WO2020147798A1 - 作为布鲁顿酪氨酸激酶抑制剂的环状分子 - Google Patents

作为布鲁顿酪氨酸激酶抑制剂的环状分子 Download PDF

Info

Publication number
WO2020147798A1
WO2020147798A1 PCT/CN2020/072551 CN2020072551W WO2020147798A1 WO 2020147798 A1 WO2020147798 A1 WO 2020147798A1 CN 2020072551 W CN2020072551 W CN 2020072551W WO 2020147798 A1 WO2020147798 A1 WO 2020147798A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
pyrazolo
group
amino
Prior art date
Application number
PCT/CN2020/072551
Other languages
English (en)
French (fr)
Inventor
王召印
姚兵
姚元山
李傲
P·K·贾达夫
曹国庆
Original Assignee
明慧医药(上海)有限公司
明慧医药(杭州)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
Priority to CN202310355998.6A priority Critical patent/CN116589465A/zh
Priority to AU2020208128A priority patent/AU2020208128B2/en
Priority to CN202080009961.2A priority patent/CN113710671B/zh
Priority to US17/423,815 priority patent/US20220081445A1/en
Application filed by 明慧医药(上海)有限公司, 明慧医药(杭州)有限公司 filed Critical 明慧医药(上海)有限公司
Priority to MX2021008632A priority patent/MX2021008632A/es
Priority to EP20741672.8A priority patent/EP3912980A4/en
Priority to CA3129841A priority patent/CA3129841C/en
Priority to BR112021014100-6A priority patent/BR112021014100A2/pt
Priority to EA202191995A priority patent/EA202191995A1/ru
Priority to KR1020217025977A priority patent/KR20210135497A/ko
Priority to JP2021542229A priority patent/JP7436994B2/ja
Priority to SG11202107886RA priority patent/SG11202107886RA/en
Publication of WO2020147798A1 publication Critical patent/WO2020147798A1/zh
Priority to IL284916A priority patent/IL284916A/en
Priority to JP2023161762A priority patent/JP2023179562A/ja

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the present invention relates to the field of small molecule drugs. Specifically, the present invention provides a novel molecule with protein tyrosine kinase inhibitory activity, as well as methods for the synthesis and use of such compounds.
  • BTK Bruton's tyrosine kinase
  • the purpose of the present invention is to provide a small molecule compound with BTK inhibitory activity.
  • the first aspect of the present invention provides a compound represented by the following formula (A), or a pharmaceutically acceptable salt thereof,
  • Ring A and Ring B are connected in parallel with each other, and each independently is a substituted or unsubstituted 5-15 membered heterocyclic ring or heteroaromatic ring;
  • Cyc 1 is selected from the following group: substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted Substituted C 3 -C 10 cycloalkenyl, substituted or unsubstituted 5-15 membered heterocyclic group, substituted or unsubstituted 5-15 membered heteroaryl, substituted or unsubstituted C 6 -C 10 aryl base;
  • Cyc 2 is selected from the following group: substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted Substituted C 3 -C 10 cycloalkenyl, substituted or unsubstituted 5-15 membered heterocyclic group, substituted or unsubstituted 5-15 membered heteroaryl, substituted or unsubstituted C 6 -C 10 aryl base;
  • Z is (CR 2 R 3 ) n , n is 0, 1, 2, 3 , 4, 5 or 6;
  • Y is (CR 4 R 5 ) m , and m is 0, 1, 2, 3, 4, 5 or 6;
  • U is (CR 6 R 7 ) r , and r is 0, 1, 2 or 3;
  • n, m and r are not 0 at the same time;
  • R 2 , R 3 , R 4 , R 5 , R 6 and R 7 are each independently selected from the group consisting of H, NH 2 , OH, halogen, substituted or unsubstituted C 1 -C 6 alkyl; or R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are connected to any two carbon atoms and the intervening ring skeleton atoms together to form a C 3 -C 8 carbocyclic ring, or a 4-8 membered heterocycle Ring, wherein the heteroatom of the heterocyclic ring is selected from: S, O, or NR f ; R f is H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 6 -C 20 aryl Group, or C 3 -C 14 heteroaryl; and at least one of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 and R 9 is
  • W is selected from the following group: N, O, S or chemical bond;
  • X is -C(R 8 R 9 )-
  • R 8 is selected from the group consisting of H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 6 -C 20 aryl, or A substituted or unsubstituted C 3 -C 14 heteroaryl group, CO 2 H, C(O)NR f 2 ;
  • R 9 is selected from the group consisting of H, OH or -[C(R 10 )(R 11 )] k -OH;
  • R 9 is OH or -[C(R 10 )(R 11 )] k -OH;
  • R 10 and R 11 are each independently selected from the group consisting of H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 6- C 10 aryl, or substituted or unsubstituted C 3 -C 14 heteroaryl; or R 10 and R 11 and the carbon atoms to which they are connected together form a C 3 -C 8 carbocyclic ring, or a 4-8 membered heterocyclic ring,
  • the heteroatom can be sulfur, oxygen, or NR f ;
  • k is 1, 2, 3, 4, 5 or 6;
  • heteroaromatic ring, heteroaryl group, heterocyclic ring, and heterocyclic group each independently contain 1-4 heteroatoms selected from N, O and S;
  • substitution refers to the group being substituted by one or more (for example, 2, 3, 4, etc.) substituents selected from the following group: halogen, C1-C6 alkyl, halogenated C1 -C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, halogenated C3-C8 cycloalkyl, oxo, -CN, hydroxyl, hydroxy-C1 -C6 alkyl, amino, carboxyl, C6-C10 aryl, halogenated C6-C10 aryl, unsubstituted or substituted with a substituent selected from the group having 1-3 selected from N, S and O Heteroatomic 5-10 membered heteroaryl: halogen, phenyl.
  • L 1 -Cyc 1 -L 2 -Cyc 2 is:
  • R 12 , R 13 , R 14 and R 15 are each independently H, halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted
  • the substituted C 3 -C 10 cycloalkyl group, the substituted or unsubstituted C 3 -C 10 heterocycloalkyl group, and the heteroatoms are selected from N, O, S;
  • L 1 -Cyc 1 -L 2 -Cyc 2 is
  • V is N or -CR 16 -; wherein, R 16 is selected from the group consisting of H, halogen-substituted or unsubstituted C 1 -C 6 alkyl, and substituted or unsubstituted C 3 -C 10 cycloalkyl.
  • the dashed line is a chemical bond or none; each of A 1 , A 2 , A 3 , A 4 , A 5 , A 6 , A 7 , A 8 and A 9 is each independently O, S, N, NH, CH or CH 2 ;
  • the wavy line indicates the connection site
  • each substitutable site of the above group may include a substituent, wherein the definition of the substituent is as described above.
  • the Has a structure selected from the following group:
  • the compound has a structure represented by the following formula (B):
  • R 12 , R 13 , R 14 and R 15 are each independently selected from the following group: H, halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, A substituted or unsubstituted C 3 -C 10 cycloalkyl group, a substituted or unsubstituted 5-15 membered heterocyclic group.
  • the compound has a structure represented by the following formula (C):
  • V is N or -CR 16 -;
  • R 12 , R 13 , R 14 and R 15 are each independently selected from the following group: H, halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, Substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted 5-15 membered heterocyclic group;
  • R 16 is H, halogen substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl.
  • the It is substituted cyclohexyl or epoxypentyl.
  • the compound has a structure represented by the following formula (D), (E) or (F):
  • the compound is selected from the following group:
  • the second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising (1) the compound described in the first aspect of the present invention, or its stereoisomers or tautomers, or its pharmaceutically acceptable Salt, hydrate or solvate; and (2) a pharmaceutically acceptable carrier.
  • the third aspect of the present invention provides a compound as described in the first aspect of the present invention, or its stereoisomer or tautomer, or its pharmaceutically acceptable salt, hydrate or solvate or
  • the use of the pharmaceutical composition according to the second aspect of the present invention is characterized in that it is used to prepare drugs for preventing and/or treating abnormal activity of BTK and diseases related to abnormal activity of BTK mutant (such as C481S).
  • the disease or condition is selected from bladder cancer, brain tumor, breast cancer, uterine cancer, colorectal cancer, esophageal cancer, liver cancer, follicular lymphoma, melanoma, hematological malignancies, Myeloma, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, and B-cell-derived lymphoid malignancies, B-cell proliferative disorders: diffuse B-cell lymphoma, follicular lymphoma, chronic lymphocytes Lymphoma, chronic lymphocytic leukemia, B-cell young lymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom's macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, Extranodal marginal zone B-cell lymphoma, intranodal marginal zone B-cell lymphoma, mantle cell lymphoma, mediast
  • the disease or condition is selected from autoimmune diseases and inflammatory diseases including rheumatoid arthritis, psoriatic arthritis, osteoarthritis and juvenile arthritis; and the hepatitis includes autoimmunity Hepatitis; the cystitis includes interstitial cystitis; the allergic reaction includes allergies, type I hypersensitivity, allergic rhinitis; the bronchitis includes bronchiolitis; the enteritis includes colitis and proctitis
  • the dermatitis includes atopic dermatitis, scleroderma, and psoriasis; the myelitis includes acute disseminated encephalomyelitis; the gastritis includes gastroenteritis; the nephritis includes pyelonephritis; the rhinitis includes sinuses inflammation.
  • the inventors unexpectedly discovered a compound represented by formula A.
  • the compound has unexpected inhibitory activity against BTK, especially the BTK C481S mutant, and can be used to treat cancers, allergic diseases, autoimmune diseases, inflammatory diseases, etc. mediated by BTK, and for B- Cell lymphoma and leukemia, especially for cancer patients who are resistant to existing drugs, have excellent therapeutic effects.
  • BTK BTK C481S mutant
  • alkyl by itself or as part of another substituent refers to a linear (ie unbranched) or branched chain, or cyclic hydrocarbon group, or a combination thereof, which may be saturated, mono- or Polyunsaturated, which may include divalent or multivalent groups, have a specified number of carbon atoms (ie, C 1 -C 10 refers to one to ten carbon atoms).
  • saturated hydrocarbon groups include, but are not limited to, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, cyclohexylmethyl Groups such as n-pentyl, n-hexyl, n-heptyl, n-octyl, etc., and equivalents and isomers.
  • An unsaturated alkyl group is an alkyl group having one or more double bonds or triple bonds.
  • unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3- (1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and higher homologs and isomers.
  • An alkyl group defined as a hydrocarbyl group is referred to as "homoalkyl”.
  • the alkyl group is optionally substituted with one or more halogen atoms.
  • fluoroalkyl refers to an alkyl group as defined above, in which one or more hydrogen atoms are replaced by fluorine atoms.
  • the alkyl group (or alkylene group) usually has 1 to 24 carbon atoms, and a group having 10 or less carbon atoms is preferred in the present invention.
  • “Lower alkyl” or “lower alkylene” refers to a shorter chain alkyl or alkylene group, usually having eight or fewer carbon atoms.
  • the alkylene group is optionally substituted with one or more halogen atoms.
  • alkynyl refers to a carbon chain containing at least one carbon-carbon triple bond, which may be linear or branched, or a combination thereof.
  • alkynyl groups include ethynyl, propargyl, 3-methyl-1-pentynyl, 2-heptynyl, and the like.
  • the alkynyl group is optionally substituted with one or more halogen atoms.
  • cycloalkyl refers to a monocyclic or bicyclic saturated carbocyclic ring, each having 3 to 10 carbon atoms.
  • the "fused analog" of cycloalkyl refers to a single ring fused with an aryl or heteroaryl group, where the point of attachment is in the non-aromatic part. Examples of cycloalkyl and fused analogs thereof include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydronaphthyl, decahydronaphthyl, indenyl and the like.
  • the cycloalkyl group is optionally substituted with one or more halogen atoms.
  • alkoxy refers to a straight or branched chain alkoxy group having the indicated number of carbon atoms.
  • C 1-6 alkoxy for example, includes methoxy, ethoxy, propoxy, isopropoxy and the like.
  • heteroalkyl by itself or in combination with another term refers to a stable linear or branched chain composed of at least one carbon atom and at least one heteroatom selected from O, N, P, Si, and S , Or cyclic hydrocarbon group, or a combination thereof, wherein the nitrogen atom, phosphorus atom or sulfur atom can be optionally oxidized and the nitrogen atom can be optionally quaternized.
  • the heteroatoms O, N, P, S, and Si can be placed at any position within the heteroalkyl group or at the position where the alkyl group is connected to the rest of the molecule.
  • heteroalkylene by itself or in combination with other terms refers to a divalent group derived from a heteroalkyl group, such as, but not limited to, -CH 2 -CH 2 -S-CH 2 -CH 2- And -CH 2 -S-CH 2 -CH 2 -NH-CH 2 -.
  • the heteroatoms can be at either end or both ends of the chain (for example, alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, etc.).
  • the writing direction of the linking group formula does not indicate the orientation of the linking group.
  • the molecular formula -C(O)OR'- means -C(O)OR'- and -R'OC(O)-.
  • the heteroalkyl group used herein includes those groups that are connected to the rest of the molecule through a heteroatom, such as -C(O)R', -C(O)NR', -NR'R", -OR' , -SR' and/or -SO 2 R'.
  • heteroalkyl is mentioned, and specific heteroalkyls such as -NR'R" are mentioned later, it should be understood that the terms heteroalkyl and -NR'R" is not repeated and not mutually exclusive. On the contrary, these specific heteroalkyls are cited for clarity. Therefore, the term “heteroalkyl” should not be interpreted herein to exclude specific heteroalkyls such as -NR'R".
  • cycloalkoxy refers to a cycloalkyl group as defined above bound to an oxygen atom, such as cyclopropoxy.
  • fluoroalkoxy refers to an alkoxy group as defined above in which one or more hydrogen atoms are replaced by fluorine.
  • aryl refers to a monocyclic or bicyclic aryl group containing only carbon atoms.
  • fused analogue of an aryl group refers to a fusion of an aryl group and a monocyclic cycloalkyl group or a monocyclic heterocyclic group, wherein the point of attachment is at the aryl group.
  • aryl groups and fused ring analogs thereof include phenyl, naphthyl, indanyl, indenyl, tetrahydronaphthyl, 2,3-dihydrobenzofuranyl, dihydrobenzopyranyl, 1, 4-Benzodioxanyl and the like.
  • heteroaryl refers to a monocyclic or bicyclic aryl group containing at least one heteroatom selected from N, O, and S.
  • fused analog of a heteroaryl group refers to the fusion of a heteroaryl group and a monocyclic cycloalkyl group or a monocyclic heterocyclic group, wherein the point of attachment is at the aryl part.
  • heteroaryl groups include pyrrolyl, isazolyl, isothiazolyl, pyrazolyl, pyridyl, oxazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, imidazolyl, triazolyl, tetrazole Group, furanyl, triazinyl, thienyl, pyrimidinyl, pyridazinyl, pyrazinyl, benzoxazolyl, benzothiazolyl, benzimidazolyl, benzofuranyl, benzothienyl, furan And (2,3-b) pyridyl, quinolyl, indolyl, isoquinolyl, etc.
  • the defined alkyl, aryl and heteroaryl groups are unsubstituted or substituted with at least one substituent selected from the group consisting of substituents.
  • the substituents are selected from the group consisting of halogen atoms, alkyl groups having 1 to 6 carbon atoms, alkoxy groups having 1 to 6 carbon atoms, haloalkyl groups having 1 to 6 carbon atoms, and having 1 to 6 carbon atoms.
  • heterocycle or “heterocyclic” or “heterocycloalkyl” or “heterocyclyl” refers to a saturated, partially saturated or unsaturated group (but not aromatic), It has a single ring or fused ring (including bridged ring systems and spiro ring systems, with 1 to 10 carbon atoms and 1 to 4 heteroatoms selected from nitrogen, sulfur or oxygen in the ring.
  • fused ring system one or more Each ring can be cycloalkyl, aryl or heteroaryl, as long as the point of attachment is through a non-aromatic ring.
  • the nitrogen and/or sulfur atoms of the heterocyclic group are optionally oxidized to provide N-oxide, sulfinyl and sulfonyl moieties.
  • heterocyclic groups and their fused analogs include pyrrolidinyl, piperidinyl, piperazinyl, imidazolidinyl, 2,3-dihydrofuran (2,3-b) Pyridyl, benzoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolinyl, etc.
  • the term also includes non-aromatic partially unsaturated mono Rings, such as 2- or 4-pyridones or N-substituted-(1H,3H)-pyrimidine-2,4-diones (N-substituted uracils) connected by a nitrogen atom.
  • non-aromatic partially unsaturated mono Rings such as 2- or 4-pyridones or N-substituted-(1H,3H)-pyrimidine-2,4-diones (N-substituted uracils) connected by a nitrogen atom.
  • substituted heterocyclic or “substituted heterocycloalkyl” or “substituted heterocyclyl” refers to a heterocyclic group substituted with 1 to 5 (such as 1 to 3) substituents The substituents are the same as those defined by substituted cycloalkyl.
  • halo or halogen by itself or as part of another substituent refers to a fluorine, chlorine, bromine or iodine atom.
  • haloalkyl is meant to include monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl means including, but not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl Base etc.
  • Prodrug refers to the substance that is transformed into the parent drug in the body. In some cases, prodrugs are often used because they are easier to administer than the parent drug. For example, prodrugs can be bioavailable when taken orally but the parent drug cannot. In the pharmaceutical composition, the prodrug may also have a higher solubility than the parent drug.
  • An example of a prodrug, but not limited to it, can be any compound of formula I being administered in the form of an ester (prodrug) to promote transport across the cell membrane. Water solubility in the cell membrane is harmful to migration, and once it is water-soluble In the cell, the ester is then metabolized and hydrolyzed to the active substance carboxylic acid.
  • Another example of a prodrug may be a short peptide (polyamino acid) to which an acid group is bonded, wherein the peptide is metabolized to release the active part.
  • the compounds of the present invention contain one or more asymmetric centers, they can be used as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and single diastereomers.
  • a compound of the present invention or a compound of formula (A) it should be understood to include all these isomeric forms.
  • Some compounds of formula (A) may contain one or more than one ring system, so cis- and trans-isomers may exist.
  • the present invention is intended to encompass all these cis- and trans-isomers.
  • ketones known as keto-enol tautomers and their enol forms. Both individual tautomers and mixtures thereof are included in the compound of formula (A).
  • the compound of formula (A) can be separated into diastereoisomeric pairs of enantiomers, for example, by fractional crystallization from a suitable solvent, such as methanol or ethyl acetate or a mixture thereof.
  • a pair of enantiomers thus obtained can be separated into individual stereoisomers by conventional methods, for example, using an optically active amine or acid as a resolving reagent or in a chiral HPLC column.
  • any enantiomer of the compound of general formula (A) can be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • the compound of formula (A) may also include a series of stable isotope labeled analogs.
  • one or more protons in the compound of formula (A) can be replaced with a deuterium atom, thereby providing a deuterated analog with improved pharmacological activity.
  • the term "pharmaceutically acceptable salt” refers to a non-toxic acid or alkaline earth metal salt of the compound of formula I. These salts can be prepared in situ during the final separation and purification of the compound of the general formula I, or prepared by reacting a suitable organic or inorganic acid or base with a basic or acidic functional group, respectively.
  • Representative salts include, but are not limited to: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate , Camphorate, camphorsulfonate, digluconate, cyclopentane propionate, lauryl sulfate, ethanesulfonate, glucose heptanoate, glycerophosphate, hemisulfate, heptanoic acid Salt, caproate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate , 2-naphthyl sulfonate, oxalate, pamoate, pectinate, thiocyanate, 3-phenylpropionate, picrate, pivalate, propionate, Succinate, sulf
  • nitrogen-containing basic groups can be quaternized with the following reagents: alkyl halides, such as methyl, ethyl, propyl, butyl chloride, bromide and iodide; dialkyl sulfate , Such as dimethyl, diethyl, dibutyl and dipentyl sulfate; long chain halides such as decyl, lauryl, myristyl and stearyl chloride, bromide and iodide; aralkyl Base halides such as benzyl and phenethyl bromide. This results in a water-soluble or oil-soluble or dispersible product.
  • alkyl halides such as methyl, ethyl, propyl, butyl chloride, bromide and iodide
  • dialkyl sulfate Such as dimethyl, diethyl, dibutyl and dipentyl sulfate
  • long chain halides
  • the base addition salt can be prepared in situ during the final separation and purification of the compound of general formula I, or the carboxylic acid moiety can be combined with a suitable base (such as pharmaceutically acceptable metal cation hydroxide, carbonate or carbonic acid). Hydrogen salt) or ammonia, or organic primary, secondary or tertiary amines.
  • Pharmaceutically acceptable salts include, but are not limited to, alkali metal and alkaline earth metal-based cations, such as sodium, lithium, potassium, calcium, magnesium, aluminum salts, etc., and non-toxic ammonium, quaternary ammonium and amine cations, including , But not limited to: ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, etc.
  • Other representative organic amines used to form base addition salts include diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like.
  • references to compounds of formula (A) also include pharmaceutically acceptable salts.
  • the preparation for oral administration can also be a hard gelatin capsule in which the active ingredient is mixed with an inert solid diluent, such as calcium carbonate, calcium phosphate or kaolin, or a soft gelatin capsule, in which the active ingredient is mixed with a water or oil medium, such as peanut oil , Liquid paraffin or olive oil.
  • an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin
  • a soft gelatin capsule in which the active ingredient is mixed with a water or oil medium, such as peanut oil , Liquid paraffin or olive oil.
  • Aqueous suspensions contain the active substance mixed with excipients suitable for preparing aqueous suspensions.
  • excipients are suspending agents, such as sodium carboxymethyl cellulose, methyl cellulose, hydroxypropyl methyl cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum arabic; dispersing agent or wetting agent
  • the agent can be a naturally occurring phospholipid, such as lecithin, or a condensation product of alkylene oxide and fatty acid, such as polyoxyethylene stearate, or ethylene oxide and long-chain aliphatic alcohol, such as seventeen
  • the condensation products of heptadecaethylene-oxycetanol, or the condensation products of ethylene oxide and partial esters derived from fatty acids and hexitols for example, polyoxyethylene sorbitol monooleate, or cyclic
  • Aqueous suspensions may also contain one or more preservatives, such as ethyl p-hydroxybenzoate or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one Or a variety of sweeteners, such as sucrose, saccharin or aspartame.
  • preservatives such as ethyl p-hydroxybenzoate or n-propyl p-hydroxybenzoate
  • coloring agents such as ethyl p-hydroxybenzoate or n-propyl p-hydroxybenzoate
  • flavoring agents such as a variety of sweeteners, such as sucrose, saccharin or aspartame.
  • sweeteners such as sucrose, saccharin or aspartame.
  • Oily suspensions can be formulated by suspending active ingredients in vegetable oils, such as peanut oil, olive oil, sesame oil or coconut oil, or in mineral oils, such as liquid paraffin. Oily suspensions may contain thickeners such as beeswax, hard paraffin or cetyl alcohol. The above-mentioned sweeteners can be added, and flavoring agents can be added to provide a palatable oral preparation. These compositions can be preserved by adding antioxidants such as ascorbic acid.
  • Dispersible powders and granules suitable for the preparation of aqueous suspensions by the addition of water provide active ingredients mixed with dispersing or wetting agents, suspending agents and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are shown by the examples already mentioned above. Additional excipients may also be present, such as sweetening agents, flavoring agents, and coloring agents.
  • the pharmaceutical composition of the present invention may also be in the form of an oil-in-water emulsion.
  • the oil phase can be a vegetable oil, such as olive oil or peanut oil, or a mineral oil, such as liquid paraffin or a mixture of these.
  • Suitable emulsifiers can be naturally occurring phospholipids, such as soybeans, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitol monooleate, and the condensation of the above partial esters with ethylene oxide Products such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs can be formulated with sweetening agents, such as glycerin, propylene glycol, sorbitol or sucrose. Such preparations may also contain a demulcent, preservative, flavoring and coloring agent.
  • the pharmaceutical composition may be in the form of a sterile injectable aqueous or oily suspension. This suspension can be formulated by known techniques using those suitable dispersing or wetting agents and suspending agents mentioned above.
  • the sterile injection preparation may also be a sterile injection solution or suspension prepared with a non-toxic parenterally acceptable diluent or solvent, for example, a solution prepared with 1,3-butanediol.
  • Acceptable vehicles and solvents that can be used are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, non-volatile oils are often used as solvents or suspension media. Any mild non-volatile oil used for this purpose can be used, including synthetic mono or diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injections.
  • the compounds of the invention can also be administered intranasally or by inhalation, usually in the form of dry powder from a dry powder inhaler (alone, as a mixture, for example, a dry blend containing lactose, or as a mixed component granule, for example, with phospholipids Such as phosphatidylcholine mixed), or aerosol sprayer from a pressurized container, pump, ejector, atomizer (I atomizer that uses an electric current to generate a fine mist) or a nebulizer, with or without suitable propulsion Agents such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane.
  • the powder may include a bioadhesive such as chitosan or cyclodextrin.
  • a pressurized container, pump, ejector, atomizer or sprayer contains a solution or suspension of the compound of the present invention, the solution or suspension contains, for example, ethanol, aqueous ethanol, or is used for dispersion, solubilization, or prolonged release activity Suitable alternative reagents, as solvent propellants and optional surfactants, such as sorbitan trioleate, oleic acid or oligolactic acid.
  • the drug product Before use in the form of a dry powder or suspension formulation, the drug product is micronized to a size suitable for inhalation delivery (usually less than 5 microns).
  • pulverization method such as spiral jet milling, fluidized bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
  • Capsules for inhalers or insufflators can be formulated into powders containing the compound of the present invention, suitable powders such as lactose or starch, and Performance modifiers such as L-leucine, mannitol or magnesium stearate. Lactose may be in anhydrous or monohydrate form, with the latter being preferred. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
  • a suitable solution formulation used in a nebulizer that generates a fine mist using an electrodynamic method may contain the compound of the present invention from 10 g to 20 mg per activation, and the activation volume can vary from 11 to 1001.
  • a typical formulation may contain a compound of formula (A), propylene glycol, sterile water, ethanol and sodium chloride.
  • Alternative solvents that can be used in place of propylene glycol include glycerin and polyethylene glycol.
  • Suitable flavoring agents such as menthol and levomenthol, or sweetening agents, such as saccharin or sodium saccharin, can be added to those formulations of the present invention for inhalation/intranasal administration.
  • Formulations for inhalation/intranasal administration can be formulated for immediate use and/or modified release, for example, using poly(DL-lactic acid-glycolic acid (PGLA).
  • Modified release formulations include delayed release, sustained release, and pulsed release , Controlled release, targeted release and programmed release.
  • the dosage unit is determined by a valve that provides a metered amount.
  • the unit of the present invention is usually arranged to administer a metered dose or "spray" containing 0.001 to 10 mg of the compound of formula (A).
  • the total daily dose is usually from 0.001 to 10 mg, and can be administered in a single dose or more generally as divided doses throughout the day.
  • the compound of formula (A) can also be administered rectally as a suppository.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at normal temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at normal temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at normal temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the drug.
  • Such substances are cocoa butter and polyethylene glycol.
  • creams, ointments, gels, solutions or suspensions, etc. containing the compound of formula (A) are applied.
  • topical applications should include mouthwashes and gargles).
  • a dosage level of about 0.01 mg to about 140 mg/kg body weight per day is useful in the treatment of the above conditions, or about 0.5 mg to about 7 g per patient per day.
  • inflammation can be effectively treated by administering about 0.01 to 50 mg of the compound per kilogram of body weight per day, or about 0.5 mg to 3.5 g per patient per day, preferably 2.5 mg to 1 g per person per day.
  • the amount of active ingredient that can be combined with the carrier material to produce a single dosage form will vary according to the host being treated and the specific mode of administration.
  • a preparation for oral administration to humans may contain 0.5 mg to 5 g of active agent, which is compounded with a suitable and convenient amount of carrier material.
  • the carrier material may vary from about 5% to about 95% of the total composition.
  • Dosage unit forms generally contain about 1 mg to about 500 mg of active ingredient, usually 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg or 1000 mg.
  • the specific dosage level for any particular patient depends on many factors, including age, weight, general health, gender, diet, time of administration, route of administration, excretion rate, drug combination, and the person being treated The severity of the specific disease.
  • the compounds of the present invention can be used to treat diseases related to the abnormal activity of BTK and BTK mutants (such as C481S).
  • the present invention also includes a method of treating a patient's disease using a method of administering a therapeutically effective amount of a compound of formula (A) to a patient.
  • the compounds of the present invention are useful for the treatment of diseases with abnormal cell growth and/or apoptosis disorders, such as mesothioloma, bladder cancer, pancreatic cancer, skin cancer, head or neck cancer, skin Or intraocular melanoma, ovarian cancer, breast cancer, uterine cancer, endometrial fallopian tube cancer, cervical cancer, vagina cancer, vulvar cancer, bone cancer, cervical cancer, colon cancer, rectal cancer, anal area cancer, stomach cancer, stomach cancer Intestinal cancer (stomach, colorectal and duodenum), chronic lymphocytic leukemia, esophageal cancer, small intestine cancer, endocrine system cancer, thyroid cancer, cancer of parathyroid cancer, adrenal cancer, soft tissue cancer, urethral cancer , Penile cancer, testicular cancer, hepatocellular carcinoma (liver cancer and bile duct), primary or secondary central nervous system tumors, primary or secondary brain tumors, Hodgkin's sarcoma, chronic
  • mesothelioma (mesothioloma), bladder cancer, pancreatic cancer, skin cancer, head or neck cancer, skin or intraocular melanoma, breast cancer, uterine cancer, fallopian tube cancer, endometrial cancer.
  • cervical cancer vaginal cancer, vulvar cancer, bone cancer, ovarian cancer, cervical cancer, colon cancer, rectal cancer, anal cancer, gastric cancer, gastrointestinal cancer (stomach, colorectal and duodenum), chronic Lymphocytic leukemia, esophageal cancer, small bowel cancer, endocrine system cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, testicular cancer, hepatocellular carcinoma (liver cancer and bile duct), primary Primary or secondary central nervous system tumors, primary or secondary brain tumors, Hodgkin’s disease, chronic or acute leukemia, chronic myeloid leukemia, lymphocytic lymphoma, lymphoblastic leukemia, follicles Lymphoma, T-cell or B-cell-derived lymphoid malignancies, melanoma, multiple myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate
  • the compound of formula (A) can also be used to treat autoimmune diseases and inflammatory diseases, for example, inflammatory bowel disease, arthritis, lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis and juvenile arthritis, Still’s disease, diabetes, myasthenia gravis, Hashimoto’s thyroiditis, Odder’s thyroiditis, Graves’ disease, Sjogren’s syndrome, multiple sclerosis, GuiUain-Barre syndrome, acute Disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylitis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, Abdominal disease, Goodpasture's syndrome (Goodpasture's syndrome), idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome (Reiter'
  • the subject is administered an anticancer agent.
  • the anticancer agent is a mitogen-activated protein kinase signaling pathway inhibitor, such as U0126, PD98059, PD184352, PD0325901, ARRY-142886, SB239063, SP600125, BAY 43-9006, wortmannin, or LY294002.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound of formula (A) itself or at least one compound of formula (A) in combination with one or more pharmaceutically acceptable excipients.
  • compositions of the present invention those suitable for oral, parenteral, nasal, transdermal or transdermal, rectal, translingual, ocular or respiratory administration, especially tablets or Sugar lozenges, sublingual tablets, sachets, packets, gelatin capsules, sublingual agents (glossettes), lozenges, suppositories, creams, ointments, skin gels, drinkable or can Ampoule for injection.
  • the dosage can vary according to the patient's sex, age and weight, route of administration, nature of indication, or any related treatment. In one or more administrations, the dosage ranges from 0.01 mg to 1 gram per 24 hours.
  • the present invention also relates to a combination of formula (A) and one or more anticancer agents selected from the group consisting of cytotoxic agents, mitotic toxins, antimetabolites, proteasome inhibitors and kinase inhibitors, and It relates to the use of this type of combination in the preparation of drugs for treating cancer.
  • the compounds of the present invention can also be used in combination with radiotherapy for the treatment of cancer.
  • the compound of formula (A) is also expected to be used as a chemotherapeutic agent combined with a therapeutic agent.
  • the therapeutic agent includes, but is not limited to, angiogenesis inhibitors, antiproliferative agents, other kinase inhibitors, and other receptor tyrosine kinase inhibitors , Aurora kinase inhibitors, Polo-like kinase inhibitors, bcr-abl kinase inhibitors, growth factor inhibitors, COX-2 inhibitors, non-steroidal anti-inflammatory drugs (NSAIDS), antimitotic agents, alkylating agents , Antimetabolites, embedded antibiotics, platinum-containing reagents, growth factor inhibitors, ionizing radiation, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, immunomodulators, immune preparations, antibodies, hormone therapy , Retinoid/deltoid plant alkaloids, proteasome inhibitors, HSP-90 inhibitors, histone deacetylase inhibitors (HDAC) inhibitor
  • Angiogenesis inhibitors include, but are not limited to, EGFR inhibitors, PDGFR inhibitors, VEGFR inhibitors, TTE2 inhibitors, IGF1R inhibitors, matrix metalloproteinase-2 (MMP-2) inhibitors, matrix metalloproteinase-9 (MMP -9) Inhibitors, thrombospondin analogs, such as thrombospondin-1 and N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-IIe-Arg-Pro-NHCH 2 CH 3 or salts thereof And N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-Ile-Arg-PrO-NHCH 2 CH 3 analogues, such as N-Ac-GlyVal-D-AIle-Ser-GIn-Ile-Arg -ProNHCH 2 CH 3 or its salt.
  • EGFR inhibitors include, but are not limited to, Iressa (gefitinib), Tarceva (erlotinib or OSI-774), icotinib, cetuximab (cetuximab) Anti), EMD-7200, ABX-EGF, HR3, IgA antibody, TP-38 (IVAX), EGFR fusion protein, EGF-vaccine, AZD9291, CO1686, anti-EGFr immunoliposome and Tykerb (lapatinib).
  • PDGFR inhibitors include, but are not limited to, CP-673, 451 and CP-868596.
  • VEGFR inhibitors include, but are not limited to, Avastin (bevacizumab), sarcomat (Sunitinib, SU11248), NEXAVAR (Sorafenib, BAY43-9006), CP-547, 632, axitinib (AG13736), apatinib, cabotinib, Iressa (vandetanib, ZD-6474), AEE788, AZD-2171, VEGFtrap, Vattaranib (PTK- 787, ZK-222584), pegastani, M862, pazopanib (GW786034), BC-00016, ABT-869 and angiozyme.
  • thrombospondin analogs include, but are not limited to, ABT-510.
  • BCL inhibitors include, but are not limited to, obatoclax and navitoclax, ABT199.
  • Aurora kinase inhibitors include, but are not limited to, VX-680, AZD-1152, and MLN-8054.
  • polo-like kinase inhibitors include, but are not limited to, BI-2536.
  • BCR-ABL kinase inhibitors include, but are not limited to, Gleevec (imatinib), ponatinib, nilotinib, and dasatinib (BMS354825).
  • platinum-containing reagents include, but are not limited to, cisplatin, Burr (carboplatin), eplatin, lobaplatin, nedaplatin, lerxidine (oxaliplatin) or satraplatin.
  • mTOR inhibitors include, but are not limited to, CCI-779, rapamycin, temsirolimus, everolimus, RAD001, INK-128, and ridaforolimus.
  • HSP-90 inhibitors include, but are not limited to, geldanamycin, radicicol, 17-AAG, KOS-953, 17-DMAG, CNF-101, CNF-1010, 17-AAG-nab , NCS-683664, Mycograb, CNF-2024, PU3, PU24FC1, VER49009, IPI-504, SNX-2112 and STA-9090.
  • HDAC histone deacetylase inhibitors
  • MEK inhibitors include, but are not limited to, PD325901, ARRY-142886, ARRY-438162, and PD98059.
  • CDK inhibitors include, but are not limited to, flavopyridol, MCS-5A, CVT-2584, seliciclib (CYC-202, R-roscovitine), ZK -304709, PHA-690509, BMI-1040, GPC-286199, BMS-387,032, PD0332991 and AZD-5438.
  • COX-2 inhibitors include, but are not limited to, CELEBREX TM (celecoxib), parecoxib, delacoxib, ABT-963, MK-663 (etoricoxib), COX-189 (Ro Mecoxib), BMS347070, RS57067, NS-398, Valdecoxib, Parecoxib, Rofecoxib, SD-8381, 4-Methyl-2-(3,4-Dimethylphenyl)-1- (4-Sulfamoyl-phenyl-1H-pyrrole, T-614, JTE-522, S-2474, SVT-2016, CT-3, SC-58125 and Arcoxia (etoricoxib).
  • non-steroidal anti-inflammatory drugs include, but are not limited to, salicylate (Amigesic), diflunisal (Dolobid), ibuprofen (ibuprofen), ketoprofen (Orudis), Nabumetone (Relafen), Piroxicam (Feldene), Naproxen (Aleve, Naproxen), Diclofenac (Voltarin), Indomethacin (Indomethacin), Sulindac (Clinoril), Tolmetin ( Tolectin), etodolac (iodine value), ketorolac (Toradol) and oxaprozin (Daypro).
  • salicylate Amigesic
  • diflunisal Dolobid
  • ibuprofen ibuprofen
  • ketoprofen Orudis
  • Nabumetone Relafen
  • Piroxicam Piroxicam
  • Naproxen Aleve, Naproxen
  • Diclofenac Voltarin
  • Indomethacin
  • ERBB2 receptor inhibitors include, but are not limited to, CP-724-714, CI-1033, (canetinib), Herceptin (trastuzumab), Omitarg (2C4, petuzumab) -petuzumab), TAK-165, GW-572016 (Ionafarnib) GW-282974, EKB-569, PI-166, dHER2 (HER2 vaccine), APC8024 (HER2 vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3 , AS HER2's trifunctional bispecific antibody, monoclonal antibody AR-209 and monoclonal antibody 2B-1.
  • alkylating agents include, but are not limited to, nitrogen mustard N-oxides, cyclophosphamide, ifosfamide, chloroetan, chlorambucil, melphalan, busulfan, dibromide Mannitol, Carboquinone, Cetepa, Ramustine, Nimustine, Temozolomide, AMD-473, Hexamethylmelamine, AP-5280, Apaziquone, Brostallicin, Bendamustine, carmustine, estramustine, formustine, glufosfamide, KW-2170, horsefosfamide, dulcitol dibromide and, carmustine (BCNU), lomo Stine (CCNU), busulfan, trioxifan, dacarbazine and temozolomide.
  • nitrogen mustard N-oxides cyclophosphamide
  • ifosfamide chloroetan
  • chlorambucil chlorambucil
  • melphalan
  • antimetabolites include, but are not limited to, methotrexate, 6-mercaptopurine nucleoside, mercaptopurine, uracil analogs, such as 5-fluorouracil (5-FU) alone or in combination with folinic acid, tegafur , UFT, Deoxyfluridine, Carmofur, Cytarabine, Cytarabine, Enoxabine, SI, Libitai (pemetrexed disodium, LY231514, MTA), Gemcitabine (gemcitabine), Fludarabine, 5-azacytidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethnylcytidine, cytarabine, hydroxyurea, TS-I, Melphalan, Nelarabine, Loratrox, ocfosate, disodium premetrexed, pentostatin, pelitrexol, raltitrexed, tri
  • antibiotics include embedded antibiotics, but are not limited to axorubicin, actinomycin (such as actinomycin D), amrubicin, annamycin, adriamycin, bleomycin a, bleomycin b, daunorubicin, doxorubicin, ixarucin, epirbucin, glarbuicin, idarubicin, mitomycin C, naphthalene Morubicin, neocarcinogen, pelomycin, pirarubicin, phalaenopsis, butylate, streptozocin, valrubicin, netstatin and combinations thereof.
  • actinomycin such as actinomycin D
  • amrubicin annamycin
  • adriamycin bleomycin a
  • bleomycin b daunorubicin
  • doxorubicin doxorubicin
  • ixarucin epirbucin
  • topoisomerase inhibitors include, but are not limited to, one or more agents selected from the following group: arubicin, amonafide, belotecan, camptothecin, 10-Hydroxycamptothecin, 9-Aminocamptothecin, flutecan (diflomotecan), irinotecan hydrochloride (Camptosar), edotecarin, epirubicin (Ellence), etoposide, exenotecan ( exatecan), gematecan, letotecan, orathecin (Supergen), BN-80915, mitoxantrone, pirabucin (pirarbucin), pixantrone (pixantrone), rubotecan, sobuzoxane, SN-38, tafluposide and topotecan.
  • agents selected from the following group arubicin, amonafide, belotecan, camptothecin, 10-Hydroxycamptothec
  • antibodies include, but are not limited to, rituximab, cetuximab, bevacizumab, Trastuzimab, specific CD40 antibody, and specific IGF1R antibody.
  • hormone therapy examples include, but are not limited to, Exemestane (Arnosine), Leuprolide Acetate, Anastrozole (Rainind), Fosrelin (Nored), Goserelin ( goserelin), docalciferol, fadrozole, formestane, tamoxifen citrate (tamoxifen), constellation, abarex, TRELSTAR, finasteride, fulvestrant, trop Remifene, raloxifene, lasoxifene, letrozole, flutamide, bicalutamide, megesterol, mifepristone, nilutamide, dexamethasone, strong Corticosteroids
  • retinoids/deltoids examples include, but are not limited to, seocalcitol (EB1089, CB1093), lexacalcitrol (KH 1060), fenretinide, allivide Aliretinoin, Bexarotene and LGD-1550.
  • plant alkaloids examples include, but are not limited to, vincristine, vinblastine, vindesine, and vinorelbine.
  • proteasome inhibitors include, but are not limited to, bortezomib (Valcade), MGL32, NPI-0052 and PR-171.
  • immune agents include, but are not limited to, interferon and many other immune enhancers.
  • Interferons include interferon alpha, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma-1a, interferon gamma-1b (Actimmune) or interferon gamma-nl and combinations thereof.
  • accelerators include filgrastin, lentinan, sizofilan, TheraCys, ubenimex, WF-10, aldesleukin, alemtuzumab , BAM-002, decarbazine, daclizumab, denileukin, gemtuzumab ozogamicin, ibritumomab, imiquimod, Lenograstim, lentinan, melanoma vaccine (Corixa), molgramostim, OncoVAC-CL, sargaramostim, tasonamine (tasonermin), tecleukin, thymalasin, tositumomab, Virulizin, Z-100, Epratuzumab, mitumomab, Ogovo Monoclonal antibody (oregovomab), petumomab (pemtumomab, Y-muHMFGl), Provenge (Dendreon), STING activator
  • a biological response modifier is an agent that regulates the defense mechanism or biological response of a living organism (such as the survival, growth, or differentiation of tissue cells) to guide it to have anti-tumor activity.
  • Such drugs include Krestin, Lentinan, Sizofrran, Streptomyces and Ubenimex.
  • pyrimidine analogs include, but are not limited to, 5-fluorouracil, fluorouridine, deoxyfluridine, ratitrexed, cytarabine (cytarabine C), cytarabine, Fludarabine and gemcitabine.
  • purine analogs include, but are not limited to, mercaptopurine and thioguanine.
  • immunomodulators include, but are not limited to, thalidomide and lenalidomide.
  • anti-mitotic agents include, but are not limited to, paclitaxel, docetaxel, albumin paclitaxel (ABRAXANE), epothilone D (KOS-862) and ZK-EPO.
  • the compound of the present invention can be prepared by the following reaction formula:
  • the compounds of the present invention can be prepared by chemical synthesis methods, examples of which are shown below. It should be understood that the order of the steps in the process can be changed, those specifically mentioned reagents, solvents and reaction conditions can be replaced, and if necessary, the reactive sites can be protected and deprotected.
  • DBU refers to 1,8-diazabicyclo[5.4.0]undec-7-ene
  • DIBAL refers to diisobutylaluminum hydride
  • DIEA diisopropylethylamine
  • DMAP refers to N,N-di Methylaminopyridine
  • DME refers to 1,2-dimethoxyethane
  • DMF refers to N,N-dimethylformamide
  • DMPE refers to 1,2-bis(dimethylphosphino)ethane
  • DMSO refers to Dimethyl sulfoxide
  • DPPB refers to 1,4-bis(diphenylphosphino)butane
  • dppe refers to 1,2-bis(diphenylphosphino)ethane
  • dppf refers to 1,1'-bis(diphenyl) Phosphino)ferrocene
  • dppm refers to 1,1'-bis(diphenylphos
  • Step 2 Optical activity test: (1S,3R)-3-acetoxycyclohexyl(R)-3,3,3-trifluoro-2-methoxy-2-phenylpropionate
  • Step 3 (1R,3R)-3-(3-(4-phenoxyphenyl)-4-((triphenyl-5-phosphinyl)amino)-1H-pyrazolo[3,4 -d]pyrimidin-1-yl)cyclohexyl acetate and (1R,3R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4- d)pyrimidin-1-yl)cyclohexyl acetate
  • triphenylphosphine (12.44g, 47.41mmol)
  • 150ml of ultra-dry tetrahydrofuran solvent to a fully dried 250ml three-necked flask, and place the flask in an ice bath.
  • diisopropyl azodicarboxylate (9.59 g, 47.41 mmol) was added dropwise to the system. After the dripping is finished, continue to stir for 40 minutes, white precipitate precipitates, continue to stir for 10 minutes.
  • Step 4 (1R,3R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexylacetic acid ester
  • Step 5 (1R,3R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazole[3,4-d]pyrimidin-1-yl)cyclohex-1- alcohol
  • Step 2 (1S,3R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl-1 -alcohol
  • Step 1 (R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexan-1-one
  • Step 2 (1S,3R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-methan Cyclohexane-1-ol and (1R,3R)-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )-1-Methylcyclohexane-1-ol
  • anhydrous cerium trichloride (0.35g, 1.41mmol) and 5ml of ultra-dry tetrahydrofuran were added to a fully dried 25ml flask. After stirring for 2 hours at room temperature, the flask was placed in a dry ice-ethanol bath, and when the temperature of the system dropped to around minus 70 degrees Celsius, a 1M ether solution of methyl lithium (1.41 mL, 1.41 mmol) was slowly added to the system.
  • Example 3A (1S,3R)-3-(4-amino-3-(4- Phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-methylcyclohexane-1-ol 50mg;
  • Example 3B (1R,3R)-3 -(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-methylcyclohexane-1-ol, 50mg , The total yield: 68%.
  • Step 1 3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclopentanone
  • anhydrous cerium trichloride 15mg, 0.62mmol
  • 10ml of ultra-dry tetrahydrofuran were added to a 25ml two-necked reaction flask. After stirring for 1 hour at room temperature, the reaction system was placed in a dry ice-ethanol bath to cool. After cooling to minus 75 degrees Celsius, methyl lithium ether solution (1.33M, 0.48mL, 0.62mmol) was added dropwise to the reaction system.
  • Step 1 4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl]cyclohexanone-1-one
  • Step 2 (1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl]-1-methan Cyclohexane-1-ol and (1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl ]-1-Methylcyclohexane-1-ol
  • anhydrous cerium trichloride (0.29g, 1.18mmol) and 2ml of anhydrous tetrahydrofuran solvent in a fully dried 25ml two-necked flask, and stir at room temperature for 1 hour.
  • reaction solution was placed in a dry ice-ethanol bath to cool, when it reached minus 75 degrees Celsius, 1M methyl lithium (1.18mL, 1.18mmol) in ether was added dropwise to the system, and kept at minus 75 degrees Celsius for 2 hours Then, the intermediate 4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl]cyclohexanone-1- A solution of ketone (100mg, 0.30mmol) in 5 ml of tetrahydrofuran. After the addition, continue to react at this temperature for 15 minutes, remove the dry ice-ethanol bath, and continue the reaction for 1 hour (natural temperature rise). TLC shows that the reaction is complete.
  • Step 4 (1s,4s)-4-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-1- Group) cyclohexan-1-ol and (1r,4r)-4-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclohexan-1-ol
  • Diisopropyl azodicarboxylate (1.26 g, 6.22 mmol) was slowly added dropwise to the reaction solution, and the reaction solution was stirred overnight after the addition was completed. The end of the reaction was monitored by TLC and LCMS. Then add 20 ml of saturated ammonium chloride solution to remove most of the tetrahydrofuran, add 20 ml of water, extract with dichloromethane (30 mL ⁇ 3), combine the organic phases, wash with saturated brine (50 mL ⁇ 1), and dry with anhydrous sodium sulfate.
  • Example 11A (1s, 4s)-4-(4-amino-3-( 4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexan-1-ol 700mg, yield: 53%;
  • Example 11B (1r,4r)-4-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ring Hexan-1-ol 100mg, yield: 8%.
  • Step 1 4-(4-Amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-ylcyclohex-1- ketone
  • Step 2 (1s,4s)-4-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-1- Group)-1-methylcyclohexan-1-ol and (1r,4r)-4-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[ 3,4-d)pyrimidin-1-yl)-1-methylcyclohexan-1-ol
  • anhydrous cerium trichloride (0.24 g, 0.96 mmol) and 3 ml of anhydrous tetrahydrofuran to a fully dried 25 ml two-necked flask, and stir at room temperature for 1 hour. Then the reaction solution was placed in a dry ice-ethanol bath and cooled to about minus 70 degrees Celsius, and 1.6M methyl lithium ether solution (0.60 mL, 0.96 mmol) was added dropwise thereto. The resulting reaction solution was continuously stirred at this temperature for 1 hour.
  • Example 12A (1s, 4s)-4-(4-amino-3-(4-(4-fluorophenoxy) )Phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-methylcyclohexan-1-ol 25mg, yield: 25%;
  • Example 12B (1r,4r )-4-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-methyl Cyclohexan-1-ol 23mg, yield: 2
  • Example 13A ( ⁇ )cis-3-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclopentane-1-ol 1.80g, yield: 46%;
  • Example 13B ( ⁇ )trans-3-(4-amino-3-(4-(4-fluorophenoxy) (Yl)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclopentane-1-ol 200 mg, yield: 5%.
  • Step 1 3-(4-Amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclopentane-1 -ketone
  • Step 2 ( ⁇ )cis-3-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )-1-methylcyclopentan-1-ol and ( ⁇ )trans-3-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo[3, 4-d]pyrimidin-1-yl)-1-methylcyclopentan-1-ol
  • anhydrous cerium trichloride (0.49g, 1.98mmol) and 3ml of anhydrous tetrahydrofuran were added to a fully dried 25ml two-necked flask and stirred at room temperature for 1 hour.
  • a 1.6M methyl lithium ether solution (1.24 mL, 1.98 mmol) is added dropwise to it, and after the addition is complete, continue to stir at this temperature 1 hour.
  • Example 14A ( ⁇ )cis-3-(4-amino-3-(4-(4-fluorophenoxy)phenyl)-1H-pyrazolo [3,4-d]pyrimidin-1-yl)-1-methylcyclopentan-1-ol 80mg, yield: 40%;
  • Example 14B ( ⁇ )trans-3-(4-amino-3 -(4-(4-Fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-methylcyclopentan-1-ol 70mg, yield: 35%.
  • Step 1 (1R,3R)-3-(3-iodo-4-((triphenyl-5-phosphoalkylene)amino)-1H-pyrazolo[3,4-d]pyrimidine-1- Yl]cyclohexyl acetate
  • Step 2 (1R,3R)-3-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl acetate
  • reaction solution was added to 1000 ml of water, extracted with ethyl acetate (1000 mL ⁇ 3), combined the organic phases, filtered through silica gel, washed with petroleum ether, washed with saturated sodium chloride solution (200 mL ⁇ 1), and dried with anhydrous sodium sulfate. It was filtered, concentrated, and purified by silica gel column (petroleum ether) to obtain 18.00 g of product, yield: 60%.
  • Step 5 (1R,3R)-3-(4-amino-3-(4-(3-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-1- Yl)cyclohexyl acetate
  • TLC monitors the end of the reaction, slowly add 100 ml of 0.5N aqueous hydrochloric acid to the system, extract with dichloromethane (30 mL ⁇ 3) and combine the organic phases, wash the organic phase with saturated brine (50 mL ⁇ 1), and dry with anhydrous sodium sulfate , Filtration, concentration, the crude product is directly thrown into the step.
  • Step 3 (1R,3R)-3-(4-amino-3-(2-fluoro-4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexyl acetate
  • Step 4 (1R,3R)-3-(4-amino-3-(2-fluoro-4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexan-1-ol
  • Step 3 (1R,3R)-3-(4-amino-3-(4-(2-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexyl acetate
  • Step 4 (1R,3R)-3-(4-amino-3-(4-(2-fluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexan-1-ol
  • Step 3 (1R,3R)-3-(4-amino-3-(4-(2,6-difluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexyl acetate
  • Step 4 (1R,3R)-3-(4-amino-3-(4-(2,6-difluorophenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexan-1-ol
  • Step 2 2-(4-(2-Fluoro-3-methoxyphenoxy)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborane
  • Step 3 (1R,3R)-3-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclohexyl acetate
  • Step 4 (1R,3R)-3-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclohexan-1-ol
  • Step 1 (1s,4s)-4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexan-1-ol
  • Step 2 (1s,4s)-4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclohexan-1-ol
  • Step 1 (1R,4R)-4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclohexyl 4-nitrobenzoate
  • Step 2 (1R,4R)-4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclohexan-1-ol
  • Step 2 3-(4-Amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclopentane-1-ol
  • reaction liquid was heated to reflux and reacted for 1 hour.
  • LCMS monitored the end of the reaction.
  • Step 2 ( ⁇ )trans-3-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d ]Pyrimidin-1-yl)cyclopentane-1-ol
  • Step 4 1-(3,6-Dihydro-2H-pyran-3-yl)-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine-4 -amine
  • Step 5 5-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)tetrahydro-2H-pyran-3, 4-diol
  • Step 2 3-(3-Iodo-4-((triphenylphosphino)amino)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl acetate
  • Step 3 3-(4-Amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl acetate
  • Step 5 3-(4-Amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexanol
  • the reaction solution was concentrated to remove the organic solvent.
  • the aqueous phase was extracted 3 times with 100 ml of dichloromethane. After the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a residue.
  • Step 6 cis-3-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H pyrazoline[3,4-d]pyrimidine-1 -Base) cyclohexanol
  • Step 2 1-(Cyclohex-2-en-1-yl)-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4 -d]pyrimidin-4-amine
  • Step 3 3-(4-Amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexane-1,2-diol
  • Step 1 1-(Cyclohex-2-en-1-yl)-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4 -d]pyrimidin-4-amine
  • Step 2 4-(4-Amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl )Cyclohexane-1,2-diol
  • the 1-(cyclohex-2-en-1-yl)-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[ 3,4-d]pyrimidin-4-amine (30mg, 0.070mmol), N-methylmorpholine-N-oxide (10mg, 0.084mmol) dissolved in 2ml tert-butanol and 0.5ml water mixed solvent, ice Potassium osmate (0.5 mg, 0.0014 mmol) was added to the reaction system under the bath, and the reaction solution was stirred at room temperature for 16 hours.
  • Step 1 1-Cyclohexyl-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 1 tert-butyl((3,4-dihydro-2H-pyran-2-yl)methoxy)diphenylsilane
  • Step 3 1-(6-(((tert-butyldiphenylsilyl)oxy)methyl)tetrahydro-2H-pyran-3-yl)-3-iodo-1H-pyrazolo[3 ,4-d]pyrimidin-4-amine
  • Step 4 1-(6-(((tert-butyldiphenylsilyl)oxy)methyl)tetrahydro-2H-pyran-3-yl)-3-(4-(2-fluoro-3 -Methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 5 (5-(4-Amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-1- (Yl)tetrahydro-2H-pyran-2-yl)methanol
  • Step 4 2-(4-(2-Fluoro-3-(methoxy-d3)phenoxy)phenyl)-4,4,5,5-tetramethyl-1,3,2-diox Cyclopentane
  • Step 6 (1R,3S)-3-(4-amino-3-(4-(2-fluoro-3-(methoxy-d3)phenoxy)phenyl)-1H-pyrazolo[3 ,4-d)pyrimidin-1-yl)cyclohexanol
  • Step 1 trans-3-(3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-4-((triphenyl-15-phosphino)amino)-1H -Pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl 4-nitrobenzoate
  • Step 3 Trans-3-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazoline[3,4-d]pyrimidine- 1-yl)cyclohexanol
  • Step 4 (1S,3S)-3-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazoline [3,4-d ]Pyrimidin-1-yl)cyclohexanol
  • the BTK/BTK (C481S) inhibitory activity of the compound of formula A was determined by Reaction Biology Corporation (One Great Valley Parkway, Malvern, PA, USA) in Malvern, Pennsylvania, USA.
  • the human full-length BTK/BTK (C481S) enzyme was used, and the substrate was a 20 ⁇ M peptide substrate [KVEKIGEGTYGVVYK].
  • the concentration of ATP used in the determination is 10 ⁇ M, staurosporine is used as a standard, and the IC 50 is 3.94 nM.
  • Table 1 shows the inhibitory activity of the compounds of the Examples on BTK/BTK (C481S) enzymes
  • the concentration of ATP in the labeled embodiment is 30uM;
  • the concentration of ATP in the labeled embodiment is 100uM;
  • the concentration of ATP in the labeled embodiment is 10uM;
  • In the labeled embodiment The concentration of ATP is 50uM; "-" means no test.
  • the CellTiter-Glo method was used to detect the inhibitory activity of the Examples against human diffuse large B lymphoma cells TMD8 cultured in vitro, and the IC 50 values are shown in Table 3 below.
  • Example 1 Compound IC 50 (nM) Example 1 20
  • Example 3A 208 Example 3B 200
  • Example 5 29 Example 6 17
  • Example 7 504 Example 8A 90
  • Example 9B 37 Example 10 145
  • Example 16 41 Example 17 33
  • Example 18 183 Example 19 0.5
  • Example 20 8.6 Example 22 8
  • Example 1 Oral administration at a dose of 30 mg/kg once a day, or oral administration at a dose of 10 ⁇ 5 mg/kg twice a day has a significant inhibitory effect on the growth of TMD8 subcutaneous xenograft tumors, and 22 days after administration, it has an effect on tumor growth
  • the inhibition rates (TGI) were 49.6% and 52.7%.
  • Example 1 Oral administration at a dose of 30 mg/kg once a day has no significant effect on animal body weight. After oral administration at a dose of 10 mg/kg twice a day for 8 consecutive days, the animal’s body weight has decreased, so adjust the dose to 5mg/kg, continued administration for 14 days.
  • Figure 1 shows the solvent control and TMD8 subcutaneous xenograft tumor volume (mean ⁇ standard deviation) of each administration group during the administration period. The results showed that 22 days after the initial administration, the compound of the present invention showed a significant therapeutic effect, and its therapeutic effect was comparable to the positive drug ibrutinib.
  • Figure 2 shows the change of the solvent control and the animal body weight (mean ⁇ standard deviation) of each administration group during the administration period. The results showed that the compound of Example 1 of the present invention exhibited significant anti-tumor effects while not having a significant impact on animal body weight.
  • Example 1 of the present invention has a significant anti-tumor effect, and significantly inhibits the growth of tumors without significantly affecting the body weight of animals.

Abstract

本发明描述了具有蛋白酪氨酸激酶抑制活性的新型分子,以及该化合物的合成和使用方法。具体地,本发明描述了式(A)化合物或其药学上可接受的盐、水合物或溶剂合物,以及该化合物的合成和使用方法。

Description

作为布鲁顿酪氨酸激酶抑制剂的环状分子 技术领域
本发明涉及小分子药物领域,具体地,本发明提供了一种具有蛋白酪氨酸激酶抑制活性的新型分子,以及此类化合物的合成和使用方法。
背景技术
蛋白激酶是人类酶中的最大家族,涵盖超过500种蛋白质。布鲁顿酪氨酸激酶(BTK)是酪氨酸激酶Tec家族的成员,并且是早期B细胞发育和成熟B细胞活化、信号转导、和存活的调节剂。BTK已成为用于治疗B细胞淋巴瘤、白血病和自身免疫疾病的新分子靶标。因此,本领域迫切需要提供更多具有BTK抑制活性的小分子化合物。
发明内容
本发明的目的是提供一种具有BTK抑制活性的小分子化合物。
本发明的第一方面,提供了一种如下式(A)所示的化合物,或其药学上可接受的盐,
Figure PCTCN2020072551-appb-000001
其中:
A环和B环互相并联,且各自独立地为取代或未取代的5-15元杂环或杂芳环;
Cyc 1选自下组:取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 2-C 6烯基、取代或未取代的C 3-C 10环烯基、取代的或未取代的5-15元杂环基、取代的或未取代的5-15元杂芳基、取代或未取代的C 6-C 10芳基;
Cyc 2选自下组:取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 2-C 6烯基、取代或未取代的C 3-C 10环烯基、取代的或未取代的5-15元杂环基、取代的或未取代的5-15元杂芳基、取代或未取代的C 6-C 10芳基;
L 1、L 2各自独立地选自下组:化学键、N、O、S、-S(=O)、-S(=O) 2、C(=O)、-C(O)NH-;
Z为(CR 2R 3) n,n为0、1、2、3、4、5或6;
Y为(CR 4R 5) m,m为0、1、2、3、4、5或6;
U为(CR 6R 7) r,r为0、1、2或3;
且n、m和r不同时为0;
R 2、R 3、R 4、R 5、R 6和R 7各自独立地选自下组:H、NH 2、OH、卤素、取代或未 取代的C 1-C 6烷基;或R 2、R 3、R 4、R 5、R 6、R 7和R 8中任意两个与其相连的碳原子以及间隔的环骨架原子共同形成C 3-C 8碳环,或4-8元的杂环,其中,所述杂环的杂原子选自:S、O、或NR f;R f为H、C 1-C 10烷基、C 3-C 10环烷基、C 6-C 20芳基、或C 3-C 14杂芳基;且R 2、R 3、R 4、R 5、R 6、R 7、R 8和R 9中至少有一个是OH或-[C(R 10)(R 11)] k-OH;
W选自下组:N、O、S或化学键;
X为-C(R 8R 9)-;
R 8选自下组:H、取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 6-C 20芳基、或取代或未取代的C 3-C 14杂芳基、CO 2H、C(O)NR f 2
R 9选自下组:H、OH或-[C(R 10)(R 11)] k-OH;
或R 8和R 9共同构成=O;
且当R 8为H时,R 9为OH或-[C(R 10)(R 11)] k-OH;
R 10和R 11各自独立地选自下组:H、取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 6-C 10芳基、或取代或未取代的C 3-C 14杂芳基;或R 10和R 11与其相连的碳原子共同形成C 3-C 8碳环,或4-8元的杂环,其中杂原子可以是硫、氧、或NR f
k为1、2、3、4、5或6;
除非特别说明,上述的杂芳环、杂芳基、杂环、杂环基各自独立地含有1-4个选自N,O和S的杂原子;
除非特别说明,所述的取代指基团被一个或多个(例如2个、3个、4个等)选自下组的取代基所取代:卤素、C1-C6烷基、卤代的C1-C6烷基、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、氧代、-CN、羟基、羟基-C1-C6烷基、氨基、羧基、C6-C10芳基、卤代的C6-C10芳基、未取代或被选自下组的取代基取代的具有1-3个选自N、S和O的杂原子的5-10元杂芳基:卤素、苯基。
在另一优选例中,L 1-Cyc 1-L 2-Cyc 2为:
Figure PCTCN2020072551-appb-000002
其中,R 12,R 13,R 14和R 15分别独立为H,卤素,取代或未取代的C 1-C 6烷基、取代或未取代的C 1-C 6烷氧基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 3-C 10的杂环烷基,杂原子选择N,O,S;
在另一优选例中,L 1-Cyc 1-L 2-Cyc 2
Figure PCTCN2020072551-appb-000003
V为N或-CR 16-;其中,R 16选自下组:H、卤素取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基。
在另一优选例中,所述的
Figure PCTCN2020072551-appb-000004
具有如下结构:
Figure PCTCN2020072551-appb-000005
其中,虚线为化学键或无;各A 1、A 2、A 3、A 4、A 5、A 6、A 7、A 8和A 9各自独立地为O、S、N、NH、CH或CH 2
波浪线表示连接位点;
且上述基团的各个可取代位点可以包括取代基,其中,取代基的定义如上文中所述。
在另一优选例中,所述的
Figure PCTCN2020072551-appb-000006
具有选自下组的结构:
Figure PCTCN2020072551-appb-000007
在另一优选例中,所述的化合物具有如下式(B)所示的结构:
Figure PCTCN2020072551-appb-000008
其中,
R 12、R 13、R 14和R 15各自独立地选自下组:H、卤素、取代或未取代的C 1-C 6烷基、取代或未取代的C 1-C 6烷氧基、取代或未取代的C 3-C 10环烷基、取代或未取代的5-15元的杂环基。
在另一优选例中,所述的化合物具有如下式(C)所示的结构:
Figure PCTCN2020072551-appb-000009
式(C)中,V为N或-CR 16-;
R 12、R 13、R 14和R 15各自独立地选自下组:H、卤素、取代或未取代的C 1-C 6烷基、取代或未取代的C 1-C 6烷氧基、取代或未取代的C 3-C 10环烷基、取代或未取代的5-15元的杂环基;
R 16为H、卤素取代或非取代的C 1-C 6烷基、取代或非取代的C 3-C 10环烷基。
在另一优选例中,
Figure PCTCN2020072551-appb-000010
为选自下组的基团:
Figure PCTCN2020072551-appb-000011
在另一优选例中,
Figure PCTCN2020072551-appb-000012
Figure PCTCN2020072551-appb-000013
在另一优选例中,
Figure PCTCN2020072551-appb-000014
Figure PCTCN2020072551-appb-000015
在另一优选例中,所述的
Figure PCTCN2020072551-appb-000016
为取代的环己基或环氧戊烷基。
在另一优选例中,所述的化合物具有如下式(D)、(E)或(F)所示的结构:
Figure PCTCN2020072551-appb-000017
在另一优选例中,所述化合物选自下组:
1).(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己-1-醇(实施例1)
2)(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例2)
3)(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇/(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇(实施例3)
4)3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-(三氟甲基)环己-1-醇(实施例4)
5)3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊醇(实施例5)
6)3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊醇;(实施例6)
7)3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-(三氟甲基)环戊醇(实施例7)
8)(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇/(1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇(实施例8)
9)(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇/(1r,4r)-4(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇(实施例9)
10)4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-环丙基环己烷-1-醇(实施例10)
11)(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇/(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例11)
12)(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇和(1r,4R)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇(实施例12)
13)顺式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇 和反式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(实施例13)
14)(±)顺式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇/(±)反式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇(实施例14)
15)(1R,3R)-3-(4-胺基-3-(4-(3-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇;(实施例15)
16)(1R,3R)-3-(4-胺基-3-(2-氟-4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇;(实施例16)
17)(1R,3R)-3-(4-胺基-3-(4-(3-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇;(实施例17)
18)(1R,3R)-3-(4-氨基-3-(4-(2,6-二氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例18)
19)(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例19)
20)(1s,4s)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例20)
21)(1r,4r)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例21)
22)顺式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(实施例22)
23)反式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(实施例23)
24)5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)四氢-2H-吡喃-3,4-二醇(实施例24)
25)3-(4-氨基-3-(4-(3-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(实施例25)
26)顺式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(实施例26)
27)3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1,2-二醇(实施例27)
28)4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1,2-二醇(实施例28)
30)(5-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)四氢-2H-吡喃-2-基)甲醇(实施例30)
31)(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-(甲氧基-d3)苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇(实施例31)
32)(1S,3S)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑啉[3,4-d]嘧啶-1-基)环己醇(实施例32)
33)3-(4-(4-氨基-1-((1R,3R)-3-羟基环己基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯氧基)-2-氟苯酚(实施例33)
本发明的第二方面,提供了一种药物组合物,其包含(1)本发明第一方面所述的化合物、或其立体异构体或互变异构体、或其药学上可接受的盐、水合物或溶剂化物;和(2)药学上可接受的载体。
本发明的第三方面,提供了一种如本发明第一方面所述的化合物、或其立体异构体或互变异构体、或其药学上可接受的盐、水合物或溶剂化物或如本发明第二方面所述的药物组合物的用途,其特征在于,用于制备预防和/或治疗BTK的异常活性以及BTK突变体(如C481S)异常活性相关的疾病的药物。
在另一优选例中,所述的疾病或病症选自膀胱癌,脑肿瘤,乳腺癌,子宫癌,结肠直肠癌,食道癌,肝脏癌症,滤泡性淋巴瘤,黑色素瘤,恶性血液病,骨髓瘤,卵巢癌,非小细胞肺癌,前列腺癌,小细胞肺癌,和B-细胞来源的淋巴恶性肿瘤,B细胞增殖性病症:弥漫性B细胞淋巴瘤、滤泡性淋巴瘤、慢性淋巴细胞淋巴瘤、慢性淋巴细胞白血病、B细胞幼淋巴细胞白血病、淋巴质浆细胞淋巴瘤/瓦尔登斯特伦氏巨球蛋白血症、脾脏边缘带淋巴瘤、浆细胞性骨髓瘤、浆细胞瘤、结外边缘带B细胞淋巴瘤、结内边缘带B细胞淋巴瘤、外套细胞淋巴瘤、纵隔(胸腺)大B细胞淋巴瘤、血管内大B细胞淋巴瘤、原发性渗出性淋巴瘤、伯基特氏淋巴瘤/白血病或淋巴瘤样肉芽肿病。
在另一优选例中,所述的疾病或病症选自自身免疫疾病和炎性疾病包括类风湿关节炎、银屑病关节炎、骨性关节炎和幼年型关节炎;所述肝炎包括自身免疫性肝炎;所述膀胱炎包括间质性膀胱炎;所述过敏反应包括过敏症、I型超敏反应、过敏性鼻炎;所述支气管炎包括细支气管炎;所述肠炎包括结肠炎、直肠炎;所述皮炎包括特异性皮炎、硬皮病、银屑病;所述脊髓炎包括急性播散性脑脊髓炎;所述胃炎包括胃肠炎;所述肾炎包括肾盂肾炎;所述鼻炎包括鼻窦炎。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
图1.给药期间溶剂对照及各给药组TMD8皮下异种移植瘤体积(平均值±标准误);
图2.给药期间溶剂对照及各给药组动物体重(平均值±标准误)。
具体实施方式
本发明人经过长期而深入的研究,意外地发现了一种如式A所示的化合物。所述的化合物对BTK,特别是BTK C481S突变体抑制具有出乎意料的抑制活性,可用于治疗由BTK介导的癌症,过敏性疾病、自身免疫疾病、炎性疾病等,以及用于B-细胞淋巴瘤和白血病,特别是针对现有药物产生耐药性的癌症病人具有优异的治疗效果。基于上述发现,发明人完成了本发明。
术语
本文所用的缩写具有化学和生物学领域中的常规含义。
除非另外说明,术语“烷基”本身或作为另一取代基的一部分是指直链(即无支链的)或支 链、或环状烃基、或其组合,其可以是饱和的、单或多不饱和的,可包括二价或多价基团,具有指定数量的碳原子(即C 1-C 10是指一至十个碳原子)。饱和烃基的示例包括,但不限于,如甲基、乙基、n-丙基、异丙基、n-丁基、t-丁基、异丁基、仲丁基、环己基、环己基甲基、环丙基甲基等基团,如n-戊基、n-己基、n-庚基、n-辛基等同系物和异构体。不饱和烷基是具有一个或多个双键或三键的烷基。不饱和烷基的示例包括,但不限于,乙烯基、2-丙烯基、巴豆基、2-异戊烯基、2-(丁二烯基)、2,4-戊二烯基、3-(1,4-戊二烯基)、乙炔基、1-和3-丙炔基、3-丁炔基,以及高级同系物和异构体。限定为烃基的烷基称为“同烷基(homoalkyl)”。所述烷基任选地被一个或多个卤素原子取代。
术语“氟代烷基”是指如上定义的烷基,其中一个或多个氢原子被氟原子取代。
术语“亚烷基”本身或作为另一取代基的一部分是指衍生自烷基的二价基团,例如,但不限于,-CH 2CH 2CH 2CH 2-、-CH 2CH=CHCH 2-、-CH 2C≡CCH 2-、-CH 2CH 2CH(CH 2CH 2CH 3)CH 2-。烷基(或亚烷基)通常具有1自24个碳原子,本发明优选具有10或更少碳原子的基团。“低级烷基”或“低级亚烷基”是指链更短的烷基或亚烷基,通常具有八个或更少的碳原子。所述亚烷基任选地被一个或多个卤素原子取代。
术语“炔基”是指含有至少一个碳碳三键的碳链,其可以是线性或支链的、或其组合。炔基的示例包括乙炔基、炔丙基、3-甲基-1-戊炔基、2-庚炔基等。所述炔基任选地被一个或多个卤素原子取代。
术语“环烷基”是指单环或双环饱和碳环,各自具有3至10碳原子。环烷基的“稠和类似物”是指单环与芳基或杂芳基稠和,其中,连接位点在非芳香部分。环烷基及其稠和类似物的示例包括环丙基、环丁基、环戊基、环己基、四氢萘基、十氢萘基、二氢茚基等。所述环烷基任选地被一个或多个卤素原子取代。
术语“烷氧基”是指具有示出碳原子数的直链或支链烷氧基团。C 1-6烷氧基,例如,包括甲氧基、乙氧基、丙氧基、异丙氧基等。
除非另外说明,术语“杂烷基”本身或与另一术语组合是指由至少一个碳原子和至少一个选自O、N、P、Si、S的杂原子构成的稳定的直链或支链、或环烃基、或其组合,其中,氮原子、磷原子或硫原子可任选地被氧化以及氮原子可任选地被季胺化。杂原子O、N、P、S和Si可置于杂烷基内的任意位置或置于烷基与该分子的其余部分相连的位置。例如包括,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、-CH=CH-O-CH 3、-Si(CH 3) 3、-CH 2-CH=N-OCH 3、-CH=CH-N(CH 3)-CH 3、-O-CH 3、-O-CH 2-CH 3和-CN。最多两个或三个杂原子可以是连续的。例如,-CH 2-NH-OCH 3和-CH 2-O-Si(CH 3) 3。类似地,术语“杂亚烷基”本身或与其它术语组合是指衍生自杂烷基的二价基团,例如,但不限于,-CH 2-CH 2-S-CH 2-CH 2-和-CH 2-S-CH 2-CH 2-NH-CH 2-。对于杂亚烷基,杂原子可在链的任一端或两端(例如,亚烷基氧基、亚烷基二氧基、亚烷基胺基、亚烷基二氨基等)。此外,对于亚烷基和杂亚烷基连接基团,连接基团分子式的书写方向不表示连接基团的取向。例如,分子式-C(O)OR'-表示-C(O)OR'-和-R'OC(O)-。如上所述,本文所用的杂烷基包括通过杂原子连接在分子其余部分的那些基团,例如-C(O)R'、-C(O)NR'、-NR'R"、-OR'、-SR'和/或-SO 2R'。在述及“杂烷基”,随后述及如-NR'R”等具体杂烷基之处,应理解,术语杂烷基和-NR'R”不重复且不互相排斥。相反,引用这些具体的杂烷基以便更清晰。因而,术语“杂烷基”不应该在本文中解释为排除如-NR'R”等特定杂烷基。
术语“环烷氧基”是指结合于氧原子的如上定义的环烷基,如环丙氧基。
术语“氟烷氧基”是指一个或多个氢原子被氟取代的如上定义的烷氧基。
术语“芳基”是指仅含有碳原子的单环或双环芳基。芳基的“稠和类似物”是指芳基和单环的环烷基或单环的杂环基稠和,其中连接点位于芳基部分。芳基及其稠环类似物的例子包括苯基、萘基、茚满基、茚基、四氢萘基、2,3-二氢苯并呋喃基、二氢苯并吡喃基、1,4-苯并二噁烷基等。
术语“杂芳基”是指含有至少一个选自N、O和S的杂原子的单环或双环芳基。杂芳基的“稠和类似物”是指杂芳基和单环的环烷基或单环的杂环基稠和,其中连接点位于芳基部分。杂芳基的例子包括吡咯基、异唑基、异噻唑基、吡唑基、吡啶基、噁唑基、噁二唑基、噻二唑基、噻唑基、咪唑基、三唑基、四唑基、呋喃基、三嗪基、噻吩基、嘧啶基、哒嗪基、吡嗪基、苯并噁唑基、苯并噻唑基、苯并咪唑基、苯并呋喃基、苯并噻吩基、呋喃并(2,3-b)吡啶基、喹啉基、吲哚基、异喹啉基等。
所定义的烷基、芳基和杂芳基是未取代的或被至少一个选自取代基组成的组的取代基所取代。
所述取代基选自下组:卤原子、具有1至6个碳原子的烷基、具有1至6个碳原子的烷氧基、具有1至6个碳原子的卤代烷基、具有1至6个碳原子的卤代烷氧基、氰基、具有2至6个碳原子的炔基、具有1至6个碳原子的烷酰基、具有3至7个环原子的环烷基、杂芳基、芳基、具有7-10个碳原子的芳烷氧基、芳基羰基、氨基羰基、具有2至5个碳原子的烯基、具有1至6个碳原子的烷硫基、氨基亚磺酰基、氨基磺酰基、羟基、-SF 5、具有1至4个碳原子的羟基烷基、硝基、氨基、羧基、具有2至5个碳原子的烷氧羰基、具有1至4个碳原子的烷氧基烷基、具有1-4个碳原子的烷基磺酰基、具有1至4个碳原子的烷酰基氨基、具有1至6个碳原子的烷酰基(烷基)氨基、在烷酰基和烷基部分均具有1至6个碳原子的烷酰基胺基烷基、在烷酰基和各烷基部分均具有1至6个碳原子的烷酰基(烷基)胺基烷基、具有1至4个碳原子的烷基磺酰基胺基、具有1至6个碳原子的单烷基胺基羰基或二烷基胺基羰基、具有1至6个碳原子的单烷基胺基亚磺酰基或双烷基胺基亚磺酰基、具有1至6个碳原子的单烷基胺基磺酰基或双烷基胺基磺酰基、具有1至4个碳原子的胺基烷基、具有1至6个碳原子的单烷基胺基或二烷基胺基、在各烷基部分均有1至6个碳原子的单烷基胺基烷基或二烷基胺基烷基、具有7至10个碳原子的芳烷基、在烷基部分具有1至4个碳原子的杂芳烷基、在烷氧基部分具有从1至4个碳原子的杂芳基烷氧基和具有1至4个碳原子的烷基磺酰胺基。
如本文所用,术语“杂环”或“杂环的”或“杂环烷基”或“杂环基”是指饱和的、部分饱和的或不饱和的基团(但不是芳香性的),具有单环或稠环(包括桥环体系和螺环体系,环内具有1至10个碳原子和1至4个选自氮、硫或氧的杂原子,在稠环体系中,一个或多个环可以是环烷基、芳基或杂芳基,只要连接点通过非芳香性环。在一实施例中,杂环基团的氮原子和/或硫原子任选地被氧化,以提供N-氧化物、亚磺酰基和磺酰基部分。“杂环基”及其稠和类似物的例子包括吡咯烷基、哌啶基、哌嗪基、咪唑烷基、2,3-二氢呋喃(2,3-b)并吡啶基、苯并噁嗪基、四氢喹啉基、四氢异喹啉基、二氢吲哚基等。该术语也包括非芳香性的部分不饱和的单环,如通过氮原子连接的2-或4-吡啶酮或N-取代的-(1H,3H)-嘧啶-2,4-二酮类(N-取代的尿嘧啶)。
如本文所用,术语“取代的杂环的”或“取代的杂环烷基”或“取代的杂环基”是指被1到5(如1至3)个取代基所取代的杂环基团,所述取代基与取代的环烷基所定义的取代 基相同。
除非另外说明,术语“卤代的”或“卤素”本身或作为另一取代基的一部分是指氟、氯、溴或碘原子。另外,术语“卤代烷基”是指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”是指包括,但不限于,三氟甲基、2,2,2-三氟乙基、4-氯丁基、3-溴丙基等。
“前药”是指在体内转化为母体药物物质。在某些状况下,由于前药比母体药物更容易施用,因此经常使用前药。例如,前药经口服可为生物可利用的而母体药物不能。在药物组合物中,前药也可具有较母体药物更高的溶解度。前药的例子,但不限于,可以是式I化合物中的任一化合物以酯(前药)的形式施用,以促进跨细胞膜传输,在细胞膜中水溶性对迁移有害,而一旦处于水溶性有益的细胞内,该酯随后代谢水解为活性物质羧酸。前药的另一例子可以是键合酸基的短肽(聚氨基酸),其中,肽经代谢以释放活性部分。
光学异构体、非对映体、几何异构体和互变异构体
由于本发明的化合物含有一个或多个不对称中心,因此可作为外消旋体和外消旋混合物,单一对映体,非对映体混合物和单一非对映体。当提及本发明化合物或式(A)化合物时,应当理解为包括所有这些异构形式。
一些本文所述的化合物含有烯属(olefinic)双键,除非另有说明,否则是指包括E和Z两种几何异构体。
一些式(A)化合物可包含一个或多于一个的环体系,因此可存在顺式-和反式-异构体。本发明旨在包含所有这些顺式-和反式-异构体。
本文描述的一些化合物可存在不同的与氢原子连接的位点,被称为互变异构体。这样的例子可以是称为酮-烯醇互变异构体的酮和其烯醇形式。单个互变异构体以及其混合物均包括在式(A)化合物中。
式(A)化合物可被分离成对映异构体的非对映体对(diastereoisomeric pairs),例如通过从合适的溶剂,如甲醇或乙酸乙酯或它们的混合物中分级结晶。一对如此得到的对映异构体可通过常规方法,例如使用光学活性的胺或酸作为拆分试剂或在手性HPLC柱中来分离成单独的立体异构体。
或者,通式(A)化合物的任何对映异构体可通过使用光学纯原料或已知构型的试剂立体定向合成获得。
此外,式(A)化合物还可以包括一系列稳定同位素标记的类似物。例如,式(A)化合物中的一个或一个以上的质子可以被替换为氘原子,从而可提供药理活性改善的氘代类似物。
盐与剂型
如本文所用,术语“药学上可接受的盐”是指通式I化合物的非毒性酸或碱土金属盐。这些盐可在最终分离和纯化通式I化合物时原位制得、或分别将合适的有机或无机酸或碱与碱性或酸性官能团反应制得。代表性的盐包括,但不限于:乙酸盐、己二酸盐、藻酸盐、柠檬酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、硫酸氢盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、二葡糖酸盐、环戊烷丙酸盐、十二烷基硫酸盐、乙磺酸盐、葡萄糖庚酸盐、甘油磷酸盐、半硫酸盐、庚酸盐、己酸盐、富马酸盐、盐酸盐、氢溴酸盐、氢碘酸盐、2-羟基乙磺酸盐、乳酸盐、马来酸盐、甲磺酸盐、烟酸盐、2-萘基磺酸盐、草酸盐、双羟萘酸盐、果胶酸盐、硫氰酸盐、3-苯基丙酸盐、苦味酸盐、新戊酸 盐、丙酸盐、琥珀酸盐、硫酸盐、酒石酸盐、硫氰酸盐、对甲苯磺酸盐和十一烷酸盐。此外,含氮的碱性基团可被如下试剂季铵盐化:烷基卤化物,如甲基、乙基、丙基、丁基的氯化物、溴化物和碘化物;二烷基硫酸盐,如二甲基、二乙基、二丁基和二戊基硫酸酯;长链卤化物如癸基、月桂基、肉豆蔻基和硬脂基的氯化物、溴化物和碘化物;芳烷基卤化物如苄基和苯乙基溴化物等。由此得到水溶性或油溶性或可分散产品。可被用于形成药学上可接受的酸加成盐的酸的例子包括如盐酸、硫酸、磷酸的无机酸,和如草酸、马来酸、甲磺酸、琥珀酸、柠檬酸的有机酸。碱加成盐可在最终分离和纯化通式I的化合物时原位制得、或使羧酸部分分别与合适的碱(如药学上可接受的金属阳离子的氢氧化物,碳酸盐或碳酸氢盐)或氨、或有机伯、仲或叔胺反应制得。药学上可接受的盐包括,但不限于,基于碱金属和碱土金属的阳离子,如钠、锂、钾、钙、镁、铝的盐等,以及无毒的铵、季铵和胺阳离子,包括,但不限于:铵、四甲基铵、四乙基铵、甲胺、二甲胺、三甲胺、三乙胺、乙胺等。其它代表性的用于形成碱加成盐的有机胺包括二乙胺、乙二胺、乙醇胺、二乙醇胺、哌嗪等。
应理解,如本文所用,提及式(A)化合物也包括药学上可接受的盐。
用于口服的制剂也可以为硬明胶胶囊,其中活性成分与惰性固体稀释剂混合,例如,碳酸钙,磷酸钙或高岭土,或者为软明胶胶囊,其中活性成分与水或油介质混合,例如花生油,液体石蜡或橄榄油。
水性悬浮液含有与适于制备水性悬浮液的赋形剂混合的活性物质。这样的赋形剂为悬浮剂,例如羧甲基纤维素钠,甲基纤维素,羟丙基甲基纤维素,藻酸钠,聚乙烯吡咯烷酮,黄蓍胶和阿拉伯树胶;分散剂或润湿剂可以是天然存在的磷脂,例如卵磷脂,或环氧烷(alkylene oxide)与脂肪酸的缩合产物,例如聚氧乙烯硬脂酸酯,或环氧乙烷与长链脂族醇,例如十七碳乙烯-氧鲸蜡醇(heptadecaethylene–oxycetanol)的缩合产物,或环氧乙烷与衍生自脂肪酸和己糖醇的偏酯的缩合产物,例如,聚氧乙烯山梨糖醇单油酸酯,或环氧乙烷与衍生自脂肪酸和己糖醇酐的偏酯的缩合产物,例如,聚乙烯脱水山梨醇单油酸酯。水性混悬剂也可含有一种或多种防腐剂,例如对羟基苯甲酸乙酯或对羟基苯甲酸正丙酯,一种或多种着色剂,一种或多种调味剂,以及一种或多种甜味剂,如蔗糖,糖精或阿斯巴甜。
油性悬浮液可将活性成分悬浮在植物油中,例如花生油,橄榄油,芝麻油或椰子油,或悬浮在矿物油,如液体石蜡中配制。油性悬浮液可含有增稠剂,例如蜂蜡,硬石蜡或鲸蜡醇。可加入上述的甜味剂,可加入矫味剂以提供可口的口服制剂。这些组合物可通过加入抗氧化剂如抗坏血酸来保存。
适合通过添加水来制备水性悬浮液的可分散粉剂和颗粒剂提供与分散剂或润湿剂,悬浮剂和一种或多种防腐剂混合的活性成分。合适的分散剂或润湿剂和助悬剂由上文已提及的例子示出。也可以存在额外的赋形剂,例如甜味剂,调味剂和着色剂。
本发明的药物组合物也可以是水包油的乳液形式。油相可以是植物油,例如橄榄油或花生油,或矿物油,例如液体石蜡或这些的混合物。合适的乳化剂可以是天然存在的磷脂,例如大豆,卵磷脂以及衍生自脂肪酸和己糖醇酐的酯或偏酯,例如山梨糖醇单油酸酯,以及上述偏酯与环氧乙烷的缩合产物,例如聚氧乙烯失水山梨醇单油酸酯。该乳剂也可含有甜味剂和矫味剂。
糖浆和酏剂(elixirs)可以用甜味剂配制,例如甘油,丙二醇,山梨醇或蔗糖。这样的制剂也可含有缓和剂,防腐剂,调味剂和着色剂。该药物组合物可以是无菌可注射的水性或油性悬浮液的形式。该悬浮液可通过已知技术,使用上述那些合适的分散剂 或润湿剂和悬浮剂来配制。无菌注射制剂也可以是利用无毒的胃肠外可接受的稀释剂或溶剂配制的无菌注射溶液或悬浮液,例如1,3-丁二醇配制的溶液。可以使用的可接受载体和溶剂为水,林格氏溶液和等渗氯化钠溶液。此外,无菌,不挥发性油通常用作溶剂或悬浮介质。用于此目的的任何温和不挥发性油都可以使用,包括合成的单或双甘油酯。此外,脂肪酸如油酸可用在注射剂的制备中。
本发明的化合物也可以经鼻内或通过吸入给药,通常是来自干粉吸入器的干粉形式(单独,作为混合物,例如,含乳糖的干燥共混物,或作为混合组分颗粒,例如与磷脂如磷脂酰胆碱混合),或来自加压容器,泵,喷射器,雾化器(使用电流来产生细雾优选的I雾化器)或喷雾器的气溶胶喷雾器,使用或不使用合适的推进剂,如1,1,1,2-四氟乙烷或1,1,1,2,3,3,3-七氟丙烷。对于鼻内使用,粉末可以包括生物粘附剂,例如壳聚糖或环糊精。
加压容器、泵、喷射器、雾化器或喷雾器包含本发明化合物的溶液或悬浮液,所述溶液或悬浮液包含例如,乙醇,含水乙醇,或用于分散,增溶,或延长释放活性的适宜替代试剂,作为溶剂的推进剂和可选的表面活性剂,如脱水山梨糖醇三油酸酯,油酸或低聚乳酸。
在以干粉或悬浮液制剂的形式使用前,将药物产品微粉化至适于吸入递送(通常小于5微米)的尺寸。
可以通过任何适当的粉碎方法实现,例如螺旋喷射研磨,流化床喷射研磨,超临界流体加工以形成纳米颗粒,高压均化,或喷雾干燥。
用于吸入器或吹入器的胶囊剂(例如由明胶或羟丙基甲基纤维素制备),泡罩和药盒可配制成含有本发明化合物的粉末,合适的粉质如乳糖或淀粉以及性能改性剂,如L-亮氨酸,甘露醇或硬脂酸镁。乳糖可以是无水的或单水合物形式,优选后者。其它合适的赋形剂包括葡聚糖,葡萄糖,麦芽糖,山梨醇,木糖醇,果糖,蔗糖和海藻糖。
在使用电流动力学方法产生细雾的雾化器中使用的合适溶液制剂可含有每次启动从10g到20mg的本发明化合物,启动体积可从11到1001变化。典型制剂可包含式(A)化合物,丙二醇,无菌水,乙醇和氯化钠。可被用于代替丙二醇的替代溶剂包括甘油和聚乙二醇。
合适的调味剂,如薄荷醇和左薄荷脑,或甜味剂,例如糖精或糖精钠,可以加入到本发明的用于吸入/鼻内给药的那些制剂中。
用于吸入/鼻内给药的制剂可以配制成即刻使用的和/或改进释放的,例如,使用聚(DL-乳酸-乙醇酸(PGLA)。改进释放制剂包括延迟释放,持续释放,脉冲释放,受控释放,靶向释放和程序释放。
在干粉吸入器和气雾剂的情况下,剂量单位是通过提供计量量的阀来确定。本发明的单位通常安排为施用含0.001至10毫克式(A)化合物的计量剂量或“喷雾”。每日总剂量通常从0.001至10毫克,可在一天内以单剂量或更通常作为分批剂量施用。
式(A)化合物也可以栓剂形式进行药物的直肠给药。这些组合物可以通过将药物与合适的无刺激性赋形剂混合制备,该赋形剂在常温下为固体,但在直肠温度下为液体,因此将在直肠中融化以释放药物。这类物质为可可脂和聚乙二醇。
对于局部使用,施用含式(A)化合物的霜剂,软膏剂,凝胶剂,溶液或悬浮液等。(对于本申请的目的,局部应用应包括漱口剂和含漱剂)。
每天约0.01毫克到约140毫克/千克体重的剂量水平在上述条件的治疗中是有用的,或每名患者每天约0.5mg至约7g是有用。例如,可通过每天每公斤体重施用约 0.01至50毫克化合物,或每天对每名患者施用约0.5mg至3.5克,优选每人每天2.5毫克至1克化合物来有效治疗炎症。
可与载体材料组合产生单一剂型的活性成分的量将根据所治疗的宿主和具体给药模式变化。例如,用于人口服给药的制剂可含有0.5毫克至5g活性剂,活性剂与适当方便数量的载体材料复合,载体材料可在总的组合物的约5%到约95%范围变化。剂量单位形式一般含有约1毫克至约500毫克的活性成分,通常为25毫克,50毫克,100毫克,200毫克,300毫克,400毫克,500毫克,600毫克,800毫克或1000毫克。
然而,应当理解,对于任何特定患者的具体剂量水平取决于多种因素,包括年龄,体重,一般健康状况,性别,饮食,给药时间,给药途径,排泄速率,药物组合以及正在接受治疗的具体疾病的严重程度。
应用
本发明的化合物可用于治疗与BTK的异常活性以及BTK突变体(如C481S)相关的疾病。
本发明还包括使用对患者施用治疗有效量的式(A)化合物的方法来治疗患者疾病的方法。
更特别地,本发明的化合物对于治疗异常细胞生长和/或凋亡失调的疾病是有用的,如间皮瘤(mesothioloma),膀胱癌,胰腺癌,皮肤癌,头部或颈部癌症,皮肤或眼内黑色素瘤,卵巢癌,乳腺癌,子宫癌,子宫内膜输卵管癌,子宫颈癌,阴道癌,外阴癌,骨癌,宫颈癌,结肠癌,直肠癌,肛门区域癌,胃癌,胃肠道癌(胃,结直肠和十二指肠),慢性淋巴细胞性白血病,食管癌,小肠癌,内分泌系统癌,甲状腺癌,甲状旁腺癌的癌腺,肾上腺癌,软组织癌,尿道癌,阴茎癌,睾丸癌,肝细胞癌(肝癌和胆汁导管),原发性或继发性中枢神经系统肿瘤,原发性或继发性脑肿瘤,霍奇金肉瘤,慢性或急性白血病,慢性髓细胞性白血病,淋巴细胞性淋巴瘤,成淋巴细胞性白血病,T-细胞或B-细胞来源的滤泡性淋巴瘤,黑色素瘤,多发性骨髓瘤,口腔癌,卵巢癌,非小细胞肺癌,前列腺癌,小细胞肺癌,肾和输尿管癌,肾细胞癌,肾盂癌,中枢神经系统肿瘤,原发性中枢神经系统淋巴瘤,非何杰金氏淋巴瘤,脊轴肿瘤,脑干神经胶质瘤,垂体腺瘤,肾上腺皮质癌,胆囊癌,脾癌,胆管癌,纤维肉瘤,神经母细胞瘤,眼癌(retinoblasitoma),或其组合。另一实施方案包括对患者的间皮瘤(mesothioloma),膀胱癌,胰腺癌,皮肤癌,头部或颈部癌症,皮肤或眼内黑色素瘤,乳腺癌,子宫癌,输卵管癌,子宫内膜癌,子宫颈癌,阴道,外阴癌,骨癌,卵巢癌,子宫颈癌,结肠癌,直肠癌,肛区癌,胃癌,胃肠道癌(胃,结直肠和十二指肠),慢性淋巴细胞性白血病,食管癌,小肠癌,内分泌系统癌,甲状腺癌,甲状旁腺癌,肾上腺癌,软组织肉瘤,尿道癌,阴茎癌,睾丸癌,肝细胞癌(肝癌和胆汁导管),原发性或继发性中枢神经系统肿瘤,原发性或继发性脑肿瘤,霍奇金病,慢性或者急性白血病,慢性髓细胞性白血病,淋巴细胞性淋巴瘤,成淋巴细胞性白血病,滤泡性淋巴瘤,T细胞或B-细胞来源的淋巴恶性肿瘤,黑色素瘤,多发性骨髓瘤,口腔癌,卵巢癌,非小细胞肺癌,前列腺癌,小细胞肺癌,肾和输尿管癌,肾细胞癌,肾盂癌,在中枢神经系统,原发性中枢神经系统淋巴瘤,非何杰金氏淋巴瘤,脊轴肿瘤,脑干神经胶质瘤,垂体腺瘤,肾上腺皮质癌,胆囊癌,脾癌,胆管癌,纤维肉瘤,神经母细胞瘤,眼癌(retinoblasitoma)或上述一种或多种癌症组合的治疗方法,所述方法包括给予治疗有效量的式(A)化合物。
式(A)化合物也可用于治疗自身免疫疾病和炎性疾病,例如,炎性肠病,关节炎,狼疮,类风湿关节炎,银屑病关节炎,骨性关节炎和幼年型关节炎,斯蒂尔病,糖尿病,重症肌无力,桥本氏甲状腺炎,奥德氏甲状腺炎,格雷夫斯病,干燥综合征,多发性硬化,格林-巴利综合征(GuiUain-Barre syndrome)、急性播散性脑脊髓炎、阿狄森氏病(Addison's disease)、眼球斜视痉挛综合征(opsoclonus-myoclonus syndrome)、强直性脊柱炎、抗磷脂抗体综合征、再生障碍性贫血、自身免疫性肝炎、腹部疾病、古德帕斯彻氏综合征(Goodpasture's syndrome)、特发性血小板减少性紫癜、视神经炎、硬皮病、原发性胆汁性肝硬化、莱特尔氏综合征(Reiter's syndrome)、高安动脉炎(Takayasu's arteritis)、颞动脉炎、温抗体型自身免疫性溶血性贫血(warm autoimmune hemolytic anemia)、韦格纳氏肉芽肿病(Wegener's granulomatosis)、银屑病、全身毛发脱失、贝切特氏病(Behcet's disease)、慢性疲劳、自律神经失调、子宫内膜异位症、间质性膀胱炎、神经性肌强直、硬皮病、外阴痛、移植、输血、过敏反应、过敏症、I型超敏反应、过敏性结膜炎、过敏性鼻炎、特应性皮炎、哮喘、阑尾炎、眼睑炎、细支气管炎、支气管炎、滑囊炎、宫颈炎、胆管炎、胆囊炎、结肠炎、结膜炎、膀胱炎、泪腺炎、皮炎、皮肌炎、脑炎、心内膜炎、子宫内膜炎、肠炎、小肠结肠炎、上髁炎、附睾炎、筋膜炎、纤维组织炎、胃炎、胃肠炎、肝炎、化脓性汗腺炎、喉炎、乳腺炎、脑膜炎、脊髓炎、心肌炎、肌炎、肾炎、卵巢炎、睾丸炎、骨炎、耳炎、胰腺炎、腮腺炎、心包炎、腹膜炎、咽炎、肋膜炎、静脉炎、肺炎(pneumonitis)、肺炎(pneumonia)、直肠炎、前列腺炎、肾盂肾炎、鼻炎、输卵管炎、鼻窦炎、口炎、滑膜炎、腱炎、扁桃体炎、葡萄膜炎、阴道炎、血管炎或外阴炎。
在一些实施例中,如果所述对象患有癌症,除了上述化合物之外,给该对象施用抗癌剂。在一实施例中,所述抗癌剂为丝裂原活化蛋白激酶信号通路抑制剂,例如U0126、PD98059、PD184352、PD0325901、ARRY-142886、SB239063、SP600125、BAY 43-9006、渥曼青霉素、或LY294002。
本发明还涉及包括至少一种式(A)化合物本身或至少一种式(A)化合物与一种或多种药学上可接受的赋形剂结合的药物组合物。
根据本发明的药物组合物,可以更特别地提及的是那些适合于口服、肠胃外、经鼻、经皮或穿皮、直肠、经舌、眼部或呼吸道给药,特别是片剂或糖锭剂、舌下片剂、香囊剂、药包(packets)、明胶胶囊剂、舌下剂(glossettes)、锭剂、栓剂、乳膏剂、软膏剂、皮肤凝胶剂、可饮用或可注射的安瓿。
剂量可根据患者的性别,年龄和体重,给药途径,适应症的性质,或者任何相关的治疗而变化,在一次或多次施用中,剂量范围从每24小时0.01毫克至1克。
此外,本发明还涉及式(A)与一种或多种抗癌剂的组合,所述抗癌剂选自细胞毒性剂,有丝分裂毒素,抗代谢物,蛋白酶体抑制剂和激酶抑制剂,并且涉及该类型的组合在制备用于治疗癌症的药物中的用途。
本发明的化合物也可与治疗癌症的放疗联用。
式(A)化合物也可望用作与治疗剂相组合的化疗剂,治疗剂包括,但不限于,血管生成抑制剂,抗增殖剂,其他激酶抑制剂,其他受体酪氨酸激酶抑制剂,极光(aurora)激酶抑制剂,Polo样激酶抑制剂,bcr-abl激酶抑制剂,生长因子抑制剂,COX-2抑制剂,非甾体抗炎药(NSAIDS),抗有丝分裂剂,烷化剂,抗代谢物,嵌入抗生素,含铂试剂,生长因子抑制剂,电离辐射,细胞周期抑制剂,酶,拓扑异构酶抑制剂,生物反应调节剂,免疫调节剂,免疫制剂,抗体,激素疗法,类维生素A/三角肌植物生物碱,蛋白酶体抑制剂,HSP-90抑制剂,组蛋白脱乙酰酶抑制剂(HDAC)抑制剂,嘌呤类似物,嘧啶类似物,MEK抑制剂,CDK抑制剂,ERBB2受体抑制剂,mTOR抑制剂,BCL抑制剂,MCL抑制剂及其组合,PD1抗体, PDL1抗体,CTLA4抗体,IDO抑制剂,TDO抑制剂,A2a拮抗剂,精氨酸酶抑制剂,以及其它抗肿瘤剂。
血管生成抑制剂包括,但不限于,EGFR抑制剂,PDGFR抑制剂,VEGFR抑制剂,TTE2抑制剂,IGF1R抑制剂,基质金属蛋白酶-2(MMP-2)抑制剂,基质金属蛋白酶-9(MMP-9)抑制剂,血小板反应蛋白类似物,例如血小板反应蛋白-1和N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-IIe-Arg-Pro-NHCH 2CH 3或其盐和N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-Ile-Arg-PrO-NHCH 2CH 3类似物,如N-Ac-GlyVal-D-AIle-Ser-GIn-Ile-Arg-ProNHCH 2CH 3或其盐。
EGFR抑制剂的例子包括,但不限于,易瑞沙(吉非替尼),特罗凯(厄洛替尼或OSI-774),埃克替尼,西妥昔单抗(西妥昔单抗),EMD-7200,ABX-EGF,HR3,IgA抗体,TP-38(IVAX),EGFR融合蛋白,EGF-疫苗,AZD9291,CO1686,抗EGFr免疫脂质体和Tykerb(拉帕替尼)。
PDGFR抑制剂的例子包括,但不限于,CP-673,451和CP-868596。
VEGFR抑制剂的例子包括,但不限于,阿瓦斯丁(贝伐单抗),纾癌特(舒尼替尼,SUl1248),多吉美(索拉非尼,BAY43-9006),CP-547,632,阿西替尼(AG13736),阿帕替尼,卡博替尼,易瑞莎(凡德他尼,ZD-6474),AEE788,AZD-2171,VEGF trap,瓦他拉尼(PTK-787,ZK-222584),哌加他尼,M862,帕唑帕尼(GW786034),BC-00016,ABT-869和血管酶(angiozyme)。
血小板反应蛋白类似物的例子包括,但不限于,ABT-510。
BCL抑制剂的例子包括,但不限于,奥巴克拉(obatoclax)和那维克拉(navitoclax),ABT199。
极光激酶抑制剂的例子包括,但不限于,VX-680,AZD-1152和MLN-8054。polo样激酶抑制剂的例子包括,但不限于,BI-2536。
BCR-ABL激酶抑制剂的例子包括,但不限于,格列卫(伊马替尼),泼那替尼(ponatinib),尼洛替尼和达沙替尼(BMS354825)。
含铂试剂的例子包括,但不限于,顺铂,伯尔(卡铂),依铂,洛铂,奈达铂,乐沙定(奥沙利铂)或沙铂。
mTOR抑制剂的例子包括,但不限于,CCI-779,雷帕霉素,替西罗莫司,依维莫司,RAD001,INK-128和里达福洛莫司(ridaforolimus)。
HSP-90抑制剂的例子包括,但不限于,格尔德霉素,根赤壳菌素,17-AAG,KOS-953,17-DMAG,CNF-101,CNF-1010,17-AAG-nab,NCS-683664,迈考拉伯(Mycograb),CNF-2024,PU3,PU24FC1,VER49009,IPI-504,SNX-2112和STA-9090。
组蛋白脱乙酰酶抑制剂(HDAC)的例子包括,但不限于,辛二酰苯胺异羟肟酸(SAHA),MS-275,丙戊酸,TSA,LAQ-824,特拉泼星(Trapoxin),图拔星(tubacin),图拔他汀(tubastatin),ACY-1215和缩酚酸肽(Depsipeptide)。
MEK抑制剂的例子包括,但不限于,PD325901,ARRY-142886,ARRY-438162和PD98059。
CDK抑制剂的例子包括,但不限于,夫拉平度(flavopyridol),MCS-5A,CVT-2584,色立昔布(seliciclib)(CYC-202,R-洛扣瓦汀(roscovitine)),ZK-304709,PHA-690509,BMI-1040,GPC-286199,BMS-387,032,PD0332991和AZD-5438。
COX-2抑制剂的例子包括,但不限于,CELEBREX TM(塞来昔布),帕瑞考昔,地拉考昔,ABT-963,MK-663(依托考昔),COX-189(罗美昔布),BMS347070,RS57067,NS-398,伐地考昔,帕雷考昔,罗非昔布,SD-8381,4-甲基-2-(3,4-二甲基苯基)-1-(4-氨磺酰基-苯基-1H-吡咯,T-614,JTE-522,S-2474,SVT-2016,CT-3,SC-58125和Arcoxia(依托考昔)。
非甾体类抗炎药(NSAID)的例子包括,但不限于,双水杨酯(Amigesic),二氟尼柳 (Dolobid),布洛芬(布洛芬),酮洛芬(Orudis),萘丁美酮(Relafen),吡罗昔康(Feldene),萘普生(Aleve,萘普生),双氯芬酸(扶他林),吲哚美辛(消炎痛),舒林酸(Clinoril),托美丁(Tolectin),依托度酸(碘值),酮咯酸(Toradol)和奥沙普秦(Daypro)。
ERBB2受体抑制剂的例子包括,但不限于,CP-724-714,CI-1033,(卡奈替尼),赫赛汀(曲妥珠单抗),Omitarg(2C4,佩图珠单抗-petuzumab),TAK-165,GW-572016(Ionafarnib)GW-282974,EKB-569,PI-166,dHER2(HER2疫苗),APC8024(HER2疫苗),抗HER/2neu双特异性抗体,B7.her2IgG3,AS HER2的三功能双特异性抗体,单克隆抗体AR-209和单克隆抗体2B-1。
烷基化剂的例子包括,但不限于,氮芥N-氧化物,环磷酰胺,异环磷酰胺,氯乙环磷酰胺,苯丁酸氮芥,美法仑,白消安,二溴甘露醇,卡波醌,塞替派,雷莫司汀,尼莫司汀,替莫唑胺,AMD-473,六甲蜜胺,AP-5280,阿帕昆(apaziquone),布洛塔星(brostallicin),苯达莫司汀,卡莫司汀,雌莫司汀,福莫司汀,葡磷酰胺,KW-2170,马磷酰胺,二溴卫矛醇以及,卡莫司汀(BCNU),洛莫司汀(CCNU),白消安,曲奥舒凡,氮烯咪胺和替莫唑胺。
抗代谢物的例子包括,但不限于,甲氨蝶呤,6-巯基嘌呤核苷,巯基嘌呤,尿嘧啶类似物,如5-氟尿嘧啶(5-FU)单独或组合使用亚叶酸,替加氟,UFT,去氧氟尿苷,卡莫氟,阿糖胞苷,阿糖胞苷,依诺他滨,S-I,力比泰(培美曲塞二钠,LY231514,MTA),健择(吉西他滨),氟达拉滨,5-氮杂胞苷,卡培他滨,克拉屈滨,氯法拉滨,地西他滨,依氟鸟氨酸,ethnylcytidine,阿糖胞苷,羟基脲,TS-I,美法仑,奈拉滨,洛拉曲克,ocfosate,培美曲塞二钠(disodium premetrexed),喷司他丁,pelitrexol,雷替曲塞,triapine,三甲曲沙,阿糖腺苷,长春新碱,长春瑞滨,霉酚酸,噻唑呋林,利巴韦林,EICAR,羟基脲和去铁胺。
抗生素的实例包括嵌入抗生素,但不限于,阿柔比星,放线菌素(如放线菌素D),氨柔比星,阿那米星(annamycin),阿霉素,博莱霉素a,博来霉素b,柔红霉素,多柔比星,依沙芦星,表柔比星(epirbucin),格拉布星(glarbuicin),伊达比星,丝裂霉素C,奈莫柔比星,新制癌菌素,培洛霉素,吡柔比星,蝴蝶霉素,丁斯酯,链佐星,戊柔比星,净司他丁和其组合。
拓扑异构酶抑制剂的例子包括,但不限于,一种或多种选自下组的试剂:阿柔比星,氨萘非特(amonafide),贝洛替康(belotecan),喜树碱,10-羟基喜树碱,9-氨基喜树碱,氟替康(diflomotecan),伊立替康盐酸盐(Camptosar),edotecarin,表柔比星(Ellence),依托泊苷,依喜替康(exatecan),吉马替康,勒托替康,orathecin(Supergen),BN-80915,米托蒽醌,匹拉布星(pirarbucin),匹杉琼(pixantrone),卢比替康,索布佐生,SN-38,他弗泊苷(tafluposide)和托泊替康。
抗体的例子包括,但不限于,利妥昔单抗,西妥昔单抗,贝伐单抗,特拉图单抗(Trastuzimab),特异性CD40抗体和特异性IGF1R抗体。
激素疗法的实例包括,但不限于,依西美坦(阿诺新),醋酸亮丙瑞林,阿那曲唑(瑞宁得),福斯林(fosrelin,诺雷德),戈舍瑞林(goserelin),度骨化醇,法倔唑,福美坦,他莫昔芬柠檬酸盐(他莫昔芬),康士得,阿巴瑞克,TRELSTAR,非那雄胺,氟维司群,托瑞米芬,雷洛昔芬,拉索昔芬,来曲唑,氟他胺,比卡鲁胺,甲地孕酮(megesterol),米非司酮,尼鲁米特,地塞米松,强的松等皮质激素
类维生素A/三角肌(deltoids)的例子包括,但不限于,西奥骨化醇(seocalcitol)(EB1089,CB1093),来沙骨化醇(lexacalcitrol,KH 1060),芬维A胺,阿利微A酸(Aliretinoin),蓓萨罗丁和LGD-1550。
植物生物碱的实例包括,但不限于,长春新碱,长春碱,长春地辛和长春瑞滨。
蛋白酶体抑制剂的实例包括,但不限于,硼替佐米(万珂),MGL32,NPI-0052和PR-171。
免疫制剂的实例包括,但不限于,干扰素和许多其它免疫促进剂。干扰素包括干扰素α,干扰素α-2a,干扰素α-2b,干扰素β,干扰素γ-1a中,干扰素γ-1b(Actimmune)或干扰素γ-nl及其组合。其他促进剂包括非尔司亭(filgrastin),香菇多糖(lentinan),西佐喃(sizofilan),TheraCys,乌苯美司(ubenimex),WF-10,阿地白介素(aldesleukin),阿仑单抗,BAM-002,达卡巴嗪(decarbazine),赛尼哌(daclizumab),地尼白介素(denileukin),吉妥单抗(gemtuzumab ozogamicin),替伊莫(ibritumomab),咪喹莫特(imiquimod),来格司亭(lenograstim),香菇多糖(lentinan),黑色素瘤疫苗(melanoma vaccine)(Corixa公司),莫拉司亭(molgramostim),OncoVAC-CL,沙莫司亭(sargaramostim),他索纳明(tasonermin),tecleukin,thymalasin,托西莫单抗(tositumomab),维如利金(Virulizin),Z-100,依帕珠单抗(epratuzumab),米妥莫单抗(mitumomab),奥戈伏单抗(oregovomab),佩图莫单抗(pemtumomab,Y-muHMFGl),普罗文奇(Provenge)(Dendreon公司),STING激活剂,IDO抑制剂,精氨酸代谢酶抑制剂,CTLA4(细胞毒性淋巴细胞抗原4)抗体以及能阻断CTLA4的试剂,PD1抗体或PD-L1等免疫检查点蛋白抑制剂或抗体。
生物反应调节剂的例子是调节有生命的生物体的防御机制或生物反应(如组织细胞的存活、生长或分化)以引导其具有抗肿瘤活性的试剂。这样的药物包括云芝胞内多糖(Krestin),香菇多糖,西佐弗兰(sizofrran),溶链菌和乌苯美司。
嘧啶类似物的例子包括,但不限于,5-氟尿嘧啶,氟尿苷,去氧氟尿苷,拉地曲德(Ratitrexed),阿糖胞苷(阿糖胞苷C),阿糖胞苷,氟达拉滨和吉西他滨。
嘌呤类似物的例子包括,但不限于,巯基嘌呤和硫鸟嘌呤。
免疫调节剂的例子包括,但不限于,沙利度胺和来那度胺。
抗有丝分裂剂的例子包括,但不限于,紫杉醇,多西他赛,白蛋白紫杉醇(ABRAXANE),埃坡霉素D(KOS-862)和ZK-EPO。
合成
本发明的化合物可通过如下反应式制备:
合成流程
Figure PCTCN2020072551-appb-000018
本发明的化合物可以通过化学合成的方法制备,其实例在下文示出。应理解的是,所述过程中的步骤的顺序可以改变,那些具体提及的试剂、溶剂和反应条件可以替换,如有必要,易反应的部位可被保护和脱保护。
下面的缩写具有如下所示的意义。DBU是指1,8-二氮杂双环[5.4.0]十一碳-7-烯;DIBAL表示二异丁基氢化铝;DIEA指二异丙基乙胺;DMAP是指N,N-二甲基氨基吡啶;DME指1,2-二甲氧基乙烷;DMF指N,N-二甲基甲酰胺;DMPE是指1,2-双(二甲基膦基)乙烷;DMSO表示二甲亚砜;DPPB指1,4-双(二苯基膦基)丁烷;dppe指1,2-双(二苯基膦基)乙烷;dppf指 1,1'-双(二苯基膦基)二茂铁;dppm指1,1'-双(二苯基膦基)甲烷;DIAD指偶氮二甲酸二异丙酯;EDCI表示1-(3-二甲基氨基丙基)-3-乙基碳二亚胺;HATU表示2-(7-氮杂-1H-苯并三唑-1-基)-1,1,3,3-四甲基脲六氟磷酸盐;HMPA表示六甲基磷酰胺;IPA是指异丙醇;LDA是指二异丙基氨基锂;LHMDS是指二(三甲基硅基)氨基锂;LAH表示氢化铝锂;NCS指N-氯琥珀酰亚胺;PyBOP是指苯并三唑-1-基-氧基三吡咯烷基磷苯并三唑六氟磷酸盐;TDA-I是指三(2-(2-甲氧基乙氧基)乙基)胺;DCM指二氯甲烷;TEA是指三乙胺,TFA是指三氟乙酸;THF是指四氢呋喃;NCS指N-氯琥珀酰亚胺;NMM是指N-甲基吗啉;NMP是指N-甲基吡咯烷酮;PPh 3指三苯基膦,rt是指室温。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
实施例1
(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000019
步骤1:(1R,3S)-3-羟基环己基乙酸酯
Figure PCTCN2020072551-appb-000020
氮气保护下,向一个烘干的100毫升二口烧瓶中,加入顺-1,3-环己二醇(1.00g,8.61mmol)和Candida Antarctica lipase B(CALB)(180mg)。另取一个50毫升二口烧瓶,向其中加入乙酸4-氯苯酯(2.20g,12.91mmol)以及30毫升甲苯溶液,用氮气脱气5分钟后,将该溶液加入到上述反应瓶中。室温搅拌过夜,TLC监测反应,8小时后反应结束,除去溶剂,粗产品经硅胶柱纯化(石油醚:乙酸乙酯=3:1),得到产品1.20g,产率为88%。
1H NMR(400MHz,CDCl 3):δ4.78-4.71(m,1H),3.74-3.67(m,1H),2.25-2.21(m,1H),2.05(s,3H),1.92-1.89(m,2H),1.83-1.80(m,2H),1.44-1.23(m,4H).
步骤2:光学活性测试:(1S,3R)-3-乙酰氧基环己基(R)-3,3,3-三氟-2-甲氧基-2-苯基丙酸酯
Figure PCTCN2020072551-appb-000021
在一个25毫升烧瓶中,加入(1R,3S)-3-羟基环己基乙酸酯(15mg,0.10mmol),吡啶(15mg,0.19mmol)以及5毫升二氯甲烷。在冰水浴冷却下,向体系中加入(S)-3,3,3-三氟-2-甲氧基-2-苯基丙酰氯(24mg,0.10mmol)的2毫升二氯甲烷溶液,TLC监测反应,原料消失后,制备TLC纯化(石油醚:乙酸乙酯=3:1),得35mg产品,产率:99%。
1H NMR(400MHz,Acetone-d 6):δ7.56-7.54(m,2H),7.49-7.46(m,3H),5.13-5.06(m,1H),4.81-4.73(m,1H),3.57(s,3H),2.30-2.25(m,1H),2.08(m,1H),1.93(s,3H),1.90-1.84(m,2H),1.54-1.42(m,3H),1.40-1.29(m,1H).
19F NMR(400MHz,Acetone-d 6):δ-72.71(s).
步骤3:(1R,3R)-3-(3-(4-苯氧基苯基)-4-((三苯基-5-亚膦基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯和(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯
Figure PCTCN2020072551-appb-000022
氮气保护下,向一充分烘干的250毫升三口烧瓶中,加入三苯基膦(12.44g,47.41mmol),150毫升超干四氢呋喃溶剂,并把烧瓶置于冰浴中。冷却至0摄氏度后,向体系中逐滴加入偶氮二甲酸二异丙酯(9.59g,47.41mmol)。滴加完后,继续搅拌40分钟,有白色沉淀析出,继续搅拌10分钟。向体系中依次加入3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(6.04g,19.91mmol)和(1R,3S)-3-羟基环己基乙酸酯(3.00g,18.96mmol),加完之后,体系变为澄清浅黄色体系。室温搅拌过夜,TLC监测反应结束。向体系中加入5毫升饱和氯化铵溶液淬灭反应。除去四氢呋喃,加入100毫升二氯甲烷,50毫升饱和食盐水,分液,二氯甲烷萃取(50mL×2),合并有机相,无水硫酸钠干燥。过滤,滤液中加入硅胶旋干,硅胶柱纯化(石油醚(含10%二氯甲烷):乙酸乙酯=100:1~100:2),得到化合物(1R,3R)-3-(3-(4-苯氧基苯基)-4-((三苯基-5-亚膦基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯3.00g,产率:20%;用二氯甲烷:甲醇=100:1~100:2,得到产物(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯3.40g,产率:40%。
(1R,3R)-3-(3-(4-苯氧基苯基)-4-((三苯基-5-亚膦基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基):
1H NMR(400MHz,CDCl 3):δ8.43-8.40(d,2H),8.07(s,1H),7.81-7.78(m,6H),7.52-7.50(m,3H),7.43-7.37(m,8H),7.13-7.07(m,5H),5.35(m,1H),5.12-5.08(m,1H),2.57-2.50(m,1H),2.19-2.15(m,1H),2.19(s,3H),1.9-1.98(m,1H),1.93-1.90(m,1H),1.84-1.74(m,1H),1.66-1.61(m,1H),1.43-1.40(m,1H).
MS ESI:m/z=704,[M+H] +.
(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯:
1H NMR(400MHz,CDCl 3):δ8.39(s,1H),7.67-7.64(m,2H),7.41-7.37(m,2H),7.17-7.14(m,3H),7.09-7.07(m,2H),5.52(br-s,2H),5.35-5.30(m,1H),5.18-5.10(m,1H),2.51-2.44(m,1H),2.23-2.20(m,1H),2.13(s,3H),2.11-2.08(m,2H),1.97-1.77(m,4H),1.66-1.58(m,1H).
MS ESI:m/z=444,[M+H] +.
步骤4:(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯
Figure PCTCN2020072551-appb-000023
在一烘干的25毫升烧瓶中,加入(1R,3R)-3-(3-(4-苯氧基苯基)-4-((三苯基-5-亚膦基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)(0.30g,0.43mmol),再加入5毫升(乙酸/水v/v=1/1)混合溶剂,加热回流。TLC监测反应,待原料反应完毕,加入二氯甲烷萃取(10mL×3),合并有机相,经硅胶柱纯化(二氯甲烷:甲醇=100:1~100:2)得到产品180mg,产率:95%。
1H NMR(400MHz,CDCl 3):δ8.39(s,1H),7.67-7.64(m,2H),7.41-7.37(m,2H),7.17-7.14(m,3H),7.09-7.07(m,2H),5.52(br-s,2H),5.35-5.30(m,1H),5.18-5.10(m,1H),2.51-2.44(m,1H),2.23-2.20(m,1H),2.13(s,3H),2.11-2.08(m,2H),1.97-1.77(m,4H),1.66-1.58(m,1H).
MS ESI:m/z=444,[M+H] +.
步骤5:(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000024
在一50毫升单口烧瓶中,加入(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(1.50g,3.38mmol),30毫升甲醇。搅拌下,向其中加入无水LiOH(0.25g,10.28mmol),室温搅拌过夜。TLC监测反应结束。过滤除去LiOH,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1~100:3)纯化,得到产品1.30g,产率:96%。
1H NMR(400MHz,CDCl 3):δ8.36(s,1H),7.67-7.63(m,2H),7.41-7.37(m,2H),7.19-7.14(m,3H),7.09-7.07(m,2H),5.67(br-s,2H),5.28-5.23(m,1H),4.40(m,1H),2.41-2.34(m,1H),2.15-2.07(m,4H),1.84-1.73(m,2H),1.69-1.61(m,1H).
MS ESI:m/z=402,[M+H] +.
实施例2
(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000025
步骤1:(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)苯甲酸环己酯
Figure PCTCN2020072551-appb-000026
在一25毫升烘干的烧瓶中,加入三苯基膦(131mg,0.50mmol),10毫升四氢呋喃,搅拌至澄清。在冰水浴冷却下,向烧瓶中慢慢滴加偶氮二甲酸二异丙酯(101mg,0.50mmol),保持5摄氏度以下反应1小时,析出白色沉淀。继续搅拌10分钟后,向体系中加入苯甲酸(61mg,0.50mmol)和(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己-1-醇(100mg,0.25mmol)的四氢呋喃混合溶液,滴加完毕,撤去冰浴,使反应液自然升至室温。反应2个小时后TLC监测反应结束,加入硅胶,旋干,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品110mg,产率:87%。
1H NMR(400MHz,Acetone-d 6):δ8.27(s,1H),8.05-8.03(m,2H),7.78-7.74(m,2H),7.64-7.61(m,1H),7.52-7.48(m,2H),7.46-7.42(m,2H),7.21-7.17(m,3H),7.13-7.11(m,2H),6.38(br-s,2H),5.25-5.17(m,1H),5.04-4.96(m,1H),2.50-2.45(m,1H),2.42-2.30(m,1H),2.28-2.20(m,1H),2.18-2.11(m,1H),1.76-1.66(m,4H).
MS ESI:m/z=506,[M+H] +.
步骤2:(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000027
在一25毫升烧瓶中,加入(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)苯甲酸环己酯(100mg,0.20mmol),5毫升甲醇,向其中分批加入无水氢氧化锂(14mg,0.59mmol),室温搅拌过夜。TLC监测反应结束。过滤除去氢氧化锂,加入硅 胶,旋干,经硅胶柱(二氯甲烷:甲醇=100:2~100:3)纯化,得到产品75mg,产率:94%。
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.67-7.64(m,2H),7.41-7.37(m,2H),7.17-7.14(m,3H),7.09-7.07(m,2H),5.47(br-s,2H),4.94-4.87(m,1H),3.91-3.86(m,1H),2.37-2.33(m,1H),2.21-2.12(m,2H),2.03-1.6(m,4H),1.52-1.44(m,1H).
MS ESI:m/z=402,[M+H] +.
实施例3
(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇和(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇
Figure PCTCN2020072551-appb-000028
步骤1:(R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-酮
Figure PCTCN2020072551-appb-000029
在一25毫升烧瓶中,加入(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己-1-醇(0.30g,0.68mmol),5毫升超干二氯甲烷,置于冰水浴中,然后加入Dess-Martin试剂(0.43g,1.02mmol),室温搅拌过夜,TLC监测反应,若原料还未反应完毕,补加0.5当量的Dess-Martin试剂。反应结束后,过滤除去白色不溶物,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品230mg,产率:85%。
1H NMR(400MHz,Acetone-d 6):δ8.26(s,1H),7.76-7.74(m,2H),7.46-7.42(m,2H),7.21-7.11(m,5H),6.40(br-s,2H),5.28-5.21(m,1H),3.18-3.09(m,1H),2.76-2.72(m,1H),2.57-2.49(m,1H),2.42-2.33(m,2H),2.25-2.08(m,2H),1.91-1.80(m,1H).
MS ESI:m/z=400,[M+H] +.
步骤2:(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇和(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇
Figure PCTCN2020072551-appb-000030
氮气保护下,向一充分烘干的25毫升烧瓶中,加入无水三氯化铈(0.35g,1.41mmol),5毫升超干四氢呋喃。室温下搅拌2小时后,将烧瓶放入干冰-乙醇浴中,待体系温度降至零下七十摄氏度附近,向体系中慢慢加入甲基锂的1M乙醚溶液(1.41mL,1.41mmol)。反应液在此温度下反应90分钟后,向体系中加入(R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-酮(0.12g,0.35mmol)的2毫升超干四氢呋喃溶液,并继续反应6个小时。TLC监测反应结束。加入硅胶,旋干,经硅胶柱(二氯甲烷:甲醇=100:2~100:3)纯化,依次得到实施例3A:(1S,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇50mg;实施例3B:(1R,3R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己烷-1-醇,50mg,总产率:68%。
实施例3A:
1H NMR(400MHz,Acetone-d 6):δ8.25(s,1H),7.76-7.74(m,2H),7.46-7.42(m,2H),7.21-7.16(m,3H),7.13-7.11(m,2H),6.36(br-s,2H),4.97-4.89(m,1H),3.96(br-s,1H),2.29-2.23(m,1H),2.01-1.86(m,4H),1.74-1.71(m,1H),1.62-1.56(m,2H),1.35(s,3H).
MS ESI:m/z=416.2,[M+H] +.
实施例3B:
1H NMR(400MHz,Acetone-d 6):δ8.24(s,1H),7.76-7.74(m,2H),7.46-7.42(m,2H),7.21-7.15(m,3H),7.13-7.11(m,2H),6.32(br-s,2H),5.27-5.19(m,1H),3.49(br-s,1H),2.18-2.12(m,1H),1.98-1.91(m,4H),1.72-1.69(m,2H),1.49-1.41(m,1H),1.29(s,3H).
MS ESI:m/z=416.2,[M+H] +.
实施例4
3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-(三氟甲基)环己-1-醇
Figure PCTCN2020072551-appb-000031
在一25毫升烘干的烧瓶中,加入(R)-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-酮(30mg,0.09mmol),三氟甲基三甲基硅烷(13mg,0.09mmol),2毫升四氢呋喃。室温搅拌下,向体系中加入3mg氟化铯,室温搅拌3小时后,加入2毫升4N盐酸溶液,再搅拌2小时后,加入5毫升水,二氯甲烷萃取(5mL×2),合并有机相,无水硫酸钠干燥,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品10mg,产率:21%。
1H NMR(400MHz,CDCl 3):δ8.47(s,1H),8.08(br-s,1H),7.71-7.70(m,2H),7.47-7.38(m,2H),7.21-7.08(m,5H),5.66(br-s,2H),5.47(s,1H),2.42-2.41(m,2H),2.1-2.0(m,3H),1.9-1.85(m,2H),1.75-1.72(m,1H).
MS ESI:m/z=470.17,[M+H] +.
实施例5
3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊醇
Figure PCTCN2020072551-appb-000032
3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(600mg,2.00mmol)和1,3-环戊二醇(200mg,2.00mmol),三苯基膦(924mg,3.50mmol)溶于20毫升无水四氢呋喃中,放置于冰水浴中搅拌,滴加偶氮二甲酸异丙酯(712mg,3.50mmol)。滴加完毕后,溶液在冰水浴下继续搅拌1小时,TLC显示原料消失。反应液直接浓缩后经硅胶柱(二氯甲烷:甲醇=20:1)纯化,得到产品500mg,收率:64%。
1H NMR(400MHz,DMSO-d 6):δ8.24(s,1H),7.68-7.66(m,2H),7.46-7.42(m,2H),7.21-7.12(m,5H),5.22-5.14(m,1H),4.94(d,1H),4.24-4.20(m,1H),2.43-2.36(m,1H),2.21-2.14(m,1H),2.08-2.00(m,2H),1.92-1.84(m,1H),1.82-1.74(m,1H).
MS ESI:m/z=388.1,[M+H] +.
实施例6
3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊醇
Figure PCTCN2020072551-appb-000033
步骤1:3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊酮
Figure PCTCN2020072551-appb-000034
将实施例5(500mg,1.29mmol)溶于二氯甲烷15毫升中。在冰水浴冷却下,向体系中加入Dess-Martin氧化剂(548mg,1.29mmol),搅拌2小时后,再补加Dess-Martin氧化剂(273mg,0.64mmol)。TLC监测反应,待原料消失后,向体系中加入5毫升饱和碳酸氢钠溶液淬灭反应,分液,二氯甲烷萃取(20mL×1),合并有机相,干燥,过滤,浓缩后经硅胶柱(二氯甲烷:甲醇=20:1)纯化,得到产品500mg,收率:100%。
MS ESI:m/z=386.1,[M+H] +.
步骤2:3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊醇
Figure PCTCN2020072551-appb-000035
氩气保护下,向一个25毫升两口反应瓶中加入无水三氯化铈(153mg,0.62mmol)以及10毫升超干四氢呋喃。在室温下搅拌1小时后,将反应体系置于干冰-乙醇浴中冷却。待冷却到零下75摄氏度后,向反应体系中逐滴加入甲基锂的乙醚溶液(1.33M,0.48mL,0.62mmol)。所得溶液在此温度下继续搅拌1小时后,向反应体系中逐滴加入3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊酮的四氢呋喃溶液(40mg,0.1mmol/0.5mL THF)。滴加完毕后,此温度下继续反应1.5小时,TLC显示有新点生成。用10毫升饱和氯化铵溶液淬灭反应,乙酸乙酯(20mL×1),有机相经无水硫酸钠干燥,过滤,浓缩后经硅胶柱(二氯甲烷:甲醇=20:1)纯化,得到产品6mg,收率:15%。
1H NMR(400MHz,CDCl 3):δ8.36(s,1H),7.65(m,2H),7.41-7.37(m,2H),7.19-7.14(m,3H),7.08(m,2H),5.71-5.63(m,1H),5.60(br-s,2H),2.52-2.45(m,1H),2.42-2.37(m,1H),2.28-2.11(m,4H),1.52(s,3H).
MS ESI:m/z=402.1,[M+H] +.
实施例7
3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-(三氟甲基)环戊醇
Figure PCTCN2020072551-appb-000036
3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊酮(39mg,0.10mmol)、氟化铯(0.15mg)和三氟甲基三甲基硅烷(15mg,0.10mmol)加入到反应瓶中,然后加入1毫升超干四氢呋喃。溶液由无色变成橘色,室温下搅拌3小时。加入4N盐酸水溶液(0.06mL),继续搅拌2小时。TLC和LCMS监测反应完毕。向反应液中加入2毫升水淬灭反应,乙酸乙酯萃取(5mL×3),合并有机相,用无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=20:1)纯化,得到产品10mg,收率:22%。
1H NMR(400MHz,CDCl 3):δ8.36(s,1H),7.63(m,2H),7.42-7.37(m,2H),7.20-7.17(m,1H),7.16-7.13(m,2H),7.10-7.07(m,2H),5.70(br-s,2H),5.55-5.50(m,1H),2.76-2.69(m,1H),2.56-2.49(m,1H),2.40(m,1H),2.36-2.30(m,1H),2.15-2.11(m,2H).
MS ESI:m/z=456.1,[M+H] +.
实施例8
(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇和(1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇
Figure PCTCN2020072551-appb-000037
在一个充分烘干的50毫升二口烧瓶中,加入三苯基膦(0.52g,1.98mmol),15毫升超干四氢呋喃溶剂,并置于冰乙醇浴中冷却。然后,向体系中滴入偶氮二甲酸二异丙酯(0.40g,0.39mL,1.98mmol),继续在此温下搅拌约30分钟,此时可以看到体系变为白色沉淀状。向上述反应液中加3-(4-苯氧苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(0.30g,0.99mmol)和1,4-环己二醇(顺式:反式=1:0.7,核磁含量)(114mg,0.99mmol)的10毫升四氢呋喃溶液。反应30分钟后,TLC监测反应,原料1,4-环己二醇已经消失。向反应体系中加入20毫升饱和氯化铵溶液,除去绝大部分四氢呋喃,二氯甲烷萃取(20mL×3),合并有机相,饱和食盐水(50mL×1)洗涤,无水硫酸钠干燥,过滤,加入硅胶拌样,经硅胶柱(二氯甲醇/甲醇体系)甲醇含量为1%时,得到产品实施例8A:(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇100mg,产率:26%;甲醇含量为2%时,得到产品实施例8B:(1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇30mg,产率:10%。
实施例8A:
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.67-7.63(m,2H),7.40-7.39(m,2H),7.19-7.07(m,5H),5.46(br-s,2H),4.84-4.76(m,1H),3.86-3.79(m,1H),2.28-2.05(m,6H),1.57-1.56(m,2H).
MS ESI:m/z=402,[M+H] +.
实施例8B:
1H NMR(400MHz,CDCl 3):δ8.43(s,1H),7.48-7.41(m,2H),7.40-7.39(m,2H),7.28-7.27(m,1H),7.24-7.16(m,4H),5.24(br-s,2H),4.22-4.16(m,1H),3.91-3.86(m,1H),2.45-2.35(m,2H),2.14-2.11(m,2H),1.95-1.92(m,2H),1.40-1.31(m,2H).
MS ESI:m/z=402,[M+H] +.
实施例9
(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇和(1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇
Figure PCTCN2020072551-appb-000038
步骤1:4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]环己酮-1-酮
Figure PCTCN2020072551-appb-000039
氩气保护下,在一个烘干的25毫升烧瓶中,加入化合物(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-醇(0.20g,0.45mmol),3毫升无水二氯甲烷溶剂,并将其置于冰-乙醇浴中。搅拌下,向其中滴入Dess-Martin试剂(0.23g,0.53mmol)的3毫升二氯甲烷溶液,加完之后,继续搅拌5小时。TLC监测反应结束,向体系中加入适量硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品150mg,产率:83%。
1H NMR(400MHz,CDCl 3):δ8.32(s,1H),7.59-7.57(m,2H),7.34-7.30(m,2H),7.12-7.06(m,3H),7.03-7.00(d,2H),5.53(br-s,2H),5.23-5.20(m,1H),2.62-2.49(m,6H),2.34-2.30(m,2H).
MS ESI:m/z=400,[M+H] +.
步骤2:(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇和(1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇
Figure PCTCN2020072551-appb-000040
在氩气保护下,在一个充分烘干的25毫升二口烧瓶中,加入无水三氯化铈(0.29g,1.18mmol),2毫升无水四氢呋喃溶剂,室温下搅拌1小时。将此反应液置于干冰-乙醇浴中冷却,待至零下75摄氏度左右时,向体系中逐滴加入1M甲基锂(1.18mL,1.18mmol)的乙醚溶液,并保持零下75摄氏度反应2小时后,向其中加入中间体4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]环己酮-1-酮(100mg,0.30mmol)的5毫升四氢呋喃溶液。加完之后继续在此温度下反应15分钟,撤去干冰-乙醇浴,继续反应1小时(自然升温),TLC显示反应结束。向体系中加入15毫升0.5N盐酸水溶液,二氯甲烷萃取(10mL×3),合并有机相,饱和碳酸氢钠水溶液(10mL×2)洗涤,水洗(20mL×2),饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品实施例9A:(1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇50mg,产率:42%;实施例9B:(1r,4r)-4(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-甲基环己烷-1-醇50mg,产率:42%。
实施例9A:
1H NMR(400MHz,CDCl 3):δ8.37(s,1H),7.66(d,J=8.6Hz,2H),7.39(t,J=8.0Hz,2H),7.16(dd,J=12.2Hz,8.0Hz,3H),7.08(d,J=7.7Hz,2H),5.49(br-s,2H),4.79-4.72(m,1H),2.55-2.45(m,2H),1.94-1.79(m,4H),1.75-1.67(m,2H),1.32(s,3H).
MS ESI:m/z=416,[M+H] +.
实施例9B:
1H NMR(400MHz,CDCl 3):δ8.37(s,1H),7.65(d,J=8.7Hz,2H),7.39(dd,J=8.5,7.5Hz,2H),7.21-7.12(m,3H),7.08(dd,J=8.6,1.0Hz,2H),5.53(s,2H),4.91-4.76(m,1H),2.33–2.18(m,2H),2.09-2.01(m,2H),1.92(d,J=12.8Hz,2H),1.77(dd,J=13.1,3.8Hz,2H),1.43(s,3H).
MS ESI:m/z=416,[M+H] +.
实施例10
4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]-1-环丙基环己烷-1-醇
Figure PCTCN2020072551-appb-000041
氩气保护下,在一充分烘干的50毫升二口烧瓶中,加入中间体4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基]环己-1-酮(0.30g,0.75mmol),30毫升无水四氢呋喃,并将反应瓶置于冰-乙醇浴中。待冷至负十度左右时,向体系中逐滴加入1M环丙基溴化镁的四氢呋喃溶液(7.51mL,7.51mmol)。滴加完后,撤去冰-乙醇浴,使之自然升至室温。室温下继续反应过夜。TLC监测反应结束。向体系中加入10毫升0.5N稀盐酸水溶液,二氯甲烷萃取(10mL×3),合并有机相,碳酸氢钠水溶液洗涤(10mL×3),水洗(10mL×3),饱和氯化钠溶液洗涤(20mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品250mg,产率:76%。
1H NMR(400MHz,CDCl 3):δ8.35(s,1H),7.65(m,2H),7.45-7.32(m,2H),7.19-7.04(m,5H),5.70(br-s,2H),4.98-4.84(m,0.3H),4.77(m,0.7H),2.67-2.44(m,1.6H),2.44-2.32(m,0.75H),2.18-1.53(m,6H),1.06-0.84(m,1H),0.40(d,J=6.9Hz,4H).
MS ESI:m/z=442,[M+H] +.
实施例11
(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇和(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000042
步骤1:1-溴-4-(4-氟苯氧基)苯
Figure PCTCN2020072551-appb-000043
氩气保护下,向一个充分烘干的500毫升三口烧瓶内依次加入对溴碘苯(30.00g,106.04mmol),4-氟苯酚(17.83g,159.06mmol),碳酸铯(69.10g,212.08mmol),二甲氨基乙酸(0.82g,7.95mmol),碘化亚铜(0.40g,2.12mmol)和350毫升1,4-二氧六环。升温至90摄氏度。保持该温度反应8小时后,TLC显示原料已反应完全。向体系中加入200毫升水,分液,乙酸乙酯萃取(50mL×3),合并有机相,饱和食盐水洗涤(300mL×1),无水硫酸钠干燥,过滤,加入硅胶拌样,经硅胶柱(石油醚)纯化,得到产品21.00g,产率:73%。
1H NMR(400MHz,CDCl 3):δ7.42(d,J=9.0Hz,2H),7.08-7.00(m,2H),7.00-6.93(m,2H),6.84(d,J=9.0Hz,2H).
步骤2:(4-(4-氟苯氧基)苯基)硼酸
Figure PCTCN2020072551-appb-000044
氩气保护下,向一250毫升二口烧瓶中,加入中间体1-溴-4-(4-氟苯氧基)苯(2.00g,7.49mmol),100毫升无水四氢呋喃,并将反应瓶置于干冰-乙醇浴中冷却。向体系中逐滴加入2.4M的正丁基锂的正己烷溶液(4.06mL,9.73mmol),继续搅拌40分钟后,向体系中滴入三异丙氧硼。TLC监测反应完全后,将反应体系缓慢倒入100毫升饱和氯化铵溶液中,乙酸乙酯萃取(50mL×3),合并有机相,饱和食盐水洗涤(100mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=97:3~55:45)纯化,得到产品1.50g,产率:86%。
1H NMR(400MHz,CDCl 3):δ8.16(d,J=8.6Hz,2H),7.10-6.94(m,8H).
步骤3:3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000045
在一个烘干的25毫升二口烧瓶中,加入3-碘代-1H-吡唑并[3,4-d]嘧啶-4-胺(5.00g,19.16mmol),中间体(4-(4-氟苯氧基)苯基)硼酸(8.89g,38.31mmol),和磷酸钾(12.20g, 57.47mmol)以及150毫升1,4-二氧六环和50毫升水,抽换氩气三次,然后加入四三苯基膦钯(3.32g,2.87mmol),再抽换氩气三次。升温至回流。约三小时后,TLC显示反应结束。向体系中加入150毫升水,搅拌10分钟后,过滤。粗产品用甲醇和乙酸乙酯(甲醇:乙酸乙酯=1:5,10mL)打浆,得到产品4.31g,产率:70%。
1H NMR(400MHz,DMSO-d 6):δ13.45(s,1H),8.21(s,1H),7.66(d,J=8.5Hz,2H),7.27(t,J=8.7Hz,2H),7.21-7.16(m,2H),7.13(d,J=8.5Hz,2H),6.74(br-s,2H).
MS ESI:m/z=322,[M+H] +.
步骤4:(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇和(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000046
氩气保护下,在一个充分干燥的100毫升二口烧瓶中,加入中间体3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(1.00g,3.11mmol),1,4-环己二醇(顺式:反式=1:0.7,核磁含量)(0.72g,6.22mmol),三苯基膦(1.63g,6.22mmol)和40毫升无水四氢呋喃,并将反应液置于冰乙醇浴中冷却。向反应液中慢慢滴加偶氮二甲酸二异丙酯(1.26g,6.22mmol),滴加完后反应液搅拌过夜。TLC和LCMS监测反应结束。然后加入20毫升饱和氯化铵溶液,除去大部分四氢呋喃,加入20毫升水,二氯甲烷萃取(30mL×3),合并有机相,饱和食盐水洗涤(50mL×1),无水硫酸钠干燥,过滤,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1.2~100:10)纯化,得到产品实施例11A:(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇700mg,产率:53%;实施例11B:(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇100mg,产率:8%。
实施例11A:
1H NMR(400MHz,CDCl 3):δ8.34(s,1H),7.66(d,J=8.7Hz,2H),7.10-7.06(m,6H),5.90(br-s,2H),4.83-4.78(m,1H),4.12(m,1H),2.55-2.49(m,2H),2.01(m,2H),2.42-2.41(m,2H),2.04-1.99(m,2H).
MS ESI:m/z=420,[M+H] +.
实施例11B:
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.64(d,J=8.7Hz,2H),7.12-7.05(m,6H),5.45(br-s,2H),4.82-4.76(m,1H),3.86–3.75(m,2H),2.27-2.05(m,7H).
MS ESI:m/z=420,[M+H] +.
实施例12
(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己- 1-醇和(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇
Figure PCTCN2020072551-appb-000047
步骤1:4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基环己-1-酮
Figure PCTCN2020072551-appb-000048
氩气保护下,在一干燥的50毫升的二口烧瓶中加入化合物(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(0.60g,1.43mmol),15毫升超干二氯甲烷。将反应液置于冰乙醇浴中冷却,然后逐滴加入Dess-Martin试剂(0.72g,1.69mmol)的15毫升二氯甲烷溶液。加完继续搅拌约4小时后,LCMS显示原料反应已完全。故向体系中加入10毫升饱和氯化铵溶液,用二氯甲烷萃取(15mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1.2~100:4)纯化,得到产品500mg,产率:83%。
MS ESI:m/z=418,[M+H] +.
步骤2:(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇和(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇
Figure PCTCN2020072551-appb-000049
氩气保护下,在一个充分干燥的25毫升二口烧瓶中,加入无水三氯化铈(0.24g,0.96mmol),3毫升无水四氢呋喃,室温搅拌1小时。然后将反应液置于干冰-乙醇浴中冷却至零下70摄氏度左右,向其中逐滴加入1.6M甲基锂的乙醚溶液(0.60mL,0.96mmol)。所得反应液继续在此温度下搅拌1小时。然后向反应液中滴加4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基环己-1-酮(0.10g,0.24mmol)的3毫升四氢呋喃溶液。加完后继续搅拌1.5小时后,TLC显示原料已反应完全。向体系中加入3 毫升饱和氯化铵溶液淬灭反应,除去四氢呋喃。乙酸乙酯萃取(5mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(5mL×3),无水硫酸钠干燥,过滤,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1.2~100:5~100:10)纯化,得到产品实施例12A:(1s,4s)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇25mg,产率:25%;实施例12B:(1r,4r)-4-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环己-1-醇23mg,产率:23%。
实施例12A:
1H NMR(400MHz,CDCl 3):δ8.34(s,1H),7.66-7.64(d,2H),7.12-7.05(m,6H),5.73(br-s,2H),4.79-4.71(m,1H),2.54-2.44(m,2H),1.91-1.84(m,4H),1.72-1.66(m,2H),1.32(s,3H).
MS ESI:m/z=434,[M+H] +.
实施例12B:
1H NMR(400MHz,CDCl 3):δ8.37(s,1H),7.66-7.64(d,2H),7.13-7.06(m,6H),5.58(br-s,2H),4.88-4.80(m,1H),2.30-2.20(m,2H),2.07-2.03(m,2H),1.93-1.90(m,2H),1.79-1.72(m,2H),1.43(s,3H).
MS ESI:m/z=434,[M+H] +.
实施例13
(±)顺式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇和(±)反式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000050
氩气保护下,在一干燥的100毫升三口烧瓶中,加入中间体3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(3.10g,9.65mmol),三苯基膦(2.56g,9.74mmol),抽换氩气三次,并将体系置于冰-乙醇浴中。向体系中加入1,3-环戊二醇(0.99g,9.65mmol)的10毫升超干四氢呋喃溶液,然后向体系中慢慢滴加偶氮二甲酸二异丙酯(1.95g,9.65mmol),滴加完毕,继续反应3小时。LCMS监测反应完全。将反应液倒入130毫升水中,乙酸乙酯萃取(100mL×3),合并有机相,有机相用饱和食盐水洗涤(200mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱纯化,得到实施例13A:(±)顺式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇1.80g,产率:46%;实施例13B:(±)反式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇200mg,产率:5%。
实施例13A:
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.63(m,2H),7.15-6.97(m,6H),5.56(br-s, 2H),5.47(m,1H),4.44(s,1H),2.52-2.34(m,2H),2.32-2.16(m,1H),2.20(m,1H),2.12-2.00(m,1H),1.92-1.81(m,1H).
MS ESI:m/z=406,[M+H] +.
实施例13B:
MS ESI:m/z=406,[M+H] +.
实施例14
(±)顺式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇和(±)反式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇
Figure PCTCN2020072551-appb-000051
步骤1:3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-酮
Figure PCTCN2020072551-appb-000052
氩气保护下,在一干燥的50毫升的二口烧瓶中,加入(±)顺式-3-(4-胺基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(0.50g,1.23mmol),加入15毫升超干二氯甲烷,并将反应液置于冰乙醇浴中冷却。然后向反应液中逐滴加入Dess-Martin试剂(0.62g,1.46mmol)的15毫升二氯甲烷溶液。加完继续搅拌反应,LCMS监测反应,待原料反应完全后,向体系中加入10毫升饱和氯化铵溶液,二氯甲烷萃取(15mL×3),合并有机相,饱和食盐水洗涤(30mL×1),无水硫酸钠干燥,过滤,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1.2~100:5)纯化,得到产品430mg,产率:86%。
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.63(m,2H),7.14-7.01(m,6H),5.69-5.62(m,1H),5.58(br-s,2H),3.02-2.96(m,1H),2.82-2.68(m,2H),2.60-2.54(m,2H),2.45-2.32(m,1H).
19F NMR(376MHz,CDCl 3):δ-118.79(s).
MS ESI:m/z=404,[M+H] +.
步骤2:(±)顺式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇和(±)反式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇
Figure PCTCN2020072551-appb-000053
氩气保护下,在一个充分干燥的25毫升二口烧瓶中,加入无水三氯化铈(0.49g,1.98mmol),3毫升无水四氢呋喃,室温搅拌1小时。将反应液置于干冰-乙醇浴中冷却至零下70摄氏度左右时,向其中逐滴加入1.6M甲基锂的乙醚溶液(1.24mL,1.98mmol),滴加完毕后继续在此温度下搅拌1小时。然后向反应液中滴加中间体3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-酮(0.20g,0.50mmol)的3毫升四氢呋喃溶液。加完后继续搅拌1.5小时。TLC显示原料已反应完全。向体系中加入3毫升饱和氯化铵水溶液淬灭反应,除去大部分四氢呋喃。乙酸乙酯萃取(5mL×3),合并有机相,饱和氯化钠溶液洗涤(5mL×3),无水硫酸钠干燥,过滤,加入硅胶拌样,经硅胶柱(二氯甲烷:甲醇=100:1.2~100:10)纯化,得到产品实施例14A:(±)顺式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇80mg,产率:40%;实施例14B:(±)反式-3-(4-氨基-3-(4-(4-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-1-甲基环戊-1-醇70mg,产率:35%。
实施例14A:
1H NMR(400MHz,CDCl 3):δ8.39(s,1H),7.67-7.65(m,2H),7.13-7.05(m,6H),5.64(br-s,2H),5.54-5.48(m,1H),2.49-2.41(m,1H),2.39-2.33(m,2H),2.22-2.18(m,1H),2.08-2.02(m,1H),1.80-1.69(m,1H),1.45(s,3H).
19F NMR(376MHz,CDCl 3):δ-118.82(s).
MS ESI:m/z=420,[M+H] +.
实施例14B
1H NMR(400MHz,CDCl 3):δ8.37(s,1H),7.66-7.64(m,2H),7.13-7.03(m,6H),5.71-5.63(m,1H),5.49(br-s,2H),2.52-2.37(m,1H),2.29-2.26(m,1H),2.25-2.13(m,3H),1.88-1.83(m,1H),1.52(s,3H).
19F NMR(376MHz,CDCl 3):δ-118.90(s).
MS ESI:m/z=420,[M+H] +.
实施例15
(1R,3R)-3-(4-胺基-3-(4-(3-氟苯氧基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000054
步骤1:(1R,3R)-3-(3-碘-4-((三苯基-5-磷亚烷基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基]环己基乙酸酯
Figure PCTCN2020072551-appb-000055
在冰乙醇浴冷却下,向一干燥的250毫升三口烧瓶中,加入(1R,3S)-3-羟基环己基乙酸酯(1.00g,6.32mmol),3-碘代-1H-吡唑并[3,4-d]嘧啶-4-胺(3.30g,12.64mmol),Ph 3P(4.97g,18.96mmol),100毫升超干四氢呋喃。然后向反应液中滴加DIAD(3.84g,18.96mmol)。继续在此温度下搅拌5小时,TLC显示原料反应完毕。向反应液中加入100毫升水,乙酸乙酯萃取(100mL×2),合并有机相。有机相用饱和食盐水洗涤(100mL×1),无水硫酸钠干燥。过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=5:1)纯化,得到产品1.20g,产率:60%。
MS ESI:m/z=662,[M+H] +.
步骤2:(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯
Figure PCTCN2020072551-appb-000056
氩气保护下,向一25毫升圆底烧瓶中,加入(1R,3R)-3-(3-碘-4-((三苯基-5-磷亚烷基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基]环己基乙酸酯(100mg,0.15mmol),乙酸和水各15毫升。将反应液加热回流2小时,LCMS检测反应完毕。用饱和碳酸氢钠水溶液50毫升中和反应体系,乙酸乙酯萃取(20mL×3),合并有机相。有机相用饱和氯化钠溶液洗涤(30mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品58mg,产率:99%。
1H NMR(400MHz,DMSO-d 6):δ8.20(s,1H),6.57(s,2H),5.17(s,1H),4.97-4.79(m,1H),2.28-2.17(m,1H),2.07(s,3H),1.99(d,J=13.6Hz,1H),1.89(dd,J=13.4,8.4Hz,2H),1.79(d,J=13.3Hz,1H),1.70(d,J=3.3Hz,2H),1.64-1.55(m,1H).
MS ESI:m/z=402,[M+H] +.
步骤3:1-(4-溴苯氧基)-3-氟苯
Figure PCTCN2020072551-appb-000057
在一烘干的1000毫升三口烧瓶中,加入对溴碘苯(36.80g,130.08mmol),间氟苯酚(15.31g,136.58mmol),N,N-二([1,1’-联苯]-2-基)草酰胺(2.55g,6.50mmol),碘化亚铜(1.24g,6.50mmol),磷酸钾(55.22g,260.15mmol)以及500毫升超干二甲基亚砜。抽换氩气三次,升温至100℃反应10小时后,TLC显示反应结束。将此反应液加入到1000毫升水中,乙酸乙酯萃取(1000mL×3),合并有机相,经硅胶过滤,石油醚洗涤,饱和氯化钠溶液洗涤(200mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油 醚)纯化,得到产品18.00g,产率:60%。
1H NMR(400MHz,CDCl 3):δ7.65(d,J=8.9Hz,0.4H),7.46(d,J=8.9Hz,1.6H),7.28(td,J=8.3,6.7Hz,1H),6.92(d,J=8.9Hz,1.6H),6.85-6.74(m,2.4H),6.70(dt,J=10.1,2.3Hz,1H).
步骤4:(4-(3-氟苯氧基)苯基)硼酸
Figure PCTCN2020072551-appb-000058
氩气保护下,向一充分烘干的250毫升三口烧瓶中,加入100毫升超干四氢呋喃,1-(4-溴苯氧基)-3-氟苯(2.00g,7.49mmol),并将反应液置于干冰-乙醇浴冷却至零下70摄氏度。向反应液中缓慢滴加正丁基锂乙醚溶液(1.6M,9.73mmol),加完之后,继续保持在此温度反应2小时。然后向反应液中加入三异丙基氧硼(1.69g,8.99mmol)。反应5小时后,TLC板显示原料几乎全部转化为产物。向反应液中加入100毫升1N稀盐酸,二氯甲烷萃取(100mL×3),合并有机相,饱和食盐水洗涤(50mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=3:1)纯化,得到产品700mg,产率:50%。
1H NMR(400MHz,CDCl 3):δ8.21(d,J=8.6Hz,2H),7.33(td,J=8.3,6.7Hz,1H),7.13(d,J=8.6Hz,2H),6.87(dtd,J=7.6,5.0,2.5Hz,2H),6.80(dt,J=10.0,2.3Hz,1H).
步骤5:(1R,3R)-3-(4-胺基-3-(4-(3-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯
Figure PCTCN2020072551-appb-000059
在一充分烘干的25毫升二口烧瓶中,依次加入(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(0.10g,0.25mmol),(4-(3-氟苯氧基)苯基)硼酸(0.12g,0.50mmol)和磷酸钾(0.16g,0.75mmol),10毫升1,4-二氧六环和5毫升水。将反应液置换氩气三次后,升温至回流,向体系中迅速加入四三苯基膦钯(0.10g,0.05mmol)。反应2个小时,LCMS显示反应结束。将反应液冷却,向体系中加入10毫升水,二氯甲烷萃取(8mL×3),合并有机相,饱和食盐水洗涤(8mL×3),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品50mg,产率:40%。
MS ESI:m/z=462,[M+H] +.
步骤6:(1R,3R)-3-(4-胺基-3-(4-(3-氟苯氧基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000060
在一25毫升烧瓶中,加入(1R,3R)-3-(4-胺基-3-(3-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯(50mg,0.11mmol),LiOH(13mg,0.54mmol)以及5毫升甲醇。室温下搅拌过夜,LCMS监测反应结束。过滤除去LiOH,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品48mg,产率:99%。
1H NMR(400MHz,CDCl 3):δ8.30(s,1H),7.62(d,J=8.6Hz,2H),7.25(td,J=8.3,6.7Hz,1H),7.11(d,J=8.6Hz,2H),6.79(ddd,J=8.3,6.3,2.2Hz,2H),6.70(dt,J=10.1,2.3Hz,1H),5.54(br-s,2H),5.28-5.08(m,1H),4.34(m,1H),2.37-2.23(m,1H),2.18-1.85(m,4H),1.76-1.66(m,2H),1.61-1.54(m,1H).
MS ESI:m/z=420,[M+H] +.
实施例16
(1R,3R)-3-(4-胺基-3-(2-氟-4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000061
步骤1:1-溴-2-氟-4-苯氧基苯
Figure PCTCN2020072551-appb-000062
在一个500毫升三口烧瓶中,加入4-溴-3-氟苯酚(10.00g,52.36mmol),苯硼酸(12.77g,104.71mmol),三乙胺(10.60g,104.71mmol),无水醋酸铜(4.754g,52.356mmol)和20g
Figure PCTCN2020072551-appb-000063
分子筛,300mL二氯甲烷。所得反应液置于氧气氛围中反应过夜,TLC监测反应结束。反应液经硅藻土过滤,用去500毫升二氯甲烷洗涤,浓缩,经硅胶柱(石油醚)纯化,得到产品6.00g,产率:50%。
1H NMR(400MHz,CDCl 3):δ7.45(dd,J=8.7,8.0Hz,1H),7.38(dd,J=8.5,7.5Hz,2H),7.18(t,J=7.4Hz,1H),7.03(dd,J=8.6,1.0Hz,2H),6.76(dd,J=9.8,2.7Hz,1H),6.69(ddd,J=8.8,2.7,1.1Hz,1H).
步骤2:(2-氟-4-苯氧基苯基)硼酸
Figure PCTCN2020072551-appb-000064
在一充分干燥的100毫升烧瓶中,加入1-(4-溴苯氧基)-3-氟苯(1.00g,3.74mmol),50毫升超干四氢呋喃,并将反应液置于干冰-乙醇浴冷却至零下70摄氏度。向反应液中缓缓滴加正丁基锂的正己烷溶液(1.6M,4.87mmol)。滴加完后继续保持该反应下反应2小时后,向反应液中加入三异丙氧硼(0.85g,4.49mmol)。TLC监测反应结束,向体系中缓缓加入100毫升0.5N的盐酸水溶液,二氯甲烷萃取(30mL×3)合并有机相,有机相用饱和食盐水(50mL×1)洗涤,无水硫酸钠干燥,过滤,浓缩,粗产品直接投下步。
步骤3:(1R,3R)-3-(4-胺基-3-(2-氟-4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯
Figure PCTCN2020072551-appb-000065
在一25毫升二口烧瓶中加入(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(0.25g,0.62mmol),(2-氟-4-苯氧基苯基)硼酸(0.19g,0.81mmol)和磷酸钾(0.30g,1.87mmol),10毫升1,4-二氧六环和5毫升水。置换氩气三次后,向体系中加入四三苯基膦钯(144mg,0.13mmol),并升温至回流。反应2小时后结束。向反应体系中加入15毫升水,二氯甲烷萃取(20mL×3),合并有机相,饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品150mg,产率:52%。
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.52(t,J=8.5Hz,1H),7.42(dd,J=8.4,7.6Hz,2H),7.22(t,J=7.4Hz,1H),7.10(d,J=7.6Hz,2H),6.94(dd,J=8.5,2.3Hz,1H),6.86(dd,J=11.2,2.4Hz,1H),5.35(br-s,3H),5.15(ddd,J=16.0,11.3,4.4Hz,1H),2.51-2.40(m,1H),2.22(d,J=13.7Hz,1H),2.13(s,3H),2.11-2.08(m,1H),2.08-1.99(m,1H),1.99-1.71(m,4H).
MS ESI:m/z=462,[M+H] +.
步骤4:(1R,3R)-3-(4-胺基-3-(2-氟-4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000066
在一25毫升烧瓶中,加入(1R,3R)-3-(4-胺基-3-(2-氟-4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯(100mg,0.22mmol),LiOH(20mg,0.87mmol)以及5毫升甲醇,室温搅拌过夜。TLC监测反应完毕后,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品80mg,产率:80%。
1H NMR(400MHz,CDCl 3):δ8.33(s,1H),7.51(t,J=8.5Hz,1H),7.41(t,J=7.9Hz,2H),7.21(t,J=7.4Hz,1H),7.09(d,J=7.9Hz,2H),6.92(dd,J=8.5,2.0Hz,1H),6.88-6.80(m,1H),5.60(br-s,2H),5.33-5.19(m,1H),4.39(m,1H),2.35-2.28(m,1H),2.17-2.12(m,1H),2.10-2.03(m,2H),2.03-1.93(m,1H),1.89-1.70(m,2H),1.65-1.58(m,1H).
MS ESI:m/z=420,[M+H] +.
实施例17
(1R,3R)-3-(4-氨基-3-(4-(2-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000067
步骤1:1-(4-溴苯氧基)-2-氟苯
Figure PCTCN2020072551-appb-000068
在一250毫升单口烧瓶中,加入邻氟苯酚(2.00g,17.84mmol),对溴苯硼酸(7.17g,35.68mmol),醋酸铜(4.75g,17.84mmol)和150毫升超干二氯甲烷和20克4A分子筛,并将反应液置于氧气氛围下。在室温搅拌下,向反应体系中逐滴加入三乙胺(3.61g,35.68mmol)。反应约10小时后,TLC显示反应结束。将反应液直接硅藻土过滤,150毫升石油醚洗涤,浓缩,经硅胶柱(石油醚)纯化,得到产品2.30g,产率:48%。
1H NMR(400MHz,CDCl 3):δ7.41(d,J=8.9Hz,2H),7.22-7.01(m,4H),6.85(d,J=8.9Hz,2H).
步骤2:
Figure PCTCN2020072551-appb-000069
氩气保护下,向一干燥的50毫升二口烧瓶中,加入1-(4-溴苯氧基)-2-氟苯(0.50g,1.87mmol)以及20毫升超干四氢呋喃。所得反应液经干冰-乙醇浴冷却后,向其中逐滴加入正丁基锂的正己烷溶液(1.6M,1.52mL,2.43mmol)。滴加完毕后继续搅拌30分钟。然后慢慢向反应液中加入三异丙氧基硼酸酯(0.42g,2.25mmol)。继续反应2小时,TLC显示反应结束。向反应液中加入15毫升饱和氯化铵水溶液,乙酸乙酯萃取(15mL×3),合并有机相,饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=3:1)纯化,得到产品200mg,产率:46%。
步骤3:(1R,3R)-3-(4-氨基-3-(4-(2-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸 环己酯
Figure PCTCN2020072551-appb-000070
氩气保护下,向一25毫升二口烧瓶中加入(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(0.50g,1.25mmol),(4-(2-氟苯氧基)苯基)硼酸(0.38g,1.62mmol),磷酸钾(0.79g,3.74mmol),四三苯基膦钯(0.29g,0.25mmol),20毫升1,4-二氧六环和10毫升水。所得反应液加热至回流反应1.5小时,TLC显示反应已完毕。向反应体系中加入15毫升水,二氯甲烷萃取(10mL×3),合并有机相,饱和食盐水(20mL×1)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1过出)纯化,得到产品390mg,产率:69%。
MS ESI:m/z=462,[M+H] +.
步骤4:(1R,3R)-3-(4-氨基-3-(4-(2-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000071
在一25毫升单口烧瓶中,加入(1R,3R)-3-(4-氨基-3-(4-(2-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯(0.30g,0.65mmol),氢氧化锂(0.05g,1.95mmol),3毫升甲醇。反应液室温搅拌过夜。TLC显示反应完毕。向反应体系中加入5毫升水,二氯甲烷萃取(5mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:2)纯化,得到产品250mg,产率:90%。
1H NMR(400MHz,CDCl 3):δ8.36(s,1H),7.64(d,J=8.7Hz,1H),7.40-7.32(m,0.5H),7.23(m,1H),7.21-7.14(m,3H),7.13-7.10(m,2H),7.05(d,J=7.7Hz,0.5H),5.59(br-s,2H),5.29-5.22(m,1H),4.40(m,1H),2.39-2.32(m,1H),2.21-1.97(m,4H),1.81-1.73(m,2H),1.68-1.61(m,1H).
19F NMR(376MHz,CDCl 3):δ-130.18(s).
MS ESI:m/z=420,[M+H] +.
实施例18
(1R,3R)-3-(4-氨基-3-(4-(2,6-二氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000072
步骤1:2-(4-溴苯氧基)-1,3-二氟苯
Figure PCTCN2020072551-appb-000073
在一250毫升烧瓶中,加入2,6-二氟苯酚(2.00g,15.37mmol),对溴苯硼酸(6.18g,30.75mmol),20克
Figure PCTCN2020072551-appb-000074
分子筛,醋酸铜(4.75g,15.37mmol)和150毫升超干二氯甲烷,并将反应液置于氧气氛围下。室温搅拌下,向上述反应液中逐滴加入三乙胺(3.11g,30.75mmol)。继续在室温反应3小时,TLC显示反应结束。反应液经硅藻土过滤,浓缩,经硅胶柱(石油醚)纯化,得到产品2.40g,产率:55%。
步骤2:(4-(2,6-二氟苯氧基)苯基)硼酸
Figure PCTCN2020072551-appb-000075
在一干燥的25毫升二口烧瓶中,加入2-(4-溴苯氧基)-1,3-二氟苯(0.50g,1.75mmol),15毫升超干四氢呋喃,并置于干冰-乙醇浴中冷至零下70摄氏度。向反应体系中逐滴加入正丁基锂的正己烷溶液(1.6M,1.43mL,2.28mmol)。滴加完毕后继续搅拌30分钟,然后向反应体系中加入三异丙氧基硼酸酯(0.40g,2.11mmol)。继续反应约2小时后,TLC显示反应完全后,向反应体系中加入15毫升饱和氯化铵溶液,用乙酸乙酯萃取(15mL×3),合并有机相,并用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=3:1)纯化,得到产品350mg,产率:80%。
1H NMR(400MHz,CDCl 3):δ7.40(d,J=9.1Hz,2H),7.21-7.10(m,1H),7.08-6.95(m,2H),6.83(d,J=9.0Hz,2H).
步骤3:(1R,3R)-3-(4-氨基-3-(4-(2,6-二氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯
Figure PCTCN2020072551-appb-000076
在氩气保护下,向一个50毫升的二口烧瓶中,加入(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(0.25g,0.62mmol),(4-(2,6-二氟苯氧基)苯基)硼酸(0.20g,0.81mmol),磷酸钾(0.40g,1.87mmol),四三苯基膦钯(144mg,0.13mmol),20毫升1,4-二氧六环和10毫升水。将反应液温度升至100摄氏度,反应1.5小时。LCMS显示反应结束。向反应体系中加入15毫升0.1N盐酸水溶液,乙酸乙酯洗涤(15mL×3),合并有机相,并用饱和氯化钠溶液洗涤(15mL×3),无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品240mg,产率:80%。
MS ESI:m/z=480,[M+H] +.
步骤4:(1R,3R)-3-(4-氨基-3-(4-(2,6-二氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000077
在一25毫升单口烧瓶中,加入(1R,3R)-3-(4-氨基-3-(4-(2,6-二氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯(0.20g,0.42mmol),LiOH(30mg,1.25mmol),3毫升甲醇。反应液在室温搅拌过夜。TLC监测反应结束。向反应瓶中加入10毫升水,乙酸乙酯萃取(10mL×3),合并有机相,饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品170mg,产率:93%。
1H NMR(400MHz,CDCl 3):δ8.37(d,J=1.5Hz,1H),7.63(d,J=8.6Hz,1H),7.49-7.38(m,1H),7.34-7.29(m,1H),7.23-7.14(m,1H),7.11-6.99(m,3H),5.51(br-s,1H),5.35(br-s,1H),5.31-5.20(m,1H),4.40(s,1H),2.50-2.24(m,1H),2.14-1.97(m,4H),1.82(m,2H),1.68-1.54(m,1H).
MS ESI:m/z=438,[M+H] +.
实施例19
(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000078
步骤1:1-(4-碘苯氧基)-2-氟-3-甲氧基苯
Figure PCTCN2020072551-appb-000079
在一干燥的100毫升单口烧瓶中,加入2-氟-3-甲氧基苯酚(1.00g,7.04mmol),4-碘苯硼酸(3.49g,14.07mmol),醋酸铜(4.75g,7.04mmol),4A分子筛10克和超干二氯甲烷50毫升,逐滴加入三乙胺(1.42g,14.07mmol)。将上述反应液置于氧气氛围下,室温搅拌过夜。TLC监测反应结束。反应液经硅藻土过滤,经硅胶柱(石油醚:乙酸乙酯=9:1)纯化,得到产品1.50g,产率:62%。
1H NMR(400MHz,CDCl 3):δ7.59(d,J=8.9Hz,2H),7.02(td,J=8.4,2.2Hz,1H),6.80(dd,J=11.5,4.3Hz,1H),6.75(d,J=8.8Hz,2H),6.64(ddd,J=8.3,6.9,1.4Hz,1H),3.92(s,3H).
步骤2:2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷
Figure PCTCN2020072551-appb-000080
氮气保护下,在一干燥的25毫升二口烧瓶中,依次加入PdCl 2(dppf)(6mg,0.01mmol),醋酸钾(85mg,0.87mmol)和频哪硼酸酯(148mg,0.58mmol),1-(4-碘苯氧基)-2-氟-3-甲氧基苯(100mg,0.29mmol)和10毫升DMSO。将反应液升温至85摄氏度并保持此温度反应约1.5小时后,TLC显示反应结束。将反应液冷却至室温,加入15毫升水,二氯甲烷萃取(10mL×3),合并有机相,饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=3:1)纯化,得到产品44mg,产率:45%。
1H NMR(400MHz,CDCl 3):δ7.81-7.70(m,2H),7.01(td,J=8.4,2.2Hz,1H),6.96(d,J=8.5Hz,2H),6.81-6.76(m,1H),6.66(ddd,J=8.3,6.9,1.4Hz,1H),3.92(s,3H),1.33(s,12H).
步骤3:(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯
Figure PCTCN2020072551-appb-000081
在一50毫升二口烧瓶中,加入(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(0.30g,0.75mmol),2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(0.30g,0.90mmol),碳酸钾(0.40g,2.24mmol),四三苯基膦钯(173mg,0.15mmol)。抽换氩气三次后,向反应体系中加入1,4-二氧六环20毫升和水10毫升,并升温至回流。继续反应1小时后,LCMS监测反应结束。将反应液冷却至室温,加入10毫升水,二氯甲烷萃取(10mL×3),合并有机相,饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品250mg,产率:70%。
MS ESI:m/z=492,[M+H] +.
步骤4:(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000082
在一25毫升单口烧瓶中,加入(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯(0.25g,0.51mmol),氢氧化锂(37mg,1.53mmol)和5毫升甲醇。所得反应液室温下搅拌过夜。TLC监测反应结束。向反应体系中加入5毫升水,二氯甲烷萃取(5mL×3),合并有机相,饱和食盐水(10mL×1)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品200mg,产率88%。
1H NMR(400MHz,CDCl 3):δ8.35(s,1H),7.63(d,J=8.7Hz,2H),7.13(d,J=8.6Hz,2H),7.07(td,J=8.4,2.1Hz,1H),6.89-6.81(m,1H),6.73(ddd,J=8.3,6.9,1.5Hz,1H),5.60(br-s,2H),5.26(m,1H),4.44-4.36(m,1H),3.94(s,3H),2.52-2.24(m,1H),2.29-1.95(m,5H),1.84-1.80(m,1H),1.65-1.60(m,1H).
19F NMR(376MHz,CDCl 3):δ-152.86(s).
MS ESI:m/z=450,[M+H] +.
实施例20
(1s,4s)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000083
步骤1:(1s,4s)-4-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000084
氩气保护下,向一干燥的500毫升二口烧瓶中,加入3-碘代-1H-吡唑并[3,4-d]嘧啶-4-胺(5.00g,19.16mmol),1,4-环己二醇(顺式:反式=1:0.7,核磁含量)(4.45g,38.3mmol),三苯基膦(10.05g,38.31mmol)以及超干四氢呋喃200毫升。将所得反应液置于冰-乙醇浴中冷却后,向其中逐滴加入偶氮二甲酸二异丙酯(7.75g,38.31mmol),加完后,继续反应5小时。LCMS监测完全反应后,过滤反应液中淡黄色固体,得到4.50g。所得母液浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品0.80g,总产率:73%。
MS ESI:m/z=360,[M+H] +.
步骤2:(1s,4s)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000085
在一50毫升二口烧瓶中,加入(1s,4s)-4-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(80mg,0.22mmol),2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(97mg,0.29mmol),碳酸钾(118mg,0.67mmol),四三苯基膦钯(51mg,0.05mmol)。抽换氩气三次后,再依次加入1,4-二氧六环5毫升和水2.5毫升,并升温至回流。反应液反应1小时后,LCMS监测反应结束。将反应液冷却至室温,向其中加入5毫升水,二氯甲烷萃取(5mL×3),合并有机相,饱和食盐水(5mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品65mg,产率:65%。
1H NMR(400MHz,CDCl 3):δ8.37(s,1H),7.63(d,J=8.7Hz,2H),7.13(d,J=8.6Hz,2H),7.07(td,J=8.4,2.1Hz,1H),6.84(t,J=7.2Hz,1H),6.77-6.69(m,1H),5.44(br-s,2H),4.87-4.74(m,1H),3.94(s,3H),3.82(m,1H),2.30-1.96(m,6H),1.55(m,2H).
19F NMR(376MHz,CDCl 3):δ-152.83(s).
MS ESI:m/z=450,[M+H] +.
实施例21
(1r,4r)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000086
步骤1:(1R,4R)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基4-硝基苯甲酸酯
Figure PCTCN2020072551-appb-000087
在一个烘干的25毫升二口烧瓶中,加入(1s,4s)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(80mg,0.18mmol),4-硝基苯甲酸(59mg,0.36mmol),三苯基膦(93mg,0.36mmol)和5毫升超干四氢呋喃,并将反应液置于冰-乙醇浴中冷却。然后慢慢滴加偶氮二甲酸二异丙酯(72mg,0.36mmol)。滴加完,继续反应3小时。LCMS显示反应结束,向反应液中加入5毫升水,二氯甲烷萃取(5mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品80mg,产率:78%。
MS ESI:m/z=599,[M+H] +.
步骤2:(1R,4R)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000088
在一25毫升单口烧瓶中,加入(1r,4r)-4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)- 1H-吡唑并[3,4-d]嘧啶-1-基)环己基4-硝基苯甲酸酯(80mg,0.13mmol),LiOH(13mg,0.54mmol),甲醇5毫升。反应液在室温下反应3小时,TLC和LCMS监测反应结束。向反应液中加入5毫升水,二氯甲烷萃取(5mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(展开剂:二氯甲烷:甲醇=100:1-100:2)纯化,得到产品50mg,产率:83%。
1H NMR(400MHz,CDCl 3):δ8.29(s,1H),7.57(d,J=8.5Hz,2H),7.05(d,J=8.5Hz,2H),6.99(td,J=8.4,2.0Hz,1H),6.76(t,J=7.9Hz,1H),6.66(dd,J=8.2,7.0Hz,1H),5.52(br-s,2H),4.76-4.70(m,1H),4.07-4.05(m,1H),3.86(s,3H),2.45(qd,J=12.8,3.5Hz,2H),1.94(d,J=9.8Hz,2H),1.81-1.76(m,2H),1.74-1.68(m,2H).
MS ESI:m/z=450,[M+H] +.
实施例22
3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000089
步骤1:3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000090
氩气保护下,向一干燥的500毫升二口烧瓶中,加入3-碘代-1H-吡唑并[3,4-d]嘧啶-4-胺(5.00g,19.16mmol),1,3-环戊二醇(顺式和反式的混合物)(3.91g,38.10mmol),三苯基膦(10.05g,38.31mmol)和超干四氢呋喃200毫升。将上述反应液置于冰-乙醇浴中冷却后,向其中逐滴加入偶氮二甲酸二异丙酯(7.75g,38.31mmol),滴加完后,继续反应5小时。LCMS监测反应结束。将反应液中产生的淡黄色沉淀过滤,得到3.80g。滤液浓缩,经硅胶柱(二氯甲烷:甲醇=100:1~100:2)纯化,得到产品200mg,总产率:60%。
MS ESI:m/z=346,[M+H] +.
步骤2:3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000091
氩气保护下,向一50毫升二口烧瓶中依次加入3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(0.25g,0.72mmol),2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(0.32g,0.94mmol),碳酸钾(0.30g,2.17mmol),四三苯基膦钯(0.17g,0.15mmol),1,4-二氧六环20毫升和水10毫升。将所得反应液升温至回流并反应1小时。LCMS监测反应结束。将反应液冷却至室温,向其中加入10毫升水,二氯甲烷萃取(10mL×3),合并有机相,饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品200mg,产率:63%。
1H NMR(400MHz,CDCl 3):δ8.38(s,1H),7.62(d,J=8.8Hz,2H),7.12(d,J=8.6Hz,2H),7.08(td,J=8.4,2.1Hz,1H),6.84(ddd,J=8.3,6.9,1.4Hz,1H),6.74(ddd,J=8.3,6.9,1.4Hz,1H),5.75(d,J=10.2Hz,1H),5.55(br-s,2H),5.46(dd,J=14.2,8.4Hz,1H),4.46-4.45(m,1H),3.94(s,3H),2.50-2.35(m,2H),2.32-2.23(m,1H),2.24-2.17(m,1H),2.10-2.01(m,1H),1.94-1.78(m,1H).
19F NMR(376MHz,CDCl 3):δ-152.82(s).
MS ESI:m/z=436,[M+H] +.
实施例23
(±)反式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000092
步骤1:3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊基4-硝基苯甲酸酯
Figure PCTCN2020072551-appb-000093
在一个烘干的25毫升二口烧瓶中,加入(±)顺式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(50mg,0.12mmol),4-硝基苯甲酸(38mg,0.23mmol),三苯基膦(60mg,0.23mmol)以及5毫升超干四氢呋喃。将反应液置于冰-乙醇浴中冷却。然后慢慢滴加偶氮二甲酸二异丙酯(46mg,0.23mmol)。滴加完毕后,继续反应3小时。LCMS显示反应结束。向反应液中加入5毫升水,二氯甲烷萃取(5mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品56mg,产率:80%。
MS ESI:m/z=585,[M+H] +.
步骤2:(±)反式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000094
在一25毫升单口烧瓶中,加入3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊基4-硝基苯甲酸酯(56mg,0.10mmol),LiOH(7mg,0.29mmol),甲醇3毫升。所得反应液在室温反应3小时。TLC和LCMS检测反应结束。向反应液中加入3毫升水,二氯甲烷萃取(3mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1-100:2)纯化,得到产品30mg,产率:90%。
1H NMR(400MHz,CDCl 3):δ8.34(s,1H),7.62(d,J=8.6Hz,2H),7.12(d,J=8.6Hz,2H),7.07(td,J=8.4,2.0Hz,1H),6.84(t,J=7.4Hz,1H),6.74(t,J=7.6Hz,1H),5.82(br-s,2H),5.65–5.57(m,1H),4.76–4.63(m,1H),3.94(s,3H),2.56-2.09(m,6H).
19F NMR(376MHz,CDCl 3):δ-152.83(s).
MS ESI:m/z=436,[M+H] +.
实施例24
5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)四氢-2H-吡喃-3,4-二醇
Figure PCTCN2020072551-appb-000095
步骤1:2-(烯丙氧基)乙醛
Figure PCTCN2020072551-appb-000096
向一100毫升烧瓶中加入3-(烯丙氧基)丙烷-1,2-二醇(1.00g,7.57mmol),二氯甲烷30毫升,水15毫升。然后向反应液中加入高碘酸钠(1.94g,9.08mmol),室温搅拌。 TLC监测反应结束。加入20毫升水,分液,二氯甲烷萃取(50mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩后直接投下一步。
步骤2:1-(烯丙氧基)丁-3-烯-2-醇
Figure PCTCN2020072551-appb-000097
在一充分烘干的2000毫升三口烧瓶内,加入2-(烯丙氧基)乙醛(15.00g,粗品),超干THF 1000毫升,并将反应液置于冰-乙醇浴内冷却。在此温度下,向反应液中滴加乙烯基溴化镁的四氢呋喃溶液(1M,179.78mL,179.78mmol),1小时后滴完。继续搅拌4小时,TLC显示反应完毕。向反应体系中加入100毫升水淬灭反应,旋去大部分四氢呋喃。向反应瓶中加入300毫升二氯甲烷,100毫升水,分液,二氯甲烷萃取(150mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(石油醚:乙酸乙酯=7:3)纯化,得到产品7.20g,产率:35%。
1H NMR(400MHz,CDCl 3):δ5.99-5.74(m,2H),5.41-5.24(m,2H),5.20(dd,J=10.7,1.3Hz,2H),4.39-4.27(m,1H),4.04(dt,J=5.7,1.3Hz,2H),3.51(dd,J=9.7,3.4Hz,1H),3.34(dd,J=9.6,8.0Hz,1H),2.49(d,J=3.4Hz,1H).
步骤3:3,6-二氢-2H-吡喃-3-醇
Figure PCTCN2020072551-appb-000098
氩气保护下,向一干燥的500毫升三口烧瓶中,加入1-(烯丙氧基)丁-3-烯-2-醇(7.20g,56.18mmol),350毫升超干二氯甲烷,Grubbs二代催化剂(1.19g,1.40mmol)。反应液于室温下反应9小时。TLC监测反应结束,反应液直接经硅胶柱(石油醚:乙酸乙酯=4:1)纯化,得到产品5.00g,产率:90%。
1H NMR(400MHz,CDCl 3):δ6.04-5.96(m,1H),5.96-5.90(m,1H),4.28-4.04(m,2H),3.98(dd,J=5.5,2.5Hz,1H),3.85(ddd,J=11.8,2.8,0.7Hz,1H),3.75(dd,J=11.8,3.0Hz,1H),1.97(d,J=9.3Hz,1H).
步骤4:1-(3,6-二氢-2H-吡喃-3-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000099
在一烘干的25毫升二口烧瓶内,加入3,6-二氢-2H-吡喃-3-醇(0.14g,1.40mmol),3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(0.42g,1.40mmol),三苯基膦(0.73g,2.80mmol),10毫升超干四氢呋喃,并将反应液置于冰-乙醇浴内冷却。然后缓慢向瓶中滴加偶氮二甲酸二异丙酯(0.57g,2.80mmol)。反应1小时后,LCMS监测反应结束。向反应瓶中加入10毫升水,二氯甲烷萃取(20mL×3),合并有机相,饱和食盐水(30mL×1)洗涤,无水硫酸钠干燥,过 滤,浓缩,经硅胶柱(二氯甲烷:甲醇=40:1)纯化,得到产品50mg,产率:95%。
1H NMR(400MHz,CDCl 3):δ8.39(s,1H),7.64(d,J=8.7Hz,2H),7.39(dd,J=8.5,7.5Hz,2H),7.22-7.10(m,3H),7.09-7.04(m,2H),6.13(ddd,J=10.4,4.8,2.2Hz,1H),6.05(dd,J=10.4,2.1Hz,1H),5.63(ddd,J=10.1,5.1,2.5Hz,1H),5.52(br-s,2H),4.36(ddd,J=16.8,5.2,2.4Hz,1H),4.29-4.15(m,2H),4.08(dd,J=11.0,7.4Hz,1H).
MS ESI:m/z=386,[M+H] +.
步骤5:5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)四氢-2H-吡喃-3,4-二醇
Figure PCTCN2020072551-appb-000100
向一个25毫升二口烧瓶中加入5毫升去离子水,5毫升叔丁醇,K 3Fe(CN) 6(0.13g,0.39mmol),K 2CO 3(54mg,0.39mmol),K 2OsO 2(OH) 4(24mg,0.07mmol)和1-(3,6-二氢-2H-吡喃-3-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(50mg,0.13mmol),剧烈搅拌。反应4小时后经LCMS监测反应结束。向反应体系中加入10毫升乙酸乙酯,过滤,向滤液中加入15毫升水,分液,乙酸乙酯萃取(15mL×2),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=20:1)纯化,得到产品18mg,产率:36%。
1H NMR(400MHz,DMSO-d 6):δ8.24(s,1H),7.66(d,J=8.7Hz,2H),7.51-7.37(m,2H),7.15(ddd,J=11.1,9.7,4.2Hz,5H),5.00(td,J=10.6,4.8Hz,1H),4.90(d,J=3.8Hz,1H),4.85(d,J=6.6Hz,1H),4.33-4.20(m,1H),3.86(d,J=8.5Hz,3H),3.65(t,J=11.0Hz,1H),3.57(d,J=11.8Hz,1H).
MS ESI:m/z=420,[M+H] +.
实施例25
3-(4-氨基-3-(4-(3-氟苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇
Figure PCTCN2020072551-appb-000101
氩气保护下,向一25毫升二口烧瓶中依次加入3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环戊烷-1-醇(0.50g,1.45mmol),(4-(3-氟苯氧基)苯基)硼酸(0.40g,1.74mmol),磷酸钾(0.92g,4.35mmol),四三苯基膦钯(72mg,0.22mmol),10毫升1,4-二氧六环和5毫升水。反应液升温至回流并反应1小时,LCMS显示反应结束。向反应体系中加入10毫升水,二氯甲烷萃取(10mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩,经硅胶柱(二氯甲烷:甲醇=100:1)纯化,得到产品360mg,产率:62%。
1H NMR(400MHz,CDCl 3):δ8.39(s,1H),7.68(d,J=8.7Hz,2H),7.33(td,J=8.3,6.7Hz, 1H),7.21-7.14(m,2H),6.87(td,J=8.4,2.4Hz,2H),6.79(dt,J=10.0,2.4Hz,1H),5.75(d,J=10.2Hz,1H),5.56(s,2H),5.47(ddd,J=16.4,7.3,3.1Hz,1H),4.52-4.39(m,1H),2.58-2.35(m,2H),2.32-2.19(m,2H),2.09-2.04(m,1H),1.95-1.82(m,1H).
19F NMR(376MHz,CDCl 3):δ-110.43(s).
MS ESI:m/z=406,[M+H] +.
实施例26
顺式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇
Figure PCTCN2020072551-appb-000102
步骤1:3-羟基乙酸环己酯
Figure PCTCN2020072551-appb-000103
将4-氯苯基乙酸酯(84.2g,495mmol)溶于无水甲苯(800mL)中,置换氮气后,向其中加入环己烷-1,3-二醇(38.3g,330mmol)和Candida Antarctica lipase B(CALB)(7.6g),再次置换氮气。反应液室温搅拌16小时,抽滤除去固体,滤液减压浓缩得到残余物,残余物经柱层析(乙酸乙酯:石油醚=0-100%)纯化,得到产品47.2g,产率:91%。
1H NMR(400MHz,CDCl 3)5.14-4.72(m,1H),4.03-3.67(m,1H),2.25-2.20(m,1H),2.02(s,3H),1.91-1.59(m,4H),1.44-1.25(m,3H).
步骤2:3-(3-碘-4-((三苯基亚膦基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯
Figure PCTCN2020072551-appb-000104
在氮气保护下,将3-碘-1H-吡唑并[3,4-d]嘧啶-4-胺(22.8g,27.5mmol),3-羟基乙酸环己酯(15.2g,96.2mmol)和三苯基膦(45.8g,174.9mmol)溶于无水四氢呋喃(300mL)中,冰浴下滴加偶氮二甲酸二异丙酯(35.3g,174.9mmol),控制滴加速度维持反应体系内温不高于10摄氏度。滴加完毕后,反应液室温搅拌16小时。反应液减压浓缩得到残余物,残余物经柱层析(石油醚:乙酸乙酯=0-100%)纯化,得到粗产品35.8g。
步骤3:3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯
Figure PCTCN2020072551-appb-000105
将3-(3-碘-4-((三苯基亚膦基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙酸环己酯(74.1g,27.4mmol)溶于100毫升乙酸,并加入100毫升水。反应液130摄氏度回流1小时。反应液浓缩除去大部分乙酸,加100毫升水稀释,并用80毫升二氯甲烷萃取2次。有机相合并后,无水硫酸钠干燥,过滤,减压浓缩得到残余物,残留物用100毫升甲苯带剩余溶残乙酸3次得到粗产品44.5g。
MS ESI:m/z=402,[M+H] +
步骤4:3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000106
将(1R)-3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯粗品(44.5g,27.4mmol)溶于150毫升甲醇和150毫升四氢呋喃的混合溶液中,加入一水合氢氧化锂(13.9g,331mmol)。反应液于室温搅拌16小时。抽滤除去固体,滤液浓缩,硅胶柱纯化(二氯甲烷:甲醇=0-100%)得到产品8.4g,三步产率:27%。
MS ESI:m/z=360,[M+H] +
步骤5:3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000107
将3-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇(8.4g,23.4mmol),2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(9.7g,28.8mmol),四三苯基膦钯(2.6g,2.3mmol)和碳酸钾(8.1g,58.5mmol)悬浮150毫升1,4-二氧六环和80毫升水的混合溶剂中,氮气置换三次,120度加热16小时,反应液变澄清。反应液浓缩除去有机溶剂,水相用100毫升二氯甲烷萃取3次,有机相合并后,无水硫酸钠干燥,过滤,减压浓缩得到残余物,残余物经硅胶柱(二氯甲烷:甲醇=0-100%)纯化得到产品10.3g,产率:81%。
MS ESI:m/z=450,[M+H] +
步骤6:顺式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H吡唑啉[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000108
(1R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇(18.1g,40mmol)经过制备HPLC纯化得到产品9.3g,产率:51%。
1H NMR(400MHz,CDCl 3)δ8.36(s,1H),7.65-7.61(m,2H),7.13-7.04(m,3H),6.86-6.81(m,1H),6.75-6.71(m,1H),5.29(brs,2H),4.91-4.86(m,1H),3.93(s,3H),3.89-3.84(m,1H),2.35-2.32(m,1H),2.19-2.11(m,1H),2.20-1.91(m,4H),1.50-1.47(m,2H).
MS ESI:m/z=450,[M+H] +.
实施例27
3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷‐1,2‐二醇
Figure PCTCN2020072551-appb-000109
步骤1:3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000110
将3-碘代-1H-吡唑并[3,4-d]嘧啶-4-胺(5.0g,19.2mmol),2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(7.7g,23.0mmol),PdCl 2(dppf)(1.4g,1.9mmol)和磷酸钾(8.1g,38.3mmol)分散在36毫升N,N-二甲基甲酰胺和18毫升水的混合溶剂中,氮气置换三次,升温至120摄氏度,反应16小时。向反应液中加入50毫升水,过滤,滤渣用甲醇(20mL×1)洗涤,减压干燥得到产品2.5g,产率:37%。
MS ESI:m/z=352,[M+H] +
步骤2:1-(环己-2-烯-1-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000111
在氮气保护下,将3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(351mg,1mmol),环己‐2‐烯醇(118mg,1.2mmol)和三苯基膦(655mg,2.5mmol)溶于20毫升无水四氢呋喃中,冰浴下滴加偶氮二甲酸二异丙酯(606mg,3mmol)控制滴加速度,使反应体系温度不高于10摄氏度。滴加完毕后,反应液室温搅拌16小时。反应液减压浓缩得到残余物,残余物经反相柱层析(甲醇:二氯甲烷=0‐100%)纯化得到产品220mg,产率:51%。
MS ESI:m/z=432,[M+H] +
步骤3:3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷‐1,2‐二醇
Figure PCTCN2020072551-appb-000112
在氮气保护下,将1-(环己-2-烯-1-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(200mg,0.46mmol),N-甲基吗啉-N-氧化物(108mg,0.92mmol)溶于10毫升叔丁醇和5毫升水混合溶剂中,冰浴下加入锇酸钾(0.5mg,0.0014mmol),反应液室温搅拌16小时。向反应液中加入10毫升饱和亚硫酸,并用乙酸乙酯(10mL×2)萃取,有机相合并后,无水硫酸钠干燥,减压浓缩得到残余物,残余物经过制备HPLC纯化得到产品110mg,产率:52%。
1H NMR(400MHz,DMSO-d 6):δ8.19(s,1H),7.64-7.66(m,2H),7.2-7.0(m,6H),4.9-4.87(m,1H),4.60(s,1H),4.45-4.43(m,2H),3.99-3.97(m,2H),3.89(s,3H),1.94-1.90(m,1H),1.81-1.76(m,3H),1.55-1.49(m,2H).
MS ESI:m/z=466,[M+H] +
实施例28
4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1,2-二醇
Figure PCTCN2020072551-appb-000113
步骤1:1-(环己-2-烯-1-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000114
在氮气保护下,将3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(50mg,0.14mmol),环己‐3‐烯醇(17mg,0.17mmol)和三苯基膦(74mg,0.28mmol)溶于3毫升无水四氢呋喃中,冰浴下滴加偶氮二甲酸二异丙酯(57mg,0.28mmol)控制滴加速度控制反应体系内部温度不高于10度。滴加完毕后,反应液室温搅拌16小时。反应液减压浓缩得到残余物,残余物经柱层析(甲醇:二氯甲烷=0-100%)纯化得到产品30mg,产率:50%。
MS ESI:m/z=432,[M+H] +
步骤2:4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1,2-二醇
Figure PCTCN2020072551-appb-000115
在氮气保护下,将1-(环己-2-烯-1-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(30mg,0.070mmol),N-甲基吗啉-N-氧化物(10mg,0.084mmol)溶于2毫升叔丁醇和0.5毫升水混合溶剂中,冰浴下向反应体系中加入锇酸钾(0.5mg,0.0014mmol),反应液室温搅拌16小时。向反应液中加入10毫升饱和亚硫酸,反应体系用乙酸乙酯(10mL×2)萃取,有机相合并后,无水硫酸钠干燥,减压浓缩得到残余物,残余物经过制备HPLC纯化,得到产品Peak 1:3mg,产率:9%;及产品Peak 2:3mg,产率:9%。
Peak 1:
1H NMR(400MHz,CDCl 3)δ8.38(s,1H),7.65-7.61(m,2H),7.14-7.11(m,2H),7.09-7.05(m,1H),6.86-6.82(m,1H),6.76-6.72(m,1H),5.52(s,2H),5.14-4.97(m,1H),4.44(brs,1H),3.96-3.94(m,4H),3.86-3.80(m,1H),2.67-2.61(m,1H)2.57-2.48(m,1H),2.25-2.19(m,2H),1.94-1.84(m,2H),1.73-1.66(m,1H).
MS ESI:m/z=466,[M+H] +
Peak 2:
1H NMR(400MHz,CDCl 3)δ8.37(s,1H),7.64-7.60(m,2H),7.14-7.10(m,2H),7.08-7.04(m,1H),6.86-6.82(m,1H),6.76-6.71(m,1H),5.39(s,2H),5.26-5.20(m,1H),4.22(brs,1H),3.99(m,3H),3.87-3.84(m,1H),2.42-2.36(m,1H),2.31-2.35(m,2H),2.17-2.06(m,2H),1.96-1.88(m,2H).
MS ESI:m/z=466,[M+H] +
实施例29(对比化合物)
1-环己基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000116
步骤1:1-环己基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000117
将1-(环己-2-烯-1-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(190mg,0.44mmol),溶于20毫升甲醇中,加入10%湿钯碳(20mg),氢气(15psi)置换三次后室温反应过夜。硅藻土过滤,滤液浓缩得到残余物,残余物经制备HPLC纯化得到产品130mg,产率:68%。
1H NMR(400MHz,CDCl 3)δ8.37(s,1H),7.65-7.63(m,2H),7.14-7.06(m,2H),7.04(m,1H),6.85-6.81(m,1H),6.75-6.70(m,1H),5.43(s,2H),4.78-7.75(m,1H),3.99(s,3H),2.11-2.02(m,4H),1.95-1.91(m,2H),1.77-1.74(m,1H),1.54-1.49(m,2H),1.34-1.31(m,1H).
MS ESI:m/z=434,[M+H] +
实施例30
(5-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)四氢-2H-吡喃-2-基)甲醇
Figure PCTCN2020072551-appb-000118
步骤1:叔丁基((3,4-二氢-2H-吡喃-2-基)甲氧基)二苯基硅烷
Figure PCTCN2020072551-appb-000119
将(3,4-二氢-2H-吡喃-2-基)甲醇(1.0g,8.7mmol)溶解到二氯甲烷(20mL)中,将叔丁基二苯基氯硅烷(3.35g,12.2mmol),咪唑(1.43g,20.3mmol)依次加入到反应体系中,该反应体系在室温条件下搅拌过夜。将二氯甲烷(50mL)加入到反应液,用水(100mL×1)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得到残余物,残余物经柱层析(乙酸乙酯:石油醚=0-100%)纯化,得到产品2.3g,产率:75%。
1H NMR(400MHz,CDCl 3):δ7.71-7.68(m,4H),7.45-7.37(m,6H),6.37-6.35(d,J=8Hz,1H),4.68-4.65(m,1H),3.96-3.93(m,1H),3.83-3.77(m,1H),3.71-3.67(m,1H),2.08-2.00(m,1H),1.99-1.92(m,2H),1.74-1.71(m,1H),1.02(s,9H).
步骤2:6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-醇
Figure PCTCN2020072551-appb-000120
将叔丁基((3,4-二氢-2H-吡喃-2-基)甲氧基)二苯基硅烷(1.3g,3.7mmol)加入到四氢呋喃(20mL)中,氮气保护下,降温至-78摄氏度后,将硼烷二甲硫醚络合物(1.8mL,10M,18.0mmol)滴加到反应液中,滴加完毕后,自然升至室温搅拌过夜。向反应体系中缓慢滴加1N的氢氧化钠水溶液至无硼烷气体放出后,加入30%双氧水(5mL)至反应液中,反应液在45摄氏度搅拌2小时。向反应体系中加入水(50mL)后用乙酸乙酯(50mL×1)萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩得到残余物,残余物经反相柱层析(乙腈:水=5-95%)纯化,得到产品1.0g,产率:73%。
步骤3:1-(6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-基)-3-碘-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000121
将3-碘代-1H-吡唑并[3,4-d]嘧啶-4-胺(0.3g,1.15mmol)加入到四氢呋喃(20mL)中,氮气保护下,将6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-醇(555mg,1.50 mmol),三苯基膦(753mg,2.87mmol)加入到反应液,0摄氏度下将偶氮二甲酸二异丙酯(697mg,3.45mmol)滴加到反应液中,反应体系在室温条件下搅拌过夜。减压浓缩得到残余物,残余物经反相柱层析(乙腈:水=5-95%)纯化得到产品330mg,产率:47%。
MS ESI:m/z=614,[M+H] +
步骤4:1-(6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
Figure PCTCN2020072551-appb-000122
将1-(6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-基)-3-碘-1H-吡唑并[3,4-d]嘧啶-4-胺(16.4g,26.8mmol)加入到1,4二氧六环(160mL)和水(40mL)的混合溶剂中,将2-(4-(2-氟-3-甲氧基苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(8.95g,26.8mmol),碳酸钾(11.1g,80.4mmol)加入到反应体系中,氮气保护下,将四(三苯基膦)钯(1.55g,1.34mmol)加入反应液,该反应体系在100摄氏度搅拌过夜。减压浓缩得到残余物,残余物经柱层析(甲醇:二氯甲烷=0-100%)纯化,得到产品Peak 1:1.4g,产率:7%;及产品Peak2:1.0g,产率:5%。
Peak 1:
1H NMR(400MHz,CDCl 3):δ8.34(s,1H),7.68-7.65(m,4H),7.62-7.60(m,2H),7.41-7.32(m,6H),7.11-7.09(m,2H),7.07-7.04(m,1H),6.86-6.82(m,1H),6.76-6.72(m,1H),5.57(brs,2H),4.83-4.82(m,1H),4.49-4.45(m,1H),3.94(s,3H),3.87-3.82(m,2H),3.71-3.69(m,2H),2.62-2.57(m,1H),2.28-2.22(m,1H),2.14-2.08(m,1H),1.82(m,1H),1.03(s,9H).
MS ESI:m/z=704,[M+H] +
Peak 2:
1H NMR(400MHz,CDCl 3):δ8.36(s,1H),7.71-7.68(m,4H),7.64-7.61(m,2H),7.44-7.37(m,6H),7.14-7.12(m,2H),7.09-7.04(m,1H),6.86-6.81(m,1H),6.76-6.72(m,1H),5.51(brs,2H),4.97-4.92(m,1H),4.16-4.11(m,1H),4.00-3.94(m,1H),3.93(s,3H),3.81-3.77(m,1H),3.66-3.60(m,2H),2.46-2.36(m,1H),2.26-2.23(m,1H),1.95(m,1H),1.70-1.60(m,1H),1.07(s,9H).
MS ESI:m/z=704,[M+H] +
步骤5:(5-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)四氢-2H-吡喃-2-基)甲醇
Figure PCTCN2020072551-appb-000123
将1-(6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺Peak 1(1.4g,2.0mmol)加入到四氢呋喃(20mL)中,将四丁基氟化铵的四氢呋喃溶液(2.6mL,1M,2.6mmol)加入到反应液,在室温条件下搅拌过夜。减压浓缩得到残余物,残余物经反相柱层析(乙腈:水=5-95%)纯化,并且经手性SFC纯化得到产品Peak 1-1:400mg,产率:43%,及产品Peak 1-2:400mg,产率:43%。
Peak 1-1:
1H NMR(400MHz,CDCl 3):δ8.35(s,1H),7.65-7.63(m,2H),7.13-7.05(m,3H),6.86-6.82(m,1H),6.77-6.72(m,1H),5.73(s,2H),4.89(s,1H),4.57-4.54(m,1H),3.97-3.93(m,1H),3.93(s,3H),3.74-3.66(m,3H),2.61-2.56(m,1H),2.33-2.30(m,1H),2.20-2.00(m,1H),1.57-1.59(m,1H).
MS ESI:m/z=466,[M+H] +
Peak 1-2:
1H NMR(400MHz,CDCl 3):δ8.37(s,1H),7.65-7.63(m,2H),7.13-7.05(m,3H),6.86-6.82(m,1H),6.77-6.72(m,1H),5.73(s,2H),4.89(s,1H),4.57-4.54(m,1H),3.97-3.93(m,1H),3.93(s,3H),3.74-3.66(m,3H),2.61-2.56(m,1H),2.33-2.30(m,1H),2.20-2.00(m,1H),1.57-1.59(m,1H).
MS ESI:m/z=466,[M+H] +
将1-(6-(((叔丁基二苯基硅烷基)氧基)甲基)四氢-2H-吡喃-3-基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺Peak 2(1.0g,1.4mmol)加入到四氢呋喃(20mL)中,将四丁基氟化铵的四氢呋喃溶液(1.8mL,1M,1.8mmol)加入到反应液,在室温条件下搅拌过夜。减压浓缩得到残余物,残余物经反相柱层析(乙腈:水=5-95%)纯化,并且经手性SFC纯化得到产品Peak 2-1:200mg,产率:30%,及产品Peak 2-2:200mg,产率:30%。
Peak 2-1
1H NMR(400MHz,CDCl 3):δ8.35(s,1H),7.63-7.60(m,2H),7.14-7.05(m,3H),6.87-6.83(m,1H),6.74-6.72(m,1H),5.74(s,2H),4.89-4.63(m,1H),4.20-4.16(m,1H),4.03-3.97(m,1H),3.97(s,3H),3.71-3.58(m,3H),2.46-2.42(m,1H),2.27-2.24(m,1H),1.83-1.80(m,1H),1.73-1.72(m,1H).
MS ESI:m/z=466,[M+H] +
Peak 2-2:
1H NMR(400MHz,CDCl 3):δ8.35(s,1H),7.63-7.60(m,2H),7.14-7.05(m,3H),6.87-6.83(m,1H),6.74-6.72(m,1H),5.74(s,2H),4.89-4.63(m,1H),4.20-4.16(m,1H),4.03-3.97(m, 1H),3.97(s,3H),3.71-3.58(m,3H),2.46-2.42(m,1H),2.27-2.24(m,1H),1.83-1.80(m,1H),1.73-1.72(m,1H).
MS ESI:m/z=466,[M+H] +
实施例31
(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-(甲氧基-d3)苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000124
步骤1:1-氟-2-(甲氧基-d3)苯
Figure PCTCN2020072551-appb-000125
氮气保护,冰浴下向2-氟苯酚(2.0g,17.9mmol),三苯基膦(7.2g,35.8mmol)和氘代甲醇-d4(946mg,26.8mmol)的无水四氢呋喃(20mL)溶液中逐滴加入偶氮二甲酸二异丙酯(7.2g,35.8mmol)后室温反应过夜。反应体系减压浓缩得到残余物,残余物经柱层析(乙酸乙酯:石油醚=0-100%)纯化,得到产品1.1g,产率:35%。
1H NMR(400MHz,CDCl 3)δ7.16-7.04(m,2H),7.03-6.90(m,2H).
步骤2:2-氟-3-(甲氧基-d3)苯酚
Figure PCTCN2020072551-appb-000126
在氮气保护下,将1-氟-2-(甲氧基-d3)苯(1.3g,10.3mmol)和五甲基二亚乙基三胺(2.6mL,12.4mmol)溶解在无水四氢呋喃(15mL)中。反应体系降温至-78摄氏度,缓慢滴加正丁基锂正己烷溶液(5.0mL,2.5M,12.4mmol),继续反应2小时后,维持此温度,缓慢滴加硼酸三异丙酯(2.9mL,12.4mmol)。自然恢复至室温后继续反应16小时。向反应体系中滴加乙酸(0.9mL,10.5mmol)淬灭反应,滴加完毕后,反应瓶置于冰水浴中,滴加30%双氧水(1.6mL,2.4mmol)。反应中析出大量黏稠固体。反应混合物在室温下搅拌30分钟,向反应体系中加入水(20mL)稀释后,用乙酸乙酯(20mL×2)萃取。有机相合并后用饱和亚硫酸氢钠溶液(30mL×1)洗涤,无水硫酸钠干燥,减压浓缩得到残余物。残留物经柱层析(石油醚:乙酸乙酯=0-100%)纯化得到产品636mg,产率:48%。
1H NMR(400MHz,CDCl 3)δ7.02-6.96(m,1H),6.71-6.65(m,1H),6.61-6.56(m,1H),5.36(brs,1H).
步骤3:1-(4-溴苯氧基)-2-氟-3-(甲氧基-d3)苯
Figure PCTCN2020072551-appb-000127
将2-氟-3-(甲氧基-d3)苯酚(636mg,4.4mmol),4-溴苯硼酸(1.3g,6.6mmol),无水醋酸铜(798mg,4.4mmol)和干燥的粉末状4A分子筛(3.0g)分散在二氯甲烷(10mL)中,缓慢滴加三乙胺(887mg,8.8mmol)。反应液在氧气氛围下(15psi),室温反应16小时。反应液经硅藻土过滤除去不溶物,滤液减压浓缩得到残余物,残余物经柱层析(石油醚:乙酸乙酯=0-100%)纯化,得到产品1.0g,产率:77%。
1H NMR(400MHz,CDCl 3)δ7.47-7.44(m,2H),7.09-7.07(m,1H),6.92-6.90(m,2H),6.86-6.71(m,1H),6.68-6.66(m,1H).
步骤4:2-(4-(2-氟-3-(甲氧基-d3)苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂环戊环
Figure PCTCN2020072551-appb-000128
将1-(4-溴苯氧基)-2-氟-3-(甲氧基-d3)苯(1.0g,3.3mmol),联硼酸频那醇酯(1.0g,4.0mmol),PdCl 2(dppf)(219mg,0.3mmol)和乙酸钾(655mg,6.7mmol)分散在1,4-二氧六环(15mL)中,置换氮气三次后,升温至110摄氏度反应16小时。反应体系降温后,硅藻土过滤除去不溶物,滤液减压浓缩得到残余物,残余物经柱层析(乙酸乙酯:石油醚=0-100%)纯化,得到产品940mg,产率:78%。
1H NMR(300MHz,CDCl 3)δ7.83(d,J=8.4Hz,2H),7.09-6.99(m,3H),6.85-6.80(m,1H),6.73-6.67(m,1H),1.38(s,12H).
步骤5:(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000129
将(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己基乙酸酯粗品溶于甲醇(40mL)和四氢呋喃(40mL)的混合溶液中,加入一水合氢氧化锂(1.6g,39.2mmol)。反应液于室温搅拌16小时。抽滤除去固体,滤液减压浓缩得到残余物,残余物经柱层析(二氯甲烷:甲醇=0-100%)纯化得到产品910mg。
MS ESI:m/z=360,[M+H] +
步骤6:(1R,3S)-3-(4-氨基-3-(4-(2-氟-3-(甲氧基-d3)苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000130
将(1R,3R)-3-(4-胺基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)环己醇(300mg,0.84mmol),2-(4-(2-氟-3-(甲氧基-d3)苯氧基)苯基)-4,4,5,5-四甲基-1,3,2-二氧杂环戊环(347mg,1.0mmol),四三苯基膦钯(92mg,0.08mmol)和碳酸钾(231mg,1.68mmol)分散在1,4-二氧六环(15mL)和水(1mL)的混合溶剂中,置换氮气三次后,升温至120度反应16小时。反应体系降温后,减压浓缩除去有机溶剂,水相用二氯甲烷(20mL×2)萃取,有机相合并后,无水硫酸钠干燥,过滤,减压浓缩得到残余物。残余物经过制备HPLC纯化得到产品110mg,产率:29%。
1H NMR(400MHz,CDCl 3)δ8.36(s,1H),7.64(d,J=8.8Hz,2H),7.13(d,J=8.8Hz,2H),7.09-7.03(m,1H),6.85-6.80(m,1H),6.74-6.70(m,1H),5.58(brs,2H),5.29-5.23(m,1H),4.40(s,1H),2.39-2.32(m,1H),2.15-2.05(m,3H),1.98-1.73(m,3H),1.67-1.60(m,1H).
MS ESI:m/z=453,[M+H] +
实施例32
(1S,3S)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑啉[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000131
步骤1:反式-3-(3-(4-(2-氟-3-甲氧基苯氧基)苯基)-4-((三苯基-15-膦叉基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基4-硝基苯甲酸酯
Figure PCTCN2020072551-appb-000132
将顺式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己-1-醇(3.3g,9.55mmol)溶于四氢呋喃(60mL)中,向其中依次加入对硝基苯甲酸(1.60g,9.55mmol),三苯基膦(4.81g,18.38mmol)后,降温至0摄氏度,氮气保护下,滴加偶氮二甲酸二异丙酯(4.45g,22.05mmol),滴加完毕后,自然恢复至室温反应2小时 后,向反应体系中加入饱和食盐水(100mL)淬灭反应,二氯甲烷(50mL x 3)萃取,有机相合并后,无水硫酸钠干燥,过滤,滤液减压浓缩得到残余物,残余物经柱层析(石油醚:乙酸乙酯=0-100%)纯化得到粗产品6.8g。
MS ESI:m/z=859,[M+H] +
步骤2:反式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基4-硝基苯甲酸酯
Figure PCTCN2020072551-appb-000133
将反式-3-(3-(4-(2-氟-3-甲氧基苯氧基)苯基)-4-((三苯基-15-膦叉基)氨基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基4-硝基苯甲酸酯粗品(5.6g)溶于乙酸(20mL)中,并向其中加入水(100mL)稀释后,升温至130摄氏度,回流反应12小时。反应液减压浓缩除去大部分溶剂,向体系中加入水(100mL)后,用二氯甲烷(50mL x 3)萃取,有机相合并后,无水硫酸钠干燥,减压浓缩得到残余物,残余物用甲苯(100mL x 3)带剩余的溶残乙酸,得到粗产品6.8g。
MS ESI:m/z=599,[M+H] +
步骤3:反式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑啉[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000134
将反式(trans)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基4-硝基苯甲酸酯粗品(6.8g)四氢呋喃(40mL)和水(10mL)的混合溶液中,向其中加入氢氧化锂一水合物(1.0g,23.8mmol)。室温反应2小时后,向反应体系中加入饱和氯化钠溶液(100mL),并用二氯甲烷(100mL x 3)萃取,有机相合并后,无水硫酸钠干燥,过滤,减压浓缩得到残余物,残余物经柱层析(二氯甲烷:甲醇=0-100%)纯化得到产品1.6g,三步产率:48.5%。
MS ESI:m/z=450,[M+H] +
步骤4:(1S,3S)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑啉[3,4-d]嘧啶-1-基)环己醇
Figure PCTCN2020072551-appb-000135
将反式-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑啉[3,4-d]嘧啶-1-基)环己醇(1.6g,3.56mmol)经手性SFC制备纯化得到产品640mg,产率:80%。
1H NMR(400MHz,CDCl3)δ8.37(s,1H),7.65-7.62(m,2H),7.14-7.04(m,3H),6.85-6.81(m,1H),6.75-6.71(m,1H),5.41(brs,2H),5.25(m,1H),4.40(m,1H),3.94(s,3H),2.36-2.33(m,1H),2.14-2.01(m,4H),1.80-1.77(m,2H),1.66(m,1H).
MS ESI:m/z=450,[M+H] +
实施例33
(1R,3R)-3-(4-(4-氨基-1-(3-羟基环己基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯氧基)-2-氟苯酚
Figure PCTCN2020072551-appb-000136
将(1R,3R)-3-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑啉[3,4-d]嘧啶-1-基)环己醇(450mg,1.0mmol)溶于二氯甲烷(10mL)中,降温至0摄氏度,向其中缓慢滴加1N三溴化硼的二氯甲烷溶液(1.5mL,1.5mmol),自然升至室温反应1小时,向反应体系中加入二氯甲烷(50mL)稀释,并用饱和碳酸氢钠溶液(100mL x 3)洗涤,有机相使用无水硫酸钠干燥,过滤,减压浓缩得到残余物,残余物经制备HPLC纯化得到产品240mg,产率:55%。
1H NMR(400MHz,DMSO-d6):11.12(brs,1H),8.22(s,1H),7.64(d,J=8.4Hz,2H),7.11(d,J=8.4Hz,2H),7.03-6.99(m,1H),6.86-6.81(m,1H),6.67-6.63(m,1H),7.21-6.33(m,2H),5.12-5.06(m,1H),4.64(brs,1H),4.14(brs,1H),2.18-2.07(m,1H),1.92-1.83(m,4H),1.69-1.59(m,2H),1.51-1.45(m,1H).
MS ESI:m/z=436,[M+H] +
生物化学评价
式A化合物的BTK/BTK(C481S)抑制活性在美国宾夕法尼亚州马尔文市大峡谷干道的反应生物公司(Reaction Biology Corporation,One Great Valley Parkway,Malvern,PA,USA)测定。使用人全长的BTK/BTK(C481S)酶,底物为20μM的肽底物[KVEKIGEGTYGVVYK]。测定用的ATP浓度为10μM,星形孢菌素用作标准品,IC 50为3.94nM。
如下表1为诸实施例化合物对BTK/BTK(C481S)酶的抑制活性
Figure PCTCN2020072551-appb-000137
其中,一些实施例在BTK/BTK(C481S)酶上的IC 50值如下表2所示:
Figure PCTCN2020072551-appb-000138
(a)标记的实施例中ATP的浓度为30uM;(b)标记的实施例中ATP的浓度为100uM;(c)标记的实施例中ATP的浓度为10uM;(d)标记的实施例中ATP的浓度为50uM;“-”表示没有测试。
结果显示,相较于环上无含羟基取代基的对照化合物29,本申请的各个化合物展现出更好的针对BTK(野生型和突变型)的抑制活性。
采用CellTiter-Glo方法检测实施例对体外培养的人弥漫大B淋巴瘤细胞TMD8的抑制活性,IC 50值如下表3所示。
如下表3为诸实施例化合物对TMD8细胞的抑制活性
化合物 IC 50(nM)
实施例1 20
实施例3A 208
实施例3B 200
实施例5 29
实施例6 17
实施例7 504
实施例8A 90
实施例9A 72
实施例9B 37
实施例10 145
实施例16 41
实施例17 33
实施例18 183
实施例19 0.5
实施例20 8.6
实施例22 8
体内药效学评价
在人弥漫大B淋巴瘤TMD8细胞NOD/SCID小鼠皮下移植瘤模型中,检测实施例1对肿瘤生长的抑制作用,肿瘤体积及动物体重如下图1及图2所示。实施例1在30mg/kg剂量下每天口服给药一次,或在10→5mg/kg剂量下每天口服给药两次,对TMD8皮下移植瘤的生长具有明显抑制作用,给药22天后对肿瘤生长的抑制率(TGI)分别为49.6%和52.7%。实施例1在30mg/kg剂量下每天口服给药一次对动物体重无明显影响,在10mg/kg剂量下每天口服给药两次、连续给药8天后,动物体重有所下降,故调整剂量至5mg/kg,继续给药14天。
图1显示了给药期间溶剂对照及各给药组TMD8皮下异种移植瘤体积(平均值±标准差)。结果显示,在初次给药22天后,本发明化合物表现出了显著的治疗效果,且其治疗效果与阳性药物依鲁替尼相当。
图2显示了给药期间溶剂对照及各给药组动物体重(平均值±标准差)的变化。结果显示,本发明实施例1化合物在表现出显著的抗肿瘤效果同时,并没有对动物体重产生明显影响。
根据实验结果,本发明实施例1具有显著的抗肿瘤效果,显著的抑制了肿瘤的生长而没有对动物体重产生明显影响。

Claims (10)

  1. 一种如下式(A)所示的化合物,或其药学上可接受的盐,
    Figure PCTCN2020072551-appb-100001
    其中:
    A环和B环互相并联,且各自独立地为取代或未取代的5-15元杂环或杂芳环;
    Cyc 1选自下组:取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 2-C 6烯基、取代或未取代的C 3-C 10环烯基、取代的或未取代的5-15元杂环基、取代的或未取代的5-15元杂芳基、取代或未取代的C 6-C 10芳基;
    Cyc 2选自下组:取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 2-C 6烯基、取代或未取代的C 3-C 10环烯基、取代的或未取代的5-15元杂环基、取代的或未取代的5-15元杂芳基、取代或未取代的C 6-C 10芳基;
    L 1、L 2各自独立地选自下组:化学键、N、O、S、-S(=O)、-S(=O) 2、C(=O)、-C(O)NH-;
    Z为(CR 2R 3) n,n为0、1、2、3、4、5或6;
    Y为(CR 4R 5) m,m为0、1、2、3、4、5或6;
    U为(CR 6R 7) r,r为0、1、2或3;
    且n、m和r不同时为0;
    R 2、R 3、R 4、R 5、R 6和R 7各自独立地选自下组:H、NH 2、OH、卤素、取代或未取代的C 1-C 6烷基;或R 2、R 3、R 4、R 5、R 6、R 7和R 8中任意两个与其相连的碳原子以及间隔的环骨架原子共同形成C 3-C 8碳环,或4-8元的杂环,其中,所述杂环的杂原子选自:S、O、或NR f;R f为H、C 1-C 10烷基、C 3-C 10环烷基、C 6-C 20芳基、或C 3-C 14杂芳基;且R 2、R 3、R 4、R 5、R 6、R 7、R 8和R 9中至少有一个是OH或-[C(R 10)(R 11)] k-OH;
    W选自下组:N、O、S或化学键;
    X为-C(R 8R 9)-;
    R 8选自下组:H、取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 6-C 20芳基、或取代或未取代的C 3-C 14杂芳基、CO 2H、C(O)NR f 2
    R 9选自下组:H、OH或-[C(R 10)(R 11)] k-OH;
    或R 8和R 9共同构成=O;
    且当R 8为H时,R 9为OH或-[C(R 10)(R 11)] k-OH;
    R 10和R 11各自独立地选自下组:H、取代或未取代的C 1-C 6烷基、取代或未取代的C 3-C 10环烷基、取代或未取代的C 6-C 10芳基、或取代或未取代的C 3-C 14杂芳基;或R 10和R 11与其相连的碳原子共同形成C 3-C 8碳环,或4-8元的杂环,其中杂原子可以是硫、 氧、或NR f
    k为1、2、3、4、5或6;
    除非特别说明,上述的杂芳环、杂芳基、杂环、杂环基各自独立地含有1-4个选自N,O和S的杂原子;
    除非特别说明,所述的取代指基团被一个或多个(例如2个、3个、4个等)选自下组的取代基所取代:卤素、C1-C6烷基、卤代的C1-C6烷基、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、氧代、-CN、羟基、羟基-C1-C6烷基、氨基、羧基、C6-C10芳基、卤代的C6-C10芳基、未取代或被选自下组的取代基取代的具有1-3个选自N、S和O的杂原子的5-10元杂芳基:卤素、苯基。
  2. 如权利要求1所述的化合物,其特征在于,所述的
    Figure PCTCN2020072551-appb-100002
    具有如下结构:
    Figure PCTCN2020072551-appb-100003
    其中,虚线为化学键或无;各A 1、A 2、A 3、A 4、A 5、A 6、A 7、A 8和A 9各自独立地为O、S、N、NH、CH或CH 2
    波浪线表示连接位点;
    且上述基团的各个可取代位点可以包括取代基,其中,取代基的定义如上文中所述。
  3. 如权利要求1所述的化合物,其特征在于,所述的
    Figure PCTCN2020072551-appb-100004
    具有选自下组的结构:
    Figure PCTCN2020072551-appb-100005
  4. 如权利要求1所述的化合物,其特征在于,所述的化合物具有如下式(B)所示的结构:
    Figure PCTCN2020072551-appb-100006
    其中,
    R 12、R 13、R 14和R 15各自独立地选自下组:H、卤素、取代或未取代的C 1-C 6烷基、取代或未取代的C 1-C 6烷氧基、取代或未取代的C 3-C 10环烷基、取代或未取代的5-15元的杂环基。
  5. 权利要求1所述化合物,其特征在于,所述的化合物具有如下式(C)所示的结构:
    Figure PCTCN2020072551-appb-100007
    式(C)中,V为N或-CR 16-;
    R 12、R 13、R 14和R 15各自独立地选自下组:H、卤素、取代或未取代的C 1-C 6烷基、取代或未取代的C 1-C 6烷氧基、取代或未取代的C 3-C 10环烷基、取代或未取代的5-15元的杂环基;
    R 16为H、卤素取代或非取代的C 1-C 6烷基、取代或非取代的C 3-C 10环烷基。
  6. 权利要求1所述化合物,其特征在于,所述的化合物具有如下式(D)、(E)或(F)所示的结构:
    Figure PCTCN2020072551-appb-100008
  7. 权利要求1所述的化合物,其特征在于,所述化合物选自下组:
    Figure PCTCN2020072551-appb-100009
    Figure PCTCN2020072551-appb-100010
    Figure PCTCN2020072551-appb-100011
    Figure PCTCN2020072551-appb-100012
    Figure PCTCN2020072551-appb-100013
    Figure PCTCN2020072551-appb-100014
  8. 一种药物组合物,其包含(1)权利要求1-7任一所述的化合物、或其立体异构体或互变异构体、或其药学上可接受的盐、水合物或溶剂化物;和(2)药学上可接受的载体。
  9. 如权利要求1-7任一所述的化合物、或其立体异构体或互变异构体、或其药学上可接受的盐、水合物或溶剂化物或如权利要求6所述的药物组合物的用途,其特征在于,用于制备预防和/或治疗BTK的异常活性以及BTK突变体(如C481S)异常活性相关的疾病的药物。
  10. 如权利要求9所述的用途,其特征在于,所述的疾病或病症选自膀胱癌,脑肿瘤,乳腺癌,子宫癌,结肠直肠癌,食道癌,肝脏癌症,滤泡性淋巴瘤,黑色素瘤,恶性血液病,骨髓瘤,卵巢癌,非小细胞肺癌,前列腺癌,小细胞肺癌,和B-细胞来源的淋巴恶性肿瘤,B细胞增殖性病症:弥漫性B细胞淋巴瘤、滤泡性淋巴瘤、慢性淋巴细胞淋巴瘤、慢性淋巴细胞白血病、B细胞幼淋巴细胞白血病、淋巴质浆细胞淋巴瘤/瓦尔登斯特伦氏巨球蛋白血症、脾脏边缘带淋巴瘤、浆细胞性骨髓瘤、浆细胞瘤、结外边缘带B细胞淋巴瘤、结内边缘带B细胞淋巴瘤、外套细胞淋巴瘤、纵隔(胸腺)大B细胞淋巴瘤、血管内大B细胞淋巴瘤、原发性渗出性淋巴瘤、伯基特氏淋巴瘤/白血病或淋巴瘤样肉芽肿病。
PCT/CN2020/072551 2019-01-18 2020-01-16 作为布鲁顿酪氨酸激酶抑制剂的环状分子 WO2020147798A1 (zh)

Priority Applications (14)

Application Number Priority Date Filing Date Title
EP20741672.8A EP3912980A4 (en) 2019-01-18 2020-01-16 CYCLIC MOLECULES SERVING AS BRUTON'S TYROSINE KINASE INHIBITOR
CN202080009961.2A CN113710671B (zh) 2019-01-18 2020-01-16 作为布鲁顿酪氨酸激酶抑制剂的环状分子
US17/423,815 US20220081445A1 (en) 2019-01-18 2020-01-16 Cyclic molecules as bruton's tyrosine kinase inhibitor
BR112021014100-6A BR112021014100A2 (pt) 2019-01-18 2020-01-16 Moléculas cíclicas como inibidor de tirosina quinase de bruton
MX2021008632A MX2021008632A (es) 2019-01-18 2020-01-16 Moléculas cíclicas como inhibidores de la tirosina cinasa de bruton.
AU2020208128A AU2020208128B2 (en) 2019-01-18 2020-01-16 Cyclic molecules as bruton's tyrosine kinase inhibitor
CA3129841A CA3129841C (en) 2019-01-18 2020-01-16 Cyclic molecules as bruton's tyrosine kinase inhibitor
CN202310355998.6A CN116589465A (zh) 2019-01-18 2020-01-16 作为布鲁顿酪氨酸激酶抑制剂的环状分子
EA202191995A EA202191995A1 (ru) 2019-01-18 2020-01-16 Циклические молекулы в качестве ингибитора тирозинкиназы брутона
KR1020217025977A KR20210135497A (ko) 2019-01-18 2020-01-16 브루톤 티로신 키나아제 억제제로서의 고리형 분자
JP2021542229A JP7436994B2 (ja) 2019-01-18 2020-01-16 ブルトン型チロシンキナーゼ阻害剤としての環状分子
SG11202107886RA SG11202107886RA (en) 2019-01-18 2020-01-16 Cyclic molecules as bruton's tyrosine kinase inhibitor
IL284916A IL284916A (en) 2019-01-18 2021-07-18 Cyclic molecules as Bruton's tyrosine kinase inhibitors
JP2023161762A JP2023179562A (ja) 2019-01-18 2023-09-25 ブルトン型チロシンキナーゼ阻害剤としての環状分子

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910049183.9 2019-01-18
CN201910049183.9A CN111454268B (zh) 2019-01-18 2019-01-18 作为布鲁顿酪氨酸激酶抑制剂的环状分子

Publications (1)

Publication Number Publication Date
WO2020147798A1 true WO2020147798A1 (zh) 2020-07-23

Family

ID=71613532

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/072551 WO2020147798A1 (zh) 2019-01-18 2020-01-16 作为布鲁顿酪氨酸激酶抑制剂的环状分子

Country Status (13)

Country Link
US (1) US20220081445A1 (zh)
EP (1) EP3912980A4 (zh)
JP (2) JP7436994B2 (zh)
KR (1) KR20210135497A (zh)
CN (3) CN111454268B (zh)
AU (1) AU2020208128B2 (zh)
BR (1) BR112021014100A2 (zh)
CA (1) CA3129841C (zh)
EA (1) EA202191995A1 (zh)
IL (1) IL284916A (zh)
MX (1) MX2021008632A (zh)
SG (1) SG11202107886RA (zh)
WO (1) WO2020147798A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021136219A1 (en) * 2020-01-02 2021-07-08 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Btk inhibitors
WO2022012510A1 (zh) * 2020-07-15 2022-01-20 成都海博为药业有限公司 一种作为btk抑制剂的化合物及其制备方法与用途

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114573586B (zh) * 2020-11-28 2023-11-03 杭州和正医药有限公司 一种抑制布鲁顿酪氨酸激酶活性的多环化合物、药物组合物及其应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1520298A (zh) * 2001-03-22 2004-08-11 �����ʩ���عɷݹ�˾ 作为治疗剂的吡唑并嘧啶类
CN101610676A (zh) * 2006-09-22 2009-12-23 药品循环公司 布鲁顿酪氨酸激酶的抑制剂
CN102656173A (zh) * 2009-10-12 2012-09-05 药品循环公司 布鲁顿酪氨酸激酶抑制剂
CN105732638A (zh) * 2016-01-22 2016-07-06 成都倍特药业有限公司 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法
CN107827892A (zh) * 2017-10-27 2018-03-23 上海应用技术大学 一种非受体酪氨酸激酶小分子抑制剂及其应用

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL148718A0 (en) * 1999-09-17 2002-09-12 Abbott Gmbh & Co Kg Pyrazolopyrimidines as therapeutic agents
WO2005074603A2 (en) * 2004-02-03 2005-08-18 Abbott Laboratories Aminobenzoxazoles as therapeutic agents
EP1979002A2 (en) * 2005-12-19 2008-10-15 OSI Pharmaceuticals, Inc. Combination of igfr inhibitor and anti-cancer agent
EP2139487B1 (en) * 2007-03-28 2015-11-11 Pharmacyclics LLC Inhibitors of bruton's tyrosine kinase
JP5369183B2 (ja) * 2008-07-16 2013-12-18 ファーマサイクリックス,インク. 固形腫瘍の治療用のブルートンのチロシンキナーゼの阻害剤
US9138436B2 (en) * 2011-07-13 2015-09-22 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
EP2548877A1 (en) * 2011-07-19 2013-01-23 MSD Oss B.V. 4-(5-Membered fused pyridinyl)benzamides as BTK-inhibitors
US20140377285A1 (en) * 2011-11-08 2014-12-25 Intellikine, Llc Treatment regimens using multiple pharmaceutical agents
WO2013113097A1 (en) * 2012-01-31 2013-08-08 Beta Pharma Canada Inc. Cyclic molecules as bruton's tyrosine kinase inhibitors
CN104379586B (zh) * 2012-05-31 2018-01-16 制药科学公司 蛋白激酶抑制剂
DK2861599T3 (da) * 2012-06-18 2020-03-02 Principia Biopharma Inc Reversible kovalente pyrrolo- eller pyrazolopyrimidiner, der er nyttige til behandling af cancer og autoimmunsygdomme
MX2016009403A (es) * 2014-02-03 2016-09-16 Cadila Healthcare Ltd Compuestos heterociclicos.
CN105017256A (zh) * 2014-04-29 2015-11-04 浙江导明医药科技有限公司 多氟化合物作为布鲁顿酪氨酸激酶抑制剂
WO2016004272A1 (en) * 2014-07-02 2016-01-07 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
CN106999494A (zh) * 2014-08-01 2017-08-01 药品循环有限公司 布鲁顿酪氨酸激酶的抑制剂
TW201613926A (en) * 2014-08-01 2016-04-16 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
CN108431007B (zh) * 2015-09-16 2022-06-07 洛克索肿瘤学股份有限公司 用于治疗癌症的作为btk抑制剂的吡唑并嘧啶衍生物
CN107344940B (zh) * 2016-05-06 2020-04-21 广东东阳光药业有限公司 Btk抑制剂及其用途
CN109153680B (zh) * 2016-07-07 2021-04-23 株式会社大熊制药 4-氨基吡唑并[3,4-d]嘧啶基氮杂双环衍生物及含其的药物组合物
WO2018089786A1 (en) * 2016-11-11 2018-05-17 Millennium Pharmaceuticals, Inc. Atg7 inhibitors and the uses thereof
GB201705971D0 (en) * 2017-04-13 2017-05-31 Cancer Res Tech Ltd Inhibitor compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1520298A (zh) * 2001-03-22 2004-08-11 �����ʩ���عɷݹ�˾ 作为治疗剂的吡唑并嘧啶类
CN101610676A (zh) * 2006-09-22 2009-12-23 药品循环公司 布鲁顿酪氨酸激酶的抑制剂
CN102656173A (zh) * 2009-10-12 2012-09-05 药品循环公司 布鲁顿酪氨酸激酶抑制剂
CN105732638A (zh) * 2016-01-22 2016-07-06 成都倍特药业有限公司 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法
CN107827892A (zh) * 2017-10-27 2018-03-23 上海应用技术大学 一种非受体酪氨酸激酶小分子抑制剂及其应用

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021136219A1 (en) * 2020-01-02 2021-07-08 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Btk inhibitors
EP4087845A4 (en) * 2020-01-02 2024-01-24 Dizal Jiangsu Pharmaceutical Co Ltd BTK INHIBITORS
WO2022012510A1 (zh) * 2020-07-15 2022-01-20 成都海博为药业有限公司 一种作为btk抑制剂的化合物及其制备方法与用途

Also Published As

Publication number Publication date
CN113710671B (zh) 2023-04-28
CN116589465A (zh) 2023-08-15
MX2021008632A (es) 2021-10-26
AU2020208128B2 (en) 2023-05-25
CA3129841A1 (en) 2020-07-23
JP2022523480A (ja) 2022-04-25
CN113710671A (zh) 2021-11-26
US20220081445A1 (en) 2022-03-17
IL284916A (en) 2021-09-30
BR112021014100A2 (pt) 2021-09-21
SG11202107886RA (en) 2021-08-30
EP3912980A4 (en) 2022-12-07
EA202191995A1 (ru) 2021-11-18
EP3912980A1 (en) 2021-11-24
KR20210135497A (ko) 2021-11-15
CN111454268B (zh) 2023-09-08
CA3129841C (en) 2023-09-26
JP7436994B2 (ja) 2024-02-22
CN111454268A (zh) 2020-07-28
JP2023179562A (ja) 2023-12-19
AU2020208128A1 (en) 2021-09-09

Similar Documents

Publication Publication Date Title
JP5985658B2 (ja) ブルトンチロシンキナーゼ阻害薬としての環状分子
EP2785720B1 (en) Heterocyclic amides compounds which are hdac6 inhibitors and used as anti-tumoral agents
WO2020147798A1 (zh) 作为布鲁顿酪氨酸激酶抑制剂的环状分子
WO2013185202A1 (en) Apoptosis inducers
TW200820972A (en) 4-methylpyridopyrimidinone compounds
US9226923B2 (en) Spirocyclic molecules as protein kinase inhibitors
US8946445B2 (en) Heterocyclic molecules as apoptosis inducers

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: 20741672

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021542229

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021014100

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 3129841

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020208128

Country of ref document: AU

Date of ref document: 20200116

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021014100

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210716

ENP Entry into the national phase

Ref document number: 2020741672

Country of ref document: EP

Effective date: 20210818