CN115707705A - Aromatic heterocyclic compound, preparation method and application thereof - Google Patents

Aromatic heterocyclic compound, preparation method and application thereof Download PDF

Info

Publication number
CN115707705A
CN115707705A CN202110961711.5A CN202110961711A CN115707705A CN 115707705 A CN115707705 A CN 115707705A CN 202110961711 A CN202110961711 A CN 202110961711A CN 115707705 A CN115707705 A CN 115707705A
Authority
CN
China
Prior art keywords
alkyl
cycloalkyl
compound
haloalkyl
preparation
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.)
Pending
Application number
CN202110961711.5A
Other languages
Chinese (zh)
Inventor
何婷
任云
刘金明
田强
宋宏梅
葛均友
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority to CN202110961711.5A priority Critical patent/CN115707705A/en
Publication of CN115707705A publication Critical patent/CN115707705A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides aromatic heterocyclic compounds, a preparation method and application thereof. Specifically, the invention provides a compound shown in formula I or pharmaceutically acceptable salts, stereoisomers, tautomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs thereof, and a pharmaceutical composition, a preparation method and application thereof in preventing and/or treating tumors or autoimmune diseases.

Description

Aromatic heterocyclic compound, preparation method and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to an aromatic heterocyclic compound, and a preparation method and application thereof.
Background
Bruton's Tyrosine Kinase (BTK), a member of the non-receptor protein tyrosine kinase Tec family, is expressed in all hematopoietic cells except T cells and terminally differentiated plasma cells, mainly in B cells and myeloid cells. BTK is a key kinase in the B cell antigen receptor (BCR) signaling pathway, and is mainly involved in the transduction and amplification of various intracellular and extracellular signals in B lymphocytes, including Toll-like receptor (TLR), chemokine and Fc receptor signaling, regulating the proliferation, differentiation and apoptosis of normal B cells.
BTK is abnormally expressed in various B cell tumors, so that BCR signal pathways are abnormally activated, and various malignant lymphomas such as chronic lymphocytic leukemia, mantle cell lymphoma and the like are initiated. At present, clinical verification shows that BTK is a safe and effective excellent target for B cell tumor represented by chronic lymphocytic leukemia. In the field of autoimmune diseases, as early as the basic research of the eighth ninety years of the last century, it has been found that BTK deficient mice are difficult to suffer from autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. Abnormal activation of BCR signaling pathway due to BTK overexpression can lead to obvious increase of the number of autoreactive B cells, and secretion of a large amount of autoantibodies induces autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus.
Currently, there are 5 BTK inhibitors listed worldwide, namely Ibrutinib (Ibrutinib), acarabutinib (zanubrutinib), tirabrutinib (tiabratinib) and orbutiib (orelburtinib), which are BTK C481S site covalent irreversible inhibitors, and the approved indications mainly belong to B cell malignancies. In recent years, the application of BTK inhibitors to autoimmune diseases such as multiple sclerosis, systemic lupus erythematosus, pemphigus etc. has achieved positive results in clinical studies. For example, GDC-0853 most progressed to stage III in multiple sclerosis and BMS-986142 most progressed to stage II in Sjogren syndrome and rheumatoid arthritis.
However, with the clinical use of BTK inhibitors, the problem of C481S resistance mutations occurs in 80% of relapsed patients, resulting in poor prognostic survival in the patients. Therefore, there is significant clinical and market value in developing a new generation of BTK inhibitors that can effectively overcome resistance to the C481S mutation.
Disclosure of Invention
The invention aims to provide a compound with a novel structure and an inhibiting effect on BTK, in particular to a BTK-C481S mutant, a pharmaceutical composition and a preparation method thereof, and application of the compound in preventing and/or treating diseases mediated by BTK wild type or C481S mutation, such as tumors or autoimmune diseases.
In one aspect, the present application provides a compound having the structure of formula (I), or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
Figure BDA0003222255240000021
x is selected from N and CH;
l is selected from-C (O) -NH-) -NH-C (O) -, -C 1-6 alkylene-NH-C (O) -and-C 1-6 alkylene-C (O) -NH-;
when X is N, L is not-C 1-6 alkylene-NH-C (O) -;
R 1 is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl, 3-12 membered heterocyclyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution;
each R 2 Each independently selected from H, halogen, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl, -OR 5 and-NR 5 (R 6 );
R 3 Is selected from C 6-10 Aryl and 5-10 membered heteroaryl, said aryl and heteroaryl optionally substituted with one or more R 7 Substitution;
R 4 selected from H, halogen, nitro, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -C 1-6 alkyl-OH, -C 1-6 alkyl-O-C 1-6 Alkyl, -OR 8 、-SR 8 、-S(O)R 8 、-S(O) 2 R 8 、-NR 9 (R 10 )、-C(O)NR 9 (R 10 )、-C(O)R 11 、-S(O) 2 -NR 9 (R 10 )、C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted with one or more halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 5 and R 6 Each independently selected from H and C 1-6 An alkyl group; or,
R 5 and R 6 Together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl;
R 7 selected from H, halogen, cyano, nitro, oxo, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl, -OR 12 、-NR 12 (R 13 ) 3-12 membered heterocyclic group, C 6-10 Aryl and 5-10 membered heteroaryl;
R 8 selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted by one or more substituents selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 9 selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 10 is selected from H and C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted by one or more substituents selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl; or,
R 9 、R 10 together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl which may optionally be substituted by one or more groups selected from oxo, halogen, hydroxy, amino, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl and-O-C 1-6 Alkyl substituent substitution;
R 11 is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more substituents selected from the group consisting of halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 12 is selected from C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl, 3-12 membered heterocyclyl, C 6-10 Aryl and 5-10 membered heteroaryl, said cycloalkyl, heterocyclyl, aryl and heteroaryl being optionally substituted by one or more substituents selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 13 selected from H and C 1-6 An alkyl group;
m is 0, 1,2,3 or 4.
In some embodiments, R 1 Is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl, 3-12 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from H, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -C 1-6 alkyl-OH, -C 1-6 alkyl-O-C 1-6 Alkyl, -OR 8 、-SR 8 、-S(O)R 8 、-S(O) 2 R 8 、-NR 9 (R 10 )、-C(O)NR 9 (R 10 )、-C(O)R 11 and-S (O) 2 -NR 9 (R 10 );
R 8 Selected from H, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 9 selected from H, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 10 selected from H, C 1-6 Alkyl and C 1-6 A haloalkyl group; or,
R 9 、R 10 together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl which may optionally be substituted by one or more groups selected from oxo, halogen, hydroxy, amino, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl and-O-C 1-6 Alkyl substituent substitution;
R 11 is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 A substituent of a haloalkyl group.
In some embodiments, R 1 Is selected from C 1-6 Alkyl radical, C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from H, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR 8 、-C(O)NR 9 (R 10 ) and-C (O) R 11
R 8 Selected from H, C 1-4 Alkyl and C 1-4 A haloalkyl group;
R 9 selected from H, C 1-4 Alkyl and C 1-4 A haloalkyl group;
R 10 is selected from H and C 1-4 Alkyl and C 1-4 A haloalkyl group;
R 11 is selected from C 1-6 Alkyl radical, C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-4 Alkyl and C 1-4 Substituted with a haloalkyl.
In some embodiments, R 1 Is selected from C 1-6 Alkyl radical, C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from H, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, hydroxy, -C (O) NR 9 (R 10 ) and-C (O) R 11
R 9 Is selected from C 1-4 An alkyl group;
R 10 is selected from C 1-4 An alkyl group;
R 11 is selected from C 1-6 Alkyl and C 3-6 Cycloalkyl, said alkyl and cycloalkyl being optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-4 Alkyl and C 1-4 A substituent of a haloalkyl group.
In some embodiments, R 1 Is selected from C 1-4 Alkyl radical, C 5-6 Cycloalkyl and 5-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from fluorine, chlorine, C 1-6 Alkyl radical, C 1-6 Haloalkyl, hydroxy, -C (O) NR 9 (R 10 ) and-C (O) R 11
R 9 Is selected from C 1-4 An alkyl group;
R 10 is selected from C 1-4 An alkyl group;
R 11 is selected from C 1-4 Alkyl and C 3-6 Cycloalkyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more hydroxy groups.
In some embodiments, R 1 Selected from isopropyl group,
Figure BDA0003222255240000041
Cyclopentyl, cyclohexyl, tetrahydropyranyl, piperidinyl,
Figure BDA0003222255240000042
In some embodiments, the piperidine ring described above
Figure BDA0003222255240000045
And is ortho to the ring substituent. In some embodiments, the piperidine ring described above
Figure BDA0003222255240000043
And the substituent on the ring is meta. In some embodiments, the piperidine ring described above
Figure BDA0003222255240000044
Para to the ring substituents.
In some embodiments, each R is 2 Each independently selected from H and halogen.
In some embodiments, each R is independently selected from R, and R 2 Each independently selected from H, fluorine and chlorine.
In some embodiments, R 2 Is H.
In some embodiments, R 2 Is fluorine.
In some embodiments, R 3 Is selected from C 6-10 Aryl and 5-6 membered heteroaryl, said aryl and heteroaryl optionally substituted with one or more R 7 Substitution;
R 7 selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR 12 and-NR 12 (R 13 );
R 12 Is selected from C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 13 is selected from H and C 1-6 An alkyl group.
In some embodiments, R 3 Is selected from C 6-10 Aryl and 5-6 membered heteroaryl, optionally substituted with one or more R 7 Substitution;
R 7 selected from halogen, C 1-6 Haloalkyl and-OR 12
R 12 Is selected from C 1-6 An alkyl group.
In some embodiments, R 3 Selected from phenyl and pyridyl optionally substituted by one or more R 7 Substitution;
R 7 selected from fluorine, chlorine, C 1-4 Haloalkyl and-OR 12
R 12 Is selected from C 1-4 An alkyl group.
In some embodiments, R 3 Selected from phenyl and pyridyl, said phenyl and pyridyl being optionally substituted with one or more substituents selected from fluoro, trifluoromethyl and methoxy.
In some embodiments, R 3 Is selected from
Figure BDA0003222255240000051
In some embodiments, X is N.
In some embodiments, X is CH.
In some embodiments of the present invention, the substrate is, L is selected from the group consisting of-C (O) -NH-, -NH-C (O) -, and-C 1-6 alkylene-C (O) -NH-.
In some embodiments, L is-C (O) -NH-.
In some embodiments, -L-R 3 Is composed of
Figure BDA0003222255240000052
In some embodiments of the present invention, the substrate is, L is selected from-C (O) -NH-, -NH-C (O) -, -C 1-4 alkylene-NH-C (O) -and-C 1-4 alkylene-C (O) -NH-.
In some embodiments, L is selected from the group consisting of-C (O) -NH-and-C 1-4 alkylene-NH-C (O) -.
In some embodiments, -L-R 3 Is selected from
Figure BDA0003222255240000053
In some embodiments, the compound has the structure shown in formula II-a:
Figure BDA0003222255240000054
wherein R is 1 、R 2 、R 3 And m is as described in any of the preceding.
In some embodiments, R in formula II-a 1 Is selected from C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 5-6 Cycloalkyl and 5-6 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted with one or more hydroxyl groups.
In some embodiments, R in formula II-a 1 Selected from isopropyl, tetrahydropyranyl,
Figure BDA0003222255240000061
In some embodiments, the compound has a structure represented by formula II-b:
Figure BDA0003222255240000062
wherein R is 1 、R 2 、R 3 And m is as described in any of the preceding.
In some embodiments, R in formula II-b 1 Is selected from C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 5-6 Cycloalkyl and 5-6 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted with one or more R 4 Substitution;
R 4 is selected from C 1-4 Alkyl, hydroxy and-C (O) R 11
R 11 Is selected from C 1-4 An alkyl group optionally substituted with one or more hydroxyl groups; preferably, R 11 Selected from unsubstituted C 1-4 An alkyl group.
In some embodiments, R in formula II-b 1 Selected from isopropyl, tetrahydropyranyl,
Figure BDA0003222255240000063
Figure BDA0003222255240000064
In some embodiments, the piperidine ring described above
Figure BDA0003222255240000065
Para to the ring substituents.
In some embodiments, the compound has a structure represented by formula II-c:
Figure BDA0003222255240000066
wherein R is 1 、R 2 、R 3 And m is as described in any of the preceding.
In some embodiments, R in formula II-c 1 Is selected from C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 5-6 Cycloalkyl and 5-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 is selected from C 1-4 Haloalkyl, hydroxy, -C (O) NR 9 (R 10 ) and-C (O) R 11
R 9 Is selected from C 1-4 An alkyl group;
R 10 is selected from C 1-4 An alkyl group;
R 11 is selected from C 1-4 Alkyl and C 3-6 Cycloalkyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more hydroxyl groups; preferably, R 11 Selected from unsubstituted C 1-4 An alkyl group.
In some embodiments, R in formula II-c 1 Selected from isopropyl, cyclopentyl, tetrahydropyranyl,
Figure BDA0003222255240000071
Figure BDA0003222255240000072
In some embodiments, R in formula II-c 1 Selected from isopropyl, cyclopentyl, tetrahydropyranyl,
Figure BDA0003222255240000073
Figure BDA0003222255240000074
In some embodiments, the piperidine ring described above
Figure BDA0003222255240000075
And is ortho to the ring substituent. In some embodiments, the piperidine ring described above
Figure BDA0003222255240000076
And the substituent on the ring is meta. In some embodiments, the piperidine ring described above
Figure BDA0003222255240000077
Para to the ring substituents.
In the above embodiments of the present invention, the groups of all embodiments can be suitably selected in any combination, thereby obtaining different general formula ranges or specific embodiments. These ranges and schemes are within the scope of the present invention.
In some embodiments, the compound is selected from
Figure BDA0003222255240000078
Figure BDA0003222255240000081
Figure BDA0003222255240000091
In another aspect, the present application provides a pharmaceutical composition comprising a compound of any one of the first aspects, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, and optionally one or more excipients.
The purpose of the pharmaceutical compositions described herein is to facilitate administration to an organism, facilitate absorption of the active ingredient and thus exert biological activity. Wherein the excipient refers to an additive except the main drug in the drug preparation. The composition has stable properties, no incompatibility with main drug, no side effect, no influence on curative effect, no deformation at room temperature, no crack, mildew, moth-eaten feeling, no harm to human body, no physiological effect, no chemical or physical effect with main drug, no influence on content determination of main drug, etc.
The pharmaceutical composition may be administered by the following route: parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular routes, or as inhalants.
The pharmaceutical composition can be prepared into various suitable dosage forms according to the administration route. When administered orally, the pharmaceutical composition may be formulated into any orally acceptable dosage form including, but not limited to, tablets, capsules, aqueous solutions or suspensions. When applied topically to the skin, the pharmaceutical composition may be formulated in a suitable ointment, lotion or cream formulation. The pharmaceutical compositions may also be administered in the form of sterile injectable preparations, including sterile injectable aqueous or oleaginous suspensions or solutions.
The effectiveness of the pharmaceutical composition and whether administration is suitable for treating a disease or medical condition in an individual can be determined by suitable in vitro or in vivo assays. Typically, an effective amount of the pharmaceutical composition sufficient to achieve a prophylactic or therapeutic effect is about 0.001 mg/kg body weight/day to about 10,000mg/kg body weight/day. Suitably, the dose is from about 0.01 mg/kg body weight/day to about 1000mg/kg body weight/day. The dosage range may be about 0.01 to 1000mg/kg of subject body weight per day, every second day, or every third day, more usually 0.1 to 500mg/kg of subject body weight.
In another aspect, the present application provides a use of a compound of any one of the first aspects, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, or a pharmaceutical composition of the foregoing, in the manufacture of an inhibitor of BTK (bruton' S tyrosine kinase) wild type or BTK-C481S mutant.
In another aspect, the present application provides a compound of any one of the first aspects, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, or a pharmaceutical composition of the foregoing, for use in inhibiting BTK (bruton' S tyrosine kinase) wild-type or BTK-C481S mutant.
In another aspect, the present application provides a method of inhibiting BTK (bruton' S tyrosine kinase) wild-type or BTK-C481S mutant, comprising administering to a subject an effective amount of a compound of any one of the first aspects of the present application, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the foregoing.
In another aspect, the present application provides a use of a compound of any one of the first aspects, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, or a pharmaceutical composition of the foregoing, for the manufacture of a medicament for the prevention and/or treatment of a disease mediated by BTK (bruton' S tyrosine kinase) wild-type or BTK-C481S mutant.
In another aspect, the present application provides a compound of any one of the first aspects, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the foregoing, for use in the prevention and/or treatment of a disease mediated by BTK (bruton' S tyrosine kinase) wild-type or BTK-C481S mutant.
In another aspect, the present application provides a method for the prevention and/or treatment of a disease mediated by BTK (bruton' S tyrosine kinase) wild-type or BTK-C481S mutant, comprising administering to a subject in need thereof an effective amount of a compound of any one of the first aspects of the present application, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the foregoing.
In some embodiments, the disease mediated by BTK (bruton' S tyrosine kinase) wild type or BTK-C481S mutant described herein is a tumor or an autoimmune disease.
In another aspect, the present application provides a process for the preparation of a compound of the first aspect selected from the following:
the method comprises the following steps: process for producing Compound II-c
Figure BDA0003222255240000111
Wherein, X 1 Is halogen, such as Cl, br or I, preferably Br; z is a linear or branched member 1 、Z 2 Is a boronic acid or boronic ester group; PG (Picture experts group) 1 Represents a protecting group including, but not limited to, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), acetyl, p-methoxybenzylamine (PMB), 2, 4-Dimethoxybenzylamine (DMB), methyl, benzyl, and the like; r is 1 ' is selected from C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl, 3-12 membered heterocycloalkenyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, alkenyl, cycloalkenyl, heterocycloalkenyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution; r is 1 、R 2 、R 3 、R 4 And m is as defined in any one of the first aspect;
preparation of Compounds 1-3: carrying out coupling reaction and optional hydrogenation reaction on the compound 1-1 to obtain a compound 1-3; preferably, when R is 1 ' is optionally substituted by one or more R 4 Substituted C 2-6 Alkenyl radical, C 3-10 (ii) cycloalkenyl or 3-12 membered heterocycloalkenyl, the hydrogenation reaction is carried out;
preparation of Compounds 1-5: carrying out coupling reaction on the compound 1-3 and the compound 1-4 to obtain a compound 1-5;
preparation of Compounds 1-7: carrying out ring closure reaction on the compounds 1-5 and the compounds 1-6 to obtain compounds 1-7;
preparation of Compounds 1-9: carrying out substitution reaction on the compounds 1-7 and the compounds 1-8 to obtain compounds 1-9;
preparation of Compound II-c: compounds 1-9 are subjected to deprotection reactions to give compounds II-c.
The specific conditions of each reaction described above can be performed with reference to textbooks in the art or related prior arts, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The coupling reaction carried out in the preparation of the compounds 1 to 3 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably bis (triphenylphosphine) palladium dichloride. The base may be an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene, or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is preferably carried out at 80 to 120 ℃ and preferably at 110 ℃. The coupling reaction may be carried out under microwave heating conditions. In some embodiments, the coupling reaction is carried out for 1 to 24 hours, such as 3 hours.
The hydrogenation reaction carried out in the preparation of the compounds 1 to 3 is preferably carried out in the presence of a metal catalyst and an organic solvent. The metal catalyst may be selected from palladium on carbon, palladium hydroxide, platinum dioxide, preferably palladium on carbon. The organic solvent can be selected from methanol, ethanol, isopropanol, tetrahydrofuran, ethyl acetate, acetic acid or mixture thereof, preferably methanol. The hydrogenation reaction is carried out under a suitable protective atmosphere (e.g., hydrogen atmosphere). The hydrogenation reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The hydrogenation reaction is preferably carried out for 2 to 16 hours.
The coupling reaction carried out in the preparation of the compounds 1 to 5 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a copper catalyst, such as copper acetate, copper trifluoromethanesulfonate, copper sulfate, cuprous bromide, preferably copper acetate. The base may be an organic base such as N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably pyridine. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from methanol, ethanol, t-butanol, dichloromethane, 1, 2-dichloroethane, dimethylsulfoxide, N-dimethylformamide, preferably 1, 2-dichloroethane. The coupling reaction is preferably carried out in the presence of a water scavenger, such as anhydrous magnesium sulfate, anhydrous sodium sulfate,
Figure BDA0003222255240000121
Molecular sieves, preferably
Figure BDA0003222255240000122
And (3) a molecular sieve. The coupling reaction is carried out in a suitable gas environment, such as air and oxygen. The coupling reaction is preferably carried out at 25 to 70 ℃, preferably 60 ℃. The coupling reaction is preferably carried out for 6 to 24 hours, for example 7 hours.
The ring-closing reaction carried out in the preparation of the compounds 1 to 7 is preferably carried out in the presence of an organic solvent and a base. The organic solvent may be selected from toluene, 1, 4-dioxane, methanol, ethanol and n-butanol and any combination thereof, preferably n-butanol. The base may be selected from triethylamine and N, N-diisopropylethylamine, preferably N, N-diisopropylethylamine. The ring closure reaction is preferably carried out at 80 to 120 ℃ and preferably at 110 ℃. The ring closure reaction is preferably carried out for 1 to 6 hours, for example 2 hours.
The substitution reaction carried out in the preparation of the compounds 1 to 9 is preferably carried out in the presence of a suitable organic solvent, a base and a condensing agent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide, N-dimethylformamide, and any combination thereof, preferably tetrahydrofuran. The condensing agent can be selected from 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, and a Cartesian condensing agent, preferably a Cartesian condensing agent. The base may be selected from N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably N, N-diisopropylethylamine. The substitution reaction is preferably carried out at 25 to 50 ℃, preferably 40 ℃. The substitution reaction is preferably carried out for 1 to 12 hours, for example 6 hours.
The deprotection reaction carried out in the preparation of compounds II-c is preferably carried out in the presence of a suitable acid. The acid may be selected from hydrochloric acid, trifluoroacetic acid, trifluoromethanesulfonic acid, preferably trifluoroacetic acid. The deprotection reaction is preferably carried out at 60 to 100 ℃, preferably 80 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 4 hours.
The method 2 comprises the following steps: process for producing Compound II-c
Figure BDA0003222255240000131
Wherein, X 1 Is halogen, such as Cl, br or I, preferably Br; z 1 、Z 2 Is a boronic acid or boronic ester group; LG (Ligno-lead-acid) 1 Represents a leaving group including, but not limited to, a halogen atom, trifluoromethanesulfonate, etc.; PG (PG) 1 、PG 2 Represents a protecting group including, but not limited to, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), acetyl, p-methoxybenzylamine (PMB), 2, 4-Dimethoxybenzylamine (DMB), 2- (trimethylsilanyl) ethoxymethyl (SEM), methyl, benzyl, and the like; r is 1 ' is selected from C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl, 3-12 membered heterocycloalkenyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, alkenyl, cycloalkenyl, heterocycloalkenyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution; r 1 、R 2 、R 3 、R 4 And m is as defined in any one of the first aspect;
preparation of Compounds 2-3: carrying out substitution reaction on the compound 2-1 and the compound 2-2 to obtain a compound 2-3;
chemical combinationPreparation of substances 2 to 5: carrying out coupling reaction and optional hydrogenation reaction on the compound 2-3 to obtain a compound 2-5; preferably, when R is 1 ' is optionally substituted by one or more R 4 Substituted C 2-6 Alkenyl radical, C 3-10 (ii) cycloalkenyl or 3-12 membered heterocycloalkenyl, the hydrogenation reaction is carried out;
preparation of Compounds 2-7: performing ring closure reaction on the compound 2-5 and the compound 2-6 to obtain a compound 2-7;
preparation of Compounds 2-9: carrying out substitution reaction on the compound 2-7 and the compound 2-8 to obtain a compound 2-9;
preparation of Compounds 2-10: carrying out deprotection reaction on the compounds 2-9 to obtain compounds 2-10;
preparation of Compound II-c: and carrying out coupling reaction on the compound 2-10 and the compound 2-11 to obtain a compound II-c.
The specific conditions of each reaction described above can be performed with reference to textbooks in the art or related prior arts, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The substitution reaction carried out in the preparation of the compounds 2-3 is preferably carried out in the presence of a suitable organic solvent and a base. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide, N-dimethylformamide, and any combination thereof, preferably N, N-dimethylformamide. The base may be selected from N, N-diisopropylethylamine, triethylamine, cesium carbonate and potassium carbonate, preferably cesium carbonate. The substitution reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The substitution reaction is preferably carried out for 6 to 20 hours, for example 16 hours.
The coupling reaction carried out in the preparation of the compounds 2 to 5 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably bis (triphenylphosphine) palladium dichloride. The base may be an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene, or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is preferably carried out at 80 to 120 ℃ and preferably at 100 ℃. In some embodiments, the coupling reaction is carried out for 1 to 24 hours, such as 16 hours.
The hydrogenation reaction carried out in the preparation of the compounds 2 to 5 is preferably carried out in the presence of a metal catalyst and an organic solvent. The metal catalyst can be selected from palladium carbon, palladium hydroxide and platinum dioxide, and palladium carbon is preferred. The organic solvent can be selected from methanol, ethanol, isopropanol, tetrahydrofuran, ethyl acetate, acetic acid or mixture thereof, preferably methanol. The hydrogenation reaction is carried out under a suitable protective atmosphere (e.g., hydrogen atmosphere). The hydrogenation reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The hydrogenation reaction is preferably carried out for 2 to 16 hours.
The ring-closing reaction carried out in the preparation of compounds 2 to 7 is preferably carried out in the presence of an organic solvent and a base. The organic solvent may be selected from toluene, 1, 4-dioxane, methanol, ethanol and n-butanol and any combination thereof, preferably n-butanol. The base may be selected from triethylamine and N, N-diisopropylethylamine, preferably N, N-diisopropylethylamine. The ring closure reaction is preferably carried out at from 80 to 120 ℃ and preferably at 110 ℃. The ring closure is preferably carried out for 1 to 6 hours, for example 2 hours.
The substitution reaction carried out in the preparation of compounds 2 to 9 is preferably carried out in the presence of a suitable organic solvent, a base and a condensing agent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide, N-dimethylformamide, and any combination thereof, preferably tetrahydrofuran. The condensing agent can be selected from 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, and a Cartesian condensing agent, preferably a Cartesian condensing agent. The base may be selected from N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably N, N-diisopropylethylamine. The substitution reaction is preferably carried out at 25 to 50 ℃, preferably at 25 ℃. The substitution reaction is preferably carried out for 1 to 12 hours, for example 2 hours.
The deprotection reaction carried out in the preparation of compounds 2 to 10 is preferably carried out in the presence of a suitable acid. The acid may be selected from hydrochloric acid, trifluoroacetic acid, trifluoromethanesulfonic acid, preferably trifluoroacetic acid. The reaction is preferably carried out at 60 to 100 ℃, preferably 70 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 5 hours.
The coupling reaction carried out in the preparation of the compounds II-c is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a copper catalyst, such as cupric acetate, copper trifluoromethanesulfonate, cupric sulfate, cuprous bromide, preferably cupric acetate. The base may be an organic base such as N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably pyridine. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from methanol, ethanol, t-butanol, dichloromethane, 1, 2-dichloroethane, dimethyl sulfoxide, N-dimethylformamide, preferably 1, 2-dichloroethane. The coupling reaction is preferably carried out in the presence of a water scavenger, such as anhydrous magnesium sulfate, anhydrous sodium sulfate,
Figure BDA0003222255240000151
Molecular sieves, preferably
Figure BDA0003222255240000152
And (3) a molecular sieve. The coupling reaction is carried out in a suitable gaseous environment, such as air and oxygen. The coupling reaction is preferably carried out at 25 to 70 ℃, preferably 40 ℃. The coupling reaction is preferably carried out for 6 to 24 hours, for example 6 hours.
The method 3 comprises the following steps: process for producing Compound II-a
Figure BDA0003222255240000153
Wherein, X 1 、X 2 Is halogen, such as Cl, br or I, preferably Br; z 1 Is a boronic acid or boronic ester group; PG (PG) 1 、PG 2 Represents a protecting group including, but not limited to, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), acetyl, p-methoxybenzylamine (PMB), 2, 4-Dimethoxybenzylamine (DMB), 2- (trimethylsilanyl) ethoxymethyl (SEM), methyl, benzyl, and the like; r 1 ' is selected from C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl, 3-12 membered heterocycloalkenyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, alkenyl, cycloalkenyl, heterocycloalkenyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution; r 1 、R 2 、R 3 、R 4 And m is as defined in any one of the first aspect;
preparation of Compounds 3-3: performing coupling reaction on the compound 3-1 and the compound 3-2 to obtain a compound 3-3;
preparation of Compounds 3-4: carrying out deprotection reaction on the compound 3-3 to obtain a compound 3-4;
preparation of Compounds 3-6: carrying out substitution reaction on the compound 3-4 and the compound 3-5 to obtain a compound 3-6;
preparation of Compounds 3-7: carrying out deprotection reaction on the compound 3-6 to obtain a compound 3-7;
preparation of Compounds 3-8: halogenating the compound 3-7 to obtain a compound 3-8
Preparation of Compound II-a: carrying out coupling reaction and optional hydrogenation reaction on the compound 3-8 to obtain a compound II-a; preferably, when R is 1 ' is optionally substituted by one or more R 4 Substituted C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl or 3-12 membered heterocycloalkenyl, the hydrogenation is carried out.
The specific conditions of each reaction described above can be performed with reference to textbooks in the field or related prior art, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The coupling reaction carried out in the preparation of the compound 3-3 is preferably carried out in the presence of a metal catalyst, a base and a ligand. The metal catalyst may be a copper catalyst, such as cuprous iodide, cuprous bromide, cuprous oxide, preferably cuprous iodide. The base is selected from potassium phosphate, potassium hydroxide, sodium tert-butoxide, cesium carbonate, preferably potassium phosphate. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from methanol, ethanol, t-butanol, dichloromethane, 1, 2-dichloroethane, dimethylsulfoxide, N-dimethylformamide, preferably N, N-dimethylformamide. The coupling reaction is preferably carried out in the presence of a ligand, such as 1, 10-phenanthroline, L-proline, trans-N, N ' -dimethyl-1, 2-cyclohexanediamine, N ' -dimethylethylenediamine, preferably trans-N, N ' -dimethyl-1, 2-cyclohexanediamine. The coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction may be carried out under microwave heating conditions. The coupling reaction is preferably carried out at 90 to 140 ℃, preferably 125 ℃. The coupling is preferably continued for 1 to 6 hours, for example 2 hours.
The deprotection reaction carried out in the preparation of compounds 3-4 is preferably carried out in the presence of a suitable acid and an organic solvent. The organic solvent can be selected from dichloromethane, chloroform, toluene, preferably dichloromethane. The acid may be selected from hydrochloric acid, boron tribromide, boron trichloride, preferably boron tribromide. The deprotection reaction is preferably carried out at 0-40 ℃, preferably 25 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 3 hours.
The substitution reaction carried out in the preparation of the compounds 3 to 6 is preferably carried out in the presence of a suitable organic solvent, a base and a condensing agent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide, N-dimethylformamide, and any combination thereof, preferably tetrahydrofuran. The condensing agent can be selected from 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, and a Cartesian condensing agent, preferably a Cartesian condensing agent. The base may be selected from N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably N, N-diisopropylethylamine. The substitution reaction is preferably carried out at 25 to 70 ℃, preferably 60 ℃. The substitution reaction is preferably carried out for 1 to 12 hours, for example 6 hours.
The deprotection reaction carried out in the preparation of compounds 3-7 is preferably carried out in the presence of a suitable acid. The acid may be selected from hydrochloric acid, trifluoroacetic acid, trifluoromethanesulfonic acid, preferably trifluoroacetic acid. The deprotection reaction is preferably carried out at 60 to 100 ℃, preferably 80 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 4 hours.
The halogenation reaction carried out in the preparation of compounds 3 to 8 is preferably carried out in the presence of an organic solvent and a halogenating agent. The halogenating agent may be N-bromosuccinimide, bromine, N-chlorosuccinimide, N-iodosuccinimide and elemental iodine, preferably N-bromosuccinimide. The solvent includes N, N-dimethylformamide, N-methylpyrrolidone, methanol, ethanol, tetrahydrofuran, acetonitrile, 1, 4-dioxane, and the like, and a mixed solvent of acetonitrile and tetrahydrofuran is preferred. The halogenation reaction is preferably carried out at a temperature of from-20 ℃ to room temperature, preferably 0 ℃. The halogenation reaction is preferably carried out for 1 to 6 hours, for example 2 hours.
The coupling reaction carried out in the preparation of the compound II-a is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst is a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis-triphenylphosphine palladium dichloride, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II). The base is an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent, which may be 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene, or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is preferably carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is preferably carried out at 80 to 120 ℃, preferably 95 ℃. The coupling reaction is preferably carried out for 1 to 24 hours, for example 6 hours.
The hydrogenation reaction carried out in the preparation of the compound II-a is preferably carried out in the presence of a metal catalyst and an organic solvent. The metal catalyst can be selected from palladium carbon, palladium hydroxide and platinum dioxide, and palladium carbon is preferred. The organic solvent can be selected from methanol, ethanol, isopropanol, tetrahydrofuran, ethyl acetate, acetic acid or mixture thereof, preferably methanol. The hydrogenation reaction is carried out under a suitable protective atmosphere (e.g., hydrogen atmosphere). The hydrogenation reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The hydrogenation reaction is preferably carried out for 2 to 16 hours.
The method 4 comprises the following steps: process for producing compound II-a
Figure BDA0003222255240000171
Wherein, X 1 、X 2 Is halogen, such as Cl, br or I, preferably Br; z is a linear or branched member 1 Is a boronic acid or boronic ester group; LG (Ligno-lead-acid) 1 、LG 2 Represents a leaving group including, but not limited to, a halogen atom, trifluoromethanesulfonate, etc.; PG (PG) 1 、PG 2 Represents a protecting group including, but not limited to, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), acetyl, p-methoxybenzylamine (PMB), 2, 4-Dimethoxybenzylamine (DMB), 2- (trimethylsilanyl) ethoxymethyl (SEM), methyl, benzyl, and the like; r 1 ' selected from C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl, 3-12 membered heterocycloalkenyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, alkenyl, cycloalkenyl, heterocycloalkenyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution; r 1 、R 2 、R 3 、R 4 And m is as defined in any one of the first aspects;
preparation of Compounds 4-3: carrying out substitution reaction on the compound 4-1 and the compound 4-2 to obtain a compound 4-3;
preparation of Compounds 4-5: carrying out substitution reaction on the compound 4-3 and the compound 4-4 to obtain a compound 4-5;
preparation of Compounds 4-7: carrying out coupling reaction and optional hydrogenation reaction on the compound 4-5 to obtain a compound 4-7; preferably, when R is 1 ' is optionally substituted by one or more R 4 Substituted C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl or 3-12 membered heterocycloalkenyl, the hydrogenation reaction is carried out;
preparation of Compounds 4-8: carrying out deprotection reaction on the compound 4-7 to obtain a compound 4-8;
preparation of Compound II-a: and carrying out coupling reaction on the compound 4-8 and the compound 4-9 to obtain a compound II-a.
The specific conditions of each reaction described above can be performed with reference to textbooks in the art or related prior arts, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The substitution reaction carried out in the preparation of the compound 4-3 is preferably carried out in the presence of a suitable organic solvent and a base. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide, N-dimethylformamide, and any combination thereof, preferably N, N-dimethylformamide. The base may be selected from N, N-diisopropylethylamine, triethylamine, sodium hydride, cesium carbonate and potassium carbonate, preferably sodium hydride. The substitution reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The substitution reaction is preferably carried out for 1 to 20 hours, for example 2 hours.
The substitution reaction carried out in the preparation of the compounds 4-5 is preferably carried out in the presence of a suitable organic solvent and a base. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide, N-dimethylformamide, and any combination thereof, preferably N, N-dimethylformamide. The base may be selected from N, N-diisopropylethylamine, triethylamine, pyridine, potassium carbonate, cesium carbonate, preferably potassium carbonate. The substitution reaction is preferably carried out at 80 to 140 ℃, preferably 130 ℃. The substitution reaction is preferably carried out for 1 to 12 hours, for example 2 hours.
The coupling reaction carried out in the preparation of the compounds 4 to 7 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex, bistriphenylphosphine dichloropalladium, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II). The base may be an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is preferably carried out in a suitable organic solvent which may be selected from 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is preferably carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is preferably carried out at 80 to 120 ℃, preferably at 90 ℃. The coupling reaction is preferably carried out for 1 to 24 hours, for example 16 hours.
The hydrogenation reaction carried out in the preparation of the compounds 4 to 7 is preferably carried out in the presence of a metal catalyst and an organic solvent. The metal catalyst can be selected from palladium carbon, palladium hydroxide and platinum dioxide, and palladium carbon is preferred. The organic solvent can be selected from methanol, ethanol, isopropanol, tetrahydrofuran, ethyl acetate, acetic acid or mixture thereof, preferably methanol. The hydrogenation reaction is carried out under a suitable protective atmosphere (e.g., hydrogen atmosphere). The hydrogenation reaction is preferably carried out at from 0 to 40 ℃, preferably at 25 ℃. The hydrogenation reaction is preferably carried out for 2 to 16 hours.
The deprotection reaction carried out in the preparation of compounds 4-8 is preferably carried out in the presence of a suitable acid. The acid may be selected from hydrochloric acid, trifluoroacetic acid, trifluoromethanesulfonic acid, preferably trifluoroacetic acid. The deprotection reaction is preferably carried out at 25 to 100 ℃, preferably 25 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 3 hours.
The coupling reaction carried out in the preparation of the compound II-a is preferably carried out in the presence of a metal catalyst, a base and a ligand. The metal catalyst may be a copper catalyst, such as cuprous iodide, cuprous bromide, cuprous oxide, preferably cuprous iodide. The base is selected from potassium phosphate, potassium hydroxide, sodium tert-butoxide, cesium carbonate, preferably cesium carbonate. The coupling reaction is carried out in a suitable organic solvent which may be selected from methanol, ethanol, tert-butanol, dichloromethane, 1, 2-dichloroethane, dimethyl sulfoxide, N-dimethylformamide, preferably N, N-dimethylformamide. The coupling reaction is preferably carried out in the presence of a ligand, such as 1, 10-phenanthroline, L-proline, trans-N, N ' -dimethyl-1, 2-cyclohexanediamine, N ' -dimethylethylenediamine, preferably trans-N, N ' -dimethyl-1, 2-cyclohexanediamine. The coupling reaction is preferably carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction may be carried out under microwave heating conditions. The coupling reaction is preferably carried out at from 90 to 140 ℃, preferably at 120 ℃. The coupling reaction is preferably carried out for 1 to 6 hours, for example 2 hours.
The method 5 comprises the following steps: process for producing Compound II-b
Figure BDA0003222255240000191
Wherein Z is 1 Is a boronic acid or boronic ester group; r is 1 、R 2 、R 3 And m is as defined in any one of the first aspects;
preparation of Compound II-b: the compound 4-8 and the compound 5-1 are subjected to coupling reaction to obtain a compound II-b.
The specific conditions of each reaction described above can be performed with reference to textbooks in the field or related prior art, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The coupling reaction carried out in the preparation of the compounds II-b is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a copper catalyst, such as cupric acetate, copper trifluoromethanesulfonate, cupric sulfate, cuprous bromide, preferably cupric acetate. The base may be an organic base such as N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably pyridine. The coupling reactionIt should be carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from methanol, ethanol, t-butanol, dichloromethane, 1, 2-dichloroethane, dimethyl sulfoxide, N-dimethylformamide, preferably 1, 2-dichloroethane. The coupling reaction is preferably carried out in the presence of a water scavenger, such as anhydrous magnesium sulfate, anhydrous sodium sulfate,
Figure BDA0003222255240000201
Molecular sieves, preferably
Figure BDA0003222255240000202
And (3) a molecular sieve. The coupling reaction is carried out in a suitable gaseous environment, such as air and oxygen. The coupling reaction is preferably carried out at 25 to 70 ℃, preferably 40 ℃. The coupling reaction is preferably carried out for 6 to 24 hours, for example 5 hours.
The method 6 comprises the following steps: process for producing Compound II-b
Figure BDA0003222255240000203
Wherein X 1 、X 2 Is halogen, such as Cl, br or I, preferably Br; z is a linear or branched member 1 、Z 2 、Z 3 Is a boronic acid, boronic ester or potassium trifluoroborate group; PG (Picture experts group) 1 Represents a protecting group including, but not limited to, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), acetyl, p-methoxybenzylamine (PMB), 2, 4-Dimethoxybenzylamine (DMB), 2- (trimethylsilanyl) ethoxymethyl (SEM), methyl, benzyl, and the like; r 1 ' is selected from C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl, 3-12 membered heterocycloalkenyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, alkenyl, cycloalkenyl, heterocycloalkenyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution; r is 1 、R 2 、R 3 、R 4 And m is as defined in any one of the first aspect;
preparation of Compound 6-3: carrying out coupling reaction on the compound 6-1 and the compound 6-2 to obtain a compound 6-3;
preparation of Compounds 6-5: carrying out coupling reaction on the compound 6-3 and the compound 6-4 to obtain a compound 6-5;
preparation of Compounds 6-6: carrying out deprotection reaction on the compound 6-5 to obtain a compound 6-6;
preparation of Compounds 6-7: the compound 6-6 is halogenated to obtain a compound 6-7
Preparation of Compound II-b: carrying out coupling reaction and optional hydrogenation reaction on the compound 6-7 to obtain a compound II-b; preferably, when R is 1 ' is optionally substituted by one or more R 4 Substituted C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl or 3-12 membered heterocycloalkenyl, the hydrogenation is carried out.
The specific conditions of each reaction described above can be performed with reference to textbooks in the art or related prior arts, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The coupling reaction carried out in the preparation of the compound 6-3 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst is a copper catalyst, such as cupric acetate, copper trifluoromethanesulfonate, cupric sulfate, cuprous bromide, preferably cupric acetate. The base is an organic base such as N, N-diisopropylethylamine, triethylamine, pyridine, 4-dimethylaminopyridine, preferably pyridine. The coupling reaction is carried out in a suitable organic solvent which may be selected from methanol, ethanol, tert-butanol, dichloromethane, 1, 2-dichloroethane, dimethyl sulfoxide, N-dimethylformamide, preferably 1, 2-dichloroethane. The coupling reaction is preferably carried out in the presence of a water scavenger, such as anhydrous magnesium sulfate, anhydrous sodium sulfate,
Figure BDA0003222255240000211
Molecular sieves, preferably
Figure BDA0003222255240000212
And (3) a molecular sieve. The coupling reaction is carried out in a suitable gaseous environment, such as air and oxygen. The coupling reaction is preferably carried out at 25 to 70 ℃, preferably 35 ℃. The coupling reaction is preferably carried out for 6 to 24 hours, for example 16 hours.
The coupling reaction carried out in the preparation of the compound 6-5 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex, bistriphenylphosphine dichloropalladium, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II). The base may be an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is preferably carried out in a suitable organic solvent which may be selected from 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is preferably carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction may be carried out under microwave heating conditions. The coupling reaction is preferably carried out at 80 to 120 ℃, preferably at 90 ℃. The coupling reaction is preferably carried out for 1 to 24 hours, for example 1 hour.
The deprotection reaction carried out in the preparation of compounds 6-6 is preferably carried out in the presence of a suitable acid. The acid may be selected from hydrochloric acid, trifluoroacetic acid, trifluoromethanesulfonic acid, preferably trifluoroacetic acid. The deprotection reaction is preferably carried out at 25 to 100 ℃, preferably 70 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 12 hours.
The halogenation reaction carried out in the preparation of compounds 6-7 is preferably carried out in the presence of an organic solvent and a halogenating agent. The halogenating agent may be N-bromosuccinimide, bromine, N-chlorosuccinimide, N-iodosuccinimide and elemental iodine, preferably N-bromosuccinimide. The solvent includes N, N-dimethylformamide, methanol, ethanol, dichloromethane, tetrahydrofuran, acetonitrile, 1, 4-dioxane, etc., and dichloromethane is preferred. The halogenation reaction is preferably carried out at a temperature of from-20 ℃ to room temperature, preferably room temperature. The halogenation reaction is preferably carried out for 1 to 6 hours, for example 2 hours.
The coupling reaction carried out in the preparation of the compounds II-b is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex, bistriphenylphosphine dichloropalladium, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II). The base may be an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent, which may be 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene, or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction may be carried out under microwave heating conditions. The coupling reaction is preferably carried out at 80 to 120 ℃, preferably 95 ℃. The coupling reaction is preferably carried out for 1 to 24 hours, for example 3 hours.
The hydrogenation reaction carried out in the preparation of the compound II-b is preferably carried out in the presence of a metal catalyst and an organic solvent. The metal catalyst may be selected from palladium on carbon, palladium hydroxide, platinum dioxide, preferably palladium on carbon. The organic solvent can be selected from methanol, ethanol, isopropanol, tetrahydrofuran, ethyl acetate, acetic acid or mixture thereof, preferably methanol. The hydrogenation reaction is carried out under a suitable protective atmosphere (e.g., hydrogen atmosphere). The hydrogenation reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The hydrogenation reaction is preferably carried out for 2 to 16 hours.
The method 7 comprises the following steps: process for producing Compound II-b
Figure BDA0003222255240000221
Wherein, X 1 、X 2 Is halogen, such as Cl, br or I, preferably Br; z is a linear or branched member 1 Is a boronic acid, boronic ester or potassium trifluoroborate group; PG (Picture experts group) 1 Represents a protecting group including, but not limited to, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), acetyl, p-methoxybenzylamine (PMB), 2, 4-Dimethoxybenzylamine (DMB), 2- (trimethylsilanyl) ethoxymethyl (SEM), methyl, benzyl, and the like; r is 1 ' selected from C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 3-10 Cycloalkenyl, 3-12 membered heterocycloalkenyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, alkenyl, cycloalkenyl, heterocycloalkenyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution; r 1 、R 2 、R 3 、R 4 And m is as defined in any one of the first aspects;
preparation of Compound 7-3: carrying out coupling reaction and optional hydrogenation reaction on the compound 7-1 to obtain a compound 7-3;
preparation of Compounds 7-5: performing coupling reaction on the compound 7-3 and the compound 7-4 to obtain a compound 7-5;
preparation of Compound II-b: the compound 7-5 undergoes a deprotection reaction to give the compound II-b.
The specific conditions of each reaction described above can be performed with reference to textbooks in the art or related prior arts, and the present invention is not particularly limited thereto. For reference, the following method can be employed.
The coupling reaction carried out in the preparation of the compound 7-3 is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst may be a palladium metal catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex, bistriphenylphosphine dichloropalladium, chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II), preferably chlorine (2-dicyclohexylphosphino-2 ',4',6' -triisopropyl-1, 1' -biphenyl) [2- (2 ' -amino-1, 1' -biphenyl) ] palladium (II). The base may be an inorganic base such as potassium phosphate, potassium acetate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene, or a mixed solvent of the above organic solvent and water, for example, a mixed solvent of 1, 4-dioxane and water. The coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is preferably carried out at 80 to 120 ℃, preferably 95 ℃. The coupling reaction is preferably carried out for 1 to 24 hours, for example 5 hours.
The hydrogenation reaction carried out in the preparation of the compound 7-3 is preferably carried out in the presence of a metal catalyst and an organic solvent. The metal catalyst may be selected from palladium on carbon, palladium hydroxide, platinum dioxide, preferably palladium on carbon. The organic solvent can be selected from methanol, ethanol, isopropanol, tetrahydrofuran, ethyl acetate, acetic acid or mixture thereof, preferably methanol. The hydrogenation reaction is carried out under a suitable protective atmosphere (e.g., hydrogen atmosphere). The hydrogenation reaction is preferably carried out at 0 to 40 ℃, preferably 25 ℃. The hydrogenation reaction is preferably carried out for 2 to 16 hours.
The coupling reaction carried out in the preparation of the compound 7-5 is preferably carried out in the presence of a metal catalyst, a base and a ligand. The metal catalyst may be a copper catalyst, such as cuprous iodide, cuprous bromide, cuprous oxide, preferably cuprous iodide. The base is selected from potassium phosphate, potassium hydroxide, sodium tert-butoxide, cesium carbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent. In some embodiments, the organic solvent may be selected from methanol, ethanol, t-butanol, dichloromethane, 1, 2-dichloroethane, dimethyl sulfoxide, N-dimethylformamide, preferably N, N-dimethylformamide. The coupling reaction is preferably carried out in the presence of a ligand, such as 1, 10-phenanthroline, L-proline, trans-N, N ' -dimethyl-1, 2-cyclohexanediamine, N ' -dimethylethylenediamine, preferably trans-N, N ' -dimethyl-1, 2-cyclohexanediamine. The coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction may be carried out under microwave heating conditions. The coupling reaction is effectively carried out at 90-140 c, preferably 120 c. The coupling reaction is preferably carried out for 1 to 6 hours, for example 3 hours.
The deprotection reaction carried out in the preparation of compound II-b is preferably carried out in the presence of a suitable acid. The acid may be selected from hydrochloric acid, trifluoroacetic acid, trifluoromethanesulfonic acid, preferably trifluoroacetic acid. The deprotection reaction is preferably carried out at 25 to 100 ℃, preferably 80 ℃. The deprotection reaction is preferably carried out for 2 to 12 hours, for example 4 hours.
Definition of
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to those techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to those skilled in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
As used herein, the term "alkylene" denotes a saturated divalent hydrocarbon group, preferably having 1,2,3,4, 5 or 6 carbon atoms, such as "C" as used herein 1-6 Alkylene group "," C 2-6 Alkylene group "," C 1-4 Alkylene "and the like, and specific examples include, but are not limited to, methylene, ethylene, propylene, and butylene.
The term "alkyl" as used herein is defined as a linear or branched saturated aliphatic hydrocarbon, preferably an alkyl group having 1,2,3,4, 5 or 6 carbon atoms, such as "C" as used herein 1-6 Alkyl group "," C 2-6 Alkyl group "," C 1-4 Alkyl group and C 1-2 Alkyl "and the like, including but not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, and n-hexyl. When the alkyl group is substituted with 1 or more (such as 1 to 3) halogens, it is said to be "haloalkyl" herein, e.g. "halo C 1-6 Alkyl group "," halogeno C 1-4 Alkyl group "," halogeno C 1-2 Alkyl "etc", specific examples include but are not limited to CH 2 F、CHF 2 、CF 3 、CCl 3 、C 2 F 5 、C 2 Cl 5 、CH 2 CF 3 、CH 2 Cl or-CH 2 CH 2 CF 3 And so on.
The term "alkenyl" as used herein means a linear or branched monovalent hydrocarbon group comprising one double bond, preferably an alkenyl group having 2,3,4, 5 or 6 carbon atoms, such as "C" as used herein 2-6 Alkenyl group "," C 2-4 Specific examples of alkenyl groups include, but are not limited to, ethenyl, 1-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 2-methyl-2-propenyl, and 4-methyl-3-pentenyl.
As used herein, the term "alkenylene" means a straight or branched divalent carbon chain having one or more carbon-carbon double bonds, preferably an alkenylene group having 2,3,4, 5 or 6 carbon atoms, such as "C" as used herein 2-6 Alkenylene group and C 2-4 And alkenylene "and specific examples include, but are not limited to, vinylene, propenylene, and the like. When the compounds of the invention contain alkenylene groups, the compounds may be present in pure E (entgegen) form, in pure Z (ipsilateral (zusammen)) form or in any mixture thereof.
As used herein, the term "cycloalkyl" refers to a saturated monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, or bicyclic, including spiro, fused or bridged systems (such as bicyclo [ 1.1.1)]Pentyl, bicyclo [2.2.1]Heptyl, bicyclo [3.2.1]Octyl or bicyclo [5.2.0]Nonyl, decalinyl, etc.). Cycloalkyl groups having 3 to 10 carbon atoms are preferred. For example, "C 3-10 Cycloalkyl group "," C 3-8 Cycloalkyl group "," C 3-6 Cycloalkyl ", specific examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
As used herein, the term "cycloalkeneThe group "refers to a monocyclic or polycyclic (such as bicyclic) hydrocarbon ring containing one or more carbon-carbon double bonds. Cycloalkenyl groups having 3 to 10 carbon atoms are preferred. For example, "C 3-10 Cycloalkenyl group "," C 3-8 Cycloalkenyl group "," C 3-6 Cycloalkenyl groups ". Specific examples include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, or cycloheptenyl.
As used herein, the term "heterocyclyl" refers to a saturated (i.e., heterocycloalkyl) or partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) monocyclic or polycyclic cyclic hydrocarbon group containing at least one heteroatom selected from N, O, and S, such as "3-12 membered heterocyclyl", "3-10 membered heterocyclyl", "3-6 membered monoheterocyclyl", "7-10 membered biheterocyclyl", and the like, as used herein. Specific examples include, but are not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, dioxolyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, 2-aza-spiro [3.5] non-7-yl, and 8-oxabicyclo [3.2.1] oct-3-yl.
As used herein, the term "heterocycloalkenyl" refers to a monocyclic or polycyclic cyclic hydrocarbon group containing at least one heteroatom selected from N, O, and S, and one or more double bonds, such as "3-12 membered heterocycloalkenyl", "3-10 membered heterocycloalkenyl", "3-6 membered mono-heterocycloalkenyl", "7-10 membered bis-heterocycloalkenyl", and the like, as used herein. Specific examples include, but are not limited to, 1,2,3,4-tetrahydropyridinyl, 1,2,3,6-tetrahydropyridinyl, 3, 6-dihydro-2H-pyranyl, and 1, 4-dioxa-spiro [4.5] dec-7-enyl.
As used herein, the term "aryl" refers to a group formed by an all-carbon monocyclic or fused polycyclic aromatic ring having a conjugated pi-electron system with one hydrogen atom removed. For example, "C" as used herein 6-10 Aryl ", specific examples include, but are not limited to, phenyl or naphthyl.
As used herein, the term "heteroaryl" refers to a group formed by a monocyclic or fused polycyclic aromatic ring containing at least one heteroatom selected from N, O and S, with the loss of one hydrogen atom. For example, "5-to 10-membered heteroaryl", "5-to 6-membered heteroaryl", and the like, as used herein, specific examples include, but are not limited to, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like.
As used herein, the term "halogen" includes F, cl, br or I.
The term "substituted" means that one or more (e.g., one, two, three, or four) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the current circumstances is not exceeded and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally substituted with \8230;" substituted ", the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of the list of substituents, one or more hydrogens on the carbon (to the extent of any hydrogens present) may be replaced individually and/or together with an independently selected optional substituent. If a nitrogen of a substituent is described as being optionally substituted with one or more of a list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogens present) may each be replaced with an independently selected optional substituent.
If a substituent is described as "independently selected from" a group of groups, each substituent is selected independently of the other. Thus, each substituent may be the same or different from another (other) substituent.
As used herein, the term "one or more" means 1 or more than 1, such as 2,3,4, 5 or 10, under reasonable conditions.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
When a bond of a substituent is shown through a bond connecting two atoms in a ring, then such substituent may be bonded to any ring atom in the substitutable ring.
The invention also includes all pharmaceutically acceptable isotopically-labeled compounds, which are identical to those of the present invention, except that one or more atoms are replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number prevailing in nature. Of isotopes suitable for inclusion in the compounds of the invention examples include, but are not limited to, isotopes of hydrogen (e.g., deuterium (g) (ii)), (iii) 2 H) Tritium (a) 3 H) ); isotopes of carbon (e.g. of 11 C、 13 C and 14 c) (ii) a Isotopes of chlorine (e.g. of chlorine) 36 Cl); isotopes of fluorine (e.g. of fluorine) 18 F) (ii) a Isotopes of iodine (e.g. of iodine) 123 I and 125 i) (ii) a Isotopes of nitrogen (e.g. of 13 N and 15 n); isotopes of oxygen (e.g. of 15 O、 17 O and 18 o); isotopes of phosphorus (e.g. of phosphorus) 32 P); and isotopes of sulfur (e.g. of sulfur) 35 S)。
As used herein, the term "stereoisomer" refers to an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., one, two, three, or four) asymmetric centers, they can give rise to racemic mixtures, single enantiomers, diastereomeric mixtures, and individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as mixtures of two or more structurally different forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. It is understood that the scope of this application encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
In this contextCan use the solid line
Figure BDA0003222255240000261
Solid wedge shape
Figure BDA0003222255240000262
Or virtual wedge shape
Figure BDA0003222255240000263
Carbon-carbon bonds of the compounds of the invention are depicted. The use of a solid line to depict bonds to asymmetric carbon atoms is intended to indicate that all possible stereoisomers (e.g., particular enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of solid or dashed wedges to depict bonds to asymmetric carbon atoms is intended to indicate that the stereoisomers shown are present. When present in a racemic mixture, solid and dotted wedges are used to define the relative stereochemistry, not the absolute stereochemistry. Unless otherwise indicated, the compounds of the present invention are intended to exist in the form of stereoisomers, including cis and trans isomers, optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof. The compounds of the invention may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio.
It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, where appropriate, in the form of a pharmaceutically acceptable derivative thereof. In the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, solvates, N-oxides, metabolites or prodrugs, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof.
For a review of suitable Salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: properties, selection, and Use" (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise a polar solvent, such as in particular water, methanol or ethanol, as structural element of the crystal lattice of the compound. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
Those skilled in the art will appreciate that not all nitrogen-containing heterocycles are capable of forming N-oxides, since the available lone pair is required for oxidation of the nitrogen to the oxide; one skilled in the art will recognize nitrogen-containing heterocycles that are capable of forming N-oxides. Those skilled in the art will also recognize that tertiary amines are capable of forming N-oxides. Synthetic methods for preparing N-oxides of heterocycles and tertiary amines are well known to those skilled in the art and include oxidation of heterocycles and tertiary amines with peroxy acids such as peracetic and m-chloroperbenzoic acid (MCPBA), hydrogen peroxide, alkyl hydroperoxides such as t-butyl hydroperoxide, sodium perborate, and dioxiranes (dioxiranes) such as dimethyldioxirane. These methods for preparing N-oxides have been widely described and reviewed in the literature, see for example: T.L.Gilchrist, comprehensive Organic Synthesis, vol.7, pp748-750; a.r.katitzky and a.j.boulton, eds., academic Press; and g.w.h.cheeseman and e.s.g.werstink, advances in Heterocyclic Chemistry, vol.22, pp 390-392, a.r.kattritzky and a.j.boulton, eds., academic Press.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., substances formed in vivo upon administration of the compounds of the present invention. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds made by the process of contacting the compounds of the present invention with a mammal for a time sufficient to produce a metabolite thereof.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Additional information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", volume 14, ACS Symposium Series (t.higuchi and v.stella). Prodrugs of the invention may be prepared, for example, by replacing appropriate functional groups present in a compound of the invention with certain moieties known to those skilled in the art as "pro-moieties", for example as described in "Design of Prodrugs", h.
As used herein, the term "treating" means reversing, alleviating the progression of, or one or more symptoms of, the disorder or condition to which such term applies.
As used herein, the term "prevention" refers to inhibiting and delaying the onset of a disease, and includes not only prevention prior to the development of the disease, but also prevention of recurrence of the disease after treatment.
As used herein, the term "effective amount" (e.g., "therapeutically effective amount" or "prophylactically effective amount") refers to an amount of active ingredient that will achieve the desired effect to some extent upon administration, e.g., to alleviate one or more symptoms of the treated condition or to prevent the occurrence of the condition or symptoms thereof.
As used herein, "individual", "subject" includes human or non-human animals. Exemplary human individuals or subjects include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
Advantageous effects of the invention
The invention provides a compound with a novel structure and an inhibiting effect on BTK, in particular to a BTK-C481S mutant, a pharmaceutical composition thereof, a preparation method thereof and application thereof in preventing and/or treating diseases mediated by BTK wild type or C481S mutation, such as tumors or autoimmune diseases. In vitro test results show that the compound has stronger inhibition effect on BTK-C481S, and part of the compounds inhibit IC (integrated Circuit) of BTK-C481S 50 In that<1000nM, preferably<500nM, more preferably<100nM, more preferably<50nM, more preferably<25nM. In some embodiments, compounds of the invention inhibit IC in vitro on human lymphoma REC-1 cells 50 In that<100 μ M, more preferably<10 μ M, more preferably<1 μ M, more preferably<500nM, more preferably<100nM, more preferably<50nM, more preferably<25nM。
Detailed Description
Embodiments of the present invention will be described in detail below with reference to examples, but those skilled in the art will appreciate that the following examples are only illustrative of the present invention and should not be construed as limiting the scope of the present invention. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
In this application, when chemical names and structural formulae are inconsistent, the structural formulae should be taken as a control unless the context suggests that the chemical name and not the structural formula is correct.
Abbreviations in the context of the present invention have the following meanings:
Figure BDA0003222255240000281
Figure BDA0003222255240000291
when the chemical names and structural formulae of the compounds in the following examples are inconsistent, the structural formulae should be taken as the basis unless the context suggests that the chemical names are correct. The structural formulae of the compounds described in the following examples are given by 1 H-NMR or MS. 1 The H-NMR analyzer is Bruker 400MHz NMR spectrometer, and the solvent is CD 3 OD、CDCl 3 Or DMSO-d 6 The internal standard substance is TMS, and all delta values are expressed by ppm values. The MS measurement instrument is an Agilent 6120B mass spectrometer, and the ion source is ESI.
The reaction process is monitored by TLC or LC-MS, a developing agent system comprises a dichloromethane and methanol system, a normal hexane and ethyl acetate system and a petroleum ether and ethyl acetate system, and the volume ratio of solvents can be adjusted according to different polarities of compounds. To obtain an appropriate value of the specific shift (Rf) or Retention Time (RT), an appropriate amount of triethylamine or the like may be added to the developing solvent. TLC was performed using an aluminum plate (20X 20 cm) manufactured by Merck, and GF254 silica gel (0.4 to 0.5 mm) for thin layer chromatography manufactured by Qingdao ocean chemical industry.
The separation and purification of the reaction product are carried out by CC or PHPLC. The CC uses 200-300 mesh silica gel as a carrier. The system of eluents comprises: dichloromethane and methanol system, petroleum ether and ethyl acetate system, the volume ratio of solvent is regulated according to different polarities of the compound, and small amount of triethylamine can be added for regulation. PHPLC uses two conditions: the preparation method 1 comprises the following steps: the instrument model is as follows: agilent 1260, column: waters Xbridge Prep C 18 OBD (19 mm. Times.150 mm. Times.5.0. Mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0mL/min; detection wavelength: 214nm; a mobile phase A:100% acetonitrile; and (3) mobile phase B:0.05% aqueous ammonium bicarbonate; elution gradient: 0min:10% A,90% B;16.0min:90% A,10% B; the preparation method 2 comprises the following steps: the instrument model is as follows: agilent 1260, column: waters SunAire Prep C 18 OBD (19 mm. Times.150 mm. Times.5.0. Mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0mL/min; detection wavelength: 214nm; a mobile phase A:100% acetonitrile; and (3) mobile phase B:100% water (containing 0.05% formic acid); elution gradient: 0min:10% A,90% B;16.0min:90% A,10% B.
Unless otherwise specified, the reaction temperatures of the examples are room temperature (20 ℃ C. To 30 ℃ C.).
The reagents used in the present invention are commercially available from Acros Organics, aldrich Chemical Company, shanghai Teber Chemical science and technology, inc., and the like.
Preparation of an intermediate:
intermediate preparation example 1: preparation of (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid
Figure BDA0003222255240000301
The first step is as follows: preparation of 4-bromo-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (trifluoromethyl) pyridin-2-amine (10g, 61.69mmol) and N, N-dimethylaniline (15.10g, 123.37mmol) were dissolved in anhydrous tetrahydrofuran (150 mL), a tetrahydrofuran solution of 4-bromobenzoyl chloride (13.54g, 61.69mmol) was slowly dropped at 0 ℃ and, after the dropping, the reaction was continued at 0 ℃ for half an hour and at room temperature for 2 hours. The reaction solution was poured into water, extracted three times with ethyl acetate, the organic phases were combined and washed three times with saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =6/1 (v/v)) to obtain the title compound of this step (16.55 g, yield: 74%).
MS m/z(ESI):345.0[M+H] +
The second step is that: preparation of 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4-bromo-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (12.55g, 34.55mmol), pinacol diborate (10.63g, 41.46mmol) and potassium acetate (6.92g, 69.09mmol) were added to 1, 4-dioxane (125 mL), replaced with nitrogen three times, bis (triphenylphosphine) palladium dichloride (484.96mg, 0.69mmol) was added, and the reaction was carried out at 90 ℃ for 5 hours. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =5/1 (v/v)) to obtain the title compound of this step (15.3 g, yield: 99%).
MS m/z(ESI):393.2[M+H] +
The third step: preparation of (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid
4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (15.3g, 34.33mmol) was added to tetrahydrofuran (75 mL) and water (15 mL), sodium periodate (22.03g, 102.99mmol) was further added, and after stirring for half an hour, dilute hydrochloric acid (3M, 12.02mL) was added and reacted at room temperature for 4 hours. The reaction solution was poured into an appropriate amount of water, extracted with ethyl acetate, the organic phase was washed with water, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was slurried with methylene chloride and filtered to obtain the title compound of this step (9.5 g, yield: 89%).
MS m/z(ESI):311.1[M+H] +
Intermediate preparation example 2: preparation of 4- (4-amino-7-bromo-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Figure BDA0003222255240000311
The first step is as follows: preparation of 4- (4-methoxy-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4-bromo-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (1g, 2.75mmol) and 4-methoxy-5H-pyrrolo [3,2-d ] pyrimidine (360.15mg, 2.29mmol) were dissolved in N, N-dimethylformamide (6 mL), potassium phosphate (983.65mg, 4.59mmol) was added, after nitrogen substitution, cuprous iodide (441.29mg, 2.29mmol) and trans-N, N' -dimethyl-1, 2-cyclohexanediamine (329.58mg, 2.29mmol) were added, and the mixture was heated by microwave at 125 ℃ for 2 hours. After the completion of the reaction, the crude product obtained by concentrating the reaction liquid was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =2/1 (v/v)) to obtain the title compound of the present step (300 mg, yield: 30%).
MS m/z(ESI):414.1[M+H] +
The second step is that: preparation of 4- (4-hydroxy-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-methoxy-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (600mg, 1.38mmol) was dissolved in dichloromethane (8 mL) and boron tribromide (1.04g, 4.14mmol) was slowly added dropwise at 0 ℃ and, after completion of the dropwise addition, reacted at room temperature for 3 hours. After completion of the reaction, methanol was added dropwise in an ice bath until no gas was generated, the pH was adjusted to weak alkalinity with a saturated sodium bicarbonate solution, extraction was performed with dichloromethane/methanol =10/1 (v/v), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to obtain the title compound of the present step (500 mg, yield: 86%).
MS m/z(ESI):400.1[M+H] +
The third step: preparation of 4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-hydroxy-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (600mg, 1.43mmol), 1, 8-diazabicyclo [5.4.0] undec-7-ene (726.28mg, 2.85mmol), and the carbocondensing agent (1.23g, 5.71mmol) were added to tetrahydrofuran (10 mL), stirred at room temperature for 15 minutes, and 2, 4-dimethoxybenzylamine (1.51g, 8.56mmol) was added dropwise, and reacted at 60 ℃ for 6 hours. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: dichloromethane/methanol =50/1 (v/v)) to obtain the title compound of this step (750 mg, yield: 86%).
MS m/z(ESI):549.2[M+H] +
The fourth step: preparation of 4- (4-amino-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (800mg, 1.17mmol) was dissolved in trifluoroacetic acid (8 mL) and reacted at 80 ℃ for 4 hours. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: dichloromethane/methanol =20/1 (v/v)) to obtain the title compound of the present step (500 mg, yield: 975%).
MS m/z(ESI):399.1[M+H] +
The fifth step: preparation of 4- (4-amino-7-bromo-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (450mg, 1.07mmol) was dissolved in acetonitrile (5 mL), and a solution of N-bromosuccinimide (212.23mg, 1.18mmol) in tetrahydrofuran (6 mL) was added dropwise at 0 ℃ and then replaced with nitrogen three times, followed by reaction at 0 ℃ for 2 hours. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: dichloromethane/methanol =8/1 (v/v)) to obtain the title compound of the present step (475 mg, yield: 93%).
MS m/z(ESI):477.0[M+H] +
Intermediate preparation example 3: preparation of 7- (1, 1-trifluoropropan-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
Figure BDA0003222255240000321
The first step is as follows: preparation of 7-bromo-4-chloro-5- ((2- (trimethylsilyl) ethoxy) methyl) -5H-pyrrolo [3,2-d ] pyrimidine
7-bromo-4-chloro-5H-pyrrolo [3,2-d ] pyrimidine (500mg, 2.04mmol) was dissolved in N, N-dimethylformamide (4 mL), and sodium hydride (98.08mg, 2.45mmol) and 2- (trimethylsilyl) ethoxymethyl chloride (502.03mg, 2.86mmol) were added and reacted at room temperature for 2 hours. After completion of the reaction, water and ethyl acetate were added for extraction, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound of this step (700 mg, yield: 90%).
MS m/z(ESI):362.0[M+H] +
The second step is that: preparation of 7-bromo-N- (2, 4-dimethoxybenzyl) -5- ((2- (trimethylsilyl) ethoxy) methyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
7-bromo-4-chloro-5- ((2- (trimethylsilyl) ethoxy) methyl) -5H-pyrrolo [3,2-d ] pyrimidine (740mg, 1.94mmol), 2, 4-dimethoxybenzylamine (409.34mg, 2.33mmol) and potassium carbonate (810.48mg, 5.81mmol) were added to N, N-dimethylformamide (4 mL) and reacted at 130 ℃ for 2 hours. Water and ethyl acetate were added for extraction, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound of this step (880 mg, yield: 87%).
MS m/z(ESI):493.1[M+H] +
The third step: preparation of N- (2, 4-dimethoxybenzyl) -7- (3, 3-trifluoroprop-1-en-2-yl) -5- ((2- (trimethylsilyl) ethoxy) methyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
7-bromo-N- (2, 4-dimethoxybenzyl) -5- ((2- (trimethylsilyl) ethoxy) methyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (550mg, 1.03mmol), 1- (trifluoromethyl) hexyleneboronic acid ester (335.48mg, 1.44mmol) and potassium carbonate (446.86mg, 3.08mmol) were added to 1, 4-dioxane (8 mL) and water (2 mL), nitrogen was replaced, chlorine (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (84.82mg, 0.10 mmol) was added, and the reaction was carried out at 90 ℃ for 16 hours. Extraction with water and ethyl acetate, organic phase combination and concentration of the resulting crude product were purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =2/1 (v/v)) to give the title compound of this step (250 mg, yield: 46%).
MS m/z(ESI):509.2[M+H] +
The fourth step: preparation of 7- (3, 3-trifluoroprop-1-en-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
N- (2, 4-Dimethoxybenzyl) -7- (3, 3-trifluoroprop-1-en-2-yl) -5- ((2- (trimethylsilyl) ethoxy) methyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (210mg, 0.39mmol) was dissolved in trifluoroacetic acid (5 mL) and reacted at room temperature for 3 hours. After completion of the reaction, concentrated sodium hydroxide solution was added to adjust the pH to a weakly basic state, extraction was performed with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to obtain the title compound of this step (85 mg, yield: 90%).
MS m/z(ESI):229.1[M+H] +
The fifth step: preparation of 7- (1, 1-trifluoropropan-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
7- (3, 3-trifluoroprop-1-en-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (89.47mg, 0.37mmol) was dissolved in tetrahydrofuran (3 mL) and methanol (3 mL), palladium on carbon (90mg, 10%) was added, hydrogen gas was substituted, and the reaction was carried out at room temperature for 4 hours. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound of this step (80 mg, yield: 89%).
MS m/z(ESI):231.1[M+H] +
Intermediate preparation example 4: preparation of 4- (7-amino-3- (piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Figure BDA0003222255240000341
The first step is as follows: preparation of 5- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) -3, 4-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester
Methyl 4-amino-3-bromo-1H-pyrazole-5-carboxylate (1.05g, 4.54mmol), N-tert-butoxycarbonyl-3, 4-dihydropyridine-5-boronic acid pinacol ester (1.42g, 4.54mmol), and potassium carbonate (1.90g, 13.63mmol) were added to a mixed solvent of 1, 4-dioxane (4 mL) and water (1 mL), and subjected to nitrogen substitution, bis-triphenylphosphine palladium dichloride (325mg, 0.45mmol) was added, and the reaction was carried out at 99 ℃ for 16 hours. The reaction solution was concentrated, diluted with ethyl acetate, washed with saturated brine, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/1 (v/v)) to obtain the title compound of this step (800 mg, yield: 52%).
MS m/z(ESI):323.2[M+H] +
The second step is that: preparation of 3- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester
Tert-butyl 5- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) -3, 4-dihydropyridine-1 (2H) -carboxylate (684mg, 2.02mmol) was dissolved in methanol (20 mL), palladium on carbon (200mg, 10%) was added, hydrogen gas was substituted, and the mixture was stirred at room temperature for 16 hours. The reaction mixture was filtered through Celite, and the filtrate was concentrated to give the title compound of this step (650 mg, yield: 94%).
MS m/z(ESI):325.2[M+H] +
The third step: preparation of 3- (4-amino-5- (methoxycarbonyl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester
(4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid (745mg, 2.28mmol), 3- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester (650mg, 1.90mmol), copper acetate (768mg, 3.81mmol) and pyridine (304mg, 3.81mmol) were added to 1, 2-dichloroethane (20 mL), and
Figure BDA0003222255240000351
molecular sieves (650 mg), oxygen substitution, and reaction at 40 ℃ for 16 hours. The reaction solution was filtered through celite, the filter cake was rinsed 2 to 3 times with dichloromethane, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/1 (v/v)) to obtain the title compound of this step (800 mg, yield: 68%).
MS m/z(ESI):589.2[M+H] +
The fourth step: preparation of tert-butyl 3- (7-hydroxy-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
Tert-butyl 3- (4-amino-5- (methoxycarbonyl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate (630mg, 1.02mmol), N-diisopropylethylamine (1 mL), and formamidine acetate (160mg, 1.53mmol) were added to N-butanol (1 mL), and reacted at 110 ℃ for 5 hours. The reaction solution was concentrated, diluted with ethyl acetate, washed with saturated brine, and the organic phase was dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/1 (v/v)) to obtain the title compound of this step (500 mg, yield: 80%).
MS m/z(ESI):584.2[M+H] +
The fifth step: preparation of tert-butyl 3- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
Tert-butyl 3- (7-hydroxy-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate (700mg, 1.14mmol), 2, 4-dimethoxybenzylamine (385mg, 2.28mmol) and a catide (305mg, 1.48mmol) were dissolved in acetonitrile (20 mL), and 1, 8-diazabicyclo [5.4.0] undec-7-ene (435mg, 1.71mmol) was added and reacted at room temperature for 16 hours. The reaction solution was concentrated, diluted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate for organic phase, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/1 (v/v)) to obtain the title compound of this step (700 mg, yield: 75%).
MS m/z(ESI):733.3[M+H] +
And a sixth step: preparation of 4- (7-amino-3- (piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Tert-butyl 3- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate (293mg, 0.40mmol) was dissolved in trifluoroacetic acid (5 mL) and reacted at 70 ℃ for 16 hours. The reaction solution was concentrated, saturated aqueous sodium hydrogencarbonate solution was added to adjust the pH to slightly alkaline, and extraction was performed with methylene chloride, and the organic phases were combined, dried over anhydrous sodium sulfate, and filtered, and the filtrate was concentrated to obtain the title compound of this step (193 mg, yield: 99%).
MS m/z(ESI):483.2[M+H] +
Intermediate preparation example 5: preparation of 4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Figure BDA0003222255240000361
The first step is as follows: preparation of 4- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester
The title compound of this step (1.47 g, yield: 32%) was synthesized in a similar manner to the procedure described in the first step of intermediate preparation example 4, substituting N-Boc-1,2,5, 6-tetrahydropyridine-4-boronic acid pinacol ester for N-t-butoxycarbonyl-3, 4-dihydropyridine-5-boronic acid pinacol ester of the first step of intermediate preparation example 4.
MS m/z(ESI):323.2[M+H] +
The second step is that: preparation of 4- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester
The title compound of this step (1.30 g, yield: 93%; 93%) was synthesized in a similar manner to the method described in the second step of intermediate preparation example 4 by substituting 4- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester for 5- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) -3, 4-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester in the second step of intermediate preparation example 4
MS m/z(ESI):325.2[M+H] +
The third step: preparation of tert-butyl 4- (4-amino-5- (methoxycarbonyl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate
The title compound (717 mg, yield: 99%) of this step was synthesized in analogy to the procedure described in the third step of intermediate preparation 4, by substituting 4- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester in the third step of intermediate preparation 4 for 3- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester in intermediate preparation 4.
MS m/z(ESI):589.2[M+H] +
The fourth step: preparation of tert-butyl 4- (7-hydroxy-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
The title compound of this step was synthesized (337 mg, yield: 58%) by the method described in the fourth step of intermediate preparation example 4 by substituting 4- (4-amino-5- (methoxycarbonyl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester for 3- (4-amino-5- (methoxycarbonyl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylic acid tert-butyl ester in the fourth step of intermediate preparation example 4.
MS m/z(ESI):584.2[M+H] +
The fifth step: preparation of tert-butyl 4- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
The title compound (340 mg, yield: 60%) of this step was synthesized in analogy to the procedure described in the fifth step of intermediate preparation example 4, with 4- (7-hydroxy-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylic acid tert-butyl ester in place of 3- (7-hydroxy-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylic acid tert-butyl ester in the fifth step of intermediate preparation example 4.
MS m/z(ESI):733.3[M+H] +
And a sixth step: preparation of 4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of this step was synthesized in a similar manner to the procedure described in the sixth step of intermediate preparation 4, using 4- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylic acid tert-butyl ester in place of 3- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylic acid tert-butyl ester in the sixth step of intermediate preparation 4 (73 mg, yield: 99%).
MS m/z(ESI):483.2[M+H] +
Intermediate preparation example 6: preparation of 7-bromo-N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
Figure BDA0003222255240000371
The first step is as follows: preparation of N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
2, 4-Dimethoxybenzylamine (5.50g, 32.56mmol), 4-chloro-5H-pyrrolo [3,2-d ] pyrimidine (5.05g, 32.56mmol) and N, N-diisopropylethylamine (12.73g, 97.68mmol) were dissolved in N-butanol (50 mL) and reacted at 110 ℃ for 12 hours. The crude product obtained by concentrating the reaction solution was slurried with ethyl acetate to obtain the title compound of this step (6 g, yield: 62%).
MS m/z(ESI):285.1[M+H] +
The second step: preparation of 7-bromo-N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
N- (2, 4-Dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (5g, 16.71mmol) was dissolved in methylene chloride (20 mL), and N-bromosuccinimide (3.00g, 16.71mmol) was added and reacted at room temperature for 2 hours. The reaction solution was poured into water, filtered, and the filter cake was dried to obtain the title compound of this step (4 g, yield: 62%).
MS m/z(ESI):363.0[M+H] +
Intermediate preparation example 7: preparation of 3- (1, 1-trifluoropropan-2-yl) -1H-pyrazolo [4,3-d ] pyrimidin-7-amine
Figure BDA0003222255240000381
The first step is as follows: preparation of 4-amino-3-bromo-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole-5-carboxylic acid methyl ester
4-amino-3-bromo-1H-pyrazole-5-carboxylic acid methyl ester (3.06g, 13.63mmol) and cesium carbonate (5.38g, 16.36mmol) were added to N, N-dimethylformamide (30 mL), and 2- (trimethylsilyl) ethoxymethyl chloride (3.44g, 20.45mmol) was added under ice-bath and reacted at room temperature for 16 hours. The reaction mixture was extracted with brine and ethyl acetate, the organic phase was washed with water, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give a crude product, which was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether =1/5 (v/v)) to give the title compound of this step (3.6 g, yield: 75%).
MS m/z(ESI):350.0[M+H] +
The second step: preparation of methyl 4-amino-3- (3, 3-trifluoroprop-1-en-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole-5-carboxylate
Methyl 4-amino-3-bromo-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole-5-carboxylate (8g, 21.70mmol), 1- (trifluoromethyl) ethylboronic acid hexanol ester (5.07g, 21.70mmol), and potassium carbonate (9.09g, 65.09mmol) were added to 1, 4-dioxane (80 mL) and water (16 mL), displaced with nitrogen three times, bis triphenylphosphine palladium dichloride (1.54g, 2.17mmol) was added, and reacted at 100 ℃ for 16 hours. The reaction solution was concentrated, an appropriate amount of water was added, extraction was performed four times with ethyl acetate, the organic layers were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =2/1 (v/v)) to obtain the title compound of this step (4.6 g, yield: 55%).
MS m/z(ESI):366.1[M+H] +
The third step: preparation of methyl 4-amino-3- (1, 1-trifluoropropan-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole-5-carboxylate
Methyl 4-amino-3- (3, 3-trifluoroprop-1-en-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole-5-carboxylate (3.1lg, 8.21mmol) was dissolved in methanol (45 mL), palladium on carbon (900mg, 10%) was added, hydrogen gas was substituted, and the reaction was carried out at room temperature for 16 hours. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound of this step (2.9 g, yield: 91%).
MS m/z(ESI):368.2[M+H] +
The fourth step: preparation of 3- (1, 1-trifluoropropan-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [4,3-d ] pyrimidin-7-ol
Methyl 4-amino-3- (1, 1-trifluoropropan-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole-5-carboxylate (3.00g, 7.76mmol) and formamidine acetate (1.22g, 11.63mmol) were added to N, N-diisopropylethylamine (20 mL) and N-butanol (20 mL) and reacted at 110 ℃ for 2 hours. The reaction solution was diluted with dichloromethane, washed with water, and the crude product obtained by concentrating the organic phase was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =2/1 (v/v)) to obtain the title compound of this step (1.9 g, yield: 64%).
MS m/z(ESI):363.1[M+H] +
The fifth step: preparation of N- (2, 4-dimethoxybenzyl) -3- (1, 1-trifluoropropan-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [4,3-d ] pyrimidin-7-amine
3- (1, 1-trifluoropropan-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [4,3-d ] pyrimidin-7-ol (294.74mg, 0.78mmol) was dissolved in tetrahydrofuran (5 mL), and 1, 8-diazabicyclo [5.4.0] undec-7-ene (294.82mg, 1.16mmol), a Cartesian condensing agent (207.08mg, 1.00mmol) and 2, 4-dimethoxybenzylamine (260.97mg, 1.55mmol) were added and reacted at room temperature for 2 hours. The reaction solution was diluted with dichloromethane, washed with water, and the crude product obtained by concentration of the organic phase was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =3/1 (v/v)) to obtain the title compound of this step (260 mg, yield: 62%).
MS m/z(ESI):512.2[M+H] +
And a sixth step: preparation of 3- (1, 1-trifluoropropan-2-yl) -1H-pyrazolo [4,3-d ] pyrimidin-7-amine
N- (2, 4-Dimethoxybenzyl) -3- (1, 1-trifluoropropan-2-yl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [4,3-d ] pyrimidin-7-amine (421.05mg, 0.78mmol) was dissolved in trifluoroacetic acid (5 mL) and reacted at 70 ℃ for 5 hours. The reaction solution was concentrated, diluted with methanol, adjusted to pH =8 with a saturated sodium bicarbonate solution, and the crude product obtained by the concentration was purified by silica gel column chromatography (eluent: dichloromethane/methanol =10/1 (v/v)) to obtain the title compound of the present step (100 mg, yield: 52%).
MS m/z(ESI):232.1[M+H] +
Intermediate preparation example 8: preparation of (3-fluoro-4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid
Figure BDA0003222255240000401
The first step is as follows: preparation of 4-bromo-2-fluoro-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of this step was synthesized in a similar manner to the procedure described for the first step of intermediate preparation example 1, substituting 4-bromo-2-fluoro-benzoyl chloride for 4-bromobenzoyl chloride of the first step of intermediate preparation example 1 (1.4 g, yield: 65%).
MS m/z(ESI):363.0[M+H] +
The second step: preparation of 2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of this step was synthesized in analogy to the procedure described in the second step of intermediate preparation example 1, using 4-bromo-2-fluoro-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide instead of 4-bromo-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide in the second step of intermediate preparation example 1 (200 mg, yield: 59%).
MS m/z(ESI):411.1[M+H] +
The third step: preparation of (3-fluoro-4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid
The title compound of this step was synthesized in a similar manner to the procedure described in the third step of intermediate preparation example 1 by substituting 2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide in the third step of intermediate preparation example 1 with 2-fluoro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (200 mg, yield: 80%).
MS m/z(ESI):329.1[M+H] +
Intermediate preparation example 9: preparation of 4-bromo-N- (2-fluorophenyl) benzamide
Figure BDA0003222255240000402
The title compound (600 mg, yield: 81%) of this step was synthesized in a similar manner to the procedure described in the first step of intermediate preparation example 1, substituting 2-fluoroaniline for 4- (trifluoromethyl) pyridin-2-amine of the first step of intermediate preparation example 1.
MS m/z(ESI):294.0[M+H] +
Intermediate preparation example 10: preparation of 4-amino-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Figure BDA0003222255240000411
The first step is as follows: preparation of 4-nitro-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound (550 mg, yield: 73%) of this step was synthesized in a similar manner to the procedure described for the first step of intermediate preparation example 1, substituting 4-nitrobenzoyl chloride for 4-bromobenzoyl chloride of the first step of intermediate preparation example 1.
MS m/z(ESI):312.1[M+H] +
The second step: preparation of 4-amino-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4-Nitro-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (550mg, 1.68mmol), iron powder (473.57mg, 8.39mmol) and ammonium chloride (45.36mg, 0.84mmol) were added to ethanol (14 mL) and water (7 mL) and reacted at 80 ℃ for 2 hours. The reaction solution was filtered, the cake was washed with ethyl acetate, the combined filtrates were extracted four times with ethyl acetate, the combined organic layers were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was slurried with methylene chloride and petroleum ether to give the title compound of this step (450 mg, yield: 91%).
MS m/z(ESI):282.1[M+H] +
Intermediate preparation example 11: preparation of 2-hydroxy-1- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) ethan-1-one
Figure BDA0003222255240000412
2-Hydroxyacetic acid (597.65mg, 7.86mmol), 1,2,3, 6-tetrahydropyridine-4-boronic acid pinacol ester (1.26g, 6.05mmol) and triethylamine (1.22g, 12.09mmol) were sequentially added to tetrahydrofuran (30 mL), the reaction mixture was cooled to 0 ℃ and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (2.98g, 7.86mmol) was added in portions and then reacted at 25 ℃ for 12 hours. The residue obtained by concentrating the reaction solution was extracted three times with water and ethyl acetate, and the organic phase was washed successively with a saturated aqueous potassium carbonate solution, 1N dilute hydrochloric acid and saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound of this step (1.5 g, yield: 93%).
MS m/z(ESI):268.2[M+H] +
Intermediate preparation example 12: preparation of (4- ((5-fluoro-2-methoxybenzamide) methyl) phenyl) boronic acid
Figure BDA0003222255240000421
The first step is as follows: preparation of N- (4-bromobenzyl) -5-fluoro-2-methoxybenzamide
5-fluoro-2-methoxy-benzoic acid (22.86g, 131.69mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (33.15g, 171.19mmol), 1-hydroxybenzotriazole (17.97g, 131.69mmol) and N, N-diisopropylethylamine (34.38g, 263.37mmol) were dissolved in dichloromethane (20 mL), stirred for 20 minutes, p-bromobenzylamine (25g, 131.69mmol) was added, and the reaction was carried out at room temperature for 12 hours. The reaction solution was concentrated, extracted with water and ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was slurried with ethyl acetate and methyl tert-butyl ether to give the title compound of this step (42 g, yield: 90%).
MS m/z(ESI):338.0[M+H] +
The second step is that: preparation of 5-fluoro-2-methoxy-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzyl) benzamide
N- (4-bromobenzyl) -5-fluoro-2-methoxybenzamide (42g, 117.99mmol), pinacol diborate (33.29g, 129.79mmol), potassium acetate (23.39g, 235.98mmol) and bis (triphenylphosphine) palladium dichloride (4.18g, 5.90mmol) were added to 1, 4-dioxane (40 mL) and reacted at 80 ℃ for 4 hours while displacing nitrogen 3 times. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether =3/1 (v/v)) to obtain the title compound of this step (40 g, yield: 88%).
MS m/z(ESI):386.2[M+H] +
The third step: preparation of (4- ((5-fluoro-2-methoxybenzamide) methyl) phenyl) boronic acid
5-fluoro-2-methoxy-N- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzyl) benzamide (0.7 g, 1.78mmol) and sodium periodate (1.15g, 5.34mmol) were added to tetrahydrofuran (15 mL) and water (3 mL), stirred at room temperature for half an hour, diluted hydrochloric acid (1N, 1.78mL) was added, and reacted at room temperature for 2 hours. Water and ethyl acetate were added for extraction, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound of this step (0.53 g, yield: 97%).
MS m/z(ESI):304.1[M+H] +
Example (b):
example 1: preparation of 4- (7-amino-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 1)
Figure BDA0003222255240000431
The first step is as follows: preparation of methyl 4-amino-3- (3, 6-dihydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylate
Methyl 4-amino-3-bromo-1H-pyrazole-5-carboxylate (1.02g, 4.54mmol), 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester (1.17g, 5.45mmol) and potassium carbonate (1.90g, 13.63mmol) were added to 1, 4-dioxane (6 mL) and water (1 mL), bis-triphenylphosphine palladium dichloride (325.52mg, 0.45mmol) was added and the mixture was heated with a microwave at 110 ℃ for reaction for 3 hours. After completion of the reaction, an appropriate amount of water was added, extracted four times with ethyl acetate, and the organic phase was concentrated to give a crude product which was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =3/1 (v/v)) to obtain the title compound of the present step (400 mg, yield: 38%).
MS m/z(ESI):224.1[M+H] +
The second step: preparation of 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylic acid methyl ester
4-amino-3- (3, 6-dihydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylic acid methyl ester (400mg, 1.70mmol) was dissolved in methanol (8 mL), palladium on carbon (200mg, 10%) was added, and after three times of replacement with hydrogen, the reaction was carried out at room temperature for 5 hours in a hydrogen atmosphere. After completion of the reaction, the reaction mixture was filtered through Celite, and the filtrate was concentrated to give the title compound of the present step (240 mg, yield: 85%).
MS m/z(ESI):226.1[M+H] +
The third step: preparation of methyl 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate
Methyl 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylate (210mg, 913.68. Mu. Mol), (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid (477.09mg, 1.46mmol),
Figure BDA0003222255240000441
Molecular sieves (100 mg), copper acetate (276.39mg, 1.37mmol) and pyridine (146.00mg, 1.83mmol) were added to 1, 2-dichloroethane (8 mL) and reacted at 60 ℃ for 7 hours. After completion of the reaction, the crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/4 (v/v)) to obtain the title compound of the present step (330 mg, yield: 70%).
MS m/z(ESI):490.2[M+H] +
The fourth step: preparation of 4- (7-hydroxy-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Methyl 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate (330mg, 0.64mmol), N-diisopropylethylamine (165.63mg, 1.28mmol) and formamidine acetate (134.70mg, 1.28mmol) were added to N-butanol (8 mL) and reacted at 110 ℃ for 2 hours. After completion of the reaction, the crude product obtained by concentrating the reaction liquid was purified by silica gel column chromatography (eluent: dichloromethane/methanol =10/1 (v/v)) to obtain the title compound of the present step (171 mg, yield: 52%).
MS m/z(ESI):485.1[M+H] +
The fifth step: preparation of 4- (7- ((2, 4-dimethoxybenzyl) amino) -3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (7-hydroxy-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (171mg, 0.33mmol), a Cartesian condensing agent (138.28mg, 0.67mmol), 2, 4-dimethoxybenzylamine (84.96mg, 0.50mmol) and 1, 8-diazabicyclo [5.4.0] undec-7-ene (170.63mg, 0.67mmol) were added to tetrahydrofuran (8 mL) and the mixture was reacted with nitrogen at 40 ℃ for 6 hours under nitrogen atmosphere while replacing nitrogen 3 times. After completion of the reaction, an appropriate amount of water and ethyl acetate were added for extraction, the organic layers were combined, and the crude product obtained by concentration of the organic layer was purified by silica gel column chromatography (eluent: dichloromethane/methanol =20/1 (v/v)) to obtain the title compound of the present step (150 mg, yield: 64%).
MS m/z(ESI):634.2[M+H] +
And a sixth step: preparation of 4- (7-amino-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (7- ((2, 4-dimethoxybenzyl) amino) -3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (80mg, 0.13mmol) was dissolved in trifluoroacetic acid (6 mL) and reacted at 80 ℃ for 4 hours. The crude product obtained by concentrating the reaction liquid was purified by high performance liquid chromatography (preparation method 2) to obtain the title compound (45 mg, yield: 70.0%) of this step.
MS m/z(ESI):484.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.44(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.35(d,J=5.6Hz,1H),8.28(d,J=8.8Hz,2H),7.67(d,J=8.4Hz,2H),7.59(d,J=5.2Hz,1H),6.72(s,2H),4.10-3.94(m,2H),3.54(td,J=11.2,3.6Hz,3H),2.04(ddd,J=13.6,11.2,4.4Hz,4H)。
Example 2: preparation of 4- (7-amino-3-isopropyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 2)
Figure BDA0003222255240000451
The first step is as follows: preparation of 4-amino-3- (prop-1-en-2-yl) -1H-pyrazole-5-carboxylic acid methyl ester
The title compound of this step was synthesized in a similar manner to the procedure described in the first step of example 1, using isopropenylboronic acid pinacol ester instead of 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester of the first step of example 1 (1.02 g, yield: 80%).
MS m/z(ESI):182.1[M+H] +
The second step is that: preparation of methyl 4-amino-3- (prop-1-en-2-yl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate
The title compound of this step (1.1 g, yield: 32%) was synthesized in analogy to the procedure described in the third step of example 1, substituting 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylic acid methyl ester for 4-amino-3- (prop-1-en-2-yl) -1H-pyrazole-5-carboxylic acid methyl ester in the third step of example 1.
MS m/z(ESI):446.1[M+H] +
The third step: preparation of methyl 4-amino-3-isopropyl-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate
The title compound of this step was synthesized in a similar manner to the procedure described in the second step of example 1, using methyl 4-amino-3- (prop-1-en-2-yl) -1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate instead of methyl 4-amino-3- (3, 6-dihydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylate of the second step in example 1 (480 mg, yield: 60%).
MS m/z(ESI):448.2[M+H] +
The fourth step: preparation of 4- (7-hydroxy-3-isopropyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
In analogy to the procedure described for the fourth step of example 1, the title compound of this step was synthesized using 4-amino-3-isopropyl-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylic acid methyl ester instead of 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1- (4- (4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylic acid methyl ester of the fourth step in example 1 (300 mg, yield: 72%).
MS m/z(ESI):443.1[M+H] +
The fifth step: preparation of 4- (7- ((2, 4-dimethoxybenzyl) amino) -3-isopropyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of this step was synthesized in a similar manner to the procedure described in the fifth step of example 1 by using 4- (7-hydroxy-3-isopropyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide instead of 4- (7-hydroxy-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4-trifluoromethylpyridin-2-yl) benzamide in the fifth step of example 1 (180 mg, yield: 90%).
MS m/z(ESI):592.2[M+H] +
And a sixth step: preparation of 4- (7-amino-3-isopropyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of the present step was synthesized in analogy to the procedure described in the sixth step of example 1, with 4- (7- ((2, 4-dimethoxybenzyl) amino) -3-isopropyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide replacing the sixth step of example 1 with 4- (7- ((2, 4-dimethoxybenzyl) amino) -3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (180 mg, yield: 90%).
MS m/z(ESI):442.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.37(s,1H),8.27(d,J=8.4Hz,2H),7.66(d,J=8.4Hz,2H),7.59(d,J=5.2Hz,1H),6.82(s,2H),3.50-3.43(m,1H),1.45(d,J=6.8Hz,6H)。
Example 3: preparation of 4- (7-amino-3- (1, 1-trifluoropropan-2-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (compound 3)
Figure BDA0003222255240000471
(4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid (193.04mg, 0.62mmol) and 3- (1, 1-trifluoropropan-2-yl) -1H-pyrazolo [4, 3-d)]Pyrimidin-7-amine (150mg, 0.62mmol) was dissolved in 1, 2-dichloroethane (5 mL) and added
Figure BDA0003222255240000472
Molecular sieves (100 mg), copper acetate (226.14mg, 1.23mmol) and pyridine (98.50mg, 1.23) were reacted at 40 ℃ for 6 hours. The reaction solution was concentrated, diluted with methylene chloride, washed with water, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by high performance liquid chromatography (preparation method 1) to obtain the title compound of this step (5 mg, yield: 2%).
MS m/z(ESI):496.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,1H),8.71(d,J=5.2Hz,1H),8.58(s,1H),8.39(s,1H),8.28(d,J=8.4Hz,2H),7.70(d,J=8.8Hz,2H),7.58(d,J=5.2Hz,1H),6.91(s,2H),4.55-4.07(m,1H),1.68(d,J=7.2Hz,3H)。
Example 4: preparation of 4- (7-amino-3- (1, 1-trifluoropropan-2-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -2-fluoro-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 4)
Figure BDA0003222255240000473
The title compound (10 mg, yield: 8%) of this step was synthesized in analogy to the procedure described in example 3, substituting (3-fluoro-4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid for (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) boronic acid in example 3.
MS m/z(ESI):514.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.39(s,1H),8.69(d,J=5.2Hz,1H),8.55(s,1H),8.40(s,1H),7.93(t,J=8.0Hz,1H),7.66-7.57(m,2H),7.53(dd,J=8.4,2.0Hz,1H),7.02(br,2H),4.32(dt,J=16.4,8.4Hz,1H),1.67(d,J=7.2Hz,3H)。
Example 5: preparation of 4- (7-amino-3- (1-isobutyrylpiperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 5)
Figure BDA0003222255240000481
Isobutyric acid (11mg, 0.12mmol), 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (47mg, 0.12mmol) and N, N-diisopropylethylamine (1695g, 0.12mmol) were dissolved in tetrahydrofuran (2 mL), and after stirring at room temperature for 15 minutes, 4- (7-amino-3- (piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (40mg, 0.083mmol) was added and reacted at room temperature for 16 hours. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 1) to obtain the title compound of this step (6 mg, yield: 13%).
MS m/z(ESI):553.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.71(d,J=5.2Hz,1H),8.57(s,1H),8.36(d,J=7.6Hz,1H),8.27(d,J=8.4Hz,2H),7.66(d,J=8.4Hz,2H),7.60-7.55(m,1H),7.26-6.44(m,2H),4.77-4.67(m,0.41H),4.32-4.18(m,0.94H),4.06-3.95(m,0.45H),3.78(s,1H),3.73(s,0.70H),3.61-3.51(m,0.62H),3.19-3.09(m,1.27H),3.02-2.79(m,2H),2.25-2.16(m,1H),2.15-1.95(m,1H),1.93-1.75(m,1H),1.64-1.42(m,1H),1.10-1.02(m,3H),0.95(dd,J=37.2,6.4Hz,3H)。
Example 6: preparation of 3- (7-amino-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) -N, N-dimethylpiperidine-1-carboxamide (compound 6)
Figure BDA0003222255240000482
4- (7-amino-3- (piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (70mg, 0.14mmol) and N, N-diisopropylethylamine (35.99mg, 0.28mmol) were dissolved in dichloromethane (3 mL), and N, N-dimethylcarbonyl chloride (16.47mg, 0.15mmol) was added and reacted at room temperature for 16 hours. The crude product obtained by concentrating the reaction liquid was purified by high performance liquid chromatography (preparation method 2) to obtain the title compound (20 mg, yield: 25%) of this step.
MS m/z(ESI):554.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.42-8.35(m,1H),8.28(d,J=8.8Hz,2H),7.67(t,J=7.2Hz,2H),7.59(d,J=5.2Hz,1H),6.84(s,2H),3.93(d,J=12.8Hz,1H),3.60(d,J=12.8Hz,1H),3.34-3.25(m,2H),3.16-3.06(m,1H),2.80-2.73(m,6H),2.22(d,J=10.4Hz,1H),1.94(ddd,J=15.6,12.4,3.6Hz,1H),1.86-1.76(m,1H),1.72-1.57(m,1H)。
Example 7: preparation of 4- (3- (1-acetylpiperidin-3-yl) -7-amino-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 7)
Figure BDA0003222255240000491
The title compound (5 mg, yield: 18%) of this step was synthesized in a similar manner to the procedure described in example 5, using acetic acid instead of isobutyric acid in example 5.
MS m/z(ESI):525.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.38(d,J=7.2Hz,1H),8.28(d,J=7.2Hz,2H),7.68(dd,J=8.4,2.8Hz,2H),7.59(d,J=5.2Hz,1H),6.84(s,2H),4.18-4.84(m,1H),3.96-3.51(m,1H),3.15(t,J=11.2Hz,1H),3.04-2.81(m,1H),2.72-2.66(m,1H),2.38-2.31(m,1H),2.19-2.28(m,1H),2.05(d,J=12.4Hz,3H),1.91-1.72(m,1H),1.71-1.41(m,1H)。
Example 8: preparation of 4- (7-amino-3- (1- (2-hydroxyacetyl) piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 8)
Figure BDA0003222255240000501
The title compound (8 mg, yield: 16%) of this step was synthesized in a similar manner to the method described in example 5, using 2-glycolic acid instead of isobutyric acid in example 5.
MS m/z(ESI):541.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.73(d,J=5.2Hz,1H),8.59(s,1H),8.38(d,J=5.6Hz,1H),8.28(d,J=8.8Hz,2H),7.73-7.64(m,2H),7.59(d,J=5.2Hz,1H),6.85(s,2H),4.77-3.94(m,5H),3.80-3.42(m,1H),3.26-2.84(m,2H),2.20-2.31(m,1H),1.95-2.14(m,1H),1.75-1.86(m,1H),1.44(s,1H)。
Example 9: preparation of 4- (7-amino-3- (1- (1-hydroxycyclopropane-1-formyl) piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (compound 9)
Figure BDA0003222255240000502
The title compound (5 mg, yield: 12%) of this step was synthesized in a similar manner to the procedure described in example 5, using 1-hydroxycyclopropane-1-carboxylic acid instead of isobutyric acid in example 5.
MS m/z(ESI):567.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.73(d,J=5.2Hz,1H),8.59(s,1H),8.38(s,1H),8.28(d,J=8.8Hz,2H),7.68(d,J=8.6Hz,1H),7.59(d,J=5.1Hz,1H),6.36(s,1H),4.73(s,3H),2.24(d,J=11.6Hz,1H),2.00(d,J=10.9Hz,1H),1.85(d,J=13.7Hz,1H),1.61(s,1H),0.96(s,1H),0.76(s,1H)。
Example 10: preparation of 4- (7-amino-3- (1- ((R) -2-hydroxypropionyl) piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 10)
Figure BDA0003222255240000511
The title compound (16 mg, yield: 28%) of this step was synthesized in a similar manner to the method described in example 5, using (R) -2-hydroxypropionic acid instead of isobutyric acid in example 5.
MS m/z(ESI):555.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.73(d,J=5.2Hz,1H),8.59(s,1H),8.38(d,J=4.4Hz,1H),8.28(d,J=8.4Hz,2H),7.68(d,J=8.8Hz,2H),7.59(d,J=5.2Hz,1H),6.87(s,2H),4.49(s,1H),4.40-4.23(m,1H),3.54(dd,J=24.4,13.2Hz,1H),3.23-2.95(m,2H),2.23(s,1H),2.14-1.97(m,1H),1.82(d,J=15.6Hz,1H),1.70-1.37(m,2H),1.37-1.10(m,4H)。
Example 11: preparation of 4- (7-amino-3- (1- (2, 2-trifluoroethyl) piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 11)
Figure BDA0003222255240000512
4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (73mg, 0.14mmol) and potassium carbonate (21mg, 0.15mmol) were added to acetonitrile (2 mL), followed by addition of 2, 2-trifluoroethyltrifluoromethanesulfonate (47mg, 0.20mmol), and reacted at room temperature for 4 hours. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 1) to obtain the title compound (17 mg, yield: 22%) of this step.
MS m/z(ESI):565.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.71(d,J=5.2Hz,1H),8.57(s,1H),8.36(s,1H),8.26(d,J=8.8Hz,2H),7.65(d,J=8.4Hz,2H),7.60-7.54(m,1H),6.89(s,1H),3.27-3.20(m,2H),3.11(dd,J=15.2,6.8Hz,1H),3.05(d,J=11.2Hz,2H),2.58-2.52(m,2H),2.03(td,J=9.2,8.0,4.0Hz,4H)。
Example 12: preparation of 4- (7-amino-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) -N, N-dimethylpiperidine-1-carboxamide (compound 12)
Figure BDA0003222255240000521
4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (210.53mg, 0.41mmol) and N, N-diisopropylethylamine (107.19mg, 0.83mmol) were dissolved in tetrahydrofuran (10 mL), and N, N-dimethylformamide chloride (49.53mg, 0.46mmol) was slowly added and reacted at room temperature for 4 hours. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 1) to obtain the title compound (50 mg, yield: 21%) of this step.
MS m/z(ESI):554.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.40(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.37(d,J=5.6Hz,1H),8.28(d,J=8.4Hz,2H),7.66(t,J=6.8Hz,2H),7.57(d,J=5.2,1H),6.73(s,2H),3.69(t,J=14.4Hz,2H),3.30(ddd,J=15.2,7.6,3.6Hz,1H),2.92(t,J=11.2Hz,2H),2.77(s,6H),2.14-2.03(m,2H),1.94(qd,J=12.4,3.6Hz,2H)。
Example 13: preparation of 4- (7-amino-3- (1-isobutyrylpiperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 13)
Figure BDA0003222255240000522
4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (63.16mg, 0.12mmol) and isobutyric acid (22.36mg, 0.25mmol) were dissolved in tetrahydrofuran (10 mL), and 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (61.80mg, 0.16mmol) and N, N-diisopropylethylamine (32.16mg, 0.25mmol) were added and reacted at room temperature for 6 hours. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by high performance liquid chromatography to give the title compound of this step (13 mg, yield: 18%).
MS m/z(ESI):553.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.36(s,1H),8.28(d,J=8.4Hz,2H),7.67(d,J=8.8Hz,2H),7.58(d,J=5.2Hz,1H),6.84(s,2H),4.49(d,J=11.6Hz,2H),4.08(d,J=12.8Hz,2H),2.94(dt,J=13.6,6.8Hz,1H),2.82(t,J=12.4Hz,1H),2.12(t,J=15.6Hz,2H),1.86(dd,J=56.0,10.2Hz,2H),1.09-0.99(m,6H)。
Example 14: preparation of 4- (7-amino-3- (1- (2-hydroxyacetyl) piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 14)
Figure BDA0003222255240000531
The title compound (25 mg, yield: 37%) of this step was synthesized in a similar manner to the method described in example 13, using 2-glycolic acid instead of isobutyric acid in example 13.
MS m/z(ESI):541.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.36(d,J=5.6Hz,1H),8.28(d,J=8.8Hz,2H),7.67(d,J=8.8Hz,2H),7.65-7.47(m,1H),6.84(s,2H),4.56(dt,J=10.8,5.6Hz,1H),4.44(d,J=12.8Hz,1H),4.24-4.08(m,2H),3.81(d,J=13.2Hz,1H),3.43(ddd,J=15.2,7.6,3.6Hz,1H),3.20(t,J=12.0Hz,1H),2.93(dd,J=25.2,13.6Hz,1H),2.10(d,J=13.2Hz,2H),1.78-2.04(m,2H)。
Example 15: preparation of 4- (7-amino-3- (1- (1-hydroxycyclopropane-1-formyl) piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (compound 15)
Figure BDA0003222255240000541
The title compound (15 mg, yield: 20%) of this step was synthesized in a similar manner to the method described in example 13, using 1-hydroxycyclopropane-1-carboxylic acid instead of isobutyric acid in example 13.
MS m/z(ESI):567.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.37(s,1H),8.28(d,J=8.8Hz,2H),7.67(d,J=8.8Hz,2H),7.59(d,J=5.2Hz,1H),6.82(s,2H),6.36(s,1H),4.51(s,3H),3.51-3.39(m,1H),3.15(s,2H),2.09(t,J=11.6Hz,2H),1.94(s,2H),0.93(s,2H),0.79(d,J=2.4Hz,2H)。
Example 16: preparation of 4- (3- (1-acetylpiperidin-4-yl) -7-amino-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 16)
Figure BDA0003222255240000542
The title compound (18 mg, yield: 26%) of this step was synthesized in a similar manner to the method described in example 13, substituting acetic acid for isobutyric acid in example 13.
MS m/z(ESI):525.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(d,J=15.6Hz,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.36(s,1H),8.29(t,J=10.4Hz,2H),7.66(t,J=7.6Hz,2H),7.58(d,J=5.2Hz,1H),6.69(s,2H),4.46(d,J=13.2Hz,1H),3.93(t,J=14.4Hz,1H),3.41(ddd,J=13.6,7.2,3.2Hz,1H),3.26(dd,J=18.4,7.2Hz,1H),2.81(t,J=11.4Hz,1H),2.14-2.01(m,5H),2.00-1.90(m,1H),1.81(ddd,J=15.6,12.0,5.2Hz,1H)。
Example 17: preparation of (R) -4- (7-amino-3- (1- (2-hydroxypropionyl) piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 17)
Figure BDA0003222255240000551
The title compound (18 mg, yield: 25%) of this step was synthesized in a similar manner to the method described in example 13, using (R) -2-hydroxypropionic acid instead of isobutyric acid in example 13.
MS m/z(ESI):555.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.36(s,1H),8.28(d,J=8.4Hz,2H),7.72-7.64(m,2H),7.59(d,J=5.2Hz,1H),6.72(s,2H),4.88(dd,J=12.4,6.8Hz,1H),4.40-4.55(m,2H),4.14(d,J=12.0Hz,1H),3.43(t,J=11.2Hz,1H),3.31-3.16(m,1H),2.89(dd,J=24.4,12.0Hz,1H),2.24-1.71(m,4H),1.28-1.10(m,3H)。
Example 18: (S) -4- (7-amino-3- (1- (2-hydroxypropionyl) piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (compound 18)
Figure BDA0003222255240000552
The title compound (18 mg, yield: 25%) of this step was synthesized in a similar manner to the method described in example 13, using (S) -2-hydroxypropionic acid instead of isobutyric acid in example 13.
MS m/z(ESI):555.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.72(d,J=5.2Hz,1H),8.59(s,1H),8.36(s,1H),8.28(d,J=8.8Hz,2H),7.67(d,J=8.8Hz,2H),7.59(d,J=5.2Hz,1H),6.82(s,2H),4.88(dd,J=12.4,6.8Hz,1H),4.59-4.36(m,2H),4.11(d,J=12.4Hz,1H),3.38-3.50(m,1H),3.30-3.17(m,1H),2.89(dd,J=24.8,11.6Hz,1H),2.17-1.77(m,4H),1.28-1.12(m,3H)。
Example 19: preparation of 4- (3- (1-acetylpiperidin-4-yl) -7-amino-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (2-fluorophenyl) benzamide (compound 19)
Figure BDA0003222255240000561
The first step is as follows: preparation of tert-butyl 4- (4-amino-1- (4- ((2-fluorophenyl) carbamoyl) phenyl) -5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate
Tert-butyl 4- (4-amino-5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate (551mg, 1.61mmol), 4-bromo-N- (2-fluorophenyl) benzamide (600mg, 1.94mmol), cuprous iodide (310.68mg, 1.61mmol), trans-N, N' -dimethyl-1, 2-cyclohexanediamine (232.04mg, 1.61mmol) and potassium carbonate (450.93mg, 3.23mmol) were added to N, N-dimethylformamide (5 mL), nitrogen was substituted, and the reaction was heated with microwave at 110 ℃ for 2 hours. After completion of the reaction, the reaction solution was poured into water, extracted four times with ethyl acetate, and the crude product obtained by concentration of the organic phase was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/2 (v/v)) to obtain the title compound of the present step (900 mg, yield: 93%).
MS m/z(ESI):538.2[M+H] +
The second step: preparation of tert-butyl 4- (1- (4- ((2-fluorophenyl) carbamoyl) phenyl) -7-hydroxy-1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
In a similar manner to the procedure described in the fourth step of example 1, the title compound of this step was synthesized using tert-butyl 4- (4-amino-1- (4- ((2-fluorophenyl) carbamoyl) phenyl) -5- (methoxycarbonyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate instead of methyl 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1- (4- (4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate of the fourth step in example 1 (820 mg, yield: 92%).
MS m/z(ESI):533.2[M+H] +
The third step: preparation of tert-butyl 4- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((2-fluorophenyl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
The title compound of this step was synthesized in a similar manner to the procedure described in the fifth step of example 1 using tert-butyl 4- (1- (4- ((2-fluorophenyl) carbamoyl) phenyl) -7-hydroxy-1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate instead of 4- (7-hydroxy-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4-trifluoromethylpyridin-2-yl) benzamide of the fifth step in example 1 (650 mg, yield: 63%).
MS m/z(ESI):682.3[M+H] +
The fourth step: preparation of 4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (2-fluorophenyl) benzamide
Tert-butyl 4- (7- ((2, 4-dimethoxybenzyl) amino) -1- (4- ((2-fluorophenyl) carbamoyl) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate (630mg, 0.74mmol) was dissolved in trifluoroacetic acid (8 mL) and reacted at 80 ℃ for 4 hours. The reaction solution was concentrated, the pH was adjusted to weak alkalinity with triethylamine, and the resulting crude product was further concentrated and purified by silica gel column chromatography (eluent: dichloromethane/methanol =8/1 (v/v)) to obtain the title compound of this step (100 mg, yield: 28%).
MS m/z(ESI):432.2[M+H] +
The fifth step: preparation of 4- (3- (1-acetylpiperidin-4-yl) -7-amino-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (2-fluorophenyl) benzamide
Glacial acetic acid (25.31mg, 0.42mmol), 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (120.20 mg, 0.31mmol), and N, N-diisopropylethylamine (53.94mg, 0.42mmol) were dissolved in tetrahydrofuran (6 mL) at 0 deg.C, stirred for half an hour, and then 4- (7-amino-3- (piperidin-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (2-fluorophenyl) benzamide (98.90mg, 0.21mmol) was added and reacted at room temperature for 1 hour. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 1) to obtain the title compound of this step (32 mg, yield: 31%).
MS m/z(ESI):474.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.28(d,J=8.8Hz,1H),8.36(s,1H),8.21(d,J=8.4Hz,2H),7.80-7.52(m,3H),7.46-7.21(m,3H),7.16-6.51(m,2H),4.46(d,J=13.1Hz,1H),3.94(d,J=13.1Hz,1H),3.26(t,J=11.6Hz,1H),2.80(t,J=11.6Hz,1H),2.23-1.56(m,8H)。
Example 20: preparation of 4- (7-amino-3- (4-hydroxycyclohexyl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (compound 20)
Figure BDA0003222255240000581
The first step is as follows: preparation of methyl 4-amino-3-bromo-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylate
The title compound of this step was synthesized (500 mg, yield: 31%) in analogy to the procedure described in the third step of example 1, using 4-amino-3-bromo-1H-pyrazole-5-carboxylic acid methyl ester instead of 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazole-5-carboxylic acid methyl ester in the third step of example 1.
MS m/z(ESI):484.0[M+H] +
The second step is that: preparation of 4- (3-bromo-7-hydroxy-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
In analogy to the procedure described for the fourth step of example 1, the title compound of this step was synthesized using 4-amino-3-bromo-1- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylic acid methyl ester instead of 4-amino-3- (tetrahydro-2H-pyran-4-yl) -1- (4- (4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) -1H-pyrazole-5-carboxylic acid methyl ester of the fourth step in example 1 (494 mg, yield: 99%).
MS m/z(ESI):479.0[M+H] +
The third step: preparation of 4- (3-bromo-7- ((2, 4-dimethoxybenzyl) amino) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of this step was synthesized in a similar manner to the procedure described in the fifth step of example 1 by using 4- (3-bromo-7-hydroxy-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide instead of 4- (7-hydroxy-3- (tetrahydro-2H-pyran-4-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4-trifluoromethylpyridin-2-yl) benzamide of the fifth step of example 1 (183 mg, yield: 28%).
MS m/z(ESI):628.1[M+H] +
The fourth step: preparation of 4- (7- ((2, 4-dimethoxybenzyl) amino) -3- (1, 4-dioxaspiro [4.5] dec-7-en-8-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (3-bromo-7- ((2, 4-dimethoxybenzyl) amino) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (170mg, 0.27mmol), 1, 4-dioxaspiro [4,5] dec-7-ene-8-boronic acid pinacol ester (108mg, 0.41mmol), bistriphenylphosphine palladium dichloride (19mg, 0.03mmol) and potassium phosphate (86mg, 0.41mmol) were added to a mixed solvent of 1, 4-dioxane (10 mL) and water (1 mL), and reacted at 99 ℃ for 12 hours under nitrogen substitution. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =1/1 (v/v)) to obtain the title compound of this step (144 mg, yield: 77%).
MS m/z(ESI):688.2[M+H] +
The fifth step: preparation of 4- (7- ((2, 4-dimethoxybenzyl) amino) -3- (1, 4-dioxaspiro [4.5] decan-8-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (7- ((2, 4-Dimethoxybenzyl) amino) -3- (1, 4-dioxaspiro [4.5] dec-7-en-8-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (154mg, 0.22mmol) was dissolved in a mixed solvent of methanol (3 mL) and ethyl acetate (3 mL), palladium on carbon (77mg, 10%) was added, hydrogen substitution was performed, and the reaction was performed at room temperature under a hydrogen atmosphere for 16 hours. The reaction mixture was filtered through Celite, and the filtrate was concentrated to give the title compound of this step (143 mg, yield: 93%).
MS m/z(ESI):690.3[M+H] +
And a sixth step: preparation of 4- (7-amino-3- (4-oxocyclohexyl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (7- ((2, 4-Dimethoxybenzyl) amino) -3- (1, 4-dioxaspiro [4.5] decan-8-yl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (143mg, 0.21mmol) was dissolved in trifluoroacetic acid (5 mL) and reacted at 70 ℃ for 5 hours with nitrogen substitution. The reaction solution was concentrated, the system pH was adjusted to weak alkaline by adding ammonia water, extraction was performed with ethyl acetate, the organic phase was dried over anhydrous sodium sulfate, filtration was performed, and the crude product obtained by concentrating the filtrate was purified with a preparative thin layer chromatography silica gel plate (developing solvent: ethyl acetate/acetonitrile =3/1 (v/v)) to obtain the title compound of the present step (102 mg, yield: 99%).
MS m/z(ESI):496.2[M+H] +
The seventh step: preparation of 4- (7-amino-3- (4-hydroxycyclohexyl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (7-amino-3- (4-oxocyclohexyl) -1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (127mg, 0.21mmol) was dissolved in anhydrous tetrahydrofuran (3 mL), and sodium borohydride (12mg, 0.31mmol) was added thereto, and the mixture was replaced with nitrogen and reacted at room temperature for 1 hour. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 1) to obtain the title compound of this step (40 mg, yield: 19%).
MS m/z(ESI):498.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,1H),8.71(d,J=5.2Hz,1H),8.57(s,1H),8.34(s,1H),8.28-8.23(m,2H),7.65(td,J=6.8,2.0Hz,2H),7.59-7.55(m,1H),6.78(s,2H),4.63(d,J=4.4Hz,1H),3.50(ddt,J=10.4,8.4,4.4Hz,1H),3.01(tt,J=11.6,3.6Hz,1H),2.13-2.02(m,2H),1.97(dd,J=12.4,3.2Hz,2H),1.84-1.73(m,2H),1.39-1.29(m,2H)。
Example 21: preparation of 4- (7-amino-3-cyclopentyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (compound 21)
Figure BDA0003222255240000601
The first step is as follows: preparation of methyl 2-cyano-2-cyclopentanylideneacetate
Methyl 2-cyanoacetate (3.03g, 30.28mmol) and cyclopentanone (2.57g, 30.28mmol) were dissolved in toluene (20 mL), and ammonium acetate (1.17g, 15.14mmol) and glacial acetic acid (1.23g, 20.46mmol) were added and reacted at 110 ℃ for 12 hours. The crude product obtained by concentrating the reaction solution was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =3/1 (v/v)) to obtain the title compound of this step (4 g, yield: 79%).
MS m/z(ESI):331.1[2M+H] +
The second step is that: preparation of methyl 2-cyano-2-cyclopentylacetate
Methyl 2-cyano-2-cyclopentanylideneacetate (2.11g, 12.11mmol) was dissolved in methanol (30 mL), palladium on carbon (1.47g, 10%) was added, and the mixture was replaced with hydrogen and reacted at room temperature for 12 hours. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound of this step (1.9 g, yield: 89%).
MS m/z(ESI):168.1[M+H] +
The third step: (E) Preparation of methyl (E) -2-cyano-2-cyclopentyl-2- ((4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) azo) acetate
4-amino-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (300mg, 1.01mmol) was dissolved in 1N hydrochloric acid (1 mL) at room temperature, 1M aqueous solution of sodium nitrite (84.76mg, 1.22mmol) was slowly dropped thereinto, after 1 hour of reaction at room temperature, the reaction solution was cooled to 0 ℃ and slowly dropped thereinto a solution of methyl 2-cyano-2-cyclopentylacetate (196.20mg, 1.111mmol) in ethanol (2 mL) and water (1 mL), and potassium acetate was added thereto in portions to keep the reaction system neutral, and after completion of addition, the reaction was carried out at 0 ℃ for 2 hours. After the completion of the reaction, a saturated ammonium chloride solution was added, extraction was performed with ethyl acetate, the combined organic layers were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =3/1 (v/v)) to obtain the title compound of the present step (374.2 mg, yield: 76%).
MS m/z(ESI):460.2[M+H] +
The fourth step: (Z) -N- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) cyclopentane carbohydrazone cyanide
Methyl (E) -2-cyano-2-cyclopentyl-2- ((4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) azo) acetate (350mg, 0.72mmol) was dissolved in tetrahydrofuran (10 mL), and 8N NaOH solution (731.05mg, 18.09mmol) was slowly added dropwise at 0 ℃ and reacted at room temperature for 1 hour. Saturated ammonium chloride solution was added, extraction was performed 4 times with ethyl acetate, and the organic phases were combined, washed successively with 10% aqueous citric acid solution, saturated sodium bicarbonate and saturated brine, dried over anhydrous magnesium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =4/1 (v/v)) to obtain the title compound of this step (250.3 mg, yield: 82%).
MS m/z(ESI):402.1[M+H] +
The fifth step: preparation of 4- (4-amino-5-cyano-3-cyclopentyl-1H-pyrazol-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
(Z) -N- (4- ((4- (trifluoromethyl) pyridin-2-yl) carbamoyl) phenyl) cyclopentanecarbonylhydrazone cyanide (180mg, 0.43mmol) was dissolved in t-butanol (8 mL), potassium t-butoxide (169.01mg, 1.49mmol) was added, stirring was carried out at room temperature for 15 minutes, 2-bromoacetonitrile (129.04mg, 1.07mmol) was added, and the reaction was carried out at room temperature for 3 hours. The reaction solution was concentrated to give the title compound of this step (100.2 mg, yield: 51%).
MS m/z(ESI):441.2[M+H] +
And a sixth step: preparation of 4- (7-amino-3-cyclopentyl-1H-pyrazolo [4,3-d ] pyrimidin-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-5-cyano-3-cyclopentyl-1H-pyrazol-1-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (85mg, 0.18mmol) was dissolved in methanol (6 mL), and formamidine acetate (192.79mg, 1.83mmol) was added and reacted at 70 ℃ for 10 hours. The crude product obtained by concentrating the reaction liquid was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate =2/1 (v/v)) to obtain the title compound of the present step (45.3 mg, yield: 50%).
MS m/z(ESI):468.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,1H),8.70(d,J=4.4Hz,1H),8.57(s,1H),8.47-8.14(m,3H),7.60(dd,J=33.2,6.0Hz,3H),6.77(s,2H),3.47(dd,J=24.4,16.0Hz,1H),2.04(dd,J=21.2,13.2Hz,4H),1.87-1.69(m,4H)。
Example 22: preparation of 4- (4-amino-7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 22)
Figure BDA0003222255240000621
The first step is as follows: preparation of 4- (4-amino-7- (3, 6-dihydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-7-bromo-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (500mg, 0.52mmol), 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester (173.76mg, 0.79mmol), and potassium carbonate (219.06mg, 1.57mmol) were added to 1, 4-dioxane (10 mL) and water (2 mL), nitrogen was replaced three times, and chlorine (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (41.58mg, 0.052mmol) was added and reacted at 95 ℃ for 6 hours. After the completion of the reaction, the crude product obtained by concentrating the reaction liquid was purified by silica gel column chromatography (eluent: dichloromethane/methanol =9/1 (v/v)) to obtain the title compound of the present step (180 mg, yield: 68%).
MS m/z(ESI):481.2[M+H] +
The second step is that: preparation of 4- (4-amino-7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-7- (3, 6-dihydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (180mg, 0.34mmol) was dissolved in methanol (8 mL), palladium on charcoal (40mg, 10%) was added, replaced with hydrogen, and reacted at room temperature for 5 hours. The reaction solution was filtered through celite, and the crude product obtained by concentrating the filtrate was purified by high performance liquid chromatography (preparation condition 2) to obtain the title compound of this step (25 mg, yield: 15%).
MS m/z(ESI):483.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.42(d,J=9.2Hz,1H),8.71(d,J=5.2Hz,1H),8.58(s,1H),8.29(dd,J=30.0,27.2Hz,3H),7.64(s,1H),7.58(d,J=8.4Hz,3H),6.08(s,2H),4.10-3.93(m,2H),3.62-3.48(m,2H),3.11(ddd,J=11.6,8.4,3.6Hz,1H),2.04-1.94(m,2H),1.82(qd,J=12.4,4.0Hz,2H)。
Example 23: preparation of 4- (4-amino-7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 23)
Figure BDA0003222255240000631
The first step is as follows: preparation of 4- (4-amino-7- (prop-1-en-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
Using isopropenylboronic acid pinacol ester instead of 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester of the first step in example 22, the title compound of this step was synthesized (270 mg, yield: 93%) in a similar manner to the procedure described in the first step of example 22.
MS m/z(ESI):439.1[M+H] +
The second step: preparation of 4- (4-amino-7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound (20 mg, yield: 8%) of this step was synthesized in analogy to the procedure described in the second step of example 22, with 4- (4-amino-7- (prop-1-en-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide replacing the second step of example 22 with 4- (4-amino-7- (3, 6-dihydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (4-amino-7- (prop-1-en-2-yl) -5H-pyrrol-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide.
MS m/z(ESI):441.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,1H),8.71(d,J=5.2Hz,1H),8.54(d,J=26.4Hz,1H),8.24(dd,J=20.8,12.4Hz,3H),7.58(dd,J=6.4,4.4Hz,4H),6.30-5.75(s,2H),3.19(td,J=13.6,6.4Hz,1H),1.36(d,J=6.8Hz,6H)。
Example 24: preparation of 4- (4-amino-7- (4-hydroxycyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 24)
Figure BDA0003222255240000641
The first step is as follows: preparation of 4- (4-amino-7- (1, 4-dioxaspiro [4.5] dec-7-en-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-7-bromo-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (2g, 2.51mmol), 1, 4-dioxaspiro [4,5] dec-7-ene-8-boronic acid pinacol ester (1.06g, 3.77mmol), and potassium carbonate (1.05g, 7.54mmol) were added to a mixed solvent of 1, 4-dioxane (8 mL) and water (2 mL), nitrogen was replaced, and chlorine (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (207.99mg, 0.25mmol) was added, and the reaction was carried out at 95 ℃ for 12 hours. After completion of the reaction, the reaction solution was concentrated to give a crude product, which was purified by silica gel column chromatography (eluent: dichloromethane/methanol =10/1 (v/v)), to give the title compound of the present step (618 mg, yield: 41%).
MS m/z(ESI):537.2[M+H] +
The second step is that: preparation of 4- (4-amino-7- (1, 4-dioxaspiro [4.5] decan-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
The title compound of the present step (500 mg, yield: 88%) was synthesized in analogy to the procedure described in the second step of example 22, using 4- (4-amino-7- (1, 4-dioxaspiro [4.5] dec-7-en-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide to replace the 4- (4-amino-7- (3, 6-dihydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide in the second step of example 22.
MS m/z(ESI):539.2[M+H] +
The third step: preparation of 4- (4-amino-7- (4-oxocyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-7- (1, 4-dioxaspiro [4.5] decan-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (450mg, 0.75mmol) was dissolved in dioxane hydrochloride (4M, 10 mL) and reacted at room temperature for 4 hours. The reaction mixture was concentrated to give the title compound of this step (105 mg, yield: 42%).
MS m/z(ESI):495.2[M+H] +
The fourth step: preparation of 4- (4-amino-7- (4-hydroxycyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide
4- (4-amino-7- (4-oxocyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (50mg, 0.09mmol) was dissolved in methanol (4 mL) and tetrahydrofuran (2 mL), and sodium borohydride (10.48mg, 0.27mmol) was added at 0 ℃ and reacted at 0 ℃ for 3 hours. After completion of the reaction, saturated ammonium chloride was added thereto for quenching, the reaction solution was concentrated, water and ethyl acetate were added thereto for extraction, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: dichloromethane/methanol =10/1 (v/v)) and further purified by preparative thin layer chromatography silica gel plate (developing solvent: dichloromethane/methanol =20/1 (v/v)) to obtain the title compound of the present step (15 mg, yield: 32%).
MS m/z(ESI):497.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.70(d,J=5.2Hz,1H),8.57(s,1H),8.24(d,J=7.8Hz,3H),7.56(t,J=7.2Hz,4H),6.00(s,2H),4.58(d,J=4.4Hz,1H),3.47(dd,J=9.6,5.2Hz,1H),2.77(t,J=12.0Hz,1H),2.04(t,J=13.2Hz,2H),1.93(d,J=9.6Hz,2H),1.57(dd,J=25.2,10.8Hz,2H),1.35-1.15(m,2H)。
Example 25: preparation of N- (4- (4-amino-7- (1, 1-trifluoropropan-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (Compound 25)
Figure BDA0003222255240000651
(4- ((5-fluoro-2-methoxybenzamide) methyl) phenyl) boronic acid (79.00mg, 0.25mmol) and 7- (1, 1-trifluoropropan-2-yl) -5H-pyrrolo [3, 2-d)]Dissolving 4-pyrimidineamine (50mg, 0.2mmol) in 1, 2-dichloroethane (5 mL), adding pyridine (32.97mg, 0.4 mmol),
Figure BDA0003222255240000652
Molecular sieves (10 mg) and copper acetate (75.70mg, 0.4 mmol) were reacted at 40 ℃ for 5 hours. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 2) to obtain the title compound (4 mg, yield: 4%) of this step.
MS m/z(ESI):488.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.96(s,1H),8.25(d,J=18.0Hz,1H),7.77(d,J=21.6Hz,1H),7.54(dd,J=24.4,7.6Hz,4H),7.37(d,J=4.8Hz,1H),7.23(dd,J=9.2,4.0Hz,1H),6.67(s,1H),5.87(s,1H),5.58(s,1H),4.63(d,J=5.6Hz,2H),4.16-3.99(m,1H),3.94(s,3H),1.64-1.43(m,3H)。
Example 26: preparation of 4- (4-amino-7- (1, 1-trifluoropropan-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) -N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (Compound 26)
Figure BDA0003222255240000661
4-bromo-N- (4- (trifluoromethyl) pyridin-2-yl) benzamide (224.89mg, 0.62mmol) and 7- (1, 1-trifluoropropan-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (100mg, 0.41mmol) were dissolved in N, N-dimethylformamide (2 mL), potassium iodide (138.40mg, 0.83mmol) was added, nitrogen was replaced, cuprous iodide (87.33mg, 0.45mmol) and cesium carbonate (407.48mg, 1.24mmol) were added, and the mixture was heated by microwave at 120 ℃ for 2 hours. Water and ethyl acetate were added for extraction, the organic phases were combined and the resulting crude product was concentrated and purified by high performance liquid chromatography (preparation method 2) to obtain the title compound of this step (4 mg, yield: 2%).
MS m/z(ESI):495.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,1H),8.72(d,J=5.2Hz,1H),8.58(s,1H),8.38-8.14(m,3H),7.94(s,1H),7.61(dd,J=16.4,6.8Hz,3H),6.13(s,2H),4.05(dd,J=16.8,8.0Hz,1H),1.58(d,J=7.2Hz,3H)。
Example 27: preparation of N- (4- (4-amino-7- (1- (2-hydroxyacetyl) piperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (Compound 27)
Figure BDA0003222255240000671
The first step is as follows: preparation of 5- (4-bromophenyl) -N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
Reacting N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d]Pyrimidine-4-amine (5.49g, 18.92mmol) and 4-bromobenzoic acid (8g, 37.84mmol) were dissolved in 1, 2-dichloroethane (80 mL), and copper acetate (6.94g, 37.84mmol) and,
Figure BDA0003222255240000672
Molecular sieve (2 g) and pyridine (3.02g, 37.84mmol), and reacting for 16 hours at 35 ℃ by oxygen substitution 3 times. The reaction solution was filtered, the filter cake was washed with dichloromethane, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: dichloromethane/methanol =20/1 (v/v)) to obtain the title compound of the present step (4.3 g, yield: 49%).
MS m/z(ESI):439.1[M+H] +
The second step is that: preparation of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
5- (4-bromophenyl) -N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (1.2g, 2.46mmol), ((5-fluoro-2-methoxybenzamide) methyl) potassium trifluoroborate (822.88mg, 2.70mmol) and potassium carbonate (1.03g, 7.38mmol) were added to 1, 4-dioxane (20 mL) and water (4 mL), replaced with nitrogen, chlorine (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (195.13mg, 0.25mmol) was added, and the mixture was heated by microwave at 90 ℃ for 1 hour. The reaction solution was concentrated, extracted with water and dichloromethane, the organic phases were combined, washed with water, and the crude product obtained by concentrating the organic phase was purified by silica gel column chromatography (eluent: dichloromethane/methanol =20/1 (v/v)) to obtain the title compound of this step (920 mg, yield: 66%).
MS m/z(ESI):542.2[M+H] +
The third step: preparation of N- (4- (4-amino-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (1.1 g, 1.97mmol) was added to trifluoroacetic acid (2 mL) and reacted at 70 ℃ for 12 hours. The reaction solution was concentrated, diluted with dichloromethane, washed with saturated aqueous sodium bicarbonate solution, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: dichloromethane/methanol =20/1 (v/v)) to obtain the title compound of the present step (560 mg, yield: 69%).
MS m/z(ESI):392.1[M+H] +
The fourth step: preparation of N- (4- (4-amino-7-bromo-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4- (4-amino-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (770 mg, 1.91mmol) was dissolved in methylene chloride (20 mL), and N-bromosuccinimide (343.07mg, 1.91mmol) was added and reacted at 25 ℃ for 2 hours. The reaction solution was diluted with methylene chloride, washed with a saturated aqueous solution of sodium sulfite, and the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound of the present step (700 mg, yield: 74%).
MS m/z(ESI):470.1[M+H] +
The fifth step: preparation of N- (4- (4-amino-7- (1- (2-hydroxyacetyl) -1,2,3, 6-tetrahydropyridin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4- (4-amino-7-bromo-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (50mg, 0.10 mmol), 2-hydroxy-1- (4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) ethan-1-one (42.60mg, 0.15mmol), and potassium carbonate (42.30mg, 0.30mmol) were added to 1, 2-dioxane (5 mL) and water (1 mL), nitrogen was replaced, chlorine (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (8.02010mg, 0.02010mmol) was added, and the mixture was heated at 95 ℃ for 3 hours. The reaction mixture was concentrated, and methylene chloride and methanol were added to dissolve the reaction mixture, followed by filtration to remove inorganic salts, and the filtrate was concentrated to give the title compound of this step (50 mg, yield: 65%).
MS m/z(ESI):531.2[M+H] +
And a sixth step: preparation of N- (4- (4-amino-7- (1- (2-hydroxyacetyl) piperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4- (4-amino-7- (1- (2-hydroxyacetyl) -1,2,3, 6-tetrahydropyridin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (50mg, 0.090mmol) was dissolved in methanol (5 mL), palladium on carbon (100mg, 10%) was added, replaced with hydrogen, and the reaction was carried out at room temperature for 5 hours. The reaction solution was filtered through Celite, and the crude product obtained by concentrating the filtrate was purified by high performance liquid chromatography (preparation method 2) to obtain the title compound (5 mg, yield: 10%) of the present step.
MS m/z(ESI):533.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ9.03(t,J=6.4Hz,1H),8.47(s,1H),8.42(s,1H),7.68(t,J=10.4Hz,2H),7.63(d,J=7.2Hz,2H),7.51-7.44(m,1H),7.37-7.29(m,1H),7.16(dd,J=9.2,4.4Hz,1H),4.61(t,J=6.0Hz,2H),4.32(s,1H),4.20(d,J=12.4Hz,1H),4.00-3.87(m,4H),3.76(d,J=15.2Hz,1H),3.61-3.44(m,4H),2.25-2.17(m,1H),2.04-1.93(m,2H),1.59-1.44(m,2H)。
Example 28: preparation of N- (4- (4-amino-7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (Compound 28)
Figure BDA0003222255240000691
The first step is as follows: preparation of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) -2-methylpropan-1-one
7-bromo-N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine (530.99mg, 1.32mmol), 2-methyl-1- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) propan-1-one (580mg, 1.97mmol) and potassium carbonate (551.04mg, 3.95mmol) were added to water (2 mL) and 1, 4-dioxane (6 mL), nitrogen was replaced, chlorine (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (104.44mg, 0.13mmol) was added, and reacted at 95 ℃ for 5 hours. The reaction solution was concentrated to obtain a residue, which was dissolved in methylene chloride and methanol, filtered to remove inorganic salts, and the filtrate was concentrated to obtain the title compound of the present step (130 mg, yield: 22%).
MS m/z(ESI):436.2[M+H] +
The second step: preparation of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) -2-methylpropan-1-one
1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) -2-methylpropan-1-one (152.63mg, 0.33mmol) was dissolved in methanol (4 mL), palladium on charcoal (40mg, 10%) was added, and the reaction was carried out under hydrogen at room temperature for 5 hours. The reaction mixture was filtered through Celite, and the filtrate was concentrated to give the title compound of this step (100 mg, yield: 88%).
MS m/z(ESI):438.2[M+H] +
The third step: preparation of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4-bromobenzyl) -5-fluoro-2-methoxybenzamide (250.20mg, 0.60mmol) and 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) -2-methylpropan-1-one (150mg, 0.31mmol) were dissolved in N, N-dimethylformamide (3 mL), potassium carbonate (129.23mg, 0.93mmol) was added, nitrogen substitution was performed, cuprous iodide (59.36mg, 0.31mmol) and trans-N, N' -dimethyl-1, 2-cyclohexanediamine (46.20mg, 0.31mmol) were added, and microwave heating was performed at 120 ℃ for 3 hours. The reaction mixture was diluted with water and ethyl acetate, filtered, the filtrate was extracted with ethyl acetate, the organic layers were combined, washed successively with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and the crude product obtained by concentrating the filtrate was purified by silica gel column chromatography (eluent: dichloromethane/methanol =15/1 (v/v)) to obtain the title compound of this step (80 mg, yield: 36%).
MS m/z(ESI):695.3[M+H] +
The fourth step: preparation of N- (4- (4-amino-7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (80mg, 0.092mmol) was dissolved in trifluoroacetic acid (8 mL) and reacted at 80 ℃ for 4 hours. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation condition 2) to obtain the title compound (25 mg, yield: 47%) of this step.
MS m/z(ESI):545.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.92(t,J=6.0Hz,1H),8.67(s,1H),7.92(s,1H),7.67-7.45(m,5H),7.36(td,J=8.8,3.2Hz,1H),7.20(dd,J=9.2,4.4Hz,1H),4.58(t,J=12.0Hz,3H),4.08(d,J=12.4Hz,1H),3.90(s,3H),3.12(d,J=11.6Hz,2H),2.91(dt,J=13.2,6.8Hz,1H),2.62(t,J=12.4Hz,1H),2.08-1.86(m,2H),1.68-1.41(m,2H),1.01(s,6H)。
Example 29: preparation of N- (4- (4-amino-7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (Compound 29)
Figure BDA0003222255240000711
The first step is as follows: preparation of 7- (3, 6-dihydro-2H-pyran-4-yl) -N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
Example 32 was repeated in analogy to 2-methyl-1- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) propan-1-one from the first step in example 28 by replacing the 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester with
The title compound (240 mg, yield: 88%) was synthesized according to the method described in the first step.
MS m/z(ESI):367.2[M+H] +
The second step is that: preparation of N- (2, 4-dimethoxybenzyl) -7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
The title compound of this step was synthesized in a similar manner to the procedure described in the second step of example 28 by substituting 7- (3, 6-dihydro-2H-pyran-4-yl) -N- (2, 4-dimethoxybenzyl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine for 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) -2-methylpropan-1-one of the second step of example 28 (200 mg, yield: 86%).
MS m/z(ESI):369.2[M+H] +
The third step: preparation of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound (120 mg, yield: 30%) of this step was synthesized in analogy to the procedure described in the third step of example 28, using N- (2, 4-dimethoxybenzyl) -7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) -2-methylpropan-1-amine instead of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-4-one in the third step of example 28.
MS m/z(ESI):626.3[M+H] +
The fourth step: preparation of N- (4- (4-amino-7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound (10 mg, yield: 11%) of this step was synthesized in a similar manner to the method described in the fourth step of example 28 by substituting N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (tetrahydro-2H-pyran-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide for N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide used in the fourth step of example 28.
MS m/z(ESI):476.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.90(t,J=6.0Hz,1H),8.20(s,1H),7.52(dd,J=9.2,3.2Hz,2H),7.48-7.40(m,2H),7.34(ddd,J=10.4,8.4,3.2Hz,1H),7.23-7.16(m,1H),5.73(s,2H),4.57(t,J=12.0Hz,2H),4.08-3.76(m,5H),3.53-3.42(m,2H),3.16-2.99(m,1H),1.93(d,J=12.8Hz,2H),1.79(ddd,J=24.8,12.4,4.4Hz,2H)。
Example 30: preparation of N- (4- (7- (1-acetylpiperidin-4-yl) -4-amino-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (Compound 30)
Figure BDA0003222255240000721
The first step is as follows: preparation of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) ethan-1-one
The title compound of this step (400 mg, yield: 85%) was synthesized in a similar manner to the procedure described in the first step of example 28 by substituting 1-acetyl-5, 6-dihydro-2H-pyridine-4-boronic acid pinacol ester for 2-methyl-1- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) propan-1-one of the first step of example 28.
MS m/z(ESI):408.2[M+H] +
The second step: preparation of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) ethan-1-one
The title compound (320 mg, yield: 80%) of this step was synthesized in analogy to the method described in the second step of example 28 by substituting 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) ethan-1-one for 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) -2-methylpropan-1-one of the second step of example 28.
MS m/z(ESI):410.2[M+H] +
The third step: preparation of N- (4- (7- (1-acetylpiperidin-4-yl) -4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound of this step was synthesized in analogy to the procedure described in the third step of example 28 by using 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) ethan-1-one instead of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-one in the third step of example 28 (120 mg, yield: 25%).
MS m/z(ESI):667.3[M+H] +
The fourth step: preparation of N- (4- (7- (1-acetylpiperidin-4-yl) -4-amino-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound of the present step was synthesized in a similar manner to the procedure described in the fourth step of example 28 using N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide instead of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide used in the fourth step of example 28 (10 mg, yield: 16%).
MS m/z(ESI):517.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.90(s,1H),8.20(s,1H),7.52(dd,J=8.8,4.4Hz,3H),7.47(s,1H),7.43(d,J=8.4Hz,2H),7.38-7.32(m,1H),7.19(dd,J=9.2,4.4Hz,1H),5.74(s,2H),4.59(d,J=6.0Hz,2H),4.46(t,J=15.6Hz,1H),3.91(d,J=5.6Hz,4H),3.17(s,2H),2.66(d,J=2.4Hz,1H),1.99(d,J=21.6Hz,5H),1.78-1.51(m,2H)。
Example 31: preparation of N- (4- (4-amino-7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (compound 31)
Figure BDA0003222255240000741
The first step is as follows: preparation of N- (2, 4-dimethoxybenzyl) -7- (prop-1-en-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
Using isopropenylboronic acid pinacol ester instead of 2-methyl-1- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) propan-1-one of the first step in example 28, the title compound of this step (300 mg, yield: 84%) was synthesized in a similar manner to the procedure described in the first step of example 28.
MS m/z(ESI):325.2[M+H] +
The second step is that: preparation of N- (2, 4-dimethoxybenzyl) -7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-4-amine
The title compound of this step (300 mg, yield: 93%) was synthesized in analogy to the procedure described in the second step of example 28, using N- (2, 4-dimethoxybenzyl) -7- (prop-1-en-2-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine instead of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) -2-methylpropan-1-one of the second step of example 28.
MS m/z(ESI):327.2[M+H] +
The third step: preparation of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound (120 mg, yield: 18%) of this step was synthesized in a similar manner to the procedure described in the third step of example 28, substituting N- (2, 4-dimethoxybenzyl) -7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-4-amine for 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) -2-methylpropan-1-one of the third step of example 28.
MS m/z(ESI):584.3[M+H] +
The fourth step: preparation of N- (4- (4-amino-7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound (6 mg, yield: 11%) of this step was synthesized in analogy to the procedure described in the fourth step of example 28 by substituting N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7-isopropyl-5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide for N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide used in the fourth step of example 28.
MS m/z(ESI):434.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.91(s,1H),8.21(s,1H),7.54-7.48(m,3H),7.46-7.40(m,3H),7.39-7.30(m,1H),7.19(dd,J=9.2,4.4Hz,1H),5.73(s,2H),4.59(d,J=6.0Hz,2H),3.90(d,J=4.8Hz,3H),3.23-3.12(m,1H),1.33(d,J=6.8Hz,6H)。
Example 32: preparation of N- (4- (4-amino-7- (4-hydroxycyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (compound 32)
Figure BDA0003222255240000751
The first step is as follows: preparation of N- (2, 4-dimethoxybenzyl) -7- (1, 4-dioxaspiro [4.5] dec-7-en-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
The title compound of this step (600 mg, yield: 69%) was synthesized in analogy to the procedure described in the first step of example 28, by substituting 1, 4-dioxaspiro [4,5] dec-7-ene-8-boronic acid pinacol ester for 2-methyl-1- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridin-1 (2H) -yl) propan-1-one of the first step in example 28.
MS m/z(ESI):423.2[M+H] +
The second step is that: preparation of N- (2, 4-dimethoxybenzyl) -7- (1, 4-dioxaspiro [4.5] decan-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
The title compound (600 mg, yield: 71%) of this step was synthesized in a similar manner to the procedure described in the second step of example 28, using N- (2, 4-dimethoxybenzyl) -7- (1, 4-dioxaspiro [4.5] dec-7-en-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine instead of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -3, 6-dihydropyridin-1 (2H) -yl) -2-methylpropan-1-one of the second step of example 28.
MS m/z(ESI):425.2[M+H] +
The third step: preparation of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1, 4-dioxaspiro [4.5] decan-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound of this step (60 mg, yield: 19%) was synthesized in analogy to the procedure described in the third step of example 28, using N- (2, 4-dimethoxybenzyl) -7- (1, 4-dioxaspiro [4.5] decan-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine instead of 1- (4- (4- ((2, 4-dimethoxybenzyl) amino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-one in the third step of example 28.
MS m/z(ESI):682.3[M+H] +
The fourth step: preparation of N- (4- (4-amino-7- (4-oxocyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
The title compound of this step was synthesized in a similar manner to the procedure described in the fourth step of example 28 using N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1, 4-dioxaspiro [4.5] decan-8-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide in place of N- (4- (4- ((2, 4-dimethoxybenzyl) amino) -7- (1-isobutyrylpiperidin-4-yl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide of the fourth step in example 28 (40 mg, yield: 68%).
MS m/z(ESI):488.2[M+H] +
The fifth step: preparation of N- (4- (4-amino-7- (4-hydroxycyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide
N- (4- (4-amino-7- (4-oxocyclohexyl) -5H-pyrrolo [3,2-d ] pyrimidin-5-yl) benzyl) -5-fluoro-2-methoxybenzamide (80mg, 0.15mmol) was dissolved in methanol (6 mL), and sodium borohydride (11.34mg, 0.30mmol) was added at 0 ℃ to react at 0 ℃ for 3 hours. The crude product obtained by concentrating the reaction solution was purified by high performance liquid chromatography (preparation method 2) to obtain the title compound (10 mg, yield: 13%) of this step.
MS m/z(ESI):490.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.91(t,J=6.0Hz,1H),8.63(s,1H),7.81(s,1H),7.51(ddd,J=19.2,13.6,8.4Hz,5H),7.36(ddd,J=9.2,7.6,3.2Hz,1H),7.20(dd,J=9.2,4.4Hz,1H),4.60(d,J=6.0Hz,2H),3.90(d,J=3.2Hz,3H),2.78(t,J=12.0Hz,1H),1.94(d,J=4.4Hz,4H),1.50(dd,J=23.2,12.8Hz,2H),1.31(dd,J=23.6,9.6Hz,2H)。
Biological examples
Experimental example 1: compound testing for BTK _ C481S kinase activity
An experimental system:
enzyme: BTK _ C481S (SignalChem)
HTRF kit: HTRF KinEASE TM -TK(Cisbio Catalog)
Experimental parameters:
enzyme (BTK _ C481S) concentration: 2nM;
substrate (TK-Substrate-Biotin) concentration: 1 mu M;
Streptavidin-XL665 concentration: 0.0625. Mu.M;
TK Antibody-Cryptate concentration: 1X;
ATP:15μM
reaction buffer: 50mM HEPES, 5mM MgCl 2 、1mM DTT、50mM SEB
Compound and enzyme pre-incubation time: room temperature for 20 minutes
Reaction time: room temperature 90 minutes
Detection time: room temperature for 60 minutes
Parameters of the microplate reader: BMG PHERAStar HTRF
The experimental method comprises the following steps:
the BTK enzyme is respectively preincubated with test compounds (100 nM, 30nM and 10 nM) with different concentrations for 20min at 25 ℃, added with TK-substrate-biotin/ATP mixed working solution, incubated for reaction for 90min at 25 ℃, finally added with the mixed working solution of 2 XSstrevidin-XL 665 and TK Antibody-Cryptate, incubated for 60min at 25 ℃ and detected by a microplate reader for fluorescence signal Ratio (Ratio).
Data processing:
and (3) calculating the percentage inhibition rate of the compounds with different concentrations according to the following formula by taking a solvent group (DMSO) as a negative control and taking an enzyme reaction buffer solution group as a blank control:
percent inhibition = (1- (different concentrations of compound Ratio-blank Ratio)/(negative control Ratio-blank Ratio)) × 100%;
when the percent inhibition is between 30-80%, the half Inhibitory Concentration (IC) of the compound is calculated according to the following formula 50 ) Or the range:
IC 50 x = X (1-percent inhibition/percent inhibition, where X is the tested concentration of compound at which the inhibition is between 30-80%.
The experimental results are as follows:
the inhibitory effect of the compound on BTK _ C481S was determined according to the above method, and the results are shown in table 1.
TABLE 1 inhibitory Activity of Compounds of the present invention against BTK _ C481S
Compound numbering BTK-C481S(IC 50 ,nM)
1 7.2
2 16.0
3 19.4
6 90.6
7 30.7
8 15.4
9 20.2
10 26.1
11 11.0
12 47.1
13 41.8
14 8.5
15 16.3
16 18.3
17 15.2
18 10.8
20 2.1
21 13.3
22 9.4
23 22.0
24 2.7
25 25.1
26 13.5
28 8.6
29 5.7
30 3.7
31 4.6
32 3.0
And (4) conclusion: in the BTK _ C481S Kinase Assay, the compounds of the invention show strong inhibitory activity.
Experimental example 2: test for inhibition of proliferation Activity of Compounds on REC-1 cells
An experimental system:
cell name/manufacturer: REC-1/ATCC
Kit name/manufacturer:
Figure BDA0003222255240000791
Luminescent Cell Viability Assay,Promega
experimental parameters:
cell number: 10000 cells/well
Culture medium: RPMI-1640+10% of FBS +1%
Content of DMSO: 0.25 percent
Compound incubation conditions: 37 ℃,5% of CO 2
Incubation time: 96h
Detecting the temperature: RT (reverse transcription)
A detection instrument: thermo Varioskan Flash
The experimental steps are as follows:
in vitro monolayer culture of REC-1 cells in RPMI-1640 medium at 10% FBS +1% P/S at 37 ℃ with a content of 5% CO 2 Air incubator. Cells in logarithmic growth phase were plated in 96-well plates at 10000/well, and pre-diluted compound was added, negative control group of cells with compound in DMSO concentration, blank control group of medium without cells, at 37 ℃ and 5% CO 2 After incubation in an air incubator for 96h, the detection reagent CellTiter-Glo was added to each well and the relative chemiluminescent unit value (RLU) was read in the microplate reader chemiluminescent detection mode.
Data processing:
the percent inhibition of the compounds at different concentrations was calculated according to the following formula:
percent inhibition = (1- (chemiluminescence of test compound)Number-blank chemiluminescent signal value)/(negative-blank chemiluminescent signal value)). Times.100%, half maximal Inhibitory Concentration (IC) of the compound was calculated using Graphpad 8.3 according to a four parameter model fitting curve 50 )。
The experimental results are as follows:
the proliferation inhibitory activity of the compounds on REC-1 cells was determined according to the above method, and the results are shown in table 2.
TABLE 2 inhibition of the proliferative Activity of the Compounds of the invention on REC-1 cells
Figure BDA0003222255240000792
Figure BDA0003222255240000801
And (4) conclusion: the compound of the invention has stronger proliferation inhibition activity on REC-1 cells.
Experimental example 3: anti-IgM stimulated BTK phosphorylation inhibition assay
An experimental system:
cell name/manufacturer: ramos/ATCC
Kit name/manufacturer:
Figure BDA0003222255240000802
experimental parameters:
cell number: 1X 10 6 Cells/wells
Culture medium: RPMI-1640+10% of FBS +1%
Content of DMSO: 0.25 percent
Compound incubation conditions: 37 ℃,5% of CO2
Incubation time: 2h
Detecting the temperature: RT (reverse transcription)
A detection instrument: BIO-RAD Universal Hood II
The experimental steps are as follows:
in vitro monolayer culture of Ramos cells in RPMI-1640 medium at 10% FBS +1% P/S at 37 ℃ with 5% CO 2 Air incubator. Inoculating cells in logarithmic growth phase into a sterile 24-well plate with a density of 1 × 10 cells per well 6 The cells were then treated with prediluted compound, the blank and anti-IgM control were treated with compound DMSO at the same concentration, at 37 ℃ and 5% CO 2 After 2h incubation in an air incubator, adding anti-IgM with a final concentration of 20 mug/mL into each hole except a blank group, carrying out centrifugation and cell collection after 30 min treatment, adding 100 mug cell lysate to the cells, carrying out cell lysis on ice, shaking once every 5 min, carrying out cell lysis for 15 min, and then placing an EP tube into a precooling 4 ℃ centrifuge to centrifuge at 12000rpm for 10min. After centrifugation, the supernatant was transferred to a new 1.5mL EP tube, 5. Mu.L of the supernatant was quantitated using the BCA kit, and after quantitation, the protein concentration of the other supernatants was diluted to the minimum protein concentration using cell lysis buffer, and finally, 10. Mu.L of 5 Xloading buffer was added to 40. Mu.L of each tube, and heated in a 99 ℃ metal bath for 10min.
NuPAGE Bis-Tris protein pre-gel 10. Mu.L of sample was added to each well and run at 170V for 40 min. Transferring the PVDF membrane to a PVDF membrane at 300mA for 100 minutes, sealing with 5% skimmed milk powder for 1 hour, cutting the PVDF membrane according to a marker, correspondingly placing the cut PVDF membrane in Phospho-Btk (Tyr 223) (D9T 6H) rabbit monoclonal antibody, btk (D3H 5) rabbit monoclonal antibody and GAPDH (D16H 11) rabbit monoclonal antibody which are prepared by 5% BSA, incubating overnight in a refrigerator shaker at 4 ℃, taking out the PVDF membrane, cleaning with TBST, placing in anti-rabbit IgG prepared with 5% skimmed milk powder, incubating in the HRP antibody for 1 hour at room temperature, taking out and cleaning with TBST; and uniformly adding the developing solution on the surface of the membrane, and placing the membrane into a BIO-RAD Universal Hood II for developing and imaging.
Data processing:
the imaging picture is subjected to gray scale analysis by using image J, and the obtained gray scale value is used for calculating the percentage inhibition rate of the compounds with different concentrations according to the following formula: percent inhibition = (Anti-IgM control band grayscale value-compound group band grayscale value)/(Anti-IgM control band grayscale value-blank band grayscale value) × 100%.
The experimental results are as follows:
the compound was assayed for inhibition of BTK phosphorylation by Anti-IgM stimulated Ramos cells as described above, and the results are shown in Table 3.
TABLE 3 inhibition of Anti-IgM stimulated BTK phosphorylation in Ramos cells by Compounds of the invention
Figure BDA0003222255240000821
And (4) conclusion: the compound has stronger inhibition effect on Ramos cell BTK phosphorylation.
Although specific embodiments of the invention have been described in detail, it will be appreciated by those skilled in the art that, based upon the overall teachings of the disclosure, various modifications and alternatives to those details could be developed and still be encompassed by the present invention. The full scope of the invention is given by the appended claims and any equivalents thereof.

Claims (13)

1. A compound, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, said compound having the structure of formula (I), wherein:
Figure FDA0003222255230000011
x is selected from N and CH;
l is selected from-C (O) -NH-, -NH-C (O) -, -C 1-6 alkylene-NH-C (O) -and-C 1-6 alkylene-C (O) -NH-;
when X is N, L is not-C 1-6 alkylene-NH-C (O) -;
R 1 is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl, 3-12 membered heterocyclyl, C 6-10 Aryl and 5-6 membered heteroaryl, said alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl optionally substituted with one or more R 4 Substitution;
each R 2 Each independently selected from H, halogen, cyano、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl, -OR 5 and-NR 5 (R 6 );
R 3 Is selected from C 6-10 Aryl and 5-10 membered heteroaryl, said aryl and heteroaryl optionally substituted with one or more R 7 Substitution;
R 4 selected from H, halogen, nitro, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -C 1-6 alkyl-OH, -C 1-6 alkyl-O-C 1-6 Alkyl, -OR 8 、-SR 8 、-S(O)R 8 、-S(O) 2 R 8 、-NR 9 (R 10 )、-C(O)NR 9 (R 10 )、-C(O)R 11 、-S(O) 2 -NR 9 (R 10 )、C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted with one or more halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 5 and R 6 Each independently selected from H and C 1-6 An alkyl group; or,
R 5 and R 6 Together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl;
R 7 selected from H, halogen, cyano, nitro, oxo, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl, -OR 12 、-NR 12 (R 13 ) 3-12 membered heterocyclic group, C 6-10 Aryl and 5-10 membered heteroaryl;
R 8 selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted by one or more substituents selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 9 selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 10 selected from H and C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said cycloalkyl and heterocyclyl being optionally substituted by one or more substituents selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl; or,
R 9 、R 10 together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl which may optionally be substituted by one or more groups selected from oxo, halogen, hydroxy, amino, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl and-O-C 1-6 Alkyl substituent substitution;
R 11 is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more substituents selected from the group consisting of halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a haloalkyl;
R 12 is selected from C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-10 Cycloalkyl, 3-12 membered heterocyclyl, C 6-10 Aryl and 5-10 membered heteroaryl, said cycloalkyl, heterocyclyl, aryl and heteroaryl being optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a substituent of haloalkyl;
R 13 selected from H and C 1-6 An alkyl group;
m is 0, 1,2,3 or 4.
2. The compound of claim 1, wherein R 1 Is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl, 3-12 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from H, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -C 1-6 alkyl-OH, -C 1-6 alkyl-O-C 1-6 Alkyl, -OR 8 、-SR 8 、-S(O)R 8 、-S(O) 2 R 8 、-NR 9 (R 10 )、-C(O)NR 9 (R 10 )、-C(O)R 11 and-S (O) 2 -NR 9 (R 10 );
R 8 Selected from H, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 9 selected from H, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 10 selected from H and C 1-6 Alkyl and C 1-6 A haloalkyl group; or,
R 9 、R 10 together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl which may optionally be substituted by one or more groups selected from oxo, halogen, hydroxy, amino, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl and-O-C 1-6 Alkyl substituent;
R 11 is selected from C 1-6 Alkyl radical, C 3-10 Cycloalkyl and 3-12 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-6 Alkyl and C 1-6 Substituted with a substituent of haloalkyl;
preferably, R 1 Is selected from C 1-6 Alkyl radical, C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from H, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR 8 、-C(O)NR 9 (R 10 ) and-C (O) R 11
R 8 Selected from H, C 1-4 Alkyl and C 1-4 A haloalkyl group;
R 9 selected from H, C 1-4 Alkyl and C 1-4 A haloalkyl group;
R 10 is selected from H and C 1-4 Alkyl and C 1-4 A haloalkyl group;
R 11 is selected from C 1-6 Alkyl radical, C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, saidAlkyl, cycloalkyl and heterocyclyl are optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-4 Alkyl and C 1-4 Substituted with a haloalkyl;
preferably, R 1 Is selected from C 1-6 Alkyl radical, C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from H, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, hydroxy, -C (O) NR 9 (R 10 ) and-C (O) R 11
R 9 Is selected from C 1-4 An alkyl group;
R 10 is selected from C 1-4 An alkyl group;
R 11 is selected from C 1-6 Alkyl and C 3-6 Cycloalkyl, said alkyl and cycloalkyl being optionally substituted by one or more groups selected from halogen, hydroxy, cyano, C 1-4 Alkyl and C 1-4 Substituted with a haloalkyl;
preferably, R 1 Is selected from C 1-4 Alkyl radical, C 5-6 Cycloalkyl and 5-6 membered heterocyclyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted by one or more R 4 Substitution;
R 4 selected from fluorine, chlorine, C 1-6 Alkyl radical, C 1-6 Haloalkyl, hydroxy, -C (O) NR 9 (R 10 ) and-C (O) R 11
R 9 Is selected from C 1-4 An alkyl group;
R 10 is selected from C 1-4 An alkyl group;
R 11 is selected from C 1-4 Alkyl and C 3-6 Cycloalkyl, said alkyl, cycloalkyl and heterocyclyl being optionally substituted with one or more hydroxyl groups;
preferably, R 1 Selected from isopropyl group,
Figure FDA0003222255230000031
Cyclopentyl, cyclohexyl,
Figure FDA0003222255230000032
Tetrahydropyranyl, piperidinyl,
Figure FDA0003222255230000033
Figure FDA0003222255230000034
Preferably, the piperidine ring mentioned above
Figure FDA0003222255230000035
Ortho to the ring substituents;
preferably, the piperidine ring mentioned above
Figure FDA0003222255230000036
Meta to the substituent on the ring;
preferably, the piperidine ring mentioned above
Figure FDA0003222255230000037
Para to the ring substituents.
3. The compound of claim 1 or 2, wherein each R 2 Each independently selected from H and halogen;
preferably, each R is 2 Each independently selected from H, fluorine and chlorine;
preferably, R 2 Is H;
preferably, R 2 Is fluorine.
4. The compound of any one of claims 1-3, wherein R 3 Is selected from C 6-10 Aryl and 5-6 membered heteroaryl, said aryl and heteroaryl optionally substituted with one or more R 7 Substitution;
R 7 selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -OR 12 and-NR 12 (R 13 );
R 12 Is selected from C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 13 selected from H and C 1-6 An alkyl group;
preferably, R 3 Is selected from C 6-10 Aryl and 5-6 membered heteroaryl, said aryl and heteroaryl optionally substituted with one or more R 7 Substitution;
R 7 selected from halogen, C 1-6 Haloalkyl and-OR 12
R 12 Is selected from C 1-6 An alkyl group;
preferably, R 3 Selected from phenyl and pyridyl optionally substituted by one or more R 7 Substitution;
R 7 selected from fluorine, chlorine, C 1-4 Haloalkyl and-OR 12
R 12 Is selected from C 1-4 An alkyl group;
preferably, R 3 Selected from phenyl and pyridyl optionally substituted with one or more substituents selected from fluoro, trifluoromethyl and methoxy;
preferably, R 3 Is selected from
Figure FDA0003222255230000041
5. The compound of any one of claims 1-4, wherein X is N.
6. The compound of any one of claims 1-4, wherein X is CH.
7. The compound of claim 5, wherein L is selected from the group consisting of-C (O) -NH-, -NH-C (O) -and-C 1-6 alkylene-C (O) -NH-;
preferably, L is-C (O) -NH-;
preferably, -L-R 3 Is composed of
Figure FDA0003222255230000042
8. The compound of claim 6, wherein L is selected from the group consisting of-C (O) -NH-, -NH-C (O) -, -C 1-4 alkylene-NH-C (O) -and-C 1-4 alkyl-C (O) -NH-;
preferably, L is selected from the group consisting of-C (O) -NH-and-C 1-4 alkylene-NH-C (O) -;
preferably, -L-R 3 Is selected from
Figure FDA0003222255230000043
9. The compound of any one of claims 1-8, having the structure:
Figure FDA0003222255230000051
wherein R is 1 、R 2 、R 3 And m is as defined in any one of claims 1 to 8.
10. A compound according to any one of claims 1 to 9 selected from
Figure FDA0003222255230000052
Figure FDA0003222255230000061
11. A pharmaceutical composition comprising a compound of any one of claims 1-10, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, and optionally one or more excipients.
12. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition according to claim 11, for the manufacture of a medicament for the prevention and/or treatment of a disease mediated by BTK (bruton' S tyrosine kinase) wild type or BTK-C481S mutant.
13. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition according to claim 11, for the preparation of a BTK (bruton' S tyrosine kinase) wild-type or BTK-C481S mutant inhibitor.
CN202110961711.5A 2021-08-20 2021-08-20 Aromatic heterocyclic compound, preparation method and application thereof Pending CN115707705A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110961711.5A CN115707705A (en) 2021-08-20 2021-08-20 Aromatic heterocyclic compound, preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110961711.5A CN115707705A (en) 2021-08-20 2021-08-20 Aromatic heterocyclic compound, preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN115707705A true CN115707705A (en) 2023-02-21

Family

ID=85212715

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110961711.5A Pending CN115707705A (en) 2021-08-20 2021-08-20 Aromatic heterocyclic compound, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN115707705A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023165570A1 (en) * 2022-03-03 2023-09-07 深圳市塔吉瑞生物医药有限公司 Cycloalkyl or heterocyclyl substituted heteroaryl compound, and composition and use thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023165570A1 (en) * 2022-03-03 2023-09-07 深圳市塔吉瑞生物医药有限公司 Cycloalkyl or heterocyclyl substituted heteroaryl compound, and composition and use thereof

Similar Documents

Publication Publication Date Title
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN105732636B (en) Heteroaromatic compounds and their use in medicine
CN110627796B (en) Nitrogenous heterocyclic derivative and application thereof in medicine
AU2018208676A1 (en) Macrocyclic compounds as trk kinase inhibitors
CN115192577A (en) KRAS mutein inhibitors
CN110698481B (en) Heteroaryl pyridone and aza-pyridone amide compounds
CN105566321B (en) Heteroaromatic compounds and their use in medicine
CN105732637B (en) Heteroaromatic compounds and their use in medicine
EP3653625B1 (en) Fused ring derivative having mgat-2 inhibitory activity
CN102918043A (en) Pyrrolopyrimidine compounds as inhibitors of CDK4/6
JP6827959B2 (en) Their use as tricyclic compounds and phosphodiesterase inhibitors
WO2018170204A1 (en) 9,10,11,12-tetrahydro-8h-[1,4]diazepino[5&#39;,6&#39;:4,5]thieno[3,2-f]quinolin-8-one compounds and uses thereof
BR112021009610A2 (en) compound, pharmaceutical composition, use of a compound or composition, and, method of inhibiting necrosis, ferroptosis, human rip1 or related indications
AU2017319430A1 (en) N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
EP4129996A1 (en) Novel aminopyrimidine egfr inhibitor
KR102667331B1 (en) Aminopyridine derivatives and their use as selective alk-2 inhibitors
US20200071326A1 (en) Tam kinase inhibitors
WO2022217042A1 (en) Naphthyl-substituted quinoline-4(1h)-ones and related compounds and their use in treating medical conditions
CN112292374A (en) Novel phosphoinositide 3-kinase inhibitor and preparation method and application thereof
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
CN115466273A (en) Substituted alkynyl heterocyclic compounds
TW202227452A (en) Fused azaheterocyclic compounds, preparation methods, methods of inhibiting tyrosine kinase, pharmaceutical composition and uses thereof
CN115707705A (en) Aromatic heterocyclic compound, preparation method and application thereof
TW202115036A (en) Bridged heterocyclyl-substituted pyrimidines and their preparation methods and pharmaceutical uses
TW202346297A (en) Compounds with activity of anti-kras-mutated tumors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication