CN112480101B - IRAK4 kinase inhibitor, preparation and application thereof - Google Patents

IRAK4 kinase inhibitor, preparation and application thereof Download PDF

Info

Publication number
CN112480101B
CN112480101B CN202010962799.8A CN202010962799A CN112480101B CN 112480101 B CN112480101 B CN 112480101B CN 202010962799 A CN202010962799 A CN 202010962799A CN 112480101 B CN112480101 B CN 112480101B
Authority
CN
China
Prior art keywords
alkyl
cancer
substituted
unsubstituted
group
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.)
Active
Application number
CN202010962799.8A
Other languages
Chinese (zh)
Other versions
CN112480101A (en
Inventor
段文虎
丁健
陈运
谢华
张惠斌
周金培
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Institute of Materia Medica of CAS
Original Assignee
Shanghai Institute of Materia Medica of CAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Institute of Materia Medica of CAS filed Critical Shanghai Institute of Materia Medica of CAS
Publication of CN112480101A publication Critical patent/CN112480101A/en
Application granted granted Critical
Publication of CN112480101B publication Critical patent/CN112480101B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/5381,4-Oxazines, e.g. morpholine ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring

Abstract

The invention provides an IRAK4 kinase inhibitor, a preparation method and an application thereof, and specifically provides a compound represented by the formula(I) The compound, stereoisomer, geometric isomer, tautomer, pharmaceutically acceptable salt, prodrug, hydrate or solvate, and preparation method and application thereof. Compared with the IRAK4 inhibitor in the prior art, the compound has obviously improved activity, so that the compound can be used for preparing IRAK4 mediated related diseases such as cancer, inflammatory diseases, autoimmune diseases and the like for prevention and/or treatment.

Description

IRAK4 kinase inhibitor, preparation and application thereof
Technical Field
The invention relates to the field of pharmaceutical chemistry, in particular to a compound with IRAK4 kinase inhibitory activity, and preparation and application thereof.
Background
Interleukin-1 receptor-associated kinase 4 (IRAK 4) is a serine/threonine kinase, located directly downstream of the interleukin-1 (IL-1) receptor family (IL-1, IL-18 and IL-33 receptors) and Toll-like receptors (TLRs, except TLR 3), and is a key signaling node for transduction of innate immunity. Upon ligand binding, IL-1 family receptors and TLRs recruit scaffolds to engage the protein myeloid differentiation primary response gene 88 (MyD 88) via a conserved Toll/IL receptor (TIR) domain. MyD88 in turn recruits IRAK4 using homotypic interactions with the death domain (death domain), thereby activating downstream signaling pathways such as NF-. Kappa.B and AP-1.IRAK4 has a dual kinase and backbone role, mediating downstream signaling pathways by forming a larger signaling complex, the MyD corpuscle, with MyD88 and IRAK 1.IRAK4 is constitutively activated and IL-1 stimulation does not significantly increase its intrinsic activity on Pellino1 in HEK293 cells expressing the IL-1 receptor, human THP1 monocytes stimulated by Pam3CSK4 or primary human macrophages. TLR7 and TLR9 mediated IFN- α/β production was abolished in IRAK4 kinase inactivated plasmacytoid dendritic cells. It is clear that the kinase activity of IRAK4 is not only essential for the induction of pro-inflammatory cytokines and chemokines by TLRs (2, 4,5,7 and 9), but also for the IFN-. Alpha./β induction mediated by TLR7 and TLR 9. Although IRAK4 occupies an extremely important place in innate immunity, studies have shown that the TIR-MyD88-IRAK4 signaling pathway is essential for protective immunity against a minority of purulent bacteria, but is redundant for host defenses against most natural infections.
Abnormal activation of the innate immune system is an important feature of many chronic autoimmune diseases. For example, the anti-acidifying antibody immune complex characteristic of Rheumatoid Arthritis (RA) and the anti-nucleic acid immune complex characteristic of Systemic Lupus Erythematosus (SLE) mediate signaling pathways through TLRs. In addition, activation of TLRs can drive B cells to differentiate into antibody-producing plasma cells, which are used to activate the adaptive immune system. Genetically modified mice with IRAK4 deletion or expressing kinase-inactivated IRAK4 exhibit some degree of impaired immune response, such as TNF α and IL-6 induced production when induced by bacterial Lipopolysaccharide (LPS). These mice were also resistant to experimentally induced arthritis, atherosclerosis, and MOG-induced encephalomyelitis. IRAK4 kinase inactivated mice were also shown to be resistant to the development of alzheimer's disease, a process thought to be due to reduced IL-1 production and signaling. Small molecule inhibitors of IRAK4 have been used to inhibit TLR-induced inflammatory signaling in vitro and in vivo. Studies have shown that IRAK4 inhibitors can reduce gout-like inflammation in uric acid-induced peritonitis models, ischemia-induced inflammation in 5/6 nephrectomized rats, and a mouse model of lupus erythematosus. Therefore, IRAK4 is considered to be an important pharmacological target for the treatment of chronic inflammatory diseases.
In activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL), leucine at MyD88 265 mutated to proline (L265P) in 29% of cases, the most prominent activating mutation in ABC DLBCL, where IRAK4 assumes most of the MyD88 function. The compounds ND-2110 and ND-2158 from Nimbus have outstanding antitumor effect in a mouse model of xenografted OCI-Ly10, and simultaneously have good combined drug effect with BTK inhibitor ibrutinib and BCL-2 inhibitor ABT-199.
Pancreatic ductal carcinoma (PDAC) overall survival rate of no more than 6% for 5 years. Studies have shown that PDAC patients positive for pIRAK4 have a higher chance of postoperative recurrence and a poorer prognosis than p-IRAK4 negative tumor patients. Through shRNA and CRISPR/Cas9n technologies, researchers find that gemcitabine and 5-fluorouracil are more effective in killing pancreatic ductal carcinoma cells after inhibiting IRAK 4. It has also been found that IRAK1/4 can drive the production of inflammatory cytokines and chemokines, resulting in the metastasis, invasion and proliferation of cancer-associated fibroblasts (CAFs). Another study showed that by knockout of mouse IRAK4, mouse lung tumor numbers were effectively reduced and cellular composition of bronchoalveolar perfusion (BALF) was not altered.
Disclosure of Invention
The invention aims to provide a compound shown as a formula (I) with IRAK4 inhibitory activity, a stereoisomer, a geometric isomer, a tautomer, a pharmaceutically acceptable salt, a prodrug, a hydrate or a solvate thereof, and a preparation method and application thereof.
In a first aspect of the present invention, there is provided a compound represented by formula (I), a stereoisomer, a geometric isomer, a tautomer, a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate thereof, or a solvate thereof:
Figure BDA0002681115790000021
wherein the content of the first and second substances,
x, Y, Z or W are independently selected from CH or N; and when X, Y, Z or W is CH, said H atom may be substituted by a substituent selected from the group consisting of: halogen, C1-C3 alkyl, C2-C6 acyl, C1-C3 alkoxy, trifluoromethoxy, trifluoroethoxy;
ring a is a 3-8 membered saturated heterocyclic ring (including monocyclic, fused or spiro) containing 1-2 heteroatoms selected from N, O and S, optionally substituted with one or more halogen, oxo, carboxyl, cyano, hydroxyl, substituted or unsubstituted 5-6 membered heterocyclic ring, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, said substitution being substituted with one or more halogen, C1-C3 alkyl or hydroxyl;
R 1 is selected from-NR 2 R 3 Substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C3-C10 cycloalkyl, substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted 5-10 membered heteroaryl, substituted or unsubstituted 3-11 membered saturated or partially saturated heterocyclic group, said substituents being represented by R a Representing;
each R is a Each independently selected from halogen, oxo, - (C0-C3 alkyl) -CN, - (C0-C3 alkyl) -OH, - (C0-C3 alkyl) -COOH, - (C0-C3 alkyl) C (= O) OR 5 - (C0-C3 alkyl) C (= O) R 5 - (C0-C3 alkyl) NR 5 R 6 C (= O) (C1-C3 alkyl), - (C0-C3 alkyl) C (= O) NR 5 R 6 - (C0-C3 alkyl) S (= O) NR 5 R 6 - (C0-C3 alkyl) S (= O) 2 NR 5 R 6 - (C0-C3 alkyl) S (= O) 2 (C1-C3 alkyl), - (C0-C3 alkyl) OP (= O) (OC 1-C3 alkyl) 2 Substituted or unsubstituted C1-C3 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C1-C6 alkylthio, said substitution being with one or more halogens;
R 2 and R 3 Each independently selected from the group consisting of a hydrogen atom, a substituted or unsubstituted C1-C3 alkyl group, a substituted or unsubstituted C3-C10 cycloalkyl group, - (C0-C3 alkyl) -6-to 10-membered aryl group, - (C0-C3 alkyl) -5-to 10-membered heteroaryl group, - (C0-C3 alkyl) -saturated or partially saturated 4-to 10-memberedHeterocyclyl, said substitution being substituted with halogen, hydroxy, amino, cyano or amido;
or R 2 And R 3 May form, together with the nitrogen atom to which they are attached, a 4-to 8-membered heterocyclic ring optionally substituted with one or more R a Substitution;
R 5 、R 6 each independently selected from the group consisting of a hydrogen atom, a substituted or unsubstituted C1-C6 alkyl group, a substituted or unsubstituted C3-C10 cycloalkyl group, and a substituted or unsubstituted 4-6 membered heterocyclic ring, said substitution being by one or more of a C1-C3 alkyl group, a hydroxyl group, a halogen, a carboxylic acid, a C2-C6 carboxylic acid ester.
In another preferred embodiment, R 2 And R 3 Together with the nitrogen atom to which they are attached form a 4-7 membered heterocyclic ring, optionally substituted with one or more R b Substitution;
R b independently selected from halogen, oxo, cyano, hydroxy, - (C0-C3 alkyl) C (= O) NHR 5 R 6 - (C0-C3 alkyl) -NR 5 R 6 Substituted or unsubstituted C1-C3 alkyl, said substitution being by one or more halogen, oxo, cyano, hydroxy.
In another preferred embodiment, R 1 Is a substituted or unsubstituted 6-10 membered aryl, substituted or unsubstituted 5-10 membered heteroaryl, and said aryl or heteroaryl is optionally substituted with one or more R c Substitution;
R c independently selected from halogen, cyano, hydroxyl, -COOH, -C (= O) OR 5 、-C(=O)R 5 C (= O) (C1-C3 alkyl), - (C0-C3 alkyl) NR 5 R 6 - (C0-C3 alkyl) C (= O) NR 5 R 6 - (C0-C3 alkyl) S (= O) NR 5 R 6 - (C0-C3 alkyl) S (= O) 2 NR 5 R 6 、-S(=O) 2 (C1-C3 alkyl), - (C0-C3 alkyl) OP (= O) (OC 1-C3 alkyl) 2 Substituted or unsubstituted C1-C3 alkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C1-C6 alkylthio, said substitution being by one or more halogen or hydroxy.
In another preferred embodiment, R 1 Is substituted or unsubstitutedA 5-6 membered aromatic heterocycle optionally substituted with one or more R c And (4) substitution.
In another preferred embodiment, the ring a is a substituted or unsubstituted group selected from the group consisting of:
Figure BDA0002681115790000031
wherein each p is independently 1, 2 or 3.
In another preferred embodiment, R is 1 Selected from the group consisting of: -NR 2 R 3 Substituted or unsubstituted C3-C6 cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted 5-to 10-membered heteroaryl, substituted or unsubstituted 3-to 6-membered saturated or partially saturated heterocyclyl, substituted or unsubstituted (3-to 6-membered saturated or partially saturated heterocyclyl) and 5-to 6-membered heteroaryl.
In another preferred embodiment, ring a is selected from the group consisting of:
Figure BDA0002681115790000032
R 4 selected from the group consisting of a hydrogen atom, a substituted or unsubstituted C1-C6 alkyl group, a substituted or unsubstituted C3-C10 cycloalkyl group, said substitution being by one or more halogens.
In a further preferred embodiment of the method,
Figure BDA0002681115790000041
selected from the group consisting of:
Figure BDA0002681115790000042
in another preferred embodiment, the compound is selected from the compounds shown in table 1.
In a second aspect of the present invention, there is provided a pharmaceutical composition comprising the following components:
1) A therapeutically effective amount of one or more compounds according to the first aspect of the present invention, stereoisomers, geometric isomers, tautomers thereof, pharmaceutically acceptable salts thereof, prodrugs thereof, hydrates thereof, or solvates thereof; and
2) A pharmaceutically acceptable carrier or excipient.
In another preferred embodiment, the pharmaceutical composition further comprises one or more active substances selected from the group consisting of: immunosuppressants, glucocorticoids, non-steroidal anti-inflammatory drugs, vinca alkaloids, paclitaxel, DNA damaging agents, bcl-2 inhibitors, BTK inhibitors, JAK inhibitors, hsp90 inhibitors, ALK inhibitors, FLT3 inhibitors, PI3K inhibitors, and SYK inhibitors.
In a third aspect of the present invention, there is provided a use of a compound according to the first aspect of the present invention, a stereoisomer, a geometric isomer, a tautomer, a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate thereof or a solvate thereof, or a pharmaceutical composition according to the second aspect of the present invention, for a use selected from the group consisting of:
1) For the preparation of a medicament for the prevention and/or treatment of IRAK4 mediated diseases;
2) Preparing a medicament for inhibiting IRAK 4.
In another preferred embodiment, the medicament is further for inhibiting a protein kinase selected from the group consisting of: FLT3, RET, VEGFR, erbB2, or a combination thereof.
In another preferred embodiment, said IRAK4 mediated disease is selected from the group consisting of: cancer, autoimmune diseases, inflammatory diseases and thromboembolic diseases.
In another preferred embodiment, wherein the cancer is selected from the group consisting of: diffuse large B-cell lymphoma, multiple myeloma, mantle cell lymphoma, waldenstrom's macroglobulinemia, acute myelogenous leukemia, chronic lymphocytic leukemia, small lymphocytic lymphoma, pancreatic ductal carcinoma.
In another preferred embodiment, wherein the autoimmune and inflammatory diseases are selected from the group consisting of: rheumatoid arthritis, osteoarthritis, juvenile arthritis, chronic obstructive pulmonary disease, multiple sclerosis, systemic lupus erythematosus, psoriasis, psoriatic arthritis, crohn's disease, ulcerative colitis, and irritable bowel syndrome.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Detailed Description
The inventor has long and intensive research and provides an IRAK4 inhibitor compound with a novel structure, and the compound has obviously improved activity compared with the IRAK4 inhibitor in the prior art, so that the compound can be used for preparing IRAK4 mediated related diseases such as cancer, inflammatory diseases, autoimmune diseases and the like for preventing and/or treating. Based on the above findings, the inventors have completed the present invention.
Term(s) for
As used herein, the term "heterocyclyl" is a cyclic group having 1, 2,3, 4, or 5 heteroatoms selected from the group consisting of: o, N or S.
In this context, the alkyl group is preferably an aliphatic alkyl group and may be a straight-chain alkyl group, a branched-chain alkyl group, a spirocycloalkyl group, a bridged cycloalkyl group, an alkenylalkyl group, an alkynylalkyl group, a cycloalkyl group, a cycloalkenyl group, a cycloalkynyl group, an alkoxyalkyl group, an alkoxyacylalkyl group, a cycloalkylalkyl group, including, without limitation: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, allyl, propargyl, cyclobutenyl, cyclohexenyl; expressions in the form of "C1-C8" are intended to include the corresponding group having 1, 2,3, 4,5, 6, 7 or 8 carbon atoms, e.g., "C1-C8 alkyl" refers to an alkyl group having 1, 2,3, 4,5, 6, 7 or 8 carbon atoms, and "C2-C10 alkenyl" refers to an alkenyl group having 2,3, 4,5, 6, 7, 8, 9 or 10 carbon atoms. In particular, the term "C0 alkyl" as used in the claims of the present invention refers to the case where the group is a bond.
In this context, the alkenyl group is preferably an ethenyl group, a propenyl group, a butenyl group, a styryl group, a phenylpropenyl group, or the like.
In this context, the cycloalkyl group may be a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 10 carbon atoms, preferably 3 to 8 carbon atoms, and more preferably the cycloalkyl group comprises 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentenyl, cyclohexyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups.
The heterocyclic group means a saturated or partially saturated monocyclic or polycyclic cyclic substituent including a 4 to 10-membered heterocyclic group, and the heterocyclic group is a saturated or unsaturated monocyclic, fused, spiro, fused, bridged ring or the like containing one or more hetero atoms (nitrogen, oxygen, sulfur) therein. The heterocyclic group described herein includes, but is not limited to, groups selected from the group consisting of: morpholine rings, piperidine rings, piperazine rings, N-alkyl or acyl substituted piperazine rings, homopiperazine rings, N-alkyl or acyl substituted homopiperazine rings, pyrrole, tetrahydropyrrole, 7H-purine and the like.
The aryl group refers to a 6 to 10 membered all carbon monocyclic or fused polycyclic (i.e., rings which share adjacent pairs of carbon atoms) group, and the group has a conjugated pi-electron system, such as phenyl and naphthyl. The aryl ring may be fused to a heterocyclyl, heteroaryl or cycloalkyl ring, non-limiting examples of which include benzimidazole, benzothiazole, benzoxazole, benzisoxazole, benzopyrazole, quinoline, benzindole, chroman.
The heteroaryl group refers to a heteroaromatic system containing 1 to 4 heteroatoms, 5 to 10 ring atoms, wherein the heteroatoms include oxygen, sulfur, and nitrogen. Heteroaryl is preferably 5-or 6-membered, for example furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl and the like. The heteroaryl group can be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring.
Unless otherwise specified, the structural formulae depicted herein are intended to include all tautomeric, enantiomeric and stereoisomeric forms (e.g., enantiomers, diastereomers, geometric isomers or conformational isomers): for example, R, S configuration containing asymmetric centers, (Z), (E) isomers and conformational isomers of (Z), (E) of double bonds. Thus, individual stereochemical isomers, tautomers or enantiomers, diastereomers or geometric isomers or conformational isomers or mixtures of tautomers of the compounds of the present invention are within the scope of the present invention.
The term "tautomer" means that structural isomers having different energies may exceed the low energy barrier and thus be converted to each other. For example, proton tautomers (i.e., proton shift changes) include interconversion by proton shift, such as 1H-indazole and 2H-indazole, 1H-benzo [ d ] imidazole and 3H-benzo [ d ] imidazole, and valence tautomers include interconversion by some recombination of bonding electrons.
Herein, the pharmaceutically acceptable salt is not particularly limited, and preferably includes: inorganic acid salts, organic acid salts, alkylsulfonic acid salts and arylsulfonic acid salts; the inorganic acid salt comprises hydrochloride, hydrobromide, nitrate, sulfate, phosphate and the like; the organic acid salt comprises formate, acetate, propionate, benzoate, maleate, fumarate, succinate, tartrate, citrate and the like; the alkyl sulfonate includes methyl sulfonate, ethyl sulfonate and the like; the aryl sulfonate includes benzene sulfonate, p-toluene sulfonate and the like.
Herein, the pharmaceutically acceptable solvate of the compound represented by the general formula (I) is not particularly limited, and preferably includes: solvates of the compounds represented by the general formula (I) with water, ethanol, isopropanol, ether, acetone, etc.
Compound (I)
The invention provides a compound shown as a formula (I), a stereoisomer, a geometric isomer, a tautomer, a pharmaceutically acceptable salt, a prodrug, a hydrate or a solvate thereof,
Figure BDA0002681115790000061
wherein each group is as defined above.
In addition toIn a preferred embodiment, in the compound, X, Y, Z, W, ring A and R 1 Each of which is a group corresponding to a specific compound described in table 1.
In another preferred embodiment, the compound is preferably the compound prepared in the examples.
In another preferred embodiment, the compound is selected from the compounds listed in table 1.
TABLE 1
Figure BDA0002681115790000062
Figure BDA0002681115790000071
Figure BDA0002681115790000081
Figure BDA0002681115790000091
Figure BDA0002681115790000101
Figure BDA0002681115790000111
Figure BDA0002681115790000121
Figure BDA0002681115790000131
Salt form
As used herein, the term "pharmaceutically acceptable salt" refers to a salt formed by a compound of the present invention and an acid or base, which is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, and the like; organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid and the like; and amino acids such as proline, phenylalanine, aspartic acid, glutamic acid, etc.
Another preferred class of salts are those of the compounds of the invention with bases, for example alkali metal salts (e.g. sodium or potassium), alkaline earth metal salts (e.g. magnesium or calcium), ammonium salts (e.g. lower alkanolammonium salts and other pharmaceutically acceptable amine salts), for example methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butylamine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, triethanolamine salts, and amine salts formed from morpholine, piperazine, lysine, respectively.
The term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio. "hydrate" refers to a complex formed by the coordination of a compound of the present invention with water.
The term "prodrug" includes a class of compounds which are biologically active or inactive in nature and which undergo metabolic or chemical reactions in the body to become transformed, or a salt or solution of the compound, when administered by an appropriate method. The prodrugs include, but are not limited to, carboxylate, carbonate, phosphate, nitrate, sulfate, sulfone, sulfoxide, amide, carbamate, azo, phosphoramide, glucoside, ether, acetal forms of the compounds.
Preparation of the Compounds of the invention
The following reaction schemes illustrate the preparation of the compounds of the present invention.
Reaction scheme I
Figure BDA0002681115790000141
The general synthetic method of the compound is shown as a reaction scheme I. The method comprises the following steps:
1. the compound 1a/1b and methyl magnesium bromide are subjected to Grignard reaction at-78 ℃ to obtain a compound 2a/2b;
2. performing a ring closure reaction on the compound 2a/2b under the condition of potassium tert-butoxide to obtain a compound 3a/3b;
3. carrying out nitration reaction on the compound 3a/3b under the action of nitric acid to obtain a compound 4a/4b
4. The compound 4a is subjected to Suzuki coupling reaction under the action of a palladium catalyst, and the compound 4b is subjected to substitution reaction under the action of alkali, so that a different form of a formula 5 can be obtained;
5. reducing the compound 5 under palladium carbon/hydrogen or reducing the compound under the condition of iron powder/ammonium chloride to obtain a compound 6;
6. the compound 6 reacts under the conditions of 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride and 1-hydroxybenzotriazole condensing agent to obtain a target compound 7.
Reaction scheme II
Figure BDA0002681115790000142
The general synthetic method of the compound is shown as a reaction route II. The method comprises the following steps:
1. the compound II-1 is debrominated under the action of n-butyllithium and reacts with dimethylcyclopropane to obtain a compound II-2;
2. the compound II-2 is subjected to ring closure under the action of potassium tert-butoxide to obtain a compound II-3;
3. carrying out nitration reaction on the compound II-3 under the action of acetic anhydride/copper nitrate to obtain a compound II-4;
4. carrying out substitution reaction on the compound II-4 under the action of organic base to obtain a compound II-5;
5. reducing the compound II-5 under palladium carbon/hydrogen to obtain a compound II-6;
6. and reacting the compound II-6 under the conditions of 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride and 1-hydroxybenzotriazole condensing agent to obtain a target compound II-7.
Pharmaceutical compositions containing compounds of formula (I)
The invention also relates to a pharmaceutical composition, which comprises one or more compounds selected from the compounds shown in the formula (I), pharmaceutically acceptable salts, prodrugs, hydrates and solvates thereof, in a therapeutically effective amount, and optionally a pharmaceutically acceptable carrier, and can be used for treating diseases related to the activity or expression level of IRAK 4. The pharmaceutical composition may be prepared in various forms according to different administration routes.
Because the compound has excellent inhibitory activity on IRAK4, the compound and various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates thereof, and a pharmaceutical composition containing the compound as a main active ingredient can be used for treating, preventing and relieving diseases related to the activity or expression amount of IRAK4, such as preventing and/or treating diseases related to abnormal expression of IRAK4 signal pathways. According to the prior art, the compounds of the invention are useful for the treatment of the following diseases: cancer, autoimmune diseases, inflammatory diseases and thromboembolic diseases, preferably, the compounds can be used for the treatment of the following diseases: diffuse large B-cell lymphoma, multiple myeloma, mantle cell lymphoma, fahrenheit macroglobulinemia, acute myelogenous leukemia, chronic lymphocytic leukemia, small lymphocytic lymphoma, pancreatic ductal carcinoma, rheumatoid arthritis, osteoarthritis, juvenile arthritis, chronic obstructive pulmonary disease, multiple sclerosis, systemic lupus erythematosus, psoriasis, psoriatic arthritis, crohn's disease, ulcerative colitis, and irritable bowel syndrome.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically 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 composition contains 1-2000mg of a compound of the invention per dose, more preferably, 5-200mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties include cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g., propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (e.g., propylene glycol, glycerol, mannitol, sorbitol, etc.), and the like
Figure BDA0002681115790000151
) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
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 mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) Fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) Disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) Wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, 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 using 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 delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed 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, especially cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain 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, 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 vehicles 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 desired.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
When using pharmaceutical compositions, a safe and effective amount of a compound of the present invention is administered to a mammal (e.g., a human) in need of treatment, wherein the administration is a pharmaceutically acceptable and effective dose, and the daily dose for a human of 60kg body weight is usually 1 to 2000mg, preferably 5 to 500mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally according to conventional conditions, or according to conditions recommended by the manufacturer. Unless otherwise indicated, percentages and parts are by weight.
Intermediate 1
6-fluoro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran (prepared according to the method reported in WO 2017/108723)
Figure BDA0002681115790000161
Step 1: preparation of methyl 2- (2, 4-difluorophenyl) acetate
Figure BDA0002681115790000162
Dissolving 2- (2, 4-difluorophenyl) acetic acid (5 g,29.0 mmol) in 100mL of methanol, adding 5mL of concentrated sulfuric acid, refluxing for reaction overnight, concentrating under reduced pressure, dissolving the residue in ethyl acetate, washing with water, washing with saturated sodium bicarbonate solution, washing with saturated salt water, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure to dryness to obtain 5g of light yellow oily substance, and directly putting into the next reaction.
Step 2:1- (2, 4-difluorophenyl) -2-methylpropan-2-ol
Figure BDA0002681115790000171
Methyl 2- (2, 4-difluorophenyl) acetate (5g, 26.8mmol) was dissolved in anhydrous tetrahydrofuran (60 mL), replaced with argon for protection, and a methyl magnesium bromide solution (27mL, 3M diethyl ether solution, 80.5 mmol) was added dropwise at-78 ℃ and after completion of the addition, the reaction was allowed to cool to room temperature after 10 minutes and the reaction was completed by TLC. Saturated ammonium chloride solution was added dropwise thereto in an ice bath to quench, ethyl acetate (150 mL) was extracted with water, and the ethyl acetate layer was washed with saturated brine and dried over anhydrous sodium sulfate. Vacuum concentrating to obtain light yellow oily matter, and directly adding into the next reaction. 1 H NMR(300MHz,Chloroform-d)δ7.24–7.15(m,1H),6.90–6.73(m,2H),2.77(d,J=1.6Hz,2H),1.23(s,6H).
And step 3: preparation of 6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran
Figure BDA0002681115790000172
Dissolving 1- (2, 4-difluorophenyl) -2-methylpropan-2-ol (5.41g, 29.0mmol) in anhydrous tetrahydrofuran (120 mL), adding potassium tert-butoxide (8.15g, 72.6 mmol), carrying out reflux reaction for 2 hours, detecting the completion of the reaction by TLC, concentrating under reduced pressure, adding ethyl acetate (150 mL) and water for extraction, washing an ethyl acetate layer with saturated common salt water, drying with anhydrous sodium sulfate, filtering, and carrying out silica gel column chromatography (ethyl acetate/petroleum ether =1% elution) to obtain 2.62g of colorless oily matter. Three-step reactionThe yield is as follows: 49 percent. 1 H NMR(400MHz,Chloroform-d)δ7.02(ddt,J=8.1,5.8,1.1Hz,1H),6.57–6.37(m,2H),2.95(t,J=1.4Hz,2H),1.47(s,6H).
And 4, step 4: preparation of 6-fluoro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran
Figure BDA0002681115790000173
Dissolving 6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran (2.62g, 15.7 mmol) in 50mL dichloromethane, dropwise adding nitric acid (2.4 mL) in ice bath, reacting at room temperature until TLC detection reaction is finished, extracting ethyl acetate (150 mL) and water, washing an ethyl acetate layer with saturated sodium bicarbonate solution, washing with saturated common salt water, drying with anhydrous sodium sulfate, filtering, and performing silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =3% -10%) to obtain 2.29g of orange yellow solid. Yield: and 69 percent. 1 H NMR(300MHz,DMSO-d 6 )δ8.04(dt,J=8.2,1.3Hz,1H),6.97(d,J=12.5Hz,1H),3.06(d,J=1.4Hz,2H),1.45(s,6H).
Intermediate 2
6-chloro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran (prepared according to the method reported in WO 2017/108723)
Figure BDA0002681115790000174
Step 1: preparation of methyl 2- (4-chloro-2-fluorophenyl) acetate
Figure BDA0002681115790000175
Dissolving 2- (4-chloro-2-fluorophenyl) acetic acid (5 g,26.5 mmol) in 50mL of methanol, adding 3mL of concentrated sulfuric acid, refluxing for reaction overnight, concentrating under reduced pressure, dissolving the residue in ethyl acetate, washing with water, washing with a saturated sodium bicarbonate solution, washing with a saturated common salt solution, drying over anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to dryness to obtain 5g of a pale yellow oily substance. 1 H NMR(400MHz,Chloroform-d)δ7.20(t,J=8.2Hz,1H),7.10(dt,J=9.8,2.3Hz,2H),3.71(s,3H),3.64(d,J=1.4Hz,2H).
Step 2: preparation of 1- (4-chloro-2-fluorophenyl) -2-methylpropan-2-ol
Figure BDA0002681115790000181
Methyl 2- (4-chloro-2-fluorophenyl) acetate (5.37g, 26.5 mmol) is dissolved in anhydrous tetrahydrofuran (60 mL), replaced by argon for protection, methyl magnesium bromide solution (26.5mL, 3M diethyl ether solution) is added dropwise at-78 ℃, after dropwise addition, the reaction is carried out for 15 minutes, the reaction is carried out at room temperature for 1 hour, and the reaction is detected by TLC to be finished. The mixture was quenched by dropwise addition of a saturated ammonium chloride solution in an ice bath, extracted with ethyl acetate (200 mL) and water, and the ethyl acetate layer was washed with saturated brine and dried over anhydrous sodium sulfate. Vacuum concentrating to dry to obtain light yellow oil. 1 H NMR(400MHz,Chloroform-d)δ7.20(dd,J=9.2,7.3Hz,1H),7.13–7.05(m,2H),2.78(d,J=1.6Hz,2H),1.24(d,J=1.0Hz,6H).
And step 3: preparation of 6-chloro-2, 2-dimethyl-2, 3-dihydrobenzofuran
Figure BDA0002681115790000182
Dissolving 1- (4-chloro-2-fluorophenyl) -2-methylpropan-2-ol (5.37g, 26.5 mmol) in anhydrous tetrahydrofuran (125 mL), adding potassium tert-butoxide (7.43g, 66.2 mmol), refluxing for overnight reaction, detecting by TLC after reaction, concentrating under reduced pressure, adding ethyl acetate (150 mL) and water for extraction, washing the ethyl acetate layer with saturated common salt water, drying over anhydrous sodium sulfate, filtering, and performing silica gel column chromatography (ethyl acetate/petroleum ether =1% elution) to obtain 3.57g of colorless oily substance. The yield of the three-step reaction is as follows: 74 percent. 1 H NMR(400MHz,Chloroform-d)δ7.05(dt,J=7.9,1.1Hz,1H),6.81(dd,J=7.9,1.9Hz,1H),6.74(d,J=1.8Hz,1H),2.98(s,2H),1.49(s,6H).
And 4, step 4: preparation of 6-chloro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran
Figure BDA0002681115790000183
Dissolving 6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran (3.57g, 19.5 mmol) in 40mL dichloromethane, dropwise adding nitric acid (2.4 mL) at room temperature, reacting at room temperature until TLC detection reaction is finished, extracting with ethyl acetate (150 mL) and water, washing with ethyl acetate layer saturated sodium bicarbonate solution, washing with saturated salt solution, drying with anhydrous sodium sulfate, filtering, performing silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =1% -10%) to obtain 4.45g of orange yellow solid, and recrystallizing with petroleum ether to obtain 2.02g of white needle-shaped solid. Yield: 45 percent. 1 H NMR(400MHz,Chloroform-d)δ7.84(s,1H),6.82(s,1H),3.04(d,J=1.3Hz,2H),1.52(s,6H).
Intermediate 3
2- (2- ((tert-butoxycarbonyl) amino) pyridin-4-yl) oxazole-4-carboxylic acid
Figure BDA0002681115790000184
Step 1: preparation of tert-butyl (4-bromopyridin-2-yl) carbamate
Figure BDA0002681115790000191
2-amino-4-bromopyridine (0.4g, 2.31mmol), di-tert-butyl dicarbonate (605mg, 2.77mmol) and 4-dimethylaminopyridine (423mg, 3.47mmol) were dissolved in dichloromethane (12 mL) and reacted overnight at room temperature, after completion of the TLC detection reaction,
silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =3% -20%) to obtain 354mg of white solid. Yield: 56 percent. 1 H NMR(300MHz,Chloroform-d)δ8.22(d,J=1.8Hz,1H),8.10–8.04(m,1H),7.97(s,1H),7.11(dd,J=5.4,1.7Hz,1H),1.53(s,9H).
Step 2: preparation of tert-butyl (4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridin-2-yl) carbamate
Figure BDA0002681115790000192
Tert-butyl (4-bromopyridin-2-yl) carbamate (0.2g, 732. Mu. Mol), pinacol diboron (223mg, 878. Mu. Mol) and potassium acetate (215mg, 2.20mmol) were dissolved in tetrahydrofuran (12 mL), and then under argon gas substitution protection, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (21mg, 29.2. Mu. Mol) was added thereto, followed by reflux reaction overnight, TLC detection reaction completed, filtration with celite, concentration, and direct charge to the next reaction.
And 3, step 3: preparation of ethyl 2- (2- ((tert-butoxycarbonyl) amino) pyridin-4-yl) oxazole-4-carboxylate
Figure BDA0002681115790000193
Ethyl 2-chlorooxazole-4-carboxylate (110mg, 626. Mu. Mol), (4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridin-2-yl) carbamic acid tert-butyl ester (240mg, 751. Mu. Mol), sodium carbonate (199mg, 1.88. Mu. Mol) and [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (23mg, 31. Mu. Mol), ethylene glycol dimethyl ether/water (10 mL/1 mL) were added to the reaction flask, replaced several times with argon, and reacted overnight at 90 ℃. The reaction was completed by TLC detection, filtered through celite, extracted with ethyl acetate (30 mL) and water, washed with saturated brine and dried over anhydrous sodium sulfate. Filtration and silica gel column chromatography (ethyl acetate/petroleum ether =1 elution) gave 121mg of a white solid. Yield: 58 percent. 1 H NMR(300MHz,DMSO-d 6 )δ10.15(s,1H),9.06(d,J=1.2Hz,1H),8.49–8.40(m,2H),7.56(dd,J=5.2,1.5Hz,1H),4.34(q,J=7.0Hz,2H),1.50(d,J=1.1Hz,9H),1.32(t,J=7.1Hz,3H).
And 4, step 4: preparation of 2- (2- ((tert-butoxycarbonyl) amino) pyridin-4-yl) oxazole-4-carboxylic acid
Figure BDA0002681115790000201
Ethyl 2- (2- ((tert-butoxycarbonyl) amino) pyridin-4-yl) oxazole-4-carboxylate (118mg, 356. Mu. Mol) was dissolved in tetrahydrofuran/methanol/water (4 mL/2mL/1 mL), and lithium hydroxide monohydrate (45mg, 1.07mmol) was added and reacted at room temperature for 20 minutesAfter TLC detection reaction, concentration, pH adjustment to weak acidity with 1N hydrochloric acid solution, solid precipitation, suction filtration and drying to obtain 83mg of white solid. Yield: 77 percent. 1 H NMR(300MHz,DMSO-d 6 )δ8.93(s,1H),8.49–8.39(m,2H),7.54(dd,J=5.2,1.5Hz,1H),1.50(s,9H).
Example 1
2- (2-Aminopyridin-4-yl) -N- (6- (4- (hydroxymethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-1)
Figure BDA0002681115790000202
Step 1: preparation of (1- (2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) piperidin-4-yl) methanol
Figure BDA0002681115790000203
6-fluoro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran (200mg, 94.7 mu mol) and 4-hydroxymethyl piperidine (218mg, 1.89mmol) are dissolved in N, N-dimethylformamide (5 mL), potassium carbonate (262mg, 1.89mmol) is added, reaction is carried out for two hours at 40 ℃, TLC detection reaction is finished, ethyl acetate (25 mL) and water are added for extraction, an ethyl acetate layer is washed with water, saturated salt is washed with water, anhydrous sodium sulfate is dried, suction filtration is carried out, concentration is carried out till dryness, and the mixture is directly put into the next reaction.
Step 2: preparation of (1- (5-amino-2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl) piperidin-4-yl) methanol
Figure BDA0002681115790000204
The reaction product obtained in the above step was dissolved in methanol (30 mL), 10% palladium on carbon (42 mg) was added, and the reaction was carried out at normal temperature for 6 hours under a hydrogen atmosphere, and after completion of the TLC detection reaction, the reaction was filtered with suction using celite, concentrated, and subjected to silica gel column chromatography (dichloromethane/methanol =1 to 50 gradient elution) to obtain 208mg of an off-white solid. The two-step yield: 79 percent. 1 H NMR(300MHz,DMSO-d 6 )δ6.49(s,1H),6.30(s,1H),4.46(t,J=5.3Hz,1H),4.15(s,2H),3.27(t,J=5.8Hz,2H),2.96(d,J=11.3Hz,2H),2.80(s,2H),2.39(t,J=11.2Hz,2H),1.70(d,J=12.3Hz,2H),1.47-1.35(m,1H)1.31(s,6H),1.22(td,J=12.4,11.1,3.3Hz,2H).
And 3, step 3: preparation of tert-butyl (4- (4- ((6- (4- (hydroxymethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) carbamoyl) oxazol-2-yl) pyridin-2-yl) carbamate
Figure BDA0002681115790000211
2- (2- ((tert-butoxycarbonyl) amino) pyridin-4-yl) oxazole-4-carboxylic acid (80mg, 263. Mu. Mol), 1- (5-amino-2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl) piperidin-4-yl) methanol (69mg, 251. Mu. Mol), 1-ethyl- (3-dimethylaminopropyl) carbonyl diimine hydrochloride (58mg, 301. Mu. Mol) and 1-hydroxybenzotriazole (41mg, 301. Mu. Mol) were dissolved in N, N-dimethylformamide (5 mL), N-diisopropylethylamine (104. Mu.L, 627. Mu. Mol) was added thereto, reaction was carried out at room temperature for 4 hours, TLC detection was completed, ethyl acetate (25 mL) and water were added for extraction, the ethyl acetate layer was washed with water, brine was dried over anhydrous sodium sulfate, filtered, and silica gel column chromatography (dichloromethane/methanol =2% elution) was carried out to obtain 80mg of a pale yellow solid. Yield: 56 percent. 1 H NMR(300MHz,DMSO-d 6 )δ10.19(s,1H),9.98(s,1H),8.99(s,1H),8.49(s,1H),8.45(d,J=5.2Hz,1H),8.18(s,1H),7.67–7.59(m,1H),6.69(s,1H),4.53(t,J=5.1Hz,1H),2.96(d,J=27.2Hz,4H),2.65(t,J=10.3Hz,2H),1.80(d,J=10.0Hz,2H),1.49(s,9H),1.42-1.35(m,6H).
Step 4: preparation of 2- (2-aminopyridin-4-yl) -N- (6- (4- (hydroxymethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000212
Reacting (4- (4- ((6- (4- (hydroxymethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) amino)Carbamoyl) oxazol-2-yl) pyridin-2-yl) carbamic acid tert-butyl ester (35mg, 62 μmol) was dissolved in dichloromethane (4.5 mL), trifluoroacetic acid (1.5 mL) was added and the reaction was allowed to react at room temperature for two hours, TLC detected for completion of the reaction, concentrated, extracted with saturated sodium bicarbonate and dichloromethane, the dichloromethane layer was dried over anhydrous sodium sulfate, filtered, concentrated, and purified by preparative thin layer chromatography (dichloromethane/methanol =16 as developing reagent) to give 26mg of a yellow solid. 1 H NMR(300MHz,DMSO-d 6 )δ9.97(s,1H),8.92(s,1H),8.22(s,1H),8.09(d,J=5.3Hz,1H),7.16–6.98(m,2H),6.69(s,1H),6.39(s,2H),4.73(t,J=5.3Hz,1H),3.43(s,2H),2.96(d,J=23.8Hz,4H),2.67(t,J=11.0Hz,2H),1.80(d,J=11.4Hz,2H),1.73–1.52(m,3H),1.40(s,6H).
Example 2
(R) -N- (6- (3-aminopiperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxamide trifluoroacetate (I-2)
Figure BDA0002681115790000221
Step 1: preparation of R) - (tert-butyl 4- (4- ((6- (3- ((tert-butoxycarbonyl) amino) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl-tert-butyl) carbamoyl) oxazol-2-yl) pyridin-2-yl) carbamate
Figure BDA0002681115790000222
Tert-butyl (R) - (4- (4- ((6- (3- ((tert-butoxycarbonyl) amino) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl tert-butyl) carbamoyl) oxazol-2-yl) pyridin-2-yl) carbamate can be prepared by a similar method as in step 4 of example 1. 1 H NMR(400MHz,Chloroform-d)δ10.15(s,1H),8.65(s,1H),8.41(d,J=5.2Hz,1H),8.37(s,1H),8.31(s,1H),7.99(s,1H),7.63(dd,J=5.2,1.5Hz,1H),6.60(s,1H),3.14(s,1H),3.02(s,2H),2.77(s,3H),1.90(s,1H),1.67(s,3H),1.55(s,9H),1.47(s,6H),1.19(s,9H).
Step 2: preparation of (R) -N- (6- (3-aminopiperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxamide trifluoroacetate
Tert-butyl (R) - (4- (4- ((6- (3- ((tert-butoxycarbonyl) amino) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl tert-butyl) carbamoyl) oxazol-2-yl) pyridin-2-yl) carbamate was dissolved in dichloromethane (4 mL), trifluoroacetic acid (1 mL) was added thereto and reacted for two hours at room temperature, and after completion of the reaction by TLC, the mixture was concentrated to dryness under reduced pressure, slurried with methanol/ether and suction-filtered to give 34mg of a pale yellow solid. 1 H NMR(400MHz,DMSO-d 6 )δ9.69(s,1H),9.07(s,1H),8.15(d,J=6.1Hz,1H),8.06(s,4H),7.75(s,2H),7.33(s,1H),7.19(dd,J=6.1,1.6Hz,1H),6.67(s,1H),3.46–3.33(m,2H),3.13(d,J=10.8Hz,1H),3.01(s,2H),2.85(d,J=11.9Hz,1H),2.77–2.61(m,2H),2.08(s,1H),1.91(d,J=13.3Hz,1H),1.80(d,J=11.3Hz,1H),1.50(d,J=8.6Hz,1H),1.42(d,J=4.1Hz,6H).
Example 3
N- (6- (4- (aminomethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxamide trifluoroacetate (I-3)
Figure BDA0002681115790000231
The trifluoroacetate salt of N- (6- (4- (aminomethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxamide may be prepared by a similar method as in example 2. 1 H NMR(400MHz,DMSO-d 6 )δ9.88(s,1H),9.05(s,1H),8.21–8.09(m,2H),7.86(s,5H),7.32(s,1H),7.18(dd,J=6.1,1.6Hz,1H),6.74(s,1H),2.98(d,J=24.2Hz,4H),2.81(t,J=6.4Hz,2H),2.68(t,J=11.5Hz,2H),1.91(d,J=12.5Hz,2H),1.74(s,1H),1.49(q,J=11.0Hz,2H),1.41(s,6H).
Example 4
2- (2-Aminopyridin-4-yl) -N- (6- (4-cyanopiperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-4)
Figure BDA0002681115790000232
2- (2-Aminopyridin-4-yl) -N- (6- (4-cyanopiperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that described in example 1. 1 H NMR(400MHz,Chloroform-d)δ9.93(s,1H),8.35(s,2H),8.23(d,J=5.3Hz,1H),7.20(dd,J=5.3,1.4Hz,1H),7.17–7.11(m,1H),6.55(s,1H),3.49(s,2H),3.12(d,J=12.2Hz,2H),3.08–2.98(m,2H),2.75(t,J=11.9Hz,3H),2.44–2.23(m,4H),1.48(s,6H).
Example 5
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6-morpholino-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-5)
Figure BDA0002681115790000233
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6-morpholino-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 1. 1 H NMR(400MHz,Chloroform-d)δ10.16(s,1H),8.34(d,J=6.7Hz,2H),8.19(d,J=5.4Hz,1H),7.13(d,J=5.3Hz,1H),6.97(s,1H),6.63(s,1H),4.90(s,2H),3.25(t,J=6.0Hz,4H),3.04(s,2H),2.84(t,J=6.0Hz,4H),1.48(s,6H).
Example 6
2- (2-Aminopyridin-4-yl) -N- (6- (3- (hydroxymethyl) pyrrolidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-6)
Figure BDA0002681115790000241
2- (2-Aminopyridin-4-yl) -N- (6- (3- (hydroxymethyl) pyrrolidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 1. 1 H NMR(400MHz,Chloroform-d)δ9.71(s,1H),8.33(s,1H),8.24(s,1H),8.17(d,J=5.4Hz,1H),7.27(d,J=1.2Hz,1H),7.19(dd,J=5.4,1.4Hz,1H),6.63(s,1H),5.14(s,2H),3.98–3.81(m,2H),3.26(td,J=8.7,3.2Hz,1H),3.13–2.96(m,4H),2.87(q,J=8.5Hz,1H),2.58(d,J=7.2Hz,1H),1.99–1.85(m,1H),1.47(s,6H).
Example 7
2- (2-Aminopyridin-4-yl) -N- (6- (4- (2, 2-difluoroethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-7)
Figure BDA0002681115790000242
Step 1: preparation of 1- (2, 2-difluoroethyl) piperazine hydrochloride
Figure BDA0002681115790000243
1-tert-butyloxycarbonyl piperazine (1g, 5.37mmol) and 2, 2-difluoroethanol (528mg, 6.44mmol) are dissolved in anhydrous dichloromethane, triethylamine and argon gas are added, trifluoromethanesulfonic anhydride is slowly added dropwise under ice bath, the mixture is reacted for 30 minutes in ice bath, and the mixture is turned to room temperature for reaction overnight. And (3) performing TLC detection, extracting dichloromethane and water, washing a dichloromethane layer, washing with saturated salt water, drying with anhydrous sodium sulfate, filtering, concentrating to dryness, dissolving in dioxane (10 mL), performing ice bath, adding a dioxane solution (10 mL,4.5 mol/L) of hydrochloric acid, turning to room temperature for reaction for three hours, performing ice bath after the reaction is finished, and performing suction filtration while the solution is cold to obtain 123mg of a yellow-brown solid. Yield: 12 percent. 1 H NMR(400MHz,DMSO-d 6 )δ9.78(s,2H),6.45(tt,J=54.5,4.2Hz,1H),3.44(td,J=15.3,4.1Hz,2H),3.36–3.17(m,8H).
And 2, step: preparation of 6- (4- (2, 2-difluoroethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-amine
Figure BDA0002681115790000251
6- (4- (2, 2-difluoroethyl) piperazin-1-yl) -2, 2-dimethyl-2 can be prepared by a method analogous to that of example 1, steps 1 and 2,3-dihydrobenzofuran-5-amine. 1 H NMR(400MHz,Chloroform-d)δ6.58(s,1H),6.49(s,1H),5.92(tt,J=55.9,4.4Hz,1H),3.48(s,6H),2.94–2.85(m,6H),1.43(s,6H).
And step 3: preparation of 2- (2-aminopyridin-4-yl) -N- (6- (4- (2, 2-difluoroethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000252
2- (2-Aminopyridin-4-yl) -N- (6- (4- (2, 2-difluoroethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-amine can be prepared from 6- (4- (2, 2-difluoroethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-amine by a reaction analogous to example 1, steps 3, 4. 1 H NMR(400MHz,Chloroform-d)δ9.96(s,1H),8.34(s,1H),8.31(s,1H),7.31(dd,J=5.3,1.4Hz,1H),7.14(t,J=1.0Hz,1H),6.63(s,1H),5.99(tt,J=55.7,4.2Hz,1H),4.72(s,2H),3.49(s,2H),3.06–2.99(m,2H),2.97–2.91(m,8H),1.48(s,6H).
Example 8
2- (2-Aminopyridin-4-yl) -N- (6- (4- (2-hydroxypropan-2-yl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-8)
Figure BDA0002681115790000253
2- (2-Aminopyridin-4-yl) -N- (6- (4- (2-hydroxypropan-2-yl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 1. 1 H NMR(300MHz,DMSO-d 6 )δ10.06(s,1H),8.89(q,J=1.5Hz,1H),8.27(s,1H),8.11(d,J=5.3Hz,1H),7.60(s,1H),7.28(s,1H),7.16–6.94(m,2H),6.72(d,J=2.5Hz,1H),6.48(s,2H),2.97(d,J=18.3Hz,4H),2.70(t,J=11.6Hz,2H),2.34(d,J=11.5Hz,1H),2.07(q,J=12.2,11.8Hz,2H),1.85(d,J=12.6Hz,2H),1.40(d,J=2.6Hz,6H).
Example 9
2- (2-Aminopyridin-4-yl) -N- (6- (4-hydroxypiperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-9)
Figure BDA0002681115790000261
2- (2-Aminopyridin-4-yl) -N- (6- (4-hydroxypiperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that described in example 1. 1 H NMR(400MHz,DMSO-d 6 )δ10.14(s,1H),8.91(s,1H),8.18(s,1H),8.10(d,J=5.3Hz,1H),7.24–7.04(m,2H),6.68(s,1H),6.30(s,2H),5.02(s,1H),3.72(s,1H),3.00(s,2H),2.95–2.84(m,2H),2.72(t,J=10.7Hz,2H),1.97(d,J=12.0Hz,2H),1.81(q,J=10.2,9.1Hz,2H),1.40(s,6H).
Example 10
2- (2-Aminopyridin-4-yl) -N- (6- (4- (cyanomethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-10)
Figure BDA0002681115790000262
2- (2-Aminopyridin-4-yl) -N- (6- (4- (cyanomethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 1. 1 H NMR(400MHz,Chloroform-d)δ9.58(s,1H),8.35(d,J=8.1Hz,2H),8.19(d,J=5.4Hz,1H),7.27(s,1H),7.19(dd,J=5.3,1.5Hz,1H),6.61(s,1H),5.09(s,2H),3.15(d,J=11.8Hz,2H),3.03(s,2H),2.70(t,J=11.5Hz,2H),2.54(d,J=4.2Hz,2H),2.03–1.81(m,5H),1.48(s,6H).
Example 11
2- (2-Aminopyridin-4-yl) -N- (6- (4- (2-hydroxyethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-11)
Figure BDA0002681115790000271
2- (2-Aminopyridin-4-yl) -N- (6- (4- (2-hydroxyethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 1. 1 H NMR(400MHz,DMSO-d 6 )δ9.91(s,1H),8.92(s,1H),8.19(s,1H),8.11(d,J=5.3Hz,1H),7.14(d,J=5.3Hz,1H),7.05(s,1H),6.72(s,1H),6.37(s,2H),4.65(s,1H),3.62(s,2H),3.00(s,3H),2.97–2.55(m,10H),1.41(s,6H).
Example 12
N- (6- (4- (2-amino-2-oxoethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxylic acid amide (I-12)
Figure BDA0002681115790000272
Step 1: preparation of 4- (2-amino-2-oxoethyl) piperazine-1-carboxylic acid tert-butyl ester
Figure BDA0002681115790000273
1-tert-butyloxycarbonyl piperazine (550mg, 2.95mmol) and bromoacetamide (611mg, 4.43mmol) were dissolved in tetrahydrofuran (20 mL), N-diisopropylethylamine (763mg, 5.91mmol) was added and reacted at room temperature overnight, TLC detection was completed, extraction was performed with ethyl acetate and water, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to dryness to obtain 464mg of a white solid. The yield thereof was found to be 64%. 1 H NMR(400MHz,DMSO-d 6 )δ7.17(d,J=30.9Hz,2H),2.86(s,2H),2.36(t,J=4.9Hz,4H),1.39(s,9H).
Step 2: preparation of 2- (piperazin-1-yl) acetamide trifluoroacetate
Figure BDA0002681115790000274
Tert-butyl 4- (2-amino-2-oxoethyl) piperazine-1-carboxylate (150mg, 616. Mu. Mol) was dissolved in dichloromethane (8 mL) and addedAdding trifluoroacetic acid (1.5 mL), reacting at room temperature for two hours, detecting by TLC to finish the reaction, concentrating to dryness, adding ether/methanol, pulping, and filtering to obtain 209mg of white solid. Yield: 91 percent. 1 H NMR(400MHz,DMSO-d 6 )δ9.01(s,2H),7.55(d,J=79.8Hz,2H),3.47(s,2H),3.26(t,J=5.1Hz,4H),3.06(s,4H).
Step preparation of 32- (4- (5-amino-2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl) piperazin-1-yl) acetamide
Figure BDA0002681115790000281
2- (4- (5-amino-2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl) piperazin-1-yl) acetamide may be prepared by a similar method to that of example 1, steps 1 and 2. 1 H NMR(400MHz,DMSO-d 6 )δ7.16(d,J=16.7Hz,2H),6.52(s,1H),6.32(s,1H),4.18(s,2H),2.90(s,2H),2.83(s,2H),2.77(s,4H),2.57(s,4H),1.33(s,6H).
And 4, step 4: preparation of N- (6- (4- (2-amino-2-oxoethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxamide
Figure BDA0002681115790000282
N- (6- (4- (2-amino-2-oxoethyl) piperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) -2- (2-aminopyridin-4-yl) oxazole-4-carboxamide can be prepared from 2- (4- (5-amino-2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl) piperazin-1-yl) acetamide by a similar reaction as in example 1, steps 3, 4. 1 H NMR(400MHz,DMSO-d 6 )δ9.91(s,1H),8.91(s,1H),8.21(s,1H),8.14(d,J=5.3Hz,1H),7.30(s,1H),7.17(s,1H),7.12(dd,J=5.3,1.4Hz,1H),7.06(s,1H),6.70(s,1H),6.41(s,2H),3.04(s,2H),3.01(s,2H),2.86(d,J=4.7Hz,4H),2.78(s,4H),1.41(s,6H).
Example 13
2- (2-Aminopyridin-4-yl) -N- (6- (4-carbamoylpiperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-13)
Figure BDA0002681115790000283
2- (2-Aminopyridin-4-yl) -N- (6- (4-carbamoylpiperazin-1-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 1. 1 H NMR(300MHz,DMSO-d 6 )δ9.81(s,1H),8.91(d,J=2.7Hz,1H),8.25(s,1H),8.04(d,J=4.9Hz,1H),7.10(t,J=3.2Hz,2H),6.69(d,J=2.8Hz,1H),6.35(s,2H),4.40(s,1H),3.00(s,3H),2.62(t,J=11.5Hz,2H),1.95–1.55(m,4H),1.40(d,J=3.8Hz,6H),1.14(d,J=3.6Hz,6H).
Example 14
2- (2-Aminopyridin-4-yl) -N- (2- (hydroxymethyl) -6- (4- (hydroxymethyl) piperidin-1-yl) -2-methyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-14)
Figure BDA0002681115790000291
Step 1: preparation of methyl 3- (2, 4-difluorophenyl) -2-hydroxy-2-methylpropionate
Figure BDA0002681115790000292
Adding magnesium chips (2.47g, 104mmol) and iodine simple substance (147mg, 579 mu mol) into a two-necked bottle, replacing argon, adding 30mL of anhydrous ether, heating to keep the solution slightly boiling, slowly adding 1- (bromomethyl) -2, 4-difluorobenzene (6 g,28.9 mmol), after the addition is finished, keeping boiling and stirring for half an hour to obtain (2, 4-difluorobenzyl) magnesium bromide for later use. Dissolving methyl 2-oxopropionate (2.96g, 28.9 mmol) in anhydrous ether, stirring at-78 deg.C for 30 min under argon protection, slowly adding freshly prepared (2, 4-difluorobenzyl) magnesium bromide, reacting at room temperature for 2hr, detecting by TLC, adding ammonium chloride solution to quench, filtering off insoluble substances, extracting with ethyl acetate and water, washing ethyl acetate layer with saturated common salt water, and adding anhydrous sulfurSodium salt was dried, filtered, concentrated under reduced pressure, and subjected to gel column chromatography (ethyl acetate/petroleum ether elution) to give 2.88g of a yellow oil. Yield: and 43 percent. 1 H NMR(400MHz,Chloroform-d)δ7.24(dd,J=8.6,6.5Hz,1H),6.88–6.75(m,2H),3.79(s,3H),3.18–3.07(m,2H),2.94(dd,J=13.9,1.2Hz,1H),1.50(s,3H).
Step 2: preparation of 6-fluoro-2-methyl-2, 3-dihydrobenzofuran-2-carboxylic acid
Figure BDA0002681115790000293
Methyl 3- (2, 4-difluorophenyl) -2-hydroxy-2-methylpropionate (2.88g, 12.51mmol) was dissolved in anhydrous tetrahydrofuran (75 mL), potassium tert-butoxide (3.51g, 31.28mmol) was added and reacted overnight at 50 deg.C, after completion of the reaction by TLC, ethyl acetate (100 mL) was added to separate the organic layer, dried over anhydrous sodium sulfate, filtered and concentrated to dryness to give a tan solid. 1 H NMR(400MHz,DMSO-d 6 )δ12.99(s,1H),7.21–7.13(m,1H),6.77–6.51(m,2H),3.46(d,J=16.0Hz,1H),3.11(d,J=16.0Hz,1H),1.59(s,3H).
And step 3: 6-fluoro-2-methyl-2, 3-dihydrobenzofuran-2-carboxylic acid methyl ester
Figure BDA0002681115790000294
6-fluoro-2-methyl-2, 3-dihydrobenzofuran-2-carboxylic acid (500mg, 2.55mmol) was dissolved in N, N-dimethylformamide (10 mL), cesium carbonate (765mg, 3.82mmol) was added, iodomethane (434mg, 3.06mmol) was added under stirring, and the reaction was carried out at room temperature for 5 hours, after which the reaction was completed by TLC. Ethyl acetate and water were added for extraction, and the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and subjected to gel column chromatography (elution with ethyl acetate/petroleum ether =1 = 50) to give 444mg of a pale yellow oil. Yield: 83 percent.
And 4, step 4: preparation of methyl 6-fluoro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-carboxylate
Figure BDA0002681115790000301
6-fluoro-2-methyl-2, 3-dihydrobenzofuran-2-carboxylic acid (444mg, 2.11mmol) is dissolved in dichloromethane (8 mL), nitric acid (0.5 mL) is slowly added dropwise at room temperature to react at room temperature until the TLC detection reaction is finished, water dichloromethane and water are added to extract, a dichloromethane layer is separated, saturated common salt is washed with water for three times, and anhydrous sodium sulfate is dried. Filtration, concentration under reduced pressure, and gel column chromatography (gradient elution with ethyl acetate/petroleum ether =1 =20 to 1. Yield: 42 percent. 1 H NMR(300MHz,Chloroform-d)δ7.93(d,J=7.5Hz,1H),6.70(d,J=11.2Hz,1H),3.81(d,J=1.5Hz,3H),3.65(d,J=16.3Hz,1H),3.17(d,J=16.3Hz,1H),1.76(s,2H).
And 5: preparation of (6-fluoro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-yl) methanol
Figure BDA0002681115790000302
Methyl 6-fluoro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-carboxylate (227mg, 889 μmol) was dissolved in tetrahydrofuran/ethanol (8 mL/2 mL), sodium borohydride (101mg, 2.67mmol) and lithium chloride (113mg, 2.67mmol) were added under ice bath, the mixture was allowed to react at room temperature for 3 hours, TLC detection was completed, saturated ammonium chloride solution was quenched, dichloromethane was added for extraction, the organic layer was separated, washed with saturated common salt water, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and subjected to column chromatography (gradient elution ethyl acetate/petroleum ether = 1) to obtain 200mg of a brown-yellow oil. Yield: 99 percent. 1 H NMR(300MHz,DMSO-d 6 )δ8.03(d,J=8.1Hz,1H),6.95(dt,J=11.0,2.4Hz,1H),5.19(dq,J=8.0,4.9,4.1Hz,1H),3.60–3.40(m,2H),2.92(d,J=16.2Hz,1H),1.38(q,J=2.4Hz,3H).
Step 6: preparation of (1- (2- (hydroxymethyl) -2-methyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) piperidin-4-yl) methanol
Figure BDA0002681115790000303
Dissolving (6-fluoro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-yl) methanol and 4-hydroxymethylpiperidine (29mg, 248 mu mol) in DMF (4 mL), adding potassium carbonate (57mg, 413 mu mol), reacting at room temperature overnight, detecting by TLC after the reaction is finished, adding ethyl acetate (20 mL), washing twice with water, washing with saturated salt water, drying with anhydrous sodium sulfate, filtering, concentrating to dryness, and directly putting into the next reaction without purification.
And 7: preparation of (1- (5-amino-2- (hydroxymethyl) -2-methyl-2, 3-dihydrobenzofuran-6-yl) piperidin-4-yl) methanol
Figure BDA0002681115790000311
Dissolving the product in methanol (18 mL), adding 5% palladium carbon (20 mg), reacting for 6 hours in hydrogen atmosphere, detecting by TLC after the reaction is finished, filtering by using kieselguhr, concentrating under reduced pressure to dryness, and directly putting the product into the next step without purification.
And 8: preparation of 2- (2-aminopyridin-4-yl) -N- (2- (hydroxymethyl) -6- (4- (hydroxymethyl) piperidin-1-yl) -2-methyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000312
2- (2-aminopyridin-4-yl) -N- (2- (hydroxymethyl) -6- (4- (hydroxymethyl) piperidin-1-yl) -2-methyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared from (1- (5-amino-2- (hydroxymethyl) -2-methyl-2, 3-dihydrobenzofuran-6-yl) piperidin-4-yl) methanol by a similar method as in steps 3,4 of example 1. 1 H NMR(300MHz,DMSO-d 6 )δ9.97(s,1H),8.90(s,1H),8.21(s,1H),8.10(d,J=5.2Hz,1H),7.09(dd,J=5.3,1.5Hz,1H),7.06(s,1H),6.67(s,1H),6.34(s,2H),5.04(t,J=5.8Hz,1H),4.70(t,J=5.4Hz,1H),3.43(dd,J=5.8,3.5Hz,4H),3.25–3.16(m,1H),2.94(d,J=11.3Hz,2H),2.83(d,J=15.8Hz,1H),2.67(t,J=11.0Hz,2H),1.81(d,J=11.3Hz,2H),1.72–1.47(m,3H),1.33(s,3H).
Example 15
2- (2-Aminopyridin-4-yl) -N- (2- (hydroxymethyl) -2-methyl-6-morpholino-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-15)
Figure BDA0002681115790000313
2- (2-Aminopyridin-4-yl) -N- (2- (hydroxymethyl) -2-methyl-6-morpholino-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 14. 1 H NMR(300MHz,DMSO-d 6 )δ9.86(s,1H),8.92(s,1H),8.15(d,J=10.2Hz,2H),7.03(s,2H),6.72(s,1H),6.39(s,2H),5.05(s,1H),3.86(s,4H),3.43(s,2H),3.20(d,J=16.2Hz,1H),2.82(d,J=5.3Hz,4H),1.34(s,3H).
Example 16
2- (2-Aminopyridin-4-yl) -N- (7-morpholino-3-oxo-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-6-yl) oxazole-4-carboxamide (I-16)
Figure BDA0002681115790000321
Step 1: preparation of 7-fluoro-6-nitro-2H-benzo [ b ] [1,4] oxazin-3 (4H) -one
Figure BDA0002681115790000322
The 7-fluoro-2H-1, 4-benzoxazine-3 (4H) -one (1g, 5.98mmol) and 80% sulfuric acid (10 mL) were iced to-10 ℃ and 65% nitric acid (580mg, 5.98mmol) and 80% sulfuric acid (1 mL) were slowly added to the reaction solution, and the reaction was carried out at room temperature for 30 minutes, after the TLC detection reaction was completed, the reaction solution was poured into crushed ice, stirred and filtered to obtain g of a yellow solid. 1 H NMR(300MHz,DMSO-d 6 )δ11.03(s,1H),7.64(d,J=7.5Hz,1H),7.27(d,J=12.3Hz,1H),4.79(s,2H),4.03(q,J=7.1Hz,2H),1.99(s,3H),1.18(t,J=7.1Hz,3H).
And 2, step: 7-morpholino-6-nitro-2H-benzo [ b ] [1,4] oxazin-3 (4H) -one
Figure BDA0002681115790000323
Reacting 7-fluoro-6-nitro-2H-benzo [ b][1,4]Dissolving oxazine-3 (4H) -ketone (100mg, 471 mu mol) in N, N-dimethylformamide (1 mL) and morpholine (1 mL), reacting at 50 ℃ for 2 hours, detecting by TLC after the reaction is finished, adding ethyl acetate, washing twice, washing with saturated salt water, drying with anhydrous sodium sulfate, filtering, concentrating to dryness, and directly putting into the next step for reaction. 1 H NMR(300MHz,Chloroform-d)δ7.30–7.24(m,1H),6.61(d,J=5.3Hz,1H),4.33(q,J=5.0Hz,2H),3.86(q,J=4.9Hz,4H),3.44(s,2H),2.97(q,J=4.8Hz,4H).
And 3, step 3: 6-amino-7-morpholino-2H-benzo [ b ] [1,4] oxazin-3 (4H) -one
Figure BDA0002681115790000324
Dissolving the product in methanol (10 mL), adding 5% palladium carbon (20 mg), reacting for 6 hours under hydrogen atmosphere, detecting by TLC after the reaction is finished, filtering by using kieselguhr, concentrating under reduced pressure to dryness, and directly putting into the next step for reaction. 1 H NMR(300MHz,DMSO-d 6 )δ10.42(s,1H),6.56(d,J=1.8Hz,1H),6.29(d,J=1.7Hz,1H),4.64(s,2H),4.37(d,J=1.8Hz,2H),3.71(t,J=4.4Hz,4H),2.71(t,J=4.5Hz,4H).
And 4, step 4: preparation of 2- (2-aminopyridin-4-yl) -N- (7-morpholino-3-oxo-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-6-yl) oxazole-4-carboxamide
Figure BDA0002681115790000331
Can be prepared by a similar method to that of steps 3,4 of example 1 from 6-amino-7-morpholino-2H-benzo [ b ]][1,4]Preparation of 2- (2-aminopyridin-4-yl) -N- (7-morpholino-3-oxo-3, 4-dihydro-2H-benzo [ b ] using oxazin-3 (4H) -one][1,4]Oxazin-6-yl) oxazole-4-carboxamide. 1 H NMR(300MHz,DMSO-d 6 )δ10.76(s,1H),9.95(s,1H),9.03(s,1H),8.17(d,J=5.7Hz,1H),8.08(s,1H),7.19(s,1H),7.13(d,J=5.9Hz,1H),7.00(s,1H),4.55(s,2H),2.84(s,4H).
Example 17
2- (2-Aminopyridin-4-yl) -N- (7- (4- (hydroxymethyl) piperidin-1-yl) -3-oxo-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-6-yl) oxazole-4-carboxamide (I-16)
Figure BDA0002681115790000332
2- (2-Aminopyridin-4-yl) -N- (7- (4- (hydroxymethyl) piperidin-1-yl) -3-oxo-3, 4-dihydro-2H-benzo [ b ] can be prepared by a similar procedure as in example 16][1,4]Oxazin-6-yl) oxazole-4-carboxamide. 1 H NMR(300MHz,DMSO-d 6 )δ10.72(s,1H),10.06(s,1H),9.00(d,J=2.1Hz,1H),8.10(d,J=5.1Hz,2H),7.19(d,J=8.5Hz,2H),6.93(s,1H),4.74(s,1H),4.53(d,J=2.1Hz,2H),2.93(d,J=11.0Hz,2H),2.69(t,J=11.0Hz,2H),1.79(d,J=11.3Hz,2H),1.64(d,J=11.9Hz,2H),1.58(s,1H).
Example 18
2- (2-Aminopyridin-4-yl) -N- (7-morpholino-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-6-yl) oxazole-4-carboxamide (I-18)
Figure BDA0002681115790000333
Can be prepared by a similar method to that of example 16 from 7-fluoro-3, 4-dihydro-2H-benzo [ b ]][1,4]Preparation of 2- (2-aminopyridin-4-yl) -N- (7-morpholino-3, 4-dihydro-2H-benzo [ b ] oxazine][1,4]Oxazin-6-yl) oxazole-4-carboxamide. 1 H NMR(300MHz,DMSO-d 6 )δ10.01(s,1H),8.91(d,J=1.8Hz,1H),8.19–8.07(m,1H),7.72(s,1H),7.03(s,2H),6.68(d,J=1.9Hz,1H),6.39(s,2H),5.88(s,1H),4.10(s,2H),3.84(s,4H),2.76(s,4H).
Example 19
2- (2-Aminopyridin-4-yl) -N- (7- (4- (hydroxymethyl) piperidin-1-yl) -3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-6-yl) oxazole-4-carboxamide (I-19)
Figure BDA0002681115790000341
Can be prepared by a method analogous to that of example 26 from 7-fluoro-3, 4-dihydro-2H-benzo [ b ]][1,4]Preparation of 2- (2-aminopyridin-4-yl) -N- (7- (4- (hydroxymethyl) piperidin-1-yl) -3, 4-dihydro-2H-benzo [ b ] oxazines][1,4]Oxazin-6-yl) oxazole-4-carboxamide. 1 H NMR(300MHz,DMSO-d 6 )δ10.13(s,1H),8.89(d,J=2.1Hz,1H),8.09(d,J=5.3Hz,1H),7.73(s,1H),7.14–7.02(m,2H),6.62(s,1H),6.37(s,2H),5.82(s,1H),4.71(s,1H),4.09(s,2H),2.86(d,J=11.1Hz,2H),2.65(d,J=11.1Hz,2H),1.77(s,2H),1.55(s,1H),1.26(d,J=19.2Hz,2H).
Example 20
2- (2-Aminopyridin-4-yl) -N- (2-isopropyl-6-morpholino-1-oxoisoindol-5-yl) oxazole-4-carboxamide (I-20)
Figure BDA0002681115790000342
Step 1: preparation of 6-chloro-2-isopropyl-5-nitroisoindol-1-one
Figure BDA0002681115790000343
Methyl 2- (bromomethyl) -5-chloro-4-nitrobenzoate (prepared according to the method reported in WO 2013/079505) (200mg, 648. Mu. Mol) was dissolved in methanol (8 mL), triethylamine (108. Mu.L, 777. Mu. Mol) and isopropylamine (66. Mu.L, 777. Mu. Mol) were added, the reaction was carried out at 70 ℃ for 6 hours, TLC was performed to complete the reaction, ethyl acetate was extracted with 1mol/L aqueous hydrochloric acid solution, the aqueous layer was extracted twice with ethyl acetate, and dried over anhydrous sodium sulfate. Concentrating to dryness. Directly putting into the next reaction. 1 H NMR(300MHz,DMSO-d 6 )δ8.34(s,1H),8.00(s,1H),4.54(s,2H),4.47–4.35(m,1H),1.26–1.22(m,6H).
Step 2: 2-isopropyl-6-morpholino-5-nitroisoindol-1-one
Figure BDA0002681115790000351
6-chloro-2-isopropyl-5-nitroisoindol-1-one (100mg, 392. Mu. Mol) and morpholine (69. Mu.L, 785. Mu. Mol), N, N-diisopropylethylamine (129. Mu.L, 785. Mu. Mol) were dissolved in dimethyl sulfoxide (2 mL), reacted overnight at 90 ℃, cooled to room temperature, extracted with ethyl acetate and water, the ethyl acetate layer was washed with water, washed with saturated saline, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and subjected to gel column chromatography (ethyl acetate/petroleum ether elution) to give 32mg of an orange oil. Yield: 26 percent. 1 H NMR(300MHz,Chloroform-d)δ7.81(s,1H),7.65(d,J=4.3Hz,1H),4.68(s,1H),4.37(d,J=5.0Hz,2H),3.85(s,4H),3.08(s,4H),1.36–1.25(m,6H).
And step 3: 5-amino-2-isopropyl-6-morpholinoisoindol-1-one
Figure BDA0002681115790000352
Preparation of 5-amino-2-isopropyl-6-morpholinoisoindol-1-one from 2-isopropyl-6-morpholino-5-nitroisoindol-1-one can be made by a method analogous to example 1, step 2. 1 H NMR(300MHz,Chloroform-d)δ7.54(s,1H),6.80(s,1H),4.66(d,J=7.0Hz,1H),4.24(s,2H),3.89(d,J=5.4Hz,4H),2.96(d,J=4.8Hz,4H),1.31–1.27(m,6H).
And 4, step 4: preparation of 2- (2-aminopyridin-4-yl) -N- (2-isopropyl-6-morpholino-1-oxoisoindol-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000353
2- (2-Aminopyridin-4-yl) -N- (2-isopropyl-6-morpholino-1-oxoisoindol-5-yl) oxazole-4-carboxamide can be prepared from 5-amino-2-isopropyl-6-morpholinoisoindol-1-one by a similar method to Steps 3,4 of example 1. 1 H NMR(300MHz,DMSO-d 6 )δ10.29(s,1H),9.02(s,1H),8.59(s,1H),8.14(d,J=5.4Hz,1H),7.57(s,1H),7.04(d,J=9.1Hz,2H),6.39(s,2H),4.42(s,2H),3.90(s,4H),2.92(s,4H),1.21(d,J=6.8Hz,6H).
Example 21
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6-morpholino-2, 3-dihydrofuro [2,3-b ] pyridin-5-yl) oxazole-4-carboxamide (I-21)
Figure BDA0002681115790000361
Step 1: preparation of 3-bromo-2, 6-difluoropyridine
Figure BDA0002681115790000362
3-bromo-2, 6-dichloropyridine (2.8g, 12.3mmol) and cesium fluoride (7.5g, 49.3mmol) were dissolved in dimethyl sulfoxide (35 mL), reacted at 80 ℃ for 8 hours, extracted with ethyl acetate and water, the ethyl acetate layer was washed twice with water, with saturated brine and dried over anhydrous sodium sulfate. Filtration, concentration under reduced pressure and gel column chromatography (petroleum ether elution) gave 1.01g of a colorless oil. 1 H NMR(400MHz,Chloroform-d)δ8.15–7.97(m,1H),6.79(dq,J=8.3,2.7,2.2Hz,1H).
Step 2: preparation of 1- (2, 6-difluoropyridin-3-yl) -2-methylpropan-2-ol
Figure BDA0002681115790000363
Dissolving 3-bromo-2, 6-difluoropyridine (1.01g, 5.21mmol) in anhydrous tetrahydrofuran (60 mL), replacing with argon for protection, adding n-butyllithium (2.2mL, 6.25mmol) at-78 ℃, adding dimethyl propylene oxide (0.53mL, 6.25mmol) and boron trifluoride diethyl etherate solution (0.75mL, 6.25mmol) after 30 minutes, reacting at room temperature for 1 hour, adding saturated ammonium chloride for quenching, extracting with ethyl acetate, washing with saturated common salt water, and drying with anhydrous sodium sulfate. Filtered, concentrated under reduced pressure, and subjected to gel column chromatography (gradient elution with ethyl acetate/petroleum ether =10% -15%) to obtain 146mg of yellow oil. 1 H NMR(400MHz,Chloroform-d)δ7.82(dt,J=9.3,7.9Hz,1H),6.79(ddd,J=8.0,2.9,0.6Hz,1H),2.78(s,2H),1.26(d,J=0.8Hz,7H).
And step 3: preparation of 6-fluoro-2, 2-dimethyl-2, 3-dihydrofuro [2,3-b ] pyridine
Figure BDA0002681115790000364
1- (2, 6-Difluoropyridin-3-yl) -2-methylpropan-2-ol (146mg, 779. Mu. Mol) and potassium tert-butoxide (218mg, 1.95mmol) were dissolved in anhydrous tetrahydrofuran (6 mL) and reacted at room temperature for 1 hour, TLC detection was completed, and extraction was performed with ethyl acetate and water, the ethyl acetate layer was washed with water, washed with saturated brine and dried over anhydrous sodium sulfate. Filtration, concentration under reduced pressure, and gel column chromatography (ethyl acetate/petroleum ether =5% elution) gave 67mg of a colorless oil. Yield: 51 percent. 1 H NMR(400MHz,Chloroform-d)δ7.45(tt,J=7.8,1.2Hz,1H),6.35(dd,J=7.8,1.5Hz,1H),2.99(dd,J=2.0,1.2Hz,2H),1.51(s,6H).
And 4, step 4: preparation of 6-fluoro-2, 2-dimethyl-5-nitro-2, 3-dihydrofuro [2,3-b ] pyridine
Figure BDA0002681115790000365
Reacting 6-fluoro-2, 2-dimethyl-2, 3-dihydrofuro [2,3-b ]]Pyridine (136mg, 813. Mu. Mol) was dissolved in acetic acid/acetic anhydride (4 mL/4 mL), copper nitrate (457 mg, 2.44mmol) was added in ice bath, the mixture was allowed to warm to room temperature for overnight reaction, TLC detection was performed for completion of the base reaction, ethyl acetate and water were extracted, the ethyl acetate layer was washed with water, brine was washed with saturated sodium chloride, and dried over anhydrous sodium sulfate. Filtering, decompressing, concentrating, and carrying out silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =5% -15%) to obtain 80mg of colorless oily substance. Yield: 46 percent. 1 H NMR(400MHz,Chloroform-d)δ8.28(dt,J=8.8,1.5Hz,1H),3.12(t,J=1.7Hz,2H),1.59(s,6H).
And 5: preparation of 2, 2-dimethyl-6-morpholino-5-nitro-2, 3-dihydrofuro [2,3-b ] pyridine
Figure BDA0002681115790000371
Reacting 6-fluoro-2, 2-dimethyl-5-nitro-2, 3-dihydrofuranAnd [2,3-b ]]Pyridine (40mg, 188. Mu. Mol) was dissolved in morpholine (1 mL) and N, N-dimethylformamide (1 mL) and reacted at room temperature for 2 hours, TLC detected completion of the reaction, extracted with ethyl acetate and water, the ethyl acetate layer was washed with water, washed with saturated brine and dried over anhydrous sodium sulfate. Filtering, concentrating under reduced pressure to dryness, and directly putting into the next reaction. 1 H NMR(400MHz,DMSO-d 6 )δ8.18(s,1H),3.65(t,J=4.5Hz,4H),2.99(s,2H),1.46(s,6H).
Step 6: preparation of 2, 2-dimethyl-6-morpholino-2, 3-dihydrofuro [2,3-b ] pyridin-5-amine
Figure BDA0002681115790000372
Prepared by a method analogous to that of example 1, step 2 from 2, 2-dimethyl-6-morpholino-5-nitro-2, 3-dihydrofuro [2,3-b ]]Preparation of 2, 2-dimethyl-6-morpholino-2, 3-dihydrofuro [2,3-b ] using pyridine]Pyridin-5-amine. 1 H NMR(400MHz,Chloroform-d)δ6.89(t,J=1.1Hz,1H),3.88–3.80(m,4H),3.45(s,2H),3.13–3.04(m,4H),2.93(d,J=1.1Hz,2H).
And 7: preparation of 2- (2-aminopyridin-4-yl) -N- (2, 2-dimethyl-6-morpholino-2, 3-dihydrofuro [2,3-b ] pyridin-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000373
2, 2-dimethyl-6-morpholino-2, 3-dihydrofuro [2,3-b ] can be prepared by a method similar to steps 3,4 of example 1]Preparation of 2- (2-aminopyridin-4-yl) -N- (2, 2-dimethyl-6-morpholino-2, 3-dihydrofuro [2,3-b ] using pyridin-5-amine]Pyridin-5-yl) oxazole-4-carboxamide. 1 H NMR(400MHz,Chloroform-d)δ9.36(s,1H),8.50(s,1H),8.33(s,1H),8.23(d,J=5.3Hz,1H),7.24(s,1H),7.09(s,1H),4.72(s,2H),3.95(s,4H),3.09(t,J=4.2Hz,4H),3.05(s,2H),1.50(s,6H).
Example 22
2- (2-Aminopyridin-4-yl) -N- (6- (4- (hydroxymethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrofuro [2,3-b ] pyridin-5-yl) oxazole-4-carboxamide (I-22)
Figure BDA0002681115790000381
2- (2-Aminopyridin-4-yl) -N- (6- (4- (hydroxymethyl) piperidin-1-yl) -2, 2-dimethyl-2, 3-dihydrofuro [2,3-b ] can be prepared by a similar method to that in example 31]Pyridin-5-yl) oxazole-4-carboxamide. 1 H NMR(400MHz,DMSO-d 6 )δ9.46(s,1H),8.97(s,1H),8.41(d,J=2.9Hz,1H),8.16–8.04(m,1H),7.09(d,J=3.5Hz,2H),6.50(s,2H),4.70(s,1H),3.41(s,2H),3.16(d,J=11.6Hz,2H),3.03(s,2H),2.72(t,J=11.2Hz,2H),1.78(d,J=10.4Hz,2H),1.57(t,J=10.1Hz,3H),1.43(s,6H).
Example 23
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (1-methyl-1H-pyrazol-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-23)
Figure BDA0002681115790000382
Step 1: preparation of 4- (2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) -1-methyl-1H-pyrazole
Figure BDA0002681115790000383
6-chloro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran (0.1g, 0.439mmol), 1-methylpyrazole-4-boronic acid pinacol ester (0.183g, 0.878mmol) and cesium carbonate (0.286g, 0.878mmol) were dissolved in dioxane/water (4 mL/0.5 mL), and [1,1' -bis (diphenylphosphino) ferrocene was added under an argon stream]Palladium dichloride (33mg, 0.043 mmol), reaction at 100 ℃ overnight, detection by TLC, suction filtration with celite, dilution with ethyl acetate (15 mL), washing with water and saturated brine once each, drying over anhydrous sodium sulfate, filtration, concentration under reduced pressure, and chromatography on silica gel column (gradient elution ethyl acetate/petroleum ether = 0-5%) to give 119mg of a yellow oil. Yield: 99 percent. 1 H NMR(400MHz,Chloroform-d)δ7.71(s,1H),7.54(d,J=10.6Hz,2H),6.69(s,1H),3.93(s,3H),3.06(s,2H),1.52(s,6H).
And 2, step: preparation of 2, 2-dimethyl-6- (1-methyl-1H-pyrazol-4-yl) -2, 3-dihydrobenzofuran-5-amine
Figure BDA0002681115790000391
Dissolving 4- (2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) -1-methyl-1H-pyrazole (101mg, 0.369mmol) in methanol (10 mL), adding palladium/carbon (30 mg), reacting in water bath at 30 ℃ for 6 hours under hydrogen atmosphere, detecting by TLC, filtering with diatomite, concentrating under reduced pressure, and directly putting into the next reaction without further purification. 1 H NMR(400MHz,Chloroform-d)δ7.65(s,1H),7.53(s,1H),6.60(d,J=9.9Hz,2H),3.94(s,3H),2.95(s,2H),1.46(s,6H).
And step 3: preparation of 2- (2-aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (1-methyl-1H-pyrazol-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000392
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (1-methyl-1H-pyrazol-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared from 2, 2-dimethyl-6- (1-methyl-1H-pyrazol-4-yl) -2, 3-dihydrobenzofuran-5-amine by a similar method to that of steps 3,4 of example 1. 1 H NMR(400MHz,DMSO-d 6 )δ9.58(s,1H),8.88(s,1H),8.11(d,J=5.1Hz,1H),7.97(s,1H),7.70(s,1H),7.50(d,J=10.9Hz,1H),7.02(d,J=4.6Hz,2H),6.82(s,1H),6.36(d,J=4.6Hz,1H),3.85(s,3H),3.03(s,2H),1.44(s,6H).
Example 24
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-24)
Figure BDA0002681115790000393
Step 1: preparation of 4- (2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) pyridine
Figure BDA0002681115790000394
4- (2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) pyridine can be prepared from 6-chloro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran by a similar method to step 1 in example 23. 1 H NMR(400MHz,Chloroform-d)δ8.70–8.58(m,2H),7.93(t,J=1.3Hz,1H),7.24–7.14(m,2H),6.60(s,1H),3.12(d,J=1.3Hz,2H),1.55(s,6H).MS(ESI,m/z):[M+H] + :271.2。
Step 2: preparation of 2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-amine
Dissolving 4- (2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran-6-yl) pyridine (0.1g, 0.370mmol) in ethanol/water (6 mL/2 mL), adding reduced iron powder (0.124g, 2.20mmol) and ammonium chloride (0.119g, 2.20mmol), reacting in an oil bath at 50 ℃ for 1.5 hours, detecting the completion of the reaction by TLC, carrying out suction filtration on kieselguhr, concentrating under reduced pressure, dissolving ethyl acetate (15 mL), extracting with 1mol/L diluted hydrochloric acid (15 mL), collecting an aqueous layer, adjusting the saturated sodium carbonate solution to be alkaline, extracting with ethyl acetate (15mL. Multidot.2), drying with anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to obtain 75mg of a light yellow solid. Yield: 84 percent. 1 H NMR(400MHz,Chloroform-d)δ8.72–8.59(m,2H),7.46–7.36(m,2H),6.64(s,1H),6.53(s,1H),3.32(brs,2H),2.99(s,2H),1.47(s,6H).
And step 3: preparation of 2- (2-aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000401
2- (2-Aminopyridin-4-yl) -N- (2, 2-Dihydrobenzofuran-5-amine can be prepared from 2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-Dihydrobenzofuran-5-amine by analogy with Steps 3,4 in example 12-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide. 1 H NMR(400MHz,DMSO-d 6 )δ9.72(s,1H),8.76(s,1H),8.64–8.52(m,2H),8.08(d,J=5.2Hz,1H),7.52–7.34(m,3H),7.04–6.89(m,2H),6.77(s,1H),6.35(s,2H),3.09(s,2H),1.46(s,6H).
Example 25
2- (2-Aminopyridin-4-yl) -N- (6- (furan-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-25)
Figure BDA0002681115790000402
2- (2-Aminopyridin-4-yl) -N- (6- (furan-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.61(s,1H),8.87(s,1H),8.20–8.06(m,1H),7.95(t,J=1.2Hz,1H),7.77(t,J=1.7Hz,1H),7.50(s,1H),7.12–6.97(m,2H),6.86(s,1H),6.82(dd,J=1.9,0.9Hz,1H),6.38(s,2H),3.05(s,2H),1.44(s,6H).
Example 26
2- (2-Aminopyridin-4-yl) -N- (6- (3, 5-dimethylisoxazol-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-26)
Figure BDA0002681115790000411
2- (2-Aminopyridin-4-yl) -N- (6- (3, 5-dimethylisoxazol-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,Chloroform-d)δ8.68(s,1H),8.29(d,J=2.6Hz,2H),8.20(d,J=5.4Hz,1H),7.13(dd,J=5.4,1.4Hz,1H),7.03(s,1H),6.55(s,1H),4.88(s,2H),3.11(s,2H),2.33(d,J=2.0Hz,3H),2.20(d,J=2.0Hz,3H),1.52(s,6H).
Example 27
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-3-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-27)
Figure BDA0002681115790000412
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-3-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,Chloroform-d)δ8.73(s,1H),8.68(d,J=4.6Hz,2H),8.27(s,1H),8.17(d,J=5.3Hz,1H),8.08(s,1H),7.77(d,J=7.8Hz,1H),7.41(dd,J=7.8,4.8Hz,1H),7.06(d,J=5.3Hz,1H),6.97(s,1H),6.69(s,1H),4.76(s,2H),3.11(s,2H),1.53(s,6H).
Example 28
2- (2-Aminopyridin-4-yl) -N- (6-morpholino-3H-spiro [ benzofuran-2, 4' -piperidin ] -5-yl) oxazole-4-carboxamide (I-28)
Figure BDA0002681115790000413
Step 1: preparation of 2, 2-trifluoro-1- (6-fluoro-5-nitro-3H-spiro [ benzofuran-2, 4 '-piperidin ] -1' -yl) ethan-1-one
Figure BDA0002681115790000414
6-fluoro-5-nitro-3H-spiro-tert-butyl [ benzofuran-2, 4' -piperidine ] can be prepared by a method similar to Steps 1 to 5 of example 29]-1' -carboxylic acid tert-butyl ester. 1 H NMR(400MHz,Chloroform-d)δ7.96(d,J=7.6Hz,1H),6.65(d,J=11.2Hz,1H),4.39(d,J=13.7Hz,1H),3.92(d,J=14.4Hz,1H),3.64(t,J=12.9Hz,1H),3.36(t,J=12.7Hz,1H),3.08(s,2H),2.09(d,J=14.1Hz,2H),1.93–1.76(m,2H).
Step 2: preparation of 2, 2-trifluoro-1- (6-morpholino-5-nitro-3H-spiro [ benzofuran-2, 4 '-piperidin ] -1' -yl) ethan-1-one
Figure BDA0002681115790000421
2, 2-trifluoro-1- (6-fluoro-5-nitro-3H-spiro [ benzofuran-2, 4' -piperidine)]-1' -yl) ethan-1-one (259mg, 0.744mmol) was dissolved in morpholine (5 mL), reacted at 50 ℃ for 45 minutes, water (25 mL) was added to precipitate a solid, filtered, and dried under vacuum to give 303mg of a yellow solid. Yield: 98 percent. 1 H NMR(400MHz,Chloroform-d)δ7.85(s,1H),6.51(s,1H),4.37(d,J=13.9Hz,1H),3.87(d,J=5.1Hz,5H),3.64(t,J=12.7Hz,1H),3.36(t,J=12.8Hz,1H),3.03(d,J=6.0Hz,6H),2.06(d,J=14.1Hz,2H),1.84(d,J=13.4Hz,2H).
And step 3: preparation of 1- (5-amino-6-morpholino-3H-spiro [ benzofuran-2, 4 '-piperidin ] -1' -yl) -2, 2-trifluoroethan-1-one
Figure BDA0002681115790000422
2, 2-trifluoro-1- (6-morpholino-5-nitro-3H-spiro [ benzofuran-2, 4' -piperidine)]-1' -yl) ethane-1-one (303mg, 0.729mmol) is dissolved in ethanol/water (24 mL/8 mL), reduced iron powder (244mg, 4.38mmol) and ammonium chloride (234mg, 4.38mmol) are added, oil bath reaction is carried out at 50 ℃ for 1.5 hours, TLC detection reaction is finished, kieselguhr suction filtration is carried out, reduced pressure concentration is carried out, ethyl acetate is dissolved (100 mL), saturated common salt water washing is carried out, anhydrous sodium sulfate is dried, filtration is carried out, and reduced pressure concentration is carried out to obtain 277mg of brown yellow solid. Yield: 99 percent. 1 H NMR(400MHz,Chloroform-d)δ6.60(s,1H),6.53(s,1H),4.33(d,J=13.2Hz,1H),3.84(t,J=4.6Hz,5H),3.65(t,J=12.8Hz,1H),3.36(t,J=12.5Hz,1H),2.93(s,2H),2.87(t,J=4.6Hz,4H),2.03(d,J=13.6Hz,2H),1.73(t,J=13.0Hz,3H).
And 4, step 4: preparation of 2- (2-aminopyridin-4-yl) -N- (6- (pyridin-4-yl) -1'- (2, 2-trifluoroacetyl) -3H-spiro [ benzofuran-2, 4' -piperidin ] -5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000423
2- (2-Aminopyridin-4-yl) -N- (6- (pyridin-4-yl) -1'- (2, 2-trifluoroacetyl) -3H-spiro [ benzofuran-2, 4' -piperidine ] -5-yl) oxazole-4-carboxamide can be prepared from 1- (5-amino-6-morpholino-3H-spiro [ benzofuran-2, 4 '-piperidine ] -1' -yl) -2, 2-trifluoroethane-1-one by a method analogous to steps 3,4 of example 1. MS (ESI): 573.3[ M ] +H ].
And 5: preparation of 2- (2-aminopyridin-4-yl) -N- (6-morpholino-3H-spiro [ benzofuran-2, 4' -piperidin ] -5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000431
2- (2-Aminopyridin-4-yl) -N- (6-morpholino-3H-spiro [ benzofuran-2, 4' -piperidine) can be prepared by a similar method to step 6 in example 29]-5-yl) oxazole-4-carboxamide. 1 H NMR(400MHz,DMSO-d 6 )δ9.86(s,1H),8.92(s,1H),8.18(s,1H),8.14(d,J=5.2Hz,1H),7.12–6.98(m,2H),6.78(s,1H),6.38(s,2H),3.95–3.80(m,4H),2.99(s,2H),2.86(dq,J=18.6,4.6Hz,6H),2.71–2.59(m,2H),1.68(dq,J=15.5,10.5,9.4Hz,4H).
Example 29
2- (2-Aminopyridin-4-yl) -N- (6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4' -piperidin ] -5-yl) oxazole-4-carboxamide trifluoroacetate (I-29)
Figure BDA0002681115790000432
Step 1: preparation of 4- (4-chloro-2-fluorobenzyl) -4-hydroxypiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0002681115790000433
Adding magnesium chips (2.18g, 35.8mmol) and iodine particles (151 mg) into a two-necked bottle, adding anhydrous ether (25 mL) under the protection of argon, heating to reflux, slowly dropwise adding 1- (bromomethyl) -4-chloro-2-fluorobenzene (8.0g, 35.8mmol), and keeping reflux reaction for 30 minutes after dropwise adding to prepare the (4-chloro-2-fluorobenzyl) magnesium bromide. Tert-butyl 4-oxopiperidine-1-carboxylate (5.94g, 29.8mmol) was dissolved in diethyl ether (190 mL), -78 lowThe prepared 4-chloro-2-fluorobenzyl) magnesium bromide is dripped under warm bath, and the temperature is changed to room temperature for reaction for 2 hours. Water and ethyl acetate (200 mL) were added, the organic layer was separated, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and subjected to silica gel column chromatography (methanol/dichloromethane =5% elution) to give 5.18g of a pale yellow solid. Yield: 51 percent. 1 H NMR(400MHz,Chloroform-d)δ7.16(t,J=8.0Hz,1H),7.11(d,J=1.9Hz,1H),7.08(q,J=2.0,1.6Hz,1H),3.85(s,2H),3.09(t,J=12.5Hz,2H),2.78(d,J=1.6Hz,2H),1.49(s,2H),1.45(s,9H).
Step 2: preparation of 6-chloro-3H-spiro [ benzofuran-2, 4 '-piperidine ] -1' -carboxylic acid tert-butyl ester
Figure BDA0002681115790000441
Tert-butyl 4- (4-chloro-2-fluorobenzyl) -4-hydroxypiperidine-1-carboxylate (2.91g, 8.46mmol) was dissolved in anhydrous tetrahydrofuran (100 mL), reacted at 65 ℃ for 3 hours, added with water and ethyl acetate, the organic layer was separated, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and subjected to silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =0 to 10%) to obtain 1.10g of a white solid. Yield: 40 percent. 1 H NMR(400MHz,Chloroform-d)δ7.03(d,J=7.9Hz,1H),6.80(dd,J=7.9,1.9Hz,1H),6.75(d,J=1.8Hz,1H),3.74(s,2H),3.52–3.31(m,2H),2.94(s,2H),1.89(d,J=13.4Hz,2H),1.69(td,J=13.9,12.0,4.4Hz,2H),1.47(s,9H).
And 3, step 3: preparation of 6-chloro-3H-spiro [ benzofuran-2, 4' -piperidine ] trifluoroacetate
Figure BDA0002681115790000442
Reacting 6-chloro-3H-spiro [ benzofuran-2, 4' -piperidine]Tert-butyl (300mg, 0.926mmol) of the-1' -carboxylate was dissolved in methylene chloride (10 mL), and trifluoroacetic acid (3 mL) was added to react at room temperature for 1.5 hours. Concentrated under reduced pressure, slurried with diethyl ether, and filtered to obtain 280mg of a white solid. Yield: 89 percent. 1 H NMR(400MHz,DMSO-d 6 )δ8.70(d,J=38.2Hz,2H),7.22(d,J=7.8Hz,1H),6.89(d,J=9.3Hz,2H),3.20(s,4H),3.08(s,2H),2.08–1.86(m,4H).
And 4, step 4: preparation of 1- (6-chloro-3H-spiro [ benzofuran-2, 4 '-piperidin ] -1' -yl) -2, 2-trifluoroethane-1-one
Figure BDA0002681115790000443
Reacting 6-chloro-3H-spiro [ benzofuran-2, 4' -piperidine]Trifluoroacetic acid salt (200 mg) was added to a two-necked flask, dichloromethane (8 mL) and N, N-diisopropylethylamine (294 μ 9, 1.78mmol) were added under argon protection, trifluoroacetic anhydride was added after ice bath for 10 minutes, the mixture was allowed to turn to room temperature for reaction for 3 hours, water and dichloromethane were added, the organic layer was separated, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and dried in vacuum to obtain 200mg of oil, which was directly put into the next reaction without further purification. 1 H NMR(400MHz,Chloroform-d)δ7.06(dd,J=7.9,1.3Hz,1H),6.84(dd,J=7.9,1.9Hz,1H),6.78(d,J=1.9Hz,1H),4.35(d,J=13.3Hz,1H),3.88(d,J=14.2Hz,1H),3.65(ddd,J=14.3,12.0,2.7Hz,1H),3.45–3.29(m,1H),3.08–2.92(m,2H),2.14–1.98(m,2H),1.89–1.70(m,2H).
And 5: preparation of 1- (6-chloro-5-nitro-3H-spiro [ benzofuran-2, 4 '-piperidin ] -1' -yl) -2, 2-trifluoroethan-1-one
Figure BDA0002681115790000444
1- (6-chloro-3H-spiro [ benzofuran-2, 4' -piperidine)]-1' -yl) -2, 2-trifluoroethane-1-one (72mg, 0.225mmol) was dissolved in dichloromethane (3 mL), concentrated nitric acid (0.5 mL) was added for reaction for 2 hours, water and dichloromethane were added, the organic layer was separated, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and purified by preparative TLC to give 46mg of a pale yellow oil. Yield: 56 percent. 1 H NMR(400MHz,Chloroform-d)δ7.86(t,J=1.3Hz,1H),6.92(s,1H),4.39(d,J=13.6Hz,1H),3.92(d,J=14.2Hz,1H),3.64(t,J=12.8Hz,1H),3.35(t,J=12.7Hz,1H),3.08(d,J=1.3Hz,2H),2.20–2.01(m,2H),1.96–1.76(m,2H).
And 6: preparation of 6-chloro-5-nitro-3H-spiro [ benzofuran-2, 4' -piperidine ]
Figure BDA0002681115790000451
1- (6-chloro-5-nitro-3H-spiro [ benzofuran-2, 4 '-piperidin ] -1' -yl) -2, 2-trifluoroethane-1-one (46mg, 0.126mmol) was dissolved in methanol, potassium carbonate (52 mg) was added to react at room temperature for 40 minutes, water and methylene chloride (containing 5% methanol) were added to separate an organic layer, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and directly charged into the next reaction without further purification.
And 7: preparation of 6-chloro-5-nitro-3H-spiro [ benzofuran-2, 4 '-piperidine ] -1' -carboxylic acid tert-butyl ester
Figure BDA0002681115790000452
The product obtained above was dissolved in dioxane (3 mL), di-tert-butyl dicarbonate (35mg, 0.152mmol) was added, the reaction was carried out at room temperature for 2 hours, water and ethyl acetate were added, the organic layer was separated, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain 56mg of a yellow oil. 1 H NMR(400MHz,Chloroform-d)δ7.84(t,J=1.2Hz,1H),6.89(s,1H),3.93–3.74(m,2H),3.37(t,J=11.9Hz,2H),3.03(d,J=1.4Hz,2H),1.92(d,J=13.6Hz,2H),1.74(td,J=14.1,12.2,4.5Hz,2H),1.48(s,9H).
And 8: preparation of 5-nitro-6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4 '-piperidine ] -1' -carboxylic acid tert-butyl ester
Figure BDA0002681115790000453
Reacting 6-chloro-5-nitro-3H-spiro [ benzofuran-2, 4' -piperidine]-1 '-Carboxylic acid tert-butyl ester (405mg, 1.10mmol), pyridine-4-boronic acid (540mg, 4.39mmol) and cesium carbonate (716mg, 2.20mmol) were dissolved in dioxane/water (20 mL/2 mL), and [1,1' -bis (diphenylphosphino) ferrocene ] was added under an argon stream]Palladium dichloride (80mg, 0.110mmol), reacting at 100 ℃ overnight, detecting by TLC after the reaction is finished, filtering by diatomite, diluting by ethyl acetate (100 mL), washing by water and saturated saline solution once respectively, and removing waterDried over sodium sulfate, filtered, concentrated under reduced pressure, and chromatographed on silica gel (ethyl acetate/petroleum ether =30% elution) to give 291mg of a yellow solid. Yield: and 76 percent. 1 H NMR(400MHz,Chloroform-d)δ8.71–8.61(m,2H),7.99–7.90(m,1H),7.24–7.16(m,2H),6.67(s,1H),3.81(s,2H),3.39(t,J=11.9Hz,2H),3.11(s,2H),1.96(d,J=13.4Hz,2H),1.78(td,J=14.1,12.3,4.4Hz,3H),1.48(s,9H).
And step 9: preparation of 5-amino-6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4 '-piperidine ] -1' -carboxylic acid tert-butyl ester
Figure BDA0002681115790000454
Reacting 5-nitro-6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4' -piperidine]-1' -carboxylic acid tert-butyl ester (42mg, 0.102mmol) is dissolved in ethanol/water (1.5 mL/0.5 mL), reduced iron powder (34mg, 0.612mmol) and ammonium chloride (33mg, 0.612mmol) are added, an oil bath reaction is carried out at 50 ℃ for 1.5 hours, after the TLC detection reaction is finished, kieselguhr is filtered, reduced pressure concentration is carried out, ethyl acetate is dissolved (15 mL), saturated common salt water washing is carried out, anhydrous sodium sulfate is dried, filtration is carried out, and reduced pressure concentration is carried out to obtain 38mg of yellow solid. Yield: 98 percent. 1 H NMR(400MHz,Chloroform-d)δ8.71–8.60(m,2H),7.46–7.35(m,2H),6.64(s,1H),6.57(s,1H),3.75(s,2H),3.39(t,J=11.9Hz,2H),2.97(d,J=1.1Hz,2H),1.91(d,J=13.6Hz,2H),1.78–1.63(m,4H),1.47(s,9H).
Step 10: preparation of 2- (2-aminopyridin-4-yl) -N- (6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4' -piperidin ] -5-yl) oxazole-4-carboxamide trifluoroacetate
Figure BDA0002681115790000461
Can be prepared by a method analogous to steps 3,4 of example 1 from 5-amino-6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4' -piperidine]Preparation of 2- (2-Aminopyridin-4-yl) -N- (6- (pyridin-4-yl) -3H-spiro [ benzofuran-2, 4 '-piperidine) from (tert-butyl) -1' -carboxylate]-5-yl) oxazole-4-carboxamide trifluoroacetate salt. 1 H NMR(400MHz,DMSO-d 6 )δ10.03(s,1H),8.91(s,1H),8.83–8.66(m,3H),8.63(s,1H),8.12(d,J=6.5Hz,3H),7.67(d,J=5.5Hz,2H),7.41(s,1H),7.39(d,J=1.6Hz,1H),7.21(dd,J=6.4,1.6Hz,1H),6.94(s,1H),3.23(s,6H),2.03(dt,J=18.4,12.3Hz,4H).
Example 30
2- (2-Aminopyridin-4-yl) -N- (6- (1-methyl-1H-pyrazol-4-yl) -3H-spiro [ benzofuran-2, 4' -piperidin ] -5-yl) oxazole-4-carboxamide trifluoroacetate (I-30)
Figure BDA0002681115790000462
2- (2-Aminopyridin-4-yl) -N- (6- (1-methyl-1H-pyrazol-4-yl) -3H-spiro [ benzofuran-2, 4' -piperidine) can be prepared by a similar method to that in example 29]-5-yl) oxazole-4-carboxamide trifluoroacetate salt. 1 H NMR(400MHz,DMSO-d 6 )δ9.71(s,1H),9.01(s,1H),8.68(d,J=42.0Hz,2H),8.16(d,J=6.4Hz,1H),7.98(s,3H),7.72(s,1H),7.47(s,1H),7.40(s,1H),7.24(d,J=6.2Hz,1H),6.93(s,1H),3.84(s,3H),3.23(s,4H),3.15(s,2H),2.12–1.88(m,4H).
Example 31
2- (2-Aminopyridin-4-yl) -N- (6- (1-isopropyl-1H-pyrazol-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-31)
Figure BDA0002681115790000471
2- (2-Aminopyridin-4-yl) -N- (6- (1-isopropyl-1H-pyrazol-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.55(s,1H),8.89(s,1H),8.09(d,J=5.2Hz,1H),8.06(s,1H),7.71(s,1H),7.50(s,1H),7.02(d,J=5.2Hz,2H),6.86(s,1H),6.35(s,2H),4.47(hept,J=6.7Hz,1H),3.04(s,2H),1.44(s,6H),1.37(d,J=6.7Hz,6H).
Example 32
2- (2-Aminopyridin-4-yl) -N- (6- (4-chloro-2-fluorophenyl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-32)
Figure BDA0002681115790000472
2- (2-Aminopyridin-4-yl) -N- (6- (4-chloro-2-fluorophenyl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,Chloroform-d)δ8.67(s,1H),8.26(s,1H),8.20(d,J=5.3Hz,1H),8.14(s,1H),7.31(q,J=9.3,8.0Hz,3H),7.08(d,J=5.4Hz,1H),6.90(s,1H),6.67(s,1H),4.79(s,2H),3.11(s,2H),1.52(s,6H).
Example 33
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (2-methylpyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-33)
Figure BDA0002681115790000473
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (2-methylpyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,Chloroform-d)δ8.89(s,1H),8.65(d,J=5.1Hz,1H),8.26(d,J=4.0Hz,2H),8.17(d,J=5.3Hz,1H),7.22(d,J=5.2Hz,1H),7.04(d,J=5.3Hz,1H),6.85(s,1H),6.68(s,1H),5.05(s,2H),3.11(s,2H),2.63(s,3H),1.52(s,6H).
Example 34
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (6-methylpyridin-3-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-34)
Figure BDA0002681115790000481
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (6-methylpyridin-3-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.62(s,1H),8.77(s,1H),8.46(d,J=2.3Hz,1H),8.08(d,J=5.3Hz,1H),7.71(dd,J=8.0,2.4Hz,1H),7.43(s,1H),7.28(d,J=8.0Hz,1H),6.96(s,1H),6.94(dd,J=5.3,1.5Hz,1H),6.73(s,1H),6.34(s,2H),3.08(s,2H),2.46(s,3H),1.46(s,6H).
Example 35
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (6- (trifluoromethyl) pyridin-3-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-35)
Figure BDA0002681115790000482
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (6- (trifluoromethyl) pyridin-3-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.85(s,1H),8.78(d,J=2.1Hz,1H),8.75(s,1H),8.17–8.01(m,2H),7.93(d,J=8.1Hz,1H),7.37(s,1H),6.98(s,1H),6.96(dd,J=5.3,1.4Hz,1H),6.86(s,1H),6.34(s,2H),3.10(s,2H),1.47(s,6H).
Example 36
2- (2-Aminopyridin-4-yl) -N- (6- (2-methoxypyridin-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-36)
Figure BDA0002681115790000491
2- (2-Aminopyridin-4-yl) -N- (6- (2-methoxypyridin-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,Chloroform-d)δ8.98(s,1H),8.31(d,J=5.3Hz,1H),8.27(s,1H),8.25(s,1H),8.18(d,J=5.4Hz,1H),7.10(d,J=5.4Hz,1H),7.02–6.95(m,1H),6.93(s,1H),6.86(s,1H),6.68(s,1H),4.86(s,2H),4.00(s,3H),3.11(s,2H),1.52(s,6H).
Example 37
2- (2-Aminopyridin-4-yl) -N- (6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-37)
Figure BDA0002681115790000492
Step 1: preparation of 6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-amine
Figure BDA0002681115790000493
6-fluoro-2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran (60mg, 0.284mmol) is dissolved in ethanol/water (6 mL/2 mL), reduced iron powder (95mg, 1.70mmol) and ammonium chloride (91mg, 1.70mmol) are added, the mixture is subjected to oil bath reaction at 50 ℃ for 2 hours, TLC detection reaction is finished, diatomite is subjected to suction filtration and reduced pressure concentration, ethyl acetate is dissolved (15 mL), 1mol/L diluted hydrochloric acid is extracted (15 mL), a water layer is collected, a saturated sodium carbonate solution is adjusted to be alkaline, ethyl acetate (15mL x 2) is extracted, anhydrous sodium sulfate is dried, filtration and reduced pressure concentration is carried out to obtain 42mg of brown oily matter. Yield: 82 percent. 1 H NMR(400MHz,Chloroform-d)δ6.61(dt,J=9.1,1.2Hz,1H),6.44(d,J=11.1Hz,1H),3.35(s,2H),2.90(t,J=1.4Hz,2H),1.44(s,6H).
And 2, step: preparation of 2- (2-aminopyridin-4-yl) -N- (6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000494
2- (2-Aminopyridin-4-yl) -N- (6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide may be prepared from 6-fluoro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-amine by a method analogous to Steps 3,4 of example 1. 1 H NMR(400MHz,DMSO-d 6 )δ9.69(s,1H),8.90(s,1H),8.18–8.02(m,1H),7.41(d,J=8.0Hz,1H),7.05(d,J=4.1Hz,2H),6.73(d,J=10.9Hz,1H),6.36(s,2H),3.00(s,2H),1.43(s,6H).
Example 38
2- (2-Aminopyridin-4-yl) -N- (6-chloro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-38)
Figure BDA0002681115790000501
2- (2-Aminopyridin-4-yl) -N- (6-chloro-2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 37. 1 H NMR(400MHz,DMSO-d 6 )δ9.63(s,1H),8.92(s,1H),8.10(dd,J=5.1,1.0Hz,1H),7.65(s,1H),7.05(dd,J=6.5,1.3Hz,2H),6.94(s,1H),6.34(s,2H),3.04(s,2H),1.43(s,6H).
Example 39
2- (2-Aminopyridin-4-yl) -N- (6- (6-cyanopyridin-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-39)
Figure BDA0002681115790000502
2- (2-Aminopyridin-4-yl) -N- (6- (6-cyanopyridin-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.90(s,1H),8.77(s,1H),8.73(s,1H),8.08(d,J=5.9Hz,1H),8.05(s,2H),7.33(s,1H),6.98(d,J=4.2Hz,2H),6.85(s,1H),6.33(s,2H),3.10(s,2H),1.47(s,6H).
Example 40
2- (2-Aminopyridin-4-yl) -N- (6- (6-methoxypyridin-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-40)
Figure BDA0002681115790000503
2- (2-Aminopyridin-4-yl) -N- (6- (6-methoxypyridin-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.54(s,1H),8.76(s,1H),8.19(d,J=2.4Hz,1H),8.07(d,J=8.5Hz,1H),7.75(dd,J=8.5,2.5Hz,1H),7.47(s,1H),6.97(t,J=1.1Hz,1H),6.93(dd,J=5.2,1.5Hz,1H),6.89–6.81(m,1H),6.72(s,1H),6.31(s,2H),3.86(s,3H),3.08(s,2H),1.46(s,6H).
EXAMPLE 41
2- (2-Aminopyridin-4-yl) -N- (6- (1-ethyl-1H-pyrazol-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-41)
Figure BDA0002681115790000511
2- (2-Aminopyridin-4-yl) -N- (6- (1-ethyl-1H-pyrazol-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.52(s,1H),8.87(s,1H),8.17–8.07(m,1H),8.02(s,1H),7.71(s,1H),7.51(s,1H),7.10–6.96(m,2H),6.83(s,1H),6.33(s,2H),4.13(q,J=7.3Hz,2H),3.04(s,2H),1.44(s,6H),1.35(t,J=7.3Hz,3H).
Example 42
(R) -2- (2-Aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -1-oxo-6- (pyridin-4-yl) isoindolin-5-yl) oxazole-4-carboxylic acid amide (I-42)
Figure BDA0002681115790000512
Step 1: preparation of (R) -6-chloro-2- (2-fluoro-3-hydroxy-3-methylbutyl) -5-nitroisoindol-1-one
Figure BDA0002681115790000513
Methyl 2- (bromomethyl) -5-chloro-4-nitrobenzoate (prepared according to the method reported in WO 2013/079505) (975mg, 3.16mmol) and (R) -4-amino-3-fluoro-2-methylbutan-2-ol (prepared according to the method reported in WO 2015/103453) (459mg, 3.79mmol) were dissolved in methanol (30 mL) and placed in a sealed tube, triethylamine (527. Mu.5) was added, 70 was reacted for 4 hours, and the pressure was reducedConcentrating, dissolving with ethyl acetate (50 mL), washing with 1mol/L HCl solution, extracting the aqueous layer with ethyl acetate (15mL. Multidot.2), combining the organic layers, drying over anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to obtain 903mg of pale yellow solid. Yield: 90 percent. 1 H NMR(400MHz,DMSO-d 6 )δ8.01(s,1H),7.91(s,1H),4.72(d,J=17.9Hz,1H),4.65–4.41(m,2H),4.27(ddd,J=37.0,15.1,2.2Hz,1H),3.64(ddd,J=16.3,15.0,9.1Hz,1H),1.34(d,J=1.8Hz,6H).
Step 2: preparation of (R) -2- (2-fluoro-3-hydroxy-3-methylbutyl) -5-nitro-6- (pyridin-4-yl) isoindol-1-one
Figure BDA0002681115790000521
(R) -6-chloro-2- (2-fluoro-3-hydroxy-3-methylbutyl) -5-nitroisoindol-1-one (300mg, 0.947mmol), pyridine-4-boronic acid (349mg, 2.84mmol) and cesium carbonate (617mg, 1.89mmol) were dissolved in dioxane/water (10 mL/1 mL) and [1,1' -bis (diphenylphosphino) ferrocene ] was added under argon flow]Palladium dichloride (69mg, 0.095mmol), reaction at 95 ℃ overnight, detection by TLC, suction filtration with celite, dilution with ethyl acetate (50 mL), washing with water and saturated brine once, drying over anhydrous sodium sulfate, filtration, concentration under reduced pressure, and silica gel column chromatography (methanol/dichloromethane = 0-3% gradient elution) to obtain 191mg of a yellow solid. Yield: 56 percent. 1 H NMR(400MHz,Chloroform-d)δ8.83–8.63(m,2H),8.04(s,1H),7.90(s,1H),7.27(d,J=1.7Hz,1H),4.81(d,J=18.0Hz,1H),4.67–4.46(m,2H),4.30(ddd,J=36.8,15.1,2.2Hz,1H),3.78–3.60(m,1H),1.35(d,J=1.7Hz,6H).
And 3, step 3: preparation of (R) -5-amino-2- (2-fluoro-3-hydroxy-3-methylbutyl) -6- (pyridin-4-yl) isoindolin-1-one
Figure BDA0002681115790000522
(R) -2- (2-fluoro-3-hydroxy-3-methylbutyl) -5-nitro-6- (pyridin-4-yl) isoindol-1-one (188mg, 0.523mmol) was dissolved in ethanol/water (10 mL/3.3 mL), and reduced iron powder was added(175mg, 3.14mmol) and ammonium chloride (168mg, 3.14mmol), oil bath reaction at 50 ℃ for 1 hour, TLC detection reaction completion, suction filtration of diatomite, concentration under reduced pressure, dissolution of ethyl acetate (45 mL), washing with saturated saline, drying with anhydrous sodium sulfate, filtration, and concentration under reduced pressure to obtain 65mg of yellow solid. Yield: 38 percent. 1 H NMR(400MHz,Methanol-d 4 )δ8.68–8.53(m,2H),7.62–7.49(m,2H),7.46(s,1H),6.91(s,1H),4.64–4.36(m,3H),4.17–4.00(m,1H),3.68(td,J=15.2,9.3Hz,1H),1.28(d,J=1.7Hz,6H).
And 4, step 4: preparation of (R) -2- (2-aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -1-oxo-6- (pyridin-4-yl) isoindolin-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000523
(R) -2- (2-aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -1-oxo-6- (pyridin-4-yl) isoindolin-5-yl) oxazole-4-carboxamide can be prepared from (R) -5-amino-2- (2-fluoro-3-hydroxy-3-methylbutyl) -6- (pyridin-4-yl) isoindolin-1-one by a similar method to that described in example 1, steps 3, 4. 1 H NMR(400MHz,DMSO-d 6 )δ9.91(s,1H),8.89(s,1H),8.81–8.60(m,2H),8.19(s,1H),8.09(d,J=5.2Hz,1H),7.68(s,1H),7.57(d,J=5.0Hz,2H),6.95(s,1H),6.90(d,J=5.1Hz,1H),6.36(s,2H),4.95(s,1H),4.76–4.59(m,2H),4.48(dd,J=49.5,9.1Hz,1H),3.99(dd,J=38.6,14.9Hz,1H),3.74(q,J=15.0,14.5Hz,1H),1.21(dd,J=14.2,3.8Hz,6H).
Example 43
2- (2-Aminopyridin-4-yl) -N- (6- (5-fluoropyridin-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide (I-43)
Figure BDA0002681115790000531
2- (2-Aminopyridin-4-yl) -N- (6- (5-fluoropyridin-3-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4 can be prepared by a similar method to that in example 24-a carboxamide. 1 H NMR(400MHz,DMSO-d 6 )δ8.68(s,1H),8.54(t,J=2.2Hz,2H),8.29(s,1H),8.18(d,J=5.3Hz,1H),8.04(s,1H),7.60–7.49(m,1H),7.10(dd,J=5.4,1.4Hz,1H),7.04–6.95(m,1H),6.69(s,1H),4.75(s,2H),3.22–3.04(m,2H),1.53(s,6H).
Example 44
2- (2-Aminopyridin-4-yl) -N- (6- (3-fluoropyridin-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-44)
Figure BDA0002681115790000532
2- (2-Aminopyridin-4-yl) -N- (6- (3-fluoropyridin-4-yl) -2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ8.64(s,1H),8.62(d,J=1.6Hz,1H),8.54(d,J=4.9Hz,1H),8.26(s,1H),8.18(d,J=5.3Hz,1H),8.00(s,1H),7.37(t,J=5.6Hz,1H),7.08(dd,J=5.3,1.4Hz,1H),6.90(s,1H),6.70(s,1H),4.88(s,2H),3.12(s,2H),1.53(s,6H).
Example 45
4- (6- (2- (2-aminopyridin-4-yl) oxazole-4-carboxamido) -5-ethoxy-1-methyl-1H-benzo [ d ] imidazol-2-yl) -2, 2-dimethylbutanoic acid methyl ester (I-45)
Figure BDA0002681115790000541
Step 1: preparation of 5-chloro-2-ethoxy-4-nitroaniline
Figure BDA0002681115790000542
2-amino-4-chloro-5-nitrophenol (2.0 g,10.6 mmol) was dissolved in N, N-dimethylformamide (40 mL), potassium carbonate (2.2 g,15.9 mmol) and bromoethane (1.392g, 12.7 mmol) were added, reaction was carried out overnight at room temperature, water and ethyl acetate (150 mL) were added, the organic layer was separated, and water and saturated brine were each washed once and anhydrousDried over sodium sulfate, filtered and concentrated under reduced pressure to give 2.0g of a brown solid. Yield: 87 percent. 1 H NMR(400MHz,DMSO-d 6 )δ7.54(s,1H),6.73(s,1H),6.49(s,2H),4.10(q,J=6.9Hz,2H),1.36(t,J=6.9Hz,3H).
And 2, step: preparation of tert-butyl (5-chloro-2-ethoxy-4-nitrophenyl) carbamate
Figure BDA0002681115790000543
5-chloro-2-ethoxy-4-nitroaniline (2.0 g, 9.23mmol) was dissolved in dichloromethane (30 mL), di-tert-butyl dicarbonate (2.42g, 11.1 mmol) and 4-dimethylaminopyridine (11 mg) were added, the reaction was refluxed for 3 hours, concentrated under reduced pressure, and subjected to silica gel column chromatography (ethyl acetate/petroleum ether =1% elution) to give 1.68g of a golden yellow solid. Yield: 57 percent. 1 H NMR(400MHz,Chloroform-d)δ8.38(s,1H),7.51(s,1H),4.17(q,J=7.0Hz,2H),1.53(d,J=14.0Hz,12H).
And step 3: preparation of tert-butyl (2-ethoxy-5- (methylamino) -4-nitrophenyl) carbamate
Figure BDA0002681115790000544
Tert-butyl (5-chloro-2-ethoxy-4-nitrophenyl) carbamate (895mg, 2.83mmol) and an alcoholic solution of methylamine (40 mL) were added to a sealed tube, and the mixture was reacted for 36 hours in a 70-tube column, followed by concentration under reduced pressure and silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =5% -10%) to obtain 600mg of an orange solid. Yield: 68 percent. 1 H NMR(400MHz,DMSO-d 6 )δ8.35(q,J=4.9Hz,1H),8.20(s,1H),7.62(s,1H),7.48(s,1H),4.06(q,J=6.9Hz,2H),2.93(d,J=4.9Hz,3H),1.50(s,9H),1.36(t,J=6.9Hz,3H).
And 4, step 4: preparation of tert-butyl (4-amino-2-ethoxy-5- (methylamino) phenyl) carbamate
Figure BDA0002681115790000551
Dissolving (2-ethoxy-5- (methylamino) -4-nitrophenyl) carbamic acid tert-butyl ester (565mg, 1.81mmol) in methanol (25 mL), adding 10% palladium carbon (200 mg) and ammonium formate (686mg, 10.9mmol), reacting at 60 ℃ for 2 hours, detecting by TLC after the reaction is finished, leaching by using kieselguhr, concentrating under reduced pressure, dissolving ethyl acetate (45 mL), washing by using saturated salt water, drying by using anhydrous sodium sulfate, filtering, concentrating under reduced pressure to obtain 485mg of blue-purple oily substance, and directly putting into the next reaction without further purification. 1 H NMR(400MHz,DMSO-d 6 )δ7.42(s,1H),6.63(s,1H),6.31(s,1H),4.37(s,2H),4.22(s,1H),3.85(s,2H),2.64(s,3H),1.43(s,9H),1.26(t,J=6.9Hz,3H).
And 5: preparation of methyl 4- (6- ((tert-butoxycarbonyl) amino) -5-ethoxy-1-methyl-1H-benzo [ d ] imidazol-2-yl) -2, 2-dimethylbutyrate
Figure BDA0002681115790000552
Tert-butyl (4-amino-2-ethoxy-5- (methylamino) phenyl) carbamate (485mg, 1.72mmol) and 5-methoxy-4, 4-dimethyl-5-oxopentanoic acid (315mg, 1.81mmol) were dissolved in N, N-dimethylformamide (15 mL), 2- (7-azabenzotriazole) -N, N' -tetramethyluronium hexafluorophosphate (983mg, 2.59mmol) and N, N-diisopropylethylamine (712 μ 7, 4.31mmol) were added, the mixture was reacted overnight at room temperature, ethyl acetate (60 mL) was diluted, water and saturated saline were washed once each, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure to give a residue dissolved in acetic acid (15 mL), reacted at room temperature for 12 hours, added water and ethyl acetate (60 mL), the organic layer was separated, saturated sodium bicarbonate and saturated common salt were washed with water, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, silica gel (ethyl acetate/petroleum ether =20% -40% gradient column chromatography) was eluted to give 391mg of a white solid. Yield: and 54 percent. 1 H NMR(400MHz,DMSO-d 6 )δ7.79(s,2H),7.14(s,1H),4.06(q,J=7.1Hz,2H),3.65(d,J=1.4Hz,3H),3.59(d,J=1.4Hz,3H),2.74(t,J=8.4Hz,2H),2.03–1.92(m,2H),1.48(d,J=1.5Hz,9H),1.37(td,J=6.9,1.4Hz,3H),1.21(d,J=1.5Hz,6H).
And 6: preparation of methyl 4- (6-amino-5-ethoxy-1-methyl-1H-benzo [ d ] imidazol-2-yl) -2, 2-dimethylbutyrate
Figure BDA0002681115790000553
Reacting 4- (6- ((tert-butoxycarbonyl) amino) -5-ethoxy-1-methyl-1H-benzo [ d]Imidazol-2-yl) -2, 2-dimethylbutanoic acid methyl ester (387mg, 0.922mmol) was dissolved in dichloromethane (20 mL), trifluoroacetic acid (5 mL) was added to react at room temperature for 1 hour, concentrated under reduced pressure, ethyl acetate and saturated sodium bicarbonate solution were added to extract, the organic layer was separated, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 283mg of a brown solid. Yield: 96 percent. 1 H NMR(400MHz,DMSO-d 6 )δ6.93(s,1H),6.69–6.56(m,1H),4.65(s,2H),3.98(q,J=6.9Hz,2H),3.59(d,J=1.5Hz,3H),3.55(d,J=1.4Hz,3H),2.66(t,J=8.5Hz,2H),1.93(t,J=8.4Hz,2H),1.42–1.30(m,3H),1.21(s,6H).
And 7: preparation of methyl 4- (6- (2- (2-aminopyridin-4-yl) oxazole-4-carboxamido) -5-ethoxy-1-methyl-1H-benzo [ d ] imidazol-2-yl) -2, 2-dimethylbutyrate
Figure BDA0002681115790000561
Can be prepared by a method similar to steps 3 and 4 of example 1 from 4- (6-amino-5-ethoxy-1-methyl-1H-benzo [ d]Preparation of methyl imidazol-2-yl) -2, 2-dimethylbutyrate 4- (6- (2- (2-aminopyridin-4-yl) oxazole-4-carboxamido) -5-ethoxy-1-methyl-1H-benzo [ d]Imidazol-2-yl) -2, 2-dimethylbutanoic acid methyl ester. 1 H NMR(400MHz,DMSO-d 6 )δ9.66(s,1H),8.96(s,1H),8.41(s,1H),8.12(d,J=5.2Hz,1H),7.29(s,1H),7.03(d,J=5.7Hz,2H),6.39(s,2H),4.20(q,J=6.9Hz,2H),3.69(s,3H),3.60(s,3H),2.85–2.71(m,2H),2.08–1.94(m,2H),1.50(t,J=6.9Hz,3H),1.22(s,6H).
Example 46
2- (2-Aminopyridin-4-yl) -N- (6-methoxy-2- (piperidin-4-yl) -2H-indazol-5-yl) oxazole-4-carboxamide trifluoroacetate salt (I-46)
Figure BDA0002681115790000562
Step 1: preparation of 2-fluoro-4-methoxy-5-nitrobenzaldehyde
Figure BDA0002681115790000563
The concentrated sulfuric acid (1.3 mL) is bathed in ice salt to-15 ℃, 2-fluoro-4-methoxybenzaldehyde (3.0g, 19.5 mmol) is added, concentrated nitric acid is slowly dripped, the temperature of the reaction liquid is controlled to be below-10 ℃, the reaction is kept for 45 minutes, the reaction liquid is poured into crushed ice, a large amount of solid is separated out, the solid is filtered and dried in vacuum, and the solid obtained is pulped with petroleum ether, filtered and dried in vacuum to obtain 3.49g of yellow white solid. Yield: and 90 percent. 1 H NMR(400MHz,Chloroform-d)δ10.21(s,1H),8.46(d,J=7.2,1H),6.87(d,J=11.5Hz,1H),4.06(s,3H).
Step 2: preparation of 2-azido-4-methoxy-5-nitrobenzaldehyde
Figure BDA0002681115790000564
2-fluoro-4-methoxy-5-nitrobenzaldehyde (2.0g, 10.0mmol) was dissolved in dimethyl sulfoxide (10 mL), sodium azide (1.31g, 20.1mmol) was added to the solution to react at room temperature for 30 minutes, water and ethyl acetate (250 mL) were added to the solution, and the ethyl acetate layer was washed once with water and saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain 2.13g of a brown solid. Yield: 95 percent. 1 H NMR(400MHz,Chloroform-d)δ10.19(s,1H),8.45(s,1H),6.80(s,1H),4.09(s,3H).
And step 3: preparation of 4- (6-methoxy-5-nitro-2H-indazol-2-yl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0002681115790000571
2-azido-4-methoxy-5-nitrobenzaldehyde (1.0g, 4.50mmol) was dissolved in dichloro-benzeneMethane (20 mL), 4-aminopiperidine-1-carboxylic acid tert-butyl ester (902mg, 4.50mmol) was dissolved in methylene chloride and added to the reaction mixture
Figure BDA0002681115790000572
Molecular sieves (2 g) reacted at room temperature for 1.5 hours and then supplemented with tert-butyl 4-aminopiperidine-1-carboxylate (270mg, 1.35mmol) and
Figure BDA0002681115790000573
molecular sieve (2 g), reacting for 14 hours at room temperature, filtering with diatomite, concentrating under reduced pressure, dissolving in anhydrous toluene (30 mL), reacting for 1 hour at 120 ℃, concentrating under reduced pressure, and performing silica gel column chromatography (gradient elution with ethyl acetate/petroleum ether =20% -40%) to obtain 1.46g of yellow solid. Yield: 86 percent. 1 H NMR(400MHz,Chloroform-d)δ8.21(s,1H),8.07(s,1H),7.10(s,1H),4.52(tt,J=11.6,4.0Hz,1H),4.33(s,2H),3.96(s,3H),2.94(s,2H),2.32–2.18(m,2H),2.08(qd,J=12.2,4.6Hz,2H),1.48(s,9H).
And 4, step 4: preparation of 4- (5-amino-6-methoxy-2H-indazol-2-yl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0002681115790000574
Dissolving 4- (6-methoxy-5-nitro-2H-indazol-2-yl) piperidine-1-carboxylic acid tert-butyl ester (233mg, 0.675mmol) in ethanol/water (12 mL/4 mL), adding reduced iron powder (226mg, 4.05mmol) and ammonium chloride (217mg, 4.05mmol), reacting in an oil bath at 50 ℃ for 6 hours, detecting the reaction by TLC, carrying out suction filtration on kieselguhr, concentrating under reduced pressure, dissolving ethyl acetate (60 mL), washing with water and saturated saline water once, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, and directly putting into the next step without further purification. 1 H NMR(400MHz,Chloroform-d)δ7.61(d,J=0.9Hz,1H),6.93(s,1H),6.75(s,1H),4.42(tt,J=11.7,4.1Hz,1H),4.25(d,J=31.8Hz,2H),3.91(s,3H),2.91(s,2H),2.19(d,J=12.9Hz,2H),2.04(s,3H),1.48(s,9H).
And 5: preparation of 2- (2-aminopyridin-4-yl) -N- (6-methoxy-2- (piperidin-4-yl) -2H-indazol-5-yl) oxazole-4-carboxamide trifluoroacetate salt
Figure BDA0002681115790000575
2- (2-Aminopyridin-4-yl) -N- (6-methoxy-2- (piperidin-4-yl) -2H-indazol-5-yl) oxazole-4-carboxamide trifluoroacetate can be prepared from tert-butyl 4- (5-amino-6-methoxy-2H-indazol-2-yl) piperidine-1-carboxylate by a similar method as in example 2. 1 H NMR(400MHz,DMSO-d 6 )δ9.52(s,1H),9.11(s,1H),8.81(d,J=11.1Hz,1H),8.57(s,2H),8.37(s,1H),8.14(d,J=6.3Hz,1H),7.84(s,2H),7.41(s,1H),7.27(d,J=6.2Hz,1H),7.17(s,1H),4.82–4.69(m,1H),4.01(s,3H),3.46(d,J=12.6Hz,2H),3.22–3.06(m,2H),2.42–2.10(m,4H).
Example 47
2- (2-Aminopyridin-4-yl) -N- (2- (hydroxymethyl) -2-methyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-47)
Figure BDA0002681115790000581
Step 1: preparation of (6-chloro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-yl) methanol
Figure BDA0002681115790000582
(6-chloro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-yl) methanol can be prepared by a method similar to steps 1 to 5 in example 14. 1 H NMR(400MHz,Chloroform-d)δ7.85(d,J=1.6Hz,1H),6.86(s,1H),3.76(d,J=5.6Hz,1H),3.66(d,J=7.3Hz,1H),3.40–3.31(m,1H),2.99–2.90(m,1H),1.84(dd,J=7.3,5.7Hz,1H),1.48(s,3H),1.26(dd,J=7.5,6.8Hz,1H).
Step 2: preparation of (2-methyl-5-nitro-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-2-yl) methanol
Figure BDA0002681115790000583
6-chloro-2-methyl-5-nitro-2, 3-dihydrobenzofuran-2-yl) methanol (58mg, 0.238mmol), pyridine-4-boronic acid (88mg, 0.714mmol) and cesium carbonate (155mg, 0.476mmol) were dissolved in dioxane/water (5 mL/0.5 mL) and [1,1' -bis (diphenylphosphino) ferrocene was added under argon flow]Palladium dichloride (17mg, 0.024mmol), reacted at 100 ℃ overnight, after completion of TLC detection, filtered through celite, diluted with ethyl acetate (30 mL), washed once with water and saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and chromatographed on silica gel (ethyl acetate/petroleum ether = 60%) to give 58mg of a pale yellow solid. Yield: 85 percent. 1 H NMR(400MHz,Chloroform-d)δ8.68–8.57(m,2H),7.94(t,J=1.3Hz,1H),7.20–7.14(m,2H),6.54(s,1H),3.82(d,J=12.0Hz,1H),3.67(d,J=12.1Hz,1H),3.45(dd,J=16.2,1.3Hz,1H),3.01(dd,J=16.2,1.2Hz,1H),2.52(s,1H),1.51(s,3H).
And step 3: preparation of (5-amino-2-methyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-2-yl) methanol
Figure BDA0002681115790000584
Dissolving (2-methyl-5-nitro-6- (pyridine-4-yl) -2, 3-dihydrobenzofuran-2-yl) methanol (50mg, 0.175mmol) in ethanol/water (6 mL/2 mL), adding reduced iron powder (58mg, 1.05mmol) and ammonium chloride (56mg, 1.05mmol), performing oil bath reaction at 50 ℃ for 1 hour, detecting the reaction by TLC, performing suction filtration by using kieselguhr, concentrating under reduced pressure, dissolving ethyl acetate (20 mL), washing with saturated salt water, drying with anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to obtain 38mg of yellow solid. Yield: 85 percent. 1 H NMR(400MHz,Chloroform-d)δ8.70–8.60(m,2H),7.44–7.36(m,2H),6.65(s,1H),6.54(s,1H),3.75–3.61(m,2H),3.25(d,J=16.2Hz,1H),2.88(d,J=15.9Hz,1H),1.44(s,3H).
And 4, step 4: preparation of 2- (2-aminopyridin-4-yl) -N- (2- (hydroxymethyl) -2-methyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide
Figure BDA0002681115790000591
2- (2-Aminopyridin-4-yl) -N- (2- (hydroxymethyl) -2-methyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared from (5-amino-2-methyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-2-yl) methanol by a similar method as that described in example 1, steps 3, 4. 1 H NMR(400MHz,DMSO-d 6 )δ9.70(s,1H),8.75(s,1H),8.65–8.51(m,2H),8.08(d,J=5.3Hz,1H),7.47–7.40(m,2H),7.39(s,1H),7.03–6.91(m,2H),6.75(s,1H),6.33(s,2H),5.13(t,J=5.8Hz,1H),3.49(qd,J=11.5,5.8Hz,2H),3.28(s,1H),3.17(d,J=5.3Hz,1H),2.92(d,J=16.3Hz,1H),1.39(s,3H).
Example 48
2- (2-Aminopyrimidin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-48)
Figure BDA0002681115790000592
2- (2-Aminopyrimidin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 24. 1 H NMR(400MHz,Methanol-d 4 )δ8.56(s,1H),8.55–8.50(m,2H),8.45(d,J=5.2Hz,1H),7.55–7.48(m,2H),7.45(s,1H),7.28(d,J=5.1Hz,1H),6.77(s,1H),1.51(s,6H).
Example 49
2- (6-Aminopyrimidin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-49)
Figure BDA0002681115790000601
2- (6-Aminopyrimidin-4-yl) -N- (2, 2-dimethyl-6- (pyridin-4-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method as in example 24. 1 H NMR(400MHz,Chloroform-d)δ8.82–8.69(m,3H),8.65(d,J=1.2Hz,1H),8.34(s,1H),8.13(s,1H),7.42(d,J=5.5Hz,2H),6.86(d,J=1.2Hz,1H),6.70(s,1H),5.59(s,2H),3.12(d,J=1.2Hz,2H),1.53(s,6H).
Example 50
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (pyrimidin-5-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-50)
Figure BDA0002681115790000602
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (pyrimidin-5-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that described in example 24. 1 H NMR(400MHz,Chloroform-d)δ9.25(s,1H),8.85(s,2H),8.53(s,1H),8.28(s,1H),8.18(d,J=5.4Hz,1H),7.87(s,1H),7.12(d,J=5.9Hz,1H),7.01(s,1H),6.70(s,1H),4.72(s,2H),3.12(s,2H),1.53(s,6H).
Example 51
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (2-methylpyrimidin-5-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxylic acid amide (I-51)
Figure BDA0002681115790000603
2- (2-Aminopyridin-4-yl) -N- (2, 2-dimethyl-6- (2-methylpyrimidin-5-yl) -2, 3-dihydrobenzofuran-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.84(s,1H),8.75(s,1H),8.70(s,2H),8.08(d,J=5.3Hz,1H),7.32(s,1H),6.99(d,J=4.7Hz,2H),6.83(s,1H),6.31(s,2H),3.09(s,2H),2.60(s,3H),1.47(s,6H).
Example 52
(R) -2- (2-Aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -1-oxo-6- (pyridin-3-yl) isoindolin-5-yl) oxazole-4-carboxylic acid amide (I-52)
Figure BDA0002681115790000611
(R) -2- (2-Aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -1-oxo-6- (pyridin-3-yl) isoindolin-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that described in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.82(s,1H),8.88(s,1H),8.70(d,J=2.3Hz,1H),8.67(dd,J=4.8,1.6Hz,1H),8.20(s,1H),8.09(d,J=5.2Hz,1H),7.96(dt,J=7.8,2.0Hz,1H),7.67(s,1H),7.56(dd,J=7.9,4.8Hz,1H),6.93(t,J=1.1Hz,1H),6.89(dd,J=5.3,1.4Hz,1H),6.34(s,2H),4.94(s,1H),4.75–4.59(m,2H),4.58–4.39(m,1H),4.08–3.91(m,1H),3.74(td,J=15.6,9.4Hz,1H),1.19(dd,J=4.5,1.6Hz,6H).
Example 53
(R) -2- (2-Aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -6- (1-methyl-1H-pyrazol-4-yl) -1-oxoisoindol-5-yl) oxazole-4-carboxamide (I-53)
Figure BDA0002681115790000612
(R) -2- (2-Aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -6- (1-methyl-1H-pyrazol-4-yl) -1-oxoisoindol-5-yl) oxazole-4-carboxamide can be prepared by a similar method to that in example 24. 1 H NMR(400MHz,DMSO-d 6 )δ9.82(s,1H),8.99(s,1H),8.30(s,1H),8.20(s,1H),8.13(d,J=5.5Hz,1H),7.85(s,1H),7.72(s,1H),7.00(d,J=3.3Hz,2H),6.37(s,2H),4.93(s,1H),4.69–4.55(m,2H),4.47(dd,J=49.8,9.8Hz,1H),4.09–3.86(m,4H),3.72(td,J=15.4,9.2Hz,1H),1.19(d,J=4.2Hz,6H).
Example 54
(R) -2- (2-aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -6- (6-methylpyridin-3-yl) -1-oxoisoindol-5-yl) oxazole-4-carboxamide (I-54)
Figure BDA0002681115790000621
Can be obtained by analogy with that in example 24Method for the preparation of (R) -2- (2-aminopyridin-4-yl) -N- (2- (2-fluoro-3-hydroxy-3-methylbutyl) -6- (6-methylpyridin-3-yl) -1-oxoisoindol-5-yl) oxazole-4-carboxamide. 1 H NMR(400MHz,Methanol-d 4 )δ8.60(d,J=2.3Hz,1H),8.57(s,1H),8.42(s,1H),8.04(d,J=5.5Hz,1H),7.89(dd,J=8.0,2.4Hz,1H),7.78(s,1H),7.52(d,J=8.1Hz,1H),7.10(s,1H),7.02(dd,J=5.5,1.6Hz,1H),4.78–4.65(m,2H),4.62–4.48(m,1H),4.16(dd,J=37.9,14.8Hz,1H),3.80(td,J=15.2,9.6Hz,1H),2.66(s,3H),1.31(s,6H).
Example 55
In this example, the following comparative compounds 1-4 were synthesized using a similar method.
Figure BDA0002681115790000622
Comparative Compound 1 is identical to Compound I-5, except that the ortho position to the pyridine N atom is a methyl group. 1 H NMR(400MHz,DMSO-d 6 )δ9.99(s,1H),9.02(s,1H),8.72(d,J=5.1Hz,1H),8.19(s,1H),7.89–7.81(m,1H),7.75(dd,J=5.2,1.6Hz,1H),6.76(s,1H),3.89(dd,J=5.7,3.2Hz,4H),3.01(s,2H),2.84(t,J=4.5Hz,4H),2.59(s,3H),1.41(s,6H).
Figure BDA0002681115790000623
Comparative Compound 2 is identical to Compound I-1, except that the pyridine ortho to the N atom is a methyl group. 1 H NMR(300MHz,DMSO-d 6 )δ10.01(s,1H),9.00(s,1H),8.68(d,J=5.0Hz,1H),8.22(s,1H),7.80(d,J=6.7Hz,2H),6.70(s,1H),4.63(t,J=5.1Hz,1H),3.43(d,J=5.3Hz,2H),3.00(s,2H),2.94(d,J=11.1Hz,2H),2.68(t,J=10.7Hz,2H),2.60(s,3H),1.91–1.77(m,2H),1.68–1.51(m,3H),1.41(s,6H).
Figure BDA0002681115790000624
Comparative Compound 3 is the same as Compound I-5 except thatThe ortho position of the pyridine N atom is not substituted. 1 H NMR(400MHz,Chloroform-d)δ10.00(s,1H),8.95–8.76(m,2H),8.36(d,J=25.4Hz,2H),8.01–7.86(m,2H),6.65(s,1H),4.01(t,J=4.5Hz,4H),3.04(s,2H),2.93(dd,J=5.5,3.3Hz,4H),1.49(s,6H).
Figure BDA0002681115790000631
Comparative Compound 4 is identical to Compound I-5, except that the amino group ortho to the pyridine N atom is substituted with an acetyl group. 1 H NMR(400MHz,DMSO-d 6 )δ10.81(s,1H),9.91(s,1H),9.01(s,1H),8.81(s,1H),8.57(dd,J=5.1,0.8Hz,1H),8.20(s,1H),7.64(dd,J=5.2,1.6Hz,1H),6.75(s,1H),3.87(dd,J=5.9,3.2Hz,4H),3.01(s,2H),2.83(dd,J=5.4,3.3Hz,4H),2.14(s,3H),1.41(s,6H)。
Test example 1
Inhibition of IRAK4 kinase Activity by Compounds
1. Test method
Using Z' -LYTE TM The kinase kit detects the kinase inhibitory activity of the compound. The method mainly comprises the following steps: compound solutions or solvent controls at different concentrations were added at 2.5 μ L/well in 384-well plates, with two replicate wells per concentration. The solvent control components were three: 0% phosphorylation, 0% inhibition and 100% phosphorylation. The enzyme and substrate were diluted with kinase buffer to a mixture of 1.5 ng/well and 2. Mu.M/well, and the mixture was used in the test compound group and the 0% phosphorylated and 0% inhibited groups, with 5. Mu.L per well. In the 100% phosphorylation group, 5. Mu.L of the phosphorylated substrate solution was added to each well at a concentration of 2. Mu.M/well. The test compound group and the 0% inhibition group were added with 2.5. Mu.L of ATP (final concentration: 400. Mu.M) to initiate the reaction, and the 384-well plate was placed in an oven at 27 ℃ for 1 hour. Adding secondary reaction reagent, 5 μ L per well, performing oven reaction at 27 deg.C for 1h, adding 5 μ L stop reagent to stop reaction, and reading plate with fluorescence detection microplate reader (Synergy 2, BIOTEK) to respectively read the emitted light at 400nM excitation light, 445nM wavelength and 520nM wavelength. Inhibition rate and half inhibition IC 50 The ratio of the two emitted lights and the percentage of phosphorylation were determined.
2. Results of the experiment
As can be seen from Table 2, the compound of the invention has an obvious inhibitory effect on IRAK4 kinase, wherein the inhibitory activity of the compound I-5 on IRAK4 is obviously improved compared with that of the comparative compounds 1,3 and 4, and the inhibitory activity of the compound I-1 is obviously improved compared with that of the comparative compound 2, which shows that the N atom ortho amino group of pyridine has an important effect on the inhibitory activity of IRAK4, and the activity of the compound is obviously deteriorated after the N atom ortho amino substituent of pyridine is substituted.
TABLE 2 detection of IRAK4 kinase Activity by Compounds
Figure BDA0002681115790000632
Figure BDA0002681115790000641
Test example 2
Growth inhibition assay of OCI-LY-10 cell line (MYD 88L 265P mutant-containing cells) with Compounds
The Cell proliferation inhibition effect of the compound was examined by CCK-8 (Cell Counting Kit-8) staining. The CCK-8 dyeing method comprises the following steps: OCI-LY-10 cells in logarithmic growth phase are inoculated into 96-well culture plates according to the growth rate of the cells at different concentrations in 90 mu L/well, after the cells grow overnight, compounds with different concentrations in 10 mu L/well are added, each concentration is provided with three multiple wells, and corresponding solvent control wells and blank control wells without cells are provided. Cells are placed in an incubator for compound action for 72h, then cell culture fluid is discarded, and 10 mu L/hole of CCK-8 reagent is added. And (3) putting the 96-well plate back to the cell culture box for reaction for 2h, and then placing the 96-well plate under a 450nm wavelength of an enzyme labeling instrument for determining the OD value. The inhibition rate of the compound on cell growth is calculated by the formula: inhibition% = (control OD value-administered OD value)/control OD value × 100%. Half the inhibition IC 50 The values were determined by fitting inhibition curves with a four-parameter method using a microplate reader with software. EXAMPLE 23 (I-23) Compound IC 50 The value was 0.291. + -. 0.096. Mu.M; EXAMPLE 24 (I-24) Compound IC 50 The value is 1.586. + -. 0.280. Mu.M。
Test example 3
Growth inhibition experiment of compound on U2932 cell line (without MYD 88L 265P mutant cell)
The Cell proliferation inhibition effect of the compound was examined by CCK-8 (Cell Counting Kit-8) staining. The CCK-8 dyeing method comprises the following steps: OCI-LY-10 cells in logarithmic growth phase are inoculated into 96-well culture plates according to the growth rate of the cells at different concentrations in 90 mu L/well, after the cells grow overnight, compounds with different concentrations in 10 mu L/well are added, each concentration is provided with three multiple wells, and corresponding solvent control wells and blank control wells without cells are provided. Cells are placed in an incubator for compound action for 72h, then cell culture fluid is discarded, and 10 mu L/hole of CCK-8 reagent is added. And (3) putting the 96-well plate back to the cell culture box for reaction for 2h, and then placing the 96-well plate under a 450nm wavelength of an enzyme labeling instrument for determining the OD value. The inhibition rate of the compound on cell growth is calculated by the formula: inhibition% = (control OD value-administered OD value)/control OD value × 100%. Half the inhibition IC 50 The values were determined by fitting inhibition curves to the microplate reader with software using a four-parameter method. EXAMPLE 24 (I-24) Compound IC 50 Values greater than 50 μ M indicate that the compound of example 24 (I-24) has selective inhibitory effects on MYD 88L 265P mutant diffuse large B-cell lymphoma.
Test example 4
Test of inhibition of tyrosine kinase Activity by Compounds
The inhibition of a series of kinase activities by the compounds was examined by ELISA. This series of kinases includes VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-alpha, PDGFR-beta, RET, C-Kit, FLT3, EGFR, erbB2, erbB4, src, abl, EPH-A2, IGF1R, IR, FGFR1, FGFR2, FGFR3, FGFR4, BTK, FAK, CSF1R and ITK, all available from Europhins Inc.
The ELISA main steps are as follows: enzyme reaction substrate Poly (Glu, tyr) was treated with potassium ion-free PBS 4:1 After reaction for 12-16h at 37 ℃, the liquid in the wells is discarded after the enzyme-linked immunosorbent assay plate is coated. And washing the plate with T-PBS for three times, and drying the ELISA plate in a 37 ℃ oven for 1-2h for later use. Add a reaction buffer diluted ATP (5 mM final concentration) solution, compound or solvent control to each well, followed by addition of a kinase starterAnd (4) performing dynamic reaction, and performing shaking table reaction at 37 ℃ for 1h. The plate was washed three times with T-PBS and reacted for 0.5h at 37 ℃ with the addition of the antibody PY 99. After washing the plate with T-PBS for three times, adding horseradish peroxidase labeled goat anti-mouse IgG for shaking table reaction at 37 ℃ for 0.5h. Discarding the liquid in the hole, washing the plate again, adding 100 mu L/hole of OPD color development liquid, and reacting for 1-10min at 25 ℃ in a dark place. Addition of H 2 SO 4 The reaction is stopped, and the reading is carried out by using a microplate reader with adjustable wavelength, wherein the wavelength is 490nm. The inhibition rate calculation formula is as follows: inhibition% = [1- (compound OD value-no enzyme control OD value)/(negative control OD value-no enzyme control OD value) ]]×100%。IC 50 The values were determined by fitting inhibition curves with a four-parameter method using a microplate reader with software.
As can be seen from Table 3, the compounds of example 23 (I-23) and example (I-24) have better selectivity on tyrosine kinase spectrum and simultaneously have inhibition effects on VEGFR-3, RET, FLT3 and ErbB 2.
TABLE 3 detection of the Activity of Compounds on the series of tyrosine kinases
Figure BDA0002681115790000661
Test example 5
Compounds FLT3 kinase and FLT3-ITD, FLT3 D835Y Mutant kinase activity assay
The inhibition of the kinase activity by the compound was calculated by measuring the ability of the kinase to phosphorylate substrates by Enzyme-Linked Immunosorbent Assay (ELISA). The kinase adopts Flt-3 and Flt-3 ITD And Flt-3 D835Y (available from Eurofins). The ELISA main steps are as follows: enzyme reaction substrate Poly (Glu, tyr) 4:1 Diluting to 2.5 mu g/hole with PBS without potassium ions, reacting at 37 ℃ for 12-16h, and coating an ELISA plate for later use. Reaction buffer (50mM HEPES pH 7.4, 20mM MgCl) was added to each well 2 ,0.1mM MnCl 2 ,0.2mM Na 3 VO 4 1mM DTT) was added to a compound or solvent control, followed by addition of kinase to initiate the reaction, followed by shaking at 37 ℃ for 1h. The plate was washed three times with T-PBS, and the antibody PY99 was added to the plate and the plate was subjected to shake reaction at 37 ℃ for 0.5 hour. After washing the plate with T-PBS, addHorse radish peroxidase-labeled goat anti-mouse IgG was subjected to shake reaction at 37 ℃ for 0.5h. After washing the plate again, the content of H is 0.03% 2 O 2 And 2mg/mL OPD developing solution, and reacting for 1-10min at 25 ℃ in a dark place. Addition of 2M H 2 SO 4 The reaction was stopped and read using a tunable wavelength microplate reader (SpectraMax Plus384, molecular Devices) at a wavelength of 490nm. IC (integrated circuit) 50 Values were obtained from the inhibition curves.
As can be seen from the results in Table 4, example 24 (I-24) showed no effect on FLT3 kinase, FLT3-ITD and FLT3 D835Y The mutant kinases all have high inhibitory activity.
TABLE 4 Compounds FLT3 kinase and FLT3-ITD, FLT3 D835Y Mutant kinase activity assay
Compound (I) Flt-3 IC 50 (nM) Flt-3 ITD IC 50 (nM) FLT3 D835Y IC 50 (nM)
I-24 1.7±0.1 3.5±2.1 0.3±0.1
Quizartinib 3.8±0.8 2.6±0.4 278.0±68.4
Test example 6
Effect of Compounds on cell proliferation of leukemia cell lines
1. Experimental materials
Leukemia cell lines: acute myelogenous leukemia cell strain MV4-11 (expression FLT3-ITD mutant gene)
MOLM-3 (expressing FLT3-ITD mutant gene and wild type FLT3 gene)
2. Test method
Leukemia cells in logarithmic growth phase were seeded at appropriate density into 96-well culture plates at 90. Mu.L per well, after overnight incubation, compounds at different concentrations (1. Mu.M for initial concentration, 5-fold dilution) were added for 72hr, and a solvent control group (negative control) was set. After the compound acts on cells for 72 hours, the influence of the compound on cell proliferation is detected by a CCK-8 cell counting kit (Shanghai Liji Biotech), 10 mu L of CCK-8 reagent is added into each hole, the hole is placed in an incubator at 37 ℃ for 2 to 4 hours, then a SpectraMax 190 reading is carried out by a full-wavelength micro-hole plate enzyme-labeling instrument, and the measurement wavelength is 450nm. The inhibition (%) of the compound on tumor cell growth was calculated using the following formula: inhibition (%) = (OD negative control well-OD administration well)/OD negative control well × 100%. IC (integrated circuit) 50 The values were determined by regression with a four parameter method using a microplate reader random plus software.
3. Results of the experiment
As can be seen from the results in Table 5, example 24 (I-24) showed significant inhibition of cell proliferation activity of acute myeloid leukemia cell lines MV4-11 and MOLM-3 expressing FLT3-ITD mutation.
TABLE 5 Effect of Compounds on MV4-11 and MOLM-3 cell proliferation
Compound (I) MV4-11 IC 50 (nM) MOLM-13IC 50 (nM)
I-24 <3.9 6.0±1.9
Quizartinib 4.8±2.7 9.8±4.0
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (14)

1. A compound of formula (I), a pharmaceutically acceptable salt thereof:
Figure FDA0003898573040000011
wherein the content of the first and second substances,
x is CH or N; y, Z or W are each independently CH; and said H atom may be replaced by a substituent selected from the group consisting of: halogen, C1-C3 alkyl, C2-C6 acyl, C1-C3 alkoxy, trifluoromethoxy, trifluoroethoxy;
ring a is selected from the group consisting of:
Figure FDA0003898573040000012
R 1 is a substituted or unsubstituted nitrogen-containing 5-6 membered heteroaryl; wherein the number of nitrogen atoms is 1-4, and the substituent is represented by R a Representing;
each R is a Are each independently selected from halogen, - (C0-C2 alkyl) -OH, - (C0-C1 alkyl) -COOH, - (C0-C1 alkyl) C (= O) OR 5 - (C0-C2 alkyl) NR 5 R 6 - (C0-C1 alkyl) C (= O) NR 5 R 6 Substituted or unsubstituted C1-C2 alkyl, substituted or unsubstituted methoxy; the substitution refers to substitution by one or more halogens;
R 5 、R 6 each independently selected from a hydrogen atom, a substituted or unsubstituted C1-C6 alkyl group; the substitution refers to substitution by one or more of C1-C3 alkyl, hydroxyl, halogen, carboxylic acid.
2. The compound, pharmaceutically acceptable salt thereof, of claim 1, wherein R is 1 Is a substituted or unsubstituted nitrogen-containing 5-6 membered heteroaryl; wherein the number of nitrogen atoms is 1-4, and the substituent is represented by R a Represents;
each R is a Each independently selected from halogen, - (C0-C1 alkyl) -OH, - (C0-C1 alkyl) -COOH, - (C0-C1 alkyl) C (= O) OR 5 - (C0-C1 alkyl) NR 5 R 6 Substituted or unsubstituted C1-C2 alkyl, substituted or unsubstituted methoxy; the substitution refers to substitution by one or more halogens.
3. A compound of formula (I), a pharmaceutically acceptable salt thereof:
Figure FDA0003898573040000013
wherein the content of the first and second substances,
x is CH or N; y, Z or W are each independently CH; and said H atom may be replaced by a substituent selected from the group consisting of: halogen, C1-C3 alkyl, C2-C6 acyl, C1-C3 alkoxy, trifluoromethoxy, trifluoroethoxy;
ring a is selected from the group consisting of:
Figure FDA0003898573040000021
R 4 selected from the group consisting of a hydrogen atom, a substituted or unsubstituted C1-C6 alkyl group, a substituted or unsubstituted C3-C10 cycloalkyl group, said substitution being by one or more halogens;
R 1 is selected from-NR 2 R 3 Substituted or unsubstituted 5-10 membered heteroaryl, substituted or unsubstituted 3-11 membered saturated or partially saturated heterocyclyl, said substituents consisting of R a Represents;
each R is a Are each independently selected from halogen, - (C0-C2 alkyl) -OH, - (C0-C1 alkyl) -COOH, - (C0-C1 alkyl) C (= O) OR 5 - (C0-C2 alkyl) NR 5 R 6 - (C0-C1 alkyl) C (= O) NR 5 R 6 Substituted or unsubstituted C1-C2 alkyl, substituted or unsubstituted methoxy; the substitution refers to substitution by one or more halogens;
R 2 and R 3 Each independently selected from a hydrogen atom, a substituted or unsubstituted C1-C3 alkyl group, said substitution being by halogen, hydroxy, amino or cyano;
or R 2 And R 3 May form, together with the nitrogen atom to which they are attached, a 4-to 8-membered heterocyclic ring optionally substituted with one or more R a Substitution;
R 5 、R 6 each independently selected from a hydrogen atom, a substituted or unsubstituted C1-C6 alkyl group; the substitution refers to substitution by one or more of C1-C3 alkyl, hydroxyl, halogen, carboxylic acid, C2-C6 carboxylate.
4. The compound, pharmaceutically acceptable salt thereof, according to claim 3,
R 1 is selected from-NR 2 R 3 Substituted or unsubstituted 5-10 membered heteroaryl, substituted or unsubstituted 3-11 membered saturated or partially saturated heterocyclyl, said substituents consisting of R a Represents;
each R is a Are each independently selected from halogen, - (C0-C1 alkyl) -OH, - (C0-C1 alkyl) -COOH, - (C0-C1 alkyl) C (= O) OR 5 - (C0-C1 alkyl) NR 5 R 6 - (C0-C1 alkyl) C (= O) NR 5 R 6 Substituted or unsubstituted C1-C2 alkyl, substituted or unsubstituted methoxy; the substitution refers to substitution by one or more halogens;
R 2 and R 3 Each independently selected from a hydrogen atom, a substituted or unsubstituted C1-C3 alkyl group, said substitution being by halogen, hydroxy, amino or cyano;
or R 2 And R 3 May form together with the nitrogen atom to which they are attached a 4-8 membered heterocyclic ring optionally substituted with one or more R a Substitution;
R 5 、R 6 each independently selected from a hydrogen atom, a substituted or unsubstituted C1-C6 alkyl group; the substitution refers to substitution by one or more of C1-C3 alkyl, hydroxyl, halogen, carboxylic acid.
5. The compound, pharmaceutically acceptable salt thereof, of claim 3, wherein R is 2 And R 3 Together with the nitrogen atom to which they are attached form a 4-7 membered heterocyclic ring, optionally substituted with one or more R a Substitution;
R a independently selected from halogen, hydroxy, - (C0-C1 alkyl) C (= O) NHR 5 - (C0-C2 alkyl) -NR 5 R 6 Substituted or unsubstituted C1-C2 alkyl, said substitution being by one or more halogens.
6. A compound selected from the group consisting of, or a pharmaceutically acceptable salt thereof,
Figure FDA0003898573040000022
Figure FDA0003898573040000031
Figure FDA0003898573040000041
Figure FDA0003898573040000051
7. a pharmaceutical composition comprising the following components:
1) A therapeutically effective amount of one or more compounds of any one of claims 1-6, pharmaceutically acceptable salts thereof; and
2) A pharmaceutically acceptable carrier or excipient.
8. The pharmaceutical composition of claim 7, further comprising one or more active agents selected from the group consisting of: immunosuppressants, glucocorticoids, non-steroidal anti-inflammatory drugs, vinca alkaloids, paclitaxel, DNA damaging agents, bcl-2 inhibitors, BTK inhibitors, JAK inhibitors, hsp90 inhibitors, ALK inhibitors, FLT3 inhibitors, PI3K inhibitors, and SYK inhibitors.
9. Use of a compound according to claim 1, or a pharmaceutically acceptable salt thereof, for the preparation of a pharmaceutical composition for the prevention or treatment of FLT3 or FLT3-ITD mediated indications.
10. The use of claim 9, wherein the indication is selected from the group consisting of: myelodysplastic syndrome, neurofibroma type I, multiple myeloma, glioblastoma, liver cancer, cervical cancer, lymphoma, bone metastasis, hormone refractory prostate cancer, hormone dependent prostate cancer, thyroid tumor, medullary thyroid carcinoma, mesothelioma, glioblastoma, including bone metastasis, merkel cell carcinoma, genitourinary tract tumors, merkel cell carcinoma, bladder cancer, papillary thyroid carcinoma, breast cancer, soft tissue sarcoma, glioma, neuroendocrine tumor, renal cell carcinoma, advanced solid tumors, undifferentiated astrocytic tumors, gastrointestinal stromal tumors, peller-linden tract syndrome, pancreatic cancer, pancreatic endocrine cancer, central nervous system tumors, metastatic renal cancer, endometrioid carcinoma, endometrioid adenocarcinoma, lung cancer, colorectal cancer, ovarian cancer, rhabdomyosarcoma, melanoma, retinoblastoma, central and peripheral nervous system tumors, acute leukemia, chronic leukemia, biliary tract cancer, bronchial cancer, esophageal cancer, testicular cancer, skin cancer, neuroblastoma, interstitial cell lymphoma, and large cell degenerative cell lymphoma.
11. The use of claim 9, wherein the indication is selected from the group consisting of: myelodysplastic syndrome, acute myeloid leukemia, neurofibroma type I, multiple myeloma, glioblastoma, non-small cell lung cancer, hepatocellular carcinoma, cervical cancer, lymphoma, bone metastasis, hormone refractory prostate cancer, hormone dependent prostate cancer, thyroid tumor, medullary thyroid cancer, mesothelioma, glioblastoma, including bone metastasis cancer, merkel cell cancer, genitourinary tract tumor, merkel cell cancer, bladder cancer, papillary thyroid cancer, breast cancer, soft tissue sarcoma, glioma, neuroendocrine tumor, renal cell carcinoma, advanced solid tumor, undifferentiated astrocytic cancer, gastrointestinal stromal tumor, von hippel-lindau syndrome, small cell lung cancer, pancreatic endocrine cancer, central nervous system tumor, metastatic renal cancer, endometrioid carcinoma, colorectal cancer, ovarian cancer, rhabdomyosarcoma, melanoma, retinoblastoma, central and peripheral nervous system tumors, chronic leukemia, esophageal cancer, bronchial cancer, testicular cancer, skin cancer, oral cancer, neuroblastoma, large cell lymphoma.
12. Use of a compound according to any one of claims 1 to 6, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 7, for the manufacture of a medicament for the prevention and/or treatment of IRAK4 mediated diseases.
13. Use of a compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, for the preparation of a pharmaceutical composition for the treatment or prevention of an IRAK 4-mediated indication.
14. The use of claim 13, wherein the indication is selected from the group consisting of: myelodysplastic syndrome, acute myeloid leukemia, cancer.
CN202010962799.8A 2019-09-12 2020-09-14 IRAK4 kinase inhibitor, preparation and application thereof Active CN112480101B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910867313 2019-09-12
CN201910867313X 2019-09-12

Publications (2)

Publication Number Publication Date
CN112480101A CN112480101A (en) 2021-03-12
CN112480101B true CN112480101B (en) 2022-11-25

Family

ID=74866134

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010962799.8A Active CN112480101B (en) 2019-09-12 2020-09-14 IRAK4 kinase inhibitor, preparation and application thereof

Country Status (2)

Country Link
CN (1) CN112480101B (en)
WO (1) WO2021047677A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022156788A1 (en) * 2021-01-22 2022-07-28 武汉人福创新药物研发中心有限公司 Benzimidazole compound and use thereof
WO2024017381A1 (en) * 2022-07-22 2024-01-25 深圳众格生物科技有限公司 Compound for inhibiting irak4 activity and use thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE465731T1 (en) * 2003-10-23 2010-05-15 Ab Science 2-AMINOARYLOXAZOLE COMPOUNDS AS TYROSINE KINASE INHIBITORS
US20070004660A1 (en) * 2005-06-10 2007-01-04 Baumann Christian A Synergistic Modulation of Flt3 Kinase Using Alkylquinolines and Alkylquinazolines
KR102066297B1 (en) * 2011-10-14 2020-01-14 암비트 바이오사이언시즈 코포레이션 Heterocyclic compounds and use thereof as modulators of type iii receptor tyrosine kinases
EP3092226B1 (en) * 2014-01-10 2019-03-13 Aurigene Discovery Technologies Limited Indazole compounds as irak4 inhibitors
MX2016017147A (en) * 2014-06-20 2017-06-07 Aurigene Discovery Tech Ltd Substituted indazole compounds as irak4 inhibitors.
CU20180006A7 (en) * 2015-07-15 2018-06-05 Aurigene Discovery Tech Ltd INDAZAOL AND AZAINDAZOL COMPOUNDS AS IRAK-4 INHIBITORS
HRP20230657T8 (en) * 2017-03-31 2023-11-10 Aurigene Oncology Limited Compounds and compositions for treating hematological disorders
WO2018234342A1 (en) * 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag Isoindolinone derivatives as irak4 modulators
WO2018234343A1 (en) * 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag Benzofurans as irak4 modulators

Also Published As

Publication number Publication date
WO2021047677A1 (en) 2021-03-18
CN112480101A (en) 2021-03-12

Similar Documents

Publication Publication Date Title
US11845760B2 (en) PRMT5 inhibitors
CN113544128A (en) KRAS-G12C inhibitors
CN106928219B (en) Nitrogen-containing fused heterocyclic compound, preparation method, intermediate, composition and application
CN113242857A (en) Preparation and application of imidazo aromatic ring compounds
EP3638680B1 (en) Heteroaromatic compounds as vanin inhibitors
CN112480101B (en) IRAK4 kinase inhibitor, preparation and application thereof
AU2019237329B2 (en) Substituted imidazolidin-2-one derivatives as PRMT5 inhibitors
WO2019189732A1 (en) Optically active crosslinked cyclic secondary amine derivative
CN111499634A (en) Quinazoline compound and application thereof in medicine
AU2017265769B2 (en) Novel 5H-Pyrrolo(2,3-D)Pyrimidin-6(7H)-one derivative
CN117062816A (en) Tricyclic-amide-bicyclic PRMT5 inhibitors
CN117177972A (en) PARP1 inhibitors and uses thereof
CN111978317A (en) Imidazopyridine MNK1/MNK2 kinase inhibitor and preparation method and application thereof
CN114555597A (en) Isocitrate Dehydrogenase (IDH) inhibitors
CN117858877A (en) C-linked inhibitors of ENL/AF9 YEATS
WO2023008577A1 (en) Ras/raf binding inhibitor compound
EP4342895A1 (en) Heteroaryl derivative compound and use thereof
CN111978318A (en) Imidazopyridine MNK1/MNK2 kinase inhibitor and preparation method and application thereof
WO2024054749A2 (en) Inhibitors of enl/af9 yeats and flt3
WO2023236960A1 (en) Carboxamide derivative having rsk inhibitory effect, pharmaceutical composition comprising same, and use thereof
KR20240046408A (en) Pyrimidin derivatives as protein kinase inhibitors and pharmaceutical compositions for treating disease containing the same
NZ730758B2 (en) Macrocyclic rip2 kinase inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant