WO2019085996A1 - 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物 - Google Patents

作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物 Download PDF

Info

Publication number
WO2019085996A1
WO2019085996A1 PCT/CN2018/113683 CN2018113683W WO2019085996A1 WO 2019085996 A1 WO2019085996 A1 WO 2019085996A1 CN 2018113683 W CN2018113683 W CN 2018113683W WO 2019085996 A1 WO2019085996 A1 WO 2019085996A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
μmol
reaction
isomer
Prior art date
Application number
PCT/CN2018/113683
Other languages
English (en)
French (fr)
Inventor
陈新海
陈兆国
张丽
于衍新
周凯
胡伯羽
王校飞
胡国平
黎健
陈曙辉
Original Assignee
南京明德新药研发股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京明德新药研发股份有限公司 filed Critical 南京明德新药研发股份有限公司
Priority to BR112020008991-5A priority Critical patent/BR112020008991A2/pt
Priority to US16/760,740 priority patent/US11377444B2/en
Priority to EP18872475.1A priority patent/EP3712157A4/en
Priority to JP2020524756A priority patent/JP7340519B2/ja
Priority to CN201880071134.9A priority patent/CN111315750B/zh
Priority to RU2020118222A priority patent/RU2771201C2/ru
Publication of WO2019085996A1 publication Critical patent/WO2019085996A1/zh

Links

Classifications

    • 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
    • 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
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic 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
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to a series of pyridopyrimidine compounds, and their use in the preparation of mTORC1/2 double kinase activity inhibitor related drugs, in particular to the compound of formula (IV), its tautomer or its pharmaceutically acceptable
  • the use of the accepted salt in the preparation of a drug related to mTORC1/2 double kinase activity inhibitor in particular to the compound of formula (IV), its tautomer or its pharmaceutically acceptable
  • the invention relates to a signal pathway which plays a key role in tumor proliferation, invasion and metastasis and anti-apoptosis, namely phosphatidylinositol 3-kinase (PI3K)-AKT-mammalian rapamycin protease mTOR signaling pathway.
  • PI3K phosphatidylinositol 3-kinase
  • PI3K vascular endothelial growth factor
  • HGF human growth factor
  • Ang1 vascular protein I
  • FGF fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • HGF human growth factor
  • Ang1 vascular protein I
  • RTK receptor tyrosine kinase
  • PI3K activation is the production of a second messenger PIP3 on the plasma membrane, which binds to the PH domain-containing signaling proteins AKT and PDK1 (phosphoinositide dependent kinase-1), causing PDK1 to phosphorylate the AKT protein Ser308 to cause AKT activation.
  • Other substrates for PDK1 also include PKC (protein kinase C), S6K (p70S6), and SGK (serum/glucocorticoid regulated kinases).
  • AKT also known as protein kinase B (PKB) is a major effector downstream of PI3K.
  • AKT Activated AKT regulates cell function by phosphorylating downstream factors such as various enzymes, kinases, and transcription factors. AKT exerts anti-apoptotic effects by phosphorylating target proteins through various downstream pathways.
  • PTEN phosphatase and tensin homology deleted on chromosome 10.
  • PTEN is a tumor suppressor gene that causes gene mutations or deletions in a wide range of human tumors.
  • PTEN is a PIP3-phosphatase that, in contrast to PI3K, converts PIP3 to PIP2 by dephosphorylation. PTEN reduces the activation of AKT and blocks all downstream signaling events regulated by AKT.
  • mTOR As a downstream substrate of AKT, mTOR is relatively conservative in evolution and integrates various signals of nutrition, energy and growth factors, and participates in biological processes such as transcription, protein translation, ribosome synthesis and apoptosis, and plays an important role in cell growth. Important role. It has two highly homologous complexes, Tor binds to KOG01 to form mTORC1, mTOR and AVO1/AVO2/AVO3/ and LST8 form mTORC2.mTOR which is insensitive to rapamycin. Phosphorylation of downstream target protein S40S ribosomal S6 Protein kinases, such as S6K1 and 4EBP1, regulate downstream protein translation.
  • mTOR binds to eIF3, phosphorylates S6K1, activates S6K1 from eIF3, and further phosphorylates cellular substrates such as p70S6 to promote protein translation and expression.
  • 4EBP1 binds to eukaryotic transcriptional initiation factor 4E and inhibits its activity. When mtor phosphorylates 4E-BP1, it activates and separates from eif-4e to achieve eukaryotic cell transcription.
  • mTORC2 phosphorylates AKT, thereby upregulating its kinase activity.
  • this signaling pathway is used in a variety of solid tumors such as breast cancer, prostate cancer, lung cancer, colon cancer, pancreatic cancer, liver cancer, stomach cancer, colorectal cancer, kidney cancer, thyroid cancer, meningitis and acute and chronic lymphocytic leukemia. , Merkel cell tumors, etc. And it is closely related to treatment tolerance and poor prognosis. It can be seen that the development of the signaling pathway of PI3K/AKT/MTOR by developing fine molecular compounds has a good development prospect.
  • the present invention aims to find a dual mTOR small molecule compound targeting drug which has good activity and exhibits excellent effects and effects.
  • US20170281637 discloses compound AZD2014, which belongs to the mTORC1 & mTORC2 kinase inhibitor, and has the structural formula shown below:
  • the present invention provides a compound of the formula (IV), a pharmaceutically acceptable salt thereof and an isomer thereof,
  • R 1 is H
  • R 2 is Me
  • R 1 , R 2 and N on the morpholine ring form a 5- to 6-membered heterocycloalkyl group
  • n is selected from 1 and 2;
  • Ring A is selected from the group consisting of phenyl and 6-10 membered heteroaryl
  • R 4 is selected from H
  • R 5 is selected from H
  • R 4 and R 5 are bonded together to form a 5- to 6-membered heterocycloalkyl group
  • D 1 , D 2 , D 3 and D 4 are each independently selected from the group consisting of a single bond, -CH 2 -, -CH 2 CH 2 - and -O-, and at least one of D 1 , D 2 , D 3 and D 4 is not Is a single bond wherein the -CH 2 - or -CH 2 CH 2 - is optionally substituted with 1 or 2 R;
  • D 5 , D 6 , D 7 and D 8 are each independently selected from the group consisting of a single bond, —CH 2 —, —O— and —NH—, and at least one of D 5 , D 6 , D 7 and D 8 is not a single bond.
  • -CH 2 - is optionally substituted by 1 or 2 R
  • -NH- is optionally substituted by R;
  • T 1 is selected from CH and N;
  • R is independently selected from the group consisting of F, Cl, Br, I, OH, NH 2 , C 1-3 alkyl, C 1-3 alkoxy, and C 3-6 cycloalkyl, wherein said C 1-3 alkane a group, a C 1-3 alkoxy group and a C 3-6 cycloalkyl group are optionally substituted by 1 or 2 R';
  • R' is independently selected from the group consisting of F, Cl, Br, I, OH, and NH 2 ;
  • the present invention provides the above compound, a pharmaceutically acceptable salt thereof and an isomer thereof, which are selected from the group consisting of
  • R 1 is H
  • R 2 is Me
  • R 1 , R 2 and N on the morpholine ring form a 5- to 6-membered heterocycloalkyl group
  • Ring A is selected from the group consisting of phenyl and 6-10 membered heteroaryl
  • R 4 is selected from H
  • R 5 is selected from H
  • R 4 and R 5 are bonded together to form a 5- to 6-membered heterocycloalkyl group
  • D 1 , D 2 , D 3 and D 4 are each independently selected from the group consisting of a single bond, -CH 2 -, -CH 2 CH 2 - and -O-, and at least one of D 1 , D 2 , D 3 and D 4 is not Is a single bond wherein the -CH 2 - or -CH 2 CH 2 - is optionally substituted with 1 or 2 R;
  • D 5 , D 6 , D 7 and D 8 are each independently selected from the group consisting of a single bond, —CH 2 —, —O— and —NH—, and at least one of D 1 , D 2 , D 3 and D 4 is not a single bond.
  • -CH 2 - is optionally substituted by 1 or 2 R
  • -NH- is optionally substituted by R;
  • T 1 is selected from CH and N;
  • R is independently selected from the group consisting of F, Cl, Br, I, OH, NH 2 , C 1-3 alkyl, C 1-3 alkoxy, and C 3-6 cycloalkyl, wherein said C 1-3 alkane a group, a C 1-3 alkoxy group and a C 3-6 cycloalkyl group are optionally substituted by 1 or 2 R';
  • R' is independently selected from the group consisting of F, Cl, Br, I, OH, and NH 2 ;
  • the present invention provides a compound of the formula (I), a pharmaceutically acceptable salt thereof and an isomer thereof,
  • R 1 is selected from H
  • R 2 is selected from Me
  • R 1 and R 2 are bonded together to form a 5- to 6-membered heterocycloalkyl group
  • Ring A is selected from the group consisting of phenyl and 6-10 membered heteroaryl
  • R 4 is selected from H
  • R 5 is selected from H
  • D 1 , D 2 , D 3 and D 4 are each independently selected from the group consisting of a single bond, -CH 2 -, -CH 2 CH 2 - and -O-, wherein the -CH 2 - or -CH 2 CH 2 - Elected by 1 or 2 R;
  • D 5 , D 6 , D 7 and D 8 are each independently selected from the group consisting of a single bond, -CH 2 -, -O- and -NH-, wherein said -CH 2 - is optionally substituted by 1 or 2 R, - NH-optionally substituted by R;
  • T 1 is selected from CH and N;
  • T 2 is selected from the group consisting of a single bond, CH 2 and -O-;
  • R is independently selected from the group consisting of F, Cl, Br, I, OH, NH 2 , C 1-3 alkyl, C 1-3 alkoxy, and C 3-6 cycloalkyl, wherein said C 1-3 alkane a group, a C 1-3 alkoxy group and a C 3-6 cycloalkyl group are optionally substituted by 1 or 2 R';
  • R' is independently selected from the group consisting of F, Cl, Br, I, OH, and NH 2 ;
  • the R is independently selected from the group consisting of F, Cl, Br, I, OH, NH 2 , Me, Et, Wherein Me, E, Optionally substituted with 1 or 2 R', other variables are as defined herein.
  • the above R is independently selected from the group consisting of F, Cl, Br, I, OH, NH 2 , Me, CF 3 , E, t and Other variables are as defined by the present invention.
  • the structural unit Selected from Other variables are as defined by the present invention.
  • R 1 and R 2 are linked together, and the structural unit Selected from Other variables are as defined by the present invention.
  • the above R 3 is selected from the group consisting of NH 2 , 1H-pyrazolyl and 1H-1,2,4-triazolyl, wherein the NH 2 , 1H-pyrazolyl and 1H-1,2,4-triazolyl are optionally substituted by 1, 2 or 3 R, other variables as defined by the present invention
  • the above R 3 is selected from the group consisting of NH 2 , 1H-pyrazolyl and 1H-1,2,4-triazolyl, wherein the NH 2 , The 1H-pyrazolyl and 1H-1,2,4-triazolyl are optionally substituted by 1, 2 or 3 R, and other variables are as defined in the present invention.
  • the above R 3 is selected from the group consisting of NH 2 , Wherein the NH 2 , Optionally substituted by 1, 2 or 3 R, other variables are as defined by the present invention.
  • the above R 3 is selected from the group consisting of NH 2 , Wherein the NH 2 , Optionally substituted by 1, 2 or 3 R, other variables are as defined by the present invention.
  • R 3 is selected from the group consisting of NH 2 ,
  • Other variables are as defined by the present invention.
  • R 3 is selected from the group consisting of NH 2 ,
  • Other variables are as defined by the present invention.
  • Ring A is selected from the group consisting of phenyl, benzo[d]oxazole, quinolyl and quinazolinyl, and other variables are as defined herein.
  • the ring A is selected from the group consisting of Other variables are as defined by the present invention.
  • the ring A is selected from the group consisting of Other variables are as defined by the present invention.
  • the structural unit Selected from Other variables are as defined by the present invention.
  • the structural unit Selected from Other variables are as defined by the present invention.
  • the above D 1 , D 2 , D 3 and D 4 are each independently selected from the group consisting of a single bond, -CH 2 -, -CH 2 CH 2 -, -O-, and And at least one of D 1 , D 2 , D 3 and D 4 is not a single bond, and other variables are as defined in the present invention.
  • the above D 5 , D 6 , D 7 and D 8 are each independently selected from the group consisting of a single bond, —CH 2 —, —O—, —NH—, And at least one of D 5 , D 6 , D 7 and D 8 is not a single bond, and other variables are as defined in the present invention.
  • the structural unit Selected from Other variables are as defined by the present invention.
  • the above compounds, pharmaceutically acceptable salts thereof, and isomers thereof are selected from the group consisting of:
  • R 1 , R 2 , R 3 , R 4 and R 5 are as defined above, and X is selected from -CH 2 -, -NH-, -O-, And -S-.
  • the present invention also provides a compound represented by the following formula, a pharmaceutically acceptable salt thereof, and an isomer thereof,
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the above compound, a pharmaceutically acceptable salt thereof and an isomer thereof as an active ingredient, and a pharmaceutically acceptable carrier.
  • the present invention also provides the use of the above compound, a pharmaceutically acceptable salt thereof, and an isomer thereof, or a pharmaceutical composition as described above, for the preparation of a medicament for treating a disease associated with mTOR double kinase.
  • the present invention also provides the use of the above compound, a pharmaceutically acceptable salt thereof, and an isomer thereof or the above composition for the preparation of a medicament for treating breast cancer, breast cancer, head and neck cancer, and colorectal cancer.
  • the compounds of the present invention were tested for mTORC1/2 kinase activity, and the data showed that the compounds of the present invention have significant and unexpected mTOR kinase inhibitory activity, which is superior to the current clinical compound AZD2014.
  • the compound of the present invention has obvious proliferation inhibitory activity against MCF-7, N87 and OE-21 cells, and has certain proliferation inhibitory activity against HT-29 cells.
  • the PK results show that the bioavailability of the compounds of the present invention is close to 100% and is a well developed, orally administrable molecule.
  • some compounds have the same efficacy as AZD2014, and the compounds of the present invention have the potential to become a variety of tumor suppressors.
  • pharmaceutically acceptable salt refers to a salt of a compound of the invention prepared from a compound having a particular substituent found in the present invention and a relatively non-toxic acid or base.
  • a base addition salt can be obtained by contacting a neutral amount of such a compound with a sufficient amount of a base in a neat solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts.
  • an acid addition salt can be obtained by contacting a neutral form of such a compound with a sufficient amount of an acid in a neat solution or a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogencarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and an organic acid salt, such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and me
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing an acid group or a base by conventional chemical methods.
  • such salts are prepared by reacting these compounds in water or an organic solvent or a mixture of the two via a free acid or base form with a stoichiometric amount of a suitable base or acid.
  • the compounds of the invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including the cis and trans isomers, the (-)- and (+)-p-enantiomers, the (R)- and (S)-enantiomers, and the diastereomeric a conformation, a (D)-isomer, a (L)-isomer, and a racemic mixture thereof, and other mixtures, such as enantiomerically or diastereomeric enriched mixtures, all of which belong to It is within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in the substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the invention.
  • enantiomer or “optical isomer” refer to stereoisomers that are mirror images of one another.
  • cis-trans isomer or “geometric isomer” is caused by the inability to freely rotate a single bond due to a double bond or a ring-forming carbon atom.
  • diastereomer refers to a stereoisomer in which the molecule has two or more chiral centers and the molecules are in a non-mirrored relationship.
  • wedge-shaped dashed keys Represents the absolute configuration of a solid center with straight solid keys
  • straight dashed keys Indicates the relative configuration of the stereocenter, using wavy lines Indicates a wedge solid key Or wedge-shaped dotted key Or with wavy lines Represents a straight solid key And straight dashed keys
  • tautomer or “tautomeric form” mean that the different functional isomers are in dynamic equilibrium at room temperature and can be rapidly converted into each other. If tautomers are possible (as in solution), the chemical equilibrium of the tautomers can be achieved.
  • proton tautomers also known as prototropic tautomers
  • prototropic tautomers include interconversions by proton transfer, such as keto-enol isomerization and imine-enes. Amine isomerization.
  • the valence tautomer includes the mutual transformation of some of the bonding electrons.
  • keto-enol tautomerization is the interconversion between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
  • the terms "enriched in one isomer”, “isomer enriched”, “enriched in one enantiomer” or “enantiomeric enriched” refer to one of the isomers or pairs
  • the content of the oligo is less than 100%, and the content of the isomer or enantiomer is 60% or more, or 70% or more, or 80% or more, or 90% or more, or 95% or more, or 96% or more, or 97% or more, 98% or more, 99% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% or more, or greater than or equal to 99.9%.
  • the term “isomer excess” or “enantiomeric excess” refers to the difference between the two isomers or the relative percentages of the two enantiomers. For example, if one of the isomers or enantiomers is present in an amount of 90% and the other isomer or enantiomer is present in an amount of 10%, the isomer or enantiomeric excess (ee value) is 80%. .
  • optically active (R)- and (S)-isomers as well as the D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If an enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or by derivatization with a chiral auxiliary wherein the resulting mixture of diastereomers is separated and the auxiliary group cleaved to provide pure The desired enantiomer.
  • a diastereomeric salt is formed with a suitable optically active acid or base, followed by conventional methods well known in the art.
  • the diastereomers are resolved and the pure enantiomer is recovered.
  • the separation of enantiomers and diastereomers is generally accomplished by the use of chromatography using a chiral stationary phase, optionally in combination with chemical derivatization (eg, formation of an amino group from an amine). Formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms that make up the compound.
  • radiolabeled compounds can be used, such as tritium (3 H), iodine -125 (125 I) or C-14 (14 C).
  • hydrogen can be replaced by heavy hydrogen to form a deuterated drug.
  • the bond composed of barium and carbon is stronger than the bond composed of common hydrogen and carbon.
  • deuterated drugs have reduced side effects and increased drug stability. Enhance the efficacy and prolong the biological half-life of the drug. Alterations of all isotopic compositions of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
  • pharmaceutically acceptable carrier refers to any formulation or carrier medium that is capable of delivering an effective amount of an active substance of the present invention, does not interfere with the biological activity of the active substance, and has no toxic side effects to the host or patient, including water, oil, Vegetables and minerals, cream bases, lotion bases, ointment bases, etc. These bases include suspending agents, tackifiers, transdermal enhancers and the like. Their formulations are well known to those skilled in the cosmetic or topical pharmaceutical arts.
  • substituted means that any one or more hydrogen atoms on a particular atom are replaced by a substituent, and may include variants of heavy hydrogen and hydrogen, as long as the valence of the particular atom is normal and the substituted compound is stable. of.
  • Oxygen substitution does not occur on the aromatic group.
  • optionally substituted means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemically achievable.
  • any variable eg, R
  • its definition in each case is independent.
  • the group may optionally be substituted with at most two R, and each case has an independent option.
  • combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • linking group When the number of one linking group is 0, such as -(CRR) 0 -, it indicates that the linking group is a single bond.
  • one of the variables When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly linked. For example, when L represents a single bond in A-L-Z, the structure is actually A-Z.
  • a substituent When a substituent is vacant, it means that the substituent is absent. For example, when X is vacant in AX, the structure is actually A.
  • the substituent can be attached to more than one atom on a ring, the substituent can be bonded to any atom on the ring, for example, a structural unit. It is indicated that the substituent R can be substituted at any position on the cyclohexyl group or cyclohexadiene.
  • substituents When the listed substituents are not indicated by which atom is attached to the substituted group, such a substituent may be bonded through any atom thereof, for example, a pyridyl group as a substituent may be passed through any one of the pyridine rings. A carbon atom is attached to the substituted group.
  • the medium linking group L is -MW-, and at this time, -MW- can be connected in the same direction as the reading order from left to right to form ring A and ring B. It is also possible to connect the ring A and the ring B in a direction opposite to the reading order from left to right. Combinations of the linking groups, substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • heterocyclyl means a stable monocyclic, bicyclic or tricyclic ring containing heteroatoms or heteroatoms which are saturated, which contain carbon atoms and 1, 2, 3 or 4 independently selected Ring heteroatoms from N, O and S.
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O) p , p is 1 or 2).
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the heterocyclic ring can be attached to the side groups of any hetero atom or carbon atom to form a stable structure.
  • heterocycles described herein can undergo substitutions at the carbon or nitrogen sites.
  • the nitrogen atom in the heterocycle is optionally quaternized.
  • a preferred embodiment is that when the total number of S and O atoms in the heterocycle exceeds 1, these heteroatoms are not adjacent to each other. Another preferred embodiment is that the total number of S and O atoms in the heterocycle does not exceed one.
  • heteroaryl means a stable 5,6,7 membered monocyclic or bicyclic or aromatic ring of a 7,8, 9 or 10 membered bicyclic heterocyclyl containing carbon atoms and 1,2. 3 or 4 ring heteroatoms independently selected from N, O and S.
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2). It is worth noting that the total number of S and O atoms on the aromatic heterocycle does not exceed one.
  • a bridged ring is formed when one or more atoms (ie, C, O, N, or S) join two non-adjacent carbon or nitrogen atoms.
  • Preferred bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and one carbon-nitrogen group. It is worth noting that a bridge always converts a single ring into a three ring. In the bridged ring, a substituent on the ring can also be present on the bridge.
  • a hetero atom may occupy a position where a heterocycloalkyl group is bonded to the rest of the molecule.
  • the 5-6 membered heterocycloalkyl group includes 5-membered and 6-membered heterocycloalkyl groups.
  • 5-6 membered heterocycloalkyl examples include, but are not limited to, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothiophenyl (including tetrahydrothiophen-2-yl and tetrahydrothiophen-3-yl, etc.) , tetrahydrofuranyl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2-piperidinyl and 3-piperidinyl, etc.), piperazinyl (including 1 - piperazinyl and 2-piperazinyl, etc.), morpholinyl (including 3-morpholinyl and 4-morpholinyl, etc.), dioxoalkyl, dithiaalkyl, isoxazolidinyl, isothiazole Alkyl, 1,2-oxazinyl, 1,2-thiaziny
  • 6-10 membered heteroaryl ring and “6-10 membered heteroaryl” are used interchangeably and the term “6-10 membered heteroaryl” means 6 to 10 rings.
  • a 6-10 membered heteroaryl group can be attached to the remainder of the molecule through a heteroatom or carbon atom.
  • the 6-10 membered heteroaryl group includes 6-8 members, 6-7 members, 6-9 members, 6 members, and 10 membered heteroaryl groups.
  • Examples of the 6-10 membered heteroaryl group include, but are not limited to, furyl groups (including 2-furyl and 3-furyl groups, etc.), pyridyl groups (including 2-pyridyl, 3-pyridyl and 4-pyridyl groups, etc.) ), pyrazinyl, pyrimidinyl (including 2-pyrimidinyl and 4-pyrimidinyl, etc.), benzothiazolyl (including 5-benzothiazolyl, etc.), fluorenyl, benzimidazolyl (including 2-benzo Imidazolyl, etc., benzoxazolyl, indenyl (including 5-indenyl, etc.), isoquinolyl (including 1-isoquinolinyl and 5-isoquinolinyl, etc.), quinoxalinyl (including 2-quinoxalinyl and 5-quinoxalinyl, etc.) or quinolyl (including 3-quinolyl and 6-quinolinyl, etc.).
  • 5-6 membered heteroaryl ring and “5-6 membered heteroaryl” are used interchangeably and the term “5-6 membered heteroaryl” means from 5 to 6 ring atoms.
  • the nitrogen atom is optionally quaternized
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O) p , p is 1 or 2).
  • a 5-6 membered heteroaryl group can be attached to the remainder of the molecule through a heteroatom or carbon atom.
  • the 5-6 membered heteroaryl group includes a 5-membered and a 6-membered heteroaryl group.
  • Examples of the 5-6 membered heteroaryl include, but are not limited to, pyrrolyl (including N-pyrrolyl, 2-pyrrolyl, and 3-pyrrolyl, etc.), pyrazolyl (including 2-pyrazolyl and 3-pyridyl) Izozolyl, etc., imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl and 5-imidazolyl, etc.), oxazolyl (including 2-oxazolyl, 4-oxazolyl and 5- Oxazolyl, etc.), triazolyl (1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl and 4H-1, 2,4-triazolyl, etc.), tetrazolyl, isoxazolyl (3-isoxazo
  • C 3-6 cycloalkyl means a saturated cyclic hydrocarbon group consisting of 3 to 6 carbon atoms which is a monocyclic and bicyclic system including C 3-6 cycloalkyl C 3-5 , C 4-5 and C 5-6 cycloalkyl, etc.; which may be monovalent, divalent or multivalent.
  • Examples of C 3-6 cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • heteroalkyl by itself or in conjunction with another term refers to a stable straight chain, branched hydrocarbon radical or combination thereof, having a number of carbon atoms and at least one heteroatom.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the hetero atom or heteroatom group may be located at any internal position of the heterohydrocarbyl group, including where the hydrocarbyl group is attached to the rest of the molecule, but the terms "alkoxy", “alkylamino” and “alkylthio” (or thioalkoxy).
  • alkyl groups which are attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • examples include, but are not limited to, -CH 2 -CH 2 -O-CH 3 , -CH 2 -CH 2 -NH-CH 3 , -CH 2 -CH 2 -N(CH 3 )-CH 3 , -CH 2 -S -CH 2 -CH 3 , -CH 2 -CH 2 , -S(O)-CH 3 , -CH 2 -CH 2 -S(O) 2 -CH 3 .
  • Up to two heteroatoms may be consecutive, for example, -CH 2 -NH-OCH 3.
  • alkyl is used to denote a straight or branched saturated hydrocarbon group, which may be monosubstituted (eg, -CH 2 F) or polysubstituted (eg, -CF 3 ), and may be monovalent (eg, Methyl), divalent (such as methylene) or polyvalent (such as methine).
  • alkyl group include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl). , t-butyl), pentyl (eg, n-pentyl, isopentyl, neopentyl) and the like.
  • C 1-3 alkyl is used to denote a straight or branched saturated hydrocarbon group consisting of 1 to 3 carbon atoms.
  • the C 1-3 alkyl group includes C 1-2 and C 2-3 alkyl groups and the like; it may be monovalent (such as methyl), divalent (such as methylene) or polyvalent (such as methine).
  • Examples of the C 1 - 3 alkyl group include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
  • C 1-3 alkoxy denotes those alkyl groups containing from 1 to 3 carbon atoms attached to the remainder of the molecule through an oxygen atom.
  • the C 1-3 alkoxy group includes C 1-2 , C 2-3 , C 3 and C 2 alkoxy groups and the like.
  • Examples of the C 1-3 alkoxy group include, but are not limited to, a methoxy group, an ethoxy group, a propoxy group (including n-propoxy group and isopropoxy group), and the like.
  • a cycloalkyl group includes any stable cyclic or polycyclic hydrocarbon group, any carbon atom which is saturated, may be monosubstituted or polysubstituted, and may be monovalent, divalent or multivalent.
  • Examples of such cycloalkyl groups include, but are not limited to, cyclopropyl, norbornyl, [2.2.2]bicyclooctane, [4.4.0]bicyclononane, and the like.
  • halo or “halogen”, by itself or as part of another substituent, denotes a fluorine, chlorine, bromine or iodine atom.
  • haloalkyl is intended to include both monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is intended to include, but is not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Wait.
  • examples of haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • alkoxy represents attached through an oxygen bridge
  • C 1-6 alkoxy groups include C 1, C 2, C 3 , C 4, C 5 , and C 6 alkoxy groups.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy and S- Pentyloxy.
  • aryl denotes a polyunsaturated, aromatic hydrocarbon substituent which may be monosubstituted or polysubstituted, which may be monovalent, divalent or polyvalent, which may be monocyclic or polycyclic ( For example, 1 to 3 rings; at least one of which is aromatic), they are fused together or covalently linked.
  • heteroaryl refers to an aryl (or ring) containing one to four heteroatoms. In an illustrative example, the heteroatoms are selected from the group consisting of B, N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom is optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • aryl or heteroaryl groups include phenyl, naphthyl, biphenyl, pyrrolyl, pyrazolyl, imidazolyl, pyrazinyl, oxazolyl, phenyl-oxazolyl, isomerism Azyl, thiazolyl, furyl, thienyl, pyridyl, pyrimidinyl, benzothiazolyl, indolyl, benzimidazolyl, indolyl, isoquinolyl, quinoxalinyl, quinolinyl, 1 -naphthyl, 2-naphthyl, 4-biphenylyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl
  • C n-n+m or C n -C n+m includes any one of n to n+m carbons, for example, C 1-12 includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 , and C 12 , also including any range of n to n+m, for example, C 1-12 includes C 1 - 3 , C 1-6 , C 1-9 , C 3-6 , C 3-9 , C 3-12 , C 6-9 , C 6-12 , and C 9-12 , etc.; similarly, n to n
  • the +m element indicates that the number of atoms on the ring is n to n+m, for example, the 3-12 element ring includes a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-
  • a 10-membered ring, a 11-membered ring, and a 12-membered ring and includes any one of n to n+m, for example, a 3-12-membered ring including a 3-6-membered ring, a 3-9-membered ring, and a 5-6-membered ring. Ring, 5-7 membered ring, 6-7 membered ring, 6-8 membered ring, and 6-10 membered ring.
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, combinations thereof with other chemical synthetic methods, and those well known to those skilled in the art. Equivalent alternatives, preferred embodiments include, but are not limited to, embodiments of the invention.
  • the compounds of the invention may be used in a variety of uses or indications, including but not limited to the specific uses or indications listed herein.
  • the solvent used in the present invention is commercially available.
  • Embodiments of the invention relate to neutral separation using high performance liquid chromatography.
  • the present invention employs the following abbreviations: aq for water; HATU for O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate ; EDC stands for N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride; m-CPBA stands for 3-chloroperoxybenzoic acid; eq stands for equivalent, equivalent; CDI stands for Carbonyldiimidazole; DCM stands for dichloromethane; PE stands for petroleum ether; DIAD stands for diisopropyl azodicarboxylate; DMF stands for N,N-dimethylformamide; DMSO stands for dimethyl sulfoxide; EtOAc stands for acetic acid Esters; EtOH for ethanol; MeOH for methanol; CBz for benzyloxycarbonyl, an amine protecting group; BOC for
  • the compounds of the invention are based on conventional nomenclature in the art or Software naming, commercially available compounds using the supplier catalog name.
  • Peak position of compound 18-1 3.032 min (chiral column: AD-3 150 ⁇ 4.6 mm, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.5 mL/min, column temperature: 40 °C).
  • Peak position of compound 18-2 3.587 min (chiral column: AD-3 150 ⁇ 4.6 mm, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.5 mL/min, column temperature: 40 °C).
  • Methyltriphenylphosphonium bromide (102 g, 285 mmol, 2 eq) and potassium t-butoxide (1 M, 257 mL, 1.8 eq) were dissolved in tetrahydrofuran (500 mL), and reacted for 1 hour at room temperature, and compound 30a (10.0 g, 143 mmol) was added. , 10.7 mL, 1 eq), reacted at room temperature for 17 hours, and then distilled at 80 ° C to distill the compound 30b.
  • Peak position of compound 32-1 2.105 min (chiral column: AD-3 100 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.8 mL/min, Column temperature: 40 ° C)
  • Peak position of compound 32-2 2.632 min (chiral column: AD-3 100 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.8 mL/min, Column temperature: 40 ° C)
  • the compound methyltriphenylphosphonium bromide (32.4 g, 90.8 mmol, 1.6 eq) was dissolved in anhydrous tetrahydrofuran (400 mL) of potassium tert-butoxide (11.5 g, 102 mmol, 1.8 eq) at room temperature (20 ° C), The mixed solution was reacted at room temperature for 3 hours, and then 33a (10.0 g, 56.8 mmol, 1 eq) was added dropwise to the above solution, and the mixed solution was reacted at 20 ° C for 17 hours.
  • Peak position of compound 33-1 1.672 min (chiral column: AD-3 100 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% isopropanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.8 mL/ Min, column temperature: 40 ° C)
  • Peak position of compound 33-2 3.338 min (chiral column: AD-3 100 x 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% isopropanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.8 mL/ Min, column temperature: 40 ° C)
  • Peak position of compound 34-1 2.407 min (chiral column: OD-3 50 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: A: CO2, B: ethanol (0.05% diethylamine), maintaining 5% B 0.2 Min, then the content of B increased from 5% gradient to 40% in 1.4min, then kept 40%B1.05min, and finally kept 5%B 0.35min, flow rate: 4mL/min, column temperature: 40°C)
  • Peak position of compound 34-2 1.807 min (chiral column: OD-3 50 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: A: CO 2 , B: ethanol (0.05% diethylamine), maintaining 5% B 0.2min, then the content of B increased from 5% gradient to 40% in 1.4min, then kept 40%B 1.05min, and finally kept 5%B 0.35min, flow rate: 4mL/min, column temperature: 40°C)
  • Lithium tetrahydrogen aluminum (3.91 g, 103 mmol, 2 eq) was dissolved in tetrahydrofuran (100 mL), and compound 38a (5.00 g, 51.5 mmol, 3.37 mL, 1 eq) was added, and the mixture was reacted at 45 ° C for 18 hours, after completion of the reaction, It was quenched by adding water (4.00 mL), 15% NaOH (4.00 mL), and water (8.00 mL), and the mixture was stirred for 0.5 hour, filtered, and dried under reduced pressure to give 38b.
  • the compound 40a (0.28 g, 1.13 mmol, 1 eq) was dissolved in trifluoroacetic acid (5.00 mL) and dichloromethane (10.0 mL), and the mixture was stirred at room temperature for 2 hours. After completion of the reaction, the mixture was dried under reduced pressure to give 40b.
  • the compound 41 (120 mg, 237 ⁇ mol, 1 eq) was dissolved in methanol (5.00 mL), and a solution of potassium hydrogen persulfate (291 mg, 474 ⁇ mol, 2 eq) (5.00 mL) was added dropwise, and reacted at room temperature for 30 hours. Purified by high performance liquid chromatography to give 43.
  • 0.3 mL of water, 0.3 mL of a 15% aqueous sodium hydroxide solution, and 1 mL of water were successively added dropwise to the reaction mixture, and the mixture was filtered, and the filtrate was washed with 10 mL of ethyl acetate. The filtrate was concentrated to give 45 g.
  • Peak position of compound 45-1 0.950 min (chiral column: AD-3 50 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 4 mL/min, column Temperature: 40 ° C).
  • Peak position of compound 45-2 1.168 min (chiral column: AD-3 50 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 4 mL/min, column Temperature: 40 ° C).
  • Peak position of compound 46-1 5.297 min (chiral column: AD-3 150 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% methanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.5 mL/min, Column temperature: 40 ° C).
  • Peak position of compound 46-2 6.265 min (chiral column: AD-3 150 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% methanol (0.05% diethylamine) + carbon dioxide, flow rate: 2.5 mL/min, Column temperature: 40 ° C).
  • Peak position of compound 47-1 1.617 min (chiral column: AD-3 50 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 4 mL/min, column Temperature: 40 ° C).
  • Peak position of compound 47-2 2.000 min (chiral column: AD-3 50 ⁇ 4.6 mm, ID, 3 ⁇ m, mobile phase: 40% ethanol (0.05% diethylamine) + carbon dioxide, flow rate: 4 mL/min, column Temperature: 40 ° C).
  • Methyltriphenylphosphonium bromide (83.0 g, 232 mmol, 2.0 eq) and potassium t-butoxide (26.01, 232 mmol, 2.0 eq) were dissolved in tetrahydrofuran (1.00 L), reacted at 20 ° C for 1 hour, and compound 48a was added ( 10.0 g, 116.16 mmol, 1.0 eq), reacted at 20 ° C for 17 hours, and then distilled at 80 ° C under atmospheric pressure to distill the compound 48b.
  • Peak position of compound 48-1 3.301 min (chiral column: AS-H 150 ⁇ 4.6 mm, ID, 5 ⁇ m, mobile phase: A: CO2, B: ethanol (0.05% diethylamine), maintaining 5% B 0.5 Min, then the content of B increased from 5% gradient to 40% in 3.5 min, then 40% B 2.5 min, and finally 5% B 1.5 min flow rate: 3 mL/min, column temperature: 40 ° C).
  • the compound 53b (0.25 g, 1.04 mmol, 1 eq) was dissolved in hydrochloric acid / ethyl acetate (10.0 mL, 2M), and the reaction was stirred at 30 ° C for 15 hours. After completion of the reaction, the mixture was dried under reduced pressure to give 53%. step.
  • Peak position of compound 53-1 2.589 min (chiral column: AS-3 150 ⁇ 4.6 mm, mobile phase: A: CO2, B: methanol (0.05% diethylamine), flow rate: 25 mL/min, column temperature: 40 ° C).
  • Peak position of compound 53-2 5.877 min (chiral column: AS-3 150 ⁇ 4.6 mm, mobile phase: A: CO2, B: methanol (0.05% diethylamine), flow rate: 25 mL/min, column temperature: 40 ° C).
  • the labeled mTOR kinase was stably expressed in HEK-293 cells.
  • Ligand beads were blocked with excess biotin and washed with buffer (1% bovine serum albumin, 0.05% Tween 20 ml, 1 ml dithiothreitol) to wash away unbound ligand and non-specific binding Ligand
  • the 11 affinity beads were resuspended in buffer (1 x PBS, 0.05% Tween 20, 0.5 ⁇ m non-biotin affinity ligand) and incubated for 30 minutes at room temperature.
  • the compounds of the invention have significant and unexpected unexpected mTOR kinase inhibitory activity.
  • Luciferase in the Cell-Titer-Glo reagent uses fluorescein, oxygen and ATP as reaction substrates to produce oxidized fluorescein and release energy in the form of light. Since ATP is required for the luciferase reaction, the total amount of light produced by the reaction is proportional to the total amount of ATP in the cell viability.
  • MCF-7 cell line ATCC-CRL-22
  • HT-29 cell line ATCC-HTB-38
  • OE21 ECACC-96062201
  • NCI-N87 cell line ATCC-CRL-5822
  • Cell culture medium (RPMI 1640 medium (Invitrogen #1868546; 10% serum Invitrogen #1804958; L-glutamine 1 ⁇ , Invitrogen #1830863; double-resistant Hyclone #J170012))
  • Multi-purpose microplate reader (Envision Reader)
  • the mother liquor of the compound was 10 mM, and the compound was diluted with DMSO to have an initial concentration of 4 mM.
  • Compounds were added to the compound compound mother liquor plate, 9 ⁇ L per well.
  • the cells were centrifuged at 1000 rpm for 1 min, and the cell plates were placed in a 37 ° C, 5% CO 2 incubator for 3 days.
  • test results are shown in Table 2
  • Table 2 Screening test results of in vitro cell proliferation inhibitory activity of the compound of the present invention
  • the compound of the present invention has obvious inhibitory activity on proliferation of MCF-7, N87 and OE-21 cells, and has certain proliferation inhibitory activity on HT-29 cells.
  • test compound was mixed with 5% DMSO/95% 10% polyoxyethylene castor oil (Cremophor EL), vortexed and sonicated to prepare an approximately clear solution of 1 mg/mL, and the microporous membrane was filtered and used.
  • 18 to 20 grams of Balb/c female mice were selected and administered as a candidate compound solution at a dose of 1 or 2 mg/kg.
  • the test compound was mixed with 1% Tween 80, 9% polyethylene glycol 400, 90% aqueous solution, vortexed and sonicated to prepare an approximate clear solution of 1 mg/mL, and the microporous membrane was filtered and used.
  • test compound exhibits the same or even better pharmacokinetic properties as the reference compound; the bioavailability of the compound of the present invention is close to 100%, which is a good developable orally administered molecule.
  • mice female BALB/c nude mice, 6-8 weeks old, weighing 18-22 g; supplier: Shanghai Xipuer-Beikai Experimental Animal Co., Ltd. Experimental methods and procedures:
  • Human breast cancer MCF-7 cells (ECACC, Cat. No. 86012803) were cultured in vitro in a single layer with EMEM (EBSS) + 2 mM glutamic acid + 1% Non Essential Amino Acids (NEAA) medium plus 10% fetal bovine serum. 100 U/mL penicillin and 100 ⁇ g/mL streptomycin were cultured in a 37 ° C 5% CO 2 incubator. Passage was routinely digested with trypsin-EDTA twice a week. When the cell saturation is 80%-90%, when the number reaches the requirement, the cells are collected, counted, and inoculated.
  • EBSS EMEM
  • NEAA Non Essential Amino Acids
  • estrogen tablets (0.18 mg) were inoculated into the left back of each mouse.
  • 0.2 mL (1 ⁇ 10 7 ) MCF-7 cells (with matrigel, volume ratio of 1:1) were subcutaneously inoculated into the right back of each mouse, and the group administration was started when the average tumor volume reached 142 mm 3 .
  • test compound was formulated into a clear solution of 0.75 mg/mL, 1.5 mg/mL, and 3 mg/mL, and the vehicle was 5% DMSO + 30% polyethylene glycol 300 + 65% water.
  • the experimental indicator is to investigate whether tumor growth is inhibited, delayed or cured.
  • Tumor diameters were measured twice a week using vernier calipers.
  • TGI (%) [1- (mean tumor volume at the end of administration of a treatment group - mean tumor volume at the start of administration of the treatment group) / (mean tumor volume at the end of treatment of the solvent control group - The mean tumor volume at the start of treatment in the solvent control group)] ⁇ control group.
  • T/C% TRTV/CRTV group started treatment (TRTV: treatment group RTV; CRTV: negative control group RTV).
  • T weight and C weight respectively indicate the tumor weight of the drug-administered group and the vehicle control group.
  • Statistical analysis included mean and standard error (SEM) of tumor volume at each time point for each group.
  • the treatment group showed the best therapeutic effect on the 21st day after the administration at the end of the trial, and therefore statistical analysis was performed based on this data to evaluate the difference between the groups.
  • T-test was used for comparison between the two groups, and one-way ANOVA was used for comparison between three groups or groups. If there was a significant difference in F values, the Games-Howell method was used for the test. If there is no significant difference in F values, the Dunnet (2-sided) method is used for analysis. All data analysis was performed with SPSS 17.0. A significant difference was considered at p ⁇ 0.05.
  • the c.p value is based on the tumor volume.
  • AZD2014 was significantly different from the solvent control group at 15 mg/kg and Example 23 at a dose of 30 mg/kg, with a TGI of 104% and 98%.

Abstract

本发明公开了一系列吡啶并嘧啶类化合物,及其制备mTORC 1/2双激酶活性抑制剂相关药物中的应用,具体公开了式(IV)所示化合物、其互变异构体或其药学上可接受的盐在制备mTORC1/2双激酶活性抑制剂相关药物中的应用。

Description

作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物
相关申请的引用
本申请主张如下优先权:
CN201711080753.8,申请日2017-11-06;
CN201810136962.8,申请日2018-02-09;
CN201810661825.6,申请日2018-06-25。
技术领域
本发明涉及一系列吡啶并嘧啶类化合物,及其在制备mTORC1/2双激酶活性抑制剂相关药物中的应用,具体涉及式(IV)所示化合物、其互变异构体或其药学上可接受的盐在制备mTORC1/2双激酶活性抑制剂相关药物中的应用。
背景技术
肿瘤,特别是恶性肿瘤是目前危害人类健康的最严重一大类疾病之一,随着科技的进步和人们对肿瘤治疗研究的越来越深入,在肿瘤的发生,发展机制上和肿瘤的治疗方面取得了飞速的进展。很多新的机制和生物标着物被发现。本发明涉及到一条对肿瘤的增殖、侵润转移和抗凋亡起关键作用的信号通路,即磷脂酰肌醇3激酶(PI3K)-AKT-哺乳动物雷帕霉素蛋白酶mTOR信号通路。
PI3K的活化很大程度上参与到靠近其质膜内侧的底物。多种生长因子和信号传导复合物,包括成纤维细胞生长因子(FGF)、血管内皮生长因子(VEGF)、人生长因子(HGF)、血管位蛋白I(Ang1)和胰岛素都能启始PI3K的激活过程。这些因子激活受体酪氨酸激酶(RTK),从而引起自磷酸化。PI3K激活的结果是在质膜上产生第二信使PIP3,PIP3与细胞内含有PH结构域的信号蛋白AKT和PDK1(phosphoinositide dependent kinase-1)结合,促使PDK1磷酸化AKT蛋白的Ser308导致AKT活化。其它PDK1的底物还包括PKC(蛋白激酶C)、S6K(p70S6)和SGK(serum/glucocorticoid regulated kinases)。AKT,亦称为蛋白激酶B(PKB),是PI3K下游主要的效应物。活化的AKT通过磷酸化多种酶、激酶和转录因子等下游因子,进而调节细胞的功能。AKT通过下游多种途径对靶蛋白进行磷酸化而发挥抗凋亡作用。PTEN(phosphatase and tensin homology deleted on chromosome 10),是一种抑癌基因,在广泛的人类肿瘤中发生基因突变或缺失。PTEN是一个PIP3-磷酸酶,与PI3K的功能相反,它可以通过去磷酸化将PIP3转变为PIP2。PTEN可减少AKT的活化而阻止所有由AKT调控的下游信号传导事件。mTOR作为AKT下游底物,进化上相对保守,可整合营养、能量及生长因子的多种信号,参与基因的转录、蛋白质翻译、核糖体合成和细胞凋亡等生物过程,在细胞生长中发挥极为重要的作用。其有两种高度同源的复合物,Tor与KOG01结合形成mTORC1,mTOR与AVO1/AVO2/AVO3/和LST8形成对雷帕霉素不敏感的mTORC2.mTOR通过磷酸化下游靶蛋白S40S核糖体S6蛋白激酶,比如S6K1和4EBP1来调节下游蛋白翻译。mTOR与eIF3结合,磷酸化S6K1,使S6K1 从eIF3上释放而被活化,进一步磷酸化细胞底物,如p70S6促进蛋白质翻译及表达。4EBP1与真核转录启动因子4E结合并抑制其活性,当mtor磷酸化4E-BP1后,使其活化与eif-4e分离,实现真核细胞转录。mTORC2能磷酸化AKT,从而上调其激酶活性。
由上可见,PI3K/AKT/mTOR信号通路上游出现任何的突变或过度表达,都会导致下游一系列级联反应,最终导致肿瘤的发生,发展和转移。而mTOR处于信号通路的枢纽,对mTORC1和mTORC2的抑制能很好的阻断信号的传递,从而达到控制肿瘤的发展。
研究发现,此信号通路在多种实体瘤,如乳腺癌、***癌、肺癌、结肠癌、胰腺癌、肝癌、胃癌、结直肠癌、肾癌、甲状腺癌、脑膜炎癌和急慢性淋巴细胞白血病,梅克尔细胞瘤等。并且与治疗耐受和不良预后紧密相关。由此可见,通过开发细分子化合物实现对PI3K/AKT/MTOR的信号通路的抑制,具有良好的开发前景。
本发明旨在发现一种双mTOR小分子化合物靶向药物,此类化合物具有良好的活性,并表现出了优异的效果和作用。
US20170281637公开化合物AZD2014,属于mTORC1&mTORC2激酶抑制剂,其结构式如下所示:
Figure PCTCN2018113683-appb-000001
发明内容
本发明提供了式(IV)所示化合物、其药学上可接受的盐及其异构体,
Figure PCTCN2018113683-appb-000002
其中,
R 1为H;
R 2为Me;
或者,R 1、R 2和吗啉环上的N形成一个5~6元杂环烷基;
R 3选自NH 2
Figure PCTCN2018113683-appb-000003
C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-,其中所述C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-任选被1、2或3个R取代;
n选自1和2;
环A选自苯基和6~10元杂芳基;
R 4选自H;
R 5选自H;
或者,R 4与R 5连接在一起形成一个5~6元杂环烷基;
D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-和-O-,且D 1、D 2、D 3和D 4至少一个不为单键,其中所述-CH 2-或-CH 2CH 2-任选被1或2个R取代;
D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-和-NH-,且D 5、D 6、D 7和D 8至少一个不为单键,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
T 1选自CH和N;
T 2选自-CH 2-、-NH-、-O-、
Figure PCTCN2018113683-appb-000004
-S-和-C(=O)NH-,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
R分别独立地选自F、Cl、Br、I、OH、NH 2、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基,其中所述C 1-3烷基、C 1-3烷氧基和C 3-6环烷基任选被1或2个R’取代;
R’分别独立地选自F、Cl、Br、I、OH和NH 2
所述C 1-3杂烷基、5~6元杂芳基和6~10元杂芳基分别包含1、2或3个独立选自-O-、-S-、-NH-、N、-C(=O)-、-C(=O)NH-和-C(=S)NH-的杂原子或杂原子团。
本发明提供上述化合物、其药学上可接受的盐及其异构体,其选自
Figure PCTCN2018113683-appb-000005
其中,
R 1为H;
R 2为Me;
或者,R 1、R 2和吗啉环上的N形成一个5~6元杂环烷基;
R 3选自NH 2
Figure PCTCN2018113683-appb-000006
C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-,其中所述C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-任选被1、2或3个R取代;
环A选自苯基和6~10元杂芳基;
R 4选自H;
R 5选自H;
或者,R 4与R 5连接在一起形成一个5~6元杂环烷基;
D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-和-O-,且D 1、D 2、D 3和D 4至少一个不为单键,其中所述-CH 2-或-CH 2CH 2-任选被1或2个R取代;
D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-和-NH-,且D 1、D 2、D 3和D 4至少一个不为单键,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
T 1选自CH和N;
T 2选自-CH 2-、-NH-、-O-、
Figure PCTCN2018113683-appb-000007
-S-和-C(=O)NH-,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
R分别独立地选自F、Cl、Br、I、OH、NH 2、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基,其中所述C 1-3烷基、C 1-3烷氧基和C 3-6环烷基任选被1或2个R’取代;
R’分别独立地选自F、Cl、Br、I、OH和NH 2
所述C 1-3杂烷基、5~6元杂芳基和6~10元杂芳基分别包含1、2或3个独立选自-O-、-S-、-NH-、N、-C(=O)-、-C(=O)NH-和-C(=S)NH-的杂原子或杂原子团。
本发明提供了式(Ⅰ)所示化合物、其药学上可接受的盐及其异构体,
Figure PCTCN2018113683-appb-000008
其中,
R 1选自H;
R 2选自Me;
或者,R 1与R 2连接在一起,形成一个5~6元杂环烷基;
R 3选自NH 2
Figure PCTCN2018113683-appb-000009
C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-,其中所述C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-任选被1、2或3个R取代;
环A选自苯基和6~10元杂芳基;
R 4选自H;
R 5选自H;
D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-和-O-,其中所述-CH 2-或-CH 2CH 2-任选被1或2个R取代;
D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-和-NH-,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
T 1选自CH和N;
T 2选自单键、CH 2和-O-;
R分别独立地选自F、Cl、Br、I、OH、NH 2、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基,其中所述C 1-3烷基、C 1-3烷氧基和C 3-6环烷基任选被1或2个R’取代;
R’分别独立地选自F、Cl、Br、I、OH和NH 2
所述C 1-3杂烷基、5~6元杂芳基和6~10元杂芳基分别包含1、2或3个独立选自-O-、-S-、-NH-、N、-C(=O)-、-C(=O)NH-和-C(=S)NH-的杂原子和杂原子团。
本发明的一些方案中,上述R分别独立地选自F、Cl、Br、I、OH、NH 2、Me、Et、
Figure PCTCN2018113683-appb-000010
其中所述Me、E、
Figure PCTCN2018113683-appb-000011
任选被1或2个R’取代,其它变量如本发明所定义。
本发明的一些方案中,上述R分别独立地选自F、Cl、Br、I、OH、NH 2、Me、CF 3、E、
Figure PCTCN2018113683-appb-000012
t和
Figure PCTCN2018113683-appb-000013
其它变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018113683-appb-000014
选自
Figure PCTCN2018113683-appb-000015
其它变量如本发明所定义。
本发明的一些方案中,上述R 1与R 2连接在一起,结构单元
Figure PCTCN2018113683-appb-000016
选自
Figure PCTCN2018113683-appb-000017
其它变量如本发 明所定义。
本发明的一些方案中,上述R 3选自NH 2
Figure PCTCN2018113683-appb-000018
Figure PCTCN2018113683-appb-000019
1H-吡唑基和1H-1,2,4-***基,其中所述NH 2
Figure PCTCN2018113683-appb-000020
Figure PCTCN2018113683-appb-000021
1H-吡唑基和1H-1,2,4-***基任选被1、2或3个R取代,其它变量如本发明所定义
本发明的一些方案中,上述R 3选自NH 2
Figure PCTCN2018113683-appb-000022
1H-吡唑基和1H-1,2,4-***基,其中所述NH 2
Figure PCTCN2018113683-appb-000023
1H-吡唑基和1H-1,2,4-***基任选被1、2或3个R取代,其它变量如本发明所定义。
本发明的一些方案中,上述R 3选自NH 2
Figure PCTCN2018113683-appb-000024
Figure PCTCN2018113683-appb-000025
其中所述NH 2
Figure PCTCN2018113683-appb-000026
Figure PCTCN2018113683-appb-000027
任选被1、2或3个R取代,其它变量如本发明所定义。
本发明的一些方案中,上述R 3选自NH 2
Figure PCTCN2018113683-appb-000028
Figure PCTCN2018113683-appb-000029
其中所述NH 2
Figure PCTCN2018113683-appb-000030
任选被1、2或3个R取代,其它变量如本发明所定义。
本发明的一些方案中,上述R 3选自NH 2
Figure PCTCN2018113683-appb-000031
Figure PCTCN2018113683-appb-000032
其它变量如本发明所定义。
本发明的一些方案中,上述R 3选自NH 2
Figure PCTCN2018113683-appb-000033
Figure PCTCN2018113683-appb-000034
其它变量如本发明所定义。
本发明的一些方案中,上述环A选自苯基、苯并[d]噁唑、喹啉基和喹唑啉基,其它变量如本发明所定 义。
本发明的一些方案中,上述环A选自
Figure PCTCN2018113683-appb-000035
Figure PCTCN2018113683-appb-000036
其它变量如本发明所定义。
本发明的一些方案中,上述环A选自
Figure PCTCN2018113683-appb-000037
Figure PCTCN2018113683-appb-000038
其它变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018113683-appb-000039
选自
Figure PCTCN2018113683-appb-000040
Figure PCTCN2018113683-appb-000041
Figure PCTCN2018113683-appb-000042
其它变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018113683-appb-000043
选自
Figure PCTCN2018113683-appb-000044
Figure PCTCN2018113683-appb-000045
Figure PCTCN2018113683-appb-000046
其它变量如本发明所定义。
本发明的一些方案中,上述D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-、-O-和
Figure PCTCN2018113683-appb-000047
且D 1、D 2、D 3和D 4至少一个不为单键,其它变量如本发明所定义。
本发明的一些方案中,上述D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-、-NH-、
Figure PCTCN2018113683-appb-000048
Figure PCTCN2018113683-appb-000049
且D 5、D 6、D 7和D 8至少一个不为单键,其它变量如本发明所定义。
本发明的一些方案中,其中结构单元
Figure PCTCN2018113683-appb-000050
选自
Figure PCTCN2018113683-appb-000051
Figure PCTCN2018113683-appb-000052
Figure PCTCN2018113683-appb-000053
其它变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018113683-appb-000054
选自
Figure PCTCN2018113683-appb-000055
Figure PCTCN2018113683-appb-000056
Figure PCTCN2018113683-appb-000057
其它变量如本发明所定义。
本发明的一些方案中,上述化合物、其药学上可接受的盐及其异构体,其选自:
Figure PCTCN2018113683-appb-000058
其中,
R 1、R 2、R 3、R 4和R 5如上述所定义,X选自-CH 2-、-NH-、-O-、
Figure PCTCN2018113683-appb-000059
和-S-。
本发明还有一些方案是由上述变量任意组合而来。
本发明还提供了下式所示化合物、其药学上可接受的盐及其异构体,
Figure PCTCN2018113683-appb-000060
Figure PCTCN2018113683-appb-000061
Figure PCTCN2018113683-appb-000062
Figure PCTCN2018113683-appb-000063
本发明还提供了一种药物组合物,包括治疗有效量的上述化合物、其药学上可接受的盐及其异构体作为活性成分以及药学上可接受的载体。
本发明还提供了上述化合物、其药学上可接受的盐及其异构体或上述的药物组合物在制备治疗与mTOR双激酶相关疾病的药物中的应用。
本发明还提供了上述化合物、其药学上可接受的盐及其异构体或上述组合物在制备治疗乳腺癌,乳腺癌,头颈癌,结直肠癌药物中的应用。
技术效果:
本发明化合物的进行mTORC1/2激酶活性测试,数据显示本发明化合物具有显著甚至意料不到的mTOR激酶抑制活性,优于目前的临床化合物AZD2014。
本发明化合物对MCF-7,N87和OE-21细胞增殖抑制活性明显,对HT-29细胞具有一定的增殖抑制活性。
PK结果显示,本发明的化合物生物利用度接近100%,是很好的可开发的口服给药的分子。
在MCF-7移植瘤模型上,部分化合物药效与AZD2014相当,本专利发明的化合物有成为多种肿瘤抑制剂之潜力。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和 反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,用楔形实线键
Figure PCTCN2018113683-appb-000064
和楔形虚线键
Figure PCTCN2018113683-appb-000065
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2018113683-appb-000066
和直形虚线键
Figure PCTCN2018113683-appb-000067
表示立体中心的相对构型,用波浪线
Figure PCTCN2018113683-appb-000068
表示楔形实线键
Figure PCTCN2018113683-appb-000069
或楔形虚线键
Figure PCTCN2018113683-appb-000070
或用波浪线
Figure PCTCN2018113683-appb-000071
表示直形实线键
Figure PCTCN2018113683-appb-000072
和直形虚线键
Figure PCTCN2018113683-appb-000073
本发明的化合物可以存在特定的。除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括一些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮两个互变异构体之间的互变。
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然 后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。术语“药学上可接受的载体”是指能够递送本发明有效量活性物质、不干扰活性物质的生物活性并且对宿主或者患者无毒副作用的任何制剂或载体介质代表性的载体包括水、油、蔬菜和矿物质、膏基、洗剂基质、软膏基质等。这些基质包括悬浮剂、增粘剂、透皮促进剂等。它们的制剂为化妆品领域或局部药物领域的技术人员所周知。
“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。当一个取代基可以连接到一个环上的一个以上原子时,这种取代基可以与这个环上的任意原子相键合,例如,结构单元
Figure PCTCN2018113683-appb-000074
表示取代基R可在环己基或者环己二烯上的任意一个位置发生取代。当所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,吡啶基作为取代基可以通过吡啶环上任意一个碳原子连接到被取代的基团上。当所列举的连接基团没有指明其连接方向,其连接方向是任意的,例如,
Figure PCTCN2018113683-appb-000075
中连 接基团L为-M-W-,此时-M-W-既可以按与从左往右的读取顺序相同的方向连接环A和环B构成
Figure PCTCN2018113683-appb-000076
也可以按照与从左往右的读取顺序相反的方向连接环A和环B构成
Figure PCTCN2018113683-appb-000077
所述连接基团、取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
除非另有规定,“杂环基”意指稳定的含杂原子或杂原子团的单环、双环或三环,它们是饱和的,它们包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子。氮和硫杂原子可任选被氧化(即NO和S(O) p,p是1或2)。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。该杂环可以附着到任何杂原子或碳原子的侧基上从而形成稳定的结构。如果产生的化合物是稳定的,本文所述的杂环可以发生碳位或氮位上的取代。杂环中的氮原子任选地被季铵化。一个优选方案是,当杂环中S及O原子的总数超过1时,这些杂原子彼此不相邻。另一个优选方案是,杂环中S及O原子的总数不超过1。如本文所用,术语“杂芳基”意指稳定的5、6、7元单环或双环或7、8、9或10元双环杂环基的芳香环,它包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。值得注意的是,芳香杂环上S和O原子的总数不超过1。当一个或多个原子(即C、O、N或S)连接两个不相邻的碳原子或氮原子时形成桥环。优选的桥环包括但不限于:一个碳原子、两个碳原子、一个氮原子、两个氮原子和一个碳-氮基。值得注意的是,一个桥总是将单环转换成三环。桥环中,环上的取代基也可以出现在桥上。
除非另有规定,术语“5-6元杂环烷基”本身或者与其他术语联合分别表示由5至6个环原子组成的饱和环状基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子,其中氮原子任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O) p,p是1或2)。其包括单环和双环体系,其中双环体系包括螺环、并环和桥环。此外,就该“5-6元杂环烷基”而言,杂原子可以占据杂环烷基与分子其余部分的连接位置。所述5-6元杂环烷基包括5元和6元杂环烷基。5-6元杂环烷基的实例包括但不限于吡咯烷基、吡唑烷基、咪唑烷基、四氢噻吩基(包括四氢噻吩-2-基和四氢噻吩-3-基等)、四氢呋喃基(包括四氢呋喃-2-基等)、四氢吡喃基、哌啶基(包括1-哌啶基、2-哌啶基和3-哌啶基等)、哌嗪基(包括1-哌嗪基和2-哌嗪基等)、吗啉基(包括3-吗啉基和4-吗啉基等)、二噁烷基、二噻烷基、异噁唑烷基、异噻唑烷基、1,2-噁嗪基、1,2-噻嗪基、六氢哒嗪基、高哌嗪基或高哌啶基等。
除非另有规定,本发明术语“6-10元杂芳环”和“6-10元杂芳基”可以互换使用,术语“6-10元杂芳基”是表示由6至10个环原子组成的具有共轭π电子体系的环状基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子。其可以是单环、稠合双环或稠合三环体系,其中各个环均为芳香性的。 其中氮原子任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O) p,p是1或2)。6-10元杂芳基可通过杂原子或碳原子连接到分子的其余部分。所述6-10元杂芳基包括6-8元、6-7元、6-9元、6元和10元杂芳基等。所述6-10元杂芳基的实例包括但不限于呋喃基(包括2-呋喃基和3-呋喃基等)、吡啶基(包括2-吡啶基、3-吡啶基和4-吡啶基等)、吡嗪基、嘧啶基(包括2-嘧啶基和4-嘧啶基等)、苯并噻唑基(包括5-苯并噻唑基等)、嘌呤基、苯并咪唑基(包括2-苯并咪唑基等)、苯并噁唑基、吲哚基(包括5-吲哚基等)、异喹啉基(包括1-异喹啉基和5-异喹啉基等)、喹喔啉基(包括2-喹喔啉基和5-喹喔啉基等)或喹啉基(包括3-喹啉基和6-喹啉基等)。
除非另有规定,本发明术语“5-6元杂芳环”和“5-6元杂芳基”可以互换使用,术语“5-6元杂芳基”表示由5至6个环原子组成的具有共轭π电子体系的单环基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子。其中氮原子任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O) p,p是1或2)。5-6元杂芳基可通过杂原子或碳原子连接到分子的其余部分。所述5-6元杂芳基包括5元和6元杂芳基。所述5-6元杂芳基的实例包括但不限于吡咯基(包括N-吡咯基、2-吡咯基和3-吡咯基等)、吡唑基(包括2-吡唑基和3-吡唑基等)、咪唑基(包括N-咪唑基、2-咪唑基、4-咪唑基和5-咪唑基等)、噁唑基(包括2-噁唑基、4-噁唑基和5-噁唑基等)、***基(1H-1,2,3-***基、2H-1,2,3-***基、1H-1,2,4-***基和4H-1,2,4-***基等)、四唑基、异噁唑基(3-异噁唑基、4-异噁唑基和5-异噁唑基等)、噻唑基(包括2-噻唑基、4-噻唑基和5-噻唑基等)、呋喃基(包括2-呋喃基和3-呋喃基等)、噻吩基(包括2-噻吩基和3-噻吩基等)、吡啶基(包括2-吡啶基、3-吡啶基和4-吡啶基等)、吡嗪基或嘧啶基(包括2-嘧啶基和4-嘧啶基等)。
除非另有规定,“C 3-6环烷基”表示由3至6个碳原子组成的饱和环状碳氢基团,其为单环和双环体系,所述C 3-6环烷基包括C 3-5、C 4-5和C 5-6环烷基等;其可以是一价、二价或者多价。C 3-6环烷基的实例包括,但不限于,环丙基、环丁基、环戊基、环己基等。
在一些实施例中,术语“杂烷基”本身或者与另一术语联合表示稳定的直链的、支链的烃原子团或其组合物,有一定数目的碳原子和至少一个杂原子组成。在一个典型实施例中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。杂原子或杂原子团可以位于杂烃基的任何内部位置,包括该烃基附着于分子其余部分的位置,但术语“烷氧基”、“烷氨基”和“烷硫基”(或硫代烷氧基)属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些烷基基团。实例包括但不限于-CH 2-CH 2-O-CH 3、-CH 2-CH 2-NH-CH 3、-CH 2-CH 2-N(CH 3)-CH 3、-CH 2-S-CH 2-CH 3、-CH 2-CH 2、-S(O)-CH 3、-CH 2-CH 2-S(O) 2-CH 3。至多两个杂原子可以是连续的,例如-CH 2-NH-OCH 3
除非另有规定,术语“烷基”用于表示直链或支链的饱和烃基,可以是单取代(如-CH 2F)或多取代的(如-CF 3),可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。烷基的例子包括甲基(Me),乙基(Et),丙基(如,n-丙基和异丙基),丁基(如,n-丁基,异丁基,s-丁基,t-丁基),戊基(如,n-戊基,异戊基,新戊基)等。
除非另有规定,术语“C 1-3烷基”用于表示直链或支链的由1至3个碳原子组成的饱和碳氢基团。所述C 1-3烷基包括C 1-2和C 2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C 1- 3烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)等。
除非另有规定,术语“C 1-3烷氧基”表示通过一个氧原子连接到分子的其余部分的那些包含1至3个碳原子的烷基基团。所述C 1-3烷氧基包括C 1-2、C 2-3、C 3和C 2烷氧基等。C 1-3烷氧基的实例包括但不限于甲氧基、乙氧基、丙氧基(包括正丙氧基和异丙氧基)等。
除非另有规定,环烷基包括任何稳定的环状或多环烃基,任何碳原子都是饱和的,可以是单取代或多取代的,可以是一价、二价或者多价。这些环烷基的实例包括,但不限于,环丙基、降冰片烷基、[2.2.2]二环辛烷、[4.4.0]二环癸烷等。
除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。此外,术语“卤代烷基”意在包括单卤代烷基和多卤代烷基。例如,术语“卤代(C 1-C 4)烷基”意在包括但不仅限于三氟甲基、2,2,2-三氟乙基、4-氯丁基和3-溴丙基等等。除非另有规定,卤代烷基的实例包括但不仅限于:三氟甲基、三氯甲基、五氟乙基,和五氯乙基。
“烷氧基”代表通过氧桥连接的具有特定数目碳原子的上述烷基,除非另有规定,C 1-6烷氧基包括C 1、C 2、C 3、C 4、C 5和C 6的烷氧基。烷氧基的例子包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基和S-戊氧基。
除非另有规定,术语“芳基”表示多不饱和的芳族烃取代基,可以是单取代或多取代的,可以是一价、二价或者多价,它可以是单环或多环(比如1至3个环;其中至少一个环是芳族的),它们稠合在一起或共价连接。术语“杂芳基”是指含有一至四个杂原子的芳基(或环)。在一个示范性实例中,杂原子选自B、N、O和S,其中氮和硫原子任选地被氧化,氮原子任选地被季铵化。杂芳基可通过杂原子连接到分子的其余部分。芳基或杂芳基的非限制性实施例包括苯基、萘基、联苯基、吡咯基、吡唑基、咪唑基、吡嗪基、噁唑基、苯基-噁唑基、异噁唑基、噻唑基、呋喃基、噻吩基、吡啶基、嘧啶基、苯并噻唑基、嘌呤基、苯并咪唑基、吲哚基、异喹啉基、喹喔啉基、喹啉基、1-萘基、2-萘基、4-联苯基、1-吡咯基、2-吡咯基、3-吡咯基、3-吡唑基、2-咪唑基、4-咪唑基、吡嗪基、2-噁唑基、4-噁唑基、2-苯基-4-噁唑基、5-噁唑基、3-异噁唑基、4-异噁唑基、5-异噁唑基、2-噻唑基、4-噻唑基、5-噻唑基、2-呋喃基、3-呋喃基、2-噻吩基、3-噻吩基、2-吡啶基、3-吡啶基、4-吡啶基、2-嘧啶基、4-嘧啶基、5-苯并噻唑基、嘌呤基、2-苯并咪唑基、5-吲哚基、1-异喹啉基、5-异喹啉基、2-喹喔啉基、5-喹喔啉基、3-喹啉基和6-喹啉基。上述任意一个芳基和杂芳基环系的取代基选自下文所述的可接受的取代基。
除非另有规定,C n-n+m或C n-C n+m包括n至n+m个碳的任何一种具体情况,例如C 1-12包括C 1、C 2、C 3、C 4、C 5、C 6、C 7、C 8、C 9、C 10、C 11、和C 12,也包括n至n+m中的任何一个范围,例如C 1-12包括C 1- 3、C 1-6、C 1-9、C 3-6、C 3-9、C 3-12、C 6-9、C 6-12、和C 9-12等;同理,n元至n+m元表示环上原子数为n至n+m 个,例如3-12元环包括3元环、4元环、5元环、6元环、7元环、8元环、9元环、10元环、11元环、和12元环,也包括n至n+m中的任何一个范围,例如3-12元环包括3-6元环、3-9元环、5-6元环、5-7元环、6-7元环、6-8元环、和6-10元环等。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明的化合物可以有多种用途或适应症,包括但不限于本申请所列举的具体用途或适应症。
本发明所使用的溶剂可经市售获得。
本发明各实施例涉及高效液相色谱分离的均采用中性分离。
本发明采用下述缩略词:aq代表水;HATU代表O-(7-氮杂苯并***-1-基)-N,N,N',N'-四甲基脲六氟磷酸盐;EDC代表N-(3-二甲基氨基丙基)-N'-乙基碳二亚胺盐酸盐;m-CPBA代表3-氯过氧苯甲酸;eq代表当量、等量;CDI代表羰基二咪唑;DCM代表二氯甲烷;PE代表石油醚;DIAD代表偶氮二羧酸二异丙酯;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;EtOH代表乙醇;MeOH代表甲醇;CBz代表苄氧羰基,是一种胺保护基团;BOC代表叔丁基羰基是一种胺保护基团;HOAc代表乙酸;NaCNBH 3代表氰基硼氢化钠;r.t.代表室温;O/N代表过夜;THF代表四氢呋喃;Boc 2O代表二-叔丁基二碳酸酯;TFA代表三氟乙酸;DIPEA代表二异丙基乙基胺;SOCl 2代表氯化亚砜;CS 2代表二硫化碳;TsOH代表对甲苯磺酸;NFSI代表N-氟-N-(苯磺酰基)苯磺酰胺;n-Bu 4NF代表氟化四丁基铵;iPrOH代表2-丙醇;mp代表熔点;LDA代表二异丙基胺基锂;Pd(PPh 3) 4代表四(三苯基膦)钯;IV代表静脉注射;PO代表口服。
本发明化合物依据本领域常规命名原则或者
Figure PCTCN2018113683-appb-000078
软件命名,市售化合物采用供应商目录名称。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
实施例1
Figure PCTCN2018113683-appb-000079
Figure PCTCN2018113683-appb-000080
第一步
将化合物1a(20.0g,104mmol,1.00eq)和浓氨水(200mL,1.45mol,14.0eq)密闭于高压釜中,130℃下搅拌24小时,压力约为0.9MPa。将反应液浓缩,得到化合物1b。
MS-ESI计算值[M+H] +173和175,实测值173和175。
1H NMR(400MHz,DMSO-d 6)δ:8.03(d,J=8.0Hz,1H),7.56(br s,2H),6.61(d,J=8.0Hz,1H)。
第二步
将化合物1b(17.0g,98.5mmol,1.00eq)、氯化铵(10.5g,197mmol,2.00eq)、1-羟基苯并***(13.3g,98.5mmol,1.00eq)、1-(3-二甲胺丙基)-3-乙基碳二亚胺盐酸盐(18.9g,98.5mmol,1.00eq)和二异丙基乙胺(38.2g,296mmol,3.00eq)溶于N,N-二甲基甲酰胺(200.0mL)中。混合物在20℃下搅拌16小时。反应完成后,溶剂减压旋干,加水(200mL),乙酸乙酯萃取(200mL×3),合并的有机相用无水硫酸钠干燥,过滤,柱层析(1:1石油醚/乙酸乙酯,R f=0.4),得到化合物,乙酸乙酯(50mL)打浆十分钟,得到化合物1c。
1H NMR(400MHz,DMSO-d 6)δ:7.96(d,J=8.0Hz,2H),7.62(br s,2H),7.40(br s,1H),6.61(d,J=8.0Hz,1H)。
第三步
将化合物1c(8.00g,46.6mmol,1.00eq)和草酰氯(7.1g,56.0mmol,4.9mL,1.00eq)依次加入甲苯(200mL)中。混合物在110℃下搅拌15小时。冷却至室温,过滤,干燥。得到化合物1d。
1H NMR(400MHz,DMSO-d 6)δ:8.24(d,J=8.0Hz,1H),7.30(d,J=8.0Hz,1H)。
第四步
将化合物1d(6.00g,30.4mmol,1.00eq)和二异丙基乙胺(11.8g,91.1mmol,15.9mL,3.00eq)依次加入甲苯(100mL)中。混合物在70℃下搅拌半小时。冷却至室温,将三氯氧磷(14.0g,91.1mmol, 8.5mL,3.00eq)滴入混合物中。混合物在100℃下搅拌2小时。冷却至室温,浓缩,柱层析(3:1石油醚/乙酸乙酯,Rf=0.4),得到化合物1e。
1H NMR(400MHz,DMSO-d 6)δ:8.45(d,J=8.0Hz,1H),7.63(d,J=8.3Hz,1H)。
第五步
将化合物1e(1.90g,8.10mmol,1.00eq)、(S)-2-甲基***啉(819mg,8.10mmol,1.00eq)和二异丙基乙胺(2.09g,16.2mmol,2.83mL,2.00eq)溶于二氯甲烷(50mL)中,所得溶液在25℃反应2小时。反应完成后,浓缩,柱层析(3:1石油醚/乙酸乙酯),得到化合物1f。
1H NMR(400MHz,DMSO-d 6)δ:8.47(d,J=8.8Hz,1H),7.55(d,J=8.8Hz,1H),4.71-4.72(m,1H),4.12-4.09(m,1H),3.92-3.91(m,1H),3.84-3.74(m,1H),3.73-3.64(m,2H),3.54-3.53(m,1H),1.46(d,J=6.8Hz,3H)。
第六步
将化合物1f(1.2g,4.01mmol,1.00eq)、化合物1g(1.15g,4.41mmol,1.10eq)、四三苯基膦钯(232mg,200μmol,0.05eq)和碳酸钾(1.66g,12.0mmol,3.00eq)溶于水(24mL)和1,4-二氧六环(120mL)中,在氮气保护下,60℃反应5小时,反应完成后,浓缩掉溶剂,加水(30mL)稀释后用乙酸乙酯萃取(50mL×2),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,柱层析(100%乙酸乙酯)得化合物1h。
1H NMR(400MHz,DMSO-d 6)δ:8.71(s,1H),8.67(d,J=4.8Hz,1H),8.55(d,J=8.8Hz,1H),8.39(d,J=8.0Hz,1H),8.14(d,J=8.8Hz,1H),8.01(d,J=8.0Hz,1H),7.68(t,J=7.6Hz,1H),4.75(d,J=6.4Hz,1H),4.17-4.15(m,1H),3.94-3.92(m,1H),3.87-3.77(m,1H),3.72(s,2H),3.59-3.57(m,1H),2.86-2.84(m,3H),1.49(d,J=6.8Hz,3H)。
第七步
将化合物1h(50mg,126μmol,1eq)、1i(18.8mg,126μmol,1eq)和DIPEA(16.2mg,126μmol,21.89μL,1eq)溶于DMSO(3mL),混合溶液在70℃反应20小时。反应完全,反应液经高效液相色谱法纯化得化合物1。
MS-ESI计算值[M+H] +475,实测值475。
1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.32-8.30(m,2H),7.96(d,J=7.2Hz,1H),7.72-7.61(m,2H),4.52(br s,1H),4.09-3.92(m,6H),3.91-3.82(m,3H),3.76-3.74(m,3H),2.99(s,3H),1.49(d,J=6.8Hz,3H),0.81(s,2H),0.71(s,2H)。
实施例2
Figure PCTCN2018113683-appb-000081
Figure PCTCN2018113683-appb-000082
将化合物1(40mg,84.3μmol,1.00eq)溶于130℃的二甲苯(2mL)中,然后将劳森试剂(37.5mg,92.7μmol,1.10eq)分批加入上述反应液中,氮气置换三次,然后在氮气保护在130℃下反应17小时,反应完成后,溶剂减压旋干,粗产品加水(10mL),二氯甲烷萃取(10mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,残留物经制备薄层色谱法分离(100%乙酸乙酯),此粗产品经制备液相色谱分离,得化合物2。
MS-ESI计算值[M+H] +491,实测值491。
1H NMR(400MHz,CDCl 3)δ:8.47(s,1H),8.15(s,1H),8.05(d,J=7.8Hz,1H),8.02-7.95(m,2H),7.47-7.36(m,2H),4.30(s,1H),4.04-3.83(m,5H),3.80-3.78(m,4H),3.73-3.60(m,3H),3.33(d,J=4.8Hz,3H),1.41(d,J=6.8Hz,3H),0.80-0.74(m,2H),0.60-0.59(m,2H)。
实施例3
Figure PCTCN2018113683-appb-000083
第一步
将化合物1f(0.5g,1.37mmol,1eq)、3a(246mg,1.37mmol,1eq)、四(三苯基膦)钯(79.2mg,68.53μmol,0.05eq)和无水碳酸钠(436mg,4.11mmol,3eq)溶于二氧六环(6.0mL)和水(2.0mL)中,氮气置换三次,然后在氮气保护70℃下反应3小时。反应结束后,溶剂减压旋干,剩余固体加水(20mL)、乙酸乙酯(10mL×3)稀释,过滤、分液,合并有机相,无水硫酸钠干燥,过滤旋干,柱层析(3:1石油醚/乙酸乙酯)得到化合物3b。
第二步
将化合物3b(180mg,375μmol,1eq)、1i(56.0mg,375μmol,1eq,HCl)和DIPEA(145mg,1.12mmol,196μL,3eq)溶于DMSO(3mL)中,然后在70℃下反应15小时。反应完成后,3c不做处理直接进行下一步反应。
第三步
将化合物3c(30.0mg,65.0μmol,1mL,1eq)、3d(10.9mg,130μmol,2eq,HCl)、DIPEA(25.2mg,195μmol,34.0μL,3eq)和HATU(49.4mg,130μmol,2eq)溶于DMSO(2mL)中,然后在27℃下反应20小时。反应完成后,经高效液相色谱法分离得到化合物3。
MS-ESI计算值[M+H] +491,实测值491。
1H NMR:(400MHz,CDCl 3)δ:9.29(s,1H),8.49(s,1H),8.13(br d,J=7.6Hz,1H),7.98(d,J=8.4Hz,1H),7.8(d,J=7.6Hz,1H),7.49(t,J=7.6Hz,1H),7.37-7.46(m,1H),4.22-4.37(m,1H),3.89-4.06(m,3H),3.75-3.89(m,9H),3.60-3.73(m,3H),1.41(d,J=6.4Hz,3H),0.74-0.81(m,2H),0.61(br s,2H)。
实施例4
Figure PCTCN2018113683-appb-000084
Figure PCTCN2018113683-appb-000085
第一步
将化合物4a(180mg,375μmol,1eq)、1i(56.0mg,375μmol,1eq,HCl)和DIPEA(145mg,1.12mmol,196μL,3eq)溶于DMSO(3mL),在70℃下反应15小时。反应完成后,得到化合物4b,直接进行下一步反应。
MS-ESI计算值[M+H] +476,实测值476。
第二步
将化合物4b(178mg,374.31μmol,1eq)和LiOH(23.56mg,561.47μmol,1.5eq)溶于二甲基亚砜(3mL)中,室温反应24小时,LC-MS监测发现没有得到化合物3。在反应液中补加氢氧化钠(29.95mg,748.63μmol,2eq),继续反应20小时。反应结束后,所得混合物直接经高效液相色谱法纯化得到化合物4c。
第三步
将化合物4c(20mg,42.9μmol,1eq)、4d(5.99mg,47.2μmol,4.13μL,1.1eq)和DMF(314μg,4.29μmol,0.33μL,0.1eq)溶于二氯甲烷(2mL)中,所得溶液在室温下反应1小时。反应完成后得到化合物4e,不做处理直接进行下一步反应。
第四步
将化合物4f(60mg,125.01μmol,1eq)加入到4e(86.9mg,1.25mmol,10eq,HCl)和DIPEA(194mg,1.50mmol,261μL,12eq)的二氯甲烷(2mL)溶液中,然后在室温下反应2小时。经高效液相色谱法纯化得到4。
MS-ESI计算值[M+H] +477,实测值477。
1H NMR(400MHz,CD 3OD)δ:8.57(s,1H),8.34-8.31(m,2H),7.90(d,J=7.6Hz,1H),7.57-7.78(m,2H),4.62-4.60(m,3H),4.04-4.02(m,2H),4.01-3.92(m,3H),3.87-3.85(m,3H),3.78-3.76(m,2H),1.51(d,J=6.8Hz,3H),0.93-0.61(m,4H)。
实施例5
Figure PCTCN2018113683-appb-000086
第一步
1h(300mg,754μmol,1.00eq)、5a(301mg,1.13mmol,1.50eq)、四三苯基膦钯(43.6mg,37.7μmol,0.05eq)和碳酸钠(240mg,2.26mmol,3.00eq)溶入水(3mL)和1,4-二氧六环(10mL)中,在氮气保护下,90℃反应16小时,反应完成后,浓缩掉溶剂,加水(10mL)稀释,乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,柱层析(100%乙酸乙酯,R f=0.4)得黄色固体(200mg)纯度63.8%,产率:27%。其中的40mg用制备高效液相色谱分离纯化得到化合物5b
MS-ESI计算值[M+H] +502,实测值502。
1H NMR(400MHz,CD 3OD)δ:8.70(s,1H),8.44(d,J=8.8Hz,1H),8.41(br d,J=8.0Hz,1H),8.02-7.92(m,2H),7.66(t,J=8.0Hz,1H),7.35-7.25(m,1H),4.68-4.66(m,1H),4.21-4.210(m,1H),4.05-3.96(m,5H),3.90-3.72(m,4H),3.00(s,3H),2.92(br s,2H),2.56(s,2H),1.94(t,J=6.4Hz,2H),1.53(d,J=6.4Hz,3H)。
第二步
将钯碳(10mg,含量10%,水分:50%)加入到5b(80mg,160μmol,1.00eq)的甲醇(10mL)溶液中,氢气置换三次。混合物在氢气氛围(15psi)和20℃下反应16小时。过滤,滤饼用10mL甲醇洗涤,滤液浓缩。粗品用制备高效液相色谱分离纯化得到化合物5。
MS-ESI计算值[M+H] +504,实测值504。
1H NMR(400MHz,CD 3OD)δ:8.73(s,1H),8.48(d,J=8.4Hz,1H),8.45(br d,J=8.0Hz,1H),8.02(d,J=8.4Hz,1H),7.98(br d,J=8.0Hz,1H),7.66(t,J=8.0Hz,1H),4.78-4.76(m,1H),4.26-4.24(m,1H),4.05-3.94(m,5H),3.89-3.70(m,4H),3.00(s,3H),2.94-2.82(m,1H),2.12-1.99(m,4H),1.92-1.90(m,2H),1.78-1.67(m,2H),1.57(d,J=6.8Hz,3H)。
实施例6
Figure PCTCN2018113683-appb-000087
第一步
将化合物1h(50.0mg,126μmol,1.00eq)、化合物6a(32.5mg,189μmol,1.50eq,0.5当量草酸盐),N,N-二异丙基乙胺(48.7mg,377μmol,3.00eq)溶于二甲亚砜(1.00mL)中,然后将反应液在70℃下搅拌16小时。反应完成后,用制备高效液相色谱分离纯化得到化合物6。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,DMSO-d 6)δ:8.64(s,2H),8.31(d,J=7.6Hz,1H),8.24(d,J=8.4Hz,1H),7.95(d,J=7.6Hz,1H),7.69(d,J=8.4Hz,1H),7.62(t,J=7.6Hz,1H),4.47-4.45(m,1H),3.95-3.82(m,6H),3.79-3.70(m,1H),3.69-3.50(m,7H),2.84(d,J=4.8Hz,3H),1.76(br t,J=4.8Hz,4H),1.38(d,J=6.8Hz,3H)。
实施例7
Figure PCTCN2018113683-appb-000088
Figure PCTCN2018113683-appb-000089
将化合物1h(50.0mg,126μmol,1.00eq)、化合物7a(32.5mg,189μmol,1.50eq,0.5当量草酸盐)和二异丙基乙胺(48.7mg,377μmol,3.00eq)溶于二甲亚砜(1.00mL)中,然后将反应液在70℃下搅拌16小时。反应完成后,用制备高效液相色谱分离纯化得到化合物7。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,DMSO-d 6)δ:8.70-8.60(m,2H),8.30(br d,J=8.0Hz,1H),8.22(d,J=8.4Hz,1H),7.95(d,J=8.0Hz,1H),7.68(d,J=8.4Hz,1H),7.62(t,J=8.0Hz,1H),4.47-4.27(m,5H),3.96-3.56(m,10H),2.84(d,J=4.4Hz,3H),1.83(br t,J=5.2Hz,4H),1.37(d,J=6.8Hz,3H)。
实施例8
Figure PCTCN2018113683-appb-000090
将化合物1h(20mg,50.3μmol,1.00eq)、8a(13.4mg,75.4μmol,1.50eq,HCl)和N,N-二异丙基乙基胺(19.5mg,151μmol,26.3μL,3.00eq)溶于二甲基亚砜(1mL),混合溶液在70℃反应40小时。反应完全后,反应液经高效液相色谱法分离得到化合物8。
MS-ESI计算值[M+H] +503,实测值503。
1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.33(d,J=8.0Hz,1H),8.26(d,J=8.0Hz,1H),7.95(d,J=8.0Hz,1H),7.67-7.61(m,2H),4.51(br d,J=7.03Hz,1H),4.13-4.05(m,2H),4.02-3.85(m,7H),3.82-3.69(m,3H),3.65(s,2H),2.99(s,3H),1.92(t,J=7.2Hz,2H),1.69(t,J=5.6Hz,4H),1.48(d,J=6.8Hz,3H)。
实施例9
Figure PCTCN2018113683-appb-000091
将化合物9a(100mg,175μmol,1eq)、1i(26.28mg,175.67μmol,1eq,HCl)和DIPEA(68.1mg,527μmol,91.8μL,3eq)溶于DMSO(5mL)中,然后在70℃下反应18小时。反应完成后,用水(10mL)将反应液稀释,然后用乙酸乙酯(15mL×5)萃取,分液,合并的有机相用无水硫酸钠干燥,过滤旋干,残留物经高效液相色谱法纯化得到化合物9。
MS-ESI计算值[M+H] +487,实测值487。
1H NMR(400MHz,CDCl 3)δ:8.63(s,1H),8.22(d,J=8.0Hz,1H),8.14(d,J=8.0Hz,1H),7.98(d,J=8.0Hz,1H),7.50-7.61(m,2H),6.56(s,1H),4.59(s,2H),3.69-4.23(m,10H),3.07(d,J=4.8Hz,3H),2.06-2.20(m,2H),1.98-2.06(m,2H),0.90(s,2H),0.69(s,2H)。
实施例10
Figure PCTCN2018113683-appb-000092
将化合物1h(50.0mg,126μmol,1.00eq)、10a(20.9mg,188μmol,1.50eq)和三乙胺(38.2mg,377μmol,52.3μL,3.00eq)溶于二甲亚砜(1.00mL)中,氮气置换三次,然后将反应液在70℃下,搅拌12小时,反应 完成后,经高效液相色谱法纯化得化合物10。
MS-ESI计算值[M+H] +473,实测值473。
1H NMR(400MHz,CD 3OD)δ:8.61(s,1H),8.32(d,J=8.0Hz,1H),8.23(d,J=8.4Hz,1H),7.95(d,J=8.0Hz,1H),7.59-7.66(m,2H),4.49-4.47(m,1H),4.00-4.07(m,4H),4.00-3.92(m,2H),3.89-3.87(m,1H),3.82-3.67(m,3H),2.99(s,3H),1.49-1.46(m,7H),0.43(s,4H)。
实施例11
Figure PCTCN2018113683-appb-000093
第一步
将化合物11a(1.0g,3.41mmol,1eq)、11b(510mg,3.41mmol,1eq,HCl)溶于无水二氯甲烷(80mL),然后加入DIPEA(441mg,3.41mmol,594μL,1eq),使混合溶液于20℃反应18小时。反应完全,减压浓缩,滤渣用制备薄层色谱法纯化(1:1石油醚/乙酸乙酯)得到化合物11c。
MS-ESI计算值[M+H] +311,312和313,实测值311,312和313。
1H NMR(400MHz,CDCl 3)δ:8.14(d,J=8.8Hz,1H),7.36(d,J=8.8Hz,1H),4.84(s,2H),3.97(d,J=11.2Hz,2H),3.83(d,J=11.2Hz,2H),2.24-2.16(m,2H),2.10-2.01(m,2H)。
第二步
将化合物11c(200mg,643μmol,1eq)、11d(167mg,643μmol,1eq)、K 2CO 3(266mg,1.93mmol,3eq)和Pd(PPh 3) 4(37.1mg,32.1μmol,0.05eq)溶于无水二氧六环(30mL)和水(6mL),氮气置换三次,使混合溶液在氮气环境下于90℃反应2小时。反应完全,将反应液减压浓缩后加水20mL,用乙酸乙酯(20mL×3)萃取,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,滤渣用板层析纯化(100%乙酸乙酯)得到化合物11e。
MS-ESI计算值[M+H] +409和411,实测值409和411。
第三步
将化合物11e(120mg,294μmol,1eq)、1i(33.2mg,222μmol,HCl)和DIPEA(37.9mg,294μmol,51.1μL,1eq)溶于DMSO(6mL),使混合溶液在氮气环境在于70℃反应17小时。反应完全,经高效液相色谱法纯化得到化合物11。
MS-ESI计算值[M+H] +486,实测值486。
1H NMR(400MHz,CD 3OD)δ:8.38(d,J=8.4Hz,1H),8.24(br s,1H),8.03(d,J=1.2Hz,1H),7.94-7.89(m,1H),7.71(d,J=8.4Hz,1H),7.43(d,J=8.4Hz,1H),4.83(s,1H),4.06-3.97(m,4H),3.94(s,2H),3.87(t,J=4.8Hz,2H),3.80(br d,J=10.4Hz,2H),2.17-2.00(m,4H),0.86-0.80(m,2H),0.76-0.70(m,2H)。
实施例12
Figure PCTCN2018113683-appb-000094
将化合物1h(80mg,201μmol,1eq)、12a(28.8mg,201μmol,25.7μL,1eq)、二异丙基乙胺(26.0mg,201μmol,35.0μL,1eq)溶于二甲基亚砜(4.00mL),使混合溶液在70℃反应1.5小时。反应完全,反应液经高效液相色谱法纯化得化合物12。
MS-ESI计算值[M+H] +505,实测值505。
1H NMR(400MHz,CD 3OD)δ:8.62-8.60(m,1H),8.31(d,J=8.0Hz,1H),8.24(d,J=8.4Hz,1H),7.95(d,J=8.0 Hz,1H),7.65-7.60(m,2H),4.50(br d,J=6.8Hz,1H),4.09-4.02(m,8H),4.00-3.93(m,2H),3.90-3.84(m,1H),3.81-3.68(m,3H),2.99(s,3H),1.80-1.74(m,4H),1.48(d,J=6.8Hz,3H)。
实施例13
Figure PCTCN2018113683-appb-000095
将化合物1h(50.0mg,126μmol,1eq)、13a(19.8mg,126μmol,1eq)和N,N-二异丙基乙胺(48.7mg,377μmol,65.7μL,3eq)溶于二甲基亚砜(2mL),在70℃下反应16小时。反应完成后,经高效液相色谱法纯化,得到化合物13。
MS-ESI[M+H]计算值:519,实测值519。
1H NMR(400MHz,CDCl 3)δ:8.63(s,1H),8.24(d,J=8.00Hz,1H),8.06(d,J=8.00Hz,1H),7.98(d,J=8.00Hz,1H),7.49-7.64(m,2H),6.52(br s,1H),4.40(br d,J=6.8Hz,1H),4.02(br d,J=10.8Hz,3H),3.83-3.94(m,6H),3.66-3.83(m,7H),3.08(d,J=4.8Hz,3H),1.80-1.93(m,2H),1.73(br s,2H),1.50(d,J=6.8Hz,3H)。
实施例14
Figure PCTCN2018113683-appb-000096
Figure PCTCN2018113683-appb-000097
第一步
将化合物14a(0.08g,359μmol,1eq)、频那醇双硼酸酯(109mg,430μmol,1.2eq)、Pd(dppf)Cl 2(10.5mg,14.4μmol,0.04eq)和乙酸钾(106mg,1.08mmol,3eq)溶于20mL的无水二氧六环,使混合溶液在氮气保护下于90℃反应22小时。反应完全。然后反应液浓缩得到14b。
MS-ESI计算值[M+H] +270,实测值270。
第二步
将化合物1f(0.1g,334μmol,1eq)、14b(99.3mg,367μmol,1.1eq)、四三苯基膦钯(19.3mg,16.7μmol,0.05eq)和碳酸钾(139mg,1.00mmol,3eq)溶于无水二氧六环(20mL)和水(4mL),使混合溶液在氮气保护下于70℃反应3.5小时。当反应完全,冷却后,向反应液中加入10mL水和60mL(20mL×3)的乙酸乙酯进行萃取。然后有机相用无水硫酸钠干燥、浓缩。柱层析分离得到14c。
MS-ESI计算值[M+H] +407,409实测值407,409。
第三步
将化合物14c(0.04g,93.4μmol,1eq)、1i(13.9mg,93.4μmol,1eq,HCl)和二异丙基胺(12.1mg,93.4μmol,16.3μL,1eq)溶于二甲基亚砜(3mL),使反应液于70℃下反应17小时。反应完全后,将反应液经高效液相色谱法纯化得化合物14。
MS-ESI计算值[M+H] +484,实测值484。
1H NMR(400MHz,CD 3OD)δ:8.30(d,J=8.4Hz,1H),8.21(s,1H),8.08(dd,J=1.6,8.4Hz,1H),8.01(d,J=9.2Hz,1H),7.80(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),6.90(d,J=8.8Hz,1H),4.52(br s,1H),4.07-3.94(m,6H),3.91-3.84(m,3H),3.80-3.72(m,3H),1.49(d,J=6.8Hz,3H),0.84-0.79(m,2H),0.72(s,2H)。
实施例15
Figure PCTCN2018113683-appb-000098
第一步
将化合物1h(70m g,176μmol,1eq)、15a(40.2mg,176μmol,1eq)和二异丙基乙胺(22.7mg,176μmol,30.7μL,1eq)溶于二甲基亚砜(5mL),使混合溶液在70℃反应17小时。反应完全,反应冷却后,向反应液中加入10mL水和30mL的乙酸乙酯进行萃取。然后向有机相中加入水萃取多余的二甲基亚砜,有机相用无水硫酸钠干燥,浓缩。板层析(0/1石油醚/乙酸乙酯),得到化合物15b。
MS-ESI计算值[M+H] +590,实测值590。
第二步
将化合物15b(100mg,169μmol,1eq)溶于乙酸乙酯(3mL),然后向上述溶液中加入盐酸/乙酸乙酯(4M,3mL,70.8eq),使反应液于20℃下反应3小时。反应完全后将反应液浓缩,然后加入水10mL和乙酸乙酯45mL(15mL×3)进行萃取,有机相用无水硫酸钠干燥,浓缩。取少量反应液经高效液相色谱法纯化得化合物15。
MS-ESI计算值[M+H] +490,实测值490。
1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.36-8.29(m,2H),7.96(d,J=7.8Hz,1H),7.69(d,J=8.4Hz,1H),7.64(t,J=7.8Hz,1H),4.62(br d,J=6.4Hz,1H),4.19-3.86(m,7H),3.81-3.72(m,5H),3.65(br d,J=9.2Hz,2H),3.53(br d,J=8.0Hz,2H),2.99(s,3H),1.51(d,J=6.8Hz,3H)。
实施例16
Figure PCTCN2018113683-appb-000099
第一步
将化合物16a(0.1g,446μmol,1eq),频那醇硼酸酯(136mg,536μmol,1.2eq),Pd(dppf)Cl 2(13.1mg,17.9μmol,0.04eq)和乙酸钾(131mg,1.34mmol,3eq)溶于10mL的无水二氧六环,使混合溶液在氮气保护下于90℃反应2.5小时。当质谱检测有30%的产物生成,停止反应。然后反应液浓缩得到化合物16b。
MS-ESI计算值[M+H] +272,实测值272。
第二步
将化合物1f(0.1g,334μmol,1eq)、16b(90.6mg,334μmol,1eq)、四三苯基膦钯(19.3mg,16.7μmol,0.05eq)和碳酸钾(139mg,1.00mmol,3eq)溶于无水二氧六环(20mL)和水(4mL),使混合溶液在氮气保护下于70℃反应15小时。当反应完全,冷却后,向反应液中加入10mL水和60mL(20mL×3)的乙酸乙酯进行萃取。然后有机相用无水硫酸钠干燥,浓缩。柱层析分离(甲醇/乙酸乙酯),得到化合物16c。
MS-ESI计算值[M+H] +408,410实测值408,410。
第三步
将化合物16c(0.1g,153μmol,1eq)、1i(22.9mg,153μmol,1eq,HCl)和二异丙基胺(19.7mg,153μmol,26.6μL,1eq)溶于二甲基亚砜(4mL),使反应液于70℃下反应15小时。反应完全后,将反应液经高效液相色谱法纯化得化合物16。
MS-ESI计算值[M+H] +485,实测值485。
1H NMR(400MHz,CD 3OD)δ:9.21(s,1H),8.61-8.55(m,2H),8.23(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.61(d,J=9.6Hz,1H),4.50(br d,J=6.8Hz,1H),4.04(q,J=4.4Hz,2H),4.00-3.92(m,4H),3.90-3.84(m,3H),3.80-3.69(m,3H),1.48(d,J=6.8Hz,3H),0.86-0.78(m,2H),0.75-0.69(m,2H)。
实施例17
Figure PCTCN2018113683-appb-000100
第一步
将化合物17a(100mg,448μmol,1.0eq),双联频那醇硼酸酯(125mg,493μmol,1.1eq),乙酸钾(132mg,1.34mmol,3.0eq)和Pd(dppf)Cl 2(16.4mg,22.4μmol,0.05eq)溶于无水二氧六环(5mL),换气三次,使混合溶液在氮气环境下于100℃反应17小时。反应完全后,将反应液直接用于下一步。
MS-ESI计算值[M+H] +271,实测值271。
第二步
往含有化合物17b的上述反应液中加入化合物17c(134mg,448μmol,1.0eq)、四(三苯基膦)钯(25.9mg,22.4μmol,0.05eq)和碳酸钠(143mg,1.34mmol,3.0eq),溶入水(2mL)和1,4-二氧六环(5mL)中,在氮气保护下,70℃反应4小时,反应完成后,浓缩掉溶剂,加水(10mL)稀释,乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,层析板分离(纯乙酸乙酯),得17d。
MS-ESI计算值[M+H] +407,409实测值407,409。
1H NMR(400MHz,CDCl 3)δ:8.60(s,1H),8.14(d,J=8.8Hz,1H),8.10(br d,J=7.8Hz,1H),7.85(d,J=7.8Hz,1H),7.79(d,J=8.8Hz,1H),7.62(d,J=2.0Hz,1H),7.54-7.45(m,1H),6.68(d,J=2.0Hz,1H),4.62(br d,J=4.3Hz,1H),4.15(br d,J=12.5Hz,1H),3.98-3.92(m,1H),3.81-3.67(m,4H),1.53(d,J=6.8Hz,3H)。
第三步
将化合物17d(30mg,58.3μmol,1.00eq)、1i(13.1mg,87.5μmol,1.50eq)和DIPEA(22.6mg,175μmol,3.00eq)溶于DMSO(3mL),使混合溶液在70℃反应17小时。反应完全,反应液经高效液相色谱法纯化得化合物17。
MS-ESI计算值[M+H] +484,实测值484。
1H NMR(400MHz,CDCl 3)δ:8.48(s,1H),8.02(d,J=8.0Hz,1H),7.97(d,J=8.3Hz,1H),7.79(br d,J=7.8Hz,1H),7.59(d,J=2.3Hz,1H),7.49-7.42(m,2H),6.67(d,J=2.0Hz,1H),4.30(br s,1H),4.01-3.56(m,12H),1.40(d,J=6.8Hz,3H),0.79-0.73(m,2H),0.61(br s,2H)。
实施例18-1&18-2
Figure PCTCN2018113683-appb-000101
将化合物1h(100mg,251μmol,1eq)、18a(34.1mg,251μmol,1eq,HCl)和N,N-二异丙基乙胺(97.5mg,754μmol,131μL,3eq)溶于二甲基亚砜(2mL),在70℃下反应18小时。反应完成后,经高效液相色谱法纯化得到消旋体取50mg进行手性分离得到化合物18-1和18-2。
MS-ESI计算值[M+H] +461,实测值461。
化合物18-1出峰位置:3.032min(手性柱:AD-3 150×4.6mm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:2.5mL/min,柱温:40℃)。
18-1核磁: 1H NMR(400MHz,CDCl 3)δ:8.58(br s,1H),7.84-8.23(m,3H),7.41-7.59(m,2H),6.52(s,1H),4.39 (br s,1H),3.63-4.02(m,10H),3.04-3.63(m,2H),2.99(d,J=4.8Hz,3H),1.46(s,3H),0.57-0.92(m,2H)。
化合物18-2出峰位置:3.587min(手性柱:AD-3 150×4.6mm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:2.5mL/min,柱温:40℃)。
18-2核磁: 1H NMR(400MHz,CDCl 3)δ:8.49-8.82(m,1H),7.82-8.23(m,3H),7.42-7.58(m,2H),6.53(br s,1H),4.39(br s,1H),3.60-4.06(m,10H),3.03-3.60(m,2H),2.99(d,J=4.8Hz,3H),1.46(s,3H),0.54-0.99(m,2H)。
实施例19
Figure PCTCN2018113683-appb-000102
第一步
将化合物19a(50mg,223μmol,1eq)、化合物19b(37.5mg,446μmol,40.8μL,2eq)、对甲苯磺酸(2.14mg,22.3μmol,1.59μL,0.1eq)溶于四氢呋喃(2mL)中,在氮气保护下,75℃反应3小时,反应完成后,浓缩掉溶剂,加水(40mL)稀释和饱和碳酸氢钠溶液(10mL),乙酸乙酯萃取(20mL×3),合并有机相,无水 硫酸钠干燥,过滤,减压旋干,得化合物19c。
MS-ESI计算值[M+H] +308,310实测值308,310。
第二步
将化合物19c(70mg,227μmol,1eq)、双联频那醇硼酸酯(57.7mg,227μmol,1eq)、Pd(dppf)Cl 2(8.31mg,11.4μmol,0.05eq)、乙酸钠(66.9mg,681μmol,3eq)溶入1,4-二氧六环(3mL)中,在氮气保护下,100℃反应16小时,反应完成后,得19e,不做处理直接投下一步。
MS-ESI计算值[M+H] +356,实测值356。
第三步
将化合物19d(80.7mg,227μmol,1eq)、化合物19e(68.0mg,227μmol,1eq)、四三苯基膦钯(13.1mg,11.4μmol,0.05eq)和碳酸钠(72.2mg,681μmol,3eq)溶入水(1mL)和1,4-二氧六环(3mL)中,在氮气保护下,90℃反应4小时,反应完成后,浓缩掉溶剂,加水(30mL)稀释,乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,柱层析(100%乙酸乙酯)纯化得19f。
MS-ESI计算值[M+H] +492,494实测值492,494。
第四步
将化合物19f(90mg,166μmol,1eq)、19g(24.9mg,166μmol,1eq,HCl)和DIPEA(64.6mg,499μmol,87.0μL,3eq)溶于DMSO(2mL),使混合溶液在70℃反应16小时。反应完全,得19h,不做处理,直接投下一步反应。
MS-ESI计算值[M+H] +569,实测值569。
第五步
将HCl(6M,166.48μL,6eq)加入化合物19h(94.7mg,166μmol,1eq)的DMSO(2mL)溶液,使混合溶液在20℃反应66小时。反应完全后,经高效液相色谱法提纯得19。
MS-ESI计算值[M+H] +485,实测值485。
1H NMR(400MHz,CDCl 3)δ:8.88(s,1H),8.02-8.35(m,4H),7.42-7.77(m,3H),4.40(br s,1H),4.01(br d,J=10.4Hz,3H),3.82-3.95(m,6H),3.65-3.82(m,3H),1.50(d,J=6.8Hz,3H),0.83(s,2H),0.59-0.69(m,2H)。
实施例20
Figure PCTCN2018113683-appb-000103
Figure PCTCN2018113683-appb-000104
将化合物1f(60mg,151μmol,1eq)和20a(22.6mg,151μmol,1eq)溶解于0.8毫升四氢呋喃中,加入叔丁醇钾(29.0mg,302μmol,2eq),加入Ruphos-Pd-G3(12.6mg,15.1μmol,0.1eq),反应液用氮气置换2分种后于80℃下反应1小时,反应完全,向反应液中加入1毫升水,经高效液相色谱法纯化得20。
1H NMR(400MHz,CDCl 3)δ:8.18(s,1H),8.09(d,J=8.0Hz,2H),7.62-7.53(m,2H),7.21(s,1H),4.51-4.18(m,3H),4.08-3.95(m,2H),3.90-3.75(m,6H),3.64(s,2H),3.08(d,J=5.0Hz,3H),1.31(s,3H),1.09-1.02(m,2H),0.90(s,2H)。
实施例21
Figure PCTCN2018113683-appb-000105
第一步
将化合物21a(5.46g,24.9mmol,3.27mL,1eq)和碳酸氢钠(6.27g,74.6mmol,2.90mL,3eq)溶于无水四氢呋喃(80mL),然后滴入21b(4.26g,74.6mmol,5.17mL,3eq),使混合溶液在氮气保护下于20℃反应16小时。反应完全,将反应液过滤,然后滤液浓缩得到21c。
第二步
将化合物21c(0.3g,1.25mmol,1eq)、21d(476mg,1.87mmol,1.5eq)、Pd(dppf)Cl 2(36.6mg,50.0μmol,0.04eq)和乙酸钾(368mg,3.75mmol,3eq)溶于无水二氧六环(25mL),使混合溶液在氮气保护下于90℃反应2小时。当反应完全,反应冷却后,减压浓缩,然后向反应液中加入15mL水和60mL(20mL×3)的乙酸乙酯进行萃取。然后有机相用无水硫酸钠干燥,浓缩。柱层析分离(42.5%--石油醚/乙酸乙酯),得到21e。MS-ESI计算值[M+H] +288,实测值288。
1H NMR(400MHz,CDCl 3)δ:7.95-7.89(m,2H),7.85(d,J=7.6Hz,1H),7.42-7.33(m,1H),2.83(qt,J=3.6,7.07Hz,1H),1.28(s,12H),.83-0.76(m,2H),0.54-0.59(m,2H)。
第三步
将化合物1f(0.1g,334μmol,1eq)、21e(125mg,435μmol,1.3eq)、二异丙基胺四三苯基膦钯(23.2mg,20.1μmol,0.06eq)和碳酸钾(139mg,1.00mmol,3eq)溶于无水二氧六环(10mL)和水(2mL),使反应液于70℃下反应4小时。反应完全后,将反应液减压浓缩,然后柱层析分离(20.2%--甲醇/乙酸乙酯)得21f。MS-ESI计算值[M+H] +424,426实测值424,426。
第四步
将化合物21f(0.09g,212μmol,1eq)、1i(31.8mg,212μmol,1eq,HCl)和二异丙基胺(27.4mg,212μmol,37.0μL,1eq)溶于二甲基亚砜(4mL),使反应液于70℃下反应15小时。反应完全后,将反应液经高效液相色谱法纯化得21。
MS-ESI计算值[M+H] +501,实测值501。
1H NMR(400MHz,CD 3OD)δ:8.60(s,1H),8.31(d,J=8Hz,1H),8.28(d,J=8.4Hz,1H),4.52(br d,J=6.0Hz,1H),4.07-3.93(m,6H),3.89-3.83(m,3H),3.80-3.71(m,3H),2.92-2.91(m,1H),1.48(d,J=6.8Hz,3H),0.90-0.79(m,4H),0.74-0.68(m,4H)。
实施例22
Figure PCTCN2018113683-appb-000106
Figure PCTCN2018113683-appb-000107
第一步
将化合物22a(5g,24.9mmol,1eq)和化合物22b(3.79g,29.9mmol,2.61mL,1.2eq)溶于二氯甲烷(50mL)中,滴加DMF(18.2mg,249μmol,19.1μL,0.01eq),室温反应2小时,反应完成后,得化合物22c。
第二步
将化合物22c(5.46g,29mmol,1eq)、化合物22d(7.28g,99.5mmol,10.5mL,4eq)和DIPEA(12.9g,99.5mmol,17.3mL,4eq)溶于二氯甲烷(50mL),室温反应5小时,反应完成后,水洗(50mL×3),有机相用无水硫酸钠干燥,过滤,减压旋干得22e。
MS-ESI计算值[M+H] +256,258,实测值256,258。
第三步
将化合物22e(300mg,1.17mmol,1eq)、化合物22f(356.9mg,1.41mmol,1.2eq)、醋酸钾(344mg,3.51mmol,3eq)和Pd(dppf)Cl 2(42.9mg,58.6μmol,0.05eq)溶于1,4-二氧六环(10mL),在氮气保护下,反应液在90℃反应4小时,反应完成后,浓缩掉溶剂,加水(30mL)稀释,乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,柱层析(40%乙酸乙酯),得22g。
MS-ESI计算值[M+H] +304,实测值304。
第四步
将化合物1f(98.7mg,330μmol,1eq)、22g(100mg,330μmol,1eq)、碳酸钠(105mg,989μmol,3eq)和四三苯基膦钯(19.1mg,16.5μmol,0.05eq)溶于1,4-二氧六环(3mL)和水(1mL),在氮气保护下,反应液在90℃反应5小时,反应完成后,浓缩掉溶剂,加水(20mL)稀释,乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,柱层析(100%乙酸乙酯)纯化得22h。
MS-ESI计算值[M+H] +440,442实测值440,442。
第五步
将化合物22h(60.0mg,136μmol,1eq)、1i(20.4mg,136μmol,1eq,HCl)和DIPEA(52.9mg,409μmol,71.3μL,3eq)溶于DMSO(3mL),使混合溶液70℃反应15小时,经高效液相色谱法提纯得22。
MS-ESI计算值[M+H] +517,实测值517。
1H NMR(400MHz,CDCl 3)δ:8.53(s,1H),8.19(d,J=8.0Hz,1H),8.06(d,J=8.6Hz,1H),7.94(d,J=7.8Hz,1H),7.45-7.64(m,2H),6.28(br s,1H),4.32-4.48(m,1H),3.62-4.24(m,12H),1.43-1.61(m,12H),0.58-0.95(m,4H)。
实施例23
Figure PCTCN2018113683-appb-000108
第一步
将化合物23a(23.0g,328mmol,24.5mL,1.0eq)和二碘化锌(5.20g,16.4mmol,0.05eq)溶解于200毫升二氯甲烷中,降内温至0℃,加入三甲基硅氰(39.1g,393mmol,49mL,1.2eq),反应液于25℃下反应18小时,反应完全,浓缩反应液并加入50毫升乙腈和50毫升盐酸水溶液(1M),于25℃搅拌5分钟。乙酸乙酯萃取,干燥有机相,过滤,浓缩,蒸发残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得23b。
1H NMR(400MHz,CDCl 3)δ:4.30(s,1H),2.62-2.59(m,2H),2.33-2.30(m,2H),1.95-1.79(m,2H)。
第二步
0℃条件下,向装有四氢铝锂(9.38g,247mmol,1.5eq)的四氢呋喃溶液(300mL)中加入23b(16.0g,164 mmol,1eq),并在20℃下反应18小时。反应完全,依次向反应液中加入水(9.38mL)、15%氢氧化钠(9.38mL)和水(28.1mL),搅拌15分钟,过滤,浓缩得23c。
1H NMR(400MHz,CDCl 3)δ:2.72(s,2H),2.12-1.76(m,7H),1.7-1.63(m,1H),1.49-1.33(m,1H)。
第三步
0℃条件下,向装有23c(2.0g,19.8mmol,1eq)和二异丙基乙基胺(4.0g,31.0mmol,5.4mL,1.6eq)的二氯甲烷(20.0mL)溶液中加入氯乙酰氯(2.23g,19.8mmol,1.6mL,1.0eq)。反应液于20℃下反应2小时。反应完全,浓缩反应液,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得23d。
1H NMR(400MHz,CDCl 3)δ:6.99(s,1H),4.17-4.02(m,2H),3.50(d,J=6.0Hz,2H),2.71(s,1H),2.14-1.98(m,4H),1.81-1.71(m,1H),1.64-1.49(m,1H)。
第四步
将化合物23d(2.2g,12.4mmol,1eq)加入无水四氢呋喃(100mL)中,降内温至0℃,加入氢化钠(1.49g,37.2mmol,60%纯度,3eq)。反应液于20℃下反应18小时,反应完全,向反应中加入水(15.0mL),用乙酸乙酯(20mL×3)萃取,合并有机相,干燥,过滤,浓缩得23e。
1H NMR(400MHz,CDCl 3)δ:6.96(s,1H),4.15(s,2H),3.44-3.35(m,2H),2.27-2.16(m,2H),2.09-2.01(m,2H),1.95-1.86(m,1H),1.74-1.63(m,1H)。
第五步
0℃条件下,向装有四氢铝锂(645mg,17mmol,2eq)的四氢呋喃溶液(30mL)中加入23e(1.2g,8.5mmol,1eq),并在20℃下反应18小时。反应完全,依次向反应液中加入水(0.7mL)、15%氢氧化钠(0.7mL)和水(2.1mL),搅拌15分钟,过滤,浓缩得23f。
1H NMR(400MHz,CDCl 3)δ:3.61-3.51(m,2H),2.87-2.76(m,4H),2.03-1.97(m,4H),1.86-1.82(m,1H),1.62-1.54(m,1H)。
第六步
将化合物23f(53mg,414μmol,1.1eq),1h(150mg,377μmol,1eq)和DIPEA(48mg,377μmol,66μL,1eq)溶于DMSO(4mL),使混合溶液70℃反应18小时。反应完全,反应液经高效液相色谱法纯化得23。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,CDCl 3)δ:8.63(s,1H),8.22(d,J=7.6Hz,1H),8.05(d,J=8.4Hz,1H),7.97(d,J=7.6Hz,1H),7.65-7.41(m,2H),6.53(br s,1H),4.40(d,J=6.8Hz,1H),4.12-3.95(m,3H),3.93-3.83(m,4H),3.83-3.68(m,5H),3.06(d,J=4.8Hz,3H),2.07-2.04(m,,4H),1.91-1.80(m,1H),1.77-1.70(m,1H),1.50(d,J=6.8Hz,3H)。
实施例24
Figure PCTCN2018113683-appb-000109
第一步
将化合物24a(23.0g,273mmol,24.2mL,1eq)和二碘化锌(4.36g,13.7mmol,0.05eq)溶解于200毫升二氯甲烷中,降内温至0℃,加入三甲基硅氰(32.6g,328mmol,41.1mL,1.2eq),反应液于25℃下反应18小时,反应完全,浓缩反应液并加入50毫升乙腈和50毫升盐酸水溶液(1M),于25℃搅拌5分钟。乙酸乙酯萃取,干燥有机相,过滤,浓缩,蒸发残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得24b。
1H NMR(400MHz,CDCl 3)δ:3.27-3.20(m,1H),2.10-2.06(m,4H),1.87-1.80(m,4H)。
第二步
0℃条件下,向装有四氢铝锂(8.19g,216mmol,1.5eq)的四氢呋喃溶液(300mL)中加入24b(16g,144mmol,1eq),并在20℃下反应18小时。反应完全,依次向反应液中加入水(8.19mL)、15%氢氧化钠(8.19mL)和水(25mL),搅拌15分钟,过滤,浓缩得24c。
1H NMR(400MHz,CDCl 3)δ:2.74(s,2H),1.90-1.55(m,9H),1.54-1.47(m,2H)。
第三步
0℃条件下,向装有24c(2.0g,17.4mmol,1eq)和二异丙基乙基胺(3.37g,26.1mmol,4.54mL,1.5eq)的二氯甲烷(20mL)溶液中加入氯乙酰氯(1.96g,17.4mmol,1.38mL,1.0eq)。反应液于20℃下反应2小时。反应完全,浓缩反应液,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得24d。
1H NMR(400MHz,CDCl 3)δ:7.03(s,1H),4.09(s,2H),3.44(d,J=5.6Hz,2H),2.00(s,1H),1.89-1.77(m,2H), 1.67-1.64(m,6H)。
第四步
将化合物24d(2.6g,13.6mmol,1eq)加入无水四氢呋喃(100mL)中,降内温至0℃,加入氢化钠(1.63g,40.7mmol,60%,3eq)。反应液于20℃下反应18小时,反应完全,向反应中加入水(15.0mL),用乙酸乙酯(20mL×3)萃取,合并有机相,干燥,过滤,浓缩得24e。
1H NMR(400MHz,CDCl 3)δ:6.79(s,1H),4.19(s,2H),3.38-3.33(m,2H),1.74-1.64(m,8H)。
第五步
0℃条件下,向装有四氢铝锂(880.32mg,23.2mmol,2eq)的四氢呋喃溶液(50mL)中加入24e(1.8g,11.6mmol,1eq),并在20℃下反应18小时。反应完全,依次向反应液中加入水(0.9mL)、15%氢氧化钠(0.9mL)和水(2.7mL),搅拌15分钟,过滤,浓缩得粗品24f。
第六步
将化合物24f(159mg,1.13mmol,3eq)、1h(150mg,377μmol,1eq)和DIPEA(97.5mg,754μmol,131μL,2eq)溶于DMSO(4mL),使混合溶液在70℃反应18小时。反应完全,反应液经高效液相色谱法纯化得24。
MS-ESI计算值[M+H] +503,实测值503。
1H NMR(400MHz,CDCl 3)δ:8.62(s,1H),8.22(d,J=8.0Hz,1H),8.05(d,J=8.4Hz,1H),7.97(d,J=8.0Hz,1H),7.57(t,J=8.0Hz,1H),7.51(d,J=8.4Hz,1H),6.46(s,1H),4.38(d,J=5.6Hz,1H),4.00(d,J=10.8Hz,2H),3.93-3.64(m,10H),3.06(d,J=4.8Hz,3H),1.85-1.70(m,6H),1.64(s,2H),1.49(d,J=6.8Hz,3H)。
实施例25
Figure PCTCN2018113683-appb-000110
Figure PCTCN2018113683-appb-000111
第一步
将化合物25a(100mg,446μmol,1.0eq),双联频那醇硼酸酯(136mg,536μmol,1.2eq),乙酸钾(131mg,1.34mmol,3.0eq)和Pd(dppf)Cl 2(16.4mg,22.4μmol,0.05eq)溶于无水二氧六环(5mL),换气三次,使混合溶液在氮气环境下于90℃反应16小时。反应完全后得25b的反应液,将反应液直接用于下一步。
MS-ESI计算值[M+H] +190,实测值190。
第二步
往含有化合物25b的上述反应液中加入化合物25c(133mg,446μmol,1.0eq)、四三苯基膦钯(25.8mg,22.3μmol,0.05eq),碳酸钠(142mg,1.34mmol,3.0eq)溶入水(2mL)和1,4-二氧六环(5mL)中,在氮气保护下,在80℃反应17小时,反应完成后,浓缩掉溶剂,加水(10mL)稀释,乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,柱层析(1:10甲醇/二氯甲烷),得25d。
MS-ESI计算值[M+H] +408,410实测值408,410。
第三步
将化合物25d(30mg,73.5μmol,1.00eq)、1i(10.0mg,66.8μmol,1.00eq)和DIPEA(28.5mg,221μmol,3.00eq)溶于DMSO(2mL),使混合溶液70℃反应17小时。反应完全,反应液经高效液相色谱法纯化得化合物25。
MS-ESI计算值[M+H] +485,实测值485。
1H NMR(400MHz,CDCl 3)δ:9.01(s,1H),8.22(br d,J=8.0Hz,1H),8.09(s,1H),8.01(d,J=8.4Hz,1H),7.75 (d,J=8.4Hz,1H),7.50(d,J=8.4Hz,1H),5.18(s,2H),4.31(br s,1H),4.02-3.54(m,12H),1.41(d,J=6.4Hz,3H),0.77(s,2H),0.61(br s,2H)。
实施例26
Figure PCTCN2018113683-appb-000112
第一步
将化合物26a(150mg,639μmol,1.00eq)、化合物26b(57.4mg,639μmol,1.00eq)和二异丙基乙胺(248mg,1.92mmol,2.83mL,3.00eq)溶于二氯甲烷(10mL)中,20℃中反应2小时。反应完成后,浓缩,柱层析(1:5甲醇/二氯甲烷),得到26c。
第二步
将化合物26c(0.12g,386μmol,1.00eq)、化合物26d(90.6mg,347μmol,0.90eq)、四三苯基膦钯(22.3mg,19.3μmol,0.05eq)和碳酸钠(123mg,1.16mmol,3.00eq)溶入水(3mL)和1,4-二氧六环(10mL)中,在氮气保护下,在80℃反应16小时,反应完成后,浓缩掉溶剂,加水(10mL)稀释,乙酸乙酯萃取(20mL× 3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,柱层析(1:5甲醇/二氯甲烷)纯化得26e。
第三步
将化合物26e(70mg,171μmol,1.00eq)、1i(30.7mg,205μmol,1.20eq)和DIPEA(66.2mg,512μmol,89μL,3.00eq)溶于DMSO(3mL),使混合溶液在70℃反应17小时。反应完全,反应液经高效液相色谱法纯化得26。
MS-ESI计算值[M+H] +487,实测值487。
1H NMR(400MHz,CDCl 3)δ:8.62(s,1H),8.21(d,J=7.8Hz,1H),8.06(d,J=8.4Hz,1H),7.98(d,J=7.8Hz,1H),7.56(t,J=7.8Hz,1H),7.49(d,J=8.4Hz,1H),6.59(br s,1H),4.48(br s,2H),4.13(br d,J=12.8Hz,2H),4.06(br s,2H),3.94(br s,2H),3.90-3.84(m,2H),3.62(br d,J=11.6Hz,2H),3.06(d,J=4.8Hz,3H),2.06-1.90(m,4H),0.89-0.82(m,2H),0.69(s,2H)。
实施例27
Figure PCTCN2018113683-appb-000113
将化合物15(150mg,306μmol,1eq)和甲醛(11.96mg,398μmol,11.0μL,1.3eq)溶于二氯乙烷(10mL)和醋酸(2mL),然后加入氰基硼氢化钠(38.5mg,613μmol,2eq),使混合溶液20℃反应18小时。反应完全,反应冷却后,将反应液减压浓缩残留物经高效液相色谱法纯化得27。
MS-ESI计算值[M+H] +504,实测值504。
1H NMR(400MHz,CD 3OD)δ:8.63(s,1H),8.36-8.30(m,2H),7.96(d,J=8.0Hz,1H),7.71(d,J=8.6Hz,1H),7.65(t,J=7.8Hz,1H),4.63(br s,2H),4.17-3.85(m,7H),3.83-3.67(m,8H),2.99(s,3H),2.61(s,3H),1.51(d,J=6.8Hz,3H)。
实施例28
Figure PCTCN2018113683-appb-000114
第一步
将化合物三苯基甲基溴化膦(25.9g,72.6mmol,1.6eq)溶于350毫升四氢呋喃中,20℃条件下,加入叔丁醇钾(1M,81.7mL,1.8eq),反应液于20℃下反应3小时,向反应液中加入28a(8.0g,45.4mmol,1eq),反应18小时。反应完全,向其中加入水(200mL)和乙酸乙酯(300mL)萃取,有机相用饱和食盐水(100mL×3)洗涤,干燥有机相,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得28b。
1H NMR(400MHz,CDCl 3)δ:7.36-7.31(m,5H),4.87-4.85(m,2H),4.46(s,2H),,4.45-4.08(m,1H),2.89-2.86(m,2H),2.78-2.73(m,2H)。
第二步
向装有28b(1.5g,8.61mmol,640μL,1eq)和N-(2-羟乙基)甲酸叔丁酯(1.67g,10.3mmol,1.60mL,1.2eq)的乙腈溶液(14mL)中加入NIS(2.32g,10.3mmol,1.2eq),并在20℃下反应4小时。反应完全,依次向 反应液中加入水(20mL)和乙酸乙酯(30mL)萃取,干燥,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)28c。
1H NMR(400MHz,CDCl 3)δ:7.33-7.30(m,5H),5.01-4.97(m,1H),4.44-4.43(m,2H),3.79-3.71(m,1H),3.33-3.32(m,3H),2.45-2.40(m,5H),2.03-2.02(m,1H),1.46(s,9H)。
第三步
将化合物28c(1.8g,3.90mmol,1eq)加入无水四氢呋喃(50mL)中,降内温至0℃,加入氢化钠(312mg,7.80mmol,60%,2eq)。反应液于20℃下反应18小时,反应完全,向反应中加入水(50.0mL),用乙酸乙酯(50mL)萃取,合并有机相,干燥,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得28d。
1H NMR(400MHz,CDCl 3)δ:7.33-7.20(m,5H),4.63-4.09(m,3H),3.52-3.50(m,2H),3.37(s,1H),3.30(s,2H),3.18(s,1H),2.35(s,1H),2.28-2.15(m,1H),1.97-1.85(m,2H),1.43-1.28(m,9H)。
第四步
将化合物28d(2.6g,13.6mmol,1eq)加入乙酸乙酯(15mL)中,向其中加入湿钯碳(0.1g,10%)。反应液用氢气置换三次并在此氛围(15psi)下于25℃下反应2小时,反应完全,过滤,浓缩得28e,粗品直接用于下一步。
第五步
向装有28e(360mg,1.48mmol,1eq)和乙酸乙酯(4mL)的反应瓶中加入HCl/EtOAc(4M,10mL,27eq),并在20℃下反应2小时。反应完全,浓缩得粗品28f,直接用于下一步。
第六步
将化合物28f(210mg,1.47mmol,1eq)和DIPEA(381mg,2.95mmol,513μL,2eq)溶于二氯甲烷(5mL)中,向反应液中加入氯甲酸苄酯(302mg,1.77mmol,251μL,1.2eq)。反应于20℃反应18小时。反应完全,向反应中加入水(30.0mL),用乙酸乙酯(20mL×3)萃取,合并有机相,干燥,过滤,浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得28g。
1H NMR(400MHz,CDCl 3)δ:7.43-7.29(m,5H),5.23-5.10(m,2H),4.55-4.10(m,1H),3.61(s,2H),3.55(s,1H),3.47-3.45(m,2H),3.34(s,1H),2.60-2.30(m,2H),1.95-1.93(m,2H)。
第七步
将化合物28g(320mg,1.15mmol,1eq)溶于二氯甲烷(5mL)中,向反应液中加入DMP(636mg,1.50mmol,1.3eq)。反应于20℃反应2小时。反应完全后浓缩,残渣用硅胶色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得28h。
1H NMR(400MHz,CDCl 3)δ:7.46-7.30(m,5H),5.16(s,2H),3.71(s,2H),3.62(s,2H),3.58-3.52(m,2H),3.15-3.07(m,2H),2.98(s,2H)。
第八步
将化合物28h(200mg,727μmol,1eq)溶于二氯甲烷(1mL)中,向反应液中加入N,N-二乙基三氟化硫(703mg,4.36mmol,576μL,6eq),反应于20℃反应18小时。反应完全,向反应中加入水(40.0mL),用二氯甲烷(30mL×3)萃取,合并有机相,干燥,过滤,浓缩,残渣用硅胶柱色谱法纯化(石油醚/乙酸乙酯=100:1-1:1)得28i。
第九步
将化合物28i(180mg,606μmol,1eq)加入甲醇(5mL)中,向其中加入湿钯碳(0.01g,10%)。反应液用氢气置换三次并在此氛围(15psi)下于20℃下反应2小时,反应完全,过滤,浓缩得28j,粗品直接用于下一步。
第十步
将化合物28j(42mg,257μmol,1eq)、1h(102mg,257μmol,1eq)和N,N-二异丙基乙基胺(66.5mg,514μmol,89.7μL,2eq)溶于DMSO(1mL),使混合溶液在70℃反应18小时。反应完全,反应液经高效液相色谱法纯化得28。
MS-ESI计算值[M+H] +525,实测值525。
1H NMR(400MHz,CDCl 3)δ:8.54(s,1H),8.15(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.89(d,J=8.0Hz,1H),7.54-7.41(m,2H),6.38(br s,1H),4.36(br s,1H),4.06-3.84(m,6H),3.79-3.53(m,6H),2.99(d,J=4.8Hz,3H),2.73-2.46(m,4H),1.44(d,J=6.8Hz,3H)。
实施例29
Figure PCTCN2018113683-appb-000115
将化合物29a(109mg,257μmol,1eq)、29b(42mg,257μmol,1eq)和N,N-二异丙基乙基胺(66.5mg,514.82μmol,89.7μL,2eq)溶于DMSO(1mL),使混合溶液在70℃反应18小时。反应完全,反应液经高效 液相色谱法纯化得29。
MS-ESI计算值[M+H] +551,实测值551。
1H NMR(400MHz,CDCl 3)δ:8.57(s,1H),8.20(d,J=8.0Hz,1H),8.07(d,J=8.4Hz,1H),7.98(d,J=8.0Hz,1H),7.61-7.47(m,2H),6.56(s,1H),4.43(d,J=6.4Hz,1H),4.12-4.05(m,2H),4.03-3.92(m,4H),3.88-3.83(m,1H),3.80-3.70(m,5H),3.01-2.92(m,1H),2.76-2.57(m,4H),1.53-1.50(m,3H),0.98-0.87(m,2H),0.76-0.60(m,2H)。
实施例30
Figure PCTCN2018113683-appb-000116
第一步
将甲基三苯基溴化磷(102g,285mmol,2eq)和叔丁醇钾(1M,257mL,1.8eq)溶于四氢呋喃(500mL)中,室温反应1小时,加入化合物30a(10.0g,143mmol,10.7mL,1eq),室温反应17小时候后,80℃蒸馏,馏分为化合物30b。
1H NMR(400MHz,CDCl 3)δ:4.51(dt,J=1.13,2.32Hz,2H),3.71-3.48(m,2H),2.61-2.46(m,2H),1.81-1.74(m,2H).
第二步
将化合物30b(30.0g,17.6mmol,1eq)、化合物30c(3.70g,21.1mmol,1.2eq)、N-碘琥珀酰亚胺(4.76g,21.1mmol,1.2eq)溶于四氢呋喃(10.0mL),室温反应24小时,反应完成后,浓缩掉溶剂,用水(100 mL)稀释,乙酸乙酯萃取(50.0mL×3),有机相无水硫酸钠干燥,过滤,减压旋干,柱层析(20%乙酸乙酯)得化合物30d。
MS-ESI计算值[M+H] +370,实测值270。
第三步
将化合物30d(30.9g,2.44mmol,1eq)、氢化钠(195mg,4.87mmol,60%纯度,2eq)溶于四氢呋喃(10.0mL),65℃反应16小时,反应完成后,加水(50.0mL)淬灭,乙酸乙酯萃取(50.0mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,得化合物30e。
MS-ESI计算值[M+H] +242,实测值142。
第四步
将化合物30e(0.18g,746μmol,1eq)溶于盐酸/乙酸乙酯(20.0mL),搅拌反应5小时,反应完成后,减压旋干,得30f。
MS-ESI计算值[M+H] +142,实测值142。
第五步
将化合物1h(141mg,354μmol,0.25eq),30f(200mg,1.42mmol,1eq,HCl),DIPEA(549mg,4.25mmol,740μL,3eq)溶于DMSO(5.00mL),使混合溶液70℃反应16小时,经高效液相色谱法提纯得30。
MS-ESI计算值[M+H] +503,实测值503。
1H NMR(400MHz,CDCl 3)δ:8.65(s,1H),8.22(br d,J=7.8Hz,1H),8.07(d,J=8.4Hz,1H),7.99(br d,J=7.8Hz,1H),7.63-7.48(m,2H),6.56(br s,1H),4.81(br d,J=13.6Hz,2H),4.42(br d,J=5.6Hz,1H),4.08-3.97(m,1H),3.96-3.67(m,6H),3.58(d,J=11.6Hz,1H),3.18(br d,J=13.2Hz,1H),3.08(d,J=4.8Hz,3H),2.26-1.93(m,4H),1.93-1.77(m,2H),1.52(d,J=6.8Hz,3H),1.36(br d,J=6.8Hz,3H).
实施例31
Figure PCTCN2018113683-appb-000117
Figure PCTCN2018113683-appb-000118
第一步
将化合物1h(100mg,251μmol,1.00eq),31a(36.0mg,251μmol,1.00eq),DIPEA(97.5mg,754μmol,131μL,3.00eq)溶于DMSO(3.00mL),使混合溶液90℃反应18小时。反应完全,反应液经高效液相色谱法纯化得31。
MS-ESI计算值[M+H] +505,实测值505。
1H NMR(400MHz,CDCl 3)δ:8.57(br s,1H),8.11(br d,J=7.2Hz,1H),7.95(br d,J=8.4Hz,1H),7.91(br d,J=8.0,1H),7.48(br t,J=8.0,1H),7.41(br d,J=8.4Hz,1H),6.53(br s,1H),4.31(br s,1H),4.01-3.52(m,14H),2.99(d,J=4.8Hz,3H),1.67(br s,4H),1.41(br d,J=6.8Hz,3H).
实施例32-1&32-2
Figure PCTCN2018113683-appb-000119
将化合物1h(280mg,704μmol,1.00eq),32a(101mg,703.8μmol,1.00eq),DIPEA(273mg,2.11mmol,131μL,3.00eq)溶于DMSO(5mL),使混合溶液80℃反应18小时。反应完全,反应液经高效液相色谱法纯化,进一步经SFC手性分离得到化合物32-1和32-2。
化合物32-1出峰位置:2.105min(手性柱:AD-3 100×4.6mm,I.D.,3μm,流动相:40%乙醇(0.05% 二乙胺)+二氧化碳,流速:2.8mL/min,柱温:40℃)
化合物32-1 MS-ESI计算值[M+H] +505,实测值505。
化合物32-1  1H NMR(400MHz,CDCl 3)δ:.56(s,1H),8.13(br d,J=8.0Hz,1H),7.98(d,J=8.0z,1H),7.90(d,J=7.8Hz,1H),7.54-7.39(m,2H),6.50(br s,1H),4.34(br d,J=7.2Hz,1H),4.03-3.55(m,16H),2.99(d,J=4.8Hz,3H),2.09-1.87(m,2H),1.42(d,J=6.8Hz,3H).
化合物32-2出峰位置:2.632min(手性柱:AD-3 100×4.6mm,I.D.,3μm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:2.8mL/min,柱温:40℃)
化合物32-2 MS-ESI计算值[M+H] +505,实测值505。
化合物32-2  1H NMR(400MHz,CDCl 3)δ:.56(s,1H),8.13(d,J=8.0Hz,1H),7.98(d,J=8.4Hz,1H),7.90(d,J=7.8Hz,1H),7.53-7.42(m,2H),6.52(br s,1H),4.34(br d,J=6.4Hz,1H),4.04-3.54(m,16H),2.98(d,J=4.8Hz,3H),2.08-1.88(m,2H),1.42(d,J=6.8Hz,3H).
实施例33-1&33-2
Figure PCTCN2018113683-appb-000120
Figure PCTCN2018113683-appb-000121
第一步
将化合物甲基三苯基溴化磷(32.4g,90.8mmol,1.6eq)室温下(20℃)溶于叔丁醇钾(11.5g,102mmol,1.8eq)的无水四氢呋喃(400mL),上述混合溶液在室温下反应3个小时,然后向上述溶液中滴加33a(10.0g,56.8mmol,1eq),使混合溶液20℃反应17小时。反应完全,TLC板显示有新化合物点产生,向反应液中加水(100mL),并用乙酸乙酯萃取(200mL×3),有机相用无水硫酸钠处理,减压浓缩,柱层析分离(50%--乙酸乙酯/石油醚),得到33b。
1H NMR(400MHz,CDCl3)δ:7.23-7.13(m,5H),4.74(br s,2H),4.34-4.27(m,2H),4.00-3.92(m,1H),2.80-2.71(m,2H),2.68-2.58(m,2H).
第二步
将化合物33b(8.7g,49.9mmol,1eq),N-Boc乙醇胺(9.66g,59.9mmol,9.29mL,1.2eq)溶于乙腈(90.0mL),然后向上述溶液中加入N-碘琥珀酰亚胺(13.5g,59.9mmol,1.2eq),使混合溶液20℃反应18小时。反应完全,TLC板显示有新化合物点产生,向反应液中加水(30.0mL),并用乙酸乙酯萃取(30mL×2),有机相用无水硫酸钠处理,减压浓缩,柱层析分离(35.5%--乙酸乙酯/石油醚),得到33c。
1H NMR(400MHz,CDCl3)δ:7.37-7.32(m,5H),5.03(br d,J=10.8Hz,1H),4.44(d,J=9.6Hz,2H),3.79-3.67(m,1H),3.34(br d,J=5.6Hz,3H),2.59-2.37(m,3H),2.31-2.11(m,2H),2.08-1.98(m,1H),1.52-1.42(m,9H).
第三步
将化合物33c(4g,8.67mmol,1eq)溶于N,N-二甲基乙酰胺(160mL),然后向上述溶液中在冰浴下加入氢化钠(694mg,60%纯度,2eq),使反应液于20℃下反应17小时。反应完全后,将反应液倒入饱和氯 化铵水溶液中(30mL),向反应液中加水(60.0mL),并用乙酸乙酯萃取(50mL×3),有机相用无水硫酸钠处理,减压浓缩,柱层析分离(30%--乙酸乙酯/石油醚),得到33d。
1H NMR(400MHz,CDCl3)δ:7.38-7.32(m,5H),4.47-4.40(m,2H),4.23-4.15(m,1H),3.62-3.53(m,2H),3.45(s,1H),3.41-3.32(m,2H),3.25(s,1H),2.43(br s,1H),2.35-2.27(m,1H),2.04(s,2H),1.46(d,J=5.2Hz,9H).
第四步
将化合物33d(1.00g,3.00mmol,1eq)溶于甲醇(30.0mL),然后在氮气环境下向上述溶液中氢氧化钯(421mg,20%纯度,0.2eq),然后用氢气置换几次,使反应液用氢气于50℃,50psi条件下反应50小时。反应完全后,将反应液过滤,滤液减压浓缩得到33e。
1H NMR(400MHz,CDCl3)δ:4.06-3.87(m,1H),3.51(br s,2H),3.42-3.36(m,2H),3.34-3.26(m,2H),3.21-3.14(m,1H),2.47-2.38(m,1H),2.35-2.25(m,1H),1.92-1.79(m,2H),1.39(s,9H).
第五步
将化合物33e(0.28g,1.15mmol,1eq)溶于乙酸乙酯(10.0mL),然后向上述溶液中加入盐酸/乙酸乙酯(4M,10mL,34.8eq),使反应液20℃条件下反应6小时。反应完全后,将反应液减压浓缩得到33f。
第六步
将化合物1h(0.3g,754μmol,1eq)溶于二甲基亚砜(10.0mL),然后向上述溶液中加入N,N-二异丙基乙胺(292mg,2.26mmol,3eq)和33f(162mg,902μmol,1.20eq,HCl),使反应液于70℃下反应25小时。反应完全后,向反应液中加水(12mL),并用乙酸乙酯萃取(10.0mL×3),有机相用无水硫酸钠处理,减压浓缩,柱层析分离(12.8%--甲醇/二氯甲烷),经手性拆分得到33-1和33-2。
化合物33-1出峰位置:1.672min(手性柱:AD-3 100×4.6mm,I.D.,3μm,流动相:40%异丙醇(0.05%二乙胺)+二氧化碳,流速:2.8mL/min,柱温:40℃)
化合物33-1 MS-ESI计算值[M+H] +505,实测值505。
化合物33-1  1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.34(d,J=8.4Hz,1H),8.29(d,J=8.4Hz,1H),7.96(d,J=8.4Hz,1H),7.68(d,J=8.4Hz,1H),7.66-7.62(m,1H),4.54(br d,J=7.2Hz,1H),4.12(br s,1H),4.04-3.93(m,3H),3.93-3.70(m,10H),3.02-2.97(m,3H),2.56-2.52(m,2H),1.96-1.92(m,2H),1.51(d,J=6.8Hz,3H).
化合物33-2出峰位置:3.338min(手性柱:AD-3 100×4.6mm,I.D.,3μm,流动相:40%异丙醇(0.05%二乙胺)+二氧化碳,流速:2.8mL/min,柱温:40℃)
化合物33-2 MS-ESI计算值[M+H] +505,实测值505。
化合物33-2  1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.33(d,J=7.8Hz,1H),8.28(d,J=8.4Hz,1H),7.96(d,J=7.8Hz,1H),7.67(d,J=8.4Hz,1H),7.66-7.62(m,1H),4.57(br d,J=7.2Hz,1H),4.44-4.36(m,1H),4.08-3.95(m,4H),3.95-3.85(m,3H),3.82-3.70(m,5H),2.99(s,3H),2.43-2.40(m,2H),2.04-1.95(m,2H),1.51(d,J=6.8Hz,3H).
实施例34-1&34-2
Figure PCTCN2018113683-appb-000122
第一步
将化合物34a(0.27g,1.11mmol,1eq)溶于无水四氢呋喃(20.0mL),然后在0℃下向上述混合溶液中小心加入氢化钠(133mg,60%纯度,3eq),和碘甲烷(1.19g,522μL,7.55eq),反应液逐渐升至室温,在室温下反应3个小时。反应完全,TLC板显示有新化合物点产生,将反应液减压浓缩,得到34b。
第二步
将化合物34b(0.10g,389μmol,1eq)溶于乙酸乙酯(8mL),然后向上述溶液中加入盐酸/乙酸乙酯(4M,8.0mL,82.34eq),使混合溶液20℃反应18小时。反应完全,TLC板显示有新化合物点产生,反应液减压浓缩,得到34c。
第三步
将化合物1h(0.1g,251μmol,1eq)溶于二甲基亚砜(5.00mL),然后向上述溶液中加入N,N-二异丙基乙胺(65.0mg,87.6μL,2eq)和34c(73.0mg,377μmol,1.5eq,HCl),使反应液于70℃下反应22小时。反应完全后,向反应液中加水(20.0mL),并用乙酸乙酯萃取(15.0mL×3),有机相用无水硫酸钠处理,减压浓缩,柱层析分离(100%--乙酸乙酯/石油醚),经手性拆分得到34-1和34-2。
化合物34-1出峰位置:2.407min(手性柱:OD-3 50×4.6mm,I.D.,3μm,流动相:A:CO2,B:乙醇(0.05%二乙胺),保持5%B 0.2min,然后在1.4min内B的含量由5%梯度增加到40%,然后保持40%B1.05min,最后保持5%B 0.35min,流速:4mL/min,柱温:40℃)
化合物34-1 MS-ESI计算值[M+H] +519,实测值519。
化合物34-1  1H NMR(400MHz,CD 3OD)δ:8.64-8.60(m,1H),8.34(d,J=8.4Hz,1H),8.29(d,J=8.4Hz,1H),7.96(dd,J=1.2,8.0Hz,1H),7.68(d,J=8.4Hz,1H),7.66-7.61(m,1H),4.55(br d,J=7.2Hz,1H),4.06-3.93(m,3H),3.92-3.70(m,10H),3.28(s,3H),2.99(s,3H),2.56-2.48(m,2H),1.96-1.86(m,2H),1.51(d,J=6.8Hz,3H).
化合物34-2出峰位置:1.807min(手性柱:OD-3 50×4.6mm,I.D.,3μm,流动相:A:CO 2,B:乙醇(0.05%二乙胺),保持5%B 0.2min,然后在1.4min内B的含量由5%梯度增加到40%,然后保持40%B 1.05min,最后保持5%B 0.35min,流速:4mL/min,柱温:40℃)
化合物34-2 MS-ESI计算值[M+H] +519,实测值519。
化合物34-2  1H NMR(400MHz,CD 3OD)δ:8.62(s,1H),8.33(d,J=8.03Hz,1H),8.29(d,J=8.4Hz,1H),7.96(d,J=8.4Hz,1H),7.68(d,J=8.4Hz,1H),7.66-7.62(m,1H),4.57(br d,J=6.0Hz,1H),4.08-3.97(m,5H),3.94-3.2(m,8H),3.27(s,3H),2.99(s,3H),2.39-2.27(m,2H),2.09-2.01(m,2H),1.51(d,J=6.8Hz,3H).
实施例35
Figure PCTCN2018113683-appb-000123
第一步
将化合物1f(2g,6.0mmol,1eq),35a(993mg,6.0mmol,1eq),碳酸钠(1.9g,18.1mmol,3eq),二氯(二三苯基膦)钯(211mg,301μmol,0.05eq)溶于无水二氧六环(35mL)和水(7.0mL)中,上述溶液在氮气环境下于70℃反应19小时。反应完全,将反应液减压浓缩,然后加水(15mL),并用乙酸乙酯萃取(20mL×3),有机相用无水硫酸钠处理,减压收集,柱层析分离(甲醇/乙酸乙酯;R f=0.28)得到35b。
MS-ESI计算值[M+H] +384和386,实测值384和386。
第二步
将化合物35b(0.1g,261μmol,1eq),30f(73.6mg,414μmol,1.6eq),N,N-二异丙基乙胺(33.7mg,261μmol,1eq)溶于二甲基亚砜(5.00mL),使反应液于70℃下反应40小时。反应完全后,向反应液过滤,并经 高效液相色谱法纯化得35。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,CD 3OD)δ:8.70(s,1H),8.36(d,J=8.0Hz,1H),8.30(d,J=8.8Hz,1H),8.04(d,J=8.0Hz,1H),7.68(d,J=8.8Hz,1H),7.68-7.64(m,1H),4.88(s,2H),4.56(d,J=6.4Hz,1H),4.04-3.96(m,2H),3.90(dd,J=2.8,11.6Hz,1H),3.84(dd,J=3.2,11.6Hz,1H),3.77-3.75(m,3H),3.59(d,J=11.6Hz,1H),3.14(dd,J=1.6,13.6Hz,1H),2.20-2.08(m,2H),2.08-1.96(m,2H),1.92-1.76(m,2H),1.50(d,J=6.8Hz,3H),1.36(d,J=6.8Hz,3H)。
实施例36
Figure PCTCN2018113683-appb-000124
第一步
将钠(3.66g,159mmol,3.77mL,2eq)溶于乙醇(147mL),加入36a(9g,79.6mmol,8.49mL,1eq),再加入化合物36b(16.1g,79.6mmol,8.11mL,1eq),使混合溶液80℃反应3小时。然后旋蒸除去乙醇,并倒入水(500mL)中,用乙酸乙酯(100mL)萃取,用无水硫酸钠干燥,过滤,减压旋干,柱层析(20%乙酸乙酯)纯化得化合物36c。
1H NMR(400MHz,CDCl 3)δ:4.26-4.14(m,2H),2.67-2.56(m,4H),2.20-2.04(m,2H),1.28-1.25(m,3H)。
第二步
将四氢锂铝(1.98g,52.2mmol,2eq)溶于四氢呋喃(100mL),加入化合物36c(4.00g,26.1mmol,1eq), 使混合溶液45℃反应18小时,反应完成后,在0-20℃依次加水(2.00mL)、15%NaOH(2.00mL)、水(6.00mL)淬灭,搅拌0.5小时,过滤,减压旋干,得化合物36d。
1H NMR(400MHz,CDCl 3)δ:3.75(s,2H),2.97(s,2H),1.93-1.73(m,6H)。
第三步
将化合物36d(0.5g,4.34mmol,1eq)溶于二氯甲烷(10.0mL),加入碳酸钠(920mg,8.68mmol,2eq)和化合物36e(539mg,4.77mmol,380μL,1.1eq),使混合溶液20℃反应1小时,反应完成后,减压旋干,柱层析(20%甲醇/二氯甲烷)得化合物36f。
1H NMR(400MHz,CDCl 3)δ:7.00(s,1H),4.11-4.08(m,3H),3.54-4.54(m,5H),3.04-3.01(m,1H),1.98-1.94(m,2H),1.82-1.75(m,4H)。
第四步
将化合物36f(0.22g,1.15mmol,1eq)溶于四氢呋喃(22.0mL),加入氢化钠(138mg,3.44mmol,60%,3eq),使混合溶液20℃反应1小时,反应完成后,减压旋干,柱层析(20%甲醇/二氯甲烷)得化合物36g。
MS-ESI计算值[M+H] +156,实测值156。
第五步
将化合物36g(0.35g,2.26mmol,1eq)溶于四氢呋喃(10.0mL),加入四氢锂铝(257mg,6.77mmol,3eq),使混合溶液25℃反应18小时,反应完成后,在0-20℃依次加水(0.26mL)、15%NaOH(0.26mL)、水(0.78mL)淬灭,搅拌0.5小时,过滤,减压旋干,得化合物36h。
MS-ESI计算值[M+H] +142,实测值142。
第六步
将化合物36h(0.1g,261μmol,1eq),35b(55.18mg,391μmol,1.5eq),DIPEA(101mg,782μmol,136μL,3eq)溶于DMSO(3.00mL),使混合溶液70℃反应16小时,经高效液相色谱法提纯得36。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,CDCl 3)δ:8.70(br s,1H),8.24(br d,J=7.6Hz,1H),8.08-7.03(m,2H),7.59(t,J=7.6Hz,1H),7.49(d,J=8.4Hz,1H),6.70(br s,1H),5.76(br s,1H),4.40(br s,1H),4.16(br d,J=12.4Hz,2H),4.02(br s,2H),3.99-3.81(m,5H),3.81-3.61(m,5H),1.92(br s,6H),1.49(d,J=6.8Hz,3H)。
实施例37
Figure PCTCN2018113683-appb-000125
将化合物1h(0.1g,251μmol,1eq),36h(53.2mg,377μmol,1.5eq),DIPEA(97.5mg,754μmol,131μL,3eq)溶于DMSO(5.00mL),使混合溶液70℃反应16小时,经高效液相色谱法提纯得37。
MS-ESI计算值[M+H] +503,实测值503。
1H NMR(400MHz,CDCl 3)δ:8.64(br s,1H),8.19(br d,J=7.2Hz,1H),8.06(br d,J=8.4Hz,1H),7.98(br d,J=7.6Hz,1H),7.57(t,J=7.6Hz,1H),7.49(d,J=8.4Hz,1H),6.70(br s,1H),4.40(br s,1H),4.16(br d,J=12.0Hz,2H),4.02(br s,2H),3.96-3.84(m,4H),3.84-3.59(m,6H),3.07(d,J=4.8Hz,3H),2.11-1.84(m,6H),1.49(d,J=6.8Hz,3H)。
实施例38
Figure PCTCN2018113683-appb-000126
Figure PCTCN2018113683-appb-000127
第一步
将四氢锂铝(3.91g,103mmol,2eq)溶于四氢呋喃(100mL),加入化合物38a(5.00g,51.5mmol,3.37mL,1eq),使混合溶液45℃反应18小时,反应完成后,在0-20℃依次加水(4.00mL)、15%NaOH(4.00mL)、水(8.00mL)淬灭,搅拌0.5小时,过滤,减压旋干,得38b。
MS-ESI计算值[M+H] +102,实测值102。
第二步
将化合物38b(4g,39.6mmol,1eq)溶于二氯甲烷(10.0mL),加入碳酸钠(8.38g,79.1mmol,2eq)和化合物38c(4.47g,39.6mmol,3.15mL,1.0eq),使混合溶液20℃反应1小时,反应完成后,减压旋干,柱层析(20%甲醇/二氯甲烷)得化合物38d。
MS-ESI计算值[M+H] +178,实测值178。
1H NMR(400MHz,CDCl 3)δ:7.27(s,1H),4.09(s,2H),3.44(d,J=5.2Hz,2H),3.32(d,J=5.6Hz,2H),2.83-2.81(m,1H),0.52(d,J=3.6Hz,4H)。
第三步
将化合物38d(4.70g,26.5mmol,1eq)溶于四氢呋喃(470mL),加入氢化钠(3.18g,79.4mmol,60%纯度,3eq),使混合溶液20℃反应1小时,反应完成后,加入盐酸调节pH至7,加入无水硫酸钠干燥,得化合物38e。
MS-ESI计算值[M+H] +142,实测值142。
第四步
将化合物38e(0.5g,3.54mmol,1eq)溶于四氢呋喃(10mL),加入四氢锂铝(269mg,7.08mmol,2eq),使混合溶液20℃反应1小时,反应完成后,在0-20℃依次加水(0.26mL)、15%NaOH(0.26mL)、水(0.78mL)淬灭,搅拌0.5小时,过滤,减压旋干,得化合物38f。
MS-ESI计算值[M+H] +128,实测值128。
第五步
将化合物38f(48.2mg,126μmol,1eq),35b(159.84mg,1.26mmol,10eq),DIPEA(48.7mg,377μmol,65.7μL,3eq)溶于DMSO(5.00mL),使混合溶液70℃反应16小时,经高效液相色谱法提纯得38。
MS-ESI计算值[M+H] +475,实测值475。
1H NMR(400MHz,CDCl 3)δ:8.69(br s,1H),8.25(br d,J=7.2Hz,1H),8.06-8.03(m,2H),7.71-7.40(m,2H),6.63(br s,1H),5.80(br s,1H),4.48-4.17(m,3H),4.07-3.94(m,2H),3.87(br s,4H),3.83-3.62(m,4H),3.55(s,2H),1.48(br d,J=6.4Hz,3H),0.85-0.31(m,4H)。
实施例39
Figure PCTCN2018113683-appb-000128
第一步
将化合物38f(141mg,354μmol,0.25eq),1h(200mg,1.42mmol,1eq,HCl),DIPEA(549mg,4.25mmol,740μL,3eq)溶于DMSO(5.00mL),使混合溶液70℃反应16小时,经高效液相色谱法提纯得39。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,CDCl 3)δ:8.64(br s,1H),8.21(br d,J=7.6Hz,1H),8.06(d,J=8.4Hz,1H),7.99(br d,J=7.6Hz,1H),7.58(t,J=7.6Hz,1H),7.50(br d,J=8.4Hz,1H),6.59(br s,1H),4.38(br d,J=6.8Hz,1H),4.34-4.12(m, 2H),4.07-3.96(m,2H),3.96-3.81(m,5H),3.81-3.61(m,3H),3.56(s,2H),3.08(d,J=4.8Hz,3H),1.49(br d,J=6.8Hz,3H),0.80-0.54(m,4H)。
实施例40
Figure PCTCN2018113683-appb-000129
第一步
将化合物40a(0.28g,1.13mmol,1eq)溶于三氟乙酸(5.00mL)和二氯甲烷(10.0mL),室温搅拌反应2小时,反应完成后,减压旋干,得40b。
MS-ESI计算值[M+H] +148,实测值148。
第二步
将化合物40b(300mg,1.15mmol,1eq,TFA)、化合物40c(320mg,804μmol,0.7eq)、DIPEA(445mg,3.45mmol,600μL,4eq)溶于二甲基亚砜(5.00mL),70℃反应16小时,反应完成后,经高效液相色谱法提纯得40d。
MS-ESI计算值[M+H] +509,实测值509。
第三步
将化合物40d(100mg,197μmol,1eq)、对甲苯磺酰氯(37.5mg,197μmol,1eq)和氢化钠(15.7mg,393μmol,60%,2eq)溶于DMF(10.0mL),室温反应16小时,反应完成后,经高效液相色谱法提纯得40。
MS-ESI计算值[M+H] +491,实测值491。
1H NMR(400MHz,CDCl 3)δ:8.65(s,1H),8.23(br d,J=7.8Hz,1H),8.09(d,J=8.4Hz,1H),7.99(br d,J=7.8Hz,1H),7.62-7.53(m,2H),6.60(br s,1H),4.66(d,J=6.4Hz,2H),4.57-4.41(m,3H),4.32-4.14(m,2H),4.08- 3.85(m,5H),3.83-3.75(m,5H),3.08(d,J=4.8Hz,3H),1.52(d,J=6.8Hz,3H)。
实施例41
Figure PCTCN2018113683-appb-000130
第一步
将化合物41a(70.0mg,138umol,1eq)、甲烷磺酰氯(0.8g,6.98mmol,541μL,50.7eq)和三乙胺(27.9mg,275μmol,38.3μL,2eq)溶于二氯甲烷(10.0mL)中,室温反应16小时,反应完成后,减压旋干,得化合物41b。
MS-ESI计算值[M+H] +665,实测值665。
第二步
将化合物41b(59.9mg,90.26μmol,1eq)、四正丁基碘化铵(3.33mg,9.03μmol,0.1eq)、硫化钠(21.1mg,271μmol,11.4μL,3eq)溶于N,N’-二甲基甲酰胺(5.00mL),在氮气保护下,70℃反应18小时,反应完成后,水洗(50.0mL×3),经高效液相色谱法提纯得41。
MS-ESI计算值[M+H] +507,实测值507。
1H NMR(400MHz,CDCl 3)δ:8.67(br s,1H),8.20(br d,J=8.0Hz,1H),8.10-8.03(m,1H),7.99(br d,J=8.0Hz,1H),7.60-7.52(m,2H),6.64(br s,1H),4.46(br d,J=5.6Hz,1H),4.42-4.33(m,1H),4.19(br d,J=13.2Hz,1H),4.09-3.91(m,3H),3.91-3.69(m,7H),3.42(br d,J=10.0Hz,2H),3.06(d,J=4.8Hz,3H),3.00(br d,J=8.0Hz,2H),1.53(br d,J=6.8Hz,3H)。
实施例42
Figure PCTCN2018113683-appb-000131
将化合物41(70.0mg,138μmol,1eq)溶于甲醇(20.0mL)中,滴加单过硫酸氢钾(84.9mg,138μmol,1eq)的水溶液(10.0mL),室温反应1小时,反应完成后,经高效液相色谱法提纯得42。
MS-ESI计算值[M+H] +523,实测值523。
1H NMR(400MHz,CDCl 3)δ:8.72-7.51(m,1H),8.11(br d,J=7.2Hz,1H),8.00(d,J=8.4Hz,1H),7.93(br d,J=7.8Hz,1H),7.55-7.43(m,2H),4.49-4.33(m,1H),4.20(s,2H),3.94(br d,J=8.4Hz,3H),3.82-3.74(m,1H),3.74-3.60(m,5H),3.52(br d,J=13.6Hz,2H),3.28-3.06(m,2H),3.00-2.94(m,3H),1.44(d,J=6.8Hz,3H)。
实施例43
Figure PCTCN2018113683-appb-000132
将化合物41(120mg,237μmol,1eq)溶于甲醇(5.00mL)中,滴加单过硫酸氢钾(291mg,474μmol,2 eq)的水溶液(5.00mL),室温反应30小时,反应完成后,经高效液相色谱法提纯得43。
MS-ESI计算值[M+H] +539,实测值539。
1H NMR(400MHz,CDCl 3)δ:8.58(br s,1H),8.14(br d,J=8.0Hz,1H),8.02(d,J=8.4Hz,1H),7.91(br d,J=7.6Hz,1H),7.60-7.44(m,2H),6.57(br s,1H),4.42(br d,J=6.0Hz,1H),4.27-3.85(m,10H),3.82-3.60(m,6H),2.99(d,J=4.8Hz,3H),1.46(d,J=6.8Hz,3H)。
实施例44
Figure PCTCN2018113683-appb-000133
将化合物35b(150mg,391μmol,1eq)、23f(128mg,782μmol,1eq)和DIPEA(152mg,1.17mmol,3eq)溶于DMSO(3.00mL),混合溶液在90℃反应20小时。反应完全,反应液经高效液相色谱法纯化得44。
MS-ESI计算值[M+H] +475,实测值475。
1H NMR(400MHz,CDCl 3)δ:8.66(br s,1H),8.18(br d,J=7.6Hz,1H),7.99(d,J=8.4Hz,1H),7.95(br d,J=7.6Hz,1H),7.51(t,J=7.6Hz,1H),7.46(br d,J=8.4Hz,1H),6.72-6.11(m,1H),5.64(br s,1H),4.35(br s,1H),3.95-3.57(m,11H),2.07-1.77(m,7H),1.45(br d,J=6.8Hz,3H)。
实施例45-1&45-2
Figure PCTCN2018113683-appb-000134
第一步
将TMSCN(18.9g,191mmol,1.5eq)、三氟化硼***络合物(18.1g,127mmol,1.0eq)依次加入化合物45a(13.0g,127mmol,1.0eq)的二氯甲烷(250mL)溶液中,20℃反应15小时,反应完成后,浓缩,柱层析(2:1石油醚/乙酸乙酯)得45b。
1H NMR(400MHz,CDCl 3)δ:3.34(d,J=12.0Hz,1H),3.19(s,1H),3.16(dd,J=1.6,12.0Hz,1H),3.13-3.01(m,2H),2.59-2.47(m,1H),2.32(td,J=8.8,12.8Hz,1H)。
第二步
在0摄氏度下,将硼烷四氢呋喃(1M,157mL,1.5eq)滴加进化合物45b的四氢呋喃(200mL)溶液中,混合溶液在20℃反应17小时。10mL甲醇加入反应液中淬灭反应,反应液浓缩得到45c,粗品直接用于下一步。
第三步
在0摄氏度下,将化合物45d(8.48g,75.1mmol,1.0eq)滴加进化合物45c(10.0g,75.1mmol,1.0eq)和二异丙基乙胺(19.4g,150mmol,2.0eq)的二氯甲烷(150mL)中,混合溶液在20℃反应3小时。5mL水加入反应液中淬灭反应,反应液浓缩,柱层析(100%乙酸乙酯)得45e。
1H NMR(400MHz,CDCl 3)δ:7.12-6.88(m,1H),4.06-3.99(m,2H),3.55(d,J=6.0Hz,2H),3.01-2.84(m,3H),2.82(d,J=11.2Hz,1H),2.68(dd,J=1.6,11.6Hz,1H),2.08-1.99(m,1H),1.81(td,J=9.2,13.2Hz,1H)。
第四步
在0摄氏度下,将叔丁醇钾(9.63g,85.8mmol,3.0eq)加入化合物45e(6.0g,28.6mmol,1.0eq)的四氢呋喃(500mL)溶液中,混合溶液在20℃反应2小时。反应液浓缩,柱层析(1:10甲醇/乙酸乙酯)得45f。
1H NMR(400MHz,CDCl 3)δ:6.43(br s,1H),4.26(s,2H),3.59-3.39(m,2H),3.13-3.00(m,2H),2.96-2.78(m,2H),2.47-2.32(m,1H),2.02-1.88(m,1H).
第五步
在0度下,将四氢铝锂(329mg,8.66mmol,1.5eq)加入化合物45f(1.0g,5.77mmol,1.0eq)的无水四氢呋喃(30.0mL)中,混合溶液在20℃反应1小时。往反应液中依次滴加0.3mL水,0.3mL15%氢氧化钠水溶液,1mL水,过滤,滤饼用10mL乙酸乙酯淋洗,滤液浓缩,得到45g,粗品直接用于下一步。
第六步
将化合物45g(92.1mg,578μmol,2.3eq),45h(100mg,251μmol,1.00eq),DIPEA(97.5mg,754μmol,3.00eq)溶于DMSO(3.00mL),使混合溶液80℃反应18小时。反应完全,反应液加水(10.0mL)稀释后用乙酸乙酯萃取(20.0mL×3),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,制备层析板纯化(100%乙酸乙酯)得45i。进一步经SFC手性分离得到化合物45-1和45-2。
化合物45-1 MS-ESI计算值[M+H] +521,实测值521。
化合物45-1出峰位置:0.950min(手性柱:AD-3 50×4.6mm,I.D.,3μm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:4mL/min,柱温:40℃)。
化合物45-1  1H NMR(400MHz,CDCl 3)δ:8.57(s,1H),8.13(br d,J=7.6Hz,1H),7.98(d,J=8.4Hz,1H),7.90(d,J=8.0Hz,1H),7.52-7.42(m,2H),6.57(br s,1H),4.35(br d,J=6.8Hz,1H),4.22(d,J=13.2Hz,1H),4.13(br s,1H),3.95-3.61(m,10H),2.98(d,J=4.8Hz,3H),2.97-2.78(m,4H),2.21(td,J=6.0,12.4Hz,1H),1.99-1.88(m,1H),1.43(d,J=6.8Hz,3H)。
化合物45-2 MS-ESI计算值[M+H] +521,实测值521。
化合物45-2出峰位置:1.168min(手性柱:AD-3 50×4.6mm,I.D.,3μm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:4mL/min,柱温:40℃)。
化合物45-2  1H NMR(400MHz,CDCl 3)δ:8.57(s,1H),8.13(br d,J=7.6Hz,1H),7.98(d,J=8.4Hz,1H),7.90(d, J=8.0Hz,1H),7.52-7.43(m,2H),6.54(br s,1H),4.34(br d,J=6.8Hz,1H),4.23(br d,J=13.2Hz,1H),4.17(br s,1H),3.95-3.60(m,10H),2.99(d,J=4.8Hz,3H),2.97-2.76(m,4H),2.20(td,J=6.0,12.4Hz,1H),2.00-1.89(m,1H),1.42(d,J=6.8Hz,3H).
实施例46-1&46-2
Figure PCTCN2018113683-appb-000135
将过硫酸氢钾(402mg,653μmol,0.85eq)的水溶液(40.0mL)滴加到化合物46a(400mg,768μmol,1.0eq)的甲醇(60.0mL)溶液中,25℃反应1.5小时。10毫升饱和硫代硫酸钠溶液淬灭反应,浓缩至体积大约为50毫升,用乙酸乙酯萃取(30mL×3),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,柱层析(1:10甲醇/乙酸乙酯)得消旋体,进一步经SFC手性分离得到化合物46-1和46-2。
化合物46-1出峰位置:5.297min(手性柱:AD-3 150×4.6mm,I.D.,3μm,流动相:40%甲醇(0.05%二乙胺)+二氧化碳,流速:2.5mL/min,柱温:40℃)。
化合物46-1 MS-ESI计算值[M+H] +537,实测值537。
化合物46-1  1H NMR(400MHz,CDCl 3)δ:8.69(br s,1H),8.13-7.91(m,3H),7.53-7.40(m,2H),4.46-4.22(br d,J=6.8Hz,2H),4.14(br d,J=12.2Hz,1H),3.97-3.62(m,10H),3.17(br s,1H),2.97(d,J=4.8Hz,6H),2.68(br d,J=6.4Hz,1H),2.49-2.38(m,1H),1.43(d,J=6.8Hz,3H).
化合物46-2出峰位置:6.265min(手性柱:AD-3 150×4.6mm,I.D.,3μm,流动相:40%甲醇(0.05%二乙胺)+二氧化碳,流速:2.5mL/min,柱温:40℃)。
化合物46-2 MS-ESI计算值[M+H] +537,实测值537。
化合物46-2  1H NMR(400MHz,CDCl 3)δ:8.70(br s,1H),8.13-7.91(m,3H),7.55-7.39(m,2H),4.45-4.22(m,2H),4.20-4.08(m,1H),3.94-3.63(m,10H),3.17(br s,1H),2.97(d,J=4.8Hz,6H),2.78-2.61(m,1H),2.44(br d,J=14.1Hz,1H),1.41(d,J=6.8Hz,3H)。
实施例47-1&47-2
Figure PCTCN2018113683-appb-000136
将过硫酸氢钾(779mg,1.27mmol,3.3eq)的水溶液(30.0mL)滴加到化合物47a(200mg,384μmol,1.0eq)的甲醇(30.0mL)溶液中,25℃反应5小时。10毫升饱和硫代硫酸钠溶液淬灭反应,浓缩至体积大约为40毫升,用二氯甲烷萃取(50.0mL×4),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,制备层析板纯化(1:20甲醇/二氯甲烷)得消旋中间体,进一步经SFC手性分离得到化合物47-1和47-2。
化合物47-1 MS-ESI计算值[M+H] +553,实测值553。
化合物47-1出峰位置:1.617min(手性柱:AD-3 50×4.6mm,I.D.,3μm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:4mL/min,柱温:40℃)。
化合物47-1  1H NMR(400MHz,CDCl 3)δ:8.65(s,1H),8.23(d,J=8.0Hz,1H),8.10(d,J=8.4Hz,1H),7.99(d,J=8.0Hz,1H),7.64-7.55(m,2H),6.58(br s,1H),4.66-4.34(m,3H),4.07-3.55(m,10H),3.45-3.18(m,4H),3.08(d,J=4.8Hz,3H),2.57-2.45(m,1H),2.38-2.26(m,1H),1.53(d,J=6.8Hz,3H)。
化合物47-2 MS-ESI计算值[M+H] +553,实测值553。
化合物47-2出峰位置:2.000min(手性柱:AD-3 50×4.6mm,I.D.,3μm,流动相:40%乙醇(0.05%二乙胺)+二氧化碳,流速:4mL/min,柱温:40℃)。
化合物47-2  1H NMR(400MHz,CDCl 3)δ:8.65(s,1H),8.23(d,J=8.0Hz,1H),8.10(d,J=8.4Hz,1H),7.99(d,J=8.0Hz,1H),7.64-7.55(m,2H),6.58(br s,1H),4.66-4.34(m,3H),4.07-3.55(m,10H),3.45-3.18(m,4H),3.08(d,J=4.8Hz,3H),2.57-2.45(m,1H),2.38-2.26(m,1H),1.53(d,J=6.8Hz,3H)。
实施例48-1&48-2
Figure PCTCN2018113683-appb-000137
第一步
将甲基三苯基溴化磷(83.0g,232mmol,2.0eq)和叔丁醇钾(26.01,232mmol,2.0eq)溶于四氢呋喃(1.00L)中,20℃反应1小时,加入化合物48a(10.0g,116.16mmol,1.0eq),20℃反应17小时后,80℃常压蒸馏,馏分为化合物48b。
1H NMR(400MHz,CDCl 3)δ:5.05-4.91(m,2H),4.26(s,2H),3.90(t,J=6.8Hz,2H),2.56(br t,J=6.8Hz,2H)。
第二步
将化合物48b(6.00g,28.5mmol,4%purity,1eq)、化合物48c(5.0g,28.5mmol,1.0eq)、N-碘代琥珀酰亚胺(6.42g,28.5mmol,1.0eq)溶于四氢呋喃(80.0mL),60℃反应17小时,反应完成后,用100毫升饱和硫代硫酸钠水溶液淬灭,乙酸乙酯萃取(150mL×3),有机相无水硫酸钠干燥,过滤,减压旋干,柱层析(1:1乙酸乙酯/石油醚)得化合物48d。
MS-ESI计算值[M+H] +386,实测386。
第三步
将化合物48d(0.2g,519μmol,1.0eq)、氢化钠(41.5mg,1.04mmol,60%purity,2.0eq)溶于N,N-二甲基甲酰胺(10.0mL),25℃反应16小时,反应完成后,加水(1.00mL)淬灭,浓缩,加10.0毫升水稀释, 乙酸乙酯萃取(10.0mL×3),合并有机相,无水硫酸钠干燥,过滤,减压旋干,得48e。
第四步
将化合物48e(0.15g,583μmol,1eq)溶于盐酸/乙酸乙酯(10.0mL,2M),25℃搅拌反应17小时,反应完成后,减压旋干,得48f。
第五步
将化合物48f(90.0mg,465μmol,1.85eq),化合物48g(100mg,251μmol,1.0eq),DIPEA(97.5mg,754μmol,3.0eq)溶于DMSO(5.00mL),使混合溶液80℃反应18小时,经高效液相色谱法提纯得消旋体,进一步经SFC手性分离得到化合物48-1和48-2。
化合物48-1出峰位置:3.301min(手性柱:AS-H 150×4.6mm,I.D.,5μm,流动相:A:CO2,B:乙醇(0.05%二乙胺),保持5%B 0.5min,然后在3.5min内B的含量由5%梯度增加到40%,然后保持40%B 2.5min,最后保持5%B 1.5min流速:3mL/min,柱温:40℃)。
化合物48-1 MS-ESI计算值[M+H] +519,实测值519。
化合物48-1  1H NMR(400MHz,CDCl 3)δ:8.54(s,1H),8.13(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.89(d,J=8.0Hz,1H),7.49(t,J=8.0Hz,1H),7.45(d,J=8.4Hz,1H),6.50(br s,1H),4.93(br s,1H),4.59(br d,J=13.2Hz,1H),4.33(br d,J=6.4Hz,1H),3.94-3.54(m,12H),3.23(d,J=13.6Hz,1H),2.99(d,J=5.2Hz,3H),2.14-2.00(m,1H),1.98-1.88(m,1H),1.42(d,J=6.8Hz,3H),1.29(br d,J=6.4Hz,3H)。
化合物48-2出峰位置:3.608min(手性柱:AS-H 150×4.6mm,I.D.,5μm,流动相:A:CO2,B:乙醇(0.05%二乙胺),保持5%B 0.5min,然后在3.5min内B的含量由5%梯度增加到40%,然后保持40%B 2.5min,最后保持5%B 1.5min流速:3mL/min,柱温:40℃)。
化合物48-2 MS-ESI计算值[M+H] +519,实测值519。
化合物48-2  1H NMR(400MHz,CDCl 3)δ:8.54(s,1H),8.13(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.89(d,J=8.0Hz,1H),7.49(t,J=8.0Hz,1H),7.45(d,J=8.4Hz,1H),6.50(br s,1H),4.93(br s,1H),4.59(br d,J=13.2Hz,1H),4.33(br d,J=6.4Hz,1H),3.94-3.54(m,12H),3.23(d,J=13.6Hz,1H),2.99(d,J=5.2Hz,3H),2.14-2.00(m,1H),1.98-1.88(m,1H),1.42(d,J=6.8Hz,3H),1.29(br d,J=6.4Hz,3H)。
实施例49
Figure PCTCN2018113683-appb-000138
Figure PCTCN2018113683-appb-000139
将化合物35b(100mg,261μmol,1.00eq),24f(92.6mg,522μmol,2.00eq),DIPEA(101mg,782μmol,136μL,3.00eq)溶于DMSO(2.00mL),使混合溶液80℃反应40小时。反应完全,反应液经高效液相色谱法纯化得49。
MS-ESI计算值[M+H] +489,实测值489。
1H NMR(400MHz,CDCl 3)δ:8.62(s,1H),8.18(br d,J=7.6Hz,1H),7.97(d,J=8.4Hz,1H),7.93(br d,J=8.0Hz,1H),7.51(t,J=8.0Hz,1H),7.43(d,J=8.4Hz,1H),6.52(br s,1H),5.63(br s,1H),4.29(br s,1H),3.95-3.57(m,12H),1.70(br d,J=8.4Hz,8H),1.41(d,J=6.8Hz,3H)。
实施例50-1&50-2
Figure PCTCN2018113683-appb-000140
将化合物35b(80.0mg,208μmol,1.00eq)、50a(44.9mg,250μmol,1.20eq)和DIPEA(80.8mg,625μmol,109μL,3eq)溶于DMSO(2.00mL),混合溶液在80℃反应22小时。反应完全,反应液经高效液相色谱法纯化得消旋体,进一步经SFC拆分得50-1和50-2。
50-1出峰位置:3.491min(手性柱:OJ-3 100×4.6mm,I.D.,3μm,流动相:A:CO2,B:甲醇(0.05%二乙胺),在4.5min内B的含量由5%梯度增加到40%,然后保持5%B 1.0min流速:2.8mL/min,柱温: 40℃)。
50-1 MS-ESI计算值[M+H] +491,实测值491。
50-1  1H NMR(400MHz,CDCl 3)δ:8.63(s,1H),8.18(br d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.94(d,J=80Hz,1H),7.51(t,J=7.2Hz,1H),7.46(d,J=8.0Hz,1H),6.61(br s,1H),5.69(br s,1H),4.33(br d,J=6.4Hz,1H),3.97-3.59(m,16H),2.07-1.85(m,2H),1.41(d,J=6.8Hz,3H)。
50-2出峰位置:3.846min(手性柱:OJ-3 100×4.6mm,I.D.,3μm,流动相:A:CO2,B:甲醇(0.05%二乙胺),在4.5min内B的含量由5%梯度增加到40%,然后保持5%B 1.0min流速:2.8mL/min,柱温:40℃)。
50-2 MS-ESI计算值[M+H] +491,实测值491。
50-2  1H NMR(400MHz,CDCl 3)δ:8.63(s,1H),8.18(br d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.94(d,J=80Hz,1H),7.51(t,J=7.2Hz,1H),7.46(d,J=8.0Hz,1H),6.61(br s,1H),5.69(br s,1H),4.33(br d,J=6.4Hz,1H),3.97-3.59(m,16H),2.07-1.85(m,2H),1.41(d,J=6.8Hz,3H)。
实施例51
Figure PCTCN2018113683-appb-000141
将化合物35b(100mg,261μmol,1.00eq),1i(46.8mg,313μmol,1.20eq),DIPEA(101mg,782μmol,136μL,3.00eq)溶于DMSO(2.00mL),使混合溶液80℃反应18小时。反应完全,反应液经高效液相色谱法纯化得51。
MS-ESI计算值[M+H] +461,实测值461。
1H NMR(400MHz,CDCl 3)δ:8.60(s,1H),8.18(br d,J=8.0Hz,1H),7.98(d,J=8.4Hz,1H),7.92(br d,J=8.0Hz, 1H),7.51(t,J=8.0Hz,1H),7.43(d,J=8.4Hz,1H),6.46(br s,1H),5.62(br s,1H),4.30(br s,1H),4.05-3.59(m,12H),1.40(d,J=6.8Hz,3H),0.80-0.73(m,2H),0.61(br s,2H)。
实施例52
Figure PCTCN2018113683-appb-000142
第一步
将化合物52a(900mg,2.11mmol,1eq)溶于甲醇(20.0mL),加入氢氧化钯(591mg,421μmol,0.2eq),在氢气50psi下,使混合溶液50℃反应42小时。然后过滤,减压旋干,得化合物52b。
1H NMR(400MHz,CDCl 3)δ:3.80-3.58(m,6H),3.52-3.50(m,1H),3.51(s,1H),3.50-3.38(m,4H),1.49(s,9H)。
第二步
将化合物52b(500mg,2.02mmol,1eq)的盐酸乙酸乙酯溶液(20.0mL,2M),在25℃反应1小时,反 应完成后,减压旋干,得粗品化合物52c。
1H NMR(400MHz,CD 3OD)δ:4.15-4.10(m,2H),3.71-3.63(m,4H),3.31(s,2H),3.23-3.17(m,2H)。
第三步
将化合物52c(350mg,1.91mmol,1eq),52d(531mg,1.33mmol,0.7eq),DIPEA(668mg,5.17mmol,0.9mL,2.71eq)溶于DMSO(7.00mL),使混合溶液80℃反应23小时,经高效液相色谱法提纯得52e。
MS-ESI计算值[M+H] +509,实测值509。
1H NMR(400MHz,CDCl 3)δ:8.61(s,1H),8.21(br d,J=8.0Hz,1H),8.08(d,J=8.4Hz,1H),7.99(d,J=8.0Hz,1H),7.60-7.54(m,2H),4.46(br d,J=4.8Hz,1H),4.34-3.45(m,19H),3.07(d,J=4.8Hz,3H),1.52(d,J=6.8Hz,3H)。
第四步
将化合物52e(300mg,590μmol,1eq)溶于乙醇(15.0mL)和水(15.0mL),加入氢氧化钠(118mg,2.95mmol,5eq),使混合溶液105℃反应285小时,经高效液相色谱法提纯得黄色固体52f。
MS-ESI计算值[M+H] +496,实测值496。
1H NMR(400MHz,DMSO-d 6)δ:8.78(s,1H),8.28(br d,J=8.0Hz,1H),8.23(d,J=8.4Hz,1H),8.05(br d,J=8.0Hz,1H),7.68(d,J=8.4Hz,1H),7.58(t,J=8.0Hz,1H),4.66-4.39(m,3H),3.96-3.41(m,14H),1.38(d,J=6.8Hz,3H)。
第五步
将化合物52f(78mg,157μmol,1eq),氯化铵(33.7mg,630μmol,4eq),DIPEA(81.4mg,630μmol,110μL,4eq),EDCI(60.4mg,315μmol,2eq),HOBt(42.5mg,315μmol,2eq)溶于二氯甲烷(10.0mL)中,使混合溶液30℃反应16小时,反应完成后,加水(3mL)淬灭后浓缩掉溶剂,加水(20.0mL)稀释后用乙酸乙酯(20.0mL×3)和甲醇(5.00mL)萃取,合并的有机相用无水硫酸钠干燥,过滤,减压旋干,得52g。
MS-ESI计算值[M+H] +495,实测值495。
第六步
将化合物52g(87.4mg,177μmol,1eq),TosCl(50.5mg,265μmol,1.5eq),氢化钠(35.3mg,884μmol,60%纯度,5eq)溶于DMF(4.00mL),使混合溶液30℃反应24小时,经高效液相色谱法提纯得52。
MS-ESI计算值[M+H] +477,实测值477。
1H NMR(400MHz,CDCl 3)δ:8.62(s,1H),8.19(br d,J=8.0Hz,1H),8.00(d,J=8.4Hz,1H),7.94(br d,J=8.0Hz,1H),7.54-7.45(m,2H),6.54(br s,1H),5.73(br s,1H),4.57(d,J=6.8Hz,2H),4.47-4.35(m,3H),4.21-4.07(m,2H),3.96-3.65(m,10H),1.43(d,J=6.8Hz,3H)。
实施例53-1&53-2
Figure PCTCN2018113683-appb-000143
第一步
在零度下,将戴斯马丁试剂(1.31g,3.08mmol,1.5eq)加入化合物53a(0.5g,2.06mmol,1.0eq)的二氯甲烷(5mL)溶液中,25℃下反应4小时。反应液浓缩,粗品用10.0mL的1M氢氧化钠溶液稀释,乙酸乙酯萃取(20.0mL×3),有机相用无水硫酸钠干燥,过滤,减压旋干,柱层析(1:4乙酸乙酯/石油醚)得53b。
1H NMR(400MHz,CDCl 3)δ:3.71(br t,J=4.8Hz,2H),3.55(s,2H),3.50(br d,J=4.8Hz,2H),3.21-3.08(m,2H),3.06-2.91(m,2H),1.49(s,9H)。
第二步
将化合物53b(0.25g,1.04mmol,1eq)溶于盐酸/乙酸乙酯(10.0mL,2M),30℃搅拌反应15小时,反应完成后,减压旋干,得53c,粗品直接用于下一步。
第三步
将化合物53d(400mg,1.01mmol,1.0eq),化合物53c(200mg,1.13mmol,1.12eq),DIPEA(390mg,302umol,3.0eq)溶于DMSO(10.0mL),使混合溶液80℃反应17小时,溶液用20.0毫升水稀释,乙酸乙酯萃取(20mL×3),有机相无水硫酸钠干燥,过滤,减压旋干,柱层析(100%乙酸乙酯)得53e。
1H NMR(400MHz,CDCl 3)δ:8.53(s,1H),8.15(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.89(d,J=8.0Hz,1H),7.52-7.48(m,1H),7.48-7.44(m,1H),6.41(br s,1H),4.34(br d,J=7.2Hz,1H),4.03-3.90(m,4H),3.82-3.59(m,8H),3.13-3.00(m,4H),2.99(d,J=4.8Hz,3H),1.42(d,J=6.8Hz,3H)。
第四步
将盐酸羟胺(41.5mg,596.9μmol,3eq)加到化合物53e(100mg,199μmmol,1.0eq)的甲醇(3.00mL)溶液中,70℃下搅拌1小时。反应液浓缩,粗品用5毫升水稀释,乙酸乙酯萃取(10.0mL×3),有机相经无水硫酸钠干燥,过滤,减压旋干,制备层析板纯化(100%乙酸乙酯)得53f。
第五步
将氢氧化钠(101mg,2.51mmol,10.0eq),对甲苯磺酰氯(479mg,2.51mmol,10.0eq)依次加入化合物53f(130mg,251μmol,1.0eq)的丙酮(40mL)和水(4mL)溶液中,30℃反应16小时,反应液浓缩,粗品用10毫升水稀释,乙酸乙酯洗涤(20mL×3),有机相弃去,水相浓缩,制备层析板纯化(1:10甲醇/乙酸乙酯)得消旋体,进一步经SFC手性分离得到化合物53-1和53-2。
化合物53-1出峰位置:2.589min(手性柱:AS-3 150×4.6mm,流动相:A:CO2,B:甲醇(0.05%二乙胺),流速:25mL/min,柱温:40℃)。
化合物53-1 MS-ESI计算值[M+H] +518,实测值518。
化合物53-1  1H NMR(400MHz,CDCl 3)δ:8.55(s,1H),8.13(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.90(d,J=8.0Hz,1H),7.53-7.43(m,2H),6.52(br s,1H),5.55(br s,1H),4.34(br d,J=6.4Hz,1H),3.97-3.58(m,12H),3.45-3.37(m,2H),2.98(d,J=5.2Hz,3H),2.56-2.41(m,2H),1.42(d,J=6.8Hz,3H)。
化合物53-2出峰位置:5.877min(手性柱:AS-3 150×4.6mm,流动相:A:CO2,B:甲醇(0.05%二乙胺),流速:25mL/min,柱温:40℃)。
化合物53-2 MS-ESI计算值[M+H] +518,实测值518。
化合物53-1  1H NMR(400MHz,CDCl 3)δ:8.55(s,1H),8.13(d,J=8.0Hz,1H),7.99(d,J=8.4Hz,1H),7.90(d,J=8.0Hz,1H),7.53-7.43(m,2H),6.52(br s,1H),5.55(br s,1H),4.34(br d,J=6.4Hz,1H),3.97-3.58(m,12H),3.45-3.37(m,2H),2.98(d,J=5.2Hz,3H),2.56-2.41(m,2H),1.42(d,J=6.8Hz,3H)。
实施例54
Figure PCTCN2018113683-appb-000144
Figure PCTCN2018113683-appb-000145
第一步
将化合物1f(150mg,501μmol,1.00eq)、54a(82.1mg,451μmol,0.90eq)、双(三苯基膦)二氯化钯(17.6mg,25.1μmol,0.05eq)和碳酸钠(106mg,1.0mmol,2.00eq)溶于水(2.00mL)和1,4-二氧六环(4.00mL)中,在氮气保护下,70℃反应15.5小时,反应完成后,浓缩掉溶剂,加水(5mL)稀释后用乙酸乙酯萃取(10.0mL×3),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,得54b。
第二步
将化合物54b(100mg,249μmol,1.00eq)、1i(33.9mg,299μmol,1.2eq)和DIPEA(96.7mg,748μmol,130μl,3eq)溶于DMSO(3.00mL),混合溶液在80℃反应17小时。反应完全,反应液经高效液相色谱法纯化得54。
MS-ESI计算值[M+H] +478,实测值478。
1H NMR(400MHz,CDCl 3)δ:8.20-8.13(m,2H),8.00(d,J=8.4Hz,1H),7.45(d,J=8.4Hz,1H),7.01(d,J=8.4Hz,1H),4.79(s,2H),4.36(br s,1H),4.08-3.63(m,15H),1.47(d,J=6.4Hz,3H),0.85(s,2H),0.70(br s,2H)。
实施例55
Figure PCTCN2018113683-appb-000146
Figure PCTCN2018113683-appb-000147
第一步
将化合物1f(100mg,334μmol,1.00eq),化合物55a(65.5mg,334μmol,1.00eq),双(三苯基膦)二氯化钯(11.7mg,16.7μmol,0.05eq)和碳酸钠(70.9mg,669μmol,2.00eq)溶于水(1.00mL)和1,4-二氧六环(3.00mL)中,在氮气保护下,80℃反应15小时,反应完成后,浓缩掉溶剂,加水(15.0mL)稀释后用乙酸乙酯萃取(50.0mL×3),合并的有机相用无水硫酸钠干燥,过滤,减压旋干,得55b。
第二步
将化合物55b(168mg,404μmol,1.00eq),1i(54.4mg,364μmol,0.90eq)和DIPEA(157mg,1.21mmol,211μL,3.00eq)溶于DMSO(3.00mL),混合溶液在80℃反应15小时。反应完全后,浓缩掉溶剂,得55c,粗品直接用于下一步。
第三步
将化合物55c(99.0mg,201μmol,1.00eq),55d(24.5mg,363μmol,1.80eq),DIPEA(104mg,806μmol,140μL,4.00eq),EDCI(57.9mg,302μmol,1.50eq),HOBt(40.8mg,302μmol,1.50eq)溶于DMSO(4.00mL),使混合溶液30℃反应22小时。反应完全,反应液经高效液相色谱法纯化得55。
MS-ESI计算值[M+H] +505,实测值505。
1H NMR(400MHz,CDCl 3)δ:8.75(br s,1H),8.51(br d,J=8.0Hz,1H),7.94(br d,J=8.4Hz,1H),7.76(br s,1H),7.50(br d,J=8.4Hz,1H),7.03(br d,J=8.4Hz,1H),4.29(br s,1H),3.97(s,3H),3.96-3.51(m,12H),2.98(br d,J=4.4Hz,3H),1.38(br d,J=6.8Hz,3H),0.76(br s,2H),0.61(br s,2H)。
实施例56
Figure PCTCN2018113683-appb-000148
将化合物56c(99.0mg,201μmol,1.00eq),氯化铵(43.1mg,806μmol,4.00eq),DIPEA(104mg,806μmol,140μL,4.00eq),EDCI(57.9mg,302μmol,1.50eq),HOBt(40.8mg,302μmol,1.50eq)溶于DMSO(4.00mL),使混合溶液30℃反应22小时。反应完全,反应液经高效液相色谱法纯化得56。
MS-ESI计算值[M+H] +491,实测值491。
1H NMR(400MHz,CDCl 3)δ:8.76(d,J=2.4Hz,1H),8.55(dd,J=2.4,8.8Hz,1H),7.93(d,J=8.8Hz,1H),7.67(br s,1H),7.50(d,J=8.8Hz,1H),7.06(d,J=8.8Hz,1H),5.75(br s,1H),4.28(br d,J=5.6Hz,1H),3.99(s,3H),3.97-3.36(m,12H),1.38(d,J=6.8Hz,3H),0.79-0.73(m,2H),0.61(br s,2H)。
实验例1:体外评价mTOR激酶抑制活性
实验材料:
本实验测试于DiscoverX,所有材料和方法均来自DiscoverX。
实验操作:
激酶活性分析。
1将标记的mTOR激酶稳定的表达于HEK-293细胞中。
3室温条件下,用生物素化小分子配体处理链霉亲和素磁珠30分钟,产生用于激酶分析的亲和树脂;
4配体珠用过量的生物素阻断并用缓冲液(1%牛血清白蛋白,0.05%吐温20毫升,1毫升二硫苏糖醇)洗涤,清洗掉未结合的配体和非特异性结合的配体;
5激酶配体亲和珠,组装,测试化合物三者的结合反应均在缓冲液(20%封闭缓冲液,0.17x磷酸盐缓冲液,0.05%吐温20,6毫升二硫苏糖醇)中实现;
6测试化合物溶于二甲基亚砜中;
7所有用于测量的化合物都是溶解于DMSO,然后将化合物直接稀释到浓度为0.9%。
8溶液置于384孔聚丙烯板,每个体积为0.02毫升;
9室温条件下,摇晃1小时;
10用缓冲液洗(1x PBS,0.05%吐温20)洗涤
11亲和珠再次悬浮于缓冲液(1x PBS,0.05%吐温20,0.5μm的非生物素亲和配体)并在室温孵化30分钟。
12用qPCR测量洗脱液中激酶的浓度。
实验结果:
表1 mTORC1和mTORC2激酶复合物活性测试结果
Figure PCTCN2018113683-appb-000149
Figure PCTCN2018113683-appb-000150
Figure PCTCN2018113683-appb-000151
“ND”表示未检测。
结论:本发明化合物具有显著甚至意料不到的mTOR激酶抑制活性。
实验例2细胞增殖抑制活性评价:
实验目的:检测待测化合物对细胞增殖抑制活性。
实验原理:Cell-Titer-Glo试剂中的荧光素酶利用荧光素、氧和ATP作为反应底物,产生氧化荧光素,并以光的形式释放能量。由于荧光素酶反应需要ATP,因而反应产生的光的总量和反应细胞活力的ATP数总量成正比。
实验材料:
细胞系:MCF-7细胞系(ATCC-CRL-22),HT-29细胞系(ATCC-HTB-38),OE21(ECACC-96062201),NCI-N87细胞系(ATCC-CRL-5822)
细胞培养基:(RPMI 1640培养基(Invitrogen#1868546;10%血清Invitrogen#1804958;左旋谷酰胺1×,Invitrogen#1830863;双抗Hyclone#J170012))
Cell
Figure PCTCN2018113683-appb-000152
发光法细胞活力检测试剂盒(Promega#G7573)
384孔细胞培养板(Greiner#E15103MA)
化合物板(LABCYTE#0006346665)
CO 2培养箱(Thermo#371)
Vi-cell细胞计数仪(Beckman Coulter)
移液器(Eppendorf)
移液管(Greiner)
移液枪(Eppendorf)
多功能酶标仪(Envision Reader)
ECHO Liquid-handling workstation(Labcyte-ECHO555)
实验步骤和方法:
2.1第1天:
按照细胞种板示意图在384或96孔板中分别按每孔1000个细胞,25μL每孔的密度种板,边缘孔不种细胞补25μLPBS。
2.2第0天:
(1)化合物母液为10mM,用DMSO稀释化合物使其初始浓度为4mM。在化合物化合物母液板板上加入化合物,每孔9μL。
(2)用ECHO液体工作站做化合物稀释并向细胞板每孔加入125nL化合物,第2列和23列细胞孔每孔加125nL DMSO,第1列和24列PBS孔每孔加125nL DMSO。
(3)细胞板每孔补加25μL培养基,最终细胞板每孔为50μL,化合物浓度为1μM,3倍稀释,10个浓度,左右复孔,DMSO终浓度为0.25%。
2.3加好化合物后,1000rpm离心1min,将细胞板放置于37℃、5%CO 2培养箱中培养3天。
2.4第3天:
从培养箱中取出细胞板,在室温下平衡30分钟。向每孔加入25μL Cell-Titer-Glo试剂,振摇一分钟使它被充分混匀,1000rpm离心1分钟。10分钟后,在PerkinElmer Envision上读板,设置荧光读取时间0.2秒。试验结果:试验结果如表2
表2:本发明化合物体外细胞增殖抑制活性筛选试验结果
Figure PCTCN2018113683-appb-000153
Figure PCTCN2018113683-appb-000154
Figure PCTCN2018113683-appb-000155
“ND”表示未检测。
结论:本发明化合物对MCF-7,N87和OE-21细胞增殖抑制活性明显,对HT-29细胞具有一定的增殖抑制活性。
实验例3:药代动力学评价
1.实验方法
受试化合物与5%DMSO/95%10%聚氧乙烯蓖麻油(Cremophor EL)混合,涡旋并超声,制备得到1mg/mL近似澄清溶液,微孔滤膜过滤后备用。选取18至20克的Balb/c雌性小鼠,静脉注射给予候选化合物溶液,剂量为1或2mg/kg。受试化合物与1%吐温80,9%聚乙二醇400,90%水溶液混合,涡旋并超声,制备得到1mg/mL近似澄清溶液,微孔滤膜过滤后备用。选取18至20克的Balb/c雌性小鼠,口服给予候选化合物溶液,剂量为2或10mg/kg。收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
测试结果:
测试结果见表3~表8。
表3 实施例化合物血浆中的药物代谢动力学(PK)参数
Figure PCTCN2018113683-appb-000156
Figure PCTCN2018113683-appb-000157
表4 实施例化合物血浆中的PK参数
Figure PCTCN2018113683-appb-000158
Figure PCTCN2018113683-appb-000159
表5 实施例化合物血浆中的PK参数
Figure PCTCN2018113683-appb-000160
表6 实施例化合物血浆中的PK参数
Figure PCTCN2018113683-appb-000161
Figure PCTCN2018113683-appb-000162
表7 实施例化合物血浆中的PK参数
Figure PCTCN2018113683-appb-000163
Figure PCTCN2018113683-appb-000164
表8 实施例化合物血浆中的PK参数
Figure PCTCN2018113683-appb-000165
“--”是指未测试或未获得数据。
试验结论:受试化合物展现了与参比化合物相同甚至更优的药代动力学性质;本发明的化合物生物利用度接近100%,是很好的可开发的口服给药的分子
实验例4:人乳腺癌MCF-7细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学研究:
实验目的:研究待测化合物对人乳腺癌MCF-7细胞皮下异种移植瘤在BALB/c裸小鼠模型体内的药效
实验动物:雌性BALB/c裸小鼠,6-8周龄,体重18-22克;供应商:上海西普尔-必凯实验动物有限公司实验方法与步骤:
4.1细胞培养
人乳腺癌MCF-7细胞(ECACC,货号:86012803)体外单层培养,培养条件为EMEM(EBSS)+2mM谷氨酸+1%Non Essential Amino Acids(NEAA)培养基中加10%胎牛血清,100U/mL青霉素和100μg/mL链霉素,37℃5%CO 2孵箱培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%,数量到达要求时,收取细胞,计数,接种。
4.2肿瘤细胞接种(肿瘤接种)
在接种细胞前3天,将***片(0.18mg)接种于每只小鼠的左后背。将0.2mL(1×10 7个)MCF-7细胞(加基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到142mm 3时开始分组给药。
4.3受试物的配制:
受试化合物配制成0.75mg/mL、1.5mg/mL和3mg/mL的澄清溶液,溶媒为5%DMSO+30%聚乙二醇300+65%水。
4.4肿瘤测量和实验指标
实验指标是考察肿瘤生长是否被抑制、延缓或治愈。每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用TGI(%)或相对肿瘤增殖率T/C(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)=【1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)】×对照组。
相对肿瘤增殖率T/C(%):计算公式如下:T/C%=TRTV/CRTV组开始治疗时平(TRTV:治疗组RTV;CRTV:阴性对照组RTV)。根据肿瘤测量的结果计算出相对肿瘤体积(relative tumor volume,RTV),计算公式为RTV=V t/V 0,其中V 0是分组给药时(即d 0)测量所得平均肿瘤体积,V t为某一次测量时的平均肿瘤体积,TRTV与CRTV取同一天数据。
在实验结束后将检测肿瘤重量,并计算T/重量百分比,T重量和C重量分别表示给药组和溶媒对照组的瘤重。
4.5统计分析
统计分析,包括每个组的每个时间点的肿瘤体积的平均值和标准误(SEM)。治疗组在试验结束时给药后第21天表现出最好的治疗效果,因此基于此数据进行统计学分析评估组间差异。两组间比较用T-test进行分析,三组或多组间比较用one-way ANOVA进行分析,如果F值有显著性差异,应用Games-Howell法进行检验。如果F值无显著性差异,应用Dunnet(2-sided)法进行分析。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
4.6试验结果
在MCF-7移植瘤模型上,部分化合物药效与AZD2014相当。
表9 化合物对人乳腺癌MCF-7细胞皮下异种移植瘤模型的抑瘤效果
Figure PCTCN2018113683-appb-000166
注:
“--”不需计算
a.平均值±SEM。
b.肿瘤生长抑制由T/C和TGI(TGI(%)=[1-(T 21-T 0)/(V 21-V 0)]×100)计算。
c.p值根据肿瘤体积。
4.7试验结论
与溶媒组相比,在人乳腺癌裸鼠移植瘤模型中,AZD2014在15mg/kg和实施例23在30mg/kg的剂量下与溶剂对照组相比有显著性差异,TGI分别为104%和98%。

Claims (21)

  1. 式(IV)所示化合物、其药学上可接受的盐及其异构体,
    Figure PCTCN2018113683-appb-100001
    其中,
    R 1为H;
    R 2为Me;
    或者,R 1、R 2和吗啉环上的N形成一个5~6元杂环烷基;
    R 3选自NH 2
    Figure PCTCN2018113683-appb-100002
    C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-,其中所述C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-任选被1、2或3个R取代;
    n选自1和2;
    环A选自苯基和6~10元杂芳基;
    R 4为H;
    R 5为H;
    或者,R 4与R 5连接在一起形成一个5~6元杂环烷基;
    D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-和-O-,且D 1、D 2、D 3和D 4至少一个不为单键,其中所述-CH 2-或-CH 2CH 2-任选被1或2个R取代;
    D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-和-NH-,且D 5、D 6、D 7和D 8至少一个不为单键,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
    T 1选自CH和N;
    T 2选自-CH 2-、-NH-、-O-、
    Figure PCTCN2018113683-appb-100003
    -S-和-C(=O)NH-,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
    R分别独立地选自F、Cl、Br、I、OH、NH 2、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基,其中所述C 1-3烷基、C 1-3烷氧基和C 3-6环烷基任选被1或2个R’取代;
    R’分别独立地选自F、Cl、Br、I、OH和NH 2
    所述C 1-3杂烷基、5~6元杂芳基和6~10元杂芳基分别包含1、2或3个独立选自-O-、-S-、-NH-、N、-C(=O)-、-C(=O)NH-和-C(=S)NH-的杂原子或杂原子团。
  2. 根据权利要求1所述化合物、其药学上可接受的盐及其异构体,其选自
    Figure PCTCN2018113683-appb-100004
    其中,
    R 1为H;
    R 2为Me;
    或者,R 1、R 2和吗啉环上的N形成一个5~6元杂环烷基;
    R 3选自NH 2
    Figure PCTCN2018113683-appb-100005
    C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-,其中所述C 1-3杂烷基、5~6元杂芳基和C 3-6环烷基-NH-C(=O)-任选被1、2或3个R取代;
    环A选自苯基和6~10元杂芳基;
    R 4选自H;
    R 5选自H;
    或者,R 4与R 5连接在一起形成一个5~6元杂环烷基;
    D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-和-O-,且D 1、D 2、D 3和D 4至少一个不为单键,其中所述-CH 2-或-CH 2CH 2-任选被1或2个R取代;
    D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-和-NH-,且D 1、D 2、D 3和D 4至少一个不为单键,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
    T 1选自CH和N;
    T 2选自-CH 2-、-NH-、-O-、
    Figure PCTCN2018113683-appb-100006
    -S-和-C(=O)NH-,其中所述-CH 2-任选被1或2个R取代,-NH-任选被R取代;
    R分别独立地选自F、Cl、Br、I、OH、NH 2、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基,其中所述C 1-3烷基、C 1-3烷氧基和C 3-6环烷基任选被1或2个R’取代;
    R’分别独立地选自F、Cl、Br、I、OH和NH 2
    所述C 1-3杂烷基、5~6元杂芳基和6~10元杂芳基分别包含1、2或3个独立选自-O-、-S-、-NH-、N、-C(=O)-、-C(=O)NH-和-C(=S)NH-的杂原子或杂原子团。
  3. 根据权利要求2所述化合物、其药学上可接受的盐及其异构体,其中,R分别独立地选自F、Cl、Br、I、OH、NH 2、Me、Et、
    Figure PCTCN2018113683-appb-100007
    其中所述Me、Et、
    Figure PCTCN2018113683-appb-100008
    任选被1或2个R’取代。
  4. 根据权利要求3所述化合物、其药学上可接受的盐及其异构体,其中,R分别独立地选自F、Cl、Br、I、OH、NH 2、Me、CF 3、Et、
    Figure PCTCN2018113683-appb-100009
  5. 根据权利要求1~4任意一项所述化合物、其药学上可接受的盐及其异构体,其中,结构单元
    Figure PCTCN2018113683-appb-100010
    选自
    Figure PCTCN2018113683-appb-100011
  6. 根据权利要求1~4任意一项所述化合物、其药学上可接受的盐及其异构体,其中,R 3选自
    Figure PCTCN2018113683-appb-100012
    Figure PCTCN2018113683-appb-100013
    1H-吡唑基和1H-1,2,4-***基,其中所述NH 2
    Figure PCTCN2018113683-appb-100014
    1H-吡唑基和1H-1,2,4-***基任选被1、2或3个R取代。
  7. 根据权利要求6所述化合物、其药学上可接受的盐及其异构体,其中,R 3选自NH 2
    Figure PCTCN2018113683-appb-100015
    Figure PCTCN2018113683-appb-100016
    其中所述NH 2
    Figure PCTCN2018113683-appb-100017
    Figure PCTCN2018113683-appb-100018
    任选被1、2或3个R取代。
  8. 根据权利要求7所述化合物、其药学上可接受的盐及其异构体,其中,R 3选自NH 2
    Figure PCTCN2018113683-appb-100019
    Figure PCTCN2018113683-appb-100020
  9. 根据权利要求1~4任意一项所述化合物、其药学上可接受的盐及其异构体,其中,环A选自苯基、苯并[d]噁唑、喹啉基和喹唑啉基。
  10. 根据权利要求9所述化合物、其药学上可接受的盐及其异构体,其中,环A选自
    Figure PCTCN2018113683-appb-100021
    Figure PCTCN2018113683-appb-100022
  11. 根据权利要求1或10所述化合物、其药学上可接受的盐及其异构体,其中,结构单元
    Figure PCTCN2018113683-appb-100023
    选自
    Figure PCTCN2018113683-appb-100024
    Figure PCTCN2018113683-appb-100025
  12. 根据权利要求2或10所述化合物、其药学上可接受的盐及其异构体,其中,结构单元
    Figure PCTCN2018113683-appb-100026
    选自
    Figure PCTCN2018113683-appb-100027
    Figure PCTCN2018113683-appb-100028
  13. 根据权利要求1~4任意一项所述化合物、其药学上可接受的盐及其异构体,其中,D 1、D 2、D 3和D 4分别独立地选自单键、-CH 2-、-CH 2CH 2-、-O-和
    Figure PCTCN2018113683-appb-100029
    且D 1、D 2、D 3和D 4至少一个不为单键。
  14. 根据权利要求1~4任意一项所述化合物、其药学上可接受的盐及其异构体,其中,D 5、D 6、D 7和D 8分别独立地选自单键、-CH 2-、-O-、-NH-、
    Figure PCTCN2018113683-appb-100030
    且D 5、D 6、D 7和D 8至少一个不为单键。
  15. 根据权利要求1~4任意一项所述化合物、其药学上可接受的盐及其异构体,其中,结构单元
    Figure PCTCN2018113683-appb-100031
    选自
    Figure PCTCN2018113683-appb-100032
    Figure PCTCN2018113683-appb-100033
  16. 根据权利要求1~9任意一项所述化合物、其药学上可接受的盐及其异构体,其选自:
    Figure PCTCN2018113683-appb-100034
    其中,
    R 1、R 2、R 3、R 4和R 5如权利要求1~7所定义;
    X选自-CH 2-、-CF 2-、
    Figure PCTCN2018113683-appb-100035
    -NH-、
    Figure PCTCN2018113683-appb-100036
    和-S-;
    Y选自-CH 2-和-CH 2CH 2-。
  17. 根据权利要求1或3~9任意一项所述化合物、其药学上可接受的盐及其异构体,其选自:
    Figure PCTCN2018113683-appb-100037
    其中,
    R 1、R 2、R 3、R 4和R 5如权利要求1或3~9所定义。
  18. 下式所示化合物、其药学上可接受的盐及其异构体,
    Figure PCTCN2018113683-appb-100038
    Figure PCTCN2018113683-appb-100039
    Figure PCTCN2018113683-appb-100040
    Figure PCTCN2018113683-appb-100041
  19. 一种药物组合物,包括治疗有效量的根据权利要求1~18任意一项所述的化合物、其药学上可接受的盐及其异构体作为活性成分以及药学上可接受的载体。
  20. 根据权利要求1~18任意一项所述的化合物、其药学上可接受的盐及其异构体或根据权利要求19所述的药物组合物在制备治疗与mTOR双激酶相关疾病的药物中的应用。
  21. 根据权利要求1~18任意一项所述的化合物、其药学上可接受的盐及其异构体或根据权利要求19所述的药物组合物在制备治疗乳腺癌,乳腺癌,头颈癌,结直肠癌药物中的应用。
PCT/CN2018/113683 2017-11-06 2018-11-02 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物 WO2019085996A1 (zh)

Priority Applications (6)

Application Number Priority Date Filing Date Title
BR112020008991-5A BR112020008991A2 (pt) 2017-11-06 2018-11-02 compostos de piridopirimidina que atuam como inibidores duais de mtorc 1/2
US16/760,740 US11377444B2 (en) 2017-11-06 2018-11-02 Pyridopyrimidine compounds acting as mTORC 1/2 dual inhibitors
EP18872475.1A EP3712157A4 (en) 2017-11-06 2018-11-02 PYRIDOPYRIMIDINE COMPOUNDS ACTING AS DOUBLE KINASE MTORC 1/2 INHIBITORS
JP2020524756A JP7340519B2 (ja) 2017-11-06 2018-11-02 mTORC1/2二重阻害剤としてのピリドピリミジン系化合物
CN201880071134.9A CN111315750B (zh) 2017-11-06 2018-11-02 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物
RU2020118222A RU2771201C2 (ru) 2017-11-06 2018-11-02 ПИРИДОПИРИМИДИНОВЫЕ СОЕДИНЕНИЯ, ВЫПОЛНЯЮЩИЕ ФУНКЦИЮ ДВОЙНЫХ ИНГИБИТОРОВ mTORC 1/2

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201711080753.8 2017-11-06
CN201711080753 2017-11-06
CN201810136962.8 2018-02-09
CN201810136962 2018-02-09
CN201810661825.6 2018-06-25
CN201810661825 2018-06-25

Publications (1)

Publication Number Publication Date
WO2019085996A1 true WO2019085996A1 (zh) 2019-05-09

Family

ID=66331374

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/113683 WO2019085996A1 (zh) 2017-11-06 2018-11-02 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物

Country Status (7)

Country Link
US (1) US11377444B2 (zh)
EP (1) EP3712157A4 (zh)
JP (1) JP7340519B2 (zh)
CN (1) CN111315750B (zh)
BR (1) BR112020008991A2 (zh)
RU (1) RU2771201C2 (zh)
WO (1) WO2019085996A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020147842A1 (zh) * 2019-01-18 2020-07-23 南京明德新药研发有限公司 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用
WO2020224585A1 (zh) * 2019-05-06 2020-11-12 南京明德新药研发有限公司 一种mTORC1/2双激酶活性抑制剂的盐型、晶型及其制备方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101360746A (zh) * 2005-11-22 2009-02-04 库多斯药物有限公司 作为mTOR抑制剂的吡啶并-、吡唑并-和嘧啶并-嘧啶衍生物
CN101558067A (zh) * 2006-08-23 2009-10-14 库多斯药物有限公司 作为mtor抑制剂的2-甲基吗啉吡啶并、吡唑并和嘧啶并-嘧啶衍生物
CN103030653A (zh) * 2011-09-30 2013-04-10 上海恒瑞医药有限公司 杂芳基并嘧啶类衍生物及其可药用盐、其制备方法及其在医药上的应用
US20170281637A1 (en) 2006-08-23 2017-10-05 Kudos Pharmaceuticals Limited Pyrido-, Pyrazo- and Pyrimido-Pyrimidine Derivatives as mTOR Inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5161102B2 (ja) * 2005-11-22 2013-03-13 クドス ファーマシューティカルズ リミテッド mTOR阻害剤としてのピリドピリミジン、ピラゾピリミジンおよびピリミドピリミジン誘導体
JP2011529920A (ja) 2008-07-31 2011-12-15 ジェネンテック, インコーポレイテッド ピリミジン化合物、組成物及び使用方法
GB201004200D0 (en) * 2010-03-15 2010-04-28 Univ Basel Spirocyclic compounds and their use as therapeutic agents and diagnostic probes

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101360746A (zh) * 2005-11-22 2009-02-04 库多斯药物有限公司 作为mTOR抑制剂的吡啶并-、吡唑并-和嘧啶并-嘧啶衍生物
CN101558067A (zh) * 2006-08-23 2009-10-14 库多斯药物有限公司 作为mtor抑制剂的2-甲基吗啉吡啶并、吡唑并和嘧啶并-嘧啶衍生物
US20170281637A1 (en) 2006-08-23 2017-10-05 Kudos Pharmaceuticals Limited Pyrido-, Pyrazo- and Pyrimido-Pyrimidine Derivatives as mTOR Inhibitors
CN103030653A (zh) * 2011-09-30 2013-04-10 上海恒瑞医药有限公司 杂芳基并嘧啶类衍生物及其可药用盐、其制备方法及其在医药上的应用

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020147842A1 (zh) * 2019-01-18 2020-07-23 南京明德新药研发有限公司 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用
WO2020224585A1 (zh) * 2019-05-06 2020-11-12 南京明德新药研发有限公司 一种mTORC1/2双激酶活性抑制剂的盐型、晶型及其制备方法

Also Published As

Publication number Publication date
RU2771201C2 (ru) 2022-04-28
EP3712157A4 (en) 2021-04-28
RU2020118222A (ru) 2021-12-08
BR112020008991A2 (pt) 2020-11-17
JP7340519B2 (ja) 2023-09-07
EP3712157A1 (en) 2020-09-23
CN111315750B (zh) 2022-12-23
RU2020118222A3 (zh) 2021-12-08
US20200339568A1 (en) 2020-10-29
CN111315750A (zh) 2020-06-19
JP2021501778A (ja) 2021-01-21
US11377444B2 (en) 2022-07-05

Similar Documents

Publication Publication Date Title
US11639353B2 (en) Cyclobutanes- and azetidine-containing mono and spirocyclic compounds as αV integrin inhibitors
JP7005582B2 (ja) ブルトン型チロシンキナーゼ(btk)インヒビターとしての多フルオロ置換化合物
JP7394074B2 (ja) 治療用化合物
JP6609631B2 (ja) 縮合環ヘテロアリール化合物及びtrk抑制剤としての用途
CN111606908B (zh) Jak抑制剂化合物及其用途
CN105693720B (zh) 作为trk激酶抑制剂的大环化合物
CN104910161B (zh) 作为jak抑制剂的吡唑并嘧啶化合物和方法
EP3919483A1 (en) Benzopyridone heterocyclic compound and use thereof
TW201823249A (zh) Menin-mll相互作用之稠合二環抑制劑
CN110156786A (zh) 嘧啶并环化合物及其制备方法和应用
TW201443023A (zh) 作爲rock抑制劑之酞□酮及異喹啉酮
TW201414737A (zh) 作爲激酶抑制劑之咪唑并三□甲腈
JP6948659B1 (ja) ピリダジニルチアアゾールカルボキシアミド化合物
CN114072407A (zh) 靶向prmt5的化合物
WO2023138583A1 (zh) 杂环类化合物、药物组合物及其应用
TW202110848A (zh) 取代的稠合雙環類衍生物、其製備方法及其在醫藥上的應用
WO2022033562A1 (zh) 用于治疗重症肺炎的jak抑制剂化合物
WO2019085996A1 (zh) 作为mTORC1/2双激酶抑制剂的吡啶并嘧啶类化合物
WO2020259703A1 (zh) 一种吡唑酮并嘧啶类化合物、其制备方法及应用
CN113286594B (zh) 吡啶并嘧啶类化合物在制备治疗鼻咽癌药物中的应用
WO2023236947A1 (zh) 取代的哒嗪-3-甲酰胺化合物作为tyk2抑制剂
WO2024017381A1 (zh) 一种抑制irak4活性的化合物及其应用
TW202333663A (zh) Rxfp1促效劑
WO2022135390A1 (zh) 己酮糖激酶抑制剂及其用途
CN116600808A (zh) 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用

Legal Events

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

Ref document number: 18872475

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020524756

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018872475

Country of ref document: EP

Effective date: 20200608

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112020008991

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112020008991

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20200506