WO2017080338A1 - 丙烯酸类衍生物、其制备方法及其在医药上的用途 - Google Patents

丙烯酸类衍生物、其制备方法及其在医药上的用途 Download PDF

Info

Publication number
WO2017080338A1
WO2017080338A1 PCT/CN2016/101768 CN2016101768W WO2017080338A1 WO 2017080338 A1 WO2017080338 A1 WO 2017080338A1 CN 2016101768 W CN2016101768 W CN 2016101768W WO 2017080338 A1 WO2017080338 A1 WO 2017080338A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
halogen
aryl
alkyl
cycloalkyl
Prior art date
Application number
PCT/CN2016/101768
Other languages
English (en)
French (fr)
Inventor
关东亮
盛首一
白骅
Original Assignee
浙江海正药业股份有限公司
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 浙江海正药业股份有限公司 filed Critical 浙江海正药业股份有限公司
Priority to EP16863515.9A priority Critical patent/EP3378861B1/en
Priority to CN201680020252.8A priority patent/CN107428758B/zh
Priority to US15/766,692 priority patent/US10519148B2/en
Priority to CA3011391A priority patent/CA3011391C/en
Priority to JP2018524291A priority patent/JP6592197B2/ja
Priority to KR1020187014684A priority patent/KR102099159B1/ko
Publication of WO2017080338A1 publication Critical patent/WO2017080338A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention belongs to the field of medicine, and in particular relates to a novel acrylic derivative, a stereoisomer thereof, a tautomer thereof or a pharmaceutically acceptable salt thereof, a process for preparing the same, a pharmaceutical composition containing the same, and the same Use as a therapeutic, especially as an estrogen receptor antagonist or an estrogen receptor alpha downregulator.
  • the estrogen receptor (ER) is a ligand-activated transcriptional regulatory protein that mediates the induction of various biological effects through interaction with endogenous estrogens, including 17 ⁇ -estradiol and Estrone.
  • ER has been found to have two subtypes of estrogen receptor alpha (ER alpha, ESR1 and NR3A) and estrogen receptor beta (ER beta, ESR2 and NR3b).
  • Estrogen receptor alpha and estrogen receptor beta are members of the steroid hormone receptor, which is a member of the nuclear receptor family. Similar to the mechanism of nuclear receptors, ER ⁇ is composed of six functional domains (named AF), a ligand-activated transcription factor, because it binds to a specific ligand, including endogenous estrogen 17 ⁇ .
  • Estradiol (E2) binds to the genomic sequence to form a complex, an estrogen receptor responsive element and a co-regulatory factor, to regulate the transcription of the targeted gene.
  • the ER ⁇ gene is located at 6q25.1 and encodes the 595A protein, resulting in different subtypes depending on the cleavage site and the starting point of transcriptional transcription.
  • the receptor also contains an N-terminal (A/B) domain, a hinge region (D, which links the C and E domains), and a carbon terminus ( F domain).
  • A/B N-terminal domain
  • D which links the C and E domains
  • F domain carbon terminus
  • the binding of estrogen to the receptor can lead to a variety of cellular changes, and its regulatory mechanisms can be divided into two pathways: the genomic and non-genomic pathways.
  • the ER-mediated genomic pathway involves the formation of an estrogen receptor dimer, binding to the ERE in the estrogen-regulated gene promoter, mediating the aggregation of other regulatory proteins to the promoter, and ultimately leading to an increase in the mRNA level of the gene. Or lower.
  • estrogen-mediated non-genomic pathway estrogen can react with estrogen-binding proteins present in or adjacent to the cell membrane of ERs and even cell membranes without ERs.
  • Estrogen's cellular responses through non-genomic pathways can increase intracellular calcium and NO levels, as well as activation of a variety of intracellular kinases, including MAPK, PI3K, PKA, and PKC, which phosphorylate and activate nER.
  • ER and/or progesterone receptors in approximately 70% of breast cancer patients indicates that the growth of this tumor cell is hormone-dependent, and the growth of other tumors such as ovarian cancer and endometrial cancer is also dependent on ER ⁇ .
  • the treatment of these diseases can inhibit ER signaling by various means, including antagonizing the binding of ligands and ER, antagonizing or downregulating ER ⁇ , and blocking the synthesis of estrogen.
  • ER ⁇ and ER ⁇ are expressed in endocrine tumors such as adrenal cortical tumors, pancreatic cancer, prostate cancer and thyroid cancer, digestive system tumors such as colon cancer, esophageal cancer, liver cancer and pancreatic cancer, and lung cancer.
  • ESR1 mutation may be one of the causes of drug resistance in patients with metastatic ER-positive breast cancer. (Toy et al., Nat. Genetic 2013, 45: 1439-1445; Li, S. et al Cell Rep. 4, 1116-1130 (2013)).
  • tumor growth has ER-dependent activity, so a mechanism for selectively down-regulating ER ⁇ provides a better method to block ER ⁇ activity and mediate early metastasis and tolerance.
  • Medicinal cancer is a mechanism for selectively down-regulating ER ⁇ provides a better method to block ER ⁇ activity and mediate early metastasis and tolerance.
  • SERD selective estrogen receptor downregulator
  • GDC-0810 and GDC-0927 of Gene Tec respectively, in clinical phase II and Clinical Phase I
  • Azlikon's AZD-9496 in Phase I
  • WO2011156518, WO2012037410, WO2015082990 also discloses a series of patent applications for SERD, including WO2011156518, WO2012037410, WO2015082990, and the like.
  • WO2011156518 WO2012037410
  • WO2015082990 WO2015082990
  • One of the objects of the present invention is to provide an acrylic derivative having an estrogen receptor antagonistic action different from the prior art structure.
  • the invention provides a compound of the formula (I), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof:
  • R 1 and R 2 are each independently selected from a hydrogen atom or a halogen, wherein the halogen is preferably F;
  • R 3 is selected from the group consisting of:
  • a cycloalkyl, heterocyclic, aryl or heteroaryl group wherein said cycloalkyl, heterocyclyl, aryl or heteroaryl group is further further selected from one or more selected from the group consisting of hydroxyl groups, halogens, Haloalkyl, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NR 5 R 6 , -C(O)NR 5 R 6 , -C( Substituting the substituents of O) R 7 , -SO 2 R 7 , -C(O)OR 7 or -NR 5 C(O)R 6 ;
  • alkyl wherein said alkyl is further substituted with one or more substituents selected from hydroxy, nitro, cyano, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NR 5 Substituted by a substituent of R 6 , -C(O)NR 5 R 6 , -C(O)R 7 , -SO 2 R 7 , -C(O)OR 7 or -NR 5 C(O)R 6 , Wherein the alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl group is further optionally further selected from one or more selected from the group consisting of halogen, hydroxy, nitro, cyano, alkoxy, -C(O Substituting a substituent of NR 5 R 6 , -C(O)R 7 , -SO 2 R 7 , -C(O)OR 7 or -NR 5 C(O)R 6 wherein said
  • Each R 4 is independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a trifluoromethyl group, a cyano group, -C(O)NR 5 R 6 , -C(O)R 7 , -SO 2 R 7 , —C(O)OR 7 or —NR 5 C(O)R 6 , wherein said alkyl or alkoxy group is further further selected from one or more selected from the group consisting of halogen, —C(O)NR 5 Substituted with a substituent of R 6 , -C(O)R 7 , -SO 2 R 7 , -C(O)OR 7 or -NR 5 C(O)R 6 ;
  • R 5 is selected from a hydrogen atom or an alkyl group
  • R 6 is selected from a hydrogen atom, an alkyl group, a cycloalkyl group, an aryl group or a heteroaryl group, wherein the alkyl group, cycloalkyl group, aryl group or heteroaryl group is further further selected from one or more selected from the group consisting of hydroxyl groups, Halogen, haloalkyl, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NR 8 R 9 , -C(O)NR 8 R 9 ,- Substituted by a substituent of C(O)R 10 , -SO 2 R 10 , -C(O)OR 10 or -NR 8 C(O)R 9 ;
  • R 5 and R 6 together with the atom to which they are attached form a 4 to 8 membered heterocyclic group, wherein said heterocyclic group is optionally further selected from one or more selected from the group consisting of alkyl, halogen, hydroxy, cyano, and nitrate.
  • Base cycloalkyl, heterocyclic, aryl, heteroaryl, -NR 8 R 9 , -C(O)NR 8 R 9 , -C(O)R 10 , -SO 2 R 10 , -C( O) substituted with a substituent of OR 10 or -NR 8 C(O)R 9 ;
  • R 7 is selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group or a heteroaryl group, wherein the alkyl group, cycloalkyl group, heterocyclic group, aryl group or heteroaryl group is optionally further one Or a plurality selected from the group consisting of hydroxyl, halogen, haloalkyl, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NR 8 R 9 , -C(O Substituting a substituent of NR 8 R 9 , -C(O)R 10 , -SO 2 R 10 , -C(O)OR 10 or -NR 8 C(O)R 9 ;
  • R 8 , R 9 and R 10 are each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group or a heteroaryl group, wherein the alkyl group, cycloalkyl group, heterocyclic group, aryl group Or a heteroaryl group optionally further selected from one or more selected from the group consisting of hydroxyl, halogen, haloalkyl, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxy Substituted by a substituent of an acid or a carboxylic acid ester;
  • n 0, 1, 2, 3 or 4.
  • the compound of the formula (I), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof which is represented by the formula (II) a compound, a stereoisomer, a tautomer thereof, or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 3 , R 4 and n are as defined in the formula (I).
  • the compound of the formula (I) or (II), or a stereoisomer, tautomer thereof or a pharmaceutically acceptable salt thereof wherein:
  • the alkyl group is preferably a C 1 -C 10 alkyl group
  • the alkoxy group is preferably a C 1 -C 10 alkoxy group
  • the cycloalkyl group is preferably a C 3 -C 12 cycloalkyl group
  • the heterocyclic group is preferably a C 3 -C 10 heterocyclic group
  • the aryl group is preferably a C 6 -C 10 aryl group
  • the heteroaryl group is preferably a 5- to 10-membered heteroaryl group.
  • each R 4 is independently selected from a hydrogen atom, a C 1 -C 3 alkyl group, a halogen, an alkoxy group, a trifluoromethyl group or a cyano group.
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein R 3 is a cyclopropyl group, Wherein the cyclopropyl group is further further selected from one or more selected from the group consisting of hydroxyl, halogen, haloalkyl, nitro, cyano, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl a group, -NR 5 R 6 , -C(O)N R 5 R 6 , -C(O)R 7 , -SO 2 R 7 , -C(O)OR 7 or -NR 5 C(O)R 6 Substituted by a substituent, which is preferably substituted by halogen, more preferably by F;
  • R 5 , R 6 and R 7 are as defined in the formula (I).
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein R 3 is selected from cyclopentyl Or a cyclohexyl group, wherein the cyclopentyl or cyclohexyl group is further optionally further selected from one or more selected from the group consisting of a hydroxyl group, a halogen, a halogenated alkyl group, a nitro group, a cyano group, an alkyl group, an alkoxy group, a cycloalkyl group, and a heterocyclic ring.
  • R 5 , R 6 and R 7 are as defined in the formula (I).
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein R 3 is an alkyl group, wherein The alkyl group is further selected from one or more selected from the group consisting of hydroxyl, nitro, cyano, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NR 5 R 6 , -C(O Substituted by a substituent of NR 5 R 6 , -C(O)R 7 , -SO 2 R 7 , -C(O)OR 7 or -NR 5 C(O)R 6 wherein the alkoxy group is a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally further substituted with one or more F;
  • R 5 , R 6 and R 7 are as defined in the formula (I).
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein:
  • R 1 and R 2 are each independently selected from halogen, wherein the halogen is preferably F;
  • R 3 is selected from the group consisting of:
  • R 4 is a hydrogen atom.
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein:
  • R 1 and R 2 are each independently selected from halogen, preferably is F;
  • R 3 is selected from cyclopropyl, cyclopentyl, cyclohexyl, C 3 -C 10 heterocyclyl, C 6 -C 10 aryl or 5-member-10-membered heteroaryl, wherein said cyclopropyl, cyclo A pentyl group, a cyclohexyl group, a C 3 -C 10 heterocyclic group, a C 6 -C 10 aryl group or a 5-membered-10 membered heteroaryl group is optionally further substituted with one or more halogens, wherein the halogen is preferably F, Cl or Br, more preferably F;
  • Each R 4 is independently selected from C 1 -C 6 alkyl or halogen, wherein the halogen is preferably F, Cl or Br, more preferably F.
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein:
  • R 1 and R 2 are each independently selected from halogen, preferably F;
  • R 3 is selected from cyclopropyl, wherein said cyclopropyl group is further substituted by one or more halogens, said halogen being preferably F;
  • Each R 4 is independently selected from C 1 -C 6 alkyl or halogen, preferably F.
  • the compound of the formula (I) or (II), a stereoisomer thereof, a tautomer thereof, or a pharmaceutically acceptable salt thereof wherein:
  • R 1 and R 2 are each independently selected from halogen, preferably F;
  • R 3 is:
  • R 4 is F.
  • Exemplary compounds of the invention include, but are not limited to, the compounds described in Table 1:
  • Another aspect of the present invention provides a process for the preparation of a compound of the formula (I) or a salt thereof, comprising the steps of:
  • the compound of the formula (Ia) is reacted with the formula (1b) in the presence of palladium acetate and tri-o-tolylphosphine under basic conditions to give a compound of the formula (IB); a compound of the formula (Ic) and a formula (I) Id) compound is reacted under basic conditions to obtain a compound of the formula (IA); a compound of the formula (IA) is reacted with a compound of the formula (IB) under acidic conditions, and further ester hydrolysis is carried out to obtain a compound of the formula (I);
  • X is a halogen, preferably bromine
  • Ra is an alkyl group
  • R b is a leaving group, preferably a halogen and a sulfonate, more preferably Br or a mesylate
  • definitions of R 1 to R 4 and n As described in the general formula (I).
  • Another aspect of the present invention provides a process for the preparation of a compound of the formula (II) or a salt thereof, comprising the steps of:
  • the compound of the formula (IIa) is reacted with a compound of the formula (Id) under basic conditions to give a compound of the formula (IIA); formula (IIA)
  • the compound is reacted with a compound of the formula (IB) under acidic conditions, and further ester hydrolyzed to obtain a compound of the formula (II);
  • R a is an alkyl group
  • R b is a leaving group, preferably a halogen and a sulfonate, more preferably Br or a mesylate
  • R 1 , R 2 , R 3 , R 4 and n are as defined. Said in the formula (I).
  • the reagent for providing acidic conditions is an inorganic acid or an organic acid
  • the inorganic acid is preferably hydrochloric acid, sulfuric acid or phosphoric acid, more preferably hydrochloric acid
  • the organic acid is preferably derived from formic acid or acetic acid, more preferably acetic acid.
  • the basic condition is provided by an organic base or an inorganic base
  • the organic base is preferably selected from the group consisting of diisopropylethylamine, diisopropylamine, pyridine, triethylamine, piperidine, N-methylpiperazine, 4- Dimethylaminopyridine is more preferably diisopropylamine and triethylamine
  • the inorganic base is preferably selected from the group consisting of sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, more preferably sodium carbonate and sodium hydroxide.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound of the formula (I) or (II) or a stereoisomer, tautomer thereof or A pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, excipient or combination thereof.
  • the composition optionally further comprises an antioxidant or a metal chelating agent.
  • the present invention also provides a method for selectively downregulating an estrogen receptor, which comprises a compound of the formula (I) or (II) or a stereoisomer, tautomer thereof or The pharmaceutically acceptable salt, or a pharmaceutical composition thereof, is contacted with an estrogen receptor, wherein the estrogen receptor is preferably an estrogen receptor alpha.
  • the present invention also provides a compound of the formula (I) or (II), or a stereoisomer, tautomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in the treatment of a female Use of a medicament for hormone receptor-mediated diseases, wherein the disease is preferably cancer, wherein the cancer is preferably breast cancer and gynecological cancer, wherein the gynecological cancer is preferably ovarian cancer and endometrial cancer.
  • the estrogen receptor is preferably an estrogen receptor alpha.
  • the present invention also provides a compound of the formula (I) or (II) or a stereoisomer, tautomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for preparing a selective estrogen
  • the use in the receptor down-regulation is preferably an estrogen receptor alpha down-regulation agent.
  • the present invention also provides a compound of the formula (I) or (II), or a stereoisomer, tautomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, which may be combined with a Or a combination of a plurality of other suitable anti-tumor drugs for treating an estrogen receptor-mediated disease, wherein the disease is preferably cancer, wherein the cancer is preferably a breast cancer or a gynecological cancer, wherein the gynecological cancer
  • the ovarian cancer or endometrial cancer wherein the estrogen receptor is preferably an estrogen receptor alpha
  • the other antitumor drug comprises an alkylating agent, an antimetabolite, a natural product having antitumor activity. And its derivatives, cytotoxic drugs or blocking immune cell migration drugs.
  • Suitable other antineoplastic agents include alkylating agents (but not limited to nitrogen mustard, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uraramustine, nitrogen Mustard, cyclophosphamide, ifosfamide, melphalan, chlorambucil, piperac bromide, quetiapine, busulfan, carmustine, lomustine, stellate, dacabar And temozolomide.
  • alkylating agents but not limited to nitrogen mustard, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes
  • alkylating agents such as uraramustine, nitrogen Mustard, cyclophosphamide, ifosfamide, melphalan, chlorambucil, piperac bromide, quetiapine, busulfan, carmustine, lomustine, stellate
  • Suitable other anti-tumor drugs include, for example, antimetabolites (including but not limited to folic acid antagonists, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors), such as methotrexate, 5-fluorouracil, fluorouracil , cytarabine, 6-mercaptopurine, 6-thiouracil, fludarabine phosphate, pentastatin and gemcitabine.
  • antimetabolites including but not limited to folic acid antagonists, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors
  • methotrexate including but not limited to folic acid antagonists, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors
  • methotrexate including but not limited to folic acid antagonists, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors
  • Suitable other anti-tumor drugs include, for example, certain natural products having antitumor activity and derivatives thereof (e.g., vinca alkaloids, antitumor antibiotics, enzymes, lymphokines, etc.), such as vinblastine, vincristine, vinblastine , bleomycin, actinomycin D, daunorubicin, doxorubicin, epirubicin, idarubicin, cytarabine, paclitaxel, pucamycin, deoxycofumycin, Mitomycin-C, L-asparaginase, interferon (especially IFN-a), etoposide and teniposide.
  • certain natural products having antitumor activity and derivatives thereof e.g., vinca alkaloids, antitumor antibiotics, enzymes, lymphokines, etc.
  • vinblastine vincristine
  • vinblastine bleomycin
  • actinomycin D actinomycin D
  • daunorubicin doxor
  • anti-tumor drugs include cytotoxic drugs including noviben, CPT-11, anastrozole, letrozole, capecitabine and droloxifene, topoisomerase.
  • Inhibitor, procarbazine, mitre Indole, platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutics; folinic acid; tegafur and hematopoietic growth factors are also suitable.
  • anti-tumor drugs include antibody therapeutic drugs such as trastuzumab, antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1 or cytokine antibodies (IL-10, IGF- ⁇ ).
  • Drugs that block immune cell migration such as chemokine receptor antagonists, including CCR2 and CCR4; drugs that enhance the immune system, such as adjuvants or adoptive T cell metastases; anticancer vaccines, including dendritic cells , synthetic peptides, DNA vaccines and recombinant viruses.
  • alkyl as a group or part of a group is meant to include C 1 -C 20 linear or branched aliphatic hydrocarbon group with a chain. It is preferably a C 1 -C 10 alkyl group, more preferably a C 1 -C 6 alkyl group, and most preferably a C 1 -C 3 alkyl group.
  • alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, sec-butyl, n-pentyl, 1, 1-di Methylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1 -ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethyl Butyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl Wait.
  • the alkyl group can be substituted or unsubstituted.
  • Alkynyl as a group or part of a group refers to an aliphatic hydrocarbon group containing a carbon-carbon triple bond, which may be straight or branched. Preference is given to C 2 -C 10 alkynyl groups, more preferably C 2 -C 6 alkynyl groups, most preferably C 2 -C 4 alkynyl groups. Examples of alkynyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, 1-, 2- or 3-butynyl, and the like. An alkynyl group can be substituted or unsubstituted.
  • Cycloalkyl means a saturated or partially saturated monocyclic, fused, bridged, and spiro carbon ring, ie, includes a monocyclic cycloalkyl, a fused cycloalkyl, a bridged cycloalkyl, and a spirocycloalkyl. It is preferably a C 3 -C 12 cycloalkyl group, more preferably a C 3 -C 8 cycloalkyl group, and most preferably a C 3 -C 6 cycloalkyl group.
  • Examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
  • the alkenyl group, the cyclooctyl group and the like are preferably a cyclopropyl group or a cyclohexenyl group.
  • “Spirocycloalkyl” means a polycyclic group of 5 to 18 members, two or more cyclic structures, and a single ring sharing a carbon atom (referred to as a spiro atom), and the ring contains one or more A double bond, but none of the rings have a fully conjugated ⁇ -electron aromatic system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spirocycloalkyl group is classified into a monospiro, a spiro- or a spirocycloalkyl group, preferably a mono- and bi-spirocycloalkyl group, preferably 4 yuan/5 yuan, 4, depending on the number of common spiro atoms between the rings. Yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6 yuan.
  • spirocycloalkyl include, but are not limited to, spiro[4.5]decyl, spiro[4.4]decyl, spiro[3.5]decyl, spiro[2.4]heptyl.
  • “Fused cycloalkyl” means 5 to 18 members, an all-carbon polycyclic group containing two or more cyclic structures that share a carbon atom with each other, and one or more rings may contain one or more double bonds, However, none of the rings have a fully conjugated ⁇ -electron aromatic system, preferably 6 to 12 members, more preferably 7 to 10 members. Depending on the number of constituent rings, it may be classified into a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic ring, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group.
  • fused cycloalkyl include, but are not limited to, bicyclo[3.1.0]hexyl, bicyclo[3.2.0]hept-1-enyl, bicyclo[3.2.0]heptyl, ten Hydronaphthyl or tetradecafluorophenanyl.
  • “Bridge cycloalkyl” means 5 to 18 members, containing two or more cyclic structures, sharing two all-carbon polycyclic groups that are not directly bonded to each other, and one or more rings may contain one or A plurality of double bonds, but none of the rings have a fully conjugated ⁇ -electron aromatic system, preferably 6 to 12 members, more preferably 7 to 10 members. It is preferably 6 to 14 members, more preferably 7 to 10 members. According to the number of constituent rings, it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups. It is selected as a double ring, a three ring or a four ring, and more preferably a double ring or a triple ring.
  • bridged cycloalkyl include, but are not limited to: (1s, 4s)-bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, (1s,5s)-bicyclic [3.3.1] Mercapto, bicyclo [2.2.2] octyl, (1r, 5r)-bicyclo[3.3.2] fluorenyl.
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocyclyl ring, wherein the ring to which the parent structure is attached is a cycloalkyl group, non-limiting examples include indanyl, tetrahydronaphthyl , benzocycloheptyl and the like.
  • the cycloalkyl group may optionally be substituted or unsubstituted.
  • Heterocyclyl “heterocyclic” or “heterocyclic” are used interchangeably herein to refer to a non-aromatic heterocyclic group wherein one or more of the ring-forming atoms are heteroatoms such as oxygen,
  • the nitrogen, sulfur atom and the like include a monocyclic ring, a fused ring, a bridged ring and a spiro ring, that is, a monocyclic heterocyclic group, a bridged heterocyclic group, a fused heterocyclic group and a spiroheterocyclic group.
  • heterocyclyl includes, but are not limited to, morpholinyl, thiomorpholinyl, tetrahydropyranyl, 1,1-dioxo-thiomorpholinyl, piperidinyl, 2-oxo- Piperidinyl, pyrrolidinyl, 2-oxo-pyrrolidinyl, piperazin-2-one, 8-oxa-3-aza-bicyclo[3.2.1]octyl and piperazinyl.
  • the heterocyclic group may be substituted or unsubstituted.
  • “Spiroheterocyclyl” means a polycyclic group of 5 to 18 members, two or more cyclic structures, and a single ring sharing one atom with each other, and the ring contains one or more double bonds, but no An aromatic system having a fully conjugated ⁇ -electron, wherein one or more ring atoms are selected from the group consisting of nitrogen, oxygen or S(O) m (where m is selected from 0, 1 or 2), and the remaining ring atoms are carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spiroheterocyclyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of shared spiro atoms between the ring and the ring, and is preferably a monospiroheterocyclic group and a dispiroheterocyclic group. More preferably, it is 4 yuan / 4 yuan, 4 yuan / 5 yuan, 4 yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6-membered monospiroheterocyclic group.
  • spiroheterocyclyl include, but are not limited to, 1,7-dioxaspiro[4.5]fluorenyl, 2-oxa-7-azaspiro[4.4]decyl, 7-oxa Spiro[3.5]decyl and 5-oxaspiro[2.4]heptyl.
  • “Fused heterocyclic group” means an all-carbon polycyclic group containing two or more cyclic structures that share a pair of atoms with each other, and one or more rings may contain one or more double bonds, but none of the rings have complete A conjugated ⁇ -electron aromatic system in which one or more ring atoms are selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining ring atoms are carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group preferably a bicyclic or tricyclic ring, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group.
  • fused heterocyclic groups include, but are not limited to, octahydropyrrolo[3,4-c]pyrrolyl, octahydro-1H-isoindenyl, 3-azabicyclo[3.1.0 Hexyl, octahydrobenzo[b][1,4]dioxine.
  • “Bridge heterocyclyl” means 5 to 14 members, 5 to 18 members, containing two or more cyclic structures, sharing two polycyclic groups which are not directly connected to each other, and one or more rings may be used.
  • bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclic group preferably a bicyclic ring, a tricyclic ring or a tetracyclic ring, and more preferably a bicyclic ring or a tricyclic ring.
  • fused heterocyclic groups include, but are not limited to, 2-azabicyclo[2.2.1]heptyl, 2-azabicyclo[2.2.2]octyl and 2-azabicyclo [3.3.2] ⁇ .
  • the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring wherein the ring to which the parent structure is attached is a heterocyclic group.
  • the heterocyclic group may optionally be substituted or unsubstituted.
  • Aryl means a carbocyclic aromatic system containing one or two rings wherein the rings may be joined together in a fused manner.
  • aryl includes aryl groups such as phenyl, naphthyl, tetrahydronaphthyl.
  • the aryl group is a C 6 -C 10 aryl group, more preferably the aryl group is a phenyl group and a naphthyl group, and most preferably a phenyl group.
  • the aryl group can be substituted or unsubstituted.
  • the "aryl” may be fused to a heteroaryl, heterocyclyl or cycloalkyl group, wherein the parent structure is attached to an aryl ring, non-limiting examples include, but are not limited to:
  • Heteroaryl means an aromatic 5 to 6 membered monocyclic or 9 to 10 membered bicyclic ring which may contain from 1 to 4 atoms selected from nitrogen, oxygen and/or sulfur.
  • heteroaryl include, but are not limited to, furyl, pyridyl, 2-oxo-1,2-dihydropyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thienyl, isoxazolyl , oxazolyl, oxadiazolyl, imidazolyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, 1,2,3-thiadiazolyl, benzodiazepine Oxacyclopentenyl, benzimidazolyl, fluorenyl, isodecyl, 1,3-dioxo-isoindenyl, quinoly
  • Heteroaryl groups can be substituted or unsubstituted.
  • the heteroaryl ring can be fused to an aryl, heterocyclic or cycloalkyl ring wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples include, but are not limited to:
  • Alkoxy means a group of (alkyl-O-). Among them, the alkyl group is defined in the relevant definition herein. Alkoxy groups of C 1 -C 6 are preferred. Examples thereof include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy and the like.
  • Hydrophilicity refers to an -OH group.
  • Halogen means fluoro, chloro, bromo and iodo, preferably chloro, bromo and iodo.
  • Amino means -NH 2 .
  • Niro means -NO 2 .
  • Benzyl refers to -CH 2 - phenyl.
  • Carboxy refers to -C(O)OH.
  • Carboxylic acid ester group means -C(O)O(alkyl) or (cycloalkyl) wherein alkyl, cycloalkyl are as defined above.
  • BWL refers to the rate of weight loss (%), and when BWL is negative, it indicates a decrease in body weight of the test animals.
  • Substituted refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • substituted or “substituted”, unless otherwise indicated, may be substituted by one or more groups selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane.
  • “Pharmaceutically acceptable salt” refers to certain salts of the above compounds which retain their original biological activity and are suitable for pharmaceutical use.
  • the pharmaceutically acceptable salt of the compound of the present invention may be a metal salt, an amine salt formed with a suitable acid, a metal salt preferably an alkali metal or an alkaline earth metal salt, and suitable acids include inorganic acids and organic acids such as acetic acid, benzenesulfonic acid, Benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, hydrochloric acid, isethionethane, lactic acid, malic acid, maleic acid, mandelic acid, Sulfonic acid, nitric acid, phosphoric acid, succinic acid, sulfuric acid, tartaric acid, p-toluenesulfonic acid, and the like. Particularly preferred It is hydrochloric acid, hydrobromic
  • “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiologically pharmaceutically acceptable carriers and Shape agent.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • Figure 1 is a graph showing the tumor growth inhibition rate of the compound of Example 1 in Test Example 4 on breast cancer MCF-7 tumor-bearing mice.
  • Figure 2 is a graph showing the tumor growth inhibition rate of the compound of Example 12 in Test Example 4 on breast cancer MCF-7 tumor-bearing mice.
  • Figure 3 is a graph showing the change in body weight loss rate of the compound of Example 1 in Test Example 4 for breast cancer MCF-7 tumor-bearing mice.
  • Figure 4 is a graph showing the change in body weight loss rate of the compound of Example 12 in Test Example 4 for breast cancer MCF-7 tumor-bearing mice.
  • Mass spectrometry was measured by LC/MS.
  • the ionization method was ESI or APCI. Unless otherwise specified, MS m/z (ESI) was measured at a ratio of 100%.
  • Thin layer chromatography silica gel plate uses Yantai Yellow Sea HSGF254 or Qingdao GF254 silica gel plate.
  • the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm.
  • the specification for thin layer chromatography separation and purification is 0.4mm. ⁇ 0.5mm.
  • CD 3 OD Deuterated methanol.
  • the solution in the reaction means an aqueous solution.
  • the compound is purified using a silica gel column chromatography eluent system and thin layer chromatography, wherein the eluent system is selected from the group consisting of: A: cyclohexane and ethyl acetate systems; B: dichloromethane and methanol systems; Petroleum ether and ethyl acetate systems; wherein the volume ratio of the solvent varies depending on the polarity of the compound, and may also be adjusted by adding a small amount of an acidic or alkaline agent such as acetic acid or triethylamine.
  • A cyclohexane and ethyl acetate systems
  • B dichloromethane and methanol systems
  • Petroleum ether and ethyl acetate systems wherein the volume ratio of the solvent varies depending on the polarity of the compound, and may also be adjusted by adding a small amount of an acidic or alkaline agent such as acetic acid or triethylamine.
  • the synthesis method according to the first step to the second step of the first method of Example 1 was repeated, except that the first step employed 1-(1H-indol-3-yl)propan-2-amine 1d and different ( a bromomethyl)cycloalkyl compound, a bromine-substituted hydroxyalkyl compound, a (bromomethyl)aryl compound, a (bromomethyl)heteroaryl compound or a (bromomethyl)heterocyclic compound, the second step
  • the obtained product is further passed through the reaction conditions of the third and fourth steps of the first method of the first embodiment to obtain the product of the examples 2-11, wherein the (bromomethyl)cycloalkyl compound is selected from (bromomethyl) Cyclopropane, (bromomethyl)cyclopentane, (bromomethyl)cyclohexane, 1-(bromomethyl)-1-fluorocyclopentane, 1-(bromomethyl)-1-fluorocyclohex
  • Lithium aluminum hydride (0.95 g, 25.0 mmol) was dissolved in 20 mL of anhydrous tetrahydrofuran, and (Z)-4-fluoro-3-(2-nitroprop-1-en-1-yl)- 1H- ⁇ 12b (1.374 g, 6.20 mmol) was dissolved in 10 mL of tetrahydrofuran and then heated to reflux for 6 hours.
  • reaction solution was successively added with 0.95 mL of water, 0.95 mL of sodium hydroxide solution (15%) and 19 mL of water to quench the reaction, and anhydrous magnesium sulfate (5.0 g) was stirred for 15 minutes, filtered, and washed with tetrahydrofuran (5 mL ⁇ 3) The cake was concentrated under reduced pressure to give 1-(4-fluoro-1H-indol-3-yl)propan-2-amine 12c (1.20 g, brown oil).
  • 1-(4-Fluoro-1H-indol-3-yl)propan-2-amine 12c (1.19 g, 6.20 mmol), (1-fluorocyclopropyl)methyl methanesulfonate under argon 1j (1.43g, 8.50mmol) and diisopropylethylamine (1.54mL, 9.30mmol) were dissolved in 12mL of 1,4 dioxane, heated to 100 ° C for 5 hours, cooled to room temperature, concentrated under reduced pressure.
  • TR-FRET Fluorescence resonance energy transfer
  • PV3551 Indirect fluorescent labeling of the receptor by GST linkage to the receptor by detecting the TR-FRET effect between the chromophore (Tb-anti-GST antibody) and the fluorescent ligand (Fluormone ES2) on the fluorescently labeled ER ⁇
  • the weakening is used to evaluate the competitive coordination ability between the test compound and the fluorescent ligand and the receptor.
  • the instrument we used was the Beckman Coulter BioPAPTR FRD microfluidic instrument.
  • test compound 120 nL was ultrasonically dispersed into a black small-capacity 384-well assay plate;
  • MCF-7 human breast cancer cell line
  • MCF-7 cells are used in the recovery experiment from frozen cells (about 5x10 6 th) over use.
  • the MCF-7 frozen cell strain (Sigma D5921) purchased from Sigma was stored in DMEM medium containing 2 mM L-glutamic acid.
  • 5% (v/v) Charcoal/Dextran-treated bovine serum embryo cells were added to the resuscitated MCF-7 cells, and the cell concentration was determined using a Coulter Counter.
  • test compound solution was prepared for different concentrations of test solutions (10 mM, 0.1 mM, 1 ⁇ , 0.01 ⁇ ).
  • concentration of test compounds 10 mM, 0.1 mM, 1 ⁇ , 0.01 ⁇ .
  • concentration of test compound and MCF-7 cell culture medium 40 ⁇ L
  • 20 ⁇ L of 11.1% (v/v) aqueous formaldehyde solution phosphoric acid PBS buffer
  • concentration of formaldehyde in the solution was 3.7% (v /v).
  • the level of estrogen receptor ERa in MCF-7 cells was calculated by measuring the fluorescence emission intensities of the two emission bands at 594 nm (24 hour time point) and 488 nm (5 hour time point) of the well plates by Cellomics Arrayscan. The average fluorescence emission intensity of each cell is positively correlated with the ER ⁇ receptor level of the cell.
  • the obtained raw fluorescence test data is obtained by means of data processing software such as Origin or Genedata.
  • IC 50 concentration of test compound downregulation of the estrogen receptor ER ⁇ by semi suppressed this value refers to the fluorescence emission intensity was reduced by 50% when the concentration of the test compound the maximum mean fluorescence intensity, measured as IC 50 in Table 3 is shown.
  • IC 50 range is: 0.1nM ⁇ A ⁇ 10nM, 10nM ⁇ B ⁇ 250nM, 250nM ⁇ C ⁇ 1000nM;
  • the compound of the present invention can coordinate well with the estrogen receptor, and has a good down-regulation effect on ER ⁇ .
  • Cell Counting Kit 8 Cell Counting Kit-8, Cat# CK04-13, Dojindo
  • Benchtop microplate reader SpectraMax M5Microplate Reader, Molecular Devices
  • MCF-7 human breast cancer cell line (purchased at Shanghai Cell Resource Center of Chinese Academy of Sciences)
  • the compound is diluted with DMSO to a final concentration of 10 mM;
  • test compound was diluted with the medium to the corresponding action concentration set, and the cells were added at 25 ⁇ L/well.
  • the final concentration of the compound was started from 1 ⁇ M, 4 times gradient dilution, 9 concentration points;
  • tumor cell growth inhibition rate % [(A c - A s ) / (A c - A b )] ⁇ 100%
  • a c negative control OA (cell + CCK-8 + DMSO)
  • a b positive control OA (medium + CCK-8 + DMSO)
  • Table 4 Compounds of the invention IC 50 values for inhibition of MCF-7 cells.
  • the compounds of the present invention have significant inhibitory effects on MCF-7 cells.
  • Test Example 4 Test of growth inhibitory effect of the compound of the present invention on MCF-7 tumor-bearing SCID mouse xenografts
  • This test was used to evaluate the growth inhibitory effect of oral administration of test substances on MCF-7 tumor-bearing SCID mice for 21 consecutive days.
  • Solvent 20% PEG400, 80% deionized water
  • test substance Preparation of the test substance: Weigh the appropriate amount of the test substance, dissolve it in PEG400 (20%), dissolve it, add 80% of the sterilized deionized water, and shake evenly. The test substance is freshly prepared daily before administration.
  • MCF-7 cells were cultured in RPMI 1640 medium containing 10% fetal bovine serum. Culture in a 37 ° C, 5% CO 2 incubator. Logarithmic growth phase cells were taken before inoculation, washed with 0.25% trypsin and washed with PBS, and the cells were resuspended in serum-free medium to adjust the cell concentration to 7.5 ⁇ 10 ⁇ 7 cells/mL (1:1 Matrigel, Extracellular Matrix Proteins). , 356234, BD).
  • mice were inoculated with 0.2 mL of cell suspension (1.5 x 10 ⁇ 7 cells/mouse) under sterile conditions on the right side of each mouse. Estrogen was administered subcutaneously after inoculation.
  • the tumor grows to a volume of about 150 to 250 mm 3 , mice with similar tumor volumes and good shape are selected (the shape is as single a sphere as possible, no irregular shape or multiple tumors are gathered together), and 10 mice in each group.
  • Each group of animals was given a test substance once a day (qd) according to the body weight of the animals, and administered orally (po) for 21 consecutive days, and the animal body weight per day was recorded.
  • the formation of tumors at the inoculation site of each group of animals was observed.
  • the long diameter (Y) and short diameter (X) of the tumor nodules were measured twice a week using vernier calipers and calculated according to the following formula:
  • V (X 2 Y)/2.
  • Evaluation index of antitumor activity tumor growth inhibition rate TGI (%), relative tumor growth rate T/C (%).
  • TGI (%): TGI (% ) (V c -V t) / V c ⁇ 100.
  • V c is the tumor volume of the model control group
  • V t is the tumor volume of the test subject group.
  • Relative tumor volume V n /V 0 .
  • V 0 is the tumor volume at the time of group administration
  • V n is the tumor volume at the time of measurement.
  • T/C (%) T RTV / C RTV ⁇ 100.
  • T RTV was the treatment group RTV
  • C RTV was the negative control group RTV.
  • TGI% Tumor growth inhibition rate
  • Example 1 and Example 12 of the present invention were established on breast cancer-based MCF-7 cells within 21 days at doses of 30 mg/kg and 50 mg/kg.
  • the tumor model in mice has a significant growth inhibition effect.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

本发明涉及一种通式(I)所示的丙烯酸类衍生物、其立体异构体、互变异构体或其可药用的盐,它们的制备方法,含有它们的药物组合物,以及它们作为治疗剂,特别是作为选择性雌激素受体下调剂(SERD)的用途,其中通式(I)各取代基与说明书中的定义相同。

Description

丙烯酸类衍生物、其制备方法及其在医药上的用途 技术领域
本发明属于医药领域,具体涉及一种新的丙烯酸类衍生物、其立体异构体、互变异构体或其可药用的盐,它们的制备方法,含有它们的药物组合物,以及它们作为治疗剂、特别是作为雌激素受体拮抗剂或雌激素受体α下调剂的用途。
背景技术
雌激素受体(estrogen receptor,ER)是配体激活的转录调节蛋白,它通过与内源性雌激素的相互作用介导多种生物效应的诱导,内源性雌激素包括17β-雌二醇和雌酮。已发现ER具有两种亚型雌激素受体α(ERα、ESR1和NR3A)和雌激素受体β(ERβ、ESR2和NR3b)。雌激素受体α和雌激素受体β是甾体激素受体的成员,其为细胞核受体家族中的一员。与细胞核受体的机构很相似,ERα是由6个功能结构域(命名为A-F)组成,为配体激活的转录因子,因为在其与特定的配体结合后,包括内源性雌激素17β雌二醇(E2),与基因组序列结合成复合物,即雌激素受体应答元件和共调节因子结合,来调节靶向基因的转录。ERα基因位于6q25.1,编码595A蛋白,根据剪切位点和转录转录起始点的不同,而得到不同的亚型。除了DNA结合域(域C)和配体结合域(域E)之外,受体还包含N-端(A/B)域,铰链区(D,其连接C和E域)和碳端(F域)。ERα和ERβ的C和E结构域具有一致性,A/B,D和F域中一致性较低。两种受体都与女性生殖道的调节和生长有关系,同时在中枢神经系统、心脑血管系统和骨代谢中也发挥着重要的作用。雌激素与受体结合可以导致多种细胞的变化,其调节机制可以划分为两种途径:基因组和非基因组途径。ER介导的基因组途径包括雌激素受体二聚体的形成,与雌激素调控基因启动子中的ERE的结合,介导其他调节蛋白向启动子的聚集,最后导致该基因mRNA水平的升高或降低。雌激素介导的非基因组途径中,雌激素能过与存在于或邻近ERs的细胞膜,甚至无ERs的细胞膜的雌激素结合蛋白进行反应。雌激素通过非基因组途径导致的细胞反应,可以使细胞内的钙离子和NO水平增加,以及多种细胞内激酶的激活,包括MAPK、PI3K、PKA和PKC,使nER磷酸化而激活。
约70%的乳腺癌患者中表达ER和/或黄体酮受体,表明此肿瘤细胞的生长为激素依赖性的,其他的肿瘤如卵巢癌和子宫内膜癌的生长也对ERα具有依赖性。对于这些疾病的治疗可以通过各种方式来抑制ER信号传导,包括拮抗配体和ER的结合,拮抗或下调ERα,阻断雌激素的合成等。同时在内分泌肿瘤如肾上腺皮质肿瘤、胰腺癌、前列腺癌和甲状腺癌,消化道系统肿瘤如结肠癌、食管癌、肝癌和胰腺癌,以及肺癌中都表达ERα和ERβ。虽然上述的治疗方法在ER阳性的肿瘤患者中起了一定的作用,但也会产生耐药性,最近报道,ESR1的突变可能是转移性ER阳性的乳腺癌患者产生耐药性的原因之一(Toy et al.,Nat.Genetic 2013,45:1439-1445;Li,S.et al Cell Rep.4,1116-1130(2013))。然而论述的可能的耐药性机制中,肿瘤的生长存在ER依赖性活性,因此通过选择性下调ERα的机制提供了一种较好的方法来阻滞ERα活性介导早期的、转移性和耐药性的癌症。
目前已经公开了多个可作为选择性雌激素受体下调剂(selective estrogen receptor downregulator(degrader),SERD)的药物,其中包括基因特克公司的GDC-0810和GDC-0927分别处于临床II期和临床I期;阿斯利康公司的AZD-9496,处于临床I期,同时也公开了一系列的SERD的专利申请,包括WO2011156518、WO2012037410、WO2015082990等。但是,仍有必要研究和开发新的雌激素受体α下调剂。
发明内容
本发明的目的之一在于提供一种与现有技术结构不同的,具有雌激素受体拮抗作用的丙烯酸类衍生物。
因此,根据本发明的第一方面,本发明提供了一种通式(I)所示的化合物、其立体异构体、互变异构体、或者它们的可药用盐:
Figure PCTCN2016101768-appb-000001
其中:
R1和R2各自独立地选自氢原子或卤素,其中所述的卤素优选为F;
R3选自如下基团:
(i)环烷基、杂环基、芳基或杂芳基,其中所述的环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代;
(ii)烷基,其中所述的烷基进一步被一个或多个选自羟基、硝基、氰基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中所述的烷氧基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自卤素、羟基、硝基、氰基、烷氧基、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中所述的卤素优选为F;
每个R4各自独立地选自氢原子、卤素、烷基、烷氧基、三氟甲基、氰基、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6,其中所述的烷基或烷氧基任选进一步被一个或多个选自卤素、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代;
R5选自氢原子或烷基;
R6选自氢原子、烷基、环烷基、芳基或杂芳基,其中所述的烷基、环烷基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR8R9、-C(O)NR8R9、-C(O)R10、-SO2R10、-C(O)OR10或-NR8C(O)R9的取代基所取代;
或者,R5和R6与连接它们的原子一起形成4~8元杂环基,其中所述的杂环基任选进一步被一个或多个选自烷基、卤素、羟基、氰基、硝基、环烷基、杂环基、芳基、杂芳基、-NR8R9、-C(O)NR8R9、-C(O)R10、-SO2R10、-C(O)OR10或-NR8C(O)R9的取代基所取代;
R7选自氢原子、烷基、环烷基、杂环基、芳基或杂芳基,其中所述烷基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR8R9、-C(O)NR8R9、-C(O)R10、-SO2R10、-C(O)OR10或-NR8C(O)R9的取代基所取代;
R8、R9和R10各自独立地选自氢原子、烷基、环烷基、杂环基、芳基或杂芳基,其中所述烷基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代 烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、羧酸或羧酸酯的取代基所取代;且
n为0、1、2、3或4。
在本发明的另一实施方案中,通式(I)所述的化合物、其立体异构体、互变异构体、或者其的可药用的盐,其为通式(II)所述的化合物、其立体异构体、互变异构体、或者其的可药用的盐:
Figure PCTCN2016101768-appb-000002
其中:R1、R2、R3、R4和n的定义如通式(I)中所述。
在本发明的一个优选实施方案中,通式(I)或(II)所述的化合物、或其立体异构体、互变异构体、或者它们的可药用盐,其中:
所述的烷基优选为C1-C10烷基;
所述的烷氧基优选为C1-C10烷氧基;
所述的环烷基优选为C3-C12环烷基;
所述的杂环基优选为C3-C10杂环基;
所述的芳基优选为C6-C10芳基;
所述的杂芳基优选为5元-10元杂芳基。
在本发明的一个优选实施方案中,通式(I)或(II)所述的化合物、或其立体异构体、互变异构体、或者它们的可药用盐,其中每个R4各自独立地选自氢原子、C1-C3烷基、卤素、烷氧基、三氟甲基或氰基。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中R3为环丙基,其中所述的环丙基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)N R5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中优选被卤素取代,更优选被F取代;且
R5、R6、R7的定义如通式(I)中所述。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中R3选自环戊基或环己基,其中所述的环戊基或环己基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中优选被卤素取代,更优选被F取代;且
R5、R6、R7的定义如通式(I)中所述。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中R3为烷基,其中所述的烷基进一步被一个或多个选自羟基、硝基、氰基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中所述的烷氧基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个F所取代;且
R5、R6、R7的定义如通式(I)中所述。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中:
R1和R2各自独立地选自卤素,其中所述卤素优选为F;
R3选自如下基团:
(i)环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基,其中所述的环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基任选进一步被一个或多个卤素所取代,其中所述卤素优选为F;
(ii)C1-C10烷基,其中所述的烷基进一步被一个或多个羟基所取代;
R4为氢原子。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中:
R1和R2各自独立地选自卤素,优选为F;
R3选自环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基,其中所述的环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基任选进一步被一个或多个卤素所取代,其中所述的卤素优选为F、Cl或Br,更优选为F;
每个R4各自独立地选自C1-C6烷基或卤素,其中所述的卤素优选为F、Cl或Br,更优选为F。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中:
R1和R2各自独立地选自卤素,优选为F;
R3选自环丙基,其中所述的环丙基进一步被一个或多个卤素所取代,所述卤素优选为F;
每个R4各自独立地选自C1-C6烷基或卤素,优选为F。
在本发明的一个实施方案中,通式(I)或(II)所述化合物、其立体异构体、互变异构体、或者它们的可药用盐,其中:
R1和R2各自独立地选自卤素,优选为F;
R3为:
Figure PCTCN2016101768-appb-000003
R4为F。
本发明的典型化合物包括但不限于如表1所述化合物:
表1:实施例1-13化合物的结构及命名
Figure PCTCN2016101768-appb-000004
Figure PCTCN2016101768-appb-000005
Figure PCTCN2016101768-appb-000006
或其立体异构体、互变异构体、或者它们的可药用盐。
本发明的另一方面提供了通式(I)所述的化合物或其盐的制备方法,包括以下步骤:
Figure PCTCN2016101768-appb-000007
通式(Ia)化合物在碱性条件下,在醋酸钯和三邻甲苯基膦存在下,与通式(1b)反应,得到通式(IB)化合物;通式(Ic)化合物与通式(Id)化合物在碱性条件下反应,得到通式(IA)化合物;通式(IA)化合物与通式(IB)化合物在酸性条件下反应,进一步酯水解,得到通式(I)化合物;
其中:X为卤素,优选为溴;Ra为烷基;Rb为离去基团,优选为卤素和磺酸酯,更优选为Br或甲磺酸酯;R1~R4和n的定义如通式(I)中所述。
本发明的另一方面提供了通式(II)所述的化合物或其盐的制备方法,包括以下步骤:
Figure PCTCN2016101768-appb-000008
通式(IIa)化合物与通式(Id)化合物在碱性条件下反应,得到通式(IIA)化合物;通式(IIA) 化合物与通式(IB)化合物在酸性条件下反应,进一步酯水解,得到通式(II)化合物;
其中:Ra为烷基;Rb为离去基团,优选为卤素和磺酸酯,更优选为Br或甲磺酸酯;R1、R2、R3、R4和n的定义如通式(I)中所述。
上述制备方法中,提供酸性条件的试剂为无机酸或有机酸,无机酸优选自盐酸、硫酸、磷酸,更优选为盐酸;有机酸优选自甲酸、乙酸,更优选为乙酸。
上述制备方法中,碱性条件由有机碱或无机碱提供,有机碱优选选自二异丙基乙胺、二异丙胺、吡啶、三乙胺、哌啶、N-甲基哌嗪、4-二甲氨吡啶,更优选为二异丙胺和三乙胺;无机碱优选选自碳酸钠、碳酸钾、碳酸铯、氢氧化钠、氢氧化钾,更优选为碳酸钠和氢氧化钠。
更近一步,本发明提供一种药物组合物,所述的药物组合物包含有效剂量的通式(I)或(II)所述的化合物或其立体异构体、互变异构体或其可药用的盐,及可药用的载体、赋形剂或它们的组合。该组合物任选进一步包括抗氧化剂或金属螯合剂。
本发明还提供一种选择性下调雌激素受体的方法,该方法包括将一种通式(I)或(II)所述的化合物或其立体异构体、互变异构体或其可药用的盐,或其药物组合物与雌激素受体相接触,其中所述的雌激素受体优选为雌激素受体α。
本发明还提供一种通式(I)或(II)所述的化合物或其立体异构体、互变异构体或其可药用的盐,或其药物组合物在制备用于治疗雌激素受体介导的疾病的药物中的用途,其中所述的疾病优选为癌症,其中所述的癌症优选为乳腺癌和妇科癌症,其中所述的妇科癌症优选为卵巢癌和子宫内膜癌,其中所述的雌激素受体优选为雌激素受体α。
本发明还提供一种通式(I)或(II)所述的化合物或其立体异构体、互变异构体或其可药用的盐,或其药物组合物在制备选择性雌激素受体下调剂中的用途,优选为雌激素受体α下调剂。
本发明还提供一种通式(I)或(II)所述的化合物或其立体异构体、互变异构体或其可药用的盐,或其药物组合物,其可以与一种或多种其他适宜的抗肿瘤药物联合用药治疗雌激素受体介导的疾病,其中所述的疾病为优选为癌症,其中所述的癌症优选为乳腺癌或妇科癌症,其中所述的妇科癌症优选为卵巢癌或子宫内膜癌,其中所述的雌激素受体优选为雌激素受体α,其中所述其他抗肿瘤药物包括烷化剂、抗代谢类药物、具有抗肿瘤活性的天然产物及其衍生物、细胞毒素类药物或阻滞免疫细胞迁移类药物。
适宜的其他抗肿瘤药物包括烷化剂(但不限于氮芥、环乙亚胺(ethylenimine)衍生物、磺酸烷基酯、亚硝基脲和三氮烯),例如乌拉莫司汀、氮芥、环磷酰胺、异环磷酰胺、美法仑、苯丁酸氮芥、哌泊溴烷、赛替派、白消安、卡莫司汀、洛莫司汀、链佐星、达卡巴嗪和替莫唑胺。
适宜的其他抗肿瘤药物也包括例如抗代谢类药物(包括但不限于叶酸拮抗剂、嘧啶类似物、嘌呤类似物和腺苷脱氨酶抑制剂),例如甲氨喋呤、5-氟尿嘧啶、氟尿嘧啶、阿糖胞苷、6-巯基嘌呤、6-硫尿嘧啶、磷酸氟达拉滨、喷司他丁和吉西他滨。
适宜的其他抗肿瘤药物还包括例如某些具有抗肿瘤活性的天然产物及其衍生物(例如长春花生物碱、抗肿瘤抗生素、酶、淋巴因子等),例如长春碱、长春新碱、长春酰胺、博来霉素、放线菌素D、柔红霉素、多柔比星、表柔比星、伊达比星、阿糖胞苷、紫杉醇、普卡霉素、脱氧考福霉素、丝裂霉素-C、L-天冬酰胺酶、干扰素(尤其IFN-a)、依托泊苷和替尼泊苷。
适宜的其他抗肿瘤药物还包括细胞毒素类药物,所述细胞毒素类药物包括诺维本、CPT-11、阿那曲唑、来曲唑、卡培他滨和屈洛昔芬、拓扑异构酶抑制剂、丙卡巴肼、米托 蒽醌、铂配位络合物例如顺铂和卡铂;生物反应调节物;生长抑制剂;抗激素治疗药物;亚叶酸;替加氟和造血生长因子也适用。
此外,适宜的其他抗肿瘤药物还包括抗体治疗药物例如曲妥单抗、共同刺激性分子的抗体例如CTLA-4、4-1BB和PD-1或细胞因子的抗体(IL-10,IGF-β等);阻滞免疫细胞迁移的药物,例如趋化因子受体拮抗剂,包括CCR2和CCR4;还包括增强免疫系统的药物,例如辅药或过继T细胞转移;抗癌疫苗,包括树突细胞、合成肽、DNA疫苗和重组病毒。
本领域技术人员公知的安全有效的给予大多数化疗药物(抗肿瘤药物)的方法,以及它们的给药标准在标准文献中已有论述,如“physicians desk reference”(PDR,e.g.1996edition,medical Economics Company,Montvale,NJ)中阐述了多种化疗药物的给药方法,其公开内容通过引用结合到本文中。
除非另有说明,否则本发明在说明书和权利要求书中所使用的部分术语定义如下:
“烷基”当作一基团或一基团的一部分时是指包括C1-C20直链或者带有支链的脂肪烃基团。优选为C1-C10烷基,更优选为C1-C6烷基,最优选为C1-C3烷基。烷基基团的实施例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代或未取代的。
“炔基”作为一基团或一基团的一部分时是指含有一个碳碳三键的脂肪烃基团,可为直链也可以带有支链。优先选择的是C2-C10的炔基,更优选C2-C6炔基,最优选C2-C4炔基。炔基基团的实施例包括,但不限于乙炔基、1-丙炔基、2-丙炔基、1-、2-或3-丁炔基等。炔基可以是取代或未取代的。
“环烷基”是指饱和或部分饱和的单环、稠环、桥环和螺环的碳环,即包括单环环烷基、稠环烷基、桥环烷基和螺环烷基。优选为C3-C12环烷基,更优选为C3-C8环烷基,最优选为C3-C6环烷基。单环环烷基的实施例包括但不限于环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等,优选环丙基、环己烯基。
“螺环烷基”指5至18元,两个或两个以上环状结构,且单环之间彼此共用一个碳原子(称螺原子)的多环基团,环内含有1个或多个双键,但没有一个环具有完全共轭的π电子的芳香系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺、双螺或多螺环烷基,优选为单螺和双螺环烷基,优选为4元/5元、4元/6元、5元/5元或5元/6元。“螺环烷基”的非限制性实例包括但不限于:螺[4.5]癸基、螺[4.4]壬基、螺[3.5]壬基、螺[2.4]庚基。
“稠环烷基”指5至18元,含有两个或两个以上环状结构彼此公用一对碳原子的全碳多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香系统,优选为6至12元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。“稠环烷基”的非限制性实例包括但不限于:二环[3.1.0]己基、二环[3.2.0]庚-1-烯基、二环[3.2.0]庚基、十氢化萘基或十四氢菲基。
“桥环烷基”指5至18元,含有两个或两个以上环状结构,彼此共用两个不直接相连接碳原子的全碳多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香系统,优选为6至12元,更优选为7至10元。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优 选为双环、三环或四环,更有选为双环或三环。“桥环烷基”的非限制性实例包括但不限于:(1s,4s)-二环[2.2.1]庚基、二环[3.2.1]辛基、(1s,5s)-二环[3.3.1]壬基、二环[2.2.2]辛基、(1r,5r)-二环[3.3.2]癸基。
所述环烷基环可以稠合于芳基、杂芳基或杂环基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基任选可以是取代的或未取代的。
“杂环基”、“杂环”或“杂环的”在本申请中可交换使用,都是指非芳香性杂环基,其中一个或多个成环的原子是杂原子,如氧、氮、硫原子等,包括单环、稠环、桥环和螺环,即包含单环杂环基、桥杂环基、稠杂环基和螺杂环基。优选具有5至7元单环或7至10元双-或三环,其可以包含1,2或3个选自氮、氧和/或硫中的原子。“杂环基”的实例包括但不限于吗啉基,硫代吗啉基,四氢吡喃基,1,1-二氧代-硫代吗啉基,哌啶基,2-氧代-哌啶基,吡咯烷基,2-氧代-吡咯烷基,哌嗪-2-酮,8-氧杂-3-氮杂-双环[3.2.1]辛基和哌嗪基。杂环基可以是取代或未取代的。
“螺杂环基”指5至18元,两个或两个以上环状结构,且单环之间彼此共用一个原子的多环基团,环内含有1个或多个双键,但没有一个环具有完全共轭的π电子的芳香系统,其中一个或多个环原子选自氮、氧或S(O)m(其中m选自0、1或2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。“螺杂环基”的非限制性实例包括但不限于:1,7-二氧杂螺[4.5]癸基、2-氧杂-7-氮杂螺[4.4]壬基、7-氧杂螺[3.5]壬基和5-氧杂螺[2.4]庚基。
“稠杂环基”指含有两个或两个以上环状结构彼此公用一对原子的全碳多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香系统,,其中一个或多个环原子选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。“稠杂环基”的非限制性实例包括但不限于:八氢吡咯并[3,4-c]吡咯基、八氢-1H-异吲哚基,3-氮杂二环[3.1.0]己基,八氢苯并[b][1,4]二噁英(dioxine)。
“桥杂环基”指5至14元,5至18元,含有两个或两个以上环状结构,彼此共用两个不直接相连接的原子的多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香系统,其中一个或多个环原子选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。“稠杂环基”的非限制性实例包括但不限于:2-氮杂二环[2.2.1]庚基,2-氮杂二环[2.2.2]辛基和2-氮杂二环[3.3.2]癸基。
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基。杂环基任选可以是取代的或未取代的。
“芳基”是指含有一个或者两个环的碳环芳香系统,其中所述环可以以稠合的方式连接在一起。术语“芳基”包括比如苯基、萘基、四氢萘基的芳香基团。优选芳基为C6-C10芳基,更优选芳基为苯基和萘基,最优选为苯基。芳基可以是取代或未取代的。所述“芳基”可与杂芳基、杂环基或环烷基稠合,其中与母体结构连接在一起的为芳基环,非限制性实例包括但不限于:
Figure PCTCN2016101768-appb-000009
“杂芳基”是指芳香族5至6元单环或9至10元双环,其可以包含1至4个选自氮、氧和/或硫中的原子。“杂芳基”的实施例包括但不限于呋喃基,吡啶基,2-氧代-1,2-二氢吡啶基,哒嗪基,嘧啶基,吡嗪基,噻吩基,异噁唑基,噁唑基,噁二唑基,咪唑基,吡咯基,吡唑基,三唑基,四唑基,噻唑基,异噻唑基,1,2,3-噻二唑基,苯并间二氧杂环戊烯基,苯并咪唑基,吲哚基,异吲哚基,1,3-二氧代-异吲哚基,喹啉基,吲唑基,苯并异噻唑基,苯并噁唑基和苯并异噁唑基。杂芳基可以是取代或未取代的。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,非限制性实例包括但不限于:
Figure PCTCN2016101768-appb-000010
“烷氧基”是指(烷基-O-)的基团。其中,烷基见本文有关定义。C1-C6的烷氧基为优先选择。其实例包括,但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基等。
“羟基”指-OH基团。
“卤素”是指氟、氯、溴和碘,优选氯、溴和碘。
“氨基”指-NH2
“氰基”指-CN。
“硝基”指-NO2
“苄基”指-CH2-苯基。
“羧基”指-C(O)OH。
“羧酸酯基”指-C(O)O(烷基)或(环烷基),其中烷基、环烷基的定义如上所述。
“BWL”指体重下降率(%),当BWL为负值时,表示测试动物体重降低。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
本说明书所述的“取代”或“取代的”,如无特别指出,均是指可被一个或多个选自以下的基团取代:烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、疏基、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氨基、卤代烷基、羟烷基、羧基、羧酸酯基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6,其中,R5~R7的定义如通式(I)中所述。
“可药用的盐”是指上述化合物能保持原有生物活性并且适合于医药用途的某些盐类。本发明化合物的可药用的盐可以为金属盐、与合适的酸形成的胺盐,金属盐优选碱金属、碱土金属盐,合适的酸包括无机酸和有机酸,例如乙酸、苯磺酸、苯甲酸、樟脑磺酸、柠檬酸、乙磺酸、富马酸、葡糖酸、谷氨酸、氢溴酸、盐酸、羟乙磺酸、乳酸、苹果酸、马来酸、扁桃酸、甲磺酸、硝酸、磷酸、琥珀酸、硫酸、酒石酸、对甲苯磺酸等。特别优选 的是盐酸、氢溴酸、磷酸和硫酸,最优选的是盐酸盐。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
附图说明
图1为测试例4中实施例1化合物对乳腺癌MCF-7荷瘤小鼠肿瘤生长抑制率图。
图2为测试例4中实施例12化合物对乳腺癌MCF-7荷瘤小鼠肿瘤生长抑制率图。
图3为测试例4中实施例1化合物对乳腺癌MCF-7荷瘤小鼠体重下降率变化图。
图4为测试例4中实施例12化合物对乳腺癌MCF-7荷瘤小鼠体重下降率变化图。
具体实施方式
以下结合实施例进一步描述本发明,应当理解的是,这些实施例并不构成对本发明范围的限制。
制备实施例
以下实施例给出了本发明代表性化合物的制备及相关结构鉴定数据。
1H NMR图谱是用Bruker仪器(400MHz)测定而得,化学位移用ppm表示。使用四甲基硅烷内标准(0.00ppm)。1H NMR的表示方法:s=单峰,d=双重峰,t=三重峰,m=多重峰,br=变宽的,dd=双重峰的双重峰,dt=三重峰的双重峰。若提供偶合常数时,其单位为Hz。
质谱是用LC/MS仪测定得到,离子化方式可为ESI或APCI,如无特殊说明,MS m/z(ESI)均为比例为100%的测定值。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
柱层析使用烟台黄海硅胶200~300目硅胶为载体。
在下列实例中,除非另有指明,所有温度为摄氏温度;除非另有指明,各种起始原料和试剂来自市售或者是根据已知的方法合成,市售原料和试剂均不经进一步纯化直接使用;除非另有指明,市售厂家包括但不限于Aldrich Chemical Company,ABCR GmbH&Co.KG,Acros Organics,广赞化工科技有限公司,景颜化工科技有限公司和上海常丰生物科技有限公司等。
CD3OD:氘代甲醇。
CDCl3:氘代氯仿。
DMSO-d6:氘代二甲基亚砜。
若实施例中无特殊说明,反应均在氩气氛下进行。
氩气氛由反应瓶连接一个约1L容积的氩气气球提供。
若实施例中无特殊说明,反应中的溶液是指水溶液。
对化合物进行纯化,采用硅胶柱层析洗脱剂体系和薄层色谱法,其中洗脱剂体系选自:A:环己烷和乙酸乙酯体系;B:二氯甲烷和甲醇体系;C:石油醚和乙酸乙酯体系;其中溶剂的体积比根据化合物的极性不同而不同,也可以加入少量的酸性或碱性试剂进行调节,如醋酸或三乙胺等。
实施例1
(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸
Figure PCTCN2016101768-appb-000011
方法一、
第一步
Figure PCTCN2016101768-appb-000012
第二步
Figure PCTCN2016101768-appb-000013
第三步
Figure PCTCN2016101768-appb-000014
第四步
Figure PCTCN2016101768-appb-000015
第一步
(E)-3-(3,5-二氟-4-甲酰基苯基)丙烯酸甲酯
将4-溴-2,6-二氟苯甲醛1a(5g,22.6mmol,根据WO2014191726公开的方法制备得到),三乙胺(6.31ml,45.2mmol),醋酸钯(254mg,1.13mmol)和三邻甲苯基膦(688mg,2.26mmol)溶于100mL二甲基甲酰胺中,搅拌下加入丙烯酸甲酯1b(2.91mL,33.9mmol),反应液加热至80℃,反应5小时。将反应液冷却,将体系溶剂浓缩至干,加入水(30mL),用乙酸乙酯(50mL×3)萃取,合并的有机相依次用2N盐酸(10mL),饱和食盐水洗涤(20mL×2),无水硫酸钠干燥,过滤,减压下浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:环己烷和乙酸乙酯体系),得到(E)-3-(3,5-二氟-4-甲酰基苯基)丙烯酸甲酯1c(3.73g,黄色固体),产率:73%。
MS m/z(ESI):227.2[M+1]
1H NMR(400MHz,CDCl3):δ=10.34(s,1H),7.57(d,J=16.0Hz,1H),7.13(d,J=9.2Hz,2H),6.52(d,J=16.0Hz,1H),3.84(s,3H)
第二步
(R)-N-((1-氟环丙基)甲基)-1-(1H-吲哚-3-基)丙-2-胺
氩气保护下,将(R)-1-(1H-吲哚-3-基)丙-2-胺1d(1.74g,10mmol,购买于上海常丰生物科技有限公司)溶于30mL二氧六环中,加入二异丙胺(1.51g,15mmol),加入1-(溴甲基)-1-氟环丙烷1e(1.67g,11mmol,购买于上海广赞化工科技有限公司),75℃反应24小时。将反应液冷却至室温,加入少量水5mL淬灭反应,无水硫酸钠干燥,过滤,滤液在减压下浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:二氯甲烷和甲醇体系),得到(R)-N-((1-氟环丙基)甲基)-1-(1H-吲哚-3-基)丙-2-胺1f(0.73g,黄色固体),产率:30%。
MS m/z(ESI):247.5[M+1]
第三步
(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基苯基)丙烯酸甲酯
氩气保护下,将(R)-N-((1-氟环丙基)甲基)-1-(1H-吲哚-3-基)丙-2-胺1f(0.73g,3mmol)溶于10mL甲苯中,加入(E)-3-(3,5-二氟-4-甲酰基苯基)丙烯酸甲酯1c(0.678g,3mmol),加入20mL甲苯,加入乙酸(0.9g,15mmol),80℃反应5小时,冷却至室温,加入少量水(5mL)淬灭反应,无水硫酸钠干燥,过滤,滤液在减压下浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:二氯甲烷和甲醇体系),得到(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基苯基)丙烯酸甲酯1g(0.87g,黄色固体),产率:60%。
MS m/z(ESI):455.5[M+1]
第四步
(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸
将(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基苯基)丙烯酸甲酯1g(0.87g,1.91mmol)溶于16.5mL四氢呋喃和甲醇(V/V=2/1)的混合溶液,缓慢加入5mL7.5M氢氧化钠溶液中,室温下反应2小时,加入少量水,用6N盐酸调PH=3,乙酸乙酯萃取(20mL×3),合并的有机相依次通过无水硫酸钠干燥,过滤,滤液在减压下浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:二氯甲烷和甲醇体系),得到(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸1(0.76g,黄色固体),产率:90%。
MS m/z(ESI):440.9[M+1](100%);441.9[M+1](27.8%)
1H NMR(400MHz,CD3OD)δ=7.61(d,J=16.1Hz,1H),7.49(d,J=7.5Hz,1H),7.34-7.21(m,3H),7.13-7.00(m,2H),6.59(d,J=15.8Hz,1H),5.79(br.s.,1H),5.50(s,1H),3.95-3.84(m,1H),3.56-3.43(m,1H),3.14(dd,J=4.6,15.7Hz,1H),3.07-2.94(m,1H),2.79(dd,J=6.5,15.8Hz,1H),1.32(d,J=6.5Hz,3H),1.19-1.01(m,2H),0.76-0.61(m,2H)
方法二、
Figure PCTCN2016101768-appb-000016
第一步
(1-氟环丙基)甲醇
将1-氟环丙烷甲酸1h(3.12g,30.0mmol)溶于75mL乙醚中,冷却至0℃,分批加入氢化铝锂(1.37g,36.0mmol),然后在0℃反应1小时。向反应液中依次加入1.3mL水、1.3mL氢氧化钠溶液(15%)和2.6mL水,搅拌10分钟,加入无水硫酸镁干燥,过滤,滤饼用乙醚洗涤,滤液减压浓缩,得到(1-氟环丙基)甲醇1i(2.05g,无色液体),产率:76%。1H NMR(400MHz,CDCl3)δ=3.89-3.76(m,2H),2.33-2.16(m,1H),1.14-1.02(m,2H),0.73-0.63(m,2H)
第二步
(1-氟环丙基)甲基甲磺酸酯
将(1-氟环丙基)甲醇1i(1.69g,18.8mmol)溶于25mL二氯甲烷中,加入三乙胺(3.1mL,22.6mmol),氩气保护冷却至-10℃,滴加甲烷磺酰氯(2.26g,19.7mmol),然后在0℃反应1小时。向反应液中依次加入15mL淬灭反应,分层,有机相用水(15mL)洗涤,无水硫酸镁干燥,过滤,减压浓缩,得到(1-氟环丙基)甲基甲磺酸酯1j(3.02g,无色油状物),产率:95.5%。
1H NMR(400MHz,CDCl3)δ=4.51-4.41(m,2H),3.12-3.06(m,3H),1.22(td,J=7.2,18.1Hz,2H),0.86(q,J=7.7Hz,2H)
第三步
(R)-N-((1-氟环丙基)甲基)-1-(1H-吲哚-3-基)丙-2-胺
氩气保护下,将(1-氟环丙基)甲基甲磺酸酯1j(3.02g,18.0mmol)、(R)-1-(1H-吲哚-3-基)丙-2-胺1d(2.84g,16.3mmol)和二异丙基乙胺(5.40mL,32.6mmol)溶于30mL 1,4二氧六环中,加热至100℃反应5小时,冷却至室温,加入硅胶过滤,滤饼用用乙酸乙酯(10mL×2)洗涤,滤液减压浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:二氯甲烷和甲醇体系),得到(R)-N-((1-氟环丙基)甲基)-1-(1H-吲哚-3-基)丙-2-胺1f(1.67g,黄褐色固体),产率:42%。
MS m/z(ESI):247.5[M+1]
重复实施例1方法一中第一步、第三步至第四步的步骤,得到实施例1化合物,(E)-3-(3,5-二氟-4-((1R,3R)-2-((1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚 -1-基)苯基)丙烯酸1。
实施例2-11
重复根据实施例1方法一中第一步至第二步的合成方法,不同的是,第二步中采用1-(1H-吲哚-3-基)丙-2-胺1d与不同的(溴甲基)环烷基化合物、溴取代的羟烷基化合物、(溴甲基)芳基化合物、(溴甲基)杂芳基化合物或(溴甲基)杂环基化合物反应,第二步得到的产物进一步通过实施例1方法一中第三步和第四步的反应条件,得到实施例2-11的产物,其中所述的(溴甲基)环烷基化合物选自(溴甲基)环丙烷、(溴甲基)环戊烷、(溴甲基)环己烷,1-(溴甲基)-1-氟环戊烷、1-(溴甲基)-1-氟环己烷,所述的溴取代的羟烷基化合物为1-溴-2-甲基丙-2-醇,所述的(溴甲基)芳基化合物选自1-(溴甲基)-2-氟苯和1-(溴甲基)-4-氟苯,所述的(溴甲基)杂芳基化合物选自3-(溴甲基)吡啶,所述的(溴甲基)杂环基化合物选自3-(溴甲基)氧杂环丁烷。具体信息如下表2所示:
表2:实施例2-11化合物结构及其确认数据
Figure PCTCN2016101768-appb-000017
Figure PCTCN2016101768-appb-000018
Figure PCTCN2016101768-appb-000019
实施例12
(E)-3-(3,5-二氟-4-((1R,3R)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸
Figure PCTCN2016101768-appb-000020
第一步
(Z)-4-氟-3-(2-硝基丙-1-烯-1-基)-1H-吲哚
将4-氟-1H-吲哚-3-甲醛12a(1.0g,6.1mmol)、14.2mL硝基乙烷、乙酸铵(235mg,3.05mmol)溶于6.0mL乙酸中,氩气保护下,110℃反应6小时。反应液减压浓缩,加入20mL乙酸乙酯,用饱和碳酸氢钠溶液(15mL)洗涤,水相用乙酸乙酯(15mL)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到(Z)-4-氟-3-(2-硝基丙-1-烯-1-基)-1H-吲哚12b(1.374g,棕色固体),产率:100%。
MS m/z(ESI):220.9[M+1]
第二步
1-(4-氟-1H-吲哚-3-基)丙烷-2-胺
将氢化铝锂(0.95g,25.0mmol)溶于20mL无水四氢呋喃中,冰浴下缓慢加入(Z)-4-氟-3-(2-硝基丙-1-烯-1-基)-1H-吲哚12b(1.374g,6.20mmol)溶于10mL四氢呋喃的溶液,然后加热至回流反应6小时。向反应液中依次加入0.95mL水、0.95mL氢氧化钠溶液(15%)和19mL水淬灭反应,无水硫酸镁(5.0g)搅拌15分钟,过滤,用四氢呋喃(5mL×3)洗涤滤饼,滤液减压浓缩,得到1-(4-氟-1H-吲哚-3-基)丙烷-2-胺12c(1.20g,褐色油状物),产率:100%。
MS m/z(ESI):193.0[M+1]
第三步
1-(1-(4-氟-1H-吲哚-3-基)-N-((1-氟环丙基)甲基)丙-2-胺
氩气保护下,将1-(4-氟-1H-吲哚-3-基)丙烷-2-胺12c(1.19g,6.20mmol)、(1-氟环丙基)甲基甲磺酸酯1j(1.43g,8.50mmol)和二异丙基乙胺(1.54mL,9.30mmol)溶于12mL 1,4二氧六环中,加热至100℃反应5小时,冷却至室温,减压浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:二氯甲烷:甲醇体系),得到1-(1-(4-氟-1H-吲哚-3-基)-N-((1-氟环丙基)甲基)丙-2-胺12d(975mg,褐色油状物),产率:60%。
MS m/z(ESI):265.0[M+1]
第四步
(E)-3-(3,5-二氟-4-((1R,3R)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯
(E)-3-(3,5-二氟-4-((1S,3S)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯
氩气保护下,将1-(1-(4-氟-1H-吲哚-3-基)-N-((1-氟环丙基)甲基)丙-2-胺12d(975mg,3.69mmol)、(E)-3-(3,5-二氟-4-甲酰基苯基)丙烯酸甲酯1c(848mg,3.69mmol)和乙酸(0.422mL,7.38mmol)溶于10mL甲苯中,85℃反应7小时。冷却至室温,减压下浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:石油醚:乙酸乙酯体系),所得油状物用10mL四氢呋喃和正己烷(V/V=1/1)的混合溶剂打浆,有大量固体析出,过滤,滤饼用2mL四氢呋喃和正己烷(V/V=1/1)的混合溶剂洗涤,滤饼干燥,得到(E)-3-(3,5-二氟-4-(5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯(322mg,白色固体),产率:19.2%,进一步通过采用超临界流体色谱法(SFC),用制备设备和手性柱对手性异构体进行拆分(手性柱:ChiralCel OJ,250×30mm I.D.5μm;流动相:A为CO2,B为甲醇(40%);流速为60mL/min),得到(E)-3-(3,5-二氟-4-((1R,3R)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯12e(163.4mg,白色固体)和(E)-3-(3,5-二氟-4-((1S,3S)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯12f(168.47mg,白色固体)。
12e:MS m/z(ESI):472.9[M+1]
12f:MS m/z(ESI):472.9[M+1]
第五步
(E)-3-(3,5-二氟-4-((1R,3R)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸
将(E)-3-(3,5-二氟-4-((1R,3R)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯12e(163.4mg,0.346mmol)溶于3mL四氢呋喃和甲醇(V/V=2/1)混合溶剂中,缓慢加入7.5M氢氧化钠溶液(0.46mL),室温反应1小时,用1M盐酸溶液调反应液pH=4,减压浓缩除去溶剂,加入15mL水和15mL乙酸乙酯,分层,水相用乙酸乙酯(15mL)萃取,合并有机相,无水硫酸镁干燥,过滤,滤液在减压下浓缩,得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:石油醚:乙酸乙酯体系),得到(E)-3-(3,5-二氟-4-((1R,3R)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸12(54mg,淡黄色固体),产率:34%。
MS m/z(ESI):458.9[M+1]
1H NMR(400MHz,CD3OD)δ=7.59(d,J=16.1Hz,1H),7.25(d,J=9.8Hz,2H),7.03-6.91(m,2H),6.63(dd,J=7.7,10.9Hz,1H),6.56(d,J=16.1Hz,1H),5.45(s,1H),3.76-3.66(m,1H),3.26-3.14(m,2H),2.89-2.70(m,2H),1.20(d,J=6.5Hz,3H),0.96(d,J=19.1Hz,2H),0.62-0.52(m,2H)
实施例13
(E)-3-(3,5-二氟-4-((1S,3S)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸
Figure PCTCN2016101768-appb-000021
第一步
(E)-3-(3,5-二氟-4-((1S,3S)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸
将(E)-3-(3,5-二氟-4-((1S,3S)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸甲酯12f(168.47mg,0.356mmol)溶于3mL四氢呋喃和甲醇(V/V=2/1)的混合溶剂中,缓慢加入7.5M氢氧化钠溶液(0.475mL),室温反应1小时,用1M盐酸溶液调反应液pH=4,减压浓缩除去溶剂,加入15mL水和15mL乙酸乙酯,分层,水相用乙酸乙酯(15mL)萃取,合并有机相,无水硫酸镁干燥,过滤,滤液在减压下浓缩, 得到的残留物通过硅胶柱层析进一步分离纯化(洗脱剂:石油醚:乙酸乙酯体系),得到(E)-3-(3,5-二氟-4-((1S,3S)-5-氟-2-(1-氟环丙基)甲基)-3-甲基-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)苯基)丙烯酸13(70mg,淡黄色固体),产率:42.9%。
MS m/z(ESI):458.9[M+1]
1H NMR(400MHz,CD3OD)δ=7.61(d,J=16.1Hz,1H),7.26(d,J=10.0Hz,2H),7.05-6.92(m,2H),6.65(dd,J=7.9,10.7Hz,1H),6.57(d,J=16.1Hz,1H),5.47(s,1H),3.72(d,J=5.8Hz,1H),3.28-3.16(m,2H),2.91-2.71(m,2H),1.21(d,J=6.5Hz,3H),0.98(d,J=18.8Hz,2H),0.58(d,J=8.5Hz,2H)
生物学评价
测试例1本发明化合物与雌激素受体ERα配位作用测试
本发明利用Lantha ScreenTM时控荧光共振能量转移技术(TR-FRET)对化合物和分离得到的雌激素受体ERα配体作用域之间的竞争性配位作用进行评价。在TR-FRET配位评价技术中用的荧光团(Fluormone ES2,产品序号P2645)和重组人雌激素受体ERα配体作用域(产品序号PV4543)是从Invitrogen公司购买。本测试的设计原理如下,雌激素受体ERα-LBD(GST)和含有荧光团的配体形成一个受体/荧光团复合物,然后加入铽(Tb)标记的抗GST抗体(产品序号PV3551),通过和受体上的GST链接来实现受体的间接荧光标记,通过检测荧光标记ERα上的发色团(Tb-抗GST抗体)和荧光配体(Fluormone ES2)之间的TR-FRET效应的减弱来评价测试化合物和荧光配体同受体之间的竞争性配位能力。我们用了如下的样品制备和测试方法对合成化合物进行了上述测试。我们所用的仪器是Beckman Coulter BioPAPTR FRD微流动工作仪。
(1)将120nL测试化合物通过超声分散到黑色小容量384孔分析板;
(2)在ES2的缓冲液中制备1x ERα-LBD/Tb-抗GST抗体复合物并在其中培养20分钟;
(3)测试之前往上述ERα-LBD/Tb-抗GST抗体复合物溶液中再加入1x ES2待用;
(4)将步骤3制备的ERα-LBD/Tb-抗GST抗体复合物溶液12μL加入到分析板孔中;
(5)将分析板避光保存,在室温下培养一个小时;
(6)在337nm的激发光下,用BMG Phera STAR检测490nm和520nm两个波段的发射光。
用Labcyte Echo550将在微板上配制好的各种不同浓度(10mM,0.1mM,1μM和10nM)的测试化合物转移到分析板上。每个测试化合物的120nL的DMSO溶液被加入到分析板孔中,十二个不同浓度被分别测试(100,29.17,10.42,2.083,1,0.292,0.104,0.02083,0.01,0.0029,0.00104,0.001μM)。得到的TR-FRET原始荧光测试数据借助于Origin或是Genedata等数据处理软件得到拟合曲线。我们用每个化合物的半抑制浓度IC50来表征测试化合物与雌激素受体ERα的竞争性配位作用。IC50代表示踪荧光团(ES2)与雌激素受体配位作用下降50%时所推算的测试化合物浓度,测定的IC50如表3所示。
测试例2MCF-7细胞对ERα下调降解测试
本发明优选化合物对ERα蛋白水平的下调作用是在人体管乳腺癌细胞株MCF-7中通过免疫荧光分析来评价的。实验所用到的MCF-7细胞是直接从冷冻细胞(约5x106个)复苏过来用的。从Sigma购买的MCF-7冷冻细胞株(Sigma D5921)是保存在2mM L-谷氨酸的DMEM培养液中。在复苏的MCF-7细胞中加入5%(v/v)Charcoal/Dextran处理过的牛血清胚胎细胞,并用Coulter Counter来测定细胞浓度。
测试所用的细胞用培养液稀释到3.75×104个/mL,移取40uL/孔上述细胞液到黑色透明底的384孔板上,然后孔板在37℃,5%的CO2下培养过夜。10mM的测试化合物溶液被用 于不同浓度测试液(10mM,0.1mM,1μΜ,0.01μΜ)的配制。往各种不同浓度的测试化合物和MCF-7细胞培养液(40μL)中加入20μL的11.1%(v/v)的甲醛水溶液(磷酸PBS缓冲液),最后溶液中甲醛的浓度为3.7%(v/v)。细胞在室温下固定20分钟后,用250μL的PBS/Proclin洗两次,再加入40μL的PBS/Proclin后于4℃下冷藏。蛋白的免疫染色分析法是用自动AutoElisa试剂盒完成的。从各个板孔中移取PBS/Proclin液,然后加入40μL含0.5%TweenTM20(v/v)的PBS进行细胞透性化处理,一小时后,孔板用250μL的PBS/0.05%Tween20/Proclin清洗三遍,加入20μL的ERα兔单克隆抗体(Thermofisher)的PBS/TweenTM 20/3%(w/v)BSA溶液(1:1000)。分析孔板在4℃下培养过夜,用250uL的PBS/TweenTM 20/Proclin清洗三次,然后每孔中加入20μL的山羊抗兔IgG AlexaFluor 594或是山羊抗兔IgG AlexaFluor 488抗体(含有Hoechst染色剂)的PBS/TweenTM 20/3%(w/v)BSA溶液(1:5000),体系在室温下培养一个小时。分析孔板用250μL的PBS/0.05%(v/v)TweenTM 20/Proclin清洗三次,加入20μL的PBS溶液,孔板于4℃下避光保存。通过Cellomics Arrayscan检测孔板594nm(24小时时间点)和488nm(5小时时间点)两个发射波段的荧光发射强度来计算MCF-7细胞中雌激素受体ERα的水平。每个细胞的平均荧光发射强度与该细胞的ERα受体水平正相关。得到的原始荧光测试数据借助于Origin或是Genedata等数据处理软件得到拟合曲线。我们用半抑制浓度IC50来表征测试化合物的对雌激素受体ERα的下调作用,这个数值是指荧光发射强度降低为平均最大荧光强度的50%时测试化合物的浓度,测定的IC50如表3所示。
表3雌激素受体ERα配位分析测试结果和ERα下调降解试验测试结果
Figure PCTCN2016101768-appb-000022
备注:IC50范围为:0.1nM≤A<10nM,10nM≤B<250nM,250nM≤C≤1000nM;
结论:本发明化合物与雌激素受体能够较好的配位,且对于ERα具有较好的下调作用。
测试例3本发明化合物对MCF-7细胞抑制IC50值的测定
1.试剂和耗材
细胞计数试剂盒8(Cell Counting Kit-8,Cat#CK04-13,Dojindo)
96孔培养板(Cat#3599,Corning Costar)
培养基和胎牛血清(GIBCO)
台式酶标仪(SpectraMax M5Microplate Reader,Molecular Devices)
MCF-7人乳腺癌细胞株(购买于中科院上海细胞资源中心)
2.试剂配制
培养基的配制:MEM+10%FBS+0.01mg/ml人重组胰岛素(human recombinant insulin)
化合物的制备:用DMSO稀释化合物使终浓度为10mM;
3.实验步骤
(1)收集对数生长期细胞,计数,用完全培养基重新悬浮细胞,调整细胞浓度至合适浓度(依照细胞密度优化试验结果确定),接种96孔板,每孔加100μL细胞悬液。细胞在 37℃,100%相对湿度,5%CO2培养箱中孵育24小时;
(2)用培养基将待测化合物稀释至所设置的相应作用浓度,按25μL/孔加入细胞。化合物的作用终浓度从1μM开始,4倍梯度稀释,9个浓度点;
(3)细胞置于37℃,100%相对湿度,5%CO2培养箱中孵育72小时;
(4)吸弃培养基,加入含10%CCK-8的完全培养基置于37℃培养箱中孵育1~5小时;
(5)轻轻震荡后在SpectraMax M5Microplate Reader上测定450nm波长处的吸光度,以650nm处吸光度作为参比,计算抑制率。
4.数据处理
按下式计算药物对肿瘤细胞生长的抑制率:肿瘤细胞生长抑制率%=[(Ac-As)/(Ac-Ab)]×100%
As:样品的OA(细胞+CCK-8+待测化合物)
Ac:阴性对照的OA(细胞+CCK-8+DMSO)
Ab:阳性对照的OA(培养基+CCK-8+DMSO)
运用软件Graphpad Prism 5并采用计算公式log(inhibitor)vs.response-variable slope进行IC50曲线拟合并计算出本发明优选化合物对MCF-7细胞抑制IC50值。
表4:本发明化合物对MCF-7细胞抑制IC50值。
实施例编号 IC50(nM)
1 0.34
12 1.45
结论:本发明化合物对MCF-7细胞具有明显的抑制作用。
测试例4本发明化合物对MCF-7荷瘤SCID鼠移植瘤的生长抑制作用的测试
1.实验目的
本测试用来评价连续21天每天一次口服给予受试物对MCF-7荷瘤SCID鼠移植瘤的生长抑制作用
2.受试物配制
溶剂:20%PEG400,80%去离子水;
受试物的配制:称取适量受试物,溶解于PEG400(20%),溶解后加入80%量灭菌去离子水,震荡均匀。受试物每天于给药前新鲜配制。
3.实验动物
品种和品系:SCID鼠,SPF,雌性,7~9周龄(16~22克),100只,购买于北京华阜康生物科技股份有限公司,健康状况良好的100只用于实验,适应环境时间5~7天。
4.MCF-7肿瘤细胞培养
MCF-7细胞培养于含10%胎牛血清的RPMI1640培养基中。培养在37℃,5%CO2培养箱内。接种前取对数生长期细胞,以0.25%胰蛋白酶消化后PBS洗涤,用不含血清的培养基重新悬浮细胞计数,调整细胞浓度至7.5×10^7cells/mL(1:1Matrigel,Extracellular Matrix Proteins,356234,BD)。
5.动物接种及分组
每个小鼠在无菌状态下,右侧腋下皮下接种0.2mL细胞悬液(1.5×10^7cells/mouse)。接种后皮下给予雌激素。待肿瘤长至体积150~250mm3左右时,选出肿瘤体积相近、形状较好的小鼠(形状尽量为单一圆球形,无不规则的形状或多个肿瘤聚在一起)每组10只。
6.动物给药和观察
各组动物按下表每天固定时间根据动物体重给予受试物1天1次(qd),口服给药(po),连续21天,并记录每天动物体重。
观察各组动物接种部位肿瘤的形成状况,每周2次用游标卡尺测量肿瘤结节的长径(Y)和短径(X),并按如下公式计算:
肿瘤结节的体积(V):V=(X2Y)/2。
抗肿瘤活性的评价指标:肿瘤生长抑制率TGI(%),相对肿瘤增殖率T/C(%)。
肿瘤生长抑制率TGI(%):TGI(%)=(Vc-Vt)/Vc×100。其中Vc为模型对照组肿瘤体积,Vt为受试物组肿瘤体积。
相对肿瘤体积(relative tumor volume,RTV):RTV=Vn/V0。其中V0为分组给药时的肿瘤体积,Vn为测量时的肿瘤体积。
相对肿瘤增殖率T/C(%):T/C(%)=TRTV/CRTV×100。其中TRTV为治疗组RTV,CRTV为阴性对照组RTV。
7.结果
表5 本发明化合物对乳腺癌MCF-7荷瘤小鼠肿瘤生长抑制率(TGI%)
Figure PCTCN2016101768-appb-000023
由表5、图1、图2、图3和图4可知,在30mg/kg和50mg/kg剂量下,本发明实施例1和实施例12化合物在21天内对基于乳腺癌MCF-7细胞建立小鼠体内肿瘤模型具有明显的生长抑制作用。

Claims (21)

  1. 一种通式(I)所示的化合物或其立体异构体、互变异构体或其可药用的盐:
    Figure PCTCN2016101768-appb-100001
    其中:
    R1和R2各自独立地选自氢原子或卤素;
    R3选自如下基团:
    (i)环烷基、杂环基、芳基或杂芳基,其中所述的环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代;
    (ii)烷基,其中所述的烷基进一步被一个或多个选自羟基、硝基、氰基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中所述的烷氧基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自卤素、羟基、硝基、氰基、烷氧基、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代;
    每个R4各自独立地选自氢原子、卤素、烷基、烷氧基、三氟甲基、氰基、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6,其中所述的烷基或烷氧基任选进一步被一个或多个选自卤素、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代;
    R5选自氢原子或烷基;
    R6选自氢原子、烷基、环烷基、芳基或杂芳基,其中所述的烷基、环烷基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR8R9、-C(O)NR8R9、-C(O)R10、-SO2R10、-C(O)OR10或-NR8C(O)R9的取代基所取代;
    或者,R5和R6与连接它们的原子一起形成4~8元杂环基,其中所述的杂环基任选进一步被一个或多个选自烷基、卤素、羟基、氰基、硝基、环烷基、杂环基、芳基、杂芳基、-NR8R9、-C(O)NR8R9、-C(O)R10、-SO2R10、-C(O)OR10或-NR8C(O)R9的取代基所取代;
    R7选自氢原子、烷基、环烷基、杂环基、芳基或杂芳基,其中所述烷基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR8R9、-C(O)NR8R9、-C(O)R10、-SO2R10、-C(O)OR10或-NR8C(O)R9的取代基所取代;
    R8、R9和R10各自独立地选自氢原子、烷基、环烷基、杂环基、芳基或杂芳基,其中所述烷基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、羧酸或羧酸酯的取代基所取代;且
    n为0、1、2、3或4。
  2. 根据权利要求1所述的化合物或其立体异构体、互变异构体或其可药用的盐,其为通式(II)所述的化合物或其立体异构体、互变异构体或其可药用的盐:
    Figure PCTCN2016101768-appb-100002
    其中:R1、R2、R3、R4和n的定义如权利要求1中所述。
  3. 根据权利要求1或2所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中:
    所述的烷基为C1-C10烷基;
    所述的烷氧基为C1-C10烷氧基;
    所述的环烷基为C3-C12环烷基;
    所述的杂环基为C3-C10杂环基;
    所述的芳基为C6-C10芳基;
    所述的杂芳基为5元-10元杂芳基。
  4. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中R1和R2为F。
  5. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中每个R4各自独立地选自氢原子、C1-C3烷基、卤素、烷氧基、三氟甲基或氰基。
  6. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中R3为环丙基,其中所述的环丙基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中优选被卤素取代,更优选被F取代;且
    R5、R6、R7的定义如权利要求1中所述。
  7. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中R3选自环戊基或环己基,其中所述的环戊基或环己基任选进一步被一个或多个选自羟基、卤素、卤代烷基、硝基、氰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中优选被卤素取代,更优选被F取代;且
    R5、R6、R7的定义如权利要求1中所述。
  8. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中R3为烷基,其中所述的烷基进一步被一个或多个选自羟基、硝基、氰基、烷氧基、环烷基、杂环基、芳基、杂芳基、-NR5R6、-C(O)NR5R6、-C(O)R7、-SO2R7、-C(O)OR7或-NR5C(O)R6的取代基所取代,其中所述的烷氧基、环烷基、杂环基、芳基或杂芳基任选进一步被一个或多个F所取代;且
    R5、R6、R7的定义如权利要求1中所述。
  9. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用 的盐,其中:
    R1和R2各自独立地选自卤素,其中所述卤素优选为F;
    R3选自如下基团:
    (i)环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基,其中所述的环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基任选进一步被一个或多个卤素所取代,其中所述卤素优选为F;
    (ii)C1-C10烷基,其中所述的烷基进一步被一个或多个羟基所取代;
    R4为氢原子。
  10. 根据权利要求1~3任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中:
    R1和R2各自独立地选自卤素,优选为F;
    R3选自环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基,其中所述的环丙基、环戊基、环己基、C3-C10杂环基、C6-C10芳基或5元-10元杂芳基任选进一步被一个或多个卤素所取代,其中所述的卤素优选为F、Cl或Br,更优选为F;
    每个R4各自独立地选自C1-C6烷基或卤素,其中所述的卤素优选为F、Cl或Br,更优选为F。
  11. 根据权利要求10所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中:
    R1和R2各自独立地选自卤素,优选为F;
    R3选自环丙基,其中所述的环丙基进一步被一个或多个卤素所取代,所述卤素优选为F;
    每个R4各自独立地选自C1-C6烷基或卤素,优选为F。
  12. 根据权利要求10所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中:
    R1和R2各自独立地选自卤素,优选为F;
    R3为:
    Figure PCTCN2016101768-appb-100003
    R4为F。
  13. 根据权利要求1~12任一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,其中所述的化合物选自:
    Figure PCTCN2016101768-appb-100004
    Figure PCTCN2016101768-appb-100005
  14. 根据权利要求1所述的通式(I)化合物的制备方法,所述方法包括:
    Figure PCTCN2016101768-appb-100006
    通式(IA)和通式(IB)在酸性条件下反应,进一步酯水解,得到通式(I)化合物;
    其中:Ra为烷基;
    R1、R2、R3、R4和n的定义如权利要求1中所述。
  15. 根据权利要求2所述的通式(II)化合物的制备方法,所述方法包括:
    Figure PCTCN2016101768-appb-100007
    通式(IIA)和通式(IB)在酸性条件下反应,进一步酯水解,得到通式(II)化合物;
    其中:Ra为烷基;
    R1、R2、R3、R4和n的定义如权利要求1中所述。
  16. 一种药物组合物,所述的药物组合物含有有效剂量的根据权利要求1~13任何一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,及可药用的载体、赋形剂或它们的组合。
  17. 根据权利要求16所述的药物组合物,该药物组合物进一步包括抗氧化剂或金属螯合剂。
  18. 一种选择性下调雌激素受体的方法,该方法包括将利要求1~13任何一项所述的化 合物或其立体异构体、互变异构体或其可药用的盐,或根据权利要求16~17任何一项所述的药物组合物与雌激素受体相接触,优选为雌激素受体α。
  19. 根据权利要求1~13任何一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,或根据权利要求16~17任何一项所述的药物组合物在制备用于治疗雌激素受体介导的疾病的药物中的用途,其中所述的疾病为癌症,其中所述的癌症优选为乳腺癌或妇科癌症,其中所述的妇科癌症优选为卵巢癌或子宫内膜癌,其中所述的雌激素受体优选为雌激素受体α。
  20. 根据权利要求1~13中任何一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,或根据权利要求16~17任何一项所述的药物组合物在制备选择性雌激素受体下调剂中的用途,其中所述的选择性雌激素受体下调剂优选为雌激素受体α下调剂。
  21. 根据权利要求1~13任何一项所述的化合物或其立体异构体、互变异构体或其可药用的盐,或根据权利要求16~17任何一项所述的药物组合物,与一种或多种其他抗肿瘤药物联合用于制备治疗雌激素受体介导的疾病的药物中的用途,其中所述的疾病为癌症,其中所述的癌症优选为乳腺癌或妇科癌症,其中所述的妇科癌症优选为卵巢癌或子宫内膜癌,其中所述的雌激素受体优选为雌激素受体α,其中所述其他抗肿瘤药物包括烷化剂、抗代谢类药物、具有抗肿瘤活性的天然产物及其衍生物、细胞毒素类药物或阻滞免疫细胞迁移类药物。
PCT/CN2016/101768 2015-11-12 2016-10-11 丙烯酸类衍生物、其制备方法及其在医药上的用途 WO2017080338A1 (zh)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP16863515.9A EP3378861B1 (en) 2015-11-12 2016-10-11 Acrylic acid derivative, preparation method and use in medicine thereof
CN201680020252.8A CN107428758B (zh) 2015-11-12 2016-10-11 丙烯酸类衍生物、其制备方法及其在医药上的用途
US15/766,692 US10519148B2 (en) 2015-11-12 2016-10-11 Acrylic acid derivative, preparation method and use in medicine thereof
CA3011391A CA3011391C (en) 2015-11-12 2016-10-11 Acrylic acid derivative, preparation method and use in medicine thereof
JP2018524291A JP6592197B2 (ja) 2015-11-12 2016-10-11 アクリル酸誘導体、製造方法、および医薬としてのその使用
KR1020187014684A KR102099159B1 (ko) 2015-11-12 2016-10-11 아크릴산 유도체, 그의 제조방법 및 의약에 있어서의 그의 사용방법

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510771111 2015-11-12
CN201510771111.7 2015-11-12

Publications (1)

Publication Number Publication Date
WO2017080338A1 true WO2017080338A1 (zh) 2017-05-18

Family

ID=58694739

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/101768 WO2017080338A1 (zh) 2015-11-12 2016-10-11 丙烯酸类衍生物、其制备方法及其在医药上的用途

Country Status (8)

Country Link
US (1) US10519148B2 (zh)
EP (1) EP3378861B1 (zh)
JP (1) JP6592197B2 (zh)
KR (1) KR102099159B1 (zh)
CN (1) CN107428758B (zh)
CA (1) CA3011391C (zh)
TW (1) TWI615393B (zh)
WO (1) WO2017080338A1 (zh)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017136688A1 (en) 2016-02-05 2017-08-10 Inventisbio Inc. Selective estrogen receptor degraders and uses thereof
WO2018001232A1 (zh) * 2016-06-29 2018-01-04 浙江海正药业股份有限公司 丙烯酸类衍生物及其制备方法和其在医药上的用途
CN107814798A (zh) * 2016-09-14 2018-03-20 四川科伦博泰生物医药股份有限公司 3‑取代丙烯酸类化合物及其制备方法和用途
US10118910B2 (en) 2015-12-09 2018-11-06 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10125135B2 (en) 2016-04-20 2018-11-13 Astrazeneca Ab Chemical compounds
US10131663B2 (en) 2016-10-24 2018-11-20 Astrazeneca Ab Chemical compounds
US10149839B2 (en) 2016-07-25 2018-12-11 Astrazeneca Ab Chemical compounds
US10221173B2 (en) 2017-01-30 2019-03-05 Astrazeneca Ab Chemical compounds
CN110746340A (zh) * 2019-11-07 2020-02-04 上海易恩化学技术有限公司 一种5-甲氧基-2-甲基色胺的合成方法
US10703747B2 (en) 2016-10-24 2020-07-07 The Board of Directors of the University of Illinois Benzothiophene-based selective mixed estrogen receptor downregulators
WO2020238733A1 (zh) 2019-05-24 2020-12-03 浙江海正药业股份有限公司 丙烯酸类衍生物的晶型及其制备方法和用途
US10959989B2 (en) 2016-04-01 2021-03-30 Recurium Ip Holdings, Llc Estrogen receptor modulators
US11040964B2 (en) 2019-02-27 2021-06-22 Ferro Therapeutics, Inc. Compounds and methods of use
US11098040B2 (en) 2018-02-28 2021-08-24 Ferro Therapeutics, Inc. Compounds and methods of use
US11278532B2 (en) 2019-08-06 2022-03-22 Recurium Ip Holdings, Llc Estrogen receptor modulators for treating mutants
CN116574054A (zh) * 2023-05-15 2023-08-11 南京中医药大学 选择性雌激素受体下调剂化合物、制备方法及用途

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112019013814A2 (pt) 2017-01-06 2020-01-21 G1 Therapeutics Inc método para tratamento de câncer ou de um tumor em um indivíduo, composição farmacêutica, combinação, e, kit.
AU2021269612A1 (en) * 2020-05-15 2023-01-05 Jiangsu Simcere Pharmaceutical Co., Ltd. Pyrrolidine compound and use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014191726A1 (en) * 2013-05-28 2014-12-04 Astrazeneca Ab Chemical compounds

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005206570A1 (en) * 2004-01-23 2005-08-04 Novartis Vaccines And Diagnostics, Inc. Tetrahydrocarboline compounds as anticancer agents
US8198292B2 (en) * 2008-07-03 2012-06-12 Osteogenex, Inc. Vinpocetine and eburnamonine derivatives for promoting bone growth, treating renal damage and cancer, and devices thereof
PL2580210T3 (pl) 2010-06-10 2017-09-29 Seragon Pharmaceuticals, Inc. Modulatory receptora estrogenowego i ich zastosowania
GB2483736B (en) 2010-09-16 2012-08-29 Aragon Pharmaceuticals Inc Estrogen receptor modulators and uses thereof
SG11201604501TA (en) 2013-12-06 2016-07-28 Hoffmann La Roche Estrogen receptor modulator for the treatment of locally advanced or metastatic estrogen receptor positive breast cancer
CN113004273A (zh) * 2016-02-05 2021-06-22 益方生物科技(上海)股份有限公司 选择性雌激素受体降解物及其用途

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014191726A1 (en) * 2013-05-28 2014-12-04 Astrazeneca Ab Chemical compounds

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11447461B2 (en) 2015-12-09 2022-09-20 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10118910B2 (en) 2015-12-09 2018-11-06 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US11072595B2 (en) 2015-12-09 2021-07-27 The Board of Trustees of lhe University of Illinois Benzothiophene-based selective estrogen receptor downregulator compounds
US10807964B2 (en) 2015-12-09 2020-10-20 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10377735B2 (en) 2015-12-09 2019-08-13 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10647724B2 (en) 2016-02-05 2020-05-12 Inventisbio Inc. Selective estrogen receptor degraders and uses thereof
KR102479822B1 (ko) 2016-02-05 2022-12-21 인벤티스바이오 엘엘씨 선택적 에스트로겐 수용체 분해제 및 이들의 용도
KR20180104161A (ko) * 2016-02-05 2018-09-19 인벤티스바이오 인크. 선택적 에스트로겐 수용체 분해제 및 이들의 용도
WO2017136688A1 (en) 2016-02-05 2017-08-10 Inventisbio Inc. Selective estrogen receptor degraders and uses thereof
US11014936B2 (en) 2016-02-05 2021-05-25 Inventisbio Llc Selective estrogen receptor degraders and uses thereof
EP3411034A4 (en) * 2016-02-05 2019-08-28 Inventisbio Inc. SELECTIVE ESTROGEN RECEPTOR DERIVATIVES AND USES THEREOF
US11065233B2 (en) 2016-04-01 2021-07-20 Recurium Ip Holdings, Llc Estrogen receptor modulators
US11065234B2 (en) 2016-04-01 2021-07-20 Recurium Ip Holdings, Llc Estrogen receptor modulators
US10959989B2 (en) 2016-04-01 2021-03-30 Recurium Ip Holdings, Llc Estrogen receptor modulators
US10125135B2 (en) 2016-04-20 2018-11-13 Astrazeneca Ab Chemical compounds
WO2018001232A1 (zh) * 2016-06-29 2018-01-04 浙江海正药业股份有限公司 丙烯酸类衍生物及其制备方法和其在医药上的用途
US10149839B2 (en) 2016-07-25 2018-12-11 Astrazeneca Ab Chemical compounds
CN107814798A (zh) * 2016-09-14 2018-03-20 四川科伦博泰生物医药股份有限公司 3‑取代丙烯酸类化合物及其制备方法和用途
CN107814798B (zh) * 2016-09-14 2020-11-03 四川科伦博泰生物医药股份有限公司 3-取代丙烯酸类化合物及其制备方法和用途
US10961241B2 (en) 2016-10-24 2021-03-30 Astrazeneca Ab Chemical compounds
US10703747B2 (en) 2016-10-24 2020-07-07 The Board of Directors of the University of Illinois Benzothiophene-based selective mixed estrogen receptor downregulators
US10131663B2 (en) 2016-10-24 2018-11-20 Astrazeneca Ab Chemical compounds
US10590130B2 (en) 2016-10-24 2020-03-17 Astrazeneca Ab Chemical compounds
US10221173B2 (en) 2017-01-30 2019-03-05 Astrazeneca Ab Chemical compounds
US10590132B2 (en) 2017-01-30 2020-03-17 Astrazeneca Ab Selective estrogen receptor down-regulators
US11046689B2 (en) 2017-01-30 2021-06-29 Astrazeneca Ab Selective estrogen receptor down-regulators
US11098040B2 (en) 2018-02-28 2021-08-24 Ferro Therapeutics, Inc. Compounds and methods of use
US11040964B2 (en) 2019-02-27 2021-06-22 Ferro Therapeutics, Inc. Compounds and methods of use
CN114929696A (zh) * 2019-05-24 2022-08-19 浙江海正药业股份有限公司 丙烯酸类衍生物的晶型及其制备方法和用途
WO2020238733A1 (zh) 2019-05-24 2020-12-03 浙江海正药业股份有限公司 丙烯酸类衍生物的晶型及其制备方法和用途
US11278532B2 (en) 2019-08-06 2022-03-22 Recurium Ip Holdings, Llc Estrogen receptor modulators for treating mutants
CN110746340A (zh) * 2019-11-07 2020-02-04 上海易恩化学技术有限公司 一种5-甲氧基-2-甲基色胺的合成方法
CN116574054A (zh) * 2023-05-15 2023-08-11 南京中医药大学 选择性雌激素受体下调剂化合物、制备方法及用途

Also Published As

Publication number Publication date
CN107428758A (zh) 2017-12-01
US20180291019A1 (en) 2018-10-11
US10519148B2 (en) 2019-12-31
EP3378861B1 (en) 2021-08-04
EP3378861A1 (en) 2018-09-26
CN107428758B (zh) 2020-08-04
KR102099159B1 (ko) 2020-04-10
EP3378861A4 (en) 2019-06-26
JP2018535216A (ja) 2018-11-29
TWI615393B (zh) 2018-02-21
KR20180080245A (ko) 2018-07-11
CA3011391A1 (en) 2017-05-18
CA3011391C (en) 2021-02-09
TW201716406A (zh) 2017-05-16
JP6592197B2 (ja) 2019-10-16

Similar Documents

Publication Publication Date Title
WO2017080338A1 (zh) 丙烯酸类衍生物、其制备方法及其在医药上的用途
TWI651321B (zh) 丙烯酸類衍生物及其製備方法和其在醫藥上的用途
JP6820254B2 (ja) 治療用化合物およびその使用
JP5492565B2 (ja) Janusキナーゼ阻害剤としての置換複素環
JP2019518767A (ja) Cxcr4阻害剤およびその使用
KR20210098960A (ko) Helios의 소분자 분해제 및 사용 방법
JP2019516757A (ja) Cbp/ep300の複素環式インヒビターおよびがんの処置におけるそれらの使用
JP2021506838A (ja) 非環式cxcr4阻害剤およびその使用
TW200906834A (en) Axl kinase inhibitors
TWI393719B (zh) 蛋白質激酶之抑制劑
JP2012524076A (ja) プロテインキナーゼck2活性に関連する障害の治療方法
WO2023099624A1 (en) Annulated 2-amino-3-cyano thiophenes and derivatives for the treatment of cancer
JP2024506858A (ja) Gpr84アンタゴニストおよびその使用
US11304929B2 (en) Tosylacetate based compounds and derivatives thereof as PHGDH inhibitors
WO2018228474A1 (zh) 聚(adp-核糖)聚合酶抑制剂、制备方法及用途
CA3069602A1 (en) Formylpyridine derivative having fgfr4 inhibitory activity, preparation method therefor and use thereof
CN110650961B (zh) Parp抑制剂、其药物组合物、制备方法和应用
CN105473590A (zh) 用于治疗增殖性障碍的1h-吡唑并[3,4-b]吡啶衍生物及其药物组合物
TWI699366B (zh) 治療或預防癌症的化合物
US11261184B2 (en) [1,6]naphthyridine compounds and derivatives as CDK8/CDK19 inhibitors
WO2022197913A1 (en) Bicyclic derivatives which can be used to treat cancer
TW201914999A (zh) 1,2-二氫-1,6-萘啶類衍生物、其製備方法、其藥物組合物及其在醫藥上的用途
WO2023173083A1 (en) Tetrahydroindole derivatives as egfr and/or her2 inhibtors useful for the treatment of cancer

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: 16863515

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15766692

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2018524291

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20187014684

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2016863515

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016863515

Country of ref document: EP

Effective date: 20180612

WWE Wipo information: entry into national phase

Ref document number: 3011391

Country of ref document: CA