CN117043163A - Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof - Google Patents

Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof Download PDF

Info

Publication number
CN117043163A
CN117043163A CN202280022476.8A CN202280022476A CN117043163A CN 117043163 A CN117043163 A CN 117043163A CN 202280022476 A CN202280022476 A CN 202280022476A CN 117043163 A CN117043163 A CN 117043163A
Authority
CN
China
Prior art keywords
methyl
cancer
compound
alkyl
mmol
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
CN202280022476.8A
Other languages
Chinese (zh)
Inventor
张海生
卓鉥
代长贵
程辉敏
方磊
牛春意
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.)
Siegershenko Shenzhen Co ltd
Original Assignee
Siegershenko Shenzhen 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 Siegershenko Shenzhen Co ltd filed Critical Siegershenko Shenzhen Co ltd
Publication of CN117043163A publication Critical patent/CN117043163A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

Pyrrolo-pyrimidine or pyrrolo-pyridine derivatives and medical application thereof. Specifically, the compound has a structure shown in a formula I, has a good inhibition effect on Focal Adhesion Kinase (FAK) and simultaneously inhibits related signal paths, and can be used for preparing medicines for treating or preventing cancers, pulmonary hypertension, pathological angiogenesis and other related diseases, and especially for treating diseases caused by excessive or abnormal cell proliferation, such as tumors or cancers.

Description

Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof Technical Field
The invention belongs to the field of medicines, and particularly relates to pyrrolopyrimidine or pyrrolopyridine derivatives and medical application thereof, in particular to application in preparing medicines for treating or preventing cancers, pulmonary hypertension and pathological angiogenesis related diseases.
Background
Focal Adhesion Kinase (FAK), also known as PTK2 (protein tyrosine kinase 2), is a non-receptor tyrosine kinase located at the junction of multiple signaling pathways and is activated by integrins, growth factor receptors, G-protein coupled receptors, cytokines. In addition to being involved in signaling as a cytoplasmic kinase, related studies have also shown that FAK plays an important role in the nucleus. FAK can promote p53 degradation through ubiquitination, leading to cancer cell growth and proliferation. Tang et al reported that FAK can also modulate the expression of GATA4 and IL-33, thereby reducing inflammatory responses and immune evasion. In the microenvironment of tumors, intracellular FAK can regulate the formation of new blood vessels, affecting the blood supply of tumors.
FAK is widely expressed in vivo, plays an important role in cell growth, proliferation, migration and adhesion, and is involved in embryo development and occurrence and development of diseases (cancer, cardiovascular diseases and the like). Overexpression of FAK is found in many types of cancers, including colon cancer, breast cancer, prostate cancer, thyroid cancer, neuroblastoma, ovarian cancer, cervical cancer, brain cancer, head and neck cancer, liver cancerEsophageal cancer, pancreatic cancer, lung cancer, gastric cancer and acute leukemia. High expression of FAK often predicts a poor prognosis. For example, studies have found that GTPase RHOA Y42C Mutations are one of the most common function-acquired mutations among diffuse gastric cancers, while RHOA Y42C Mutant mice are sensitive to FAK inhibitors, suggesting that inhibition of FAK activity may be a new strategy for treating diffuse gastric cancer.
During FAK function, binding of transmembrane integrin receptors to the extracellular matrix (ECM) recruits FAK to sites of integrin aggregation. FAK does not interact directly with integrins, but rather binds to cell membranes and other adhesion proteins through its carboxy-terminal FAT domain. Once recruited, FAK in the inactive state activates its catalytic activity through autophosphorylation of Y397. After phosphorylation, FAK acts as a molecular scaffold, src family kinases can be recruited, src can phosphorylate the Y576 and Y577 sites of FAK, further enhancing FAK activity while facilitating recruitment of downstream SH2 domain containing proteins such as Grb2 and PI3K, and the like. SOS formation complexes can be further recruited upon binding of Grb2 to FAK to further activate the downstream Ras-MAPK signaling pathway.
Based on the above, FAK and its signaling pathway related targets are considered potential targets for anticancer drug development. While inhibitors against FAK are currently not marketed, only some have entered clinical stages, such as Defactinib, IN10018, GSK-2256098, etc. Therefore, the development of new compounds that modulate the FAK signaling pathway is of great importance.
Disclosure of Invention
The invention provides an Focal Adhesion Kinase (FAK) inhibitor compound, a preparation method thereof, a medicament or a composition containing the compound, and a method for treating diseases caused by excessive or abnormal cell proliferation, such as cancers by using the compound, so that the Focal Adhesion Kinase (FAK) inhibitor compound has good clinical application prospect. The compound disclosed by the invention has good inhibition effect on FAK kinase activity, also has the effect of reducing activated YAP, and simultaneously has better drug generation, drug effect and toxicological characteristics.
In a first aspect of the invention there is provided a compound of formula I, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof,
Wherein,
ring a is independently selected from: C3-C20 heteroaryl and C6-C20 aryl, such as 5-6 membered heteroaryl and phenyl;
the B ring is independently selected from: C3-C20 heteroaryl and C6-C20 aryl, such as 5-6 membered heteroaryl and phenyl; preferably, ring B is a six membered aryl or heteroaryl group;
l is independently selected from: bond or CH 2 Preferably, L is CH 2
X is independently selected from CH or N, preferably X is N;
R 1 independently selected from: halogen, -C (=O) N (CH) 3 ) 2 、-C(=O)NHCH 3 、-C(=O)NH 2 -CH (=o), -COOH, CN, C1-C6 alkyl, -CF 3 、-CHF 2 、-CH 2 F、-CO 2 CH 3
R 2 Independently selected from: -N (R) 5 )S(O) m R 6 、-P(=O)R' 5 R' 6 、-S(O) m NR 5 R 6 、-C(=O)NR 5 R 6 、-NR 5 C(=O)R 6 、-C(=O)R 5 、-C(=O)OR 5 、-OC(=O)R 5 、-S(O) m R 5
R 3 Each independently selected from: -H, -OH, -halogen, -CF 3 、-CHF 2 、-CH 2 F、-CN、-NO 2 、-NR 5 R 6 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3R 9 Substitution;
R 4 each independently selected from: H. -OR 5 Halogen, -CF 3 、-CHF 2 、-CH 2 F、-CN、-NO 2 、-NR 5 R 6 、-C(=O)NR 5 R 6 、-C(=O)NR 5 OR 6 、-C(R 5 )=NR 6 、-NR 5 C(=O)R 6 、-C(=O)R 5 、-C(=O)C(=O)R 5 、-C(=O)OR 5 、-OC(=O)R 5 、-OC(=O)OR 5 、-P(=O)R' 5 R' 6 、-S(=O)(=NR 5 )R 6 、-S(O) m R 5 、-NR 5 S(O) m R 6 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3R 9 Substitution;
or any two adjacent R 4 And C5-C7 cycloalkyl, 5-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, which together with the atoms to which they are attached; wherein the C5-C7 cycloalkyl, 5-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl may be substituted with 1-3R 9 Substitution;
R 5 、R 6 、R' 5 and R'. 6 Each independently selected from: H. -OH, halogen, -CF 3 、-CHF 2 、-CH 2 F、-CN、-NO 2 、-CH 2 CF 3 、-NR 7 R 8 、-S(O) m R 7 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; or at NR 7 R 8 Wherein R is 7 And R is 8 Together with the N atom to which it is attached form a 3-10 membered heterocyclyl (including bridged and spiro rings); wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3R 9 Substitution;
R 9 each independently selected from: H. -OH, oxo (=o), halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 、-OR 10 、-C(=O)R 10 、-OC(=O)R 10 、-OC(=O)R 10 、-OC(=O)OR 10 、-C(=O)NR 10 R 11 、-NR 10 C(=O)NR 11 R 12 、-NR 10 R 11 、-NR 10 C(=O)R 11 、-NR 10 S(O) m R 11 、-S(O) m R 10 、-S(O) m NR 10 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1 to 3 groups selected from the group consisting of: C1-C6 alkyl, halogen, -OH, -CN, -NO 2 、-CHF 2 、-CH 2 CF 3 、-CF 3 、-C(O)R 13 、-C(O)NR 13 R 14 、-S(O) m R 13 、-S(O) m NR 13 R 14 Substitution;
R 7 、R 8 、R 10 、R 11 、R 12 、R 13 and R is 14 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1 to 3 groups selected from the group consisting of: -OH, halogen, -CN, -NO 2 、-NH 2 、-CHF 2 、-CH 2 CF 3 、-CF 3 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, -C (O) - (C1-C6 alkoxy), C3-C12 cycloalkyl, 3-12 membered heterocycloalkyl, C1-C6 alkylamine;
n and n' are each independently selected from 0, 1, 2, 3 or 4;
m is independently selected from 0, 1 or 2.
In some preferred embodiments of the present invention,selected from:
wherein p is 0, 1 or 2;
R 4 definitions of (A) are as described above。
In some preferred embodiments, the compound has a structure represented by formula II or formula III
Wherein,
p is 0, 1 or 2;
R 1 、R 2 、R 3 、R 4 the definitions of B and n' are as described above.
In some preferred embodiments, the compound has a structure of formula IV or formula V
Wherein,
R 1 、R 2 、R 3 、R 4 the definitions of B and n' are as described above, R 5 And R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein said C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy.
Among the compounds of the present application, the compounds of formula IV and V have better FAK kinase inhibitory activity than the same class of molecules; compounds of formula III, R meta to the amide bond on the benzene ring in formula IV 4 Compounds of the formula V or compounds of the formula V which are alkoxy (C1-C6-alkoxy, in particular methoxy) can improve the phase relative to the same moleculeSelectivity for other kinases including, but not limited to, PYK 2.
In some preferred embodiments, the compound has one of the following structures:
wherein,
R 1 、R 2 、R 3 、R 4 definition of R is as above 5 And R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-C10 heteroaryl, R' is selected from C1-C6 alkyl; wherein said C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy.
In some preferred embodiments, R 1 Independently selected from: F. cl, -C (=O) NH 2 -CH (=o), -COOH, -CN, C1-C6 alkyl, -CF 3 、-CHF 2 、-CH 2 F。
In some preferred embodiments, the B ring is selected from: phenyl, pyridyl or pyrazinyl.
In some preferred embodiments, each R 4 Independently selected from: H. halogen, C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl, -C (O) NR 5 R 6 The method comprises the steps of carrying out a first treatment on the surface of the Wherein,R 5 and R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein said C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy.
In some preferred embodiments, R 1 Independently selected from: F. cl, -C (=O) NH 2 -CH (=o), -COOH, -CN, methyl, -CF 3 、-CHF 2 、-CH 2 F, performing the process; and/or
Each R is 4 Independently selected from: F. cl, C1-C6 alkoxy, C3-C6 cycloalkyl, 3-6 membered heterocyclyl, -C (=O) NH (C1-C6 alkyl), -C (=O) NH (C3-C6 cycloalkyl), -C (=O) NH (3-6 membered heterocyclyl); wherein the C1-C6 alkyl, C1-C6 alkoxy, C3-C6 cycloalkyl, 3-6 membered heterocyclyl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy; and/or
R 2 Independently selected from: -N (R) 5 )S(O) m R 6 、-P(=O)R' 5 R' 6 、-S(O) m R 5 The method comprises the steps of carrying out a first treatment on the surface of the Wherein R is 5 、R 6 、R' 5 And R'. 6 Each independently selected from: H. -OH, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl; wherein m is 1 or 2; and/or
R 3 Each independently selected from: s (O) 2 CH 3 、-NCH 3 S(O) 2 CH 3 、-NHS(O) 2 CH 3 、-P(=O)(CH 3 ) 2 、-P(=O)(OH) 2 The method comprises the steps of carrying out a first treatment on the surface of the And/or
Selected from:
in some preferred embodiments A, B, L, X, R 1 、R 2 、R 3 、R 4 N and n' are groups corresponding to each specific compound in the examples.
In some preferred embodiments, the compound has a structure of one of the following groups or is selected from the group consisting of:
in some preferred embodiments, the compound is a compound as shown in the examples.
In a second aspect of the invention, there is provided a pharmaceutical composition comprising a compound as described in the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof; and a pharmaceutically acceptable carrier, diluent or excipient; preferably, the pharmaceutical composition further comprises one or more selected from the group consisting of: chemotherapeutic agents, PD-1 inhibitors, PD-1 antibodies, PD-L1 inhibitors, PD-L1 antibodies, ALK inhibitors, PI3K inhibitors, BTK inhibitors, EGFR antibodies, VEGFR inhibitors, VEGFR antibodies, HDAC inhibitors, CDK inhibitors, MEK inhibitors, akt inhibitors, mTOR inhibitors, SHP2 inhibitors, KRAS G12C inhibitors, KRAS G12D inhibitors, KRAS G12V inhibitors, C-MET inhibitors, her2 antibodies, claudin18.2 antibodies.
In a third aspect, the present invention provides the use of a compound according to the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to the second aspect, for the manufacture of a medicament for the prevention or treatment of a disease associated with FAK; preferably, the FAK-associated disease is cancer, pulmonary hypertension, or pathological angiogenesis; more preferably, the cancer is selected from: skin cancer, bone cancer, glioma, breast cancer, adrenal cancer, bladder cancer, esophageal cancer, cancer of the head or neck, liver cancer, parathyroid cancer, penile cancer, small intestine cancer, thyroid cancer, urinary tract cancer, cervical cancer, endometrial cancer, fallopian tube cancer, renal pelvis cancer, vaginal cancer, vulvar cancer, chronic or acute leukemia, colon cancer, melanoma, hematological malignancy, hodgkin's lymphoma, lung cancer, lymphocytic lymphoma, central nervous system tumor (CNS), ovarian cancer, pancreatic cancer, pituitary adenoma, prostate cancer, soft tissue sarcoma, gastric cancer, uterine cancer.
In a fourth aspect of the invention, there is provided a process for the preparation of a compound as described in the first aspect comprising the steps of
s 4) in an inert solvent and in the presence of a catalyst, reacting the compound I-1 with the compound I-2 to obtain a compound shown in a formula I;
wherein X' is halogen;
preferably, the method further comprises the step of:
s 1) reacting compound I-3 with I-4 in the presence of a base (e.g., DIPEA) in an inert solvent (e.g., I-PrOH) to give a compound of formula I-5;
s 2) in an inert solvent (e.g. DMF), a catalyst (e.g. Pd (PPh) 3 ) 2 Cl 2 And CuI) and under alkaline (such as DIPEA) conditions, reacting the compound I-5 with I-6 to obtain a compound of formula I-7;
s 3) reacting the compound I-7 in the presence of an inert solvent (such as NMP) and a base (such as tBuOK) to obtain a compound of formula I-1;
in the method, in the process of the invention,
x ', X ", and X'" are each independently halogen (e.g., cl, br, I);
ring A, ring B, X, L, R 1 、R 2 、R 3 、R 4 The definition of n and n' is as described above.
In a fifth aspect of the invention, there is provided a method of treating a FAK-related disease, the method comprising administering to a subject identified or diagnosed as having a FAK-related disease a therapeutically effective amount of a compound according to the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to the second aspect.
In a sixth aspect of the invention, there is provided a method for inhibiting FAK kinase activity in a cell or a subject, the method comprising the step of contacting the cell or administering to the subject a compound of the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to the second aspect; preferably, the cell is a mammalian cell; preferably, the subject is a mammal; more preferably a human.
In a seventh aspect, the present invention provides a compound according to the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, oxynitride, metabolite, or a pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to the second aspect, for use in the manufacture of a medicament for the prevention or treatment of a disease associated with YAP, in particular a disease associated with YAP positive; preferably, the YAP-related disease is cancer; more preferably, the cancer is selected from: skin cancer, bone cancer, glioma, breast cancer, adrenal cancer, bladder cancer, esophageal cancer, cancer of the head or neck, liver cancer, parathyroid cancer, penile cancer, small intestine cancer, thyroid cancer, urinary tract cancer, cervical cancer, endometrial cancer, fallopian tube cancer, renal pelvis cancer, vaginal cancer, vulvar cancer, chronic or acute leukemia, colon cancer, melanoma, hematological malignancy, hodgkin's lymphoma, lung cancer, lymphocytic lymphoma, central nervous system tumor (CNS), ovarian cancer, pancreatic cancer, pituitary adenoma, prostate cancer, soft tissue sarcoma, gastric cancer, uterine cancer.
In an eighth aspect of the invention, there is provided the use of a compound as described in the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or a pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition as described in the second aspect, for the manufacture of a medicament for the prevention or treatment of a disease associated with FAK and YAP, in particular a disease associated with FAK and YAP positivity; preferably, the FAK and YAP related disease is cancer; more preferably, the cancer is selected from: skin cancer, bone cancer, glioma, breast cancer, adrenal cancer, bladder cancer, esophageal cancer, cancer of the head or neck, liver cancer, parathyroid cancer, penile cancer, small intestine cancer, thyroid cancer, urinary tract cancer, cervical cancer, endometrial cancer, fallopian tube cancer, renal pelvis cancer, vaginal cancer, vulvar cancer, chronic or acute leukemia, colon cancer, melanoma, hematological malignancy, hodgkin's lymphoma, lung cancer, lymphocytic lymphoma, central nervous system tumor (CNS), ovarian cancer, pancreatic cancer, pituitary adenoma, prostate cancer, soft tissue sarcoma, gastric cancer, uterine cancer.
In a ninth aspect of the invention there is provided a method of treating a YAP (particularly YAP positive) associated disease, the method comprising administering to a subject identified or diagnosed as having a YAP (particularly YAP positive) associated disease a therapeutically effective amount of a compound as described in the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, oxynitride, metabolite or a pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition as described in the second aspect.
In a tenth aspect of the invention, there is provided a method for reducing activated YAP in a cell or a subject, the method comprising the step of contacting the cell or administering to the subject a compound of the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to the second aspect; preferably, the cell is a mammalian cell; preferably, the subject is a mammal; more preferably a human.
In another aspect, there is provided a method of treating FAK and YAP related disorders, the method comprising administering to a subject identified or diagnosed as having FAK and YAP related disorders a therapeutically effective amount of a compound according to the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to the second aspect.
In another aspect, there is provided a method for inhibiting FAK kinase activity in a cell or subject, and reducing activated YAP in a cell or subject, the method comprising the step of contacting the cell or administering to the subject a compound as described in the first aspect, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, oxynitride, metabolite, or a pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, or a pharmaceutical composition as described in the second aspect; preferably, the cell is a mammalian cell; preferably, the subject is a mammal; more preferably a human.
It is understood that within the scope of the present invention, the above-described technical features of the present invention and technical features specifically described below (e.g., in the examples) may be combined with each other to constitute new or preferred technical solutions. And are limited to a space, and are not described in detail herein.
Drawings
Figure 1 shows a morphological comparison of normal organoid cells with diffuse gastric carcinoma organoid cells.
FIG. 2 shows the effect of Compound 4 (2.5. Mu.M cultured for 48 h) on proliferation and morphology of diffuse gastric cancer organoids.
FIG. 3 shows the effect of Compound 6 (2.5. Mu.M cultured for 48 h) on proliferation and morphology of diffuse gastric cancer organoids.
FIG. 4 shows the effect of Compound 8 (2.5. Mu.M culture for 48 h) on proliferation and morphology of diffuse gastric cancer organoids.
FIG. 5 shows the effect of Compound 9 (2.5. Mu.M cultured for 48 h) on proliferation and morphology of diffuse gastric cancer organoids.
Figure 6 shows the effect of compound 9 and Defactinib on diffuse gastric carcinoma organoid cell proliferation.
Fig. 7 shows the effect of compound 9 on diffuse gastric carcinoma organoid FAK and YAP protein activity.
Figure 8 shows the effect of compound 10 on diffuse gastric carcinoma organoid FAK and YAP protein activity.
Fig. 9 shows the effect of compounds 8 and 9 on FAK and YAP protein activity of human diffuse gastric cancer tumor cell lines.
Figure 10 shows the effect of Defactinib, IN10018 and compounds 24, 25, 26, 27, 28 on FAK and YAP protein activity of human diffuse gastric cancer tumor cell lines.
Figure 11 shows the effect of compound 9 and Defactinib on proliferation of human diffuse gastric cancer tumor cell lines.
Detailed Description
The present inventors have conducted extensive and intensive studies and have unexpectedly found a class of compounds having a superior FAK kinase inhibitory activity while simultaneously achieving YAP reduction in activation. In addition, the compounds have better pharmacodynamic/pharmacokinetic properties. On this basis, the present application has been completed.
Terminology
In the present application, unless otherwise indicated, terms used have the ordinary meanings known to those skilled in the art.
When substituents are described by conventional formulas written from left to right, the substituents also include chemically equivalent substituents obtained when writing formulas from right to left. For example, -CH 2 O-is equivalent to-OCH 2 -。
The term "alkyl" by itself or as part of another substituent refers to a straight or branched chain hydrocarbon radical having the indicated number of carbon atoms (i.e., C1-C6 refers to containing 1, 2, 3, 4, 5, or 6 carbon atoms). Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, and the like. In the present application, alkyl is also intended to include substituted alkyl groups, i.e. one or more positions in the alkyl group are substituted, in particular 1 to 4 substituents, and may be substituted at any position.
The term "alkoxy" refers to a straight or branched or cyclic alkyl group attached through an ether oxygen, the free valence of which results from the ether oxygen. Alkoxy is preferably C1-C6 alkoxy, more preferably C1-C3 alkoxy. Representative examples include (but are not limited to): methoxy, ethoxy, propoxy, isopropoxy, butoxy and the like.
The term "alkenyl" means a straight or branched hydrocarbon group containing one or more double bonds and having the indicated number of carbon atoms. For example, "C2-C6 alkenyl" refers to groups containing 2 to 6 carbon atoms. Alkenyl groups include, but are not limited to: ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, heptenyl, octenyl and the like.
The term "alkynyl" refers to a straight or branched hydrocarbon radical containing one or more triple bonds and having the indicated number of carbon atoms. For example, "C2-C6 alkynyl" refers to groups containing 2 to 6 carbon atoms. Alkynyl groups include, but are not limited to: ethynyl, propynyl, butynyl, and the like.
The term "cycloalkyl" is intended to include saturated monocyclic (e.g., C3-C8), bicyclic (e.g., C5-C12 fused bicyclic, C5-C12 membered spirobicyclic) or polycyclic cyclic alkyl groups, "C3-C6 cycloalkyl" refers to containing 3 to 6 carbon atoms, and "C3-C12 cycloalkyl" refers to containing 3 to 12 carbon atoms. Cycloalkyl is preferably C3-C12 cycloalkyl, more preferably C3-C6 cycloalkyl. Representative cycloalkyl groups of the present invention include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, Etc. It is understood that substituted or unsubstituted cycloalkyl groups, such as branched cycloalkyl groups (e.g., 1-methylcyclopropyl and 2-methylcyclopropyl), are included in the definition of "cycloalkyl".
The term "cycloalkenyl" refers to cycloalkyl groups as defined above and further containing 1 or more double bonds, including but not limited to cyclopentenyl, cyclohexenyl.
The term "heterocyclyl" generally refers to a stable monocyclic (e.g., 3-8 membered, i.e., 3-, 4-, 5-, 6-, 7-, or 8-membered) or bicyclic (e.g., 5-12-, 5-, 6-, 7-, 8-, 9-, 10-, 11-, or 12-membered) or polycyclic (e.g., 7-14-, 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered) heterocycle, including fused-, spiro-, and/or bridged ring structures, which are saturated, partially unsaturated, and which contain carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from N, O and S. The term also includes polycyclic groups formed by the fusion of a heterocyclic ring with an aromatic ring, such as a benzene ring. "heterocyclyl" may be substituted or unsubstituted. The nitrogen and sulfur heteroatoms as ring atoms may optionally be oxidized. The nitrogen atom is substituted or unsubstituted (i.e., N or NR, where R is H or another substituent if defined). The heterocycle may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. If the resulting compound is stable, the heterocyclyl groups described herein may be substituted on a carbon or nitrogen atom. The nitrogen in the heterocycle may optionally be quaternized. Preferably, when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to each other. Preferably, the total number of S and O atoms in the heterocycle is no greater than 1. The heterocyclic group may be attached to any heteroatom or carbon atom residue of a ring or ring system molecule. Examples of heterocyclyl groups include, but are not limited to: azetidinyl, pyrrolidinyl, oxetanyl, pyrazolinyl, imidazolinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, hexahydroazepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1, 3-dioxanyl, and tetrahydro-1, 1-dioxythiophene, and the like. Wherein the heterocyclic groups of the spiro ring, the condensed ring and the bridged ring are optionally connected with other groups through single bonds, or are further connected with other cycloalkyl groups, heterocyclic groups, aryl groups and heteroaryl groups through any two or more atoms on the ring in a parallel ring mode.
The term "aryl", alone or as part of a larger moiety such as "aralkyl", "aralkoxy" or "aryloxyalkyl", refers to a monocyclic, bicyclic or tricyclic ring system (preferably 6-10 membered aromatic rings) having a total of 5 to 15 ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. "aryl" may be substituted or unsubstituted. In certain embodiments of the present invention, "aryl" refers to an aromatic ring system including, but not limited to: phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl and tetrahydronaphthyl. The aryl group may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring. The fused aryl group may be attached to another group at a suitable position on the cycloalkyl ring or aromatic ring. The connection lines drawn from the ring system indicate that the bond may be attached to any suitable ring atom. Aryl groups may be optionally substituted or unsubstituted.
The term "heteroaryl" refers to a heteroaromatic system containing 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, nitrogen and sulfur. Heteroaryl is preferably a 5-to 10-membered ring, more preferably 5-or 6-membered ring, such as pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazolyl, tetrazolyl, and the like. "heteroaryl" groups may be substituted or unsubstituted.
The term "halogen" includes fluorine, chlorine, bromine and iodine.
Unless otherwise indicated, it is assumed that any heteroatom in an underfilling state has sufficient hydrogen atoms to complement its valence.
In the present invention, the term "substituted" means that one or more hydrogen atoms on a particular group are replaced with a particular substituent. The specific substituents are those described in the foregoing for each of the examples or are those found in each of the examples. Unless otherwise specified, a substituted group may have a substituent selected from a specific group at any substitutable site of the group, which may be the same or different at each position, i.e., each substituent is independent of the other. Those skilled in the art will appreciate that combinations of substituents contemplated by the present invention are those that are stable or chemically achievable. Typical substitutions include, but are not limited to, one or more of the following groups: such as hydrogen, deuterium, halogen (e.g., a single halogen substituent or a multiple halogen substituent, the latter such as trifluoromethyl or containing Cl) 3 Alkyl group), cyano group, nitro group, oxo groupSuch as =o), trifluoromethyl, trifluoromethoxy, cycloalkyl, alkenyl, alkynyl, heterocyclyl, aryl, heteroaryl, OR a 、SR a 、S(=O)R e 、S(=O) 2 R e 、P(=O) 2 R e 、S(=O) 2 OR e ,P(=O) 2 OR e 、NR b R c 、NR b S(=O) 2 R e 、NR b P(=O) 2 R e 、S(=O) 2 NR b R c 、P(=O) 2 NR b R c 、C(=O)OR d 、C(=O)R a 、C(=O)NR b R c 、OC(=O)R a 、OC(=O)NR b R c 、NR b C(=O)OR e 、NR d C(=O)NR b R c 、NR d S(=O) 2 NR b R c 、NR d P(=O) 2 NR b R c 、NR b C(=O)R a Or NR b P(=O) 2 R e Wherein R is a Can independently represent hydrogen, deuterium, alkyl, cycloalkyl, alkenyl, alkynyl, heterocyclyl, aryl or heteroaryl, R b 、R c And R is d Can independently represent hydrogen, deuterium, alkyl, cycloalkyl, heterocycle or aromatic ring, or R b And R is c Together with the N atom, may form a heterocyclic ring; r is R e Can independently represent hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, heterocyclyl, aryl or heteroaryl. Typical substituents described above, such as alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocyclyl, aryl or heteroaryl groups, may be optionally substituted. Such as (but not limited to): halogen, hydroxy, cyano, carboxyl (-COOH), C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-12 membered heterocyclyl, aryl, heteroarylAryl, C1-C8 aldehyde, C2-C10 acyl, C2-C10 ester, amino, C1-C6 alkoxy, C1-C10 sulfonyl, C1-C6 ureido, and the like.
Yes-associated protein (YAP) is a proto-oncoprotein which exists in the cytoplasm in an inactive form and when activated, it migrates to the nucleus and activates transcription of genes associated with cell division and apoptosis. YAP is one of the downstream regulatory proteins in the Hippo signaling pathway, which cooperates with the transcription coactivator TAZ to direct gene expression by controlling the TEAD transcription factor family. The Hippo signaling pathway is an evolutionarily conserved signaling pathway that plays a key role in organ development, epithelial homeostasis, tissue regeneration, wound healing and immunomodulation. Deregulation of the Hippo pathway and YAP/TAZ-TEAD activity has been associated with a variety of diseases, the most prominent of which is cancer.
In the present invention, "YAP positive" means that the YAP content in the nucleus reaches or exceeds a preset content.
In the present invention, "FAK positive" means that the intracellular phosphorylated FAK content reaches or exceeds a preset level.
Active ingredient
As used herein, the term "compound of the invention" or "active ingredient of the invention" is used interchangeably to refer to a compound of formula I, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, isotope or prodrug thereof.
In the present invention, the compound of formula I has the following structure:
therein, A, B, L, X, R 1 、R 2 、R 3 、R 4 The definition of n and n' is as described above.
Preferably, the compound has a structure represented by formula II or formula III
Wherein p, R 1 、R 2 、R 3 、R 4 The definitions of B and n' are as described above.
Preferably, the compound has a structure represented by formula IV or formula V
Wherein,
R 1 、R 2 、R 3 、R 4 the definitions of B and n' are as described above, R 5 And R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein said C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy.
Preferably, R 1 Independently selected from: F. cl, -C (=O) NH 2 -CH (=o), -COOH, CN, methyl, -CF 3 、-CHF 2 、-CH 2 F;
Ring B is a six membered aryl or heteroaryl group; preferably, the B ring is selected from: phenyl, pyridyl or pyrazinyl;
R 4 each independently selected from: H. halogen, C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl, -C (O) NR 5 R 6 The method comprises the steps of carrying out a first treatment on the surface of the Wherein R is 5 And R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl;
R 2 independently selected from: -N (R) 5 )S(O) m R 6 、-P(=O)R' 5 R' 6 、-S(O) m R 5
Wherein R is 5 、R 6 、R' 5 And R'. 6 Each independently selected from: H. OH, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl;
wherein m is 1 or 2;
preferably, R 3 Each independently selected from: s (O) 2 CH 3 、-NCH 3 S(O) 2 CH 3 、-NHS(O) 2 CH 3 、-P(=O)(CH 3 ) 2 、-P(=O)(OH) 2
Salts which may be formed with the compounds of the present invention are also within the scope of the present invention. Unless otherwise indicated, the compounds of the present invention are understood to include salts thereof. The term "salt" as used herein refers to salts formed with inorganic or organic acids and bases in the acid or base form. Furthermore, when the compound of the present invention contains a basic moiety, it includes, but is not limited to, pyridine or imidazole, and an acidic moiety, including, but not limited to, carboxylic acids, the possible formation of zwitterions ("inner salts") are included within the term "salts". Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, for example, in isolation or purification steps during the preparation process. The compounds of the invention may form salts, for example, by reacting compound I with an amount of, for example, an equivalent of, an acid or base, salting out in a medium, or lyophilizing in aqueous solution.
The compounds of the present invention contain basic fragments, including but not limited to amine or pyridine or imidazole rings, which may form salts with organic or inorganic acids. Typical acids that may be salified include acetates (e.g., with acetic acid or trihaloacetic acid, such as trifluoroacetic acid), adipates, alginates, ascorbates, aspartate, benzoate, benzenesulfonate, bisulfate, borate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, diglycolate, dodecyl sulfate, ethane sulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, caproate, hydrochloride, hydrobromide, hydroiodide, hydroxyethanesulfonate (e.g., 2-hydroxyethanesulfonate), lactate, maleate, mesylate, naphthalene sulfonate (e.g., 2-naphthalene sulfonate), nicotinate, nitrate, oxalate, pectate, persulfate, phenylpropionate (e.g., 3-phenylpropionate), phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate (e.g., formed with sulfuric acid), sulfonate, tartrate, thiocyanate, toluene sulfonate such as p-toluenesulfonate, dodecanoate, and the like.
Certain compounds of the present invention may contain acidic moieties, including but not limited to carboxylic acids, that may form salts with various organic or inorganic bases. Typical base-forming salts include ammonium salts, alkali metal salts such as sodium, lithium, potassium salts, alkaline earth metal salts such as calcium, magnesium salts, and salts with organic bases (e.g., organic amines), such as benzathine, dicyclohexylamine, hydrabamine (salts with N, N-bis (dehydroabietyl) ethylenediamine), N-methyl-D-glucamine, N-methyl-D-glucamide, t-butylamine, and salts with amino acids such as arginine, lysine, and the like. Basic nitrogen-containing groups can be combined with halide quaternary ammonium salts, such as small molecule alkyl halides (e.g., methyl, ethyl, propyl and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl and dipentyl sulfates), long chain halides (e.g., decyl, dodecyl, tetradecyl and tetradecyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenyl bromides), and the like.
Prodrugs and solvates of the compounds of the invention are also within the scope of coverage. The term "prodrug" as used herein refers to a compound that undergoes chemical conversion by metabolic or chemical processes to produce a compound, salt, or solvate of the invention when used in the treatment of a related disorder. The compounds of the present invention include solvates, such as hydrates.
The compounds, salts or solvates of the present invention, may exist in tautomeric forms (e.g., amides and imine ethers). All of these tautomers are part of the present invention.
Stereoisomers of all compounds (e.g., those having asymmetric carbon atoms which may be present as a result of various substitutions), including enantiomeric and diastereoisomeric forms thereof, are contemplated as falling within the scope of the present invention. The individual stereoisomers of the compounds of the invention may not be present simultaneously with the other isomers (e.g., having particular activity as one pure or substantially pure optical isomer), or may be mixtures, such as racemates, or mixtures with all or a portion of the other stereoisomers. The chiral center of the present invention has two configurations, S or R, defined by the International Association of theory and application chemistry (IUPAC) 1974. The racemic forms can be resolved by physical methods, such as fractional crystallization, or by separation of crystals by derivatization into diastereomers, or by chiral column chromatography. Individual optical isomers may be obtained from the racemates by suitable methods, including but not limited to conventional methods, such as salt formation with an optically active acid followed by recrystallization.
The compounds of the present invention are prepared, isolated and purified in sequence to give the compounds in an amount of 90% by weight or more, for example 95% or more and 99% or more ("very pure" compounds), as listed in the text description. Such "very pure" compounds of the invention are also included herein as part of the invention.
All configurational isomers of the compounds of the present invention are within the scope of coverage, whether in mixtures, pure or very pure form. The definition of compounds in the present invention includes both the cis (Z) and the trans (E) olefin isomers, as well as the cis and trans isomers of carbocycles and heterocycles.
Throughout the specification, groups and substituents may be selected to provide stable fragments and compounds.
Specific functional groups and chemical term definitions are described in detail below. For the purposes of the present invention, chemical elements are described in conjunction with Periodic Table of the Elements, CAS version, handbook of Chemistry and Physics,75 th Ed.. The definition of specific functional groups is also described herein. Furthermore, the basic principles of organic chemistry and specific functional groups and reactivities are described in "Organic Chemistry", thomas Sorrell, university Science Books, sausalato 1999, which is incorporated by reference in its entirety.
Certain compounds of the invention may exist in specific geometric or stereoisomeric forms. The present invention encompasses all compounds, including cis and trans isomers, R and S enantiomers, diastereomers, (D) isomers, (L) isomers, racemic mixtures, and other mixtures thereof. In addition, an asymmetric carbon atom may represent a substituent such as an alkyl group. All isomers and mixtures thereof are encompassed by the present invention.
According to the invention, the mixture of isomers may contain various isomer ratios. For example, in a mixture of only two isomers, there may be a combination of: all ratios of 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98:2, 99:1, or 100:0 isomers are within the scope of the invention. Similar ratios, as well as ratios for more complex mixtures of isomers, are within the scope of the present invention, as would be readily understood by one of ordinary skill in the art.
The present invention also includes isotopically-labeled compounds, equivalent to those disclosed herein as original compounds. In practice, however, it will often occur that one or more atoms are replaced by atoms of a different atomic weight or mass number than the one . Examples of isotopes that can be listed as compounds of the invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes, respectively, such as 2 H、 3 H、 13 C、 11 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F and F 36 Cl. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates thereof, wherein isotopes or other isotopic atoms containing such compounds are within the scope of the present invention. Certain isotopically-labeled compounds of the present invention, e.g 3 H and 14 radioisotopes of C are also useful in, among other things, tissue distribution experiments of drugs and substrates. Tritium, i.e. tritium 3 H and carbon-14, i.e 14 C, their preparation and detection are relatively easy. Is the first choice in isotopes. In addition, heavier isotopic substitutions such as deuterium, i.e 2 H may be preferred in some cases because of its good metabolic stability, which may be advantageous in certain therapies, such as increasing half-life or decreasing dosage in vivo. Isotopically-labeled compounds can be prepared by conventional methods by using readily available isotopically-labeled reagents in place of non-isotopically-labeled reagents using the protocols disclosed in the examples.
If one is to design the synthesis of a particular enantiomer of a compound of the invention, it may be prepared by asymmetric synthesis or by derivatization with chiral auxiliary, separating the resulting diastereomeric mixture and removing the chiral auxiliary to give the pure enantiomer. Alternatively, if the molecule contains a basic functional group, such as an amino acid, or an acidic functional group, such as a carboxyl group, diastereomeric salts can be formed therewith using an appropriate optically active acid or base, and then the resulting mixture can be separated by conventional means such as fractional crystallization or chromatography to give the pure enantiomer.
As described herein, the compounds of the present invention may be substituted with any number of substituents or functional groups to extend their inclusion. In general, the term "substituted", whether appearing before or after the term "optional", in the formulas of the present invention includes substituents, means that the specified structural substituent is substituted for the hydrogen radical. When multiple of a particular structure are substituted at a position with multiple particular substituents, the substituents may be the same or different at each position. The term "substitution" as used herein includes all permissible organic compound substitutions. In a broad sense, permissible substituents include acyclic, cyclic, branched, unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic organic compounds. In the present invention, the heteroatom nitrogen may have a hydrogen substituent or any of the permissible organic compounds described hereinabove to supplement the valence state thereof. Furthermore, the present invention is not intended to be limited in any way to allow substitution of organic compounds. The present invention recognizes that the combination of substituents and variable groups is very good in the treatment of diseases in the form of stable compounds. The term "stable" as used herein refers to a compound that is stable for a period of time sufficient to maintain structural integrity of the compound, preferably for a period of time sufficient to be effective, as used herein for the purposes described above.
Metabolites of the compounds and pharmaceutically acceptable salts thereof of the present application, as well as prodrugs that can be converted in vivo to structures of the compounds and pharmaceutically acceptable salts thereof of the present application are also encompassed by the claims of the present application.
Pharmaceutical compositions and methods of administration
The compounds of formula (I), or enantiomers, diastereomers, racemates, tautomers, stereoisomers, geometric isomers, nitrogen oxides, metabolites or pharmaceutically acceptable salts, hydrates, solvates, isotopes or prodrugs thereof, and pharmaceutical compositions comprising the compounds of formula (I), or enantiomers, diastereomers, racemates, tautomers, stereoisomers, geometric isomers, nitrogen oxides, metabolites or pharmaceutically acceptable salts, hydrates, solvates, isotopes or prodrugs thereof, are useful for the prevention and/or treatment of: cancer, pulmonary hypertension, pathologic angiogenesis.
The compounds of formula (I), or enantiomers, diastereomers, racemates, tautomers, stereoisomers, geometric isomers, nitroxides, metabolites or pharmaceutically acceptable salts, hydrates, solvates, isotopes or prodrugs thereof, may be used in combination with other known medicaments for treating or ameliorating similar conditions. When administered in combination, the mode and dosage of administration of the original drug may remain unchanged, while the compound of formula (I) is administered simultaneously or first or second; or preparing the compound of formula (I), or enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitrogen oxide, metabolite or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, and other medicaments into a single preparation, and simultaneously administering the single preparation. When the compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, oxynitride, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, is administered simultaneously with one or more other drugs, a pharmaceutical composition containing one or more known drugs simultaneously with the compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, oxynitride, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof may be preferably used. Drug combinations also include administration of a compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, with one or more other known drugs, over an overlapping period of time. When a compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, is used in combination with one or more other drugs, the dosage of the compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, may be lower than the dosage of the known drugs alone.
Drugs or active ingredients that may be used in combination with the compounds of formula (I), or enantiomers, diastereomers, racemates, tautomers, stereoisomers, geometric isomers, nitrogen oxides, metabolites, or pharmaceutically acceptable salts, hydrates, solvates, isotopes or prodrugs thereof, include, but are not limited to: chemotherapeutic agents, PD-1 inhibitors, PD-1 antibodies, PD-L1 inhibitors, PD-L1 antibodies, ALK inhibitors, PI3K inhibitors, BTK inhibitors, EGFR antibodies, VEGFR inhibitors, VEGFR antibodies, HDAC inhibitors, CDK inhibitors, MEK inhibitors, akt inhibitors, mTOR inhibitors, SHP2 inhibitors, KRAS G12C inhibitors, KRAS G12D inhibitors, KRAS G12V inhibitors, C-MET inhibitors, her2 antibodies, claudin18.2 antibodies.
The compounds of formula (I), or enantiomers, diastereomers, racemates, tautomers, stereoisomers, geometric isomers, nitroxides, metabolites or pharmaceutically acceptable salts, hydrates, solvates, isotopes or prodrugs thereof, may also be used in combination with therapeutic agents for the treatment of pulmonary arterial hypertension (PHA). The PHA therapeutic is preferably a vasodilator, such as epoprostenol, tadalafil, ambrisentan, or the like.
Dosage forms of the pharmaceutical composition of the present invention include (but are not limited to): injection, tablet, capsule, aerosol, suppository, pellicle, dripping pill, external liniment, controlled release or sustained release preparation, or nanometer preparation.
The pharmaceutical compositions of the present invention comprise a safe and effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical compositions contain 1-2000mg of the compound of the invention per dose, more preferably 10-1000mg of the compound of the invention per dose. Preferably, the "one dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" means: one or more compatible solid or liquid filler or gel materials which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. "compatible" as used herein means that the components of the composition are capable of blending with and between the compounds of the present invention without significantly reducing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g. stearic acid, magnesium stearate), calcium sulphate, vegetable oils (e.g. soya oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g. propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (e.g. tween ) Wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizing agents, antioxidants, preservatives, pyrogen-free water and the like.
"excipient" refers to an adjunct to the drug formulation other than the primary drug, and may also be referred to as an adjuvant.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) Fillers or compatibilizers, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, e.g., glycerin; (d) Disintegrants, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) a slow solvent, such as paraffin; (f) an absorption accelerator, e.g., a quaternary amine compound; (g) Wetting agents, such as cetyl alcohol and glycerol monostearate; (h) an adsorbent, for example, kaolin; and (i) a lubricant, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared with coatings and shells, such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be released in a delayed manner in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxes. The active compound may also be in the form of microcapsules with one or more of the above excipients, if desired.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compound, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of these substances and the like.
In addition to these inert diluents, the compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar-agar or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms of the compounds of the present invention for topical administration include ointments, powders, patches, sprays and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The methods of treatment of the present invention may be administered alone or in combination with other therapeutic means or therapeutic agents.
When a pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is applied to a mammal (e.g., a human) in need of treatment, wherein the dose at the time of administration is a pharmaceutically effective dose, and the daily dose is usually 1 to 2000mg, preferably 50 to 1000mg, for a human having a body weight of 60 kg. Of course, the particular dosage should also take into account factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled practitioner.
The invention also provides a preparation method of the pharmaceutical composition, which comprises the following steps: a pharmaceutically acceptable carrier is admixed with a compound of formula (I) or a crystalline form, pharmaceutically acceptable salt, hydrate or solvate thereof according to the invention, thereby forming a pharmaceutical composition.
Preparation method
The following schemes and examples describe methods for preparing compounds of formula I. The starting materials and intermediates are purchased from commercial sources, prepared by known procedures, or otherwise described. In some cases, the order of the steps of the reaction scheme may be altered to promote the reaction or to avoid unwanted side reaction products.
The process for preparing the compound of the formula (I) according to the present invention is described in more detail below, but these specific processes do not limit the present invention in any way. The compounds of the present invention may also optionally be conveniently prepared by combining the various synthetic methods described in this specification or known in the art, such combination being readily apparent to those skilled in the art to which the present invention pertains.
Typically, in the preparation scheme, each reaction is carried out in a suitable solvent, typically under inert gas, at 0 to 90℃for a period of typically 2 to 24 hours.
Preferably, the compounds of the present invention are prepared by the following method
s 1) reacting compound I-3 with I-4 in the presence of a base (e.g., DIPEA) in an inert solvent (e.g., I-PrOH) to give a compound of formula I-5;
s 2) in an inert solvent (e.g. DMF), a catalyst (e.g. Pd (PPh) 3 ) 2 Cl 2 And CuI) and under alkaline (such as DIPEA) conditions, reacting the compound I-5 with I-6 to obtain a compound of formula I-7;
s 3) reacting the compound I-7 in the presence of an inert solvent (such as NMP) and a base (such as tBuOK) to obtain a compound of formula I-1;
s 4) reacting the compound I-1 with I-2 in an inert solvent (such as a catalyst) to obtain a compound of formula I;
in the method, in the process of the invention,
x ', X ", and X'" are each independently halogen (e.g., cl, br, I);
ring A, ring B, X, L, R 1 、R 2 、R 3 、R 4 The definition of n and n' is as described above.
It will also be appreciated by those skilled in the art that in the methods described herein, the intermediate compound functional groups may need to be protected by appropriate protecting groups. Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting groups is described in detail in Greene, t.w. and p.g.m. wuts, protective Groups in Organi Synthesis, (1999), 4th Ed. The protecting group may also be a polymeric resin.
The reagents or materials used in the present invention are commercially available or available in the manner set forth in the literature report.
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental methods, in which specific conditions are not noted in the following examples, are generally conducted under conventional conditions or under conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
The invention has the following main advantages:
(1) The compounds of the present invention have excellent inhibition ability on FAK kinase and/or can significantly reduce/decrease activated YAP; especially when two adjacent R's are on ring A 4 When the compound ring structure is formed with the ring A, the activity of the compound ring A is greatly improved; in addition, R 4 Is alkoxy or amido and R 2 In the case of N-methyl-sulfamide, the activity can also be improved obviously.
(2) The compound has lower toxic and side effects.
(3) The compound has better pharmacodynamics and pharmacokinetics.
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental procedure, which does not address the specific conditions in the examples below, is generally followed by routine conditions such as Sambrook et al, molecular cloning: conditions described in the laboratory Manual (New York: cold Spring Harbor Laboratory Press, 1989) or as recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred methods and materials described herein are presented for illustrative purposes only.
The structure of the compounds of the present invention is determined by Nuclear Magnetic Resonance (NMR) and liquid chromatography-mass spectrometry (LC-MS).
NMR is to makeThe solvent was determined to contain deuterated dimethyl sulfoxide (DMSO-d) as detected by Bruker AVANCE-400 and Bruker AVANCE-500 nuclear magnetic instruments 6 ) Deuterated acetone (CD) 3 COCD 3 ) Deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), etc., the internal standard being Tetramethylsilane (TMS), the chemical shifts being measured in parts per million (ppm).
Liquid chromatography (LC-MS) was performed using an Agilent 1260 mass spectrometer. HPLC was determined using an Agilent 1100 high pressure chromatograph (Microsorb 5 micron C18 100 x 3.0mm column).
The thin layer chromatography silica gel plate is Qingdao GF254 silica gel plate, TLC is 0.15-0.20mm, and the preparation thin layer chromatography is 0.4-0.5 mm. Column chromatography generally uses Qingdao silica gel 200-300 mesh silica gel as carrier.
The starting materials in the examples of the present invention are known and commercially available, or may be synthesized using or according to literature reported in the art.
Except for the special descriptions, all reactions of the invention are carried out by continuous magnetic stirring under the protection of dry inert gas (such as nitrogen or argon), and the reaction temperature is in degrees centigrade.
The following abbreviations are used throughout this application
THF: tetrahydrofuran (THF)
DCM: dichloromethane (dichloromethane)
PE: petroleum ether
Na 2 CO 3 : sodium carbonate
MeOH: methanol
HCl: hydrochloric acid
Pd(PPh 3 ) 4 : tetratriphenylphosphine palladium
K 2 CO 3 : potassium carbonate
H 2 O: water and its preparation method
TEA: triethylamine
DIEA: n, N-diisopropylethylamine
DMF: n, N-dimethylformamide
DMSO: dimethyl sulfoxide
NaBH 4 : sodium borohydride
Sn 2 (Bu-n) 6 : hexahexylditin
CuI: cuprous iodide
Cs 2 CO 3 : cesium carbonate
K 3 PO 4 : potassium phosphate
Pd 2 (dba) 3 : tris (dibenzylideneacetone) dipalladium
Pd/C: palladium carbon
Xantphos: 2-dicyclohexylphosphorus-2, 4, 6-triisopropylbiphenyl
EA: acetic acid ethyl ester
Boc 2 O: di-tert-butyl dicarbonate
Pd(dppf) 2 Cl 2 : [1,1' -bis (diphenylphosphine) ferrocene]Palladium dichloride
NaH: sodium hydrogen
CH 3 I: methyl iodide
L-Proline: l-proline
L-Selectride: trisec-butyl lithium borohydride
Examples
Example 1
Preparation of methyl 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carboxylate
Step 1: synthesis of N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
N- (3- (aminomethyl) pyrazin-2-yl) -N-methylmethanesulfonamide (5.0 g,19.8 mmol), 2, 4-dichloro-5-iodopyrimidine (5.0 g,18.2 mmol) and DIPEA (12.0 mL,72.6 mmol) were reacted at 0deg.C(100 mL) the solution was stirred for 3.5h. TLC (petroleum ether/ethyl acetate=5/1) showed that after completion of the reaction mixture was poured into water (150 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed with saturated brine (100 mL) and dried over anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and distilled off in a rotary manner to give a crude product. The crude product was purified by FCC (petroleum ether/ethyl acetate=10/1) to give N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide 7.54g.ms m/z (ESI): 455.2[ m+h)] +
Step 2: synthesis of 3- (2-chloro-4- ((((3- (N-methyl-sulfamido) pyrazin-2-yl) methyl) amino) pyrimidin-5-yl) propionate
Pd (PPh) was added separately to a solution of N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (3.0 g,6.6 mmol) and methyl propiolate (1.6 g,19.7 mmol) in DMF (30 mL) under nitrogen 3 ) 2 Cl 2 (70 mg), cuI (126 mg,0.661 mmol) and DIPEA (3.3 mL,20.0 mmol). The reaction was then stirred in an oil bath at 70℃for 6h under nitrogen protection. LCMS showed that after the reaction was completed, the reaction solution was suction filtered, then the filtrate was poured into water (200 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed 3 times with saturated brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered by suction and concentrated to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give 3- (2-chloro-4- ((((3- (N-methylsulfonylmethyl) pyrazin-2-yl) methyl) amino) pyrimidin-5-yl) propionate 1.76g.ms m/z (ESI): 411.3[ m+h)] +
Step 3: synthesis of methyl 2-chloro-7- (2- (N-methyl-sulfamido) benzyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carboxylate
To a solution of 3- (2-chloro-4- ((((3- (N-methylsulfonylmethyl) sulfanomide) pyrazin-2-yl) methyl) amino) pyrimidin-5-yl) propionate (1.23 g,3.04 mmol) in NMP (12 mL) at 0deg.C was added tBuOK (169 mg,3.64 mmol) the reaction solution was then warmed to room temperature and stirred overnight TLC (Petroleum ether/ethyl acetate)=5/1) showed that after the reaction was completed, the reaction solution was concentrated and dissolved 3 times with ethyl acetate (200 mL). After the organic phases were combined, the organic phase was washed 3 times with water (100 mL) and then 1 time with saturated brine (100 mL). Anhydrous Na for organic phase 2 SO 4 Drying, filtering and concentrating the dried product to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give 2-chloro-7- (2- (N-methyl-methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ]Pyrimidine-6-carboxylic acid methyl ester 230mg. MS m/z (ESI): 411.3[ M+H ]] +
Step 4: synthesis of methyl 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carboxylate
To 2-chloro-7- (2- (N-methyl-sulfamido) benzyl) -7H-pyrrolo [2,3-d ] at room temperature]To a solution of pyrimidine-6-carboxylic acid methyl ester (145 mg, 0.460 mmol) in 2-butanol (14 mL) was added TsOH.H 2 O (97 mg,0.510 mmol) and then the reaction was stirred for 8h at 115 ℃. TLC (DCM/meoh=15:1) showed that after the reaction was complete, the reaction was concentrated and dissolved with DCM (10 mL). The organic phase was washed 3 times with water (10 mL) and then 1 time with saturated brine (10 mL). Anhydrous Na for organic phase 2 SO 4 Drying, filtering and concentrating the dried product to obtain a crude product. The crude product was purified by flash column chromatography (DCM/meoh=100/1-20/1) to give 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d]Pyrimidine-6-carboxylic acid methyl ester 43.9mg. 1 H NMR(400MHz,DMSO-d 6 )δ10.07(s,1H),8.99(s,1H),8.51(d,J=2.4Hz,1H),8.43(d,J=2.4Hz,1H),8.26-8.19(m,1H),7.84(d,J=8.8Hz,2H),7.70(d,J=8.8Hz,2H),7.34(s,1H),6.05(s,2H),3.70(s,3H),3.37(s,3H),3.22(s,3H),2.75(d,J=4.4Hz,3H).MS m/z(ESI):525.5[M+H] +
Example 2
Preparation of 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carboxylic acid
To 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] under room temperature conditions]To a solution of pyrimidine-6-carboxylic acid methyl ester (130 mg,0.248 mmol) in methanol/water (5 mL/2.5 mL) was added LiOH H 2 O (104 mg,2.48 mmol). The reaction was then stirred overnight at 50 ℃. TLC (DCM/meoh=10:1) showed that after the reaction was complete the reaction was concentrated and taken up with H 2 O (20 mL) was dissolved. The solution was adjusted to pH 2 with 1M HCl and the precipitated yellow solid was filtered to give crude product, which was purified by flash column chromatography (DCM/meoh=100/1-10/1) to give 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylsulphonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-6-carboxylic acid 125mg. 1 H NMR(400MHz,DMSO-d 6 )δ10.03(s,1H),8.96(s,1H),8.50(d,J=2.4Hz,1H),8.43(d,J=2.8Hz,1H),8.23(dd,J=8.8,4.0Hz,1H),7.85(d,J=8.8Hz,2H),7.70(d,J=8.8Hz,2H),7.27(s,1H),6.02(s,2H),3.35(s,3H),3.21(s,3H),2.75(d,J=4.4Hz,3H)。MS m/z(ESI):511.5[M+H] +
Example 3
Preparation of N, N-dimethyl-2- (((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carboxamide
2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-6-carboxylic acid (80 mg,0.157 mmol) and dimethylamine hydrochloride (20 mg,0.245 mmol) were dissolved in DMF (1.5 mL), HATU (90 mg,0.237 mmol) and DIPEA (0.1 mL,0.60 mmol) were added and then reacted at room temperature After 2h, LCMS monitored completion of the reaction, the reaction was quenched with water (10 mL) and extracted 3 times with EA (20 mL), the organic phase was washed with saturated brine, and dried over anhydrous Na 2 SO 4 The organic phase was dried, and the dried product was filtered with suction and concentrated to give the crude product which was purified by column chromatography (MeOH/dcm=0-10%) to give N, N-dimethyl-2- (((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-6-carboxamide 82.0mg. 1 H NMR(400MHz,DMSO-d 6 )δ9.88(s,1H),8.86(s,1H),8.52(d,J=2.4Hz,1H),8.48(d,J=2.4Hz,1H),8.41-8.19(m,1H),7.84(d,J=8.8Hz,2H),7.72(d,J=8.8Hz,2H),6.82(s,1H),5.81(s,2H),3.31(s,3H),3.17(s,3H),3.11-3.04(m,3H),2.87-2.79(m,3H),2.76(d,J=4.4Hz,3H).MS m/z(ESI):538.5[M+H] +
Example 4
Preparation of N-methyl-4- ((6-methyl-7- (((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) benzamide
Step 1: synthesis of 3N- (3- ((((2-chloro-5- (propynyl-1)) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
Pd (PPh) was added separately to a solution of N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (586.3 mg,1.29 mmol) and tributyl (prop-1-yn-1-yl) stannane (850.0 mg,2.58 mmol) in toluene (20 mL) under nitrogen 3 ) 4 (150.0 mg,0.13 mmol) and CuI (24.75 mg,0.13 mmol) were added and the reaction was stirred in an oil bath at 100deg.C under nitrogen for 2h. LCMS showed that after the reaction was completed, the reaction solution was suction filtered, then poured into water (10 mL) and extracted 2 times with ethyl acetate (10 mL). The organic phases were combined and washed with saturated brine (20 mL) and dried over anhydrous Na 2 SO 4 Drying the organic phase and pumping the dried productFiltering and concentrating to obtain crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give N- (3- ((((2-chloro-5- (propynyl-1)) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide 230mg. MS m/z (ESI): 367.1[ M+H ]] +
Step 2: synthesis of N- (3- ((2-chloro-6-methyl-7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide
To a solution of N- (3- ((((2-chloro-5- (propynyl-1)) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (230 mg,0.63 mmol) in DMF (10 mL) was added DBU (0.85 mL,6.3 mmol) at 0deg.C. Then the reaction solution was stirred for 5 hours while heating to 60 ℃. TLC (petroleum ether/ethyl acetate=5/1) showed that after the reaction was completed, the reaction solution was concentrated and dissolved with ethyl acetate (5 mL). The organic phase was washed 3 times with water (5 mL) and then 1 time with saturated brine (10 mL). Anhydrous Na for organic phase 2 SO 4 Drying, filtering and concentrating the dried product to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=4/1) to give N- (3- ((2-chloro-6-methyl-7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 80mg. MS m/z (ESI): 367.1[ M+H ] ] +
Step 3: synthesis of N-methyl-4- ((6-methyl-7- (((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) benzamide
To N- (3- ((2-chloro-6-methyl-7H-pyrrolo [2, 3-d) at room temperature]To a solution of pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methyl methanesulfonamide (50 mg,0.14 mmol) in 2-butanol (5 mL) was added para-aminobenzamide (22.54 mg,0.15 mmol) and TsOH.H 2 O (26.6 mg,0.14 mmol). Then the reaction solution was stirred for 8 hours while heating to 120 ℃. TLC (DCM/meoh=15:1) showed that after the reaction was complete, the reaction was concentrated and dissolved with DCM (5 mL). The organic phase was washed 3 times with water (5 mL) and then 1 time with saturated brine (10 mL). Anhydrous Na for organic phase 2 SO 4 Drying, filtering and concentrating the dried product to obtain a crude product. The crude product was purified by flash column chromatography (DCM/meoh=100/1-20/1) to give N-methyl-4- ((6-methyl-7- (((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) benzamide 25mg. 1 H NMR(400MHz,DMSO-d 6 )δ9.66(s,1H),8.64(s,1H),8.56(d,J=2.4Hz,1H),8.52(d,J=2.5Hz,1H),8.18(q,J=4.1Hz,1H),6.31(d,J=1.1Hz,1H),5.70(s,2H),3.29(s,3H),3.22(s,3H),2.75(d,J=4.5Hz,3H),2.24(s,3H).MS m/z(ESI):481.2[M+H] +
Example 5
Preparation of 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carboxamide
2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylmethylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-6-carboxylic acid (30 mg,0.06 mmol), HATU (33.5 mg,0.09 mmol) and DIPEA (20. Mu.L, 0.12 mmol) were dissolved in DMF (1 mL), reacted at room temperature for 1H, aqueous ammonia (20. Mu.L, 0.12 mmol) was added, reacted at room temperature for 2H, water was added after completion of the reaction was monitored, DCM was added to extract, the concentrated extract was dried and the concentrate was purified by column chromatography to give 2- ((4- (methylcarbamoyl) phenyl) amino) -7- ((3- (N-methylsulfonylmethyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-6-carboxamide 10mg. 1 H NMR(400MHz,DMSO)δ9.90(s,1H),8.91(s,1H),8.44(d,J=2.3Hz,1H),8.36(d,J=2.5Hz,1H),8.18(d,J=4.8Hz,1H),7.92(s,1H),7.81(d,J=8.8Hz,2H),7.66(d,J=8.8Hz,2H),7.23(s,1H),7.19(s,1H),6.00(s,2H),3.33(s,4H),3.16(s,3H),2.71(d,J=4.5Hz,3H).MS m/z(ESI):510.0[M+H] +
Example 6
Preparation of 4- ((6-cyano-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
Step 1 Synthesis of N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
2, 4-dichloro-5-iodopyrimidine (1.2 g,4.37 mmol) was dissolved in DMF (12 mL), DIPEA (1.3 mL,7.94 mmol) and N- (3- (aminomethyl) pyrazin-2-yl) -N-methylmethanesulfonamide (0.85 g,3.97 mmol) were added at 0deg.C, reaction was carried out for 2h, water was added after completion of the reaction, ethyl acetate was added to extract, the extract was concentrated by drying, and the concentrate was purified by column chromatography to give N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide 1.0g, MS m/z (ESI): 454.8[ M+H ] ] +
Step 2 Synthesis of N- (3- (((2-chloro-5- (3, 3-diethoxyprop-1-yn-1-yl) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (960 mg,2.11 mmol), 3-diethoxyprop-1-yne (0.48 mL,2.75 mmol), cuI (81 mg,0.42 mmol), pd (PPh) 2 Cl 2 (150 mg,0.21 mmol) and DIPEA (10 mL) were dissolved in DMF (10 mL), reacted for 14h at 65℃and monitored for completion, water was added after the reaction was completed, ethyl acetate was added for extraction, the extract was concentrated by drying, and the concentrate was purified by column chromatography to give N- (3- (((2-chloro-5- (3, 3-diethoxyprop-1-yn-1-yl) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylsulfonamide (950 mg, MS m/z (ESI): 455.0[ M+H)] +
Step 3 Synthesis of N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
N- (3- (((2-chloro-5- (3, 3-diethoxyprop-1-yn-1-yl) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (950 mg,2.09 mmol) was dissolved in THF (9.5 mL), TBAF (12.6 mL,12.55 mmol) was added, the reaction was allowed to proceed overnight at room temperature, water was added after completion of the reaction, ethyl acetate was added to extract, the extract was dried, and the concentrate was purified by column chromatography The polycondensate gives N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 400mg. MS m/z (ESI) 454.9[ M+H ]] +
Step 4 Synthesis of N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide
N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl pyrazin-2-yl) -N-methyl methanesulfonamide (200 mg,0.44 mmol) was dissolved in dioxane (2 mL), concentrated hydrochloric acid (0.5 mL) was added under ice bath, reaction was performed at room temperature for 30min, water was added after completion of the reaction, saturated sodium carbonate solution was added to adjust pH to neutral, ethyl acetate was added for extraction, the extract was concentrated by drying, and the concentrate was purified by column chromatography to give N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 167mg. MS m/z (ESI) 380.9[ M+H ]] +
Step 5 Synthesis of N- (3- ((2-chloro-6- ((hydroxyimino) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide
N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl pyrazin-2-yl) -N-methyl methanesulfonamide (167 mg,0.44 mmol) and hydroxylamine hydrochloride (80 mg,1.15 mmol) were dissolved in absolute ethanol (4 mL), reacted at 50℃for 3H, the reaction mixture was concentrated after completion of the reaction, water was added, and saturated sodium hydrogencarbonate solution was added to adjust the pH to neutral, and the filter cake was filtered and washed with water to give N- (3- ((2-chloro-6- ((hydroxyimino) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 154mg. MS m/z (ESI): 395.9[ M+H ]] +
Step 6 Synthesis of N- (3- ((2-chloro-6-cyano-7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
N- (3- ((2-chloro-6- ((hydroxyimino) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl pyrazin-2-yl) -N-methyl methanesulfonamide (154 mg,0.39 mmol) was dissolved in DCE (3 mL), CDI (316 mg,1.95 mmol) was added under ice bath, reacted at room temperature for 1.5 hours, the reaction mixture was concentrated after the completion of the reaction, and then purified by column chromatography to obtainTo N- (3- ((2-chloro-6-cyano-7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 132mg. MS m/z (ESI): 377.9[ M+H ]] +
Step 7 Synthesis of 4- ((6-cyano-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
N- (3- ((2-chloro-6-cyano-7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl pyrazin-2-yl) -N-methyl methanesulfonamide (100 mg,0.27 mmol), 4-amino-N-methyl benzamide (47.8 mg,0.32 mmol) and TsOH (50.5 mg,0.27 mmol) were dissolved in sec-butanol (5 mL), refluxed at 115℃for 5H, monitored for completion, cooled to room temperature, added with water, extracted with ethyl acetate, the extract was dried and the concentrate purified by column chromatography to give 4- ((6-cyano-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-2-yl) amino) -N-methylbenzamide 33mg. 1 H NMR(400MHz,DMSO)δ10.10(s,1H),8.98(s,1H),8.61(d,J=2.5Hz,1H),8.56(d,J=2.5Hz,1H),8.23(d,J=4.5Hz,1H),7.77(d,J=8.9Hz,2H),7.71(d,J=8.9Hz,2H),7.54(s,1H),5.80(s,2H),3.30–3.25(m,3H),3.20(s,3H),2.75(d,J=4.5Hz,3H).MS m/z(ESI):492.1[M+H] +
Example 7
Preparation of 4- ((6-formyl-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
Step 1 Synthesis of 4- ((6- (Diethoxymethyl) -7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methyl methanesulfonamide (70 mg,0.15 mmol), 4-amino-N-methylbenzamide (27.8 mg,0.19 mmol), BINAP (19 mg,0.03 mmol), pd 2 (dba) 3 (14 mg,0.02 mmol) and sodium t-butoxide (30 mg,0.31 mmol) were dissolved in dioxane (7 mL), nitrogen protected, reacted at 100deg.C for 4.5H, monitored for completion, water was added, ethyl acetate was added to extract, the concentrate was dried, and the concentrate was purified by column chromatography to give 4- ((6- (diethoxymethyl) -7- ((3- (N-methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-methylbenzamide 47mg, MS m/z (ESI): 569.1[ M+H ]] +
Step 2 Synthesis of 4- ((6-formyl-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
4- ((6- (Diethoxymethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -N-methylbenzamide (47 mg,0.08 mmol) was dissolved in dioxane (0.7 mL), concentrated hydrochloric acid (0.09 mL) was added under ice bath, reaction was performed at room temperature for 10min, water was added after completion of the reaction, saturated sodium carbonate solution was further added to adjust pH to neutral, ethyl acetate was added to extract, the extract was dried, and the concentrate was purified by column chromatography to give 4- ((6-formyl-7- ((3- (N-methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-methylbenzamide 10mg. 1 H NMR(400MHz,DMSO)δ10.20(s,1H),9.63(s,1H),9.08(s,1H),8.50(d,J=2.5Hz,1H),8.40(d,J=2.5Hz,1H),8.23(d,J=5.0Hz,1H),7.84(d,J=8.9Hz,2H),7.71(d,J=8.8Hz,2H),7.56(s,1H),5.98(s,2H),3.40(s,4H),3.20(s,3H),2.75(d,J=4.5Hz,3H).MS m/z(ESI):495.1[M+H] +
Example 8
Preparation of 4- ((6-formyl-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide
Step 1: synthesis of 3-methoxy-N-methyl-4-nitrobenzamide
3-methoxy-4-nitrobenzoic acid (600 mg,3.04 mmol), HATU (1.73 g,4.57 mmol) and DIPEA (1 mL,6.08 mmol) were dissolved in DMF (6 mL), reacted at room temperature for 1h, methylamine hydrochloride (400 mg,6.08 mmol) was added, reacted at room temperature for 3h, water was added after the reaction was monitored to be complete, ethyl acetate was added for extraction, the extract was concentrated by drying, and the concentrate was purified by column chromatography to give 251mg of 3-methoxy-N-methyl-4-nitrobenzamide. MS m/z (ESI): 211.0[ M+H ] ] +
Step 2: synthesis of 4-amino-3-methoxy-N-methylbenzamide
3-methoxy-N-methyl-4-nitrobenzamide (251 mg,1.20 mmol), iron powder (160 mg,2.99 mmol) and ammonium chloride (160 mg,2.99 mmol) were dissolved in methanol (12 mL) and water (3 mL), reacted at 70℃for 12 hours, a small amount of water was added to the reaction system after monitoring the reaction was completed, suction filtration was performed, the filtrate was concentrated, and the concentrate was purified by column chromatography to give 193mg of 4-amino-3-methoxy-N-methylbenzamide. MS m/z (ESI): 181.1[ M+H ]] +
Step 3: synthesis of 4- ((6- (Diethoxymethyl) -7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide
N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methyl methanesulfonamide (57 mg,0.13 mmol), 4-amino-3-methoxy-N-methylbenzamide (27.2 mg,0.15 mmol), BINAP (16 mg,0.03 mmol), pd 2 (dba) 3 (12 mg,0.01 mmol) and sodium t-butoxide (24 mg,0.25 mmol) were dissolved in dioxane (3 mL), nitrogen protected, reacted at 100deg.C for 4.5H, monitored for completion, water was added, ethyl acetate was added to extract, the concentrate was dried, and the concentrate was purified by column chromatography to give 4- ((6- (diethoxymethyl) -7- ((3- (N-methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide 40mg, MS m/z (ESI): 599.1[ M+H ]] +
Step 4: synthesis of 4- ((6-formyl-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide
4- ((6- (Diethoxymethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -3-methoxy-N-methylbenzamide (40 mg,0.07 mmol) was dissolved in dioxane (0.6 mL), concentrated hydrochloric acid (0.07 mL) was added under ice bath, reaction was performed at room temperature for 30min, water was added after completion of the reaction, saturated sodium carbonate solution was further added to adjust pH to neutral, ethyl acetate was added to extract, the extract was concentrated by drying, and the concentrate was purified by column chromatography to give 4- ((6-formyl-7- ((3- (N-methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide 12mg. 1 H NMR(400MHz,DMSO)δ9.64(s,1H),9.06(s,1H),8.50(d,J=2.5Hz,1H),8.44–8.37(m,2H),8.31(dd,J=11.9,6.5Hz,2H),7.56(s,1H),7.48(d,J=1.8Hz,1H),7.38(m,1H),5.97(s,2H),3.91(s,3H),3.38(s,3H),3.19(s,3H),2.77(d,J=4.5Hz,3H).MS m/z(ESI):525.1[M+H] +
Example 9
Synthesis of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide preparation
Step 1: synthesis of N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl pyrazin-2-yl) -N-methyl methanesulfonamide (45 mg,0.12 mmol) was dissolved in DCM (1 mL), BAST (24. Mu.L) was added under ice bath, reacted at room temperature for 5H, water was added after completion of the reaction, dichloromethane was added for extraction, the extract was concentrated, and the concentrate was purified by column chromatography to give N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide 34mg, MS m/z (ESI): 403.0[ M+H ]] +
Step 2: synthesis of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl pyrazin-2-yl) -N-methyl methanesulfonamide (28 mg,0.07 mmol), 4-amino-N-methyl benzamide (13 mg,0.08 mmol) and p-toluenesulfonic acid (13 mg,0.07 mmol) were dissolved in sec-butanol (3 mL), reacted at 115℃for 14H, the reaction was monitored to completion, the system was cooled to room temperature, water was added, ethyl acetate was added to extract, the extract was concentrated by drying, and the concentrate was purified by column chromatography to give 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-2-yl) amino) -N-methylbenzamide 17mg. 1 H NMR(400MHz,DMSO)δ9.88(s,1H),8.89(s,1H),8.54(d,J=2.4Hz,1H),8.47(d,J=2.5Hz,1H),8.19(d,J=4.5Hz,1H),7.75(d,J=8.9Hz,2H),7.66(d,J=8.9Hz,2H),7.19(s,1H),6.94(s,1H),5.81(s,2H),3.29(s,3H),3.20(s,3H),2.74(d,J=4.5Hz,3H).MS m/z(ESI):517.0[M+H] +
Example 10
Preparation of 4- ((6-chloro-7- ((2- (N-methyl methylsulfonyl) pyridin-3-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
Step 1: synthesis of 2, 6-dichloro-7H-pyrrolo [2,3-d ] pyrimidine
2-chloro-5, 7-dihydro-6H-pyrrolo [2,3-d ] at 0deg.C]Pyrimidin-6-one (1.0 g,5.8 mmol) was dissolved in POCl 3 In (10 mL) solution, slowly heating to 110 ℃ and stirring for reaction for 1h, concentrating and spin-drying the reaction solution after TLC detection reaction is completed, diluting and dissolving the reaction solution with DCM (20 mL), adding saturated NaHCO 3 The solution (15 mL) was extracted, the combined organic phases were separated, the organic phase was washed with saturated aqueous NaCl (20 mL), the organic phase was concentrated by rotation and purified by column chromatography (PE/ea=from 0 to 50%) to give 26-dichloro-7H-pyrrolo [2,3-d]Pyrimidine 600mg. MS m/z (ESI): 188.0[ M+H ]] +
Step 2: synthesis of N- (3- ((2, 6-dichloro-7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyridin-2-yl) -N-methyl methanesulfonamide
At room temperature, 2, 6-dichloro-7H-pyrrolo [2,3-d]Pyrimidine (600 mg,3.2 mmol) and N- (3- (chloromethyl) pyridin-2-yl) -N-methylmethanesulfonamide (900 mg,3.8 mmol) were dissolved in DMF (5 mL) and K was added 2 CO 3 (660 mg,6.4 mmol). The mixture was then stirred for 8 hours at 45 ℃. LCMS showed that after the reaction was complete, the reaction mixture was quenched with water (100 mL) and slowly extracted 2 times with DCM (80 mL). The combined organic phases were washed with brine (100 mL) and with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by ISCO column chromatography (petroleum ether/ethyl acetate=3/1) to give N- (3- ((2, 6-dichloro-7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyridin-2-yl) -N-methyl methanesulfonamide 200mg, MS m/z (ESI): 386.0[ M+H ]] +
Step 3: synthesis of 4- ((6-chloro-7- ((2- (N-methyl methylsulfonyl) pyridin-3-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methylbenzamide
To N- (3- ((2, 6-dichloro-7H-pyrrolo [2, 3-d) at room temperature]To a solution of pyrimidin-7-yl) methyl-pyridin-2-yl) -N-methyl methanesulfonamide (0.1 g,0.26 mmol) in 2-butanol (5 mL) was added para-aminobenzamide (46.7 mg,0.31 mmol) and TsOH.H 2 O (49.3 mg,0.26 mmol). Then the reaction solution was stirred for 8 hours while heating to 120 ℃. TLC (DCM/meoh=15:1) showed that after the reaction was complete, the reaction was concentrated and dissolved with DCM (5 mL). The organic phase was washed with water (10 mL) followed by brine (10 mL). Anhydrous Na for organic phase 2 SO 4 And (5) drying, filtering and concentrating the dried product to obtain a crude product. The crude product was purified by flash column chromatography (DCM/meoh=from 100/1 to 20/1) to give 4- ((6-chloro-7- ((2- (N-methyl methylsulfonyl) pyridin-3-yl) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-2-yl) amino) -N-methylbenzamide 51mg. 1 H NMR(400MHz,DMSO-d 6 )δ9.84(s,1H),8.78(s,1H),8.49(dd,J=4.7,1.7Hz,1H),8.19(d,J=4.6Hz,1H),7.79(d,J=8.8Hz,2H),7.69(d,J=8.8Hz,2H),7.36(dd,J=7.8,4.7Hz,1H),7.13(dd,J=7.8,1.6Hz,1H),6.74(s,1H),5.58(s,2H),3.27(s,3H),3.17(s,3H),2.75(d,J=4.5Hz,3H).MS m/z(ESI):500.3[M+H] +
Example 11
Preparation of 4- ((6- (difluoromethyl) -7- (2- (dimethylphosphoryl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide
Step 1: synthesis of 2- (dimethylphosphinoyl) benzonitrile
2-iodobenzonitrile (6.90 g,30.1 mmol) and phosphorus dimethyloxide (2.50 g,45.2 mmol) were dissolved in acetonitrile (150 mL) and Pd was added sequentially under nitrogen protection 2 (dba) 3 (900mg,1.50mmol)、Xant-Phos(1.20g,3.01mmol)、Cs 2 CO 3 (12.0 g,37.0 mmol) and triethylamine (21 ml,150 mmol) were then reacted at constant temperature to 85℃for 19h. After the TLC detection reaction is completed, the reaction liquid is filtered by suction, the filter cake is washed by acetonitrile for 3 times, and the filtrate is concentrated to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give 4.80g of 2- (dimethylphosphinoyl) benzonitrile as a yellow solid. MS m/z (ESI) 180.2[ M+H ]] +
Step 2: synthesis of (2- (aminomethyl) phenyl) dimethylphosphine oxide
2- (dimethyl phosphinoyl) benzonitrile (4.80 g,28.2 mmol) was dissolved in tetrahydrofuran (60 ml) and cooled to 0 ℃. Borane tetrahydrofuran complex (84.6 ml,1.00mol/L,84.6 mmol) was slowly added dropwise to the reaction system under nitrogen protection, slowly warmed to room temperature and stirred for 2h. After completion of the TLC detection the reaction solution was slowly quenched with methanol (100 mL) and concentrated. The crude product was purified by flash column chromatography (dichloromethane/methanol=10/1) to give (2- (aminomethyl) phenyl) dimethylphosphine 1.30g, yellow brown gum. MS m/z (ESI): 184 .1[M+H] +
Step 3: synthesis of (2- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) phenyl) dimethylphosphine oxide
2, 4-dichloro-5-iodopyrimidine (1.58 g,5.46 mmol) and DIPEA (1.80 mL,10.9 mmol) were dissolved in DMF (10 mL), cooled to-5-0deg.C, and then a solution of (2- (aminomethyl) phenyl) dimethylphosphine oxide (1.30 g,5.46 mmol) in DMF (5 mL) was added dropwise. After completion of the dropwise addition, the reaction was carried out at 0℃for 2 hours, water was added after completion of the reaction, dichloromethane was added to extract, and the extract was dried and concentrated to obtain a crude product, which was purified by flash column chromatography (petroleum ether/ethyl acetate=20/1) to give (2- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) phenyl) dimethylphosphine 1.35g, a white solid, MS m/z (ESI): 422.9[ M+H)] +
Step 4: synthesis of (2- ((((2-chloro-5- (3, 3-diethoxyprop-1-ynyl) pyrimidin-4-yl) amino) methyl) phenyl) dimethylphosphine oxide
(2- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) phenyl) dimethylphosphine (1.35 g,3.20 mmol) was dissolved in DMF (50 mL) Pd (PPh) was added separately under nitrogen protection 3 )Cl 2 (225mg,0.32mmol)、PPh 3 (42 mg,0.16 mmol), cuprous iodide (122 mg,0.64 mmol), triethylamine (1.33 mL,9.61 mmol) and 3, 3-diethoxyprop-1-yne (440 mg,3.42 mmol). Reacting at 60 ℃ for 4 hours, pumping and filtering the reaction system after TLC monitoring reaction is complete, and pouring filtrate into 10% NH 4 Cl aqueous solution (100 mL), ethyl acetate extraction, concentration to obtain crude product. The crude product was purified by flash column chromatography (dichloromethane/methanol=10/1) to give (2- ((((2-chloro-5- (3, 3-diethoxyprop-1-ynyl) pyrimidin-4-yl) amino) methyl) phenyl) dimethylphosphine oxide 665mg, MS m/z (ESI): 422.1[ M+H)] +
Step 5: synthesis of (2- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) phenyl) dimethylphosphine oxide
(2- ((((2-chloro-5- (3, 3-diethoxyprop-1-ynyl) pyrimidin-4-yl) amino) methyl) phenyl) dimethylphosphine oxide (665 mg,1.58 mmol) was dissolved in tetrahydrofuran (40 mL), TBAF (9.50 mL,9.50mmol,1 mol/L) was added and the reaction was allowed to proceed to 65℃for 2h.TLC to monitor completion of the reactionThe reaction solution was concentrated to give a crude product, which was purified by flash column chromatography (dichloromethane/methanol=20/1) to give (2- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) phenyl) dimethyl phosphine oxide crude 1.59g, brown oil. MS m/z (ESI): 422.1[ M+H ]] +
Step 6: synthesis of 2-chloro-7- (2- (dimethylphosphoryl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carbaldehyde
(2- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-7-yl) methyl phenyl) dimethylphosphine (1.59 g, crude) was dissolved in 1, 4-dioxane (13 mL), concentrated hydrochloric acid (8.00 mL,12 mol/L) was added in portions, stirred at room temperature for 30min, and after completion of LCMS monitoring the reaction, water (30 mL) was added to dilute the reaction solution, followed by extraction with ethyl acetate (50 mL) 3 times. The organic phase was washed with saturated brine, then dried over anhydrous sodium sulfate, the dried product was filtered, and concentrated to give a crude product. The crude product was purified by flash column chromatography (dichloromethane/methanol=15/1) to give 2-chloro-7- (2- (dimethylphosphoryl) benzyl) -7H-pyrrolo [2,3-d ]980mg of pyrimidine-6-carbaldehyde crude product was found to be a brown oil. MS m/z (ESI) 348.1[ M+H ]] +
Step 7: synthesis of (2- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) phenyl) dimethylphosphine oxide
2-chloro-7- (2- (dimethylphosphoryl) benzyl) -7H-pyrrolo [2,3-d]Pyrimidine-6-carbaldehyde (980 mg, crude product) was dissolved in methylene chloride (20 mL), cooled to 0℃and then DAST (750 mL) was added dropwise thereto, and the mixture was stirred at room temperature for 2h. After completion of LCMS monitoring, the reaction was quenched by slowly dropping the solution into water, extracting 3 times with ethyl acetate (50 mL), drying the organic phase over anhydrous sodium sulfate, suction-filtering the dried product and concentrating to obtain crude product. The crude product was purified by flash column chromatography (dichloromethane/methanol=20/1) to give 2- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) phenyl) dimethylphosphine 128mg, pale yellow solid. MS m/z (ESI) 370.1[ M+H ]] +
Step 8: synthesis of 4-amino-3-methoxy-N-methylbenzamide
4-amino-3-methoxybenzoic acid (2.00 g,11.9 mmol) and methyl acetateAmine hydrochloride (8.08 g,119.6 mmol) was dissolved in DMF (100 mL), sodium carbonate (12.7 g,119.6 mmol) was added in portions followed by HATU (9.10 g,23.9 mmol) and reacted overnight at room temperature. After the completion of the TLC monitoring, the reaction solution was added to water, followed by extraction 3 times with ethyl acetate (50 mL), and the organic phase was dried over anhydrous sodium sulfate, and the dried product was suction-filtered and concentrated to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=1/1) to give 1.42g of 4-amino-3-methoxy-N-methylbenzamide as a white solid. MS m/z (ESI): 181.1[ M+H ] ] +
Step 9: synthesis of 4- ((6- (difluoromethyl) -7- (2- (dimethylphosphoryl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide
4-amino-3-methoxy-N-methylbenzamide (50 mg,0.28 mmol), 2- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) phenyl dimethyl phosphine oxide (100 mg,0.27 mmol) and p-toluene sulfonic acid (55 mg,0.32 mmol) were dissolved in n-butanol (2.00 mL). The reaction mixture was heated to 115℃and reacted overnight. After completion of the LCMS detection reaction, the reaction mixture was poured into water (10 mL) and extracted 3 times with ethyl acetate (20 mL). The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the dried product was filtered and concentrated to give a crude product. The crude product was purified by flash column chromatography (dichloromethane/methanol=20/1) and pre-TLC to give 4- ((6- (difluoromethyl) -7- (2- (dimethylphosphoryl) benzyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide 3.5mg, yellow solid. MS m/z (ESI): 513.2[ M+H ]] +
Example 12
Preparation of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-alkyl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-alkyl) amino) -3-methoxy-N-methylbenzamide
Step 1: synthesis of N- (3-cyanopyrazin-2-yl) -N-methylmethanesulfonamide
3-Chloropyrazine-2-carbonitrile (8.40 g,60.2 mmol), N-methyl-sulfamide (8.00 g,73.3 mmol) and K 2 CO 3 A mixture of (9.00 g,65.1 mmol) of acetonitrile (100 mL) was stirred at 85deg.C for 12 hours. TLC (petroleum ether/ethyl acetate=1/1) showed that after the reaction was completed, the reaction solution was suction-filtered, the filter cake was washed 3 times with ACN (50 mL), and the filtrate was concentrated to give a crude product. The crude product was purified by flash chromatography (FCC, petroleum ether/ethyl acetate=3/1) to give N- (3-cyanopyrazin-2-yl) -N-methylmethanesulfonamide (13.3 g, crude). MS m/z (ESI): =213.04 [ m+h ]] +
Step 2: synthesis of N- (3- (aminomethyl) pyrazin-2-yl) -N-methylmethanesulfonamide
To a mixture of N- (3-cyanopyrazin-2-yl) -N-methylmethanesulfonamide (13.3 g) and 1M HCl (2.4 mL) in MeOH (150 mL) was added 10% Pd/C (5.50 g) at room temperature. H 2 3 times of displacement, and then the reaction solution was subjected to H 2 Stirring was carried out for 12h under (15 psi) conditions. LCMS showed that after the reaction was completed, the reaction was suction filtered through celite and the filter cake was washed with MeOH (50 mL). Concentrating the filtrate to obtain a crude product. The crude product was slurried in DCM (100 mL), suction filtered and dried to give N- (3- (aminomethyl) pyrazin-2-yl) -N-methylmethanesulfonamide (HCl, 5.26g, 34.6%). MS m/z (ESI): 217.04[ M+H ] ] +
Step 3: synthesis of N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
A solution of N- (3- (aminomethyl) pyrazin-2-yl) -N-methylmethanesulfonamide (HCl, 5.26g,20.83 mmol), 2, 4-dichloro-5-iodopyrimidine (5.73 g,20.84 mmol) and DIPEA (6.88 mL,41.7 mmol) in DMF (60 mL) was stirred at 0deg.C for 2h. TLC (petroleum ether/ethyl acetate=5/1) showed that after the reaction was completed, the reaction mixture was poured into water (150 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed 3 times with saturated brine (100 mL) and then with anhydrous Na 2 SO 4 Drying, filtering the dried product and steaming the product to obtain a crude product. The crude product was purified by FCC (petroleum ether/ethyl acetate=10/1) to give N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (5.88 g,12.9mmol,62.1%)。MS m/z(ESI):454.9[M+H] +
step 4: synthesis of N- (3- (((2-chloro-5- (3, 3-diethoxyprop-1-yn-1-yl) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
Pd (PPh) was added separately to a solution of N- (3- ((((2-chloro-5-iodopyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (5.88 g,12.9 mmol) and 3, 3-diethoxyprop-1-yne (2.49 g,19.4 mmol) in DMF (80 mL) under nitrogen 3 ) 2 Cl 2 (907mg,1.29mmol)、CuI(494mg,2.59mmol)、PPh 3 (170 mg,0.644 mmol) and TEA (5.40 mL,38.8 mmol). The reaction was then stirred in an oil bath at 60℃for 2h under nitrogen protection. LCMS showed that after the reaction was filtered off with suction, the reaction was poured into water (200 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed 3 times with saturated brine (100 mL) and then with anhydrous Na 2 SO 4 Drying, suction filtering and concentrating the dried product to obtain crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give N- (3- (((2-chloro-5- (3, 3-diethoxyprop-1-yn-1-yl) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (8.00 g, crude product). MS m/z (ESI): 455.0[ M+H ]] +
Step 5: synthesis of N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
To a solution of N- (3- (((2-chloro-5- (3, 3-diethoxyprop-1-yn-1-yl) pyrimidin-4-yl) amino) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide (8.00 g, crude) in THF (40 mL) was added TBAF (1M, 78mL,78.0 mmol) at 25 ℃. The reaction solution was then stirred in an oil bath at 60℃for 1h. TLC (petroleum ether/ethyl acetate=5/1) showed that after the reaction was completed, the reaction solution was concentrated and dissolved with ethyl acetate (200 mL). The organic phase was washed 3 times with water (100 mL) and then 1 time with saturated brine (100 mL). Anhydrous Na for organic phase 2 SO 4 Drying, filtering and concentrating the dried product to obtain a crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) purification to give N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-acyl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide (2.53 g,5.56mmol, 43.0%). MS m/z (ESI): 455.0[ M+H ]] +
Step 6: synthesis of N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide
At 25℃to N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]To a solution of pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methyl methanesulfonamide (2.53 g,5.56 mmol) in 1, 4-dioxane (40 mL) was added 1M aqueous HCl (1M, 16 mL). The reaction solution was then stirred at 25℃for 2h. LCMS showed that after the reaction was complete, the reaction mixture was poured into water (80 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed with saturated brine (100 mL) and with anhydrous Na 2 SO 4 And (5) drying. The dried product was filtered and evaporated to give N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide (2.48 g, crude). MS m/z (ESI): 381.0[ M+H ] ] +
Step 7: synthesis of N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
At 0deg.C, to N- (3- ((2-chloro-6-formyl-7H-pyrrolo [2, 3-d)]To a solution of pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methylsulfonylamines (2.48 g, crude) in DCM (40 mL) was slowly added DAST (2.50 mmol,19.3 mmol) dropwise. The reaction solution was then stirred at 25℃for 2h. LCMS showed that after the reaction was completed, the reaction was quenched by slowly dropping into water (100 mL) and extracted 2 times with DCM (80 mL). The combined organic phases were washed with brine (100 mL) and then with anhydrous Na 2 SO 4 And (5) drying. The dried product was filtered off with suction and concentrated to give the crude product. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-acyl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide (1.38 g,3.43mmol,61.7%, two-step yield). MS m/z (ESI): 403.0[ M+H ]] +
Step 8: synthesis of N- (3- ((6- (difluoromethyl) -2- ((1-isopropyl-3-methyl-1H-pyrazol-5-yl) amino) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen ]Pd (OAc) was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (100 mg,0.248 mmol) and 1-isopropyl-3-methyl-1H-pyrazol-5-amine (52.0 mg,0.372 mmol) in 1, 4-dioxane (3 mL), respectively 2 (12 mg,0.05 mmol), BINAP (62 mg,0.099 mmol) and Cs 2 CO 3 (242 mg,0.774 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and concentrated to obtain crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give N- (3- ((6- (difluoromethyl) -2- ((1-isopropyl-3-methyl-1H-pyrazol-5-yl) amino) -7H-pyrrolo [2, 3-d)]Pyrimidine-7-acyl) methyl) pyrazin-2-alkyl) -N-methyl methanesulfonamide 56mg. 1 HNMR(400MHz,DMSO-d 6 )9.07(s,1H),8.75(s,1H),8.51(d,J=4.00Hz,1H),8.45(d,J=4.00Hz,1H),7.16(m,1H),5.73(m,1H),5.62(s,2H),3.15(d,J=8Hz,6H),1.17(d,J=8Hz,6H).MS m/z(ESI):506.1[M+H] +
Example 13
Preparation of N- (3- ((6- (difluoromethyl) -2- ((2-oxoindol-6-alkyl) amino) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen ]Pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylPd (OAc) was added to a solution of sulfonamide (80 mg, 0.199mmol) and 6-aminoindol-2-one (45.0 mg,0.298 mmol) in 1, 4-dioxane (2 mL), respectively 2 (9 mg,0.04 mmol), BINAP (50 mg,0.08 mmol) and Cs 2 CO 3 (195 mg,0.579 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and dried over anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered by suction and concentrated to obtain crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give N- (3- ((6- (difluoromethyl) -2- ((2-oxoindol-6-alkyl) amino) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 12mg. MS m/z (ESI): 515.1[ M+H ]] +
Example 14
Preparation of N- (3- ((6- (difluoromethyl) -2- ((2-methoxy-4-morpholinophenyl) amino) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd (OAc) was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (80 mg, 0.1998 mmol) and 2-methoxy-4-morpholinoaniline (62.2 mg,0.298 mmol) in 1, 4-dioxane (2 mL), respectively 2 (9 mg,0.04 mmol), BINAP (50 mg,0.08 mmol) and Cs 2 CO 3 (195 mg,0.579 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and then with anhydrous Na 2 SO 4 Drying the organic phase, and then suction-filtering and concentrating the dried product to obtainTo crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give N- (3- ((6- (difluoromethyl) -2- ((2-oxoindol-6-alkyl) amino) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide 45mg. 1 HNMR(400MHz,DMSO-d 6 )8.74(s,1H),8.54(d,J=4.00Hz,1H),8.48(d,J=4.00Hz,1H),7.84(s,1H),7.69(d,J=8.00Hz,1H),7.17(t,J=64.0Hz,1H),6.86(s,1H),6.61(s,1H),6.36–6.33(m,1H),5.70(m,2H),3.77(s,3H),3.74(t,J=4.00Hz,4H),3.24(s,3H),3.18(s,3H),3.05(t,J=4.00Hz,4H).MS m/z(ESI):575.1[M+H] +
Example 15
Preparation of N- (3- ((6- (difluoromethyl) -2- ((2-methoxy-5-methyl-4- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-alkyl) phenyl ] amino)) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd (OAc) was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (80 mg, 0.199mmol) and 2-methoxy-5-methyl-4- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) aniline (69.0 mg,0.298 mmol) in 1, 4-dioxane (2 mL), respectively 2 (9 mg,0.04 mmol), BINAP (50 mg,0.08 mmol) and Cs 2 CO 3 (195 mg,0.579 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered with suction and concentrated to give the crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give N- (3- ((6- (difluoromethyl) -2- ((2-methoxy-5-methyl-4- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-alkyl) phenyl)]Amino group) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonamide. 1 HNMR(400MHz,DMSO-d 6 )8.82(s,1H),8.54(d,J=4Hz,1H),8.48(d,J=4Hz,1H),7.99(s,1H),7.93(s,1H),8.22(s,1H),7.14(t,J=64.0Hz,1H),6.90(s,1H),6.68(s,1H),3.78(s,3H),3.25(s,3H),3.17(s,5H),2.75(s,2H),2.42(s,3H),2.36(s,2H),2.07(s,3H),1.90(s,1H)。MS m/z(ESI):599.2[M+H] +
Example 16
Preparation of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-alkyl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-alkyl) amino) -3-methoxy-N-methylbenzamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd (OAc) was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (100 mg,0.248 mmol) and 4-amino-3-methoxy-N-methylbenzamide (67.1 mg,0.372 mmol) in 1, 4-dioxane (3 mL), respectively 2 (12 mg,0.05 mmol), BINAP (62 mg,0.099 mmol) and Cs 2 CO 3 (242 mg,0.774 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and concentrated to give crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-alkyl) amino) -3-methoxy-N-methylbenzamide 45mg. 1 HNMR(400MHz,DMSO-d 6 )8.90(s,1H),8.55(d,J=2.56Hz,1H),8.47(d,J=4.00Hz,1H),8.31-8.26(m,1H),8.09(s,1H),7.47(d,J=2.00Hz,1H),7.37-7.34(m,1H),7.21(d,J=52Hz,1H),5.96(s,1H),3.92(s,3H),3.30(s,3H),2.78(d,J=4.00Hz,1H)。MS m/z(ESI):547.1[M+H] +
Example 17
Preparation of (4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-alkyl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N- (1-methylpiperidin-4-yl) benzamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd (OAc) was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (40 mg,0.099 mmol) and 4-amino-3-methoxy-N- (1-methyl-piperidin-4-yl) benzamide (35.0 mg,0.149 mmol) in 1, 4-dioxane (1 mL), respectively 2 (5 mg,0.02 mmol), BINAP (25 mg,0.04 mmol) and Cs 2 CO 3 (97 mg,0.298 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered with suction and concentrated to give the crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-alkyl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -3-methoxy-N- (1-methylpiperidin-4-yl) benzamide 23mg. 1 HNMR(400MHz,DMSO-d 6 )8.90(s,1H),8.52(m,1H),8.45(d,J=4Hz,1H),8.31(d,J=12Hz,1H),8.22(m,1H),8.09(s,1H),7.45(m,1H),7.39-7.37(m,1H),7.18(t,J=56Hz,1H),5.79(s,2H),3.90(s,3H),3.27(s,3H),3.18(s,3H),1.94-1.87(m,2H),1.75-1.72(m,2H),1.27(m,1H),1.24-1.20(m,5H),0.85-0.70(m,2H).MS m/z(ESI):630.2[M+H] +
Example 18
Preparation of 7- ((6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide
Step 1: synthesis of 2-chloro-5-iodo-N- (4-methoxybenzyl) pyrimidin-4-amine
At 0 ℃, PMBNH 2 A solution of (5.00 g,36.4 mmol), 2, 4-dichloro-5-iodopyrimidine (10.0 g,36.4 mmol) and DIPEA (9.00 mL,54.6 mmol) in DMF (100 mL) was stirred for 2h. TLC (petroleum ether/ethyl acetate=5/1) showed that after the reaction was completed, the reaction mixture was poured into water (200 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed with saturated brine (100 mL 3 times with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and concentrated to give crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give 12.0g of 2-chloro-5-iodo-N- (4-methoxybenzyl) pyrimidin-4-amine. MS m/z (ESI): 375.9[ M+H ]] +
Step 2: synthesis of 2-chloro-5- (3, 3-diethoxy-1-propyl-1-alkyl) -N- (4-methoxybenzyl) pyrimidin-4-amine
2-chloro-5-iodo-N- (4-methoxybenzyl) pyrimidin-4-amine (12.0 g,31.95 mmol) and 3, 3-diethoxypropyne (6.41 g,50.0 mmol) were added to DMF (120 mL), pd (PPh) was added to the mixture at 25 ℃ 3 )Cl 2 (2.34g,3.20mmol)、CuI(1.27g,6.66mmol)、PPh 3 (436 mg,1.66 mmol) and TEA (10 mL,100 mmol). The reaction mixture was then stirred under nitrogen at 60 ℃ for 2h. LCMS showed that after the reaction was complete, the reaction mixture was poured into water (200 mL) and extracted 2 times with ethyl acetate (100 mL). The combined organic phases were washed 3 times with brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and evaporated to give a residue. The residue passes throughFlash column chromatography (petroleum ether/ethyl acetate=10/1) afforded 2-chloro-5- (3, 3-diethoxy-1-propyl-1-yl) -N- (4-methoxybenzyl) pyrimidin-4-amine 13.0g. MS m/z (ESI): 376.0[ M+H ]] +
Step 3: synthesis of 2-chloro-6- (diethoxymethyl) -7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ] pyrimidine
To a solution of 2-chloro-5- (3, 3-diethoxy-1-propyl-1-yl) -N- (4-methoxybenzyl) pyrimidin-4-amine (13.0 g, crude) in THF (50 mL) at 25℃was added TBAF (1M, 105mL,105 mmol). The reaction mixture was stirred at 60℃for 1h. TLC (petroleum ether/ethyl acetate=5/1) showed that after the reaction was completed, the reaction was concentrated and then dissolved with EtOAc (250 mL). The above solution was washed 3 times with water (100 mL) and then 1 time with saturated brine (150 mL). Anhydrous Na for organic phase 2 SO 4 Drying, then filtering and evaporating the dried product to obtain a crude compound. The crude compound was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give 2-chloro-6- (diethoxymethyl) -7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d]7.93g of pyrimidine. MS m/z (ESI): 376.0[ M+H ]] +
Step 4: synthesis of 2-chloro-7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ] pyrimidine-6-carbaldehyde
To 2-chloro-6- (diethoxymethyl) -7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ] at 25 ℃]To a solution of pyrimidine (7.93 g,21.1 mmol) in 1, 4-hexacyclic ring (80 mL) was added 6M hydrochloric acid (36 mL). The reaction solution was then stirred at 25℃for 1h. LCMS showed that after the reaction was complete, the reaction was poured into water (100 mL) and extracted 2 times with EtOAc (150 mL). The combined organic phases were washed with brine (200 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give 2-chloro-7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ]]Pyrimidine-6-carbaldehyde (5.34 g, crude). MS M/Z (ESI): 302.0 (M+H) +
Step 5: synthesis of 2-chloro-6- (difluoromethyl) -7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ] pyrimidine
To 2-chloro-7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ] at 0deg.C]PyrimidineDAST (7.00 mL,53.1 mmol) was added dropwise to a solution of 6-carbaldehyde (5.34 g, crude) in DCM (50 mL). After the completion of the dropwise addition, the reaction mixture was heated to 25℃and stirred for 2 hours. LCMS showed that after the reaction was completed, the reaction mixture was slowly quenched with water (100 mL) and extracted 2 times with DCM (80 mL). The organic phases were combined and washed with saturated brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. Concentrating the dried product to obtain a crude compound. The crude compound was purified by flash column chromatography (petroleum ether/ethyl acetate=10/1) to give 2-chloro-6- (difluoromethyl) -7- (4-methoxybenzyl) -7H-pyrrolo [2, 3-d)]4.50g of pyrimidine. MS m/z (ESI) + :324.0[M+H] + . Step 6: synthesis of 2-chloro-6- (difluoromethyl) -7H-pyrrolo [2,3-d]Pyrimidine
To 2-chloro-7- (4-methoxybenzyl) -7H-pyrrolo [2,3-d ] at 0deg.C]Pyrimidine-6-carbaldehyde (4.50 g,13.9 mmol) in ACN/H 2 O=5: to the mixture in 1 (60 mL), CAN (56 g,992 mmol) was added. The mixture was then stirred at 0 ℃ for 12 hours. LCMS showed that after the reaction was complete, the reaction mixture was quenched with water (100 mL) and slowly extracted 2 times with DCM (80 mL). The combined organic phases were washed with brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by FCC (petroleum ether/ethyl acetate=10/1) to give 2-chloro-6-difluoromethyl-7H-pyrrolo [2,3-d]Pyrimidine 2.0g, MS m/z (ESI): 204.02 (M+H) +
Step 7: synthesis of (3- (methylsulfonyl) phenyl) methanol
To a mixture of methyl 3- (methylsulfonyl) benzoate (4.0 g,18.6 mmol) in THF (60 mL) was added LAH (780 mg,20.5 mmol) at 25 ℃. The mixture was then stirred at 25 ℃ for 12 hours. LCMS showed that after the reaction was complete, the reaction mixture was quenched with water (100 mL) and slowly extracted 2 times with DCM (80 mL). The combined organic phases were washed with brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by FCC (petroleum ether/ethyl acetate=5/1) to give (3- (methylsulfonyl) phenyl) methanol 3.0g, ms m/z (ESI):186.0[M+H] +
Step 8: synthesis of 1- (chloromethyl) -3- (methylsulfonyl) benzene
To methyl (3- (methylsulfonyl) phenyl) methanol (3.0 g 16.1 mmol) at 25℃in CH 2 Cl 2 SOCl was added to the mixture in (20 mL) 2 (2.3 g,20.5 mmol). The mixture was then stirred at 25 ℃ for 12 hours. LCMS showed that after the reaction was complete, the reaction mixture was quenched with water (100 mL) and slowly extracted 2 times with DCM (80 mL 2). The combined organic phases were washed with brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by FCC (petroleum ether/ethyl acetate=5/1) to give 2.8g of 1- (chloromethyl) -3- (methylsulfonyl) benzene, ms m/z (ESI): 186.0[ M+H ]] +
Step 9: synthesis of 2-chloro-6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidine
2-chloro-6- (difluoromethyl) -7H-pyrrolo [2,3-d]Pyrimidine (2.0 g,9.8 mmol) and 1- (chloromethyl) -3- (methylsulfonyl) benzene (1.98 g), dissolved in ACN (20 mL), was added K 2 CO 3 (2.7 g,19.6 mmol) and then stirred at 45℃for 8 hours. LCMS showed that after the reaction was complete, the reaction mixture was quenched with water (100 mL) and slowly extracted 2 times with DCM (80 mL). The combined organic phases were washed with brine (100 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by FCC (petroleum ether/ethyl acetate=3/1) to give 2-chloro-6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d]Pyrimidine 2.8g, MS m/z (ESI): 371.02[ M+H ]] +
Step 10: synthesis of 7- ((6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide
To 2-chloro-6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ] at 120 DEG C]Pyrimidine (100 mg,0.27 mmol) and 7-amino-N-mixturesTo methyl 2, 3-dihydrobenzofuran-4-carboxamide (77.8 mg,0.41 mmol) in 1, 4-dioxane (3 mL) was added Cs 2 CO 3 (260mg,0.81mmol)、Pd(OAc) 2 (12 mg,0.054 mmol) and BINAP (67 mg,0.11 mmol). The mixture was then stirred at 120℃for 8 hours. LCMS showed that after the reaction was complete, the reaction mixture was cooled and quenched with water (100 mL) and slowly extracted 2 times with EA (80 mL). The organic phases were combined and washed with brine (100 mL) followed by anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by FCC (petroleum ether/ethyl acetate=3/1) to give 7- ((6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide 30mg. 1 HNMR(400MHz,DMSO-d 6 )8.91(s,1H),8.32(d,J=8Hz,1H),8.26-8.23(m,1H),8.14(s,1H),7.83-7.77(m,2H),7.60-7.56(m,1H),7.45-7.38(m,3H),7.31(t,J=52Hz,1H),7.00(m,1H),5.60(s,2H),3.12(s,3H),2.76(d,J=8Hz,3H).MS m/z(ESI):527.02[M+H] +
Example 19
Preparation of 4- ((6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide
Under the protection of nitrogen, 2-chloro-6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2,3-d ]Pyrimidine (100 mg,0.27 mmol) and 4-amino-3-methoxy-N-methylbenzamide (69 mg,0.41 mmol) were dissolved in 1, 4-dioxane (3 mL), and Cs was added 2 CO 3 (260mg,0.81mmol)、Pd(OAc) 2 (12 m,0.054 mmol) and BINAP (67 mg,0.11 mmol), and then the reaction system was slowly warmed to 120℃and stirred for 8 hours. LCMS showed that after the reaction was complete, the reaction mixture was cooled to room temperature and quenched with water (100 mL) and slowly extracted with ethyl acetate (80 mL)Taking 2 times. The organic phases were combined and washed with brine (100 mL) followed by anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and evaporated to give a residue. The residue was purified by FCC (petroleum ether/ethyl acetate=1/1) to give 4- ((6- (difluoromethyl) -7- (3- (methylsulfonyl) benzyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -3-methoxy-N-methylbenzamide 75mg. 1 HNMR(400MHz,DMSO-d 6 )8.85(s,1H),8.33(s,1H),8.05-8.01(m,1H),7.83-7.74(m,3H),7.60(t,J=8Hz,1H),7.41-7.38(m,3H),7.28(t,J=56Hz,1H),7.10(d,J=8Hz,1H),6.97-6.96(m,1H),4.49(t,J=8Hz,1H),3.40(t,J=8Hz,2H),2.72(d,J=8Hz,3H).MS m/z(ESI):515.02[M+H] +
Example 20
Preparation of 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide method
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd (OAc) was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (100 mg,0.248 mmol) and 7-amino-N-methyl-2, 3-dihydrobenzofuran-4-carboxamide (71.5 mg,0.372 mmol) in 1, 4-dioxane (3 mL), respectively 2 (12 mg,0.05 mmol), BINAP (62 mg,0.099 mmol) and Cs 2 CO 3 (242 mg,0.774 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered with suction and concentrated to give the crude compound. Purification of the crude product by flash column chromatography (petroleum ether/ethyl acetate=0/1) gives 7- ((6- (difluoromethyl) -7- ((3)- (N-methyl-sulfamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d]Pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide 76mg. 1 HNMR(400MHz,DMSO-d 6 )8.90(s,1H),8.55(d,J=2.56Hz,1H),8.84(s,1H),8.55-8.48(m,2H),8.22(s,1H),8.10(m,1H),7.76(d,J=8.00Hz,1H),7.32(d,J=52Hz,1H),7.08(d,J=8Hz,1H),6.92(s,1H),5.76(s,2H),4.53(t,J=12.0Hz,2H),3.42(t,J=8.00Hz,2H),3.33(s,3H),3.25(s,3H),2.74(d,J=4.00Hz,3H)。MS m/z(ESI):559.1[M+H] +
Example 21
Preparation of 4- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2-fluoro-5-methoxy-N- (1-methylpiperidin-4-yl) benzamide
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (100 mg,0.25 mmol) and 4-amino-2-fluoro-5-methoxy-N- (1-methylpiperidin-4-yl) benzamide (88.6 mg,0.3 mmol) in 1, 4-dioxane (10 mL), respectively 2 (dba) 3 (23.8 mg,0.025 mmol), BINAP (31.2 mg,0.05 mmol), and tBuONa (48.0 mg,0.5 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (20 mL). The combined organic phases were washed with saturated brine (20 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered with suction and concentrated to give the crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/100) to give 56mg of 4-amino-2-fluoro-5-methoxy-N- (1-methylpiperidin-4-yl) benzamide. 1 HNMR(400MHz,DMSO-d 6 )δ8.95(s,1H),8.55(d,J=2.4Hz,1H),8.47(d,J=2.4Hz,1H),8.24(d,J=13.1Hz,1H),8.19(s,1H),7.94(d,J=5.1Hz,1H),7.33–7.07(m,2H),6.99(s,1H),5.83(s,2H),3.90(s,3H),3.78(s,1H),3.30(s,5H),3.19(s,4H),2.91(s,2H),2.33(s,5H),1.83(d,J=10.6Hz,2H),1.62(dd,J=21.2,10.8Hz,2H)。MS m/z(ESI):648.2[M+H] +
Example 22
Preparation of 7- ((6-formyl-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide
Step 1: synthesis of 4-bromo-2, 3-dihydrobenzofuran-7-amine
2, 3-Dihydrobenzofuran-7-amine (5 g,40.0 mmol) was dissolved in DMF (60 mL), NBS (7.2 g,40.7 mmol) was added in portions, the reaction was carried out at room temperature for 3h, water was added after completion of the reaction, dichloromethane was added for extraction, the extract was dried and concentrated, and 4-bromo-2, 3-dihydrobenzofuran-7-amine 7.2g was obtained by column chromatography, MS m/z (ESI): 214.0[ M+H ] ] +
Step 2: synthesis of 7-amino-2, 3-dihydrobenzofuran-4-carboxylic acid ethyl ester
Dissolving 4-bromo-2, 3-dihydrobenzofuran-7-amine (3.5 g,16.28 mmol), cobaltosic octacarbonyl (5.5 g,16.28 mmol), palladium acetate (0.18 g,0.81 mmol), xantPhos (0.94 g,1.63 mmol) and DMAP (7.9 g,65.12 mmol) in toluene (26 mL) and ethanol (9 mL), reacting at 105 ℃ for 12h under nitrogen, monitoring the reaction completion, adding water, extracting with ethyl acetate, drying the concentrated extract, purifying by column chromatography to obtain ethyl 7-amino-2, 3-dihydrobenzofuran-4-carboxylate 1.84g, MS m/z (ESI): 208.1[ M+H ]] +
Step 3: synthesis of 7-amino-2, 3-dihydrobenzofuran-4-carboxylic acid
7-amino-2, 3-Dihydrobenzofuran-4-carboxylic acid ethyl ester (500 mg,2.42 mmol) was dissolved in THF (10 mL) and water (10 mL), naOH (290 mg,7.25 mmol) was added, the reaction was carried out at 60℃overnight, and the reaction was concentrated after completion of the reactionThe pH of the solution was adjusted to be weakly acidic by adding a 2M hydrochloric acid solution, followed by suction filtration to obtain 340mg of 7-amino-2, 3-dihydrobenzofuran-4-carboxylic acid. MS m/z (ESI) 180.0[ M+H ]] +
Step 4: synthesis of 7-amino-N-methyl-2, 3-dihydrobenzofuran-4-carboxamide
7-amino-2, 3-dihydrobenzofuran-4-carboxylic acid (340 mg,1.90 mmol), HATU (866 mg,2.28 mmol) and methylamine hydrochloride (256 mg,3.80 mmol) were dissolved in DMF (5 mL), DIPEA (1.26 mL,7.60 mmol) was added, the reaction was allowed to proceed at room temperature for 12h, water was added after completion of the reaction, dichloromethane was added to extract, the concentrated extract was dried and column chromatography was performed to give 3-methoxy-N-methyl-4-nitrobenzamide 300mg, MS m/z (ESI): 193.1[ M+H ] ] +
Step 5: synthesis of 7- ((6- (Diethoxymethyl) -7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide
N- (3- ((2-chloro-6- (diethoxymethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methylmethanesulfonamide (220 mg,0.48 mmol), 3-methoxy-N-methyl-4-nitrobenzamide (140 mg,0.73 mmol), BINAP (60 mg,0.10 mmol), pd 2 (dba) 3 (45 mg,0.05 mmol) and sodium t-butoxide (94 mg,0.97 mmol) were dissolved in dioxane (22 mL), nitrogen protected, reacted for 4.5H at 100deg.C, monitored for completion, water was added, ethyl acetate was added to extract, the concentrated extract was dried and purified by column chromatography to give 7- ((6- (diethoxymethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide 210mg, MS m/z (ESI): 611.2[ M+H ]] +
Step 6: synthesis of 7- ((6-formyl-7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide
7- ((6- (Diethoxymethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofurans-4-carboxamide (210 mg,0.34 mmol) was dissolved in dioxane (2 mL) and water (2 mL), TFA (1 mL) was added under ice-bath, reaction was carried out at room temperature for 1H, water was added after monitoring the reaction was complete, dichloromethane was added for extraction, the concentrated extract was dried and purified by column chromatography to give 7- ((6-formyl-7- ((3- (N-methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-methyl-2, 3-dihydrobenzofuran-4-carboxamide 27mg. 1 H NMR(500MHz,DMSO)δ9.58(s,1H),8.98(s,1H),8.75(s,1H),8.47(d,J=2.2Hz,1H),8.39(d,J=2.3Hz,1H),8.12(d,J=4.6Hz,1H),7.66(d,J=8.3Hz,1H),7.51(s,1H),7.08(d,J=8.3Hz,1H),5.87(s,2H),4.48(t,J=8.7Hz,2H),3.39(t,J=8.6Hz,2H),3.32(s,3H),3.15(s,3H),2.71(d,J=4.5Hz,3H).MS m/z(ESI):537.1[M+H] +
Example 23
Preparation of 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-ethyl-2, 3-dihydrobenzofuran-4-carboxamide
Step 1: synthesis of 4-methyl-7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylate
To N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d) under nitrogen]Pd was added to a solution of pyrimidine-7-acyl-methyl) pyrazin-2-alkyl) -N-methylmethanesulfonamide (540 mg,1.35 mmol) and ethyl 7-amino-2, 3-dihydrobenzofuran-4-carboxylate (335.3 mg,1.62 mmol) in 1, 4-dioxane (30 mL), respectively 2 (dba) 3 (128.2 mg,0.14 mmol), BINAP (168.0 mg,0.27 mmol) and tBuONa (259.5 mg,2.74 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (50mL) and then using anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and concentrated to give crude compound. Flash column chromatography (DCM/meoh=0-10%) purification yielded methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylate 320mg, MS m/z (ESI): 560.1[ M+H ]] +
Step 2: synthesis of methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylic acid
To methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) under ambient conditions]Pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylate (320 mg,0.56 mmol) in methanol/water (10 mL/20 mL) was added LiOH H 2 O (40.1 mg,1.68 mmol). The reaction was then stirred overnight at 50 ℃. TLC (DCM/meoh=10:1) showed that after the reaction was complete the reaction was concentrated and taken up with H 2 O (20 mL) was dissolved. 1M HCl adjusted solution Ph to 2, with yellow solid precipitated, crude filtered, purified by flash column chromatography (DCM/MeOH=from 100/1 to 10/1) to give methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylic acid 280mg, MS m/z (ESI): 546.1[ M+H ]] +
Step 3: synthesis of 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-ethyl-2, 3-dihydrobenzofuran-4-carboxamide
The compound methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylic acid (60.0 mg,0.12 mmol) and ethylamine hydrochloride (30.1 mg,0.37 mmol) were dissolved in DMF (10 mL), HATU (91.3 mg,0.24 mmol) and DIPEA (0.12 mL,0.72 mmol) were added to the reaction, then reacted at room temperature for 2h, and after completion of the LCMS monitoring The reaction was quenched with water (10 mL) then extracted 3 times with EA (10 mL), and the combined organic phases were washed 1 time with saturated NaCl and with anhydrous Na 2 SO 4 The organic phase was dried and the dried product was concentrated by filtration and purified by column chromatography on a concentrate (MeOH/dcm=from 0 to 10%) to give 7- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-ethyl-2, 3-dihydrobenzofuran-4-carboxamide 32.0mg. 1 H NMR(400MHz,DMSO-d 6 )δ8.84(s,1H),8.55(d,J=2.4Hz,1H),8.48(d,J=2.4Hz,1H),8.23(s,1H),8.13(t,J=5.6Hz,1H),7.75(d,J=8.4Hz,1H),7.31–7.05(m,2H),6.92(s,1H),5.75(s,2H),4.52(t,J=8.8Hz,2H),3.42(t,J=8.8Hz,2H),3.26–3.21(m,5H),3.19(s,3H),1.10(t,J=7.2Hz,3H).MS m/z(ESI):573.2[M+H] +
Example 24
Preparation of 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-methoxy-2, 3-dihydrobenzofuran-4-carboxamide
The compound methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylic acid (50 mg,0.09 mmol), O-methylhydroxylamine hydrochloride (23.4 mg,0.28 mmol), HATU (68.5 mg,0.18 mmol), DMAP (1.0 mg,0.01 mmol) and DIPEA (0.1 mL,0.54 mmol) were added to DMF (6 mL) and then reacted at room temperature for 2h, after completion of LCMS monitoring the reaction, the reaction was quenched with water (10 mL) and extracted 2 times with EA (20 mL), the organic phase was washed 1 time with saturated aqueous NaCl solution and then with anhydrous Na 2 SO 4 Drying, filtering, concentrating the dried product, and subjecting the concentrate to column chromatography to obtain 7- ((6- (difluoromethyl) -7- ((3- (N-methyl sulfonamide) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N-methoxy-2, 3-dihydrobenzenesAnd 25mg of furan-4-carboxamide. 1 H NMR(400MHz,DMSO-d 6 )δ11.39(s,1H),8.84(s,1H),8.55(d,J=2.3Hz,1H),8.48(d,J=2.4Hz,1H),8.27(s,1H),7.77(d,J=8.4Hz,1H),7.19(t,J=53.3Hz,1H),6.98(d,J=8.5Hz,1H),6.93(s,1H),5.76(s,2H),4.54(t,J=8.8Hz,2H),3.68(s,3H),3.41(t,J=8.6Hz,2H),3.26(s,3H),3.19(s,3H).MS m/z(ESI):575.2[M+H] +
Example 25
Preparation of 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N-isobutyl-2, 3-dihydrobenzofuran-4-carboxamide
The compound methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylic acid (60.0 mg,0.11 mmol), isobutylamine (24.1 mg,0.33 mmol), HATU (83.7 mg,0.22 mmol), DMAP (1.5 mg,0.01 mmol) and DIPEA (0.12 mL,0.66 mmol) were added to DMF (6 mL) and then reacted at room temperature for 2h, after completion of LCMS monitoring the reaction, the reaction was quenched with water (10 mL), extracted 3 times with EA (10 mL), the organic phase was washed with saturated aqueous NaCl solution, then with anhydrous Na 2 SO 4 Dried, and the dried product concentrated by filtration, and finally purified by column chromatography on a concentrate (MeOH/dcm=0-10%) to give 7- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) ]Pyrimidin-2-yl) amino) -N-isobutyl-2, 3-dihydrobenzofuran-4-carboxamide 35.1mg. 1 H NMR(400MHz,DMSO-d 6 )δ8.83(s,1H),8.54(d,J=2.3Hz,1H),8.48(d,J=2.4Hz,1H),8.26(s,1H),8.14(t,J=5.8Hz,1H),7.74(d,J=8.4Hz,1H),7.25(t,J=53.3Hz,53.3Hz,1H),7.08(d,J=8.4Hz,1H),6.92(s,1H),5.75(s,2H),4.52(t,J=8.7Hz,2H),3.41(t,J=8.7Hz,2H),3.24(s,3H),3.18(s,3H),3.03(t,J=6.4Hz,2H),1.81(dp,J=13.7,6.9Hz,1H),0.88(d,J=6.7Hz,6H).MS m/z(ESI):601.2[M+H] +
Example 26
Preparation of 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -N- (1-methylpiperidin-4-yl) -2, 3-dihydrobenzofuran-4-carboxamide
The compound methyl 7- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxylic acid (100 mg,0.18 mmol), N-methylpiperidin-4-amine (62.8 mg,0.55 mmol), HATU (137.0 mg,0.36 mmol), DMAP (2.4 mg,0.018 mmol) and DIPEA (0.17 mL,1.08 mmol) were added to DMF (10 mL) and then reacted at room temperature for 2h after completion of LCMS monitoring the reaction, quenched with water (10 mL), extracted 3 times with EA (20 mL), the organic phase was washed with saturated brine, then with anhydrous Na 2 SO 4 The organic phase was dried, the dried product was concentrated by filtration and purified by column chromatography of the concentrate (MeOH/dcm=from 0 to 10%) to give 7- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -N- (1-methylpiperidin-4-yl) -2, 3-dihydrobenzofuran-4-carboxamide 64.0mg. 1 H NMR(400MHz,DMSO-d 6 )δ8.83(s,1H),8.54(d,J=2.4Hz,1H),8.48(d,J=2.4Hz,1H),8.26(s,1H),8.01(d,J=7.6Hz,1H),7.76(d,J=8.4Hz,1H),7.30–7.05(m,2H),6.92(s,1H),5.75(s,2H),4.52(t,J=8.8Hz,2H),3.75(s,1H),3.40(t,J=8.8Hz,3H),3.24(s,3H),3.18(s,3H),2.93–2.83(m,2H),2.33–2.13(m,5H),1.78(d,J=10.8Hz,2H),1.61(d,J=9.9Hz,2H)。MS m/z(ESI):642.2[M+H] +
Example 27
Preparation of N-methyl-4- ((7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) benzamide
Step 1: synthesis of 3- (N-methyl-methylsulfonyl) pyrazine-2-carboxylic acid
The compound N- (3-cyanopyrazin-2-yl) -N-methylmethanesulfonamide (3 g,14.15 mmol) was dissolved in EtOH (30 ml), 10% NaOH (60 ml) was added, the reaction solution was refluxed at 100℃for 16h by condensation, the reaction solution was dried by LCMS after monitoring the reaction was completed, concentrated hydrochloric acid was added to adjust the pH to 5, DCM (100 ml) was extracted 6 times, the organic phase was washed with saturated NaCl, the organic phase was dried over anhydrous sodium sulfate, and the dried product was concentrated by filtration to give 2.5g of a pale yellow oily crude product, 3- (N-methylmethanesulfonamido) pyrazine-2-carboxylic acid. MS m/z (ESI) 232.0[ M+H ]] +
Step 2: synthesis of methyl 3- (N-methylsulfonyl) pyrazine-2-carboxylate
Compound 3- (N-methylmethanesulfonamido) pyrazine-2-carboxylic acid (1 g,4.32 mmol) was dissolved in DMF (20 ml) followed by K 2 CO 3 (1.19 g,8.64 mmol), meI (2.45 g,7.28 mmol), reaction at 25℃for 1h, LCMS monitoring reaction completion, reaction was diluted with 20mL of water, extracted 4 times with EtOAc (50 mL), combined organic phases washed 1 time with saturated NaCl, then dried over anhydrous sodium sulfate, the dried product was concentrated by filtration and the concentrate was purified by column chromatography (PE: EA=1:1) to give 500mg of methyl 3- (N-methylsulfonamidyl) pyrazine-2-carboxylate as a pale yellow oil. MS m/z (ESI): 246.1[ M+H ] ] +
Step 3: synthesis of N- (3- (hydroxymethyl) pyrazin-2-yl) -N-methylmethanesulfonamide
The compound methyl 3- (N-methylmethanesulfonamido) pyrazine-2-carboxylate (1.0 g,4.1 mmol) was dissolved in THF (20 ml) at 0deg.C, naBH was added 4 (1.19 g,8.64 mmol), heating the reaction solution to 25deg.C, reacting for 1h, LCMS monitoring, diluting the reaction solution with methanol (10 ml), extracting with EtOAc (10 ml) for 3 times, washing the organic phase with saturated NaCl, drying the organic phase with anhydrous sodium sulfate, filtering and concentrating the dried product, and introducingThe concentrate was purified by column chromatography (PE: ea=4:1) to give 270mg of N- (3- (hydroxymethyl) pyrazin-2-yl) -N-methylmethanesulfonamide as a pale yellow oily solid. MS m/z (ESI): 218.0[ M+H ]] +
Step 4: synthesis of N- (3- (chloromethyl) pyrazin-2-yl) -N-methylmethanesulfonamide
The compound N- (3- (hydroxymethyl) pyrazin-2-yl) -N-methylmethanesulfonamide (270 mg,1.23 mmol) was dissolved in DCM (5 ml) at 0deg.C and SOCl was slowly added dropwise 2 (0.26 ml,3.7 mmol) and heating the reaction to 25deg.C, stirring overnight, LCMS monitoring the reaction, directly steaming the reaction solution, adding DCM (10 ml) for dilution, and adding saturated NaHCO 3 (10 mL) of the solution was extracted, the organic phase was separated, then washed with saturated NaCl (10 mL), and the organic phase was dried over anhydrous sodium sulfate, and the dried product was concentrated by filtration, and the concentrate was purified by column chromatography (PE: EA=4:1) to give 170mg of N- (3- (chloromethyl) pyrazin-2-yl) -N-methylmethanesulfonamide as a pale yellow oily solid. MS m/z (ESI) 236.0[ M+H ] ] +
Step 5: synthesis of 2-chloro-7- (((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2,3-d ] pyrimidine)
Sodium hydride (1.95 g,48.8 mmol) was added to 2-chloro-7H-pyrrolo [2,3-d ] in an ice water bath]Pyrimidine (5.0 g,32.5 mmol) in THF (50 mL). After stirring at 0℃for 30 minutes, 2- (trisilyl) ethoxymethyl chloride (6.5 g,39.0 mmol) was added thereto. Then, the reaction apparatus was allowed to stand at room temperature and stirred for 2 hours, and after the disappearance of the starting material was detected by TLC, ice water (10 mL) was added to the reaction system to separate an organic phase. The aqueous phase was extracted 3 times with ethyl acetate (50 mL). The combined organic phases were backwashed with water (50 mL) and the organic phases were dried over anhydrous sodium sulfate, the dried product was filtered, concentrated and dried in vacuo to give 2-chloro-7- (((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine (5.9 g), MS m/z (ESI): 284.1[ M+H ]] + . The compound was used directly in the next reaction without purification
Step 6: synthesis of 2-chloro-6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2,3-d ] pyrimidine
To a 250 mL three neck round bottom flask was added bis (pinacolato) diboron (3.9 g,15.4 mmol), 4-di-tert-butyl-2, 2-bipyridine (273.8 mg,1.02 mmol), 1, 5-cyclooctadiene methoxy iridium dimer (342.6 mg,0.51mmol and n-hexane (40 mL) in this order, after dissolving, stirring at 50℃for 10min, then 2-chloro-7- (((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2, 3-d) was added to the mixture ]A solution of pyrimidine (2.9 g,10.2 mmol) in tetrahydrofuran (12 mL). Subsequently, the reaction system was stirred at 90℃for 1 hour. After LC-MS monitored the disappearance of starting material, the heating was stopped. After the reaction mixture was cooled to room temperature, ice water (100 mL) was added to the reaction mixture to dilute it. The organic phase was separated and the aqueous phase was extracted 3 times with ethyl acetate (60 mL). The combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the resulting solid residue was purified by column chromatography over silica gel (eluent: petroleum ether/dichloromethane=2/1) to give 2-chloro-6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2,3-d]Pyrimidine 1.8g. MS m/z (ESI): 410.2[ M+H ]] +
Step 7: synthesis of 2-chloro-6- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2,3-d ] pyrimidine
To a 250mL three neck round bottom flask was added copper 2-thiophenecarboxylate (186.9 g,0.98 mmol), 1, 10-phenanthroline (353.2 mg,1.96 mmol), lithium hydroxide monohydrate (820.7 mg,19.6 mmol), 1- (trifluoromethyl) -1, 2-phenyliodic-3- (1) -one (4.0 g,9.8 mmol) and 2-chloro-6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrole [2,3-d ]Pyrimidine (3.9 g,11.74 mmol). Subsequently, the reaction apparatus was evacuated to replace nitrogen gas, and repeated three times. Then, methylene chloride (120 mL) was added. After the mixture was dissolved by stirring, the reaction mixture was stirred at 45℃for 10 hours. After the reaction solution was cooled to room temperature, it was concentrated under reduced pressure to obtain a solid residue, which was then separated and purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=1/1). The product was collected and concentrated under reduced pressure to give 2-chloro-6- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrroleAnd [2,3-d ]]Pyrimidine 802mg. MS m/z (ESI): 352.0[ M+H ]] +
Step 8: synthesis of 2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidine
The compound 2-chloro-6- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -7H-pyrrolo [2,3-d]Pyrimidine (0.82 g,2.03 mmol) was dissolved in dichloromethane (20 mL) and trifluoroacetic acid (20 mL) was added. Stirring was carried out at 60℃for 2 hours. Subsequently, the reaction solution was concentrated under reduced pressure. To the resulting brown residue was added methanol (40 mL), water (20 mL), and potassium carbonate (1.38 g,10.00 mmol). After dissolution by stirring, the mixture was stirred at 60℃for 2 hours. After the reaction solution was cooled to room temperature, it was concentrated under reduced pressure to remove methanol from the reaction solution. The resulting liquid was extracted 3 times with ethyl acetate (20 mL) and the organic phases were combined. The organic phase was backwashed 3 times with saturated brine (20 mL), then dried over anhydrous sodium sulfate, and finally concentrated in vacuo to give 2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] as a white solid ]Pyrimidine 0.68g, without purification, was used directly in the next reaction. MS m/z (ESI): 222.0[ M+H ]] +
Step 9: synthesis of N- (3- ((2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methylmethanesulfonamide
The compound 2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] is reacted at 45 DEG C]Pyrimidine (120 mg,0.54 mmol) and N- (3- (chloromethyl) pyrazin-2-yl) -N-methylmethanesulfonamide (153.0 mg,0.65 mmol) were dissolved in ACN (20 mL) and K was added 2 CO 3 (149.0 mg,1.08 mmol). The mixture was then stirred at 90℃for 8 hours. LCMS showed that after the reaction was complete, the reaction mixture was quenched with water (10 mL) and slowly extracted 2 times with DCM (10 mL). The combined organic phases were washed with brine (20 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried. The dried product was filtered and concentrated and the concentrate was purified by FCC (petroleum ether/ethyl acetate=3/1) to give N- (3- ((2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methyl-methanesulfonamide 100mg, MS m/z (ESI): 421.0[ M+H ]] +
Step 10: synthesis of N-methyl-4- ((7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) benzamide
Under the protection of nitrogen, N- (3- ((2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2, 3-d)]Pd was added to a solution of pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methyl methanesulfonamide (130 mg,0.31 mmol) and 4-aminobenzamide (69.5 mg,0.46 mmol) in 1, 4-dioxane (15 mL), respectively 2 (dba) 3 (27.5 mg,0.03 mmol), BINAP (37.3 mg,0.06 mmol) and tBuONa (59.6 mg,0.62 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (30 mL). The combined organic phases were washed with saturated brine (20 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered with suction and concentrated to give the crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give N-methyl-4- ((7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -6- (trifluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) benzamide 54mg. 1 HNMR(400MHz,DMSO-d 6 )δ10.02(s,1H),8.98(s,1H),8.58(d,J=2.4Hz,1H),8.48(d,J=2.4Hz,1H),8.22(q,J=4.3Hz,1H),7.73(d,J=8.9Hz,2H),7.68(d,J=8.9Hz,2H),7.24(s,1H),5.83(s,2H),3.32(s,3H),3.22(s,3H),2.75(d,J=4.5Hz,3H).MS m/z(ESI):535.1[M+H] +
Example 28
Preparation of N-methyl-7- ((7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -6- (trifluoromethyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxamide
Under the protection of nitrogen, N- (3- ((2-chloro-6- (trifluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl) pyrazin-2-yl) -N-methyl methanesulfonatePd was added to a solution of amide (100 mg,0.24 mmol) and 7-amino-N-methyl-2, 3-dihydrobenzofuran-4-carboxamide (69.1 mg,0.36 mmol) in 1, 4-dioxane (10 mL), respectively 2 (dba) 3 (21.9 mg,0.024 mmol), BINAP (29.8 mg,0.048 mmol) and tBuona (46.1 mg,0.48 mmol). Then, the reaction solution was stirred at 120℃for 12 hours under nitrogen protection. LCMS showed that after the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted 2 times with ethyl acetate (10 mL). The combined organic phases were washed with saturated brine (20 mL) and then with anhydrous Na 2 SO 4 The organic phase is dried, and the dried product is filtered and concentrated to obtain crude compound. The crude product was purified by flash column chromatography (petroleum ether/ethyl acetate=0/1) to give N-methyl-7- ((7- ((3- (N-methyl methylsulfonylamino) pyrazin-2-yl) methyl) -6- (trifluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -2, 3-dihydrobenzofuran-4-carboxamide 46mg. 1 HNMR(400MHz,DMSO-d 6 )δ8.90(s,1H),8.57(d,J=2.4Hz,1H),8.49(d,J=2.4Hz,1H),8.45(s,1H),8.11(q,J=4.3Hz,1H),7.64(d,J=8.4Hz,1H),7.21(s,1H),7.06(d,J=8.4Hz,1H),5.74(s,2H),4.50(t,J=8.8Hz,2H),3.41(t,J=8.8Hz,2H),3.25(s,3H),3.19(s,3H),2.74(d,J=4.5Hz,3H)。MS m/z(ESI):577.2[M+H] +
Example 29
Preparation of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2-fluoro-N-isobutoxy-5-methoxybenzamide
Step 1: synthesis of methyl 4-amino-2-fluoro-5-methoxybenzoate
4-amino-2-fluoro-5-methoxybenzoic acid (500 mg,2.70 mmol) was dissolved in THF (4 mL) and methanol (1 mL), 2M (2 mL,4.05 mmol) trimethylsilane (2 mL,4.05 mmol) was added dropwise under ice bath, reacted at room temperature for 3h, the completion of the reaction was monitored, the acetic acid quenching reaction was added, and the reaction was concentratedColumn chromatography of the reaction solution to give methyl 4-amino-2-fluoro-5-methoxybenzoate 506mg, MS m/z (ESI) 200.0[ M+H ]] +
Step 2: synthesis of methyl 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2-fluoro-5-methoxybenzoate
N- (3- ((2-chloro-6- (difluoromethyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-7-yl) methyl-pyrazin-2-yl) -N-methylmethanesulfonamide (180 mg,0.45 mmol), methyl 4-amino-2-fluoro-5-methoxybenzoate (106 mg,0.54 mmol), BINAP (56 mg,0.09 mmol), pd 2 (dba) 3 (41 mg,0.04 mmol) and sodium t-butoxide (86 mg,0.90 mmol) were dissolved in dioxane (18 mL), and reacted at 100℃for 4.5h under nitrogen, and after the completion of the reaction, the reaction mixture was poured into water (30 mL) and extracted 2 times with ethyl acetate (15 mL). The organic phases were combined and then washed with saturated brine (30 mL) and then with anhydrous Na 2 SO 4 The organic phase was dried, the dried product was filtered with suction and concentrated, and the concentrate was purified by column chromatography to give methyl 4- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -2-fluoro-5-methoxybenzoate 160mg, MS m/z (ESI) 566.2[ M+H ]] +
Step 3: synthesis of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2-fluoro-5-methoxybenzoic acid
Methyl 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidine-2-yl) amino) -2-fluoro-5-methoxybenzoate (160 mg,0.28 mmol) was dissolved in THF (5 mL) and water (5 mL), naOH (34 mg,0.85 mmol) was added, the reaction was carried out overnight at room temperature, the reaction mixture was concentrated after the completion of the reaction, 2M hydrochloric acid solution was then added to adjust the pH to weak acidity, the reaction mixture was concentrated, and 4- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d) was obtained by column chromatography]Pyrimidin-2-yl) amino) -2-fluoro-5-methoxybenzoic acid 80mg. MS m/z (ESI): 552.1[ M+H ]] +
Step 4: synthesis of 4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2,3-d ] pyrimidin-2-yl) amino) -2-fluoro-N-isobutoxy-5-methoxybenzamide
4- ((6- (difluoromethyl) -7- ((3- (N-methyl methylsulfonyl) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -2-fluoro-5-methoxybenzoic acid (80 mg,0.15 mmol), HATU (67 mg,0.17 mmol) and O-isobutylamine hydrochloride (37 mg,0.29 mmol) were dissolved in DMF (3 mL), DIPEA (0.1 mL,0.58 mmol) was added and reacted at room temperature for 12h, after completion of the reaction mixture was poured into water (30 mL) and extracted 2 times with ethyl acetate (15 mL). The organic phases were combined and then washed with saturated brine (30 mL) followed by anhydrous Na 2 SO 4 The organic phase was dried, the dried product was filtered with suction and concentrated, and the concentrate was chromatographed to give 4- ((6- (difluoromethyl) -7- ((3- (N-methylsulfonamido) pyrazin-2-yl) methyl) -7H-pyrrolo [2, 3-d)]Pyrimidin-2-yl) amino) -2-fluoro-N-isobutoxy-5-methoxybenzamide 63mg. 1 H NMR(400MHz,DMSO)δ11.22(s,1H),8.94(s,1H),8.53(d,J=2.3Hz,1H),8.45(d,J=2.4Hz,1H),8.26–8.15(m,2H),7.20(s,1H),7.15(d,J=6.2Hz,1H),6.98(s,1H),5.82(s,2H),3.88(s,3H),3.64(d,J=6.6Hz,2H),3.28(s,3H),3.18(s,3H),1.90(m,1H),0.92(d,J=6.6Hz,6H).MS m/z(ESI):623.2[M+H] +
Biological evaluation of Compounds
Test example 1: in vitro enzymatic inhibitory Activity of the Compounds of the invention
1. Reagent, consumable and instrument
2. Experimental procedure
1) Add 50. Mu.L of compound to 384 well dilution plates.
2) Compounds of each column were treated with DMSO at 1: serial dilutions were performed 3, 10 points per dilution plus DMSO-only control.
3) Diluted compound solutions in each row were transferred to 0.1 μl to 384 assay plates using Echo, containing 2 replicates per column.
4) mu.L of 2 Xenzyme solution was added to the assay plate and centrifuged at 1000rpm for 1 min. Incubate at 25℃for 15 min.
5) mu.L of 2 Xsubstrate solution was added to 384-well assay plates.
6) Incubate at 25℃for 60 min.
7) mu.L of Sa-XL665 solution and 5. Mu.L of TK antibody-Eu 3 were mixed + Added to the assay plate. Centrifuge at 1000rpm for 1 minute.
8) Incubate at 25℃for 60 min.
9) The fluorescent signal was read on an Envision 2104 plate reader.
3. Data analysis
1) For each screening plate, mean data and Standard Deviation (SD) of DMSO and 100nM Defactinib (as control) were calculated
2) Percent inhibition of compound (% inh) =100 (max-sample value)/(max-min)
3) The IC50 was calculated using the nonlinear regression equation of XLfit 5.3.1, as follows:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
log (concentration of Compound)
Inhibition ratio (% inh)
Top and Bottom unit consistent with Y unit
log IC50 is consistent with the unit of X
Hillslope, gradient coefficient or gradient
4. Experimental results
FAK kinase activity IC50 activity data for specific compounds are shown in table 1.
TABLE 1
The results show that the compound of the invention has better inhibition effect on FAK and IC thereof 50 Are between 0.001 mu M and 10 mu M, i.e. have better in vitro FAK enzymatic inhibition activity.
Test example 2: pharmacodynamic evaluation in compound in vitro Diffuse Gastric Cancer (DGC) organoid model
The experimental steps are as follows: organoids were amplified in 6 well plates (7 Matrigel aliquots per well) for 2-3 days prior to assessment. After passaging, 1000 cells were seeded in 5. Mu.L Matrigel aliquots into each well of 96-well plates containing 100. Mu.L 50% L-WRN medium. After 24 hours of organoid culture, the compound was added. The compound was dissolved in DMSO to a final concentration of 10 μm stock solution, then the stock solution was diluted to 1000 times the use concentration with DMSO, then the medium was added to dilute to the use concentration, the dilution was added to the culture well plate, and after one addition of the compound, the medium was not replaced later nor was the compound re-added. The organoids were then cultured for the indicated number of days, and then the effect of the compounds was evaluated IN three ways, all with DMSO as negative control and either difactinib (Defactinib) or IN10018 as positive control.
1. The cell morphology is observed through a microscope, and the normal cell and the diffuse gastric cancer organoid model are different in cell morphology, as shown in fig. 1, the normal organoid morphology is in a spherical vacuum shape, and the diffuse gastric cancer organoid is in a solid grape-shaped irregular shape, so that observing the influence of a drug on the cell morphology is also one of important characterization for evaluating the drug effect; experimental results show that the compound can restore cell morphology in an in vitro DGC organoid model, as shown in figures 1-5 (partial compound results of the invention are not shown), the compound 4, 6, 8 and 9 can restore cell morphology when being treated in the DGC organoid model at the concentration of 2.5 mu M, the effect of the compound 8 and 9 is obviously superior to that of the Defactinib, and simultaneously, the cell proliferation inhibition effect which is better than that of the Defactinib is obviously observed.
2. Cell viability was assessed using reagents and protocols from CellTiter-Glo (Promega G7570). mu.L CellTiter-Glo reagent and 50. Mu.L medium were mixed and added to each well, and the plate was gently shaken at room temperature for 30 minutes to dissolve the matrigel and read on a Tecan plate reader.
Experimental results show that the compounds of the invention can inhibit the cell growth of the human diffuse organoid cell model. As shown in fig. 6, compound 9 can inhibit cell proliferation more significantly than Defactinib at 2.5 μm.
3. After cell culture and drug administration culture for 48 hours by the method, cells were lysed, proteins were extracted, protein detection was performed by Western Blot technique, grey value analysis of protein bands was performed after gel imaging, and the effect of compounds on phosphorylated FAK (P-FAK (Y397)) and activated YAP in DGC organoid model was examined. (see Haish Zhang, cancer Discovery,2020 for details). The results are shown in FIGS. 7-8.
Experimental results show that the compound can inhibit the phosphorylation of FAK (inhibit FAK kinase activity and reduce phosphorylated FAK) in a DGC organoid model, and simultaneously reduce activated YAP and show dose dependence. Whereas Defactinib does not inhibit YAP activity.
Test example 3: pharmacodynamics evaluation of compound in vitro human diffuse gastric cancer tumor cell line SNU668 model
1. Cell plating
SNU-668 cells (supplier: KCLB; cat. No. 00668) grown to 70% -80%, the culture supernatant was aspirated, and the supernatant was rinsed once with PBS for aspiration;
b.10cm dish is added with 1ml of 0.25% pancreatin, evenly shaken left and right and up and down, and put into an incubator for digestion for 2-3min;
c. taking out the culture dish, adding 2-3ml of complete culture medium to stop digestion, transferring to a 15ml centrifuge tube, and centrifuging at 1000rpm for 5min;
d. the supernatant was aspirated, resuspended in 1ml of complete medium and counted;
e. the 12-well plate was removed, 150000 cells and 900. Mu.L of medium were plated per well, and placed in a cell incubator for overnight culture.
2. Cell administration
a. Taking out small molecule medicine mother liquor Defactinib, IN10018, compound 24, compound 25, compound 26, compound 27, compound 28 and DMSO in advance, blowing for several times, and mixing uniformly;
b. each small molecule is administered at a concentration of 1 μm or 2.5 μm per well, and each small molecule is diluted to 10 μm or 25 μm with 1mM or 2.5mM stock, respectively, using culture medium;
c. taking out the culture plate with the well-planted cells, marking the serial number of each hole, respectively sucking 100 mu L of small molecules and concentration into the holes by using a 200 mu L pipetting gun, and adding the small molecules and the concentration into the holes around the holes drop by drop;
d. After all the holes are added, the medicine is slightly shaken for two times to be uniformly diffused, the time is recorded, and the medicine is put back into the cell incubator.
3. Protein extraction
a. After 24 hours or 48 hours of administration, the culture medium is discarded, the PBS is washed and discarded;
b. cells were lysed by adding RIPA lysate containing phosphatase inhibitor and PMSF into well plate, centrifuging at 12000rpm for 5min, collecting supernatant, measuring protein concentration and adjusting protein concentration, adding 4×loading buffer, and boiling.
WB detection
SDS-PAGE electrophoresis was performed: 80v,30mm,120v,60min; transferring: 80v,60min; closing: 5% skimmed milk 1h, primary antibodies (non-p-YAP (1:1500, abcam), p-FAK Y397 (1:1000, CST), actin (1:10000, CST) formulated with 5% BSA. TBST, incubated overnight at 4 ℃, secondary antibodies (coat-anti-mouse-IgG and coat-anti-rabit-IgG, 1:3000, protein) formulated with 5% skimmed milk, incubated for 1-2h at RT.
The results are shown in fig. 9-10, and show that the compounds of the present invention can inhibit FAK phosphorylation (inhibit FAK kinase activity, reduce phosphorylated FAK) while reducing activated YAP in the SNU668 model of human diffuse gastric cancer tumor cell line. Whereas Defactinib and IN10018 have no effect of inhibiting YAP activity.
1000 cells/well were plated on 96-well cell plates according to the method of culture and administration of SNU668 described above. The following day, 0.5uM and 2.5 uM of Defactinib and compound 9 were added and cell viability was assessed on days 2, 4 and 6 post-dosing using reagents and protocols from CellTiter-Glo (Promega G7570). mu.L CellTiter-Glo reagent and 50. Mu.L medium were mixed and added to each well, and the plates were read on a Tecan plate reader by gently shaking the plates for 30 minutes at room temperature.
The experimental result shows that the compound 9 can inhibit the cell growth of the human diffuse gastric cancer tumor cell line model. As shown in fig. 11, compound 9 can inhibit cell proliferation more significantly than Defactinib at 0.5 μm and 2.5 μm.
Thus, the compounds of the application may be used for the treatment of diseases, especially cancer, by inhibiting FAK kinase activity and/or reducing activated YAP.
All documents mentioned in this disclosure are incorporated by reference in this disclosure as if each were individually incorporated by reference. Further, it will be appreciated that various changes and modifications may be made by those skilled in the art after reading the above teachings, and such equivalents are intended to fall within the scope of the application as defined in the appended claims.

Claims (13)

  1. A compound of formula I, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof,
    wherein,
    ring a is independently selected from: C3-C20 heteroaryl and C6-C20 aryl, such as 5-6 membered heteroaryl and phenyl;
    the B ring is independently selected from: C3-C20 heteroaryl and C6-C20 aryl, such as 5-6 membered heteroaryl and phenyl; preferably, ring B is a six membered aryl or heteroaryl group;
    l is independently selected from: bond or CH 2 Preferably, L is CH 2
    X is independently selected from CH or N, preferably X is N;
    R 1 independently selected from: halogen, -C (=O) N (CH) 3 ) 2 、-C(=O)NHCH 3 、-C(=O)NH 2 -CH (=o), -COOH, CN, C1-C6 alkyl, -CF 3 、-CHF 2 、-CH 2 F、-CO 2 CH 3
    R 2 Independently selected from: -N (R) 5 )S(O) m R 6 、-P(=O)R' 5 R' 6 、-S(O) m NR 5 R 6 、-C(=O)NR 5 R 6 、-NR 5 C(=O)R 6 、-C(=O)R 5 、-C(=O)OR 5 、-OC(=O)R 5 、-S(O) m R 5
    R 3 Each independently selected from: -H, -OH, halogen, -CF 3 、-CHF 2 、-CH 2 F、-CN、-NO 2 、-NR 5 R 6 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkyneA group, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3R 9 Substitution;
    R 4 each independently selected from: -H, -OR 5 Halogen, -CF 3 、-CHF 2 、-CH 2 F、-CN、-NO 2 、-NR 5 R 6 、-C(=O)NR 5 R 6 、-C(=O)NR 5 OR 6 、-C(R 5 )=NR 6 、-NR 5 C(=O)R 6 、-C(=O)R 5 、-C(=O)C(=O)R 5 、-C(=O)OR 5 、-OC(=O)R 5 、-OC(=O)OR 5 、-P(=O)R' 5 R' 6 、-S(=O)(=NR 5 )R 6 、-S(O) m R 5 、-NR 5 S(O) m R 6 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3R 9 Substitution;
    or any two adjacent R 4 And C5-C7 cycloalkyl, 5-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, which together with the atoms to which they are attached; wherein the C5-C7 cycloalkyl, 5-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl may be substituted with 1-3R 9 Substitution;
    R 5 、R 6 、R' 5 and R'. 6 Each independently selected from: H. -OH, halogen,-CF 3 、-CHF 2 、-CH 2 F、-CN、-NO 2 、-CH 2 CF 3 、-NR 7 R 8 、-S(O) m R 7 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; or at NR 7 R 8 Wherein R is 7 And R is 8 Together with the N atom to which it is attached form a 3-10 membered heterocyclyl (including bridged and spiro rings); wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3R 9 Substitution;
    R 9 each independently selected from: H. -OH, oxo (=o), halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 、-OR 10 、-C(=O)R 10 、-OC(=O)R 10 、-OC(=O)R 10 、-OC(=O)OR 10 、-C(=O)NR 10 R 11 、-NR 10 C(=O)NR 11 R 12 、-NR 10 R 11 、-NR 10 C(=O)R 11 、-NR 10 S(O) m R 11 、-S(O) m R 10 、-S(O) m NR 10 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1 to 3 groups selected from the group consisting of: C1-C6 alkyl, halogen, -OH, -CN, -NO 2 、-CHF 2 、-CH 2 CF 3 、-CF 3 、-C(O)R 13 、-C(O)NR 13 R 14 、-S(O) m R 13 、-S(O) m NR 13 R 14 Substitution;
    R 7 、R 8 、R 10 、R 11 、R 12 、R 13 and R is 14 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein the C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1 to 3 groups selected from the group consisting of: -OH, halogen, -CN, -NO 2 、-NH 2 、-CHF 2 、-CH 2 CF 3 、-CF 3 C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, -C (O) - (C1-C6 alkoxy), C3-C12 cycloalkyl, 3-12 membered heterocycloalkyl, C1-C6 alkylamine;
    n and n' are each independently selected from 0, 1, 2, 3 or 4;
    m is independently selected from 0, 1 or 2.
  2. The compound of claim 1, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof,selected from:
    wherein p is 0, 1 or 2;
    R 4 is defined as in claim 1.
  3. The compound of claim 1, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, wherein the compound has a structure of formula II, formula III, formula IV, or formula V
    Wherein,
    p is 0, 1 or 2;
    R 1 、R 2 、R 3 、R 4 、R 5 、R 6 the definitions of B and n' are as defined in claim 1.
  4. The compound of claim 1, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, wherein R 1 Independently selected from: F. cl, -C (=O) NH 2 -CH (=o), -COOH, -CN, C1-C6 alkyl, -CF 3 、-CHF 2 、-CH 2 F。
  5. The compound of any one of claims 1-4, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, wherein ring B is selected from the group consisting of: phenyl, pyridyl or pyrazinyl.
  6. The compound of any one of claims 1-5, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, wherein R 4 Independently selected from: H. halogen, C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl, -C (O) NR 5 R 6 The method comprises the steps of carrying out a first treatment on the surface of the Wherein R is 5 And R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein said C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy.
  7. The compound of any one of claims 1-6, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, wherein R 1 Independently selected from: F. cl, -C (=O) NH 2 -CH (=o), -COOH, -CN, methyl, -CF 3 、-CHF 2 、-CH 2 F, performing the process; and/or
    R 4 Each independently selected from: F. cl, C1-C6 alkoxy, C3-C6 cycloalkyl, 3-6 membered heterocyclyl, -C (=O) NH (C1-C6 alkyl), -C (=O) NH (C3-C6 cycloalkyl), -C (=O) NH (3-6 membered heterocyclyl); wherein the C1-C6 alkyl, C1-C6 alkoxy, C3-C6 cycloalkyl, 3-6 membered heterocyclyl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy; and/or
    R 2 Independently selected from: -N (R) 5 )S(O) m R 6 、-P(=O)R' 5 R' 6 、-S(O) m R 5 The method comprises the steps of carrying out a first treatment on the surface of the Wherein R is 5 、R 6 、R' 5 And R'. 6 Each independently selected from: H. -OH, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-10 heteroaryl; wherein m is 1 or 2; and/or
    R 3 Each independently selected from: s (O) 2 CH 3 、-NCH 3 S(O) 2 CH 3 、-NHS(O) 2 CH 3 、-P(=O)(CH 3 ) 2 、-P(=O)(OH) 2 The method comprises the steps of carrying out a first treatment on the surface of the And/or
    Selected from:
  8. the compound of any one of claims 1-7, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, wherein the compound has a structure of one of the group consisting of:
    wherein,
    R 1 、R 2 、R 3 、R 4 is defined as in formula I, formula II, formula III, formula IV or formula V, R 5 And R is 6 Each independently selected from: H. C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-C12 heterocyclyl, C3-C12 cycloalkenyl, C6-C10 aryl, 5-C10 heteroaryl, R' is selected from C1-C6 alkyl; wherein said C1-C6 alkyl, C1-C6 alkoxy, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C10 aryl, 5-10 membered heteroaryl may be substituted with 1-3 substituents selected from the group consisting of: halogen, -CF 3 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CN、-NO 2 C1-C6 alkyl, C1-C6 alkoxy;
    preferably, the compound has one of the following structures:
  9. a pharmaceutical composition comprising a compound of any one of claims 1-8, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof; and a pharmaceutically acceptable carrier, diluent or excipient; preferably, the pharmaceutical composition further comprises one or more selected from the group consisting of: chemotherapeutic agents, PD-1 inhibitors, PD-1 antibodies, PD-L1 inhibitors, PD-L1 antibodies, ALK inhibitors, PI3K inhibitors, BTK inhibitors, EGFR antibodies, VEGFR inhibitors, VEGFR antibodies, HDAC inhibitors, CDK inhibitors, MEK inhibitors, akt inhibitors, mTOR inhibitors, SHP2 inhibitors, KRAS G12C inhibitors, KRAS G12D inhibitors, KRAS G12V inhibitors, C-MET inhibitors, her2 antibodies, claudin18.2 antibodies.
  10. Use of a compound according to any one of claims 1-8, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope or prodrug thereof, or a pharmaceutical composition according to claim 9, for the manufacture of a medicament for the prevention or treatment of a FAK-related disease; preferably, the FAK-associated disease is cancer, pulmonary hypertension, or pathological angiogenesis; more preferably, the cancer is selected from: skin cancer, bone cancer, glioma, breast cancer, adrenal cancer, bladder cancer, esophageal cancer, cancer of the head or neck, liver cancer, parathyroid cancer, penile cancer, small intestine cancer, thyroid cancer, urinary tract cancer, cervical cancer, endometrial cancer, fallopian tube cancer, renal pelvis cancer, vaginal cancer, vulvar cancer, chronic or acute leukemia, colon cancer, melanoma, hematological malignancy, hodgkin's lymphoma, lung cancer, lymphocytic lymphoma, central nervous system tumor (CNS), ovarian cancer, pancreatic cancer, pituitary adenoma, prostate cancer, soft tissue sarcoma, gastric cancer, uterine cancer.
  11. A process for preparing a compound as claimed in claim 1, comprising the steps of
    s 4) in an inert solvent and in the presence of a catalyst, reacting the compound I-1 with the compound I-2 to obtain a compound shown in a formula I;
    wherein X' is halogen;
    preferably, the method further comprises the step of:
    s 1) reacting compound I-3 with I-4 in the presence of a base (e.g., DIPEA) in an inert solvent (e.g., I-PrOH) to give a compound of formula I-5;
    s 2) in an inert solvent (e.g. DMF), a catalyst (e.g. Pd (PPh) 3 ) 2 Cl 2 And CuI) and under alkaline (such as DIPEA) conditions, reacting the compound I-5 with I-6 to obtain a compound of formula I-7;
    s 3) reacting the compound I-7 in the presence of an inert solvent (such as NMP) and a base (such as tBuOK) to obtain a compound of formula I-1;
    in the method, in the process of the invention,
    x ', X ", and X'" are each independently halogen (e.g., cl, br, I);
    ring A, ring B, X, L, R 1 、R 2 、R 3 、R 4 The definitions of n and n' are as defined in claim 1.
  12. A method of treating a FAK-related disease, comprising administering to a subject identified or diagnosed as having a FAK-related disease a therapeutically effective amount of a compound of any one of claims 1-8, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or a pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, or the pharmaceutical composition of claim 9.
  13. A method for inhibiting FAK kinase activity in a cell or a subject, comprising the step of contacting the cell or administering to the subject the compound of any one of claims 1-8, or an enantiomer, diastereomer, racemate, tautomer, stereoisomer, geometric isomer, nitroxide, metabolite, or pharmaceutically acceptable salt, hydrate, solvate, isotope, or prodrug thereof, or the pharmaceutical composition of claim 9; preferably, the cell is a mammalian cell; preferably, the subject is a mammal; more preferably a human.
CN202280022476.8A 2021-09-08 2022-09-08 Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof Pending CN117043163A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2021110507775 2021-09-08
CN202111050777 2021-09-08
PCT/CN2022/117868 WO2023036252A1 (en) 2021-09-08 2022-09-08 Pyrrolopyrimidine or pyrrolopyridine derivative and medical use thereof

Publications (1)

Publication Number Publication Date
CN117043163A true CN117043163A (en) 2023-11-10

Family

ID=85507197

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280022476.8A Pending CN117043163A (en) 2021-09-08 2022-09-08 Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof

Country Status (2)

Country Link
CN (1) CN117043163A (en)
WO (1) WO2023036252A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117105916B (en) * 2023-10-23 2024-01-16 希格生科(深圳)有限公司 Benzofuran compound and medical application thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1713806B1 (en) * 2004-02-14 2013-05-08 Irm Llc Compounds and compositions as protein kinase inhibitors
CA2714177A1 (en) * 2008-02-06 2009-08-13 Novartis Ag Pyrrolo [2,3-d] pyridines and use thereof as tyrosine kinase inhibitors
UY33227A (en) * 2010-02-19 2011-09-30 Novartis Ag PIRROLOPIRIMIDINE COMPOUNDS AS INHIBITORS OF THE CDK4 / 6
CN103059030B (en) * 2012-12-28 2015-04-29 北京师范大学 Pyrimidine compound with effect of adhesion kinase inhibition and preparation method and application thereof
US20160222014A1 (en) * 2013-09-10 2016-08-04 Asana Biosciences, Llc Compounds for regulating fak and/or src pathways
JP2022528047A (en) * 2019-03-22 2022-06-08 ショウヤオ ホールディングス(ベイジン) カンパニー, リミテッド Wee1 inhibitor, its manufacture and use

Also Published As

Publication number Publication date
WO2023036252A1 (en) 2023-03-16

Similar Documents

Publication Publication Date Title
JP2022506887A (en) Nitrogen-containing condensed heterocyclic SHP2 inhibitor compound, production method and use
JP6726677B2 (en) Substituted 2-H-pyrazole derivatives as anticancer agents
TWI713952B (en) Compounds and compositions as inhibitors of bromodomain proteins
EP2121692B1 (en) Substituted heterocycles as janus kinase inhibitors
JP6457623B2 (en) 2,4-disubstituted 7H-pyrrolo [2,3-d] pyrimidine derivatives, process for their preparation and use in medicine
CN107151249B (en) Pteridinone derivative as FLT3 inhibitor and application thereof
WO2021088945A1 (en) Compound as shp2 inhibitor and use thereof
WO2015127872A1 (en) 2,4-disubstituted phenylene-1,5-diamine derivatives and applications thereof, and pharmaceutical compositions and pharmaceutically acceptable compositions prepared therefrom
RU2633694C2 (en) Dyetherned phenylaminopyrimidine and pharmaceutical composition containing such connection
CN114430739A (en) EGFR inhibitor, composition and preparation method thereof
WO2010085597A1 (en) Macrocyclic compounds and their use as kinase inhibitors
TWI707855B (en) Novel imidazopyridazine compounds and their use
CN115109078A (en) Pyrimidopyridine inhibitor and preparation method and application thereof
CN116390728B (en) Quinazoline derivative, preparation method and application thereof
WO2021115457A1 (en) Pyrazolo[1,5-a]pyridine compound, preparation method therefor and use thereof
CN113302196A (en) EGFR inhibitor and composition and application thereof
JP7420403B2 (en) Compounds used as kinase inhibitors and their applications
CN110028509B (en) Pyrrolopyrimidines as selective JAK2 inhibitors, and synthesis method and application thereof
WO2020215998A1 (en) Pyrimido five-membered heterocyclic compound and use thereof as mutant idh2 inhibitor
CN108191837A (en) PI3K α/mTOR bidifly enzyme inhibitors and its pharmaceutical composition and application
WO2020125513A1 (en) Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine
CN115260187A (en) Pyridone compounds and uses thereof
CN117043163A (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof
TW202033520A (en) Fused ring derivatives used as fgfr4 inhibitors
CN112823159B (en) Aromatic heterocyclic compounds with kinase inhibitory activity

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination