CN107759602B - Compound containing conjugated allene structure, pharmaceutical composition and application thereof - Google Patents

Compound containing conjugated allene structure, pharmaceutical composition and application thereof Download PDF

Info

Publication number
CN107759602B
CN107759602B CN201710639753.0A CN201710639753A CN107759602B CN 107759602 B CN107759602 B CN 107759602B CN 201710639753 A CN201710639753 A CN 201710639753A CN 107759602 B CN107759602 B CN 107759602B
Authority
CN
China
Prior art keywords
compound
alkyl
reaction
added
cancer
Prior art date
Legal status (The legal status 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 status listed.)
Expired - Fee Related
Application number
CN201710639753.0A
Other languages
Chinese (zh)
Other versions
CN107759602A (en
Inventor
赵玉军
陈德恒
严子琴
郭德祥
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Institute of Materia Medica of CAS
Original Assignee
Shanghai Institute of Materia Medica of CAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Institute of Materia Medica of CAS filed Critical Shanghai Institute of Materia Medica of CAS
Publication of CN107759602A publication Critical patent/CN107759602A/en
Application granted granted Critical
Publication of CN107759602B publication Critical patent/CN107759602B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The invention relates to a compound containing a conjugated allene structure, a pharmaceutical composition and application thereof, in particular to a compound represented by a general formula (I) or a salt thereof, a pharmaceutical composition thereof and application thereof as a BTK inhibitor and/or a B cell activation inhibitor for preventing or treating diseases related to B cell activity abnormity and/or BTK. The compound has better lethal effect on cancer cells such as lymph cancer, breast cancer, liver cancer, intestinal cancer, gastric cancer, lung cancer, cervical cancer and the like, and has potential for treating related cancers and autoimmune diseases.

Description

Compound containing conjugated allene structure, pharmaceutical composition and application thereof
Technical Field
The invention relates to a compound containing conjugated allene structure fragments and application thereof, in particular to a small molecular compound with a conjugated allene structure or salt thereof, a pharmaceutical composition thereof and application thereof as a B cell activation inhibitor for preventing or treating diseases related to abnormal B cell activity.
Background
The chemical structure of carbonyl conjugated alkene, carbonyl conjugated alkyne can form covalent bond with amino acid residue of cysteine, lysine and the like in protein, and is widely applied in the design and development of non-reversible small molecule drugs, small molecule drugs containing carbonyl conjugated alkene have wide application in clinic, for example, the following 7 approved commercially available drugs all contain carbonyl conjugated alkene fragment, EGFR kinase inhibitor Afatinib (Afatinib) and Osimetinib are both approved by the food and drug administration of the United states for treating non-small cell lung cancer, BTK kinase inhibitor Ibrutinib (Ibrutinib) also contains carbonyl conjugated alkene fragment, which is approved for treating lymphoma, 5- α -reductase inhibitor Dutasteride (Dutasteride) is mainly used for treating prostatic hypertrophy, male pattern alopecia, seborrheic alopecia, hereditary alopecia, while 5- α -reductase inhibitor Finasteride mainly contains carbonyl conjugated alkene fragment, Keaparic-kinase (Finarcoside) is mainly used for treating benign prostatic hyperplasia, the two inhibitors also contain carbonyl conjugated alkene-kinase, and the potential of reversible cysteine-conjugated alkyne-kinase for treating benign prostatic hyperplasia, kidney disease has been studied in the clinical stage of benign prostatic hyperplasia, kidney disease, and also has potential for treating benign prostatic hyperplasia.
Figure BDA0001365649630000011
WO2008039218a2, WO2008121742a2 and WO2010009342a2 disclose carbonyl-conjugated olefin-and carbonyl-conjugated alkyne-containing compounds represented by the general formula (a):
Figure BDA0001365649630000021
in the general formula (A), LaARepresents CH2O, NH or S, ArARepresents a substituted or unsubstituted aryl or a substituted or unsubstituted heteroaryl group, YARepresents an optionally substituted radical selected from the group consisting of alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, ZARepresents CO, OCO, NHCO, CS, etc., R7-AAnd R8-AIndependently represent H, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C1-C4 heteroalkyl, substituted or unsubstituted C3-C6 cycloalkyl, substituted or unsubstituted C2-C6 heterocycloalkyl, or R7-AAnd R8-ATaken together to form a bond, R6-ARepresents H, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C1-C4 heteroalkyl, C1-C6 alkoxyalkyl, C1-C8 alkylaminoalkyl, substituted or unsubstituted aryl, or the like.
WO2008060301a2 discloses carbonyl conjugated alkenes, carbonyl conjugated alkynes containing compounds comprising a 6-hydroxypurine backbone represented by general formula (B):
Figure BDA0001365649630000022
in the general formula (B), Q1BAnd Q2BIndependently selected from CX1B、CX2BAnd nitrogen, Q3BRepresents nitrogen or CH, X1BAnd X2BIndependently selected from hydrogen, (C1-C6) alkyl, cyano, halogen and the like, R1BSelected from hydrogen and (C1-C6) alkyl, yB represents 0 or an integer of 1-3, R2BAnd R3BIndependently selected from hydrogen and (C1-C6) alkyl, R4BSelected from alkyl, heterocycle, aryl and heteroAryl, etc., R5BSelected from alkyl, heterocyclic, substituted heterocyclic, and the like.
WO2007142755a2 discloses compounds represented by the general formula (C):
Figure BDA0001365649630000023
in the general formula (C), XCSelected from nitrogen and CR8C,R8CSelected from hydrogen, halogen, and substituted, unsubstituted alkyl, and the like, Q1CSelected from O, S, etc., ZCSelected from oxygen, sulfur and NY5C,Y5CSelected from hydrogen, substituted or unsubstituted alkyl, and the like, Q2C、Q3CAnd Q4CIndependently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, and the like, R2CSelected from hydrogen, substituted or unsubstituted alkyl, and the like, nCRepresents 0, 1,2, 3 or 4.
WO2003037890A2 discloses compounds represented by the general formula (D):
Figure BDA0001365649630000031
in the general formula (D), R1DSelected from hydrogen, substituted or unsubstituted alkyl,
Figure BDA0001365649630000032
Etc. R2DRepresents a substituted or unsubstituted alkyl group, a substituted or unsubstituted alkenyl group, a substituted or unsubstituted alkynyl group or the like, YDSelected from O, C-NO2And the group of S, ZDIs selected from
Figure BDA0001365649630000033
Etc. in which AD、DD、EDAnd MDEach independently represents CR12DN and N-oxide, R12DRepresents a group selected from hydrogen, halogen, amino, hydroxyl, cyano, etc., XDRepresents a group selected from O, C-NO2Radicals of S, etc., R6DRepresents a group selected from a substituted or unsubstituted cycloalkyl group, a substituted or unsubstituted heteroalkyl group, a substituted or unsubstituted aryl group, etc.; symbol a indicated by a dotted lineDRepresents any one of a single bond or a double bond, nDRepresents an integer selected from 0, 1 and 2.
CN102918040B discloses a compound represented by the general formula (E):
Figure BDA0001365649630000034
in the general formula (E), LErepresents-O-, -S-, -SO-, -SO2-、-NH-、-C(O)-、-CH2-O-、-O-CH2-、-CH2-or-CH (OH) -, ring 1 represents a4 to 7-membered cyclic group which may be substituted with 1 to 5 substituents each independently selected from a halogen atom, C1-4 alkyl, C1-4 alkoxy, cyano, C1-4 haloalkyl, C1-4 haloalkoxy, wherein when the number of substituents on ring 1 is 2, the substituents may form a4 to 7-membered cyclic group together with the atom constituting ring 1 to which they are bonded, and ring 2 represents a group which may be substituted with 1 to 3-K-R2Substituted 4-to 7-membered saturated heterocyclic ring, mERepresents 0, 1 or 2, nERepresents 0, 1,2, 3 or 4, R1ERepresents a halogen atom, a C1-4 alkyl group, a C1-4 alkoxy group, a C1-4 haloalkyl group or a C1-4 haloalkoxy group.
WO2013010868A1 discloses compounds represented by the general formula (F):
Figure BDA0001365649630000035
x in the general formula (F)FRepresents CH, O, N or S, YFRepresents C (R)6F) N, O or S, Z is CH, N or a bond, AFRepresents CH or N, B1FRepresents nitrogen or (CR)7F),B2FRepresents nitrogen or (CR)8F),B3FRepresents nitrogen or (CR)9F),B4FRepresents nitrogen or (CR)10F),R1FRepresents R11FC(O)、R12FS(O)、R13FSO2Or optionally substituted with R14FSubstituted C1-6 alkyl, R2FRepresents H, C1-3 alkyl or C3-7 cycloalkyl, R3FRepresents H, C1-6 alkyl or C3-7 cycloalkyl, R2FAnd R3FMay form, together with the N and C atoms to which they are attached, a C3-7 heterocycloalkyl group optionally substituted by one or more fluorine atoms, hydroxy, C1-3 alkyl, C1-3 alkoxy or oxo groups, R4FRepresents H or C1-3 alkyl, R5FRepresents H, halogen, cyano, C1-4 alkyl, C1-3 alkoxy, C3-C6 cycloalkyl, any alkyl group optionally substituted by 1 or more halogens, or R5FRepresents a C6-10 aryl group or a C2-C6 heterocycloalkyl group, R6FRepresents H or C1-3 alkyl, or R5FAnd R6FForm C3-7 cycloalkenyl or C2-C6 heterocycloalkenyl, each of which is optionally substituted with C1-3 alkyl or 1 or more halogens, R7FRepresents H, halogen and C1-3 alkoxy, R8FRepresents H and C1-3 alkyl, or R7FAnd R8FTo form C6-10 aryl or C1-9 heteroaryl, R9FAnd R10FRespectively represent H, halogen and C1-3 alkoxy, R11FIndependently selected from C1-6 alkyl, C2-6 alkenyl or C2-6 alkynyl, each alkyl, alkenyl or alkynyl being optionally substituted with one or more groups selected from hydroxy, C1-4 alkyl, C3-7 cycloalkyl, C1-4 alkylamino, di-C1-4 alkylamino, C1-3 alkoxy, C3-7 cycloalkoxy, C6-10 aryl, C3-7 heterocycloalkyl, or R11FRepresents C1-3 alkyl-C (O) -S-C1-3 alkyl or C1-5 heteroaryl optionally substituted by one or more groups selected from halogen, cyano, R12FAnd R13FIndependently represent C2-6 alkenyl or C2-6 alkynyl, both optionally substituted by one or more groups selected from hydroxy, C1-4 alkyl, C3-7 cycloalkyl, C1-4 alkylamino, di-C1-4 alkylamino, C1-3 alkoxy, C3-7 cycloalkoxy, C6-10 aryl, C3-7 heterocyclic aryl, or R12FAnd R13FIndependently represent C1-5 heteroaryl optionally substituted by one or more groups selected from halogen and cyano, R14FIndependently selected from halogen, cyano, or C3-7 cycloalkyl, C1-4 alkylamino, di-C1-4 alkylamino, C1-3 alkoxy, C3-7 cycloalkoxy, C6-10 aryl, C3-7 heteroaryl, and C3-7 heterocycloalkyl, optionally substituted with one or more groups selected from hydroxy, C1-4 alkyl, C3-7 cycloalkyl, C1-4 alkylamino, di-C1-4 alkylamino, C1-3 alkoxy, C3-7 cycloalkoxy, C6-10 aryl, C3-7 heteroaryl, and C2-6 alkenyl or C2-6 alkynyl.
WO2014025976A1 discloses compounds represented by the general formula (G):
Figure BDA0001365649630000041
in the general formula (G), wherein AGOne of the following structures is shown,
Figure BDA0001365649630000042
R1Grepresents N (R)3G)2Or H, CyGRepresents aryl or heteroaryl, each of which is represented by R2GSubstituted and optionally substituted by halogen, halogeno-C1-4 alkyl, C1-4 alkyl and C1-4 alkoxy, R2GSelected from the group consisting of optionally substituted halogen, halogenated C1-4 alkyl, C1-4 alkyl, R3G-S(O)mG-, cyano, -C (O) -N (R)3G)2Or L substituted by C1-4 alkoxyG-ArGC1-6 alkyl and C1-6 alkoxy, LGRepresents a chemical bond selected from O,>C(O)、-(CH2)nG-、-O-(CH2)nG-、-N(R3G)-、-N(R3G)-(CH2)nG-、-(CH2)nG-N(R3G)-、-C(O)-N(R3G)-、-C(O)-N(R3G)-(CH2)nG-、-N(R3G)-C(O)-N(R3G)-、-N(R3G)-C(O)-、-S(O)mG-N(R3G)-、-N(R3G)-S(O)mGA linking group of-etc., ArGRepresents a carbocyclic, heterocyclic or heteroaryl group, X1GRepresents a chemical bond or- (CH)2)nGA linking group of (A), YGRepresents C7-C10 spiro, monocyclic or bicyclic nitrogen-containing heterocycles, carbocycles, aryls optionally containing 0 to 1 ring nitrogen atom, each of which is substituted by one R4GSubstituted, R4GRepresents one of the following groups:
Figure BDA0001365649630000051
nGrepresents 1,2, 3 or 4, mGRepresents 0, 1 or 2, R3GRepresents H or C1-4 alkyl, R5GRepresents H, C1-4 alkoxy, C1-4 alkyl, C1-4 alkyl, C1-4 alkoxy, - (CH)2)nG-heterocycles and heterocycles, each heterocycle being optionally substituted by halogen, hydroxy or R3G-S(O)mG-substitution.
However, little research has been done on the biological activity and pharmacological properties of compounds containing conjugated dienyl structural fragments. Conjugated allenes differ from conjugated alkenes and alkynes in chemical structure and properties. Therefore, unlike compounds containing conjugated alkene, or conjugated alkyne moieties, the compounds containing conjugated allene moieties are not known for their biological activity and pharmacological properties, and are subject to uncertainty in all aspects of their properties.
Disclosure of Invention
The invention aims to develop a small molecular compound with a conjugated allene structure.
In order to achieve the aim, the invention firstly synthesizes a compound containing conjugated allene segments, and researches the activity of the compound in tumor cell strains. The results show that the compound containing the conjugated allene structure has a good lethal effect on cancer cells such as B cell lymphoma, breast cancer, liver cancer, intestinal cancer, gastric cancer, lung cancer, cervical cancer and the like for the first time, and the compound containing the conjugated allene structure has potential for treating related cancers and autoimmune diseases, so that the invention is completed.
One aspect of the present invention relates to a compound represented by the general formula (I):
Figure BDA0001365649630000061
in the general formula (I):
w is selected from the following groups:
Figure BDA0001365649630000062
Figure BDA0001365649630000063
Figure BDA0001365649630000064
preferably selected from
Figure BDA0001365649630000065
n is 0, 1 or 2;
m is 0, 1,2, 3 or 4;
preferably n + m is different from 0, more preferably n + m is 2,3 or 4;
l represents-O-, -CO-NH-, -NH-CO-, -O-CH2-、-CO-NH-CH2-、-NR6-、-NR6-CH2-、-CH2-NR6-, -S-, -CH (OH) -or-CH2-; preferably, L represents-O-or-NH-CO-;
R1represents a C6-C12 aryl or a 5-10 membered monocyclic or bicyclic heteroaryl group containing 1-4 heteroatoms selected from N, O, S; the aryl and heteroaryl groups are optionally substituted with 1 to 6 substituents selected from halogen, cyano, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy, amino, -NH (C1-C4 alkyl), -N (C1-C4 alkyl) (C1-C4 alkyl), -NHCO- (C1-C4 alkyl), -NHCO- (C1-C4 alkoxy), preferably optionally substituted with 1 to 2 substituents selected from halogen, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy and amino; the aryl group is preferably phenyl, and the heteroaryl group is preferably pyridyl, benzopyridyl or indolyl;
R2、R3and R4Each independently selected from hydrogen and C1-C4 alkyl, said C1-C4 alkyl being optionally substituted with 1-3 substituents selected from halogen, hydroxy, amino, C1-C4 alkoxy, -NH- (C1-C4 alkyl), -N (C1-C4 alkyl) (C1-C4 alkyl), -NHCO- (C1-C4 alkyl), -NHCO- (C1-C4 alkoxy), preferably, optionally substituted with 1-3 substituents selected from halogen, hydroxy, amino, C1-C4 alkoxy; preferably, R2、R3And R4Are all hydrogen;
or R2And R3Together with the carbon to which they are attached form a 4-8 membered ring, optionally containing 1-3 heteroatoms selected from N, O and S, and optionally substituted with 1-3 heteroatoms selected from halogen, hydroxy, amino, C1-C4 alkoxy, (C1-C4 alkyl) -NH2Substituent of-NH- (C1-C4 alkyl), -N (C1-C4 alkyl) (C1-C4 alkyl);
B1、B2、B3and B4Each independently represents N or-C (R)5) Preferably, B1、B2、B3And B4Up to two are N, more preferably up to one is N;
R5each independently selected from hydrogen, halogen, cyano, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy, amino, -NH- (C1-C4 alkyl), -N (C1-C4 alkyl) (C1-C4 alkyl), -NHCO- (C1-C4 alkyl) and-NHCO- (C1-C4 alkoxy), preferably each independently selected from hydrogen, halogen, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C4-C4 alkyl, halogen substituted C4-C4 alkoxy and amino, or two adjacent R, C, H, C, N-C-S-C5Together with the carbon atoms to which they are attached form a phenyl ring or a 5-6 membered heteroaromatic ring containing 1-2 heteroatoms selected from N, O and S;
R6each independently selected from hydrogen or C1-C4 alkyl, preferably from hydrogen, methyl and ethyl.
In one embodiment, the compound of formula (I) is a compound of formula (I-1):
Figure BDA0001365649630000071
wherein R is1、R2、R3、R4、B1、B2、B3、B4N, m, X and W are as defined for formula (I).
In one embodiment, the compound of formula (I) is a compound of formula (I-2):
Figure BDA0001365649630000072
wherein R is1、R2、R3、R4、B1、B2、B3、B4N, m, X and W are as defined for formula (I).
In one embodiment, the compound of formula (I) is a compound selected from the following formulae (I-3) to (I-7):
Figure BDA0001365649630000081
wherein, L and R1Is as defined in formula (I);
B2represents N or-C (R)5);
R5Each independently selected from hydrogen, halogen, cyano, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy, amino, -NH- (C1-C4 alkyl), -N (C1-C4 alkyl) (C1-C4 alkyl), -NHCO- (C1-C4 alkyl), and-NHCO- (C1-C4 alkoxy), preferably each independently selected from hydrogen, halogen, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy, and amino.
In one embodiment, the compound of formula (I) is selected from the following compounds:
Figure BDA0001365649630000082
Figure BDA0001365649630000091
Figure BDA0001365649630000101
Figure BDA0001365649630000111
Figure BDA0001365649630000121
Figure BDA0001365649630000131
Figure BDA0001365649630000141
Figure BDA0001365649630000151
Figure BDA0001365649630000161
Figure BDA0001365649630000171
Figure BDA0001365649630000181
another aspect of the present invention relates to a pharmaceutical composition comprising the above-described compound of the present invention or a salt thereof. The pharmaceutical composition may optionally further comprise pharmaceutically acceptable excipients which may be, for example, binders, fillers (diluents), disintegrants, suspensions, suspending agents, slow (controlled) release agents, lyoprotectants, coatings, enteric materials, lubricants (glidants, antiadherents), sweeteners, flavors, plasticizers, opacifiers, solubilizers, humectants, solvents, osmo-regulators, colorants, pigments, surfactants (emulsifiers), water-soluble matrices, fat-soluble matrices, oleaginous matrices, pore-forming agents, gels, preservatives, buffers, chelating agents, antioxidants and the like.
In one embodiment, the pharmaceutical composition is a BTK inhibitor.
In one embodiment, the pharmaceutical composition is a B cell activation inhibitor, in particular a B cell growth inhibitor.
In one embodiment, the pharmaceutical composition is a medicament for preventing or treating diseases associated with B cell activation, particularly B cell growth, and BTK. Another aspect of the present invention relates to a method for preventing and/or treating a disease associated with activated B cells, particularly B cell growth, and/or BTK, the method comprising administering to a subject in need thereof a therapeutically effective amount of the above-described compound of the present invention or a salt thereof. The subject is preferably a mammal, such as a human.
Another aspect of the present invention relates to the use of a compound of the present invention or a salt thereof as described above for the preparation of a medicament for the prevention and/or treatment of diseases associated with activated B cells, in particular B cell growth, and/or BTK.
In one embodiment, the disease associated with activated B cells and/or BTK is an allergic disease, an autoimmune disease, an inflammatory disease, a thromboembolic disease, or a cancer.
In one embodiment, the cancer is non-hodgkin's lymphoma, breast, liver, bowel, stomach, pancreas, lung, and cervix.
Another aspect of the present invention relates to a compound of the present invention or a salt thereof as described above or a pharmaceutical composition as described above for use in the prevention and/or treatment of diseases associated with activated B cells, in particular B cell growth, and/or BTK.
The present invention will be described in detail below.
In the present invention, halogen means fluorine, chlorine, bromine and iodine.
In the present invention, the C1-C4 alkyl group means a straight-chain or branched C1-C4 alkyl group including methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl and the like.
In the present invention, the C1-C4 alkoxy group means a straight or branched C1-4 alkoxy group including methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy, tert-butoxy and the like.
In the present invention, the halogen-substituted C1-C4 alkyl group means a group in which a C1-C4 alkyl group is substituted with 1 or 2 or more halogen atoms, and examples thereof include a fluoromethyl group, a chloromethyl group, a bromomethyl group, an iodomethyl group, a difluoromethyl group, a trifluoromethyl group, a 1-fluoroethyl group, a 2-fluoroethyl group, a pentafluoroethyl group, a 1-fluoropropyl group, a 2-fluoropropyl group, a 3-fluoropropyl group, a 1-chloropropyl group, a 2-chloropropyl group, a 3-chloropropyl group, a 1-fluorobutyl group, a 2-fluorobutyl group, a 3-fluorobutyl group, a4, 4, 4-trifluorobutyl group, and a 4-bromobutyl group.
In the present invention, the halogen-substituted C1-C4 alkoxy group means a group in which a C1-C4 alkoxy group is substituted with 1 or 2 or more halogen atoms, and examples thereof include trifluoromethoxy group, trichloromethoxy group, chloromethoxy group, bromomethoxy group, fluoromethoxy group, difluoromethoxy group, dimethoxy group, fluoromethoxy group, difluoromethoxy group, dibromomethoxy group, 2-fluoroethoxy group, 2,2, 2-trifluoroethylmethoxy group, 2,2, 2-trichloroethylmethoxy group, 3-chloropropoxy group, 2-fluoro-3-chloropropoxy group, 1-fluorobutoxy group, 2-fluorobutoxy group, 3-fluorobutoxy group and 3-chlorobutoxy group.
In the present invention, the C6-C12 aryl group means a 6-to 12-membered aromatic carbocyclic group, and examples thereof include phenyl, quinonyl, naphthalene-1, 4-diketonyl, naphthyl, anthryl, phenanthryl, azulenyl, acenaphthenyl, fluorenyl and the like.
In the present invention, the 5-10 membered monocyclic or bicyclic heteroaryl group containing 1-4 heteroatoms selected from N, O, S means an aromatic 5-7 membered monocyclic heterocyclic group and 8-10 membered bicyclic heterocyclic group containing 1-4 heteroatoms, for example, examples thereof include pyrrolyl, furyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furazanyl, oxadiazolyl, thiadiazolyl, thiazinyl, benzofuranyl, benzoisothiofuranyl, isoindolyl, benzo [ b ] thienyl, benzo [ c ] thienyl, benzimidazolyl, purinyl, indazolyl, benzoxazolyl, benzoisoxazolyl, benzothiazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, cinnolinyl and the like.
In the present invention, the 4-to 8-membered ring optionally containing 1 to 3 heteroatoms selected from N, O, S means a 4-to 8-membered saturated carbocyclic ring or a 4-to 8-membered saturated heterocyclic ring containing 1 to 3 heteroatoms selected from N, O, S, for example, examples thereof include a cyclobutane ring, a cyclopentane ring, a cyclohexane ring, a cycloheptane ring, a cyclooctane ring, an oxetane ring, a tetrahydrofuran ring, a tetrahydropyran ring, an oxepane ring, an acridine ring, a pyrrolidine ring, a piperidine ring, an azepane ring, an azocane ring, a tetrahydrothiophene ring, a1, 4-diazepane ring, a1, 3-dioxepane ring, a1, 4-dioxepane ring, a1, 5-diazacyclooctane ring, a homomorpholine ring, a1, 5-azacyclooctane ring and the like.
In the present invention, the heteroaromatic ring containing 1 to 2 heteroatoms selected from N, O and S means a 5-to 6-membered aromatic heterocycle containing 1 to 2 heteroatoms selected from N, O and S, and examples thereof include a pyrrole ring, a furan ring, an imidazole ring, a pyrazole ring, a pyridine ring, a pyrimidine ring, a pyrazine ring, a pyridazine ring, a thiophene ring, an oxazole ring, an isoxazole ring, a thiazole ring, and an isothiazole ring.
[ isomers ]
In the present invention, unless otherwise specified, the structure of the present invention includes all isomers, for example, alkyl groups include straight-chain alkyl groups and branched-chain alkyl groups, further, geometrical isomers (E-form, Z-form, cis-form, trans-form) in double bonds, rings, fused rings, optical isomers (R and S-forms, α and β forms, enantiomers, diastereomers) resulting from the presence of asymmetric carbon atoms and the like, optically active forms (D, L, D, L-forms), optical isomers, mixtures thereof in any ratio, and racemic mixtures thereof are included in the present invention.
In the present invention, unless otherwise specified, the meanings of the symbols are those familiar to those skilled in the art, for example
Figure BDA0001365649630000201
Indicating that the chemical bond is directed to the front side of the paper (β configuration);
Figure BDA0001365649630000202
indicating that the chemical bond is directed toward the back of the paper (α configuration) "/" indicating that the chemical bond may be directed toward the front or back, or a mixture of α and β configurations in any proportion thereof.
The compound represented by the general formula (I) can be converted into a corresponding salt by a known method. The salt is preferably water soluble. Suitable salts include alkali metal (potassium, sodium, etc.) salts, alkaline earth metal (calcium, magnesium, etc.) salts, and ammonium salts, pharmaceutically acceptable salts of organic amines (tetramethylammonium, triethylamine, methylamine, cyclopentylamine, benzylamine, phenethylamine, piperidine, monoethanolamine, diethanolamine, tris (hydroxymethyl) aminomethane, lysine, arginine, N-methyl-D-glucamine, etc.), acid adduct salts (inorganic acid salts (hydrochloride, hydrobromide, hydroiodide, sulfate, phosphate, nitrate, etc.), organic acid salts (acetate, trifluoroacetate, lactate, tartrate, oxalate, fumarate, maleate, benzoate, citrate, methanesulfonate, ethanesulfonate, benzenesulfonate, toluenesulfonate, isethionate, glucuronate, gluconate, etc.)), and the like.
The compounds represented by the general formula (I) and salts thereof may also exist in the form of solvates thereof. The solvate is preferably low in toxicity and water-soluble. As a suitable solvate, for example, a solvate of a solvent such as water or an alcohol (e.g., ethanol) may be cited, and a plurality of crystal forms may exist.
In addition, the compounds represented by the general formula (I) and salts thereof may also exist in the form of prodrugs thereof. The prodrug is a compound which can be converted into a compound represented by the general formula (I) in vivo by reaction with an enzyme and/or gastric acid or the like. Examples of the prodrug of the compound represented by the general formula (I) include compounds in which a hydroxyl group of the compound represented by the general formula (I) is acylated, alkylated, phosphorylated, borated (for example, compounds in which a hydroxyl group of the compound of the present invention is acetylated, palmitoylated, propionylated, pivaloylated, succinylated, fumarylated, malonylated, dimethylaminocarbonylated, and the like); compounds in which the carboxyl group of the compound represented by the general formula (I) is esterified or amidated (for example, compounds in which the carboxyl group of the compound represented by the general formula (I) is esterified with ethyl, isopropyl, phenyl, hydroxymethyl, dimethylaminomethyl, pivaloyloxymethyl, ethoxycarbonyloxyethyl, peptide, (5-methyl-2-oxo-1, 3-dioxopentyl) methyl, cyclohexyloxycarbonylethyl, formamido, etc.); and the like. These compounds can be prepared by known methods. The prodrug of the compound represented by the general formula (I) may be either a hydrate or a non-hydrate. The prodrug of the compound represented by the general formula (I) may be a compound represented by the general formula (I) under physiological conditions as described in "Guangchua bookstore, 1990, development of pharmaceuticals", volume 7, molecular design, pp 163-198 ".
Further, the compound represented by the general formula (I) may be an isotope (e.g., deuterium, tritium, etc.),11C、13C、14C、13N、15N、15O、18O35S、18F、36Cl、123I、125I) And the like.
The isomers (including optical isomers, tautomers and mixtures thereof in any ratio), solvates and prodrugs of the compounds represented by the general formula (I) and salts thereof are included in the present invention as equivalent alternatives. The optical isomers in the present invention include not only compounds with 100% purity but also other optical isomers less than 50%.
[ Process for producing the Compound of the present invention ]
The compound represented by the general formula (I) of the present invention can be prepared by one of the following methods. The method comprises the following steps: compound a1 can be prepared by the condensation reaction of intermediate a2 and intermediate A3, as shown in scheme 1. The condensation reaction conditions may be the synthesis conditions reported in Angew. chem. int. Ed.2014,53, 7491-7494. The reaction conditions after modification and optimization based on the above reaction conditions can also be used as reaction conditions for preparing A1 by condensation of A2 and A3.
Figure BDA0001365649630000211
Reaction scheme 1
In reaction scheme 1, L, R1、R2、B1、B2、B3、B4N, m and W are as defined for formula (I), R3And R4Are both hydrogen.
The second method comprises the following steps: compound a1 can be prepared by condensation reaction of intermediate a2 and intermediate B1 as shown in scheme 2. The condensation reaction conditions may be those reported in Journal of Organic Chemistry 2002,67, 6575-6578. The modified and optimized reaction conditions based on the reaction conditions can also be used as the reaction conditions for preparing A1 by condensation of A2 and B1.
Figure BDA0001365649630000221
Reaction formula 2
In reaction scheme 2, L, R1、R2、R3、R4、B1、B2、B3、B4N, m and W are as defined for formula (I).
The third method comprises the following steps: compound a1 can be prepared by coupling reaction of intermediate a2, intermediate B2 and carbon monoxide over a catalyst [ e.g. tetrakis (triphenylphosphine) palladium) ]. The coupling reaction conditions may be Organic Letters,2009, Vol.11, No.13, 2900-; reaction conditions reported in Journal of Organic Chemistry,2002,67, 6575-6578. The reaction conditions after modification and optimization based on the above reaction conditions may also be used as the reaction conditions for preparing a 1.
Figure BDA0001365649630000222
Reaction formula 3
In reaction scheme 3, L, R1、R2、R3、R4、B1、B2、B3、B4N, m and W are as defined for formula (I).
The method four comprises the following steps: compound a1 can be synthesized as shown in equation 4:
(1) under the action of alkali, A1 reacts with chloroacetyl chloride to obtain B3;
(2) b3 reacts with triphenyl phosphine to obtain B4;
(3) b4 can generate a Wittig reagent B5 under the action of alkali;
(6) reaction of B5 with acid chloride B6 affords a 1.
The reaction conditions in the above steps may be those reported in Tetrahedron Letters 56(2015) 3281-3284. Wherein the base is selected from organic and inorganic bases, such as triethylamine, diisopropylethylamine, N-methylmorpholine, sodium ethoxide, and mixtures thereof,
Sodium methoxide, sodium hydroxide, potassium hydroxide, sodium tert-butoxide, potassium tert-butoxide, lithium diisopropylamide, butyl lithium and the like. The reaction conditions after modification and optimization based on the above reaction conditions may also be used as the reaction conditions for preparing a 1.
Figure BDA0001365649630000231
Reaction formula 4
In reaction formula 4, L, R1、R2、R3、R4、B1、B2、B3、B4N, m and W are as defined for formula (I).
The key intermediate A2 of the compound of the present invention can be a commercially available product or can be synthesized by or by referring to methods in the prior art, for example, the synthesis can be one of the following methods.
The method comprises the following steps: intermediate a2 was synthesized as shown in equation 5:
(1) boric acid A5 or boric acid ester A6 and bromide A7 are subjected to Suzuki coupling reaction to obtain an intermediate A4; the Suzuki coupling reaction conditions can be reaction conditions disclosed in Chemical Reviews 1995,95(7), 2457-2483 and org.processres.Dev.2011,15, 1018-1026, and the modified and optimized reaction conditions based on the reaction conditions can also be used as the reaction conditions for preparing A2;
(2) deprotection reactions were carried out to remove the Cbz protecting group contained in a4 to afford intermediate a 2. For example, the deprotection method described in Protective Grups in Organic Synthesis, Wiley, New York,1999 can be used, or appropriate modifications and optimizations can be made based on literature procedures.
Figure BDA0001365649630000232
Reaction formula 5
In reaction formula 5, R1、L、B1、B2、B3、B4N, m and W are as defined for formula (I).
The second method comprises the following steps: intermediate a2 can be prepared by the following route, as shown in scheme 6:
(1) intermediate A10 and compound A9 are connected through a Mitsunobu reaction to obtain intermediate A8; the Mitsunobu reaction can adopt the method reported in Strategic Applications of Named Reactions in organic Synthesis,2005, Elsevier Inc press and ACS Med. chem. Lett.2016,7,198-203, and can be modified and optimized appropriately based on the literature methods;
(2) deprotection to remove the Boc group of A8 gave A2. The deprotection reaction can be carried out by the method of deprotecting groups described in Protective Grups in Organic Synthesis, Wiley, New York,1999, or can be modified and optimized appropriately based on literature methods.
Figure BDA0001365649630000241
Reaction formula 6
In reaction formula 6, R1、L、B1、B2、B3、B4N, m and W are as defined for formula (I).
Intermediates a5 and a6 can be commercially available products or synthesized using or referencing methods known in the art, for example, they can be synthesized by the following route, as shown in scheme 7: the halogenated aromatic hydrocarbon A11 is transformed and metallized by using n-butyl lithium, and then reacts with triisopropyl borate to obtain boric acid A5 by acidification; or coupling the trifluoromethyl sulfonate A12 of halogenated aromatic ring and aryl alcohol with bis-pinacol borate under the catalysis of a palladium catalyst to obtain the pinacol borate A6. The synthesis methods of the boric acid A5 and the borate A6 can be the methods disclosed in J.org.chem.2002,67,5394-5397 and J.org.chem.1995,60,7508-7510, and can be modified and optimized appropriately based on the literature methods.
Figure BDA0001365649630000242
Reaction formula 7
In reaction formula 7, R1、L、B1、B2、B3And B4Is the same as in formula (I), and X is Br, I or OTf.
Intermediate a7 may be a commercially available product or synthesized using or referencing methods known in the art, for example, it may be synthesized by the following route, as shown in scheme 8:
(1) condensing 2- (3-chloropyrazinyl) methylamine with compound C1(N-Cbz protected α -amino acid, β -amino acid or γ -amino acid) to give amide C2;
(2) amide C2 in phosphorus oxychloride (POCl)3) Can be dehydrated and cyclized under the action of other reagents to obtain a cyclized product C3;
(3) brominating the cyclization product C3 by N-bromosuccinimide (NBS) to obtain a bromination product C4;
(4) ammonolysis of the brominated product C4 with aqueous ammonia affords compound C5, which is part of a representative a7 structure.
The above-mentioned amide condensation, cyclization, bromination and aminolysis reactions can be prepared by the method described in Comprehensive organic transformations, A Guide to Functional Group Preparations, 2 nd edition (RichardC. Larock, John Wiley & Sons Inc,1999), or by appropriate modification and combination of the methods shown in the examples. The synthetic route can also adopt the reaction conditions in the patent WO2013/010868, and can also be appropriately modified and optimized on the basis of the literature method.
Figure BDA0001365649630000251
Reaction formula 8
In the reaction formula 8, n, m and W are as defined in the general formula (I).
Intermediate a10 may be a commercially available product or synthesized using or referencing methods known in the art, for example, it may be synthesized by the following route, as shown in scheme 9: boronic acid a5, or boronic esters a6 and a13, were subjected to Suzuki coupling to afford intermediate a 10. The Suzuki coupling reaction can be carried out by the methods disclosed in Chemical Reviews 1995,95(7): 2457-2483 and documents such as org. Process Res. Dev.2011,15, 1018-1026, and can be appropriately modified and optimized on the basis of the documents.
Figure BDA0001365649630000261
Reaction formula 9
In reaction formula 9, R1、L、B1、B2、B3、B4And W is as defined for formula (I), and X is Br, I or OTf.
A13 is aryl ring compound substituted by halogeno or trifluoromethyl sulfonate, and the aryl rings are 5-6 membered monocyclic heterocycle or 8-10 fused ring heterocycle compound. A13 can be a commercially available product or can be synthesized by methods known in the art, for example, according to Comprehensive Heterocyclic Chemistry III. Editors-in-Chief AlanR.Katritzky, Christopher A.Ramsden, Eric F.V.Scriven, and Richard J.K.Taylor, Elsevier Ltd. Press, 2008 and synthetic methods in the literature cited in this document.
Drawings
Fig. 1 is a graph showing the cellular activities of ibrutinib and compound 4 according to the present invention measured in B-cell lymphoma Raji cell lines.
Detailed Description
The present invention will be described in detail below with reference to examples, but the present invention is not limited to these examples.
General purification of compounds preparative chromatographic conditions: the purification instrument is used for preparing high performance liquid phase by Shimadzu semi-preparationChromatography (by HPLC, model LC-20AR) on a SunAire Prep C18OBR columnTM5 uM. The mobile phases used for the HPLC work were water (0.1% trifluoroacetic acid) and acetonitrile (0.1% trifluoroacetic acid), the mobile phase gradient being a 1% increase in the proportion of acetonitrile per minute. During purification, the initial acetonitrile ratio is changed correspondingly according to the property of the compound.
1H NMR represents hydrogen spectrum of hydrogen atom signal spectrum measured by a nuclear magnetic resonance apparatus.1H NMR immediately following in brackets, CDCl3Representing the dissolution sample solvent as deuterated chloroform, MeOD-d4 representing the dissolution sample solvent as tetradeuterated methanol, DMSO-d6Representative of the dissolution sample solvent is hexadeuterodimethylsulfoxide. 400MHz represents the frequency of the electromagnetic waves used by the NMR spectrometer used for the test.
ESI-MS represents the mass spectrometer instrument using a probe in electrospray ionization (ESI) mode. [ M + H ]]+The latter figure is the theoretical value of mass-to-charge ratio after ionization of the species.
The specific rotation was measured by a Rudolph Autopol VI polarimeter, model INS-QC-76.
CH2Cl2Represents dichloromethane, MeOH represents methanol, Cbz represents a benzyloxycarbonyl protecting group, and Boc represents a tert-butoxycarbonyl protecting group.
The names of compounds used in this specification are generally named according to IUPAC nomenclature, and also according to common names.
Preparative example 1 synthesis of boronic acid, boronic ester:
the method comprises the following steps: synthesis of N- (2-pyridyl) 4- (4,4,5,5-tetramethyl- [1,3,2] dioxaboronyl) -benzamide (C007)
Figure BDA0001365649630000271
Step one, synthesizing p-bromobenzoyl chloride: in a 250mL glass flask, p-bromobenzoic acid (3g,15mmol), N, N-dimethylformamide (DMF, 0.4mL) and dichloromethane (CH) were added2 Cl 250 mL). Cooling the reaction solution to zero degree with ice water bath, adding thionyl chloride (SOCl)2,1.32mL,18mmol). The reaction was warmed to room temperature and stirred overnight. Evaporation to dryness was carried out by rotary evaporation and the crude product was purified by co-evaporation with toluene (30 mL. times.2) to give a colorless liquid which was used directly in the next reaction.
Step two, synthesizing N- (2-pyridyl) p-bromobenzamide (C004): all colorless liquid obtained in the previous step was put into a 250mL glass flask and dissolved in dry tetrahydrofuran (THF, 20 mL). This solution was transferred to a 250mL flask containing 4-N, N-dimethylaminopyridine (DMAP, 0.183g,1.5mmol), 2-aminopyridine (1.41g,15mmol), diisopropylethylamine (DIEA, 3.2mL,18mmol) and dry tetrahydrofuran (30 mL). The mixed solution was heated to 50 ℃ and stirred overnight. Tetrahydrofuran was evaporated by rotary evaporation and the residue was dissolved in ethyl acetate (300 mL). The organic phase was extracted twice with hydrochloric acid solution (0.2N,100mL) and twice with aqueous sodium hydroxide solution (0.2N,100 mL). Finally, the organic phase was washed with saturated brine, dried and concentrated to give the title compound C004 as a solid (2.67 g). The product was used directly in the next reaction without further purification.1H NMR(CDCl3400MHz) 8.37(d, J-8.40 Hz,1H),8.30-8.26(m,1H),7.81(d, J-8.40 Hz,2H),7.80-7.72(m,1H),7.64(d, J-8.40 Hz,2H),7.21-7.06(m,1H), ESI-MS theoretical calculation C12H10 79BrN2O[M+H]+277.0; the experiment found that 277.0.
Step three: synthesis of N- (2-pyridyl) 4- (4,4,5,5-tetramethyl- [1,3,2] dioxolane boryl) benzamide (C007): in a 250mL glass flask, C004(960mg,3mmol), bis-pinacolato borate (1.5g,6.0mmol), potassium acetate (KOAc, 1.2g,12mmol) and dry 1, 4-dioxane (20mL) were added. The reaction solution was deoxygenated, and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (210mg,0.3mmol) was added, and deoxygenated again. The reaction solution was heated to 90 ℃ and stirred overnight. The reaction was quenched with 50mL of water and extracted three times with ethyl acetate (50 mL). The combined organic phases were washed with saturated brine, dried, concentrated and purified over silica gel column. Purification afforded the title compound, C007, as a solid (925 mg).
1H NMR(CDCl3,400MHz):8.86(s,br,1H),8.42(d,J=8.39Hz,1H),8.29(d,J=4.62Hz,1H),793(s,4H),7.81-7.74(m,1H),7.09(dd, J ═ 6.78,5.37Hz,1H),1.37(s, 12H). Theoretical calculation of ESI-MS C18H22BN2O3[M+H]+325.2; the test result is 325.3.
The second method comprises the following steps: synthesis of N- (2- (4-trifluoromethyl) pyridyl) 4- (4,4,5,5-tetramethyl- [1,3,2] dioxaboronyl) -benzamide (C012)
Figure BDA0001365649630000281
Step one, synthesizing p-bromobenzoyl chloride: in a 250mL glass flask, p-bromobenzoic acid (3g,15mmol), N, N-dimethylformamide (0.4mL) and dichloromethane (50mL) were added. The reaction solution was cooled to zero degrees with an ice-water bath, and thionyl chloride (1.32mL,18mmol) was added. The reaction was warmed to room temperature and stirred overnight. Evaporation to dryness was carried out by rotary evaporation and the crude product was purified by co-evaporation with toluene (30 mL. times.2) to give a colorless liquid which was used directly in the next reaction.
Step two, synthesizing N- (2- (4-trifluoromethyl) pyridyl) p-bromobenzamide (C009): all colorless liquid obtained in the above reaction step was placed in a 250mL glass flask and dissolved in dry tetrahydrofuran (20 mL). This solution was transferred to a 250mL flask containing 4-N, N-dimethylaminopyridine (0.183g,1.5mmol), 2-amino-4-trifluoromethylpyridine (1.41g,15mmol), diisopropylethylamine (3.2mL,18mmol) and dry tetrahydrofuran (30 mL). The mixed solution was heated to 50 ℃ and stirred overnight. Tetrahydrofuran was evaporated by rotary evaporation and dissolved in ethyl acetate (300 mL). The organic phase was extracted twice with hydrochloric acid solution (0.2N,100mL) and twice with aqueous sodium hydroxide solution (0.2N,100 mL). Finally, the organic phase was washed with saturated brine, dried and concentrated to obtain the objective compound C009 as a solid (3.93 g). The product was used directly in the next reaction without further purification.
1H NMR(CDCl3400MHz) 8.99(s,1H),8.67(s,1H),8.39(d, J ═ 5.00Hz,1H),7.80(d, J ═ 8.35Hz,2H),7.64(d, J ═ 8.35Hz,2H),7.29(d, J ═ 4.75Hz, 1H). Theoretical calculation of ESI-MS C13H9 79BrF3N2O[M+H]+345.0; experimental survey345.1 percent.
Step three, synthesizing N- (2- (4-trifluoromethyl) pyridyl) 4- (4,4,5,5-tetramethyl- [1,3,2] dioxaboronyl) benzamide (C012): in a 250mL glass flask, C009(1.70g,5mmol), bis-pinacolato borate (2.54g,10.0mmol), potassium acetate (2.0g,20mmol) and dry 1, 4-dioxane (30mL) were added. The reaction solution was deoxygenated, and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (350mg,0.5mmol) was added, and deoxygenated again. The reaction solution was heated to 90 ℃ and stirred overnight. The reaction was quenched with 50mL of water and extracted three times with ethyl acetate (50 mL). The organic phases were combined, washed with saturated brine, dried, concentrated and purified by means of a silica gel column to give the objective compound C012 as a viscous liquid (1.65g), which was gradually solidified by leaving it in a refrigerator at-20 ℃.
1H NMR(CDCl3400MHz) 9.00-8.90(br,1H),8.73(s,1H),8.47(d, J-5.08 Hz,1H),8.00-7.90(m,2H),7.31(d, J-5.08 Hz,1H),1.37(s, 12H). Theoretical calculation of ESI-MS C19H21BF3N2O3[M+H]+393.2; the experiment found that 393.3.
The third method comprises the following steps: synthesis of N- (2- (4-trifluoromethyl) pyridyl) 2-fluoro-4- (4,4,5,5-tetramethyl- [1,3,2] dioxaboronyl) benzamide (C019)
Figure BDA0001365649630000291
Step one, synthesizing 2-fluoro-4-bromobenzoyl chloride: in a 250mL glass flask, 2-fluoro-4-bromobenzoic acid (3.28g,15mmol), N, N-dimethylformamide (0.4mL) and dichloromethane (50mL) were added. The reaction solution was cooled to zero degrees with an ice-water bath, and thionyl chloride (1.32mL,18mmol) was added. The reaction was warmed to room temperature and stirred overnight. Evaporation to dryness was carried out by rotary evaporation and the crude product was purified by co-evaporation with toluene (30 mL. times.2) to give a colorless liquid which was used directly in the next reaction.
Step two, synthesizing N- (2- (4-trifluoromethyl) pyridyl) 2-fluoro-4-bromobenzamide (C016): all colorless liquid obtained in the above reaction step was placed in a 250mL glass flask and dissolved in dry tetrahydrofuran (20 mL). This solution was transferred to a 250mL flask containing 4-N, N-dimethylaminopyridine (0.183g,1.5mmol), 2-amino-4-trifluoromethylpyridine (2.43g,15mmol), diisopropylethylamine (3.2mL,18mmol) and dry tetrahydrofuran (30 mL). The mixed solution was heated to 50 ℃ and stirred overnight. Tetrahydrofuran was evaporated by rotary evaporation and dissolved in ethyl acetate (300 mL). The organic phase was extracted twice with hydrochloric acid solution (0.2N,100mL) and twice with aqueous sodium hydroxide solution (0.2N,100 mL). Finally, the organic phase was washed with saturated brine, dried and concentrated to give the objective compound C016 as a solid (3.51 g). The product was used directly in the next reaction without further purification.
1H NMR(CDCl3400MHz) 9.20-9.10(br,1H),8.67(s,1H),8.51(d, J-5.10 Hz,1H),8.06(t, J-8.39 Hz,1H),7.51(d, J-8.45 Hz,1H),7.44(d, J-11.56 Hz,1H),7.33(d, J-5.01 Hz, 1H). Theoretical calculation of ESI-MS C13H8 79BrF4N2O[M+H]+363.0; the test result is 363.1.
Step three, N- (2- (4-trifluoromethyl) pyridyl) 2-fluoro-4- (4,4,5,5-tetramethyl- [1,3,2] dioxaboronyl) benzamide (C019): in a 250mL glass flask, C016(1.81g,5mmol), bis-pinacolato borate (2.54g,10.0mmol), potassium acetate (2.0g,20mmol) and dry 1, 4-dioxane (30mL) were added. The reaction solution was deoxygenated, and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (350mg,0.5mmol) was added, and deoxygenated again. The reaction solution was heated to 90 ℃ and stirred overnight. The reaction was quenched with 50mL of water and extracted three times with ethyl acetate (50 mL). The organic phases were combined, washed with saturated brine, dried, concentrated and purified by silica gel column to give the objective compound C019 as a solid (2.46 g).
1H NMR(CDCl3400MHz) 9.40-9.22(br,1H),8.70(s,1H),8.56-8.40(m,1H),8.14(t, J ═ 6.76Hz,1H),7.74(d, J ═ 7.30Hz 1H),7.63(d, J ═ 12.34Hz,1H),7.36-7.22(m,1H),1.36(s, 12H). Theoretical calculation of ESI-MS C19H20BF4N2O3[M+H]+411.2; the test result is 444.3.
The method four comprises the following steps: synthesis of 2- (4-phenoxyphenyl) -4,4,5, 5-Tetramethyl- [1,3,2] dioxaborolan (4,4,5, 5-tetramethylyl-2- (4-phenoxy-phenyl) - [1,3,2] dioxaborolane, A)
Figure BDA0001365649630000301
4-Bromobiphenyl ether (6.2g,25.0mmol) and isopropanol pinacol borate (10.3mL,50mmol) were dissolved in dry tetrahydrofuran, cooled well to-78 ℃ under nitrogen, n-butyllithium (2.5M,18mL,45mmol) was added dropwise, stirred for 6 hours after the addition was completed, and slowly warmed to-40 ℃ for 12 hours. The reaction solution was added to a saturated ammonium chloride solution, and extracted three times with ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, evaporated to dryness by rotary evaporation and purified on a column to give the desired compound 5.4g with a yield of 72%.1H NMR(CDCl3,400MHz):7.78(d,J=8.5Hz,2H),7.35(t,J=8.0Hz,2H),7.13(t,J=7.3Hz,1H),7.0 3(d,J=8.3Hz,2H),6.98(d,J=8.6Hz,2H),1.34(s,12H)。
Preparation example 2 Synthesis of Key intermediate N-Cbz- (S) -3- (1-bromo, 8-aminoimidazo [1,5-a ] pyrazinyl) substituted-2-pyrrolidines (S8)
Figure BDA0001365649630000302
2- (3-Chloropyrazin-2-yl) methanamine (S4)
Step one, synthesizing 2- (3-chloropyrazinyl) methyl bromide (S2): 2-chloro-3-methylpyrazine (S1,7.72g,60mmol) and N-bromosuccinimide (NBS, 14.87g,84mmol) were dissolved in carbon tetrachloride (CCl)4100 mL). Adding benzoyl peroxide (Bz) in one time2O21.45g,6mmol) and the reaction was refluxed overnight. Then, the reaction solution was cooled to room temperature, filtered to remove the solid, and the solid was washed with carbon tetrachloride to obtain CCl4And (3) solution. The carbon tetrachloride solutions were combined and concentrated to give 13.27g of a mixture containing S2, which was used in the next reaction without purification.
Step two, synthesizing 2- (3-chloropyrazinyl) methyl-substituted-2-isoindole-1, 3-dione (S3): 13.27g of the mixture containing S2 in the previous step was dissolved in anhydrous N, N-dimethylformamide (150 mL). To the solution was added potassium phthalimide (22.2g), and the solution was heated to 60 ℃ for reaction overnight. After the reaction was completed, N-dimethylformamide was distilled off by a rotary evaporator, and water and methylene chloride were added. The aqueous phase was extracted with dichloromethane. The organic phases were combined, dried and concentrated to give a mixture containing S3. The mixture was used in the next reaction without further purification.
Step three, synthesizing 2- (3-chloropyrazinyl) methylamine: the mixture containing S3 in the previous step was placed in a 250mL glass bottle, and 100mL of methanol was added and mixed. Add 85% hydrazine hydrate (H) to the flask2N-NH2)6.6g, and the reaction solution was heated to 60 ℃ to react for 2 hours. Cooling, the solid was filtered on a Buchner funnel, the filter cake was washed with methanol (500mL), the resulting methanol solution was concentrated and the resulting mixture was dissolved in dichloromethane (300 mL). The mixture was filtered again through a Buchner funnel, the filter cake was washed with dichloromethane, and the resulting dichloromethane solution was concentrated and purified with a silica gel column to give 3-chloro-2-pyrazinylmethylamine (S4) (3.53g, 21% overall yield over 3 steps).1HNMR(CDCl3,400MHz):8.48(d,J=2.31Hz,2H),8.26(d,J=2.31Hz,2H),4.14(s,3H)。
N- (3-Chloropyrazin-2-ylmethyl) N-Cbz-prolinamide (S5)
Step four, S4(0.90g,7.3mmol), N-Cbz proline (2.74g,11mmol) and dichloromethane (20mL) were added to a 100mL glass vial. The reaction flask was cooled with ice water and then triethylamine (Et) was added3N, 4mL,38mmol) and stirred for 10 min. 2- (7-Benzotolyltriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (HATU,3.33g,8.8mmol) was added in one portion and stirred at room temperature overnight. The reaction was quenched with water, the aqueous phase was extracted with dichloromethane, and the organic phases were combined. The organic phase was washed with saturated brine, dried, concentrated and purified by silica gel column to give the objective compound S5 as an oily liquid (4.35 g).1HNMR(CDCl3400MHz) 8.40-8.24(m,1H),8.28(s,1H),7.46-7.00(m,5H),5.30-4.95(m,2H),4.80-4.30(m,3H),3.75-3.40(m,2H),2.40-2.10(m,2H),2.10-1.75(m, 2H). Theoretical calculation of ESI-MS C18H19 35ClN4NaO3[M+Na]+397.1; the test result is 397.1.
N-Cbz- (S) -3- (8-chloroimidazo [1,5-a ] pyrazinyl) substituted-2-pyrrolidines (S6)
Step five, in a 250mL glass vial, S5(8.64g,23.1mmol),1, 3-dimethyl-2-imidazolidinone (7.5mL,69.3mmol) and dry acetonitrile (50mL) were added. Cooling the reaction flask and the reaction solution in the flask to 0 deg.C, and adding phosphorus oxychloride (POCl)313.5mL,92.4mmol) and warmed to 60-65 deg.c for reaction overnight. Finally, the reaction solution was poured into 25% aqueous ammonia (50mL), and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried, concentrated and purified by silica gel column to give the title compound S6 as a white solid (5.89 g).
1H NMR(CDCl3400MHz) 8.15(d, J ═ 4.40Hz,0.5H),7.79(s,0.5H),7.68(s,0.5H),7.62-7.54(m,0.5H),7.36-7.16(m,3H),7.15-7.05(m,1H),6.96(d, J ═ 5.65Hz,0.5H),6.73(d, J ═ 7.54Hz,1H),5.32-5.23(m,0.5H),5.15(t, J ═ 6.29Hz,0.5H),5.06(d, J ═ 12.26Hz,0.5H),4.96(d, J ═ 11.89Hz,1H),4.64(d, J ═ 11.89, 0.5H), 3.80H, 3.5H, 3.58H, 2.2H), 2.15-1H (m,2H), 2.5H, 1H, 2H, 1H, 15(d, 2H). Theoretical calculation of ESI-MS C18H18 35ClN4O2[M+H]+357.1; the test result is 357.3.
N-Cbz- (S) -3- (1-bromo, 8-chloroimidazo [1,5-a ] pyrazinyl) substituted-2-pyrrolidines (S7)
Step six, in a 250mL glass bottle, S6(5.89g,16.5mmol) and dry N, N-dimethylformamide (40mL) were added. The flask and the reaction solution in the flask were cooled to 0 ℃ and N-bromosuccinimide (3.22g,18.2mmol) was added. The reaction was slowly warmed to room temperature and reacted overnight. The reaction was quenched with saturated sodium thiosulfate, the aqueous phase was extracted with ethyl acetate, and the organic phases were combined and washed with saturated brine, dried, concentrated, and purified by silica gel column to give the title compound S7 as a yellow solid (3.19 g).
1H NMR(CDCl3400MHz) 8.21(d, J-4.84 Hz,0.5H),7.41(d, J-4.63 Hz,0.5H),7.39-7.14(m,5H),6.95-6.80(m,1H),5.25-4.90(m,2.6H),4.68(d, J-11.44 Hz,0.4H),3.80-3.56(m,2H),2.60-2.20(m,3H),2.10-1.90(m, 1H). Theoretical calculation of ESI-MS C18H17 79Br35ClN4O2[M+H]+435.0; 435.1 is measured.
N-Cbz- (S) -3- (1-bromo, 8-aminoimidazo [1,5-a ] pyrazinyl) substituted-2-pyrrolidines (S8)
Step seven, into a 25mL thick-walled glass vial, S7(0.5g,1.1mmol) and isopropanol (16mL) were added. To the solution was added 4mL of concentrated ammonia, then the tube was closed and heated in an oil bath at 80 ℃ overnight. The reaction solution was cooled and then concentrated, and the resulting mixture was purified by a silica gel column to obtain the objective compound S8 as a white solid (137 mg).
1H NMR(CDCl3400MHz) 7.60(d, J-4.82 Hz,0.5H),7.40-7.10(m,4.5H),7.04-6.92(m,1H),6.88(d, J-6.37 Hz,0.6H),6.69(d, J-4.71 Hz,0.4H),5.20-4.90(m,2.6H),4.75(d, J-11.90 Hz,0.4H),3.80-3.50(m,2H),2.40-2.10(m,3H),2.06-1.86(m, 1H). Theoretical calculation of ESI-MS C18H19 79BrN5O2[M+H]+416.1; the experiment found that 416.3.
EXAMPLE 1 Synthesis of the Compound (S) -N- (pyridin-2-yl) para- (3- (1- (2, 3-butynoyl) pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (1)
Figure BDA0001365649630000331
The method comprises the following steps: synthesis of benzyl (S) -2- ((1- (4- (pyridin-2-ylcarbamoyl) phenyl) -8-amino) imidazo [1,5-a ] pyrazin-2-yl) pyrrolidin-1-ylcarboxylate (C018)
In a 100mL glass bottle, 4- (4,4,5,5-tetramethyl- [1,3,2] is added]Dioxoboronyl) -benzoic acid 2-pyridylamine amide (213mg,0.66mmol), S8(137mg,0.33mmol) and 1, 2-dimethoxyethane (4 mL). To the above solution was added aqueous sodium carbonate (2M,2mL) and the solution was deoxygenated. Then, add [1,1' -bis (diphenylphosphino) ferrocene to the reaction flask]Palladium dichloride dichloromethane complex (24mg) and oxygen removed again. The reaction was refluxed overnight, cooled to room temperature and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried, concentrated and purified by silica gel column to give the title compound C018(110 mg). ESITheoretical calculation of MS C30H28N7O3[M+H]+534.2; the experiment result shows that 534.2.
Step two: synthesis of (S) -N- (pyridin-2-yl) -4- (3- (pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (C020)
In a 50mL glass vial, C018(110mg) was added. A solution of hydrobromic acid in acetic acid (33%, 3mL) was added via syringe. The reaction was stirred at room temperature for 1 hour and quenched by the addition of 10mL of water. This solution was purified by HPLC to give the trifluoroacetate salt of the target compound C020, which was lyophilized to give 60mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 10%, retention time t1/27.6 min.
1H NMR (MeOD-d4,400mhz):8.47(d, J ═ 5.53Hz,1H),8.40-8.32(m,1H),8.28(d, J ═ 8.24Hz,2H),8.01(d, J ═ 8.60Hz,1H),7.98-7.92(m,3H),7.58(t, J ═ 6.62Hz,1H),7.17(d, J ═ 5.86Hz,1H),5.43(t, J ═ 7.41Hz,1H),3.70-3.60(m,1H),3.60-3.50(m,1H),2.70-2.60(m,1H),2.55-2.42(m,1H),2.42-2.32(m,1H),2.32-2.20(m, 1H). ESI-MS on C22H22N7Theoretical calculation of O [ M + H]+400.5; the test result is 400.3.
Step three: synthesis of (S) -N- (pyridin-2-yl) para- (3- (1- (2, 3-butynoyl) pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (1)
In a 50mL glass bottle, 3-butynoic acid (9mg,0.1mmol), 2-chloro-1-methylpyridine iodide (24mg,0.1mmol) and dry dichloromethane (3mL) were added and the solution was stirred at room temperature for 1 hour. A solution of C020 in trifluoroacetate (30mg) and triethylamine (0.3mL,2.1mmol) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 1 hour. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of the target compound 1, and lyophilized to give 2.4mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 10%, retention time t1/214.6 min.
1H NMR(MeOD-d4,400MHz):8.47(d,J=6.10Hz,1H),8.41-8.35(m,1H),8.30-8.22(m,2H),8.04-7.95(m,2H),7.92-7.86(m,2H),7.62-7.55(m,1H),7.09-7.05(m,1H),6.09(t,J=6.48Hz,1H),5.57-5.48(m,1H),5.305.15(m,2H),4.00-3.90(m,1H),3.90-3.80(m,1H),2.54-2.40(m,2H),2.35-2.25(m,1H),2.25-2.10(m, 1H). Theoretical calculation of ESI-MS C26H24N7O2[M+H]+466.2; 466.3 is measured by experiments. Specific rotation degree
Figure BDA0001365649630000341
(MeOH)。
EXAMPLE 2 Synthesis of the Compound (S) -N- (4-trifluoromethylpyridin-2-yl) para- (3- (1- (2, 3-Butadienoyl) pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (2)
Figure BDA0001365649630000342
The method comprises the following steps: synthesis of benzyl (S) -2- ((1- (4- (4-trifluoromethylpyridin-2-ylcarbamoyl) phenyl) -8-amino) imidazo [1,5-a ] pyrazin-2-yl) pyrrolidin-1-ylcarboxylate (C014)
In a 100mL glass bottle, 4- (4,4,5,5-tetramethyl- [1,3,2] is added]Dioxoboronyl) -benzoic acid 2- (4-trifluoromethyl) pyridylamine amide (470mg,1.2mmol), S8(246mg,0.6mmol) and 1, 2-dimethoxyethane (6 mL). Adding Na into the solution2CO3Aqueous (2M,3mL) and deoxygenated the solution. Then, add [1,1' -bis (diphenylphosphino) ferrocene to the reaction flask]Palladium dichloride dichloromethane complex (42mg) and oxygen removed again. The reaction was refluxed overnight, cooled to room temperature and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried, concentrated and purified by silica gel column to give the objective compound C014(350 mg). Theoretical calculation of ESI-MS C31H27F3N7O3[M+H]+602.2; the test result is 602.3.
Step two: synthesis of (S) -N- (4-trifluoromethylpyridin-2-yl) 4- (3- (pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (C017)
To a 50mL glass bottle, C014(210mg) was added. A solution of hydrobromic acid in acetic acid (33%, 4mL) was added via syringe. The reaction was stirred at room temperature for 1 hour and quenched by the addition of 10mL of water. This is achieved byThe solution was purified by HPLC to give the trifluoroacetate salt of the target compound C017, and lyophilized to give 150mg of solid. HPLC purification conditions: initial acetonitrile proportion 15%, retention time t1/215.0 min.
1H NMR (MeOD-d4,400mhz) 8.65 to 8.86(m,2H),8.18(d, J ═ 8.19Hz,2H),7.95(d, J ═ 5.89Hz,1H),7.90(d, J ═ 8.19Hz,2H),7.43(d, J ═ 4.96Hz,1H),7.15(d, J ═ 5.89Hz,1H),5.44(t, J ═ 7.43Hz,1H),3.70 to 3.60(m,1H),3.60 to 3.50(m,1H),2.74 to 2.60(m,1H),2.54 to 2.42(m,1H),2.42 to 2.32(m,1H),2.32 to 2.20(m, 1H). Theoretical calculation of ESI-MS C23H21F3N7O[M+H]+468.2; the test result is 468.2.
Step three: synthesis of (S) -N- (4-trifluoromethylpyridin-2-yl) para- (3- (1- (2, 3-butynoyl) pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (2)
In a 50mL glass bottle, 3-butynoic acid (10mg,0.12mmol), 2-chloro-1-methylpyridine iodide (38mg,0.15mmol) and dry dichloromethane (3mL) were added and the solution was stirred at room temperature for 1 hour. A solution of C017 trifluoroacetate (40mg) and triethylamine (0.3mL,2.1mmol) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 1 hour. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of the target compound 2, which was lyophilized to give 8.0mg of a solid. HPLC purification conditions: initial acetonitrile proportion 15%, retention time t1/226.2 minutes.
1H NMR (MeOD-d4,400mhz) 8.65 to 8.55(m,2H),8.18(d, J ═ 8.24Hz,2H),8.02(d, J ═ 5.92Hz,1H),7.85(d, J ═ 8.24Hz,2H),7.44(d, J ═ 5.08,1H),7.06(d, J ═ 5.92Hz,1H),6.09(t, J ═ 6.52Hz,1H),5.60 to 5.50(m,1H),5.30 to 5.20(m,2H),4.00 to 3.90(m,1H),3.90 to 3.80(m,1H),2.54 to 2.40(m,2H),2.36 to 2.22(m,1H),2.22 to 2.10(m, 1H). Theoretical calculation of ESI-MS C27H23F3N7O2[M+H]+534.2; the experiment result shows 534.1. Specific rotation degree
Figure BDA0001365649630000351
EXAMPLE 3 Synthesis of the Compound (S) -N- (4-trifluoromethylpyridin-2-yl) 2-fluoro para- (3- (1- (2, 3-butenoyl) pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (3)
Figure BDA0001365649630000352
The method comprises the following steps: synthesis of benzyl (S) -2- ((1- (3-fluoro-4- (4-trifluoromethylpyridin-2-ylcarbamoyl) phenyl) -8-amino) imidazo [1,5-a ] pyrazin-2-yl) -pyrrolidin-1-ylcarboxylate (C022)
In a 100mL glass bottle, 2-fluoro-4- (4,4,5,5-tetramethyl- [1,3, 2)]Dioxoboronyl) -benzoic acid 2- (4-trifluoromethyl) pyridylamine amide (220mg,0.5mmol), S8(100mg,0.24mmol) and 1, 2-dimethoxyethane (6 mL). To the above solution was added aqueous sodium carbonate (2M,4mL) and the solution was deoxygenated. Then, add [1,1' -bis (diphenylphosphino) ferrocene to the reaction flask]Palladium dichloride dichloromethane complex (24mg) and oxygen removed again. The reaction was refluxed overnight, cooled to room temperature and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried, concentrated and purified by silica gel column to give the title compound C022(156 mg). Theoretical calculation of ESI-MS C31H26F4N7O3[M+H]+620.2; the experiment shows that 620.0
Step two: synthesis of (S) -N- (4-trifluoromethylpyridin-2-yl) 2-fluoro-4- (3- (pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (C024)
In a 50mL glass bottle, C022(156mg) was added. A solution of hydrobromic acid in acetic acid (33%, 3mL) was added via syringe. The reaction was stirred at room temperature for 1 hour and quenched by the addition of 10mL of water. This solution was purified by HPLC to give the trifluoroacetate salt of target compound C024, which was lyophilized to give 62.9mg of solid. HPLC purification conditions: initial acetonitrile proportion of 20%, retention time t1/213.5 min.
1H NMR(MeOD-d4,400MHz):8.64-8.58(m,2H),8.05(t,J=7.77Hz,1H),7.95(d,J=5.88Hz,1H),7.75-7.65(m,2H),7.46(d,J=5.04Hz,1H),7.17(d,J=5.88Hz,1H),5.43(t,J=7.41Hz,1H),3.70-3.60(m,1H),3.60-3.50(m,1H),2.72-2.60(m,1H),2.52-2.40(m,1H),2.40-2.32(m,1H),2.32-2.20(m, 1H). Theoretical calculation of ESI-MS C23H20F4N7O[M+H]+486.2; the test result is 486.2.
Step three: synthesis of (S) -N- (4-trifluoromethylpyridin-2-yl) 2-fluoro-para- (3- (1- (2, 3-butynoyl) pyrrolidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (3)
In a 50mL glass bottle, 3-butynoic acid (8.4mg,0.1mmol), 2-chloro-1-methylpyridine iodide (24mg,0.1mmol) and dry dichloromethane (3mL) were added and the solution was stirred at room temperature for 1 hour. A solution of C024 in trifluoroacetate (40mg) and triethylamine (0.2mL,1.4mmol) in dichloromethane (3mL) was added to the above reaction flask and stirred at room temperature for 1 hour. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of the target compound 3, and lyophilized to give 13.0mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 20%, retention time t1/222.9 min.
1H NMR (MeOD-d4,400mhz) 8.65-8.55(m,2H),8.10-7.96(m,2H),7.70-7.60(m,2H),7.50-7.42(m,1H),7.07(d, J ═ 5.93Hz,1H),6.09(t, J ═ 6.53Hz,1H),5.60-5.50(m,1H),5.30-5.18(m,2H),4.00-3.90(m,1H),3.90-3.80(m,1H),2.54-2.40(m,2H),2.34-2.22(m,1H),2.20-2.10(m, 1H). Theoretical calculation of ESI-MS C27H22F4N7O2[M+H]+552.2; the experiment found that the product is 552.1.
EXAMPLE 4 Synthesis of the Compound (R) -2, 3-butenoyl 3- (3- (4-phenoxyphenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidine (4)
Figure BDA0001365649630000371
The method comprises the following steps: 3-iodo-1H-pyrazolo [3,4-d ] pyrimidin-4-amine (B)
1H- [3,4-d]Pyrazolopyrimidine-4-amine (5.3g,39mmol) and N-iodosuccinimide (13.3g,59mmol) were suspended in N, N-dimethylformamide and heated to 80 ℃ under nitrogen for 12 hours. After the reaction is finished, cooling to room temperature, adding water, precipitating a large amount of solid,the mixture was filtered through a buchner funnel, and the solid was washed twice with a saturated sodium thiosulfate solution, water, and hot ethanol, respectively, and then dried by an oil pump to obtain 7.8g of the target compound with a yield of 77%.1H NMR(DMSO-d6,400MHz):8.15(s,1H)。
Step two: 3- (4-Phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidine-4-amine (C)
Mixing 3-iodo-1H- [3,4-d]Pyrazolopyrimidine-4-amine (435mg,1.7mmol) in N, N-dimethylformamide/H2Adding 2- (4-phenoxyphenyl) -4,4,5, 5-tetramethyl- [1,3,2] into the mixed solvent of O]Cyclopentaborane dioxide (1.5g,5.0mmol), potassium phosphate (531mg,2.5mmol) and [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride dichloromethane complex (69mg,0.08mmol) was heated to 120 ℃ under nitrogen for 12 hours. After the reaction, water was added and a large amount of solid was precipitated. Extraction with ethyl acetate was carried out three times, the organic phases were combined, washed with water three times, dried by adding anhydrous sodium sulfate, the solvent was dried by spinning, and column purification was carried out to obtain the objective compound 187mg in 37% yield.1H NMR(DMSO-d6400MHz) 13.56(s,1H),8.22(s,1H),7.66(d, J ═ 8.7Hz,2H),7.43(t, J ═ 8.1Hz,2H),7.21-7.11(m, 5H). Theoretical calculation of ESI-MS C17H13N5O[M+H]+304.1, the experiment found: 304.3.
step three: (R) -1- (piperidin-3-yl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-D ] pyrimidine-4-amine (D)
Reacting 3- (4-phenoxyphenyl) -1H- [3,4-d]Pyrazolopyrimidine-4-amine (185mg,0.61mmol), (S) -1-tert-butoxycarbonyl-3-hydroxypiperidine (246mg,1.22mmol) and triphenylphosphine (640mg,2.44mmol) were dissolved in dry tetrahydrofuran, diisopropyl azodicarboxylate (0.6mL,3.05mmol) was added dropwise, and the mixture was heated to 40 ℃ for 12 hours. After the reaction is finished, the tetrahydrofuran is removed by spinning, and the 3- [ 4-amino-3- (4-phenoxyphenyl) -1- [3,4-d is obtained by column purification]Pyrazolopyrimidinyl]Tert-butyl piperidinecarboxylate. This was dissolved in methylene chloride (6mL), and trifluoroacetic acid (6mL) was added to the solution to conduct a reaction at room temperature for 12 hours. The reaction progress was monitored by HPLC. After the reaction is finished, removing trifluoroacetic acid by spinning, adding water and methanol, separating by an HPLC preparative column, and freeze-drying to obtain 111mg of the trifluoroacetate salt of the target compound with the yield of 38%. HPLC purification conditions: initial acetonitrile ratio25% retention time t 1/210 minutes.
1H NMR(MeOD-d4400MHz) 8.43(s,1H),7.72(d, J-8.5 Hz,2H),7.42(t, J-8.0 Hz,2H),7.23-7.09(m,5H),5.31-5.22(m,1H),3.77-3.65(m,2H),3.45-3.38(m,1H),3.29-3.20(m,1H),2.40-2.23(m,2H),2.19-2.09(m,1H),2.02-1.90(m, 1H). Theoretical calculation of ESI-MS C22H22N6O[M+H]+387.2, the experiment found: 387.1.
step four: (R) -2, 3-butenoyl 3- (3- (4-phenoxyphenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidine (4)
In a 50mL glass bottle, 3-butynoic acid (10mg,0.12mmol), 2-chloro-1-methylpyridine iodide (38mg,0.15mmol) and dry dichloromethane (3mL) were added, and the reaction solution was stirred at room temperature for 1 hour. 1-3-piperidine- (4-phenoxyphenyl) -1H- [3,4-d]A solution of pyrazolopyrimidine-4-amine trifluoroacetate (40mg,0.1mmol) and triethylamine (0.1mL,0.72mmol) in dichloromethane (3mL) was added to the above reaction flask and stirred at room temperature for 1 hour. The dichloromethane was evaporated using a rotary evaporator. The resulting dope was purified by HPLC to give the trifluoroacetate salt of target compound 4, and lyophilized to give 7mg (15%) of a solid. HPLC purification conditions: initial acetonitrile proportion of 20%, retention time t1/230.0 min.
1H NMR(MeOD-d4400 MHz: 8.39-8.45(m,1H),7.65-7.72(m,2H),7.39-7.45(m,2H),7.14-7.23(m,3H),7.08-7.14(m,2H),6.26(t, J ═ 5.97Hz,0.5H),6.09(t, J ═ 6.64Hz,0.5H),5.198-5.26(m,2H),4.60-4.45(m,0.7H),4.40-4.15(m,1H),4.10-3.95(m,0.6H), 3.95-385 (m,0.3H),3.85-3.70(m,0.3H),3.65-3.56(m,0.5H),3.56-3.40(m,0.5H), 2.30H, 1.20-20H), theoretical calculation values (m,1H, 2H), 1.5H, 1H, 20H, 1H, 20-20H, 1H, 20H, 126H25N6O2[M+H]+453.2, the experiment found: 453.1. specific rotation degree
Figure BDA0001365649630000381
(MeOH)。
Synthetic route to key intermediate C044 of preparation example 3:
Figure BDA0001365649630000382
step one, synthesizing (R) -3- [ (3-chloro-pyrazine-2-methylene) -carbamoyl ] -piperidine-1-formic acid tert-butyl ester (C032)
In a 100mL glass bottle, S4(0.60g,4.8mmol), N-Boc- (R) -3-carboxylic acid piperidine (1.37g,5.9mmol) and dichloromethane (30mL) were added. The reaction flask was cooled with ice water, and then triethylamine (3mL,19.2mmol) was added and stirred for 10 min. 2- (7-Benzotolyltriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (HATU,2.24g,5.9mmol) was added in one portion and stirred at room temperature overnight. The reaction was quenched with water, the aqueous phase was extracted with dichloromethane, and the organic phases were combined. The organic phase was washed with saturated brine, dried, concentrated and purified by silica gel column. Purification gave the title compound C032 as an oily liquid (1.52 g).1H NMR (CDCl3,400MHz) 8.45(s,1H),8.32(s,1H),7.10-6.90(br,1H),4.80-4.60(m,2H),4.00-3.80(m,1H),3.20-3.00(m,1H),3.00-2.80(m,1H),2.50-2.30(m,1H),2.00-1.90(m,1H),1.90-1.60(m,4H),1.45(s, 9H). Theoretical calculation of ESI-MS C16H23 35ClN4NaO3[M+Na]+377.1, found experimentally: 377.1.
step two, synthesizing (R) -3- [ (3-chloro-pyrazine-2-methylene) -carbamoyl ] -piperidine-1-carboxylic acid benzyl ester (C036)
In a 100mL glass vial, C032(1.52g,4.3mmol) and dichloromethane (5mL) were added. Trifluoroacetic acid (5mL) was added by syringe, followed by stirring at room temperature for 1 hour. The solvent and trifluoroacetic acid were evaporated using a rotary evaporator. The resulting crude was placed in a 100mL glass vial and anhydrous tetrahydrofuran (30mL) and triethylamine (2.5mL,18mmol) were added. The reaction flask was cooled with ice water, benzyl chloroformate (1.1g,6.5mmol) was added via syringe and stirred at room temperature overnight. The reaction was quenched with water, the aqueous phase was extracted with ethyl acetate and the organic phases were combined. The organic phase was washed with brine, then dried over anhydrous sodium sulfate and concentrated using a rotary evaporator. The remaining crude product was purified by silica gel column to give C036(1.1 g).
1H NMR(CDCl3,400MHz):8.40(s,1H),8.31(s,1H),7.45-7.25(m,5H),5.20-5.04(m,2H),4.804.57(m,2H),4.23-4.10(m,1H),4.10-3.80(m,1H),3.28-3.15(m,1H),3.12-2.84(m,1H),2.54-2.35(m,1H),2.22-2.06(m,1H),2.06-1.65(m,3H),1.62-1.40(m, 1H). Theoretical calculation of ESI-MS C19H21 35ClN4NaO3[M+Na]+As 411.1, experimentally determined: 411.3.
step three, synthesizing (R) -3- (8-chloro- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-benzyl formate (CDA-114)
Reacting 3- [ (3-chloro-pyrazine-2-methylene) -carbamoyl]-piperidine-1-carboxylic acid benzyl ester (C036, 1.10g,2.84mmol) was dissolved in acetonitrile, 1, 3-dimethyl-2-imidazolidinone (0.97g,8.51mmol) was added, cooled to 0 deg.C, phosphorus oxychloride (1.74g,11.40mmol) was added, and after the addition, the temperature was raised to 60 deg.C for 12 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, and then slowly added to 50mL of 25% aqueous ammonia ice. Extraction was performed with ethyl acetate (100 mL. times.3), and the organic phases were combined, washed once with saturated brine, dried over anhydrous sodium sulfate, the solvent was removed by rotation, and the mixture was passed through a silica gel column to obtain the objective compound CDA-114(524 mg).1H NMR(CDCl3400MHz) of 7.85 to 7.65(m,2H),7.50 to 7.30(m,5H),7.25 to 7.10(m,1H),5.24 to 5.04(m,2H),4.50 to 4.18(m,2H),3.28 to 2.84(m,3H),2.24 to 2.07(m,2H),1.97 to 1.83(m,1H),1.80 to 1.55(m, 1H). Theoretical calculation of ESI-MS C19H19 35ClN4O2[M+H]+371.1, the experiment found: 371.4.
step four, synthesizing (R) -3- (1-bromo-8-chloro- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-carboxylic acid benzyl ester (CDA-116)
Reacting 3- (8-chloro- [1, 5-a)]Imidazopyrazin-3-yl) -piperidine-1-carboxylic acid benzyl ester (CDA-114, 514mg,1.39mmol) was dissolved in 5mL of DMF, cooled to 0 deg.C and NBS (272mg,1.53mmol) added overnight at room temperature. After completion of the reaction, 30mL of a saturated sodium thiosulfate solution was added to the reaction mixture, extraction was performed with ethyl acetate (100 mL. times.3), the organic phases were combined, washed with water (50 mL. times.3), dried over anhydrous sodium sulfate, the solvent was removed by rotation, and the mixture was purified by a silica gel column to obtain the objective compound CDA-116(628 mg).1H NMR(CDCl3,400MHz):7.80-7.65(m,1H),7.50-7.25(m,5H),7.20-7.05(m,1H),5.27-5.04(m,2H),4.49-4.19(m,2H),3.24-2.83(m,3H),2.18-2.10(m,2H),1.95-1.82(m,1H),1.70-1.50(m,1H) In that respect Theoretical calculation of ESI-MS C19H18 35Cl79BrN4O2[M+H]+449.0, the experiment found: 449.2.
step five, synthesizing (R) -3- (1-bromo-8-amino- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-carboxylic acid benzyl ester (C044)
To a 25mL thick-walled glass vial, CDA-116(0.53g,1.1mmol), DMSO (2mL) and isopropanol (12mL) were added. Concentrated ammonia (6mL) was added to the solution, then the tube was closed and heated in an oil bath at 85 ℃ overnight. The reaction solution was cooled and then concentrated, and the resulting mixture was purified by a silica gel column to obtain the objective compound C044(389 mg).
1H NMR(CDCl3400MHz) of 7.50 to 7.20(m,5H),7.10 to 6.80(m,2H),6.10 to 5.80(m,2H),5.20 to 5.00(m,2H),4.50 to 4.10(m,2H),3.20 to 2.70(m,3H),2.20 to 1.70(m,3H),1.70 to 1.50(m, 1H). Theoretical calculation of ESI-MS C19H21 79BrN5O2[M+H]430.1, experimentally determined: 430.2.
synthetic route to key intermediate C049 of preparation example 4:
Figure BDA0001365649630000401
step one, synthesizing (S) -2- [ (3-chloro-pyrazine-2-methylene) -carbamoyl ] -piperidine-1-carboxylic acid tert-butyl ester (C038)
In a 100mL glass bottle, S4(0.60g,4.8mmol), N-Boc- (S) -2-carboxylic acid piperidine (1.37g,6.0mmol) and dichloromethane (30mL) were added. The reaction flask was cooled with ice water, and then triethylamine (3mL,19.2mmol) was added and stirred for 10 min. 2- (7-Benzotolyltriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (HATU,2.28g,6.0mmol) was added in one portion and stirred at room temperature overnight. The reaction was quenched with water, the aqueous phase was extracted with dichloromethane, and the organic phases were combined. The organic phase was washed with saturated brine, dried, concentrated and purified by silica gel column to give the objective compound C038 as an oily liquid (1.59 g).1H NMR(CDCl3,400MHz):8.42(s,1H),8.31(s,1H),7.40-7.25(br,1H),5.00-4.40(m,3H),4.20-4.00(m,1H),3.00-2.73(m,1H),2.40-2.20(m,1H),1.80-1.20(m5H),1.50(s, 9H). Theoretical calculation of ESI-MS C16H23 35ClN4NaO3[M+Na]+377.1, found experimentally: 377.4.
step two, synthesizing (S) -2- [ (3-chloro-pyrazine-2-methylene) -carbamoyl ] -piperidine-1-carboxylic acid benzyl ester (C040)
In a 100mL glass bottle, C038(1.59g,4.5mmol) and dichloromethane (5mL) were added. Trifluoroacetic acid (5mL) was added by syringe, followed by stirring at room temperature for 1 hour. The solvent and trifluoroacetic acid were evaporated using a rotary evaporator. The resulting crude was placed in a 100mL glass vial and anhydrous tetrahydrofuran (20mL) and triethylamine (2.5mL,18mmol) were added. The reaction flask was cooled with ice-water, benzyl chloroformate (1.2g,6.75mmol) was added via syringe and stirred at room temperature overnight. The reaction was quenched with water, the aqueous phase was extracted with ethyl acetate and the organic phases were combined. The organic phase was washed with brine, then dried over anhydrous sodium sulfate and concentrated using a rotary evaporator. The remaining crude product was purified by silica gel column to give C040(1.29 g).
1H NMR(CDCl3400MHz) 8.36(s,1H),8.30(s,1H),7.48-7.05(m,5H),5.30-5.10(m,2H),5.05-4.85(m,1H),4.85-4.70(m,1H),4.70-4.50(m,1H),4.30-4.00(m,1H),3.10-2.90(m,1H),2.50-2.30(m,1H),1.80-1.37(m, 5H). Theoretical calculation of ESI-MS C19H21 35ClN4NaO3[M+Na]+As 411.1, experimentally determined: 411.1.
step three, synthesizing (S) -2- (8-chloro- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-benzyl formate (C042)
2- [ (3-chloro-pyrazine-2-methylene) -carbamoyl ] -piperidine-1-carboxylic acid benzyl ester (C036, 1.29g,3.5mmol) was dissolved in acetonitrile, 1, 3-dimethyl-2-imidazolidinone (1.14mL,10.5mmol) was added, cooling to 0 deg.C, phosphorus oxychloride (2.05mL,14mmol) was added, and after the addition, the temperature was raised to 60 deg.C for 12 hours. After the reaction was completed, the reaction mixture was cooled to room temperature, and the reaction mixture was slowly added to a mixture of 30mL of concentrated aqueous ammonia and ice. Extraction with ethyl acetate (100 mL. times.3) was performed, and the organic phases were combined, washed once with saturated brine, dried over anhydrous sodium sulfate, the solvent was removed by rotation, and the mixture was subjected to silica gel column to obtain the objective compound C042(756 mg).
1H NMR(CDCl3400MHz, 8.10-7.80(m,1H),7.80(s,1H),7.45-7.30(m,5H),7.26-7.05(m,1H),5.90-5.75(br,1H),5.30-5.05(m,2H),4.10-3.90(m,1H),2.70(td, J ═ 13.25,2.29Hz,1H),2.55-2.30(m,2H),2.10-1.90(m,1H),1.90-1.75(m,1H),1.76-1.65(m,1H),1.65-1.50(m, 1H). Theoretical calculation of ESI-MS C19H20 35ClN4O2[M+H]+371.1, the experiment found: 371.3.
step four, synthesizing (S) -2- (1-bromo-8-chloro- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-carboxylic acid benzyl ester (C048)
Benzyl 2- (8-chloro- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-carboxylate (C042, 969mg,2.6mmol) was dissolved in 25mL of DMF, cooled to 0 deg.C and NBS (513mg,2.9mmol) added overnight at room temperature. After completion of the reaction, 30mL of a saturated sodium thiosulfate solution was added to the reaction mixture, extraction was performed with ethyl acetate (100 mL. times.3), the organic phases were combined, washed with water (50 mL. times.3), dried over anhydrous sodium sulfate, the solvent was removed by rotation, and the mixture was purified by a silica gel column to obtain the objective compound C048(650 mg).
1H NMR(CDCl3400MHz) 8.10-7.80(m,1H),7.50-7.30(m,5H),7.23-7.07(m,1H),5.85-5.70(m,1H),5.28-5.10(m,2H),4.06-3.94(m,1H),2.73(td, J ═ 13.43,1.84Hz,1H),2.53-2.38(m,1H),2.38-2.25(m,1H),2.03-1.89(m,1H),1.86-1.67(m,2H),1.63-1.46(m, 1H). Theoretical calculation of ESI-MS C19H19 79Br35ClN4O2[M+H]+449.1, the experiment found: 449.1.
step five, synthesizing (S) -2- (1-bromo-8-amino- [1,5-a ] imidazopyrazin-3-yl) -piperidine-1-carboxylic acid benzyl ester (C049)
To a 25mL thick-walled glass vial, C048(0.50g,1.0mmol), DMSO (2mL) and isopropanol (12mL) were added. Concentrated ammonia (6mL) was added to the solution, then the tube was closed and heated in an oil bath at 85 ℃ overnight. The reaction solution was cooled and then concentrated, and the resulting mixture was purified by a silica gel column to obtain the objective compound C049.
1H NMR(CDCl3,400MHz):7.60-7.25(m,5H),7.05-6.80(m,1H),6.00-5.60(m,3H),5.28-5.09(m,2H),4.00(d,J=13.2Hz,1H),2.79(dt,J=13.3Hz,J=2.1Hz,1H),2.50-2.22(m,2H),2.00-1.84(m,1H),1.82-1.64(m,2H),1.62-1.45(m, 1H). Theoretical calculation of ESI-MS C19H21 79BrN5O2[M+H]+430.1, the experiment found: 430.2.
example 5: synthesis of (R) -N- (pyridin-2-yl) para- (3- (1- (2, 3-butenoyl) piperidin-3-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (5)
Figure BDA0001365649630000421
The method comprises the following steps: (R) -benzyl 3- ((1- (4- (pyridin-2-ylcarbamoyl) phenyl) -8-amino) imidazo [1,5-a ] pyrazin-3-yl) -piperidin-1-ylcarboxylate (C047)
In a 100mL glass bottle, 4- (4,4,5,5-tetramethyl- [1,3,2] is added]Dioxocyclopentanobutyl) -benzoic acid 2-pyridylamine amide (583mg,1.8mmol), C044(389mg,0.90mmol) and 1, 2-dimethoxyethane (6 mL). To the above solution was added aqueous sodium carbonate (2M,6mL) and the solution was deoxygenated. Then, add [1,1' -bis (diphenylphosphino) ferrocene to the reaction flask]Palladium dichloride dichloromethane complex (73mg.0.09mmol) and oxygen again. The reaction was refluxed overnight, cooled to room temperature and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with saturated brine, dried, concentrated and purified over silica gel column. Purification gave the title compound C047(498 mg). Theoretical calculation of ESI-MS C31H30N7O3[M+H]+548.2; the test result is 548.2.
Step two: synthesis of (R) -N- (pyridin-2-yl) para (3- (piperidin-3-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (CDA-118)
In a 50mL glass vial, C047(498mg) was added. A solution of hydrobromic acid in acetic acid (33%, 6mL) was added via syringe. The reaction was stirred at room temperature for 1 hour and quenched by the addition of 20mL of water. This solution was purified by HPLC to give the trifluoroacetate salt of the desired compound CDA-118, which was lyophilized to give 170mg of solid.
HPLC purification conditions: initial acetonitrile proportion of 5%, retention time t1/212.5 min.1H NMR(MeOD-d4,400MHz):8.55-8.43(m,2H),8.31(d, J ═ 8.2Hz,2H),8.02-7.92(m,4H),7.66(t, J ═ 6.7Hz,1H),7.11(d, J ═ 5.9Hz,1H),3.92-3.81(m,1H),3.69(dd, J ═ 12.6Hz, J ═ 3.5Hz,1H)3.60(dd, J ═ 12.6Hz, J ═ 9.6Hz,1H),3.50-3.40(m,1H),3.30-3.18(m,1H),2.36-2.20(m,1H),2.10-1.90(m, 3H). Theoretical calculation of ESI-MS C23H24N7O[M+H]+414.2; the test result is 414.3.
Step three: synthesis of (R) -N- (pyridin-2-yl) para- (3- (1- (2, 3-butenoyl) piperidin-3-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (5)
In a 50mL glass bottle, 3-butynoic acid (8mg,0.09mmol), 2-chloro-1-methylpyridine iodide (38mg,0.17mmol) and dry dichloromethane (3mL) were added and the solution was stirred at room temperature for 1 hour. A solution of CDA-118 in trifluoroacetate (40mg,0.076mmol) and triethylamine (0.1mL) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 1 hour. The viscous product obtained by evaporating dichloromethane with a rotary evaporator was purified by HPLC to obtain the trifluoroacetate salt of the objective compound 5, which was then lyophilized to obtain 11.2mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 10%, retention time t1/216.5 min.1HNMR (MeOD-d4,400mhz):8.47(d,1H, J ═ 5.4Hz),8.33(t,1H, J ═ 7.8Hz),8.26(d,2H, J ═ 8.2Hz),7.99(d,1H, J ═ 8.6Hz),7.96-7.85(m,3H),7.55(t,1H, J ═ 6.5Hz),7.06(d,1H, J ═ 6.0Hz),6.30-6.15(m,1H),5.30-5.10(m,2H),4.65-4.55(m,1H),4.35-4.22(m,1H),4.17-4.09(m,1H),3.87-3.76(m,1H),3.45-3.35(m,1H), 1.84-1H, 1.84(m,1H), 1.17-4.84 (m,1H). Theoretical calculation of ESI-MS C27H26N7O2[M+H]+480.2; the test result is 480.2.
Example 6: synthesis of (S) -N- (pyridin-2-yl) para- (3- (1- (2, 3-butenoyl) piperidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (6)
Figure BDA0001365649630000431
The method comprises the following steps: (S) -benzyl 2- ((1- (4- (pyridin-2-ylcarbamoyl) phenyl) -8-amino) imidazo [1,5-a ] pyrazin-3-yl) piperidin-1-ylcarboxylate (C050)
In a 100mL glass vial, 4- (4,4,5,5-tetramethyl- [1,3,2] dioxaboronyl) -benzoic acid 2-pyridylaminamide (608mg,2.0mmol), C049(400mg,1.0mmol) and 1, 2-dimethoxyethane (6mL) were added. To the above solution was added aqueous sodium carbonate (2M,6mL) and the solution was deoxygenated. Then, the [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (81mg.0.1mmol) was added to the reaction flask and oxygen was again removed. The reaction was refluxed overnight, cooled to room temperature and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with saturated brine, dried, concentrated and purified over silica gel column. Purification yielded the target compound C050(515 mg).
1H NMR(CDCl3400MHz) 8.90-8.80(br,1H),8.42(d, J ═ 8.31Hz,1H),8.31(d, J ═ 4.35Hz,1H),8.06(d, J ═ 7.98Hz,2H),7.85(d, J ═ 7.90Hz,2H),7.79(t, J ═ 7.62Hz,1H),7.44-7.30(m,5H),7.15-7.06(m,1H),7.05-6.90(m 1H),5.90-5.72(br,1H),5.30-5.10(m,3H),4.04(d, J ═ 12.56Hz,1H),2.91(t, J ═ 12.88Hz,1H),2.60-2.44(m, 2.44H), 2.04 (d, J ═ 12.56Hz,1H),2.91(t, J ═ 12.88, 1H),2.60-2.44(m, 6H), 6.66H, 1H), 1H, 6H, 1H, 6, 1. Theoretical calculation of ESI-MS C31H30N7O3[M+H]+548.2; the test result is 548.1.
Step two: synthesis of (S) -N- (pyridin-2-yl) para- (3- (piperidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (C053)
In a 50mL glass bottle, C050(500mg) was added. A solution of hydrobromic acid in acetic acid (33%, 10mL) was added via syringe. The reaction was stirred at room temperature for 1 hour and quenched by the addition of 20mL of water. This solution was purified by HPLC to give the trifluoroacetate salt of the objective compound C053 (515 mg).
HPLC purification conditions: initial acetonitrile proportion of 5%, retention time t1/213 minutes.1H NMR (MeOD-d4,400mhz) 8.48(d,1H, J ═ 5.5Hz),8.39(t,1H, J ═ 8.0Hz),8.28(d,2H, J ═ 8.2Hz),8.05-7.92(m,4H),7.59(t,1H, J ═ 6.6Hz),7.15(d,1H, J ═ 5.8Hz),3.64-3.56(m,1H),3.34-3.26(m,2H),2.40-2.25(m,1H),2.15-1.80(m, 5H). Theoretical calculation of ESI-MS C23H24N7O[M+H]+414.2; the experiment found that 414.2.
Step three: synthesis of (S) -N- (pyridin-2-yl) para- (3- (1- (2, 3-butenoyl) piperidin-2-yl) -8-aminoimidazo [1,5-a ] pyrazin-1-yl) benzamide (6)
In a 50mL glass vial, 3-butynoic acid (13mg) and dry dichloromethane (3mL) were added. To the above solution was added 2-chloro-1-methylpyridine iodide (23 mg). The solution was stirred at room temperature for 1 hour. A solution of C053 in trifluoroacetate (44mg) and triethylamine (0.1mL) in dichloromethane (3mL) was added to the above reaction flask and stirred at room temperature for 40 minutes. The viscous product obtained by evaporating dichloromethane with a rotary evaporator was purified by HPLC to obtain the trifluoroacetate salt of the objective compound 6, which was then lyophilized to obtain 6.8mg of a solid.
HPLC purification conditions: initial acetonitrile proportion of 10%, retention time t1/220.5 min.1H NMR (MeOD-d4,400mhz):8.48(d,1H, J ═ 5.7Hz),8.36(t,1H, J ═ 8.0Hz),8.27(d,2H, J ═ 8.2Hz),8.00-7.90(m,3H),7.91-7.81(m,1H),7.57(t,1H, J ═ 6.6Hz),7.04(d,1H, J ═ 5.8Hz),6.27(t,2H, J ═ 6.55Hz),5.26(d,2H, J ═ 6.55Hz),4.10-3.90(m,1H),3.50-3.20(m,1H),2.50-2.30(m,2H),2.10-1.90(m,1H), 1.91-76 (m,1H), 1.58(m, 1H). Theoretical calculation of ESI-MS C27H26N7O2[M+H]+480.2; the experiment found that 480.3. Specific rotation degree
Figure BDA0001365649630000451
(MeOH)。
EXAMPLE 7 Synthesis of the Compound (R) -2, 3-butenoyl 3- (3- (4-phenoxy-2-fluoro-phenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidine (7)
Figure BDA0001365649630000452
The method comprises the following steps: synthesis of 2-fluoro-4-bromophenyl-phenyl ether (C041)
In a 50mL dry glass bottle, p-bromom-fluorophenol (400mg,2mmol), phenylboronic acid (490mg,4mmol), anhydrous cupric acetate (437mg,2.4mmol) and dry molecular sieve (C: (R) (R))
Figure BDA0001365649630000453
2g) In that respect Dry dichloromethane (10mL) was added to the bottle and finally triethylamine (1.5mL) was added. After the reaction was stirred at room temperature for 24 hours, the reaction solution was filtered through celite to remove solids. The celite was rinsed with dichloromethane (200mL), the organic phases were combined and concentrated, and the resulting reaction crude was purified with normal phase silica gel column to give the title compound C041(140 mg).1H NMR(CDCl3,400MHz):7.46(t,1H,J=8.34Hz),7.42-7.36(m,2H),7.18(t,1H,J=7.41Hz),7.04(d,2H,J=7.86Hz),6.77(dd,1H,J=2.57Hz,J=9.76Hz),6.70(d,1H,J=8.81Hz)。
Step two: synthesis of 2- (4-phenoxy-2-fluoro-phenyl) -4,4,5, 5-tetramethyl- [1,3,2] dioxaborolan (C051)
In a 100mL dry glass bottle, 2-fluoro-4-bromophenyl-phenyl ether (700mg,2.62mmol), bis-pinacol boronate (1.32g,5.2mmol), potassium acetate (KOAc, 1.03g,10.5mmol) and dry 1, 4-dioxane (15mL) were added. Removing oxygen from the reaction solution, and adding [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride dichloromethane complex (182mg,0.26mmol) and oxygen removed again. The reaction solution was heated to 90 ℃ and stirred overnight. The reaction was quenched with 50mL of water and extracted three times with ethyl acetate (50 mL). The combined organic phases were washed with saturated brine, dried, concentrated and purified over silica gel column. Purification afforded the title compound C051 as a solid (1.30 g).1H NMR(CDCl3400MHz) 7.68(t,1H, J ═ 7.61Hz),7.38(d,2H, J ═ 7.71Hz),7.17(t,1H, J ═ 7.46Hz),7.04(d,2H, J ═ 8.05Hz),6.76(d,1H, J ═ 8.21Hz),6.61(d,1H, J ═ 10.71Hz),1.35(s, 12H). Theoretical calculation of ESI-MS C18H21BFO3[M+H]315.2, found experimentally.
Step three: synthesis of 3- (4-phenoxy-2-fluoro-phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (C054)
Mixing 3-iodo-1H- [3,4-d]Pyrazolopyrimidine-4-amine (520mg,2.0mmol) in N, N-dimethylformamide/H2O (12mL-8mL), C051(1.30g,4.1mmol), potassium phosphate (1.27g,6mmol) and [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride dichloromethane complex (163mg,0.2mmol) was heated to 120 ℃ under nitrogen for 12 hours. After the reaction is finished, adding water and a large amount of solidAnd (4) precipitating. Extracting with ethyl acetate (50mL) for three times, combining organic phases, washing with water for three times, adding anhydrous sodium sulfate for drying, spin-drying the solvent, and purifying by column chromatography to obtain the target compound C054184 mg.
1H NMR(CDCl3400MHz) 7.56-7.40(m,4H),7.21(t,1H, J ═ 7.72Hz),7.18(d,2H, J ═ 8.20Hz),7.01(d,1H, J ═ 11.61Hz),6.92(d,1H, J ═ 9.10 Hz). Theoretical calculation of ESI-MS C17H13FN5O[M+H]+322.1, found experimentally: 322.1.
step four: synthesis of (R) -1- (piperidin-3-yl) -3- (4-phenoxy-2-fluoro-phenyl) -1H-pyrazolo [3,4-d ] pyrimidine-4-amine (C065)
C054(184mg,0.60mmol), (S) -1-tert-butoxycarbonyl-3-hydroxypiperidine (241mg,1.2mmol) and triphenylphosphine (628mg,2.4mmol) were dissolved in dry tetrahydrofuran, diisopropyl azodicarboxylate (2.42g,12mmol) was added dropwise, and the mixture was heated to 40 ℃ for 12 hours. After the reaction is finished, the tetrahydrofuran is removed by spinning, and the tert-butyl 3- [ 4-amino-3- (4-phenoxy-2-fluoro-phenyl) -1- [3,4-d ] pyrazolopyrimidinyl ] piperidinecarboxylate is obtained by column purification. This was dissolved in methylene chloride (3mL), and trifluoroacetic acid (8mL) was added to the solution to conduct a reaction at room temperature for 12 hours. After the reaction is finished, removing trifluoroacetic acid by spinning, adding water and methanol, separating by an HPLC (high performance liquid chromatography) preparative column, and freeze-drying to obtain 95mg of trifluoroacetate of the target compound C065.
HPLC purification conditions: initial acetonitrile proportion 25%, retention time t1/214 minutes.1H NMR(MeOD-d4400MHz) 8.43(s,1H),7.65(t,1H, J ═ 8.5Hz),7.46(t,2H, J ═ 7.7Hz),7.25(t,1H, J ═ 7.4Hz),7.15(d,2H, J ═ 8.0Hz),6.97(d,1H, J ═ 8.9Hz),6.91(d,1H, J ═ 11.5Hz),5.38-5.24(m,1H),3.78-3.68(m,2H),3.50-3.40(m,1H),3.30-3.16(m,1H),2.40-2.24(m,2H),2.22-2.10(m,1H),2.06-1.90(m, 1H). Theoretical calculation of ESI-MS C22H22FN6O[M+H]405.2, experimentally determined: 405.1.
step five: (R) -2, 3-butenoyl 3- (3- (4-phenoxy-2-fluoro-phenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidine (7)
3-Butynoic acid (16.8mg,0.2mmol) was placed in a 50mL glass vial, dried dichloromethane (3mL) was added to dissolve the substrate, then 2-chloro-1-methylpyridine iodide (29mg,0.13mmol) was added, and the reaction solution was stirred at room temperature for 1 hour. A solution of C065(50mg,0.1mmol) and triethylamine (0.1mL,0.72mmol) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 40 minutes. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of the target compound 7, which was lyophilized to give 18mg of a solid.
HPLC purification conditions: initial acetonitrile proportion of 30%, retention time t1/218 minutes.1H NMR(MeOD-d4400MHz):8.45-8.37(m,1H),7.68-7.58(m,1H),7.66-7.58(m,1H),7.46(t,2H, J ═ 7.85Hz),7.25(t,1H, J ═ 7.43Hz),7.16(d,2H, J ═ 7.98Hz),6.97(d,1H, J ═ 8.68Hz),6.91(d,1H, J ═ 11.54Hz),6.24(t,0.5H, J ═ 6.15Hz),6.08(t,0.5H, J ═ 6.15Hz),5.22-5.05(m,1H),4.60-4.40(m,0.6H),4.40-4.10(m,0.8H),4.10-3.80(m, 3.70H), 3.70H, 3.70 (m-3.70H), 3.3.70H, 3.70 (m-3.3.50H), 3.3.50H, 3.70H, 3.3, 3.5H, 3.1H, 326H24FN6O2[M+H]+471.2, experimentally determined: 471.3.
example 8: synthesis of the Compound (S) -2, 3-butenoyl-2- (1- (4-phenoxyphenyl) -8-aminoimidazo [1,5-a ] pyrazin-3-yl) -1-pyrrolidine (8)
Figure BDA0001365649630000471
The method comprises the following steps: synthesis of the Compound benzyl (S) -2- (1- (4-phenoxyphenyl) -8-aminoimidazo [1,5-a ] pyrazin-3-yl) -pyrrolidin-1-ylcarboxylate (C063)
In a 100mL glass bottle, 2- (4-phenoxyphenyl) -4,4,5, 5-tetramethyl- [1,3,2]Dioxolane (580mg,2.0mmol), S8(400mg,1.0mmol) and 1, 2-dimethoxyethane (6 mL). To the above solution was added aqueous sodium carbonate (2M,6mL) and the solution was deoxygenated. Then, add [1,1' -bis (diphenylphosphino) ferrocene to the reaction flask]Palladium dichloride dichloromethane complex (70mg.0.1mmol) and deoxygenated again. The reaction was refluxed overnight, cooled to room temperature, and the aqueous phase was extracted three times with ethyl acetate (50 mL. times.3). The organic phases were combined, washed with saturated brine and driedDried, concentrated and purified over silica gel column. Purification gave the title compound C063(320 mg). Theoretical calculation of ESI-MS C30H28N5O3[M+H]+506.2; the test result is 506.1.
Step two: synthesis of 1- (4-phenoxyphenyl) -3- (pyrrolidin-2-yl) -8-amino-imidazo [1,5-a ] pyrazine (C067)
In a 50mL glass bottle, C063(320mg) was added. A solution of hydrobromic acid in acetic acid (33%, 6mL) was added via syringe. The reaction was stirred at room temperature for 1 hour and quenched by the addition of 20mL of water. This solution was purified by HPLC to give the trifluoroacetate salt of the target compound C067, which was lyophilized to give 148mg of solid.
HPLC purification conditions: initial acetonitrile proportion 15%, retention time t1/216 minutes.1H NMR (MeOD-d4,400mhz):7.90(d,1H, J ═ 5.90Hz),7.70(d,2H, J ═ 8.49Hz),7.41(t,2H, J ═ 7.85Hz),7.24-7.05(m,6H),5.41(t,1H, J ═ 7.41Hz),3.70-3.59(m,1H),3.59-3.50(m,1H),2.70-2.58(m,1H),2.54-2.40(m,1H),2.40-2.18(m, 2H). Theoretical calculation of ESI-MS C22H22N5O[M+H]+372.2; the test result is 372.3.
Step three: synthesis of the Compound (S) -2, 3-butenoyl-2- (1- (4-phenoxyphenyl) -8-aminoimidazo [1,5-a ] pyrazin-3-yl) -1-pyrrolidine (8)
In a 50mL glass vial, 3-butynoic acid (17mg,0.2mmol) and dry dichloromethane (3mL) were added followed by 2-chloro-1-methylpyridine iodide (30mg,0.13mmol) and the solution was stirred at room temperature for 1 hour. A solution of C067 trifluoroacetate salt (50mg,0.1mmol) and diisopropylethylamine (0.2mL) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 40 minutes. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of target compound 8, and lyophilized to give 14.8mg of a solid. HPLC purification conditions: initial acetonitrile proportion 25%, retention time t1/218 minutes.1H NMR(MeOD-d4,400MHz):7.96(d,1H,J=5.94Hz),7.63(d,2H,J=8.50Hz),7.41(t,2H,J=7.83Hz),7.22-7.08(m,5H),7.00(d,1H,J=5.91Hz),6.07(t,1H,J=6.53Hz),5.20-4.80(m,1H),5.30-5.18(m,2H),3.98-3.88(m,1H),3.88-3.78(m,1H),2.60-2.33(m,2H) 2.30-2.20(m,1H),2.20-2.08(m, 1H). Theoretical calculation of ESI-MS C26H24N5O2[M+H]+438.2; the test result is 438.1.
EXAMPLE 9 Synthesis of the Compound (R) -2, 3-butynenoyl 3- (3- (4- (pyridin-2-ylcarbamoyl) phenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidine (9)
Figure BDA0001365649630000481
Step one, synthesizing (R) -3- (3-iodo-4-amino-1H-pyrazolo [3,4-d ] pyrimidine-1-yl) piperidine carboxylic acid tert-butyl ester (CDA-132)
3-iodo-1H- [3,4-d ] pyrazolopyrimidin-4-amine (B,200mg,0.77mmol) was dissolved in dry tetrahydrofuran, and triphenylphosphine (603mg,2.30mmol), (S) -1-tert-butoxycarbonyl-3-hydroxypiperidine (310mg,1.54mmol) was added. Diisopropyl azodicarboxylate (0.92mL,4.60mmol) was added dropwise under nitrogen. After the addition was complete, the mixture was stirred at 40 ℃ for 12 hours. After the reaction, tetrahydrofuran was removed by rotation, and the mixture containing CDA-132 was purified by column chromatography to obtain 580 mg.
Step two, synthesizing (R) -3- (3- (4- (pyridine-2-yl carbamoyl) phenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidine-1-yl) piperidine carboxylic acid tert-butyl ester (CDA-133):
the reaction flask was charged with the mixture containing CDA-132 of the previous step (580mg), N- (pyridin-2-yl) -4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzamide (500mg,1.54mmol), ethylene glycol dimethyl ether (6mL,2mol/L), and sodium carbonate solution (3mL, 2M). After oxygen was removed and nitrogen purged, [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (63mg,0.08mmol) was added, heated to 95 ℃ under nitrogen protection and stirred for 12 hours. After cooling, 50mL of water was added, extraction was carried out three times with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, and after rotary removal of the solvent, column purification was carried out to obtain 360mg of a mixture containing CDA-133.
Step three Synthesis of (R) -4- (4-amino-1- (piperidin-3-yl) -1H-pyrazolo [3,4-d ] pyrimidin-3-yl) -N- (pyridin-2-yl) benzamide (CDA-136):
the last step comprisesThe mixture of CDA-133 was dissolved in 3mL of dichloromethane, 6mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 12 hours. After the solvent was removed by evaporation, water and methanol were added, and HPLC purification gave the trifluoroacetate salt of CDA-136, which was lyophilized to give 254mg of solid. The total yield of the three steps is 63 percent. HPLC purification conditions: initial acetonitrile proportion of 10%, retention time t 1/210 minutes.1HNMR(MeOD-d4400MHz 8.50(s,1H),8.48(s,1H),8.37(t,1H, J ═ 7.94Hz),8.26(d,2H, J ═ 7.94Hz),8.04(d,1H, J ═ 8.53Hz),7.94(d,2H, J ═ 7.94Hz),7.58(t,1H, J ═ 6.44Hz),5.44-5.28(m,1H),3.81-3.74(m,2H),3.54-3.42(m,1H),3.32-3.22(m,1H),2.46-2.28(m,2H),2.24-2.14(m,1H),2.12-1.98(m, 1H). Theoretical calculation of ESI-MS C22H22N8O[M+H]+415.2; the experiment shows that: 415.2.
step four Synthesis of the Compound (R) -2, 3-butenoyl-3- (3- (4- (pyridin-2-ylcarbamoyl) phenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidine (9):
3-Butynoic acid (10mg,0.13mmol) was dissolved in dry dichloromethane, 2-chloro-1-methylpyridine iodide (25mg,0.11mmol) was added, and the mixture was stirred at room temperature for 1 hour. A solution of CDA-136 trifluoroacetate (50mg,0.1mmol) and diisopropylethylamine (0.2mL) in dry dichloromethane (3mL) was added and the reaction was allowed to proceed at room temperature for 1 hour. After removal of the solvent by evaporation, water and methanol were added and purified by HPLC to give the trifluoroacetate salt of compound 9, which was lyophilized to give 23mg of solid. The yield of this step was 41%. HPLC purification conditions: the initial acetonitrile proportion was 10%, the retention time t ═ 18 minutes.1H NMR(MeOD-d4400MHz 8.44(s,1H),8.42(s,1H),8.22(d,2H, J-8.5 Hz),8.18-8.08(m,2H),7.92(d,2H, J-6.76 Hz),7.45-7.35(m,1H),6.30-6.23(m,0.6H),6.12-6.04(m,0.4H),5.28-4.93(m,3H),4.53-4.45(m,0.5H),4.32-4.22(m,0.5H),4.21-4.10(m,0.5H),4.08-4.01(m,0.5H),3.99-3.87(m,0.5H),3.71-3.62(m,0.5H), 3.52-3.52 (m, 3.83-2H), 2.06-1H, 2H, 1H, 4.3H, 4H, 4.3H, 4H, 4.5H, 4.3. Theoretical calculation of ESI-MS C26H24N8O2[M+H]+481.2; the experiment shows that: 481.2.
example 10: synthesis of (S) -2, 3-butenoyl-2- (1- (4-phenoxyphenyl) -8-aminoimidazo [1,5-a ] pyrazin-3-yl) piperidine (10)
Figure BDA0001365649630000501
The method comprises the following steps: synthesis of the Compound benzyl (S) -2- (1- (4-phenoxyphenyl) -8-aminoimidazo [1,5-a ] pyrazin-3-yl) -piperidin-1-ylcarboxylate (C092)
In a 100mL glass bottle, 2- (4-phenoxyphenyl) -4,4,5, 5-tetramethyl- [1,3,2]Dioxolane (414mg,1.4mmol), C049(280mg,0.65mmol) and 1, 2-dimethoxyethane (8 mL). To the above solution was added aqueous sodium carbonate (2M,4mL) and the solution was deoxygenated. Then, add [1,1' -bis (diphenylphosphino) ferrocene to the reaction flask]Palladium dichloride dichloromethane complex (50mg.0.07mmol) and deoxygenated again. The reaction was refluxed overnight, cooled to room temperature, and the aqueous phase was extracted three times with ethyl acetate (50 ml. reaction. the organic phases were combined and washed with saturated brine, dried, concentrated and purified by silica gel column to give the objective compound C092(220 mg).1H NMR(CDCl3400MHz) 7.63(d,2H, J ═ 8.17Hz),7.43-7.30(m,7H),7.20-7.05(m,5H),7.00-6.85(m,1H),5.27-4.97(m,4H),4.08-3.96(m,1H),2.88(t,1H, J ═ 12.80Hz),2.61-2.45(m,1H),2.45-2.32(m,1H),2.02-1.50(m, 5H). Theoretical calculation of ESI-MS C31H30N5O3[M+H]+520.2; the experiment result is 520.2.
Step two: synthesis of 1- (4-phenoxyphenyl) -3- (piperidin-2-yl) -8-amino-imidazo [1,5-a ] pyrazine (C096)
In a 50mL glass bottle, C092(220mg) was added. A solution of hydrobromic acid in acetic acid (33%, 8mL) was added via syringe. After stirring at room temperature for 1 hour, 20mL of water was added to quench. This solution was purified by HPLC to give the trifluoroacetate salt of the target compound C096, which was lyophilized to give 148mg of solid. HPLC purification conditions: initial acetonitrile proportion 15%, retention time t1/216 minutes.1H NMR (MeOD-d4,400mhz):7.92(d,1H, J ═ 5.94Hz),7.70(d,2H, J ═ 8.56Hz),7.43(t,2H, J ═ 7.89Hz),7.25 to 7.06(m,6H),5.16 to 4.90(m,1H),3.65 to 3.54(m,1H),3.40 to 3.21(m,1H),2.37 to 2.26(m,1H),2.14 to 1.97(m,3H),1.94 to 1.78(m, 2H). Theoretical calculation of ESI-MS C23H24N5O[M+H]+386.2; the test result is 386.2.
Step three: synthesis of the Compound (S) -2, 3-butenoyl-2- (1- (4-phenoxyphenyl) -8-aminoimidazo [1,5-a ] pyrazin-3-yl) -1-piperidine (10)
In a 50mL glass vial, 3-butynoic acid (17mg,0.2mmol) and dry dichloromethane (3mL) were added followed by 2-chloro-1-methylpyridine iodide (30mg,0.13mmol) and the solution was stirred at room temperature for 1 hour. A solution of C096 trifluoroacetate salt (50mg,0.1mmol) and diisopropylethylamine (0.2mL) in dichloromethane (3mL) was added to the above reaction flask and stirred at room temperature for 40 minutes. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of the target compound 10, which was lyophilized to give 18.3mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 30%, retention time t1/219.5 min.1H NMR (MeOD-d4,400mhz) 7.85 to 7.75(m,1H),7.73 to 7.65(m,2H),7.45 to 7.38(m,2H),7.24 to 7.14(m,3H),7.14 to 7.07(m,2H),7.01 to 6.94(m,1H),6.26(t,1H, J ═ 6.62Hz),6.29 to 6.14(m,1H),5.25(d,2H, J ═ 6.50Hz),4.10 to 3.83(m,1H),2.50 to 2.27(m,2H),2.10 to 1.93(m,1H),1.90 to 1.73(m, 3H). Theoretical calculation of ESI-MS C27H26N5O2[M+H]+452.2; the experiment found that 452.1.
Example 11: synthesis of the Compound (R) -2, 3-butynenoyl 3- (3- (4-phenoxyphenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) pyrrolidine
Figure BDA0001365649630000511
Step one, synthesizing (R) -3- (3- (4-phenoxyphenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidine-1-yl) pyrrolidine carboxylic acid tert-butyl ester (CDA-148-1):
intermediate C (100mg,0.33mmol) was dissolved in dry tetrahydrofuran and triphenylphosphine (346mg,1.32mmol) and (S) -1-tert-butoxycarbonyl-3-hydroxypyrrolidine (125mg,0.66mmol) were added. Diisopropyl azodicarboxylate (0.40mL,1.98mmol) was added dropwise under nitrogen. After the addition was complete, the mixture was stirred at 40 ℃ for 12 hours. After the reaction, tetrahydrofuran was removed by rotation, and the mixture containing CDA-148-1 (400 mg) was obtained by column purification.
Step two, synthesizing (R) -3- (4-phenoxyphenyl) -1- (pyrrolidine-3-yl) -1H-pyrazolo [3,4-d ] pyrimidine-4-amine (CDA-148):
the mixture containing CDA-148-1 obtained in the previous step was dissolved in 4mL of dichloromethane, 4mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 12 hours. After the solvent was removed by evaporation, water and methanol were added, and HPLC purification gave the trifluoroacetate salt of CDA-148, which was lyophilized to give 120mg of solid. The yield of the two steps is 74 percent. HPLC purification conditions: the initial acetonitrile proportion was 15%, and the retention time t was 15 minutes.1HNMR(MeOD-d4400MHz) 8.45(s,1H),7.73(d,2H, J-8.50 Hz),7.39(t,2H, J-7.92 Hz),7.20-7.05(m,5H),5.89-5.81(m,1H),3.96-3.85(m,2H),3.83-3.74(m,1H),3.67-3.57(m,1H),2.73-2.61(m,1H),2.60-2.50(m, 1H). Theoretical calculation of ESI-MS C21H20N6O[M+H]+373.2; the experiment shows that: 373.1.
step three: synthesis of the Compound (R) -2, 3-butynoyl3- (3- (4-phenoxyphenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) pyrrolidine (11)
In a 50mL glass vial, 3-butynoic acid (11mg,0.14mmol) and dry dichloromethane (3mL) were added followed by 2-chloro-1-methylpyridine iodide (31.5mg,0.14mmol) and the solution was stirred at room temperature for 1 hour. A solution of CDA-148 in trifluoroacetate (60mg,0.12mmol) and diisopropylethylamine (0.2mL) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 30 min. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of the target compound 11, which was lyophilized to give 23.8mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 20%, retention time t1/222.5 minutes.1H NMR (MeOD-d4,400MHz):8.45-8.40(m,1H),7.71-7.64m,2H),7.44-7.37(m,2H),7.22-7.12(m,3H),7.12-7.05(m,2H),6.10(t,0.5H, J ═ 6.53Hz),6.02(t,0.5H, J ═ 6.53Hz),5.75-5.62(m,1H),5.30-5.22(m,2H),4.22-3.82(m,3.5H),3.81-3.66(m,0.5H),2.69-2.47(m, 2H). Theoretical calculation of ESI-MS C25H23N6O2[M+H]+439.2; the test result is 439.1.
Example 12: synthesis of the Compound 1- (3- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) azetidin-1-yl) -2, 3-buty-len-1-one
Figure BDA0001365649630000531
Step one, synthesis of tert-butyl 3- (3- (4-phenoxyphenyl) -4-amino-1H-pyrazolo [3,4-d ] pyrimidin-1-yl) azetidinecarboxylate (CDB 004):
intermediate C (100mg,0.33mmol) and cesium carbonate (215mg,0.66mmol) were dissolved in dry DMF. Tert-butyl 3- (methylsulfonyloxy) azetidinecarboxylate (166mg,0.66mmol) was dissolved in 2mL dry DMF under nitrogen, added dropwise to the system and heated to 75 deg.C with stirring for 24 h. After the reaction, water was added, and extraction was carried out three times with ethyl acetate, and the organic phases were combined, washed three times with saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column to obtain 45mg of the objective compound CDB 004.
Step two, synthesis of 3- (4-phenoxyphenyl) -1- (azetidin-3-yl) -1H-pyrazolo [3,4-d ] pyrimidine-4-amine (CDB 007):
CDB004(45mg,0.10mmol) was dissolved in 2mL of dichloromethane, 2mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 12 hours. After removal of the solvent by evaporation, water and methanol were added and purified by HPLC to give the trifluoroacetate salt of CDB007 which was lyophilized to give 45mg of solid. The yield of the two steps is 29 percent. HPLC purification conditions: the initial acetonitrile proportion was 20%, the retention time t ═ 11 minutes.1H NMR(MeOD-d4400MHz) 8.43(s,1H),7.76(d,2H, J ═ 8.56Hz),7.44-7.38(m,2H),7.22-7.15(m,3H),7.13-7.08(m,2H),6.08-5.99(m,1H),4.80-4.63(m, 4H). Theoretical calculation of ESI-MS C20H18N6O[M+H]+359.2; the experiment shows that: 359.1.
step three, synthesizing 1- (3- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidine-1-yl) azetidin-1-yl) -2, 3-butralin-1-one (12):
in a 25mL glass vial, 3-butynoic acid (10mg,0.12mmol) and dry dichloromethane (3mL) were added followed by 2-chloro-1-methylpyridine iodide (28mg,0.12mmol) and the solution was stirred at room temperature for 1 hour.A solution of the trifluoroacetate salt of CDB007 (45mg,0.09mmol) and diisopropylethylamine (0.2mL) in dichloromethane (3mL) was added to the reaction flask and stirred at room temperature for 60 minutes. The dichloromethane was evaporated using a rotary evaporator and the resulting dope was purified by HPLC to give the trifluoroacetate salt of target compound 12, and lyophilized to give 15mg of a solid. HPLC purification conditions: initial acetonitrile proportion of 20%, retention time t1/222.5 minutes.1H NMR (MeOD-d4,400mhz):8.41(s,1H),7.73(d,2H, J ═ 8.72Hz),7.45-7.38(m,2H),7.23-7.15(m,3H),7.14-7.09(m,2H),5.93-5.85(m,1H),5.81(t,1H, J ═ 6.68Hz),5.32(d,2H, J ═ 6.72Hz),4.83-4.74(m,2H),4.64-4.53(m, 2H). Theoretical calculation of ESI-MS C24H20N6O2[M+H]+425.3; the test result is 425.3.
The structures of the above compounds are summarized as follows:
Figure BDA0001365649630000541
Figure BDA0001365649630000551
Figure BDA0001365649630000561
by the above-described synthesis method, the following compounds can also be synthesized:
Figure BDA0001365649630000562
Figure BDA0001365649630000571
Figure BDA0001365649630000581
Figure BDA0001365649630000591
Figure BDA0001365649630000601
Figure BDA0001365649630000611
Figure BDA0001365649630000621
Figure BDA0001365649630000631
Figure BDA0001365649630000641
Figure BDA0001365649630000651
Figure BDA0001365649630000661
Figure BDA0001365649630000671
Figure BDA0001365649630000681
Figure BDA0001365649630000691
Figure BDA0001365649630000701
experimental example 1: method for testing cell activity of compound
A20 mM compound stock solution was prepared by dissolving a sample to be tested in 100% dimethyl sulfoxide. Compounds were diluted with 100% dimethylsulfoxide to the highest concentration required for the experiment (2.5mM or 62.5 μ M).
Add 5. mu.L of compound solution to 96-well flat bottom clear cell culture plates and dilute 2-fold sequentially with cell culture medium (containing 10% fetal calf serum) to a gradient of desired compound concentration (44.6. mu.M-0.0218. mu.M or 1.116-0.544 nM). Finally, 70 μ L of the cell solution to be tested was added to each well at a cell density of 3000-. In the experiment, two control groups were set, in addition to the compound tested: 1) cells and medium were added, but no compound control; 2) the medium containing 10% fetal bovine serum was added, but the cell-free and compound-free group. After culturing the 96-well plate in a 37 ℃ cell culture chamber containing 5% carbon dioxide for 4 days, 14. mu.L of CCK-8 reagent was added to each well, followed by incubation at 37 ℃ for 2 to 4 hours. The 96-well plate was read with a TECAN microplate reader, and the absorbance at a wavelength of 450nm was taken.
The effect of different concentrations of compound on cell activity was calculated using the following formula: [ experimental group absorbance-absorbance of medium containing 10% fetal bovine serum (cell-free and compound-free group) ]/[ absorbance of cell-free and compound-free group-absorbance of medium containing 10% fetal bovine serum (cell-free and compound-free group) ] × 100%.
The data were processed using GraphPad Prism 6 software, and the IC was obtained50The values are the compound concentrations corresponding to a 50% inhibition of cell growth.
For example: based on the above test method for cell growth inhibitory activity of compounds, the cell growth inhibitory activity measured in the Raji cell line of B-cell lymphoma is shown in fig. 1. Wherein, IC of Compound 450IC value of 0.36 + -0.05 μ M and Ibrutinib (Ibrutinib)50The value is 3.42 +/-1.61 mu M, and the activity of the compound 4 is superior to that of ibrutinib.
For example, based on the above test methods for cell growth inhibitory activity of compounds 2-4 and Ibrutinib (Ibrutinib) in B-cell lymphoma Raji cell line, B-cell lymphoma NAMALWA cell line, intestinal cancer cell lines LOVO, HCT-116, liver cancer cell line HepG2, breast cancer cell line MDA-MB-231, cervical cancer cell lineCell growth inhibitory Activity IC in Hela cell line, gastric cancer cell line NCI-N87 and Lung cancer cell lines NCI-H1975 and NCI-H165050The values are given in the table below. On the whole, the growth inhibition activity of the compounds 2-4 on the tumor cell strains is better than that of ibrutinib.
Figure BDA0001365649630000711
As can be seen from the data in the table, the growth inhibitory activity of the compounds 2 and 3 on B cells is significantly higher than that of the marketed drug Ibrutinib (Ibrutinib), and the average value of IC50 of Ibrutinib is 85 times of the IC50 value of the compound 2, which indicates that the cell growth inhibitory activity of the compound 2 in the B cell lymphoma Raji cell strain has significant superiority.
Compared with ibrutinib, the cell growth inhibitory activity (IC50) of the compounds 2 and 3 in B-cell lymphoma cell strains NAMALWA, liver cancer cell strains HepG2, breast cancer cell strains MDA-MB-231, intestinal cancer cell strains HCT-116, cervical cancer cell strains Hela, stomach cancer cell strains NCI-N87, lung cancer cell strains NCI-H1975 and NCI-H1650 is improved by more than 20-100, which shows that the cell growth inhibitory activity of the compounds 2 and 3 is obviously improved in other cancer cell strains as well as B-cell lymphoma cell strains.
Based on the cell activity test method of the compounds, IC of the compounds 1-10 and ibrutinib in Raji cell line of B cell lymphoma, NAMALWA cell line of B cell lymphoma and HCT-116 cell line of intestinal cancer is tested50The values are given in the table below.
Figure BDA0001365649630000721
Experimental example 2: method for testing kinase activity of compound BTK
The test method is based on the principle of solid-phase bead adsorption induced fluorescence deviation and is used for testing the kinase activity.
First, KR buffer solution (10mM Tris-HCl,10mM MgCl) was used2,0.01%Tween-20,0.05%NaN3,1mM DTT,2mM MnCl2pH 7.2) to a concentration of 0.4U/mL. Secondly, benefit fromTest compound concentrations were 2mM to 63.2nM diluted with DMSO gradient. The DMSO solution of the diluted test compound described above was then diluted 50-fold with KR buffer solution on a 384-well plate. The gradient of test compound formulated as described above ensures a final concentration gradient of test compound in the 384 well plates of 10. mu.M to 0.316nM per well.
In a 384 well plate, 5. mu.L of the above test compound concentration gradient solution and 5. mu.L of 0.4U/mL BTK kinase solution were added to each well. The test compound and BTK kinase were mixed well for 60 minutes at room temperature, and then 5 μ L of fluorescently labeled polypeptide substrate (200nM) in KR buffer was added to each well, so that the final concentration of polypeptide substrate per well was 50 nM. Finally, 5. mu.L of 20. mu.M ATP in KR buffer was added to each well to give a final ATP concentration of 5. mu.M per well. After the phosphorylation reaction was carried out at room temperature for 2 hours, the reaction was quenched. After incubation at room temperature for 60 minutes, the fluorescence polarization values were read using a microplate reader. Since the phosphorylated substrate is adsorbed and immobilized by the solid phase bead, the difference (mP) in the fluorescence polarization value of 535nM can be read both horizontally and vertically. Based on the value of mP for the reference well without ATP added, Δ mP per well values can be calculated. The IC of the test compound can be determined by processing the data with GraphPad Prism 6 software and plotting Δ mP against the concentration gradient50The value is obtained.
Based on the above method, the BTK kinase inhibitory IC of the synthesized compound was determined50The values are as follows:
Figure BDA0001365649630000731

Claims (18)

1. a compound represented by the general formula (I):
Figure FDA0002404493110000011
in the general formula (I):
w is selected from the following groups:
Figure FDA0002404493110000012
Figure FDA0002404493110000013
n is 0, 1 or 2;
m is 0, 1,2, 3 or 4;
n + m is 2,3 or 4;
l represents-O-, -CO-NH-, -NH-CO-, -NR6-, -S-or-CH2-;
R1Represents a C6-C12 aryl or a 5-10 membered monocyclic heteroaryl group containing 1-4 heteroatoms selected from N, O, S; the aryl and heteroaryl groups are optionally substituted with 1-2 substituents selected from halogen, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen-substituted C1-C4 alkyl, halogen-substituted C1-C4 alkoxy, and amino;
R2、R3and R4Each independently selected from hydrogen and C1-C4 alkyl, said C1-C4 alkyl being optionally substituted with 1-3 substituents selected from halogen, hydroxy, amino, C1-C4 alkoxy, -NH- (C1-C4 alkyl), -N (C1-C4 alkyl) (C1-C4 alkyl);
B1、B2、B3and B4Each independently represents N or-C (R)5);
R5Each independently selected from hydrogen, halogen, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy, and amino;
R6each independently selected from hydrogen or C1-C4 alkyl.
2. The compound or salt according to claim 1, wherein,
w is selected from
Figure FDA0002404493110000014
3. A compound or a salt thereof as claimed in claim 1 wherein L represents-O-or-NH-CO-.
4. The compound according to claim 1, wherein the aryl group is a phenyl group and the heteroaryl group is a pyridyl group, or a salt thereof.
5. The compound or salt according to claim 1, wherein R is2、R3And R4Are both hydrogen.
6. The compound or salt according to claim 1, wherein B is1、B2、B3And B4At most two of which are N.
7. The compound or salt according to claim 6, wherein B is1、B2、B3And B4At most one of which is N.
8. The compound or salt according to claim 1, wherein R is6Each independently selected from hydrogen, methyl and ethyl.
9. The compound according to any one of claims 1 to 8, wherein the compound of the general formula (I) is a compound of the general formula (I-1):
Figure FDA0002404493110000021
wherein R is1、R2、R3、R4、B1、B2、B3、B4N, m, X and W are as defined in the corresponding claims.
10. The compound according to any one of claims 1 to 8, wherein the compound of the general formula (I) is a compound of the general formula (I-2):
Figure FDA0002404493110000022
wherein R is1、R2、R3、R4、B1、B2、B3、B4N, m, X and W are as defined in the corresponding claims.
11. The compound according to any one of claims 1 to 8, wherein the compound of the general formula (I) is a compound selected from the following general formulae (I-3) to (I-7):
Figure FDA0002404493110000023
Figure FDA0002404493110000031
wherein, L and R1Are as defined in the corresponding claims;
B2represents N or-C (R)5);
R5Each independently selected from hydrogen, halogen, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, halogen substituted C1-C4 alkyl, halogen substituted C1-C4 alkoxy, and amino.
12. A compound according to claim 1, or a salt thereof, wherein the compound of formula (I) is selected from the following compounds:
Figure FDA0002404493110000032
Figure FDA0002404493110000041
Figure FDA0002404493110000051
Figure FDA0002404493110000061
Figure FDA0002404493110000071
Figure FDA0002404493110000081
Figure FDA0002404493110000091
Figure FDA0002404493110000101
Figure FDA0002404493110000111
Figure FDA0002404493110000121
Figure FDA0002404493110000131
13. a pharmaceutical composition comprising a compound of any one of claims 1-12 or a salt thereof.
14. The pharmaceutical composition of claim 13, wherein said pharmaceutical composition is a BTK inhibitor.
15. The pharmaceutical composition of claim 13, wherein said pharmaceutical composition is a B cell activation inhibitor.
16. Use of a compound of any one of claims 1 to 12 or a salt thereof for the manufacture of a medicament for the prevention and/or treatment of a disease associated with abnormal B cell activity and/or BTK.
17. The use of claim 16, wherein the disease associated with abnormal B cell activity and/or BTK is an allergic disease, an autoimmune disease, an inflammatory disease, a thromboembolic disease, or a cancer.
18. The use of claim 17, wherein the cancer is one or more selected from non-hodgkin's lymphoma, breast cancer, liver cancer, intestinal cancer, stomach cancer, pancreatic cancer, lung cancer and cervical cancer.
CN201710639753.0A 2016-08-17 2017-07-31 Compound containing conjugated allene structure, pharmaceutical composition and application thereof Expired - Fee Related CN107759602B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610679711 2016-08-17
CN2016106797115 2016-08-17

Publications (2)

Publication Number Publication Date
CN107759602A CN107759602A (en) 2018-03-06
CN107759602B true CN107759602B (en) 2020-04-21

Family

ID=61265253

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710639753.0A Expired - Fee Related CN107759602B (en) 2016-08-17 2017-07-31 Compound containing conjugated allene structure, pharmaceutical composition and application thereof

Country Status (1)

Country Link
CN (1) CN107759602B (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110776511B (en) * 2018-07-26 2022-12-30 正大天晴药业集团股份有限公司 Quinoline ring-containing BTK inhibitors
CA3112656A1 (en) 2018-09-14 2020-03-19 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Furo[3,4-b]pyrrole-containing btk inhibitor
CN109134472B (en) * 2018-09-20 2020-02-11 浙江理工大学 Synthesis process of 3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidine-4-amine
CN112142745B (en) * 2019-06-27 2023-03-21 杭州和正医药有限公司 Casein kinase 1 epsilon inhibitor, pharmaceutical composition and application thereof
CN111018865B (en) * 2019-10-17 2021-01-15 山东大学 1-substituted benzyl pyrazolopyrimidine derivative and preparation method and application thereof
TW202220994A (en) * 2020-08-17 2022-06-01 大陸商貝達藥業股份有限公司 Bicyclic compounds, pharmaceutical compositions comprising the same and use thereof
CN115073468B (en) * 2021-03-15 2023-12-22 药雅科技(上海)有限公司 Preparation and application of imidazopyrazines BTK inhibitor
CN113968860B (en) * 2021-11-05 2023-06-06 杭州医学院 Reversible BTK inhibitor and synthesis method and application thereof
CN114044796A (en) * 2021-11-18 2022-02-15 鄂尔多斯应用技术学院 Stereoselective synthesis method of tetra-substituted allyl azide

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101674834A (en) * 2007-03-28 2010-03-17 环状药物公司 inhibitors of bruton's tyrosine kinase
CN102918040A (en) * 2010-05-31 2013-02-06 小野药品工业株式会社 Purinone derivative
WO2015058084A1 (en) * 2013-10-18 2015-04-23 Medivation Technologies, Inc. Heterocyclic compounds and methods of use
CN104603124A (en) * 2012-08-10 2015-05-06 勃林格殷格翰国际有限公司 Heteroaromatic compounds as bruton's tyrosine kinase (btk) inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101674834A (en) * 2007-03-28 2010-03-17 环状药物公司 inhibitors of bruton's tyrosine kinase
CN102918040A (en) * 2010-05-31 2013-02-06 小野药品工业株式会社 Purinone derivative
CN104603124A (en) * 2012-08-10 2015-05-06 勃林格殷格翰国际有限公司 Heteroaromatic compounds as bruton's tyrosine kinase (btk) inhibitors
WO2015058084A1 (en) * 2013-10-18 2015-04-23 Medivation Technologies, Inc. Heterocyclic compounds and methods of use

Also Published As

Publication number Publication date
CN107759602A (en) 2018-03-06

Similar Documents

Publication Publication Date Title
CN107759602B (en) Compound containing conjugated allene structure, pharmaceutical composition and application thereof
KR20220119088A (en) KRAS mutant protein inhibitor
CN105358550B (en) Histone deacetylase inhibitors
KR102057877B1 (en) Nitrogenous heterocyclic derivatives and their application in drugs
CN111484477A (en) Benzopyridone heterocyclic compound and application thereof
CN113286794A (en) KRAS mutein inhibitors
CN111285850A (en) Isoindoline compounds, preparation method thereof, pharmaceutical composition and application thereof
WO2001007411A1 (en) Biarylurea derivatives
CN108069976A (en) A kind of nitrogenous macrocyclic compounds, its preparation method, pharmaceutical composition and application
CN113748114A (en) Quinazoline compound and application thereof in medicine
EP3124486B1 (en) Substituted nitrogen-containing heterocyclic derivatives, pharmaceutical compositions comprising the same and applications of antitumor thereof
JP6887430B2 (en) Deuterated Brigatinib derivatives, pharmaceutical compositions containing such derivatives, and their use.
TW202003466A (en) ANTAGONISTS OF HUMAN INTEGRIN α4β7
EP2411368A1 (en) Nicotinamide derivatives, preparation thereof, and therapeutic use thereof as anticancer drugs
KR20200039616A (en) New inhibitor of cyclin-dependent kinase CDK9
CN111094288A (en) Fused ring derivatives having MGAT-2 inhibitory activity
TW201920196A (en) Heteroaromatic compounds as VANIN inhibitors
CN112824420B (en) Compounds useful as EGFR kinase inhibitors and uses thereof
WO2015049629A1 (en) Imidazoquinoline compounds as bromodomain inhibitors
CN111714628A (en) Small molecule PD-1/PD-L1 inhibitor, pharmaceutical composition of small molecule PD-1/PD-L1 inhibitor and PD-L1 antibody and application of small molecule PD-1/PD-L1 inhibitor and pharmaceutical composition
CA3047136A1 (en) Inhibitors of mutant isocitrate dehydrogenases and compositions and methods thereof
CN114127063A (en) Pyrimido five-membered heterocyclic compounds and application thereof as mutant IDH2 inhibitor
WO2022048631A1 (en) Compound having antitumor activity and use thereof
CN114315818A (en) Benzamide compound and application thereof
TWI729094B (en) Endosulfonamide compounds and methods of use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20200421

Termination date: 20210731

CF01 Termination of patent right due to non-payment of annual fee