WO2013104263A1 - 多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用 - Google Patents

多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用 Download PDF

Info

Publication number
WO2013104263A1
WO2013104263A1 PCT/CN2012/088016 CN2012088016W WO2013104263A1 WO 2013104263 A1 WO2013104263 A1 WO 2013104263A1 CN 2012088016 W CN2012088016 W CN 2012088016W WO 2013104263 A1 WO2013104263 A1 WO 2013104263A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
methyl
benzopyran
hydrogen
compound
Prior art date
Application number
PCT/CN2012/088016
Other languages
English (en)
French (fr)
Inventor
丁红霞
李靖
孟坤
Original Assignee
北京盛诺基医药科技有限公司
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 北京盛诺基医药科技有限公司 filed Critical 北京盛诺基医药科技有限公司
Priority to US14/371,866 priority Critical patent/US9221781B2/en
Priority to KR1020147022558A priority patent/KR102030207B1/ko
Priority to BR112014016635A priority patent/BR112014016635B8/pt
Priority to CA2860999A priority patent/CA2860999C/en
Priority to EP12864757.5A priority patent/EP2803665B1/en
Priority to JP2014551506A priority patent/JP6113751B2/ja
Priority to ES12864757.5T priority patent/ES2627316T3/es
Priority to AU2012365712A priority patent/AU2012365712B2/en
Priority to MX2014008526A priority patent/MX364833B/es
Publication of WO2013104263A1 publication Critical patent/WO2013104263A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/26Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3
    • C07D311/28Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only
    • C07D311/30Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only not hydrogenated in the hetero ring, e.g. flavones
    • 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

Definitions

  • the present invention relates to polyhydroxybenzopyrone compounds and salts or prodrugs thereof, and to pharmaceutical compositions containing such compounds for use in the manufacture of a medicament for the prevention and/or treatment of neoplastic diseases.
  • Estrogen is a group of hormones associated with many key physiological functions in the human body.
  • the functions of estrogen include promoting female sexual organ development, preparing for breast and uterus during pregnancy, and breastfeeding after childbirth. Estrogen also plays an important role in maintaining proper cardiovascular function and bone density. It is well known that estrogen can stimulate cell proliferation, which may increase the risk of cancer in women, especially breast cancer and uterine cancer. Estrogen regulates cellular function by binding to estrogen receptors in target cells. Two estrogen receptors (ERs), ER- ⁇ and ER-P have been found in human cells.
  • N-terminal domain A/B domain
  • C domain mid-DNA binding domain
  • D/E/F domain C-terminal ligand binding Domain
  • the N-terminal domain has a non-ligand-dependent activation function (AF-1), which interacts with a coactivator and transcriptionally activates a target gene in the absence of a ligand.
  • AF-1 non-ligand-dependent activation function
  • DNA binding domains play an important role in receptor dimerization and binding to specific DNA sequences.
  • the C-terminal ligand binding domain mediates ligand binding and has a ligand-dependent transcriptional activation function (AF-2) that activates gene transcription in the presence of a ligand.
  • AF-1 non-ligand-dependent activation function
  • AF-2 ligand-dependent transcriptional activation function
  • ER- ⁇ The full-length ER- ⁇ is a protein with a molecular weight of 66 kDa and is called ER-a66.
  • ER-a66 contains all three functional domains.
  • the splice variant of ER-a66 was later discovered and named ER-a46.
  • ER-a46 has a molecular weight of approximately 46 kDa, which lacks the N-terminal AF-1 domain of hER-a66.
  • ER-a36 a new 36 kDa ER- ⁇ variant, ER-a36, was discovered. It lacks the N-terminal AF-1 domain of ER-a66 and the C-terminal AF-2 domain (see wang et al, Biochem. Biophys. Res. Commun.
  • ER-a66 is generally considered Transcription activates its target gene to mediate estrogen-stimulated cell proliferation.
  • the binding of estrogen to ER-a66 activates the transcriptional activation domain of ER-a66, thereby stimulating the expression of downstream target genes and ultimately leading to cell proliferation.
  • ER-a46 has been shown to mediate rapid NO synthesis initiated by membranes and stimulated by estrogen (see Li et al, Proc. Natl. Acad. Sci. USA 100: 4807-4812, (2003)) and ER-a46 lacking the AF-1 domain was found to inhibit AF-1 activity of ER-a66 (see Flouriot, G., EMBO, 19, 4688-4700, (2000)).
  • ER-a36 deletion of AF-1 and The AF-2 transcriptional activation domain which acts as a dominant negative inhibitor, inhibits the AF-1 and AF-2 functions of ER-a and ER- ⁇ .
  • ER-a36 is mainly distributed on the cell membrane and mediates Membrance-induced conduction of mitogenic estrogen signaling, which stimulates cell proliferation (see wang et al, Biochem. Biophys. Res. Commun. 336, 1023-1027, (2005); wang et al, Proc. Natl. Acad. Sci. USA 103: 9063-9068, (2006) In-depth studies have shown that estrogen signaling is mediated through traditional nuclear transcriptional activation pathways and non-traditional membrane-primed signaling pathways.
  • ER-a66 and ER-a46 appear to act primarily in the nucleus, whereas ER-a36 appears to act primarily through the nucleus. It has also been shown that ER-a36 lacks the helix 8-12 of the ligand binding domain possessed by the original ER-a66, which completely alters the specificity of ER-a36 ligand binding. Therefore, ER-a36 may bind different ligands to ER-a66 and ER- ⁇ . Since diseases associated with estrogen receptors still affect many people, there is an urgent need to find a novel compound and a pharmaceutical composition thereof for preventing and/or treating these related diseases. Summary of the invention
  • the present invention provides a class of polyhydroxybenzopyrone compounds and salts or prodrugs thereof which are useful for regulating the function of the novel estrogen receptor ER-a36, and the formula (I):
  • R 1 selected from hydrogen, (CC 6) alkyl containing one or more halogen atoms (CC 6) alkyl;
  • R 2, R 3, R 4 , R 5 and R 6 may be independently selected from hydrogen, (dC 4) alkyl, containing one or more substituted with halogen atom (dC 4) alkyl, halo, cyano, containing a Or a plurality of halogen atom-substituted (dC 6 ) alkoxy groups; but R 2 , R 3 , R 4 , R 5 and R 6 may not be hydrogen at the same time;
  • R 4 may not be a chlorine atom.
  • Figure 1 shows the results of Western blotting of ER-a66, ER-a46 and ER-a36 expression in human breast cancer samples.
  • Lane 1 normal breast tissue; tract 2: invasive ductal carcinoma; tract 3: invasive ductal carcinoma; tract 4: invasive ductal carcinoma; tract 5: invasive lobular carcinoma; tract 6: invasive lobular carcinoma; Non-invasive ductal carcinoma.
  • Figure 2 (top panel) shows the results of immunofluorescence staining of MDA-MB-231 cells.
  • MDA-MB-231 cells are ER-negative breast cancer cell lines lacking ER-a66 and ER-a46.
  • MDA-MB-231 cells were stained with an antibody that specifically binds to ER-a36 (left panel labeled "ER-a36 Ab” in the figure: positive staining is shown in green). Using 4,6-diindol-2-phenylindole for cells The core is stained. (The middle panel labeled "DAPI'' in the figure: the positive staining is shown in blue.) The combined staining signals are labeled as "merging.” When the antibody is preincubated with the immunogenic polypeptide bound to the antibody, The results were negative (bottom).
  • Figure 3 shows the results of Western blotting of ER-0136 expressing different tumor cell lines.
  • Lane 1 293 human renal epithelial cell lines transiently overexpressing ER-0136; Human-derived human breast cancer SK-BR-3 cell line; Lane 5-7: Human breast cancer MCF-7 cell line from different laboratory sources; Lane 8-9: Human leukemia HL-60 cells from different laboratory sources Strain 10-11: Human leukemia MV-4-11 cell line from different laboratory sources; Lane 12-13: Human granulocyte leukemia K562 cells from different laboratory sources; Lane 14: Human liver cancer A2780 cells; : Human liver cancer HEL-7402 cells; Lane 16: Human liver cancer HEL-9204 cells; Lane 17: Primary liver cancer Hep-11 cells derived from patients; Lane 18: Primary liver cancer Hep-12 cells derived from patients.
  • Figure 8 shows the detection of compound 1 in vitro against gastric cancer BGC-823 cells, lung cancer H460 cells by MTT assay, Growth inhibition of colon cancer LS174T cells, pancreatic cancer PANC-1 and prostate cancer PC-3 cells. The results showed that Compound 1 had significant growth inhibitory effects on these tumor cells in a dose-dependent manner.
  • Figure 9 shows the comparison with tamoxifen (0.7 mg/mouse/day), compound 1 (0.7 mg/mouse/day) and negative control blank vehicle (0.2 mL/mouse/day), respectively.
  • the average tumor weight (histogram a) of the tumor-bearing human breast cancer BCAP-37 tumor strain after 20 days of continuous administration showed the inhibition of tumor growth of the compound.
  • Figure 10 shows that when rituximab was used respectively Positive control, growth curve of tumors administered to nude mice bearing tumor-bearing human B lymphoma Daudi cells for 21 days in a compound 1 (0.7 mg/mouse/day) and a negative control blank vehicle (0.2 mL/mouse/day).
  • Figure 11 shows when the positive control medroxyprogesterone acetate (120 mg/kg), and the low (17.5 mg/kg), medium (35 mg/kg), high dose (70 mg/kg) and negative of the compound 1 were respectively negative.
  • Control blank vehicle (0.2 mL/mouse/day) on tumor-bearing human endometrial carcinoma Ishikawa tumor strain nude mice Continued administration after 20 days the average tumor weight. Remarkably the growth inhibitory effects of compounds on tumor tumor of mice, and better than the positive control.
  • a benzopyrone compound a pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutical composition comprising the same, which are useful in Regulates the function of the novel estrogen receptor ER-a36, and prevents and/or treats diseases mediated by the ER-0136 receptor, such as cancer.
  • the invention provides a compound of formula (I):
  • R 1 selected from hydrogen, (CC 6) alkyl containing one or more halogen atoms (CC 6) alkyl;
  • R 2, R 3, R 4 , R 5 and R 6 may be independently selected from hydrogen, (dC 4) alkyl, containing one or more substituted with halogen atom (dC 4) alkyl,, cyano, containing a Or a plurality of atom-substituted (dC 4 ) alkoxy groups, but R 2 , R 3 , R 4 , R 5 and R 6 may not be hydrogen at the same time; and when R 1 is a methyl group and R 3 and R 5 are hydrogen R 4 may not be a chlorine atom.
  • One embodiment of the present invention comprises a group of compounds having the structure of formula (I), referred to as a compound ( ⁇ ), wherein the compound has the formula:
  • R 2, R 3, R 4 , R 5 and R 6 are independently selected from hydrogen, (C r C 4) alkyl, containing one or more halogen atoms, substituted with (C r C 4) alkyl, Su, cyano a group having one or more atom-substituted (C r C 4 ) alkoxy groups; but R 2 , R 3 , R 4 , R 5 and R 6 may not be hydrogen at the same time.
  • One embodiment of the present invention comprises a group of compounds of the formula (I), referred to as a compound ( ⁇ ), wherein the group of compounds has the formula:
  • R 2, R 3, R 4 , R 5 and R 6 may be independently selected from hydrogen, (dC 4) alkyl, containing one or more substituted with halogen atom (dC 4) alkyl,, cyano, containing a Or a plurality of atom-substituted (dC 4 ) alkoxy groups; but R 2 , R 3 , R 4 , R 5 and R 6 may not be hydrogen at the same time.
  • R 4 may not be a chlorine atom.
  • Particularly preferred compounds of the formula (I) of the present invention include, but are not limited to, the following compounds:
  • substitution means that a hydrogen atom in a molecule is replaced by a different atom or molecule.
  • An atom or molecule that replaces a hydrogen atom is referred to as a "substituent.”
  • the minimum and maximum values of the carbon atom content in the hydrocarbon group are indicated by a prefix, for example, the prefix (C a -C b )alkyl group indicates any alkyl group having from “a” to "b” carbon atoms.
  • (dQ alkyl refers to an alkyl group containing from 1 to 6 carbon atoms.
  • alkoxy refers to a straight or branched, monovalent, saturated fat bonded to an oxygen atom.
  • alkyl refers to a straight chain or a branch. Chained, monovalent, saturated aliphatic chains including, but not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, pentyl, isopentyl, hexyl, and the like.
  • halogen or "halogen atom” means a chlorine, bromine, fluorine and iodine atom or group.
  • heteroaryl means that one or more of the carbon atoms have been replaced by a hetero atom such as nitrogen, oxygen or sulfur.
  • a hetero atom such as nitrogen, oxygen or sulfur.
  • Monocyclic or polycyclic aromatic hydrocarbon groups If a heteroaryl group contains more than one hetero atom, these heteroatoms may be the same or different.
  • Heteroaryl groups include, but are not limited to, benzofuranyl, benzothienyl Benzimidazolyl, benzoxazolyl, benzothiazolyl, benzopyranyl, furan , imidazolyl, oxazolyl, fluorenyl, fluorenyl, isobenzofuranyl, isodecyl, isoquinolinyl, isothiazolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, Oxime, oxazolyl, fluorenyl, pteridinyl, fluorenyl, pyranyl, pyridyl, pyrazolyl, fluorenyl, pyrido[3,4-b]decyl, pyridine , pyrimidinyl, pyrrolyl, quinolyl, quinolyl, quinoxalinyl, thiadiazolyl, thiatriazo
  • oxo refers to a carbonyl group formed by the combination of a carbon atom and an oxygen atom.
  • Prodrugs and solvates of the compounds of the invention are also contemplated.
  • prodrug refers to a pre-drug a compound that, after being administered, is chemically converted in the body by metabolic or chemical processes (for example, placed under physiological pH conditions or by the activity of an enzyme) to release the active drug.
  • metabolic or chemical processes for example, placed under physiological pH conditions or by the activity of an enzyme
  • Prodrugs of the invention may also include metabolic precursors of the compounds of the invention, which may not be active when administered to a subject, but may be in vivo Converted to a compound of formula (I) of the invention or a salt and/or solvate thereof. Prodrugs can also be naturally occurring or chemically synthesized compounds.
  • the compounds of formula (I) of the present invention may exist in unsolvated as well as solvated forms of pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is contemplated that the present invention encompasses all solvated and unsolvated forms.
  • the solvate of the compound of formula (I) is preferably a hydrate.
  • All stereoisomers of the compounds of the invention such as isomers which may exist due to asymmetric carbon atoms on the R substituent of the compound of formula (I), including both enantiomeric and diastereomeric forms, are within the scope of the invention The scope. All stereoisomers of the compounds of formula (I) and mixtures thereof, including racemic mixtures, are also part of the invention. In addition, all geometric isomers and positional isomers are also included. For example, if the compound of the formula (I) contains a double bond, both the cis and trans forms and mixtures thereof are also included in the scope of the present invention.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physicochemical differences by methods well known to those skilled in the art, such as chromatography and/or fractional crystallization. Separation of enantiomers can be carried out by reaction with an optically active compound, conversion of the enantiomeric mixture to a mixture of diastereomers, followed by separation of the diastereomers, followed by separate diastereoisomers. The isomer is converted (e.g., hydrolyzed) to the corresponding pure enantiomer.
  • certain of the compounds of formula (I) may be atropisomers (e.g., substituted biaryls) and are also part of the present invention.
  • pharmaceutically acceptable means that a carrier, carrier, diluent, adjuvant, and/or salt formed is generally chemically or physically compatible with the other ingredients that constitute a pharmaceutical dosage form, and is physiologically Compatible with the receptor.
  • salt and “pharmaceutically acceptable salt” refer to a compound of formula (I) or a stereoisomer thereof, or a prodrug thereof and an inorganic
  • an acid and/or a base salt formed from an organic acid and a base and also a zwitterionic salt (internal salt), and also a quaternary ammonium salt such as an alkylammonium salt.
  • These salts can be obtained directly in the final isolation and purification of the compounds. It may also be obtained by mixing a compound represented by the formula (I), or a stereoisomer thereof, or a prodrug thereof with an appropriate amount (e.g., equivalent) of a certain amount of an acid or a base.
  • These salts may be precipitated in a solution and collected by filtration, or recovered after evaporation of the solvent, or may be obtained by lyophilization after reaction in an aqueous medium.
  • Acid addition salts include, for example, hydrobromide, hydroiodide, hydrochloride, sulfate, hydrogen sulfate, nitrate, acetate
  • oxalate alginate, ascorbate, aspartate, butyrate, camphorate, camphorsulfonate, cyclopentane Propionate, digluconate, ethanesulfonate, 2-hydroxyethanesulfonate, 2-naphthalenesulfonate, nicotinate, persulfate, 3-phenylpropionate, picrate, Penic acid salt, propionate, salicylate, besylate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, toluene Acid salt, citrate, maleate, fumarate, succinate, tartrate, thiocyanate, naphthate, methanesulfonate, glucoheptonate, lactobionate, dodecyl Sulfonates, adipates, and other similar salts.
  • a basic salt for example, a salt formed when the R substituent contains an acidic moiety such as a carboxyl group or a phenolic hydroxyl group
  • an ammonium salt for example, an alkali metal salt (for example, a sodium salt, a lithium salt, and a potassium salt), and an alkaline earth metal salt (for example, a calcium salt).
  • magnesium salts salts with organic bases (such as organic amines) (eg dibenzylethylenediamine, dicyclohexylamine, sulphamine, N-methyl-D-glucosamine, tert-butylamine), and amino acids Such as arginine, a salt formed by lysine, and the like.
  • quaternary ammonium salts with nitrogen-containing alkaline agents including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and Other analogues. Further examples are found in the references of this patent, Berge, et al., J. Pharm. Sci., 66, 1-19 (1977). The compounds of formula (I) may also exist as tautomers in equilibrium, and all such forms are embraced within the scope of the invention.
  • the invention includes isotopically-labeled compounds of formula (I), said isotopically-labeled compounds being the same as the compounds listed herein, but wherein one or more of the atoms are replaced by another atom
  • the atomic mass or mass of the atom is different from the atomic mass or mass number that is common in nature.
  • Isotopes which may be introduced into the compounds of formula (I) include hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, SP 2 H, 3 H, 13 C, 14 C, 15 N, 17 0, 18 0, 31 P , 32 P, 35 S, 18 F and 36 C1.
  • Compounds of formula (I), and stereoisomers and prodrugs thereof, containing such isotopes and/or other atomic isotopes, as well as pharmaceutically acceptable salts of such compounds, stereoisomers or prodrugs, are intended to be encompassed within the scope of the invention within.
  • Certain isotopically-labeled compounds of formula (I), such as those labeled with radioisotopes such as 3 H and 14 C, can be used in tissue distribution analysis of compounds and or substrates. Since the isotope of yttrium (ie) and the isotope of carbon 14 (ie, 14 C) are relatively easy to prepare and detect, we prefer the 3 H isotope and the 14 C isotope.
  • isotopes such as deuterated (ie, 2 H)
  • the isotopically-labeled compound of formula (I) can generally be prepared by methods known to those of ordinary skill in the art, such as the replacement of non-isotopically labeled reagents with isotopically labeled reagents.
  • the compounds of the present invention are modulators of the novel estrogen receptor ER-a36 and can be used to modulate the function of ER-a36 in cells in vitro and in vivo.
  • the compounds of formula (I) according to the invention can be used Prevention and/or treatment of related diseases mediated by ER-0136, especially related oncological diseases.
  • methods of modulating the function of ER-a36 in a cell comprising administering a compound of formula (I) to a cell expressing ER-0136.
  • the cells can endogenously express ER-0136 or exogenously express ER-a36 by genetic engineering, and can simultaneously or not express other estrogen receptors (such as ER-a66, ER-a46 and ER-).
  • the cell endogenously expresses ER-a36.
  • the The cells are cancer cells that endogenously express ER-a36.
  • Cancer cells expressing ER-0136 include, but are not limited to, breast cancer cells, leukemia cells, liver cancer cells, lymphoma cells, lung cancer cells, myeloma cells, prostate cancer cells, ovarian cancer cells, endometrial cancer cells, colon cancer cells. And gastric cancer cells.
  • the cell expressing ER-0136 is a breast cancer cell, leukemia cell, liver cancer cell, lymphoma cell, endometrial cancer cell, and ovarian cancer cell endogenously expressing ER-0136.
  • Breast cancer cells expressing ER-a36 include, but are not limited to, MCF7, MDA-MB-231, and SKBR-3 cells.
  • Leukemia cells expressing ER-a36 include, but are not limited to, K562, MV-4-11, SUM159, HL-60 and Molt-4 cells.
  • Endometrial cancer cells that express ER-a36 include, but are not limited to, HeclA cells.
  • Hepatoma cells expressing ER-a36 include, but are not limited to, A2780, BEL7402, BEL7404, HEL-9204, Hep2G, Hep3B and primary liver cancer stem cell Hep-12 derived from a patient.
  • Lymphoma cells expressing ER-a36 include, but are not limited to, Daudi. Expression of endogenous ER-a36 can be increased or decreased by treatment with one or more agents.
  • the invention provides a method of making a cell expressing exogenous ER-a36.
  • the cells can be prepared by genetic engineering methods known to those of ordinary skill in the (; See Samb k, etc., Molecular Cloning, A Laboratory Manual ( 2d Ed 1989) (Cold Spring Harbor Laboratory)) 0 Briefly, a first outer prepared The ER-a36 gene is inserted into an expression vector, and the expression vector is transfected into a host cell, and then the host cell is grown in a culture medium suitable for expressing exogenous ER-0136. .
  • ER-0136 The gene sequence of human ER-0136 is disclosed in Wang et al., Biochem. Biophys. Res. Commun. 336, 1023-1027 (2005) (GenBank Accession No. BX640939).
  • Cells expressing exogenous ER-a36 may or may not express endogenous ER-a36.
  • the level of expression of endogenous or exogenous ER-a36 in the cell can be increased or decreased by treatment with one or more other agents. These agents include, for example, serum, ⁇ 2 ⁇ (7 ⁇ -estradiol), tamoxifen, and fulvestrant (ICI 182, 780).
  • the compound of the formula (I) according to the present invention can be used for the preparation of a medicament for preventing and/or treating cancer associated with ER-a36, including but not limited to anal cancer, cholangiocarcinoma, bladder cancer, bone cancer, intestinal cancer (colon cancer) , rectal cancer), brain cancer, breast cancer, carcinoid, cervical cancer, endocrine-related cancer, endometrial cancer, eye cancer, gallbladder cancer, head and neck cancer, Kaposi's sarcoma cancer, kidney cancer, laryngeal cancer, leukemia, liver cancer , lung cancer, lymphoma, melanoma, mesothelioma, myeloma, neuroendocrine cancer, esophageal cancer, ovarian cancer, pancreatic cancer, penile cancer, prostate cancer, skin cancer, soft tissue sarcoma cancer, spinal cord cancer, stomach cancer, testicular cancer, Thyroid cancer, vaginal cancer, vulvar cancer or uterine cancer.
  • the cancer associated with ER-a36 includes breast cancer, cervical cancer, colon cancer, endometrial cancer, leukemia, liver cancer, lymphoma, lung cancer, myeloma, ovarian cancer, prostate cancer, gastric cancer , pancreatic cancer, kidney cancer, melanoma, thyroid cancer, soft tissue sarcoma cancer, or uterine cancer.
  • the cancer associated with ER-a36 includes breast cancer, liver cancer, lymphoma, prostate cancer, gastric cancer, lung cancer, colon cancer, pancreatic cancer, endometrial cancer, ovarian cancer, and leukemia.
  • the subject may be a mammal, such as a dog, cat, cow, sheep, horse or human, preferably a human.
  • the necessary therapeutic amount of the drug will vary depending on the particular disease and can be readily determined by one of ordinary skill in the art having the benefit of the present disclosure.
  • one or more compounds of the invention may be used in combination with one another.
  • the compounds of the invention may be used in combination with any other active agent for the preparation of a medicament or pharmaceutical composition that modulates cellular function or treats a disease. If a group of compounds is used, the compounds can be administered to the subject simultaneously, separately or sequentially.
  • the compounds of the invention may be used in combination with one or more other anticancer agents.
  • Anticancer agents that can be used include, but are not limited to, alkylating agents, nitrogen mustards, folic acid antagonists, guanidine antagonists, pyrimidine antagonists, spindle toxins, topoisomerase inhibitors, apoptosis inducers, blood vessels Production inhibitors, ghost white toxin, nitrosourea, antimetabolites, protein synthesis inhibitors, kinase inhibitors, antiestrogens, cisplatin, carboplatin, interferon, asparaginase, leuprolide, Flutamide, megestrol, mitomycin, bleomycin, doxorubicin, irinotecan and paclitaxel.
  • the anticancer agent is an antiestrogen drug, such as tamoxifen and fulvestrant (ICI 182, 780).
  • a compound of formula (I), or a stereoisomer or prodrug thereof, or a pharmaceutically acceptable salt of a stereoisomer or prodrug of said compound may be administered as a medicament A form of the composition comprising a pharmaceutically acceptable carrier, carrier or diluent. They are also useful in the preparation of a medicament for the prevention and/or treatment of a disease associated with ER-0136 in a subject.
  • the pharmaceutical compositions of the invention are also useful for treating diseases in animals.
  • a general veterinarian can administer a compound of the invention, or a veterinary salt thereof, or a veterinary solvent or a prodrug thereof, in a suitable acceptable formulation, based on experience in the art. The veterinarian can determine the most appropriate dosage and route of administration for an animal. If a plurality of active compounds are administered in combination, the active compounds may be administered sequentially, separately or sequentially in a certain order.
  • Method of Preparing Compounds The compounds of formula (I) can be prepared by various synthetic methods. A typical preparation process is listed below. Unless otherwise indicated, the definitions of 1 ⁇ , 1 2 , 1 3 , 1 4 , 1 5 , 16 are as previously described. It will be apparent to those skilled in the art that the exact method of preparation of the compounds can vary slightly depending on the chemical structure.
  • the compound of the formula (I) can be produced in several steps.
  • Compound L is prepared from compound i_ and catalyzed by a Lewis acid by a Houben-Hersch reaction (modified Friedel-Glysylation).
  • Lewis acids suitable for the reaction include, for example, anhydrous zinc chloride, anhydrous aluminum trichloride, ferric chloride, titanium tetrachloride, tin chloride, boron trifluoride etherate complex and the like.
  • the reaction is usually carried out at a temperature of from about 0 ° C to about 120 ° C for 1 to 20 hours.
  • Compound X can be prepared by condensation of compound L with various substituted compounds ⁇ in an inert solvent.
  • Suitable inert solvents for this reaction include ethers such as DME, 1,2-diethoxyethane, THF, 1,4-dioxane, DMF, hydrazine, hydrazine-dimethylacetamide, pyridine, hydrazine -Methyl-2-pyrrolidone.
  • the reaction can be carried out under basic conditions such as potassium hydroxide, potassium carbonate, cesium carbonate, sodium hydride, sodium methoxide, potassium t-butoxide, DBU 1,8-diazabicyclo-bicyclo (5, 4, 0)-7-undecene), n-butyllithium, LDA (lithium diisopropylamide), LHMDS (lithium hexamethyldisilazide) and the like.
  • a stoichiometric or catalytic amount of a phase transfer catalyst such as 18-crown-6, TBAB (tetrabutylammonium bromide), TBAF (tetrabutylammonium fluoride) or the like may be added during the reaction.
  • the reaction temperature is usually between about 0 ° C and about 140 ° C, preferably at the reflux temperature of the solvent for 1 to 20 hours.
  • the isopentenyl bromide can be reacted with the compound iL under basic conditions to prepare a compound.
  • Suitable solvents for the reaction include, for example, methanol, DMF (N,N-dimethylformamide), THF (tetrahydrofuran), water, toluene, DME (1,2-dimethoxyethane), and mixed solvents. Such as methanol-water, DMF-water, tetrahydrofuran-water and the like.
  • the solvent used in the reaction is preferably water.
  • Bases suitable for the reaction include, for example, potassium hydroxide, potassium carbonate, cesium carbonate, sodium methoxide, sodium hydride, potassium t-butoxide, DBU (1,8-diazabicyclo-bicyclo(5,4,0) -7-undecene), n-butyllithium, LDA (lithium diisopropylamide), LHMDS (lithium hexamethyldisilazide) and the like.
  • the reaction temperature is usually between about 0 ° C and about 100 ° C and the reaction time is from 1 to 20 hours.
  • Compound ill can be prepared by reacting compound ill with isopentenyl bromide under basic conditions.
  • suitable solvents for the reaction include, for example, methanol, DMF (N,N-dimethylformamide), THF (tetrahydrofuran), water, toluene, DME (1,2-dimethoxyethane), and mixed solvents.
  • methanol-water, DMF-water, tetrahydrofuran-water and the like The solvent used in the reaction is preferably water.
  • Bases suitable for the reaction include, for example, potassium hydroxide, potassium carbonate, cesium carbonate, sodium methoxide, sodium hydride, potassium t-butoxide, DBU (1,8-diazabicyclo-bicyclic (5,4,0) -7-undecene), n-butyllithium, LDA (lithium diisopropylamide), LHMDS (lithium hexamethyldisilazide) and the like.
  • the reaction temperature is usually between about 0 ° C and about 100 ° C and the reaction time is from 1 to 20 hours.
  • Compound ⁇ can be prepared by condensation of compound IL with various substituted acid chlorides iii in an inert solvent.
  • Suitable inert solvents for this reaction include ethers such as DME, 1,2-diethoxyethane, THF, 1,4-dioxane, DMF, hydrazine, hydrazine-dimethylacetamide, pyridine, hydrazine -Methyl-2-pyrrolidone.
  • the reaction can be carried out under basic conditions, such as potassium hydroxide, potassium carbonate, cesium carbonate, sodium hydride, sodium methoxide, potassium t-butoxide, DBU 1,8-diazabicyclo-bicyclo (5,4,0 )-7-undecene), n-butyllithium, LDA (lithium diisopropylamide), LHMDS (lithium hexamethyldisilazide) and the like.
  • a stoichiometric or catalytic amount of a phase transfer catalyst such as 18-crown-6, TBAB (tetrabutylammonium bromide), TBAF (tetrabutylammonium fluoride), or the like may be added during the reaction.
  • the temperature of the reaction is usually between about 0 ° C and about 140 ° C, preferably at the reflux temperature of the solvent for 1 to 20 hours.
  • R 1 of the compound of formula (I) is hydrogen, it can be prepared according to the following embodiment
  • P is a protecting group for a hydroxyl group.
  • the compound is removed by removing the protecting group of the compound.
  • the method of removal varies according to the nature of the protection. See mainly "Protective Groups in Organic Synthesis” (Greene T. W et al. John Wiley & Sons, New York, 1991).
  • Preferred protecting groups are benzyl, benzoyl, benzyloxycarbonyl, TBDMS (tert-butyldimethylsilyl), THP (tetrahydropyranyl), methyl, MOM (methoxymethyl), PMB (p-methoxybenzyl) and the like.
  • the compound siL is reacted with isopentenyl bromide under basic conditions to prepare a compound.
  • Suitable solvents for the reaction include, for example, methanol, DMF (N,N-dimethylformamide), THF (tetrahydrofuran), water, toluene, DME (1,2-dimethoxyethane), and mixed solvents.
  • methanol-water, DMF-water, tetrahydrofuran-water and the like Such as methanol-water, DMF-water, tetrahydrofuran-water and the like.
  • the solvent used in the reaction is preferably water.
  • Bases suitable for the reaction include, for example, potassium hydroxide, potassium carbonate, cesium carbonate, sodium methoxide, sodium hydride, potassium t-butoxide, DBU (1,8-diazabicyclo-bicyclo(5,4,0) -7-undecene), n-butyllithium, LDA (lithium diisopropylamide), LHMDS (lithium hexamethyldisilazide) and the like.
  • the reaction temperature is usually between about 0 ° C and about 100 ° C and the reaction time is from 1 to 20 hours. detailed description
  • room temperature or ambient temperature means in the range of 18-25 ° C
  • the solvent is removed using a rotary evaporator under reduced pressure.
  • the reaction process was monitored by thin layer chromatography (TLC).
  • TLC thin layer chromatography
  • the reaction time is for illustrative purposes only.
  • the structure and purity of the compound are confirmed by at least one of the following techniques: TLC, mass spectrometry, nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC).
  • the yields are for illustrative purposes only.
  • Step 2 Preparation of 2-(4-trifluoromethylphenyl)-3-methoxy-5,7-dihydroxy-4H-benzopyran-4-one 2-methoxy-1-( 2,4,6-Trihydroxyphenyl)ethanone (30 g, 151 mmol) and 4-trifluoromethylbenzoyl chloride (37.5 g, 180 mmol) dissolved in 250 mL of dry pyridine, DBU was added dropwise at room temperature (53.2, 350 mmol). After the dropwise addition was completed, the reaction system was heated to 75 ° C, and the reaction was stirred overnight. The reaction mixture was cooled to room temperature, and most of the solvent was removed under reduced pressure, and the residue was poured into ice-cooled diluted hydrochloric acid.
  • reaction system was reacted at room temperature overnight, and the pH was adjusted to 6 with 2N hydrochloric acid. Right, then extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution and dried over anhydrous sodium sulfate.
  • Step 2 Preparation of 2-(3,4-difluorophenyl) -3 -Methoxy-5,7-dihydroxy-8-(3-methyl-2-buten-1-yl)-4H-benzopyran-4-one (Compound 10) under nitrogen protection, 2-methoxy small [2,4,6-trihydroxy-3,5-bis(3-methyl-2-butenyl)phenyl]ethanone (250 mg, 0.94 mmol), anhydrous Potassium powder (779 mg, 5.63 mmol), TBAB (tetrabutylammonium bromide, 454 mg, 1.41 mmol) and 3,4-difluorobenzoyl chloride (331 mg, 1.88 mmol) in toluene (GO mL) , reflux reaction for 6 hours.
  • Compound 10 2-methoxy small [2,4,6-trihydroxy-3,5-bis(3-methyl-2-butenyl)phenyl]ethanone (250 mg, 0.94 mmol)
  • Example 14 Expression of ER- ⁇ variants in human breast cancer samples
  • a film pre-coated with human breast cancer cell tissue was purchased from ProSci Incorporated (Poway, CA).
  • the membrane was detected with an anti-ER-a36 antibody and an HRP-conjugated secondary antibody which specifically recognized ER-a36, and was visualized with an enhanced chemical luminescence (ECL) detector (available from Amersham Pharmacia Biotech).
  • ECL enhanced chemical luminescence
  • the label on the same membrane was eluted, and the anti-estrogen receptor-a antibody H222 (Novocastra Laboratories Ltd, UK), which recognizes all three subtypes of ER- ⁇ , ER-a66, ER-a46 and ER-a36, was used. Provide) for testing.
  • Figure 1 shows that three estrogen receptor subtypes of ER-a66, ER-a46 and ER-a36 are not expressed in normal breast tissue (lane 1), but they are in a sample of invasive ductal carcinoma (lane 2). , a sample of invasive lobular carcinoma (lane 5) and non-invasive ductal carcinoma (lane 7) were expressed.
  • ER-a36 was expressed in another sample of invasive ductal carcinoma (lane 4) and invasive lobular carcinoma (lane 6).
  • Dao 2 and Dao 3 are invasive ductal carcinoma tissues from 2 different patients.
  • Lanes 5 and 6 are invasive lobular carcinoma tissues from two different patients, respectively.
  • Example 15 Expression of ER-a36 in ER-negative breast cancer cell line MDA-MB-231
  • MDA-MB-231 is a well-known cell line lacking ER-a66 and ER-a46 (see Relevance of breast cancer cell lines as models for breast tumours: an update. Marc Lacroix, Guy Leclercq, Brest Cancer Research and Treatment 83: 249-289 (2004)).
  • MDA-MB-231 cells are from the American Type Culture Collection (ATCC). Place MDA-MB-231 cells in Dulbecco's Modified Eagle Medium (DMEM) and 10% fetal bovine blood The clear 8-well BIOCOAT slide (supplied by BD Science Discovery Labware) was incubated at 37 ° C and 5% CO 2 atmosphere for 12 hours.
  • DMEM Dulbecco's Modified Eagle Medium
  • BIOCOAT slide supplied by BD Science Discovery Labware
  • the cells were then washed twice with sterile phosphate buffered saline (PBS) and fixed with 4% paraformaldehyde in PBS (pH 7.4) for 30 minutes at room temperature. Thereafter, the cells were washed with PBS and ruptured with 0.5% (v/v) Triton X-100 for 10 minutes. The cells were washed again with PBS, and 3% serum in PBS was added and blocked at room temperature for 1 hour.
  • PBS sterile phosphate buffered saline
  • PBS 4% paraformaldehyde in PBS
  • Figure 2 shows that MDA-MB-231 cells were stained positive by anti-ER-a36 antibody. To further confirm the confidence of the experiment, no specific staining was shown by incubation of the anti-ER-0136 antibody pre-incubated with the immunogenic polypeptide (Fig. 2, lower panel), indicating the specificity of the antibody.
  • Example 16 Western blotting detection of ER-0136 expression on different tumor cell lines
  • the experimental cells e.g., MDB-MA-231, medium 10%-FBS-DMEM
  • the experimental cells were cultured at 37 ° C and 5% CO 2 conditions. After each well of the cells was grown to 60% to 90% fullness, the cells were collected and centrifuged at 4,300 rpm for 5 minutes at 4 °C. After removing the supernatant, an appropriate amount of lysate and Lysis buffer containing 1% ⁇ -40 and 0.7 mM EDTA were added, followed by addition of a protease inhibitor, and cleavage for 30 minutes to 1 hour under ice bath conditions. The cells were again centrifuged at 14,000 rpm for 15 minutes, and the supernatant was taken and protein quantitation was performed.
  • Lane 1 293 human renal epithelial cell lines transiently overexpressing ER-0136; lanes 2-4: human breast cancer SK-BR-3 cell lines from different laboratory sources; lanes 5-7: human mammary glands from different laboratory sources Cancer MCF-7 cell line; Lane 8-9: Human leukemia HL-60 cell line from different laboratory sources; Lane 10-11: Human leukemia MV-4-11 cell line from different laboratory sources; Lane 12-13: Different laboratory-derived human chronic myeloid leukemia K562 cells; tract 14: human liver cancer A2780 cells; tract 15: human liver cancer HEL-7402 cells; tract 16: human liver cancer HEL-9204 cells; tract 17: primary cells derived from patients Liver cancer Hep-11 cells; Lane 18: Primary liver cancer Hep-12 cells derived from patients.
  • Example 17 Inhibition of compound growth on different breast cancer cells in vitro
  • MDA-MB-231 cells were stored at 10% under 37 ° C and 5% CO 2 atmosphere. Fetal bovine serum in DMEM. The cells were seeded at a density of 6 x 10 3 /well in 96-well plates. The test compound was dissolved in DMSO and applied to MDA at a concentration of 0, 0.3 ⁇ , 0.5 ⁇ , 1 ⁇ , 2 ⁇ , 3 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ , 30 ⁇ , 50 ⁇ and 100 ⁇ . MB-231 cells were 72 hours. The CellTiter-Glo® Luminescence Cell Viability Assay Kit (Promega) was then used to detect cells treated with the compound and the luminescence values were recorded using Envision.
  • MCF7 cells are breast cancer cell lines that highly express ER-66, ER-46 and ER-36. (Relevance of breast cancer cell lines as models for breast tumours: an update. Marc Lacroix, Guy Leclercq, Breast Cancer Research and Treatment (2004) 83, 249-289; Wang et al., Proc. Natl. Acad. Sci. USA103 :9063-9068 (2006)).
  • MCF7 cells were derived from ATCC and maintained at 37 ° C, 5% CO 2 atmosphere and in Dulbecco's Modified Eagle Medium (DMEM) and 10% fetal bovine serum.
  • DMEM Dulbecco's Modified Eagle Medium
  • the cells were seeded at a density of 6 x 10 3 /well in 96-well plates.
  • the test compound was dissolved in DMSO and applied to MCF7 cells at a concentration of 0, 0.3 ⁇ , 0.5 ⁇ , 1 ⁇ , 2 ⁇ , 3 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ , 30 ⁇ , 50 ⁇ and 100 ⁇ . 72 hours.
  • the CellTiter-Glo® Luminescence Cell Viability Assay Kit (Promega) was then used to detect cells treated with the compound and the luminescence values were recorded using Envision.
  • MDA-MB-231 cells MCF7 cells tamoxifen a) 20.90 ⁇ 1.51 ) 22.55 ⁇ 4.15
  • NA stands for no activity and its IC 5Q value is greater than 100 ⁇ .
  • Leukemia K562 was obtained from ATCC and maintained at 37 ° C, 5% C0 2 atmosphere and in IMDM and 10% fetal bovine serum. The cells were seeded at a density of 6 x 10 3 /well in 96-well plates. The test compound was dissolved in DMSO and applied to ⁇ 562 cells at a concentration of 0, 0.3 ⁇ , 0.5 ⁇ , 1 ⁇ , 2 ⁇ , 3 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ , 30 ⁇ , 50 ⁇ and 100 ⁇ . 72 hours. The CellTiter-Glo® Luminescent Cell Viability Assay Kit (Promega) was then used to detect cells treated with the compound and the luminescence values were recorded using Envision.
  • Example 19 Inhibition of Compound Growth on Human B Lymphoma Daudi Cells in Vitro
  • Human B lymphoma Daudi cells were obtained from ATCC and maintained at 37 ° C, 5% CO 2 atmosphere and in IMDM and 10% fetal bovine serum. The cells were seeded at a density of 6 x 10 3 /well in 96-well plates. The test compound was dissolved in DMSO and applied to ⁇ 562 cells at a concentration of 0, 0.3 ⁇ , 0.5 ⁇ , 1 ⁇ , 2 ⁇ , 3 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ , 30 ⁇ , 50 ⁇ and 100 ⁇ . 72 hours. The CellTiter-Glo® Luminescence Cell Viability Assay Kit (Promega) was then used to detect cells treated with the compound and the luminescence values were recorded using Envision. The results of in vitro effects of the partial compounds of the present invention on the cell viability of leukemia K562 cells and human B lymphoma Daudi cells are listed in Table 4 below.
  • Gleevec and cytarabine are positive control compounds for the K562 cell model and the Daudi cell model, respectively.
  • ND stands for no measurement.
  • NA stands for no activity and its IC 5Q value is greater than 100 ⁇ .
  • Example 20 Inhibition of in vitro growth of compounds on acute leukemia cells
  • Acute myeloid leukemia cell HL-60 was obtained from ATCC and maintained at 37 ° C, 5% C0 2 atmosphere and in IMDM and 10% fetal bovine serum. The cells were seeded at a density of 6 x 10 3 /well in 96-well plates. Test compounds dissolved in DMSO, at the concentration of 0, 10_ 4 M, 10_ 5 M, 10_ 6 M, 10 "7 M, 10_ 8 M acting on the HL-60 cells for 72 hours. Then MTT assay was used to detect the OD Value, the percentage of inhibition of cell proliferation was calculated.
  • Example 21 Inhibition of growth of compounds on hepatoma cells in vitro
  • Hepatoma cell line HL-60 was obtained from ATCC and maintained at 37 ° C, 5% C0 2 atmosphere and in DMEM and 10% 10% NCS and 5 ( ⁇ g/ml KANA. Cells were seeded at a density of 6 x 10 3 /well In a 96-well plate, the test compound was dissolved in DMSO at a concentration of 0, 10" 4 M, 10" 5 M, 10" 6 M, 10" 7 M, 1 (T 8 M was applied to BEL-7402 The cells were subjected to 72 hours. Then, the OD value was measured by the SRB method, and the percentage of inhibition of cell proliferation was calculated. The results of the growth inhibitory effect of some of the compounds of the present invention on liver cancer cells are shown in Table 6.
  • the gastric cancer cell BGC-823 was seeded at a density of 3 ⁇ 10 3 /well in a 96-well plate and maintained in a DMEM medium containing 2.5% CS-FBS in phenol red at 37 ° C in a 5% CO 2 atmosphere. 24 hours.
  • the test compound was dissolved in DMSO and applied to BGC-823 cells at a concentration of 0, 1, 2, 4, 8, 10, 20 ⁇ for 72 hours. The OD value was then measured by the MTT method to calculate the percentage of inhibition of cell proliferation. The results are shown in Figure 4.
  • Example 23 Inhibition of growth of compounds on lung cancer cells in vitro
  • Lung cancer cells H460 were seeded at a density of 4.0 ⁇ 10 3 /well in 96-well plates, and cultured in a phenol red-free 1640 medium containing 2.5% CS-FBS at 37 ° C in a 5% CO 2 atmosphere. hour.
  • the test compound was dissolved in DMSO and applied to H460 cells at a concentration of 0, 1, 2, 4, 8, 10, 20 ⁇ for 72 hours. Then, the OD value was used to detect the OD value, and the percentage of inhibition of cell proliferation was calculated. The results are shown in Figure 5.
  • Example 24 Inhibition of in vitro growth of compounds on colon cancer cells
  • Colon cancer cells LS174T were seeded at a density of 4.5 ⁇ 10 3 /well in 96-well plates and maintained in a phenol red-free 1640 medium containing 2.5% CS-FBS at 37 ° C in a 5% CO 2 atmosphere. 24 hours. Test compounds were dissolved in DMSO and applied to LS174T cells for 72 hours at concentrations of 0, 1, 2, 4, 8, 10, 20 ⁇ M. Then, the OD value was used to detect the OD value, and the percentage of inhibition of cell proliferation was calculated. The results are shown in Figure 6.
  • Example 25 Inhibition of compound growth on pancreatic cancer cells in vitro
  • Prostate cancer cell line PC-3 was seeded in a 96-well plate at a density of 3 ⁇ 10 3 cells/well, and incubated with F12K medium containing 10% fetal calf serum for 24 hours at 37 ° C in a 5% CO 2 atmosphere.
  • the test compound was dissolved in DMSO and applied to PC-3 cells at a concentration of 0, 1, 2, 4, 8, 10, 20 ⁇ for 72 hours. Then, the OD value was used to detect the OD value, and the percentage of inhibition of cell proliferation was calculated. The results are shown in Figure 8.
  • In Vivo Evaluation Example 27 Growth Inhibition of Compounds on Human Breast Cancer BCAP-37 Cell Transplanted Tumor in Nude Mice
  • Test compounds were administered to nude mice bearing breast cancer xenografts to test their effect of inhibiting tumor growth.
  • Tumor tissues were extracted from nude mice bearing BCAP-37 cells and cut into small pieces. Several pieces of tumor tissue were implanted into the armpit of the right forelimb of female nude mice. After implantation, the ⁇ 2 ⁇ solution was injected once a day for 6 days in a dose of 7 ⁇ ⁇ per nude mouse to stimulate the growth of the tumor in the mice. From day 7 onwards, the tumor-bearing nude mice were orally administered with a corn oil solution of the test compound at a dose of 35 mg/kg.
  • Example 28 Growth inhibition of compound on human B lymphoma Daudi cell xenografts in nude mice
  • Test compounds were administered to nude mice bearing human B lymphoma Daudi cell xenografts to test their effect of inhibiting tumor growth.
  • Human B lymphoma Daudi cells are derived from ATCC. After 5 passages, lx lO 7 cells were added to 0.2 mL of Matrigel and implanted subcutaneously into the armpit of the right forelimb of male nude mice. When tumors in nude mice reached 150-200 mm 3 , the tumor-bearing nude mice were randomly divided into groups of ten. The mixed oil gavage administration group was used as a negative control, and rituximab was intravenously administered as a positive control group, and the test compound was dissolved in a mixed oil for intragastric administration.
  • the administration period was 21 consecutive days, and a mixed oil suspension (35 mg/kg) of the test compound was intragastrically administered to the test group nude mice every day.
  • the rituximab was administered to the positive control group twice a week.
  • Anti-(20 mg/kg) while daily intragastric administration of negative control group, blank mixed oil solvent Times.
  • Tumor volume and body weight of nude mice were measured twice weekly during the dosing cycle. Tumor growth curves were plotted as tumor volume and time of administration ( Figure 10) to assess the inhibitory effect of compounds on tumor growth.
  • Example 29 Growth inhibition of compound on human endometrial carcinoma Ishikawa cell xenografts in nude mice
  • Test compounds were administered to nude mice bearing endometrial cancer xenografts to test their effect of inhibiting tumor growth.
  • Tumor tissues were extracted from nude mice bearing Ishikawa cells and cut into small pieces. Several pieces of tumor tissue were implanted into the armpit of the right forelimb of female nude mice. After implantation, the ⁇ 2 ⁇ solution was injected once a day for 6 days in a dose of 7 ⁇ ⁇ per nude mouse to stimulate the growth of the tumor in the mice. From the 7th day, the tumor-bearing nude mice were orally administered with a mixed oil solution of the test compound at a dose of 35 mg/kg.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyrane Compounds (AREA)

Abstract

本发明提供了一类具有式(I)结构的多羟基苯并吡喃酮类化合物及其药学上可接受的盐或前药,以及含有该类化合物的药物组合物。所述化合物可用于调控新型雌激素受体ER-α36,能用于预防和/或治疗由ER-α36受体介导的相关肿瘤疾病,如乳腺癌,白血病和肝癌等。

Description

多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用 技术领域
本发明涉及多羟基苯并吡喃酮类化合物及其盐或前药, 以及含有这类化合物的药物组合 物, 它们在用于制备预防和 /或治疗肿瘤疾病的药物中的用途。 背景技术
雌激素是与人体中许多关键生理功能有关的一组激素。 雌激素的功能包括促进女性性器 官发育、 为怀孕时的乳腺和子宫以及分娩后的母乳喂养做好充分准备。 雌激素在维持适当的 心血管功能及骨密度方面也起着重要的作用。 众所周知, 雌激素可以剌激细胞增殖, 由此可 能增加女性患上癌症的危险, 特别是乳腺癌与子宫癌。 雌激素通过与靶细胞中的雌激素受体相结合来调节细胞功能。 在人体细胞中已发现两种 雌激素受体 (ERs), ER-α和 ER-P。 他们具有相似的蛋白质结构, 每种均拥有三个独立却相 互作用的功能结构域: N末端结构域 (A/B结构域)、 中段 DNA结合结构域 (C结构域) 以 及 C末端配体结合结构域 ( D/E/F结构域)。 N末端结构域具有非配体依赖性激活功能 (AF-1 ), 可与共激活因子相互作用, 在缺乏配体情况下转录激活靶基因。 DNA结合结构域在受体二聚 化以及与特定 DNA序列结合方面具有重要作用。 C末端配体结合域可介导配体结合并具有配 体依赖性转录激活功能 (AF-2), 可在配体存在时激活基因转录。 全长的 ER-α是分子量为 66 kDa的蛋白, 被称为 ER-a66。 ER-a66包含全部三种功能结 构域。 后来人们又发现了 ER-a66的剪接变异体, 将其命名为 ER-a46。 ER-a46的分子量约为 46 kDa, 其缺少 hER-a66的 N末端 AF-1结构域。 最近又发现了一个新的 36 kDa的 ER-α变 异体, ER-a36。它缺少 ER-a66的 N末端 AF-1结构域及 C端 AF-2结构域(参见 wang等人, Biochem. Biophys. Res. Commun. 336, 1023-1027, (2005 。 通常认为 ER-a66 通过转录激活其靶基因来介导雌激素剌激的细胞增殖。 雌激素与 ER-a66的结合可激活 ER-a66的转录激活结构域, 从而剌激下游靶基因的表达, 并最终导致 细胞增殖。 ER-a46被证明可介导由膜启动且受雌激素剌激的快速 NO合成 (参见 Li等人, Proc. Natl. Acad. Sci. USA 100: 4807-4812, (2003))。 并且人们也发现缺失 AF-1 结构域的 ER-a46会抑制 ER-a66的 AF-1活性 (参见 Flouriot, G., EMBO, 19, 4688-4700, (2000))。 由 于 ER-a36缺失 AF-1和 AF-2转录激活结构域, 可将其作为显性负面抑制剂来抑制 ER-a和 ER-β的 AF-1和 AF-2功能。 另外, ER-a36主要分布在细胞膜上, 并且介导膜引发的促有丝 分裂雌激素信号的传导, 所述雌激素信号传导剌激细胞增殖。 (参见 wang 等人, Biochem. Biophys. Res. Commun. 336, 1023-1027, (2005); wang等人, Proc. Natl. Acad. Sci. USA 103: 9063-9068, (2006) ) 深入研究表明, 雌激素信号是通过传统的细胞核转录激活通路与非传统的膜启动的信号 传导通路来进行介导。 ER-a66和 ER-a46似乎主要在细胞核内起作用, 而 ER-a36似乎主要 通过在细胞核外起作用。 另据显示, ER-a36缺少原始 ER-a66带有的配体结合结构域的螺旋 8-12, 这完全改变了 ER-a36配体结合的特异性。 因此, ER-a36可能与 ER-a66和 ER-β结合不同的配体。 由于与雌激素受体相关的疾病仍然影响着许多人, 目前急需找到一种新型的, 用于预防 和 /或治疗这些相关疾病的化合物及其药物组合物。 发明内容
本发明提供了可用于调控新型雌激素受体 ER-a36的功能的一类多羟基苯并吡喃酮类化 合物及其盐或前药, 以及含该 式 (I) 所示:
Figure imgf000004_0001
其中:
R1可选自氢, (C C6)烷基, 含有一个或多个卤原子取代的 (C C6)烷基;
R2, R3, R4, R5和 R6可独立选自氢, (d-C4)烷基, 含有一个或多个卤原子取代的 (d-C4) 烷基, 卤素, 氰基, 含有一个或多个卤原子取代的 (d-C6)烷氧基;; 但 R2, R3, R4, R5和 R6 不可同时为氢;
且当 R1为甲基, R3和 R5为氢时, R4不可为氯原子。 附图说明
图 1显示了 ER-a66、 ER-a46和 ER-a36在人乳腺癌样品中表达的蛋白免疫印迹结果。 道 1 : 正常乳腺组织; 道 2: 浸润性导管癌; 道 3: 浸润性导管癌; 道 4: 侵袭性导管癌; 道 5: 浸润性小叶癌; 道 6: 浸润性小叶癌; 道 7: 非侵袭性导管癌。 图 2(上图)为 MDA-MB-231细胞的免疫荧光染色结果。 MDA-MB-231细胞是缺乏 ER-a66 和 ER-a46的 ER阴性乳腺癌细胞系。用与 ER-a36特异性结合的抗体对 MDA-MB-231细胞进 行染色 (图中标为 "ER-a36 Ab"的左图: 正染色显示为绿色)。 用 4,6-二脒 -2-苯基吲哚对细胞 核进行染色。 (图中标为" DAPI' '的中图: 正染色显示为蓝色)。合并的染色信号被标为"合并"。 当将所述抗体与结合到所述抗体上的免疫原多肽预温育时, 结果显示为阴性 (下图)。 图 3 显示了不同肿瘤细胞株表达 ER-0136的蛋白免疫印迹结果。道 1 :瞬时过表达 ER-0136 的 293人肾上皮细胞株; 道 2-4: 不同实验室来源的人乳腺癌 SK-BR-3细胞株; 道 5-7: 不同实验室来源的人乳腺癌 MCF-7细胞株; 道 8-9: 不同实验室来源的人白血病 HL-60 细胞株; 道 10-11 : 不同实验室来源的人白血病 MV-4-11细胞株; 道 12-13 : 不同实验 室来源的人慢性粒细胞白血病 K562细胞; 道 14: 人肝癌 A2780细胞; 道 15 : 人肝癌 HEL-7402细胞; 道 16: 人肝癌 HEL-9204细胞; 道 17: 来源于病人的原代肝癌 Hep-11 细胞; 道 18: 来源于病人的原代肝癌 Hep-12细胞。 图 4至图 8分别是用 MTT法检测化合物 1 在体外对胃癌 BGC-823细胞、肺癌 H460 细胞、 结肠癌 LS174T细胞、 胰腺癌 PANC-1和前列腺癌 PC-3细胞的生长抑制作用。 结果显示化合物 1 对这些肿瘤细胞都有显著的生长抑制作用, 且呈较好的剂量依赖关 系。 IC50在 1~4 μΜ。 图 9显示了当分别以阳性对照他莫昔芬 (0.7mg/鼠 /天), 化合物 1 (0.7 mg/鼠 /天) 以及 阴性对照空白溶媒(0.2 mL/鼠 /天)对荷瘤人乳腺癌 BCAP-37瘤株裸鼠连续给药 20天后的平 均瘤重 (直方图 a), 显示了化合物的肿瘤生长的抑制作用。 图 10显示了当分别以利妥昔单抗为阳性对照, 以化合物 1 (0.7 mg/鼠 /天) 以及阴性对 照空白溶媒 (0.2 mL/鼠 /天) 对荷瘤人 B淋巴瘤 Daudi细胞裸鼠连续给药 21天的肿瘤的生长 曲线图。 图 11显示了当分别以阳性对照醋酸甲孕酮( 120 mg/kg),和化合物 1的低( 17.5 mg/kg)、 中 (35 mg/kg)、 高剂量 (70 mg/kg)以及阴性对照空白溶媒 (0.2 mL/鼠 /天) 对荷瘤人子宫内膜 癌 Ishikawa瘤株裸鼠连续给药 20天后的平均瘤重。化合物 1对荷瘤小鼠的肿瘤生长有显著地 抑制作用, 且优于阳性对照药物。 发明详述
化合物及其衍生物 在本发明的某些实施方式中提供了一类苯并吡喃酮类化合物及其药学上可接受的盐或 前药, 以及含该类化合物的药物组合物, 它们可用于调控新型雌激素受体 ER-a36的功能, 预 防和 /或治疗由 ER-0136受体介导的疾病, 如癌症, 等。
在某些实施方式中, 本发明提供了式 (I) 的化合物:
Figure imgf000006_0001
及其药学上可接受的盐、 前药或药物组合物, 其中:
R1可选自氢, (C C6)烷基, 含有一个或多个卤原子取代的 (C C6)烷基;
R2, R3, R4, R5和 R6可独立选自氢, (d-C4)烷基, 含有一个或多个卤原子取代的 (d-C4) 烷基, 素, 氰基, 含有一个或多个 原子取代的 (d-C4)烷氧基, 但 R2, R3, R4, R5和 R6 不可同时为氢; 且当 R1为甲基, R3和 R5为氢时, R4不可为氯原子。 本发明的一个实施方式, 包括一组带式 (I) 结构的化合物, 称为化合物 (Π) , 其所述 化合物具有下式:
Figure imgf000006_0002
其中:
R2, R3, R4, R5和 R6可独立选自氢, (CrC4)烷基, 含有一个或多个卤原子取代的 (CrC4) 烷基, 素, 氰基, 含有一个或多个 原子取代的 (CrC4)烷氧基; 但 R2, R3, R4, R5和 R6 不可同时为氢。 本发明的一个实施方式, 包括一组带式 (I) 结构的化合物, 称为化合物 (ΠΙ) , 其所述 化合物组具有下式:
Figure imgf000007_0001
( III ) 其中:
R2, R3, R4, R5和 R6可独立选自氢, (d-C4)烷基, 含有一个或多个卤原子取代的 (d-C4) 烷基, 素, 氰基, 含有一个或多个 原子取代的 (d-C4)烷氧基; 但 R2, R3, R4, R5和 R6 不可同时为氢。
且当 R3和 R5为氢时, R4不可为氯原子。
本发明特别优选的式 (I) 结构的化合物包括但不限于下列化合物:
2-(4-三氟甲基苯基 )-3,5,7-三羟基 -8-P-甲基 -2-丁烯 -1-基) - 4H-苯并吡喃 -4-酮;
2-(4-氟苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3-氟 -4-氯苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(4-氯苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(4-三氟甲氧基苯基) -3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3,4-二氯苯基) -3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3-三氟甲基 -4-氯苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基) - 4H-苯并吡喃 -4-酮;
2-(4-溴苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3,4-二氟苯基) -3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮; 2-(4-三氟甲基苯基 )-3-甲氧基 -5,7-二羟基 -8-P-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮; 2-(4-三氟甲氧基苯基) -3-甲氧基 -5,7-二羟基 -8 3-甲基 -2-丁烯 -1-基) - 4H-苯并吡喃 -4-酮; 2-(3-三氟甲基 -4-氯苯基 )-3-甲氧基 -5,7-二羟基 -8-P-甲基 -2-丁烯 -1-基) - 4H-苯并吡喃 -4- 酮;
2-(4-溴苯基 )-3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯小基)- 4H-苯并吡喃 -4-酮。 本文中提供的化合物和衍生物可以根据 IUPAC (国际纯粹与应用化学联合会)或 CAS (化 学文摘服务社, Columbus, OH)命名系统命名。 以下是本说明书中使用的术语的定义。 除非另外说明, 本文中为基团或术语提供的初始 定义适用于整篇说明书的该基团或术语, 无论是单独的或是作为另一基团的一部分。
"取代"是指分子中的氢原子被其它不同的原子或分子所替换。 替换氢原子的原子或分子 称之为 "取代基"。 碳氢基团中碳原子含量的最小值和最大值通过前缀表示, 例如, 前缀 (Ca-Cb)烷基表明任 何含 "a"至" b"个碳原子的烷基。 因此, 例如, (d-Q烷基是指包含 1-6个碳原子的烷基。 术语"烷氧基 "是指与一个氧原子键合的直链或带有支链的、 单价的、 饱和脂肪链, 包括 但不限于如甲氧基、 乙氧基、 丙氧基、 丁氧基、 异丁氧基、 叔丁氧基以及其它类似基团。 术语"烷基"指直链或带有支链的、 单价的、 饱和脂肪链, 包括但不限于如, 甲基、 乙基、 丙基、 异丙基、 丁基、 异丁基、 戊基、 异戊基、 己基以及其它类似基团。 术语"卤素"或"卤原子 "是指氯、 溴、 氟和碘原子或基团。 术语"杂芳基 "是指其中一个或多个碳原子已被如氮、 氧或硫等杂原子取代的单环或多环 芳香烃基团。 如果杂芳基含有不止一个杂原子, 则这些杂原子可能相同, 也可能是不同的。 杂芳基包括但不限于如苯并呋喃基、 苯并噻吩基、 苯并咪唑基、 苯并噁唑基、 苯并噻唑基、 苯并吡喃基、 呋喃基、 咪唑基、 吲唑基、 吲嗉基、 吲哚基、 异苯并呋喃基、 异吲哚基、 异喹 啉基、 异噻唑基、 异噁唑基、 萘啶基、 噁二唑基、 噁嗉基、 噁唑基、 酞嗉基、 蝶啶基、 嘌吟 基、 吡喃基、 吡嗉基、 吡唑基、 哒嗉基、 吡啶并 [3,4-b]吲哚基、 吡啶基、 嘧啶基、 吡咯基、 喹嗉基、 喹啉基、 喹喔啉基、 噻二唑基、 噻三唑基、 噻唑基、 噻吩基、 三嗉基、 三唑基、 咕 吨基以及其它类似基团。 术语"氧代"是指碳原子和氧原子结合形成的羰基基团。 本发明化合物的前药和溶剂化物也在考虑之内。 术语"前药"是指一种作为药物前体的化 合物, 该化合物在被服用后, 在体内通过代谢或化学过程 (例如, 被置于生理 pH条件下或通 过酶的活性)发生化学转化, 释放活性药物。 关于前药的合成与使用的讨论, 参见 T. Higuchi 禾口 W. Stella的文章: "Prodrugs as Novel Delivery Systems" vol. 14 of the ACS Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987。这两篇文章的内容均被引用入本文中。本发明的"前药" 也可包括本发明化合物的代谢前体, 该类前药在对主体给药时可能不具有活性, 但可在体内 转化为本发明的式(I)化合物或其盐和 /或溶剂化物。前药也可以是天然存在或者化学合成的 化合物。 本发明的式(I)化合物可以以非溶剂化形式或者诸如水, 乙醇等的可药用溶剂的溶剂化 形式存在, 且可预期的是本发明包括所有溶剂化和非溶剂化的形式。 式 (I)化合物的溶剂化 物优选为水化物。 本发明化合物的所有立体异构体, 例如由于式 (I) 化合物的 R取代基上的不对称碳原 子而可能存在的异构体, 包括对映体和非对映体形式, 均属于本发明的范围。 所有式 (I)所 示的化合物的立体异构体及其混合物, 包括外消旋混合物也都是本发明的一部分。 另外, 也 包括所有的几何异构体和位置异构体。 例如: 如果式 (I)所示的化合物含有双键, 则其顺式 与反式两种形式及其混合物也都包括在本发明的范围之内。 非对映异构体混合物可以通过本领域普通技术人员熟知的方法(如色谱法和 /或分步结晶 法) 基于它们的物理化学差异来分离为它们的各个非对映异构体。 对映异构体的分离可通过 与具有光学活性的化合物反应, 将对映异构体混合物转化成非对映异构体混合物, 然后分离 非对映异构体, 再将单独的非对映异构体转化 (如水解) 成相应的纯对映异构体。 另外, 某 些式 (I) 所示的化合物可以为阻转异构体 (如, 取代的联芳烃), 也是本发明中的一部分。 术语"药学上可接受的"是指某载体、 运载物、 稀释剂、 辅料, 和 /或所形成的盐通常在化 学上或物理上与构成某药物剂型的其它成分相兼容, 并在生理上与受体相兼容。 术语"盐"和"可药用的盐"是指式(I)所示的化合物或其立体异构体, 或其前药与无机和
/或有机酸和碱形成的酸式和 /或碱式盐, 也包括两性离子盐 (内盐), 还包括季铵盐, 例如烷 基铵盐。 这些盐可以是在化合物的最后分离和纯化中直接得到。 也可以是通过将式 (I)所示 化合物, 或其立体异构体, 或其前药, 与一定数量的酸或碱适当 (例如等当量) 进行混合而 得到。 这些盐可能在溶液中形成沉淀而以过滤方法收集, 或在溶剂蒸发后回收而得到, 或在 水介质中反应后冷冻干燥制得。 酸加成的盐包括如氢溴酸盐、 氢碘酸盐、 盐酸盐、 硫酸盐、 硫酸氢盐、 硝酸盐、 乙酸盐
(包括与乙酸或三卤乙酸如三氟乙酸形成的盐)、草酸盐、藻酸盐、抗坏血酸盐、天冬氨酸盐、 丁酸盐、 樟脑酸盐、 樟脑磺酸盐、 环戊烷丙酸盐、 二葡糖酸盐、 乙磺酸盐、 2-羟基乙磺酸盐、 2-萘磺酸盐、 烟酸盐、 过硫酸盐、 3-苯基丙酸盐、 苦味酸盐、 新戊酸盐、 丙酸盐、 水杨酸盐、 苯磺酸盐、 棕榈酸盐、 硬脂酸盐、 月桂酸盐、 硼酸盐、 苯甲酸盐、 乳酸盐、 磷酸盐、 甲苯磺 酸盐、 柠檬酸盐、 马来酸盐、 延胡索酸盐、 琥珀酸盐、 酒石酸盐、 硫氰酸盐、 萘酸盐、 甲磺 酸盐、 葡庚糖酸盐、 乳糖酸盐、 十二烷基磺酸盐、 己二酸盐以及其它类似盐。 碱性盐 (例如在 R取代基含有酸性部分如羧基或酚羟基的情形所形成的盐) 包括铵盐, 碱金属盐(例如钠盐、 锂盐和钾盐), 碱土金属盐(例如钙盐和镁盐), 与有机碱(如有机胺) 形成的盐 (例如二苄基乙二胺、 二环己胺、 哈胺、 N-甲基 -D-葡糖胺、 叔丁胺), 以及与氨基 酸如精氨酸、 赖氨酸形成的盐等。 也包括与含氮碱性试剂形成季铵盐, 包括但不限于铵、 四 甲基铵、 四乙铵、 甲胺、 二甲胺盐、 三甲基胺盐、 三乙胺盐、 乙胺以及其它类似物。 其他例 子详见本专利的参考文献 Berge, et al., J. Pharm. Sci., 66, 1-19 (1977)。 所述的式(I)化合物还可以作为处于平衡中的互变异构体存在, 且所有这些形式都包括 在本发明的范围内。 本发明的一种实施方式中, 本发明包括了同位素标记的式(I)化合物, 所述同位素标记 化合物是指与本文中所列化合物相同, 但是其中的一个或多个原子被另一个原子取代, 该原 子的原子质量或质量数不同于自然界中常见的的原子质量或质量数。 可以引入式 (I)化合物 中的同位素包括包括氢、 碳、 氮、 氧、 磷、 氟和氯, SP 2H, 3H、 13C、 14C、 15N、 170、 180、 31P、 32P、 35S、 18F和 36C1。 含有上述同位素和 /或其它原子同位素的式 (I) 的化合物及其 立体异构体和前药, 以及该化合物、 立体异构体或者前药的可药用的盐均应包含在本发明范 围之内。 某些同位素标记的式 (I) 化合物, 例如那些被如 3H和 14C等放射性同位素标记的化合 物,可被用作化合物和或底物的组织分布分析中。由于含氚(即 )的同位素和碳 14 (即 14C) 的同位素相对容易制备与检测, 我们优选 3H的同位素和 14C的同位素。 另外, 一些同位素, 如氘代 (即 2H), 由于有更好的代谢稳定性 (如提高体内半衰期、 或者降低给药剂量) 从而 提供某些治疗优势, 因此, 可在某些情况下优选。 所述同位素标记的式 (I)化合物通常可用 本领域普通技术人员已知的方法诸如用同位素标记的试剂替代非同位素标记的试剂来制备。 发明用途 本发明所述的化合物是新型雌激素受体 ER-a36的调节剂,并可用于调节 ER-a36在体外 和体内细胞中的功能, 因此, 本发明所述式 (I)化合物可用于预防和 /或治疗由 ER-0136介导 的相关疾病, 尤其是相关的肿瘤疾病。 在本发明的某些实施方式中, 提供了调节 ER-a36在细胞中的功能的方法, 该方法包括 将式 (I)的化合物作用于一个表达 ER-0136的细胞。所述细胞可内源性表达 ER-0136或通过基因 工程来外源性地表达 ER-a36,而且,可同时表达或不表达其它雌激素受体(如 ER-a66、 ER-a46 和 ER-βλ 在某一个实施方式中, 所述细胞内源性地表达 ER-a36。 在优选实施方式中, 所述 细胞是内源性地表达 ER-a36的癌细胞。 表达 ER-0136的癌细胞包括但不限于如乳腺癌细胞、 白血病细胞、 肝癌细胞、 淋巴瘤细胞、 肺癌细胞、 骨髓瘤细胞、 前列腺癌细胞、 卵巢癌细胞、 子宫内膜癌细胞、 结肠癌细胞和胃癌细胞。在更优选的实施方式中, 所述表达 ER-0136的细胞 是内源性地表达 ER-0136的乳腺癌细胞、 白血病细胞、 肝癌细胞、 淋巴瘤细胞、 子宫内膜癌细 胞和卵巢癌细胞。表达 ER-a36的乳腺癌细胞包括但不限于 MCF7、 MDA-MB-231和 SKBR-3 细胞。 表达 ER-a36的白血病细胞包括但不限于 K562, MV-4-11 , SUM159 , HL-60和 Molt-4 细胞。表达 ER-a36的子宫内膜癌细胞包括但不限于 HeclA细胞。表达 ER-a36的肝癌细胞包 括但不限于 A2780, BEL7402, BEL7404, HEL-9204 , Hep2G, Hep3B和来源于病人的原代 肝癌干细胞 Hep-12。 表达 ER-a36的淋巴瘤细胞包括但不限于 Daudi。 内源性 ER-a36的表达 可以通过用一种或多种试剂处理而增加或减少。 这些试剂包括如血清、 Ε2β(17β-雌二醇)、 他 莫昔芬和氟维司群 (ICI 182,780)。 在另一实施方式中, 本发明提供了表达外源性 ER-a36的细胞的制备方法。 该细胞可用 本领域普通技术人员已知的基因工程方法制备 (;参见 Samb k等, Molecular Cloning, A Laboratory Manual (2d Ed. 1989) (Cold Spring Harbor Laboratory)) 0简要来说, 先制备一个外源 性 ER-a36基因, 并将其插入至一个表达载体中, 再将所述表达载体转染到宿主细胞内, 然后 将宿主细胞放入适于表达外源性 ER-0136的培养液中生长。人 ER-0136的基因序列公开在 Wang 等人, Biochem. Biophys. Res. Commun. 336, 1023-1027 (2005) (GenBank登录号 BX640939)。 表达外源性 ER-a36的细胞可以表达或不表达内源性 ER-a36。细胞中内源性或外源性 ER-a36 的表达水平可以通过用一种或多种其它试剂处理而增加或减少。 这些试剂包括如血清、 Ε2β( 7β-雌二醇)、 他莫昔芬和氟维司群 (ICI 182,780)。 因此, 本发明所述的式 (I)化合物可用于制备预防和 /或治疗与 ER-a36相关癌症的药物, 包括但不限于肛门癌、 胆管癌、 膀胱癌、 骨癌、 肠癌 (结肠癌、 直肠癌)、 脑癌、 乳腺癌、 类 癌、 宫颈癌、 内分泌相关癌症、 子宫内膜癌、 眼癌、 胆囊癌、 头颈癌、 卡波济肉瘤癌、 肾癌、 喉癌、 白血病、 肝癌、 肺癌、 淋巴瘤、 黑色素瘤、 间皮瘤、 骨髓瘤、 神经内分泌癌、 食管癌、 卵巢癌、 胰腺癌、 阴茎癌、 前列腺癌、 皮肤癌、 软组织肉瘤癌、 脊髓癌、 胃癌、 睾丸癌、 甲 状腺癌、 阴道癌、 外阴癌或子宫癌。在更优选的实施方式中, 所述与 ER-a36相关癌症包括乳 腺癌、 宫颈癌、 结肠癌、 子宫内膜癌、 白血病、 肝癌、 淋巴瘤、 肺癌、 骨髓瘤、 卵巢癌、 前 列腺癌、 胃癌、 胰腺癌、 肾癌、 黑色素瘤、 甲状腺癌、 软组织肉瘤癌、 或子宫癌。。 在更加优 选的实施方式中, 所述与 ER-a36相关癌症包括乳腺癌, 肝癌、 淋巴瘤、 前列腺癌、 胃癌、 肺 癌、 结肠癌、 胰腺癌、 子宫内膜癌、 卵巢癌和白血病。 所述受试对象可以为哺乳动物, 如狗、 猫、 牛、 羊、 马或人, 优选人。 所述药物的必需 治疗量根据具体疾病不同而变化并且可由获益于本公开的本领域普通技术人员容易地确定。 在某些实施方式中, 本发明的一种或多种化合物可以彼此联合使用。 也可选择将本发明 的化合物与任何其它的活性试剂结合使用, 用于制备调控细胞功能或治疗疾病的药物或药物 组合物。 如果使用的是一组化合物, 则可将这些化合物同时、 分别或有序地对受试对象进行 给药。 在某些实施方式中, 本发明的化合物可以与一种或多种其它抗癌剂联合使用。 可联用的 抗癌剂包括但不限于烷化剂、 氮芥类药物、 叶酸拮抗剂、 嘌吟拮抗剂、 嘧啶拮抗剂、 纺锤体 毒素、 拓扑异构酶抑制剂、 凋亡诱导剂、 血管生成抑制剂、 鬼白毒素、 亚硝基脲、 抗代谢物、 蛋白合成抑制剂、 激酶抑制剂、 抗雌激素药、 顺铂、 卡铂、 干扰素、 天冬酰胺酶、 亮丙瑞林、 氟他胺、 甲地孕酮、 丝裂霉素、 博莱霉素、 阿霉素、 依立替康和紫杉醇。 在某一个实施方式 中, 所述抗癌剂是抗雌激素药物, 诸如他莫昔芬和氟维司群 (ICI 182,780)。 在本发明某些实施方式中, 式 (I)所示的化合物、 或其立体异构体或前药, 或所述化合物 的立体异构体或前药的一种可药用盐可以以药物组合物的形式, 其中包含可药用载体、 运载 剂或稀释剂。 它们也可用于制备预防和 /或治疗受试对象与 ER-0136相关的疾病的药物。 在某些实施方式中, 本发明的药物组合物也可用于治疗动物疾病。 普通兽医可根据从业 经验将本发明的一种化合物、 或其可兽用的盐、 或其可兽用的溶剂或其前药以合适的可接受 的制剂形式给药。 兽医可决定对某一动物最适合的剂量和给药途径。 如果将多种活性化合物组合起来给药, 则这些活性化合物可以被同时、 分别或以一定顺 序依次给药。 化合物的制备方法 式 (I)的化合物可以通过不同的合成方法来制备。 典型的制备过程如下所列。 除非有其它 表示, 1^, 1 2, 1 3, 1 4, 1 5, 1 6的定义如前所述。 对于本专业技术人员显而易见的是, 化合物的确切制备方法是可根据化学结构而略有不 同的。 而且, 在下面所阐述的多数制备过程中, 利用常规的保护基团 (以 P来表示) 来保护 不稳定或活泼基团是必要的。 上述保护基团的性质, 以及它们引入或去除的制备方法都是公 开的技术。 (实例见 Greene T. W. "有机合成中的保护基团", John Wiley & Sons,纽约, 1991 )。 以下所列的方案 1至方案 3以及相关描述仅作为本发明所述的式(I)化合物的制备方法的某 些实例, 并非限制本发明的范围。
Figure imgf000013_0001
Figure imgf000013_0002
方案 1
根据以上实施方案 1, 式 (I)的化合物可通过几个步骤制备。 化合物 L是由化合物 i_和 在路易斯酸的催化下, 经霍本-赫施反应 (改良傅-克酰基化反应) 而制备得到。 适用于该反 应的路易斯酸包括, 如无水氯化锌, 无水三氯化铝, 三氯化铁, 四氯化钛, 氯化锡, 三氟化 硼乙醚复合物等等。 该反应通常在约 0°C至约 120°C之间反应 1至 20个小时。 将化合物 L与各种取代的化合物 ήι在惰性溶剂中缩合反应可制备化合物 X。适用于该反 应的惰性溶剂包括醚, 如 DME, 1,2-二乙氧基乙烷, THF, 1,4-二氧六环, DMF, Ν,Ν-二甲 基乙酰胺, 吡啶, Ν-甲基 -2-吡咯烷酮。 该反应可在碱性条件下进行, 如氢氧化钾, 碳酸钾, , 碳酸铯, 氢化钠, 甲醇钠, 叔丁醇钾, DBU 1,8-二氮杂二环 -双环 (5,4,0)-7-十一烯), 正丁基锂, LDA (二异丙基氨基锂), LHMDS (六甲基二硅基胺基锂) 等等。 反应时可加入化学当量的或 催化量的相转移催化剂, 如 18-冠 -6, TBAB (四丁基溴化铵), TBAF (四丁基氟化铵)等等。 反 应的温度通常在约 0°C至约 140°C之间, 优选为在溶剂回流温度下反应 1至 20个小时。 将异戊烯基溴与化合物 iL在碱性条件下反应, 可制备 合物。 适用于该反应的溶剂 包括, 如甲醇, DMF(N,N-二甲基甲酰胺), THF (四氢呋喃), 水, 甲苯, DME(1,2-二甲氧基乙 烷), 以及混合溶剂, 如甲醇 -水, DMF-水, 四氢呋喃-水等等。在反应中使用的溶剂优选为水。 适用于该反应的碱包括,如氢氧化钾,碳酸钾,碳酸铯, 甲醇钠,氢化钠,叔丁醇钾, DBU(1,8- 二氮杂二环 -双环 (5,4,0)-7-十一烯), 正丁基锂, LDA (二异丙基氨基锂), LHMDS (六甲基二 硅基胺基锂) 等等。 反应温度通常在约 0°C至约 100°C之间, 反应时间为 1至 20个小时。
在化学合成策略上, 异戊烯基的引入和苯并吡喃酮母环的关环反应是本发明化合物合成 的关键步骤。 根据不同取代基的特性, 这两个关键步骤的先后顺序也可适当调整。 因此, 本 发明中的化合物也可采用方案 2来制备。
Figure imgf000014_0001
Figure imgf000014_0002
方案 2
在方案 2中,各类型反应所涉及的反应条件均与方案 1中类似。将化合物 ill在碱性条件 下异戊烯基溴反应, 可制备化合物^。 适用于该反应的溶剂包括, 如甲醇, DMF(N,N-二甲 基甲酰胺), THF(四氢呋喃), 水, 甲苯, DME(1,2-二甲氧基乙烷), 以及混合溶剂, 如甲醇- 水, DMF-水, 四氢呋喃-水等等。 在反应中使用的溶剂优选为水。 适用于该反应的碱包括, 如氢氧化钾,碳酸钾,碳酸铯,甲醇钠,氢化钠,叔丁醇钾, DBU(1,8-二氮杂二环 -双环 (5,4,0)-7- 十一烯), 正丁基锂, LDA (二异丙基氨基锂), LHMDS (六甲基二硅基胺基锂)等等。 反应温 度通常在约 0°C至约 100°C之间, 反应时间为 1至 20个小时。
将化合物 I L与各种取代酰氯 iii在惰性溶剂中缩合反应可制备化合物 ιίί。 适用于该反 应的惰性溶剂包括醚, 如 DME, 1,2-二乙氧基乙烷, THF, 1,4-二氧六环, DMF, Ν,Ν-二甲 基乙酰胺, 吡啶, Ν-甲基 -2-吡咯烷酮。 该反应可在碱性条件下进行, 如氢氧化钾, 碳酸钾, 碳酸铯, 氢化钠, 甲醇钠, 叔丁醇钾, DBU 1,8-二氮杂二环 -双环 (5,4,0)-7-十一烯), 正丁基锂, LDA (二异丙基氨基锂), LHMDS (六甲基二硅基胺基锂) 等等。 反应时可加入化学当量的或 催化量的相转移催化剂, 如 18-冠 -6, TBAB (四丁基溴化铵), TBAF (四丁基氟化铵)等等。 反 应的温度通常在约 0°C至约 140°C之间, 优选为在溶剂回流温度下反应 1至 20个小时。 当式 (I) 化合物的 R1为氢时, 其可依据以下实施方案 3来制备。
Figure imgf000015_0001
― ( xiiL)
方案 3
在方案 3中, P为羟基的保护基团。将化合物 的保护基脱除可得到化合物 。根据保 护基的不同, 去除的方法也相应变化, 主要参见"有机合成中的保护基团 "(Greene T. W等著. John Wiley & Sons, 纽约, 1991 ) 其中的方法。 优选的保护基为苄基, 苯甲酰基, 苄氧羰基, TBDMS (叔丁基二甲基硅基), THP (四氢吡喃基), 甲基, MOM (甲氧基甲基), PMB (对甲氧基 苄基)等。
化合物 siL与异戊烯基溴在碱性条件下反应, 可制备化合物^。适用于该反应的溶剂包 括,如甲醇, DMF(N,N-二甲基甲酰胺), THF (四氢呋喃),水, 甲苯, DME(1,2-二甲氧基乙烷), 以及混合溶剂, 如甲醇 -水, DMF-水, 四氢呋喃-水等等。 在反应中使用的溶剂优选为水。 适 用于该反应的碱包括, 如氢氧化钾, 碳酸钾, 碳酸铯, 甲醇钠, 氢化钠, 叔丁醇钾, DBU(1,8- 二氮杂二环 -双环 (5,4,0)-7-十一烯), 正丁基锂, LDA (二异丙基氨基锂), LHMDS (六甲基二 硅基胺基锂) 等等。 反应温度通常在约 0°C至约 100°C之间, 反应时间为 1至 20个小时。 具体实施方式
通过以下非限制性的实施例对本发明进行阐述, 除非另有所述, 否则: 室温或环境温度 是指 18-25°C范围内, 溶剂是在减压条件下使用旋转蒸发仪来脱除的, 反应过程是通过薄层 层析法 (TLC) 进行监测的。 反应时间仅用于阐述说明。 化合物的结构和纯度通过至少以下 一种技术确认: TLC, 质谱, 核磁共振 (NMR) , 高效液相色谱 (HPLC) 。 所述产率仅用 于阐述说明。
实施例 1
2-(4-三氟甲基苯基 )-3,5,7-三羟基 -8-P-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮 (化合物 1) 步骤 1 : 制备 2-甲氧基 -1-(2,4,6-三羟基苯基)乙酮 将间苯三酚 (35.1 g, 279 mmol) 溶于乙醚 (500 mL ) 中, 在冰水浴条件下, 加入无水 氯化锌 (8 g, 59 mmol ) 和 2-甲氧基乙腈 (18 g, 253 mmol) 。 将干燥的氯化氢气体通入到 反应体系中, 剧烈搅拌 5小时。 在这个过程中, 会有沉淀析出。 过滤收集沉淀物, 并将其溶 于水中, 加热回流 3小时。 冷却后, 收集粉红色沉淀, 并用水重结晶可得到白色目标化合物 ( 45 g,产率 81%)。 1H NMR (400 MHz, DMSO- 6): δ=12.14 (s, 2H), 10.41 (s, 1H), 5.79 (s, 2H), 4.56 (s, 2H), 3.32 (s, 3H)。 步骤 2: 制备 2-(4-三氟甲基苯基 )-3-甲氧基 -5,7-二羟基 -4H-苯并吡喃 -4-酮 将 2-甲氧基 -1-(2,4,6-三羟基苯基)乙酮(30 g,151 mmol)和 4-三氟甲基苯甲酰氯 (37.5 g, 180 mmol) 溶于 250 mL干燥的吡啶中, 室温滴加 DBU (53.2, 350 mmol)。 滴加完毕后, 将反应体 系加热至 75 °C, 搅拌反应过夜。 将反应混合物冷却至室温, 并减压除去大部分溶剂, 残余物 倒入到冰浴冷却的稀盐酸溶液中。 用 500 mL乙酸乙酯萃取 3次, 合并的萃取液用 300 mL 的 2N碳酸钠水溶液洗涤。 用无水硫酸钠干燥后, 浓缩, 粗产品用石油醚 /乙酸乙酯 (10: 1 ) 重 结晶得到目标化合物 (21 g, 产率 40%)。 步骤 3 : 制备 2 4-三氟甲基苯基 )-3,5,7-三羟基 -4H-苯并吡喃 -4-酮 在 250 mL的三颈烧瓶中, 将 2-(4-三氟甲基苯基 )-3-甲氧基 -5,7-二羟基 -4H-苯并吡喃 -4- 酮 (10 g, 28.3 mmol)溶于 150mL二氯甲烷中。 在 0 °C下慢慢滴加三溴化硼 (21.2 g, 84.9 mmol)。 加完后, 反应体系升至室温反应 4小时, 然后用 80 mL冰水淬灭反应, 用 500 mL乙酸乙酯萃 取 3次。 合并的萃取液用饱和氯化钠水溶液洗涤 1次, 无水硫酸钠干燥。 过滤浓缩后, 粗产 物用 20 mL的乙酸乙酯 /石油醚(1 : 10 )打浆后,过滤干燥得到黄色目标化合物(7 g,产率 70%)。 步骤 4: 制备 2-(4-三氟甲基苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮 (化 合物 1)
将化合物 2-(4-三氟甲基苯基 )-3,5,7-三羟基 -4H-苯并吡喃 -4-酮 (3.38 g, 10 mmol) 和碳酸 铯 (33g, 100 mmol)溶于 100 mL水中, 在冰水浴条件下滴加异戊烯基溴 (1.9 g, 13mmol)。 加完 后, 反应体系在室温下反应过夜, 用 2N 的盐酸调 pH至 6左右, 然后用乙酸乙酯萃取 2次。 合并有机相, 饱和氯化钠溶液洗涤后, 用无水硫酸钠干燥。 过滤浓缩后, 粗产物用硅胶柱层 析纯化,以乙酸乙酯 /石油醚(1 :25 )为洗脱液,得黄色目标化合物 508 mg,产率 U^/o ^H-NMR (300 MHz, DMSO- 6): δ=12.20 (s, 1Η), 10.87 (brs, 1H), 10.07 (brs, 1H), 8.35 (d, 2H, J= 8.1 Hz), 7.94 (d, 2H, J=7.7 Hz), 6.33 (s, 1H), 5.19 (t, 1H, J= 5.4 Hz), 3.45 (d, 2H, J=6.0 Hz), 1.75 (s, 3H), 1.64 (s, 3H); LC-MS (ESI, m/z): 407.0 [M+H]"0 参照实施例 1的制备方法, 化合物 2至化合物 8是以中间体 2-甲氧基 -1-(2,4,6-三羟基苯 基)乙酮为原料与各种不同取代的烷基酰氯、 芳基酰氯或杂芳基酰氯反应, 采用类似的方法制 备得到, 具体如下表 1所示。 表 1
Figure imgf000017_0001
实施例 9
2-(4-三氟甲基苯基 )-3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯小基) - 4H-苯并吡喃 -4-酮 (化合物 9) 将制备化合物 1时的中间体 2-(4-三氟甲基苯基 )-3-甲氧基 -5,7-二羟基 -4H-苯并吡喃 -4-酮 (500 mg, 1.42 mmol)和碳酸铯 (4.95g, 15 mmol)溶于 25 ml水中, 在冰水浴条件下滴加异戊烯 基溴 (220 mg, 1.5 mmol)。 加完后, 反应体系在室温下反应过夜, 用 2N 的盐酸调 pH至 6左 右, 然后用乙酸乙酯萃取 2次。 合并有机相, 饱和氯化钠溶液洗涤后, 用无水硫酸钠干燥。 过滤浓缩后, 粗产物用硅胶柱层析纯化, 以乙酸乙酯 /石油醚为洗脱液, 得黄色目标化合物 95 mg,产率 16.5%。 1H-NMR (300 MHz, DMSO- 6): δ=12.41 (IH, s), 10.92 (IH, brs), 8.20 (2H, d, J= 8.1 Hz), 7.96 (2H, d, J=7.7 Hz), 6.34 (IH, s), 5.15 (IH, t, J= 5.4 Hz), 3.84 (3H, s), 3.41 (2H, d, J=6.0 Hz), 1.68 (3H, s), 1.62 (3H, s); LC-MS (ESI, m/z): 421.1 [M+H] -。 实施例 10
2-P,4-二氟苯基) -3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮 (化合物 10) 步骤 1 : 制备 2-甲氧基小[2,4,6-三羟基 -3-(3-甲基丁 -2-烯)苯基]乙酮 将化合物 2-甲氧基 -1-(2,4,6-三羟基苯基)乙酮 (2.0 g, 10.09 mmol) 溶于 5%氢氧化钾水溶 液中(1.132 g, 20.18 mmol), 然后在冰浴条件下, 慢慢滴加异戊烯基溴 (1.504 g,10.09 mmol)。 反应混合物在室温下反应 2小时, 然后倒入到冰水中, 用 1N盐酸调节 pH至 2左右, 再用乙 酸乙酯萃取 3次。 合并有机相, 用无水硫酸钠干燥, 过滤浓缩后, 粗产物用硅胶柱层析纯化 (以二氯甲烷: 甲醇 =100:1为洗脱体系) 得目标化合物 (0.5g, 产率 18.6%) 。 ^ NMR OO MHz, DMSO- 6): δ=13.70 (s, IH), 10.70 (s, IH), 10.33 (9s, IH), 5.97 (s, IH), 5.08 (s, IH), 4.56 (s, 2H), 3.30 (s, 3H), 3.02 (m,2H), 1.66 (s, 3H), 1.57 (s, 3H). 步骤 2: 制备 2-(3,4-二氟苯基) -3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4- 酮 (化合物 10) 在氮气保护下, 将 2-甲氧基小[2,4,6-三羟基 -3,5-二 (3-甲基 -2-丁烯小基)苯基]乙酮 (250 mg, 0.94 mmol), 无水碳酸钾粉末(779 mg, 5.63 mmol), TBAB (四丁基溴化铵, 454 mg, 1.41 mmol) 和 3,4-二氟苯甲酰氯 (331 mg, 1.88 mmol) 溶于甲苯中 GO mL) 中, 回流反应 6小时。 冷却后, 去除甲苯, 加入 20 mL水。 水溶液用乙酸乙酯萃取。 合并的有机相用水和 饱和食盐水洗涤, 用无水硫酸钠干燥, 去除溶剂后得到褐色残余物。 将残余物溶于 20 mL甲 醇-水混合液 (;比例 4: 1), 加入氢氧化钾 (l g)。 混合物加热回流 2小时, 冷却到室温, 并用 1N 盐酸溶液酸化至 pH=4, 用二氯甲烷萃取三次。 合并的有机相用无水硫酸钠干燥并去除溶剂。 粗产物用硅胶柱层析(洗脱液: 乙酸乙酯 /石油醚 =1 : 50)纯化得到目标化合物(13.1 mg, 收 率 3.59%)。 JH NMR (400 MHz, CDC13): δ=12.45 (s, IH), 7.98-7.89 (m, 2H), 7.35-7.30 (m, IH), 6.34 (s, 2H), 5.25 (br, IH), 3.89 (s, 3H), 3.55 (d, J = 6.8 Hz, 2H), 1.84 (s, 3H), 1.77 (s, 3H); LC-MS: 390.1 (MH)+; Purity: 98.6% (254 nm). 参照实施例 9的制备方法, 化合物 11和化合物 13以各自相应的中间体为原料与异戊烯 基溴发生单取代反应而制备得到。 具体如下表 2所示。 表 2
Figure imgf000019_0001
生物活性测试方法 本发明的式 (I) 化合物的活性是通过以下分析方法测试的。 实施例 14: ER-α变体在人乳腺癌试样中的表达
自 ProSci Incorporated (Poway, CA) 公司处购买了一个预先涂抹了人乳腺癌细胞组织的 薄膜。 用特异性识别 ER-a36的抗 ER-a36抗体和 HRP共轭第二抗体检测薄膜, 再用增强化 学发光 (ECL)检测剂 (Amersham Pharmacia Biotech提供)进行显像。 洗脱该同一薄膜上的标记, 并用可识别 ER-α所有三种亚型—— ER-a66、 ER-a46和 ER-a36的抗雌激素受体 -a抗体 H222 (Novocastra Laboratories Ltd, UK公司提供)进行检测。 图 1显示了 ER-a66、 ER-a46和 ER-a36 三种雌激素受体亚型在正常乳腺组织中 (道 1)没有表达,但它们在浸润性导管癌的一个试样 (道 2)、 浸润性小叶癌的一个试样 (道 5)和非侵袭性导管癌 (道 7)中都有表达。 另外, ER-a36在侵 袭性导管癌 (道 4)和浸润性小叶癌的另一试样 (道 6)中有表达。其中,道 2和道 3是分别来自 2 个不同患者的浸润性导管癌组织。道 5和道 6是分别来自 2个不同患者的浸润性小叶癌组织。 此结果表明 ER-a36在正常乳腺组织中不表达, 而其在不表达 ER-a66和 ER-a46的 ER阴性 乳腺癌样本中有表达。 实施例 15: ER-a36在 ER阴性乳腺癌细胞系 MDA-MB-231中表达
MDA-MB-231 是一种大家熟知的缺失 ER-a66和 ER-a46 的细胞系 (;参见 Relevance of breast cancer cell lines as models for breast tumours: an update. Marc Lacroix, Guy Leclercq, Brest Cancer Research and Treatment 83: 249-289 (2004))。 MDA-MB-231细胞来自美国菌种保藏中心 (ATCC)。 将 MDA-MB-231细胞放在含有 Dulbecco改良 Eagle培养基 (DMEM)和 10%胎牛血 清的 8孔 BIOCOAT载玻片 (BD Science Discovery Labware 公司提供)中, 在 37°C和 5% C02 气氛中培养 12小时。然后所述细胞用无菌磷酸盐缓冲液 (PBS)冲洗 2次, 并在室温用 4%多聚 甲醛的 PBS(pH 7.4)溶液固定 30分钟。其后,所述细胞用 PBS冲洗,并用 0.5% (体积 /体积) Triton X-100通透破膜 10分钟。 再用 PBS冲洗细胞, 并加入 3%血清的 PBS溶液, 在室温下封闭 1 小时。 在所述载玻片上加入 ER-0136特异性抗体, 或加入预先与可结合 ER-0136特异性抗体的 免疫原多肽孵育 30分钟的相同抗体在室温下孵育 1小时,然后用含 0.5% Triton X- 100的 PBS (PBST)冲洗 3次, 再用异硫氰酸荧光素 (FTIC)标记的第二抗体孵育。最后, 所述玻片用 PBST 冲洗 3次, PBS冲洗 1次, 再加入免疫荧光标记 (Molecular Probes, Eugene, OR公司提供), 在 Nikon E600显微镜下检测并通过 MRC-1024共聚成像系统 (Bio-Rad公司提供)采集图像。 图 2(上图)显示了 MDA-MB-231细胞被抗 ER-a36抗体染色呈阳性。为了进一步证实该实验的可 信度,通过将被免疫原多肽预孵育过的抗 ER-0136抗体孵育后未显示出任何染色 (图 2, 下图), 表明所述抗体的特异性。 实施例 16: 蛋白免疫印迹法检测 ER-0136在不同肿瘤细胞株上的表达情况
在 37°C和 5%C02条件培养实验细胞(如 MDB-MA-231 , 培养基为 10%-FBS-DMEM)。 待每孔细胞长到 60%--90%满度, 收集细胞, 并于 4°C, 4300rpm条件下离心 5分钟。 除去上 清后, 加入适量裂解液以及含 1% ΝΡ-40和 0.7 mM EDTA的 Lysis缓冲液, 再加入蛋白酶抑 制剂, 与冰浴条件下裂解 30分钟至 1小时。 再次以 14000rpm离心 15分钟, 取上清液, 并进 行蛋白定量。 蛋白免疫印迹法通用程序: 在预制胶或自配胶上进行转膜、 电泳、 抗 ERa-36 抗体封闭、 洗脱、 二抗封闭、 洗脱、 最后在暗室下进行压片曝光, 显示结果。 图 3 显示了不 同肿瘤细胞株表达 ER-0136的蛋白免疫印迹结果。 道 1 : 瞬时过表达 ER-0136的 293人肾上皮 细胞株; 道 2-4: 不同实验室来源的人乳腺癌 SK-BR-3细胞株; 道 5-7: 不同实验室来源的人 乳腺癌 MCF-7细胞株; 道 8-9: 不同实验室来源的人白血病 HL-60细胞株; 道 10-11 : 不同 实验室来源的人白血病 MV-4-11细胞株; 道 12-13: 不同实验室来源的人慢性粒细胞白血病 K562细胞;道 14:人肝癌 A2780细胞;道 15:人肝癌 HEL-7402细胞;道 16:人肝癌 HEL-9204 细胞; 道 17: 来源于病人的原代肝癌 Hep-11细胞; 道 18: 来源于病人的原代肝癌 Hep-12细 胞。 实施例 17: 化合物对不同乳腺癌细胞的体外生长抑制作用
A: 对 ER-阴性乳腺癌 MDA-MB-231细胞的体外 CellTiter-Glo®ATP发光法细胞活力检测 在 37°C和 5% C02气氛条件下将 MDA-MB-231细胞保存在含有 10%胎牛血清的 DMEM 中。 细胞以 6xl03 个 /孔的密度接种在 96孔板上。 将测试化合物溶于 DMSO中, 并以浓度为 0, 0.3 μΜ, 0.5 μΜ, 1 μΜ, 2 μΜ, 3 μΜ, 5 μΜ, 10 μΜ, 20 μΜ, 30 μΜ, 50 μΜ and 100 μΜ 作用于 MDA-MB-231 细胞 72 小时。 然后用 CellTiter-Glo®发光法细胞活力检测试剂盒 (Promega)来检测被化合物作用后的细胞, 并用 Envision记录下发光值。
B: 对 ER-阳性乳腺癌 MCF-7细胞的体外 CellTiter-Glo®ATP发光法细胞活力检测 MCF7 细胞是高表达 ER- 66, ER- 46 and ER- 36 的乳腺癌细胞系。 (Relevance of breast cancer cell lines as models for breast tumours: an update. Marc Lacroix, Guy Leclercq, Breast Cancer Research and Treatment (2004) 83, 249-289; Wang et al., Proc. Natl. Acad. Sci. U.S.A.103:9063-9068 (2006)). MCF7细胞来自于 ATCC, 并且维持在 37°C, 5% C02气氛以及 在 Dulbecco改良 Eagle培养基 (DMEM)和 10%胎牛血清中。细胞以 6xl03 个 /孔的密度接种在 96孔板上。将测试化合物溶于 DMSO中,并以浓度为 0, 0.3 μΜ, 0.5 μΜ, 1 μΜ, 2 μΜ, 3 μΜ, 5 μΜ, 10 μΜ, 20 μΜ, 30 μΜ, 50 μΜ and 100 μΜ作用于 MCF7细胞 72小时。 然后用 CellTiter-Glo®发光法细胞活力检测试剂盒 (Promega)来检测被化合物作用后的细胞, 并用 Envision记录下发光值。
本发明部分化合物对不同乳腺癌细胞的细胞活力影响结果列举在下表 3中。
表 3
对乳腺癌细胞活力的抑制作用 IC5Q M)
化合物编号
MDA-MB-231细胞 MCF7细胞 他莫昔芬 a) 20.90±1.51 ) 22.55±4.15
1 1.066 2.012
2 NAC) 3.167
3 2.867 8.603
4 4.325 16.011
5 4.476 3.485
6 1.875 6.854
7 9.303 7.542
9 NA NA
10 NA 31.46
a) : 他莫昔芬作为阳性对照化合物。
b) : 当化合物被测试的次数大于 3次, IC5Q 值用平均值 ±标准差来表示。
c) : NA代表没有活性, 其 IC5Q 值大于 100 μΜ。
d): ND 代表没有测定。 实施例 18: 化合物对慢性白血病 K562细胞的体外生长抑制作用
A: 慢性白血病 K562细胞的体外 CellTiter-Glo®ATP发光法细胞活力检测
白血病 K562来自于 ATCC, 并维持在 37°C, 5% C02气氛以及在 IMDM和 10%胎牛血 清中。 细胞以 6xl03 个 /孔的密度接种在 96孔板上。 将测试化合物溶于 DMSO中, 并以浓度 为 0, 0.3 μΜ, 0.5 μΜ, 1 μΜ, 2 μΜ, 3 μΜ, 5 μΜ, 10 μΜ, 20 μΜ, 30 μΜ, 50 μΜ and 100 μΜ作用于 Κ562细胞 72小时。 然后用 CellTiter-Glo®发光法细胞活力检测试剂盒 (Promega) 来检测被化合物作用后的细胞, 并用 Envision记录下发光值。 实施例 19: 化合物对人 B淋巴细胞瘤 Daudi细胞的体外生长抑制作用
A: 人 B淋巴细胞瘤 Daudi细胞的体外 CellTiter-Glo®ATP发光法细胞活力检测
人 B淋巴细胞瘤 Daudi细胞来自于 ATCC,并维持在 37°C, 5% C02气氛以及在 IMDM和 10%胎牛血清中。细胞以 6xl03 个 /孔的密度接种在 96孔板上。将测试化合物溶于 DMSO中, 并以浓度为 0, 0.3 μΜ, 0.5 μΜ, 1 μΜ, 2 μΜ, 3 μΜ, 5 μΜ, 10 μΜ, 20 μΜ, 30 μΜ, 50 μΜ and 100 μΜ作用于 Κ562细胞 72小时。 然后用 CellTiter-Glo®发光法细胞活力检测试剂盒 (Promega)来检测被化合物作用后的细胞, 并用 Envision记录下发光值。 本发明部分化合物体外对白血病 K562细胞和人 B淋巴细胞瘤 Daudi细胞的细胞活力影响结 果列举在下表 4中。
表 4
体外对细胞活力的抑制作用 Κ5ο (μΜ)
化合物编号
Κ562细胞 Daudi细胞 格列卫 a) 0.751 ND )
阿糖胞苷 ND 10.033
1 1.035 0.824
2 6.139 9.234
3 1.849 2.909
4 0.974 2.638
5 1.475 1.486
6 0.935 1.983
7 6.140 4.068
10 ΝΑ 21.75
11 ΝΑ NA
a) : 格列卫和阿糖胞苷分别作为 K562细胞模型和 Daudi细胞模型 的阳性对照化合物。
b) : ND 代表没有测定。
c): NA代表没有活性, 其 IC5Q 值大于 100 μΜ。 实施例 20: 化合物对急性白血病细胞的体外生长抑制作用
Α: 体外 ΜΤΤ法检测化合物对急性粒细胞白血病细胞 HL-60细胞增殖的抑制作用
急性粒细胞白血病细胞 HL-60来自于 ATCC,并维持在 37°C, 5% C02气氛以及在 IMDM 和 10%胎牛血清中。 细胞以 6xl03 个 /孔的密度接种在 96孔板上。 将测试化合物溶于 DMSO 中, 并以浓度为 0, 10_4M, 10_5M, 10_6M, 10"7M, 10_8M作用于 HL-60细胞 72小时。 然后 用 MTT法来检测 OD值, 计算细胞增殖被抑制的百分率。 B: 体外 MTT法检测化合物对急性淋巴母细胞白血病细胞 Molt-4细胞增殖的抑制作用 急性淋巴母细胞白血病细胞 Molt-4来自于 ATCC, 并维持在 37°C, 5% C02气氛以及在 RPMI-1640和 10%胎牛血清中。 细胞以 6xl03 个 /孔的密度接种在 96孔板上。 将测试化合物 溶于 DMSO中, 并以浓度为 0, 10"4M, 10"5M, 10"6M, 10"7M, 10—8M作用于 Molt-4细胞 72 小时。 然后用 MTT法来检测 OD值, 计算细胞增殖被抑制的百分率。 本发明部分化合物对不同白血病细胞的细胞活力影响结果列举在表 5中。
表 5
以 10—6M浓度化合物对细胞增殖的抑制百分率 (%)
化合物编号
HL-60细胞 Molt-4细胞 阿霉素 a) 79.0 90.1
1 60.7 43.7
2 NAb) NA
3 28.5 14.4
4 NA 14.7
5 59.8 69.4
6 61.9 67.4
7 NA 57.0
9 NA NA
11 NA NA
a) : 以阿霉素分别作为阳性对照化合物。
b) : NA代表没有活性, 化合物在 10—6M浓度下对细胞增殖的抑制百分 率小于 10%。 实施例 21 : 化合物对肝癌细胞的体外生长抑制作用
A: 体外 SRB法检测化合物对肝癌细胞 BEL-7402细胞增殖的抑制作用
肝癌细胞 HL-60来自于 ATCC, 并维持在 37°C, 5% C02气氛以及在 DMEM禾口 10%10%NCS以及 5(^g/mlKANA中。细胞以 6xl03 个 /孔的密度接种在 96孔板上。将测试化 合物溶于 DMSO中, 并以浓度为 0, 10"4M, 10"5M, 10"6M, 10"7M, 1(T8M作用于 BEL-7402 细胞 72小时。 然后用 SRB法来检测 OD值, 计算细胞增殖被抑制的百分率。 本发明部分化合物对肝癌细胞的生长抑制作用结果列举在表 6中
表 6
以 10—6M浓度化合物对细胞增殖的抑制百分率 (%)
化合物编号
BEL-7402细胞 阿霉素 a) 55.2
1 23.7
2 21.8
3 28.6
4 46.4
5 15.6
6 14.9
7 19.8
9 11.7
10 17.4
a): 阿霉素作为阳性对照化合物。 实施例 22: 化合物对胃癌细胞的体外生长抑制作用
A: 体外 MTT法检测化合物对胃癌细胞 BGC-823细胞增殖的抑制作用
将胃癌细胞 BGC-823以 3xl03 个 /孔的密度接种在 96孔板上, 并在 37°C, 5% C02气氛 中, 用含有 2.5% CS-FBS的无酚红的 DMEM培养基养 24小时。 将测试化合物溶于 DMSO 中, 并以浓度为 0, 1, 2, 4, 8, 10, 20 μΜ作用于 BGC-823细胞 72小时。 然后用 MTT法 来检测 OD值, 计算细胞增殖被抑制的百分率。 结果见附图 4。 实施例 23 : 化合物对肺癌细胞的体外生长抑制作用
A: 体外 MTT法检测化合物对肺癌细胞 H460细胞增殖的抑制作用
将肺癌细胞 H460以 4.0xl03 个 /孔的密度接种在 96孔板上,并在 37°C, 5% C02气氛中, 用含有 2.5% CS-FBS的无酚红的 1640培养基养 24小时。将测试化合物溶于 DMSO中, 并以 浓度为 0, 1, 2, 4, 8, 10, 20 μΜ作用于 H460细胞 72小时。然后用 ΜΤΤ法来检测 OD值, 计算细胞增殖被抑制的百分率。 结果见附图 5。 实施例 24: 化合物对结肠癌细胞的体外生长抑制作用
Α: 体外 ΜΤΤ法检测化合物对结肠癌细胞 LS174T细胞增殖的抑制作用
将结肠癌细胞 LS174T以 4.5χ103 个 /孔的密度接种在 96孔板上, 并在 37°C, 5% C02 气氛中, 用含有 2.5% CS-FBS的无酚红的 1640培养基养 24小时。 将测试化合物溶于 DMSO 中, 并以浓度为 0, 1, 2, 4, 8, 10, 20 μΜ作用于 LS174T细胞 72小时。 然后用 ΜΤΤ法来 检测 OD值, 计算细胞增殖被抑制的百分率。 结果见附图 6。 实施例 25 : 化合物对胰腺癌细胞的体外生长抑制作用
Α: 体外 ΜΤΤ法检测化合物对胰腺癌细胞 PANC-1细胞增殖的抑制作用 将胰腺癌细胞 PANC-1以 3xl03 个 /孔的密度接种在 96孔板上, 并在 37°C, 5% C02气 氛中,用含有 2.5% CS-FBS的无酚红的 1640培养基养 24小时。将测试化合物溶于 DMSO中, 并以浓度为 0, 1, 2, 4, 8, 10, 20 μΜ作用于 PANC-1细胞 72小时。 然后用 ΜΤΤ法来检 测 OD值, 计算细胞增殖被抑制的百分率。 结果见附图 7。 实施例 26: 化合物对前列腺细胞的体外生长抑制作用
Α: 体外 ΜΤΤ法检测化合物对前列腺细胞 PC-3细胞增殖的抑制作用
将前列腺癌细胞 PC-3以 3χ103 个 /孔的密度接种在 96孔板上, 并在 37°C, 5% C02气氛 中, 用含有 10%胎牛血清的 F12K培养基养 24小时。 将测试化合物溶于 DMSO中, 并以浓 度为 0, 1, 2, 4, 8, 10, 20 μΜ作用于 PC-3细胞 72小时。 然后用 ΜΤΤ法来检测 OD值, 计算细胞增殖被抑制的百分率。 结果见附图 8。 体内评价 实施例 27: 化合物对裸鼠体内人乳腺癌 BCAP-37细胞移植瘤的生长抑制作用
将测试化合物作用于带有乳腺癌移植瘤的裸鼠, 以测试它们抑制肿瘤生长的效果。 从具 有 BCAP-37细胞的裸鼠体内提取肿瘤组织, 切成小片。将几片肿瘤组织植入雌性裸鼠右前肢 的腋下。 植入后, 以每只裸鼠 7 μ§的剂量连续 6天每天一次对其注射 Ε2β溶液, 以剌激肿瘤 在受注鼠体内的生长。 从第 7天开始, 以 35 mg/kg的剂量给荷瘤裸鼠灌胃于被测化合物的玉 米油溶液。 使用他莫昔芬作为阳性对照, 玉米油作为阴性对照, 将被测化合物溶于玉米油溶 液中 (20 mg/mL) 中。 连续 15-21天, 每天向裸鼠给药 35 mg/kg的被测化合物和他莫昔芬, 或玉米油。 裸鼠死亡后, 对其解剖并取出肿瘤组织称重。 用以下公式百分比计算肿瘤组织的 生长抑制率: 肿瘤的生长抑制率 = (阴性对照组肿瘤平均重量-被测化合物治疗过的肿瘤平 均重量) /阴性对照组肿瘤的平均重量。 将实验结果作柱状图, 列于图 9中。 实施例 28: 化合物对裸鼠体内人 B淋巴细胞瘤 Daudi细胞移植瘤的生长抑制作用
将测试化合物作用于带有人 B淋巴细胞瘤 Daudi细胞移植瘤的裸鼠, 以测试它们抑制 肿瘤生长的效果。 人 B淋巴细胞瘤 Daudi细胞来源于 ATCC。 5次传代后的 l x lO7个细胞加 入 0.2 mL Matrigel后一起皮下植入到雄性裸鼠右前肢的腋下。当裸鼠体内的肿瘤达到 150~200 mm3时, 将荷瘤裸鼠随机分组, 每十只为一组。 使混合油灌胃给药组作为阴性对照, 静脉给 予利妥昔单抗作为阳性对照组, 被测化合物溶于混合油中灌胃给药。 给药周期为连续 21天, 每天向测试组裸鼠灌胃给药被测化合物的混合油悬浮液(35mg/kg)—次; 每周两次次向阳性 对照组注射给药利妥昔单抗 (20 mg/kg) , 同时每天灌胃给予阴性对照组空白混合油溶媒一 次。 在给药周期中, 每周测量两次肿瘤体积和裸鼠体重。 以肿瘤体积和给药时间做肿瘤生长 曲线图 (图 10) , 评估化合物对肿瘤生长的抑制作用。 实施例 29: 化合物对裸鼠体内人子宫内膜癌 Ishikawa细胞移植瘤的生长抑制作用
将测试化合物作用于带有子宫内膜癌移植瘤的裸鼠, 以测试它们抑制肿瘤生长的效果。 从具有 Ishikawa细胞的裸鼠体内提取肿瘤组织, 切成小片。 将几片肿瘤组织植入雌性裸鼠右 前肢的腋下。 植入后, 以每只裸鼠 7 μ§的剂量连续 6天每天一次对其注射 Ε2β溶液, 以剌激 肿瘤在受注鼠体内的生长。 从第 7天开始, 以 35 mg/kg的剂量给荷瘤裸鼠灌胃于被测化合物 的混合油溶液。 使用醋酸甲孕酮作为阳性对照, 混合油作为阴性对照, 将被测化合物溶于混 合油溶液中 (20 mg/mL) 中。 连续 15-21天, 每天向裸鼠给药 35 mg/kg的被测化合物和醋酸 甲孕酮, 或混合油。 裸鼠死亡后, 对其解剖并取出肿瘤组织称重。 用以下公式百分比计算肿 瘤组织的生长抑制率: 肿瘤的生长抑制率 = (阴性对照组肿瘤平均重量 -被测化合物治疗过 的肿瘤平均重量) /阴性对照组肿瘤的平均重量。 将实验结果作柱状图, 列于图 11中。

Claims

1、 式 (I) 所示的化合物或其药学上可接受的盐, 溶剂合物或前药,
Figure imgf000027_0001
其中:
R1可选自氢, (C C6)烷基, 含有一个或多个卤原子取代的 (C C6)烷基;
R2, R3, R4, R5和 R6可独立选自氢, (d-C4烷基, 含有一个或多个卤原子取代的 (d-C4) 烷基, 素, 氰基, 含有一个或多个 原子取代的 (CrC4)烷氧基; 但 R2, R3, R4, R5和 R6 不可同时为氢;
且当 R1为甲基, R3和 R5为氢时, R4不可为氯原子。
2、 如权利要求 1所述的式 (I) 化合物或其药学上可接受的盐, 溶剂合物或前药, 其中:
Figure imgf000027_0002
其中:
R1可选自氢, 甲基, 乙基, 三氟甲基, 二氟甲基;
R2, R3, R4, R5和 R6可独立选自氢, 甲基, 乙基, 丙基, 异丙基, 丁基, 卤素, 氰基,, 含有一个或多个卤原子取代的 (d-C4)烷基, 含有一个或多个卤原子取代的 (d-C4)烷氧基; 但 R2, R3, R4, R5和 R6不可同时为氢;
且当 R1为甲基, R3和 R5为氢时, R4不可为氯原子。
3、 如权利要求 2所述的化合物, 当 R1为氢时, 其化学结构如式 (Π) 所示,
Figure imgf000028_0001
其中 :
R2, R3, R4, R5和 R6可独立选自氢, 甲基, 乙基, 丙基, 异丙基, 丁基, 卤素, 氰基,, 含有一个或多个卤原子取代的 (d-C4)烷基, 含有一个或多个卤原子取代的 (d-Q烷氧基; 但 R2, R3, R4, R5和 R6不可同时为氢。
4、 如权利要求 2所述的化合物, 当 R1为甲基时, 其化学结构如式 (ΠΙ) 所示,
Figure imgf000028_0002
其中 :
R2, R3, R4, R5和 R6可独立选自氢, 甲基, 乙基, 丙基, 异丙基, 丁基, 卤素, 氰基, 含有一个或多个卤原子取代的 (d-C4)烷基, 含有一个或多个卤原子取代的 (d-Q烷氧基, 但 R2, R3, R4, R5和 R6不可同时为氢;
且当 R3和 R5为氢时, R4不可为氯原子。 5、 如权利要求 1-4所述, 本发明优选的化合物可选自:
2-(4-三氟甲基苯基 )-3,5,7-三羟基 -8-P-甲基 -2-丁烯 -1-基) - 4H-苯并吡喃 -4-酮;
2-(4-氟苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3-氟 -4-氯苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(4-氯苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(4-三氟甲氧基苯基) -3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3,4-二氯苯基) -3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3-三氟甲基 -4-氯苯基 )-3,5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮; 2-(4-溴苯基 )-3,
5,7-三羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮;
2-(3,4-二氟苯基) -3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮; 2-(4-三氟甲基苯基 )-3-甲氧基 -5,7-二羟基 -8-P-甲基 -2-丁烯 -1-基)- 4H-苯并吡喃 -4-酮; 2-(4-三氟甲氧基苯基) -3-甲氧基 -5,7-二羟基 -8 3-甲基 -2-丁烯 -1-基) - 4H-苯并吡喃 -4-酮; 2-(3-三氟甲基 -4-氯苯基 )-3-甲氧基 -5,7-二羟基 -8-P-甲基 -2-丁烯小基)- 4H-苯并吡喃 -4- 酮;
2-(4-溴苯基 )-3-甲氧基 -5,7-二羟基 -8-(3-甲基 -2-丁烯小基)- 4H-苯并吡喃 -4-酮。
6、 如权利要求 1-5所述化合物, 或其药学上可接受的盐、 溶剂合物或者前体药物的有效剂 组成的药物组合物, 其中含有一个或多个可药用辅料。
7、 如权利要求 1-5所述的化合物, 或其药学上可接受的盐、 溶剂合物或者前体药物, 及其药 物组合物在制备预防或治疗与 ER-0136有关的肿瘤疾病的药物中的应用。
8、 如权利要求 7所述的应用, 其中所述的癌症选自胆管癌、 膀胱癌、 骨癌、 肠癌 (结肠癌、 直肠癌)、 脑癌、 乳腺癌、 宫颈癌、 子宫内膜癌、 头颈癌、 卡波济肉瘤癌、 肾癌、 喉癌、 白血 病、 肝癌、 肺癌、 淋巴瘤、 黑色素瘤、 间皮瘤、 骨髓瘤、 神经内分泌癌、 食管癌、 卵巢癌、 胰腺癌、 阴茎癌、 前列腺癌、 皮肤癌、 软组织肉瘤癌、 脊髓癌、 胃癌、 睾丸癌、 甲状腺癌、 或子宫癌。
9、 如权利要求 8所述的应用, 其中所述的癌症选自乳腺癌、 宫颈癌、 结肠癌、 子宫内膜癌、 白血病、 肝癌、 淋巴瘤、 肺癌、 骨髓瘤、 卵巢癌、 前列腺癌、 胃癌、 胰腺癌、 肾癌、 黑色素 瘤、 甲状腺癌、 软组织肉瘤癌、 或子宫癌。
10、 如权利要求 9所述的应用, 其中所述的癌症是乳腺癌, 肝癌、 淋巴瘤、 前列腺癌、 胃癌、 肺癌、 结肠癌、 胰腺癌、 子宫内膜癌、 卵巢癌和白血病。
PCT/CN2012/088016 2012-01-13 2012-12-31 多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用 WO2013104263A1 (zh)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US14/371,866 US9221781B2 (en) 2012-01-13 2012-12-31 Synthesis of polyhydroxy chromenone compounds and their anti-tumor effects
KR1020147022558A KR102030207B1 (ko) 2012-01-13 2012-12-31 폴리하이드록시 크로메논 화합물의 합성 및 이의 항종양 효과
BR112014016635A BR112014016635B8 (pt) 2012-01-13 2012-12-31 composto, composição, e, uso de um composto
CA2860999A CA2860999C (en) 2012-01-13 2012-12-31 Synthesis of polyhydroxy chromenone compounds and their anti-tumor effects
EP12864757.5A EP2803665B1 (en) 2012-01-13 2012-12-31 Synthesis of polyhydroxy benzopyran ketone compound and anti-tumor effect thereof
JP2014551506A JP6113751B2 (ja) 2012-01-13 2012-12-31 ポリヒドロキシベンゾピランケトン化合物の合成およびその抗腫瘍効果
ES12864757.5T ES2627316T3 (es) 2012-01-13 2012-12-31 Síntesis del compuesto de polihidroxi benzopirano cetona y efecto antitumoral del mismo
AU2012365712A AU2012365712B2 (en) 2012-01-13 2012-12-31 Synthesis of polyhydroxy benzopyran ketone compound and anti-tumor effect thereof
MX2014008526A MX364833B (es) 2012-01-13 2012-12-31 Sintesis de compuestos polihidroxi cromenona y sus efectos anti-tumor.

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201210011031.8 2012-01-13
CN201210011031 2012-01-13
CN201210573072.6 2012-12-25
CN201210573072.6A CN103204838B (zh) 2012-01-13 2012-12-25 多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用

Publications (1)

Publication Number Publication Date
WO2013104263A1 true WO2013104263A1 (zh) 2013-07-18

Family

ID=48752268

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/088016 WO2013104263A1 (zh) 2012-01-13 2012-12-31 多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用

Country Status (11)

Country Link
US (1) US9221781B2 (zh)
EP (1) EP2803665B1 (zh)
JP (1) JP6113751B2 (zh)
KR (1) KR102030207B1 (zh)
CN (1) CN103204838B (zh)
AU (1) AU2012365712B2 (zh)
BR (1) BR112014016635B8 (zh)
CA (1) CA2860999C (zh)
ES (1) ES2627316T3 (zh)
MX (1) MX364833B (zh)
WO (1) WO2013104263A1 (zh)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101784990B1 (ko) * 2016-01-13 2017-10-12 주식회사 파이브지티 얼굴인식을 이용한 홈 오토메이션 제어 시스템 및 그 방법
CN107216303B (zh) * 2016-07-15 2020-04-21 北京盛诺基医药科技股份有限公司 氟可拉定的合成方法
CN107216302B (zh) * 2016-07-15 2020-02-21 北京盛诺基医药科技股份有限公司 一种氟可拉定的合成方法
AU2017313753B2 (en) 2016-08-16 2022-01-27 The Trustees Of Columbia University In The City Of New York GABA(A) receptor modulators and methods to control airway hyperresponsiveness and inflammation in asthma
US10266510B2 (en) 2017-01-18 2019-04-23 King Abdulaziz University Antimicrobial and cytotoxic compounds and methods for treating cancer, a bacterial infection, and/or a fungal infection
CN108947949B (zh) * 2017-05-19 2022-08-19 泰州华元医药科技有限公司 抗焦虑氘代化合物及其医药用途
CN109020938A (zh) * 2018-08-17 2018-12-18 昆明龙津药业股份有限公司 一种杨梅素的制备方法
CN110054605A (zh) * 2019-05-31 2019-07-26 北京盛诺基医药科技有限公司 一种淫羊藿素中间体的制备方法
CN110092770A (zh) * 2019-05-31 2019-08-06 北京盛诺基医药科技有限公司 一种黄酮化合物中间体的制备方法
CN112569222A (zh) * 2020-12-31 2021-03-30 赣南医学院 三氟淫羊藿素在制备改善疼痛、肿胀及运动功能的药物中的应用
CN112851615B (zh) * 2021-01-21 2023-04-07 厦门大学 一种异戊烯基黄酮的制备及其作为治疗宫颈癌药物的应用
CN113717138B (zh) * 2021-10-21 2023-02-24 沈阳药科大学 一类氮芥类色原酮衍生物与应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101104611A (zh) * 2007-04-29 2008-01-16 殷正丰 一种3-甲氧基黄酮类化合物,其制备方法和应用
CN102018698A (zh) * 2008-04-18 2011-04-20 盛诺基医药科技有限公司 治疗雌激素受体相关疾病的化合物及其方法

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1786864A (zh) 2004-12-10 2006-06-14 上海迪比特实业有限公司 一种计算机安全认证方法
JP2007332695A (ja) 2006-06-16 2007-12-27 Advanced Media Inc 電子錠装置及び音声認証ロッカーシステム
WO2008100977A2 (en) 2007-02-14 2008-08-21 N.V. Organon Carbamates therapeutic release agents as amidase inhibitors
CN102038673B (zh) * 2009-10-20 2012-07-25 北京珅奥基医药科技有限公司 羟基苯并吡喃酮类化合物在制备治疗白血病药物中的应用
CN102558164B (zh) 2010-12-31 2016-06-15 北京盛诺基医药科技有限公司 苯并吡喃酮类雌激素受体调节剂

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101104611A (zh) * 2007-04-29 2008-01-16 殷正丰 一种3-甲氧基黄酮类化合物,其制备方法和应用
CN102018698A (zh) * 2008-04-18 2011-04-20 盛诺基医药科技有限公司 治疗雌激素受体相关疾病的化合物及其方法

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
"ACS Symposium Series", vol. 14, article "Produgs as Novel Delivery Systems"
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BIOCHEM. BIOPHYS. RES. COMMUN., vol. 336, 2005, pages 1023 - 1027
FLOURIOT, G., EMBO, vol. 19, 2000, pages 4688 - 4700
GREENE T. W.: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
GREENE T.W: "protective groups in organic synthesis", 1991, JOHN WILEY & SONS
LI ET AL., PROC. NATL. ACAD. SCI., USA, vol. 100, 2003, pages 4807 - 4812
MARC LACROIX; GUY LECLERCQ: "Relevance of breast cancer cell lines as models for breast tumors: an update", BREAST CANCER RESEARCH AND TREATMENT, vol. 83, 2004, pages 249 - 289, XP002502163, DOI: doi:10.1023/B:BREA.0000014042.54925.cc
MARC LACROIX; GUY LECLERCQ: "Relevance of breast cancer cell lines as models for breast tumours: an update", BREAST CANCER RESEARCH AND TREATMENT, vol. 83, 2004, pages 249 - 289, XP002502163, DOI: doi:10.1023/B:BREA.0000014042.54925.cc
SAMBOOK: "Molecular Cloning, A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
See also references of EP2803665A4 *
WANG ET AL., BIOCHEM. BIOPHYS. RES.COMMUN, vol. 336, 2005, pages 1023 - 1027
WANG ET AL., BIOCHEM. BIOPHYS. RES.COMMUN., vol. 336, 2005, pages 1023 - 1027
WANG ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 103, 2006, pages 9063 - 9068
WANG ET AL., PROC.NATL.ACAD.SCI., USA, vol. 103, 2006, pages 9063 - 9068

Also Published As

Publication number Publication date
CA2860999A1 (en) 2013-07-18
US20150011618A1 (en) 2015-01-08
CN103204838B (zh) 2017-06-09
JP6113751B2 (ja) 2017-04-12
JP2015503596A (ja) 2015-02-02
BR112014016635B1 (pt) 2021-07-13
BR112014016635B8 (pt) 2021-08-24
BR112014016635A8 (pt) 2017-07-04
EP2803665A4 (en) 2015-07-08
ES2627316T3 (es) 2017-07-27
KR20140109498A (ko) 2014-09-15
CA2860999C (en) 2019-12-31
CN103204838A (zh) 2013-07-17
US9221781B2 (en) 2015-12-29
KR102030207B1 (ko) 2019-10-08
EP2803665B1 (en) 2017-05-10
MX364833B (es) 2019-05-08
AU2012365712B2 (en) 2017-04-20
EP2803665A1 (en) 2014-11-19
AU2012365712A1 (en) 2014-07-24
MX2014008526A (es) 2014-11-25
BR112014016635A2 (pt) 2017-06-13

Similar Documents

Publication Publication Date Title
WO2013104263A1 (zh) 多羟基苯并吡喃酮类化合物的合成及其抗肿瘤作用
US9090583B2 (en) Benzopyrone estrogen receptor regulator
CN102018698B (zh) 治疗雌激素受体相关疾病的化合物及其方法
US10398689B2 (en) Benzopiperidine derivative, preparation method thereof and medical use thereof
CN105008343A (zh) 作为选择性雌激素受体降解剂的苯并噻吩衍生物及其组合物
US10315990B2 (en) Isothiocyanate compounds and isothiocyanate XPO1 protein inhibitor drugs thereof
JP5938400B2 (ja) ピロリル置換ジヒドロインドール−2−オン誘導体、調製方法及びそれらの用途
JP6674381B2 (ja) 癌治療のためのMcl−1調節化合物
JP2016515585A (ja) 癌、神経障害及び線維性障害の治療のための、脂質フラン、ピロール及びチオフェン化合物
CN110240587A (zh) 一类芳基二氟苄基醚类化合物、制备方法及用途
WO2014169697A1 (zh) 长春碱类衍生物及其制备方法和应用
CN113956236B (zh) 邻苯二甲酰亚胺类化合物及其用途
WO2023088257A1 (zh) 苦鬼臼毒素衍生物、其药物组合物及用途
CN103755673A (zh) 治疗雌激素受体相关疾病的化合物及其方法
WO2023141852A1 (zh) Cdk2抑制剂及其制备方法和用途
CN108186630A (zh) 靛红类似物在制备抗肿瘤药物中的应用
KR20220104734A (ko) 올레아난 신남아미드 유도체 및 이의 제조 방법과 용도
CN108912113A (zh) 一种化合物及其作为选择性pi3k抑制剂在制备治疗肿瘤的药物中的应用

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: 12864757

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2012864757

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012864757

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2860999

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 14371866

Country of ref document: US

Ref document number: MX/A/2014/008526

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2014551506

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2012365712

Country of ref document: AU

Date of ref document: 20121231

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20147022558

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014016635

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014016635

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140703