WO2020253762A1 - 吲唑类衍生物、其制备方法及其在医药上的应用 - Google Patents

吲唑类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2020253762A1
WO2020253762A1 PCT/CN2020/096744 CN2020096744W WO2020253762A1 WO 2020253762 A1 WO2020253762 A1 WO 2020253762A1 CN 2020096744 W CN2020096744 W CN 2020096744W WO 2020253762 A1 WO2020253762 A1 WO 2020253762A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
general formula
racemate
tautomer
Prior art date
Application number
PCT/CN2020/096744
Other languages
English (en)
French (fr)
Inventor
樊兴
杨方龙
严晶晶
吴晓
贺峰
陶维康
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
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 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to CA3143367A priority Critical patent/CA3143367A1/en
Priority to AU2020294642A priority patent/AU2020294642A1/en
Priority to BR112021025601A priority patent/BR112021025601A2/pt
Priority to US17/618,767 priority patent/US20220267304A1/en
Priority to KR1020227000865A priority patent/KR20220024476A/ko
Priority to CN202080038970.4A priority patent/CN113874367A/zh
Priority to JP2021575257A priority patent/JP2022537330A/ja
Priority to EP20827542.0A priority patent/EP3988547A4/en
Priority to MX2021015486A priority patent/MX2021015486A/es
Publication of WO2020253762A1 publication Critical patent/WO2020253762A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • 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
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present disclosure belongs to the field of medicine, and relates to an indazole derivative, its preparation method and its application in medicine.
  • the present disclosure discloses its use as an estrogen receptor modulator to prevent and/or treat estrogen receptor-mediated Or the use of dependent diseases or disorders, said diseases are particularly preferably breast cancer.
  • Breast cancer is one of the most common malignant tumors in women. According to GLOBALCAN statistics in 2012 (CA CANCER J CLIN 2015; 65:87–108), there are approximately 1.7 million new cancer cases and 520,000 deaths worldwide each year. Both morbidity and mortality rank first among female malignant tumors. According to the 2017 China Cancer Registry Annual Report released by the National Cancer Center, breast cancer ranks first in the incidence of female malignant tumors, with about 279,000 new cases each year, and an annual increase of about 2%.
  • endocrine therapy occupies an important position.
  • aromatase inhibitors which can inhibit the conversion of androgens into estrogen and reduce the level of estrogen in the body
  • SERM selective estrogen receptor modulators
  • SERM selective estrogen receptors modulator
  • SERM selective estrogen receptor degrader
  • SELD selective estrogen receptor degrader
  • endocrine therapy is the first choice for estrogen receptor-positive breast cancer
  • about 30% of patients receiving adjuvant therapy will relapse, and almost all patients with metastatic breast cancer will develop resistance and progress.
  • the mechanisms of resistance to endocrine therapy are mainly divided into two categories. One is focused on the estrogen receptor signaling pathway itself, including the activation mutation, amplification, and fusion with other genes of the gene encoding the estrogen receptor. Other mechanisms include the activation of signal pathways that cross-react with the estrogen receptor signaling pathway, such as the growth factor receptor pathway, etc. (Nat Rev Clin Oncol. 2015 Oct ; 12(10):573-83).
  • ESR1 gene mutations were detected in 11-55% of estrogen receptor-positive metastatic breast cancer patients who had received aromatase inhibitor therapy. Further research found that the mutant receptor can be independent of estrogen. Phosphorylation, play a transcriptional role, so that estrogen-dependent MCF7-inoculated tumors can no longer rely on estrogen to grow in the body, and the mutant receptor will make SERM tamoxifen (tamoxifen) and SERD fulvestrant (fulvestrant) Reduced activity. Therefore, mutations in the ESR1 gene may be one of the mechanisms of drug resistance in estrogen-positive breast cancer (Nat Rev Clin Oncol. 2015 Oct; 12(10): 573-83 and Nat Genet 2013; 45:1439–45).
  • ESR1 gene mutations mostly occur in patients with metastatic estrogen receptor-positive breast cancer who have progressed after AIs treatment. These patients are no longer sensitive to AIs treatment. Therefore, it is necessary to develop an estrogen receptor targeting ESR1 gene mutations. Antagonist.
  • the first-in-class estrogen receptor covalent binding antagonist H3B-6545 developed by Eisai Company has strong inhibitory activity on wild-type and mutant estrogen receptors, and can be covalently bound to the receptor To exert the effect for a longer time, the first and second clinical trials are currently underway.
  • Currently published patents for estrogen receptor antagonists targeting ESR1 gene mutations are WO2016196346 and WO2016196342.
  • the purpose of the present disclosure is to provide a compound represented by the general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof Form, or its pharmaceutically acceptable salt:
  • G is CH or N
  • Z is selected from bond, CR 5 R 6 , -O-(CH 2 )t- or -NR 7 -(CH 2 )t-;
  • Ring A is selected from cycloalkyl or heterocyclic group
  • R b is selected from a hydrogen atom or an alkyl group
  • R 1 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclic, aryl and heteroaryl;
  • R 2 is selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, amino, cyano, nitro, carboxy, aldehyde, hydroxy, hydroxyalkyl, cycloalkyl, aryl and heteroaryl;
  • R 3 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, cyano, amino, nitro, carboxy, aldehyde, hydroxyl, hydroxyalkyl, NR 13 C (O) R 14 , C(O)NR 13 R 14 , SO 2 R 15 , cycloalkyl, heterocyclic, aryl and heteroaryl;
  • R 4 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, cyano, amino, nitro, carboxy, aldehyde, hydroxyl, hydroxyalkyl, cycloalkyl , Heterocyclyl, aryl and heteroaryl;
  • R 5 and R 6 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, cyano, amino, nitro, carboxy, aldehyde, hydroxyl, hydroxyalkyl, ring Alkyl, heterocyclyl, aryl and heteroaryl;
  • R 7 is selected from hydrogen atom, alkyl group, haloalkyl group, hydroxyalkyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group;
  • R 8 and R 9 are the same or different, and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 8 and R 9 together with the nitrogen atom to which they are connected form a heterocyclic group, wherein the heterocyclic group optionally contains 1 or 2 identical or different nitrogen atoms selected from N, O and S heteroatoms, and the heterocyclic group is optionally selected from alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, One or more substituents in the aryl and heteroaryl groups;
  • R 10 and R 11 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, cyano, amino, nitro, carboxy, aldehyde, hydroxyl, hydroxyalkyl, ring Alkyl, heterocyclyl, aryl and heteroaryl;
  • R 12 is selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 13 and R 14 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, cyano, amino, nitro, carboxy, aldehyde, hydroxyl, hydroxyalkyl, ring Alkyl, heterocyclyl, aryl and heteroaryl;
  • R 15 is selected from hydrogen atom, alkyl group, haloalkyl group, hydroxyalkyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group;
  • n 0 or 1
  • n 0, 1, 2 or 3;
  • q 0, 1, 2, 3, 4 or 5;
  • s 0, 1, 2, or 3 and
  • t 0, 1, 2, 3, 4, 5, or 6.
  • ring A is selected from C 3 -C 6 cycloalkyl, 3 to 6 membered heterocyclic group; said heterocyclic group contains 1 to 3 selected from N, O Or S heteroatom.
  • the compound represented by the general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer, Or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound represented by the general formula (II) or a tautomer, a mesoisomer, a racemate, an enantiomer, or a non-pair Enantiomers or their mixture forms, or their pharmaceutically acceptable salts:
  • the compound represented by general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer Or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound represented by the general formula (III) or a tautomer, meso, racemate, enantiomer, or non-pair Enantiomers or their mixture forms, or their pharmaceutically acceptable salts:
  • the compound represented by general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer Or its mixture form, or its pharmaceutically acceptable salt which is a compound represented by the general formula (IV) or its tautomer, meso, racemate, enantiomer, or non-pair Enantiomers or their mixture forms, or their pharmaceutically acceptable salts:
  • G, Z, R 1 , R 2 , R 3 , R 4 , R 8 , R 9 , R b , n, s and q are as defined in general formula (I);
  • R 8 and R 9 form a heterocyclic group together with the nitrogen atom to which they are connected, wherein the heterocyclic group optionally contains 1 to 2 identical or different N atoms in addition to one nitrogen atom.
  • the heterocyclic group is optionally selected from alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl , Aryl and heteroaryl groups are substituted by one or more substituents; more preferably, the heterocyclic group is optionally substituted by hydroxy or hydroxyalkyl; further preferably, the heterocyclic group is optionally Replaced by hydroxyl.
  • the compound represented by general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 3 is selected from hydrogen atom, halogen, alkyl, cyano, haloalkyl, NR 13 C(O)R 14 , C(O)NR 13 R 14 and SO 2 R 15 ; R 13 and R 14 are the same or different, and are each independently selected from a hydrogen atom and an alkyl group; R 15 is selected from a hydrogen atom and an alkyl group.
  • the compound represented by general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 4 is the same or different, and each is independently selected from a hydrogen atom, a halogen, an alkyl group, and an alkoxy group, preferably a hydrogen atom.
  • the heterocyclic group optionally contains 1 to 2 identical or different heteroatoms selected from N, O and S, and the heterocyclic group is optionally selected from Alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloal
  • Typical compounds of general formula (I) include but are not limited to:
  • Another aspect of the present disclosure provides a compound represented by general formula (IA) or its tautomer, meso, racemate, enantiomer, diastereomer or In the form of a mixture, or a pharmaceutically acceptable salt thereof,
  • R w is an amino protecting group, preferably tetrahydropyranyl
  • R 8 and R 9 are the same or different, and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 8 and R 9 together with the nitrogen atom to which they are connected form a heterocyclic group, wherein the heterocyclic group optionally contains 1 or 2 identical or different nitrogen atoms selected from N, O and S heteroatoms, and the heterocyclic group is optionally selected from alkyl, alkoxy, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, -COOR 16 -, cycloalkyl , Substituted by one or more substituents in heterocyclyl, aryl and heteroaryl;
  • R 10 and R 11 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl, haloalkyl, alkoxy, cyano, amino, nitro, carboxy, aldehyde, hydroxyl, hydroxyalkyl, ring Alkyl, heterocyclyl, aryl and heteroaryl;
  • R 12 is selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 16 is selected from hydrogen atom, alkyl group, haloalkyl group and hydroxyalkyl group;
  • Rings A, G, Z, R 1 , R 2 , R 3 , R 4 , R b , n, m, s, and q are as defined in the general formula (I).
  • Another aspect of the present disclosure provides a compound represented by the general formula (IIA) or its tautomer, meso, racemate, enantiomer, diastereomer or In the form of a mixture, or a pharmaceutically acceptable salt thereof,
  • R w is an amino protecting group, preferably tetrahydropyranyl
  • R is as defined in the general formula (IA);
  • Rings A, G, Z, R 1 , R 2 , R 3 , R 4 , R b , n, m, s, and q are as defined in the general formula (II).
  • Another aspect of the present disclosure provides a compound represented by general formula (IIIA) or its tautomer, meso, racemate, enantiomer, diastereomer or In the form of a mixture, or a pharmaceutically acceptable salt thereof,
  • R w is an amino protecting group, preferably tetrahydropyranyl
  • R is as defined in the general formula (IA);
  • G, Z, R 1 , R 2 , R 3 , R 4 , R b , n, s, and q are as defined in the general formula (III).
  • Another aspect of the present disclosure provides a compound represented by the general formula (IVA) or its tautomer, meso, racemate, enantiomer, diastereomer or In the form of a mixture, or a pharmaceutically acceptable salt thereof,
  • R w is an amino protecting group, preferably tetrahydropyranyl
  • G, Z, R 1 , R 2 , R 3 , R 4 , R b , R 8 , R 9 , n, s, and q are as defined in the general formula (IV).
  • Typical compounds of general formula (IA) include but are not limited to:
  • Another aspect of the present disclosure provides a method for preparing a compound represented by the general formula (I) or its tautomer, meso, racemate, enantiomer, and diastereomer Or a method in the form of a mixture thereof, or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound or its tautomer, mesosome, racemate, enantiomer, diastereomer or its mixture form, or its pharmaceutically acceptable salt is deprotected under acidic conditions Reaction to obtain the compound of general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture form thereof, or its pharmaceutically acceptable salt;
  • R w is an amino protecting group, preferably tetrahydropyranyl or tert-butoxycarbonyl;
  • R is as defined in the general formula (IA);
  • Another aspect of the present disclosure provides a method for preparing a compound represented by general formula (II) or its tautomer, meso, racemate, enantiomer, and diastereomer Or a method in the form of a mixture thereof, or a pharmaceutically acceptable salt thereof, the method comprising:
  • R w is an amino protecting group, preferably tetrahydropyranyl or tert-butoxycarbonyl;
  • R is as defined in general formula (IIA);
  • Ring A, G, Z, R 1 , R 2, R 3, R 4, R a, R b, n, m, s and q are as formula (II) as defined above.
  • Another aspect of the present disclosure provides a method for preparing a compound represented by general formula (III) or its tautomer, meso, racemate, enantiomer, and diastereomer Or a method in the form of a mixture thereof, or a pharmaceutically acceptable salt thereof, the method comprising:
  • R w is an amino protecting group, preferably tetrahydropyranyl or tert-butoxycarbonyl;
  • R is as defined in general formula (IIIA);
  • G, Z, R 1, R 2, R 3, R 4, R a, R b, n, s and q are as formula (III) as defined above.
  • Another aspect of the present disclosure provides a method for preparing a compound represented by the general formula (IV) or its tautomer, meso, racemate, enantiomer, and diastereomer Or a method in the form of a mixture thereof, or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound of general formula (IVA) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture form thereof, or its pharmaceutically acceptable salt is acidic Deprotection reaction occurs under conditions to obtain the compound of general formula (IV) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or Its medicinal salt;
  • R w is an amino protecting group, preferably tetrahydropyranyl or tert-butoxycarbonyl;
  • G, Z, R 1 , R 2 , R 3 , R 4 , R 8 , R 9 , R b , n, s and q are as defined in the general formula (IV).
  • compositions which contains a therapeutically effective dose of the compounds represented by the above general formulas (I) to (IV) or their tautomers, mesosomes, racemics Isomers, enantiomers, diastereomers or mixtures thereof, or pharmaceutically acceptable salts, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present disclosure also relates to a method for preparing the above-mentioned pharmaceutical composition, which comprises combining the compounds represented by the general formulas (I) to (IV) or their tautomers, mesosomes, racemates, and The enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof are mixed with a pharmaceutically acceptable carrier, diluent or excipient.
  • the present disclosure further relates to compounds represented by general formulas (I) to (IV) or tautomers, mesoisomers, racemates, enantiomers, diastereomers or mixtures thereof Use of the form, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing the same in the preparation of an estrogen receptor modulator.
  • the present disclosure further relates to compounds represented by general formulas (I) to (IV) or tautomers, mesoisomers, racemates, enantiomers, diastereomers or mixtures thereof Form, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing the same in the preparation of a medicament for the prevention and/or treatment of estrogen receptor-mediated or dependent diseases or disorders; wherein the estrogen receptor mediates
  • the leading or dependent disease or condition is preferably cancer, more preferably breast cancer, ovarian cancer, endometrial cancer, prostate cancer or uterine cancer; further preferably breast cancer.
  • the present disclosure further relates to a compound represented by formula (I) to (IV) or its tautomer, meso, racemate, enantiomer, diastereomer Isomers or mixtures thereof, or pharmaceutically acceptable salts thereof.
  • the present disclosure further relates to a compound represented by the general formula (I) to (IV) of the present disclosure or its tautomer, meso, racemate, enantiomer, diastereomer
  • the construct or its mixture form, or its pharmaceutically acceptable salt, is used as a medicine for treating estrogen receptor-mediated or dependent diseases or disorders; wherein the estrogen receptor-mediated or dependent diseases or
  • the disorder is preferably cancer, more preferably breast cancer, ovarian cancer, endometrial cancer, prostate cancer or uterine cancer; further preferably breast cancer.
  • the present disclosure further relates to a method of treating estrogen receptor-mediated or dependent diseases or conditions, the method comprising administering to a patient in need thereof a therapeutically effective dose of the general formulas (I) to (IV) of the present disclosure
  • This method shows outstanding efficacy and few side effects.
  • the estrogen receptor-mediated or dependent disease or condition is preferably cancer, more preferably breast cancer, ovarian cancer, endometrial cancer, prostate cancer or uterine cancer; further preferably breast cancer.
  • the active compound can be prepared in a form suitable for administration by any appropriate route, and the active compound is preferably in a unit dose form, or a form in which the patient can self-administer in a single dose.
  • the expression of the unit dose of the compound or composition of the present invention can be tablet, capsule, cachet, bottled syrup, powder, granule, lozenge, suppository, regenerating powder or liquid preparation.
  • the dosage of the compound or composition used in the treatment method of the present invention will generally vary with the severity of the disease, the weight of the patient, and the relative efficacy of the compound.
  • a suitable unit dose can be 0.1-1000 mg.
  • the pharmaceutical composition of the present invention may contain one or more auxiliary materials selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients Wait.
  • auxiliary materials selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients Wait.
  • the composition may contain 0.1 to 99% by weight of the active compound.
  • the pharmaceutical composition containing the active ingredient may be in a form suitable for oral administration, such as tablets, dragees, lozenges, water or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or Elixirs.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions. Such compositions can contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents and preservatives, In order to provide pleasing and delicious medicinal preparations.
  • the tablet contains the active ingredient and non-toxic pharmaceutically acceptable excipients suitable for the preparation of tablets for mixing.
  • excipients can be inert excipients; granulating and disintegrating agents; and lubricants.
  • These tablets may be uncoated or may be coated by known techniques that mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract, thereby providing a sustained release effect over a longer period of time.
  • water-soluble taste masking substances can be used.
  • Oral preparations can also be provided by soft gelatin capsules in which the active ingredient is mixed with an inert solid diluent, or the active ingredient is mixed with a water-soluble carrier.
  • Aqueous suspensions contain the active substance and excipients suitable for the preparation of aqueous suspensions for mixing. Such excipients are suspending agents, dispersing agents or wetting agents.
  • the active ingredient of the aqueous suspension may be a dispersible powder or granules. By adding water, the active ingredient is mixed with one or more dispersing agents, wetting agents or suspending agents.
  • the aqueous suspension may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
  • Oil suspensions can be formulated by suspending the active ingredients in vegetable oil or mineral oil.
  • the oil suspension may contain thickening agents.
  • the above-mentioned sweeteners and flavoring agents can be added to provide a palatable preparation. These compositions can be preserved by adding antioxidants.
  • dispersible powders and granules suitable for preparing water suspensions can be provided with active ingredients and dispersing or wetting agents for mixing, suspending agents or one or more preservatives. Suitable dispersing or wetting agents and suspending agents can illustrate the above examples. Other excipients such as sweetening agents, flavoring agents and coloring agents may also be added. These compositions are preserved by adding antioxidants such as ascorbic acid.
  • the pharmaceutical composition of the present disclosure may also be in the form of an oil-in-water emulsion.
  • the oil phase can be vegetable oil or mineral oil or a mixture thereof.
  • Suitable emulsifiers can be naturally occurring phospholipids.
  • Sweeteners can be used.
  • Such preparations may also contain a demulcent, a preservative, a coloring agent and an antioxidant.
  • the pharmaceutical composition of the present disclosure may be in the form of a sterile injectable aqueous solution.
  • Acceptable solvents or solvents that can be used are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injection preparation may be a sterile oil-in-water injection microemulsion in which the active ingredient is dissolved in an oil phase.
  • the injection or microemulsion can be injected into the patient's bloodstream by local large injections.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM. 5400 intravenous pump.
  • the pharmaceutical composition of the present disclosure may be in the form of a sterile injection water or oil suspension for intramuscular and subcutaneous administration.
  • the suspension can be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents mentioned above.
  • the sterile injection preparation may also be a sterile injection solution or suspension prepared in a parenterally acceptable non-toxic diluent or solvent.
  • sterile fixed oil can be conveniently used as a solvent or suspension medium.
  • fatty acids can also be used to prepare injections.
  • the compounds of the present disclosure can be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid in the rectum and thus will melt in the rectum to release the drug.
  • the dosage of the drug depends on many factors, including but not limited to the following factors: the activity of the specific compound used, the age of the patient, the weight of the patient, the health of the patient, and the behavior of the patient , The patient’s diet, time of administration, mode of administration, rate of excretion, combination of drugs, etc.; in addition, the best mode of treatment such as the mode of treatment, the daily dosage of compound (I) or the amount of pharmaceutically acceptable salt
  • the type can be verified according to the traditional treatment plan.
  • alkyl refers to a saturated aliphatic hydrocarbon group, which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 12 carbon atoms, more preferably containing 1 to 6 carbons Atom of the alkyl group.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 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-dimethylbutyl, 1,3 -Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -Methylhexyl, 3-methylhexyl, 4-methylhe
  • a lower alkyl group containing 1 to 6 carbon atoms non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Group, n-pentyl, 1,1-dimethylpropyl, 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-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Group, 2,3-dimethylbutyl, etc.
  • Alkyl groups may be substituted or unsubstituted. When substituted, the substituents may be substituted at any available attachment point.
  • the substituents are preferably one or more of the following groups, which are independently selected from alkanes Group, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkane Oxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxy, or carboxylate.
  • alkylene refers to a saturated linear or branched aliphatic hydrocarbon group, which has 2 residues derived from the removal of two hydrogen atoms from the same carbon atom or two different carbon atoms of the parent alkane, which is A straight or branched chain group containing 1 to 20 carbon atoms, preferably containing 1 to 12 carbon atoms, more preferably an alkylene group containing 1 to 6 carbon atoms.
  • Non-limiting examples of alkylene groups include, but are not limited to, methylene (-CH 2 -), 1,1-ethylene (-CH(CH 3 )-), 1,2-ethylene (-CH 2 -) CH 2 )-, 1,1-propylene (-CH(CH 2 CH 3 )-), 1,2-propylene (-CH 2 CH(CH 3 )-), 1,3-propylene (-CH 2 CH 2 CH 2 -), 1,4-butylene (-CH 2 CH 2 CH 2 CH 2 -), etc.
  • the alkylene group may be substituted or unsubstituted. When substituted, the substituent may be substituted at any available point of attachment.
  • the substituent is preferably independently optionally selected from alkyl, alkenyl, alkynyl , Alkoxy, alkylthio, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy Substituted by one or more substituents in the group, cycloalkylthio, heterocycloalkylthio and oxo.
  • alkenyl refers to an alkyl compound containing a carbon-carbon double bond in the molecule, wherein the definition of the alkyl group is as described above.
  • the alkenyl group may be substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from hydrogen atoms, alkyl groups, alkoxy groups, halogens, halogenated alkyl groups, hydroxyl groups, One or more substituents of hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclic, aryl and heteroaryl are substituted.
  • alkynyl refers to an alkyl compound containing a carbon-carbon triple bond in the molecule, wherein the definition of the alkyl group is as described above.
  • the alkynyl group may be substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from hydrogen atoms, alkyl groups, alkoxy groups, halogens, halogenated alkyl groups, hydroxyl groups, It is substituted by one or more substituents among hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclic, aryl and heteroaryl.
  • alkoxy refers to -O- (alkyl) and -O- (unsubstituted cycloalkyl), where alkyl and cycloalkyl are as defined herein.
  • alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthio, carboxyl or carboxylate.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent.
  • the cycloalkyl ring contains 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms (e.g., 3, 4, 5). , 6, 7, 8, 9, 10, 11, 12), more preferably 3 to 6 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene Groups, cyclooctyl, etc.; polycyclic cycloalkyls include spiro, fused, and bridged cycloalkyls.
  • spirocycloalkyl refers to a polycyclic group that shares one carbon atom (called a spiro atom) between 5- to 20-membered monocyclic rings. It may contain one or more double bonds, but none of the rings have complete conjugate ⁇ electronic system. It is preferably 6 to 14 yuan (for example, 6, 7, 8, 9, 10, 11, 12, 13, 14 yuan), and more preferably 7 to 10 yuan.
  • the spirocycloalkyl group is classified into a single spirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a single spirocycloalkyl group and a bispirocycloalkyl group. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered monospirocycloalkyl.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to a 5- to 20-membered all-carbon polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ electron system. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan (for example, 7, 8, 9 or 10 yuan).
  • bicyclic, tricyclic, tetracyclic or polycyclic condensed cycloalkyls preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicyclic alkyl.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to a 5- to 20-membered, all-carbon polycyclic group with any two rings sharing two carbon atoms that are not directly connected. It may contain one or more double bonds, but no ring has complete Conjugated ⁇ electron system. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan (for example, 7, 8, 9 or 10 yuan). According to the number of constituent rings, it can be classified into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyls, preferably bicyclic, tricyclic or tetracyclic, and more preferably bicyclic or tricyclic.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring includes the above-mentioned cycloalkyl groups (for example, spirocycloalkyl, fused cycloalkyl, and bridged cycloalkyl) fused on an aryl, heteroaryl or heterocycloalkyl ring, where it is connected to the parent structure
  • the ring together is a cycloalkyl group, non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl and the like.
  • Cycloalkyl groups may be optionally substituted or unsubstituted.
  • the substituents are preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthio, oxo, carboxy, or carboxylate.
  • groups are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthi
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, which contains 3 to 20 ring atoms, one or more of which is selected from nitrogen, oxygen, S, S( O) or S(O) 2 heteroatoms, but not including the ring part of -OO-, -OS- or -SS-, and the remaining ring atoms are carbon. It preferably contains 3 to 12 ring atoms, of which 1 to 4 are heteroatoms (for example, 1, 2, 3, and 4); more preferably, it contains 3 to 6 ring atoms (for example, 3, 4, 5, 6).
  • Non-limiting examples of monocyclic heterocyclic groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidine Group, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, etc., preferably piperidinyl and pyrrolidinyl.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclic group refers to a polycyclic heterocyclic group that shares one atom (called a spiro atom) between 5- to 20-membered monocyclic rings, wherein one or more ring atoms are selected from nitrogen, oxygen, S, and S (O) or S(O) 2 heteroatoms, and the remaining ring atoms are carbon. It can contain one or more double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan (for example, 7, 8, 9 or 10 yuan).
  • the spiro heterocyclic group is classified into a single spiro heterocyclic group, a dispiro heterocyclic group or a polyspiro heterocyclic group, preferably a single spiro heterocyclic group and a dispiro heterocyclic group. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered monospiro heterocyclic group.
  • spiroheterocyclic groups include:
  • fused heterocyclic group refers to a 5- to 20-membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system.
  • One or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system, where one or more of the ring atoms are heteroatoms selected from nitrogen, oxygen, S, S(O) or S(O) 2 , and the remaining ring atoms are carbon. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan (for example, 7, 8, 9 or 10 yuan).
  • fused heterocyclic groups include:
  • bridged heterocyclic group refers to a 5- to 14-membered polycyclic heterocyclic group with any two rings sharing two atoms that are not directly connected. It may contain one or more double bonds, but none of the rings has a complete common A conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen, S, S(O) or S(O) 2 and the remaining ring atoms are carbon. It is preferably 6 to 14 yuan, more preferably 7 to 10 yuan (for example, 7, 8, 9 or 10 yuan).
  • bridged heterocyclic groups include:
  • the heterocyclic ring includes the aforementioned heterocyclic group (such as spiro heterocyclic group, fused heterocyclic group and bridged heterocyclic group) fused to an aryl, heteroaryl or cycloalkyl ring, wherein the parent structure is connected to The ring together is a heterocyclic group, non-limiting examples of which include:
  • the heterocyclic group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthio, oxo, carboxy, or carboxylate.
  • aryl refers to a 6 to 14-membered all-carbon monocyclic or fused polycyclic (that is, rings sharing adjacent pairs of carbon atoms) with a conjugated ⁇ -electron system, preferably 6 to 10 members, such as benzene Base and naphthyl.
  • the aryl ring includes the above-mentioned aryl group fused to a heteroaryl, heterocyclic or cycloalkyl ring, wherein the ring connected to the parent structure is an aryl ring, and non-limiting examples thereof include:
  • Aryl groups may be substituted or unsubstituted.
  • the substituents are preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio group, carboxyl group or carboxylate group, preferably phenyl group.
  • heteroaryl refers to a heteroaromatic system containing 1 to 4 heteroatoms and 5 to 14 ring atoms, where the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • Heteroaryl groups are preferably 5 to 10 members (for example, 5, 6, 7, 8, 9 or 10 members), more preferably 5 or 6 members, such as imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl , Oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc., preferably imidazolyl, pyrazolyl, pyrimidinyl or thiazolyl; more preferably pyrazolyl or thiazole base.
  • the heteroaryl ring includes the above-mentioned heteroaryl group fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring connected to the parent structure is a heteroaryl ring, and non-limiting examples thereof include:
  • the heteroaryl group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , Heterocycloalkylthio, carboxyl or carboxylate.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group, where the alkyl group is as defined above.
  • haloalkyl refers to an alkyl substituted by halogen, where alkyl is as defined above.
  • deuterated alkyl refers to an alkyl group substituted with a deuterium atom, where the alkyl group is as defined above.
  • hydroxy refers to the -OH group.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • amino refers to -NH 2 .
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • aldehyde group refers to -CHO.
  • carboxylate group refers to -C(O)O (alkyl) or -C(O)O (cycloalkyl), where alkyl and cycloalkyl are as defined above.
  • acyl halide refers to a compound containing a -C(O)-halogen group.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may but need not be present, and the description includes the case where the heterocyclic group is substituted by an alkyl group and the case where the heterocyclic group is not substituted by an alkyl group .
  • 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 with 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 can determine (by experiment or theory) possible or impossible substitutions without too much effort. For example, an amino group or a hydroxyl group with free hydrogen may be unstable when combined with a carbon atom with an unsaturated (eg, olefinic) bond.
  • “Pharmaceutical composition” means a mixture containing one or more of the compounds described herein or their physiologically/pharmaceutically acceptable salts or prodrugs and other chemical components, and other components such as physiological/pharmaceutically acceptable carriers And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, facilitate the absorption of the active ingredients and then exert the biological activity.
  • “Pharmaceutically acceptable salt” refers to the salt of the compound of the present disclosure. Such salt is safe and effective when used in the body of a mammal, and has due biological activity.
  • X is selected from A, B, or C
  • X is selected from A, B and C
  • X is A, B or C
  • X is A, B and C
  • other different terms are all expressed In the same sense, it means that X can be any one or more of A, B, and C.
  • the compounds of the present disclosure may also include isotopic derivatives thereof.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound whose structure differs only in the presence of one or more isotopically enriched atoms.
  • 18 F-fluorine label 18 F isotope
  • 11 C-, 13 C-, or 14 C-rich Compounds in which a set of carbon ( 11 C-, 13 C-, or 14 C-carbon label; 11 C-, 13 C-, or 14 C- isotope) replace carbon atoms are within the scope of the present disclosure.
  • Such compounds can be used, for example, as analytical tools or probes in biological assays, or as tracers for in vivo diagnostic imaging of diseases, or as tracers for pharmacodynamics, pharmacokinetics, or receptor studies.
  • Deuterated compounds can generally retain activity comparable to that of non-deuterated compounds, and when deuterated at certain specific sites, they can achieve better metabolic stability, thereby obtaining certain therapeutic advantages (such as increased in vivo half-life or reduced dosage requirements) ).
  • the term "therapeutically effective amount” refers to a sufficient amount of a drug or agent that is non-toxic but can achieve the desired effect.
  • the determination of the effective amount varies from person to person, depends on the age and general condition of the subject, and also depends on the specific active substance. The appropriate effective amount in a case can be determined by those skilled in the art according to routine experiments.
  • the present disclosure provides a new type of estrogen receptor antagonist represented by the general formula (I), and it is found that the compound of this type of structure has good in vitro activity. Compared with the prior art, the alpha contained in this type of structure , ⁇ -unsaturated amide compounds, which can inactivate the estrogen receptor by specifically binding to the cysteine of the estrogen receptor ligand binding domain.
  • the compound represented by the general formula (I) of the present disclosure or its tautomer, meso, racemate, enantiomer, diastereomer, or mixture form thereof, or pharmacologically thereof includes the following steps:
  • the compound of general formula (IA) or its tautomer, mesosome, racemate, enantiomer, diastereomer or its mixture form, or its pharmaceutically acceptable salt in acid The deprotection reaction occurs under conditions to obtain a compound of general formula (I) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or Its medicinal salt;
  • R w is an amino protecting group; R is as defined in the general formula (IA);
  • R w is an amino protecting group
  • R is as defined in general formula (IIA);
  • Ring A, G, Z, R 1 , R 2, R 3, R 4, R a, R b, n, m, s and q are as formula (II) as defined above.
  • R w is an amino protecting group
  • R is as defined in general formula (IIIA);
  • G, Z, R 1, R 2, R 3, R 4, R a, R b, n, s and q are as formula (III) as defined above.
  • the compound of general formula (IVA) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture form thereof, or its pharmaceutically acceptable salt is acidic Deprotection reaction occurs under conditions to obtain the compound of general formula (IV) or its tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or Its medicinal salt;
  • R w is an amino protecting group
  • G, Z, R 1 , R 2 , R 3 , R 4 , R 8 , R 9 , R b , n, s and q are as defined in the general formula (IV).
  • the reagents that provide acidic conditions in the above solution include but are not limited to pyridine hydrobromide, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid or methanesulfonic acid, preferably hydrochloric acid.
  • the amino protecting group is selected from tetrahydropyranyl, tert-butoxycarbonyl, acetyl, benzyl, allyl and p-methoxybenzyl, preferably tetrahydropyranyl.
  • the above reaction is preferably carried out in a solvent.
  • the solvents used include but are not limited to: acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water, N,N- Dimethylformamide or its mixed solvent.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • NMR was measured with Bruker AVANCE-400 nuclear magnetic instrument, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), and the internal standard was four Methylsilane (TMS).
  • MS uses Agilent 1200/1290 DAD-6110/6120 Quadrupole MS liquid-mass spectrometer (manufacturer: Agilent, MS model: 6110/6120 Quadrupole MS); waters ACQuity UPLC-QD/SQD (manufacturer: waters, MS Model: waters ACQuity Qda Detector/waters SQ Detector); THERMO Ultimate 3000-Q Exactive (manufacturer: THERMO, MS model: THERMO Q Exactive).
  • HPLC High performance liquid chromatography analysis uses Agilent HPLC 1200DAD, Agilent HPLC 1200VWD and Waters HPLC e2695-2489 high pressure liquid chromatograph.
  • HPLC preparation uses Waters 2545-2767, Waters 2767-SQ Detecor2, Shimadzu LC-20AP and Gilson GX-281 preparative chromatographs.
  • CombiFlash rapid preparation instrument uses Combiflash Rf200 (TELEDYNE ISCO).
  • the thin layer chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate, the size of the silica gel plate used in thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm, and the size of thin layer chromatography separation and purification products is 0.4mm ⁇ 0.5mm.
  • the silica gel column chromatography generally uses Yantai Huanghai silica gel 200-300 mesh silica gel as the carrier.
  • the known starting materials of the present disclosure can be synthesized by or according to methods known in the art, or can be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc., Darui Chemicals and other companies.
  • reaction can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • the argon atmosphere or nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon with a volume of about 1L.
  • the hydrogen atmosphere refers to the reaction flask connected to a hydrogen balloon with a volume of about 1L.
  • the pressure hydrogenation reaction uses Parr 3916EKX hydrogenator and Qinglan QL-500 hydrogen generator or HC2-SS hydrogenator.
  • the hydrogenation reaction is usually evacuated, filled with hydrogen, and repeated three times.
  • the microwave reaction uses the CEM Discover-S 908860 microwave reactor.
  • the solution refers to an aqueous solution.
  • reaction temperature is room temperature, which is 20°C to 30°C.
  • the monitoring of the reaction progress in the examples adopts thin-layer chromatography (TLC).
  • the developing reagent used in the reaction, the eluent system of column chromatography used in the purification of the compound and the developing reagent system of thin-layer chromatography include: A: Dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of triethylamine and Adjust with alkaline or acidic reagents such as acetic acid.
  • Example 1 Using the synthetic route of Example 1, replace the raw material 1h in the fourth step with (E)-4-bromo-N,N-dimethylbut-2-enamide 5d (using the implementation on page 39 of the specification in the patent application US2016347717 (Prepared by the method disclosed in Example 1), 1 g of the raw material was replaced with 5c to obtain the title compound 5e (60 mg) with a yield of 50%.
  • Example 7 Using the synthetic route of Example 7, the first step raw material 7a was replaced with (1-aminocyclohexyl) methanol 8a (prepared by the well-known method "Journal of Medicinal Chemistry, 2004, 47(18), 4613-4626” ), the title compound 8b (650 mg) was prepared with a yield of 68%.
  • Dissolve compound 1d (150mg, 0.5mmol) in methyltetrahydrofuran (15mL), add double pinacol borate (140mg, 0.6mmol) and tetrakistriphenylphosphine platinum (28mg, 0.02mmol), and replace the argon Gas was blown 3 times, and the temperature was raised to 85°C and stirred for 3 hours.
  • compound 9b (167 mg, 0.4 mmol), bistriphenylphosphorus palladium dichloride (31 mg, 0.05 mmol), cesium carbonate (300 mg, 0.9 mmol) and water (1 mL) were added and stirred at room temperature overnight.
  • the raw material 1g in the fourth step was replaced with compound 11f, and the raw material 1h was replaced with 11b, to obtain the title compound 11g (50mg) with a yield of 86%.
  • the raw material 1g in the fourth step was replaced with compound 11f, and the raw material 1h was replaced with 13a to obtain the title compound 13b (41 mg) with a yield of 81%.
  • the raw material 1g in the fourth step was replaced with compound 11f, and the raw material 1h was replaced with 5d to obtain the title compound 14a (80 mg) with a yield of 79%.
  • the raw material 1g in the fourth step was replaced with compound 11f, and the raw material 1h was replaced with 2a to obtain the title compound 15a (49 mg) with a yield of 72%.
  • Dissolve compound 11a (0.5g, 3.0mmol) in dichloromethane (10mL), add triethylamine (0.4g, 3.6mmol), tert-butylamine (0.2g, 3.0mmol), 1-(3-dimethylamino) at room temperature
  • triethylamine 0.4g, 3.6mmol
  • tert-butylamine 0.2g, 3.0mmol
  • Propyl)-3-ethylcarbodiimide hydrochloride 0.7 g, 3.6 mmol
  • 1-hydroxybenzotriazole 0.5 g, 3.6 mmol
  • the raw material 1g in the fourth step was replaced with compound 11f, and the raw material 1h was replaced with 16a to obtain the title compound 16b (47 mg) with a yield of 83%.
  • the raw material 1g in the fourth step was replaced with compound 11f, and the raw material 1h was replaced with 17a to obtain the title compound 17b (42 mg) with a yield of 73%.
  • Example 19 Using the synthetic route of Example 19, the third step raw material 19c was replaced with N-(4-iodophenyl)acetamide 20a (using the well-known method "Journal of Organic Chemistry, 2018, 83(2), 930-938" Prepared), the title compound 20b (70 mg) was prepared with a yield of 61%.
  • Example 19 Using the synthetic route of Example 19, the third step raw material 19c was replaced with 4-iodo-N-methylbenzamide 21a (using the well-known method "Journal of the American Chemical Society, 2013, 135(12), 4628- 4631"), the title compound 21b (50 mg) was prepared, and the yield was 44%.
  • the third step raw material 19c was replaced with 1-iodo-4-(trifluoromethyl)benzene 22a to obtain the title compound 22b (80 mg) with a yield of 69%.
  • Example 19 Using the synthetic route of Example 19, the third step raw material 19c was replaced with 1-iodo-4-(methylsulfonyl)benzene 23a (prepared by the well-known method "Organic Letters, 2014, 16(9), 2306-2309" Rather), the title compound 23b (60 mg) was prepared with a yield of 51%.
  • the first step raw material 9d was replaced with compound 11f to obtain the title compound 25a (37 mg) with a yield of 83%.
  • the first step raw material 9d was replaced with compound 11f, and the raw material 10a was replaced with acrylic acid 26a to obtain the title compound 26b (30 mg) with a yield of 73%.
  • the second step raw material 10b was replaced with compound 26b to obtain the title compound 26 (3 mg) with a yield of 12%.
  • Example 2 Using the synthetic route of Example 1, the first step raw material 1b was replaced with (S)-3-hydroxypiperidine-1-carboxylic acid tert-butyl ester 27a (using the well-known method "Tetrahedron, 2007, 63(2), 331 -336"), the title compound 27b (531 mg) was prepared, and the yield was 92%.
  • the fourth step raw material 1g was replaced with compound 27d, and the raw material 1h was replaced with compound 5d to obtain the title compound 27e (65 mg) with a yield of 64%.
  • the first step raw material 10a was replaced with compound 26a to obtain the title compound 28a (50 mg) with a yield of 63%.
  • the first step raw material 1b was replaced with (R)-3-hydroxypyrroline-1-carboxylic acid tert-butyl ester 29a to obtain the title compound 29b (0.9g) with a yield of 86%.
  • the first step raw material 1b was replaced with (S)-3-hydroxypyrroline-1-carboxylic acid tert-butyl ester 30a to obtain the title compound 30b (0.5 g) with a yield of 50%.
  • the fourth step raw material 1g was replaced with compound 30d, and the raw material 1h was replaced with compound 5d to obtain the title compound 30e (70 mg) with a yield of 62%.
  • the first step raw material 1b was replaced with (R)-3-aminopiperidine-1-carboxylic acid tert-butyl ester 31a to obtain the title compound 31b (0.8 g) with a yield of 57%.
  • the raw material 1g in the fourth step was replaced with compound 31d, and the raw material 1h was replaced with compound 5d to obtain the title compound 31 (20 mg) with a yield of 39%.
  • the first step raw material 1b was replaced with 4-aminopiperidine-1-carboxylic acid tert-butyl ester 33a to obtain the title compound 33b (0.2 g) with a yield of 20%.
  • the raw material 1g in the fourth step was replaced with compound 33d, and the raw material 1h was replaced with compound 5d to obtain the title compound 33e (70 mg) with a yield of 86%.
  • the first step raw material 1b was replaced with (R)-pyrrolin-3-ylcarbamate tert-butyl 34a to obtain the title compound 34b (0.8 g) with a yield of 70%.
  • the first step raw material 1b was replaced with (S)-pyrrolin-3-ylcarbamate tert-butyl 35a to obtain the title compound 35b (0.8 g) with a yield of 69%.
  • the raw material 1g in the fourth step was replaced with compound 35d, and the raw material 1h was replaced with compound 5d to obtain the title compound 35e (40 mg) with a yield of 61%.
  • the raw material 1g in the fourth step was replaced with compound 36d, and the raw material 1h was replaced with compound 5d to obtain the title compound 36e (70 mg) with a yield of 86%.
  • the raw material 1g in the fourth step was replaced with compound 37e, and the raw material 1h was replaced with compound 5d to obtain the title compound 37f (75 mg) with a yield of 79%.
  • the second step raw material 1c was replaced with compound 37b to obtain the title compound 38a (100 mg) with a yield of 80%.
  • Test Example 1 The inhibitory effect of the compound of the present disclosure on the activity of the estrogen receptor reporter gene
  • MCF7 cells MCF7/ERE-luc (ATCC, HTB-22) expressing the luciferase reporter gene ERE-luc (synthesized by Jinweizhi Biotechnology Co., Ltd.) controlled by the estrogen receptor response element use 10% fetal bovine serum and 500 ⁇ g /ml G418 MEM (GE Healthcare, SH30024.01) medium for culture.
  • ERE-luc luciferase reporter gene
  • MCF7/ERE-luc cells were seeded in a 96-well plate at a density of 30,000 cells/well using an incomplete MEM medium containing 10% activated carbon-treated fetal bovine serum (BioSun, BS-0004-500).
  • the inhibitory effect of the compounds in the present disclosure on the activity of the estrogen receptor reporter gene was determined by the above test.
  • Graghpad Prism was used to plot the chemiluminescence signal value and the logarithmic concentration of the compound.
  • the measured IC 50 value is shown in Table 1 below.
  • Example number IC 50 (nM) 1 1 2 3 3 4 4 4 5 3 6 0.5 7 38 8 3 9 2 10 8 11 3 12 2 13 3 14 2 15 3 16 5 17 6 18 6 19 4 twenty two twenty four twenty three 13 twenty four 9
  • the compound of the present disclosure has a significant inhibitory effect on the estrogen receptor reporter gene.
  • Test Example 2 The inhibitory effect of the compound of the present disclosure on the proliferation of MCF7 cells
  • MCF7 cells (ATCC, HTB-22) were cultured in MEM (GE Healthcare, SH30024.01) complete medium containing 10% fetal bovine serum.
  • MEM GE Healthcare, SH30024.01
  • MCF7 cells were seeded in a 96-well plate at a density of 3,000 cells/well using complete medium, with 100 ⁇ l cell suspension per well, and placed in a 37°C, 5% CO 2 cell incubator overnight. The next day, the medium was aspirated, and each well was replaced with 135 ⁇ l of MEM incomplete medium containing 2% fetal bovine serum. At the same time, each well was added with 15 ⁇ l of different concentrations of test compound prepared with incomplete medium.
  • the final concentration of the compound was Starting from 100 nM, perform 4-fold gradient dilution at 9 concentration points, set a blank control containing 0.5% DMSO, and place it in a 37°C, 5% CO 2 cell incubator for 144 hours.
  • On the eighth day take out the 96-well cell culture plate and add 150 ⁇ l to each well Luminescent Cell Viability Assay (Promega, G7573), after standing at room temperature for 10 minutes, use a multi-label microplate microplate reader (PerkinElmer, VICTOR 3) to read the luminescence signal value, and use Graphpad Prism software to calculate according to the compound concentration and luminescence signal value The IC 50 value of the compound's inhibitory activity.
  • the compound of the present disclosure has a significant inhibitory effect on the proliferation of MCF7 cells.
  • Test Example 3 The inhibitory effect of the compound of the present disclosure on the proliferation of MCF7 cells expressing ER ⁇ mutant
  • the purpose of this experiment is to determine the inhibitory activity of the compounds of the present disclosure on the proliferation of ERa mutant MCF7 cells.
  • mutants ER ⁇ Y537S and ER ⁇ D538G of human estrogen receptor ⁇ (ER ⁇ ) protein were obtained by site-directed mutation using the cDNA (Accession No. NM000125) of the wild-type ESR1 gene as a template by double-primer PCR.
  • the primer sequences used for mutation are as follows (the nucleotides underlined are the sites of mutation):
  • the cDNA of mutant ESR1 was cloned into the target lentiviral vector pCDH-CMV-MCS-EF1-Puro (SBI, CD510B-1). Then the lentiviral plasmid with the mutant ESR1 gene sequence and the lentiviral packaging plasmids pVSV-G and pCMV-dR8.91 (SBI, LV500A-1) were transfected by Lipofectamine 3000 Transfection Reagent (ThermoFisher Scientific, Cat#L3000075) HEK-293T cells (ATCC, CRL-3216).
  • the MCF7 cells expressing the ERa mutant were cultured in MEM (GE Healthcare, SH30024.01) complete medium containing 10% fetal bovine serum.
  • MEM GE Healthcare, SH30024.01
  • complete medium containing 10% fetal bovine serum.
  • cells were seeded in a 96-well plate at a density of 3,000 cells/well using complete medium, with 100 ⁇ l cell suspension per well, and placed in a 37°C, 5% CO 2 cell incubator overnight.
  • the medium was aspirated, and each well was replaced with 135 ⁇ l of MEM incomplete medium containing 2% fetal bovine serum.
  • each well was added with 15 ⁇ l of different concentrations of test compound prepared with incomplete medium.
  • the final concentration of the compound was Starting from 100 nM, perform 4-fold gradient dilution at 9 concentration points, set a blank control containing 0.5% DMSO, and place it in a 37°C, 5% CO 2 cell incubator for 144 hours.
  • On the eighth day take out the 96-well cell culture plate and add 150 ⁇ l to each well Luminescent Cell Viability Assay (Promega, G7573), after standing at room temperature for 10 minutes, use a multi-label microplate microplate reader (PerkinElmer, VICTOR 3) to read the luminescence signal value, and use Graphpad Prism software to calculate according to the compound concentration and luminescence signal value The IC 50 value of the compound's inhibitory activity.
  • the compounds of the present disclosure have a significant inhibitory effect on the proliferation of MCF7 cells expressing ER ⁇ mutants.
  • Test Example 4 Determination of the inhibitory activity of the compound of the present invention on Na v 1.5
  • Na V 1.5 ion channel is stably expressed on HEK293 cells. After the Na V 1.5 current is stable, comparing the Na V 1.5 current before and after the compound application, the influence of the compound on the Na V 1.5 ion channel can be obtained.
  • Patch clamp amplifier patch clamp PC-505B (WARNER instruments)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (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)
  • Peptides Or Proteins (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

吲唑类衍生物、其制备方法及其在医药上的应用。特别地,涉及通式(I)所示的吲唑类衍生物、其制备方法及含有该衍生物的药物组合物,以及其作为雌激素受体调节剂预防和/或治疗雌激素受体介导的或依赖性的疾病或病症的用途,所述的疾病特别优选乳腺癌。其中通式(I)中的各取代基与说明书中的定义相同。

Description

吲唑类衍生物、其制备方法及其在医药上的应用 技术领域
本公开属于医药领域,涉及一种吲唑类衍生物、其制备方法及其在医药上的应用,本公开公开了其作为雌激素受体调节剂预防和/或治疗雌激素受体介导的或依赖性的疾病或病症的用途,所述的疾病特别优选乳腺癌。
背景技术
乳腺癌是女性最常见的恶性肿瘤之一,据2012年GLOBALCAN统计数据显示(CA CANCER J CLIN 2015;65:87–108),全球一年约有170万新发癌症病例,52万死亡病例,无论发病率和死亡率都居女性恶性肿瘤首位。国家癌症中心发布的2017年《中国肿瘤登记年报》显示,乳腺癌居女性恶性肿瘤发病率首位,每年新发病例约27.9万,并以每年2%左右的速度递增。
约有70%的乳腺癌患者为雌激素受体(estrogen receptor,ER)阳性乳腺癌。在这部分乳腺癌患者的治疗中,内分泌治疗(endocrine therapy)占有重要地位。内分泌治疗主要分三类,分别是:芳香化酶抑制剂(aromatase inhibitor,AI),能够抑制雄激素转化为雌激素,降低体内雌激素的水平;选择性雌激素受体调节剂(selective estrogen receptor modulator,SERM),拮抗雌激素受体的活性;和选择性雌激素受体降解剂(selective estrogen receptor degrader,SERD),不仅可以拮抗雌激素受体的活性,还能够促进受体的降解(Pharmacol Ther.2017 Dec 28)。虽然内分泌治疗是雌激素受体阳性乳腺癌的首选治疗,但约有30%接受辅助治疗的病人会发生复发,而几乎所有的转移性乳腺癌病人都会产生耐药而发生进展。对内分泌治疗产生耐药的机制主要分两类,一类集中在雌激素受体信号通路本身,包括编码雌激素受体的基因ESR1的激活突变、扩增、与其他基因的融合,雌激素受体共调解因子和下游控制细胞周期因子的失调等,另一类机制包括与雌激素受体信号通路有交叉反应的信号通路的激活,如生长因子受体通路等(Nat Rev Clin Oncol.2015 Oct;12(10):573-83)。
2013年两项研究,在11~55%的接受过芳香化酶抑制剂治疗的雌激素受体阳性转移性乳腺癌病人中检测到了ESR1基因突变,进一步研究发现突变受体可以不依赖雌激素发生磷酸化,发挥转录作用,使雌激素依赖的MCF7接种的肿瘤在体内可以不再依赖雌激素生长,而且突变受体会使SERM他莫昔芬(tamoxifen)和SERD氟维司群(fulvestrant)的活性降低。因此ESR1基因突变可能是雌激素阳性乳腺癌发生耐药的机制之一(Nat Rev Clin Oncol.2015 Oct;12(10):573-83and Nat Genet 2013;45:1439–45)。在随后进行的多个研究中,都在雌激素受体阳性转移性乳腺癌病人中发现了一定比例的ESR1基因突变,突变比例大约在30%左右。在BOLERO-2临床试验中发现,经过AIs治疗后进展的雌激素受体阳性转移性乳腺 癌病人的ctDNA中有29%存在ER Y537S和ER D538G突变。在依西美坦(exemestane)单用组,发生突变病人的无进展生存期(progression free survival,PFS)和总生存期(overall survival,OS)都比没有发生突变的病人短(Nat Genet 2013;45:1446–51)。
综上所述,ESR1基因突变大多发生在经过AIs治疗而进展的转移性雌激素受体阳性乳腺癌病人中,这些病人对AIs治疗不再敏感,因此需要开发针对ESR1基因突变的雌激素受体拮抗剂。
Eisai公司开发的first-in-class的雌激素受体共价结合拮抗剂H3B-6545对野生型和突变型雌激素受体都有较强的抑制活性,且能够通过和受体的共价结合发挥更长时间的药效,目前正在进行临床一二期试验。目前公开的针对ESR1基因突变的雌激素受体拮抗剂的专利有WO2016196346和WO2016196342。
发明内容
本公开的目的在于提供一种通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐:
Figure PCTCN2020096744-appb-000001
其中:
G为CH或N;
Z选自键、CR 5R 6、-O-(CH 2)t-或-NR 7-(CH 2)t-;
环A选自环烷基或杂环基;
R a选自-CH 2CH=CHC(O)NR 8R 9、-C(O)CH=CR 10R 11和-C(O)C≡CR 12
R b选自氢原子或烷基;
R 1各自相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 2选自氢原子、卤素、烷基、卤代烷基、烷氧基、氨基、氰基、硝基、羧基、醛基、羟基、羟烷基、环烷基、芳基和杂芳基;
R 3各自相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、NR 13C(O)R 14、C(O)NR 13R 14、SO 2R 15、环烷基、杂环基、芳基和杂芳基;
R 4各自相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 5和R 6相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 7选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 8和R 9相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
或者,R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R 10和R 11相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 12选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 13和R 14相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 15选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
m为0或1;
n为0、1、2或3;
q为0、1、2、3、4或5;
s为0、1、2或3且
t为0、1、2、3、4、5或6。
在本公开一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中环A选自C 3-C 6环烷基、3至6元的杂环基;所述的杂环基含有1~3个选自N、O或S的杂原子。
在本公开的另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中环A选自环丙基、环戊基、环己基、四氢吡咯基或哌啶基。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中环A选自
Figure PCTCN2020096744-appb-000002
Figure PCTCN2020096744-appb-000003
优选
Figure PCTCN2020096744-appb-000004
Figure PCTCN2020096744-appb-000005
在本公开一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐,其为通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐:
Figure PCTCN2020096744-appb-000006
其中
环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如通式(I)中所定义。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,其为通式(III)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐:
Figure PCTCN2020096744-appb-000007
其中:G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如通式(I)中所定义。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,其为通式(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐:
Figure PCTCN2020096744-appb-000008
其中:
G、Z、R 1、R 2、R 3、R 4、R 8、R 9、R b、n、s和q如通式(I)中所定义;
优选地,R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;更优选地,所述的杂环基任选被羟基或羟烷基取代;进一步优选地,所述的杂环基任选被羟基取代。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中G为N原子。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 1为选自卤素或氢原子。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 2为卤代烷基,优选C 1-C 6卤代烷基,更优选-CH 2CF 3
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 3选自氢原子、卤素、烷基、氰基、卤代烷基、NR 13C(O)R 14、C(O)NR 13R 14和SO 2R 15;R 13和R 14相同或不同,且各自独立地选自氢原子和烷基;R 15选自氢原子和烷基。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 4相同或不同,各自独立地选自氢原子、卤素、烷基和烷氧基,优选氢原子。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中Z选自键、-O-、-O-CH 2-和-NH-。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R a选自-CH 2CH=CHC(O)NR 8R 9、-C(O)CH=CR 10R 11和-C(O)C≡CR 12;R 8和R 9相同或不同,且各自独立地选自氢原子、烷基、环烷基或杂环基;或者R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;R 10和R 11相同或不同,各自独立地为氢原子或烷基;R 12选自氢原子或烷基。
在本公开另一个优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R a选自-CH 2CH=CHC(O)N(CH 3) 2、-CH 2CH=CHC(O)NH(CH 3)、-CH 2CH=CHC(O)NHC(CH 3) 3、-C(O)CH=CH 2、-C(O)C≡CCH 3
Figure PCTCN2020096744-appb-000009
Figure PCTCN2020096744-appb-000010
优选地,R a选自-CH 2CH=CHC(O)N(CH 3) 2、-CH 2CH=CHC(O)NH(CH 3)、-CH 2CH=CHC(O)NHC(CH 3) 3、-C(O)CH=CH 2、-C(O)C≡CCH 3
Figure PCTCN2020096744-appb-000011
Figure PCTCN2020096744-appb-000012
通式(I)的典型化合物,包括但不限于:
Figure PCTCN2020096744-appb-000013
Figure PCTCN2020096744-appb-000014
Figure PCTCN2020096744-appb-000015
Figure PCTCN2020096744-appb-000016
Figure PCTCN2020096744-appb-000017
Figure PCTCN2020096744-appb-000018
Figure PCTCN2020096744-appb-000019
Figure PCTCN2020096744-appb-000020
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐。
本公开的另一方面,提供一种通式(IA)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,
Figure PCTCN2020096744-appb-000021
其为合成通式(I)的中间体,
其中:
R w为氨基保护基,优选四氢吡喃基;
R选自-CH 2CH=CHC(O)NR 8R 9、-C(O)CH=CR 10R 11或-C(O)C≡CR 12
R 8和R 9相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
或者,R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、-COOR 16-、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R 10和R 11相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 12选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 16选自氢原子、烷基、卤代烷基和羟烷基;
环A、G、Z、R 1、R 2、R 3、R 4、R b、n、m、s和q如通式(I)中所定义。
本公开的另一方面,提供一种通式(IIA)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,
Figure PCTCN2020096744-appb-000022
其为合成通式(II)的中间体,
其中:
R w为氨基保护基,优选四氢吡喃基;
R如通式(IA)中所定义;
环A、G、Z、R 1、R 2、R 3、R 4、R b、n、m、s和q如通式(II)中所定义。
本公开的另一方面,提供一种通式(IIIA)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,
Figure PCTCN2020096744-appb-000023
其为合成通式(III)的中间体,
其中:
R w为氨基保护基,优选四氢吡喃基;
R如通式(IA)中所定义;
G、Z、R 1、R 2、R 3、R 4、R b、n、s和q如通式(III)中所定义。
本公开的另一方面,提供一种通式(IVA)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,
Figure PCTCN2020096744-appb-000024
其为合成通式(IV)的中间体,
其中:
R w为氨基保护基,优选四氢吡喃基;
G、Z、R 1、R 2、R 3、R 4、R b、R 8、R 9、n、s和q如通式(IV)中所定义。
通式(IA)的典型化合物,包括但不限于:
Figure PCTCN2020096744-appb-000025
Figure PCTCN2020096744-appb-000026
Figure PCTCN2020096744-appb-000027
Figure PCTCN2020096744-appb-000028
Figure PCTCN2020096744-appb-000029
Figure PCTCN2020096744-appb-000030
Figure PCTCN2020096744-appb-000031
Figure PCTCN2020096744-appb-000032
Figure PCTCN2020096744-appb-000033
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐。
本公开另一方面,提供一种制备根据通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2020096744-appb-000034
化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(I)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基,优选四氢吡喃基或叔丁氧羰基;
R如通式(IA)中所定义;
环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如通式(I)中所定义。
本公开另一方面,提供一种制备根据通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2020096744-appb-000035
通式(IIA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(II)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基,优选四氢吡喃基或叔丁氧羰基;
R如通式(IIA)中所定义;
环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如通式(II)中所定义。
本公开另一方面,提供一种制备根据通式(III)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2020096744-appb-000036
通式(IIIA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对 映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(III)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基,优选四氢吡喃基或叔丁氧羰基;
R如通式(IIIA)中所定义;
G、Z、R 1、R 2、R 3、R 4、R a、R b、n、s和q如通式(III)中所定义。
本公开另一方面,提供一种制备根据通式(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐的方法,该方法包括:
Figure PCTCN2020096744-appb-000037
通式(IVA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(IV)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基,优选四氢吡喃基或叔丁氧羰基;
G、Z、R 1、R 2、R 3、R 4、R 8、R 9、R b、n、s和q如通式(IV)中所定义。
本公开的另一方面涉及一种药物组合物,其含有治疗有效剂量的上述各通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。本公开还涉及一种制备上述药物组合物的方法,其包括将各通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐与药学上可接受的载体、稀释剂或赋形剂相混合。
本公开进一步涉及通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐或包含其的药物组合物在制备雌激素受体调节剂中的用途。
本公开进一步涉及通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐或包含其的药物组合物在制备预防和/或治疗雌激素受体介导的或依赖性的疾病或病症的药物中的用途;其中所述雌激素受体介导的或依赖性的疾病或病症优选为癌症,更优选为乳腺癌、卵巢癌、子宫内膜癌、前列腺癌或子宫癌;进一步优选乳 腺癌。
本公开进一步涉及一种用作药物的通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐。
本公开进一步涉及一种本公开的通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其作为治疗雌激素受体介导的或依赖性的疾病或病症的药物;其中所述雌激素受体介导的或依赖性的疾病或病症优选为癌症,更优选为乳腺癌、卵巢癌、子宫内膜癌、前列腺癌或子宫癌;进一步优选乳腺癌。
本公开进一步涉及一种治疗雌激素受体介导的或依赖性的疾病或病症的方法,该方法包括向需要其的患者施用治疗有效剂量的本公开的通式(I)至(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐。该方法表现出突出的疗效和较少的副作用。其中所述雌激素受体介导的或依赖性的疾病或病症优选为癌症,更优选为乳腺癌、卵巢癌、子宫内膜癌、前列腺癌或子宫癌;进一步优选乳腺癌。
可将活性化合物制成适合于通过任何适当途径给药的形式,活性化合物优选是以单位剂量的方式,或者是以患者可以以单剂自我给药的方式。本发明化合物或组合物的单位剂量的表达方式可以是片剂、胶囊、扁囊剂、瓶装药水、药粉、颗粒剂、锭剂、栓剂、再生药粉或液体制剂。
本发明治疗方法中所用化合物或组合物的剂量通常将随疾病的严重性、患者的体重和化合物的相对功效而改变。不过,作为一般性指导,合适的单位剂量可以是0.1~1000mg。
本发明的药物组合物除活性化合物外,可含有一种或多种辅料,所述辅料选自以下成分:填充剂(稀释剂)、粘合剂、润湿剂、崩解剂或赋形剂等。根据给药方法的不同,组合物可含有0.1至99重量%的活性化合物。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。这些赋形剂可以是惰性赋形剂;造粒剂和崩解剂;和润滑剂。这些片剂可以不包衣或可通过掩盖药物的味道或在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。例如,可使用水溶性味道掩蔽物质。也可用其中活性成分与惰性固体稀释剂混合,或其中活性成分与水溶性载体混合的软明胶胶囊提供口服制剂。
水悬浮液含有活性物质和用于混合的适宜制备水悬浮液的赋形剂。此类赋形 剂是悬浮剂、分散剂或湿润剂。水混悬液的活性成分可以是可分散粉末或颗粒。通过加入水,活性成分与一种或多种分散剂、湿润剂或悬浮剂混合。水混悬液还可以含有一种或多种防腐剂、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂。
油混悬液可通过使活性成分悬浮于植物油或矿物油中配制而成。油悬浮液可含有增稠剂。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂保存这些组合物。
通过加入水可使适用于制备水混悬的可分散粉末和颗粒提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂可说明上述的例子。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
本公开的药物组合物也可以是水包油乳剂的形式。油相可以是植物油或矿物油或其混合物。适宜的乳化剂可以是天然产生的磷脂。可用甜味剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本公开的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳。可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本公开化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。这种装置的实例是Deltec CADD-PLUS.TM.5400型静脉注射泵。
本公开的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在肠胃外可接受的无毒稀释剂或溶剂中制备的无菌注射溶液或混悬液。此外,可方便地用无菌固定油作为溶剂或悬浮介质此外,脂肪酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本公开化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行为、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合等;另外,最佳的治疗方式如治疗的模式、通式化合物(I)的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链 基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、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、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、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。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“亚烷基”指饱和的直链或支链脂肪族烃基,其具有2个从母体烷的相同碳原子或两个不同的碳原子上除去两个氢原子所衍生的残基,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子,更优选含有1至6个碳原子的亚烷基。亚烷基的非限制性实例包括但不限于亚甲基(-CH 2-)、1,1-亚乙基(-CH(CH 3)-)、1,2-亚乙基(-CH 2CH 2)-、1,1-亚丙基(-CH(CH 2CH 3)-)、1,2-亚丙基(-CH 2CH(CH 3)-)、1,3-亚丙基(-CH 2CH 2CH 2-)、1,4-亚丁基(-CH 2CH 2CH 2CH 2-)等。亚烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选独立地任选选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、硫醇、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、杂环烷硫基和氧代基中的一个或多个取代基所取代。
术语“烯基”指分子中含有碳碳双键的烷基化合物,其中烷基的定义如上所述。烯基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自氢原子、烷基、烷氧基、卤素、卤代烷基、羟基、羟烷基、 氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代。
术语“炔基”指分子中含有碳碳三键的烷基化合物,其中烷基的定义如上所述。炔基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自氢原子、烷基、烷氧基、卤素、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基和环烷基的定义如本文所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子(例如3、4、5、6、7、8、9、10、11、12个),更优选包含3至6个碳原子。单环环烷基的非限制性实例包括环丙基、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;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其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电子系统。优选为6至14元(例如6、7、8、9、10、11、12、13、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环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2020096744-appb-000038
术语“稠环烷基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中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电子系统。优选为6至14元,更优选为7至10元(例如7、8、9或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:
Figure PCTCN2020096744-appb-000039
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其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电子系统。优选为6至14元,更优选为7至10元(例如7、8、9或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:
Figure PCTCN2020096744-appb-000040
所述环烷基环包括上述环烷基(例如螺环烷基、稠环烷基和桥环烷基)稠合于芳基、杂芳基或杂环烷基环上,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。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧、S、S(O)或S(O) 2的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子(例如1、2、3和4个);更优选包含3至6个环原子(例如3、4、5、6个)。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等,优选哌啶基、吡咯烷基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧、S、S(O)或S(O) 2的杂原子,其余环原子为碳。其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电子系统。优选为6至14元,更优选为7至10元(例如7、8、9或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:
Figure PCTCN2020096744-appb-000041
术语“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,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电子系统,其中一个或多个环原子为选自氮、氧、S、S(O)或S(O) 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(例如7、8、9或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:
Figure PCTCN2020096744-appb-000042
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其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电子系统,其中一个或多个环原子为选自氮、氧、S、S(O)或S(O) 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(例如7、8、9或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。桥杂环基的非限制性实例包括:
Figure PCTCN2020096744-appb-000043
所述杂环基环包括上述杂环基(例如螺杂环基、稠杂环基和桥杂环基)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,其非限制性实例包括:
Figure PCTCN2020096744-appb-000044
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,例如苯基和萘基。所述芳基环包括上述芳基稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2020096744-appb-000045
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基,优选苯基。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元(例如5、6、7、8、9或10元),更优选为5元或6元,例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等,优选为咪唑基、吡唑基、嘧啶基或噻唑基;更优选为吡唑基或噻唑基。所述杂芳基环包括上述杂芳基稠合于芳基、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,其非限制性实例包括:
Figure PCTCN2020096744-appb-000046
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、 杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
术语“卤代烷基”指被卤素取代的烷基,其中烷基如上所定义。
术语“氘代烷基”指被氘原子取代的烷基,其中烷基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
术语“羧基”指-C(O)OH。
术语“醛基”指-CHO。
术语“羧酸酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基和环烷基如上所定义。
术语“酰卤”指含有-C(O)-卤素的基团的化合物。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“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。
“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。
“可药用盐”是指本公开化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
本公开中“X选自A、B、或C”、“X选自A、B和C”、“X为A、B或C”、“X为A、B和C”等不同用语均表达了相同的意义,即表示X可以是A、B、C中任意一种或几种。
本公开的化合物还可包含其同位素衍生物。术语“同位素衍生物”指结构不同仅在于存在一种或多种同位素富集原子的化合物。例如,具有本公开的结构,除了用“氘”或“氚”代替氢,或者用 18F-氟标记( 18F同位素)代替氟,或者用 11C-、 13C-、或 14C-富集的碳( 11C-、 13C-、或者 14C-碳标记; 11C-、 13C-、或 14C-同位素) 代替碳原子的化合物处于本公开的范围内。这样的化合物可用作例如生物学测定中的分析工具或探针,或者可以用作疾病的体内诊断成像示踪剂,或者作为药效学、药动学或受体研究的示踪剂。氘代物通常可以保留与未氘代的化合物相当的活性,并且当氘代在某些特定位点时可以取得更好的代谢稳定性,从而获得某些治疗优势(如体内半衰期增加或剂量需求减少)。
针对药物或药理学活性剂而言,术语“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于受试者的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
本公开提供了一种新型结构通式(I)所示的雌激素受体拮抗剂,并发现该类结构的化合物具有很好的体外活性,与现有技术相比该类结构中含有的α,β不饱和键的酰胺化合物,它们可通过与雌激素受体配体结合域的半胱氨酸的特异性结合而使雌激素受体失活。
本公开化合物的合成方法
为了完成本公开的目的,本公开采用如下技术方案:
方案一
本公开通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2020096744-appb-000047
通式(IA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(I)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基;R如通式(IA)中所定义;
环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如通式(I)中所定义。
方案二
Figure PCTCN2020096744-appb-000048
通式(IIA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对 映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(II)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基;
R如通式(IIA)中所定义;
环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如通式(II)中所定义。
方案三
Figure PCTCN2020096744-appb-000049
通式(IIIA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(III)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基;
R如通式(IIIA)中所定义;
G、Z、R 1、R 2、R 3、R 4、R a、R b、n、s和q如通式(III)中所定义。
方案四
Figure PCTCN2020096744-appb-000050
通式(IVA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(IV)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
其中:
R w为氨基保护基;
G、Z、R 1、R 2、R 3、R 4、R 8、R 9、R b、n、s和q如通式(IV)中所定义。
以上方案提供酸性条件的试剂包括但不限于吡啶氢溴酸盐、三氟乙酸、甲酸、乙酸、盐酸、硫酸或甲磺酸,优选为盐酸。
所述氨基保护基选自四氢吡喃基、叔丁氧羰基、乙酰基、苄基、烯丙基和对甲氧苄基,优选为四氢吡喃基。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、甲苯、四氢呋喃、二氯甲烷、二甲基亚砜、1,4-二氧六环、水、N,N-二甲基甲酰胺或其混合溶剂。
具体实施方式
以下结合实施例进一步描述本公开,但这些实施例并非限制着本公开的范围。
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10 -6(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6)、氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS)。
MS的测定用Agilent 1200/1290 DAD-6110/6120 Quadrupole MS液质联用仪(生产商:Agilent,MS型号:6110/6120 Quadrupole MS);waters ACQuity UPLC-QD/SQD(生产商:waters,MS型号:waters ACQuity Qda Detector/waters SQ Detector);THERMO Ultimate 3000-Q Exactive(生产商:THERMO,MS型号:THERMO Q Exactive)。
高效液相色谱法(HPLC)分析使用Agilent HPLC 1200DAD、Agilent HPLC 1200VWD和Waters HPLC e2695-2489高压液相色谱仪。
手性HPLC分析测定使用Agilent 1260 DAD高效液相色谱仪。
高效液相制备使用Waters 2545-2767、Waters 2767-SQ Detecor2、Shimadzu LC-20AP和Gilson GX-281制备型色谱仪。
手性制备使用Shimadzu LC-20AP制备型色谱仪。
CombiFlash快速制备仪使用Combiflash Rf200(TELEDYNE ISCO)。
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。
硅胶柱色谱法一般使用烟台黄海硅胶200~300目硅胶为载体。
激酶平均抑制率及IC 50值的测定用NovoStar酶标仪(德国BMG公司)。
本公开的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH&Co.KG、Acros Organics、Aldrich Chemical Company、韶远化学科技(Accela ChemBio Inc)、达瑞化学品等公司。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂,纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷/甲醇体系,B:正己烷/乙酸乙酯体系,C:石油醚/乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1
(E)-1-吗啉基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮1
Figure PCTCN2020096744-appb-000051
第一步
(1-(((5-碘吡啶-2-基)氧基)甲基)环丙基)氨基甲酸叔丁酯1c
将氢化钠(0.4g,10.7mmol)溶于N,N-二甲基甲酰胺(20mL),于室温加入1-(羟甲基)环丙基氨基甲酸叔丁酯1b(1.0g,5.3mmol,采用公知的方法“Journal of Organic Chemistry,2002,67(11),3965-3968”制备而得),加完后缓慢加入2-氟-5- 碘吡啶1a(1.8g,8.0mmol)。室温搅拌2小时后停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物1c(2.4g),产率:86%。
MS m/z(ESI):391.0[M+1]。
第二步
(Z)-(1-(((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基甲酸叔丁酯1f
将3-氟-1-(四氢-2H-吡喃-2-基)-5-(4,4,4-三氟丁-1-炔-1-基)-1H-吲唑1d(1.8g,5.5mmol,采用专利申请WO2018098305中说明书第84页的实施例3公开的方法制备得到)溶于甲基四氢呋喃(40mL),加入双联频哪醇硼酸酯(1.7g,6.6mmol)和四三苯基膦铂(137mg,0.1mmol),抽换氩气3次,升温至85℃搅拌3小时。冷却至室温,加入化合物1c(2.0g,5.2mmol)、双三苯基磷二氯化钯(741mg,1.1mmol)、碳酸铯(3.6g,11.0mmol)和水(1mL),室温搅拌过夜。加入碘苯1e(1.2g,6.1mmol)和氢氧化钾(1.5g,27.6mmol),抽换氩气3次,升温至85℃搅拌2小时后冷却至室温停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物1f(3.0g),产率:88%。
MS m/z(ESI):667.2[M+1]。
第三步
(Z)-(1-(((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)-1-胺1g
将化合物1f(1.8g,2.7mmol)溶于二氯甲烷(15mL),加入三氟乙酸(3mL),室温搅拌反应5小时,停止反应。反应液减压浓缩,用饱和碳酸氢钠溶液(100mL)将反应液调至pH 8左右,无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题产物1g粗品(1.4g),产率:89%,产物不经纯化直接进行下一步反应。
第四步
(E)-1-吗啉基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮1i
将化合物1g(1.7g,2.8mmol)溶于N,N-二甲基甲酰胺(20mL),于室温加入二异丙基乙胺(1.1g,8.5mmol),然后加入(E)-4-溴-1-吗啉基丁-2-烯-1-酮1h(0.7g,2.8mmol,采用专利申请US2016347717中说明书第65页的实施例15公开的方法制备得到),搅拌反应2小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(50mL×2),合并有机相,用饱和氯化钠溶液洗涤(50mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物1i(1.3g),产率:65%。
MS m/z(ESI):720.2[M+1]。
第五步
(E)-1-吗啉基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮1
将化合物1i(2.0g,2.8mmol)溶于甲醇(5mL),加入盐酸(12N,10mL)搅拌反应3小时。停止并冷却反应,浓缩反应液,加入饱和碳酸氢钠溶液(15mL),用二氯甲烷萃取(50mL×4),合并有机相,依次用水洗(30mL×3),饱和氯化钠溶液洗涤(50mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物1(1.3g),产率:73%。
MS m/z(ESI):636.2[M+1]。
1H NMR(400MHz,CD 3OD)7.65(d,2H),7.49(d,1H),7.30-7.22(m,7H),6.82-6.76(m,1H),6.60-6.52(m,2H),4.15(s,2H),3.62-3.39(m,12H),0.76-0.64(m,4H)。
实施例2
(E)-1-(吡咯啉-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮2
Figure PCTCN2020096744-appb-000052
第一步
(E)-1-(吡咯啉-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮2b
采用实施例1的合成路线,将第四步原料1h替换为(E)-4-溴-1-(吡咯啉-1-基)丁-2-烯-1-酮2a(采用专利申请US2016347717中说明书第58页的实施例11公开的方法制备得到),制得标题化合物2b(51mg),产率69%。
MS m/z(ESI):704.3[M+1]。
第二步
(E)-1-(吡咯啉-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮2
采用实施例1的合成路线,将第五步原料1i替换为化合物2b,制得标题化合物2(20mg),产率65%。
MS m/z(ESI):620.3[M+1]。
1H NMR(400MHz,CD 3OD)7.65(d,2H),7.50(d,1H),7.31-7.21(m,7H),6.84-6.80(m,1H),6.59(d,1H),6.38(d,1H),4.16(s,2H),3.61-3.39(m,8H),0.92-0.60(m,4H),0.72-0.64(m,4H)。
实施例3
(E)-1-(哌啶-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮3
Figure PCTCN2020096744-appb-000053
第一步
(E)-1-(哌啶-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮3b
采用实施例1的合成路线,将第四步原料1h替换为(E)-4-溴-1-(哌啶-1-基)丁-2-烯-1-酮3a(采用专利申请US2016347717中说明书第58页的实施例12公开的方法制备得到),制得标题化合物3b(40mg),产率74%。
MS m/z(ESI):718.4[M+1]。
第二步
(E)-1-(哌啶-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮3
采用实施例1的合成路线,将第五步原料1i替换为化合物3b,制得标题化合物3(15mg),产率61%。
MS m/z(ESI):634.3[M+1]。
1H NMR(400MHz,CD 3OD)7.65(d,2H),7.50(d,1H),7.30-7.21(m,7H),6.74-6.70(m,1H),6.60-6.55(m,2H),5.36-5.35(m,1H),4.36(s,2H),3.56-3.53(m,5H),3.46-3.39(m,2H),2.05(s,1H),1.67-1.54(m,3H),0.92-0.68(m,6H)。
实施例4
(E)-1-(哌嗪-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮4
Figure PCTCN2020096744-appb-000054
第一步
4-((E)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯酰)哌嗪-1-羧酸叔丁酯4b
采用实施例1的合成路线,将第四步原料1h替换为(E)-4-(4-溴丁-2-烯酰基)哌嗪-1-羧酸叔丁酯4a(采用专利申请WO2014160200中说明书第129页的实施例32公开的方法制备得到),制得标题化合物4b(61mg),产率72%。
MS m/z(ESI):819.4[M+1]。
第二步
(E)-1-(哌嗪-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮4
采用实施例1的合成路线,将第五步原料1i替换为化合物4b,制得标题化合物4(20mg),产率41%。
MS m/z(ESI):635.3[M+1]。
1H NMR(400MHz,CD 3OD)7.65(d,2H),7.50(d,1H),7.30-7.20(m,7H),6.82-6.77(m,1H),6.59-6.53(m,2H),4.15(s,2H),3.69-3.67(m,4H),3.53-3.41(m,4H),2.96-2.94(m,4H),0.72-0.62(m,4H)。
实施例5
(E)-N,N-二甲基-4-(甲基(1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯酰胺5
Figure PCTCN2020096744-appb-000055
第一步
(1-(((5-碘吡啶-2-基)氧基)甲基)环丙基(甲基)氨基甲酸叔丁酯5a
将化合物1c(0.5g,1.3mmol)溶于N,N-二甲基甲酰胺(25mL),于室温加入氢化钠(0.1g,2.6mmol),加完后缓慢加入碘甲烷(0.3g,1.9mmol)。室温搅拌2小时后停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物5a(0.5g),产率:87%。
MS m/z(ESI):405.1[M+1]。
第二步
(Z)-甲基(1-(((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基甲酸叔丁酯5b
采用实施例1的合成路线,将第二步原料1c替换为化合物5a,制得标题化合物5b(89mg),产率85%。
MS m/z(ESI):681.3[M+1]。
第三步
(Z)-N-甲基-1-(((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)-1-胺5c
采用实施例1的合成路线,将第三步原料1f替换为化合物5b,制得标题化合物5c(60mg),产率70%。
第四步
(E)-N,N-二甲基-4-(甲基(1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯酰胺5e
采用实施例1的合成路线,将第四步原料1h替换为(E)-4-溴-N,N-二甲基丁-2-烯酰胺5d(采用专利申请US2016347717中说明书第39页的实施例1公开的方法制备得到),原料1g替换为5c制得标题化合物5e(60mg),产率50%。
第五步
(E)-N,N-二甲基-4-(甲基(1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯酰胺5
采用实施例1的合成路线,将第五步原料1i替换为化合物5e,制得标题化合物5(15mg),产率28%。
MS m/z(ESI):608.2[M+1]。
1H NMR(400MHz,CDCl 3)9.59(s,1H),7.65(d,2H),7.33(d,1H),7.28-7.23(m,6H),7.10(d,1H),6.79-6.75(m,1H),6.46(d,1H),6.35(d,1H),4.22(s,2H),3.48(d,2H),3.38-3.35(m,2H),3.05(d,6H),2.38(s,3H),0.73-0.63(m,4H)。
实施例6
(E)-4-(乙基(1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)-N,N-二甲基丁-2-烯酰胺6
Figure PCTCN2020096744-appb-000056
第一步
乙基(1-(((5-碘吡啶-2-基)氧基)甲基)环丙基氨基甲酸叔丁酯6a
采用实施例5的合成路线,将第一步试剂碘甲烷替换为碘乙烷,制得标题化合物6a(58mg),产率88%。
第二步
(Z)-乙基(1-(((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基甲酸叔丁酯6b
采用实施例1的合成路线,将第二步原料1c替换为化合物6a,制得标题化合物6b(85mg),产率80%。
第三步
(Z)-N-乙基-1-(((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)-1-胺6c
采用实施例1的合成路线,将第三步原料1f替换为化合物6b,制得标题化合物6c(65mg),产率76%。
第四步
(E)-4-(乙基(1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)-N,N-二甲基丁-2-烯酰胺6d
采用实施例5的合成路线,将第四步原料5c替换为化合物6c,制得标题化合物6d(70mg),产率59%。
MS m/z(ESI):706.3[M+1]。
第五步
(E)-4-(乙基(1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)-N,N-二甲基丁-2-烯酰胺6
采用实施例1的合成路线,将第五步原料1i替换为化合物6d,制得标题化合物6(15mg),产率28%。
MS m/z(ESI):622.3[M+1]。
1H NMR(400MHz,CDCl 3)9.74(s,1H),7.66(d,2H),7.31(d,1H),7.28-7.25(m,6H),7.21(d,1H),6.86-6.82(m,1H),6.46-6.36(m,2H),4.19(s,2H),3.53(d,2H),3.35-3.30(m,2H),3.06(d,6H),2.73-2.70(m,2H),1.05-1.02(m,3H),0.73-0.61(m,4H)。
实施例7
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环戊基)氨基)丁-2-烯酰胺7
Figure PCTCN2020096744-appb-000057
Figure PCTCN2020096744-appb-000058
第一步
1-(((5-碘吡啶-2-基)氧基)甲基)环戊基-1-胺7b
将叔丁醇钾(0.4g,3.5mmol)溶于四氢呋喃(10mL),于室温加入(1-氨基环戊基)甲醇7a(0.4g,3.5mmol,采用公知的方法“Journal of the American Chemical Society,2004,126(46),15195-15201”制备而得),加完后缓慢加入化合物1a(0.8g,3.5mmol)。室温搅拌2小时后停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物7b(0.7g),产率:66%。
MS m/z(ESI):319.1[M+1]。
第二步
(E)-4-((1-(((5-碘吡啶-2-基)氧基)甲基)环戊基)氨基)-N,N-二甲基丁-2-烯酰胺7c
将化合物7b(100mg,0.3mmol)溶于N,N-二甲基甲酰胺(10mL),于室温加入二异丙基乙胺(41mg,0.3mmol),然后加入化合物5d(60g,0.3mmol),搅拌反应2小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(50mL×2),合并有机相,饱和氯化钠溶液洗涤(50mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物7c(86mg),产率:67%。
第三步
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环戊基)氨基)丁-2-烯酰胺7d
将化合物1d(50mg,0.2mmol)溶于甲基四氢呋喃(20mL),加入双联频哪醇硼酸酯(47mg,0.2mmol),四三苯基膦铂(10mg,0.01mmol),抽换氩气3次,升温至85℃搅拌3小时。冷却至室温,加入化合物7c(66mg,0.2mmol)、双三苯基磷二氯化钯(11mg,0.02mmol)、碳酸铯(150mg,0.5mmol)和水(0.2mL),室温搅拌过夜。加入化合物1e(63mg,0.3mmol)和氢氧化钾(26mg,0.5mmol),抽换氩气3次,升 温至85℃搅拌2小时后冷却至室温停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物7d(84mg),产率:78%。
第四步
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环戊基)氨基)丁-2-烯酰胺7
将化合物7d(30mg,0.04mmol)溶于甲醇(1mL),加入盐酸(12N,2mL)搅拌反应3小时。停止并冷却反应,浓缩反应液,加入饱和碳酸氢钠溶液(15mL),用二氯甲烷萃取(10mL×4),合并有机相,依次用水洗(10mL×3),饱和氯化钠溶液洗涤(10mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物7(10mg),产率:41%。
MS m/z(ESI):622.3[M+1]。
1H NMR(400MHz,CDCl 3)7.61-7.45(m,3H),7.29-7.01(m,7H),6.54-6.42(m,3H),3.95(s,2H),3.42-3.39(m,1H),3.17-3.14(m,1H),2.91(s,1H),2.77(s,1H),2.62(s,3H),2.26(s,3H),1.63-1.52(m,4H),1.49-1.38(m,4H)。
实施例8
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环己基)氨基)丁-2-烯酰胺8
Figure PCTCN2020096744-appb-000059
第一步
1-(((5-碘吡啶-2-基)氧基)甲基)环己基-1-胺8b
采用实施例7的合成路线,将第一步原料7a替换为(1-氨基环己基)甲醇8a(采用公知的方法“Journal of Medicinal Chemistry,2004,47(18),4613-4626”制备而得), 制得标题化合物8b(650mg),产率68%。
MS m/z(ESI):333.1[M+1]。
第二步
(E)-4-((1-(((5-碘吡啶-2-基)氧基)甲基)环己基)氨基)-N,N-二甲基丁-2-烯酰胺8c
采用实施例7的合成路线,将第二步原料7b替换为化合物8b,制得标题化合物8c(500mg),产率58%。
MS m/z(ESI):444.2[M+1]。
第三步
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环己基)氨基)丁-2-烯酰胺8d
采用实施例7的合成路线,将第三步原料7c替换为化合物8c,制得标题化合物8d(250mg),产率94%。
MS m/z(ESI):720.4[M+1]。
第四步
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环己基)氨基)丁-2-烯酰胺8
采用实施例7的合成路线,将第四步原料7d替换为化合物8d,制得标题化合物8(80mg),产率36%。
MS m/z(ESI):636.2[M+1]。
1H NMR(400MHz,CD 3OD)7.72(d,1H),7.65(s,1H),7.49(dd,1H),7.33(dd,2H),7.31-7.24(m,5H),6.80(d,1H),6.68-6.65(m,2H),4.53(s,2H),3.85(d,2H),3.46-3.38(m,2H),3.08(s,3H),2.98(s,3H),2.06(d,2H),1.75-1.64(m,3H),1.64-1.47(m,4H),1.31(m,1H)。
实施例9
(E)-N,N-二甲基-4-(4-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺9
Figure PCTCN2020096744-appb-000060
Figure PCTCN2020096744-appb-000061
第一步
4-((5-碘吡啶-2-基)氧基)哌啶-1-羧酸叔丁酯9b
将氢化钠(0.6g,14.9mmol)溶于N,N-二甲基甲酰胺(30mL),于室温加入4-羟基哌啶-1-羧酸叔丁酯9a(1.0g,5.0mmol,采用公知的方法“Journal of the American Chemical Society,2018,140(1),155-158”制备而得),加完后缓慢加入化合物1a(2.2g,9.9mmol)。室温搅拌2小时后停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物9b(0.6g),产率:30%。
第二步
(Z)-4-((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-羧酸叔丁酯9c
将化合物1d(150mg,0.5mmol)溶于甲基四氢呋喃(15mL),加入双联频哪醇硼酸酯(140mg,0.6mmol)和四三苯基膦铂(28mg,0.02mmol),抽换氩气3次,升温至85℃搅拌3小时。冷却至室温,加入化合物9b(167mg,0.4mmol)、双三苯基磷二氯化钯(31mg,0.05mmol)、碳酸铯(300mg,0.9mmol)和水(1mL),室温搅拌过夜。加入化合物1e(187mg,0.9mmol)和氢氧化钾(129mg,2.3mmol),抽换氩气3次,升温至85℃搅拌2小时后冷却至室温停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物9c(210mg),产率:67%。
MS m/z(ESI):681.3[M+1]。
第三步
(Z)-3-氟-1-(四氢-2H-吡喃-2-基)-5-(4,4,4-三氟-2-苯基-1-(6-(哌啶-4-基氧基)吡啶-3-基)丁-1-烯-1-基)-1H-吲唑9d
将化合物9c(120mg,0.2mmol)溶于二氯甲烷(5mL),加入三氟乙酸(1mL),室温搅拌反应5小时,停止反应。反应液减压浓缩,用饱和碳酸氢钠溶液(10mL) 将反应液调至pH 8左右,无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题产物9d粗品(100mg),产率:98%,产物不经纯化直接进行下一步反应。
第四步
(E)-N,N-二甲基-4-(4-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺9e
将化合物9d(100mg,0.2mmol)溶于N,N-二甲基甲酰胺(5mL),于室温加入二异丙基乙胺(67mg,0.5mmol),然后加入化合物5d(31mg,0.2mmol)搅拌反应2小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物9e(85mg),产率:71%。
第五步
(E)-N,N-二甲基-4-(4-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)
吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺9
将化合物9e(80mg,0.1mmol)溶于甲醇(5mL),加入盐酸(12N,3mL)搅拌反应3小时。停止并冷却反应,浓缩反应液,加入饱和碳酸氢钠溶液(15mL),用二氯甲烷萃取(10mL×4),合并有机相,依次用水洗(10mL×3),饱和氯化钠溶液洗涤(10mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物9(25mg),产率:36%。
MS m/z(ESI):608.4[M+1]。
1H NMR(400MHz,CDCl 3)9.91(br,1H),7.68(d,2H),7.45(d,1H),7.33(s,1H),7.24-7.15(m,5H),6.82(s,2H),6.47(d,1H),5.31(s,1H),3.86(s,2H),3.49(s,2H),3.80-3.31(m,2H),3.15-3.12(s,3H),3.11(s,3H),3.05(s,3H),2.26-2.18(m,4H)。
实施例10
(Z)-1-(4-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-炔-1-酮10
Figure PCTCN2020096744-appb-000062
Figure PCTCN2020096744-appb-000063
第一步
(Z)-1-(4-((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-炔-1-酮10b
将化合物9d(60mg,0.1mmol)溶于N,N-二甲基甲酰胺(3mL),于室温加入三乙胺(52mg,0.5mmol)、丁二炔酸10a(17mg,0.2mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(30mg,0.2mmol)和1-羟基苯并三唑(24mg,0.2mmol)搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物10b(45mg),产率:67%。
第二步
(Z)-1-(4-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-炔-1-酮10
将化合物10b(45mg,0.07mmol)溶于甲醇(5mL),加入盐酸(12N,3mL)搅拌反应3小时。停止并冷却反应,浓缩反应液,加入饱和碳酸氢钠溶液(15mL),用二氯甲烷萃取(10mL×4),合并有机相,依次用水洗(10mL×3),饱和氯化钠溶液洗涤(10mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物10(15mg),产率:38%。
MS m/z(ESI):563.3[M+1]。
1H NMR(400MHz,CDCl 3)9.67(br,1H),7.65(d,2H),7.45(d,1H),7.34(m,1H),7.21-7.17(m,4H),6.50(d,1H),5.11(s,1H),3.94-3.92(m,1H),3.80-3.71(m,4H),3.37-3.29(m,2H),2.02(s,3H),1.94-1.75(m,4H)。
实施例11
(E)-N-甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺11
Figure PCTCN2020096744-appb-000064
第一步
(E)-4-溴-N-甲基丁-2-烯酰胺11b
将(E)-4-溴丁-2-烯酸11a(0.5g,3.0mmol)溶于二氯甲烷(5mL),于室温加入三乙胺(0.3g,3.3mmol)、甲胺(0.1g,3.3mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.6g,3.3mmol)、1-羟基苯并三唑(0.4g,3.3mmol)搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物11b(0.4g),产率:77%。
第二步
(R)-3-((5-碘吡啶-2-基)氧基)哌啶-1-羧酸叔丁酯11d
采用实施例1的合成路线,将第一步原料1b替换为(R)-3-羟基哌啶-1-羧酸叔丁酯11c(采用公知的方法“Tetrahedron,2011,67(7),1485-1500”制备而得),制得标题化合物11d(653mg),产率91%。
第三步
(3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-羧酸叔丁酯11e
采用实施例1的合成路线,将第二步原料1c替换为化合物11d,制得标题化合物11e(200mg),产率64%。
MS m/z(ESI):680.9[M+1]。
第四步
3-氟-1-(四氢-2H-吡喃-2-基)-5-((Z)-4,4,4-三氟-2-苯基-1-(6-(((R)-哌啶-3-基)氧基)吡啶-3-基)丁-1-烯-1-基)-1H-吲唑11f
采用实施例1的合成路线,将第三步原料1f替换为化合物11e,制得标题化合物11f(170mg),产率100%。
第五步
(E)-N-甲基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺11g
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为11b,制得标题化合物11g(50mg),产率86%。
第六步
(E)-N-甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺11
采用实施例1的合成路线,将第五步原料1i替换为化合物11g,制得标题化合物11(20mg),产率45%。
MS m/z(ESI):594.2[M+1]。
1H NMR(400MHz,CD 3OD)7.65(d,2H),7.49(d,1H),7.31-7.19(m,6H),6.68-6.62(m,2H),6.30(d,1H),5.39(s,1H),3.88(s,2H),3.67(s,1H),3.48-3.38(m,3H),3.26-3.13(m,2H),3.01(s,1H),2.80(s,3H),2.06(s,2H),1.86-1.73(m,2H)。
实施例12
(E)-1-吗啉基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮12
Figure PCTCN2020096744-appb-000065
Figure PCTCN2020096744-appb-000066
第一步
(E)-1-吗啉基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮12a
采用实施例1的合成路线,将第四步原料1g替换为化合物11f制得标题化合物12a(50mg),产率79%。
第二步
(E)-1-吗啉基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮12
采用实施例1的合成路线,将第五步原料1i替换为化合物12a,制得标题化合物12(20mg),产率42%。
MS m/z(ESI):650.3[M+1]。
1H NMR(400MHz,CDCl 3)10.10(s,1H),7.67-7.62(m,3H),7.50(d,1H),7.27-7.11(m,6H),6.79(s,2H),6.48(s,1H),5.38(s,1H),3.93(s,2H),3.70(s,7H),3.55(s,2H),3.38-3.31(m,3H),3.09-2.82(m,2H),1.85-1.65(m,4H)。
实施例13
(E)-N-(四氢-2H-吡喃-4-基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺13
Figure PCTCN2020096744-appb-000067
第一步
(E)-4-溴-N-(四氢-2H-吡喃-4-基)丁-2-烯酰胺13a
将(E)-4-溴丁-2-烯酸11a(1.0g,6.1mmol)溶于二氯甲烷(10mL),于室温加入三乙胺(1.8g,18.2mmol)、四氢吡喃-4-胺(0.6g,6.1mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(1.3g,6.7mmol)和1-羟基苯并三唑(0.9g,6.7mmol)搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物13a(1.2g),产率:79%。
第二步
(E)-N-(四氢-2H-吡喃-4-基)-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺13b
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为13a,制得标题化合物13b(41mg),产率81%。
第三步
(E)-N-(四氢-2H-吡喃-4-基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺13
采用实施例1的合成路线,将第五步原料1i替换为化合物13b,制得标题化合物13(20mg),产率32%。
MS m/z(ESI):664.3[M+1]。
1H NMR(400MHz,DMSO-d 6)12.68(s,1H),7.92-7.90(d,1H),7.60-7.59(m,1H),7.52-7.50(m,1H),7.23-7.14(m,6H),6.50-6.43(m,2H),5.97-5.93(d,1H),4.86-4.79(m,1H),3.79-3.74(m,3H),3.47-3.38(m,2H),3.33-3.32(m,1H),3.00-2.98(m,2H),2.80-2.78(m,1H),2.55-2.50(m,1H),2.00-1.24(m,12H)。
实施例14
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺14
Figure PCTCN2020096744-appb-000068
Figure PCTCN2020096744-appb-000069
第一步
(E)-N,N-二甲基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺14a
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为5d,制得标题化合物14a(80mg),产率79%。
第二步
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺14
采用实施例1的合成路线,将第五步原料1i替换为化合物14a,制得标题化合物14(10mg),产率27%。
MS m/z(ESI):608.3[M+1]。
1H NMR(400MHz,CDCl 3)9.77-7.75(br,1H),7.68-7.62(m,2H),7.46-7.43(m,1H),7.36-7.30(m,2H),7.27-7.18(d,5H),6.87-6.76(m,2H),6.80-6.48(m,1H),5.42-5.28(m,1H),3.97-3.67(m,5H),3.38-3.31(m,2H),3.12-3.07(m,6H),2.31-1.69(m,4H)。
实施例15
(E)-1-(吡咯啉-1-基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮15
Figure PCTCN2020096744-appb-000070
Figure PCTCN2020096744-appb-000071
第一步
(E)-1-(吡咯啉-1-基)-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮15a
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为2a,制得标题化合物15a(49mg),产率72%。
第二步
(E)-1-(吡咯啉-1-基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮15
采用实施例1的合成路线,将第五步原料1i替换为化合物15a,制得标题化合物15(20mg),产率47%。
MS m/z(ESI):634.3[M+1]。
1H NMR(400MHz,CD 3OD)7.63(d,2H),7.49(dd,1H),7.29-7.19(m,7H),6.79-6.74(m,1H),6.49-6.43(m,2H),5.02-4.98(m,1H),3.56-3.54(m,2H),3.47-3.44(m,4H),3.25(d,2H),2.92(d,1H),2.65-2.64(m,1H),2.37-2.35(m,2H),1.94-1.85(m,6H),1.64-1.52(m,2H)。
实施例16
(E)-N-(叔丁基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺16
Figure PCTCN2020096744-appb-000072
Figure PCTCN2020096744-appb-000073
第一步
(E)-4-溴-N-(叔丁基)丁-2-烯酰胺16a
将化合物11a(0.5g,3.0mmol)溶于二氯甲烷(10mL),于室温加入三乙胺(0.4g,3.6mmol)、叔丁胺(0.2g,3.0mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.7g,3.6mmol)和1-羟基苯并三唑(0.5g,3.6mmol)搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物16a(0.3g),产率:39%。
第二步
(E)-N-(叔丁基)-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺16b
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为16a,制得标题化合物16b(47mg),产率83%。
第三步
(E)-N-(叔丁基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺16
采用实施例1的合成路线,将第五步原料1i替换为化合物16b,制得标题化合物16(23mg),产率28%。
MS m/z(ESI):636.5[M+1]。
1H NMR(400MHz,CD 3OD)7.67(d,2H),7.50(dd,1H),7.32-7.22(m,7H),6.64-6.55(m,2H),6.33(d,1H),5.40(s,1H),3.88(s,2H),3.69(d,1H),3.52-3.41(m,3H),3.24(d,1H),3.02-2.99(m,1H),2.10-2.07(m,2H),1.87-1.75(m,2H),1.36(s,9H)。
实施例17
(E)-N-环戊基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺17
Figure PCTCN2020096744-appb-000074
第一步
(E)-4-溴-N-环戊基丁-2-烯酰胺17a
将化合物11a(0.5g,3.0mmol)溶于二氯甲烷(10mL),于室温加入三乙胺(0.4g,3.6mmol)、环戊胺(0.3g,3.0mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.7g,3.6mmol)和1-羟基苯并三唑(0.5g,3.6mmol),搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物17a(0.3g),产率:49%。
第二步
(E)-N-环戊基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺17b
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为17a,制得标题化合物17b(42mg),产率73%。
第三步
(E)-N-环戊基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺17
采用实施例1的合成路线,将第五步原料1i替换为化合物17b,制得标题化合物17(21mg),产率38%。
MS m/z(ESI):648.3[M+1]。
1H NMR(400MHz,DMSO-d 6)12.75-12.68(m,1H)7.88-7.86(d,1H),7.61-7.59(m,2H),7.53-7.50(m,1H),7.23-7.15(m,6H),6.47-6.41(m,2H),5.96-5.93(d,1H), 4.85-4.79(m,1H),4.02-3.97(m,1H),3.47-3.39(m,2H),2.99-2.98(m,2H),2.80-2.79(m,1H),2.56-2.50(m,2H),2.30-2.25(m,1H),1.99-1.27(m,12H)。
实施例18
(E)-1-(哌嗪-1-基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮18
Figure PCTCN2020096744-appb-000075
第一步
4-((E)-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰基)哌嗪-1-羧酸叔丁酯18a
采用实施例1的合成路线,将第四步原料1g替换为化合物11f,原料1h替换为化合物4a,制得标题化合物18a(51mg),产率76%。
第二步
(E)-1-(哌嗪-1-基)-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯-1-酮18
采用实施例1的合成路线,将第五步原料1i替换为化合物18a,制得标题化合物18(21mg),产率31%。
MS m/z(ESI):649.3[M+1]。
1H NMR(400MHz,CD 3OD)7.67(d,2H),7.50(d,1H),7.34-7.20(m,6H),6.92(d,1H),6.78-6.61(m,2H),5.36(s,1H),3.94-3.88(m,5H),3.50-3.39(m,6H),3.28(s,2H),2.23-1.88(m,5H),1.34-1.30(m,3H)。
实施例19
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-(4-氟苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺19
Figure PCTCN2020096744-appb-000076
第一步
(R)-5-碘-2-(哌啶-3-基氧基)吡啶19a
将化合物11d(1.0g,2.5mmol)溶于二氯甲烷(15mL),加入三氟乙酸(3mL),室温搅拌反应5小时,停止反应。反应液减压浓缩,用饱和碳酸氢钠溶液(100mL)将反应液调至pH 8左右,无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题产物19a粗品(0.8g),产率:100%,产物不经纯化直接进行下一步反应。
第二步
(R,E)-4-(3-((5-碘吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺19b
将化合物19a(752mg,2.5mmol)溶于N,N-二甲基甲酰胺(20mL),于室温加入二异丙基乙胺(1.6g,12.4mmol),然后加入化合物5d(470mg,2.5mmol)搅拌反应2小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(50mL×2),合并有机相,饱和氯化钠溶液洗涤(50mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物19b(0.7g),产率:68%。
MS m/z(ESI):416.1[M+1]。
第三步
(E)-N,N-二甲基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-(4-氟苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺19d
将化合物1d(50mg,0.2mmol)溶于甲基四氢呋喃(10mL),加入双联频哪醇硼酸酯(47mg,0.2mmol)和四三苯基膦铂(4mg,0.003mmol),抽换氩气3次,升温至85℃搅拌3小时。冷却至室温,加入化合物19b(57mg,0.1mmol)、双三苯基磷二 氯化钯(20mg,0.03mmol)、碳酸铯(128mg,0.4mmol)和水(0.5mL),室温搅拌过夜。加入4-氟碘苯19c(68mg,0.3mmol)和氢氧化钾(43mg,0.8mmol),抽换氩气3次,升温至85℃搅拌2小时后冷却至室温停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物19d(50mg),产率:46%。
MS m/z(ESI):710.3[M+1]。
第四步
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-(4-氟苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺19
将化合物19d(50mg,0.07mmol)溶于甲醇(2mL),加入盐酸(12N,1mL)搅拌反应3小时。停止并冷却反应,浓缩反应液,加入饱和碳酸氢钠溶液(15mL),用二氯甲烷萃取(10mL×4),合并有机相,依次用水洗(10mL×3),饱和氯化钠溶液洗涤(10mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物19(15mg),产率:34%。
MS m/z(ESI):626.2[M+1]。
1H NMR(400MHz,CD 3OD)7.70(s,1H),7.65(s,1H),7.52(dd,1H),7.32-7.26(m,4H),7.02-6.97(t,2H),6.91-6.87(m,1H),6.65-6.61(d,2H),5.44(s,1H),3.94(s,3H),3.50-3.38(m,4H),3.14(s,3H),3.12-3.05(m,1H),3.01(s,3H),2.11-2.09(m,2H),1.89-1.77(m,2H)。
实施例20
(E)-4-((R)-3-((5-((Z)-2-(4-乙酰氨基苯基)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺20
Figure PCTCN2020096744-appb-000077
第一步
(E)-4-((3R)-3-((5-((Z)-2-(4-乙酰氨基苯基)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺20b
采用实施例19的合成路线,将第三步原料19c替换为N-(4-碘苯基)乙酰胺20a(采用公知的方法“Journal of Organic Chemistry,2018,83(2),930-938”制备而得),制得标题化合物20b(70mg),产率61%。
第二步
(E)-4-((R)-3-((5-((Z)-2-(4-乙酰氨基苯基)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺20
采用实施例19的合成路线,将第四步原料19d替换为化合物20b,制得标题化合物20(22mg),产率41%。
MS m/z(ESI):665.2[M+1]。
1H NMR(400MHz,CD 3OD)7.67(d,2H),7.48-7.44(m,3H),7.28(dd,2H),7.18(d,2H),6.82(d,1H),6.67-6.59(m,2H),5.28(s,1H),3.76(s,2H),3.49-3.39(m,4H),3.25(d,1H),3.10(s,3H),3.03(d,1H),2.99(s,3H),2.11(s,3H),1.99-1.94(m,2H),1.86-1.74(m,2H)。
实施例21
4-((Z)-1-(6-(((R)-1-((E)-4-(二甲氨基)-4-氧代丁-2-烯-1-基)哌啶-3-基)氧基)吡啶-3-基)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)丁-1-烯-2-基)-N-甲基苯甲酰胺21
Figure PCTCN2020096744-appb-000078
第一步
4-((Z)-1-(6-(((R)-1-((E)-4-(二甲氨基)-4-氧代丁-2-烯-1-基)哌啶-3-基)氧基)吡啶-3-基)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)丁-1-烯-2-基)-N-甲基苯甲酰胺21b
采用实施例19的合成路线,将第三步原料19c替换为4-碘-N-甲基苯甲酰胺21a(采用公知的方法“Journal of the American Chemical Society,2013,135(12),4628-4631”制备而得),制得标题化合物21b(50mg),产率44%。
第二步
4-((Z)-1-(6-(((R)-1-((E)-4-(二甲氨基)-4-氧代丁-2-烯-1-基)哌啶-3-基)氧基)吡啶-3-基)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)丁-1-烯-2-基)-N-甲基苯甲酰胺21
采用实施例19的合成路线,将第四步原料19d替换为化合物21b,制得标题化合物21(10mg),产率32%。
MS m/z(ESI):665.3[M+1]。
1H NMR(400MHz,CD 3OD)7.71-7.69(m,4H),7.50(d,1H),7.36-7.29(m,4H),6.88(d,1H),6.65-6.58(m,2H),5.39(s,1H),3.97-3.91(m,2H),3.69(d,1H),3.49-3.44(m,3H),3.06(s,3H),3.05-3.00(m,2H),2.99(s,3H),2.89(s,3H),2.10-2.03(m,2H),1.86-1.74(m,2H)。
实施例22
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-(4-(三氟甲基)苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺22
Figure PCTCN2020096744-appb-000079
第一步
(E)-N,N-二甲基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-(4-(三氟甲基)苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺22b
采用实施例19的合成路线,将第三步原料19c替换为1-碘-4-(三氟甲基)苯22a,制得标题化合物22b(80mg),产率69%。
第二步
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-(4-(三氟甲基)苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺22
采用实施例19的合成路线,将第四步原料19d替换为化合物22b,制得标题化合物22(20mg),产率31%。
MS m/z(ESI):676.2[M+1]。
1H NMR(400MHz,CD 3OD)7.70(dd,2H),7.57-7.55(m,3H),7.47(d,2H),7.32(dd,2H),6.88(d,1H),6.65-6.59(m,2H),5.37(s,1H),3.91(d,2H),3.50(q,2H),3.32(s,4H),3.12(s,3H),3.00(s,3H),2.09-2.04(m,2H),1.84(d,2H)。
实施例23
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-(4-(甲基磺酰基)苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺23
Figure PCTCN2020096744-appb-000080
第一步
(E)-N,N-二甲基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-(4-(甲基磺酰基)苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺23b
采用实施例19的合成路线,将第三步原料19c替换为1-碘-4-(甲磺酰基)苯23a(采用公知的方法“Organic Letters,2014,16(9),2306-2309”制备而得),制得标题化合物23b(60mg),产率51%。
第二步
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-(4-(甲基磺酰基)苯基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺23
采用实施例19的合成路线,将第四步原料19d替换为化合物23b,制得标题化 合物23(10mg),产率28%。
MS m/z(ESI):686.2[M+1]。
1H NMR(400MHz,CD 3OD)7.83(d,2H),7.67(s,2H),7.53-7.50(m,3H),7.30(d,2H),6.89(dd,1H),6.66-6.59(m,2H),5.38(s,1H),3.97-3.91(m,3H),3.70-3.65(m,1H),3.50(q,3H),3.28(d,1H),3.11(s,6H),2.99(s,3H),2.10-2.01(m,2H),1.94-1.86(m,2H)。
实施例24
(E)-4-((R)-3-((5-((Z)-2-(4-氰基苯基)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺24
Figure PCTCN2020096744-appb-000081
第一步
(E)-4-((3R)-3-((5-((Z)-2-(4-氰基苯基)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺24b
采用实施例19的合成路线,将第三步原料19c替换为4-碘苯甲腈24a,制得标题化合物24b(80mg),产率73%。
第二步
(E)-4-((R)-3-((5-((Z)-2-(4-氰基苯基)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)-N,N-二甲基丁-2-烯酰胺24
采用实施例19的合成路线,将第四步原料19d替换为化合物24b,制得标题化合物24(20mg),产率38%。
MS m/z(ESI):633.2[M+1]。
1H NMR(400MHz,CD 3OD)7.67-7.61(m,4H),7.47(d,1H),7.45(d,2H),7.33(d,2H),6.89(d,1H),6.66-6.61(m,2H),5.44(s,1H),3.94-3.91(m,2H),3.71(d,1H), 3.52-3.45(m,3H),3.25(d,1H),3.12(s,3H),3.05(d,1H),3.01(s,3H),2.11-2.04(m,2H),1.90-1.77(m,2H)。
实施例25
(R,Z)-1-(3-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-炔-1-酮25
Figure PCTCN2020096744-appb-000082
第一步
1-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-炔-1-酮25a
采用实施例10的合成路线,将第一步原料9d替换为化合物11f,制得标题化合物25a(37mg),产率83%。
第二步
(R,Z)-1-(3-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-炔-1-酮25
采用实施例10的合成路线,将第二步原料10b替换为化合物25a,制得标题化合物25(4mg),产率15%。
MS m/z(ESI):563.2[M+1]。
1H NMR(400MHz,CDCl 3)9.38(s,1H),7.68-7.65(m,2H),7.43-7.12(m,8H),6.46-6.39(m,1H),5.02(d,1H),3.95-3.77(m,2H),3.33-3.00(m,4H),2.13-1.93(m,4H),1.69(d,3H)。
实施例26
(R,Z)-1-(3-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丙-2-烯-1-酮26
Figure PCTCN2020096744-appb-000083
第一步
1-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丙-2-烯-1-酮26b
采用实施例10的合成路线,将第一步原料9d替换为化合物11f,原料10a替换为丙烯酸26a,制得标题化合物26b(30mg),产率73%。
第二步
(R,Z)-1-(3-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丙-2-烯-1-酮26
采用实施例10的合成路线,将第二步原料10b替换为化合物26b,制得标题化合物26(3mg),产率12%。
MS m/z(ESI):551.2[M+1]。
1H NMR(400MHz,CDCl 3)9.31(s,1H),7.68(d,2H),7.43(d,2H),7.29-7.27(m,5H),7.20-7.09(m,1H),6.40(d,1H),6.39-6.20(m,1H),5.49-5.37(m,2H),4.95(s,1H),3.72(s,1H),3.68(d,2H),3.39(d,1H),3.36-3.31(m,2H),2.04-1.90(m,4H)。
实施例27
(E)-N,N-二甲基-4-((S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺27
Figure PCTCN2020096744-appb-000084
Figure PCTCN2020096744-appb-000085
第一步
(S)-3-((5-碘吡啶-2-基)氧基)哌啶-1-羧酸叔丁酯27b
采用实施例1的合成路线,将第一步原料1b替换为(S)-3-羟基哌啶-1-羧酸叔丁酯27a(采用公知的方法“Tetrahedron,2007,63(2),331-336”制备而得),制得标题化合物27b(531mg),产率92%。
第二步
(3S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-羧酸叔丁酯27c
采用实施例1的合成路线,将第二步原料1c替换为化合物27b,制得标题化合物27c(190mg),产率76%。
MS m/z(ESI):680.9[M+1]。
第三步
3-氟-1-(四氢-2H-吡喃-2-基)-5-((Z)-4,4,4-三氟-2-苯基-1-(6-(((S)-哌啶-3-基)氧基)吡啶-3-基)丁-1-烯-1-基)-1H-吲唑27d
采用实施例1的合成路线,将第三步原料1f替换为化合物27c,制得标题化合物27d(85mg),产率100%。
第四步
(E)-N,N-二甲基-4-((3S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺27e
采用实施例1的合成路线,将第四步原料1g替换为化合物27d,原料1h替换为化合物5d,制得标题化合物27e(65mg),产率64%。
第五步
(E)-N,N-二甲基-4-((S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丁-2-烯酰胺27
采用实施例1的合成路线,将第五步原料1i替换为化合物27e,制得标题化合物27(15mg),产率43%。
MS m/z(ESI):607.9[M+1]。
1H NMR(400MHz,CDCl 3)9.36(s,1H),7.69-7.67(m,1H),7.64-7.63(d,1H),7.45-7.43(m,1H),7.35-7.32(d,1H),7.28-7.19(m,4H),7.08-7.06(d,1H),6.89-6.82(m,1H),6.46-6.41(m,1H),5.39-5.36(m,1H),5.08-5.02(br,1H),3.38-3.30(m,2H),3.22-3.16(m,2H),3.05(s,3H),3.10(s,3H),2.92-2.86(m,1H),2.67-2.61(m,1H),2.34-2.24(m,2H),2.07-1.77(m,4H),1.53-1.48(m,1H)。
实施例28
(Z)-1-(4-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丙-2-烯-1-酮28
Figure PCTCN2020096744-appb-000086
第一步
(Z)-1-(4-((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丙-2-烯-1-酮28a
采用实施例10的合成路线,将第一步原料10a替换为化合物26a,制得标题化合物28a(50mg),产率63%。
第二步
(Z)-1-(4-((5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)哌啶-1-基)丙-2-烯-1-酮28
采用实施例10的合成路线,将第二步原料10b替换为化合物28a,制得标题化合物28(10mg),产率23%。
MS m/z(ESI):551.2[M+1]。
1H NMR(400MHz,CDCl 3)9.47(br,1H),7.60(d,2H),7.35(d,1H),7.28(d,1H),7.26-7.19(m,5H),6.59(dd,1H),6.49(d,1H),6.29(d,1H),5.76(d,1H),5.14(m,1H),3.89-3.53(m,5H),3.34(t,2H),1.99-1.94(m,2H),1.77-1.66(m,2H)。
实施例29
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-基)丁-2-烯酰胺29
Figure PCTCN2020096744-appb-000087
第一步
(R)-3-((5-碘吡啶-2-基)氧基)吡咯啉-1-羧酸叔丁酯29b
采用实施例1的合成路线,将第一步原料1b替换为(R)-3-羟基吡咯啉-1-羧酸叔丁酯29a,制得标题化合物29b(0.9g),产率86%。
第二步
(3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-羧酸叔丁酯29c
采用实施例1的合成路线,将第二步原料1c替换为化合物29b,制得标题化合物29c(260mg),产率37%。
第三步
3-氟-1-(四氢-2H-吡喃-2-基)-5-((Z)-4,4,4-三氟-2-苯基-1-(6-(((R)-吡咯啉-3-基)氧基)吡啶-3-基)丁-1-烯-1-基)-1H-吲唑29d
采用实施例1的合成路线,将第三步原料1f替换为化合物29c,制得标题化合物29d(93mg),产率99%。
第四步
(E)-N,N-二甲基-4-((3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-基)丁-2-烯酰胺29e
采用实施例1的合成路线,将第四步原料1g替换为化合物29d,原料1h替换为化合物5d,制得标题化合物29e(60mg),产率54%。
MS m/z(ESI):678.3[M+1]。
第五步
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-基)丁-2-烯酰胺29
采用实施例1的合成路线,将第五步原料1i替换为化合物29e,制得标题化合物29(15mg),产率29%。
MS m/z(ESI):594.3[M+1]。
1H NMR(400MHz,CDCl 3)12.72(s,1H),7.70-7.65(m,2H),7.60-7.50(d,1H),7.25-7.20(m,5H),7.19-7.15(m,2H),6.55-6.45(m,3H),5.17-5.10(m,1H),3.50-3.35(m,2H),3.15-3.10(m,2H),2.95(s,3H),2.83(s,3H),2.70-2.60(m,2H),2.50-2.45(m,1H),2.35-2.25(m,1H),2.15-2.05(m,1H),1.70-1.60(m,1H)。
实施例30
(E)-N,N-二甲基-4-((S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-基)丁-2-烯酰胺30
Figure PCTCN2020096744-appb-000088
第一步
(S)-3-((5-碘吡啶-2-基)氧基)吡咯啉-1-羧酸叔丁酯30b
采用实施例1的合成路线,将第一步原料1b替换为(S)-3-羟基吡咯啉-1-羧酸叔丁酯30a,制得标题化合物30b(0.5g),产率50%。
第二步
(3S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-羧酸叔丁酯30c
采用实施例1的合成路线,将第二步原料1c替换为化合物30b,制得标题化合物30c(260mg),产率85%。
第三步
3-氟-1-(四氢-2H-吡喃-2-基)-5-((Z)-4,4,4-三氟-2-苯基-1-(6-(((S)-吡咯啉-3-基)氧基)吡啶-3-基)丁-1-烯-1-基)-1H-吲唑30d
采用实施例1的合成路线,将第三步原料1f替换为化合物30c,制得标题化合物30d(100mg),产率98%。
第四步
(E)-N,N-二甲基-4-((3S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-基)丁-2-烯酰胺30e
采用实施例1的合成路线,将第四步原料1g替换为化合物30d,原料1h替换为化合物5d,制得标题化合物30e(70mg),产率62%。
MS m/z(ESI):678.3[M+1]。
第五步
(E)-N,N-二甲基-4-((S)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)吡咯啉-1-基)丁-2-烯酰胺30
采用实施例1的合成路线,将第五步原料1i替换为化合物30e,制得标题化合物30(10mg),产率23%。
MS m/z(ESI):593.9[M+1]。
1H NMR(400MHz,CDCl 3)9.58(s,1H),7.67-7.65(m,2H),7.46(d,1H),7.34-7.30(m,2H),7.27-7.16(m,4H),6.87-6.77(m,2H),6.59(d,1H),5.53(s,1H),4.14(s,1H),3.93(s,3H),3.39-3.31(m,2H),3.29-3.03(m,7H),2.41-2.28(m,4H)。
实施例31
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-基)丁-2-烯酰胺31
Figure PCTCN2020096744-appb-000089
Figure PCTCN2020096744-appb-000090
第一步
(R)-3-((5-碘吡啶-2-基)氨基)哌啶-1-羧酸叔丁酯31b
采用实施例1的合成路线,将第一步原料1b替换为(R)-3-氨基哌啶-1-羧酸叔丁酯31a,制得标题化合物31b(0.8g),产率57%。
第二步
(3R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-羧酸叔丁酯31c
采用实施例1的合成路线,将第二步原料1c替换为化合物31b,制得标题化合物31c(210mg),产率75%。
第三步
(R,Z)-N-(哌啶-3-基)-5-(4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-胺31d
采用实施例1的合成路线,将第三步原料1f替换为化合物31c,制得标题化合物31d(42mg),产率92%。
第四步
(E)-N,N-二甲基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-基)丁-2-烯酰胺31
采用实施例1的合成路线,将第四步原料1g替换为化合物31d,原料1h替换为化合物5d,制得标题化合物31(20mg),产率39%。
MS m/z(ESI):607.3[M+1]。
1H NMR(400MHz,CDCl 3)9.26(s,1H),7.68(s,1H),7.55-7.54(m,1H),7.44-7.41(m,1H),7.36-7.31(m,1H),7.28-7.23(m,4H),6.92-6.80(m,2H),6.45-6.41(m,1H),6.15-6.13(m,1H),5.39-5.37(m,1H),3.35-3.28(m,2H),3.17-3.15(m,2H),3.07(s,3H),3.02(s,3H),2.70-2.68(m,1H),2.47-2.45(m,1H),2.47-2.45(m,1H),2.06-2.04(m,2H),1.34-1.29(m,4H)。
实施例32
(E)-1-吗啉基-4-((R)-3-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-基)丁-2-烯-1-酮32
Figure PCTCN2020096744-appb-000091
采用实施例31的合成路线,将第四步原料5d替换为化合物1h,制得标题化合物32(8mg),产率19%。
MS m/z(ESI):649.2[M+1]。
1H NMR(400MHz,CDCl 3)9.25(s,1H),7.68(s,1H),7.56-7.52(m,1H),7.45-7.40(m,1H),7.36-7.32(m,1H),7.28-7.21(m,4H),6.94-6.82(m,2H),6.44-6.37(m,1H),6.15-6.07(m,1H),5.39-5.35(m,1H),3.76-3.46(m,8H),3.36-3.26(m,2H),3.17-3.05(m,2H),2.99-2.92(m,1H),2.65-2.03(m,7H),0.95-0.87(m,1H)。
实施例33
(E)-N,N-二甲基-4-(4-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-基)丁-2-烯酰胺33
Figure PCTCN2020096744-appb-000092
第一步
4-((5-碘吡啶-2-基)氨基)哌啶-1-羧酸叔丁酯33b
采用实施例1的合成路线,将第一步原料1b替换为4-氨基哌啶-1-羧酸叔丁酯 33a,制得标题化合物33b(0.2g),产率20%。
第二步
(Z)-4-((5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-羧酸叔丁酯33c
采用实施例1的合成路线,将第二步原料1c替换为化合物33b,制得标题化合物33c(80mg),产率64%。
MS m/z(ESI):680.3[M+1]。
第三步
(Z)-N-(哌啶-4-基)-5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-胺33d
采用实施例1的合成路线,将第三步原料1f替换为化合物33c,制得标题化合物33d(68mg),产率100%。
第四步
(E)-N,N-二甲基-4-(4-((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-基)丁-2-烯酰胺33e
采用实施例1的合成路线,将第四步原料1g替换为化合物33d,原料1h替换为化合物5d,制得标题化合物33e(70mg),产率86%。
第五步
(E)-N,N-二甲基-4-(4-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氨基)哌啶-1-基)丁-2-烯酰胺33
采用实施例1的合成路线,将第五步原料1i替换为化合物33e,制得标题化合物33(15mg),产率24%。
MS m/z(ESI):607.7[M+1]。
1H NMR(400MHz,CDCl 3)7.63(s,1H),7.59(s,1H),7.38-7.32(m,4H),7.24(d,2H),7.23-7.02(m,2H),6.77(s,2H),6.47(d,1H),5.36(s,1H),3.93(s,1H),3.83(s,2H),3.39-3.29(m,6H),3.12(s,3H),3.09(s,3H),2.42(m,2H),2.07(s,2H)。
实施例34
(E)-N,N-二甲基-4-(((R)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基)丁-2-烯酰胺34
Figure PCTCN2020096744-appb-000093
Figure PCTCN2020096744-appb-000094
第一步
(R)-(1-(5-碘吡啶-2-基)吡咯啉-3-基)氨基甲酸叔丁酯34b
采用实施例1的合成路线,将第一步原料1b替换为(R)-吡咯啉-3-基氨基甲酸叔丁酯34a,制得标题化合物34b(0.8g),产率70%。
第二步
((3R)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基甲酸叔丁酯34c
采用实施例1的合成路线,将第二步原料1c替换为化合物34b,制得标题化合物34c(85mg),产率83%。
MS m/z(ESI):665.9[M+1]。
第三步
(3R)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-胺34d
采用实施例1的合成路线,将第三步原料1f替换为化合物34c,制得标题化合物34d(60mg),产率82%。
第四步
(E)-N,N-二甲基-4-(((3R)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基)丁-2-烯酰胺34e
采用实施例1的合成路线,将第四步原料1g替换为化合物34d,原料1h替换为化合物5d,制得标题化合物34e(45mg),产率63%。
第五步
(E)-N,N-二甲基-4-(((R)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基)丁-2-烯酰胺34
采用实施例1的合成路线,将第五步原料1i替换为化合物34e,制得标题化合物 34(9mg),产率23%。
MS m/z(ESI):592.9[M+1]。
1H NMR(400MHz,CDCl 3)9.53(s,1H),7.67(s,2H),7.38(d,1H),7.30-7.22(m,6H),6.95-6.87(m,2H),6.45(d,1H),6.07(d,1H),3.63-3.59(m,1H),3.54-3.48(m,3H),3.41-3.28(m,3H),3.23-3.19(m,1H),3.06(d,6H),2.27-2.14(m,2H),1.89-1.81(m,2H)。
实施例35
(E)-N,N-二甲基-4-(((S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基)丁-2-烯酰胺35
Figure PCTCN2020096744-appb-000095
第一步
(S)-(1-(5-碘吡啶-2-基)吡咯啉-3-基)氨基甲酸叔丁酯35b
采用实施例1的合成路线,将第一步原料1b替换为(S)-吡咯啉-3-基氨基甲酸叔丁酯35a,制得标题化合物35b(0.8g),产率69%。
第二步
((3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基甲酸叔丁酯35c
采用实施例1的合成路线,将第二步原料1c替换为化合物35b,制得标题化合物35c(86mg),产率84%。
MS m/z(ESI):666.3[M+1]。
第三步
(3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-胺35d
采用实施例1的合成路线,将第三步原料1f替换为化合物35c,制得标题化合物35d(55mg),产率80%。
第四步
(E)-N,N-二甲基-4-(((3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基)丁-2-烯酰胺35e
采用实施例1的合成路线,将第四步原料1g替换为化合物35d,原料1h替换为化合物5d,制得标题化合物35e(40mg),产率61%。
第五步
(E)-N,N-二甲基-4-(((S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)吡咯啉-3-基)氨基)丁-2-烯酰胺35
采用实施例1的合成路线,将第五步原料1i替换为化合物35e,制得标题化合物35(8mg),产率23%。
MS m/z(ESI):592.9[M+1]。
1H NMR(400MHz,CDCl 3)9.47(s,1H),7.67(s,2H),7.39(dd,1H),7.34(dd,1H),7.28-7.22(m,5H),6.96-6.87(m,2H),6.45(dd,1H),6.07(dd,1H),3.63-3.59(m,1H),3.54-3.46(m,3H),3.41-3.28(m,3H),3.22-3.19(m,1H),3.07(d,6H),2.20-2.15(m,2H),1.88-1.81(m,2H)。
实施例36
(E)-N,N-二甲基-4-((1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-4-基)氨基)丁-2-烯酰胺36
Figure PCTCN2020096744-appb-000096
Figure PCTCN2020096744-appb-000097
第一步
(1-(5-碘吡啶-2-基)哌啶-4-基)氨基甲酸叔丁酯36b
采用实施例1的合成路线,将第一步原料1b替换为哌啶-4-基氨基甲酸叔丁酯36a,制得标题化合物36b(0.3g),产率28%。
第二步
(Z)-(1-(5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-4-基)氨基甲酸叔丁酯36c
采用实施例1的合成路线,将第二步原料1c替换为化合物36b,制得标题化合物36c(120mg),产率74%。
MS m/z(ESI):679.9[M+1]。
第三步
(Z)-1-(5-(4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-4-胺36d
采用实施例1的合成路线,将第三步原料1f替换为化合物36c,制得标题化合物36d(68mg),产率100%。
第四步
(E)-N,N-二甲基-4-((1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-4-基)氨基)丁-2-烯酰胺36e
采用实施例1的合成路线,将第四步原料1g替换为化合物36d,原料1h替换为化合物5d,制得标题化合物36e(70mg),产率86%。
第五步
(E)-N,N-二甲基-4-((1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-4-基)氨基)丁-2-烯酰胺36
采用实施例1的合成路线,将第五步原料1i替换为化合物36e,制得标题化合物36(15mg),产率24%。
MS m/z(ESI):607.7[M+1]。
1H NMR(400MHz,CD 3OD)7.70(s,1H),7.56-7.53(m,1H),7.48-7.42(m,1H),7.35-7.32(m,1H),7.30-7.26(m,6H),7.00-6.98(d,1H),6.92-6.89(m,1H),6.72-6.64(m,1H),4.25-4.21(d,2H),3.92-3.90(d,2H),3.57-3.39(m,4H),3.17-3.11(m,4H),3.01(s,3H),2.24-2.22(m,2H),1.67-1.67(m,2H)。
实施例37
(E)-N,N-二甲基-4-(甲基((S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基)丁-2-烯酰胺37
Figure PCTCN2020096744-appb-000098
第一步
(S)-(1-(5-碘吡啶-2-基)哌啶-3-基)氨基甲酸叔丁酯37b
将碳酸钾(1.5g,20.5mmol)溶于N,N-二甲基甲酰胺(10mL),于室温加入(S)-哌啶-3-基氨基甲酸叔丁酯37a(2.0g,10.0mmol),加完后缓慢加入化合物1a(2.2g,10.0mmol)。加热至100℃搅拌4小时后停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物37b(3.5g),产率:87%。
第二步
(S)-(1-(5-碘吡啶-2-基)哌啶-3-基)(甲基)氨基甲酸叔丁酯37c
将氢化钠(12mg,0.5mmol)溶于N,N-二甲基甲酰胺(10mL),于室温加入化合物37b(100mg,0.2mmol),加完后缓慢加入碘甲烷(28mg,0.2mmol)。室温搅拌2 小时后停止反应。反应液减压浓缩,残余物用薄层色谱法以展开剂体系B纯化所得残余物,得到标题产物37c(95mg),产率:92%。
MS m/z(ESI):418.1[M+1]。
第三步
甲基((3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基甲酸叔丁酯37d
采用实施例1的合成路线,将第二步原料1c替换为化合物37c,制得标题化合物37d(150mg),产率71%。
MS m/z(ESI):694.4[M+1]。
第四步
(3S)-N-甲基-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-胺37e
采用实施例1的合成路线,将第三步原料1f替换为化合物37d,制得标题化合物37e(80mg),产率93%。
第五步
(E)-N,N-二甲基-4-(甲基((3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基)丁-2-烯酰胺37f
采用实施例1的合成路线,将第四步原料1g替换为化合物37e,原料1h替换为化合物5d,制得标题化合物37f(75mg),产率79%。
第六步
(E)-N,N-二甲基-4-(甲基((S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基)丁-2-烯酰胺37
采用实施例1的合成路线,将第五步原料1i替换为化合物37f,制得标题化合物37(22mg),产率25%。
MS m/z(ESI):621.2[M+1]。
1H NMR(400MHz,CDCl 3)10.51(s,1H),7.71(s,1H),7.62(s,1H),7.74-7.31(m,4H),7.29-7.21(m,4H),6.80(s,2H),6.65(dd,1H),4.71(dd,1H),4.04(d,2H),3.87(d,2H),3.41(d,2H),3.35-3.31(m,2H),3.06(d,6H),2.84(s,3H),2.29(d,1H),2.02-1.92(m,2H),1.69(d,1H)。
实施例38
(E)-1-吗啉基-4-(((S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基)丁-2-烯-1-酮38
Figure PCTCN2020096744-appb-000099
Figure PCTCN2020096744-appb-000100
第一步
((3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基甲酸叔丁酯38a
采用实施例1的合成路线,将第二步原料1c替换为化合物37b,制得标题化合物38a(100mg),产率80%。
MS m/z(ESI):680.4[M+1]。
第二步
(3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-胺38b
采用实施例1的合成路线,将第三步原料1f替换为化合物38a,制得标题化合物38b(35mg),产率82%。
第三步
(E)-1-吗啉基-4-(((3S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基)丁-2-烯-1-酮38c
采用实施例1的合成路线,将第四步原料1g替换为化合物38b,制得标题化合物38c(75mg),产率79%。
第四步
(E)-1-吗啉基-4-(((S)-1-(5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)哌啶-3-基)氨基)丁-2-烯-1-酮38
采用实施例1的合成路线,将第五步原料1i替换为化合物38c,制得标题化合物38(9mg),产率25%。
MS m/z(ESI):649.3[M+1]。
1H NMR(400MHz,CDCl 3)9.92(s,1H),7.65(s,2H),7.37(d,1H),7.30-7.22(m,6H),6.95-6.88(m,2H),6.40(d,2H),4.15(d,1H),3.76-3.49(m,7H),3.53-3.49(m,4H),3.32-3.29(m,2H),3.00-2.95(m,1H),2.86-2.81(m,1H),2.73-2.69(m,1H),1.99-1.94(m,1H),1.78-1.72(m,1H),1.58-1.48(m,1H),1.33-1.27(m,2H)。
实施例39
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯酰胺39
Figure PCTCN2020096744-appb-000101
第一步
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯酰胺39a
采用实施例1的合成路线,将第四步原料1h替换为5d,制得标题化合物39a(53mg),产率:65%。
第二步
(E)-N,N-二甲基-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-酰胺39
采用实施例1的合成路线,将第五步原料1i替换为化合物39a,制得标题化合物39(24mg),产率:62%。
MS m/z(ESI):593.9[M+1]。
1H NMR(400MHz,CD 3OD)7.63(d,2H),7.41-7.08(m,8H),6.83-6.76(m,1H),6.48-6.36(m,2H),4.12(s,2H),3.49-3.48(m,2H),3.36-3.31(m,2H),3.09(s,3H),3.03(s,3H),0.67-0.55(m,4H)。
实施例40
(E)-1-(3-羟基氮杂环丁-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮40
Figure PCTCN2020096744-appb-000102
Figure PCTCN2020096744-appb-000103
第一步
(E)-4-溴-1-(3-羟基氮杂环丁-1-基)丁-2-烯1-酮40a
将化合物11a(0.5g,3.0mmol)溶于二氯甲烷(5mL),于室温加入三乙胺(0.3g,3.3mmol)、3-羟基氮杂环丁烷(0.2g,3.3mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.6g,3.3mmol)和1-羟基苯并三唑(0.4g,3.3mmol),搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物40a(0.5g),产率:72%。
第二步
(E)-1-(3-羟基氮杂环丁-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮40b
采用实施例1的合成路线,将第四步原料1h替换为40a,制得标题化合物40b(43mg),产率:51%。
第三步
(E)-1-(3-羟基氮杂环丁-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮40
采用实施例1的合成路线,将第五步原料1i替换为化合物40b,制得标题化合物40(27mg),产率:67%。
MS m/z(ESI):622.1[M+1]。
1H NMR(400MHz,CD 3OD)7.71-7.66(m,2H),7.53-7.51(m,1H),7.35-7.21(m,8H),6.71-6.65(m,2H),4.63-4.48(m,2H),4.43(s,2H),4.29-4.25(m,1H),4.05-4.00(m,3H),3.85-3.81(m,1H),3.48-3.33(m,2H),1.17-1.11(m,4H)。
实施例41
(E)-1-((S)-3-羟基吡咯烷-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮41
Figure PCTCN2020096744-appb-000104
第一步
(S,E)-4-溴-1-(3-羟基吡咯烷-1-基)丁-2-烯1-酮41a
将化合物11a(0.5g,3.0mmol)溶于二氯甲烷(5mL),于室温加入三乙胺(0.3g,3.3mmol)、(S)-3-羟基吡咯烷(0.2g,3.3mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.6g,3.3mmol)和1-羟基苯并三唑(0.4g,3.3mmol),搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物41a(0.5g),产率:73%。
第二步
(E)-1-((S)-3-羟基吡咯烷-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮41b
采用实施例1的合成路线,将第四步原料1h替换为41a,制得标题化合物41b(41mg),产率:57%。
第三步
(E)-1-((S)-3-羟基吡咯烷-1-基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮41
采用实施例1的合成路线,将第五步原料1i替换为化合物41b,制得标题化合物41(37mg),产率:69%。
MS m/z(ESI):636.2[M+1]。
1H NMR(400MHz,CD 3OD)7.71-7.66(m,2H),7.53-7.51(m,1H),7.35-7.22(m,8H),6.71-6.66(m,2H),4.44(s,2H),4.07-4.00(m,2H),3.74-3.67(m,2H),3.55-3.51(m,5H),2.21-2.19(m,2H),1.19-1.13(m,4H)。
实施例42
(E)-1-((R)-2-(羟甲基)吗啉基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮42
Figure PCTCN2020096744-appb-000105
第一步
(R,E)-4-溴-1-(2-(羟甲基)吗啉基)丁-2-烯1-酮42a
将化合物11a(0.5g,3.0mmol)溶于二氯甲烷(5mL),于室温加入三乙胺(0.3g,3.3mmol)、(R)-吗啉-2-基甲醇(0.4g,3.3mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.6g,3.3mmol)和1-羟基苯并三唑(0.4g,3.3mmol),搅拌反应12小时。停止并冷却反应,加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×2),合并有机相,饱和氯化钠溶液洗涤(20mL×4),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层色谱法以展开剂体系A纯化所得残余物,得到标题产物42a(0.5g),产率:63%。
第二步
(E)-1-((R)-2-(羟甲基)吗啉基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮42b
采用实施例1的合成路线,将第四步原料1h替换为42a,制得标题化合物42b(31mg),产率:47%。
第三步
(E)-1-((R)-2-(羟甲基)吗啉基)-4-((1-(((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)甲基)环丙基)氨基)丁-2-烯-1-酮42
采用实施例1的合成路线,将第五步原料1i替换为化合物42b,制得标题化合物42(27mg),产率:59%。
MS m/z(ESI):666.1[M+1]。
1H NMR(400MHz,CD 3OD)7.67-7.65(m,2H),7.52-7.50(m,1H),7.31-7.26(m,7H),6.81-6.77(m,1H),6.61-6.59(m,2H),4.45-4.42(m,1H),4.30(s,2H),3.99-3.93(m,2H),3.61-3.42(m,9H),3.03-3.00(m,1H),0.81-0.72(m,4H)。
实施例43
(E)-1-吗啉基-4-((2-((5-((Z)-4,4,4-三氟-1-(3-氟-1H-吲唑-5-基)-2-苯基丁-1-烯-1-基)吡啶-2-基)氧基)乙基)氨基)丁-2-烯-1-酮43
Figure PCTCN2020096744-appb-000106
采用实施例1的合成路线,将第四步原料1g替换为43a(采用专利申请“WO2018098305中说明书第77页的实施例1”公开的方法制备得到),制得标题化合物43(21mg),产率:42%。
MS m/z(ESI):610.2[M+1]。
1H NMR(400MHz,CD 3OD)7.69-7.67(m,2H),7.53-7.50(m,1H),7.32-7.20(m,7H),6.80-6.59(m,3H),4.36-4.34(m,2H),3.66-3.58(m,10H),3.48-3.40(m,3H),3.13-3.10(m,2H)。
生物学评价
以下结合测试例进一步描述解释本公开,但这些实施例并非意味着限制本公开的范围。
测试例1、本公开化合物对雌激素受体报告基因活性的抑制作用
1、实验目的
本实验的目的是测试本公开化合物对雌激素受体报告基因活性的抑制作用,根据IC 50大小评价化合物的体外活性。
2、实验方法
表达雌激素受体反应元件控制的荧光素酶报告基因ERE-luc(金唯智生物科技有限公司合成)的MCF7细胞MCF7/ERE-luc(ATCC,HTB-22)使用含有10%胎牛血清和500μg/ml G418的MEM(GE Healthcare,SH30024.01)培养基进行培养。实验第一天,使用含有10%活性炭处理的胎牛血清(BioSun,BS-0004-500)的MEM 不完全培养基将MCF7/ERE-luc细胞以30,000个/孔的密度种于96孔板,每孔100μl细胞悬液,放置37℃,5%CO 2的细胞培养箱培养过夜。第二天,每孔加入10μl用上述不完全培养基配制的β-雌二醇(SIGMA,E2758-250MG)和不同浓度的待测化合物,β-雌二醇的终浓度是0.1nM,化合物的终浓度是从10μM开始进行10倍梯度稀释的9个浓度点,设置含有0.5%DMSO的空白对照,放置37℃,5%CO 2的细胞培养箱培养20小时。第三天,取出96孔板,每孔加入100μl ONE-Glo TMLuciferase Assay system(Promega,E6110)检测荧光素酶的活性,室温放置3分钟至细胞充分裂解后,使用多标记微孔板酶标仪(PerkinElmer,VICTOR 3)读取发光信号值,用Graphpad Prism软件根据化合物的浓度和发光信号值计算化合物抑制活性的IC 50值。
3、测试结果
本公开中化合物对雌激素受体报告基因活性的抑制作用通过以上的试验进行测定,用Graghpad Prism对化学发光信号值与化合物的对数浓度作图,测得的IC 50值见下表1。
表1 本公开中化合物对雌激素受体报告基因活性的抑制作用IC 50
实施例编号 IC 50(nM)
1 1
2 3
3 4
4 4
5 3
6 0.5
7 38
8 3
9 2
10 8
11 3
12 2
13 3
14 2
15 3
16 5
17 6
18 6
19 4
22 24
23 13
24 9
25 3
26 3
27 7
28 4
29 3
30 3
31 6
32 28
33 3
34 3
35 4
36 5
37 8
38 5
39 2
40 2
41 3
42 0.7
结论:本公开化合物对雌激素受体报告基因具有明显的抑制作用。
测试例2、本公开化合物对MCF7细胞增殖的抑制效应
1、实验目的
本实验的目的是测定本公开化合物对MCF7细胞增殖的抑制活性,根据IC 50大小评价化合物的体外活性。
2、实验方法
MCF7细胞(ATCC,HTB-22)用含有10%胎牛血清的MEM(GE Healthcare,SH30024.01)完全培养基进行培养。实验第一天,使用完全培养基将MCF7细胞以3,000个/孔的密度种于96孔板,每孔100μl细胞悬液,放置37℃,5%CO 2的细胞培养箱培养过夜。第二天吸掉培养基,每孔更换为135μl含有2%胎牛血清的MEM不完全培养基,同时每孔加入15μl用不完全培养基配制的不同浓度的待测化合物,化合物的终浓度是从100nM开始进行4倍梯度稀释的9个浓度点,设置含有0.5%DMSO的空白对照,放置37℃,5%CO 2的细胞培养箱培养144小时。第八天,取出96孔细胞培养板,每孔加入150μl
Figure PCTCN2020096744-appb-000107
Luminescent Cell Viability Assay(Promega,G7573),室温放置10分钟后,使用多标记微孔板酶标仪(PerkinElmer,VICTOR 3)读取发光信号值,用Graphpad Prism软件根据化合物的浓度和发光信号值计算化合物抑制活性的IC 50值。
3、数据分析
用Graghpad Prism对化学发光信号值与化合物的对数浓度作图,得出化合物的 IC 50值。结果参见表2。
表2 本公开化合物对MCF7细胞增殖的抑制效应的IC 50
实施例编号 IC 50(nM) 最大抑制(%)
1 0.5 100
2 0.6 112
3 2 101
4 1.5 113
5 0.1 96
6 0.15 94
7 2 102
8 0.1 113
9 0.1 110
10 0.3 104
11 0.5 110
12 0.4 108
13 0.6 88
14 0.8 108
15 0.2 107
16 0.8 102
17 0.6 96
18 1 100
19 0.4 97
21 45 90
22 4 103
23 1 97
24 1 98
25 0.5 109
26 0.3 103
27 0.3 86
28 0.1 90
29 0.4 93
30 0.2 93
31 0.7 97
32 5 100
33 0.3 105
34 0.2 103
35 1.1 105
36 0.5 104
37 0.8 98
38 0.8 96
39 0.2 113
40 0.1 115
41 0.1 115
42 0.03 105
结论:本公开化合物对MCF7细胞增殖具有明显的抑制作用。
测试例3、本公开化合物对表达ERα突变体MCF7细胞增殖的抑制效应
1、实验目的
本实验的目的是测定本公开化合物对表达ERα突变体MCF7细胞增殖的抑制活性。
2、实验方法
定点突变和细胞系构建
人雌激素受体α(estrogen receptor α,ERα)蛋白的突变体ERα Y537S与ERα D538G使用双引物PCR的方式以野生型ESR1基因的cDNA(Accession No.NM000125)为模板进行定点突变获得。突变所使用的引物序列如下(下划线标出的核苷酸为突变的位点):
Figure PCTCN2020096744-appb-000108
将突变体ESR1的cDNA克隆至目标慢病毒载体pCDH-CMV-MCS-EF1-Puro(SBI,CD510B-1)上。然后将带有突变体ESR1基因序列的慢病毒质粒以及慢病毒包装质粒pVSV-G和pCMV-dR8.91(SBI,LV500A-1)通过Lipofectamine 3000 Transfection Reagent(ThermoFisher Scientific,Cat#L3000075)转染到HEK-293T细胞(ATCC,CRL-3216)中。转染后48小时,将带有病毒的含10%FBS的DMEM高糖培养基(GE Healthcare,SH30243.01)上清过滤,超速离心获得病毒沉淀,用适量的含10%FBS、非必需氨基酸(SIGMA,M7145-100ML)、丙酮酸钠(SIGMA,S8636-100ML)的MEM培养基(GE Healthcare,SH30024.01)重悬溶解后,加入到MCF7细胞(ATCC,HTB-22)中,并加入终浓度为8μg/ml的聚凝胺(SIGMA,H9268-5G)孵育过夜。转染两天后,在细胞培养液中加入1μg/ml的嘌呤霉素 (invitrogen,A11138-03)进行抗性筛选,约两周后得到能够稳定表达ERαY537S与ERαD538G突变体的MCF7细胞系。
细胞增殖抑制实验
将表达ERα突变体的MCF7细胞用含有10%胎牛血清的MEM(GE Healthcare,SH30024.01)完全培养基进行培养。实验第一天,使用完全培养基将细胞以3,000个/孔的密度种于96孔板,每孔100μl细胞悬液,放置37℃,5%CO 2的细胞培养箱培养过夜。第二天吸掉培养基,每孔更换为135μl含有2%胎牛血清的MEM不完全培养基,同时每孔加入15μl用不完全培养基配制的不同浓度的待测化合物,化合物的终浓度是从100nM开始进行4倍梯度稀释的9个浓度点,设置含有0.5%DMSO的空白对照,放置37℃,5%CO 2的细胞培养箱培养144小时。第八天,取出96孔细胞培养板,每孔加入150μl
Figure PCTCN2020096744-appb-000109
Luminescent Cell Viability Assay(Promega,G7573),室温放置10分钟后,使用多标记微孔板酶标仪(PerkinElmer,VICTOR 3)读取发光信号值,用Graphpad Prism软件根据化合物的浓度和发光信号值计算化合物抑制活性的IC 50值。
表3 本公开化合物对表达ERα突变体MCF7细胞增殖的抑制效应的IC 50
Figure PCTCN2020096744-appb-000110
Figure PCTCN2020096744-appb-000111
结论:本公开化合物对表达ERα突变体MCF7细胞增殖具有明显的抑制作用。
测试例4、本发明化合物对Na v1.5抑制活性的测定
本实验的目的是为了调查化合物在离体实验中对Na V1.5离子通道的影响。Na V1.5离子通道稳定地表达在HEK293细胞上。在Na V1.5电流稳定后,比较化合物应用前后Na V1.5电流的大小,可以得到化合物对Na V1.5离子通道的影响。
1实验材料及仪器
1)膜片钳放大器:patch clamp PC-505B(WARNER instruments)
2)数模转换器:Digidata 1440A(Axon CNS)
3)微操控仪:MP-225(SUTTER instrument)
4)倒置显微镜:TL4(Olympus)
5)玻璃微电极拉制仪:PC-10(NARISHIGE)
6)微电极玻璃毛细管:B12024F(武汉微探科学仪器有限公司)
7)二甲基亚砜(DMSO):D2650(Sigma-Aldrich)
2实验步骤
2.1化合物配制
配制细胞内外液的化合物除用于酸碱滴定的NaOH和KOH外,均从Sigma(St.Louis,MO)公司购买。
细胞外液为:NaCl,137mM;KCl,4mM;CaCl 2,1.8mM;MgCl 2,1mM;HEPES,10;葡萄糖10mM;pH 7.4(NaOH滴定)。
细胞内液为:天冬氨酸,140mM;MgCl 2,2mM;EGTA 11mM;HEPES,10mM;pH 7.2(CsOH滴定)。
测试化合物的保存浓度为9mM,溶于二甲基亚砜(DMSO)。测试当天再溶于细胞外液,配制成要求浓度。
2.2手动膜片钳测试过程
1)化合物配制成指定浓度的溶液后,按浓度由高到低顺序将药液依次加入各个管道,并对各个管道进行标记。
2)将细胞转移到灌流槽中,用细胞外液进行灌流。将细胞内液分批少量储存于-80℃冰箱,实验当天融化。电极用PC-10(Narishige,Japan)拉制。全细胞膜片钳记录,噪音用采样频率的五分之一进行过滤。
3)化合物均采用利用自身重力的灌流系统进行灌流。每个浓度至少测试两个细胞。在电流稳定(或5分钟)后,再比较化合物使用前后的电流大小变化来计算化合物的阻断作用。
4)清洗灌流槽。按药液浓度有高到低进行冲洗,每个浓度药液冲洗20秒。最后用细胞外液冲洗1分钟。
2.3测试电压方程(resting(静息状态))及结果
将细胞钳制在–80mV,然后用持续8毫秒方波去极化到Vhalf,然后用20毫秒的方波超极化到-120mV,再用20毫秒的方波去极化到-10mV,以得到Na V1.5电流。这一程序每15秒重复一次。检测-10mV方波引发的最大电流,待其稳定后,灌流测试化合物,当反应稳定后,计算阻断的强度。
3数据分析
资料存于计算机系统做分析,资料采集和分析用pCLAMP 10(Molecular Devices,Union City,CA)。电流稳定指连续4次扫描的峰值电流的绝对平均值超过200pA而CV值小于10%,或者连续4次扫描的峰值电流的平均值在200pA和50pA之间而且CV值小于30%。电流稳定后最后4个电流峰值的均值用来计算化合物在此溶度的作用。
本发明化合物对Na v1.5的抑制活性通过以上的试验进行测定,测得的IC 50值见下表4。
表4 本发明化合物对Na v1.5通道活性抑制的IC 50
实施例编号 IC 50(uM)
1 >10
39 8.2
40 11.5
41 5.3
43 2.7
结论:本发明中的化合物1、39-41对Na v1.5通道的抑制效果弱于实施例化合物43,可见心脏毒性风险明显低于实施例化合物43。
药代动力学评价
测试例5、本公开实施例化合物的BALB/C裸鼠药代动力学测试
1、摘要
以BALB/C裸鼠为受试动物,应用LC/MS/MS法测定了BALB/C裸鼠灌胃给予本公开化合物后不同时刻血浆中的药物浓度。研究本公开化合物在BALB/C裸鼠体内的药代动力学行为,评价其药动学特征。
2、试验方案
2.1试验药品及仪器
实施例1化合物、实施例14化合物、实施例15化合物、实施例39-42化合物。
LC/MS/MS的仪器来源:LC/MS/MS(API4000三重四极杆串联质谱仪(No.3)(美国Applied Biosystems公司)、Shimadzu LC-30AD超高效液相色谱系统(日本Shimadzu公司)。
2.2试验动物
BALB/C裸鼠63只,雌性,平均分成7组,9只为1组,购自杰思捷实验动物有限公司提供,动物生产许可证号SCXK(沪)2013-0006。
2.3药物配制
对于实施例14,称取适量样品,加5%体积的DMSO、5%体积的吐温80和90%体积的生理盐水配制成0.1mg/mL的无色澄清透明液体。
对于实施例1、15、39-42,称取适量样品,加9%体积的PEG400、0.5%体积的吐温80和90.5%体积的0.5%CMC-Na配制成1.5mg/mL的无色澄清透明液体。
2.4给药
禁食一夜后分别灌胃给药,给药体积0.2ml/10g,实施例14化合物给药剂量为2mg/kg,实施例1化合物、实施例15化合物、实施例39-42化合物给药剂量为30mg/kg。
3、操作
Balb/C裸鼠63只,雌性;禁食一夜后灌胃给药。
对于实施例化合物1、39,于给药后0.25、0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时采血0.1ml(每个时间点3只动物),置于肝素(国药集团化学试剂有限公司)处理的采血管中,3500rpm离心10分钟分离血浆,于-20℃保存。测定药物灌胃给药后裸鼠血浆中的待测化合物含量:取给药后各时刻的裸鼠血浆25μL,加入内标溶液喜树碱(中国生物制品检定所)30μL(100ng/mL)和乙腈225μL,涡旋混合5分钟,离心10分钟(3700-4000转/分钟),血浆样品取上清液0.1-0.5μL进行LC/MS/MS分析。
对于实施例化合物14、15,于给药后0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时采血0.1ml(每个时间点3只动物),置于肝素化试管中,3500rpm离心10分钟分离血浆,于-20℃保存。测定药物灌胃给药后裸鼠血浆中的待测化合物含量:取给药后各时刻的裸鼠血浆25μL,加入内标溶液喜树碱30μL(100ng/mL)和乙腈200μL,涡旋混合5分钟,离心10分钟(3700转/分钟),血浆样品取上清液0.25-1μL 进行LC/MS/MS分析。
对于实施例化合物40-42,于给药后0.25、0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时采血0.1ml(每个时间点3只动物),置于EDTA-K2(上海泰坦科技股份有限公司)抗凝试管中,10000rpm离心1分钟(4℃),1小时内分离血浆,于-20℃保存,采血至离心过程在冰浴条件下操作。测定药物灌胃给药后裸鼠血浆中的待测化合物含量:取给药后各时刻的裸鼠血浆25μL,加入内标溶液喜树碱50μL(100ng/mL)和乙腈200μL,涡旋混合5分钟,离心10分钟(3700转/分钟),血浆样品取上清液1μL进行LC/MS/MS分析。4、BALB/C裸鼠药代动力学参数结果
本公开实施例化合物的药代动力学参数如下表5。
Figure PCTCN2020096744-appb-000112
结论:本公开化合物的药代吸收良好,具有明显的药代吸收效果。
测试例6、雌激素受体ERα野生型和ERαY537S突变型共价修饰生物学评价1、实验目的
本实验的目的是测定本公开化合物对雌激素受体ERα野生型和ERαY537S突变型的共价修饰作用。
2、实验方法
雌激素受体ERα野生型和ERαY537S突变型的配体结合区域(LBD,ligand binding domain,aa296-554)由大肠杆菌表达并纯化。将2μM ERα野生型或ERαY537S突变型蛋白和10μM化合物加入到含有50mM Tris-HCl,pH7.5,150mM NaCl,1mM TCEP,5%丙三醇的缓冲液中混匀,置于4℃孵育24小时后,进行高分辨质谱(安捷伦科技公司,Agilent Q-tof 6530)检测。或者将1μM ERα野生型或ERαY537S突变型蛋白和3μM化合物加入到含有50mM Tris-HCl,pH7.5,150mM NaCl,1mM TCEP,5%丙三醇的缓冲液中混匀,置于37℃孵育15分钟后,进行高分辨质谱检测。在质谱检测结果中分子量为蛋白与化合物之和的峰即为共价修饰产物,通过计算未结合化合物蛋白与总蛋白的比值算出共价修饰的百分比,结果如下表6。
表6 本公开化合物对雌激素受体ERα野生型和ERαY537S突变型共价修饰24小时后的共价修饰比
Figure PCTCN2020096744-appb-000113
Figure PCTCN2020096744-appb-000114
结论:测试化合物对ERα野生型或ERαY537S突变型蛋白均具有很好的共价修饰作用。

Claims (21)

  1. 一种通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐:
    Figure PCTCN2020096744-appb-100001
    其中:
    G为CH或N;
    Z选自键、CR 5R 6、-O-(CH 2) t-或-NR 7-(CH 2) t-;
    环A选自环烷基或杂环基;
    R a选自-CH 2CH=CHC(O)NR 8R 9、-C(O)CH=CR 10R 11和-C(O)C≡CR 12
    R b选自氢原子或烷基;
    R 1各自相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 2选自氢原子、卤素、烷基、卤代烷基、烷氧基、氨基、氰基、硝基、羧基、醛基、羟基、羟烷基、环烷基、芳基和杂芳基;
    R 3各自相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、NR 13C(O)R 14、C(O)NR 13R 14、SO 2R 15、环烷基、杂环基、芳基和杂芳基;
    R 4各自相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 5和R 6相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 7选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 8和R 9相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    或者,R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R 10和R 11相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 12选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 13和R 14相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 15选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    m为0或1;
    n为0、1、2或3;
    q为0、1、2、3、4或5;
    s为0、1、2或3且
    t为0、1、2、3、4、5或6。
  2. 根据权利要求1所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中环A选自C 3-C 6环烷基或3至6元的杂环基;所述的杂环基含有1~3个选自N、O或S的杂原子;优选地,环A选自环丙基、环戊基、环己基、四氢吡咯基或哌啶基。
  3. 根据权利要求1或2所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,其中环A选自
    Figure PCTCN2020096744-appb-100002
  4. 根据权利要求1或2所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,其为通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐:
    Figure PCTCN2020096744-appb-100003
    其中:
    环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如权利要求1所定义。
  5. 根据权利要求1至4中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,其为通式(III)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐:
    Figure PCTCN2020096744-appb-100004
    其中:G、Z、R 1、R 2、R 3、R 4、R a、R b、n、s和q如权利要求1中所定义。
  6. 根据权利要求1至5中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中G为N原子。
  7. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 1为选自卤素或氢原子。
  8. 根据权利要求1至7中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 2为卤代烷基,优选C 1-C 6卤代烷基,更优选-CH 2CF 3
  9. 根据权利要求1至8中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 3选自氢原子、氰基、卤素、烷基、卤代烷基、NR 13C(O)R 14、C(O)NR 13R 14和SO 2R 15;R 13和R 14相同或不同,且各自独立地选自氢原子和烷基;R 15选自氢原子和烷基。
  10. 根据权利要求1至9中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R 4相同或不同,各自独立地选自氢原子、卤素、烷基和烷氧基,优选氢原子。
  11. 根据权利要求1至10中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中Z选自键、-O-、-O-CH 2-和-NH-。
  12. 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R a选自-CH 2CH=CHC(O)NR 8R 9、-C(O)CH=CR 10R 11和-C(O)C≡CR 12
    R 8和R 9相同或不同,且各自独立地选自氢原子、烷基、环烷基或杂环基;
    或者R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R 10和R 11相同或不同,各自独立地为氢原子或烷基;
    R 12选自氢原子或烷基。
  13. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中R a选自-CH 2CH=CHC(O)N(CH 3) 2、-CH 2CH=CHC(O)NH(CH 3)、-CH 2CH=CHC(O)NHC(CH 3) 3、-C(O)CH=CH 2、-C(O)C≡CCH 3
    Figure PCTCN2020096744-appb-100005
    Figure PCTCN2020096744-appb-100006
    优选地,R a选自-CH 2CH=CHC(O)N(CH 3) 2、-CH 2CH=CHC(O)NH(CH 3)、-CH 2CH=CHC(O)NHC(CH 3) 3、-C(O)CH=CH 2、-C(O)C≡CCH 3
    Figure PCTCN2020096744-appb-100007
    Figure PCTCN2020096744-appb-100008
  14. 根据权利要求1至13中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,其中所述化合物选自:
    Figure PCTCN2020096744-appb-100009
    Figure PCTCN2020096744-appb-100010
  15. 一种通式(IA)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,
    Figure PCTCN2020096744-appb-100011
    其中:
    R w为氨基保护基,优选四氢吡喃基;
    R选自-CH 2CH=CHC(O)NR 8R 9、-C(O)CH=CR 10R 11或-C(O)C≡CR 12
    R 8和R 9相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    或者,R 8和R 9与相连接的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、卤素、氨基、氰基、硝基、羟基、羟烷基、-COOR 16-、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R 10和R 11相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羧基、醛基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 12选自氢原子、烷基、卤代烷基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 16选自氢原子、烷基、卤代烷基和羟烷基;
    环A、G、Z、R 1、R 2、R 3、R 4、R b、n、m、s和q如权利要求1中所定义。
  16. 根据权要求15所述的通式(IA)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐,其中所述化合物选自:
    Figure PCTCN2020096744-appb-100012
    Figure PCTCN2020096744-appb-100013
    Figure PCTCN2020096744-appb-100014
  17. 一种制备根据权利要求1至14中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐的方法,该方法包括:
    Figure PCTCN2020096744-appb-100015
    通式(IA)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐在酸性条件下发生脱保护反应,得到通式(I)化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐;
    其中:
    R w为氨基保护基,优选四氢吡喃基;
    R如权利要求15中所定义;
    环A、G、Z、R 1、R 2、R 3、R 4、R a、R b、n、m、s和q如权利要求1中所定义。
  18. 一种药物组合物,其含有治疗有效量的根据权利要求1至14中任一项所 述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  19. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐或根据权利要求18所述的药物组合物在制备雌激素受体调节剂中的用途。
  20. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐或根据权利要求18所述的药物组合物在制备预防和/或治疗雌激素受体介导的或依赖性的疾病或病症的药物中的用途。
  21. 根据权利要求20所述的用途,其中所述雌激素受体介导的或依赖性的疾病或病症为癌症,优选为乳腺癌、卵巢癌、子宫内膜癌、前列腺癌或子宫癌,更优选乳腺癌。
PCT/CN2020/096744 2019-06-19 2020-06-18 吲唑类衍生物、其制备方法及其在医药上的应用 WO2020253762A1 (zh)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA3143367A CA3143367A1 (en) 2019-06-19 2020-06-18 Indazole derivative, preparation method therefor, and pharmaceutical application thereof
AU2020294642A AU2020294642A1 (en) 2019-06-19 2020-06-18 Indazole derivative, preparation method therefor, and pharmaceutical application thereof
BR112021025601A BR112021025601A2 (pt) 2019-06-19 2020-06-18 Derivado de indazole, método de preparação do mesmo, e aplicação farmacêutica do mesmo
US17/618,767 US20220267304A1 (en) 2019-06-19 2020-06-18 Indazole derivative, preparation method therefor, and pharmaceutical application thereof
KR1020227000865A KR20220024476A (ko) 2019-06-19 2020-06-18 인다졸 유도체, 이의 제조 방법, 및 이의 약제학적 적용
CN202080038970.4A CN113874367A (zh) 2019-06-19 2020-06-18 吲唑类衍生物、其制备方法及其在医药上的应用
JP2021575257A JP2022537330A (ja) 2019-06-19 2020-06-18 インダゾール誘導体、その製造方法及び医薬上のその使用
EP20827542.0A EP3988547A4 (en) 2019-06-19 2020-06-18 INDAZOLE DERIVATIVE, METHOD FOR PREPARATION AND PHARMACEUTICAL APPLICATION
MX2021015486A MX2021015486A (es) 2019-06-19 2020-06-18 Derivado de indazol, metodo de preparacion y aplicacion farmaceutica del mismo.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910530981.3 2019-06-19
CN201910530981 2019-06-19

Publications (1)

Publication Number Publication Date
WO2020253762A1 true WO2020253762A1 (zh) 2020-12-24

Family

ID=74040676

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/096744 WO2020253762A1 (zh) 2019-06-19 2020-06-18 吲唑类衍生物、其制备方法及其在医药上的应用

Country Status (11)

Country Link
US (1) US20220267304A1 (zh)
EP (1) EP3988547A4 (zh)
JP (1) JP2022537330A (zh)
KR (1) KR20220024476A (zh)
CN (1) CN113874367A (zh)
AU (1) AU2020294642A1 (zh)
BR (1) BR112021025601A2 (zh)
CA (1) CA3143367A1 (zh)
MX (1) MX2021015486A (zh)
TW (1) TW202115022A (zh)
WO (1) WO2020253762A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022127904A1 (zh) * 2020-12-18 2022-06-23 江苏恒瑞医药股份有限公司 一种吲唑类衍生物的药学上可接受的盐、结晶形式及其制备方法
WO2023284790A1 (zh) * 2021-07-13 2023-01-19 江苏恒瑞医药股份有限公司 选择性雌激素受体共价拮抗剂与cdk4/6抑制剂联合在制备治疗乳腺癌药物中的用途
CN115960082A (zh) * 2021-10-13 2023-04-14 长春金赛药业有限责任公司 一种四取代的烯烃化合物、其制备方法及其在医药上的应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014160200A1 (en) 2013-03-13 2014-10-02 Dana-Farber Cancer Institute, Inc. Ras inhibitors and uses thereof
US20160347717A1 (en) 2015-05-29 2016-12-01 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
WO2016196342A1 (en) 2015-05-29 2016-12-08 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
US20180141913A1 (en) * 2016-11-24 2018-05-24 Eisai R & D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
WO2018098251A1 (en) * 2016-11-24 2018-05-31 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014160200A1 (en) 2013-03-13 2014-10-02 Dana-Farber Cancer Institute, Inc. Ras inhibitors and uses thereof
US20160347717A1 (en) 2015-05-29 2016-12-01 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
WO2016196342A1 (en) 2015-05-29 2016-12-08 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
WO2016196346A1 (en) 2015-05-29 2016-12-08 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
US20180141913A1 (en) * 2016-11-24 2018-05-24 Eisai R & D Management Co., Ltd. Tetrasubstituted alkene compounds and their use
WO2018098305A1 (en) 2016-11-24 2018-05-31 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use for the treatment of breast cancer
WO2018098251A1 (en) * 2016-11-24 2018-05-31 Eisai R&D Management Co., Ltd. Tetrasubstituted alkene compounds and their use

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
CA CANCER J CLIN, vol. 65, 2015, pages 87 - 108
GOVEK STEVEN P; NAGASAWA JOHNNY Y; DOUGLAS KARENSA L; LAI ANDILIY G; KAHRAMAN MEHMET; BONNEFOUS CELINE; APARICIO ANNA M; DARIMONT : "Optimization of an indazole series of selective estrogen receptor degraders: Tumor regression in a tamoxifen-resistant breast cancer xenograft.", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 25, no. 22, 3 October 2015 (2015-10-03), pages 5163 - 5167, XP029321431, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2015.09.074 *
JOURNAL OF MEDICINAL CHEMISTRY, vol. 47, no. 18, 2004, pages 4613 - 4626
JOURNAL OF ORGANIC CHEMISTRY, vol. 67, no. 11, 2002, pages 3965 - 3968
JOURNAL OF ORGANIC CHEMISTRY, vol. 83, no. 2, 2018, pages 930 - 938
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 126, no. 46, 2004, pages 15195 - 15201
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 135, no. 12, 2013, pages 4628 - 4631
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 140, no. 1, 2018, pages 155 - 158
NAT GENET, vol. 45, 2013, pages 1446 - 51
NAT REV CLIN ONCOL., vol. 12, no. 10, October 2015 (2015-10-01), pages 573 - 83
ORGANIC LETTERS, vol. 16, no. 9, 2014, pages 2306 - 2309
PHARMACOL THER., 28 December 2017 (2017-12-28)
See also references of EP3988547A4
TETRAHEDRON, vol. 63, no. 2, 2007, pages 331 - 336
TETRAHEDRON, vol. 67, no. 7, 2011, pages 1485 - 1500
XIAOLING PUYANG, FURMAN CRAIG, ZHENG GUO ZHU, WU ZHENHUA J., BANKA DEEPTI, AITHAL KIRAN, AGOULNIK SERGEI, BOLDUC DAVID M., BUONAMI: "Discovery of Selective Estrogen Receptor Covalent Antagonists for the Treatment of ERαWT and ERαMUT Breast Cancer", CANCER DISCOVERY, vol. 8, no. 9, 1 September 2018 (2018-09-01), pages 1176 - 1193, XP055767015, ISSN: 2159-8274, DOI: 10.1158/2159-8290.CD-17-1229 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022127904A1 (zh) * 2020-12-18 2022-06-23 江苏恒瑞医药股份有限公司 一种吲唑类衍生物的药学上可接受的盐、结晶形式及其制备方法
WO2023284790A1 (zh) * 2021-07-13 2023-01-19 江苏恒瑞医药股份有限公司 选择性雌激素受体共价拮抗剂与cdk4/6抑制剂联合在制备治疗乳腺癌药物中的用途
CN115960082A (zh) * 2021-10-13 2023-04-14 长春金赛药业有限责任公司 一种四取代的烯烃化合物、其制备方法及其在医药上的应用
CN115960082B (zh) * 2021-10-13 2024-06-07 长春金赛药业有限责任公司 一种四取代的烯烃化合物、其制备方法及其在医药上的应用

Also Published As

Publication number Publication date
AU2020294642A1 (en) 2022-02-03
MX2021015486A (es) 2022-01-24
BR112021025601A2 (pt) 2022-02-01
JP2022537330A (ja) 2022-08-25
TW202115022A (zh) 2021-04-16
KR20220024476A (ko) 2022-03-03
CA3143367A1 (en) 2020-12-24
EP3988547A1 (en) 2022-04-27
EP3988547A4 (en) 2022-07-27
CN113874367A (zh) 2021-12-31
US20220267304A1 (en) 2022-08-25

Similar Documents

Publication Publication Date Title
WO2021197464A1 (zh) 稠合咪唑类衍生物、其制备方法及其在医药上的应用
JP6759514B2 (ja) ブロモドメインに対して活性な化合物
CN109311870B (zh) 雌激素受体调节剂
JP4948403B2 (ja) ヒストン・デアセチラーゼの新インヒビターとしての置換インドリルアルキルアミノ誘導体
WO2020238791A1 (zh) 氢化吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2020253762A1 (zh) 吲唑类衍生物、其制备方法及其在医药上的应用
WO2022017339A1 (zh) 稠合哒嗪类衍生物、其制备方法及其在医药上的应用
KR20220151616A (ko) 에스트로겐 수용체-연관 질환의 치료 방법
TW202003495A (zh) 苯並哌啶或雜芳基並哌啶類衍生物、其製備方法及其在醫藥上的應用
KR102569031B1 (ko) Nr2b nmda 수용체 길항제로서의 피롤로피리미딘 유도체
EP2119704A1 (en) Acylguanidine derivative
WO2021121210A1 (zh) 并环类衍生物、其制备方法及其在医药上的应用
WO2020169042A1 (zh) 6-氧代-1,6-二氢哒嗪类前药衍生物、其制备方法及其在医药上的应用
EP2928466B1 (en) Use of maleimide derivatives for preventing and treating leukemia
WO2021139756A1 (zh) 三环四氢异喹啉类衍生物、其制备方法及其在医药上的应用
TW202317564A (zh) Cdk2抑制劑及其製備方法和用途
CN115594695A (zh) 大环类化合物、其制备方法及其在医药上的应用
WO2020221272A1 (zh) 吲哚类大环衍生物、其制备方法及其在医药上的应用
TW202033520A (zh) 用作fgfr4抑制劑的稠環衍生物
WO2020114519A1 (zh) 作为cdk-hdac双通路抑制剂的杂环化合物
TW201835079A (zh) 6-吡唑-[1,2,4]三唑並[4,3-α]吡啶-3-醯胺類衍生物、其製備方法及其在醫藥上的應用
RU2817737C2 (ru) Производное индазола, способ его получения и его фармацевтическое применение
CN112851583A (zh) 新型苯并氮杂䓬类化合物、组合物及其用途
CN107428682A (zh) 酰胺类衍生物、其制备方法及其在医药上的用途
WO2023274280A1 (zh) 一种联苯类衍生物抑制剂的晶型及其制备方法

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: 20827542

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3143367

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021575257

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021025601

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20227000865

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020827542

Country of ref document: EP

Effective date: 20220119

ENP Entry into the national phase

Ref document number: 112021025601

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20211217

ENP Entry into the national phase

Ref document number: 2020294642

Country of ref document: AU

Date of ref document: 20200618

Kind code of ref document: A