WO2017070878A1 - C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 - Google Patents
C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 Download PDFInfo
- Publication number
- WO2017070878A1 WO2017070878A1 PCT/CN2015/093161 CN2015093161W WO2017070878A1 WO 2017070878 A1 WO2017070878 A1 WO 2017070878A1 CN 2015093161 W CN2015093161 W CN 2015093161W WO 2017070878 A1 WO2017070878 A1 WO 2017070878A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- substituted
- amino acid
- unsubstituted
- triptolide
- group
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
- 0 CC(C)[C@]1([C@]2OC(C(*)N*)=O)O[C@@]1[C@@]1O[C@]1([C@@]1[C@](C)CC3)[C@]22O[C@]2C[C@@]1C(CO1)=C3C1=O Chemical compound CC(C)[C@]1([C@]2OC(C(*)N*)=O)O[C@@]1[C@@]1O[C@]1([C@@]1[C@](C)CC3)[C@]22O[C@]2C[C@@]1C(CO1)=C3C1=O 0.000 description 6
- AANDWUPHUWBAEV-ORGQIQBHSA-N CC(C)[C@]1([C@H]2O)O[C@H]1[C@@H]1O[C@]1([C@@H]1[C@H](C)CC3)[C@]22O[C@H]2C[C@H]1C(CO1)=C3C1=O Chemical compound CC(C)[C@]1([C@H]2O)O[C@H]1[C@@H]1O[C@]1([C@@H]1[C@H](C)CC3)[C@]22O[C@H]2C[C@H]1C(CO1)=C3C1=O AANDWUPHUWBAEV-ORGQIQBHSA-N 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/34—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
- A61K31/343—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/365—Lactones
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/58—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
- A61K31/585—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin containing lactone rings, e.g. oxandrolone, bufalin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D303/00—Compounds containing three-membered rings having one oxygen atom as the only ring hetero atom
- C07D303/02—Compounds containing oxirane rings
- C07D303/04—Compounds containing oxirane rings containing only hydrogen and carbon atoms in addition to the ring oxygen atoms
- C07D303/06—Compounds containing oxirane rings containing only hydrogen and carbon atoms in addition to the ring oxygen atoms in which the oxirane rings are condensed with a carbocyclic ring system having three or more relevant rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D307/00—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
- C07D307/77—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D493/00—Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
- C07D493/22—Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains four or more hetero rings
Definitions
- the invention belongs to the field of natural medicine and medicinal chemistry, and particularly relates to a novel triptolide derivative, in particular to a C14-position hydroxyl esterified triptolide amino acid derivative, a method and a use for preparing the same.
- Triptolide also known as triptolide, is a kind of three epoxy groups and one ⁇ , ⁇ unsaturated five-membered one isolated from Tripterygium wilfordii.
- a rosin-type diterpene lactone compound having a unique configuration of a lactone ring structure, having a molecular formula of C 20 H 24 O 6 having a molecular weight of 360.4, and a structural formula represented by the following formula.
- triptolide has significant biological activities such as anti-tumor, anti-inflammatory, immunosuppressive, and anti-male fertility. Triptolide can not only prolong the survival time of mouse leukemia L615, but also has obvious therapeutic effect on human leukemia. The complete remission rate is 40% (18/45) [Xia Zhilin et al. Pharmacological and clinical research of triptolide. China Journal of Pharmacology 1992, (6) 427-431]. Clinically used to treat psoriasis, rheumatoid arthritis, leukemia, kidney disease, etc.
- triptolide has high toxicity, its therapeutic window is narrow, and it has great side effects on the digestive system, genitourinary system and blood system. Therefore, clinical development research has been Certain restrictions. Due to the unique chemical structure and biological activity of triptolide, it has attracted great interest from chemical and pharmacologists in various countries. In order to reveal the mechanism of action and structure-activity relationship of triptolide, and to find high-efficiency and low-toxic derivatives, several research groups conducted a systematic study on triptolide and its analogues, and synthesized a series of triptolide derivatives. [Zhang Fan et al. Progress in structural modification of triptolide. Journal of Pharmaceutical Sciences.
- Minnelide is a water-soluble triptolide prodrug synthesized by scientists at the University of Minnesota in the United States. It can be converted to triptolide in vivo and in vitro. In some animal models, Minnelide showed good anticancer effects and less toxic effects. For example, in a nude mouse model of pancreatic cancer, Minnelide prolongs the survival of mice by reducing the volume of the tumor and reducing the spread of the tumor, and Minnelide does not exhibit significant toxic effects at an effective dose that causes tumor regression.
- triptolide is entering Phase I clinical trials [Chugh R, et al. A preclinical evaluation of Minnelide as a therapeutic agent against pancreatic cancer. Sci Transl Med. 2012; 4 (156): 156ra 139.].
- no triptolide or its derivatives have entered Phase II clinical trials. Therefore, researchers still need to further study and explore the safe application of triptolide.
- Clinically relevant methods as well as new high-efficiency, low-toxic triptolide derivatives.
- the pharmacokinetic characteristics of triptolide oral and intravenous injections suggest that triptolide has a short elimination half-life, undergoes rapid absorption, distribution, metabolism and elimination in vivo, and plasma concentration and drug concentration of various tissues change with time.
- One of the objects of the present invention is to provide a novel C14-position hydroxyesterified triptolide amino acid derivative of the formula (I), or a pharmaceutically acceptable adduct or complex thereof, characterized by a single amino acid esterification, a salt, a decomposition product, and a metabolite, and a novel triptolide-esterified triptolide amino acid derivative characterized by dipeptide esterification of the novel C14-position, or a pharmaceutically acceptable adduct thereof, or a compound thereof Substances, salts, decomposition products and metabolites:
- R 1 is selected from a substituted or unsubstituted C 2 -C 20 alkyl group, a substituted or unsubstituted C 2 -C 20 olefin group, a substituted or unsubstituted C 4 -C 20 conjugated olefin group, a substituted or unsubstituted C 3 -C 7 cycloalkyl or cycloalkenyl group, a substituted or unsubstituted aromatic hydrocarbon group, a substituted or unsubstituted heterocyclic group and an aromatic heterocyclic group, a substituted or unsubstituted amino acid a side chain alkyl group; the amino acid may be racemic or optically pure (left-handed or right-handed); the substituent is selected from the group consisting of halogen, amine group, C 1 -C 6 substituted amine group, nitro group, cyano group, hydroxyl group, C 1 -C 6 alkoxy, fluorenyl, C
- R 2 and R 3 are selected from H, substituted or unsubstituted C 1 -C 20 alkyl, substituted or unsubstituted C 2 -C 20 alkene, substituted or unsubstituted C 4 -C 20 Co-decene olefin group, substituted or unsubstituted C 3 -C 7 cycloalkyl or cycloalkyl group, substituted or unsubstituted aromatic hydrocarbon group, substituted or unsubstituted heterocyclic group and aromatic heterocyclic group, substituted Or an unsubstituted amino acid side chain alkyl group, the amino acid may be racemic or optically pure (left-handed or right-handed); the substituent is selected from the group consisting of halogen, amine group, C 1 -C 6 substituted amine group, nitro group, Cyano, hydroxy, C 1 -C 6 alkoxy, fluorenyl, C 1 -C 6 alkylthio; R 2 and R
- R 4 is selected from substituted or unsubstituted C 2 -C 20 alkyl, substituted or unsubstituted C 2 -C 20 alkene, substituted or unsubstituted C 4 -C 20 conjugated alkene group, Substituted or unsubstituted C 3 -C 7 cycloalkyl or cycloalkenyl, substituted or unsubstituted arene, substituted or unsubstituted heterocyclic and aromatic heterocyclic, substituted or unsubstituted amino a side chain alkyl group, the amino acid may be racemic or optically pure (left-handed or right-handed); the substituent is selected from the group consisting of halogen, amine group, C 1 -C 6 substituted amine group, nitro group, cyano group, hydroxyl group, C 1 -C 6 alkoxy, fluorenyl, C 1 -C 6 alkylthio;
- R 5 and R 6 are selected from H, substituted or unsubstituted C 1 -C 20 alkyl, substituted or unsubstituted C 2 -C 20 alkene, substituted or unsubstituted C 4 -C 20 Co-decene olefin group, substituted or unsubstituted C 3 -C 7 cycloalkyl or cycloalkyl group, substituted or unsubstituted aromatic hydrocarbon group, substituted or unsubstituted heterocyclic group and aromatic heterocyclic group, substituted Or an unsubstituted amino acid side chain alkyl group, the amino acid may be racemic or optically pure and include levorotatory and dextrorotatory; the substituent is selected from the group consisting of halogen, amine group, C 1 -C 6 substituted amine group, Nitro, cyano, hydroxy, C 1 -C 6 alkoxy, fluorenyl, C 1 -C 6 alkylthio.
- a second object of the present invention is to provide a process for preparing a single amino acid esterification of the present invention, wherein (I) and a dipeptide esterified a compound of the formula (II) C14-position hydroxylated triptolide amino acid derivative, which follows the route below:
- the single amino acid esterification of the C14-position hydroxyl esterified triptolide amino acid derivative of the formula (I) can be obtained from a triptolide with the corresponding amino acid R 2 R 3 NCHR 1 CO 2 H in a condensing agent or a catalyst. In the presence of a suitable solvent, condensed esterification at a suitable temperature; or it may be condensed and esterified by triptolide and the corresponding amino acid acyl chloride R 2 R 3 NCHR 1 COCl in the presence of an alkaline reagent.
- amino acid R 2 R 3 NCHR 1 CO 2 H is activated to form an active ester intermediate, and then reacted with triptolide to form a C14-hydroxyesterified triptolide amino acid derivative (I);
- the amino acid R 2 R 3 NCHR 1 CO 2 H is activated to form a mixed anhydride intermediate, which is then reacted with triptolide to form a C14-hydroxyesterified triptolide amino acid derivative (I).
- R 1 , R 2 , R 3 , amino acid R 2 R 3 NCHR 1 CO 2 H, R 1 , R 2 , R 3 , amino acid acid chloride R 2 R 3 NCHR 1 COCl, R 1 , R 2 , R 3 , the active ester intermediates R 1 , R 2 , R 3 , and the mixed anhydride intermediates R 1 , R 2 , R 3 are the same as defined above in the formula (I).
- the present invention also provides a method for preparing a triptolide amino acid derivative esterified with a C14-position hydroxydipeptide of the formula (II) of the present invention: a triptolide amino acid derivative esterified with a C14-position hydroxydipeptide (II)
- the C14-position hydroxyl-esterified triptolide amino acid derivative (I) can be obtained by the above method, and in the amino acid R 2 R 3 NCHR 1 CO 2 H, R 2 or R 3 is a protecting group.
- the protecting groups for these amino groups are known to those skilled in the art and can be readily found in literature sources, such as Greene and Wuts, "Protective Groups in Organic Synthesis” Third Edition, John Wiley & Sons Press, New York, NY, 1999. All of them are incorporated herein by reference.
- the protecting group is then removed to give a derivative of the formula Ia, which is then condensed with the corresponding amino acid R 5 R 6 NCHR 4 CO 2 H in the presence of a condensing agent or catalyst in a suitable solvent at an appropriate temperature.
- COCl is formed by condensation amidation in the presence of an alkaline reagent; or the amino acid R 5 R 6 NCHR 4 CO 2 H is activated to form an active ester intermediate, and then the triptolide amino acid derivative esterified with a C14-position hydroxyl group ( Ia) reaction formation; the amino acid R 5 R 6 NCHR 4 CO 2 H can also be activated to form a mixed anhydride intermediate, which is then reacted with a C14-hydroxy esterified triptolide amino acid derivative (Ia).
- R 5 or R 6 is a protecting group, the compound can be further deprotected to give compound IIa.
- R 1 , R 2 , R 3 , R 4 , R 5 and R 6 amino acid R 2 R 3 NCHR 1 CO 2 H, R 1 , R 2 , R 3 , amino acid acid chloride R 2 R 3 NCHR 1 in RCO, R 1 , R 2 , R 3 , active ester intermediates R 1 , R 2 , R 3 , and mixed anhydride intermediates R 1 , R 2 , R 3 are as defined above in formula (II) .
- R 5 R 6 NCHR 4 CO 2 H in R 4 , R 5 , R 6 , amino acid acyl chloride R 5 R 6 NCHR 4 COCl in R 4 , R 5 , R 6 , active ester intermediate R 4 , R 5 , R 6 , and the mixed anhydride intermediates R 4 , R 5 , R 6 are the same as defined above in formula (II).
- a third object of the present invention is to provide a pharmaceutical composition comprising at least one compound of the present invention, and optionally a pharmaceutically acceptable excipient selected from the group consisting of Formula (I) A triptolide amino acid derivative esterified with a C14-position hydroxy single amino acid ester and a triptolide amino acid derivative esterified with a C14-position hydroxydipeptide of the formula (II).
- a fourth object of the present invention is to provide a use of a compound of the present invention or a pharmaceutical composition comprising the same for the preparation of a medicament, particularly an antitumor medicament. Accordingly, 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, human cervical cancer, glioma, Nasopharyngeal cancer, laryngeal cancer, esophageal cancer, middle ear tumor, prostate cancer, etc.
- the compound is selected from the group consisting of a triptolide amino acid derivative of a C14-position hydroxy single amino acid of the formula (I) and a triptolide amino acid derivative esterified with a C14-position hydroxydipeptide of the formula (II). .
- a fifth object of the present invention is to provide a use of a compound of the present invention or a pharmaceutical composition comprising the same for the preparation of a medicament for treating an autoimmune disease.
- the autoimmune disease is particularly selected from autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, psoriasis, and kidney disease, which are associated with abnormal immune function of cells such as T cells or B cells.
- the compound is selected from the group consisting of a triptolide amino acid derivative of a C14-position hydroxy single amino acid of the formula (I) and a triptolide amino acid derivative esterified with a C14-position hydroxydipeptide of the formula (II). .
- the present invention relates to a novel amino acid esterification of the general formula (I) and a dipeptide esterification of the novel C14-position hydroxyesterified triptolide Amino acid derivatives or pharmaceutically acceptable adducts, complexes, salts, decomposition products and metabolites thereof:
- R 1 is selected from substituted or unsubstituted C 2 -C 20 alkyl, substituted or unsubstituted C 2 -C 20 alkene, substituted or unsubstituted C 4 -C 20 conjugated alkene group, substituted or not Substituted C 3 -C 7 cycloalkyl or cycloalkenyl, substituted or unsubstituted arene, substituted or unsubstituted heterocyclic and aromatic heterocyclic, substituted or unsubstituted amino acid side chain alkyl, amino acid Is racemic or optically pure (left-handed or right-handed); substituents are selected from halogen, amine, C 1 -C 6 substituted amine, nitro, cyano, hydroxy, C 1 -C 6 alkane Oxyl, fluorenyl, C 1 -C 6 alkylthio.
- R 2 , R 3 , R 4 , R 5 and R 6 are selected from H, substituted or unsubstituted C 1 -C 20 alkyl, substituted or unsubstituted C 2 -C 20 alkene, substituted or unsubstituted a C 4 -C 20 conjugated alkene group, a substituted or unsubstituted C 3 -C 7 cycloalkyl or cycloalkenyl group, a substituted or unsubstituted aromatic hydrocarbon group, a substituted or unsubstituted heterocyclic group and an aromatic heterocyclic group a substituted or unsubstituted amino acid side chain alkyl group, the amino acid may be racemic or optically pure (left-handed or right-handed); the substituent is selected from the group consisting of halogen, amine group, C 1 -C 6 substituted amine group, Nitro, cyano, hydroxy, C 1 -C 6 alkoxy, fluorenyl, C 1
- the invention relates to compounds of Formula I or Formula II wherein R 1 , R 4 are selected from substituted or unsubstituted C 2 -C 20 alkyl. R 1 and R 4 may be the same or different.
- the invention relates to compounds of Formula I or Formula II wherein R 2 , R 3 , R 5 and R 6 are selected from H, substituted or unsubstituted C 1 -C 20 alkyl. R 2 , R 3 , R 5 and R 6 may be the same or different.
- the invention relates to compounds of Formula I or Formula II wherein R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are selected from substituted or unsubstituted C 2 -C 20 An alkene-based, substituted or unsubstituted C 4 -C 20 conjugated alkene group.
- R 1 , R 2 , R 3 R 4 , R 5 and R 6 may be the same or different.
- the invention relates to compounds of Formula I or Formula II wherein R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are selected from substituted or unsubstituted C 3 -C 7 A cycloalkyl or cycloalkenyl group.
- R 1 , R 2 , R 3 , R 4 , R 5 and R 6 may be the same or different.
- the invention relates to compounds of Formula I or Formula II wherein R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are selected from substituted or unsubstituted arenes.
- R 1 , R 2 , R 3 , R 4 , R 5 and R 6 may be the same or different.
- the invention relates to compounds of Formula I or Formula II wherein R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are selected from substituted or unsubstituted heterocyclic or aromatic Heterocyclic group.
- R 1 , R 2 , R 3 , R 4 , R 5 and R 6 may be the same or different.
- the invention relates to compounds of Formula I or Formula II wherein R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are selected from substituted or unsubstituted amino acid side chain alkyl groups .
- R 1 , R 2 , R 3 , R 4 , R 5 and R 6 may be the same or different.
- triptolide amino acid derivative of the present invention is shown below. These examples are only intended to further illustrate the invention and are not intended to limit the scope of the invention.
- the invention particularly preferably exemplifies the following compounds of formula (I):
- the invention particularly preferably comprises the following compound of formula (II):
- the present invention relates to compounds of the formula (I) and formula (II) of the invention in the form of their salts, solvates, hydrates, adducts, complexes, polymorphs or prodrugs.
- C 1 -C 20 alkyl refers to a straight or branched, substituted or unsubstituted alkane group containing from 1 to 20 carbon atoms.
- Examples of C 1 -C 20 alkane groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl and n-icosane. base.
- C 2 -C 20 alkyl refers to a straight or branched, substituted or unsubstituted alkane group containing from 2 to 20 carbon atoms.
- Examples of C 2 -C 20 alkane groups include, but are not limited to, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, n-pentyl, n-hexyl and n-icosyl.
- C 2 -C 20 olefinic group means a straight or branched, substituted or unsubstituted alkene group having 2 to 20 carbon atoms.
- Examples of C 2 -C 20 alkene groups include, but are not limited to, vinyl, allyl, and eicosyl groups.
- C 4 -C 20 conjugated alkene group means a straight or branched, substituted or unsubstituted conjugated olefin group having 4 to 20 carbon atoms.
- Examples of the C 4 -C 20 conjugated alkene group include, but are not limited to, a copolybutadienyl group, a (retinyl) group, and a (9Z)-octadecyl (yl)-9-olefin group.
- C 3 -C 7 cycloalkyl or cycloalkenyl refers to a hydrocarbyl group of a 3-7 membered monocyclic ring system having a saturated or unsaturated ring, and the C 3 -C 7 cycloalkyl group may be a cyclopropyl group or a cyclobutyl group.
- Base cyclopentyl, cyclohexyl, cycloheptyl, cyclopropenyl and cyclohexenyl.
- aromatic hydrocarbon group means an aromatic hydrocarbon group free of hetero atoms, including an aromatic hydrocarbon group, an aromatic hydrocarbon alkyl group and an alkyl aromatic hydrocarbon group.
- heterocyclic aromatic hydrocarbon group means an aromatic hydrocarbon group containing a hetero atom, and includes a heterocyclic aromatic hydrocarbon group, a heterocyclic arenealkyl group, and an alkylheteroaryl arene group. Heteroatoms refer to nitrogen, oxygen and sulfur.
- the heterocyclic aromatic hydrocarbon group may contain one or several hetero atoms.
- amino acid refers to both natural and unnatural amino acids.
- halogen means fluoro, chloro, bromo or iodo.
- C 1 -C 6 substituted amine refers to -NC 1 -C 6 alkyl and -NC 3 -C 6 cycloalkyl.
- C 1 -C 6 alkoxy refers to -OC 1 -C 6 alkyl and -OC 3 -C 6 cycloalkyl.
- C 1 -C 6 alkylthio refers to -SC 1 -C 6 alkyl and -SC 3 -C 6 cycloalkyl.
- pharmaceutically acceptable adducts and complexes of the compounds of formula (I) and formula (II) means that the compounds of the invention further interact with other small molecules or biomacromolecules by non-chemical or non-covalent molecules. The product of the combination.
- the term "pharmaceutically acceptable salts of the compounds of formula (I) and formula (II)” refers to organic acid salts of the compounds of the invention with pharmaceutically acceptable anionic organic acids, which Organic acid salts include, but are not limited to, tosylate, mesylate, malate, acetate, citrate, malonate, tartrate, succinate, benzoate, ascorbate, milk Acid salts, alpha-ketoglutarate and alpha-glycerophosphate; also suitable inorganic salts, including but not limited to hydrochlorides, sulfates, nitrates, hydrogencarbonates and carbonates, phosphates, Hydrobromide, hydroiodide, and the like.
- 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 that 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 bioabsorbability and availability).
- Differences in stability can result in chemical reactivity (eg, differential oxidation such that when formulated from one polymorph, the formulation fades faster than when formed from another polymorph) or mechanical properties (eg, when stored) Changes in the kinetically favorable polymorphic tablet granules converted to thermodynamically more stable polymorphs) or both (for example, tablets of one polymorph are 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, the shape or size distribution of its particles.
- hydrate refers to a compound of the invention or a salt thereof, further comprising a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
- prodrug refers to a derivative that 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, for example, 1 Burger's Medicinal Chemistry and Drug Discovery (1995) 172-178, 949-982 (Manfred E. Wolff, ed. 5) and J. Rautio's Prodrugs and Targeted Delivery (2011) 31- 60 (Wiley-VCH, Methods and Principles in Medicinal Chemistry, Vol. 47) and those described in G. Thomas's Fundamentals of Medicinal Chemistry (2003) 195-200 (Wiley).
- the triptolide derivative of the compound of the present invention has the general formula I (having ten chiral centers, nine chiral centers originating from triptolide, one chiral center derived from amino acids); and formula II (having Eleven chiral centers, nine chiral centers are derived from triptolide, and two chiral centers are derived from the stereochemical structure shown by the structural formula of amino acids.
- the definitions and conventions of stereochemistry used herein generally follow the McGraw-Hill Dictionary of Chemical Terms (SP Parker, Ed., McGraw-Hill Book Company, New York, 1984); 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 encompasses any treatment for a patient's condition, including: (a) preventing a disease or condition in a disease or condition that has not yet been diagnosed; (b) inhibiting the symptoms of the disease, ie preventing Its development; or (c) to alleviate the disease It causes disease or symptom deterioration.
- the compounds of the present invention can be prepared according to conventional organic chemical synthesis methods.
- the general preparation method of the compound of the general formula (I) of the present invention is as follows:
- the single amino acid esterification of the C14-position hydroxyl esterified triptolide amino acid derivative of the formula (I) can be isolated from the naturally extracted triptolide (TPL) and the corresponding amino acid R 2 R 3 NCHR 1 CO 2 H In the presence of a condensing agent or a catalyst, it is condensed and esterified at a suitable temperature in a suitable solvent; it can also be obtained from an alkaline reagent by triptolide and the corresponding amino acid acyl chloride R 2 R 3 NCHR 1 COCl.
- the amino acid R 2 R 3 NCHR 1 CO 2 H may also be activated to form a mixed anhydride intermediate, which is then reacted with triptolide to form a C14-hydroxyesterified triptolide amino acid derivative (I).
- R 1 , R 2 , R 3 , amino acid R 2 R 3 NCHR 1 CO 2 H, R 1 , R 2 , R 3 , amino acid acid chloride R 2 R 3 NCHR 1 COCl, R 1 , R 2 , R 3 , the active ester intermediates R 1 , R 2 , R 3 , and the mixed anhydride intermediate R 1 , R 2 , R 3 are the same as defined above in the formula (I).
- the triptolide amino acid derivative (II) esterified with a C14-position hydroxydipeptide can be obtained by the above method, and the amino acid derivative (I) of the C14-position hydroxyl esterified triptolide is used, and the amino acid R 2 R 3 is used.
- R 2 or R 3 is a protecting group.
- the protecting groups for these amino groups are known to those skilled in the art and can be readily found in literature sources, such as Greene and Wuts, "Protective Groups in Organic Synthesis” Third Edition, John Wiley & Sons Press, New York, NY, 1999. All of them are incorporated herein by reference.
- the protecting group is then removed to give a derivative of the formula Ia, which is then condensed with the corresponding amino acid R 5 R 6 NCHR 4 CO 2 H in the presence of a condensing agent or catalyst in a suitable solvent at an appropriate temperature.
- Amidated to a C14-position hydroxydipeptide esterified triptolide amino acid derivative (II); a derivative of the formula Ia and a corresponding amino acid acyl chloride R 5 R 6 NCHR 4 may also be obtained by removal of a protecting group.
- COCl is formed by condensation amidation in the presence of an alkaline reagent; or the amino acid R 5 R 6 NCHR 4 CO 2 H is activated to form an active ester intermediate, and then the triptolide amino acid derivative esterified with a C14-position hydroxyl group ( Ia) reaction formation; the amino acid R 5 R 6 NCHR 4 CO 2 H can also be activated to form a mixed anhydride intermediate, which is then reacted with a C14-hydroxy esterified triptolide amino acid derivative (Ia).
- R 5 and R 6 are a protecting group, the compound can be further deprotected to give compound IIa.
- R 1 , R 2 , R 3 , R 4 , R 5 and R 6 amino acid R 2 R 3 NCHR 1 CO 2 H, R 1 , R 2 , R 3 , amino acid acid chloride R 2 R 3 NCHR 1 in RCO, R 1 , R 2 , R 3 , active ester intermediates R 1 , R 2 , R 3 , and mixed anhydride intermediates R 1 , R 2 , R 3 are as defined above in formula (II) .
- the above reaction is generally carried out in the presence of an alkali or an alkaline reagent.
- the base here may be, but not limited to, an organic base.
- Solvents used include, but are not limited to, aprotic polar solvents such as dichloromethane (DCM), dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMA), N -methylpyrrole (NMP) or tetrahydrofuran (THF), and the like.
- aprotic polar solvents such as dichloromethane (DCM), dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMA), N -methylpyrrole (NMP) or tetrahydrofuran (THF), and the like.
- the reaction temperature of the above reaction is generally from 0 ° C to 50 ° C. It generally varies depending on the starting materials used and the base used.
- the starting material for the preparation reaction is triptolide (TPL).
- TPL triptolide
- Protecting groups are those which, once attached to an active moiety (e.g., a hydroxyl or amino group), prevent these moieties from being interfered by subsequent reactions and which can be removed by conventional methods after the reaction.
- hydroxy protecting groups include, but are not limited to, alkyl, benzyl, allyl, triphenyl Methyl (ie, triphenylmethyl), acyl (eg, benzoyl, acetyl or HOOC-X"-CO-, X" is an alkylene, alkenylene, cycloalkylene or aroma a silyl group (for example, trimethylsilyl, triethylsilyl and tert-butyldimethylsilyl), an alkoxycarbonyl group, an aminocarbonyl group (for example, dimethylaminocarbonyl, Methyl ethylaminocarbonyl and phenylaminocarbonyl), alkoxymethyl, benzyloxymethyl and alky
- amino protecting groups include, but are not limited to, alkoxycarbonyl, alkanoyl, aryloxycarbonyl, aryl substituted alkyl, 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 Edition, 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 pharmaceutical compositions comprising the compounds of formula I and formula II of the invention.
- the present invention provides a pharmaceutical composition comprising at least one of the compounds of Formula I or Formula II of the present invention as described above, and optionally a pharmaceutically acceptable excipient.
- Methods of preparing various pharmaceutical compositions containing a certain amount of active ingredient are known, or will be apparent to those skilled in the art from this disclosure. As described by Remington's Pharmaceutical Sciences, Martin, E. W., ed., Mack Publishing Company, 19th ed. (1995). Methods of preparing the pharmaceutical compositions include the incorporation of suitable pharmaceutical excipients, carriers, diluents and the like.
- the pharmaceutical preparations of the invention are prepared in a known manner, including conventional methods of mixing, dissolving or lyophilizing.
- the compounds of the invention may be formulated into pharmaceutical compositions and administered to a patient in a variety of ways suitable for the mode of administration selected, such as orally or parenterally (by intravenous, intramuscular, topical or subcutaneous routes).
- the compounds of the invention may be administered systemically, for example, orally, in association 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. use.
- Such compositions and preparations should contain at least 0.01% of active compound.
- the ratio of such compositions and formulations may of course vary and may range from about 0.1% to about 99% by weight of a given unit dosage form.
- the amount of active compound is such that an effective dosage level can 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 disintegrating agent 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 disintegrating agent 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, wintergreen or cherry
- 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.
- Dispersing agents in glycerol, liquid polyethylene glycols, triacetin and mixtures thereof, and oils can also be 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.
- Proper fluidity can be maintained, for example, by liposome formation, by maintaining the desired particle size in the case of a dispersing agent, or by the use of a surfactant.
- the action of preventing microorganisms can be produced by various antibacterial and antifungal agents such as parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
- 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 absorbent (for example, aluminum monostearate and gelatin).
- Sterile injectable solutions are prepared by combining the required active compound in a suitable solvent with the various other ingredients enumerated above, followed by filter sterilization.
- the preferred preparation methods are vacuum drying and lyophilization techniques which result in a powder of the active ingredient plus any additional ingredients present in the previously sterile filtration solution. .
- 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 such as 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 active ingredient selected, but also on the mode of administration, the nature of the condition to be treated, and the age and condition of the patient, ultimately depending on the attending physician or clinical The doctor's 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 plurality of capsules or tablets.
- the amount of unit dose of the active ingredient may vary or be adjusted between about 0.01 mg to about 1000 mg or more, depending on the particular treatment involved.
- Figure 1 is an immunoblot showing the levels of XPB and Pol II protein in tumor cells and normal cells.
- Figure 2 is an immunoblot showing the inhibition of XP- and Pol II activity of human THP-1 leukemia cells by 14-D-Valine-TPL.
- Figure 3 is an immunoblot showing the detection of 14-D-Valine-TPL inhibition of c-myc activity in human K562 leukemia cells.
- TPL triptolide
- D-Boc-Valine dextrorotatory (N-Boc) valine
- DIC N, N'-diisopropylcarbodiimide
- DMAP 4-dimethylamino Pyridine
- DCM dichloromethane
- TFA trifluoroacetic acid
- the first step thuring triptolide TPL (500 mg, 1.39 mmol, 1.0 eq.), dextrorotatory (N-Boc) valine (1500 mg, 6.91 mmol, 5 eq.) in dichloromethane (20 mL), cooled To 0 ° C. N,N'-diisopropylcarbodiimide (4 mL, 13 mmol, 9.35 eq.) and 4-dimethylaminopyridine (130 mg, 1.07 mmol, 0.8 eq) were added to the mixture at 0 °C.
- reaction was carried out at 25 ° C for 24 hours, and the reaction mixture was diluted with ethyl acetate, washed with water, and then washed with saturated aqueous ammonium chloride, dried and concentrated to give a crude product. Further purification by preparative silica gel column gave a white solid-D-Boc-Valine-TPL (14-D-Boc-valine-triptolide) (250 mg) in a yield of 32.2%.
- Step 2 Dissolve the product from the previous step, D-Boc-Valine-TPL (14-dextro-Boc-valine-triptolide) (200 mg) in dichloromethane (15 mL), add 3 mL three Fluoroacetic acid. After the addition was completed, the reaction solution was reacted at 25 ° C for 3.5 hours. The reaction mixture was diluted with methylene chloride and washed with aqueous sodium hydrogen carbonate, then dried and concentrated to give crude. Further purification by preparative high performance liquid chromatography gave a white solid D-Valine-TPL (14-d- valine-triptolide) (120 mg) in a yield of 73%.
- D-Valine-TPL 14-dextro-valine-triptolide
- L-Boc-Val L-(N-Boc)proline
- HATU 2-(7-azobenzene Triazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
- DIPEA N,N-diisopropylethylamine
- DCM dichloromethane
- TFA trifluoroacetic acid.
- Step 2 The product of the previous step, 14-L-Boc-Valine-D-Valine-TPL (14-L-(N-Boc)-valine-D-valine-triptolide) (85 mg) ) Dissolved in dichloromethane (5 mL) and 1 mL of trifluoroacetic acid was added dropwise. After the addition was completed, the reaction solution was reacted at 25 ° C for 3.5 hours. The reaction mixture was diluted with methylene chloride and washed with aqueous sodium hydrogen carbonate, then dried and concentrated to give crude. Further purification by preparative high performance liquid chromatography gave 14-L-Valine-D-Valine-TPL (14-L-valine-d-valine-triptolide) (51.5 mg) as a white solid.
- Compound 14-L-Valine-TPL (14-L-valine-triptolide) was synthesized as in Example 1, and L-Boc-Valine (L-B-N-Boc-valine) was used instead of D-Boc.
- -Valine-TPL (R-N-Boc-valine.
- Compound 14-L-Valine-L-Valine-TPL (14-L-valine-L-valine-triptolide) was synthesized as in Example 2, using 14-L-Valine-TPL (14-left-handed) Proline-triptolide, Example 3) instead of 14-D-Valine-TPL (14-d-valine-triptolide, Example 1), with L-Boc-Valine (left-handed-N) -Boc-valine) instead of D-Boc-Valine-TPL (dextrorotatory N-Boc-valine).
- Compound 14-D-Valine-D-Valine-TPL (14-D-valine-d-valine-triptolide was synthesized as in Example 2, using D-Boc-Valine (D-Boc-Valine) -Boc-valine) instead of L-Boc-Valine-TPL (L-N-Boc-valine).
- TPL triptolide
- L-Boc-O-TBDMS-Tyrosine L-(N-Boc-O-tert-butyldimethylsilyl) tyrosine
- DIC N, N'-diisopropyl Carbodiimide
- DMAP 4-dimethylaminopyridine
- DCM dichloromethane
- TFA trifluoroacetic acid.
- TBAF tetrabutylammonium fluoride
- D-Valine-TPL 14-dextro-valine-triptolide
- L-Boc-Phenylalanine L-(octa)phenylalanine
- HATU 2-(7-azobenzotriazine) Azole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
- DIPEA N,N-diisopropylethylamine
- L-Tyrosine-TPL 14-L-tyrosine-triptolide
- L-Boc-O-TBDMS-Tyrosine L-(N-Boc-O-tert-butyl) Methylsilyl)tyrosine replaces D-Boc-Valine-TPL (dextrorotatory N-Boc-valine).
- Example 7 Comparison of the therapeutic window of the compound 14-D-Valine-TPL of the present invention with triptolide (TPL)
- Leukemia cell lines Sup-B15 (Ph + acute lymphocytic leukemia), CEM (acute lymphocytic leukemia, ALL), Molt-4 (acute lymphocytic leukemia, ALL).
- Normal blood cell samples from peripheral blood of healthy volunteers.
- Primary reagent 14-D-Valine-TPL of the invention.
- Main instruments cell culture incubator, microplate reader
- the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum.
- Different concentrations of 14-D-Valine-TPL were added, mixed, and cultured in a carbon dioxide (5% CO 2 ) cell incubator at 37 ° C for 72 hours.
- the viable cell concentration was then determined by the MTT method.
- the cell viability of the control group (without compound treatment) was set to 100%, and the cell viability (%) after the action of the compound and the half-growth inhibition concentration (72-hour IC50 value) of the leukemia cells at 72 hours and the therapeutic window index were calculated.
- Treatment window index normal blood cell IC50 value / leukemia cell IC50 value.
- a therapeutic window index of >1 indicates a therapeutic window.
- Compounds are more toxic to tumor cells than normal blood cells, and the larger the therapeutic window, the better the selectivity of the compound for tumor cells.
- the compound 14-D-Valine-TPL of the present invention has a better therapeutic window as shown in Table 1 below, and triptolide has substantially no therapeutic window.
- Table 1 shows that triptolide (TPL) has only a small therapeutic window for Molt-4 cells (2.0), while the other three leukemia cells Jurkat, CEM, and Sup-B15 have a therapeutic window of ⁇ 1, respectively, 0.64. 0.93 and 0.94 indicate that triptolide is essentially non-selective to normal cells and tumor cytotoxicity, which is consistent with the results reported in the literature that TPL has essentially no therapeutic window.
- the 14-D-Valine-TPL of the present invention has a therapeutic window index of 5.54-18.58 for four different types of human leukemia cells, indicating that 14-D-Valine-TPL has a larger therapeutic window for all four leukemias. , wherein the therapeutic window index for Molt-4 cells is as high as 18.58.
- Example 8 Determination of the activity of the compound 14-D-Valine-TPL of the present invention against leukemia in vitro
- Leukemia cell lines human KG-1a (acute myeloid leukemia, AML-M0), THP-1 (acute myeloid leukemia, AML-M5) NB4 (acute promyelocytic leukemia, AML), Kasumi-1 (acute marrow) Leukemia M2 type, AML-M2), KG-1 (acute myeloid leukemia, AML), Jurkat (acute lymphocytic leukemia, ALL), H9 (acute lymphocytic leukemia, ALL).
- Primary reagent 14-D-Valine-TPL of the invention.
- Main instruments cell culture incubator, microplate reader
- the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum.
- Different concentrations of 14-D-Valine-TPL were added, mixed, and cultured in a carbon dioxide (5% CO 2 ) cell incubator at 37 ° C for 72 hours.
- the viable cell concentration was then determined by the MTT method.
- the cell viability of the control group (without compound treatment) was set to 100%, and the cell viability (%) after the action of the compound and the half-growth inhibitory concentration of the leukemia cells at 72 hours (72-hour IC 50 value) were calculated.
- Table 2 shows that 14-D-Valine-TPL of the present invention can induce cell death of human acute myeloid leukemia and acute lymphocytic leukemia and inhibit the growth of these leukemia cells.
- Example 9 Activity assay of anti-human leukemia in vivo of compound 14-D-Valine-TPL mice of the present invention
- NOD/SCID mice were purchased from the Shanghai Animal Center of the Chinese Academy of Sciences
- Leukemia cell lines human KG-1a (acute myeloid leukemia, AML-M0), THP-1 (acute monocytic leukemia, AML-M5), purchased from the ATCC library.
- the 14-D-Valine-TPL trifluoroacetate salt of the present invention was first dissolved in sterile PBS at a concentration of 10 mg/ml and then diluted with sterile deionized water to the desired working concentration.
- 7-week-old female NOD/SCID was inoculated subcutaneously in the subcutaneous area of 1 ⁇ 10 7 cells (0.2 ml).
- the left side was human acute myeloid leukemia KG-1a cells, and the right side was human acute monocytic leukemia THP- 1 cell.
- the tumor tumors were grown to a length of about 0.5 cm and randomly divided into groups of 3 each.
- the control group was treated with equal amount of PBS, 14-D-Valine-TPL component 0.2 mg/kg body weight and 0.4 mg/kg body weight, and administered intragastrically twice a day (8 o'clock in the morning and 4 o'clock in the afternoon). , continuous treatment for 14 days.
- mice were sacrificed, tumor tissues were taken, and the tumor weight was measured.
- the main organs and tissues such as liver, spleen, heart, lung, large intestine and small intestine were examined.
- Table 3 and Table 4 show that 14-D-Valine-TPL of the present invention can significantly inhibit the growth of two different types of human acute myeloid leukemia cell lines KG-1a and THP-1 in mice, and has a dose-dependent effect.
- the 0.4-D-Valine-TPL dose of 0.4 mg per kilogram of body weight can completely abolish the KG-1a and THP-1 xenografts, but the body weight of the mice is not significantly reduced.
- the mice are dissected to check the heart, lung, liver, spleen and large intestine. There were no obvious abnormalities in the main organs such as the small intestine.
- Example 10 Determination of anti-human leukemia activity in 14-L-Valine-L-Valine-TPL (14-LLV-TPL) mice
- NOD/SCID mice were purchased from the Shanghai Animal Center of the Chinese Academy of Sciences
- Leukemia cell line human KG-1a (acute myeloid leukemia, AML-M0) was purchased from the ATCC library.
- the 14-LLV-TPL trifluoroacetate salt of the present invention was first dissolved in sterile PBS at a concentration of 10 mg/ml and then diluted with sterile deionized water to the desired working concentration.
- the main organs and tissues such as liver, spleen, heart, lung, large intestine and small intestine were examined.
- Table 5 shows that 14-LLV-TPL of the present invention can significantly inhibit the growth of human acute myeloid leukemia cell line KG-1a in mice, and there is a significant dose-dependent effect.
- Example 11 Determination of anti-human leukemia activity in 14-L-Valine-D-Valine-TPL (14-DLV-TPL) mice
- NOD/SCID mice were purchased from the Shanghai Animal Center of the Chinese Academy of Sciences
- Leukemia cell line human KG-1a (acute myeloid leukemia, AML-M0) was purchased from the ATCC library.
- the 14-DLV-TPL trifluoroacetate salt of the present invention was first dissolved in sterile PBS at a concentration of 10 mg/ml and then diluted with sterile deionized water to the desired working concentration.
- the main organs and tissues such as liver, spleen, heart, lung, large intestine and small intestine were examined.
- Table 6 shows that 14-DLV-TPL of the present invention can significantly inhibit the growth of human acute myeloid leukemia cell line KG-1a in mice, and there is a significant dose-dependent effect.
- Example 12 Determination of anti-human leukemia activity in 14-L-Phenylalanyl-L-tyrosine-TPL (14-LPT-TPL) mice
- NOD/SCID mice were purchased from the Shanghai Animal Center of the Chinese Academy of Sciences
- Leukemia cell line human KG-1a (acute myeloid leukemia, AML-M0) was purchased from the ATCC library.
- the 14-LPT-TPL trifluoroacetate of the present invention was first dissolved in sterile PBS at a concentration of 10 mg/ml and then diluted with sterile deionized water to the desired working concentration.
- the main organs and tissues such as liver, spleen, heart, lung, large intestine and small intestine were examined.
- Table 7 shows that 14-LPT-TPL of the present invention can significantly inhibit the growth of human acute myeloid leukemia cell line KG-1a in mice, and there is a significant dose-dependent effect.
- Example 13 Determination of anti-human leukemia activity in 14-D-Valine-D-Valine-TPL (14-DDV-TPL) mice
- mice NOD/SCID mice were purchased from the Shanghai Animal Center of the Chinese Academy of Sciences.
- Leukemia cell line Human K562/ADR (chronic myeloid leukemia, CML) was purchased from the ATCC library.
- Reagents The 14-DDV-TPL trifluoroacetate salt of the present invention was first dissolved in sterile PBS at a concentration of 10 mg/ml and then diluted with sterile deionized water to the desired working concentration.
- Main instruments cell culture incubator, microplate reader
- the main organs and tissues such as liver, spleen, heart, lung, large intestine and small intestine were examined.
- Table 8 shows that 14-DDV-TPL of the present invention can significantly inhibit the growth of human chronic myeloid leukemia cell line K562/ADR organelle in mice, and there is a significant dose-dependent effect.
- Example 14 Determination of anti-human leukemia activity in 14-L-Valine-TPL (14-LV-TPL) mice
- NOD/SCID mice were purchased from the Shanghai Animal Center of the Chinese Academy of Sciences
- Leukemia cell line Human K562/ADR (chronic myeloid leukemia, CML) was purchased from the ATCC library.
- the 14-LV-TPL trifluoroacetate salt of the present invention was first dissolved in sterile PBS at a concentration of 10 mg/ml and then diluted with sterile deionized water to the desired working concentration.
- the main organs and tissues such as liver, spleen, heart, lung, large intestine and small intestine were examined.
- Table 9 shows that 14-LV-TPL of the present invention can significantly inhibit the growth of human chronic myeloid leukemia cell line K562/ADR organelle in mice, and there is a significant dose-dependent effect.
- Example 15 Inhibition of T lymphocyte and B lymphocyte activity by 14-D-Valine-TPL in vitro
- Leukemia cell lines Jurkat (T lymphocytes), H9 (T lymphocytes), CEM (B lymphocytes), and Sup-B15 (B lymphocytes).
- the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum.
- Different concentrations of 14-D-Valine-TPL were added, mixed, and cultured in a carbon dioxide (5% CO 2 ) cell incubator at 37 ° C for 72 hours.
- the viable cell concentration was then determined by the MTT method.
- the cell viability of the control group (without compound treatment) was set to 100%, and the cell viability (%) after the action of the compound and the half-growth inhibitory concentration of the leukemia cells at 72 hours (72-hour IC 50 value) were calculated.
- Table 10 shows that 14-D-Valine-TPL of the present invention can significantly inhibit the growth of these T lymphocytes and B lymphocytes.
- Example 16 Expression of transcription factor TFIIH subunit XPB and RNA polymerase II (Pol II) in different types of tumor cells and normal cells
- TFIIH subunit XPB xeroderma pigmentosum group B
- RNA polymerase II RNA polymerase II
- Leukemia cell lines Nalm6 (lymphoma), Jurkat (acute lymphocytic leukemia), THP-1 (acute monocytic leukemia), KG-1a (acute myeloid leukemia), HL-60 (acute myeloid leukemia), NB4 (Acute promyelocytic leukemia), U937 (acute monocytic leukemia). Normal blood cell samples were obtained from volunteers.
- Figure 1 shows the results of immunoblotting to detect the expression levels of XPB and Pol II in different types of tumor cells and normal cells. Among them, 1: normal blood cells; 2: Nalm6; 3: Jurkat; 4: THP-1; 5: KG-1a; 6: HL-60; 7: NB4; 8: U937.
- Example 17 14-D-Valine-TPL inhibits XPB and Pol II activity in human THP-1 leukemia cells
- XPB and Pol II are key target molecules for the pharmacological action of triptolide [Titov DV, et al. XPB, a subunit of TFIIH, is a target of the natural product triptolide. Nature chemical biology. 2011, 7:182 -188].
- the present invention uses cell culture techniques and immunoblotting techniques to detect the effect of 14-D-Valine-TPL on the activity of XPB and Pol II in leukemia cells.
- Leukemia cell line human THP-1 leukemia cell line (acute myeloid leukemia-M5, AML-M5).
- the well-growth leukemia cells were inoculated into the wells of a 6-well cell culture plate at a density of 1 ⁇ 10 6 /ml.
- the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum.
- Different concentrations of 14-D-Valine-TPL were added, mixed, and cultured in a carbon dioxide (5% CO 2 ) cell incubator at 37 ° C for 48 hours. Cellular proteins were then extracted and XPB and Pol II expression levels were detected by immunoblotting.
- Figure 2 shows the results of 14-D-Valine-TPL inhibition of XPB and Pol II activity in human THP-1 leukemia cells by cell culture technique and immunoblotting
- Example 18 14-D-Valine-TPL inhibits c-myc oncogene activity in human K562 leukemia cells
- oncogene c-myc is also one of the key target molecules for triptolide to exert anticancer pharmacological effects [Stéphane Vis conclusions, et al. Triptolide is an inhibitor of RNA polymerase I and II-dependent transcription leading predominantly down-regulation Of short-lived mRNA. Mol Cancer Ther 2009; 8: 2780-2790].
- the present invention uses cell culture techniques and immunoblotting techniques to examine the effect of 14-D-Valine-TPL on c-myc activity in leukemia cells.
- Leukemia cell line human K562 leukemia cell line (chronic myeloid leukemia).
- the well-growth leukemia cells were inoculated into the wells of a 6-well cell culture plate at a density of 1 ⁇ 10 6 /ml.
- the culture broth was a 1640 cell culture medium containing 10% fetal bovine serum.
- Different concentrations of 14-D-Valine-TPL were added, mixed, and cultured in a carbon dioxide (5% CO 2 ) cell incubator at 37 ° C for 48 hours. Cellular proteins were then extracted and c-myc protein expression levels were detected by immunoblotting.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
- Medicinal Preparation (AREA)
- Steroid Compounds (AREA)
- Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
Abstract
Description
| 14-D-Valine-TPL | TPL | |||
| 白血病细胞系 | IC50(nM/L) | 治疗窗 | IC50 | 治疗窗 |
| Jurkat | 36.301 | 5.54 | 33.829 | 0.64 |
| CEM | 25.488 | 7.89 | 23.286 | 0.93 |
| Molt-4 | 10.824 | 18.58 | 10.718 | 2.03 |
| Sup-B15 | 20.674 | 9.73 | 23.153 | 0.94 |
| 正常血细胞 | ||||
| No1 | 202.706 | 29.289 | ||
| No2 | 198.826 | 20.772 | ||
| No3 | 205.784 | 9.383 | ||
| No4 | 199.835 | 8.968 | ||
| No5 | 213.719 | 20.072 | ||
| No6 | 189.872 | 20.445 | ||
| No7 | 203.546 | 19.515 | ||
| No8 | 184.509 | 9.710 | ||
| No9 | 210.835 | 57.443 | ||
| 合计 | 201.07 | 21.733 |
| 14-D-Val-TPL | 14-L-Val-TPL | 14-L-Val-14-L-Val-TPL | |
| 细胞系 | IC50(nM/L) | IC50(nM/L) | IC50(nM/L) |
| KG-1 | 49.0 | 8.86 | |
| KG-1a | 31.0 | 20 | |
| THP-1 | 16.0 | 5.0 | |
| HL-60 | 65.8 | ||
| U937 | 60.0 | ||
| Jurkat | 36.301 | ||
| CEM | 25.488 | ||
| Molt-4 | 10.824 | ||
| Sup-B15 | 20.674 |
| 14-D-Valine-TPL | |
| 免疫细胞系 | IC50(nM/L) |
| Jurkat(T淋巴细胞) | 36.301 |
| CEM(B淋巴细胞) | 25.488 |
| Molt-4(T淋巴细胞) | 10.824 |
| Sup-B15(B淋巴细胞) | 20.674 |
Claims (23)
- 通式(I)和通式(II)的C14-位羟基酯化的雷公藤甲素氨基酸衍生物,其中R1和R4选自有取代或无取代的C2-C20烷基、有取代或无取代的C2-C20烯烃基,有取代或无取代的C4-C20的共扼烯烃基,有取代或无取代的C3-C7环烷基或环烯烃基,有取代或无取代的芳烃基,有取代或无取代的杂环基和芳香杂环基,有取代或无取代的氨基酸侧链烷基;氨基酸可以是消旋的,也可以是光学纯(左旋或右旋)的;取代基选自卤素,胺基,C1-C6取代胺基,硝基,氰基,羟基,C1-C6烷氧基,巯基,C1-C6烷硫基;其中R2,R3,R5和R6选自H、有取代或无取代的C1-C20烷基、有取代或无取代的C2-C20烯烃基,有取代或无取代的C4-C20的共扼烯烃基,有取代或无取代的C3-C7环烷基或环烯烃基,有取代或无取代的芳烃基,有取代或无取代的杂环基和芳香杂环基,有取代或无取代的氨基酸侧链烷基,氨基酸可以是消旋的,也可以是光学纯(左旋或右旋)的;取代基选自卤素,胺基,C1-C6取代胺基,硝基,氰基,羟基,C1-C6烷氧基,巯基,C1-C6烷硫基。R2和R3,R5和R6可以是BOC,Fmoc,Bn等保护基。R2和R3相同或不相同。R5和R6相同或不相同。
- 根据权利要求1所述的C14-位羟基酯化的雷公藤甲素氨基酸衍生物的药学上可接受的加成物或者复合物。
- 根据权利要求1所述的C14-位羟基酯化的雷公藤甲素氨基酸衍生物的药学上可接受的盐。
- 根据权利要求1所述的C14-位羟基酯化的雷公藤甲素氨基酸衍生物的分解和代谢产物。
- 根据权利要求1的雷公藤甲素的C14-位羟基酯化的雷公藤甲素氨基酸衍生物的药学上可接受的加成物、复合物、盐、以及分解和代谢产物,其中R1和R4选自有取代或无取代的C2-C20烷基、有取代或无取代的C2-C20烯烃基,取代或无取代的C4-C20的共扼烯烃基,有取代或无取代的C3-C7环烷基或环烯烃基,有取代或无取代的芳烃基,有取代或无取代的杂环基和芳香杂环基,有取代或无取代的氨基酸侧链烷基,氨基酸可以是消旋的,也可以是光学纯(左旋或右旋);取代基选自卤素,胺基,C1-C6取代胺基,硝基,氰基,羟基,C1-C6烷氧基,巯基,C1-C6烷硫基;R2和R3,R5和R6是或不是保护基;R2和R3相同或不相同;R5和R6相同或不相同;其中R2,R3,R5和R6选自H、有取代或无取代的C1-C20烷基、有取代或无取代的C2-C20烯烃基,取代或无取代的C4-C20的共扼烯烃 基,有取代或无取代的C3-C7环烷基或环烯烃基,有取代或无取代的芳烃基,有取代或无取代的杂环基和芳香杂环基,有取代或无取代的氨基酸侧链烷基,氨基酸可以是消旋的,也可以是光学纯(左旋或右旋)的;取代基选自卤素,胺基,C1-C6取代胺基,硝基,氰基,羟基,C1-C6烷氧基,巯基,C1-C6烷硫基。
- 根据权利要求1-4任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物,其中R1和R2选自取代或无取代的芳烃基;取代基选自卤素,胺基,C1-C6取代胺基,硝基,氰基,羟基,C1-C6烷氧基,巯基,C1-C6烷硫基;取代基可选择一个或数个同时存在。
- 一种药物组合物,其中包含权利要求1-7中任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物和任选的药学上可以接受的赋形剂。
- 权利要求1-7中的任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物在制备抗肿瘤药物中的用途。
- 权利要求1-7任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物作为抗肿瘤治疗剂的用途。
- 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1-7任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物。
- 根据权利要求11或12所述的用途,其特征在于,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。
- 根据权利要求13所述的方法,其特征在于,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。
- 根据权利要求1-7中的任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物在制备抗免疫性疾病药物中的用途。
- 根据权利要求16的用途,其特征在于,所述免疫性疾病选自与免疫细胞功能异常相关的自 身免疫性疾病。
- 根据权利要求16的用途,其特征在于,所述免疫性疾病选自与T细胞或B细胞功能异常相关的自身免疫性疾病.
- 根据权利要求16的用途,其特征在于,所述免疫性疾病的选择范围包括类风湿性关节炎、系统性红斑狼疮、银屑病、肾病。
- 根据权利要求1-7中的任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物在制备XPB或Pol II异常表达相关疾病药物中的用途。
- 根据权利要求20的用途,其特征在于,所述XPB或Pol II异常表达相关疾病选自与XPB或Pol II过表达相关的肿瘤。
- 根据权利要求1-7中的任一项的C14-位羟基酯化的雷公藤甲素氨基酸衍生物或其药学上可接受的加成物、复合物、盐、以及分解和代谢产物在制备致癌基因c-myc异常表达相关疾病药物中的用途。
- 根据权利要求22的用途,其特征在于,所述c-myc异常表达相关疾病选自与c-myc过表达相关的肿瘤。
Priority Applications (11)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| HK18105555.6A HK1246279B (zh) | 2015-10-29 | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 | |
| EP15906944.2A EP3248981B1 (en) | 2015-10-29 | 2015-10-29 | C14-hydroxyl esterified amino acid derivatives of triptolide, and preparation method and use thereof |
| CA2977559A CA2977559C (en) | 2015-10-29 | 2015-10-29 | C14-hydroxyl esterified amino acid derivative of triptolide, and preparation method and use thereof |
| ES15906944T ES2909799T3 (es) | 2015-10-29 | 2015-10-29 | Derivados aminoacídicos de triptólido esterificados en el hidroxilo C14 y método de preparación y uso de los mismos |
| JP2017548297A JP6827942B2 (ja) | 2015-10-29 | 2015-10-29 | トリプトリドのc14ヒドロキシルエステル化アミノ酸誘導体、ならびにその製造方法および使用 |
| DK15906944.2T DK3248981T3 (da) | 2015-10-29 | 2015-10-29 | C14-hydroxyl-forestrede aminosyrederivater af triptolid, og fremstillingsfremgangsmåde og anvendelse deraf |
| US15/551,695 US10238623B2 (en) | 2015-10-29 | 2015-10-29 | C14-hydroxyl esterified amino acid derivative of triptolide, and preparation method and use thereof |
| CN201580074254.0A CN107207525B (zh) | 2015-10-29 | 2015-10-29 | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 |
| AU2015413013A AU2015413013B2 (en) | 2015-10-29 | 2015-10-29 | C14-hydroxyl esterified amino acid derivatives of triptolide, and preparation method and use thereof |
| KR1020177025330A KR102452412B1 (ko) | 2015-10-29 | 2015-10-29 | 트리프톨리드의 c14-히드록실 에스테르화 아미노산 유도체, 및 그의 제조 방법 및 용도 |
| PCT/CN2015/093161 WO2017070878A1 (zh) | 2015-10-29 | 2015-10-29 | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 |
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| PCT/CN2015/093161 WO2017070878A1 (zh) | 2015-10-29 | 2015-10-29 | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2017070878A1 true WO2017070878A1 (zh) | 2017-05-04 |
Family
ID=58631173
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2015/093161 Ceased WO2017070878A1 (zh) | 2015-10-29 | 2015-10-29 | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 |
Country Status (10)
| Country | Link |
|---|---|
| US (1) | US10238623B2 (zh) |
| EP (1) | EP3248981B1 (zh) |
| JP (1) | JP6827942B2 (zh) |
| KR (1) | KR102452412B1 (zh) |
| CN (1) | CN107207525B (zh) |
| AU (1) | AU2015413013B2 (zh) |
| CA (1) | CA2977559C (zh) |
| DK (1) | DK3248981T3 (zh) |
| ES (1) | ES2909799T3 (zh) |
| WO (1) | WO2017070878A1 (zh) |
Cited By (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN109021061A (zh) * | 2018-09-29 | 2018-12-18 | 郭可点 | 雷公藤甲素靶向前药及其制备方法和应用 |
| WO2023143330A1 (zh) * | 2022-01-25 | 2023-08-03 | 杭州卫本医药科技有限公司 | 具有杀伤myc阳性细胞功能雷公藤二萜类环氧化合物、及其制备方法和应用 |
Families Citing this family (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN110003304B (zh) * | 2019-04-22 | 2022-02-22 | 湖北大学 | 一种水溶性雷公藤甲素衍生物及其制备方法和用途 |
| CN110229210B (zh) * | 2019-06-25 | 2022-07-12 | 河南中医药大学 | 雷公藤红素的氨基酸共晶物及其制备方法和用途 |
| CN115397843A (zh) * | 2020-04-15 | 2022-11-25 | 杭州卫本医药科技有限公司 | 用于筛选药物的靶标多肽和筛选方法 |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1246121A (zh) * | 1996-03-01 | 2000-03-01 | 法玛吉尼西斯公司 | 免疫抑制化合物及方法 |
| WO2002074759A1 (en) * | 2001-03-15 | 2002-09-26 | Pharmagenesis, Inc. | Amino acid derivatives of triptolide compounds as immune modulators and anticancer agent |
| WO2008087202A1 (en) * | 2007-01-18 | 2008-07-24 | Pierre Fabre Medicament | Triptolide derivatives for use in the treatment of acute myeloid leukemia |
Family Cites Families (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| ATE222910T1 (de) | 1997-05-23 | 2002-09-15 | Aventis Pharma Inc | Triptolid-derivate nützlich zur behandlung von autoimmunkrankheiten |
| CN100374443C (zh) * | 2004-03-29 | 2008-03-12 | 北京键凯科技有限公司 | 亲水性聚合物-雷公藤提取物的结合物及其药物组合物 |
| CN103816548A (zh) * | 2014-03-11 | 2014-05-28 | 北京林业大学 | 靶向亲水性聚合物-雷公藤甲素结合物 |
-
2015
- 2015-10-29 EP EP15906944.2A patent/EP3248981B1/en active Active
- 2015-10-29 CA CA2977559A patent/CA2977559C/en active Active
- 2015-10-29 CN CN201580074254.0A patent/CN107207525B/zh active Active
- 2015-10-29 DK DK15906944.2T patent/DK3248981T3/da active
- 2015-10-29 KR KR1020177025330A patent/KR102452412B1/ko active Active
- 2015-10-29 ES ES15906944T patent/ES2909799T3/es active Active
- 2015-10-29 US US15/551,695 patent/US10238623B2/en active Active
- 2015-10-29 WO PCT/CN2015/093161 patent/WO2017070878A1/zh not_active Ceased
- 2015-10-29 AU AU2015413013A patent/AU2015413013B2/en active Active
- 2015-10-29 JP JP2017548297A patent/JP6827942B2/ja active Active
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1246121A (zh) * | 1996-03-01 | 2000-03-01 | 法玛吉尼西斯公司 | 免疫抑制化合物及方法 |
| WO2002074759A1 (en) * | 2001-03-15 | 2002-09-26 | Pharmagenesis, Inc. | Amino acid derivatives of triptolide compounds as immune modulators and anticancer agent |
| WO2008087202A1 (en) * | 2007-01-18 | 2008-07-24 | Pierre Fabre Medicament | Triptolide derivatives for use in the treatment of acute myeloid leukemia |
Non-Patent Citations (1)
| Title |
|---|
| See also references of EP3248981A4 * |
Cited By (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN109021061A (zh) * | 2018-09-29 | 2018-12-18 | 郭可点 | 雷公藤甲素靶向前药及其制备方法和应用 |
| CN109021061B (zh) * | 2018-09-29 | 2019-07-12 | 郭可点 | 雷公藤甲素靶向前药及其制备方法和应用 |
| WO2023143330A1 (zh) * | 2022-01-25 | 2023-08-03 | 杭州卫本医药科技有限公司 | 具有杀伤myc阳性细胞功能雷公藤二萜类环氧化合物、及其制备方法和应用 |
Also Published As
| Publication number | Publication date |
|---|---|
| AU2015413013B2 (en) | 2020-11-05 |
| AU2015413013A1 (en) | 2017-09-07 |
| CA2977559C (en) | 2022-04-12 |
| HK1246279A1 (zh) | 2018-09-07 |
| CN107207525B (zh) | 2018-11-27 |
| JP2018536619A (ja) | 2018-12-13 |
| US10238623B2 (en) | 2019-03-26 |
| EP3248981A1 (en) | 2017-11-29 |
| CN107207525A (zh) | 2017-09-26 |
| ES2909799T3 (es) | 2022-05-10 |
| KR102452412B1 (ko) | 2022-10-06 |
| CA2977559A1 (en) | 2017-05-04 |
| DK3248981T3 (da) | 2022-03-28 |
| EP3248981A4 (en) | 2018-07-18 |
| JP6827942B2 (ja) | 2021-02-10 |
| US20180036277A1 (en) | 2018-02-08 |
| EP3248981B1 (en) | 2022-03-09 |
| KR20180073517A (ko) | 2018-07-02 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2019223718A1 (zh) | 一种免疫调节剂 | |
| WO2017070878A1 (zh) | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 | |
| WO2013079024A1 (zh) | 齐墩果酸酰胺化衍生物、及其制备方法和应用 | |
| EP2805954B1 (en) | 7-substituted hanfangichin b derivative, and preparation method and use thereof | |
| CN113698401A (zh) | β-榄香烯大环衍生物及其制备方法和应用 | |
| EP2786990B1 (en) | 2-aminated methylene or 2-esterified methylene tanshinone derivatives, and preparation method and application thereof | |
| WO2012175002A1 (zh) | 10-甲氧基喜树碱衍生物、制备方法和用途 | |
| US9353146B2 (en) | Acylation derivatives of paridis saponins I, preparation method therefor and application thereof | |
| CN108484640B (zh) | 一种抗肿瘤的细胞凋亡蛋白抑制剂 | |
| CN104039794B (zh) | 7‑位取代的汉防己乙素衍生物、及其制备方法和应用 | |
| CN103946217B (zh) | 2‑位胺化亚甲基或2‑位酯化亚甲基丹参酮衍生物、及其制备方法和应用 | |
| CN102241648A (zh) | 抗多药耐药紫杉烷类衍生物及其制备方法和应用 | |
| CN104334571B (zh) | 重楼皂苷i的酰化衍生物、及其制备方法和应用 | |
| EP3917933A1 (en) | Duocarmycin analogues | |
| CN103450164B (zh) | 格尔德霉素衍生物及其制备方法和用途 | |
| CN101855216A (zh) | 三尖杉宁碱衍生物、及其制法和其药物组合物与用途 | |
| CN103635476B (zh) | 高三尖杉酯碱的酰化衍生物、及其制备方法和应用 | |
| HK1246279B (zh) | C14-位羟基酯化的雷公藤甲素氨基酸衍生物、及其制备方法和应用 | |
| 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 |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 15906944 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 15551695 Country of ref document: US |
|
| ENP | Entry into the national phase |
Ref document number: 2977559 Country of ref document: CA |
|
| REEP | Request for entry into the european phase |
Ref document number: 2015906944 Country of ref document: EP |
|
| ENP | Entry into the national phase |
Ref document number: 2015413013 Country of ref document: AU Date of ref document: 20151029 Kind code of ref document: A |
|
| ENP | Entry into the national phase |
Ref document number: 20177025330 Country of ref document: KR Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2017548297 Country of ref document: JP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |