinoma cells), SW620 (human colon adenocarcinoma cells), SW480 ( Human colon cancer cells), MGC 803 (human gastric cancer cells).
  • HCC human hepatocellular carcinoma, HCC
  • A549 human lung cancer
  • MCF-7 breast cancer
  • PANC-1 pancreatic cancer
  • PC-3 prostate cancer
  • MG63 osteosarcoma
  • AGS gastric cancer
  • Huh7 human liver cancer cells
  • Becap37 human breast cancer cells
  • Hela human cervical cancer cells
  • RKO human colon adenocarcino
  • Main instruments Cell culture incubator, microplate reader.
  • the culture solution was a DMEM high glucose cell culture medium containing 10% fetal calf serum. Place in a carbon dioxide (5% C0 2 ) cell incubator at 37 °C for 24 hours, then add different concentrations of berbamines, mix and continue to set carbon dioxide (5% C0 2 ) cell incubator 37 °C Cultivate for 72 hours. Then, the concentration of the living cells was measured by the MTT method, and the cell viability (%) after the action of the drug was calculated. In this experiment, the cell viability of the control group (without compound treatment) was set to 100%.
  • Table 2 shows that the berbamine derivatives of the present invention are capable of inducing death of human solid tumor cells and inhibiting the growth of these tumor cells, and the activity of the berbamine derivatives of the present invention against solid tumor cells is markedly enhanced as compared with the small indamine itself, wherein the present invention
  • the small imide compound (3) has a 6-fold increase in anti-PANC-1 (pancreatic cancer) activity and a 6-fold increase in anti-MCF-7 (breast cancer) activity, and is resistant to HepG2 (human hepatocellular carcinoma, HCC).

Description

小檗胺的二酰亚胺化衍生物、 其制备方法和应用 技术领域
本发明属于天然药物及药物化学领域, 涉及新型小檗胺衍生物, 特别是小檗胺的二酰亚胺化衍生物, 还涉及制备这些化合物的方法、 包 含该化合物的组合物及其在制备抗肿瘤药物中的用途。 背景技术
小檗胺 ( berbamine, BBM ) 又称为 6,6,,7-三曱氧基-2,2,-二甲基 berbaman-12-醇 ( 656',7-trimethoxy-2,2'-dimethylberbaman-12-ol ) , 是从 中国中草药小檗属植物中提取的一种双苄基异喹啉类生物碱。 小檗
Figure imgf000003_0001
Berbamine Oxyacanthine Cocsuline
小檗胺 尖刺碱 防己苏林
CAS: 478-61-5 CAS: 15352-74-6 CAS: 26279-88-9
Figure imgf000004_0001
Tetrandrine Fangchinoline Cocsoline 汉防己甲萦 防己醇灵 防己索林
CAS: 518-34-3 CAS: 43 4352-70-4
Figure imgf000004_0002
Cycieanine Thalrugosaminine
环轮藤宁 Mol. Wt: 560.73 绉唐松草宁 Mol. Wt.: 652.78 CAS: 518-94-5 CAS: 22226-73-9
小檗胺及其类似物
小檗胺具有刺激髓细胞增殖作用, 能提高造血干细胞集落因子
( GCSF ) 的含量, 促进骨髓造血干细胞和粒祖细胞的增殖, 并向粒系 细胞分化, 促进白细胞的增生(林传荣等, 升白胶(小檗胺)治疗化疗 性白细胞减少症的临床观察, 中成药, 1994, 16 (7): 29 ) 。
小檗胺通过诱导细胞凋亡和影响细胞周期的方式抑制前列腺癌 PC- 3细胞的增殖, 并具有时间和浓度的依赖性。 (孙鵬等, 小檗胺 诱导前列腺癌 PC-3细胞凋亡及及机制, 中华实验外科杂志, 2007 , 24 (8): 957 ) 。
小檗胺在体外对 K562细胞具有明显的增殖抑制作用及明确的 诱导凋亡作用, 并呈时间-浓度依赖关系; 在荷瘤棵鼠体内, 小檗胺 同样具有显著的抑制 K562细胞生长的作用,尤其能下调瘤体组织细 胞 bcr/abl mRNA的表达水平。 (吴东等, 小檗胺对 K562细胞体外 及体内作用的实验研究, 中华实验血液学杂志, 2005 , 13 (3): 373 )。 小檗胺对细胞毒性 T淋巴细胞有抑制作用, 对小鼠体外自然杀伤 细胞活性有明显促进作用, 在体内外可诱生出较高水平白细胞介素 II
(IL-2), 可避免用大剂量 IL- 2治疗肿瘤时引起的毒副反应。 实验证明, 小檗胺对辐射损伤小鼠免疫系统有良好防护作用 (刘新等, 小檗胺对 BALB/C小鼠的免疫调节作用, 中国医科大学学报, 1996, 25 (3): 229; 罗崇念等, 小檗胺对小鼠脾细胞毒性 Τ淋巴细胞活性的抑制作用, 中国 药理学与毒理学杂志, 1995 , 9 (2): 159-160; 葛明珠等, 盐酸小檗胺对 辐射小鼠的免疫防护作用的实验研究,免疫学杂志, 1998 , 14 (4): 238 )。
小檗胺诱导人白血病 Jurkat细胞凋亡机制也有研究与报道。 其结果 表明, 小檗胺能选择性抑制人白血病 Jurkat细胞凋亡, 使细胞周期阻滞 于 S期, 同时细胞 caspase-3蛋白表达增高。 且随着药物作用浓度从 0.5 ug/mL增加到 10 ug/mL, 细胞存活率由 93.69%降低为 14.85%, 在此作 用浓度范围内小檗胺对正常人外周血白细胞无明显细胞毒作用。 (董志 煜等,小檗胺诱导人白血病 Jurkat细胞凋亡的实验研究,中国肿瘤, 2007 , 16 ( 9 ) :722 ) 。 中国已经批准盐酸小檗胺片的上市销售,用于治疗各种原因引起的 白细胞减少症, 包括预防癌症放疗、 化疗后白细胞的减少。
也有报道小檗胺对细胞增殖的抑制作用。例如小檗胺及其某些衍生 物对脑恶性胶质瘤细胞、 人宫颈癌细胞、 腹水癌细胞及黑色素瘤细胞有 明显的抑制作用 (张金红等, 小檗胺及其衍生物的结构对宫颈癌
( CHeLa ) 细胞生长增殖的影响, 南开大学学 ^艮 (自然科学), 1996 , 29 ( 2 ): 89; 张金红等, 小檗胺及其衍生物对恶性黑色素瘤细胞增殖的影 响, 中草药, 1997, 28 (8): 483; 张金红等, 小檗胺衍生物 (EBB )体 内抗肿瘤作用初探, 中草药, 1998 , 29 (4): 243; 段江燕等, 小檗胺类 化合物对黑色瘤细胞内钙调蛋白水平的影响,中草药,2002 , 33 (1): 59 )。 其中 [0-(4-乙氧基)-丁基]-小檗胺 (EBB ) 为专一性强的 CaM拮抗剂, 专一性系数高出小檗胺 6.5倍。 EBB诱导肺癌细胞凋亡, 同时维护主要 器官细胞正常生物功能(段江燕等, 〔0-(4-乙氧基) -丁基]-小檗胺诱导肺 癌细胞凋亡的初探, 山西师范大学学报(自然科学版) , 2001 , 15 (4): 55 ) 。 另一小檗胺衍生物为 0-丹磺酞基小檗胺(DB ) , 其含有疏水性 萤光基团。 DB对依赖于 CaM的红细胞膜 Ca2++Mg2+ ATPase抑制活性 比小檗胺强 25倍; DB对细胞内的颗粒酶磷酸二酯酶活性有明显的抑制 作用, 且存在剂量与活性的关系。 另外, 还发现 DB对肺癌细胞的作用 比小檗胺强, 而对人胚肺细胞细胞毒性小, 其抑制肺癌细胞除了与抑制 原癌基因有关, 也与控制失活的抑癌基因有关(张金红等, 钙调素拮抗 剂 0-丹酰基基小檗胺对磷酸二酯酶及肺细胞增殖的影响, 南开大学学 报 (自然科学) , 2001, 34 (3): 64 ) 。
本发明人在专利 CN 101273989A中描述了一类小檗胺衍生物在 用于制备治疗抗肿瘤药物中的应用, 主要涉及苯曱酰基和苄基衍生 物。
迄今为止, 所报道的小檗胺化合物只能短暂抑制肿瘤细胞生长, 不 能完全清除肿瘤。尤其是对血液系统恶性肿瘤如白血病、多发性骨髓瘤、 淋巴瘤, 和实体肿瘤如肝癌、 肺癌、 乳腺癌、 前列腺癌、 骨肉瘤等。 显 而易见, 抗肿瘤活性更高的小檗胺^^生物仍有待继续研究与开发。 发明内容
本发明的一个目的是提供通式 (I)的新型小檗胺二酰亚胺化衍生 物或其
Figure imgf000006_0001
R!选自 H、 素、 胺基、 硝基、 氰基、 羟基、 巯基、 取代或无取代 的 C C6烷基、 取代或无取代的 CrC6 ^氧基、 取代或无取代的 - C6 烷疏基、 取代或无取代的 CrC6烷胺基、 取代或无取代的 C3-C7环烷基;
A选自直链或支链的、 取代或无取代的、 任选地被选自 0、 N和 S 的杂原子中断的亚烷基- ( CH2 ) n -, 其中, n 1-15的整数;
W、 X、 Y和 Z独立地选自取代或无取代的次甲基 CH、 亚曱基 C 和选自 0、 N和 S的杂原子, 其中 W、 X、 Y和 Z中至少两个为 CH或 CH2;
所述 " 是由选自如下的取代基取代: !¾素、 胺基、 硝基、 氰 基、 羟基和巯基。
优选地, 通式 (I)中 W = X = Y Z = CH。
优选地, 通式 (I)中 A为无取代的亚烷基- (CH2 ) n
在一个优选的实施方案中, 本发明提供通式(I-a )的小檗胺的邻苯 二甲酰亚胺化衍生物,
Figure imgf000007_0001
式(l-a)
其中个基团如式 (I)中所定义。
当 W、 X、 Y和 Z中有一个或两个杂原子并且 A为无取代的烷基- ( CH2 ) n -时, 本发明提供的是通式(I- b )的小檗胺的芳香杂环二甲酰 亚胺化衍生物:
Figure imgf000007_0002
其中个基团如通式 (I)中所定义。
本发明的另一个目的是提供制备本发明通式 I化合物的方法:
Figure imgf000007_0003
III 化合物 (I)由式 (m)化合物与式 (Π)化合物反应制得 , 式 (III)中 Rl5 A, W, X, Y, Z与上文在通式 (I)中的定义相同; LG为离去基团, 其可以是 但不限于卤素、 磺酸酯基等。
本发明的另一个目的是提供包含本发明化合物的药物组合物, 所 述药物组合物包括至少一种本发明化合物, 和任选的药学上可以接受 的!!武形剂。
本发明的另一个目的是提供本发明化合物或包含谅化合物的药 物组合物在制备药物、 特别是抗肿瘤药物中的用途。 相应地, 本发明 提供一种治疗肿瘤患者的方法, 包括给予需要治疗的患者治疗有效 量的至少一种本发明的化合物。 所述肿瘤特别选自白血病、 多发性 骨髓瘤、 淋巴瘤、 肝癌、 胃癌、 乳腺癌、 胆管细胞癌、 胰腺癌、 肺 癌、 大肠癌、 骨肉瘤、 黑色素瘤、 前列腺癌等。
本发明还涉及用于治疗肿瘤的本发明的化合物。 具体实施方式
本发明涉及通式 (I)的新型小檗胺的二曱酰亚胺化衍生物或其药 学上
Figure imgf000008_0001
选自 H、 素、 胺基、 硝基、 氰基、 羟基、 巯基、 取代或无取代 的 Cr C6烷基、 取代或无取代的 (!-^烷氧基、 取代或无取代的 CrC6 烷硫基、 取代或无取代的 Cr6烷胺基、 取代或无取代的 C3-C7环烷基;
A 选自直链或支链的、 取代或无取代的、 任选地被选自 0、 N、 S 的杂原子中断的亚烷基- (CH2 ) n - , 其中 n = 1-15的整数;
W、 X、 Y和 Z独立地选自取代或无取代的次甲基 CH、 亚曱基 CH2 和选自 0、 N和 S的杂原子, 其中 W、 X、 Y和 Z中至少两个为 CH或 CH2;
所述 "取代" 是由选自如下的取代基取代: 素、 胺基、 硝基、 氰 基、 羟基和巯基。
当 W = X = Y = Z = CH和 A为无取代的亚烷基- (C¾ ) n -时, 本 发明提供的是通式 (I- a ) 的小檗胺的邻苯二甲酰亚胺化衍生物。
Figure imgf000009_0001
当 W、 X、 Y和 Z中有一个或两个杂原子并且 A为无取代的烷基 ( CH2 ) n -时, 本发明提供的是通式(I-b )的小檗胺的芳香杂环二甲酰 亚胺化衍生物 ·,
Figure imgf000009_0002
在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 n为 1-15的 整数, 但1^为氢时 n不为 2。
在一种实施方式中, 本发明涉及式 I- a的化合物, 其中 n为 1-15的 整数, 但1¾ 为氢时 n不为 3。
在一种实施方式中, 本发明涉及式 I- a的化合物, 其中 n为 1-10的 整数。
在一种实施方式中, 本发明涉及式 I- a的化合物, 其中 n为 1-10的 整数, 但 为氢时 n不为 2。
在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 n为 1-10的 整数, 但1^为氢时 n不为 3。 在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 η为 1-8的 整数。
在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 n为 1-8的 整数, 但 为氢时 n不为 2。
在一种实施方式中, 本发明涉及式 i-a的化合物, 其中 n为 1-8的 整数, 但!^为氢时 n不为 3。
在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 n为 1-7的 整数。
在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 n为 1-7的 整数, 但1^为氢时 n不为 2。
在一种实施方式中, 本发明涉及式 I-a的化合物, 其中 n为 1-7的 整数, 但1^为氢时 n不为 3。
在一种实施方式中,本发明涉及式 I-a和 I-b的化合物,其中 n为 1、 2、 3、 4、 5、 6、 7、 8、 9、 10、 11、 12、 13、 14或 15 , 或者 n为这些 数字中任意两个为端值组成的范围中的整数, 例如 n为 4-7、 4-8、 4-9、
4- 10、 4-11、 4-12, 4-13、 4-14、 4-15、 5-7、 5-8、 5-9、 5-10、 5-11、 5-12、
5- 13、 5-14、 5-15等。
在一种实施方式中, 本发明涉及通式 I的化合物, 选自 H、 C C6 烷氧基、 1¾代03-:6烷氧基、 CrC6烷硫基、 代(^-( 6烷硫基、 CrC6 烷基、 代(^- C6烷基、 CrC7环烷基、 代( 3- C7环烷基、 1¾素、 硝基、 任选由一个或两个 CrC6烷基取代的氨基和氰基。
在一种实施方式中, 本发明涉及通式 I的化合物, R1为 H、 CrC6 烷氧基、 CrC6烷硫基、 CrC6烷基、 C3-C7环烷基、 1¾素、 硝基、 任选 由一个或两个 CrC6烷基取代的氨基或氰基。
在一种实施方式中, 本发明涉及通式 I的化合物, 1^为 11、 CrC6烷 氧基、 CrC6烷基、 CrC7环烷基或卤素。
在一种实施方式中, 本发明涉及通式 I的化合物, 1^为 11、 CrC3烷 氧基、 C C3烷基或 C5-C6坏烷基。
在一种实施方式中, 本发明涉及通式 I的化合物, 1^为 11、 曱氧基 或甲基。
在一种实施方式中, 本发明涉及通式 I的化合物, 1¾ 为11。
在一种实施方式中,本发明涉及通式 I的化合物,其中 选自取代 或无取代的 crc6烷基, 所述 "取代" 是由选自如下的取代基取代: 鹵 素、 胺基、 硝基、 氰基、 羟基和巯基。 。
在一种实施方式中,本发明涉及通式 I的化合物, 其中 选自取代 或无取代的 CrC6烷氧基, 所述 "取代" 是由选自如下的取代基取代: 卤素、 胺基、 硝基、 氰基、 羟基和巯基。
在一种实施方式中, 本发明涉及通式 I的化合物, 其中!^选自取代 或无取代的 CrC6烷硫基, 所述 "取代" 是由选自如下的取代基取代: 卤素、 胺基、 硝基、 氰基、 羟基和巯基。
在一种实施方式中, 本发明涉及通式 I的化合物, 其中 1^选自取代 或无取代的 CrC ^胺基, 所述 "取代" 是由选自如下的取代基取代: 鹵素、 胺基、 硝基、 氰基、 羟基和巯基。
在一种实施方式中, 本发明涉及通式 I的化合物, 其中 1^选自取代 或无取代的 CrC7环烷基所述 "取代" 是由选自如下的取代基取代: 卤 素、 胺基、 硝基、 氰基、 羟基和巯基。
在一种实施方式中, 本发明通式 I化合物中 1^取代基的位置在 α 位。
在一种实施方式中, 本发明通式 I化合物中!^取代基的位置在 β 位。
在一种实施方式中,本发明通式 I化合物中 取代基的位置在 ex和 β位。
在一种实施方式中, 本发明涉及通式 I的化合物, 其中 Α选自直链 或支链的、 取代或无取代的、 任选地被选自 0、 N、 S的杂原子中断的 亚烷基- (CH2 ) n -, 其中 n = 1-15的整数。
在另一种实施方式中, 本发明特别优选如下的式 I化合物或其药学 上可接受的盐:
Figure imgf000012_0001
12-0- ((1,3-二氧代异吲哚啉 -2-基)-甲基)-小檗胺 (化合物 BS-BE- 001),
Figure imgf000012_0002
12-0- (5- (1,3-二氧代异吲哚啉 -2-基)-戊基 )-小檗胺(化合物 BS-BE- 002),
Figure imgf000012_0003
12-〇-(7- (1,3- BS-BE-003 ,
Figure imgf000012_0004
12- 0-(3- (3,4-吡啶二甲酰亚胺基) -丙基) -小檗胺 (化合物 BS-BE-004) 剂化物、 水合物、 加成物、 复合物、 多晶型物或前药。
如本文所使用, 术语" CrC6烷基"是指含有 1-6个碳原子的直链 或支链烃基。 CrC6烷基的例子包括但不限于甲基、 乙基、 正丙基、 异丙基、 叔丁基、 正戊基和正己基。
术语 "CrC6烷氧基 "是指 -0- crc6烷基。
术语 "Cr C6烷硫基 "是指 -S- C c6烷基。
术语" C3- C7环烷基 "是指具有饱和环的 3-7 元单环系统的烃, C3-C7环烷基可以为环丙基、 环丁基、 环戊基、 环己基、 环庚基。
术语" 1¾素 "是指氟、 氯、 溴或碘。
本发明中的 "取代基" 可以是一个或者多个, 取决于带有取代基的 基团上的氢的数量和化学基固组合的稳定性。 当存在多个取代基时, 这 些取代基可以相同或不同。
如本文所使用, 术语"式 (I)化合物的药学上可以接受的盐"的例子是 由形成药学上可以接受的阴离子的有机酸形成的有机酸盐,这些盐包括 是但不限于曱苯磺酸盐、 曱磺酸盐、 苹果酸盐、 醋酸盐、 柠檬酸盐、 丙 二酸盐、 酒石酸盐、 琥珀酸盐、 笨甲酸盐、 抗坏血酸益、 乳酸盐、 α-酮 戊二酸盐和 α-甘油磷酸盐; 也可形成合适的无机盐, 这些盐包括但不限 于盐酸盐、 硫酸盐、 硝酸盐、 碳酸氢盐和碳酸盐、 磷酸盐、 氢溴酸盐、 氢 酸盐等。
药学上可以接受的盐可使用本领域熟知的标准程序获得, 例如, 通 过将足量的碱性化合物和提供药学上可以接受的阴离子的合适的酸反 应生成。
如本文所使用 , 术语"多晶型物 "是指本发明的化合物或其复合 物的固体晶体形式。 相同化合物的不同的多晶型物可以显示不同的 物理、化学和 /或光谱性质。不同的物理性质包括但不限于稳定性(例 如, 对热或光) 、 可压缩性和密度 (对于配制制剂和产品生产是重 要的) 和溶解速率 (其可以影响生物利用度) 。 稳定性的不同会造 成化学反应性 (例如差异氧化, 使得当由一种多晶型物构成时比由 另一多晶型物构成时剂型更快地褪色) 或机械性能 (例如, 储存时 作为动力学有利的多晶型物的片剂碎末转化成热力学更加稳定的多 晶型物) 或两者 (例如, 一种多晶型物的片剂在高潮湿度时更加容 易破碎) 中的变化。 多晶型物的不同的物理性质可以影响它们的加 工。 例如, 一种多晶型物可能比另一种更可能形成溶剂化物或可能 比另一种更加难以过滤或洗去杂质, 这是由于例如其颗粒的形状或 大小分布。
如本文所使用, 术语"水合物 "是指本发明的化合物或其盐, 其 进一步包含通过非共价分子间力结合的化学计量的或非化学计量的 量的水。
如本文所使用, 除非另外说明, 术语"前药"是指可以在生物学 条件 (体外或体内) 下水解、 氧化或进行其他反应以提供本发明的 化合物的衍生物。 前药仅在生物学条件下经过该反应成为活性化合 物, 或者它们在它们不反应的形式中具有活性。 通常可以使用公知 的方法制备前药, 例如 1 Burger's Medicinal Chemistry and Drug Discovery (1995) 172- 178, 949- 982 (Manfred E. Wolff编, 第 5版)中描 述的那些方法。
本发明化合物中小檗胺环部分具有式 I 结构式所显示的立体化 学结构。 本文使用的立体化学的定义和约定一般遵循 MCGRAW- HILL DICTIONARY OF CHEMICAL TERMS (S. P. Parker, Ed., McGraw-Hill Book Company, New York, 1984); 和 ELIEL, E.和 WILEN, S., STEREOCHEMISTRY OF ORGANIC COMPOUNDS (John Wiley & Sons, Inc., New York, 1994)。许多 有机化合物以光学活性形式存在, 即它们具有旋转平面偏光的平面 的能力。
本文使用的术语"治疗"一般是指获得需要的药理和 /或生理效 应。 该效应根据完全或部分地预防疾病或其症状, 可以是预防性的; 和 /或根据部分或完全稳定或治愈疾病和 /或由于疾病产生的副作用, 可以是治疗性的。 本文使用的"治疗"涵盖了对患者疾病的任何治疗, 包括: ( a)预防易感染疾病或症状但还没诊断出患病的患者所发生的 疾病或症状; (b)抑制疾病的症状, 即阻止其发展; 或(c)緩解疾病的 症状, 即, 导致疾病或症状退化。
本发明的化合物可以按照常规的有机化学合成方法制备。例如,本 发明涉及一种制备式 (I)化合物的方法,
Figure imgf000015_0001
III II I 其中化合物 (I)由式 (III)化合物与式 (Π)化合物反应制得, 式中 Α, Β, W, X,Y, Z 和1^与上文在式 (I)中的定义相同; LG为离去基团, 可以是 但不限于卤素 (如: 氯、 溴、 碘) 、 磺酸酯基(如: 甲磺酸酯基、 对甲 苯磺酸酯基) 等。
该反应一般在碱存在下进行, 这里碱可以是但不限于钠、 氢化钠、 氢氧化钠或氢氧化钾等。
该反应一般在溶剂中进行。 吏用的溶剂包括但不限于非质子极性溶 剂, 例如二曱基亚砜(DMSO ) 、 二曱基甲酰胺(DMF )或六甲基磷酰 胺(ΗΜΡΤ ) 等。
该反应的反应温度一般为 0 °C至室温,一般随所用的反应原料以及 碱的不同而变化。
式 (Π)化合物为天然产物经分离提取得到的小檗胺, 可以在市场上 购买获得。
N-(2-氯乙基) -邻苯二曱酰亚胺、 N-(5-溴戊基)-邻苯二甲酰亚胺、 N-(7- 溴庚基)-邻苯二曱酰亚胺, N-(3-溴丙基)-吡啶二甲酰亚胺等。
式 (III)化合物可以通过常规方法合成制得, 例如通过任选取代的邻 苯二曱酰亚胺或吡啶二甲酰亚胺与亚烷基二! ¾ (如亚烷基二氯或二溴) 或亚烷基二磺酸酯 (如曱磧酸酯或甲苯磺酸酯)在碱存在下反应制得。 反应物亚烷基二! ¾或亚烷基二磺酸酯可以通过亚烷基二醇的 I¾化或酯 化反应制得。
常规的化学转换可用于实施本发明。 本领域的技术人员可以决 定用于这些化学转换的适当的化学剂、 溶剂、 保护基和反应条件。 相关信息描述于 , 例如 , R. Larock , Comprehensive Organic Transformations , VCH 出版商(1989); T.W. Greene和 P.G.M. Wuts , Protective Groups in Organic Synthesis , 第 3版, John Wiley and Sons( 1999); L. Fieser和 M. Fieser, Fieser and Fieser 's Reagents for Organic Synthesis , John Wiley and Sons(1994); 及 L. Paquette编辑的 Encyclopedia of Reagents for Organic Synthesis , John Wiley and Sons( 1995)及其后来的版本。 保护基指那些一旦连接活性部分 (例如, 羟基或氨基), 防止这些 羟基保护基的例子包括但不限于, 烷基、 苯甲基、 烯丙基、 三苯曱 基(即,三苯基甲基)、酰基 (例如,苯甲酰基、 乙酰基或 HOOC- X"-CO-, X"为亚烷基、 亚链烯基、 亚环烷基或亚芳基)、 甲硅烷基 (例如, 三甲 基甲硅烷基、 三乙基曱硅烷基和叔丁基二曱基曱硅烷基)、 烷氧基羰 基、 氨基羰基 (例如, 二甲基氨基齪基、 甲基乙氨基齪基和苯基氨基 羰基)、 烷氧曱基、 苯甲氧曱基和烷基巯曱基。 氨基保护基的例子包 括但不限于, 烷氧基羰基、 烷酰基、 芳氧基羰基、 芳基取代的烷基 等。 羟基和氨基保护基已在 T.W. Greene和 P.G..M. Wuts , Protective Groups in Organic Synthesis , 第 2版, John Wiley and Sons(1991)中讨 论。 羟基和氨基保护基都可在反应后通过常规的方法去除。 本发明还提供了包含本发明通式 I化合物的药物组合物。
本发明提供了这样的药物组合物, 其包含至少一种如上所述的 本发明的通式 I化合物, 和任选的药学上可以接受的赋形剂。
制备各种含有一定量的活性成分的药物组合物的方法是已知 的, 或根据本发明的公开内容对于本领域技术人员是显而易见的。 ¾口 REMINGTON'S PHARMACEUTICAL SCIENCES, Martin, E.W., ed., Mack Publishing Company, 19th ed. (1995)所述, 制备所述药物组合物的方 法包括掺入适当的药学赋形剂、 载体、 稀释剂等。
以已知的方法制造本发明的药物制剂, 包括常规的混合、 溶解 或冻干方法。
本发明的化合物可以制成药物组合物, 并向患者以适于选定的施用 方式的各种途径施用, 例如口服或肠胃外(通过静脉内、 肌内、 局部或 皮下途径) 。
因此, 本发明的化合物结合药学上可以接受的载体(如惰性稀释剂 或可同化的可食用的载体)可以全身施用, 例如, 口服。 它们可以封闭 在硬或软壳的明胶胶嚢中, 可以压为片剂。 对于口服治疗施用, 活性化 合物可以结合一种或多种赋形剂,并以可吞咽的片剂、颊含片剂、含片、 胶嚢剂、 酏剂、 悬浮剂、 糖浆、 圓片等的形式使用。 这种组合物和制剂 应该包含至少 0.1%的活性化合物。 这种组合物和制剂的比例当然可以 变化, 可以占给定的单位剂型重量的大约 1%至大约 99%。 在这种治疗 有用的组合物中, 活性化合物的量使得能够获得有效剂量水平。
片剂、 含片、 丸剂、 胶嚢剂等也可以包含: 粘合剂, 如黄蓍胶、 阿 拉伯胶、 玉米淀粉或明胶; 赋形剂, 如磷酸氢二钙; 崩解剂, 如玉米淀 粉、 马铃薯淀粉、 藻酸等; 润滑剂, 如硬脂酸镁; 和甜味剂, 如蔗糖、 果糖、 乳糖或阿司帕坦; 或调味剂, 如薄荷、 冬青油或樱桃香味。 当单 位剂型是胶嚢时, 除了上面类型的材料, 它还可以包含液体载体, 如植 物油或聚乙二醇。 各种其他材料可以存在, 作为包衣, 或以其他方式改 变固体单位剂型的物理形式。 例如, 片剂、 丸剂或胶嚢剂可以用明胶、 蜡、 虫胶或糖等包衣。 糖浆或酏剂可以包含活性化合物, 蔗糖或果糖作 为甜味剂, 对羟苯甲酸甲酯或对羟苯曱酸丙酯作为防腐剂, 染料和调味 剂 (如樱桃香料或桔子香料)。 当然, 用于制备任何单位剂型的任何材 料应该是药学上可以接受的且以应用的量基本上无毒。 此外, 活性化合 物可以掺入緩释制剂和緩释装置中。
活性化合物也可以通过输注或注射来静脉内或腹膜内施用。可以制 备活性化合物或其盐的水溶液, 任选地混和无毒的表面活性剂。 也可以 制备在甘油、 液体聚乙二醇、 甘油三乙酸酯及其混合物以及油中的分散 剂。 在普通的储存和使用条件下, 这些制剂包含防腐剂以防止微生物生 长。
适于注射或输注的药物剂型可以包括包含适于无菌的可注射或可 输注的溶液或分散剂的即时制剂的活性成分(任选封装在脂质体中)的 无菌水溶液或分散剂或无菌粉末。 在所有情况下, 最终的剂型在生产和 储存条件下必须是无菌的、 液体的和稳定的。 液体载体可以是溶剂或液 体分散介质, 包括, 例如水、 乙醇、 多元醇(例如, 甘油、 丙二醇、 液 体聚乙二醇等) 、 植物油、 无毒的甘油酯及其合适的混合物。 可以维持 合适的流动性, 例如, 通过脂质体的形成, 通过在分散剂的情况下维持 所需的粒子大小, 或通过表面活性剂的使用。 可以通过各种抗细菌剂和 抗真菌剂 (如对羟苯甲酸酯、 氯丁醇、 苯酚、 山梨酸、 硫柳汞等)产生 预防微生物的作用。 在许多情况下, 优选包括等渗剂, 如糖、 緩冲剂或 氯化钠。 通过使用延緩吸收剂的组合物(例如, 单硬脂酸铝和明胶)可 以产生可注射的组合物的延长吸收。
通过将合适的溶剂中的需要量的活性化合物与需要的上面列举的 各种其他成分结合, 然后进行过滤灭菌, 制备无菌可注射溶液。 在用于 制备无菌注射溶液的无菌粉末的情况下,优选的制备方法是真空千燥和 冷冻干燥技术,这会产生活性成分加上任何另外需要的以前无菌过滤溶 液中存在的成分的粉末。
有用的固体载体包括粉碎的固体(如滑石、 粘土、 微晶纤维素、 二 氧化硅、 氧化铝等) 。 有用的液体载体包括水、 乙醇或乙二醇或水-乙 醇 /乙二醇混合物, 本发明的化合物可以任选在无毒的表面活性剂的帮 助下以有效含量溶解或分散在其中。 可以加入佐剂 (如香味)和另外的 抗微生物剂来优化对于给定用途的性质。
增稠剂 (如合成的聚合物、 脂肪酸、 脂肪酸盐和酯、 脂肪醇、 改性 纤维素或改性无机材料)也可和液体载体用于形成可涂覆的糊剂、凝胶、 软膏、 肥皂等, 直接用于使用者的皮肤上。 化合物或其活性盐或衍生物的治疗需要量, 不仅取决于选择的特定 的盐,而且取决于施药方式、待治疗的疾病的本质和患者的年龄和状态, 最终取决于在场医师或临床医生的决定。
上述制剂可以以单位剂型存在, 该单位剂型是含有单位剂量的 物理分散单元, 适于向人体和其它哺乳动物体给药。 单位剂型可以 是胶嚢或片剂, 或是很多胶嚢或片剂。 根据所涉及的具体治疗, 活 性成分的单位剂量的量可以在大约 0.1到大约 1000亳克或更多之间 进行变化或调整。 药物、 特别是抗肿瘤药物中的用途。 相应地, 本发明提供一种治疗 肿瘤患者的方法, 包括给予需要治疗的患者治疗有效量的至少一种 本发明的化合物。 本发明的小檗胺衍生物或其药学上可接受的盐例 如可用于治疗白血病、 多发性骨髓瘤、 淋巴瘤、 肝癌、 胃癌、 乳腺 癌、 胆管细胞癌、 胰腺癌、 肺癌、 大肠癌、 骨肉瘤、 黑色素瘤、 人 宫颈癌、 神经胶质瘤、 鼻咽癌、 喉癌、 食管癌、 中耳肿瘤、 前列腺 癌等肿瘤。
在下列实施例中, 将更加具体地解释本发明。 但应理解, 下列 实施例旨在说明本发明而不对本发明的范围构成任何限制。
的合成方法获得 实施例 1:
Figure imgf000019_0001
在氮气保护气氛中, 在 0 °C下, 向溶于 Ν,Ν-二曱基甲酰胺 (5 ml)的 二盐酸小檗胺 (205 mg, 0.3 mmol)中加入 aH (48 mg, 1.2 mmol), 搅拌 1 小时后, 加入 2-氯曱基 -异吲哚啉 -1,3-二酮 (88 mg, 0.45 mmol)。 使反应 液加热至 80 °C过夜。 然后真空蒸发反应混合物, 并通过制备性薄层色 谱法纯化, 产生白色或淡黄色化合物 (BS- BE-001) (1 1.5 mg, 5.0 %)。
LC/MS m/z: M+l 768.3 100% (纯度)。
'Η NMR (CDC13) δ: 7.882-7.865 (dd, 2Η, J=6.0 Hz, 5.5 Hz),
7.749-7.732 (dd, 2H, J=5.5 Hz, 6.0 Hz), 7.264 (s , IH) , 7.019-6.998 (dd, 1H,J=8.5 Hz, 8.0 Hz) , 6.919-6.903 (d, IH, 7=7.5 Hz), 6.713-6.698 (d, IH, •7=7.5 Hz), 6.626-6.614 (m, IH), 6.527 (s, IH), 6.420-6.385 (m, IH), 6.266 (s, 1H), 5.954 (s, 1H)? 5.766-5.717 (m, 2H), 3.850 (s, 2H), 3.750 (s, 3H), 3.610 (s, 3H), 3.487-3.473 (m, IH), 3.396 (s, IH), 3.241-3.203 (m, 2H), 3.1 13 (s, 3H), 3.012-2.768 (m, 6H), 2.566 (s, 3H), 2.532 (s, 1H),
2.383-2.271 (m, IH), 2.216 (s, IH), 1.795-1.725 (m, 2H)。 实施例
Figure imgf000020_0001
在氮气保护气氛中, 在 0 °C下, 向溶于 Ν,Ν-二甲基曱酰胺 (5 ml)的 二盐酸小檗胺 (205 mg, 0.3 mmol)中加入 aH (48 mg, 1.2 mmol), 搅拌 1 小时后, 加入 2- (5-溴戊基) -异吲哚啉- 1,3-二酮(133 mg, 0.45 mmol)。 使 反应液加热至 80 °C过夜。 然后真空蒸发反应混合物, 并通过制备薄层 色谱法纯化以产生白色或淡黄色化合物 (BS-BE- 002) (98.4 mg, 39.8 %)。
LC/MS m/z: M+l 824.2 100% (纯度)。
]H NMR (CDCI3) δ: 7.826-7.809 (dd, 2H, J=5.5 Hz, 5.5 Hz),
7.692-7.680 (dd, 2H, 7=6.0 Hz, 5.5 Hz), 7.262-7.243 (m, 2H)5 7.084-7.071 (d, 1H, J= 6.5 Hz), 6.799-6.733 (m, 2H), 6.623-6.607 (d, IH, J=8.0 Hz), 6.529 (s, IH), 6.395 (s, IH), 6.272 (s, IH), 5.971 (s, IH), 4.069-4.042 (t, 2H =6.5 Hz, 7.0 Hz), 3.783 (s, 2H), 3.750 (s, 3H), 3.718-3.689 (t, 2H, «7=7.0 Hz, 7.5 Hz), 3.610 (s, IH), 3.399 (s, IH), 3.245-3.208 (m, 2H), 3.121 (s, 3H), 3.023-2.779 (m, 6H), 2.569 (s, 3 H), 2.540 (s, I H), 2.370-2.338 (m, 1 H), 2.250 (s, 3H), 1.926-1.869 (m, 2H), 1 .788〜1.682 (m, 3H), 1.570-1 .507 (m, 2H)。 实施例 3:
Figure imgf000021_0001
将 1 ,7-二溴庚烷和邻苯二酰亚胺钾 (1 .0 g, 5.4 mmol)在 N,N-二曱基 甲酰胺 (8 ml)中的混合物加热至 80°C过夜。冷却后, 用乙酸乙酯 (50 mL) 和水 (50 mL)稀释反应混合物。 有机层经无水硫酸钠干燥, 并真空浓缩 通过硅胶柱色谱纯化 (1,2 g, 70%)。
在氮气保护气氛中, 在 0 °C下, 向溶于 Ν,Ν-二曱基甲酰胺 (5 ml)的 二盐酸小檗胺 (205 mg, 0.3 mmol)中加入 NaH (48 mg, 1 .2 mmol) , 搅拌 1 小时后, 加入 2- (7-溴庚基) -异吲哚啉- 1 ,3-二酮(133 mg, 0.45 mmol)。 使 反应液加热至 80 °C过夜。 然后真空蒸发反应混合物, 并通过制备薄层 色谱法纯化以产生白色或淡黄色化合物 (BS-BE-003) (20.2 mg, 7.9 %)。
LC/MS m/z: M+l 852.4 100% (纯度)。
!H画 R (CDCI3) δ: 7.837-7.820 (dd, 2H, J=5.5 Hz, 5.5 Hz),
7.704-7.687 (dd, 2H, J=6.0 Hz, 5.5 Hz), 7.266-7.247 (m, 1 H), 7.099-7.086 (d, IH, J^6.5 Hz), 6.810-6.753 (m, 2H), 6.625〜6.612 (d, 1 H, J=6,5 Hz), 6.53 1 (s, I H), 6.385 (s, I H), 6.275 (s, IH), 5.971 (s, 1H), 4.055-4.028 (t, 2H; •7=6.5 Hz, 7.0 Hz), 3.863-3.848 (d, 2H, J=7.5 Hz), 3.750 (s, 3H),
3.679〜3.650 (t, 2H 5 7=7.0 Hz, 7.5 Hz), 3.607 (s, 3H)5 3,41 l (s, I H),
3.282-3.213 (m, 2H), 3.122 (s, 3H); 3.043-2.783 (m , 7H), 2.588 (s, IH), 2.570 (s, 3H), 2.400 (s, I H), 2.254 (s, 3H), 1 .847-1.704 (m, 2H), 1.685-1.657 (m, 2H), 1.48卜 1.400 (m , 6H)。 实施例 4: 化合物 (BS-BE-004)的合成 s Nal. eCN
v 、Br 向乙腈(35 mL)中加入 1,3-二溴丙烷(1.0 g, 5mmol) , Nal(3.0 g 20mmoi), 反应加热至 80 °C搅拌 3小时, 得到 1,3-二碘丙烷产物粗品。 此产物粗品不经纯化可直接用于下一步反应。
Figure imgf000022_0001
向乙腈 (20 mL)中加入 3,4-吡啶二甲酰亚胺 (740 mg, 5 mmol), 2C03 (828 mg, 6mmol), 随后加入 1,3-二碘丙烷 (5 mmol), 加热至 80。C反应 16小时。反应结束后将反应液过滤,滤液旋干后得到的粗产品经柱纯化 后得到黄色固体产物, N- ( 3-碘丙基) -3,4-吡啶二甲酰亚胺 (200 mg, 13%)u
Figure imgf000022_0002
在氮气保护气氛中, 在 0 °C下, 向溶于 Ν,Ν-二曱基曱酰胺 (5 mL) 的二盐酸小檗胺 (305 mg, 0.5 mmol)中分批加入 NaH (80 mg, 2 mmol),升 至室温搅拌 0.5小时后,加入 N-( 3-碘丙基)-3,4-吡啶二曱酰亚胺(160 mg, 0.5 mmol)。 4吏反应液加热至 80 °C过夜。 反应结束后将反应液倒入冰水 中, 过滤得到的固体粗品产物通过制备性薄层色谱法纯化, 得到棕色化 合物 (BS-BE- 004) (35 mg, 5.0 %)。
MS m/z: M+l 797.4 LC: 2.835 min (84.99%)。
lH NMR (CDC13) δ : 9.123(s, 1H), 8.993-8.972 (d, 1H, =8.4 Hz), 8,194 8.164 (d, IH, J=8.4 Hz), 7.099^7.084 (d, 1H? =6.0 Hz), 6.813-6.756 (m, 4H), 6.628-6.613 (d, IH, 6.0 Hz), 6.542 (s, 1H), 6.391 (s, IH), 6.282 (s, 1H), 5.973 (s, 1H), 4.326-3.985 (m, 8H), 3.853-3.132 (m, 2H), 3.752 (s 3H), 3.702〜3.678 (m, 4H), 3.673 (s, 3H), 3.413(s, 2H), 3.122 (s, 3H), 2.588 (s, 2H), 2.570 (s, 3H), 2.254 (s, 3H), 1.847-1.704 (m, 2H)。 实施例 5: 本发明的小檗胺二酰亚胺化衍生物抗白血病活性测定
(1) 实验材料
白血病细胞株: 人 K562白血病细胞系 (慢性髓系白血病, CML)、 K562/adr (耐药慢性髓系白血病, CML)、 NB4 (急性早幼粒细胞白血病, AML)、 Kasumi- 1 (急性髓系白血病 M2型, AML- M2)、 Jurkat (急性淋巴 细胞白血病, ALL)、 H9 (急性淋巴细胞白血病, ALL;)。
试剂:
小檗胺 (BBM)标准品购自四川什邡普康生化有限公司,
本发明小檗胺衍生物:
12- 0- (( 1 ,3 -二氧代异 I哚淋— 2-基)-曱基) -小檗胺 (化合物 BS- BE- 001 ),
12-C 5- (1,3-二氧代异吲哚啉 -2-基)-戊基) -小檗胺 (化合物 BS- BE-002),
12-0- (7- ( 1 ,3-二氧代异吲哚啉 -2-基)-庚基) - 'j、檗胺 (化合物 BS- BE- 003), 12- 0- (3- (3,4-吡啶二甲酰亚胺基)-丙基) -小檗胺 (化合物 BS- BE- 004)
主要仪器: 细胞培养箱, 酶标仪
(2) 实验方法
取生长良好的白血病细胞 6000个, 接种到 96孔细胞培养板孔内。 培养液为含 10%胎牛血清的 1640细胞培养液。 加入不同浓度的小檗胺 类化合物, 混匀后 , 置于二氧化碳( 5%C02 ) 细胞培养箱 37 °C培养 72 小时。 然后用 MTT法测定活细胞浓度。 在本实验中对照组 (不加化合 物处理) 细胞活力设为 100%, 并计算出化合物作用后细胞活力 (%) 和 72小时白血病细胞半数生长抑制浓度 (72小时 IC5。值)。
(3) 实验结果
实验结果见表 1。 表 i显示本发明的小檗胺衍生物能诱导人慢性髓 系白血病、急性髓系白血病和急性淋巴细胞白血病细胞死亡和抑制这些 白血病细胞生长, 与小檗胺本身比较, 本发明小檗胺衍生物抗白血病细 胞活性明显增强,其中本发明小檗胺衍生物 (BS- BE-003)抗人 K562慢性 髓系白血病活性提高 5倍以上, 抗 K562/adr (耐药慢性髓系白血病)活性 提高 6倍以上。
实施例 5: 本发明小檗胺衍生物抗人多发性骨髓瘤和淋巴瘤细胞活性测 定
(1) 实验材料
骨髓瘤和淋巴瘤细胞株: U266(多发性骨髓瘤), RPMI8226(多发性 骨髓瘤), DOHH2(淋巴瘤)。
试剂: 同实施例 4
主要仪器: 细胞培养箱, 酶标仪。
(2) 实验方法
取生长良好的上述肿瘤细胞 6000个,接种到 96孔细胞培养板孔内。 培养液为含 10%胎牛血清的 1640细胞培养液。 加入不同浓度的小檗胺 类化合物, 混勾后, 置于二氧化碳(5%C02 ) 细胞培养箱 37。C培养 72 小时。 然后用 MTT法测定活细胞浓度。 在本实 ^:中对照组(不加化合 物处理) 细胞活力设为 100%, 并计算出化合物作用后细胞活力 (%) 和 72小时白血病细胞半数生长抑制浓度 (72小时 IC5。值)。
(3) 实验结果
实验结果见表 1。 表 1显示本发明小檗胺的二酰亚胺化衍生物能诱 导人骨髓瘤和淋巴瘤细胞死亡和抑制这些肿瘤细胞生长,与小檗胺本身 比较, 新型小檗胺衍生物抗骨髓瘤和淋巴瘤细胞活性明显增强, 其中本 发明的小檗胺衍生物 (BS- BE- 003)抗 RPMI8226(多发性骨髓瘤)活性提高 近 6倍。
表 1: 小檗胺的二酰亚胺化衍生物对白血病, 淋巴瘤和多发性骨髓 瘤细胞半数生长抑制浓度测定 (72小时, IC5G值)
K562 562a NB4 Kasumi Jurkat H9 RPMI dr -1 8226 BBM 3」 3.53 3 1.3 1.9 5.04 1.17
BS-BE-001 0.98 0.64 1.8 3.36 1.8 2.94 0.01
BS-BE-002 1.02 1.15 2.17
BS-BE-003 0.58 0.59 0.97 1.14 0.75 1.9 0.20
BS-BE-004 10 5.14 16.24 9.16 9.46 733 3.04
实施例 6: 本发明小檗胺的二酰亚胺化衍生物抗人实体瘤作用测定
(1) 实验材料
人实体瘤细胞株: HepG2(人肝细胞肝癌, HCC)、 A549(人肺癌)、 MCF- 7(乳腺癌)、 PANC-1(胰腺癌)、 PC-3(前列腺癌)、 MG63(骨肉瘤)、 AGS (胃癌)、 Huh7(人肝癌细胞)、 Becap37(人乳腺癌细胞)、 Hela (人宫颈 癌细胞)、 RKO(人结肠腺癌细胞)、 SW620(人结肠腺癌细胞)、 SW480(人 结肠癌细胞)、 MGC 803(人胃癌细胞)。
试剂: 同实施例 4
主要仪器: 细胞培养箱, 酶标仪。
(2) 实验方法
取生长良好的人实体瘤细胞 4000个,接种到 96孔细胞培养板孔内。 培养液为含 10%胎牛血清的 DMEM高糖细胞培养液。 置于二氧化碳 ( 5%C02 )细胞培养箱 37 °C培养 24小时, 然后, 加入不同浓度的小檗 胺类化合物, 混匀后, 继续置二氧化碳(5%C02 ) 细胞培养箱 37 °C培 养 72小时。 然后用 MTT法测定活细胞浓度, 并计算出药物作用后细胞 活力(% )。在本实验中对照组(不加化合物处理)细胞活力设为 100%。
(3) 实验结果
实验结果见表 2。 表 2显示本发明的小檗胺衍生物能诱导人实体瘤 细胞死亡和抑制这些肿瘤细胞生长, 与小檗胺本身比较, 本发明小檗胺 衍生物抗实体瘤细胞活性明显增强,其中本发明小檗胺的二酰亚胺化合 物 (3)抗 PANC- 1(胰腺癌)活性提高 6倍以上, 抗 MCF- 7(乳腺癌)活性提 高近 6倍, 抗 HepG2(人肝细胞肝癌, HCC)和 Becap37(人乳腺癌细胞) 活性提高近 5倍, 抗 MG63(骨肉瘤)活性提高近 8倍, 抗 MGC 803(人胃 癌细胞)活性提高近 7倍。 表 2 小檗胺的二酰亚胺化衍生物对人实体瘤细胞半数生长抑制浓 度测定 (72小时, IC5Q值)
Figure imgf000026_0001
表 2 (续):
RKO SW620 SW480 PC-3 AGS MG63 MGC803
BBM 2.34 1.1 5.74 3.7 5.84 9.18 4.96
BS-BE-001 2.93 1.13 4.68 3.95 3.49 4.83 2.38
BS-BE-003 0.72 0.57 1.63 2.7 1.35 1.21 0.74
BS-BE-004 18 4.63 8.58 >16 8.55 16 13.5

Claims

权 利 要 求 书
1.通式 (I)的小檗胺的二酰亚胺化衍生物或其药学上可接受的盐:
Figure imgf000027_0001
其中, 选自 H、 i¾素、 胺基、 硝基、 氰基、 羟基、 巯基、 取代或无取 代的 CrC6烷基、 取代或无取代的 CrC6烷氧基、 取代或无取代的 CrC6 烷硫基、 取代或无取代的 CrC6烷胺基、 取代或无取代的 C3-C7环烷基; A选自直链或支链的、 取代或无取代的、 任选地被选自 0、 N、 S的杂 原子中断的亚烷基- (CH2 ) n -, 其中 n 为 1-15的整数;
W, X, Y和 Z选自取代或无取代的次甲基 CH、 亚甲基 CH2和选自 0、 N和 S的杂原子, 其中 W、 X、 Y和 Z中至少两个为 CH或 CH2;
所述 "取代" 是由选自如下的取代基取代: 素、 胺基、 硝基、 氰基、 羟基和巯基。
2. 式 (I- a)小檗胺的邻苯二甲酰亚胺化衍生物或其药学上可接受 的盐:
Figure imgf000027_0002
式(l-a)
其中, n为 1-15的整数; R^ B与权利要求 1通式 (I)中的定义相同。
3.式 (I-b)小檗胺的芳香杂环二曱酰亚胺化衍生物或其药学上可 接受的盐:
Figure imgf000028_0001
式 (l-b)
其中, 芳香杂环 (Het ) 中含有一个或两个选自 0、 N和 S的杂原子, R, , n和 B与权利要求 1通式 (I)中的定义相同。
4. 根据权利要求 1-3的小檗胺的二酰亚胺化衍生物或其药学上可接 受的盐, 其中 n为 1-10的整数。
5. 根据权利要求 1-3的小檗胺的二酰亚胺化衍生物或其药学上可接 受的盐, 其中 n为 1-8的整数。
6. 根据权利要求 1-3的小檗胺的二酰亚胺化衍生物或其药学上可接 受的盐, 其中 n为 1-7的整数。
7. 根据权利要求 1-6任一项的小檗胺的二酰亚胺化衍生物或其药学 上可接受的盐, 其中 R1为 H、 CrC6烷氧基、 CrC6烷硫基、 CrC6烷基、 CrC7环烷基、 素、 硝基、 任选由一个或两个 CrC6烷基取代的氨基或 氰基。
8. 根据权利要求 1-6任一项的小檗胺的二酰亚胺化衍生物或其药学 上可接受的盐, 其中 为 11、 - ( 6烷氧基、 - C6烷基、 CrC7环烷基 或卤素。
9. 根据权利要求 1-6任一项的小檗胺的二酰亚胺化衍生物或其药学 上可接受的盐,其中 为 H、 CrC3烷氧基、 CrC3烷基或 C5- 6环烷基。
10. 根据权利要求 1-6任一项的小檗胺的二酰亚胺化衍生物或其药学 上可接受的盐, 其中 为11、 曱氧基或曱基。
11. 根据权利要求 1-6任一项的小檗胺的二酰亚胺化衍生物或其药 学上可接受的盐, 其中 为 11。
12. 根据权利要求 1的小檗胺的二酰亚胺化衍生物或其药学上可 接受的盐, 选自下述化合物或其药学上可接受的盐:
Figure imgf000029_0001
13. 一种制备通式 I化合物的方法,
Figure imgf000030_0001
Ifl I!
包括将式 III化合物与式 II化合物反应, 生成式 I化合物, 其中化 合物 I、 II、 ΙΠ中的 A, B, W, X, Y, Z、
Figure imgf000030_0002
n如权利要求 1-11中任一项 所定义, LG为离去基团。
14. 根据权利要求 13的方法, 其中离去基团 LG为卤素原子或磺 酸酯基。
15. 一种药物组合物, 其中包含权利要求 1-12中任一项的小檗胺 的二酰亚胺化衍生物或其药学上可接受的益和任选的药学上可以接 受的赋形剂。
16. 权利要求 1-12中的任一项的小檗胺的二酰亚胺化衍生物或其 药学上可接受的盐在制备抗肿瘤药物中的用途。
17. 一种治疗肿瘤患者的方法,包括给予需要治疗的患者治疗有 效量的根据权利要求 1-12任一项的小檗胺的二酰亚胺化衍生物或其 药学上可接受的盐。
18. 作为抗肿瘤治疗剂的权利要求 1-12任一项的小檗胺的二酰 亚胺化衍生物或其药学上可接受的盐。
19. 根据权利要求 16、 17或 18的用途、 方法或小檗胺的二酰亚 胺化衍生物, 其中, 所述肿瘤选自白血病、 多发性骨髓瘤、 淋巴瘤、 肝癌、 胃癌、 乳腺癌、 胆管细胞癌、 胰腺癌、 肺癌、 大肠癌、 骨肉 瘤、 人宫颈癌、 神经胶质瘤、 鼻咽癌、 喉癌、 食管癌、 中耳肿瘤、 黑色素瘤和前列腺癌。
Figure imgf000031_0001
PCT/CN2011/078905 2010-08-27 2011-08-25 小檗胺的二酰亚胺化衍生物、其制备方法和应用 WO2012025054A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2013525130A JP5701387B2 (ja) 2010-08-27 2011-08-25 ベルバミンのジカルボキシミド誘導体、その調製方法及び使用
CN201180004924.3A CN102712655B (zh) 2010-08-27 2011-08-25 小檗胺的二酰亚胺化衍生物、其制备方法和应用
DK11819429.9T DK2610257T3 (en) 2010-08-27 2011-08-25 DI-IMIDED DERIVATIVE OF BERBAMINE AND METHOD FOR PREPARING AND USING THEREOF
US13/819,111 US9249137B2 (en) 2010-08-27 2011-08-25 Dicarboximide derivatives of berbamine, the preparation and use thereof
EP11819429.9A EP2610257B1 (en) 2010-08-27 2011-08-25 Diimidated derivative of berbamine, and preparation method therefor and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2010076423 2010-08-27
CNPCT/CN2010/076423 2010-08-27

Publications (1)

Publication Number Publication Date
WO2012025054A1 true WO2012025054A1 (zh) 2012-03-01

Family

ID=45722906

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2011/078905 WO2012025054A1 (zh) 2010-08-27 2011-08-25 小檗胺的二酰亚胺化衍生物、其制备方法和应用

Country Status (5)

Country Link
US (1) US9249137B2 (zh)
EP (1) EP2610257B1 (zh)
JP (1) JP5701387B2 (zh)
DK (1) DK2610257T3 (zh)
WO (1) WO2012025054A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103012421A (zh) * 2012-12-06 2013-04-03 刘力 异喹啉类药物及其制备和用途
CN114031623A (zh) * 2021-11-12 2022-02-11 山西医科大学 一种c14位氨基取代粉防己碱衍生物及其制备和应用

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5695200B2 (ja) 2010-09-10 2015-04-01 ハンジョウ ベンシェン ファーマシューティカル シーオー., エルティーディー.Hangzhou Bensheng Pharmaceutical Co., Ltd. 複素環アミノベルバミン誘導体、その調製方法及び使用
JP7016102B2 (ja) * 2018-12-06 2022-02-04 キリンホールディングス株式会社 T細胞又はnk細胞の製造方法、t細胞又はnk細胞の培養用培地、t細胞又はnk細胞の培養方法、未分化t細胞の未分化状態を維持する方法及びt細胞又はnk細胞の増殖促進剤

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0499723A (ja) * 1990-08-17 1992-03-31 Kaken Shiyouyaku Kk ウイルス・ゲノム不活化剤
CN101273989A (zh) 2008-04-09 2008-10-01 浙江大学 一类小檗胺衍生物及其盐的应用
US20100298369A1 (en) * 2009-05-19 2010-11-25 David Horne Berbamine derivatives

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH02111777A (ja) * 1988-10-20 1990-04-24 Kaken Shiyouyaku Kk ビスベンジルイソキノリン誘導体
JPH032183A (ja) * 1989-05-31 1991-01-08 Kaken Shiyouyaku Kk ビスベンジルイソキノリン誘導体
ZA923816B (en) * 1991-05-30 1993-11-25 Bristol Myers Squibb Co Prepartion of 6-0-alkylesamicin a derivatives
JPH06211661A (ja) * 1993-01-12 1994-08-02 Kaken Shiyouyaku Kk 抗炎症剤
JPH11180873A (ja) * 1997-12-22 1999-07-06 Kaken Shoyaku Kk NF−κB活性阻害剤
JP4596792B2 (ja) * 2004-02-24 2010-12-15 あすか製薬株式会社 5−ht1a作動作用と5−ht3拮抗作用を併有する薬剤
CN1810247A (zh) * 2005-01-28 2006-08-02 中国医学科学院血液学研究所 小檗胺衍生物ebb在抑制人体肿瘤细胞侵袭转移的药物应用

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0499723A (ja) * 1990-08-17 1992-03-31 Kaken Shiyouyaku Kk ウイルス・ゲノム不活化剤
CN101273989A (zh) 2008-04-09 2008-10-01 浙江大学 一类小檗胺衍生物及其盐的应用
US20100298369A1 (en) * 2009-05-19 2010-11-25 David Horne Berbamine derivatives

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"BURGER'S MEDICINAL CHEMISTRY AND DRUG DISCOVERY", 1995, pages: 172 - 178,949-
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
"MCGRAW-HILL DICTIONARY OF CHEMICAL TERMS", 1984, MCGRAW-HILL BOOK COMPANY
"REMINGTON'S PHARMACEUTICAL SCIENCES", 1995, MACK PUBLISHING COMPANY
DONG ZHIYU ET AL.: "The experimental study of berbamine on inducing apoptosis of human leukemia Jurkat cells", CHINESE TUMOR, vol. 16, no. 9, 2007, pages 722
DUAN JIANGYAN ET AL.: "The influence of berbamine compounds on calmodulin protein level within melanoma cells", CHINESE HERBAL MEDICINE, vol. 33, no. 1, 2002, pages 59
DUAN JIANGYAN ET AL.: "The preliminary exploration of [O-(4-ethoxy)-butyl]-berbamine for induction of lung cancer cell apoptosis", JOURNAL OF SHANXI NORMAL UNIVERSITY (NATURAL SCIENCE EDITION, vol. 15, no. 4, 2001, pages 55
ELIEL, E.; WILEN, S.: "STEREOCHEMISTRY OF ORGANIC COMPOUNDS", 1994, JOHN WILEY & SONS
GE MINGZHU ET AL.: "The experimental study of immune protection action of berbamine on irradiated mice", JOURNAL OF IMMUNOLOGY, vol. 14, no. 4, 1998, pages 238
L. FIESER; M. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
LIN CHUANRONG ET AL.: "The clinical observations on the treatment of chemotherapy-induced leukopenia with Shengbei'an (berbamine", PREPARED CHINESE MEDICINE, vol. 16, no. 7, 1994, pages 29
LIU XIN ET AL.: "The immune regulation action of berbamine on BALB/C mice", JOURNAL OF CHINA MEDICAL UNIVERSITY, vol. 25, no. 3, 1996, pages 229
LUO CHONGNIAN ET AL.: "The inhibition of berbamine on mice splenocytes cytotoxic T lymphocyte activity", CHINESE JOURNAL OF PHARMACOLOGY AND TOXICOLOGY, vol. 9, no. 2, 1995, pages 159 - 160
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
See also references of EP2610257A4 *
SUN PENG ET AL.: "The effect of berbamine inducing apoptosis of prostate cancer PC-3 cells and the mechanism", CHINESE JOURNAL OF EXPERIMENTAL SURGERY, vol. 24, no. 8, 2007, pages 957
T. W. GREENE; P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS
T.W. GREENE; PG.M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS
WU DONG ET AL.: "The experimental study of the actions of berbamine on K562 cells in vitro and in vivo", JOURNAL OF CHINESE EXPERIMENTAL HEMATOLOGY, vol. 13, no. 3, 2005, pages 373
ZHANG JINHONG ET AL.: "The influence of the calmodulin antagonist O-Dansyl berbamine on phosphodiesterase and pulmonary cell proliferation", ACTA SCIENTIARUM NATURALIUM UNIVERSITY NANKAIENSIS, vol. 34, no. 3, 2001, pages 64
ZHANG JINHONG ET AL.: "The influence of the structures of the berbamine and its derivatives on cervical carcinoma (CHeLa) cells proliferation", ACTA SCIENTIARUM NATURALIUM UNIVERSITY NANKAIENSIS, vol. 29, no. 2, 1996, pages 89
ZHANG JINHONG ET AL.: "The preliminary exploration of the in vivo antitumor effect of the berbamine derivatives (EBB", CHINESE HERBAL MEDICINE, vol. 29, no. 4, 1998, pages 243
ZHANG JINHONG: "The influence of the berbamine and its derivatives on malignant melanoma cell proliferation", CHINESE HERBAL MEDICINE, vol. 28, no. 8, 1997, pages 483

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103012421A (zh) * 2012-12-06 2013-04-03 刘力 异喹啉类药物及其制备和用途
CN103012421B (zh) * 2012-12-06 2016-01-20 刘力 异喹啉类药物及其制备和用途
CN114031623A (zh) * 2021-11-12 2022-02-11 山西医科大学 一种c14位氨基取代粉防己碱衍生物及其制备和应用

Also Published As

Publication number Publication date
EP2610257A1 (en) 2013-07-03
US20130158068A1 (en) 2013-06-20
DK2610257T3 (en) 2016-02-01
EP2610257A4 (en) 2014-04-16
JP2013536204A (ja) 2013-09-19
JP5701387B2 (ja) 2015-04-15
EP2610257B1 (en) 2015-10-28
US9249137B2 (en) 2016-02-02

Similar Documents

Publication Publication Date Title
US20210163464A1 (en) Pyridine compound
JP5695200B2 (ja) 複素環アミノベルバミン誘導体、その調製方法及び使用
WO2022017533A1 (zh) 用作cdk7激酶抑制剂的化合物及其应用
TWI481604B (zh) Novel 5-fluorouracil derivatives
TWI453201B (zh) 可作為Hsp90之抑制劑之異吲哚衍生物
WO2019042444A1 (zh) 一类抑制并降解酪氨酸蛋白激酶alk的化合物
WO2020001420A1 (zh) 一类细胞坏死抑制剂及其制备方法和用途
JP6298768B2 (ja) 7−置換ハンファンギチンb誘導体、その調製方法及び使用
WO2011020288A1 (zh) 取代酰肼类化合物及其应用
WO2012025054A1 (zh) 小檗胺的二酰亚胺化衍生物、其制备方法和应用
US20190100495A1 (en) Substituted tetrahydroisoquinoline ethylbenzamide anti-cancer agents
EP2897952A1 (en) 4-(indolyl or benzimidazolyl)amino-2-(2-(indol-3-yl)ethyl)aminopyrimidines useful for the treatment of cancer
CN112125914B (zh) 5-取代的小檗胺衍生物,其制备方法和应用
WO2013185613A1 (zh) 重楼皂苷i的酰化衍生物、及其制备方法和应用
WO2013023620A1 (zh) 高三尖杉酯碱的胺化衍生物、及其制备方法和应用
WO2013079017A1 (zh) 2-位烷基或芳香基取代的丹参酮衍生物、及其制备方法和应用
WO2014000686A1 (zh) 嘧啶并苯并氮杂卓类化合物及其作为抗肿瘤药物的应用
CN102712655A (zh) 小檗胺的二酰亚胺化衍生物、其制备方法和应用
CN113480520B (zh) 一种异喹啉-1,3(2h,4h)-二酮衍生物及其应用
KR20140048433A (ko) 신규한 스트렙토클로린 유도체 및 이를 유효성분으로 하는 암 질환의 예방 및 치료용 약학조성물
US10471154B2 (en) Spirocyclic indolone polyethylene glycol carbonate compound, composition, preparation method and use thereof
TWI344462B (en) Indole derivatives having an apoptosis-inducing effect
TW201741305A (zh) 4,9-二側氧基-4,9-二氫萘并[2,3-b]呋喃-3-羧醯胺衍生物及其用於治療增生性疾病與感染性疾病之用途
US20130109724A1 (en) N-aminopyrrolylmethyltetrahydropyridines as anti-cancer agents
WO2013023622A1 (zh) 高三尖杉酯碱的酰化衍生物、及其制备方法和应用

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180004924.3

Country of ref document: CN

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

Ref document number: 11819429

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013525130

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13819111

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011819429

Country of ref document: EP